Skip to main content
Clinical Microbiology Reviews logoLink to Clinical Microbiology Reviews
. 2004 Oct;17(4):697–728. doi: 10.1128/CMR.17.4.697-728.2004

Mycoplasma pneumoniae and Its Role as a Human Pathogen

Ken B Waites 1,*, Deborah F Talkington 2
PMCID: PMC523564  PMID: 15489344

Abstract

Mycoplasma pneumoniae is a unique bacterium that does not always receive the attention it merits considering the number of illnesses it causes and the degree of morbidity associated with it in both children and adults. Serious infections requiring hospitalization, while rare, occur in both adults and children and may involve multiple organ systems. The severity of disease appears to be related to the degree to which the host immune response reacts to the infection. Extrapulmonary complications involving all of the major organ systems can occur in association with M. pneumoniae infection as a result of direct invasion and/or autoimmune response. The extrapulmonary manifestations are sometimes of greater severity and clinical importance than the primary respiratory infection. Evidence for this organism's contributory role in chronic lung conditions such as asthma is accumulating. Effective management of M. pneumoniae infections can usually be achieved with macrolides, tetracyclines, or fluoroquinolones. As more is learned about the pathogenesis and immune response elicited by M. pneumoniae, improvement in methods for diagnosis and prevention of disease due to this organism may occur.

INTRODUCTION

The first mycoplasma to be isolated in culture was the bovine pleuropneumonia agent now known as Mycoplasma mycoides subsp. mycoides, which was described initially by Nocard and Roux in 1898 (315). Over the next 50 years, evidence accumulated relating to the importance of the parasitic bacteria known at that time as pleuropneumonia-like organisms (PPLO) in various diseases of animals and their possible involvement in human disease. In the 1930s Klieneberger introduced the concept that mycoplasmas were “L-forms” of bacteria lacking cell walls and living symbiotically with other, walled bacteria (232). This theory started a spirited debate pitting those who believed that mycoplasmas were unique species against those who believed that mycoplasmas were wall-less variants of other known bacterial species and were undeserving of a unique taxonomic representation. This controversy was not completely settled until the 1960s, when guanine-plus-cytosine (G+C) content assays and DNA-DNA hybridization assays showed that mycoplasmas were indeed unique forms of life and lacked the ability to produce cell walls under any circumstances. A summary of the mycoplasma and ureaplasma species known to occur in humans, excluding occasional isolates or mycoplasmas of animal origin that sometimes infect humans, is shown in Table 1.

TABLE 1.

Mycoplasmas isolated from humansa

Organism Primary site of colonization
Role in diseaseb Reference(s)
Respiratory tract Genitourinary tract
Acholeplasma laidlawii + No 119
Mycoplasma buccale + No 154
Mycoplasma faucium + No 154
Mycoplasma fermentans + + Yes? 118
Mycoplasma hominis + Yes 118
Mycoplasma genitalium + Yes 423
Mycoplasma lipophilum + No 100
Mycoplasma orale + No 409
Mycoplasma pirum ? ? No 101
Mycoplasma penetrans + ? 270
Mycoplasma primatum + + No 99
Mycoplasma salivarium + No 118
Mycoplasma pneumoniae + Yes 68
Mycoplasma spermatophilum + No 186
Ureaplasma urealyticum + Yes 350, 376
Ureaplasma parvum + Yes 350, 376
a

The organisms shown in this table represent those species that have been shown to occur in humans, with humans as their primary host. This listing excludes occasional isolates and those known to be primarily of animal origin that have been recovered from humans in isolated instances, usually in association with immunocompromise.

b

In immunocompetent persons.

Dienes and Edsall detected the first mycoplasma isolated from humans in a Bartholin's gland abscess in 1937 (104). This mycoplasma was probably the organism we now know as Mycoplasma hominis. Other human mycoplasmas, including Mycoplasma fermentans. Mycoplasma salivarium, and T-strains, later known as ureaplasmas, had been described by the 1950s. The organism eventually known to be Mycoplasma pneumoniae was first isolated in tissue culture from the sputum of a patient with primary atypical pneumonia by Eaton et al. in 1944, and thereafter it became known as the Eaton agent (115). Tests on volunteers and field studies conducted in the 1950s and early 1960s provided solid evidence that the Eaton agent caused lower respiratory tract infections in humans (66, 69, 268), but it was considered to be a virus until it became clear that antibiotics could be effective against it. In 1961 Marmion and Goodburn postulated that the Eaton agent was a PPLO and not a virus (281). Chanock et al. succeeded in culturing the Eaton agent on cell-free medium (68) and proposed the taxonomic designation M. pneumoniae in 1963 (67).

The numerous commensal mycoplasmal species that commonly inhabit the human oropharynx, especially the most common species, Mycoplasma orale and M. salivarium, may occasionally cause diagnostic confusion with M. pneumoniae if they happen to find their way to the lower respiratory tract or if diagnostic specimens from the lower respiratory tract are contaminated with oropharyngeal secretions. However, with rare exceptions, usually involving immunosuppressed persons, the oropharyngeal commensal mycoplasmal species are not pathogenic.

Among human mycoplasmas, M. pneumoniae is by far the best known and most carefully studied. Much has been learned during the past several years about its cell biology, the host immune response that it elicits, laboratory techniques for detection, disease epidemiology, and its role as a respiratory tract pathogen. These developments are summarized in this review.

MOLLICUTE TAXONOMY AND CLASSIFICATION

The term “mycoplasma” (Greek; “mykes” = fungus and “plasma” = formed) emerged in the 1950s (117) and replaced the older PPLO terminology. The allusion to a fungus-like growth pattern in the name “mycoplasma” happens to describe only the growth of M. mycoides, but the term was nevertheless adopted and has persisted to this day. In the 1960s, mycoplasmas were designated members of a class named Mollicutes, which derives from Latin words meaning soft (“mollis”) and skin (“cutis”). The current taxonomic designations included in class Mollicutes comprise 4 orders, 5 families, 8 genera, and about 200 known species that have been detected in humans, vertebrate animals, arthropods, and plants. M. pneumoniae is a member of the family Mycoplasmataceae and order Mycoplasmatales (435).

Members of the class Mollicutes are characterized by their small genomes consisting of a single circular chromosome containing 0.58 to 2.2 Mbp, a low G+C content (23 to 40 mol%), and the permanent lack of a cell wall (213). The taxonomy of this class has been extensively revised based on 16S rRNA analysis and is discussed in greater detail elsewhere (213). Studies of 16S rRNA sequences suggest that mycoplasmas are most closely related to the gram-positive eubacterial subgroup that includes the bacilli, streptococci, and lactobacilli. According to Maniloff (278), the Mollicutes diverged from the Streptococcus branch of gram-positive bacteria with low G+C contents and relatively small bacterial genomes about 605 million years ago. Their small genomes are now believed to be the result of a gradual reduction in genome size from a common ancestor in a process known as degenerative evolution (278). The nature of the selective pressure that led to the evolution of Mollicutes is not precisely known. Figure 1 shows the mycoplasma phylogeny reconstructed from 16S rRNA sequence comparisons.

FIG. 1.

FIG. 1.

Phylogeny of mycoplasmas reconstructed from 16S rRNA sequence comparisons. Branch lengths are proportional to evolutionary distance (the number of base changes per 1,000 nucleotides). The scale at the bottom denotes the branch distance corresponding to five base changes per 100 nucleotides. Reprinted from reference 278 with permission.

CELL BIOLOGY

Mycoplasmas represent the smallest self-replicating organisms, in both cellular dimensions and genome size, that are capable of cell-free existence (444). Individual spindle-shaped cells of M. pneumoniae are 1 to 2 μm long and 0.1 to 0.2 μm wide, compared with a typical bacillus of 1 to 4 μm in length and 0.5 to 1.0 μm in width. Accordingly, the M. pneumoniae cell volume is less than 5% of that of a typical bacillus. The small size and volume of mycoplasmal cells allow them to pass through 0.45-μm-pore-size filters that are commonly used to filter sterilize media. The small cellular mass also means that mycoplasmas cannot be detected by light microscopy, and they do not produce visible turbidity in liquid growth media. Typical colonies of M. pneumoniae, shown in Fig. 2, rarely exceed 100 μm in diameter when cultivated on enriched medium such as SP4 agar and require examination under a stereomicroscope to visualize their morphological features.

FIG. 2.

FIG. 2.

Spherical colonies of M. pneumoniae growing on SP4 agar. Magnification, ×95.

The genome of M. pneumoniae was completely sequenced in 1996 and shown to consist of 816,394 bp with 687 genes (187). In contrast, the Escherichia coli genome comprises 4,600,000 bp and about 4,300 genes, making it more than five times larger than that of M. pneumoniae (356).

The small genome of M. pneumoniae and its limited biosynthetic capabilities are responsible for many of the biological characteristics and requirements for complex medium supplementation in order for the organism to be cultivated in vitro. Mollicutes have no ability to synthesize peptidoglycan cell walls, since the genes responsible for these processes are not present in the genome. The lack of a rigid cell wall confers pleomorphism on the cells and makes them unable to be classified as cocci or bacilli in the manner of conventional eubacteria. Mollicutes have never been found as freely living organisms in nature, since they depend on a host cell to supply them with the things they need for their parasitic existence. Another characteristic of most mollicutes and all members of the genus Mycoplasma is the requirement for sterols in artificial growth media, supplied by the addition of serum. Sterols are necessary components of the triple-layered mycoplasmal cell membrane that provide some structural support to the osmotically fragile mycoplasma.

Maintenance of osmotic stability is especially important in mollicutes due to the lack of a rigid cell wall. Although these organisms can flourish within an osmotically stable environment in their chosen eukaryotic host, they are extremely susceptible to desiccation, a fact that has a great impact on the need for proper handling of clinical specimens from which cultural isolation is to be attempted and the need for close contact for transmission of infection from person to person by airborne droplets. Another structural component of the M. pneumoniae cell that is important for extracellular survival is a protein network that provides a cytoskeleton to support the cell membrane.

One aspect of M. pneumoniae cell biology that is not widely appreciated is the fact that this organism, along with several other mollicute species, may elaborate capsular material external to the cell membrane. The first report indicating the presence of capsular material in M. pneumoniae appeared in 1976 in a review of its ultrastructure as determined by electron microscopy (444). It was suggested that this capsular material may have a role in adherence, but this has not been conclusively proven (444).

M. pneumoniae possesses very limited metabolic and biosynthetic activities for proteins, carbohydrates, and lipids in comparison to conventional bacteria. Like other mollicutes, it scavenges for nucleic acid precursors and apparently does not synthesize purines or pyrimidines de novo (91). For many years it has been known that fermentation of glucose to lactic acid by means of substrate phosphorylation effected by phosphoglyceric acid kinase and pyruvate kinase is a means by which M. pneumoniae generates ATP. Beyond that, there has been considerable speculation about which enzyme systems were actually present. Much of what is now known about the metabolic properties of M. pneumoniae has been made possible through annotation of its genome and direct identification of the genes encoding enzyme systems responsible for various metabolic pathways (91, 187). However, some interesting and perhaps unexpected findings have occurred since the sequence and annotation of the genome have been published. For example, there is genomic evidence for enzymes such as arginine deiminase, even though biochemical activity evidenced by ammonia production has not been directly observed in M. pneumoniae (328). Pollack et al. (327) have recently reviewed the central carbohydrate pathways of mollicutes and reported that M. pneumoniae possesses all 10 reactions of glycolysis but that the tricarboxylic acid cycle and a complete electron transport chain containing cytochromes are absent. Thus, the lactic acid end product of fermentation is still relatively reduced with electrons whose energy could be trapped if the pyruvate precursor could be diverted to the tricarboxylic acid cycle.

M. pneumoniae reduces tetrazolium either aerobically or anaerobically, and this has been one of several characteristics that have been used historically to identify the species and distinguish it from commensal mycoplasmas of the oropharynx. The availability of nucleic acid amplification techniques such as the PCR assay has made older methods of identification such as tetrazolium reduction and hemadsorption with guinea pig erythrocytes less important than they were previously. A pathway listing all of the relevant enzymes encoded in the M. pneumoniae genome is available at www.bork.embl-heidelberg.de/Annot/MP/ (91). Another unique property of Mycoplasma spp. is the use of the universal stop codon UGA as a codon for tryptophan (200).

M. pneumoniae, like other mollicutes, has developed specialized reproductive cycles as a result of its adaptation to existence with a limited genome and a parasitic life style that requires attachment to host cells (91). It reproduces by binary fission, temporally linked with duplication of its attachment organelle, which migrates to the opposite pole of the cell during replication and before nucleoid separation (15). A graphic model proposed for M. pneumoniae cell division, illustrating the formation and migration of the attachment organelles, is shown in Fig. 3. M. pneumoniae has been shown to bind and glide on glass and other solid surfaces, with the organism moving with the attachment organelle at the leading end. Neither genomic analysis nor electron microscopy of M. pneumoniae has demonstrated the presence of structures such as flagella or pili, suggesting that gliding motility occurs by an unknown mechanism involving the attachment organelle (290).

FIG. 3.

FIG. 3.

Proposed scheme for cell division and duplication of the terminal attachment structure in M. pneumoniae. Reprinted from reference 375 with permission.

PATHOGENESIS OF DISEASE

Mycoplasmas are primarily mucosal pathogens, living a parasitic existence in close association with epithelial cells of their host, usually in the respiratory or urogenital tracts. M. pneumoniae exclusively parasitizes humans, whereas some of the other human mycoplasmas have also been recovered from nonhuman primates. A comprehensive review of the molecular biology and pathogenicity of mycoplasmas that includes considerable information devoted to M. pneumoniae was published in 1998 by Razin et al. (345), and the reader is referred to that excellent summary for details at the cellular and subcellular levels of how mycoplasmas cause disease in their eukaryotic hosts.

Cytadherence

Evidence accumulated since the 1960s through animal models as well as in vitro cell and organ culture systems indicates that attachment of M. pneumoniae to the respiratory epithelium is a prerequisite for later events that culminate in production of disease (403). This close interaction between the mycoplasma and host cells protects it from removal by the host's mucociliary clearance mechanism and allows it to produce a variety of local cytotoxic effects.

Because M. pneumoniae is primarily an extracellular pathogen that depends on close association with host cells to survive, it has evolved a complex and specialized attachment organelle to facilitate its parasitic existence, as shown by electron microscopy (Fig. 4 and 5). This attachment organelle consists of a specialized tip structure with a central core of a dense rod-like central filament surrounded by a lucent space that is enveloped by an extension of the organism's cell membrane. The tip structure is actually a network of adhesins, interactive proteins, and adherence accessory proteins that cooperate structurally and functionally to mobilize and concentrate adhesins at the tip of the organism. The host cell ligand for mycoplasmal adhesins has not been characterized conclusively, although sialoglycoconjugates and sulfated glycolipids have been implicated (247, 347). Recent data (89) have shown that two proteins expressed on the M. pneumoniae cell surface, elongation factor TU and pyruvate dehydrogenase E1 β, are also involved in binding M. pneumoniae to fibronectin, a very common component of eukaryotic cell surfaces, basement membranes, and the extracellular matrix.

FIG. 4.

FIG. 4.

Scanning electron micrograph of M. pneumoniae cells. Whole mycoplasmas are shown, with terminal attachment structures indicated by arrowheads. Reprinted from reference 245 with permission.

FIG. 5.

FIG. 5.

Transmission electron micrograph of M. pneumoniae-infected hamster tracheal ring, demonstrating the close association of the attachment structure to the epithelium (arrow). (Copyright J. L. Jordan and D. C. Krause.)

The P1 adhesin is a 170-kDa protein concentrated in the attachment tip that is now known to be the major structure responsible for interaction of M. pneumoniae with host cells (27, 30, 31, 82, 88, 90, 196, 240, 241). In addition to its presence in the attachment organelle, lower concentrations of P1 are also widespread on the cell's surface (15, 375). Loss of P1 activity through spontaneous mutation or by trypsin treatment results in avirluence by reduced adherence of mycoplasmas to eukaryotic cells (26). Spontaneous reversion to the cytadhering phenotype is accompanied by the reappearance of the implicated proteins, restoration of structurally and functionally intact tips, and return of full infectivity. Further proof for the functional role of P1 as an adhesin comes from evidence that monoclonal antibodies against P1 block adherence in a hamster model of mycoplasma respiratory disease, whereas antibodies produced against some other M. pneumoniae proteins have no effect on attachment (242).

Jacobs (202) postulated that the immunodominant epitopes of the M. pneumoniae adhesins differ from the highly conserved adherence-mediating domains, thereby suggesting that one reason for the lack of protective immunity against reinfection even in the midst of a serological response could lie in the fact that antibodies directed against the variable domains are unlikely to generate effective cytadherence-blocking antibodies. Studies by Baseman and coworkers suggest that the P1 protein expression alone is not sufficient to mediate adherence of M. pneumoniae to certain host cells and cause disease and that the cooperative activity of other proteins is needed (29, 243, 244). P30 is one of several additional proteins that have been implicated in the adherence process, based on the knowledge that antibodies developed against P30 can block M. pneumoniae hemadsorption (29, 88, 298). Balish and Krause (15) recently suggested that P30 may be involved in gliding motility as well as coordination of cell division with biogenesis of the attachment organelle.

Other structures produced by M. pneumoniae that have been studied as mediators in cytadherence in M. pneumoniae include proteins HMW1, HMW2, HMW3, HMW4, HMW5, P90, and P65, which, in addition to P30, are believed to participate in the establishment of the polar structure. Once this polar structure is established, an independently assembled complex of proteins B, C, and P1 is drawn to the structure to complete formation of the functional terminal attachment organelle (15). A more in-depth discussion of mycoplasma interactions with host cells and the process of cytadherence at the subcellular level was published recently by Rottem (354).

Intracellular Localization

Mycoplasmas are known primarily as mucosal pathogens that reside extracellularly on epithelial surfaces. However, during the past few years, the potential for several mycoplasmal species to fuse with and enter host cells that are not normally phagocytic has been demonstrated (355). Such an occurrence should not be unexpected for microorganisms lacking a rigid cell wall that are typically closely associated with host cell surfaces. Rottem (355) has summarized current knowledge concerning the features that enable M. penetrans, a mycoplasma of uncertain pathological significance that has been isolated from urine specimens from human immunodeficiency virus-infected persons, and M. fermentans to invade host cells, some of which may be relevant to enhance understanding of similar events that may occur with M. pneumoniae. Dallo and Baseman (87) recently described the ability of M. pneumoniae to survive, synthesize DNA, and undergo cell replication in artificial cell culture systems over a 6-month period.

An intracellular existence that sequesters M. pneumoniae could facilitate the establishment of latent or chronic states, circumvent mycoplasmacidal immune mechanisms, facilitate its ability to cross mucosal barriers and gain access to internal tissues, and impair the efficacy of some drug therapies, accounting for difficulty in eradicating the mycoplasmas under clinical conditions (28, 355, 403, 420). Fusion of the mycoplasmal cell membrane with that of the host may also result in release of various hydrolytic enzymes produced by the mycoplasma as well as insertion of mycoplasmal membrane components into the host cell membrane, a process that could potentially alter receptor recognition sites and affect cytokine induction and expression (355). At present, the extent to which M. pneumoniae invades and replicates intracellularly in vivo is not known. Therefore, the clinical significance of these theoretical events associated with cell fusion remains to be proven.

Cytotoxicity and Inflammation

Internalization of a cell-associated mycoplasma into a host cell is not a prerequisite for the initiation of local cytotoxic events and clinical manifestation of disease, although cytadherence in the respiratory tract is the initiating event in disease production by M. pneumoniae. It is not known precisely how M. pneumoniae injures the respiratory epithelial cell after attachment, but a number of biochemical and immunological properties of the organism that are likely to be involved have been described. Close approximation of the organism to the host cells, facilitated by the adhesin proteins, appears to be important to facilitate localized tissue disruption and cytotoxicity. Unlike many human pathogens, M. pneumoniae is not known to produce any exotoxins. Hydrogen peroxide and superoxide radicals synthesized by M. pneumoniae act in concert with endogenous toxic oxygen molecules generated by host cells to induce oxidative stress in the respiratory epithelium (420). Consistent with the small genome, M. pneumoniae lacks some enzymes that are associated with virulence of other bacteria, such as superoxide dismutase and catalase. Hydrogen peroxide production in M. pneumoniae occurs as a result of a flavin-terminated electron transport chain (420). Hydrogen peroxide has been known to be important as a virulence factor in M. pneumoniae since Somerson et al. showed it to be the molecule that confers hemolytic activity (385). The ultrastructural effects of peroxide on host cells such as erythrocytes include loss of reduced glutathione, denaturation of hemoglobin, peroxidation of erythrocyte lipids, and eventually lysis of the cells. Almagor et al. (8) suggested that superoxide anion produced by M. pneumoniae acts to inhibit catalase in host cells, thereby reducing the enzymatic breakdown of peroxides produced endogenously and by the mycoplasma, rendering the host cell more susceptible to oxidative damage. M. pneumoniae hemadsorption and lysis of guinea pig erythrocytes, which are low in endogenous catalase, are also mediated by peroxide (420). This property was adapted for use as a diagnostic test to presumptively distinguish M. pneumoniae from other commensal mycoplasmas that are commonly found in the human respiratory tract, which do not produce hydrogen peroxide and therefore do not hemadsorb in this manner.

Host cell lactoferrin acquisition by M. pneumoniae is yet another possible means by which local injury may occur, through generation of highly reactive hydroxy radicals resulting from the introduction of iron complexes in a microenvironment rendered locally acidic by cellular metabolism that also includes hydrogen peroxide and superoxide anion (419).

Mammalian cells parasitized by M. pneumoniae exhibit a number of cytopathic effects that may occur as a result of the local damage mediated biochemically following cytadherence. M. pneumoniae infection leads to deterioration of cilia in the respiratory epithelium, both structurally and functionally. Cells may lose their cilia entirely, appear vacuolated, and show a reduction in oxygen consumption, glucose utilization, amino acid uptake, and macromolecular synthesis, ultimately resulting in exfoliation of all or parts of the infected cells (80, 83) These subcellular events can be translated into some of the clinical manifestations of respiratory tract infection that are associated with this organism, such as the persistent, hacking cough that is so commonly associated with M. pneumoniae.

Once M. pneumoniae reaches the lower respiratory tract, the organism may be opsonized by complement or antibodies. Macrophages become activated, begin phagocytosis, and undergo chemotactic migration to the site of infection. High percentages of neutrophils and lymphocytes are present in alveolar fluid. CD4+ T lymphocytes, B lymphocytes, and plasma cells infiltrate the lung (65, 318), manifested radiologically as pulmonary infiltrates. Further amplification of the immune response in association with lymphocyte proliferation, production of immunoglobulins, and release of tumor necrosis factor alpha (TNF-α), gamma interferon (IFN-γ), and various interleukins (including interleukin-1β [IL-1β], IL-2, IL-4, IL-5, IL-6, IL-8, IL-10, and IL-18) occurs, based on evidence from clinical and in vitro studies and from animal models (193, 195, 263, 308, 309, 406, 420, 447, 448). Many of these reactive substances will have elevated levels in both alveolar fluid and serum (233, 263, 406, 448). Yang et al. (448) recently reported that M. pneumoniae infection of human epithelial carcinoma cells in vitro resulted in increased levels of IL-8 and TNF-α mRNAs and that both proteins were secreted into culture medium. The major proinflammatory cytokine IL-1β mRNA also increased and the corresponding protein was synthesized, but its secretion was cell type specific due to an endogenous caspase-1 inhibitory component in the lung epithelial cells studied (447).

The role of cytokines and other reactive substances in the pathogenesis of M. pneumoniae lung disease has been a topic of considerable interest during the past several years, and a number of clinical studies involving humans as well as investigations based on animal models have been reported. Current evidence from human and animal studies suggests that cytokine production and lymphocyte activation may either minimize disease through the enhancement of host defense mechanisms or exacerbate disease through immunological lesion development. Thus, the more vigorous the cell-mediated immune response and cytokine stimulation, the more severe the clinical illness and pulmonary injury (78, 201, 305, 338, 342, 404-406). This concept of immune-mediated lung disease provides a basis for consideration of immunomodulatory therapeutics in addition to conventional antimicrobial therapies in management of disease due to M. pneumoniae.

Asthma and Other Chronic Lung Conditions

The release of proinflammatory cytokines in association with M. pneumoniae infection has also been implicated as a possible mechanism leading to or exacerbating underlying chronic pulmonary diseases such as bronchial asthma. The concept that chronic infection with M. pneumoniae might play a role in the pathogenesis of asthma was speculated on over 30 years ago (42). The relevant questions are whether M. pneumoniae is a primary cause of asthma or whether mycoplasmal infection is at least a cofactor in its development. Appreciation of the pathogenesis of chronic murine respiratory mycoplasmosis, a naturally occurring mycoplasmal infection of rodents mediated by Mycoplasma pulmonis that is slowly progressive, is greatly influenced by heredity, and has characteristics that are similar in some ways to those of asthma in humans, gives further credence to the potential for mycoplasmas to cause longstanding lung disorders such as asthma (60).

Multiple lines of evidence suggest why M. pneumoniae may play a role in the pathogenesis of asthma beyond simple, acute exacerbation. M. pneumoniae can be detected by PCR and/or culture more often from the airways of patients with chronic, stable asthma than from matched control patients. Kraft et al. (238) detected M. pneumoniae by PCR in respiratory secretions of 10 of 18 stable adult asthmatics (56%) and in only 1 of 11 healthy controls. In another study, throat cultures for M. pneumoniae were positive in 24.7% of children and adults with asthma exacerbation, compared with 5.7% of healthy controls (158). However, other studies of children with acute asthma exacerbation showed that while rhinoviruses and respiratory syncytial virus may be detected frequently, M. pneumoniae played a minor role and was detected in just a few patients (48, 155, 414). Limitations of some of these studies were the use of complement fixation tests alone to identify patients with M. pneumoniae (414) and inclusion of very young children in whom viral bronchiolitis instead of asthma may have been present (155).

Treatment of asthma patients in whom M. pneumoniae has been detected with macrolide antimicrobials resulted in improvement in pulmonary function tests in comparison with asthma patients who did not have evidence of M. pneumoniae in airways (239), owing perhaps to both the antibacterial and the anti-inflammatory effects of macrolides. Macrolides are known to reduce airway hyperresponsiveness in asthmatic patients, attenuate pulmonary inflammation by protecting ciliated epithelium against oxidative damage, stabilize cell membranes, and decrease sputum purulence (133). Mycoplasmas have been detected by PCR in airways even when cultures and serological results are negative, suggesting that low numbers of organisms may evade detection by the immune system (239). The lack of a measurable serological response may also facilitate the organism's persistence in the lower respiratory tract.

Lung abnormalities, including reduced pulmonary clearance and airway hyperresponsiveness, may persist for weeks to months after an infection with M. pneumoniae (227, 279, 295, 359, 377). Marc et al. (279) reported abnormalities in pulmonary function tests in up to 50% of children, and Kim et al. (227) described abnormal computerized axial tomography studies for 37% of children months to years after an episode of M. pneumoniae respiratory tract infection, thus establishing the ability of mycoplasmas to induce chronic and possibly permanent lung damage long after resolution of respiratory tract symptoms.

In animal models of M. pneumoniae infection, Hardy et al. (178) demonstrated that an initial pneumonia lasted 3 to 4 weeks, similar to the case for human disease, characterized by histological lung inflammation and elevated cytokine and chemokine levels. At 530 days following inoculation of M. pneumoniae into the respiratory tract, 78% of mice demonstrated peribronchial and perivascular mononuclear infiltrates that were significantly more pronounced than those in controls by a histopathological severity score, concomitantly with increased airway reactivity and obstruction. Such information provides further evidence for the potential for this organism to produce chronic lung disease of clinical significance.

M. pneumoniae is known to induce a number of the inflammatory mediators implicated in the pathogenesis of asthma that may play a role in exacerbations, which often include wheezing (74, 126, 373, 377). Esposito et al. (126) studied 225 children with an acute episode of wheezing, 16 of whom had mycoplasmal infection as determined by serology and/or PCR on nasopharyngeal secretions, and compared them to 8 asymptomatic children with M. pneumoniae and 8 uninfected controls. Children with wheezing and acute M. pneumoniae infection had a statistically significant increase in IL-5 compared to children with M. pneumoniae who were asymptomatic and to the controls without wheezing. Those authors therefore proposed that M. pneumoniae might trigger the wheezing process by means of IL-5 secretion in persons who are genetically predisposed or are otherwise susceptible. This seems plausible since IL-5 is the cytokine that has been shown to be essential for development of airway hyperresponsiveness in association with infection caused by respiratory syncytial virus (370, 371).

Hardy et al. (179) provided further evidence that the presence of M. pneumoniae in the lower respiratory tract stimulates production of a wide array of inflammatory mediators, including TNF-α, IFN-γ, IL-6, and IL-8, using a murine model of infection. By plethysmography, intranasal inoculation of live M. pneumoniae into mice triggered greater pulmonary airflow resistance or obstruction for a longer duration than what was observed in animals that were inoculated with dead organisms or SP4 broth alone. Significant airflow resistance persisted for the entire 28-day observation period, even after histological evidence of pulmonary inflammation subsided.

M. pneumoniae can be associated with significantly greater numbers of mast cells in patients with chronic asthma, according to one study (284), and experimental evidence from a rodent mast cell line suggests that the organism can induce activation of mast cells with release of serotonin and β-hexosaminidase (193). Elevated serum immunoglobulin E (IgE) levels as well as production of IgE specific to M. pneumoniae or common allergens may also occur during mycoplasmal infection in children with onset of asthma. Koh et al. (233) showed that levels of the cytokine IL-4 and the ratio of IL-4 to IFN-γ were significantly higher in children with M. pneumoniae than in those with pneumococcal pneumonia or uninfected controls, suggesting that a TH2-like cytokine response represents a favorable condition for IgE production.

