Recombinant Bacteroides thetaiotaomicron 50S ribosomal protein L3 (rplC)

Shipped with Ice Packs
In Stock

Description

Introduction to Recombinant Bacteroides thetaiotaomicron 50S Ribosomal Protein L3 (rplC)

Recombinant Bacteroides thetaiotaomicron 50S ribosomal protein L3 (rplC) refers to a genetically engineered version of the L3 ribosomal protein from the bacterium Bacteroides thetaiotaomicron. This protein is a crucial component of the bacterial ribosome, specifically part of the 50S subunit, which plays a vital role in protein synthesis. The recombinant form is produced through genetic engineering techniques, allowing for the study of its structure, function, and potential applications in biotechnology and microbiology.

Background on Bacteroides thetaiotaomicron

Bacteroides thetaiotaomicron is a prominent gut commensal bacterium known for its ability to degrade complex polysaccharides, contributing significantly to the human gut microbiome's metabolic capabilities . Its genetic and metabolic versatility makes it an attractive model for studying microbial interactions and metabolic pathways.

Role of Ribosomal Protein L3

Ribosomal protein L3 is essential for the assembly and function of the bacterial ribosome. In bacteria, L3 mutations have been associated with resistance to certain antibiotics, such as linezolid and tiamulin, which target the peptidyl transferase center (PTC) of the ribosome . The protein's structure and interactions with rRNA are critical for maintaining the ribosome's integrity and function.

Recombinant Production of L3 Protein

Recombinant production of the L3 protein involves cloning the rplC gene into a suitable expression vector and expressing it in a host organism, often Escherichia coli. This method allows for the purification and characterization of the protein in large quantities, facilitating detailed biochemical and structural studies .

Research Findings and Applications

While specific research on recombinant Bacteroides thetaiotaomicron L3 protein is limited, studies on similar proteins in other bacteria highlight their importance in understanding ribosomal function and antibiotic resistance mechanisms. The ability to engineer and study these proteins can lead to insights into novel antimicrobial strategies and biotechnological applications.

Table: Comparison of Ribosomal Protein L3 Across Different Bacteria

BacteriumFunction of L3Known Mutations/Resistance
Escherichia coliEssential for ribosome assembly and functionMutations linked to resistance against linezolid and tiamulin
Bacteroides thetaiotaomicronNot specifically studied for antibiotic resistancePotential role in polysaccharide metabolism and symbiosis
Staphylococcus spp.Associated with resistance to oxazolidinonesMutations impact ribosomal function

Future Directions

Future research on recombinant Bacteroides thetaiotaomicron L3 protein could focus on its structural characterization, interactions with other ribosomal components, and potential roles in polysaccharide metabolism. This could provide insights into how modifications of ribosomal proteins might influence bacterial fitness and symbiotic relationships within the gut microbiome.

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference during order placement for customized preparation.
Lead Time
Delivery times vary depending on the purchase method and location. Please consult your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard blue ice packs unless otherwise requested. 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 default glycerol concentration is 50%, provided as a guideline for customers.
Shelf Life
Shelf life depends on 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. Avoid repeated freeze-thaw cycles.
Tag Info
The tag type is determined during manufacturing.
The tag type is determined during the production process. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
rplC; BT_2727; 50S ribosomal protein L3
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-205
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Bacteroides thetaiotaomicron (strain ATCC 29148 / DSM 2079 / NCTC 10582 / E50 / VPI-5482)
Target Names
rplC
Target Protein Sequence
MPGLLGKKIG MTSVFSADGK NVPCTVIEAG PCVVTQVKTV EKDGYAAVQL GFQDKKEKHT TKPLMGHFKR AGVTPKRHLA EFKEFETELN LGDTITVEMF NDATFVDVVG TSKGKGFQGV VKRHGFGGVG QATHGQHNRA RKPGSIGACS YPAKVFKGMR MGGQLGGDRV TVQNLQVLKV IADHNLLLIK GSIPGCKGSI VIIEK
Uniprot No.

Target Background

Function
A key rRNA-binding protein, it directly interacts near the 3'-end of the 23S rRNA, initiating the assembly of the 50S ribosomal subunit.
Database Links

KEGG: bth:BT_2727

STRING: 226186.BT_2727

Protein Families
Universal ribosomal protein uL3 family

Q&A

Basic Research Questions

  • What is the function of the 50S ribosomal protein L3 (rplC) in Bacteroides thetaiotaomicron?

The 50S ribosomal protein L3 in B. thetaiotaomicron serves multiple critical functions in ribosome assembly and protein synthesis. As in other bacterial species, L3 is one of the first ribosomal proteins to be assembled onto the 23S rRNA and is essential for initiating the assembly of the 50S ribosomal subunit . The protein contains a branched loop that extends close to the peptidyl transferase center (PTC), the active site for peptide bond formation during protein synthesis . This proximity to the PTC allows L3 to influence the conformation of rRNA at the active site, thereby modulating peptidyl transferase activity.

