Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Apr 1.
Published in final edited form as: Semin Perinatol. 2013 Apr;37(2):94–101. doi: 10.1053/j.semperi.2013.01.005

Ureaplasma and BPD

Suhas G Kallapur 1, Boris W Kramer 2, Alan H Jobe 1
PMCID: PMC3628630  NIHMSID: NIHMS453367  PMID: 23582963

Abstract

Ureaplasma is an organism with low virulence and is a commensal of the lower genito-urinary tract in females. From here, it can gain entry in the amniotic fluid to cause inflammation in the amniotic compartment during pregnancy. Ureaplasma spp. are the most common organisms isolated from women with chorioamnionitis. Ureaplasma spp. are associated with increased risk for preterm labor and morbidity in the preterm neonate. However, there is some controversy regarding the importance of Ureaplasma in the pathogenesis of bronchopulmonary dysplasia (BPD). This article will review the microbiology of Ureaplasma, host innate immune responses, and the pathology of lung injury in animal models of Ureaplasma chorioamnionitis. We will review epidemiological studies of Ureaplasma and BPD in preterm infants and efficacy of antibiotics in preventing preterm labor and BPD.


The Ureaplasmas are generally regarded as commensals of the lower genital tract in both males and females1,2. However, Ureaplasma infection of the upper genital tract during pregnancy is associated with adverse pregnancy outcomes including preterm births and neonatal morbidity3.

Microbiology of Ureaplasma Species

Ureaplasma spp. are among the smallest free-living, self-replicating microorganisms. They have an extremely low G+C content of 25.5% (or the highest A+T composition) within open reading frames of any prokaryotes sequenced to date. Ureaplasmas have evolved from Gram-positive bacteria by degenerative evolution to lose the peptidoglycan cell wall4. As their name suggests, Ureaplasmas utilize urea as their sole source of carbon, producing ammonia as a metabolic product5. Currently, there are 2 identified species of Ureaplasmas that infect humans that are divided into 14 antigenically distinct Serovars6. Prior to 2000, the 14 Serovars were considered to be the single species U. urealyticum. Their separation into U. parvum (Serovars 1, 3, 6 and 14) and U. urealyticum (Serovars 2, 4, 5, 7–13) is based on phylogenetic evidence7. However, the new taxonomic classification has not been fully adopted in the literature and the 14 Serovars are often referred to as U. urealyticum.

Virulence of Ureaplasma Species

Several proteins have been proposed as virulence factors. The mba gene encodes for MBA, the major surface-exposed lipoprotein. MBA is thought to be the major virulence factor of Ureaplasma spp. and is the predominant antigen recognized by the host immune system during infection4. Ureaplasmas can alter the expression of their MBA in order to evade host immune responses and maintain chronicity of infection810. Ureaplasma Phospholipase A and C activities were identified by in vitro hydrolysis of an artificial phosphate ester11. The phospholipases could potentially generate prostaglandins – a known trigger of labor11. Similarly, an IgA protease activity, which could destroy mucosal IgA, was demonstrated in Ureaplasma12. However, genome analyses of all the Ureaplasma Serovars did not find sequences for either the IgA protease or phospholipase A or C13. However there was a gene encoding a phospholipase D domain containing protein13. The urease activity of Ureaplasma generates ammonia from the cleavage of urea5, which can cause toxicity to host tissues due to change in pH. To date there is no definitive identification of specific virulence factors in Ureaplasma.

Recent studies demonstrated that horizontal gene transfer occurs in Ureaplasma isolated from clinical samples, resulting in genetic hybrid forms of Ureaplasma Serovars, implying unstable genotypes during the course of infection14. Nevertheless, there is no evidence of Serovar specific pathogenic effects or severity of infection10,13. An experiment in the sheep illustrates the instability of Ureaplasma phenotype. Sheep were given an intra-amniotic injection with either a virulent or non-virulent-derived Ureaplasma clones (both Ureaplasma parvum Serovar 6) at 55 days of gestation. Virulence was defined as Ureaplasma recovered from amniotic fluid of sheep with severe chorioamnionitis or minimal chorioamnionitis. Although both Ureaplasma isolates caused chronic colonization and fetal inflammation, the severity of chorioamnionitis or fetal inflammation was not different after 70d of colonization of the fetal compartment15. Therefore the original phenotype of Ureaplasma virulence was not sustained during the chronic infection. Virulence and persistence will also be influenced by the ability of microorganisms to form biofilms. The majority of clinical isolates of Ureaplasma form biofilms16.

Ureaplasma as Perinatal Pathogens Causing Preterm Birth

The microbial invasion of amniotic cavity associated with preterm birth is relatively unique among infectious diseases in humans. The microbes are mostly opportunistic commensal vaginal organisms of low pathogenicity in otherwise healthy women. Polymicrobial growth is common from amniotic fluid from chorioamnionitis, and Ureaplasma spp. are the organisms most frequently isolated, although they rarely cause infections elsewhere13,17. The strongest evidence that Ureaplasma can cause preterm labor is from experiments in Rhesus macaques. Intra-amniotic injection of Ureaplasma parvum or the related organism Mycoplasma hominis induced chorioamnionitis, fetal inflammation and preterm labor18. In human pregnancies, several studies demonstrate the association of Ureaplasma species and preterm labor. Using PCR amplification of 16S ribosomal DNA, Ureaplasma species were the most common microbial genus identified in amniotic fluid of women with preterm premature rupture of membranes17. These organisms can be isolated in the amniotic fluid in the second trimester and can remain clinically silent for several weeks to months19. Gerber et al sampled amniotic fluid by amniocentesis from 254 asymptomatic women at 15–17 weeks' gestation, and Ureaplasma species were identified in 11% subjects. Preterm labor occurred in 59% of Ureaplasma-positive women compared with 4% Ureaplasma-negative women. Preterm births were also significantly higher in Ureaplasma-positive women compared to Ureaplasma-negative women20. Isolation of Ureaplasma from the amniotic fluid was associated with chorioamnionitis and inflammatory mediators in the amniotic fluid – known triggers of preterm labor21,22. The current hypothesis for infection related preterm labor is that the organisms from lower genital tract (e.g. Ureaplasma) ascend through the cervix into the choriodecidual space. The localized inflammation in this region traverses through the chorion-amnion and disseminates in the amniotic fluid. This leads to a generalized inflammation of the amnion and the chorio-decidua resulting in the production of pro-inflammatory mediators such as interleukin-1beta (IL-1β), IL-6 and prostaglandins that ultimately initiate preterm labor23.