There are a relatively small number of very limited studies that have implicated M. pneumoniae in other chronic lung conditions. Respiratory tract infections with bacteria such as Streptococcus pneumoniae, Haemophilus influenzae, and Moraxella catarrhalis have been associated with acute exacerbations of chronic obstructive pulmonary disease (COPD) for many years. Over 20 years ago, Gump et al. (176) and later Buscho et al. (54) and Smith et al. (383) made some of the earliest observations that mycoplasmal infections could be associated with some cases of COPD exacerbation. However, little attention was given to this possibility for several years. Since the late 1990s, some investigations have been undertaken to assess the possible contribution of M. pneumoniae and Chlamydophila pneumoniae in COPD (260, 261, 293). Mogulkoc et al. (293) used serology to assess the presence of M. pneumoniae and C. pneumoniae in 49 ambulatory patients with acute purulent exacerbations of COPD. They found evidence of acute C. pneumoniae in 11 patients (22%), sometimes in association with other bacteria. M. pneumoniae infection was detected in only three patients (6%). Lieberman et al. (260) also used serology to assess the presence of M. pneumoniae, evaluating a group of 219 patients hospitalized with acute exacerbations of COPD. A total of 34 patients (14.2%) had serological evidence of acute M. pneumoniae infection, making it the third most common bacterial pathogen detected. More than one agent was detected in one-third of the cases. The findings of this study were particularly significant in that they showed that respiratory viruses and atypical bacteria, mainly Legionella species and M. pneumoniae, were involved in most cases and that the classical bacteria were responsible for a minority of cases. These same investigators (261) further described 34 hospitalizations of COPD patients with evidence of M. pneumoniae and showed that 3 had pneumonia, 3 required intensive care management, and 1 died. Since the majority of these patients had serological evidence of an additional pathogen and since the study relied entirely upon serological measurements, it is impossible to determine the precise contribution of M. pneumoniae to these clinical conditions. Specific short-term treatment with drugs known to be active against mycoplasmas did not appear to be beneficial in reducing the duration of hospitalization, but this is not too surprising in view of the chronicity of many mycoplasmal infections and the difficulty in their eradication in many instances.

The occurrence of bacterial infections of the respiratory tract is considered the main cause of progressive pulmonary failure in patients with cystic fibrosis (124). Although the role of M. pneumoniae in community acquired infections of the lower respiratory tract is well known, very little information is available about its occurrence and pathogenic significance in patients with cystic fibrosis. Petersen et al. (324) detected M. pneumoniae antibody by complement fixation (CF) in only 2 of 332 episodes of acute exacerbations in patients with cystic fibrosis. Subsequently, Efthimiou et al. (121) noted a fourfold rise in antibody titers against M. pneumoniae. Coxiella burnetii, and various viruses in a small number of young adults with cystic fibrosis who experienced deterioration in lung function and increase in lower respiratory tract symptoms. Ong et al. (317) and Pribble et al. (335) detected antibodies against M. pneumoniae in 1 of 19 and in 4 of 80 acute pulmonary exacerbations, respectively. Although these studies were limited in the respect that serology was the sole means of assessing the presence of mycoplasmas and the test methods employed present some difficulty in proper interpretation to define a recent infection, taken together they suggest that mycoplasmas may occur but are fairly uncommon causes of these complications in persons with cystic fibrosis. Emre et al. (124) confirmed this assumption in that they were unable to demonstrate the presence of M. pneumoniae by PCR of oropharyngeal secretions in 16 patients, and only 1 of 16 showed serological evidence of recent infection. C. pneumoniae was detected by nasopharyngeal culture in 4 of 32 cases (12.5%), and three of these four patients had elevated IgM or IgG antibodies that were suggestive of acute infection. Clearly, more work must be done to clarify the importance of M. pneumoniae and other atypical bacteria in the epidemiology and pathogenesis of exacerbations of chronic lung diseases, using a more comprehensive diagnostic strategy that would include direct tests for the presence of the organism by PCR so that very low numbers of organisms might be detected in the airways, culture, and serology.

Immune Response and Immunomodulatory Effects

M. pneumoniae possesses both protein and glycolipid antigens that elicit antibody responses in infected individuals. The P1 protein is the target of many of the antibodies that are produced by the host in response to the M. pneumoniae infection, and it has also served as a target for development of serological assays. Following an initial infection, the normal immune system responds by rapidly producing antibodies that peak after 3 to 6 weeks, followed by a gradual decline over months to years. As a result of the long incubation period, an antibody response is often evident by the time symptoms appear. Elevation of M. pneumoniae-specific IgM alone can often be interpreted as evidence of acute infection, since this antibody typically appears within 1 week of the initial infection and approximately 2 weeks before IgG antibody (286, 380). However, the presence of IgM is considered most significant in pediatric populations, where there have been fewer opportunities for repeated exposures. Adults who have been infected repeatedly over a period of years may not respond to mycoplasma antigens with a brisk IgM response (434). In these cases, reinfection leads directly to an IgG response; therefore, the absence of a positive IgM test does rule out an acute infection. When it does occur, the IgM response may persist for months or years following infection (445), and in these cases a positive IgM test result may not reflect a current or recent infection.

IgA, while often overlooked as a diagnostic antibody class, may actually be a better indicator of recent infections in all age groups (380). IgA antibodies are produced early in the course of disease, rise quickly to peak levels, and decrease earlier than IgM or IgG (166, 440). Research into IgA responses in adult and pediatric populations and assessment of antibody levels in other body fluids such as urine are warranted. The importance of an intact immune response in protection against mycoplasmal disease is apparent in view of prolonged disease and dissemination in persons with hypogammaglobulinemia (410).

In addition to M. pneumoniae-specific antibodies, a variety of cross-reactive antibodies may develop in association with M. pneumoniae infection. The extensive sequence homology of the M. pneumoniae adhesin proteins and glycolipids of the cell membrane with mammalian tissues is a well-known example of molecular mimicry that may trigger autoimmune disorders that involve multiple organ systems through formation of antibodies against substances such as myosin, keratin, fibrinogen, brain, liver, kidney, smooth muscle, and lung tissues (16). Mycoplasmal adhesins also exhibit amino acid sequence homologies with human CD4 and class II major histocompatibility complex lymphocyte proteins, which could generate autoreactive antibodies and trigger cell killing and immunosuppression (353). Cold agglutinins and their historical use as a crude diagnostic test for M. pneumoniae infection are discussed in a subsequent section. Circulating immune complexes also occur during acute phases of M. pneumoniae diseases (80, 291).

Specific T-cell-mediated immunity is also involved in the host reaction to infection by M. pneumoniae. Lymphocytes from persons known to have had a prior mycoplasmal infection will undergo blast transformation when cultured in the presence of M. pneumoniae (134). Leukocytes from individuals with M. pneumoniae infections will show evidence of chemotaxis in the presence of the organism, and these individuals will respond with IFN-α in their blood (285, 302).

A property of many species of mycoplasmas that affects the immune responsiveness of the host is their propensity for mitogenic stimulation of B and T lymphocytes, thereby inducing autoimmune disease through the activation of anti-self T cells or polyclonal B lymphocytes (403). This property is associated with the ability of M. pneumoniae to stimulate production of cytokines in the initiation of the acute inflammatory response as described above.

Antigenic Variation

Many mycoplasmal species that infect animals or humans are known for their ability to induce chronic disease states in which clearing of the organism is extremely difficult. Therefore, these organisms must have evolved means by which they can successfully evade or modulate the host immune response. As mentioned above, intracellular localization and immunomodulatory activities are possible means to this end. Another mechanism that has been extensively studied in many other bacteria is variation in surface antigens. High-frequency phase and antigenic variation of surface adhesin proteins made possible by DNA rearrangements in truncated and sequence-related copies of the P1 adhesin genes that are dispersed throughout the genome has been described for M. pneumoniae (396, 397). Recombinational events among the repetitive elements themselves and with regions of the three-gene P1 adhesin operon promote diversity and altered specificities and affinities and maximize the coding potential of the limited mycoplasma genome (31, 451).

EPIDEMIOLOGY

Geographic Prevalence and Seasonality of Disease

Soon after the identification of M. pneumoniae as the etiological agent of primary atypical pneumonia in the early 1960s, considerable interest arose in elucidation and characterization of its incidence, prevalence, mode of spread, and spectrum of disease. M. pneumoniae infections can involve both the upper and the lower respiratory tracts and occur both endemically and epidemically worldwide in children and adults. Climate and geography are not thought to be of major significance. Even though most available data concerning the occurrence of M. pneumoniae infections have come from studies performed in the United States, Europe, and Japan, seroprevalence investigations using CF technology in arctic and tropical zones have also indicated the presence of M. pneumoniae antibody, suggesting that populations in these regions have had infections due to this organism (164, 214, 398). In the United States, there is no national surveillance system tracking M. pneumoniae infections; therefore, much of what we know about rates of endemic disease comes from population-based studies relying primarily on serological measurements (146, 148, 150, 171, 296, 297). Foy (143) used CF antibody determinations and culture to show that M. pneumoniae was responsible for 15 to 20% of all cases of community-acquired pneumonia, or two cases per 1,000 persons on an annual basis between 1962 and 1975, in Seattle, Wash. Additional retrospective serological studies performed in Denmark showed a pattern of M. pneumoniae infections over a 50-year period from 1946 through 1995 with endemic disease transmission punctuated with cyclic epidemics every 3 to 5 years, similar to what was observed in the United States (143, 267). Additional studies in North America and Europe performed over the past 3 decades have also reported similar trends (106, 143, 144, 146, 181, 207, 252, 266, 267, 313, 340). Although the incidence of disease does not vary greatly by season, the proportion of patients with pneumonia due to M. pneumoniae is greatest during the summer in temperate climates due to the lower incidence of other respiratory pathogens at this time (5, 283, 334). Outbreaks of M. pneumoniae infections also tend to occur in the summer or early fall (5, 131, 403). The long incubation period and relatively low transmission rate have been implicated in the prolonged duration of epidemics of M. pneumoniae infections (137).

Layani-Milon et al. (252) used data obtained by PCR assays on nasal swabs to define the incidence of M. pneumoniae and several respiratory viruses among persons presenting with evidence of acute respiratory tract infection in a region of France over a 5-year period. The distributions of organisms varied from year to year, with M. pneumoniae ranking second to influenza A virus as the most frequent pathogen encountered during the surveillance period. The rates of M. pneumoniae infections varied from year to year, unrelated to rates of other pathogens, suggesting cycles of epidemics as have been described in earlier studies. In some instances, both viral and mycoplasmal organisms were detected simultaneously in the same specimens.

Some recent changes in the incidence of M. pneumoniae infections described by Lind et al. (267), in which high numbers of cases occurred between epidemics without a return to lower endemic levels, have led to speculation as to the reasons for this occurrence. Jacobs (202) suggested that the availability of new information on virulence factors and the P1 adhesin along with the deciphering of the complete genome might enhance understanding of why these changes have occurred. Dorigo-Zetsma et al. (107) genotyped M. pneumoniae clinical isolates and grouped them into eight subtypes within two genomic groups based on P1 adhesin subtypes. Studies from Germany (207) and Japan (365) have shown that different P1 adhesins subtypes may be operating in the development and cycling times of M. pneumoniae epidemics. Such gene divergences within the P1 adhesin and development of subtype-specific antibodies following initial infection might account for the frequency of reinfections, which may be due to another subtype (202). Further work by Cousin-Allery et al. (86) and Dumke et al. (114), using techniques including restriction fragment length polymorphism, rapid amplified polymorphic DNA analysis, multilocus sequence typing, Western blotting, and two-dimensional gel electrophoresis to characterize over 200 M. pneumoniae isolates collected over several years from several European countries and the United States, showed that M. pneumoniae is a rather uniform microorganism, such that most of the isolates could be classified into two groups or subtypes based on the sequences of the P1 adhesin gene, the ORF6 gene, and the P65 gene and by a typical DNA restriction fragment pattern. Both studies found that one or the other of the two subgroups tended to predominate in specific geographical regions to some extent and that there were changes over time with respect to which subgroups most of the isolates belonged to. Dumke et al. (114) found four variants, which were identical to one another but did not belong to either subtype. These studies support the earlier findings (207, 365) that suggested that different M. pneumoniae subtypes might be operative in the cycling times of epidemics. Ovyn et al. (320) described the use of nucleic acid sequence-based amplification to classify 24 M. pneumoniae isolates into two types, yielding results in general agreement with those obtained by the other techniques.

Historically, M. pneumoniae has not been considered part of the normal flora of humans, and its detection by culture could usually be considered abnormal and of etiological significance if in a person with a clinical condition known to be caused by the organism. However, it can persist for variable periods in the respiratory tract following infections that have resolved clinically with appropriate antimicrobial therapy (143). The usual explanation for such persistence has been that the organism attaches strongly to and invades epithelial cells and that macrolide or tetracycline antibiotics commonly used for treating mycoplasmal infections are bacteriostatic and unable to kill all of the organisms. Surveillance studies using culture and/or PCR indicate that a prolonged asymptomatic carrier state may occur in some persons, providing a reservoir for spread of the organism to others (109, 143, 162, 254). Gnarpe et al. (162) demonstrated that 13.5% of 758 healthy volunteers harbored the organism. During a subsequent period of 11 months, the incidence of M. pneumoniae decreased to 4.6% of 499 volunteers, indicating the fluctuating occurrence of this organism over time.

Powerful molecular techniques such as PCR have extremely high sensitivity, theoretically being able to detect a single organism or a single copy of the targeted gene when purified DNA is used; this greatly exceeds the detection threshold of culture, which is approximately 100 to 1,000 cells under optimum conditions (53, 211, 343, 434). Since the mid-1990s, the widespread use of PCR for studies of M. pneumoniae has greatly enhanced, but also complicated, our understanding of its epidemiology, necessitating a reconsideration of the meaning of a “gold standard” for M. pneumoniae diagnosis.

Disease Transmission

M. pneumoniae can be transmitted through aerosols from person to person, and disease has been produced experimentally by aerosol inoculation (80). Persons with active mycoplasmal infection will carry the organisms in the nose, throat, trachea, and sputum, indicating diffuse involvement. Spread of organisms is greatly facilitated by the ubiquitous cough. Since the organisms tend to be associated with desquamated cells, relatively large droplets may be required for transmission, as evidenced by the close personal contact typical of outbreak settings, e.g., schools, military barracks, and institutions. M. pneumoniae infections commonly spread gradually among family members within a household (109, 137, 403). In view of the intimate contact needed for droplet transmission and the slow (6-h) generation time of M. pneumoniae, 1 to 3 weeks of incubation for each case is typical, and several cycles may be necessary before intrafamily transmission is complete. Some studies have reported incubation periods from common-source outbreaks of as short as 4 days (363), whereas others (145) have reported longer incubations, with a median of 23 days with intrafamilial spread, where smaller inocula may be involved and transmission may be less effective until the index case has exhibited symptoms for several days. Foy et al. (145) reported that 39% of family contacts may eventually become infected with M. pneumoniae, many asymptomatically. Dorigo-Zetsma et al. (109) found that among 79 asymptomatic household contacts of 30 index cases with acute respiratory tract infection due to M. pneumoniae, 15% harbored the organism, with a significantly greater number being children under 15 years of age. A novel mathematical model for investigating the control and spread of disease transmission has recently been applied to assess the effects of interventions in outbreaks of M. pneumoniae in closed communities (288).

Disease Outbreaks

Numerous outbreaks of M. pneumoniae infections have been documented in the community or in closed or semiclosed settings such as military bases (120, 131, 167, 168, 292), hospitals (141, 221, 229), religious communities (253, 299), and facilities for the mentally or developmentally disabled (197, 228, 391). In outbreaks where many more persons, usually living close together in military barracks or similar situations, are exposed to M. pneumoniae aerosols simultaneously, the rate of spread within a facility appears to be higher than in single-family households.

Attack rates of M. pneumoniae among military recruits and other closed or semiclosed populations can be quite high, with reports ranging from 25 to 71% in some settings (5, 120, 131, 228). Some studies have shown M. pneumoniae to be the leading cause of bacterial pneumonia among hospitalized and nonhospitalized military personnel (168, 170). Although long-term morbidity is uncommon, these outbreaks can be very disruptive and can consume significant resources. Strategies to control these outbreaks have included cohorting and use of antibiotics for symptomatic persons and for prophylaxis.

Demographics and Spectrum of Disease

Serological studies performed in the 1960s and 1970s, evaluating the attack rates of M. pneumoniae according to sex and broken down into various age groups, have yielded mixed results, with slight gender differences apparent between some age groups. Overall, there appears to be little reason to suspect that males and females have greatly differing susceptibilities to M. pneumoniae infections (145, 280, 314).

M. pneumoniae causes up to 40% or more of cases of community-acquired pneumonias and as many as 18% of cases requiring hospitalization in children (5, 47, 137, 156, 180, 182, 199, 273, 357, 403, 446). Older studies relying upon serology and culture reported M. pneumoniae pneumonia to be somewhat uncommon in children aged less than 5 years and greatest among school-aged children 5 to 15 years of age, with a decline after adolescence and on into adulthood (5, 143-145). However, M. pneumoniae may occur endemically and occasionally epidemically in older persons, as well as in children under 5 years of age (47, 143, 180, 199, 446). Detection of the organism in 23% of community-acquired pneumonias in children 3 to 4 years of age in a study performed in the United States during the 1990s (47) and documentation of its occurrence in children less than 4 years of age in France (252), without significant differences in infection rates for other children or adults, may reflect the greater number of young children who attend day care centers on a regular basis than in previous years and the ease with which young children share respiratory secretions with older household members or contacts. These recent findings may also reflect improved detection abilities that were unavailable in the 1960s and 1970s, when the first descriptions of M. pneumoniae epidemiology and age distribution were published. Although M. pneumoniae is generally not considered to be a neonatal pathogen, Ursi et al. (426) described probable transplacental transmission of M. pneumoniae, documented by PCR, in the nasopharyngeal aspirate of a neonate with congenital pneumonia.

Whereas pneumonia may be the most severe type of M. pneumoniae infection, the most typical syndrome, especially in children, is tracheobronchitis, often accompanied by a variety of upper respiratory tract manifestations. Esposito et al. (125) demonstrated acute M. pneumoniae infection in 44 of 184 children with nonstreptococcal pharyngitis (23%), using the criteria of an elevated IgM antibody titer or a fourfold increase in IgG antibody titer and/or a positive PCR assay on the nasopharyngeal aspirate. Since the testing for M. pneumoniae was performed only on specimens that were negative for Streptococcus pyogenes, it is possible that an even greater proportion of infections due to M. pneumoniae might have been detected, since some cases may be mixed. No other clinical manifestations or laboratory analyses other than a history of recurrent episodes of pharyngitis were useful in predicting the presence of M. pneumoniae.

M. pneumoniae infection is ordinarily mild, and many adult cases may be asymptomatic, whereas this is much less common in children, perhaps reflecting some degree of protective immunity for reinfections. Moreover, subsequent infections may be more common following initial mild infections as opposed to infection in which pneumonia develops, perhaps due to lesser stimulation of the immune response (147).

Although most mycoplasma infections occur among outpatients (hence the colloquial term “walking pneumonia”), M. pneumoniae is a significant cause of bacterial pneumonia in adults requiring hospitalization in the United States. Marston et al. (283) reported that M. pneumoniae was definitely responsible for 5.4% and possibly responsible for 32.5% of 2,776 cases of community-acquired pneumonia in hospitalized adults in a two-county region of Ohio, using CF antibody determinations for detection. Extrapolation of these data nationally provides an estimated 18,700 to 108,000 cases of pneumonia in hospitalized adults due to M. pneumoniae annually in the United States. Since the majority of patients with pneumonia in the United States are treated as outpatients, the total number of pneumonias due to M. pneumoniae is almost certainly many times greater, and as many as half of all infections in adults may even be asymptomatic. An additional striking finding of the study by Marston et al. (283) was their observation that the incidence of pneumonias due to M. pneumoniae in hospitalized adults increased with age, and it was second only to S. pneumoniae in elderly persons (Fig. 6).

FIG. 6.

FIG. 6.

Data from an active surveillance study performed in Ohio in 1991, showing age-specific rates of community-acquired pneumonia due to the major bacterial pathogens. M. pneumoniae infections were diagnosed by seroconversion, using CF tests. These data demonstrate that M. pneumoniae causes a relatively large proportion of pneumonias of sufficient severity to warrant hospitalization among persons younger than 50 years but that it is also an important cause of pneumonia in older age groups. Sp, S. pneumoniae; Mp, M. pneumoniae; Lp, L. pneumophila; Cp, C. pneumoniae. Reprinted from reference 283 with permission of the publisher.

Another study of hospitalized adults with community-acquired pneumonias performed in Israel (334), which used commercial serological kits to detect antibodies, showed M. pneumoniae to be second only to S. pneumoniae, and it was responsible for 29.2% of pneumonias overall.

CLINICAL SYNDROMES

Respiratory Tract Infections

The clinical entity of pneumonia eventually proven to be caused by M. pneumoniae was recognized many years before the actual identity and nature of the etiological agent were established. The first clues to differentiate pneumonia eventually proven to be due to mycoplasma from classical pneumococcal pneumonia came from the observations that some cases failed to respond to treatment with sulfonamides or penicillin. The lack of response to antimicrobial therapy was deemed “atypical,” and the condition was thought likely to be a primary form of lung disease of uncertain etiology; hence, the term “primary atypical pneumonia” was coined. This term, along with “walking pneumonia,” has been used widely by physicians and the lay public to denote mycoplasmal respiratory disease.

M. pneumoniae infections may be manifested in the upper respiratory tract, the lower respiratory tract, or both. The frequency of nonspecific upper respiratory tract infection manifestations has varied among numerous studies published since the mid-1960s, with some reports indicating that as many as 50% of patients with M. pneumoniae infection present with upper respiratory tract illness (132). Symptomatic disease typically develops gradually over a period of several days, often persisting for weeks to months. The most common manifestations include sore throat, hoarseness, fever, cough which is initially nonproductive but later may yield small to moderate amounts of nonbloody sputum, headache, chills, coryza, myalgias, earache, and general malaise (80, 137, 273, 392, 403). Dyspnea may be evident in more severe cases, and the cough may take on a pertussis-like character, causing patients to complain of chest soreness from protracted coughing (80). Inflammation of the throat may be present, especially in children, with or without cervical adenopathy, and conjunctivitis and myringitis sometimes occur (7, 125, 152). Children under 5 years of age are most likely to manifest coryza and wheezing, and progression to pneumonia is relatively uncommon, whereas older children aged 5 to 15 years are more likely to develop bronchopneumonia, involving one or more lobes, sometimes requiring hospitalization (137, 273, 392). Mild infections and asymptomatic conditions are particularly common in adults, and bronchopneumonia involving one or more lobes develops in 3 to 10% of infected persons (61). As mentioned above, M. pneumoniae is an important cause of pneumonia sufficiently severe to require hospitalization, especially in elderly persons (282, 283, 334). Several studies from the 1960s and 1970s indicate that M. pneumoniae may cause up to 5% of cases of bronchiolitis in young children (102, 112, 161, 271).

Chest auscultation may show scattered or localized rhonchi and expiratory wheezes. Since the alveoli are usually spared, rales and frank consolidation are fairly uncommon unless atelectasis is widespread. In uncomplicated cases, the acute febrile period lasts about a week, while the cough and lassitude may persist for 2 weeks or even longer. The duration of symptoms and signs will generally be shorter if antimicrobial treatment is initiated early in the course of illness (80).

It is important for clinicians to understand that the clinical presentation of M. pneumoniae respiratory disease is often similar to what is also seen with other atypical pathogens, particularly C. pneumoniae, various respiratory viruses, and bacteria such as S. pneumoniae. M. pneumoniae may also be present in the respiratory tract concomitantly with other pathogens (47, 109, 137, 180, 182, 252, 446), and there is some evidence from humans and animal models indicating that infection with M. pneumoniae may precede and somehow intensify subsequent infections with various respiratory viruses and bacteria, including S. pyogenes and Neisseria meningitidis (78). Potential explanations for such a synergistic effect include immunosuppression or alteration in respiratory tract flora due to the presence of M. pneumoniae (78, 255, 269, 358). Children with functional asplenia and immune system impairment due to sickle cell disease, other conditions such as Down syndrome, and various immunosuppressive states are at risk of developing more fulminant pneumonia due to M. pneumoniae (38, 137, 151, 192, 273, 378, 403).

Children with hypogammaglobulinemia are also known to be at greater risk for development of respiratory and joint infections due to M. pneumoniae, demonstrating the importance of functional humoral immunity in protection against infections due to this organism (137, 351, 403, 410). Roifman et al. (351) reported that 18 of 23 patients with hypogammaglobulinemia had one or more episodes of acute respiratory illness during which Ureaplasma urealyticum. M. orale, or M. pneumoniae was isolated from sputum. Resolution occurred followed institution of specific antibiotic therapy and elimination of the mycoplasmas. M. pneumoniae was isolated from the joint of a patient with arthritis and from six patients with chronic lung disease. Clinical improvement, albeit transient, coincided with negative mycoplasma culture results. There are a few case reports of M. pneumoniae infections in pediatric AIDS patients (49, 210), but is not known whether the incidence or severity of pulmonary or extrapulmonary M. pneumoniae infections in AIDS patients is increased significantly or how any immunosuppressed state specifically affects host resistance to M. pneumoniae infection. Fulminant infections with multiple organ involvement and deaths due to M. pneumoniae, usually in otherwise healthy adults and children, have been reported but are uncommon (73, 93, 103, 149, 198, 364, 372, 402, 403, 416, 442).

Extrapulmonary Manifestations

As many as 25% of persons infected with M. pneumoniae may experience extrapulmonary complications at variable time periods after onset of or even in the absence of respiratory illness. Autoimmune reactions have been suggested to be responsible for many of the extrapulmonary complications associated with mycoplasmal infection (403). However, the availability of PCR has greatly enhanced understanding of how M. pneumoniae can disseminate throughout the body. The presence of M. pneumoniae in extrapulmonary sites such as blood, synovial fluid and cerebrospinal fluid, pericardial fluid, and skin lesions has been documented by PCR as well as culture, so direct invasion must always be considered (23, 220, 235, 306, 360). However, the frequency of direct invasion of these sites is unknown because the organism is rarely sought for clinical purposes. It is also important to realize that extrapulmonary complications can be seen before, during, or after pulmonary manifestations or can occur in the complete absence of any respiratory symptoms (62).

Central nervous system (CNS) complications are recognized as among the most common of extrapulmonary manifestations of M. pneumoniae infection (384) and have been known to occur since the first report appeared in 1943, even before the true identity of the causative organism was known (56). Approximately 6 to 7% of hospitalized patients with serologically confirmed cases of M. pneumoniae pneumonia may experience neurological complications of varying severity (237, 294, 332, 387). Such complications have included encephalitis, cerebellar syndrome and polyradiculitis, cranial nerve palsies, aseptic meningitis or meningoencephalitis, acute disseminated encephalomyelitis, coma, optic neuritis, diplopia, mental confusion, and acute psychosis secondary to encephalitis (39, 98, 159, 226, 264, 331, 386, 392). A number of motor deficiencies have also been described, including cranial nerve palsy, brachial plexus neuropathy, ataxia, choreoathetosis, and ascending paralysis (Guillain-Barré Syndrome) (2, 4, 9, 12, 39, 44, 223, 226, 319). Encephalitis has been the most common neurological manifestation in children (237). Most patients with neurological complications experience them 1 to 2 weeks after the onset of respiratory signs, but 20% of patients or more have no preceding or concomitant diagnosis of respiratory infection (333). This figure may be higher yet in children (413).

Most of the first descriptions of CNS complications were based on serology and later on occasional isolation of M. pneumoniae from the respiratory tract rather than the CNS. The lack of clear evidence that mycoplasmas were actually present in neurological tissues led to theories that damage to brain tissue occurred as a result of cross-reacting or autoimmune antibodies (129, 142) and even to concern that neurological infections by other bacterial pathogens were causing false-positive mycoplasmal serology (230). The potential role of immunological sequelae of M. pneumoniae infection that can lead to neurological complications cannot be discounted, and some CNS complications are very likely due to this mechanism as opposed to direct invasion (312, 323). Antibodies against galactocerebroside, a component of CNS myelin, has been detected in 100% of patients with M. pneumoniae and CNS involvement and in only 25% of those without CNS involvement (312). Postinfectious leukoencephalopathy due to M. pneumoniae also suggests a role for autoimmunity in some cases (325).

Proof that viable organisms or M. pneumoniae DNA can be detected directly in neural tissues and CSF provides convincing evidence that this organism does indeed disseminate from the respiratory tract in some instances (3, 105, 198, 251, 307, 401, 403, 415). Neurological manifestations associated with M. pneumoniae infections usually resolve completely, but they can result in chronic debilitating deficits in motor or mental function (384). These conditions can be severe and life threatening. Rautonen et al. (341) reported that children with M. pneumoniae were seven times more likely to die or have severe neurological sequelae than other children, second only to cases of herpes simplex virus infection. The presence of peripheral neurological sequelae such as radiculitis and transverse myelitis has been identified as a risk factor for chronic CNS sequelae (59, 331). Central nervous system effects due to mycoplasmas have been reviewed in detail by Talkington (401).

Whereas neurological disorders may be the most severe extrapulmonary manifestations of M. pneumoniae infections, dermatological disorders, including erythematous maculopapular and vesicular rashes, are perhaps the most common clinically significant complications, occurring in up to 25% of patients. Although the disorders are usually self-limited, severe forms of Stevens-Johnson syndrome, conjunctivitis, ulcerative stomatitis, and bullous exanthems have been reported, and the organism has been detected directly in the cutaneous lesions (70, 71, 256, 276, 395). Clinicians should keep in mind that the presence of erythematous maculopapular rashes in M. pneumoniae patients can also be caused by a number of antibiotics commonly used to treat respiratory tract infections.

Nonspecific myalgias, arthralgias, and polyarthropathies occur in approximately 14% of patients with acute M. pneumoniae infection and may sometimes persist for long periods (7). Septic arthritis with detection of the organism directly in synovial fluid has been reported numerous times, occurring most commonly in hypogammaglobulinemic patients but sometimes in immunocompetent persons (92, 183, 209, 249, 304, 329, 351, 374, 408, 410, 441). In view of the well-known associations of animal mycoplasmas and their ability to produce chronic, naturally occurring infection of the joints and the experimental models, joint involvement by M. pneumoniae is not unexpected. Little attention has been paid to invasive infections of bones due to M. pneumoniae, but at least one report of osteomyelitis in a splenectomized patient with hypogammaglobulinemia has been attributed to M. pneumoniae, in which the infection was detected by PCR (250).

Cardiac complications associated with M. pneumoniae are relatively uncommon, but involvement has been reported at rates of from 1 to 8.5% in persons with serological evidence of infection, somewhat more commonly in adults than in children (287, 330). Pericarditis, myocarditis, and pericardial effusion with and without cardiac tamponade have all been described, and the organism has been detected in pericardial fluid (24, 122, 128, 171, 218, 248, 287, 364, 384, 400). According to one study (330), almost half of the patients with M. pneumoniae infection had symptoms or signs of heart abnormalities an average of 16 months later.

