1
|
Huang MK, Yang YL, Hui L, Chen XL, Liu T, Jiang YM. Epidemiological characteristics and clinical antibiotic resistance analysis of Ureaplasma urealyticum infection among women and children in southwest China. BMC Infect Dis 2024; 24:849. [PMID: 39169329 PMCID: PMC11340039 DOI: 10.1186/s12879-024-09760-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 08/19/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND The aim of this study was to investigate the epidemiological characteristics and antibiotic resistance patterns of Ureaplasma urealyticum (UU) infection among women and children in southwest China. METHODS A total of 8,934 specimens, including urogenital swabs and throat swabs were analyzed in this study. All samples were tested using RNA-based Simultaneous Amplification and Testing (SAT) methods. Culture and drug susceptibility tests were performed on UU positive patients. RESULTS Among the 8,934 patients, the overall positive rate for UU was 47.92%, with a higher prevalence observed among women of reproductive age and neonates. The majority of UU positive outpatients were women of reproductive age (88.03%), while the majority of UU positive inpatients were neonates (93.99%). Overall, hospitalization rates due to UU infection were significantly higher in neonates than in women. Further analysis among neonatal inpatients revealed a higher incidence of preterm birth and low birth weight in UU positive inpatients (52.75% and 3.65%, respectively) than in UU negative inpatients (44.64% and 2.89%, respectively), especially in very preterm and extremely preterm neonates. Moreover, the incidence rate of bronchopulmonary dysplasia (BPD) among hospitalized neonatal patients was significantly higher in the UU positive group (6.89%) than in the UU negative group (4.18%). The drug susceptibility tests of UU in the neonatology, gynecology and obstetrics departments exhibited consistent sensitivity patterns to antibiotics, with high sensitivity to tetracyclines and macrolides, and low sensitivity to fluoroquinolones. Notably, UU samples collected from the neonatology department exhibited significantly higher sensitivity to azithromycin and erythromycin (93.8% and 92.9%, respectively) than those collected from the gynecology and obstetrics departments. CONCLUSIONS This study enhances our understanding of the current epidemiological characteristics and antibiotic resistance patterns of UU infection among women and children in southwest China. These findings can aid in the development of more effective intervention, prevention and treatment strategies for UU infection.
Collapse
Affiliation(s)
- Meng-Ke Huang
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renmin Road South, Chengdu, Sichuan, 610041, P. R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yun-Long Yang
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renmin Road South, Chengdu, Sichuan, 610041, P. R. China
| | - Lu Hui
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renmin Road South, Chengdu, Sichuan, 610041, P. R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, China
| | - Xiao-Lin Chen
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renmin Road South, Chengdu, Sichuan, 610041, P. R. China
| | - Ting Liu
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renmin Road South, Chengdu, Sichuan, 610041, P. R. China.
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, China.
- State Key Laboratory of Biotherapy and Cancer Center, National Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China.
| | - Yong-Mei Jiang
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renmin Road South, Chengdu, Sichuan, 610041, P. R. China.
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
2
|
Chen X, Huang X, Zhou Q, Kang H, Qiu H, Shi L, Tang H, Zeng S. Association between Ureaplasma urealyticum colonization and bronchopulmonary dysplasia in preterm infants: a systematic review and meta-analysis. Front Pediatr 2024; 12:1436568. [PMID: 39175806 PMCID: PMC11338929 DOI: 10.3389/fped.2024.1436568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 07/29/2024] [Indexed: 08/24/2024] Open
Abstract
Background Bronchopulmonary dysplasia (BPD) is the most prevalent chronic lung disease in preterm infants. Studies have shown that Ureaplasma urealyticum (UU) infection is linked to its pathogenesis. However, it remains controversial whether UU colonization in preterm infants increases the risk of developing BPD. Objective This study aimed to conduct a systematic review and meta-analysis to summarize the correlation between UU and BPD. Methods We searched PubMed, Embase, the Cochrane Library, Web of Science, Wanfang Database, Chinese National Knowledge Infrastructure Database, Chinese Science and Technique Journal Database, and the China Biology Medicine disc from their inception to March 15, 2024. We included cohort and case-control studies investigating the association between UU infections and BPD in preterm infants, adhering to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. The Newcastle-Ottawa Scale was used for quality assessment. The outcome was defined as the continued need for oxygen or respiratory support at 28 days after birth (BPD28) or at 36 weeks postmenstrual age (BPD36). Considering the potential publication bias in observational studies, we used a random-effects meta-analysis model, assessed heterogeneity (I2), performed subgroup analyses, evaluated publication bias, and graded the quality of evidence. Results The meta-analysis included 36 cohort studies encompassing 5,991 participants. Among these, 20 reported on BPD28, 13 on BPD36, and 3 on both. The results indicated a significant association between UU colonization and BPD28 (odds ratio (OR): 2.26, 95% confidence interval (CI): 1.78-2.85, P < 0.00001, 23 studies, very low certainty of evidence) and BPD36 (OR: 2.13, 95% CI: 1.47-3.07, P < 0.0001, 16 studies, very low certainty of evidence). Conclusion There is a correlation between UU colonization and the development of BPD in preterm infants. Future research should prioritize well-designed, large-scale, high-quality randomized controlled trials (RCTs) to comprehensively assess the risk of BPD in neonates following UU infection and to provide stronger evidence for clinical screening and prevention strategies to improve the prognosis of affected newborns. Systematic Review Registration https://www.crd.york.ac.uk/, identifier (CRD42024524846).
Collapse
Affiliation(s)
- Xianhong Chen
- Shenzhen Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Xunbin Huang
- Division of Neonatology, Longgang District Central Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Qiujing Zhou
- Division of Neonatology, Longgang District Central Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Houxin Kang
- Neonatal·Child Critical Child Health Care Division, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi Tujia and Miao Autonomous Prefecture, Hubei, China
| | - Huixian Qiu
- Division of Neonatology, Longgang District Central Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Lindong Shi
- Division of Neonatology, Longgang District Central Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Hong Tang
- Division of Neonatology, Shenzhen Yantian District People’s Hospital, Shenzhen, Guangdong, China
| | - Shujuan Zeng
- Division of Neonatology, Longgang District Central Hospital of Shenzhen, Shenzhen, Guangdong, China
| |
Collapse
|
3
|
Silwedel C, Laube M, Speer CP, Glaser K. The Role of Ureaplasma Species in Prenatal and Postnatal Morbidity of Preterm Infants: Current Concepts. Neonatology 2024; 121:627-635. [PMID: 38934167 DOI: 10.1159/000539613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Ureaplasma species are considered commensals of the adult urogenital tract. Yet, in pregnancy, Ureaplasma parvum and Ureaplasma urealyticum have been associated with chorioamnionitis and preterm birth. In preterm infants, Ureaplasma respiratory tract colonization has been correlated with the development of bronchopulmonary dysplasia and has been implicated in the pathogenesis of other complications of prematurity. Controversies on the impact of Ureaplasma exposure on neonatal morbidity, however, remain, and recommendations for screening practices and therapeutic management in preterm infants are missing. SUMMARY In this review, we outline clinical and experimental evidence of Ureaplasma-driven fetal and neonatal morbidity, critically examining inconsistencies across some of the existing studies. We explore underlying mechanisms of Ureaplasma-associated neonatal morbidity and discuss gaps in the current understanding including the interplay between Ureaplasma and the maternal urogenital tract and the preterm airway microbiome. Ultimately, we highlight the importance of adequate diagnostics and review the potential efficacy of anti-infective therapies. KEY MESSAGES There is strong evidence that perinatal Ureaplasma exposure is causally related to the development of bronchopulmonary dysplasia, and there are conclusive data of the role of Ureaplasma in the pathogenesis of neonatal central nervous system infection. Observational and experimental findings indicate immunomodulatory capacities that might promote an increased risk of secondary infections. The burden of Ureaplasma exposure is inversely related to gestational age - leaving the tiniest babies at highest risk. A better knowledge of contributing pathogen and host factors and modulating conditions remains paramount to define screening and treatment recommendations allowing early intervention in preterm infants at risk.
Collapse
Affiliation(s)
- Christine Silwedel
- Department of Pediatrics, University Hospital of Würzburg, Würzburg, Germany
| | - Mandy Laube
- Center for Pediatric Research Leipzig, Division of Neonatology, Department of Pediatrics, University of Leipzig, Leipzig, Germany
| | - Christian P Speer
- Department of Pediatrics, University Hospital of Würzburg, Würzburg, Germany
| | - Kirsten Glaser
- Center for Pediatric Research Leipzig, Division of Neonatology, Department of Pediatrics, University of Leipzig, Leipzig, Germany
- Division of Neonatology, Department of Women's and Children's Health, University of Leipzig Medical Center, Leipzig, Germany
| |
Collapse
|
4
|
Ma J, Wang Z, Luo C, Xi J, Wang X, Hu Y, Zhu C, Jin Z. Epidemiological and Clinical Characteristics of Neonatal Ureaplasma urealyticum Infection. Infect Drug Resist 2024; 17:1323-1332. [PMID: 38585416 PMCID: PMC10995099 DOI: 10.2147/idr.s452014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 03/23/2024] [Indexed: 04/09/2024] Open
Abstract
Purpose To understand the epidemiology and clinical features of Ureaplasma urealyticum (UU) infection in hospitalized neonates due to vertical transmission from mother to child. Methods Respiratory secretions were collected from neonates hospitalized in the neonatology department of the Maternal and Child Health Hospital of Hubei Province from July 2020 to June 2022, and PCR was used to detect UU-DNA in respiratory secretions. The neonates were divided into UU-positive and UU-negative groups, the epidemiological and clinical characteristics of two groups, were statistically analyzed. Results A total of 7257 hospitalized neonates were included in this study, of whom 561 were UU positive and 6696 were UU negative, with a UU detection rate of 7.73%. The detection rate among female neonates was higher than male neonates, and the highest detection rate was found in the period from 1-7 days after birth; the detection rate was highest in spring and fall, and the lowest in winter, but the overall difference was not statistically significant (P>0.05). Compared with the UU-negative group, neonates in the UU-positive group were more likely to be preterm, have a lower birth weight, be delivered vaginally, and have maternal preterm rupture of membranes. In addition, neonates in the UU-positive group were more likely to be co-infected with pathogens and to have complications related to UU infections, which were all statistically significant (P<0.05). Conclusion Neonatal UU infections are detected more frequently in female infants, with the highest detection rate occurring in 1-7 days after birth, and the most prevalent periods for infection being spring and fall. Vaginal delivery and premature rupture of membranes may lead to an increased risk of vertical UU transmission from mother to child, and UU infection is strongly associated with preterm labor, low birth weight, pathogen co-infection, and related complications.
