Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Jun 1.
Published in final edited form as: J Matern Fetal Neonatal Med. 2018 Jan 17;32(12):1978–1991. doi: 10.1080/14767058.2017.1422714

IN VIVO EVIDENCE OF INFLAMMASOME ACTIVATION DURING SPONTANEOUS LABOR AT TERM

Bogdan Panaitescu 1,2, Roberto Romero 1,3,4,5, Nardhy Gomez-Lopez 1,2,6, Yi Xu 1,2, Yaozhu Leng 1,2, Eli Maymon 1,2, Percy Pacora 1,2, Offer Erez 1,2, Lami Yeo 1,2, Sonia S Hassan 1,2, Chaur-Dong Hsu 2
PMCID: PMC6050140  NIHMSID: NIHMS956854  PMID: 29295667

Abstract

Objective

Upon inflammasome activation, the adaptor protein of the inflammasome apoptosis-associated speck-like protein containing a CARD (ASC) forms intracellular specks, which can be released into the extracellular space. The objectives of this study were to investigate whether: 1) extracellular ASC is present in the amniotic fluid of women who delivered at term; 2) amniotic fluid ASC concentrations are greater in women who underwent spontaneous labor at term than in those who delivered at term in the absence of labor; and 3) amniotic epithelial and mesenchymal cells can form intracellular ASC specks in vitro.

Methods

This retrospective cross-sectional study included amniotic fluid samples from 41 women who delivered at term in the absence of labor (n=24) or underwent spontaneous labor at term (n=17). Amniotic epithelial and mesenchymal cells were also isolated from the chorioamniotic membranes from a separate group of women delivered at term (n=3), in which ASC speck formation was assessed by confocal microscopy. Monocytes from healthy individuals were used as positive controls for ASC speck formation (n=3).

Results

1) The adaptor protein of the inflammasome ASC is detectable in the amniotic fluid of women who delivered at term; 2) Amniotic fluid ASC concentration was higher in women who underwent spontaneous labor at term than in those who delivered at term without labor; and 3) Amniotic epithelial and mesenchymal cells are capable of forming ASC specks and filaments in vitro.

Conclusion

Amniotic fluid ASC concentrations are increased in women who underwent spontaneous labor at term. Amniotic epithelial and mesenchymal cells are capable of forming ASC specks, suggesting that these cells are a source of extracellular ASC in the amniotic fluid. These findings provide in vivo evidence that there is inflammasome activation in the amniotic cavity during the physiological process of labor at term.

Keywords: Apoptosis-associated speck-like protein containing a CARD (ASC), ASC speck, amnion, caspase-1, chorion, chorioamniotic membranes, cytokine, cryopyrin, labor, interleukin-1 beta, IL-18, intra-amniotic inflammation, biomarker, NOD-like receptor (NLR), NLR Family Pyrin Domain Containing 3 protein (NLRP3), normal pregnancy, parturition, preterm labor, pattern recognition receptor (PRR), PYD And CARD Domain-Containing Protein (PYCARD), sterile inflammation

INTRODUCTION

In most women, spontaneous labor at term occurs in the absence of intra-amniotic infection [1, 2] and, therefore, is considered a sterile inflammatory process [3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13]. This process is characterized by the increased bioavailability of cytokines [14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28] and chemokines [29, 30, 31, 32, 33, 34] in the amniotic fluid, maternal circulation [35, 36, 37], and reproductive tissues [27, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52]. In addition, labor is accompanied by the infiltration of innate (e.g. neutrophils and macrophages) inflammatory cells into the cervix [45, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62], myometrium [55, 63, 64, 65, 66, 67, 68, 69], and chorioamniotic membranes [41, 70, 71, 72, 73]. The latter tissues also contain infiltrating adaptive effector immune cells such as T cells [72, 74] and, to a lesser extent, B cells [75]. The mechanisms responsible for the sterile inflammatory process of labor at term are not fully understood.

Sterile inflammation is induced by danger signals derived from necrosis or cellular stress [76], termed damage-associated molecular patterns (DAMPs) [77, 78] or alarmins [79], which can activate the inflammasome [80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91]. Inflammasomes are cytoplasmic multi-protein complexes composed of: 1) a sensor molecule or pattern recognition receptor (PRR) [e.g. nucleotide-binding oligomerization domain-like receptors (NLRs)], 2) the adaptor protein [apoptosis-associated speck-like protein (ASC) or PYD and CARD domain containing protein (PYCARD)], and 3) pro-caspase-1 [92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126]. Upon activation, the inflammasome complex induces the auto-catalytic cleavage of pro-caspase-1 into its active form [92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126] which, in turn, can cleave pro-IL-1β and pro-IL-18 into their mature and secreted bioactive forms [127, 128, 129, 130, 131, 132, 133, 134, 135, 136, 137]. Active forms of caspase-1 are also required to induce a specific type of pro-inflammatory programmed cell death termed pyroptosis [138, 139, 140].

Inflammasomes are implicated in the mechanisms that lead to the sterile inflammatory process of labor at term [141, 142, 143, 144, 145] and preterm [146, 147]. Inflammasome activation includes two steps: initiation of the nuclear factor kappa B (NF-κB) pathway [148, 149] and assembly of the inflammasome complex [149]. Upon inflammasome activation, the ASC protein assembles into a large intracellular complex termed “speck” that consists of multimers of ASC dimers [150, 151]. ASC specks, however, can also be released to the extracellular space and remain stable for extended periods of time [152], where they can serve as danger signals amplifying the inflammatory response [153, 154]. Recently, we showed that intracellular ASC specks are abundant in the chorioamniotic membranes and choriodecidual leukocytes from women who underwent spontaneous labor at term [145]. However, whether amniotic cells can form ASC specks and release them into the amniotic fluid during the physiological process of labor (i.e. in vivo evidence of inflammasome activation) is unknown.

In the study herein, we investigated whether: 1) extracellular ASC is present in the amniotic fluid of women who delivered at term; 2) amniotic fluid ASC concentrations are greater in women who underwent spontaneous labor at term than in those who delivered at term in the absence of labor; and 3) amniotic epithelial and mesenchymal cells can form intracellular ASC specks in vitro.

MATERIALS AND METHODS

Study population

This is a retrospective cross-sectional study conducted by searching our clinical database and bank of biological samples which included 41 patients who had amniotic fluid samples obtained by trans-abdominal amniocentesis. Women were grouped into the following: 1) subjects who delivered at term in the absence of labor (n=24); and 2) subjects who underwent spontaneous labor at term (n=17). The chorioamniotic membranes from a separate group of women who delivered at term (n=3) were also collected for primary cell culture isolation of amniotic epithelial and mesenchymal cells. As positive controls, monocytes were isolated from healthy individuals (n=3). Both amniotic primary cells and monocytes were used for the in vitro determination of intra- and extracellular ASC specks. Women who had positive cultures for microorganisms and high concentrations of IL-6 (≥2.6 ng/mL) [1, 155, 156, 157, 158, 159, 160, 161, 162, 163, 164, 165, 166, 167, 168, 169, 170, 171, 172, 173, 174, 175, 176, 177, 178, 179, 180, 181, 182] were excluded. Women with multiple gestations, or those who had fetuses affected with chromosomal and/or sonographic abnormalities were also excluded. Maternal and neonatal data were obtained by retrospective clinical chart review.

All patients provided written informed consent to donate additional amniotic fluid or chorioamniotic membrane samples for research purposes, according to protocols approved by the Institutional Review Boards of the Detroit Medical Center (Detroit, MI, USA), 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, U. S. Department of Health and Human Services (NICHD/NIH/DHHS).

Clinical definitions

Gestational age was determined by the date of 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. Spontaneous term labor was defined as the presence of regular uterine contractions with a frequency of at least 1 every 10 mins, and cervical change after 37 weeks of gestation. Acute histologic chorioamnionitis was diagnosed based on the presence of inflammatory cells in the chorionic plate and/or chorioamniotic membranes [183, 184, 185, 186], while acute funisitis was diagnosed by the presence of neutrophils in the wall of umbilical vessels and/or Wharton’s jelly, using previously described criteria [183, 184, 187, 188, 189].

Amniotic fluid sample collection

Amniotic fluid samples were obtained by trans-abdominal amniocentesis under ultrasound guidance. Women at term (≥37 weeks) not in labor underwent amniocentesis to either assess fetal lung maturity prior to cesarean delivery or for research purposes. Women who underwent spontaneous labor at term underwent amniocentesis to assess fetal lung maturity or to detect the presence or absence of intra-amniotic infection/inflammation, as described above. Samples of amniotic fluid were transported to the laboratory in a sterile capped syringe, centrifuged at 1300 × g for 10 min at 4 °C, and the supernatant was stored at −70 °C until use.

Determination of ASC concentrations in the amniotic fluid

Concentrations of ASC (also termed PYCARD) in the amniotic fluid were determined by using a sensitive and specific enzyme-linked immunosorbent assay (ELISA) kit obtained from LifeSpan BioSciences (Seattle, WA, USA). This ELISA kit was initially validated in our laboratory prior the execution of this study. Amniotic fluid concentrations of ASC were obtained by interpolation from the standard curve. The inter- and intra-assay coefficients of variation were 5.004% and 8.614%, respectively. The sensitivity of the assay was 0.131 ng/mL.

Primary cell culture of amniotic epithelial and mesenchymal cells

Immediately after collection, the amnion membrane was manually peeled from the underlying chorion layer of chorioamniotic membranes and then cut into 2×2cm pieces. Amniotic mesenchymal cells (AMCs) were obtained by digesting the amnion fragments in 1mg/mL collagenase A (Sigma-Aldrich, St. Louis, MO) at 37°C with gentle shaking for 3 hours. The tissues were then filtered through a 100µm cell strainer (Falcon; Corning Life Sciences, Durham, NC) and centrifuged at 200 × g for 10 min. AMCs were suspended in Dulbecco’s modified Eagle’s medium (DMEM; Life Technologies, Grand Island, NY) containing 10% fetal bovine serum (FBS, Life Technologies) and 100U/mL penicillin and streptomycin antibiotics (Life Technologies). Amniotic epithelial cells (AECs) were obtained by rinsing the amnion fragments in 0.05% (w/v) trypsin/EDTA (Life Technologies) and incubating such fragments with 15mL of fresh trypsin/EDTA at 37°C with gentle shaking for 10 min. The trypsin digestion supernatant was discarded and the amnion fragments were placed into fresh trypsin/EDTA solution and further digested at 37°C for 40 min with gentle shaking. The total digestion/incubation process was repeated twice. In order to stop digestion, FBS was added to the supernatant between each incubation period. Finally, the supernatant was centrifuged for 10 min at 200 × g. Isolated AECs and AMCs were then cultured in DMEM containing 10% FBS and 100U/mL penicillin and streptomycin at 37°C with 5% CO2. All experiments were performed with cells from passage 2-3.

Isolation of peripheral blood mononuclear cells

Peripheral blood mononuclear cells (PBMCs) were isolated using the density gradient reagent Histopaque 1077 (Sigma-Aldrich), according to the manufacturer’s instructions. Briefly, 6mL of peripheral blood were layered on top of 6mL of Histopaque 1077 and centrifuged at 450 × g for 30 min with no break at room temperature. PBMCs were collected from the interphase of the plasma and gradient and washed with 1X phosphate-buffered saline (1X PBS; Life Technologies). PBMCs were then resuspended in RPMI 1640 medium (Life Technologies) supplemented with 10% FBS and 1% of penicillin/streptomycin at a density of 1×106 cells/mL.

ASC speck formation and immunofluorescence

PBMCs were seeded into a four-well Lab-Tek chamber slide (Thermo Fisher Scientific, Rochester, NY) at 5×105 cells/well and cultured at 37°C with 5% CO2 for one hour to allow the monocytes to attach to the slide. Following incubation, the culture medium was gently aspirated and monocytes were incubated with 1µg/mL of lipopolysaccharide (LPS; Escherichia coli 0111:B4; Sigma-Aldrich) alone for 2 hours followed by addition of 20µM of nigericin (catalog number N7142, Sigma-Aldrich) for an additional hour. Non-treated monocytes were used as a negative control.

Amniotic epithelial and mesenchymal cells were plated into four-well chamber slides (BD Falcon, Bedford, MA) at 1×104 cells per well. Amniotic epithelial and mesenchymal cells were cultured overnight (approximately 18 hours) with 1µg/mL of LPS (Escherichia coli 0111:B4) followed by addition of 20µM of nigericin for an additional hour.