Hemolytic anemia is recognized as a rare but severe complication of mycoplasmal pneumonia, occurring more often in children than in adults (70, 122, 381). The mechanism by which M. pneumoniae causes this complication has been attributed to cross-reacting cold agglutinins (80, 122, 381, 403, 424). Two cases of aplastic anemia associated with M. pneumoniae have also been reported (390). A recent report suggests that thrombotic thrombocytopenic purpura associated with M. pneumoniae infection may be the result of cross-reactive antibodies inactivating plasma von Willebrand factor-cleaving protease (23). Fulminant infection leading to fatal disseminated intravascular coagulation has also been reported (73), as has a case of priapism in a 12-year-old boy that was felt to be due to the hypercoagulable state that sometimes occurs in association with M. pneumoniae infection (192). If subclinical forms of hemolytic anemia and intravascular coagulation are considered, over 50% of patients with M. pneumoniae infections may be affected.

Acute glomerulonephritis, renal failure, tubulointerstitial nephritis, and IgA nephropathy, as well as other conditions, have been sporadically reported in association with M. pneumoniae infections (217, 235, 321, 369, 430). Kanayama et al. (217) reported cases of IgA nephropathy in persons in whom the mycoplasma infection was diagnosed serologically. Attempts to demonstrate mycoplasma antigen in damaged renal tissue by immunohistochemical techniques have not been uniformly successful, once again leading to theories that an antibody-mediated pathogenesis is responsible (217, 431). A recent attempt to use PCR to identify mycoplasmas in renal tissue from four children with acute nephritis concomitant with serological evidence of recent M. pneumoniae infection also failed (360). However, the presence of mycoplasma antigen has been demonstrated by immunoperoxidase staining in renal tissue in a patient with acute interstitial nephritis (10).

M. pneumoniae infection may be associated with a variety of nonspecific complaints related to the gastrointestinal system. These include nausea, vomiting, and diarrhea. Rarely, cholestatic hepatitis and pancreatitis have been associated with respiratory infections (11, 174, 381, 386).

Up to one-third of patients with M. pneumoniae infection may have nonspecific ear symptoms, including otitis externa, otitis media, and myringitis (301, 392). Acute rhabdomyolysis was recently reported in association with M. pneumoniae infection in a 15-year-old patient (41). Ocular manifestations have been reported in children occasionally and include conjunctivitis, anterior uveitis, optic neuropathy, retinitis and retinal hemorrhages, iritis, and optic disk swelling, with or without permanent degradation of vision (289, 362). M. pneumoniae has been isolated from the urogenital tracts of males and females and has been cultured from a tubo-ovarian abscess (165). Given the apparent ability of the organism to invade the bloodstream, infections in almost any organ system are possible. Higuchi et al. (184, 185) reported the detection of M. pneumoniae by PCR in ruptured atherosclerotic plaques and stenotic heart valves and speculated about its possible association, along with C. pneumoniae, as a risk factor for embolization and myocardial infarction. Additional data are necessary to determine the significance of these preliminary reports.

DIAGNOSIS

General Laboratory Features

Clinical laboratory findings are seldom diagnostic for M. pneumoniae infection. About one-third of persons with lower respiratory tract infections may have leukocytosis (392). An elevated erythrocyte sedimentation rate may also be observed (45). Gram staining of sputum may show mononuclear cells or neutrophils and normal flora. There are no hepatic or renal abnormalities typical of M. pneumoniae infection, although the hemolytic anemia that develops in some patients may be reflected in the hemogram.

Prior to the widespread availability of commercialized antibody assays, and even before the precise bacteriological characterization of the etiological agent was known, clinicians sometimes used the presence of cold agglutinins to confirm their clinical suspicions of primary atypical pneumonia, which was also known as cold-agglutinin disease. Cold agglutinins are actually IgM antibodies that are produced 1 to 2 weeks after initial infection in about 50% of M. pneumoniae infections and may persist for several weeks. One theory is that cold agglutinins are the result of cross-reactive autoantibodies developed against the I antigen of human erythrocytes during acute mycoplasmal infection. Another theory is that they develop directly as a result of antigenic alteration of the erythrocytes caused by M. pneumoniae. The latter theory is supported by the facts that M. pneumoniae is known to interact with and adsorb to human erythrocytes and to produce a hemolysin and because cold agglutinins can be induced in rabbits inoculated with human erythrocytes that were preincubated with M. pneumoniae but not in rabbits inoculated with either M. pneumoniae or erythrocytes alone (16).

Two varieties of cold agglutinin determination have been used. The simplest version of the test was what is referred to as “bedside cold agglutinins.” This consists of collecting blood from a patient suspected of having mycoplasmal pneumonia into a tube containing citrate or another suitable anticoagulant and placing it in ice water in a freezer for approximately 30 s. The tube is then examined for coarse agglutination by being tilted on its side. On warming, the agglutination should resolve, but it can be reproduced by repeating the cooling procedure. A more precise test is to determine the cold agglutinin titer by reacting doubling dilutions of patient sera suspected to contain the cold agglutinins with human type O erythrocytes in the cold and determining the highest dilution at which agglutination occurs. The cold agglutinin response often correlates directly with the severity of pulmonary involvement; patients with extensive lobar involvement will usually have a titer of ≥1:32, whereas those with only minimal illness may not develop cold agglutinins. However, cold agglutinins also occur in persons who have various bacterial and rickettsial infections as well as in those with influenza virus and adenovirus infections (16). Due to the availability of antibody assays that are based on immunological reagents specific for M. pneumoniae, the popularity of testing serum for the presence of cold agglutinins has declined. If used, this test should be limited to persons in whom mycoplasmal pneumonia is a strong possibility. Under these circumstances, a positive test provides reasonable supportive evidence of a mycoplasmal etiology, on the basis of which clinical management decisions can be made.

Radiographic Findings

Primary atypical pneumonia due to M. pneumoniae can be extremely variable and mimic a wide variety of lung diseases. The inflammatory response elicited by M. pneumoniae causes interstitial mononuclear inflammation in the lungs that may be manifested radiographically as diffuse, reticular infiltrates of bronchopneumonia in the perihilar regions or lower lobes, usually with a unilateral distribution, and hilar adenopathy. Bilateral involvement may occur in about 20% of cases (137). However, lobar consolidation with bilateral alveolar involvement has been described, and the degree of consolidation may exceed what would be expected based on the severity of clinical manifestations (97). Among patients with M. pneumoniae pneumonia who require hospitalization, up to 10.9% will require mechanical ventilation (282).

Pathological Findings

Histopathological examinations from fatal cases of M. pneumoniae pneumonia, biopsy material, examination of tissues from animal models, and tracheal organ cultures have shown lesions of the epithelial lining of the mucosal surfaces with ulceration and destruction of ciliated epithelium of bronchi and bronchioles. Edema of bronchial and bronchiolar walls; bronchiolar and alveolar infiltrates of macrophages, lymphocytes, neutrophils, plasma cells, and fibrin; and bronchiolitis obliterans have been described (65, 77, 93, 259, 277, 352, 372, 432). Type II pneumocyte hyperplasia and diffuse alveolar damage have also been reported. Pleura may contain patches of fibrinous exudates. Pleural effusions and diffuse alveolar damage sometimes occur in association with more severe cases, and long-term sequelae such as pleural scarring, bronchiectasis, and pulmonary fibrosis have been reported (40, 72, 97, 222, 338, 372). Lung abscesses may also occur (258, 379). Recent studies utilizing animal models developed to investigate the potential role of M. pneumoniae in chronic lung conditions such as asthma (discussed above) have also provided insights into the histopathological aspects of M. pneumoniae lung disease (178, 179). Immunosuppressed persons with M. pneumoniae infection may lack pulmonary infiltrates, further attesting to the importance of the host immune response in lesion development (151).

Microbiological Tests

The lack of rapid and accurate diagnostic laboratory tests to detect M. pneumoniae infection has hampered understanding of the epidemiology as well as contributed to unawareness of the potential clinical significance of this common pathogen by many physicians. From the microbiologist's point of view, one might argue in favor of the need to routinely use the best means available to identify persons with acute M. pneumoniae respiratory tract infection under all circumstances. However, from a practical standpoint, one must also consider therapy that will generally be provided empirically. Thus, the performance of diagnostic tests for M. pneumoniae in a typical ambulatory care setting may be unnecessary much of the time. This approach has been taken in the Infectious Disease Society of America diagnosis and treatment guidelines for community-acquired pneumonias (25). However, in the event of an illness in which M. pneumoniae is suspected and that is of sufficient severity as to require hospitalization, especially if the patient has any sort of immune deficiency or underlying condition that may make an unfavorable outcome more likely, attempts to detect M. pneumoniae infection are justified. Unfortunately, most of the diagnostic methods that are currently available for detection of M. pneumoniae infections are somewhat better suited for use in epidemiological studies as opposed to direct management of individual patients due to their prolonged turnaround time, limited availability, and cost. Among currently available test modalities, each has limitations.

Descriptions of the physiological, serological, and morphological procedures for detection and identification of mycoplasmas in clinical specimens have been summarized and updated to include more of the molecular-based methods in Molecular and Diagnostic Procedures in Mycoplasmology, vol. 2, published in 1996 (421). Waites et al. (434, 438, 439) have summarized current methods for clinical diagnosis and susceptibility testing of human mycoplasmas and have discussed commercial products used for culture and serology. Razin (343) has also provided an overview of diagnosis of mycoplasmal infections with commentary on various molecular-based methods. Brief summaries and commentary on culture-based and non-culture-based methods for detection of infections due to M. pneumoniae are presented here.

Culture.

Culture of M. pneumoniae from the respiratory tract and other body sites is laborious and expensive, requiring serial blind passages, specialized and expensive growth media, and incubation periods of up to several weeks. Compared to molecular techniques such as PCR, its analytic sensitivity may be no more than 60% in experienced laboratories, even when rigorous adherence to procedures known to enhance cultural isolation are used (199). However, when positive, culture has the advantage of being 100% specific, provided that appropriate additional procedures are used to identify the organism isolated to species level. The persistence of M. pneumoniae for variable lengths of time following acute infection also makes it difficult in some cases to assess the significance of a positive culture or assay without additional confirmatory tests such as serology, as discussed above. For the reasons cited above, culture is rarely used for routine diagnosis and/or management of M. pneumoniae infections. In view of the many limitations of culture for detection of M. pneumoniae, if attempted it should be augmented by additional diagnostic methods such as PCR and/or serology.

The lack of reliable commercially prepared media in the past effectively prevented many clinical laboratories from offering M. pneumoniae detection by culture, even before alternative techniques such as PCR existed. If culture is attempted, isolation of M. pneumoniae from nasopharyngeal or throat swabs or lower respiratory tract specimens should be considered clinically significant in most instances but should be correlated with the presence of clinical respiratory disease due to the possibility of asymptomatic carriage. Due to the organism's sensitivity to adverse environmental conditions, proper specimen collection, storage, and transport are critical for maintaining viability for culture processing and DNA extraction. Currently recommended methods for specimen collection, transport medium, transport and storage conditions, selection and preparation of growth medium, inoculation, incubation, and organism identification for patients suspected of having M. pneumoniae infections have been described in considerable detail in reference texts (434, 438, 439), and only a brief commentary will be provided here.

Isolation of M. pneumoniae from essentially any type of clinical specimen from the respiratory tract, as well as from other body fluids and tissues that are suitable for cultivation of conventional bacteria, is possible. SP4 medium, developed over 20 years ago by Tully et al. (422), has become the most successful and widely used broth and agar medium for cultivating M. pneumoniae for clinical purposes. It is sold commercially in the United States by Remel Laboratories (Lenexa, Kans.). Detection of M. pneumoniae by culture is predicated on its characteristic hydrolysis of glucose, with a resultant acidic shift, after 4 days or more of incubation in broth containing a phenol red pH indicator. Initial specimens, as well as broths with color change and blind subcultures, should be transferred to SP4 agar, incubated, and examined under a stereomicroscope at regular intervals to look for development of spherical colonies of up to 100 μm in diameter (Fig. 2). Incubation of M. pneumoniae broth cultures under atmospheric conditions at 37°C is satisfactory, but agar plates will yield the best colonial growth if 5% CO2 is provided and plates are sealed to prevent loss of moisture during prolonged incubation. A biphasic medium consisting of an agar slant in a screw-cap bottle to which broth is added is sold by Irvine Scientific in the United States under the trade name Mycotrim RS.

Although the common commensal mycoplasmal species from the oropharynx have different metabolic properties, growth rates, and requirements that distinguish them from M. pneumoniae, it is necessary to perform additional tests to conclusively prove the identification of a mycoplasma from the respiratory tract or another body site as M. pneumoniae. Such procedures for species identification include hemadsorption with guinea pig erythrocytes, reduction of tetrazolium, agar growth inhibition with homologous antisera, epi-immunofluorescence or immunoperoxidase staining, immunoblotting with monoclonal antibodies, metabolism inhibition tests, and PCR assays. Unfortunately, none of these methods has been adapted for development of a commercial product that can be easily purchased and used in a diagnostic laboratory. Hemadsorption assays and tetrazolium reduction can theoretically be accomplished in most clinical laboratories that do not have capabilities for molecular diagnostic testing. However, the cost, the technical expertise required, and the very limited availability of the reagents for the immunologically based methods of species identification effectively preclude their use outside specialized reference or research laboratories.

Antigen detection techniques.

Rapid assays for direct antigenic detection of M. pneumoniae in respiratory tract specimens have included direct immunofluorescence, counterimmunoelectrophoresis, immunoblotting, and antigen capture enzyme immunoassay (157, 188, 189, 231, 234). The utility and general acceptance of these techniques have been reduced by low sensitivity and cross-reactivity with other mycoplasmas found in the respiratory tract. Considering that the concentration of M. pneumoniae cells typically found in sputum specimens from infected patients is approximately 102 to 106 CFU/ml and that antigen detection techniques may be able to detect about 103 to 104 CFU/100 μl of specimen, nonamplified antigen detection assays are at the limit of sensitivity for detection and are not recommended for diagnostic purposes when superior technology utilizing nucleic acid amplification is possible. Although not sold in the United States at present, commercial antigen detection tests for M. pneumoniae are available in some European countries.

DNA probes.

DNA hybridization techniques for the diagnosis of M. pneumoniae infection were developed in the early 1980s and had about the same diagnostic sensitivity as antigen detection techniques. These methods have been reviewed by Razin (344). The 16S rRNA genes have been widely used as targets, as have probes consisting of rDNA. GenProbe (San Diego, Calif.) previously sold a 125I-labeled DNA probe for an rRNA sequence specific for M. pneumoniae, but the more recently available amplification techniques, such as the PCR assay, that do not involve radioisotopes and have greater sensitivity led to its discontinuation.

PCR.

Development of molecular-based testing such as the PCR assay has lessened the importance of culture as a means for detecting M. pneumoniae. Studies since the late 1980s using simulated clinical specimens, animal models, and later clinical trials have validated the ability of PCR to detect M. pneumoniae, often in conjunction with serology and/or culture (43, 46, 47, 53, 96, 108, 111, 127, 180, 199, 211, 215, 239, 337, 382, 417, 443). The same types of clinical specimens that can undergo culture can also be tested by PCR. The use of two different targets can maximize the ability to detect the organism. The conventional PCR procedure currently used at the Centers for Disease Control and Prevention has been adapted from the procedure originally described by Bernet et al. (43), using primers derived from the M. pneumoniae ATPase gene. They have also developed a real-time PCR which targets this gene. Other sequences, primarily the P1 adhesin and conserved regions of 16S rRNA, have also been utilized (96, 211, 236, 275, 344, 402, 427, 429, 442). Table 2 lists selected original references for M. pneumoniae nucleic acid amplification assays that describe different target regions of the genome. For additional information and tabulations of various nucleic acid amplification assays according to respiratory specimen tested, or by publication with or without validation data, the reader is referred to three recent publications on this topic (95, 272, 300). Among the other advantages of PCR are that it can be used to detect mycoplasmas in tissue that has already been processed for histological examination or in cultures that are contaminated where culture is impossible, it requires only one specimen, it can be completed in 1 day, it may be positive earlier in infection than serology, and it does not require viable organisms (402, 434, 442). Specific advantages of RNA-based amplification techniques are the high sensitivity that can be achieved due to the large number of rRNA copies per mycoplasmal cell and the fact that its detection is more indicative of viable mycoplasmas in a clinical sample (95).

TABLE 2.

Selected original references on M. pneumoniae nucleic acid amplification assays

Authors Reference Gene target PCR product size (bp)
Bernet et al. 43 ATPase operon gene 144
Jensen et al. 211 P1 gene 153
Van Kuppeveld et al. 428 16S RNA 277
de Barbeyrac et al. 96 P1 gene 466
Luneberg et al. 275 tuf gene 950
Ramirez et al. 339 P1 gene 375

It is difficult to compare results of one study utilizing PCR for epidemiological or diagnostic purposes directly with another because of the varied specimen types, DNA extraction and amplification techniques, primer selection, and reference standards used for comparison. Most techniques are basically similar, but they may differ in targeted sequences and primers. However, comparison of the PCR technique with culture and/or serology has yielded varied results, and large-scale experience with this procedure is still limited for M. pneumoniae.

In view of the enhanced analytical sensitivity of the PCR assay over culture, a positive PCR result and negative culture can be easily explained. However, in a case with a negative PCR assay and a positive culture (or serology), the presence of inhibitors or some other technical problem with the PCR assay must be considered (127, 199, 346). Reznikov et al. (346) showed that PCR inhibition was much more likely to occur with nasopharyngeal aspirates than with throat swabs and recommended the latter specimen for diagnostic purposes for M. pneumoniae. Dorigo-Zetsma et al. (108) performed a comprehensive examination with 18 patients with M. pneumoniae respiratory tract infection detected by PCR or serology and showed that sputum was the specimen that was most likely to be PCR positive (62.5%, versus 41% for nasopharynx, 28% for throat swabs, and 44% for throat washes). Dilution of samples may sometimes overcome inhibition of PCR, but this may also diminish the sensitivity because the nucleic acid is diluted along with any inhibitors that may be present. There are also commercial reagents for nucleic acid purification that are effective in removing most inhibitors of amplification in PCR assays.

There is justified concern when the PCR assay is used as the sole means of detection for surveillance purposes without culture, serology, or clinical data because most studies using PCR have not attempted to do any type of quantitation. Since it is not known with certainty whether there is a specific threshold quantity of M. pneumoniae in respiratory tract tissues that can differentiate colonization from infection, a positive result by PCR may overestimate the clinical importance of M. pneumoniae as a pathogen if the population sampled has a high carriage rate and because of the propensity of this organism to cocirculate with other bacterial and viral pathogens. Williamson et al. (443) suggested a threshold of 104 genomic DNA copies per ml of throat washing as a threshold for differentiating clinical infection and carriage. Dorigo-Zetsma et al. (110) also proposed that the number of organisms may be related to the severity of clinical illness. Using a semiquantitative nested PCR on dilutions of nucleic acid extracted from throat washings of patients with M. pneumoniae respiratory disease, they calculated an organism load of 20 to 3,830 CFU/ml of throat sample. The mean M. pneumoniae load in samples from hospitalized patients was significantly higher than that in samples from nonhospitalized subjects. These studies are evidence of progress being made in the characterization of the microecological niche occupied by M. pneumoniae in the human respiratory tract and the optimum tools for its assessment, but much more remains to be done to fully appreciate and understand the true meaning of these findings.

Dorigo-Zetsma et al. (108) suggested that elderly adults with pneumonia who are PCR positive and serologically negative for M. pneumoniae might be deficient in antibody response due to a natural decline in the humoral immune system as part of the aging process (160). This was also observed in other seroepidemiological studies (94, 181). A positive PCR test and negative serology could also mean that the specimen was collected too early in the course of illness to allow sufficient time for antibody to develop. In view of the difficulty in the interpretation of serological data because of high background IgG level and sometimes prolonged persistence of IgM, there is also the possibility of false-positive serological data when concomitant PCR testing is negative, depending on the test methods and interpretive criteria. In the case of antibiotic treatment prior to specimen collection, the PCR result may be negative as soon as 24 h following treatment, but serological results may remain positive.

Further refinements to traditional PCR assays, such as combining PCR with a hybridization step, employment of a two-step (nested) procedure in which there is reamplification of a PCR product with a second primer set, real-time detection using specific probes with matched internal controls to evaluate polymerase inhibition, and quantitation of M. pneumoniae in acute and subclinical infections, will be very important (1, 177, 202, 403).

Until PCR assays can be standardized, made available at a reasonable cost, and sold commercially as complete diagnostic kits suitable for use in hospital-based or other reference laboratories, this method of diagnosis is unlikely to gain widespread use for detection of M. pneumoniae infection for clinical as opposed to epidemiological purposes in the United States. Further development of multiplex PCR tests to detect other atypical pathogens such as C. pneumoniae simultaneously with M. pneumoniae may make this type of test more attractive and practical for routine use in diagnostic laboratories. Such endeavors have been undertaken already, but no diagnostic products are being sold commercially in the United States thus far (84, 173, 418). Hardegger et al. (177) found that a real-time PCR assay was equal to a conventional nested PCR with regard to sensitivity in detection of M. pneumoniae in clinical samples, allowing for quantitation of the amplified product during PCR combined with a reduction in hands-on time. Development of quantitative PCR assays will be beneficial in facilitating better understanding of the carrier state associated with M. pneumoniae. Other modifications of the PCR assay for detection of M. pneumoniae, in which amplification is carried out in sealed glass capillary tubes, have been described by Honda et al. (194).

Serology.

Despite its drawbacks for use with immunosuppressed persons who are unable to mount an antibody response, serological diagnosis of M. pneumoniae respiratory infections has long been the cornerstone of M. pneumoniae diagnosis and for epidemiological studies because of the relative lack of sensitivity and time-consuming nature of culture. Also, the carrier state that may occur in an unknown percentage of persons in the absence of acute infection can potentially confound interpretation of PCR test results. Serum is easy to collect, store, and ship, but the need for acute- and convalescent-phase specimens and the complex and time-consuming nature of many of the serological assays that have been used in the past have limited acceptance of serology for routine diagnostic testing. Some of the newer and improved commercial assays have overcome some of these limitations. In view of these considerations, it is advisable to test simultaneously for both IgM and IgG in paired specimens collected 2 to 3 weeks apart for the most accurate diagnosis of recent or current M. pneumoniae infection, especially in adults (411). A fourfold or greater rise in antibody titer indicates a current or recent infection. The late elevation of IgG that sometimes occurs, the high seroprevalence of IgG antibodies that persist for long periods in persons with a history of M. pneumoniae infection, and the lack of an IgM response in adults complicate and impose serious limitations on the use of serology as a sole means for diagnosis of M. pneumoniae infections (343). Thus, a logical approach would be to incorporate PCR and serological studies for IgG and IgM for optimum diagnosis of M. pneumoniae infections.

Historically, the CF test gained early popularity among laboratories that routinely ran CF tests for viral agents, and it was the sole means for detecting antibodies to M. pneumoniae for many years. However, mycoplasmas are much more antigenically complex than viruses, leading to nonspecific reactions. The glycolipids of M. genitalium are highly cross-reactive with M. pneumoniae due to shared lipid antigens (265), and this also causes problems for CF tests. Sera from patients with bacterial meningitis also tend to have high mycoplasmal CF titers. Cross-reactions of CF antigens with other organ-specific antigens unrelated to microorganisms may also occur.

Kenny et al. (225) reported that among patients with M. pneumoniae culture-positive, X-ray-proven pneumonia, 53% showed a fourfold titer increase and 36% showed antibody titers of ≥32. With both high titers and high stationary-phase titers as criteria, the sensitivity of the CF test was 90% and the specificity was 88%. Single titers of >32 are sometimes considered to be indicative of recent infection. However, this endpoint varies greatly among laboratories, and antibodies to glycolipid antigens may persist for at least a year after infection (343). Confirmation of CF test results with Western immunoblots can aid in interpretation but greatly adds to the time and expense of testing. Since CF measures mainly IgM and to a minor extent IgG, its diagnostic value may be limited to initial M. pneumoniae infections (203).

CF tests have largely been replaced by improved methods using alternative technologies that are less time-consuming and labor-intensive. The target antigens vary among tests; some are protein, while others are glycolipid. Whether there is a significant cross-reactivity with these M. pneumoniae antibody test systems and M. genitalium has not been established with certainty, but it seems somewhat unlikely. A great many publications describing a number of different techniques for measuring antibody response to M. pneumoniae infection have appeared since the early 1980s, but many of the assays have never been developed for commercial distribution. Some have been used for seroepidemiology studies, however. Several of the existing commercial kits have been evaluated by using CF as the reference method. Considering its lack of antibody class distinction and tendency for cross-reactivity with other microorganisms, CF is not really suitable as a reference standard. Appreciation of this fact makes interpretation of the specificity and sensitivity results of comparative studies problematic. Some recent studies evaluating commercial kits have merely compared one method or product with another without consideration of CF data, but since results and conclusions from some studies are based upon assay of a single acute-phase serum sample while others have used paired specimens, direct comparisons and extrapolations from multiple studies become rather complex and are not always feasible.

Assay formats adapted for commercial distribution include indirect immunofluorescence assay (IFA), particle agglutination (PA) assay, and enzyme-linked immunoassay (EIA). A German company, Genzyme Virotech GmbH, has developed a Western blot assay for detection of IgG and IgA, but this test is not sold in the United States. Summaries of the major commercial serological test kits and their various assay formats have been recently published (434, 439), so explicit details of the various methods and their procedures are not provided in this review.

IFAs for M. pneumoniae consist of antigen affixed to glass slides. Specific antibody is detected after staining with anti-human IgM or IgG fluorochrome conjugate. These kits provide accurate, quantitative serological data, but their interpretation is subjective and a fluorescence microscope is necessary. Results can be affected by the presence of rheumatoid factor and high M. pneumoniae-specific IgG antibody levels, and additional procedures are required to validate IgM results in these settings (403).

The PA tests in use today mainly utilize latex or gelatin as the carrier particles that are incubated with test serum. If the serum contains specific antibodies, the particles agglutinate, resulting in a visible reaction. PA tests use a mixture of M. pneumoniae antigens to detect both IgG and IgM simultaneously. Several PA products that provide qualitative results that can be visualized on a card or quantitative data that can be read in a microtiter plate format can be purchased commercially (13, 22, 116, 219, 262, 286), but not all of the products are available in the United States. Lieberman et al. (262) concluded that the Sero M. pneumoniae antibody capture EIA kit was better than the Fujirebio Serodia Myco II PA kit for diagnosis of current M. pneumoniae infection when a single serum sample was tested, but they stressed that testing of acute- and convalescent-phase sera is required for precise diagnosis, even with the EIA. Barker et al. (22) also compared the Serodia Myco II PA kit with IgM-specific antibody capture EIA and IFA. They determined that this PA test was not as specific for detection of IgM as the other tests. Aubert et al. (14) felt that PA tests performed about as well as EIAs and IFAs, but Matas et al. (286) reported the Meridian ImmunoCard EIA was able to detect lower levels of IgM antibodies than either the Serodia Myco II kit or CF tests. Karppelin et al. (219) evaluated the Serofast (International Microbio, Toulon, France) and found it to be less sensitive than CF and EIA. Echevarria et al. (116), compared the Serofast kit to an antibody capture EIA by using sera that were shown to be either positive or negative by CF. Both tests were shown to be sensitive and specific, but the performance of the antibody capture EIA for detection of IgM diminished with older persons. Based on evidence presented to date, PA assays do not offer any advantages over other techniques such as EIAs or IFAs, except possibly their ease and simplicity of performance.

EIAs are the most widely used commercial mycoplasma serology tests in the United States. They are amenable to a variety of assay conditions suitable for testing large or small numbers of sera, detect very small amounts of antibody, require serum volumes of ≤100 μl, can be made isotype specific, and are generally more sensitive than CF (434). EIAs are more sensitive for detecting acute infection than culture and can be comparable in sensitivity to PCR, provided that a sufficient time has elapsed since infection for antibody to develop and that the patient has a functional immune system. Crude multiantigen preparations, purified proteins (including the P1 adhesin), μ-capture approaches, purified glycolipids, and synthetic peptides have all been used as targets (22, 64, 116, 190, 191, 203, 303, 399, 425, 445). Patient sera are incubated with the solid-phase antigen, and bound antibodies are visualized by using substrate and enzyme-labeled conjugates directed against the primary antibody. The amount of conjugate reacting is proportional to the levels of antibody present in the patient's serum, measured quantitatively with a spectrophotometer. Most EIAs are sold in 96-well microtiter plate formats, although some can be obtained as breakaway microwell strips which allow smaller numbers of sera to be tested eco-omically.

Two EIAs are packaged as qualitative membrane-based procedures for the detection of single test specimens. These are truly rapid EIAs (10 min or less) and are simple to perform. The ImmunoCard (Meridian Diagnostics, Cincinnati, Ohio) is an IgM-only assay that is simple to read, and it is especially useful for testing pediatric samples. As are typical of IgM-only assays based on M. pneumoniae protein antigens, the specificity may be somewhat compromised for patients with autoimmune disease and there are the limitations in interpreting the results. Studies using sera from confirmed M. pneumoniae cases have also shown that the ImmunoCard performed better than the CF test (6, 286, 412). The Remel EIA is a membrane-based assay that detects IgM and IgG simultaneously and has shown good sensitivity and specificity when compared to other tests, including IFA, the GenBio ImmunoWell EIA, the Diasorin EIA, the Meridian ImmunoCard, and CF (130, 412). The manufacturers have endorsed the use of a single assay with the Remel EIA for diagnosis of acute M. pneumoniae infections in young persons, and other manufacturers have suggested that high titers of IgM or IgG above a specified cutoff value may be used to indicate acute infection. However, we contend that acute- and convalescent-phase sera are necessary for greatest accuracy, but we acknowledge the fact that the value of the single-point-of-care tests offered by both the Remel and Meridian kits is lost if paired specimens are required. The cost of materials to perform these rapid EIAs exceeds U.S. $10 for each patient tested.

ANTIMICROBIAL SUSCEPTIBILITIES AND CHEMOTHERAPY

Antimicrobial Susceptibility Profiles

A summary of the in vitro activities of several antimicrobial agents against M. pneumoniae is shown in Table 3. M. pneumoniae is inhibited by tetracyclines, macrolides, ketolides, and fluoroquinolones, with little variation in MICs among clinical isolates (435, 436). Thus, in vitro susceptibility testing is not indicated for routine patient management purposes for infections of the respiratory tract. Other agents that are active at the bacterial ribosome, such as streptogramins, chloramphenicol, and aminoglycosides, may also show in vitro inhibitory activity against M. pneumoniae but are not widely used for therapeutic purposes against this organism. Clindamycin may be effective in vitro, but limited reports suggest that it may not be active in vivo, and it has not been considered a first-line treatment (80). Due to the lack of a cell wall, all mycoplasmas are innately resistant to all beta-lactams and glycopeptides. Sulfonamides, trimethoprim, polymixins, nalidixic acid, and rifampin are also inactive. Linezolid is the prototype agent of the oxazolidinone class. These agents are much less active against M. pneumoniae than the other agents that inhibit protein synthesis (224). New quinolones such as moxifloxacin, gatifloxacin, garenoxacin, gemifloxacin, and sparfloxacin tend to have somewhat greater in vitro activity than older agents such as ciprofloxacin, ofloxacin, and levofloxacin, although MICs of all fluoroquinolones are severalfold higher than those of macrolides (224, 435-437). Fluoroquinolones have been shown to be bactericidal for M. pneumoniae, whereas macrolides and tetracyclines are primarily bacteriostatic (36, 113, 435, 437).

TABLE 3.

MIC ranges of various antimicrobials against M. pneumoniaea

Drug MIC (μg/ml)
Tetracycline 0.63-0.25
Doxycycline 0.016-2
Erythromycin ≤0.001-0.016
Roxithromycin ≤0.01
Dirithromycin 0.12-0.5
Clarithromycin ≤0.001-0.125
Azithromycin ≤0.001-0.125
Josamycin ≤0.01-0.125
Telithromycin 0.008-0.06
Cethromycin ≤0.001-0.016
Clindamycin ≤0.008-2
Lincomycin 4-8
Pristinamycin 0.02-0.05
Chloramphenicol 2
Gentamicin 4
Ciprofloxacin 0.5-4
Ofloxacin 0.05-2
Levofloxacin 0.063-2
Sparfloxacin ≤0.008-0.5
Grepafloxacin 0.06-0.25
Trovafloxacin ≤0.008-0.5
Gatifloxacin ≤0.016-0.25
Moxifloxacin 0.06-0.25
Gemifloxacin ≤0.008-0.125
Garenoxacin 0.008-0.125
Gentamicin 4
Quinupristin-dalfopristin 0.008-0.06
Linezolid >64
Evernimicin 2.0-4.0
a

Data were compiled from multiple published studies in which different methodologies, and often different antimicrobial concentrations, were used.

The extent of naturally occurring, clinically significant acquired antimicrobial resistance of M. pneumoniae to any drug class is not known, but it is generally believed to be uncommon. Treatment failures have been rarely reported for microbiologically proven cases of M. pneumoniae infection, and recent studies evaluating macrolides, tetracyclines, or fluoroquinolones indicate comparable in vitro activities against isolates obtained from North America and Europe, with no change from data reported earlier other than some minor variations that may be due to differences in testing methods, media, and inocula (32, 34, 311, 435). However, organisms from acute M. pneumoniae infections are seldom sought by culture, they are almost never tested for in vitro susceptibilities for clinical purposes, and most in vitro studies reported to date tested relatively small numbers of clinical isolates from limited geographic areas. Organisms can be shed in respiratory secretions for long periods after acute infection, and high-level-macrolide-resistant strains have been isolated following erythromycin treatment, but resistance did not appear to affect clinical outcome (310, 394). Macrolide-resistant M. pneumoniae mutants can easily be selected in vitro (311, 393, 394); such mutants typically exhibit the macrolide-lincosamide-streptogramin B-type resistance, rendering lincosamides and streptogramin B inactive in addition to the macrolides.

Experimental observations of two laboratory-derived erythromycin-resistant M. pneumoniae mutants indicated that macrolide resistance can occur due to point mutations leading to A-to-G transitions in the peptidyl transferase loop of domain V of the 23S rRNA gene at positions 2063 and 2064, which reduces the affinity of these antibiotics for the ribosomes (274, 316). The likelihood of M. pneumoniae developing resistance to macrolides by this mechanism under natural conditions may be enhanced, since there is only a single rRNA operon in the M. pneumoniae genome (163). No plasmids or erm genes to mediate ribosomal modification, or any enzymes that break down macrolides, have ever been described for M. pneumoniae. The well-documented occurrences of the tetM determinant associated with the conjugative transposon Tn916, which confers resistance to the tetracyclines, in M. hominis and Ureaplasma spp. (34, 348, 349) and of mutations in DNA gyrase and topoisomerase that confer fluoroquinolone resistance (34, 37) prove the potential for mycoplasmas to acquire resistance to these drug classes. However, to date no such naturally occurring resistance to tetracyclines or fluoroquinolones in M. pneumoniae has been reported in the published medical literature to our knowledge. Whether the bactericidal effect of various fluoroquinolones against M. pneumoniae shown in vitro is more effective in preventing persistence of the organism in respiratory secretions after clinical resolution has not been documented.

In Vitro Susceptibility Testing

Even though susceptibility testing is not necessary to guide treatment of M. pneumoniae infections, such procedures are needed in order to evaluate new or investigational antimicrobial agents in comparison to existing drugs, occasionally for systemic infections (especially if the host is immunosuppressed), if treatment is not successful clinically, and/or for eradication of the organism from a normally sterile site. There are no official guidelines for performance, interpretation, or quality control of in vitro susceptibility tests for human mycoplasmas published by the National Committee for Clinical Laboratory Standards (NCCLS). However, a subcommittee of the NCCLS was established in 2001 to address this, and work is currently under way to develop in vitro susceptibility protocols and quality control ranges for representative drug classes for M. pneumoniae. M. hominis, and Ureaplasma spp. In the absence of NCCLS-sanctioned methods, the Mycoplasma Chemotherapy Working Team of the International Research Program on Comparative Mycoplasmology has formulated methods for determination of MICs by broth dilution and agar dilution. These methods have been summarized by Waites et al. (434). Procedures for determination of MICs for human mycoplasmas are also available in other publications, which can be used as a basis for development of laboratory protocols (33, 407). Many aspects of susceptibility testing of mycoplasmas are identical to the procedures that have been developed and standardized for conventional bacteria. However, the fastidious nature, complex growth requirements, and incubation length and conditions for mycoplasmal growth have to be considered in optimizing testing for these organisms. Broth- and agar-based methods for determining MICs have been adapted for testing mycoplasmas, and some general comments regarding the procedures to be performed are relevant for both techniques. As with all bacteria, the inoculum of organisms is important. Since mycoplasmas do not produce turbidity in liquid medium, it is somewhat difficult and complex to determine the number of viable cells present in an active culture. Experience has shown that MICs are not greatly affected when M. pneumoniae taken from frozen stocks is used. Thus, some laboratories prefer to test a frozen stock culture that was serially diluted previously to determine the number of viable cells and then dilute it to an inoculum containing 104 to 105 color-changing units/ml for use directly after a 2-h prewarming period (435, 436). SP4 medium and Hayflick's modified medium (pH 7.4 to 7.6) have been used for determining MICs for M. pneumoniae, with similar results (35, 434, 435). MIC test mixtures should be incubated at 37°C. Broth-based test mixtures can be incubated under atmospheric conditions, whereas agar plates should be supplemented with 5% CO2 for optimum colony development. Mandatory controls that must be used for all types of MIC assays include a sterility control, a growth control, and solvent control. Simultaneous testing of an isolate of M. pneumoniae for which MICs of the drugs of interest are known and consistent is also advisable. Due to the slow growth of M. pneumoniae, MIC test mixtures have to be incubated for 5 days or more and examined daily. No breakpoints to designate susceptibility or resistance have been designated for mycoplasmas. However, some inference about the meaning of the MICs obtained can be made based on breakpoints established for other bacteria and the achievable concentrations within the body for the drugs being tested. Generally speaking, if the MIC is ≤1 μg/ml, the drug may potentially be active against M. pneumoniae.

The agar dilution method of susceptibility testing has the advantages of having a stable endpoint over time, unlike broth dilution, where color change endpoints tend to shift over time. Agar dilution is suitable for testing large numbers of organisms at once. Testing small numbers of isolates by agar dilution is not very practical, since several individual agar plates must be prepared for each drug to encompass all of the dilutions that may be needed and media must be used when fresh. This method of testing is ideally performed by using a Steers replicator to deliver the inoculum to the agar plate, with a goal of generating 30 to 300 colonies in the growth control (434). The MIC is read as the lowest concentration of drug that prevents colony formation when colonies are evident on the growth control plate.

The broth microdilution technique is based on the principle that a constant number of organisms are added to serial doubling concentrations of antimicrobial agents diluted in broth in a 96-well microtiter plate. Broth microdilution is the most practical and widely used method of susceptibility testing for M. pneumoniae. Multiple antimicrobials can be tested in the same microtiter plate at several different dilutions. The inoculated microtiter plates are sealed with an adhesive cover to prevent drying out and are incubated until the growth control shows a color change. The MIC is read as the lowest concentration of drug that inhibits growth at the time the growth control first shows evidence of growth. Minimal bactericidal concentrations can be tested directly by diluting and subculturing wells in the microtiter plate that show no evidence of growth (color change) into fresh medium. When the minimal bactericidal concentration is within two dilutions of the MIC (fourfold), the agent may be considered bactericidal (435).

Waites et al. (435) have described methods for performing time-kill assays in which multiple dilutions of antimicrobials are incubated in broth with inocula of M. pneumoniae. Bactericidal activity over time is assessed by removal of small volumes of broth daily for up to a week and subculturing to agar plates. Colony counts performed on the subcultures will demonstrate the extent of bacterial killing and the time of exposure to the antimicrobial that is necessary to achieve this effect. Traditionally, a reduction of 99.9% or 3 log10 dilutions of the original inoculum has been used to define bactericidal as opposed to bacteriostatic activity (435).

Treatment of Infections Due to M. pneumoniae

When M. pneumoniae infections were first described soon after the organism was characterized and isolated in culture in the 1960s, there was some sentiment that antibiotic therapy was unnecessary since the disease is self-limiting most of the time. We now know that administration of antimicrobials will generally produce satisfactory results, with a marked reduction in duration of respiratory symptoms compared to nontreatment. Treatment of M. pneumoniae infections on a case-by-case basis has been guided primarily by its well-known and consistent susceptibilities to drugs in the macrolide, tetracycline, and fluoroquinolone classes that are available for oral administration. Appreciation of M. pneumoniae as a significant respiratory tract pathogen and the numerous new drugs in the macrolide and fluoroquinolone classes that have become available since the early 1990s have led to the performance of several trials involving adults and children with community-acquired pneumonia in which the clinical efficacies of various antimicrobials were evaluated. Many published clinical trials have been able to identify relatively small numbers of community-acquired pneumonias proven to be caused by M. pneumoniae and have usually relied upon serological diagnosis, although some recent studies have incorporated culture and/or PCR and thereby improved the numbers of microbiologically proven cases. The use of serology alone as a diagnostic measure precludes determination of whether a treatment regimen actually eradicates the organism, and thus data regarding the microbiological efficacy of any antimicrobial regimen are relatively sparse.

Newer fluoroquinolones are being used extensively for treatment of respiratory tract infections in adults since they can be used empirically to treat infections due to mycoplasmas, chlamydiae, legionellae, M. catarrhalis, and S. pneumoniae (including penicillin- and macrolide-resistant organisms); they have an acceptable safety profile; and they can be given orally once daily with favorable results. However, fluoroquinolones are not recommended for use in children due to possible toxicity to developing cartilage. Likewise, tetracyclines are not approved for use in children younger than 8 years of age. Macrolides are generally considered the treatments of choice for M. pneumoniae in both adults and children.

Table 4 summarizes several comparative and noncomparative studies in which identification of M. pneumoniae infections in adults or children with community-acquired pneumonia was specifically sought and in which the clinical and/or bacteriological response was documented for this organism. The results of these studies have generally shown that newer agents such as clarithromycin or azithromycin are as effective clinically as erythromycin, with cure rates consistently exceeding 90% when such data are available. These recent clinical trials evaluating treatment regimens for M. pneumoniae have not included untreated controls, so the cure rates for various drugs cannot account for the possibility of spontaneous resolution, which may occur in approximately 10 days (137). However, a treatment arm of one study (139) consisted of beta-lactams alone, and all 22 patients with M. pneumoniae infection identified serologically who received these agents achieved clinical cure, suggesting that these infections can indeed resolve spontaneously, since these drugs would have no effect on this organism. However, there could possibly have been another undetected pathogen involved that was inhibited by these agents, thus accounting for a favorable outcome.

TABLE 4.

Clinical trials for treatment of community-acquired pneumonia in which tests to identify infections with M. pneumoniae were included

Authors Reference Patients Drug(s) M. pneumoniae infections
No. of cases Detection method(s) Outcome and/or comment
Cassell et al. 63 Adults Clarithromycin (14 days) vs erythromycin (14 days) 15 Culture, serology Clinical cure was achieved in 4 of 4 (100%) and 3 of 3 (100%) subjects receiving clarithromycin or erythromycin, respectively
Block et al. 47 Children Clarithromycin (10 days) vs erythromycin (10 days) 69 Culture, PCR, serology Mycoplasma eradication was achieved in 9 of 9 (100%) culture-positive evaluable subjects who received clarithromycin vs 4 of 4 (100%) who received erythromycin; eradication rates based on PCR alone were 19 of 22 (86%) vs 24 of 26 (92%) for clarithromycin and erythromycin, respectively (however, significance of a positive PCR assay after treatment is unknown)
Wubbel et al. 446 Children Azithromycin (5 days) vs erythromycin (10 days) or amoxicillin-clavulanate (10 days) 12 Culture, PCR, serology All subjects with mycoplasma infection were classified as having clinical cure after treatment with any agent; however, 1 subject in the azithromycin group had a positive follow-up culture and 1 had a positive PCR
Harris et al. 180 Children Azithromycin (5 days) vs erythromycin (10 days) or amoxicillin-clavulanate (10 days) 124 Culture, PCR, serology Mycoplasma eradication was achieved in 14 of 14 (100%) culture-positive evaluable subjects who received azithromycin vs 4 of 6 (67%) with erythromycin and 0 of 1 with amoxicillin-clavulanate
Schonwald et al. 368 Adults Azithromycin (3 vs 5 days) 43 Serology A 1.5-g total dose of azithromycin was equally effective when administered as a 3- or a 5-day regimen and resulted in clinical cure for all mycoplasmal infections that were encountered
Schonwald et al. 367 Adults Azithromycin (single dose vs 3 days) 24 Serology A single 1.5-g dose of azithromycin was equally effective as a 3-day regimen for treatment of mycoplasmal infection
Schonwald et al. 366 Adults Azithromycin (5 days) vs erythromycin (10 days) 55 Serology Therapeutic success was achieved for all subjects with mycoplasma infection who received either drug
Kaku et al. 216 Adults Roxithromycin 13 Culture, serology Clinical efficacy was documented in 12 of 13 (93%) cases; a bacteriological effect was achieved in 4 of 6 (67%) cases
Patel et al. 322 Adults Moxifloxacin (10 days) 29 Culture, serology A bacteriological response occurred in 27 of 29 (93%) subjects with mycoplasmal infection
Finch et al. 140 Adults Moxifloxacin (7-14 days) vs amoxicillin-clavulanate with or without clarithromycin (7-14 days) 30 Serology Clinical success was documented 21-28 days after the end of treatment in 12 of 13 (93%) subjects who received moxifloxacin and in 16 of 17 (94%) who received the comparator agents
Plouffe et al. 326 Adults Ofloxacin vs standard treatments with a variety of agents for different durations 38 Serology Clinical improvement or cure occurred in 13 of 14 (88%) subjects who were serologically positive for mycoplasma and in 20 of 22 (91%) who received various other treatments
File et al. 138 Adults Gemifloxacin (7 days) vs trovafloxacin (7 days) 88 Culture, serology Clinical success for mycoplasmal infections was documented in 41 of 44 (94%) subjects who received gemifloxacin and in 38 of 40 (95%) who received trovafloxacin
File et al. 139 Adults Levofloxacin (7-14 days) vs ceftriaxone (7-14 days) or cefuroxime (7-14 days) 41 Serology Clinical improvement or cure was documented in 19 of 19 (100%) subjects treated with levofloxacin and in 22 of 22 (100%) who received either ceftriaxone or cefuroxime; the apparent response to cephalosporins may represent self-limited disease or the presence of an undiagnosed copathogen
Carbon et al. 57 Adults Telithromycin (7-10 days) 8 Serology All eight patients with mycoplasmal infection had clinical cure
Pullman et al. 336 Adults Telithromycin, trovafloxacin 93 Serology, culture, PCR Clinical cure was documented in 4 of 4 subjects with mycoplasmal infection who received telithromycin and in 5 of 5 who received trovafloxacin

Newer macrolides are generally preferred over erythromycin due to their greater tolerability, once- or twice-daily dosing requirements, and shorter treatment duration in the case of azithromycin, even though their costs are considerably greater. The extremely high potency of azithromycin against M. pneumoniae and its long half-life probably account for its ability to cure M. pneumoniae infections with very short treatment courses, and possibly even with a single 1.5-g dose, despite the relatively slow growth of the organism (367, 368). Overall, azithromycin and the investigational ketolide cethromycin (ABT-773) are the most potent drugs against M. pneumoniae in terms of MICs (436). Recently published clinical trials evaluating the investigational ketolide telithromycin alone (57) or in comparison with the fluoroquinolone trovafloxacin (336) for treatment of pneumonia in adults demonstrated that this ketolide produced clinical cure rates of 100 and 93.3%, respectively, for persons whose infection was due to M. pneumoniae.

Some recent studies evaluating fluoroquinolones for treatment of community-acquired pneumonias in adults have included diagnostic tests, primarily serology, for M. pneumoniae (138-140, 322, 326). The general conclusions of these studies are that newer fluoroquinolones such as levofloxacin and moxifloxacin are effective clinically against M. pneumoniae infections, as one might expect from their in vitro potencies. Specific treatment dosage recommendations and therapeutic regiments for M. pneumoniae infections in adults and children are provided in reference texts (433).

Relatively little data are available regarding outcomes of antimicrobial treatment of severely ill children or adults requiring hospitalization for M. pneumoniae pneumonia or treatment of immunosuppressed persons with M. pneumoniae infection. Eradication of M. pneumoniae from persons with immunosuppression can be extremely difficult, requiring prolonged therapy, even when the organisms are susceptible to the expected agents. This difficulty highlights the fact that mycoplasmas are inhibited but not killed by most commonly used antimicrobial agents in concentrations achievable in vivo.

Limited information from case reports suggests that high-dose steroid therapy may be effective in reversing neurological symptoms in children with complicated M. pneumoniae infection (175), and some clinicians recommend use of steroids in combination with an antibiotic that can penetrate the CNS, such as doxycycline or chloramphenicol (105, 208). Azithromycin and clarithromycin are commonly used for M. pneumoniae respiratory infections, and both have been used successfully in M. pneumoniae CNS infections, even though macrolides penetrate poorly into the CNS (175, 384). A recent review of steroid therapy in M. pneumoniae CNS cases suggests that it is often beneficial, particularly in severe cases. Of 14 patients with severe M. pneumoniae CNS disease, 11 (78%) were reported to have a complete or near complete recovery, a better outcome than reported for an earlier series of patients who did not receive steroids (58). The value of using steroids to treat Stevens-Johnson syndrome caused by M. pneumoniae has not been clearly established (257).

Both plasmapheresis and intravenous immunoglobulin therapy might be considered if steroid therapy is ineffective for cases of acute disseminated encephalomyelitis, which is considered to have an important immune component (384). A trial of intravenous immunoglobulin in a critically ill patient with encephalitis and M. pneumoniae pneumonia was associated with neurological improvement within 48 h of treatment (361). A patient with M. pneumoniae infection suffering from bilateral optic neuritis as well as acute Guillain-Barré syndrome recovered after plasmapheresis (325). A survey of commercial intravenous immunoglobulin therapy preparations found that most preparations had significant activity against M. pneumoniae (246).

Antibiotic therapy for persons with asthma and documented infection with M. pneumoniae or other organisms that are amenable to such treatments may prove useful. However, it should be emphasized that antibiotic administration should ideally be reserved for those asthmatic persons who are actually shown to harbor the organism.

A number of trials of prophylactic antibiotics to control outbreaks of M. pneumoniae disease have been performed, with varied success. While some earlier studies used tetracyclines (212), recent investigations used the newer macrolides. Azithromycin has been used effectively for prophylaxis against clinical infection with M. pneumoniae during outbreaks (197, 228) with a 75% protective efficacy against illnesses resulting in physician visits among hospital employees according to one study (197). Weekly oral azithromycin (500 mg) had a 64% protective efficacy based on data from serological tests against M. pneumoniae infections in U.S. marines (169). These findings support the use of prophylactic antibiotics that are highly active against M. pneumoniae in outbreak settings.

VACCINES

Interest in development of a vaccine for M. pneumoniae has been evident since the early 1960s, soon after the organism was successfully identified and isolated in culture and much of its epidemiology had become apparent. Interest was fostered by the lack of natural protective immunity following primary infection, prolonged carriage, and propensity for outbreaks of infections in military camps, schools, and hospitals. Development of a vaccine also seemed promising in view of the facts that the organism is rather homogeneous antigenically and there appears to be some protection against reinfection (81, 85, 135, 136). Animal models have been used extensively to improve understanding of the natural immune response to M. pneumoniae infection and how it might be modified by immunization (17-20, 75-77, 123, 153, 449, 450). Ellison et al. (123) provided an in-depth discussion of protective antigens, protective immune mechanisms, and prior vaccine strategies for M. pneumoniae, summarizing the work performed in this area through the early 1990s.

Considering that attachment and initiation of local damage at the cellular and subcellular levels are responsible for M. pneumoniae disease, the logical vaccine strategy is to prevent attachment and thereby prevent initiation of disease. The initial studies with human volunteers used formalin-inactivated vaccines, but their protective efficacy results were generally disappointing for a variety of reasons that have been explored in depth by Ellison et al. (123). Perhaps the most significant event related to volunteer immunizations was the experience of some immunized volunteers who developed more severe illness after experimental challenge with live mycoplasmas. This occurrence has also been documented in animal models after administration of inactivated as well as component vaccines (77, 79, 206), signifying that immunization sensitizes the host in some way through a cell-mediated immune response. Development of live attenuated vaccines administered intranasally or by aerosol inhalation in animal models showed some protective efficacy against disease upon rechallenge with virulent strains, suggesting that stimulation of local mucosal defense mechanisms is important (123, 135, 449). Development of a serum antibody response after mucosal immunization may or may not occur, depending on the immunogen (123, 135, 450). Unfortunately, the live attenuated vaccines never made it to human use due to concern over residual virulence of the vaccine strain of M. pneumoniae. Similar problems were encountered with temperature-sensitive mutant vaccines (51, 172, 388, 389). Other vaccine candidates have included acellular protein and polysaccharide components and recombinant DNA (50, 52, 123, 204, 206). While the importance of the P1 adhesin in mediating M. pneumoniae cytadherence and initiation of disease cannot be denied, animal studies using P1 as a vaccine antigen have not demonstrated protective efficacy (76, 204-206). Experimental animal studies involving mice (449, 450) hamsters (18, 75-77), guinea pigs (206), and chimpanzees (21, 153) have continued through the 1980s and 1990s, but to our knowledge there have been no recent clinical trials in humans with any newer versions of M. pneumoniae vaccines, and there is no indication that any type of vaccine will be approved for use against M. pneumoniae any time soon.

FUTURE NEEDS

Multifaceted approaches for detection and characterization of M. pneumoniae and its associated diseases by using PCR, serology, and culture, augmented by knowledge obtained from the complete genome sequence, have been applied in epidemiological investigations, animal models of disease, evaluation of diagnostic reagents, and clinical trials of antimicrobial agents. As a result, our understanding of this organism's cell biology, interactions with host cells, mechanisms of disease production, evasion of host defenses, disease transmission, and contribution to chronic lung diseases and of the efficacy of new antimicrobial treatments have improved, with much knowledge accumulated during the past decade. Despite these many advances, much is still unknown about this tiny bacterium, which is among the smallest of all free-living life forms. Most mycoplasma infections in clinical settings never have a microbiological diagnosis because rapid, sensitive, specific, and reasonably priced methods for its direct detection are not readily available in many physician offices or hospital laboratories. From a practical standpoint, serological tests are the only means by which M. pneumoniae infections are diagnosed on a wide scale, and this method has a number of limitations. A reliable and user-friendly amplified or nonamplified method for detection of the mycoplasma or its nucleic acid in clinical specimens would be of immense importance for patient diagnosis and management, for furthering knowledge of a potential role in chronic lung disease, and for evaluation of microbiological efficacy in clinical trials of antibiotics. Development of a safe vaccine that offers protective immunity might also go a long way towards reducing the extent of M. pneumoniae infections, particularly in-high risk populations such as those in the military, schools, hospitals, and other institutions where large numbers of people dwell in close proximity.

Acknowledgments

The use of trade names is for identification only and does not constitute endorsement by the Public Health Service or by the U.S. Department of Health and Human Resources.

