Hostname: page-component-76fb5796d-qxdb6 Total loading time: 0 Render date: 2024-04-26T05:42:12.174Z Has data issue: false hasContentIssue false

Role of staphylococci in mastitis in sheep

Published online by Cambridge University Press:  19 August 2019

Natalia G. C. Vasileiou
Affiliation:
Veterinary Faculty, University of Thessaly, 43100 Karditsa, Greece
Dimitris C. Chatzopoulos
Affiliation:
Veterinary Faculty, University of Thessaly, 43100 Karditsa, Greece
Stela Sarrou
Affiliation:
Veterinary Faculty, University of Thessaly, 43100 Karditsa, Greece University Hospital of Larissa, 41110 Larissa, Greece
Ilektra A. Fragkou
Affiliation:
Veterinary Faculty, University of Thessaly, 43100 Karditsa, Greece
Angeliki I. Katsafadou
Affiliation:
Veterinary Faculty, University of Thessaly, 43100 Karditsa, Greece
Vasia S. Mavrogianni
Affiliation:
Veterinary Faculty, University of Thessaly, 43100 Karditsa, Greece
Efthimia Petinaki
Affiliation:
University Hospital of Larissa, 41110 Larissa, Greece
George C. Fthenakis*
Affiliation:
Veterinary Faculty, University of Thessaly, 43100 Karditsa, Greece
*
Author for correspondence: George C. Fthenakis, Email: gcf@vet.uth.gr
Rights & Permissions [Opens in a new window]

Abstract

Staphylococci have been isolated from various sites of the body of healthy sheep, as well as from many infections of those animals, the main one being mastitis. The objective of this review is to appraise the importance and significance of staphylococci in causing mastitis in ewes. The review includes a brief classification and taxonomy of staphylococci and describes the procedures for their isolation and identification, as well as their virulence determinants and the mechanisms of resistance to antibacterial agents. Various staphylococcal species have been implicated in staphylococcal mastitis and the characteristics of isolates are discussed with regards to potential virulence factors. Staphylococcal mastitis is explicitly described, with reference to sources of infection, the course of the disease and the relevant control measures. Finally, the potential significance of staphylococci present in ewes’ milk for public health is discussed briefly.

Type
Review Article
Copyright
Copyright © Hannah Dairy Research Foundation 2019 

Staphylococcal mastitis in sheep was first studied towards the end of the 19th century by the French veterinarian and microbiologist Edmund Nocard, who described the experience of a practicing veterinarian of the time as follows: ‘This disease is rightly considered to be the plague of the cheese-making flocks; it is not rare to see one tenth of the animals affected by the terrible evil; those, which recover, are invariably lost for milk-production. All the treatments have been tried equally unsuccessfully; this, what the shepherds have found the best to save the animal, is to cut open the udder in different directions from the start of the disease and to treat the wounds with any detergent lotion. The majority of veterinarians of the country consider this disease as a simple mastitis, caused by the milk engorgement and the knocks given on the mammary gland by the milkers; but the owners refuse to believe in these possible causes of the disease and the majority of them thinks that this mastitis is related to anthrax. This opinion should be rejected, because the animals never exhibit, during their life or after their death, the signs or lesions of anthrax; microscopic examination of the blood has never allowed me to see bacteria …’ (Nocard, Reference Nocard1887).

Nocard (Reference Nocard1887) described staphylococcal mastitis from pathological and microbiological examinations. He isolated the causative microorganism from the milk of an affected ewe and described its morphological, cultural and biochemical characteristics. Also, he studied the effects of experimental intramammary inoculation of mammary secretion from an affected mammary gland, by means of which he successfully reproduced a pathological condition similar to the natural disease. Thereafter, Bridre (Reference Bridre1907) undertook a systematic investigation of the disease in dairy flocks in France. He found that the incidence of the disease was approximately 5%, with a mortality of 20%. Further, he attempted to protect sheep by means of immunisation.

In general, the onset and outcome of staphylococcal infections are dependent on the combination of the virulence of the invading isolate and the defence abilities of the host. Protection of a host against staphylococci is to a large degree dependent upon (a) the integrity of the skin and mucous membranes, which form an important barrier to entrance of staphylococci into the body, and (b) the number and functionality of leucocytes, which are important for phagocytosing and destroying the invading bacteria (Murray et al., Reference Murray, Pearson, Coombs, Flexman, Golledge, Speers, Dyer, McLellan, Reilly, Bell, Bowen and Christiansen2008). Moreover, recent work has also presented a role for anti-staphylococcal antibodies, which seem to clear staphylococcal isolates more effectively (Vasileiou, Reference Vasileiou2019, Vasileiou et al., Reference Vasileiou, Chatzopoulos, Cripps, Ioannidi, Gougoulis, CHouzouris, Lianou, Calvo Gonzalez-Valerio, Guix Vallverdu, Argyros, Cesio, Font, Mavrogianni, Petinaki and Fthenakis2019a).

Staphylococcal infections are usually preceded by colonisation and are characterised by intense tissue inflammation. They occur when the bacteria enter through skin or mucous membrane breakdowns (e.g. injuries, ulcerations, surgical incisions). The invading bacteria multiply locally and produce enzymes and toxins, leading in tissue destruction, influx of polymorphonuclear cells, severe tissue damage and abscess formation, which characterise the staphylococcal infections (Lowy, Reference Lowy1998). By means of this mechanism and in accord with the characteristics of staphylococci, local infections occur initially at the point of entry. Thereafter, more severe infections, consequently to bacterial dissemination in tissues or haematogenously can occur in other areas of the body of the host.

Staphylococci have been isolated from various sites of the body of healthy sheep (online Supplementary Table S1). In sheep, the most important problem caused by staphylococci is mastitis, clinical or subclinical. Moreover, the bacteria have been implicated also in various other disorders (online Supplementary Table S1), the significance of which varies. Some of these occur frequently, e.g. impetigo or abscesses; some sporadically, e.g. vaginal infections or abortion; whilst others have been described only experimentally, e.g. osteomyelitis or rhinosinusitis.

In dairy sheep flocks, mammary infections have an obvious financial significance (Selvaggi et al., Reference Selvaggi, D’ Alessandro and Dario2017), due to the reduction in milk yield, the downgrading of milk quality and the rejection of milk consequently to antibiotic administration. Mammary infections are important also in meat production flocks, as reduced milk yield of ewes has been shown to lead to suboptimal growth of their lambs (Fthenakis and Jones, Reference Fthenakis and Jones1990a). Other costs associated with the disease include those for replacement animals and the relevant veterinary expenses. Mastitis is also a great welfare concern (European Food Safety Authority, 2014).

The objective of this review is to appraise the importance and significance of staphylococci in mastitis in ewes. Although staphylococci are important as causal agents for the disease, there is a lack of a systematic review of relevant knowledge.

Methodology

The review includes primarily references published in journals cited in the Web of Knowledge database (http://www.webofknowledge.com); papers published in these journals have been refereed. Various search terms have been employed to identify relevant publications (e.g. ‘sheep’, ‘goat*’, ‘mastitis’, ‘somatic cell count*’, ‘milk’, ‘staphylococcus’, ‘teat’, ‘carriage’, ‘carrier’). Subsequently, the full papers have been retrieved through the websites of the respective journals. Citations of other material are scarce; these include a few references to book chapters and doctoral theses (i.e. material that has been reviewed) and one reference to edited conference material. References cited in the review appear at the end of the review, and additional supporting references are included in the online Supplementary File.

The microorganism

Classification – taxonomy

Taxonomically, the genus Staphylococcus (S.) is classified in the bacterial family Staphylococcaceae (order: Bacillales, class: Bacilli, phylum: Firmicutes, domain: Bacteria). Currently, the genus includes more than 50 species, with many having subspecies. Apart from speciation, various other taxonomic schemes have been proposed for staphylococci, which are based on phenotypic characteristics or molecular findings.

A well-known approach refers to the reaction of staphylococcal species to the coagulase test, which identifies whether a staphylococcal isolate produces the exoenzyme coagulase: staphylococcal species are classified as coagulase-positive or coagulase-negative, with a third class, coagulase-variable, having been established recently. Another approach refers to natural susceptibility of the species to novobiocin: staphylococcal species are classified as novobiocin-sensitive or novobiocin-resistant. Further, clustering into groups based on results of 16s rRNA sequencing has also been performed and 11 groups have been distinguished (Table 1).

Table 1. Clustering of staphylococcal species into 11 groups based on results of 16s rRNA sequencing

Note. Reports regarding other species, e.g. S. leei (coagulase +ve), S. lyticans, S. pseudolugdunensis, have also been published, but their valid taxonomic status has not yet been confirmed.

Source: Euzeby (Reference Euzeby1997).

Isolation and identification

The procedure of isolation of staphylococci starts with culture of the clinical samples, which for mastitis are, usually, milk samples collected from ewes under examination or from the bulk milk tank. Less often, other types of samples may need to be processed, e.g. swabs from mammary lesions or udder skin, or material from the teat duct or mammary tissue samples (Fragkou et al., Reference Fragkou, Boscos and Fthenakis2014).

Enrichment of samples is not necessary. They can be inoculated directly onto conventional (e.g. blood agar) or selective (e.g. Chapman's medium) media. The plates should be incubated at 35–37 °C for up to 48 h (Fragkou et al., Reference Fragkou, Boscos and Fthenakis2014, Vasileiou et al., Reference Vasileiou, Cripps, Ioannidi, Chatzopoulos, Gougoulis, Sarrou, Orfanou, Politis, Calvo Gonzalez-Valerio, Argyros, Mavrogianni, Petinaki and Fthenakis2018b). If no apparent growth has occurred the media can be reincubated for a further 24 h. Due to the tolerance of staphylococci in increased salt concentrations (as high as 10%), mannitol salt agar is an appropriate selective medium (e.g. Chapman's medium) for growth and isolation. Chromogenic agar can be used for the isolation of S. aureus (Ariza-Miguel et al., Reference Ariza-Miguel, Oniciuc, Sanz, Fernandez-Natal, Hernandez and Rodriguez-Lazaro2015). False negative results may always occur with culture technologies. A prolonged transportation time or inappropriate maintenance of samples can trigger bacterial survival mechanisms, e.g. slime production and biofilm formation. In such cases, the bacteria do not produce obvious colonies when transferred onto agar plates (Skrlin, Reference Skrlin2016). Further, in long-standing infections, staphylococci may not grow on solid media (Ehrlich et al., Reference Ehrlich, Demeo, Palmer, Sauber, Altman, Altman, Sotereanos, Conti, Baratz, Maale, Hu, Post, Nistico, Kreft, Hall-Stoodley, Costerton, Stoodley, Ehrlich, DeMeo, Costerton and Winkler2014).

Evaluation of the appearance of colonies is the first step in identification of staphylococci. Staphylococcal species usually form distinctive colonies on sheep blood agar, being smooth and butyrous, with a low convex profile and an entire edge (Winn and Koneman, Reference Winn and Koneman2006). Most Staphylococcus aureus isolates are pale light yellow colonies, due to staphyloxanthin, a carotenoid pigment with a notable role in pathogenicity of the bacteria (Lan et al., Reference Lan, Cheng, Dunman, Missiakas and He2010). Other species, most often, produce white-coloured colonies.

Microscopically, Gram-staining confirms a Gram-positive microorganism. Staphylococcal colonies are differentiated from other Gram-positive bacteria (e.g. streptococci, enterococci) by the catalase test, which detects cytochrome oxidase enzymes, staphylococci show a positive reaction in the test. This is followed by the coagulase test, which serves to distinguish coagulase-positive isolates (S. anaerobius, S. aureus, S. delphini, S. hyicus, S. intermedius, S. lutrae, S. pseudintermedius and S. schleiferi subsp. coagulans).

