Recombinant Geobacter sulfurreducens 30S ribosomal protein S4 (rpsD)

Shipped with Ice Packs
In Stock

Description

Introduction to Recombinant Geobacter sulfurreducens 30S Ribosomal Protein S4 (rpsD)

Recombinant Geobacter sulfurreducens 30S ribosomal protein S4 (rpsD) is a crucial component of the ribosomal machinery in the bacterium Geobacter sulfurreducens, which is known for its unique metabolic capabilities, particularly in the reduction of metals and extracellular electron transfer. This protein plays an essential role in the assembly and function of the ribosome, facilitating protein synthesis.

Overview of Geobacter sulfurreducens

Geobacter sulfurreducens is a gram-negative bacterium that thrives in anaerobic environments, utilizing metals as electron acceptors. It has garnered significant attention due to its potential applications in bioremediation and bioenergy production, particularly in microbial fuel cells. The organism's ability to transfer electrons to insoluble metal oxides and electrodes makes it an important model for studying bioelectrochemical systems.

Structure and Function of rpsD

Molecular Structure

The rpsD gene encodes the 30S ribosomal protein S4, which is part of the small subunit of the ribosome. This protein is integral to the structure of the ribosome, contributing to its stability and function during translation. The protein's structure typically includes several conserved domains that are critical for its interaction with ribosomal RNA and other ribosomal proteins.

Role in Protein Synthesis

Ribosomal protein S4 is involved in maintaining the proper conformation of the ribosome, which is essential for accurate translation of mRNA into proteins. It interacts with ribosomal RNA, stabilizing the structure necessary for peptide bond formation.

Recombinant Production of rpsD

Expression Systems

Recombinant production of rpsD can be achieved using various expression systems, including bacterial systems such as Escherichia coli. The gene can be cloned into expression vectors that facilitate high-level production and subsequent purification of the protein.

Purification Techniques

Purification methods often include affinity chromatography, where tags such as His-tags are used to isolate the recombinant protein from other cellular components. Following purification, techniques like SDS-PAGE can be employed to confirm the identity and purity of rpsD.

Research Findings on rpsD

Recent studies have focused on understanding the functional implications of rpsD within Geobacter sulfurreducens. Key findings include:

  • Gene Expression Analysis: Transcriptomic studies indicate that rpsD expression is regulated under different environmental conditions, reflecting its role in adapting to changes in nutrient availability and electron acceptors .

  • Functional Studies: Knockout experiments have shown that disruption of rpsD affects growth rates and protein synthesis efficiency, underscoring its importance in cellular metabolism .

  • Interactions with Other Proteins: Research has highlighted potential interactions between rpsD and other ribosomal proteins, suggesting a cooperative role in ribosome assembly and function .

Data Table: Characteristics of Recombinant Geobacter sulfurreducens 30S Ribosomal Protein S4

CharacteristicDescription
OrganismGeobacter sulfurreducens
Gene NamerpsD
Protein TypeRibosomal Protein S4
Molecular WeightApproximately 15 kDa
RoleEssential for ribosome assembly and function
Expression SystemEscherichia coli (commonly used)
Purification MethodAffinity chromatography

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 preparation.

Lead Time
Delivery times vary depending on the purchasing method and location. Please consult your local distributor for precise delivery estimates.

Note: Standard shipping includes 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 collect the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquotting for long-term storage at -20°C/-80°C. Our standard glycerol concentration is 50% and may serve as a reference.
Shelf Life
Shelf life depends on various factors, including storage conditions, buffer composition, temperature, and the protein's inherent 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
Upon receipt, store at -20°C/-80°C. Aliquot 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. To specify a tag type, please inform us; we will prioritize its development.

Synonyms
rpsD; GSU283230S ribosomal protein S4
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-208
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Geobacter sulfurreducens (strain ATCC 51573 / DSM 12127 / PCA)
Target Names
rpsD
Target Protein Sequence
MARYTGPSCR LCRRENMELF LKGERCYTDK CAIKRRNYPP GQHGQGRPKV SNYGVQLREK QKVRRIYGIL EKQFRSYFQE ADRLKGVTGE NLLSLLERRL DNVVYRLGFA ASRTEARILV RHNHFTLNGK KANIPSIQLR AGDVVELKEK SRKIACINES LDAVVRRGIP QWLELEKDAY KGVVKTLPVR EDITMPIQEQ LIVELYSK
Uniprot No.

