Recombinant Mycoplasma gallisepticum Translation initiation factor IF-2 (infB), partial

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 during order placement for customized preparation.
Lead Time
Delivery times vary depending on the purchasing method and location. Please consult your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard blue ice packs unless dry ice shipping is requested in advance. Additional fees apply for dry ice shipping.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to collect the contents. Reconstitute the protein in sterile deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50%, which can serve as a guideline.
Shelf Life
Shelf life depends on various factors, including storage conditions, buffer components, temperature, and the protein's inherent stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
The tag type is determined during the manufacturing process.
The tag type is determined during production. If you require a specific tag, please inform us; we will prioritize its development.
Synonyms
infB; MYCGA1150; MGA_0821Translation initiation factor IF-2
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Protein Length
Partial
Purity
>85% (SDS-PAGE)
Species
Mycoplasma gallisepticum (strain R(low / passage 15 / clone 2))
Target Names
infB
Uniprot No.

Target Background

Function
A crucial component in initiating protein synthesis, this protein protects formylmethionyl-tRNA from spontaneous hydrolysis and facilitates its binding to 30S ribosomal subunits. It also participates in GTP hydrolysis during 70S ribosomal complex formation.
Database Links

KEGG: mga:MGA_0821

Protein Families
TRAFAC class translation factor GTPase superfamily, Classic translation factor GTPase family, IF-2 subfamily
Subcellular Location
Cytoplasm.

Q&A

What is the role of Translation Initiation Factor IF-2 (infB) in Mycoplasma gallisepticum?

Translation Initiation Factor IF-2 (infB) plays a critical role in protein synthesis in Mycoplasma gallisepticum by facilitating the binding of initiator tRNA (fMet-tRNA) to the ribosomal P-site during translation initiation. Unlike its counterparts in more complex organisms, M. gallisepticum infB operates within a minimalist genome context, making it particularly interesting for understanding essential bacterial processes. The protein participates in GTP-dependent positioning of the initiator tRNA and helps establish the reading frame for accurate protein synthesis. In M. gallisepticum, which causes significant respiratory disease in poultry, infB may have unique structural features adapted to the organism's parasitic lifestyle and growth conditions. The pathogen's ability to cause respiratory disease and production losses in poultry populations worldwide makes understanding its core translational machinery particularly relevant for developing targeted interventions .

How does M. gallisepticum IF-2 differ structurally from IF-2 proteins in other bacterial species?

M. gallisepticum IF-2 exhibits several structural differences compared to IF-2 proteins from other bacterial species, reflecting adaptations to the organism's minimalist genome and parasitic lifestyle. While maintaining the conserved GTP-binding domain common to all IF-2 proteins, M. gallisepticum IF-2 shows variability in its N-terminal domain, which typically contains species-specific sequences. The protein likely contains modifications that support M. gallisepticum's ability to invade host cells, a property demonstrated in studies showing this pathogen can enter both human epithelial cells and chicken embryo fibroblasts within 20 minutes of exposure . The structural adaptations may also relate to the organism's temperature sensitivity, a feature exploited in attenuated vaccines like ts-11. Unlike IF-2 proteins from free-living bacteria, M. gallisepticum IF-2 may have evolved features that allow it to function efficiently under the resource-limited conditions of intracellular parasitism, potentially affecting ribosome binding affinity or interaction with other translation factors. Researchers should note that these structural differences may impact expression strategies when producing recombinant versions of the protein .

How can researchers verify the functional activity of purified recombinant M. gallisepticum IF-2?

Verifying the functional activity of purified recombinant M. gallisepticum IF-2 requires assays that assess its core biochemical functions. The primary activity test should evaluate GTP binding and hydrolysis capabilities using either radioactive [γ-³²P]GTP hydrolysis assays or fluorescent GTP analogs that change spectral properties upon binding. For a more comprehensive functional assessment, researchers should employ in vitro translation assays using either E. coli or mycoplasma-derived ribosomal components to measure IF-2's ability to facilitate initiator tRNA binding to ribosomes. This can be quantified using filter-binding techniques with radiolabeled fMet-tRNA. Circular dichroism spectroscopy provides valuable information about the protein's secondary structure, confirming proper folding essential for function. For researchers investigating M. gallisepticum pathogenesis, cell invasion assays similar to those described for intact M. gallisepticum can determine whether recombinant IF-2 affects host cell interactions . When conducting functional verification, temperature-dependent activity profiling is particularly important, especially when comparing wild-type versus ts-11 vaccine strain-derived IF-2, as temperature sensitivity may reflect functional differences relevant to attenuation. Control experiments should include heat-denatured protein samples and, where possible, comparison with native IF-2 purified directly from M. gallisepticum cultures .

What are the optimal storage conditions for maintaining stability of recombinant M. gallisepticum IF-2?

Maintaining stability of recombinant M. gallisepticum IF-2 during storage requires careful attention to buffer composition, temperature, and handling protocols. Optimal storage conditions include buffer systems containing 50 mM Tris-HCl or HEPES at pH 7.5, supplemented with 150-300 mM NaCl to maintain solubility, 15-20% glycerol as a cryoprotectant, 1-5 mM DTT or 2-mercaptoethanol to prevent oxidation of cysteine residues, and 2-5 mM MgCl₂ to stabilize nucleotide-binding regions. For short-term storage (1-2 weeks), the protein can be maintained at 4°C with minimal loss of activity, while long-term storage requires flash-freezing in liquid nitrogen and storage at -80°C in single-use aliquots to avoid repeated freeze-thaw cycles. Stability testing should include periodic assessment of both structural integrity via circular dichroism and functional activity through GTP binding assays. Researchers should note that addition of protease inhibitors (such as PMSF or commercial cocktails) may be necessary for preparations stored at 4°C. Lyophilization represents an alternative for long-term storage but requires extensive optimization to prevent activity loss during the freeze-drying process. When designing stability studies, researchers should establish a time course of activity retention under various storage conditions, testing at multiple timepoints (0, 1, 2, 4, 8, 12 weeks) to generate reliable stability profiles .

How can recombinant M. gallisepticum IF-2 be used as a target for developing novel antimicrobials against avian mycoplasma infections?

Recombinant M. gallisepticum IF-2 offers significant potential as a target for developing novel antimicrobials against avian mycoplasma infections. The essential nature of translation initiation in bacterial survival makes IF-2 an attractive target, as compounds that selectively inhibit this protein could disrupt protein synthesis in M. gallisepticum while potentially sparing host processes. High-throughput screening methods using purified recombinant IF-2 can identify small molecule inhibitors through assays measuring GTP binding/hydrolysis, interaction with initiator tRNA, or formation of the 30S initiation complex. Researchers should establish a screening cascade that progresses from biochemical assays to cell-based systems using both laboratory-adapted strains and field isolates. Structure-based drug design approaches require solving the crystal or cryo-EM structure of M. gallisepticum IF-2, which can guide rational design of inhibitors targeting unique features of the mycoplasma protein. These could exploit structural differences between bacterial and eukaryotic initiation factors to achieve selectivity. When developing antimicrobial strategies, researchers should assess effects on M. gallisepticum's capacity for epithelial cell invasion, as this ability contributes to pathogenesis and persistence . Additionally, any potential inhibitors should be evaluated in the gentamicin invasion assay to determine if they reduce mycoplasma internalization or affect intracellular survival over 48-hour periods, similar to studies documented with intact organisms .

What role might recombinant M. gallisepticum IF-2 play in developing next-generation vaccines against M. gallisepticum?

Recombinant M. gallisepticum IF-2 has significant potential for next-generation vaccine development through several strategic approaches. As a conserved protein essential for bacterial survival, IF-2 represents a promising subunit vaccine candidate that could elicit protective immunity without the risks associated with live attenuated vaccines. Immunization studies should evaluate different adjuvant formulations to optimize both humoral and cell-mediated immune responses against IF-2. The protein could also be incorporated into existing vaccine platforms; for example, the ts-11 attenuated vaccine strain has already demonstrated capacity as a vector for expressing heterologous proteins like chicken IFN-gamma . A similar approach could incorporate modified IF-2 variants to enhance immunogenicity. When designing vaccination strategies, researchers should monitor both systemic antibody responses through methods like rapid serum agglutination (RSA) and cellular immune responses via IFN-gamma production in spleen cultures, as studies with other M. gallisepticum vaccines have shown these parameters correlate with protection . Notably, research with ts-11 expressing chicken IFN-gamma showed enhanced cellular immunity despite minimal antibody response, suggesting potential for similar immunomodulatory approaches with IF-2-based vaccines . For vaccine efficacy studies, researchers should assess protection against both colonization and clinical disease, and evaluate heterophil infiltration in tracheal epithelium as a marker of protective mucosal immunity.

How might structural studies of recombinant M. gallisepticum IF-2 contribute to understanding mycoplasma evolution and host adaptation?

Structural studies of recombinant M. gallisepticum IF-2 can provide valuable insights into mycoplasma evolution and host adaptation through comparative analyses with translation initiation factors from other organisms. X-ray crystallography or cryo-electron microscopy of purified IF-2 would reveal unique structural features that reflect M. gallisepticum's adaptation to its minimal genome and parasitic lifestyle. Domain architecture comparison between M. gallisepticum IF-2 and homologs from free-living bacteria would highlight modifications that evolved during genome reduction, a hallmark of mycoplasma evolution. The binding interface between IF-2 and initiator tRNA may display adaptations specific to the mycoplasma translational apparatus, potentially explaining how these organisms maintain translation efficiency despite reduced genetic resources. Researchers should combine structural data with molecular dynamics simulations to understand how M. gallisepticum IF-2 functions under various conditions, including temperature ranges relevant to its poultry hosts. When conducting evolutionary analyses, researchers should examine whether structural features correlate with M. gallisepticum's documented ability to invade host cells, a property demonstrated in both human epithelial cells and chicken embryo fibroblasts . This invasive capability distinguishes M. gallisepticum from many other mycoplasmas and may relate to specialized protein functions. The temperature sensitivity of certain M. gallisepticum strains, particularly the ts-11 vaccine strain, likely reflects structural adaptations in key proteins like IF-2, making comparative structural analyses between wild-type and attenuated strains particularly informative .

Can recombinant M. gallisepticum IF-2 be used to develop improved diagnostic assays for M. gallisepticum infection?

Recombinant M. gallisepticum IF-2 holds significant potential for developing improved diagnostic assays with enhanced specificity and sensitivity compared to current methods. ELISA-based detection systems using purified recombinant IF-2 as the capture antigen could detect antibodies produced during M. gallisepticum infection, similar to successful approaches documented with other mycoplasma species like M. suis using recombinant antigens (rMSG1 and rHspA1) . When designing such assays, researchers should evaluate both IgG and IgM responses to capture early and persistent antibody production. Current serological testing for mycoplasmas often relies on crude antigens purified from infected animals, which introduces variability and ethical concerns . Recombinant IF-2-based assays would eliminate these issues, providing standardized reagents independent of animal experiments. For assay development, researchers should determine optimal cutoff values through ROC curve analysis using well-characterized positive and negative field samples. Specificity testing must include sera against related avian mycoplasmas to ensure the assay distinguishes M. gallisepticum from commensal species. For validation, comparison with existing diagnostic methods such as PCR and traditional serology is essential, calculating Cohen's kappa coefficients to assess agreement between tests, similar to the methodology used for validating M. suis recombinant antigen ELISAs . The diagnostic performance should be evaluated using field samples with varying bacterial loads to establish correlations between antibody levels, bacterial detection by PCR, and clinical parameters like respiratory signs or egg production losses.

How can researchers overcome solubility issues when expressing recombinant M. gallisepticum IF-2?

Solubility challenges with recombinant M. gallisepticum IF-2 require systematic optimization strategies addressing multiple expression parameters. Researchers frequently observe inclusion body formation when expressing mycoplasma proteins in E. coli, necessitating modifications to expression conditions. Lowering induction temperature to 16-20°C significantly improves proper folding by slowing protein synthesis, allowing chaperones to assist folding. Similarly, reducing inducer concentration (0.1-0.2 mM IPTG versus standard 1 mM) and expressing in rich media like Terrific Broth rather than LB can enhance soluble yields. For construct design, researchers should consider expressing individual domains separately if the full-length protein proves consistently insoluble. Fusion partners known to enhance solubility, such as maltose-binding protein (MBP), SUMO, or thioredoxin, often outperform simple His-tags for mycoplasma proteins. If inclusion bodies persist despite optimization, controlled refolding protocols using stepwise dialysis from denaturing conditions can recover active protein. For high-throughput optimization, researchers should implement parallel small-scale expressions varying cell lines (BL21, Rosetta, Arctic Express), induction parameters, and growth media compositions, followed by solubility screening via SDS-PAGE of supernatant versus pellet fractions. When encountering persistent solubility issues, co-expression with molecular chaperones like GroEL/ES or DnaK/J/GrpE often proves beneficial for proteins from organisms with different folding environments, like mycoplasmas .

What strategies help resolve contamination issues during purification of recombinant M. gallisepticum IF-2?

Resolving contamination issues during purification of recombinant M. gallisepticum IF-2 requires identifying the nature of contaminants and implementing targeted strategies for their removal. Common contaminants include host cell proteins with affinity for nickel columns, nucleic acids complexed with the target protein, and proteolytic fragments of IF-2 itself. For E. coli proteins that co-purify with His-tagged IF-2, washing IMAC columns with moderate imidazole concentrations (40-60 mM) before elution removes weakly bound contaminants. Including 1-5 mM ATP in wash buffers disrupts interactions with heat shock proteins like DnaK that frequently contaminate recombinant preparations. For nucleic acid contamination, evident as high A260/A280 ratios, increasing NaCl concentration to 500-750 mM in wash buffers disrupts nucleic acid-protein interactions. Treatment with Benzonase nuclease (25 U/mL) during initial lysis effectively degrades contaminating DNA/RNA. To address proteolytic degradation, researchers should add protease inhibitor cocktails during all purification steps and minimize processing time, keeping samples cold throughout. For persistent contamination with similarly sized proteins, orthogonal purification techniques like ion exchange chromatography using a salt gradient can separate proteins based on charge differences. For final polishing, size exclusion chromatography not only removes aggregates but separates contaminants of different molecular weights. Researchers should verify purity through multiple analytical methods including SDS-PAGE, Western blotting, and mass spectrometry to ensure complete removal of all significant contaminants .

How can researchers troubleshoot loss of activity during purification and storage of recombinant M. gallisepticum IF-2?

Activity loss during purification and storage of recombinant M. gallisepticum IF-2 often stems from specific molecular events that can be systematically addressed through targeted interventions. Oxidation of cysteine residues represents a common cause of inactivation, requiring inclusion of reducing agents such as DTT (1-5 mM) or TCEP (0.5-2 mM) in all buffers, with the latter offering greater stability for long-term storage. Nucleotide-binding proteins like IF-2 typically require Mg²⁺ (2-5 mM) for structural stability; its absence can lead to progressive denaturation. Researchers should implement activity assays at each purification step to track specific activity, identifying stages where function declines. When activity loss occurs despite maintaining reducing conditions and essential cofactors, protein aggregation may be responsible, detectable through dynamic light scattering or analytical size exclusion chromatography. For storage, flash-freezing in liquid nitrogen with cryoprotectants (15-20% glycerol or 10% sucrose) minimizes damage from ice crystal formation. Researchers should establish a thermal stability profile using differential scanning fluorimetry to determine optimal buffer conditions that maximize the protein's melting temperature. For difficult preparations, additives like arginine (50-100 mM) can stabilize partially folded intermediates without interfering with downstream applications. When testing functional activity, researchers should employ multiple assay formats including GTP binding, ribosome interaction, and initiator tRNA binding to comprehensively assess different functional aspects that might be differentially affected by purification and storage conditions .

What are the main challenges in designing effective experiments to study interactions between recombinant M. gallisepticum IF-2 and host cellular components?

Studying interactions between recombinant M. gallisepticum IF-2 and host cellular components presents several methodological challenges requiring careful experimental design. The first significant hurdle involves maintaining physiologically relevant conditions that reflect the environment where natural interactions occur. Researchers must determine whether interactions should be studied using purified components or in cellular contexts, with each approach offering distinct advantages. For in vitro studies with purified components, binding partners must be identified through techniques like pull-down assays with recombinant IF-2 as bait against host cell lysates, followed by mass spectrometry identification of interacting proteins. Surface plasmon resonance or isothermal titration calorimetry can then quantify binding kinetics and thermodynamics between IF-2 and identified partners. For cellular studies, researchers face challenges in distinguishing direct interactions from indirect effects, requiring approaches like proximity labeling (BioID or APEX) to identify proteins within nanometer-scale distances of IF-2 in relevant cellular contexts. When investigating IF-2's potential role in host cell invasion, researchers can adapt the gentamicin invasion assay developed for intact M. gallisepticum, including proper controls to distinguish between intracellular and extracellular protein using dual-fluorescence confocal microscopy techniques . All interaction studies should include controls addressing potential artifacts from recombinant protein tags, ideally comparing results with tagged and tag-cleaved versions or using different tag positions (N-terminal versus C-terminal). When interpreting results, researchers must consider that in vitro interactions may not reflect the complex environment of the host-pathogen interface during natural infection .

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.