Next, all of the cells were fixed using 4% paraformaldehyde (Electron Microscopy Sciences, Hatfield, PA) for 20 min at room temperature, rinsed with 1X PBS, and permeabilized using 0.25% Triton X-100 (Promega, Madison, WI) for 5 min at room temperature. Prior to staining, non-specific antibody interactions were blocked using serum-free protein blocker (Cat#X09090; DAKO North America, Carpinteria, CA) for 30 min at room temperature. Cells were then stained with a rabbit anti-human ASC (Cat#AG-25B-0006-C100; Adipogen, San Diego, CA) antibody at room temperature for one hour. Rabbit IgG was used as a negative control. Following staining, cells were washed with 1X PBS containing 0.1% Tween 20 (PBST) (Sigma-Aldrich). After blocking for 10 min with 10% goat serum (KPL, Gaithersburg, MD), secondary goat anti-rabbit IgG–Alexa Fluor 594 (Life Technologies) was added and cells were incubated for one hour at room temperature in the dark. Finally, cells were washed with 1X PBST and mounted using ProLong Diamond Antifade Mountant with DAPI (Life Technologies). Immunofluorescence was visualized using a Zeiss LSM 780 laser scanning confocal microscope (Carl Zeiss Microscopy, Jena, Germany) at the Microscopy, Imaging, and Cytometry Resources Core at the Wayne State University School of Medicine (http://micr.med.wayne.edu/).

Statistical Analysis

Statistical analysis was performed using SPSS version 19 (IBM Corp, Armonk, NY, USA) or GraphPad Prism (La Jolla, CA, USA) software. Normality tests were used to determine whether the data were normally distributed. Mann-Whitney U tests were used to compare continuous non-parametric variables between groups. Fisher’s exact tests were used to compare proportions between groups. Unpaired t tests were used to compare continuous parametric variables. A p value of ≤0.05 was considered statistically significant.

RESULTS

Characteristics of the study population

A total of 41 amniotic fluid samples from women who underwent transabdominal amniocentesis with (n=17) or without (n=24) spontaneous labor at term were included in this study. Demographic and clinical characteristics of the study population are displayed in Table 1. There were no significant differences in the median maternal age, body mass index (BMI), and gestational age at delivery between the groups. Most of the women included in this study were African-American and delivered healthy neonates (Apgar score ≥8 [190, 191, 192]). All of the women who underwent spontaneous labor at term delivered vaginally (59%, 10/17) and all of the women who delivered at term in the absence of labor underwent a cesarean section (100%, 24/24). The median gestational age at amniocentesis was modestly shorter in the term no labor group [38.35 weeks (37.15-39)] than in the spontaneous labor at term group [39.3 (37.8-39.9) weeks] (p=0.044). Acute histologic chorioamnionitis was more frequent in women who underwent spontaneous labor at term (29.4%, 5/17) than in those who delivered at term in the absence of labor (0%, 0/24) (p=0.008).

Table 1.

Clinical and demographic characteristics of the study population

Term no labor
(N=24)
Spontaneous labor at term
(N=17)
P-value
Maternal age (years)a 26.5 (22.5-29.75) 24 (22-28.5) 0.338
Body mass index (kg/m2)a 33.3 (25.3-36.6) 27.3 (20.4-34) 0.075
Raceb
 African-American 70.8% (17/24) 94% (16/17) 0.11
 Caucasian 20.8% (5/24) 6% (1/17) 0.372
 Hispanic 4.2% (1/24) 0 1
 Other 4.2% (1/24) 0 1
Gestational age at amniocentesis (weeks)a 38.35 (37.15-39) 39.3 (37.8-39.9) 0.044
Delivery routeb
 Vaginal 0 59% (10/17) <0.0001
 Cesarean section 100% (24/24) 41% (7/17) <0.0001
Gestational age at delivery (weeks)a 38.5 (37.4-39.08) 39.3 (37.9-39.9) 0.051
Birthweighta 3158 (2991-3445) 3080 (2845-3378) 0.121
Apgar score at 1 mina 9 (8-9) 8 (8-9) 0.597
Apgar score at 5 mina 9 (9-9) 9 (9-9) 0.831
Histopathological placental findingsb
 Acute histologic chorioamnionitis 0 29.4% (5/17) 0.008
 Acute funisitis 4.2% (1/24) 29.4% (5/17) 0.065

Data are given as median (interquartile range) and percentage (n/N)

a

Mann-Whitney U test.

b

Fisher’s exact test.

Amniotic fluid ASC concentration is increased in women with spontaneous labor at term

ASC is a cytoplasmic protein; however, upon inflammasome activation, specks are formed and can be released to the extracellular space and remain stable for extended periods of time [152]. As readout for inflammasome activation, we determined the presence of extracellular ASC protein in the amniotic fluid. All of the amniotic fluid samples from women who delivered at term (41/41) had detectable levels of the extracellular ASC protein (Figure 1). Yet, the amniotic fluid concentration of ASC was greater in women who underwent spontaneous labor at term than in those who delivered at term in the absence of labor (Figure 1, p=0.01).

Figure 1.

Figure 1

Amniotic fluid ASC concentrations (ng/mL) in women who delivered at term in the absence of labor (n= 24) and women who underwent spontaneous labor at term (n=17). Red line represents the mean ± SEM.

Amniotic epithelial and mesenchymal cells form intra- and extracellular ASC specks in vitro

First, we evaluated whether primary cultures of amniotic epithelial and mesenchymal cells could form intracellular ASC specks in vitro. Upon inflammasome stimulation (LPS+ nigericin), ASC speck formation was observed in monocytes (positive controls, Figure 2A–B) as well as in amniotic epithelial and mesenchymal cells (Figure 2C–D). In monocytes, ASC specks were observed in the cytoplasm as a large red dot (arrows, Figure 2B). Isotype controls did not show any signal, as expected (Figure 2A). In amniotic mesenchymal cells, the ASC protein was observed in the cytoplasm and nucleus (yellow arrow, Figure 2D) and forming ASC filaments (white arrow, Figure 2D), which are created prior to ASC speck formation and inflammasome activation [193, 194, 195, 196, 197]. Isotype controls did not show any signal, as expected (Figure 2C). In the same fashion, the ASC protein in amniotic epithelial cells was observed in the cytoplasm and nucleus (yellow arrow, Figure 2F) and forming ASC speck-like structures (white arrow, Figure 2F). Isotype controls did not show any signal, as expected (Figure 2E). These results show that amniotic epithelial and mesenchymal cells are capable of forming intracellular ASC specks upon inflammasome activation.

Figure 2.

Figure 2

ASC speck formation in amniotic epithelial and mesenchymal primary cells. Monocytes and amniotic cells were incubated with lipopolysaccharide (1µg/mL) and nigericin (20µM) and ASC speck formation was assessed by confocal microscopy. (A-B) Monocytes isolated from the peripheral blood of healthy individuals were used as positive controls (n=3). (C-F) Amniotic epithelial and mesenchymal cells were isolated from the chorioamniotic membranes from women who delivered at term in the absence of labor (n=3). Right panel: ASC signal is shown in red. Left panel: isotype controls. The blue signal is DAPI (nuclei). White arrows show ASC specks/filaments and yellow arrows show cytoplasmic/nuclear ASC. 400× magnification. ASC, apoptosis-associated speck-like protein containing a CARD.

DISCUSSION

Principal findings of the study

In the current study, we report that: 1) extracellular ASC was present in the amniotic fluid of women who delivered at term; 2) amniotic fluid ASC concentrations were greater in women who underwent spontaneous labor at term than in those who delivered at term in the absence of labor; and 3) amniotic epithelial and mesenchymal cells formed intracellular ASC specks and filaments in vitro.

A role for the inflammasome in spontaneous labor at term

Evidence supporting the participation of the inflammasome in the mechanisms of spontaneous labor at term [141] includes the following: 1) caspase 1, the predominant inflammasome-activated caspase [98, 198], is present in the amniotic fluid and its concentrations are higher in women with spontaneous labor at term than in those without labor [142]; 2) amniotic fluid concentrations of IL-1β are increased in women who underwent spontaneous labor at term compared to those who delivered at term in the absence of labor [14, 15, 16, 42]; 3) amniotic fluid concentrations of IL-18 are greater in term pregnancies than in the second trimester [199]; 4) the chorioamniotic membranes from women who underwent spontaneous labor at term display increased expression of the inflammasome sensor molecule NLRP3 (NLR Family Pyrin Domain Containing 3 protein or cryopyrin), contain high amounts of active forms of caspase-1, and release elevated concentrations of mature IL-1β compared to those from women who delivered at term in the absence of labor [143]; and 5) ASC/caspase-1 complexes (i.e. inflammasome assembly) are greater in the chorioamniotic membranes and choriodecidual leukocytes from women who underwent spontaneous labor at term than in those without labor. Herein, we provide further in vivo evidence that there is inflammasome activation in the amniotic cavity of women who underwent spontaneous labor at term. Specifically, we showed that amniotic epithelial and mesenchymal cells can form ASC speck-like structures and filaments, which are created prior to ASC speck formation upon inflammasome activation [193, 194, 195, 196, 197]. Such ASC specks can then be released and found extracellularly in the amniotic cavity, where they are elevated in women who underwent spontaneous labor at term. In the amniotic cavity, extracellular ASC can function as a danger signal to amplify the sterile inflammatory process that accompanies the process of labor [3, 4, 5, 6, 7, 8, 9, 10]. Collectively, these findings provide solid evidence that there is inflammasome activation during the physiological process of spontaneous labor at term.

A role for inflammasome-processed IL-1β in spontaneous labor at term

The biochemical function of the inflammasome is to activate caspase-1 [127, 128, 129, 133, 134, 137] which, in turn, will lead to the maturation of IL-1β [92, 94, 98, 99, 102, 113, 118, 125, 126, 200]. In line with the findings reported herein, we previously showed that the chorioamniotic membranes express IL1B and release the mature protein form, and that such a process was increased in the chorioamniotic membranes from women who had undergone spontaneous labor at term than in those without labor [143]. The biological functions of bioactive IL-1β during the process of labor comprise: 1) stimulation of prostaglandin biosynthesis by the human amnion [201], decidual cells [202], chorioamniotic membranes [203], and myometrial cells [204, 205], 2) induction of the expression of cyclooxygenase-2 by human myometrial cells [206], 3) upregulation of the expression of matrix metalloproteinases (e.g. MMP-9) by human cervical smooth muscle cells [207] and the amnion [208]; and 4) induction of pro-inflammatory cytokines by decidual cells [209]. Indeed, systemic administration of IL-1β induces preterm labor and birth in mice [17, 210] and monkeys [211, 212, 213, 214, 215, 216, 217, 218], confirming the central role of this cytokine in the mechanisms responsible for labor at term.

CONCLUSION

The adaptor protein of the inflammasome ASC, which is released upon inflammasome activation, is detectable in the amniotic fluid of women who delivered at term and increased in those who underwent spontaneous labor at term. Amniotic epithelial and mesenchymal cells are capable of forming ASC specks and filaments in vitro, suggesting that these cells are a source of extracellular ASC in the amniotic fluid. These findings provide in vivo evidence that there is inflammasome activation in the amniotic cavity during the physiological process of labor at term.

Acknowledgments

This research was supported, in part, by the Perinatology Research Branch (PRB), 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 (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. We gratefully acknowledge the PRB Translational Research laboratory for their contributions to the execution of this study. We thank the physicians and nurses from the Center for Advanced Obstetrical Care and Research and the Intrapartum Unit for their help in collecting human samples. We also thank staff members of the PRB Clinical Laboratory and the PRB Histology/Pathology Unit for the processing and examination of the pathological sections.

Footnotes

DECLARATION OF INTEREST STATEMENT

The authors report no conflicts of interest.

