Skip to main content

Diversity and pathogenesis of Staphylococcus aureus from bovine mastitis: current understanding and future perspectives

Abstract

Staphylococcus aureus is a leading cause of bovine mastitis worldwide. Despite some improved understanding of disease pathogenesis, progress towards new methods for the control of intramammary infections (IMI) has been limited, particularly in the field of vaccination. Although herd management programs have helped to reduce the number of clinical cases, S. aureus mastitis remains a major disease burden. This review summarizes the past 16 years of research on bovine S. aureus population genetics, and molecular pathogenesis that have been conducted worldwide. We describe the diversity of S. aureus associated with bovine mastitis and the geographical distribution of S. aureus clones in different continents. We also describe studies investigating the evolution of bovine S. aureus and the importance of host-adaptation in its emergence as a mastitis pathogen. The available information on the prevalence of virulence determinants and their functional relevance during the pathogenesis of bovine mastitis are also discussed. Although traits such as biofilm formation and innate immune evasion are critical for the persistence of bacteria, the current understanding of the key host-pathogen interactions that determine the outcome of S. aureus IMI is very limited. We suggest that greater investment in research into the genetic and molecular basis of bovine S. aureus pathogenesis is essential for the identification of novel therapeutic and vaccine targets.

Background

Bovine mastitis is a multifactorial inflammatory disease that depends on a combination of animal-, environmental-, and pathogen-related factors. Visible abnormalities in the milk, swelling or tenderness of the udder are signs of clinical mastitis while no overt signs are observed in subclinical mastitis. S. aureus is a well-studied opportunistic pathogen frequently associated with subclinical mastitis and responsible for large economic losses due to reduced milk quality and production [1]. S. aureus spreads among cows during milking, requiring a cooperative approach to reduce dissemination to healthy animals [2]. Bovine isolates of S. aureus are also a leading cause of foodborne diseases with bulk tank and raw milk products important vehicles for bacterial transmission to humans [3].

The importance of the human-animal-environment One Health approach for investigating disease transmission and control has been well highlighted [4] but concrete strategies are still needed to reduce the burden of infectious diseases and the impact of antimicrobial resistance in livestock and humans. Indeed, implementation of the One Health approach is essential to improve animal welfare, enhance food safety, and promote human health. An important consideration is the capacity for transmission of S. aureus among livestock and humans or host-switching events leading to the emergence of new pathogenic or resistant clones [5, 6].

S. aureus of bovine origin has long been a focus of microbiological research. However, the scientific knowledge gained has yet to be translated into effective vaccines, therapeutics or rapid, inexpensive diagnostics that can be applied for better disease control [7]. Limited insights into the pathogenesis of mastitis will result from analysis of S. aureus strains in isolation. It is imperative that we examine the bacterial interaction with the host to identify novel therapeutics to fight this old disease. This review has compiled worldwide studies on the population diversity and virulence of S. aureus of bovine origin that have been published in the last 16 years. Articles describing the clonal complexes (CCs) or spa types identified among bovine S. aureus were included to reveal the worldwide distribution of the pathogen. Overall, there are remarkable differences in virulence profiles among field isolates and some virulence factors display ruminant-specific features like LukMF’ suggesting a potential role during intramammary infection (IMI). Studies that shed light on the mechanism of virulence factors made by S. aureus from bovine IMI are described with emphasis on adhesins and toxins whose function has been experimentally demonstrated (Fig. 1).

Fig. 1
figure 1

Proteins that have been implicated in the pathogenesis of Staphylococcus aureus of bovine origin. Surface proteins interact with host proteins to promote bacterial adhesion and invasion. Biofilm-associated protein (Bap) interferes in the internalization pathway mediated by FnBP and contributes to biofilm formation, a process that also depends on the ica locus. Toxins such as alpha and beta hemolysins, promote necrosis of the mammary gland tissue and lyse bovine erythrocytes to use hemoglobin as a source of iron. Leukotoxin LukMF´ binds to neutrophils present in the bovine milk. Staphylococcal superantigens activate T-cells resulting in the release of various pro-inflammatory cytokines. Figure created by authors using resources from Smart Servier Medical Art and BioRender

Main text

Worldwide distribution of clones

From 2004 to early 2021, 79 articles were published on the epidemiology of bovine S. aureus isolated in 31 countries. However, the 67 articles that used multilocus sequence typing (MLST) or spa typing, two commonly used methods for the molecular typing of S. aureus isolates, were included in this review (Table 1). CC97 was the most disseminated genotype, identified in 15 different countries of America, Asia, Europe, and Africa. CC97 has been reported as the dominant lineage in Chile [8], Brazil [9, 10], Japan [11], and the USA [8]. It has been reported that IMI cases caused by CC97 strains lead to asymptomatic, subclinical or persistent infections, increasing the challenge of pathogen control in dairy herds [11, 12]. Based on genomic comparison, a higher virulence potential was predicted for CCs 151 and 97 compared to other bovine-associated S. aureus lineages [13], a finding that is consistent with functional and in vivo studies carried out [14, 15].

Table 1 Clonal complexes and spa types of bovine isolates of Staphylococcus aureus disseminated worldwide

CC1 and CC5 also have a global distribution, each being reported among cows in ten countries. Although these lineages are associated with human infections [77, 78] they have recently emerged in cows via host switch events [79]. It is reported that CC1 is less likely to cause clinical mastitis in cows [80]. The CC126, CC130, and CC133 strains were less widely distributed, in four, two, and six countries, respectively. CC133 and CC130 were localized in Europe with CC130 found in Germany and the UK and CC133 identified in four countries (Portugal, Italy, Germany, and Norway) in addition to Brazil and Canada. Although CC133 is most related to small ruminants [31], it was also isolated from cows with mastitis [44, 81]. CC126 was identified in Brazil [21], the USA [8], and Italy [45] and frequently recovered from subclinical mastitis.

Several CCs such as CC97, CC705, CC398, CC479, and CC8 were shared between proximal countries Germany, Italy, and Switzerland, consistent with cross-border movement of animals. Of note, CC479 strains were associated with severe bovine mastitis cases [80, 82] and demonstrated to induce a stronger pro-inflammatory response from bovine mammary epithelial cells (bMEC) than other bovine associated lineages such as CC151 [83]. Unlike CC479 and CC705 that show high prevalence in only Northern Italy, Germany, and Switzerland [32, 33, 46, 47, 61, 62], methicillin-resistant isolates of CC398 have been reported in eight countries across three continents: the USA, China, Belgium, Egypt, Germany, Iran, Italy, and Switzerland [34, 43, 47, 48, 84]. The ST398 MRSA lineage emerged in pigs but is now widely identified in other animal hosts including ruminants, poultry, horses, dogs, and cats [85]. It is reported that CC398 is a more promiscuous clone compared to other CCs [86], and therefore may represent a greater threat to public and animal health, especially considering the prevalence of MRSA strains.

CC8 is a global human lineage associated with an array of different diseases and includes the major epidemic clone USA300 [87]. Recently, CC8 S. aureus strains have been found in association with bovines [88] and reported in eleven countries: Algeria, Australia, Belgium, Canada, China, Germany, Italy, Japan, Switzerland, and the USA. It was suggested that the bovine sequence type (ST) 8 strains were the likely result of a recent human to bovine host jump [63, 89]. These data are important to evaluate the transmission risk for people working in the farm and milking environments and for consumers of raw dairy products [35]. Studies conducted in Uganda and Hungary identified the same lineage in both milkers and cows, suggesting transmission events between species, although the direction of transfer (cow-human or human-cow) was not defined [39, 73].

Taken together, this literature review identified a wide variety of S. aureus genotypes in dairy cattle worldwide. Considering the region of interest, it might help to formulate strategies to understand and reduce the infection spread of S. aureus strains. The most disseminated genotypes (CCs) globally were CC97 (15 countries), CC1 (10 countries), CC5 (10 countries), CC8 (8 countries), and CC398 (8 countries). Some lineages may be the result of human to bovine host switching events.

Population genomics of bovine S. aureus host-adaptation

The wide availability of inexpensive high-throughput sequencing has resulted in an explosion of whole genomes sequences for S. aureus and several population genomic studies of bovine strains have been published since the first genome of a bovine S. aureus strain RF122 was reported [90]. Overall, as indicated from population studies summarized in the previous section, a limited number of widely distributed S. aureus clones are responsible for the great majority of S. aureus mastitis cases worldwide [5, 86]. These clones have evolved through human to bovine host-switching events that have happened since the Neolithic era after domestication led to increased opportunities for transmission among humans and livestock [86, 91]. After a host-jump into cows from humans, S. aureus has undergone host-adaptation via gene acquisition, loss and diversification to allow it to survive in an anatomically and physiologically distinct niche [86, 90, 92]. For example, mobile genetic elements (MGE) including staphylococcal pathogenicity islands and bacteriophages which encode bovine-specific effectors of virulence are widely disseminated among bovine S. aureus clones and encode known and putative virulence factors including superantigens, von Willebrand binding protein (vWBP), and LukMF’ [92,93,94]. These effectors represent some of the best characterized factors involved in the pathogenesis of S. aureus mastitis with particular importance in both innate and acquired immune evasion [95, 96].

In addition to gene acquisition, loss of gene function has been a hallmark of bovine and ovine S. aureus strains that are adapting to their ruminant host species, as genes which are unessential for survival or fitness in the new host acquire nonsense or frameshift mutations that corrupt the open reading frame resulting in truncated or untranslated proteins [90, 92]. In addition, diversification of existing genes can occur that is associated with adaptation to a distinct host environment. For example, in bovine S. aureus strains, genes associated with carbohydrate utilization were identified to be under diversifying selective pressure suggesting adaptive evolution. Consistent with this genetic signature, bovine strains were better able to utilize lactose, the major source of carbohydrate in bovine milk, than S. aureus strains from humans or birds [86]. In another study, extensive recombination events were associated with the evolution of a subtype of CC97 (ST71), which led to the acquisition of several genes encoding proteins that promoted human innate immune evasion [97].

A recent study from Richardson et al. estimated the number of host-switching events that have occurred during the evolutionary history of S. aureus and although most jumps have occurred from humans into cows [89], several instances of reverse host-switches from cows back into humans have been identified [86]. For example, a study of the CC97 clone has identified at least two host-switch events from cows into humans that led to the emergence of a new human pathogenic clone that has spread around the world [6]. In addition, the major human epidemic clone in Southeast Asia (ST59) is predicted to have originated in cows [91, 98]. In each case, the strains have adapted to a human host species by the acquisition of genes required for human adaptation such as the immune evasion cluster of the Sa3int phage family which encodes factors that mediate human-specific innate immune evasion [6]. Richardson et al. identified a set of putative MGE that was associated with host-switching events suggesting an important role in host-adaptation [86] and providing avenues for future research into the host-pathogen interactions important for the colonization of dairy cows and the pathogenesis of mastitis.

Pathogenesis of bovine S. aureus

Adhesion to bovine mammary cells

Colonization is a key step in bacterial pathogenesis and bovine S. aureus has evolved in many ways to facilitate adhesion to different host cell types [98, 99]. One family of S. aureus adhesins comprises the microbial surface component recognizing adhesive matrix molecules (MSCRAMMs), which are cell-wall anchored proteins that share structural features like an N-terminal folded domain responsible for ligand binding, and a wall-spanning region followed by a sorting signal located at the C-terminal that anchor the protein to the cell wall [100].

Adhesion to and invasion of bovine mammary epithelial cells is mainly promoted by the fibronectin-binding protein MSCRAMMs FnBPA and FnBPB, [101]. Fibronectin acts as a bridge that connects FnBPA to the α5β1 integrin present on the cell surface [102, 103]. It has been demonstrated that the FnBP-integrin interaction induces the assembly of a cytosolic protein complex that modulates cytoskeleton rearrangement and mediates bacterial uptake [104]. S aureus may also adhere to fibrinogen, elastin, and plasminogen in an FnBP-dependent manner [105,106,107] that could promote colonization of different tissues and spread to other anatomical sites. Laboratory strains lacking FnBPs have a lower ability to colonize mouse mammary glands under suckling pressure, confirming that FnBPs confer a competitive advantage in vivo and may be considered as virulence factors for mammary gland colonization [108]. Overexpression of the fnb genes results in a higher invasion of bovine mammary epithelial cells (bMEC) [109]. Expression of FnBPB but not FnBPA is also related to increased invasiveness of isolates representative of ST8 [110]. However, the presence of fnb genes is not a prerequisite for cell adhesion because bacteria still retain the ability to invade even if both genes are absent [99], or if a truncated protein is produced [110] suggesting that additional factors may also be involved.

The high prevalence reported for the fnbA gene in bovine isolates [111,112,113] may reflect the importance of this adhesin in the pathogenesis of bovine mastitis. In contrast, a considerable discrepancy (1.5–100%) in the prevalence of the fnbB gene was reported in bovine S. aureus isolates, even among isolates from the same region that may in part reflect allelic variation that precluded PCR amplification [25, 114, 115].

Bovine isolates of S. aureus produce other MSCRAMMs that may promote colonization [109, 116, 117]. For example, clumping factors A and B (ClfA and ClfB) are fibrinogen-binding proteins that act as adhesins and have several defined roles in colonization and pathogenesis [100, 118, 119]. Although adherence to human endothelial cells requires fibrinogen to mediate the interaction between ClfA and the host integrin α5β3 [120], adherence to bovine epithelial cells occurs in a fibrinogen-independent manner via the annexin A2 receptor [121]. ClfA expressed by S. aureus also inhibits phagocytosis through the cleavage of C3b in a process mediated by an interaction with the complement regulator factor I [122]. It is yet to be confirmed if a similar inhibition mechanism is utilized by bovine S. aureus isolates. The clfA gene is usually described as highly prevalent (63.7-100%) in bacterial isolates of dairy cattle across all investigated countries with some disparities seen in the Netherlands (21%) [99, 123, 124]. The frequency of the clfB gene is reported as between 50 and 100% in bovine S. aureus isolates [25, 99, 111].

The collagen adhesin (Cna) protein is a MSCRAMM that not only has a role in adhesion but also participates in immune evasion during human infection [125]. Again, there is considerable variation in the prevalence of the cna gene observed among S. aureus isolates in dairy herds around the world [26, 111, 114]. Considering that collagen is highly prevalent in udder tissue, the expression of the cna gene by S. aureus of bovine origin could be important for adherence. Of note, it has been demonstrated that the cna gene has been acquired by the ST71 lineage of CC97 through recombination, conferring the capacity for adherence to collagen in vitro [97].

Other less studied fibrinogen-binding proteins have been described in S. aureus of bovine origin like the serine-aspartate repeat proteins (Sdr), extracellular fibrinogen binding protein (Efb), and the iron-regulated adhesin IsdA. Although there is no clear role for sdrCDE in mastitis the sdrD gene correlates with a high prevalence of IMI [126]. In addition, a strong association between the srdD and sdrE genes and clinical mastitis has been reported [127]. The efb gene has also been reported to have a high prevalence in field isolates [123]. During human infection, the interaction of Efb with fibrinogen creates a protective shield with an anti-phagocytic role indicating an immunosuppressive role [53]. Although IsdA binds to fibrinogen and fibronectin [128], it has been well-studied as a vaccine target for mastitis control along with other iron-regulated proteins such as IsdB and IsdH due to their high immunogenicity, gene expression during IMI and presence of antibodies in milk of animals naturally infected with S. aureus [7, 129, 130].

Bovine isolates of S. aureus produce a carbohydrate-based surface component when cultured in milk whey resulting in enhancement of bacterial adherence to bovine mammary cells and increased virulence in a murine model of mastitis [131, 132]. Although the carbohydrate-based surface component was never confirmed as a true capsule, other authors described encapsulated isolates as less adherent compared to acapsular strains [133, 134]. Internalization by MAC-T cells was also lower in encapsulated isolates while acapsular mutants persisted longer in host cells compared to the wild-type strains [135]. A one herd study reported that acapsular isolates belonging to ST9 exhibit high invasive capacity, a phenotype that was suggested to contribute to the dissemination of bacteria among lactating cows [136].

In summary, the expression of a large array of surface proteins with the ability to bind to extracellular matrix proteins by bovine S. aureus highlights the different strategies that have evolved to promote colonization and pathogenesis. Studies have reported the high prevalence of particular adhesins among bovine isolates from intercontinental herds, but further functional analysis is required, along with improved understanding of the synergy between the adhesins, and the regulation of gene expression, to provide important insights into the progression of intramammary infection.

Biofilm formation and bovine mastitis

S. aureus of bovine origin is usually described as a biofilm producer [109, 137, 138], a trait that is usually related to bacterial persistence and increased tolerance to antibiotics [139]. The biofilm has a complex structure consisting of many cell layers embedded in an extracellular matrix, which in S. aureus consists mainly of the polysaccharide intercellular adhesin (PIA), a poly-β (1–6)-N-acetylglucosamine (PNAG) produced by the ica locus. Besides PIA, the biofilm matrix is also composed of several MSCRAMMs, such as FnBPs, ClfA, ClfB, and Protein A, that promote bacterial adhesion to host cells to initiate biofilm formation (56). The production of S. aureus biofilm has not yet been demonstrated in vivo in IMI, despite immunological detection of certain components such as slime and PIA from mammary gland samples [134, 140].

There are many differences in the production of biofilm among bovine isolates of different genetic origins. Generally, strong biofilm producers belong to the agrIII group [113, 141] and invade epithelial cells to a lesser degree compared to planktonic or low biofilm producers [142, 143]. agrI-type isolates are moderate biofilm producers, highly invasive, and are frequently isolated from subclinical mastitis in contrast to the agrII group that produces less biofilm, has reduced invasiveness, and is more related to clinical mastitis [144,145,146,147]. Nevertheless, differences in the level of biofilm formation among isolates from the same lineage are also observed [23, 24].

Milk and lactose can positively influence biofilm formation by S. aureus agrII bovine isolates due to the production of PIA [141, 144, 148]. In addition, milk and lactose stimulate the expression of the lactose transporter EII and the teichoic acid biosynthesis protein B (TagB) that is regulated by Rbf, the repressor of biofilm [148,149,150]. Teichoic acids are an important component of the staphylococcal biofilm extracellular matrix and establish electrostatic interactions with PIA [151].

S. aureus can cause mastitis in cows in different stages of lactation and infections frequently persist over the ongoing lactation [152, 153]. A non-lactating period, also called the dry period, allows the recovery of the cow’s mammary gland and improves milk production in the following lactation. Additionally, dry cow therapy is recommended to reduce the risk of staphylococcal infections. High biofilm producers are likely to persist during the cow’s dry period [23] due to their efficient adherence to the mammary epithelium. Also, S. aureus isolated from milk is more likely to produce biofilm compared to bacteria isolated from teat skin and milking unit liners [154]. In a three-year follow-up of a dairy herd, S. aureus strains that produced more biofilm and higher amounts of PNAG showed higher within-herd prevalence and persistence [136]. These strains also presented reduced cellular cytotoxicity and high invasiveness. A less prevalent phenotype whose persistence was related to the formation of dendrites instead of biofilm was also described by the authors. These findings show that although biofilm production is usually described as a primordial factor that contributes to persistence other bacterial traits may also support chronic infections during bovine mastitis.

The impact of ica deletion on biofilm production can be compensated by the expression of a surface protein known as the biofilm-associated protein (Bap) that promotes adhesion of S. aureus to abiotic and biotic surfaces [155]. Isolates expressing Bap adhere to epithelial cells but are less invasive due to the interaction between Bap and the Gp96 receptor expressed by mammary epithelial cells, which interferes with the internalization pathway mediated by FnBP [99, 156]. Furthermore, isolates harboring ica and bap are strong biofilm producers and are more resistant to antibiotics than isolates harboring only ica or bap [155]. However, Bap-positive strains can lose the ability to form biofilm when grown in milk, probably due to the stabilization of Bap in the presence of calcium [49]. At lower concentrations of calcium, Bap is cleaved into fragments that form amyloid fibers providing a scaffold for biofilm development [157]. Taken together, these data indicate that S. aureus of bovine origin forms biofilm through a PIA-dependent manner during the lactating period but a PIA-independent mechanism via Bap might play a role during the dry cow period when concentrations of calcium in the udder are low.

Toxins as important virulence factors in mastitis

A wide array of different secreted toxins has been implicated in S. aureus disease pathogenesis [158]. Superantigens (SAgs) are a family of potent immunostimulatory exotoxins produced by S. aureus that are known for their ability to circumvent normal immune function. They bind as a bridge to major histocompatibility complex class II molecules and specific Vβ segments of T-cell receptors, resulting in their proliferation, differentiation into effector cells, and massive cytokine release [159]. As a result, several T cells that share the same Vβ segment may be activated, independently of antigen specificity [160]. SAgs in bovine strains of S. aureus include TSST-1, staphylococcal enterotoxins (SE), and staphylococcal enterotoxin-like proteins (SEl) that are mostly encoded on MGE [88, 93].

Genomic analysis revealed variation in the gene content of SAgs of bovine isolates from 57 distinct sequence types (ST) [161] in concordance with the findings reported for isolates around the world [90, 162, 163]. However, there are disparities in the distribution of the enterotoxin gene cluster (egc), which is highly prevalent within CC30, CC151, and CC45, and the pathogenicity island SaPIbov, which is primarily associated with CC133 and CC151 [161]. The prevalence of enterotoxins is also highly variable in isolates belonging to different geographical locations [21, 124, 164], which is explained by the wide variety of S. aureus genotypes found in dairy cattle [124], besides differences in environmental and management factors in each geographical area [165].

The presence of specific enterotoxin genes has been linked to acute and clinical bovine mastitis [23, 166] although seg and sei were found in persistent subtypes recovered from cows presenting subclinical mastitis [136]. Additionally, seh and sek tend to be more frequent in isolates causing subclinical mastitis, while sed and sej are mostly associated with persistent mastitis [166]. Other studies reported that neither the genes sea and sed [167] nor the set of genes sea, seb, see [168] were detected in isolates causing subclinical mastitis in cows on dairy farms in Spain [167] or the USA [168]. Due to the large repertoire of enterotoxins present in S. aureus more studies are needed before gene signatures can be associated with the outcome of mastitis in dairy cows.

Current evidence indicates the involvement of SAgs in the pathogenesis of bovine mastitis. Enterotoxin M and H contribute to inflammation, necrosis, and/or apoptosis of bovine mammary epithelial cells [169, 170], pathology also observed upon injection of SEC into the mammary glands of mice [171]. However, the high concentrations of toxins used in those studies may not mimic the in vivo condition and therefore are unlikely to occur during intramammary infection. It was demonstrated that most SAgs produced by S. aureus RF122 present mitogenic activity for bovine T cells, even at low concentrations [161]. In this study cows challenged with a SAg-deficient strain demonstrated an inability to develop clinical mastitis but had similar somatic cell counts and milk quality compared to wild-type RF122-infected cows, highlighting the functional importance of SAgs during bovine infection.

S. aureus also produces numerous membrane-damaging toxins that compromise host-cell function in vitro and are involved in iron acquisition and host immune evasion [172]. α-hemolysin (Hla) binds to the ADAM10 receptor and assembles into a β-barrel transmembrane pore in the cell surface, promoting the release of small molecules that lead to tissue necrosis [173]. Phagocytized bacteria secrete Hla to lyse bovine endothelial cells, an event that may free intracellular bacteria to target other cells [174]. β-hemolysin (Hlb) also damages the secretory epithelial cells but has less cytotoxicity, with a synergistic effect seen with a combination of both toxins [175]. The hydrolysis of sphingomyelin by β-hemolysin increases host cell permeability with progressive loss of cell surface charge, rendering the cells more susceptible to the action of α-hemolysin. Additionally, β-hemolysin exerts lymphotoxic effects and lyses bovine erythrocytes contributing to iron acquisition [172]. δ-hemolysin (Hld) is a small peptide that is translocated across the plasma membrane at low concentrations, forming a transient structure that perturbs the bilayer and induces ionic efflux [176]. Hld belongs to the family of phenol-soluble modulins (PSM), toxins that are highly cytolytic and have proinflammatory activities. A high expression of hld but no other psm genes was reported in the well-studied bovine strains RF122 and N305 [177]. In the same study, the authors showed reduced cytokine production in bacteria expressing psmαβhld, suggesting that an attenuated immune response is related to chronic mastitis.

The hemolytic activities of Hlb and Hld produced by S. aureus isolates recovered from clinical mastitis when grown in the whey of skimmed milk suggest a role for Hld in IMI [178] but there is experimental evidence that supports the relevance of the other toxins in bovine mastitis. Antibodies raised against α- but not β-hemolysin produced by bovine S. aureus strains protect rabbits from mastitis [179]. Intramammary injections of Hla in lactating rabbits disrupt the architecture of the mammary gland, with necrotic lesions seen at high doses while injection of purified Hlb causes an influx of polymorphonuclear leukocytes (PMN) into the alveoli besides edema [180]. These findings were further corroborated by studies showing less necrosis and greater preservation of tissue structure 24 h post-challenge in a murine model with a Δhla/Δhlb mutant [181]. Compared to the parental strain that caused 60% mortality, no mortality was seen in mice inoculated with the double mutant. Cows infected with Δhla and Δhlb mutants were able to eliminate the bacteria and suffer only mild inflammation compared to the severe clinical signs presented by animals infected with the parental strains [182]. Taken together, these studies reveal the importance of Hla and Hlb in bacterial virulence during IMI.

Genes involved in the synthesis of hemolysins are frequently found in bovine isolates of S. aureus [166, 183, 184] and nearly 50% of those isolated from subclinical disease show hemolytic activity [163, 185]. There is no correlation between hemolysin expression and bacterial lineage since strains from the same ST often demonstrate both high and low levels of gene expression and activity [186, 187]. S. aureus isolated from cows with subclinical mastitis that shows higher expression of hld are more likely to be nonpersistent during either lactation or through the dry period [23]. Also, isolates causing clinical mastitis show higher hemolytic activity and prevalence of hemolysin genes when compared to isolates recovered from subclinical mastitis [163, 188], which may result in greater damage of the bovine mammary secretory epithelial cells. In addition, the presence of single nucleotide polymorphisms within the promoter region of hla is likely to be associated with a high α-hemolysin expression by S. aureus isolates associated with severe bovine mastitis, a finding that may be used as a marker to discriminate strain virulence [189].

Leukocidins secreted by S. aureus are bicomponent toxins that target white blood cells leading to the formation of pores that disrupt host cell function allowing S. aureus to escape from the immune system [190]. The leukocidin S subunit binds to a receptor in the host cell membrane, followed by F subunit recruitment and oligomerization into an octameric structure creating a β-barrel channel inserted in the host cell lipid bilayer. The presence of interchangeable classes of S and F subunits in bovine S. aureus raises the hypothesis that leukocidins may present distinct activities according to the cell target during the infective process [191]. LukMF’ is restricted to S. aureus strains associated with animals such as bovines and ovines [192] although the prevalence of the lukM and lukF genes varies considerably (4–96%) among S. aureus isolates recovered from cows [82, 193, 194]. Among all bovine-associated S. aureus lineages, CC97 showed a high prevalence of the genes lukMF [11, 195]. Also, there are reports that associate gene presence with certain clones such as CC151, CC479, and CC133 [36]. Even though the genes have been detected in S. aureus isolated from subclinical mastitis [24, 82, 167], expression is more often detected in those recovered from clinical mastitis or lineages that carry a nonsense mutation in the repressor of toxins Rot [82]. The presence of LukMF´ and antibodies in milk and the serum of goats that received intramammary infusion with a high leukocidin-producing strain is evidence of the functional role of these toxins during mastitis [196].

The high cytotoxicity of LukMF’ may be conferred by the high efficiency of LukM for cell receptors present on bovine leukocytes, neutrophils, and macrophages [197]. Interestingly, no inflammation was seen in cows that received intramammary injections of purified LukMF’ [198]. Later, it was shown that the expression of CCR1 on bovine, but not on human neutrophils was the cause for species-specific killing by LukMF’ [199]. Further studies challenged cattle with high and intermediate LukMF’ producing strains and correlated the higher level of expression and the presence of milk antibodies against LukM with clinical mastitis severity [96]. Taken together, these studies strongly support the functional significance of LukMF’ in the pathogenesis of bovine mastitis.

Other leukotoxins such as γ-hemolysin (Hlg) and LukED are produced by bovine S. aureus but vary in their activities against bovine cells. These toxins recognize certain receptors in neutrophils but compared to LukMF’ they are expressed in lower levels during in vitro growth [96]. Hlg is composed of the F subunit HlgB that combines with HlgC or HlgA to form the active toxin [200]. The effect of HlgAB on bovine PMN is greater than HlgBC [191]. The same authors showed that reduced activity of the leukotoxin LukED was boosted when LukD was replaced by LukM. The genes hlg and lukED are frequently reported in bovine S. aureus [184, 195, 201] but more studies are needed to provide data on their role during IMI.

In summary, bovine strains of S. aureus produce a plethora of toxins that target different host cells and have wide ranging roles during infection including nutrient acquisition and immune evasion. If in the past studies were focused on detecting their presence, now they have started to reveal the molecular functions of toxins and their interplay with the host. The immunogenicity of some toxins has been shown supporting their use as therapeutic agents for fighting IMI. Though higher levels of toxin expression are more likely to be associated with more severe symptoms of mastitis it is still unknown if their complementary and/or redundant activities are needed during IMI progression.

Conclusions and future directions

S. aureus has been the subject of intense investigations in the veterinary field and is probably the most studied causative agent of bovine mastitis, a disease with a huge impact on the dairy supply chain. Here we compiled findings specifically related to important virulence factors of bovine S. aureus. Considering the 27 countries from which the studies have been published, CC97 and t267 are the most disseminated lineages. There is no clear pattern of distribution of adhesion or toxin genes in bovine S. aureus isolates making the outcome of association studies of mastitis and novel therapeutic development complicated. Although the production of biofilm has been reported for every field isolate, more evidence is needed to confirm in vivo biofilm formation and the specific role of Bap in bovine S. aureus antibiotic-resistant biofilm production. Except for leukocidin LukMF’, few virulence determinants have been correlated with clinical severity.

There is still much must be done to improve animal health, diagnosis, and control of mastitis in dairy herds. 3D cell culture is emerging as a powerful technology to replicate mammary gland structural complexity, allowing advances in the pathophysiology of mastitis and reducing the dependence on animal models. Organoid cultures open new windows to explore the molecular crosstalk between host and pathogen, host and lineage-specific traits that affect disease outcome, and the contribution of the host microenvironment to the expression of the virulence factors highly prevalent in field isolates. This may shed light on the S. aureus biology of bovine mastitis and eventually open new perspectives aimed at better controlling bovine mastitis.

While numerous genomes have been sequenced, bovine S. aureus contains many genes of unknown function. The construction of transposon (Tn) mutant libraries will deepen our understanding of the pathogen´s biology, unveil new virulence factors and mechanisms of bacterial resistance to antibiotics allowing more efficient strategies for bacterial control. The co-existence of different clonal complexes within dairy herds is a challenge to the control of mastitis. Tn mutagenesis may also be used to identify lineage-specific traits that contribute to the success of clinically relevant clones.

Antibiotics are frequently used to prevent and treat bovine mastitis. With the emergence of antimicrobial-resistant clones and consumer demand for food safety, farmers must rely on strategies other than herd sanitation to control intramammary infections. Although vaccination is available, multifactorial causes like poor antigens and gaps in the understanding of the immune response hinder the development of effective vaccines against S. aureus. Over the last years, there has been an increase in the search for alternative treatment methods using natural products, bacteriocins, bacteriophages, and nanoparticles. Despite showing promise, the inhibitory activity seen in vitro is not always sustained when the therapy is tested in vivo regardless of the animal model used, an interference probably caused by milk components. This can be circumvented using nanocarriers that bring stability, solubility, tissue permeability, or controlled release of the antibiotic drug which can be used in formulations to eradicate planktonic or sessile bacteria. However, there is still a long road to successful clinical translation.

Availability of data and materials

Not applicable.

Abbreviations

Bap:

Biofilm-associated protein

bMEC:

Bovine mammary epithelial cells

CC:

Clonal complex

Clf:

Clumping factor

FnBP:

Fibronectin-binding protein

IMI:

Intramammary infection

LukMF’:

Bicomponent leukocidin LukMF’

MAC-T:

Mammary alveolar cells-large T antigen

MSCRAMMs:

Microbial surface component recognizing adhesive matrix molecules

MLST:

Multilocus sequence type

MGE:

Mobile genetic elements

PIA:

Polysaccharide intercellular adhesin

PNAG:

Poly-β (1–6)-N-acetylglucosamine

Rbf:

Repressor of biofilm

SAgs:

Superantigens

TagB:

Teichoic acid biosynthesis protein B (TagB)

vWBP:

von Willebrand binding protein

References

  1. Gonçalves JL, Kamphuis C, Martins C, Barreiro JR, Tomazi T, Gameiro AH, et al. Bovine subclinical mastitis reduces milk yield and economic return. Livest Sci. 2018;210:25–32. https://doi.org/10.1016/j.livsci.2018.01.016.

    Article  Google Scholar 

  2. Gomes F, Henriques M. Control of bovine mastitis: old and recent therapeutic Approaches. Curr Microbiol. 2016;72:377–82. https://doi.org/10.1007/s00284-015-0958-8.

    Article  CAS  PubMed  Google Scholar 

  3. Fagundes H, Barchesi L, Filho AN, Ferreira LM, Oliveira CA. Occurrence of Staphylococcus aureus in raw milk produced in dairy farms in São Paulo state, Brazil. Braz J Microbiol. 2010;41:376–80. https://doi.org/10.1590/S1517-83822010000200018.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Guardabassi L, Butaye P, Dockrell DH, Fitzgerald JR, Kuijper EJ, ESCMID Study Group for Veterinary Microbiology (ESGVM). One Health: a multifaceted concept combining diverse approaches to prevent and control antimicrobial resistance. Clin Microbiol Infect. 2020;26:1604–5. https://doi.org/10.1016/j.cmi.2020.07.012.

    Article  CAS  PubMed  Google Scholar 

  5. Fitzgerald JR. Livestock-associated Staphylococcus aureus: origin, evolution and public health threat. Trends Microbiol. 2012;20:192–8. https://doi.org/10.1016/j.tim.2012.01.006.

    Article  CAS  PubMed  Google Scholar 

  6. Spoor LE, McAdam PR, Weinert LA, Rambaut A, Hasman H, Aarestrup FM, et al. Livestock origin for a human pandemic clone of community-associated methicillin-resistant Staphylococcus aureus. mBio. 2013;4. https://doi.org/10.1128/mbio.00356-13.

  7. Rainard P, Foucras G, Fitzgerald JR, Watts JL, Koop G, Middleton JR. Knowledge gaps and research priorities in Staphylococcus aureus mastitis control. Transbound Emerg Dis. 2018;65(Suppl 1):149–65. https://doi.org/10.1111/tbed.12698.

    Article  PubMed  Google Scholar 

  8. Smith EM, Green LE, Medley GF, Bird HE, Fox LK, Schukken YH, et al. Multilocus sequence typing of intercontinental bovine Staphylococcus aureus isolates. J Clin Microbiol. 2005;43:4737–43. https://doi.org/10.1128/jcm.43.9.4737-4743.2005.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Aires-de-Sousa M, Parente CE, Vieira-da-Motta O, Bonna IC, Silva DA, de Lencastre H. Characterization of Staphylococcus aureus isolates from buffalo, bovine, ovine, and caprine milk samples collected in Rio de Janeiro State, Brazil. Appl Environ Microbiol. 2007;73:3845–9. https://doi.org/10.1128/aem.00019-07.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Rabello RF, Moreira BM, Lopes RMM, Teixeira LM, Riley LW, Castro ACD. Multilocus sequence typing of Staphylococcus aureus isolates recovered from cows with mastitis in Brazilian dairy herds. J Med Microbiol. 2007;56:1505–11. https://doi.org/10.1099/jmm.0.47357-0.

    Article  CAS  PubMed  Google Scholar 

  11. Hata E, Kobayashi H, Nakajima H, Shimizu Y, Eguchi M. Epidemiological analysis of Staphylococcus aureus isolated from cows and the environment of a dairy farm in Japan. J Vet Med Sci. 2010;72:647–52. https://doi.org/10.1292/jvms.09-0452.

    Article  PubMed  Google Scholar 

  12. Hata E. Bovine mastitis outbreak in Japan caused by methicillin-resistant Staphylococcus aureus New York/Japan clone. J Vet Diagn Invest. 2016;28:291–8. https://doi.org/10.1177/1040638716643126.

    Article  CAS  PubMed  Google Scholar 

  13. Naushad S, Nobrega DB, Naqvi SA, Barkema HW, De Buck J. Genomic analysis of bovine Staphylococcus aureus isolates from milk to elucidate diversity and determine the distributions of antimicrobial and virulence genes and their association with mastitis. mSystems. 2020;5:e00063-20. https://doi.org/10.1128/msystems.00063-20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Guinane CM, Sturdevant DE, Herron-Olson L, Otto M, Smyth DS, Villaruz AE, et al. Pathogenomic analysis of the common bovine Staphylococcus aureus clone (ET3): emergence of a virulent subtype with potential risk to public health. J Infect Dis. 2008;197:205–13. https://doi.org/10.1086/524689.

    Article  CAS  PubMed  Google Scholar 

  15. Murphy MP, Niedziela DA, Leonard FC, Keane OM. The in vitro host cell immune response to bovine-adapted Staphylococcus aureus varies according to bacterial lineage. Sci Rep. 2019;9:6134. https://doi.org/10.1038/s41598-019-42424-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Titouche Y, Hakem A, Houali K, Meheut T, Vingadassalon N, Ruiz-Ripa L, et al. Emergence of methicillin-resistant Staphylococcus aureus (MRSA) ST8 in raw milk and traditional dairy products in the Tizi Ouzou area of Algeria. J Dairy Sci. 2019;102:6876–84. https://doi.org/10.3168/jds.2018-16208.

  17. Zaatout N, Ayachi A, Kecha M, Kadlec K. Identification of staphylococci causing mastitis in dairy cattle from Algeria and characterization of Staphylococcus aureus. J Appl Microbiol. 2019;127:1305–14. https://doi.org/10.1111/jam.14402.

    Article  CAS  PubMed  Google Scholar 

  18. Schabauer A, Pinior B, Gruber CM, Firth CL, Käsbohrer A, Wagner M, et al. The relationship between clinical signs and microbiological species, spa type, and antimicrobial resistance in bovine mastitis cases in Austria. Vet Microbiol. 2018;227:52–60. https://doi.org/10.1016/j.vetmic.2018.10.024.

    Article  PubMed  Google Scholar 

  19. Bardiau M, Yamazaki K, Duprez JN, Taminiau B, Mainil JG, Ote I. Genotypic and phenotypic characterization of methicillin-resistant Staphylococcus aureus (MRSA) isolated from milk of bovine mastitis. Lett Appl Microbiol. 2013;57:181–6. https://doi.org/10.1111/lam.12099.

    Article  CAS  PubMed  Google Scholar 

  20. Nemeghaire S, Argudín MA, Haesebrouck F, Butaye P. Epidemiology and molecular characterization of methicillin-resistant Staphylococcus aureus nasal carriage isolates from bovines. BMC Vet Res. 2014;10:153. https://doi.org/10.1186/1746-6148-10-153.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Bonsaglia ECR, Silva NCC, Rossi BF, Camargo CH, Dantas STA, Langoni H, et al. Molecular epidemiology of methicillin-susceptible Staphylococcus aureus (MSSA) isolated from milk of cows with subclinical mastitis. Microb Pathog. 2018;124:130–5. https://doi.org/10.1016/j.micpath.2018.08.031.

    Article  CAS  PubMed  Google Scholar 

  22. Rossi BF, Bonsaglia ECR, Castilho IG, Dantas STA, Salina A, Langoni H, et al. Genotyping of long-term persistent Staphylococcus aureus in bovine subclinical mastitis. Microb Pathog. 2019;132:45–50. https://doi.org/10.1016/j.micpath.2019.04.031.

    Article  CAS  PubMed  Google Scholar 

  23. Veh KA, Klein RC, Ster C, Keefe G, Lacasse P, Scholl D, Roy JP, Haine D, Dufour S, Talbot BG, Ribon AO, Malouin F. Genotypic and phenotypic characterization of Staphylococcus aureus causing persistent and nonpersistent subclinical bovine intramammary infections during lactation or the dry period. J Dairy Sci. 2015;98:155–68. https://doi.org/10.3168/jds.2014-8044.

    Article  CAS  PubMed  Google Scholar 

  24. Pichette-Jolette S, Millette G, Demontier E, Bran-Barrera D, Cyrenne M, Ster C, et al. Partial prediction of the duration and the clinical status of Staphylococcus aureus bovine intramammary infections based on the phenotypic and genotypic analysis of isolates. Vet Microbiol. 2019;228:188–95. https://doi.org/10.1016/j.vetmic.2018.11.024.

    Article  CAS  PubMed  Google Scholar 

  25. Zhang L, Gao J, Barkema HW, Ali T, Liu G, Deng Y, et al. Virulence gene profiles: alpha-hemolysin and clonal diversity in Staphylococcus aureus isolates from bovine clinical mastitis in China. BMC Vet Res. 2018;14:63. https://doi.org/10.1186/s12917-018-1374-7.

  26. Ren Q, Liao G, Wu Z, Lv J, Chen W. Prevalence and characterization of Staphylococcus aureus isolates from subclinical bovine mastitis in southern Xinjiang, China. J Dairy Sci. 2020;103:3368–80. https://doi.org/10.3168/jds.2019-17420.

    Article  CAS  PubMed  Google Scholar 

  27. Wang W, Lin X, Jiang T, Peng Z, Xu J, Yi L, et al. prevalence and characterization of Staphylococcus aureus cultured from raw milk taken from dairy cows with mastitis in Beijing, China. Front Microbiol. 2018;9:1123. https://doi.org/10.3389/fmicb.2018.01123.

    Article  PubMed  PubMed Central  Google Scholar 

  28. El-Ashker M, Gwida M, Tomaso H, Monecke S, Ehricht R, El-Gohary F, et al. Staphylococci in cattle and buffaloes with mastitis in Dakahlia Governorate, Egypt. J Dairy Sci. 2015;98:7450–9. https://doi.org/10.3168/jds.2015-9432.

    Article  CAS  PubMed  Google Scholar 

  29. Zayda MG, Matsuda Y, Hammad AM, Honjoh K, Elbagory AM, Miyamoto T. Molecular characterisation of methicillin-resistant (MRSA) and methicillin-susceptible (MSSA) Staphylococcus aureus isolated from bovine subclinical mastitis and Egyptian raw milk cheese. Int Dairy J. 2020;104:104646–55. https://doi.org/10.1016/j.idairyj.2020.104646.

  30. Gindonis V, Taponen S, Myllyniemi AL, Pyörälä S, Nykäsenoja S, Salmenlinna S, et al. Occurrence and characterization of methicillin-resistant staphylococci from bovine mastitis milk samples in Finland. Acta Vet Scand. 2013;55:61. https://doi.org/10.1186/1751-0147-55-61.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Bar-Gal GK, Blum SE, Hadas L, Ehricht R, Monecke S, Leitner G. Host-specificity of Staphylococcus aureus causing intramammary infections in dairy animals assessed by genotyping and virulence genes. Vet Microbiol. 2015;176:143–54. https://doi.org/10.1016/j.vetmic.2015.01.007.

    Article  CAS  PubMed  Google Scholar 

  32. Fessler A, Scott C, Kadlec K, Ehricht R, Monecke S, Schwarz S. Characterization of methicillin-resistant Staphylococcus aureus ST398 from cases of bovine mastitis. J Antimicrob Chemother. 2010;65:619–25. https://doi.org/10.1093/jac/dkq021.

    Article  CAS  PubMed  Google Scholar 

  33. Spohr M, Rau J, Friedrich A, Klittich G, Fetsch A, Guerra B, et al. Methicillin-resistant Staphylococcus aureus (MRSA) in three dairy herds in southwest Germany. Zoonoses Public Health. 2011;58:252–61. https://doi.org/10.1111/j.1863-2378.2010.01344.x.

    Article  CAS  PubMed  Google Scholar 

  34. Kadlec K, Entorf M, Peters T. Occurrence and characteristics of livestock-associated methicillin-resistant Staphylococcus aureus in quarter milk samples from dairy cows in Germany. Front Microbiol. 2019 Jun 12;10:1295. https://doi.org/10.3389/fmicb.2019.01295.

  35. Sheet OH, Grabowski NT, Klein G, Reich F, Abdulmawjood A. Characterisation of mecA gene negative Staphylococcus aureus isolated from bovine mastitis milk from northern Germany. Folia Microbiol. 2019;64:845–55. https://doi.org/10.1007/s12223-019-00698-z.

    Article  CAS  Google Scholar 

  36. Schlotter K, Ehricht R, Hotzel H, Monecke S, Pfeffer M, Donat K. Leukocidin genes lukF-P83 and lukM are associated with Staphylococcus aureus clonal complexes 151, 479 and 133 isolated from bovine udder infections in Thuringia, Germany. Vet Res. 2012;43:42. https://doi.org/10.1186/1297-9716-43-42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Monecke S, Kuhnert P, Hotzel H, Slickers P, Ehricht R. Microarray-based study on virulence-associated genes and resistance determinants of Staphylococcus aureus isolates from cattle. Vet Microbiol. 2007;125:128–40. https://doi.org/10.1016/j.vetmic.2007.05.016.

    Article  CAS  PubMed  Google Scholar 

  38. Schlotter K, Huber-Schlenstedt R, Gangl A, Hotzel H, Monecke S, Müller E, et al. Multiple cases of methicillin-resistant CC130 Staphylococcus aureus harboring mecC in milk and swab samples from a Bavarian dairy herd. J Dairy Sci. 2014;97:2782–8. https://doi.org/10.3168/jds.2013-7378.

    Article  CAS  PubMed  Google Scholar 

  39. Juhász-Kaszanyitzky E, Jánosi S, Somogyi P, Dán A, van der Graaf-van Bloois L, van Duijkeren E, et al. MRSA transmission between cows and humans. Emerg Infect Dis. 2007;13:630–2. https://dx.doi.org/10.3201%2Feid1304.060833.

  40. Mitra SD, Velu D, Bhuvana M, Krithiga N, Banerjee A, Shome R, et al. Staphylococcus aureus spa type t267, clonal ancestor of bovine subclinical mastitis in India. J Appl Microbiol. 2013;114:1604–15. https://doi.org/10.1111/jam.12186.

    Article  CAS  PubMed  Google Scholar 

  41. Sheela P, Isloor S, Rathnamma D, Veeregowda BM, Shambulingappa B, et al. Characterisation by spa typing of Staphylococcus aureus isolates originating from bovine and bubaline mastitis in southern India. Indian J Comp Microbiol Immunol Infect Dis. 2019;40:21–30. https://doi.org/10.5958/0974-0147.2019.00004.7.

    Article  Google Scholar 

  42. Mahanti A, Joardar SN, Bandyopadhyay S, Banerjee J, Ghosh S, Batabyal K, et al. Characterization of methicillin-resistant and enterotoxins producing Staphylococcus aureus in bovine milk in India. J Sci Food Agric; 2020. 100017. https://doi.org/10.1016/j.jafr.2019.100017.

  43. Saei HD, Panahi M. Genotyping and antimicrobial resistance of Staphylococcus aureus isolates from dairy ruminants: differences in the distribution of clonal types between cattle and small ruminants. Arch Microbiol. 2020;202:115–25. https://doi.org/10.1007/s00203-019-01722-z.

    Article  CAS  Google Scholar 

  44. Smyth DS, Feil EJ, Meaney WJ, Hartigan PJ, Tollersrud T, Fitzgerald JR, et al. Molecular genetic typing reveals further insights into the diversity of animal-associated Staphylococcus aureus. J Med Microbiol. 2009;58:1343–53. https://doi.org/10.1099/jmm.0.009837-0.

    Article  PubMed  Google Scholar 

  45. Luini M, Cremonesi P, Magro G, Bianchini V, Minozzi G, Castiglioni B, et al. Methicillin-resistant Staphylococcus aureus (MRSA) is associated with low within-herd prevalence of intra-mammary infections in dairy cows: Genotyping of isolates. Vet Microbiol. 2015;178:270–4. https://doi.org/10.1016/j.vetmic.2015.05.010.

    Article  CAS  PubMed  Google Scholar 

  46. Mazzilli M, Piccinini R, Scali F, Zecconi A. Pattern characterization of genes involved in non-specific immune response in Staphylococcus aureus isolates from intramammary infections. Res Vet Sci. 2015;103:54–9. https://doi.org/10.1016/j.rvsc.2015.09.007.

    Article  CAS  PubMed  Google Scholar 

  47. Barberio A, Mazzolini E, Dall’Ava B, Rosa G, Brunetta R, Zandonà L, et al. A longitudinal case study on dissemination of ST398 methicillin-resistant Staphylococcus aureus within a dairy cow herd. Foodborne Pathog Dis. 2019;16:761–8. https://doi.org/10.1089/fpd.2019.2622.

    Article  CAS  PubMed  Google Scholar 

  48. Cortimiglia C, Luini M, Bianchini V, Marzagalli L, Vezzoli F, Avisani D, et al. Prevalence of Staphylococcus aureus and of methicillin-resistant S. aureus clonal complexes in bulk tank milk from dairy cattle herds in Lombardy region (Northern Italy). Epidemiol Infect. 2016;144:3046–51. https://doi.org/10.1017/s0950268816001576.

    Article  CAS  PubMed  Google Scholar 

  49. Snel GG, Monecke S, Ehricht R, Piccinini R. Molecular characteristics of bap-positive Staphylococcus aureus strains from dairy cow mastitis. J Dairy Res. 2015;82:312–6. https://doi.org/10.1017/S0022029915000199.

    Article  CAS  PubMed  Google Scholar 

  50. Johler S, Macori G, Bellio A, Acutis PL, Gallina S, Decastelli L. Characterization of Staphylococcus aureus isolated along the raw milk cheese production process in artisan dairies in Italy. J Dairy Sci. 2018;101:2915–20. https://doi.org/10.3168/jds.2017-13815.

    Article  CAS  PubMed  Google Scholar 

  51. Baba K, Ishihara K, Ozawa M, Usui M, Hiki M, Tamura Y, et al. Prevalence and mechanism of antimicrobial resistance in Staphylococcus aureus isolates from diseased cattle, swine and chickens in Japan. J Vet Med Sci. 2012;74:561–5. https://doi.org/10.1292/jvms.11-0357.

    Article  CAS  PubMed  Google Scholar 

  52. Sato T, Usui M, Konishi N, Kai A, Matsui H, Hanaki H, et al. Closely related methicillin-resistant Staphylococcus aureus isolates from retail meat, cows with mastitis, and humans in Japan. PLoS ONE. 2017;12:e0187319. https://doi.org/10.1371/journal.pone.0187319.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Ko DS, Seong WJ, Kim D, Kim EK, Kim NH, Lee CY, Kim JH, Kwon HJ. Molecular prophage typing of Staphylococcus aureus isolates from bovine mastitis. J Vet Sci. 2018;19:771–81. https://doi.org/10.4142/jvs.2018.19.6.771.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Hwang SY, Park YK, Koo HC, Park YH. spa typing and enterotoxin gene profile of Staphylococcus aureus isolated from bovine raw milk in Korea. J Vet Sci.2010;11:125 – 31. https://doi.org/10.4142/jvs.2010.11.2.125.

  55. Song JW, Yang SJ, Shin S, Seo KS, Park YH, Park KT. Genotypic and phenotypic characterization of methicillin-resistant Staphylococcus aureus isolated from bovine mastitic milk in Korea. J Food Prot. 2016;79:1725–32. https://doi.org/10.4315/0362-028x.jfp-16-067.

    Article  CAS  PubMed  Google Scholar 

  56. Jørgensen HJ, Mørk T, Caugant DA, Kearns A, Rørvik LM. Genetic variation among Staphylococcus aureus strains from Norwegian bulk milk. Appl Environ Microbiol. 2005;71:8352–61. https://doi.org/10.1128/aem.71.12.8352-8361.2005.

    Article  PubMed  PubMed Central  Google Scholar 

  57. Zastempowska E, Orczykowska-Kotyna M, Lassa H. Isolation of nuc mutant isolates of Staphylococcus aureus from bovine clinical mastitis. Vet J. 2014;200:446–8. https://doi.org/10.1016/j.tvjl.2014.03.029.

    Article  CAS  PubMed  Google Scholar 

  58. Conceição T, de Lencastre H, Aires-de-Sousa M. Healthy bovines as reservoirs of major pathogenic lineages of Staphylococcus aureus in Portugal. Microb Drug Resist. 2017;23:845–51. https://doi.org/10.1089/mdr.2017.0074.

    Article  CAS  PubMed  Google Scholar 

  59. Antók FI, Mayrhofer R, Marbach H, Masengesho JC, Keinprecht H, Nyirimbuga V, et al. Characterization of antibiotic and biocide resistance genes and virulence factors of Staphylococcus species associated with bovine mastitis in Rwanda. Antibiotics. 2019;9:1. https://doi.org/10.3390/antibiotics9010001.

    Article  CAS  PubMed Central  Google Scholar 

  60. Lundberg Å, Nyman AK, Aspán A, Börjesson S, Unnerstad HE, Waller KP. Udder infections with Staphylococcus aureus, Streptococcus dysgalactiae, and Streptococcus uberis at calving in dairy herds with suboptimal udder health. J Dairy Sci.2016;99:2102-17. https://doi.org/10.3168/jds.2015-9487.

  61. Stalder U, Stephan R, Corti S, Bludau M, Maeschli A, Klocke P, et al. Short communication: Staphylococcus aureus isolated from colostrum of dairy heifers represent a closely related group exhibiting highly homogeneous genomic and antimicrobial resistance features. J Dairy Sci. 2014;97:4997–5000. https://doi.org/10.3168/jds.2013-7721.

    Article  CAS  PubMed  Google Scholar 

  62. Käppeli N, Morach M, Corti S, Eicher C, Stephan R, Johler S. Staphylococcus aureus related to bovine mastitis in Switzerland: Clonal diversity, virulence gene profiles, and antimicrobial resistance of isolates collected throughout 2017. J Dairy Sci. 2019;102:3274–81. https://doi.org/10.3168/jds.2018-15317.

    Article  CAS  PubMed  Google Scholar 

  63. Resch G, François P, Morisset D, Stojanov M, Bonetti EJ, Schrenzel J, et al. Human-to-bovine jump of Staphylococcus aureus CC8 is associated with the loss of a β-hemolysin converting prophage and the acquisition of a new staphylococcal cassette chromosome. PLoS ONE. 2013;8:e58187. https://doi.org/10.1371/journal.pone.0058187.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Moser A, Stephan R, Corti S, Johler S. Comparison of genomic and antimicrobial resistance features of latex agglutination test-positive and latex agglutination test-negative Staphylococcus aureus isolates causing bovine mastitis. J Dairy Sci. 2013;96:329–34. https://doi.org/10.3168/jds.2012-5944.

    Article  CAS  PubMed  Google Scholar 

  65. Tasara T, Cernela N, Stephan R. Function impairing mutations in blaZ and blaR genes of penicillin-susceptible Staphylococcus aureus strains isolated from bovine mastitis. Schweiz Arch Tierheilkd. 2013;155:359–63. https://doi.org/10.1024/0036-7281/a000471.

    Article  CAS  PubMed  Google Scholar 

  66. Hummerjohann J, Naskova J, Baumgartner A, Graber HU. Enterotoxin-producing Staphylococcus aureus genotype B as a major contaminant in Swiss raw milk cheese. J Dairy Sci. 2014;97:1305–12. https://doi.org/10.3168/jds.2013-7643.

    Article  CAS  PubMed  Google Scholar 

  67. Rüegsegger C, Sihto J. Toxic bovine mastitis caused by Staphylococcus aureus in twin cows. Schweiz Arch Tierheilkd. 2014;156:539–42. https://doi.org/10.1024/0036-7281/a000644.

    Article  PubMed  Google Scholar 

  68. Johler S, Weder D, Bridy C, Huguenin MC, Robert L, Hummerjohann J, et al. Outbreak of staphylococcal food poisoning among children and staff at a Swiss boarding school due to soft cheese made from raw milk. J Dairy Sci. 2015;98:2944–8. https://doi.org/10.3168/jds.2014-9123.

    Article  CAS  PubMed  Google Scholar 

  69. Merz A, Stephan R, Johler S. Staphylococcus aureus isolates from goat and sheep milk seem to be closely related and differ from isolates detected from bovine milk. Front Microbiol. 2016;14:7:319. https://doi.org/10.3389/fmicb.2016.00319.

  70. Leuenberger A, Sartori C, Boss R, Resch G, Oechslin F, Steiner A, et al. Genotypes of Staphylococcus aureus: on-farm epidemiology and the consequences for prevention of intramammary infections. J Dairy Sci. 2019;102:3295–309. https://doi.org/10.3168/jds.2018-15181.

    Article  CAS  PubMed  Google Scholar 

  71. Ben Said M, Abbassi MS, Bianchini V, Sghaier S, Cremonesi P, Romanò A, et al. Genetic characterization and antimicrobial resistance of Staphylococcus aureus isolated from bovine milk in Tunisia. Lett Appl Microbiol. 2016;63:473–81. https://doi.org/10.1111/lam.12672.

    Article  CAS  PubMed  Google Scholar 

  72. Klibi A, Jouini A, Gómez P, Slimene K, Ceballos S, Torres C, et al. Molecular characterization and clonal diversity of methicillin-resistant and -susceptible Staphylococcus aureus isolates of milk of cows with clinical mastitis in Tunisia. Microb Drug Resist. 2018;24:1210–6. https://doi.org/10.1089/mdr.2017.0278.

    Article  CAS  PubMed  Google Scholar 

  73. Kateete DP, Kabugo U, Baluku H, Nyakarahuka L, Kyobe S, Okee M, et al. Prevalence and antimicrobial susceptibility patterns of bacteria from milkmen and cows with clinical mastitis in and around Kampala, Uganda. PLoS ONE. 2013;8:e63413. https://doi.org/10.1371/journal.pone.0063413.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Jamrozy DM, Fielder MD, Butaye P, Coldham NG. Comparative genotypic and phenotypic characterisation of methicillin-resistant Staphylococcus aureus ST398 isolated from animals and humans. PLoS ONE. 2012;7:e40458. https://doi.org/10.1371/journal.pone.0040458.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Paterson GK, Morgan FJ, Harrison EM, Peacock SJ, Parkhill J, Zadoks RN, et al. Prevalence and properties of mecC methicillin-resistant Staphylococcus aureus (MRSA) in bovine bulk tank milk in Great Britain. J Antimicrob Chemother. 2014;69:598–602. https://doi.org/10.1093/jac/dkt417.

    Article  CAS  PubMed  Google Scholar 

  76. Haran KP, Godden SM, Boxrud D, Jawahir S, Bender JB, Sreevatsan S. Prevalence and characterization of Staphylococcus aureus, including methicillin-resistant Staphylococcus aureus, isolated from bulk tank milk from Minnesota dairy farms. J Clin Microbiol. 2012;50:688–95. https://doi.org/10.1128/JCM.05214-11.

  77. Layer F, Ghebremedhin B, Konig W, Konig B. Heterogeneity of methicillin-susceptible Staphylococcus aureus strains at a German University Hospital implicates the circulating-strain pool as a potential source of emerging methicillin-resistant S. aureus clones. J Clin Microbiol. 2006;44:2179–85. https://doi.org/10.1128/jcm.02321-05.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Haag AF, Fitzgerald JR, Penadés JR. Staphylococcus aureus in animals. Microbiol Spectr. 2019;7(3). https://doi.org/10.1128/microbiolspec.gpp3-0060-2019.

  79. Alba P, Feltrin F, Cordaro G, Porrero MC, Kraushaar B, Argudín MA, et al. Livestock-associated methicillin-resistant and methicillin-susceptible Staphylococcus aureus sequence type (CC)1 in European farmed animals: high genetic relatedness of isolates from Italian cattle herds and humans. PLoS ONE. 2015;10:e0137143. https://doi.org/10.1371/journal.pone.0137143.g001.

    Article  PubMed  PubMed Central  Google Scholar 

  80. Hoekstra J, Zomer AL, Rutten VPMG, Benedictus L, Stegeman A, Spaninks MP, et al. Genomic analysis of European bovine Staphylococcus aureus from clinical versus subclinical mastitis. Sci Rep. 2020;10:18172. https://doi.org/10.1038/s41598-020-75179-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Silva NCC, Guimarães FF, Manzi MP, Budri PE, Gómez-Sanz E, Benito D, et al. Molecular characterization and clonal diversity of methicillin-susceptible Staphylococcus aureus in milk of cows with mastitis in Brazil. J Dairy Sci. 2013;96:6856–62. https://doi.org/10.3168/jds.2013-6719.

    Article  CAS  PubMed  Google Scholar 

  82. Hoekstra J, Rutten V, Sommeling L, Van Werven T, Spaninks M, Duim B, et al. High production of LukMF’ in Staphylococcus aureus field strains is associated with clinical bovine mastitis. Toxins. 2018;10:200. https://doi.org/10.3390/toxins10050200.

    Article  CAS  PubMed Central  Google Scholar 

  83. Hoekstra J, Rutten VPMG, Lam TJGM, Van Kessel KPM, Spaninks MP, Stegeman JA, et al. Activation of a bovine mammary epithelial cell line by ruminant-associated Staphylococcus aureus is Lineage Dependent. Microorganisms. 2019;7:668. https://doi.org/10.3390/microorganisms7120688.

    Article  CAS  Google Scholar 

  84. Guardabassi L, Larsen J, Weese JS, Butaye P, Battisti A, Kluytmans J, et al. Public health impact and antimicrobial selection of meticillin-resistant staphylococci in animals. J Glob Antimicrob Resist. 2013;1:55–62. https://doi.org/10.1016/j.jgar.2013.03.011.

    Article  CAS  PubMed  Google Scholar 

  85. McCarthy AJ, Lindsay JA, Loeffler A. Are all methicillin-resistant Staphylococcus aureus (MRSA) equal in all hosts? Epidemiological and genetic comparison between animal and human MRSA. Vet Dermatol. 2012;23:267–75. https://doi.org/10.1111/j.1365-3164.2012.01072.x.

    Article  PubMed  Google Scholar 

  86. Richardson EJ, Bacigalupe R, Harrison EM, Weinert LA, Lycett S, Vrieling M, et al. Gene exchange drives the ecological success of a multi-host bacterial pathogen. Nat Ecol Evol. 2018;2:1468–78. https://doi.org/10.1038/s41559-018-0617-0.

    Article  PubMed  PubMed Central  Google Scholar 

  87. Tenover FC, Goering RV. Methicillin-resistant Staphylococcus aureus strain USA300: origin and epidemiology. J Antimicrob Chemother. 2009;64:441–6. https://doi.org/10.1093/jac/dkp241.

    Article  CAS  PubMed  Google Scholar 

  88. Peton V, Le Loir Y. Staphylococcus aureus in veterinary medicine. Infect Genet Evol. 2014;21:602–15. https://doi.org/10.1016/j.meegid.2013.08.011.

    Article  PubMed  Google Scholar 

  89. Sakwinska O, Giddey M, Moreillon M, Morisset D, Waldvogel A, Moreillon P. Staphylococcus aureus host range and human-bovine host shift. Appl Environ Microbiol. 2011;77:5908–15. https://doi.org/10.1128/aem.00238-11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Herron-Olson L, Fitzgerald JR, Musser JM, Kapur V. Molecular correlates of host specialization in Staphylococcus aureus. PLoS ONE. 2007;2:e1120. https://doi.org/10.1371/journal.pone.0001120.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Weinert LA, Welch JJ, Suchard MA, Lemey P, Rambaut A, Fitzgerald JR. Molecular dating of human-to-bovid host jumps by Staphylococcus aureus reveals an association with the spread of domestication. Biol Lett. 2012;8:829–32. https://doi.org/10.1098/rsbl.2012.0290.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Guinane CM, Ben Zakour NL, Tormo-Mas MA, Weinert LA, Lowder BV, Cartwright RA, et al. Evolutionary genomics of Staphylococcus aureus reveals insights into the origin and molecular basis of ruminant host adaptation. Genome Biol Evol. 2010;2:454–66. https://doi.org/10.1093/gbe/evq031.

  93. Fitzgerald JR, Monday SR, Foster TJ, Bohach GA, Hartigan PJ, Meaney WJ, et al. Characterization of a putative pathogenicity island from bovine Staphylococcus aureus encoding multiple superantigens. J Bacteriol. 2001;183:63–70. https://doi.org/10.1128/JB.183.1.63-70.2001.

  94. Viana D, Blanco J, Tormo-Más MA, Selva L, Guinane CM, Baselga R, et al. Adaptation of Staphylococcus aureus to ruminant and equine hosts involves SaPI-carried variants of von Willebrand factor-binding protein. Mol Microbiol. 2010;77:1583–94. https://doi.org/10.1111/j.1365-2958.2010.07312.x.

    Article  CAS  PubMed  Google Scholar 

  95. Cheng AG, McAdow M, Kim HK, Bae T, Missiakas DM, Schneewind O. Contribution of coagulases towards Staphylococcus aureus disease and protective immunity. PLoS Pathog. 2010;6:e1001036. https://doi.org/10.1371/journal.ppat.1001036.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Vrieling M, Boerhout EM, van Wigcheren GF, Koymans KJ, Mols-Vorstermans TG, de Haas CJ, et al. LukMF’ is the major secreted leukocidin of bovine Staphylococcus aureus and is produced in vivo during bovine mastitis. Sci Rep. 2016;6:37759. https://doi.org/10.1038/srep37759.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Spoor LE, McAdam PR, Weinert LA, Rambaut A, Hasman H, Aarestrup FM, et al. Livestock origin for a human pandemic clone of community-associated methicillin-resistant Staphylococcus aureus. mBio. 13;4:e00356-13. https://doi.org/10.1128/mbio.00356-13.

  98. Ward MJ, Goncheva M, Richardson E, McAdam PR, Raftis E, Kearns A, et al. Identification of source and sink populations for the emergence and global spread of the East-Asia clone of community-associated MRSA. Genome Biol. 2016;17:160. https://doi.org/10.1186/s13059-016-1022-0.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Castilho IG, Dantas STA, Langoni H, Araújo JP Jr, Fernandes A Jr, Alvarenga FCL, Maia L, Cagnini DQ, Rall VLM. Host-pathogen interactions in bovine mammary epithelial cells and HeLa cells by Staphylococcus aureus isolated from subclinical bovine mastitis. J Dairy Sci. 2017;100(8):6414–21. https://doi.org/10.3168/jds.2017-12700.

    Article  CAS  PubMed  Google Scholar 

  100. Foster TJ, Geoghegan JA, Ganesh VK, Höök M. Adhesion, invasion and evasion: the many functions of the surface proteins of Staphylococcus aureus. Nat Rev Microbiol. 2014;12:49–62. https://doi.org/10.1038/nrmicro3161.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Lammers A, Nuijten PJ, Smith HE. The fibronectin-binding proteins of Staphylococcus aureus are required for adhesion to and invasion of bovine mammary gland cells. FEMS Microbiol Lett. 1999;180:103–9. https://doi.org/10.1111/j.1574-6968.1999.tb08783.x.

    Article  CAS  PubMed  Google Scholar 

  102. Sinha B, François PP, Nüsse O, Foti M, Hartford OM, Vaudaux P, et al. Fibronectin-binding protein acts as Staphylococcus aureus invasin via fibronectin bridging to integrin alpha5beta1. Cell Microbiol. 1999;1:101–17. https://doi.org/10.1046/j.1462-5822.1999.00011.x.

    Article  CAS  PubMed  Google Scholar 

  103. Fowler T, Wann ER, Joh D, Johansson S, Foster TJ, Höök M. Cellular invasion by Staphylococcus aureus involves a fibronectin bridge between the bacterial fibronectin-binding MSCRAMMs and host cell beta1 integrins. Eur J Cell Biol. 2000;79:672–9. https://doi.org/10.1078/0171-9335-00104.

    Article  CAS  PubMed  Google Scholar 

  104. Agerer F, Lux S, Michel A, Rohde M, Ohlsen K, Hauck CR. Cellular invasion by Staphylococcus aureus reveals a functional link between focal adhesion kinase and cortactin in integrin-mediated internalisation. J Cell Sci. 2005;118:2189–200. https://doi.org/10.1242/jcs.02328.

    Article  CAS  PubMed  Google Scholar 

  105. Dziewanowska K, Patti JM, Deobald CF, Bayles KW, Trumble WR, Bohach GA. Fibronectin binding protein and host cell tyrosine kinase are required for internalization of Staphylococcus aureus by epithelial cells. Infect Immun. 1999;67:4673–8. https://doi.org/10.1128/iai.67.9.4673-4678.1999.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Roche FM, Downer R, Keane F, Speziale P, Park PW, Foster TJ. The N-terminal A domain of fibronectin-binding proteins A and B promotes adhesion of Staphylococcus aureus to elastin. J Biol Chem. 2004;279:38433–40. https://doi.org/10.1074/jbc.m402122200.

    Article  CAS  PubMed  Google Scholar 

  107. Pietrocola G, Nobile G, Gianotti V, Zapotoczna M, Foster TJ, Geoghegan JA, et al. Molecular interactions of human plasminogen with fibronectin-binding protein B (FnBPB), a fibrinogen/fibronectin-binding protein from Staphylococcus aureus. J Biol Chem. 2016;291:18148–62. https://doi.org/10.1074/jbc.M116.731125.

  108. Brouillette E, Talbot BG, Malouin F. The fibronectin-binding proteins of Staphylococcus aureus may promote mammary gland colonization in a lactating mouse model of mastitis. Infect Immun. 2003;71:2292–5. https://doi.org/10.1128/IAI.71.4.2292-2295.2003.

  109. Pereyra EA, Picech F, Renna MS, Baravalle C, Andreotti CS, Russi R, Calvinho LF, Diez C, Dallard BE. Detection of Staphylococcus aureus adhesion and biofilm-producing genes and their expression during internalization in bovine mammary epithelial cells. Vet Microbiol. 2016;183:69–77. https://doi.org/10.1016/j.vetmic.2015.12.002.

    Article  CAS  PubMed  Google Scholar 

  110. Capra E, Cremonesi P, Pietrelli A, Puccio S, Luini M, Stella A, et al. Genomic and transcriptomic comparison between Staphylococcus aureus strains associated with high and low within herd prevalence of intra-mammary infection. BMC Microbiol. 2017;17:21. https://doi.org/10.1186/s12866-017-0931-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Klein RC, Fabres-Klein MH, Brito MA, Fietto LG, Ribon AOB. Staphylococcus aureus of bovine origin: genetic diversity, prevalence and the expression of adhesin-encoding genes. Vet Microbiol. 2012;160:183–8. https://doi.org/10.1016/j.vetmic.2012.05.025.

    Article  CAS  PubMed  Google Scholar 

  112. Xu J, Tan X, Zhang X, Xia X, Sun H. The diversities of staphylococcal species, virulence and antibiotic resistance genes in the subclinical mastitis milk from a single Chinese cow herd. Microb Pathog. 2015;88:29–38. https://doi.org/10.1016/j.micpath.2015.08.004.

    Article  CAS  PubMed  Google Scholar 

  113. Khoramrooz SS, Mansouri F, Marashifard M, Malek Hosseini SA, Akbarian Chenarestane-Olia F, Ganavehei B, Gharibpour F, Shahbazi A, Mirzaii M, Darban-Sarokhalil D. Detection of biofilm related genes, classical enterotoxin genes and agr typing among Staphylococcus aureus isolated from bovine with subclinical mastitis in southwest of Iran. Microb Pathog. 2016;97:45–51. https://doi.org/10.1016/j.micpath.2016.05.022.

    Article  CAS  PubMed  Google Scholar 

  114. Gogoi-Tiwari J, Waryah CB, Eto KY, Tau M, Wells K, Costantino P, et al. Relative distribution of virulence-associated factors among Australian bovine Staphylococcus aureus isolates: potential relevance to development of an effective bovine mastitis vaccine. Virulence. 2015;6:419–23. https://doi.org/10.1080/21505594.2015.1043508.

    Article  PubMed  PubMed Central  Google Scholar 

  115. Wang D, Zhang L, Zhou X, He Y, Yong C, Shen M, et al. Antimicrobial susceptibility, virulence genes, and randomly amplified polymorphic DNA analysis of Staphylococcus aureus recovered from bovine mastitis in Ningxia, China. J Dairy Sci. 2016;99:9560–9. https://doi.org/10.3168/jds.2016-11625.

    Article  CAS  PubMed  Google Scholar 

  116. Ster C, Gilbert FB, Cochard T, Poutrel B. Transcriptional profiles of regulatory and virulence factors of Staphylococcus aureus of bovine origin: oxygen impact and strain-to-strain variations. Mol Cell Probes. 2005;19:227–35. https://doi.org/10.1016/j.mcp.2005.01.002.

    Article  CAS  PubMed  Google Scholar 

  117. Misra N, Wines TF, Knopp CL, McGuire MA, Tinker JK. Expression, immunogenicity and variation of iron-regulated surface protein A from bovine isolates of Staphylococcus aureus. FEMS Microbiol Lett. 2017;364:fnx082. https://doi.org/10.1093/femsle/fnx082.

    Article  CAS  PubMed Central  Google Scholar 

  118. McDevitt D, Francois P, Vaudaux P, Foster TJ. Molecular characterization of the clumping factor (fibrinogen receptor) of Staphylococcus aureus. Mol Microbiol. 1994;11:237–48. https://doi.org/10.1111/j.1365-2958.1994.tb00304.x.

    Article  CAS  PubMed  Google Scholar 

  119. Ní Eidhin D, Perkins S, Francois P, Vaudaux P, Höök M, Foster TJ. Clumping factor B (ClfB), a new surface-located fibrinogen-binding adhesin of Staphylococcus aureus. Mol Microbiol. 1998;30:245–57. https://doi.org/10.1046/j.1365-2958.1998.01050.x.

    Article  PubMed  Google Scholar 

  120. McDonnell CJ, Garciarena CD, Watkin RL, McHale TM, McLoughlin A, Claes J, et al. Inhibition of major integrin α αVβ3 reduces Staphylococcus aureus attachment to sheared human endothelial cells. J Thromb Haemost. 2016;14:2536–47. https://doi.org/10.1111/jth.13501.

    Article  CAS  PubMed  Google Scholar 

  121. Ashraf S, Cheng J, Zhao X. Clumping factor A of Staphylococcus aureus interacts with AnnexinA2 on mammary epithelial cells. Sci Rep. 2017;7:40608. https://doi.org/10.1038/srep40608.

  122. Hair PS, Echague CG, Sholl AM, Watkins JA, Geoghegan JA, Foster TJ, et al. Clumping factor A interaction with complement factor I increases C3b cleavage on the bacterial surface of Staphylococcus aureus and decreases complement-mediated phagocytosis. Infect Immun. 2010;78:1717–27. https://doi.org/10.1128/iai.01065-09.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Ikawaty R, Brouwer E, Van Duijkeren E, Mevius D, Verhoef J, Fluit A. Virulence factors of genotyped bovine mastitis Staphylococcus aureus isolates in the Netherlands. Int J Dairy Sci. 2010;5:60–70. https://doi.org/10.3923/ijds.2010.60.70.

    Article  CAS  Google Scholar 

  124. Monistero V, Graber HU, Pollera C, Cremonesi P, Castiglioni B, Bottini E, et al. Isolates from bovine mastitis in eight countries: genotypes, detection of genes encoding different toxins and other virulence genes. Toxins. 2018;10:247. https://doi.org/10.3390/toxins10060247.

    Article  CAS  PubMed Central  Google Scholar 

  125. Kang M, Ko YP, Liang X, Ross CL, Liu Q, Murray BE, et al. Collagen-binding microbial surface components recognizing adhesive matrix molecules (MSCRAMM) of Gram-positive bacteria inhibit complement activation via the classical pathway. J Biol Chem. 2013;288:20520–31. https://doi.org/10.1074/jbc.m113.454462.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Magro G, Biffani S, Minozzi G, Ehricht R, Monecke S, Luini M, et al. Virulence genes of Staphylococcus aureus from dairy cow mastitis and contagiousness risk. Toxins. 2017;9:195. https://doi.org/10.3390/toxins9060195.

    Article  CAS  PubMed Central  Google Scholar 

  127. Xue H, Lu H, Zhao X. Sequence diversities of serine-aspartate repeat genes among Staphylococcus aureus isolates from different hosts presumably by horizontal gene transfer. PLoS ONE. 2011;6:e20332. https://doi.org/10.3390/toxins9060195.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Clarke SR, Wiltshire MD, Foster SJ. IsdA of Staphylococcus aureus is a broad spectrum, iron-regulated adhesin. Mol Microbiol. 2004;51:1509–19. https://doi.org/10.1111/j.1365-2958.2003.03938.x.

    Article  CAS  PubMed  Google Scholar 

  129. Ster C, Beaudoin F, Diarra MS, Jacques M, Malouin F, Lacasse P. Evaluation of some Staphylococcus aureus iron-regulated proteins as vaccine targets. Vet Immunol Immunopathol. 2010;136:311–8. https://doi.org/10.1016/j.vetimm.2010.03.010.

    Article  CAS  PubMed  Google Scholar 

  130. Nagasawa Y, Uchida I, Tanabe F, Hirose A, Sugawara K, Kiku Y, et al. Intramammary infection caused by Staphylococcus aureus increases IgA antibodies to iron-regulated surface determinant-A, -B, and -H in bovine milk. Vet Immunol Immunopathol. 2021;235:110235. https://doi.org/10.1016/j.vetimm.2021.110235.

    Article  CAS  PubMed  Google Scholar 

  131. Mamo W, Lindahl M, Jonsson P. Enhanced virulence of Staphylococcus aureus from bovine mastitis induced by growth in milk whey. Vet Microbiol. 1991;27:371–84. https://doi.org/10.1016/0378-1135(91)90161-8.

    Article  CAS  PubMed  Google Scholar 

  132. Mamo W, Fröman G. Adhesion of Staphylococcus aureus to bovine mammary epithelial cells induced by growth in milk whey. Microbiol Immunol. 1994;38:305–8. https://doi.org/10.1111/j.1348-0421.1994.tb01781.x.

    Article  CAS  PubMed  Google Scholar 

  133. Cifrian E, Guidry AJ, O’Brien CN, Nickerson SC, Marquardt WW. Adherence of Staphylococcus aureus to cultured bovine mammary epithelial cells. J Dairy Sci. 1994;77:970–83. https://doi.org/10.3168/jds.S0022-0302(94)77033-8.

  134. Hensen SM, Pavicić MJ, Lohuis JA, Poutrel B. Use of bovine primary mammary epithelial cells for the comparison of adherence and invasion ability of Staphylococcus aureus strains. J Dairy Sci. 2000;83:418–29. https://doi.org/10.3168/jds.s0022-0302(00)74898-3.

    Article  CAS  PubMed  Google Scholar 

  135. Tuchscherr L, Heitmann V, Hussain M, Viemann D, Roth J, von Eiff C, et al. Staphylococcus aureus small-colony variants are adapted phenotypes for intracellular persistence. J Infect Dis. 2010;202:1031–40. https://doi.org/10.1086/656047.

    Article  PubMed  Google Scholar 

  136. Grunert T, Stessl B, Wolf F, Sordelli DO, Buzzola FR, Ehling-Schulz M. Distinct phenotypic traits of Staphylococcus aureus are associated with persistent, contagious bovine intramammary infections. Sci Rep. 2018;8:15968. https://doi.org/10.1038/s41598-018-34371-1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Babra C, Tiwari JG, Pier G, Thein TH, Sunagar R, Sundareshan S, Isloor S, Hegde NR, de Wet S, Deighton M, Gibson J, Costantino P, Wetherall J, Mukkur T. The persistence of biofilm-associated antibiotic resistance of Staphylococcus aureus isolated from clinical bovine mastitis cases in Australia. Folia Microbiol. 2013;58:469–74. https://doi.org/10.1007/s12223-013-0232-z.

    Article  CAS  Google Scholar 

  138. Felipe V, Morgante CA, Somale PS, Varroni F, Zingaretti ML, Bachetti RA, Correa SG, Porporatto C. Evaluation of the biofilm forming ability and its associated genes in Staphylococcus species isolates from bovine mastitis in Argentinean dairy farms. Microb Pathog. 2017;104:278–86. https://doi.org/10.1016/j.micpath.2017.01.047.

    Article  CAS  PubMed  Google Scholar 

  139. Melchior MB, Fink-Gremmels J, Gaastra W. Comparative assessment of the antimicrobial susceptibility of Staphylococcus aureus isolates from bovine mastitis in biofilm versus planktonic culture. J Vet Med B Infect Dis Vet Public Health. 2006;53:326–32. https://doi.org/10.1111/j.1439-0450.2006.00962.x.

    Article  CAS  PubMed  Google Scholar 

  140. Schönborn S, Krömker V. Detection of the biofilm component polysaccharide intercellular adhesin in Staphylococcus aureus infected cow udders. Vet Microbiol. 2016;196:126–8. https://doi.org/10.1016/j.vetmic.2016.10.023.

    Article  CAS  PubMed  Google Scholar 

  141. Fabres-Klein MH, Caizer Santos MJ, Contelli Klein R, Nunes de Souza G, de Oliveira Barros Ribon A. An association between milk and slime increases biofilm production by bovine Staphylococcus aureus. BMC Vet Res. 2015;11:3. https://doi.org/10.1186/s12917-015-0319-7.

    Article  PubMed  PubMed Central  Google Scholar 

  142. Torres G, Vargas K, Sánchez-Jiménez M, Reyes-Velez J, Olivera-Angel M. Genotypic and phenotypic characterization of biofilm production by Staphylococcus aureus strains isolated from bovine intramammary infections in Colombian dairy farms. Heliyon. 2019;5:e02535. https://doi.org/10.1016/j.heliyon.2019.e02535.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Bohl LP, Isaac P, Breser ML, Orellano MS, Correa SG, Tolosa de Talamoni NG, Porporatto C. 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. 2021;152:104604. https://doi.org/10.1016/j.micpath.2020.104604.

    Article  CAS  PubMed  Google Scholar 

  144. Melchior MB, van Osch MH, Graat RM, van Duijkeren E, Mevius DJ, Nielen M, Gaastra W, Fink-Gremmels J. Biofilm formation and genotyping of Staphylococcus aureus bovine mastitis isolates: evidence for lack of penicillin-resistance in Agr-type II strains. Vet Microbiol. 2009;137:83–9. https://doi.org/10.1016/j.vetmic.2008.12.004.

    Article  CAS  PubMed  Google Scholar 

  145. Bardiau M, Detilleux J, Farnir F, Mainil JG, Ote I. Associations between properties linked with persistence in a collection of Staphylococcus aureus isolates from bovine mastitis. Vet Microbiol. 2014;169:74–9. https://doi.org/10.1016/j.vetmic.2013.12.010.

    Article  PubMed  Google Scholar 

  146. Bardiau M, Caplin J, Detilleux J, Graber H, Moroni P, Taminiau B, Mainil JG. 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. 2016;185:1–6. https://doi.org/10.1016/j.vetmic.2016.01.003.

    Article  CAS  PubMed  Google Scholar 

  147. Rossi BF, Bonsaglia ECR, Pantoja JCF, Santos MV, Gonçalves JL, Fernandes Júnior A, Rall VLM. Association between the accessory gene regulator (agr) group and the severity of bovine mastitis caused by Staphylococcus aureus. J Dairy Sci. 2021;104:3564–8. https://doi.org/10.3168/jds.2020-19275.

    Article  CAS  PubMed  Google Scholar 

  148. Xue T, Chen X, Shang F. Short communication: Effects of lactose and milk on the expression of biofilm-associated genes in Staphylococcus aureus strains isolated from a dairy cow with mastitis. J Dairy Sci. 2014;97:6129–34. https://doi.org/10.3168/jds.2014-8344.

    Article  CAS  PubMed  Google Scholar 

  149. Sharer MV, Su C, Hegde NV, Jayarao BM, Sordillo LM. Differential expression of the lactose transporter gene affects growth of Staphylococcus aureus in milk. J Dairy Sci. 2003;86:2373–81. https://doi.org/10.3168/jds.S0022-0302(03)73831-4.

    Article  CAS  PubMed  Google Scholar 

  150. Cue D, Lei MG, Luong TT, Kuechenmeister L, Dunman PM, O’Donnell S, Rowe S, O’Gara JP, Lee CY. Rbf promotes biofilm formation by Staphylococcus aureus via repression of icaR, a negative regulator of icaADBC. J Bacteriol. 2009;191:6363–73. https://doi.org/10.1128/JB.00913-09.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Formosa-Dague C, Feuillie C, Beaussart A, Derclaye S, Kucharíková S, Lasa I, Van Dijck P, Dufrêne YF. Sticky matrix: adhesion mechanism of the staphylococcal polysaccharide intercellular adhesin. ACS Nano. 2016;10:3443–52. https://doi.org/10.1021/acsnano.5b07515.

    Article  CAS  PubMed  Google Scholar 

  152. Waller PK, Bengtsson B, Lindberg A, Nyman A, Ericsson Unnerstad H. Incidence of mastitis and bacterial findings at clinical mastitis in Swedish primiparous cows—influence of breed and stage of lactation. Vet Microbiol. 2009;134:89–94. https://doi.org/10.1016/j.vetmic.2008.09.004.

    Article  Google Scholar 

  153. Abebe R, Hatiya H, Abera M, Megersa B, Asmare K. Bovine mastitis: Prevalence, risk factors and isolation of Staphylococcus aureus in dairy herds at Hawassa milk shed, South Ethiopia. BMC Vet Res. 2016;12:270. https://doi.org/10.1186/s12917-016-0905-3.

    Article  PubMed  PubMed Central  Google Scholar 

  154. Fox LK, Zadoks RN, Gaskins CT. Biofilm production by Staphylococcus aureus associated with intramammary infection. Vet Microbiol. 2005;107:295–9. https://doi.org/10.1016/j.vetmic.2005.02.005.

    Article  CAS  PubMed  Google Scholar 

  155. Cucarella C, Tormo MA, Ubeda C, Trotonda MP, Monzón M, Peris C, Amorena B, Lasa I, Penadés JR. Role of biofilm-associated protein Bap in the pathogenesis of bovine Staphylococcus aureus. Infect Immun. 2004;72:2177–85. https://doi.org/10.1128/IAI.72.4.2177-2185.2004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Valle J, Latasa C, Gil C, Toledo-Arana A, Solano C, Penadés JR, Lasa I. Bap, a biofilm matrix protein of Staphylococcus aureus prevents cellular internalization through binding to GP96 host receptor. PLoS Pathog. 2012;8:e1002843. https://doi.org/10.1371/journal.ppat.1002843.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Taglialegna A, Navarro S, Ventura S, Garnett JA, Matthews S, Penades JR, Lasa I, Valle J. Staphylococcal Bap proteins build amyloid scaffold biofilm matrices in response to environmental signals. PLoS Pathog. 2016;12:e1005711. https://doi.org/10.1371/journal.ppat.1005711.

  158. Tam K, Torres VJ. Staphylococcus aureus secreted toxins and extracellular enzymes. Microbiol Spectr. 2019;7(2):10.1128. https://doi.org/10.1128/microbiolspec.GPP3-0039-2018.

  159. Tollersrud T, Kampen AH, Kenny K. Staphylococcus aureus enterotoxin D is secreted in milk and stimulates specific antibody responses in cows in the course of experimental intramammary infection. Infect Immun. 2006;74:3507–12. https://doi.org/10.1128/IAI.01726-05.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Actor JK. Role of Major histocompatibility complex in the immune response. In: Elsevier’s Integrated Review Immunology and Microbiology. 2012. p. 33–41.

  161. Wilson GJ, Tuffs SW, Wee BA, Seo KS, Park N, Connelley T, et al. Bovine Staphylococcus aureus superantigens stimulate the entire T cell repertoire of cattle. Infect Immun. 2018;86:1–16. https://doi.org/10.1128/IAI.00505-18.

    Article  Google Scholar 

  162. Larsen HD, Aarestrup FM, Jensen NE. Geographical variation in the presence of genes encoding superantigenic exotoxins and β-hemolysin among Staphylococcus aureus isolated from bovine mastitis in Europe and USA. Vet Microbiol. 2002;85:61–7. https://doi.org/10.1016/s0378-1135(01)00478-3.

  163. Elsayed MS, El-Bagoury AEM, Dawoud MA. Phenotypic and genotypic detection of virulence factors of Staphylococcus aureus isolated from clinical and subclinical mastitis in cattle and water buffaloes from different farms of Sadat City in Egypt. Vet World. 2015;8:1051–8. https://doi.org/10.14202/vetworld.2015.1051-1058.

    Article  PubMed  PubMed Central  Google Scholar 

  164. Wang D, Zhang L, Yong C, Shen M, Ali T, Shahid M, et al. Relationships among superantigen toxin gene profiles, genotypes, and pathogenic characteristics of Staphylococcus aureus isolates from bovine mastitis. J Dairy Sci. 2017;100:4276–86. doi:https://doi.org/10.3168/jds.2016-12405.

    Article  CAS  PubMed  Google Scholar 

  165. Da Silva ER, Do Carmo LS, Da Silva N. Detection of the enterotoxins A, B, and C genes in Staphylococcus aureus from goat and bovine mastitis in Brazilian dairy herds. Vet Microbiol. 2005;106:103–7. https://doi.org/10.1016/j.vetmic.2004.12.005.

  166. Haveri M, Roslöf A, Rantala L, Pyörälä S. Virulence genes of bovine Staphylococcus aureus from persistent and nonpersistent intramammary infections with different clinical characteristics. J Appl Microbiol. 2007;103:993–1000. https://doi.org/10.1111/j.1365-2672.2007.03356.x.

    Article  CAS  PubMed  Google Scholar 

  167. Fueyo JM, Mendoza MC, Rodicio MR, Muñiz J, Alvarez MA, Martín MC. Cytotoxin and pyrogenic toxin superantigen gene profiles of Staphylococcus aureus associated with subclinical mastitis in dairy cows and relationships with macrorestriction genomic profiles. J Clin Microbiol. 2005;43:1278–84. https://doi.org/10.1128/JCM.43.3.1278-1284.2005.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Oliveira L, Rodrigues AC, Hulland C, Ruegg PL. Enterotoxin production, enterotoxin gene distribution, and genetic diversity of Staphylococcus aureus recovered from milk of cows with subclinical mastitis. Am J Vet Res. 2011;72:1361–8. https://doi.org/10.2460/ajvr.72.10.1361. 

  169. Liu Y, Chen W, Ali T, Alkasir R, Yin J, Liu G, Han B. Staphylococcal enterotoxin H induced apoptosis of bovine mammary epithelial cells in vitro. Toxins. 2014;6:3552–67. https://doi.org/10.3390/toxins6123552.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Zhao Y, Tang J, Yang D, Tang C, Chen J. Staphylococcal enterotoxin M induced inflammation and impairment of bovine mammary epithelial cells. J Dairy Sci. 2020;103:8350–9. https://doi.org/10.3168/jds.2019-17444.

    Article  CAS  PubMed  Google Scholar 

  171. Fang R, Cui J, Cui T, Guo H, Ono HK, Park CH, et al. Staphylococcal enterotoxin C is an important virulence factor for mastitis. Toxins. 2019;11:1–13. https://doi.org/10.3390/toxins11030141.

    Article  CAS  Google Scholar 

  172. Huseby M, Shi K, Kent Brown C, Digre J, Mengistu F, Keun SS, et al. Structure and biological activities of beta toxin from Staphylococcus aureus. J Bacteriol. 2007;189:8719–26. https://doi.org/10.1128/JB.00741-07.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Von Hoven G, Rivas AJ, Neukirch C, Klein S, Hamm C, Qin Q, et al. Dissecting the role of ADAM10 as a mediator of Staphylococcus aureus α-toxin action. Biochem J. 2016;473:1929–40. https://doi.org/10.1042/BCJ20160062.

    Article  CAS  Google Scholar 

  174. Vann JM, Proctor RA. Cytotoxic effects of ingested Staphylococcus aureus on bovine endothelial cells: role of S. aureus α-hemolysin. Microb Pathog. 1988;4:443–53. https://doi.org/10.1016/0882-4010(88)90029-0.

  175. Cifrian E, Guidry AJ, Bramley AJ, Norcross NL, Bastida-Corcuera FD, Marquardt WW. Effect of staphylococcal β toxin on the cytotoxicity, proliferation and adherence of Staphylococcus aureus to bovine mammary epithelial cells. Vet Microbiol. 1996;48:187–98. https://doi.org/10.1016/0378-1135(95)00159-x.

  176. Verdon J, Girardin N, Lacombe C, Berjeaud JM, Héchard Y. δ-Hemolysin, an update on a membrane-interacting peptide. Peptides. 2009;30:817–23. https://doi.org/10.1016/j.peptides.2008.12.017.

    Article  CAS  PubMed  Google Scholar 

  177. Deplanche M, Alekseeva L, Semenovskaya K, Fu CL, Dessauge F, Finot L, et al. Staphylococcus aureus phenol-soluble modulins impair interleukin expression in bovine mammary epithelial cells. Infect Immun. 2016;84:1682–92. http://dx.doi.org/10.1128 /IAI.01330-15.

  178. Ali-Vehmas T, Vikerpuur M, Pyörälä S, Atroshi F. Characterization of hemolytic activity of Staphylococcus aureus strains isolated from bovine mastitic milk. Microbiol Res. 2001;155:339–44. https://doi.org/10.1016/S0944-5013(01)80013-6.

  179. Adlam C, Ward PD, Turner WH. Effect of immunization with highly purified Panton-Valentine leucocidin and delta-toxin on staphylococcal mastitis in rabbits. J Comp Pathol. 1980;90:265–74. https://doi.org/10.1016/0021-9975(80)90063-8.

  180. Ward PD, Adlam C, McCartney AC, Arbuthnott JP, Thorley CM. A histopathological study of the effects of highly purified staphylococcal alpha and beta toxins on the lactating mammary gland and skin of the rabbit. J Comp Pathol. 1979:169–77. https://doi.org/10.1016/0021-9975(79)90056-2.

  181. Bramley AJ, Patel AH, O’Reilly M, Foster R, Foster TJ. Roles of alpha-toxin and beta-toxin in virulence of Staphylococcus aureus for the mouse mammary gland. Infect Immun. 1989;57:2489–94. https://doi.org/10.1128/iai.57.8.2489-2494.1989.

  182. Kenny K, Bastida FD, Norcross NL. Secretion of alpha-hemolysin by bovine mammary isolates of Staphylococcus aureus. Can. J Vet Res. 1992;56:265–8.

    CAS  Google Scholar 

  183. Ote I, Taminiau B, Duprez JN, Dizier I, Mainil JG. Genotypic characterization by polymerase chain reaction of Staphylococcus aureus isolates associated with bovine mastitis. Vet Microbiol. 2011;153:285–92. https://doi.org/10.1016/j.vetmic.2011.05.042.

    Article  CAS  PubMed  Google Scholar 

  184. Fursova K, Sorokin A, Sokolov S, Dzhelyadin T, Shulcheva I, Shchannikova M, et al. Virulence factors and phylogeny of Staphylococcus aureus associated with bovine mastitis in Russia based on genome sequences. Front Vet Sci. 2020;7:1–10. https://doi.org/10.3389/fvets.2020.00135.

    Article  Google Scholar 

  185. Rajić Savić N, Katić V, Velebit B. Characteristics of coagulase-positive staphylococci isolated from milk in cases of subclinical mastitis. Acta Vet Brno. 2014;64:115–23. https://doi.org/10.2478/acve-2014-0012.

    Article  Google Scholar 

  186. Li T, Lu H, Wang X, Gao Q, Dai Y, Shang J, et al. Molecular characteristics of Staphylococcus aureus causing bovine mastitis between 2014 and 2015. Front Cell Infect Microbiol. 2017;7 APR:1–10. https://doi.org/10.3389/fcimb.2017.00127.

    Article  CAS  Google Scholar 

  187. Tavares A, Nielsen JB, Boye K, Rohde S, Paulo AC, Westh H, et al. Insights into alpha-hemolysin (Hla) evolution and expression among Staphylococcus aureus clones with hospital and community origin. PLoS ONE. 2014;9. https://doi.org/10.1371/journal.pone.0098634.

  188. Silva ER, Da, Boechat JUD, Martins JCD, Ferreira WPB, Siqueira AP, Silva N, Da. Hemolysin production by Staphylococcus aureus species isolated from mastitic goat milk in Brazilian dairy herds. Small Rumin Res. 2005;56:271–5. https://doi.org/10.1016/j.smallrumres.2004.04.011.

    Article  Google Scholar 

  189. Hall JW, Ji Y. Identification of predominant SNPs as a novel method for genotyping bovine Staphylococcus aureus isolates. Virulence. 2012;3:98–102. https://doi.org/10.4161/viru.3.1.18724.

    Article  PubMed  Google Scholar 

  190. Alonzo IIIF, Torres VJ. The bicomponent pore-forming leucocidins of Staphylococcus aureus. Microbiol Mol Biol R. 2014;78:199–230. https://doi.org/10.1128/MMBR.00055-13.

    Article  CAS  Google Scholar 

  191. Barrio MB, Rainard P, Prévost G. LukM/LukF’-PV is the most active Staphylococcus aureus leukotoxin on bovine neutrophils. Microb Infect. 2006;8:2068–74. https://doi.org/10.1016/j.micinf.2006.03.004.

    Article  CAS  Google Scholar 

  192. Rainard P, Corrales JC, Barrio MB, Cochard T, Poutrel B. Leucotoxic activities of Staphylococcus aureus strains isolated from cows, ewes, and goats with mastitis: Importance of LukM/LukF’-PV leukotoxin. Clin Vaccine Immunol. 2003;10:272–7. https://doi.org/10.1128/CDLI.10.2.272-277.2003.

    Article  CAS  Google Scholar 

  193. Mekonnen SA, Lam TJGM, Hoekstra J, Rutten VPMG, Tessema TS, Broens EM, et al. Characterization of Staphylococcus aureus isolated from milk samples of dairy cows in smallholder farms of North-Western Ethiopia. BMC Vet Res. 2018;14:246. https://doi.org/10.1186/s12917-018-1558-1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Yamada T, Tochimaru N, Nakasuji S, Hata E, Kobayashi H, Eguchi M, et al. Leukotoxin family genes in Staphylococcus aureus isolated from domestic animals and prevalence of lukM–lukF-PV genes by bacteriophages in bovine isolates. Vet Microbiol. 2005;110:97–103. https://doi.org/10.1016/j.vetmic.2005.07.006.

    Article  CAS  PubMed  Google Scholar 

  195. Schmidt T, Kock MM, Ehlers MM. Molecular characterization of Staphylococcus aureus isolated from bovine mastitis and close human contacts in South African dairy herds: genetic diversity and inter-species host transmission. Front Microbiol. 2017;8:511. https://doi.org/10.3389/fmicb.2017.00511.

    Article  PubMed  PubMed Central  Google Scholar 

  196. Rainard P. Staphylococcus aureus leucotoxin LukM/F’ is secreted and stimulates neutralising antibody response in the course of intramammary infection. Vet Res. 2007;38:685–96. https://doi.org/10.1051/vetres:2007026.

    Article  PubMed  Google Scholar 

  197. Fromageau A, Gilbert FB, Prévost G, Rainard P. Binding of the Staphylococcus aureus leucotoxin LukM to its leucocyte targets. Microb Pathog. 2010;49:354–62. https://doi.org/10.1016/j.micpath.2010.07.002.

    Article  CAS  PubMed  Google Scholar 

  198. Fromageau A, Cunha P, Gilbert FB, Rainard P. Purified Staphylococcus aureus leukotoxin LukM/F’ does not trigger inflammation in the bovine mammary gland. Microb Pathogen. 2011;51:396–401. https://doi.org/10.1016/j.micpath.2011.09.005.

    Article  CAS  Google Scholar 

  199. Vrieling M, Koymans KJ, Heesterbeek DAC, Aerts PC, Rutten VPMG, de Haas CJC, et al. Bovine Staphylococcus aureus secretes the leukocidin LukMF’ to kill migrating neutrophils through CCR1. mBio. 2015;6:e00335-15. https://doi.org/10.1128/mBio.00335-15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  200. Divyakolu S, Chikkala R, Ratnakar KS, Sritharan V. Hemolysins of Staphylococcus aureus—an update on their biology, role in pathogenesis and as targets for anti-virulence therapy. Adv Infect Dis. 2019;9:80–104. https://doi.org/10.4236/aid.2019.92007.

    Article  CAS  Google Scholar 

  201. Yang F, Zhang S, Shang X, Li H, Zhang H, Cui D, et al. Detection and molecular characterization of methicillin-resistant Staphylococcus aureus isolated from subclinical bovine mastitis cases in China. J Dairy Sci. 2020;103:840–5. https://doi.org/10.3168/jds.2019-16317.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The corresponding authors thank the members of their labs past and present who have contributed to our understanding of this topic.

Funding

J.R.F. was funded by institute strategic grant funding ISP2: BBS/E/D/20002173 and BBS/E/D/20002174 from the Biotechnology and Biological Sciences Research Council (United Kingdom), and a Wellcome Trust collaborative award 201531/Z/16/Z. The funders had no role in the design of the study, or in the analysis, and interpretation of data or writing of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

AOBR and JRF designed the concept of this article. BC, ACP, APA, MHF, AOBR, JRF, and TAOM contributed to the literature review and writing of the manuscript. LSR contributed to figure generation. All authors read and approved the final version to be published.

Corresponding authors

Correspondence to J. Ross Fitzgerald or Andrea de Oliveira Barros Ribon.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Nor applicable.

Competing interests

Not applicable.

Additional information

Publisher’s Note

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

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

Campos, B., Pickering, A.C., Rocha, L.S. et al. Diversity and pathogenesis of Staphylococcus aureus from bovine mastitis: current understanding and future perspectives. BMC Vet Res 18, 115 (2022). https://doi.org/10.1186/s12917-022-03197-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12917-022-03197-5

Keywords