Recombinant Rhodopirellula baltica Polyribonucleotide nucleotidyltransferase (pnp), partial

Shipped with Ice Packs
In Stock

Description

Overview of Rhodopirellula baltica

R. baltica is a member of the phylum Planctomycetes, characterized by its complex cell structure, which includes intracellular membranes. This bacterium is of significant interest due to its ecological role and potential applications in biotechnology. It is known for its ability to degrade complex organic compounds and form biofilms, which are crucial for its survival and interaction with the environment .

Polyribonucleotide Nucleotidyltransferase (PNP) Function

PNP enzymes are generally involved in the processing and degradation of RNA. They catalyze the transfer of nucleotidyl groups from nucleoside triphosphates to the 3'-hydroxyl group of RNA, leading to the formation of polyribonucleotides. This process is crucial for RNA turnover and the regulation of gene expression in bacteria.

Research Findings and Data

While specific data on the recombinant PNP from R. baltica is not available, research on R. baltica has highlighted its unique genetic and metabolic features. For example, R. baltica possesses a diverse set of transport proteins, including sodium-dependent symporters and proton-dependent antiporters, which are essential for its survival in marine environments .

Table: Transport Proteins in Rhodopirellula baltica

Type of Transport ProteinPercentage of Total Transporters
Channels12%
Secondary Carriers33%
Primary Active Transport41%
OtherVariable

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your preferred format in order notes for customized preparation.
Lead Time
Delivery times vary depending on the purchasing method and location. Please contact your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard blue ice packs unless dry ice shipping is requested in advance. Additional fees apply for dry ice shipping.
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%, which 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. Aliquot for multiple uses 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; we will prioritize its development.
Synonyms
pnp; RB5804; Polyribonucleotide nucleotidyltransferase; EC 2.7.7.8; Polynucleotide phosphorylase; PNPase
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Protein Length
Partial
Purity
>85% (SDS-PAGE)
Species
Rhodopirellula baltica (strain DSM 10527 / NCIMB 13988 / SH1)
Target Names
pnp
Uniprot No.

Target Background

Function

This protein is involved in mRNA degradation. It catalyzes the processive 3'- to 5'-direction phosphorolysis of single-stranded polyribonucleotides.

Database Links

KEGG: rba:RB5804

STRING: 243090.RB5804

Protein Families
Polyribonucleotide nucleotidyltransferase family
Subcellular Location
Cytoplasm.

Q&A

What expression systems are commonly used for producing Recombinant R. baltica PNPase?

Recombinant R. baltica PNPase can be produced using several expression systems, each with distinct advantages depending on your experimental requirements. The most common expression hosts include:

  • E. coli: This prokaryotic expression system offers high yield, rapid growth, and cost-effectiveness. It's particularly suitable for initial characterization studies and when large quantities of protein are needed.

  • Yeast: Systems such as Saccharomyces cerevisiae or Pichia pastoris provide eukaryotic post-translational modifications while maintaining relatively high expression levels.

  • Baculovirus: This insect cell-based system offers superior folding capability and post-translational modifications for complex proteins.

  • Mammalian cell systems: These provide the most authentic post-translational modifications and protein folding but typically yield lower quantities of protein compared to other systems .

The choice of expression system should be guided by your specific research questions. For basic biochemical characterization, E. coli systems may be sufficient, while studies investigating structural properties or requiring specific modifications might benefit from eukaryotic expression systems. Regardless of the chosen system, the recombinant protein typically achieves ≥85% purity as determined by SDS-PAGE analysis .

What purification strategies are most effective for R. baltica PNPase?

The purification of recombinant R. baltica PNPase typically employs a multi-step chromatographic approach to achieve high purity (≥85% as determined by SDS-PAGE) . Based on established protocols for similar proteins, an effective purification strategy would include:

  • Initial capture: Immobilized metal affinity chromatography (IMAC) using a histidine tag is highly effective for initial purification. Nickel or cobalt resins capture the His-tagged protein from the crude lysate.

  • Intermediate purification: Ion exchange chromatography (IEX) helps remove contaminants with different charge properties. The choice between cation or anion exchange depends on the PNPase's isoelectric point.

  • Polishing step: Size exclusion chromatography (SEC) separates the target protein based on molecular size, removing aggregates and degradation products.

