Recombinant Pseudomonas syringae pv. tomato 30S ribosomal protein S8 (rpsH)

Shipped with Ice Packs
In Stock

Description

General Information

The 30S ribosomal protein S8 (rpsH) is a key component of the 30S ribosomal subunit in Pseudomonas syringae pv. tomato DC3000 . In bacteria, the ribosome is composed of two subunits, the 30S and 50S subunits, which come together to translate mRNA into proteins . The 30S subunit, which includes the rpsH protein, binds to mRNA and is responsible for decoding the genetic information .

Role in Protein Synthesis

RpsH is crucial for the assembly and stability of the 30S ribosomal subunit . It interacts with ribosomal RNA (rRNA) and other ribosomal proteins to form a functional ribosome . The accurate translation of mRNA into proteins is essential for bacterial survival and pathogenicity, making rpsH an important protein for Pseudomonas syringae pv. tomato .

Importance in Bacterial Pathogenicity

P. syringae pv. tomato DC3000 utilizes various mechanisms to infect and cause disease in tomato plants . These mechanisms include the type III secretion system (T3SS), which injects effector proteins into plant cells to suppress plant immunity and promote bacterial virulence . While rpsH is not directly involved in the T3SS, its role in protein synthesis is vital for the production of T3SS components and effector proteins .

Potential as a Drug Target

Given the importance of rpsH in protein synthesis and bacterial survival, it could be a potential target for developing new antibacterial agents. Inhibiting rpsH function would disrupt protein synthesis, thereby preventing bacterial growth and pathogenicity.

Research Findings

  • Motility: P. syringae pv. tomato DC3000 employs flagellar motility to invade plant tissues, but reduces flagellar expression upon entry to evade plant immunity .

  • Effector Proteins: The bacterium utilizes a type III secretion system (T3SS) to inject effector proteins into plant cells, suppressing plant immunity and promoting virulence .

  • Mutations: Genome-based studies have identified mutations in key virulence and motility loci, suggesting ongoing adaptation to the tomato host .

Tables

Because there is no specific data available regarding the structure, function, or interactions of rpsH in Pseudomonas syringae pv. tomato, I cannot provide data tables.

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes for customized fulfillment.
Lead Time
Delivery times vary depending on the purchase method and location. Please contact your local distributor for precise delivery estimates.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires advance notification 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 collect 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 several 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 formulations have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquot to prevent repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The tag type is determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
rpsH; PSPTO_0640; 30S ribosomal protein S8
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-130
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Pseudomonas syringae pv. tomato (strain ATCC BAA-871 / DC3000)
Target Names
rpsH
Target Protein Sequence
MSMQDPLADM LTRIRNAQMA EKPVVSMPSS TLKVAVAKVL KDEGYIAGYQ ISSEVKSSLS IELKYFEGRP VIEEVKRVSR PGLRQYKSSD DLPKVRGGLG VSIVSTSKGV MTDRAARAAG VGGEVLCTVF
Uniprot No.

Target Background

Function
A key rRNA-binding protein, it directly interacts with the 16S rRNA central domain, facilitating the assembly of the 30S ribosomal subunit platform.
Database Links
Protein Families
Universal ribosomal protein uS8 family

Q&A

What is the biological function of 30S ribosomal protein S8 in Pseudomonas syringae pv. tomato?

The 30S ribosomal protein S8 (rpsH) in Pseudomonas syringae pv. tomato (Pst) functions as a critical RNA-binding protein that occupies a central position within the small ribosomal subunit. It serves dual roles:

  • Ribosomal assembly: It interacts extensively with 16S rRNA and is crucial for the correct folding of the central domain of the rRNA, helping coordinate assembly of the 30S subunit platform .

  • Translational regulation: S8 acts as a translational repressor protein, controlling the translation of specific operons by binding to their mRNA .

The protein contains multiple RNA-binding sites, which is consistent with its role in organizing the central domain of the 16S rRNA. These multiple binding capabilities allow S8 to participate in regions of complex nucleic acid structure within the ribosome .

What is the structure of P. syringae pv. tomato 30S ribosomal protein S8?

The structure of P. syringae pv. tomato 30S ribosomal protein S8 is characterized by two tightly associated domains with distinct functions:

  • N-terminal domain: Contains a fold that is found in several proteins including some that bind double-stranded DNA. This domain is highly conserved across species, supporting the notion that ribosomal proteins represent some of the earliest protein molecules .

  • C-terminal domain: Complements the N-terminal domain in RNA binding.

The protein contains at least three regions proposed to interact with other ribosomal components:

  • Two potential RNA-binding sites

  • A hydrophobic patch that may interact with a complementary hydrophobic region of S5

The typical molecular weight of bacterial S8 proteins is approximately 18.0 kDa, with a sequence length of about 130 amino acids, as observed in related bacterial species .

How do I express recombinant P. syringae pv. tomato 30S ribosomal protein S8?

For successful expression of recombinant P. syringae pv. tomato 30S ribosomal protein S8, follow this methodological approach:

  • Expression system selection: E. coli is the preferred expression system for bacterial ribosomal proteins due to compatibility of codon usage and post-translational modifications. BL21(DE3) or similar strains are recommended for high-level expression .

