Vous êtes sur la page 1sur 9

Published December 5, 2014

Mammary tissue damage during bovine mastitis: Causes and control1

X. Zhao*2 and P. Lacasse†

*Department of Animal Science, McGill University, Ste. Anne de Bellevue, Québec, H9X 3V9, Canada;
and †Dairy and Swine Research and Development Centre, Agriculture and Agri-Food Canada,
PO Box 90 STN Lennoxville, Sherbrooke, Quebec, J1M 1Z3, Canada

ABSTRACT: Mastitis, an inflammatory reaction of whereas other bacteria are able to invade and multiply
the mammary gland that is usually caused by a micro- within the bovine mammary epithelial cells before caus-
bial infection, is recognized as the most costly disease ing cell death. In addition, mastitis is characterized by
in dairy cattle. Decreased milk production accounts for an influx of somatic cells, primarily polymorphonuclear
approximately 70% of the total cost of mastitis. Mam- neutrophils, into the mammary gland. With more im-
mary tissue damage reduces the number and activity mune cells migrating into the mammary gland and the
of epithelial cells and consequently contributes to de- breakdown of the blood-milk barrier, damage to the
creased milk production. Mammary tissue damage has mammary epithelium worsens. It is well known that
been shown to be induced by either apoptosis or necro- breakdown of the extracellular matrix can lead to death
sis. These 2 distinct types of cell death can be distin- of the epithelial cells. Meanwhile, polymorphonuclear
guished by morphological, biochemical, and molecular neutrophils can harm the mammary tissue by releasing
changes in dying cells. Both bacterial factors and host reactive oxygen intermediates and proteolytic enzymes.
immune reactions contribute to epithelial tissue dam- In vitro and in vivo studies suggest that the use of
age. During infection of the mammary glands, the tis- antioxidants and other protective compounds in masti-
sue damage can initially be caused by bacteria and their tis control programs is worth investigating, because
products. Certain bacteria produce toxins that destroy they may aid in alleviating damage to secretory cells
cell membranes and damage milk-producing tissue, and thus reduce subsequent milk loss.

Key words: bovine, mammary gland, mastitis, epithelial cell, apoptosis, tissue damage

©2008 American Society of Animal Science. All rights reserved. J. Anim. Sci. 2008. 86(Suppl. 1):57–65
doi:10.2527/jas.2007-0302

INTRODUCTION cows in given herds (Wilson et al., 1997; Pitkala et al.,


2004). The cost of subclinical mastitis is very difficult
Mastitis is defined as an inflammation of the mam- to quantify, but most experts agree that subclinical
mary gland. Mastitis usually occurs primarily in re- mastitis costs the average dairy farmer more than does
sponse to intramammary bacterial infection, but also clinical mastitis. Assuming a 45% prevalence of subclin-
to intramammary mycoplasmal, fungal, or algal infec- ical mastitis, the cost has been estimated in the range
tions. Mechanical trauma, thermal trauma, and chemi- of $180 to $320 per case (Wilson et al., 1997). Approxi-
cal insult predispose the gland to intramammary infec- mately 70% of this cost is associated with a reduction
tion (IMI). Occurrence of mastitis depends on the inter- in milk production. A large portion of it results from
irreversible damage to the mammary tissue (Oliver and
action of host, agent, and environmental factors.
Calvinho, 1995). Although antibiotics are very useful
Mastitis is the most costly infectious disease of dairy
to treat the infection, they do not directly protect the
cattle. The prevalence of mastitis in dairy cattle is rela-
gland from being damaged.
tively high. Subclinical mastitis is the main form of This review is a compilation of some major findings
mastitis in modern dairy herds, exceeding 20 to 50% of concerning tissue damage during mastitis. We describe
the current understanding of how bacteria, polymor-
phonuclear neutrophils (PMN), and host proteases and
1
cytokines contribute to tissue damage. Finally, research
Presented at the Eighth International Workshop on the Biology
aimed at the reduction of tissue damage is discussed.
of Lactation in Farm Animals held in Pirassununga, Brazil, August
21–23, 2006.
2
OVERVIEW OF MASTITIS
Corresponding author: xin.zhao@mcgill.ca
Received May 25, 2007. Bovine mammary glands are exposed to diverse bac-
Accepted August 20, 2007. teria throughout lactation and in nonlactating periods.

57
58 Zhao and Lacasse

Pathogens commonly isolated from mastitic milk can cium-dependent endonuclease (Arends et al., 1990). Ap-
be classified as noncontagious (most are environmental) pearance of this oligonucleosomal DNA laddering in
and contagious microorganisms. The former include stained gels is now widely used to detect apoptosis.
Streptococcus uberis, Streptococcus dysgalactiae, Esche- Enzymatic labeling of DNA strand breaks also allows
richia coli, and coagulase-negative staphylococcus spe- for histological detection of apoptosis in the tissue by a
cies, whereas the latter include Staphylococcus aureus terminal deoxynucleotidyl transferase-mediated dUTP
and Streptococcus agalactiae. The teat and streak canal nick-end labeling (TUNEL) assay. In addition, loss of
are the first line of defense of the mammary gland. The asymmetry in cell membrane phospholipids during
keratin lining in the streak canal provides a physical apoptosis allows Annexin-V staining to be used to detect
and chemical barrier against bacterial penetration (Ca- exposed phosphatidylserine as a marker for apoptosis.
puco et al., 1992). Bacteria may escape the natural Furthermore, specific caspases, specific factors in apop-
defense mechanisms by multiplication along the streak totic pathways, and cleavage of caspase substrates have
canal (especially after milking), or by propulsion into been explored to detect apoptosis. Unlike apoptosis, ne-
the teat cistern by vacuum fluctuations at the teat end crosis refers to the accidental death of cells in living
during milking. The infection occurs after bacteria gain tissue. Necrosis may follow a wide variety of injuries,
entrance to the mammary gland via the teat canal. both physical (e.g., cuts, burns, bruises) and biological
After bacteria overcome the anatomical defense, they (e.g., effects of disease-causing agents). The sign of ne-
must evade the cellular and humoral defense mecha- crosis, dead tissue, is called a lesion. It begins with
nisms of the mammary gland to establish disease (Sor- swelling of the cytoplasm and mitochondria caused by a
dillo and Streicher, 2002). If the infection is not elimi- loss of membrane integrity and ends with total cell lysis.
nated, bacterial levels in the mammary gland eventu- These apparently clear-cut definitions between
ally rise to a level at which they begin to damage the apoptosis and necrosis, however, are likely oversimpli-
mammary epithelium. As infection persists, the num- fied. For example, the apoptotic bodies as well as the
ber of somatic cells in milk continues to increase and, remaining cell fragments, if not quickly removed by
concomitantly, tissue damage is worsened. The alveoli phagocytes or neighboring cells, will ultimately swell
in the gland start to lose structural integrity and the and finally lyse. This process is called secondary necro-
blood-milk barrier is breached. This allows extracellu- sis. In addition, the positive or negative interconnection
lar fluid to enter the gland and mix with the milk. between autophagy and apoptosis (Gozuacik and Kim-
Visible changes in the milk and the udder start to occur. chi, 2004) adds additional complexity to apoptosis re-
These can include external swelling, reddening of the search.
gland, and clotting and wateriness of the milk. By defi- Cytotoxicity is also commonly used in the literature
nition, this is the start of clinical symptoms. In brief, to describe tissue damage, but does not infer a specific
bovine mammary epithelial cells can be damaged dur- cellular mechanism. Rather, it indicates the cell-killing
ing IMI by 1) release of a range of cellular and extracel- property of a chemical compound or a mediator cell.
lular products from bacterial pathogens; 2) lysosomal Many assays are available for measuring cytotoxicity,
enzymes and oxidative products released from phago- and most of them are based on increased plasma mem-
cytes during phagocytosis of invading organisms, and brane permeability or increased uptake of dyes by dying
3) proteases from blood and cytokines released during cells or dead cells. The results from cytotoxicity assays
the immune response. vary considerably depending on the method used (Fel-
lows and O’Donovan 2007). In general, cytotoxicity
APOPTOSIS AND NECROSIS assays can be used to detect necrosis and the late stage
of apoptosis, but not the early stage of apoptosis. Spe-
Multiple-assay methodologies and different terminol- cifically for bovine mammary studies, lactate dehydro-
ogies have been used by different research groups to genase activity and N-acetyl-β-D-glucosaminidase
study and describe tissue damage during mastitis. (NAGase) activity have commonly been used as mark-
Thus, a brief introduction to types of cell death is war- ers for tissue damage. The former enzyme is a stable
ranted. There are 2 distinct types of cell death, cytoplasmic enzyme present in all cells. It is rapidly
apoptosis and necrosis, which may be distinguished by released outside the cell when the plasma membrane
morphological, biochemical, and molecular changes in is damaged. Located within lysosomes of mammary epi-
dying cells. Apoptosis is a process of deliberate suicide thelial cells, NAGase is released from damaged mam-
of a cell in multicellular organisms. The process of mary epithelial cells (Kitchen et al., 1980). However,
apoptosis was originally distinguished from necrosis on a small fraction of milk NAGase can also come from
the basis of cellular ultrastructure (Kerr et al., 1972). leukocytes (Capuco et al., 1986).
Apoptosis may be identified by a characteristic pattern
of morphological changes, including nuclear and cyto- TISSUE DAMAGE: HISTOLOGICAL
plasmic condensation, nuclear fragmentation, and for- OBSERVATION
mation of apoptotic bodies (Strange et al., 1992). These
changes are associated with cleavage of chromatin into Histological analyses have been widely used since
discretely sized oligonucleosome fragments by a cal- the 1970s and are still being used today for assessing
Tissue damage and mastitis 59
damage to secretory tissue in the bovine mammary reported a massive PMN infiltration and necrosis of
gland caused by mastitis pathogens. For example, Be- secretory tissues. In addition, Heald (1979) observed
nites et al. (2002) examined the mammary parenchyma that mammary tissues from lactating cows inoculated
from 184 slaughtered dairy cows for the existence of with Staph. aureus exhibited less milk synthesis and
microorganisms and histopathological changes. Of all secretion, as evidenced by more interalveolar stroma
the samples from which microorganisms were isolated, and involuting alveolar epithelium and less alveolar
only 3.1% did not show histological changes. The re- luminal space compared with uninfected contralateral
maining 96.9% of samples showed an inflammatory re- controls. Moreover, these changes were associated with
sponse (i.e., edema, mammary epithelial cell damage, replacement of secretory tissue with nonsecretory tis-
and PMN infiltration), tissue repair process, or both. sue (Nickerson and Heald, 1981). Similarly, a study
In contrast, 33.9% of samples from glands without evi- with dry cows revealed that quarters with intramam-
dence of microorganisms (n = 56) showed no histological mary Staph. aureus infection had a greater degree of
changes. These results clearly indicate that the pres- mammary involution with a greater proportion of inter-
ence of microorganisms is associated with tissue alveolar stroma and a lesser proportion of alveolar lu-
damage. men compared with uninfected quarters (Sordillo and
Escherichia coli is one of the most important patho- Nickerson, 1988). Finally, a challenge study with heif-
gens causing mastitis in dairy cows (Bradley, 2002), ers by Trinidad et al. (1990) showed that mammary
resulting in inflammation that ranges from subacute to glands infected by Staph. aureus also exhibited fewer
peracute. Necrosis of the mammary epithelium occurs alveolar epithelial and luminal areas but more interal-
during severe, naturally occurring clinical E. coli masti- veolar stroma than controls. From these results, we are
tis, as well as during severe experimental E. coli masti- relatively confident in concluding that Staph. aureus
tis. In moderate cases of E. coli mastitis, there is mini- infection causes necrosis of the secretory tissues and
mal alveolar tissue damage, as shown by Frost et al. that the damaged secretory tissue is replaced with non-
(1980). In that experiment, the main changes were su- secretory tissue.
perficial and confined to the epithelium of the teat sinus,
lactiferous sinus, and large ducts, without serious APOPTOSIS DURING MASTITIS
involvement of the secretory tissue. There were some
lesions of the basement membrane underlying the dam- The induction of apoptosis by bacterial pathogens
aged epithelium but damage was localized. In very se- is a well-established cellular process (Weinrauch and
vere cases, the infection progressed via the ductile sys- Zychlinsky, 1999). Apoptosis is a key feature of mam-
tem to produce a limited inflammatory reaction but mary gland development and function. In addition,
with an extensive involvement of the secretory tissue apoptosis is critical for removing the milk-secreting al-
(Frost and Brooker, 1986). In its most severe form with veolar epithelial cells during lactation and postlacta-
uncontrolled bacterial multiplication, all lactiferous si- tional involution (Capuco and Akers, 1999; Capuco et
nus epithelia were lost, interstitial tissue became hem- al., 2003). However, conclusive and direct evidence for
orrhagic, and often the animal died of toxemia within involvement of apoptosis during mastitis has been pro-
a few hours of infection (Burvenich et al., 2003). The vided only by Long et al. (2001). Escherichia coli-in-
severity of the disease varies considerably from cow to fected mammary glands were biopsied, with the re-
cow. Kornalijnslijper et al. (2004) reported that 6 h after sulting tissues processed for RNA, protein, and histo-
intramammary inoculation, the bacterial counts, which logical examinations. Both mRNA and protein analyses
were significantly correlated with severity of the dis- indicated up-regulation of the proapoptotic factors Bax
ease, varied considerably (i.e., up to 800 fold). They and IL-1β-converting enzyme, and a down-regulation
concluded that bacterial growth in the phase before of the antiapoptotic factor Bcl-2. Further, induction of
massive influx of PMN is a major determinant in the a 92-kDa gelatinase was observed by gelatin zymogra-
final severity of experimental E. coli mastitis. To deter- phy. Finally, the number of apoptotic epithelial cells per
mine whether the endotoxin lipopolysaccharide (LPS) 10 microscopic sections, as determined by the TUNEL
is responsible for E. coli-mediated tissue damage, Ca- assay, increased from 1.8 ± 0.5 to 8.8 ± 2.8 cells. In
puco et al. (1985) cultured lactating mammary tissue addition, there was evidence for induction of apoptosis
explants in the presence of E. coli endotoxin and a labo- by other mastitis pathogens. For example, Sheffield
ratory culture filtrate of E. coli. Results indicated that (1997) reported a 5-fold induction of a putative marker
endotoxin had no effect, but the culture filtrate dam- of apoptosis, testosterone-repressed prostate mucin-2
aged tissue and decreased milk synthesis and secretion. mRNA, after experimental infection of the bovine mam-
One of the most common types of chronic mastitis is mary gland with Strep. agalactiae. Finally, in vitro
caused by Staph. aureus. Histopathological responses of studies indicated that Staph. aureus caused apoptosis
lactating tissue to experimental or naturally occurring in a bovine mammary cell line (Bayles et al., 1998).
Staph. aureus mastitis were extensively studied during Nevertheless, the effect of Staph. aureus and Strep.
the 1970s and 1980s. Chandler and Reid (1973) exam- agalactiae on apoptosis of mammary tissue still needs
ined mammary parenchymal tissue samples from lac- to be confirmed in vivo. It is worthwhile to indicate
tating cows infected naturally with Staph. aureus and that apoptosis does not cause major histomorphological
60 Zhao and Lacasse

changes, as observed in traditional mastitis studies; all types of biomolecules (e.g., DNA, proteins, lipids,
however, it does reduce cell numbers in mammary tis- and carbohydrates) and therefore induce tissue injury.
sue. In addition, apoptosis may cause tissue damage in Bovine PMN have primary (azurophilic), secondary,
a delayed fashion through secondary necrosis (Medan and tertiary granules (Paape et al., 2002, 2003). These
et al., 2002). intracellular granules contain bactericidal peptides,
The clearance processes of apoptotic cells during mas- proteins, and enzymes such as elastase, other protein-
titis are not totally understood. The clearance of apop- ases, and myeloperoxidase that are released into phago-
totic cells by phagocytes, such as macrophages, has cytic vacuoles or the extracellular environment
been shown. In addition, mammary epithelial cells are (Faurschou and Borregaard, 2003). Proteolytic enzymes
known to be capable of phagocytosing apoptotic cells in in PMN include neutral and acidic proteases. Elastase
species other than cattle (Monks et al., 2002). Because (EC 3.4.21.36) and cathepsin G (EC 3.4.21.20) are the
methods for detection of apoptosis reveal only the cells predominant enzymes in PMN (Bank and Ansorge,
involved in the process, it is quite possible that 2001). Other proteases in PMN include the thiol prote-
apoptosis during mastitis is underestimated. ase cathepsin B (EC 3.4.22.1) and the acid protease
cathepsin D (EC 3.4.23.5), as reported by Owen and
INVOLVEMENT OF NEUTROPHILS Campbell (1999). Furthermore, activated bovine PMN
IN TISSUE DAMAGE can express matrix metalloproteinase (MMP)-9 (Li et
al., 1999).
Milk PMN usually have lower total proteolytic enzy-
Following detection of pathogen invasion into the
matic activities than peripheral blood PMN (Prin-Ma-
mammary gland, macrophages and epithelial cells re-
thieu et al., 2002), indicating that PMN release many
lease chemoattractants. These agents trigger the mi-
proteases during migration into the mammary gland.
gration of leukocytes, mainly PMN, from the blood to-
Using an E. coli mastitis model, Haddadi et al. (2006)
ward the mammary gland and increase their propor-
observed that milk PMN had lower cathepsin and colla-
tions from a basal level of 5 to 25% to approximately genase activities than peripheral blood PMN. Thus,
90% of total cells in the milk (Leitner et al., 2000; Riollet PMN appear to use up part of these enzymes during
et al., 2000a). These PMN are considered as the second their migration to cross the endothelium, extracellular
line of defense of the mammary gland. The presence of matrix (ECM), and epithelium (Faurschou and Borreg-
functional PMN is crucial to the host defense against aard, 2003). In contrast, Le Roux et al. (2003) reported
bacterial pathogens (Paape et al., 2003). that elastase was higher in milk than in blood. Elastase
The main functions of PMN are to engulf pathogens is localized in azurophilic granules of PMN, and its
and destroy them via oxygen-dependent and oxygen- increase in milk is probably due to proinflammatory
independent systems. At the same time, PMN can po- cytokines such as IL-6, IL-2, and tumor necrosis factor-
tentially harm the mammary gland. The exact mecha- α, which increase the transcription of serine proteases,
nism by which PMN damage bovine epithelial cells dur- including elastase and cathepsin G (Le Roux et al.,
ing mastitis is still not fully understood. Neutrophils 2003). Thus, the amount and variety of enzymes re-
may promote tissue injury and disturb mammary func- leased by PMN into milk seem to be affected by the
tion, via reactive oxygen metabolite generation (i.e., the migration and activation of PMN.
respiratory burst) and granular enzyme release (i.e., The enzymes involved in bovine mammary tissue de-
degranulation; Paape et al., 2002). struction were investigated by Mehrzad et al. (2005)
During experimentally induced Staph. aureus masti- by using an endotoxin-induced mastitis model. They
tis, migration of PMN through the secretory epithelium reported that mastitic milk proteases hydrolyzed ca-
was correlated with extensive morphological damage sein, gelatin, collagen, hemoglobin, mammary gland
(Harmon and Heald, 1982). Direct evidence that PMN membrane proteins, and lactoferrin, indicating that
damage mammary tissue was provided by Capuco et mastitic milk proteases have a broad spectrum of activi-
al. (1986). Neutrophils isolated from mammary glands ties. Further, the direct involvement of proteases in
of nulliparous heifers given an injection of E. coli endo- epithelial cell damage was demonstrated by the fact
toxin were incubated with mammary tissues from non- that coincubation of normal mammary tissue with mas-
infected quarters. Microscopic examination indicated titic milk, but not normal milk, caused tissue degrada-
that epithelial cell damage resulted from treatment tion (Mehrzad et al., 2005). Therefore, proteases re-
with intact, lysed, or phagocytosing PMN. The greatest leased by PMN are likely involved in mammary tissue
morphological damage resulted from treatment with damage during mastitis.
phagocytosing PMN. This observation was confirmed It is unlikely that the diapedesis process itself is di-
in an in vitro coculture system with mammary epithe- rectly involved in epithelial damage. Under normal con-
lial cells and PMN. Activated blood PMN were cytotoxic ditions, PMN can migrate into mammary glands with-
for mammary epithelial cells (Ledbetter et al., 2001; out damaging the tissue. Further, PMN diapedesis it-
Lauzon et al., 2005), possibly via the release of extracel- self did not cause detectable epithelial cell damage in
lular reactive oxygen species, such as hydroxyl radicals an in vitro study (Lin et al., 1995). However, prolonged
(Boulanger et al., 2002). Oxidative stress can damage diapedesis of leukocytes could cause damage to mam-
Tissue damage and mastitis 61
mary parenchymal tissue, resulting in decreased pro- bovine mammary tissue (Capuco et al., 1985) or mam-
duction of milk (Harmon and Heald, 1982; Sordillo and mary epithelial cells (Boulanger et al., 2002). However,
Nickerson, 1988). This damage may occur through sev- the possibility for involvement of LPS in E. coli-induced
eral mechanisms, including premature activation dur- cell damage cannot be totally ruled out, because LPS
ing migration, extracellular release of toxic products stimulated the expression of IL-1 by MAC-T cells, an
during the killing of some microbes, removal of infected established bovine mammary epithelial cell line (Boud-
or damaged host cells and debris as a first step in tissue jellab et al., 2000). In addition, LPS increased expres-
remodeling, or failure to terminate acute inflamma- sion of urokinase-type plasminogen activator (uPA;
tory responses. Ohta et al., 2000). Therefore, it is plausible that LPS
The PMN-induced damage is probably somewhat con- induces apoptosis or necrosis in mammary epithelial
tained. After ingestion and release of their chemicals, cells indirectly through induction of proteases or proin-
most of the milk PMN perish by induction of apoptosis. flammatory cytokines. At this time, however, there is
This is followed by the migration of macrophages into no experimental evidence to support this notion, pre-
the mammary gland and engulfment of PMN by these sumably because of the previous lack of sensitive ana-
macrophages. Through this process, damaging chemi- lytical methods.
cals are walled off within dying PMN, which are then Staphylococcus aureus produces toxins that destroy
ingested by macrophages to minimize further damage cell membranes, directly damage milk-producing tis-
to tissue (Paape et al., 2002, 2003). sue, and induce necrosis in bovine mammary glands
(Chandler and Reid, 1973; Heald, 1979; Nickerson and
INVOLVEMENT OF BACTERIA Heald, 1981; Sordillo and Nickerson, 1988; Trinidad et
IN TISSUE DAMAGE al., 1990). Initially, the bacteria damage tissues lining
the teat and gland cisterns within the quarter. If un-
There is increasing evidence that pathogens use vari- checked, they invade the duct system and establish
ous mechanisms to impinge upon cell death pathways. deep-seated pockets of infection in the milk-secreting
A number of pathogens are armed with an array of cells (i.e., alveoli). This is followed by the walling-off of
virulence determinants, which interact with key compo- bacteria by scar tissue and the formation of abscesses.
nents of a host cell’s death pathways or interfere with Moreover, Bayles et al. (1998) provided evidence that
regulation of transcription factors monitoring cell sur- Staph. aureus induced apoptosis in bovine mammary
vival. These virulence factors induce cell death by a gland epithelial cells. They showed that 2 h after the
variety of mechanisms, which include 1) pore-forming internalization of one Staph. aureus mastitis isolate,
toxins, which interact with the host cell membrane and MAC-T cells exhibited detachment from the matrix,
permit the leakage of cellular components; 2) toxins rounding, a mottled cell membrane, and vacuolation
that express their enzymatic activity in the host cytosol; of the cytoplasm, all of which are indicative of cells
3) effector proteins delivered directly into host cells undergoing apoptosis. By 18 h, the majority of the MAC-
by a highly specialized type-III secretory system; 4) T cell population exhibited an apoptotic morphology.
superantigens that target immune cells, and 5) other Other evidence for apoptosis was the existence of a
modulators of host cell death (Weinrauch and Zychlin- characteristic DNA ladder pattern and the positive TU-
sky, 1999). Much progress has been made in under- NEL labeling of apoptotic cells. These results clearly
standing the role of apoptosis and necrosis in response demonstrate that after internalization, Staph. aureus
to bacterial infection. escapes the endosome and induces apoptosis in mam-
Escherichia coli produces a number of proteinases, mary epithelial cells. The apoptosis depends on factors
including collagenolytic enzymes, which contribute to regulated by the global virulence gene regulators Agr
the degradation of ECM components (Haddadi et al., and Sar (Wesson et al., 1998), as well as by caspases 8
2005, 2006). There is no doubt that these proteolytic and 3 (Wesson et al., 2000). Although there is still no
activities contribute to the apoptosis and necrosis of direct evidence showing that Staph. aureus induces
mammary epithelial cells. However, the degree to apoptosis of bovine mammary epithelial cells in vivo,
which they contribute is still unknown. this seems likely.
Endotoxin LPS is considered a key factor for the Staphylococcus aureus produces a wide variety of exo-
pathogenicity of gram-negative bacteria. Accordingly, proteins that contribute to its ability to colonize and
intramammary infusion of E. coli LPS is often used to cause disease in mammalian hosts. Nearly all strains
study events occurring during E. coli mastitis, because examined secrete a group of enzymes and cytotoxins,
it mimics the symptoms of naturally occurring mastitis which includes 4 hemolysins (α, β, λ, and δ), nucleases,
without microorganism invasion and toxin production, proteases, lipases, hyaluronidase, and collagenase. The
which could cause direct damaging effects on the mam- main function of these proteins may be to convert local
mary epithelial cells (Oliver and Calvinho, 1995). When host tissues into the nutrients required for bacterial
LPS was used to induce an inflammatory response in growth. Some strains produce one or more additional
the mammary gland, no lesions were observed in the exoproteins, which include toxic shock syndrome toxin-
mammary glands (Hill, 1991). Similarly, LPS does not 1, staphylococcal enterotoxins, exfoliative toxins, and
cause tissue damage in vitro in explants of lactating leukocidin (Dinges et al., 2000). Strains isolated from
62 Zhao and Lacasse

cases of bovine mastitis express α, β, λ, and δ toxins, involved in the activation of plasminogen to plasmin
leukocidins, enterotoxin, and coagulase (Bramley et al., (Green and Lund, 2005).
1989; Matsunaga et al., 1993). Among all the Staph. The plasmin-plasminogen activator system in bovine
aureus cytotoxins, α-hemolysin has been the most care- milk is closely correlated with gradual involution (Pol-
fully examined. It can induce both apoptosis and necro- itis, 1996). Plasmin directly degrades matrix proteins
sis in eukaryotic cells, depending on the dosage given such as fibrin and laminin and also activates MMP
(reviewed by Weinrauch and Zychlinsky, 1999). At low precursors, such as pro-MMP-3, MMP-9, and MMP-
doses, the toxin binds to specific, but as yet unidentified, 13 (Green and Lund, 2005). The increase in plasmin
cell surface receptors and produces small pores that activity during mastitis is linked to the permeability
selectively facilitate the release of monovalent ions, re- of the milk epithelial barrier during inflammation. In
sulting in characteristic DNA fragmentation and cell addition, numerous activators of plasmin and its zymo-
death. At high doses, α-hemolysin nonspecifically ab- gen come from the bloodstream, PMN, and bacteria.
sorbs to the lipid bilayer and forms larger pores in the During mastitis, the serine protease activator concen-
membrane that are Ca2+ permissive, which results in tration, including those for plasmin and plasminogen
massive necrosis without DNA fragmentation (Wein- in blood and also in milk, increases sharply (Le Roux
rauch and Zychlinsky, 1999). Whether this also applies et al., 2003; Moussaoui et al., 2004). Polymorphonuclear
to bovine mammary epithelial cells is unknown. Other neutrophils have a pool of plasminogen activators, such
bacterial toxins could also have damaging effects on as uPA and, to a lesser extent, tissue plasminogen acti-
bovine mammary tissues, but they have not been stud- vators (Moir et al., 2001). Several bacteria, such as
ied intensively. Kuroishi et al. (2003) inoculated mam- Staph. aureus, E. coli, and Salmonella typhimurium,
mary glands with staphylococcal enterotoxin C and ob- express a plasminogen receptor on their surfaces,
served epithelial cellular degeneration, including inva- which, through immobilization of plasminogen, en-
gination and cytoplasmic vacuolation of epithelial cells. hances plasminogen activation into plasmin (Lahteen-
It is not clear whether this effect is due to increased maki et al., 2001).
production of the superoxide by migrated PMN or How much plasmin contributes to tissue damage dur-
whether it is a direct effect of staphylococcal enterotoxin ing mastitis is debatable. Mammary epithelial cell via-
C on mammary epithelial cells. bility depends on attachment to the ECM, so it is rea-
Streptococcus dysgalactiae is an environmental sonable to postulate that ECM degradation is involved
pathogen that causes bovine IMI. Almeida and Oliver in tissue damage and cell death during mastitis. Ex-
(1995) assayed 2 mastitis strains of Strep. dysgalactiae pression of MMP-9, stromelysin-1 mRNA, and uPA
for their ability to invade, multiply, and induce damage mRNA was increased in association with apoptosis dur-
to MAC-T cells. They demonstrated that the invasion ing E. coli mastitis (Long et al., 2001). Matrix metallo-
process did not appear to affect the viability of mam- protease-9 can be produced by bovine PMN (Li et al.,
mary epithelial cells but that cellular damage was in- 1999) and MAC-T cells (Long et al., 2001). Stromelysin-
duced, as indicated by the release of increasing amounts 1 contributed to the breakdown of most ECM compo-
of lactate dehydrogenase (Almeida and Oliver, 1995). nents, including laminin and collagen type IV (Rudolph-
Owen and Matrisian, 1998). Other proteases that have
been reported to increase in milk from mastitic cows
INVOLVEMENT OF PLASMA PROTEINS
include MMP-2 and MMP-9 (Raulo et al., 2002; Lauzon
AND CYTOKINES IN TISSUE DAMAGE
et al., 2006), as well as a 120-kDa gelatinase (Mehrzad
et al., 2005; Lauzon et al., 2006). Mehrzad et al. (2005)
At the onset of mastitis, increased permeability of also tested proteolytic activity directly in normal mam-
the blood-milk mammary epithelial barrier leads first mary tissue of mastitic cows. They found that lac-
to an influx of serum constituents, such as plasminogen toserum from mastitic cows was more proteolytic than
and numerous other enzymes, and second to a massive normal lactoserum. Lactoserum from mastitic cows ex-
recruitment of somatic cells, in particular, PMN (Le foliated the cells and surrounding proteins, leaving a
Roux et al., 2003). Milk contains 2 proteinase systems nude dense collagen network. These differences re-
derived from blood, one of which is involved in dissolv- sulted from protease contents and activities, which
ing blood clots (i.e., plasmin) and the other in defense were significantly higher in mastitic milk (Mehrzad et
against invasive microorganisms (i.e., lysosomal pro- al., 2005). They concluded that the proteolysis and tis-
teinases of somatic cells). Whereas plasmin is the prin- sue damage observed were largely due to MMP. How-
cipal proteinase in good-quality milk, other proteinases, ever, the possible contribution from other nonplasmin
including cathepsins and elastase, are probably also proteases and plasmin should not be ignored.
active, particularly as the somatic cell count of milk Cytokines are central mediators of inflammatory
increases (Kelly et al., 2006; Leitner et al., 2006). This events during IMI. Bacteria release potent toxins that
is supported by the observation that the protease activi- trigger white blood cells and epithelial cells in the mam-
ties of plasmin and mastitic milk differ (Mehrzad et mary gland to secrete cytokines (Paape et al., 2003).
al., 2005). In addition, mammary epithelial cells also Intramammary challenge with E. coli or Staph. aureus
express certain MMP and serine proteases, which are has elicited differential innate immune responses in
Tissue damage and mastitis 63
terms of clinical symptoms and milk cytokine profiles ide to hydrogen peroxide. Hydrogen peroxide is con-
at the protein level (Bannerman et al., 2004; Riollet et verted to water by the enzyme glutathione peroxidase
al., 2000b) and at mRNA levels (Lee et al., 2006). The (Ward et al., 1988). Those 2 enzymes effectively control
difference in cytokine profiles may underlie the differ- most free radicals within the cytosol. Accordingly, the
ences in the sequela of symptoms for these 2 types use of antioxidants to prevent oxidative stress has been
of mastitis. studied in several types of inflammation (Cuzzocrea et
Very little work has been conducted to determine the al., 2004).
role of cytokines in the regulation of tissue damage Using a coculture model of activated bovine PMN
during mastitis. Cytokines recruit PMN that function and MAC-T cells, Lauzon et al. (2005) found that 3
as phagocytes at the site of infection. During a study antioxidants (i.e., catechin, deferoxamine, or glutathi-
on the use of recombinant bovine (rb) IL-8 as a potential one ethyl ester) could partially or totally eliminate the
therapeutic agent for subclinical mastitis of dairy cows, damaging effect of activated bovine PMN to MAC-T
Takahashi et al. (2005) observed significant increases cells, indicating that antioxidants may be effective tools
in milk somatic cell counts and in chemiluminescence for protecting mammary tissue against PMN-induced
activity after intramammary injection of rbIL-8. Al- oxidative stress during bovine mastitis. The protective
though they did not study the tissue damage, the high effects of these 3 antioxidants on PMN-induced damage
levels of milk somatic cell counts and free radical pro- to mammary cells were further evaluated in vivo by
duction would certainly have led to tissue damage. It using an endotoxin-induced mastitis model. The extent
has previously been reported that rbIL-8 can induce of cell damage was evaluated by measuring milk levels
the migration of PMN through an in vitro model of the of lactate dehydrogenase and NAGase activity. Intra-
endo-epithelial barrier consisting of the bovine aortic mammary infusions of catechin or glutathione ethyl
endothelial cell line and MAC-T cells (Lee and Zhao, ester did not exert any protective effect, whereas infu-
2000). Other cytokines, such as tumor necrosis factor- sion of deferoxamine, a chelator of iron, decreased milk
α and IL-1, induce apoptosis in a variety of cell types, lactate dehydrogenase and NAGase activity, indicating
including bovine endothelial cells (Mebmer et al., 1999) a protective effect against PMN-induced damage. The
and human mammary epithelial cells (Burow et al., protective effect of deferoxamine was also evident from
1999). Levels of these cytokines increase during E. coli a lower level of haptoglobin in milk. The proteolytic
mastitis (Bannerman et al., 2004), and it is tempting activity of mastitic milk was not influenced by the pres-
to assume that they also induce apoptosis in bovine ence of deferoxamine. Overall, these results indicate
mammary epithelial cells. A range of cytokines are also that local infusion of deferoxamine may be an effective
known to promote a wide variety of functions of PMN, tool to protect mammary tissue against PMN-induced
including adhesion, surface receptor expression, free oxidative stress during bovine mastitis (Lauzon et
radical production, and release of lysosomal constit- al., 2006).
uents (Paape et al., 2003). Therefore, the effects of cy-
tokines on tissue damage are more likely to be mediated SUMMARY
through recruitment and activation of PMN.
Despite considerable advances in the understanding
REDUCTION OF TISSUE DAMAGE of the pathogenesis of bovine mastitis, the disease con-
tinues to be an economically important problem in the
Although the mechanisms underlying tissue damage dairy industry throughout the world. The ideal means
during mastitis have not been fully delineated, there of dealing with mastitis is to prevent it from occurring.
are likely opportunities for pharmacological interven- However, even under the best prevention and control
tion to block the proteolytic or oxidative cascade within programs, mastitis will still occur. Reducing economic
the inflamed gland to reduce tissue damage during mas- losses resulting from mastitis is still a daunting chal-
titis. Up to now, most work in this area has been focused lenge. Although it is accepted that neutrophils, bacte-
on the reduction of free radicals, because results from ria, and host proteases and cytokines contribute to
research using in vitro models indicate that LPS stimu- mammary tissue damage during mastitis, many finer
lates PMN-induced damage to mammary epithelial details are still unknown. Mammary tissue damage
cells by producing superoxide (Boulanger et al., 2002). during IMI is probably underestimated because of the
The major free radicals found in biological systems lack of sensitive and noninvasive detection methods.
are superoxide, hydrogen peroxide, hydroxyl radical, Early detection of IMI is a prerequisite for initiating
and fatty acid radicals (Ward et al., 1988). Hydrogen any intervention measure to minimize the mammary
peroxide is found primarily in the cytosol of cells, and tissue damage. The mechanisms responsible for mam-
fatty acid radicals are found primarily in cell mem- mary epithelium and tissue damage during mastitis
branes (Ward et al., 1988). Superoxide and hydroxyl are still not well known, but they include both apoptosis
radicals can be found in both cell components (Paape and necrosis. Understanding the biochemical and cellu-
et al., 2003). Because free radicals are extremely toxic lar changes that occur in the gland during mastitis will
to cells, the body has developed a sophisticated antioxi- ultimately lead to a means of manipulating mammary
dant system. Superoxide dismutase converts superox- functions to minimize the damage from mastitis. In
64 Zhao and Lacasse

addition to the use of antibiotics to treat mastitis, other Faurschou, M., and N. Borregaard. 2003. Neutrophil granules and
secretory vesicles in inflammation. Microb. Infect. 5:1317–1327.
measures for reducing tissue damage may be a cost-
Fellows, M. D., and M. R. O’Donovan. 2007. Cytotoxicity in cultured
effective way to reduce the losses caused by mastitis. mammalian cells is a function of the method used to estimate
it. Mutagenesis 22:275–280.
LITERATURE CITED Frost, A. J., and B. E. Brooker. 1986. Hyperacute Escherichia coli
mastitis of cattle in the immediate post-partum period. Aust.
Almeida, R. A., and S. P. Oliver. 1995. Invasion of bovine mammary Vet. J. 63:327–331.
epithelial cells by Streptococcus dysgalactiae. J. Dairy Sci. Frost, A. J., A. W. Hill, and B. E. Brooker. 1980. The early pathogene-
78:1310–1317. sis of bovine mastitis due to Escherichia coli. Proc. R. Soc. Lond.
Arends, M. J., R. G. Morris, and A. H. Wyllie. 1990. Apoptosis: The B. Biol. Sci. 209:419–431.
role of endonuclease. Am. J. Pathol. 136:593–608. Gozuacik, D., and A. Kimchi. 2004. Autophagy as a cell death and
Bank, U., and S. Ansorge. 2001. More than destructive: Neutrophil tumor suppressor mechanism. Oncogene 23:2891–2906.
derived serine proteases in cytokine bioactivity control. J. Leu- Green, K. A., and L. R. Lund. 2005. ECM degrading proteases and
koc. Biol. 69:197–206. tissue remodeling in the mammary gland. Bioessays 27:894–903.
Bannerman, D. D., M. J. Paape, J. W. Lee, X. Zhao, J. C. Hope, and Haddadi, K., F. Moussaoui, I. Hebia, F. Laurent, and Y. Le Roux.
P. Rainard. 2004. Escherichia coli and Staphylococcus aureus 2005. E. coli proteolytic activity in milk and casein breakdown.
elicit different innate immune responses following intramam- Reprod. Nutr. Dev. 45:485–496.
mary infection. Clin. Diagn. Lab. Immunol. 11:463–472. Haddadi, K., C. Prin-Mathieu, F. Moussaoui, G. C. Faure, F. Van-
Bayles, K. W., C. A. Wesson, L. E. Liou, L. K. Fox, G. A. Bohach, groenweghe, C. Burvenich, and Y. Le Roux. 2006. Polymorpho-
and W. R. Trumble. 1998. Intracellular Staphylococcus aureus nuclear neutrophils and Escherichia coli proteases involved in
escapes the endosome and induces apoptosis in epithelial cells. proteolysis of casein during experimental E. coli mastitis. Int.
Infect. Immun. 66:336–342. Dairy J. 16:639–647.
Benites, N. R., J. L. Guerra, P. A. Melville, and E. O. da Costa. 2002. Harmon, R. J., and C. W. Heald. 1982. Migration of polymorphonu-
Aetiology and histopathology of bovine mastitis of a spontaneous clear leukocytes into the bovine mammary gland during experi-
occurrence. J. Vet. Med. B 49:366–370. mentally induced Staphylococcus aureus mastitis. Am. J. Vet.
Boudjellab, N., H. S. Chan-Tang, and X. Zhao. 2000. Bovine interleu- Res. 43:992–998.
kin-1 expression by cultured mammary epithelial cells (MAC- Heald, C. W. 1979. Morphometric study of experimentally induced
T) and its involvement in the release of MAC-T derived interleu- Staphylococcus aureus mastitis in the cow. Am. J. Vet. Res.
kin-8. Comp. Physiol. Biochem. 127:191–199. 40:1294–1298.
Hill, A. W. 1991. Somatic cells—Friends or foes? Pages 217–232 in
Boulanger, V., X. Zhao, and P. Lacasse. 2002. Protective effect of
New Insights into the Pathogenesis of Mastitis. C. Burvenich,
melatonin and catalase in bovine neutrophil-induced model of
G. Vandeputte-Van Messom, and A. W. Hill, ed. Flemish Vet.
mammary cell damage. J. Dairy Sci. 85:562–569.
J., Merelbeke, Belgium.
Bradley, A. J. 2002. Bovine mastitis: An evolving disease. Vet. J.
Kelly, A. L., F. O’Flaherty, and P. F. Fox. 2006. Indigenous proteolytic
163:1–13.
enzymes in milk: A brief overview of the present state of knowl-
Bramley, A. J., A. H. Patel, M. O’Reilly, R. Foster, and T. J. Foster.
edge. Int. Dairy J. 16:563–572.
1989. Roles of alpha-toxin and beta-toxin in virulence of Staphy-
Kerr, J. F. R., A. H. Wyllie, and A. R. Currie. 1972. Apoptosis: A
lococcus aureus for the mouse mammary gland. Infect. Immun.
basic biological phenomenon with wide-ranging implications in
57:2489–2494.
tissue kinetics. Br. J. Cancer 26:239–257.
Burow, M. E., Y. Tang, B. M. Collins-Burow, S. Krajewski, J. C.
Kitchen, B. J., G. Middleton, I. G. Durward, R. J. Andrews, and M.
Reed, J. A. McLachlan, and B. S. Bechman. 1999. Effects of
C. Salmon. 1980. Mastitis diagnostic tests to estimate mammary
environmental estrogens on tumor necrosis factor α-mediated
gland epithelial cell damage. J. Dairy Sci. 63:978–983.
apoptosis in MCF-7 cells. Carcinogenesis 20:2057–2061.
Kornalijnslijper, J. E., A. J. J. M. Daemen, T. van Werven, T. A.
Burvenich, C., V. van Merris, J. Mehrzad, A. Diez-Fraile, and L.
Niewold, V. P. M. G. Rutten, and E. N. Noordhuizen-Stassen.
Duchateau. 2003. Severity of E. coli mastitis is mainly deter-
2004. Bacterial growth during the early phase of infection deter-
mined by cow factors. Vet. Res. 34:521–564.
mines the severity of experimental Escherichia coli mastitis in
Capuco, A. V., and R. M. Akers. 1999. Mammary involution in dairy
dairy cows. Vet. Microbiol. 10:177–186.
animals. J. Mammary Gland Biol. Neoplasia 4:137–144.
Kuroishi, T., K. Komine, K. Asai, J. Kobayashi, K. Watanabe, T.
Capuco, A. V., S. A. Bright, J. W. Pankey, D. L. Wood, R. H. Miller,
Yamaguchi, S. Kamata, and K. Kumagai. 2003. Inflammatory
and J. Bitman. 1992. Increased susceptibility to intramammary
responses of bovine polymorphonuclear neutrophils induced by
infection following removal of teat canal keratin. J. Dairy Sci. staphylococcal enterotoxin C via stimulation of mononuclear
75:2126–2130. cells. Clin. Diagn. Lab. Immunol. 10:1011–1018.
Capuco, A. V., S. E. Ellis, S. A. Hale, E. Long, R. A. Erdman, X. Zhao, Lahteenmaki, K., P. Kuusela, and T. K. Korhonen. 2001. Bacterial
and M. J. Paape. 2003. Lactation persistency: Insights from plasminogen activators and receptors. FEMS Microbiol. Rev.
mammary cell proliferation studies. J. Anim. Sci. 81(Suppl. 25:531–552.
3):18–31. Lauzon, K., X. Zhao, A. Bouetard, L. Delbecchi, B. Paquette, and
Capuco, A. V., M. J. Paape, and S. C. Nickerson. 1986. In vitro study P. Lacasse. 2005. Antioxidants to prevent bovine neutrophil-
of polymorphonuclear leukocyte damage to mammary tissues of induced mammary epithelial cell damage. J. Dairy Sci.
lactating cows. Am. J. Vet. Res. 47:663–668. 88:4295–4303.
Capuco, A. V., M. J. Paape, J. J. Smith, and D. A. Loeffler. 1985. In Lauzon, K., X. Zhao, and P. Lacasse. 2006. Deferoxamine reduces
vitro effect of bacterial toxins on lactating bovine mammary tissue damage during endotoxin-induced mastitis in dairy cows.
tissue. J. Dairy Sci. 68(Suppl. 1):206. J. Dairy Sci. 89:3846–3857.
Chandler, R. L., and I. M. Reid. 1973. Ultrastructure and associated Ledbetter, T. K., M. J. Paape, and L. W. Douglas. 2001. Cytotoxic
observations in clinical cases of mastitis in cattle. J. Comp. Pa- effects of peroxynitrite, polymorphonuclear neutrophils, free
thol. 83:233–241. radical scavengers, inhibitors of myeloperoxidase, and inhibitors
Cuzzocrea, S., C. Thiemermann, and D. Salvemini. 2004. Potential of nitric oxide synthase on bovine mammary secretory epithelial
therapeutic effect of antioxidant therapy in shock and inflam- cells. Am. J. Vet. Res. 62:286–293.
mation. Curr. Med. Chem. 11:1147–1162. Lee, J.-W., D. D. Bannerman, M. J. Paape, M.-K. Huang, and X.
Dinges, M. M., P. M. Orwin, and P. M. Schlievert. 2000. Exotoxins Zhao. 2006. Characterization of cytokine expression in milk so-
of Staphylococcus aureus. Clin. Microbiol. Rev. 13:16–34. matic cells during intramammary infections with Escherichia
Tissue damage and mastitis 65
coli or Staphylococcus aureus by real-time PCR. Vet. Res. neutrophil leukocytes. J. Mammary Gland Biol. Neoplasia
37:219–229. 7:109–121.
Lee, J., and X. Zhao. 2000. Recombinant human interleukin-8, but Pitkala, A., M. Haveri, S. Pyorala, V. Myllys, and T. Honkanen-
not human interleukin-1β, induces bovine neutrophil migration Buzalski. 2004. Bovine mastitis in Finland 2001—Prevalence,
in an in vitro co-culture system. Cell Biol. Int. 24:889–895. distribution of bacteria, and antimicrobial resistance. J. Dairy
Leitner, G., O. Krifucks, U. Merin, Y. Lavi, and N. Silanikove. 2006. Sci. 87:2433–2441.
Interactions between bacteria type, proteolysis of casein and Politis, I. 1996. Plasminogen activator system: Implications for mam-
physico-chemical properties of bovine milk. Int. Dairy J. mary cell growth and involution. J. Dairy Sci. 79:1097–1107.
16:648–654. Prin-Mathieu, C., Y. Le Roux, G. C. Faure, F. Laurent, M. C. Béné,
Leitner, G., E. Shoshani, O. Krifucks, M. Chaffer, and A. Saran. 2000. and F. Moussaoui. 2002. Enzymatic activities of bovine periph-
Milk leucocyte population patterns in bovine udder infection of eral blood leukocytes and milk polymorphonuclear neutrophils
different etiology. J. Vet. Med. B. 47:581–589. during intramammary inflammation caused by lipopolysaccha-
Le Roux, Y., F. Laurent, and F. Moussaoui. 2003. Polymorphonuclear ride. Clin. Diagn. Lab. Immunol. 9:812–817.
proteolytic activity and milk composition change. Vet. Res. Raulo, S. M., T. Sorsa, T. Tervahartiala, T. Latvanen, E. Pirila, J.
34:629–645. Hirvonen, and P. Maisi. 2002. Increase in milk metalloprotei-
Li, X., X. Zhao, and S. Ma. 1999. Secretion of 92 kDa gelatinase nase activity and vascular permeability in bovine endotoxin-
(MMP-9) by bovine neutrophils. Vet. Immunol. Immunopathol. induced and naturally occurring Escherichia coli mastitis. Vet.
67:247–258. Immunol. Immunopathol. 85:137–145.
Lin, Y., L. Xia, J. D. Turner, and X. Zhao. 1995. Morphological obser- Riollet, C., P. Rainard, and B. Poutrel. 2000a. Cells and cytokines in
vation of neutrophil diapedesis across bovine mammary gland inflammatory secretions of bovine mammary gland. Adv. Exp.
epithelium in vitro. Am. J. Vet. Res. 56:203–207. Med. Biol. 480:247–258.
Long, E., A. V. Capuco, D. L. Wood, T. Sonstegard, G. Tomita, M. J. Riollet, C., P. Rainard, and B. Poutrel. 2000b. Differential induction
Paape, and X. Zhao. 2001. Escherichia coli induces apoptosis and of complement fragment C5a and inflammatory cytokines during
proliferation of mammary cells. Cell Death Differ. 8:808–816. intramammary infections with Escherichia coli and Staphylococ-
Matsunaga, T., S. Kamata, N. Kakiichi, and K. Uchida. 1993. Charac- cus aureus. Clin. Diagn. Lab. Immunol. 7:161–167.
teristics of Staphylococcus aureus isolated from peracute, acute Rudolph-Owen, L. A., and L. M. Matrisian. 1998. Matrix metallopro-
and chronic bovine mastitis. J. Vet. Med. Sci. 55:297–300. teinases in remodeling of the normal and neoplastic mammary
Mebmer, U. K., V. A. Briner, and J. Pfeilschifter. 1999. Tumor necrosis gland. J. Mam. Gland Biol. Neoplasia 3:177–189.
factor-α and lipopolysaccharide induce apoptotic cell death in Sheffield, L. G. 1997. Mastitis increases growth factor messenger
bovine glomerular endothelial cells. Kidney Int. 55:2322–2337. ribonucleic acid in bovine mammary glands. J. Dairy Sci.
Medan, D., L. Wang, X. Yang, S. Dokka, V. Castranova, and Y. Rojana- 80:2020–2024.
sakul. 2002. Induction of neutrophil apoptosis and secondary Sordillo, L. M., and S. C. Nickerson. 1988. Morphologic changes in
necrosis during endotoxin-induced pulmonary inflammation in the bovine mammary gland during involution and lactogenesis.
mice. J. Cell. Physiol. 191:320–326. Am. J. Vet. Res. 49:1112–1120.
Sordillo, L. M., and K. L. Streicher. 2002. Mammary gland immunity
Mehrzad, J., C. Desrosiers, K. Lauzon, G. Robitaille, X. Zhao, and P.
and mastitis susceptibility. J. Mammary Gland Biol. Neoplasia
Lacasse. 2005. Proteases involved in mammary tissue damage
7:135–146.
during endotoxin-induced mastitis in dairy cows. J. Dairy Sci.
Strange, R., F. Li, S. Saurer, A. Burkhardt, and R. R. Friis. 1992.
88:211–222.
Apoptotic cell death and tissue remodeling during mouse mam-
Moir, E., N. A. Booth, B. Bennett, and L. A. Robbie. 2001. Polymorpho-
mary gland involution. Development 115:49–58.
nuclear leukocytes mediate endogenous thrombus lysis via a u-
Takahashi, H., T. Komatsu, K. Hodate, R. Horino, and Y. Yokomizo.
PA-dependent mechanism. Br. J. Haematol. 113:72–80.
2005. Effect of intramammary injection of RbIL-8 on milk levels
Monks, J., F. J. Geske, L. Lehman, and V. A. Fadok. 2002. Do inflam-
of somatic cell count, chemiluminescence activity and shedding
matory cells participate in mammary gland involution? J. Mam-
patterns of total bacteria and S. aureus in Holstein cows with
mary Gland Biol. Neoplasia 7:163–176.
naturally Infected-subclinical mastitis. J. Vet. Med. B 52:32–37.
Moussaoui, F., F. Vangroenweghe, K. Haddadi, Y. Le Roux, F. Trinidad, P., S. C. Nickerson, and R. W. Adkinson. 1990. Histopathol-
Laurent, L. Duchateau, and C. Burvenich. 2004. Proteolysis in ogy of staphylococcal mastitis in unbred dairy heifers. J. Dairy
milk during experimental Escherichia coli mastitis. J. Dairy Sci. Sci. 73:639–647.
87:2923–2931. Ward, P. A., J. S. Warren, and K. J. Johnson. 1988. Oxygen radicals,
Nickerson, S. C., and C. W. Heald. 1981. Histopathologic response inflammation, and tissue injury. Free Radical Bio. Med.
of the bovine mammary gland to experimentally induced Staphy- 5:403–408.
lococcus aureus infection. Am. J. Vet. Res. 42:1351–1355. Weinrauch, Y., and A. Zychlinsky. 1999. The induction of apoptosis
Ohta, S., K. Niiya, N. Sakuragawa, and H. Fuse. 2000. Induction of by bacterial pathogens. Annu. Rev. Microbiol. 53:155–187.
urokinase-type plasminogen activator by lipopolysaccharide in Wesson, C. A., J. Deringer, L. E. Liou, K. W. Bayles, G. A. Bohach,
PC-3 human prostatic cancer cells. Thromb. Res. 97:343–347. and W. R. Trumble. 2000. Apoptosis induced by Staphylococcus
Oliver, S. P., and L. F. Calvinho. 1995. Influence of inflammation on aureus in epithelial cells utilizes a mechanism involving Cas-
mammary gland metabolism and milk composition. J. Anim. pases 8 and 3. Infect. Immun. 68:2998–3001.
Sci. 73(Suppl. 2):18–33. Wesson, C. A., L. E. Liou, K. M. Todd, G. A. Bohach, W. R. Trumble,
Owen, C. A., and E. J. Campbell. 1999. The cell biology of leukocyte and K. Bayles. 1998. Staphylococcus aureus Agr and Sar global
mediated proteolysis. J. Leukoc. Biol. 65:137–150. regulators influence internalization and induction of apoptosis.
Paape, M., D. Bannerman, X. Zhao, and J. Lee. 2003. The bovine Infect. Immun. 66:5238–5243.
neutrophil: Structure and function in blood and milk. Vet. Res. Wilson, D. J., R. N. Gonzales, and H. H. Das. 1997. Bovine mastitis
34:597–627. pathogens in New York and Pennsylvania: Prevalence and ef-
Paape, M., J. Mehrzad, X. Zhao, J. Detilleux, and C. Burvenich. 2002. fects on somatic cell count and milk production. J. Dairy Sci.
Defense of the bovine mammary gland by polymorphonuclear 80:2592–2598.

Vous aimerez peut-être aussi