3,747
Views
28
CrossRef citations to date
0
Altmetric
Review article

Clinical expression, epidemiology, and monitoring of Mycoplasma gallisepticum and Mycoplasma synoviae: an update

, ORCID Icon &
Pages 2-18 | Received 28 Feb 2021, Accepted 13 Jun 2021, Published online: 21 Sep 2021

ABSTRACT

Mycoplasma gallisepticum (MG) and Mycoplasma synoviae (MS) are of clinical and economic importance for the global poultry industry. Many countries and integrations are involved in monitoring programmes to control both mycoplasma species. This review provides an extensive historic overview of the last seven decades on the development of the knowledge regarding the factors that influence the clinical expression of the disease, the epidemiology, and monitoring of both MG and MS. This includes the detection of new virulent strains, studies unravelling the transmission routes, survival characteristics, and the role of other avian hosts. Also the role of molecular typing tests in unravelling epidemiology and factors that complicate the interpretation of test results is discussed. The latter includes the presence of heterologous mycoplasma infections, the use of heterologous oil-emulsion vaccines, and the use of antibiotic treatments. Also the occurrence of MG and MS strains with low virulence and the use of live and/or inactivated MS and MS vaccines are discussed.

This article is part of the following collections:
Golden Anniversary Reviews in Avian Pathology

Introduction

The clinical and economic importance of Mycoplasma gallisepticum (MG) in the poultry industry was already acknowledged in the early 1950s (Floyd et al., Citation1952; Crawley & Fahey, Citation1955). For Mycoplasma synoviae (MS), the clinical impact for the poultry industry was also recognized in early reports (Olson, Adler et al., Citation1964). Since then, an increase of economic impact has been recognized due to the identification and emergence of new pathogenic strains (Kerr & Olson, Citation1969; Morrow, Bell et al., Citation1990; Landman & Feberwee, Citation2001, Citation2012; Landman & Bronneberg, Citation2003). The first description of avian mycoplasmas dates from 1936 (Nelson, Citation1936) and was related to chickens with respiratory disease from which coccobaciliform bodies were isolated (Nelson, Citation1935); they were later renamed pleuropneumonia-like organisms (Barber & Fabricant, Citation1962). This respiratory disease was later defined as chronic respiratory disease (CRD) in chickens and infectious sinusitis in turkeys (Floyd et al., Citation1952), and the aetiological agent identified as MG (Edward & Kanarek, Citation1960). Infectious synovitis was reported for the first time in the 1950s (Olson et al., Citation1954; Chalquest & Fabricant, Citation1960) and the aetiological agent was later identified as MS (Olson, Adler et al., Citation1964; Olsen, Kerr et al., Citation1964). Most important was the evidence that both mycoplasma species could exhibit a variance in clinical expression (Ghazikhanian et al., Citation1973; Lin & Kleven, Citation1982a), especially in the presence of other respiratory agents, and that synergistic effects with other respiratory agents had negative effects on poultry performance (Fahey & Crawley, Citation1954b; Gross, Citation1961; Corstvet & Sadler, Citation1966; Springer et al., Citation1974; Rhoades, Citation1977). Studies unravelling their transmission routes and epidemiology contributed to the development and implementation of the first monitoring and control programmes for MG (Crawley & Fahey, Citation1955; Chute et al., Citation1965). The knowledge regarding the expression of clinical disease, epidemiology, and the implementation of monitoring programmes was crucial for the control of both species in the commercial poultry industry in the last six decades, and contributed to the economic sustainability of the poultry industry of today. This paper gives an overview on the historical and current knowledge regarding clinical expression of the disease, the epidemiology, and monitoring of MG and MS.

Clinical expression

Clinical expression of MG

MG is associated with respiratory disease, egg production losses, reduced hatchability, and significant downgrading of carcasses at slaughter due to the systemic effects (Stipkovits & Kempf, Citation1996; Kleven, Citation1997; Levisohn & Kleven, Citation2000). Besides commercial poultry, clinical outbreaks in backyard poultry and other avian species including wild birds have also been reported (Madden et al., Citation1967; Cookson & Shivaprasad, Citation1994; Ley et al., Citation1996; Benčina et al., Citation2003). In the early 1950s it was shown that MG was responsible for turkey sinusitis, CRD in chickens, egg production losses up to 50% during 7–14 days not recovering up to the expected production level, reduced eggshell quality, and reduced hatchability due to late embryo mortality in chickens (Markham & Wong, Citation1952; Fahey & Crawley, Citation1954b). Other observations confirmed that MG alone was able to induce disease and that the expression of clinical signs caused by MG varied greatly between infections with different isolates (Gross, Citation1961; Truscott et al., Citation1974; Jordan, Citation1975; Rodriguez & Kleven, Citation1980; Yoder, Citation1986). Also, strains with differences in tissue tropism and/or virulence are described such as the MG R strain, MG S6 strain, and WVU 907 strain. The MG R strain is a strain isolated from turkeys with sinusitis (Markham & Wong, Citation1952), while the MG S6 strain was isolated from the brains of turkeys with infectious sinusitis and neurological signs (Zander, Citation1961). More evidence of the involvement of MG in neurological signs in turkeys was reported over the years (Thomas et al., Citation1966; Chin et al., Citation1991; Wyrzykowski et al., Citation2013). Furthermore, MG is also reported in relation to joint disease (Wannop et al., Citation1971; Morrow et al., Citation1997). In 1974 for the first time, a MG strain of low virulence was reported (Truscott et al., Citation1974). This strain induced a low number of reactors in serological tests, while no clinical signs were observed (Truscott et al., Citation1974; Yoder, Citation1986; Dingfelder et al., Citation1991). In 1981, the Mycoplasma strain WVU 907 was designated as a MG strain of low virulence (Sahu & Olson, Citation1981; Yoder, Citation1986; Kempf et al., Citation1997).

Clinical expression of MS

The clinical and economic relevance of MS for the commercial poultry industry was evidenced in several early studies. MS monitoring and control programmes have already been running in reproduction stock for a long time and include culling of MS-positive flocks and/or streaming of infected products. However, in the last two decades, the identification of new virulent MS strains, with economic impact on poultry production, increased the attention for the control of MS in the commercial poultry industry in general (Landman, Citation2014). Different from MG, until now only subclinical infections have been reported in backyard poultry and other wild free-flying birds such as pigeons, crows, and sparrows (Benčina et al., Citation1988a, Citation1988b; Bradbury et al., Citation2001b; Michiels et al., Citation2016). MS is known as the cause of infectious synovitis, respiratory disease, egg production losses, and eggshell abnormalities in commercial poultry. The first report involving MS was related to infectious synovitis (Olson, Kerr et al., Citation1964). Since this finding, MS infection has been widely reported as the causative agent of infectious synovitis in turkeys and chickens (Kerr & Olson, Citation1969; Morrow, Bell et al., Citation1990; Landman & Feberwee, Citation2001, Citation2012; Landman & Bronneberg, Citation2003). Also, the involvement of MS in respiratory disease has been shown in several studies. MS was recovered from the respiratory tract of birds without clinical disease (Olson, Adler et al., Citation1964), while other studies showed that MS infections were related to egg production losses, airsacculitis in broilers and turkeys, and reduced broiler performance (King et al., Citation1973; Vardaman, Reece et al., Citation1973; Bradbury & Howell, Citation1975; Goren, Citation1978; Lott et al., Citation1978; Macowan et al., Citation1982; Osorio et al., Citation2007). Moreover, tissue tropism seemed relevant as studies showed that some MS isolates were more related to the presence of joint lesions, while other isolates were more related to the presence of lesions in the respiratory tract (Kleven et al., Citation1975). In 2009, a MS isolate related to egg production losses, eggshell apex abnormalities (EAA), and reduced eggshell strength was reported for the first time (Feberwee et al., Citation2009). Broiler-breeder birds seemed less susceptible for the production of eggs with EAA than commercial layers (Feberwee & Landman, Citation2010). From 2009 onwards, several reports on MS related to EAA, egg production losses and/or reduced eggshell quality, and increased second-quality eggs followed and showed the worldwide presence of such strains (Catania et al., Citation2010; Ranck et al., Citation2010; Gole et al., Citation2012; Jeon et al., Citation2014; Kursa et al., Citation2019; Lorenc et al., Citation2019; Cisneros-Tamayo et al., Citation2020). Moreover, other reports also evidenced the amyloid-inducing potential of MS (Landman & Feberwee, Citation2001).

Factors influencing clinical expression MG and MS

The role of other respiratory pathogens, including viral vaccine strains, in aggravating disease or the expression of CRD in the presence of inapparent MS and MG infections, was acknowledged as early as 1955 (Markham & Wong, Citation1952; Crawley & Fahey, Citation1955; Gross, Citation1961). During the last four decades the significant role of several respiratory agents complicating disease induced by MG has been confirmed. The synergistic effect of MG with other bacteria, such as Haemophilus gallinarum (Avibacterium paragallinarum), Mycoplasma meleagridis, Pasteurella gallinarum, and Escherichia coli, was evidenced (Gross, Citation1958; Kato, Citation1965; Reis & Yamamoto, Citation1971; Chu & Uppal, Citation1975; Bradbury, Citation1984; Droual et al., Citation1992). Other studies evidenced the role of respiratory viruses including vaccine strains such as Newcastle disease virus (NDV), infectious bronchitis virus (IBV), and avian metapneumovirus in the aggravation of the disease in the presence of MG (Corstvet & Sadler, Citation1966; Sato et al., Citation1970; Nonomura & Sato, Citation1971; Omuro et al., Citation1971; Rodriguez & Kleven, Citation1980; Naylor et al., Citation1992; Leigh et al., Citation2012). The most recent studies show the synergistic effect between a H3N8 low pathogenic avian influenza virus and MG (Stipkovits, Egyed et al., Citation2012; Stipkovits, Glavits et al., Citation2012). Respiratory bacteria (such as M. meleagridis, Pasteurella gallinarum, E. coli, O. rhinotracheale), and respiratory viruses, including vaccine strains, have also been shown to complicate disease induced by MS (Kleven et al., Citation1972; Springer et al., Citation1974; Bradbury & Howell, Citation1975; Kleven, Citation1975; Rhoades, Citation1977; Goren, Citation1978; Hopkins & Yoder, Citation1984; Droual et al., Citation1992; Khehra et al., Citation1999; Zorman-Rojs et al., Citation2000; Landman & Feberwee, Citation2004; Raviv et al., Citation2007; Feberwee et al., Citation2009; Feberwee & Landman, Citation2010; Roussan et al., Citation2011). In a recent paper, a frequent association of detection of M. pullorum and MS was found in cases with eggshell apex abnormalities (Cisneros-Tamayo et al., Citation2020). However, further studies have to confirm the synergistic role of M. pullorum. The degree of synergism between MS and IBV strains seems to be influenced by the virulence of the IBV isolate (Hopkins & Yoder, Citation1982; Landman & Feberwee, Citation2004). Besides synergism with other respiratory agents, for both MG and MS, it was also shown that disease severity may be enhanced due to immunosuppression (Vardaman, Landreth et al., Citation1973; Giambrone et al., Citation1977; Morrison et al., Citation1977) and/or adverse climate and house conditions (Sato et al., Citation1973; Yoder et al., Citation1977; Kempf et al., Citation1988).

Epidemiology

Knowledge about the epidemiology of MG and MS is vital for the design of monitoring and control programmes of both species. It includes knowledge about the transmission routes, survival of MG and MS in the environment, and the occurrence of MG and MS in avian species other than chickens and turkeys. Also, the development of molecular typing methods has contributed to the knowlegde regarding the epidemiology of MG and MS.

