Open access peer-reviewed chapter

Bovine Mastitis: Part I

Written By

Oudessa Kerro Dego

Submitted: 15 October 2019 Reviewed: 27 July 2020 Published: 02 September 2020

DOI: 10.5772/intechopen.93483

From the Edited Volume

Animal Reproduction in Veterinary Medicine

Edited by Faruk Aral, Rita Payan-Carreira and Miguel Quaresma

Chapter metrics overview

1,129 Chapter Downloads

View Full Metrics

Abstract

Bovine mastitis is one of the most important bacterial diseases of dairy cattle throughout the world. Mastitis is responsible for great economic losses to the dairy producer and to the milk processing industry resulting from reduced milk production, alterations in milk composition, discarded milk, increased replacement costs, extra labor, treatment costs, and veterinary services. Economic losses due to bovine mastitis are estimated to be $2 billion in the United States, $400 million in Canada (Canadian Bovine Mastitis and Milk Quality Research Network-CBMQRN) and $130 million in Australia per year. Many factors can influence the development of mastitis; however, inflammation of the mammary gland is usually a consequence of adhesion, invasion, and colonization of the mammary gland by one or more mastitis pathogens such as Staphylococcus aureus, Streptococcus uberis, and Escherichia coli.

Keywords

  • mastitis
  • bovine
  • Staphylococcus
  • Streptococcus

1. Introduction

Bovine mastitis is one of the most important bacterial diseases of dairy cattle throughout the world. Mastitis is responsible for major economic losses to the dairy producer and milk processing industry resulting from reduced milk production, alterations in milk composition, discarded milk, increased replacement costs, extra labor, treatment costs, and veterinary services [1]. Annual economic losses due to bovine mastitis are estimated to be $2 billion in the United States [2], $400 million in Canada (Canadian Bovine Mastitis and Milk Quality Research Network-CBMQRN), and $130 million in Australia [3]. Many factors including host, pathogen, and environmental factors influence the development of mastitis; however, inflammation of the mammary gland is usually a consequence of adhesion, invasion, and colonization of the mammary gland by one or more contagious (Staphylococcus aureus, Streptocococcus agalactiae, Corynebacterium bovis, Mycoplasmsa bovis, etc.) or environmental (coliform bacteria, environmental Streptococcus spp. and some coagulase negative Staphylococcus spp., many other minor pathogens) mastitis pathogens.

Advertisement

2. Etiology of mastitis

Over 135 various microorganisms have been identified from bovine mastitis. The most common bovine mastitis pathogens are classified as contagious and environmental mastitis pathogens [4]. This classification depends upon their distribution in their natural habitat and mode of transmission from their natural habitat to the mammary glands of dairy cows [5]. It is important to mention that all pathogens lists as environmental or contagious may not be strictly environmental or strictly contagious; some of them may transmit both ways. Environmental mastitis pathogens exist in the cow’s environment, and they can cause infection at any time. Environmental mastitis pathogens are difficult to control because they are in the environment of dairy cows and can transmit to the mammary glands at any time, whereas contagious mastitis pathogens exist in the infected udder or on the teat skin and transmit from infected to non-infected udder during milking by milker’s hand or milking machine liners. Environmental mastitis pathogens include a wide range of organisms, including coliform bacteria (Escherichia coli, Klebsiella spp., Enterobacter spp., and Citrobacter spp), environmental Streptococcus spp. (Streptococcus uberis, Streptococcus dysgalactiae, Streptococcus equi, Streptococcus zooepidemicus, Streptococcus equinus, Streptococcus canis, Streptococcus parauberis, and others), Trueperella pyogenes, which was previously called Arcanobacterium pyogenes or Corynebacterium pyogenes and environmental coagulase-negative Staphylococcus species (CNS) (S. chromogenes, S. simulans, S. epidermidis, S. xylosus, S. haemolyticus, S. warneri, S. sciuri, S. lugdunensis, S. caprae, S. saccharolyticus, and others) [46, 7, 8, 9] and others such as Pseudomonas, Proteus, Serratia, Aerococcus, Listeria, Yeast and Prototheca that are increasingly found as mastitis-causing pathogens on some farms [10, 11].

Contagious mastitis pathogens primarily exist in the infected mammary glands or on the cow’s teat skin and transmit from infected to non-infected mammary glands during milking by milker’s hand or milking machine liners. Mycoplasma spp. may spread from cow to cow through aerosol transmission and invade the udder subsequent to bacteremia. The most frequent contagious mastitis pathogens are coagulase-positive Staphylococcus aureus, Streptococcus agalactiae, Mycoplasma bovis, and Corynebacterium bovis [11, 12]. The prevalence of mastitis caused by these different mastitis pathogens varies depending on herd management practices, geographical location, and other environmental conditions [13]. These different causative agents of mastitis have a multitude of virulence factors that make treatment and prevention of mastitis difficult.

2.1 Environmental mastitis pathogens

It is important to mention that all environmental mastitis pathogens may not be strictly environmental, and some of them may transmit both ways (contagious and environmental). However, the vast majority of these organisms are in the environment of dairy cows, and they transmit from these environmental sources to the udder of a cow at any time of the lactation cycle.

2.1.1 Streptococcus uberis mastitis

Streptococcus uberis is one of the environmental mastitis pathogens that accounts for a significant proportion of subclinical and clinical mastitis in lactating and non-lactating cows and heifers [14]. This organism is commonly found in the bedding material, which facilitates infection of mammary glands at any time [15]. Some report also indicated the possibility of contagious transmission of Streptococcus uberis [16].

S. uberis has various mechanisms of virulence that increases the chances of this organism establishing infection. These include a capsule, which evades phagocytosis, adherence to, and invasion into mammary epithelial cells [17, 18]. S. uberis adheres to epithelial cells using different mechanisms, including the formation of pedestals [19] and bridge formation through Streptococcus uberis adhesion molecule (SUAM) and lactoferrin [20, 21, 22]. This attachment is specific and mediated through a bridge formation between Streptococcus uberis adhesion molecule (SUAM) [2324] on S. uberis surface and lactoferrin, which is in the mammary secretion and has a receptor on the mammary epithelial surface [20, 22]. This interaction creates a molecular bridge that enhances S. uberis adherence to and internalization into mammary epithelial cells most likely via caveolae-dependent endocytosis and potentially allows S. uberis to evade host defense mechanisms [22, 24]. These factors increase the pathogenicity of S. uberis to cause mastitis. The sua gene is conserved among strains of S. uberis isolated from geographically diverse areas [9, 13], and a sua deletion mutant of S. uberis is defective in adherence to and internalization into mammary epithelial cells [14].

2.1.2 Coagulase-negative Staphylococcus species (CNS)

More recently, coagulase-negative Staphylococcus species (CNS) such as S. chromogenes, S. simulans, S. xylosus, S. haemolyticus, S. hyicus, and S. epidermidis are increasingly isolated from bovine milk [7, 25, 26, 27] with S. chromogenes being the most increasingly diagnosed species as a cause of subclinical mastitis. Staphylococcus chromogenes [28] and other CNS [4, 8] have been shown to cause subclinical infections in dairy cows that reduce the prevalence of contagious mastitis pathogens.

Staphylococcus chromogenes is most commonly isolated from mammary secretions rather than from the environment itself [8, 29]. S. chromogenes consistently isolated from the cow’s udder and teat skin [30], and some studies showed that it causes long-lasting, persistent subclinical infections [26]. The CNS causes high somatic cell counts in milk on some dairy farms [29, 31]. Woodward et al. [32] evaluated the normal teat skin flora and found that 25% of the isolates exhibited the ability to prevent the growth of some mastitis pathogens. An in vitro study conducted on S. chromogenes showed that this organism could inhibit the growth of major mastitis-causing pathogens such as Staph. aureus, Strep. dysgalactiae, and Strep. uberis [28]. In a study conducted on conventional and organic Canadian dairy farms, CNS were found in 20% of the clinical samples [33]. Recently, mastitis caused by CNS increasingly became more problematic in dairy herds [30, 34, 35, 36]. However, mastitis caused by CNS is less severe compared to mastitis caused by Staphylococcus aureus [26].

2.1.3 Coliform mastitis

Coliform bacteria such as Escherichia, Klebsiella, and Enterobacter are a common cause of mastitis in dairy cows [37]. The most common species, isolated in more than 80% of cases of coliform mastitis, is Escherichia coli [38, 39]. E. coli usually infects the mammary glands during the dry period and progresses to inflammation and clinical mastitis during the early lactation with local and sometimes severe systemic clinical manifestations. Some reports indicated that the severity of E. coli mastitis is mainly determined by cow factors rather than by virulence factors of E. coli [40]. However, recent molecular and genetic studies showed that the pathogenicity of E. coli is entirely dependent on the FecA protein that enables E. coli to actively uptake iron from ferric-citrate in the mammary gland [41]. The severity of the clinical mastitis and peak E. coli counts in mammary secretions are positively correlated. Intramammary infection with E. coli induced expression and release of pro-inflammatory cytokines [42, 43]. Recently, it has been shown with mouse mastitis models that IL-17A and Th17 cells are instrumental in the defense against E. coli intramammary infection [44, 45]. However, the role of IL-17 in bovine E. coli mastitis is not well defined. The result of recent vaccine efficacy study against E. coli mastitis suggested that cell-mediated immune response has more protective effect than humoral response [46]. However, the cytokine signaling pathways that lead to efficient bacterial clearance are not clearly defined.

2.2 Contagious mastitis pathogens

2.2.1 Coagulase-positive Staphylococcus aureus

Coagulase-positive Staphylococcus aureus is one of the most common contagious mastitis pathogens in dairy cows, with an estimated incidence rate of 43–74% [47, 48]. Staphylococcus aureus is grouped under the family Staphylococcaceae and genus Staphylococcus. It is a gram-positive, catalase and coagulase-positive, non-spore forming, oxidase negative, non-motile, cluster-forming, and facultative anaerobe [49]. The coagulase test is not an absolute test for the confirmation of the diagnosis of S. aureus from the cases of bovine mastitis, but more than 95% of all coagulase-positive staphylococci from bovine mastitis belong to S. aureus [50]. Other coagulase-positive species include S. aureus subsp. anaerobius causes lesion in sheep; S. pseudintermedius causes pyoderma, pustular dermatitis, pyometra, otitis externa, and other infections in dogs and cats; S. schleiferi subsp. coagulans causes otitis externa (inflammation of the external ear canal) in dogs; S. hyicus is coagulase variable (some strains are positive and some others are negative), species that causes mastitis in dairy cows, exudative epidermitis (greasy pig disease) in pigs; and S. delphini causes purulent cutaneous lesions in dolphins.

S. aureus can infect many host species, including humans. In humans, S. aureus causes a wide variety of illnesses ranging from mild skin infection to a life-threatening systemic infection. It has been reported that certain strains of S. aureus with specific tissue tropism can be adapted to infect specific tissue such as the mammary gland [51]. Furthermore, a study by McMillan [52] showed distinct lineages of S. aureus in bovine, ovine, and caprine species. S. aureus strains can be host specific, meaning that they are found more commonly in a specific species [51]. Some studies showed that S. aureus that causes mastitis belong to certain dominant clones, which are frequently responsible for clinical and subclinical mastitis in a herd at certain geographic areas, indicating that the control measures may need to be directed against specific clones in a given area [53, 54, 55]. However, because S. aureus is such a big problem in human health, cross-infection has been an important research topic. Several studies have reported cases of cross-infection in several different species [56, 57, 58]. In the dairy industry, there have been reports of human origin methicillin-resistant S. aureus infecting bovine mammary glands [59, 60]. These studies add to the unease that strains can gain new mutations or virulence factors and adapt to cross the interspecies boundary relatively rapidly [61].

Although the incidence of S. aureus mastitis can be reduced with hygienic milking practices and a good management system, it is still a major problem for dairy farms, with a prevalence of 66% among farms tested in the United States [62]. The prevalence of S. aureus mastitis varies from farm to farm because of variation in hygienic milking practices and overall farm management differences on the application of control measures for contagious mastitis pathogens. Good hygiene in the milking parlor can significantly reduce the occurrence of new S. aureus mastitis in the herd, but it does not remove existing cases within a herd [63]. Neave et al. concluded that it is nearly impractical to keep all udder quarters of dairy cows free of all pathogens at all times. Since this early observation by Neave et al. [63], many studies have confirmed that management practices can reduce new cases of intramammary infection (IMI) [9, 64] but cannot eliminate existing infections. In the United States, the prevalence of clinical and subclinical S. aureus mastitis ranged from 10 to 45% [65] and 15 to 75%, respectively.

2.2.1.1 Virulence factors of S. aureus

Staphylococcus aureus has many virulence factors that can be grouped broadly into two major classes. These include (1) secretory factors which are surface localized structural components that serve as virulence factors and (2) secretory virulence factors which are produced by bacteria cells and secreted out of cells and act on different targets in the host body. Both non-secretory and secretory virulence factors together help this pathogen to evade the host’s defenses and colonize mammary glands.

2.3 Non-secretory factors

Some of surface localized structural components that serve as virulence factors include membrane-bound proteins, which include collagen-binding protein, fibrinogen-binding protein, elastin-binding protein, penicillin-binding protein, and lipoteichoic acid. Similarly, cell wall-bound factors such as peptidoglycan, lipoteichoic acid, teichoic acid, protein A, β-Lactamase, and proteases serve as non-secretory virulence factors. Other cell surface-associated virulence factors include exopolysaccharides, which comprises capsule, slime, and biofilm. Overall, S. aureus has over 24 surface proteins and 13 secreted proteins that are involved in immune evasion [66] and about 15–26 proteins for biofilm formation [67, 68].

Surface proteins, such as staphylococcal protein A (SpA), clumping factors A and B (ClfA and ClfB) [69, 70, 71], fibrinogen-binding proteins [72], iron-regulated surface determinants (IsdA, IsdB, and IsdH) [69, 73], fibronectin-binding proteins A and B [74], biofilm associated protein (BAP) and exopolysaccharides (capsule, slime, and biofilms) [75, 76, 77, 78, 79], play roles in S. aureus adhesion to and invasion into host cells [80]. The BAP expression enhances biofilm production and the BAP gene is only found in S. aureus strain from bovine origin [81, 82, 83]. Evaluation of BAP gene of S. aureus from bovine and human isolates using polymerase chain reaction restriction fragment length polymorphism (PCR-RFLP) showed that bovine and human isolates are not closely related [84]. Thus, some host-specific evolutionary factors may have been developed between both strain types.

Biofilms are considered an important virulence factor in the pathogenesis of bovine S. aureus mastitis [77, 78]. Slime, an extracellular polysaccharide layer, acts as a barrier against phagocytosis and antimicrobials. It also helps with adhesion to a surface [85]. If a biofilm forms in a mammary gland, it will protect those bacteria from antimicrobials and the host’s immune system [77, 78]. In addition, once the biofilm matures and the immune attack has subsided, the biofilm can break open and allow reinfection of the mammary gland [86]. There are many contributors to biofilm production, such as polysaccharide intercellular adhesin (PIA) also known as poly-N-acetyl-β (1-6)-glucosamine (PNAG), MSCRAMMS, teichoic acids, and extracellular DNA (eDNA) [75, 76] that are known to help these bacteria cells to hold onto a surface [87]. Various proteins encoded by intercellular adhesion loci such as icaA, icaB, icaC, and icaD are involved in PIA production which in turn result in biofilm formation [75, 76]. Vasudevan et al. [88] evaluated the correlation of slime production and presence of the intercellular adhesion (ica) genes with biofilm production. These authors [88] found that all tested isolates were positive for icaA and icaD genes, and most tested isolates produce slime, but not all slime positives produced biofilms in vitro. Similarly, a study in Poland found that all isolates were positive for icaA and icaD [80] genes. While adhesion is promoted with biofilm production, the bap gene prevents the invasion of host cells [83]. Despite the presence of the ica gene strongly support biofilm production, the presence of the ica gene is not mandatory for biofilm production since S. aureus lacking ica gene can still produce biofilm through other microbial surface components recognizing adhesive matrix molecules (MSCRAM) and secreted proteins [89, 90].

2.4 Secretory factors

Some of the known secretory virulence factors are toxins which include staphylococcal enterotoxins, non-enteric exfoliative toxins, toxic shock syndrome toxin 1, leucocidin, and hemolysins (alpha, beta, delta, and gamma) [91, 92]. Similarly, enzymes such as coagulase, staphylokinase, DNAase, phosphatase, lipase, phospholipase, and hyaluronidase serve as virulence factors of S. aureus [93].

2.4.1 Hemolysins

S. aureus isolates from bovine mastitis produce alpha (α), beta (β), gamma (γ), and delta (δ) hemolysins that cause hemolysis of red blood cells of the host [94] and all are antigenically distinct. α-hemolysin is a pore-forming toxin that binds to a disintegrin and metalloproteinase domain-containing protein-10 (ADAM10) receptor resulting in pore formation and cellular necrosis [95, 96]. It is also known to increase the inflammatory response and decrease macrophage function [97]. α-hemolysin damages the plasma membrane of the epithelial cell resulting in leakages of low-molecular-weight molecules from the cytosol and death of the cell [98]. It is produced by 20–50% of strains from bovine IMI [99]. A study reported that the α-hemolysin might be required for a cell to cell interaction during biofilm formation [100]. β-hemolysin hydrolyzes the sphingomyelin present in the plasma membrane resulting in increased permeability with progressive loss of cell surface charge [101]. It is produced by 75–100% of S. aureus strains from bovine IMI [99]. α-hemolysin expression requires specific growth conditions in vitro because its growth is inhibited by agar [102]. α-hemolysin producing strains cause complete hemolysis of sheep red blood cells, whereas β-hemolysin producing strains cause partial hemolysis within 24 h of incubation at 37°C [103]. Partial hemolysis caused by β-hemolysin becomes completely lysed after further storage at 4–15°C, which is also expressed as hot-cold lysis [104]. β-hemolysin producing strains are the most frequent isolates from animals [105]. δ-hemolysin causes complete hemolysis of red blood cells of wide range of species including human, rabbit, sheep, horse, rat, guinea pig, and some fish erythrocytes. δ-hemolysin migrates more slowly through agar than the α-hemolysin so the effect takes longer time to express. Double (α- and β-) hemolysin producing strains caused complete hemolysis in the middle with partial hemolysis on the peripheral area around each colony [105]. γ-hemolysin is produced by almost every strain of S. aureus, but γ-hemolysin is not identifiable on blood agar plates, due to the inhibitory effect of agar on toxin activity [106].

2.4.2 Enterotoxins Enterotoxins

These toxins are heat stable and can resist pasteurization. S. aureus produces staphylococcal enterotoxins A, B, C, D, E, G, H, I, and J–Q as well as toxic shock syndrome toxin 1 (tsst-1) [105, 107, 108]. Enterotoxins can get into the food chain through the consumption of contaminated food and cause food poisoning [109]. Staphylococcal enterotoxins tend to contaminate dairy products and cause foodborne illness [110, 111]. Staphylococcal enterotoxins G to Q (SEG–SEQ) are prevalent among S. aureus isolates from cases of bovine mastitis and are also implicated in the pathogenesis of mastitis. Some of these toxins are known to function as superantigens that cause increased immunological reactivity in the host [110]. Some studies showed that about 20% of S. aureus isolates from IMI produce toxic shock syndrome toxin-1 [109, 112]. Toxic shock syndrome toxin causes toxic shock syndrome and can be fatal [113]. Besides the superantigenic effect of enterotoxins, their role in the pathogenesis of mastitis is unknown. It may be specific to each strain or area based on selective pressures in the habitat [114]. Enterotoxin prevalence seems to vary between geographical regions. The strains producing enterotoxin C have been isolated relatively frequently from cases of bovine mastitis [108, 115, 116].

Enterotoxins are believed to have a role in the development of mastitis since S. aureus isolates from cases of mastitis had a high prevalence of enterotoxins than isolates from milk of cows without mastitis [117, 118]; however, staphylococcal enterotoxins expressions are controlled by several regulatory elements [119] that respond to a variety of different micro-environmental stimuli and the exact mechanisms by which enterotoxins contribute to the development of mastitis are not clearly known and yet to be determined.

In addition to specific virulence factors, Staphylococcus aureus also possesses different mechanisms or traits such as biofilm formation, adhesion to and invasion into mammary epithelial cells, and formation of small colony variant (SCV) that enable this pathogen to resist host defense mechanisms. The ability of S. aureus to invade mammary epithelial cells during mastitis plays a significant role in the pathogenesis of S. aureus. Internalized bacteria can hide from the host’s immune system inside the host cell and continue to multiply inside the host cell [120]. There may be many mechanisms that S. aureus uses to invade into host cells, and each mechanism can be strain dependent. S. aureus strains have a fibronectin-binding protein that can link to the fibronectin on the mammary epithelial cell surface. Fibronectin binding protein is thought to be a common way for the bacteria cells to invade bovine mammary epithelial cells. Fibronectin-binding protein-deficient strains cannot invade host cells [121]. The presence of a capsule prevents adherence to epithelial cells [122, 123].

Adhesion is the first step in the formation of biofilm or the invasion of host cells, which protects the bacteria from the host immune system and facilitates chronic infection [124]. Adhesion is dependent on surface proteins called adhesins, which help the bacterium to recognize and attach to host cells. Staphylococci are coated with a wide variety of surface proteins that help them to adhere to host cells and extracellular matrix components. Microbial surface components recognizing adhesive matrix molecules (MSCRAMMs) of the host are the most common surface proteins that are involved in adhesion [124]. The ability to bind to host tissue or the host’s cell surface is a pivotal part of the bacteria’s pathogenicity because adhesion is typically the first step in the invasion and biofilm formation [125, 126].

Adhesion to and invasion into epithelial cells [124], intracellular survival in macrophages [127], and epithelial cells allow them to avoid detection by the host immune system and resist treatment with antibiotics [120]. Due to its poor response to treatments, S. aureus infections often become chronic with a low cure rate [128]. Treatment of Staphylococcus aureus mastitis with cloxacillin cured only 25% of the clinical cases and 40% of subclinical cases in the study by Tyler and Baggot [129]. Staphylococcus aureus also has a known ability to form biofilms [77, 78, 86] and acquire antimicrobial-resistance genes via horizontal resistance gene transfer, which enables this bacterium to develop antimicrobial resistance [130, 131].

The mode of transmission from infected mammary glands or colonized udder skin to healthy mammary glands is through contact during milking procedures with milker’s hand, towel, and milking machine [58]. S. aureus usually causes subclinical or chronic infections and is difficult to clear with antibiotic treatment [132].

2.4.3 Streptococcus agalactiae

The most important virulence factor of S. agalactiae is the capsular polysaccharide [133], which protects this bacterium from being engulfed by macrophages and subsequently phagocytosed [133]. Another virulence factor of S. agalactiae is the Rib protein, which confers resistance to proteases. Emaneini et al. [133] found that the Rib encoding gene (rib) was detected in 89% of the isolates from bovine origin. Streptococcus agalactiae causes persistent infections that are usually difficult to clear without antibiotic treatment [134]. Though Streptococcus agalactiae is highly contagious, it has good response to treatment with antibiotics, which makes it possible to eliminate from herds with current mastitis control measures [129]. Since the adoption of hygienic milking practices, the incidence of mastitis caused by S. agalactiae has dramatically decreased and is now rarely observed in dairy herds [135].

2.4.4 Mycoplasma mastitis

Mastitis caused by Mycoplasma spp. is a growing concern in the United States. It is believed that this organism has been underreported due to the difficulty of isolation by culture method [136]. The incidence of Mycoplasma mastitis varies across the globe, with a 3.2% prevalence rate in the United States that may increase to 14.4% in larger herd size of greater than 500 cows [47, 48, 62, 137]. A risk factor for Mycoplasma mastitis increase with herd size, and most of the Mycoplasma mastitis cases are subclinical infections with outbreaks linked to asymptomatic carriers [138]. Pathogenesis of most Mycoplasma spp. infection is characterized by adherence to and internalization into host cells resulting in colonization of the host with immune modulation without causing severe disease [138]. Mycoplasma species lack a cell wall, thus not sensitive to beta-lactam antibiotics, but showed sensitivity to non-beta-lactam antibiotics [139].

Advertisement

3. Routes of entry of mastitis pathogens to the udder

In general, it is believed that mastitis pathogens gain entrance to the udder through teat opening into the teat canal and from the teat canal into the intramammary area during the reverse flow of milk due to vacuum pressure fluctuation of the milking machine [9]. However, the detailed mechanism of mastitis pathogen colonization of the mammary gland may vary among species of bacteria and the virulence factors associated with particular strain in each species. An example of this is in some cases; it has been shown that E. coli can penetrate the teat canal without the reverse flow of milk [9]. Some of the major mastitis pathogens, such as E. coli [140], Staphylococcus aureus, and Streptococcus uberis [20, 21, 22] can adhere to and subsequently invade into the mammary epithelial cells. This adherence and subsequent invasion into mammary epithelial cells allow them to persist in the intracellular area as well as to escape the host immune defenses attack and action of antimicrobial drugs [120, 140, 141, 142, 143, 144]. Dogan et al. [145] compared E. coli strains known to cause chronic infections with strains known to cause acute infections and found that chronic strains were more invasive to the epithelial cells, leading to the difficulty in clearance and persistent infection compared to acute strains. S. aureus enters the mammary gland through the teat opening and subsequently multiply in the mammary gland where they may form biofilms, attach to, and internalize into the mammary epithelial cells causing inflammation of mammary glands characterized by swelling, degeneration of epithelial cells, and epithelial erosions and ulcers [146, 147].

