1
|
Manuel G, Twentyman J, Noble K, Eastman AJ, Aronoff DM, Seepersaud R, Rajagopal L, Adams Waldorf KM. Group B streptococcal infections in pregnancy and early life. Clin Microbiol Rev 2024:e0015422. [PMID: 39584819 DOI: 10.1128/cmr.00154-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2024] Open
Abstract
SUMMARYBacterial infections with Group B Streptococcus (GBS) are an important cause of adverse outcomes in pregnant individuals, neonates, and infants. GBS is a common commensal in the genitourinary and gastrointestinal tracts and can be detected in the vagina of approximately 20% of women globally. GBS can infect the fetus either during pregnancy or vaginal delivery resulting in preterm birth, stillbirth, or early-onset neonatal disease (EOD) in the first week of life. The mother can also become infected with GBS leading to postpartum endometritis, and rarely, maternal sepsis. An invasive GBS infection of the neonate may present after the first week of life (late-onset disease, LOD) through transmission from caregivers, breast milk, and other sources. Invasive GBS infections in neonates can result in sepsis, pneumonia, meningitis, neurodevelopmental impairment, death, and lifelong disability. A policy of routine screening for GBS rectovaginal colonization in well-resourced countries can trigger the administration of intrapartum antibiotic prophylaxis (IAP) when prenatal testing is positive, which drastically reduces rates of EOD. However, many countries do not routinely screen pregnant women for GBS colonization but may administer IAP in cases with a high risk of EOD. IAP does not reduce rates of LOD. A global vaccination campaign is needed to reduce the significant burden of invasive GBS disease that remains among infants and pregnant individuals. In this narrative review, we provide a comprehensive overview of the global impact of GBS colonization and infection, virulence factors and pathogenesis, and current and future prophylactics and therapeutics.
Collapse
Affiliation(s)
- Gygeria Manuel
- Department of Obstetrics & Gynecology, University of Washington, Seattle, Washington, USA
| | - Joy Twentyman
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Kristen Noble
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Alison J Eastman
- Department of Obstetrics and Gynecology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - David M Aronoff
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Ravin Seepersaud
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Lakshmi Rajagopal
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
- Global Health, University of Washington, Seattle, Washington, USA
| | - Kristina M Adams Waldorf
- Department of Obstetrics & Gynecology, University of Washington, Seattle, Washington, USA
- Global Health, University of Washington, Seattle, Washington, USA
| |
Collapse
|
2
|
Mureanu N, Bowman AM, Porter-Wright IA, Verma P, Efthymiou A, Nicolaides KH, Scotta C, Lombardi G, Tribe RM, Shangaris P. The Immunomodulatory Role of Regulatory T Cells in Preterm Birth and Associated Pregnancy Outcomes. Int J Mol Sci 2024; 25:11878. [PMID: 39595948 PMCID: PMC11593591 DOI: 10.3390/ijms252211878] [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: 10/03/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024] Open
Abstract
Spontaneous preterm birth (sPTB), defined as live birth before 37 weeks of gestational age, is associated with immune dysregulation and pro-inflammatory conditions that profoundly impact newborn health. The question of immune integrity at the maternal-foetal interface is a focus of recent studies centring not only sPTB but the conditions often affiliated with this outcome. Regulatory T cells (Tregs) play a critical anti-inflammatory role in pregnancy, promoting foetal tolerance and placentation. Due to this gestational role, it is hypothesised that decreased or dysfunctional Tregs may be implicated in cases of sPTB. This review examines studies comparing Treg presence in healthy term pregnancies and those with sPTB-associated conditions. Conflicting findings across different conditions and within sPTB itself have been identified. However, notable findings from the research indicate increased proinflammatory cytokines in pregnancies suffering from premature rupture of membranes (pPROM), chorioamnionitis, infection, preeclampsia, and gestational diabetes (GDM). Additionally, reduced Treg levels were identified in preeclampsia, GDM, and pPROM as well as chorioamnionitis presenting with increased Treg dysfunctionality. Treg deficiencies may contribute to health issues in preterm newborns. Current sPTB treatments are limited, underscoring the potential of in utero therapies targeting inflammation, including T cell interventions. Future research aims to establish consensus on the role of Tregs in sPTB and associated conditions and advancing understanding of mechanisms leading to Treg deficiencies in adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Nicoleta Mureanu
- School of Life Course & Population Sciences, King’s College London, 10th Floor North Wing, St Thomas’ Hospital, London SE1 7EH, UK; (N.M.); (A.M.B.); (A.E.); (K.H.N.); (R.M.T.)
- Harris Birthright Research Centre for Fetal Medicine, King’s College London, London SE1 7EH, UK
- Faculty of Medicine, Department of Obstetrics and Gynaecology, Carol Davila University of Medicine and Pharmacy, Bulevardul Eroii Sanitari 8, 050474 Bucharest, Romania
| | - Amanda M. Bowman
- School of Life Course & Population Sciences, King’s College London, 10th Floor North Wing, St Thomas’ Hospital, London SE1 7EH, UK; (N.M.); (A.M.B.); (A.E.); (K.H.N.); (R.M.T.)
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE1 7EH, UK; (I.A.P.-W.); (P.V.); (C.S.); (G.L.)
| | - Imogen A. Porter-Wright
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE1 7EH, UK; (I.A.P.-W.); (P.V.); (C.S.); (G.L.)
| | - Priya Verma
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE1 7EH, UK; (I.A.P.-W.); (P.V.); (C.S.); (G.L.)
| | - Athina Efthymiou
- School of Life Course & Population Sciences, King’s College London, 10th Floor North Wing, St Thomas’ Hospital, London SE1 7EH, UK; (N.M.); (A.M.B.); (A.E.); (K.H.N.); (R.M.T.)
- Harris Birthright Research Centre for Fetal Medicine, King’s College London, London SE1 7EH, UK
| | - Kypros H. Nicolaides
- School of Life Course & Population Sciences, King’s College London, 10th Floor North Wing, St Thomas’ Hospital, London SE1 7EH, UK; (N.M.); (A.M.B.); (A.E.); (K.H.N.); (R.M.T.)
- Harris Birthright Research Centre for Fetal Medicine, King’s College London, London SE1 7EH, UK
| | - Cristiano Scotta
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE1 7EH, UK; (I.A.P.-W.); (P.V.); (C.S.); (G.L.)
- Department of Life Sciences, Centre for Inflammation Research and Translational Medicine, Brunel University London, London UB8 3PH, UK
| | - Giovanna Lombardi
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE1 7EH, UK; (I.A.P.-W.); (P.V.); (C.S.); (G.L.)
| | - Rachel M. Tribe
- School of Life Course & Population Sciences, King’s College London, 10th Floor North Wing, St Thomas’ Hospital, London SE1 7EH, UK; (N.M.); (A.M.B.); (A.E.); (K.H.N.); (R.M.T.)
| | - Panicos Shangaris
- School of Life Course & Population Sciences, King’s College London, 10th Floor North Wing, St Thomas’ Hospital, London SE1 7EH, UK; (N.M.); (A.M.B.); (A.E.); (K.H.N.); (R.M.T.)
- Harris Birthright Research Centre for Fetal Medicine, King’s College London, London SE1 7EH, UK
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE1 7EH, UK; (I.A.P.-W.); (P.V.); (C.S.); (G.L.)
| |
Collapse
|
3
|
Tripathy S, Burd I, Kelleher MA. Membrane inflammasome activation by choriodecidual Ureaplasma parvum infection without intra-amniotic infection in a Non-Human Primate model†. Biol Reprod 2024; 110:971-984. [PMID: 38335245 PMCID: PMC11094395 DOI: 10.1093/biolre/ioae027] [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: 09/27/2023] [Revised: 12/29/2023] [Accepted: 02/02/2024] [Indexed: 02/12/2024] Open
Abstract
Intrauterine infection is a significant cause of neonatal morbidity and mortality. Ureaplasma parvum is a microorganism commonly isolated from cases of preterm birth and preterm premature rupture of membranes (pPROM). However, the mechanisms of early stage ascending reproductive tract infection remain poorly understood. To examine inflammation in fetal (chorioamnionic) membranes we utilized a non-human primate (NHP) model of choriodecidual U. parvum infection. Eight chronically catheterized pregnant rhesus macaques underwent maternal-fetal catheterization surgery at ~105-112 days gestation and choriodecidual inoculation with U. parvum (105 CFU/mL, n =4) or sterile media (controls; n = 4) starting at 115-119 days, repeated at 5-day intervals until C-section at 136-140 days (term=167 days). The average inoculation to delivery interval was 21 days, and Ureaplasma infection of the amniotic fluid (AF) was undetectable in all animals. Choriodecidual Ureaplasma infection resulted in increased fetal membrane expression of MMP-9 and PTGS2, but did not result in preterm labor or increased concentrations of AF pro-inflammatory cytokines. However, membrane expression of inflammasome sensors, NLRP3, NLRC4, AIM2, and NOD2, and adaptor ASC (PYCARD) gene expression were significantly increased. Gene expression of IL-1β, IL-18, IL-18R1 , CASPASE-1, and pro-CASPASE-1 protein increased with Ureaplasma infection. Downstream inflammatory genes MYD88 and NFκB (Nuclear factor kappa-light-chain-enhancer of activated B cells) were also significantly upregulated. These results demonstrate that choriodecidual Ureaplasma infection, can cause activation of inflammasome complexes and pathways associated with pPROM and preterm labor prior to microbes being detectable in the AF.
Collapse
Affiliation(s)
- Sudeshna Tripathy
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
| | - Irina Burd
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Meredith A Kelleher
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
| |
Collapse
|
4
|
Tripathy S, Burd I, Kelleher MA. Membrane Inflammasome Activation by Choriodecidual Ureaplasma parvum Infection without Intra-Amniotic Infection in an NHP Model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.18.557989. [PMID: 37781578 PMCID: PMC10541100 DOI: 10.1101/2023.09.18.557989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Intrauterine infection is a significant cause of preterm labor and neonatal morbidity and mortality. Ureaplasma parvum is the micro-organism most commonly isolated from cases of preterm birth and preterm premature rupture of membranes (pPROM). However, the mechanisms during the early stages of ascending reproductive tract infection that initiate maternal-fetal inflammatory pathways, preterm birth and pPROM remain poorly understood. To examine inflammation in fetal (chorioamnionic) membranes in response to Ureaplasma parvum infection, we utilized a novel in vivo non-human primate model of early choriodecidual infection. Eight chronically catheterized pregnant rhesus macaques underwent maternal-fetal catheterization surgery at 105-112 days gestation and choriodecidual inoculation with Ureaplasma parvum (10 5 cfu/mL of a low passaged clinical isolate, serovar 1; n=4) or saline/sterile media (Controls; n=4) starting at 115-119 days gestation, repeated every 5 days until scheduled cesarean-section at 136-140d gestation (term=167d). The average inoculation to delivery interval was 21 days and Ureaplasma infection of the amniotic fluid was undetectable by culture and PCR in all animals. Inflammatory mediators in amniotic fluid (AF) were assessed by Luminex, ELISA and multiplex assays. RNA was extracted from the chorion and amnionic membranes for single gene analysis (qRT-PCR) and protein expression was determined by Western blot and immunohistochemistry. Our NHP model of choriodecidual Ureaplasma infection, representing an early-stage ascending reproductive tract infection without microbial invasion of the amniotic cavity, resulted in increased fetal membrane protein and gene expression of MMP-9 and PTGS2, but did not result in preterm labor (no increase in uterine contractility) or increased concentrations of amniotic fluid pro-inflammatory cytokines (IL-1β, IL-6, IL-8, IL-18, TNF-α). However, membrane expression of inflammasome sensor molecules, NLRP3, NLRC4, AIM2 and NOD2, and the adaptor protein ASC ( PYCARD ) gene expression were significantly increased in the Ureaplasma group when compared to non-infected controls. Gene expression of IL-1 β, IL-18, the IL-18R1 receptor , CASPASE-1 and pro-CASPASE-1 protein were also increased in the fetal membranes with Ureaplasma infection. Downstream inflammatory signaling genes MYD88 was also significantly upregulated in both the amnion and chorion, along with a significant increase in NFKB in the chorion. These results demonstrate that even at the early stages of ascending reproductive tract Ureaplasma infection, activation of inflammasome complexes and pathways associated with degradation of chorioamnionic membrane integrity are present. This study therefore provides experimental evidence for the importance of the early stages of ascending Ureaplasma infection in initiating processes of pPROM and preterm labor. These findings have implications for the identification of intrauterine inflammation before microbes are detectable in the amniotic fluid (sterile inflammation) and the timing of potential treatments for preterm labor and fetal injury caused by intrauterine infection.
Collapse
|
5
|
Kılınçdemir Turgut Ü, Tola EN, Sezik M. The association between preterm delivery and postpartum bleeding in otherwise uncomplicated pregnancies. J Turk Ger Gynecol Assoc 2022; 23:177-183. [PMID: 35781761 PMCID: PMC9450917 DOI: 10.4274/jtgga.galenos.2022.2021-11-7] [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: 11/24/2021] [Accepted: 05/20/2022] [Indexed: 12/01/2022] Open
Abstract
Objective The primary aim was to investigate whether preterm delivery was an independent risk factor for blood or blood products transfusion in the intrapartum or postpartum period, considered as a proxy for severe obstetric bleeding. Material and Methods Throughout a 9-month-period, 216 uncomplicated singleton deliveries were included in a cross-sectional study after exclusion of severe maternal and fetal morbidity, such as chorioamnionitis, and use of medications including tocolytics. Maternal and neonatal data were evaluated and compared across preterm (between 24 0/7-36 6/7 weeks’ gestation) and term (between 37 0/7-41 6/7 weeks’ gestation) deliveries. Primary and secondary outcomes were requirement for blood or blood products transfusion until discharge and change in hemoglobin value and hematocrit from baseline to postpartum hour 6, respectively. Logistic regression models were constructed to evaluate the effect of preterm delivery on the primary outcome. Results There were 90 (41.7%) preterm deliveries with an overall cesarean section rate of 77.8%. Preterm delivery was not an independent risk factor for the primary outcome, when route of delivery, maternal body-mass index, antenatal steroid administration, and baseline (admission) platelet and leukocyte counts were controlled for [adjusted risk ratio, 2.46; 95% confidence interval (CI), 0.69-8.77; p=0.16]. Subgroup analysis, including cesarean deliveries, revealed a similar result (adjusted risk ratio, 1.65; 95% CI, 0.42-6.48; p=0.47). Secondary outcomes, including decrease in mean or percent values of hemoglobin and hematocrit measurements, were also similar across preterm and term groups, both after vaginal and cesarean delivery (for all comparisons, p>0.05). Conclusion Preterm delivery is not independently associated with increased requirement for blood transfusions or decreased hemoglobin and hematocrit values following otherwise uncomplicated vaginal or cesarean delivery of singletons.