Collapse
Affiliation(s)
- Jun’e Ma
- Department of Clinical Laboratory, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430070, People’s Republic of China
| | - Zhenhui Wang
- Department of Clinical Laboratory, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430070, People’s Republic of China
| | - Chuanjin Luo
- State Key Laboratory of Virology College of Life Sciences, Wuhan University, Wuhan, 430060, People’s Republic of China
| | - Jin’ou Xi
- Department of Clinical Laboratory, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430070, People’s Republic of China
| | - Xiaojing Wang
- Department of Clinical Laboratory, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430070, People’s Republic of China
| | - Yan Hu
- Department of Clinical Laboratory, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430070, People’s Republic of China
| | - Chengliang Zhu
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, People’s Republic of China
| | - Zhengjiang Jin
- Department of Clinical Laboratory, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430070, People’s Republic of China
| |
Collapse
|
5
|
Viscardi RM, Magder LS, Terrin ML, Davis NL. Predicting the likelihood of lower respiratory tract Ureaplasma infection in preterms. Arch Dis Child Fetal Neonatal Ed 2023; 108:250-255. [PMID: 36261143 PMCID: PMC10113605 DOI: 10.1136/archdischild-2022-324192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 10/06/2022] [Indexed: 11/04/2022]
Abstract
OBJECTIVE To develop predictive models of Ureaplasma spp lower airway tract infection in preterm infants. METHODS A dataset was assembled from five cohorts of infants born <33 weeks gestational age (GA) enrolled over 17 years (1999-2016) with culture and/or PCR-confirmed tracheal aspirate Ureaplasma status in the first week of life (n=415). Seventeen demographic, obstetric and neonatal factors were analysed including admission white blood cell (WBC) counts. Best subset regression was used to develop three risk scores for lower airway Ureaplasma infection: (1) including admission laboratory values, (2) excluding admission laboratory values and (3) using only data known prenatally. RESULTS GA and rupture of membranes >72 hours were significant predictors in all 3 models. When all variables including admission laboratory values were included in the regression, WBC count was also predictive in the resulting model. When laboratory values were excluded, delivery route was found to be an additional predictive factor. The area under the curve for the receiver operating characteristic indicated high predictive ability of each model to identify infants with lower airway Ureaplasma infection (range 0.73-0.77). CONCLUSION We developed predictive models based on clinical and limited laboratory information available in the perinatal period that can distinguish between low risk (<10%) and high risk (>40%) of lower airway Ureaplasma infection. These may be useful in the design of phase III trials of therapeutic interventions to prevent Ureaplasma-mediated lung disease in preterm infants and in clinical management of at-risk infants.
Collapse
Affiliation(s)
- Rose Marie Viscardi
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Laurence S Magder
- Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Michael L Terrin
- Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Natalie L Davis
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
6
|
Van Mechelen K, van Westering-Kroon E, Hütten M, Mahieu L, Villamor E. Placing Ureaplasma within the Context of Bronchopulmonary Dysplasia Endotypes and Phenotypes. CHILDREN (BASEL, SWITZERLAND) 2023; 10:children10020256. [PMID: 36832386 PMCID: PMC9955705 DOI: 10.3390/children10020256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/17/2023] [Accepted: 01/29/2023] [Indexed: 02/03/2023]
Abstract
Different pathophysiological pathways (endotypes), leading to very preterm birth may result in distinct clinical phenotypes of bronchopulmonary dysplasia (BPD). Ureaplasma is a unique player in the pathogenesis of BPD. The interaction between factors inherent to Ureaplasma (virulence, bacterial load, duration of exposure), and to the host (immune response, infection clearance, degree of prematurity, respiratory support, concomitant infections) may contribute to BPD development in a variable manner. The data reviewed herein support the hypothesis that Ureaplasma, as a representative of the infectious/inflammatory endotype, may produce pulmonary damage predominantly in parenchyma, interstitium, and small airways. In contrast, Ureaplasma may have a very limited role in the pathogenesis of the vascular phenotype of BPD. In addition, if Ureaplasma is a key factor in BPD pathogenesis, its eradication by macrolides should prevent BPD. However, various meta-analyses do not show consistent evidence that this is the case. The limitations of current definitions and classifications of BPD, based on respiratory support needs instead of pathophysiology and phenotypes, may explain this and other failures in strategies aimed to prevent BPD. The precise mechanisms through which Ureaplasma infection leads to altered lung development and how these pathways can result in different BPD phenotypes warrant further investigation.
Collapse
Affiliation(s)
- Karen Van Mechelen
- Department of Pediatrics, Maastricht University Medical Center, School for Oncology and Reproduction (GROW), Maastricht University, 6229 HX Maastricht, The Netherlands
| | - Elke van Westering-Kroon
- Department of Pediatrics, Maastricht University Medical Center, School for Oncology and Reproduction (GROW), Maastricht University, 6229 HX Maastricht, The Netherlands
| | - Matthias Hütten
- Department of Pediatrics, Maastricht University Medical Center, School for Oncology and Reproduction (GROW), Maastricht University, 6229 HX Maastricht, The Netherlands
| | - Ludo Mahieu
- Department of Neonatology, Antwerp University Hospital, 2650 Edegem, Belgium
| | - Eduardo Villamor
- Department of Pediatrics, Maastricht University Medical Center, School for Oncology and Reproduction (GROW), Maastricht University, 6229 HX Maastricht, The Netherlands
- Correspondence:
| |
Collapse
|
7
|
Ureaplasma-Driven Neonatal Neuroinflammation: Novel Insights from an Ovine Model. Cell Mol Neurobiol 2023; 43:785-795. [PMID: 35334011 PMCID: PMC9957905 DOI: 10.1007/s10571-022-01213-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 03/14/2022] [Indexed: 11/03/2022]
Abstract
Ureaplasma species (spp.) are considered commensals of the adult genitourinary tract, but have been associated with chorioamnionitis, preterm birth, and invasive infections in neonates, including meningitis. Data on mechanisms involved in Ureaplasma-driven neuroinflammation are scarce. The present study addressed brain inflammatory responses in preterm lambs exposed to Ureaplasma parvum (UP) in utero. 7 days after intra-amniotic injection of UP (n = 10) or saline (n = 11), lambs were surgically delivered at gestational day 128-129. Expression of inflammatory markers was assessed in different brain regions using qRT-PCR and in cerebrospinal fluid (CSF) by multiplex immunoassay. CSF was analyzed for UP presence using ureB-based real-time PCR, and MRI scans documented cerebral white matter area and cortical folding. Cerebral tissue levels of atypical chemokine receptor (ACKR) 3, caspases 1-like, 2, 7, and C-X-C chemokine receptor (CXCR) 4 mRNA, as well as CSF interleukin-8 protein concentrations were significantly increased in UP-exposed lambs. UP presence in CSF was confirmed in one animal. Cortical folding and white matter area did not differ among groups. The present study confirms a role of caspases and the transmembrane receptors ACKR3 and CXCR4 in Ureaplasma-driven neuroinflammation. Enhanced caspase 1-like, 2, and 7 expression may reflect cell death. Increased ACKR3 and CXCR4 expression has been associated with inflammatory central nervous system (CNS) diseases and impaired blood-brain barrier function. According to these data and previous in vitro findings from our group, we speculate that Ureaplasma-induced caspase and receptor responses affect CNS barrier properties and thus facilitate neuroinflammation.
Collapse
|
8
|
In Vitro
Activity of Delafloxacin and Finafloxacin against Mycoplasma hominis and
Ureaplasma
Species. Microbiol Spectr 2022; 10:e0009922. [PMID: 35532225 PMCID: PMC9241805 DOI: 10.1128/spectrum.00099-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The in vitro activity of two new fluoroquinolones, delafloxacin and finafloxacin, were evaluated against M. hominis and Ureaplasma spp. The MICs of delafloxacin, finafloxacin, and two classical fluoroquinolones (moxifloxacin and levofloxacin) were tested against 29 M. hominis and 67 Ureaplasma spp. isolates using the broth microdilution method. The molecular mechanisms underlying fluoroquinolone resistance were also investigated. Delafloxacin exhibited low MICs against M. hominis and Ureaplasma spp., including the levofloxacin-resistant isolates. For M. hominis, delafloxacin showed low MIC90 value of 1 μg/mL (MIC range, <0.031 -1 μg/mL) compared to 8 μg/mL for finafloxacin, 16 μg/mL for moxifloxacin, and 32 μg/mL for levofloxacin. For U. parvum and U. urealyticum, delafloxacin had low MIC90 values (U. parvum, 2 μg/mL; U. urealyticum, 4 μg/mL) compared to 16 -32 μg/mL for finafloxacin, 16 μg/mL for moxifloxacin, and 32 - >32 μg/mL for levofloxacin. The two mutations GyrA S153L and ParC S91I were commonly identified in fluoroquinolone-resistant M. hominis, and ParC S83L was the most frequent mutation identified in fluoroquinolone-resistant Ureaplasma spp. Delafloxacin displayed lower MICs against fluoroquinolone-resistant isolates of both M. hominis and Ureaplasma spp. that have mutations in the quinolone resistance determining regions (QRDRs) than the two classical fluoroquinolones. Delafloxacin is a promising fluoroquinolone with low MICs against fluoroquinolone-resistant M. hominis and Ureaplasma spp. Our study confirms the potential clinical use of delafloxacin in treating antimicrobial-resistant M. hominis and Ureaplasma spp. infections. IMPORTANCE Fluoroquinolone resistance in Mycoplasma hominis and Ureaplasma spp. is on the rise globally, which has compromised the efficacy of the currently available antimicrobial agents. This study evaluated the antimicrobial activity of two new fluoroquinolones, delafloxacin and finafloxacin, for the first time, against M. hominis and Ureaplasma spp. clinical isolates. Delafloxacin and finafloxacin displayed different antimicrobial susceptibility profiles against M. hominis and Ureaplasma spp. in vitro. Delafloxacin was found to be more effective against M. hominis and Ureaplasma spp. than three classical fluoroquinolones (finafloxacin, moxifloxacin, and levofloxacin). Finafloxacin displayed activity similar to moxifloxacin but superior to levofloxacin against M. hominis and Ureaplasma spp. Our findings demonstrate that delafloxacin is a promising fluoroquinolone with outstanding activity against fluoroquinolone-resistant M. hominis and Ureaplasma spp.
Collapse
|
9
|
Gallini F, De Rose DU, Coppola M, Pelosi MS, Cota F, Bottoni A, Ricci D, Romeo DM, Spanu T, Maggio L, Mercuri E, Vento G. Maternal Ureaplasma/Mycoplasma colonization during pregnancy and neurodevelopmental outcomes for preterm infants. Front Pediatr 2022; 10:893812. [PMID: 36046478 PMCID: PMC9420904 DOI: 10.3389/fped.2022.893812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 07/19/2022] [Indexed: 11/25/2022] Open
Abstract
INTRODUCTION Ureaplasma (U.) and Mycoplasma (M.) species have been related to pregnancy complications (including preterm birth) and worse neonatal outcomes. The aim of our work is to evaluate neurodevelopmental outcomes in preterm infants born to mothers with Ureaplasma/Mycoplasma colonization during pregnancy. METHODS Preterm infants with gestational age (GA) of ≤ 30 weeks were included in a retrospective follow-up study. To evaluate the effects of maternal vaginal colonization, we divided preterm infants into two groups: exposed and unexposed infants. All infants were assessed at 24 ± 3 months of age using Griffith's Mental Developmental Scales (GMDS). RESULTS Among 254 preterm infants, only 32 infants (12.6%) were exposed to U. /M. colonization during pregnancy. Exposed infants and unexposed ones had a similar Griffith's Developmental Quotient (106 ± 27.2 vs. 108.9 ± 19.5, respectively), without significant differences (p = 0.46). However, exposed infants had a significantly poorer outcome than their unexposed peers in terms of locomotor abilities (100.7 ± 28.3 exposed vs. 111.5 ± 26.1 unexposed, p = 0.03). CONCLUSION For visual and hearing impairment, exposed and unexposed infants had similar incidences of cognitive and motor impairment. However, exposed infants had significantly lower locomotor scores than unexposed peers.
Collapse
Affiliation(s)
- Francesca Gallini
- Neonatology Unit, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario "Agostino Gemelli" Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy.,Università Cattolica del Sacro Cuore, Milan, Italy
| | - Domenico Umberto De Rose
- Neonatology Unit, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario "Agostino Gemelli" Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy.,Neonatal Intensive Care Unit, Medical and Surgical Department of Fetus-Newborn-Infant, "Bambino Gesù" Children's Hospital Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Maria Coppola
- Neonatology Unit, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario "Agostino Gemelli" Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Maria Sofia Pelosi
- Neonatology Unit, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario "Agostino Gemelli" Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Francesco Cota
- Neonatology Unit, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario "Agostino Gemelli" Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy.,Università Cattolica del Sacro Cuore, Milan, Italy
| | - Anthea Bottoni
- Neonatology Unit, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario "Agostino Gemelli" Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Daniela Ricci
- National Centre of Services and Research for the Prevention of Blindness and Rehabilitation of Low Vision Patients-International Agency for the Prevention of Blindness (IAPB) Italia Onlus, Rome, Italy.,Pediatric Neurology Unit, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario "Agostino Gemelli" Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Domenico Marco Romeo
- Università Cattolica del Sacro Cuore, Milan, Italy.,Pediatric Neurology Unit, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario "Agostino Gemelli" Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Teresa Spanu
- Università Cattolica del Sacro Cuore, Milan, Italy.,Department of Laboratory and Infectious Sciences, Fondazione Policlinico Universitario "Agostino Gemelli" Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Luca Maggio
- Università Cattolica del Sacro Cuore, Milan, Italy.,Neonatology Unit, S. Camillo-Forlanini Hospital, Rome, Italy
| | - Eugenio Mercuri
- Università Cattolica del Sacro Cuore, Milan, Italy.,Pediatric Neurology Unit, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario "Agostino Gemelli" Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Giovanni Vento
- Neonatology Unit, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario "Agostino Gemelli" Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy.,Università Cattolica del Sacro Cuore, Milan, Italy
| |
Collapse
|
10
|
Xing N, Zhao Z, Li Q, Dong Y, Li J, Zhang S. Ureaplasma parvum meningitis following atypical choroid plexus papilloma resection in an adult patient: a case report and literature review. BMC Infect Dis 2021; 21:1276. [PMID: 34930148 PMCID: PMC8690907 DOI: 10.1186/s12879-021-06975-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 12/13/2021] [Indexed: 11/30/2022] Open
Abstract
Background While Ureaplasma parvum has previously been linked to the incidence of chorioamnionitis, abortion, premature birth, and perinatal complications, there have only been rare reports of invasive infections of the central nervous system (CNS) in adults. Owing to its atypical presentation and the fact that it will yield sterile cultures using conventional techniques, diagnosing U. parvum meningitis can be challenging. Case presentation We describe a case of U. parvum meningitis detected in an adult patient following surgical brain tumor ablation. After operation, the patient experienced epilepsy, meningeal irritation, and fever with unconsciousness. Cerebrospinal fluid (CSF) analysis showed leukocytosis (484 * 106 /L), elevated protein levels (1.92 g/L), and decreased glucose concentrations (0.02 mmol/L). Evidence suggested that the patient was suffering from bacterial meningitis. However, no bacterial pathogens in either CSF or blood were detected by routine culture or serology. The symptoms did not improve with empirical antibiotics. Therefore, we performed metagenomic next-generation sequencing (mNGS) to identify the etiology of the meningitis. Ureaplasma parvum was detected by mNGS in CSF samples. To the best of our knowledge, this case is the first reported instance of U. parvum meningitis in an adult patient in Asian. After diagnosis, the patient underwent successful moxifloxacin treatment and recovered without complications. Conclusions As mNGS strategies can enable the simultaneous detection of a diverse array of microbes in a single analysis, they may represent a valuable means of diagnosing the pathogens responsible for CNS infections and other clinical conditions with atypical presentations.
Collapse
Affiliation(s)
- Na Xing
- Department of Endocrinology, The Fourth Hospital of Hebei Medical University, No. 12, Jiankang Road, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Zhenxiang Zhao
- Department of Neurosurgery, The Fourth Hospital of Hebei Medical University, No. 12, Jiankang Road, Shijiazhuang, 050000, Hebei, People's Republic of China.
| | - Qingjing Li
- Clinical Laboratory, The Fourth Hospital of Hebei Medical University, No. 12, Jiankang Road, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Yalan Dong
- Department of Neurosurgery, The Fourth Hospital of Hebei Medical University, No. 12, Jiankang Road, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Jianfeng Li
- Department of Neurosurgery, The Fourth Hospital of Hebei Medical University, No. 12, Jiankang Road, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Shuping Zhang
- Department of Oncology, The Seventh People's Hospital of Hebei Province, No. 389, Jungong Road, Dingzhou, 073000, Hebei, People's Republic of China
| |
Collapse
|
11
|
Spencer NR, Radnaa E, Baljinnyam T, Kechichian T, Tantengco OAG, Bonney E, Kammala AK, Sheller-Miller S, Menon R. Development of a mouse model of ascending infection and preterm birth. PLoS One 2021; 16:e0260370. [PMID: 34855804 PMCID: PMC8638907 DOI: 10.1371/journal.pone.0260370] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 11/08/2021] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Microbial invasion of the intraamniotic cavity and intraamniotic inflammation are factors associated with spontaneous preterm birth. Understanding the route and kinetics of infection, sites of colonization, and mechanisms of host inflammatory response is critical to reducing preterm birth risk. OBJECTIVES This study developed an animal model of ascending infection and preterm birth with live bacteria (E. coli) in pregnant CD-1 mice with the goal of better understanding the process of microbial invasion of the intraamniotic cavity and intraamniotic inflammation. STUDY DESIGN Multiple experiments were conducted in this study. To determine the dose of E. coli required to induce preterm birth, CD-1 mice were injected vaginally with four different doses of E. coli (103, 106, 1010, or 1011 colony forming units [CFU]) in 40 μL of nutrient broth or broth alone (control) on an embryonic day (E)15. Preterm birth (defined as delivery before E18.5) was monitored using live video. E. coli ascent kinetics were measured by staining the E. coli with lipophilic tracer DiD for visualization through intact tissue with an in vivo imaging system (IVIS) after inoculation. The E. coli were also directly visualized in reproductive tissues by staining the bacteria with carboxyfluorescein succinimidyl ester (CFSE) prior to administration and via immunohistochemistry (IHC) by staining tissues with anti-E. coli antibody. Each pup's amniotic fluid was cultured separately to determine the extent of microbial invasion of the intraamniotic cavity at different time points. Intraamniotic inflammation resulting from E. coli invasion was assessed with IHC for inflammatory markers (TLR-4, P-NF-κB) and neutrophil marker (Ly-6G) for chorioamnionitis at 6- and 24-h post-inoculation. RESULTS Vaginally administered E. coli resulted in preterm birth in a dose-dependent manner with higher doses causing earlier births. In ex vivo imaging and IHC detected uterine horns proximal to the cervix had increased E. coli compared to the distal uterine horns. E. coli were detected in the uterus, fetal membranes (FM), and placenta in a time-dependent manner with 6 hr having increased intensity of E. coli positive signals in pups near the cervix and in all pups at 24 hr. Similarly, E. coli grew from the cultures of amniotic fluid collected nearest to the cervix, but not from the more distal samples at 6 hr post-inoculation. At 24 hr, all amniotic fluid cultures regardless of distance from the cervix, were positive for E. coli. TLR-4 and P-NF-κB signals were more intense in the tissues where E. coli was present (placenta, FM and uterus), displaying a similar trend toward increased signal in proximal gestational sacs compared to distal at 6 hr. Ly-6G+ cells, used to confirm chorioamnionitis, were increased at 24 hr compared to 6 hr post-inoculation and control. CONCLUSION We report the development of mouse model of ascending infection and the associated inflammation of preterm birth. Clinically, these models can help to understand mechanisms of infection associated preterm birth, determine targets for intervention, or identify potential biomarkers that can predict a high-risk pregnancy status early in pregnancy.
Collapse
Affiliation(s)
- Nicholas R. Spencer
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| | - Enkhtuya Radnaa
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| | - Tuvshintugs Baljinnyam
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| | - Talar Kechichian
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| | - Ourlad Alzeus G. Tantengco
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
- Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines Manila, Manila, Philippines
| | - Elizabeth Bonney
- Department of Obstetrics and Gynecology, University of Vermont, Burlington, VT, United States of America
| | - Ananth Kumar Kammala
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| | - Samantha Sheller-Miller
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| | - Ramkumar Menon
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| |
Collapse
|
12
|
Ferreira G, Santander A, Savio F, Guirado M, Sobrevia L, Nicolson GL. SARS-CoV-2, Zika viruses and mycoplasma: Structure, pathogenesis and some treatment options in these emerging viral and bacterial infectious diseases. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166264. [PMID: 34481867 PMCID: PMC8413106 DOI: 10.1016/j.bbadis.2021.166264] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 07/22/2021] [Accepted: 08/30/2021] [Indexed: 01/28/2023]
Abstract
The molecular evolution of life on earth along with changing environmental, conditions has rendered mankind susceptible to endemic and pandemic emerging infectious diseases. The effects of certain systemic viral and bacterial infections on morbidity and mortality are considered as examples of recent emerging infections. Here we will focus on three examples of infections that are important in pregnancy and early childhood: SARS-CoV-2 virus, Zika virus, and Mycoplasma species. The basic structural characteristics of these infectious agents will be examined, along with their general pathogenic mechanisms. Coronavirus infections, such as caused by the SARS-CoV-2 virus, likely evolved from zoonotic bat viruses to infect humans and cause a pandemic that has been the biggest challenge for humanity since the Spanish Flu pandemic of the early 20th century. In contrast, Zika Virus infections represent an expanding infectious threat in the context of global climate change. The relationship of these infections to pregnancy, the vertical transmission and neurological sequels make these viruses highly relevant to the topics of this special issue. Finally, mycoplasmal infections have been present before mankind evolved, but they were rarely identified as human pathogens until recently, and they are now recognized as important coinfections that are able to modify the course and prognosis of various infectious diseases and other chronic illnesses. The infectious processes caused by these intracellular microorganisms are examined as well as some general aspects of their pathogeneses, clinical presentations, and diagnoses. We will finally consider examples of treatments that have been used to reduce morbidity and mortality of these infections and discuss briefly the current status of vaccines, in particular, against the SARS-CoV-2 virus. It is important to understand some of the basic features of these emerging infectious diseases and the pathogens involved in order to better appreciate the contributions of this special issue on how infectious diseases can affect human pregnancy, fetuses and neonates.
Collapse
Affiliation(s)
- Gonzalo Ferreira
- Laboratory of Ion Channels, Biological Membranes and Cell Signaling, Department of Biophysics, Faculty of Medicine, Universidad de la República, Montevideo, Uruguay.
| | - Axel Santander
- Laboratory of Ion Channels, Biological Membranes and Cell Signaling, Department of Biophysics, Faculty of Medicine, Universidad de la República, Montevideo, Uruguay
| | - Florencia Savio
- Laboratory of Ion Channels, Biological Membranes and Cell Signaling, Department of Biophysics, Faculty of Medicine, Universidad de la República, Montevideo, Uruguay
| | - Mariana Guirado
- Department of Infectious Diseases, Faculty of Medicine, Universidad de la República, Montevideo, Uruguay
| | - Luis Sobrevia
- Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaeology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Seville E-41012, Spain; Medical School (Faculty of Medicine), São Paulo State University (UNESP), Brazil; University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston QLD 4029, Queensland, Australia; Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen (UMCG), 9713GZ Groningen, the Netherlands
| | - Garth L Nicolson
- Department of Molecular Pathology, The Institute for Molecular Medicine, Huntington Beach, CA, USA
| |
Collapse
|
13
|
Abstract
The recently recognized connection between the gut microbiota and pulmonary disease has been termed the gut-lung axis. However, broader connections link the gut and the lungs and these organ systems are tightly interrelated in both homeostasis and disease. This concept is often ignored in the compartmentalized treatment of pulmonary or gastrointestinal disease. In newborns, the most severe gastrointestinal complication of prematurity, necrotizing enterocolitis, and the most severe pulmonary complication, bronchopulmonary dysplasia, both produce significant systemic morbidity. In this review, we highlight the often neglected pathophysiology of the gut-lung axis contributes to increased risk of bronchopulmonary dysplasia in premature infants with necrotizing enterocolitis.
Collapse
Affiliation(s)
| | - Namasivayam Ambalavanan
- Department of Pediatrics, Division of Neonatology, University of Alabama, Birmingham, United Kingdom.
| |
Collapse
|
14
|
Razak A, Alshehri N. Azithromycin for preventing bronchopulmonary dysplasia in preterm infants: A systematic review and meta-analysis. Pediatr Pulmonol 2021; 56:957-966. [PMID: 33382534 DOI: 10.1002/ppul.25230] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 11/30/2020] [Accepted: 12/11/2020] [Indexed: 12/29/2022]
Abstract
CONTEXT Azithromycin has anti-Ureaplasma and anti-inflammatory properties that might help reduce lung injury in preterm infants. OBJECTIVE To test the efficacy and safety of prophylactic or therapeutic azithromycin in preventing bronchopulmonary dysplasia (BPD) in preterm infants with unknown or proven Ureaplasma status. METHODS We searched PubMed, Web of Science, and Cochrane Library until 13 September 2020. Two authors independently assessed the eligibility, risk of bias, and extracted the data. We performed a random-effects model meta-analysis to yield pooled relative risk (RR) or mean difference (MD) with 95% confidence interval (CI). We used the Cochrane GRADE methodology for summarizing the results. RESULTS We included five randomized clinical trials. The meta-analysis revealed no significant differences in BPD (RR, 0.92; 95% CI, 0.71, 1.19; low-quality evidence), death (RR, 0.75; 95% CI, 0.52, 1.10; low-quality evidence), and BPD or death (RR, 0.90; 95% CI, 0.74, 1.10; low-quality evidence). However, a significantly lower BPD or death (RR, 0.83; 95% CI, 0.70, 0.99) and a trend toward lower BPD (RR, 0.83; 95% CI, 0.66, 1.03) with azithromycin therapy was noted in Ureoplasma positive neonates. No differences in secondary outcomes were noted, except for significantly lower supplemental oxygen days with azithromycin (MD, -6.06; 95% CI, -7.40, -4.72; moderate-quality evidence). The test for subgroup differences between short (<7 days) and long (>7 days) course of azithromycin were nonsignificant for all the outcomes. CONCLUSION Low-quality evidence suggests azithromycin therapy reduces BPD or death in preterm infants with positive Ureoplasma, but not in all preterm infants.
Collapse
Affiliation(s)
- Abdul Razak
- Division of Neonatology, Department of Pediatrics, Princess Nourah Bint Abdulrahman University, King Abdullah bin Abdulaziz University Hospital, Riyadh, Saudi Arabia
| | - Norah Alshehri
- Division of Neonatology, Department of Pediatrics, Princess Nourah Bint Abdulrahman University, King Abdullah bin Abdulaziz University Hospital, Riyadh, Saudi Arabia
| |
Collapse
|
15
|
Rittenschober-Böhm J, Habermüller T, Waldhoer T, Fuiko R, Schulz SM, Pimpel B, Goeral K, Witt A, Berger A, Pichler K. Maternal Vaginal Ureaplasma spp. Colonization in Early Pregnancy Is Associated with Adverse Short- and Long-Term Outcome of Very Preterm Infants. CHILDREN (BASEL, SWITZERLAND) 2021; 8:276. [PMID: 33916723 PMCID: PMC8066242 DOI: 10.3390/children8040276] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/25/2021] [Accepted: 04/01/2021] [Indexed: 11/16/2022]
Abstract
Vaginal colonization with Ureaplasma (U.) spp. has been shown to be associated with adverse pregnancy outcome; however, data on neonatal outcome are scarce. The aim of the study was to investigate whether maternal vaginal colonization with U. spp. in early pregnancy represents a risk factor for adverse short- or long-term outcome of preterm infants. Previously, 4330 pregnant women were enrolled in an observational multicenter study, analyzing the association between vaginal U. spp. colonization and spontaneous preterm birth. U. spp. colonization was diagnosed via PCR analysis from vaginal swabs. For this study, data on short-term outcome were collected from medical records and long-term outcome was examined via Bayley Scales of Infant Development at 24 months adjusted age. Two-hundred-and-thirty-eight children were born <33 weeks gestational age. After exclusion due to asphyxia, malformations, and lost-to-follow-up, data on short-term and long-term outcome were available from 222 and 92 infants, respectively. Results show a significant association between vaginal U. spp. colonization and severe intraventricular hemorrhage (10.4% vs. 2.6%, p = 0.03), retinopathy of prematurity (21.7% vs. 10.3%, p = 0.03), and adverse psychomotor outcome (24.3% vs. 1.8%, OR 13.154, 95%CI 1.6,110.2, p = 0.005). The data suggest an association between vaginal U. spp. colonization in early pregnancy and adverse short- and long-term outcome of very preterm infants.
Collapse
Affiliation(s)
- Judith Rittenschober-Böhm
- Division of Neonatology, Intensive Care and Neuropediatrics, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria; (T.H.); (R.F.); (B.P.); (K.G.); (A.B.); (K.P.)
| | - Tanja Habermüller
- Division of Neonatology, Intensive Care and Neuropediatrics, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria; (T.H.); (R.F.); (B.P.); (K.G.); (A.B.); (K.P.)
| | - Thomas Waldhoer
- Department of Epidemiology, Center of Public Health, Medical University of Vienna, 1090 Vienna, Austria;
| | - Renate Fuiko
- Division of Neonatology, Intensive Care and Neuropediatrics, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria; (T.H.); (R.F.); (B.P.); (K.G.); (A.B.); (K.P.)
| | - Stefan M. Schulz
- Department of Pathology, Medical University of Vienna, 1090 Vienna, Austria;
| | - Birgit Pimpel
- Division of Neonatology, Intensive Care and Neuropediatrics, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria; (T.H.); (R.F.); (B.P.); (K.G.); (A.B.); (K.P.)
| | - Katharina Goeral
- Division of Neonatology, Intensive Care and Neuropediatrics, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria; (T.H.); (R.F.); (B.P.); (K.G.); (A.B.); (K.P.)
| | - Armin Witt
- Division of Obstetrics and Feto-Maternal Medicine, Department of Obstetrics and Gynecology, Medical University of Vienna, 1090 Vienna, Austria;
| | - Angelika Berger
- Division of Neonatology, Intensive Care and Neuropediatrics, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria; (T.H.); (R.F.); (B.P.); (K.G.); (A.B.); (K.P.)
| | - Karin Pichler
- Division of Neonatology, Intensive Care and Neuropediatrics, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria; (T.H.); (R.F.); (B.P.); (K.G.); (A.B.); (K.P.)
| |
Collapse
|
16
|
Moschino L, Duci M, Fascetti Leon F, Bonadies L, Priante E, Baraldi E, Verlato G. Optimizing Nutritional Strategies to Prevent Necrotizing Enterocolitis and Growth Failure after Bowel Resection. Nutrients 2021; 13:nu13020340. [PMID: 33498880 PMCID: PMC7910892 DOI: 10.3390/nu13020340] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/18/2021] [Accepted: 01/19/2021] [Indexed: 02/07/2023] Open
Abstract
Necrotizing enterocolitis (NEC), the first cause of short bowel syndrome (SBS) in the neonate, is a serious neonatal gastrointestinal disease with an incidence of up to 11% in preterm newborns less than 1500 g of birth weight. The rate of severe NEC requiring surgery remains high, and it is estimated between 20–50%. Newborns who develop SBS need prolonged parenteral nutrition (PN), experience nutrient deficiency, failure to thrive and are at risk of neurodevelopmental impairment. Prevention of NEC is therefore mandatory to avoid SBS and its associated morbidities. In this regard, nutritional practices seem to play a key role in early life. Individualized medical and surgical therapies, as well as intestinal rehabilitation programs, are fundamental in the achievement of enteral autonomy in infants with acquired SBS. In this descriptive review, we describe the most recent evidence on nutritional practices to prevent NEC, the available tools to early detect it, the surgical management to limit bowel resection and the best nutrition to sustain growth and intestinal function.
Collapse
MESH Headings
- Enterocolitis, Necrotizing/complications
- Enterocolitis, Necrotizing/diagnosis
- Enterocolitis, Necrotizing/prevention & control
- Enterocolitis, Necrotizing/surgery
- Failure to Thrive/prevention & control
- Humans
- Infant
- Infant Nutritional Physiological Phenomena
- Infant, Newborn
- Infant, Premature
- Infant, Premature, Diseases/diagnosis
- Infant, Premature, Diseases/prevention & control
- Infant, Premature, Diseases/surgery
- Intestines/surgery
- Short Bowel Syndrome/etiology
- Short Bowel Syndrome/prevention & control
Collapse
Affiliation(s)
- Laura Moschino
- Neonatal Intensive Care Unit, Department of Women’s and Children’s Health, University Hospital of Padova, 35128 Padova, Italy; (L.M.); (L.B.); (E.P.); (E.B.)
| | - Miriam Duci
- Pediatric Surgery Unit, Department of Women’s and Children’s Health, University Hospital of Padova, 35128 Padova, Italy; (M.D.); (F.F.L.)
| | - Francesco Fascetti Leon
- Pediatric Surgery Unit, Department of Women’s and Children’s Health, University Hospital of Padova, 35128 Padova, Italy; (M.D.); (F.F.L.)
| | - Luca Bonadies
- Neonatal Intensive Care Unit, Department of Women’s and Children’s Health, University Hospital of Padova, 35128 Padova, Italy; (L.M.); (L.B.); (E.P.); (E.B.)
| | - Elena Priante
- Neonatal Intensive Care Unit, Department of Women’s and Children’s Health, University Hospital of Padova, 35128 Padova, Italy; (L.M.); (L.B.); (E.P.); (E.B.)
| | - Eugenio Baraldi
- Neonatal Intensive Care Unit, Department of Women’s and Children’s Health, University Hospital of Padova, 35128 Padova, Italy; (L.M.); (L.B.); (E.P.); (E.B.)
| | - Giovanna Verlato
- Neonatal Intensive Care Unit, Department of Women’s and Children’s Health, University Hospital of Padova, 35128 Padova, Italy; (L.M.); (L.B.); (E.P.); (E.B.)
- Correspondence: ; Tel.: +39-0498211428
| |
Collapse
|
17
|
Sun T, Fu J. Analysis of the Clinical Features of Intrauterine Ureaplasma urealyticum Infection in Preterm Infants: A Case-Control Study. Front Pediatr 2021; 9:774150. [PMID: 34956983 PMCID: PMC8696116 DOI: 10.3389/fped.2021.774150] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 11/15/2021] [Indexed: 11/14/2022] Open
Abstract
Objective: To analyze the clinical characteristics of intrauterine Ureaplasma urealyticum (UU) infection in premature infants. Method: In this single-center retrospective case-control study, 291 preterm infants born in our hospital and hospitalized in our department and gestational age no more than 32 weeks, birth weight no more than 2000 g were included from January 2019 to January 2021. Lower respiratory tract secretion, gastric fluid and urine were collected for UU RNA detection within 48 h after birth. Intrauterine UU infection is defined by at least one positive UU-PCR test of secreta or excreta of preterm infants after birth. The UU infection group included 86 preterm infants and the non-UU infection group included 205 preterm infants. We compared their clinical features, hemogram changes and disease outcomes using statistical analyses. Results: The clinical characteristics of premature infants such as the duration of oxygen use and ventilator use in hospital were significantly prolonged in the UU infection group (P < 0.05). The levels of leukocytes, platelet and procalcitonin in the UU infection group were significantly higher than in the non-UU infection group (P < 0.05). In terms of preterm complications, only the incidences of bronchopulmonary dysplasia, retinopathy of prematurity and metabolic bone disease in premature infants in the UU infection group were significantly higher than those in the non-UU infection group (P < 0.05). The mode of delivery, maternal premature rupture of membranes, and postnatal leukocyte level were independent risk factors for UU infection, while gestational hypertension was a protective factor for UU infection. The level of leukocytes in postnatal hemogram of premature infants could be used as a diagnostic index of UU infection, but the diagnostic accuracy was poor. Conclusion: In our study, UU infection can increase the incidence of bronchopulmonary dysplasia, retinopathy of prematurity and metabolic bone disease in preterm infants, but have no effect on the incidence of necrotizing enterocolitis, intracranial hemorrhage, white matter damage and other diseases in preterm infants. For high-risk premature infants, UU should be detected as soon as possible after birth, early intervention and drug treatment necessarily can improve the prognosis as much as possible.
Collapse
Affiliation(s)
- Tong Sun
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jianhua Fu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
18
|
Zöllkau J, Pieper DH, Pastuschek J, Makarewicz O, Mentzel HJ, Dawczynski K, Schleußner E. Lethal Neonatal Respiratory Failure by Perinatal Transmission of Ureaplasma Parvum after Maternal PPROM. Z Geburtshilfe Neonatol 2020; 225:361-365. [PMID: 33339061 DOI: 10.1055/a-1326-2719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
A primiparous pregnant woman was admitted due to preterm premature rupture of membranes (PPROM) at 27+0 week of gestational age (WGA). Conventional vaginal microbiological analysis had no pathological finding. Management decisions based on national guidelines included antenatal corticoids, tocolytics and antibiotics. Unstoppable efforts of preterm labor in 28+0 WGA and supposed amniotic infection syndrome necessitated emergency cesarean section. The preterm infant underwent NICU therapy, developed an early-onset neonatal sepsis and therapy-refractory pulmonary insufficiency with consecutive right heart failure, resulting in death on the 36th day of life. Microbiota analyses by 16Sr DNA sequencing was performed from maternal vaginal swabs and from neonatal pharyngeal swabs. Maternal antibiotic treatment resulted in depletion of physiological vaginal colonization with Lactobacillus crispatus. Ureaplasma parvum became the dominant vaginal microorganism at delivery and was detected in high relative abundance in the neonatal specimen. Progressive radiological air-space changes and interstitial pathologies associated with Ureaplasma infection (bronchopulmonary dysplasia type III) were seen early at the 3rd and distinctly from 14th day of life. This clearly demonstrates the need of vaginal colonization diagnostics in PPROM patients and awareness of the consecutive risks in the preterm. Vaginal microbiome analysis may allow individualized and targeted maternal and fetal diagnostic, prophylactic and therapeutic strategies to identify, protect and treat the high-risk neonates after PPROM.
Collapse
Affiliation(s)
- Janine Zöllkau
- Obstetrics, Jena University Hospital, Jena, Germany.,Center for Sepsis Control and Case (CSCC), Jena University Hospital, Jena, Germany
| | - Dietmar H Pieper
- Microbial Interactions and Processes Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Jana Pastuschek
- Obstetrics, Jena University Hospital, Jena, Germany.,Center for Sepsis Control and Case (CSCC), Jena University Hospital, Jena, Germany
| | - Oliwia Makarewicz
- Center for Sepsis Control and Case (CSCC), Jena University Hospital, Jena, Germany.,Institute for Infectious Diseases and Infection Control (IIMK), Jena University Hospital, Jena, Germany
| | - Hans-Joachim Mentzel
- Department of Radiology, Section Pediatric Radiology, Jena University Hospital, Jena, Germany
| | - Kristin Dawczynski
- Center for Sepsis Control and Case (CSCC), Jena University Hospital, Jena, Germany.,Department of Pediatrics, Section Neonatology, Jena University Hospital, Jena, Germany
| | - Ekkehard Schleußner
- Obstetrics, Jena University Hospital, Jena, Germany.,Center for Sepsis Control and Case (CSCC), Jena University Hospital, Jena, Germany
| |
Collapse
|
19
|
Conde-Agudelo A, Romero R, Jung EJ, Garcia Sánchez ÁJ. Management of clinical chorioamnionitis: an evidence-based approach. Am J Obstet Gynecol 2020; 223:848-869. [PMID: 33007269 PMCID: PMC8315154 DOI: 10.1016/j.ajog.2020.09.044] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/08/2020] [Accepted: 09/24/2020] [Indexed: 02/09/2023]
Abstract
This review aimed to examine the existing evidence about interventions proposed for the treatment of clinical chorioamnionitis, with the goal of developing an evidence-based contemporary approach for the management of this condition. Most trials that assessed the use of antibiotics in clinical chorioamnionitis included patients with a gestational age of ≥34 weeks and in labor. The first-line antimicrobial regimen for the treatment of clinical chorioamnionitis is ampicillin combined with gentamicin, which should be initiated during the intrapartum period. In the event of a cesarean delivery, patients should receive clindamycin at the time of umbilical cord clamping. The administration of additional antibiotic therapy does not appear to be necessary after vaginal or cesarean delivery. However, if postdelivery antibiotics are prescribed, there is support for the administration of an additional dose. Patients can receive antipyretic agents, mainly acetaminophen, even though there is no clear evidence of their benefits. Current evidence suggests that the administration of antenatal corticosteroids for fetal lung maturation and of magnesium sulfate for fetal neuroprotection to patients with clinical chorioamnionitis between 24 0/7 and 33 6/7 weeks of gestation, and possibly between 23 0/7 and 23 6/7 weeks of gestation, has an overall beneficial effect on the infant. However, delivery should not be delayed to complete the full course of corticosteroids and magnesium sulfate. Once the diagnosis of clinical chorioamnionitis has been established, delivery should be considered, regardless of the gestational age. Vaginal delivery is the safer option and cesarean delivery should be reserved for standard obstetrical indications. The time interval between the diagnosis of clinical chorioamnionitis and delivery is not related to most adverse maternal and neonatal outcomes. Patients may require a higher dose of oxytocin to achieve adequate uterine activity or greater uterine activity to effect a given change in cervical dilation. The benefit of using continuous electronic fetal heart rate monitoring in these patients is unclear. We identified the following promising interventions for the management of clinical chorioamnionitis: (1) an antibiotic regimen including ceftriaxone, clarithromycin, and metronidazole that provides coverage against the most commonly identified microorganisms in patients with clinical chorioamnionitis; (2) vaginal cleansing with antiseptic solutions before cesarean delivery with the aim of decreasing the risk of endometritis and, possibly, postoperative wound infection; and (3) antenatal administration of N-acetylcysteine, an antioxidant and antiinflammatory agent, to reduce neonatal morbidity and mortality. Well-powered randomized controlled trials are needed to assess these interventions in patients with clinical chorioamnionitis.
Collapse
Affiliation(s)
- Agustin Conde-Agudelo
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, and U.S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, and U.S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI; Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI; Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI; Detroit Medical Center, Detroit, MI; Department of Obstetrics and Gynecology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL.
| | - Eun Jung Jung
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, and U.S. Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Ángel José Garcia Sánchez
- Department of Biomedical and Diagnostic Sciences, Faculty of Medicine, University of Salamanca, Salamanca, Spain
| |
Collapse
|
20
|
Viscardi RM, Terrin ML, Magder LS, Davis NL, Dulkerian SJ, Waites KB, Ambalavanan N, Kaufman DA, Donohue P, Tuttle DJ, Weitkamp JH, Hassan HE, Eddington ND. Randomised trial of azithromycin to eradicate Ureaplasma in preterm infants. Arch Dis Child Fetal Neonatal Ed 2020; 105:615-622. [PMID: 32170033 PMCID: PMC7592356 DOI: 10.1136/archdischild-2019-318122] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 01/26/2020] [Accepted: 02/25/2020] [Indexed: 12/22/2022]
Abstract
OBJECTIVE To test whether azithromycin eradicates Ureaplasma from the respiratory tract in preterm infants. DESIGN Prospective, phase IIb randomised, double-blind, placebo-controlled trial. SETTING Seven level III-IV US, academic, neonatal intensive care units (NICUs). PATIENTS Infants 240-286 weeks' gestation (stratified 240-266; 270-286 weeks) randomly assigned within 4 days following birth from July 2013 to August 2016. INTERVENTIONS Intravenous azithromycin 20 mg/kg or an equal volume of D5W (placebo) every 24 hours for 3 days. MAIN OUTCOME MEASURES The primary efficacy outcome was Ureaplasma-free survival. Secondary outcomes were all-cause mortality, Ureaplasma clearance, physiological bronchopulmonary dysplasia (BPD) at 36 weeks' postmenstrual age, comorbidities of prematurity and duration of respiratory support. RESULTS One hundred and twenty-one randomised participants (azithromycin: n=60; placebo: n=61) were included in the intent-to-treat analysis (mean gestational age 26.2±1.4 weeks). Forty-four of 121 participants (36%) were Ureaplasma positive (azithromycin: n=19; placebo: n=25). Ureaplasma-free survival was 55/60 (92% (95% CI 82% to 97%)) for azithromycin compared with 37/61 (61% (95% CI 48% to 73%)) for placebo. Mortality was similar comparing the two treatment groups (5/60 (8%) vs 6/61 (10%)). Azithromycin effectively eradicated Ureaplasma in all azithromycin-assigned colonised infants, but 21/25 (84%) Ureaplasma-colonised participants receiving placebo were culture positive at one or more follow-up timepoints. Most of the neonatal mortality and morbidity was concentrated in 21 infants with lower respiratory tract Ureaplasma colonisation. In a subgroup analysis, physiological BPD-free survival was 5/10 (50%) (95% CI 19% to 81%) among azithromycin-assigned infants with lower respiratory tract Ureaplasma colonisation versus 2/11 (18%) (95% CI 2% to 52%) in placebo-treated infants. CONCLUSION A 3-day azithromycin regimen effectively eradicated respiratory tract Ureaplasma colonisation in this study. TRIAL REGISTRATION NUMBER NCT01778634.
Collapse
Affiliation(s)
- Rose Marie Viscardi
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Michael L Terrin
- Department of Epidemiology and Preventive Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Laurence S Magder
- Department of Epidemiology and Preventive Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Natalie L Davis
- Department of Pediatrics, University of Maryland Baltimore, Baltimore, Maryland, USA
| | - Susan J Dulkerian
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Ken B Waites
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | - David A Kaufman
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Pamela Donohue
- Department of Pediatrics, Johns Hopkins Medicine, Baltimore, Maryland, USA
| | - Deborah J Tuttle
- Department of Pediatrics, Christiana Care Health System, Newark, Delaware, USA
| | - Jorn-Hendrik Weitkamp
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Hazem E Hassan
- University of Maryland School of Pharmacy, Baltimore, Maryland, USA
| | | |
Collapse
|
21
|
|
22
|
Intra-Amniotic Infection with Ureaplasma parvum Causes Preterm Birth and Neonatal Mortality That Are Prevented by Treatment with Clarithromycin. mBio 2020; 11:mBio.00797-20. [PMID: 32576673 PMCID: PMC7315120 DOI: 10.1128/mbio.00797-20] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Preterm birth is the leading cause of neonatal morbidity and mortality worldwide. Multiple etiologies are associated with preterm birth; however, 25% of preterm infants are born to a mother with intra-amniotic infection, most commonly due to invasion of the amniotic cavity by Ureaplasma species. Much research has focused on establishing a link between Ureaplasma species and adverse pregnancy/neonatal outcomes; however, little is known about the taxonomy of and host response against Ureaplasma species. Here, we applied a multifaceted approach, including human samples, in vivo models, and in vitro manipulations, to study the maternal-fetal immunobiology of Ureaplasma infection during pregnancy. Furthermore, we investigated the use of clarithromycin as a treatment for this infection. Our research provides translational knowledge that bolsters scientific understanding of Ureaplasma species as a cause of adverse pregnancy/neonatal outcomes and gives strong evidence for the use of clarithromycin as the recommended treatment for women intra-amniotically infected with Ureaplasma species. Intra-amniotic infection is strongly associated with adverse pregnancy and neonatal outcomes. Most intra-amniotic infections are due to Ureaplasma species; however, the pathogenic potency of these genital mycoplasmas to induce preterm birth is still controversial. Here, we first laid out a taxonomic characterization of Ureaplasma isolates from women with intra-amniotic infection, which revealed that Ureaplasma parvum is the most common bacterium found in this clinical condition. Next, using animal models, we provided a causal link between intra-amniotic inoculation with Ureaplasma species and preterm birth. Importantly, the intra-amniotic inoculation of Ureaplasma species induced high rates of mortality in both preterm and term neonates. The in vivo potency of U. parvum to induce preterm birth was not associated with known virulence factors. However, term-derived and preterm-derived U. parvum isolates were capable of inducing an intra-amniotic inflammatory response. Both U. parvum isolates invaded several fetal tissues, primarily the fetal lung, and caused fetal inflammatory response syndrome. This bacterium was also detected in the placenta, reproductive tissues, and most severely in the fetal membranes, inducing a local inflammatory response that was replicated in an in vitro model. Importantly, treatment with clarithromycin, a recently recommended yet not widely utilized antibiotic, prevented the adverse pregnancy and neonatal outcomes induced by U. parvum. These findings shed light on the maternal-fetal immunobiology of intra-amniotic infection.
Collapse
|
23
|
Prophylactic Intra-Uterine β-Cyclodextrin Administration during Intra-Uterine Ureaplasma parvum Infection Partly Prevents Liver Inflammation without Interfering with the Enterohepatic Circulation of the Fetal Sheep. Nutrients 2020; 12:nu12051312. [PMID: 32380648 PMCID: PMC7284867 DOI: 10.3390/nu12051312] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/27/2020] [Accepted: 04/29/2020] [Indexed: 12/20/2022] Open
Abstract
Chorioamnionitis can lead to inflammation and injury of the liver and gut, thereby predisposing patients to adverse outcomes such as necrotizing enterocolitis (NEC). In addition, intestinal bile acids (BAs) accumulation is causally linked to NEC development. Plant sterols are a promising intervention to prevent NEC development, considering their anti-inflammatory properties in the liver. Therefore, we investigated whether an intra-amniotic (IA) Ureaplasma parvum (UP) infection affected the liver and enterohepatic circulation (EHC) and evaluated whether an IA administered plant sterol mixture dissolved in β-cyclodextrin exerted prophylactic effects. An ovine chorioamnionitis model was used in which liver inflammation and the EHC were assessed following IA UP exposure in the presence or absence of IA prophylactic plant sterols (a mixture of β-sitosterol and campesterol dissolved in β-cyclodextrin (carrier)) or carrier alone. IA UP exposure caused an inflammatory reaction in the liver, histologically seen as clustered and conflated hepatic erythropoiesis in the parenchyma, which was partially prevented by IA administration of sterol + β-cyclodextrin, or β-cyclodextrin alone. In addition, IA administration of β-cyclodextrin prior to UP caused changes in the expression of several hepatic BAs transporters, without causing alterations in other aspects of the EHC. Thereby, the addition of plant sterols to the carrier β-cyclodextrin did not have additional effects.
Collapse
|
24
|
Wang Q, Wang K, Zhang Y, Lu C, Yan Y, Huang X, Zhou J, Chen L, Wang D. Neonatal Ureaplasma parvum meningitis: a case report and literature review. Transl Pediatr 2020; 9:174-179. [PMID: 32477918 PMCID: PMC7237967 DOI: 10.21037/tp.2020.02.04] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Ureaplasma parvum (U. parvum) is common commensal in the female genitourinary tract. Despite U. parvum has been associated with chorioamnionitis, abortion, prematurity and perinatal complications, the invasive central nervous system (CNS) infection is rare in neonates. Diagnosis of U. parvum meningitis can be difficult for the atypical presentations and sterile cultures by conventional methods. Metagenomic next-generation sequencing (mNGS) could identify a broad range of human pathogens in a target-independent manner. Here, we performed mNGS to search for the infectious etiology in a term infant presenting with fever and seizure. U. parvum genome was identified by mNGS and further confirmed by PCR in the same cerebrospinal fluid (CSF) sample. As the quick and timely diagnosis, the baby was successfully treated with erythromycin for 4 weeks without complication. The clinical follow-up has showed that the physical and mental development are normal. In conclusion, mNGS may a promising diagnostic technology for U. parvum meningitis. As mNGS is able to identify diverse microbes in a single run, it could be a useful strategy to detection the clinical causative pathogens with atypical features in neonates.
Collapse
Affiliation(s)
- Qiu Wang
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Kai Wang
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Yuanbo Zhang
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Chaosheng Lu
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Yana Yan
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Xiaoxia Huang
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Jing Zhou
- Department of Obstetrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Lijiang Chen
- Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Dan Wang
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| |
Collapse
|
25
|
Heymans C, de Lange IH, Hütten MC, Lenaerts K, de Ruijter NJE, Kessels LCGA, Rademakers G, Melotte V, Boesmans W, Saito M, Usuda H, Stock SJ, Spiller OB, Beeton ML, Payne MS, Kramer BW, Newnham JP, Jobe AH, Kemp MW, van Gemert WG, Wolfs TGAM. Chronic Intra-Uterine Ureaplasma parvum Infection Induces Injury of the Enteric Nervous System in Ovine Fetuses. Front Immunol 2020; 11:189. [PMID: 32256485 PMCID: PMC7089942 DOI: 10.3389/fimmu.2020.00189] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 01/24/2020] [Indexed: 01/18/2023] Open
Abstract
Background: Chorioamnionitis, inflammation of the fetal membranes during pregnancy, is often caused by intra-amniotic (IA) infection with single or multiple microbes. Chorioamnionitis can be either acute or chronic and is associated with adverse postnatal outcomes of the intestine, including necrotizing enterocolitis (NEC). Neonates with NEC have structural and functional damage to the intestinal mucosa and the enteric nervous system (ENS), with loss of enteric neurons and glial cells. Yet, the impact of acute, chronic, or repetitive antenatal inflammatory stimuli on the development of the intestinal mucosa and ENS has not been studied. The aim of this study was therefore to investigate the effect of acute, chronic, and repetitive microbial exposure on the intestinal mucosa, submucosa and ENS in premature lambs. Materials and Methods: A sheep model of pregnancy was used in which the ileal mucosa, submucosa, and ENS were assessed following IA exposure to lipopolysaccharide (LPS) for 2 or 7 days (acute), Ureaplasma parvum (UP) for 42 days (chronic), or repetitive microbial exposure (42 days UP with 2 or 7 days LPS). Results: IA LPS exposure for 7 days or IA UP exposure for 42 days caused intestinal injury and inflammation in the mucosal and submucosal layers of the gut. Repetitive microbial exposure did not further aggravate injury of the terminal ileum. Chronic IA UP exposure caused significant structural ENS alterations characterized by loss of PGP9.5 and S100β immunoreactivity, whereas these changes were not found after re-exposure of chronic UP-exposed fetuses to LPS for 2 or 7 days. Conclusion: The in utero loss of PGP9.5 and S100β immunoreactivity following chronic UP exposure corresponds with intestinal changes in neonates with NEC and may therefore form a novel mechanistic explanation for the association of chorioamnionitis and NEC.
Collapse
Affiliation(s)
- Cathelijne Heymans
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| | - Ilse H de Lange
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands.,Department of Pediatrics, School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, Netherlands
| | - Matthias C Hütten
- Department of Pediatrics, School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, Netherlands.,Neonatology, Department of Pediatrics, Maastricht University Medical Center, Maastricht, Netherlands.,Neonatology, Department of Pediatrics, University Hospital Aachen, Aachen, Germany.,Neonatology, Department of Pediatrics, University Children's Hospital Würzburg, Würzburg, Germany
| | - Kaatje Lenaerts
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| | - Nadine J E de Ruijter
- Department of Pediatrics, School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, Netherlands
| | - Lilian C G A Kessels
- Department of Pediatrics, School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, Netherlands
| | - Glenn Rademakers
- Department of Pathology, School for Oncology and Developmental Biology (GROW), Maastricht University Medical Center, Maastricht, Netherlands
| | - Veerle Melotte
- Department of Pathology, School for Oncology and Developmental Biology (GROW), Maastricht University Medical Center, Maastricht, Netherlands
| | - Werend Boesmans
- Department of Pathology, School for Oncology and Developmental Biology (GROW), Maastricht University Medical Center, Maastricht, Netherlands.,Biomedical Research Institute (BIOMED), Hasselt University, Hasselt, Belgium
| | - Masatoshi Saito
- Division of Obstetrics and Gynecology, University of Western Australia, Perth, WA, Australia.,Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Haruo Usuda
- Division of Obstetrics and Gynecology, University of Western Australia, Perth, WA, Australia.,Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Sarah J Stock
- Usher Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Owen B Spiller
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Michael L. Beeton
- Division of Neonatology/Pulmonary Biology, The Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, United States
| | - Matthew S Payne
- Division of Obstetrics and Gynecology, University of Western Australia, Perth, WA, Australia
| | - Boris W Kramer
- Department of Pediatrics, School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, Netherlands.,Neonatology, Department of Pediatrics, Maastricht University Medical Center, Maastricht, Netherlands
| | - John P Newnham
- Division of Obstetrics and Gynecology, University of Western Australia, Perth, WA, Australia
| | - Alan H Jobe
- Division of Obstetrics and Gynecology, University of Western Australia, Perth, WA, Australia.,Division of Neonatology/Pulmonary Biology, The Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, United States
| | - Matthew W Kemp
- Division of Obstetrics and Gynecology, University of Western Australia, Perth, WA, Australia.,Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan.,School of Veterinary and Life Sciences, Murdoch University, Perth, WA, Australia
| | - Wim G van Gemert
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands.,Pediatric Surgery, Department of Surgery, Maastricht University Medical Center, Maastricht, Netherlands.,Department of Surgery, University Hospital Aachen, Aachen, Germany
| | - Tim G A M Wolfs
- Department of Pediatrics, School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, Netherlands.,Department of Biomedical Engineering (BMT), School for Cardiovascular Diseases (CARIM), Maastricht University, Maastricht, Netherlands
| |
Collapse
|
26
|
Silwedel C, Speer CP, Härtel C, Glaser K. Ureaplasma-Driven Neuroinflammation in Neonates: Assembling the Puzzle Pieces. Neonatology 2020; 117:665-672. [PMID: 33271546 PMCID: PMC7949233 DOI: 10.1159/000512019] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 09/22/2020] [Indexed: 12/12/2022]
Abstract
Ureaplasma species (spp.) are commonly regarded as low-virulence colonizers of the genitourinary tract. Intrauterine Ureaplasma infection, however, has been associated with chorioamnionitis and preterm birth. The overall impact of a neonatal Ureaplasma colonization is yet to be understood. High pathogen prevalence and frequent neurological morbidities particularly in immature preterm infants call for an assessment of the significance of Ureaplasma spp. in neonatal neuroinflammation. This narrative review summarizes clinical data, animal studies, and in vitro results to elucidate potential Ureaplasma-associated neurological morbidities as well as underlying mechanisms. Increasing evidence indicates an involvement of Ureaplasma spp. in invasive central nervous system infections, suggesting a meticulous ability of Ureaplasma spp. to interfere with immune defense mechanisms. Ultimately, Ureaplasma spp. should be considered as relevant pathogens in neonatal neuroinflammation.
Collapse
Affiliation(s)
- Christine Silwedel
- University Children's Hospital, University of Wuerzburg, Wuerzburg, Germany,
| | - Christian P Speer
- University Children's Hospital, University of Wuerzburg, Wuerzburg, Germany
| | - Christoph Härtel
- University Children's Hospital, University of Wuerzburg, Wuerzburg, Germany
| | - Kirsten Glaser
- University Children's Hospital, University of Wuerzburg, Wuerzburg, Germany.,Department of Women and Child Health, Center for Pediatric Research Leipzig, Division of Neonatology, University of Leipzig, Leipzig, Germany
| |
Collapse
|
27
|
Silwedel C, Speer CP, Haarmann A, Fehrholz M, Claus H, Schlegel N, Glaser K. Ureaplasma Species Modulate Cytokine and Chemokine Responses in Human Brain Microvascular Endothelial Cells. Int J Mol Sci 2019; 20:ijms20143583. [PMID: 31336668 PMCID: PMC6678482 DOI: 10.3390/ijms20143583] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/11/2019] [Accepted: 07/18/2019] [Indexed: 12/17/2022] Open
Abstract
Ureaplasma species are common colonizers of the adult genitourinary tract and often considered as low-virulence commensals. Intraamniotic Ureaplasma infections, however, facilitate chorioamnionitis and preterm birth, and cases of Ureaplasma-induced neonatal sepsis, pneumonia, and meningitis raise a growing awareness of their clinical relevance. In vitro studies are scarce but demonstrate distinct Ureaplasma-driven impacts on immune mechanisms. The current study addressed cytokine and chemokine responses upon exposure of native or lipopolysaccharide (LPS) co-stimulated human brain microvascular endothelial cells (HBMEC) to Ureaplasma urealyticum or U. parvum, using qRT-PCR, RNA sequencing, multi-analyte immunoassay, and flow cytometry. Ureaplasma exposure in native HBMEC reduced monocyte chemoattractant protein (MCP)-3 mRNA expression (p < 0.01, vs. broth). In co-stimulated HBMEC, Ureaplasma spp. attenuated LPS-evoked mRNA responses for C-X-C chemokine ligand 5, MCP-1, and MCP-3 (p < 0.05, vs. LPS) and mitigated LPS-driven interleukin (IL)-1α protein secretion, as well as IL-8 mRNA and protein responses (p < 0.05). Furthermore, Ureaplasma isolates increased C-X-C chemokine receptor 4 mRNA levels in native and LPS co-stimulated HBMEC (p < 0.05). The presented results may imply immunomodulatory capacities of Ureaplasma spp. which may ultimately promote chronic colonization and long-term neuroinflammation.
Collapse
Affiliation(s)
- Christine Silwedel
- University Children's Hospital, University of Wuerzburg, Josef-Schneider-Str. 2, 97080 Wuerzburg, Germany.
| | - Christian P Speer
- University Children's Hospital, University of Wuerzburg, Josef-Schneider-Str. 2, 97080 Wuerzburg, Germany
| | - Axel Haarmann
- Department of Neurology, University of Wuerzburg, Josef-Schneider-Str. 11, 97080 Wuerzburg, Germany
| | - Markus Fehrholz
- University Children's Hospital, University of Wuerzburg, Josef-Schneider-Str. 2, 97080 Wuerzburg, Germany
| | - Heike Claus
- Institute for Hygiene and Microbiology, University of Wuerzburg, Josef-Schneider-Str. 2, 97080 Wuerzburg, Germany
| | - Nicolas Schlegel
- Department of Surgery I, University of Wuerzburg, Oberduerrbacherstr. 6, 97080 Wuerzburg, Germany
| | - Kirsten Glaser
- University Children's Hospital, University of Wuerzburg, Josef-Schneider-Str. 2, 97080 Wuerzburg, Germany
| |
Collapse
|
28
|
Ureaplasma species modulate cell adhesion molecules and growth factors in human brain microvascular endothelial cells. Cytokine 2019; 121:154737. [PMID: 31158700 DOI: 10.1016/j.cyto.2019.154737] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 05/23/2019] [Accepted: 05/27/2019] [Indexed: 12/20/2022]
Abstract
Ureaplasma species (spp.) are considered commensals of the adult urogenital tract, but may cause chorioamnionitis and preterm birth as well as sepsis and meningitis in neonates. Pathomechanisms in Ureaplasma-driven neuroinflammation are largely unknown. This study addressed mRNA and protein expression of intercellular and vascular cell adhesion molecules (ICAM-1, VCAM-1), granulocyte-colony stimulating factor (G-CSF), and vascular endothelial growth factor (VEGF) in native or lipopolysaccharide (LPS) co-stimulated human brain microvascular endothelial cells (HBMEC) exposed to Ureaplasma (U.) urealyticum or U. parvum. Ureaplasma spp. reduced G-CSF mRNA (p < 0.05) and protein expression (p < 0.01) and increased VEGF mRNA levels (p < 0.01) in native HBMEC. Upon co-stimulation, Ureaplasma isolates enhanced LPS-evoked VEGF and ICAM-1 mRNA expression (p < 0.05), but mitigated G-CSF and VCAM-1 mRNA responses (p < 0.05). In line with previous findings, our results indicate an ability of Ureaplasma spp. to compromise blood-brain barrier integrity, mitigate immune defense, and subdue neuroprotective mechanisms. This may facilitate intracerebral inflammation, allow chronic infections, and promote brain injury. More pronounced effects in co-stimulated cells may indicate an immunomodulatory capacity of Ureaplasma spp.
Collapse
|
29
|
Silwedel C, Fehrholz M, Speer CP, Ruf KC, Manig S, Glaser K. Differential modulation of pulmonary caspases: Is this the key to Ureaplasma-driven chronic inflammation? PLoS One 2019; 14:e0216569. [PMID: 31067276 PMCID: PMC6506144 DOI: 10.1371/journal.pone.0216569] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 04/23/2019] [Indexed: 12/26/2022] Open
Abstract
Although accepted agents in chorioamnionitis and preterm birth, the role of Ureaplasma species (spp.) in inflammation-driven morbidities of prematurity, including the development of bronchopulmonary dysplasia, remains controversial. To add to scarce in vitro data addressing the pro-inflammatory capacity of Ureaplasma spp., pulmonary epithelial-like A549 cells and human pulmonary microvascular endothelial cells (HPMEC) were incubated with Ureaplasma (U.) urealyticum, U. parvum, and Escherichia coli lipopolysaccharide (LPS). Ureaplasma isolates down-regulated caspase mRNA levels in A549 cells (caspase 8: p<0.001, 9: p<0.001, vs. broth), while increasing caspase protein expression, enzyme activity, and cell death in HPMEC (active caspase 3: p<0.05, caspase 8: p<0.05, active caspase 9: p<0.05, viability: p<0.05). LPS, contrarily, induced caspase mRNA expression in HPMEC (caspase 3: p<0.01, 4: p<0.001, 5: p<0.001, 8: p<0.001, vs. control), but not in A549 cells, and did not affect enzyme activity or protein levels in either cell line. LPS, but neither Ureaplasma isolate, enhanced mRNA expression of pro-inflammatory interleukin (IL)-6 in both A549 (p<0.05, vs. control) and HPMEC (p<0.001) as well as tumor necrosis factor-α (p<0.01), IL-1β (p<0.001), and IL-8 (p<0.05) in HPMEC. We are therefore the first to demonstrate a differential modulation of pulmonary caspases by Ureaplasma spp. in vitro. Ureaplasma-driven enhanced protein expression and activity of caspases in pulmonary endothelial cells result in cell death and may cause structural damage. Down-regulated caspase mRNA in pulmonary epithelial cells, contrarily, may indicate Ureaplasma-induced inhibition of apoptosis and prevent effective immune responses. Both may ultimately contribute to chronic Ureaplasma colonization and long-term pulmonary inflammation.
Collapse
Affiliation(s)
- Christine Silwedel
- University Children´s Hospital, University of Wuerzburg, Wuerzburg, Germany
- * E-mail:
| | - Markus Fehrholz
- University Children´s Hospital, University of Wuerzburg, Wuerzburg, Germany
| | - Christian P. Speer
- University Children´s Hospital, University of Wuerzburg, Wuerzburg, Germany
| | - Katharina C. Ruf
- University Children´s Hospital, University of Wuerzburg, Wuerzburg, Germany
| | - Steffi Manig
- Institute of Hygiene and Microbiology, University of Wuerzburg, Wuerzburg, Germany
| | - Kirsten Glaser
- University Children´s Hospital, University of Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
30
|
Glaser K, Gradzka-Luczewska A, Szymankiewicz-Breborowicz M, Kawczynska-Leda N, Henrich B, Waaga-Gasser AM, Speer CP. Perinatal Ureaplasma Exposure Is Associated With Increased Risk of Late Onset Sepsis and Imbalanced Inflammation in Preterm Infants and May Add to Lung Injury. Front Cell Infect Microbiol 2019; 9:68. [PMID: 31001484 PMCID: PMC6454044 DOI: 10.3389/fcimb.2019.00068] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 03/04/2019] [Indexed: 12/14/2022] Open
Abstract
Background: Controversy remains concerning the impact of Ureaplasma on preterm neonatal morbidity. Methods: Prospective single-center study in very low birth weight infants <30 weeks' gestation. Cord blood and initial nasopharyngeal swabs were screened for Ureaplasma parvum and U. urealyticum using culture technique and polymerase chain reaction. Neonatal outcomes were followed until death or discharge. Multi-analyte immunoassay provided cord blood levels of inflammatory markers. Using multivariate regression analyses, perinatal Ureaplasma exposure was evaluated as risk factor for the development of bronchopulmonary dysplasia (BPD), other neonatal morbidities until discharge and systemic inflammation at admission. Results: 40/103 (39%) infants were positive for Ureaplasma in one or both specimens, with U. parvum being the predominant species. While exposure to Ureaplasma alone was not associated with BPD, we found an increased risk of BPD in Ureaplasma-positive infants ventilated ≥5 days (OR 1.64; 95% CI 0.12–22.98; p = 0.009). Presence of Ureaplasma was associated with a 7-fold risk of late onset sepsis (LOS) (95% CI 1.80–27.39; p = 0.014). Moreover, Ureaplasma-positive infants had higher I/T ratios (b 0.39; 95% CI 0.08–0.71; p = 0.014), increased levels of interleukin (IL)-17 (b 0.16; 95% CI 0.02–0.30; p = 0.025) and matrix metalloproteinase 8 (b 0.77; 95% CI 0.10–1.44; p = 0.020), decreased levels of IL-10 (b −0.77; 95% CI −1.58 to −0.01; p = 0.043) and increased ratios of Tumor necrosis factor-α, IL-8, and IL-17 to anti-inflammatory IL-10 (p = 0.003, p = 0.012, p < 0.001). Conclusions: Positive Ureaplasma screening was not associated with BPD. However, exposure contributed to BPD in infants ventilated ≥5 days and conferred an increased risk of LOS and imbalanced inflammatory cytokine responses.
Collapse
Affiliation(s)
- Kirsten Glaser
- University Children's Hospital, University of Wuerzburg, Wuerzburg, Germany
| | | | | | | | - Birgit Henrich
- Institute of Medical Microbiology and Hospital Hygiene, University Clinic of Heinrich-Heine University Duesseldorf, Duesseldorf, Germany
| | - Ana Maria Waaga-Gasser
- Department of Surgery I, Molecular Oncology and Immunology, University of Wuerzburg, Wuerzburg, Germany
| | - Christian P Speer
- University Children's Hospital, University of Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
31
|
Naeem A, Ahmed I, Silveyra P. Bronchopulmonary Dysplasia: An Update on Experimental Therapeutics. EUROPEAN MEDICAL JOURNAL 2019. [DOI: 10.33590/emj/10313109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic inflammatory lung disease that affects thousands of newborns and infants every year. Although it is accepted that BPD results from lung damage and inflammation triggered by mechanical ventilation and hyperoxia, the causes and molecular events leading to lung damage and arrested development remain unknown. While recent advances in neonatal care have improved the survival of very low-weight infants, the rates of BPD have not improved accordingly. This is mainly due to our limited understanding of the disease’s pathogenesis and the effective therapeutic options available. Current therapeutics for BPD involve ventilation management, steroid treatment, and administration of various agents, such as pulmonary surfactant, caffeine, vitamin A, nitric oxide, and stem cells. However, the efficacy of these agents in preventing and ameliorating BPD symptoms varies depending on the populations studied and the disease stage. As the field moves towards personalised therapeutic approaches, this review summarises clinical and experimental studies conducted in various models, aiming to increase understanding of the cellular and molecular mechanisms by which these agents can prevent or treat BPD. Due to the increasing number of extremely premature infants, it is imperative that we continue to work towards understanding the mechanisms of BPD pathogenesis and generating more effective therapeutic options.
Collapse
Affiliation(s)
- Anika Naeem
- Pulmonary Immunology and Physiology Laboratory (PIP), Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Imtiaz Ahmed
- Pulmonary Immunology and Physiology Laboratory (PIP), Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Patricia Silveyra
- Pulmonary Immunology and Physiology Laboratory (PIP), Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| |
Collapse
|
32
|
Silwedel C, Haarmann A, Fehrholz M, Claus H, Speer CP, Glaser K. More than just inflammation: Ureaplasma species induce apoptosis in human brain microvascular endothelial cells. J Neuroinflammation 2019; 16:38. [PMID: 30764830 PMCID: PMC6374915 DOI: 10.1186/s12974-019-1413-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 01/24/2019] [Indexed: 02/07/2023] Open
Abstract
Background Ureaplasma species (spp.) are commonly regarded as low-virulent commensals but may cause invasive diseases in immunocompromised adults and in neonates, including neonatal meningitis. The interactions of Ureaplasma spp. with host defense mechanisms are poorly understood. This study addressed Ureaplasma-driven cell death, concentrating on apoptosis as well as inflammatory cell death. Methods Human brain microvascular endothelial cells (HBMEC) were exposed to Ureaplasma (U.) urealyticum serovar 8 (Uu8) and U. parvum serovar 3 (Up3). Resulting numbers of dead cells as well as mRNA levels and enzyme activity of key agents in programmed cell death were assessed by flow cytometry, RNA sequencing, and qRT-PCR, respectively. xCELLigence data were used for real-time monitoring of changes in cell adhesion properties. Results Both Ureaplasma isolates induced cell death (p < 0.05, vs. broth). Furthermore, Ureaplasma spp. enhanced mRNA levels for genes in apoptosis, including caspase 3 (Up3 p < 0.05, vs. broth), caspase 7 (p < 0.01), and caspase 9 (Up3 p < 0.01). Caspase 3 activity was increased upon Uu8 exposure (p < 0.01). Vice versa, Ureaplasma isolates downregulated mRNA levels for proteins involved in inflammatory cell death, namely caspase 1 (Uu8 p < 0.01, Up3 p < 0.001), caspase 4 (Uu8 p < 0.05, Up3 p < 0.01), NOD-like receptor pyrin domain-containing 3 (Uu8 p < 0.05), and receptor-interacting protein kinase 3 (p < 0.05). Conclusions By inducing apoptosis in HBMEC as main constituents of the blood-brain barrier, Ureaplasma spp. may provoke barrier breakdown. Simultaneous suppression of inflammatory cell death may additionally attenuate host defense strategies. Ultimate consequence could be invasive and long-term CNS infections by Ureaplasma spp. Electronic supplementary material The online version of this article (10.1186/s12974-019-1413-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Christine Silwedel
- University Children's Hospital, University of Wuerzburg, Josef-Schneider-Str. 2, 97080, Wuerzburg, Germany.
| | - Axel Haarmann
- Department of Neurology, University of Wuerzburg, Josef-Schneider-Str. 11, 97080, Wuerzburg, Germany
| | - Markus Fehrholz
- University Children's Hospital, University of Wuerzburg, Josef-Schneider-Str. 2, 97080, Wuerzburg, Germany
| | - Heike Claus
- Institute for Hygiene and Microbiology, University of Wuerzburg, Josef-Schneider-Str. 2, 97080, Wuerzburg, Germany
| | - Christian P Speer
- University Children's Hospital, University of Wuerzburg, Josef-Schneider-Str. 2, 97080, Wuerzburg, Germany
| | - Kirsten Glaser
- University Children's Hospital, University of Wuerzburg, Josef-Schneider-Str. 2, 97080, Wuerzburg, Germany
| |
Collapse
|
33
|
Dong Y, Glaser K, Speer CP. Late-onset sepsis caused by Gram-negative bacteria in very low birth weight infants: a systematic review. Expert Rev Anti Infect Ther 2019; 17:177-188. [PMID: 30640556 DOI: 10.1080/14787210.2019.1568871] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Very low birth weight (VLBW) infants are highly susceptible to late-onset sepsis (LOS). Compared to Gram-positive bacteria, Gram-negative bacteria are less common to cause LOS, but are associated with a more severe clinical manifestation, higher mortality, and increased risk of neonatal morbidity. So far, the clinical picture of late-onset Gram-negative sepsis (LOGNS) in VLBW infants has not been elucidated. An up-to-date and thorough understanding of the clinical picture of LOGNS in VLBW infants is important to optimize current anti-sepsis protocols. Areas covered: Literature published in the last three decades was searched for data on the epidemiology, pathogen profile, risk factors, clinical manifestations, laboratory parameters, mortality, and short-term and long-term morbidity of LOGNS in VLBW infants. Expert opinion: Gram-negative bacteria are major contributors of neonatal morbidity and mortality in VLBW infants with LOS, potentially posing a significant disease burden. Unravelling the pathogen-specific clinical picture of LOGNS and the underlying mechanisms is of particular interest. VLBW infants may differ from more mature neonates in terms of disease burden and clinical course of LOGNS. Epidemiologic studies aided by advanced molecular techniques may help to develop anti-sepsis protocols specialized for VLBW infants, with strategies targeting Gram-negative bacteria.
Collapse
Affiliation(s)
- Ying Dong
- a University Children's Hospital , University of Wuerzburg , Wuerzburg , Germany.,b Department of Neonatology , Children's Hospital of Fudan University , Shanghai , China
| | - Kirsten Glaser
- a University Children's Hospital , University of Wuerzburg , Wuerzburg , Germany
| | - Christian P Speer
- a University Children's Hospital , University of Wuerzburg , Wuerzburg , Germany
| |
Collapse
|
34
|
Silwedel C, Speer CP, Haarmann A, Fehrholz M, Claus H, Buttmann M, Glaser K. Novel insights into neuroinflammation: bacterial lipopolysaccharide, tumor necrosis factor α, and Ureaplasma species differentially modulate atypical chemokine receptor 3 responses in human brain microvascular endothelial cells. J Neuroinflammation 2018; 15:156. [PMID: 29792190 PMCID: PMC5966865 DOI: 10.1186/s12974-018-1170-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 04/19/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Atypical chemokine receptor 3 (ACKR3, synonym CXCR7) is increasingly considered relevant in neuroinflammatory conditions, in which its upregulation contributes to compromised endothelial barrier function and may ultimately allow inflammatory brain injury. While an impact of ACKR3 has been recognized in several neurological autoimmune diseases, neuroinflammation may also result from infectious agents, including Ureaplasma species (spp.). Although commonly regarded as commensals of the adult urogenital tract, Ureaplasma spp. may cause invasive infections in immunocompromised adults as well as in neonates and appear to be relevant pathogens in neonatal meningitis. Nonetheless, clinical and in vitro data on Ureaplasma-induced inflammation are scarce. METHODS We established a cell culture model of Ureaplasma meningitis, aiming to analyze ACKR3 variances as a possible pathomechanism in Ureaplasma-associated neuroinflammation. Non-immortalized human brain microvascular endothelial cells (HBMEC) were exposed to bacterial lipopolysaccharide (LPS) or tumor necrosis factor-α (TNF-α), and native as well as LPS-primed HBMEC were cultured with Ureaplasma urealyticum serovar 8 (Uu8) and U. parvum serovar 3 (Up3). ACKR3 responses were assessed via qRT-PCR, RNA sequencing, flow cytometry, and immunocytochemistry. RESULTS LPS, TNF-α, and Ureaplasma spp. influenced ACKR3 expression in HBMEC. LPS and TNF-α significantly induced ACKR3 mRNA expression (p < 0.001, vs. control), whereas Ureaplasma spp. enhanced ACKR3 protein expression in HBMEC (p < 0.01, vs. broth control). Co-stimulation with LPS and either Ureaplasma isolate intensified ACKR3 responses (p < 0.05, vs. LPS). Furthermore, stimulation wielded a differential influence on the receptor's ligands. CONCLUSIONS We introduce an in vitro model of Ureaplasma meningitis. We are able to demonstrate a pro-inflammatory capacity of Ureaplasma spp. in native and, even more so, in LPS-primed HBMEC, underlining their clinical relevance particularly in a setting of co-infection. Furthermore, our data may indicate a novel role for ACKR3, with an impact not limited to auto-inflammatory diseases, but extending to infection-related neuroinflammation as well. AKCR3-induced blood-brain barrier breakdown might constitute a potential common pathomechanism.
Collapse
Affiliation(s)
- Christine Silwedel
- University Children's Hospital, University of Wuerzburg, Josef-Schneider-Str. 2, 97080, Wuerzburg, Germany.
| | - Christian P Speer
- University Children's Hospital, University of Wuerzburg, Josef-Schneider-Str. 2, 97080, Wuerzburg, Germany
| | - Axel Haarmann
- Department of Neurology, University of Wuerzburg, Josef-Schneider-Str. 11, 97080, Wuerzburg, Germany
| | - Markus Fehrholz
- University Children's Hospital, University of Wuerzburg, Josef-Schneider-Str. 2, 97080, Wuerzburg, Germany
| | - Heike Claus
- Institute for Hygiene and Microbiology, University of Wuerzburg, Josef-Schneider-Str. 2, 97080, Wuerzburg, Germany
| | - Mathias Buttmann
- Department of Neurology, University of Wuerzburg, Josef-Schneider-Str. 11, 97080, Wuerzburg, Germany.,Department of Neurology, Caritas Hospital, Uhlandstr. 7, 97980, Bad Mergentheim, Germany
| | - Kirsten Glaser
- University Children's Hospital, University of Wuerzburg, Josef-Schneider-Str. 2, 97080, Wuerzburg, Germany
| |
Collapse
|
35
|
Ureaplasma isolates differentially modulate growth factors and cell adhesion molecules in human neonatal and adult monocytes. Cytokine 2018; 105:45-48. [PMID: 29455108 DOI: 10.1016/j.cyto.2018.01.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 01/29/2018] [Accepted: 01/30/2018] [Indexed: 01/15/2023]
Abstract
Generally regarded as commensal bacteria, the pathogenicity of Ureaplasma has often been considered low. Controversy remains concerning the clinical relevance of Ureaplasma infection in the pathogenesis of inflammation-related morbidities. Recently, we demonstrated Ureaplasma-driven pro-inflammatory cytokine responses in human monocytes in vitro. We hypothesized that Ureaplasma may induce further inflammatory mediators. Using qRT-PCR and multi-analyte immunoassay, we assessed the expression of granulocyte-colony stimulating factor (G-CSF), vascular endothelial growth factor (VEGF), intercellular adhesion molecule 1 (ICAM-1) and vascular cell adhesion molecule 1 (VCAM-1) in term neonatal and adult monocytes exposed to Ureaplasma urealyticum serovar 8 (Uu8) and U. parvum serovar 3 (Up3). Ureaplasma significantly induced VEGF mRNA in neonatal (Up3: p < 0.05, versus broth control) and adult monocytes (Uu8: p < 0.05) as well as ICAM-1 mRNA in neonatal cells (p < 0.05 each). As far as protein expression was concerned, Up3 stimulated VEGF release in both monocyte subsets (p < 0.01) and enhanced secretion of ICAM-1 protein in neonatal monocytes (p < 0.05). In adult cells, ICAM-1 protein release was increased upon exposure to both isolates (Uu8: p < 0.05, Up3: p < 0.01). Ureaplasma-induced responses did not significantly differ from corresponding levels mediated by E. coli lipopolysaccharide (LPS). The stimulatory effects were dose-dependent. Ureaplasma infection, on the contrary, did not affect G-CSF and VCAM-1 expression. Of note, co-infection of LPS-primed neonatal monocytes with Ureaplasma enhanced LPS-induced ICAM-1 release (Uu8: p < 0.05). Our results confirm Ureaplasma-driven pro-inflammatory activation of human monocytes in vitro, demonstrating a differential modulation of growth factors and cell adhesion molecules, that might promote unbalanced monocyte responses and adverse immunomodulation.
Collapse
|