Skip to main content

REVIEW article

Front. Vet. Sci., 06 March 2024
Sec. Veterinary Infectious Diseases

Mycoplasma bovis mastitis in dairy cattle

  • 1Department of Animal Science, The University of Tennessee, Knoxville, TN, United States
  • 2College of Veterinary Medicine, The University of Tennessee, Knoxville, TN, United States

Mycoplasma bovis has recently been identified increasingly in dairy cows causing huge economic losses to the dairy industry. M. bovis is a causative agent for mastitis, pneumonia, endometritis, endocarditis, arthritis, otitis media, and many other clinical symptoms in cattle. However, some infected cows are asymptomatic or may not shed the pathogen for weeks to years. This characteristic of M. bovis, along with the lack of adequate testing and identification methods in many parts of the world until recently, has allowed the M. bovis to be largely undetected despite its increased prevalence in dairy farms. Due to growing levels of antimicrobial resistance among wild-type M. bovis isolates and lack of cell walls in mycoplasmas that enable them to be intrinsically resistant to beta-lactam antibiotics that are widely used in dairy farms, there is no effective treatment for M. bovis mastitis. Similarly, there is no commercially available effective vaccine for M. bovis mastitis. The major constraint to developing effective intervention tools is limited knowledge of the virulence factors and mechanisms of the pathogenesis of M. bovis mastitis. There is lack of quick and reliable diagnostic methods with high specificity and sensitivity for M. bovis. This review is a summary of the current state of knowledge of the virulence factors, pathogenesis, clinical manifestations, diagnosis, and control of M. bovis mastitis in dairy cows.

1 Introduction

Mycoplasma bovis, which was formerly known as Mycoplasma agalactiae subsp. bovis (1), is a causative agent of several diseases in cattle and other farmed ruminants including mastitis, pneumonia, endocarditis, arthritis, otitis, meningitis, and reproductive problems both in bulls and cows (26). M. bovis mastitis is an emerging dairy cattle disease that poses a significant challenge globally due to its highly contagious nature and resistance to antimicrobials (7). Despite the isolation of various Mycoplasma spp. from milk samples of cows with mastitis, M. bovis is the most common causative agent (8, 9).

In the United States of America (U.S.) alone, economic losses due to M. bovis mastitis is estimated to be above $100 million annually (10). This mainly results from increased somatic cell counts (SCC), decreased milk production, culling, and treatment costs. Since SCC is an indicator of milk quality and udder health, SCC beyond legal limits in the U.S. results in regulatory measures such as suspension of operation permits (11). Culling of infected cows has proven costly in the U.S. with the rate reaching as high as 70% (12). The prevalence of M. bovis mastitis is high in large dairy operations especially in the U.S. dairy industry where several large scale dairy operations exist (13, 14). Currently adopted mastitis control programs which involve environmental sanitation, proper milking procedures and udder health (15) are partly ineffective due to transmission of M. bovis through respiratory route and semen or seminal fluid (16, 17) as well as calf to cow or vice-versa (18). Given the difficulty in diagnosis due to the fastidious growth nature and subclinical infection, with infected cows apparently appearing healthy, the disease can remain undetected for long time in dairy farms with significant economic losses (19).

Although M. bovis mastitis was first reported in the early 1960s (2) and has since been problematic, effective control tools such as vaccine or prophylactic therapy or treatment is yet to be developed. This is largely attributed to the knowledge gap in the critically important virulence factors and pathogenesis mechanisms of M. bovis mastitis.

With increasing antimicrobial resistance problem (20), accurate diagnosis is crucial to avoid the use of broad-spectrum antimicrobial drugs and to conduct targeted treatment. Among the challenges in the control and prevention of M. bovis infections is developing effective and sustainable control tools and rapid and reliable pen-side diagnostic tool with high specificity and sensitivity that can be used at farm level (21). This review is a succinct summary of current state of knowledge in virulence factors, pathogenesis, clinical manifestations, diagnosis, and control measures of M. bovis mastitis in dairy cows.

2 Virulence factors

2.1 Adhesion and invasion

Mycoplasmas lack cell walls and have exposed membrane proteins. The exposed membrane proteins primarily interact with host surfaces and enable the bacteria to adhere to the host mucosal surfaces and are also necessary for the bacteria to acquire nutrients from their surroundings and evade their host’s immune response (22).

Adhesion is an essential virulence attribute in mycoplasmas since adhesion mutants are avirulent (23). M. bovis utilizes a 48.8 kDa receptor TrmFO that binds to host fibronectin, an extracellular matrix glycoprotein (24). Other isolates also express key adhesins such as α-enolase, a hypothetical lipoprotein with adhesin activity (P27), variable surface lipoprotein A (VpmaX), and fructose-1,6-biophasphate aldolase (25, 26). A cytoadhesive surface exposed protein in certain strains is expressed to surmount the highly tight epithelial junctions during infection in organs such as lung (27). Attachment only, however, does not constitute internalization as some M. bovis cells were shown adhered onto and others internalized into calf turbinate cells in a single in vitro infection (28, 29). Burki and co-authors showed that M. bovis enters host cells through non-classical endocytic pathway (28) which involves invagination of plasma membrane to internalize pathogens (30). This pathway is used by host cells to uptake various fluids and solutes but M. bovis takes this to its advantage. Autophagy is a highly conserved self-destructive process in eukaryotic cells aimed to remove faulty organelles, misfolded proteins and pathogens (31). However, M. bovis prevents autophagy in bovine mammary epithelial cells to replicate in an intracellular environment while also avoiding clearance by host immune responses and antimicrobial agents (32, 33). M. bovis exerts this effect through blocking autophagic flux which involves recognition of intracellular M. bovis by receptors, delivery to enzyme-bound membranes and final transport to lysosomes, a degradation machinery (33).

2.2 Variable surface proteins

Among the characteristics that increases the virulence of M. bovis is the collection of immunodominant variable surface proteins (Vsps). These surface lipoproteins are highly variable in their size and coding sequences (34, 35). Due to different surface lipoprotein variants potentially interacting with the host immune system at any given time, immune responses against these Vsps are not effective. Furthermore, the pathogenicity of M. bovis is significantly increased because these Vsps enable it to avoid detection and clearance by a host immune system (36).

2.2.1 Nucleases

Mycoplasmas do not have biosynthetic mechanisms to synthesize nucleic acid precursors and depend on cellular nucleases to generate nucleotide precursors (37). Nucleases are components of the mycoplasmal membrane that hydrolytically cleave the phosphodiester backbone of DNA and they play important role in acquisition of the host nucleic acids. Various Mycoplasma cellular nucleases have been characterized which are believed to be important for generating nucleotides and hence expected to contribute to virulence (38). Endonucleases cleave the phosphodiester bond in the middle of chains within the polynucleotide whereas the exonucleases selectively cleave the polynucleotide chain either at the 5′ or 3′ ends (39).

2.2.2 Biofilm formation

It has been demonstrated that M. bovis produces biofilms; however, the level of adhesion and effectiveness of the biofilms vary between different strains depending on the surface lipoprotein (Vsp) expression. Biofilms of M. bovis increase heat resistance at 50°C and desiccation tolerance but are not any more resistant to antimicrobials compared to planktonic cells (40).

2.2.3 Nucleomodulin secretion

Nucleomodulins are effector proteins secreted by bacteria that can interact with the host DNA and serve to regulate gene transcription to favor the pathogenesis of the bacteria (41). The MbovP475 lipoprotein is secreted by M. bovis and binds the promoters of the cell cycle central regulatory genes, CRYAB and MCF2L2 genes and downregulates their expression in bovine macrophage cell line (42) resulting in decreases in bovine macrophage cell line viability.

2.3 Metabolites

M. bovis synthesizes hydrogen peroxide (H2O2) which has the potential to react with iron and copper ions to produce cytotoxic hydroxyl radicals (43). This is possible through the NADH oxidase enzyme expressed by M. bovis which reduce oxygen to H2O2 on top of its adhesin role (44). Reactive oxygen species produced by M. bovis can cause varying degree of damage in bovine mammary epithelial cells including apoptosis (45).

3 Pathogenesis of Mycoplasma bovis mastitis and clinical symptoms

The most common clinical manifestations of M. bovis mastitis includes udder swelling and abnormal milk appearance ranging from watery and flaky milk to thick purulent inflammatory fluid (46). However, some cows may show no outward symptoms although having subclinical mastitis with or without shedding the bacterium (4750). M. bovis mastitis causes an increase in SCC. Among the initial reactions during intramammary challenge infection include high SCC and acute phase proteins (51). These authors reported increased production of serum amyloid A and lipopolysaccharide -binding protein in experimentally induced M. bovis intramammary infection. It is possible that M. bovis spreads from one site of infection to another via blood circulation. This was demonstrated by isolation of M. bovis from previously uninfected cows which were challenged experimentally by the intramammary route (46). Although shedding and re-infection is a possibility due to contagious nature of the organism, M. bovis from a milk of mastitic cows had identical pulsed field gel electrophoresis pattern with those isolated from other body parts such as eyes, nasal cavities and ears which is strong indication of internal dissemination (52).

Transmission of M. bovis has been linked to colostrum, milk, semen, air-borne, and intrauterine routes (16, 5355). Udder-to-udder is thought to be the primary route by which the infection is transmitted between cows (12, 56). Although M. bovis intramammary infection is widespread in lactating dairy cows, it is also not uncommon in dry cows (57).

Several aspects of M. bovis mastitis differs from other major mastitis pathogens. Unlike coliform mastitis, which is environmental, M. bovis mastitis is contagious which means it can be transmitted from infected cows to healthy cows during milking time. The major contagious mastitis pathogens include Staphylococcus aureus, Streptococcus agalactiae, and Mycoplasma spp. (13). Compared to Staphylococcus aureus and E. coli, M. bovis weakly affects mRNA expression in bovine mammary epithelial cells (58). According to the USDA, M. bovis is more prevalent in large (500+) dairy operations compared to other contagious mastitis pathogens such as Streptococcus agalactiae (13).

4 Host immune responses against Mycoplasma bovis infection

4.1 Innate immunity

Epithelial cells are the major cell types in the bovine mammary gland and are the first line of defense. Upon first encounter, M. bovis adhere to and invade mammary gland epithelial cells (59) and bovine mammary epithelial cells respond by upregulated expression of proinflammatory cytokines such as interleukin (IL)-6 and IL-8 and TNF-α (58, 60). Autophagy is one of the mechanisms these cells employ to eliminate invading bacteria, however under-expression of autophagy related proteins has been demonstrated in M. bovis infected bovine mammary epithelial cells (32, 33).

M. bovis mastitis is characterized by massive recruitment of neutrophils into the milk spaces of the mammary gland (61). Similarly, M. bovis has been shown to stimulate production of neutrophils extracellular traps (NETs) and possess the membrane nuclease, MnuA which degrades the NETs through either exo- or endonuclease activity (38, 62). In addition, another membrane nuclease of M. bovis (MBOV_RS02825) has been demonstrated to degrade NETs and cause apoptosis in macrophages (63). NETs are considered part of the innate immune response which have DNA as a major structural component to trap and stop pathogenic bacteria from spreading (64). Degradation of the NETs by mycoplasmal nucleases have a bifold advantage including avoiding the entrapment and opsonophagocytic killing by neutrophils and scavenging the nucleotide precursors (65). Furthermore, M. bovis promote neutrophil apoptosis to ensure its persistence and systemic dissemination.

M. bovis has been observed to elicit proinflammatory cytokine and chemokine responses in infected hosts which may weaken the host and increase the pathogenicity of the bacteria (66). Opsonization is necessary for phagocytosis of M. bovis by macrophages and neutrophils, but M. bovis can combat this through surface antigen variation, and biofilm formation (40). Macrophages kill M. bovis via phagocytosis up on opsonization support from IgG1 and IgG2 (67). In a study conducted to determine M. bovis-bovine viral diarrhea virus (BVDV) synergism during infection, M. bovis induced apoptosis and cytotoxicity in bovine macrophages (68). In contrary, another study reported significant reduction in apoptosis in macrophages induced by M. bovis possibly as a mechanism of survival (69).

M. bovis inhibits proliferation of peripheral blood mononuclear cells (PBMCs) to evade the immune system and cause chronic infections (70). However, in contrary another study reported an increase in the expression of TNF-α, IL-12 and IFN-γ and increased proliferative responses in PBMCs stimulated with M. bovis (71). Following intramammary inoculation with M. bovis, milk from infected quarters exhibited increased SCCs, yet there was not a significant difference between levels of PBMCs or mononuclear cells in the stimulated and unstimulated mammary lymph nodes (72). In fact, there was a decrease in the mRNA levels of innate-immunity related genes from blood mononuclear cells following intramammary infection with M. bovis, such as complement factor D (CFD), ficolin 1 (FCN1), and tumor necrosis factor superfamily member 13 (TNFSF13) (72). This alteration of the host transcriptome likely contributes to the chronic nature of many M. bovis infections.

4.2 Adaptive immunity

M. bovis antigens activate host CD4+, CD8+, γδ T- cells, B- cells, and leukocytosis (73, 74). In addition, M. bovis also induces IgG1and IgG2 responses (75). IgG1, however, has a lower opsonin effect which does not activate a strong humoral immune response in the host and resulting in persistence of M. bovis infections for long periods of time (76). The effect of cytokines, such as interferon gamma (IFN-γ), that encourage cell death but also cytokines that characterize the Th2 response and slow recovery of tissues likely contribute to the pathogenicity of M. bovis and make it more difficult for the host to recover from infections (77).

5 Diagnosis of Mycoplasma bovis

5.1 Culture

Microbial culture has traditionally been used to definitively diagnose M. bovis. However, the longer time it takes to grow makes culture unfavorable to make a rapid diagnosis. Most common specimens to diagnose M. bovis includes milk, bronchioalveolar lavage, deep nasopharyngeal swabs, joint fluids, and semen. Mycoplasma plates are incubated at 37°C in 5% CO2 for 7–10 days and colonies are typically characterized by a ‘fried egg appearance’ when observed under light microscope (78). This is unfavorable where rapid diagnosis is needed to isolate infected animals, limit further dissemination of infections and commence appropriate antibiotic therapy (79). Due to the longer days required by M. bovis, sometimes the colonies are overgrown by other bacteria to the extent that they cover M. bovis colonies and makes it difficult to observe under microscope (80). Not only they are overgrown on the plates, but they are also overgrown in the milk since mycoplasmas have limited capability to multiply in milk (81).

Mycoplasmas have one of the smallest genome sizes (0.58–1.38 Mbp) which likely renders them needy of nutrients such as amino acids and fatty acids (22, 82). Another challenge with M. bovis culture is its detection limit which is greater than or equal to 272 CFU/mL (83) meaning any number of mycoplasma cells less than the detection limit could possibly be overlooked. Culture also fails to differentiate other non-pathogenic mollicutes such as Acholeplasma which exhibit same ‘fried egg’ appearance as M. bovis and can potentially contaminate M. bovis samples (84).

Bulk tank milk samples were used in several studies to estimate prevalence and other research purposes (8588). The major problem with bulk tank milk sampling is collecting a representative amount of sample out of 200 to 2,600 gallons of tanks which could massively dilute the amount of Mycoplasma cells. Usually, about 10–40 mL of milk sample is collected (85, 87), centrifuged and only 100–200 μL are spread on the Mycoplasma plates. Some laboratories culture milk samples directly (12), however it has been demonstrated that centrifugation and resuspension can potentially increase the rate of recovery (89). Occasionally, enrichment of milk samples in Mycoplasma broth before culture is practiced (90). Milk samples need to be processed immediately after collection to maximize the likelihood of positive diagnosis or need to be frozen although freezing has been shown to cause 1–2 log10 reduction (91). In addition, multiple sampling is greatly advised due to intermittent shedding behavior of the pathogen in mastitis cases (49, 92). However, there are also studies which were conducted based on one-time bulk tank milk sampling (85, 93).

5.2 Polymerase chain reaction

PCR is such a sensitive method in M. bovis diagnosis that it detects as low as 10 CFUs from broth cultures (94, 95). Compared to culture, enzyme-linked immunosorbent assay (ELISA), sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE), and nucleic acid hybridization, PCR was found to be superior in terms of high specificity, sensitivity and rapidity (96). However, expensive reagents and equipment are the major setbacks to apply in small-scale laboratories and farm environments. The application of PCR in M. bovis diagnosis brought about several advantages. It bypasses the tiresome culturing and samples can directly be screened for M. bovis DNA. In comparison to the culture method which only detects live organisms, PCR can detect the DNA from dead microorganisms which can have several implications. This is true, for instance, perhaps if milk samples are stored in a freezer for long time and M. bovis cells are no longer viable (91). In some cases, pre-enrichment is recommended before performing PCR assays for as many as 4 days (97). This might save time and resource in terms of avoiding culturing negative samples, however is not helpful when rapid diagnosis is needed.

UvrC gene has widely been used as a target in conventional and real-time PCR in M. bovis detection in bulk tank milk and lung samples (87, 98, 99). It is one of housekeeping genes in M. bovis which encodes an enzyme that mediates excision DNA repair system (82, 100). Furthermore, this gene has been recommended for use in routine laboratory diagnosis of M. bovis (101). Realtime PCR has been shown to be more sensitive than conventional PCR and was able to amplify as low as 40 copies of the target gene (98). The CT values in real-time and quantitative PCR (qPCR) can be used as a predictive tool for M. bovis isolation (97).

A multiplex PCR which separately detects different species such as M. bovis, M. bovigenitalium, and M. californicum in a same sample which would greatly reduce efforts and time spent to perform multiple reactions (102). Furthermore, bacterial species such as Mannheimia hemolytica are usually isolated alongside M. bovis in respiratory diseases (103). Multiplex quantitative PCR has been proven to specifically detect and quantify these respiratory pathogens (104, 105). Nested-PCR, where two sets of primers are employed, is preferred to increase sensitivity and specificity of Mycoplasma detection (106, 107). PCR has also been employed to identify antibiotic resistance genes in M. bovis (108, 109).

5.3 Indirect and direct ELISA

Unlike culture and PCR, ELISA detects the anti-M. bovis antibodies in the host serum or milk from past or recent infections as a result of humoral immune responses. ELISA has widely been used on bulk tank milk samples to detect anti-M. bovis antibodies in the milk for diagnostic, prevalence, and retrospective studies (85, 88, 110, 111). A MilA ELISA with a specificity and sensitivity as high as 94.2 and 96.6%, respectively, has been used to estimate M. bovis mastitis prevalence from bulk tank milk samples (112). This ELISA, in which MilA membrane protein was used as a coating antigen, was first developed in Australia (113). Furthermore, the milA gene was expressed on the surface of a phage and was used as antigen in indirect ELISA which was reported to be inexpensive and convenient compared to the MilA peptide protein (114). However, bulk tank milk is not a fully representative sample for a herd since M. bovis can cause various disease manifestations in various age groups (115). This necessitates considering blood samples in certain cases where the young stock is suspected to harbor the infection or they are being newly introduced to herd. ELISA might not necessarily indicate active M. bovis infection as positive ELISA results turns negative in PCR when both methods were used in same herds (85, 116). Thus, ELISA can be used as a biosecurity tool before introducing newcomers to the herd (110) and as a surveillance tool to monitor and confirm eradication of M. bovis infections in some nations (117). It also plays role in testing immunogenicity of novel proteins in a study conducted to investigate M. bovis pathogenesis and its protective antigens (118).

The use of Indirect ELISA to detect anti-M. bovis antibodies is challenged by cross-reactivity from other Mycoplasma spp. such as M. agalactiae (119). Indirect ELISA might not be as rapid as desired sometimes, since it takes 1–2 weeks for the animal to mount humoral immune response (seroconversion) and results could possibly turn negative in this time window (78), thus it needs to be used in conjunction with other tests. Once antibodies are produced by the host, however, ELSIA is not affected by the intermittent shedding behavior of M. bovis in the milk which indicates it is important to use culture, PCR and/or ELISA together, whenever possible, since they complement each other. Unlike indirect ELISA, the use of direct ELISA in M. bovis diagnosis is limited. There are not many direct ELISAs reported, however one study reported membrane protein P48 based monoclonal antibodies has been shown to specifically detect M. bovis without cross-reactivity with related species such as M. agalactiae (120).

5.4 MALDI-TOF MS

Matrix-Assisted Laser Desorption Ionization – Time of Flight Mass Spectrometry (MALDI-TOF MS) has lately been adapted as a rapid tool to accurately diagnose microbes. The principle behind the method is formation of ions from intact bacterial cells using laser light ionization (121). Samples are obtained from colonies on agar plates, mixed with matrix solution, and introduced to a mass spectrometer ion source. Results are analyzed against archived references or with colonies from known bacteria. Following a comparison with the 16S rDNA PCR as gold standard, MALDI-TOF has been in agreement for 97.8% species-level and 99.6% genus-level for aerobes and 95.3% species-level and 100% genus-level for anaerobes which shows that it is a preferable method to diagnose bacteria of veterinary interest (122). The suitability of MALDI-TOF to distinguish between phylogenetically closest sub-species in human and ruminant mycoplasmas has been confirmed (123). Although some studies recommend MALDI-TOF as promising test for routine diagnosis of M. bovis, the disadvantage lies in its dependence on enrichment which can take 2 to 4 days (124, 125). The starting material for MALDI-TOF could also be a secretome extraction or a protein from M. bovis. Zubair et al. used extracted secretome from M. bovis HB0801 strain to predict 8 proteins related to a virulence which signifies the importance of MALDI-TOF in identifying potential diagnostic and vaccine targets (126).

Limited library of mycoplasmas in the MALDI-TOF databases has been reported as one of the challenges in using this method to detect M. bovis (125). Initial MALDI-TOF installation and instrumentation is also believed to be expensive which makes its application at large scale level difficult (127). Therefore, MALDI-TOF is commonly used more as a confirmatory technique rather than a routine diagnostic tool.

5.5 Loop-mediated isothermal amplification

The LAMP is a high sensitivity method which utilizes a set of 2 to 3 primers that can produce several copies (108) of target DNA in less than an hour since it bypasses the denaturation step (128). As the name indicates, LAMP amplification occurs within a constant temperature. Unlike the conventional PCR, LAMP does not need thermocyclers and can easily be done in heating block (129). Following amplification, results can be read in 2% agarose gel electrophoresis; using SYBR green I staining or based on turbidity of the reaction mixture (129, 130). UvrC-based LAMP in M. bovis has been demonstrated to have 10-fold higher sensitivity compared to PCR with 100 and 74% sensitivity and specificity, respectively, (129), however later on other researchers improved the specificity to 90.9% (130). Other M. bovis genes such as oppD (encodes oligopeptide permease D), gltX (glutamate transfer RNA ligase), gyrB (gyrase B subunit) and 16 s rRNA were also employed to show sensitive and specific detection of M. bovis using LAMP (131, 132).

6 Control and prevention measures

Globally, DNA amplification techniques used for detection and identification of bacteria have only become widely globally accessible within the last 30 years, making it difficult to trace the exact time and route by which M. bovis first spread around the world (94). The first definitive identification of M. bovis infection was in 1961 in the U.S. (2). From there, the pathogen was thought to have been spread to other countries through movement of cattle and cattle products (7).

The strategy toward the control and prevention of M. bovis mastitis, or M. bovis infection in general, depends on the country. New Zealand, which is the latest country to report M. bovis mastitis in 2017, prefers a nationwide complete eradication program (117). However, other endemic countries endeavor to contain infections at the farm level through culling or isolating infected animals (133). Finland, for example, pursues a voluntary control program involving farmers since M. bovis is regarded as one of less serious diseases (134).

Currently, the best-known method for controlling M. bovis is mere prevention of exposure to the pathogen and other infected cows. Screening of original herd before purchasing new cows is worthwhile as well as quarantine of new cows which adds extra layer of security. In addition, isolation and culling of infected cows are necessary measures to effectively control the disease and minimize outbreaks (42). However, advances in rapid and accessible tests to detect M. bovis on dairy farms are necessary to control and prevent outbreaks from occurring more effectively.

6.1 Use of antimicrobials

There are currently no known effective treatments against M. bovis available for use. One of the main reasons for this is the growing incidence of antimicrobial resistant bacterial strains. Drugs such as tiamulin, enrofloxacin, danofloxacin, and florfenicol has been reported to have low minimum inhibitory concentration against M. bovis (20). However, in the last 2 decades, M. bovis have shown less susceptibility to antimicrobial agents like fluoroquinolones (135). Furthermore, lack of cell wall makes the organism resistant to commonly used antimicrobials such as penicillin and cephalosporins. Macrolides such as tylosin and tilmicosin which were traditionally used to treat Mycoplasma infections has gradually become less effective (136). Recent trends indicate that antimicrobial resistance against other common antimicrobials, such as tetracyclines, has been increasing as broadly reviewed elsewhere (20). Some natural compounds have been shown to be promising and may further be developed to produce effective therapeutic options (137).

6.2 Vaccines

Many potential vaccine candidates have been developed but are not available for widespread commercial use. For example, autogenous vaccines have been developed (138); however, such vaccines are useful only for a single farm, limiting their potential for large-scale use. Other vaccines showed efficacy in studies but failed to elicit any protective effects in field trials as they failed to reduce the incidence of M. bovis cases (139, 140). There are currently no commercially available effective vaccines that prevent the incidence of M. bovis infection. Given the fact that M. bovis causes pneumonia in the feedlot cattle (141), considerable number of vaccine works has been done using feedlot cattle as a model. Prysliak and co-authors developed a sub-unit vaccine using the highly conserved glyceraldehyde-3-phosphate dehydrogenase (GAPDH) protein; however, subsequent controlled experimental efficacy evaluation showed that it did not confer protection against experimental challenge (142). Similarly, intranasal inoculation of total protein extract and membrane fractions from M. bovis triggered strong humoral immune responses but failed to protect against experimental challenge infection (143). Another challenge in vaccine development against M. bovis mastitis is strain variations of M. bovis (6). M. bovis also expresses antigenically variable surface proteins (34, 144) and thus necessitates developing vaccines from highly conserved immunogenic proteins. Despite several trials in the past, conserved immunogenic molecules which can elicit protective immune responses against M. bovis mastitis are yet to be identified.

7 Perspectives and future directions

In summary, effective control tools such as vaccine or prophylactic or therapeutic drugs against M. bovis mastitis are not available. Currently adopted mastitis control programs which involve environmental sanitation, proper milking procedures, and udder health are partly ineffective due to transmission of M. bovis through the respiratory route, semen or seminal fluid as well as calf to cow or vice-versa.

Rapid diagnosis is extremely important for the identification and culling of infected animals before infection spreads through the herd. This is particularly true in farm settings where purchased animals need to be screened or where segregation and culling of infected animals is much needed. Therefore, rapid and accurate pen-side diagnostic tests or combination of tests are needed. Given the difficulty in diagnosis due to the fastidious growth nature and subclinical infection, the disease can remain undetected for a long time in dairy farms with significant economic losses. Accurate diagnosis and antimicrobial susceptibility tests are also important to conduct targeted treatment which reduces the emergence of antimicrobial resistance. Whenever microbial culture is used for diagnosis, suspected animals awaiting results should be segregated from other herd members. Immediate culturing of milk samples is highly encouraged to increase the likelihood of detection since refrigeration and freezing lowers the survival of Mycoplasma cells (145). Cows with mastitis are usually asymptomatic and sometimes M. bovis is detected from healthy animals (146). This is also best exemplified by sudden occurrence of infection signs such as lameness and mastitis in yet closed dairy herds (147). Therefore, multiple sampling and regular screening of existing herd members is extremely important depending on how often new heifers are purchased. To overcome the problem of intermittent shedding of the Mycoplasma in the milk, indirect ELISA could be of great help since it depends on the antibodies rather than the detection of the antigen itself. MALDI-TOF is an advanced, rapid, and accurate method to detect M. bovis from various clinical samples. Its expensiveness and requiring sophisticated instrumentation and expertise renders it difficult to recommend for large-scale use. Finally, uniform recommendation should be put forward regarding which tests can be bundled together and yield complete diagnosis of M. bovis for different clinical samples collected from the animal body organs affected.

One of the major reasons for the failure to develop an effective control tool is limited knowledge of the virulence factors of M. bovis and the pathogenesis of M. bovis mastitis in dairy cows. Therefore, developing effective and sustainable control tools such as vaccines or prophylactic or therapeutic drugs, or any other innovative intervention tool using advanced molecular biology and cellular and molecular immunology approaches is required.

The combined economic and welfare impact of M. bovis infections prompted extensive search for effective and sustainable control tools such as vaccines, prophylactic and therapeutic solutions (21) while also antimicrobial resistance is increasing (20). Vaccine attempts has been futile due to mainly the knowledge gap in the pathogenesis and virulence mechanism of M. bovis. Novel vaccine or antimicrobial drugs will be out of reach if conserved immunogenic antigens or therapeutic agents are not discovered.

Hygienic husbandry practices during milking, feeding, and overall rearing is of paramount significance.

Author contributions

AG: Writing – review & editing, Writing – original draft. SD: Writing – review & editing, Writing – original draft. BG: Writing – review & editing. OK: Writing – review & editing, Conceptualization, Supervision.

Funding

The author (s) declare that no financial support was received for the research, authorship, and/or publication of this article.

Acknowledgments

The authors thank UT Dairy Heath Research Group for their constructive comments and suggestions.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Askaa, G, and Ernø, H. Note: elevation of Mycoplasma agalactiae subsp. bovis to species rank: Mycoplasma boυis (Hale et al.) comb. nov. Int J Syst Evol Microbiol. (1976) 26:323–5.

Google Scholar

2. Hale, HH, Helmboldt, CF, Plastridge, WN, and Stula, EF. Bovine mastitis caused by a Mycoplasma species. Cornell Vet. (1962) 52:582–91.

Google Scholar

3. Suwanruengsri, M, Uemura, R, Kanda, T, Fuke, N, Nueangphuet, P, Pornthummawat, A, et al. Production of granulomas in Mycoplasma bovis infection associated with meningitis-meningoencephalitis, endocarditis, and pneumonia in cattle. J Vet Diagn Invest. (2022) 34:68–76. doi: 10.1177/10406387211053254

PubMed Abstract | Crossref Full Text | Google Scholar

4. Foster, AP, Naylor, RD, Howie, NM, Nicholas, RA, and Ayling, RD. Mycoplasma bovis and otitis in dairy calves in the United Kingdom. Vet J. (2009) 179:455–7. doi: 10.1016/j.tvjl.2007.10.020

PubMed Abstract | Crossref Full Text | Google Scholar

5. Kanda, T, Tanaka, S, Suwanruengsri, M, Sukmawinata, E, Uemura, R, Yamaguchi, R, et al. Bovine endocarditis associated with Mycoplasma bovis. J Comp Pathol. (2019) 171:53–8. doi: 10.1016/j.jcpa.2019.07.003

PubMed Abstract | Crossref Full Text | Google Scholar

6. Aebi, M, Bodmer, M, Frey, J, and Pilo, P. Herd-specific strains of Mycoplasma bovis in outbreaks of mycoplasmal mastitis and pneumonia. Vet Microbiol. (2012) 157:363–8. doi: 10.1016/j.vetmic.2012.01.006

PubMed Abstract | Crossref Full Text | Google Scholar

7. Nicholas, RA . Bovine mycoplasmosis: silent and deadly. Vet Rec. (2011) 168:459–62. doi: 10.1136/vr.d2468

PubMed Abstract | Crossref Full Text | Google Scholar

8. Allan, M, Britten, EDT, and Justine, E. Britten, editor why species matter? Dramatic Revelations in Mycoplasma Mastitis Management with Molecular Diagnostics. NMC Annual Meeting Proceedings; (2020).

Google Scholar

9. Gioia, G, Addis, MF, Santisteban, C, Gross, B, Nydam, DV, Sipka, AS, et al. Mycoplasma species isolated from bovine milk collected from US dairy herds between 2016 and 2019. J Dairy Sci. (2021) 104:4813–21. doi: 10.3168/jds.2020-19171

PubMed Abstract | Crossref Full Text | Google Scholar

10. Nicholas, RA, and Ayling, RD. Mycoplasma bovis: disease, diagnosis, and control. Res Vet Sci. (2003) 74:105–12. doi: 10.1016/S0034-5288(02)00155-8

Crossref Full Text | Google Scholar

11. USDA-APHIS–VS–CEAH–NAHMS. Determining U.S. Milk quality using bulk tank somatic cell counts, 2019, Info Sheet, Fort Collins, CO. (2021). Available at: https://www.aphis.usda.gov/animal_health/nahms/dairy/downloads/dairy_monitoring/btscc_2019infosheet.pdf (Accessed February 24, 2024).

Google Scholar

12. Gonzalez, RN, Sears, PM, Merrill, RA, and Hayes, GL. Mastitis due to Mycoplasma in the state of New York during the period 1972-1990. Cornell Vet. (1992) 82:29–40.

Google Scholar

13. USDA-APHIS. Prevalence of contagious mastitis pathogens on U.S. dairy operations In: Agriculture. USA: Department of Agriculture (2008)

Google Scholar

14. Fernando Ulloa, DB, Jara, Ronald, García, Eduardo, Kruze, Juan, and Mella, Armin, editor. Case report: Mycoplasma mastitis outbreaks in two Chilean dairy herds. NMC Annual Meeting Proceedings; (2021).

Google Scholar

15. National Mastitis Council (NMC). Recommended Mastitis Control Program – North American version. Documents and reports, New Prague, MN. (2022). Available at: https://www.nmconline.org/documents/ (Accessed February 25, 2024).

Google Scholar

16. Haapala, V, Pohjanvirta, T, Vähänikkilä, N, Halkilahti, J, Simonen, H, Pelkonen, S, et al. Semen as a source of Mycoplasma bovis mastitis in dairy herds. Vet Microbiol. (2018) 216:60–6. doi: 10.1016/j.vetmic.2018.02.005

PubMed Abstract | Crossref Full Text | Google Scholar

17. Pardon, B , editor. Mastitis pathogens revisited: Mycoplasma spp. NMC annual meeting proceedings. (2020); 2020.

Google Scholar

18. Timonen, AAE, Autio, T, Pohjanvirta, T, Häkkinen, L, Katholm, J, Petersen, A, et al. Dynamics of the within-herd prevalence of Mycoplasma bovis intramammary infection in endemically infected dairy herds. Vet Microbiol. (2020) 242:108608. doi: 10.1016/j.vetmic.2020.108608

PubMed Abstract | Crossref Full Text | Google Scholar

19. Nicholas, RA, Fox, LK, and Lysnyansky, I. Mycoplasma mastitis in cattle: to cull or not to cull. Vet J. (2016) 216:142–7. doi: 10.1016/j.tvjl.2016.08.001

Crossref Full Text | Google Scholar

20. Lysnyansky, I, and Ayling, RD. Mycoplasma bovis: mechanisms of resistance and trends in antimicrobial susceptibility. Front Microbiol. (2016) 7:595. doi: 10.3389/fmicb.2016.00595

Crossref Full Text | Google Scholar

21. Calcutt, MJ, Lysnyansky, I, Sachse, K, Fox, LK, Nicholas, RAJ, and Ayling, RD. Gap analysis of Mycoplasma bovis disease, diagnosis and control: an aid to identify future development requirements. Transbound Emerg Dis. (2018) 65:91–109. doi: 10.1111/tbed.12860

PubMed Abstract | Crossref Full Text | Google Scholar

22. Razin, S, Yogev, D, and Naot, Y. Molecular biology and pathogenicity of mycoplasmas. Microbiol Mol Biol Rev. (1998) 62:1094–156. doi: 10.1128/MMBR.62.4.1094-1156.1998

PubMed Abstract | Crossref Full Text | Google Scholar

23. Krause, DC, Leith, DK, Wilson, RM, and Baseman, JB. Identification of Mycoplasma pneumoniae proteins associated with hemadsorption and virulence. Infect Immun. (1982) 35:809–17. doi: 10.1128/iai.35.3.809-817.1982

PubMed Abstract | Crossref Full Text | Google Scholar

24. Guo, Y, Zhu, H, Wang, J, Huang, J, Khan, FA, Zhang, J, et al. Trm FO, a fibronectin-binding Adhesin of Mycoplasma bovis. Int J Mol Sci. (2017) 18:1732. doi: 10.3390/ijms18081732

PubMed Abstract | Crossref Full Text | Google Scholar

25. Shitamori, F, Uemura, R, Kanda, T, and Sueyoshi, M. The presence of adhesion factors NOX, α-enolase, Trm FO, P 27, and Vpma X in Mycoplasma bovis wild isolates in Japan. Open Vet J. (2022) 12:782–6. doi: 10.5455/OVJ.2022.v12.i6.1

PubMed Abstract | Crossref Full Text | Google Scholar

26. Huang, J, Zhu, H, Wang, J, Guo, Y, Zhi, Y, Wei, H, et al. Fructose-1, 6-bisphosphate aldolase is involved in Mycoplasma bovis colonization as a fibronectin-binding adhesin. Res Vet Sci. (2019) 124:70–8. doi: 10.1016/j.rvsc.2019.02.010

PubMed Abstract | Crossref Full Text | Google Scholar

27. Zhu, X, Dong, Y, Baranowski, E, Li, X, Zhao, G, Hao, Z, et al. Mbov_0503 encodes a novel Cytoadhesin that facilitates Mycoplasma bovis interaction with tight junctions. Microorganisms. (2020) 8. doi: 10.3390/microorganisms8020164

PubMed Abstract | Crossref Full Text | Google Scholar

28. Bürki, S, Gaschen, V, Stoffel, MH, Stojiljkovic, A, Frey, J, Kuehni-Boghenbor, K, et al. Invasion and persistence of Mycoplasma bovis in embryonic calf turbinate cells. Vet Res. (2015) 46:53. doi: 10.1186/s13567-015-0194-z

PubMed Abstract | Crossref Full Text | Google Scholar

29. Rottem, S . Interaction of mycoplasmas with host cells. Physiol Rev. (2003) 83:417–32. doi: 10.1152/physrev.00030.2002

Crossref Full Text | Google Scholar

30. Cossart, P, and Helenius, A. Endocytosis of viruses and bacteria. Cold Spring Harb Perspect Biol. (2014) 6:972. doi: 10.1101/cshperspect.a016972

Crossref Full Text | Google Scholar

31. Glick, D, Barth, S, and Macleod, KF. Autophagy: cellular and molecular mechanisms. J Pathol. (2010) 221:3–12. doi: 10.1002/path.2697

PubMed Abstract | Crossref Full Text | Google Scholar

32. Xu, M, Liu, Y, Mayinuer, T, Lin, Y, Wang, Y, Gao, J, et al. Mycoplasma bovis inhibits autophagy in bovine mammary epithelial cells via a PTEN/PI3K-Akt-mTOR-dependent pathway. Front Microbiol. (2022) 13:935547. doi: 10.3389/fmicb.2022.935547

PubMed Abstract | Crossref Full Text | Google Scholar

33. Liu, Y, Deng, Z, Xu, S, Liu, G, Lin, Y, Khan, S, et al. Mycoplasma bovis subverts autophagy to promote intracellular replication in bovine mammary epithelial cells cultured in vitro. Vet Res. (2021) 52:130. doi: 10.1186/s13567-021-01002-z

PubMed Abstract | Crossref Full Text | Google Scholar

34. Lysnyansky, I, Rosengarten, R, and Yogev, D. Phenotypic switching of variable surface lipoproteins in Mycoplasma bovis involves high-frequency chromosomal rearrangements. J Bacteriol. (1996) 178:5395–401. doi: 10.1128/jb.178.18.5395-5401.1996

PubMed Abstract | Crossref Full Text | Google Scholar

35. Beier, T, Hotzel, H, Lysnyansky, I, Grajetzki, C, Heller, M, Rabeling, B, et al. Intraspecies polymorphism of vsp genes and expression profiles of variable surface protein antigens (Vsps) in field isolates of Mycoplasma bovis. Vet Microbiol. (1998) 63:189–203. doi: 10.1016/S0378-1135(98)00238-7

PubMed Abstract | Crossref Full Text | Google Scholar

36. Buchenau, I, Poumarat, F, Le Grand, D, Linkner, H, Rosengarten, R, and Hewicker-Trautwein, M. Expression of Mycoplasma bovis variable surface membrane proteins in the respiratory tract of calves after experimental infection with a clonal variant of Mycoplasma bovis type strain PG45. Res Vet Sci. (2010) 89:223–9. doi: 10.1016/j.rvsc.2010.03.014

PubMed Abstract | Crossref Full Text | Google Scholar

37. Minion, FC, Jarvill-Taylor, KJ, Billings, DE, and Tigges, E. Membrane-associated nuclease activities in mycoplasmas. J Bacteriol. (1993) 175:7842–7. doi: 10.1128/jb.175.24.7842-7847.1993

PubMed Abstract | Crossref Full Text | Google Scholar

38. Sharma, S, Tivendale, KA, Markham, PF, and Browning, GF. Disruption of the membrane nuclease gene (MBOVPG45_0215) of Mycoplasma bovis greatly reduces cellular nuclease activity. J Bacteriol. (2015) 197:1549–58. doi: 10.1128/JB.00034-15

PubMed Abstract | Crossref Full Text | Google Scholar

39. Mason, PA, and Cox, LS. The role of DNA exonucleases in protecting genome stability and their impact on ageing. Age (Dordr). (2012) 34:1317–40. doi: 10.1007/s11357-011-9306-5

PubMed Abstract | Crossref Full Text | Google Scholar

40. McAuliffe, L, Ellis, RJ, Miles, K, Ayling, RD, and Nicholas, RAJ. Biofilm formation by Mycoplasma species and its role in environmental persistence and survival. Microbiology. (2006) 152:913–22. doi: 10.1099/mic.0.28604-0

PubMed Abstract | Crossref Full Text | Google Scholar

41. Hanford, HE, Von Dwingelo, J, and Abu, KY. Bacterial nucleomodulins: a coevolutionary adaptation to the eukaryotic command center. PLoS Pathog. (2021) 17:e1009184. doi: 10.1371/journal.ppat.1009184

PubMed Abstract | Crossref Full Text | Google Scholar

42. Zhao, G, Lu, D, Wang, S, Zhang, H, Zhu, X, Hao, Z, et al. Novel mycoplasma nucleomodulin Mbov P 475 decreased cell viability by regulating expression of CRYAB and MCF2L2. Virulence. (2022) 13:1590–613. doi: 10.1080/21505594.2022.2117762

PubMed Abstract | Crossref Full Text | Google Scholar

43. Khan, LA, Miles, RJ, and Nicholas, RA. Hydrogen peroxide production by Mycoplasma bovis and Mycoplasma agalactiae and effect of in vitro passage on a Mycoplasma bovis strain producing high levels of H2O2. Vet Res Commun. (2005) 29:181–8. doi: 10.1023/B:VERC.0000047506.04096.06

PubMed Abstract | Crossref Full Text | Google Scholar

44. Zhao, G, Zhang, H, Chen, X, Zhu, X, Guo, Y, He, C, et al. Mycoplasma bovis NADH oxidase functions as both a NADH oxidizing and O (2) reducing enzyme and an adhesin. Sci Rep. (2017) 7:44. doi: 10.1038/s41598-017-00121-y

PubMed Abstract | Crossref Full Text | Google Scholar

45. Liu, Y, Zhou, M, Xu, S, Khan, MA, Shi, Y, Qu, W, et al. Mycoplasma bovis-generated reactive oxygen species and induced apoptosis in bovine mammary epithelial cell cultures. J Dairy Sci. (2020) 103:10429–45. doi: 10.3168/jds.2020-18599

PubMed Abstract | Crossref Full Text | Google Scholar

46. Bennett, RH, and Jasper, DE. Bovine mycoplasmal mastitis from intramammary inoculations of small numbers of Mycoplasma bovis: local and systemic antibody response. Am J Vet Res. (1980) 41:889–92.

Google Scholar

47. Hazelton, M, Morton, J, Parker, A, Sheehy, P, Bosward, K, Malmo, J, et al. Whole dairy herd sampling to detect subclinical intramammary Mycoplasma bovis infection after clinical mastitis outbreaks. Vet Microbiol. (2020) 244:108662. doi: 10.1016/j.vetmic.2020.108662

PubMed Abstract | Crossref Full Text | Google Scholar

48. Punyapornwithaya, V, Fox, LK, Hancock, DD, Gay, JM, and Alldredge, JR. Association between an outbreak strain causing Mycoplasma bovis mastitis and its asymptomatic carriage in the herd: a case study from Idaho, USA. Prev Vet Med. (2010) 93:66–70. doi: 10.1016/j.prevetmed.2009.08.008

Crossref Full Text | Google Scholar

49. Biddle, MK, Fox, LK, and Hancock, DD. Patterns of mycoplasma shedding in the milk of dairy cows with intramammary mycoplasma infection. J Am Vet Med Assoc. (2003) 223:1163–6. doi: 10.2460/javma.2003.223.1163

PubMed Abstract | Crossref Full Text | Google Scholar

50. Vähänikkilä, N, Pohjanvirta, T, Haapala, V, Simojoki, H, Soveri, T, Browning, GF, et al. Characterisation of the course of Mycoplasma bovis infection in naturally infected dairy herds. Vet Microbiol. (2019) 231:107–15. doi: 10.1016/j.vetmic.2019.03.007

PubMed Abstract | Crossref Full Text | Google Scholar

51. Kauf, AC, Rosenbusch, RF, Paape, MJ, and Bannerman, DD. Innate immune response to intramammary Mycoplasma bovis infection. J Dairy Sci. (2007) 90:3336–48. doi: 10.3168/jds.2007-0058

PubMed Abstract | Crossref Full Text | Google Scholar

52. Biddle, MK, Fox, LK, Evans, MA, and Gay, CC. Pulsed-field gel electrophoresis patterns of Mycoplasma isolates from various body sites in dairy cattle with Mycoplasma mastitis. J Am Vet Med Assoc. (2005) 227:455–9. doi: 10.2460/javma.2005.227.455

PubMed Abstract | Crossref Full Text | Google Scholar

53. Gille, L, Evrard, J, Callens, J, Supré, K, Grégoire, F, Boyen, F, et al. The presence of Mycoplasma bovis in colostrum. Vet Res. (2020) 51:54. doi: 10.1186/s13567-020-00778-w

PubMed Abstract | Crossref Full Text | Google Scholar

54. Hermeyer, K, Peters, M, Brügmann, M, Jacobsen, B, and Hewicker-Trautwein, M. Demonstration of Mycoplasma bovis by immunohistochemistry and in situ hybridization in an aborted bovine fetus and neonatal calf. J Vet Diagn Invest. (2012) 24:364–9. doi: 10.1177/1040638711435145

PubMed Abstract | Crossref Full Text | Google Scholar

55. Kanci, A, Wawegama, NK, Marenda, MS, Mansell, PD, Browning, GF, and Markham, PF. Reproduction of respiratory mycoplasmosis in calves by exposure to an aerosolised culture of Mycoplasma bovis. Vet Microbiol. (2017) 210:167–73. doi: 10.1016/j.vetmic.2017.09.013

Crossref Full Text | Google Scholar

56. González, RN, and Wilson, DJ. Mycoplasmal mastitis in dairy herds. Vet Clin North Am Food Anim Pract. (2003) 19:199–221. doi: 10.1016/S0749-0720(02)00076-2

Crossref Full Text | Google Scholar

57. Otter, A, Wright, T, Leonard, D, Richardson, M, and Ayling, R. Mycoplasma bovis mastitis in dry dairy cows. Vet Rec. (2015) 177:601–2. doi: 10.1136/vr.h6663

PubMed Abstract | Crossref Full Text | Google Scholar

58. Gondaira, S, Higuchi, H, Iwano, H, Nishi, K, Nebu, T, Nakajima, K, et al. Innate immune response of bovine mammary epithelial cells to Mycoplasma bovis. J Vet Sci. (2018) 19:79–87. doi: 10.4142/jvs.2018.19.1.79

PubMed Abstract | Crossref Full Text | Google Scholar

59. Josi, C, Bürki, S, Stojiljkovic, A, Wellnitz, O, Stoffel, MH, and Pilo, P. Bovine epithelial in vitro infection models for Mycoplasma bovis. Front Cell Infect Microbiol. (2018) 8:329. doi: 10.3389/fcimb.2018.00329

PubMed Abstract | Crossref Full Text | Google Scholar

60. Yang, J, Liu, Y, Lin, C, Yan, R, Li, Z, Chen, Q, et al. Regularity of toll-like receptors in bovine mammary epithelial cells induced by Mycoplasma bovis. Front Vet Sci. (2022) 9:846700. doi: 10.3389/fvets.2022.846700

PubMed Abstract | Crossref Full Text | Google Scholar

61. Schneider, P, Brill, R, Schouten, I, Nissim-Eliraz, E, Lysnyansky, I, and Shpigel, NY. Lipoproteins are potent activators of nuclear factor kappa B in mammary epithelial cells and virulence factors in Mycoplasma bovis mastitis. Microorganisms. (2022) 10, 1–14. doi: 10.3390/microorganisms10112209

PubMed Abstract | Crossref Full Text | Google Scholar

62. Mitiku, F, Hartley, CA, Sansom, FM, Coombe, JE, Mansell, PD, Beggs, DS, et al. The major membrane nuclease Mnu a degrades neutrophil extracellular traps induced by Mycoplasma bovis. Vet Microbiol. (2018) 218:13–9. doi: 10.1016/j.vetmic.2018.03.002

PubMed Abstract | Crossref Full Text | Google Scholar

63. Zhang, H, Zhao, G, Guo, Y, Menghwar, H, Chen, Y, Chen, H, et al. Mycoplasma bovis MBOV_RS02825 encodes a secretory nuclease associated with cytotoxicity. Int J Mol Sci. (2016) 17, 1–18. doi: 10.3390/ijms17050628

Crossref Full Text | Google Scholar

64. Brinkmann, V, Reichard, U, Goosmann, C, Fauler, B, Uhlemann, Y, Weiss, DS, et al. Neutrophil extracellular traps kill bacteria. Science. (2004) 303:1532–5. doi: 10.1126/science.1092385

Crossref Full Text | Google Scholar

65. Cacciotto, C, and Alberti, A. Eating the enemy: Mycoplasma strategies to evade neutrophil extracellular traps (NETs) promoting bacterial nucleotides uptake and inflammatory damage. Int J Mol Sci. (2022) 23, 1–18. doi: 10.3390/ijms232315030

PubMed Abstract | Crossref Full Text | Google Scholar

66. Valsala, R, Rana, R, Remesh, AT, Thankappan, S, and Behera, S. Effect of Mycoplasma bovis on production of pro-inflammatory cytokines by peripheral blood mononuclear cells. Adv Anim Vet Sci. (2017) 5:400–4. doi: 10.17582/journal.aavs/2017/5.10.400.404

Crossref Full Text | Google Scholar

67. Howard, CJ, and Taylor, G. Interaction of mycoplasmas and phagocytes. Yale J Biol Med. (1983) 56:643–8.

Google Scholar

68. Burgi, N, Josi, C, Burki, S, Schweizer, M, and Pilo, P. Mycoplasma bovis co-infection with bovine viral diarrhea virus in bovine macrophages. Vet Res. (2018) 49:2. doi: 10.1186/s13567-017-0499-1

PubMed Abstract | Crossref Full Text | Google Scholar

69. Maina, T, Prysliak, T, and Perez-Casal, J. Mycoplasma bovis delay in apoptosis of macrophages is accompanied by increased expression of anti-apoptotic genes, reduced cytochrome C translocation and inhibition of DNA fragmentation. Vet Immunol Immunopathol. (2019) 208:16–24. doi: 10.1016/j.vetimm.2018.12.004

PubMed Abstract | Crossref Full Text | Google Scholar

70. Suleman, M, Cyprian, FS, Jimbo, S, Maina, T, Prysliak, T, Windeyer, C, et al. Mycoplasma bovis-induced inhibition of bovine peripheral blood mononuclear cell proliferation is ameliorated after blocking the immune-inhibitory programmed death 1 receptor. Infect Immun. (2018) 86, 1–15. doi: 10.1128/IAI.00921-17

PubMed Abstract | Crossref Full Text | Google Scholar

71. Gondaira, S, Higuchi, H, Iwano, H, Nakajima, K, Kawai, K, Hashiguchi, S, et al. Cytokine mRNA profiling and the proliferative response of bovine peripheral blood mononuclear cells to Mycoplasma bovis. Vet Immunol Immunopathol. (2015) 165:45–53. doi: 10.1016/j.vetimm.2015.03.002

PubMed Abstract | Crossref Full Text | Google Scholar

72. Gondaira, S, Nishi, K, Tanaka, T, Yamamoto, T, Nebu, T, Watanabe, R, et al. Immunosuppression in cows following Intramammary infusion of Mycoplasma bovis. Infect Immun. (2020) 88, 1–10. doi: 10.1128/IAI.00521-19

PubMed Abstract | Crossref Full Text | Google Scholar

73. Dudek, K, and Bednarek, D. T- and B-cell response analysis following calf immunisation with experimental Mycoplasma Bovis vaccine containing Saponin and lysozyme dimer. J Vet Res. (2017) 61:433–7. doi: 10.1515/jvetres-2017-0060

PubMed Abstract | Crossref Full Text | Google Scholar

74. Dudek, K, Bednarek, D, and Szacawa, E. Evaluation of immune response in seropositive cattle for Mycoplasma bovis. Bull Vet Inst Pulawy. (2011) 55:631–4.

Google Scholar

75. Prysliak, T, Maina, T, Yu, L, Suleman, M, Jimbo, S, and Perez-Casal, J. Induction of a balanced IgG1/IgG2 immune response to an experimental challenge with Mycoplasma bovis antigens following a vaccine composed of Emulsigen™, IDR peptide 1002, and poly I: C. Vaccine. (2017) 35:6604–10. doi: 10.1016/j.vaccine.2017.10.037

Crossref Full Text | Google Scholar

76. Askar, H, Chen, S, Hao, H, Yan, X, Ma, L, Liu, Y, et al. Immune evasion of Mycoplasma bovis. Pathogens. (2021) 10:297. doi: 10.3390/pathogens10030297

PubMed Abstract | Crossref Full Text | Google Scholar

77. Vanden Bush, TJ, and Rosenbusch, RF. Characterization of the immune response to Mycoplasma bovis lung infection. Vet Immunol Immunopathol. (2003) 94:23–33. doi: 10.1016/S0165-2427(03)00056-4

Crossref Full Text | Google Scholar

78. Parker, AM, Sheehy, PA, Hazelton, MS, Bosward, KL, and House, JK. A review of mycoplasma diagnostics in cattle. J Vet Intern Med. (2018) 32:1241–52. doi: 10.1111/jvim.15135

PubMed Abstract | Crossref Full Text | Google Scholar

79. Fox, LK . Mycoplasma mastitis: causes, transmission, and control. Vet Clin North Am Food Anim Pract. (2012) 28:225–37. doi: 10.1016/j.cvfa.2012.03.007

Crossref Full Text | Google Scholar

80. Quinn, PJ, Markey, BK, Leonard, FC, Hartigan, P, Fanning, S, and Fitzpatrick, E. Veterinary microbiology and microbial disease. US: John Wiley & Sons (2011).

Google Scholar

81. Parker, AM, House, JK, Hazelton, MS, Bosward, KL, Mohler, VL, Maunsell, FP, et al. Milk acidification to control the growth of Mycoplasma bovis and Salmonella Dublin in contaminated milk. J Dairy Sci. (2016) 99:9875–84. doi: 10.3168/jds.2016-11537

PubMed Abstract | Crossref Full Text | Google Scholar

82. Nicholas, R, Ayling, R, and McAuliffe, L. Mycoplasma diseases of ruminants. UK: CAB International (2008).

Google Scholar

83. Sachse, K, Salam, HS, Diller, R, Schubert, E, Hoffmann, B, and Hotzel, H. Use of a novel real-time PCR technique to monitor and quantitate Mycoplasma bovis infection in cattle herds with mastitis and respiratory disease. Vet J. (2010) 186:299–303. doi: 10.1016/j.tvjl.2009.10.008

PubMed Abstract | Crossref Full Text | Google Scholar

84. Hazelton, MS, Morton, JM, Parker, AM, Bosward, KL, Sheehy, PA, Dwyer, CJ, et al. Mycoplasma bovis and other Mollicutes in replacement dairy heifers from Mycoplasma bovis-infected and uninfected herds: a 2-year longitudinal study. J Dairy Sci. (2020) 103:11844–56. doi: 10.3168/jds.2020-18921

PubMed Abstract | Crossref Full Text | Google Scholar

85. Hurri, E, Ohlson, A, Lundberg, Å, Aspán, A, Pedersen, K, and Tråvén, M. Herd-level prevalence of Mycoplasma bovis in Swedish dairy herds determined by antibody ELISA and PCR on bulk tank milk and herd characteristics associated with seropositivity. J Dairy Sci. (2022) 105:7764–72. doi: 10.3168/jds.2021-21390

PubMed Abstract | Crossref Full Text | Google Scholar

86. Justice-Allen, A, Trujillo, J, Goodell, G, and Wilson, D. Detection of multiple Mycoplasma species in bulk tank milk samples using real-time PCR and conventional culture and comparison of test sensitivities. J Dairy Sci. (2011) 94:3411–9. doi: 10.3168/jds.2010-3940

PubMed Abstract | Crossref Full Text | Google Scholar

87. Liapi, M, Botsaris, G, Arsenoglou, C, Markantonis, N, Michael, C, Antoniou, A, et al. Rapid detection of Mycoplasma bovis, Staphylococcus aureus and Streptococcus agalactiae in cattle bulk tank milk in Cyprus and relations with somatic cell counts. Pathogens. (2021) 10, 1–9. doi: 10.3390/pathogens10070841

PubMed Abstract | Crossref Full Text | Google Scholar

88. McAloon, CI, McAloon, CG, Tratalos, J, O'Grady, L, McGrath, G, Guelbenzu, M, et al. Seroprevalence of Mycoplasma bovis in bulk milk samples in Irish dairy herds and risk factors associated with herd seropositive status. J Dairy Sci. (2022) 105:5410–9. doi: 10.3168/jds.2021-21334

PubMed Abstract | Crossref Full Text | Google Scholar

89. Punyapornwithaya, V, Fox, LK, Gay, GM, Hancock, DD, and Alldredge, JR. Short communication: the effect of centrifugation and resuspension on the recovery of Mycoplasma species from milk. J Dairy Sci. (2009) 92:4444–7. doi: 10.3168/jds.2009-2182

PubMed Abstract | Crossref Full Text | Google Scholar

90. Boothby, JT, Mueller, R, Jasper, DE, and Thomas, CB. Detecting Mycoplasma bovis in milk by enzyme-linked immunosorbent assay, using monoclonal antibodies. Am J Vet Res. (1986) 47:1082–4.

Google Scholar

91. Biddle, MK, Fox, LK, Hancock, DD, Gaskins, CT, and Evans, MA. Effects of storage time and thawing methods on the recovery of Mycoplasma species in milk samples from cows with intramammary infections. J Dairy Sci. (2004) 87:933–6. doi: 10.3168/jds.S0022-0302(04)73237-3

PubMed Abstract | Crossref Full Text | Google Scholar

92. Dudek, K, Bednarek, D, Ayling, RD, and Szacawa, E. Immunomodulatory effect of Mycoplasma bovis in experimentally infected calves. Bull Vet Inst Pulawy. (2013) 57:499–506. doi: 10.2478/bvip-2013-0087

Crossref Full Text | Google Scholar

93. Liu, Y, Xu, S, Li, M, Zhou, M, Huo, W, Gao, J, et al. Molecular characteristics and antibiotic susceptibility profiles of Mycoplasma bovis associated with mastitis on dairy farms in China. Prev Vet Med. (2020) 182:105106. doi: 10.1016/j.prevetmed.2020.105106

Crossref Full Text | Google Scholar

94. Hotzel, H, Demuth, B, Sachse, K, Pflitsch, A, and Pfützner, H. Detection of Mycoplasma bovis using in vitro deoxyribonucleic acid amplification. Rev Sci Tech. (1993) 12:581–91. doi: 10.20506/rst.12.2.700

PubMed Abstract | Crossref Full Text | Google Scholar

95. Cremonesi, P, Vimercati, C, Pisoni, G, Perez, G, Ribera, AM, Castiglioni, B, et al. Development of DNA extraction and PCR amplification protocols for detection of Mycoplasma bovis directly from milk samples. Vet Res Commun. (2007) 31:225–7. doi: 10.1007/s11259-007-0011-x

PubMed Abstract | Crossref Full Text | Google Scholar

96. Sachse, K, Pfützner, H, Hotzel, H, Demuth, B, Heller, M, and Berthold, E. Comparison of various diagnostic methods for the detection of Mycoplasma bovis. Rev Sci Tech. (1993) 12:571–80. doi: 10.20506/rst.12.2.701

PubMed Abstract | Crossref Full Text | Google Scholar

97. Andrés-Lasheras, S, Zaheer, R, Ha, R, Lee, C, Jelinski, M, and McAllister, TA. A direct qPCR screening approach to improve the efficiency of Mycoplasma bovis isolation in the frame of a broad surveillance study. J Microbiol Methods. (2020) 169:105805. doi: 10.1016/j.mimet.2019.105805

PubMed Abstract | Crossref Full Text | Google Scholar

98. Behera, S, Rana, R, Gupta, PK, Kumar, D, Sonal, RV, et al. Development of real-time PCR assay for the detection of Mycoplasma bovis. Tropl Anim Health Prod. (2018) 50:875–82. doi: 10.1007/s11250-018-1510-1

PubMed Abstract | Crossref Full Text | Google Scholar

99. Arcangioli, MA, Chazel, M, Sellal, E, Botrel, MA, Bezille, P, Poumarat, F, et al. Prevalence of Mycoplasma bovis udder infection in dairy cattle: preliminary field investigation in Southeast France. N Z Vet J. (2011) 59:75–8. doi: 10.1080/00480169.2011.552856

PubMed Abstract | Crossref Full Text | Google Scholar

100. Subramaniam, S, Bergonier, D, Poumarat, F, Capaul, S, Schlatter, Y, Nicolet, J, et al. Species identification of Mycoplasma bovis and Mycoplasma agalactiae based on the uvr C genes by PCR. Mol Cell Probes. (1998) 12:161–9. doi: 10.1006/mcpr.1998.0160

PubMed Abstract | Crossref Full Text | Google Scholar

101. Thomas, A, Dizier, I, Linden, A, Mainil, J, Frey, J, and Vilei, EM. Conservation of the uvr C gene sequence in Mycoplasma bovis and its use in routine PCR diagnosis. Vet J. (2004) 168:100–2. doi: 10.1016/S1090-0233(03)00186-2

PubMed Abstract | Crossref Full Text | Google Scholar

102. Parker, AM, House, JK, Hazelton, MS, Bosward, KL, and Sheehy, PA. Comparison of culture and a multiplex probe PCR for identifying Mycoplasma species in bovine milk, semen and swab samples. PloS One. (2017) 12:e0173422. doi: 10.1371/journal.pone.0173422

PubMed Abstract | Crossref Full Text | Google Scholar

103. Valeris-Chacin, R, Powledge, S, McAtee, T, Morley, PS, and Richeson, J. Mycoplasma bovis is associated with Mannheimia haemolytica during acute bovine respiratory disease in feedlot cattle. Front Microbiol. (2022) 13:946792. doi: 10.3389/fmicb.2022.946792

PubMed Abstract | Crossref Full Text | Google Scholar

104. Pansri, P, Katholm, J, Krogh, KM, Aagaard, AK, Schmidt, LMB, Kudirkiene, E, et al. Evaluation of novel multiplex qPCR assays for diagnosis of pathogens associated with the bovine respiratory disease complex. Vet J. (2020) 256:105425. doi: 10.1016/j.tvjl.2020.105425

PubMed Abstract | Crossref Full Text | Google Scholar

105. Conrad, CC, Daher, RK, Stanford, K, Amoako, KK, Boissinot, M, Bergeron, MG, et al. A sensitive and accurate recombinase polymerase amplification assay for detection of the primary bacterial pathogens causing bovine respiratory disease. Front Vet Sci. (2020) 7:208. doi: 10.3389/fvets.2020.00208

PubMed Abstract | Crossref Full Text | Google Scholar

106. Justice-Allen, A, Trujillo, J, Corbett, R, Harding, R, Goodell, G, and Wilson, D. Survival and replication of Mycoplasma species in recycled bedding sand and association with mastitis on dairy farms in Utah. J Dairy Sci. (2010) 93:192–202. doi: 10.3168/jds.2009-2474

PubMed Abstract | Crossref Full Text | Google Scholar

107. Baird, S, Carman, J, and Dinsmore, R. Detection and identification of Mycoplasma from bovine mastitis infections using a nested polymerase chain reaction. J Vet Diagn Invest. (1999) 11:432–5. doi: 10.1177/104063879901100507

Crossref Full Text | Google Scholar

108. Amram, E, Mikula, I, Schnee, C, Ayling, RD, Nicholas, RA, Rosales, RS, et al. 16S rRNA gene mutations associated with decreased susceptibility to tetracycline in Mycoplasma bovis. Antimicrob Agents Chemother. (2015) 59:796–802. doi: 10.1128/AAC.03876-14

PubMed Abstract | Crossref Full Text | Google Scholar

109. Lysnyansky, I, Mikula, I, Gerchman, I, and Levisohn, S. Rapid detection of a point mutation in the par C gene associated with decreased susceptibility to fluoroquinolones in Mycoplasma bovis. Antimicrob Agents Chemother. (2009) 53:4911–4. doi: 10.1128/AAC.00703-09

PubMed Abstract | Crossref Full Text | Google Scholar

110. Parker, AM, House, JK, Hazelton, MS, Bosward, KL, Morton, JM, and Sheehy, PA. Bulk tank milk antibody ELISA as a biosecurity tool for detecting dairy herds with past exposure to Mycoplasma bovis. J Dairy Sci. (2017) 100:8296–309. doi: 10.3168/jds.2016-12468

PubMed Abstract | Crossref Full Text | Google Scholar

111. Petersen, MB, Wawegama, NK, Denwood, M, Markham, PF, Browning, GF, and Nielsen, LR. Mycoplasma bovis antibody dynamics in naturally exposed dairy calves according to two diagnostic tests. BMC Vet Res. (2018) 14:258. doi: 10.1186/s12917-018-1574-1

PubMed Abstract | Crossref Full Text | Google Scholar

112. Salgadu, A, Firestone, SM, Watt, A, Thilakarathne, DS, Condello, AK, Siu, D, et al. Evaluation of the mil a ELISA for the diagnosis of herd infection with Mycoplasma bovis using bulk tank milk and estimation of the prevalence of M. bovis in Australia. Vet Microbiol. (2022) 270:109454. doi: 10.1016/j.vetmic.2022.109454

PubMed Abstract | Crossref Full Text | Google Scholar

113. Wawegama, NK, Browning, GF, Kanci, A, Marenda, MS, and Markham, PF. Development of a recombinant protein-based enzyme-linked immunosorbent assay for diagnosis of Mycoplasma bovis infection in cattle. Clin Vaccine Immunol. (2014) 21:196–202. doi: 10.1128/CVI.00670-13

PubMed Abstract | Crossref Full Text | Google Scholar

114. Farzaneh, M, Derakhshandeh, A, Al-Farha, AAA, Petrovski, K, and Hemmatzadeh, F. A novel phage-displayed mil a ELISA for detection of antibodies against M. bovis in bovine milk. J Appl Microbiol. (2022) 133:1496–505. doi: 10.1111/jam.15655

PubMed Abstract | Crossref Full Text | Google Scholar

115. Petersen, MB, Krogh, K, and Nielsen, LR. Factors associated with variation in bulk tank milk Mycoplasma bovis antibody-ELISA results in dairy herds. J Dairy Sci. (2016) 99:3815–23. doi: 10.3168/jds.2015-10056

PubMed Abstract | Crossref Full Text | Google Scholar

116. Gogoi-Tiwari, J, Tiwari, HK, Wawegama, NK, Premachandra, C, Robertson, ID, Fisher, AD, et al. Prevalence of Mycoplasma bovis infection in calves and dairy cows in Western Australia. Vet Sci. (2022) 9, 1–11. doi: 10.3390/vetsci9070351

Crossref Full Text | Google Scholar

117. Cowled, BD, Sergeant, ESG, Leslie, EEC, Crosbie, A, Burroughs, A, Kingston, O, et al. Use of scenario tree modelling to plan freedom from infection surveillance: Mycoplasma bovis in New Zealand. Prev Vet Med. (2022) 198:105523. doi: 10.1016/j.prevetmed.2021.105523

PubMed Abstract | Crossref Full Text | Google Scholar

118. Chen, X, Huang, J, Zhu, H, Guo, Y, Khan, FA, Menghwar, H, et al. P 27 (MBOV_RS03440) is a novel fibronectin binding adhesin of Mycoplasma bovis. Int J Med Microbiol. (2018) 308:848–57. doi: 10.1016/j.ijmm.2018.07.006

PubMed Abstract | Crossref Full Text | Google Scholar

119. Sun, Z, Fu, P, Wei, K, Zhang, H, Zhang, Y, Xu, J, et al. Identification of novel immunogenic proteins from Mycoplasma bovis and establishment of an indirect ELISA based on recombinant E1 beta subunit of the pyruvate dehydrogenase complex. PloS One. (2014) 9:e88328. doi: 10.1371/journal.pone.0088328

PubMed Abstract | Crossref Full Text | Google Scholar

120. Fu, P, Sun, Z, Zhang, Y, Yu, Z, Zhang, H, Su, D, et al. Development of a direct competitive ELISA for the detection of Mycoplasma bovis infection based on a monoclonal antibody of P 48 protein. BMC Vet Res. (2014) 10:42. doi: 10.1186/1746-6148-10-42

PubMed Abstract | Crossref Full Text | Google Scholar

121. Holland, RD, Wilkes, JG, Rafii, F, Sutherland, JB, Persons, CC, Voorhees, KJ, et al. Rapid identification of intact whole bacteria based on spectral patterns using matrix-assisted laser desorption/ionization with time-of-flight mass spectrometry. Rapid Commun Mass Spectrom. (1996) 10:1227–32. doi: 10.1002/(SICI)1097-0231(19960731)10:10<1227::AID-RCM659>3.0.CO;2-6

PubMed Abstract | Crossref Full Text | Google Scholar

122. Randall, LP, Lemma, F, Koylass, M, Rogers, J, Ayling, RD, Worth, D, et al. Evaluation of MALDI-ToF as a method for the identification of bacteria in the veterinary diagnostic laboratory. Res Vet Sci. (2015) 101:42–9. doi: 10.1016/j.rvsc.2015.05.018

Crossref Full Text | Google Scholar

123. Pereyre, S, Tardy, F, Renaudin, H, Cauvin, E, del Prá Netto Machado, L, Tricot, A, et al. Identification and subtyping of clinically relevant human and ruminant mycoplasmas by use of matrix-assisted laser desorption ionization-time of flight mass spectrometry. J Clin Microbiol. (2013) 51:3314–23. doi: 10.1128/JCM.01573-13

PubMed Abstract | Crossref Full Text | Google Scholar

124. Bokma, J, Van Driessche, L, Deprez, P, Haesebrouck, F, Vahl, M, Weesendorp, E, et al. Rapid identification of Mycoplasma bovis strains from bovine Bronchoalveolar lavage fluid with matrix-assisted laser desorption ionization-time of flight mass spectrometry after enrichment procedure. J Clin Microbiol. (2020) 58, 1–10. doi: 10.1128/JCM.00004-20

PubMed Abstract | Crossref Full Text | Google Scholar

125. McDaniel, AJ, and Derscheid, RJ. MALDI-TOF mass spectrometry and high-resolution melting PCR for the identification of Mycoplasma bovis isolates. BMC Vet Res. (2021) 17:170. doi: 10.1186/s12917-021-02870-5

PubMed Abstract | Crossref Full Text | Google Scholar

126. Zubair, M, Muhamed, SA, Khan, FA, Zhao, G, Menghwar, H, Faisal, M, et al. Identification of 60 secreted proteins for Mycoplasma bovis with secretome assay. Microb Pathog. (2020) 143:104135. doi: 10.1016/j.micpath.2020.104135

PubMed Abstract | Crossref Full Text | Google Scholar

127. Singhal, N, Kumar, M, Kanaujia, PK, and Virdi, JS. MALDI-TOF mass spectrometry: an emerging technology for microbial identification and diagnosis. Front Microbiol. (2015) 6, 1–16. doi: 10.3389/fmicb.2015.00791

PubMed Abstract | Crossref Full Text | Google Scholar

128. Soroka, M, Wasowicz, B, and Rymaszewska, A. Loop-mediated isothermal amplification (LAMP): the better sibling of PCR? Cells. (2021) 10, 1–20. doi: 10.3390/cells10081931

PubMed Abstract | Crossref Full Text | Google Scholar

129. Bai, Z, Shi, L, Hu, C, Chen, X, Qi, J, Ba, X, et al. Development of a loop-mediated isothermal amplification assay for sensitive and rapid detection of Mycoplasma bovis. Afr J Biotechnol. (2011) 10:12333–8.

Google Scholar

130. Higa, Y, Uemura, R, Yamazaki, W, Goto, S, Goto, Y, and Sueyoshi, M. An improved loop-mediated isothermal amplification assay for the detection of Mycoplasma bovis. J Vet Med Sci. (2016) 78:1343–6. doi: 10.1292/jvms.15-0459

PubMed Abstract | Crossref Full Text | Google Scholar

131. Ashraf, A, Imran, M, Yaqub, T, Tayyab, M, Shehzad, W, Mingala, CN, et al. Development and validation of a loop-mediated isothermal amplification assay for the detection of Mycoplasma bovis in mastitic milk. Folia Microbiol (Praha). (2018) 63:373–80. doi: 10.1007/s12223-017-0576-x

PubMed Abstract | Crossref Full Text | Google Scholar

132. Appelt, S, Aly, SS, Tonooka, K, Glenn, K, Xue, Z, Lehenbauer, TW, et al. Development and comparison of loop-mediated isothermal amplification and quantitative polymerase chain reaction assays for the detection of Mycoplasma bovis in milk. J Dairy Sci. (2019) 102:1985–96. doi: 10.3168/jds.2018-15306

PubMed Abstract | Crossref Full Text | Google Scholar

133. Haapala, V, Vähänikkilä, N, Kulkas, L, Tuunainen, E, Pohjanvirta, T, Autio, T, et al. Mycoplasma bovis infection in dairy herds-risk factors and effect of control measures. J Dairy Sci. (2021) 104:2254–65. doi: 10.3168/jds.2020-18814

PubMed Abstract | Crossref Full Text | Google Scholar

134. Autio, T, Tuunainen, E, Nauholz, H, Pirkkalainen, H, London, L, and Pelkonen, S. Overview of control programs for cattle diseases in Finland. Front Vet Sci. (2021) 8:688936. doi: 10.3389/fvets.2021.688936

PubMed Abstract | Crossref Full Text | Google Scholar

135. Klein, U, de Jong, A, Youala, M, El Garch, F, Stevenin, C, Moyaert, H, et al. New antimicrobial susceptibility data from monitoring of Mycoplasma bovis isolated in Europe. Vet Microbiol. (2019) 238:108432. doi: 10.1016/j.vetmic.2019.108432

PubMed Abstract | Crossref Full Text | Google Scholar

136. Lerner, U, Amram, E, Ayling, RD, Mikula, I, Gerchman, I, Harrus, S, et al. Acquired resistance to the 16-membered macrolides tylosin and tilmicosin by Mycoplasma bovis. Vet Microbiol. (2014) 168:365–71. doi: 10.1016/j.vetmic.2013.11.033

PubMed Abstract | Crossref Full Text | Google Scholar

137. Soehnlen, MK, Tran, MA, Lysczek, HR, Wolfgang, DR, and Jayarao, BM. Identification of novel small molecule antimicrobials targeting Mycoplasma bovis. J Antimicrob Chemother. (2011) 66:574–7. doi: 10.1093/jac/dkq503

PubMed Abstract | Crossref Full Text | Google Scholar

138. Dudek, K, Bednarek, D, Ayling, RD, Kycko, A, Szacawa, E, and Karpinska, TA. An experimental vaccine composed of two adjuvants gives protection against Mycoplasma bovis in calves. Vaccine. (2016) 34:3051–8. doi: 10.1016/j.vaccine.2016.04.087

PubMed Abstract | Crossref Full Text | Google Scholar

139. Boothby, JT, Jasper, DE, and Thomas, CB. Experimental intramammary inoculation with Mycoplasma bovis in vaccinated and unvaccinated cows: effect on milk production and milk quality. Can J Vet Res. (1986) 50:200–4.

Google Scholar

140. Soehnlen, MK, Aydin, A, Lengerich, EJ, Houser, BA, Fenton, GD, Lysczek, HR, et al. Blinded, controlled field trial of two commercially available Mycoplasma bovis bacterin vaccines in veal calves. Vaccine. (2011) 29:5347–54. doi: 10.1016/j.vaccine.2011.05.092

Crossref Full Text | Google Scholar

141. Arcangioli, MA, Duet, A, Meyer, G, Dernburg, A, Bézille, P, Poumarat, F, et al. The role of Mycoplasma bovis in bovine respiratory disease outbreaks in veal calf feedlots. Vet J. (2008) 177:89–93. doi: 10.1016/j.tvjl.2007.03.008

PubMed Abstract | Crossref Full Text | Google Scholar

142. Prysliak, T, van der Merwe, J, and Perez-Casal, J. Vaccination with recombinant Mycoplasma bovis GAPDH results in a strong humoral immune response but does not protect feedlot cattle from an experimental challenge with M. bovis. Microb Pathog. (2013) 55:1–8. doi: 10.1016/j.micpath.2012.12.001

PubMed Abstract | Crossref Full Text | Google Scholar

143. Mulongo, M, Prysliak, T, and Perez-Casal, J. Vaccination of feedlot cattle with extracts and membrane fractions from two Mycoplasma bovis isolates results in strong humoral immune responses but does not protect against an experimental challenge. Vaccine. (2013) 31:1406–12. doi: 10.1016/j.vaccine.2012.12.055

PubMed Abstract | Crossref Full Text | Google Scholar

144. Li, Y, Zheng, H, Liu, Y, Jiang, Y, Xin, J, Chen, W, et al. The complete genome sequence of Mycoplasma bovis strain Hubei-1. PloS One. (2011) 6:e20999. doi: 10.1371/journal.pone.0020999

PubMed Abstract | Crossref Full Text | Google Scholar

145. Boonyayatra, S, Fox, LK, Besser, TE, Sawant, A, and Gay, JM. Effects of storage methods on the recovery of Mycoplasma species from milk samples. Vet Microbiol. (2010) 144:210–3. doi: 10.1016/j.vetmic.2009.12.014

PubMed Abstract | Crossref Full Text | Google Scholar

146. Penterman, PM, Holzhauer, M, van Engelen, E, Smits, D, and Velthuis, AGJ. Dynamics of Mycoplasma bovis in Dutch dairy herds during acute clinical outbreaks. Vet J. (2022) 283-284:105841. doi: 10.1016/j.tvjl.2022.105841

PubMed Abstract | Crossref Full Text | Google Scholar

147. Wilson, DJ, Skirpstunas, RT, Trujillo, JD, Cavender, KB, Bagley, CV, and Harding, RL. Unusual history and initial clinical signs of Mycoplasma bovis mastitis and arthritis in first-lactation cows in a closed commercial dairy herd. J Am Vet Med Assoc. (2007) 230:1519–23. doi: 10.2460/javma.230.10.1519

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: Mycoplasma bovis , Mycoplasma bovis mastitis, dairy cows, subclinical mastitis, virulence factors, pathogenesis, clinical mastitis, intramammary infection

Citation: Gelgie AE, Desai SE, Gelalcha BD and Kerro Dego O (2024) Mycoplasma bovis mastitis in dairy cattle. Front. Vet. Sci. 11:1322267. doi: 10.3389/fvets.2024.1322267

Received: 16 October 2023; Accepted: 21 February 2024;
Published: 06 March 2024.

Edited by:

Camila Hamond, Animal and Plant Health Inspection Service (USDA), United States

Reviewed by:

Hidetoshi Higuchi, Rakuno Gakuen University, Japan
Tarja Pohjanvirta, Finnish Food Authority, Finland

Copyright © 2024 Gelgie, Desai, Gelalcha and Kerro Dego. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Oudessa Kerro Dego, okerrode@utk.edu

ORCID: Sarah E. Desai, orcid.org/0009-0000-9631-6584

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.