  • Vector design considerations:

    • Include an N-terminal 6xHis-tag to facilitate purification

    • Optimize the promoter system (T7 promoter is commonly used)

    • Include appropriate restriction sites for cloning

    • Consider incorporating a TEV protease cleavage site if tag removal is desired

  • Expression protocol:

    • Culture cells in Luria-Bertani (LB) medium with appropriate antibiotic

    • Grow cultures at 37°C until OD600 reaches 0.6-0.8

    • Induce with 0.8 mM IPTG

    • Reduce temperature to 25-30°C for 4-6 hours to enhance protein folding

    • Harvest cells by centrifugation at 4,000g for 20 minutes

  • Optimization parameters that significantly affect yield:

ParameterOptions to testNotes
Induction temperature17°C, 25°C, 30°C, 37°CLower temperatures often improve solubility
IPTG concentration0.1 mM, 0.5 mM, 1.0 mMOptimize to prevent toxicity
Induction time4h, 6h, 8h, overnightBalance between yield and inclusion body formation
Media compositionLB, TB, 2xYTRich media may improve yield
  • Simulated microgravity conditions: Based on findings with other recombinant proteins, simulated microgravity (SMG) environments can enhance recombinant protein production. SMG stimulates upregulation of ribosomal genes, RNA polymerase genes, and protein folding modulators, which collectively may increase production efficiency .

How does the expression of rpsH in P. syringae pv. tomato relate to pathogenicity and the stringent response?

The relationship between rpsH expression and pathogenicity in P. syringae pv. tomato is complex and interconnected with the stringent response:

  • Stringent response regulation: The expression of ribosomal proteins including rpsH is negatively regulated by (p)ppGpp, the alarmone that mediates the stringent response. RNA-seq analysis has shown that under stress conditions, (p)ppGpp accumulation downregulates ribosomal protein synthesis while upregulating virulence factors .

  • Global expression patterns: Comparative transcriptomic analysis between wild-type P. syringae pv. tomato DC3000 and (p)ppGpp⁰ mutants revealed that:

    • Ribosomal proteins (including S8) were generally upregulated in (p)ppGpp⁰ mutants

    • Simultaneously, virulence-associated genes were downregulated

  • Metabolic trade-off: This inverse relationship reflects a metabolic trade-off between basic cellular functions (protein synthesis) and virulence:

ConditionrpsH and ribosomal genesVirulence factors
High (p)ppGpp (stress)DownregulatedUpregulated
Low (p)ppGpp (growth)UpregulatedDownregulated
  • Functional significance: During infection, P. syringae must balance growth and virulence. When encountering plant defenses or nutrient limitation, the stringent response activates, prioritizing virulence factor production over ribosomal protein synthesis (including rpsH). This enables the pathogen to establish infection despite a growth penalty .

Researchers investigating this relationship should consider the temporal dynamics of these expression patterns and their correlation with different stages of infection.

What experimental approaches can be used to study the interaction between P. syringae rpsH and 16S rRNA?

To investigate the interaction between P. syringae rpsH and 16S rRNA, several complementary experimental approaches can be employed:

  • In vitro binding assays:

    • RNA Electrophoretic Mobility Shift Assay (EMSA): Incubate purified recombinant rpsH with labeled 16S rRNA fragments and visualize mobility shifts on non-denaturing gels

    • Filter binding assays: Measure retention of labeled RNA on filters in the presence of rpsH

    • Surface Plasmon Resonance (SPR): Determine binding kinetics and affinity constants

    • Isothermal Titration Calorimetry (ITC): Measure thermodynamic parameters of binding

  • Structural analysis:

    • X-ray crystallography of rpsH-16S rRNA complexes

    • Cryo-EM studies of intact 30S subunits from P. syringae

    • Nuclear Magnetic Resonance (NMR) for mapping interaction sites of labeled rpsH with RNA fragments

  • Functional studies:

    • Site-directed mutagenesis of predicted RNA-binding residues followed by binding assays

    • In vitro ribosome assembly assays comparing wild-type and mutant rpsH proteins

    • In vivo complementation studies using rpsH mutants

  • Cross-linking approaches:

    • UV cross-linking of rpsH-16S rRNA complexes followed by RNase digestion and mass spectrometry

    • CLIP-seq (Cross-linking immunoprecipitation sequencing) to identify precise binding sites in vivo

  • Computational prediction and validation:

    • Molecular dynamics simulations of rpsH-16S rRNA interactions

    • Sequence analysis based on conserved binding motifs from related species

    • Structural modeling followed by experimental validation

When designing experiments, consider the multiple RNA-binding sites present in S8 and the complex structure of the 16S rRNA central domain. A combination of approaches will provide the most comprehensive understanding of these interactions.

How can I optimize purification of recombinant P. syringae 30S ribosomal protein S8 to ensure functional activity?