L3 plays a crucial role in maintaining the structural integrity of the ribosome's catalytic center. Along with ribosomal proteins L2 and L4, L3 is considered an indispensable component for the formation of the PTC . These proteins do not directly participate in catalysis but rather stabilize and maintain the conformation of rRNA at the active site. This structural contribution is essential for the proper functioning of the ribosome during translation.

In B. thetaiotaomicron specifically, the L3 protein likely contributes to the organism's ability to adapt to changing nutrient conditions in the gut environment. As B. thetaiotaomicron is known for its extensive capacity to utilize various polysaccharides, efficient protein synthesis machinery is essential for rapid adaptation to different carbon sources .

  • How is the rplC gene organized in the B. thetaiotaomicron genome?

The rplC gene encoding the L3 ribosomal protein in B. thetaiotaomicron is organized within a conserved operon structure similar to that observed in other bacterial species. Based on comparative genomic analysis with E. coli, the rplC gene is likely located in the S10 operon, which is part of the str-spc region of the bacterial chromosome . In E. coli, this operon encodes 11 ribosomal proteins, and a similar organization is expected in B. thetaiotaomicron due to the highly conserved nature of ribosomal genes across bacterial species.

The rplC gene is typically positioned upstream of the rplD gene, which encodes the L4 ribosomal protein . This genetic organization is functionally significant because the L4 protein strongly regulates the expression of the S10 operon through a feedback mechanism. When L4 is in excess, it can bind to the mRNA of the S10 operon and inhibit further translation, thereby maintaining appropriate stoichiometry of ribosomal proteins.

In the context of B. thetaiotaomicron's genome, which contains 88 polysaccharide utilization loci (PULs) covering 18% of its genome , the ribosomal protein operons represent essential housekeeping genes that are consistently expressed to support the rapid protein synthesis needs of this metabolically versatile organism.

  • What is the structural significance of ribosomal protein L3 in the bacterial ribosome?

Ribosomal protein L3 holds remarkable structural significance in the bacterial ribosome, serving as a cornerstone for ribosome assembly and function. The most critical structural feature of L3 is its unique conformation, where the main part of the protein is positioned on the surface of the 50S ribosomal subunit, while a branched loop extends deep into the ribosome, reaching close to the peptidyl transferase center (PTC) . This architectural arrangement allows L3 to influence activities at the ribosome's functional core while maintaining connections to the ribosome's outer surface.

L3 is one of the first proteins to be incorporated during ribosome assembly, highlighting its role as a nucleation site for 50S subunit formation. Reconstitution experiments have demonstrated that only L3 and L24 are capable of initiating assembly of the E. coli 50S ribosomal subunit . This early incorporation is essential for creating the scaffold onto which other ribosomal proteins and rRNA segments can properly fold and assemble.

The positioning of L3 relative to the PTC makes it particularly relevant for antibiotic interactions. Many antibiotics that target the bacterial ribosome, including linezolid (an oxazolidinone) and tiamulin (a pleuromutilin), bind to the PTC where L3's branched loop extends . Consequently, structural alterations in L3 can affect antibiotic binding and contribute to resistance mechanisms.

Computational modeling of L3 mutations in E. coli has been used to assess changes in 50S structure and antibiotic binding, revealing how subtle alterations in L3 can propagate through the ribosomal structure to affect both rRNA conformation and antibiotic susceptibility .

  • How does B. thetaiotaomicron L3 protein compare to its homologs in other bacterial species?

The B. thetaiotaomicron L3 protein shares significant sequence and structural homology with L3 proteins from other bacterial species, particularly within conserved functional domains. While the search results don't provide a direct comparison of B. thetaiotaomicron L3 with other bacterial L3 proteins, we can infer similarities based on the conserved nature of ribosomal proteins across bacterial species.

When comparing ribosomal proteins across different bacterial species, the highest conservation is typically observed in regions that interact with rRNA or participate in critical functional processes. In L3, the branched loop that extends toward the peptidyl transferase center (PTC) is particularly well-conserved because of its essential role in ribosome function.

Table 1: Comparative analysis of L3 protein features across bacterial species

FeatureB. thetaiotaomicron L3 (inferred)E. coli L3Staphylococcus aureus L3Brachyspira spp. L3
Proximity to PTCBranched loop extends to PTCBranched loop extends to PTC Branched loop extends to PTC Branched loop extends to PTC
Role in antibiotic resistancePotentially involved in resistance to PTC-targeting antibioticsMutations linked to tiamulin and linezolid resistance Mutations linked to linezolid resistance Mutations linked to tiamulin/valnemulin resistance
Assembly roleLikely early assembly proteinOne of first proteins to be assembled Early assembly proteinEarly assembly protein
Regulatory functionMay influence gene expression in polysaccharide utilizationModulates peptidyl transferase activity Modulates peptidyl transferase activityModulates peptidyl transferase activity

As a member of the Bacteroidetes phylum, B. thetaiotaomicron's L3 protein likely contains unique adaptations that reflect its evolutionary history and ecological niche in the mammalian gut. These adaptations may include specific amino acid substitutions that optimize protein synthesis under the anaerobic, nutrient-rich conditions of the gut environment, particularly given B. thetaiotaomicron's specialization in utilizing complex polysaccharides .

  • What are the characteristic domains and motifs in B. thetaiotaomicron L3 protein?

The B. thetaiotaomicron L3 protein contains several characteristic domains and motifs that are essential for its function in ribosome assembly and protein synthesis. While the search results don't provide specific information about B. thetaiotaomicron L3 domains, we can infer these features based on the highly conserved nature of ribosomal proteins and information available about L3 proteins in other bacterial species.

The most distinctive structural feature of L3 is its extended loop structure that projects toward the peptidyl transferase center (PTC) of the ribosome . This loop contains highly conserved residues that interact with the 23S rRNA to stabilize the conformation of the PTC. Mutations in this loop region have been associated with antibiotic resistance in various bacterial species, highlighting its functional importance .

L3 typically contains a globular domain that sits on the surface of the ribosome, anchoring the protein to the 50S subunit. This domain interacts with other ribosomal proteins and rRNA elements to maintain the structural integrity of the ribosome. The globular domain also contains binding sites for regulatory factors that may modulate ribosome function under different physiological conditions.

In the context of B. thetaiotaomicron's ecological niche as a gut symbiont specialized in polysaccharide degradation, the L3 protein may contain specific adaptations that optimize ribosome function under varying nutrient conditions. While L3 itself is not directly involved in polysaccharide utilization, its role in protein synthesis makes it integral to the expression of the numerous enzymes and transporters encoded within B. thetaiotaomicron's 88 polysaccharide utilization loci (PULs) .

Comparative analysis of L3 sequences across Bacteroides species would likely reveal conserved motifs that reflect the shared evolutionary history of these gut symbionts, as well as variable regions that may contribute to species-specific adaptations to different host environments or dietary patterns.

Advanced Research Questions

  • What methodologies are most effective for expressing recombinant B. thetaiotaomicron 50S ribosomal protein L3?

Expressing recombinant B. thetaiotaomicron 50S ribosomal protein L3 presents several challenges due to its role as an essential ribosomal protein. Based on methodologies used for other ribosomal proteins, several approaches can be recommended for optimal expression.

A plasmid exchange system similar to that described for E. coli L3 provides an effective methodology . This approach involves creating a host strain with a chromosomal deletion of the native L3 gene, complemented by a plasmid-borne wild-type L3 gene. The recombinant L3 variants can then be introduced on a second, compatible plasmid, followed by selection for replacement of the wild-type plasmid. This methodology allows for the exclusive expression of the recombinant L3 protein without competition from the wild-type protein.

For the expression protocol, the following steps are recommended:

  • Clone the B. thetaiotaomicron rplC gene into an expression vector with an appropriate promoter. For anaerobic bacteria like B. thetaiotaomicron, promoters active under anaerobic conditions should be selected.

  • Include a purification tag (e.g., His-tag, FLAG-tag) that doesn't interfere with protein folding or function. Placement at the C-terminus is often preferred to minimize interference with ribosome assembly.

  • Transform the expression construct into an appropriate host system. While E. coli is commonly used, expression in B. thetaiotaomicron itself or in closely related Bacteroides species may improve protein folding and stability.

  • Induce protein expression under conditions that mimic the anaerobic gut environment. Consider using minimal media supplemented with carbon sources relevant to B. thetaiotaomicron's natural habitat.

  • For purification, use gentle lysis conditions to preserve protein structure, followed by affinity chromatography based on the chosen purification tag.

Expression success can be verified using Western blot analysis with antibodies specific to L3 or to the purification tag . Additionally, mass spectrometry can confirm the identity of the expressed protein, as demonstrated in the E. coli L3 expression system .

  • How can mutations in B. thetaiotaomicron L3 be engineered to study antibiotic resistance mechanisms?

Engineering mutations in B. thetaiotaomicron L3 to study antibiotic resistance mechanisms requires a systematic approach focusing on regions known to influence antibiotic interactions. Based on research with E. coli L3, mutations should be targeted to the loops of L3 near the peptidyl transferase center (PTC), as these regions are most likely to affect antibiotic binding .

A methodological approach for engineering and studying L3 mutations would include:

  • Identification of target residues: Analyze the sequence and predicted structure of B. thetaiotaomicron L3, focusing on residues equivalent to those implicated in antibiotic resistance in other bacterial species. The branched loop of L3 that extends near the PTC should be prioritized .