REFERENCES

  • 1.Abele-Horn, M., U. Busch, H. Nitschko, E. Jacobs, R. Bax, F. Pfaff, B. Schaffer, and J. Heesemann. 1998. Molecular approaches to diagnosis of pulmonary diseases due to Mycoplasma pneumoniae. J. Clin. Microbiol. 36:548-551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Abele-Horn, M., W. Franck, U. Busch, H. Nitschko, R. Roos, and J. Heesemann. 1998. Transverse myelitis associated with Mycoplasma pneumoniae infection. Clin. Infect. Dis. 26:909-912. [DOI] [PubMed] [Google Scholar]
  • 3.Abramovitz, P., P. Schvartzman, D. Harel, I. Lis, and Y. Naot. 1987. Direct invasion of the central nervous system by Mycoplasma pneumoniae: a report of two cases. J. Infect. Dis. 155:482-487. [DOI] [PubMed] [Google Scholar]
  • 4.Agustin, E. T., V. Gill, and B. A. Cunha. 1994. Mycoplasma pneumoniae meningoencephalitis complicated by diplopia. Heart Lung 23:436-437. [PubMed] [Google Scholar]
  • 5.Alexander, E. R., H. M. Foy, G. E. Kenny, R. A. Kronmal, R. McMahan, E. R. Clarke, W. A. MacColl, and J. T. Grayston. 1966. Pneumonia due to Mycoplasma pneumoniae. Its incidence in the membership of a co-operative medical group. N. Engl. J. Med. 275:131-136. [DOI] [PubMed] [Google Scholar]
  • 6.Alexander, T. S., L. D. Gray, J. A. Kraft, D. S. Leland, M. T. Nikaido, and D. H. Willis. 1996. Performance of Meridian ImmunoCard Mycoplasma test in a multicenter clinical trial. J. Clin. Microbiol. 34:1180-1183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Ali, N. J., M. Sillis, B. E. Andrews, P. F. Jenkins, and B. D. Harrison. 1986. The clinical spectrum and diagnosis of Mycoplasma pneumoniae infection. Q. J. Med. 58:241-251. [PubMed] [Google Scholar]
  • 8.Almagor, M., I. Kahane, and S. Yatziv. 1984. Role of superoxide anion in host cell injury induced by Mycoplasma pneumoniae infection. A study in normal and trisomy 21 cells. J. Clin. Investig. 73:842-847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Al-Mateen, M., M. Gibbs, R. Dietrich, W. G. Mitchell, and J. H. Menkes. 1988. Encephalititis lethargica-like illness in a girl with mycoplasma infection. Neurology 38:1155-1158. [DOI] [PubMed] [Google Scholar]
  • 10.Andrews, P. A., C. M. Lloyd, M. C. Webb, and S. H. Sacks. 1994. Acute interstitial nephritis associated with Mycoplasma pneumoniae infection. Nephrol. Dial. Transplant. 9:564-566. [DOI] [PubMed] [Google Scholar]
  • 11.Arav-Boger, R., A. Assia, Z. Spirer, Y. Bujanover, and S. Reif. 1995. Cholestatic hepatitis as a main manifestation of Mycoplasma pneumoniae infection. J. Pediatr. Gastroenterol. Nutr. 21:459-460. [DOI] [PubMed] [Google Scholar]
  • 12.Arnold, S. E. 1987. Psychosis and Mycoplasma pneumoniae. Hillside J. Clin. Psychiatr. 9:231-235. [PubMed] [Google Scholar]
  • 13.Aubert, G., B. Pozzetto, O. G. Gaudin, J. Hafid, A. D. Mbida, and A. Ros. 1992. Evaluation of five commercial tests: complement fixation, microparticle agglutination, indirect immunofluorescence, enzyme-linked immunosorbent assay and latex agglutination, in comparison to immunoblotting for Mycoplasma pneumoniae serology. Ann. Biol. Clin. (Paris) 50:593-597. [PubMed] [Google Scholar]
  • 14.Aubert, G., B. Pozzetto, J. Hafid, and O. G. Gaudin. 1992. Immunoblotting patterns with Mycoplasma pneumoniae of serum specimens from infected and non-infected subjects. J. Med. Microbiol. 36:341-346. [DOI] [PubMed] [Google Scholar]
  • 15.Balish, M. F., and D. C. Krause. 2002. Cytadherence and the cytoskeleton, p. 491-518. In S. Razin and R. Herrman (ed.), Molecular biology and pathogenicity of mycoplasmas. Kluwer Academic/Plenum Publishers, New York, N.Y.
  • 16.Barile, M. F. 1979. Mycoplasma-tissue cell interactions, p. 425-474. In J. G. Tully and R. F. Whitcomb (ed.), The mycoplasmas II. Human and animal mycoplasmas, vol. 2. Academic Press, New York, N.Y.
  • 17.Barile, M. F., D. K. Chandler, H. Yoshida, M. W. Grabowski, R. Harasawa, and S. Razin. 1988. Parameters of Mycoplasma pneumoniae infection in Syrian hamsters. Infect. Immun. 56:2443-2449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Barile, M. F., D. K. Chandler, H. Yoshida, M. W. Grabowski, and S. Razin. 1988. Hamster challenge potency assay for evaluation of Mycoplasma pneumoniae vaccines. Infect. Immun. 56:2450-2457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Barile, M. F., M. W. Grabowski, K. Kapatais-Zoumbois, B. Brown, P. C. Hu, and D. K. Chandler. 1994. Protection of immunized and previously infected chimpanzees challenged with Mycoplasma pneumoniae. Vaccine 12:707-714. [DOI] [PubMed] [Google Scholar]
  • 20.Barile, M. F., M. W. Grabowski, P. J. Snoy, and D. K. Chandler. 1987. Superiority of the chimpanzee animal model to study the pathogenicity of known Mycoplasma pneumoniae and reputed mycoplasma pathogens. Isr. J. Med. Sci. 23:556-560. [PubMed] [Google Scholar]
  • 21.Barile, M. F., K. Kapatais-Zoumbos, P. Snoy, M. W. Grabowski, M. Sneller, L. Miller, and D. K. Chandler. 1994. Experimentally induced septic arthritis in chimpanzees infected with Mycoplasma hominis. Mycoplasma pneumoniae, and Ureaplasma urealyticum. Clin. Infect. Dis. 18:694-703. [DOI] [PubMed] [Google Scholar]
  • 22.Barker, C. E., M. Sillis, and T. G. Wreghitt. 1990. Evaluation of Serodia Myco II particle agglutination test for detecting Mycoplasma pneumoniae antibody: comparison with mu-capture ELISA and indirect immunofluorescence. J. Clin. Pathol. 43:163-165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Bar Meir, E., H. Amital, Y. Levy, A. Kneller, Y. Bar-Dayan, and Y. Shoenfeld. 2000. Mycoplasma-pneumoniae-induced thrombotic thrombocytopenic purpura. Acta Haematol. 103:112-115. [DOI] [PubMed] [Google Scholar]
  • 24.Barrett-Conner, E. 1972. Anemia and infection. Am. J. Med. 52:242-253. [DOI] [PubMed] [Google Scholar]
  • 25.Bartlett, J. G., S. F. Dowell, L. A. Mandell, T. M. File, S. M. Musher, and M. J. FIne. 2000. Practice guidelines for the management of community acquired pneumonia in adults. Clin. Infect. Dis. 31:347-382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Baseman, J. B. 1993. The cytadhesins of Mycoplasma pneumoniae and M. genitalium, p. 243-259. In S. Rottem and I. Kahane (ed.), Subcellular biochemistry. Plenum Press, New York, N.Y. [DOI] [PubMed]
  • 27.Baseman, J. B., M. Banai, and I. Kahane. 1982. Sialic acid residues mediate Mycoplasma pneumoniae attachment to human and sheep erythrocytes. Infect. Immun. 38:389-391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Baseman, J. B., M. Lange, N. L. Criscimagna, J. A. Giron, and C. A. Thomas. 1995. Interplay between mycoplasmas and host target cells. Microb. Pathog. 19:105-116. [DOI] [PubMed] [Google Scholar]
  • 29.Baseman, J. B., J. Morrison-Plummer, D. Drouillard, B. Puleo-Scheppke, V. V. Tryon, and S. C. Holt. 1987. Identification of a 32-kilodalton protein of Mycoplasma pneumoniae associated with hemadsorption. Isr. J. Med. Sci. 23:474-479. [PubMed] [Google Scholar]
  • 30.Baseman, J. B., S. P. Reddy, and S. F. Dallo. 1996. Interplay between mycoplasma surface proteins, airway cells, and the protean manifestations of mycoplasma-mediated human infections. Am. J. Respir. Crit. Care Med. 154:S137-S144. [DOI] [PubMed] [Google Scholar]
  • 31.Baseman, J. B., and J. G. Tully. 1997. Mycoplasmas: sophisticated, reemerging, and burdened by their notoriety. Emerg. Infect. Dis. 3:21-32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Bebear, C., M. Dupon, H. Renaudin, and B. de Barbeyrac. 1993. Potential improvements in therapeutic options for mycoplasmal respiratory infections. Clin. Infect. Dis. 17(Suppl. 1):S202-S207. [DOI] [PubMed] [Google Scholar]
  • 33.Bebear, C., and J. A. Robertson. 1996. Determination of minimal inhibitory concentration, p. 189-197. In J. G. Tully and S. Razin (ed.), Molecular and diagnostic procedures in mycoplasmology, vol. II. Academic Press, New York, N.Y.
  • 34.Bebear, C. M., and C. Bebear. 2002. Antimycoplasmal agents, p. 545-566. In S. Razin and R. Herrmann (ed.), Molecular biology and pathogenicity of mycoplasmas. Kluwer Academic/Plenum Publishers, New York, N.Y.
  • 35.Bebear, C. M., H. Renaudin, A. Boudjadja, and C. Bebear. 1998. In vitro activity of BAY-8039, a new fluoroquinolone, against mycoplasmas. Antimicrob. Agents Chemother. 42:703-704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Bebear, C. M., H. Renaudin, T. Schaeverbeke, F. LeBlanc, and C. Bebear. 2000. Comparative activities of telithromycin (HMR 3647), levofloxacin and other antimicrobial agents against human mycoplasmas. Antimicrob. Agents Chemother. 47:1980-1982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Bebear, C. M., J. Renaudin, A. Charron, B. Renaudin, B. de Barbeyrac, T. Schaeverbeke, and C. Bebear. 1999. Mutations in the gyrA. parC, and parE genes associated with fluoroquinolone resistance in clinical isolates of Mycoplasma hominis. Antimicrob. Agents Chemother. 43:954-956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Becton, D. L., H. S. Friedman, J. Kurtzberg, S. Chaffee, J. M. Falletta, and T. R. Kinney. 1986. Severe Mycoplasma pneumonia in three sisters with sickle cell disease. Pediatr. Hematol. Oncol. 3:259-265. [DOI] [PubMed] [Google Scholar]
  • 39.Behan, P. O., R. G. Feldman, J. M. Segerra, and I. T. Draper. 1986. Neurological aspects of mycoplasmal infection. Acta Neurol. Scand. 74:314-322. [DOI] [PubMed] [Google Scholar]
  • 40.Benisch, B. M., A. Fayemi, and M. A. Gerber. 1972. Mycoplasmal pneumonia in a patient with rheumatic heart disease. Am. J. Clin. Pathol. 58:343-348. [DOI] [PubMed] [Google Scholar]
  • 41.Berger, R. P., and R. M. Wadowksy. 2000. Rhabdomyolysis associated with infection by Mycoplasma pneumoniae: a case report. Pediatrics 105:433-436. [DOI] [PubMed] [Google Scholar]
  • 42.Berkovich, S., S. J. Millian, and R. D. Snyder. 1970. The association of viral and mycoplasma infections with reurrence of wheezing in the asthmatic child. Ann. Allergy 28:43-49. [PubMed] [Google Scholar]
  • 43.Bernet, C., M. Garret, B. de Barbeyrac, C. Bebear, and J. Bonnet. 1989. Detection of Mycoplasma pneumoniae by using the polymerase chain reaction. J. Clin. Microbiol. 27:2492-2496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Beskind, D. L., and S. M. Keim. 1994. Choreoathetotic movement disorder in a boy with Mycoplasma pneumoniae encephalitis. Ann. Emerg. Med. 23:1375-1378. [DOI] [PubMed] [Google Scholar]
  • 45.Biberfield, G., T. Johnsson, and J. Johnsson. 1965. Studies on Mycoplasma pneumoniae infection in Sweden. Acta Pathol. Microbiol. Scand. 63:469-475. [DOI] [PubMed] [Google Scholar]
  • 46.Blackmore, T. K., M. Reznikov, and D. L. Gordon. 1995. Clinical utility of the polymerase chain reaction to diagnose Mycoplasma pneumoniae infection. Pathology 27:177-181. [DOI] [PubMed] [Google Scholar]
  • 47.Block, S., J. Hedrick, M. R. Hammerschlag, G. H. Cassell, and J. C. Craft. 1995. Mycoplasma pneumoniae and Chlamydia pneumoniae in pediatric community-acquired pneumonia: comparative efficacy and safety of clarithromycin vs. erythromycin ethylsuccinate. Pediatr. Infect. Dis. J. 14:471-477. [DOI] [PubMed] [Google Scholar]
  • 48.Brouard, J., F. Freymuth, F. Toutain, N. Bach, A. Vabret, S. Gouarin, J. Petitjean, and J. F. Duhamel. 2002. Role of viral infections and Chlamydia pneumoniae and Mycoplasma pneumoniae infections in asthma in infants and young children. Epidemiologic study of 118 children. Arch. Pediatr. 9(Suppl. 3):365s-371s. [DOI] [PubMed] [Google Scholar]
  • 49.Brouard, J., J. Petitjean, F. Freymuth, and J. F. Duhamel. 1989. Mycoplasma pneumoniae respiratory infection in a child presenting with positive HIV 1 serology. Arch. Fr. Pediatr. 46:155-156. [PubMed] [Google Scholar]
  • 50.Brunner, H. 1981. Protective efficacy of Mycoplasma pneumoniae polysaccharides. Isr. J. Med. Sci. 17:678-681. [PubMed] [Google Scholar]
  • 51.Brunner, H., H. Greenberg, W. D. James, R. L. Horswood, and R. M. Chankock. 1973. Decreased virulence and protective effect of genetically stable temperature sensitive mutants of Mycoplasma pneumoniae. Ann. N. Y. Acad. Sci. 225:436-452. [Google Scholar]
  • 52.Brunner, H., and B. Prescott. 1982. Effect of Mycoplasma pnuemoniae polysaccharides and glycolipids on prophylaxis of experimental disease. Semin. Infect. Dis. 4:190-197. [Google Scholar]
  • 53.Buck, G. E., L. C. O'Hara, and J. T. Summersgill. 1992. Rapid, sensitive detection of Mycoplasma pneumoniae in simulated clinical specimens by DNA amplification. J. Clin. Microbiol. 30:3280-3283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Buscho, R. O., D. Saxtan, P. S. Shultz, E. Finch, and M. A. Mufson. 1978. Infections with viruses and Mycoplasma pneumoniae during exacerbations of chronic bronchitis. J. Infect. Dis. 137:377-383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Reference deleted.
  • 56.Campbell, T. A., P. S. Strong, and G. S. Grier. 1943. Primary atypical pneumonia: two hundred cases at Ft. Eustin, Virginia. JAMA 122:723-729. [Google Scholar]
  • 57.Carbon, C., S. Moola, I. Velancsics, B. Leroy, M. Rangaraju, and P. Decosta. 2003. Telithromycin 800 mg once daily for seven to ten days is an effective and well-tolerated treatment for community-acquired pneumonia. Clin. Microbiol. Infect. 9:691-703. [DOI] [PubMed] [Google Scholar]
  • 58.Carpenter, T. C. 2002. Corticosteroids in the treatment of etriatal encephalitis complicating Mycoplasma pneumoniae pneumonia: possible benefit of intravenous immunoglobulin. Pediatr. Infect. Dis. J. 20:534-545. [DOI] [PubMed] [Google Scholar]
  • 59.Carstensen, H., and K. O. Nilsson. 1987. Neurological complications associated with Mycoplasma pneumoniae infection in children. Neuropediatrics 18:57-58. [DOI] [PubMed] [Google Scholar]
  • 60.Cassell, G. H. 1998. Infectious causes of chronic inflammatory diseases and cancer. Emerg. Infect. Dis. 4:475-487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Cassell, G. H., W. A. Clyde, Jr., and J. K. Davis. 1985. Mycoplasma respiratory infections, p. 65-106. In S. Razin and M. F. Barile (ed.), The mycoplasmas, vol. 4. Academic Press, New York, N.Y.
  • 62.Cassell, G. H., and B. C. Cole. 1981. Mycoplasmas as agents of human disease. N. Engl. J. Med. 304:80-89. [DOI] [PubMed] [Google Scholar]
  • 63.Cassell, G. H., J. Drnec, K. B. Waites, M. S. Pate, L. B. Duffy, H. L. Watson, and J. C. McIntosh. 1991. Efficacy of clarithromycin against Mycoplasma pneumoniae. J. Antimicrob. Chemother. 27(Suppl. A):47-59. [DOI] [PubMed] [Google Scholar]
  • 64.Cassell, G. H., G. Gambill, and L. B. Duffy. 1996. ELISA in respiratory infections of humans, p. 123-136. In J. G. Tully and S. Razin (ed.), Molecular and diagnostic procedures in diagnostic mycoplasmology. Academic Press, New York, N.Y.
  • 65.Chan, E. D., and C. H. Welsh. 1995. Fulminant Mycoplasma pneumoniae pneumonia. West. J. Med. 162:133-142. [PMC free article] [PubMed] [Google Scholar]
  • 66.Chanock, R. M., M. K. Cook, H. H. Fox, R. H. Parrott, and B. J. Huebner. 1960. Serologic evidence of infection with Eaton agent in lower respiratory illness of childhood. N. Engl. J. Med. 262:648-654. [DOI] [PubMed] [Google Scholar]
  • 67.Chanock, R. M., L. Dienes, M. D. Eaton, D. G. Edward, E. A. Freundt, L. Hayflick, J. F. P. Hers, K. E. Jensen, C. Liu, B. P. Marmion, M. A. Mufson, P. F. Smith, N. L. Somerson, and D. Taylor-Robinson. 1963. Mycoplasma pneumoniae: proposed nomenclature for atypical pneumonia organism (Eaton agent). Science 140:662. [DOI] [PubMed] [Google Scholar]
  • 68.Chanock, R. M., L. Hayflick, and M. F. Barile. 1962. Growth on artificial medium of an agent associated with atypical pneumonia and its identification as a PPLO. Proc. Natl. Acad. Sci. USA 48:41-49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Chanock, R. M., D. M. Rifkind, H. M. Kravetz, V. Knight, and K. M. Johnson. 1961. Respiratory disease in volunteers infected with Eaton agent: a preloiminary report. Proc. Natl. Acad. Sci. USA 47:887-890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Cherry, J. D. 1993. Anemia and mucocutaneous lesions due to Mycoplasma pneumoniae infections. Clin. Infect. Dis. 17(Suppl. 1):S47-S51. [DOI] [PubMed] [Google Scholar]
  • 71.Cherry, J. D., and R. C. Welliver. 1976. Mycoplasma pneumoniae infections of adults and children. West. J. Med. 125:47-55. [PMC free article] [PubMed] [Google Scholar]
  • 72.Chester, A., J. Kane, and V. Garagusi. 1975. Mycoplasma pneumonia with bilateral pleural effusions. Am. Rev. Respir. Dis. 112:451-456. [DOI] [PubMed] [Google Scholar]
  • 73.Chryssanthopoulos, C., M. Eboriadou, K. Monti, V. Soubassi, and K. Sava. 2001. Fatal disseminated intravascular coagulation caused by Mycoplasma pneumoniae.Pediatr. Infect. Dis. J. 20:634-635. [DOI] [PubMed] [Google Scholar]
  • 74.Chu, H. W., M. Kraft, J. E. Krause, M. D. Rex, and R. J. Martin. 2000. Substance P and its receptor neurokinin 1 expression in asthmatic airways. J. Allergy Clin. Immunol. 106:713-722. [DOI] [PubMed] [Google Scholar]
  • 75.Cimolai, N., A. C. Cheong, B. J. Morrison, and G. P. Taylor. 1996. Mycoplasma pneumoniae reinfection and vaccination: protective oral vaccination and harmful immunoreactivity after re-infection and parenteral immunization. Vaccine 14:1479-1483. [DOI] [PubMed] [Google Scholar]
  • 76.Cimolai, N., D. G. Mah, G. P. Taylor, and B. J. Morrison. 1995. Bases for the early immune response after rechallenge or component vaccination in an animal model of acute Mycoplasma pneumoniae pneumonitis. Vaccine 13:305-309. [DOI] [PubMed] [Google Scholar]
  • 77.Cimolai, N., G. P. Taylor, D. Mah, and B. J. Morrison. 1992. Definition and application of a histopathological scoring scheme for an animal model of acute Mycoplasma pneumoniae pulmonary infection. Microbiol. Immunol. 36:465-478. [DOI] [PubMed] [Google Scholar]
  • 78.Cimolai, N., D. Wensley, M. Seear, and E. T. Thomas. 1995. Mycoplasma pneumoniae as a cofactor in severe respiratory infections. Clin. Infect. Dis. 21:1182-1185. [DOI] [PubMed] [Google Scholar]
  • 79.Clyde, W. A., Jr. 1971. Immunopathology of experimental Mycoplasma pneumoniae disease. Infect. Immun. 4:757-763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Clyde, W. A., Jr. 1979. Mycoplasma pneumonaie infections of man, p. 275-306. In J. G. Tully and R. F. Whitcomb (ed.), The mycoplasmas II. Human and animal mycoplasmas, vol. II. Academic Press, New York, N.Y.
  • 81.Clyde, W. A., Jr. 1971. Mycoplasma pneumoniae pneumonia. Mil. Med. 136:20-22. [PubMed] [Google Scholar]
  • 82.Collier, A. M. 1983. Attachment by mycoplasmas and its role in disease. Rev. Infect. Dis. 5(Suppl. 4):S685-S691. [DOI] [PubMed] [Google Scholar]
  • 83.Collier, A. M., and J. B. Baseman. 1973. Organ culture techniques with mcoplasmas. Ann. N. Y. Acad. Sci. 225:277-289. [Google Scholar]
  • 84.Corsaro, D., M. Valassina, D. Venditti, V. Venard, A. Le Faou, and P. E. Valensin. 1999. Multiplex PCR for rapid and differential diagnosis of Mycoplasma pneumoniae and Chlamydia pneumoniae in respiratory infections. Diagn. Microbiol. Infect. Dis. 35:105-108. [DOI] [PubMed] [Google Scholar]
  • 85.Couch, R. B. 1964. Infection with artificially propagated Eaton agent (Mycoplasma pneumoniae): implications for development of attenuated vaccine for cold agglutinin-positive pneumonia. JAMA 187:442-447. [PubMed] [Google Scholar]
  • 86.Cousin-Allery, A., A. Charron, B. de Barbeyrac, G. Fremy, J. Skov Jensen, H. Renaudin, and C. Bebear. 2000. Molecular typing of Mycoplasma pneumoniae strains by PCR-based methods and pulsed-field gel electrophoresis. Application to French and Danish isolates. Epidemiol. Infect. 124:103-111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Dallo, S. F., and J. B. Baseman. 2000. Intracellular DNA replication and long-term survival of pathogenic mycoplasmas. Microb. Pathog. 29:301-309. [DOI] [PubMed] [Google Scholar]
  • 88.Dallo, S. F., A. Chavoya, and J. B. Baseman. 1990. Characterization of the gene for a 30-kilodalton adhesion-related protein of Mycoplasma pneumoniae. Infect. Immun. 58:4163-4165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Dallo, S. F., T. R. Kannan, M. W. Blaylock, and J. B. Baseman. 2002. Elongation factor Tu and E1 beta subunit of pyruvate dehydrogenase complex act as fibronectin binding proteins in Mycoplasma pneumoniae.Mol Microbiol. 46:1041-1051. [DOI] [PubMed] [Google Scholar]
  • 90.Dallo, S. F., C. J. Su, J. R. Horton, and J. B. Baseman. 1988. Identification of P1 gene domain containing epitope(s) mediating Mycoplasma pneumoniae cytoadherence. J. Exp. Med. 167:718-723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Dandekar, T., B. Snel, S. Schmidt, W. Lathe, M. Suyama, M. Huynen, and P. Bork. 2002. Comparative genome analysis of the mollicutes, p. 255-278. In S. Razin and R. Herrmann (ed.), Molecular biology and pathogenesis of mycoplasmas. Kluwer Academic/Plenum Publishers, New York, N.Y.
  • 92.Davis, C. P., S. Cochran, J. Lisse, G. Buck, A. R. DiNuzzo, T. Weber, and J. A. Reinarz. 1988. Isolation of Mycoplasma pneumoniae from synovial fluid samples in a patient with pneumonia and polyarthritis. Arch. Intern. Med. 148:969-970. [PubMed] [Google Scholar]
  • 93.Daxboeck, F., G. Brunner, H. Popper, R. Krause, K. Schmid, G. J. Krejs, and C. Wenisch. 2002. A case of lung transplantation following Mycoplasma pneumoniae infection. Eur. J. Clin. Microbiol. Infect. Dis. 21:318-322. [DOI] [PubMed] [Google Scholar]
  • 94.Daxboeck, F., K. Kircher, R. Krause, H. Heinzl, C. Wenisch, and G. Stanek. 2002. Effect of age on antibody titer to Mycoplasma pneumoniae. Scand. J. Infect. Dis. 34:577-579. [DOI] [PubMed] [Google Scholar]
  • 95.Daxboeck, F., R. Krause, and C. Wenisch. 2003. Laboratory diagnosis of Mycoplasma pneumoniae infection. Clin. Microbiol. Infect. 9:263-273. [DOI] [PubMed] [Google Scholar]
  • 96.de Barbeyrac, B., C. Bernet-Poggi, F. Febrer, H. Renaudin, M. Dupon, and C. Bebear. 1993. Detection of Mycoplasma pneumoniae and Mycoplasma genitalium in clinical samples by polymerase chain reaction. Clin. Infect. Dis. 17(Suppl. 1):S83-S89. [DOI] [PubMed] [Google Scholar]
  • 97.Decanq, H. G., Jr., and F. A. Lee. 1965. Mycoplasma pneumoniae pneumonia. Massive pulmonary involvement and pleural effusion. JAMA 194:1010-1011. [DOI] [PubMed] [Google Scholar]
  • 98.Decaux, G., M. Szyper, M. Ectors, A. Cornil, and L. Franken. 1980. Central nervous system complications of Mycoplasma pneumoniae. J. Neurol. Neurosurg. Psychiatr. 43:883-887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Del Giudice, R. A., T. R. Carski, M. F. Barile, R. M. Lemcke, and J. G. Tully. 1971. Proposal for classifying human strain navel and related simian mycoplasmas as Mycoplasma primatum sp. nov. J. Bacteriol. 108:439-445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Del Giudice, R. A., R. H. Purcell, T. R. Carski, and R. M. Chanock. Characterization of a new Mycoplasma species of human origin. Int. J. Syst. Bacteriol. 24:147-153.
  • 101.Del Giudice, R. A., J. G. Tully, D. L. Rose, and R. M. Cole. 1985. Mycoplasma pirum sp. nov., a terminal structured mollicute from cell cultures. Int. J. Syst. Bacteriol. 35:285-291. [Google Scholar]
  • 102.Denny, F. W., W. A. Clyde, Jr., and W. P. Glezen. 1971. Mycoplasma pneumoniae disease: clinical spectrum, pathophysiology, epidemiology, and control. J. Infect. Dis. 123:74-92. [DOI] [PubMed] [Google Scholar]
  • 103.De Vos, M., L. Van Nimmen, and G. Baele. 1974. Disseminated intravascular coagulation during a fatal Mycoplasma pneumoniae infection. Acta Haematol. 52:120-125. [DOI] [PubMed] [Google Scholar]
  • 104.Dienes, L., and G. Edsall. 1937. Observations on the L-organisms of Klieneberger. Proc. Soc. Exp. Biol. Med. 36:740-744. [Google Scholar]
  • 105.Dionisio, D., M. Valassina, S. Mata, R. Rossetti, A. Vivarelli, F. C. Esperti, M. Benvenuti, C. Catalani, and M. Uberti. 1999. Encephalitis caused directly by Mycoplasma pneumoniae. Scand. J. Infect. Dis. 31:506-509. [DOI] [PubMed] [Google Scholar]
  • 106.Dominguez, A., S. Minguell, J. Torres, A. Serrano, J. Vidal, and L. Salleras. 1996. Community outbreak of acute respiratory infection by Mycoplasma pneumoniae. Eur. J. Epidemiol. 12:131-134. [DOI] [PubMed] [Google Scholar]
  • 107.Dorigo-Zetsma, J. W., J. Dankert, and S. A. Zaat. 2000. Genotyping of Mycoplasma pneumoniae clinical isolates reveals eight P1 subtypes within two genomic groups. J. Clin. Microbiol. 38:965-970. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Dorigo-Zetsma, J. W., R. P. Verkooyen, H. P. van Helden, H. van der Nat, and J. M. van den Bosch. 2001. Molecular detection of Mycoplasma pneumoniae in adults with community-acquired pneumonia requiring hospitalization. J. Clin. Microbiol. 39:1184-1186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Dorigo-Zetsma, J. W., B. Wilbrink, H. van der Nat, A. I. Bartelds, M. L. Heijnen, and J. Dankert. 2001. Results of molecular detection of Mycoplasma pneumoniae among patients with acute respiratory infection and in their household contacts reveals children as human reservoirs. J. Infect. Dis. 183:675-678. [DOI] [PubMed] [Google Scholar]
  • 110.Dorigo-Zetsma, J. W., S. A. Zaat, A. J. Vriesema, and J. Dankert. 1999. Demonstration by a nested PCR for Mycoplasma pneumoniae that M. pneumoniae load in the throat is higher in patients hospitalised for M. pneumoniae infection than in non-hospitalised subjects. J. Med. Microbiol. 48:1115-1122. [DOI] [PubMed] [Google Scholar]
  • 111.Dorigo-Zetsma, J. W., S. A. Zaat, P. M. Wertheim-van Dillen, L. Spanjaard, J. Rijntjes, G. van Waveren, J. S. Jensen, A. F. Angulo, and J. Dankert. 1999. Comparison of PCR, culture, and serological tests for diagnosis of Mycoplasma pneumoniae respiratory tract infection in children. J. Clin. Microbiol. 37:14-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Dowdle, W. R., J. A. Stewart, J. T. Heyward, and R. Q. Robinson. 1967. Mycoplasma pneumoniae infections in a children's population: a five-year study. Am. J. Epidemiol. 85:137-146. [DOI] [PubMed] [Google Scholar]
  • 113.Duffy, L. B., D. M. Crabb, X. Bing, and K. B. Waites. 2003. Bactericidal activity of levofloxacin against Mycoplasma pneumoniae. J. Antimicrob. Chemother. 52:527-528. [DOI] [PubMed] [Google Scholar]
  • 114.Dumke, R., I. Catrein, E. Pirkil, R. Herrmann, and E. Jacobs. 2003. Subtyping of Mycoplasma pneumoniae isolates based on extended genome sequencing and on expression profiles. Int. J. Med. Microbiol. 292:513-525. [DOI] [PubMed] [Google Scholar]
  • 115.Eaton, M. D., G. Meikejohn, and W. Van Herick. 1944. Studies on the etiology of primary atypical pneumonia: a filterable agent transmissible to cotton rats, hamsters, and chick embryos. J. Exp. Med. 79:649-667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Echevarria, J. M., P. Leon, P. Balfagon, J. A. Lopez, and M. V. Fernandez. 1990. Diagnosis of Mycoplasma pneumoniae infection by microparticle agglutination and antibody-capture enzyme-immunoassay. Eur. J. Clin. Microbiol. Infect. Dis. 9:217-220. [DOI] [PubMed] [Google Scholar]