Collapse
Affiliation(s)
- Ümran Kılınçdemir Turgut
- Department of Obstetrics and Gynecology, Süleyman Demirel University Faculty of Medicine, Isparta, Turkey
| | - Esra Nur Tola
- Department of Obstetrics and Gynecology, İstanbul Medipol University Faculty of Medicine, İstanbul, Turkey
| | - Mekin Sezik
- Department of Obstetrics and Gynecology, Süleyman Demirel University Faculty of Medicine, Isparta, Turkey
| |
Collapse
|
6
|
Olmos-Ortiz A, Hernández-Pérez M, Flores-Espinosa P, Sedano G, Helguera-Repetto AC, Villavicencio-Carrisoza Ó, Valdespino-Vazquez MY, Flores-Pliego A, Irles C, Rivas-Santiago B, Moreno-Verduzco ER, Díaz L, Zaga-Clavellina V. Compartmentalized Innate Immune Response of Human Fetal Membranes against Escherichia coli Choriodecidual Infection. Int J Mol Sci 2022; 23:ijms23062994. [PMID: 35328414 PMCID: PMC8949057 DOI: 10.3390/ijms23062994] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/21/2022] [Accepted: 03/02/2022] [Indexed: 12/29/2022] Open
Abstract
An infectious process into the uterine cavity represents a major endangered condition that compromises the immune privilege of the maternal-fetal unit and increases the risk for preterm birth (PTB) and premature rupture of membranes (PROM). Fetal membranes are active secretors of antimicrobial peptides (AMP), which limit bacterial growth, such as Escherichia coli. Nevertheless, the antibacterial responses displayed by chorioamniotic membranes against a choriodecidual E. coli infection have been briefly studied. The objective of this research was to characterize the profile of synthesis, activity, and spatial distribution of a broad panel of AMPs produced by fetal membranes in response to E. coli choriodecidual infection. Term human chorioamniotic membranes were mounted in a two independent compartment model in which the choriodecidual region was infected with live E. coli (1 × 105 CFU/mL). Amnion and choriodecidual AMP tissue levels and TNF-α and IL-1β secretion were measured by the enzyme-linked immunosorbent assay. The passage of bacterium through fetal membranes and their effect on structural continuity was followed for 24 h. Our results showed that E. coli infection caused a progressive mechanical disruption of the chorioamniotic membranes and an activated inflammatory environment. After the challenge, the amnion quickly (2-4 h) induced production of human beta defensins (HBD)-1, HBD-2, and LL-37. Afterwards (8-24 h), the amnion significantly produced HBD-1, HBD-2, HNP-1-3, S100A7, sPLA2, and elafin, whereas the choriodecidua induced LL-37 synthesis. Therefore, we noticed a temporal- and tissue-specific pattern regulation of the synthesis of AMPs by infected fetal membranes. However, fetal membranes were not able to contain the collagen degradation or the bacterial growth and migration despite the battery of produced AMPs, which deeply increases the risk for PTB and PROM. The mixture of recombinant HBDs at low concentrations resulted in increased bactericidal activity compared to each HBD alone in vitro, encouraging further research to study AMP combinations that may offer synergy to control drug-resistant infections in the perinatal period.
Collapse
Affiliation(s)
- Andrea Olmos-Ortiz
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología (INPer), Mexico City 11000, Mexico; (A.O.-O.); (M.H.-P.); (P.F.-E.); (G.S.); (A.C.H.-R.); (Ó.V.-C.); (A.F.-P.)
| | - Mayra Hernández-Pérez
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología (INPer), Mexico City 11000, Mexico; (A.O.-O.); (M.H.-P.); (P.F.-E.); (G.S.); (A.C.H.-R.); (Ó.V.-C.); (A.F.-P.)
| | - Pilar Flores-Espinosa
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología (INPer), Mexico City 11000, Mexico; (A.O.-O.); (M.H.-P.); (P.F.-E.); (G.S.); (A.C.H.-R.); (Ó.V.-C.); (A.F.-P.)
| | - Gabriela Sedano
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología (INPer), Mexico City 11000, Mexico; (A.O.-O.); (M.H.-P.); (P.F.-E.); (G.S.); (A.C.H.-R.); (Ó.V.-C.); (A.F.-P.)
| | - Addy Cecilia Helguera-Repetto
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología (INPer), Mexico City 11000, Mexico; (A.O.-O.); (M.H.-P.); (P.F.-E.); (G.S.); (A.C.H.-R.); (Ó.V.-C.); (A.F.-P.)
| | - Óscar Villavicencio-Carrisoza
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología (INPer), Mexico City 11000, Mexico; (A.O.-O.); (M.H.-P.); (P.F.-E.); (G.S.); (A.C.H.-R.); (Ó.V.-C.); (A.F.-P.)
| | | | - Arturo Flores-Pliego
- Departamento de Inmunobioquímica, Instituto Nacional de Perinatología (INPer), Mexico City 11000, Mexico; (A.O.-O.); (M.H.-P.); (P.F.-E.); (G.S.); (A.C.H.-R.); (Ó.V.-C.); (A.F.-P.)
| | - Claudine Irles
- Departamento de Fisiología y Desarrollo Celular, INPer, Mexico City 11000, Mexico;
| | | | | | - Lorenza Díaz
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico;
| | - Verónica Zaga-Clavellina
- Departamento de Fisiología y Desarrollo Celular, INPer, Mexico City 11000, Mexico;
- Correspondence: ; Tel.: +52-55-5520-9900 (ext. 478)
| |
Collapse
|
7
|
Menon R, Dixon CL, Cayne S, Radnaa E, Salomon C, Sheller-Miller S. Differences in cord blood extracellular vesicle cargo in preterm and term births. Am J Reprod Immunol 2022; 87:e13521. [PMID: 35007379 DOI: 10.1111/aji.13521] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 12/14/2021] [Accepted: 12/29/2021] [Indexed: 11/28/2022] Open
Abstract
OBJECTIVE This study determined the cord plasma-derived extracellular vesicle (exosomes; 30-160 nm particles) proteomic profile in patients who had spontaneous preterm birth (PTB) or preterm premature rupture of membranes (pPROM), compared to those who delivered at term regardless of labor status. METHODS This is a cross-sectional analysis of a retrospective cohort that quantified and determined the proteomic cargo content of exosomes present in cord blood plasma samples in PTB or pPROM, and normal term in labor (TL) or term not in labor (TNIL) pregnancies. Exosomes were isolated by differential centrifugation followed by size exclusion chromatography. Exosomes were characterized by nanoparticle tracking analysis (quantity and size) and markers (dot blots for exosome markers). The exosomal proteomic profile was identified by liquid chromatography-mass spectrometry (LC-MS/MS). Ingenuity pathway analysis determined canonical pathways and biofunctions associated with dysregulated proteins. RESULTS Cord plasma exosomes have similar quantity and exhibit both tetraspanin and ESCRT protein markers specific of exosomes regardless of the conditions. Proteomics analysis exhibited several similar markers as well as very unique markers in exosomes from each condition; however, bioinformatics analysis revealed a generalized and non-specific inflammatory condition represented in exosomes from different condition that is not indicative of any specific underlying biological functions indicative of an underlying pathology. CONCLUSIONS Compared to maternal plasma and amniotic fluid exosomes, the value of cord plasma derived exosomes is limited. Quantity, character, and proteomic cargo contents in exosomes or the pathways and functions represented by differentially expressed proteins do not distinguish specific conditions regarding normal and abnormal parturition. The value of cord plasma exosome proteomic cargo has limited value as an indicator of an underlying physiology or as a biomarker of fetal well-being.
Collapse
Affiliation(s)
- Ramkumar Menon
- Division of Basic and Translation Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Christopher Luke Dixon
- Division of Basic and Translation Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Samir Cayne
- Division of Basic and Translation Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Enkhtuya Radnaa
- Division of Basic and Translation Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Carlos Salomon
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital, The University of Queensland, Brisbane, Australia.,Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepción, Concepción, Chile
| | - Samantha Sheller-Miller
- Division of Basic and Translation Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| |
Collapse
|
8
|
Shynlova O, Boros-Rausch A, Farine T, Adams Waldorf KM, Dunk C, Lye SJ. Decidual Inflammation Drives Chemokine-Mediated Immune Infiltration Contributing to Term Labor. THE JOURNAL OF IMMUNOLOGY 2021; 207:2015-2026. [PMID: 34526377 DOI: 10.4049/jimmunol.2100493] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 07/30/2021] [Indexed: 01/14/2023]
Abstract
Infiltration of maternal peripheral leukocytes into the uterine tissues is a critical event occurring before, during, and after term labor (TL). In this article, we investigate the contribution of uterine smooth muscle (myometrium) and pregnant endometrium (decidua) to the inflammatory process during human TL. We hypothesize that labor-related physiological inflammation is orchestrated by uterine-secreted cytokines, which dually activate the uterine vascular endothelium and maternal leukocytes to promote their adhesion and infiltration into the uterus. Using Luminex and ELISA assays, we examine a full range of cytokines (45 proteins) in media conditioned by primary decidual and myometrial cells from TL and term not in labor (TNL) women. The effect of conditioned media on the activation of human uterine microvascular endothelial cells was measured by qPCR and on peripheral leukocytes by flow cytometry. Transendothelial migration of calcein-labeled primary leukocytes toward media was assessed by fluorometry. Stromal decidual cells secrete significantly higher levels of multiple cytokines compared with myometrial cells (p < 0.05) and significantly more cytokines during TL than TNL. These cytokines activate uterine microvascular endothelial cells through the upregulation of cell adhesion molecule VCAM-1 and peripheral leukocytes by upregulation of CD11b. Furthermore, multiple cytokines secreted from the TL decidua and myometrium significantly increase migration of granulocytes, monocytes, and lymphocytes compared with TNL (p < 0.05), which was blocked by a broad-spectrum chemokine inhibitor (FX125L). These data reveal the critical role for decidual- and myometrial-secreted cytokines in the activation of inflammatory pathways leading to labor. We suggest that these pathways represent targets for therapeutic intervention during preterm labor.
Collapse
Affiliation(s)
- Oksana Shynlova
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada; .,Department of Physiology, University of Toronto, Ontario, Canada.,Department of Obstetrics and Gynecology, University of Toronto, Ontario, Canada; and
| | - Adam Boros-Rausch
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.,Department of Physiology, University of Toronto, Ontario, Canada
| | - Tali Farine
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.,Department of Physiology, University of Toronto, Ontario, Canada
| | | | - Caroline Dunk
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Stephen J Lye
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.,Department of Physiology, University of Toronto, Ontario, Canada.,Department of Obstetrics and Gynecology, University of Toronto, Ontario, Canada; and
| |
Collapse
|
9
|
Cappelletti M, Presicce P, Feiyang M, Senthamaraikannan P, Miller LA, Pellegrini M, Sim MS, Jobe AH, Divanovic S, Way SS, Chougnet CA, Kallapur SG. The induction of preterm labor in rhesus macaques is determined by the strength of immune response to intrauterine infection. PLoS Biol 2021; 19:e3001385. [PMID: 34495952 PMCID: PMC8452070 DOI: 10.1371/journal.pbio.3001385] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 09/20/2021] [Accepted: 08/04/2021] [Indexed: 12/18/2022] Open
Abstract
Intrauterine infection/inflammation (IUI) is a major contributor to preterm labor (PTL). However, IUI does not invariably cause PTL. We hypothesized that quantitative and qualitative differences in immune response exist in subjects with or without PTL. To define the triggers for PTL, we developed rhesus macaque models of IUI driven by lipopolysaccharide (LPS) or live Escherichia coli. PTL did not occur in LPS challenged rhesus macaques, while E. coli–infected animals frequently delivered preterm. Although LPS and live E. coli both caused immune cell infiltration, E. coli–infected animals showed higher levels of inflammatory mediators, particularly interleukin 6 (IL-6) and prostaglandins, in the chorioamnion-decidua and amniotic fluid (AF). Neutrophil infiltration in the chorio-decidua was a common feature to both LPS and E. coli. However, neutrophilic infiltration and IL6 and PTGS2 expression in the amnion was specifically induced by live E. coli. RNA sequencing (RNA-seq) analysis of fetal membranes revealed that specific pathways involved in augmentation of inflammation including type I interferon (IFN) response, chemotaxis, sumoylation, and iron homeostasis were up-regulated in the E. coli group compared to the LPS group. Our data suggest that the intensity of the host immune response to IUI may determine susceptibility to PTL.
Collapse
Affiliation(s)
- Monica Cappelletti
- Divisions of Neonatology and Developmental Biology, UCLA Mattel Children’s Hospital, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Pietro Presicce
- Divisions of Neonatology and Developmental Biology, UCLA Mattel Children’s Hospital, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Ma Feiyang
- Department of Molecular, Cell and Developmental Biology Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
- Institute for Quantitative and Computational Biosciences–Collaboratory, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Paranthaman Senthamaraikannan
- Division of Neonatology and Pulmonary Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Lisa A. Miller
- California National Primate Research Center, University of California Davis, Davis, California, United States of America
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
- Institute for Quantitative and Computational Biosciences–Collaboratory, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Myung S. Sim
- Department of Medicine Statistics Core, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Alan H. Jobe
- Division of Neonatology and Pulmonary Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Senad Divanovic
- Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Sing Sing Way
- Infectious Diseases, Cincinnati Children’s Hospital Research Foundation, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Claire A. Chougnet
- Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Suhas G. Kallapur
- Divisions of Neonatology and Developmental Biology, UCLA Mattel Children’s Hospital, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
10
|
Lee JH, Park CW, Moon KC, Park JS, Jun JK. The Inflammatory Milieu of Amniotic Fluid Increases with Chorio-Deciduitis Grade in Inflammation-Restricted to Choriodecidua, but Not Amnionitis, of Extra-Placental Membranes. J Clin Med 2021; 10:3041. [PMID: 34300208 PMCID: PMC8307834 DOI: 10.3390/jcm10143041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/04/2021] [Accepted: 07/06/2021] [Indexed: 12/27/2022] Open
Abstract
No information exists about whether intra-amniotic inflammatory response increases with a chorio-deciduitis grade in the context of both inflammation-restricted to chorio-decidua and amnionitis of extra-placental membranes among spontaneous preterm births. The objective of current study is to examine this issue. A study population included 195 singleton pregnant women with chorio-deciduitis, and who spontaneously delivered at preterm (21.6~35.7 weeks) within 7 days of amniocentesis. We examined intra-amniotic inflammatory response according to the chorio-deciduitis grade in the context of inflammation restricted to chorio-decidua and amnionitis of extra-placental membranes. Intra-amniotic inflammatory response was measured by MMP-8 concentration (ng/mL) and WBC-count (cells/mm3) in amniotic-fluid (AF). Inflammation restricted to chorio-decidua and amnionitis were present in 47.7% (93/195) and 52.3% (102/195) of cases, respectively. Median AF MMP-8 concentration and WBC-count significantly increased with chorio-deciduitis grade in the context of inflammation restricted to chorio-decidua. However, there was no significant difference in median AF MMP-8 concentration and WBC-count between chorio-deciduitis grade-1 and grade-2 in the context of amnionitis. The inflammatory milieu of AF increases with chorio-deciduitis grade in inflammation-restricted to chorio-decidua, but not amnionitis, of extra-placental membranes. This finding suggests that a chorio-deciduitis grade may have little effect on the intensification of intra-amniotic inflammatory response in the context of amnionitis of extra-placental membranes.
Collapse
Affiliation(s)
- Joon Hyung Lee
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul 03080, Korea; (J.H.L.); (J.S.P.); (J.K.J.)
| | - Chan-Wook Park
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul 03080, Korea; (J.H.L.); (J.S.P.); (J.K.J.)
- Medical Research Center, Institute of Reproductive Medicine and Population, Seoul National University, Seoul 03080, Korea
| | - Kyung Chul Moon
- Department of Pathology, Seoul National University College of Medicine, Seoul 03080, Korea;
| | - Joong Shin Park
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul 03080, Korea; (J.H.L.); (J.S.P.); (J.K.J.)
| | - Jong Kwan Jun
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul 03080, Korea; (J.H.L.); (J.S.P.); (J.K.J.)
- Medical Research Center, Institute of Reproductive Medicine and Population, Seoul National University, Seoul 03080, Korea
| |
Collapse
|
11
|
Li M, Brokaw A, Furuta AM, Coler B, Obregon-Perko V, Chahroudi A, Wang HY, Permar SR, Hotchkiss CE, Golos TG, Rajagopal L, Adams Waldorf KM. Non-human Primate Models to Investigate Mechanisms of Infection-Associated Fetal and Pediatric Injury, Teratogenesis and Stillbirth. Front Genet 2021; 12:680342. [PMID: 34290739 PMCID: PMC8287178 DOI: 10.3389/fgene.2021.680342] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 05/25/2021] [Indexed: 12/25/2022] Open
Abstract
A wide array of pathogens has the potential to injure the fetus and induce teratogenesis, the process by which mutations in fetal somatic cells lead to congenital malformations. Rubella virus was the first infectious disease to be linked to congenital malformations due to an infection in pregnancy, which can include congenital cataracts, microcephaly, hearing impairment and congenital heart disease. Currently, human cytomegalovirus (HCMV) is the leading infectious cause of congenital malformations globally, affecting 1 in every 200 infants. However, our knowledge of teratogenic viruses and pathogens is far from complete. New emerging infectious diseases may induce teratogenesis, similar to Zika virus (ZIKV) that caused a global pandemic in 2016-2017; thousands of neonates were born with congenital microcephaly due to ZIKV exposure in utero, which also included a spectrum of injuries to the brain, eyes and spinal cord. In addition to congenital anomalies, permanent injury to fetal and neonatal organs, preterm birth, stillbirth and spontaneous abortion are known consequences of a broader group of infectious diseases including group B streptococcus (GBS), Listeria monocytogenes, Influenza A virus (IAV), and Human Immunodeficiency Virus (HIV). Animal models are crucial for determining the mechanism of how these various infectious diseases induce teratogenesis or organ injury, as well as testing novel therapeutics for fetal or neonatal protection. Other mammalian models differ in many respects from human pregnancy including placentation, labor physiology, reproductive tract anatomy, timeline of fetal development and reproductive toxicology. In contrast, non-human primates (NHP) most closely resemble human pregnancy and exhibit key similarities that make them ideal for research to discover the mechanisms of injury and for testing vaccines and therapeutics to prevent teratogenesis, fetal and neonatal injury and adverse pregnancy outcomes (e.g., stillbirth or spontaneous abortion). In this review, we emphasize key contributions of the NHP model pre-clinical research for ZIKV, HCMV, HIV, IAV, L. monocytogenes, Ureaplasma species, and GBS. This work represents the foundation for development and testing of preventative and therapeutic strategies to inhibit infectious injury of human fetuses and neonates.
Collapse
Affiliation(s)
- Miranda Li
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA, United States
- Department of Biological Sciences, Columbia University, New York, NY, United States
| | - Alyssa Brokaw
- Department of Global Health, University of Washington, Seattle, WA, United States
| | - Anna M. Furuta
- Department of Global Health, University of Washington, Seattle, WA, United States
| | - Brahm Coler
- Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Veronica Obregon-Perko
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Ann Chahroudi
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
- Center for Childhood Infections and Vaccines of Children’s Healthcare of Atlanta and Emory University, Atlanta, GA, United States
| | - Hsuan-Yuan Wang
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, United States
| | - Sallie R. Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, United States
| | - Charlotte E. Hotchkiss
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States
| | - Thaddeus G. Golos
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, United States
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, United States
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Lakshmi Rajagopal
- Department of Global Health, University of Washington, Seattle, WA, United States
- Department of Pediatrics, University of Washington, Seattle, WA, United States
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Kristina M. Adams Waldorf
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA, United States
- Department of Global Health, University of Washington, Seattle, WA, United States
- Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
12
|
McCartney SA, Kapur R, Liggitt HD, Baldessari A, Coleman M, Orvis A, Ogle J, Katz R, Rajagopal L, Adams Waldorf KM. Amniotic fluid interleukin 6 and interleukin 8 are superior predictors of fetal lung injury compared with maternal or fetal plasma cytokines or placental histopathology in a nonhuman primate model. Am J Obstet Gynecol 2021; 225:89.e1-89.e16. [PMID: 33412130 DOI: 10.1016/j.ajog.2020.12.1214] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/20/2020] [Accepted: 12/30/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Intra-amniotic infection or inflammation is common in early preterm birth and associated with substantial neonatal lung morbidity owing to fetal exposure to proinflammatory cytokines and infectious organisms. Amniotic fluid interleukin 8, a proinflammatory cytokine, was previously correlated with the development of neonatal bronchopulmonary dysplasia, but whether amniotic fluid cytokines or placental pathology more accurately predicts neonatal lung pathology and morbidity is unknown. We have used a pregnant nonhuman primate model of group B Streptococcus infection to study the pathogenesis of intra-amniotic infection, bacterial invasion of the amniotic cavity and fetus, and microbial-host interactions. In this nonhuman primate model, we have studied the pathogenesis of group B Streptococcus strains with differing potential for virulence, which has resulted in a spectrum of intra-amniotic infection and fetal lung injury that affords the opportunity to study the inflammatory predictors of fetal lung pathology and injury. OBJECTIVE This study aimed to determine whether fetal lung injury is best predicted by placental histopathology or the cytokine response in amniotic fluid or maternal plasma. STUDY DESIGN Chronically catheterized pregnant monkeys (Macaca nemestrina, pigtail macaque) at 116 to 125 days gestation (term at 172 days) received a choriodecidual inoculation of saline (n=5), weakly hemolytic group B Streptococcus strain (n=5, low virulence), or hyperhemolytic group B Streptococcus strain (n=5, high virulence). Adverse pregnancy outcomes were defined as either preterm labor, microbial invasion of the amniotic cavity, or development of the fetal inflammatory response syndrome. Amniotic fluid and maternal and fetal plasma samples were collected after inoculation, and proinflammatory cytokines (tumor necrosis factor alpha, interleukin beta, interleukin 6, interleukin 8) were measured by a multiplex assay. Cesarean delivery was performed at the time of preterm labor or within 1 week of inoculation. Fetal necropsy was performed at the time of delivery. Placental pathology was scored in a blinded fashion by a pediatric pathologist, and fetal lung injury was determined by a semiquantitative score from histopathology evaluating inflammatory infiltrate, necrosis, tissue thickening, or collapse scored by a veterinary pathologist. RESULTS The principal findings in our study are as follows: (1) adverse pregnancy outcomes occurred more frequently in animals receiving hyperhemolytic group B Streptococcus (80% with preterm labor, 80% with fetal inflammatory response syndrome) than in animals receiving weakly hemolytic group B Streptococcus (40% with preterm labor, 20% with fetal inflammatory response syndrome) and in controls (0% preterm labor, 0% fetal inflammatory response syndrome); (2) despite differences in the rate of adverse pregnancy outcomes and fetal inflammatory response syndrome, fetal lung injury scores were similar between animals receiving the weakly hemolytic group B Streptococcus strains and animals receiving the hyperhemolytic group B Streptococcus strains; (3) fetal lung injury score was significantly correlated with peak amniotic fluid cytokines interleukin 6 and interleukin 8 but not tumor necrosis factor alpha or interleukin 1 beta; and (4) fetal lung scores were poorly correlated with maternal and fetal plasma cytokine levels and placental pathology. CONCLUSION Amniotic fluid interleukin 6 and interleukin 8 levels were superior predictors of fetal lung injury than placental histopathology or maternal plasma cytokines. This evidence supports a role for amniocentesis in the prediction of neonatal lung morbidity owing to intra-amniotic infection, which cannot be provided by cytokine analysis of maternal plasma or placental histopathology.
Collapse
Affiliation(s)
- Stephen A McCartney
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA
| | - Raj Kapur
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle Children's Hospital, Seattle, WA
| | - H Denny Liggitt
- Department of Comparative Medicine, University of Washington, Seattle, WA
| | - Audrey Baldessari
- Washington National Primate Research Center, University of Washington, Seattle, WA
| | - Michelle Coleman
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA
| | - Austyn Orvis
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA
| | - Jason Ogle
- Washington National Primate Research Center, University of Washington, Seattle, WA
| | - Ronit Katz
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA
| | - Lakshmi Rajagopal
- Department of Pediatrics, Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA; Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA
| | - Kristina M Adams Waldorf
- Department of Obstetrics and Gynecology and Global Health, University of Washington, Seattle, WA; Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA; Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA; Sahlgrenska Academy, University of Gothenburg, Sweden.
| |
Collapse
|
13
|
Coleman M, Orvis A, Wu TY, Dacanay M, Merillat S, Ogle J, Baldessari A, Kretzer NM, Munson J, Boros-Rausch AJ, Shynlova O, Lye S, Rajagopal L, Adams Waldorf KM. A Broad Spectrum Chemokine Inhibitor Prevents Preterm Labor but Not Microbial Invasion of the Amniotic Cavity or Neonatal Morbidity in a Non-human Primate Model. Front Immunol 2020; 11:770. [PMID: 32425945 PMCID: PMC7203489 DOI: 10.3389/fimmu.2020.00770] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 04/06/2020] [Indexed: 12/11/2022] Open
Abstract
Leukocyte activation within the chorioamniotic membranes is strongly associated with inflammation and preterm labor (PTL). We hypothesized that prophylaxis with a broad-spectrum chemokine inhibitor (BSCI) would downregulate the inflammatory microenvironment induced by Group B Streptococcus (GBS, Streptococcus agalactiae) to suppress PTL and microbial invasion of the amniotic cavity (MIAC). To correlate BSCI administration with PTL and MIAC, we used a unique chronically catheterized non-human primate model of Group B Streptococcus (GBS)-induced PTL. In the early third trimester (128–138 days gestation; ~29–32 weeks human pregnancy), animals received choriodecidual inoculations of either: (1) saline (N = 6), (2) GBS, 1–5 × 108 colony forming units (CFU)/ml; N = 5), or (3) pre-treatment and daily infusions of a BSCI (10 mg/kg intravenous and intra-amniotic) with GBS (1–5 × 108 CFU/ml; N = 4). We measured amniotic cavity pressure (uterine contraction strength) and sampled amniotic fluid (AF) and maternal blood serially and cord blood at delivery. Cesarean section was performed 3 days post-inoculation or earlier for PTL. Data analysis used Fisher's exact test, Wilcoxon rank sum and one-way ANOVA with Bonferroni correction. Saline inoculation did not induce PTL or infectious sequelae. In contrast, GBS inoculation typically induced PTL (4/5, 80%), MIAC and fetal bacteremia (3/5; 60%). Remarkably, PTL did not occur in the BSCI+GBS group (0/4, 0%; p = 0.02 vs. GBS), despite MIAC and fetal bacteremia in all cases (4/4; 100%). Compared to the GBS group, BSCI prophylaxis was associated with significantly lower cytokine levels including lower IL-8 in amniotic fluid (p = 0.03), TNF-α in fetal plasma (p < 0.05), IFN-α and IL-7 in the fetal lung (p = 0.02) and IL-18, IL-2, and IL-7 in the fetal brain (p = 0.03). Neutrophilic chorioamnionitis was common in the BSCI and GBS groups, but was more severe in the BSCI+GBS group with greater myeloperoxidase staining (granulocyte marker) in the amnion and chorion (p < 0.05 vs. GBS). Collectively, these observations indicate that blocking the chemokine response to infection powerfully suppressed uterine contractility, PTL and the cytokine response, but did not prevent MIAC and fetal pneumonia. Development of PTL immunotherapies should occur in tandem with evaluation for AF microbes and consideration for antibiotic therapy.
Collapse
Affiliation(s)
- Michelle Coleman
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States
| | - Austyn Orvis
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States
| | - Tsung-Yen Wu
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA, United States
| | - Matthew Dacanay
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA, United States
| | - Sean Merillat
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States
| | - Jason Ogle
- Washington National Primate Center, University of Washington, Seattle, WA, United States
| | - Audrey Baldessari
- Washington National Primate Center, University of Washington, Seattle, WA, United States
| | - Nicole M Kretzer
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA, United States
| | - Jeff Munson
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, United States
| | | | - Oksana Shynlova
- Department of Physiology, University of Toronto, Toronto, ON, Canada.,Department of Obstetrics & Gynaecology, University of Toronto, Toronto, ON, Canada
| | - Stephen Lye
- Department of Physiology, University of Toronto, Toronto, ON, Canada.,Department of Obstetrics & Gynaecology, University of Toronto, Toronto, ON, Canada
| | - Lakshmi Rajagopal
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States.,Department of Pediatrics, University of Washington, Seattle, WA, United States.,Department of Global Health, University of Washington, Seattle, WA, United States
| | - Kristina M Adams Waldorf
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA, United States.,Department of Global Health, University of Washington, Seattle, WA, United States
| |
Collapse
|
14
|
Cappelletti M, Presicce P, Kallapur SG. Immunobiology of Acute Chorioamnionitis. Front Immunol 2020; 11:649. [PMID: 32373122 PMCID: PMC7177011 DOI: 10.3389/fimmu.2020.00649] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 03/23/2020] [Indexed: 12/19/2022] Open
Abstract
Acute chorioamnionitis is characterized by neutrophilic infiltration and inflammation at the maternal fetal interface. It is a relatively common complication of pregnancy and can have devastating consequences including preterm labor, maternal infections, fetal infection/inflammation, fetal lung, brain, and gastrointestinal tract injury. In this review, we will discuss current understanding of the pathogenesis, immunobiology, and mechanisms of this condition. Most commonly, acute chorioamnionitis is a result of ascending infection with relatively low-virulence organisms such as the Ureaplasma species. Furthermore, recent vaginal microbiome studies suggest that there is a link between vaginal dysbiosis, vaginal inflammation, and ascending infection. Although less common, microorganisms invading the maternal-fetal interface via hematogenous route (e.g., Zika virus, Cytomegalovirus, and Listeria) can cause placental villitis and severe fetal inflammation and injury. We will provide an overview of the knowledge gleaned from different animal models of acute chorioamnionitis and the role of different immune cells in different maternal-fetal compartments. Lastly, we will discuss how infectious agents can break the maternal tolerance of fetal allograft during pregnancy and highlight the novel future therapeutic approaches.
Collapse
Affiliation(s)
- Monica Cappelletti
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, United States
| | - Pietro Presicce
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, United States
| | - Suhas G Kallapur
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
15
|
Schwenkel G, Romero R, Slutsky R, Motomura K, Hsu CD, Gomez-Lopez N. HSP70: an alarmin that does not induce high rates of preterm birth but does cause adverse neonatal outcomes. J Matern Fetal Neonatal Med 2020; 34:4110-4118. [PMID: 31906756 DOI: 10.1080/14767058.2019.1706470] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Objective: Preterm labor and birth are strongly associated with sterile intra-amniotic inflammation, a clinical condition that is proposed to be initiated by danger signals, or alarmins. The aim of this study was to investigate whether the intra-amniotic administration of the alarmin heat shock protein 70 (HSP70) induces preterm labor/birth and adverse neonatal outcomes.Methods: Pregnant mice received an intra-amniotic injection of 200 ng (n = 8), 400 ng (n = 6), 500 ng (n = 10), or 1 µg of HSP70 (n = 6). Control mice were injected with saline (n = 10). Following injection, the rates of preterm labor/birth and neonatal mortality were recorded. Neonatal weights at weeks 1, 2, and 3 were also recorded.Results: The intra-amniotic injection of 400 ng [late preterm birth 16.7 ± 16.7% (1/6)], 500 ng [early and late preterm birth 10 ± 10% (1/10) each], or 1 µg [early preterm birth 16.7 ± 16.7% (1/6)] of HSP70 induced low rates of preterm/birth. However, the intra-amniotic injection of 500 ng or 1 µg of HSP70 induced significantly higher rates of neonatal mortality compared to controls [saline 14.2% (10/74), 200 ng 9.8% (6/61), 400 ng 17.9% (9/45), 500 ng 28.8% (23/78), and 1 µg 21.4% (13/49)]. Neonates born to dams injected with 200, 500 ng, or 1 µg HSP70 were leaner than controls (p ≤ .05).Conclusion: Intra-amniotic administration of the alarmin HSP70 did not induce high rates of preterm labor/birth; yet, it did indeed result in adverse neonatal outcomes.
Collapse
Affiliation(s)
- George Schwenkel
- 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, U.S. Department of Health and Human Services, Bethesda, MD, USA, and Detroit, MI, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - 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, U.S. Department of Health and Human Services, Bethesda, MD, USA, and Detroit, MI, USA.,Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA.,Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA.,Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
| | - Rebecca Slutsky
- 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, U.S. Department of Health and Human Services, Bethesda, MD, USA, and Detroit, MI, USA
| | - Kenichiro Motomura
- 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, U.S. Department of Health and Human Services, Bethesda, MD, USA, and Detroit, MI, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Chaur-Dong Hsu
- 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, U.S. Department of Health and Human Services, Bethesda, MD, USA, and Detroit, MI, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA.,Department of Physiology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Nardhy Gomez-Lopez
- 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, U.S. Department of Health and Human Services, Bethesda, MD, USA, and Detroit, MI, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA.,Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
16
|
Cheah FC, Lai CH, Tan GC, Swaminathan A, Wong KK, Wong YP, Tan TL. Intrauterine Gardnerella vaginalis Infection Results in Fetal Growth Restriction and Alveolar Septal Hypertrophy in a Rabbit Model. Front Pediatr 2020; 8:593802. [PMID: 33553066 PMCID: PMC7862757 DOI: 10.3389/fped.2020.593802] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 12/16/2020] [Indexed: 11/15/2022] Open
Abstract
Background: Gardnerella vaginalis (GV) is most frequently associated with bacterial vaginosis and is the second most common etiology causing intrauterine infection after Ureaplasma urealyticum. Intrauterine GV infection adversely affects pregnancy outcomes, resulting in preterm birth, fetal growth restriction, and neonatal pneumonia. The knowledge of how GV exerts its effects is limited. We developed an in vivo animal model to study its effects on fetal development. Materials and Methods: A survival mini-laparotomy was conducted on New Zealand rabbits on gestational day 21 (28 weeks of human pregnancy). In each dam, fetuses in the right uterine horn received intra-amniotic 0.5 × 102 colony-forming units of GV injections each, while their littermate controls in the left horn received sterile saline injections. A second laparotomy was performed seven days later. Assessment of the fetal pups, histopathology of the placenta and histomorphometric examination of the fetal lung tissues was done. Results: Three dams with a combined total of 12 fetuses were exposed to intra-amniotic GV, and 9 fetuses were unexposed. The weights of fetuses, placenta, and fetal lung were significantly lower in the GV group than the saline-inoculated control group [mean gross weight, GV (19.8 ± 3.8 g) vs. control (27.9 ± 1.7 g), p < 0.001; mean placenta weight, GV (5.5 ± 1.0 g) vs. control (6.5 ± 0.7 g), p = 0.027; mean fetal lung weight, GV (0.59 ± 0.11 g) vs. control (0.91 ± 0.08 g), p = 0.002. There was a two-fold increase in the multinucleated syncytiotrophoblasts in the placenta of the GV group than their littermate controls (82.9 ± 14.9 vs. 41.6 ± 13.4, p < 0.001). The mean alveolar septae of GV fetuses was significantly thicker than the control (14.8 ± 2.8 μm vs. 12.4 ± 3.8 μm, p = 0.007). Correspondingly, the proliferative index in the interalveolar septum was 1.8-fold higher in the GV group than controls (24.9 ± 6.6% vs. 14.2 ± 2.9%, p = 0.011). The number of alveoli and alveolar surface area did not vary between groups. Discussion: Low-dose intra-amniotic GV injection induces fetal growth restriction, increased placental multinucleated syncytiotrophoblasts and fetal lung re-modeling characterized by alveolar septal hypertrophy with cellular proliferative changes. Conclusion: This intra-amniotic model could be utilized in future studies to elucidate the acute and chronic effects of GV intrauterine infections.
Collapse
Affiliation(s)
- Fook-Choe Cheah
- Department of Pediatrics, Universiti Kebangsaan Malaysia Medical Center, Kuala Lumpur, Malaysia
| | - Chee Hoe Lai
- Department of Pediatrics, Universiti Kebangsaan Malaysia Medical Center, Kuala Lumpur, Malaysia
| | - Geok Chin Tan
- Department of Pathology, Universiti Kebangsaan Malaysia Medical Center, Kuala Lumpur, Malaysia
| | - Anushia Swaminathan
- Department of Pediatrics, Universiti Kebangsaan Malaysia Medical Center, Kuala Lumpur, Malaysia
| | - Kon Ken Wong
- Department of Microbiology, Universiti Kebangsaan Malaysia Medical Center, Kuala Lumpur, Malaysia
| | - Yin Ping Wong
- Department of Pathology, Universiti Kebangsaan Malaysia Medical Center, Kuala Lumpur, Malaysia
| | - Tian-Lee Tan
- Department of Pediatrics, Universiti Kebangsaan Malaysia Medical Center, Kuala Lumpur, Malaysia
| |
Collapse
|
17
|
Stinson LF, Payne MS. Infection-mediated preterm birth: Bacterial origins and avenues for intervention. Aust N Z J Obstet Gynaecol 2019; 59:781-790. [PMID: 31617207 DOI: 10.1111/ajo.13078] [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] [Received: 07/23/2019] [Revised: 08/30/2019] [Accepted: 09/03/2019] [Indexed: 01/07/2023]
Abstract
Preterm birth (PTB) is globally the leading cause of death and disability in children under five years of age. Intra-amniotic infection is well recognised as a major cause of PTB. Importantly, it is the most common cause of extreme PTB (birth prior to 28 weeks gestation), which is frequently associated with a wide range of serious neonatal morbidities. Recent developments in next generation sequencing technologies, combined with many years of culture-based microbiological data have allowed us to gain a deeper understanding of the pathogenesis of infection-mediated PTB. In particular, studies have revealed numerous potential routes to intra-amniotic infection beyond the classically described ascending vaginal route. Currently, antibiotic therapy is standard treatment for suspected or confirmed intra-amniotic infection, although its use in this context has had mixed success due to problems ranging from inappropriate antibiotic selection in relation to the target organism/s, to poor transplacental drug passage. In this review, we will draw together evidence from animal models and human studies to characterise pathways to intra-amniotic infection. We will then thoroughly outline current therapeutic protocols for cases of intra-amniotic infection and suggest potential new avenues for treatment.
Collapse
Affiliation(s)
- Lisa F Stinson
- School of Molecular Sciences, The University of Western Australia, Perth, Australia
| | - Matthew S Payne
- Division of Obstetrics and Gynaecology, Faculty of Health & Medical Sciences, The University of Western Australia, Perth, Australia
| |
Collapse
|
18
|
Bonney EA, Johnson MR. The role of maternal T cell and macrophage activation in preterm birth: Cause or consequence? Placenta 2019; 79:53-61. [PMID: 30929747 DOI: 10.1016/j.placenta.2019.03.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 03/06/2019] [Indexed: 12/18/2022]
Abstract
The role of the immune system in term (TL) and preterm labor (PTL) is unknown. Despite the fact that globally, PTL remains the most important cause of childhood mortality. Infection, typically of the fetal membranes, termed chorioamnionitis, is the best-understood driver of PTL, but the mechanisms underpinning other causes, including idiopathic and stretch-induced PTL, are unclear, but may well involve activation of the maternal immune system. The final common pathway of placental dysfunction, fetal membrane rupture, cervical dilation and uterine contractions are highly complex processes. At term, choriodecidual rather than myometrial inflammation is thought to drive the onset of labor and similar findings are present in different types of PTL including idiopathic PTL. Although accumulated data has confirmed an association between the immune response and preterm birth, there is yet a need to understand if this response is an initiator or a consequence of tissue-level dysregulation. This review focuses on the potential role of macrophages and T cells in innate and adaptive immunity relevant to preterm birth in humans and animal models.
Collapse
Affiliation(s)
- Elizabeth A Bonney
- Department of Obstetrics, Gynecology and Reproductive Sciences University of Vermont, Larner College of Medicine, Burlington, VT, USA.
| | - Mark R Johnson
- Faculty of Medicine, Department of Surgery & Cancer, Imperial College, London, United Kingdom
| |
Collapse
|
19
|
Suff N, Karda R, Diaz JA, Ng J, Baruteau J, Perocheau D, Tangney M, Taylor PW, Peebles D, Buckley SMK, Waddington SN. Ascending Vaginal Infection Using Bioluminescent Bacteria Evokes Intrauterine Inflammation, Preterm Birth, and Neonatal Brain Injury in Pregnant Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:2164-2176. [PMID: 30036519 PMCID: PMC6168615 DOI: 10.1016/j.ajpath.2018.06.016] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 06/08/2018] [Accepted: 06/19/2018] [Indexed: 12/11/2022]
Abstract
Preterm birth is a serious global health problem and the leading cause of infant death before 5 years of age. At least 40% of cases are associated with infection. The most common way for pathogens to access the uterine cavity is by ascending from the vagina. Bioluminescent pathogens have revolutionized the understanding of infectious diseases. We hypothesized that bioluminescent Escherichia coli can be used to track and monitor ascending vaginal infections. Two bioluminescent strains were studied: E. coli K12 MG1655-lux, a nonpathogenic laboratory strain, and E. coli K1 A192PP-lux2, a pathogenic strain capable of causing neonatal meningitis and sepsis in neonatal rats. On embryonic day 16, mice received intravaginal E. coli K12, E. coli K1, or phosphate-buffered saline followed by whole-body bioluminescent imaging. In both cases, intravaginal delivery of E. coli K12 or E. coli K1 led to bacterial ascension into the uterine cavity, but only E. coli K1 induced preterm parturition. Intravaginal administration of E. coli K1 significantly reduced the proportion of pups born alive compared with E. coli K12 and phosphate-buffered saline controls. However, in both groups of viable pups born after bacterial inoculation, there was evidence of comparable brain inflammation by postnatal day 6. This study ascribes specific mechanisms by which exposure to intrauterine bacteria leads to premature delivery and neurologic inflammation in neonates.
Collapse
Affiliation(s)
- Natalie Suff
- Gene Transfer Technology Group, University College London, London, United Kingdom; Preterm Birth Group, Department of Maternal and Fetal Medicine, Institute for Women's Health, University College London, London, United Kingdom
| | - Rajvinder Karda
- Gene Transfer Technology Group, University College London, London, United Kingdom
| | - Juan A Diaz
- Gene Transfer Technology Group, University College London, London, United Kingdom
| | - Joanne Ng
- Gene Transfer Technology Group, University College London, London, United Kingdom
| | - Julien Baruteau
- Gene Transfer Technology Group, University College London, London, United Kingdom; Department of Metabolic Medicine, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Dany Perocheau
- Gene Transfer Technology Group, University College London, London, United Kingdom
| | - Mark Tangney
- SynBio Centre, University College Cork, Cork, Ireland
| | - Peter W Taylor
- School of Pharmacy, University College London, London, United Kingdom
| | - Donald Peebles
- Preterm Birth Group, Department of Maternal and Fetal Medicine, Institute for Women's Health, University College London, London, United Kingdom
| | - Suzanne M K Buckley
- Gene Transfer Technology Group, University College London, London, United Kingdom.
| | - Simon N Waddington
- Gene Transfer Technology Group, University College London, London, United Kingdom; MRC Antiviral Gene Therapy Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
20
|
Rogers LM, Anders AP, Doster RS, Gill EA, Gnecco JS, Holley JM, Randis TM, Ratner AJ, Gaddy JA, Osteen K, Aronoff DM. Decidual stromal cell-derived PGE 2 regulates macrophage responses to microbial threat. Am J Reprod Immunol 2018; 80:e13032. [PMID: 30084522 DOI: 10.1111/aji.13032] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 07/16/2018] [Indexed: 12/30/2022] Open
Abstract
PROBLEM Bacterial chorioamnionitis causes adverse pregnancy outcomes, yet host-microbial interactions are not well characterized within gestational membranes. The decidua, the outermost region of the membranes, is a potential point of entry for bacteria ascending from the vagina to cause chorioamnionitis. We sought to determine whether paracrine communication between decidual stromal cells and macrophages shaped immune responses to microbial sensing. METHOD OF STUDY Decidual cell-macrophage interactions were modeled in vitro utilizing decidualized, telomerase-immortalized human endometrial stromal cells (dTHESCs) and phorbol ester-differentiated THP-1 macrophage-like cells. The production of inflammatory mediators in response to LPS was monitored by ELISA for both cell types, while phagocytosis of bacterial pathogens (Escherichia coli and Group B Streptococcus (GBS)) was measured in THP-1 cells or primary human placental macrophages. Diclofenac, a non-selective cyclooxygenase inhibitor, and prostaglandin E2 (PGE2 ) were utilized to interrogate prostaglandins as decidual cell-derived paracrine immunomodulators. A mouse model of ascending chorioamnionitis caused by GBS was utilized to assess the colocalization of bacteria and macrophages in vivo and assess PGE2 production. RESULTS In response to LPS, dTHESC and THP-1 coculture demonstrated enhancement of most inflammatory mediators, but a potent suppression of macrophage TNF-α generation was observed. This appeared to reflect a paracrine-mediated effect of decidual cell-derived PGE2 . In mice with GBS chorioamnionitis, macrophages accumulated at sites of bacterial invasion with increased PGE2 in amniotic fluid, suggesting such paracrine effects might hold relevance in vivo. CONCLUSION These data suggest key roles for decidual stromal cells in modulating tissue responses to microbial threat through release of PGE2 .
Collapse
Affiliation(s)
- Lisa M Rogers
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Anjali P Anders
- Division of Newborn Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri
| | - Ryan S Doster
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | - Juan S Gnecco
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee.,Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jacob M Holley
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Tara M Randis
- Department of Pediatrics, New York University School of Medicine, New York, New York.,Department of Microbiology, New York University School of Medicine, New York, New York
| | - Adam J Ratner
- Department of Pediatrics, New York University School of Medicine, New York, New York.,Department of Microbiology, New York University School of Medicine, New York, New York
| | - Jennifer A Gaddy
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.,Department of Veteran Affairs, Tennessee Valley Healthcare Systems, Nashville, Tennessee
| | - Kevin Osteen
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee.,Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, Tennessee.,Department of Veteran Affairs, Tennessee Valley Healthcare Systems, Nashville, Tennessee
| | - David M Aronoff
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee.,Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
21
|
Protective effect of Group B Streptococcus type-III polysaccharide conjugates against maternal colonization, ascending infection and neonatal transmission in rodent models. Sci Rep 2018; 8:2593. [PMID: 29416049 PMCID: PMC5803199 DOI: 10.1038/s41598-018-20609-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 01/18/2018] [Indexed: 12/25/2022] Open
Abstract
Group B Streptococcus (GBS) is a normal inhabitant of recto-vaginal mucosae in up to 30% of healthy women. Colonization is a major risk factor for perinatal infection which can lead to severe complications such as stillbirth and neonatal invasive disease. Intra-partum antibiotic prophylaxis in colonized women is a safe and cost-effective preventive measure against early-onset disease in the first days of life, but has no effect on late-onset manifestations or on early maternal infection. Maternal immunization with capsular polysaccharide-based vaccines shows promise for the prevention of both early-onset and late-onset neonatal infections, although ability to prevent maternal colonization and ascending infection has been less studied. Here we investigated the effect of a GBS glycoconjugate vaccine since the very early stage of maternal GBS acquisition to neonatal outcome by rodent models of vaginal colonization and ascending infection. Immunization of female mice and rats with a type III glycoconjugate reduced vaginal colonization, infection of chorioamniotic/ placental membranes and bacterial transmission to fetuses and pups. Type III specific antibodies were detected in the blood and vagina of vaccinated mothers and their offspring. The obtained data support a potential preventive effect of GBS glycoconjugate vaccines during the different stages of pregnancy.
Collapse
|
22
|
Gomez-Lopez N, Romero R, Xu Y, Miller D, Unkel R, Shaman M, Jacques SM, Panaitescu B, Garcia-Flores V, Hassan SS. Neutrophil Extracellular Traps in the Amniotic Cavity of Women with Intra-Amniotic Infection: A New Mechanism of Host Defense. Reprod Sci 2017; 24:1139-1153. [PMID: 27884950 PMCID: PMC6343453 DOI: 10.1177/1933719116678690] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Neutrophil extracellular traps (NETs) control microbial infections through their antimicrobial activities attributed to DNA, histones, granules, and cytoplasmic proteins (eg, elastase). Intra-amniotic infection is characterized by the influx of neutrophils into the amniotic cavity; therefore, the aim of this study was to determine whether amniotic fluid neutrophils form NETs in this inflammatory process. METHODS Amniotic fluid samples from women with intra-amniotic infection (n = 15) were stained for bacteria detection using fluorescent dyes. Amniotic fluid neutrophils were purified by filtration. As controls, neutrophils from maternal blood samples (n = 3) were isolated by density gradients. Isolated neutrophils were plated onto glass cover slips for culture with and without 100 nM of phorbol-12-myristate-13-acetate (PMA). NET formation was assessed by 4',6-diamidino-2-phenylindole (DAPI) staining and scanning electron microscopy. Different stages of NET formation were visualized using antibodies against elastase and histone H3, in combination with DAPI staining, by confocal microscopy. Finally, maternal or neonatal neutrophils were added to amniotic fluid samples from women without intra-amniotic infection (n = 4), and NET formation was evaluated by DAPI staining. RESULTS (1) NETs were present in the amniotic fluid of women with intra-amniotic infection; (2) all of the amniotic fluid samples had detectable live and dead bacteria associated with the presence of NETs; (3) in contrast to neutrophils from the maternal circulation, amniotic fluid neutrophils did not require PMA stimulation to form NETs; (4) different stages of NET formation were observed by co-localizing elastase, histone H3, and DNA in amniotic fluid neutrophils; and (5) neither maternal nor neonatal neutrophils form NETs in the amniotic fluid of women without intra-amniotic infection. CONCLUSION NETs are detectable in the amniotic fluid of women with intra-amniotic infection.
Collapse
Affiliation(s)
- Nardhy Gomez-Lopez
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, USA and Detroit,
MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of
Medicine, Detroit, MI, USA
- Department of Immunology and Microbiology, Wayne State University School of
Medicine, Detroit, MI, USA
| | - Roberto Romero
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, USA and Detroit,
MI, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor,
MI, USA
- Department of Epidemiology and Biostatistics, Michigan State University,
East Lansing, MI, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit,
MI, USA
| | - Yi Xu
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, USA and Detroit,
MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of
Medicine, Detroit, MI, USA
| | - Derek Miller
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, USA and Detroit,
MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of
Medicine, Detroit, MI, USA
- Department of Immunology and Microbiology, Wayne State University School of
Medicine, Detroit, MI, USA
| | - Ronald Unkel
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, USA and Detroit,
MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of
Medicine, Detroit, MI, USA
| | - Majid Shaman
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, USA and Detroit,
MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of
Medicine, Detroit, MI, USA
| | - Suzanne M. Jacques
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, USA and Detroit,
MI, USA
- Department of Pathology, Hutzel Women’s Hospital/Harper University Hospital,
Wayne State University School of Medicine, Detroit, MI, USA
| | - Bogdan Panaitescu
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, USA and Detroit,
MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of
Medicine, Detroit, MI, USA
| | - Valeria Garcia-Flores
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, USA and Detroit,
MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of
Medicine, Detroit, MI, USA
| | - Sonia S. Hassan
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, USA and Detroit,
MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of
Medicine, Detroit, MI, USA
| |
Collapse
|
23
|
Harrell MI, Burnside K, Whidbey C, Vornhagen J, Adams Waldorf KM, Rajagopal L. Exploring the Pregnant Guinea Pig as a Model for Group B Streptococcus Intrauterine Infection. ACTA ACUST UNITED AC 2017; 2. [PMID: 29034376 PMCID: PMC5635843 DOI: 10.4172/2576-1420.1000109] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Infection of the amniotic cavity remains a major cause of preterm birth, stillbirth, fetal injury and early onset, fulminant infections in newborns. Currently, there are no effective therapies to prevent in utero infection and consequent co-morbidities. This is in part due to the lack of feasible and appropriate animal models to understand mechanisms that lead to in utero infections. Use of mouse and rat models do not fully recapitulate human pregnancy, while pregnant nonhuman primate models are limited by ethical considerations, technical constraints, and cost. Given these limitations, the guinea pig is an attractive animal model for studying pregnancy infections, particularly as the placental structure is quite similar to the human placenta. Here, we describe our studies that explored the pregnant guinea pig as a model to study in utero Group B Streptococci (GBS) infections. We observed that intrauterine inoculation of wild type GBS in pregnant guinea pigs resulted in bacterial invasion and dissemination to the placenta, amniotic fluid and fetal organs. Also, hyperhemolytic GBS such as those lacking the hemolysin repressor CovR/S showed increased dissemination into the amniotic fluid and fetal organs such as the fetal lung and brain. These results are similar to those observed in mouse and non-human primate models of in utero infection, and support use of the guinea pig as a model for studying GBS infections in pregnancy.
Collapse
Affiliation(s)
- Maria I Harrell
- Department of Pediatrics, University of Washington and Seattle Children's Research Institute, Seattle, Washington, United States of America
| | - Kellie Burnside
- Department of Pediatrics, University of Washington and Seattle Children's Research Institute, Seattle, Washington, United States of America
| | - Christopher Whidbey
- Department of Pediatrics, University of Washington and Seattle Children's Research Institute, Seattle, Washington, United States of America.,Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - Jay Vornhagen
- Department of Pediatrics, University of Washington and Seattle Children's Research Institute, Seattle, Washington, United States of America.,Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - Kristina M Adams Waldorf
- Department of Global Health, University of Washington, Seattle, Washington, United States of America.,Department of Obstetrics and Gynecology, School of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Lakshmi Rajagopal
- Department of Pediatrics, University of Washington and Seattle Children's Research Institute, Seattle, Washington, United States of America.,Department of Global Health, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
24
|
Gomez-Lopez N, Romero R, Arenas-Hernandez M, Panaitescu B, Garcia-Flores V, Mial TN, Sahi A, Hassan SS. Intra-amniotic administration of lipopolysaccharide induces spontaneous preterm labor and birth in the absence of a body temperature change. J Matern Fetal Neonatal Med 2017; 31:439-446. [PMID: 28139962 DOI: 10.1080/14767058.2017.1287894] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Intra-amniotic infection is associated with spontaneous preterm labor. In most cases, the infection is subclinical and bacteria are detected in the amniotic cavity rather than in the chorioamniotic membranes. The aims of this study were to establish a model of intra-amniotic lipopolysaccharide (LPS)-induced preterm labor/birth that resembles the subclinical syndrome and to compare this model to two established models of LPS-induced preterm labor/birth. METHODS Pregnant B6 mice received an intra-amniotic, intra-uterine, or intra-peritoneal injection of LPS (100 ng/amniotic sac, 15 μg/25 μL, and 15 μg/200 μL respectively) or PBS (control). Following injection, body temperature (every two hours for a 12-h period), gestational age, and the rate of preterm labor/birth were recorded. RESULTS An intra-amniotic injection of LPS resulted in preterm labor/birth [LPS 80 ± 24.79% (8/10) versus PBS 0% (0/8); p = 0.001] without causing maternal hypothermia. Intra-peritoneal [LPS 100% (8/8) versus PBS 0% (0/8); p < 0.001)] and intra-uterine [LPS 100% (8/8) versus PBS 28.57 ± 33.47% (2/7); p =0 .007] injections of LPS induced preterm labor/birth; yet, maternal hypothermia was observed. CONCLUSION Intra-amniotic injection of LPS induces preterm labor/birth in the absence of a body temperature change, which resembles the subclinical syndrome.
Collapse
Affiliation(s)
- Nardhy Gomez-Lopez
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services , Bethesda , MD , and Detroit , MI , USA.,b Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA.,c Department of Immunology and Microbiology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Roberto Romero
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services , Bethesda , MD , and Detroit , MI , USA.,d Department of Obstetrics and Gynecology , University of Michigan , Ann Arbor , MI , USA.,e Department of Epidemiology and Biostatistics , Michigan State University , East Lansing , MI , USA.,f Center for Molecular Medicine and Genetics , Wayne State University , Detroit , MI , USA
| | - Marcia Arenas-Hernandez
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services , Bethesda , MD , and Detroit , MI , USA.,b Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Bogdan Panaitescu
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services , Bethesda , MD , and Detroit , MI , USA.,b Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Valeria Garcia-Flores
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services , Bethesda , MD , and Detroit , MI , USA.,b Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Tara N Mial
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services , Bethesda , MD , and Detroit , MI , USA.,b Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Aashna Sahi
- b Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| | - Sonia S Hassan
- a Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research , Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services , Bethesda , MD , and Detroit , MI , USA.,b Department of Obstetrics and Gynecology , Wayne State University School of Medicine , Detroit , MI , USA
| |
Collapse
|
25
|
Masselli G, Perrone G, Kinkel K, Di Tola M, Laghi F, Gualdi G, Brunelli R. Are Second Trimester Apparent Diffusion Coefficient Values of the Short Uterine Cervix Associated with Impending Preterm Delivery? Radiology 2016; 280:897-904. [DOI: 10.1148/radiol.2016150670] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
26
|
Choi J, Park JW, Kim BJ, Choi YJ, Hwang JH, Lee SM. Funisitis is more common in cervical insufficiency than in preterm labor and preterm premature rupture of membranes. J Perinat Med 2016; 44:523-9. [PMID: 26812854 DOI: 10.1515/jpm-2015-0123] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 12/07/2015] [Indexed: 11/15/2022]
Abstract
OBJECTIVE To compare the frequency of histologic chorioamnionitis and funisitis among women experiencing preterm labor, preterm premature rupture of membranes (PROM) and cervical insufficiency. METHODS This retrospective cohort study included singleton pregnant women who delivered at ≤36 weeks of gestation. The patients with preterm birth were subdivided into preterm labor (n=117), preterm PROM (n=153), and cervical insufficiency (n=20). All placentas were examined for pathology, according to the criteria of Salafia with minor modification. Frequencies of histologic chorioamnionitis and funisitis were evaluated according to the causes of preterm birth. RESULTS 1) Histologic chorioamnionitis was diagnosed in 48.7% (57/117) of cases with preterm labor, 47.4% (73/153) with preterm PROM, and 75.0% (15/20) with cervical insufficiency. Funisitis was detected in 11.1% (13/117) of cases with preterm labor, 15.7% (24/153) with preterm PROM, and 40.0% (8/20) with cervical insufficiency. 2) Frequency of histologic chorioamnionitis was higher in cases with cervical insufficiency compared to preterm PROM. Frequency of funisitis was higher in cases with cervical insufficiency compared to both preterm labor and preterm PROM (P<0.05). The difference in frequency of funisitis remained significant after adjustment for gestational age at delivery and cervical dilatation at diagnosis. 3) Frequency of grade 2 funisitis was higher in cases with cervical insufficiency (35.0%, 7/20) compared to both preterm labor (6.8%, 8/117) and preterm PROM (9.8%, 15/153) (P=0.001). And the difference remained significant after adjustment for gestational age at delivery and cervical dilatation at diagnosis. CONCLUSION The highest frequency of funisitis was observed in cervical insufficiency among cases with spontaneous preterm birth.
Collapse
|
27
|
Kemp MW, Molloy TJ, Usuda H, Woodward E, Miura Y, Payne MS, Ireland DJ, Jobe AH, Kallapur SG, Stock SJ, Spiller OB, Newnham JP, Saito M. Outside-in? Acute fetal systemic inflammation in very preterm chronically catheterized sheep fetuses is not driven by cells in the fetal blood. Am J Obstet Gynecol 2016; 214:281.e1-281.e10. [PMID: 26408085 DOI: 10.1016/j.ajog.2015.09.076] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 08/25/2015] [Accepted: 09/15/2015] [Indexed: 11/17/2022]
Abstract
BACKGROUND The preterm birth syndrome (delivery before 37 weeks gestation) is a major contributor to the global burden of perinatal morbidity and death. The cause of preterm birth is complex, multifactorial, and likely dependent, at least in part, on the gestational age of the fetus. Intrauterine infection is frequent in preterm deliveries that occur at <32 weeks gestation; understanding how the fetus responds to proinflammatory insult will be an important step towards early preterm birth prevention. However, animal studies of infection and inflammation in prematurity commonly use older fetuses that possess comparatively mature immune systems. OBJECTIVE Aiming to characterize acute fetal responses to microbial agonist at a clinically relevant gestation, we used 92-day-old fetuses (62% of term) to develop a chronically catheterized sheep model of very preterm pregnancy. We hypothesized that any acute fetal systemic inflammatory responses would be driven by signaling from the tissues exposed to Escherichia coli lipopolysaccharide that is introduced into the amniotic fluid. STUDY DESIGN Eighteen ewes that were carrying a single fetus at 92 days of gestation had recovery surgery to place fetal tracheal, jugular, and intraamniotic catheters. Animals were recovered for 24 hours before being administered either intraamniotic E coli lipopolysaccharide (n = 9) or sterile saline solution (n = 9). Samples were collected for 48 hours before euthanasia and necroscopy. Fetal inflammatory responses were characterized by microarray analysis, quantitative polymerase chain reaction, and enzyme-linked immunosorbent assay. RESULTS Intraamniotic lipopolysaccharide reached the distal trachea within 2 hours. Lipopolysaccharide increased tracheal fluid interleukin-8 within 2 hours and generated a robust inflammatory response that was characterized by interleukin-6 signaling pathway activation and up-regulation of cell proliferation but no increases in inflammatory mediator expression in cord blood RNA. CONCLUSIONS In very preterm sheep fetuses, lipopolysaccharide stimulates inflammation in the fetal lung and fetal skin and stimulates a systemic inflammatory response that is not generated by fetal blood cells. These data argue for amniotic fluid-exposed tissues that play a key role in driving acute fetal and intrauterine inflammatory responses.