Thereafter, identification to species level can be by commercial assays which use modified carbohydrate fermentation tests or adaptations of standard bacteriological identification biochemical tests (Donay et al., Reference Donay, Mathieu, Fernandes, Pregermain, Bruel, Wargnier, Casin, Weill, Lagrange and Herrmann2004), or by molecular identification techniques, e.g. the Polymerase Chain Reaction (PCR) or Matrix-Assisted Laser Desorption/Ionisation-Time of Flight (MALDI-TOF). The latter analyses the protein profile of bacterial cells (Katsafadou et al., Reference Katsafadou, Tsangaris, Billinis and Fthenakis2015).

MALDI-TOF technology allows a greater number of bacterial isolates to be rapidly and accurately identified to species level (Cameron et al., Reference Cameron, Perry, Middleton, Chaffer, Lewis and Keefe2018, Hulland et al., Reference Hulland, Dufour and Munoz2018). This is particularly relevant for coagulase-negative staphylococci, the identification of which to species level is often omitted and thus these organisms are cumulatively reported to the genus level only.

Virulence factors

Pathogenicity of staphylococcal infections is based on the virulence factors of the bacteria that allow them to survive within the host and cause relevant damage. The bacteria carry a wide array of potential virulence factors (Table 2), being capable of (i) adhering to host tissues, (ii) avoiding, overcoming or invading the host immune system and (iii) releasing harmful toxins or enzymes (Sahuquillo Arce et al., Reference Sahuquillo Arce, Yarad Auad, Hernández Cabezas and Méndez-Vilas2013). During infections, the bacteria produce the respective virulence determinants in a sequence tightly coordinated by relevant regulatory systems. The expression of both virulence factors and regulatory mechanisms is controlled by specific virulence genes. Factors contributing to the pathogenicity of staphylococci can be classified as (i) bacterial cell surface components (adherence factors) and (ii) secreted variants.

Table 2. Virulence factors involved in the pathogenetic role of a typical S. aureus isolate

Adherence factors include various proteins which mainly act during the early phase of infection. Their principal function is to facilitate attachment of the bacteria to the host cell surface, simultaneously leading to the cascade host immune response evasion (Foster and Höök, Reference Foster and Höök1998). Various capsular polysaccharides, located on the bacterial cell wall, are involved in the inhibition of phagocytosis by neutrophils, while teichoic acids are implicated in bacterial adherence to the mucosal surface. Most of these are microbial surface components recognising adhesive matrix molecules including fibrinogen, fibronectin and collagens. Staphylococcal protein A (SpA), fibronectin-binding protein A and fibronectin-binding protein B (FnbpA and FnbpB), collagen-binding protein and clumping factor A and B proteins have also been determined to play a keynote role in the virulence of staphylococci (Palmqvist et al., Reference Palmqvist, Foster, Tarkowski and Josefsson2002, Heying et al., Reference Heying, van de Gevel, Que, Moreillon and Beekhuizen2007).

Slime production by staphylococcal isolates contributes to biofilm formation by these bacteria and plays an important role in the pathogenesis of staphylococcal infections and mastitis specifically (Schönborn et al., Reference Schönborn, Wente, Paduch and Krömker2017), particularly at the early stage of the infection, when the bacteria adhere to the mammary epithelial cells. Genes icaA, icaB, icaC, icaD are responsible for production of icaADBC-encoded polysaccharide intercellular adhesion and ica-independent chemically diverse slime (Cramton et al., Reference Cramton, Gerke, Schnell, Nichols and Götz1999, Reference Cramton, Ulrich, Götz and Doring2001). More specifically, the icaA gene product is a transmembrane protein which, for optimal action, requires presence of the icaD gene product. The icaC gene encodes a product contributing to chain formulation of N-acetyl-glucosamine oligomers produced by the icaAD combination (Gerke et al., Reference Gerke, Kraft, Sussmuth, Schweitzer and Götz1998); this product is also considered to be involved in translocation of the polysaccharide on cell surface (Gerke et al., Reference Gerke, Kraft, Sussmuth, Schweitzer and Götz1998). Τhe icaB-encoded protein is considered responsible for the bacterial evasion process from the phagocytosis process (Cerca et al., Reference Cerca, Jefferson, Maira-Litrán, Pier, Kelly-Quintos, Goldmann, Azeredo and Pier2007). Moreover, the clfa gene encodes a surface protein, demonstrated as clumping factor A, which has a crucial role in bacterial initial attachment and evasion of host immune responses (Stutz et al., Reference Stutz, Stephan and Tasara2011). The bap gene also encodes an important surface protein, termed ‘biofilm associated protein’; beyond its contribution to initial bacterial attachment, it has also been considered that this protein is capable to induce a polysaccharide intercellular adhesion/poly-N-acetyl glucosamine-independent slime production process, especially on abiotic surfaces (Latasa et al., Reference Latasa, Solano, Penadés and Lasa2006). Finally, the eno gene encodes a laminin-binding surface protein (Carneiro et al., Reference Carneiro, Postol, Nomizo, Reis and Brentani2004).

Secreted virulence factors are mainly present during the late phase of infection and usually have a more distinct role in microbial pathogenicity (Otto, Reference Otto2013). Based on their principal activity, secreted virulence determinants are further classified in four categories: (i) super-antigens, (ii) cytolytic toxins, (iii) exoenzymes and (iv) miscellaneous proteins. Tissue shock syndrome toxin-1 (TSST-1) and enterotoxins are the most prominent superantigens, usually causing clinical conditions of increased severity. Cytolytic toxins (i.e. α-haemolysin, β-haemolysin, γ-haemolysin, toxins of the leucocidin family) are capable of forming pores in the host cell wall, causing the osmotic leakage of cell content and, therefore, lysis of the cell; cytolysis provides the required nutrients for further growth of the bacteria. The extracellular enzymes are produced by most staphylococcal isolates and aim to inactivate host antimicrobial mechanisms, allowing bacterial dissemination. These exoenzymes include various lipases, nucleases and proteases (i.e. hyaluronidase, serine, cysteine, staphylokinase). Finally, many other proteins have been shown to have a further impact on the host immune system; staphylococcal complement inhibitor protein, extracellular fibrinogen binding protein, chemotaxis inhibitory protein and formyl peptide receptor-like protein-1 inhibitory protein are the ones most frequently detected (Otto, Reference Otto2013).

Synthesis of these factors occurs during the two growth phases of the bacteria. During the early phase, cell wall-associated factors facilitate tissue attachment and evasion of host defence system, allowing staphylococci to accumulate and, possibly, also form a biofilm. and the late phase, during which bacteria secrete a spectrum of exoproteins including proteinases, haemolysins and super-antigens, whilst, at the same time, cell wall-associated factors are downregulated, leading to enhancement of the biofilm and bacterial dissemination within the mammary gland (Novick, Reference Novick2003). The production of the various virulence factors of the bacteria is controlled by various mechanisms, following the general strategy of the microbial adhesion, invasion and proliferation. The function of regulation systems is in response of bacterial cell density (quorum-sensing) and environmental factors (e.g. nutrient availability, pH, temperature, oxygen tension). Moreover, it is also noteworthy that a virulence determinant may be under the influence of several regulatory systems that act synergistically to ensure the appropriate conditions for bacterial survival (Wang and Muir, Reference Wang and Muir2016).

In vitro studies have shown that regulation mechanisms can be classified into two major groups: (i) the two-component regulatory systems and (ii) the global transcriptional regulators (Cheung et al., Reference Cheung, Bayer, Zhang, Gresham and Xiong2004). At nucleotide level, the two-component regulatory systems include the staphylococcal accessory element (sae locus) and the accessory gene regulator (agr locus). The sae locus codes the expression of several virulence determinants, mainly contributing to bacterial adhesion and host immune evasion. The agr-coded genes seem to promote the expression of bacterial exoproteins (e.g. TSST-1, enterotoxins, serine proteinase), simultaneously reducing the synthesis of cell-wall associated proteins (e.g. SpA, FnbpA, FnbB). This system was initially described in S. aureus, in which four distinct allelic variants, agrA/Β/C/D, have been sequenced. Subsequently, presence of homologous agr-like loci in other staphylococcal species have been detected (Dufour et al., Reference Dufour, Gillet, Bes, Lina, Vandenesch, Floret, Etienne and Richet2002). Most S. aureus isolates have a third significant group of virulence genes regulators, usually referred as sigma factors (σ).

Mechanisms of resistance to antibacterial agents

A major attribute of most staphylococcal species (including S. aureus) is their extended capacity to develop, fairly rapidly, resistance to antimicrobial agents. Nowadays, multi-drug resistant staphylococci (e.g. S. aureus, S. epidermidis, S. intermedius) are commonly recovered from human or animal clinical samples (Mediavilla et al., Reference Mediavilla, Chen, Mathema and Kreiswirth2012).

Antimicrobial tolerance or resistance is linked to the genetic background of each individual isolate and develops through mutations and rearrangements within the staphylococcal genome or by acquisition of resistance determinants. The wide spectrum of staphylococcal genetic variants and the increased antibiotic pressure contribute significantly in antibiotic resistance formation. Most resistance determinants of staphylococcal genetic material are in highly volatile areas (e.g. genomic islands, plasmids) promoting the occurrence of mutations leading to antibiotic resistance (Foster, Reference Foster2017). Further, during co-infections, genetic transfer may also take place and essential resistant components may easily be transferred horizontally. Finally, genetic transfer of resistant genetic elements between bacteria belonging to different species has also been recorded (Haaber et al., Reference Haaber, Penadés and Ingmer2017).

In general, mechanisms of resistance against antibiotics developed by staphylococci are: (i) relevant modulations of cell wall permeability, (ii) enzymatic inactivation of the antimicrobial agent, (iii) modification of the antibiotic target and (iv) activation of relevant bacterial efflux pumps (McCallum et al., Reference McCallum, Berger-Bächi and Senn2010). Depending on specific antimicrobial agents, one or more of these mechanisms may be involved (Pantosti et al., Reference Pantosti, Sanchini and Monaco2007).

β-Lactams and glycopeptides are antimicrobial agents that inhibit the formation of the staphylococcal cell wall. The penicillin-binding protein (PBP) 1 and PBP 2 are the target of β-lactams, which inhibit the function of PBPs and, therefore, the formation of an intact cell wall. To overcome the effects of β-lactams, methicillin-resistant S. aureus (MRSA) isolates produce a fifth additional PBP named PBP 2a, encoded by the mecA gene, which has reduced affinity with β-lactams and remains active in the presence of β-lactams (Lovering et al., Reference Lovering, Gretes, Safadi, Danel, De Castro, Page and Strynadka2012). Glycopeptides (e.g. vancomycin) attach to the dipeptide D-ala/D-Ala and inhibit the function of PBP. S. aureus isolates with intermediate susceptibility to vancomycin have a remarkably modified architecture on their cell wall, lacking most of the crucial targets of glycopeptides (McGuinness et al., Reference McGuinness, Malachowa and DeLeo2017). Regarding resistance to tetracyclines, the main mechanism is associated with the energy-dependent efflux of tetracycline encoded by the tetA(K) and tetA(L) genes (Khosravi et al., Reference Khosravi, Jenabi and Montazeri2017); recent findings have indicated an association between presence of genes encoding for tetracycline-resistance and for biofilm-formation in staphylococcal isolates (Vasileiou et al., Reference Vasileiou, Sarrou, Papagiannitsis, Chatzopoulos, Malli, Mavrogianni, Petinaki and Fthenakis2019b). With regards to acquisition of resistance to aminoglycosides, resistant isolates are capable to release cytoplasmic aminoglycoside modifying enzymes, which inhibit ribosome binding. Finally, resistance to macrolides and to lincosamides is mainly due to ribosomal modification encoded by the erm genes.

Implication of staphylococci in mastitis in ewes

Implication in clinical mastitis

S. aureus is the primary causal agent of clinical mastitis in ewes. The incidence risk is probably less than 7% across a lactation (Bergonier and Berthelot, Reference Bergonier and Berthelot2003, Arsenault et al., Reference Arsenault, Dubreuil, Higgins and Belanger2008). Field reports have indicated that staphylococci have been isolated from up to 70% of cases of clinical mastitis in field investigations in dairy production flocks (Bergonier and Berthelot, Reference Bergonier and Berthelot2003, Mork et al., Reference Mork, Waage, Tollersrud, Kvitle and Sviland2007). In meat production flocks, staphylococci have been reported to be less frequent, but still responsible for up to 40% of cases of the disease (Arsenault et al., Reference Arsenault, Dubreuil, Higgins and Belanger2008). Other coagulase-positive staphylococcal species associated with clinical mastitis in ewes include S. anaerobius, S. hyicus, S. intermedius and S. schleiferi (Table 3).

Table 3. Selected references regarding implication of non-S. aureus staphylococcal strains in mastitis or mammary carriage in ewes

Coagulase-negative staphylococci have been isolated from cases of clinical mastitis, although much less frequently than S. aureus. These include S. epidermidis, S. saprophyticus, S. simulans, S. xylosus and S. warneri (Table 3). Further, Fthenakis and Jones (Reference Fthenakis and Jones1990b) have reported experimentally induced clinical mastitis caused by S. chromogenes.

Implication in subclinical mastitis

Subclinical mastitis is more common than clinical disease. Criteria employed for definition of subclinical mastitis are important in determining its prevalence; for example, in a field investigation, in which strict criteria had been used, prevalence of subclinical mastitis was 26% (Vasileiou et al., Reference Vasileiou, Cripps, Ioannidi, Chatzopoulos, Gougoulis, Sarrou, Orfanou, Politis, Calvo Gonzalez-Valerio, Argyros, Mavrogianni, Petinaki and Fthenakis2018b, Reference Vasileiou, Gougoulis, Riggio, Ioannidi, Chatzopoulos, Mavrogianni, Petinaki and Fthenakis2018c), whilst in other field studies, with less strict criteria, prevalence was found to be as high as 40%, although the accuracy of detection of cases has been questioned (Vasileiou et al., Reference Vasileiou, Cripps, Ioannidi, Chatzopoulos, Gougoulis, Sarrou, Orfanou, Politis, Calvo Gonzalez-Valerio, Argyros, Mavrogianni, Petinaki and Fthenakis2018b). It is also noteworthy to cite a report of epidemic occurrence of subclinical mastitis in a flock, where prevalence of the disease has been reported to be 94%, with all cases caused by coagulase-negative staphylococci (Fthenakis et al., Reference Fthenakis, Leontides, Skoufos, Taitzoglou and Tzora2004).

A clear consensus exists in the literature that coagulase-negative staphylococcal species are the primary aetiological agents of subclinical mastitis and can constitute up to 70% of bacterial isolates from cases of subclinical disease (Gelasakis et al., Reference Gelasakis, Mavrogianni, Petridis, Vasileiou and Fthenakis2015). The more frequent staphylococcal species recovered from cases of subclinical mastitis were S. chromogenes, S. epidermidis, S. simulans and S. xylosus. Other species recovered as aetiological agents of the disease less often include S. auricularis, S. capitis, S. caprae, S. cohnii, S. equorum, S. haemolyticus, S. hominis, S. lentus, S. saprophyticus, S. sciuri, S. schleiferi, S. warneri (Table 3).

S. aureus has also been recovered as the causative agent of subclinical mastitis, but much less frequently than from cases of clinical disease (Vasileiou et al., Reference Vasileiou, Cripps, Ioannidi, Chatzopoulos, Gougoulis, Sarrou, Orfanou, Politis, Calvo Gonzalez-Valerio, Argyros, Mavrogianni, Petinaki and Fthenakis2018b). Sporadically, S. aureus may account for as many as 40% of isolates from cases of clinical mastitis (Al-Majali and Jawabreh, Reference Al-Majali and Jawabreh2003).

Mammary carriage

The term mammary ‘carriage’ (or ‘carrier state’) (Verhoeven et al., Reference Verhoeven, Gagnaire, Botelho-Nevers, Grattard, Carricajo, Lucht, Pozzetto and Berthelot2014) describes the presence of bacteria in the udder with no increased somatic cell numbers (i.e. in the absence of inflammation). The term refers to bacterial flora present in the teat duct or teat cistern (Fragkou et al., Reference Fragkou, Mavrogianni, Cripps, Gougoulis and Fthenakis2007, Mavrogianni et al., Reference Mavrogianni, Cripps and Fthenakis2007). By using culture-independent methods of microbial identification (e.g. MALDI-TOF), recent work performed in cows has suggested the existence of a microbiota in the teat and the lactiferous ducts; a possible source of these bacteria can be the intestine, as it has been found that bacteria from the gut microbiota reach the mammary gland within leucocytes (e.g. dendritic cells, macrophages, lymphocytes) (Rainard, Reference Rainard2017). Vasileiou et al. (Reference Vasileiou, Cripps, Ioannidi, Chatzopoulos, Gougoulis, Sarrou, Orfanou, Politis, Calvo Gonzalez-Valerio, Argyros, Mavrogianni, Petinaki and Fthenakis2018b) have indicated that the prevalence of staphylococcal mammary carriage in ewes was 6.5%. The significance of mammary carriage is that the microorganisms might become pathogenic under the effect of various factors, which decrease defensive efficacy of hosts or promote pathogenicity of bacteria. In a previous experimental study, Fragkou et al. (Reference Fragkou, Mavrogianni, Cripps, Gougoulis and Fthenakis2007) have shown that, under the effect of factors reducing efficacy of teat defences, staphylococcal carriage could multiply, ascend to the mammary parenchyma and, ultimately, cause clinical mastitis.

Cases of false positive mammary carriage refer to isolation of bacteria at the very early stage of infection before an inflammatory reaction would be elicited (Fthenakis and Jones, Reference Fthenakis and Jones1990b), as well as in cases of contamination of milk samples during collection. Nevertheless, with experienced staff and strict maintenance of aseptic sampling, contamination should be minimal; for example, Rovai et al. (Reference Rovai, Caja, Salama, Jubert, Lázaro, Lázaro and Leitner2014), in a field investigation, found that only 2% of milk samples from ewes were contaminated.

Characteristics of isolates with regards to potential virulence factors

Typing of S. aureus from cases of clinical mastitis has produced varying results regarding the presence of agr locus. Vautor et al. (Reference Vautor, Carsenti-Dellamonica, Sabah, Mancini, Pepin and Dellamonica2007) indicated that 70% of isolates belonged to the agr group III and 13% to the agr group I. In contrast, de Almeida et al. (Reference de Almeida, de Almeida, de Mendonça and Mamizuka2013) indicated that isolates from clinical mastitis belonged mostly to the agr group I (38%) and less often to the agr group II (19%), whilst isolates from subclinical mastitis were equally distributed to these two groups (38% in each). Finally, Bar-Gal et al. (Reference Bar-Gal, Blum, Hadas, Ehricht, Monecke and Leitner2015) reported that all S. aureus isolates from mastitis, clinical or subclinical, belonged to the agr group I.

Further, there is disagreement regarding the significance of biofilm-forming staphylococci from cases of mastitis in ewes varying from 0% (Azara et al., Reference Azara, Longheu, Sanna and Tola2017) to 26% (Vautor et al., Reference Vautor, Carsenti-Dellamonica, Sabah, Mancini, Pepin and Dellamonica2007) to 40–43% (Ergun et al., Reference Ergun, Aslantas, Kirecci, Ozturk, Ceylan and Boyar2012) to 91–98% (Tel et al., Reference Tel, Aslantas, Keskin, Yilmaz and Demir2012) of relevant isolates found to form biofilms. However, phenotypic results of biofilm formation depend upon the criteria employed for assessment and, moreover, isolates may not always express a biofilm-forming phenotype, despite carrying genes associated with slime production and biofilm-formation (Vasileiou et al., Reference Vasileiou, Chatzopoulos, Gougoulis, Sarrou, Katsafadou, Spyrou, Mavrogianni, Petinaki and Fthenakis2018a). The prevalence of subclinical mastitis caused specifically by biofilm-forming staphylococci (independently of species) was found to be 15.5%, with hand-milking recognised as the most important risk factor (Vasileiou et al., Reference Vasileiou, Chatzopoulos, Gougoulis, Sarrou, Katsafadou, Spyrou, Mavrogianni, Petinaki and Fthenakis2018a).

Examination of staphylococci by Multi Locus Sequence Typing also revealed varying and inconsistent findings. Porrero et al. (Reference Porrero, Hasman, Vela, Fernandez-Garayzabal, Dominguez and Aarestrup2012) examined S. aureus strains from clinical mastitis and found that they belonged to types ST9, ST133, ST1739 and ST2011. de Almeida et al. (Reference de Almeida, de Almeida, de Mendonça and Mamizuka2013) reported that S. aureus strains from clinical mastitis belonged to types ST750, ST1728, ST1729 and ST1730, whilst those from subclinical mastitis to types ST750, ST1728 and ST1729. Finally, Bar-Gal et al. (Reference Bar-Gal, Blum, Hadas, Ehricht, Monecke and Leitner2015) reported that S. aureus strains from clinical or subclinical mastitis belonged to types ST133 and ST522. In general, S. aureus ST133 is most often isolated from samples of sheep origin (McMillan et al., Reference McMillan, Moore, McAuley, Fegan and Fox2016).

In studies which had investigated production of virulence factors by staphylococci (S. aureus or coagulase-negative isolates) from cases of mastitis, various factors or genes encoding for shock syndrome toxin-1 (Orden et al., Reference Orden, Goyache, Hernandez, Domenech, Suarez and Gomez Lucia1992b, Scherrer et al., Reference Scherrer, Corti, Muehlherr, Zweifel and Stephen2004), leucocidin (Burriel and Dagnall, Reference Burriel and Dagnall1997), Panton-Valentine toxin (Unal and Cinar, Reference Unal and Cinar2012), exfoliative toxins (Mariutti et al., Reference Mariutti, Souza, Ullah, Caruso, de Moraes, Zanphorlin, Tartaglia, Seyffert, Azevedo, Le Loir, Murakami and Arni2015), haemolysins (Azara et al., Reference Azara, Longheu, Sanna and Tola2017) and autolysin (Azara et al., Reference Azara, Longheu, Sanna and Tola2017) have been detected. The presence of these factors was, in general, more prevalent in S. aureus than in coagulase-negative isolates, which can explain the increased pathogenicity of the former.

Resistance to antibacterial agents of isolates from cases of mastitis in ewes

There is one rule for the effective treatment of mastitis: the combination of speed and efficacy. As soon as mastitis is diagnosed, effective antimicrobial agents should be administered (Mavrogianni et al., Reference Mavrogianni, Menzies, Fragkou and Fthenakis2011). Ideally, and to preserve susceptibility of pathogens to available drugs, treatment should be a narrow spectrum antibiotic known to be effective against staphylococci.

Recent evidence from around Europe does not indicate significant problems of resistance to antibiotics commonly used for cases of mastitis in sheep. Vautor et al. (Reference Vautor, Carsenti-Dellamonica, Sabah, Mancini, Pepin and Dellamonica2007) reported only sporadic resistance in S. aureus isolated in France. Onni et al. (Reference Onni, Sanna, Larsen and Tola2011), in Italy, also found limited resistance in S. epidermidis, except to penicillin, for which the resistance rate was 38%. Similar results have been observed in Turkey, where in coagulase-negative isolates from subclinical mastitis only resistance to β-lactams was noteworthy (43%), whilst there was much smaller frequency of resistance to tetracycline (11%) and even less to other agents (Ergun et al., Reference Ergun, Aslantas, Kirecci, Ozturk, Ceylan and Boyar2012). Further work in Turkey corroborated those findings, the rate of resistance to penicillin was 27% and to tetracycline 8% (Unal et al., Reference Unal, Askar, Macun, Sakarya, Altun and Yildirim2012). Martins et al. (Reference Martins, Faccioli, Bonesso, Fernandes, Oliveira, Dantas, Zafalon and Cunha2017) published similar results; 17% of isolates were resistant to penicillin and 11% to tetracycline. Finally, evidence from Greece was consistent with the above, as the frequency of resistant isolates was <25% for all the antimicrobial agents evaluated (Vasileiou et al., Reference Vasileiou, Sarrou, Papagiannitsis, Chatzopoulos, Malli, Mavrogianni, Petinaki and Fthenakis2019b). Different findings have been reported by Azara et al. (Reference Azara, Longheu, Sanna and Tola2017), who found greater resistance to tetracycline (50%) of S. aureus from clinical mastitis. In contrast to the above results, in Brazil, Franca et al. (Reference Franca, Peixoto, Cavalcante, Melo, Oliveira, Veschi, Mota and Costa2012) determined a higher resistance to amoxicillin, erythromycin, lincomycin, streptomycin and tetracycline (>35% of staphylococcal isolates tested).

Whilst the above results are indicative of the possibility for effective treatment of clinical mastitis, in vitro results of antimicrobial susceptibility do not always correspond with results of in vivo administration of an antibacterial agent. Various reasons may account for this discrepancy, e.g. pharmacokinetic limitations during clinical application, inappropriate treatment regime or iatrogenic infection occurring during treatment (Mavrogianni et al., Reference Mavrogianni, Menzies, Fragkou and Fthenakis2011).

Staphylococcal mastitis in sheep

Sources of infection

Staphylococci usually disseminate into the mammary gland from the hands of milkers (Marco Melero, Reference Marco Melero1994, Vasileiou et al., Reference Vasileiou, Chatzopoulos, Gougoulis, Sarrou, Katsafadou, Spyrou, Mavrogianni, Petinaki and Fthenakis2018a). The use of bare hands during milking contributes to transfer of bacteria to the teat, when the duct is open.

Other potential sources of bacteria include the nasopharynx of sucking lambs (Gougoulis et al., Reference Gougoulis, Kyriazakis, Tzora, Taitzoglou, Skoufos and Fthenakis2008), staphylococci already present in the teat duct (Fragkou et al., Reference Fragkou, Mavrogianni, Cripps, Gougoulis and Fthenakis2007) and staphylococci present on the udder skin (Mavrogianni et al., Reference Mavrogianni, Cripps and Fthenakis2007). Albenzio et al. (Reference Albenzio, Taibi, Caroprese, De Rosa, Muscio and Sevi2003) considered that lambs' mouths and milkers' hands were the major sources of ewe udder and milk contamination, whilst few staphylococci and streptococci were isolated from the teat cups. During suckling, the teat orifice comes into contact with the wall of the buccal cavity of lambs. As Laukova and Marounek (Reference Laukova and Marounek1992) have isolated staphylococci form the upper alimentary tract of lambs, it is evident that the bacteria can transmit to the teat during sucking As the lower part of the teat comes into contact with the pharynx of the lamb (Titchen, Reference Titchen1977) the bacteria subsequently enter the duct; perhaps; the tongue of the lamb may ‘push’ the bacteria upwards into the duct. Isolation of the microorganism after only a short (1 min) sucking activity indicates the speed by which the whole process can take place (Gougoulis et al., Reference Gougoulis, Kyriazakis, Tzora, Taitzoglou, Skoufos and Fthenakis2008). Staphylococci have been found as resident flora of the teat duct of healthy ewes (Fragkou et al., Reference Fragkou, Mavrogianni, Cripps, Gougoulis and Fthenakis2007, Mavrogianni et al., Reference Mavrogianni, Cripps and Fthenakis2007). When the microbial equilibrium is disrupted for any reason, it is possible that pathogenicity of the flora isolates increases, leading to invasion of the mammary parenchyma and disease. Further, physicochemical changes occurring of udder skin, e.g. in response to bad weather conditions or harsh teat disinfectants, may create chapping increasing the susceptibility of the mammary gland. In the epidermis, drying results in a lower lipid content thus less antibacterial fatty acids, bacteriostatic salts and proteins, as well as immunoglobulins (Noble and Somerville, Reference Noble, Somerville and Noble1974). Additionally, the reduced hydration of chapped skin, alters the skin microflora, consequently decreasing resistance to bacterial colonisation (Fox et al., Reference Fox, Nagy, Hillers, Cronrath and Ratkowsky1991). Chapping removes the acid mantle and increases teat surface area, due to excoriations and fissuring, thus providing additional surface for bacterial attachment. All these increase bacterial populations on the udder skin, thus increasing risk of infection of the mammary parenchyma (Mavrogianni et al., Reference Mavrogianni, Cripps and Fthenakis2007).

Giannakopoulos et al. (Reference Giannakopoulos, Vasileiou, Gougoulis, Cripps, Ioannidi, Chatzopoulos, Billinis, Mavrogianni, Petinaki and Fthenakis2019) have modelled for the first time areas of high risk for the development of staphylococcal mastitis, taking into account data regarding prevalence of the disease in the field and environmental and locality conditions, using the ecological niche modelling approach. Whilst the work referred to Greece, there is the possibility to extent the findings to cover other parts of the world.

Course of the disease

The general principles of mammary defences apply for staphylococcal infections. After successful ascent of staphylococci into the teat cistern and the mammary parenchyma, leucocytes constitute the main line of defence against staphylococci. These are developed immediately upon bacterial entry into the mammary gland, 2–4 h after that and 3–4 d later, respectively (Fthenakis and Jones, Reference Fthenakis and Jones1990b, Fragkou et al., Reference Fragkou, Dagleish, Papaioannou, Cripps, Boscos, Ververidis, Orfanou, Solomakos, Finlayson, Govaris, Kyriazakis and Fthenakis2010). An increased diameter of mammary vessels, a greater blood volume of blood passing through the gland increase soon after infection, and an increased permeability of the blood–milk barrier, by various mechanisms (e.g. by modulating claudins at the tight mammary junctions), lead to the transportation of an increased amount of blood constituents into the mammary gland (Barbagianni, Reference Barbagianni2016). Thus, the inflammatory response is developed and sustained. Neutrophils phagocytose bacteria and perform intracellular killing by the rapid release of reactive oxygen species: superoxide radicals and hydrogen peroxide (‘respiratory burst’) (Van Oostveldt et al., Reference Van Oostveldt, Paape, Dosogne and Burvenich2002). Neutrophils also release various antibacterial proteins, e.g. cathelicidins, which become available at the mammary parenchyma (Katsafadou et al., Reference Katsafadou, Tsangaris, Anagnostopoulos, Billinis, Barbagianni, Vasileiou, Spanis, Mavrogianni and Fthenakis2019).

Neutrophils can also kill staphylococci extracellularly, by means of extracellular traps, which have been described for the first time in sheep by Pisanu et al. (Reference Pisanu, Cubeddu, Pagnozzi, Rocca, Cacciotto, Alberti, Marogna, Uzzau and Addis2015). These represent a mesh of DNA, histones, antimicrobial proteins and proteinases that entrap and inactivate the invading staphylococci with no direct contact or engulfment necessary (Phillipson and Kubes, Reference Phillipson and Kubes2011).

Interleucins play an important role in the host response, as they regulate the influx of leucocytes in milk after bacterial invasion. Albenzio et al. (Reference Albenzio, Caroprese, Santillo, Ruggieri, Ciliberti and Sevi2012) have reported the relationships between bacteria, leucocytes and interleucins produced after bacterial invasion (TNF-α, IL-8, IL-1β, IL-10, IL-12). Concentrations of TNF-α and IL-12 were higher after invasion of pathogenic bacteria (including staphylococci); IL-8 was associated with somatic cell counts and pathogenic bacteria. Regarding IL-1β, Albenzio et al. (Reference Albenzio, Caroprese, Santillo, Ruggieri, Ciliberti and Sevi2012) have not observed any differences after intramammary staphylococcal challenge, whilst other authors have (Winter and Colditz, Reference Winter and Colditz2002, Winter et al., Reference Winter, Schilcher, Fuchs and Colditz2003).

Production of virulence factors by the invading staphylococci leads, principally, to destruction of mammary epithelial cells and leucocytes, as well as blood vessels within the mammary parenchyma and consequentially results in intramammary haemorrhages (Fthenakis and Jones, Reference Fthenakis and Jones1990b). Further, biofilm formation allows rapid diffusion of the bacteria, which thus become difficult to control. Occasionally, pathogens that had not been eliminated by the mammary defences pass into the blood circulation. Further, staphylococci can survive in leucocytes, especially if these are not fully functioning, e.g. due to lack of energy in the immediately post-partum period (Fthenakis et al., Reference Fthenakis, Mavrogianni, Gallidis and Papadopoulos2015). This could lead to bacterial accumulations and abscessation within the mammary parenchyma, whence a recrudescence of the infection can ensue (Fthenakis and Jones, Reference Fthenakis and Jones1990b).

The various virulence factors in mastitis-associated staphylococci have not been detected consistently in all isolates tested and there are significant differences even within the various species of the microorganism. This underlines the importance of the host defences in the development and outcome of an intramammary infection. Potential development of clinical mastitis or limitation of the infection to subclinical mastitis depends upon the combination of virulence factors of the invading bacteria and the efficacy of the defence mechanisms of the host. There is now evidence confirming that cellular defences are the determining host factor that may limit a virulent isolate to only cause subclinical mastitis (Barbagianni et al., Reference Barbagianni, Mavrogianni, Katsafadou, Spanos, Tsioli, Galatos, Nakou, Valasi, Gouletsou and Fthenakis2015, Mavrogianni et al., Reference Mavrogianni, Papadopoulos, Gougoulis, Gallidis, Ptochos, Fragkou, Orfanou and Fthenakis2017). Cell defences are non-specific, acting against all invading bacteria, including staphylococci. A factor that can affect efficacy of cell defences is the breed of sheep (Riggio and Portolano, Reference Riggio and Portolano2015). Potentially, subclinical mastitis can convert to clinical, if defences of the host are impaired. Complete recovery requires a fully functioning defence system of the animals, coupled with an effective course of treatment. Other possible outcomes include the development of a long-standing (‘chronic’) infection with periodic flare-ups of clinical disease, the necrosis of mammary parenchyma, leading to partial or extensive tissue slough-off, the formation of intramammary abscesses with presence of staphylococci therein and the recrudescence of disease. The role of humoral defences has not been studied in depth. Nevertheless, in a recent study, anti-staphylococcal antibodies detected after vaccination of ewes have been found to contribute to improved clearance of staphylococci, but not to fully prevent establishment of the infection (Vasileiou et al., Reference Vasileiou, Chatzopoulos, Cripps, Ioannidi, Gougoulis, CHouzouris, Lianou, Calvo Gonzalez-Valerio, Guix Vallverdu, Argyros, Cesio, Font, Mavrogianni, Petinaki and Fthenakis2019a).