Transmission routes

The economic relevance of MG related to respiratory disease, egg production losses, and reduced hatchabililty was already acknowledged in the 1950s (Floyd et al., Citation1952; Crawley & Fahey, Citation1955). Subsequently, control of this species became relevant and knowledge with regard to the transmission routes of avian mycoplasmas also became more important. In early studies it was determined that MG and MS infection led to a carrier state and that both species can be transmitted both horizontally and vertically (in ovo), and control of CRD should be based on the control of MG in the reproduction flocks (Fahey & Crawley, Citation1954b, Citation1956; Carnaghan, Citation1961; McMartin, Citation1968; Vardaman, Citation1976). Horizontal spread can occur via direct contact and via indirect contact by aerosol transmission or by the introduction of contaminated materials or persons (Fahey & Crawley, Citation1955a). Furthermore, the study of McMartin and co-workers evidenced the effect of poultry density on the rate of horizontal transmission within a flock (McMartin et al., Citation1987). Vertical transmission is a major route of spread of the infection, and is of economic relevance regarding international trade. The egg transmission rates are unpredictable and may vary between strains (Olesiuk & Van Roekel, Citation1960; Carnaghan, Citation1961; Roberts & McDaniel, Citation1967; Lin & Kleven, Citation1982b) and phase of the infection (Olesiuk & Van Roekel, Citation1960; Carnaghan, Citation1961; Roberts & McDaniel, Citation1967; Lin & Kleven, Citation1982b). The peak egg transmission rates occur about 3–4 weeks after infection and are lower in the chronic phase of the infection (Roberts & McDaniel, Citation1967). Although the percentage of vertically-infected progeny is low in the chronic phase, horizontal spreading to non-infected chickens still can occur (Fahey & Crawley, Citation1954b). In some parts of the world, the prevalence of MG has decreased considerably due to the top-down control (Ter Veen et al., Citation2021).

Survival outside the host

The understanding of reservoirs and survival of both MG and MS outside the host has improved; however, their role in the epidemiology is not yet fully elucidated (Kleven, Citation1997, Citation2008; Levisohn & Kleven, Citation2000; Landman, Citation2014). Mechanisms of spread between flocks are not yet fully understood, as unexplained outbreaks still occur and may involve direct and indirect transmission by contact, aerosol, and fomites (Fahey & Crawley, Citation1956; Olsen, Kerr et al., Citation1964; Anderson & Beard, Citation1967; Mohammed et al., Citation1987; Marois et al., Citation2000, Citation2005). Also, artificial insemination has been regarded as a route of transmission for MG (Buntz et al., Citation1986). Multi-age housing and poor hygiene management can be regarded as risk factors for the introduction and transmission of MG and MS. Studies have shown that both mycoplasma species can survive on different materials such as straw, rubber, nose, hair, dust, feed, water, and egg debris, and these materials can be regarded as sources of indirect transmission for both species. However, survival time is low on most materials (1–4 days) except for egg material in which both species can survive for several months (Chandiramani et al., Citation1966; Christensen et al., Citation1994; Abolnik & Gouws, Citation2014). Based on the latter, it can be concluded that materials contaminated with egg debris are a major risk factor in the horizontal transmission of MG and MS. Survival potential outside the host is also evidenced by the identification of biofilm formation by MG. Biofilm formation suggests a survival advantage and increased resistance to disinfectants (Chen et al., Citation2012; Wang et al., Citation2017), and is subsequently a risk factor for transmission. A recent study showed that biofilm formation may also increase the survival potency of a pathogen inside the host and, in that way, may also increase the potency for vertical transmission (Chen et al., Citation2021).

Hosts

Many studies evidenced the presence of MG and MS infections in birds other than chickens and turkeys, including different types of backyard poultry (such as pheasants, partridges, and quails), wild birds (such as sparrows, house and gold finches, pigeons, and flamingos), and waterfowl, such as ducks and geese (Madden et al., Citation1967; Tripathy et al., Citation1972; Pascucci et al., Citation1976; Yamada & Matsuo, Citation1983; Buntz et al., Citation1986; Poveda et al., Citation1986; Ley et al., Citation1996; Bradbury et al., Citation2001b; Catania et al., Citation2016; Michiels et al., Citation2016). A recent review reported the presence of MG in 56 non-poultry hosts (Sawicka et al., Citation2020). Experimentally it was also shown that transmission of MG from wildlife to chickens (Stallknecht et al., Citation1998) could occur only after direct contact (Kleven & Fletcher, Citation1983; Stallknecht et al., Citation1998). This latter finding stresses the importance of keeping commercial chicken houses free from wild birds. This also raises the question whether the increasing tendency to keep commercial chickens outdoor increases the risk of introduction of MG and/or MS via contact with wild birds and waterfowl.

Epidemiological conclusions based on molecular tests

In the past decades several molecular typing techniques have been used to unravel the epidemiology of MG and MS. These tests have helped to differentiate between vaccine and field strains, to investigate the role of wild birds in the epidemiology of MG and MS, and to investigate the role of horizontal or vertical transmission in outbreaks of MG or MS. Several studies evidenced the role of molecular epidemiology for the differentiation of vaccine and field strains and identified the persistence or spread of vaccine strains. By the use of Restriction Endonuclease Analysis (REA) MG vaccine strain could be differentiated from field strains (Yogev et al., Citation1988; Kleven, Browning et al., Citation1988; Ley et al., Citation1993). Furthermore, another study, using Random Amplified Polymorphic DNA (RAPD), indicated the persistence of a vaccine strain on a farm. In this study, RAPD patterns similar to that of vaccine strain 6/85 were identified on a farm where this vaccine had previously been used (Charlton et al., Citation1999). The use of MGc2 and MG IGSR typing showed the spread of live vaccine strain from vaccinated to non-vaccinated flocks in Jordan (Gharaibeh et al., Citation2011). Two genetic groups (A and B) of MG isolates could be differentiated, and multiple Jordan isolates could not be differentiated from the F-live vaccine strain. Only three isolates could be differentiated as field strains. As the flocks had not been vaccinated and F-live vaccine strain was commonly used in Jordan, the authors suggested it was likely that the F-live vaccine strain had spread from vaccinated to non-vaccinated farms and has, therefore, become the predominant genotype in Jordan. Also, an Egyptian study reported the spread of F-live vaccine strain from vaccinated to non-vaccinated flocks (Khalifa et al., Citation2014). In another study, the results of molecular genotyping were indicative for the potential spread of ts-11 live vaccine strain and subsequent studies suggested vertical transmission as well as a possible reversion of virulence (El Gazzar et al., Citation2011). Core genome MultiLocus Sequence Typing (CgMLST) based on 425 conserved genes showed that the ts-11-like isolates, previously obtained from outbreaks in commercial broiler flocks in North-eastern Georgia, were indeed very closely related to the ts-11 vaccine strains (Ghanem et al., Citation2018). This further suggested that the vaccine was the shared origin of this outbreak in the United States. Furthermore, this study also showed that some of the isolates originating from the UK were very closely related to 6/85 indicating a possible common origin for these isolates.

In another study, high sequence identity based on the MGc2 gene was shown between MG isolates obtained from commercial and backyard turkey flocks in Iran and some of the Iranian turkey isolates and Indian and Pakistan isolates (Rasoulinezhad et al., Citation2017). This finding indicated the presence of 6/85 vaccine-like strains in the field responsible for clinical outbreaks and which were different from the 6/85 vaccine strain. Discrimination of MS-H live vaccine and field strains by MLST demonstrated the possible spread of MS-H live vaccine from vaccinated to non-vaccinated birds (Dijkman et al., Citation2017).

Moreover, molecular tests have also been applied to elucidate the role of wild birds in the epidemiology of MG and MS. RAPD has contributed to unravel the epidemiology of outbreaks of MG in free-ranging house finches (Carpodacus mexicanus) which began in the eastern United States and spread throughout the United states, infecting not only house finches but also other songbirds (Fischer et al., Citation1997; Frasca et al., Citation1997; Ley, Berkhoff et al., Citation1997, Citation2006; Hartup et al., Citation2000; Mikaelian et al., Citation2001; Roberts, Nolan & Hill Citation2001; Roberts, Nolan, Lauerman et al., Citation2001; Wellehan, Calsamiglia et al., Citation2001; Wellehan, Zens et al., Citation2001; Pillai et al., Citation2003; Cherry et al., Citation2006). RAPD patterns of MG isolates from songbirds examined from 1994 through 1996 from 11 states, representing three host species, were identical but different from other isolates tested (Ley, Berkhoff et al., Citation1997). This finding was indicative of a clonal outbreak of MG in songbirds and suggested a single source (Ley, Berkhoff et al., Citation1997). The previous study also suggested that the outbreaks in finches were not related to MG outbreaks in commercial poultry. RAPD patterns of MG isolates involved in outbreaks of conjunctivitis in evening grosbeaks (Coccothraustes vespertinus) and pine grosbeaks (Pinicola enucleator) during the winter 1998–1999 in Canada were also identical to those of the house finches and gold finches suggesting an on-going epidemic of MG conjunctivitis in eastern North America (Mikaelian et al., Citation2001). By MLST identical sequence types were identified in backyard poultry and commercial poultry suggesting the potential spread of MG between backyard and commercial poultry (Ter Veen et al., Citation2021). VlhA sequence analysis was also shown to be not sufficiently discriminatory to distinguish between MS field and MS vaccine strains as was shown by the study of an MS isolate obtained from a captive lesser flamingo (Phoeniconaias minor) in an Italian zoo (Catania et al., Citation2016). Although no MS-H vaccination was used in the zoo, vlhA typing was unable to distinguish the MS isolate from the MS-H vaccine strain, while adding the sequence analysis of the obg gene suggested a major difference.

Finally, molecular tests have been used to elucidate the source of outbreaks with MG and MS. The use of REA showed that the MG isolate from a peacock was different from the MG isolates obtained from outbreaks in commercial poultry indicating that it was very unlikely that the outbreak in peacocks was the source of the outbreak in commercial poultry (Kleven, Morrow, et al., Citation1988). REA has also been used for MS (Morrow, Whithear et al., Citation1990) and was shown to be relevant for epidemiological questions as was demonstrated in an outbreak investigation of MS infection in North Carolina turkeys (Ley & Avakian, Citation1992) where considerable similarity in REA patterns was observed among MS field isolates suggesting that the outbreaks were related to a common source of infection. RAPD has also been applied by multiple research groups to unravel the epidemiology of MG and MS outbreaks in commercial poultry (Fan et al., Citation1995; Charlton et al., Citation1999). RAPD typing indicated that for MG in outbreaks in turkey flocks from different companies and farms in California (Charlton et al., Citation1999) there was no evidence for spread of MG between companies. It was evidenced by RAPD that a single strain was involved in outbreaks on ranches from the same company. The latter indicated the role of horizontal spread between farms from the same company and underlined the improvement of the hygiene management. RAPD and Pulsed-Field Gel Electrophoresis (PFGE) profiles of MS isolates obtained from 36 layer flocks were very homogeneous (97/99 isolates had similar PFGE and RAPD profiles), suggesting the horizontal transmission of a widely disseminated clone (Dufour-Gesbert et al., Citation2006). This also complicated determining the precise source of infection to better understand routes of transmission, and indicated that other techniques with higher discriminatory power were needed to further unravel the epidemiology of MS. The 5-gene MLST typing results of 209 MS isolates obtained from 15 different countries, different categories of poultry and farms, and type of lesions (Dijkman et al., Citation2016) showed different MLST sequence types following geographical origin supporting the hypothesis of regional population evolution. This typing scheme also showed its use for local epidemiological investigation of MS outbreaks. MLST was also used to investigate MG isolates collected from and including the years 2010–2019 and originating from MG outbreaks in Italy (Matucci et al., Citation2020). MLST clonal complex 1 was the most prevalent clonal complex detected in commercial poultry and also included strains from minor avian species suggesting a possible role of these birds in the evolution of these MG strains.

Monitoring programmes and complicating factors

Monitoring programmes and tests

Well-designed monitoring programmes are essential to be able to control horizontal and vertical spread of MG and/or MS. Although many countries implement a monitoring programme, little information about the contents and efficacy is available in the literature. Examples of successful programmes are the National Poultry Improvement Plan (NPIP) of the US Department of Agriculture (“The National Poultry Improvement Plan ”) for both MG and MS, and the European MG control programme for reproduction stock based on EU Council Directives 90/539/EEC and 2009/158/EC (EU, Citation1990, Citation2009). Both US and EU MG control programmes are based on taking a high number of samples starting at the end of the rearing period, frequent sampling in the production period, and culling of infected flocks. For the check of the rearing flocks, the NPIP requires 150 samples per house compared to 60 samples by the EU regulations. During production, the EU requires to take 60 samples per house every 90 days. The NPIP requires to test a total of 75 birds (75 serum samples) every 90 days, or 25 birds (25 serum samples) every 30 days. Breeding companies usually have very strict control programmes to keep all their grandparents and pedigree stock free from both MS and MG. In these programmes, the sampling frequency is usually higher than required by the national programmes, for example every 2–4 weeks.

The first monitoring programmes date from the early 1960s and were based on frequent seromonitoring of small flocks using the tube and rapid plate agglutination test, and the haemagglutination inhibition (HI) test, and frequent attempts to isolate MG from pipped embryos with airsacculitis (Fahey & Crawley, Citation1954b; Crawley & Fahey, Citation1955; Beckman et al., Citation1959). At that time, prevalence was high and there were no vaccines available for MG or MS. Testing of embryos with airsacculitis, using MG PCR or culture, is still performed. Nowadays, monitoring programmes are performed at a flock level and are still based on frequent blood sampling of poultry flocks and testing for the presence of antibodies against MG or MS, and/or the presence of the pathogen itself by culture or PCR (Buim et al., Citation2009; Kahya et al., Citation2010; Muhammad et al., Citation2018).

Studies also showed the potential of egg yolk for monitoring of antibodies at a flock level, but serum is much easier to process in a lab than egg yolk and results in far less biological waste (Mohammed et al., Citation1986; Brown et al., Citation1991; Kempf & Gesbert, Citation1998). Also, testing of day-old chickens (DOC) is performed when the MS or MG status of the breeder flocks is unknown. However, the interpretation of the results can be influenced by many factors, such as serological cross-reactions, antibiotic treatment, MG and/or MS vaccination of the breeder stock, and phase of the infection (Bradbury, Citation2005; Moronato et al., Citation2018). For many decades the rapid or serum plate agglutination (RPA or SPA) test and the haemagglutination (HI) test were the standard tests for the monitoring programmes of MS and MG. Difficulties regarding the interpretation of the HI test and RPA test were already recognized in early monitoring programmes (Fahey & Crawley, Citation1954a). In general, the specificity of RPA and HI tests has been shown to be high; however, several factors have been found that can decrease the specificity (see below). The percentage of RPA and HI non-specific reactions may be reduced by diluting the test serum (Ross et al., Citation1990; Feberwee et al., Citation2005, Citation2020; OIE, Citation2018). Nowadays, the RPA test is mainly used in monitoring programmes as a screening test as it is quick, relatively inexpensive, and highly sensitive due to its efficient detection of IgM antibodies (Kleven, Citation1975; Feberwee et al., Citation2005); MG and MS RPA antigens are still commercially available today. The HI test is less frequently used nowadays as the test is time-consuming, is dependent on the availability of non-commercial reagents, and cross-reactions between antigenically related strains can occur (Goren, Citation1979; Kleven, Morrow et al., Citation1988; Kleven et al., Citation2007; Dingfelder et al., Citation1991). Commercially available ELISAs are used by many laboratories, as screening test or as a confirmation test. In general, ELISA tests may be slightly less sensitive and more specific than RPA tests, and less specific but more sensitive than HI tests (Avakian et al., Citation1988; Kaszanyitzky et al., Citation1994; Kempf et al., Citation1994; Ewing, Kleven et al., Citation1996; Kempf & Gesbert, Citation1998; Feberwee et al., Citation2005).

Overall, papers show that there is no superior serological test for monitoring programmes; some studies reported that ELISA was not able to detect the antibodies against MG or MS where RPA was able to (Ewing et al., Citation1998; Kleven et al., Citation2007). On other occasions, both HI and RPA showed a low sensitivity compared to ELISA (Ewing, Lauerman et al., Citation1996). Regarding specificity, no serological test has a specificity of 100%. For this reason, it is recommended not to rely completely on only one test system (Feberwee et al., Citation2005). Further confirmation of serologic results may be by a second antibody test or by detection and identification of MG or MS by isolation or PCR (Luciano et al., Citation2011; OIE, Citation2018).

Complications from infections of other Mycoplasma species

Antibodies against heterologous Mycoplasma species that are present in the flock can interfere with serological monitoring for MG or MS, especially in the acute phase of the infection (Kempf et al., Citation1994; Ewing, Kleven et al., Citation1996; OIE, Citation2018; Feberwee et al., Citation2020). Mycoplasma species other than MG and MS, such as M. pullorum, M. gallinarum, M. gallinaceum, M. iners, M iowae, and M. imitans, are frequently found in commercial and backyard poultry (Bradbury & McClenaghan, Citation1982; Benčina, Dorrer et al., Citation1987; Benčina, Mrzel, et al., Citation1987; Wang et al., Citation1990; Bradbury et al., Citation1993, Citation2001a; Feberwee et al., Citation2020). Heterologous Mycoplasma species can share some common antigens as shown for MG, MS, and M. imitans using tests such as immunoblots, haemagglutination inhibition (HI) tests, rapid plate agglutination tests, and ELISAs (Bradley et al., Citation1988; Yogev et al., Citation1989; Avakian & Kleven, Citation1990a, Citation1990b; Bradbury et al., Citation1993). It has been shown under experimental and field conditions that this decrease of specificity due to these cross-reactions is usually transient and also depends on other factors, such as the antigens used in the test and serum dilution. Using a higher serum dilution can also lower the level of detected cross-reactions due to infections with heterologous Mycoplasma as shown for the RPA and HI test when using the 1:4 or 1:8 dilution as cut-off instead of undiluted or 1:2 diluted serum (Feberwee et al., Citation2005; OIE, Citation2018). Doubtful results for MG or MS are best investigated by performing tests with MS antigen (and vice versa) to obtain more information regarding potential cross-reactions (OIE, Citation2018).

Complications from the recent use of heterologous oil-emulsion vaccines

A transient increase of false-positive reactions in MG and MS serological tests occurs in some flocks recently vaccinated with oil-emulsion vaccines against various agents other than Mycoplasmas (Cullen & Timms, Citation1972; Glisson et al., Citation1984; Ahmad et al., Citation1988; Avakian et al., Citation1988; Yoder, Citation1989; Ross et al., Citation1990). The peak of the number of false-positive reactions is usually a few weeks post-vaccination, after which it gradually disappears in a number of weeks (Glisson et al., Citation1984). The cause is believed to be the use of products similar to serum components for the production of the Mycoplasma antigen and the production of the antigens for the inactivated vaccine, for example in cell culture or bacteriological culture. Examples of such products are horse serum, foetal calf serum and new-born calf serum (Bradbury & Jordan, Citation1973; Glisson et al., Citation1984). When chickens are injected with these antigens in oil emulsions, these also induce antibodies against the remains of the added serum components present in the vaccine. These antibodies are then detected by the similar serum components from the growth medium absorbed to the Mycoplasma cell during in vitro growth (Bradbury & Jordan, Citation1971, Citation1972). The use of purified Mycoplasma antigens is expected to decrease the rate of false-positive reactions resulting from antibodies in the test serum directed towards serum components. Using a higher serum dilution can also lower the level of false positives occurring after the recent use of an oil-inactivated vaccine, as shown for the RPA test when using the 1:4 or 1:8 dilution as cut-off instead of undiluted or 1:2 diluted serum (Ross et al., Citation1990). Another pragmatic and effective approach to distinguish the transient false positives occurring after the recent use of an oil-inactivated vaccine from true positives is to resample the suspected flock a few weeks later and perform the blood tests again. In the case of false positives in the first sampling due to the use of an inactivated vaccine these reactions usually disappear in several weeks.

Complications from antibiotic treatments

Antibiotic treatments that are effective against MG or MS may decrease or delay the antibody response to an infection, especially when the treatment is used in the early stage of the infection (Fahey & Crawley, Citation1955b; Jordan et al., Citation1989, Citation1999; Migaki et al., Citation1993; Jordan & Horrocks, Citation1996; Levisohn & Kleven, Citation2000; Stanley et al., Citation2001). It will also decrease the number of positives in the PCR and culture.

Complications from infections by low virulence strains

Many comparisons and publications have shown that RPA, HI, and ELISA tests will provide the same diagnosis in a big majority of infected and non-infected flocks. In a minority of cases, RPA, HI and/or ELISA provide conflicting results or all show a lower sensitivity than expected (Kleven et al., Citation2001), for example in the case of atypical or slow spreading MS and MG strains of low virulence (Yoder, Citation1986; Dingfelder et al., Citation1991; Ewing, Lauerman et al., Citation1996; Kempf et al., Citation1997). Kleven et al. (Citation2001) reported poor to inconsistent reactivity by RPA, ELISA, and HI-test in MS-infected turkey flocks. Experimental infection of turkeys with one of the isolates (K4463B) either by aerosol or systemically by a combination of intravenous, foot pad, and eyedrop routes showed a clear difference in serological responses. Turkeys challenged by the systemic route responded normally to all serologic tests, whereas those challenged by aerosol either responded very poorly on all serologic tests or were seronegative up to 6 weeks postchallenge, even though they were positive for MS by tracheal culture. These results suggest that turkeys may harbour an upper respiratory infection with MS, while remaining serologically negative. The study of Kempf et al. (Citation1997) also showed that challenge of chickens with a so-called atypical isolate leads to poor serological results and poor mycoplasma culture results.

Complications from the use of live or inactivated MG and MS vaccines

The use of MG or MS vaccines interferes with monitoring programmes. Both live and inactivated vaccines are expected to induce an antibody response. Inactivated MG and MS vaccines are expected to induce a strong serological response. To increase the immunogenicity of the inactivated antigens, immune-potentiating vaccine adjuvants are used (Panigraphy et al., Citation1981; Barbour & Newman, Citation1990).

The level of antibodies induced by live conventional vaccines varies and depends on, amongst other factors, the level of attenuation of the strain, way of application, and the interval between vaccination and sampling. In general, F strain-derived live vaccines induce higher levels of antibodies against MG than ts-11-derived vaccines, whereas 6/85-derived live vaccines often induce low or no detectable serological response during the first months post-vaccination (Whithear et al., Citation1990; Evans & Hafez, Citation1992; Abd-el-Motelib & Kleven, Citation1993; Ley, McLaren et al., Citation1997; Branton et al., Citation2002; Burnham et al., Citation2002; Noormohammadi et al., Citation2002; Noormohammadi & Whithear, Citation2019). The variation in serological responses to vaccination is underlined by the long-term study of Noormohammadi and Whithear (Citation2019) who reported a slowly increasing serological response to a 6/85 vaccine from 26% at 4 weeks post-vaccination to 100% at 10–36 weeks post-vaccination. In the same study, the initial 100% serological response to the ts-11 vaccine at 4 and 10 weeks post-vaccination dropped to 50–70% positives at 20–36 weeks post-vaccination. These variations in the levels of systemic antibodies after vaccination with live conventional MG or MS vaccines may complicate the diagnosis of a field challenge as seroconversions can also be caused by the variable response in time to the vaccination.

Moronato et al. (Citation2018) studied the serological response of broiler-breeders (vaccinated at week 14 with live vaccine MS-H) at 25, 30, 38, 44, 49, 54, and 59 weeks of age. No field strains were detected during this field study despite intensive monitoring. The birds were located in six barns. On average 79% and 85% of the sera were positive in the RPA and ELISA, respectively. However, there were significant differences in the percentage of positive samples over time and between barns. The difference in percentage of positive ELISA results between the barns at the same age varied from 20% to 100%. The birds of three barns had a consistently high percentage of positives at all ages where as the other barns showed a varying percentage of positives. The authors also report a high variation in ELISA titres.

The use of live vaccines also interferes with the use of culturing and PCR, for example as confirmation tests. Live MG and MS vaccines are expected to be detectable in the respiratory tract for several months to lifelong. In general, less attenuated strains can be detected in a higher percentage of the vaccinated birds and for a longer period (Kleven, Citation1981; Ley, McLaren et al., Citation1997; Noormohammadi & Whithear, Citation2019).

In case MG or MS vaccinated birds are monitored, a certain level of antibodies is already expected meaning that a challenge to a field strain could be detected based on a significant rise in antibody levels (Abd-el-Motelib & Kleven, Citation1993). For several vaccines, over the last years tests have become available that can differentiate vaccine from field strains, so-called DIVA tests (differentiating infected from vaccinated animals) (Shahid et al., Citation2014; Kreizinger et al., Citation2015; Dijkman et al., Citation2017; Zhu et al., Citation2017; Sulyok et al., Citation2019).

An alternative for conventional live MG and MS vaccines are vectorized recombinant vaccines expressing one of several proteins of MG or MS, such as a commercially available recombinant Fowl Pox vector expressing MG 40k and mgc genes (Zhang et al., Citation2010). The use of such a transient replicating vaccine that induces an immune response only to the proteins expressed from the inserted genes of Mycoplasma would cause hardly or no complications for tests used in monitoring programmes. The detectable serological response for commonly used ELISAs is expected to be very low or even absent (Zhang et al., Citation2010; Ferguson-Noel et al., Citation2012). Also, a MG PCR would only be able to detect this vaccine strain during its transient and short replication and only when the primers or probe were designed to detect the insert of the vector vaccine.

Today’s monitoring programmes and issues for consideration

Many developments have taken place since the first monitoring programmes were started. During past decades the worldwide poultry industry has grown considerably and, in parallel, flock sizes and number of birds per farm has also increased. This development increases the importance of a high quality of the diagnosis. Fortunately, the toolbox with diagnostic options and techniques has also grown. The use of the correct diagnostic tests, or combinations thereof, in the various field situations of low to high prevalence, whether or not in combination with vaccinations, requires a great deal of knowledge. shows an overview of the use of screening and confirmation tests in monitoring programmes in non-vaccinated and vaccinated flocks.

Table 1. Overview of the use of screening- and confirmation tests in monitoring programmes in non-vaccinated and vaccinated flocks.

Monitoring of non-vaccinated flocks that are expected to be free of infection is the simplest situation, as any seroconversion or detection of the pathogen indicates an infection. In this situation, false-positive results are a major concern as specificity of tests, certainly all serological tests, is not 100%. Especially when higher sample sizes are used to detect early infections of MG or MS, the risk of false-positive results increases significantly. The use of confirmation tests on sera that tested positive will further increase specificity and may further contribute to a correct decision on the MG or MS status of the flock (Martin et al., Citation1987). However, a matter of concern is that the use of combined tests to increase the specificity may decrease the sensitivity for the detection of an infection. As only positive sera are retested, a confirmation test can only lower the number of positive samples. This means that the demands of a confirmation test are not only a high specificity but also a high sensitivity. This requirement is especially important in the early phase of infections when some tests might detect the antibody response earlier than others (Avakian et al., Citation1988; Kempf et al., Citation1994; Feberwee et al., Citation2005; OIE, Citation2018). Confirmation of positive serological tests can also be performed using techniques to detect the bacterium itself, by culturing or, more commonly, by PCR (Ewing, Lauerman et al., Citation1996). Another way to increase the specificity of serological testing at flock level is to accept a low number or percentage of positive samples before a flock is considered to be positive. OIE (Citation2018) suggests 10% in the RPA MG (when no confirmation test is used). Feberwee et al. (Citation2008) and Ter Veen et al. (Citation2020) accepted one positive sample in the RPA MS using a serum dilution of 1:8.

Flocks that have only been vaccinated with an inactivated vaccine against MG or MS are expected to be positive in serological tests and negative in tests that show the presence of the pathogen. A peak of antibodies is expected shortly after the vaccination, following which the titres will gradually start to decline. A seroconversion during production or the detection of the pathogen will indicate an infection.

Most complicated is monitoring of flocks that have been vaccinated with a live, conventional vaccine. Live MG and MS vaccine strains may persist in the flock and may be detected by general MG and MS PCR tests. Live-vaccinated flocks are also expected to be seropositive to a certain extent, but the percentage of positives and titres vary in time and depend, amongst other factors, on the level of attenuation of the strain, way of application, and the interval between vaccination and sampling (Whithear et al., Citation1990; Evans & Hafez, Citation1992; Abd-el-Motelib & Kleven, Citation1993; Ley, McLaren et al., Citation1997; Branton et al., Citation2002; Burnham et al., Citation2002; Noormohammadi et al., Citation2002; Moronato et al., Citation2018; Noormohammadi & Whithear, Citation2019). Therefore, a DIVA PCR test may give more certainty to monitor field challenge as they are able to discriminate field strains from vaccine strains.

Monitoring of flocks that have only been vaccinated with a live vectored vaccine can be compared with monitoring of non-vaccinated flocks as the vaccine does not persist in the bird and it does not or hardly induce a detectable antibody response. The number of commercially available live vectored vaccines for MG and MS is still very low but that might change in the future due to the strong progress made in this area during the last decade.

To conclude, there is no monitoring programme that is suitable for all conditions; it should be suitable for the actual prevalence and whether flocks have been vaccinated or not. Whatever the situation, it is recommended not to rely solely on one test in monitoring programmes for MG and MS (Feberwee et al., Citation2005, Citation2020; Kahya et al., Citation2010; Luciano et al., Citation2011; OIE, Citation2018).

Concluding remarks

The increased knowledge, regarding the expression of clinical disease, epidemiology, vaccination, and diagnostic tools, has allowed further development of ways to control both MG and MS in the commercial poultry industry in the last six decades. Many developments have taken place since the first monitoring programmes were started, including the use of vaccines. Fortunately, the toolbox with diagnostic options and techniques has grown to meet the requirements of the industry. Recognition of new emerging strains and the synergistic effect of other factors in expressing the disease have contributed to the recognition of the clinical and economic importance of MS and MG. The latter has also contributed to the increased urge to unravel the epidemiology of both mycoplasma species to be able to improve the current monitoring and control programmes. There is no monitoring programme that is suitable for all conditions; a monitoring programme has to be suitable for the actual prevalence and whether flocks have been vaccinated or not. Whatever the situation, it is recommended not to rely solely on one test in monitoring programmes for MG and MS. Gaining more information on the expression of clinical disease caused by avian mycoplasma, its epidemiology, and expansion of well-defined collections of isolates from a wide geographical range and period in time are key for filling in the broader epidemiological picture and are crucial for further control of the disease in the commercial poultry industry. Combining all this information has led to novel and significant insights into the origin, diversification, and evolutionary process of avian mycoplasmas both on a local and global scale (Ley et al., Citation2016; Ball et al., Citation2018; De la Cruz et al., Citation2020; Matucci et al., Citation2020; Sawicka et al., Citation2020; Ter Veen et al., Citation2021). The poultry industry is expected to grow in the coming decades to be able to feed the expanding world population. Subsequently, this will lead to an increase in international trade of poultry and eggs, and the possible use of live and or inactivated vaccination programmes. With this background, tight biosecurity and the role of molecular tests with high discriminatory power as tools for unravelling epidemiological questions and knowledge on factors interfering with monitoring programmes are going to play an even more prominent role in the control of both mycoplasma species.

Supplemental material

Supplemental Material

Download MS Word (345.2 KB)

Disclosure statement

No potential conflict of interest was reported by the authors.

References

  • Abd-el-Motelib, T.Y. & Kleven, S.H. (1993). A comparative study of Mycoplasma gallisepticum vaccines in young chickens. Avian Diseases, 37, 981–987.
  • Abolnik, C. & Gouws, J. (2014). Extended survival times of Mycoplasma gallisepticum and Mycoplasma synoviae on kanekalon synthetic hair fibres. Poultry Science, 93, 8–11.
  • Ahmad, I., Kleven, S.H., Avakian, A.P. & Glisson, J.R. (1988). Sensitivity and specificity of Mycoplasma gallisepticum agglutination antigens prepared from medium with artificial liposomes substituting for serum. Avian Diseases, 32, 519–526.
  • Anderson, D.P. & Beard, C.W. (1967). Aerosol studies with avian mycoplasma. 2. Infectivity of Mycoplasma gallisepticum for chickens and turkeys. Avian Diseases, 11, 60–64.
  • Avakian, A.P. & Kleven, S.H. (1990a). Evaluation of sodium dodecyl sulfate-polyacrylamide gel electrophoresis purified proteins of Mycoplasma gallisepticum and M. synoviae as antigens in a dot-enzyme-linked immunosorbent assay. Avian Diseases, 34, 575–584.
  • Avakian, A.P. & Kleven, S.H. (1990b). The humoral immune response of chickens to Mycoplasma gallisepticum and Mycoplasma synoviae studied by immunoblotting. Veterinary Microbiology, 24, 155–169.
  • Avakian, A.P., Kleven, S.H. & Glisson, J.R. (1988). Evaluation of the specificity and sensitivity of two commercial enzyme-linked immunosorbent assay kits, the serum plate agglutination test, and the hemagglutination-inhibition test for antibodies formed in response to Mycoplasma gallisepticum. Avian Diseases, 32, 262–272.
  • Ball, C., Forrester, A. & Ganapathy, K. (2018). Co-circulation of genetically diverse population of vaccine related and unrelated respiratory mycoplasmas and viruses in UK poultry flocks with health or production problems. Veterinary Microbiology, 225, 132–138.
  • Barber, T.L. & Fabricant, J. (1962). Primary isolation of Mycoplasma organisms (PPLO) from mammalian sources. Journal of Bacteriology, 83, 1268–1273.
  • Barbour, E.K. & Newman, J.A. (1990). Preliminary data on efficacy of Mycoplasma gallisepticum vaccines containing different adjuvants in laying hens. Veterinary Immunology and Immunopathology, 26, 115–123.
  • Beckman, J.R., Dunlop, W.R. & Staples, W. (1959). Observations in the prevention of the CRD complex. Poultry Science, 38, 807–816.
  • Benčina, D., Dorrer, D. & Tadina, T. (1987). Mycoplasma species isolated from six avian species. Avian Pathology, 16, 653–664.
  • Benčina, D., Mrzel, I., Rojs, O.Z., Bidovec, A. & Dovč, A. (2003). Characterisation of Mycoplasma gallisepticum strains involved in respiratory disease in pheasants and peafowl. Veterinary Record, 152, 230–234.
  • Benčina, D., Mrzel, I., Tadina, T. & Dorrer, D. (1987). Mycoplasma species in chicken flocks with different management systems. Avian Pathology, 16, 599–608.
  • Benčina, D., Tadina, T. & Dorrer, D. (1988a). Natural infection of geese with Mycoplasma gallisepticum and Mycoplasma synoviae and egg transmission of the mycoplasmas. Avian Pathology, 17, 925–928.
  • Benčina, D., Tadina, T. & Dorrer, D. (1988b). Natural infection of ducks with Mycoplasma synoviae and Mycoplasma gallisepticum and Mycoplasma egg transmission. Avian Pathology, 17, 441–449.
  • Bradbury, J.M. (1984). Avian mycoplasma infections: prototype of mixed infections with mycoplasmas, bacteria and viruses. Annales de microbiologie, 135A, 83–89.
  • Bradbury, J.M. (2005). Summary workshop of European mycoplasma specialists. World’s Poultry Science, 61, 355–357.
  • Bradbury, J.M., Abdul-Wahab, O.M., Yavari, C.A., Dupiellet, J.P. & Bove, J.M. (1993). Mycoplasma imitans sp. nov. is related to Mycoplasma gallisepticum and found in birds. International Journal of Systematic Bacteriology, 43, 721–728.
  • Bradbury, J.M. & Howell, L.J. (1975). The response of chickens to experimental infection “em ovo” with Mycoplasma synoviae. Avian Pathology, 4, 277–286.
  • Bradbury, J.M. & Jordan, F.T. (1971). The absorption of -globulins to Mycoplasma gallisepticum and the possible role in non-specific serological reactions. Veterinary Record, 89, 318.
  • Bradbury, J.M. & Jordan, F.T. (1972). Studies on the adsorption of certain medium proteins to Mycoplasma gallisepticum and their influence on agglutination and haemagglutination reactions. Journal of Hygiene, 70, 267–278.
  • Bradbury, J.M. & Jordan, F.T. (1973). Non-specific agglutination of Mycoplasma gallisepticum. Veterinary Record, 92, 591–592.
  • Bradbury, J.M. & McClenaghan, M. (1982). Detection of mixed Mycoplasma species. Journal of Clinical Microbiology, 16, 314–318.
  • Bradbury, J.M., Yavari, C.A. & Dare, C.M. (2001a). Mycoplasmas and respiratory disease in pheasants and partridges. Avian Pathology, 30, 391–396.
  • Bradbury, J.M., Yavari, C.A. & Dare, C.M. (2001b). Detection of Mycoplasma synoviae in clinically normal pheasants. Veterinary Record, 148, 72–74.
  • Bradley, L.D., Snyder, D.B. & Van Deusen, R.A. (1988). Identification of species-specific and interspecies-specific polypeptides of Mycoplasma gallisepticum and Mycoplasma synoviae. American Journal of Veterinary Research, 49, 511–515.
  • Branton, S.L., Bearson, S.M., Bearson, B., Lott, B.D., Maslin, W.R., Collier, S.D., Pharr, G.T. & Boykin, D.L. (2002). The effects of 6/85 live Mycoplasma gallisepticum vaccine in commercial layer hens over a 43-week laying cycle on egg production, selected egg quality parameters, and egg size distribution when challenged before beginning of lay. Avian Diseases, 46, 423–428.
  • Brown, M.B., Stoll, M.L., Scasserra, A.E. & Butcher, G.D. (1991). Detection of antibodies to Mycoplasma gallisepticum in egg yolk versus serum samples. Journal of Clinical Microbiology, 29, 2901–2903.
  • Buim, M.R., Mettifogo, E., Timenetsky, J., Kleven, S. & Ferreira, A.J. (2009). Epidemiological survey on Mycoplasma gallisepticum and M. synoviae by multiplex PCR in commercial poultry. Pesquisa Veterinária Brasileira, 29, 552–556.
  • Buntz, B., Bradbury, J.M., Vuillaume, A. & Rousselot-Paillet, D. (1986). Isolation of Mycoplasma gallisepticum from geese. Avian Pathology, 15, 615–617.
  • Burnham, M.R., Branton, S.L., Peebles, E.D., Lott, B.D. & Gerard, P.D. (2002). Effects of F-strain Mycoplasma gallisepticum inoculation at twelve weeks of age on performance and egg characteristics of commercial egg-laying hens. Poultry Science, 81, 1478–1485.
  • Carnaghan, R.B. (1961). Egg transmission of infectious synovitis. Journal of Comparative Pathology and Therapeutics, 71, 279–285.
  • Catania, S., Bilato, D., Gobbo, F., Granato, A., Terregino, C., Iob, L. & Nicholas, R.A. (2010). Treatment of eggshell abnormalities and reduced egg production caused by Mycoplasma synoviae infection. Avian Diseases, 54, 961–964.
  • Catania, S., Gobbo, F., Ramirez, A.S., Guadagnini, D., Baldasso, E., Moronato, M.L. & Nicholas, R.A. (2016). Laboratory investigations into the origin of Mycoplasma synoviae isolated from a lesser flamingo (Phoeniconaias minor). BMC Veterinary Research, 12, 52.
  • Chalquest, R.R. & Fabricant, J. (1960). Pleuropneumonia-like organisms associated with synovitis in fowls. Avian Diseases, 4, 515–539.
  • Chandiramani, N.K., Van Roekel, H. & Olesiuk, O.M. (1966). Viability studies with Mycoplasma gallisepticum under different environmental conditions. Poultry Science, 45, 1029–1044.
  • Charlton, B.R., Bickford, A.A., Chin, R.P. & Walker, R.L. (1999). Randomly amplified polymorphic DNA (RAPD) analysis of Mycoplasma gallisepticum isolates from turkeys from the central valley of California. Journal of Veterinary Diagnostic Investigation, 11, 408–415.
  • Chen, H., Yu, S., Hu, M., Han, X., Chen, D., Qiu, X. & Ding, C. (2012). Identification of biofilm formation by Mycoplasma gallisepticum. Veterinary Microbiology, 161, 96–103.
  • Chen, S., Feng, Z., Sun, H., Zhang, R., Qin, T. & Peng, D. (2021). Biofilm-Formation-Related genes csgD and bcsA promote the vertical transmission of Salmonella enteritidis in chicken. Frontiers in Veterinary Science, 7, 625049.
  • Cherry, J.J., Ley, D.H. & Altizer, S. (2006). Genotypic analyses of Mycoplasma gallisepticum isolates from songbirds by random amplification of polymorphic DNA and amplified-fragment length polymorphism. Journal of Wildlife Diseases, 42, 421–428.
  • Chin, R.P., Daft, B.M., Meteyer, C.U. & Yamamoto, R. (1991). Meningoencephalitis in commercial meat turkeys associated with Mycoplasma gallisepticum. Avian Diseases, 35, 986–993.
  • Christensen, N.H., Yavari, C.A., McBain, A.J. & Bradbury, J.M. (1994). Investigations into the survival of Mycoplasma gallisepticum, Mycoplasma synoviae and Mycoplasma iowae on materials found in the poultry house environment. Avian Pathology, 23, 127–143.
  • Chu, H.P. & Uppal, P.K. (1975). Single and mixed infections of avian infectious bronchitis virus and Mycoplasma gallisepticum. Developments in Biological Standardization, 28, 101–114.
  • Chute, H.L., Cuozzo, T.R., Stauffer, T. & MacDonald, V. (1965). The commercial production of PPLO-free chickens. Canadian Veterinary Journal, 6, 19–21.
  • Cisneros-Tamayo, M., Kempf, I., Coton, J., Michel, V., Bougeard, S., de Boisseson, C., Lucas, P., Bayon-Auboyer, M.H., Chiron, G., Mindus, C. & Gautier-Bouchardon, A.V. (2020). Investigation on eggshell apex abnormality (EAA) syndrome in France: isolation of Mycoplasma synoviae is frequently associated with Mycoplasma pullorum. BMC Veterinary Research, 16, 271.
  • Cookson, K.C. & Shivaprasad, H.L. (1994). Mycoplasma gallisepticum infection in chukar partridges, pheasants, and peafowl. Avian Diseases, 38, 914–921.
  • Corstvet, R.E. & Sadler, W.W. (1966). A comparative study of single and multiple respiratory infections in the chicken: single infections (with Mycoplasma gallisepticum and Newcastle disease virus). American Journal of Veterinary Research, 27, 1721–1733.
  • Crawley, J.F. & Fahey, J.E. (1955). A proposed plan for the control of chronic respiratory disease of chickens. Poultry Science, 34, 707–716.
  • Cullen, G.A. & Timms, L.M. (1972). Diagnosis of mycoplasma infection in poultry previously vaccinated with killed adjuvant vaccines. British Veterinary Journal, 128, 94–100.
  • De la Cruz, L., Barrera, M., Rios, L., Corona-Gonzalez, B., Bulnes, C.A., Diaz-Sanchez, A.A., J, A.A., Lobo-Rivero, E. & Perez, L.J. (2020). Unraveling the global phylodynamic and phylogeographic expansion of Mycoplasma gallisepticum: understanding the origin and expansion of this pathogen in Ecuador. Pathogens (Basel, Switzerland), 9, 674.
  • Dijkman, R., Feberwee, A. & Landman, W.J. (2016). Development and evaluation of a multi-locus sequence typing scheme for Mycoplasma synoviae. Avian Pathology, 45, 426–442.
  • Dijkman, R., Feberwee, A. & Landman, W.J.M. (2017). Development, validation and field evaluation of a quantitative real-time PCR able to differentiate between field Mycoplasma synoviae and the MS-H-live vaccine strain. Avian Pathology, 46, 403–415.
  • Dingfelder, R.S., Ley, D.H., McLaren, J.M. & Brownie, C. (1991). Experimental infection of turkeys with Mycoplasma gallisepticum of low virulence, transmissibility, and immunogenicity. Avian Diseases, 35, 910–919.
  • Droual, R., Shivaprasad, H.L., Meteyer, C.U., Shapiro, D.P. & Walker, R.L. (1992). Severe mortality in broiler chickens associated with Mycoplasma synoviae and Pasteurella gallinarum. Avian Diseases, 36, 803–807.
  • Dufour-Gesbert, F., Dheilly, A., Marois, C. & Kempf, I. (2006). Epidemiological study on Mycoplasma synoviae infection in layers. Veterinary Microbiology, 114, 148–154.
  • Edward, D.G. & Kanarek, A.D. (1960). Organisms of the pleuropneumonia group of avian origin: their classification into species. Annals of the New York Academy of Sciences, 79, 696–702.
  • El Gazzar, M., Laibinis, V.A. & Ferguson-Noel, N. (2011). Characterization of a ts-11-like Mycoplasma gallisepticum isolate from commercial broiler chickens. Avian Diseases, 55, 569–574.
  • EU. (1990). Council Directive 90/539/EEC of 15 October 1990 on animal health conditions governing intra-Community trade in, and imports from third countries of, poultry and hatching eggs.
  • EU. (2009). CouncilDirective 2009/158/EC of 30 November 2009 on animal health conditions governing intra-Community trade in, and imports from third countries of, poultry and hatching eggs.
  • Evans, R.D. & Hafez, Y.S. (1992). Evaluation of a Mycoplasma gallisepticum strain exhibiting reduced virulence for prevention and control of poultry mycoplasmosis. Avian Diseases, 36, 197–201.
  • Ewing, M.L., Cookson, K.C., Phillips, R.A., Turner, K.R. & Kleven, S.H. (1998). Experimental infection and transmissibility of Mycoplasma synoviae with delayed serologic response in chickens. Avian Diseases, 42, 230–238.
  • Ewing, M.L., Kleven, S.H. & Brown, M.B. (1996). Comparison of enzyme-linked immunosorbent assay and hemagglutination-inhibition for detection of antibody to Mycoplasma gallisepticum in commercial broiler, fair and exhibition, and experimentally infected birds. Avian Diseases, 40, 13–22.
  • Ewing, M.L., Lauerman, L.H., Kleven, S.H. & Brown, M.B. (1996). Evaluation of diagnostic procedures to detect Mycoplasma synoviae in commercial multiplier-breeder farms and commercial hatcheries in Florida. Avian Diseases, 40, 798–806.
  • Fahey, J.E. & Crawley, J.F. (1954a). Studies on chronic respiratory disease of chickens IV. A hemagglutination inhibition diagnostic test. Canadian Journal of Comparative Medicine and Veterinary Science, 18, 264–272.
  • Fahey, J.E. & Crawley, J.F. (1954b). Studies on chronic respiratory disease of chickens. III. Egg transmission of a pleuropneumonia-like organism. Canadian Journal of Comparative Medicine and Science, 18, 67–75.
  • Fahey, J.E. & Crawley, J.F. (1955a). Studies on chronic respiratory disease of chickens. V. Air-borne spread of the CRD agents. Canadian Journal of Comparative Medicine and Science, 18, 264–272.
  • Fahey, J.E. & Crawley, J.F. (1955b). Studies on chronic respiratory disease of chickens VI. The effects of antibiotics on the clinical and serological course of CRD. Canadian Journal of Comparative Medicine and Science, 19, 281–286.
  • Fahey, J.E. & Crawley, J.F. (1956). Studies on chronic respiratory disease of chickens. VII. The nature of infection with the pleuropneumonia-like organisms. Canadian Journal of Comparative Medicine and Science, 20, 7–19.
  • Fan, H.H., Kleven, S.H. & Jackwood, M.W. (1995). Studies of intraspecies heterogeneity of Mycoplasma synoviae, M. meleagridis, and M. iowae with arbitrarily primed polymerase chain reaction. Avian Diseases, 39, 766–777.
  • Feberwee, A., Dijkman, R., Wiegel, J., Ter Veen, C., Bataille, H., Bouwstra, R. & de Wit, S. (2020). Rate of false positive reactions in 11 M. gallisepticum and M. synoviae serological tests in samples obtained from SPF birds inoculated with heterologous mycoplasma species. Avian Pathology, 49, 179–184.
  • Feberwee, A. & Landman, W.J. (2010). Induction of eggshell apex abnormalities in broiler breeder hens. Avian Pathology, 39, 133–137.
  • Feberwee, A., Mekkes, D.R., de Wit, J.J., Hartman, E.G. & Pijpers, A. (2005). Comparison of culture, PCR, and different serologic tests for detection of Mycoplasma gallisepticum and Mycoplasma synoviae infections. Avian Diseases, 49, 260–268.
  • Feberwee, A., de Vries, T.S. & Landman, W.J. (2008). Seroprevalence of Mycoplasma synoviae in Dutch commercial poultry farms. Avian Pathology, 37, 629–633.
  • Feberwee, A., de Wit, J.J. & Landman, W.J. (2009). Induction of eggshell apex abnormalities by Mycoplasma synoviae: field and experimental studies. Avian Pathology, 38, 77–85.
  • Ferguson-Noel, N., Cookson, K., Laibinis, V.A. & Kleven, S.H. (2012). The efficacy of three commercial Mycoplasma gallisepticum vaccines in laying hens. Avian Diseases, 56, 272–275.
  • Fischer, J.R., Stallknecht, D.E., Luttrell, P., Dhondt, A.A. & Converse, K.A. (1997). Mycoplasmal conjunctivitis in wild songbirds: the spread of a new contagious disease in a mobile host population. Emerging Infectious Diseases, 3, 69–72.
  • Floyd, S., Markham, F.S. & Wong, S.C. (1952). Pleuropneumonia-like organisms in the etiology of turkey sinusitis and chronic respiratory disease of chickens. Poultry Science, 31, 902–904.
  • Frasca, S., Jr., Hinckley, L., Forsyth, M.H., Gorton, T.S., Geary, S.J. & Van Kruiningen, H.J. (1997). Mycoplasmal conjunctivitis in a European starling. Journal of Wildlife Diseases, 33, 336–339.
  • Ghanem, M., Wang, L., Zhang, Y., Edwards, S., Lu, A., Ley, D. & El-Gazzar, M. (2018). Core genome multilocus sequence typing: a standardized approach for molecular typing of Mycoplasma gallisepticum. Journal of Clinical Microbiology, 56, 1–11.
  • Gharaibeh, S., Laibinis, V., Wooten, R., Stabler, L. & Ferguson-Noel, N. (2011). Molecular characterization of Mycoplasma gallisepticum isolates from Jordan. Avian Diseases, 55, 212–216.
  • Ghazikhanian, G., Yamamoto, R. & Cordy, D.R. (1973). Response of turkeys to experimental infection with Mycoplasma synoviae. Avian Diseases, 17, 122–136.
  • Giambrone, J.J., Eidson, C.S. & Kleven, S.H. (1977). Effect of infectious bursal disease on the response of chickens to Mycoplasma synoviae, Newcastle disease virus, and infectious bronchitis virus. American Journal of Veterinary Research, 38, 251–253.
  • Glisson, J.R., Dawe, J.F. & Kleven, S.H. (1984). The effect of oil-emulsion vaccines on the occurrence of nonspecific plate agglutination reactions for Mycoplasma gallisepticum and M. synoviae. Avian Diseases, 28, 397–405.
  • Gole, V.C., Chousalkar, K.K. & Roberts, J.R. (2012). Prevalence of antibodies to Mycoplasma synoviae in laying hens and possible effects on egg shell quality. Preventive Veterinary Medicine, 106, 75–78.
  • Goren, E. (1978). Effects of Mycoplasma synoviae infection on the state of health, reactions to vaccination and results of fattening in broiler chickens. Tijdschrift voor Diergeneeskunde, 103, 36–41.
  • Goren, E. (1979). Haemadsorption inhibition test for the identification of Mycoplasma gallisepticum and Mycoplasma synoviae. Veterinary Quarterly, 1, 158–162.
  • Gross, W.B. (1958). Symposium on chronic respiratory diseases of poultry. II. The role of Escherichia coli in the cause of chronic respiratory disease and certain other respiratory diseases. American Journal of Veterinary Research, 19, 448–452.
  • Gross, W.B. (1961). The development of “air sac disease”. Avian Diseases, 5, 431–439.
  • Hartup, B.K., Kollias, G.V. & Ley, D.H. (2000). Mycoplasmal conjunctivitis in songbirds from New York. Journal of Wildlife Diseases, 36, 257–264.
  • Hopkins, S.R. & Yoder, H.W., Jr. (1982). Influence of infectious bronchitis strains and vaccines on the incidence of Mycoplasma synoviae airsacculitis. Avian Diseases, 26, 741–752.
  • Hopkins, S.R. & Yoder, H.W., Jr. (1984). Increased incidence of airsacculitis in broilers infected with Mycoplasma synoviae and chicken-passaged infectious bronchitis vaccine virus. Avian Diseases, 28, 386–396.
  • Jeon, E.O., Kim, J.N., Lee, H.R., Koo, B.S., Min, K.C., Han, M.S., Lee, S.B., Bae, Y.J., Mo, J.S., Cho, S.H., Lee, C.H. & Mo, I.P. (2014). Eggshell apex abnormalities associated with Mycoplasma synoviae infection in layers. Journal of Veterinary Science, 15, 579–582.
  • Jordan, F.T. (1975). Avian mycoplasma and pathogenicity – a review. Avian Pathology, 4, 165–174.
  • Jordan, F.T., Forrester, C.A., Hodge, A. & Reeve-Johnson, L.G. (1999). The comparison of an aqueous preparation of tilmicosin with tylosin in the treatment of Mycoplasma gallisepticum infection of turkey poults. Avian Diseases, 43, 521–525.
  • Jordan, F.T., Gilbert, S., Knight, D.L. & Yavari, C.A. (1989). Effects of baytril, tylosin and iamulin on avian mycoplasmas. Avian Pathology, 18, 659–673.
  • Jordan, F.T. & Horrocks, B.K. (1996). The minimum inhibitory concentration of tilmicosin and tylosin for Mycoplasma gallisepticum and Mycoplasma synoviae and a comparison of their efficacy in the control of Mycoplasma gallisepticum infection in broiler chicks. Avian Diseases, 40, 326–334.
  • Kahya, S., Temelli, S., Eyigor, A. & Carli, K.T. (2010). Real-time PCR culture and serology for the diagnosis of Mycoplasma gallisepticum in chicken breeder flocks. Veterinary Microbiology, 144, 319–324.
  • Kaszanyitzky, E., Czifra, G. & Stipkovits, L. (1994). Detection of Mycoplasma gallisepticum antibodies in turkey blood samples by ELISA and by the slide agglutination and haemagglutination inhibition tests. Acta Veterinaria Hungarica, 42, 69–78.
  • Kato, K. (1965). Infectious coryza of chickens. V. Influence of Mycoplasma gallisepticum infection on chicken infected with Haemophilus gallinarum. National Institute of Animal Health Quarterly, 5, 183–189.
  • Kempf, I., Cacou, P.M., Guittet, M., Ollivier, C., Morin, M., L’Hospitalier, R. & Bennejean, G. (1988). Experimental infection with Mycoplasma gallisepticum: influence of ammonia as an exacerbating factor. Avian Pathology, 17, 601–615.
  • Kempf, I. & Gesbert, F. (1998). Comparison of serological tests for detection of Mycoplasma gallisepticum antibodies in eggs and chicks hatched from experimentally infected hens. Veterinary Microbiology, 60, 207–213.
  • Kempf, I., Gesbert, F. & Guittet, M. (1997). Experimental infection of chickens with an atypical Mycoplasma gallisepticum strain: comparison of diagnostic methods. Research in Veterinary Science, 63, 211–213.
  • Kempf, I., Gesbert, F., Guittet, M., Bennejean, G. & Stipkovits, L. (1994). Evaluation of two commercial enzyme-linked immunosorbent assay kits for the detection of Mycoplasma gallisepticum antibodies. Avian Pathology, 23, 329–338.
  • Kerr, K.M. & Olson, N.O. (1969). Pathology of chickens experimentally inoculated or contact-infected with an arthritis-producing virus. Avian Diseases, 13, 729–745.
  • Khalifa, R., Eissa, S., El-Hariri, M. & Refai, M. (2014). Sequencing analysis of Mycoplasma gallisepticum wild strains in vaccinated chicken breeder flocks. Journal of Molecular Microbiology and Biotechnology, 24, 98–104.
  • Khehra, R.S., Jones, R.C. & Bradbury, J.M. (1999). Dual infection of turkey poults with avian pneumovirus and Mycoplasma synoviae. Avian Pathology, 28, 401–404.
  • King, D.D., Kleven, S.H., Wenger, D.M. & Anderson, D.P. (1973). Field studies with Mycoplasma synoviae. Avian Diseases, 17, 722–726.
  • Kleven, S.H. (1975). Antibody response to avian mycoplasmas. American Journal of Veterinary Research, 36, 563–565.
  • Kleven, S.H. (1981). Transmissibility of the F strain of Mycoplasma gallisepticum in leghorn chickens. Avian Diseases, 25, 1005–1018.
  • Kleven, S.H. (1997). Changing expectations in the control of Mycoplasma gallisepticum. Acta Veterinaria Hungarica, 45, 299–305.
  • Kleven, S.H. (2008). Control of avian mycoplasma infections in commercial poultry. Avian Diseases, 52, 367–374.
  • Kleven, S.H., Browning, G.F., Bulach, D.M., Ghiocas, E., Morrow, C.J. & Whithear, K.G. (1988). Examination of Mycoplasma gallisepticum strains using restriction endonuclease DNA analysis and DNA-DNA hybridisation. Avian Pathology, 17, 559–570.
  • Kleven, S.H., Ferguson-Noel, N., Raviv, Z., Wooten, R. & Laibinis, V. (2007). Serological responses of chickens to low challenge doses of Mycoplasma synoviae. Avian Diseases, 51, 738–743.