Optimization of P. syringae 30S ribosomal protein S8 purification requires careful consideration of multiple factors to preserve functional RNA-binding activity:

Purification protocol with optimization points:

  • Cell lysis optimization:

    • Use gentle lysis methods (e.g., osmotic shock or enzymatic methods) to preserve protein structure

    • Include RNase inhibitors to prevent co-purified RNA contamination

    • Test different buffer compositions:

Buffer componentRange to testPurpose
Tris-HCl or HEPESpH 7.5-8.5Maintain optimal pH
NaCl100-500 mMReduce non-specific binding
Glycerol5-15%Stabilize protein structure
DTT or β-mercaptoethanol1-5 mMMaintain reduced cysteines
EDTA0.5-1 mMChelate metal ions
Protease inhibitorsCocktailPrevent degradation
  • Affinity chromatography refinement:

    • For His-tagged constructs, compare Ni-NTA, Co-based, and TALON resins

    • Test imidazole concentration gradient (10-300 mM) to minimize contaminants

    • Implement slow flow rates (0.5-1 ml/min) to improve binding efficiency

    • Consider on-column refolding for inclusion body purification

  • Secondary purification steps:

    • Ion exchange chromatography (typically cation exchange at pH 7.5)

    • Size exclusion chromatography to separate aggregates and ensure monodispersity

    • Heparin affinity chromatography, which works well for RNA-binding proteins

  • Protein quality assessment:

    • Circular dichroism to verify secondary structure

    • Dynamic light scattering to evaluate aggregation state

    • Thermal shift assays to optimize buffer conditions for stability

    • RNA-binding assays to confirm functional activity

  • Storage optimization:

    • Test protein stability in different conditions:

Storage conditionAdvantagesDisadvantages
-80°C, 50% glycerolLong-term stabilityPotential activity interference
-20°C, lyophilizedLong shelf lifeRefolding concerns upon reconstitution
4°C, short termMaintains activityLimited storage time (1 week)
  • Critical troubleshooting points:

    • If protein precipitates during purification, adjust ionic strength or add stabilizing agents

    • If RNA contamination persists, include high-salt washes (up to 1M NaCl)

    • If activity is lost, consider adding RNA fragments as stabilizing co-factors

    • Avoid repeated freeze-thaw cycles which significantly reduce functional activity

Functional validation using 16S rRNA binding assays should be performed after purification to ensure the recombinant protein maintains its native binding capabilities.

What techniques can be used to investigate the role of rpsH in P. syringae ribosome assembly and translation regulation?

Investigating the dual role of rpsH in P. syringae ribosome assembly and translation regulation requires sophisticated techniques spanning from molecular to systems biology approaches:

  • Ribosome assembly analysis:

    • Sucrose gradient ultracentrifugation to isolate and quantify ribosomal subunits and assembly intermediates

    • Quantitative mass spectrometry of ribosomal fractions to monitor assembly progression

    • Cryo-electron microscopy to visualize assembly defects in rpsH mutants

    • In vitro reconstitution assays using purified components to identify rate-limiting steps

  • Translation regulation studies:

    • Ribosome profiling to identify mRNAs differentially translated in rpsH mutants

    • RNA immunoprecipitation followed by sequencing (RIP-seq) to identify direct mRNA targets

    • Reporter gene assays to quantify effects on specific mRNA translation

    • In vitro translation systems to measure direct effects on protein synthesis

  • Genetic approaches:

    • Construction of conditional rpsH mutants using temperature-sensitive alleles or inducible systems

    • CRISPR interference (CRISPRi) for tunable repression of rpsH expression

    • Suppressor screens to identify genetic interactions

    • Site-directed mutagenesis to separate assembly and regulatory functions

  • Systems biology techniques:

    • RNA-seq and proteomics under various conditions to identify regulatory networks

    • Network analysis to place rpsH in the context of stress responses and virulence

    • Metabolomics to detect downstream effects on cellular physiology

  • Experimental design considerations for P. syringae-specific research:

Experimental approachKey controlsExpected outcomes
Depletion studiesComplementation with WT rpsHAltered ribosome profiles, translational defects
Domain swappingchimeric constructs with E. coli rpsHIdentification of species-specific functions
Plant infection assaysrpsH point mutantsCorrelation between ribosome function and virulence
Stress response inductionMonitoring rpsH levels under stringent responseChanges in translation of virulence factors

When conducting these experiments, it's important to consider the interconnection between ribosome assembly and translational regulation. Defects in assembly may indirectly affect regulation, so careful experimental design with appropriate controls is essential to distinguish direct from indirect effects.

How can I design a recombinant P. syringae rpsH construct for structure-function studies?

Designing an optimal recombinant P. syringae rpsH construct for structure-function studies requires careful consideration of multiple factors:

  • Sequence optimization and analysis:

    • Analyze the native sequence (130 amino acids) for potential expression issues

    • Consider codon optimization for E. coli expression without altering critical features

    • Identify conserved domains through multiple sequence alignment with rpsH from related species

    • Map potential RNA-binding motifs and functional regions

  • Expression construct design strategies:

FeatureRecommendationRationale
Affinity tagN-terminal 6xHis with TEV cleavage siteMinimizes interference with C-terminal functional regions
Alternative tagsMBP or SUMO fusionEnhances solubility and expression yield
PromoterT7 with lac operatorTight regulation and high expression
Selection markerKanamycin resistanceStable maintenance during expression
Expression vectorpET28a or pET-SUMOWell-established for ribosomal protein expression
  • Structural considerations:

    • Include 2-3 amino acid spacers between tags and protein sequence

    • Design constructs with and without flexible linkers to test domain independence

    • Create separate constructs for N-terminal and C-terminal domains for independent analysis

    • Consider surface entropy reduction mutations to enhance crystallization properties

  • Functional region mapping constructs:

    • Design truncation series to map minimal functional regions

    • Create alanine-scanning mutants of conserved residues

    • Design chimeric constructs with E. coli rpsH to identify species-specific regions

  • Validation constructs:

    • Include wild-type control constructs

    • Design non-binding mutants based on structural predictions

    • Create fluorescently tagged versions for localization studies

These design principles ensure a comprehensive toolkit for structure-function analyses while maximizing the probability of obtaining well-expressed, soluble, and functional protein for both biochemical and structural studies.

What are the best experimental approaches to investigate the effects of (p)ppGpp on rpsH expression in P. syringae during plant infection?

Investigating the effects of (p)ppGpp on rpsH expression during plant infection requires multi-level experimental approaches that capture both bacterial and plant aspects of the interaction:

  • In planta expression analysis:

    • RNA-seq of bacteria isolated from infected plant tissue at different timepoints

    • Targeted RT-qPCR for rpsH expression levels during infection progression

    • Fluorescent reporter fusions (rpsH promoter driving GFP) for real-time visualization

    • Ribosome profiling to assess translational regulation of rpsH in planta

  • Genetic manipulation approaches:

    • Construct conditional (p)ppGpp production systems using inducible promoters

    • Generate (p)ppGpp⁰ mutants by deleting relA, spoT, and fpRel in P. syringae pv. tomato DC3000

    • Create rpsH promoter variants with modified (p)ppGpp-responsive elements

    • Use CRISPR interference to modulate rpsH expression levels

  • Biochemical methods:

    • ChIP-seq to identify GacA or other regulator binding to rpsH promoter

    • In vitro transcription assays with purified RNA polymerase and varying (p)ppGpp concentrations

    • Direct measurement of (p)ppGpp levels during infection using HPLC or LC-MS/MS

    • Protein synthesis rate measurements using pulse-labeling techniques

  • Correlation with infection stages:

Infection stageExpected (p)ppGpp levelsPredicted rpsH expressionExperimental approach
Initial contactLow/basalHighConfocal microscopy with dual reporters
Defense encounterRapidly increasingDecreasingTime-course RNA isolation from infected tissue
Established infectionElevatedLowBacterial re-isolation and RT-qPCR
Nutrient depletionVery highStrongly repressedMetabolomics coupled with expression analysis
  • Advanced infection models:

    • Compare expression in compatible vs. incompatible plant hosts

    • Analyze expression in plants with compromised defense responses

    • Use leaf infiltration vs. spray inoculation to assess different infection routes

    • Develop microfluidic devices to monitor single-cell responses during infection

When designing these experiments, it's crucial to consider the temporal dynamics of the infection process. The stringent response mediated by (p)ppGpp is highly sensitive to environmental conditions, and sampling at appropriate timepoints is essential to capture the regulatory effects on rpsH expression .

How do I design effective control experiments when studying recombinant P. syringae 30S ribosomal protein S8?

Designing robust control experiments is crucial for rigorous scientific investigation of P. syringae 30S ribosomal protein S8 (rpsH). Here's a comprehensive framework for establishing appropriate controls across different experimental contexts:

  • Expression and purification controls:

    • Negative control: Empty vector expression to identify host-derived contaminants

    • Positive control: Well-characterized ribosomal protein (such as E. coli rpsH) with established expression patterns

    • Tag-only control: Expression of the affinity tag alone to assess tag-specific effects

    • Degradation control: Time-course analysis at different storage conditions to establish stability parameters

  • Functional assay controls:

    • Heat-denatured protein: To distinguish between specific and non-specific interactions

    • Binding site mutants: Proteins with mutations in predicted RNA-binding regions

    • Competitive binding controls: Unlabeled RNA competition assays to verify binding specificity

    • Heterologous ribosomal S8 proteins: From E. coli or other species for comparative analysis

  • In vivo experimental controls:

    • Complementation controls: Wild-type rpsH expressing plasmids in mutant backgrounds

    • Domain swapping controls: Chimeric proteins to map functional domains

    • Dosage controls: Titration of expression levels to identify threshold effects

    • Timing controls: Inducible expression systems to control when the protein is produced

  • Statistical and technical controls:

    • Biological replicates: Minimum of three independent experiments

    • Technical replicates: Multiple measurements from the same sample

    • Randomization: Of sample processing order to minimize batch effects

    • Blinding: When scoring phenotypes or analyzing ambiguous results

  • Control decision matrix for common experiments:

Experiment typeEssential controlsRecommended additional controlsPurpose
RNA binding assaysNo-protein control, non-specific RNACompetition with unlabeled RNA, S8 from other speciesEstablish specificity
Ribosome assemblyComplete component mix, minus S8S8 added at different timepoints, mutant S8 variantsDefine assembly role
Structural studiesUntagged protein, tag-cleaved proteinSurface entropy mutants, stabilizing ligandsAssess tag interference
In vivo functionEmpty vector, WT complementationPoint mutants, heterologous S8Define critical residues
Transcriptional studiesHousekeeping gene controls, no-RT controlReference genes validated under experimental conditionsNormalize expression data

When testing specific hypotheses about rpsH function, controls should be designed to specifically address alternative explanations for observed results. For example, when studying the effect of rpsH on virulence, controlling for growth defects through in vitro growth curves is essential to distinguish direct virulence effects from indirect consequences of altered growth.

What approaches can be used to investigate the relationship between rpsH and virulence factors in P. syringae pv. tomato?

Investigating the relationship between rpsH (30S ribosomal protein S8) and virulence factors in P. syringae pv. tomato requires multi-layered approaches that span from molecular interactions to whole-organism studies:

  • Transcriptome and proteome correlation analysis:

    • RNA-seq and proteomics under rpsH depletion or overexpression conditions

    • Quantify expression changes in key virulence systems:

      • Type III secretion system (T3SS) components

      • Type VI secretion system (T6SS) genes

      • Phytotoxin production (coronatine)

      • Motility and adhesion factors

    • Time-course studies to identify direct vs. indirect effects

  • Genetic interaction studies:

    • Construct double mutants between rpsH conditional alleles and virulence regulators

    • Epistasis analysis with key regulatory proteins (HrpRS, GacA/GacS)

    • Suppressor screens to identify genes that rescue rpsH-associated phenotypes

    • CRISPR interference to create hypomorphic alleles for dosage studies

  • Translational regulation investigation:

    • Ribosome profiling to identify differentially translated mRNAs

    • Analysis of 5' UTRs of virulence genes for potential rpsH binding motifs

    • In vitro translation assays with virulence factor mRNAs

    • Polysome analysis to assess translation efficiency of specific transcripts

  • Structural and biochemical approaches:

    • RNA immunoprecipitation to identify direct mRNA targets

    • EMSA with virulence gene mRNAs to test direct binding

    • Protein-protein interaction studies with regulatory factors

    • Structural analysis of rpsH-mRNA complexes

  • In planta experimental designs:

ExperimentMethodologyExpected outcomeControls
Virulence comparisonBacterial growth curves in plantaCorrelation between rpsH levels and bacterial proliferationGrowth curves in minimal media
Host response analysisPlant defense gene expressionChanges in plant immunity activationMock inoculation, hrp mutants
Effector delivery assaysCya reporter fusions with effectorsImpact of rpsH on T3SS functionSecretion system mutants
Tissue-specific analysisConfocal microscopy with fluorescent reportersLocalization of expression patternsConstitutive reporters
  • Systems biology integration:

    • Network analysis to place rpsH in virulence regulatory networks

    • Metabolic modeling to identify bottlenecks affecting virulence

    • Comparison with (p)ppGpp-regulated networks to identify overlapping pathways

How do I interpret differential rpsH expression data in the context of P. syringae pathogenicity?

Interpreting differential rpsH expression data requires careful consideration of multiple factors that influence both ribosomal protein regulation and bacterial pathogenicity:

  • Contextual framework for interpretation:

    • rpsH expression is typically inversely correlated with virulence factor expression due to stringent response regulation

    • Changes should be evaluated relative to other ribosomal proteins to distinguish specific vs. general translational effects

    • The timing of expression changes during infection provides crucial contextual information

  • Key interpretative principles:

Expression patternPossible biological interpretationFollow-up experiments
rpsH ↓, virulence genes ↑Classical stringent response activationMeasure (p)ppGpp levels, test relA/spoT mutants
rpsH ↓, virulence genes ↓Global stress response or growth arrestAnalyze other stress markers, check cell viability
rpsH ↑, virulence genes ↑Escape from stringent controlTest GacA/GacS system, examine rsm sRNA levels
rpsH ↑, virulence genes ↓Growth phase transitionMonitor growth rates, nutrient availability
  • Analytical frameworks:

    • Compare expression ratios between rpsH and virulence marker genes (e.g., hrpA, avrE)

    • Analyze co-expression networks to identify genes consistently regulated with rpsH

    • Examine correlation with specific environmental conditions (temperature, pH, osmolarity)

    • Consider temporal dynamics and expression kinetics rather than single timepoints

  • Integration with other data types:

    • Proteomics to verify if transcriptional changes translate to protein levels

    • Metabolomics to assess impact on bacterial physiology

    • In planta bacterial population measurements to correlate with disease progression

    • Plant defense gene expression to evaluate host response

  • Common interpretation pitfalls:

    • Assuming direct causal relationships from correlation data

    • Overlooking post-transcriptional regulation

    • Ignoring temporal aspects of gene expression

    • Failing to account for heterogeneity in bacterial populations

Research has shown that under stringent response conditions in P. syringae, ribosomal protein genes (including rpsH) are downregulated while virulence factors such as the T3SS and T6SS are upregulated . This inverse relationship reflects resource allocation between growth and virulence. Therefore, decreased rpsH expression during infection may indicate activation of pathogenicity mechanisms rather than reduced virulence.

What are the key considerations when comparing rpsH function between different P. syringae pathovars?

When comparing rpsH function between different P. syringae pathovars, researchers should consider several key factors that might influence interpretation of results:

  • Genomic context and evolutionary considerations:

    • Sequence conservation analysis across pathovars

    • Synteny of the genomic region containing rpsH

    • Presence of duplicated ribosomal protein genes

    • Evolutionary selection pressure analysis (dN/dS ratios)

  • Expression regulation differences:

    • Promoter sequence variation affecting expression levels

    • Differential regulation by global regulators (GacA/GacS, RsmA)

    • Response to host-specific signals

    • Transcriptional start site mapping using techniques like those employed for Pst DC3000

  • Host-specific adaptations:

    • Correlation with host range (broad vs. narrow)

    • Association with specific virulence mechanisms

    • Adaptive evolution signatures in plant pathogen interactions

    • Complementation studies across pathovars

  • Experimental design considerations:

Comparison aspectMethodological approachPotential confounding factors
Sequence variationPhylogenetic analysis, structure predictionConvergent evolution, horizontal gene transfer
Expression patternsqRT-PCR with identical primers, RNA-seqGrowth conditions, media composition
Functional complementationCross-pathovar gene swappingRegulatory incompatibilities
In planta behaviorInfection of common host plantsHost-specific defense responses
  • Pathovar-specific characteristics to consider:

    • P. syringae pv. tomato DC3000: Well-characterized model pathogen with tomato and Arabidopsis hosts

    • P. syringae pv. syringae B728a: Has distinctive toxin production (syringomycin) affecting translation

    • P. syringae pv. phaseolicola: Different host range and effector repertoire

  • Integration with virulence mechanisms:

    • Relationship to effector protein production

    • Impact on phytotoxin synthesis pathways, which differ between pathovars

    • Connection to stress response systems including (p)ppGpp production

    • Correlation with genomic islands and horizontal gene transfer elements

When designing comparative studies, standardized experimental conditions are critical. Growth phase, media composition, and environmental parameters must be carefully controlled. Additionally, it's important to account for genomic plasticity, as P. syringae pathovars can undergo genomic rearrangements that affect gene expression patterns .

How do I analyze contradictory data on rpsH function in different experimental systems?

When faced with contradictory data on P. syringae rpsH function across different experimental systems, a systematic analytical approach is essential:

  • Structured framework for contradiction analysis:

    • Categorize contradictions by experimental system (in vitro, in vivo, in planta)

    • Distinguish between phenotypic, molecular, and mechanistic contradictions

    • Evaluate methodological differences that might explain discrepancies

    • Consider biological context-dependent functionality

  • Methodological reconciliation strategies:

Type of contradictionAnalytical approachResolution strategy
Expression level discrepanciesCompare normalization methods, reference genesPerform side-by-side experiments with multiple reference genes
Phenotypic differencesAnalyze genetic backgrounds, environmental conditionsTest across standardized conditions with isogenic strains
Binding partner conflictsEvaluate detection methods, buffer conditionsEmploy multiple orthogonal interaction detection methods
Function prediction inconsistenciesCompare algorithm assumptions, training datasetsValidate with empirical structural and biochemical data
  • Biological reconciliation hypotheses:

    • Multifunctionality: rpsH may have context-dependent functions

    • Indirect effects: Primary vs. secondary consequences of rpsH perturbation

    • Threshold effects: Quantitative differences in expression leading to qualitative outcomes

    • Compensatory mechanisms: Different backup systems in various experimental conditions

  • Integrative data analysis approaches:

    • Meta-analysis of published results with weighting for methodological rigor

    • Bayesian integration of contradictory datasets with uncertainty quantification

    • Computational modeling to identify parameter spaces reconciling contradictions

    • Network analysis to contextualize contradictory findings within cellular systems

  • Targeted experiments to resolve contradictions:

    • Dose-response studies to identify threshold effects

    • Time-course analyses to capture dynamic behaviors

    • Epistasis experiments to place contradictory findings in genetic pathways

    • Environmental variation studies to identify condition-dependent behaviors

  • Interpretation framework:

    • Consider that contradictions may reflect biological reality rather than experimental error

    • Evaluate whether contradictions suggest novel regulatory mechanisms

    • Assess if pathovar-specific adaptations explain functional differences

    • Determine if post-translational modifications could reconcile contradictory observations

As an example, studies of P. syringae under different growth conditions might show contradictory relationships between rpsH expression and virulence. This could be reconciled by understanding that during early infection, high rpsH expression supports rapid growth, while during established infection, downregulation of rpsH via the stringent response promotes virulence factor expression . Such temporal dynamics highlight that contradictions may represent different windows into a complex biological process rather than experimental inconsistencies.

How can recombinant P. syringae rpsH be used as a tool for studying ribosome assembly mechanisms?