For researchers working with PNPase in functional studies, it's particularly important to verify enzymatic activity after purification. Activity assays measuring the degradation of defined RNA substrates can confirm that the purified enzyme maintains its native functionality. Additionally, ensuring the removal of nucleic acid contaminants is critical, as these can interfere with subsequent functional analyses.

How does PNPase contribute to bacterial persistence mechanisms and what experimental approaches best elucidate these functions?

PNPase plays a significant role in bacterial persistence mechanisms, particularly in response to antibiotic stress. In E. coli, PNPase has been identified as a key regulator of persister formation. Experimental evidence demonstrates that pnp knockout strains (Δpnp) exhibit significant defects in persistence to antibiotics and various stress conditions compared to wild-type strains. This persistence phenotype can be restored upon complementation with the pnp gene, confirming PNPase's direct involvement in this process .

To effectively investigate PNPase's role in persistence, researchers should consider the following experimental approaches:

  • Gene knockout and complementation studies: Constructing pnp deletion mutants and complemented strains allows for direct assessment of PNPase's contribution to persistence. This approach has successfully demonstrated that PNPase controls cellular metabolism by negatively regulating the global regulator cyclic AMP receptor protein (CRP) operon .

  • Transcriptomic analysis: RNA-seq analysis of Δpnp strains compared to wild-type reveals downstream effects of PNPase deletion. Previous studies identified 242 differentially expressed genes (166 upregulated and 76 downregulated) in Δpnp strains, with upregulated genes primarily mapping to metabolism and virulence pathways .

  • Post-transcriptional regulation studies: Investigating PNPase's interaction with the 5'-untranslated region (UTR) of target transcripts provides mechanistic insights. Reporter constructs with or without 5'-UTR regions (such as crp-lacZ) in various genetic backgrounds can demonstrate PNPase's regulatory function .

The table below summarizes key phenotypic differences observed between wild-type and Δpnp strains:

PhenotypeWild-typeΔpnp strainReference
Growth rateNormalSlightly reduced
Antibiotic persistenceNormalSignificantly defective
MotilityNormalDefective
Biofilm formationNormalEnhanced
CRP expressionBaseline3.22-fold increase

These experimental approaches collectively provide a comprehensive understanding of PNPase's role in bacterial persistence and stress response mechanisms.

What methodologies are most effective for studying the RNA degradation activity of R. baltica PNPase in vitro?

To effectively characterize the RNA degradation activity of R. baltica PNPase in vitro, researchers should employ a multi-faceted approach combining biochemical assays, structural analysis, and substrate specificity determination:

  • Phosphorolytic activity assays: The standard approach involves incubating purified PNPase with radiolabeled RNA substrates in the presence of inorganic phosphate. Reaction products can be analyzed by polyacrylamide gel electrophoresis (PAGE) followed by autoradiography. Quantitative analysis can be performed by measuring the release of nucleoside diphosphates using HPLC or coupled enzymatic assays.

  • Substrate specificity profiling: Systematically test PNPase activity against various RNA structures (single-stranded, stem-loops, double-stranded regions) and sequences to determine preference patterns. This can be complemented with competition assays using mixed substrate pools to identify high-affinity targets.

  • Kinetic characterization: Determine key kinetic parameters (Km, Vmax, kcat) under varying conditions (temperature, pH, ion concentrations) to establish the enzyme's optimal operating environment. This is particularly important for R. baltica PNPase given the marine environment of this organism, which may have adapted to function under specific salt concentrations.

  • RNA binding studies: Employ techniques such as electrophoretic mobility shift assays (EMSA), fluorescence anisotropy, or surface plasmon resonance (SPR) to characterize PNPase-RNA interactions independent of catalytic activity. This helps distinguish binding defects from catalytic defects.

  • Structure-function analysis: Combine site-directed mutagenesis with activity assays to identify critical residues for substrate recognition and catalysis. This approach is particularly valuable for comparative studies with PNPase from other bacterial species like E. coli where more extensive characterization exists.

When interpreting results, researchers should consider that PNPase activity can be modulated by various cellular factors and environmental conditions, which may explain differences observed between in vitro and in vivo functional studies.

How does R. baltica PNPase expression change throughout the organism's life cycle, and what techniques best capture these dynamics?

Rhodopirellula baltica exhibits a complex life cycle with distinct morphological phases, including swarmer cells, budding cells, single cells, and rosette formations. Understanding PNPase expression throughout this life cycle requires sophisticated temporal profiling approaches:

  • Whole genome microarray analysis: This technique has proven effective in monitoring gene expression throughout R. baltica's growth curve. Studies have shown that numerous genes, including those with potential biotechnological applications, are differentially regulated during different growth phases . While specific data for pnp expression is not directly reported in the current literature, this approach would effectively capture its expression dynamics.

  • Growth phase-specific sampling: For comprehensive profiling, samples should be collected at defined points: early exponential phase (dominated by swarmer and budding cells), transition phase (mixture of single cells, budding cells, and rosettes), and stationary phase (predominantly rosette formations) . This temporal resolution is critical for correlating PNPase expression with specific morphological transitions.

  • Quantitative RT-PCR: As a complementary approach to microarray analysis, qRT-PCR provides more sensitive quantification of pnp transcript levels across growth phases. Normalization to stable reference genes is essential for accurate comparison across time points.

  • Protein-level analysis: Western blotting or targeted proteomics (MRM/PRM) can verify whether transcriptional changes translate to altered protein levels. Previous proteome studies of R. baltica have successfully identified proteins that change in abundance throughout the life cycle .

  • Single-cell approaches: Given the heterogeneous nature of R. baltica cultures at different growth phases, techniques like single-cell RNA-seq or fluorescence in situ hybridization (FISH) could provide insights into cell-type specific expression patterns that might be masked in bulk analyses.

The metabolic adaptation observed during R. baltica's transition to stationary phase, including upregulation of stress response genes like glutathione peroxidase (RB2244), thioredoxin (RB12160), and universal stress protein (uspE, RB4742) , suggests that PNPase expression might also be modulated during this transition to support RNA turnover under stress conditions.

What approaches can be used to investigate interactions between PNPase and other components of the RNA degradation machinery in R. baltica?

Investigating PNPase interactions within the broader RNA degradation machinery requires techniques that can identify stable and transient protein-protein interactions while preserving functional associations:

  • Affinity purification coupled with mass spectrometry (AP-MS): Using tagged versions of PNPase expressed in R. baltica, researchers can isolate intact protein complexes and identify interaction partners through mass spectrometry. Both formaldehyde crosslinking (for capturing transient interactions) and native conditions should be employed to generate comprehensive interaction maps.

  • Bacterial two-hybrid system: This approach can verify specific binary interactions between PNPase and candidate partners identified through AP-MS. It's particularly useful for confirming direct physical interactions versus co-complex associations.

  • Co-immunoprecipitation (Co-IP) with RNA degradosome components: Based on known degradosome components in related bacteria, targeted Co-IP experiments can determine whether R. baltica PNPase forms similar complexes. Critical candidates include ribonuclease E (RNase E), RNA helicase (RhlB), and enolase, which form the core degradosome in many bacteria.

  • In vivo RNA immunoprecipitation (RIP): This technique can identify RNA substrates that are simultaneously bound by PNPase and other RNA-binding proteins, providing insights into cooperative RNA processing.

  • Microscopy-based approaches: Fluorescence resonance energy transfer (FRET) or bimolecular fluorescence complementation (BiFC) can visualize protein interactions in living cells, potentially revealing the subcellular localization of PNPase-containing complexes during different growth phases.

While specific information on R. baltica RNA degradosome composition is limited in the current literature, research in E. coli has demonstrated that PNPase interacts with the global regulator CRP at the post-transcriptional level, specifically targeting the 5'-UTR of the crp transcript . Similar regulatory mechanisms might exist in R. baltica, suggesting that interaction studies should extend beyond canonical RNA degradosome components to include potential regulatory partners.

How can researchers effectively use PNPase as a tool for studying bacterial persistence mechanisms?

PNPase's established role in bacterial persistence makes it a valuable tool for studying this clinically relevant phenomenon. Researchers can leverage PNPase in several ways:

  • As a genetic marker for persistence: Monitoring pnp expression levels can serve as an indicator of cells transitioning to a persister state. Constructing fluorescent reporters fused to the pnp promoter could enable real-time visualization of this transition at the single-cell level.

  • As a drug target validation system: Given that PNPase has been implicated as a target for the tuberculosis persister drug pyrazinamide, researchers can use R. baltica PNPase to validate and characterize new anti-persister compounds . This approach involves:

    • Enzyme inhibition assays with candidate compounds

    • Structural studies of PNPase-inhibitor complexes

    • Correlation of in vitro inhibition with effects on bacterial persistence

  • For comparative genomics approaches: By comparing PNPase sequence and function across diverse bacterial species, researchers can identify conserved mechanisms of persistence regulation. This comparative approach has revealed that PNPase negatively regulates the crp operon in E. coli , and similar regulatory targets might exist in R. baltica.

  • As a genetic engineering tool: Modulating PNPase levels through controlled expression systems allows researchers to manipulate persistence rates experimentally. This approach can help determine how persistence frequency impacts survival under various antibiotic treatment regimens.

The table below summarizes key experimental readouts when studying persistence using PNPase as a research tool:

Experimental ApproachReadout MeasurementsApplication
Gene expression profilingTranscriptome changes in Δpnp vs. wild-typeIdentifying PNPase-regulated pathways
Antibiotic tolerance assaysSurvival rates after antibiotic exposureQuantifying persistence phenotypes
Metabolic indicator analysisATP levels, membrane potentialAssessing metabolic state of persisters
Reporter systemsβ-galactosidase activity (crp-lacZ)Measuring regulatory effects

These approaches collectively enable researchers to dissect the molecular mechanisms underlying bacterial persistence and potentially develop new strategies to combat persistent infections.

What are the key considerations when designing experiments to study the role of PNPase in post-transcriptional regulation?

When investigating PNPase's role in post-transcriptional regulation, researchers should consider several critical experimental design factors:

  • Target transcript selection: Based on E. coli studies, PNPase regulates specific transcripts by interacting with their 5'-UTR regions. For R. baltica, researchers should:

    • Prioritize transcripts with structured 5'-UTRs

    • Focus on genes involved in stress response and metabolism

    • Consider global regulators (like crp in E. coli) as potential targets

  • Reporter system design: Construct reporter plasmids containing:

    • The native promoter of the target gene

    • Variants with and without the 5'-UTR region

    • A quantifiable reporter gene (e.g., lacZ, GFP)

    This approach has successfully demonstrated PNPase's regulatory effect on the crp operon in E. coli, where β-galactosidase activity was 8.3-fold higher in the Δpnp strain compared to wild-type when the 5'-UTR was present .

  • Growth phase considerations: R. baltica exhibits distinct morphological phases throughout its growth cycle . Experiments should include:

    • Sampling across multiple growth phases (early exponential, transition, stationary)

    • Correlation of regulatory effects with cell morphology changes

    • Assessment under different stress conditions to capture condition-specific regulation

  • RNA stability measurements: To distinguish direct effects on RNA stability from translational effects:

    • Perform RNA half-life measurements using rifampicin to inhibit transcription

    • Compare decay rates of target transcripts in wild-type versus Δpnp strains

    • Use northern blotting or qRT-PCR for quantification

  • Direct binding assays: Confirm direct PNPase-RNA interactions using:

    • RNA electrophoretic mobility shift assays (EMSA)

    • UV crosslinking followed by immunoprecipitation

    • In vitro structure probing to identify PNPase binding sites

  • Integration with global analyses: Combine targeted approaches with:

    • Transcriptome-wide analyses (RNA-seq) of wild-type versus Δpnp strains

    • Ribosome profiling to assess translational effects

    • Metabolomic profiling to identify downstream consequences

These methodological considerations will help researchers develop robust experimental designs that can effectively characterize PNPase's role in post-transcriptional regulation while accounting for the unique biological characteristics of R. baltica.

What technical challenges might researchers encounter when working with R. baltica PNPase and how can these be addressed?

Researchers working with R. baltica PNPase may encounter several technical challenges that require specific troubleshooting approaches:

  • Expression and solubility issues:

    • Challenge: As an enzyme from a marine organism, R. baltica PNPase may show suboptimal folding in standard expression systems.

    • Solution: Optimize expression conditions (temperature, induction parameters) and consider marine-mimicking buffer systems containing appropriate salt concentrations. Expression trials comparing multiple host systems (E. coli, yeast, baculovirus, and mammalian cells) are recommended to identify optimal conditions for obtaining soluble, active enzyme .

  • Functional characterization in heterologous systems:

    • Challenge: PNPase function may depend on specific RNA degradosome components or cellular factors that differ between R. baltica and model organisms.

    • Solution: Complement functional studies in heterologous systems with in vitro reconstitution using purified components. Consider creating chimeric systems where R. baltica PNPase is expressed alongside its native interacting partners.

  • Growth and cultivation of R. baltica:

    • Challenge: R. baltica exhibits complex growth phases with different cell morphologies , making consistent sample collection challenging.

    • Solution: Establish standardized growth protocols with clear markers for different growth phases. Monitor culture morphology through microscopy to confirm developmental stage before sampling.

  • RNA substrate specificity:

    • Challenge: Determining physiologically relevant RNA targets for R. baltica PNPase.

    • Solution: Combine computational prediction of structured RNA regions in the R. baltica transcriptome with experimental validation using techniques like CLIP-seq (crosslinking immunoprecipitation followed by sequencing) to identify direct PNPase binding sites.

  • Distinguishing direct from indirect effects:

    • Challenge: PNPase deletion affects multiple cellular processes, making it difficult to identify direct regulatory targets.

    • Solution: Use complementary approaches, including:

      • Controlled expression systems to perform time-course induction experiments

      • Catalytically inactive PNPase mutants to separate binding from degradation effects

      • Targeted analysis of specific transcripts using reporter constructs with defined regulatory elements

  • Structural characterization:

    • Challenge: Obtaining structural information for R. baltica PNPase.

    • Solution: Consider cryo-electron microscopy as an alternative to crystallography, particularly if the protein proves difficult to crystallize. Focus on complex structures with RNA substrates to gain functional insights.

By anticipating these challenges and implementing appropriate methodological strategies, researchers can effectively navigate the technical difficulties associated with studying this complex enzyme from a marine bacterium with unique cellular characteristics.

How does R. baltica PNPase compare to homologous enzymes from other bacterial species?

Comparative analysis of PNPase across bacterial species reveals both conserved features and species-specific adaptations that reflect diverse ecological niches and regulatory networks:

  • Structural conservation: PNPase typically contains core domains that are highly conserved across bacteria:

    • Two RNase PH domains with phosphorolytic activity

    • S1 and KH RNA-binding domains

    • While sequence-level comparison data specific to R. baltica PNPase is limited in the provided sources, functional conservation suggests structural similarities with well-characterized homologs

  • Regulatory contexts: Notable differences exist in how PNPase is integrated into regulatory networks:

    • In E. coli, PNPase negatively regulates the global regulator CRP by targeting the 5'-UTR of crp mRNA

    • In R. baltica, the specific regulatory targets remain to be fully characterized, but the organism's unique lifestyle as a marine bacterium with complex morphological transitions suggests potentially distinct regulatory roles

  • Physiological roles: PNPase contributions to stress response and persistence appear to be conserved but with species-specific manifestations:

    • E. coli Δpnp strains show defects in antibiotic persistence, motility, and altered biofilm formation

    • The role of PNPase in R. baltica's adaptation to marine environments and its complex life cycle represents an important area for future investigation

  • Degradosome associations: In many bacteria, PNPase functions as part of a multi-enzyme RNA degradosome complex:

    • In E. coli, PNPase associates with RNase E, RNA helicase RhlB, and enolase

    • The composition of potential RNA degradosome complexes in R. baltica remains to be characterized

    • The planctomycete-specific cell compartmentalization may impact degradosome localization and function

  • Environmental adaptations: As a marine organism, R. baltica PNPase likely exhibits adaptations to function optimally in its native environment:

    • Potential salt tolerance mechanisms

    • Adaptation to the temperature range of marine habitats

    • Possible roles in responses to marine-specific stressors

Comparative functional studies that express R. baltica PNPase in model organisms like E. coli, alongside reciprocal experiments with E. coli PNPase expressed in R. baltica, would provide valuable insights into both conserved functions and species-specific adaptations of this important enzyme.

What are the emerging applications of R. baltica PNPase in biotechnology and therapeutic development?

The unique properties of R. baltica PNPase, combined with its role in fundamental cellular processes, position it for several emerging biotechnological and therapeutic applications:

  • Anti-persister drug development:

    • PNPase's critical role in bacterial persistence mechanisms makes it a promising target for developing novel antimicrobials

    • The tuberculosis drug pyrazinamide has implicated PNPase as a target, suggesting similar approaches could be effective against other persistent infections

    • Structure-based drug design targeting R. baltica PNPase could yield compounds effective against marine pathogens or serve as models for targeting homologous enzymes in clinical pathogens

  • RNA processing tools:

    • The phosphorolytic activity of PNPase can be harnessed for controlled RNA degradation in vitro

    • Engineered variants with altered substrate specificity could serve as tools for selective RNA processing

    • Applications in RNA sample preparation for next-generation sequencing and other analytical techniques

  • Biosensors for environmental monitoring:

    • PNPase-based reporter systems could detect marine pollutants that affect RNA metabolism

    • The enzyme's natural adaptation to marine conditions makes R. baltica PNPase particularly suitable for developing robust biosensors for aquatic environments

  • Biofilm control strategies:

    • The observation that PNPase deletion affects biofilm formation suggests potential applications in controlling microbial colonization

    • Engineering PNPase activity could provide novel approaches to modulate biofilm development in both beneficial and harmful microbial communities

  • Synthetic biology applications:

    • Incorporating R. baltica PNPase into synthetic gene circuits could enable post-transcriptional regulation in engineered biological systems

    • The enzyme could serve as a tunable component in feedback loops controlling gene expression dynamics

  • Protein engineering platform:

    • The adaptation of R. baltica to marine environments suggests its PNPase may have unique stability properties

    • These features could be leveraged to develop engineered proteins with enhanced stability under challenging conditions

These applications extend beyond the direct use of R. baltica PNPase itself to include insights gained from studying its structure, function, and regulation that can be applied to engineering other biological systems or developing targeted interventions for clinical applications.

What experimental approaches can elucidate the role of PNPase in R. baltica's adaptation to its marine environment?

Investigating PNPase's contribution to R. baltica's marine adaptation requires integrating molecular techniques with environmental simulation approaches:

  • Comparative salt tolerance studies:

    • Challenge wild-type and Δpnp R. baltica strains with varying salinity levels to assess survival and growth

    • Measure PNPase expression and activity across salinity gradients

    • Analyze salt-dependent changes in RNA degradation patterns and target specificity

  • Temperature adaptation analysis:

    • Characterize enzymatic properties of purified R. baltica PNPase across temperature ranges typical of its marine habitat

    • Compare cold sensitivity of wild-type versus Δpnp strains, as PNPase often plays crucial roles in cold adaptation

    • Evaluate transcriptome-wide effects of PNPase deletion under different temperature regimes

  • Environmental stress response profiling:

    • Expose cultures to marine-relevant stressors (UV radiation, nutrient limitation, pollutants)

    • Monitor pnp expression dynamics during stress exposure and recovery

    • Correlate PNPase activity with R. baltica's morphological transitions under stress conditions

  • Biofilm and attachment studies:

    • Quantify the effects of PNPase deletion on attachment to surfaces mimicking marine substrates

    • Analyze biofilm architecture and extracellular matrix composition in wild-type versus Δpnp strains

    • Investigate the role of PNPase in rosette formation, a characteristic feature of R. baltica's life cycle

  • Transcriptome and RNA stability analysis under marine conditions:

    • Perform RNA-seq comparing wild-type and Δpnp strains under simulated marine conditions

    • Measure half-lives of stress-responsive transcripts to identify PNPase-dependent stability regulation

    • Focus on genes involved in osmotic regulation, nutrient acquisition, and cell wall remodeling

  • In situ validation approaches:

    • Develop reporter systems to monitor PNPase activity in natural marine samples

    • Use fluorescence microscopy to localize PNPase within R. baltica cells during different life cycle stages

    • Correlate PNPase expression with environmental parameters in natural habitats

The table below summarizes key experimental parameters for investigating PNPase's role in marine adaptation:

Environmental ParameterExperimental RangeMeasured OutcomesRelevance to Marine Adaptation
Salinity0.5-3.5% NaClGrowth rate, enzyme activity, RNA profilesOsmotic stress response
Temperature4-30°CCold sensitivity, RNA degradation kineticsSeasonal temperature fluctuation
Nutrient availabilityC:N:P ratiosPersister formation, life cycle transitionsAdaptation to nutrient limitation
UV exposure280-400 nmDNA repair efficiency, stress gene expressionSurface water adaptation

These experimental approaches would provide comprehensive insights into how PNPase contributes to R. baltica's remarkable adaptation to its marine niche while potentially revealing novel mechanisms of post-transcriptional regulation in environmental adaptation.

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.