References

  • 1.Romero R, Nores J, Mazor M, Sepulveda W, Oyarzun E, Parra M, Insunza A, Montiel F, Behnke E, Cassell GH. Microbial invasion of the amniotic cavity during term labor. Prevalence and clinical significance. J Reprod Med. 1993;38:543–8. Epub 1993/07/01. [PubMed] [Google Scholar]
  • 2.Seong HS, Lee SE, Kang JH, Romero R, Yoon BH. The frequency of microbial invasion of the amniotic cavity and histologic chorioamnionitis in women at term with intact membranes in the presence or absence of labor. Am J Obstet Gynecol. 2008;199:375 e1–5. doi: 10.1016/j.ajog.2008.06.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Romero R, Espinoza J, Goncalves LF, Kusanovic JP, Friel LA, Nien JK. Inflammation in preterm and term labour and delivery. Semin Fetal Neonatal Med. 2006;11:317–26. doi: 10.1016/j.siny.2006.05.001. Epub 2006/07/15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Haddad R, Tromp G, Kuivaniemi H, Chaiworapongsa T, Kim YM, Mazor M, Romero R. Human spontaneous labor without histologic chorioamnionitis is characterized by an acute inflammation gene expression signature. Am J Obstet Gynecol. 2006;195:394 e1–24. doi: 10.1016/j.ajog.2005.08.057. Epub 2006/08/08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Hassan SS, Romero R, Haddad R, Hendler I, Khalek N, Tromp G, Diamond MP, Sorokin Y, Malone J., Jr The transcriptome of the uterine cervix before and after spontaneous term parturition. Am J Obstet Gynecol. 2006;195:778–86. doi: 10.1016/j.ajog.2006.06.021. Epub 2006/09/05. [DOI] [PubMed] [Google Scholar]
  • 6.Norman JE, Bollapragada S, Yuan M, Nelson SM. Inflammatory pathways in the mechanism of parturition. BMC Pregnancy Childbirth. 2007;7(Suppl 1):S7. doi: 10.1186/1471-2393-7-S1-S7. Epub 2007/06/19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Hassan SS, Romero R, Tarca AL, Nhan-Chang CL, Vaisbuch E, Erez O, Mittal P, Kusanovic JP, Mazaki-Tovi S, Yeo L, Draghici S, Kim JS, Uldbjerg N, Kim CJ. 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;22:1183–93. doi: 10.3109/14767050903353216. Epub 2009/11/04. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.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;200:104 e1–11. doi: 10.1016/j.ajog.2008.08.032. Epub 2009/01/06. [DOI] [PubMed] [Google Scholar]
  • 9.Mittal P, Romero R, Tarca AL, Gonzalez J, Draghici S, Xu Y, Dong Z, Nhan-Chang CL, Chaiworapongsa T, Lye S, Kusanovic JP, Lipovich L, Mazaki-Tovi S, Hassan SS, Mesiano S, Kim CJ. Characterization of the myometrial transcriptome and biological pathways of spontaneous human labor at term. J Perinat Med. 2010;38:617–43. doi: 10.1515/JPM.2010.097. Epub 2010/07/16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Nhan-Chang CL, Romero R, Tarca AL, Mittal P, Kusanovic JP, Erez O, Mazaki-Tovi S, Chaiworapongsa T, Hotra J, Than NG, Kim JS, Hassan SS, Kim CJ. Characterization of the transcriptome of chorioamniotic membranes at the site of rupture in spontaneous labor at term. Am J Obstet Gynecol. 2010;202:462 e1–41. doi: 10.1016/j.ajog.2010.02.045. Epub 2010/05/11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Arbib N, Aviram A, Gabbay Ben-Ziv R, Sneh O, Yogev Y, Hadar E. The effect of labor and delivery on white blood cell count. J Matern Fetal Neonatal Med. 2016;29:2904–8. doi: 10.3109/14767058.2015.1110572. [DOI] [PubMed] [Google Scholar]
  • 12.Herrera CA, Stoerker J, Carlquist J, Stoddard GJ, Jackson M, Esplin S, Rose NC. Cell-free DNA, inflammation, and the initiation of spontaneous term labor. Am J Obstet Gynecol. 2017;217:583 e1–e8. doi: 10.1016/j.ajog.2017.05.027. [DOI] [PubMed] [Google Scholar]
  • 13.Phillippe M. The link between cell-free DNA, inflammation and the initiation of spontaneous labor at term. Am J Obstet Gynecol. 2017;217:501–2. doi: 10.1016/j.ajog.2017.09.003. [DOI] [PubMed] [Google Scholar]
  • 14.Romero R, Brody DT, Oyarzun E, Mazor M, Wu YK, Hobbins JC, Durum SK. Infection and labor. III. Interleukin-1: a signal for the onset of parturition. Am J Obstet Gynecol. 1989;160:1117–23. doi: 10.1016/0002-9378(89)90172-5. Epub 1989/05/01. [DOI] [PubMed] [Google Scholar]
  • 15.Romero R, Parvizi ST, Oyarzun E, Mazor M, Wu YK, Avila C, Athanassiadis AP, Mitchell MD. Amniotic fluid interleukin-1 in spontaneous labor at term. J Reprod Med. 1990;35:235–8. Epub 1990/03/01. [PubMed] [Google Scholar]
  • 16.Romero R, Mazor M, Brandt F, Sepulveda W, Avila C, Cotton DB, Dinarello CA. Interleukin-1 alpha and interleukin-1 beta in preterm and term human parturition. Am J Reprod Immunol. 1992;27:117–23. doi: 10.1111/j.1600-0897.1992.tb00737.x. Epub 1992/04/01. [DOI] [PubMed] [Google Scholar]
  • 17.Romero R, Sepulveda W, Mazor M, Brandt F, Cotton DB, Dinarello CA, Mitchell MD. The natural interleukin-1 receptor antagonist in term and preterm parturition. Am J Obstet Gynecol. 1992;167:863–72. doi: 10.1016/s0002-9378(12)80003-2. Epub 1992/10/01. [DOI] [PubMed] [Google Scholar]
  • 18.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;166:1576–87. doi: 10.1016/0002-9378(92)91636-o. Epub 1992/05/01. [DOI] [PubMed] [Google Scholar]
  • 19.Saito S, Kasahara T, Kato Y, Ishihara Y, Ichijo M. Elevation of amniotic fluid interleukin 6 (IL-6), IL-8 and granulocyte colony stimulating factor (G-CSF) in term and preterm parturition. Cytokine. 1993;5:81–8. doi: 10.1016/1043-4666(93)90027-3. Epub 1993/01/01. [DOI] [PubMed] [Google Scholar]
  • 20.Opsjln SL, Wathen NC, Tingulstad S, Wiedswang G, Sundan A, Waage A, Austgulen R. Tumor necrosis factor, interleukin-1, and interleukin-6 in normal human pregnancy. Am J Obstet Gynecol. 1993;169:397–404. doi: 10.1016/0002-9378(93)90096-2. Epub 1993/08/01. [DOI] [PubMed] [Google Scholar]
  • 21.Romero R, Gomez R, Galasso M, Mazor M, Berry SM, Quintero RA, Cotton DB. The natural interleukin-1 receptor antagonist in the fetal, maternal, and amniotic fluid compartments: the effect of gestational age, fetal gender, and intrauterine infection. Am J Obstet Gynecol. 1994;171:912–21. doi: 10.1016/s0002-9378(94)70058-3. Epub 1994/10/01. [DOI] [PubMed] [Google Scholar]
  • 22.Andrews WW, Hauth JC, Goldenberg RL, Gomez R, Romero R, Cassell GH. Amniotic fluid interleukin-6: correlation with upper genital tract microbial colonization and gestational age in women delivered after spontaneous labor versus indicated delivery. Am J Obstet Gynecol. 1995;173:606–12. doi: 10.1016/0002-9378(95)90290-2. Epub 1995/08/01. [DOI] [PubMed] [Google Scholar]
  • 23.Olah KS, Vince GS, Neilson JP, Deniz G, Johnson PM. Interleukin-6, interferon-gamma, interleukin-8, and granulocyte-macrophage colony stimulating factor levels in human amniotic fluid at term. J Reprod Immunol. 1996;32:89–98. doi: 10.1016/s0165-0378(96)00990-4. Epub 1996/11/01. [DOI] [PubMed] [Google Scholar]
  • 24.Maymon E, Ghezzi F, Edwin SS, Mazor M, Yoon BH, Gomez R, Romero R. The tumor necrosis factor alpha and its soluble receptor profile in term and preterm parturition. Am J Obstet Gynecol. 1999;181:1142–8. doi: 10.1016/s0002-9378(99)70097-9. Epub 1999/11/16. [DOI] [PubMed] [Google Scholar]
  • 25.Bowen JM, Chamley L, Keelan JA, Mitchell MD. Cytokines of the placenta and extra-placental membranes: roles and regulation during human pregnancy and parturition. Placenta. 2002;23:257–73. doi: 10.1053/plac.2001.0782. Epub 2002/04/24. [DOI] [PubMed] [Google Scholar]
  • 26.Kemp B, Winkler M, Maas A, Maul H, Ruck P, Reineke T, Rath W. Cytokine concentrations in the amniotic fluid during parturition at term: correlation to lower uterine segment values and to labor. Acta Obstet Gynecol Scand. 2002;81:938–42. doi: 10.1034/j.1600-0412.2002.811007.x. Epub 2002/10/09. [DOI] [PubMed] [Google Scholar]
  • 27.Keelan JA, Blumenstein M, Helliwell RJ, Sato TA, Marvin KW, Mitchell MD. Cytokines, prostaglandins and parturition–a review. Placenta. 2003;24(Suppl A):S33–46. doi: 10.1053/plac.2002.0948. Epub 2003/07/05. [DOI] [PubMed] [Google Scholar]
  • 28.Gotsch F, Romero R, Kusanovic JP, Erez O, Espinoza J, Kim CJ, Vaisbuch E, Than NG, Mazaki-Tovi S, Chaiworapongsa T, Mazor M, Yoon BH, Edwin S, Gomez R, Mittal P, Hassan SS, Sharma S. The anti-inflammatory limb of the immune response in preterm labor, intra-amniotic infection/inflammation, and spontaneous parturition at term: a role for interleukin-10. J Matern Fetal Neonatal Med. 2008;21:529–47. doi: 10.1080/14767050802127349. Epub 2008/07/09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.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;165:813–20. doi: 10.1016/0002-9378(91)90422-n. Epub 1991/10/01. [DOI] [PubMed] [Google Scholar]
  • 30.Romero R, Gomez R, Galasso M, Munoz H, Acosta L, Yoon BH, Svinarich D, Cotton DB. Macrophage inflammatory protein-1 alpha in term and preterm parturition: effect of microbial invasion of the amniotic cavity. Am J Reprod Immunol. 1994;32:108–13. doi: 10.1111/j.1600-0897.1994.tb01101.x. Epub 1994/09/01. [DOI] [PubMed] [Google Scholar]
  • 31.Dudley DJ, Hunter C, Mitchell MD, Varner MW. Elevations of amniotic fluid macrophage inflammatory protein-1 alpha concentrations in women during term and preterm labor. Obstet Gynecol. 1996;87:94–8. doi: 10.1016/0029-7844(95)00366-5. Epub 1996/01/01. [DOI] [PubMed] [Google Scholar]
  • 32.Athayde N, Romero R, Maymon E, Gomez R, Pacora P, Araneda H, Yoon BH. A role for the novel cytokine RANTES in pregnancy and parturition. Am J Obstet Gynecol. 1999;181:989–94. doi: 10.1016/s0002-9378(99)70337-6. Epub 1999/10/16. [DOI] [PubMed] [Google Scholar]
  • 33.Esplin MS, Romero R, Chaiworapongsa T, Kim YM, Edwin S, Gomez R, Gonzalez R, Adashi EY. Amniotic fluid levels of immunoreactive monocyte chemotactic protein-1 increase during term parturition. J Matern Fetal Neonatal Med. 2003;14:51–6. doi: 10.1080/jmf.14.1.51.56. Epub 2003/10/18. [DOI] [PubMed] [Google Scholar]
  • 34.Hamill N, Romero R, Gotsch F, Kusanovic JP, Edwin S, Erez O, Than NG, Mittal P, Espinoza J, Friel LA, Vaisbuch E, Mazaki-Tovi S, Hassan SS. Exodus-1 (CCL20): evidence for the participation of this chemokine in spontaneous labor at term, preterm labor, and intrauterine infection. J Perinat Med. 2008;36:217–27. doi: 10.1515/JPM.2008.034. Epub 2008/06/26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Unal ER, Cierny JT, Roedner C, Newman R, Goetzl L. Maternal inflammation in spontaneous term labor. Am J Obstet Gynecol. 2011;204:223 e1–5. doi: 10.1016/j.ajog.2011.01.002. Epub 2011/03/08. [DOI] [PubMed] [Google Scholar]
  • 36.Cierny JT, Unal ER, Flood P, Rhee KY, Praktish A, Olson TH, Goetzl L. Maternal inflammatory markers and term labor performance. Am J Obstet Gynecol. 2014;210:447 e1–6. doi: 10.1016/j.ajog.2013.11.038. Epub 2013/12/04. [DOI] [PubMed] [Google Scholar]
  • 37.Kucukgul S, Ozkan ZS, Yavuzkir S, Ilhan N. Investigation of the maternal and cord plasma levels of IL-1 beta, TNF-alpha and VEGF in early membrane rupture. J Matern Fetal Neonatal Med. 2016;29:2157–60. doi: 10.3109/14767058.2015.1077511. [DOI] [PubMed] [Google Scholar]
  • 38.Taniguchi T, Matsuzaki N, Kameda T, Shimoya K, Jo T, Saji F, Tanizawa O. The enhanced production of placental interleukin-1 during labor and intrauterine infection. Am J Obstet Gynecol. 1991;165:131–7. doi: 10.1016/0002-9378(91)90241-i. Epub 1991/07/01. [DOI] [PubMed] [Google Scholar]
  • 39.Fidel PL, Jr, Romero R, Ramirez M, Cutright J, Edwin SS, LaMarche S, Cotton DB, Mitchell MD. Interleukin-1 receptor antagonist (IL-1ra) production by human amnion, chorion, and decidua. Am J Reprod Immunol. 1994;32:1–7. doi: 10.1111/j.1600-0897.1994.tb00872.x. Epub 1994/08/01. [DOI] [PubMed] [Google Scholar]
  • 40.Dudley DJ, Collmer D, Mitchell MD, Trautman MS. Inflammatory cytokine mRNA in human gestational tissues: implications for term and preterm labor. J Soc Gynecol Investig. 1996;3:328–35. Epub 1996/11/01. [PubMed] [Google Scholar]
  • 41.Ammala M, Nyman T, Salmi A, Rutanen EM. The interleukin-1 system in gestational tissues at term: effect of labour. Placenta. 1997;18:717–23. doi: 10.1016/s0143-4004(97)90014-x. Epub 1997/11/19. [DOI] [PubMed] [Google Scholar]
  • 42.Keelan JA, Marvin KW, Sato TA, Coleman M, McCowan LM, Mitchell MD. Cytokine abundance in placental tissues: evidence of inflammatory activation in gestational membranes with term and preterm parturition. Am J Obstet Gynecol. 1999;181:1530–6. doi: 10.1016/s0002-9378(99)70400-x. Epub 1999/12/22. [DOI] [PubMed] [Google Scholar]
  • 43.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;66:445–9. doi: 10.1095/biolreprod66.2.445. Epub 2002/01/24. [DOI] [PubMed] [Google Scholar]
  • 44.Lonergan M, Aponso D, Marvin KW, Helliwell RJ, Sato TA, Mitchell MD, Chaiwaropongsa T, Romero R, Keelan JA. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), TRAIL receptors, and the soluble receptor osteoprotegerin in human gestational membranes and amniotic fluid during pregnancy and labor at term and preterm. J Clin Endocrinol Metab. 2003;88:3835–44. doi: 10.1210/jc.2002-021905. Epub 2003/08/14. [DOI] [PubMed] [Google Scholar]
  • 45.Osman I, Young A, Ledingham MA, Thomson AJ, Jordan F, Greer IA, Norman JE. 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;9:41–5. doi: 10.1093/molehr/gag001. Epub 2003/01/17. [DOI] [PubMed] [Google Scholar]
  • 46.Esplin MS, Peltier MR, Hamblin S, Smith S, Fausett MB, Dildy GA, Branch DW, Silver RM, Adashi EY. Monocyte chemotactic protein-1 expression is increased in human gestational tissues during term and preterm labor. Placenta. 2005;26:661–71. doi: 10.1016/j.placenta.2004.09.012. Epub 2005/08/09. [DOI] [PubMed] [Google Scholar]
  • 47.Kim GJ, Romero R, Kuivaniemi H, Tromp G, Haddad R, Kim YM, Kim MR, Nien JK, Hong JS, Espinoza J, Santolaya J, Yoon BH, Mazor M, Kim CJ. Expression of bone morphogenetic protein 2 in normal spontaneous labor at term, preterm labor, and preterm premature rupture of membranes. Am J Obstet Gynecol. 2005;193:1137–43. doi: 10.1016/j.ajog.2005.06.032. Epub 2005/09/15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Kim YM, Romero R, Chaiworapongsa T, Kim GJ, Kim MR, Kuivaniemi H, Tromp G, Espinoza J, Bujold E, Abrahams VM, Mor G. Toll-like receptor-2 and -4 in the chorioamniotic membranes in spontaneous labor at term and in preterm parturition that are associated with chorioamnionitis. Am J Obstet Gynecol. 2004;191:1346–55. doi: 10.1016/j.ajog.2004.07.009. Epub 2004/10/28. [DOI] [PubMed] [Google Scholar]
  • 49.Koga K, Mor G. Toll-like receptors at the maternal-fetal interface in normal pregnancy and pregnancy disorders. Am J Reprod Immunol. 2010;63:587–600. doi: 10.1111/j.1600-0897.2010.00848.x. Epub 2010/04/07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Koga K, Izumi G, Mor G, Fujii T, Osuga Y. Toll-like receptors at the maternal-fetal interface in normal pregnancy and pregnancy complications. Am J Reprod Immunol. 2014;72:192–205. doi: 10.1111/aji.12258. Epub 2014/04/24. [DOI] [PubMed] [Google Scholar]
  • 51.Stephen GL, Lui S, Hamilton SA, Tower CL, Harris LK, Stevens A, Jones RL. Transcriptomic profiling of human choriodecidua during term labor: inflammation as a key driver of labor. Am J Reprod Immunol. 2015;73:36–55. doi: 10.1111/aji.12328. Epub 2014/10/07. [DOI] [PubMed] [Google Scholar]
  • 52.Gomez-Lopez N, Tong WC, Arenas-Hernandez M, Tanaka S, Hajar O, Olson DM, Taggart MJ, Mitchell BF. Chemotactic activity of gestational tissues through late pregnancy, term labor, and RU486-induced preterm labor in Guinea pigs. Am J Reprod Immunol. 2015;73:341–52. doi: 10.1111/aji.12333. Epub 2014/10/21. [DOI] [PubMed] [Google Scholar]
  • 53.Liggins G. Cervical ripening as an inflammatory reaction. In: Ellwood E, Anderson A, editors. The cervix in pregnancy and labor: clinical and biochemical investigations. Edinburgh: Churchill Livingstone; 1981. pp. 1–9. [Google Scholar]
  • 54.Bokstrom H, Brannstrom M, Alexandersson M, Norstrom A. Leukocyte subpopulations in the human uterine cervical stroma at early and term pregnancy. Hum Reprod. 1997;12:586–90. doi: 10.1093/humrep/12.3.586. Epub 1997/03/01. [DOI] [PubMed] [Google Scholar]
  • 55.Mackler AM, Iezza G, Akin MR, McMillan P, Yellon SM. Macrophage trafficking in the uterus and cervix precedes parturition in the mouse. Biol Reprod. 1999;61:879–83. doi: 10.1095/biolreprod61.4.879. Epub 1999/09/24. [DOI] [PubMed] [Google Scholar]
  • 56.Kelly RW. Inflammatory mediators and cervical ripening. J Reprod Immunol. 2002;57:217–24. doi: 10.1016/s0165-0378(02)00007-4. Epub 2002/10/19. [DOI] [PubMed] [Google Scholar]
  • 57.Sakamoto Y, Moran P, Bulmer JN, Searle RF, Robson SC. Macrophages and not granulocytes are involved in cervical ripening. J Reprod Immunol. 2005;66:161–73. doi: 10.1016/j.jri.2005.04.005. Epub 2005/07/28. [DOI] [PubMed] [Google Scholar]
  • 58.Yellon SM, Ebner CA, Sugimoto Y. Parturition and recruitment of macrophages in cervix of mice lacking the prostaglandin F receptor. Biol Reprod. 2008;78:438–44. doi: 10.1095/biolreprod.107.063404. Epub 2007/11/16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Yellon SM, Oshiro BT, Chhaya TY, Lechuga TJ, Dias RM, Burns AE, Force L, Apostolakis EM. Remodeling of the cervix and parturition in mice lacking the progesterone receptor B isoform. Biol Reprod. 2011;85:498–502. doi: 10.1095/biolreprod.111.091983. Epub 2011/05/27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Clyde LA, Lechuga TJ, Ebner CA, Burns AE, Kirby MA, Yellon SM. Transection of the pelvic or vagus nerve forestalls ripening of the cervix and delays birth in rats. Biol Reprod. 2011;84:587–94. doi: 10.1095/biolreprod.110.086207. Epub 2010/11/26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Payne KJ, Clyde LA, Weldon AJ, Milford TA, Yellon SM. Residency and activation of myeloid cells during remodeling of the prepartum murine cervix. Biol Reprod. 2012;87:106. doi: 10.1095/biolreprod.112.101840. Epub 2012/08/24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Myers DA. The recruitment and activation of leukocytes into the immune cervix: further support that cervical remodeling involves an immune and inflammatory mechanism. Biol Reprod. 2012;87:107. doi: 10.1095/biolreprod.112.105049. Epub 2012/09/29. [DOI] [PubMed] [Google Scholar]
  • 63.Thomson AJ, Telfer JF, Young A, Campbell S, Stewart CJ, Cameron IT, Greer IA, Norman JE. Leukocytes infiltrate the myometrium during human parturition: further evidence that labour is an inflammatory process. Hum Reprod. 1999;14:229–36. Epub 1999/06/22. [PubMed] [Google Scholar]
  • 64.Shynlova O, Tsui P, Dorogin A, Lye SJ. Monocyte chemoattractant protein-1 (CCL-2) integrates mechanical and endocrine signals that mediate term and preterm labor. J Immunol. 2008;181:1470–9. doi: 10.4049/jimmunol.181.2.1470. [DOI] [PubMed] [Google Scholar]
  • 65.Shynlova O, Tsui P, Jaffer S, Lye SJ. Integration of endocrine and mechanical signals in the regulation of myometrial functions during pregnancy and labour. Eur J Obstet Gynecol Reprod Biol. 2009;144(Suppl 1):S2–10. doi: 10.1016/j.ejogrb.2009.02.044. [DOI] [PubMed] [Google Scholar]
  • 66.Hamilton S, Oomomian Y, Stephen G, Shynlova O, Tower CL, Garrod A, Lye SJ, Jones RL. Macrophages infiltrate the human and rat decidua during term and preterm labor: evidence that decidual inflammation precedes labor. Biol Reprod. 2012;86:39. doi: 10.1095/biolreprod.111.095505. Epub 2011/10/21. [DOI] [PubMed] [Google Scholar]
  • 67.Shynlova O, Nedd-Roderique T, Li Y, Dorogin A, Nguyen T, Lye SJ. Infiltration of myeloid cells into decidua is a critical early event in the labour cascade and post-partum uterine remodelling. J Cell Mol Med. 2013;17:311–24. doi: 10.1111/jcmm.12012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Shynlova O, Lee YH, Srikhajon K, Lye SJ. Physiologic uterine inflammation and labor onset: integration of endocrine and mechanical signals. Reprod Sci. 2013;20:154–67. doi: 10.1177/1933719112446084. [DOI] [PubMed] [Google Scholar]
  • 69.Arenas-Hernandez M, Romero R, St Louis D, Hassan SS, Kaye EB, Gomez-Lopez N. An imbalance between innate and adaptive immune cells at the maternal-fetal interface occurs prior to endotoxin-induced preterm birth. Cell Mol Immunol. 2016;13:462–73. doi: 10.1038/cmi.2015.22. Epub 2015/04/08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Osman I, Young A, Jordan F, Greer IA, Norman JE. Leukocyte density and proinflammatory mediator expression in regional human fetal membranes and decidua before and during labor at term. J Soc Gynecol Investig. 2006;13:97–103. doi: 10.1016/j.jsgi.2005.12.002. Epub 2006/01/31. [DOI] [PubMed] [Google Scholar]
  • 71.Gomez-Lopez N, Estrada-Gutierrez G, Jimenez-Zamudio L, Vega-Sanchez R, Vadillo-Ortega F. Fetal membranes exhibit selective leukocyte chemotaxic activity during human labor. J Reprod Immunol. 2009;80:122–31. doi: 10.1016/j.jri.2009.01.002. Epub 2009/05/02. [DOI] [PubMed] [Google Scholar]
  • 72.Gomez-Lopez N, Vadillo-Perez L, Hernandez-Carbajal A, Godines-Enriquez M, Olson DM, Vadillo-Ortega F. Specific inflammatory microenvironments in the zones of the fetal membranes at term delivery. Am J Obstet Gynecol. 2011;205:235 e15–24. doi: 10.1016/j.ajog.2011.04.019. Epub 2011/07/19. [DOI] [PubMed] [Google Scholar]
  • 73.Gomez-Lopez N, Vadillo-Perez L, Nessim S, Olson DM, Vadillo-Ortega F. Choriodecidua and amnion exhibit selective leukocyte chemotaxis during term human labor. Am J Obstet Gynecol. 2011;204:364 e9–16. doi: 10.1016/j.ajog.2010.11.010. Epub 2011/02/08. [DOI] [PubMed] [Google Scholar]
  • 74.Gomez-Lopez N, Vega-Sanchez R, Castillo-Castrejon M, Romero R, Cubeiro-Arreola K, Vadillo-Ortega F. Evidence for a role for the adaptive immune response in human term parturition. Am J Reprod Immunol. 2013;69:212–30. doi: 10.1111/aji.12074. Epub 2013/01/26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Xu Y, Plazyo O, Romero R, Hassan SS, Gomez-Lopez N. Isolation of Leukocytes from the Human Maternal-fetal Interface. J Vis Exp. 2015:e52863. doi: 10.3791/52863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Chen GY, Nunez G. Sterile inflammation: sensing and reacting to damage. Nat Rev Immunol. 2010;10:826–37. doi: 10.1038/nri2873. Epub 2010/11/23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Rubartelli A, Lotze MT. Inside, outside, upside down: damage-associated molecular-pattern molecules (DAMPs) and redox. Trends Immunol. 2007;28:429–36. doi: 10.1016/j.it.2007.08.004. Epub 2007/09/12. [DOI] [PubMed] [Google Scholar]
  • 78.Lotze MT, Zeh HJ, Rubartelli A, Sparvero LJ, Amoscato AA, Washburn NR, Devera ME, Liang X, Tor M, Billiar T. The grateful dead: damage-associated molecular pattern molecules and reduction/oxidation regulate immunity. Immunol Rev. 2007;220:60–81. doi: 10.1111/j.1600-065X.2007.00579.x. Epub 2007/11/06. [DOI] [PubMed] [Google Scholar]
  • 79.Oppenheim JJ, Yang D. Alarmins: chemotactic activators of immune responses. Curr Opin Immunol. 2005;17:359–65. doi: 10.1016/j.coi.2005.06.002. Epub 2005/06/16. [DOI] [PubMed] [Google Scholar]
  • 80.Martinon F, Petrilli V, Mayor A, Tardivel A, Tschopp J. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature. 2006;440:237–41. doi: 10.1038/nature04516. Epub 2006/01/13. [DOI] [PubMed] [Google Scholar]
  • 81.Mariathasan S, Weiss DS, Newton K, McBride J, O’Rourke K, Roose-Girma M, Lee WP, Weinrauch Y, Monack DM, Dixit VM. Cryopyrin activates the inflammasome in response to toxins and ATP. Nature. 2006;440:228–32. doi: 10.1038/nature04515. Epub 2006/01/13. [DOI] [PubMed] [Google Scholar]
  • 82.Dostert C, Petrilli V, Van Bruggen R, Steele C, Mossman BT, Tschopp J. Innate immune activation through Nalp3 inflammasome sensing of asbestos and silica. Science. 2008;320:674–7. doi: 10.1126/science.1156995. Epub 2008/04/12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Cassel SL, Eisenbarth SC, Iyer SS, Sadler JJ, Colegio OR, Tephly LA, Carter AB, Rothman PB, Flavell RA, Sutterwala FS. The Nalp3 inflammasome is essential for the development of silicosis. Proc Natl Acad Sci USA. 2008;105:9035–40. doi: 10.1073/pnas.0803933105. Epub 2008/06/26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Hornung V, Bauernfeind F, Halle A, Samstad EO, Kono H, Rock KL, Fitzgerald KA, Latz E. Silica crystals and aluminum salts activate the NALP3 inflammasome through phagosomal destabilization. Nat Immunol. 2008;9:847–56. doi: 10.1038/ni.1631. Epub 2008/07/08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Cassel SL, Joly S, Sutterwala FS. The NLRP3 inflammasome: a sensor of immune danger signals. Semin Immunol. 2009;21:194–8. doi: 10.1016/j.smim.2009.05.002. Epub 2009/06/09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Yamasaki K, Muto J, Taylor KR, Cogen AL, Audish D, Bertin J, Grant EP, Coyle AJ, Misaghi A, Hoffman HM, Gallo RL. NLRP3/cryopyrin is necessary for interleukin-1beta (IL-1beta) release in response to hyaluronan, an endogenous trigger of inflammation in response to injury. J Biol Chem. 2009;284:12762–71. doi: 10.1074/jbc.M806084200. Epub 2009/03/05. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Cassel SL, Sutterwala FS. Sterile inflammatory responses mediated by the NLRP3 inflammasome. Eur J Immunol. 2010;40:607–11. doi: 10.1002/eji.200940207. Epub 2010/03/05. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Leemans JC, Cassel SL, Sutterwala FS. Sensing damage by the NLRP3 inflammasome. Immunol Rev. 2011;243:152–62. doi: 10.1111/j.1600-065X.2011.01043.x. Epub 2011/09/03. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Nakahira K, Haspel JA, Rathinam VA, Lee SJ, Dolinay T, Lam HC, Englert JA, Rabinovitch M, Cernadas M, Kim HP, Fitzgerald KA, Ryter SW, Choi AM. Autophagy proteins regulate innate immune responses by inhibiting the release of mitochondrial DNA mediated by the NALP3 inflammasome. Nat Immunol. 2011;12:222–30. doi: 10.1038/ni.1980. Epub 2010/12/15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Iyer SS, He Q, Janczy JR, Elliott EI, Zhong Z, Olivier AK, Sadler JJ, Knepper-Adrian V, Han R, Qiao L, Eisenbarth SC, Nauseef WM, Cassel SL, Sutterwala FS. Mitochondrial cardiolipin is required for Nlrp3 inflammasome activation. Immunity. 2013;39:311–23. doi: 10.1016/j.immuni.2013.08.001. Epub 2013/08/21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.O’Neill LA. Cardiolipin and the Nlrp3 inflammasome. Cell Metab. 2013;18:610–2. doi: 10.1016/j.cmet.2013.10.013. Epub 2013/11/12. [DOI] [PubMed] [Google Scholar]
  • 92.Martinon F, Burns K, Tschopp J. The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol Cell. 2002;10:417–26. doi: 10.1016/s1097-2765(02)00599-3. Epub 2002/08/23. [DOI] [PubMed] [Google Scholar]
  • 93.Petrilli V, Papin S, Tschopp J. The inflammasome. Curr Biol. 2005;15:R581. doi: 10.1016/j.cub.2005.07.049. Epub 2005/08/09. [DOI] [PubMed] [Google Scholar]
  • 94.Ogura Y, Sutterwala FS, Flavell RA. The inflammasome: first line of the immune response to cell stress. Cell. 2006;126:659–62. doi: 10.1016/j.cell.2006.08.002. Epub 2006/08/23. [DOI] [PubMed] [Google Scholar]
  • 95.Sutterwala FS, Ogura Y, Flavell RA. The inflammasome in pathogen recognition and inflammation. J Leukoc Biol. 2007;82:259–64. doi: 10.1189/jlb.1206755. Epub 2007/05/02. [DOI] [PubMed] [Google Scholar]
  • 96.Mariathasan S, Monack DM. Inflammasome adaptors and sensors: intracellular regulators of infection and inflammation. Nat Rev Immunol. 2007;7:31–40. doi: 10.1038/nri1997. Epub 2006/12/23. [DOI] [PubMed] [Google Scholar]
  • 97.Stutz A, Golenbock DT, Latz E. Inflammasomes: too big to miss. J Clin Invest. 2009;119:3502–11. doi: 10.1172/JCI40599. Epub 2009/12/04. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Franchi L, Eigenbrod T, Munoz-Planillo R, Nunez G. The inflammasome: a caspase-1-activation platform that regulates immune responses and disease pathogenesis. Nat Immunol. 2009;10:241–7. doi: 10.1038/ni.1703. Epub 2009/02/18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Jha S, Ting JP. Inflammasome-associated nucleotide-binding domain, leucine-rich repeat proteins and inflammatory diseases. J Immunol. 2009;183:7623–9. doi: 10.4049/jimmunol.0902425. Epub 2009/12/17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Pedra JH, Cassel SL, Sutterwala FS. Sensing pathogens and danger signals by the inflammasome. Curr Opin Immunol. 2009;21:10–6. doi: 10.1016/j.coi.2009.01.006. Epub 2009/02/19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Lamkanfi M, Dixit VM. Inflammasomes: guardians of cytosolic sanctity. Immunol Rev. 2009;227:95–105. doi: 10.1111/j.1600-065X.2008.00730.x. Epub 2009/01/06. [DOI] [PubMed] [Google Scholar]
  • 102.Latz E. The inflammasomes: mechanisms of activation and function. Curr Opin Immunol. 2010;22:28–33. doi: 10.1016/j.coi.2009.12.004. Epub 2010/01/12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Schroder K, Tschopp J. The inflammasomes. Cell. 2010;140:821–32. doi: 10.1016/j.cell.2010.01.040. Epub 2010/03/23. [DOI] [PubMed] [Google Scholar]
  • 104.Franchi L, Munoz-Planillo R, Reimer T, Eigenbrod T, Nunez G. Inflammasomes as microbial sensors. Eur J Immunol. 2010;40:611–5. doi: 10.1002/eji.200940180. Epub 2010/03/05. [DOI] [PubMed] [Google Scholar]
  • 105.Bauernfeind F, Ablasser A, Bartok E, Kim S, Schmid-Burgk J, Cavlar T, Hornung V. Inflammasomes: current understanding and open questions. Cell Mol Life Sci. 2011;68:765–83. doi: 10.1007/s00018-010-0567-4. Epub 2010/11/13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Kersse K, Bertrand MJ, Lamkanfi M, Vandenabeele P. NOD-like receptors and the innate immune system: coping with danger, damage and death. Cytokine Growth Factor Rev. 2011;22:257–76. doi: 10.1016/j.cytogfr.2011.09.003. Epub 2011/10/15. [DOI] [PubMed] [Google Scholar]
  • 107.Gross O, Thomas CJ, Guarda G, Tschopp J. The inflammasome: an integrated view. Immunol Rev. 2011;243:136–51. doi: 10.1111/j.1600-065X.2011.01046.x. Epub 2011/09/03. [DOI] [PubMed] [Google Scholar]
  • 108.Lamkanfi M. Emerging inflammasome effector mechanisms. Nat Rev Immunol. 2011;11:213–20. doi: 10.1038/nri2936. Epub 2011/02/26. [DOI] [PubMed] [Google Scholar]
  • 109.Lamkanfi M, Dixit VM. Modulation of inflammasome pathways by bacterial and viral pathogens. J Immunol. 2011;187:597–602. doi: 10.4049/jimmunol.1100229. Epub 2011/07/08. [DOI] [PubMed] [Google Scholar]
  • 110.Broz P, Monack DM. Molecular mechanisms of inflammasome activation during microbial infections. Immunol Rev. 2011;243:174–90. doi: 10.1111/j.1600-065X.2011.01041.x. Epub 2011/09/03. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Skeldon A, Saleh M. The inflammasomes: molecular effectors of host resistance against bacterial, viral, parasitic, and fungal infections. Front Microbiol. 2011;2:15. doi: 10.3389/fmicb.2011.00015. Epub 2011/07/01. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Horvath GL, Schrum JE, De Nardo CM, Latz E. Intracellular sensing of microbes and danger signals by the inflammasomes. Immunol Rev. 2011;243:119–35. doi: 10.1111/j.1600-065X.2011.01050.x. Epub 2011/09/03. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.van de Veerdonk FL, Netea MG, Dinarello CA, Joosten LA. Inflammasome activation and IL-1beta and IL-18 processing during infection. Trends Immunol. 2011;32:110–6. doi: 10.1016/j.it.2011.01.003. Epub 2011/02/22. [DOI] [PubMed] [Google Scholar]
  • 114.Franchi L, Munoz-Planillo R, Nunez G. Sensing and reacting to microbes through the inflammasomes. Nat Immunol. 2012;13:325–32. doi: 10.1038/ni.2231. Epub 2012/03/21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Dagenais M, Skeldon A, Saleh M. The inflammasome: in memory of Dr. Jurg Tschopp. Cell Death Differ. 2012;19:5–12. doi: 10.1038/cdd.2011.159. Epub 2011/11/15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Ciraci C, Janczy JR, Sutterwala FS, Cassel SL. Control of innate and adaptive immunity by the inflammasome. Microbes Infect. 2012;14:1263–70. doi: 10.1016/j.micinf.2012.07.007. Epub 2012/07/31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Rathinam VA, Vanaja SK, Fitzgerald KA. Regulation of inflammasome signaling. Nat Immunol. 2012;13:333–42. doi: 10.1038/ni.2237. Epub 2012/03/21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Franchi L, Nunez G. Immunology. Orchestrating inflammasomes. Science. 2012;337:1299–300. doi: 10.1126/science.1229010. Epub 2012/09/18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Henao-Mejia J, Elinav E, Strowig T, Flavell RA. Inflammasomes: far beyond inflammation. Nat Immunol. 2012;13:321–4. doi: 10.1038/ni.2257. Epub 2012/03/21. [DOI] [PubMed] [Google Scholar]
  • 120.Latz E, Xiao TS, Stutz A. Activation and regulation of the inflammasomes. Nat Rev Immunol. 2013;13:397–411. doi: 10.1038/nri3452. Epub 2013/05/25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Bauernfeind F, Hornung V. Of inflammasomes and pathogens–sensing of microbes by the inflammasome. EMBO Mol Med. 2013;5:814–26. doi: 10.1002/emmm.201201771. Epub 2013/05/15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Vladimer GI, Marty-Roix R, Ghosh S, Weng D, Lien E. Inflammasomes and host defenses against bacterial infections. Curr Opin Microbiol. 2013;16:23–31. doi: 10.1016/j.mib.2012.11.008. Epub 2013/01/16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Lamkanfi M, Dixit VM. Mechanisms and functions of inflammasomes. Cell. 2014;157:1013–22. doi: 10.1016/j.cell.2014.04.007. Epub 2014/05/27. [DOI] [PubMed] [Google Scholar]
  • 124.Ulland TK, Ferguson PJ, Sutterwala FS. Evasion of inflammasome activation by microbial pathogens. J Clin Invest. 2015;125:469–77. doi: 10.1172/JCI75254. Epub 2015/02/03. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Vanaja SK, Rathinam VA, Fitzgerald KA. Mechanisms of inflammasome activation: recent advances and novel insights. Trends Cell Biol. 2015;25:308–15. doi: 10.1016/j.tcb.2014.12.009. Epub 2015/02/03. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Guo H, Callaway JB, Ting JP. Inflammasomes: mechanism of action, role in disease, and therapeutics. Nat Med. 2015;21:677–87. doi: 10.1038/nm.3893. Epub 2015/06/30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Black RA, Kronheim SR, Merriam JE, March CJ, Hopp TP. A pre-aspartate-specific protease from human leukocytes that cleaves pro-interleukin-1 beta. J Biol Chem. 1989;264:5323–6. Epub 1989/04/05. [PubMed] [Google Scholar]
  • 128.Kostura MJ, Tocci MJ, Limjuco G, Chin J, Cameron P, Hillman AG, Chartrain NA, Schmidt JA. Identification of a monocyte specific pre-interleukin 1 beta convertase activity. Proc Natl Acad Sci USA. 1989;86:5227–31. doi: 10.1073/pnas.86.14.5227. Epub 1989/07/01. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Thornberry NA, Bull HG, Calaycay JR, Chapman KT, Howard AD, Kostura MJ, Miller DK, Molineaux SM, Weidner JR, Aunins J, et al. A novel heterodimeric cysteine protease is required for interleukin-1 beta processing in monocytes. Nature. 1992;356:768–74. doi: 10.1038/356768a0. Epub 1992/04/30. [DOI] [PubMed] [Google Scholar]
  • 130.Cerretti DP, Kozlosky CJ, Mosley B, Nelson N, Van Ness K, Greenstreet TA, March CJ, Kronheim SR, Druck T, Cannizzaro LA, et al. Molecular cloning of the interleukin-1 beta converting enzyme. Science. 1992;256:97–100. doi: 10.1126/science.1373520. Epub 1992/04/03. [DOI] [PubMed] [Google Scholar]
  • 131.Gu Y, Kuida K, Tsutsui H, Ku G, Hsiao K, Fleming MA, Hayashi N, Higashino K, Okamura H, Nakanishi K, Kurimoto M, Tanimoto T, Flavell RA, Sato V, Harding MW, Livingston DJ, Su MS. Activation of interferon-gamma inducing factor mediated by interleukin-1beta converting enzyme. Science. 1997;275:206–9. doi: 10.1126/science.275.5297.206. Epub 1997/01/10. [DOI] [PubMed] [Google Scholar]
  • 132.Ghayur T, Banerjee S, Hugunin M, Butler D, Herzog L, Carter A, Quintal L, Sekut L, Talanian R, Paskind M, Wong W, Kamen R, Tracey D, Allen H. Caspase-1 processes IFN-gamma-inducing factor and regulates LPS-induced IFN-gamma production. Nature. 1997;386:619–23. doi: 10.1038/386619a0. Epub 1997/04/10. [DOI] [PubMed] [Google Scholar]
  • 133.Dinarello CA. Interleukin-1 beta, interleukin-18, and the interleukin-1 beta converting enzyme. Ann N Y Acad Sci. 1998;856:1–11. doi: 10.1111/j.1749-6632.1998.tb08307.x. Epub 1999/01/26. [DOI] [PubMed] [Google Scholar]
  • 134.Fantuzzi G, Dinarello CA. Interleukin-18 and interleukin-1 beta: two cytokine substrates for ICE (caspase-1) J Clin Immunol. 1999;19:1–11. doi: 10.1023/a:1020506300324. Epub 1999/03/18. [DOI] [PubMed] [Google Scholar]
  • 135.Sansonetti PJ, Phalipon A, Arondel J, Thirumalai K, Banerjee S, Akira S, Takeda K, Zychlinsky A. Caspase-1 activation of IL-1beta and IL-18 are essential for Shigella flexneri-induced inflammation. Immunity. 2000;12:581–90. doi: 10.1016/s1074-7613(00)80209-5. Epub 2000/06/08. [DOI] [PubMed] [Google Scholar]
  • 136.Kahlenberg JM, Lundberg KC, Kertesy SB, Qu Y, Dubyak GR. Potentiation of caspase-1 activation by the P2X7 receptor is dependent on TLR signals and requires NF-kappaB-driven protein synthesis. J Immunol. 2005;175:7611–22. doi: 10.4049/jimmunol.175.11.7611. Epub 2005/11/23. [DOI] [PubMed] [Google Scholar]
  • 137.Netea MG, van de Veerdonk FL, van der Meer JW, Dinarello CA, Joosten LA. Inflammasome-independent regulation of IL-1-family cytokines. Annu Rev Immunol. 2015;33:49–77. doi: 10.1146/annurev-immunol-032414-112306. Epub 2014/12/11. [DOI] [PubMed] [Google Scholar]
  • 138.Cookson BT, Brennan MA. Pro-inflammatory programmed cell death. Trends Microbiol. 2001;9:113–4. doi: 10.1016/s0966-842x(00)01936-3. Epub 2001/04/17. [DOI] [PubMed] [Google Scholar]
  • 139.Miao EA, Rajan JV, Aderem A. Caspase-1-induced pyroptotic cell death. Immunol Rev. 2011;243:206–14. doi: 10.1111/j.1600-065X.2011.01044.x. Epub 2011/09/03. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Shalini S, Dorstyn L, Dawar S, Kumar S. Old, new and emerging functions of caspases. Cell Death Differ. 2015;22:526–39. doi: 10.1038/cdd.2014.216. Epub 2014/12/20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Pineles BL, Romero R, Montenegro D, Tarca AL, Than NG, Hassan S, Gotsch F, Draghici S, Espinoza J, Kim CJ. “The inflammasome” in human parturition. Reproductive Sciences. 2007;14:59A. [Google Scholar]
  • 142.Gotsch F, Romero R, Chaiworapongsa T, Erez O, Vaisbuch E, Espinoza J, Kusanovic JP, Mittal P, Mazaki-Tovi S, Kim CJ, Kim JS, Edwin S, Nhan-Chang CL, Hamill N, Friel L, Than NG, Mazor M, Yoon BH, Hassan SS. Evidence of the involvement of caspase-1 under physiologic and pathologic cellular stress during human pregnancy: a link between the inflammasome and parturition. J Matern Fetal Neonatal Med. 2008;21:605–16. doi: 10.1080/14767050802212109. Epub 2008/10/02. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Romero R, Xu Y, Plazyo O, Chaemsaithong P, Chaiworapongsa T, Unkel R, Than NG, Chiang PJ, Dong Z, Xu Z, Tarca AL, Abrahams VM, Hassan SS, Yeo L, Gomez-Lopez N. A Role for the Inflammasome in Spontaneous Labor at Term. Am J Reprod Immunol. 2016 doi: 10.1111/aji.12440. Epub 2016/03/10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Gomez-Lopez N, Romero R, Xu Y, Plazyo O, Unkel R, Than NG, Chaemsaithong P, Chaiworapongsa T, Dong Z, Tarca AL, Abrahams VM, Yeo L, Hassan SS. A Role for the Inflammasome in Spontaneous Labor at Term with Acute Histologic Chorioamnionitis. Reprod Sci. 2017;24:934–53. doi: 10.1177/1933719116675058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Gomez-Lopez N, Romero R, Xu Y, Garcia-Flores V, Leng Y, Panaitescu B, Miller D, Abrahams VM, Hassan SS. Inflammasome assembly in the chorioamniotic membranes during spontaneous labor at term. Am J Reprod Immunol. 2017:77. doi: 10.1111/aji.12648. Epub 2017/02/25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Gomez-Lopez N, Romero R, Xu Y, Plazyo O, Unkel R, Leng Y, Than NG, Chaiworapongsa T, Panaitescu B, Dong Z, Tarca AL, Abrahams VM, Yeo L, Hassan SS. A Role for the Inflammasome in Spontaneous Preterm Labor With Acute Histologic Chorioamnionitis. Reprod Sci. 2017;24:1382–401. doi: 10.1177/1933719116687656. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Plazyo O, Romero R, Unkel R, Balancio A, Mial TN, Xu Y, Dong Z, Hassan SS, Gomez-Lopez N. HMGB1 Induces an Inflammatory Response in the Chorioamniotic Membranes That Is Partially Mediated by the Inflammasome. Biol Reprod. 2016;95:130. doi: 10.1095/biolreprod.116.144139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Bauernfeind FG, Horvath G, Stutz A, Alnemri ES, MacDonald K, Speert D, Fernandes-Alnemri T, Wu J, Monks BG, Fitzgerald KA, Hornung V, Latz E. Cutting edge: NF-kappaB activating pattern recognition and cytokine receptors license NLRP3 inflammasome activation by regulating NLRP3 expression. J Immunol. 2009;183:787–91. doi: 10.4049/jimmunol.0901363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Sutterwala FS, Haasken S, Cassel SL. Mechanism of NLRP3 inflammasome activation. Ann N Y Acad Sci. 2014;1319:82–95. doi: 10.1111/nyas.12458. Epub 2014/05/21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Vajjhala PR, Mirams RE, Hill JM. Multiple binding sites on the pyrin domain of ASC protein allow self-association and interaction with NLRP3 protein. J Biol Chem. 2012;287:41732–43. doi: 10.1074/jbc.M112.381228. Epub 2012/10/16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Fernandes-Alnemri T, Wu J, Yu JW, Datta P, Miller B, Jankowski W, Rosenberg S, Zhang J, Alnemri ES. The pyroptosome: a supramolecular assembly of ASC dimers mediating inflammatory cell death via caspase-1 activation. Cell Death Differ. 2007;14:1590–604. doi: 10.1038/sj.cdd.4402194. Epub 2007/06/30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Balci-Peynircioglu B, Waite AL, Schaner P, Taskiran ZE, Richards N, Orhan D, Gucer S, Ozen S, Gumucio D, Yilmaz E. Expression of ASC in renal tissues of familial mediterranean fever patients with amyloidosis: postulating a role for ASC in AA type amyloid deposition. Exp Biol Med (Maywood) 2008;233:1324–33. doi: 10.3181/0803-RM-106. Epub 2008/09/16. [DOI] [PubMed] [Google Scholar]
  • 153.Baroja-Mazo A, Martin-Sanchez F, Gomez AI, Martinez CM, Amores-Iniesta J, Compan V, Barbera-Cremades M, Yague J, Ruiz-Ortiz E, Anton J, Bujan S, Couillin I, Brough D, Arostegui JI, Pelegrin P. The NLRP3 inflammasome is released as a particulate danger signal that amplifies the inflammatory response. Nat Immunol. 2014;15:738–48. doi: 10.1038/ni.2919. Epub 2014/06/24. [DOI] [PubMed] [Google Scholar]
  • 154.Franklin BS, Bossaller L, De Nardo D, Ratter JM, Stutz A, Engels G, Brenker C, Nordhoff M, Mirandola SR, Al-Amoudi A, Mangan MS, Zimmer S, Monks BG, Fricke M, Schmidt RE, Espevik T, Jones B, Jarnicki AG, Hansbro PM, Busto P, Marshak-Rothstein A, Hornemann S, Aguzzi A, Kastenmuller W, Latz E. The adaptor ASC has extracellular and ‘prionoid’ activities that propagate inflammation. Nat Immunol. 2014;15:727–37. doi: 10.1038/ni.2913. Epub 2014/06/24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Romero R, Mazor M, Wu YK, Sirtori M, Oyarzun E, Mitchell MD, Hobbins JC. Infection in the pathogenesis of preterm labor. Semin Perinatol. 1988;12:262–79. Epub 1988/10/01. [PubMed] [Google Scholar]
  • 156.Romero R, Mazor M. Infection and preterm labor. Clin Obstet Gynecol. 1988;31:553–84. doi: 10.1097/00003081-198809000-00006. Epub 1988/09/01. [DOI] [PubMed] [Google Scholar]
  • 157.Romero R, Shamma F, Avila C, Jimenez C, Callahan R, Nores J, Mazor M, Brekus CA, Hobbins JC. Infection and labor. VI. Prevalence, microbiology, and clinical significance of intraamniotic infection in twin gestations with preterm labor. Am J Obstet Gynecol. 1990;163:757–61. doi: 10.1016/0002-9378(90)91063-i. Epub 1990/09/01. [DOI] [PubMed] [Google Scholar]
  • 158.Romero R, Ghidini A, Mazor M, Behnke E. Microbial invasion of the amniotic cavity in premature rupture of membranes. Clin Obstet Gynecol. 1991;34:769–78. doi: 10.1097/00003081-199112000-00013. Epub 1991/12/01. [DOI] [PubMed] [Google Scholar]
  • 159.Yoon BH, Romero R, Moon JB, Shim SS, Kim M, Kim G, Jun JK. Clinical significance of intra-amniotic inflammation in patients with preterm labor and intact membranes. Am J Obstet Gynecol. 2001;185:1130–6. doi: 10.1067/mob.2001.117680. Epub 2001/11/22. [DOI] [PubMed] [Google Scholar]
  • 160.Romero R, Miranda J, Chaiworapongsa T, Chaemsaithong P, Gotsch F, Dong Z, Ahmed AI, Yoon BH, Hassan SS, Kim CJ, Korzeniewski SJ, Yeo L. 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;71:330–58. doi: 10.1111/aji.12189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Romero R, Miranda J, Chaiworapongsa T, Korzeniewski SJ, Chaemsaithong P, Gotsch F, Dong Z, Ahmed AI, Yoon BH, Hassan SS, Kim CJ, Yeo L. Prevalence and clinical significance of sterile intra-amniotic inflammation in patients with preterm labor and intact membranes. Am J Reprod Immunol. 2014;72:458–74. doi: 10.1111/aji.12296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Romero R, Miranda J, Kusanovic JP, Chaiworapongsa T, Chaemsaithong P, Martinez A, Gotsch F, Dong Z, Ahmed AI, Shaman M, Lannaman K, Yoon BH, Hassan SS, Kim CJ, Korzeniewski SJ, Yeo L, Kim YM. Clinical chorioamnionitis at term I: microbiology of the amniotic cavity using cultivation and molecular techniques. J Perinat Med. 2015;43:19–36. doi: 10.1515/jpm-2014-0249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Romero R, Miranda J, Chaemsaithong P, Chaiworapongsa T, Kusanovic JP, Dong Z, Ahmed AI, Shaman M, Lannaman K, Yoon BH, Hassan SS, Kim CJ, Korzeniewski SJ, Yeo L, Kim YM. Sterile and microbial-associated intra-amniotic inflammation in preterm prelabor rupture of membranes. J Matern Fetal Neonatal Med. 2015;28:1394–409. doi: 10.3109/14767058.2014.958463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Kacerovsky M, Musilova I, Stepan M, Andrys C, Drahosova M, Jacobsson B. Detection of intraamniotic inflammation in fresh and processed amniotic fluid samples with the interleukin-6 point of care test. Am J Obstet Gynecol. 2015;213:435–6. doi: 10.1016/j.ajog.2015.05.039. [DOI] [PubMed] [Google Scholar]
  • 165.Chaemsaithong P, Romero R, Korzeniewski SJ, Dong Z, Yeo L, Hassan SS, Kim YM, Yoon BH, Chaiworapongsa T. 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;28:1510–9. doi: 10.3109/14767058.2014.961417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Combs CA, Garite TJ, Lapidus JA, Lapointe JP, Gravett M, Rael J, Amon E, Baxter JK, Brady K, Clewell W, Eddleman KA, Fortunato S, Franco A, Haas DM, Heyborne K, Hickok DE, How HY, Luthy D, Miller H, Nageotte M, Pereira L, Porreco R, Robilio PA, Simhan H, Sullivan SA, Trofatter K, Westover T, Obstetrix Collaborative Research N Detection of microbial invasion of the amniotic cavity by analysis of cervicovaginal proteins in women with preterm labor and intact membranes. Am J Obstet Gynecol. 2015;212:482 e1–e12. doi: 10.1016/j.ajog.2015.02.007. [DOI] [PubMed] [Google Scholar]
  • 167.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;29:2563–72. doi: 10.3109/14767058.2015.1094794. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Kunze M, Klar M, Morfeld CA, Thorns B, Schild RL, Markfeld-Erol F, Rasenack R, Proempeler H, Hentschel R, Schaefer WR. Cytokines in noninvasively obtained amniotic fluid as predictors of fetal inflammatory response syndrome. Am J Obstet Gynecol. 2016;215:96 e1–8. doi: 10.1016/j.ajog.2016.01.181. [DOI] [PubMed] [Google Scholar]
  • 169.Romero R, Chaemsaithong P, Korzeniewski SJ, Tarca AL, Bhatti G, Xu Z, Kusanovic JP, Dong Z, Docheva N, Martinez-Varea A, Yoon BH, Hassan SS, Chaiworapongsa T, Yeo L. Clinical chorioamnionitis at term II: the intra-amniotic inflammatory response. J Perinat Med. 2016;44:5–22. doi: 10.1515/jpm-2015-0045. Epub 2015/05/06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Chaemsaithong P, Romero R, Korzeniewski SJ, Martinez-Varea A, Dong Z, Yoon BH, Hassan SS, Chaiworapongsa T, Yeo L. 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;29:349–59. doi: 10.3109/14767058.2015.1006620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Romero R, Chaemsaithong P, Korzeniewski SJ, Kusanovic JP, Docheva N, Martinez-Varea A, Ahmed AI, Yoon BH, Hassan SS, Chaiworapongsa T, Yeo L. Clinical chorioamnionitis at term III: how well do clinical criteria perform in the identification of proven intra-amniotic infection? J Perinat Med. 2016;44:23–32. doi: 10.1515/jpm-2015-0044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Revello R, Alcaide MJ, Dudzik D, Abehsera D, Bartha JL. Differential amniotic fluid cytokine profile in women with chorioamnionitis with and without funisitis. J Matern Fetal Neonatal Med. 2016;29:2161–5. doi: 10.3109/14767058.2015.1077512. [DOI] [PubMed] [Google Scholar]
  • 173.Romero R, Chaemsaithong P, Docheva N, Korzeniewski SJ, Tarca AL, Bhatti G, Xu Z, Kusanovic JP, Dong Z, Chaiyasit N, Ahmed AI, Yoon BH, Hassan SS, Chaiworapongsa T, Yeo L. Clinical chorioamnionitis at term IV: the maternal plasma cytokine profile. J Perinat Med. 2016;44:77–98. doi: 10.1515/jpm-2015-0103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Chaemsaithong P, Romero R, Korzeniewski SJ, Martinez-Varea A, Dong Z, Yoon BH, Hassan SS, Chaiworapongsa T, Yeo L. 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;29:360–7. doi: 10.3109/14767058.2015.1006621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Martinez-Varea A, Romero R, Xu Y, Miller D, Ahmed AI, Chaemsaithong P, Chaiyasit N, Yeo L, Shaman M, Lannaman K, Cher B, Hassan SS, Gomez-Lopez N. Clinical chorioamnionitis at term VII: the amniotic fluid cellular immune response. J Perinat Med. 2017;45:523–38. doi: 10.1515/jpm-2016-0225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Oh KJ, Kim SM, Hong JS, Maymon E, Erez O, Panaitescu B, Gomez-Lopez N, Romero R, Yoon BH. Twenty-four percent of patients with clinical chorioamnionitis in preterm gestations have no evidence of either culture-proven intraamniotic infection or intraamniotic inflammation. Am J Obstet Gynecol. 2017;216:604 e1–e11. doi: 10.1016/j.ajog.2017.02.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Kacerovsky M, Radochova V, Musilova I, Stepan M, Slezak R, Andrys C, Skogstrand K, Hougaard D, Jacobsson B. Levels of multiple proteins in gingival crevicular fluid and intra-amniotic complications in women with preterm prelabor rupture of membranes. J Matern Fetal Neonatal Med. 2017:1–9. doi: 10.1080/14767058.2017.1347626. [DOI] [PubMed] [Google Scholar]
  • 178.Chaiyasit N, Romero R, Chaemsaithong P, Docheva N, Bhatti G, Kusanovic JP, Dong Z, Yeo L, Pacora P, Hassan SS, Erez O. Clinical chorioamnionitis at term VIII: a rapid MMP-8 test for the identification of intra-amniotic inflammation. J Perinat Med. 2017;45:539–50. doi: 10.1515/jpm-2016-0344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Gomez-Lopez N, Romero R, Garcia-Flores V, Xu Y, Leng Y, Alhousseini A, Hassan SS, Panaitescu B. Amniotic fluid neutrophils can phagocytize bacteria: A mechanism for microbial killing in the amniotic cavity. Am J Reprod Immunol. 2017;78 doi: 10.1111/aji.12723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Musilova I, Andrys C, Drahosova M, Soucek O, Stepan M, Bestvina T, Spacek R, Jacobsson B, Cobo T, Kacerovsky M. Intraamniotic inflammation and umbilical cord blood interleukin-6 concentrations in pregnancies complicated by preterm prelabor rupture of membranes. J Matern Fetal Neonatal Med. 2017;30:900–10. doi: 10.1080/14767058.2016.1197900. [DOI] [PubMed] [Google Scholar]
  • 181.Gomez-Lopez N, Romero R, Xu Y, Leng Y, Garcia-Flores V, Miller D, Jacques SM, Hassan SS, Faro J, Alsamsam A, Alhousseini A, Gomez-Roberts H, Panaitescu B, Yeo L, Maymon E. Are amniotic fluid neutrophils in women with intraamniotic infection and/or inflammation of fetal or maternal origin? Am J Obstet Gynecol. 2017 doi: 10.1016/j.ajog.2017.09.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Oh KJ, Hong JS, Romero R, Yoon BH. The frequency and clinical significance of intra-amniotic inflammation in twin pregnancies with preterm labor and intact membranes. J Matern Fetal Neonatal Med. 2017:1–15. doi: 10.1080/14767058.2017.1384460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.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. doi: 10.1016/j.ajog.2015.08.040. Epub 2015/10/03. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Redline RW. Classification of placental lesions. Am J Obstet Gynecol. 2015;213:S21–8. doi: 10.1016/j.ajog.2015.05.056. Epub 2015/10/03. [DOI] [PubMed] [Google Scholar]
  • 185.Revello R, Alcaide MJ, Abehsera D, Martin-Camean M, Sousa EFGM, Alonso-Luque B, Bartha JL. Prediction of chorioamnionitis in cases of intraamniotic infection by ureaplasma urealyticum in women with very preterm premature rupture of membranes or preterm labour. J Matern Fetal Neonatal Med. 2017:1–6. doi: 10.1080/14767058.2017.1330407. [DOI] [PubMed] [Google Scholar]
  • 186.Romero R, Chaemsaithong P, Docheva N, Korzeniewski SJ, Kusanovic JP, Yoon BH, Kim JS, Chaiyasit N, Ahmed AI, Qureshi F, Jacques SM, Kim CJ, Hassan SS, Chaiworapongsa T, Yeo L, Kim YM. Clinical chorioamnionitis at term VI: acute chorioamnionitis and funisitis according to the presence or absence of microorganisms and inflammation in the amniotic cavity. J Perinat Med. 2016;44:33–51. doi: 10.1515/jpm-2015-0119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Yoon BH, Romero R, Park JS, Kim M, Oh SY, Kim CJ, Jun JK. 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;183:1124–9. doi: 10.1067/mob.2000.109035. Epub 2000/11/21. [DOI] [PubMed] [Google Scholar]
  • 188.Pacora P, Chaiworapongsa T, Maymon E, Kim YM, Gomez R, Yoon BH, Ghezzi F, Berry SM, Qureshi F, Jacques SM, Kim JC, Kadar N, Romero R. Funisitis and chorionic vasculitis: the histological counterpart of the fetal inflammatory response syndrome. J Matern Fetal Neonatal Med. 2002;11:18–25. doi: 10.1080/jmf.11.1.18.25. Epub 2002/10/17. [DOI] [PubMed] [Google Scholar]
  • 189.Romero R, Chaemsaithong P, Docheva N, Korzeniewski SJ, Tarca AL, Bhatti G, Xu Z, Kusanovic JP, Chaiyasit N, Dong Z, Yoon BH, Hassan SS, Chaiworapongsa T, Yeo L, Kim YM. Clinical chorioamnionitis at term V: umbilical cord plasma cytokine profile in the context of a systemic maternal inflammatory response. J Perinat Med. 2016;44:53–76. doi: 10.1515/jpm-2015-0121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Apgar V. A proposal for a new method of evaluation of the newborn infant. Curr Res Anesth Analg. 1953;32:260–7. Epub 1953/07/01. [PubMed] [Google Scholar]
  • 191.Thavarajah H, Flatley C, Kumar S. The relationship between the five minute Apgar score, mode of birth and neonatal outcomes. J Matern Fetal Neonatal Med. 2017:1–7. doi: 10.1080/14767058.2017.1315666. [DOI] [PubMed] [Google Scholar]
  • 192.van Tetering AAC, van de Ven J, Fransen AF, Dieleman JP, van Runnard Heimel PJ, Oei SG. Risk factors of incomplete Apgar score and umbilical cord blood gas analysis: a retrospective observational study. J Matern Fetal Neonatal Med. 2017;30:2539–44. doi: 10.1080/14767058.2016.1256985. [DOI] [PubMed] [Google Scholar]
  • 193.Cai X, Chen J, Xu H, Liu S, Jiang QX, Halfmann R, Chen ZJ. Prion-like polymerization underlies signal transduction in antiviral immune defense and inflammasome activation. Cell. 2014;156:1207–22. doi: 10.1016/j.cell.2014.01.063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Lu A, Magupalli VG, Ruan J, Yin Q, Atianand MK, Vos MR, Schroder GF, Fitzgerald KA, Wu H, Egelman EH. Unified polymerization mechanism for the assembly of ASC-dependent inflammasomes. Cell. 2014;156:1193–206. doi: 10.1016/j.cell.2014.02.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Ruland J. Inflammasome: putting the pieces together. Cell. 2014;156:1127–9. doi: 10.1016/j.cell.2014.02.038. [DOI] [PubMed] [Google Scholar]
  • 196.Sahillioglu AC, Sumbul F, Ozoren N, Haliloglu T. Structural and dynamics aspects of ASC speck assembly. Structure. 2014;22:1722–34. doi: 10.1016/j.str.2014.09.011. [DOI] [PubMed] [Google Scholar]
  • 197.Dick MS, Sborgi L, Ruhl S, Hiller S, Broz P. ASC filament formation serves as a signal amplification mechanism for inflammasomes. Nat Commun. 2016;7:11929. doi: 10.1038/ncomms11929. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Srinivasula SM, Poyet JL, Razmara M, Datta P, Zhang Z, Alnemri ES. The PYRIN-CARD protein ASC is an activating adaptor for caspase-1. J Biol Chem. 2002;277:21119–22. doi: 10.1074/jbc.C200179200. Epub 2002/04/23. [DOI] [PubMed] [Google Scholar]
  • 199.Pacora P, Romero R, Maymon E, Gervasi MT, Gomez R, Edwin SS, Yoon BH. Participation of the novel cytokine interleukin 18 in the host response to intra-amniotic infection. Am J Obstet Gynecol. 2000;183:1138–43. doi: 10.1067/mob.2000.108881. Epub 2000/11/21. [DOI] [PubMed] [Google Scholar]
  • 200.He Y, Hara H, Nunez G. Mechanism and Regulation of NLRP3 Inflammasome Activation. Trends Biochem Sci. 2016;41:1012–21. doi: 10.1016/j.tibs.2016.09.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Romero R, Durum S, Dinarello CA, Oyarzun E, Hobbins JC, Mitchell MD. Interleukin-1 stimulates prostaglandin biosynthesis by human amnion. Prostaglandins. 1989;37:13–22. doi: 10.1016/0090-6980(89)90028-2. Epub 1989/01/01. [DOI] [PubMed] [Google Scholar]
  • 202.Mitchell MD, Edwin S, Romero RJ. Prostaglandin biosynthesis by human decidual cells: effects of inflammatory mediators. Prostaglandins Leukot Essent Fatty Acids. 1990;41:35–8. doi: 10.1016/0952-3278(90)90128-8. [DOI] [PubMed] [Google Scholar]
  • 203.Brown NL, Alvi SA, Elder MG, Bennett PR, Sullivan MH. Interleukin-1beta and bacterial endotoxin change the metabolism of prostaglandins E2 and F2alpha in intact term fetal membranes. Placenta. 1998;19:625–30. doi: 10.1016/s0143-4004(98)90024-8. [DOI] [PubMed] [Google Scholar]
  • 204.Hertelendy F, Romero R, Molnar M, Todd H, Baldassare JJ. Cytokine-initiated signal transduction in human myometrial cells. Am J Reprod Immunol. 1993;30:49–57. doi: 10.1111/j.1600-0897.1993.tb00601.x. Epub 1993/09/01. [DOI] [PubMed] [Google Scholar]
  • 205.Hertelendy F, Rastogi P, Molnar M, Romero R. Interleukin-1beta-induced prostaglandin E2 production in human myometrial cells: role of a pertussis toxin-sensitive component. Am J Reprod Immunol. 2001;45:142–7. doi: 10.1111/j.8755-8920.2001.450304.x. Epub 2001/03/29. [DOI] [PubMed] [Google Scholar]
  • 206.Belt AR, Baldassare JJ, Molnar M, Romero R, Hertelendy F. The nuclear transcription factor NF-kappaB mediates interleukin-1beta-induced expression of cyclooxygenase-2 in human myometrial cells. Am J Obstet Gynecol. 1999;181:359–66. doi: 10.1016/s0002-9378(99)70562-4. Epub 1999/08/24. [DOI] [PubMed] [Google Scholar]
  • 207.Watari M, Watari H, DiSanto ME, Chacko S, Shi GP, Strauss JF., 3rd Pro-inflammatory cytokines induce expression of matrix-metabolizing enzymes in human cervical smooth muscle cells. Am J Pathol. 1999;154:1755–62. doi: 10.1016/S0002-9440(10)65431-4. Epub 1999/06/11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Arechavaleta-Velasco F, Ogando D, Parry S, Vadillo-Ortega F. Production of matrix metalloproteinase-9 in lipopolysaccharide-stimulated human amnion occurs through an autocrine and paracrine proinflammatory cytokine-dependent system. Biol Reprod. 2002;67:1952–8. doi: 10.1095/biolreprod.102.004721. [DOI] [PubMed] [Google Scholar]
  • 209.Ibrahim SA, Ackerman WEt, Summerfield TL, Lockwood CJ, Schatz F, Kniss DA. Inflammatory gene networks in term human decidual cells define a potential signature for cytokine-mediated parturition. Am J Obstet Gynecol. 2016;214:284 e1–e47. doi: 10.1016/j.ajog.2015.08.075. [DOI] [PubMed] [Google Scholar]
  • 210.Romero R, Mazor M, Tartakovsky B. Systemic administration of interleukin-1 induces preterm parturition in mice. Am J Obstet Gynecol. 1991;165:969–71. doi: 10.1016/0002-9378(91)90450-6. Epub 1991/10/01. [DOI] [PubMed] [Google Scholar]
  • 211.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;171:1660–7. doi: 10.1016/0002-9378(94)90418-9. Epub 1994/12/01. [DOI] [PubMed] [Google Scholar]
  • 212.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;171:1668–72. doi: 10.1016/0002-9378(94)90419-7. Epub 1994/12/01. [DOI] [PubMed] [Google Scholar]
  • 213.Baggia S, Gravett MG, Witkin SS, Haluska GJ, Novy MJ. Interleukin-1 beta intra-amniotic infusion induces tumor necrosis factor-alpha, prostaglandin production, and preterm contractions in pregnant rhesus monkeys. J Soc Gynecol Investig. 1996;3:121–6. doi: 10.1177/107155769600300304. Epub 1996/05/01. [DOI] [PubMed] [Google Scholar]
  • 214.Vadillo-Ortega F, Sadowsky DW, Haluska GJ, Hernandez-Guerrero C, Guevara-Silva R, Gravett MG, Novy MJ. Identification of matrix metalloproteinase-9 in amniotic fluid and amniochorion in spontaneous labor and after experimental intrauterine infection or interleukin-1 beta infusion in pregnant rhesus monkeys. Am J Obstet Gynecol. 2002;186:128–38. doi: 10.1067/mob.2002.118916. Epub 2002/01/26. [DOI] [PubMed] [Google Scholar]
  • 215.Sadowsky DW, Adams KM, Gravett MG, Witkin SS, Novy MJ. Preterm labor is induced by intraamniotic infusions of interleukin-1beta and tumor necrosis factor-alpha but not by interleukin-6 or interleukin-8 in a nonhuman primate model. Am J Obstet Gynecol. 2006;195:1578–89. doi: 10.1016/j.ajog.2006.06.072. Epub 2006/11/30. [DOI] [PubMed] [Google Scholar]
  • 216.Aagaard K, Ganu R, Ma J, Hu M, Miller L, Jobe AH, Kallapur SG, Chougnet CA. Intraamniotic interleukin-1 (IL1β) induces histological choriamnionitis and alters the microbiome in a primate model of inflammatory preterm birth. Am J Obstet Gynecol. 2014;208:S218. [Google Scholar]
  • 217.Prince A, Ma J, Miller L, Hu M, Jobe AH, Chougnet CA, Kallapur SG, Aagaard K. Chorioamnionitis induced by intraamniotic injection of IL1, LPS or Ureaplasma parvum is associated with an altered microbiome in a primate model of inflammatory preterm birth. Am J Obstet Gynecol. 2014;212:S153. [Google Scholar]
  • 218.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. doi: 10.1095/biolreprod.114.124420. Epub 2014/12/30. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES