Skip to main content
Reproductive Sciences logoLink to Reproductive Sciences
. 2017 Jan 26;24(10):1382–1401. doi: 10.1177/1933719116687656

A Role for the Inflammasome in Spontaneous Preterm Labor With Acute Histologic Chorioamnionitis

Nardhy Gomez-Lopez 1,2,3,, Roberto Romero 1,4,5,6,, Yi Xu 1,2, Olesya Plazyo 1,2, Ronald Unkel 1,2, Yaozhu Leng 1,2, Nandor Gabor Than 1,7,8,9, Tinnakorn Chaiworapongsa 1,2, Bogdan Panaitescu 1,2, Zhong Dong 1,2, Adi L Tarca 1,2, Vikki M Abrahams 10, Lami Yeo 1,2, Sonia S Hassan 1,2
PMCID: PMC5933090  PMID: 28122480

Abstract

Inflammasomes are cytosolic multiprotein complexes that orchestrate inflammation in response to pathogens and endogenous danger signals. Herein, we determined whether the chorioamniotic membranes from women in spontaneous preterm labor with acute histologic chorioamnionitis (1) express major inflammasome components; (2) express caspase (CASP)-1 and CASP-4 as well as their active forms; (3) exhibit apoptosis-associated speck-like protein containing a CARD (ASC)/CASP-1 complex formation; and (4) release the mature forms of interleukin (IL)-1β and IL-18. We utilized quantitative reverse transcription polymerase chain reaction, enzyme-linked immunosorbent assay, immunoblotting, and immunohistochemistry to determine the messenger RNA (mRNA) and protein expression of major inflammasome components, nucleotide-binding oligomerization domain (NOD) proteins, and the pro- and mature/active forms of CASP-1, CASP-4, IL-1β, and IL-18. The ASC/CASP-1 complex formation was determined using an in situ proximity ligation assay. When comparing the chorioamniotic membranes from women in spontaneous preterm labor with acute histologic chorioamnionitis to those without this placental lesion, we found that (1) the mRNA of NLR family pyrin domain-containing protein (NLRP)1, NLRP3, NLR family CARD domain-containing protein 4 (NLRC4), and NOD2 were higher; (2) the NLRP3 protein was increased; (3) the mRNA and active form (p10) of CASP-1 were greater; (4) the mRNA and active form of CASP-4 were increased; (5) the mRNA and mature form of IL-1β were higher; (6) the mature form of IL-18 was elevated; and (7) ASC/CASP-1 complex formation was increased. In conclusion, spontaneous preterm labor with acute histologic chorioamnionitis is characterized by an upregulation of NLRP3 and the active form of CASP-4, as well as increased ASC/CASP-1 complex formation, which may participate in the activation of CASP-1 and the maturation of IL-1β and IL-18 in the chorioamniotic membranes. These findings provide the first evidence that supports a role for the inflammasome in the pathological inflammation implicated in spontaneous preterm labor with acute histologic chorioamnionitis.

Keywords: apoptosis-associated speck-like protein containing a CARD (ASC), caspase-1, caspase-4, cytokines, fetal inflammatory response, fever, funisitis, inflammasome assembly, intraamniotic inflammation, microbial invasion of the amniotic cavity, neutrophil, parturition, pregnancy, prematurity, preterm birth, PYD and CARD domain containing (PYCARD) protein, pyroptosis

Introduction

Preterm birth is one of the most common, yet detrimental, obstetrical syndromes16 and the leading cause of perinatal morbidity and mortality worldwide.710 Approximately 70% of all preterm births are preceded by spontaneous preterm labor,1,5,1113 a syndrome of multiple pathological processes.14 Of all the putative causes associated with spontaneous preterm labor, only intraamniotic inflammation/infection has been causally linked to preterm birth.1538 Intraamniotic inflammation/infection generally results in acute histologic chorioamnionitis;3948 thus, this placental lesion is strongly associated with preterm labor and birth.40,4953 Acute histologic chorioamnionitis can also occur in the setting of sterile intraamniotic inflammation,5458 an inflammatory process in which microorganisms cannot be detected using both cultivation and molecular microbiology techniques.5463 Sterile inflammation is induced by danger signals, termed damage-associated molecular patterns (DAMPs)64,65 or alarmins,66 derived from necrotic cells or cellular stress.67 Therefore, acute histologic chorioamnionitis is evidence of intraamniotic inflammation regardless of the presence of infection.68

Acute histologic chorioamnionitis is defined by the infiltration of neutrophils and monocytes/macrophages into the chorioamniotic membranes,45,48,6872 which is mediated by a gradient of potent chemokines, including interleukin (IL)-8,68,73,74 C-X-C motif chemokine ligand 6 (CXCL6),68,75 and C-C motif chemokine ligand 2 (CCL2)76 (also known as monocyte chemoattractant protein or MCP-1). This pathological activation of the innate immune system is observed in spontaneous preterm labor and includes high concentrations of pro-inflammatory cytokines, such as IL-1α, IL-1β, tumor necrosis factor α, and IL-6 in the amniotic fluid,7789 decidua,90,91 and umbilical cord blood.9294 Elevated concentrations of these cytokines are linked to adverse neonatal outcomes.80,82,83,92,95111 Therefore, the study herein focuses on the mechanisms implicated in the pathophysiology of acute histologic chorioamnionitis in spontaneous preterm labor.

The inflammasome is implicated in physiological112114 and pathological inflammation (ie, acute histologic chorioamnionitis115) during human parturition at term; however, its role in spontaneous preterm labor is unknown. Inflammasomes are cytoplasmic multiprotein complexes that promote an inflammatory response through the release of the mature forms of IL-1β and IL-18.116150 The inflammasome complex contains: (1) a pattern recognition receptor (PRR or sensor molecule), (2) the adaptor protein ASC (apoptosis-associated speck-like protein containing a CARD), and (3) pro-caspase-1 (pro-CASP-1).116152 Inflammasome activation prompts the release of active CASP-1 which, in turn, can participate in the processing of mature IL-1β and IL-18.153163 These events induce a pro-inflammatory programmed cell death termed pyroptosis.164166 In addition, active CASP-4 participates in the noncanonical activation of the inflammasome by activating CASP-1149,167171, which can lead to pyroptosis and the release of alarmins (eg, high mobility group box-1 or HMGB1) in a CASP-1-independent manner.172

Herein, we propose that the inflammasome is implicated in the pathological inflammation (acute histologic chorioamnionitis) associated with spontaneous preterm labor. Such a hypothesis is supported by the fact that amniotic fluid concentrations of CASP-1,113 IL-1β,78 and IL-18173 are greater in women who undergo spontaneous preterm labor with intraamniotic infection/inflammation than in those without this clinical condition. The aims of this study were to determine whether the chorioamniotic membranes from women in spontaneous preterm labor with acute histologic chorioamnionitis: (1) express major inflammasome components; (2) express CASP-1 and CASP-4 as well as their active forms; (3) exhibit ASC/CASP-1 complex formation; and (4) release the mature forms of IL-1β and IL-18.

Materials and Methods

Human Participants, Clinical Specimens, and Definitions

In order to conduct this case–control study, chorioamniotic membrane samples from women who underwent spontaneous preterm labor with (PTL-ACA) or without (PTL) acute histologic chorioamnionitis were collected from the Bank of Biological Specimens of the Detroit Medical Center, Wayne State University, and the Perinatology Research Branch (Detroit, Michigan), 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. The Institutional Review Boards of these institutions approved the collection and use of biological materials for research purposes. Participants provided written informed consent and samples were collected within 0.5 hour after delivery. Table 1 includes the demographic and clinical characteristics of the study population. Multiparous women or women with neonates having congenital or chromosomal abnormalities were excluded. 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. Preterm delivery was defined as birth prior to week 37 of gestation.

Table 1.

Demographic and Clinical Characteristics of the Study Groups.

PTL (n = 33) PTL-ACA (n = 37) P Value
Maternal age, years, median (IQR)a 24.5 (19.8-32.3) 26 (21-31.3) NS
Race, n (%)b NS
 African American 24 (72.7) 30 (81.1)
 Caucasian 4 (12.1) 5 (13.5)
 Other 5 (15.2) 2 (5.4)
Maternal weight, kg, median (IQR)a 65.5 (54-75.3) 69.5 (54-85) NS
Body mass index, kg/m2, median (IQR)a 24.6 (20.6-29) 24.4 (20.2-32.9) NS
Primiparity, n (%)b 5 (15.2) 5 (13.5) NS
Gestational age at delivery, weeks, median (IQR)a 31.7 (30-33) 31.8 (30.3-32.9) NS
Birth weight, g, median (IQR)a 1571.5 (1193.8-1928.8) 1560 (1218.8-1906.3) NS
Cesarean section, n (%)b 8 (24.2) 7 (18.9) NS
Acute histologic chorioamnionitis, n (%)b 0 (0) 37 (100) <.0001

Abbreviations: PTL, preterm labor without acute histologic chorioamnionitis; PTL-ACA, preterm labor with acute histologic chorioamnionitis; IQR, interquartile range; NS, non-significant.

aMann-Whitney U test.

bFisher’s exact test.

Placental Histopathological Examinations

Five-µm-thick sections of formalin-fixed, paraffin-embedded tissue specimens were cut and mounted on SuperFrost Plus microscope slides (Erie Scientific LLC, Portsmouth, New Hampshire). In each case, several tissue sections of the chorioamniotic membranes, umbilical cord, and placental disk were examined. After deparaffinization, slides were rehydrated, stained with hematoxylin–eosin, and evaluated by pathologists who were blinded to the clinical outcome, according to published criteria.68,69 The diagnosis of acute histologic chorioamnionitis was made when the infiltration of neutrophils into the chorionic trophoblast layer or chorioamniotic connective tissue was observed.68,69

RNA Isolation, cDNA Generation, and Quantitative Reverse Transcription Polymerase Chain Reaction Analysis

Total RNA was extracted from snap-frozen chorioamniotic membrane samples (PTL, n = 33 and PTL-ACA, n = 37) using TRIzol (Invitrogen, Life Technologies Corporation, Grand Island, New York) and Qiagen RNeasy kits (Qiagen, Gaithersburg, Maryland). The purity, concentration, and integrity of the RNA were assessed using the NanoDrop 1000 spectrophotometer (Thermo Scientific, Wilmington, Delaware, USA) and the Bioanalyzer 2100 (Agilent Technologies, Wilmington, Delaware). cDNA was generated using the SuperScript III First-Strand Synthesis System (Invitrogen) and oligo(dT)20 primers (Invitrogen). Gene expression profiling was performed on the BioMark System for high-throughput quantitative reverse transcription polymerase chain reaction (Fluidigm, San Francisco, California) and the ABI 7500 FAST Real-Time PCR System (Applied Biosystems, Life Technologies Corporation, Foster City, California) with TaqMan gene expression assays (Applied Biosystems) listed in Supplementary Table 1.

Chorioamniotic Membrane Tissue Lysates

Tissue lysates were prepared by homogenizing snap-frozen chorioamniotic membranes (PTL and PTL-ACA, n = 10 each) in 2 mL of 1X phosphate-buffered saline (PBS; Invitrogen) containing a complete protease inhibitor cocktail (Cat. No. 11697498001; Roche Applied Science, Mannheim, Germany). Next, lysates were centrifuged at 15700×g for 5 minutes at 4°C, and the supernatant was collected and stored at −80°C. Prior to enzyme-linked immunosorbent assay (ELISA) or immunoblotting, total protein determination was determined using the Quick Start Bradford Protein Assay Kit (Bio-Rad, Hercules, California).

Enzyme-Linked Immunosorbent Assays

Protein concentrations of NLR family pyrin domain-containing protein (NLRP)1, NLRP3, absent in melanoma 2 (AIM2), nucleotide-binding oligomerization domain (NOD) 2 protein, CASP-1, CASP-4, IL-18, pro-IL-1β, and mature IL-1β were determined in chorioamniotic membrane tissue lysates by immunoassays (NLRP1, NLRP3, and NOD2 ELISA kits from Cusabio, Wuhan, Hubei, P.R. China; AIM2, CASP-1, and CASP-4 ELISA kits from Cloud Clone, Houston, Texas; pro-IL-1β and IL-1β ELISA kits from R&D Systems, Minneapolis, Minnesota; and IL-18 ELISA kits from MBL International Corporation, Woburn, Massachusetts), as previously described.114 The sensitivities of the assays were <4.67 pg/mL for NLRP1, <0.039 ng/mL for NLRP3, <0.056 ng/mL for AIM2, <6.25 pg/mL for NOD2, <0.112 ng/mL for CASP-1, <0.053 ng/mL for CASP-4, 3.3 pg/mL for pro-IL-1β, <1 pg/mL for mature IL-1β, and <12.5 pg/mL for IL-18. The IL-1β ELISA kit measured about 10% of the pro-IL-1β. The immunoassays for NLR family CARD domain-containing protein 4 (NLRC4) and NOD1 did not meet our criteria for validation; instead, immunoblotting was performed.

Immunohistochemistry

Tissue sections (5-μm thick) were prepared from the chorioamniotic membranes (PTL and PTL-ACA, n = 10 each). Immunohistochemistry for NLRP1, NLRP3, NLRC4, AIM2, NOD1, NOD2, CASP-1, CASP-4, IL-1β, and IL-18 was performed, as previously described.114 Supplementary Table 2 includes the utilized primary antibodies and immunostaining conditions. Quantification of the intensity was performed using a PerkinElmer Panoramic MIDI slide scanner (PerkinElmer, Waltham, Massachusetts).

Immunoblotting

The protein quantity of NLRC4 and NOD1, as well as the active/mature forms of CASP-1, CASP-4, and IL-18, was determined by immunoblotting. Tissue lysates (50 μg per well) were subjected to 4% to 12% sodium dodecyl sulphate-polyacrylamide gel electrophoresis (Invitrogen). Separated proteins were then transferred onto nitrocellulose membranes (Bio-Rad). Next, membranes were submerged in blocking solution (5% nonfat dry milk in Tris-buffered saline containing 0.1% Tween-20, Bio-Rad or StartingBlock T20 Block Buffer, ThermoFisher Scientific, Inc, Rockford, Illinois) and probed overnight at 4°C with the following human antibodies: mouse anti-NLRC4 antibody (BioLeg-end, San Diego, California), rabbit anti-NOD1 polyclonal antibody (Enzo Life Sciences, Farmingdale, New York174), mouse anti-CASP-1 monoclonal antibody (R&D Systems), rabbit anti-CASP-4 polyclonal antibody (Abcam, Cambridge, Massachusetts), or rabbit anti-IL-18 polyclonal antibody (Santa Cruz Biotechnology, Dallas, Texas). A horseradish peroxidase-conjugated anti-mouse or anti-rabbit immunoglobulin G (IgG; Cell Signaling, Boston, Massachusetts) was used as a secondary antibody. Chemiluminescence signals were detected with ChemiGlow West Reagents (Protein Simple, Santa Clara, California), and images were acquired using the FUJIFILM LAS-4000 Imaging System (FUJIFILM North America Corporation, Valhalla, New York). Finally, nitrocellulose membranes were then stripped with Restore Plus Western Blot Stripping Buffer (Pierce Biotechnology, ThermoFisher Scientific Inc) for 15 minutes, washed with 1X PBS, blocked, and reprobed for 1 hour at room temperature with a mouse anti-beta-actin (ACTB) monoclonal antibody (Sigma-Aldrich). Chemiluminescence signals were again detected with ChemiGlow West Reagents, and images were acquired using the FUJIFILM LAS-4000 Imaging System.

In Situ Proximity Ligation Assay

ASC/CASP-1 complex formation was detected by identifying protein interactions between ASC and CASP-1 using a Duolink in situ proximity ligation assay kit (Olink Bioscience, Uppsala, Sweden), following the manufacturer’s instructions. Briefly, chorioamniotic membrane tissues (PTL and PTL-ACA, n = 7 each) were frozen in Tissue-Plus O.C.T. compound (Fisher HealthCare, Houston, Texas) immediately after collection. Cryogenic sections were cut to 10 µm and placed on glass microscope slides (Fisherbrand Superfrost Plus slides; Thermo Scientific, Waltham, Massachusetts). The sections were fixed using 4% paraformaldehyde (Electron Microscopy Sciences, Hatfield, Pennsylvania) for 20 minutes at room temperature, rinsed with 1X PBS, and permeabilized using 0.25% Triton X-100 (Promega, Madison, Wisconsin) for 5 minutes at room temperature. Prior to staining, nonspecific antibody interactions were blocked using serum-free protein blocker (Cat#X09090; DAKO North America, Carpinteria, California) for 30 minutes at room temperature. The sections were then stained at 4°C overnight with the following antibodies: rabbit anti-human ASC (Cat#AG-25B-0006-C100; Adipogen, San Diego, California) and mouse anti-human CASP-1 (Cat#MAB6251, clone 661228; R&D Systems, Minneapolis, Minnesota). Rabbit IgG and mouse IgG2A were used as negative controls, respectively. Following staining, slides were briefly washed with 1X Wash Buffer A from the Duolink kit and incubated with the provided proximity ligation assay probes for 1 hour at 37°C, followed by a second wash with Wash Buffer A. The slides were then incubated with Duolink ligase solution for 30 minutes at 37°C, washed with Wash Buffer A, and incubated for 100 minutes with Duolink polymerase solution. The slides were washed with 1X Wash Buffer B and mounted with Duolink mounting media with DAPI (4’,6-diamidino-2-phenylindole). Immunofluorescence was visualized using an Olympus BX60 fluorescence microscope (Olympus, Tokyo, Japan) at 400× magnification. Images were acquired using an Olympus DP71 camera and DP Controller software (Olympus). Semi-quantification was performed using the Duolink image analysis software. Images and a video were acquired using a Zeiss LSM 800 laser scanning confocal microscope (Carl Zeiss Microscopy, Jena, Germany) at the Microscopy, Imaging, and Cytometry Resources Core at Wayne State University School of Medicine (http://micr.med.wayne.edu/). ASC/CASP-1 complex formation was calculated by dividing the number of signals over the area of the tissue, which was expressed as pixels.

Statistical Analyses

The SPSS version19.0 software (SPSS Inc, Chicago, Illinois) was used to analyze demographic and clinical data. Comparisons between the 2 groups (PTL vs PTL-ACA) were performed using the Fisher’s exact test for proportions as well as the Mann-Whitney U test for nonnormally distributed continuous variables. Gene expressions relative to ACTB/GAPDH/RPLP0 were calculated as −ΔCt values, where ΔCt (ΔCt = Cttarget − Ctreference) was computed for each sample after averaging the Ct values over the technical replicates. Group means of gene expression were then compared using t tests from an analysis of variance linear model and the resulting P values were adjusted using the Benjamini-Hochberg procedure. Spearman correlations were used to examine the relationship between inflammasome components and products. An adjusted P value of ≤.05 was considered statistically significant.

Results

mRNA Abundance and Protein Expression of Inflammasome Components and NOD Proteins in the Chorioamniotic Membranes in Spontaneous Preterm Labor with and without Acute Histologic Chorioamnionitis

First, the expression of major inflammasome components (NLRP1, NLRP3, AIM2, and NLRC4) and NOD proteins (NOD1 and NOD2) was determined in the chorioamniotic membranes from women who had undergone spontaneous preterm labor. The chorioamniotic membranes from women who had undergone spontaneous preterm labor with and without acute histologic chorioamnionitis expressed all of the inflammasome components. The mRNA abundance of NLRP1, NLRP3, NLRC4, and NOD2 was higher in the chorioamniotic membranes from women who underwent spontaneous preterm labor with acute histologic chorioamnionitis than in those without this placental lesion (Figures 1A and 2A). The protein concentrations of NLRP3 were greater in the chorioamniotic membranes from women who underwent spontaneous preterm labor with acute histologic chorioamnionitis than in those without this placental lesion (Figure 1B). Whereas the protein concentrations of AIM2 and NOD2 were lower in the chorioamniotic membranes from women who underwent spontaneous preterm labor with acute histologic chorioamnionitis than in those without this placental lesion (Figure 1B). The protein concentrations of NLRP1, NLRC4, and NOD1 were similar between these 2 groups (Figures 1B, 2B and E).

Figure 1.

Figure 1.

Inflammasome components and NOD2 protein in the chorioamniotic membranes. A, mRNA abundance of inflammasome components and NOD2 protein in the chorioamniotic membranes from women in spontaneous preterm labor with (PTL-ACA, n = 37) or without (PTL, n = 33) acute histologic chorioamnionitis. Relative gene expressions are presented as −ΔCt values. T tests from an analysis of variance (ANOVA) linear model and the resulting P values were adjusted using the Benjamini-Hochberg procedure. B, Protein concentrations of inflammasome components and NOD2 in chorioamniotic membrane tissue lysates (n = 10 each). Mann-Whitney U tests. C, Representative immunostainings for inflammasome components and NOD2 in the chorioamniotic membranes (n = 10 each), 200× magnification. Circles denote outlier values. mRNA indicates messenger RNA; NOD, nucleotide-binding oligomerization domain; PTL, preterm labor without acute histologic chorioamnionitis; PTL-ACA, preterm labor with acute histologic chorioamnionitis.

Figure 2.

Figure 2.

NLRC4 and NOD1 in the chorioamniotic membranes. (A and D) mRNA abundance of NLRC4 and NOD1 in the chorioamniotic membranes from women in spontaneous preterm labor with (PTL-ACA, n = 37) or without (PTL, n = 33) acute histologic chorioamnionitis. Relative gene expressions are presented as −ΔCt values. T tests from an analysis of variance (ANOVA) linear model and the resulting P values were adjusted using the Benjamini-Hochberg procedure. (B and E) Protein quantity of NLRC4 and NOD1 in chorioamniotic membrane tissue lysates (n = 6-9 each). Mann-Whitney U tests. (C and F) Intensity of the immunostainings for NLRC4 and NOD1 in the chorioamniotic membranes (n = 10 each) and representative immunostainings, 200× magnifications. Mann-Whitney U tests. Circles denote outlier values. mRNA indicates messenger RNA; NLRC4, NLR family CARD domain-containing protein 4; NOD, nucleotide-binding oligomerization domain; PTL, preterm labor without acute histologic chorioamnionitis; PTL-ACA, preterm labor with acute histologic chorioamnionitis.

Immunohistochemistry analysis revealed that all of the inflammasome components and NOD proteins were expressed in the chorioamniotic membranes from women who had undergone spontaneous preterm labor with and without acute histologic chorioamnionitis. The NLRP1, NLRP3, NLRC4, and NOD2 proteins showed strong immunoreactivity in the chorionic trophoblast cells, decidual stromal cells, and amniotic mesodermal and epithelial cells (Figures 1C and 2C). However, AIM2 and NOD1 showed weak immunoreactivity in the chorionic trophoblast cells, decidual stromal cells, and amniotic mesodermal and epithelial cells (Figures 1C and 2F).

The combined increase in the mRNA abundance and protein expression of NLRP3 in the chorioamniotic membranes from women who underwent spontaneous preterm labor with acute histologic chorioamnionitis suggests that the inflammasome may participate in the pathological activation of the innate immune system, leading to preterm labor in the setting of intraamniotic inflammation.

Activation of CASP-1 and CASP-4 in the Chorioamniotic Membranes in Spontaneous Preterm Labor with Acute Histologic Chorioamnionitis

Next, we examined whether the increased concentration of NLRP3 was linked to the activation of CASP-1 and CASP-4 in the chorioamniotic membranes from women who had undergone spontaneous preterm labor with acute histologic chorioamnionitis. The mRNA abundance of CASP1 and CASP4 was greater in the chorioamniotic membranes from women who underwent spontaneous preterm labor with acute histologic chorioamnionitis than in those without this placental lesion (Figure 3A). Although the protein concentrations of CASP-1 and CASP-4 were not significantly different between the 2 groups (Figure 3B), semiquantitative analysis of immunostaining revealed that CASP-1 was increased in the chorioamniotic membranes from women who underwent spontaneous preterm labor with acute histologic chorioamnionitis (Figure 3C). Also, the protein quantities of one of the active forms of CASP-1 (p10) and the active form of CASP-4 (p20) were increased in the chorioamniotic membranes from women who had undergone spontaneous preterm labor with acute histologic chorioamnionitis (Figure 4A, D, E, and G). In contrast, pro-CASP-1 was reduced in the chorioamniotic membranes from women who had undergone spontaneous preterm labor with acute histologic chorioamnionitis (Figure 4B). The active form of CASP-1 (p20) in the chorioamniotic membranes from women who had undergone spontaneous preterm labor with acute histologic chorioamnionitis seemed to be lower than in those without this placental lesion; yet, this reduction did not reach statistical significance (P = .07, Figure 4C). The quantity of the pro-CASP-4 was not different between these 2 groups (Figure 4F). Altogether, these findings provide evidence that there is activation of CASP-1 in the chorioamniotic membranes, which is likely mediated by the inflammasome and active CASP-4 during spontaneous preterm labor with acute histologic chorioamnionitis.

Figure 3.

Figure 3.

mRNA and protein expression of inflammatory caspases in the chorioamniotic membranes. A, mRNA abundance of caspase (CASP1) and CASP4 in the chorioamniotic membranes from women in spontaneous preterm labor with (PTL-ACA, n = 37) or without (PTL, n = 33) acute histologic chorioamnionitis. Relative gene expressions are presented as −ΔCt values. T tests from an analysis of variance (ANOVA) linear model and the resulting P values were adjusted using the Benjamini-Hochberg procedure. B, Protein concentrations of CASP-1 and CASP-4 in the chorioamniotic membrane tissue lysates (n = 10 each). Mann-Whitney U tests. C, Intensity of the immunostainings for CASP-1 and CASP-4 in the chorioamniotic membranes (n = 10 each) and representative immunostainings, 200× magnifications. Mann-Whitney U tests. Circles denote outlier values. mRNA indicates messenger RNA; PTL, preterm labor without acute histologic chorioamnionitis; PTL-ACA, preterm labor with acute histologic chorioamnionitis.

Figure 4.

Figure 4.

Inflammatory caspases in the chorioamniotic membranes. Immunoblotting of caspase (CASP)-1 (A) in the chorioamniotic membranes from women in spontaneous preterm labor with (PTL-ACA, n = 7) or without (PTL, n = 7) acute histologic chorioamnionitis. Quantification of the zymogen (B) and its active forms (C and D). Immunoblotting of CASP-4 (E) in the chorioamniotic membranes from women in spontaneous preterm labor with (PTL-ACA, n = 7) or without (PTL, n = 7) acute histologic chorioamnionitis. Quantification of the zymogen (F) and its active form (G). ACTB was used as an internal control and for quantifications. Mann-Whitney U tests; PTL, preterm labor without acute histologic chorioamnionitis; PTL-ACA, preterm labor with acute histologic chorioamnionitis.

Increased mRNA Abundance and Protein Expression of IL-1β in the Chorioamniotic Membranes in Spontaneous Preterm Labor with Acute Histologic Chorioamnionitis

Active forms of CASP-1 can convert pro-IL-1β into its mature form.154156,175,176 Active CASP-4 can also induce the noncanonical activation of IL-1β.171,177181 Hence, we determined whether the activation of CASP-1 and CASP-4 was correlated with the abundance of the mature form of IL-1β in the chorioamniotic membranes from women who had undergone spontaneous preterm labor with acute histologic chorioamnionitis. The mRNA abundance of IL1B in the chorioamniotic membranes from women who underwent spontaneous preterm labor with acute histologic chorioamnionitis was increased compared to those without this placental lesion (Figure 5A). The concentration of mature IL-1β was greater in the chorioamniotic membranes from women who had undergone spontaneous preterm labor with acute histologic chorioamnionitis than in those without this placental lesion (Figure 5B). In contrast, the concentration of pro-IL-1β was reduced in preterm cases with acute histologic chorioamnionitis (Figure 5B). Furthermore, IL-1β intensity was stronger in the chorioamniotic membranes from women who underwent spontaneous preterm labor with acute histologic chorioamnionitis than in those without this placental lesion (Figure 5C). These data suggest that the active forms of CASP-1 and CASP-4 may be involved in the processing of mature IL-1β in the chorioamniotic membranes during spontaneous preterm labor with acute histologic chorioamnionitis.

Figure 5.

Figure 5.

Interleukin (IL)-1β in the chorioamniotic membranes. A, mRNA abundance of IL1B in the chorioamniotic membranes from women in spontaneous preterm labor with (PTL-ACA, n = 37) or without (PTL, n = 33) acute histologic chorioamnionitis. Relative gene expressions are presented as −ΔCt values. T tests from an analysis of variance (ANOVA) linear model and the resulting P values were adjusted using the Benjamini-Hochberg procedure. Circles denote outlier values. B, Protein concentrations of the pro- and mature form of IL-1β in the chorioamniotic membrane lysates (n = 10 each). Mann-Whitney U tests. C, Intensity of the immunostainings for IL-1β in the chorioamniotic membranes (n = 10 each) and representative immunostainings, 200× magnifications. Mann-Whitney U tests. mRNA indicates messenger RNA; PTL, preterm labor without acute histologic chorioamnionitis; PTL-ACA, preterm labor with acute histologic chorioamnionitis.

An Increase in the Protein Expression and Mature Form of IL-18 in the Chorioamniotic Membranes in Spontaneous Preterm Labor with Acute Histologic Chorioamnionitis

Active forms of CASP-1 can convert pro-IL-18 into its mature form.157 Also, CASP-4 plays a role in the activation and release of IL-18 in response to enteric pathogens (eg, Salmonella typhimurium and enteropathogenic Escherichia coli) through intracellular endotoxin sensing.179 Next, we examined whether the activation of CASP-1 and CASP-4 was linked to the release of mature IL-18 by the chorioamniotic membranes from women who underwent spontaneous preterm labor with acute histologic chorioamnionitis. In line with previously published data,173 we found that the chorioamniotic membranes from women who had undergone spontaneous preterm labor with acute histologic chorioamnionitis had a greater protein concentration of IL-18 (Figure 6B) and a higher quantity of the mature form of this cytokine (Figure 6D) than those without this placental lesion. No significant differences were observed in the mRNA abundance (Figure 6A) and immunoreactivity (Figure 6C) of IL-18 in the chorioamniotic membranes between these 2 groups. These data demonstrate that the activation of CASP-1 and CASP-4 is associated with an increase of the protein concentration and mature form of IL-18 in the chorioamniotic membranes from women who had undergone spontaneous preterm labor with acute histologic chorioamnionitis.

Figure 6.

Figure 6.

Interleukin (IL)-18 in the chorioamniotic membranes. A, mRNA abundance of IL18 in the chorioamniotic membranes from women in spontaneous preterm labor with (PTL-ACA, n = 37) or without (PTL, n = 33) acute histologic chorioamnionitis. Relative gene expressions are presented as −ΔCt values. T tests from an analysis of variance (ANOVA) linear model and the resulting P values were adjusted using the Benjamini-Hochberg procedure. B, Protein concentrations of IL-18 in the chorioamniotic membrane tissue lysates (n = 10 each). Mann-Whitney U tests. C, Intensity of the immunostainings for IL-18 in the chorioamniotic membranes (n = 10 each) and representative immunostainings, 200× magnifications. Mann-Whitney U tests. D, Immunoblotting of IL-18 and its mature form in the chorioamniotic membranes and their quantifications (n = 7-9 each). ACTB was used as an internal control and for quantifications. Mann-Whitney U tests. Circles denote outlier values. mRNA indicates messenger RNA; PTL, preterm labor without acute histologic chorioamnionitis; PTL-ACA, preterm labor with acute histologic chorioamnionitis.

A subanalysis of the protein data demonstrated positive and significant correlations between (1) active CASP-1 and mature IL-1β (Spearman ρ = .58, P = .032, Supplementary Figure 1A); (2) NLRP3 and mature IL-1β (Spearman ρ = .76, P < .001, Supplementary Figure 1B); (3) active CASP-1 and NLRP3 (Spearman ρ = .69, P = .008, Supplementary Figure 1C); (4) mature IL-18 and NLRP3 (Spearman ρ = .53, P = .036, Supplementary Figure 1D); and (5) active CASP-4 and mature IL-1β (Spearman ρ = .61, P = .019, Supplementary Figure 1E). These results suggest that all of these proteins are increased in chorioamniotic membrane samples from women who underwent spontaneous preterm labor with acute histologic chorioamnionitis compared to those without this placental lesion.

Increased ASC/CASP-1 Complex Formation in the Chorioamniotic Membranes in Spontaneous Preterm Labor with Acute Histologic Chorioamnionitis

Since there were positive correlations between the NLRP3 protein, the active form of CASP-1, and the mature forms of IL-1β and IL-18, we next investigated whether there was inflammasome assembly in the chorioamniotic membranes from women who underwent spontaneous preterm labor with acute histologic chorioamnionitis. Inflammasome assembly includes the oligomerization of the NLR protein, adaptor protein ASC, and CASP-1;149,182 therefore, we used an in situ proximity ligation assay in order to determine the formation of ASC/CASP-1 complexes. ASC/CASP-1 complexes were identified in the chorioamniotic membranes from women who underwent spontaneous preterm labor with and without acute histologic chorioamnionitis (Figure 7A and B). However, ASC/CASP-1 complexes were greater in the chorioamniotic membranes from women who had undergone spontaneous preterm labor with acute histologic chorioamnionitis than in those without this placental lesion (Figure 7C). ASC/CASP-1 complexes were not detected in isotype controls (Figure 7D and E). A 3D reconstruction shows that there are ASC/CASP-1 complexes in all of the layers of the chorioamniotic membranes from women who underwent spontaneous preterm labor with acute histologic chorioamnionitis (Supplementary Video 1). These findings provide evidence that there is inflammasome assembly in the chorioamniotic membranes during spontaneous preterm labor with acute histologic chorioamnionitis.

Figure 7.

Figure 7.

ASC/caspase (CASP)-1 complex formation in the chorioamniotic membranes. In situ proximity ligation assays for ASC and CASP-1 in the chorioamniotic membranes from women in spontaneous preterm labor with (PTL-ACA) or without (PTL) acute histologic chorioamnionitis. (A and B) Representative images of ASC/CASP-1 complexes (red signal) in PTL and PTL-ACA groups. (D and E) Representative images of isotype controls in PTL and PTL-ACA groups. The blue signal is DAPI (nuclei). 400× magnifications. C, Semi-quantification of the number of ASC/CASP-1 complexes in the chorioamniotic membranes (PTL and PTL-ACA, n = 7 each). ASC/CASP-1 complexes were calculated by dividing the number of signals over the area of the tissue, which was expressed as pixels. Mann-Whitney U tests.

Discussion

Principal Findings of the Study

When comparing the chorioamniotic membranes from women in spontaneous preterm labor with acute histologic chorioamnionitis to those without this placental lesion, we found that (1) the mRNA expression of NLRP1, NLRP3, NLRC4, and NOD2 was higher; (2) the NLRP3 protein was increased; (3) the mRNA and active form of CASP-1 were greater; (4) the mRNA and active form of CASP-4 were increased; (5) the mRNA and mature form of IL-1β were higher; (6) the mature form of IL-18 was elevated; and (7) ASC/CASP-1 complexes were increased. Altogether, these findings provide the first evidence that supports a role for the inflammasome in the pathological inflammatory process in the chorioamniotic membranes from women who had undergone spontaneous preterm labor with acute histologic chorioamnionitis.

A role for the inflammasome in the physiological112114 and pathological113,114 inflammatory process of term parturition has been previously reported. Herein, we provided the first evidence that the NLRP3 inflammasome is involved in the pathological pro-inflammatory process of preterm parturition. The NLRP3 inflammasome includes the NLRP3 protein, the adaptor molecule ASC with 2 death-fold domains (1 pyrin domain and 1 CARD), and pro-CASP-1.144,151,183,184 Several stimuli185191 including crystalline material,187,192 necrosis-derived extracellular adenosine triphosphate,193 vaccine adjuvants,194198 phospholipid cardiolipin and mitochondrial DNA,199201 bacterial toxins,193,202,203 and other DAMPs189,190 and pathogen-associated molecular patterns (PAMPs)120,204214 can induce the activation of the NLRP3 inflammasome. The process of activation of this inflammasome includes 2 steps—the priming and the oligomerization or assembly of the multiprotein complex.182,215 The first step includes the sensing of the DAMP and/or PAMP via the PRR which, in turn, induces the activation of the nuclear factor kappa B pathway, resulting in the upregulation of the NLRP3 protein and the expression of pro-IL-1β.182,215,216 The second step permits the oligomerization or assembly of the inflammasome complex, which includes the NLRP3, ASC, and CASP-1 proteins.182,215 The current study provides evidence that there is priming and assembly of the NLRP3 inflammasome complex in the chorioamniotic membranes during spontaneous preterm labor with acute histologic chorioamnionitis. It is worth mentioning that there were some ASC/CASP-1 complexes (ie, inflammasome assembly) in the chorioamniotic membranes from women who underwent spontaneous preterm labor without acute chorioamnionitis. These data suggest that the inflammasome is also involved in the process of preterm parturition in the absence of acute histologic chorioamnionitis.

Oligomerization of the inflammasome leads to the recruitment of ASC, which binds and activates pro-CASP-1 via its CARD.144,217 In the study herein, we found that the mRNA abundance, immunoreactivity, and the active form of CASP-1 (p10) are increased in the chorioamniotic membranes in spontaneous preterm labor with acute histologic chorioamnionitis. This finding is consistent with previous studies demonstrating that: (1) amniotic fluid CASP-1 concentrations are greater in women who underwent spontaneous preterm labor with intraamniotic infection/inflammation than in those who underwent preterm labor without this clinical condition;113 and (2) CASP-1 concentration and the abundance of its mature form are increased in the uteri of mice prior to inflammation-induced preterm birth (mice injected with peptidoglycan and polyinosinic-polycytidylic acid) compared to term controls.218 Together, these data suggest that during spontaneous preterm labor with acute histologic chorioamnionitis, the chorioamniotic membranes release abundant quantities of the active form of CASP-1, which is most likely mediated by the inflammasome.

CASP-4 (murine homologue CASP-11) is implicated in the activation of CASP-1 and participates in the non-cannonical activation of the inflammasome.149,172,213,219 In the current study, we found that the mRNA abundance of CASP-4 and its mature form (p20) were increased in the chorioamniotic membranes from women who underwent spontaneous preterm labor with acute histologic chorioamnionitis. These data indicate that CASP-4 could be mediating the noncanonical activation of the inflammasome in spontaneous preterm labor with acute histologic chorioamnionitis. We suggest that the activation of CASP-4 is also implicated in the induction of preterm parturition in the setting of sterile intraamniotic inflammation. This concept is based on 2 observations—(1) CASP-4 can induce the release of alarmins in a CASP-1–independent manner172 and (2) the intraamniotic administration of HMGB1 (a classic alarmin220) can induce preterm labor and birth.221

Following activation, CASP-1 converts inactive pro-IL-1β into its mature and secreted form.155,222228 IL-1β induces the expression and release of different mediators implicated in the process of labor.229235 Indeed, the administration of IL-1β causes preterm birth in mice 236,237 and monkeys,238245 confirming an essential role for this cytokine in the pathological process of labor. This effect can be abrogated by the administration of the IL-1β receptor antagonist.237 In the study herein, we demonstrated that the chorioamniotic membranes from women in spontaneous preterm labor with acute histologic chorioamnionitis released high amounts of mature IL-1β, which is most likely mediated by the active forms of CASP-1. This mature form of IL-1β will then participate in the pathological pro-inflammatory milieu that accompanies the premature process of labor. It is worth mentioning that pro-IL-1β was reduced in the chorioamniotic membranes from women in spontaneous preterm labor with acute histologic chorioamnionitis compared to those without this placental lesion, which is most likely due to the processing of this zymogen into its mature form.

The active forms of CASP-1157160,163 and CASP-4179 can also convert pro-IL-18 into its mature form. IL-18 is present in the amniotic fluid, and its concentration is greater in women who underwent spontaneous preterm labor with microbial invasion of the amniotic cavity than in those without this clinical condition.173,246 In the present study, the total concentration and mature form of IL-18 was increased in the chorioamniotic membranes from women who underwent spontaneous preterm labor with acute histologic chorioamnionitis. IL-18 is a major interferon γ inducing factor that activates Th-1 responses in T cells and natural killer cells.247254 Therefore, it is likely that IL-18 participates in the cellular pro-inflammatory milieu in the chorioamniotic membranes during spontaneous preterm labor with acute histologic chorioamnionitis.

Conclusion

Herein, we provide the first evidence that supports a role for the NLRP3 inflammasome and CASP-4 in the activation of CASP-1 and the consequent release of mature IL-1β and IL-18 by the chorioamniotic membranes from women who underwent spontaneous preterm labor with acute histologic chorioamnionitis. These findings provide insight into the mechanisms that are implicated in the pathological pro-inflammatory process of spontaneous preterm labor in acute inflammation of the chorioamniotic membranes.

Supplementary Material

Supplementary material
Supplementary material
Supplementary_tables.pdf (32.6KB, pdf)
Supplementary material
Download video file (30.4MB, avi)

Acknowledgments

The authors gratefully acknowledge Valeria Garcia-Flores, Derek Miller, Lorri McLuckie, Rona Wang, Yang Jiang, Gaurav Bhatti, and Zhonghui Xu for their contributions to the execution of this study. The authors 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. The authors also thank staff members of the PRB Clinical Laboratory and the PRB Histology/Pathology Unit for the processing and examination of 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.

Supplemental Material: Supplementary material is available for this article online.

References

  • 1. Romero R, Mazor M, Munoz H, Gomez R, Galasso M, Sherer DM. The preterm labor syndrome. Ann N Y Acad Sci. 1994;734:414–429. [DOI] [PubMed] [Google Scholar]
  • 2. Romero R. Prenatal medicine: the child is the father of the man. 1996. Prenatal Neonatal Med. 1996;1:8–11. [DOI] [PubMed] [Google Scholar]
  • 3. Romero R, Espinoza J, Kusanovic JP, et al. The preterm parturition syndrome. BJOG. 2006;113(suppl 3):17–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Gotsch F, Romero R, Erez O, et al. The preterm parturition syndrome and its implications for understanding the biology, risk assessment, diagnosis, treatment and prevention of preterm birth. J Matern Fetal Neonatal Med. 2009;22(suppl 2):5–23. [DOI] [PubMed] [Google Scholar]
  • 5. Romero R, Lockwood CJ. Pathogenesis of spontaneous preterm labor In: Creasy RK, Resnik R, Iams J, eds. Creasy and Resnik’s Maternal–Fetal Medicine: Principles and Practice. 6th ed Philadelphia, PA: Elsevier; 2009:521–543. [Google Scholar]
  • 6. Romero R. Prenatal medicine: the child is the father of the man. 1996. J Matern Fetal Neonatal Med. 2009;22(8):636–639. [DOI] [PubMed] [Google Scholar]
  • 7. Muglia LJ, Katz M. The enigma of spontaneous preterm birth. N Engl J Med. 2010;362(6):529–535. [DOI] [PubMed] [Google Scholar]
  • 8. Blencowe H, Cousens S, Oestergaard MZ, et al. National, regional, and worldwide estimates of preterm birth rates in the year 2010 with time trends since 1990 for selected countries: a systematic analysis and implications. Lancet. 2012;379(9832):2162–2172. [DOI] [PubMed] [Google Scholar]
  • 9. Hamilton BE, Hoyert DL, Martin JA, Strobino DM, Guyer B. Annual summary of vital statistics: 2010–2011. Pediatrics. 2013;131(3):548–558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Liu L, Oza S, Hogan D, et al. Global, regional, and national causes of child mortality in 2000–13, with projections to inform post-2015 priorities: an updated systematic analysis. Lancet. 2015;385(9966):430–440. [DOI] [PubMed] [Google Scholar]
  • 11. Berkowitz GS, Blackmore-Prince C, Lapinski RH, Savitz DA. Risk factors for preterm birth subtypes. Epidemiology. 1998;9(3):279–285. [PubMed] [Google Scholar]
  • 12. Moutquin JM. Classification and heterogeneity of preterm birth. BJOG. 2003;110(suppl 20):30–33. [DOI] [PubMed] [Google Scholar]
  • 13. Goldenberg RL, Culhane JF, Iams JD, Romero R. Epidemiology and causes of preterm birth. Lancet. 2008;371(9606):75–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Romero R, Dey SK, Fisher SJ. Preterm labor: one syndrome, many causes. Science. 2014;345(6198):760–765. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Bobitt JR, Ledger WJ. Unrecognized amnionitis and prematurity: a preliminary report. J Reprod Med. 1977;19(1):8–12. [PubMed] [Google Scholar]
  • 16. Bobitt JR, Ledger WJ. Amniotic fluid analysis. Its role in maternal neonatal infection. Obstet Gynecol. 1978;51(1):56–62. [PubMed] [Google Scholar]
  • 17. Miller JM, Jr, Pupkin MJ, Hill GB. Bacterial colonization of amniotic fluid from intact fetal membranes. Am J Obstet Gynecol. 1980;136(6):796–804. [DOI] [PubMed] [Google Scholar]
  • 18. Bobitt JR, Hayslip CC, Damato JD. Amniotic fluid infection as determined by transabdominal amniocentesis in patients with intact membranes in premature labor. Am J Obstet Gynecol. 1981;140(8):947–952. [DOI] [PubMed] [Google Scholar]
  • 19. Wallace RL, Herrick CN. Amniocentesis in the evaluation of premature labor. Obstet Gynecol. 1981;57(4):483–486. [PubMed] [Google Scholar]
  • 20. Wahbeh CJ, Hill GB, Eden RD, Gall SA. Intra-amniotic bacterial colonization in premature labor. Am J Obstet Gynecol. 1984;148(6):739–743. [DOI] [PubMed] [Google Scholar]
  • 21. 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]
  • 22. Romero R, Mazor M. Infection and preterm labor. Clin Obstet Gynecol. 1988;31(3):553–584. [DOI] [PubMed] [Google Scholar]
  • 23. 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(2675611):817–824. [DOI] [PubMed] [Google Scholar]
  • 24. Romero R, Avila C, Brekus CA, Morotti R. The role of systemic and intrauterine infection in preterm parturition. Ann N Y Acad Sci. 1991;622:355–375. [DOI] [PubMed] [Google Scholar]
  • 25. Watts DH, Krohn MA, Hillier SL, Eschenbach DA. The association of occult amniotic fluid infection with gestational age and neonatal outcome among women in preterm labor. Obstet Gynecol. 1992;79(3):351–357. [DOI] [PubMed] [Google Scholar]
  • 26. Gibbs RS, Romero R, Hillier SL, Eschenbach DA, Sweet RL. A review of premature birth and subclinical infection. Am J Obstet Gynecol. 1992;166(5):1515–1528. [DOI] [PubMed] [Google Scholar]
  • 27. Gomez R, Romero R, Edwin SS, David C. Pathogenesis of preterm labor and preterm premature rupture of membranes associated with intraamniotic infection. Infect Dis Clin North Am. 1997;11(1):135–176. [DOI] [PubMed] [Google Scholar]
  • 28. Romero R, Gomez R, Chaiworapongsa T, Conoscenti G, Kim JC, Kim YM. The role of infection in preterm labour and delivery. Paediatr Perinat Epidemiol. 2001;15(suppl 2):41–56. [DOI] [PubMed] [Google Scholar]
  • 29. Yoon BH, Romero R, Moon JB, et al. The frequency and clinical significance of intra-amniotic inflammation in patients with a positive cervical fetal fibronectin. Am J Obstet Gynecol. 2001;185(5):1137–1142. [DOI] [PubMed] [Google Scholar]
  • 30. 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]
  • 31. Romero R, Espinoza J, Chaiworapongsa T, Kalache K. Infection and prematurity and the role of preventive strategies. Semin Neonatol. 2002;7(4):259–274. [DOI] [PubMed] [Google Scholar]
  • 32. Shim SS, Romero R, Hong JS, et al. Clinical significance of intra-amniotic inflammation in patients with preterm premature rupture of membranes. Am J Obstet Gynecol. 2004;191(4):1339–1345. [DOI] [PubMed] [Google Scholar]
  • 33. Romero R, Gotsch F, Pineles B, Kusanovic JP. Inflammation in pregnancy: its roles in reproductive physiology, obstetrical complications, and fetal injury. Nutr Rev. 2007;65(12 pt 2):S194–S202. [DOI] [PubMed] [Google Scholar]
  • 34. 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–e296. [DOI] [PubMed] [Google Scholar]
  • 35. Lee SE, Romero R, Park CW, Jun JK, Yoon BH. The frequency and significance of intraamniotic inflammation in patients with cervical insufficiency. Am J Obstet Gynecol. 2008;198(6):633.e631–e638. [DOI] [PubMed] [Google Scholar]
  • 36. Lee SE, Romero R, Lee SM, Yoon BH. Amniotic fluid volume in intra-amniotic inflammation with and without culture-proven amniotic fluid infection in preterm premature rupture of membranes. J Perinat Med. 2010;38(1):39–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. 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]
  • 38. Kim SM, Romero R, Lee J, et al. The frequency and clinical significance of intra-amniotic inflammation in women with preterm uterine contractility but without cervical change: do the diagnostic criteria for preterm labor need to be changed? J Matern Fetal Neonatal Med. 2012;25(8):1212–1221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Blanc WA. Amniotic infection syndrome; pathogenesis, morphology, and significance in circumnatal mortality. Clin Obstet Gynecol. 1959;2:705–734. [PubMed] [Google Scholar]
  • 40. Russell P. Inflammatory lesions of the human placenta: clinical significance of acute chorioamnionitis. Am J Diagn Gynecol Obstet. 1979;2:127–137. [Google Scholar]
  • 41. Blanc WA. Pathology of the placenta and cord in ascending and in haematogenous infection. Ciba Found Symp. 1979(77):17–38. [DOI] [PubMed] [Google Scholar]
  • 42. Hillier SL, Martius J, Krohn M, Kiviat N, Holmes KK, Eschenbach DA. A case–control study of chorioamnionic infection and histologic chorioamnionitis in prematurity. N Engl J Med. 1988;319(15):972–978. [DOI] [PubMed] [Google Scholar]
  • 43. Hillier SL, Krohn MA, Kiviat NB, Watts DH, Eschenbach DA. Microbiologic causes and neonatal outcomes associated with chorioamnion infection. Am J Obstet Gynecol. 1991;165(4 pt 1):955–961. [DOI] [PubMed] [Google Scholar]
  • 44. Romero R, Salafia CM, Athanassiadis AP, et al. The relationship between acute inflammatory lesions of the preterm placenta and amniotic fluid microbiology. Am J Obstet Gynecol. 1992;166(5):1382–1388. [DOI] [PubMed] [Google Scholar]
  • 45. Redline RW, Faye-Petersen O, Heller D, Qureshi F, Savell V, Vogler C; Society for Pediatric Pathology, Perinatal Section, Amniotic Fluid Infection Nosology Committee. Amniotic infection syndrome: nosology and reproducibility of placental reaction patterns. Pediatr Dev Pathol. 2003;6(5):435–448. [DOI] [PubMed] [Google Scholar]
  • 46. Redline RW. Placental inflammation. Semin Neonatol. 2004;9(4):265–274. [DOI] [PubMed] [Google Scholar]
  • 47. Fox H, Sebire NJ. Infections and inflammatory lesions of the placenta. Pathology of the placenta. 3rd ed Edinburgh, Scotland: Elsevier Saunders; 2007:303–354. [Google Scholar]
  • 48. Benirschke K, Burton G, Baergen R. Infectious diseases. Pathology of the Human Placenta: Springer Berlin Heidelberg; 2012:557–655. [Google Scholar]
  • 49. Guzick DS, Winn K. The association of chorioamnionitis with preterm delivery. Obstet Gynecol. 1985;65(1):11–16. [PubMed] [Google Scholar]
  • 50. van Hoeven KH, Anyaegbunam A, Hochster H, et al. Clinical significance of increasing histologic severity of acute inflammation in the fetal membranes and umbilical cord. Pediatr Pathol Lab Med. 1996;16(5):731–744. [PubMed] [Google Scholar]
  • 51. Srinivas SK, Ma Y, Sammel MD, et al. Placental inflammation and viral infection are implicated in second trimester pregnancy loss. Am J Obstet Gynecol. 2006;195(3):797–802. [DOI] [PubMed] [Google Scholar]
  • 52. Srinivas SK, Ernst LM, Edlow AG, Elovitz MA. Can placental pathology explain second-trimester pregnancy loss and subsequent pregnancy outcomes? Am J Obstet Gynecol. 2008;199(4):402.e401–405. [DOI] [PubMed] [Google Scholar]
  • 53. Taguchi A, Yamashita A, Kawana K, et al. Recent progress in therapeutics for inflammation-associated preterm birth: a review [published online December 1, 2015]. Reprod Sci. pii: 1933719115618282. [DOI] [PubMed] [Google Scholar]
  • 54. 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]
  • 55. 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]
  • 56. 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]
  • 57. 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]
  • 58. 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]
  • 59. 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]
  • 60. Romero R, Chaemsaithong P, Korzeniewski SJ, et al. Clinical chorioamnionitis at term III: how well do clinical criteria perform in the identification of proven intra-amniotic infection? J Perinat Med. 2016;44(1):23–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Romero R, Chaemsaithong P, Docheva N, et al. Clinical chorioamnionitis at term IV: the maternal plasma cytokine profile. J Perinat Med. 2016;44(1):77–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Romero R, Chaemsaithong P, Docheva N, et al. Clinical chorioamnionitis at term V: umbilical cord plasma cytokine profile in the context of a systemic maternal inflammatory response. J Perinat Med. 2016;44(1):53–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Romero R, Chaemsaithong P, Docheva N, et al. 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(1):33–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Rubartelli A, Lotze MT. Inside, outside, upside down: damage-associated molecular-pattern molecules (DAMPs) and redox. Trends Immunol. 2007;28(10):429–436. [DOI] [PubMed] [Google Scholar]
  • 65. Lotze MT, Zeh HJ, Rubartelli A, et al. The grateful dead: damage-associated molecular pattern molecules and reduction/oxidation regulate immunity. Immunol Rev. 2007;220:60–81. [DOI] [PubMed] [Google Scholar]
  • 66. Oppenheim JJ, Yang D. Alarmins: chemotactic activators of immune responses. Curr Opin Immunol. 2005;17(4):359–365. [DOI] [PubMed] [Google Scholar]
  • 67. Chen GY, Nunez G. Sterile inflammation: sensing and reacting to damage. Nat Rev Immunol. 2010;10(12):826–837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. 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(suppl 4): S29–S52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Redline RW. Classification of placental lesions. Am J Obstet Gynecol. 2015;213(4 Suppl):S21–8. [DOI] [PubMed] [Google Scholar]
  • 70. Kim CJ, Romero R, Kusanovic JP, et al. The frequency, clinical significance, and pathological features of chronic chorioamnionitis: a lesion associated with spontaneous preterm birth. Mod Pathol. 2010;23(7):1000–1011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Toti P, Arcuri F, Tang Z, et al. Focal increases of fetal macrophages in placentas from pregnancies with histological chorioamnionitis: potential role of fibroblast monocyte chemotactic protein-1. Am J Reprod Immunol. 2011;65(5):470–479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Redline RW. Inflammatory response in acute chorioamnionitis. Semin Fetal Neonatal Med. 2012;17(1):20–25. [DOI] [PubMed] [Google Scholar]
  • 73. 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]
  • 74. Kacerovsky M, Drahosova M, Hornychova H, et al. Value of amniotic fluid interleukin-8 for the prediction of histological chorioamnionitis in preterm premature rupture of membranes. Neuro Endocrinol Lett. 2009;30(6):733–738. [PubMed] [Google Scholar]
  • 75. Mittal P, Romero R, Kusanovic JP, et al. CXCL6 (granulocyte chemotactic protein-2): a novel chemokine involved in the innate immune response of the amniotic cavity. Am J Reprod Immunol. 2008;60(3):246–257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Esplin MS, Romero R, Chaiworapongsa T, et al. Monocyte chemotactic protein-1 is increased in the amniotic fluid of women who deliver preterm in the presence or absence of intra-amniotic infection. J Matern Fetal Neonatal Med. 2005;17(6):365–373. [DOI] [PubMed] [Google Scholar]
  • 77. Romero R, Avila C, Santhanam U, Sehgal PB. Amniotic fluid interleukin 6 in preterm labor. Association with infection. J Clin Invest. 1990;85(5):1392–1400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. 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]
  • 79. Romero R, Yoon BH, Kenney JS, Gomez R, Allison AC, Sehgal PB. Amniotic fluid interleukin-6 determinations are of diagnostic and prognostic value in preterm labor. Am J Reprod Immunol. 1993;30(2-3):167–183. [DOI] [PubMed] [Google Scholar]
  • 80. Hillier SL, Witkin SS, Krohn MA, Watts DH, Kiviat NB, Eschenbach DA. The relationship of amniotic fluid cytokines and preterm delivery, amniotic fluid infection, histologic chorioamnionitis, and chorioamnion infection. Obstet Gynecol. 1993;81(6):941–948. [PubMed] [Google Scholar]
  • 81. Arntzen KJ, Kjollesdal AM, Halgunset J, Vatten L, Austgulen R. TNF, IL-1, IL-6, IL-8 and soluble TNF receptors in relation to chorioamnionitis and premature labor. J Perinat Med. 1998;26(1):17–26. [DOI] [PubMed] [Google Scholar]
  • 82. Yoon BH, Jun JK, Romero R, et al. Amniotic fluid inflammatory cytokines (interleukin-6, interleukin-1beta, and tumor necrosis factor-alpha), neonatal brain white matter lesions, and cerebral palsy. Am J Obstet Gynecol. 1997;177(1):19–26. [DOI] [PubMed] [Google Scholar]
  • 83. Yoon BH, Romero R, Jun JK, et al. Amniotic fluid cytokines (interleukin-6, tumor necrosis factor-alpha, interleukin-1 beta, and interleukin-8) and the risk for the development of bronchopulmonary dysplasia. Am J Obstet Gynecol. 1997;177(4):825–830. [DOI] [PubMed] [Google Scholar]
  • 84. Figueroa R, Garry D, Elimian A, Patel K, Sehgal PB, Tejani N. Evaluation of amniotic fluid cytokines in preterm labor and intact membranes. J Matern Fetal Neonatal Med. 2005;18(4):241–247. [DOI] [PubMed] [Google Scholar]
  • 85. Holst RM, Laurini R, Jacobsson B, et al. Expression of cytokines and chemokines in cervical and amniotic fluid: relationship to histological chorioamnionitis. J Matern Fetal Neonatal Med. 2007;20(12):885–893. [DOI] [PubMed] [Google Scholar]
  • 86. Kacerovsky M, Celec P, Vlkova B, et al. Amniotic fluid protein profiles of intraamniotic inflammatory response to Ureaplasma spp. and other bacteria. PLoS One. 2013;8(3):e60399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. 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]
  • 88. 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]
  • 89. Saito M, Payne MS, Miura Y, et al. Polymyxin B agonist capture therapy for intrauterine inflammation: proof-of-principle in a fetal ovine model. Reprod Sci. 2014;21(5):623–631. [DOI] [PubMed] [Google Scholar]
  • 90. Lockwood CJ, Arcuri F, Toti P, et al. Tumor necrosis factor-alpha and interleukin-1beta regulate interleukin-8 expression in third trimester decidual cells: implications for the genesis of chorioamnionitis. Am J Pathol. 2006;169(4):1294–1302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Lockwood CJ, Murk WK, Kayisli UA, et al. Regulation of interleukin-6 expression in human decidual cells and its potential role in chorioamnionitis. Am J Pathol. 2010;177(4):1755–1764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Yoon BH, Romero R, Yang SH, et al. Interleukin-6 concentrations in umbilical cord plasma are elevated in neonates with white matter lesions associated with periventricular leukomalacia. Am J Obstet Gynecol. 1996;174(5):1433–1440. [DOI] [PubMed] [Google Scholar]
  • 93. Dollner H, Vatten L, Halgunset J, Rahimipoor S, Austgulen R. Histologic chorioamnionitis and umbilical serum levels of pro-inflammatory cytokines and cytokine inhibitors. BJOG. 2002;109(5):534–539. [PubMed] [Google Scholar]
  • 94. Andrys C, Drahosova M, Hornychova H, et al. Umbilical cord blood concentrations of IL-6, IL-8, and MMP-8 in pregnancy complicated by preterm premature rupture of the membranes and histological chorioamnionitis. Neuro Endocrinol Lett. 2010;31(6):857–863. [PubMed] [Google Scholar]
  • 95. 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(1):81–88. [DOI] [PubMed] [Google Scholar]
  • 96. Yoon BH, Romero R, Park JS, et al. Fetal exposure to an intra-amniotic inflammation and the development of cerebral palsy at the age of three years. Am J Obstet Gynecol. 2000;182(3):675–681. [DOI] [PubMed] [Google Scholar]
  • 97. Hitti J, Tarczy-Hornoch P, Murphy J, Hillier SL, Aura J, Eschenbach DA. Amniotic fluid infection, cytokines, and adverse outcome among infants at 34 weeks’ gestation or less. Obstet Gynecol. 2001;98(6):1080–1088. [DOI] [PubMed] [Google Scholar]
  • 98. Moon JB, Kim JC, Yoon BH, et al. Amniotic fluid matrix metalloproteinase-8 and the development of cerebral palsy. J Perinat Med. 2002;30(4):301–306. [DOI] [PubMed] [Google Scholar]
  • 99. 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]
  • 100. Kunze M, Klar M, Morfeld CA, et al. Cytokines in noninvasively obtained amniotic fluid as predictors of fetal inflammatory response syndrome. Am J Obstet Gynecol. 2016;215(1):96.e1–96.e8. [DOI] [PubMed] [Google Scholar]
  • 101. Weeks JW, Reynolds L, Taylor D, Lewis J, Wan T, Gall SA. Umbilical cord blood interleukin-6 levels and neonatal morbidity. Obstet Gynecol. 1997;90(5):815–818. [DOI] [PubMed] [Google Scholar]
  • 102. Gomez R, Romero R, Ghezzi F, Yoon BH, Mazor M, Berry SM. The fetal inflammatory response syndrome. Am J Obstet Gynecol. 1998;179(1):194–202. [DOI] [PubMed] [Google Scholar]
  • 103. Berner R, Niemeyer CM, Leititis JU, et al. Plasma levels and gene expression of granulocyte colony-stimulating factor, tumor necrosis factor-alpha, interleukin (IL)-1beta, IL-6, IL-8, and soluble intercellular adhesion molecule-1 in neonatal early onset sepsis. Pediatr Res. 1998;44(4):469–477. [DOI] [PubMed] [Google Scholar]
  • 104. Dollner H, Vatten L, Linnebo I, Zanussi GF, Laerdal A, Austgulen R. Inflammatory mediators in umbilical plasma from neonates who develop early-onset sepsis. Biol Neonate. 2001;80(1):41–47. [DOI] [PubMed] [Google Scholar]
  • 105. Goepfert AR, Andrews WW, Carlo W, et al. Umbilical cord plasma interleukin-6 concentrations in preterm infants and risk of neonatal morbidity. Am J Obstet Gynecol. 2004;191(4):1375–1381. [DOI] [PubMed] [Google Scholar]
  • 106. An H, Nishimaki S, Ohyama M, et al. Interleukin-6, interleukin-8, and soluble tumor necrosis factor receptor-I in the cord blood as predictors of chronic lung disease in premature infants. Am J Obstet Gynecol. 2004;191(5):1649–1654. [DOI] [PubMed] [Google Scholar]
  • 107. Elsmen E, Ley D, Cilio CM, Hansen-Pupp I, Hellstrom-Westas L. Umbilical cord levels of interleukin-1 receptor antagonist and neonatal outcome. Biol Neonate. 2006;89(4):220–226. [DOI] [PubMed] [Google Scholar]
  • 108. Satar M, Turhan E, Yapicioglu H, Narli N, Ozgunen FT, Cetiner S. Cord blood cytokine levels in neonates born to mothers with prolonged premature rupture of membranes and its relationship with morbidity and mortality. Eur Cytokine Netw. 2008;19(1):37–41. [DOI] [PubMed] [Google Scholar]
  • 109. Liu J, Feng ZC. Increased umbilical cord plasma interleukin-1 beta levels was correlated with adverse outcomes of neonatal hypoxic-ischemic encephalopathy. J Trop Pediatr. 2010;56(3):178–182. [DOI] [PubMed] [Google Scholar]
  • 110. Armstrong-Wells J, Donnelly M, Post MD, Manco-Johnson MJ, Winn VD, Sebire G. Inflammatory predictors of neurologic disability after preterm premature rupture of membranes. Am J Obstet Gynecol. 2015;212(2):212.e211–212.e219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Cordeiro CN, Savva Y, Vaidya D, et al. Mathematical modeling of the biomarker milieu to characterize preterm birth and predict adverse neonatal outcomes. Am J Reprod Immunol. 2016;75(5):594–601. [DOI] [PubMed] [Google Scholar]
  • 112. Pineles BL, Romero R, Montenegro D, et al. “The inflammasome” in human parturition. Reprod Sci. 2007;14(1):59A.17636217 [Google Scholar]
  • 113. Gotsch F, Romero R, Chaiworapongsa T, et al. 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(9):605–616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. 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:10.1111/aji.12440. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Gomez-Lopez N, Romero R, Xu Y, et al. A role for the inflammasome in spontaneous labor at term with acute histologic chorioamnionitis [published online November 16, 2016]. pii: 1933719116675058. [DOI] [PMC free article] [PubMed]
  • 116. 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(2):417–426. [DOI] [PubMed] [Google Scholar]
  • 117. Petrilli V, Papin S, Tschopp J. The inflammasome. Curr Biol. 2005;15(15):R581. [DOI] [PubMed] [Google Scholar]
  • 118. Ogura Y, Sutterwala FS, Flavell RA. The inflammasome: first line of the immune response to cell stress. Cell. 2006;126(4):659–662. [DOI] [PubMed] [Google Scholar]
  • 119. Sutterwala FS, Ogura Y, Flavell RA. The inflammasome in pathogen recognition and inflammation. J Leukoc Biol. 2007;82(2):259–264. [DOI] [PubMed] [Google Scholar]
  • 120. Mariathasan S, Monack DM. Inflammasome adaptors and sensors: intracellular regulators of infection and inflammation. Nat Rev Immunol. 2007;7(1):31–40. [DOI] [PubMed] [Google Scholar]
  • 121. Stutz A, Golenbock DT, Latz E. Inflammasomes: too big to miss. J Clin Invest. 2009;119(12):3502–3511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. 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(3):241–247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Jha S, Ting JP. Inflammasome-associated nucleotide-binding domain, leucine-rich repeat proteins and inflammatory diseases. J Immunol. 2009;183(12):7623–7629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Pedra JH, Cassel SL, Sutterwala FS. Sensing pathogens and danger signals by the inflammasome. Curr Opin Immunol. 2009;21(1):10–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Lamkanfi M, Dixit VM. Inflammasomes: guardians of cytosolic sanctity. Immunol Rev. 2009;227(1):95–105. [DOI] [PubMed] [Google Scholar]
  • 126. Latz E. The inflammasomes: mechanisms of activation and function. Curr Opin Immunol. 2010;22(1):28–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Schroder K, Tschopp J. The inflammasomes. Cell. 2010;140(6):821–832. [DOI] [PubMed] [Google Scholar]
  • 128. Franchi L, Munoz-Planillo R, Reimer T, Eigenbrod T, Nunez G. Inflammasomes as microbial sensors. Eur J Immunol. 2010;40(3):611–615. [DOI] [PubMed] [Google Scholar]
  • 129. Bauernfeind F, Ablasser A, Bartok E, et al. Inflammasomes: current understanding and open questions. Cell Mol Life Sci. 2011;68(5):765–783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. 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(5-6):257–276. [DOI] [PubMed] [Google Scholar]
  • 131. Gross O, Thomas CJ, Guarda G, Tschopp J. The inflammasome: an integrated view. Immunol Rev. 2011;243(1):136–151. [DOI] [PubMed] [Google Scholar]
  • 132. Lamkanfi M. Emerging inflammasome effector mechanisms. Nat Rev Immunol. 2011;11(3):213–220. [DOI] [PubMed] [Google Scholar]
  • 133. Lamkanfi M, Dixit VM. Modulation of inflammasome pathways by bacterial and viral pathogens. J Immunol. 2011;187(2):597–602. [DOI] [PubMed] [Google Scholar]
  • 134. Broz P, Monack DM. Molecular mechanisms of inflammasome activation during microbial infections. Immunol Rev. 2011;243(1):174–190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Skeldon A, Saleh M. The inflammasomes: molecular effectors of host resistance against bacterial, viral, parasitic, and fungal infections. Front Microbiol. 2011;2:15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Horvath GL, Schrum JE, De Nardo CM, Latz E. Intracellular sensing of microbes and danger signals by the inflammasomes. Immunol Rev. 2011;243(1):119–135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. 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(3):110–116. [DOI] [PubMed] [Google Scholar]
  • 138. Franchi L, Munoz-Planillo R, Nunez G. Sensing and reacting to microbes through the inflammasomes. Nat Immunol. 2012;13(4):325–332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. Dagenais M, Skeldon A, Saleh M. The inflammasome: in memory of Dr. Jurg Tschopp. Cell Death Differ. 2012;19(1):5–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Ciraci C, Janczy JR, Sutterwala FS, Cassel SL. Control of innate and adaptive immunity by the inflammasome. Microbes Infect. 2012;14(14):1263–1270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Rathinam VA, Vanaja SK, Fitzgerald KA. Regulation of inflammasome signaling. Nat Immunol. 2012;13(4):333–332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Franchi L, Nunez G. Immunology. Orchestrating inflammasomes. Science. 2012;337(6100):1299–1300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Henao-Mejia J, Elinav E, Strowig T, Flavell RA. Inflammasomes: far beyond inflammation. Nat Immunol. 2012;13(4):321–324. [DOI] [PubMed] [Google Scholar]
  • 144. Latz E, Xiao TS, Stutz A. Activation and regulation of the inflammasomes. Nat Rev Immunol. 2013;13(6):397–411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. Bauernfeind F, Hornung V. Of inflammasomes and pathogens – sensing of microbes by the inflammasome. EMBO Mol Med. 2013;5(6):814–826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Vladimer GI, Marty-Roix R, Ghosh S, Weng D, Lien E. Inflammasomes and host defenses against bacterial infections. Curr Opin Microbiol. 2013;16(1):23–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Lamkanfi M, Dixit VM. Mechanisms and functions of inflammasomes. Cell. 2014;157(5):1013–1022. [DOI] [PubMed] [Google Scholar]
  • 148. Ulland TK, Ferguson PJ, Sutterwala FS. Evasion of inflammasome activation by microbial pathogens. J Clin Invest. 2015;125(2):469–477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149. Vanaja SK, Rathinam VA, Fitzgerald KA. Mechanisms of inflammasome activation: recent advances and novel insights. Trends Cell Biol. 2015;25(5):308–315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Guo H, Callaway JB, Ting JP. Inflammasomes: mechanism of action, role in disease, and therapeutics. Nat Med. 2015;21(7):677–687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Agostini L, Martinon F, Burns K, McDermott MF, Hawkins PN, Tschopp J. NALP3 forms an IL-1beta-processing inflammasome with increased activity in Muckle-Wells autoinflammatory disorder. Immunity. 2004;20(3):319–325. [DOI] [PubMed] [Google Scholar]
  • 152. Mariathasan S, Newton K, Monack DM, et al. Differential activation of the inflammasome by caspase-1 adaptors ASC and Ipaf. Nature. 2004;430(6996):213–218. [DOI] [PubMed] [Google Scholar]
  • 153. 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(10):5323–5326. [PubMed] [Google Scholar]
  • 154. Kostura MJ, Tocci MJ, Limjuco G, et al. Identification of a monocyte specific pre-interleukin 1 beta convertase activity. Proc Natl Acad Sci U S A. 1989;86(14):5227–5231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155. Thornberry NA, Bull HG, Calaycay JR, et al. A novel heterodimeric cysteine protease is required for interleukin-1 beta processing in monocytes. Nature. 1992;356(6372):768–774. [DOI] [PubMed] [Google Scholar]
  • 156. Cerretti DP, Kozlosky CJ, Mosley B, et al. Molecular cloning of the interleukin-1 beta converting enzyme. Science. 1992;256(5053):97–100. [DOI] [PubMed] [Google Scholar]
  • 157. Gu Y, Kuida K, Tsutsui H, et al. Activation of interferon-gamma inducing factor mediated by interleukin-1beta converting enzyme. Science. 1997;275(5297):206–209. [DOI] [PubMed] [Google Scholar]
  • 158. Ghayur T, Banerjee S, Hugunin M, et al. Caspase-1 processes IFN-gamma-inducing factor and regulates LPS-induced IFN-gamma production. Nature. 1997;386(6625):619–623. [DOI] [PubMed] [Google Scholar]
  • 159. Dinarello CA. Interleukin-1 beta, interleukin-18, and the interleukin-1 beta converting enzyme. Ann N Y Acad Sci. 1998;856:1–11. [DOI] [PubMed] [Google Scholar]
  • 160. Fantuzzi G, Dinarello CA. Interleukin-18 and interleukin-1 beta: two cytokine substrates for ICE (caspase-1). J Clin Immunol. 1999;19(1):1–11. [DOI] [PubMed] [Google Scholar]
  • 161. Sansonetti PJ, Phalipon A, Arondel J, et al. Caspase-1 activation of IL-1beta and IL-18 are essential for Shigella flexneri-induced inflammation. Immunity. 2000;12(5):581–590. [DOI] [PubMed] [Google Scholar]
  • 162. 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(11):7611–7622. [DOI] [PubMed] [Google Scholar]
  • 163. 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] [PubMed] [Google Scholar]
  • 164. Cookson BT, Brennan MA. Pro-inflammatory programmed cell death. Trends Microbiol. 2001;9(3):113–114. [DOI] [PubMed] [Google Scholar]
  • 165. Miao EA, Rajan JV, Aderem A. Caspase-1-induced pyroptotic cell death. Immunol Rev. 2011;243(1):206–214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166. Shalini S, Dorstyn L, Dawar S, Kumar S. Old, new and emerging functions of caspases. Cell Death Differ. 2015;22(4):526–539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167. Wang S, Miura M, Jung YK, Zhu H, Li E, Yuan J. Murine caspase-11, an ICE-interacting protease, is essential for the activation of ICE. Cell. 1998;92(4):501–509. [DOI] [PubMed] [Google Scholar]
  • 168. Sollberger G, Strittmatter GE, Kistowska M, French LE, Beer HD. Caspase-4 is required for activation of inflammasomes. J Immunol. 2012;188(4):1992–2000. [DOI] [PubMed] [Google Scholar]
  • 169. Kayagaki N, Wong MT, Stowe IB, et al. Noncanonical inflammasome activation by intracellular LPS independent of TLR4. Science. 2013;341(6151):1246–1249. [DOI] [PubMed] [Google Scholar]
  • 170. Hagar JA, Powell DA, Aachoui Y, Ernst RK, Miao EA. Cytoplasmic LPS activates caspase-11: implications in TLR4-independent endotoxic shock. Science. 2013;341(6151):1250–1253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171. Aachoui Y, Leaf IA, Hagar JA, et al. Caspase-11 protects against bacteria that escape the vacuole. Science. 2013;339(6122):975–978. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172. Kayagaki N, Warming S, Lamkanfi M, et al. Non-canonical inflammasome activation targets caspase-11. Nature. 2011;479(7371):117–121. [DOI] [PubMed] [Google Scholar]
  • 173. Pacora P, Romero R, Maymon E, et al. Participation of the novel cytokine interleukin 18 in the host response to intra-amniotic infection. Am J Obstet Gynecol. 2000;183(5):1138–1143. [DOI] [PubMed] [Google Scholar]
  • 174. Hoang M, Potter JA, Gysler SM, et al. Human fetal membranes generate distinct cytokine profiles in response to bacterial Toll-like receptor and nod-like receptor agonists. Biol Reprod. 2014;90(2):39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175. Martinon F, Tschopp J. Inflammatory caspases: linking an intracellular innate immune system to autoinflammatory diseases. Cell. 2004;117(5):561–574. [DOI] [PubMed] [Google Scholar]
  • 176. Martinon F, Tschopp J. Inflammatory caspases and inflammasomes: master switches of inflammation. Cell Death Differ. 2007;14(1):10–22. [DOI] [PubMed] [Google Scholar]
  • 177. Case CL, Kohler LJ, Lima JB, et al. Caspase-11 stimulates rapid flagellin-independent pyroptosis in response to Legionella pneumophila. Proc Natl Acad Sci U S A. 2013;110(5):1851–1856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178. Broz P, Monack DM. Noncanonical inflammasomes: caspase-11 activation and effector mechanisms. PLoS Pathog. 2013;9(2):e1003144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179. Knodler LA, Crowley SM, Sham HP, et al. Noncanonical inflammasome activation of caspase-4/caspase-11 mediates epithelial defenses against enteric bacterial pathogens. Cell Host Microbe. 2014;16(2):249–256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180. Casson CN, Yu J, Reyes VM, et al. Human caspase-4 mediates noncanonical inflammasome activation against gram-negative bacterial pathogens. Proc Natl Acad Sci U S A. 2015;112(21):6688–6693. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181. Yang D, He Y, Munoz-Planillo R, Liu Q, Nunez G. Caspase-11 requires the pannexin-1 channel and the purinergic P2X7 pore to mediate pyroptosis and endotoxic shock. Immunity. 2015;43(5):923–932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182. Sutterwala FS, Haasken S, Cassel SL. Mechanism of NLRP3 inflammasome activation. Ann N Y Acad Sci. 2014;1319:82–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183. Sutterwala FS, Ogura Y, Zamboni DS, Roy CR, Flavell RA. NALP3: a key player in caspase-1 activation. J Endotoxin Res. 2006;12(4):251–256. [DOI] [PubMed] [Google Scholar]
  • 184. Chae JJ, Cho YH, Lee GS, et al. Gain-of-function pyrin mutations induce NLRP3 protein-independent interleukin-1beta activation and severe autoinflammation in mice. Immunity. 2011;34(5):755–768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185. 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(5876):674–677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186. Cassel SL, Eisenbarth SC, Iyer SS, et al. The Nalp3 inflammasome is essential for the development of silicosis. Proc Natl Acad Sci U S A. 2008;105(26):9035–9040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187. Hornung V, Bauernfeind F, Halle A, et al. Silica crystals and aluminum salts activate the NALP3 inflammasome through phagosomal destabilization. Nat Immunol. 2008;9(8):847–856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188. Yamasaki K, Muto J, Taylor KR, et al. 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(19):12762–12771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189. Cassel SL, Joly S, Sutterwala FS. The NLRP3 inflammasome: a sensor of immune danger signals. Semin Immunol. 2009;21(4):194–198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190. Cassel SL, Sutterwala FS. Sterile inflammatory responses mediated by the NLRP3 inflammasome. Eur J Immunol. 2010;40(3):607–611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191. Leemans JC, Cassel SL, Sutterwala FS. Sensing damage by the NLRP3 inflammasome. Immunol Rev. 2011;243(1):152–162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192. Martinon F, Petrilli V, Mayor A, Tardivel A, Tschopp J. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature. 2006;440(7081):237–241. [DOI] [PubMed] [Google Scholar]
  • 193. Mariathasan S, Weiss DS, Newton K, et al. Cryopyrin activates the inflammasome in response to toxins and ATP. Nature. 2006;440(7081):228–232. [DOI] [PubMed] [Google Scholar]
  • 194. Kool M, Petrilli V, De Smedt T, et al. Cutting edge: alum adjuvant stimulates inflammatory dendritic cells through activation of the NALP3 inflammasome. J Immunol. 2008;181(6):3755–3759. [DOI] [PubMed] [Google Scholar]
  • 195. Li H, Willingham SB, Ting JP, Re F. Cutting edge: inflammasome activation by alum and alum’s adjuvant effect are mediated by NLRP3. J Immunol. 2008;181(1):17–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196. Eisenbarth SC, Colegio OR, O’Connor W, Sutterwala FS, Flavell RA. Crucial role for the Nalp3 inflammasome in the immunostimulatory properties of aluminium adjuvants. Nature. 2008;453(7198):1122–1126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197. Franchi L, Nunez G. The NLRP3 inflammasome is critical for aluminium hydroxide-mediated IL-1beta secretion but dispensable for adjuvant activity. Eur J Immunol. 2008;38(8):2085–2089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198. Demento SL, Eisenbarth SC, Foellmer HG, et al. Inflammasome-activating nanoparticles as modular systems for optimizing vaccine efficacy. Vaccine. 2009;27(23):3013–3021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199. Nakahira K, Haspel JA, Rathinam VA, et al. Autophagy proteins regulate innate immune responses by inhibiting the release of mitochondrial DNA mediated by the NALP3 inflammasome. Nat Immunol. 2011;12(3):222–230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200. Iyer SS, He Q, Janczy JR, et al. Mitochondrial cardiolipin is required for NLRP3 inflammasome activation. Immunity. 2013;39(2):311–323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201. O’Neill LA. Cardiolipin and the NLRP3 inflammasome. Cell Metab. 2013;18(5):610–612. [DOI] [PubMed] [Google Scholar]
  • 202. Gurcel L, Abrami L, Girardin S, Tschopp J, van der Goot FG. Caspase-1 activation of lipid metabolic pathways in response to bacterial pore-forming toxins promotes cell survival. Cell. 2006;126(6):1135–1145. [DOI] [PubMed] [Google Scholar]
  • 203. Munoz-Planillo R, Franchi L, Miller LS, Nunez G. A critical role for hemolysins and bacterial lipoproteins in Staphylococcus aureus-induced activation of the NLRP3 inflammasome. J Immunol. 2009;183(6):3942–3948. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204. Kanneganti TD, Body-Malapel M, Amer A, et al. Critical role for cryopyrin/Nalp3 in activation of caspase-1 in response to viral infection and double-stranded RNA. J Biol Chem. 2006;281(48):36560–36568. [DOI] [PubMed] [Google Scholar]
  • 205. Koo IC, Wang C, Raghavan S, Morisaki JH, Cox JS, Brown EJ. ESX-1-dependent cytolysis in lysosome secretion and inflammasome activation during mycobacterial infection. Cell Microbiol. 2008;10(9):1866–1878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206. Muruve DA, Petrilli V, Zaiss AK, et al. The inflammasome recognizes cytosolic microbial and host DNA and triggers an innate immune response. Nature. 2008;452(7183):103–107. [DOI] [PubMed] [Google Scholar]
  • 207. Thomas PG, Dash P, Aldridge JR, Jr, et al. The intracellular sensor NLRP3 mediates key innate and healing responses to influenza A virus via the regulation of caspase-1. Immunity. 2009;30(4):566–575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208. Allen IC, Scull MA, Moore CB, et al. The NLRP3 inflammasome mediates in vivo innate immunity to influenza A virus through recognition of viral RNA. Immunity. 2009;30(4):556–565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209. Duncan JA, Gao X, Huang MT, et al. Neisseria gonorrhoeae activates the proteinase cathepsin B to mediate the signaling activities of the NLRP3 and ASC-containing inflammasome. J Immunol. 2009;182(10):6460–6469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210. Joly S, Ma N, Sadler JJ, Soll DR, Cassel SL, Sutterwala FS. Cutting edge: Candida albicans hyphae formation triggers activation of the NLRP3 inflammasome. J Immunol. 2009;183(6):3578–3581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211. Ichinohe T, Lee HK, Ogura Y, Flavell R, Iwasaki A. Inflammasome recognition of influenza virus is essential for adaptive immune responses. J Exp Med. 2009;206(1):79–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212. Menu P, Vince JE. The NLRP3 inflammasome in health and disease: the good, the bad and the ugly. Clin Exp Immunol. 2011;166(1):1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213. Rathinam VA, Vanaja SK, Waggoner L, et al. TRIF licenses caspase-11-dependent NLRP3 inflammasome activation by gram-negative bacteria. Cell. 2012;150(3):606–619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214. Clay GM, Sutterwala FS, Wilson ME. NLR proteins and parasitic disease. Immunol Res. 2014;59(1-3):142–152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215. Bauernfeind FG, Horvath G, Stutz A, et al. Cutting edge: NF-kappaB activating pattern recognition and cytokine receptors license NLRP3 inflammasome activation by regulating NLRP3 expression. J Immunol. 2009;183(2):787–791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216. Franchi L, Eigenbrod T, Nunez G. Cutting edge: TNF-alpha mediates sensitization to ATP and silica via the NLRP3 inflammasome in the absence of microbial stimulation. J Immunol. 2009;183(2):792–796. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217. Fernandes-Alnemri T, Wu J, Yu JW, et al. The pyroptosome: a supramolecular assembly of ASC dimers mediating inflammatory cell death via caspase-1 activation. Cell Death Differ. 2007;14(9):1590–1604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218. Jaiswal MK, Agrawal V, Mallers T, Gilman-Sachs A, Hirsch E, Beaman KD. Regulation of apoptosis and innate immune stimuli in inflammation-induced preterm labor. J Immunol. 2013;191(11):5702–5713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219. Broz P, Ruby T, Belhocine K, et al. Caspase-11 increases susceptibility to Salmonella infection in the absence of caspase-1. Nature. 2012;490(7419):288–291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220. 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]
  • 221. Gomez-Lopez N, Romero R, Plazyo O, et al. Intra-amniotic administration of HMGB1 induces spontaneous preterm labor and birth. Am J Reprod Immunol. 2016;75(1):3–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222. Wilson KP, Black JA, Thomson JA, et al. Structure and mechanism of interleukin-1 beta converting enzyme. Nature. 1994;370(6487):270–275. [DOI] [PubMed] [Google Scholar]
  • 223. Dinarello CA. Interleukin-1. Adv Pharmacol. 1994;25:21–51. [DOI] [PubMed] [Google Scholar]
  • 224. Dinarello CA. The biological properties of interleukin-1. Eur Cytokine Netw. 1994;5(6):517–531. [PubMed] [Google Scholar]
  • 225. Dinarello CA. The interleukin-1 family: 10 years of discovery. FASEB J. 1994;8(15):1314–1325. [PubMed] [Google Scholar]
  • 226. Dinarello CA. Interleukin-1. Cytokine Growth Factor Rev. 1997;8(4):253–265. [DOI] [PubMed] [Google Scholar]
  • 227. Dinarello CA. Immunological and inflammatory functions of the interleukin-1 family. Annu Rev Immunol. 2009;27:519–550. [DOI] [PubMed] [Google Scholar]
  • 228. Creagh EM. Caspase crosstalk: integration of apoptotic and innate immune signalling pathways. Trends Immunol. 2014;35(12):631–640. [DOI] [PubMed] [Google Scholar]
  • 229. Romero R, Durum S, Dinarello CA, Oyarzun E, Hobbins JC, Mitchell MD. Interleukin-1 stimulates prostaglandin biosynthesis by human amnion. Prostaglandins. 1989;37(1):13–22. [DOI] [PubMed] [Google Scholar]
  • 230. Hertelendy F, Romero R, Molnar M, Todd H, Baldassare JJ. Cytokine-initiated signal transduction in human myometrial cells. Am J Reprod Immunol. 1993;30(2-3):49–57. [DOI] [PubMed] [Google Scholar]
  • 231. 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(3):142–147. [DOI] [PubMed] [Google Scholar]
  • 232. 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(2):359–366. [DOI] [PubMed] [Google Scholar]
  • 233. 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(6):1755–1762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234. 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]
  • 235. Stock O, Gordon L, Kapoor J, et al. Chorioamnionitis occurring in women with preterm rupture of the fetal membranes is associated with a dynamic increase in mRNAs coding cytokines in the maternal circulation. Reprod Sci. 2015;22(7):852–859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236. Romero R, Mazor M, Tartakovsky B. Systemic administration of interleukin-1 induces preterm parturition in mice. Am J Obstet Gynecol. 1991;165(4 pt 1):969–971. [DOI] [PubMed] [Google Scholar]
  • 237. Romero R, Sepulveda W, Mazor M, et al. The natural interleukin-1 receptor antagonist in term and preterm parturition. Am J Obstet Gynecol. 1992;167(4 pt 1):863–872. [DOI] [PubMed] [Google Scholar]
  • 238. 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]
  • 239. 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]
  • 240. 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(3):121–126. [DOI] [PubMed] [Google Scholar]
  • 241. Vadillo-Ortega F, Sadowsky DW, Haluska GJ, et al. 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(1):128–138. [DOI] [PubMed] [Google Scholar]
  • 242. 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(6):1578–1589. [DOI] [PubMed] [Google Scholar]
  • 243. Aagaard K, Ganu R, Ma J, et al. 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(1):S218. [Google Scholar]
  • 244. Prince A, Ma J, Miller L, et al. 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(1):S153. [Google Scholar]
  • 245. Presicce P, Senthamaraikannan P, Alvarez M, et al. Neutrophil recruitment and activation in decidua with intra-amniotic IL-1beta in the preterm rhesus macaque. Biol Reprod. 2015;92(2):56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246. Jacobsson B, Holst RM, Mattsby-Baltzer I, Nikolaitchouk N, Wennerholm UB, Hagberg H. Interleukin-18 in cervical mucus and amniotic fluid: relationship to microbial invasion of the amniotic fluid, intra-amniotic inflammation and preterm delivery. BJOG. 2003;110(6):598–603. [PubMed] [Google Scholar]
  • 247. Okamura H, Tsutsi H, Komatsu T, et al. Cloning of a new cytokine that induces IFN-gamma production by T cells. Nature. 1995;378(6552):88–91. [DOI] [PubMed] [Google Scholar]
  • 248. Ushio S, Namba M, Okura T, et al. Cloning of the cDNA for human IFN-gamma-inducing factor, expression in Escherichia coli, and studies on the biologic activities of the protein. J Immunol. 1996;156(11):4274–4279. [PubMed] [Google Scholar]
  • 249. Takeda K, Tsutsui H, Yoshimoto T, et al. Defective NK cell activity and Th1 response in IL-18-deficient mice. Immunity. 1998;8(3):383–390. [DOI] [PubMed] [Google Scholar]
  • 250. Dinarello CA, Novick D, Puren AJ, et al. Overview of interleukin-18: more than an interferon-gamma inducing factor. J Leukoc Biol. 1998;63(6):658–664. [PubMed] [Google Scholar]
  • 251. Dinarello CA. IL-18: A TH1-inducing, proinflammatory cytokine and new member of the IL-1 family. J Allergy Clin Immunol. 1999;103(1 pt 1):11–24. [DOI] [PubMed] [Google Scholar]
  • 252. Dinarello CA. Interleukin-18. Methods. 1999;19(1):121–132. [DOI] [PubMed] [Google Scholar]
  • 253. Novick D, Kim S, Kaplanski G, Dinarello CA. Interleukin-18, more than a Th1 cytokine. Semin Immunol. 2013;25(6):439–448. [DOI] [PubMed] [Google Scholar]
  • 254. Dinarello CA, Novick D, Kim S, Kaplanski G. Interleukin-18 and IL-18 Binding Protein. Front Immunol. 2013;4:289. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementary material
Supplementary material
Supplementary_tables.pdf (32.6KB, pdf)
Supplementary material
Download video file (30.4MB, avi)

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

RESOURCES