Recombinant Staphylococcus aureus Uncharacterized protein SAR0322 (SAR0322)

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder.
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes for customized preparation.
Lead Time
Delivery times vary depending on the purchase method and location. Consult your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard blue ice packs. Dry ice shipping requires advance notice 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 serves as a guideline.
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 formulations 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. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The specific tag type is determined during production. Please specify your desired tag type for preferential development.
Synonyms
SAR0322Protein-ADP-ribose hydrolase; EC 3.2.1.-
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-266
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Staphylococcus aureus (strain MRSA252)
Target Names
SAR0322
Target Protein Sequence
METLKSNKAR LEYLINDMRR ERNDNDVLVM PSSFEDLWEL YRGLANVRPA LPVSDEYLAV QDAMLSDLNR QHVTDLKDLK PIKGDNIFVW QGDITTLKID AIVNAANSRF LGCMQANHDC IDNIIHTKAG VQVRLDCAEI IRQQGRNEGV GKAKITRGYN LPAKYIIHTV GPQIRRLPVS KLNQDLLAKC YLSCLKLADQ QSLNHIAFCC ISTGVFAFPQ DEAAEIAVRT VESYLKETNS TLKVVFNVFT DKDLQLYKEA FNRDAE
Uniprot No.

Target Background

Function
This protein specifically reverses SirTM-mediated mono-ADP-ribosylation of GcvH-L by releasing ADP-ribose from the target protein. It may also modulate the response to host-derived oxidative stress.
Database Links

KEGG: sar:SAR0322

Q&A

What is SAR0322 and what is its significance in Staphylococcus aureus research?

SAR0322 (also referred to as folC in some literature) is an uncharacterized protein in Staphylococcus aureus that has gained research interest due to its potential involvement in drug resistance mechanisms. The protein is identified in the genomic annotations of S. aureus strains, particularly in strain MRSA252 (BX571856) . Research suggests that SAR0322 may function as folylpolyglutamate synthase, an enzyme involved in folate metabolism, which is critical for bacterial growth and survival. Understanding this protein's structure and function may provide insights into S. aureus pathogenesis and potential drug targets.

How does SAR0322 relate to other characterized proteins in S. aureus?

While SAR0322 remains largely uncharacterized, genomic analysis places it within the context of other S. aureus genes. Unlike well-characterized regulatory proteins such as SarA (which is present at approximately 50,000 copies per cell and functions as a DNA-binding protein responsive to redox state) , SAR0322's role in cellular processes is still being elucidated. Comparative genomic studies have identified SAR0322 as an ortholog found across different S. aureus strains, suggesting evolutionary conservation that may indicate functional importance .

What experimental approaches are typically used to study uncharacterized proteins like SAR0322?

Several methodological approaches are employed to study uncharacterized proteins:

  • Recombinant protein expression and purification: Expression in systems like E. coli or yeast to obtain sufficient quantities for biochemical and structural studies.

  • Structural analysis: X-ray crystallography, NMR spectroscopy, or cryo-EM to determine three-dimensional structure.

  • Sequence analysis: Bioinformatic approaches to identify conserved domains and predict function.

  • Gene knockout/mutation studies: Creating deletion mutants to observe phenotypic changes.

  • Protein-protein interaction studies: Yeast two-hybrid, co-immunoprecipitation, or protein microarrays to identify binding partners.

  • Transcriptomic and proteomic analyses: RNA-Seq and mass spectrometry to understand expression patterns and regulation.

These approaches collectively help determine the protein's function and significance within bacterial systems.

What evidence suggests SAR0322 may be involved in antimicrobial resistance mechanisms?

Genome-wide association studies have identified mutations in SAR0322 that correlate with drug resistance phenotypes. Specifically, the mutation H201YQE in SAR0322 has been associated with a significant drug resistance score (4.96, p-value 8.72e-04) . This statistical association suggests that alterations in this protein may contribute to resistance mechanisms. The approach used to identify this association involves:

  • Collection of genotype data from multiple S. aureus strains

  • Determination of drug resistance profiles through susceptibility testing

  • Statistical analysis to associate specific mutations with resistance phenotypes

  • Validation through comparative genomics and functional studies

This methodological pipeline, known as GWAMAR (Genome-Wide Assessment of Mutations Associated with drug Resistance), has been used to identify genetic determinants of resistance in bacterial pathogens .

How can researchers experimentally validate the role of SAR0322 mutations in antimicrobial resistance?

Validating SAR0322's role in antimicrobial resistance requires multiple experimental approaches:

  • Site-directed mutagenesis: Introducing specific mutations (e.g., H201YQE) into susceptible strains to determine if resistance is conferred.

  • Complementation studies: Expressing wild-type SAR0322 in resistant mutant strains to assess restoration of susceptibility.

  • Biochemical assays: Characterizing enzyme activity of wild-type versus mutant proteins to identify functional differences.

  • Structural biology: Determining how mutations affect protein structure and potential interactions with antimicrobial compounds.

  • Transcriptomic analysis: Examining changes in gene expression patterns associated with SAR0322 mutations.

  • In vivo infection models: Testing virulence and antibiotic response in animal models using isogenic strains differing only in SAR0322 sequence.

These approaches collectively provide robust evidence for the protein's role in resistance mechanisms rather than merely statistical associations.

What methodological approaches are recommended for expressing and purifying recombinant SAR0322 for structural studies?

Optimization of recombinant SAR0322 expression and purification typically involves:

  • Expression system selection: While E. coli is commonly used, some researchers opt for yeast expression systems for S. aureus proteins to ensure proper folding .

  • Vector optimization: Incorporating appropriate tags (e.g., His-tag) to facilitate purification while minimizing interference with protein function.

  • Expression conditions: Optimizing temperature, inducer concentration, and duration to maximize yield of soluble protein.

  • Purification strategy:

    • Initial capture using affinity chromatography (e.g., Ni-NTA for His-tagged proteins)

    • Further purification using ion exchange and size exclusion chromatography

    • Quality assessment by SDS-PAGE and Western blotting

  • Protein quality assessment: Evaluating purity (>90% is typically desired), homogeneity, and functional activity before proceeding to structural studies.

For crystallization studies, special attention should be paid to protein stability and homogeneity, potentially requiring buffer optimization through differential scanning fluorimetry or thermal shift assays.

How might the potential function of SAR0322 as folylpolyglutamate synthase be investigated experimentally?

To investigate SAR0322's putative function as folylpolyglutamate synthase:

  • Enzymatic activity assays:

    • Measure substrate conversion (dihydrofolate to tetrahydrofolate)

    • Monitor glutamylation of folate compounds using HPLC or mass spectrometry

    • Compare kinetic parameters with known folylpolyglutamate synthases

  • Complementation studies:

    • Express SAR0322 in bacterial strains with folC deletions

    • Assess restoration of growth in folate-depleted conditions

  • Structural comparisons:

    • Perform homology modeling with known folylpolyglutamate synthases

    • Identify conserved catalytic residues and substrate-binding sites

  • Inhibitor studies:

    • Test known folylpolyglutamate synthase inhibitors against purified SAR0322

    • Evaluate competitive binding using techniques like isothermal titration calorimetry

  • Metabolomic analysis:

    • Compare folate metabolite profiles in wild-type versus SAR0322 mutant strains

    • Identify accumulation or depletion of specific folate species

Such experimental validation is critical since sequence-based functional predictions require biochemical confirmation.

How is SAR0322 expression regulated in S. aureus, and how does this compare to known regulatory networks?

While specific information about SAR0322 regulation is limited, we can draw parallels with well-characterized regulatory systems in S. aureus:

  • Global regulators: Major regulatory systems in S. aureus include SarA and agr, which control virulence factor expression . SAR0322 expression may be influenced by these global regulators, particularly if it contributes to pathogenesis.

  • Response to environmental conditions: Like SarA, which responds to redox states , or SrrAB, which senses changes in the cellular redox environment , SAR0322 expression might be modulated by environmental factors relevant to infection sites.

  • Transcriptional profiling: RNA-Seq analysis comparing wild-type and regulatory mutant strains (ΔsarA, Δagr, etc.) could reveal whether SAR0322 is part of these regulons. Similar approaches have identified 390 mRNAs and 51 sRNAs differentially expressed in a ΔsarA mutant .

  • Promoter analysis: Identifying potential binding sites for known transcription factors through bioinformatic approaches and validating through techniques like ChIP-Seq, which has revealed 354 mRNAs and 55 sRNA targets of SarA in the S. aureus genome .

Understanding SAR0322's place in regulatory networks provides context for its potential role in pathogenesis and antimicrobial resistance.

What methods are most effective for studying the expression patterns of SAR0322 across different growth conditions and infection models?

To comprehensively study SAR0322 expression patterns:

  • Quantitative transcriptomics:

    • RNA-Seq to measure transcript levels across growth phases, nutrient conditions, and stress responses

    • Single-cell RNA-Seq to assess expression heterogeneity within bacterial populations

    • RT-qPCR for targeted validation of expression changes

  • Translational analysis:

    • Ribosome profiling to assess translation efficiency

    • Mass spectrometry-based proteomics to quantify protein levels

    • Western blotting with specific antibodies for targeted protein quantification

  • Reporter systems:

    • Transcriptional fusions of the SAR0322 promoter with reporter genes (e.g., GFP, luciferase)

    • Translational fusions to monitor both transcriptional and post-transcriptional regulation

  • In vivo expression:

    • Analysis of expression during infection using animal models

    • Ex vivo infection models using human tissues

    • Dual RNA-Seq to simultaneously monitor host and pathogen gene expression

  • Environmental variables to test:

    • Oxygen limitation (anaerobic vs. aerobic growth)

    • Nutrient availability (rich vs. minimal media)

    • pH variations (acidic vs. neutral environments)

    • Antibiotic exposure (sub-inhibitory concentrations)

    • Growth phase (exponential vs. stationary)

These methodological approaches provide a comprehensive view of when and where SAR0322 functions during infection.

Could SAR0322 be a potential target for vaccine development against S. aureus infections?

Evaluating SAR0322's potential as a vaccine target requires consideration of several factors:

  • Conservation and expression:

    • Analysis of genomic data indicates SAR0322 is conserved across S. aureus strains , which is desirable for broad-spectrum protection.

    • Expression levels during infection would need to be confirmed, as successful vaccine antigens must be accessible to the immune system during infection.

  • Immunogenicity assessment:

    • Recombinant SAR0322 could be tested for ability to elicit antibody responses in animal models.

    • Bioinformatic epitope prediction tools could identify potential B-cell and T-cell epitopes.

  • Protective capacity:

    • Challenge studies in vaccinated animal models to assess reduction in bacterial burden.

    • Functional assays to determine if antibodies against SAR0322 can neutralize its activity or promote opsonophagocytosis.

  • Comparative analysis with failed vaccine targets:

    • Previous S. aureus vaccine candidates like V710 (IsdB) and StaphVAX (CP5/CP8) have failed in clinical trials , highlighting the need to understand mechanisms of protection beyond antibody generation.

    • Multi-component approaches may be necessary, as demonstrated by rFSAV, which includes five S. aureus antigens .

  • Safety considerations:

    • Ensuring SAR0322 or its derivatives do not induce harmful immune responses, a concern highlighted by increased mortality observed with some experimental vaccines .

Current vaccine development approaches for S. aureus emphasize multiple antigens and both humoral and cellular immune responses, suggesting SAR0322 would likely be considered as part of a combination vaccine strategy.

What methodological approaches would be recommended to investigate SAR0322's role in S. aureus pathogenesis?

A comprehensive investigation of SAR0322's role in pathogenesis would include:

  • Genetic manipulation:

    • Creation of SAR0322 deletion mutants using techniques like allelic exchange

    • Complementation with wild-type and mutant versions of the gene

    • CRISPR-Cas9 approaches for precise genome editing

  • In vitro virulence assays:

    • Adherence to host cells or extracellular matrix components

    • Biofilm formation capacity compared to wild-type strains

    • Resistance to host defense mechanisms (e.g., antimicrobial peptides, oxidative stress)

  • Ex vivo models:

    • Survival in human blood or serum

    • Interaction with immune cells (e.g., neutrophils, macrophages)

    • Tissue explant infection models

  • Animal infection models:

    • Systemic infection models to assess dissemination and organ burden

    • Specialized models for specific infections (e.g., skin abscess, pneumonia, endocarditis)

    • Monitoring bacterial load, host response, and disease progression

  • Host response analysis:

    • Cytokine profiles in response to wild-type versus mutant strains

    • Recruitment of immune cells to infection sites

    • Adaptive immune response development

  • Multi-omics approaches:

    • Transcriptomics of both pathogen and host during infection

    • Metabolomics to identify altered metabolic pathways

    • Proteomics to assess changes in protein expression and post-translational modifications

These methodologies collectively provide a comprehensive understanding of SAR0322's contribution to S. aureus virulence and pathogenesis.

How can contradictory data about SAR0322 function be reconciled in experimental design?

Addressing contradictory findings about SAR0322 requires systematic experimental approaches:

  • Strain-specific differences:

    • Compare SAR0322 sequence and expression across different S. aureus strains (laboratory vs. clinical isolates)

    • Assess function in multiple genetic backgrounds to determine if effects are strain-dependent

  • Conditional functionality:

    • Test function under diverse environmental conditions (pH, temperature, oxygen levels)

    • Consider growth phase-dependent effects, as seen with SarA, which remains at constant levels but shows different activity based on growth phase

  • Redundancy and compensation:

    • Identify potential compensatory mechanisms that may mask phenotypes in single-gene studies

    • Construct double or triple mutants to uncover redundant functions

  • Technical considerations:

    • Standardize experimental protocols across research groups

    • Utilize multiple methodological approaches to confirm findings

    • Consider artifacts from expression systems or purification methods

  • Data integration:

    • Combine results from genomic, transcriptomic, and proteomic studies

    • Use computational modeling to reconcile seemingly contradictory observations

    • Apply Bayesian statistical approaches to weight evidence from different studies

  • Collaborative validation:

    • Establish multi-laboratory validation studies for key findings

    • Share reagents (e.g., antibodies, recombinant proteins, mutant strains) to ensure consistency

This systematic approach helps distinguish genuine biological complexity from technical artifacts or strain-specific effects.

What are the potential applications of SAR0322 structure-function studies in developing new antimicrobial strategies?

Understanding SAR0322's structure and function could lead to novel antimicrobial approaches:

  • Structure-based drug design:

    • If confirmed as folylpolyglutamate synthase, SAR0322's structure could guide development of selective inhibitors

    • Molecular docking studies to identify potential binding pockets

    • Fragment-based approaches to develop lead compounds

  • Allosteric modulation:

    • Identifying regulatory sites distinct from the active site

    • Designing molecules that lock the protein in inactive conformations

  • Antimicrobial resistance mechanisms:

    • Understanding how mutations like H201YQE affect function and resistance

    • Developing inhibitors that remain effective against resistant variants

  • Combination therapies:

    • Identifying synergistic targets in related metabolic pathways

    • Designing multi-target approaches to reduce resistance development

  • Alternative approaches:

    • Immunomodulatory strategies that target host-pathogen interactions involving SAR0322

    • Anti-virulence approaches if SAR0322 contributes to pathogenesis rather than essential functions

  • Translational considerations:

    • Assessing conservation across other pathogenic species for broad-spectrum potential

    • Evaluating specificity to minimize disruption of human microbiome

These applications build upon current antimicrobial development strategies while potentially addressing limitations of existing approaches.

What bioinformatic pipelines are recommended for analyzing SAR0322 in the context of S. aureus genomic data?

A comprehensive bioinformatic analysis of SAR0322 would include:

  • Sequence analysis pipeline:

    • Multiple sequence alignment across diverse S. aureus strains

    • Phylogenetic analysis to understand evolutionary relationships

    • Identification of conserved domains and motifs

    • Prediction of post-translational modifications

  • Structural prediction approaches:

    • Homology modeling based on related proteins with known structures

    • Ab initio modeling for unique regions

    • Molecular dynamics simulations to assess conformational flexibility

    • Prediction of protein-protein interaction interfaces

  • Genomic context analysis:

    • Examination of operonic structure and nearby genes

    • Identification of potential regulatory elements in promoter regions

    • Comparative genomics across S. aureus strains and related species

    • Analysis of horizontal gene transfer patterns

  • Integration with experimental data:

    • Incorporation of RNA-Seq data to identify co-expressed genes

    • ChIP-Seq analysis to identify potential regulators

    • Integration with proteomic data to validate expression

  • Resistance-associated mutation analysis:

    • Implementation of GWAMAR-like pipelines to associate mutations with phenotypes

    • Structural mapping of resistance-associated mutations

    • Molecular modeling to predict functional impacts of mutations

This integrated bioinformatic approach provides context for experimental studies and generates testable hypotheses about SAR0322 function.

How can researchers design experiments to distinguish between correlation and causation when studying SAR0322 mutations associated with drug resistance?

Establishing causative relationships between SAR0322 mutations and drug resistance requires:

  • Genetic modification approaches:

    • Introduce specific mutations (e.g., H201YQE) into drug-susceptible strains

    • Revert mutations in resistant strains back to wild-type

    • Use allelic exchange or CRISPR-Cas9 for precise genomic modifications

  • Phenotypic characterization:

    • Comprehensive antimicrobial susceptibility testing before and after genetic modifications

    • Growth kinetics analysis under various antibiotic concentrations

    • Competition assays between wild-type and mutant strains in the presence of antibiotics

  • Biochemical validation:

    • Express and purify wild-type and mutant proteins

    • Compare enzymatic activities and binding affinities

    • Structural studies to understand molecular mechanisms of resistance

  • Clinical correlation:

    • Screening clinical isolates for the presence of identified mutations

    • Correlating mutation frequency with treatment outcomes

    • Longitudinal studies tracking mutation emergence during therapy

  • Statistical approaches:

    • Multivariate analysis to control for confounding genetic factors

    • Bayesian networks to model causal relationships

    • Propensity score matching when analyzing clinical data

  • Reproducibility considerations:

    • Testing in multiple strain backgrounds

    • Validation across different laboratories

    • Publication of negative results to avoid publication bias

This methodological framework helps establish whether SAR0322 mutations directly cause resistance or are merely markers of resistant lineages.

How does SAR0322 compare functionally and structurally to other characterized S. aureus proteins involved in drug resistance?

A comparative analysis reveals distinct characteristics of SAR0322 compared to other S. aureus resistance proteins:

  • Comparison with established resistance determinants:

    • Unlike PBP2a (mecA), which directly reduces affinity for β-lactam antibiotics , SAR0322's potential role appears more metabolic if it functions as folylpolyglutamate synthase

    • Unlike membrane transporters that actively efflux antibiotics, SAR0322 likely affects cellular metabolism or antibiotic modification

  • Structural comparisons:

    • Putative enzymatic function differs from structural proteins like PBP2a that alter cell wall synthesis

    • Domain organization and catalytic mechanisms would be distinct from transport proteins

    • Conservation patterns across strains may reflect different selective pressures compared to surface proteins

  • Evolutionary analysis:

    • Assessment of selection pressure (dN/dS ratios) compared to established resistance proteins

    • Analysis of horizontal gene transfer patterns versus vertical inheritance

    • Identification of co-evolving residues that maintain functional interactions

  • Regulatory context:

    • Unlike resistance genes often found on mobile genetic elements, SAR0322 appears to be chromosomally encoded

    • Integration with core metabolic networks versus specialized resistance mechanisms

    • Potential interactions with global regulators like SarA or SrrAB

  • Metabolic integration:

    • Connection to folate metabolism pathways distinct from direct antibiotic targets

    • Potential indirect effects on cellular physiology compared to specific resistance mechanisms

This comparative perspective places SAR0322 in the broader context of S. aureus resistance mechanisms and highlights its unique characteristics.

What methodological approaches should be used to study potential interactions between SAR0322 and other regulatory proteins in S. aureus?

Investigating protein-protein interactions involving SAR0322 requires multiple complementary approaches:

  • Affinity-based methods:

    • Co-immunoprecipitation using antibodies against SAR0322 or potential partners

    • Tandem affinity purification to identify stable complexes

    • Bacterial two-hybrid systems to screen for interactions

    • Pull-down assays with recombinant tagged proteins

  • Proximity-based approaches:

    • Bacterial two-hybrid or split-protein complementation assays

    • Chemical cross-linking followed by mass spectrometry

    • Proximity labeling methods (e.g., BioID, APEX) adapted for bacterial systems

  • Biophysical techniques:

    • Surface plasmon resonance to measure binding kinetics

    • Isothermal titration calorimetry for thermodynamic parameters

    • Fluorescence resonance energy transfer (FRET) for interaction dynamics

    • Analytical ultracentrifugation to characterize complex formation

  • Structural studies of complexes:

    • X-ray crystallography of co-crystallized proteins

    • Cryo-electron microscopy for larger complexes

    • NMR spectroscopy for mapping interaction interfaces

  • Genetic approaches:

    • Suppressor mutation screening to identify functional interactions

    • Synthetic genetic arrays to map genetic interactions

    • Epistasis analysis between SAR0322 and regulatory genes

  • Systems biology integration:

    • Correlation of expression patterns across conditions

    • Network analysis to identify potential functional associations

    • Integration of interactome data with phenotypic studies

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.