Advertisement

4. Clinical manifestation of mastitis

Depending on clinical signs, mastitis can also be divided into clinical and subclinical mastitis. Clinical mastitis is characterized by visible inflammatory changes (abnormalities) in the mammary gland tissue such as redness, swelling, pain, increased heart, and abnormal changes in milk color (watery, bloody, and blood tinged) and consistency (clots or flakes) [9]. Clinical mastitis can be acute, peracute, subacute, or chronic. Acute mastitis is a very rapid inflammatory response characterized by systemic clinical signs which include fever, anorexia, shock, as well as local inflammatory changes in the mammary gland and milk. Peracute mastitis is manifested by a rapid onset of severe inflammation, pain, and systemic symptoms that resulted in a severely sick cow within a short period of time. Subacute mastitis is the most frequently seen form of clinical mastitis characterized by few local signs of mild inflammation in the udder and visible changes in milk such as small clots. Chronic mastitis is a long-term recurring, persistent case of mastitis that may show few symptoms of mastitis between repeated occasional flare-ups of the disease where signs are visible and can continue over periods of several months. Chronic mastitis often leads to irreversible damage to the udder from the repeated occurrences of the inflammation, and often these cows are culled.

Subclinical mastitis is the inflammation of the mammary gland that does not create visible changes in the milk or the udder. Subclinical mastitis is an infection of mammary gland characterized by non-visible inflammatory changes such as a high somatic cell count coupled with shedding of causative bacteria through milk [9]. During this inflammatory process, the milk samples showed a rapid increase of somatic cells, characterized by increased number of neutrophils in the secretion [146, 148]. Despite increased recruitment of somatic cells into infected mammary glands, evidenced by an increased number of neutrophils, infection usually does not clear but became subclinical. Intramammary infections during early lactation may become acute clinical mastitis characterized by gangrene development due congestion and thrombosis (blockage) of blood supply to the tissue but most new infection during late lactation or dry period become acute or chronic mastitis [149, 150].

The increase in somatic cell count during subclinical infections leads to a decrease in useful components in the milk, such as lactose and casein [151]. Lactose is the sugar found in milk, and casein is one of the major proteins in milk and decreases in these two components affect the quality and quantity of milk yield [9]. During mastitis, there is an increase in lipase and plasmin, which have a detrimental effect on the quantity and quality of milk due to the breakdown of milk fat and casein [9]. Subclinical infections can reduce milk production by 10–12% when just one-quarter is infected [152]. These subclinical infections cause some of the greatest unseen economic [20] losses because of their detrimental impact on production and milk quality without showing visible signs of infection [152].

Advertisement

5. Risk factors for mastitis

There are host-, pathogen-, and environmental-related risk factors that predispose dairy cows to mastitis. The host risk factors include age (parity), stage of lactation, somatic cell count, breed, the anatomy of the mammary glands/morphology of udder and teat (diameter of teat canal and conformation of the udder), and immune competence (immunity) [153] (Figure 1). The environmental risk factors include the proper functioning status of milking machine, udder trauma, sanitation, climate, nutrition, management, season, and housing condition [154] (Figure 1). The pathogen risk factors include type (bacteria, fungi, yeast, and algae), number (large number and small number), virulence (highly, moderate, or less virulent), frequency of exposure (dirty farm floor, dirty milking machine, and dirty teat drying towels frequently expose to pathogen; clean floor, clean milking machine, and clean teat drying towels less exposure to pathogens), ability to resist flushing out of the glands by milk (ability to adhere or attach to and invade or internalize into mammary epithelial cells), zoonotic (transmit from cow to human or vice versa) potential, and resistance to antimicrobials [4] (Figure 1). The warm, humid, and moist climate favors the growth of bacteria and increases the chances of intramammary infection (IMI) and mastitis development [154]. The incidence of mastitis varies from farm to farm due to the combined effects of these different factors that increase the risk of disease development.

Figure 1.

Risk factors for mastitis. SA, Staphylococcus aureus; EC, Escherichia coli; SU, Streptococcus uberis; SCC, somatic cell count; AMR, antimicrobial resistance.

Dairy cows are highly susceptible to IMI during the early dry period due to increased colonization of teat skin with bacteria. Bacterial colonization of teat increases during the early dry period because of an absence of hygienic milking practices including pre-milking washing and drying of teats [155], as well as pre- and post-milking teat dipping in antiseptic solutions [156, 157] that are known to reduce teat end colonization and infection. An udder infected during the early dry period usually manifests clinical mastitis during the transition period because of increased production of parturition inducing immunosuppressive hormones [158159], negative energy balance [160], and physical stress during calving [161].

Advertisement

6. Role of mastitis on public health

Mastitis is increasingly becoming a public health concern due to the ability of the causative bacterial pathogens and/or their products, such as enterotoxins, to enter the food supply and cause foodborne diseases [109, 162], especially through the consumption of raw milk [29] and undercooked meat of culled dairy cows due to chronic mastitis that are usually sold to the slaughter (abattoir) for meat consumption. The Center for Disease Control (CDC) estimated that roughly 48 million people in the United States a year become sick from foodborne diseases [163]. Foodborne pathogens have been detected in bulk tank milk in multiple studies [164, 165, 166, 167]. These authors found that the number of foodborne pathogens detected in bulk tank milk vary with location, management practices, hygiene, and number of animals on the farm [165]. Similarly, a study on bulk tank milk from east Tennessee and southwest Virginia by Rohrbach et al. [168] showed that 32.5% of the samples analyzed contained one or more foodborne pathogens. Even dairy producers who used proper hygienic milking practices, pre- and post-milking teat disinfectant and antibiotic dry cow therapy, had foodborne pathogens in their bulk tank milk [164]. The isolation of these foodborne pathogens from bulk tank milk samples across the United States demonstrate the threat that mastitis pathogens and zoonotic mastitis causing pathogens create on public health if raw milk is consumed or if these pathogens make it through processing.

Advertisement

7. Conclusions

Bovine mastitis is the most important multifactorial disease of dairy cattle throughout the world. Mastitis is responsible for huge economic losses to the dairy producers and milk processing industry due to reduced milk production, alterations in milk composition, discarded milk, increased replacement costs, extra labor, treatment costs, and veterinary services. Many factors including pathogen, host, and environment can influence the development of mastitis. Mastitis, the inflammation of the mammary gland is usually a consequence of adhesion, invasion, and colonization of the mammary gland by one or more mastitis pathogens such as Staphylococcus aureus, Streptococcus uberis, and Escherichia coli.