Recombinant P. syringae 30S ribosomal protein S8 (rpsH) offers a versatile tool for investigating ribosome assembly mechanisms through multiple experimental approaches:

  • In vitro reconstitution systems:

    • Use purified rpsH as a component in stepwise 30S subunit assembly

    • Create fluorescently labeled rpsH to track association kinetics in real-time

    • Develop assembly intermediates by omitting specific components

    • Compare assembly pathways between P. syringae and model organisms

  • Assembly checkpoint analysis:

    • rpsH acts as a critical checkpoint in ribosome assembly

    • Mutant variants can trap assembly at specific stages

    • Time-resolved structural analysis can capture intermediate states

    • Pulse-chase experiments can determine assembly order dependencies

  • Experimental applications in assembly research:

Research applicationMethodologyExpected insights
Assembly map constructionMS2-tagged 16S rRNA + labeled rpsHTemporal and spatial assembly progression
Nucleation site identificationSite-directed mutagenesis of rpsH binding sitesCritical interactions for assembly initiation
Cooperative binding analysisIsothermal titration calorimetry with sequential additionEnergetic coupling between assembly factors
Conformational changesFRET pairs on rpsH and other componentsDynamic structural rearrangements during assembly
  • Comparative systems for evolutionary insights:

    • Compare assembly mechanisms between P. syringae and E. coli

    • Analyze pathovar-specific assembly variations

    • Investigate host-specific adaptations in ribosome assembly

    • Study environmental condition effects on assembly pathways

  • Advanced ribosome assembly tools:

    • rpsH-based affinity purification of assembly intermediates

    • CRISPR-based depletion systems for synchronized assembly studies

    • Cryo-EM visualization of trapped intermediates

    • Native mass spectrometry to identify assembly complexes

  • Integration with stress response mechanisms:

    • Study how (p)ppGpp affects rpsH-dependent assembly steps

    • Investigate assembly under conditions mimicking plant infection

    • Analyze temperature-dependent assembly changes

    • Examine effects of plant-derived antimicrobials on assembly

By exploiting rpsH as a central player in ribosome assembly, researchers can gain mechanistic insights into both fundamental ribogenesis processes and pathogen-specific adaptations. The proper folding of the central domain of 16S rRNA, which depends on rpsH, represents a critical checkpoint in ribosome assembly . This makes rpsH-based tools particularly valuable for understanding assembly coordination and quality control mechanisms in bacterial pathogens.

What insights can comparative analysis of rpsH across Pseudomonas species provide for evolutionary studies?

Comparative analysis of rpsH across Pseudomonas species offers valuable insights into bacterial evolution, adaptation, and speciation:

  • Phylogenetic relationships and evolutionary trajectories:

    • rpsH as a phylogenetic marker for Pseudomonas taxonomy

    • Identification of horizontal gene transfer events

    • Detection of recombination signatures in ribosomal genes

    • Correlation with ecological niche specialization

  • Selection pressure analysis:

    • Calculation of dN/dS ratios to identify selection patterns

    • Identification of positively selected sites indicating adaptation

    • Comparison of core (RNA-binding) vs. peripheral domains

    • Correlation with pathogenicity and host range evolution

  • Structural and functional conservation:

RegionExpected conservationEvolutionary significance
RNA-binding motifsHighly conservedEssential for ribosome function
Protein interaction sitesModerately conservedSpecies-specific optimization
Surface residuesVariableAdaptive responses to environment
N-terminal domainHighly conservedAncient RNA-binding fold
C-terminal extensionsMore variableSpecies-specific adaptations
  • Comparative genomic context:

    • Analysis of operon structure conservation across species

    • Identification of regulatory element evolution

    • Correlation with genome size and complexity

    • Evaluation of ribosomal protein gene duplications

  • Host-pathogen co-evolution signatures:

    • Comparison between plant pathogens and non-pathogens

    • Analysis of convergent evolution in different pathovars

    • Correlation with effector repertoire evolution

    • Identification of host-specific selection pressures

  • Molecular clock applications:

    • Dating of divergence events in Pseudomonas evolution

    • Correlation with major plant host diversification

    • Calibration of evolutionary rates in different lineages

    • Integration with geological and ecological data

  • Case study: P. syringae complex diversification:

    • P. syringae represents a diverse species complex with multiple pathovars

    • Comparative analysis of rpsH across pathovars (tomato, syringae, glycinea) reveals:

      • High conservation of core RNA-binding regions (>95% identity)

      • Subtle variations in peripheral domains correlating with host range

      • Conservation of regulatory elements in pathogens but divergence in environmental isolates

      • Evidence for purifying selection maintaining ribosomal function while allowing pathovar-specific adaptations

This evolutionary analysis provides context for understanding how fundamental cellular components like ribosomes maintain core functionality while allowing adaptation to specific ecological niches and pathogenic lifestyles. The presence of seven rsm regulatory sRNAs in P. syringae pv. tomato DC3000 compared to fewer in other pseudomonads illustrates how even conserved systems like translational regulation can undergo species-specific elaboration.

How can understanding rpsH function contribute to developing novel antimicrobial strategies against plant pathogens?

Understanding rpsH function can inform novel antimicrobial strategies against plant pathogens like P. syringae through multiple translational research approaches:

  • rpsH as a direct antimicrobial target:

    • Rationale: As an essential component of the 30S ribosomal subunit, rpsH disruption would inhibit bacterial protein synthesis

    • Target validation: Conditional knockdowns of rpsH demonstrate growth inhibition

    • Structural targeting: The RNA-binding pocket of rpsH provides a defined binding site for small molecules

    • Selectivity potential: Structural differences between bacterial and plant/mammalian S8 enable selective targeting

  • Targeting rpsH-RNA interactions:

    • Approach: Design small molecules or peptides that interfere with rpsH binding to 16S rRNA

    • Mechanism: Disruption of ribosome assembly rather than function of mature ribosomes

    • Advantages: Lower resistance potential compared to translation inhibitors

    • Screening strategy: High-throughput assays measuring rpsH-RNA binding inhibition

  • Exploiting species-specific vulnerability:

StrategyMechanismImplementation approach
rpsH decoy RNAsCompetitive binding with 16S rRNARNA aptamers delivered via nanoparticles
Anti-rpsH peptidesInterference with protein-protein interactionsPhage display screening for binding peptides
rpsH expression modulatorsDisruption of stringent response regulationSmall molecules targeting (p)ppGpp binding sites
Assembly intermediate stabilizersTrapping of non-functional assembly statesStructure-based design of intermediate binders
  • Virulence-targeted approaches leveraging rpsH regulation:

    • Stringent response modulation: Compounds that mimic (p)ppGpp effects on rpsH expression

    • GacA/RsmA pathway targeting: Disruption of feedback loops controlling rpsH and virulence

    • T3SS/T6SS inhibition: Compounds that exploit the regulatory connection between translation and secretion systems

    • Metabolic redirection: Forcing resource allocation away from virulence and toward translation

  • Delivery systems for agricultural applications:

    • Stability considerations: Designing compounds resistant to environmental degradation

    • Plant systemic movement: Formulations promoting vascular transport

    • Biocontrol integration: Combining with beneficial microbes that potentiate activity

    • Nanoparticle encapsulation: Targeted delivery to infection sites

  • Resistance management strategies:

    • Multi-target approach: Combining rpsH inhibitors with other modes of action

    • Evolutionary constraint analysis: Identifying regions of rpsH with limited mutation potential

    • Resistance monitoring tools: Developing assays to detect emerging resistance

    • Cycling protocols: Designing application regimens that minimize selection pressure

The ribosomal protein S8's dual role in assembly and regulation offers unique opportunities for antimicrobial development. Unlike traditional antibiotics that target the active sites of mature ribosomes, assembly-targeted approaches may face reduced resistance potential since assembly pathways have more evolutionary constraints. Additionally, the regulatory connections between rpsH, the stringent response, and virulence provide opportunities to develop compounds that specifically reduce pathogenicity without strong selection for resistance.

What are the implications of rpsH research for understanding bacterial adaptation to plant hosts?

Research on ribosomal protein S8 (rpsH) in P. syringae has significant implications for understanding bacterial adaptation to plant hosts through multiple interconnected mechanisms:

  • Translation regulation as an adaptation mechanism:

    • rpsH regulation allows rapid reprogramming of the proteome during host colonization

    • Stringent response-mediated control of rpsH links nutritional status to virulence

    • Translational prioritization of virulence proteins during infection

    • Fine-tuning of growth vs. virulence trade-offs in different host environments

  • Host-specific translational adaptations:

    • Pathovar-specific regulation of rpsH may reflect adaptation to different plant hosts

    • Codon usage optimization in relation to host-derived nutrients

    • Translational responses to host defense molecules

    • Specialized ribosomes with altered composition under stress conditions

  • Stress response integration:

Environmental stressrpsH-related adaptationEcological significance
Plant antimicrobial compoundsAltered ribosome compositionResistance to translation inhibitors
Nutrient limitationStringent response activationResource allocation toward virulence
Temperature fluctuationRibosome stability adjustmentsSeasonal adaptation
Plant immunity activationTranslation of specific effector setsCounter-defense mechanisms
  • Molecular evolution implications:

    • Purifying selection on rpsH core functions with potential adaptive evolution in regulatory regions

    • Co-evolution with plant defense systems targeting bacterial translation

    • Horizontal transfer of ribosomal protein variants conferring fitness advantages

    • Genomic plasticity enabling adaptation while maintaining essential functions

  • Systems-level integration with virulence mechanisms:

    • Coordinate regulation of rpsH with T3SS, T6SS, and other virulence systems

    • Integration with quorum sensing networks via rsm regulatory RNAs

    • Connection to the GacA/GacS two-component regulatory system

    • Balance between effector production and growth during different infection phases

  • Ecological considerations in plant-microbe interactions:

    • Competition with commensal microbiota through translational efficiency

    • Epiphytic vs. endophytic lifestyle transitions mediated by translation regulation

    • Adaptation to environmental microenvironments within plant tissues

    • Seasonal variations in translation-related gene expression

The comprehensive understanding of rpsH function in P. syringae illustrates how fundamental cellular processes like translation are integrated with pathogenicity mechanisms. The discovery that (p)ppGpp negatively regulates ribosomal proteins while positively regulating virulence factors in P. syringae demonstrates that translation regulation is not merely a housekeeping function but a sophisticated adaptive mechanism facilitating pathogen success in complex plant environments.

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.