Vaccination against the disease

As part of udder health management, various measures can be implemented for prevention of mastitis. Most of these have a broad spectrum of efficacy and are relevant for prevention of staphylococcal mastitis as well. Such measures include various management practices, e.g. application of post-milking teat disinfection and correct use and maintenance of milking systems.

Vaccination is a targeted means for prevention of staphylococcal mastitis. In the past, inactivated vaccines with whole staphylococcal cells and/or toxoids have been developed against mastitis (Watson, Reference Watson1988), offering mainly reduction in severity of clinical signs (not reduction in incidence risk). A vaccine with inactivated S. aureusS. simulans whole cells and S. aureus exopolysaccharide antigens within liposomes reduced the incidence risk of the disease (Amorena et al., Reference Amorena, Baselga and Albizu1994). More recently, Perez et al. (Reference Perez, Prenafeta, Valle, Penades, Rota, Solano, Marco, Grillo, Lasa, Irache, Maira-Litran, Jimenez-Barbero, Costa, Pier, de Andres and Amorena2009) described the induction of antibodies against the poly-N-acetyl β−1,6 glucosamine exopolysaccharide, the main component of the extracellular matrix of staphylococcal biofilms. A vaccine has been produced which includes a S. aureus bacterin containing bacterial cells surrounded by components of the biofilm matrix. The vaccine elicits an exopolysaccharide specific antibody response offering some protection against S. aureus mastitis (Prenafeta et al., Reference Prenafeta, March, Foix, Casals and Costa2010) and has contributed to prevention of slime production by staphylococcal isolates, thus limiting their growth and dissemination within and outside the mammary gland. Vasileiou et al. (Reference Vasileiou, Chatzopoulos, Cripps, Ioannidi, Gougoulis, CHouzouris, Lianou, Calvo Gonzalez-Valerio, Guix Vallverdu, Argyros, Cesio, Font, Mavrogianni, Petinaki and Fthenakis2019a) described the efficacy of this vaccine in reducing the incidence risk of staphylococcal mastitis in an extensive field study, offering protection against S. aureus and coagulase-negative staphylococci. Autogenous staphylococcal vaccines are of limited use in specific flocks and do not have a wide applicability. There are no reports of their efficacy, as they are produced for ‘tailor-made’ administration in flocks with perceived problem of staphylococcal mastitis, without first being evaluated for efficacy.

Public health considerations

Staphylococcal enterotoxins are in a family of over 20 exotoxins, which are related and have a sequence homology (Pinchuk et al., Reference Pinchuk, Beswick and Reyes2010). The enterotoxins can cause significant diseases in people, among these food poisoning and toxic shock syndrome. Enterotoxins are mainly produced by S. aureus, although other staphylococcal species may also show enterotoxigenic properties. Staphylococcal food poisoning is an intoxication occurring consequently to consumption of foods containing sufficient amounts of one (or more) pre-formed enterotoxins (Angeles Argudin et al., Reference Angeles Argudin, Carmen Mendoza and Rosario Rodicio2010). Hence, it becomes evident that if milk collected from sheep was subjected to appropriate thermal processing leading to staphylococcal killing there would be no real danger of enterotoxin poisoning from dairy products from that milk. In contrast, the danger would increase sharply with consumption of unprocessed milk, given the frequency of staphylococcal mammary infections, especially as production of enterotoxin has been detected in S. aureus isolated from intramammary infections (Orden et al., Reference Orden, Cid, Blanco, Quiteria, Gomez Lucia and de la Fuente1992a, Reference Orden, Goyache, Hernandez, Domenech, Suarez and Gomez Lucia1992b) or the farm milk tank (Scherrer et al., Reference Scherrer, Corti, Muehlherr, Zweifel and Stephen2004).

For these same reasons, dairy products have not been considered to be implicated in the dissemination of antimicrobial resistance to consumers of milk. Nevertheless, recent studies have documented that cell-free genetic material from staphylococci resistant to antimicrobial agents, not destroyed during thermal processing of milk, could be transferred to humans (Wang et al., Reference Wang, McEntire, Zhang, Li and Doyle2012, Schwarz et al., Reference Schwarz, Loeffler and Kadlec2017). Given that resistance genes could be incorporated in other bacterial species (e.g. Streptococcus spp., Acinetobacter spp.), which are part of the normal bacterial flora of humans, dissemination of resistance genes can occur, potentially making staphylococci ‘stores’ of resistant genes and dairy products a means for their transfer.

Concluding remarks

Staphylococci are the most frequent mastitis pathogens in ewes. Extensive research has clarified various facets of their pathogenicity for the mammary gland of sheep. Some of that knowledge has been based in relevant work performed in cattle. However, the situation with cattle and sheep differs significantly, not only in terms of differences between the animal species but also between management practices in the respective industries. In relation to ovine mastitis, there are still areas, which have not been fully clarified, whilst further questions have arisen from the findings of recent work.

The identification of a supposed microbiota within the mammary gland has implications in the interpretation of the results of bacteriological testing of milk samples from cases of mastitis; much more work is needed to confirm any, or the absence of a, pathogenic role of bacteria. The microbiota has implications for the immune response of the mammary gland as these bacteria may contribute to a defensive role of the gland.

The recent detection of anti-staphylococcal antibodies in cases of mastitis raises questions regarding their significance in contributing to immunological enhancement of the defence response.

Further, the identification of biofilm-forming staphylococci (independently of species) as causal agents of mastitis, i.e. as an independent disease entity beyond staphylococcal species, raises issues regarding the dynamics of infection by these organisms. Their presence in milking parlours and relevant factors contributing in their dissemination have not been studied.

Finally, the identification, in dairy products, of cell-free genetic material of strains resistant to antimicrobial agents adds another public health concern. Pasteurised milk had previously been considered of low risk for the transfer of resistance genes, but the new findings indicate a further route for dissemination of such genes. This puts further pressure in maintaining udder health and limiting staphylococcal involvement as a mammary pathogen, with a view to minimising public health concerns.

Supplementary material

The supplementary material for this article can be found at https://doi.org/10.1017/S0022029919000591.

Footnotes

*

These authors have contributed equally and their names are listed alphabetically.