References

  1. 1. Petrovski K, Trajcev M, Buneski G. A review of the factors affecting the costs of bovine mastitis. Journal of the South African Veterinary Association. 2006;77:52-60
  2. 2. NMC. The Cost of Mastitis: Dairy Insight Research 2005/2006: Final report. 2005
  3. 3. Ismail ZB. Mastitis vaccines in dairy cows: Recent developments and recommendations of application. Veterinary world. 2017;10:1057
  4. 4. Bradley AJ. Bovine mastitis: An evolving disease. The Veterinary Journal. 2002;164:116-128
  5. 5. Calvinho LF, Oliver SP. Invasion and persistence of streptococcus dysgalactiae within bovine mammary epithelial cells. Journal of Dairy Science. 1998;81:678-686
  6. 6. Becker K, Heilmann C, Peters G. Coagulase-negative staphylococci. Clinical Microbiology Reviews. 2014;27:870-926
  7. 7. De Vliegher S, Fox LK, Piepers S, McDougall S, Barkema HW. Invited review: Mastitis in dairy heifers: Nature of the disease, potential impact, prevention, and control. Journal of Dairy Science. 2012;95:1025-1040
  8. 8. Piessens V, Van Coillie E, Verbist B, Supre K, Braem G, Van Nuffel A, et al. Distribution of coagulase-negative staphylococcus species from milk and environment of dairy cows differs between herds. Journal of Dairy Science. 2011;94:2933-2944
  9. 9. Blowey RW. Mastitis Control in Dairy Herds. 2nd ed. Cambridge, Mass, MA: CABI; 2010
  10. 10. Cameron M, Saab M, Heider L, McClure JT, Rodriguez-Lecompte JC, Sanchez J. Antimicrobial susceptibility patterns of environmental streptococci recovered from bovine milk samples in the maritime provinces of Canada. Front Vet Sci. 2016;3:79
  11. 11. Bobbo T, Ruegg PL, Stocco G, Fiore E, Gianesella M, Morgante M, et al. Associations between pathogen-specific cases of subclinical mastitis and milk yield, quality, protein composition, and cheese-making traits in dairy cows. Journal of Dairy Science. 2017;100:4868-4883
  12. 12. Barkema HW, Green MJ, Bradley AJ, Zadoks RN. Invited review: The role of contagious disease in udder health. Journal of Dairy Science. 2009;92:4717-4729
  13. 13. Oliver S, Mitchell B. Prevalence of mastitis pathogens in herds participating in a mastitis control program1. Journal of Dairy Science. 1984;67:2436-2440
  14. 14. Smith KL, Todhunter D, Schoenberger P. Environmental mastitis: Cause, prevalence, prevention1, 2. Journal of Dairy Science. 1985;68:1531-1553
  15. 15. Bramley AJ. Sources of streptococcus uberis in the dairy herd: I. Isolation from bovine faces and from straw bedding of cattle. Journal of Dairy Research. 1982;49:369-373
  16. 16. Zadoks RN, Gillespie BE, Barkema HW, Sampimon OC, Oliver SP, Schukken YH. Clinical, epidemiological and molecular characteristics of Streptococcus uberis infections in dairy herds. Epidemiology and Infection. 2003;130:335-349
  17. 17. Almeida R, Oliver S. Antiphagocytic effect of the capsule of Streptococcus uberis. Zoonoses and Public Health. 1993;40:707-714
  18. 18. Oliver S, Almeida R, Calvinho L. Virulence factors of Streptococcus uberis isolated from cows with mastitis. Zoonoses and Public Health. 1998;45:461-471
  19. 19. Matthews K, Almeida R, Oliver S. Bovine mammary epithelial cell invasion by Streptococcus uberis. Infection and Immunity. 1994;62:5641-5646
  20. 20. Almeida RA, Kerro Dego O, Headrick SI, Lewis MJ, Oliver SP. Role of Streptococcus uberis adhesion molecule in the pathogenesis of Streptococcus uberis mastitis. Veterinary Microbiology. 2015;179:332-335
  21. 21. Almeida RA, Fang W, Oliver SP. Adherence and internalization of Streptococcus uberis to bovine mammary epithelial cells are mediated by host cell proteoglycans. FEMS Microbiology Letters. 1999;177:313-317
  22. 22. Patel D, Almeida RA, Dunlap JR, Oliver SP. Bovine lactoferrin serves as a molecular bridge for internalization of Streptococcus uberis into bovine mammary epithelial cells. Veterinary Microbiology. 2009;137:297-301
  23. 23. Fang W, Oliver SP. Identification of lactoferrin-binding proteins in bovine mastitis-causing Streptococcus uberis. FEMS Microbiology Letters. 1999;176:91-96
  24. 24. Almeida RA, Luther DA, Park HM, Oliver SP. Identification, isolation, and partial characterization of a novel Streptococcus uberis adhesion molecule (SUAM). Veterinary Microbiology. 2006;115:183-191
  25. 25. Vanderhaeghen W, Piepers S, Leroy F, Van Coillie E, Haesebrouck F, De Vliegher S. Invited review: Effect, persistence, and virulence of coagulase-negative Staphylococcus species associated with ruminant udder health. Journal of Dairy Science. 2014;97:5275-5293
  26. 26. Taponen S, Pyorala S. Coagulase-negative staphylococci as cause of bovine mastitis- not so different from Staphylococcus aureus? Veterinary Microbiology. 2009;134:29-36
  27. 27. Nyman AK, Fasth C, Waller KP. Intramammary infections with different non-aureus staphylococci in dairy cows. Journal of Dairy Science. 2018;101:1403-1418
  28. 28. De Vliegher S, Opsomer G, Vanrolleghem A, Devriese L, Sampimon O, Sol J, et al. In vitro growth inhibition of major mastitis pathogens by Staphylococcus chromogenes originating from teat apices of dairy heifers. Veterinary Microbiology. 2004;101:215-221
  29. 29. Gillespie BE, Headrick SI, Boonyayatra S, Oliver SP. Prevalence and persistence of coagulase-negative Staphylococcus species in three dairy research herds. Veterinary Microbiology. 2009;134:65-72
  30. 30. Taponen S, Bjorkroth J, Pyorala S. Coagulase-negative staphylococci isolated from bovine extramammary sites and intramammary infections in a single dairy herd. The Journal of Dairy Research. 2008;75:422-429
  31. 31. Fry PR, Middleton JR, Dufour S, Perry J, Scholl D, Dohoo I. Association of coagulase-negative staphylococcal species, mammary quarter milk somatic cell count, and persistence of intramammary infection in dairy cattle. Journal of Dairy Science. 2014;97:4876-4885
  32. 32. Woodward W, Besser T, Ward A, Corbeil L. In vitro growth inhibition of mastitis pathogens by bovine teat skin normal flora. Canadian Journal of Veterinary Research. 1987;51:27
  33. 33. Levison L, Miller-Cushon E, Tucker A, Bergeron R, Leslie K, Barkema H, et al. Incidence rate of pathogen-specific clinical mastitis on conventional and organic Canadian dairy farms. Journal of Dairy Science. 2016;99:1341-1350
  34. 34. Pyorala S, Taponen S. Coagulase-negative staphylococci-emerging mastitis pathogens. Veterinary Microbiology. 2009;134:3-8
  35. 35. Taponen S, Koort J, Bjorkroth J, Saloniemi H, Pyorala S. Bovine intramammary infections caused by coagulase-negative staphylococci may persist throughout lactation according to amplified fragment length polymorphism-based analysis. Journal of Dairy Science. 2007;90:3301-3307
  36. 36. Taponen S, Liski E, Heikkila AM, Pyorala S. Factors associated with intramammary infection in dairy cows caused by coagulase-negative staphylococci, Staphylococcus aureus, Streptococcus uberis, Streptococcus dysgalactiae, Corynebacterium bovis, or Escherichia coli. Journal of Dairy Science. 2017;100:493-503
  37. 37. Hogan J, Larry SK. Coliform mastitis. Veterinary Research. 2003;34:507-519
  38. 38. Botrel MA, Haenni M, Morignat E, Sulpice P, Madec JY, Calavas D. Distribution and antimicrobial resistance of clinical and subclinical mastitis pathogens in dairy cows in Rhone-Alpes, France. Foodborne Pathogens and Disease. 2010;7:479-487
  39. 39. Bradley AJ, Leach KA, Breen JE, Green LE, Green MJ. Survey of the incidence and aetiology of mastitis on dairy farms in England and Wales. The Veterinary Record. 2007;160:253-257
  40. 40. Burvenich C, Van Merris V, Mehrzad J, Diez-Fraile A, Duchateau L. Severity of E. coli mastitis is mainly determined by cow factors. Veterinary Research. 2003;34:521-564
  41. 41. Blum SE, Goldstone RJ, Connolly JPR, Reperant-Ferter M, Germon P, Inglis NF, et al. Postgenomics characterization of an essential genetic determinant of mammary pathogenic Escherichia coli. MBio. 2018;9(2):e00423-18
  42. 42. Petzl W, Zerbe H, Gunther J, Seyfert HM, Hussen J, Schuberth HJ. Pathogen-specific responses in the bovine udder. Models and immunoprophylactic concepts. Research in Veterinary Science. 2018;116:55-61
  43. 43. Petzl W, Zerbe H, Gunther J, Yang W, Seyfert HM, Nurnberg G, et al. Escherichia coli, but not Staphylococcus aureus triggers an early increased expression of factors contributing to the innate immune defense in the udder of the cow. Veterinary Research. 2008;39:18
  44. 44. Zhao Y, Zhou M, Gao Y, Liu H, Yang W, Yue J, et al. Shifted T helper cell polarization in a murine Staphylococcus aureus mastitis model. PLoS One. 2015;10:e0134797
  45. 45. Porcherie A, Gilbert FB, Germon P, Cunha P, Trotereau A, Rossignol C, et al. IL-17A is an important effector of the immune response of the mammary gland to Escherichia coli infection. Journal of Immunology. 2016;196:803-812
  46. 46. Herry V, Gitton C, Tabouret G, Reperant M, Forge L, Tasca C, et al. Local immunization impacts the response of dairy cows to Escherichia coli mastitis. Scientific Reports. 2017;7:3441
  47. 47. USDA APHIS U. Antibiotic Use on U.S. Dairy Operations, 2002 and 2007 (infosheet, 5p, October, 2008) [Online]. 2008a. Available from: https://www.aphis.usda.gov/animal_health/nahms/dairy/downloads/dairy07/Dairy07_is_AntibioticUse_1.pdf [Accessed: 23 March 2020]
  48. 48. USDA APHIS U. United States Department of Agriculture, Animal Plant Health Inspection Service National Animal Health Monitoring System. Highlights of Dairy 2007 Part III: Reference of dairy cattle health and management practices in the United States, 2007 (info sheet 4p, October, 2008) [Online]. 2008b. Available from: https://www.aphis.usda.gov/animal_health/nahms/dairy/downloads/dairy07/Dairy07_ir_Food_safety.pdf [Accessed: 23 March 2020]
  49. 49. Takahashi T, Satoh I, Kikuchi N. Phylogenetic relationships of 38 taxa of the genus Staphylococcus based on 16S rRNA gene sequence analysis. International Journal of Systematic Bacteriology. 1999;49(Pt 2):725-728
  50. 50. Fox LK, Hancock DD. Effect of segregation on prevention of intramammary infections by Staphylococcus aureus. Journal of Dairy Science. 1989;72:540-544
  51. 51. van Leeuwen WB, Melles DC, Alaidan A, Al-Ahdal M, Boelens HA, Snijders SV, et al. Host-and tissue-specific pathogenic traits of Staphylococcus aureus. Journal of Bacteriology. 2005;187:4584-4591
  52. 52. McMillan K, Moore SC, McAuley CM, Fegan N, Fox EM. Characterization of Staphylococcus aureus isolates from raw milk sources in Victoria, Australia. BMC Microbiology. 2016;16:169
  53. 53. Graber HU, Naskova J, Studer E, Kaufmann T, Kirchhofer M, Brechbuhl M, et al. Mastitis-related subtypes of bovine Staphylococcus aureus are characterized by different clinical properties. Journal of Dairy Science. 2009;92:1442-1451
  54. 54. Capurro A, Aspan A, Artursson K, Waller KP. Genotypic variation among Staphylococcus aureus isolates from cases of clinical mastitis in Swedish dairy cows. Veterinary Journal. 2010;185:188-192
  55. 55. Anderson KL, Lyman RL. Long-term persistence of specific genetic types of mastitis-causing Staphylococcus aureus on three dairies. Journal of Dairy Science. 2006;89:4551-4556
  56. 56. Simoons-Smit A, Savelkoul P, Stoof J, Starink T, Vandenbroucke-Grauls C. Transmission of Staphylococcus aureus between humans and domestic animals in a household. European Journal of Clinical Microbiology and Infectious Diseases. 2000;19:150-152
  57. 57. Rodgers JD, McCullagh JJ, McNamee PT, Smyth JA, Ball HJ. Comparison of Staphylococcus aureus recovered from personnel in a poultry hatchery and in broiler parent farms with those isolated from skeletal disease in broilers. Veterinary Microbiology. 1999;69:189-198
  58. 58. Zadoks R, Van Leeuwen W, Kreft D, Fox L, Barkema H, Schukken Y, et al. Comparison of Staphylococcus aureus isolates from bovine and human skin, milking equipment, and bovine milk by phage typing, pulsed-field gel electrophoresis, and binary typing. Journal of Clinical Microbiology. 2002;40:3894-3902
  59. 59. 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. Veterinary Microbiology. 2007;125:128-140
  60. 60. Türkyılmaz S, Tekbıyık S, Oryasin E, Bozdogan B. Molecular epidemiology and antimicrobial resistance mechanisms of methicillin-resistant Staphylococcus aureus isolated from bovine milk. Zoonoses and Public Health. 2010;57:197-203
  61. 61. Pantosti A, Sanchini A, Monaco M. Mechanisms of antibiotic resistance in Staphylococcus aureus. Future Microbiology. 2007;2:323-334
  62. 62. USDA APHIS U. Part III: Health Management and Biosecurity in US Feedlots, 1999. US Department of Agriculture [Online]. 2000. Available from: https://www.aphis.usda.gov/animal_health/nahms/dairy/downloads/dairy07/Dairy07_ir_Food_safety.pdf [Accessed: 23 December 2020]
  63. 63. Neave F, Dodd F, Kingwill R, Westgarth D. Control of mastitis in the dairy herd by hygiene and management. Journal of Dairy Science. 1969;52:696-707
  64. 64. Hillerton J, Berry E. Treating mastitis in the cow a tradition or an archaism. Journal of Applied Microbiology. 2005;98:1250-1255
  65. 65. Sischo W, Heider LE, Miller G, Moore D. Prevalence of contagious pathogens of bovine mastitis and use of mastitis control practices. Journal of the American Veterinary Medical Association. 1993;202:595-600
  66. 66. McCarthy AJ, Lindsay JA. Genetic variation in Staphylococcus aureus surface and immune evasion genes is lineage associated: Implications for vaccine design and host-pathogen interactions. BMC Microbiology. 2010;10:173
  67. 67. Brady RA, Leid JG, Camper AK, Costerton JW, Shirtliff ME. Identification of Staphylococcus aureus proteins recognized by the antibody-mediated immune response to a biofilm infection. Infection and Immunity. 2006;74:3415-3426
  68. 68. den Reijer PM, Sandker M, Snijders SV, Tavakol M, Hendrickx AP, van Wamel WJ. Combining in vitro protein detection and in vivo antibody detection identifies potential vaccine targets against Staphylococcus aureus during osteomyelitis. Medical Microbiology and Immunology. 2017;206:11-22
  69. 69. Clarke SR, Foster SJ. Surface adhesins of Staphylococcus aureus. Advances in Microbial Physiology. 2006;51:187-224
  70. 70. Hauck CR, Ohlsen K. Sticky connections: Extracellular matrix protein recognition and integrin-mediated cellular invasion by Staphylococcus aureus. Current Opinion in Microbiology. 2006;9:5-11
  71. 71. Speziale P, Pietrocola G, Rindi S, Provenzano M, Provenza G, Di Poto A, et al. Structural and functional role of Staphylococcus aureus surface components recognizing adhesive matrix molecules of the host. Future Microbiology. 2009;4:1337-1352
  72. 72. Burke FM, McCormack N, Rindi S, Speziale P, Foster TJ. Fibronectin-binding protein B variation in Staphylococcus aureus. BMC Microbiology. 2010;10:160
  73. 73. Zecconi A, Scali F. Staphylococcus aureus virulence factors in evasion from innate immune defenses in human and animal diseases. Immunology Letters. 2013;150:12-22
  74. 74. Camussone CM, Calvinho LF. Virulence factors of Staphylococcus aureus associated with intramammary infections in cows: Relevance and role as immunogens. Revista Argentina de Microbiología. 2013;45:119-130
  75. 75. Gotz F. Staphylococcus and biofilms. Molecular Microbiology. 2002;43:1367-1378
  76. 76. Otto M. Staphylococcal biofilms. Current Topics in Microbiology and Immunology. 2008;322:207-228
  77. 77. Donlan RM, Costerton JW. Biofilms: Survival mechanisms of clinically relevant microorganisms. Clinical Microbiology Reviews. 2002;15:167-193
  78. 78. Stewart PS, Costerton JW. Antibiotic resistance of bacteria in biofilms. Lancet. 2001;358:135-138
  79. 79. Foster TJ, Geoghegan JA, Ganesh VK, Hook M. Adhesion, invasion and evasion: The many functions of the surface proteins of Staphylococcus aureus. Nature Reviews. Microbiology. 2014;12:49-62
  80. 80. Szweda P, Schielmann M, Milewski S, Frankowska A, Jakubczak A. Biofilm production and presence of Ica and bap genes in Staphylococcus aureus strains isolated from cows with mastitis in the eastern Poland. Polish Journal of Microbiology. 2012;61:65-69
  81. 81. Cucarella C, Solano C, Valle J, Amorena B, Lasa I, Penades JR. Bap, a Staphylococcus aureus surface protein involved in biofilm formation. Journal of Bacteriology. 2001;183:2888-2896
  82. 82. Lasa I, Penadés JR. Bap: A family of surface proteins involved in biofilm formation. Research in Microbiology. 2006;157:99-107
  83. 83. Valle J, Latasa C, Gil C, Toledo-Arana A, Solano C, Penadés JR, et al. Bap, a biofilm matrix protein of Staphylococcus aureus prevents cellular internalization through binding to GP96 host receptor. PLoS Pathogens. 2012;8:e1002843
  84. 84. Cucarella C, Tormo MÁ, Ubeda C, Trotonda MP, Monzón M, Peris C, et al. Role of biofilm-associated protein bap in the pathogenesis of bovine Staphylococcus aureus. Infection and Immunity. 2004;72:2177-2185
  85. 85. Milanov D, Lazić S, Vidić B, Petrović J, Bugarski D, Šeguljev Z. Slime production and biofilm forming ability by Staphylococcus aureus bovine mastitis isolates. Acta Veterinaria. 2010;60:217-226
  86. 86. Melchior MB, Vaarkamp H, Fink-Gremmels J. Biofilms: A role in recurrent mastitis infections? Veterinary Journal. 2006;171:398-407
  87. 87. Dhanawade NB, Kalorey DR, Srinivasan R, Barbuddhe SB, Kurkure NV. Detection of intercellular adhesion genes and biofilm production in Staphylococcus aureus isolated from bovine subclinical mastitis. Veterinary Research Communications. 2010;34:81-89
  88. 88. Vasudevan P, Nair MKM, Annamalai T, Venkitanarayanan KS. Phenotypic and genotypic characterization of bovine mastitis isolates of Staphylococcus aureus for biofilm formation. Veterinary Microbiology. 2003;92:179-185
  89. 89. O’Gara JP. Ica and beyond: Biofilm mechanisms and regulation in Staphylococcus epidermidis and Staphylococcus aureus. FEMS Microbiology Letters. 2007;270:179-188
  90. 90. Otto M. Staphylococcal infections: Mechanisms of biofilm maturation and detachment as critical determinants of pathogenicity. Annual Review of Medicine. 2013;64:175-188
  91. 91. Aydin A, Sudagidan M, Muratoglu K. Prevalence of staphylococcal enterotoxins, toxin genes and genetic-relatedness of foodborne Staphylococcus aureus strains isolated in the Marmara region of Turkey. International Journal of Food Microbiology. 2011;148:99-106
  92. 92. Rogolsky M. Nonenteric toxins of Staphylococcus aureus. Microbiological Reviews. 1979;43:320-360
  93. 93. Wadstrom T. Biological properties of extracellular proteins from Staphylococcus. Annals of the New York Academy of Sciences. 1974;236:343-361
  94. 94. 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. Infection and Immunity. 1989;57:2489-2494
  95. 95. Berube BJ, Bubeck WJ. Staphylococcus aureus alpha-toxin: Nearly a century of intrigue. Toxins (Basel). 2013;5:1140-1166
  96. 96. Otto M. Staphylococcus aureus toxins. Current Opinion in Microbiology. 2014;17:32-37
  97. 97. Songer JG, Post KW. Veterinary Microbiology-E-Book: Bacterial and Fungal Agents of Animal Disease. St. Louis, Missouri: Elsevier Health Sciences; 2004
  98. 98. Kerro Dego O, Nederbragt H. Factors involved in the early pathogenesis of bovine Staphylococcus aureus mastitis with emphasis on bacterial adhesion and invasion. A review. The veterinary quarterly. 2002;24:181-198
  99. 99. Sutra L, Poutrel B. Virulence factors involved in the pathogenesis of bovine intramammary infections due to Staphylococcus aureus. Journal of Medical Microbiology. 1994;40:79-89
  100. 100. Caiazza NC, O’toole G. Alpha-toxin is required for biofilm formation by Staphylococcus aureus. Journal of Bacteriology. 2003;185:3214-3217
  101. 101. Low DKR, Freer JH. Biological effects of highly purified B-lysin (sphingomyelinase) from S. aureus. FEMS letters. 1977;2:133-138
  102. 102. O’Callaghan RJ, Callegan MC, Moreau JM, Green LC, Foster TJ, Hartford OM, et al. Specific roles of alpha-toxin and beta-toxin during Staphylococcus aureus corneal infection. Infection and Immunity. 1997;65:1571-1578
  103. 103. Essmann F, Bantel H, Totzke G, Engels IH, Sinha B, Schulze-Osthoff K, et al. Staphylococcus aureus alpha-toxin-induced cell death: Predominant necrosis despite apoptotic caspase activation. Cell Death and Differentiation. 2003;10:1260-1272
  104. 104. Smyth CJ, Mollby R, Wadstrom T. Phenomenon of hot-cold hemolysis: Chelator-induced lysis of sphingomyelinase-treated erythrocytes. Infection and Immunity. 1975;12:1104-1111
  105. 105. Dinges MM, Orwin PM, Schlievert PM. Exotoxins of Staphylococcus aureus. Clinical Microbiology Reviews. 2000;13:16-34
  106. 106. Prevost G, Couppie P, Prevost P, Gayet S, Petiau P, Cribier B, et al. Epidemiological data on Staphylococcus aureus strains producing synergohymenotropic toxins. Journal of Medical Microbiology. 1995;42:237-245
  107. 107. Srinivasan V, Sawant AA, Gillespie BE, Headrick SJ, Ceasaris L, Oliver SP. Prevalence of enterotoxin and toxic shock syndrome toxin genes in Staphylococcus aureus isolated from milk of cows with mastitis. Foodborne Pathogens and Disease. 2006;3:274-283
  108. 108. Matsunaga T, Kamata S-I, Kakiichi N, Uchida K. Characteristics of Staphylococcus aureus isolated from peracute, acute and chronic bovine mastitis. Journal of Veterinary Medical Science. 1993;55:297-300
  109. 109. Hennekinne J-A, De Buyser M-L, Dragacci S. Staphylococcus aureus and its food poisoning toxins: Characterization and outbreak investigation. FEMS Microbiology Reviews. July 2012;36(4):815-836
  110. 110. Bergdoll MS, Chu FS, Borja CR, Huang I-Y, Weiss KF. The staphylococcal enterotoxins. Japanese Journal of Microbiology. 1967;11:358-368
  111. 111. Kong C, H-m N, Nathan S. Targeting Staphylococcus aureus toxins: A potential form of anti-virulence therapy. Toxins. 2016;8:72
  112. 112. Kenny K, Reiser RF, Bastida-Corcuera FD, Norcross NL. Production of enterotoxins and toxic shock syndrome toxin by bovine mammary isolates of Staphylococcus aureus. Journal of Clinical Microbiology. 1993;31:706-707
  113. 113. Todd J, Fishaut M, Kapral F, Welch T. Toxic-shock syndrome associated with phage-group-I staphylococci. The Lancet. 1978;312:1116-1118
  114. 114. Moon J, Lee A, Kang H, Lee E, Joo Y, Park YH, et al. Antibiogram and coagulase diversity in staphylococcal enterotoxin-producing Staphylococcus aureus from bovine mastitis. Journal of Dairy Science. 2007;90:1716-1724
  115. 115. Stephan R, Annemüller C, Hassan A, Lämmler C. Characterization of enterotoxigenic Staphylococcus aureus strains isolated from bovine mastitis in north-East Switzerland. Veterinary Microbiology. 2001;78:373-382
  116. 116. Cenci-Goga B, Karama M, Rossitto P, Morgante R, Cullor J. Enterotoxin production by Staphylococcus aureus isolated from mastitic cows. Journal of Food Protection. 2003;66:1693-1696
  117. 117. Piccinini R, Borromeo V, Zecconi A. Relationship between S. aureus gene pattern and dairy herd mastitis prevalence. Veterinary Microbiology. 2010;145:100-105
  118. 118. Piechota M, Kot B, Zdunek E, Mitrus J, Wicha J, Wolska MK, et al. Distribution of classical enterotoxin genes in staphylococci from milk of cows with- and without mastitis and the cowshed environment. Polish Journal of Veterinary Sciences. 2014;17:407-411
  119. 119. Fisher EL, Otto M, Cheung GYC. Basis of virulence in enterotoxin-mediated staphylococcal food poisoning. Frontiers in Microbiology. 2018;9:436
  120. 120. Almeida RA, Matthews KR, Cifrian E, Guidry AJ, Oliver SP. Staphylococcus aureus invasion of bovine mammary epithelial cells. Journal of Dairy Science. 1996;79:1021-1026
  121. 121. 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 Microbiology Letters. 1999;180:103-109
  122. 122. Cifrian E, Guidry A, O’Brien C, Marquardt W. Effect of alpha-toxin and capsular exopolysaccharide on the adherence of Staphylococcus aureus to cultured teat, ductal and secretory mammary epithelial cells. Research in Veterinary Science. 1995;58:20-25
  123. 123. Hensen S, Pavičić M, Lohuis J, Poutrel B. Use of bovine primary mammary epithelial cells for the comparison of adherence and invasion ability of Staphylococcus aureus strains. Journal of Dairy Science. 2000;83:418-429
  124. 124. Josse J, Laurent F, Diot A. Staphylococcal adhesion and host cell invasion: Fibronectin-binding and other mechanisms. Frontiers in Microbiology. 2017;8:2433
  125. 125. Loffler B, Tuchscherr L, Niemann S, Peters G. Staphylococcus aureus persistence in non-professional phagocytes. International Journal of Medical Microbiology. 2014;304:170-176
  126. 126. Moormeier DE, Bayles KW. Staphylococcus aureus biofilm: A complex developmental organism. Molecular Microbiology. 2017;104:365-376
  127. 127. Fowler T, Wann ER, Joh D, Johansson S, Foster TJ, Hook M. Cellular invasion by Staphylococcus aureus involves a fibronectin bridge between the bacterial fibronectin-binding MSCRAMMs and host cell beta-1 integrins. European Journal of Cell Biology. 2000;79:672-679
  128. 128. Abdi RD, Gillespie BE, Vaughn J, Merrill C, Headrick SI, Ensermu DB, et al. Antimicrobial resistance of Staphylococcus aureus isolates from dairy cows and genetic diversity of resistant isolates. Foodborne Pathogens and Disease. 2018;15:449-458
  129. 129. Tyler JW, Wilson RC, Dowling P. Treatment of subclinical mastitis. The Veterinary Clinics of North America. Food Animal Practice. 1992;8:17-28
  130. 130. Brüssow H, Canchaya C, Hardt W-D. Phages and the evolution of bacterial pathogens: From genomic rearrangements to lysogenic conversion. Microbiology and Molecular Biology Reviews. 2004;68:560-602
  131. 131. Owens W, Ray C, Watts J, Yancey R. Comparison of success of antibiotic therapy during lactation and results of antimicrobial susceptibility tests for bovine mastitis. Journal of Dairy Science. 1997;80:313-317
  132. 132. Carter E, Kerr D. Optimization of DNA-based vaccination in cows using green fluorescent protein and protein a as a prelude to immunization against staphylococcal mastitis. Journal of Dairy Science. 2003;86:1177-1186
  133. 133. Emaneini M, Jabalameli F, Abani S, Dabiri H, Beigverdi R. Comparison of virulence factors and capsular types of Streptococcus agalactiae isolated from human and bovine infections. Microbial Pathogenesis. 2016;91:1-4
  134. 134. Farnsworth R. Indications of Contagious and Environmental Mastitis Pathogens in a Dairy Herd. USA: Annual Meeting of National Mastitis Council; 1987
  135. 135. Hillerton JE, Berry EA. The management and treatment of environmental streptococcal mastitis. Veterinary Clinics of North America: Food Animal Practice. 2003;19:157-169
  136. 136. Nicholas R, Ayling R, McAuliffe L. Mycoplasma mastitis. Veterinary Record. 2007;160:382-382
  137. 137. USDA APHIS U. United States Department of Agriculture, Animal Plant Health Inspection Service National Animal Health Monitoring System. Injection Practices on U.S. Dairy Operations, 2007 (Veterinary Services Info Sheet 4 p, February 2009) [Online]. 2009. Available from https://www.aphis.usda.gov/animal_health/nahms/dairy/downloads/dairy07/Dairy07_is_InjectionPrac_1.pdf [Accessed: 23 March 2020]
  138. 138. Fox LK. Mycoplasma mastitis: Causes, transmission, and control. Veterinary Clinics: Food Animal Practice. 2012;28:225-237
  139. 139. Jasper DE. Bovine mycoplasmal mastitis. Advances in Veterinary Science and Comparative Medicine. 1981;25:121-157
  140. 140. Dogan B, Klaessig S, Rishniw M, Almeida R, Oliver S, Simpson K, et al. Adherent and invasive Escherichia coli are associated with persistent bovine mastitis. Veterinary Microbiology. 2006;116:270-282
  141. 141. Almeida RA, Dogan B, Klaessing S, Schukken YH, Oliver SP. Intracellular fate of strains of Escherichia coli isolated from dairy cows with acute or chronic mastitis. Veterinary Research Communications. 2011;35:89-101
  142. 142. Bayles KW, Wesson CA, Liou LE, Fox LK, Bohach GA, Trumble W. Intracellular Staphylococcus aureus escapes the endosome and induces apoptosis in epithelial cells. Infection and Immunity. 1998;66:336-342
  143. 143. Craven N, Anderson JC. Phagocytosis of Staphylococcus aureus by bovine mammary gland macrophages and intracellular protection from antibiotic action in vitro and in vivo. The Journal of Dairy Research. 1984;51:513-523
  144. 144. Zhao S, Gao Y, Xia X, Che Y, Wang Y, Liu H, et al. TGF-β1 promotes Staphylococcus aureus adhesion to and invasion into bovine mammary fibroblasts via the ERK pathway. Microbial Pathogenesis. 2017;106:25-29
  145. 145. Perez-Casal J, Prysliak T, Kerro Dego O, Potter AA. Immune responses to a Staphylococcus aureus GapC/B chimera and its potential use as a component of a vaccine for S. aureus mastitis. Veterinary Immunology and Immunopathology. 2006;109:85-97
  146. 146. Gudding R, McDonald J, Cheville N. Pathogenesis of Staphylococcus aureus mastitis: Bacteriologic, histologic, and ultrastructural pathologic findings. American Journal of Veterinary Research. 1984;45:2525-2531
  147. 147. Zecconi A, Cesaris L, Liandris E, Dapra V, Piccinini R. Role of several Staphylococcus aureus virulence factors on the inflammatory response in bovine mammary gland. Microbial Pathogenesis. 2006;40:177-183
  148. 148. Harmon R. Physiology of mastitis and factors affecting somatic cell counts1. Journal of Dairy Science. 1994;77:2103-2112
  149. 149. Keefe G. Update on control of Staphylococcus aureus and Streptococcus agalactiae for management of mastitis. The Veterinary Clinics of North America. Food Animal Practice. 2012;28:203-216
  150. 150. Zecconi A. Staphylococcus aureus mastitis: What we need to know to con. Israel Journal of Veterinary Medicine. 2010;65:93-99
  151. 151. Malek dos Reis CB, Barreiro JR, Mestieri L, MADF P, dos Santos MV. Effect of somatic cell count and mastitis pathogens on milk composition in Gyr cows. BMC Veterinary Research. 2013;9:67
  152. 152. Akers RM, Nickerson SC. Mastitis and its impact on structure and function in the ruminant mammary gland. Journal of Mammary Gland Biology and Neoplasia. 2011;16:275-289
  153. 153. Sordillo LM, Streicher KL. Mammary gland immunity and mastitis susceptibility. Journal of Mammary Gland Biology and Neoplasia. 2002;7:135-146
  154. 154. Hogan J, Smith K. 1987. A practical look at environmental mastitis. The compendium on continuing education for the practicing veterinarian (USA).
  155. 155. Gibson H, Sinclair LA, Brizuela CM, Worton HL, Protheroe RG. Effectiveness of selected premilking teat-cleaning regimes in reducing teat microbial load on commercial dairy farms. Letters in Applied Microbiology. 2008;46:295-300
  156. 156. Gleeson D, O’Brien B, Flynn J, O’Callaghan E, Galli F. Effect of pre-milking teat preparation procedures on the microbial count on teats prior to cluster application. Irish Veterinary Journal. 2009;62:461-467
  157. 157. Dufour S, Frechette A, Barkema HW, Mussell A, Scholl DT. Invited review: Effect of udder health management practices on herd somatic cell count. Journal of Dairy Science. 2011;94:563-579
  158. 158. Mordak R, Stewart PA. Periparturient stress and immune suppression as a potential cause of retained placenta in highly productive dairy cows: Examples of prevention. Acta Veterinaria Scandinavica. 2015;57:84
  159. 159. Drackley JK. ADSA foundation scholar award. Biology of dairy cows during the transition period: The final frontier? Journal of Dairy Science. 1999;82:2259-2273
  160. 160. Esposito G, Irons PC, Webb EC, Chapwanya A. Interactions between negative energy balance, metabolic diseases, uterine health and immune response in transition dairy cows. Animal Reproduction Science. 2014;144:60-71
  161. 161. Bach A. Associations between several aspects of heifer development and dairy cow survivability to second lactation. Journal of Dairy Science. 2011;94:1052-1057
  162. 162. Oliver SP, Jayarao BM, Almeida RA. Foodborne pathogens, mastitis, milk quality, and dairy food safety. In: Proceedings of National Mastitis Council (NMC), 44th meeting, January 16-19. Orlando, FL; 2005. pp. 3-27
  163. 163. Scallan E, Hoekstra RM, Angulo FJ, Tauxe RV, Widdowson M-A, Roy SL, et al. Foodborne illness acquired in the United States—Major pathogens. Emerging Infectious Diseases. 2011;17:7-15
  164. 164. Jayarao BM, Henning DR. Prevalence of foodborne pathogens in bulk tank milk1. Journal of Dairy Science. 2001;84:2157-2162
  165. 165. Gillespie BE, Oliver SP. Simultaneous detection of mastitis pathogens, Staphylococcus aureus, Streptococcus uberis, and Streptococcus agalactiae by multiplex real-time polymerase chain reaction. Journal of Dairy Science. 2005;88:3510-3518
  166. 166. Steele ML, Mcnab WB, Poppe C, Griffiths MW, Chen S, Degrandis SA, et al. Survey of Ontario bulk tank raw Milk for food-borne pathogens. Journal of Food Protection. 1997;60:1341-1346
  167. 167. Van Kessel J, Karns J, Gorski L, McCluskey B, Perdue M. Prevalence of Salmonellae, Listeria monocytogenes, and fecal coliforms in bulk tank milk on US dairies. Journal of Dairy Science. 2004;87:2822-2830
  168. 168. Rohrbach BW, Draughon FA, Davidson PM, Oliver SP. Prevalence of Listeria monocytogenes, Campylobacter jejuni, Yersinia enterocolitica, and Salmonella in bulk tank milk: Risk factors and risk of human exposure. Journal of Food Protection. 1992;55:93-97

Written By

Oudessa Kerro Dego

Submitted: 15 October 2019 Reviewed: 27 July 2020 Published: 02 September 2020