Skip to main content
Reproductive Sciences logoLink to Reproductive Sciences
. 2016 Nov 24;24(8):1139–1153. doi: 10.1177/1933719116678690

Neutrophil Extracellular Traps in the Amniotic Cavity of Women with Intra-Amniotic Infection: A New Mechanism of Host Defense

Nardhy Gomez-Lopez 1,2,3,, Roberto Romero 1,4,5,6,, Yi Xu 1,2, Derek Miller 1,2,3, Ronald Unkel 1,2, Majid Shaman 1,2, Suzanne M Jacques 1,7, Bogdan Panaitescu 1,2, Valeria Garcia-Flores 1,2, Sonia S Hassan 1,2
PMCID: PMC6343453  PMID: 27884950

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.

Keywords: cytokines, fetal inflammatory response, fever, funisitis, inflammation, intra-amniotic inflammation, labor, microbial invasion of the amniotic cavity (MIAC), neutrophil, parturition, pregnancy, preterm birth, preterm labor

Introduction

Intra-amniotic inflammation can be due to microorganisms (ie, intra-amniotic infection) or danger signals derived from necrosis and cellular stress (ie, sterile intra-amniotic inflammation).1-6 Intra-amniotic infection is frequently caused by Ureaplasma urealyticum, Gardnerella vaginalis, and other commensal organisms found in the lower genital tract.7-10 Microbial invasion of the amniotic cavity (MIAC) can elicit local and systemic inflammatory responses.11-15 Systemic maternal inflammation results in clinical chorioamnionitis, which refers to the presence of maternal fever associated with clinical signs (ie, foul-smelling discharge, uterine tenderness, and maternal and fetal tachycardia) as well as laboratory abnormalities such as leukocytosis.16,17

In humans, intra-amniotic infection is associated with a local inflammatory response,18 which is characterized by an increased amniotic fluid white blood cell (WBC) count19-23 and elevated concentrations of inflammatory mediators, such as cytokines18,24 and lipids (eg, prostaglandins).25-38 In nonhuman primates, intra-amniotic infection also results in a local inflammatory response.39-48 Neutrophils are the most abundant leukocytes in the amniotic fluid in cases of intra-amniotic infection.19 These innate immune cells seem to be fetal49 and originate from the chorionic plate;50 yet, further evidence is required to prove their fetal origin using genetic fingerprinting. Recently, using immunophenotyping, we demonstrated that amniotic fluid neutrophils express proinflammatory cytokines such as tumor necrosis factor-α, macrophage inflammatory protein (MIP)-1α, MIP-1β, interleukin (IL)-1α, IL-1β, and IL-8.51 These cytokines and chemokines have been implicated in the processes responsible for term and preterm parturition.24,52-74

Neutrophils are the first line of defense against invading pathogens and possess an arsenal of weapons utilized in the elimination of microbes.75 These innate immune cells are primarily phagocytes, capable of enveloping and killing microbes through the initiation of NADPH oxidase activity, which leads to the release of reactive oxygen species such as peroxide, superoxide, and hydroxyl radical.76,77 Neutrophils carry granules filled with enzymes such as myeloperoxidase, cathepsin G, elastase, and proteinase 3, which can be injected into the phagosome or released externally, leading to bacterial killing.75,76

Neutrophils can also undergo a specialized cell death termed neutrophil extracellular traps (NETs),78,79 which represents the final containment effort of a neutrophil to lyse pathogens.80 NETs are web-like structures composed of DNA, histones, and antimicrobial products such as neutrophil elastase.78-80 These traps eliminate microbes through their biochemical components.81 Histones (∼ 50% of the extracellular trap82) use their cationic properties to kill bacteria by permeabilizing microbial membranes.81 Extracellular DNA also has microbicidal properties as it is a powerful chelator of divalent cations.83 Therefore, the main function of NETs in immune host defense is to prevent infection by trapping and killing bacteria. The aim of this study was to investigate whether neutrophils in the amniotic cavity of women with intra-amniotic infection are capable of forming NETs.

Materials and Methods

Study Population

This was a cross-sectional study of patients who underwent transabdominal amniocentesis due to clinical indications or amniocentesis during cesarean section (Tables 1 and 2). Patients were enrolled at Hutzel Women’s Hospital of the Detroit Medical Center (Detroit, Michigan). The inclusion criteria were (1) singleton gestations and (2) sufficient viable cells (>1 × 105 cells/mL) in amniotic fluid samples. Viable cell numbers were determined using an automatic cell counter (Cellometer Auto 2000; Nexcelom Bioscience, Lawrence, Massachusetts).

Table 1.

Demographic and Clinical Characteristics of Samples Used in This Study.

Term Delivery (n = 9) Preterm Delivery (n = 6) P Value
Maternal age, years; median (IQR)a 21 (19-22) 25.5 (24.3-32.8) .028
Body mass index, kg/m2; median (IQR)a 25.6 (23.7-30.2) 33.3 (27.3-35.8) NS
Gestational age at delivery, week; median (IQR)a 39.9 (39.3-40.3) 24 (23.3-29.3) .001
Birth weight, g; median (IQR)a 3440 (3385-3635) 630 (552.5-1435) .01
Race, n (%)b NS
 African American 8 (88.9) 6 (100)
 Caucasian 0 (0) 0 (0)
 Hispanic 0 (0) 0 (0)
 Asian 1 (11.1) 0 (0)
 Other 0 (0) 0 (0)
Labor, n (%)b 9 (100) 5 (83.3) NS
Primiparity, n (%)b 5 (55.6) 1 (16.7) NS
Cesarean section, n (%)b 6 (66.7) 2 (33.3) NS
Acute maternal inflammatory response, n (%)b
 Stage 1 (acute subchorionitis) 1 (11.1) 0 (0) NS
 Stage 2 (acute chorioamnionitis) 4 (44.4) 1 (16.7) NS
 Stage 3 (acute necrotizing chorioamnionitis) 4 (44.4) 5 (83.3) NS
Acute fetal inflammatory response, n(%)b
 Stage 1 (acute phlebitis/chorionic vasculitis) 4 (44.4) 1 (16.7) NS
 Stage 2 (acute arteritis) 5 (55.6) 3 (50) NS
 Stage 3 (necrotizing funisitis) 0 (0) 2 (33.3) NS

Abbreviation: IQR, interquartile range.

aWilcoxon rank sum test.

bFisher’s exact test.

Table 2.

Clinical Characteristics of Amniotic Fluid Samples Utilized in This Study.

Sample Clinical Chorioamnionitis Viable Cell Counta (cells/mm3) Gestational Age at Amniocentesis Collection Method of Amniotic Fluid IL-6 (ng/mL) Gram Stain Amniotic Fluid Culture WBC Count (cells/mm3) Glucose (mg/dL) Gestational Age at Delivery Bacterial Live/Dead Staining Delivery Method
1 Yes 1080 39.9 Transabdominal 87.0 Negative Ureaplasma subspecies, Streptococcus anginosus 7183 <1 39.9 Positive C/S
2 Yes 3570 40.1 Transabdominal 32.3 Gram-positive cocci, Gram-variable bacilli Ureaplasma subspecies, Peptostreptococcus subspecies, Viridans streptococci, Enterococcus faecalis 3000 <1 40.3 Positive C/S
3 Yes 927 39.7 Transabdominal 30.3 Gram-positive cocci, Gram-positive bacilli Ureaplasma subspecies, S agalactiae 530 <1 39.7 Positive Vaginal
4 Yes 1040 40.9 Transabdominal 113.5 Gram-negative bacilli Ureaplasma subspecies, Mycoplasma subspecies, Fusobacterium subspecies 878 <1 40.9 Positive Vaginal
5 Yes 1100 37.7 Transabdominal 16.9 Negative Lactobacillus subspecies, Peptostreptococcus subspecies, Gardnerella vaginalis, S anginosus 900 7 37.7 Positive Vaginal
6 Yes 996 22.7 Transabdominal 334.9 Negative Fusobacterium subspecies 463 <1 23.3 Positive C/S
7 Yes 697 24.6 Transabdominal 259.4 Gram-negative bacilli Escherichia coli 609 <1 24.6 Positive Vaginal
8 No 1430 30.7 Transabdominal 60.0 Negative Ureaplasma subspecies, Mycoplasma subspecies 1400 12 30.9 Positive Vaginal
9 Yes 2220 36.6 Transabdominal 8.1 Gram-positive cocci S agalactiae 310 <1 36.7 Positive Vaginal
10 Yes 508 23.1 Transabdominal 121.0 Gram-negative bacilli Ureaplasma subspecies, G vaginalis, Bacteroides ureolyticus 750 <1 23.4 Positive C/S
11 Yes 468 20.7 Transabdominal 122.1 Gram-positive cocci Ureaplasma subspecies, S mitis, Eikenella corrodens 1150 17 20.7 Positive Vaginal
12 Yes 11 100 39.3 C/S 106.9 NA NA NA NA 39.3 Positive C/S
13 Yes 2830 37.4 C/S 237.0 NA NA NA NA 37.4 Positive C/S
14 No 26 800 40.6 C/S 120.8 NA NA NA NA 40.6 Positive C/S
15 No 206 40.1 C/S 16.7 NA NA NA NA 40.1 Positive C/S

Abbreviations: CS, cesarean section; IL, interleukin; NA, not available; WBC, white blood cell.

aViable cell count: determined with AO/PI on Cellometer Auto 2000 (Nexcelom).

Maternal and neonatal data were obtained by retrospective clinical chart review. The information retrieved included the following: use of epidural analgesia, intrapartum antibiotic administration, membrane status at the time of amniocentesis (intact or ruptured), and mode of delivery. Patients with the diagnosis of clinical chorioamnionitis (see diagnostic criteria in the subsequent section) were counseled by their treating physicians about the potential value of knowing the precise microorganism(s) involved in the suspected infection. Further management of these patients was at the discretion of the attending physician.

All patients provided written informed consent to donate additional amniotic fluid for research purposes, according to protocols approved by the Institutional Review Boards of the Detroit Medical Center, Wayne State University, and the Perinatology Research Branch, an intramural program of the Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS).

Clinical Definitions

Gestational age was determined by the last menstrual period and confirmed by ultrasound examination. The gestational age derived from sonographic fetal biometry was used if the estimation was inconsistent with menstrual dating. Clinical chorioamnionitis was diagnosed by the presence of maternal fever accompanied by 2 or more of the following criteria: (1) uterine tenderness, (2) malodorous vaginal discharge, (3) fetal tachycardia (heart rate > 160 beats/min), (4) maternal tachycardia (heart rate > 100 beats/min), and (5) maternal leukocytosis (leukocyte count >15 000 cells/mm3).11,84-86 Labor at term was defined as the presence of regular uterine contractions with a frequency of at least 1 every 10 minutes and cervical change after 37 weeks of gestation. Preterm labor was diagnosed by the presence of regular uterine contractions (at least 3 in 30 minutes) and documented cervical changes in patients with a gestational age between 20 and 36 (6/7) weeks.

Microbial invasion of the amniotic cavity was defined as a positive amniotic fluid culture.87-91 Intra-amniotic inflammation was diagnosed when the IL-6 concentration in amniotic fluid was ≥ 2.6 ng/mL.92,93 Intra-amniotic infection was defined as the presence of MIAC with intra-amniotic inflammation.3-6,10,92-102 Acute histologic chorioamnionitis was diagnosed based on the presence of inflammatory cells in the chorionic plate and/or chorioamniotic membranes,103-106 whereas acute funisitis was diagnosed by the presence of neutrophils in the wall of the umbilical vessels and/or Wharton’s jelly, using previously described criteria.103,105,107-110

Sample Collection

Amniotic fluid was retrieved by transabdominal amniocentesis under antiseptic conditions using a 22-gauge needle monitored by ultrasound. Amniotic fluid was also retrieved by amniocentesis during cesarean section under antiseptic conditions. Amniotic fluid samples were transported to the clinical laboratory in a capped sterile syringe and were cultured for aerobic and anaerobic bacteria as well as for genital mycoplasmas.10,19,111-114 Shortly after collection, the WBC count in amniotic fluid samples was determined using a hemocytometer chamber, according to previously described methods.19 Glucose concentration115 was also determined and Gram stain116 was performed in amniotic fluid samples. Cultures, WBC count, glucose concentration, and Gram Stain were not performed in those amniotic fluid samples collected during cesarean section, as these samples were collected for research purposes only. However, both IL-6 concentration and the presence of bacteria (bacterial live/dead staining) were assessed in all of the amniotic fluid samples.

Determination of IL-6 in Amniotic Fluid

Interleukin-6 concentrations in amniotic fluid samples were determined using a sensitive and specific enzyme immunoassay obtained from R&D Systems (Minneapolis, Minnesota). IL-6 concentrations were determined by interpolation from the standard curve. The interassay and intraassay coefficients of variation for IL-6 were 8.7% and 4.6%, respectively. The detection limit of the IL-6 assay was 0.09 pg/mL.

Detection of NETs by DNA Staining

NETs were discovered when isolated peripheral neutrophils were stimulated with phorbol-12-myristate-13-acetate (PMA) in vitro.78 Therefore, we used maternal peripheral neutrophils as positive controls. Peripheral blood samples were collected by venipuncture into EDTA-containing tubes from pregnant women at term in the absence of labor (n = 3). Neutrophils were isolated using the density gradient reagent Histopaque 1119 (Sigma-Aldrich; St. Louis, Missouri), according to the manufacturer’s instructions and a previously published method.117 Briefly, 6 mL of peripheral blood were layered on top of 6 mL of Histopaque 1119 and centrifuged at 800g for 20 minutes with no break at room temperature. Neutrophils were collected from the lower phase of the gradient after the peripheral blood mononuclear cell band was discarded. The collected neutrophils were further purified using a gradient composed of 85%, 80%, 75%, 70%, and 65% Percoll (GE Healthcare Life Sciences; Uppsala, Sweden) and washed with 1X PBS (Life Technologies; Grand Island, New York). Purified neutrophils were then resuspended in RPMI-1640 culture medium supplemented with 10% FBS and 1% penicillin/streptomycin (Life Technologies; hereafter referred to as “supplemented RPMI”). Neutrophils were incubated in 24-well culture plates (Corning Life Sciences, Durham, North Carolina) containing 12-mm cover slips (Fisher Scientific, Waltham, Massachusetts) at a concentration of 2 × 105 cells/0.5 mL and incubated for 1 hour at 37°C with 5% CO2. Following incubation, adherent neutrophils were stimulated using 100 nM of PMA (Sigma-Aldrich) for 2 hours at 37°C with 5% CO2. Next, paraformaldehyde (PFA; Electron Microscopy Science, Hatfield, Pennsylvania) was added to each culture plate well at a final concentration of 4% for 2 hours. The cover slips were then carefully removed from the culture plate well, rinsed with 1X PBS, and mounted onto Fisherbrand Superfrost Plus microscope slides (Thermo Scientific, Wilmington, Delaware) using ProLong Diamond Antifade Mountant with 4′,6-diamidino-2-phenylindole (DAPI; Life Technologies). Images were acquired using an Olympus BX60 fluorescence microscope (Olympus Corporation, Tokyo, Japan) equipped with an Olympus DP71 camera and DP Controller Software (Olympus Corporation).

Amniotic fluid samples were passed through a sterile 15-μm filter (Cat# 43-50015-03; pluriSelect Life Science, Leipzig, Germany) and centrifuged at 200g for 5 minutes at room temperature (n = 15). Amniotic fluid cells were then resuspended in supplemented RPMI at a concentration of 2.5 × 105 cells/0.5 mL and placed in 24-well culture plates containing 12-mm cover slips. Next, cells were incubated for 1 hour at 37°C with 5% CO2, stimulated with 100 nM of PMA for 2 hours, and fixed with 4% PFA for 2 hours. Finally, cover slips were carefully removed, rinsed with 1X PBS, and mounted onto Fisherbrand Superfrost Plus microscope slides using ProLong Diamond Antifade Mountant with DAPI. Images were acquired using an Olympus BX60 fluorescence microscope equipped with an Olympus DP71 camera and DP Controller Software.

Detection of Bacteria in Amniotic Fluid Samples

Staining of bacteria in amniotic fluid samples (n = 15) was performed as previously described,118 using the LIVE/DEAD BacLight Bacterial Viability Kit (Cat# L7007; Life Technologies) in a sterile biosafety cabinet. Briefly, 100 μL of amniotic fluid were mixed with 900 μL of sterile 1X PBS. Three microliters of the dye mix (component A and B were mixed at a 1:1 ratio) were added to the cell suspension and incubated for 15 minutes at room temperature in the dark. Next, the cells were centrifuged at 10 000g for 5 minutes, and the supernatant was discarded. The cell pellet was resuspended in 5 μL of 1X PBS, and a slide smear was prepared and air-dried. Finally, the slide was gently rinsed with 1X PBS and mounted with ProLong Diamond Antifade Mountant with DAPI. Images were acquired using an Olympus BX 60 fluorescence microscope with an Olympus DP71 camera and DP Controller Software.

Scanning Electron Microscopy

Neutrophils were isolated from maternal blood and amniotic fluid samples and then cultured on cover slips, as described above (n = 3 each). After PMA stimulation, the culture medium was aspirated and electron microscopy fixative (2.5% glutaraldehyde in 0.1 M phosphate buffer, pH 7.4; Cat# 16537-05, Electron Microscopy Science) was carefully added to the culture plate wells. Following fixation for 2 hours at 4°C, the cover slips were gently washed with 1X electron microscopy wash buffer (Sorensen’s phosphate buffer 0.2 M, pH 7.4; Cat# 11601-10, Electron Microscopy Science). The cover slips were then stored in 1X electron microscopy wash buffer and transported to the Microscopy & Image Analysis Laboratory at the University of Michigan (Ann Arbor, Michigan) (https://medicine.umich.edu/medschool/research/office-research/biomedical-research-core-facilities/microscopy-image-analysis). Images were obtained using an AMRAY 1910 Field Emission Scanning Electron Microscope (SEMTechSolutions, North Billerica, Massachusetts).

Confocal Microscopy

Neutrophils from amniotic fluid samples were cultured on cover slips, as described above (n = 3). After PFA fixation, cells were washed with 1X PBS, permeabilized with 1X PBS containing 0.25% Triton X-100 (Cat# H5141; Promega, Madison, Wisconsin) for 2 minutes, and then washed with 1X PBS again. Nonspecific antibody interactions were blocked by treating the cover slips with Dako Protein Block Serum-Free Solution (catalog number X0909; DakoCytomation, Carpinteria, California) for 30 minutes at room temperature. The cover slips were incubated for 1 hour at room temperature with a mouse anti-human neutrophil elastase (Cat# M0752; clone NP57, Dako, Denmark) and a rabbit anti-human H3 antibody (Cat# ab5103; Abcam, Cambridge, Massachusetts), and then washed with 1X PBS. Next, a second blocking step was performed by adding 10% goat serum (KPL, Gaithersburg, Maryland) for 10 minutes. The cover slips were incubated with a secondary goat anti-mouse IgG–Alexa Fluor 488 antibody (Cat# A11029; Life Technologies) and a goat anti-rabbit IgG–Alexa Fluor 594 antibody (Cat#A11072; Life Technologies) for 1 hour at room temperature in the dark. Finally, the cover slips were washed with 1X PBS and mounted onto Fisherbrand Superfrost Plus microscope slides using ProLong Diamond Antifade Mountant with DAPI. Slides were visualized on a Zeiss LSM 780 laser scanning confocal microscope (Carl Zeiss Microscopy GmbH, Jena, Germany) at the Microscopy, Imaging, and Cytometry Resources Core at the Wayne State University School of Medicine (Detroit, Michigan) (http://micr.med.wayne.edu/). Confocal Z stacks were acquired using a Plan-Apochromat 100×/1.40 Oil DIC lens with 1.5× digital zoom. Immunofluorescence signals for Alexa Fluor 594 and Alexa Fluor 488 were excited with an In Tune White Light laser tuned to 595 nm and a 488-nm line Multiline Argon laser, respectively. The DAPI signal was excited with a 405-nm diode laser.

Determination of NET Formation by Maternal and Neonatal Neutrophils Incubated with the Amniotic Fluid of Women without Intra-Amniotic Infection

Maternal or neonatal neutrophils were isolated from peripheral or cord blood, respectively, using density gradients and cultured on cover slips, as described above. Five hundred microliters of amniotic fluid from 4 singleton term pregnancies without labor or infection/inflammation (Gram stain negative, negative cultures, WBC count = 0, and IL-6 < 2.6 ng/mL) were added to the cultured neutrophils and incubated for 2 hours at 37°C with 5% CO2. Positive controls included maternal or neonatal neutrophils stimulated with PMA, as described above. Next, PFA was added to each culture plate well at a final concentration of 4% for 2 hours. The cover slips were then carefully removed from the culture plate well, rinsed with 1X PBS, and mounted onto Fisherbrand Superfrost Plus microscope slides using ProLong Diamond Antifade Mountant with DAPI. Images were acquired using an Olympus BX60 fluorescence microscope equipped with an Olympus DP71 camera and DP Controller Software.

Results

Characteristics of the Study Population

A total of 15 amniotic fluid samples from women who underwent transabdominal amniocentesis before delivery or during cesarean section were included in this study. Demographic and clinical characteristics of the study population are displayed in Table 1. All of the patients were diagnosed with intra-amniotic inflammation as they had elevated concentrations of IL-6 in the amniotic fluid (≥ 2.6 ng/mL92,93; Table 2). Nine patients underwent spontaneous labor at term (Table 1), of which 7 were diagnosed with clinical chorioamnionitis (Table 2). Five patients underwent spontaneous preterm labor and birth, and 1 patient delivered preterm in the absence of labor (Table 1). Five of the 6 patients who delivered preterm were diagnosed with clinical chorioamnionitis (Table 2). All of the patients who underwent amniocentesis for clinical purposes were diagnosed with intra-amniotic infection as they had positive cultures (ie, MIAC) and elevated concentrations of IL-6 in the amniotic fluid (≥ 2.6 ng/mL3-6,10,92-102; Table 2). All of these patients also had elevated WBC counts (≥ 50 cells/mm3)19 in the amniotic fluid (Table 2). Most of the patients (10/11) had low glucose concentrations (< 14 mg/dL)115 in the amniotic fluid (Table 2). The most common microorganisms were Ureaplasma subspecies followed by Streptococcus subspecies (Table 2).

The First Observation of NETs in the Amniotic Cavity of Women with Intra-Amniotic Inflammation

First, we investigated whether NETs were detectable in amniotic fluid samples from women with intra-amniotic inflammation. As a positive control, we stimulated neutrophils from maternal blood with PMA and observed the appearance of NETs using DAPI staining, which detects DNA.119,120 Figure 1A and 1B demonstrate that incubation of maternal neutrophils with PMA induces the formation of NETs.78,117 However, amniotic fluid neutrophils formed NETs in the absence of PMA stimulation (Figure 1C). When amniotic fluid neutrophils were stimulated with PMA, no evident increase in NET formation was observed.

Figure 1.

Figure 1.

DAPI staining of NETs in maternal peripheral blood and the amniotic fluid. Representative fluorescence microscopy images of (A) unstimulated maternal neutrophils, (B) PMA-stimulated maternal neutrophils, (C) unstimulated amniotic fluid neutrophils, and (D) PMA-stimulated amniotic fluid neutrophils. DNA is blue (DAPI). Magnification 400×. DAPI indicates 4′,6-diamidino-2-phenylindole; NET, neutrophil extracellular trap; PMA, phorbol-12-myristate-13-acetate.

Detection of NETs and Bacteria in the Amniotic Fluid

In order to investigate whether the presence of bacteria in the amniotic fluid was associated with the production of NETs, we utilized bacterial Live/Dead staining combined with DAPI staining.118 All of the amniotic fluid samples had detectable live and dead bacteria (Table 2). It is worth mentioning that the bacterial Live/Dead staining detected Ureaplasma subspecies and Mycoplasma subspecies in sample #8 (Table 2); yet, further validation of this assay is required to identify bacteria without a cell wall in the amniotic fluid. Figure 2 is a representative image of live and dead bacteria trapped by amniotic fluid NETs. These data demonstrate that all of the patients had intra-amniotic infection, and more importantly, that NETs are associated with MIAC.

Figure 2.

Figure 2.

Bacterial Live/Dead staining and DAPI staining of an amniotic fluid sample showing bacteria trapped by NETs. Separated layers show (A) DAPI staining, (B) SYTO 9 staining, (C) propidium iodide (PI) staining, and (D) a merged image. Live bacteria with intact cell membranes fluoresce green and dead bacteria with compromised membranes fluoresce red. DNA is blue (DAPI). Magnification 1000×. DAPI indicates 4′,6-diamidino-2-phenylindole; NET, neutrophil extracellular trap.

Visualization of NETs by Scanning Electron Microscopy

We further characterized the appearance of NETs in the amniotic fluid using scanning electron microscopy. Maternal peripheral neutrophils without PMA stimulation maintained a classic round morphology typical of quiescent cells80,121-123 (Figure 3A and B). Maternal peripheral neutrophils stimulated with PMA appeared amorphous and distended, with filamentous projections, possibly DNA, being released (Figure 3C and D).80,123 The morphology of amniotic fluid neutrophils resembled PMA-stimulated maternal neutrophils (Figure 3E and F). Yet, the amniotic fluid neutrophils appeared flatter and more distended without the filamentous projections observed in PMA-stimulated maternal neutrophils (Figure 3E vs. C).

Figure 3.

Figure 3.

Scanning electron microscopy of neutrophils from maternal peripheral blood and amniotic fluid samples. Unstimulated maternal neutrophils were captured at magnifications (A) 1000× and (B) 5000×, PMA-stimulated maternal neutrophils were captured at magnifications (C) 1000× and (D) 3000×, and unstimulated neutrophils from amniotic fluid were captured at magnifications (E) 1000× and (F) 4000×. PMA indicates phorbol-12-myristate-13-acetate.

Visualization of Elastase and Histone H3 in Amniotic Fluid NETs

Next, we used confocal microscopy to confirm that the structures observed in the amniotic fluid were indeed NETs. We then performed elastase and histone H3 staining of two known components of NETs,78,117,124-127 in combination with DAPI staining. In the amniotic fluid, there were neutrophils in a resting stage (Figure 4; white arrow) and at different stages of NET formation (Figures 4 and 5). In Figure 4, the yellow arrow represents an early stage of NET formation as the neutrophil shows extranuclear histone H3 and intracellular DNA and elastase. The red arrow in Figure 4 represents an intermediate stage of NET formation as elastase is bursting out of the cell; yet, DNA and histones are still contained. In Figure 5, the final stage of NET formation is observed as histone H3, elastase, and DNA burst from an amniotic fluid neutrophil. Altogether, these data demonstrate that different stages of NET formation are present in the amniotic cavity of women with intra-amniotic infection.

Figure 4.

Figure 4.

Confocal microscopy showing different stages of NET formation in amniotic fluid samples. Separated layers show (A) DAPI staining in blue, (B) neutrophil elastase staining in green, (C) histone H3 staining in red, and (D) a merged image. White arrows point to a neutrophil that is in a resting stage, yellow arrows point to a neutrophil in an early stage of NET formation, and red arrows point to a neutrophil in an intermediate stage of NET formation. Magnification 630×. DAPI indicates 4′,6-diamidino-2-phenylindole; NET, neutrophil extracellular trap.

Figure 5.

Figure 5.

Confocal microscopy showing the late stage of NET formation in amniotic fluid samples. Separated layers show (A) DAPI staining in blue, (B) neutrophil elastase staining in green, (C) histone H3 staining in red, and (D) a merged image. Magnification 630×. DAPI indicates 4′,6-diamidino-2-phenylindole; NET, neutrophil extracellular trap.

Maternal or Neonatal Neutrophils Do Not Form NETs in the Amniotic Fluid of Women without Intra-Amniotic Infection

In order to prove that amniotic fluid neutrophils do not form NETs in the absence of bacteria, maternal or neonatal neutrophils were added to amniotic fluid samples from women who delivered at term without intra-amniotic infection. PMA induced in vitro NET formation in maternal and neonatal neutrophils (Figure 6: A vs. B and D vs. E). Neonatal neutrophils formed PMA-induced NETs at a lesser extent than maternal neutrophils, as previously demonstrated (Figure 6E vs. B).128,129 However, maternal and neonatal neutrophils incubated with amniotic fluid samples from women without intra-amniotic infection did not form NETs in vitro (Figure 6C and F). These results show that maternal and neonatal neutrophils do not form NETs in the amniotic fluid in the absence of bacteria or inflammation.

Figure 6.

Figure 6.

DAPI staining of maternal and neonatal neutrophils in the amniotic fluid of women without intra-amniotic infection. Representative fluorescence microscopy images of (A) unstimulated maternal neutrophils, (B) PMA-stimulated maternal neutrophils, (C) maternal neutrophils incubated with 4 different amniotic fluid samples from women without intra-amniotic infection, (D) unstimulated neonatal neutrophils, (E) PMA-stimulated neonatal neutrophils, and (F) neonatal neutrophils incubated with 4 different amniotic fluid samples from women without intra-amniotic infection. DNA is blue (DAPI). Magnification 1000×. DAPI indicates 4′,6-diamidino-2-phenylindole; PMA, phorbol-12-myristate-13-acetate.

Discussion

Amniotic fluid neutrophils are considered to be of fetal origin,49,50 and their number is a useful marker for intra-amniotic inflammation.19 Yet, amniotic fluid neutrophils have been observed in patients with a severe maternal inflammatory response without a fetal inflammatory response (ie, funisitis and chorionic vasculitis), suggesting that, in some cases, amniotic fluid neutrophils are of maternal origin or a mixture of both fetal and maternal neutrophils. Previously, we provided evidence that amniotic fluid neutrophils are a source of antimicrobial peptides130 and cytokines51 in humans. However, their precise role in immune host defense needs to be further elucidated. Herein, we demonstrated for the first time that amniotic fluid neutrophils form NETs in patients with intra-amniotic infection. As NET formation represents a mechanism of innate immune defense against pathogens,83,131 we propose that amniotic fluid neutrophils form extracellular traps to kill microbes invading the amniotic cavity.

NET formation is induced in vivo in response to Staphylococcus aureus,79 S pyogenes,132 Escherichia coli,133 Candida albicans,134 Aspergillus nidulans,135 Leishmania amazonensis, 136 and HIV-1.137 Bacteria lacking a cell wall (eg, Mycoplasma subspecies) can also induce NET formation.138 Furthermore, NETs are abundant in placental villi from preeclamptic women.139 In the present study, we demonstrated that amniotic fluid neutrophils undergo NET formation in intra-amniotic infections mostly due to Ureaplasma subspecies and/or Streptococcus subspecies (mostly Streptococcus agalactiae or group B streptococcus [GBS]). The mechanism whereby Ureaplasma subspecies induces NET formation could involve toll-like receptor (TLR) signaling as the intra-uterine inoculation of Ureaplasma parvum increases the expression of TLRs 1, 2, and 6 in placental tissues.140

GBS induces NET formation in the genital tract (ie, cervicovaginal swabs) when inoculated vaginally.141 Incubation with this Gram-positive bacterium also induces NET formation in peripheral blood neutrophils in vitro.141 Importantly, GBS can evade NET formation by degrading the DNA matrix comprising the extracellular trap.142

We noted that some of the bacteria trapped by amniotic fluid NETs were alive at the time of staining. This finding could represent that (1) the process of NET formation was in its early stages, (2) amniotic fluid NETs could not kill bacteria efficiently, or (3) some bacteria, such as GBS, can degrade the extracellular trap. Further studies are required to investigate the mechanisms whereby Ureaplasma subspecies, Streptococcus subspecies, and other bacteria involved in intra-amniotic infection induce NET formation.

In the current study, we also demonstrated that neither maternal nor neonatal neutrophils form NETs in the amniotic fluid of women without intra-amniotic infection. This finding suggests that amniotic fluid neutrophils form NETs when bacteria invade the amniotic cavity. Yet, there is a possibility that NETs are formed in the setting of sterile intra-amniotic inflammation as alarmins (eg, high-mobility group box 1,143 monosodium urate crystals,144 and heme145) and cytokines (eg, IL-1β144 and IL-878) induce in vitro NET formation in the absence of bacteria.

It is worth mentioning that neonatal neutrophils formed fewer NETs than maternal neutrophils upon PMA stimulation. This impairment was recently attributed to a neonatal NET-inhibitory factor present in cord blood plasma, which blunts in vitro and in vivo NET formation.145

In summary, we report that amniotic fluid neutrophils undergo NET formation in women with intra-amniotic infection. This finding provides a new immune defense mechanism whereby amniotic fluid neutrophils can kill microbes invading the amniotic cavity.

Acknowledgments

The authors thank the physicians and nurses from the Center for Advanced Obstetrical Care and Research and the Intrapartum Unit at Hutzel Women's Hospital in the Detroit Medical Center for their help in collecting samples. The authors also thank staff members from the PRB Clinical Laboratory and the PRB Histology Unit for processing and preparing the pathological sections. Finally, the authors thank Tara Mial for her critical readings of the manuscript.

Footnotes

Declaration of Conflicting Interests: The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Funding: The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This research was supported, in part, by the Perinatology Research Branch, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), and, in part, with federal funds from the NICHD/NIH/DHHS under Contract No. HHSN275201300006C. This research was also supported by the Wayne State University Perinatal Initiative in Maternal, Perinatal and Child Health.

References

  • 1. Romero R, Espinoza J, Goncalves LF, Kusanovic JP, Friel L, Hassan S. The role of inflammation and infection in preterm birth. Semin Reprod Med. 2007;25(1):21–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Vrachnis N, Vitoratos N, Iliodromiti Z, Sifakis S, Deligeoroglou E, Creatsas G. Intrauterine inflammation and preterm delivery. Ann N Y Acad Sci. 2010;1205:118–122. [DOI] [PubMed] [Google Scholar]
  • 3. Romero R, Miranda J, Chaiworapongsa T, et al. A novel molecular microbiologic technique for the rapid diagnosis of microbial invasion of the amniotic cavity and intra-amniotic infection in preterm labor with intact membranes. Am J Reprod Immunol. 2014;71(4):330–358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Romero R, Miranda J, Chaiworapongsa T, et al. Prevalence and clinical significance of sterile intra-amniotic inflammation in patients with preterm labor and intact membranes. Am J Reprod Immunol. 2014;72(5):458–474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Romero R, Miranda J, Chaiworapongsa T, et al. Sterile intra-amniotic inflammation in asymptomatic patients with a sonographic short cervix: prevalence and clinical significance [Published online September 24, 2014]. J Matern Fetal Neonatal Med. 1–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Romero R, Miranda J, Chaemsaithong P, et al. Sterile and microbial-associated intra-amniotic inflammation in preterm prelabor rupture of membranes. J Matern Fetal Neonatal Med. 2015;28(12):1394–1409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Romero R, Quintero R, Oyarzun E, et al. Intraamniotic infection and the onset of labor in preterm premature rupture of the membranes. Am J Obstet Gynecol. 1988;159(3):661–666. [DOI] [PubMed] [Google Scholar]
  • 8. Romero R, Sirtori M, Oyarzun E, et al. Infection and labor. V. Prevalence, microbiology, and clinical significance of intraamniotic infection in women with preterm labor and intact membranes. Am J Obstet Gynecol. 1989;161(3):817–824. [DOI] [PubMed] [Google Scholar]
  • 9. Romero R, Gonzalez R, Sepulveda W, et al. Infection and labor. VIII. Microbial invasion of the amniotic cavity in patients with suspected cervical incompetence: prevalence and clinical significance. Am J Obstet Gynecol. 1992;167(4 pt 1):1086–1091. [DOI] [PubMed] [Google Scholar]
  • 10. Romero R, Miranda J, Kusanovic JP, et al. Clinical chorioamnionitis at term I: microbiology of the amniotic cavity using cultivation and molecular techniques. J Perinat Med. 2015;43(1):19–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Gilstrap LC, 3rd, Cox SM. Acute chorioamnionitis. Obstet Gynecol Clin North Am. 1989;16(2):373–379. [PubMed] [Google Scholar]
  • 12. Gibbs RS, Duff P. Progress in pathogenesis and management of clinical intraamniotic infection. Am J Obstet Gynecol. 1991;164(5 pt 1):1317–1326. [DOI] [PubMed] [Google Scholar]
  • 13. Lee SE, Romero R, Kim CJ, Shim SS, Yoon BH. Funisitis in term pregnancy is associated with microbial invasion of the amniotic cavity and intra-amniotic inflammation. J Matern Fetal Neonatal Med. 2006;19(11):693–697. [DOI] [PubMed] [Google Scholar]
  • 14. Lee SE, Romero R, Jung H, Park CW, Park JS, Yoon BH. The intensity of the fetal inflammatory response in intraamniotic inflammation with and without microbial invasion of the amniotic cavity. Am J Obstet Gynecol. 2007;197(3):294 e291–296. [DOI] [PubMed] [Google Scholar]
  • 15. Gotsch F, Romero R, Kusanovic JP, et al. The fetal inflammatory response syndrome. Clin Obstet Gynecol. 2007;50(3):652–683. [DOI] [PubMed] [Google Scholar]
  • 16. Gibbs RS, Blanco JD, St Clair PJ, Castaneda YS. Quantitative bacteriology of amniotic fluid from women with clinical intraamniotic infection at term. J Infect Dis. 1982;145(1):1–8. [DOI] [PubMed] [Google Scholar]
  • 17. Gibbs RS, Dinsmoor MJ, Newton ER, Ramamurthy RS. A randomized trial of intrapartum versus immediate postpartum treatment of women with intra-amniotic infection. Obstet Gynecol. 1988;72(6):823–828. [DOI] [PubMed] [Google Scholar]
  • 18. Romero R, Chaemsaithong P, Korzeniewski SJ, et al. Clinical chorioamnionitis at term II: the intra-amniotic inflammatory response. J Perinat Med. 2016;44(1):5–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Romero R, Quintero R, Nores J, et al. Amniotic fluid white blood cell count: a rapid and simple test to diagnose microbial invasion of the amniotic cavity and predict preterm delivery. Am J Obstet Gynecol. 1991;165(4 pt 1):821–830. [DOI] [PubMed] [Google Scholar]
  • 20. Romero R, Yoon BH, Mazor M, et al. The diagnostic and prognostic value of amniotic fluid white blood cell count, glucose, interleukin-6, and gram stain in patients with preterm labor and intact membranes. Am J Obstet Gynecol. 1993;169(4):805–816. [DOI] [PubMed] [Google Scholar]
  • 21. Romero R, Yoon BH, Mazor M, et al. A comparative study of the diagnostic performance of amniotic fluid glucose, white blood cell count, interleukin-6, and gram stain in the detection of microbial invasion in patients with preterm premature rupture of membranes. Am J Obstet Gynecol. 1993;169(4):839–851. [DOI] [PubMed] [Google Scholar]
  • 22. Gomez R, Romero R, Galasso M, Behnke E, Insunza A, Cotton DB. The value of amniotic fluid interleukin-6, white blood cell count, and gram stain in the diagnosis of microbial invasion of the amniotic cavity in patients at term. Am J Reprod Immunol. 1994;32(3):200–210. [DOI] [PubMed] [Google Scholar]
  • 23. Yoon BH, Yang SH, Jun JK, Park KH, Kim CJ, Romero R. Maternal blood C-reactive protein, white blood cell count, and temperature in preterm labor: a comparison with amniotic fluid white blood cell count. Obstet Gynecol. 1996;87(2):231–237. [DOI] [PubMed] [Google Scholar]
  • 24. Romero R, Grivel JC, Tarca AL, et al. Evidence of perturbations of the cytokine network in preterm labor. Am J Obstet Gynecol. 2015;213(6):836.e831–836.e818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Romero R, Emamian M, Quintero R, Wan M, Hobbins JC, Mitchell MD. Amniotic fluid prostaglandin levels and intra-amniotic infections. Lancet. 1986;1(8494):1380. [DOI] [PubMed] [Google Scholar]
  • 26. Romero R, Emamian M, Wan M, Quintero R, Hobbins JC, Mitchell MD. Prostaglandin concentrations in amniotic fluid of women with intra-amniotic infection and preterm labor. Am J Obstet Gynecol. 1987;157(6):1461–1467. [DOI] [PubMed] [Google Scholar]
  • 27. Romero R, Wu YK, Mazor M, Hobbins JC, Mitchell MD. Amniotic fluid prostaglandin E2 in preterm labor. Prostaglandins Leukot Essent Fatty Acids. 1988;34(3):141–145. [DOI] [PubMed] [Google Scholar]
  • 28. Romero R, Wu YK, Sirtori M, et al. Amniotic fluid concentrations of prostaglandin F2 alpha, 13,14-dihydro-15-keto-prostaglandin F2 alpha (PGFM) and 11-deoxy-13,14-dihydro-15-keto-11, 16-cyclo-prostaglandin E2 (PGEM-LL) in preterm labor. Prostaglandins. 1989;37(1):149–161. [DOI] [PubMed] [Google Scholar]
  • 29. Romero R, Wu YK, Mazor M, Oyarzun E, Hobbins JC, Mitchell MD. Amniotic fluid arachidonate lipoxygenase metabolites in preterm labor. Prostaglandins Leukot Essent Fatty Acids. 1989;36(2):69–75. [DOI] [PubMed] [Google Scholar]
  • 30. Bry K, Hallman M. Prostaglandins, inflammation, and preterm labor. J Perinatol. 1989;9(1):60–65. [PubMed] [Google Scholar]
  • 31. Mazor M, Wiznitzer A, Maymon E, Leiberman JR, Cohen A. Changes in amniotic fluid concentrations of prostaglandins E2 and F2 alpha in women with preterm labor. Isr J Med Sci. 1990;26(8):425–428. [PubMed] [Google Scholar]
  • 32. Hsu CD, Meaddough E, Aversa K, et al. Dual roles of amniotic fluid nitric oxide and prostaglandin E2 in preterm labor with intra-amniotic infection. Am J Perinatol. 1998;15(12):683–687. [DOI] [PubMed] [Google Scholar]
  • 33. Lee SE, Park IS, Romero R, Yoon BH. Amniotic fluid prostaglandin F2 increases even in sterile amniotic fluid and is an independent predictor of impending delivery in preterm premature rupture of membranes. J Matern Fetal Neonatal Med. 2009;22(10):880–886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Maddipati KR, Romero R, Chaiworapongsa T, et al. Eicosanomic profiling reveals dominance of the epoxygenase pathway in human amniotic fluid at term in spontaneous labor. FASEB J. 2014;28(11):4835–4846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Bhat G, Williams SM, Saade GR, Menon R. Biomarker interactions are better predictors of spontaneous preterm birth. Reprod Sci. 2014;21(3):340–350. [DOI] [PubMed] [Google Scholar]
  • 36. Menon R, Jones J, Gunst PR, et al. Amniotic fluid metabolomic analysis in spontaneous preterm birth. Reprod Sci. 2014;21(6):791–803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Park JY, Romero R, Lee J, Chaemsaithong P, Chaiyasit N, Yoon BH. An elevated amniotic fluid prostaglandin F2alpha concentration is associated with intra-amniotic inflammation/infection, and clinical and histologic chorioamnionitis, as well as impending preterm delivery in patients with preterm labor and intact membranes. J Matern Fetal Neonatal Med. 2016;29(16):2563–2572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Maddipati KR, Romero R, Chaiworapongsa T, et al. Lipidomic analysis of patients with microbial invasion of the amniotic cavity reveals up-regulation of leukotriene B4. FASEB J. 2016;30(10):3296–3307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Gravett MG, Witkin SS, Haluska GJ, Edwards JL, Cook MJ, Novy MJ. An experimental model for intraamniotic infection and preterm labor in rhesus monkeys. Am J Obstet Gynecol. 1994;171(6):1660–1667. [DOI] [PubMed] [Google Scholar]
  • 40. Witkin SS, Gravett MG, Haluska GJ, Novy MJ. Induction of interleukin-1 receptor antagonist in rhesus monkeys after intraamniotic infection with group B streptococci or interleukin-1 infusion. Am J Obstet Gynecol. 1994;171(6):1668–1672. [DOI] [PubMed] [Google Scholar]
  • 41. Gravett MG, Haluska GJ, Cook MJ, Novy MJ. Fetal and maternal endocrine responses to experimental intrauterine infection in rhesus monkeys. Am J Obstet Gynecol. 1996;174(6):1725–1731; discussion 1731-1723. [DOI] [PubMed] [Google Scholar]
  • 42. Bethea CL, Gravett MG, Sadowsky DW, Haluska GJ, Axthelm MK, Novy MJ. Amniotic fluid prolactin is decreased by experimental intrauterine infection or interleukin-1beta infusion but not via prostaglandins in pregnant rhesus macaques. Biol Reprod. 1998;58(6):1385–1393. [DOI] [PubMed] [Google Scholar]
  • 43. Gravett MG, Novy MJ, Rosenfeld RG, et al. Diagnosis of intra-amniotic infection by proteomic profiling and identification of novel biomarkers. JAMA. 2004;292(4):462–469. [DOI] [PubMed] [Google Scholar]
  • 44. Gravett MG, Adams KM, Sadowsky DW, et al. Immunomodulators plus antibiotics delay preterm delivery after experimental intraamniotic infection in a nonhuman primate model. Am J Obstet Gynecol. 2007;197(5):518.e511–518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Adams Waldorf KM, Persing D, Novy MJ, Sadowsky DW, Gravett MG. Pretreatment with toll-like receptor 4 antagonist inhibits lipopolysaccharide-induced preterm uterine contractility, cytokines, and prostaglandins in rhesus monkeys. Reprod Sci. 2008;15(2):121–127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Novy MJ, Duffy L, Axthelm MK, et al. Ureaplasma parvum or Mycoplasma hominis as sole pathogens cause chorioamnionitis, preterm delivery, and fetal pneumonia in rhesus macaques. Reprod Sci. 2009;16(1):56–70. [DOI] [PubMed] [Google Scholar]
  • 47. Grigsby PL, Novy MJ, Adams Waldorf KM, Sadowsky DW, Gravett MG. Choriodecidual inflammation: a harbinger of the preterm labor syndrome. Reprod Sci. 2010;17(1):85–94. [DOI] [PubMed] [Google Scholar]
  • 48. Grigsby PL, Novy MJ, Sadowsky DW, et al. 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(6):475.e471–475.e414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Sampson JE, Theve RP, Blatman RN, et al. Fetal origin of amniotic fluid polymorphonuclear leukocytes. Am J Obstet Gynecol. 1997;176(1 Pt 1):77–81. [DOI] [PubMed] [Google Scholar]
  • 50. Lee SD, Kim MR, Hwang PG, Shim SS, Yoon BH, Kim CJ. Chorionic plate vessels as an origin of amniotic fluid neutrophils. Pathol Int. 2004;54(7):516–522. [DOI] [PubMed] [Google Scholar]
  • 51. Martinez-Verea A, Romero R, Xu Y, et al. Clinical Chorioamnionitis at Term VII: The Amniotic Fluid Cellular Immune Response [Published online October 20, 2016]. J Perinat Med. doi:10.1515/jpm-2016-0225 [DOI] [PMC free article] [PubMed]
  • 52. Romero R, Manogue KR, Mitchell MD, et al. Infection and labor. IV. Cachectin-tumor necrosis factor in the amniotic fluid of women with intraamniotic infection and preterm labor. Am J Obstet Gynecol. 1989;161(2):336–341. [DOI] [PubMed] [Google Scholar]
  • 53. Romero R, Mazor M, Sepulveda W, Avila C, Copeland D, Williams J. Tumor necrosis factor in preterm and term labor. Am J Obstet Gynecol. 1992;166(5):1576–1587. [DOI] [PubMed] [Google Scholar]
  • 54. Romero R, Brody DT, Oyarzun E, et al. Infection and labor. III. Interleukin-1: a signal for the onset of parturition. Am J Obstet Gynecol. 1989;160(5 Pt 1):1117–1123. [DOI] [PubMed] [Google Scholar]
  • 55. Romero R, Parvizi ST, Oyarzun E, et al. Amniotic fluid interleukin-1 in spontaneous labor at term. J Reprod Med. 1990;35(3):235–238. [PubMed] [Google Scholar]
  • 56. Romero R, Mazor M, Brandt F, et al. Interleukin-1 alpha and interleukin-1 beta in preterm and term human parturition. Am J Reprod Immunol. 1992;27(3-4):117–123. [DOI] [PubMed] [Google Scholar]
  • 57. Heng YJ, Liong S, Permezel M, Rice GE, Di Quinzio MK, Georgiou HM. The interplay of the interleukin 1 system in pregnancy and labor. Reprod Sci. 2014;21(1):122–130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Romero R, Xu Y, Plazyo O, et al. A Role for the Inflammasome in Spontaneous Labor at Term [Published online March 8, 2016]. Am J Reprod Immunol. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Romero R, Ceska M, Avila C, Mazor M, Behnke E, Lindley I. Neutrophil attractant/activating peptide-1/interleukin-8 in term and preterm parturition. Am J Obstet Gynecol. 1991;165(4 pt 1):813–820. [DOI] [PubMed] [Google Scholar]
  • 60. Cherouny PH, Pankuch GA, Romero R, et al. Neutrophil attractant/activating peptide-1/interleukin-8: association with histologic chorioamnionitis, preterm delivery, and bioactive amniotic fluid leukoattractants. Am J Obstet Gynecol. 1993;169(5):1299–1303. [DOI] [PubMed] [Google Scholar]
  • 61. Osmers RG, Blaser J, Kuhn W, Tschesche H. Interleukin-8 synthesis and the onset of labor. Obstet Gynecol. 1995;86(2):223–229. [DOI] [PubMed] [Google Scholar]
  • 62. Elliott CL, Loudon JA, Brown N, Slater DM, Bennett PR, Sullivan MH. IL-1beta and IL-8 in human fetal membranes: changes with gestational age, labor, and culture conditions. Am J Reprod Immunol. 2001;46(4):260–267. [DOI] [PubMed] [Google Scholar]
  • 63. Gomez-Lopez N, Tong WC, Arenas-Hernandez M, et al. Chemotactic activity of gestational tissues through late pregnancy, term labor, and RU486-induced preterm labor in Guinea pigs. Am J Reprod Immunol. 2015;73(4):341–352. [DOI] [PubMed] [Google Scholar]
  • 64. Romero R, Gomez R, Galasso M, et al. Macrophage inflammatory protein-1 alpha in term and preterm parturition: effect of microbial invasion of the amniotic cavity. Am J Reprod Immunol. 1994;32(2):108–113. [DOI] [PubMed] [Google Scholar]
  • 65. Young A, Thomson AJ, Ledingham M, Jordan F, Greer IA, Norman JE. Immunolocalization of proinflammatory cytokines in myometrium, cervix, and fetal membranes during human parturition at term. Biol Reprod. 2002;66(2):445–449. [DOI] [PubMed] [Google Scholar]
  • 66. Osman I, Young A, Ledingham MA, et al. Leukocyte density and pro-inflammatory cytokine expression in human fetal membranes, decidua, cervix and myometrium before and during labour at term. Mol Hum Reprod. 2003;9(1):41–45. [DOI] [PubMed] [Google Scholar]
  • 67. Haddad R, Tromp G, Kuivaniemi H, et al. Human spontaneous labor without histologic chorioamnionitis is characterized by an acute inflammation gene expression signature. Am J Obstet Gynecol. 2006;195(2):394.e391–324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Hassan SS, Romero R, Haddad R, et al. The transcriptome of the uterine cervix before and after spontaneous term parturition. Am J Obstet Gynecol. 2006;195(3):778–786. [DOI] [PubMed] [Google Scholar]
  • 69. Hassan SS, Romero R, Tarca AL, et al. Signature pathways identified from gene expression profiles in the human uterine cervix before and after spontaneous term parturition. Am J Obstet Gynecol. 2007;197(3):250.e251–257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Hassan SS, Romero R, Tarca AL, et al. The transcriptome of cervical ripening in human pregnancy before the onset of labor at term: identification of novel molecular functions involved in this process. J Matern Fetal Neonatal Med. 2009;22(12):1183–1193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Bollapragada S, Youssef R, Jordan F, Greer I, Norman J, Nelson S. Term labor is associated with a core inflammatory response in human fetal membranes, myometrium, and cervix. Am J Obstet Gynecol. 2009;200(1):104.e101–111. [DOI] [PubMed] [Google Scholar]
  • 72. Mittal P, Romero R, Tarca AL, et al. Characterization of the myometrial transcriptome and biological pathways of spontaneous human labor at term. J Perinat Med. 2010;38(6):617–643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Lannon SM, Vanderhoeven JP, Eschenbach DA, Gravett MG, Adams Waldorf KM. Synergy and interactions among biological pathways leading to preterm premature rupture of membranes. Reprod Sci. 2014;21(10):1215–1227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Taguchi A, Yamashita A, Kawana K, et al. Recent Progress in Therapeutics for Inflammation-Associated Preterm Birth: A Review. Reprod Sci. 2017;24(1):7–18. [DOI] [PubMed] [Google Scholar]
  • 75. Kumar V, Sharma A. Neutrophils: Cinderella of innate immune system. Int Immunopharmacol. 2010;10(11):1325–1334. [DOI] [PubMed] [Google Scholar]
  • 76. Witko-Sarsat V, Rieu P, Descamps-Latscha B, Lesavre P, Halbwachs-Mecarelli L. Neutrophils: molecules, functions and pathophysiological aspects. Lab Invest. 2000;80(5):617–653. [DOI] [PubMed] [Google Scholar]
  • 77. Segal AW. How neutrophils kill microbes. Annu Rev Immunol. 2005;23:197–223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Brinkmann V, Reichard U, Goosmann C, et al. Neutrophil extracellular traps kill bacteria. Science. 2004;303(5663):1532–1535. [DOI] [PubMed] [Google Scholar]
  • 79. Fuchs TA, Abed U, Goosmann C, et al. Novel cell death program leads to neutrophil extracellular traps. J Cell Biol. 2007;176(2):231–241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Brinkmann V, Zychlinsky A. Beneficial suicide: why neutrophils die to make NETs. Nat Rev Microbiol. 2007;5(8):577–582. [DOI] [PubMed] [Google Scholar]
  • 81. Brinkmann V, Zychlinsky A. Neutrophil extracellular traps: is immunity the second function of chromatin? J Cell Biol. 2012;198(5):773–783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Urban CF, Ermert D, Schmid M, et al. Neutrophil extracellular traps contain calprotectin, a cytosolic protein complex involved in host defense against Candida albicans. PLoS Pathog. 2009;5(10):e1000639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Sorensen OE, Borregaard N. Neutrophil extracellular traps—the dark side of neutrophils. J Clin Invest. 2016;126(5):1612–1620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Gibbs RS, Castillo MS, Rodgers PJ. Management of acute chorioamnionitis. Am J Obstet Gynecol. 1980;136(6):709–713. [DOI] [PubMed] [Google Scholar]
  • 85. Newton ER. Chorioamnionitis and intraamniotic infection. Clin Obstet Gynecol. 1993;36(4):795–808. [DOI] [PubMed] [Google Scholar]
  • 86. Tita AT, Andrews WW. Diagnosis and management of clinical chorioamnionitis. Clin Perinatol. 2010;37(2):339–354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Romero R, Mazor M, Wu YK, et al. Infection in the pathogenesis of preterm labor. Semin Perinatol. 1988;12(4):262–279. [PubMed] [Google Scholar]
  • 88. Romero R, Mazor M. Infection and preterm labor. Clin Obstet Gynecol. 1988;31(3):553–584. [DOI] [PubMed] [Google Scholar]
  • 89. Romero R, Shamma F, Avila C, et al. Infection and labor. VI. Prevalence, microbiology, and clinical significance of intraamniotic infection in twin gestations with preterm labor. Am J Obstet Gynecol. 1990;163(3):757–761. [DOI] [PubMed] [Google Scholar]
  • 90. Romero R, Ghidini A, Mazor M, Behnke E. Microbial invasion of the amniotic cavity in premature rupture of membranes. Clin Obstet Gynecol. 1991;34(4):769–778. [DOI] [PubMed] [Google Scholar]
  • 91. Romero R, Nores J, Mazor M, et al. Microbial invasion of the amniotic cavity during term labor. Prevalence and clinical significance. J Reprod Med. 1993;38(7):543–548. [PubMed] [Google Scholar]
  • 92. Yoon BH, Romero R, Moon JB, et al. Clinical significance of intra-amniotic inflammation in patients with preterm labor and intact membranes. Am J Obstet Gynecol. 2001;185(5):1130–1136. [DOI] [PubMed] [Google Scholar]
  • 93. Romero R, Chaiworapongsa T, Savasan ZA, et al. Clinical chorioamnionitis is characterized by changes in the expression of the alarmin HMGB1 and one of its receptors, sRAGE. J Matern Fetal Neonatal Med. 2012;25(6):558–567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Madan I, Romero R, Kusanovic JP, et al. The frequency and clinical significance of intra-amniotic infection and/or inflammation in women with placenta previa and vaginal bleeding: an unexpected observation. J Perinat Med. 2010;38(3):275–279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Cruciani L, Romero R, Vaisbuch E, et al. Pentraxin 3 in amniotic fluid: a novel association with intra-amniotic infection and inflammation. J Perinat Med. 2010;38(2):161–171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Romero R, Chaiworapongsa T, Alpay Savasan Z, et al. Damage-associated molecular patterns (DAMPs) in preterm labor with intact membranes and preterm PROM: a study of the alarmin HMGB1. J Matern Fetal Neonatal Med. 2011;24(12):1444–1455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Gervasi MT, Romero R, Bracalente G, et al. Midtrimester amniotic fluid concentrations of interleukin-6 and interferon-gamma-inducible protein-10: evidence for heterogeneity of intra-amniotic inflammation and associations with spontaneous early (<32 weeks) and late (>32 weeks) preterm delivery. J Perinat Med. 2012;40(4):329–343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Combs CA, Gravett M, Garite TJ, et al. Amniotic fluid infection, inflammation, and colonization in preterm labor with intact membranes. Am J Obstet Gynecol. 2014;210(2):125.e121–125.e115. [DOI] [PubMed] [Google Scholar]
  • 99. Kacerovsky M, Musilova I, Andrys C, et al. Prelabor rupture of membranes between 34 and 37 weeks: the intraamniotic inflammatory response and neonatal outcomes. Am J Obstet Gynecol. 2014;210(4):325.e321–325.e310. [DOI] [PubMed] [Google Scholar]
  • 100. Chaemsaithong P, Romero R, Korzeniewski SJ, et al. A point of care test for the determination of amniotic fluid interleukin-6 and the chemokine CXCL-10/IP-10. J Matern Fetal Neonatal Med. 2015;28(13):1510–1519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Chaemsaithong P, Romero R, Korzeniewski SJ, et al. A point of care test for interleukin-6 in amniotic fluid in preterm prelabor rupture of membranes: a step toward the early treatment of acute intra-amniotic inflammation/infection. J Matern Fetal Neonatal Med. 2016;29(3):360–367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Chaemsaithong P, Romero R, Korzeniewski SJ, et al. A rapid interleukin-6 bedside test for the identification of intra-amniotic inflammation in preterm labor with intact membranes. J Matern Fetal Neonatal Med. 2016;29(3):349–359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. 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(4 suppl):S29–S52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Redline RW. Classification of placental lesions. Am J Obstet Gynecol. 2015;213:S21–28. [DOI] [PubMed] [Google Scholar]
  • 105. Redline RW. Inflammatory responses in the placenta and umbilical cord. Semin Fetal Neonatal Med. 2006;11(5):296–301. [DOI] [PubMed] [Google Scholar]
  • 106. Mi Lee S, Romero R, Lee KA, et al. The frequency and risk factors of funisitis and histologic chorioamnionitis in pregnant women at term who delivered after the spontaneous onset of labor. J Matern Fetal Neonatal Med. 2011;24(1):37–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Yoon BH, Romero R, Park JS, et al. The relationship among inflammatory lesions of the umbilical cord (funisitis), umbilical cord plasma interleukin 6 concentration, amniotic fluid infection, and neonatal sepsis. Am J Obstet Gynecol. 2000;183(5):1124–1129. [DOI] [PubMed] [Google Scholar]
  • 108. Park JS, Romero R, Yoon BH, et al. The relationship between amniotic fluid matrix metalloproteinase-8 and funisitis. Am J Obstet Gynecol. 2001;185(5):1156–1161. [DOI] [PubMed] [Google Scholar]
  • 109. Pacora P, Chaiworapongsa T, Maymon E, et al. Funisitis and chorionic vasculitis: the histological counterpart of the fetal inflammatory response syndrome. J Matern Fetal Neonatal Med. 2002;11(1):18–25. [DOI] [PubMed] [Google Scholar]
  • 110. Park CW, Lee SM, Park JS, Jun JK, Romero R, Yoon BH. The antenatal identification of funisitis with a rapid MMP-8 bedside test. J Perinat Med. 2008;36(6):497–502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Yoon BH, Romero R, Kim CJ, et al. Amniotic fluid interleukin-6: a sensitive test for antenatal diagnosis of acute inflammatory lesions of preterm placenta and prediction of perinatal morbidity. Am J Obstet Gynecol. 1995;172(3):960–970. [DOI] [PubMed] [Google Scholar]
  • 112. Yoon BH, Romero R, Lim JH, et al. The clinical significance of detecting Ureaplasma urealyticum by the polymerase chain reaction in the amniotic fluid of patients with preterm labor. Am J Obstet Gynecol. 2003;189(4):919–924. [DOI] [PubMed] [Google Scholar]
  • 113. DiGiulio DB, Romero R, Amogan HP, et al. Microbial prevalence, diversity and abundance in amniotic fluid during preterm labor: a molecular and culture-based investigation. PLoS One. 2008;3(8):e3056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. DiGiulio DB, Gervasi MT, Romero R, et al. Microbial invasion of the amniotic cavity in pregnancies with small-for-gestational-age fetuses. J Perinat Med. 2010;38(5):495–502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Romero R, Jimenez C, Lohda AK, et al. Amniotic fluid glucose concentration: a rapid and simple method for the detection of intraamniotic infection in preterm labor. Am J Obstet Gynecol. 1990;163(3):968–974. [DOI] [PubMed] [Google Scholar]
  • 116. Romero R, Emamian M, Quintero R, et al. The value and limitations of the gram stain examination in the diagnosis of intraamniotic infection. Am J Obstet Gynecol. 1988;159(1):114–119. [DOI] [PubMed] [Google Scholar]
  • 117. Brinkmann V, Laube B, Abu Abed U, Goosmann C, Zychlinsky A. Neutrophil extracellular traps: how to generate and visualize them. J Vis Exp. 2010(36):pii:1724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Kim MJ, Romero R, Gervasi MT, et al. Widespread microbial invasion of the chorioamniotic membranes is a consequence and not a cause of intra-amniotic infection. Lab Invest. 2009;89(8):924–936. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Lin MS, Alfi OS, Donnell GN. Differential fluorescence of sister chromatids with 4’-6-diamidino-2-phenylindole. Can J Genet Cytol. 1976;18(3):545–547. [DOI] [PubMed] [Google Scholar]
  • 120. Coleman AW, Maguire MJ, Coleman JR. Mithramycin- and 4’-6-diamidino-2-phenylindole (DAPI)-DNA staining for fluorescence microspectrophotometric measurement of DNA in nuclei, plastids, and virus particles. J Histochem Cytochem. 1981;29(8):959–968. [DOI] [PubMed] [Google Scholar]
  • 121. McCarthy DA, Bernhagen J, Liu YC, Perry JD. A rapid preparation technique for leucocytes. J Microsc. 1990;158(pt 1):63–72. [DOI] [PubMed] [Google Scholar]
  • 122. McCarthy DA, Rampton DS, Liu YC. Peripheral blood neutrophils in inflammatory bowel disease: morphological evidence of in vivo activation in active disease. Clin Exp Immunol. 1991;86(3):489–493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Lannan S, McLean A, Drost E, et al. Changes in neutrophil morphology and morphometry following exposure to cigarette smoke. Int J Exp Pathol. 1992;73(2):183–191. [PMC free article] [PubMed] [Google Scholar]
  • 124. Neeli I, Khan SN, Radic M. Histone deimination as a response to inflammatory stimuli in neutrophils. J Immunol. 2008;180(3):1895–1902. [DOI] [PubMed] [Google Scholar]
  • 125. Wang Y, Li M, Stadler S, et al. Histone hypercitrullination mediates chromatin decondensation and neutrophil extracellular trap formation. J Cell Biol. 2009;184(2):205–213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Li P, Li M, Lindberg MR, Kennett MJ, Xiong N, Wang Y. PAD4 is essential for antibacterial innate immunity mediated by neutrophil extracellular traps. J Exp Med. 2010;207(9):1853–1862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Papayannopoulos V, Metzler KD, Hakkim A, Zychlinsky A. Neutrophil elastase and myeloperoxidase regulate the formation of neutrophil extracellular traps. J Cell Biol. 2010;191(3):677–691. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Yost CC, Cody MJ, Harris ES, et al. Impaired neutrophil extracellular trap (NET) formation: a novel innate immune deficiency of human neonates. Blood. 2009;113(25):6419–6427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Marcos V, Nussbaum C, Vitkov L, et al. Delayed but functional neutrophil extracellular trap formation in neonates. Blood. 2009;114(23):4908–4911; author reply 4911-4902. [DOI] [PubMed] [Google Scholar]
  • 130. Espinoza J, Chaiworapongsa T, Romero R, et al. Antimicrobial peptides in amniotic fluid: defensins, calprotectin and bacterial/permeability-increasing protein in patients with microbial invasion of the amniotic cavity, intra-amniotic inflammation, preterm labor and premature rupture of membranes. J Matern Fetal Neonatal Med. 2003;13(1):2–21. [DOI] [PubMed] [Google Scholar]
  • 131. Mohanty T, Sjogren J, Kahn F, et al. A novel mechanism for NETosis provides antimicrobial defense at the oral mucosa. Blood. 2015;126(18):2128–2137. [DOI] [PubMed] [Google Scholar]
  • 132. Buchanan JT, Simpson AJ, Aziz RK, et al. DNase expression allows the pathogen group A Streptococcus to escape killing in neutrophil extracellular traps. Curr Biol. 2006;16(4):396–400. [DOI] [PubMed] [Google Scholar]
  • 133. Clark SR, Ma AC, Tavener SA, et al. Platelet TLR4 activates neutrophil extracellular traps to ensnare bacteria in septic blood. Nat Med. 2007;13(4):463–469. [DOI] [PubMed] [Google Scholar]
  • 134. Urban CF, Reichard U, Brinkmann V, Zychlinsky A. Neutrophil extracellular traps capture and kill Candida albicans yeast and hyphal forms. Cell Microbiol. 2006;8(4):668–676. [DOI] [PubMed] [Google Scholar]
  • 135. Bianchi M, Hakkim A, Brinkmann V, et al. Restoration of NET formation by gene therapy in CGD controls aspergillosis. Blood. 2009;114(13):2619–2622. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Guimaraes-Costa AB, Nascimento MT, Froment GS, et al. Leishmania amazonensis promastigotes induce and are killed by neutrophil extracellular traps. Proc Natl Acad Sci U S A. 2009;106(16):6748–6753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. Saitoh T, Komano J, Saitoh Y, et al. Neutrophil extracellular traps mediate a host defense response to human immunodeficiency virus-1. Cell Host Microbe. 2012;12(1):109–116. [DOI] [PubMed] [Google Scholar]
  • 138. Cacciotto C, Cubeddu T, Addis MF, et al. Mycoplasma lipoproteins are major determinants of neutrophil extracellular trap formation [Published online May 10, 2016]. Cell Microbiol. [DOI] [PubMed] [Google Scholar]
  • 139. Gupta AK, Hasler P, Holzgreve W, Gebhardt S, Hahn S. Induction of neutrophil extracellular DNA lattices by placental microparticles and IL-8 and their presence in preeclampsia. Hum Immunol. 2005;66(11):1146–1154. [DOI] [PubMed] [Google Scholar]
  • 140. Allam AB, von Chamier M, Brown MB, Reyes L. Immune profiling of BALB/C and C57BL/6 mice reveals a correlation between Ureaplasma parvum-Induced fetal inflammatory response syndrome-like pathology and increased placental expression of TLR2 and CD14. Am J Reprod Immunol. 2014;71(3):241–251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Carey AJ, Tan CK, Mirza S, et al. Infection and cellular defense dynamics in a novel 17beta-estradiol murine model of chronic human group B streptococcus genital tract colonization reveal a role for hemolysin in persistence and neutrophil accumulation. J Immunol. 2014;192(4):1718–1731. [DOI] [PubMed] [Google Scholar]
  • 142. Derre-Bobillot A, Cortes-Perez NG, Yamamoto Y, et al. Nuclease A (Gbs0661), an extracellular nuclease of Streptococcus agalactiae, attacks the neutrophil extracellular traps and is needed for full virulence. Mol Microbiol. 2013;89(3):518–531. [DOI] [PubMed] [Google Scholar]
  • 143. Tadie JM, Bae HB, Jiang S, et al. HMGB1 promotes neutrophil extracellular trap formation through interactions with Toll-like receptor 4. Am J Physiol Lung Cell Mol Physiol. 2013;304(5):L342–L349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Mitroulis I, Kambas K, Chrysanthopoulou A, et al. Neutrophil extracellular trap formation is associated with IL-1beta and autophagy-related signaling in gout. PLoS One. 2011;6(12):e29318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. Yost CC, Schwertz H, Cody MJ, et al. Neonatal NET-inhibitory factor and related peptides inhibit neutrophil extracellular trap formation. J Clin Invest. 2016;126(10):3783–3798. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Reproductive Sciences are provided here courtesy of Society for Reproductive Investigation

RESOURCES