  • Mutagenesis design: Implement site-directed mutagenesis to create specific amino acid substitutions. Consider both mutations identified in clinical isolates and rational design based on structural predictions. Include a range of mutation types: conservative substitutions, charge alterations, and size changes.

  • Expression system selection: Utilize a plasmid exchange system similar to that described for E. coli, where plasmid-carried mutated L3 genes replace wild-type L3 in a chromosomal L3 deletion strain . This approach ensures that only the mutated L3 is present in the ribosomes.

  • Phenotypic characterization: Evaluate the impact of mutations on:

    • Growth rates under different conditions (as a measure of fitness cost)

    • Minimum inhibitory concentrations (MICs) for various PTC-targeting antibiotics, particularly linezolid and tiamulin

    • Ribosome assembly efficiency

    • Translation fidelity using reporter systems

  • Structural analysis: Implement computational modeling to predict the impact of L3 mutations on 50S structure and antibiotic binding, similar to the XP Glide methodology used for E. coli L3 mutations . This can provide insights into the mechanism by which the mutations affect antibiotic susceptibility.

Table 2: Potential mutations in B. thetaiotaomicron L3 and their predicted effects based on E. coli studies

Mutation RegionExample MutationsExpected Effect on Antibiotic SusceptibilityPotential Fitness Cost
PTC-proximal loopEquivalent to E. coli mutationsReduced susceptibility to linezolid, tiamulinModerate fitness cost
Surface-exposed regionsConservative substitutionsMinimal effect on antibiotic susceptibilityLow fitness cost
rRNA interfaceCharge alterationsVariable effects on drug bindingPotentially high fitness cost
L3-L4 interfaceSize-altering mutationsPossible indirect effects via altered assemblyModerate to high fitness cost

It's essential to verify the expression and stability of mutated L3 proteins using Western blotting and mass spectrometry, as demonstrated in the E. coli L3 mutation studies .

  • What are the challenges in crystallizing recombinant B. thetaiotaomicron 50S ribosomal protein L3 for structural studies?

Crystallizing recombinant B. thetaiotaomicron 50S ribosomal protein L3 for structural studies presents several significant challenges that require specific methodological approaches to overcome. These challenges stem from both the intrinsic properties of ribosomal proteins and the specific characteristics of B. thetaiotaomicron as an anaerobic gut bacterium.

The primary challenges and their methodological solutions include:

  • Protein stability outside the ribosomal context: Ribosomal proteins like L3 have evolved to function within the complex environment of the ribosome, where they interact extensively with rRNA and other proteins. When isolated, L3 may adopt non-native conformations or aggregate. To address this:

    • Optimize buffer conditions to mimic the ribosomal environment (high Mg²⁺ concentration, appropriate pH)

    • Consider co-crystallization with synthetic rRNA fragments that represent L3's natural binding partners

    • Use stabilizing agents such as osmolytes that don't interfere with crystal formation

  • Protein solubility: L3 contains hydrophobic regions that interact with the ribosome interior, potentially leading to solubility issues when expressed recombinantly. Solutions include:

    • Design fusion constructs with solubility-enhancing tags (e.g., MBP, SUMO) that can be cleaved before crystallization

    • Perform systematic buffer optimization screening, varying ionic strength, pH, and additives

    • Implement surface entropy reduction by mutating surface-exposed flexible residues to enhance crystal contacts

  • Conformational flexibility: L3 likely contains flexible regions, particularly in the branched loop that extends toward the PTC , which can impede crystal formation. Approaches to address this include:

    • Limited proteolysis to identify and potentially remove highly flexible regions

    • Introduce targeted disulfide bonds to restrict conformational flexibility

    • Use antibody fragments (Fab) for co-crystallization to stabilize specific conformations

  • Expression challenges: As an anaerobic gut bacterium, B. thetaiotaomicron proteins may require specific conditions for proper folding. Methodological approaches include:

    • Expression in anaerobic systems or closely related host organisms

    • Consider codon optimization for the expression host while maintaining critical structural elements

    • Co-expression with specific chaperones if misfolding is observed

  • Crystal quality optimization: Once initial crystals are obtained, improving resolution requires systematic optimization:

    • Implement seeding techniques to improve crystal order

    • Consider crystallization in microgravity to reduce convection effects

    • Use post-crystallization treatments such as dehydration or annealing to improve diffraction quality

For B. thetaiotaomicron L3 specifically, it may be worthwhile to consider alternative structural approaches such as cryo-electron microscopy of reconstituted ribosomal subunits containing the recombinant L3, potentially revealing its structure in a more native context.

  • How does the interaction between L3 and 23S rRNA contribute to B. thetaiotaomicron's antibiotic susceptibility profile?

The interaction between L3 and 23S rRNA is a critical determinant of B. thetaiotaomicron's antibiotic susceptibility profile, particularly for antibiotics targeting the peptidyl transferase center (PTC). While the search results don't provide specific information about B. thetaiotaomicron, insights can be inferred from studies in other bacterial species.

The L3 protein contains a branched loop that extends close to the PTC, where many antibiotics bind . This strategic positioning allows L3 to influence antibiotic binding both directly, through potential steric interactions, and indirectly, by modulating the conformation of 23S rRNA nucleotides that form the binding site. Mutations in L3, particularly in this branched loop region, have been associated with resistance to PTC-targeting antibiotics in various bacterial species .

The specific interactions between L3 and 23S rRNA that contribute to antibiotic susceptibility include:

  • Conformational stabilization of PTC nucleotides: L3 helps maintain the proper conformation of key 23S rRNA nucleotides in the PTC. Alterations in L3 can propagate through the ribosomal structure to affect the positioning of these nucleotides, potentially reducing antibiotic binding affinity.

  • Allosteric effects on drug binding sites: Even though L3 may not directly contact certain antibiotics, its interactions with 23S rRNA can allosterically influence antibiotic binding sites. For example, mutations in L3 have been shown to affect susceptibility to tiamulin, which binds to the PTC .

  • Influence on rRNA modifications: The interaction between L3 and 23S rRNA may affect the accessibility of certain rRNA nucleotides to modification enzymes. In some bacteria, methylation of 23S rRNA has been linked to antibiotic resistance.

Methodologically, several approaches can be used to study these interactions in B. thetaiotaomicron:

  • Computational modeling: Similar to the approach used for E. coli L3 mutations , computational modeling using the XP Glide methodology can predict how L3 mutations affect the structure of the 50S subunit and antibiotic binding.

  • Site-directed mutagenesis: Creating specific mutations in the L3 protein, particularly in the PTC-proximal loop, followed by antibiotic susceptibility testing to identify regions critical for drug interactions.

  • Ribosome footprinting: This technique can identify specific 23S rRNA nucleotides protected by L3 binding, revealing potential interaction sites relevant to antibiotic susceptibility.

  • Chemical probing of rRNA structure: Methods such as SHAPE (Selective 2'-Hydroxyl Acylation analyzed by Primer Extension) can detect changes in rRNA conformation resulting from L3 mutations, providing insights into how these mutations affect antibiotic binding sites.

  • What bioinformatic approaches can be used to predict the impact of L3 mutations on ribosome function in B. thetaiotaomicron?

Predicting the impact of L3 mutations on ribosome function in B. thetaiotaomicron requires sophisticated bioinformatic approaches that integrate structural, evolutionary, and functional data. Several methodological strategies can be employed to make such predictions with high confidence.

  • Homology modeling and structural analysis:

    • Generate a homology model of B. thetaiotaomicron L3 based on crystal structures from related organisms

    • Dock the modeled L3 into available ribosome structures to identify critical interaction sites

    • Implement molecular dynamics simulations to assess the stability of wild-type and mutant L3 structures

    • Use methods similar to the XP Glide methodology described for E. coli L3 to quantify binding energy changes for antibiotics

  • Evolutionary conservation analysis:

    • Perform multiple sequence alignment of L3 proteins across bacterial species, focusing on Bacteroidetes

    • Calculate conservation scores for each amino acid position to identify functionally critical residues

    • Apply methods such as Statistical Coupling Analysis (SCA) to detect co-evolving networks of amino acids that may be functionally linked

    • Identify positions under positive or negative selection pressure, which may indicate functional constraints

  • Machine learning approaches:

    • Develop classification models trained on known L3 mutations and their phenotypic effects

    • Use feature engineering to incorporate structural information, evolutionary conservation, and physico-chemical properties

    • Implement deep learning approaches such as graph neural networks to capture the complex relationship between L3 mutations and ribosome function

    • Validate predictions using experimental data from related bacterial species

  • Network analysis of ribosomal interactions:

    • Construct interaction networks representing contacts between L3 residues and other ribosomal components

    • Perform in silico alanine scanning to identify critical interaction nodes

    • Use perturbation analysis to predict how mutations propagate effects through the ribosomal structure

    • Apply centrality measures to identify hub residues whose mutation would have widespread effects

  • Integration with experimental data:

    • Correlate computational predictions with experimental data on antibiotic resistance profiles

    • Use structure-activity relationship analysis to refine predictions based on known mutation effects

    • Implement Bayesian approaches to update prediction confidence based on new experimental evidence

Table 3: Comparison of bioinformatic approaches for predicting L3 mutation effects

For B. thetaiotaomicron specifically, these approaches should incorporate data on its unique ecological niche as a gut symbiont specialized in polysaccharide degradation , as this environmental context may influence the functional constraints on L3.

  • How can recombinant L3 be used to study B. thetaiotaomicron's adaptation to different gut environments?

Recombinant L3 protein offers a powerful tool to investigate B. thetaiotaomicron's adaptation to diverse gut environments, particularly focusing on translation regulation under varying nutrient conditions. Given B. thetaiotaomicron's remarkable capacity to utilize different polysaccharides , studying how ribosomal proteins like L3 contribute to this adaptability can provide insights into the organism's ecological success.

Methodological approaches for using recombinant L3 to study gut adaptation include:

  • Comparative expression analysis under different nutrient conditions:

    • Generate recombinant L3 variants with reporter tags (e.g., fluorescent proteins or epitope tags)

    • Expose B. thetaiotaomicron cultures to different polysaccharide sources similar to those described in the second search result (corn starch, fucoidan, mucin-type O-glycans)

    • Quantify L3 expression levels using Western blotting or flow cytometry

    • Correlate changes in L3 expression with activation of specific polysaccharide utilization loci (PULs)

  • Protein-RNA interaction studies in varied environments:

    • Use techniques such as RNA immunoprecipitation (RIP) with recombinant tagged L3 to identify changes in L3-rRNA or L3-mRNA interactions under different growth conditions

    • Apply CLIP-seq (Crosslinking Immunoprecipitation followed by sequencing) to map interaction sites at nucleotide resolution

    • Determine whether L3 might play regulatory roles beyond its structural function in the ribosome when B. thetaiotaomicron adapts to different carbon sources

  • In vitro translation system with recombinant components:

    • Develop a B. thetaiotaomicron-specific in vitro translation system using recombinant L3 and other ribosomal components

    • Assess translation efficiency of PUL-associated mRNAs under different conditions

    • Determine whether post-translational modifications of L3 occur in response to environmental changes

  • Host colonization studies with L3 variants:

    • Create B. thetaiotaomicron strains expressing tagged or mutated L3 variants

    • Colonize gnotobiotic mice with these strains and expose them to different diets

    • Monitor bacterial adaptation through proteomic and transcriptomic analyses

    • Assess competitive fitness of strains with different L3 variants under varying dietary regimes

  • Metabolic labeling to track translation dynamics:

    • Use approaches similar to the fucose analog (FucAl) labeling described in the search results

    • Apply ribosome profiling to identify changes in translation patterns when B. thetaiotaomicron is exposed to different polysaccharides

    • Determine whether L3 variants affect the translation efficiency of specific mRNA subsets

This research direction is particularly relevant given B. thetaiotaomicron's ecological niche as a gut symbiont specialized in polysaccharide degradation. The organism has 88 polysaccharide utilization loci (PULs) covering 18% of its genome , and efficient regulation of protein synthesis through ribosomal components like L3 would be essential for rapid adaptation to changing nutrient availability in the gut environment.

  • What techniques are available for studying the assembly of L3 into the 50S ribosomal subunit in B. thetaiotaomicron?

Studying the assembly of L3 into the 50S ribosomal subunit in B. thetaiotaomicron requires sophisticated techniques that can capture the temporal and spatial aspects of this complex process. As L3 is one of the first ribosomal proteins to be assembled onto the 23S rRNA , understanding its incorporation is crucial for elucidating the entire ribosome assembly pathway in this important gut symbiont.

Several methodological approaches can be employed to study L3 assembly:

  • In vitro reconstitution assays:

    • Express and purify recombinant B. thetaiotaomicron L3 with appropriate tags

    • Transcribe 23S rRNA in vitro or purify ribosomal components from cells

    • Perform sequential addition experiments to determine the order of assembly

    • Monitor assembly using techniques such as sucrose gradient centrifugation, light scattering, or native gel electrophoresis

    • Analyze assembly intermediates by cryo-electron microscopy to visualize L3 positioning

  • Pulse-chase labeling and immunoprecipitation:

    • Metabolically label newly synthesized L3 using radioactive amino acids or non-canonical amino acids

    • Chase with unlabeled amino acids and collect samples at different time points

    • Immunoprecipitate ribosomal complexes using antibodies against L3 or other ribosomal markers

    • Analyze the composition of precipitated complexes to track L3 incorporation into assembling ribosomes

  • Fluorescence-based approaches:

    • Generate fluorescently tagged L3 variants that retain functionality

    • Implement fluorescence recovery after photobleaching (FRAP) to study the dynamics of L3 incorporation

    • Use Förster resonance energy transfer (FRET) between labeled L3 and other ribosomal components to monitor assembly in real-time

    • Apply single-molecule fluorescence techniques to track individual L3 molecules during assembly

  • Time-resolved structural studies:

    • Use time-resolved chemical probing methods such as SHAPE-seq to monitor rRNA structure changes during L3 binding

    • Implement hydrogen-deuterium exchange mass spectrometry (HDX-MS) to identify protein regions involved in early assembly contacts

    • Apply time-resolved cryo-electron microscopy to capture structural snapshots during assembly

    • Develop crosslinking methods to freeze transient interactions during the assembly process

  • Genetic approaches:

    • Create conditional L3 mutants in B. thetaiotaomicron to study assembly defects

    • Implement CRISPR interference to temporarily reduce L3 expression and observe assembly intermediates

    • Use suppressor mutation analysis to identify functional interactions important for assembly

    • Develop ribosome profiling methods to monitor translation of other ribosomal proteins when L3 assembly is perturbed

Table 4: Comparison of techniques for studying L3 assembly into 50S subunits

TechniqueTemporal ResolutionSpatial ResolutionThroughputKey AdvantagesKey Limitations
In vitro reconstitutionMinutes to hoursDepends on detection methodLowControlled conditionsMay not reflect in vivo complexity
Pulse-chase labelingMinutesLowMediumCaptures in vivo dynamicsLimited temporal resolution
Fluorescence approachesSeconds to minutesNanometer scaleLow to mediumReal-time observationMay require protein modification
Time-resolved structural studiesSeconds to minutesAtomic to molecularLowHigh structural detailTechnically challenging
Genetic approachesHours to generationsLowHighIn vivo relevanceIndirect readouts of assembly

These techniques can be particularly valuable for understanding how B. thetaiotaomicron's ribosome assembly might be regulated in response to the varying nutrient conditions encountered in the gut environment, potentially linking ribosome biogenesis to the organism's impressive polysaccharide utilization capabilities .

  • How does the presence of various polysaccharides affect the expression and function of L3 in B. thetaiotaomicron?

The presence of various polysaccharides likely influences the expression and function of ribosomal protein L3 in B. thetaiotaomicron, given this organism's specialized ability to sense and utilize diverse carbohydrate sources. While the search results don't directly address this relationship, we can infer potential mechanisms based on B. thetaiotaomicron's known responses to different polysaccharide nutrition conditions .

Methodological approaches to investigate this relationship include:

  • Quantitative proteomics under different polysaccharide conditions:

    • Culture B. thetaiotaomicron in minimal media supplemented with different polysaccharides similar to those described in the search results (corn starch, fucoidan, mucin-type O-glycans)

    • Apply stable isotope labeling with amino acids in cell culture (SILAC) to quantify changes in L3 expression

    • Use targeted proteomics approaches such as selected reaction monitoring (SRM) to accurately measure L3 levels

    • Compare L3 post-translational modifications across different nutritional states

  • Transcriptional and translational regulation analysis:

    • Implement RNA-seq to measure rplC mRNA levels under different polysaccharide conditions

    • Use ribosome profiling to assess translational efficiency of the rplC gene

    • Analyze the activity of promoters controlling the S10 operon (which typically contains rplC) in response to different carbon sources

    • Investigate whether polysaccharide utilization loci (PULs) activation affects ribosomal protein expression through global regulatory mechanisms

  • Structural and functional assessment of ribosomes:

    • Isolate ribosomes from B. thetaiotaomicron grown on different polysaccharides

    • Compare the incorporation efficiency of L3 into 50S subunits

    • Measure peptidyl transferase activity using in vitro translation assays

    • Assess ribosome stability and subunit association through sedimentation analysis

  • In vivo labeling approaches:

    • Adapt the fucose analog labeling method described in the search results to specifically monitor L3 synthesis

    • Create reporter fusions to track L3 expression in real-time during polysaccharide source transitions

    • Implement pulse labeling experiments to measure L3 turnover rates under different nutritional states

When B. thetaiotaomicron encounters a new polysaccharide source, it must rapidly express the appropriate degradative enzymes and transporters encoded in its 88 polysaccharide utilization loci (PULs) . This increased protein synthesis demand might necessitate upregulation of ribosomal components, including L3, to support the translational capacity required for this adaptive response.

  • What role might L3 play in B. thetaiotaomicron's polysaccharide utilization systems?

While L3 is not directly involved in polysaccharide degradation or transport, it likely plays an indirect but crucial role in B. thetaiotaomicron's polysaccharide utilization systems through its essential function in ribosome structure and protein synthesis. The search results provide context about B. thetaiotaomicron's extensive polysaccharide utilization capabilities , which can be linked to ribosomal function.

L3's potential contributions to polysaccharide utilization can be investigated through several methodological approaches:

  • Conditional expression studies:

    • Create B. thetaiotaomicron strains with L3 under control of inducible promoters

    • Titrate L3 expression to different levels and measure the impact on expression of polysaccharide utilization loci (PULs) proteins

    • Determine whether limiting L3 availability creates bottlenecks in ribosome assembly that affect the cell's ability to respond to new polysaccharide sources

    • Assess whether L3 expression is preferentially maintained during nutrient limitation to preserve translational capacity

  • Ribosome specialization analysis:

    • Investigate whether different growth conditions lead to ribosomes with altered L3 content or modifications

    • Implement ribosome profiling to determine if L3-containing ribosomes preferentially translate specific subsets of mRNAs, particularly those encoding PUL components

    • Analyze whether L3 variants affect the translation efficiency of mRNAs with different codon usage patterns or secondary structures

    • Examine whether L3 interacts with regulatory factors that coordinate ribosome function with nutritional status

  • Molecular evolution studies:

    • Compare L3 sequences across Bacteroides species with different polysaccharide utilization capabilities

    • Identify L3 residues under positive selection that might indicate adaptation to specific translational needs

    • Analyze whether L3 coevolved with specific PUL components or regulatory systems

    • Reconstruct the evolutionary history of L3 in relation to the expansion of PULs in Bacteroides genomes

B. thetaiotaomicron contains 88 polysaccharide utilization loci (PULs) covering 18% of its genome , encoding proteins involved in sensing, importing, degrading, and regulating dietary carbohydrates. The coordinated expression of these numerous proteins requires efficient and responsive translational machinery. As an essential component of the ribosome's peptidyl transferase center , L3 directly influences protein synthesis capacity.

When B. thetaiotaomicron encounters a new polysaccharide source, it must rapidly express the appropriate suite of enzymes and transporters. This sudden demand for increased protein synthesis would rely on optimal ribosome function, where L3 plays a critical role. Additionally, the need to fine-tune protein expression based on available resources might involve specialized ribosome populations or modifications to core ribosomal proteins like L3.

The experimental approach described in the second search result, using fucose analog labeling to study protein expression under different polysaccharide conditions , could be adapted to specifically examine how L3 expression and incorporation into ribosomes changes during adaptation to new carbon sources.

  • How can isotope labeling of recombinant L3 be optimized for NMR studies of B. thetaiotaomicron ribosomal complexes?

Optimizing isotope labeling of recombinant B. thetaiotaomicron L3 for NMR studies requires careful consideration of expression systems, labeling strategies, and sample preparation techniques. This methodological approach is essential for obtaining high-quality structural data on L3 within ribosomal complexes.

A comprehensive optimization protocol would include:

  • Expression system selection and optimization:

    • Compare different expression hosts including E. coli strains optimized for isotope labeling (e.g., BL21(DE3), C41(DE3))

    • Consider cell-free expression systems for rapid production of labeled L3

    • Evaluate expression in minimal media containing isotope-labeled precursors (¹⁵N-ammonium sulfate, ¹³C-glucose, ²H₂O)

    • Optimize induction conditions (temperature, inducer concentration, duration) to maximize yield while maintaining protein folding

  • Selective labeling strategies:

    • Implement amino acid-specific labeling for targeted analysis of functional regions

    • Use segmental isotope labeling to focus on the branched loop extension of L3 that approaches the PTC

    • Apply SAIL (Stereo-Array Isotope Labeling) technology for stereospecific assignments

    • Design sparse labeling schemes to reduce spectral complexity in large protein-RNA complexes

  • Sample preparation for different NMR experiments:

    • For isolated L3 studies: Optimize buffer conditions to maintain stability while minimizing signal broadening

    • For L3-rRNA complexes: Develop methods to reconstitute defined complexes with isotope-labeled L3 and unlabeled rRNA fragments

    • For intact ribosomal subunit studies: Establish protocols for incorporating labeled L3 into otherwise unlabeled 50S subunits

    • Implement specific isotope filtering experiments to selectively observe L3 signals within complex assemblies

  • Advanced NMR methodologies for large complexes:

    • Design TROSY-based experiments to mitigate relaxation effects in large complexes

    • Implement methyl-TROSY approaches focusing on labeled methyl groups as sensitive probes

    • Apply solid-state NMR techniques for very large assemblies

    • Combine solution and solid-state NMR with other structural techniques (cryo-EM, X-ray crystallography) for integrated structural analysis

Table 5: Comparison of isotope labeling strategies for NMR studies of L3

Labeling StrategyInformation ContentTechnical ComplexitySample RequirementsSuitable NMR ExperimentsAdvantages for L3 Studies
Uniform ¹⁵NBackbone assignments, dynamicsLow2-5 mg proteinHSQC, TROSYGood for initial characterization
Uniform ¹³C/¹⁵NComplete assignmentsMedium5-10 mg proteinTriple resonanceComprehensive structure determination
Deuteration with selective protonationSpecific site informationHigh5-10 mg proteinMethyl-TROSYEnhanced sensitivity for large complexes
Segmental labelingDomain-specific informationVery highMultiple samplesStandard and specializedFocus on PTC-proximal loops
Site-specific labelingPrecise local informationExtremely highMultiple samplesFiltered/edited experimentsTargeted analysis of critical residues

For B. thetaiotaomicron L3 specifically, special consideration should be given to maintaining the protein's native conformation during the labeling and purification process. As an anaerobic gut bacterium with specific physiological requirements, protein expression conditions may need to be adjusted to ensure proper folding. Additionally, since L3 is one of the first proteins incorporated during ribosome assembly , studying its dynamic interactions with rRNA requires careful design of labeling strategies that capture its behavior in different assembly states.

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.