Recombinant Listeria monocytogenes serotype 4b Protein translocase subunit SecA 2 (secA2), 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 in order notes for customized fulfillment.
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 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 be used as a reference.
Shelf Life
Shelf life depends on several factors, including storage conditions, buffer components, 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 to prevent repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The tag type is determined during production. If a specific tag type is required, please inform us, and we will prioritize its development.
Synonyms
secA2; LMOf2365_0612Protein translocase subunit SecA 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
Listeria monocytogenes serotype 4b (strain F2365)
Target Names
secA2
Uniprot No.

Target Background

Function
SecA2 is a component of the Sec protein translocase complex, interacting with the SecYEG preprotein conducting channel. It plays a crucial role in coupling ATP hydrolysis to protein translocation across the cell membrane, functioning as an ATP-driven motor that facilitates the stepwise movement of polypeptide chains across the membrane.
Database Links
Protein Families
SecA family
Subcellular Location
Cell membrane; Peripheral membrane protein; Cytoplasmic side. Cytoplasm.

Q&A

What is the SecA2 protein and how does it differ from the canonical SecA protein in Listeria monocytogenes?

SecA2 is an auxiliary protein secretion system paralog found in Listeria monocytogenes and eight other Gram-positive pathogens. Unlike the essential canonical SecA, SecA2 is nonessential but plays a crucial role in secreting a specific subset of proteins that contribute to bacterial virulence. The SecA2 system operates in parallel to the main Sec pathway, which remains the predominant translocation system for crossing the cytoplasmic membrane in Gram-positive bacteria .

While both SecA and SecA2 participate in protein translocation, they exhibit substrate specificity differences. The standard SecA handles the majority of secreted proteins with typical N-terminal signal sequences, whereas SecA2 is responsible for the secretion of specific virulence-associated proteins including autolysins such as p60 and NamA, which have been experimentally shown to promote host colonization .

What are the characteristics of Listeria monocytogenes serotype 4b and why is it significant in SecA2 research?

Listeria monocytogenes serotype 4b is particularly significant in clinical and research contexts for several reasons:

  • It accounts for almost all major outbreaks of human listeriosis, despite being less frequently isolated from food than serotypes 1/2a and 1/2b

  • It is associated with the highest case mortality rate among L. monocytogenes serotypes

  • Phylogenetically, serotype 4b is notable for being split between lineage I and lineage II, unlike other serotypes that cluster neatly within specific lineages

  • In terms of SecA2 research, understanding its function in serotype 4b is particularly important given this serotype's epidemiological significance

The distribution of L. monocytogenes serotypes across phylogenetic lineages is as follows:

  • Lineage I: Serotypes 1/2b, 3b, 3c, and some 4b strains

  • Lineage II: Serotypes 1/2a, 1/2c, 3a, and some 4b strains

  • Lineage III: Serotypes 4a and 4c

How does SecA2 contribute to Listeria monocytogenes pathogenesis?

SecA2 contributes to L. monocytogenes pathogenesis through several key mechanisms:

  • Secretion of autolytic enzymes: SecA2 is responsible for the secretion of at least 17 proteins, with the two most abundant being p60 and N-acetylmuramidase (NamA) autolysins, which hydrolyze bacterial peptidoglycan and contribute to host colonization .

  • Cell-to-cell spread: SecA2-deficient (ΔSecA2) bacteria show reduced cell-cell spread in cultured cells, demonstrating SecA2's role in intracellular mobility and infection propagation .

  • Immune evasion: By facilitating the secretion of specific virulence factors, SecA2 may enable L. monocytogenes to subvert host immune responses .

  • In vivo persistence: SecA2-deficient bacteria are rapidly cleared after systemic infection of murine hosts, indicating SecA2's critical role in establishing persistent infection. In competitive index assays, while ΔSecA2 bacteria initially seed tissues as well as wild-type strains, they are subsequently eliminated more rapidly .

  • Morphological impacts: SecA2 mutations lead to bacterial "rough" colony morphology and cell chaining, characteristics that may affect host interaction .

What are the established methods for generating secA2 deletion mutants in Listeria monocytogenes serotype 4b?

Generating secA2 deletion mutants requires precise genetic manipulation techniques. The following methodological approach is recommended based on established protocols:

  • Allelic exchange: Create a deletion construct containing upstream and downstream flanking regions of the secA2 gene fused together, excluding the secA2 coding sequence.

  • Shuttle vector integration: Clone this construct into a temperature-sensitive shuttle vector (such as pKSV7) that contains a selectable marker.

  • Transformation and selection: Transform L. monocytogenes serotype 4b with the construct using electroporation, with initial selection for the antibiotic resistance marker at permissive temperature (30°C).

  • Chromosomal integration: Shift to non-permissive temperature (42°C) while maintaining antibiotic selection to force plasmid integration into the chromosome by homologous recombination.

  • Excision and screening: Release antibiotic selection and passage at permissive temperature to allow second recombination event, then screen for loss of antibiotic resistance.

  • Mutant confirmation: Verify secA2 deletion by PCR and sequencing, and confirm the rough colony phenotype and cell chaining characteristic of ΔsecA2 mutants .

  • Phenotypic validation: Test for known SecA2-deficient phenotypes including reduced swarming motility (approximately 92% reduction compared to wild-type) and susceptibility to lysozyme .

What assays can be used to evaluate the impact of SecA2 deficiency on Listeria monocytogenes virulence?

Multiple complementary assays should be employed to comprehensively assess the impact of SecA2 deficiency on virulence:

In vitro assays:

  • Plaque assay: This cell-to-cell spread assay reveals that ΔsecA2 mutants form plaques approximately 30% the size of wild-type strains, quantifying intracellular spread deficiency .

  • Colony morphology and microscopy: ΔsecA2 mutants display a rough colony morphology correlating with bacterial chaining observed microscopically .

  • Swarming motility assay: Using semi-solid agar, assess motility reduction (ΔsecA2 mutants typically show ~92% reduced swarming compared to wild-type) .

  • Lysozyme susceptibility test: ΔsecA2 mutants demonstrate increased susceptibility to lysozyme compared to typically resistant wild-type strains .

  • Antibiotic susceptibility assay: Test sensitivity to cell wall-acting antibiotics, as ΔsecA2 mutants often show increased susceptibility .

In vivo assays:

  • Competitive index assay: Co-infect mice with equal proportions of erythromycin-resistant (Erm^R) mutant and erythromycin-sensitive (Erm^S) wild-type bacteria, then determine ratios in harvested tissues at various time points post-infection. ΔsecA2 bacteria show equal initial tissue seeding but are subsequently cleared more rapidly .

  • Bacterial burden determination: Quantify bacterial loads in organs (liver, spleen) following infection with ΔsecA2 versus wild-type strains.

  • Survival studies: Monitor survival rates of mice infected with various doses of ΔsecA2 versus wild-type L. monocytogenes.

How can SecA2-dependent secreted proteins be identified in Listeria monocytogenes serotype 4b?

Identifying SecA2-dependent secreted proteins requires a systematic approach combining proteomics with genetic and biochemical validation:

  • Comparative secretome analysis:

    • Culture wild-type and ΔsecA2 mutant strains in minimal media to minimize background protein content

    • Collect and concentrate culture supernatants using trichloroacetic acid precipitation

    • Separate proteins using two-dimensional gel electrophoresis or liquid chromatography

    • Identify differentially secreted proteins by mass spectrometry

  • Protein localization studies:

    • Generate reporter fusions with candidate SecA2-dependent proteins

    • Compare localization patterns between wild-type and ΔsecA2 strains using immunofluorescence microscopy

    • Perform cell fractionation to detect proteins in cytoplasmic, membrane, cell wall, and secreted fractions

  • Genetic complementation:

    • Restore secA2 expression in ΔsecA2 mutants and verify restoration of protein secretion

    • Create point mutations in secA2 to identify critical residues for secretion of specific targets

  • Signal sequence analysis:

    • Analyze N-terminal signal sequences of identified SecA2-dependent proteins for unique features

    • Create signal sequence swaps between SecA2-dependent and SecA-dependent proteins to determine specificity determinants

This methodology has successfully identified 17 SecA2-dependent secreted and surface proteins in L. monocytogenes, with p60 and NamA autolysins being the most abundant .

What genetic interactions exist between secA2 and other components of the secretion machinery in Listeria monocytogenes?

The genetic interactions between secA2 and other secretion machinery components reveal complex functional relationships within the L. monocytogenes secretory pathway:

  • SecA2-SecY interaction: Whole-genome sequencing of ΔsecA2 suppressor mutants identified a mutation in secY (G408R) that partially restores motility and cellular function. This suggests functional interplay between SecA2 and the core SecYEG translocon complex .

  • SecA-SecA2 relationship: A suppressor mutation in the canonical secA gene (D559N) can partially compensate for secA2 deficiency, indicating potential overlapping functions or regulatory relationships between these paralogs .

  • WalRK regulatory system: A mutation in walI (G109R) was identified in a ΔsecA2 suppressor mutant, suggesting the WalRK two-component regulatory system may influence SecA2-dependent secretion .

  • Cell wall biosynthesis genes: Multiple suppressor mutations occurred in genes related to cell wall biosynthesis and modification, including mnaA (UDP-N-acetylglucosamine 2-epimerase) and genes similar to teichoic acid biosynthesis proteins, highlighting the connection between SecA2 function and cell envelope maintenance .

The table below summarizes key suppressor mutations identified in ΔsecA2 mutants and their phenotypic impacts:

StrainNotable mutationEncoded protein% motility (±SD)% plaque area (±SD)Colony morphology
R57G408RSecY112 (±15)87 (±13)Smooth
R60.1D559NSecA87 (±23)21 (±6)Smooth
R9MultipleVarious29 (±3)41 (±9)Rough
R23MultipleVarious55 (±13)0Smooth
R35Multiple*Various102 (±26)0Smooth
ΔsecA2N/AN/A9 (±2)29 (±9)Rough

*R35 contained a mutation in mutS (DNA mismatch repair protein), likely causing hypermutation .

How does the SecA2-dependent secretion mechanism differ between Listeria monocytogenes serotype 4b and other serotypes?

While SecA2 function is conserved across L. monocytogenes serotypes, important differences exist in SecA2-dependent secretion between serotype 4b and other serotypes:

  • Surface autolysin IspC secretion: Evidence suggests that secretion of the surface autolysin IspC is SecA2-independent in L. monocytogenes serotype 4b, unlike in other serotypes . This serotype-specific difference may contribute to the unique virulence characteristics of serotype 4b strains.

  • Promoter activity: The ispC gene in serotype 4b has been shown to have a functional promoter immediately upstream of its open reading frame, as confirmed by promoterless lacZ gene fusion and 5'RACE analysis, potentially affecting its regulation independent of SecA2 .

  • Serotype-specific autolysins: Research using monoclonal antibodies identified IspC as a serotype 4b-specific surface antigen (~77 kDa), with several antibodies showing 100% specificity for serotype 4b isolates . This suggests differences in autolysin expression or localization between serotypes.

  • Stress response differences: Expression of SecA2-dependent proteins may be differentially regulated under stress conditions between serotypes, impacting their adaptation to host environments and food processing conditions .

These differences may help explain why serotype 4b accounts for most foodborne outbreaks and has a higher case mortality rate despite being less frequently isolated from food than other serotypes.

What are the mechanistic explanations for the reduced virulence of secA2 deletion mutants in Listeria monocytogenes?

The reduced virulence of ΔsecA2 mutants stems from multiple interconnected mechanistic deficiencies:

  • Autolysin secretion defects: SecA2 deficiency prevents proper secretion of p60 and NamA autolysins, which are critical for peptidoglycan hydrolysis and remodeling. Studies confirm that restoration of virulence in Δp60 bacteria requires full-length p60 with an intact catalytic domain, indicating that peptidoglycan hydrolysis by p60 is crucial for L. monocytogenes pathogenesis .

  • Cell division and separation impairment: SecA2-dependent autolysins are required for proper separation of daughter cells after division, explaining the observed chaining phenotype in ΔsecA2 mutants. This chaining likely impedes bacterial motility and tissue penetration in vivo .

  • Cell-to-cell spread deficiency: ΔsecA2 mutants show significantly reduced plaque formation (approximately 30% of wild-type plaque size), indicating impaired ability to spread between host cells . This defect limits the bacteria's capacity to disseminate within tissues.

  • Altered cell wall integrity: SecA2 deficiency leads to increased susceptibility to lysozyme and cell wall-acting antibiotics, suggesting compromised cell wall architecture that may expose the bacteria to host defense mechanisms .

  • Immune recognition: Changes in cell surface composition due to altered protein secretion may increase recognition by host immune components, explaining the rapid clearance of ΔsecA2 bacteria in murine models despite successful initial tissue seeding .

  • Cumulative effects: Individual deletion of SecA2-dependent proteins (e.g., Δp60 and ΔNamA) causes intermediate reductions in bacterial virulence in vivo, suggesting that the complete virulence defect of ΔsecA2 mutants results from the combined loss of multiple secreted virulence factors .

What strategies can be employed to express and purify recombinant SecA2 protein from Listeria monocytogenes serotype 4b?

Expressing and purifying recombinant SecA2 from L. monocytogenes serotype 4b requires careful optimization of conditions to obtain functional protein:

  • Expression system selection:

    • E. coli systems: BL21(DE3) or similar strains with T7 RNA polymerase are recommended

    • Vector options: pET-based vectors with N-terminal or C-terminal affinity tags (His6, GST, or MBP)

    • Codon optimization: Consider optimizing the secA2 coding sequence for E. coli expression

  • Expression optimization:

    • Temperature: Lower temperatures (16-20°C) often improve SecA2 solubility

    • Induction conditions: Use lower IPTG concentrations (0.1-0.5 mM) for longer periods (16-20 hours)

    • Media supplementation: Addition of 1% glucose can help reduce basal expression

    • Co-expression considerations: Co-express with molecular chaperones (GroEL/GroES) if solubility is problematic

  • Purification strategy:

    • Initial capture: Immobilized metal affinity chromatography (IMAC) for His-tagged protein

    • Intermediate purification: Ion exchange chromatography (typically Q-Sepharose)

    • Polishing step: Size exclusion chromatography to obtain homogeneous protein

    • Buffer optimization: Include 5-10% glycerol and 1-5 mM DTT or TCEP to maintain stability

  • Protein quality assessment:

    • Purity analysis: SDS-PAGE and Western blotting with SecA2-specific antibodies

    • Structural integrity: Circular dichroism spectroscopy

    • Activity testing: ATPase assays to confirm functionality

  • Storage considerations:

    • Store at -80°C in buffer containing 20-50 mM Tris-HCl (pH 8.0), 100-300 mM NaCl, 10% glycerol, and 1 mM DTT

    • Avoid repeated freeze-thaw cycles by preparing single-use aliquots

When expressing the partial SecA2 construct, carefully select domain boundaries based on structural predictions to ensure proper folding of the recombinant fragment.

How can researchers study the interactions between SecA2 and its substrate proteins in vitro?

Several complementary techniques can be employed to characterize SecA2-substrate interactions:

  • Pull-down assays:

    • Immobilize purified His-tagged SecA2 on Ni-NTA resin

    • Incubate with L. monocytogenes cell lysate or purified candidate substrates

    • Wash extensively and elute bound complexes

    • Identify interacting partners by mass spectrometry or Western blotting

  • Surface plasmon resonance (SPR):

    • Immobilize SecA2 or substrate proteins on sensor chips

    • Measure real-time binding kinetics (k_on, k_off) and calculate affinity constants (K_D)

    • Compare binding parameters between wild-type SecA2 and mutant variants

    • Examples from similar studies show affinity constants ranging from 1.0 × 10^-7 to 6.4 × 10^-9 M for protein interactions

  • Isothermal titration calorimetry (ITC):

    • Directly measure thermodynamic parameters of SecA2-substrate binding

    • Determine binding stoichiometry, enthalpy, and entropy changes

    • Study effects of nucleotides (ATP, ADP) on binding properties

  • Fluorescence-based assays:

    • Label SecA2 and/or substrates with fluorescent probes

    • Measure fluorescence anisotropy or FRET to detect complex formation

    • Monitor real-time changes during translocation processes

  • Crosslinking studies:

    • Use chemical crosslinkers or photo-crosslinking to capture transient interactions

    • Identify crosslinked residues by mass spectrometry

    • Map interaction interfaces between SecA2 and substrates

  • ATPase activity assays:

    • Measure SecA2 ATPase activity in the presence of different substrates

    • Compare stimulation of ATPase activity by SecA2-dependent versus SecA-dependent substrates

    • Evaluate effects of signal sequence modifications on ATPase stimulation

These methods can be particularly valuable for identifying key residues in both SecA2 and its substrates that determine secretion specificity.

What approaches can be used to develop novel antimicrobials targeting the SecA2 pathway in Listeria monocytogenes?

Targeting the SecA2 pathway represents a promising approach for developing Listeria-specific antimicrobials with several strategic advantages:

  • Structure-based inhibitor design:

    • Obtain high-resolution crystal structures of L. monocytogenes SecA2

    • Identify unique structural features distinguishing SecA2 from essential SecA

    • Use in silico docking and molecular dynamics to design selective inhibitors targeting:

      • ATP binding pocket

      • Substrate binding regions

      • SecY interaction interfaces

      • Allosteric regulatory sites

  • High-throughput screening approaches:

    • Develop ATPase activity assays suitable for microplate format

    • Screen chemical libraries against purified SecA2 protein

    • Conduct secondary screens in bacterial cell cultures

    • Validate hit compounds against other SecA2-containing pathogens

  • Peptide-based inhibitors:

    • Design peptides mimicking SecA2-dependent signal sequences

    • Create peptide derivatives with enhanced binding and reduced degradation

    • Develop peptidomimetics with improved pharmacological properties

  • Antibody-based approaches:

    • Generate antibodies against surface-exposed SecA2-dependent proteins

    • Develop antibody-antibiotic conjugates for targeted delivery

    • Create bispecific antibodies targeting multiple SecA2-dependent factors

  • Genetic attenuation strategies:

    • Design secA2 variants with temperature-sensitive mutations

    • Create strains with inducible secA2 expression for vaccine development

    • Engineer bacteria expressing dominant-negative SecA2 variants

  • Evaluation and validation:

    • Test candidate compounds for efficacy against:

      • Multiple L. monocytogenes serotypes, particularly 4b

      • Biofilm formation and persistence

      • Intracellular infection models

    • Assess specificity by testing against:

      • Human Sec machinery

      • Beneficial gut microbiota

      • Other pathogens lacking SecA2

This research direction is particularly valuable as SecA2 inhibitors would specifically target virulence without disrupting essential functions, potentially reducing selective pressure for resistance development.

What are the main challenges in studying SecA2 function in Listeria monocytogenes serotype 4b, and how can they be addressed?

Researchers face several technical challenges when investigating SecA2 in L. monocytogenes serotype 4b:

  • Genetic manipulation difficulties:

    • Challenge: L. monocytogenes serotype 4b strains can be less amenable to transformation than laboratory strains

    • Solution: Optimize electroporation protocols with glycine treatment of cells, adjust field strength parameters, and use methylation-deficient plasmid preparations to improve transformation efficiency

  • Protein secretion complexity:

    • Challenge: Distinguishing SecA2-dependent secretion from canonical Sec pathway is challenging

    • Solution: Employ quantitative proteomics approaches like SILAC (stable isotope labeling with amino acids in cell culture) to precisely compare wild-type and ΔsecA2 secretomes

  • Functional redundancy:

    • Challenge: Partial functional overlap between SecA and SecA2 complicates phenotypic analysis

    • Solution: Generate conditional secA mutants in secA2-null backgrounds to distinguish unique and shared functions

  • Strain-specific variations:

    • Challenge: Variations in SecA2 function between serotype 4b clinical isolates

    • Solution: Compare multiple serotype 4b isolates from diverse sources and outbreaks to establish consistent SecA2-dependent phenotypes

  • In vivo relevance:

    • Challenge: Connecting in vitro findings to in vivo pathogenesis

    • Solution: Develop tissue-specific infection models that simulate natural infection routes for serotype 4b strains

  • Structural analysis limitations:

    • Challenge: Obtaining sufficient quantities of properly folded recombinant SecA2

    • Solution: Explore alternative expression systems including cell-free protein synthesis or baculovirus expression systems for improved yield and folding

  • Understanding suppressor mutations:

    • Challenge: Isolating and characterizing genuine suppressor mutations from adaptive mutations

    • Solution: Employ genome editing technologies to introduce specific suppressor mutations into clean genetic backgrounds for unambiguous phenotypic analysis

How can researchers troubleshoot issues with SecA2-dependent protein secretion assays?

Common challenges in SecA2-dependent secretion assays and their solutions include:

  • Protein contamination from cell lysis:

    • Problem: Cell lysis can release cytoplasmic proteins, confounding secretome analysis

    • Solution: Monitor assays for cytoplasmic marker proteins (e.g., EF-Tu, ribosomal proteins); optimize culture conditions to minimize lysis; use shorter culture times and gentle handling

  • Low abundance of SecA2-dependent proteins:

    • Problem: Some SecA2-dependent proteins may be secreted at low levels

    • Solution: Concentrate culture supernatants; use more sensitive detection methods like LC-MS/MS with multiple reaction monitoring; consider immunoprecipitation to enrich specific targets

  • Inconsistent phenotypes in ΔsecA2 mutants:

    • Problem: Spontaneous suppressor mutations can arise in ΔsecA2 strains

    • Solution: Maintain fresh stocks; verify the ΔsecA2 phenotype before each experiment; sequence key loci (secY, secA) in strains showing unexpected phenotypes

  • Difficulty detecting specific SecA2-dependent proteins:

    • Problem: Lack of specific antibodies for all SecA2-dependent proteins

    • Solution: Generate epitope-tagged versions of target proteins; develop targeted mass spectrometry assays; consider using reporter fusions

  • Media composition effects:

    • Problem: Media components can influence protein secretion profiles

    • Solution: Use defined minimal media for secretome analysis; test multiple media conditions; include appropriate controls for each condition

  • Protein degradation during secretion analysis:

    • Problem: Secreted proteases can degrade other secreted proteins

    • Solution: Include protease inhibitors in collection buffers; analyze samples at multiple time points; consider using protease-deficient strains for initial characterization

  • Cross-contamination between cellular fractions:

    • Problem: Improper fractionation can lead to misleading localization results

    • Solution: Validate fractionation protocols with known markers for each cellular compartment; use multiple complementary fractionation techniques

A systematic approach to troubleshooting, with appropriate controls for each step, is essential for reliable characterization of SecA2-dependent secretion.

What considerations are important when designing experiments to compare SecA2 function across different Listeria monocytogenes strains and serotypes?

Designing robust comparative experiments requires careful consideration of several factors:

  • Strain selection strategy:

    • Include multiple independent isolates of each serotype (especially 4b)

    • Consider epidemic and sporadic isolates separately

    • Include strains from diverse sources (clinical, food, environmental)

    • Ensure proper phylogenetic characterization of all strains

  • Genetic manipulation standardization:

    • Use identical genetic modification strategies across strains

    • Verify mutations by whole genome sequencing to detect any off-target effects

    • Create marker-free deletions when possible to avoid polar effects

    • Include complemented strains using identical expression systems

  • Experimental condition controls:

    • Standardize growth conditions (medium, temperature, growth phase)

    • Account for serotype-specific growth rate differences

    • Test multiple environmental conditions relevant to pathogenesis

    • Include appropriate stress conditions (acid, salt, temperature shifts)

  • Phenotypic assay standardization:

    • Develop quantitative assays with internal standards

    • Blind analysis to prevent experimental bias

    • Perform assays in parallel rather than sequentially

    • Include biological and technical replicates

  • Data analysis considerations:

    • Account for strain-specific baseline differences

    • Use appropriate statistical methods for multiple comparisons

    • Consider both absolute and relative changes in phenotypes

    • Integrate data from multiple assay types

  • Specific assays for cross-strain comparison:

    • Quantitative secretome analysis with identical protein extraction and identification methods

    • Standardized virulence assays in both cell culture and animal models

    • Consistent methods for measuring colony morphology and cell chaining

    • Identical assays for antibiotic and lysozyme susceptibility

  • Genomic context analysis:

    • Compare sequences of secA2 and its substrates across strains

    • Analyze regulatory regions controlling secA2 expression

    • Identify strain-specific genetic factors that might influence SecA2 function

    • Consider horizontal gene transfer events that might affect secretion system components

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.