Host Response to Ureaplasma Infection

Microbial recognition by innate immune systems can be mediated by a variety of germline-encoded receptors, including Toll-like receptors (TLRs), RIG-like receptors (RLRs), Nod-like receptors (NLRs), and cytosolic DNA sensors such as the HIN200 family member AIM224. Ureaplasma sp. lack a gram-negative or gram-positive bacterial cell wall, thus are devoid of lipopolysaccharides or peptidoglycans – the microbial products that are potent activators of TR4 (lipopolysaccharide or LPS) and the TLR2 or NOD1/2 (peptidoglycan) pathways. Nevertheless, placental leukocytes or neonatal monocytes exposed in vitro to Ureaplasma spp. induce the release of inflammatory cytokines25,26. Shimizu et al27 showed that U. parvum lipoproteins, including the multi-banded antigen activate nuclear factor-kappa B (NF-κB) in reporter cell lines via TLR1, TLR2 and TLR6 signaling. Peltier et al.28 found that the macrophage-stimulating activity from U. urealyticum is mainly due to lipoproteins, and signaling involving TLR2 or TLR4 receptors. Mechanisms of immune alterations induced by Ureaplasma in vivo have not been identified. However, the Ureaplasmas do induce antibody production in both humans and animals15,29.

Ureaplasmas are generally considered to be microorganisms with low virulence. However, it is conceivable that in immunodeficient hosts, the organism may have enhanced virulence. Surfactant protein-A (SP-A) is an innate host defense molecule that is secreted by the lung and is deficient in preterm infants30. Compared to wild type mice, SP-A deficient mice had delayed clearance of Ureaplasma from the lungs, increased of inflammatory cells and pro-inflammatory cytokine expression31. This observation may be relevant to preterm fetuses and neonates who will have low levels of SP-A and other innate host defense factors in the lungs.

Ureaplasma and Immune Modulation

Chronic infections can induce a state of immune paralysis called endotoxin tolerance. Thus leukocytes from patients with sepsis or recovering from typhoid fever can have endotoxin tolerance32, 33. We reported that in sheep repeated intra-amniotic injections of LPS (endotoxin), a constituent of cell wall of gram negative bacteria, induces tolerance to a variety of toll-like receptor agonists in the preterm fetus3436. Interestingly, chronic intra-amniotic colonization/infection by Ureaplasma also profoundly diminished responses to intra-amniotic LPS in the preterm fetal sheep37 (Figure 1). A benefit of endotoxin tolerance is decreased inflammation in the host and therefore decreased organ injury. However, endotoxin tolerance also can increase susceptibility to infections due to suppression of innate immune responses. These observations in preterm fetuses may have direct relevance to immune responses of newborns at risk of developing bronchopulmonary dysplasia (BPD).

Figure 1. Immune modulation: Chronic exposure to Ureaplasma (UP) downregulated LPS responses in preterm fetal Sheep.

Figure 1

IL-1β mRNA in fetal lung was measured by rt-PCR after different intraamniotic injections at different intervals prior to preterm delivery at 125d (80% gestation). The mean mRNA level in the control group was set to 1. mRNA levels of each group were expressed as fold increase relative to controls. Exposure to UP alone did not induce IL-1β expression but prevented LPS induced IL-1β expression (*p < 0.05 vs control). Figure based on data from37.

Ureaplasma Species and Lung Inflammation in the Developing Lung – Animal Studies

Ureaplasma infection of the developing lung has been studied primarily in sheep and non-human primates. Intra-amniotic injection of Ureaplasma in early gestation sheep resulted in efficient colonization and a 5-log increase in the Ureaplasma in amniotic fluid counts that persisted for 3 months to term with very little overt adverse effects in the ewe, consistent with a commensal-like host response15. Interestingly, after a chronic exposure to intra-amniotic Ureaplasma, all of the fetal lungs were colonized10. Although Ureaplasma was not cultured from either the fetal or maternal blood after an intra-amniotic injection, about 50% of the ewes had an IgG antibody response10,15. Although chorioamnionitis of varying severity was demonstrated in a majority of the sheep, about 10% of the fetal membranes showed no chorioamnionitis despite recovery of high titers of Ureaplasma from the amniotic fluid over 3 months10. These experiments illustrate the complexities in understanding the host response to a Ureaplasma exposure.

Recruitment to and activation of inflammatory cells in the fetal lung could be detected as early as 3d after intra-amniotic injection of live Ureaplasma in sheep38 (Figure 2). Both monocytes and neutrophils increased, and MHCII expression in the monocytes increased 14d after intra-amniotic Ureaplasma injection, consistent with maturation of the monocytic cells38. The inflammatory cell infiltration was focal in nature and no areas of consolidation or “pneumonia” were detected. This modest microscopic inflammation would not be detectable by standard X-rays. The inflammatory infiltrate was accompanied by modest increases in the pulmonary expression of the pro-inflammatory cytokines/chemokines IL-1β, IL-6 and IL-8 within 1 week that persisted for at least 6 weeks39,40.

Figure 2. Modest time-dependent fetal lung inflammation induced by UP in preterm fetal Sheep.

Figure 2

A: Intraamniotic injection of Ureaplasma parvum serovar 3 into the amniotic cavity induced an increase in IL-8 mRNA in the fetal lung. B: Neutrophils in broncho-alveolar lavage fluid. Results are given per kilogram body weight. The Ureaplasma parvum serovar 3 injection induced a pulmonary inflammation at 3 days, 7 days, 14 days or 70 days before preterm delivery. *, p <0.05 versus control. Figure based on data from38,41.

The modest lung inflammation was followed by the counter-intuitive observation of significant increases in lung gas volumes and surfactant lipids in the preterm fetal sheep. This early lung maturation was first detected in the preterm fetal lungs 3 weeks after intra-amniotic Ureaplasma injection and persisted for 10 weeks despite continuous exposures41 (Figure 3). Although these striking effects on lung physiology are consistent with clinical “lung maturation”, they probably represent “dysmaturation” since the improved lung physiology was accompanied by evidence of impaired lung development38. Fourteen days after intra-amniotic Ureaplasma injection, preterm fetal sheep delivered at 80% gestation had decreased elastic foci and increased smooth muscle around bronchioles and pulmonary artery/arterioles38 (Figure 4). These changes in elastin and smooth muscle are similar to those reported for infants with BPD42. Therefore the benefits from acute improvement in lung physiology were somewhat offset by changes in lung architecture. However, alveolar and pulmonary vascular development was no different between the controls and fetus exposed to Ureaplasma for 70 d43. Therefore the pathologic changes detected at 14d were not sustained during more extended exposures, suggesting that repair mechanisms must be operative.

Figure 3. Increases in lung gas volume induced by UP in preterm fetal Sheep.

Figure 3

The lung gas volume at 40 cm H2O increased after intraamniotic injection of Ureaplasma parvum serovar 3 at 3 days, 7 days, 14 days or 70 days before preterm delivery. *, p <0.05 versus control. . Figure based on data from38.

Figure 4. Lung remodeling induced by UP in preterm fetal Sheep.

Figure 4

Elastin foci in the lung after intraamniotic injection of Ureaplasma parvum serovar 3 after 3, 7 and 14 days. The number of elastin foci in the control group was 12 and set to 1 for ease of comparison. Changes are shown as fold increase or decrease over control. *, p <0.05 versus control. Figure based on data from38.

In Rhesus macaques, intra-amniotic injection of Ureaplasma caused chorioamnionitis, preterm labor and delivery within 15d18. Both Ureaplasma parvum and the related organism Mycoplasma hominis given by the intra-amniotic route caused fetal pneumonia characterized by increased neutrophils and macrophages and alveolar type II cell proliferation indicating injury18. In another experiment, pregnant baboons were given intra-amniotic Ureaplasma at 65% gestation. The preterm baboons were delivered operatively 2–3d later and were given mechanical ventilation for 14d. Half of the neonatal baboons cleared Ureaplasma from their airways, while the remaining half had persistent Ureaplasma colonization of the lungs44. The neonatal baboons with persistent Ureaplasma colonization had lung inflammation and worse lung function compared to those animals that cleared the Ureaplasma from the lungs. Lung pathology at autopsy of the persistently Ureaplasma positive at 14d revealed a mixed monocytic, lymphocytic and to a lesser extent neutrophilic infiltrate in the lungs44. In general, the lung pathology after intrauterine Ureaplasma exposure was more severe in non-human primates compared to sheep, suggesting species differences in susceptibility to Ureaplasma.

Ureaplasma Species and Lung Inflammation in the Developing Lung – Human Studies

The lung pathology of archived autopsy specimens from Ureaplasma infected preterm infants demonstrated increased lung fibrosis, elastic fiber accumulation, smooth muscle actin, and increased tumor necrosis factor alpha (TNFα) and transforming growth factor beta 1 (TGFβ1) immunoreactivity45,46. Elevated maternal and fetal or newborn antibody titers to Ureaplasma correlated with an increased incidence of stillbirth and infants with fatal neonatal respiratory disease29. The findings from these limited human studies are consistent with lung pathology described in animal studies.

Pathogenesis of BPD

The most predictive risk factors for development of BPD are gestational age, birth-weight, duration of respiratory support, fraction of inspired oxygen (FiO2), race and gender47. Although these risk factors hold true on a population basis, individual infants who develop BPD have different trajectories toward developing BPD. For instance, if the need for supplemental oxygen is used as a proxy for lung injury, then three patterns (or subsets) of BPD are discernible48,49. In a study of preterm infants <28 week gestation at birth, 20% of the infants received a consistently low FiO2, 38% had pulmonary deterioration (low initial FiO2 followed by increased oxygen need), and the remaining 43% had consistently high FiO2 needs48. The incidence of BPD was 17% in the consistently low FiO2 group, 51% in the pulmonary deterioration group, and 67% in the early and persistent pulmonary dysfunction group. These patterns of oxygen use are apparent soon after birth and must result from complex interactions between antenatal exposures and postnatal management50. For example, the chronic, indolent chorioamnionitis associated with very preterm birth can induce early lung maturation and cause endotoxin tolerance with a suppressed inflammatory response to a secondarily inflammatory exposure (Figure 1). However, chorioamnionitis associated with more severe lung inflammation may cause a pneumonia that is seldom distinguished clinically from respiratory distress syndrome (RDS) and/or may interfere with a surfactant treatment response51. Fetal growth restriction greatly increases the risk of BPD52. Subsequent early life experiences such as lung injury with the initiation of ventilation are further variables that contribute to the initial severity of the lung disease50. However, some antenatal exposures such as chorioamnionitis could ameliorate inflammation due to a tolerance phenomenon and have beneficial effects on surfactant production and lung maturation. Conceptually therefore, these different insults during fetal or neonatal epochs can interact in a synergistic fashion to increase the inflammation or conversely may dampen the net inflammatory exposure. The interactions between the different insults can therefore modulate the risk for BPD.

Clinical and Epidemiological Studies of Ureaplasma and BPD

Ureaplasma spp. are the organisms most frequently associated with chorioamnionitis and very preterm delivery53. However, the contribution of Ureaplasma or chorioamnionitis to BPD is less clear. Several single center studies reported the association of Ureaplasma in the respiratory secretions of newborns with the development of BPD5458. Some studies used the BPD definition of oxygen supplementation at 28d, while others used the more current definition of BPD as the need for oxygen supplementation at 36 week post-menstrual age. However, other studies of mechanically ventilated preterm infants did not associate Ureaplasma in the respiratory tract with BPD5962. Similarly, some studies reported an association of BPD with chorioamnionitis63,64, but more recent reports do not find a clear association65,66. Indeed some studies found that chorioamnionitis correlated with a decrease in the development of BPD67.

To better define the role of Ureaplasma in the development of BPD, Schelonka et al performed a meta-analysis of published studies using stringent entry criteria68. Twenty-three studies with an aggregate of 2216 infants reported BPD using the 28 week definition, and 8 studies with 751 infants reported BPD using the 36 week definition. Overall, the relative risk for BPD in Ureaplasma positive infants was 1.6 (C.I. 1.1–2.3) for BPD at 36 weeks, or 2.8 (C.I. 2.3–3.5) for BPD at 28d compared to Ureaplasma negative group. However, there was substantial heterogeneity in the studies with the greatest contribution to the BPD association from the studies reporting fewer than 100 infants, suggesting a reporting bias68.

Why might there be such heterogeneity in the studies? Different patient populations, divergent ventilator management practices, non-uniform Ureaplasma culture techniques and publication bias may contribute to the discrepant results. However, another possible explanation is that Ureaplasma may cause both some benefit and some harm, and the differential contribution of harm vs. benefit in different populations may explain the variation. Ureaplasma in preterm neonates is acquired antenatally. Experiments in sheep clearly demonstrate that chorioamnionitis caused by Ureaplasma results in increased lung compliance3941. At least one study in preterm infants also reported a decreased incidence of RDS in infants with Ureaplasma isolated from tracheal aspirates58. Further, mechanical ventilation is a major contributor to BPD and more gentle ventilation (i.e. continuous positive airway pressure or CPAP) tends to decrease the incidence of BPD6973. Therefore, it is possible that Ureaplasma induced increases in surfactant and thus the improved lung function may decrease the need for mechanical ventilation and therefore afford protection against BPD. Alternatively, Ureaplasma may be a priming insult and mechanical ventilation following Ureaplasma may increase lung injury. Indeed, a recent study of 122 preterm infants < 1000g birth weight from Japan demonstrated no association between Ureaplasma cultured in the tracheal aspirate with BPD. However, the infants that received prolonged mechanical ventilation for ≥ 2 weeks and were Ureaplasma positive had an increased risk for BPD compared to Ureaplasma negative infants on prolonged mechanical ventilation74. This synergy between Ureaplasma colonization and subsequent injurious exposures in preterm infants may partly explain the variable increased risk for BPD in different patient populations.

Treatment with Antimicrobial Agents

Since Ureaplasma is the most common organism isolated in women with chorioamnionitis it is instructive to review antibiotic trials for chorioamnionitis. Antenatal trials of antimicrobial use in the setting of preterm labor have yielded mixed results. Mercer et al.75, randomized 614 women with preterm premature rupture of membranes between 24–32 weeks to ampicillin plus erythromycin or placebo for a 7d treatment course75. The women randomized to antibiotics had a higher latency and the infants had a lower rate of RDS, necrotizing enterocolitis (NEC) and BPD. In the ORACLE I randomized trial, 4286 women with preterm premature rupture of membranes <37 weeks gestation were randomized to erythromycin only, amoxicillin plus clavulanic acid only, both, or placebo76. Any antibiotic use was associated with increased latency but only erythromycin decreased the composite outcome of death or BPD or intraventricular hemorrhage (IVH) or periventricular leukomalacia76. The amoxicillin and clavulanic acid combination increased the incidence of NEC. In the ORACLE II trial, the same antibiotic regimens (or placebo) as ORACLE I was given to 6295 women <37 week gestation with intact membranes but spontaneous preterm labor77. In this trial, antibiotic use for 10d decreased maternal infections but had no benefit for neonatal mortality or morbidity. Additionally, results of a 7-yr follow-up of the ORACLE II trial found that children whose mothers randomized to either erythromycin or amoxicillin-clavulanic acid had increased rates of cerebral palsy, suggesting the use of antibiotics during pregnancy may have some inherent risks78. The current recommendations by the American College of Obstetrics and Gynecology (ACOG) is to use antibiotics as prophylaxis for group B streptococcal infection and use broad spectrum antibiotics for women with preterm premature rupture of membranes or for women with diagnosed acute perinatal infections79. However, ACOG recommends not using antibiotics for women with preterm labor with intact membranes79.

The pharmacokinetics of maternal antibiotic treatments are not well understood in the amniotic fluid or the fetus80. In particular, the penetration of macrolides to the amniotic compartment after a systemic administration appears to be poor in sheep81, which might explain the lack of efficacy of antibiotics in women with intact membranes. However, in a Rhesus macaque model of chorioamnionitis induced by intra-amniotic injection of Ureaplasma, maternal intravenous administration of azithromycin effectively delayed preterm delivery and reduced fetal lung inflammation82. These experiments with disparate results in different species reinforce the need for further work to elucidate pharmacokinetics and efficacy of macrolides to treat the fetal compartment.

A limited number of studies report the use of macrolides for eradication of Ureaplasma in neonates at risk for developing BPD. Infants in two trials with limited numbers of infants <30 weeks gestation on mechanical ventilation were treated with erythromycin. Ureaplasma status was unknown at the time of initiation of antibiotics in the Lyon83 et al. study. In contrast, Jonsson et al.84 recruited only infants with Ureaplasma positive tracheal aspirates. Both studies failed to show a benefit of erythromycin for prevention of death or BPD. Another macrolide, azithromycin is actively concentrated in the macrophages and has anti-inflammatory effects. Ballard et al85 randomized 220 mechanically ventilated preterm infants ≤ 1250g in the first 3d of life. They treated the infants with azithromycin or placebo for up to 6 weeks. Overall, azithromycin did not protect against death or BPD, but in the subgroup of infants who had Ureaplasma isolated in tracheal aspirates, azithromycin decreased BPD from 94% in the placebo arm to 73% in the antibiotic arm. Recently, a randomized trial from Turkey evaluated clarithromycin in 74 preterm infants 750–1250g at birth who tested positive for Ureaplasma in the nasopharynx in the first 3d of life86. Clarithromycin treatment for 10d decreased the incidence of BPD. Although no significant side-effects were reported in these studies, antibiotics must be used with caution in preterm neonates since prolonged antibiotic exposure has been associated with increased rates of NEC or late onset sepsis87,88.

Summary and Conclusions

Ureaplasma species are the organisms most associated with chorioamnionitis and preterm delivery. Although there is controversy regarding the role of Ureaplasma in BPD, experimental evidence and clinical/epidemiological data demonstrate that Ureaplasma species can increase the risk for BPD in preterm infants. Antenatal exposure to Ureaplasma increases surfactant production, increases lung volumes and induces pulmonary inflammation with disordered alveolar development in preterm sheep. Experimental evidence demonstrates that Ureaplasma can suppress signaling by LPS and thus modulate the innate immune system. In preterm infants, Ureaplasma colonization can prime the respiratory tract and synergistically increase ventilator induced lung injury. Treatment with macrolide antibiotics for Ureaplasma in preterm infants has not met with much success.

Acknowledgments

Funded by NIH grant HD57869 (to SGK)

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

REFERENCES

  • 1.Cassell GH, Waites KB, Watson HL, Crouse DT, Harasawa R. Ureaplasma urealyticum intrauterine infection: role in prematurity and disease in newborns. Clin Microbiol Rev. 1993;6(1):69–87. doi: 10.1128/cmr.6.1.69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Knox CL, Allan JA, Allan JM, et al. Ureaplasma parvum and Ureaplasma urealyticum are detected in semen after washing before assisted reproductive technology procedures. Fertil Steril. 2003;80(4):921–929. doi: 10.1016/s0015-0282(03)01125-7. [DOI] [PubMed] [Google Scholar]
  • 3.Viscardi RM. Ureaplasma species: role in diseases of prematurity. Clin Perinatol. 2010;37(2):393–409. doi: 10.1016/j.clp.2009.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Glass JI, Lefkowitz EJ, Glass JS, et al. The complete sequence of the mucosal pathogen Ureaplasma urealyticum. Nature. 2000;407(6805):757–762. doi: 10.1038/35037619. [DOI] [PubMed] [Google Scholar]
  • 5.Ligon JV, Kenny GE. Virulence of ureaplasmal urease for mice. Infect Immun. 1991;59(3):1170–1171. doi: 10.1128/iai.59.3.1170-1171.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Echahidi F, Muyldermans G, Lauwers S, Naessens A. Development of monoclonal antibodies against Ureaplasma urealyticum serotypes and their use for serotyping clinical isolates. Clin Diagn Lab Immunol. 2000;7(4):563–567. doi: 10.1128/cdli.7.4.563-567.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Robertson JA, Stemke GW, Davis JW, Jr, et al. Proposal of Ureaplasma parvum sp. nov. and emended description of Ureaplasma urealyticum (Shepard et al. 1974) Robertson et al 2001. Int J Syst Evol Microbiol. 2002;52(Pt 2):587–597. doi: 10.1099/00207713-52-2-587. [DOI] [PubMed] [Google Scholar]
  • 8.Monecke S, Helbig JH, Jacobs E. Phase variation of the multiple banded protein in Ureaplasma urealyticum and Ureaplasma parvum. Int J Med Microbiol. 2003;293(2–3):203–211. doi: 10.1078/1438-4221-00239. [DOI] [PubMed] [Google Scholar]
  • 9.Zimmerman CU, Rosengarten R, Spergser J. Ureaplasma antigenic variation beyond MBA phase variation: DNA inversions generating chimeric structures and switching in expression of the MBA N-terminal paralogue UU172. Mol Microbiol. 2011;79(3):663–676. doi: 10.1111/j.1365-2958.2010.07474.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Knox CL, Dando SJ, Nitsos I, et al. The severity of chorioamnionitis in pregnant sheep is associated with in vivo variation of the surface-exposed multiple-banded antigen/gene of Ureaplasma parvum. Biol Reprod. 2010;83(3):415–426. doi: 10.1095/biolreprod.109.083121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.De Silva NS, Quinn PA. Localization of endogenous activity of phospholipases A and C in Ureaplasma urealyticum. J Clin Microbiol. 1991;29(7):1498–1503. doi: 10.1128/jcm.29.7.1498-1503.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Kilian M, Brown MB, Brown TA, Freundt EA, Cassell GH. Immunoglobulin A1 protease activity in strains of Ureaplasma urealyticum. Acta Pathol Microbiol Immunol Scand B. 1984;92(1):61–64. doi: 10.1111/j.1699-0463.1984.tb02794.x. [DOI] [PubMed] [Google Scholar]
  • 13.Paralanov V, Lu J, Duffy LB, et al. Comparative genome analysis of 19 Ureaplasma urealyticum and Ureaplasma parvum strains. BMC Microbiol. 2012;12(1):88. doi: 10.1186/1471-2180-12-88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Xiao L, Paralanov V, Glass JI, et al. Extensive horizontal gene transfer in ureaplasmas from humans questions the utility of serotyping for diagnostic purposes. J Clin Microbiol. 2011;49(8):2818–2826. doi: 10.1128/JCM.00637-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Dando SJ, Nitsos I, Kallapur SG, et al. The role of the multiple banded antigen of Ureaplasma parvum in intra-amniotic infection: major virulence factor or decoy? PLoS One. 2012;7(1):e29856. doi: 10.1371/journal.pone.0029856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Pandelidis K, McCarthy A, Chesko KL, Viscardi RM. Role of Biofilm Formation in Ureaplasma Antibiotic Susceptibility and Development of Bronchopulmonary Dysplasia in Preterm Neonates. Pediatr Infect Dis J. 2012 doi: 10.1097/INF.0b013e3182791ae0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.DiGiulio DB. Diversity of microbes in amniotic fluid. Semin Fetal Neonatal Med. 2012;17(1):2–11. doi: 10.1016/j.siny.2011.10.001. [DOI] [PubMed] [Google Scholar]
  • 18.Novy MJ, Duffy L, Axthelm MK, et al. Ureaplasma parvum or Mycoplasma hominis as sole pathogens cause chorioamnionitis, preterm delivery, and fetal pneumonia in rhesus macaques. Reprod Sci. 2009;16(1):56–70. doi: 10.1177/1933719108325508. [DOI] [PubMed] [Google Scholar]
  • 19.Perni SC, Vardhana S, Korneeva I, et al. Mycoplasma hominis and Ureaplasma urealyticum in midtrimester amniotic fluid: association with amniotic fluid cytokine levels and pregnancy outcome. Am J Obstet Gynecol. 2004;191(4):1382–1386. doi: 10.1016/j.ajog.2004.05.070. [DOI] [PubMed] [Google Scholar]
  • 20.Gerber S, Vial Y, Hohlfeld P, Witkin SS. Detection of Ureaplasma urealyticum in second-trimester amniotic fluid by polymerase chain reaction correlates with subsequent preterm labor and delivery. J Infect Dis. 2003;187(3):518–521. doi: 10.1086/368205. [DOI] [PubMed] [Google Scholar]
  • 21.Witt A, Berger A, Gruber CJ, et al. Increased intrauterine frequency of Ureaplasma urealyticum in women with preterm labor and preterm premature rupture of the membranes and subsequent cesarean delivery. Am J Obstet Gynecol. 2005;193(5):1663–1669. doi: 10.1016/j.ajog.2005.03.067. [DOI] [PubMed] [Google Scholar]
  • 22.Namba F, Hasegawa T, Nakayama M, et al. Placental features of chorioamnionitis colonized with Ureaplasma species in preterm delivery. Pediatr Res. 2010;67(2):166–172. doi: 10.1203/PDR.0b013e3181c6e58e. [DOI] [PubMed] [Google Scholar]
  • 23.Kim MJ, Romero R, Gervasi MT, et al. Widespread microbial invasion of the chorioamniotic membranes is a consequence and not a cause of intra-amniotic infection. Lab Invest. 2009;89(8):924–936. doi: 10.1038/labinvest.2009.49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Kumar H, Kawai T, Akira S. Pathogen recognition by the innate immune system. Int Rev Immunol. 2011;30(1):16–34. doi: 10.3109/08830185.2010.529976. [DOI] [PubMed] [Google Scholar]
  • 25.Estrada-Gutierrez G, Gomez-Lopez N, Zaga-Clavellina V, et al. Interaction between pathogenic bacteria and intrauterine leukocytes triggers alternative molecular signaling cascades leading to labor in women. Infect Immun. 2010;78(11):4792–4799. doi: 10.1128/IAI.00522-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Manimtim WM, Hasday JD, Hester L, et al. Ureaplasma urealyticum modulates endotoxin-induced cytokine release by human monocytes derived from preterm and term newborns and adults. Infect Immun. 2001;69(6):3906–3915. doi: 10.1128/IAI.69.6.3906-3915.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Shimizu T, Kida Y, Kuwano K. Ureaplasma parvum lipoproteins, including MB antigen, activate NF-{kappa}B through TLR1, TLR2 and TLR6. Microbiology. 2008;154(Pt 5):1318–1325. doi: 10.1099/mic.0.2007/016212-0. [DOI] [PubMed] [Google Scholar]
  • 28.Peltier MR, Freeman AJ, Mu HH, Cole BC. Characterization of the macrophage-stimulating activity from Ureaplasma urealyticum. Am J Reprod Immunol. 2007;57(3):186–192. doi: 10.1111/j.1600-0897.2006.00460.x. [DOI] [PubMed] [Google Scholar]
  • 29.Quinn PA. Evidence of an immune response to Ureaplasma urealyticum in perinatal morbidity and mortality. Pediatr Infect Dis. 1986;5(6 Suppl):S282–S287. doi: 10.1097/00006454-198611010-00018. [DOI] [PubMed] [Google Scholar]
  • 30.Korfhagen TR, LeVine AM, Whitsett JA. Surfactant protein A (SP-A) gene targeted mice. Biochim Biophys Acta. 1998;1408(2–3):296–302. doi: 10.1016/s0925-4439(98)00075-1. [DOI] [PubMed] [Google Scholar]
  • 31.Famuyide ME, Hasday JD, Carter HC, et al. Surfactant protein-A limits Ureaplasma-mediated lung inflammation in a murine pneumonia model. Pediatr Res. 2009;66(2):162–167. doi: 10.1203/PDR.0b013e3181aabd66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Monneret G, Venet F, Pachot A, Lepape A. Monitoring immune dysfunctions in the septic patient: a new skin for the old ceremony. Mol Med. 2008;14(1–2):64–78. doi: 10.2119/2007-00102.Monneret. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Lehman JS, Jr, Bassily S. Endotoxin tolerance in patients with chronic bacteremia and bacteriuria due to Salmonella. J Infect Dis. 1971;124(3):318–321. doi: 10.1093/infdis/124.3.318. [DOI] [PubMed] [Google Scholar]
  • 34.Kallapur SG, Jobe AH, Ball MK, et al. Pulmonary and systemic endotoxin tolerance in preterm fetal sheep exposed to chorioamnionitis. J Immunol. 2007;179(12):8491–8499. doi: 10.4049/jimmunol.179.12.8491. [DOI] [PubMed] [Google Scholar]
  • 35.Kramer BW, Kallapur SG, Moss TJ, et al. Intra-amniotic LPS modulation of TLR signaling in lung and blood monocytes of fetal sheep. Innate Immun. 2009;15(2):101–107. doi: 10.1177/1753425908100455. [DOI] [PubMed] [Google Scholar]
  • 36.Kramer BW, Ikegami M, Moss TJ, et al. Endotoxin-induced chorioamnionitis modulates innate immunity of monocytes in preterm sheep. Am J Respir Crit Care Med. 2005;171(1):73–77. doi: 10.1164/rccm.200406-745OC. [DOI] [PubMed] [Google Scholar]
  • 37.Kallapur SG, Kramer BW, Knox CL, et al. Chronic Fetal Exposure to Ureaplasma parvum Suppresses Innate Immune Responses in Sheep. J Immunol. 2011;187(5):2688–2695. doi: 10.4049/jimmunol.1100779. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Collins JJ, Kallapur SG, Knox CL, et al. Inflammation in fetal sheep from intra-amniotic injection of Ureaplasma parvum. Am J Physiol Lung Cell Mol Physiol. 2010;299(6):L852–L860. doi: 10.1152/ajplung.00183.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Moss TJ, Knox CL, Kallapur SG, et al. Experimental amniotic fluid infection in sheep: effects of Ureaplasma parvum serovars 3 and 6 on preterm or term fetal sheep. Am J Obstet Gynecol. 2008;198(1):122. doi: 10.1016/j.ajog.2007.06.065. e121-128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Moss TJ, Nitsos I, Knox CL, et al. Ureaplasma colonization of amniotic fluid and efficacy of antenatal corticosteroids for preterm lung maturation in sheep. Am J Obstet Gynecol. 2009;200(1):96. doi: 10.1016/j.ajog.2008.08.044. e91-96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Moss TJ, Nitsos I, Ikegami M, Jobe AH, Newnham JP. Intrauterine Ureaplasma infection accelerates fetal lung maturation and causes growth restriction in sheep. Am J Obstet Gynecol. 2005;192:1179–1186. doi: 10.1016/j.ajog.2004.11.063. [DOI] [PubMed] [Google Scholar]
  • 42.Husain AN. Pathology of arrested acinar development in postsurfactant bronchopulmonary dysplasia. Human Pathology. 1998;29(7):710–717. doi: 10.1016/s0046-8177(98)90280-5. [DOI] [PubMed] [Google Scholar]
  • 43.Polglase GR, Dalton RG, Nitsos I, et al. Pulmonary vascular and alveolar development in preterm lambs chronically colonized with Ureaplasma parvum. Am J Physiol Lung Cell Mol Physiol. 2010;299(2):L232–L241. doi: 10.1152/ajplung.00369.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Yoder BA, Coalson JJ, Winter VT, et al. Effects of antenatal colonization with ureaplasma urealyticum on pulmonary disease in the immature baboon. Pediatr Res. 2003;54(6):797–807. doi: 10.1203/01.PDR.0000091284.84322.16. [DOI] [PubMed] [Google Scholar]
  • 45.Viscardi R, Manimtim W, He JR, et al. Disordered pulmonary myofibroblast distribution and elastin expression in preterm infants with Ureaplasma urealyticum pneumonitis. Pediatr Dev Pathol. 2006;9(2):143–151. doi: 10.2350/10-05-0112.1. [DOI] [PubMed] [Google Scholar]
  • 46.Viscardi RM, Manimtim WM, Sun CC, Duffy L, Cassell GH. Lung pathology in premature infants with Ureaplasma urealyticum infection. Pediatr Dev Pathol. 2002;5(2):141–150. doi: 10.1007/s10024001-0134-y. [DOI] [PubMed] [Google Scholar]
  • 47.Laughon MM, Langer JC, Bose CL, et al. Prediction of Bronchopulmonary Dysplasia by Postnatal Age in Extremely Premature Infants. Am J Respir Crit Care Med. 2011;183(12):1715–1722. doi: 10.1164/rccm.201101-0055OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Laughon M, Allred EN, Bose C, et al. Patterns of respiratory disease during the first 2 postnatal weeks in extremely premature infants. Pediatrics. 2009;123(4):1124–1131. doi: 10.1542/peds.2008-0862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Charafeddine L, D'Angio CT, Phelps DL. Atypical chronic lung disease patterns in neonates. Pediatrics. 1999;103(4 Pt 1):759–765. doi: 10.1542/peds.103.4.759. [DOI] [PubMed] [Google Scholar]
  • 50.Kallapur SG, Jobe AH. Antenatal factors that influence postnatal lung development and injury. In: Polin RA, Fox WW, Abman SH, editors. Fetal and Neonatal physiology. 4th ed. Philadelphia, PA: W.B.: Saunders and Co.; 2011. pp. 1047–1054. [Google Scholar]
  • 51.Been JV, Rours IG, Kornelisse RF, et al. Chorioamnionitis alters the response to surfactant in preterm infants. J Pediatr. 2010;156(1):10–15. doi: 10.1016/j.jpeds.2009.07.044. e11. [DOI] [PubMed] [Google Scholar]
  • 52.Bose C, Van Marter LJ, Laughon M, et al. Fetal growth restriction and chronic lung disease among infants born before the 28th week of gestation. Pediatrics. 2009;124(3):e450–e458. doi: 10.1542/peds.2008-3249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Witt A, Berger A, Gruber CJ, et al. IL-8 concentrations in maternal serum, amniotic fluid and cord blood in relation to different pathogens within the amniotic cavity. J Perinat Med. 2005;33(1):22–26. doi: 10.1515/JPM.2005.003. [DOI] [PubMed] [Google Scholar]
  • 54.Garland SM, Bowman ED. Role of Ureaplasma urealyticum and Chlamydia trachomatis in lung disease in low birth weight infants. Pathology. 1996;28(3):266–269. doi: 10.1080/00313029600169134. [DOI] [PubMed] [Google Scholar]
  • 55.Pacifico L, Panero A, Roggini M, et al. Ureaplasma urealyticum and pulmonary outcome in a neonatal intensive care population. Pediatr Infect Dis J. 1997;16(6):579–586. doi: 10.1097/00006454-199706000-00008. [DOI] [PubMed] [Google Scholar]
  • 56.Perzigian RW, Adams JT, Weiner GM, et al. Ureaplasma urealyticum and chronic lung disease in very low birth weight infants during the exogenous surfactant era. Pediatr Infect Dis J. 1998;17(7):620–625. doi: 10.1097/00006454-199807000-00009. [DOI] [PubMed] [Google Scholar]
  • 57.Abele-Horn M, Genzel-Boroviczeny O, Uhlig T, et al. Ureaplasma urealyticum colonization and bronchopulmonary dysplasia: a comparative prospective multicentre study. Eur J Pediatr. 1998;157(12):1004–1011. doi: 10.1007/s004310050987. [DOI] [PubMed] [Google Scholar]
  • 58.Hannaford K, Todd DA, Jeffrey H, et al. Role of ureaplasma urealyticum in lung disease of prematurity. Arch Dis Child Fetal Neonatal Ed. 1999;81:F162–F167. doi: 10.1136/fn.81.3.f162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Da Silva O, Gregson D, Hammerberg O. Role of Ureaplasma urealyticum and Chlamydia trachomatis in development of bronchopulmonary dysplasia in very low birth weight infants. Pediatr Infect Dis J. 1997;16(4):364–369. doi: 10.1097/00006454-199704000-00006. [DOI] [PubMed] [Google Scholar]
  • 60.van Waarde WM, Brus F, Okken A, Kimpen JL. Ureaplasma urealyticum colonization, prematurity and bronchopulmonary dysplasia. Eur Respir J. 1997;10(4):886–890. [PubMed] [Google Scholar]
  • 61.Couroucli XI, Welty SE, Ramsay PL, et al. Detection of microorganisms in the tracheal aspirates of preterm infants by polymerase chain reaction: association of adenovirus infection with bronchopulmonary dysplasia. Pediatr Res. 2000;47(2):225–232. doi: 10.1203/00006450-200002000-00013. [DOI] [PubMed] [Google Scholar]
  • 62.Heggie AD, Bar-Shain D, Boxerbaum B, et al. Identification and quantification of ureaplasmas colonizing the respiratory tract and assessment of their role in the development of chronic lung disease in preterm infants. Pediatr Infect Dis J. 2001;20(9):854–859. doi: 10.1097/00006454-200109000-00006. [DOI] [PubMed] [Google Scholar]
  • 63.Watterberg K, Demers L, Scott S, Murphy S. Chorioamnionitis and early lung inflammation in infants in whom bronchopulmonary dysplasia develops. Pediatrics. 1996 Feb;97:210–215. [PubMed] [Google Scholar]
  • 64.Viscardi RM, Muhumuza CK, Rodriguez A, et al. Inflammatory markers in intrauterine and fetal blood and cerebrospinal fluid compartments are associated with adverse pulmonary and neurologic outcomes in preterm infants. Pediatr Res. 2004;55(6):1009–1017. doi: 10.1203/01.pdr.0000127015.60185.8a. [DOI] [PubMed] [Google Scholar]
  • 65.Van Marter LJ, Dammann O, Allred EN, et al. Chorioamnionitis, mechanical ventilation, and postnatal sepsis as modulators of chronic lung disease in preterm infants. J Pediatr. 2002;140(2):171–176. doi: 10.1067/mpd.2002.121381. [DOI] [PubMed] [Google Scholar]
  • 66.Andrews WW, Goldenberg RL, Faye-Petersen O, et al. The Alabama Preterm Birth study: polymorphonuclear and mononuclear cell placental infiltrations, other markers of inflammation, and outcomes in 23- to 32- week preterm newborn infants. Am J Obstet Gynecol. 2006;195(3):803–808. doi: 10.1016/j.ajog.2006.06.083. [DOI] [PubMed] [Google Scholar]
  • 67.Lahra MM, Beeby PJ, Jeffery HE. Intrauterine inflammation, neonatal sepsis, and chronic lung disease: a 13-year hospital cohort study. Pediatrics. 2009;123(5):1314–1319. doi: 10.1542/peds.2008-0656. [DOI] [PubMed] [Google Scholar]
  • 68.Schelonka RL, Katz B, Waites KB, Benjamin DK., Jr Critical appraisal of the role of Ureaplasma in the development of bronchopulmonary dysplasia with metaanalytic techniques. Pediatr Infect Dis J. 2005;24(12):1033–1039. doi: 10.1097/01.inf.0000190632.31565.83. [DOI] [PubMed] [Google Scholar]
  • 69.Morley CJ, Davis PG, Doyle LW, et al. Nasal CPAP or intubation at birth for very preterm infants. N Engl J Med. 2008;358(7):700–708. doi: 10.1056/NEJMoa072788. [DOI] [PubMed] [Google Scholar]
  • 70.Finer NN, Carlo WA, Walsh MC, et al. Early CPAP versus surfactant in extremely preterm infants. N Engl J Med. 2010;362(21):1970–1979. doi: 10.1056/NEJMoa0911783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Dunn MS, Kaempf J, de Klerk A, et al. Randomized trial comparing 3 approaches to the initial respiratory management of preterm neonates. Pediatrics. 2011;128(5):e1069–e1076. doi: 10.1542/peds.2010-3848. [DOI] [PubMed] [Google Scholar]
  • 72.Carlo WA. Gentle ventilation: the new evidence from the SUPPORT, COIN, VON, CURPAP, Colombian Network, and Neocosur Network trials. Early Hum Dev. 2012;88(Suppl 2):S81–S83. doi: 10.1016/S0378-3782(12)70022-1. [DOI] [PubMed] [Google Scholar]
  • 73.Sandri F, Plavka R, Ancora G, et al. Prophylactic or early selective surfactant combined with nCPAP in very preterm infants. Pediatrics. 2010;125(6):e1402–e1409. doi: 10.1542/peds.2009-2131. [DOI] [PubMed] [Google Scholar]
  • 74.Inatomi T, Oue S, Ogihara T, et al. Antenatal exposure to Ureaplasma species exacerbates bronchopulmonary dysplasia synergistically with subsequent prolonged mechanical ventilation in preterm infants. Pediatr Res. 2012;71(3):267–273. doi: 10.1038/pr.2011.47. [DOI] [PubMed] [Google Scholar]
  • 75.Mercer BM, Miodovnik M, Thurnau GR, et al. Antibiotic therapy for reduction of infant morbidity after preterm premature rupture of the membranes. A randomized controlled trial. National Institute of Child Health and Human Development Maternal-Fetal Medicine Units Network. JAMA. 1997;278(12):989–995. [PubMed] [Google Scholar]
  • 76.Kenyon SL, Taylor DJ, Tarnow-Mordi W. Broad-spectrum antibiotics for preterm, prelabour rupture of fetal membranes: the ORACLE I randomised trial. ORACLE Collaborative Group. Lancet. 2001;357(9261):979–988. doi: 10.1016/s0140-6736(00)04233-1. [DOI] [PubMed] [Google Scholar]
  • 77.Kenyon SL, Taylor DJ, Tarnow-Mordi W. Broad-spectrum antibiotics for spontaneous preterm labour: the ORACLE II randomised trial. ORACLE Collaborative Group. Lancet. 2001;357(9261):989–994. doi: 10.1016/s0140-6736(00)04234-3. [DOI] [PubMed] [Google Scholar]
  • 78.Kenyon S, Pike K, Jones DR, et al. Childhood outcomes after prescription of antibiotics to pregnant women with spontaneous preterm labour: 7-year follow-up of the ORACLE II trial. Lancet. 2008;372(9646):1319–1327. doi: 10.1016/S0140-6736(08)61203-9. [DOI] [PubMed] [Google Scholar]
  • 79.ACOG Committee Opinion No. 445: antibiotics for preterm labor. Obstet Gynecol. 2009;114(5):1159–1160. doi: 10.1097/AOG.0b013e3181c33c86. [DOI] [PubMed] [Google Scholar]
  • 80.Turner MA, Jacqz-Aigrain E, Kotecha S. Azithromycin, Ureaplasma and chronic lung disease of prematurity: a case study for neonatal drug development. Arch Dis Child. 2012;97(6):573–577. doi: 10.1136/adc.2010.195180. [DOI] [PubMed] [Google Scholar]
  • 81.Dando SJ, Nitsos I, Newnham JP, et al. Maternal administration of erythromycin fails to eradicate intrauterine ureaplasma infection in an ovine model. Biol Reprod. 2010;83(4):616–622. doi: 10.1095/biolreprod.110.084954. [DOI] [PubMed] [Google Scholar]
  • 82.Grigsby PL, Novy MJ, Sadowsky DW, et al. Maternal Azithromycin therapy for ureaplasma intra-amniotic infection delays preterm delivery and reduces fetal lung injury in a primate model. Am J Obstet Gynecol. 2012 doi: 10.1016/j.ajog.2012.10.871. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Lyon AJ, McColm J, Middlemist L, et al. Randomised trial of erythromycin on the development of chronic lung disease in preterm infants. Arch Dis Child Fetal Neonatal Ed. 1998;78(1):F10–F14. doi: 10.1136/fn.78.1.f10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Jonsson B, Rylander M, Faxelius G. Ureaplasma urealyticum, erythromycin and respiratory morbidity in high-risk preterm neonates. Acta Paediatr. 1998;87(10):1079–1084. doi: 10.1080/080352598750031428. [DOI] [PubMed] [Google Scholar]
  • 85.Ballard HO, Shook LA, Bernard P, et al. Use of azithromycin for the prevention of bronchopulmonary dysplasia in preterm infants: a randomized, double-blind, placebo controlled trial. Pediatr Pulmonol. 2011;46(2):111–118. doi: 10.1002/ppul.21352. [DOI] [PubMed] [Google Scholar]
  • 86.Ozdemir R, Erdeve O, Dizdar EA, et al. Clarithromycin in preventing bronchopulmonary dysplasia in Ureaplasma urealyticum-positive preterm infants. Pediatrics. 2011;128(6):e1496–e1501. doi: 10.1542/peds.2011-1350. [DOI] [PubMed] [Google Scholar]
  • 87.Cotten CM, Taylor S, Stoll B, et al. Prolonged duration of initial empirical antibiotic treatment is associated with increased rates of necrotizing enterocolitis and death for extremely low birth weight infants. Pediatrics. 2009;123(1):58–66. doi: 10.1542/peds.2007-3423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Kuppala VS, Meinzen-Derr J, Morrow AL, Schibler KR. Prolonged initial empirical antibiotic treatment is associated with adverse outcomes in premature infants. J Pediatr. 2011;159(5):720–725. doi: 10.1016/j.jpeds.2011.05.033. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES