Recombinant Mycoplasma mycoides subsp. mycoides SC 50S ribosomal protein L22 (rplV)

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: While we will prioritize shipping the format currently in stock, please specify any format requirements in your order notes for customized preparation.
Lead Time
Delivery times vary depending on the purchasing method and location. Please contact your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard blue ice packs unless 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 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 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
Upon receipt, store at -20°C/-80°C. Aliquot for multiple uses to prevent repeated freeze-thaw cycles.
Tag Info
Tag type is determined during the manufacturing process.
The tag type is determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
rplV; MSC_0740; 50S ribosomal protein L22
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-111
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Mycoplasma mycoides subsp. mycoides SC (strain PG1)
Target Names
rplV
Target Protein Sequence
MEAKAKLSMI RISPRKMRLV ADTIRNKAVL VAVATLKNLN KDAAEPILKL LNSAVANAVN NNGMEADKLY VKTIFVNEGP TLKRFRPRAH GRAYEIFKRT SHVVIVVSDE K
Uniprot No.

Target Background

Function
This protein specifically binds to 23S rRNA; binding is enhanced by other ribosomal proteins such as L4, L17, and L20. It plays a crucial role in the early stages of 50S ribosomal subunit assembly. It interacts with multiple domains of 23S rRNA within both the assembled 50S subunit and the complete 70S ribosome. The globular domain resides near the polypeptide exit tunnel on the subunit's exterior, while an extended β-hairpin lines the exit tunnel wall within the 70S ribosome.
Database Links
Protein Families
Universal ribosomal protein uL22 family

Q&A

What expression systems are most effective for producing recombinant M. mycoides SC L22?

For recombinant expression of M. mycoides SC L22, an E. coli-based system has proven most effective, as demonstrated in systematic surface proteome characterization studies . The recommended approach includes:

  • Codon optimization of the M. mycoides SC rplV gene for E. coli expression

  • Cloning into a vector with an N-terminal His-tag (e.g., pET28a)

  • Expression in E. coli BL21(DE3) at lower temperatures (16-18°C) after IPTG induction

  • Cultivation in Terrific Broth supplemented with sorbitol (0.5%) and betaine (2.5mM) to enhance protein solubility

This methodology has successfully produced soluble recombinant surface proteins from M. mycoides SC with yields sufficient for downstream applications including immunological studies .

How can researchers verify the structural integrity of purified recombinant L22?

Verification of structural integrity for recombinant L22 requires a multi-technique approach:

  • Circular Dichroism (CD) spectroscopy to confirm proper secondary structure elements

  • Thermal shift assays to evaluate protein stability and proper folding

  • Size Exclusion Chromatography (SEC) to confirm monomeric state and absence of aggregation

  • Limited proteolysis to assess domain organization and accessibility

  • RNA binding assays using known L22 consensus sequences to confirm functional activity

A correctly folded L22 should demonstrate specific binding to RNA motifs containing the characteristic G-C base pair and U-containing loop structure as observed in orthologous L22 proteins .

What experimental approaches can elucidate L22's RNA binding properties in M. mycoides SC?

To characterize the RNA binding properties of M. mycoides SC L22, researchers should implement:

  • RNA Immunoprecipitation (RIP) assays: Using FLAG-tagged recombinant L22 to precipitate bound RNA molecules, followed by qRT-PCR or RNA-seq analysis .

  • Electrophoretic Mobility Shift Assays (EMSA): Using synthetic RNA oligonucleotides containing predicted binding motifs similar to those identified in intron regions of genes like MDM4 .

  • Surface Plasmon Resonance (SPR): For quantitative measurement of binding affinities to different RNA motifs.

  • CLIP-seq (Cross-linking immunoprecipitation followed by sequencing): To map genome-wide binding sites of L22 in M. mycoides SC.

  • Mutational analysis: Systematic mutation of L22 consensus binding motifs to identify critical residues required for interaction.

This multi-faceted approach can identify whether M. mycoides SC L22 recognizes similar RNA motifs as described in other organisms, particularly the G-C base pair on stem structures with a U at the 3' end of the loop .

How does nucleolar stress affect L22 function in M. mycoides SC, and how can this be studied?

To investigate how nucleolar stress influences L22 function in M. mycoides SC:

  • Cellular localization studies: Using fluorescently tagged L22 to track redistribution under stress conditions.

  • Ribosome profiling: To examine changes in the translation landscape during stress.

  • RNA-seq analysis: For identifying alternative splicing events potentially regulated by L22 during stress.

  • L22 interactome analysis: Via IP-MS (Immunoprecipitation-Mass Spectrometry) under normal and stress conditions.

  • Reporter assays: Construct splicing reporters containing potential M. mycoides SC L22-regulated introns to monitor splicing regulation during stress.

Evidence from mammalian systems suggests that during nucleolar stress, L22 may relocate from the ribosome to regulate splicing of specific pre-mRNAs . This mechanism might be evolutionarily conserved in M. mycoides SC, potentially regulating stress response genes through alternative splicing.

How can researchers analyze contradictory data regarding L22's involvement in M. mycoides SC pathogenesis?

When confronted with contradictory findings about L22's role in pathogenesis:

  • Strain-specific analysis: Compare L22 sequences and expression levels across different M. mycoides SC isolates with varying virulence.

  • Temporal expression studies: Examine L22 expression at different stages of infection using RT-qPCR and proteomics.

  • Animal model validation: Use multiple animal models to validate findings, with particular attention to cattle, the natural host of M. mycoides SC .

  • Multi-omics integration: Combine transcriptomics, proteomics, and immunological data to develop a comprehensive model.

  • Meta-analysis: Systematically compare experimental methodologies across contradictory studies to identify potential sources of variation.

Experimental ApproachStrengthsLimitationsBest Application
Surface protein arraysHigh-throughput screening of multiple proteins simultaneouslyLimited to surface-exposed epitopesHumoral response profiling
Genetic knockout studiesDirect assessment of gene functionGenetic manipulation challenges in MycoplasmaValidation of essential functions
Infection modelsPhysiologically relevantVariability between animalsValidation of in vitro findings
Luminex suspension arraysMultiplex analysis of antibody responsesRequires specific equipmentMonitoring immune responses over time

What methodologies are most effective for studying L22's potential role in alternative splicing regulation in M. mycoides SC?

To investigate L22's role in splicing regulation:

  • Minigene splicing assays: Construct reporters containing M. mycoides SC genes with potential L22 binding motifs in intronic regions.

  • CRISPR-Cas9 genome editing: Generate L22 binding site deletions in the M. mycoides SC genome to assess effects on splicing patterns .

  • RNA-seq with junction analysis: Apply specialized bioinformatic algorithms to detect alternative splicing events genome-wide.

  • L22 depletion/overexpression studies: Use inducible expression systems to modulate L22 levels and monitor splicing changes.

  • In vitro splicing assays: Reconstitute splicing reactions with purified components to directly assess L22's impact.

While M. mycoides SC has a relatively compact genome with fewer introns than eukaryotes, some genes containing introns may be regulated by L22 through mechanisms similar to those observed for MDM4 in mammalian systems . Potential L22 binding motifs within these introns can be identified through bioinformatic analysis.

How can researchers develop high-throughput screening assays to identify inhibitors of L22 function in M. mycoides SC?

For developing high-throughput screening (HTS) assays targeting L22:

  • Fluorescence polarization assay: Using fluorescently labeled RNA oligonucleotides containing L22 binding motifs to screen for compounds that disrupt binding.

  • AlphaScreen technology: For detecting protein-RNA interactions in a miniaturized format suitable for HTS.

  • Split-reporter complementation assays: Linking L22-RNA binding to a bioluminescent or fluorescent readout.

  • Surface plasmon resonance (SPR) arrays: For rapid screening of multiple compounds against immobilized L22.

  • In silico screening: Using structural models of M. mycoides SC L22 to virtually screen compound libraries before experimental validation.

The screening cascade should include:

  • Primary screening at single concentration

  • Dose-response confirmation

  • Counter-screening against other ribosomal proteins

  • Functional validation in bacterial growth assays

  • Cytotoxicity assessment in mammalian cells

Hits can be prioritized based on selectivity for L22 over other RNA-binding proteins and efficacy in inhibiting M. mycoides SC growth.

How can recombinant L22 be utilized in developing improved diagnostic tests for CBPP?

Recombinant L22 from M. mycoides SC can enhance CBPP diagnostics through:

  • Multiplex serological assays: Incorporating L22 into Luminex suspension array technology alongside other immunogenic proteins enables simultaneous detection of multiple antibody responses in minute sample volumes .

  • Protein microarrays: Immobilizing L22 with other recombinant M. mycoides SC proteins on microarray surfaces allows high-throughput screening of cattle sera.

  • Lateral flow assays: Developing point-of-care diagnostics using L22-specific antibodies for rapid field testing.

  • ELISA optimization: Using purified recombinant L22 as antigen in indirect ELISA formats with enhanced sensitivity.

When developing such assays, researchers should:

  • Evaluate both IgG and IgA responses against L22

  • Determine temporal dynamics of antibody responses

  • Compare responses in vaccinated versus naturally infected animals

  • Assess cross-reactivity with other Mycoplasma species

Current research demonstrates that magnetic bead-based assays incorporating multiple M. mycoides SC surface proteins can achieve 20-fold signal separation between CBPP-positive and negative sera , suggesting L22 could contribute to improved diagnostic sensitivity and specificity.

What experimental design is optimal for investigating L22's interaction with host immune components during CBPP infection?

To study L22-host immune interactions:

  • Ex vivo infection models: Using bovine peripheral blood mononuclear cells (PBMCs) exposed to recombinant L22.

  • Antibody epitope mapping: Identifying immunodominant regions of L22 using peptide arrays and sera from CBPP-infected cattle .

  • Cytokine profiling: Measuring pro- and anti-inflammatory cytokine responses to L22 stimulation in bovine immune cells.

  • Immunization studies: Testing recombinant L22 as a subunit vaccine component and monitoring specific immune responses.

  • Protein-protein interaction studies: Identifying host proteins that interact with L22 using pull-down assays and mass spectrometry.

The experimental design should include:

  • Appropriate controls (naive and vaccinated animals)

  • Longitudinal sampling

  • Multiple immune parameters (antibody isotypes, T-cell responses)

  • Correlation with protection status

Previous studies have successfully monitored IgG, IgM, and IgA responses against M. mycoides SC surface proteins over time in vaccine studies with eight animals , providing a methodological framework for L22-specific investigations.

What are the critical parameters for optimizing L22 solubility and stability for structural studies?

For structural studies of recombinant M. mycoides SC L22:

  • Buffer optimization:

    • Screen pH range 6.5-8.5

    • Test different ionic strengths (50-300 mM NaCl)

    • Include stabilizing agents (glycerol 5-10%, reducing agents like DTT or TCEP)

  • Protein engineering approaches:

    • Construct truncated versions to remove disordered regions

    • Create fusion proteins with solubility enhancers (MBP, SUMO)

    • Introduce surface mutations to enhance solubility

  • Refolding strategies:

    • Dilution refolding from denaturing conditions

    • On-column refolding during purification

    • Chaperone co-expression systems

  • Stabilization for crystallography:

    • Complex with RNA oligonucleotides containing binding motifs

    • Addition of molecular stabilizers

    • Surface entropy reduction mutations

Data from similar ribosomal protein studies suggests maintaining reducing conditions is critical due to potentially reactive cysteine residues that can lead to non-physiological disulfide formation and aggregation.

How can researchers effectively analyze L22 post-translational modifications in M. mycoides SC?

To characterize post-translational modifications (PTMs) of L22:

  • Mass spectrometry approaches:

    • Bottom-up proteomics using multiple proteases

    • Top-down analysis of intact protein

    • Targeted MS/MS for specific modification sites

    • Parallel reaction monitoring (PRM) for quantitative analysis

  • Enrichment strategies:

    • Phosphopeptide enrichment (TiO2, IMAC)

    • Ubiquitination enrichment (K-ε-GG antibodies)

    • Acetylation-specific antibodies

    • Chemical labeling approaches

  • Site-directed mutagenesis:

    • Mutation of predicted modification sites

    • Phosphomimetic substitutions

    • Creation of modification-resistant variants

  • Temporal dynamics:

    • Analysis of modifications under different stress conditions

    • Growth phase-dependent modification patterns

PTM TypeEnrichment MethodDetection ApproachBiological Significance Assessment
PhosphorylationTiO2 chromatographyNeutral loss scanningMutation studies and functional assays
AcetylationAnti-acetyl-lysine antibodiesDiagnostic fragment ionsDeacetylase inhibitor studies
MethylationHydrophilic interaction chromatographyAccurate mass measurementMethyltransferase knockdown effects
Proteolytic processingN-terminal COFRADICProtein N-terminal sequencingProtease inhibition studies

What emerging technologies can advance our understanding of L22's role in M. mycoides SC biology?

Cutting-edge approaches for investigating L22 function include:

  • Cryo-electron microscopy: For high-resolution structural analysis of L22 within the context of the M. mycoides SC ribosome.

  • Single-molecule fluorescence techniques: To directly observe L22-RNA interactions and potential conformational changes.

  • Nanopore direct RNA sequencing: For detecting modified nucleotides within RNA molecules that interact with L22.

  • Spatially resolved transcriptomics: To map the subcellular localization of L22-bound RNAs in M. mycoides SC.

  • Synthetic biology approaches: Creating minimal M. mycoides SC systems with engineered L22 variants to test specific hypotheses about function.

These technologies could reveal previously unrecognized aspects of L22 biology, particularly its potential roles outside the ribosome in regulating gene expression through direct RNA binding .

How might L22's RNA binding properties be harnessed for novel antimicrobial development strategies?

Leveraging L22's RNA binding functions for antimicrobial development:

  • Structure-based drug design: Using crystal structures of L22-RNA complexes to design compounds that selectively interfere with binding.

  • Peptide mimetics: Developing peptides that mimic L22's RNA binding domain to competitively inhibit its function.

  • RNA decoys: Creating synthetic RNA oligonucleotides containing L22 binding motifs to sequester the protein away from its natural targets.

  • PROTAC approach: Designing bifunctional molecules that bind L22 and recruit the bacterial degradation machinery.

  • Aptamer selection: Identifying aptamers with high affinity for L22 that could interfere with its function.

These approaches would build on the understanding that L22 binds specific RNA motifs with a G-C base pair on the stem and a U at the 3' end of the loop , potentially disrupting essential regulatory networks in M. mycoides SC.

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.