References

Albenzio, M, Taibi, L, Caroprese, M, De Rosa, G, Muscio, A and Sevi, A (2003) Immune response, udder health and productive traits of machine milked and suckling ewes. Small Ruminant Research 48, 189200.Google Scholar
Albenzio, M, Caroprese, M, Santillo, A, Ruggieri, D, Ciliberti, MG and Sevi, A (2012) Immune competence of the mammary gland as affected by somatic cell and pathogenic bacteria in ewes with subclinical mastitis. Journal of Dairy Science 95, 38773887.Google Scholar
Al-Majali, AM and Jawabreh, S (2003) Period prevalence and aetiology of subclinical mastitis in Awassi sheep in southern Jordan. Small Ruminant Research 47, 243248.Google Scholar
Amorena, B, Baselga, R and Albizu, I (1994) Use of liposome-immunopotentiated exopolysaccharide as a component of an ovine mastitis staphylococcal vaccine. Vaccine 12, 243249.Google Scholar
Angeles Argudin, M, Carmen Mendoza, M and Rosario Rodicio, M (2010) Food poisoning and Staphylococcus aureus enterotoxins. Toxins 2, 1751–U342.Google Scholar
Ariza-Miguel, J, Oniciuc, EA, Sanz, I, Fernandez-Natal, I, Hernandez, M and Rodriguez-Lazaro, D (2015) Evaluation of two commercially available chromogenic media for confirmation of methicillin-resistant Staphylococcus aureus from human, animal, and food samples. International Journal of Food Microbiology 209, 2628.Google Scholar
Ariznabarreta, A, Gonzalo, C and San Primitivo, F (2002) Microbiological quality and somatic cell count of ewe milk with special reference to staphylococci. Journal of Dairy Science 85, 13701375.Google Scholar
Arsenault, J, Dubreuil, P, Higgins, R and Belanger, D (2008) Risk factors and impacts of clinical and subclinical mastitis in commercial meat-producing sheep flocks in Quebec, Canada. Preventive Veterinary Medicine 87, 373393.Google Scholar
Azara, C, Longheu, G, Sanna, G and Tola, S (2017) Biofilm formation and virulence factor analysis of Staphylococcus aureus isolates collected from ovine mastitis. Journal of Applied Microbiology 123, 372379.Google Scholar
Barbagianni, MS (2016) Experimental Study of Pregnancy Toxaemia in Ewes and its Association with Mastitis in the Post-Partum Period (PhD thesis). University of Thessaly.Google Scholar
Barbagianni, MS, Mavrogianni, VS, Katsafadou, AI, Spanos, AS, Tsioli, V, Galatos, A, Nakou, M, Valasi, I, Gouletsou, PG and Fthenakis, GC (2015) Pregnancy toxaemia as risk factor for development of mastitis in sheep during the immediately post-partum period. Small Ruminant Research 130, 246251.Google Scholar
Bar-Gal, GK, Blum, SE, Hadas, L, Ehricht, R, Monecke, S and Leitner, G (2015) Host-specificity of Staphylococcus aureus causing intramammary infections in dairy animals assessed by genotyping and virulence genes. Veterinary Microbiology 176, 143154.Google Scholar
Bergonier, D and Berthelot, X (2003) New advances in epizootiology and control of ewe mastitis. Livestock Production Science 79, 116.Google Scholar
Berube, BJ and Wardenburg, JB (2013) Staphylococcus aureus α-toxin: nearly a century of intrigue. Toxins (Basel) 6, 11401166.Google Scholar
Bridre, J (1907) La mammite gangreneuse des brebis laitieres: pathogenie et vaccination. Bulletin de la Societe Centrale de Medecine Veterinaire 61, 500506.Google Scholar
Burriel, AR (1997) Resistance of coagulase-negative staphylococci isolated from sheep to various antimicrobial agents. Research in Veterinary Science 63, 189190.Google Scholar
Burriel, AR and Dagnall, GJR (1997) Leukotoxic factors produced by staphylococci of ovine origin. Microbiological Research 152, 247250.Google Scholar
Cameron, M, Perry, J, Middleton, JR, Chaffer, M, Lewis, J and Keefe, GP (2018) Short communication: evaluation of MALDI-TOF mass spectrometry and a custom reference spectra expanded database for the identification of bovine-associated coagulase-negative staphylococci. Journal of Dairy Science 101, 590595.Google Scholar
Carneiro, CR, Postol, E, Nomizo, R, Reis, LF and Brentani, RR (2004) Identification of enolase as a laminin-binding protein on the surface of Staphylococcus aureus. Microbes and Infection 6, 604608.Google Scholar
Cerca, N, Jefferson, KK, Maira-Litrán, T, Pier, DB, Kelly-Quintos, C, Goldmann, DA, Azeredo, J and Pier, GB (2007) Molecular basis for preferential protective efficacy of antibodies directed to the poorly acetylated form of staphylococcal poly-N-acetyl-beta-(1–6)-glucosamine. Infection and Immunity 75, 34063413.Google Scholar
Cheung, AL, Bayer, AS, Zhang, G, Gresham, H and Xiong, YQ (2004) Regulation of virulence determinants in vitro and in vivo in Staphylococcus aureus. FEMS Immunology & Medical Microbiology 40, 19.Google Scholar
Cramton, SE, Gerke, C, Schnell, NF, Nichols, WW and Götz, F (1999) The intercellular adhesion (ica) locus is present in Staphylococcus aureus and is required for biofilm formation. Infection and Immunity 67, 54275433.Google Scholar
Cramton, SE, Ulrich, M, Götz, F and Doring, G (2001) Anaerobic conditions induce expression of polysaccharide intercellular adhesin in Staphylococcus aureus and Staphylococcus epidermidis. Infection and Immunity 69, 40794085.Google Scholar
Cuccuru, C, Meloni, M, Sala, E, Scaccabarozzi, L, Locatelli, C, Moroni, P and Bronzo, V (2011) Effects of intramammary infections on somatic cell score and milk yield in Sarda sheep. New Zealand Veterinary Journal 59, 128131.Google Scholar
de Almeida, LM, de Almeida, MZPRB, de Mendonça, CL and Mamizuka, EM (2013) Comparative analysis of agr groups and virulence genes among subclinical and clinical mastitis Staphylococcus aureus isolates from sheep flocks of the Northeast of Brazil. Brazilian Journal of Microbiology 44, 493498.Google Scholar
de Haas, CJ, Veldkamp, KE, Peschel, A, Weerkamp, F, Van Wamel, WJ, Heezius, EC, Poppelier, MJ, Van Kessel, KP and van Strijp, JA (2004) Chemotaxis inhibitory protein of Staphylococcus aureus, a bacterial antiinflammatory agent. The Journal of Experimental Medicine 199, 687695.Google Scholar
Deinhofer, M and Pernthaner, A (1993) Differentiation of staphylococci from ewe and goat milk samples. Deutsche Tierarztliche Wochenschrift 100, 234236.Google Scholar
de la Fuente, R, Ruiz Santa Quiteria, JA, Cid, D, Domingo, M and Suarez, G (1993) Experimental intramammary infection of ewes with Staphylococcus aureus subsp anaerobius. Research in Veterinary Science 54, 221226.Google Scholar
Donay, JL, Mathieu, D, Fernandes, P, Pregermain, C, Bruel, P, Wargnier, A, Casin, I, Weill, FX, Lagrange, PH and Herrmann, JL (2004) Evaluation of the automated phoenix system for potential routine use in the clinical microbiology laboratory. Journal of Clinical Microbiology 42, 15421546.Google Scholar
Dubin, G (2002) Extracellular proteases of Staphylococcus spp. Biological Chemistry 383, 10751086.Google Scholar
Dufour, P, Gillet, Y, Bes, M, Lina, G, Vandenesch, F, Floret, D, Etienne, J and Richet, H (2002) Community-acquired methicillin-resistant Staphylococcus aureus infections in France: emergence of a single clone that produces Panton-Valentine leucocidin. Clinical Infectious Diseases 35, 819824.Google Scholar
Ehrlich, GD, Demeo, PJ, Palmer, MP, Sauber, TJ, Altman, DS, Altman, G, Sotereanos, N, Conti, SF, Baratz, M, Maale, GE, Hu, FZ, Post, JC, Nistico, L, Kreft, R, Hall-Stoodley, L, Costerton, JW and Stoodley, P (2014) Culture-negative infections in orthopedic surgery. In Ehrlich, G, DeMeo, P, Costerton, J, Winkler, H (eds), Culture Negative Orthopedic Biofilm Infections. Springer Series on Biofilms, vol 7. Springer, Berlin pp. 1727. Springer Series on Biofilms 7.Google Scholar
Eisenbeis, J, Peisker, H, Backes, CS, Bur, S, Hölters, S, Thewes, N, Greiner, M, Junker, C, Schwarz, EC, Hoth, M, Junker, K, Preissner, KT, Jacobs, K, Herrmann, M and Bischoff, M (2017) The extracellular adherence protein (Eap) of Staphylococcus aureus acts as a proliferation and migration repressing factor that alters the cell morphology of keratinocytes. International Journal of Medical Microbiology 307, 116125.Google Scholar
Ergun, Y, Aslantas, O, Dogruer, G, Kirecci, E, Saribay, MK, Ates, CT, Ulku, A and Demir, C (2009) Prevalence and etiology of subclinical mastitis in Awassi dairy ewes in southern Turkey. Turkish Journal of Veterinary & Animal Sciences 33, 477483.Google Scholar
Ergun, Y, Aslantas, O, Kirecci, E, Ozturk, F, Ceylan, A and Boyar, Y (2012) Antimicrobial susceptibility, presence of resistance genes and biofilm formation in coagulase negative staphylococci isolated from subclinical sheep mastitis. Kafkas Universitesi Veteriner Fakultesi Dergisi 18, 449456.Google Scholar
European Food Safety Authority (2014) Scientific opinion on the welfare risks related to the farming of sheep for wool, meat and milk production. EFSA Journal 12, 39334060.Google Scholar
Euzeby, JP (1997) List of bacterial names with standing in nomenclature: a folder available on the Internet. International Journal of Systematic Bacteriology 47, 590592 (list of prokaryotic names with standing in nomenclature, Available at http://www.bacterio.net).Google Scholar
Foster, TJ (2017) Antibiotic resistance in Staphylococcus aureus. Current status and future prospects. FEMS Microbiology Reviews 41, 430444.Google Scholar
Foster, TJ and Höök, M (1998) Surface protein adhesins of Staphylococcus aureus. Trends in Microbiology 6, 484488.Google Scholar
Fox, LK, Nagy, JA, Hillers, JK, Cronrath, JD and Ratkowsky, DA (1991) Effects of postmilking teat treatment on the colonization of Staphylococcus aureus on chapped teat skin. American Journal of Veterinary Research 52, 799802.Google Scholar
Fragkou, IA, Mavrogianni, VS, Cripps, PJ, Gougoulis, DA and Fthenakis, GC (2007) The bacterial flora in the teat duct of ewes can protect against and can cause mastitis. Veterinary Research 38, 525545.Google Scholar
Fragkou, IA, Dagleish, MP, Papaioannou, N, Cripps, PJ, Boscos, CM, Ververidis, HN, Orfanou, DC, Solomakos, N, Finlayson, J, Govaris, A, Kyriazakis, I and Fthenakis, GC (2010) The induction of lymphoid follicle-like structures in the ovine teat duct following experimental infection with Mannheimia haemolytica. Veterinary Journal 184, 194200.Google Scholar
Fragkou, IA, Boscos, CM and Fthenakis, GC (2014) Diagnosis of clinical or subclinical mastitis in ewes. Small Ruminant Research 118, 8692.Google Scholar
Franca, CA, Peixoto, RM, Cavalcante, MB, Melo, NF, Oliveira, CJB, Veschi, JA, Mota, RA and Costa, MM (2012) Antimicrobial resistance of Staphylococcus spp. from small ruminant mastitis in Brazil. Pesquisa Veterinaria Brasileira 32, 747753.Google Scholar
Fthenakis, GC and Jones, JET (1990 a) The effect of experimentally induced subclinical mastitis on the milk yield of ewes and on the growth of lambs. British Veterinary Journal 146, 4349.Google Scholar
Fthenakis, GC and Jones, JET (1990 b) The effect of inoculation of coagulase negative staphylococci into the ovine mammary gland. Journal of Comparative Pathology 102, 211219.Google Scholar
Fthenakis, GC, Marples, RR, Richardson, JF and Jones, JET (1994) Some properties of coagulase-negative staphylococci isolated from cases of ovine mastitis. Epidemiology and Infection 112, 171176.Google Scholar
Fthenakis, GC, Leontides, L, Skoufos, J, Taitzoglou, IA and Tzora, A (2004) Case report: high prevalence rate of ovine mastitis, caused by coagulase-negative staphylococci and predisposed by increased gossypol consumption. Small Ruminant Research 52, 185189.Google Scholar
Fthenakis, GC, Mavrogianni, VS, Gallidis, E and Papadopoulos, E (2015) Interactions between parasitic infections and reproductive efficiency in sheep. Veterinary Parasitology 208, 5666.Google Scholar
Gelasakis, AI, Mavrogianni, VS, Petridis, IG, Vasileiou, NGC and Fthenakis, GC (2015) Mastitis in sheep – the last 10 years and the future of research. Veterinary Microbiology 185, 136146.Google Scholar