  • 117.Edward, D., G. Meiklejohn, and E. A. Freundt. 1956. The classification and nomenclature of organisms of the pleuropneumonia group. J. Gen Microbiol. 14:197-207. [DOI] [PubMed] [Google Scholar]
  • 118.Edward, D. G. 1955. A suggested classification and nomenclature for organisms of the pleuropneumonia group. Int. Bull. Bacteriol. Nomencl. 5:85-93. [Google Scholar]
  • 119.Edward, D. G., and E. A. Freundt. 1970. Amended nomenclature for a classification of strains related to “Mycoplasma laidlawii.” J. Gen Microbiol. 62:1-2. [DOI] [PubMed] [Google Scholar]
  • 120.Edwards, E. A., Y. E. Crawford, W. E. Pierce, and R. O. Peckinpaugh. 1976. A longitudinal study of Mycoplasma pneumoniae: infections in Navy recruits by isolation and seroepidemiology. Am. J. Epidemiol. 104:556-562. [DOI] [PubMed] [Google Scholar]
  • 121.Efthimiou, J., M. E. Hodson, P. Taylor, A. G. Taylor, and J. C. Batten. 1984. Importance of viruses and Legionella pneumophila in respiratory exacerbations of young adults with cystic fibrosis. Thorax 39:150-154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.el-Khatib, M., and A. M. Lerner. 1975. Myocarditis in Mycoplasma pneumoniae pneumonia. Occurrence with hemolytic anemia and extraordinary titers of cold isohemagglutinins. JAMA 231:493-494. [DOI] [PubMed] [Google Scholar]
  • 123.Ellison, J. S., L. D. Olson, and M. F. Barile. 1992. Immunity and vaccine development, p. 491-504. In J. Maniloff, R. N. McElhaney, L. R. Finch, and J. B. Baseman (ed.), Mycoplasmas: molecular biology and pathogenesis. American Society for Microbiology, Washington, D.C.
  • 124.Emre, U., M. Bernius, P. M. Roblin, P. F. Gaerlan, J. T. Summersgill, P. Steiner, J. Schachter, and M. R. Hammerschlag. 1996. Chlamydia pneumoniae infection in patients with cystic fibrosis. Clin. Infect. Dis. 22:819-823. [DOI] [PubMed] [Google Scholar]
  • 125.Esposito, S., R. Cavagna, S. Bosis, R. Droghetti, N. Faelli, and N. Principi. 2002. Emerging role of Mycoplasma pneumoniae in children with acute pharyngitis. Eur. J. Clin. Microbiol. Infect. Dis. 21:607-610. [DOI] [PubMed] [Google Scholar]
  • 126.Esposito, S., R. Droghetti, S. Bosis, L. Claut, P. Marchisio, and N. Principi. 2002. Cytokine secretion in children with acute Mycoplasma pneumoniae infection and wheeze. Pediatr. Pulmonol. 34:122-127. [DOI] [PubMed] [Google Scholar]
  • 127.Falguera, M., A. Nogues, A. Ruiz-Gonzalez, M. Garcia, and T. Puig. 1996. Detection of Mycoplasma pneumoniae by polymerase chain reaction in lung aspirates from patients with community-acquired pneumonia. Chest 110:972-976. [DOI] [PubMed] [Google Scholar]
  • 128.Farraj, R. S., R. B. McCully, J. K. Oh, and T. F. Smith. 1997. Mycoplasma-associated pericarditis. Mayo Clin. Proc. 72:33-36. [DOI] [PubMed] [Google Scholar]
  • 129.Feder, H. M., Jr., T. Watkin, S. R. Cole, and R. Quintiliani. 1981. Severe meningoencephalitis: complicating Mycoplasma pneumoniae infection in a child. Arch. Pathol. Lab. Med. 105:619-621. [PubMed] [Google Scholar]
  • 130.Fedorko, D. P., D. D. Emery, S. M. Franklin, and D. D. Congdon. 1995. Evaluation of a rapid enzyme immunoassay system for serologic diagnosis of Mycoplasma pneumoniae infection. Diagn. Microbiol. Infect. Dis. 23:85-88. [DOI] [PubMed] [Google Scholar]
  • 131.Feikin, D. R., J. F. Moroney, D. F. Talkington, W. L. Thacker, J. E. Code, L. A. Schwartz, D. D. Erdman, J. C. Butler, and M. S. Cetron. 1999. An outbreak of acute respiratory disease caused by Mycoplasma pneumoniae and adenovirus at a federal service training academy: new implications from an old scenario. Clin. Infect. Dis. 29:1545-1550. [DOI] [PubMed] [Google Scholar]
  • 132.Feizi, T., H. Maclean, R. G. Sommerville, and J. G. Selwyn. 1967. Studies on an epidemic of respiratory disease caused by Mycoplasma pneumoniae. Br. Med. J. 1:457-460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Feldman, C., R. Anderson, A. J. Theron, G. Ramafi, P. J. Cole, and R. Wilson. 1996. Roxithromycin, clarithromycin, and azithromycin attenuate the injurious effects of bioactive phospholipids on human respiratory epithelium in vitro. Inflammation 21:655-665. [DOI] [PubMed] [Google Scholar]
  • 134.Fernald, G. W. 1972. In vitro response of human lymphocytes to Mycoplasma pneumoniae. Infect. Immun. 5:552-558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Fernald, G. W., and W. A. Clyde, Jr. 1970. Protective effect of vaccines in experimental Mycoplasma pneumoniae disease. Infect. Immun. 1:559-565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Fernald, G. W., and W. P. Glezen. 1973. Humoral and cellular immune responses to an inactivated Mycoplasma pneumoniae vaccine in children. J. Infect. Dis. 127:498-504. [DOI] [PubMed] [Google Scholar]
  • 137.Ferwerda, A., H. A. Moll, and R. de Groot. 2001. Respiratory tract infections by Mycoplasma pneumoniae in children: a review of diagnostic and therapeutic measures. Eur. J. Pediatr. 160:483-491. [DOI] [PubMed] [Google Scholar]
  • 138.File, T. M., Jr., B. Schlemmer, J. Garau, M. Cupo, and C. Young. 2001. Efficacy and safety of gemifloxacin in the treatment of community-acquired pneumonia: a randomized, double-blind comparison with trovafloxacin. J. Antimicrob. Chemother. 48:67-74. [DOI] [PubMed] [Google Scholar]
  • 139.File, T. M., Jr., J. Segreti, L. Dunbar, R. Player, R. Kohler, R. R. Williams, C. Kojak, and A. Rubin. 1997. A multicenter, randomized study comparing the efficacy and safety of intravenous and/or oral levofloxacin versus ceftriaxone and/or cefuroxime axetil in treatment of adults with community-acquired pneumonia. Antimicrob. Agents Chemother. 41:1965-1972. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Finch, R., D. Schurmann, O. Collins, R. Kubin, J. McGivern, H. Bobbaers, J. L. Izquierdo, P. Nikolaides, F. Ogundare, R. Raz, P. Zuck, and G. Hoeffken. 2002. Randomized controlled trial of sequential intravenous (i.v.) and oral moxifloxacin compared with sequential i.v. and oral co-amoxiclav with or without clarithromycin in patients with community-acquired pneumonia requiring initial parenteral treatment. Antimicrob. Agents Chemother. 46:1746-1754. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Fischman, R. A., K. E. Marschall, J. W. Kislak, and D. M. Greenbaum. 1978. Adult respiratory distress syndrome caused by Mycoplasma pneumoniae. Chest 74:471-473. [DOI] [PubMed] [Google Scholar]
  • 142.Fisher, R. S., A. W. Clark, J. S. Wolinsky, I. M. Parhad, H. Moses, and M. R. Mardiney. 1983. Postinfectious leukoencephalitis complicating Mycoplasma pneumoniae infection. Arch. Neurol. 40:109-113. [DOI] [PubMed] [Google Scholar]
  • 143.Foy, H. M. 1993. Infections caused by Mycoplasma pneumoniae and possible carrier state in different populations of patients. Clin. Infect. Dis. 17(Suppl. 1):S37-S46. [DOI] [PubMed] [Google Scholar]
  • 144.Foy, H. M., M. K. Cooney, I. Allan, and G. E. Kenny. 1979. Rates of pneumonia during influenza epidemics in Seattle, 1964 to 1975. JAMA 241:253-258. [PubMed] [Google Scholar]
  • 145.Foy, H. M., J. T. Grayston, G. E. Kenny, E. R. Alexander, and R. McMahan. 1966. Epidemiology of Mycoplasma pneumoniae infection in families. JAMA 197:859-866. [PubMed] [Google Scholar]
  • 146.Foy, H. M., G. E. Kenny, M. K. Cooney, and I. D. Allan. 1979. Long-term epidemiology of infections with Mycoplasma pneumoniae. J. Infect. Dis. 139:681-687. [DOI] [PubMed] [Google Scholar]
  • 147.Foy, H. M., G. E. Kenny, M. K. Cooney, I. D. Allan, and G. van Belle. 1983. Naturally acquired immunity to pneumonia due to Mycoplasma pneumoniae. J. Infect. Dis. 147:967-973. [DOI] [PubMed] [Google Scholar]
  • 148.Foy, H. M., G. E. Kenny, R. McMahan, A. M. Mansy, and J. T. Grayston. 1970. Mycoplasma pneumoniae pneumonia in an urban area. Five years of surveillance. JAMA 214:1666-1672. [PubMed] [Google Scholar]
  • 149.Foy, H. M., C. M. Nolan, and I. D. Allan. 1983. Epidemiologic aspects of M. pneumoniae disease complications: a review. Yale J. Biol. Med. 56:469-473. [PMC free article] [PubMed] [Google Scholar]
  • 150.Foy, H. M., C. G. Nugent, G. E. Kenny, R. McMahan, and J. T. Grayston. 1971. Repeated Mycoplasma pneumoniae pneumonia after 4 and one-half years. JAMA 216:671-672. [DOI] [PubMed] [Google Scholar]
  • 151.Foy, H. M., H. Ochs, S. D. Davis, G. E. Kenny, and R. R. Luce. 1973. Mycoplasma pneumoniae infections in patients with immunodeficiency syndromes: report of four cases. J. Infect. Dis. 127:388-393. [DOI] [PubMed] [Google Scholar]
  • 152.Fransen, H., M. Forsgren, Z. Heigl, and G. Tunevall. 1969. Studies on Mycoplasma pneumoniae in patients hospitalized with acute respiratory illness. Scand. J. Infect. Dis. 1:91-98. [DOI] [PubMed] [Google Scholar]
  • 153.Franzoso, G., P. C. Hu, G. A. Meloni, and M. F. Barile. 1994. Immunoblot analyses of chimpanzee sera after infection and after immunization and challenge with Mycoplasma pneumoniae. Infect. Immun. 62:1008-1014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Freundt, E. A., D. Taylor-Robinson, R. H. Purcell, R. M. Chanock, and F. T. Black. 1974. Proposal of Mycoplasma buccale nom. nov. and Mycoplasma faucium nom. nov. for Mycoplasma orale′types' 2 and 3, respectively. Int. J. Syst. Bacteriol. 24:252-255. [Google Scholar]
  • 155.Freymuth, F., A. Vabret, J. Brouard, F. Toutain, R. Verdon, J. Petitjean, S. Gouarin, J. F. Duhamel, and B. Guillois. 1999. Detection of viral, Chlamydia pneumoniae and Mycoplasma pneumoniae infections in exacerbations of asthma in children. J. Clin. Virol. 13:131-139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Gendrel, D. 1997. Antibiotic treatment of Mycoplasma pneumoniae infections. Pediatr. Pulmonol. Suppl. 16:46-47. [PubMed] [Google Scholar]
  • 157.Gerstenecker, B., and E. Jacobs. 1993. Development of a capture-ELISA for the specific detection of Mycoplasma pneumonaie in patient's material, p. 195-205. In I. Kahane and A. Adoni (ed.), Rapid diagnosis of mycoplasmas. Plenum Press, New York, N.Y.
  • 158.Gil, J. C., R. L. Cedillo, B. G. Mayagoitia, and M. D. Paz. 1993. Isolation of Mycoplasma pneumoniae from asthmatic patients. Ann. Allergy 70:23-25. [PubMed] [Google Scholar]
  • 159.Gillberg, C. 1980. Schizophreniform psychosis in a case of Mycoplasma pneumoniae encephalitis. J. Autism Dev. Disord. 10:153-158. [DOI] [PubMed] [Google Scholar]
  • 160.Ginaldi, L., M. De Martinis, A. D'Ostilio, L. L. Marini, M. F., M. P. Corsi, and D. Quaglino. 1999. The immune system in the elderly. I. Specific humoral immunity. Immunol. Res. 20:101-188. [DOI] [PubMed] [Google Scholar]
  • 161.Glezen, W. P., F. A. Loda, W. A. Clyde, R. J. Senior, C. I. Sheaffer, W. G. Conley, and F. W. Denny. 1971. Epidemiologic patterns of acute lower respiratory tract diseases of children in a pediatric group practice. J. Pediatr. 78:397-406. [DOI] [PubMed] [Google Scholar]
  • 162.Gnarpe, J., A. Lundback, B. Sundelof, and H. Gnarpe. 1992. Prevalence of Mycoplasma pneumoniae in subjectively healthy individuals. Scand. J. Infect. Dis. 24:161-164. [DOI] [PubMed] [Google Scholar]
  • 163.Gobel, U., G. H. Butler, and E. J. Stanbridge. 1984. Comparative analysis of mycoplasma ribosomal RNA operons. Isr. J. Med. Sci. 20:762-764. [PubMed] [Google Scholar]
  • 164.Golubjatnikov, R., V. D. Allen, M. P. Olmos-Blancarte, and S. L. Inhorn. 1975. Serologic profile of children in a Mexican highland community: prevalence of complement-fixing antibodies to Mycoplasma pneumoniae, respiratory syncytial virus and parainfluenza viruses. Am. J. Epidemiol. 101:458-464. [DOI] [PubMed] [Google Scholar]
  • 165.Goulet, M., R. Dular, J. G. Tully, G. Billowes, and S. Kasatiya. 1995. Isolation of Mycoplasma pneumoniae from the human urogenital tract. J. Clin. Microbiol. 33:2823-2825. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Granstrom, M., T. Holme, A. M. Sjogren, A. Ortqvist, and M. Kalin. 1994. The role of IgA determination by ELISA in the early serodiagnosis of Mycoplasma pneumoniae infection, in relation to IgG and mu-capture IgM methods. J. Med. Microbiol. 40:288-292. [DOI] [PubMed] [Google Scholar]
  • 167.Gray, G. C., J. D. Callahan, A. W. Hawksworth, C. A. Fisher, and J. C. Gaydos. 1999. Respiratory diseases among U. S. military personnel: countering emerging threats. Emerg. Infect. Dis. 5:379-385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Gray, G. C., L. B. Duffy, R. J. Paver, S. D. Putnam, R. J. Reynolds, and G. H. Cassell. 1997. Mycoplasma pneumoniae: a frequent cause of pneumonia among U. S. Marines in southern California. Mil. Med. 162:524-526. [PubMed] [Google Scholar]
  • 169.Gray, G. C., D. C. McPhate, M. Leinonen, G. H. Cassell, E. P. Deperalta, S. D. Putnam, J. A. Karcher, M. H. Sawyer, A. Laurila, and J. D. Connor. 1998. Weekly oral azithromycin as prophylaxis for agents causing acute respiratory disease. Clin. Infect. Dis. 26:103-110. [DOI] [PubMed] [Google Scholar]
  • 170.Gray, G. C., B. S. Mitchell, J. E. Tueller, E. R. Cross, and D. E. Amundson. 1994. Pneumonia hospitalizations in the US Navy and Marine Corps: rates and risk factors for 6,522 admissions, 1981-1991. Am. J. Epidemiol. 139:793-802. [DOI] [PubMed] [Google Scholar]
  • 171.Grayston, J. T., G. E. Kenny, H. M. Foy, R. A. Kronmal, and E. R. Alexander. 1967. Epidemiological studies of Mycoplasma pneumoniae infections in civilians. Ann. N. Y. Acad. Sci. 143:436-446. [DOI] [PubMed] [Google Scholar]
  • 172.Greenberg, H., C. M. Helms, H. Brunner, and R. M. Chanock. 1974. Asymptomatic infection of adult volunteers with a temperature sensitive mutant of Mycoplasma pneumoniae. Proc. Natl. Acad. Sci. USA 71:4015-4019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Grondahl, B., W. Puppe, A. Hoppe, I. Kuhne, J. A. Weigl, and H. J. Schmitt. 1999. Rapid identification of nine microorganisms causing acute respiratory tract infections by single-tube multiplex reverse transcription-PCR: feasibility study. J. Clin. Microbiol. 37:1-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Grullich, C., T. F. Baumert, and H. E. Blum. 2003. Acute Mycoplasma pneumoniae infection presenting as cholestatic hepatitis. J. Clin. Microbiol. 41:514-515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Gucuyener, K., F. Simsek, O. Yilmaz, and A. Serdaroglu. 2000. Methyl-prednisolone in neurologic complications of Mycoplasma pneumonia. Indian J. Pediatr. 67:467-469. [DOI] [PubMed] [Google Scholar]
  • 176.Gump, D. W., C. A. Phillips, B. R. Forsyth, K. McIntosh, K. R. Lamborn, and W. H. Stouch. 1976. Role of infection in chronic bronchitis. Am. Rev. Respir. Dis. 113:465-474. [DOI] [PubMed] [Google Scholar]
  • 177.Hardegger, D., D. Nadal, W. Bossart, M. Altwegg, and F. Dutly. 2000. Rapid detection of Mycoplasma pneumoniae in clinical samples by real-time PCR. J. Microbiol. Methods 41:45-51. [DOI] [PubMed] [Google Scholar]
  • 178.Hardy, R. D., H. S. Jafri, K. Olsen, J. Hatfield, J. Iglehart, B. B. Rogers, P. Patel, G. Cassell, G. H. McCracken, and O. Ramilo. 2002. Mycoplasma pneumoniae induces chronic respiratory infection, airway hyperreactivity, and pulmonary inflammation: a murine model of infection-associated chronic reactive airway disease. Infect. Immun. 70:649-654. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Hardy, R. D., H. S. Jafri, K. Olsen, M. Wordemann, J. Hatfield, B. B. Rogers, P. Patel, L. Duffy, G. Cassell, G. H. McCracken, and O. Ramilo. 2001. Elevated cytokine and chemokine levels and prolonged pulmonary airflow resistance in a murine Mycoplasma pneumoniae pneumonia model: a microbiologic, histologic, immunologic, and respiratory plethysmographic profile. Infect. Immun. 69:3869-3876. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Harris, J. A., A. Kolokathis, M. Campbell, G. H. Cassell, and M. R. Hammerschlag. 1998. Safety and efficacy of azithromycin in the treatment of community-acquired pneumonia in children. Pediatr. Infect. Dis. J. 17:865-871. [DOI] [PubMed] [Google Scholar]
  • 181.Hauksdottir, G. S., T. Jonsson, V. Sigurdardottir, and A. Love. 1998. Seroepidemiology of Mycoplasma pneumoniae infections in Iceland 1987-96. Scand. J. Infect. Dis. 30:177-180. [DOI] [PubMed] [Google Scholar]
  • 182.Heiskanen-Kosma, T., M. Korppi, C. Jokinen, S. Kurki, L. Heiskanen, H. Juvonen, S. Kallinen, M. Sten, A. Tarkiainen, P. R. Ronnberg, M. Kleemola, P. H. Makela, and M. Leinonen. 1998. Etiology of childhood pneumonia: serologic results of a prospective, population-based study. Pediatr. Infect. Dis. J. 17:986-991. [DOI] [PubMed] [Google Scholar]
  • 183.Hernandez, L. A., G. E. Urquhart, and W. C. Dick. 1977. Mycoplasma pneumoniae infection and arthritis in man. Br. Med. J. 2:14-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Higuchi, M. L., N. Sambiase, S. Palomino, P. Gutierrez, L. M. Demarchi, V. D. Aiello, and J. A. Ramires. 2000. Detection of Mycoplasma pneumoniae and Chlamydia pneumoniae in ruptured atherosclerotic plaques. Braz. J. Med. Biol. Res. 33:1023-1026. [DOI] [PubMed] [Google Scholar]
  • 185.Higuchi Mde, L., M. H. Higuchi-Dos-Santos, H. Pierri, S. Palomino, N. V. Sambiase, J. A. Ramires, and M. Wajngarten. 2002. Mycoplasma pneumoniae and Chlamydia pneumoniae in calcified nodules of aortic stenotic valves. Rev. Inst. Med. Trop. Sao Paulo 44:209-212. [DOI] [PubMed] [Google Scholar]
  • 186.Hill, A. C. 1991. Mycoplasma spermatophilum, a new species isolated from human spermatozoa and cervix. Int. J. Syst. Bacteriol. 41:229-233. [DOI] [PubMed] [Google Scholar]
  • 187.Himmelreich, R., H. Hilbert, H. Plagens, E. Pirkl, B. C. Li, and R. Herrmann. 1996. Complete sequence analysis of the genome of the bacterium Mycoplasma pneumoniae. Nucleic Acids Res. 24:4420-4449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Hirai, Y., J. Shiode, T. Masayoshi, and Y. Kanemasa. 1991. Application of an indirect immunofluorescence test for detection of Mycoplasma pneumoniae in respiratory exudates. J. Clin. Microbiol. 29:2007-2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Hirschberg, L., and T. Holme. 1991. ELISA for detection of Mycoplasma pneumoniae antigens using monoclonal antibodies. APMIS 99:475-481. [PubMed] [Google Scholar]
  • 190.Hirschberg, L., T. Holme, and A. Krook. 1991. Human antibody response to the major adhesin of Mycoplasma pneumoniae: increase in titers against synthetic peptides in patients with pneumonia. APMIS 99:515-520. [DOI] [PubMed] [Google Scholar]
  • 191.Hirschberg, L., A. Krook, C. A. Pettersson, and T. Vikerfors. 1988. Enzyme-linked immunosorbent assay for detection of Mycoplasma pneumoniae specific immunoglobulin M. Eur. J. Clin. Microbiol. Infect. Dis. 7:420-423. [DOI] [PubMed] [Google Scholar]
  • 192.Hirshberg, S. J., R. S. Charles, and J. B. Ettinger. 1996. Pediatric priapism associated with Mycoplasma pneumoniae. Urology 47:745-746. [DOI] [PubMed] [Google Scholar]
  • 193.Hoek, K. L., G. H. Cassell, L. B. Duffy, and T. P. Atkinson. 2002. Mycoplasma pneumoniae-induced activation and cytokine production in rodent mast cells. J. Allergy Clin. Immunol. 109:470-476. [DOI] [PubMed] [Google Scholar]
  • 194.Honda, J., T. Yano, M. Kusaba, J. Yonemitsu, H. Kitajima, M. Masuoka, K. Hamada, and K. Oizumi. 2000. Clinical use of capillary PCR to diagnose Mycoplasma pneumonia. J. Clin. Microbiol. 38:1382-1384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Hsieh, C. C., R. B. Tang, C. H. Tsai, and W. Chen. 2001. Serum interleukin-6 and tumor necrosis factor-alpha concentrations in children with mycoplasma pneumonia. J. Microbiol. Immunol. Infect. 34:109-112. [PubMed] [Google Scholar]
  • 196.Hu, P. C., A. M. Collier, and J. B. Baseman. 1977. Surface parasitism by Mycoplasma pneumoniae of respiratory epithelium. J. Exp. Med. 145:1328-1343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Hyde, T. B., M. Gilbert, S. B. Schwartz, E. R. Zell, J. P. Watt, W. L. Thacker, D. F. Talkington, and R. E. Besser. 2001. Azithromycin prophylaxis during a hospital outbreak of Mycoplasma pneumoniae pneumonia. J. Infect. Dis. 183:907-912. [DOI] [PubMed] [Google Scholar]
  • 198.Ieven, M., H. Demey, D. Ursi, G. Van Goethem, P. Cras, and H. Goossens. 1998. Fatal encephalitis caused by Mycoplasma pneumoniae diagnosed by the polymerase chain reaction. Clin. Infect. Dis. 27:1552-1553. [DOI] [PubMed] [Google Scholar]
  • 199.Ieven, M., D. Ursi, H. Van Bever, W. Quint, H. G. Niesters, and H. Goossens. 1996. Detection of Mycoplasma pneumoniae by two polymerase chain reactions and role of M. pneumoniae in acute respiratory tract infections in pediatric patients. J. Infect. Dis. 173:1445-1452. [DOI] [PubMed] [Google Scholar]
  • 200.Inamine, J. M., K. C. Ho, S. Loechel, and P. C. Hu. 1990. Evidence that UGA is read as a tryptophan codon rather than as a stop codon by Mycoplasma pneumoniae. Mycoplasma genitalium, and Mycoplasma gallisepticum. J. Bacteriol. 172:504-506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Ito, S., Y. Abe, K. Kinomoto, T. Saitoh, T. Kato, Y. Kohli, M. Kuriyama, T. Sakai, and T. Ishizaki. 1995. Fulminant Mycoplasma pneumoniae pneumonia with marked elevation of serum soluble interleukin-2 receptor. Intern. Med. 34:430-435. [DOI] [PubMed] [Google Scholar]
  • 202.Jacobs, E. 2002. Mycoplasma pneumoniae disease manifestations and epidemiology, p. 519-530. In S. Razin and R. Herrman (ed.), Molecular biology and pathogenicity of mycoplasmas. Kluwer Academic/Plenum Publishers, New York, N.Y.
  • 203.Jacobs, E. 1993. Serological diagnosis of Mycoplasma pneumoniae infections: a critical review of current procedures. Clin. Infect. Dis. 17(Suppl. 1):S79-S82. [DOI] [PubMed] [Google Scholar]
  • 204.Jacobs, E., M. Drews, A. Stuhlert, C. Buttner, P. J. Klein, M. Kist, and W. Bredt. 1988. Immunological reaction of guinea-pigs following intranasal Mycoplasma pneumoniae infection and immunization with the 168 kDa adherence protein. J. Gen. Microbiol. 134:473-479. [DOI] [PubMed] [Google Scholar]
  • 205.Jacobs, E., A. Pilatschek, B. Gerstenecker, K. Oberle, and W. Bredt. 1990. Immunodominant epitopes of the adhesin of Mycoplasma pneumoniae. J. Clin. Microbiol. 28:1194-1197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Jacobs, E., A. Stuhlert, M. Drews, K. Pumpe, H. E. Schaefer, M. Kist, and W. Bredt. 1988. Host reactions to Mycoplasma pneumoniae infections in guinea-pigs preimmunized systemically with the adhesin of this pathogen. Microb. Pathog. 5:259-265. [DOI] [PubMed] [Google Scholar]
  • 207.Jacobs, E., M. Vonski, K. Oberle, O. Opitz, and K. Pietsch. 1996. Are outbreaks and sporadic respiratory infections by Mycoplasma pneumoniae due to two distinct subtypes? Eur. J. Clin. Microbiol. Infect. Dis. 15:38-44. [DOI] [PubMed] [Google Scholar]
  • 208.Jafri, H. S., and G. H. McCracken, Jr. 1999. Fluoroquinolones in paediatrics. Drugs 58(Suppl. 2):43-48. [DOI] [PubMed] [Google Scholar]
  • 209.Jansson, E., A. Backman, K. Hakkarainen, A. Miettinen, and B. Seniusova. 1983. Mycoplasmas and arthritis. Z. Rheumatol. 42:315-319. [PubMed] [Google Scholar]
  • 210.Jensen, J. S., C. Heilmann, and N. H. Valerius. 1994. Mycoplasma pneumoniae infection in a child with AIDS. Clin. Infect. Dis. 19:207. [DOI] [PubMed] [Google Scholar]
  • 211.Jensen, J. S., J. Sondergard-Andersen, S. A. Uldum, and K. Lind. 1989. Detection of Mycoplasma pneumoniae in simulated clinical samples by polymerase chain reaction. APMIS 97:1046-1048. [DOI] [PubMed] [Google Scholar]
  • 212.Jensen, K. J., L. B. Senterfit, W. E. Scully, T. J. Conway, R. F. West, and W. W. Drummy. 1967. Mycoplasma pneumoniae infections in children. An epidemiologic appraisal in families treated with oxytetracycline. Am. J. Epidemiol. 86:419-432. [DOI] [PubMed] [Google Scholar]
  • 213.Johansson, K.-E., and B. Pettersson. 2002. Taxonomy of mollicutes, p. 1-29. In S. Razin and R. Herrman (ed.), Molecular biology and pathogenicity of mycoplasmas. Kluwer Academic/Plenum Publishers, New York, N.Y.
  • 214.Joosting, A. C., R. M. Harwin, A. Coppin, P. Battaglia, and P. van der Hoef. 1976. A serological investigation of Mycoplasma pneumoniae infection on the Witwatersrand. S. Afr. Med. J. 50:2134-2135. [PubMed] [Google Scholar]
  • 215.Kai, M., S. Kamiya, H. Yabe, I. Takakura, K. Shiozawa, and A. Ozawa. 1993. Rapid detection of Mycoplasma pneumoniae in clinical samples by the polymerase chain reaction. J. Med. Microbiol. 38:166-170. [DOI] [PubMed] [Google Scholar]
  • 216.Kaku, M., S. Kohno, H. Koga, K. Ishida, and K. Hara. 1995. Efficacy of roxithromycin in the treatment of mycoplasma pneumonia. Chemotherapy 41:149-152. [DOI] [PubMed] [Google Scholar]
  • 217.Kanayama, Y., K. Shiota, K. Kotumi, Y. Ikuno, R. Yasumoto, M. Ishii, and T. Inoue. 1982. Mycoplasma pneumoniae pneumonia associated with IgA nephropathy. Scand. J. Infect. Dis. 14:231-233. [DOI] [PubMed] [Google Scholar]
  • 218.Karjalainen, J. 1990. A loud third heart sound and asymptomatic myocarditis during Mycoplasma pneumoniae infection. Eur. Heart J. 11:960-963. [DOI] [PubMed] [Google Scholar]
  • 219.Karppelin, M., K. Hakkarainen, M. Kleemola, and A. Miettinen. 1993. Comparison of three serological methods for diagnosing Mycoplasma pneumoniae infection. J. Clin. Pathol. 46:1120-1123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Kasahara, I., Y. Otsubo, T. Yanase, H. Oshima, H. Ichimaru, and M. Nakamura. 1985. Isolation and characterization of Mycoplasma pneumoniae from cerebrospinal fluid of a patient with pneumonia and meningoencephalitis. J. Infect. Dis. 152:823-825. [DOI] [PubMed] [Google Scholar]
  • 221.Kashiwagi, S., J. Hayashi, H. Nomura, W. Kajiyama, H. Ikematsu, T. Shingu, K. Hayashida, and M. Kaji. 1985. An outbreak of Mycoplasma pneumoniae infections in a hospital in Japan. Kurume Med. J. 32:293-296. [DOI] [PubMed] [Google Scholar]
  • 222.Kaufman, J. M., C. A. Cuvelier, and M. Van der Straeten. 1980. Mycoplasma pneumonia with fulminant evolution into diffuse interstitial fibrosis. Thorax 35:140-144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Keegan, B. M., N. J. Lowry, and J. Y. Yager. 1999. Mycoplasma pneumoniae: a cause of coma in the absence of meningoencephalitis. Pediatr. Neurol. 21:822-825. [DOI] [PubMed] [Google Scholar]
  • 224.Kenny, G. E., and F. D. Cartwright. 2001. Susceptibilities of Mycoplasma hominis. M. pneumoniae, and Ureaplasma urealyticum to GAR-936, dalfopristin, dirithromycin, evernimicin, gatifloxacin, linezolid, moxifloxacin, quinupristin-dalfopristin, and telithromycin compared to their susceptibilities to reference macrolides, tetracyclines, and quinolones. Antimicrob. Agents Chemother. 45:2604-2608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Kenny, G. E., G. G. Kaiser, M. K. Cooney, and H. M. Foy. 1990. Diagnosis of Mycoplasma pneumoniae pneumonia: sensitivities and specificities of serology with lipid antigen and isolation of the organism on soy peptone medium for identification of infections. J. Clin. Microbiol. 28:2087-2093. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Kikuchi, M., Y. Tagawa, H. Iwamoto, H. Hoshino, and N. Yuki. 1997. Bickerstaff's brainstem encephalitis associated with IgG anti-GQ1b antibody subsequent to Mycoplasma pneumoniae infection: favorable response to immunoadsorption therapy. J. Child Neurol. 12:403-405. [DOI] [PubMed] [Google Scholar]
  • 227.Kim, C. K., C. Y. Chung, J. S. Kim, W. S. Kim, Y. Park, and Y. Y. Koh. 2000. Late abnormal findings on high-resolution computed tomography after Mycoplasma pneumonia. Pediatrics 105:372-378. [DOI] [PubMed] [Google Scholar]
  • 228.Klausner, J. D., D. Passaro, J. Rosenberg, W. L. Thacker, D. F. Talkington, S. B. Werner, and D. J. Vugia. 1998. Enhanced control of an outbreak of Mycoplasma pneumoniae pneumonia with azithromycin prophylaxis. J. Infect. Dis. 177:161-166. [DOI] [PubMed] [Google Scholar]
  • 229.Kleemola, M., and C. Jokinen. 1992. Outbreak of Mycoplasma pneumoniae infection among hospital personnel studied by a nucleic acid hybridization test. J. Hosp. Infect. 21:213-221. [DOI] [PubMed] [Google Scholar]
  • 230.Kleemola, M., and H. Kayhty. 1982. Increase in titers of antibodies to Mycoplasma pneumoniae in patients with purulent meningitis. J. Infect. Dis. 146:284-288. [DOI] [PubMed] [Google Scholar]
  • 231.Kleemola, M., R. Raty, J. Karjalainen, W. Schuy, B. Gerstenecker, and E. Jacobs. 1993. Evaluation of an antigen-capture enzyme immunoassay for rapid diagnosis of Mycoplasma pneumoniae infection. Eur. J. Clin. Microbiol. Infect. Dis. 12:872-875. [DOI] [PubMed] [Google Scholar]
  • 232.Klieneberger, E. 1935. The natural occurrence of pleuropneumonia-like organisms in apparent symbiosis with Streptobacillus moniliformis and other bacteria. J. Pathol. Bacteriol. 40:93-105. [Google Scholar]
  • 233.Koh, Y. Y., Y. Park, H. J. Lee, and C. K. Kim. 2001. Levels of interleukin-2, interferon-gamma, and interleukin-4 in bronchoalveolar lavage fluid from patients with Mycoplasma pneumonia: implication of tendency toward increased immunoglobulin E production. Pediatrics 107:E39. [DOI] [PubMed] [Google Scholar]
  • 234.Kok, T. W., G. Varkanis, B. P. Marmion, J. Martin, and A. Esterman. 1988. Laboratory diagnosis of Mycoplasma pneumoniae infection. 1. Direct detection of antigen in respiratory exudates by enzyme immunoassay. Epidemiol. Infect. 101:669-684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Koletsky, R. J., and A. J. Weinstein. 1980. Fulminant Mycoplasma pneumoniae infection. Report of a fatal case, and a review of the literature. Am. Rev. Respir. Dis. 122:491-496. [DOI] [PubMed] [Google Scholar]
  • 236.Kong, F., S. Gordon, and G. L. Gilbert. 2000. Rapid-cycle PCR for detection and typing of Mycoplasma pneumoniae in clinical specimens. J. Clin. Microbiol. 38:4256-4259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Koskiniemi, M. 1993. CNS manifestations associated with Mycoplasma pneumoniae infections: summary of cases at the University of Helsinki and review. Clin. Infect. Dis. 17(Suppl. 1):S52-S57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Kraft, M., G. H. Cassell, J. E. Henson, H. Watson, J. Williamson, B. P. Marmion, C. A. Gaydos, and R. J. Martin. 1998. Detection of Mycoplasma pneumoniae in the airways of adults with chronic asthma. Am. J. Respir. Crit. Care Med. 158:998-1001. [DOI] [PubMed] [Google Scholar]
  • 239.Kraft, M., G. H. Cassell, J. Pak, and R. J. Martin. 2002. Mycoplasma pneumoniae and Chlamydia pneumoniae in asthma: effect of clarithromycin. Chest 121:1782-1788. [DOI] [PubMed] [Google Scholar]
  • 240.Krause, D. C. 1998. Mycoplasma pneumoniae cytadherence: organization and assembly of the attachment organelle. Trends Microbiol. 6:15-18. [DOI] [PubMed] [Google Scholar]
  • 241.Krause, D. C. 1996. Mycoplasma pneumoniae cytadherence: unravelling the tie that binds. Mol. Microbiol. 20:247-253. [DOI] [PubMed] [Google Scholar]
  • 242.Krause, D. C., and J. B. Baseman. 1983. Inhibition of Mycoplasma pneumoniae hemadsorption and adherence to respiratory epithelium by antibodies to a membrane protein. Infect. Immun. 39:1180-1186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Krause, D. C., D. K. Leith, and J. B. Baseman. 1983. Reacquisition of specific proteins confers virulence in Mycoplasma pneumoniae. Infect. Immun. 39:830-836. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244.Krause, D. C., D. K. Leith, R. M. Wilson, and J. B. Baseman. 1982. Identification of Mycoplasma pneumoniae proteins associated with hemadsorption and virulence. Infect. Immun. 35:809-817. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245.Krause, D. C., and D. Taylor-Robinson. 1992. Mycoplasmas which infect humans, p. 417-444. In J. Maniloff, R. N. McElhaney, L. R. Finch, and J. B. Baseman (ed.), Mycoplasmas: molecular biology and pathogenesis. American Society for Microbiology, Washington, D.C.
  • 246.Krause, I., R. Wu, Y. Sherer, M. Patanik, J. M. Peter, and Y. Schoenfeld. 2002. In vitro antiviral and antibacterial activity of commercial intravenous immunoglobulin preparations—a potential role for adjuvant intravenous immunoglobulin therapy in infectious diseases. Transfus. Med. 12:133-139. [DOI] [PubMed] [Google Scholar]
  • 247.Krivan, H. C., L. D. Olson, M. F. Barile, V. Ginsburg, and D. D. Roberts. 1989. Adhesion of Mycoplasma pneumoniae to sulfated glycolipids and inhibition by dextran sulfate. J. Biol. Chem. 264:9283-9288. [PubMed] [Google Scholar]
  • 248.Lambert, H. P. 1969. Infections caused by Mycoplasma pneumoniae. Br. J. Dis. Chest 63:71-82. [DOI] [PubMed] [Google Scholar]
  • 249.Lambert, H. P. 1968. Mycoplasma pneumoniae infections. J. Clin. Pathol. Suppl. 2:52-60. [PubMed] [Google Scholar]
  • 250.La Scola, B., G. Michel, and D. Raoult. 1997. Use of amplification and sequencing of 16S rRNA gene to diagnose Mycoplasma pneumoniae osteomyelitis in a patient with hypogammaglobuliemia. Clin. Infect. Dis. 24:1161-1163. [DOI] [PubMed] [Google Scholar]
  • 251.Launes, J., A. Paetau, K. Linnavuori, and M. Iivanaineu. 1997. Direct invasion of the brain parenchyma by Mycoplasma pneumoniae. Acta Neurol. Scand. 95:374. [DOI] [PubMed] [Google Scholar]
  • 252.Layani-Milon, M. P., I. Gras, M. Valette, J. Luciani, J. Stagnara, M. Aymard, and B. Lina. 1999. Incidence of upper respiratory tract Mycoplasma pneumoniae infections among outpatients in Rhone-Alpes, France, during five successive winter periods. J. Clin. Microbiol. 37:1721-1726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253.Leibowitz, Z., P. Schvartzman, L. Epstein, I. Lis, and Y. Naot. 1988. An outbreak of Mycoplasma pneumoniae pneumonia in two kibbutzim: a clinical and epidemiologic study. Isr. J. Med. Sci. 24:88-92. [PubMed] [Google Scholar]
  • 254.Leng, Z., G. E. Kenny, and M. C. Roberts. 1994. Evaluation of the detection limits of PCR for identification of Mycoplasma pneumoniae in clinical samples. Mol. Cell. Probes. 8:125-130. [DOI] [PubMed] [Google Scholar]
  • 255.Lepow, M. L., N. Balassanian, J. Emmerich, R. B. Roberts, M. S. Rosenthal, and E. Wolinsky. 1968. Interrelationships of viral, mycoplasmal, and bacterial agents in uncomplicated pneumonia. Am. Rev. Respir. Dis. 97:533-545. [DOI] [PubMed] [Google Scholar]
  • 256.Levine, D. P., and A. M. Lerner. 1978. The clinical spectrum of Mycoplasma pneumoniae infections. Med. Clin. N. Am. 62:961-978. [DOI] [PubMed] [Google Scholar]
  • 257.Levy, M., and N. H. Shear. 1991. Mycoplasma pneumoniae infections and Stevens-Johnson syndrome. Report of eight cases and review of the literature. Clin. Pediatr. 30:42-49. [DOI] [PubMed] [Google Scholar]
  • 258.Lewis, J. E., and C. Sheptin. 1972. Mycoplasmal pneumonia associated with abscess of the lung. Calif. Med. 117:69-72. [PMC free article] [PubMed] [Google Scholar]
  • 259.Libre, J. M., A. Urban, M. A. Garcia, M. A. Carrasco, and C. Murcia. 1997. Bronchiolitis obliterans organizing pneumonia associated with acute Mycoplasma pneumoniae infection. Clin. Infect. Dis. 25:1340-1342. [DOI] [PubMed] [Google Scholar]
  • 260.Lieberman, D., M. Ben-Yaakov, Z. Lazarovich, S. Hoffman, B. Ohana, M. G. Friedman, B. Dvoskin, M. Leinonen, and I. Boldur. 2001. Infectious etiologies in acute exacerbation of COPD. Diagn. Microbiol. Infect. Dis. 40:95-102. [DOI] [PubMed] [Google Scholar]
  • 261.Lieberman, D., M. Ben-Yaakov, O. Shmarkov, Y. Gelfer, R. Varshavsky, B. Ohana, Z. Lazarovich, and I. Boldur. 2002. Serological evidence of Mycoplasma pneumoniae infection in acute exacerbation of COPD. Diagn. Microbiol. Infect. Dis. 44:1-6. [DOI] [PubMed] [Google Scholar]
  • 262.Lieberman, D., S. Horowitz, O. Horovitz, F. Schlaeffer, and A. Porath. 1995. Microparticle agglutination versus antibody-capture enzyme immunoassay for diagnosis of community-acquired Mycoplasma pneumoniae pneumonia. Eur. J. Clin. Microbiol. Infect. Dis. 14:577-584. [DOI] [PubMed] [Google Scholar]
  • 263.Lieberman, D., S. Livnat, F. Schlaeffer, A. Porath, S. Horowitz, and R. Levy. 1997. IL-1beta and IL-6 in community-acquired pneumonia: bacteremic pneumococcal pneumonia versus Mycoplasma pneumoniae pneumonia. Infection 25:90-94. [DOI] [PubMed] [Google Scholar]
  • 264.Lin, W. C., P. I. Lee, C. Y. Lu, Y. C. Hsieh, H. P. Lai, C. Y. Lee, and L. M. Huang. 2002. Mycoplasma pneumoniae encephalitis in childhood. J. Microbiol. Immunol. Infect. 35:173-178. [PubMed] [Google Scholar]
  • 265.Lind, K. 1982. Serological cross-reactions between “Mycoplasma genitalium” and M. pneumoniae. Lancet ii:1158-1159. [DOI] [PubMed] [Google Scholar]
  • 266.Lind, K., and M. W. Bentzon. 1976. Epidemics of Mycoplasma pneumoniae infection in Denmark from 1958 to 1974. Int. J. Epidemiol. 5:267-277. [DOI] [PubMed] [Google Scholar]
  • 267.Lind, K., M. W. Benzon, J. S. Jensen, and W. A. Clyde, Jr. 1997. A seroepidemiological study of Mycoplasma pneumoniae infections in Denmark over the 50-year period 1946-1995. Eur. J. Epidemiol. 13:581-586. [DOI] [PubMed] [Google Scholar]
  • 268.Liu, C., M. D. Eaton, and J. T. Heyl. 1957. Studies on atypical pneumonia. II. Observations concerning the development and immunological characteristics of antibody in patients. J. Exp. Med. 109:545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 269.Liu, C., P. Jayanetra, and D. W. Voth. 1970. Effect of combined Mycoplasma pneumoniae and pneumococcal infections in hamsters. Ann. N. Y. Acad. Sci. 174:828-834. [DOI] [PubMed] [Google Scholar]
  • 270.Lo, S. C., M. M. Hayes, J. G. Tully, R. Y. Wang, H. Kotani, P. F. Pierce, D. L. Rose, and J. W. Shih. 1992. Mycoplasma penetrans sp. nov., from the urogenital tract of patients with AIDS. Int. J. Syst. Bacteriol. 42:357-364. [DOI] [PubMed] [Google Scholar]
  • 271.Loda, F. A., W. A. Clyde, Jr., W. P. Glezen, R. J. Senior, C. I. Sheaffer, and F. W. Denny, Jr. 1968. Studies on the role of viruses, bacteria, and M. pneumoniae as causes of lower respiratory tract infections in children. J. Pediatr. 72:161-176. [DOI] [PubMed] [Google Scholar]
  • 272.Loens, K., D. Ursi, H. Goossens, and M. Ieven. 2003. Molecular diagnosis of Mycoplasma pneumoniae respiratory tract infections. J. Clin. Microbiol. 41:4915-4923. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273.Luby, J. P. 1991. Pneumonia caused by Mycoplasma pneumoniae infection. Clin. Chest Med. 12:237-244. [PubMed] [Google Scholar]
  • 274.Lucier, T. S., K. Heitzman, S. K. Liu, and P. C. Hu. 1995. Transition mutations in the 23S rRNA of erythromycin-resistant isolates of Mycoplasma pneumoniae. Antimicrob. Agents Chemother. 39:2770-2773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 275.Luneberg, E., J. S. Jensen, and M. Frosch. 1993. Detection of Mycoplasma pneumoniae by polymerase chain reaction and nonradioactive hybridization in microtiter plates. J. Clin. Microbiol. 31:1088-1094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 276.Macfarlane, J. 1980. Mycoplasma pneumoniae infection, antibiotics and exanthema. Infection 8:119-120. [DOI] [PubMed] [Google Scholar]
  • 277.Maisel, J. C., L. H. Babbitt, and T. J. John. 1967. Fatal Mycoplasma pneumoniae infection with isolation of organisms from lung. JAMA 202:287-290. [PubMed] [Google Scholar]
  • 278.Maniloff, J. 1992. Phylogeny of mycoplasmas, p. 549-559. In J. Maniloff, R. N. McElhaney, L. R. Finch, and J. B. Baseman (ed.), Mycoplasmas: molecular biology and pathogenesis. American Society for Microbiology, Washington, D.C.
  • 279.Marc, E., M. Chaussain, F. Moulin, J. L. Iniguez, G. Kalifa, J. Raymond, and D. Gendrel. 2000. Reduced lung diffusion capacity after Mycoplasma pneumoniae pneumonia. Pediatr. Infect. Dis. J. 19:706-710. [DOI] [PubMed] [Google Scholar]
  • 280.Markham, J. G. 1979. Mycoplasma pneumoniae infection in 1978. N. Z. Med. J. 90:473-474. [PubMed] [Google Scholar]
  • 281.Marmion, B. P., and G. M. Goodburn. 1961. Effect of an inorganic gold salt on Eaton's primary atypical pneumonia agent and other observations. Nature 189:247-248. [DOI] [PubMed] [Google Scholar]
  • 282.Marrie, T. J. 1993. Mycoplasma pneumoniae pneumonia requiring hospitalization, with emphasis on infection in the elderly. Arch. Intern. Med. 153:488-494. [PubMed] [Google Scholar]
  • 283.Marston, B. J., J. F. Plouffe, T. M. File, Jr., B. A. Hackman, S. J. Salstrom, H. B. Lipman, M. S. Kolczak, R. F. Breiman, et al. 1997. Incidence of community-acquired pneumonia requiring hospitalization. Results of a population-based active surveillance study in Ohio. Arch. Intern. Med. 157:1709-1718. [PubMed] [Google Scholar]
  • 284.Martin, R. J., H. W. Chu, J. M. Honour, and R. J. Harbeck. 2001. Airway inflammation and bronchial hyperresponsiveness after Mycoplasma pneumoniae infection in a murine model. Am. J. Respir. Cell Mol. Biol. 24:577-582. [DOI] [PubMed] [Google Scholar]
  • 285.Martin, R. R., G. Warr, R. Couch, and V. Knight. 1973. Chemotaxis of human leukocytes: responsiveness to Mycoplasma pneumoniae. J. Lab. Clin. Med. 81:520-529. [PubMed] [Google Scholar]
  • 286.Matas, L., J. Dominguez, F. De Ory, N. Garcia, N. Gali, P. J. Cardona, A. Hernandez, C. Rodrigo, and V. Ausina. 1998. Evaluation of Meridian ImmunoCard Mycoplasma test for the detection of Mycoplasma pneumoniae-specific IgM in paediatric patients. Scand. J. Infect. Dis. 30:289-293. [DOI] [PubMed] [Google Scholar]
  • 287.Meseguer, M. A., J. A. Perez-Molina, J. Fernandez-Bustamante, R. Gomez, I. Martos, and M. C. Quero. 1996. Mycoplasma pneumoniae pericarditis and cardiac tamponade in a ten-year-old girl. Pediatr. Infect. Dis. J. 15:829-831. [DOI] [PubMed] [Google Scholar]
  • 288.Meyers, L. A., M. E. J. Newman, M. Martin, and S. Schrag. 2003. Applying network theory to epidemics: control measures for Mycoplasma pneumoniae outbreaks. Emerg. Infect. Dis. 9:204-210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 289.Milla, E., L. Zografos, and B. Piguet. 1998. Bilateral optic papillitis following Mycoplasma pneumoniae pneumonia. Ophthalmologica 212:344-346. [DOI] [PubMed] [Google Scholar]
  • 290.Miyata, M. 2002. Gliding motility of mycoplasma—a mechanism cannot be explained by today's biology. Nippon Saikingaku Zasshi 57:581-595. [PubMed] [Google Scholar]
  • 291.Mizutani, H. 1983. Immunologic responses in patients with Mycoplasma pneumoniae infections. Am. Rev. Respir. Dis. 127:175-179. [DOI] [PubMed] [Google Scholar]
  • 292.Mogabgab, W. J. 1968. Mycoplasma pneumoniae and adenovirus respiratory illnesses in military and university personnel, 1959-1966. Am. Rev. Respir. Dis. 97:345-358. [DOI] [PubMed] [Google Scholar]
  • 293.Mogulkoc, N., S. Karakurt, B. Isalska, U. Bayindir, T. Celikel, V. Korten, and N. Colpan. 1999. Acute purulent exacerbation of chronic obstructive pulmonary disease and Chlamydia pneumoniae infection. Am. J. Respir. Crit. Care Med. 160:349-353. [DOI] [PubMed] [Google Scholar]
  • 294.Mok, J. Y., J. M. Inglis, and H. Simpson. 1979. Mycoplasma pneumoniae infection. A retrospective review of 103 hospitalised children. Acta Paediatr. Scand. 68:833-839. [DOI] [PubMed] [Google Scholar]
  • 295.Mok, J. Y., P. R. Waugh, and H. Simpson. 1979. Mycoplasma pneuminia infection. A follow-up study of 50 children with respiratory illness. Arch. Dis. Child. 54:506-511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 296.Monto, A. S., E. R. Bryan, and L. M. Rhodes. 1974. The Tecumseh study of respiratory illness. VII. Further observations on the occurrence of respiratory syncytial virus and Mycoplasma pneumoniae infections. Am. J. Epidemiol. 100:458-468. [DOI] [PubMed] [Google Scholar]
  • 297.Monto, A. S., and S. K. Lim. 1971. The Tecumseh study of respiratory illness. 3. Incidence and periodicity of respiratory syncytial virus and Mycoplasma pneumoniae infections. Am. J. Epidemiol. 94:290-301. [DOI] [PubMed] [Google Scholar]
  • 298.Morrison-Plummer, J., D. K. Leith, and J. B. Baseman. 1986. Biological effects of anti-lipid and anti-protein monoclonal antibodies on Mycoplasma pneumoniae. Infect. Immun. 53:398-403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 299.Muldoon, R. L., J. Raucci, J. Kowalski, and K. Rajashekaraiah. 1982. An outbreak of Mycoplasma pneumoniae respiratory illness in a semi-closed religious commune. Ann. Emerg. Med. 11:613-615. [DOI] [PubMed] [Google Scholar]
  • 300.Murdoch, D. R. 2003. Nucleic acid amplification tests for the diagnosis of pneumonia. Clin. Infect. Dis. 36:1162-1170. [DOI] [PubMed] [Google Scholar]
  • 301.Murray, H. W., H. Masur, L. B. Senterfit, and R. B. Roberts. 1975. The protean manifestations of Mycoplasma pneumoniae infection in adults. Am. J. Med. 58:229-242. [DOI] [PubMed] [Google Scholar]
  • 302.Nakayama, T., T. Urano, M. Osano, N. Maehara, and S. Makino. 1986. Alpha interferon in the sera of patients infected with Mycoplasma pneumoniae. J. Infect. Dis. 154:904-906. [DOI] [PubMed] [Google Scholar]
  • 303.Naot, Y., E. Lis, R. Siman-Tov, and H. Brunner. 1986. Comparison of enzyme-linked immunosorbent assay and radioimmunoprecipitation test for detection of immunoglobulin A antibodies to Mycoplasma pneumoniae in nasal secretions. J. Clin. Microbiol. 24:892-893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 304.Naraqi, S., and S. A. Kabins. 1973. Mycoplasma pneumoniae monoarticular arthritis. J. Pediatr. 83:621-623. [DOI] [PubMed] [Google Scholar]
  • 305.Narita, M. 2000. Detection of Mycoplasma pneumoniae DNA in cerebrospinal fluid and local immune response. Clin. Infect. Dis. 30:405-406. [DOI] [PubMed] [Google Scholar]
  • 306.Narita, M., Y. Matsuzono, O. Itakura, T. Togashi, and H. Kikuta. 1996. Survey of mycoplasmal bacteremia detected in children by polymerase chain reaction. Clin. Infect. Dis. 23:522-525. [DOI] [PubMed] [Google Scholar]
  • 307.Narita, M., Y. Matsuzono, T. Togashi, and N. Kajii. 1992. DNA diagnosis of central nervous system infection by Mycoplasma pneumoniae. Pediatrics 90:250-253. [PubMed] [Google Scholar]
  • 308.Narita, M., H. Tanaka, S. Abe, S. Yamada, M. Kubota, and T. Togashi. 2000. Close association between pulmonary disease manifestation in Mycoplasma pneumoniae infection and enhanced local production of interleukin-18 in the lung, independent of gamma interferon. Clin. Diagn. Lab. Immunol. 7:909-914. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 309.Narita, M., H. Tanaka, S. Yamada, S. Abe, T. Ariga, and Y. Sakiyama. 2001. Significant role of interleukin-8 in pathogenesis of pulmonary disease due to Mycoplasma pneumoniae infection. Clin. Diagn. Lab. Immunol. 8:1028-1030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 310.Niitu, Y., S. Hasegawa, T. Suetake, H. Kubota, S. Komatsu, and M. Horikawa. 1970. Resistance of Mycoplasma pneumoniae to erythromycin and other antibiotics. J. Pediatr. 76:438-443. [DOI] [PubMed] [Google Scholar]
  • 311.Niitu, Y., H. Kubota, S. Hasegawa, S. Komatsu, and M. Horikawa. 1974. Susceptibility of Mycoplasma pneumoniae to antibiotics in vitro. Jpn. J. Microbiol. 18:149-155. [DOI] [PubMed] [Google Scholar]
  • 312.Nishimura, M., T. Saida, S. Kuroki, T. Kawabata, H. Obayashi, K. Saida, and T. Uchiyama. 1996. Post-infectious encephalitis with anti-galactocerebroside antibody subsequent to Mycoplasma pneumoniae infection. J. Neurol. Sci. 140:91-95. [DOI] [PubMed] [Google Scholar]
  • 313.Noah, N. D. 1976. Epidemiology of Mycoplasma pneumoniae infection in the United Kingdom: an analysis of reports to the Public Health Laboratory Service of England and Wales. Infection 4:25-28. [DOI] [PubMed] [Google Scholar]
  • 314.Noah, N. D. 1974. Mycoplasma pneumoniae infection in the United Kingdom—1967-73. Br. Med. J. 2:544-546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 315.Nocard, E., and E. R. Roux. 1898. Le microbe de la peripneumonie. Ann. Inst. Pasteur (Paris) 12:240-262. [Google Scholar]
  • 316.Okazaki, N., M. Narita, S. Yamada, K. Izumikawa, M. Umetsu, T. Kenri, Y. Sasaki, Y. Arakawa, and T. Sasaki. 2001. Characteristics of macrolide-resistant Mycoplasma pneumoniae strains isolated from patients and induced with erythromycin in vitro. Microbiol. Immunol. 45:617-620. [DOI] [PubMed] [Google Scholar]
  • 317.Ong, E. L., M. E. Ellis, A. K. Webb, K. R. Neal, M. Dodd, E. O. Caul, and S. Burgess. 1989. Infective respiratory exacerbations in young adults with cystic fibrosis: role of viruses and atypical microorganisms. Thorax 44:739-742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 318.Opitz, O., K. Pietsch, S. Ehlers, and E. Jacobs. 1996. Cytokine gene expression in immune mice reinfected with Mycoplasma pneumoniae: the role of T cell subsets in aggravating the inflammatory response. Immunobiology 196:575-587. [DOI] [PubMed] [Google Scholar]
  • 319.O'Riordan, J. I., B. Gomez-Anson, I. F. Moseley, and D. H. Miller. 1999. Long term MRI follow-up of patients with post infectious encephalomyelitis: evidence for a monophasic disease. J. Neurol. Sci. 167:132-136. [DOI] [PubMed] [Google Scholar]
  • 320.Ovyn, C., D. van Strijp, M. Ieven, D. Ursi, B. van Gemen, and H. Goossens. 1996. Typing of Mycoplasma pneumoniae by nucleic acid sequence-based amplification, NASBA. Mol. Cell. Probes 10:319-324. [DOI] [PubMed] [Google Scholar]
  • 321.Pasternack, A., H. Helin, T. Vanttinen, G. Jarventie, and T. Vesikari. 1979. Acute tubulointerstitial nephritis in a patient with Mycoplasma pneumoniae infection. Scand. J. Infect. Dis. 11:85-87. [DOI] [PubMed] [Google Scholar]
  • 322.Patel, T., J. Pearl, J. Williams, D. Haverstock, D. Church, et al. 2000. Efficacy and safety of ten day moxifloxacin 400 mg once daily in the treatment of patients with community-acquired pneumonia. Respir. Med. 94:97-105. [DOI] [PubMed] [Google Scholar]
  • 323.Pellegrini, M., T. J. O'Brien, J. Hoy, and L. Sedal. 1996. Mycoplasma pneumoniae infection associated with an acute brainstem syndrome. Acta Neurol. Scand. 93:203-206. [DOI] [PubMed] [Google Scholar]
  • 324.Petersen, N. T., N. Hoiby, C. H. Mordhorst, K. Lind, E. W. Flensborg, and B. Bruun. 1981. Respiratory infections in cystic fibrosis patients caused by virus, chlamydia and mycoplasma—possible synergism with Pseudomonas aeruginosa. Acta Paediatr. Scand. 70:623-628. [DOI] [PubMed] [Google Scholar]
  • 325.Pfausler, B., K. Engelhardt, A. Kampfl, H. Spiss, E. Taferner, and E. Schmutzhard. 2002. Post-infectious central and peripheral nervous system diseases complicating Mycoplasma pneumoniae infection. Report of three cases and review of the literature. Eur. J. Neurol. 9:93-96. [DOI] [PubMed] [Google Scholar]
  • 326.Plouffe, J. F., M. T. Herbert, T. M. File, Jr., I. Baird, J. N. Parsons, J. B. Kahn, and K. T. Rielly-Gauvin, et al. 1996. Ofloxacin versus standard therapy in treatment of community-acquired pneumonia requiring hospitalization. Antimicrob. Agents Chemother. 40:1175-1179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 327.Pollack, J. D., M. A. Myers, T. Dandekar, and R. Herrmann. 2002. Suspected utility of enzymes with multiple activities in the small genome Mycoplasma species: the replacement of the missing “household” nucleoside diphosphate kinase gene and activity by glycolytic kinases. Omics 6:247-258. [DOI] [PubMed] [Google Scholar]
  • 328.Pollack, J. D., M. V. Williams, and R. N. McElhaney. 1997. The comparative metabolism of the mollicutes (Mycoplasmas): the utility for taxonomic classification and the relationship of putative gene annotation and phylogeny to enzymatic function in the smallest free-living cells. Crit. Rev. Microbiol. 23:269-354. [DOI] [PubMed] [Google Scholar]
  • 329.Ponka, A. 1979. Arthritis associated with Mycoplasma pneumoniae infection. Scand. J. Rheumatol. 8:27-32. [PubMed] [Google Scholar]
  • 330.Ponka, A. 1979. Carditis associated with Mycoplasma pneumoniae infection. Acta Med. Scand. 206:77-86. [DOI] [PubMed] [Google Scholar]
  • 331.Ponka, A. 1980. Central nervous system manifestations associated with serologically verified Mycoplasma pneumoniae infection. Scand. J. Infect. Dis. 12:175-184. [DOI] [PubMed] [Google Scholar]
  • 332.Ponka, A. 1978. Clinical and laboratory manifestations in patients with serological evidence of Mycoplasma pneumoniae infection. Scand. J. Infect. Dis. 10:271-275. [DOI] [PubMed] [Google Scholar]
  • 333.Ponka, A. 1979. The occurrence and clinical picture of serologically verified Mycoplasma pneumoniae infections with emphasis on central nervous system, cardiac and joint manifestations. Ann. Clin. Res. 11(Suppl. 24):1-60. [PubMed] [Google Scholar]
  • 334.Porath, A., F. Schlaeffer, and D. Lieberman. 1997. The epidemiology of community-acquired pneumonia among hospitalized adults. J. Infect. 34:41-48. [DOI] [PubMed] [Google Scholar]
  • 335.Pribble, C. G., P. G. Black, J. A. Bosso, and R. B. Turner. 1990. Clinical manifestations of exacerbations of cystic fibrosis associated with nonbacterial infections. J. Pediatr. 117:200-204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 336.Pullman, J., J. Champlin, and P. S. Vrooman, Jr. 2003. Efficacy and tolerability of once-daily oral therapy with telithromycin compared with trovafloxacin for the treatment of community-acquired pneumonia in adults. Int. J. Clin. Pract. 57:377-384. [PubMed] [Google Scholar]
  • 337.Qasem, J. A., Z. U. Khan, G. Shiji, and A. S. Mustafa. 2002. Polymerase chain reaction as a sensitive and rapid method for specific detection of Mycoplasma pneumoniae in clinical samples. Microbiol. Res. 157:77-82. [DOI] [PubMed] [Google Scholar]
  • 338.Radisic, M., A. Torn, P. Gutierrez, H. A. Defranchi, and P. Pardo. 2000. Severe acute lung injury caused by Mycoplasma pneumoniae: potential role for steroid pulses in treatment. Clin. Infect. Dis. 31:1507-1511. [DOI] [PubMed] [Google Scholar]
  • 339.Ramirez, J. A., S. Ahkee, A. Tolentino, R. D. Miller, and J. T. Summersgill. 1996. Diagnosis of Legionella pneumophila. Mycoplasma pneumoniae, or Chlamydia pneumoniae lower respiratory infection using the polymerase chain reaction on a single throat swab specimen. Diagn. Microbiol. Infect. Dis. 24:7-14. [DOI] [PubMed] [Google Scholar]
  • 340.Rastawicki, W., and M. Jagielski. 1998. Electrophoretic and immunologic comparative analysis of Mycoplasma pneumoniae and Mycoplasma genitalium proteins. Med. Dosw. Mikrobiol. 50:259-267. [PubMed] [Google Scholar]
  • 341.Rautonen, J., M. Koskiniemi, and A. Vaheri. 1991. Prognostic factors in childhood acute encephalitis. Pediatr. Infect. Dis. J. 10:441-446. [DOI] [PubMed] [Google Scholar]
  • 342.Rawadi, G., S. Roman-Roman, M. Castedo, V. Dutilleul, S. Susin, P. Marchetti, M. Geuskens, and G. Kroemer. 1996. Effects of Mycoplasma fermentans on the myelomonocytic lineage. Different molecular entities with cytokine-inducing and cytocidal potential. J. Immunol. 156:670-678. [PubMed] [Google Scholar]
  • 343.Razin, S. 2002. Diagnosis of mycoplasmal infections, p. 531-544. In S. Razin and R. Herrmann (ed.), Molecular biology and pathogenicity of mycoplasmas. Kluwer Academic/Plenum Publishers, New York, N.Y.
  • 344.Razin, S. 1994. DNA probes and PCR in diagnosis of mycoplasma infections. Mol. Cell. Probes 8:497-511. [DOI] [PubMed] [Google Scholar]
  • 345.Razin, S., D. Yogev, and Y. Naot. 1998. Molecular biology and pathogenicity of mycoplasmas. Microbiol. Mol. Biol. Rev. 62:1094-1156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 346.Reznikov, M., T. K. Blackmore, J. J. Finlay-Jones, and D. L. Gordon. 1995. Comparison of nasopharyngeal aspirates and throat swab specimens in a polymerase chain reaction-based test for Mycoplasma pneumoniae. Eur. J. Clin. Microbiol. Infect. Dis. 14:58-61. [DOI] [PubMed] [Google Scholar]
  • 347.Roberts, D. D., L. D. Olson, M. F. Barile, V. Ginsburg, and H. C. Krivan. 1989. Sialic acid-dependent adhesion of Mycoplasma pneumoniae to purified glycoproteins. J. Biol. Chem. 264:9289-9293. [PubMed] [Google Scholar]
  • 348.Roberts, M. C., and G. E. Kenny. 1986. Dissemination of the tetM tetracycline resistance determinant to Ureaplasma urealyticum. Antimicrob. Agents Chemother. 29:350-352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 349.Roberts, M. C., L. A. Koutsky, D. LeBlanc, K. K. Holmes, and G. E. Kenny. 1985. Tetracycline-resistant Mycoplasma hominis strains containing streptococcal tetM sequences. Antimicrob. Agents Chemother. 28:141-143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 350.Robertson, J. A., G. W. Stemke, J. W. Davis, Jr., R. Harasawa, D. Thirkell, F. Kong, M. C. Shepard, and D. K. Ford. 2002. Proposal of Ureaplasma parvum sp. nov. and emended description of Ureaplasma urealyticum (Shepard et al. 1974). Int. J. Syst. Evol. Microbiol. 52:587-597. [DOI] [PubMed] [Google Scholar]
  • 351.Roifman, C. M., C. P. Rao, H. M. Lederman, S. Lavi, P. Quinn, and E. W. Gelfand. 1986. Increased susceptibility to Mycoplasma infection in patients with hypogammaglobulinemia. Am. J. Med. 80:590-594. [DOI] [PubMed] [Google Scholar]
  • 352.Rollins, S., T. Colby, and F. Clayton. 1986. Open lung biopsy in Mycoplasma pneumoniae pneumonia. Arch. Pathol. Lab. Med. 110:34-41. [PubMed] [Google Scholar]
  • 353.Root-Bernstein, R. S., and S. H. Hobbs. 1991. Homologies between mycoplasma adhesion peptide, CD4 and class II MHC proteins: a possible mechanism for HIV-mycoplasma synergism in AIDS. Res. Immunol. 142:519-523. [DOI] [PubMed] [Google Scholar]
  • 354.Rottem, S. 2003. Interacton of mycoplasmas with host cells. Physiol. Rev. 83:417-432. [DOI] [PubMed] [Google Scholar]
  • 355.Rottem, S. 2002. Invasion of mycoplasmas into and fusion with host cells, p. 391-402. In S. Razin and R. Herrmann (ed.), Molecular biology and pathogenicity of mycoplasmas. Kluwer Academic/Plenum Publishers, New York, N.Y.
  • 356.Rudd, K. E. 2000. EcoGene: a genome sequence database for Escherichia coli K-12. Nucleic Acids Res. 28:60-64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 357.Ruuskanen, O., H. Nohynek, T. Ziegler, R. Capeding, H. Rikalainen, P. Huovinen, and M. Leinonen. 1992. Pneumonia in childhood: etiology and response to antimicrobial therapy. Eur. J. Clin. Microbiol. Infect. Dis. 11:217-223. [DOI] [PubMed] [Google Scholar]
  • 358.Sabato, A. R., D. M. Cooper, and Y. H. Thong. 1981. Transitory depression of immune function following Mycoplasma pneumoniae infection in children. Pediatr. Res. 15:813-816. [PubMed] [Google Scholar]
  • 359.Sabato, A. R., A. J. Martin, B. P. Marmion, T. W. Kok, and D. M. Cooper. 1984. Mycoplasma pneumoniae: acute illness, antibiotics, and subsequent pulmonary function. Arch. Dis. Child. 59:1034-1037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 360.Said, M. H., M. P. Layani, S. Colon, G. Faraj, C. Glastre, and P. Cochat. 1999. Mycoplasma pneumoniae-associated nephritis in children. Pediatr. Nephrol. 13:39-44. [DOI] [PubMed] [Google Scholar]
  • 361.Sakoulas, G. 2001. Brainstem and striatal encephalitis complicating Mycoplasma pneumoniae pneumonia: possible benefit of intravenous immunoglobulin. Pediatr. Infect. Dis. J. 20:543-545. [DOI] [PubMed] [Google Scholar]
  • 362.Salzman, M. B., S. K. Sood, M. L. Slavin, and L. G. Rubin. 1992. Ocular manifestations of Mycoplasma pneumoniae infection. Clin. Infect. Dis. 14:1137-1139. [DOI] [PubMed] [Google Scholar]
  • 363.Sande, M. A., F. Gadot, and R. P. Wenzel. 1975. Point source epidemic of Mycoplasma pneumoniae infection in a prosthodontics laboratory. Am. Rev. Respir. Dis. 112:213-217. [DOI] [PubMed] [Google Scholar]
  • 364.Sands, M. J., Jr., and R. Rosenthal. 1982. Progressive heart failure and death associated with Mycoplasma pneumoniae pneumonia. Chest 81:763-765. [DOI] [PubMed] [Google Scholar]
  • 365.Sasaki, T., T. Kenri, N. Okazaki, M. Iseki, R. Yamashita, M. Shintani, Y. Sasaki, and M. Yayoshi. 1996. Epidemiological study of Mycoplasma pneumoniae infections in Japan based on PCR-restriction fragment length polymorphism of the P1 cytadhesin gene. J. Clin. Microbiol. 34:447-449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 366.Schonwald, S., M. Gunjaca, L. Kolacny-Babic, V. Car, and M. Gosev. 1990. Comparison of azithromycin and erythromycin in the treatment of atypical pneumonias. J. Antimicrob. Chemother. 25(Suppl. A):123-126. [DOI] [PubMed] [Google Scholar]
  • 367.Schonwald, S., I. Kuzman, K. Oreskovic, V. Burek, V. Skerk, V. Car, D. Bozinovic, J. Culig, and S. Radosevic. 1999. Azithromycin: single 1.5 g dose in the treatment of patients with atypical pneumonia syndrome—a randomized study. Infection 27:198-202. [DOI] [PubMed] [Google Scholar]
  • 368.Schonwald, S., V. Skerk, I. Petricevic, V. Car, L. Majerus-Misic, and M. Gunjaca. 1991. Comparison of three-day and five-day courses of azithromycin in the treatment of atypical pneumonia. Eur. J. Clin. Microbiol. Infect. Dis. 10:877-880. [DOI] [PubMed] [Google Scholar]
  • 369.Schulman, P., T. C. Piemonte, and B. Singh. 1980. Acute renal failure, hemolytic anemia, and Mycoplasma pneumoniae. JAMA 244:1823-1824. [PubMed] [Google Scholar]
  • 370.Schwarze, J., G. Cieslewicz, A. Joetham, T. Ikemura, M. Makela, A. Dakham, L. D. Schultz, M. C. Lamers, and E. W. Gelfand. 2000. Critical roles for interleukin-4 and interleukin-5 during respiratory syncytial virus infection in the development of airway hyperresponsiveness after airway sensitization. Am. J. Respir. Crit. Care Med. 162:380-386. [DOI] [PubMed] [Google Scholar]
  • 371.Schwarze, J. E., M. Makela, G. Cieslewicz, A. Dakham, M. Lahn, T. Ikemura, A. Joetham, and E. W. Gelfand. 1999. Transfer of the enhancing effect of respiratory syncytial virus infection on subsequent allergic airway sensitization by T lymphocytes. J. Immunol. 163:5729-5734. [PubMed] [Google Scholar]
  • 372.Scully, R. E., E. J. Mark, W. F. McNeely, and B. U. McNeely. 1992. Case records of the Massachusetts General Hospital, case 5—1992. N. Engl. J. Med. 326:324-336.1728737 [Google Scholar]
  • 373.Seggev, J. S., I. Lis, R. Siman-Tov, R. Gutman, H. Abu-Samara, G. Schey, and Y. Naot. 1986. Mycoplasma pneumoniae is a frequent cause of exacerbation of bronchial asthma in adults. Ann. Allergy 57:263-265. [PubMed] [Google Scholar]
  • 374.Sequeira, W., E. Jones, and D. M. Bronson. 1981. Mycoplasma pneumoniae infection with arthritis and a varicella-like eruption. JAMA 246:1936-1937. [PubMed] [Google Scholar]
  • 375.Seto, S., G. Layh-Schmitt, T. Kenri, and M. Miyata. 2001. Visualization of the attachment organelle and cytadherence proteins of Mycoplasma pneumoniae by immunofluorescence microscopy. J. Bacteriol. 183:1621-1630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 376.Shepard, M. C. 1954. The recovery of pleuropneumonia-like organisms from Negro men with and without nongonococcal urethritis. Am. J. Syph. Gonor. Vener. Dis. 38:113-124. [PubMed] [Google Scholar]
  • 377.Shimizu, T., H. Mochizuki, M. Kato, M. Shigeta, A. Morikawa, and T. Hori. 1991. Immunoglobulin levels, number of eosinophils in the peripheral blood and bronchial hypersensitivity in children with Mycoplasma pneumoniae pneumonia. Arerugi 40:21-27. [PubMed] [Google Scholar]
  • 378.Shulman, S. T., J. G. Bartlett, W. A. J. Clyde, and E. M. Ayoub. 1972. The unusual severity of mycoplasmal pneumonia in children with sickle cell disease. N. Engl. J. Med. 287:164-167. [DOI] [PubMed] [Google Scholar]
  • 379.Siegler, D. I. 1973. Lung abscess associated with Mycoplasma pneumoniae infection. Br. J. Dis. Chest 67:123-127. [DOI] [PubMed] [Google Scholar]
  • 380.Sillis, M. 1990. The limitations of IgM assays in the serological diagnosis of Mycoplasma pneumoniae infections. J. Med. Microbiol. 33:253-258. [DOI] [PubMed] [Google Scholar]
  • 381.Simonian, N., and D. Janner. 1998. Pleural effusion, hepatitis and hemolytic anemia in a twelve-year-old male child. Pediatr. Infect. Dis. J. 17:173.-174, 176-177. [DOI] [PubMed] [Google Scholar]
  • 382.Skakni, L., A. Sardet, J. Just, J. Landman-Parker, J. Costil, N. Moniot-Ville, F. Bricout, and A. Garbarg-Chenon. 1992. Detection of Mycoplasma pneumoniae in clinical samples from pediatric patients by polymerase chain reaction. J. Clin. Microbiol. 30:2638-2643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 383.Smith, C. B., R. E. Kanner, C. A. Golden, M. R. Klauber, and A. D. Renzetti, Jr. 1980. Effect of viral infections on pulmonary function in patients with chronic obstructive pulmonary diseases. J. Infect. Dis. 141:271-280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 384.Smith, R., and L. Eviatar. 2000. Neurologic manifestations of Mycoplasma pneumoniae infections: diverse spectrum of diseases. A report of six cases and review of the literature. Clin. Pediatr. 39:195-201. [DOI] [PubMed] [Google Scholar]
  • 385.Somerson, N. L., B. E. Walls, and R. M. Chanock. 1965. Hemolysin of Mycoplasma pneumoniae: tentative identification as a peroxide. Science 150:226-228. [DOI] [PubMed] [Google Scholar]
  • 386.Squadrini, F., G. Lami, F. Pellegrino, G. Pinelli, M. Bavieri, A. Fontana, and A. Bisetti. 1988. Acute hepatitis complicating Mycoplasma pneumoniae infection. J. Infect. 16:201-202. [DOI] [PubMed] [Google Scholar]
  • 387.Stallman, N. D., B. C. Allan, and M. A. Wiemers. 1976. Infection with Mycoplasma pneumoniae: clinical and serological data on 286 patients. Med. J. Aust. 1:340-343. [DOI] [PubMed] [Google Scholar]
  • 388.Steinberg, P., R. L. Horswood, H. Brunner, and R. M. Chanock. 1971. Temperature-sensitive mutants of Mycoplasma pneumoniae. II. Response of hamsters. J. Infect. Dis. 124:179-187. [DOI] [PubMed] [Google Scholar]
  • 389.Steinberg, P., R. L. Horswood, and R. M. Chanock. 1969. Temperature-sensitive mutants of Mycoplasma pneumoniae. I. In vitro biologic properties. J. Infect. Dis. 120:217-224. [DOI] [PubMed] [Google Scholar]
  • 390.Stephan, J. L., C. Galambrun, B. Pozzetto, F. Grattard, and P. Bordigoni. 1999. Aplastic anemia after Mycoplasma pneumoniae infection: a report of two cases. J. Pediatr. Hematol. Oncol. 21:299-302. [DOI] [PubMed] [Google Scholar]
  • 391.Sterner, G., G. de Hevesy, G. Tunevall, and S. Wolontis. 1966. Acute respiratory illness with Mycoplasma pneumoniae. An outbreak in a home for children. Acta Paediatr. Scand. 55:280-286. [DOI] [PubMed] [Google Scholar]
  • 392.Stevens, D., P. G. Swift, P. G. Johnston, P. J. Kearney, B. D. Corner, and D. Burman. 1978. Mycoplasma pneumoniae infections in children. Arch. Dis. Child. 53:38-42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 393.Stopler, T., and D. Branski. 1986. Resistance of Mycoplasma pneumoniae to macrolides, lincomycin and streptogramin B. J. Antimicrob. Chemother. 18:359-364. [DOI] [PubMed] [Google Scholar]
  • 394.Stopler, T., C. B. Gerichter, and D. Branski. 1980. Antibiotic-resistant mutants of Mycoplasma pneumoniae. Isr. J. Med. Sci. 16:169-173. [PubMed] [Google Scholar]
  • 395.Stutman, H. R. 1987. Stevens-Johnson syndrome and Mycoplasma pneumoniae: evidence for cutaneous infection. J. Pediatr. 111:845-847. [DOI] [PubMed] [Google Scholar]
  • 396.Su, C. J., A. Chavoya, S. F. Dallo, and J. B. Baseman. 1990. Sequence divergency of the cytadhesin gene of Mycoplasma pneumoniae. Infect. Immun. 58:2669-2674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 397.Su, C. J., S. F. Dallo, A. Chavoya, and J. B. Baseman. 1993. Possible origin of sequence divergence in the P1 cytadhesin gene of Mycoplasma pneumoniae. Infect. Immun. 61:816-822. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 398.Suhs, R. H., and H. A. Feldman. 1966. Serologic epidemiologic studies with M. pneumoniae. II. Prevalence of antibodies in several populations. Am. J. Epidemiol. 83:357-365. [DOI] [PubMed] [Google Scholar]
  • 399.Suni, J., R. Vainionpaa, and T. Tuuminen. 2001. Multicenter evaluation of the novel enzyme immunoassay based on P1-enriched protein for the detection of Mycoplasma pneumoniae infection. J. Microbiol. Methods 47:65-71. [DOI] [PubMed] [Google Scholar]
  • 400.Szymanski, M., M. Petric, F. E. Saunders, and R. Tellier. 2002. Mycoplasma pneumoniae pericarditis demonstrated by polymerase chain reaction and electron microscopy. Clin. Infect. Dis. 34:E16-E17. [DOI] [PubMed] [Google Scholar]
  • 401.Talkington, D. F. Mycoplasma and Ureaplasma in central nervous system disorders. In M. Scheld, R. Whitley, and C. Marra (ed.), Infections of the central nervous system, in press. Lippincott, Williams & Wilkins, Philadelphia, Pa.
  • 402.Talkington, D. F., W. L. Thacker, D. W. Keller, and J. S. Jensen. 1998. Diagnosis of Mycoplasma pneumoniae infection in autopsy and open-lung biopsy tissues by nested PCR. J. Clin. Microbiol. 36:1151-1153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 403.Talkington, D. F., K. B. Waites, S. B. Schwartz, and R. E. Besser. 2001. Emerging from obscurity: understanding pulmonary and exrrapulmonary syndromes, pathogenesis, and epidemiology of human Mycoplasma pneumoniae infections, p. 57-84. In W. M. Scheld, W. A. Craig, and J. M. Hughes (ed.), Emerging Infections 5. American Society for Microbiology, Washington, D.C.
  • 404.Tanaka, G., Y. Nagatomo, Y. Kai, M. Matsuyama, M. Kuroki, T. Sasaki, K. Murai, A. Okayama, and H. Tsubouchi. 2002. Mycoplasma pneumonia of identical twin sisters with different clinical courses depending on the treatment. Kansenshogaku Zasshi 76:1040-1044. [DOI] [PubMed] [Google Scholar]
  • 405.Tanaka, H., H. Koba, S. Honma, F. Sugaya, and S. Abe. 1996. Relationships between radiological pattern and cell-mediated immune response in Mycoplasma pneumoniae pneumonia. Eur. Respir. J. 9:669-672. [DOI] [PubMed] [Google Scholar]
  • 406.Tanaka, H., M. Narita, S. Teramoto, T. Saikai, K. Oashi, T. Igarashi, and S. Abe. 2002. Role of interleukin-18 and T-helper type 1 cytokines in the development of Mycoplasma pneumoniae pneumonia in adults. Chest 121:1493-1497. [DOI] [PubMed] [Google Scholar]
  • 407.Taylor-Robinson, D. 1996. Cidal activity testing, p. 199-204. In J. G. Tully and S. Razin (ed.), Molecular and diagnostic procedures in mycoplasmology, vol. II. Academic Press, New York, N.Y.
  • 408.Taylor-Robinson, D. 1981. Mycoplasmal arthritis in man. Isr. J. Med. Sci. 17:616-621. [PubMed] [Google Scholar]
  • 409.Taylor-Robinson, D., J. Canchola, H. Fox, and R. M. Chanock. 1964. A newly identified oral mycoplasma (M. orale) and its relationship to other human mycoplasmas. Am. J. Hyg. 80:135-148. [DOI] [PubMed] [Google Scholar]
  • 410.Taylor-Robinson, D., J. M. Gumpel, A. Hill, and A. J. Swannell. 1978. Isolation of Mycoplasma pneumoniae from the synovial fluid of a hypogrammaglobulinaemic patient in a survey of patients with inflammatory polyarthritis. Ann. Rheum. Dis. 37:180-182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 411.Thacker, W. L., and D. F. Talkington. 2000. Analysis of complement fixation and commercial enzyme immunoassays for detection of antibodies to Mycoplasma pneumoniae in human serum. Clin. Diagn. Lab. Immunol. 7:778-780. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 412.Thacker, W. L., and D. F. Talkington. 1995. Comparison of two rapid commercial tests with complement fixation for serologic diagnosis of Mycoplasma pneumoniae infections. J. Clin. Microbiol. 33:1212-1214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 413.Thomas, N. H., J. E. Collins, S. A. Robb, and R. O. Robinson. 1993. Mycoplasma pneumoniae infection and neurological disease. Arch. Dis. Child. 69:573-576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 414.Thumerelle, C., A. Deschildre, C. Bouquillon, C. Santos, A. Sardet, M. Scalbert, L. Delbecque, P. Debray, A. Dewilde, D. Turck, and F. Leclerc. 2003. Role of viruses and atypical bacteria in exacerbations of asthma in hospitalized children: a prospective study in the Nord-Pas de Calais region (France). Pediatr. Pulmonol. 35:75-82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 415.Tjhie, J. H., P. H. Savelkoul, and C. M. Vandenbroucke-Grauls. 1997. Polymerase chain reaction evaluation for Mycoplasma pneumoniae. J. Infect. Dis. 176:1124-1125. [DOI] [PubMed] [Google Scholar]
  • 416.Tjhie, J. H., E. M. van de Putte, K. Haasnoot, A. J. van den Brule, and C. M. Vandenbroucke-Grauls. 1997. Fatal encephalitis caused by Mycoplasma pneumoniae in a 9-year-old girl. Scand. J. Infect. Dis. 29:424-425. [DOI] [PubMed] [Google Scholar]
  • 417.Tjhie, J. H., F. J. van Kuppeveld, R. Roosendaal, W. J. Melchers, R. Gordijn, D. M. MacLaren, J. M. Walboomers, C. J. Meijer, and A. J. van den Brule. 1994. Direct PCR enables detection of Mycoplasma pneumoniae in patients with respiratory tract infections. J. Clin. Microbiol. 32:11-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 418.Ton, C. Y. W., C. Donnelly, G. Harvey, and M. Sillis. 1999. Multiplex polymerase chain reaction for the simultaneous detection of Mycoplasma pneumoniae. Chlamydia pneumoniae, and Chlamydia psittaci in respiratory samples. J. Clin. Pathol. 52:257-263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 419.Tryon, V. V., and J. B. Baseman. 1987. The acquisition of human lactoferrin by Mycoplasma pneumoniae. Microb. Pathog. 3:437-443. [DOI] [PubMed] [Google Scholar]
  • 420.Tryon, V. V., and J. B. Baseman. 1992. Pathogenic determinants and mechanisms, p. 457-471. In J. Maniloff (ed.), Mycoplasmas: molecular biology and pathogenesis. American Society for Microbiology, Washington, D.C.
  • 421.Tully, J. G., and S. Razin (ed.). 1996. Molecular and diagnostic procedures in mycoplasmology, vol. 2. Diagnostic procedures. Academic Press, New York, N.Y.
  • 422.Tully, J. G., D. L. Rose, R. F. Whitcomb, and R. P. Wenzel. 1979. Enhanced isolation of Mycoplasma pneumoniae from throat washings with a newly-modified culture medium. J. Infect. Dis. 139:478-482. [DOI] [PubMed] [Google Scholar]
  • 423.Tully, J. G., D. Taylor-Robinson, D. L. Rose, R. M. Cole, and J. M. Bove. 1983. Mycoplasma genitalium, a new species from the human urogenital tract. Int. J. Syst. Bacteriol. 33:387-396. [Google Scholar]
  • 424.Turtzo, D. F., and P. K. Ghatak. 1976. Acute hemolytic anemia with Mycoplasma pneumoniae pneumonia. JAMA 236:1140-1141. [PubMed] [Google Scholar]
  • 425.Uldum, S. A., J. S. Jensen, J. Sondergard-Andersen, and K. Lind. 1992. Enzyme immunoassay for detection of immunoglobulin M (IgM) and IgG antibodies to Mycoplasma pneumoniae. J. Clin. Microbiol. 30:1198-1204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 426.Ursi, D., J. P. Ursi, M. Ieven, M. Docx, P. Van Reempts, and S. R. Pattyn. 1995. Congenital pneumonia due to Mycoplasma pneumoniae. Arch. Dis. Child. Fetal Neonatal Ed. 72:F118-F20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 427.Ursi, J. P., D. Ursi, M. Ieven, and S. R. Pattyn. 1992. Utility of an internal control for the polymerase chain reaction. Application to detection of Mycoplasma pneumoniae in clinical specimens. APMIS 100:635-639. [DOI] [PubMed] [Google Scholar]
  • 428.van Kuppeveld, F. J., K. E. Johansson, J. M. Galama, J. Kissing, G. Bolske, E. Hjelm, J. T. van der Logt, and W. J. Melchers. 1994. 16S rRNA based polymerase chain reaction compared with culture and serological methods for diagnosis of Mycoplasma pneumoniae infection. Eur. J. Clin. Microbiol. Infect. Dis. 13:401-405. [DOI] [PubMed] [Google Scholar]
  • 429.Vekris, A., F. Bauduer, S. Maillet, C. Bebear, and J. Bonnet. 1995. Improved microplate immunoenzymatic assay of PCR products for rapid detection of Mycoplasma pneumoniae. Mol. Cell. Probes 9:25-31. [DOI] [PubMed] [Google Scholar]
  • 430.Vitullo, B. B., S. O'Regan, J. P. de Chadarevian, and B. S. Kaplan. 1978. Mycoplasma pneumonia associated with acute glomerulonephritis. Nephron 21:284-288. [DOI] [PubMed] [Google Scholar]
  • 431.Von Bonsdorff, M., A. Ponka, and T. Tornroth. 1984. Mycoplasmal pneumonia associated with mesangiocapillary glomerulonephritis type II (dense deposit disease). Acta Med. Scand. 216:427-429. [PubMed] [Google Scholar]
  • 432.Wachowski, O., S. Demirakca, K.-M. Muller, and W. Scheurlen. 2003. Mycoplasma pneumoniae associated organising pneumonia in a 10 year old boy. Arch. Dis. Child. 88:270-272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 433.Waites, K. B. 2001. Mycoplasma, p. 649-655. In D. Schlossberg (ed.), Current therapy of infectious disease. Mosby, St. Louis, Mo.
  • 434.Waites, K. B., C. M. Bebear, J. A. Robertson, D. F. Talkington, and G. E. Kenny (ed.). 2001. Cumitech 34, Laboratory diagnosis of mycoplasmal infections. American Society for Microbiology, Washington, D.C.
  • 435.Waites, K. B., D. M. Crabb, X. Bing, and L. B. Duffy. 2003. In vitro susceptibilities to and bactericidal activities of garenoxacin (BMS-284756) and other antimicrobial agents against human mycoplasmas and ureaplasmas. Antimicrob. Agents Chemother. 47:161-165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 436.Waites, K. B., D. M. Crabb, and L. B. Duffy. 2003. In vitro activities of ABT-773 and other antimicrobials against human mycoplasmas. Antimicrob. Agents Chemother. 47:39-42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 437.Waites, K. B., D. M. Crabb, and L. B. Duffy. 2003. Inhibitory and bactericidal activities of gemifloxacin and other antimicrobials against Mycoplasma pneumoniae. Int. J. Antimicrob. Agents 21:574-577. [DOI] [PubMed] [Google Scholar]
  • 438.Waites, K. B., Y. Rikihisa, and D. Taylor-Robinson. 2003. Mycoplasma and ureaplasma, p. 972-990. In P. R. Murray, E. J. Baron, J. H. Jorgensen, M. A. Pfaller, and R. H. Yolken (ed.), Manual of clinical microbiology, 8th ed. American Society for Microbiology, Washington, D.C.
  • 439.Waites, K. B., D. F. Talkington, and C. M. Bebear. 2002. Mycoplasmas, p. 201-224. In A. L. Truant (ed.), Manual of commercial methods in clinical microbiology. American Society for Microbiology, Washington, D.C.
  • 440.Watkins-Riedel, T., G. Stanek, and F. Daxboeck. 2001. Comparison of SeroMP IgA with four other commercial assays for serodiagnosis of Mycoplasma pneumoniae pneumonia. Diagn. Microbiol. Infect. Dis. 40:21-25. [DOI] [PubMed] [Google Scholar]
  • 441.Weinstein, M. P., and C. B. Hall. 1974. Mycoplasma pneumoniae infection associated with migratory polyarthritis. Am. J. Dis. Child. 127:125-126. [DOI] [PubMed] [Google Scholar]
  • 442.Williamson, J., B. P. Marmion, T. Kok, R. Antic, and R. J. Harris. 1994. Confirmation of fatal Mycoplasma pneumoniae infection by polymerase chain reaction detection of the adhesin gene in fixed lung tissue. J. Infect. Dis. 170:1052-1053. [DOI] [PubMed] [Google Scholar]
  • 443.Williamson, J., B. P. Marmion, D. A. Worswick, T. W. Kok, G. Tannock, R. Herd, and R. J. Harris. 1992. Laboratory diagnosis of Mycoplasma pneumoniae infection. 4. Antigen capture and PCR-gene amplification for detection of the mycoplasma: problems of clinical correlation. Epidemiol. Infect. 109:519-537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 444.Wilson, M. H., and A. M. Collier. 1976. Ultrastructural study of Mycoplasma pneumoniae in organ culture. J. Bacteriol. 125:332-339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 445.Wreghitt, T. G., and M. Sillis. 1985. A micro-capture ELISA for detecting Mycoplasma pneumoniae IgM: comparison with indirect immunofluorescence and indirect ELISA. J. Hyg 94:217-227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 446.Wubbel, L., L. Muniz, A. Ahmed, M. Trujillo, C. Carubelli, C. McCoig, T. Abramo, M. Leinonen, and G. H. McCracken, Jr. 1999. Etiology and treatment of community-acquired pneumonia in ambulatory children. Pediatr. Infect. Dis. J. 18:98-104. [DOI] [PubMed] [Google Scholar]
  • 447.Yang, J., W. Craig Hooper, D. J. Phillips, and D. F. Talkington. 2003. Interleukin-1beta responses to Mycoplasma pneumoniae infection are cell-type specific. Microb. Pathog. 34:17-25. [DOI] [PubMed] [Google Scholar]
  • 448.Yang, J., W. C. Hooper, D. J. Phillips, and D. F. Talkington. 2002. Regulation of proinflammatory cytokines in human lung epithelial cells infected with Mycoplasma pneumoniae. Infect. Immun. 70:3649-3655. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 449.Yayoshi, M., E. Hayatsu, and M. Yoshioka. 1989. Protective effects of Mycoplasma pneumoniae live vaccine or its hyperimmune serum on the experimental infection in mice. Kansenshogaku Zasshi 63:684-691. [DOI] [PubMed] [Google Scholar]
  • 450.Yayoshi, M., T. Sasaki, and M. Yoshioka. 1992. Relationship between an 85 kDa protein and the protective effects of Mycoplasma pneumoniae. Microbiol. Immunol. 36:455-464. [DOI] [PubMed] [Google Scholar]
  • 451.Yogev, D., G. F. Browning, and K. S. Wise. 2002. Genetic mechanisms of surface variation, p. 417-444. In S. Razin and R. Herrmann (ed.), Molecular biology and pathogenicity of mycoplasmas. Kluwer Academic/Plenum Publishers, New York, N.Y.

Articles from Clinical Microbiology Reviews are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES