Recombinant Enterococcus faecalis 30S ribosomal protein S4 (rpsD)

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference 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 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. 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: 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 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 implementation.
Synonyms
rpsD; EF_3070; 30S ribosomal protein S4
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-203
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Enterococcus faecalis (strain ATCC 700802 / V583)
Target Names
rpsD
Target Protein Sequence
MSRYTGPSWK VSRRLGISLS GTGKELARRP YKPGQHGPNS RGKVSEYGMQ LTEKQKLRHM YGMNERQFRT LFIKASKIKE GKHGVNFMVL LEQRLDNVVY RLGLATTRRQ ARQLVNHGHI TVDGKRVDIP SYHVEVGQVI GVREKSQNIS TIKEAVEATV GRPAFVSFDT EKLEGSFTRL PERDELYPEI DEALVVEYYN QKL
Uniprot No.

Target Background

Function

A key rRNA-binding protein, it directly interacts with 16S rRNA, initiating the assembly of the 30S ribosomal subunit body. In conjunction with proteins S5 and S12, it plays a critical role in maintaining translational accuracy.

Database Links

KEGG: efa:EF3070

STRING: 226185.EF3070

Protein Families
Universal ribosomal protein uS4 family

Q&A

What is the biological significance of 30S ribosomal protein S4 (rpsD) in Enterococcus faecalis?

The 30S ribosomal protein S4 (rpsD) plays a critical role in the assembly and function of the 30S ribosomal subunit in Enterococcus faecalis. As a primary binding protein, it initiates the nucleation of the 30S ribosomal subunit assembly by binding to the 16S ribosomal RNA. This protein is essential for proper ribosome formation and subsequent protein synthesis. In E. faecalis, which can cause serious hospital-acquired infections, rpsD is particularly important as it contributes to the organism's survival and pathogenicity through its role in translation and protein production . Additionally, as a conserved ribosomal protein, rpsD has been used in ribosomal multi-locus sequence typing (rMLST) for species identification of Enterococcus strains .

How is E. faecalis rpsD structurally different from other bacterial ribosomal S4 proteins?

While all bacterial S4 proteins share a common core structure that enables RNA binding and ribosome assembly, E. faecalis rpsD exhibits specific structural features that distinguish it from other bacterial homologs. The protein contains conserved RNA-binding domains alongside species-specific regions that may contribute to antibiotic resistance or adaptation mechanisms. Structural studies using X-ray crystallography and cryo-electron microscopy have revealed that these differences primarily occur in surface-exposed loops and the N-terminal domain, which can interact with species-specific partners or antibiotics. These structural variations may explain why E. faecalis demonstrates distinct patterns of antibiotic susceptibility compared to other bacterial species .

What are the standard methods for identifying and characterizing rpsD in E. faecalis isolates?

Identification and characterization of rpsD in E. faecalis typically involves a multi-faceted approach:

  • Genomic analysis: Whole-genome sequencing followed by rMLST (ribosomal multi-locus sequence typing) can reliably identify the rpsD gene and its allelic variants . For instance, in a recent study, genome sequencing provided over 10.5 million qualified reads for isolate characterization.

  • PCR amplification: Species-specific primers can be designed to target the rpsD gene for identification purposes. Using optimized primer sets similar to those developed for other E. faecalis targets, researchers can achieve high specificity and sensitivity .

  • Protein expression analysis: Western blotting with antibodies specific to rpsD or mass spectrometry-based proteomics can detect the protein itself. In a comprehensive surface protein study of E. faecalis V583, mass spectrometry identified 69 unique proteins, including ribosomal proteins, after proteolytic "shaving" of bacterial cells .

  • Functional assays: RNA binding assays and ribosome assembly tests can evaluate the functional properties of rpsD in different isolates.

What expression systems are most effective for producing recombinant E. faecalis rpsD protein?

Several expression systems have been evaluated for the production of recombinant E. faecalis rpsD, with the following comparative efficacy:

The most effective approach combines the pET expression system in E. coli BL21(DE3) with optimized induction conditions (0.5 mM IPTG, 18°C, 16 hours). This system achieves a balance between high yield and proper folding, particularly when supplemented with ribosome-associated chaperones. For improved solubility, fusion tags such as MBP (maltose-binding protein) or SUMO can be employed, though they necessitate an additional cleavage step during purification .

What purification strategies yield the highest purity recombinant rpsD protein?

A multi-step purification strategy is recommended to achieve high-purity recombinant rpsD:

  • Initial capture: Immobilized metal affinity chromatography (IMAC) using Ni-NTA resin for His-tagged rpsD, with optimization of imidazole concentration in wash buffers (20-40 mM) to remove weakly bound contaminants.

  • Intermediate purification: Ion exchange chromatography (typically cation exchange using SP Sepharose) exploiting rpsD's basic pI value.

  • Polishing step: Size exclusion chromatography using Superdex 75 or 200 columns to separate monomeric rpsD from aggregates and remaining contaminants.

  • Quality control: SDS-PAGE and western blotting for purity assessment, mass spectrometry for identity confirmation, and circular dichroism for secondary structure verification.

This approach typically yields >95% pure protein suitable for structural and functional studies. For crystallography applications, an additional hydroxyapatite chromatography step may be beneficial to remove nucleic acid contaminants that can interfere with crystallization .

How can researchers optimize the solubility of recombinant E. faecalis rpsD?

Optimization of E. faecalis rpsD solubility involves multiple strategies:

  • Expression conditions: Lower induction temperatures (16-18°C) significantly improve solubility by slowing protein synthesis and allowing proper folding.

  • Buffer optimization:

    • Addition of stabilizing agents: 5-10% glycerol, 100-300 mM NaCl

    • Chelating agents: 1-5 mM EDTA to prevent metal-catalyzed oxidation

    • Reducing agents: 1-5 mM DTT or β-mercaptoethanol to maintain reduced cysteines

  • Co-expression approaches: Co-expression with ribosomal RNA fragments that naturally bind rpsD can improve folding and solubility by mimicking the natural assembly process.

  • Solubility-enhancing tags: MBP or SUMO fusion tags positioned at the N-terminus can dramatically improve solubility, though their large size may interfere with some structural studies.

  • Refolding protocols: For cases where inclusion bodies are unavoidable, a gradual dialysis refolding protocol using decreasing concentrations of urea (8M to 0M) in the presence of the redox pair GSH/GSSG (5:1 ratio) has proven effective .

What are the critical factors in designing experiments to study rpsD-RNA interactions?

When designing experiments to study rpsD-RNA interactions, researchers should consider the following critical factors:

  • RNA preparation: Use in vitro transcribed 16S rRNA fragments containing the known S4 binding site (nucleotides 39-47, 394-405, 505-526) or synthetic oligonucleotides mimicking these regions. Ensure RNA is properly folded by heating to 65°C followed by slow cooling in the presence of magnesium ions.

  • Buffer conditions: Optimize ionic strength (typically 100-150 mM KCl) and magnesium concentration (5-10 mM) to mimic physiological conditions. Include RNase inhibitors to prevent RNA degradation during experiments.

  • Binding assays selection:

    • Filter binding assays provide quantitative Kd values but may underestimate complex stability

    • Electrophoretic mobility shift assays (EMSA) allow visualization of distinct complexes

    • Surface plasmon resonance (SPR) provides real-time binding kinetics

    • Isothermal titration calorimetry (ITC) measures thermodynamic parameters

  • Controls and validation:

    • Use known rpsD mutants with altered RNA binding as controls

    • Validate results using multiple complementary techniques

    • Compare binding of E. faecalis rpsD to homologous proteins from other species

  • Data analysis: Apply appropriate binding models (simple bimolecular interaction vs. cooperative binding) when analyzing binding isotherms. Consider using Hill coefficients to evaluate cooperative binding effects that are common in ribosomal protein-RNA interactions .

How should researchers design experiments to study the role of rpsD in antibiotic resistance?

Designing experiments to study rpsD's role in antibiotic resistance requires a systematic approach:

  • Strain selection and preparation:

    • Use clinically relevant E. faecalis strains with varying antibiotic susceptibility profiles

    • Create isogenic strains with wild-type and mutant rpsD alleles using CRISPR-Cas9 genome editing

    • Complement rpsD deletion strains with plasmid-expressed wild-type or mutant rpsD variants

  • Antibiotic susceptibility testing:

    • Determine minimum inhibitory concentrations (MICs) using broth microdilution according to CLSI guidelines

    • Conduct time-kill assays to evaluate bactericidal/bacteriostatic effects

    • Perform growth curve analyses in sub-inhibitory antibiotic concentrations

  • Molecular mechanism studies:

    • Analyze ribosome assembly efficiency using sucrose gradient centrifugation

    • Evaluate translation fidelity with dual-luciferase reporter systems

    • Assess antibiotic binding to ribosomes using radiolabeled antibiotics

  • Structural studies:

    • Perform cryo-EM analysis of ribosomes with and without bound antibiotics

    • Use hydrogen-deuterium exchange mass spectrometry to identify conformational changes

  • Data interpretation:

    • Correlate structural changes with functional outcomes

    • Distinguish direct effects (antibiotic binding site alteration) from indirect effects (altered ribosome assembly) .

What controls are essential when analyzing the impact of rpsD mutations on ribosome assembly?

When analyzing the impact of rpsD mutations on ribosome assembly, the following controls are essential:

  • Protein expression controls:

    • Western blot analysis to confirm equivalent expression levels of wild-type and mutant rpsD proteins

    • qRT-PCR to verify similar mRNA levels, eliminating transcriptional effects

  • Assembly pathway controls:

    • Include assembly intermediates analysis at different time points

    • Monitor other ribosomal proteins' incorporation to distinguish rpsD-specific effects

    • Analyze assembly in the presence of specific assembly factors

  • Functional controls:

    • In vitro translation assays to correlate assembly defects with functional outcomes

    • Polysome profiling to assess effects on translation initiation

  • Specificity controls:

    • Complementation experiments with wild-type rpsD to confirm phenotype reversibility

    • Introduction of known benign mutations as negative controls

    • Introduction of known assembly-disrupting mutations as positive controls

  • Environmental controls:

    • Test assembly at different temperatures to identify temperature-sensitive phenotypes

    • Vary ionic conditions to identify mutation effects that are environment-dependent

These controls help distinguish direct effects of rpsD mutations from indirect effects caused by altered protein stability or expression levels .

How can recombinant E. faecalis rpsD be used to develop novel antimicrobial strategies?

Recombinant E. faecalis rpsD offers several promising avenues for novel antimicrobial development:

  • Structure-based drug design: High-resolution structural data of rpsD bound to its RNA target can be used to design small molecules that disrupt this essential interaction. Virtual screening campaigns targeting the RNA-binding interface have identified several lead compounds with IC50 values in the micromolar range in preliminary studies.

  • Ribosome assembly inhibitors: Compounds that specifically bind to rpsD and prevent its incorporation into the 30S subunit can effectively inhibit bacterial growth. These assembly inhibitors represent a new class of antibiotics with a mechanism distinct from traditional translation inhibitors, potentially overcoming existing resistance mechanisms .

  • Peptide mimetics: Synthetic peptides derived from the RNA-binding domain of rpsD can competitively inhibit ribosome assembly. These peptides can be optimized for cell penetration and stability using non-natural amino acids and cyclization strategies.

  • Immunological approaches: As a surface-exposed protein in some growth conditions, recombinant rpsD can be used to develop vaccines or antibody-based therapeutics. In animal models, anti-rpsD antibodies have shown protection against E. faecalis infection when the protein becomes accessible during certain growth phases .

  • CRISPR-Cas targeting: The rpsD gene can be specifically targeted by CRISPR-Cas antimicrobials, creating selective pressure that leads to lethal mutations in this essential gene.

The most promising approach combines structural biology with medicinal chemistry to develop small molecules that can penetrate the bacterial cell wall and specifically disrupt rpsD function or incorporation into ribosomes .

What methods are most effective for studying the conformational dynamics of rpsD during ribosome assembly?

Studying the conformational dynamics of rpsD during ribosome assembly requires sophisticated biophysical techniques:

  • Single-molecule FRET (smFRET): By labeling specific residues in rpsD with donor and acceptor fluorophores, researchers can track distance changes during binding to 16S rRNA and subsequent assembly steps. This technique provides insights into transient intermediates that are not captured by static structural methods.

  • Nuclear Magnetic Resonance (NMR) spectroscopy: For smaller fragments of rpsD, NMR can provide detailed information about local structural changes and dynamics on multiple timescales. 15N-1H HSQC experiments are particularly useful for monitoring structural perturbations upon RNA binding.

  • Hydrogen-deuterium exchange mass spectrometry (HDX-MS): This technique identifies regions of rpsD that undergo changes in solvent accessibility during assembly, revealing conformational changes and protein-RNA interaction surfaces with peptide-level resolution.

  • Time-resolved cryo-electron microscopy: By capturing assembly intermediates at different time points, researchers can visualize the structural transitions of rpsD during incorporation into the 30S subunit.

  • Molecular dynamics simulations: Computational approaches can model the conformational changes of rpsD on timescales that are difficult to capture experimentally, generating hypotheses that can be tested with the above methods.

The most comprehensive approach combines multiple techniques at different resolution levels, correlating atomic-level structural changes with functional outcomes in ribosome assembly and translation .

How can researchers distinguish between direct and indirect effects of rpsD mutations on antibiotic resistance?

Distinguishing between direct and indirect effects of rpsD mutations on antibiotic resistance requires a systematic experimental approach:

  • Structural analysis:

    • Determine high-resolution structures of wild-type and mutant ribosomes with bound antibiotics

    • Identify changes in the antibiotic binding pocket versus distant conformational effects

    • Use computational modeling to predict how mutations affect antibiotic binding energetics

  • Biochemical assays:

    • Direct binding assays using purified components (e.g., isothermal titration calorimetry, surface plasmon resonance)

    • Competition assays with labeled and unlabeled antibiotics

    • Ribosome protection assays with purified components

  • Genetic approaches:

    • Introduce compensatory mutations that restore structure but maintain resistance

    • Create chimeric rpsD proteins to map resistance determinants

    • Use suppressor mutation analysis to identify interconnected residues

  • Translational fidelity assays:

    • Measure misincorporation rates, readthrough efficiency, and frameshifting

    • Correlate changes in translational accuracy with antibiotic resistance

    • Test whether resistance correlates with altered translation in the absence of antibiotics

  • Ribosome assembly analysis:

    • Quantify effects on assembly rate and efficiency

    • Identify accumulation of specific assembly intermediates

    • Test whether assembly defects correlate with resistance phenotypes

By combining these approaches, researchers can determine whether rpsD mutations confer resistance by directly affecting antibiotic binding, indirectly altering ribosome structure/function, or changing assembly pathways that impact the final ribosome configuration .

What are the common challenges in expressing recombinant E. faecalis rpsD and how can they be addressed?

Researchers frequently encounter several challenges when expressing recombinant E. faecalis rpsD:

  • Poor solubility and inclusion body formation:

    • Solution: Lower induction temperature to 16-18°C and reduce IPTG concentration to 0.1-0.3 mM

    • Alternative: Use solubility-enhancing fusion partners such as MBP, SUMO, or Thioredoxin

    • Advanced approach: Co-express with 16S rRNA fragments to promote natural folding

  • Proteolytic degradation:

    • Solution: Include protease inhibitor cocktail during purification

    • Alternative: Use protease-deficient expression strains (e.g., BL21(DE3) pLysS)

    • Advanced approach: Optimize construct design to remove exposed protease-sensitive sites

  • Co-purification of bound RNA:

    • Solution: Include high-salt washes (500 mM NaCl) during purification

    • Alternative: Add RNase A treatment step followed by additional purification

    • Advanced approach: Use anion exchange chromatography to separate protein-RNA complexes

  • Aggregation during concentration/storage:

    • Solution: Include 5-10% glycerol and 1-5 mM reducing agent in storage buffer

    • Alternative: Store at moderate concentration (<5 mg/mL) and concentrate only before use

    • Advanced approach: Use additives like arginine (50-100 mM) to prevent aggregation

  • Inactive protein despite successful purification:

    • Solution: Verify proper folding using circular dichroism spectroscopy

    • Alternative: Confirm identity and integrity by mass spectrometry

    • Advanced approach: Test RNA binding activity immediately after purification to establish baseline functionality

How can researchers troubleshoot inconsistent results in rpsD-RNA binding assays?

Inconsistent results in rpsD-RNA binding assays can stem from multiple sources:

  • RNA quality issues:

    • Problem: Degraded or improperly folded RNA

    • Diagnosis: Run RNA on denaturing gel or use bioanalyzer

    • Solution: Prepare fresh RNA, include RNase inhibitors, and verify proper folding by native gel electrophoresis

  • Protein activity variation:

    • Problem: Batch-to-batch variation in protein activity

    • Diagnosis: Use positive control RNA with known binding affinity

    • Solution: Standardize protein batches using activity assays before main experiments

  • Buffer composition effects:

    • Problem: Minor variations in pH, salt, or magnesium can dramatically affect binding

    • Diagnosis: Systematically vary buffer components

    • Solution: Use precisely controlled buffer compositions and include internal controls

  • Equipment calibration:

    • Problem: Drift in instrument calibration (particularly for fluorescence-based assays)

    • Diagnosis: Regular verification with standards

    • Solution: Include calibration standards in each experiment

  • Data analysis inconsistency:

    • Problem: Different fitting models or parameters

    • Diagnosis: Analyze the same dataset with multiple approaches

    • Solution: Establish standardized analysis protocols and use multiple fitting models to assess robustness

A systematic troubleshooting approach includes running multiple controls, standardizing reagents, and implementing consistent analysis protocols to minimize variability in binding assay results .

What strategies can address challenges in detecting subtle phenotypic changes in rpsD mutant strains?

Detecting subtle phenotypic changes in rpsD mutant strains requires sensitive approaches:

  • Growth condition optimization:

    • Challenge: Standard conditions may mask subtle phenotypes

    • Solution: Test growth under various stressors (temperature, pH, nutrient limitation)

    • Advanced approach: Use chemostats to maintain precise growth conditions for extended periods

  • High-sensitivity fitness assays:

    • Challenge: Traditional growth curves lack sensitivity for minor fitness effects

    • Solution: Use competition assays with differentially marked strains

    • Advanced approach: Implement deep sequencing-based fitness measurements (BarSeq)

  • Ribosome profiling:

    • Challenge: Global translation effects may be missed by bulk measurements

    • Solution: Use ribosome profiling to measure translation at nucleotide resolution

    • Advanced approach: Combine with RNA-seq to calculate translation efficiency for each transcript

  • Single-cell analysis:

    • Challenge: Population heterogeneity may obscure phenotypes

    • Solution: Use fluorescent reporters and flow cytometry to analyze individual cells

    • Advanced approach: Time-lapse microscopy to track phenotypes in lineages

  • Metabolomics approach:

    • Challenge: Metabolic adaptations may compensate for ribosomal defects

    • Solution: Untargeted metabolomics to identify perturbed pathways

    • Advanced approach: Flux analysis using labeled substrates to quantify pathway activities

The most effective strategy combines multiple approaches, particularly pairing sensitive fitness measurements with molecular techniques that directly assess ribosome function .

What emerging technologies are likely to advance our understanding of E. faecalis rpsD function and applications?

Several emerging technologies show particular promise for advancing E. faecalis rpsD research:

  • Cryo-electron tomography: This technique allows visualization of ribosomes in their native cellular context, revealing how rpsD mutations affect ribosome distribution and association with other cellular components. Recent advances in sub-tomogram averaging can achieve near-atomic resolution of ribosomes in situ.

  • Rapid detection methodologies: Novel approaches like recombinase polymerase amplification (RPA) combined with lateral flow strips (LFS) enable detection of E. faecalis within approximately 35 minutes with a detection limit of 10 CFU/μL. This technology could be adapted to specifically detect rpsD variants associated with antibiotic resistance .

  • Microfluidic ribosome assembly systems: These systems allow real-time observation of assembly processes with single-molecule resolution, potentially revealing transient intermediates and assembly pathways affected by rpsD mutations.

  • AlphaFold and deep learning approaches: AI-based structure prediction tools are increasingly accurate for protein-RNA complexes, allowing rapid modeling of how mutations affect rpsD-RNA interactions without requiring experimental structure determination.

  • CRISPR-based screening: High-throughput functional genomics using CRISPR interference or base editing can systematically map the functional landscape of rpsD, identifying residues critical for various aspects of its function.

These technologies, particularly when used in combination, will enable unprecedented insights into rpsD function at molecular, cellular, and population levels .

How might research on E. faecalis rpsD contribute to understanding bacterial evolution and adaptation?

Research on E. faecalis rpsD offers several unique windows into bacterial evolution and adaptation:

  • Ribosomal evolution: As a core component of the translation machinery, rpsD sequence and structural changes across bacterial species reveal evolutionary constraints and adaptations in this essential process. Comparative studies between E. faecalis rpsD and homologs from other bacteria can identify conserved functional cores versus species-specific adaptations.

  • Antibiotic resistance mechanisms: Tracking mutations in rpsD that emerge under antibiotic pressure provides insights into evolutionary pathways to resistance. The fitness costs and compensatory mutations associated with resistance-conferring changes in rpsD illuminate constraints on resistance evolution.

  • Host-pathogen co-evolution: E. faecalis transitions between commensal and pathogenic lifestyles, with potential changes in ribosome composition and function. Studying how rpsD contributes to these transitions can reveal mechanisms of bacterial adaptation to different host environments.

  • Horizontal gene transfer dynamics: The conservation of rpsD sequences can be used to track horizontal gene transfer events and reconstruct bacterial population structures. rMLST analysis, which includes rpsD alleles, has proven valuable for species identification and evolutionary studies in Enterococcus .

  • Experimental evolution approaches: Long-term evolution experiments with E. faecalis under controlled conditions can reveal how selection pressures drive changes in rpsD and the associated phenotypic consequences, providing empirical data on evolutionary trajectories and constraints.

This research contributes to fundamental evolutionary biology while also providing practical insights for predicting and addressing antibiotic resistance development .

What interdisciplinary approaches could enhance the development of rpsD-targeted therapeutics?

Developing effective rpsD-targeted therapeutics requires integration of multiple disciplines:

  • Structural biology and computational chemistry:

    • High-resolution structures of rpsD-RNA complexes provide templates for in silico screening

    • Molecular dynamics simulations identify druggable binding pockets and predict compound binding energetics

    • Fragment-based drug design identifies chemical scaffolds with optimal binding properties

  • Synthetic biology and genetic engineering:

    • CRISPR-based genome editing creates model strains for testing therapeutic hypotheses

    • Synthetic ribosome approaches test the functional consequences of rpsD modifications

    • Cell-free translation systems allow rapid screening of compounds affecting rpsD function

  • Biophysics and nanotechnology:

    • Advanced delivery systems overcome bacterial membrane barriers

    • Single-molecule techniques characterize drug-target interactions with unprecedented detail

    • Microfluidic systems enable high-throughput screening of compound libraries

  • Systems biology and bioinformatics:

    • Network analysis identifies synthetic lethal interactions with rpsD for combination therapies

    • Machine learning approaches predict compound properties and optimize lead structures

    • Multi-omics integration reveals system-wide effects of rpsD targeting

  • Clinical microbiology and epidemiology:

    • Surveillance studies identify emerging resistance patterns

    • Clinical isolate collections provide diverse targets for therapeutic validation

    • Rapid diagnostic methods enable targeted therapeutic approaches

The most promising path forward involves collaborative research teams integrating these disciplines, with particular emphasis on combining structural insights with medicinal chemistry and advanced delivery systems to develop therapeutics that can effectively target rpsD while overcoming the delivery challenges presented by the bacterial cell envelope.

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.