Gerke, C, Kraft, A, Sussmuth, R, Schweitzer, O and Götz, F (1998) Characterization of the N-acetyl-glucosaminyl-transferase activity involved in the biosynthesis of the Staphylococcus epidermidis polysaccharide intercellular adhesin. Journal of Biological Chemistry 273, 1858618593.Google Scholar
Giannakopoulos, A, Vasileiou, NGC, Gougoulis, DA, Cripps, PJ, Ioannidi, KS, Chatzopoulos, DC, Billinis, C, Mavrogianni, VS, Petinaki, E and Fthenakis, GC (2019) Use of geographical information system and ecological niche modelling for predicting potential space distribution of subclinical mastitis in ewes. Veterinary Microbiology 228, 119128.Google Scholar
Gougoulis, DA, Kyriazakis, I, Tzora, A, Taitzoglou, IA, Skoufos, J and Fthenakis, GC (2008) Effects of lamb sucking on the bacterial flora of teat duct and mammary gland of ewes. Reproduction in Domestic Animals 43, 2226.Google Scholar
Haaber, J, Penadés, JR and Ingmer, H (2017) Transfer of antibiotic resistance in Staphylococcus aureus. Trends in Microbiology 25, 893905.Google Scholar
Hariharan, H, Donachie, W, Macaldowie, C and Keefe, G (2004) Bacteriology and somatic cell counts in milk samples from ewes on a Scottish farm. Canadian Journal of Veterinary Research 68, 188192.Google Scholar
Heying, R, van de Gevel, J, Que, YA, Moreillon, P and Beekhuizen, H (2007) Fibronectin-binding proteins and clumping factor A in Staphylococcus aureus experimental endocarditis: FnBPA is sufficient to activate human endothelial cells. Thrombosis and Haemostasis 97, 617626.Google Scholar
Hu, C, Xiong, N, Zhang, Y, Rayner, S and Chen, S (2012) Functional characterization of lipase in the pathogenesis of Staphylococcus aureus. Biochemical and Biophysical Research Communications 419, 617620.Google Scholar
Hulland, C, Dufour, S and Munoz, M (2018) Milk Bacteriological Analysis Using MALDI-TOF Technology. New Prague, USA: National Mastitis Council.Google Scholar
Ibberson, CB, Jones, CL, Singh, S, Wise, MC, Hart, ME, Zurawski, DV and Horswill, AR (2014) Staphylococcus aureus hyaluronidase is a CodY-regulated virulence factor. Infection and Immunity 82, 42534264.Google Scholar
Katsafadou, AI, Tsangaris, GT, Billinis, C and Fthenakis, GC (2015) Use of proteomics in the study of microbial diseases of small ruminants. Veterinary Microbiology 181, 2733.Google Scholar
Katsafadou, AI, Tsangaris, GT, Anagnostopoulos, AK, Billinis, C, Barbagianni, MS, Vasileiou, NGC, Spanis, SA, Mavrogianni, and Fthenakis, GC (2019) Differential quantitative proteomics study of experimental Mannheimia haemolytica mastitis in sheep. Journal of Proteomics 205, 103393.Google Scholar
Khosravi, AD, Jenabi, A and Montazeri, EA (2017) Distribution of genes encoding resistance to aminoglycoside modifying enzymes in methicillin-resistant Staphylococcus aureus (MRSA) strains. Kaohsiung Journal of Medical Sciences 33, 587593.Google Scholar
Kiedrowski, ΜR, Crosby, HA, Hernandez, FJ, Malone, CL, McNamara, JO and Horswill, RA (2014) Staphylococcus aureus Nuc2 is a functional, surface-attached extracellular nuclease. PLoS One 9, e95574.Google Scholar
Kirecci, E, Ergun, Y, Dogruer, G and Saribay, MK (2009) Usefulness of the e-test for the determination of the susceptibility of Staphylococcus sp. isolated from milk of sheep and goats with subclinical mastitis to amikacin and amoxicillin-clavulanic acid. Bulletin of the Veterinary Institute in Pulawy 53, 401405.Google Scholar
Ko, YP, Liang, X, Smith, CW, Degen, JL and Höök, M (2011) Binding of Efb from Staphylococcus aureus to fibrinogen blocks neutrophil adherence. Journal of Biological Chemistry 286, 98659874.Google Scholar
Kobayashi, SD and DeLeo, FR (2013) Staphylococcus aureus protein A promotes immune suppression. mBio, e00764–13.Google Scholar
Lacey, KA, Leech, JM, Lalor, SJ, McCormack, N, Geoghegan, JA and McLoughlin, RM (2017) The Staphylococcus aureus cell wall-anchored protein clumping factor A is an important T cell antigen. Infection and Immunity 85, e0054917.Google Scholar
Lan, L, Cheng, A, Dunman, PM, Missiakas, D and He, C (2010) Golden pigment production and virulence gene expression are affected by metabolisms in Staphylococcus aureus. Journal of Bacteriology 192, 30683077.Google Scholar
Lang, S, Livesley, MA, Lambert, PA, Littler, WA and Elliott, TS (2000) Identification of a novel antigen from Staphylococcus epidermidis. FEMS Immunology and Medical Microbiology 29, 213220.Google Scholar
Larsen, HD, Aarestrup, FM and Jensen, NE (2002) Geographical variation in the presence of genes encoding superantigenic exotoxins and beta-hemolysin among Staphylococcus aureus isolated from bovine mastitis in Europe and USA. Veterinary Microbiology 85, 6167.Google Scholar
Latasa, C, Solano, C, Penadés, JR and Lasa, I (2006) Biofilm-associated proteins. Comptes Rendus Biologies 329, 849857.Google Scholar
Laukova, A and Marounek, M (1992) Physiological and biochemical characteristics of staphylococci isolated from the rumen of young calves and lambs. Zentralblatt fur Mikrobiologie 147, 489494.Google Scholar
Leitner, G, Chaffer, M, Caraso, Y, Ezra, E, Kababea, D, Winkler, M, Glickman, A and Saran, A (2003) Udder infection and milk somatic cell count, NAGase activity and milk composition-fat, protein and lactose-in Israeli-Assaf and Awassi sheep. Small Ruminant Research 49, 157164.Google Scholar
Lovering, AL, Gretes, MC, Safadi, SS, Danel, F, De Castro, L, Page, MG and Strynadka, NC (2012) Structural insights into the anti-methicillin-resistant Staphylococcus aureus (MRSA) activity of ceftobiprole. Journal of Biological Chemistry 287, 3209632102.Google Scholar
Lowy, FD (1998) Medical progress – Staphylococcus aureus infections. New England Journal of Medicine 339, 520532.Google Scholar
Madani, A, Garakani, K and Mofrad, RKM (2017) Molecular mechanics of Staphylococcus aureus adhesin, CNA, and the inhibition of bacterial adhesion by stretching collagen. PLoS One 12, e0179601.Google Scholar
Marco Melero, JC (1994) Mastitis in Laxta Breed Sheep: Epidemiology, Diagnosis and Control (Doctoral thesis). University of Zaragoza.Google Scholar
Mariutti, RB, Souza, TACB, Ullah, A, Caruso, IP, de Moraes, FR, Zanphorlin, LM, Tartaglia, NR, Seyffert, N, Azevedo, VA, Le Loir, Y, Murakami, MT and Arni, RK (2015) Crystal structure of Staphylococcus aureus exfoliative toxin D-like protein: structural basis for the high specificity of exfoliative toxins. Biochemical and Biophysical Research Communications 467, 171177.Google Scholar
Martins, KB, Faccioli, PY, Bonesso, MF, Fernandes, S, Oliveira, AA, Dantas, A, Zafalon, LF and Cunha, MDRS (2017) Characteristics of resistance and virulence factors in different species of coagulase-negative staphylococci isolated from milk of healthy sheep and animals with subclinical mastitis. Journal of Dairy Science 100, 21842195.Google Scholar
Mavrogianni, VS, Fthenakis, GC, Burriel, AR, Gouletsou, P, Papaioannou, N and Taitzoglou, IA (2004) Experimentally induced teat stenosis in dairy ewes: clinical, pathological and ultrasonographic features. Journal of Comparative Pathology 130, 7074.Google Scholar
Mavrogianni, VS, Cripps, PJ and Fthenakis, GC (2007) Bacterial flora and risk of infection of the ovine teat duct and mammary gland throughout lactation. Preventive Veterinary Medicine 79, 163173.Google Scholar
Mavrogianni, VS, Menzies, PI, Fragkou, IA and Fthenakis, GC (2011) Principles of mastitis treatment in sheep and goats. Veterinary Clinics of North America Food Animal Practice 27, 115120.Google Scholar
Mavrogianni, VS, Papadopoulos, E, Gougoulis, DA, Gallidis, E, Ptochos, S, Fragkou, IA, Orfanou, DC and Fthenakis, GC (2017) Gastrointestinal trichostrongylosis can predispose ewes to clinical mastitis after experimental mammary infection. Veterinary Parasitology 245, 7177.Google Scholar
McCallum, N, Berger-Bächi, B and Senn, MM (2010) Regulation of antibiotic resistance in Staphylococcus aureus. International Journal of Medical Microbiology 300, 118129.Google Scholar
McGuinness, WA, Malachowa, N and DeLeo, FR (2017) Vancomycin resistance in Staphylococcus aureus. Yale Journal of Biological Medicine 902, 269281.Google Scholar
McMillan, K, Moore, SC, McAuley, CM, Fegan, N and Fox, EM (2016) Characterization of Staphylococcus aureus isolates from raw milk sources in Victoria, Australia. BMC Microbiology 16, 169.Google Scholar
Mediavilla, JR, Chen, L, Mathema, B and Kreiswirth, BN (2012) Global epidemiology of community-associated methicillin resistant Staphylococcus aureus (CA-MRSA). Current Opinion in Microbiology 15, 588595.Google Scholar
Menzies, BE (2003) The role of fibronectin binding proteins in the pathogenesis of Staphylococcus aureus infections. Current Opinion in Infectious Diseases 16, 225229.Google Scholar
Mork, T, Waage, S, Tollersrud, S, Kvitle, B and Sviland, S (2007) Clinical mastitis in ewes; bacteriology, epidemiology and clinical features. Acta Veterinaria Scandinavica 49, 2330.Google Scholar
Murray, RJ, Pearson, JC, Coombs, GW, Flexman, JP, Golledge, CL, Speers, DJ, Dyer, JR, McLellan, DG, Reilly, M, Bell, JM, Bowen, SF and Christiansen, KJ (2008) Outbreak of invasive methicillin-resistant Staphylococcus aureus infection associated with acupuncture and joint injection. Infection Control and Hospital Epidemiology 29, 859865.Google Scholar
Noble, WC and Somerville, DA (1974) Skin as a habitat. In Noble, WC (ed.), Microbiology of Human Skin. Philadelphia: Saunders, pp. 378.Google Scholar
Nocard, E (1887) Note sur la mammite gangreneuse des brebis laitieres. Annales de l’ Institut Pasteur 1, 417428.Google Scholar
Novick, RP (2003) Autoinduction and signal transduction in the regulation of staphylococcal virulence. Molecular Microbiology 48, 14291449.Google Scholar
Onni, T, Sanna, G, Cubeddu, GP, Marogna, G, Lollai, S, Leori, G and Tola, S (2010) Identification of coagulase-negative staphylococci isolated from ovine milk samples by PCR-RFLP of 16S rRNA and gap genes. Veterinary Microbiology 144, 347352.Google Scholar
Onni, T, Sanna, G, Larsen, J and Tola, S (2011) Antimicrobial susceptibilities and population structure of Staphylococcus epidermidis associated with ovine mastitis. Veterinary Microbiology 148, 4550.Google Scholar
Orden, JA, Cid, D, Blanco, ME, Quiteria, JARS, Gomez Lucia, E and de la Fuente, R (1992 a) Enterotoxin and toxic shock syndrome toxin-one production by staphylococci isolated from mastitis in sheep. APMIS 100, 132134.Google Scholar
Orden, JA, Goyache, J, Hernandez, J, Domenech, A, Suarez, G and Gomez Lucia, E (1992 b) Production of staphylococcal enterotoxins and TSST-1 by coagulase-negative staphylococci isolated from ruminant mastitis. Journal of Veterinary Medicine B 39, 144148.Google Scholar
O'Riordan, K and Lee, JC (2004) Staphylococcus aureus capsular polysaccharides. Clinical Microbiology Reviews 17, 218234.Google Scholar
Otto, M (2013) Staphylococcus aureus toxins. Current Opinion in Microbiology 17, 3237.Google Scholar
Ozenc, E, Seker, E, Acar, DB, Birdane, MK, Darbaz, I and Dogan, N (2011) The importance of staphylococci and threshold value of somatic cell count for diagnosis of subclinical mastitis in Pirlak sheep at mid-lactation. Reproduction in Domestic Animals 46, 970974.Google Scholar
Palmqvist, N, Foster, T, Tarkowski, A and Josefsson, E (2002) Protein A is a virulence factor in Staphylococcus aureus arthritis and septic death. Microbial Pathogenesis 33, 239249.Google Scholar
Pantosti, A, Sanchini, A and Monaco, M (2007) Mechanisms of antibiotic resistance in Staphylococcus aureus. Future Microbiology 2, 323334.Google Scholar
Perez, MM, Prenafeta, A, Valle, J, Penades, J, Rota, C, Solano, C, Marco, J, Grillo, MJ, Lasa, I, Irache, JM, Maira-Litran, T, Jimenez-Barbero, J, Costa, L, Pier, GB, de Andres, D and Amorena, B (2009) Protection from Staphylococcus aureus mastitis associated with poly-N-acetyl beta-1,6 glucosamine specific antibody production using biofilm-embedded bacteria. Vaccine 27, 23792386.Google Scholar
Persson, Y, Nyman, AK, Soderquist, L, Tomic, N and Waller, KP (2017) Intramammary infections and somatic cell counts in meat and pelt producing ewes with clinically healthy udders. Small Ruminant Research 156, 6672.Google Scholar
Phillipson, M and Kubes, P (2011) The neutrophil in vascular inflammation. Nature Medicine 17, 13811390.Google Scholar
Pilipcincova, I, Bhide, M, Dudrikova, E and Travnicek, M (2010) Genotypic characterization of coagulase-negative staphylococci isolated from sheep milk in Slovakia. Acta Veterinaria Brno 79, 269275.Google Scholar
Pinchuk, IV, Beswick, EJ and Reyes, VE (2010) Staphylococcal enterotoxins. Toxins (Basel) 2, 21772197.Google Scholar
Pisanu, S, Cubeddu, T, Pagnozzi, D, Rocca, S, Cacciotto, C, Alberti, A, Marogna, G, Uzzau, S and Addis, MF (2015) Neutrophil extracellular traps in sheep mastitis. Veterinary Research 46, 59.Google Scholar
Porrero, MC, Hasman, H, Vela, AI, Fernandez-Garayzabal, JF, Dominguez, L and Aarestrup, FM (2012) Clonal diversity of Staphylococcus aureus originating from the small ruminants goats and sheep. Veterinary Microbiology 156, 157161.Google Scholar
Prenafeta, A, March, R, Foix, A, Casals, I and Costa, L (2010) Study of the humoral immunological response after vaccination with a Staphylococcus aureus biofilm-embedded bacterin in dairy cows: possible role of the exopolysaccharide specific antibody production in the protection from Staphylococcus aureus induced mastitis. Veterinary Immunology Immunopathology 134, 208217.Google Scholar
Rainard, P (2017) Mammary microbiota of dairy ruminants: fact or fiction? Veterinary Research 48, 25.Google Scholar
Riggio, V and Portolano, B (2015) Genetic selection for reduced somatic cell counts in sheep milk: a review. Small Ruminant Research 126, 3342.Google Scholar
Rooijakkers, SH, Milder, FJ, Bardoel, BW, Ruyken, M, van Strijp, JA and Gros, P (2007) Staphylococcal complement inhibitor: structure and active sites. Journal of Immunology 179, 29892998.Google Scholar
Rovai, M, Caja, G, Salama, AAK, Jubert, A, Lázaro, B, Lázaro, M and Leitner, G (2014) Identifying the major bacteria causing intramammary infections in individual milk samples of sheep and goats using traditional bacteria culturing and real-time polymerase chain reaction. Journal of Dairy Science 97, 53935400.Google Scholar
Rupp, R, Bergonier, D, Dion, S, Hygonenq, MC, Aurel, MR, Robert-Granie, C and Foucras, G (2009) Response to somatic cell count-based selection for mastitis resistance in a divergent selection experiment in sheep. Journal of Dairy Science 92, 12031219.Google Scholar
Sahuquillo Arce, JM, Yarad Auad, F and Hernández Cabezas, A (2013) Biofilms: a biological antimicrobial resistance system. In Méndez-Vilas, A (ed.), Microbial Pathogens and Strategies for Combating them: Science, Technology and Education. Badajoz: Formatex, pp. 6167.Google Scholar
Saratsis, P, Leontides, L, Tzora, A, Alexopoulos, C and Fthenakis, GC (1998) Incidence risk and aetiology of mammary abnormalities in dry ewes in 10 flocks in Southern Greece. Preventive Veterinary Medicine 37, 173183.Google Scholar
Scherrer, D, Corti, S, Muehlherr, JE, Zweifel, C and Stephen, R (2004) Phenotypic and genotypic characteristics of Staphylococcus aureus isolates from raw bulk-tank milk samples of goats and sheep. Veterinary Microbiology 101, 101107.Google Scholar
Schönborn, S, Wente, N, Paduch, JH and Krömker, V (2017) In vitro ability of mastitis causing pathogens to form biofilms. Journal of Dairy Research 84, 198201.Google Scholar
Schwarz, S, Loeffler, A and Kadlec, K (2017) Bacterial resistance to antimicrobial agents and its impact on veterinary and human medicine. Veterinary Dermatology 28, 82–e19.Google Scholar
Selvaggi, M, D’ Alessandro, AG and Dario, C (2017) Environmental and genetic factors affecting milk yield and quality in three Italian sheep breeds. Journal of Dairy Research 84, 2731.Google Scholar
Silversides, JA, Lappin, E and Ferguson, AJ (2010) Staphylococcal toxic shock syndrome: mechanisms and management. Current Infectious Diseases Reports 12, 392400.Google Scholar
Skrlin, J (2016) Impact of biofilm on healing and a method for identifying it in the wound. Acta Medica Croatica 70, 2932.Google Scholar
Spaan, NA, van Strijp, JAG and Torres, VJ (2017) Leukocidins: Staphylococcal bi-component pore-forming toxins find their receptors. Nature Reviews Microbiology 15, 435447.Google Scholar
Stemerding, AM, Köhl, J, Pandey, MK, Kuipers, A, Leusen, JH, Boross, P, Nederend, M, Vidarsson, G, Weersink, AY, van de Winkel, JG, van Kessel, KP and van Strijp, JA (2013) Staphylococcus aureus formyl peptide receptor-like 1 inhibitor (FLIPr) and its homologue FLIPr-like are potent FcγR antagonists that inhibit IgG-mediated effector functions. Journal of Immunology 191, 353362.Google Scholar
Stutz, K, Stephan, R and Tasara, T (2011) Spa, ClfA, and FnbA genetic variations lead to Staphaurex test-negative phenotypes in bovine mastitis Staphylococcus aureus isolates. Journal of Clinical Microbiology 49, 638646.Google Scholar
Tejada, TS, Silva, DT, Dias, PA, Conceicao, RCS, Neto, HM and Timm, CD (2012) Subclinical mastitis caused by Staphylococcus coagulase negative in meet-producing sheep. Arquivo Brasileiro de Medicina Veterinaria e Zootecnia 64, 10741076.Google Scholar
Tel, OY, Aslantas, O, Keskin, O, Yilmaz, ES and Demir, C (2012) Investigation of the antibiotic resistance and biofilm formation of Staphylococcus aureus strains isolated from gangrenous mastitis of ewes. Acta Veterinaria Hungarica 60, 189197.Google Scholar
Titchen, DA (1977) Cineradiographic studies of swallowing in the suckled lamb. Annales de Recherche Veterinaire 8, 483.Google Scholar
Unal, N and Cinar, OD (2012) Detection of staphylococcal enterotoxin, methicillin-resistant and Panton-Valentine leukocidin genes in coagulase-negative staphylococci isolated from cows and ewes with subclinical mastitis. Tropical Animal Health and Production 44, 369375.Google Scholar
Unal, N, Askar, S, Macun, HC, Sakarya, F, Altun, B and Yildirim, M (2012) Panton-Valentine leukocidin and some exotoxins of Staphylococcus aureus and antimicrobial susceptibility profiles of staphylococci isolated from milks of small ruminants. Tropical Animal Health and Production 44, 573579.Google Scholar
Van Oostveldt, K, Paape, MJ, Dosogne, H and Burvenich, C (2002) Effect of apoptosis on phagocytosis, respiratory burst and CD18 adhesion receptor expression of bovine neutrophils. Domestic Animal Endocrinology 22, 3750.Google Scholar
Vasileiou, NGC (2019) Mastitis in Ewes associated with Staphylococcus spp.: New Clinical, Epidemiological, Management, Microbiological and Zoonotic Findings and Evaluation of a Novel Vaccine against the Disease (PhD thesis). University of Thessaly.Google Scholar
Vasileiou, NGC, Chatzopoulos, DC, Gougoulis, DA, Sarrou, S, Katsafadou, AI, Spyrou, V, Mavrogianni, VS, Petinaki, E and Fthenakis, GC (2018 a) Slime-producing staphylococci as causal agents of subclinical mastitis in sheep. Veterinary Microbiology 224, 9399.Google Scholar
Vasileiou, NGC, Cripps, PJ, Ioannidi, KS, Chatzopoulos, DC, Gougoulis, DA, Sarrou, S, Orfanou, DC, Politis, A, Calvo Gonzalez-Valerio, T, Argyros, S, Mavrogianni, VS, Petinaki, E and Fthenakis, GC (2018 b) Extensive countrywide field investigation of subclinical mastitis in sheep in Greece. Journal of Dairy Science 101, 72977310.Google Scholar
Vasileiou, NGC, Gougoulis, DA, Riggio, V, Ioannidi, KS, Chatzopoulos, DC, Mavrogianni, VS, Petinaki, E and Fthenakis, GC (2018 c) Association of subclinical mastitis prevalence with sheep breeds in Greece. Journal of Dairy Research 85, 317320.Google Scholar
Vasileiou, NGC, Chatzopoulos, DC, Cripps, PJ, Ioannidi, KS, Gougoulis, DA, CHouzouris, TM, Lianou, DT, Calvo Gonzalez-Valerio, T, Guix Vallverdu, R, Argyros, S, Cesio, M, Font, I, Mavrogianni, VS, Petinaki, E and Fthenakis, GC (2019 a) Evaluation of efficacy of a biofilm-embedded bacteria-based vaccine against staphylococcal mastitis in sheep – a randomized, placebo-controlled field study. Journal of Dairy Science. in press.Google Scholar
Vasileiou, NGC, Sarrou, S, Papagiannitsis, C, Chatzopoulos, DC, Malli, E, Mavrogianni, VS, Petinaki, E and Fthenakis, GC (2019 b) Antimicrobial agent susceptibility and typing of staphylococcal isolates from subclinical mastitis in ewes. Microbial Drug Resistance. doi: 10.1089/mdr.2019.0009Google Scholar
Vautor, E, Carsenti-Dellamonica, H, Sabah, M, Mancini, G, Pepin, M and Dellamonica, P (2007) Characterization of Staphylococcus aureus isolates recovered from dairy sheep farms (agr group, adherence, slime, resistance to antibiotics). Small Ruminant Research 72, 197199.Google Scholar
Verhoeven, PO, Gagnaire, J, Botelho-Nevers, E, Grattard, F, Carricajo, A, Lucht, F, Pozzetto, B and Berthelot, P (2014) Detection and clinical relevance of Staphylococcus aureus nasal carriage: an update. Expert Review of Anti-Infective Therapy 12, 7589.Google Scholar
Wang, B and Muir, TW (2016) Regulation of virulence in Staphylococcus aureus: molecular mechanisms and remaining puzzles. Cell Chemical Biology 23, 214224.Google Scholar
Wang, H, McEntire, JC, Zhang, L, Li, X and Doyle, M (2012) The transfer of antibiotic resistance from food to humans: facts, implications and future directions. Revue Scientifique et Technique – Office International des Epizooties 31, 249260.Google Scholar
Watson, DL (1988) Vaccination against experimental staphylococcal mastitis in ewes. Research in Veterinary Science 45, 1621.Google Scholar
Wieckowska-Szakiel, M, Sadowska, B and Rózalska, B (2007) Staphylokinase production by clinical Staphylococcus aureus strains. Polish Journal of Microbiology 56, 97102.Google Scholar
Winn, WC and Koneman, EW (2006) Koneman's Color Atlas and Textbook of Diagnostic Microbiology, 6th Edn. Philadelphia: Lippincott Williams & Wilkins.Google Scholar
Winter, P and Colditz, IG (2002) Immunological responses of the lactating ovine udder following experimental challenge with Staphylococcus epidermidis. Veterinary Immunology and Immunopathology 89, 5765.Google Scholar
Winter, P and Hofer, E (1996) Coagulase-negative staphylococci as a pathogen of subclinical and clinical mastitis in three flocks of milk sheep. Tierarztliche Umschau 51, 222226.Google Scholar
Winter, P, Hoflechner, A and Baumgartner, W (1999) Resistance patterns of ovine mastitis pathogens. Berliner und Munchener Tierarztliche Wochenschrift 112, 216222.Google Scholar
Winter, P, Rammelmayr, A and Baumgartner, W (2002) Udder health status of dairy flocks during one lactation period with special regards to infections caused by coagulase-negative staphylococci. Wiener Tierarztliche Monatsschrift 89, 242248.Google Scholar
Winter, P, Schilcher, F, Fuchs, K and Colditz, IG (2003) Dynamics of experimentally induced Staphylococcus epidermidis mastitis in East Friesian milk ewes. Journal of Dairy Research 70, 157164.Google Scholar
Figure 0

Table 1. Clustering of staphylococcal species into 11 groups based on results of 16s rRNA sequencing

Figure 1

Table 2. Virulence factors involved in the pathogenetic role of a typical S. aureus isolate

Figure 2

Table 3. Selected references regarding implication of non-S. aureus staphylococcal strains in mastitis or mammary carriage in ewes

Supplementary material: PDF

Vasileiou et al. supplementary material

Vasileiou et al. supplementary material 1

Download Vasileiou et al. supplementary material(PDF)
PDF 913.1 KB