Target Background

Function

A key rRNA-binding protein, it directly binds to 16S rRNA, initiating the assembly of the 30S ribosomal subunit. In conjunction with S5 and S12, it plays a crucial role in translational accuracy.

Database Links

KEGG: gsu:GSU2832

STRING: 243231.GSU2832

Protein Families
Universal ribosomal protein uS4 family

Q&A

What is the role of ribosomal protein S4 (rpsD) in Geobacter sulfurreducens?

Ribosomal protein S4 (rpsD) in Geobacter sulfurreducens is a critical component of the 30S ribosomal subunit, serving essential functions in ribosome assembly and translation. As in other bacteria, S4 likely acts as a primary binding protein that interacts directly with 16S rRNA during the early stages of 30S subunit assembly. This protein helps establish the proper conformation of the 16S rRNA, particularly in the 5' domain, and creates binding sites for subsequent ribosomal proteins. In ribosome biogenesis, S4 functions similarly to other primary binding r-proteins by promoting long-range tertiary structure in the ribosomal RNA .

What are the recommended methods for cloning the rpsD gene from G. sulfurreducens?

For cloning the rpsD gene from G. sulfurreducens, researchers should:

  • Extract genomic DNA using specialized protocols for G. sulfurreducens, considering its Gram-negative cell wall structure

  • Design PCR primers based on the genomic sequence of G. sulfurreducens (GenBank accession number available in the complete genome sequence)

  • Include appropriate restriction sites in primers for subsequent cloning

  • Optimize PCR conditions for G. sulfurreducens' high GC content

  • Clone the amplified gene into an expression vector with an appropriate tag (His-tag is commonly used)

  • Verify the sequence to ensure no mutations were introduced during amplification

This approach should be adapted based on the genetic system available for G. sulfurreducens, which includes tools for gene manipulation and expression .

What are the optimal conditions for expressing recombinant G. sulfurreducens rpsD in E. coli?

For optimal expression of recombinant G. sulfurreducens rpsD in E. coli:

  • Select an appropriate E. coli strain (BL21(DE3) or Rosetta for potential rare codon issues)

  • Use a vector with a strong, inducible promoter (T7 or tac)

  • Include a His-tag or other affinity tag for purification

  • Grow cultures at 30°C rather than 37°C to enhance soluble protein production

  • Induce with 0.1-0.5 mM IPTG at mid-log phase (OD600 = 0.6-0.8)

  • Continue expression for 4-6 hours, or overnight at 18°C for reduced inclusion body formation

  • Monitor expression by SDS-PAGE

The expression conditions should be optimized experimentally, as G. sulfurreducens proteins may have specific requirements different from those of model organisms like E. coli. Researchers should consider the effects of growth temperature, inducer concentration, and duration of induction on both yield and solubility.

What purification strategy is most effective for obtaining high-purity rpsD protein?

The most effective purification strategy for obtaining high-purity rpsD protein involves:

Table 1: Multi-step Purification Strategy for Recombinant rpsD

StepMethodBuffer CompositionPurpose
1Immobilized metal affinity chromatography (IMAC)50 mM Tris-HCl pH 8.0, 300 mM NaCl, 5-250 mM imidazole gradientInitial capture of His-tagged protein
2Ion exchange chromatography20 mM Tris-HCl pH 8.0, 50-500 mM NaCl gradientSeparation based on charge properties
3Size exclusion chromatography20 mM Tris-HCl pH 7.5, 150 mM NaCl, 1 mM DTTFinal polishing step, assessment of oligomeric state
4Buffer exchange/dialysis20 mM HEPES pH 7.5, 100 mM KCl, 10 mM MgCl2, 5% glycerolPreparation for functional studies

Each purification step should be validated by SDS-PAGE analysis and, if necessary, Western blotting using antibodies against the purification tag or the protein itself. The final product should be assessed for purity by mass spectrometry and for proper folding by circular dichroism spectroscopy.

How can I troubleshoot issues with rpsD solubility during recombinant expression?

When encountering solubility issues with recombinant rpsD:

  • Modify expression conditions: Lower the temperature to 18-20°C during induction and reduce inducer concentration to decrease expression rate and improve folding.

  • Use solubility enhancing tags: Consider fusion partners like SUMO, MBP, or GST that can enhance solubility.

  • Adjust buffer composition: Include stabilizing agents in lysis buffers:

    • 5-10% glycerol

    • 0.1-0.5% non-ionic detergents (Triton X-100, NP-40)

    • 50-300 mM NaCl to shield ionic interactions

    • 1-5 mM reducing agents (DTT or β-mercaptoethanol)

  • Co-expression with chaperones: Express rpsD alongside molecular chaperones like GroEL/GroES to assist proper folding.

  • Adjust cell lysis conditions: Use gentler lysis methods like freeze-thaw cycles or enzymatic lysis rather than sonication.

If solubility issues persist, consider adapting purification methods from the study of other ribosomal proteins like those characterized in the 30S ribosome assembly process .

What techniques are most appropriate for studying the RNA-binding properties of G. sulfurreducens rpsD?

For studying RNA-binding properties of G. sulfurreducens rpsD, several complementary approaches should be employed:

  • Electrophoretic Mobility Shift Assays (EMSA): To determine binding affinities between purified rpsD and synthetic 16S rRNA fragments.

  • Fluorescence Anisotropy: For quantitative measurement of binding constants using fluorescently labeled RNA.

  • Surface Plasmon Resonance (SPR): To analyze real-time binding kinetics and measure association/dissociation rates.

  • RNA Footprinting: Using hydroxyl radical probing similar to methods described for RsgA interaction with 30S subunits to identify specific nucleotides protected by rpsD binding .

  • Microscale Thermophoresis (MST): For detecting interactions in solution with minimal sample consumption.

  • X-ray Crystallography or Cryo-EM: To determine the three-dimensional structure of rpsD-RNA complexes at atomic resolution, similar to structural studies conducted for 30S ribosomal assembly factors .

These methods should be complemented with mutational analysis to identify critical residues involved in RNA binding.

How does rpsD from G. sulfurreducens participate in 30S ribosomal subunit assembly?

The role of rpsD in G. sulfurreducens 30S ribosomal subunit assembly likely follows the general principles established in other bacteria:

  • Primary Binding Protein: As one of the early binding r-proteins in the 30S assembly map (similar to the Nomura assembly map), rpsD likely binds directly to 16S rRNA independently of other proteins .

  • Conformational Stabilization: Upon binding, rpsD induces conformational changes in 16S rRNA that create binding sites for secondary binding proteins.

  • Assembly Kinetics: Like other r-proteins, rpsD binding rates would be expected to correspond to its position in the assembly order, as demonstrated in pulse-labeling based quantitative mass-spectrometry experiments for other bacterial systems .

  • Coordination with Assembly Factors: The assembly process likely involves coordination with GTPase assembly factors like RsgA, which has been shown to play crucial roles in validating the architecture of the 30S decoding center .

  • Prevention of Kinetic Traps: Proper binding of rpsD helps prevent the accumulation of misfolded intermediate states of low free energy that would slowly convert into mature 30S subunits .

In G. sulfurreducens specifically, the 30S assembly process may have adaptations related to the organism's unique environmental niche and metabolism, including its ability to reduce Fe(III) and survive in anaerobic sedimentary environments .

How can we study the impact of rpsD mutations on G. sulfurreducens ribosome function and cellular physiology?

To study the impact of rpsD mutations on G. sulfurreducens ribosome function and cellular physiology:

  • Site-Directed Mutagenesis: Generate specific mutations in conserved residues of rpsD based on structural predictions and sequence alignments.

  • Genetic System Application: Utilize the available genetic system for G. sulfurreducens to introduce mutations into the chromosomal copy of rpsD .

  • Phenotypic Analysis:

    • Growth rate measurements under various conditions (different electron acceptors, temperatures, pH)

    • Fe(III) reduction rates for both soluble and insoluble forms

    • Survival during oxygen exposure and stationary phase

    • Protein synthesis rates using radioactive amino acid incorporation

  • Ribosome Profiling: Analyze the impact of mutations on global translation patterns and potential ribosome stalling sites.

  • 30S Assembly Analysis: Examine whether mutations affect ribosome assembly using sucrose gradient centrifugation and quantification of free subunits versus 70S ribosomes.

  • In vitro Translation Assays: Assess the functional capacity of mutant ribosomes using reconstituted translation systems.

  • Interactome Analysis: Use pull-down assays to identify altered interactions between mutant rpsD and other ribosomal components or assembly factors.

These approaches would provide comprehensive insights into how specific rpsD residues contribute to ribosome function and cellular fitness in G. sulfurreducens.

What approaches can be used to investigate interactions between rpsD and ribosome assembly factors in G. sulfurreducens?

To investigate interactions between rpsD and ribosome assembly factors in G. sulfurreducens:

  • Co-Immunoprecipitation (Co-IP): Using antibodies against rpsD or epitope-tagged versions to pull down associated assembly factors.

  • Chemical Cross-Linking Coupled with Mass Spectrometry (XL-MS): To capture transient interactions between rpsD and assembly factors like RsgA that may be involved in 30S maturation .

  • Cryo-EM Analysis: To visualize assembly intermediates containing rpsD and associated factors, similar to the techniques used to elucidate RsgA's mechanism of action in 30S decoding center maturation .

  • Genetic Suppressor Screens: Identify mutations in assembly factors that compensate for defects in rpsD mutants.

  • Bacterial Two-Hybrid Assays: To test direct interactions between rpsD and candidate assembly factors in vivo.

  • Surface Plasmon Resonance (SPR): For quantitative measurement of binding affinities between purified rpsD and assembly factors.

  • Hydroxyl Radical Probing: To map interaction interfaces between rpsD and assembly factors on the 30S subunit at nucleotide resolution .

  • Fluorescence Resonance Energy Transfer (FRET): Using fluorescently labeled rpsD and assembly factors to monitor interactions in real-time.

These complementary approaches would provide insights into the network of interactions that coordinate 30S assembly in G. sulfurreducens.

A.3. How can rpsD be used as a tool to study ribosome specialization in G. sulfurreducens compared to other bacteria?

Using rpsD as a tool to study ribosome specialization in G. sulfurreducens requires comparative approaches:

  • Sequence and Structure Comparison: Detailed bioinformatic analysis of rpsD sequences across bacterial species, with particular focus on:

    • Unique sequence features in Geobacteraceae

    • Adaptive changes related to lifestyle in anaerobic sedimentary environments

    • Structural predictions of G. sulfurreducens-specific elements

  • Heterologous Complementation Studies: Determine if rpsD from G. sulfurreducens can functionally replace its ortholog in model organisms like E. coli, and vice versa.

  • Domain Swapping Experiments: Create chimeric rpsD proteins with domains from different bacteria to identify regions responsible for species-specific functions.

  • Ribosome Profiling Comparison: Compare translation efficiency and accuracy across different bacterial species using identical reporter constructs.

  • Environmental Adaptation Analysis: Test how rpsD variants from different bacteria perform under conditions relevant to G. sulfurreducens ecology:

    • Fe(III) reduction conditions

    • Oxygen stress scenarios

    • Different electron acceptors

    • Various temperatures and pH values

  • Specialized Ribosome Engineering: Use G. sulfurreducens rpsD features to create specialized ribosomes with altered translation properties in model organisms.

This comparative approach would reveal how ribosomal components have evolved to support G. sulfurreducens' specialized metabolic functions, including its central role in Fe(III) reduction in sedimentary environments .

How should controls be designed for experiments involving recombinant G. sulfurreducens rpsD?

Proper experimental controls for recombinant G. sulfurreducens rpsD studies should include:

  • Expression Controls:

    • Empty vector control (same vector without rpsD gene)

    • Expression of a known, well-behaved protein using the same system

    • Wild-type rpsD expression alongside any mutant versions

  • Purification Controls:

    • Process control (applying the purification protocol to cells not expressing the target protein)

    • Stability control (testing purified protein stability under experimental conditions over time)

    • Tag-only control (expressing and purifying the tag alone)

  • Functional Assay Controls:

    • Known functional ribosomal protein S4 from a model organism (E. coli)

    • Heat-denatured rpsD to confirm activity requires proper folding

    • RNA binding assays: non-specific RNA controls and competition assays

  • Structural Analysis Controls:

    • Circular dichroism measurements before and after storage

    • Dynamic light scattering to confirm monodispersity

    • Mass spectrometry verification of protein integrity

  • In vivo Studies Controls:

    • Complementation with native G. sulfurreducens rpsD

    • Empty vector controls for phenotypic studies

    • Wild-type G. sulfurreducens strain alongside any mutants

These controls help distinguish specific effects from artifacts and provide benchmarks for evaluating experimental outcomes.

What are the critical parameters to consider when designing experiments to study rpsD interactions with 16S rRNA?

When designing experiments to study rpsD interactions with 16S rRNA, researchers should consider these critical parameters:

  • RNA Preparation Quality:

    • Ensure RNA is properly folded in biologically relevant conformations

    • Minimize contamination with RNases

    • Consider using synthetic RNA fragments representing specific 16S rRNA domains

    • Include controls with non-specific RNA

  • Buffer Composition:

    • Mg²⁺ concentration (critical for RNA structure)

    • Monovalent ion concentration (K⁺, Na⁺)

    • pH consistent with physiological conditions for G. sulfurreducens

    • Presence of molecular crowding agents to mimic cellular conditions

  • Temperature:

    • Perform experiments at temperatures relevant to G. sulfurreducens ecology

    • Consider temperature gradients to assess binding thermodynamics

  • Protein:RNA Ratio:

    • Titrate across a wide range of molar ratios

    • Consider cooperative binding effects

  • Experimental Approach Selection:

    • For kinetic studies: stopped-flow fluorescence, SPR

    • For structural studies: footprinting, X-ray crystallography, Cryo-EM

    • For binding affinity: EMSA, fluorescence anisotropy, ITC

  • Competition Assays:

    • Include other 30S proteins to assess hierarchy and cooperativity in binding

    • Test with assembly factors like RsgA that might influence rpsD-RNA interactions

  • Analysis of Binding Specificity:

    • Compare binding to cognate versus non-cognate RNA sequences

    • Map binding sites using deletion/mutation analysis of RNA

These parameters should be systematically optimized to ensure reproducible and physiologically relevant results.

How can researchers effectively integrate structural and functional data for a comprehensive understanding of G. sulfurreducens rpsD?

To effectively integrate structural and functional data for a comprehensive understanding of G. sulfurreducens rpsD:

  • Structure-Function Correlation Pipeline:

    • Obtain high-resolution structural data (X-ray crystallography or Cryo-EM)

    • Identify conserved and variable regions through comparative analysis

    • Design targeted mutations based on structural features

    • Test mutant proteins in functional assays

    • Develop computational models to predict impacts of additional mutations

  • Multi-scale Experimental Approach:

    • Atomic level: High-resolution structural studies of isolated rpsD

    • Molecular level: rpsD-RNA and rpsD-protein interaction studies

    • Ribosome level: Integration into 30S subunit and effects on assembly

    • Cellular level: Impact on translation and growth phenotypes

    • Ecological level: Performance under environmentally relevant conditions

  • Integrative Data Analysis Strategies:

    • Use molecular dynamics simulations to connect static structures to dynamic function

    • Apply machine learning approaches to identify patterns across diverse datasets

    • Develop network models of rpsD interactions within the ribosome assembly pathway

  • Visualization and Modeling Tools:

    • Generate interactive 3D models incorporating both structural and functional data

    • Use color-coding to represent functional importance of different regions

    • Create mutation-sensitivity maps based on combined structural and functional assays

  • Collaborative Cross-disciplinary Analysis:

    • Integrate expertise from structural biologists, biochemists, microbiologists, and computational scientists

    • Use standardized data formats to facilitate sharing and integration

    • Develop custom databases for G. sulfurreducens ribosomal components

This integrated approach provides a comprehensive understanding of how rpsD structure determines its function in ribosome assembly and cellular physiology, particularly in the context of G. sulfurreducens' unique metabolism involving Fe(III) reduction and survival in anaerobic environments .

How should researchers interpret contradictory results in rpsD interaction studies?

When facing contradictory results in rpsD interaction studies:

  • Systematic Variation Analysis:

    • Compare experimental conditions across contradictory results

    • Identify differences in buffer composition, especially Mg²⁺ concentration

    • Evaluate protein and RNA preparation methods for potential differences

    • Assess experimental temperatures and incubation times

  • Multiple Method Validation:

    • Apply complementary techniques to the same interaction question

    • For example, if EMSA and SPR give contradictory results, add ITC or fluorescence anisotropy

    • Prioritize methods that preserve native conditions

  • Context Dependency Evaluation:

    • Test if the presence of other ribosomal components affects the interaction

    • Consider if full-length 16S rRNA versus fragments gives different results

    • Assess whether post-translational modifications affect interactions

  • Kinetic versus Equilibrium Considerations:

    • Determine if contradictions arise from measuring different stages of the interaction

    • Distinguish between initial binding events and stable complex formation

    • Consider time-resolved measurements to capture the complete interaction profile

  • Biological Relevance Framework:

    • Evaluate which experimental system better reflects in vivo conditions

    • Consider the physiological environment of G. sulfurreducens

    • Validate with in vivo approaches when possible

  • Statistical Robustness Assessment:

    • Ensure sufficient replication across independent experiments

    • Apply appropriate statistical tests to determine significance of differences

    • Consider Bayesian approaches to integrate contradictory data

This systematic approach helps resolve contradictions and develops a more nuanced understanding of context-dependent interactions involving rpsD.

What statistical approaches are most appropriate for analyzing data from rpsD functional studies?

For analyzing data from rpsD functional studies, appropriate statistical approaches include:

Table 2: Statistical Methods for Different rpsD Experimental Data Types

Data TypeRecommended Statistical ApproachImplementation Notes
Binding assays (EMSA, SPR)Non-linear regression (Hill equation or similar)Use global fitting when possible; compare binding models with AIC or BIC
Growth curves with rpsD mutantsMixed-effects models or repeated measures ANOVAAccount for batch effects; use appropriate post-hoc tests for multiple comparisons
Fe(III) reduction rate assaysLinear regression for initial rates; non-linear regression for complete time coursesTransform data if necessary to meet assumptions; use residual analysis
Survival assays (stationary phase, oxygen)Kaplan-Meier survival analysis; log-rank test for comparisonsConsider competing risks when multiple stress factors are present
Ribosome assembly kineticsMulti-phase association/dissociation modelsUse AIC to determine optimal number of phases; bootstrap for confidence intervals
Proteomics/RNA-seqDESeq2 or limma for differential expression; GSEA for pathway analysisControl for multiple testing; consider biological replicates carefully
Structure-function correlationsPrincipal component analysis; partial least squaresValidate with cross-validation; consider dimensionality reduction

When designing experiments:

  • Calculate appropriate sample sizes using power analysis

  • Include biological replicates (different protein/RNA preparations)

  • Plan for technical replicates to assess methodological variation

  • Consider factorial designs to test interactions between variables

  • Include positive and negative controls in all statistical analyses

For complex datasets, consider consulting with a biostatistician to ensure appropriate experimental design and analysis.

How can researchers effectively compare rpsD from G. sulfurreducens with orthologs from other bacterial species?

To effectively compare rpsD from G. sulfurreducens with orthologs from other bacterial species:

  • Sequence-Based Comparative Analysis:

    • Multiple sequence alignment with diverse bacterial S4 proteins

    • Phylogenetic tree construction to visualize evolutionary relationships

    • Conservation analysis to identify G. sulfurreducens-specific features

    • Identification of selection signatures using dN/dS ratio analysis

    • Coevolution analysis with interacting partners (16S rRNA, other r-proteins)

  • Structural Comparison Approaches:

    • Homology modeling based on existing S4 structures

    • Superposition of structures to identify conformational differences

    • Electrostatic surface potential comparison

    • Molecular dynamics simulations to compare dynamic properties

    • Binding pocket analysis for RNA interaction sites

  • Functional Comparative Methods:

    • Heterologous complementation assays in model organisms

    • In vitro translation efficiency comparison using identical templates

    • Thermal stability comparison across orthologs

    • RNA binding specificity and affinity measurements

    • Assembly kinetics into 30S subunits

  • Ecological Context Integration:

    • Correlation of sequence/structural features with bacterial lifestyle

    • Comparison across bacteria with similar metabolic capabilities (other Fe(III)-reducers)

    • Analysis of rpsD adaptations in extremophiles vs. mesophiles

    • Examination of rpsD conservation in obligate vs. facultative anaerobes

  • Visualization and Analysis Tools:

    • ConSurf server for evolutionary conservation mapping onto structure

    • DALI server for structural similarity searches

    • Interactive phylogenetic trees with mapped functional/structural data

    • Network visualization of interaction partners across species

This multi-faceted comparative approach reveals how rpsD has evolved to support the unique physiology of G. sulfurreducens, particularly its adaptation to environments where Fe(III) reduction is important .

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.