SAR1419 is a Staphylococcus aureus protein annotated as a putative branched-chain amino acid carrier protein. While distinct from SAR1418, its recombinant form has been characterized in research contexts:
Recombinant SAR1419 is utilized in structural and functional studies to elucidate its role in bacterial metabolism and potential as a therapeutic target.
Recent studies highlight recombinant S. aureus proteins as critical components in experimental vaccines. For example:
This vaccine includes five antigens:
α-hemolysin (Hla)
Staphylococcal enterotoxin B (SEB)
Staphylococcal protein A (SpA)
Iron surface determinant B (IsdB-N2)
Key Findings from Preclinical Trials:
Protection: rFSAV reduced bacterial loads and pathology in murine sepsis and pneumonia models .
Immune Response: Induced antibodies neutralizing Hla toxicity and blocking SpA-mediated B-cell suppression .
Cross-Strain Efficacy: Demonstrated broad protection against diverse S. aureus strains .
SpA (Protein A):
SarA (Staphylococcal Accessory Regulator A):
Antigen Selection: Prior failures (e.g., StaphVAX, V710) underscore the need for antigens that avoid SpA-mediated antibody sequestration .
Adjuvant Strategies: rFSAV’s success highlights the importance of combining antigens that neutralize toxins (Hla) and counteract immune evasion (SpA) .
The absence of SAR1418-specific data in the provided sources suggests:
SAR1418 may be a less-studied or hypothetical protein.
Potential nomenclature discrepancies (e.g., SAR1419 vs. SAR1418).
A need for targeted studies to characterize SAR1418’s structure, function, and therapeutic relevance.
Validate SAR1418’s existence via genomic databases (e.g., UniProt, NCBI).
Conduct structural studies (X-ray crystallography, NMR) to resolve its TelA-like domain architecture.
Explore its role in antibiotic resistance or virulence using knockout strains.
SAR1418, as a TelA-like protein in S. aureus, likely plays a regulatory role similar to other SAR proteins identified in the S. aureus genome. Based on homology to other regulatory proteins in S. aureus, it may be involved in coordinating gene expression during different stages of infection. S. aureus pathogenesis involves a complex process requiring coordinated expression of cell wall adhesins during initial colonization and toxin production during tissue invasion phases . SAR1418 could potentially function within regulatory networks that control these virulence factors, similar to how the SarA protein family regulates virulence gene expression in response to changing microenvironments . The protein may exhibit DNA-binding properties, potentially featuring a winged helix structure common to many regulatory proteins in S. aureus . To determine its precise function, researchers should consider complementation studies in SAR1418 knockout mutants, followed by virulence phenotype assessment.
While the specific structure of SAR1418 has not been fully characterized, structural analysis approaches used for other S. aureus proteins provide a methodological framework. Many regulatory proteins in S. aureus, particularly those in the SarA family, feature winged helix structures with DNA-binding capabilities . Sequence alignment and structural prediction tools would be valuable first steps to identify conserved domains in SAR1418. Structural studies of SarA, SarR, SarS, and MgrA have revealed that these proteins possess variations on the winged helix motif that are critical for DNA binding and function . Based on these findings, researchers should focus on identifying potential helix-turn-helix motifs and wing regions in SAR1418 that might be important for its function. X-ray crystallography or NMR studies would provide definitive structural information and should be pursued to fully understand SAR1418's mechanism of action.
To study SAR1418 expression patterns, researchers should consider multiple complementary approaches. Quantitative RT-PCR remains the gold standard for measuring transcript levels across different growth phases and environmental conditions. Similar to studies of SarA family proteins, researchers should examine SAR1418 expression during exponential and post-exponential growth phases, as many virulence-associated genes show growth phase-dependent regulation . Reporter gene fusions (such as lacZ or GFP) to the SAR1418 promoter would enable real-time monitoring of expression in response to environmental stimuli. Western blotting with anti-SAR1418 antibodies would confirm protein-level expression changes. When designing these studies, researchers should consider that, like SarV, SAR1418 might be expressed at low or undetectable levels under standard laboratory conditions but could be significantly induced under specific conditions related to virulence or stress response .
For optimal recombinant SAR1418 production, researchers should evaluate multiple expression systems. E. coli remains the first-choice expression host due to rapid growth and high protein yields. BL21(DE3) or its derivatives are recommended for initial attempts, using vectors with inducible promoters like pET systems. For S. aureus proteins that pose folding challenges in E. coli, yeast expression systems have proven effective. Pichia pastoris or Saccharomyces cerevisiae may yield properly folded SAR1418 with mammalian-like post-translational modifications if required . Expression optimization should include testing multiple growth temperatures (15-37°C), inducer concentrations, and induction times. For SAR1418, similar to approaches used for CHIPS protein, expression in the 29-149 amino acid range with an N-terminal His-tag would facilitate purification while maintaining protein function . Cell-free expression systems represent an alternative for proteins that are toxic to cellular hosts or prone to inclusion body formation.
Purification of recombinant SAR1418 to >90% purity suitable for functional studies requires a multi-step approach. Initial capture via immobilized metal affinity chromatography (IMAC) using Ni-NTA resin is recommended if the protein is expressed with a His-tag . For SAR1418 expressed without tags, ion exchange chromatography based on the protein's theoretical isoelectric point should be employed. Size exclusion chromatography as a polishing step will separate different oligomeric states and remove aggregates. Each purification step should be optimized by analyzing fractions via SDS-PAGE. For functional studies, researchers must verify that the recombinant SAR1418 retains its native conformation using circular dichroism or limited proteolysis. If SAR1418 functions as a DNA-binding protein similar to SarA family proteins, electrophoretic mobility shift assays (EMSA) with potential target DNA sequences would confirm functionality . Researchers should aim for >90% purity as achieved with other S. aureus proteins for accurate functional characterization .
Comprehensive quality assessment of recombinant SAR1418 requires multiple analytical techniques. SDS-PAGE analysis provides basic information about purity and apparent molecular weight, while mass spectrometry confirms the exact mass and sequence integrity . Dynamic light scattering (DLS) should be employed to evaluate sample homogeneity and detect aggregation. Thermal shift assays can assess protein stability and help optimize buffer conditions for maximal stability. For SAR1418's functional integrity, DNA-binding assays would be critical if it functions similarly to other regulatory proteins in S. aureus . Activity assays should be designed based on the predicted function of SAR1418, which may include promoter binding studies if it acts as a transcriptional regulator. Size exclusion chromatography coupled with multi-angle light scattering (SEC-MALS) provides definitive information about the oligomeric state of the protein in solution, which is particularly important for proteins that function as dimers or higher-order complexes, as seen with many SarA family proteins .
To identify the SAR1418 regulon, researchers should implement complementary genomic approaches. Chromatin immunoprecipitation followed by sequencing (ChIP-seq) represents the gold standard for identifying direct DNA-binding sites of regulatory proteins in vivo. This approach requires either a specific antibody against SAR1418 or expression of epitope-tagged SAR1418 in S. aureus. RNA-seq analysis comparing wild-type and SAR1418 mutant strains under various conditions would reveal genes differentially expressed in the absence of SAR1418. Similar approaches have successfully identified genes regulated by SarA family proteins . Researchers should examine both exponential and post-exponential growth phases, as many virulence factors show growth phase-dependent regulation in S. aureus . Transcriptomic data should be validated using quantitative RT-PCR for select genes and reporter gene fusions. Researchers should consider that, like other regulatory proteins in S. aureus, SAR1418 might function within complex regulatory networks, potentially interacting with two-component regulatory systems to coordinate virulence gene expression .
Investigation of SAR1418 structure-function relationships requires systematic mutagenesis coupled with functional assays. Site-directed mutagenesis of conserved residues identified through sequence alignment with other SAR proteins should target potential DNA-binding regions. Based on structural studies of SarA family proteins, researchers should focus on basic residues within the α3-α4 helix-turn-helix motifs and conserved residues in the β2-L-β3 wing region, which are critical for DNA binding . Each mutant should be assessed for DNA-binding capability using EMSAs and for the ability to complement SAR1418 mutant phenotypes. Similar mutation studies with SarA revealed that some residues are crucial for DNA binding while others affect protein function without altering DNA binding . X-ray crystallography of SAR1418 in complex with target DNA sequences would provide definitive information about protein-DNA interactions. Hydrogen-deuterium exchange mass spectrometry (HDX-MS) offers an alternative approach to identify regions involved in DNA binding or protein-protein interactions when crystallization proves challenging.
Resolving contradictory functional data about SAR1418 requires rigorous experimental design and careful consideration of genetic backgrounds. When contradictory results emerge across studies, researchers should first examine methodological differences, including strain backgrounds, growth conditions, and assay parameters. Studies of SarA family proteins have shown that results can vary significantly between laboratory strains and clinical isolates, as demonstrated with SarS regulation . To resolve such discrepancies, researchers should test SAR1418 function across multiple clinically relevant strains rather than relying solely on laboratory strains like RN4220, which has been heavily mutagenized . Complementation studies using plasmid-borne SAR1418 should include controls to ensure expression levels match physiological conditions. Generation of clean deletion mutants using allelic exchange, rather than insertional inactivation, would minimize polar effects on downstream genes that might confound interpretations. Independent validation using complementary techniques and collaboration between laboratories would strengthen consensus findings about SAR1418 function.
SAR1418 likely integrates into the complex virulence regulatory networks in S. aureus through specific protein-protein interactions or competitive promoter binding. To map these interactions, researchers should employ yeast two-hybrid screens or co-immunoprecipitation followed by mass spectrometry to identify protein binding partners. Based on studies of SarA family proteins, potential interactors might include other regulatory proteins or components of two-component regulatory systems, which collectively control virulence gene expression in S. aureus . Researchers should investigate whether SAR1418 functions synergistically or antagonistically with established regulators like Agr, SarA, and MgrA. Competition binding assays can determine if SAR1418 competes with other regulators for the same promoter regions, similar to how SarA displaces SarR from the agr promoter . Genetic approaches involving double mutations (SAR1418 plus known regulators) would reveal epistatic relationships within the regulatory network. Researchers should examine how environmental signals modulate these interactions, as many regulatory proteins in S. aureus respond to specific microenvironmental cues during infection .
Investigating SAR1418's potential role in antibiotic resistance requires a multi-faceted approach. Comparative transcriptomics of wild-type and SAR1418 mutant strains exposed to subinhibitory antibiotic concentrations would reveal whether SAR1418 regulates genes involved in resistance mechanisms. If SAR1418 functions similarly to MgrA, which regulates autolysis and antibiotic tolerance, it might influence cell wall metabolism and susceptibility to cell wall-active antibiotics . Minimum inhibitory concentration (MIC) determinations across multiple antibiotic classes would identify specific resistance phenotypes associated with SAR1418 mutation. Time-kill assays would reveal dynamics of bacterial killing in the presence or absence of SAR1418. Researchers should examine whether SAR1418 influences the expression of efflux pumps, similar to how MgrA regulates norA expression in S. aureus. If SAR1418 affects cell wall metabolism like SarV, it might influence susceptibility to cell wall-active antibiotics through modulation of autolytic activity . Studies in diverse clinical isolates, including MRSA strains, would be essential to establish the clinical relevance of any identified resistance mechanisms.
Structural information about SAR1418 could guide rational drug design strategies targeting this protein. X-ray crystallography or cryo-electron microscopy should be employed to determine the three-dimensional structure at high resolution. If SAR1418 functions as a DNA-binding protein similar to SarA family members, researchers should focus on identifying small molecules that interfere with critical DNA-binding interfaces . Virtual screening of compound libraries against the SAR1418 structure could identify potential inhibitors for experimental validation. Fragment-based drug discovery approaches, beginning with smaller chemical fragments that bind with lower affinity, could be optimized into higher-affinity lead compounds. Researchers should consider that many SarA family proteins contain a conserved cysteine residue that functions as an oxidation sensor in MgrA . If present in SAR1418, this feature could be exploited for the design of redox-active compounds that specifically target this protein. Surface plasmon resonance or isothermal titration calorimetry would provide quantitative binding data for hit compounds. Lead optimization should focus on compounds that specifically inhibit SAR1418 function without affecting mammalian proteins, to minimize toxicity concerns.
Addressing solubility issues with recombinant SAR1418 requires systematic optimization of expression and buffer conditions. For expression optimization, researchers should test multiple fusion tags beyond His-tag, including solubility-enhancing tags like MBP, SUMO, or Thioredoxin. Co-expression with molecular chaperones (GroEL/GroES, DnaK/DnaJ) often improves folding of challenging proteins. For proteins prone to inclusion body formation, lower expression temperatures (15-20°C) and reduced inducer concentrations typically enhance soluble yields. If SAR1418 persistently forms inclusion bodies, researchers can implement refolding strategies from solubilized inclusion bodies using gradual dialysis against decreasing concentrations of denaturants. Buffer optimization should include screening of pH values (pH 5-9), salt concentrations (0-500 mM NaCl), and addition of stabilizing agents (glycerol, arginine, or specific detergents). High-throughput thermal shift assays can rapidly identify buffer conditions that maximize protein stability. If SAR1418 contains cysteine residues, like many SarA family proteins , addition of reducing agents (DTT or TCEP) might prevent aggregation caused by non-native disulfide formation.
Overcoming expression barriers for SAR1418 in heterologous systems requires addressing multiple potential limitations. Codon optimization for the expression host is essential, as S. aureus has a different codon usage bias compared to common expression hosts like E. coli. Researchers should analyze the SAR1418 sequence for rare codons and either optimize the sequence or use strains supplemented with rare tRNA genes. For proteins toxic to the expression host, tight control of expression using repressible promoters or expression in C41/C43 E. coli strains designed for toxic protein expression is recommended. If SAR1418 requires specific post-translational modifications, eukaryotic expression systems like Pichia pastoris might be more successful than bacterial systems . For proteins that affect host cell viability, cell-free expression systems bypass cellular toxicity while providing properly folded protein. If SAR1418 forms insoluble inclusion bodies despite optimization attempts, researchers can employ direct extraction and purification under denaturing conditions followed by controlled refolding, similar to approaches used for other challenging bacterial proteins. Expression as a fusion with a self-cleaving intein might improve solubility while allowing tag-free protein recovery.
Validating antibody specificity for SAR1418 requires comprehensive controls to ensure reliable detection. The gold standard control is comparing antibody reactivity between wild-type S. aureus and an isogenic SAR1418 deletion mutant, which should show absence of the specific band in the mutant. Researchers should test antibody specificity against purified recombinant SAR1418 alongside total protein extracts from S. aureus. Pre-adsorption controls, where the antibody is pre-incubated with purified SAR1418 before immunoblotting, should eliminate specific signals if the antibody is truly specific. For polyclonal antibodies, affinity purification against immobilized recombinant SAR1418 will enrich for specific antibodies while reducing background reactivity. When developing monoclonal antibodies, epitope mapping ensures recognition of accessible regions in the native protein. Cross-reactivity testing against related proteins from the same family is essential, particularly if SAR1418 shares sequence homology with other S. aureus proteins. Researchers should validate the antibody across multiple detection methods (Western blotting, immunofluorescence, ELISA) to ensure consistent performance. All validation data should be documented and reported to enhance reproducibility across research groups.