Collapse
Affiliation(s)
- Matthew W Kemp
- School of Women's and Infants' Health, The University of Western Australia, Perth, Western Australia, Australia.
| | - Timothy J Molloy
- Blood, Stem Cells and Cancer Research Programme, St Vincent's Centre for Applied Medical Research, Darlinghurst, NSW, Australia
| | - Haruo Usuda
- Division of Perinatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Eleanor Woodward
- School of Women's and Infants' Health, The University of Western Australia, Perth, Western Australia, Australia
| | - Yuichiro Miura
- School of Women's and Infants' Health, The University of Western Australia, Perth, Western Australia, Australia
| | - Matthew S Payne
- School of Women's and Infants' Health, The University of Western Australia, Perth, Western Australia, Australia
| | - Demelza J Ireland
- School of Women's and Infants' Health, The University of Western Australia, Perth, Western Australia, Australia
| | - Alan H Jobe
- School of Women's and Infants' Health, The University of Western Australia, Perth, Western Australia, Australia; Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Centre, University of Cincinnati, School of Medicine, Cincinnati, OH
| | - Suhas G Kallapur
- School of Women's and Infants' Health, The University of Western Australia, Perth, Western Australia, Australia; Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Centre, University of Cincinnati, School of Medicine, Cincinnati, OH
| | - Sarah J Stock
- MRC Centre for Reproductive Health, University of Edinburgh Queen's Medical Research Institute, Edinburgh, UK
| | - Owen B Spiller
- Cardiff University, School of Medicine, Institute of Molecular and Experimental Medicine, University Hospital of Wales, Cardiff, UK
| | - John P Newnham
- School of Women's and Infants' Health, The University of Western Australia, Perth, Western Australia, Australia
| | - Masatoshi Saito
- School of Women's and Infants' Health, The University of Western Australia, Perth, Western Australia, Australia; Division of Perinatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| |
Collapse
|
28
|
Kim CJ, Romero R, Chaemsaithong P, Chaiyasit N, Yoon BH, Kim YM. Acute chorioamnionitis and funisitis: definition, pathologic features, and clinical significance. Am J Obstet Gynecol 2015; 213:S29-52. [PMID: 26428501 PMCID: PMC4774647 DOI: 10.1016/j.ajog.2015.08.040] [Citation(s) in RCA: 618] [Impact Index Per Article: 68.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Revised: 08/12/2015] [Accepted: 08/16/2015] [Indexed: 12/25/2022]
Abstract
Acute inflammatory lesions of the placenta consist of diffuse infiltration of neutrophils at different sites in the organ. These lesions include acute chorioamnionitis, funisitis, and chorionic vasculitis and represent a host response (maternal or fetal) to a chemotactic gradient in the amniotic cavity. While acute chorioamnionitis is evidence of a maternal host response, funisitis and chorionic vasculitis represent fetal inflammatory responses. Intraamniotic infection generally has been considered to be the cause of acute chorioamnionitis and funisitis; however, recent evidence indicates that "sterile" intraamniotic inflammation, which occurs in the absence of demonstrable microorganisms induced by "danger signals," is frequently associated with these lesions. In the context of intraamniotic infection, chemokines (such as interleukin-8 and granulocyte chemotactic protein) establish a gradient that favors the migration of neutrophils from the maternal or fetal circulation into the chorioamniotic membranes or umbilical cord, respectively. Danger signals that are released during the course of cellular stress or cell death can also induce the release of neutrophil chemokines. The prevalence of chorioamnionitis is a function of gestational age at birth, and present in 3-5% of term placentas and in 94% of placentas delivered at 21-24 weeks of gestation. The frequency is higher in patients with spontaneous labor, preterm labor, clinical chorioamnionitis (preterm or term), or ruptured membranes. Funisitis and chorionic vasculitis are the hallmarks of the fetal inflammatory response syndrome, a condition characterized by an elevation in the fetal plasma concentration of interleukin-6, and associated with the impending onset of preterm labor, a higher rate of neonatal morbidity (after adjustment for gestational age), and multiorgan fetal involvement. This syndrome is the counterpart of the systemic inflammatory response syndrome in adults: a risk factor for short- and long-term complications (ie, sterile inflammation in fetuses, neonatal sepsis, bronchopulmonary dysplasia, periventricular leukomalacia, and cerebral palsy). This article reviews the definition, pathogenesis, grading and staging, and clinical significance of the most common lesions in placental disease. Illustrations of the lesions and diagrams of the mechanisms of disease are provided.
Collapse
Affiliation(s)
- Chong Jai Kim
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea; Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI
| | - Roberto Romero
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, 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, Detroit, MI.
| | - Piya Chaemsaithong
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Noppadol Chaiyasit
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Bo Hyun Yoon
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI; Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Yeon Mee Kim
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI; Department of Pathology, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Korea
| |
Collapse
|
29
|
Nan C, Dangor Z, Cutland CL, Edwards MS, Madhi SA, Cunnington MC. Maternal group B Streptococcus-related stillbirth: a systematic review. BJOG 2015; 122:1437-45. [PMID: 26177561 DOI: 10.1111/1471-0528.13527] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2015] [Indexed: 11/26/2022]
Abstract
BACKGROUND Limited epidemiological data on the association between maternal rectovaginal group B Streptococcus (GBS) colonisation and stillbirth makes assessment of antenatal interventions for GBS stillbirth difficult. OBJECTIVES To systematically review the existing literature and evaluate the incidence of GBS-related stillbirth by region up to March 2015. SEARCH STRATEGY A systematic review of the published literature was completed using PubMed/MEDLINE, EMBASE, LILACS, and Cochrane Library, with Medical Subject Headings (MeSH) and search terms based upon the Centers for Disease Control and Prevention's (CDC) Active Bacterial Core Surveillance (ABCs) GBS-related stillbirth definition and chorioamnionitis. SELECTION CRITERIA Studies reporting original data on GBS-related stillbirth occurring ≥20 weeks of gestation, with GBS confirmed by autopsy or by culture from the placenta, amniotic fluid, or other normally sterile site samples from the stillborn. DATA COLLECTION AND ANALYSIS Descriptive analyses were performed with the absolute GBS-related stillbirth rates and proportion of stillbirths attributed to GBS calculated per study where possible. Differences in stillbirth definitions did not allow for pooled estimates to be calculated. MAIN RESULTS Seventeen studies reported GBS-related stillbirth rates varying from 0.04 to 0.9 per 1000 births, with the proportion of stillbirths associated with GBS ranging from 0 to 12.1%. Most studies reported data from before the year 2000 and from high-income countries. CONCLUSIONS The sparsely available epidemiological evidence was not reported consistently, emphasising the importance of standardised stillbirth definitions and diagnostic methods to optimally assess the effectiveness of any future antenatal interventions. Timing of stillbirth, GBS serotype, and global diversity were gaps in the current evidence. TWEETABLE ABSTRACT Systematic review finds Group B Streptococcus causes up to 12.1% of stillbirths, but more research is needed.
Collapse
Affiliation(s)
- C Nan
- Cassandra Nan, Research Consultant, Maastricht, the Netherlands
| | - Z Dangor
- Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Department of Science and Technology/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand, Johannesburg, South Africa
| | - C L Cutland
- Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Department of Science and Technology/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand, Johannesburg, South Africa
| | - M S Edwards
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - S A Madhi
- Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Department of Science and Technology/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand, Johannesburg, South Africa
| | | |
Collapse
|
30
|
Musilova I, Pliskova L, Kutova R, Jacobsson B, Paterova P, Kacerovsky M. Streptococcus agalactiae in pregnancies complicated by preterm prelabor rupture of membranes. J Matern Fetal Neonatal Med 2015; 29:1036-40. [PMID: 25923242 DOI: 10.3109/14767058.2015.1038514] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE The main aim of this study was to evaluate the presence of Streptococcus agalactiae (S. agalactiae) in the vagina and the amniotic fluid in pregnancies complicated by preterm prelabor rupture of membranes (PPROM). The next aim was to evaluate the incidence of S. agalactiae early onset sepsis in newborns from PPROM pregnancies, with respect to the presence of S. agalactiae in the vagina and the amniotic fluid. METHODS Singleton gestations with PPROM between 24 + 0 and 36 + 6 were included. A vaginal swab was obtained, and amniocentesis was performed at admission. The presence of S. agalactiae in the vagina and in the amniotic fluid was assessed by culture and by real-time polymerase chain reaction, respectively. RESULTS In total, 336 women were included. The presence of S. agalactiae in the vaginal and amniotic fluid was found in 9% (31/336) and 1% (3/336) of women. One woman had S. agalactiae in the amniotic fluid but was negative for the presence of S. agalactiae in the vaginal fluid. Early onset neonatal sepsis developed in one newborn from pregnancies complicated by the presence of S. agalactiae in the amniotic fluid. CONCLUSION The presence of S. agalactiae in the vagina and amniotic fluid complicated approximately each 10th and each 100th PPROM pregnancy. Cultivation-negative findings of S. agalactiae in the vagina did not exclude the positivity of the amniotic fluid for S. agalactiae and the development of early onset sepsis in newborns.
Collapse
Affiliation(s)
- Ivana Musilova
- a Department of Obstetrics and Gynecology, Faculty of Medicine in Hradec Kralove , Charles University in Prague, University Hospital Hradec Kralove , Czech Republic
| | - Lenka Pliskova
- b Institute of Clinical Biochemistry and Diagnosis, Faculty of Medicine in Hradec Kralove, Charles University in Prague, University Hospital Hradec Kralove , Czech Republic
| | - Radka Kutova
- b Institute of Clinical Biochemistry and Diagnosis, Faculty of Medicine in Hradec Kralove, Charles University in Prague, University Hospital Hradec Kralove , Czech Republic
| | - Bo Jacobsson
- c Department of Obstetrics and Gynecology , Sahlgrenska Academy , Gothenburg , Sweden .,d Norwegian Institute of Public Health, Division of Epidemiology , Department of Genes and Environment , Oslo , Norway
| | - Pavla Paterova
- e Institute of Clinical Microbiology, Faculty of Medicine in Hradec Kralove, Charles University in Prague, University Hospital Hradec Kralove , Czech Republic , and
| | - Marian Kacerovsky
- a Department of Obstetrics and Gynecology, Faculty of Medicine in Hradec Kralove , Charles University in Prague, University Hospital Hradec Kralove , Czech Republic .,f Biomedical Research Center , University Hospital Hradec Kralove , Czech Republic
| |
Collapse
|
31
|
Presicce P, Senthamaraikannan P, Alvarez M, Rueda CM, Cappelletti M, Miller LA, Jobe AH, Chougnet CA, Kallapur SG. Neutrophil recruitment and activation in decidua with intra-amniotic IL-1beta in the preterm rhesus macaque. Biol Reprod 2015; 92:56. [PMID: 25537373 PMCID: PMC4342792 DOI: 10.1095/biolreprod.114.124420] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 09/26/2014] [Accepted: 12/22/2014] [Indexed: 11/01/2022] Open
Abstract
Chorioamnionitis, an infection/inflammation of the fetomaternal membranes, is frequently associated with preterm delivery. The mechanisms of inflammation in chorioamnionitis are poorly understood. We hypothesized that neutrophils recruited to the decidua would be the major producers of proinflammatory cytokines. We injected intra-amniotic (IA) interleukin 1beta (IL-1beta) at ∼80% gestation in rhesus macaque monkeys, Macaca mulatta, delivered the fetuses surgically 24 h or 72 h after IA injections, and investigated the role of immune cells in the chorion-amnion decidua. IA IL-1beta induced a robust infiltration of neutrophils and significant increases of proinflammatory cytokines in the chorioamnion decidua at 24 h after exposure, with a subsequent decrease at 72 h. Neutrophils in the decidua were the major source of tumor necrosis factor alpha (TNFalpha) and IL-8. Interestingly, IA IL-1beta also induced a significant increase in anti-inflammatory indoleamine 2,3-dioxygenase (IDO) expression in the decidua neutrophils. The frequency of regulatory T cells (Tregs) and FOXP3 mRNA expression in the decidua did not change after IA IL-1beta injection. Collectively, our data demonstrate that in this model of sterile chorioamnionitis, the decidua neutrophils cause the inflammation in the gestational tissues but may also act as regulators to dampen the inflammation. These results help to understand the contribution of neutrophils to the pathogenesis of chorioamnionitis-induced preterm labor.
Collapse
Affiliation(s)
- Pietro Presicce
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| | | | - Manuel Alvarez
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| | - Cesar M Rueda
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio Division of Immunobiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Monica Cappelletti
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Lisa A Miller
- California National Primate Research Center, University of California Davis, Davis, California Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California
| | - Alan H Jobe
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| | - Claire A Chougnet
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio Division of Immunobiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Suhas G Kallapur
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| |
Collapse
|
32
|
Cheung CY, Beardall MK, Anderson DF, Brace RA. Prostaglandin E2 regulation of amnion cell vascular endothelial growth factor expression: relationship with intramembranous absorption rate in fetal sheep. Am J Physiol Regul Integr Comp Physiol 2014; 307:R354-60. [PMID: 24898841 DOI: 10.1152/ajpregu.00070.2014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We hypothesized that prostaglandin E2 (PGE2) stimulates amniotic fluid transport across the amnion by upregulating vascular endothelial growth factor (VEGF) expression in amnion cells and that amniotic PGE2 concentration correlates positively with intramembranous (IM) absorption rate in fetal sheep. The effects of PGE2 at a range of concentrations on VEGF 164 and caveolin-1 gene expressions were analyzed in cultured ovine amnion cells. IM absorption rate, amniotic fluid (AF) volume, and PGE2 concentration in AF were determined in late-gestation fetal sheep during control conditions, isovolumic fetal urine replacement (low IM absorption rate), or intra-amniotic fluid infusion (high IM absorption rate). In ovine amnion cells, PGE2 induced dose- and time-dependent increases in VEGF 164 mRNA levels and reduced caveolin-1 mRNA and protein levels. VEGF receptor blockade abolished the caveolin-1 response, while minimally affecting the VEGF response to PGE2. In sheep fetuses, urine replacement reduced amniotic PGE2 concentration by 58%, decreased IM absorption rate by half, and doubled AF volume (P < 0.01). Intra-amniotic fluid infusion increased IM absorption rate and AF volume (P < 0.01), while amniotic PGE2 concentration was unchanged. Neither IM absorption rate nor AF volume correlated with amniotic PGE2 concentration under each experimental condition. Although PGE2 at micromolar concentrations induced dose-dependent responses in VEGF and caveolin-1 gene expression in cultured amnion cells consistent with a role of PGE2 in activating VEGF to mediate AF transport across the amnion, amniotic PGE2 at physiological nanomolar concentrations does not appear to regulate IM absorption rate or AF volume.
Collapse
Affiliation(s)
- Cecilia Y Cheung
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, Oregon; and Center for Developmental Health, Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon
| | - Michael K Beardall
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, Oregon; and
| | - Debra F Anderson
- Center for Developmental Health, Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon
| | - Robert A Brace
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, Oregon; and Center for Developmental Health, Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
33
|
Lyle SK. Immunology of infective preterm delivery in the mare. Equine Vet J 2014; 46:661-8. [DOI: 10.1111/evj.12243] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 02/04/2014] [Indexed: 01/22/2023]
Affiliation(s)
- S. K. Lyle
- Department of Veterinary Clinical Sciences, School of Veterinary Medicine Louisiana State University Baton Rouge Louisiana USA
| |
Collapse
|
34
|
Acosta EP, Grigsby PL, Larson KB, James AM, Long MC, Duffy LB, Waites KB, Novy MJ. Transplacental transfer of Azithromycin and its use for eradicating intra-amniotic ureaplasma infection in a primate model. J Infect Dis 2014; 209:898-904. [PMID: 24179112 PMCID: PMC3935474 DOI: 10.1093/infdis/jit578] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 09/20/2013] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Our goals were to describe azithromycin (AZI) pharmacokinetics in maternal plasma (MP), fetal plasma (FP), and amniotic fluid (AF) following intra-amniotic infection (IAI) with Ureaplasma in pregnant rhesus monkeys and to explore concentration-response relationships. METHODS Following intra-amniotic inoculation of Ureaplasma parvum, rhesus monkeys received AZI (12.5 mg/kg every 12 hours intravenously for 10 days; n = 10). Intensive pharmacokinetic sampling of MP, FP, and AF was scheduled following the first (ie, single) dose and the last (ie, multiple) dose. Noncompartmental and pharmacokinetic modeling methods were used. RESULTS The AF area under the concentration-time curve at 12 hours was 0.22 µg×h/mL following a single dose and 6.3 µg×h/mL at day 10. MP and AF accumulation indices were 8.4 and 19, respectively. AZI AF half-life following the single dose and multiple dose were 156 and 129 hours, respectively. The median MP:FP ratio in concomitantly drawn samples was 3.2 (range, 1.3-9.6; n = 9). Eradication of U. parvum occurred at 6.6 days, with a 95% effective concentration (EC95) of 39 ng/mL for the maximum AZI AF concentration. CONCLUSIONS Our study demonstrates that a maternal multiple-dose AZI regimen is effective in eradicating U. parvum IAI by virtue of intra-amniotic accumulation and suggests that antenatal therapy has the potential to mitigate complications associated with U. parvum infection in pregnancy, such as preterm labor and fetal sequelae.
Collapse
Affiliation(s)
| | - Peta L. Grigsby
- Oregon National Primate Research Center
- Division of Reproductive and Developmental Sciences
- Department of Obstetrics and Gynecology, Oregon Health and Science University, Beaverton
| | | | | | | | - Lynn B. Duffy
- Department of Pathology, University of Alabama at Birmingham
| | - Ken B. Waites
- Department of Pathology, University of Alabama at Birmingham
| | - Miles J. Novy
- Oregon National Primate Research Center
- Division of Reproductive and Developmental Sciences
- Department of Obstetrics and Gynecology, Oregon Health and Science University, Beaverton
| |
Collapse
|
35
|
Kallapur SG, Presicce P, Rueda CM, Jobe AH, Chougnet CA. Fetal immune response to chorioamnionitis. Semin Reprod Med 2014; 32:56-67. [PMID: 24390922 DOI: 10.1055/s-0033-1361823] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Chorioamnionitis is a frequent cause of preterm birth and is associated with an increased risk for injury responses in the lung, gastrointestinal tract, brain, and other fetal organs. Chorioamnionitis is a polymicrobial nontraditional infectious disease because the organisms causing chorioamnionitis are generally of low virulence and colonize the amniotic fluid often for extended periods, and the host (mother and the fetus) does not have typical infection-related symptoms such as fever. In this review, we discuss the effects of chorioamnionitis in experimental animal models that mimic the human disease. Our focus is on the immune changes in multiple fetal organs and the pathogenesis of chorioamnionitis-induced injury in different fetal compartments. As chorioamnionitis disproportionately affects preterm infants, we discuss the relevant developmental context for the immune system. We also provide a clinical context for the fetal responses.
Collapse
Affiliation(s)
- Suhas G Kallapur
- Division of Neonatology/Pulmonary Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati
| | - Pietro Presicce
- Division of Neonatology/Pulmonary Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati
| | - Cesar M Rueda
- Division of Immunobiology, The Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| | - Alan H Jobe
- Division of Neonatology/Pulmonary Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati
| | - Claire A Chougnet
- Division of Immunobiology, The Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| |
Collapse
|
36
|
Raiche E, Ouellet A, Berthiaume M, Rousseau É, Pasquier JC. Short and inflamed cervix predicts spontaneous preterm birth (COLIBRI study). J Matern Fetal Neonatal Med 2013; 27:1015-9. [PMID: 24228627 DOI: 10.3109/14767058.2013.847917] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE To develop a new strategy of predicting spontaneous preterm birth (sPTB) by combination of transvaginal ultrasound (TVUS) assessment and inflammatory proteins detection in vaginal secretions. METHODS Prospective study of 87 women referred for cervical length assessment with a standardized TVUS combined to vaginal secretions sampling. Samples were analyzed for presence of 10 cytokines. Main outcome was sPTB (<37 weeks of gestation). Associations were assessed with the chi-square, Fisher's exact test (p < 0.05) and Wald's logistic regression. RESULTS sPTB occurred in 25.3% of women at a median gestational age of 35.6 weeks of gestation. Short cervix (<25 mm) (n = 24) was associated with sPTB (p < 0.01) as interleukine (IL)-1β, IL-8 and IL-10 in vaginal secretions (p < 0.05). In multivariate analysis, short cervix and IL-8 in vaginal secretions were independently associated with sPTB (OR 3.58 (95%CI 1.02; 12.61) and 14.55 (95%CI 1.64; 128.83), respectively) as their combination (OR 4.33 (95%CI 1.25; 14.95)). By categorizing cervical length by presence of IL-8, sPTB occurred in 55.6% of women with a short inflamed cervix. CONCLUSION COLIBRI study used a novel, single-step method of vaginal secretions sampling during TVUS and demonstrated that combination of short cervix and IL-8 in vaginal secretions is a promising sPTB predictive test.
Collapse
|
37
|
Saito M, Payne MS, Miura Y, Ireland DJ, Stock S, Kallapur SG, Kannan PS, Newnham JP, Kramer BW, Jobe AH, Keelan JA, Kemp MW. Polymyxin B Agonist Capture Therapy for Intrauterine Inflammation. Reprod Sci 2013; 21:623-31. [DOI: 10.1177/1933719113508820] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Masatoshi Saito
- School of Women’s and Infants’ Health, The University of Western Australia, Perth, Western Australia
- Division of Perinatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Matthew S. Payne
- School of Women’s and Infants’ Health, The University of Western Australia, Perth, Western Australia
| | - Yuichiro Miura
- School of Women’s and Infants’ Health, The University of Western Australia, Perth, Western Australia
| | - Demelza J. Ireland
- School of Women’s and Infants’ Health, The University of Western Australia, Perth, Western Australia
| | - Sarah Stock
- School of Women’s and Infants’ Health, The University of Western Australia, Perth, Western Australia
| | - Suhas G. Kallapur
- School of Women’s and Infants’ Health, The University of Western Australia, Perth, Western Australia
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Centre, University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | - Paranthaman S. Kannan
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Centre, University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | - John P. Newnham
- School of Women’s and Infants’ Health, The University of Western Australia, Perth, Western Australia
| | - Boris W. Kramer
- School of Women’s and Infants’ Health, The University of Western Australia, Perth, Western Australia
- Department of Paediatrics, School of Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Alan H. Jobe
- School of Women’s and Infants’ Health, The University of Western Australia, Perth, Western Australia
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Centre, University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | - Jeffrey A. Keelan
- School of Women’s and Infants’ Health, The University of Western Australia, Perth, Western Australia
| | - Matthew W. Kemp
- School of Women’s and Infants’ Health, The University of Western Australia, Perth, Western Australia
| |
Collapse
|
38
|
Lee J, Romero R, Chaiworapongsa T, Dong Z, Tarca AL, Xu Y, Chiang PJ, Kusanovic JP, Hassan SS, Yeo L, Yoon BH, Than NG, Kim CJ. Characterization of the fetal blood transcriptome and proteome in maternal anti-fetal rejection: evidence of a distinct and novel type of human fetal systemic inflammatory response. Am J Reprod Immunol 2013; 70:265-84. [PMID: 23905683 DOI: 10.1111/aji.12142] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 05/07/2013] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The human fetus is able to mount a systemic inflammatory response when exposed to microorganisms. This stereotypic response has been termed the 'fetal inflammatory response syndrome' (FIRS), defined as an elevation of fetal plasma interleukin-6 (IL-6). FIRS is frequently observed in patients whose preterm deliveries are associated with intra-amniotic infection, acute inflammatory lesions of the placenta, and a high rate of neonatal morbidity. Recently, a novel form of fetal systemic inflammation, characterized by an elevation of fetal plasma CXCL10, has been identified in patients with placental lesions consistent with 'maternal anti-fetal rejection'. These lesions include chronic chorioamnionitis, plasma cell deciduitis, and villitis of unknown etiology. In addition, positivity for human leukocyte antigen (HLA) panel-reactive antibodies (PRA) in maternal sera can also be used to increase the index of suspicion for maternal anti-fetal rejection. The purpose of this study was to determine (i) the frequency of pathologic lesions consistent with maternal anti-fetal rejection in term and spontaneous preterm births; (ii) the fetal serum concentration of CXCL10 in patients with and without evidence of maternal anti-fetal rejection; and (iii) the fetal blood transcriptome and proteome in cases with a fetal inflammatory response associated with maternal anti-fetal rejection. METHOD OF STUDY Maternal and fetal sera were obtained from normal term (n = 150) and spontaneous preterm births (n = 150). A fetal inflammatory response associated with maternal anti-fetal rejection was diagnosed when the patients met two or more of the following criteria: (i) presence of chronic placental inflammation; (ii) ≥80% of maternal HLA class I PRA positivity; and (iii) fetal serum CXCL10 concentration >75th percentile. Maternal HLA PRA was analyzed by flow cytometry. The concentrations of fetal CXCL10 and IL-6 were determined by ELISA. Transcriptome analysis was undertaken after the extraction of total RNA from white blood cells with a whole-genome DASL assay. Proteomic analysis of fetal serum was conducted by two-dimensional difference gel electrophoresis. Differential gene expression was considered significant when there was a P < 0.01 and a fold-change >1.5. RESULTS (i) The frequency of placental lesions consistent with maternal anti-fetal rejection was higher in patients with preterm deliveries than in those with term deliveries (56% versus 32%; P < 0.001); (ii) patients with spontaneous preterm births had a higher rate of maternal HLA PRA class I positivity than those who delivered at term (50% versus 32%; P = 0.002); (iii) fetuses born to mothers with positive maternal HLA PRA results had a higher median serum CXCL10 concentration than those with negative HLA PRA results (P < 0.001); (iv) the median serum CXCL10 concentration (but not IL-6) was higher in fetuses with placental lesions associated with maternal anti-fetal rejection than those without such lesions (P < 0.001); (v) a whole-genome DASL assay of fetal blood RNA demonstrated differential expression of 128 genes between fetuses with and without lesions associated with maternal anti-fetal rejection; and (vi) comparison of the fetal serum proteome demonstrated 20 proteins whose abundance differed between fetuses with and without lesions associated with maternal anti-fetal rejection. CONCLUSION We describe a systemic inflammatory response in human fetuses born to mothers with evidence of maternal anti-fetal rejection. The transcriptome and proteome of this novel type of fetal inflammatory response were different from that of FIRS type I (which is associated with acute infection/inflammation).
Collapse
Affiliation(s)
- Joonho Lee
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, Detroit, MI, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Guo Y, Ma Z, Kou H, Sun R, Yang H, Smith CV, Zheng J, Wang H. Synergistic effects of pyrrolizidine alkaloids and lipopolysaccharide on preterm delivery and intrauterine fetal death in mice. Toxicol Lett 2013; 221:212-8. [PMID: 23831946 DOI: 10.1016/j.toxlet.2013.06.238] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2012] [Revised: 06/22/2013] [Accepted: 06/27/2013] [Indexed: 01/26/2023]
Abstract
Preterm birth is the leading cause of death for newborn infants, and lipopolysaccharide (LPS) is commonly used to induce preterm delivery in experimental animals. Pyrrolizidine alkaloids (PAs) are widespread and occur in foods, herbs, and other plants. This study was to investigate the synergistic effects of LPS and two representative PAs, retrorsine (RTS) and monocrotaline (MCT), on preterm delivery and fetal death. Pregnant Kunming mice were divided into seven groups: control, RTS, MCT, LPS, RTS+LPS and two MCT+LPS groups. Animals in PAs and PAs+LPS groups were dosed intragastrically with RTS (10mg/kg) or MCT (20 mg/kg or 60 mg/kg) from gestational day (GD) 9 to GD16; mice given LPS were injected intraperitoneally with 150 μg/kg on GD15.5. Latencies to delivery, numbers of pups live and dead at birth were recorded, and livers of live neonates were collected. The incidence of LPS-induced preterm birth was enhanced in dams pretreated with MCT, and combination of PAs and LPS increased fetal mortality from PAs. The enhancement of LPS-induced preterm delivery and fetal demise in animals exposed chronically to PAs and other substances found in foods and beverages consumed widely by humans merits further focused investigation.
Collapse
Affiliation(s)
- Yu Guo
- Department of Pharmacology, School of Basic Medical Science, Wuhan University, Wuhan 430071, China
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Kallapur SG, Presicce P, Senthamaraikannan P, Alvarez M, Tarantal AF, Miller LM, Jobe AH, Chougnet CA. Intra-amniotic IL-1β induces fetal inflammation in rhesus monkeys and alters the regulatory T cell/IL-17 balance. THE JOURNAL OF IMMUNOLOGY 2013; 191:1102-9. [PMID: 23794628 DOI: 10.4049/jimmunol.1300270] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Very low birth weight preterm newborns are susceptible to the development of debilitating inflammatory diseases, many of which are associated with chorioamnionitis. To define the effects of chorioamnionitis on the fetal immune system, IL-1β was administered intra-amniotically at ~80% gestation in rhesus monkeys. IL-1β caused histological chorioamnionitis, as well as lung inflammation (infiltration of neutrophils or monocytes in the fetal airways). There were large increases in multiple proinflammatory cytokine mRNAs in the lungs at 24 h postadministration, which remained elevated relative to controls at 72 h. Intra-amniotic IL-1β also induced the sustained expression of the surfactant proteins in the lungs. Importantly, IL-1β significantly altered the balance between inflammatory and regulatory T cells. Twenty-four hours after IL-1β injection, the frequency of CD3(+)CD4(+)FOXP3(+) T cells was decreased in lymphoid organs. In contrast, IL-17A-producing cells (CD3(+)CD4(+), CD3(+)CD4(-), and CD3(-)CD4(-) subsets) were increased in lymphoid organs. The frequency of IFN-γ-expressing cells did not change. In this model of a single exposure to an inflammatory trigger, CD3(+)CD4(+)FOXP3(+) cells rebounded quickly, and their frequency was increased at 72 h compared with controls. IL-17 expression was also transient. Interestingly, the T cell profile alteration was confined to the lymphoid organs and not to circulating fetal T cells. Together, these results suggest that the chorioamnionitis-induced IL-1/IL-17 axis is involved in the severe inflammation that can develop in preterm newborns. Boosting regulatory T cells and/or controlling IL-17 may provide a means to ameliorate these abnormalities.
Collapse
Affiliation(s)
- Suhas G Kallapur
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Yoo HS, Chang YS, Kim JK, Ahn SY, Kim ES, Sung DK, Jeon GW, Hwang JH, Shim JW, Park WS. Antenatal betamethasone attenuates intrauterine infection-aggravated hyperoxia-induced lung injury in neonatal rats. Pediatr Res 2013; 73:726-33. [PMID: 23493167 DOI: 10.1038/pr.2013.51] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Intrauterine infection can exacerbate postnatal hyperoxic lung injury. We hypothesized that antenatal betamethasone treatment attenuates hyperoxic lung injury aggravated by intrauterine infection in neonatal rats. METHODS Newborn Sprague-Dawley rats were divided into eight experimental groups according to (i) whether rats were exposed to normoxia (N) or hyperoxia (H, 85% oxygen) from postnatal day (P)1 to P14, (ii) whether antenatal betamethasone (0.2 mg/dose) or vehicle was administered to pregnant rats at gestation days (E)19 and E20, and (iii) whether intrauterine infection was induced or not antenatally. Intrauterine infection was induced by intracervical inoculation of Escherichia coli into pregnant rats on E19. We measured cytokine levels of tumor necrosis factor-α (TNF-α), interleukin (IL)-6, and IL-1β in P1 rat lungs and performed morphometric analyses and assessed inflammatory responses in lung tissue and bronchoalveolar lavage (BAL) at P14 by terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end labeling (TUNEL) staining and measurement of myeloperoxidase activity, collagen, and cytokine levels. RESULTS Cytokine levels in P1 rat lungs were increased by intrauterine infection, and these increases were attenuated by antenatal betamethasone. Hyperoxic lung injuries, indicated by morphometric changes and an inflammatory response in the lung and BAL fluid, were aggravated by intrauterine infection at P14. This aggravation was significantly attenuated by antenatal betamethasone. CONCLUSION Antenatal betamethasone attenuated aggravated hyperoxic lung injuries induced by intrauterine infection in neonatal rats via its anti-inflammatory actions.
Collapse
Affiliation(s)
- Hye Soo Yoo
- Department of Pediatrics, Samsung Medical Center, School of Medicine Sungkyunkwan University, Seoul, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Taylor BD, Holzman CB, Fichorova RN, Tian Y, Jones NM, Fu W, Senagore PK. Inflammation biomarkers in vaginal fluid and preterm delivery. Hum Reprod 2013; 28:942-52. [PMID: 23416276 PMCID: PMC3600841 DOI: 10.1093/humrep/det019] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Revised: 12/20/2012] [Accepted: 01/16/2013] [Indexed: 01/13/2023] Open
Abstract
STUDY QUESTION Which inflammation biomarkers detected in the vaginal fluid are most informative for identifying preterm delivery (PTD) risk? SUMMARY ANSWER Elevated interleukin (IL)-6 at mid-trimester was associated with increased odds of spontaneous PTD at <35 weeks and with PTD plus histologic chorioamnionitis (HCA), and had the greatest sensitivity for detecting these two PTD subtypes. WHAT IS KNOWN ALREADY Maternal and/or fetal inflammation play a role in some preterm deliveries, therefore inflammation biomarkers might help to identify women at greater risk. STUDY DESIGN, SIZE, DURATION We examined 1115 women from the Pregnancy Outcomes and Community Health Study, a cohort study conducted from September 1998 through June 2004, for whom data were available on mid-pregnancy inflammatory biomarkers. PARTICIPANTS/MATERIALS, SETTING, METHODS At enrollment at 16-27 weeks gestation, vaginal fluid samples were collected from a swab and 15 eluted biomarkers were measured using the Meso Scale Discovery multiplex electrochemiluminescence platform. Associations of biomarkers with PTD were examined, according to clinical circumstance, week at delivery and presence/absence of HCA. Weighted logistic regression was used to determine odds ratios (OR) and 95% confidence intervals (CI) adjusted for race. Sensitivity and specificity were compared between individual and multiple biomarkers, identified by a bootstrapping method. MAIN RESULTS AND THE ROLE OF CHANCE Elevated IL-6 (>75th percentile) displayed the strongest association with spontaneous PTD <35 weeks (OR 2.3; CI 1.3-4.0) and PTD with HCA (OR 2.8; CI 1.4-6.0). The sensitivity of IL-6 to detect spontaneous PTD <35 weeks or PTD with HCA was 0.43 and 0.51, respectively, while specificity was 0.74 and 0.75, respectively. IL-6 plus IL1β, IL-6r, tumor necrosis factor-alpha or granulocyte-macrophage colony-stimulating factor increased specificity (range 0.84-0.88), but decreased sensitivity (range 0.28-0.34) to detect both PTD subtypes. Results were similar when a combination of IL-6 and bacterial vaginosis (BV) was explored. Thus, the use of multiple biomarkers did not detect PTD subtypes with a greater sensitivity than IL-6 alone, and IL-6 is a specific but non-sensitive marker for the detection of spontaneous PTD. LIMITATIONS, REASONS FOR CAUTION Our ability to find small effect size associations between PTD and inflammation biomarkers (OR <2.0) might have been limited by the modest number of less common PTD subtypes in our population (e.g. spontaneous delivery <35 weeks, PTD accompanied by HCA) and by relatively higher variability for some cytokines, for example tumor necrosis factor-α, IL-12p70, IL-10 and granulocyte-macrophage colony-stimulating factor, that are less stable and commonly undetectable or detectable at low levels in human vaginal secretions. WIDER IMPLICATIONS OF THE FINDINGS Larger studies are needed to further explore a role of inflammation biomarkers in combination with other risk factors, including specific BV-associated organisms, for the prediction of PTD subtypes. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by the National Institute of Child Health and Human Development, National Institute of Nursing, March of Dimes Foundation, Thrasher Research Foundation and Centers for Disease Control and Prevention. The authors have no conflicts of interest.
Collapse
Affiliation(s)
- Brandie D Taylor
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, 130 Desoto Street, 5th floor Parran Hall, Pittsburgh, PA 15261, USA.
| | | | | | | | | | | | | |
Collapse
|
43
|
Natale F, Brunelli R, Bizzarri B, Castronovo A, De Curtis M. Cervical insufficiency: a new issue for guidelines on prevention of perinatal group B streptococcal disease? Pediatrics 2013; 131:e612-5. [PMID: 23296432 DOI: 10.1542/peds.2011-3677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The updated Guidelines on Prevention of Perinatal Group B Streptococcal Disease, issued by the Centers for Disease Control and Prevention, actually represent the mainstay in the prevention of neonatal early-onset group B streptococcal (GBS) sepsis. According to these guidelines, patients with possible preterm delivery are screened for GBS colonization and offered intrapartum prophylaxis only if they enter preterm labor or experience preterm premature rupture of the membranes. Nonetheless, the fulfillment of these recommendations seems to be suboptimal in clinical practice, as it is heavily influenced by the knowledge of the colonization status. We report here 2 cases of blood culture-proven, early-onset neonatal GBS sepsis involving preterm infants delivered by mothers who had midtrimester cervical insufficiency and bulging membranes. Midtrimester acute cervical insufficiency strongly predicts preterm delivery. These women are liable to miss intrapartum antibiotic prophylaxis because they typically have shorter labor, and the test results for GBS status are unlikely to be available before delivery. We believe that women with midtrimester cervical insufficiency and bulging membranes should be screened for GBS infection soon after hospital admittance if the gestational age is close to the threshold of fetal viability. A timely diagnosis of GBS colonization may not only increase the number of patients receiving targeted intrapartum antibiotic prophylaxis but would also allow consideration of the administration of antepartum antibiotic prophylaxis. Indeed, as further outlined in this report, GBS intraamniotic infection may dramatically occur before the onset of preterm labor or preterm premature rupture of the membranes.
Collapse
Affiliation(s)
- Fabio Natale
- Neonatal ICU, Department of Pediatrics and Child Neuropsychiatry, "Sapienza" University of Rome, V.le Regina Elena 324-00161, Rome, Italy.
| | | | | | | | | |
Collapse
|
44
|
Grigsby PL, Novy MJ, Sadowsky DW, Morgan TK, Long M, Acosta E, Duffy LB, Waites KB. Maternal azithromycin therapy for Ureaplasma intraamniotic infection delays preterm delivery and reduces fetal lung injury in a primate model. Am J Obstet Gynecol 2012; 207:475.e1-475.e14. [PMID: 23111115 DOI: 10.1016/j.ajog.2012.10.871] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Revised: 07/27/2012] [Accepted: 10/15/2012] [Indexed: 12/19/2022]
Abstract
OBJECTIVE We assessed the efficacy of a maternal multidose azithromycin (AZI) regimen, with and without antiinflammatory agents to delay preterm birth and to mitigate fetal lung injury associated with Ureaplasma parvum intraamniotic infection. STUDY DESIGN Long-term catheterized rhesus monkeys (n = 16) received intraamniotic inoculation of U parvum (10(7) colony-forming U/mL, serovar 1). After contraction onset, rhesus monkeys received no treatment (n = 6); AZI (12.5 mg/kg, every 12 h, intravenous for 10 days; n = 5); or AZI plus dexamethasone and indomethacin (n = 5). Outcomes included amniotic fluid proinflammatory mediators, U parvum cultures and polymerase chain reaction, AZI pharmacokinetics, and the extent of fetal lung inflammation. RESULTS Maternal AZI therapy eradicated U parvum intraamniotic infection from the amniotic fluid within 4 days. Placenta and fetal tissues were 90% culture negative at delivery. AZI therapy significantly delayed preterm delivery and prevented advanced fetal lung injury, although residual acute chorioamnionitis persisted. CONCLUSION Specific maternal antibiotic therapy can eradicate U parvum from the amniotic fluid and key fetal organs, with subsequent prolongation of pregnancy, which provides a therapeutic window of opportunity to effectively reduce the severity of fetal lung injury.
Collapse
Affiliation(s)
- Peta L Grigsby
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Beaverton, OR, USA.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Buhimschi IA, Nayeri UA, Laky CA, Razeq SA, Dulay AT, Buhimschi CS. Advances in medical diagnosis of intra-amniotic infection. ACTA ACUST UNITED AC 2012; 7:5-16. [PMID: 23530840 DOI: 10.1517/17530059.2012.709232] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Intrauterine infection is a global problem and a significant contributor to morbidity and perinatal death. The host response to infection causes an inflammatory state that acts synergistically with microbial insult to induce preterm birth and fetal damage. Prompt and accurate diagnosis of intra-amniotic infection in the asymptomatic stage of the disease is critical for improved maternal and neonatal outcomes. AREAS COVERED This article provides an overview of the most recent progress, challenges, and opportunities for discovery and clinical implementation of various maternal serum, cervicovaginal, and amniotic fluid biomarkers in pregnancies complicated by intra-amniotic infection. EXPERT OPINION Clinically relevant biomarkers are critical to the accurate diagnostic of intrauterine infection. Front-end implementation of such biomarkers will also translate in lower incidence of early-onset neonatal sepsis (EONS) which is an important determinant of neonatal morbidity and mortality associated with prematurity. However, of the hundreds of differentially expressed proteins, only few may have clinical utility and thus function as biomarkers. The small number of validation studies along with barriers to implementation of technological innovations in the clinical setting are current limitations.
Collapse
Affiliation(s)
- Irina A Buhimschi
- Yale University, Department of Obstetrics, Gynecology & Reprod. Science, New Haven, CT 06520, USA.
| | | | | | | | | | | |
Collapse
|
46
|
Kemp MW, Saito M, Kallapur SG, Jobe AH, Keelan JA, Li S, Kramer B, Zhang L, Knox C, Yaegashi N, Newnham JP. Inflammation of the fetal ovine skin following in utero exposure to Ureaplasma parvum. Reprod Sci 2012; 18:1128-37. [PMID: 22031190 DOI: 10.1177/1933719111408114] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
There is increasing evidence linking in utero infection and inflammation to preterm birth. Many commensal urogenital tract microorganisms, including the Mycoplasmas and Ureaplasmas, are commonly detected in association with preterm birth. Using an ovine model of sterile fetal inflammation, we demonstrated previously that the fetal skin generates a robust inflammatory response following in utero exposure to lipopolysaccharides from Escherichia coli. The fetal skin's response to colonization of the amniotic fluid by viable microorganisms remains unstudied. We hypothesised that in utero infection with Ureaplasma parvum serovar 3 would induce a proinflammatory response in the fetal skin. We found that (1) cultured fetal keratinocytes (the primary cellular constituent of the epidermis) respond to U. parvum exposure in vitro by increasing the expression of the chemotactant monocyte chemoattractant protein 1 (MCP-1) but not interleukin 1β (IL-1β), IL-6, IL-8, or tumor necrosis factor-α (TNF-α); (2) the fetal skin's response to 7 days of U. parvum exposure is characterized by elevated expression of MCP-1, TNF-α, and IL-10; and (3) the magnitude of inflammatory cytokine/chemokine expression in the fetal skin is dependent on the duration of U parvum exposure. These novel findings provide further support for the role of the fetal skin in the development of fetal inflammation and the preterm birth that may follow.
Collapse
Affiliation(s)
- Matthew W Kemp
- School of Women's and Infants' Health, The University of Western Australia, Perth, Australia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Williams O, Hutchings G, Hubinont C, Debauche C, Greenough A. Pulmonary effects of prolonged oligohydramnios following mid-trimester rupture of the membranes--antenatal and postnatal management. Neonatology 2012; 101:83-90. [PMID: 21934333 DOI: 10.1159/000329445] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Accepted: 05/15/2011] [Indexed: 11/19/2022]
Abstract
Mid-trimester, preterm prelabour rupture of the membranes (PPROM) with prolonged oligohydramnios remains a challenge for both obstetricians and neonatologists. Although survival rates have improved, morbidity remains common particularly due to pulmonary insufficiency and pulmonary hypertension. The aetiology of abnormal lung development is unknown but may depend critically on pulmonary vascular development. Antenatal evaluation of at-risk foetuses by three-dimensional ultrasound and MRI is possible but the techniques need to be further assessed. Antenatal corticosteroids given in cases of PPROM reduce the incidence of neonatal death, respiratory distress syndrome, intraventricular haemorrhage and necrotising enterocolitis without increasing maternal or neonatal infection. The true risk-benefit ratio of antibiotics, tocolysis and strategies to normalise amniotic fluid volume remains less clear. There is no consensus regarding the optimal ventilation strategy to support infants with pulmonary insufficiency following PPROM, and further work is required to determine whether and which pulmonary vasodilators improve long-term outcome in these infants.
Collapse
Affiliation(s)
- Olivia Williams
- Department of Neonatology, Cliniques Universitaires Saint Luc, Université Catholique de Louvain, Brussels, Belgium.
| | | | | | | | | |
Collapse
|
48
|
Adams Waldorf KM, Gravett MG, McAdams RM, Paolella LJ, Gough GM, Carl DJ, Bansal A, Liggitt HD, Kapur RP, Reitz FB, Rubens CE. Choriodecidual group B streptococcal inoculation induces fetal lung injury without intra-amniotic infection and preterm labor in Macaca nemestrina. PLoS One 2011; 6:e28972. [PMID: 22216148 PMCID: PMC3244436 DOI: 10.1371/journal.pone.0028972] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Accepted: 11/18/2011] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Early events leading to intrauterine infection and fetal lung injury remain poorly defined, but may hold the key to preventing neonatal and adult chronic lung disease. Our objective was to establish a nonhuman primate model of an early stage of chorioamnionitis in order to determine the time course and mechanisms of fetal lung injury in utero. METHODOLOGY/PRINCIPAL FINDINGS Ten chronically catheterized pregnant monkeys (Macaca nemestrina) at 118-125 days gestation (term=172 days) received one of two treatments: 1) choriodecidual and intra-amniotic saline (n=5), or 2) choriodecidual inoculation of Group B Streptococcus (GBS) 1×10(6) colony forming units (n=5). Cesarean section was performed regardless of labor 4 days after GBS or 7 days after saline infusion to collect fetal and placental tissues. Only two GBS animals developed early labor with no cervical change in the remaining animals. Despite uterine quiescence in most cases, blinded review found histopathological evidence of fetal lung injury in four GBS animals characterized by intra-alveolar neutrophils and interstitial thickening, which was absent in controls. Significant elevations of cytokines in amniotic fluid (TNF-α, IL-8, IL-1β, IL-6) and fetal plasma (IL-8) were detected in GBS animals and correlated with lung injury (p<0.05). Lung injury was not directly caused by GBS, because GBS was undetectable in amniotic fluid (~10 samples tested/animal), maternal and fetal blood by culture and polymerase chain reaction. In only two cases was GBS cultured from the inoculation site in low numbers. Chorioamnionitis occurred in two GBS animals with lung injury, but two others with lung injury had normal placental histology. CONCLUSIONS/SIGNIFICANCE A transient choriodecidual infection can induce cytokine production, which is associated with fetal lung injury without overt infection of amniotic fluid, chorioamnionitis or preterm labor. Fetal lung injury may, thus, occur silently without symptoms and before the onset of the fetal systemic inflammatory response syndrome.
Collapse
|
49
|
Effects of urocortin 2 and urocortin 3 on IL-10 and TNF-α expression and secretion from human trophoblast explants. Placenta 2011; 32:969-74. [DOI: 10.1016/j.placenta.2011.09.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Revised: 09/18/2011] [Accepted: 09/21/2011] [Indexed: 01/26/2023]
|
50
|
Abstract
Much emphasis in recent decades has been devoted to inflammation and infection as a premier causal mechanism of preterm birth. This article explores the epidemiologic, clinical, and animal data that exist to support this conceptual paradigm as well as proposed mechanisms through which to potentially mitigate the adversity of prematurity. Truly successful interventions are not likely to occur until the pathogenesis of preterm birth and the role of inflammation in causing not only parturition but also fetal and neonatal injury is fully elucidated.
Collapse
Affiliation(s)
- Jamie A Bastek
- Maternal and Child Health Research Program, Philadelphia, PA, USA
| | | | | |
Collapse
|