Skip to main content
  • Research article
  • Open access
  • Published:

Immunoproteomic analysis of the serum IgG response to cell wall-associated proteins of Staphylococcus aureus strains belonging to CC97 and CC151

Abstract

CC97 and CC151 are two of the most common Staphylococcus aureus lineages associated with bovine intramammary infection. The genotype of the infecting S. aureus strain influences virulence and the progression of intramammary disease. Strains from CC97 and CC151 encode a distinct array of virulence factors. Identification of proteins elaborated in vivo will provide insights into the molecular mechanism of pathogenesis of these lineages, as well as facilitating the development of tailored treatments and pan-lineage vaccines and diagnostics. The repertoire of genes encoding cell wall-anchored (CWA) proteins was identified for S. aureus strains MOK023 (CC97) and MOK124 (CC151); MOK023 encoded more CWA proteins than MOK124. Serum collected during an in vivo challenge trial was used to investigate whether the humoral response to cell wall proteins was strain-specific. Immunoproteomic analysis demonstrated that the humoral response in MOK023-infected cows predominantly targeted high molecular weight proteins while the response in MOK124-infected cows targeted medium or low molecular weight proteins. Antigenic proteins were identified by two-dimensional serum blotting followed by mass spectometry-based identification of immunoreactive spots, with putative antigens subsequently validated. The CWA proteins ClfB, SdrE/Bbp and IsdA were identified as immunogenic regardless of the infecting strain. In addition, a number of putative strain-specific imunogens were identified. The variation in antigens produced by different strains may indicate that these strains have different strategies for exploiting the intramammary niche. Such variation should be considered when developing novel control strategies including vaccines, therapeutics and diagnostics.

Introduction

Staphylococcus aureus is a major pathogen associated with bovine intramammary infection (IMI) and is responsible for substantial economic losses in the dairy industry [1, 2]. A wide variety of S. aureus sequence types have been associated with IMI, with clonal complex (CC) 97 and CC151 being two of the most common, globally-distributed, bovine-adapted lineages [3]. The S. aureus genome consists of a core genome, found in all strains of the species, and a variable genome, found in only some strains, with many virulence genes encoded within the variable genome. Strains within a lineage generally have a similar variable genome but this differs from the variable genome of other lineages as the S. aureus restriction-modification system limits horizontal gene transfer between lineages [4]. Significant differences in the virulence gene content of CC97 and CC151 strains have been demonstrated. CC151 strains encode a large number of toxins but few genes encoding cell wall-anchored (CWA) proteins involved in host cell adherence, internalisation and biofilm formation. In contrast, CC97 strains encode few toxins but have an array of genes encoding CWA proteins [5,6,7].

A variety of in vitro studies have demonstrated significant differences between isolates from CC97 and CC151 in a selection of virulence traits, including host cell adherence and internalisation, biofilm production and elicitation of a host immune response [8,9,10,11]. In vivo studies in which cows were challenged with strain Newbould 305 (CC97) or RF122 (CC151), demonstrated that Newbould 305 generally causes mild or sub-clinical mastitis while RF122 gives rise to severe clinical mastitis [12,13,14,15]. Indeed, a recent in vivo study directly compared the host response to S. aureus strains MOK023 (CC97) and MOK124 (CC151). Intramammary challenge with MOK124 resulted in more overt clinical signs, higher somatic cell count, higher milk IL-1β, IL-8 and anti-S. aureus IgG and a greater drop in milk yield compared to challenge with MOK023 [16]. Milk somatic cell transcriptomic analysis demonstrated that each strain induced a characteristic host response [17]. This suggests that these strains may have developed differing strategies for the exploitation of the intramammary niche and may differ in their mode of pathogenicity.

Understanding the impact of strain variation on disease epidemiology, clinical presentation and the host response to bovine IMI is crucial for the development of novel prevention and control strategies. In particular, the development of novel vaccines and/or diagnostic tools would limit the negative impact of this disease on animal health and welfare and enable a reduction in the use of antimicrobials in the dairy industry. Currently, much S. aureus research has focussed on the potential of CWA proteins as protective antigens [18,19,20,21]. These proteins are common vaccine targets due to their location on the surface of the bacterial cell and their role in host cell interaction, nutrient utilisation and immune evasion. However, human S. aureus vaccines based on CWA proteins have to-date failed to provide protection [22]. One recently elaborated explanation for this is that prior exposure to S. aureus may induce an “immune imprint” with subsequent vaccination resulting in the preferential recall of non-protective antibodies. These antibodies can compete with protective antibodies, reducing opsonophagocytosis and further impairing a protective response [23]. Despite this caveat, the identification of immunogenic proteins has a number of advantages; it enables epitope mapping to identify protective protein domains as well as enabling the identification of proteins and virulence factors expressed in vivo, which may be exploitable as therapeutic targets or diagnostic markers. However, S. aureus IMI control strategies must account for the extensive genomic diversity between bovine-adapted strains and lineages. For example, strains belonging to CC151 lack many genes encoding CWA proteins [5, 6] while ST71 strains do not encode the ica operon [5], the protein products of which are responsible for poly-N-acetylglucosamine biosynthesis [24], a major target of one of the few licenced vaccines [25].

In this study a genomic and immunoproteomic approach was used to identify the repertoire of, and interrogate the humoral immune responses to, CWA proteins expressed by CC97 and CC151 strains of S. aureus during bovine IMI. Key cell wall proteins expressed by each strain that elicited an IgG response in infected cows were identified and validated. Comparison of the immunogenic molecules elaborated in vivo by each strain facilitates the identification of pan-strain and strain-specific antigens.

Materials and methods

S. aureus strains and culture conditions

S. aureus strains MOK023 and MOK124 were recovered from milk of cows presenting with mastitis as described previously [26]. Strain-typing demonstrated they belong to ST3170 (CC97) and ST151 (CC151) respectively [5]. S. aureus strains were preserved in Trypticase Soy Broth (TSB) (LabM, Heywood, UK) supplemented with 15% (v/v) glycerol at −80 °C. When required, strains were recovered on Trypticase Soy Agar (TSA) (LabM) at 37 °C overnight. Strains were single colony purified every 7 days onto fresh TSA. For liquid cultures, strains were grown in Trypticase Soy Broth (TSB) at 37 °C and 200 rpm.

Genome assembly and annotation

Whole genome sequence data for MOK023 and MOK124 are available [5, 16], accession numbers SRS775827 and SRS2841713 respectively. Quality control checks were carried out on both forward and reverse reads using the FastQC software [27]. Draft genomes were assembled and annotated as previously described [28]. For each putative protein, subcellular location was predicted using PSORTb v3.0 [29].

Identification of putative CWA proteins

In S. aureus 25 CWA proteins have been described [30]. Two cell wall-anchoring domains exist, each the target of one of two proteolytic enzymes, Sortase A or Sortase B, responsible for anchoring the protein in the cell wall. Substrates of Sortase A have a tripartite motif, including an LPxTG cleavage motif, at the C-terminal while substrates of Sortase B have a NPQTN cleavage motif [31]. Predicted proteins containing either of the sortase cleavage motifs were identified. Subsequently, a hidden markov model, designed to directly predict Gram positive bacterial CWA proteins based on the presence of the tripartite motif [32], was used to predict if proteins carrying a sortase cleavage motif were putative CWA proteins. BLASTP was used for the identification of putative CWA proteins and determining the presence of sequence or structural variation. Structural variation in clfA, isdH and sasA for MOK124 and sasC for MOK023 was confirmed by Sanger sequencing.

Preparation of S. aureus cell wall-associated proteins

S. aureus strains were grown overnight in 5 mL of TSB at 37 °C with 200 rpm orbital shaking. The following morning, the overnight cultures were diluted 1:100 into 250 mL of fresh TSB and incubated at 37 °C, 200 rpm until mid-exponential phase (OD600nm ~0.4). Cultures were then centrifuged at 1000 × g for 30 min and the supernatant removed. Pelleted cells were resuspended in phosphate buffered saline (PBS), adjusted to an OD600nm of 10 and centrifuged at 5000 × g for 2 min. The supernatant was removed and the cell pellets resuspended in Digestion Buffer (30% raffinose in 20 mM MgCl2, 50 mM Tris–HCl, pH7.5) containing 1 X EDTA-free protease inhibitor cocktail (Sigma Aldrich, St Louis, USA). Lysostaphin from Staphylococcus staphylolyticus (Sigma Aldrich) was added to a final concentration of 0.2 mg/mL before incubation at 37 °C for 30 min. Cell wall-associated proteins were harvested by supernatant collection after centrifugation at 5000 × g for 15 min. Nucleic acids were removed by adding 100 X Protease Inhibitor Mix (GE Healthcare, Chicago, USA) and 100 X Nuclease Mix (GE Healthcare) to a final concentration of 1 X and allowing digestion for 50 min at room temperature with frequent mixing. Desalting was carried out using the PD-10 Desalting Column (GE Healthcare), following the gravity flow protocol, as per the manufacturer’s instructions. The elution step was repeated twice. For protein concentration and buffer exchange, 15 mL of prepared cell wall-associated proteins was added to a 3 kDa molecular weight cutoff filter (Amicon, Millipore, Darmstadt, Germany) and centrifuged at 4000 × g until 1 mL of supernatant remained in the unit. The flow through was discarded, and PBS added to bring sample back to 15 mL. The filter device was again centrifuged at 4000 × g until 1 mL of supernatant remained in the unit and flow through discarded. The PBS wash was repeated once and Isoelectric Focusing (IEF) Rehydration buffer (10 mM Tris, 8 M Urea, 2 M Thiourea, 4% (w/v) CHAPS, 1% (v/v) TritonX-100) or Storage buffer (6 M Urea, 2 M Thiourea, 0.1 M Tris–HCl pH 8.6, filter sterilized) was added to bring the sample back to 15 mL. The filter device was centrifuged at 4000 × g until no more flow through passed and the concentrated supernatant removed to a fresh 1.5 mL Eppendorf tube. Protein quantification was performed using the 2-D Quant kit (GE Healthcare), as per the manufacturer’s instructions.

Gel electrophoresis

Protein samples were separated by either one-dimensional (1D) or two-dimensional (2D) SDS-PAGE, as previously described [28]. Briefly, protein samples were prepared for 1D electrophoresis by adding LDS Sample Buffer (NuPAGE, Thermo Scientific, Waltham, USA) to a final concentration of 1 X, boiled for 10 min and separated on 8–12% SDS-PAGE gels using the Mini-PROTEAN Tetra Cell (Bio-Rad, Hercules, USA). Following electrophoresis, proteins were either stained by coomassie dye or immobilised onto PVDF membrane at 20 V for 1.5 h using the Bio-Rad Trans-Blot SD Semi-Dry Electrophoretic Transfer Cell. For 2D electrophoresis, protein samples were adjusted to a concentration of 60 µg in 125 µL of IEF Rehydration Buffer containing 0.8% (w/v) IPG pH 3–10 NL buffer (GE Healthcare), 15 mg/mL DeStreak Reagent (GE Healthcare) and a trace amount of bromophenol. Proteins were separated according to their isoelectric point (pI) using non-linear gradient strips (Immobiline Drystrip pH 3–10 NL; GE Healthcare) on the Agilent (Santa Clara, USA) 3100 OFFGEL Fractionator until a kVh of 8 was reached. Strips were then reduced for 10 min with agitation in Equilibration buffer (30% (v/v) glycerol, 2% (w/v) SDS, 6 M urea, 50 mM Tris–HCL pH 8.8) containing 2% (w/v) dithiothreitol (DTT) followed by alkylation for 10 min with agitation in Equilibration buffer containing 2.5% (w/v) iodoacetamide (IAA). Proteins were then electrophoresed on 10% SDS-PAGE gels using the Mini-PROTEAN Tetra cell (Bio-Rad). Following electrophoresis, gels were either stained by silver nitrate rapid staining, as described by [33], or immobilised onto PVDF membrane, as described above for 1D SDS-PAGE.

Serum blotting

Bovine sera came from a S. aureus intramammary infection trial previously described [16] and consisted of sera from 5 and 4 cows infected with MOK023 and MOK124 respectively. Serum samples were from 5 time-points over the course of infection: pre-infection (day 0) and 7, 14, 21 and 29 days post-infection. Serum from day 29 post-infection was missing for one cow (582) from the MOK124 group.

Membranes were blocked in 10% non-fat, dried milk in PBS for 1 h at room temperature followed by 3 × 10 min washes in PBS. Membranes were then incubated overnight at 4 °C with bovine sera diluted 1:2000 in PBS. Subsequently, membranes were washed in PBS as described above, before incubating in the dark for 2 h at room temperature with horseradish peroxidase (HRP)-conjugated goat anti-cow IgG (Abcam, Cambridge, UK), diluted 1:5000 in filter-sterilised 5% BSA in PBS. The membranes were subsequently washed 4 × 10 min in PBS. The membranes were incubated with SuperSignal West Pico PLUS Chemiluminescent Substrate (Thermo Scientific) and imaged using the Omega Lum C Imaging System (Aplegen, Pleasanton, USA).

Identification of immunogenic proteins

Serum blotted 2D membranes were manually aligned to the corresponding stained 2D gel. Spots identified as immunogenic were excised from the gel and each spot placed in a separate sterile eppendorf tube. Excised spots were prepared for mass spectrometry, with overnight digestion at 37 °C, as described by [34]. Following digestion, the supernatant was harvested and peptides dried in a new sterile tube.

Mass spectrometry analysis

Dried digested samples were resuspended in 0.5% (v/v) TFA before desalting using Pierce™ C18 spin tips (Thermo Scientific). Desalted peptides were resuspended in Loading Solution (0.05% (v/v) TFA, 2% (v/v) acetonitrile) and analysed on a Q Exactive™ Hybrid Quadrupole-Orbitrap™ Mass Spectrometer coupled to an UltiMate™ 3000 RSLCnano System (Thermo Scientific), according to the method of Owens et al. [35]. Mass spectrometry data were analysed using Proteome Discoverer software (v1.4; Thermo Scientific™), against protein databases created for each strain, using the SEQUEST algorithm with the following settings (i) trypsin was selected as the cleavage enzyme with up to 2 missed cleavages allowed (ii) oxidation of methionine was set as a variable modification and (iii) carbamidomethylation of cysteine was set as a fixed modification. Results were filtered using the Percolator module and only medium confidence peptides (False Discovery Rate (FDR) < 0.05) were retained. Further filtering was performed to remove proteins identified by only a single unique peptide. Where more than one protein was detected in a given excised spot, candidate proteins were identified based on (i) predicted location in the cell wall, (ii) predicted molecular weight, (iii) predicted pI and (iv) the number of unique peptides.

Recombinant protein expression and blotting

The plasmids used for recombinant protein expression are listed in Table 1. Plasmids encoding ClfA, ClfB and FnbpB were provided by Prof. Joan Geoghegan, Trinity College Dublin. Plasmids encoding IsdA and SirA were provided by Prof. Michael Murphy, The University of British Columbia while the plasmid encoding SdrD was provided by Prof. Mona Johannessen, The Arctic University of Norway. All plasmids were transformed into BL21 (DE3) competent Escherichia coli (New England BioLabs, Ipswich, USA) and the integrity of the plasmid insert confirmed by Sanger sequencing. Transformed E. coli strains were stored in Luria–Bertani (LB) Broth (Sigma Aldrich) supplemented with 15% (v/v) glycerol at −80 °C until required. Strains were recovered on LB (Sigma Aldrich) plates, containing 100 µg/mL ampicillin or 30 µg/mL kanamycin as required, at 37 °C overnight. For liquid cultures, strains were grown in LB broth, containing appropriate antibiotics as required, at 37 °C with 250 rpm orbital shaking.

Table 1 Plasmids used in this study.

For recombinant protein expression, E. coli strains were inoculated into 5 ml of LB broth, with appropriate antibiotics, and grown at 37 °C, 250 rpm for 4 h. Recombinant protein production was induced for 3 h with IPTG (final concentration given in Table 1). Following induction, cultures were centrifuged at 1000 × g for 30 min and the supernatant removed. Cell pellets were resuspended in PBS and adjusted to the same OD600nm. Cells were then centrifuged at 3500 × g for 3 min, resuspended in 1 X LDS buffer and boiled for 10 min. The whole cell lysates were separated by 1D SDS-PAGE, using 8–12% gels. Gels were stained with coomassie dye or proteins immobilised onto PVDF membrane. Blots were probed with bovine sera as described above. A lysate of untransformed E. coli and transformed, uninduced E. coli were included as controls.

Results

Identification of genes encoding putative CWA proteins of S. aureus MOK023 and MOK124

MOK023 and MOK124 were predicted to encode 19 and 10 intact S. aureus CWA proteins, respectively (Additional file 1). Neither strain encoded cna, bap, sasL, pls or sasX while fnbB, sasG, sasK and sdrD were also missing from the genome of MOK124. MOK023 encoded sdrE while MOK124 encoded bbp, the allelic variant of sdrE. Structural variation, predicted to prevent anchoring of the cognate protein in the cell wall, was identified in sasC for MOK023 and clfA, isdH, sasA, sasC, sdrC and spa for MOK124 (Additional file 1).

Characterisation of the humoral response to S. aureus cell wall-associated proteins from MOK023 and MOK124

Cell wall protein extracts from mid-exponential phase cultures of MOK023 and MOK124 were separated by 1D SDS-PAGE and immobilised onto PVDF membrane. The proteins were probed with serum from each cow infected with that strain. Figure 1A shows the results of the MOK023 proteins probed with serum from cows infected with this strain. The humoral response predominantly targeted high molecular weight proteins, with a number of immunoreactive bands > 80 kDa. All cows in this group also had a reactive band at ~50 kDa. For two cows (506 and 565) an immune response against a number of low molecular weight proteins was also evident. Figure 1B shows the results of the MOK124 cell wall proteins probed with serum from cows infected with this strain. For this group, the induced humoral response primarily targeted low molecular weight proteins, with major immunoreactive bands identified at ~25, ~30 and ~35 kDa. (Figure 1B). A few high molecular weight weakly immunoreactive bands could also be identified (~120, ~130 and ~200 kDa); however, in many cases reactivity did not increase post-infection. Immunoreactive bands could be identified in all cows from both groups prior to intramammary challenge.

Figure 1
figure 1

One-dimensional serum blots of S. aureus cell wall-associated proteins. A MOK023 cell wall-associated proteins probed with sera from MOK023 infected cows and B MOK124 cell wall-associated proteins probed with sera from MOK124 infected cows. Lane 1 = Day 0 pre-infection, Lane 2 = Day 7 post-infection, Lane 3 = Day 14 post-infection, Lane 4 = Day 21 post-infection and Lane 5 = Day 29 post-infection. Equal loading is shown by coomassie-stained gels of cell-wall extracts.

Identification of immunogenic cell wall-associated proteins from S. aureus MOK023 and MOK124

The cell wall-associated proteins of MOK023 and MOK124 were separated by 2D electrophoresis and immobilised onto PVDF membrane. Proteins from both strains were probed with serum collected pre-infection and 14 days post-infection from cows 604 (MOK023 infected) and 504 (MOK124 infected) to identify both strain-specific and common antigens. Spots that increased in intensity post-infection were considered immunogenic.

The results of the 2D silver nitrate stained gels and serum blots of MOK023 proteins probed with sera from cow 604 (MOK023 infected) and cow 504 (MOK124 infected) are shown in Figure 2. As seen in the 1D gels, antibodies raised in cow 604 primarily targeted high molecular weight (> 100 kDa) proteins (Figure 2C); however for cow 504, antibodies reacted against high, medium and low molecular weight proteins (Figure 2F). Antibodies raised in cow 504 also primarily targeted low pI proteins (Figure 2F). Candidate immunogenic antigens from MOK023 were identified by mass spectrometry analysis of spots which showed an increase in immunoreactivity post-infection. Proteins were detected in 27 excised spots, with more than one protein detected in 25 spots. However, CWA proteins could be identified in only 18 spots; the top two candidate CWA proteins identified in each spot are listed in Table 2. Additional file 2 lists all detected proteins in each of the spots.

Figure 2
figure 2

Two-dimensional silver nitrate stained gels and serum blots of MOK023 cell wall-associated proteins. A and D Silver nitrate stained gel of MOK023 cell wall-associated protein extract. Numbers in red indicate the spots where proteins were detected by mass spectrometry. Membranes were probed using day 0 (B & E) and day 14 C and F serum from cow 604 (MOK023 infected) B and C and cow 504 (MOK124 infected) E and F.

Table 2 List of MOK023 (CC97) candidate immunogenic CWA proteins detected by mass spectrometry.

The results of the 2D silver nitrate stained gels and serum blots of the MOK124 proteins probed with sera from cow 504 (MOK124 infected) and cow 604 (MOK023 infected) are shown in Figure 3. Antibodies generated by cow 504 primarily targeted low molecular weight (< 35 kDa) proteins (Figure 3C) in agreement with the 1D analysis; however, antibodies generated by cow 604 primarily targeted high molecular weight (> 100 kDa) proteins (Figure 3F) with few common immunoreactive spots. More immunoreactivity was evident at day 14 post-infection when the MOK124 proteins were probed with serum from the cow infected with the homologous strain compared to the heterologous strain. Candidate immunogenic proteins from MOK124 were identified by mass spectrometry. Of the selected immunoreactive spots, proteins were detected in 13 of the excised spots, with more than one protein detected in 12 of those spots. However, candidate CWA proteins were detected in only 4 spots; the top candidate CWA protein identified in each spot is listed in Table 3. Additional file 3 lists all detected proteins in each of the spots. While reactive antigens were evident when MOK124 cell wall-associated proteins were probed with serum from cow 604, there was insufficient protein in many corresponding spots for candidate identification and no candidate CWA proteins were identified.

Figure 3
figure 3

Two-dimensional silver nitrate stained gel and serum blots of MOK124 cell wall-associated proteins. A and D Silver nitrate stained gel of MOK124 cell wall-associated protein extract. Numbers in red indicate the spots where proteins were detected by mass spectrometry. Membranes were probed using day 0 B and E and day 14 C and F serum from cow 504 (MOK124 infected) B and C and cow 604 (MOK023 infected) E and F.

Table 3 List of MOK124 (CC151) candidate immunogenic cell wall-anchored proteins detected by mass spectrometry.

Immunoreactivity of candidate antigens

The 2D serum blotting identified a number of candidate immunogenic S. aureus CWA proteins. However, peptides matching up to 4 different CWA proteins were found in some spots and identification of specific candidates was complicated by the fact that many CWA proteins have similar molecular weight and pI and include repeat regions. Therefore, validation of candidate CWA proteins was performed. Whole cell lysates of E. coli expressing recombinant ClfA, ClfB, FnbpB, IsdA or SdrD were separated by 1D electrophoresis, immobilised onto PVDF membrane and probed with serum collected pre and post-infection from cows 604 and 504. The results of the serum blots of lysates of untransformed E. coli as well as E. coli expressing recombinant ClfA, are shown in Figure 4A and Additional file 4. ClfA was highly expressed, even in the absence of induction. Sera from both cows demonstrated minimal reactivity with rClfA pre-infection; however, a strong anti-ClfA response was induced post-infection in both cows. Cow 604 induced a weak antibody response to ClfB post-infection that was most evident 21 days post-infection (Figure 4B and Additional file 5). In contrast, cow 504 had antibodies to ClfB pre-challenge indicating prior exposure to ClfB-expressing S. aureus; reactivity increased post-infection, most notably at day 29 (Additional file 5). Both cows had minimal immunoreactivity against FnbpB pre-infection but induced a strong antibody response post-infection (Figure 4C and Additional file 6). For IsdA, strong immunoreactivity was evident for both cows pre-infection and reactivity increased post-infection (Figure 4D and Additional file 7). Both cows also generated anti-SdrD antibodies post-infection, with cow 504 showing evidence of prior exposure to SdrD (Figure 4E and Additional file 8).

Figure 4
figure 4

One-dimensional serum blots of lysates of E. coli BL21 overexpressing recombinant proteins. Lysates of E. coli BL21 overexpressing recombinant ClfA (A), ClfB (B), FnbpB (C), IsdA (D) and SdrD (E) were probed using sera from cow 604 (MOK023 infected) and cow 504 (MOK124 infected). Lane 1 = No plasmid control, Lane 2 = Uninduced control and Lane 3 = Induced with 100 μM IPTG for 3 h. Equal loading is shown by coomassie-stained gels of the whole cell lysates.

Discussion

In this study, a proteomic approach was taken to characterise the immunogenicity of CWA proteins of S. aureus strains MOK023 (CC97) and MOK124 (CC151). Genome sequence analysis demonstrated that MOK023 encoded more CWA proteins than MOK124 due to extensive decay in genes encoding CWA proteins in MOK124. This is in agreement with previous studies of strains belonging to CC151 [6, 7, 36]. Allelic variation in a number of genes encoding CWA proteins was also identified. Such variation may affect their antigenicity, as has been shown for FnbpA and FnbpB [37, 38]. The humoral immune response to cell wall proteins of these contrasting strains was characterised using pre and post-infection serum from a bovine intramammary challenge trial [16]. The results from 1D serum blots showed that the strains induced, to some extent, a strain-specific response. Cows infected with MOK023 produced antibodies that mainly reacted against high molecular weight proteins while cows infected with MOK124 produced antibodies that mainly reacted against low molecular weight proteins. The serum blots also indicated the cows had prior exposure to S. aureus; however, this was expected as the cows used in this study were colonised by S. aureus at extramammary sites at the time of infection (unpublished observation). Despite this, there was a distinct increase in serum reactivity against a number of CWA proteins after intramammary challenge. Interestingly, it was also noted in the serum blots that there was a decline in immunoreactivity to some proteins on or before day 29 post-infection, despite the fact that the cows were still infected. S. aureus is known to cause chronic, persistent infections [39, 40]. Strategies used by S. aureus for evasion of the host immune system include internalisation within host cells and biofilm formation [30, 41,42,43,44]. Thereby, S. aureus can evade intracellular killing by the phagosome, effector molecules and cells of the immune system as well as avoid killing by antibiotics. While S. aureus are internalised or biofilm-associated, limited antigens may be available for recognition by the immune system and this could be responsible for the reduced serum reactivity. Alternatively, within-host adaptation may have resulted in a change in the phenotypic traits expressed in vivo over the course of such a chronic, persistent infection [45].

In order to identify specific immunogenic proteins expressed in vivo by MOK124 and MOK023, 2D electrophoresis followed by serum blotting was used for cell wall protein separation and protein identification. Serum from cows 504 and 604 only were used to probe the cell wall-associated proteins as these 2 cows typified the IgG response within each group. Serum from a single cow from each group was used for the immunoblot analysis rather than a pooled serum sample as the genotype of S. aureus to which each cow had been previously exposed was unknown. Serum from a single cow was therefore considered preferable to identify spots that increased in intensity post-infection. However, the use of serum from a single individual may have resulted in the identification of cow-specific antigens compared to pooled serum, which may have identified a greater diversity of antigens more representative of the response in all cows. Agreement between 1 and 2D serum blotting techniques was seen. In both instances serum from cow 504 reacted with many low molecular weight proteins while serum from cow 604 reacted predominantly with high molecular weight proteins. Notably, antibodies produced by cow 604 could cross react with cell wall-associated proteins produced by MOK124 and vice versa, indicating some common immunoreactive proteins. Common immunogenic CWA proteins identified included ClfB, SdrE/Bbp and IsdA. In addition ClfA, FnbpB and SdrD were identified when MOK023 proteins were probed. Some considerations should be taken into account when interpreting these results. Firstly, CWA proteins were harvested from early to mid exponential phase cultures grown in tryptic soy broth with aeration. These growth conditions were selected as they are conducive to the in vitro expression of a number of important CWA proteins, including the FnBPs and ClfB [46, 47]. However, they do not mimic in vivo growth conditions in the mammary gland. The repertoire of S. aureus CWA proteins expressed in vivo during IMI may differ from those expressed, and hence available for detection, in the present study. Growth of the S. aureus strains in milk or under conditions more similar to those in the mammary gland, such as iron and oxygen restriction, may have revealed additional immunogenic antigens [48]. Secondly, low intensity spots were not excised for mass spectrometry analysis due to the likelihood they would contain insufficient protein for identification. Silver nitrate staining is a sensitive method for protein detection; however, it limits protein identification to spots with medium to high intensity staining. For example, no immunogenic proteins were detected when MOK124 cell wall-associated proteins were probed with serum from cow 604. An alternative, sensitive staining method, with better compatibility with mass spectrometry, may have allowed identification of additional proteins. Thirdly, more than one protein was detected by mass spectromtry in the majority of excised spots. CWA proteins were selected as the most likely immunogenic proteins due to their cell surface expression. However, CWA proteins were not the only type of proteins detected, with many cytoplasmic membrane and cytosolic proteins also identified. This is a common finding; other studies that used enzymatic-dependent cell surface shaving methods also reported cytoplasmic contamination, which may be at least partly attributable to autolysis of S. aureus cells and the subsequent reattachment of cytoplasmic proteins to the cell surface [49,50,51,52]. However, serum blotting of E. coli lysates overexpressing SirA, a cytoplasmic membrane protein commonly found in immunoreactive spots with IsdA, found no immunoreactivity to this protein (data not shown) providing some support for the selection of CWA proteins as candidate immunogens. Future work, including the construction of isogenic S. aureus deletion mutants for each of the candidate immunogens, would facilitate confirmation of the identity of the immunoreactive protein in each of the identified spots.

In order to validate a number of the candidate immunogens, plasmids expressing the CWA proteins ClfA, ClfB, FnbpB, SdrD and IsdA were transformed into E. coli for recombinant protein expression. Lysates containing the recombinant proteins were probed with sera from cows 504 and 604. Both cows had antibodies that reacted against all recombinant proteins. Serum from cow 504 could react with SdrD and FnbpB which was unexpected as MOK124 does not encode sdrD or fnbB. One likely explanation is that antibodies raised against Bbp can cross react with SdrD; these allelic variants are 87.4% identical between the two strains. Similarly, MOK124 encodes fnbA, and antibodies raised against the protein likely cross-reacted with the closely related FnbpB [53]. Non-specific interactions were also seen with some E. coli proteins, which has been reported previously [54, 55]. Purification of the recombinant proteins prior to serum blotting would likely have eliminated the detection of such bands, while also enabling downstream functional characterisation of the candidate immunogens as well as investigation of their interactions. It was notable that expression of some recombinant proteins was seen in the absence of inducer, in particular proteins expressed from pQE30. This was likely caused by promoter leakage due to insufficent copies of the lacI repressor, which is not encoded by pQE30 and is not highly expressed by E. coli BL21 DE3 cells [56].

Infection with MOK023 or MOK124 has been demonstrated to result in contrasting host response and infection signs [16]. The infecting S. aureus strain has also been demonstrated to influence the severity and outcome of bovine IMI in a variety of other studies [40, 45, 57]. These studies suggest that particular strains or genotypes express distinctive phenotypic traits during the course of infection. Therefore, different strains may have different mechanisms by which they infect or colonise the mammary gland and cause disease. In a previous study we demonstrated that MOK124 produced more toxins in vitro than MOK023 and the secreted proteins from MOK124 against which a serum IgG response was targeted during bovine IMI included a number of toxins [28]. The results from this study suggest that the serum IgG response to MOK023 targets many high molecular weight proteins, most likely CWA proteins, further supporting the hypothesis that these strains have contrasting modes of pathogenicity.

A number of the immunogens identified in this study have been previously tested as vaccine candidates designed to prevent S. aureus IMI. The ability of recombinant ClfA + IsdA and ClfA + FnBPA to induce a serum and milk IgG response in vivo has been demonstrated and the functional capacity of the antibodies demonstrated [20, 58]. Vaccine efficacy trials have also indicated the ability of these proteins to provide at least partial protection against S. aureus IMI [21, 59]. The results from the present study indicate that strains from both major bovine-adapted lineages CC97 and CC151 produce these immunogenic antigens in vivo, supporting their investigation as pan-lineage protective antigens. A number of methods, such as peptide arrays or yeast, bacterial or phage display systems, are currently available to determine the epitopes recognised by antibodies. Future work should include the application of such technologies for the identification of immunodominant regions of the candidate CWA proteins, as well as the identification of novel immunogens. However, the extensive sequence variation between strains and lineages will need to be considered with this approach.

The development of novel mastitis management and control strategies requires knowledge of the prevalent pathogens and an understanding of the mechanism by which they cause disease. New strategies must target all major strains or genotypes to maintain cow health and welfare and milk quality as dairy producers rarely have information on the genotype of the infecting strain. Therefore, this study, which may have implications for the development of vaccines against different S. aureus strains and lineages, offers a starting point for further research into the mode of pathogenicity of prevalent bovine-adapted strains as well as the utility of CWA proteins as vaccine or diagnostic candidates. However, some considerations should be made for future work. Quantitative analysis could help to determine if commonly identified proteins between different S. aureus strains share similar immunoreactivity or not. In addition, the ability of the antibodies generated to provide protection from infection must be further explored. Moreover, antibodies produced in response to one strain may cross-react with, but not cross-protect against, other S. aureus strains and this remains to be determined. The role of other antibody classes, particularly locally produced IgA and IgM should be considered as important mammary gland defense and natural antibody classes [60]. Furthermore, additional CC97 and CC151 S. aureus strains, along with strains from other important bovine-adapted lineages, should be investigated to identify common and strain-specific immunogenic proteins.

Availability of data and materials

All data generated or analysed during this study are included in this published article and its additional information files.

References

  1. Geary U, Lopez-Villalobos N, Begley N, McCoy F, O’Brien B, O’Grady L, Shalloo L (2012) Estimating the effect of mastitis on the profitability of Irish dairy farms. J Dairy Sci 95:3662–3673

    CAS  PubMed  Google Scholar 

  2. Rasmussen P, Shaw APM, Muñoz V, Bruce M, Torgerson PR (2022) Estimating the burden of multiple endemic diseases and health conditions using Bayes’ Theorem: a conditional probability model applied to UK dairy cattle. Prev Vet Med 203:105617

    PubMed  PubMed Central  Google Scholar 

  3. Yebra G, Harling-Lee JD, Lycett S, Aarestrup FM, Larsen G, Cavaco LM, Seo KS, Abraham S, Norris JM, Schmidt T, Ehlers MM, Sordelli DO, Buzzola FR, Gebreyes WA, Gonçalves JL, Dos Santos MV, Zakaria Z, Rall VLM, Keane OM, Niedziela DA, Paterson GK, Holmes MA, Freeman TC, Fitzgerald JR (2022) Multiclonal human origin and global expansion of an endemic bacterial pathogen of livestock. Proc Natl Acad Sci U S A 119:e2211217119

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Lindsay JA (2010) Genomic variation and evolution of Staphylococcus aureus. Int J Med Microbiol 300:98–103

    CAS  PubMed  Google Scholar 

  5. Budd KE, McCoy F, Monecke S, Cormican P, Mitchell J, Keane OM (2015) Extensive genomic diversity among Bovine-Adapted Staphylococcus aureus: evidence for a genomic rearrangement within CC97. PLoS One 10:e0134592

    PubMed  PubMed Central  Google Scholar 

  6. Herron-Olson L, Fitzgerald JR, Musser JM, Kapur V (2007) Molecular correlates of host specialization in Staphylococcus aureus. PLoS One 2:e1120

    PubMed  PubMed Central  Google Scholar 

  7. Monecke S, Slickers P, Ehricht R (2008) Assignment of Staphylococcus aureus isolates to clonal complexes based on microarray analysis and pattern recognition. FEMS Immunol Med Microbiol 53:237–251

    CAS  PubMed  Google Scholar 

  8. Bardiau M, Caplin J, Detilleux J, Graber H, Moroni P, Taminiau B, Mainil JG (2016) Existence of two groups of Staphylococcus aureus strains isolated from bovine mastitis based on biofilm formation, intracellular survival, capsular profile and agr-typing. Vet Microbiol 185:1–6

    CAS  PubMed  Google Scholar 

  9. Budd KE, Mitchell J, Keane OM (2016) Lineage associated expression of virulence traits in bovine-adapted Staphylococcus aureus. Vet Microbiol 189:24–31

    PubMed  Google Scholar 

  10. Murphy MP, Niedziela DA, Leonard FC, Keane OM (2019) The in vitro host cell immune response to bovine-adapted Staphylococcus aureus varies according to bacterial lineage. Sci Rep 9:6134

    PubMed  PubMed Central  Google Scholar 

  11. Zbinden C, Stephan R, Johler S, Borel N, Bünter J, Bruckmaier RM, Wellnitz O (2014) The inflammatory response of primary bovine mammary epithelial cells to Staphylococcus aureus strains is linked to the bacterial phenotype. PLoS One 9:e87374

    PubMed  PubMed Central  Google Scholar 

  12. Kauf AC, Vinyard BT, Bannerman DD (2007) Effect of intramammary infusion of bacterial lipopolysaccharide on experimentally induced Staphylococcus aureus intramammary infection. Res Vet Sci 82:39–46

    CAS  PubMed  Google Scholar 

  13. Schukken YH, Leslie KE, Barnum DA, Mallard BA, Lumsden JH, Dick PC, Vessie GH, Kehrli ME (1999) Experimental Staphylococcus aureus intramammary challenge in late lactation dairy cows: quarter and cow effects determining the probability of infection. J Dairy Sci 82:2393–2401

    CAS  PubMed  Google Scholar 

  14. Wilson GJ, Tuffs SW, Wee BA, Seo KS, Park N, Connelley T, Guinane CM, Morrison WI, Fitzgerald JR (2018) Bovine Staphylococcus aureus superantigens stimulate the entire T cell repertoire of cattle. Infect Immun 86:e00505-e518

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Hensen SM, Pavicić MJ, Lohuis JA, de Hoog JA, Poutrel B (2000) Location of Staphylococcus aureus within the experimentally infected bovine udder and the expression of capsular polysaccharide type 5 in situ. J Dairy Sci 83:1966–1975

    CAS  PubMed  Google Scholar 

  16. Niedziela DA, Murphy MP, Grant J, Keane OM, Leonard FC (2020) Clinical presentation and immune characteristics in first-lactation Holstein-Friesian cows following intramammary infection with genotypically distinct Staphylococcus aureus strains. J Dairy Sci 103:8453–8466

    CAS  PubMed  Google Scholar 

  17. Niedziela DA, Cormican P, Foucras G, Leonard FC, Keane OM (2021) Bovine milk somatic cell transcriptomic response to Staphylococcus aureus is dependent on strain genotype. BMC Genomics 22:796

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Stranger-Jones YK, Bae T, Schneewind O (2006) Vaccine assembly from surface proteins of Staphylococcus aureus. Proc Natl Acad Sci U S A 103:16942–16947

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Zuo QF, Cai CZ, Ding HL, Wu Y, Yang LY, Feng Q, Yang HJ, Wei ZB, Zeng H, Zou QM (2014) Identification of the immunodominant regions of Staphylococcus aureus fibronectin-binding protein A. PLoS One 9:e95338

    PubMed  PubMed Central  Google Scholar 

  20. Misra N, Wines TF, Knopp CL, Hermann R, Bond L, Mitchell B, McGuire MA, Tinker JK (2018) Immunogenicity of a Staphylococcus aureus-cholera toxin A(2)/B vaccine for bovine mastitis. Vaccine 36:3513–3521

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Alabdullah HA, Overgaard E, Scarbrough D, Williams JE, Mohammad Mousa O, Dunn G, Bond L, McGuire MA, Tinker JK (2020) Evaluation of the efficacy of a cholera-toxin-based Staphylococcus aureus vaccine against bovine intramammary challenge. Vaccines 9:6

    PubMed  PubMed Central  Google Scholar 

  22. Fowler VG Jr, Proctor RA (2014) Where does a Staphylococcus aureus vaccine stand? Clin Microbiol Infect 20:66–75

    PubMed  PubMed Central  Google Scholar 

  23. Tsai CM, Caldera JR, Hajam IA, Chiang AWT, Tsai CH, Li H, Díez ML, Gonzalez C, Trieu D, Martins GA, Underhill DM, Arditi M, Lewis NE, Liu GY (2022) Non-protective immune imprint underlies failure of Staphylococcus aureus IsdB vaccine. Cell Host Microbe 30:1163-1172.e6

    CAS  PubMed  PubMed Central  Google Scholar 

  24. O’Gara JP (2007) ica and beyond: biofilm mechanisms and regulation in Staphylococcus epidermidis and Staphylococcus aureus. FEMS Microbiol Lett 270:179–188

    PubMed  Google Scholar 

  25. Prenafeta A, March R, Foix A, Casals I, Costa L (2010) Study of the humoral immunological response after vaccination with a Staphylococcus aureus biofilm-embedded bacterin in dairy cows: possible role of the exopolysaccharide specific antibody production in the protection from Staphylococcus aureus induced mastitis. Vet Immunol Immunopathol 134:208–217

    CAS  PubMed  Google Scholar 

  26. Keane OM, Budd KE, Flynn J, McCoy F (2013) Pathogen profile of clinical mastitis in Irish milk-recording herds reveals a complex aetiology. Vet Rec 173:17

    CAS  PubMed  Google Scholar 

  27. FastQC: A quality control tool for high throughput sequence data. https://www.bioinformatics.babraham.ac.uk/projects/fastqc/

  28. Drumm SD, Cormican P, O’Boyle P, Owens RA, Mitchell J, Keane OM (2022) Immunoproteomic analysis of the secretome of bovine-adapted strains of Staphylococcus aureus demonstrates a strain-specific humoral response. Vet Immunol Immunopathol 249:110428

    CAS  PubMed  Google Scholar 

  29. Yu NY, Wagner JR, Laird MR, Melli G, Rey S, Lo R, Dao P, Sahinalp SC, Ester M, Foster LJ, Brinkman FS (2010) PSORTb 30: improved protein subcellular localization prediction with refined localization subcategories and predictive capabilities for all prokaryotes. Bioinformatics 26:1608–1615

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Foster TJ, Geoghegan JA, Ganesh VK, Höök M (2014) Adhesion, invasion and evasion: the many functions of the surface proteins of Staphylococcus aureus. Nat Rev Microbiol 12:49–62

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Mazmanian SK, Ton-That H, Su K, Schneewind O (2002) An iron-regulated sortase anchors a class of surface protein during Staphylococcus aureus pathogenesis. Proc Natl Acad Sci U S A 99:2293–2298

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Fimereli DK, Tsirigos KD, Litou ZI, Liakopoulos TD, Bagos PG, Hamodrakas SJ (2012) CW-PRED: A HMM-based method for the classification of cell wall-anchored proteins of gram-positive bacteria. In: Maglogiannis Ilias, Plagianakos Vassilis, Vlahavas Ioannis (eds) Artificial intelligence: theories and applications. Springer, Berlin Heidelberg, Berlin, Heidelberg

    Google Scholar 

  33. Chevallet M, Luche S, Rabilloud T (2006) Silver staining of proteins in polyacrylamide gels. Nat Protoc 1:1852–1858

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Shevchenko A, Tomas H, Havlis J, Olsen JV, Mann M (2006) In-gel digestion for mass spectrometric characterization of proteins and proteomes. Nat Protoc 1:2856–2860

    CAS  PubMed  Google Scholar 

  35. Owens RA, O’Keeffe G, Smith EB, Dolan SK, Hammel S, Sheridan KJ, Fitzpatrick DA, Keane TM, Jones GW, Doyle S (2015) Interplay between gliotoxin resistance, secretion, and the methyl/methionine cycle in Aspergillus fumigatus. Eukaryot Cell 14:941–957

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Jamrozy DM, Fielder MD, Butaye P, Coldham NG (2012) Comparative genotypic and phenotypic characterisation of methicillin-resistant Staphylococcus aureus ST398 isolated from animals and humans. PLoS ONE 7:e40458

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Burke FM, McCormack N, Rindi S, Speziale P, Foster TJ (2010) Fibronectin-binding protein B variation in Staphylococcus aureus. BMC Microbiol 10:160

    PubMed  PubMed Central  Google Scholar 

  38. Loughman A, Sweeney T, Keane FM, Pietrocola G, Speziale P, Foster TJ (2008) Sequence diversity in the A domain of Staphylococcus aureus fibronectin-binding protein A. BMC Microbiol 8:74

    PubMed  PubMed Central  Google Scholar 

  39. Peton V, Bouchard DS, Almeida S, Rault L, Falentin H, Jardin J, Jan G, Hernandez D, François P, Schrenzel J, Azevedo V, Miyoshi A, Berkova N, Even S, Le Loir Y (2014) Fine-tuned characterization of Staphylococcus aureus Newbould 305, a strain associated with mild and chronic mastitis in bovines. Vet Res 45:106

    PubMed  PubMed Central  Google Scholar 

  40. Hoekstra J, Zomer AL, Rutten V, Benedictus L, Stegeman A, Spaninks MP, Bennedsgaard TW, Biggs A, De Vliegher S, Mateo DH, Huber-Schlenstedt R, Katholm J, Kovács P, Krömker V, Lequeux G, Moroni P, Pinho L, Smulski S, Supré K, Swinkels JM, Holmes MA, Lam T, Koop G (2020) Genomic analysis of European bovine Staphylococcus aureus from clinical versus subclinical mastitis. Sci Rep 10:18172

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Brouillette E, Grondin G, Shkreta L, Lacasse P, Talbot BG (2003) In vivo and in vitro demonstration that Staphylococcus aureus is an intracellular pathogen in the presence or absence of fibronectin-binding proteins. Microb Pathog 35:159–168

    CAS  PubMed  Google Scholar 

  42. Côté-Gravel J, Malouin F (2019) Symposium review: Features of Staphylococcus aureus mastitis pathogenesis that guide vaccine development strategies. J Dairy Sci 102:4727–4740

    PubMed  Google Scholar 

  43. Bohl LP, Isaac P, Breser ML, Orellano MS, Correa SG, Tolosa de Talamoni NG, Porporatto C (2021) Interaction between bovine mammary epithelial cells and planktonic or biofilm Staphylococcus aureus: The bacterial lifestyle determines its internalization ability and the pathogen recognition. Microb Pathog 152:104604

    CAS  PubMed  Google Scholar 

  44. Cucarella C, Tormo MA, Knecht E, Amorena B, Lasa I, Foster TJ, Penadés JR (2002) Expression of the biofilm-associated protein interferes with host protein receptors of Staphylococcus aureus and alters the infective process. Infect Immun 70:3180–3186

    PubMed  PubMed Central  Google Scholar 

  45. Marbach H, Mayer K, Vogl C, Lee JYH, Monk IR, Sordelli DO, Buzzola FR, Ehling-Schulz M, Grunert T (2019) Within-host evolution of bovine Staphylococcus aureus selects for a SigB-deficient pathotype characterized by reduced virulence but enhanced proteolytic activity and biofilm formation. Sci Rep 9:13479

    PubMed  PubMed Central  Google Scholar 

  46. Greene C, McDevitt D, Francois P, Vaudaux PE, Lew DP, Foster TJ (1995) Adhesion properties of mutants of Staphylococcus aureus defective in fibronectin-binding proteins and studies on the expression of fnb genes. Mol Microbiol 17:1143–1152

    CAS  PubMed  Google Scholar 

  47. Ní Eidhin D, Perkins S, Francois P, Vaudaux P, Höök M, Foster TJ (1998) Clumping factor B (ClfB), a new surface-located fibrinogen-binding adhesin of Staphylococcus aureus. Mol Microbiol 30:245–257

    PubMed  Google Scholar 

  48. Le Maréchal C, Jan G, Even S, McCulloch JA, Azevedo V, Thiéry R, Vautor E, Le Loir Y (2009) Development of serological proteome analysis of mastitis by Staphylococcus aureus in ewes. J Microbiol Methods 79:131–136

    PubMed  Google Scholar 

  49. Nandakumar R, Nandakumar MP, Marten MR, Ross JM (2005) Proteome analysis of membrane and cell wall associated proteins from Staphylococcus aureus. J Proteome Res 4:250–257

    CAS  PubMed  Google Scholar 

  50. Dreisbach A, Hempel K, Buist G, Hecker M, Becher D, van Dijl JM (2010) Profiling the surfacome of Staphylococcus aureus. Proteomics 10:3082–3096

    CAS  PubMed  Google Scholar 

  51. Solis N, Larsen MR, Cordwell SJ (2010) Improved accuracy of cell surface shaving proteomics in Staphylococcus aureus using a false-positive control. Proteomics 10:2037–2049

    CAS  PubMed  Google Scholar 

  52. Monteiro R, Hébraud M, Chafsey I, Chambon C, Viala D, Torres C, Poeta P, Igrejas G (2015) Surfaceome and exoproteome of a clinical sequence type 398 methicillin resistant Staphylococcus aureus strain. Biochem Biophys Rep 3:7–13

    PubMed  PubMed Central  Google Scholar 

  53. Speziale P, Pietrocola G (2020) The multivalent role of fibronectin-binding proteins A and B (FnBPA and FnBPB) of Staphylococcus aureus in host infections. Front Microbiol 11:2054

    PubMed  PubMed Central  Google Scholar 

  54. Robbins JB, Myerowitz L, Whisnant JK, Argaman M, Schneerson R, Handzel ZT, Gotschlich EC (1972) Enteric bacteria cross-reactive with Neisseria meningitidis groups A and C and Diplococcus pneumoniae types I and 3. Infect Immun 6:651–656

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Lee CJ, Koizumi K, Henrichsen J, Perch B, Lin CS, Egan W (1984) Capsular polysaccharides of nongroupable streptococci that cross-react with pneumococcal group 19. J Immunol 133:2706–2711

    CAS  PubMed  Google Scholar 

  56. Kwon SK, Kim SK, Lee DH, Kim JF (2015) Comparative genomics and experimental evolution of Escherichia coli BL21(DE3) strains reveal the landscape of toxicity escape from membrane protein overproduction. Sci Rep 5:16076

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Vrieling M, Boerhout EM, van Wigcheren GF, Koymans KJ, Mols-Vorstermans TG, de Haas CJ, Aerts PC, Daemen IJ, van Kessel KP, Koets AP, Rutten VP, Nuijten PJ, van Strijp JA, Benedictus L (2016) LukMF’ is the major secreted leukocidin of bovine Staphylococcus aureus and is produced in vivo during bovine mastitis. Sci Rep 6:37759

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Pujato N, Camussone CM, Renna MS, Perrig MS, Morein B, Calvinho LF, Marcipar IS (2018) Evaluation of the humoral immune response to a multicomponent recombinant vaccine against S aureus in healthy pregnant heifers. Vet J 235:47–53

    CAS  PubMed  Google Scholar 

  59. Camussone CM, Reidel IG, Molineri AI, Cicotello J, Miotti C, Suarez Archilla GA, Curti CC, Veaute C, Calvinho LF (2022) Efficacy of immunization with a recombinant S aureus vaccine formulated with liposomes and ODN-CpG against natural S aureus intramammary infections in heifers and cows. Res Vet Sci 145:177–187

    CAS  PubMed  Google Scholar 

  60. Ezzat Alnakip M, Quintela-Baluja M, Böhme K, Fernández-No I, Caamaño-Antelo S, Calo-Mata P, Barros-Velázquez J (2014) The immunology of mammary gland of dairy ruminants between healthy and inflammatory conditions. J Vet Med 2014:659801

    PubMed  PubMed Central  Google Scholar 

  61. O’Connell DP, Nanavaty T, McDevitt D, Gurusiddappa S, Höök M, Foster TJ (1998) The fibrinogen-binding MSCRAMM (clumping factor) of Staphylococcus aureus has a Ca2+-dependent inhibitory site. J Biol Chem 273:6821–6829

    PubMed  Google Scholar 

  62. Perkins S, Walsh EJ, Deivanayagam CC, Narayana SV, Foster TJ, Höök M (2001) Structural organization of the fibrinogen-binding region of the clumping factor B MSCRAMM of Staphylococcus aureus. J Biol Chem 276:44721–44728

    CAS  PubMed  Google Scholar 

  63. Burke FM, Di Poto A, Speziale P, Foster TJ (2011) The A domain of fibronectin-binding protein B of Staphylococcus aureus contains a novel fibronectin binding site. FEBS J 278:2359–2371

    CAS  PubMed  Google Scholar 

  64. Grigg JC, Vermeiren CL, Heinrichs DE, Murphy ME (2007) Haem recognition by a Staphylococcus aureus NEAT domain. Mol Microbiol 63:139–149

    CAS  PubMed  Google Scholar 

  65. Askarian F, Ajayi C, Hanssen AM, van Sorge NM, Pettersen I, Diep DB, Sollid JU, Johannessen M (2016) The interaction between Staphylococcus aureus SdrD and desmoglein 1 is important for adhesion to host cells. Sci Rep 6:22134

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Grigg JC, Cheung J, Heinrichs DE, Murphy ME (2010) Specificity of Staphyloferrin B recognition by the SirA receptor from Staphylococcus aureus. J Biol Chem 285:34579–34588

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We would like to acknowledge technical staff in Teagasc Grange, UCD and Maynooth University for invaluable assistance with this study.

Funding

This study was funded by a grant from Teagasc, the Agriculture and Food Development Authority (0048GE). Q-Exactive mass spectrometry instrumentation was funded by a competitive infrastructure award from Science Foundation Ireland (SFI) [12/RI/2346 (3)] to Maynooth University (Prof. Sean Doyle).

Author information

Authors and Affiliations

Authors

Contributions

SDD performed genome assembly, annotation, immunoproteomics, mass spectrometry sample preparation and data analysis. PC provided mentoring and supervision of all bioinformatic work. RAO advised on mass spectrometry sample preparation and analysis and performed all mass spectrometry techniques. JM and OMK conceived the initial idea of the study and participated in its design and coordination. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Orla M. Keane.

Ethics declarations

Ethics approval and consent to participate

The study utilised samples collected during a previous study which was reviewed and approved by the University College Dublin Animal Research Ethics Committee (protocol number Leonard AREC 16 44).

Competing interests

The authors declare that they have no competing interests.

Additional information

Handling editor: Tom McNeilly.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1: Repertoire of known cell wall anchored proteins in the genome of MOK023 and MOK124.

Additional file 2: Mass spectrometry data for all MOK023 proteins identified in spots excised from 2D gels of MOK023 surface proteins.

Equivalent blots were probed with serum from cow 604 (MOK023 infected) and cow 504 (MOK124 infected) to identify immunogenic proteins.

Additional file 3: Mass spectrometry data for all MOK124 proteins identified in spots excised from 2D gels of MOK124 surface proteins.

Equivalent blots were probed with serum from cow 504 (MOK124 infected) and cow 604 (MOK023 infected) to identify immunogenic proteins.

Additional file 4: One-dimensional serum blots of lysates of E. coli BL21 (DE3) overexpressing recombinant ClfA.

Membranes were probed using sera from cow 604 (MOK023 infected) and cow 504 (MOK124 infected).

Additional file 5: One-dimensional serum blots of lysates of E. coli BL21 (DE3) overexpressing recombinant ClfB.

Membranes were probed using sera from cow 604 (MOK023 infected) and cow 504 (MOK124 infected).

Additional file 6: One-dimensional serum blots of lysates of E. coli BL21 (DE3) overexpressing recombinant FnbpB.

Membranes were probed using sera from cow 604 (MOK023 infected) and cow 504 (MOK124 infected).

Additional file 7: One-dimensional serum blots of lysates of E. coli BL21 (DE3) overexpressing recombinant IsdA.

Membranes were probed using sera from cow 604 (MOK023 infected) and cow 504 (MOK124 infected).

Additional file 8: One-dimensional serum blots of lysates of E. coli BL21 (DE3) overexpressing recombinant SdrD.

Membranes were probed using sera from cow 604 (MOK023 infected) and cow 504 (MOK124 infected).

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Drumm, S.D., Cormican, P., Owens, R.A. et al. Immunoproteomic analysis of the serum IgG response to cell wall-associated proteins of Staphylococcus aureus strains belonging to CC97 and CC151. Vet Res 54, 79 (2023). https://0-doi-org.brum.beds.ac.uk/10.1186/s13567-023-01212-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://0-doi-org.brum.beds.ac.uk/10.1186/s13567-023-01212-7

Keywords