  • Kleven, S.H., Fletcher, O.J. & Davis, R.B. (1975). Influence of strain of Mycoplasma synoviae and route of infection on development of synovitis or airsacculitis in broilers. Avian Diseases, 19, 126–135.
  • Kleven, S.H. & Fletcher, W.O. (1983). Laboratory infection of house sparrows (Passer domesticus) with Mycoplasma gallisepticum and Mycoplasma synoviae. Avian Diseases, 27, 308–311.
  • Kleven, S.H., King, D.D. & Anderson, D.P. (1972). Airsacculitis in broilers from Mycoplasma synoviae: effect on air-sac lesions of vaccinating with infectious bronchitis and Newcastle virus. Avian Diseases, 16, 915–924.
  • Kleven, S.H., Morrow, C.J. & Whithear, K.G. (1988). Comparison of Mycoplasma gallisepticum strains by hemagglutination-inhibition and restriction endonuclease analysis. Avian Diseases, 32, 731–741.
  • Kleven, S.H., Rowland, G.N. & Kumar, M.C. (2001). Poor serologic response to upper respiratory infection with Mycoplasma synoviae in turkeys. Avian Diseases, 45, 719–723.
  • Kreizinger, Z., Sulyok, K.M., Pasztor, A., Erdelyi, K., Felde, O., Povazsan, J., Korosi, L. & Gyuranecz, M. (2015). Rapid, simple and cost-effective Molecular method to differentiate the temperature sensitive (ts+) MS-H vaccine strain and wild-type Mycoplasma synoviae isolates. PLoS One, 10, e0133554.
  • Kursa, O., Pakula, A., Tomczyk, G., Pasko, S. & Sawicka, A. (2019). Eggshell apex abnormalities caused by two different Mycoplasma synoviae genotypes and evaluation of eggshell anomalies by full-field optical coherence tomography. BMC Veterinary Research, 15, 1.
  • Landman, W.J. (2014). Is Mycoplasma synoviae outrunning Mycoplasma gallisepticum? A viewpoint from the netherlands. Avian Pathology, 43, 2–8.
  • Landman, W.J. & Bronneberg, R.G. (2003). [Mycoplasma synoviae-associated amyloid arthropathy in white leghorns: case report]. Tijdschrift voor Diergeneeskunde, 128, 36–40.
  • Landman, W.J. & Feberwee, A. (2001). Field studies on the association between amyloid arthropathy and Mycoplasma synoviae infection, and experimental reproduction of the condition in brown layers. Avian Pathology, 30, 629–639.
  • Landman, W.J. & Feberwee, A. (2004). Aerosol-induced Mycoplasma synoviae arthritis: the synergistic effect of infectious bronchitis virus infection. Avian Pathology, 33, 591–598.
  • Landman, W.J. & Feberwee, A. (2012). Longitudinal field study on the occurrence of Mycoplasma synoviae in Dutch turkey flocks with lameness and experimental induction of the condition. Avian Pathology, 41, 141–149.
  • Leigh, S.A., Branton, S.L., Evans, J.D. & Collier, S.D. (2012). Effect of infection route and concurrent infectious bronchitis virus vaccination on Mycoplasma gallisepticum disease pathology in an experimental model. Avian Pathology, 41, 497–503.
  • Levisohn, S. & Kleven, S.H. (2000). Avian mycoplasmosis (Mycoplasma gallisepticum). Revue Scientifique et Technique Office International des Epizooties, 19, 425–442.
  • Ley, D.H. & Avakian, A.P. (1992). An outbreak of Mycoplasma synoviae infection in North Carolina turkeys: comparison of isolates by sodium dodecyl sulfate-polyacrylamide gel electrophoresis and restriction endonuclease analysis. Avian Diseases, 36, 672–678.
  • Ley, D.H., Avakian, A.P. & Berkhoff, J.E. (1993). Clinical Mycoplasma gallisepticum infection in multiplier breeder and meat turkeys caused by F strain: identification by sodium dodecyl sulfate-polyacrylamide gel electrophoresis, restriction endonuclease analysis, and the polymerase chain reaction. Avian Diseases, 37, 854–862.
  • Ley, D.H., Berkhoff, J.E. & Levisohn, S. (1997). Molecular epidemiologic investigations of Mycoplasma gallisepticum conjunctivitis in songbirds by random amplified polymorphic DNA analyses. Emerging Infectious Diseases, 3, 375–380.
  • Ley, D.H., Berkhoff, J.E. & McLaren, J.M. (1996). Mycoplasma gallisepticum isolated from house finches (Carpodacus mexicanus) with conjunctivitis. Avian Diseases, 40, 480–483.
  • Ley, D.H., Hawley, D.M., Geary, S.J. & Dhondt, A.A. (2016). House finch (Haemorhous mexicanus) conjunctivitis, and Mycoplasma spp. isolated from North American wild birds, 1994–2015. Journal of Wildlife Diseases, 52, 669–673.
  • Ley, D.H., McLaren, J.M., Miles, A.M., Barnes, H.J., Miller, S.H. & Franz, G. (1997). Transmissibility of live Mycoplasma gallisepticum vaccine strains ts-11 and 6/85 from vaccinated layer pullets to sentinel poultry. Avian Diseases, 41, 187–194.
  • Ley, D.H., Sheaffer, D.S. & Dhondt, A.A. (2006). Further western spread of Mycoplasma gallisepticum infection of house finches. Journal of Wildlife Diseases, 42, 429–431.
  • Lin, M.Y. & Kleven, S.H. (1982a). Pathogenicity of two strains of Mycoplasma gallisepticum in turkeys. Avian Diseases, 26, 360–364.
  • Lin, M.Y. & Kleven, S.H. (1982b). Egg transmission of two strains of Mycoplasma gallisepticum in chickens. Avian Diseases, 26, 487–495.
  • Lorenc, Z., Pasko, S., Kursa, O., Pakula, A. & Salbut, L. (2019). Spectral technique for detection of changes in eggshells caused by Mycoplasma synoviae. Poultry Science, 98, 3481–3487.
  • Lott, B.D., Drott, T.H., Vardaman, T.H. & Reece, F.N. (1978). Effect of Mycoplasma synoviae on egg quality and egg production of broiler breeders. Poultry Science, 57, 309–311.
  • Luciano, R.L., Cardoso, A.L., Stoppa, G.F., Kanashiro, A.M., de Castro, A.G. & Tessari, E.N. (2011). Comparative study of serological tests for Mycoplasma synoviae diagnosis in commercial poultry breeders. Veterinary Medicine International, 2011, 1.
  • Macowan, K.J., Randall, C.J., Jones, H.G. & Brand, T.F. (1982). Association of Mycoplasma synoviae with respiratory disease of broilers. Avian Pathology, 11, 235–244.
  • Madden, D.L., Henderson, W.H. & Moses, H.E. (1967). Case report: isolation of Mycoplasma gallisepticum from bobwhite quail (Colinus virginianus). Avian Diseases, 11, 378–380.
  • Markham, F.S. & Wong, S.C. (1952). Pleuropneumonia-like organisms in the etiology of turkey sinusitis and chronic respiratory disease of chickens. Poultry Science, 31, 902–904.
  • Marois, C., Dufour-Gesbert, F. & Kempf, I. (2000). Detection of Mycoplasma synoviae in poultry environment samples by culture and polymerase chain reaction. Veterinary Microbiology, 73, 311–318.
  • Marois, C., Picault, J.P., Kobisch, M. & Kempf, I. (2005). Experimental evidence of indirect transmission of Mycoplasma synoviae. Veterinary Research, 36, 759–769.
  • Martin, S.W., Meek, A.H. & Willeberg, P. (1987). Veterinary epidemiology, principles and methods. Ames: Iowa State University Press.
  • Matucci, A., Stefani, E., Gastaldelli, M., Rossi, I., De Grandi, G., Gyuranecz, M. & Catania, S. (2020). Molecular differentiation of Mycoplasma gallisepticum outbreaks: a last decade study on Italian farms using GTS and MLST. Vaccines (Basel), 8, 665.
  • McMartin, D.A. (1968). Latent infection of chickens with Mycoplasma gallisepticum. Veterinary Record, 83, 280.
  • McMartin, D.A., Khan, M.I., Farver, T.B. & Christie, G. (1987). Delineation of the lateral spread of Mycoplasma gallisepticum infection in chickens. Avian Diseases, 31, 814–819.
  • Michiels, T., Welby, S., Vanrobaeys, M., Quinet, C., Rouffaer, L., Lens, L., Martel, A. & Butaye, P. (2016). Prevalence of Mycoplasma gallisepticum and Mycoplasma synoviae in commercial poultry, racing pigeons and wild birds in Belgium. Avian Pathology, 45, 244–252.
  • Migaki, T.T., Avakian, A.P., Barnes, H.J., Ley, D.H., Tanner, A.C. & Magonigle, R.A. (1993). Efficacy of danofloxacin and tylosin in the control of mycoplasmosis in chicks infected with tylosin-susceptible or tylosin-resistant field isolates of Mycoplasma gallisepticum. Avian Diseases, 37, 508–514.
  • Mikaelian, I., Ley, D.H., Claveau, R., Lemieux, M. & Berube, J.P. (2001). Mycoplasmosis in evening and pine grosbeaks with conjunctivitis in Quebec. Journal of Wildlife Diseases, 37, 826–830.
  • Mohammed, H.O., Carpenter, T.E. & Yamamoto, R. (1987). Evaluation of factors associated with infection of commercial layers with Mycoplasma gallisepticum and M. synoviae. Avian Diseases, 31, 470–476.
  • Mohammed, H.O., Yamamoto, R., Carpenter, T.E. & Ortmayer, H.B. (1986). Comparison of egg yolk and serum for the detection of Mycoplasma gallisepticum and M. synoviae antibodies by enzyme-linked immunosorbent assay. Avian Diseases, 30, 398–408.
  • Moronato, M.L., Cecchinato, M., Facchetti, G., Mainenti, M., Gobbo, F. & Catania, S. (2018). Application of different laboratory techniques to monitor the behaviour of a Mycoplasma synoviae vaccine (MS-H) in broiler breeders. BMC Veterinary Research, 14, 357.
  • Morrison, C.A., Bradbury, E.M. & Garrett, R.A. (1977). A comparison of the structures of several acid-urea extracted ribosomal proteins from Escherichia coli using proton NMR. FEBS Letters, 81, 435–439.
  • Morrow, C.J., Bell, I.G., Walker, S.B., Markham, P.F., Thorp, B.H. & Whithear, K.G. (1990). Isolation of Mycoplasma synoviae from infectious synovitis of chickens. Australian Veterinary Journal, 67, 121–124.
  • Morrow, C.J., Bradbury, J.M., Gentle, M.J. & Thorp, B.H. (1997). The development of lameness and bone deformity in the broiler following experimental infection with Mycoplasma gallisepticum or Mycoplasma synoviae. Avian Pathology, 26, 169–187.
  • Morrow, C.J., Whithear, K.G. & Kleven, S.H. (1990). Restriction endonuclease analysis of Mycoplasma synoviae strains. Avian Diseases, 34, 611–616.
  • Muhammad, F., Hussain, J., Fareed, S.K., Ahmad Khan, T., Ahmad Khan, S. & Ahmad, A. (2018). Diagnosis of avian Mycoplasmas: a comparison between PCR and culture technique. Archives if Razi Institute, 73, 239–244.
  • The National Poultry Improvement Plan. From Poultry improvement.org
  • Naylor, C.J., Al-Ankari, A.R., Al-Afaleq, A.I., Bradbury, J.M. & Jones, R.C. (1992). Exacerbation of Mycoplasma gallisepticum infection in turkeys by rhinotracheitis virus. Avian Pathology, 21, 295–305.
  • Nelson, J.B. (1935). Cocco-bacilliform bodies associated with an infectious fowl coryza. Science, 82, 43–44.
  • Nelson, J.B. (1936). Studies on an uncomplicated coryza of the domestic fowl: V. A coryza of slow onset. Journal of Experimental Medicine, 63, 509–513.
  • Nonomura, I. & Sato, S. (1971). Effect of the Newcastle disease virus TCND strain on Mycoplasma gallisepticum infection in chickens. National Institute of Animal Health Quarterly, 11, 184–190.
  • Noormohammadi, A.H., Jones, J.E., Underwood, G. & Whithear, K.G. (2002). Poor systemic antibody response after vaccination of commercial broiler breeders with Mycoplasma gallisepticum vaccine ts-11 not associated with susceptibility to challenge. Avian Diseases, 46, 623–628.
  • Noormohammadi, A.H. & Whithear, K.G. (2019). Comparison of the short-term and long-term efficacies of the Mycoplasma gallisepticum vaccines ts-11 and 6/85. Avian Pathology, 48, 238–244.
  • OIE. (2018). Chapter 3.3.5. Avian Mycoplasmosis (Mycoplasma gallisepticum and Mycoplasma synoviae).
  • Olesiuk, O.M. & Van Roekel, H. (1960). Transmission of chronic respiratory disease in chickens. Avian Diseases, 4, 348–368.
  • Olson, N.O., Adler, H.E., DaMassa, A.J. & Corstvet, R.E. (1964). The effect of intranasal exposure to Mycoplasma synoviae and infectious bronchitis on development of lesions and agglutinins. Avian Diseases, 8, 623–631.
  • Olson, N.O., Bletner, J.K., Shelton, D.C., Munro, D.A. & Anderson, G.C. (1954). Enlarged joint condition in poultry caused by an infectious agent. Poultry Science, 33, 1075.
  • Olson, N.O., Kerr, K.M. & Campbell, A. (1964). Control of infectious synovitis 13. The antigen study of three strains. Avian Diseases, 8, 209–214.
  • Omuro, M., Suzuki, K., Kawamura, H. & Munakata, K. (1971). Interaction of Mycoplasma gallisepticum, mild strains of Newcastle disease virus and infectious bronchitis virus in chickens. National Institute of Animal Health Quarterly, 11, 83–93.
  • Osorio, C., Fletcher, O.J., Abdul-Aziz, T., Gonder, E., Tilley, B. & Ley, D.H. (2007). Pneumonia of turkey breeder hens associated with Mycoplasma synoviae. Avian Diseases, 51, 791–796.
  • Panigraphy, B., Grumbles, L.C. & Hall, C.F. (1981). Immunogenic potency of oil-emulsified Mycoplasma gallisepticum bacterins. Avian Diseases, 25, 821–826.
  • Pascucci, S., Maestrini, N., Govoni, S. & Prati, A. (1976). Mycoplasma synoviae in the guinea-fowl. Avian Pathology, 5, 291–297.
  • Pillai, S.R., Mays, H.L., Jr., Ley, D.H., Luttrell, P., Panangala, V.S., Farmer, K.L. & Roberts, S.R. (2003). Molecular variability of house finch Mycoplasma gallisepticum isolates as revealed by sequencing and restriction fragment length polymorphism analysis of the pvpA gene. Avian Diseases, 47, 640–648.
  • Poveda, J.B., Fernandez, A., Carranza, J., Hermoso, M. & Perea, J.A. (1986). Isolation of Mycoplasma synoviae from the red-legged partridge (Alectoris rufa). Avian Pathology, 15, 797–802.
  • Ranck, M.F., Schmidt, V., Philipp, H.C., Voss, M., Kacza, J., Richter, A., Fehlhaber, K. & Krautwald-Junghanns, M.E. (2010). Mycoplasma synoviae-associated egg-pole shell defects in laying hens. Berliner und Munchener Tierarztliche Wochenschrift, 123, 111–118.
  • Rasoulinezhad, S., Bozorgmehrifard, M.H., Hosseini, H., Sheikhi, N. & Charkhkar, S. (2017). Molecular detection and phylogenetic analysis of Mycoplasma gallisepticum from backyard and commercial turkey flocks in Iran. Veterinary Research Forum, 8, 293–298.
  • Raviv, Z., Ferguson-Noel, N., Laibinis, V., Wooten, R. & Kleven, S.H. (2007). Role of Mycoplasma synoviae in commercial layer Escherichia coli peritonitis syndrome. Avian Diseases, 51, 685–690.
  • Reis, R. & Yamamoto, R. (1971). Pathogenesis of single and mixed infections caused by Mycoplasma meleagridis and Mycoplasma gallisepticum in turkey embryos. American Journal of Veterinary Research, 32, 63–74.
  • Rhoades, K.R. (1977). Turkey sinusitis: synergism between Mycoplasma synoviae and Mycoplasma meleagridis. Avian Diseases, 21, 670–674.
  • Roberts, D.H. & McDaniel, J.W. (1967). Mechanism of egg transmission of Mycoplasma gallisepticum. Journal of Comparative Pathology, 77, 439–442.
  • Roberts, S.R., Nolan, P.M. & Hill, G.E. (2001). Characterization of Mycoplasma gallisepticum infection in captive house finches (Carpodacus mexicanus) in 1998. Avian Diseases, 45, 70–75.
  • Roberts, S.R., Nolan, P.M., Lauerman, L.H., Li, L.Q. & Hill, G.E. (2001). Characterization of the mycoplasmal conjunctivitis epizootic in a house finch population in the southeastern USA. Journal of Wildlife Diseases, 37, 82–88.
  • Rodriguez, R. & Kleven, S.H. (1980). Pathogenicity of two strains of Mycoplasma gallisepticum in broilers. Avian Diseases, 24, 800–807.
  • Ross, T., Slavik, M., Bayyari, G. & Skeeles, J. (1990). Elimination of mycoplasmal plate agglutination cross-reactions in sera from chickens inoculated with infectious bursal disease viruses. Avian Diseases, 34, 663–667.
  • Roussan, D.A., Al-Rifai, R.H., Khawaldeh, G.Y., Totanji, W.S. & Shaheen, I. (2011). Ornithobacterium rhinotracheale and Mycoplasma synoviae in broiler chickens in Jordan. Revue Scientifique et Technique Office International des Epizooties, 30, 931–937.
  • Sahu, S.P. & Olson, N.O. (1981). Characterization of an isolate of mycoplasma WVU 907 which possesses common antigens to Mycoplasma gallisepticum. Avian Diseases, 25, 943–953.
  • Sato, S., Nonomura, I., Shimizu, F., Shoya, S. & Horiuchi, T. (1970). Mixed infection with Mycoplasma gallisepticum and the B1 strain of Newcastle disease virus in chickens. National Institute of Animal Health Quarterly, 10, 58–65.
  • Sato, S., Shoya, S. & Kobayashi, H. (1973). Effect of ammonia on Mycoplasma gallisepticum infection in chickens. National Institute of Animal Health Quarterly, 13, 45–53.
  • Sawicka, A., Durkalec, M., Tomczyk, G. & Kursa, O. (2020). Occurrence of Mycoplasma gallisepticum in wild birds: a systematic review and meta-analysis. PLoS One, 15, e0231545.
  • Shahid, M.A., Markham, P.F., Marenda, M.S., Agnew-Crumpton, R. Noormohammadi, A.H. & Balish, M.F. (2014). High-resolution melting-curve analysis of obg gene to differentiate the temperature-sensitive Mycoplasma synoviae vaccine strain MS-H from non-temperature-sensitive strains. PLoS One, 9, e92215.
  • Springer, W.T., Luskus, C. & Pourciau, S.S. (1974). Infectious bronchitis and mixed infections of Mycoplasma synoviae and Escherichia coli in gnotobiotic chickens I. synergistic role in the airsacculitis syndrome. Infection and Immunity, 10, 578–589.
  • Stallknecht, D.E., Luttrell, M.P., Fischer, J.R. & Kleven, S.H. (1998). Potential for transmission of the finch strain of Mycoplasma gallisepticum between house finches and chickens. Avian Diseases, 42, 352–358.
  • Stanley, W.A., Hofacre, C.L., Speksnijder, G., Kleven, S.H. & Aggrey, S.E. (2001). Monitoring Mycoplasma gallisepticum and Mycoplasma synoviae infection in breeder chickens after treatment with enrofloxacin. Avian Diseases, 45, 534–539.
  • Stipkovits, L., Egyed, L., Palfi, V., Beres, A., Pitlik, E., Somogyi, M., Szathmary, S. & Denes, B. (2012). Effect of low-pathogenicity influenza virus H3N8 infection on Mycoplasma gallisepticum infection of chickens. Avian Pathology, 41, 51–57.
  • Stipkovits, L., Glavits, R., Palfi, V., Beres, A., Egyed, L., Denes, B., Somogyi, M. & Szathmary, S. (2012). Pathologic lesions caused by coinfection of Mycoplasma gallisepticum and H3N8 low pathogenic avian influenza virus in chickens. Veterinary Pathology, 49, 273–283.
  • Stipkovits, L. & Kempf, I. (1996). Mycoplasmoses in poultry. Revue Scientifique et Technique Office International des Epizooties, 15, 1495–1525.
  • Sulyok, K.M., Kreizinger, Z., Bekő, K., Forró, B., Marton, S., Bányai, K., Catania, S., Ellis, C., Bradbury, J., Olaogun, O.M., Kovács, A.B., Cserép, T., Gyuranecz, M. & Fenwick, B. (2019). Development of molecular methods for rapid differentiation of Mycoplasma gallisepticum vaccine strains from field isolates. Journal of Clinical Microbiology, 57, 1–12.
  • Ter Veen, C., de Wit, J.J. & Feberwee, A. (2020). Relative contribution of vertical, within-farm and between-farm transmission of Mycoplasma synoviae in layer pullet flocks. Avian Pathology, 49, 56–61.
  • Ter Veen, C., Dijkman, R., de Wit, J.J., Gyuranecz, M. & Feberwee, A. (2021). Decrease of Mycoplasma gallisepticum seroprevalence and introduction of new genotypes in Dutch commercial poultry during the years 2001–2018. Avian Pathology, 50, 52–60.
  • Thomas, L., Davidson, M. & McCluskey, R.T. (1966). Studies of PPLO infection. I. The production of cerebral polyarteritis by Mycoplasma gallisepticum in turkeys; the neurotoxic property of the Mycoplasma. Journal of Experimental Medicine, 123, 897–912.
  • Tripathy, S.B., Acharjyo, L.N., Singh, U., Ray, S.K. & Misra, S.K. (1972). Studies on an outbreak of Mycoplasma gallisepticum infection among peafowls (Pavo cristatus). British Veterinary Journal, 128, 428–431.
  • Truscott, R.B., Ferguson, A.E., Ruhnke, H.L., Pettit, J.R., Robertson, A. & Speckmann, G. (1974). An infection in chickens with a strain of Mycoplasma gallisepticum of low virulence. Canadian Journal of Comparative Medicine and Science, 38, 341–343.
  • Vardaman, T.H. (1976). The resistance and carrier status of meat-type hens exposed to Mycoplasma synoviae. Poultry Science, 55, 268–273.
  • Vardaman, T.H., Landreth, K., Whatley, S., Dreesen, L.J. & Glick, B. (1973). Resistance to Mycoplasma synoviae is bursal dependent. Infection and Immunity, 8, 674–676.
  • Vardaman, T.H., Reece, F.N. & Deaton, J.W. (1973). Effect of Mycoplasma synoviae on broiler performance. Poultry Science, 52, 1909–1912.
  • Wang, Y., Whithear, K.G. & Ghiocas, E. (1990). Isolation of Mycoplasma gallinarum and M. gallinaceum from the reproductive tract of hens. Australian Veterinary Journal, 67, 31–32.
  • Wang, Y., Yi, L., Zhang, F., Qiu, X., Tan, L., Yu, S., Cheng, X. & Ding, C. (2017). Identification of genes involved in Mycoplasma gallisepticum biofilm formation using mini-Tn4001-SGM transposon mutagenesis. Veterinary Microbiology, 198, 17–22.
  • Wannop, C.C., Butler, E.J. & Pearson, A.W. (1971). Experimental reproduction of Turkey Syndrome ’65 by infection with M. gallisepticum. Veterinary Record, 88, 30–33.
  • Wellehan, J.F., Calsamiglia, M., Ley, D.H., Zens, M.S., Amonsin, A. & Kapur, V. (2001). Mycoplasmosis in captive crows and robins from Minnesota. Journal of Wildlife Diseases, 37, 547–555.
  • Wellehan, J.F., Zens, M.S., Calsamiglia, M., Fusco, P.J., Amonsin, A. & Kapur, V. (2001). Diagnosis and treatment of conjunctivitis in house finches associated with mycoplasmosis in Minnesota. Journal of Wildlife Diseases, 37, 245–251.
  • Whithear, K.G., Soeripto, H.K.E. & Ghiocas, E. (1990). Immunogenicity of a temperature sensitive mutant Mycoplasma gaffisepticum vaccine. Australian Veterinary Journal, 67, 168–174.
  • Wyrzykowski, B., Albaric, O., Moreau, S., Nguyen, F., Fleurance, R., Belluco, S., Wyers, M. & Colle, M.A. (2013). Retrospective study of Mycoplasma gallisepticum meningoencephalitis in six turkey flocks in western France. Journal of Comparative Pathology, 148, 173–177.
  • Yamada, S. & Matsuo, K. (1983). Experimental infection of ducks with Mycoplasma synoviae. Avian Diseases, 27, 762–765.
  • Yoder, H.W., Jr. (1986). A historical account of the diagnosis and characterization of strains of Mycoplasma gallisepticum of low virulence. Avian Diseases, 30, 510–518.
  • Yoder, H.W., Jr. (1989). Nonspecific reactions to Mycoplasma serum plate antigens induced by inactivated poultry disease vaccines. Avian Diseases, 33, 60–68.
  • Yoder, H.W., Jr., Drury, L.N. & Hopkins, S.R. (1977). Influence of environment on airsacculitis: effects of relative humidity and air temperature on broilers infected with Mycoplasma synoviae and infectious bronchitis. Avian Diseases, 21, 195–208.
  • Yogev, D., Levisohn, S., Kleven, S.H., Halachmi, D. & Razin, S. (1988). Ribosomal RNA gene probes to detect intraspecies heterogeneity in Mycoplasma gallisepticum and M. synoviae. Avian Diseases, 32, 220–231.
  • Yogev, D., Levisohn, S. & Razin, S. (1989). Genetic and antigenic relatedness between Mycoplasma gallisepticum and Mycoplasma synoviae. Veterinary Microbiology, 19, 75–84.
  • Zander, D.V. (1961). Origin of S6 strain mycoplasma. Avian Diseases, 5, 154–156.
  • Zhang, G.Z., Zhang, R., Zhao, H.L., Wang, X.T., Zhang, S.P., Li, X.J., Qin, C.Z., Lv, C.M., Zhao, J.X. & Zhou, J.F. (2010). A safety assessment of a fowlpox-vectored Mycoplasma gallisepticum vaccine in chickens. Poultry Science, 89, 1301–1306.
  • Zhu, L., Konsak, B.M., Olaogun, O.M., Agnew-Crumptona, R., Kanci, A., Marenda, M.S., Browning, G.F. & Noormohammadi, A.H. (2017). Identification of a new genetic marker in Mycoplasma synoviae vaccine strain MS-H and development of a strategy using polymerase chain reaction and high-resolution melting curve analysis for differentiating MS-H from field strains. Veterinary Microbiology, 210, 49–55.
  • Zorman-Rojs, O., Zdovc, I., Benčina, D. & Mrzel, I. (2000). Infection of turkeys with Ornithobacterium rhinotracheale and Mycoplasma synoviae. Avian Diseases, 44, 1017–1022.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.