Abstract
Objectives
Inflammasomes are signaling platforms that upon sensing pathogens and sterile stressors mediate the release of mature forms of interleukin (IL)-1β and IL-18. The aims of this study were to determine: 1) the expression of major inflammasome components in the chorioamniotic membranes in spontaneous labor at term; 2) whether there are changes in the inflammasome components associated with activation of caspase-1 and caspase-4; and 3) whether these events are associated with the release of the mature forms of IL-1β and IL-18.
Methods
Chorioamniotic membranes were collected from women at term with and without spontaneous labor. mRNA abundance and protein concentrations of inflammasome components, nucleotide-binding oligomerization domain-containing (NOD)–1 and NOD–2 proteins, caspase-1 and caspase-4, IL-1β, and IL-18 were quantified by qRT-PCR (n=28-29 each), ELISA (n=10 each) or immunoblotting (n=8 each), and immunohistochemistry (n=10 each). Active caspase-1 and caspase-4, as well as mature IL-18, were determined by immunoblotting (n=4 each), and pro- and mature forms of IL-1β were determined by ELISA (n=4-7 each).
Results
Inflammasome components and NOD proteins were expressed in the chorioamniotic membranes obtained from women at term. The chorioamniotic membranes from women who had undergone labor had: 1) higher NLRP3 (NOD-like receptor family, pyrin domain–containing protein 3) and NOD1 protein concentrations; 2) greater immunoreactivity for caspase-1 and caspase-4; 3) higher quantity of the active form of caspase 1 (p20); and 4) higher mRNA abundance and protein concentrations of pro- and mature IL-1β. However, mRNA abundance and protein concentrations of the mature form of IL-18 were not increased in tissues from women who underwent labor at term.
Conclusions
Spontaneous labor at term is characterized by the expression of inflammasome components, which may participate in the activation of caspase-1 leading to the consequent cleavage and release of mature IL-1β by the chorioamniotic membranes. These results support the participation of the inflammasome in the mechanisms responsible for spontaneous parturition at term.
Keywords: amnion, caspase-1, caspase-4, chorion, cytokine, labor, IL-1β, IL-18, NOD1, NOD2, normal pregnancy, parturition, sterile inflammation, preterm labor, biomarker, chorioamniotic membranes, NLRP1, NLRP3, NLRC4, AIM2
INTRODUCTION
Spontaneous term labor is a state of physiologic sterile inflammation.1-6 The evidence in support of this concept includes the increased bioavailability of cytokines7-21 and chemokines22-27 in the amniotic fluid, maternal circulation,28-31 and reproductive tissues.20, 32-46 Moreover, parturition is accompanied by an influx of inflammatory cells, e.g., neutrophils and macrophages, into the cervix,39, 47-56 myometrium,49, 57-63 and chorioamniotic membranes.35, 64-67 T cells are also present in the chorioamniotic membranes.66, 68 The inflammatory response associated with normal spontaneous labor is considered sterile since intra-amniotic infection is absent in most women.1, 2, 69-72
The mechanisms responsible for sterile inflammation in parturition have not been elucidated, but are thought to involve inflammasomes,73-75 which are high-molecular-weight multi-subunit protein complexes found in the cytoplasm capable of inducing an inflammatory response through the production of interleukin (IL)-1β and IL-18.76-110 Their basic structure consists of–: 1) an inflammasome sensor molecule, 2) the adaptor protein ASC (an apoptosis-associated speck-like protein), and 3) pro-caspase-1(pro-CASP-1).76-110 Once activated, the inflammasome complex induces auto-catalytic cleavage of pro-CASP-1 into its mature/active form. Caspase-1 (CASP-1) can then cleave pro-IL-1β and pro-IL-18, and the newly described pro-IL-33, into the mature, secreted forms of the cytokines.111-124 In addition, CASP-1 is required for a specific type of programmed cell death induced by inflammation: pyroptosis.125-127 Recently, it was demonstrated that CASP-4 expression is required for activation of CASP-1 in ultraviolet B-irradiated keratinocytes and activated macrophages,128 suggesting that caspase-4 acts upstream of caspase-1 and the inflammasome.129
Several inflammasomes have been identified and named after their respective pattern recognition receptors (PRRs):130 NLR family pyrin domain (NLRP)1,76 NLRP3,131 NLR family caspase activation and recruitment domain (CARD) (NLRC)4 (also known as IPAF),132, 133 interferon gamma-inducible protein 16 (IFI16),134 or absent in melanoma (AIM2).135-143 Inflammasome specificity depends on which ligand(s) the PRR recognizes, and once PRR-ligand binding occurs, the inflammasomes oligomerize with other components of the multi-protein complexes and become activated.86, 94, 104, 110, 144, 145 Two additional PRRs belonging to the NLR family – the nucleotide-binding oligomerization domain-containing proteins 1 and 2 (NOD1 and NOD2) – recognize bacterial peptidoglycan segments, but do not recruit inflammasome components.90, 146-153 Instead, the NODs directly activate nuclear factor kappa B (NF-κB) proinflammatory signaling, which can induce the expression of pro-IL-1β76, 87, 131, 146, 154-158 and pro-IL-18.115 The functional combination of NOD proteins and inflammasome components (e.g., NOD2 and NLRP3) improves immune responses in murine dendritic cells.159 Moreover, inflammasome activation is associated with an increased production of eicosanoids (prostaglandins and leukotrienes), which leads to further inflammation.160
We proposed that the inflammasome participates in labor at term73, 74 and in pregnancy complications.161-171 Indeed, we demonstrated that CASP-1, the predominant inflammasome-activated caspase,82, 172 is present in the amniotic fluid and that its concentration increases as a function of gestational age.75 In addition, we found that amniotic fluid CASP-1 concentration is higher in women in spontaneous labor at term than in those without labor.75 This is mirrored by increased amniotic fluid IL-1β bioactivity and immunoreactivity in women in spontaneous labor at term.8, 9, 36 IL-18 concentration in the amniotic fluid is also higher in term pregnancies than in the second trimester.173 Collectively, this evidence supports the hypothesis that inflammasomes are involved in the physiologic sterile inflammatory process associated with spontaneous labor at term. The aims of this study were to determine whether: 1) inflammasomes are expressed in the chorioamniotic membranes from women who underwent spontaneous labor at term; 2) changes in inflammasome components are associated with activation of CASP-1 and CASP-4; and 3) these events are associated with the release of the mature forms of IL-1β and IL-18.
MATERIALS AND METHODS
Human subjects, clinical specimens, and definitions
A case control study was conducted including patients who delivered at term without labor (TNL) or at term after labor (TIL). Chorioamniotic membrane samples were collected from the Bank of Biological Specimens of the Perinatology Research Branch, NICHD/NIH/DHHS, Wayne State University, and The Detroit Medical Center (Detroit, MI, USA). The Institutional Review Boards of these institutions approved the collection and use of biological materials for research purposes. All participating women provided written informed consent, and samples were collected within 30 minutes after delivery. Demographic and clinical characteristics of these study groups are represented in Table I. Patients with multiple births or with neonates having congenital or chromosomal abnormalities were excluded. Labor was defined by the presence of regular uterine contractions at a frequency of at least two contractions every 10 minutes with cervical changes resulting in delivery.174 In each case, tissue sections of the chorioamniotic membranes were evaluated for acute histologic chorioamnionitis, according to published criteria,175, 176 by pathologists who had been blinded to the clinical outcome. Samples collected from women with acute histologic chorioamnionitis were excluded from this study.
Table I.
Demographic and clinical characteristics of the study populations
| TNL (n=29) | TIL (n=28) | P value | |
|---|---|---|---|
|
| |||
| Maternal age (years)* | 24.0 (19-35.0) | 21.5 (16.0-31.0) | 0.01 |
|
| |||
| Race** | NS | ||
| African-American | 25 (89.3%) | 24 (88.9%) | |
| Caucasian | 1 (3.6%) | 2 (7.4%) | |
| Hispanic | 1 (3.6%) | 0 (0.0%) | |
| Other | 1 (3.6%) | 1 (3.7%) | |
|
| |||
| Maternal weight (kg)* | 86 (44-136) | 66 (49-109) | 0.0002 |
|
| |||
| Body mass index (kg/m²)* | 33.9 (19.80-48.40) | 24.5 (18.30-41.20) | 0.001 |
|
| |||
| Gestational age at delivery (weeks)* | 39.4 (37.1-40.9) | 39.7 (37.1-41.7) | NS |
|
| |||
| Birth weight (grams)* | 3385 (2960-4010) | 3270 (2750-3870) | NS |
|
| |||
| Cesarean section** | 100% | 32.1% | 0.01 |
|
| |||
| Acute chorioamnionitis | 0% | 0% | NA |
Mann-Whitney test
Chi-square test
RNA isolation, cDNA generation, and qRT-PCR analysis
TRIzol® (Invitrogen™, Life Technologies Corporation, Grand Island, NY, USA) and Qiagen RNeasy® Kits (Qiagen, Gaithersburg, MD, USA) were used to extract total RNA from snap-frozen chorioamniotic membrane tissues (TNL, n=29, and TIL, n=28). RNA purity and concentration were assessed with the NanoDrop® 1000 spectrophotometer (Thermo Scientific, Wilmington, DE, USA), and RNA integrity was evaluated with the Bioanalyzer 2100 (Agilent Technologies, Wilmington, DE, USA). The SuperScript® III First-Strand Synthesis System (Invitrogen) and oligo(dT)20 primers (Invitrogen) were utilized to generate cDNA. Gene expression profiling was performed on the BioMark™ System for high-throughput qRT-PCR (Fluidigm, San Francisco, CA, USA) and on the ABI 7500 FAST Real-Time PCR System (Applied Biosystems®, Life Technologies Corporation, Foster City, CA, USA) with TaqMan® gene expression assays (Applied Biosystems) listed in Table II.
Table II.
TaqMan® assays used for gene expression profiling
| Gene Symbol | Protein Name | Assay ID |
|---|---|---|
| ACTB | Actin beta | Hs99999903_m1 |
| GAPDH | Glyceraldehyde-3-phosphate dehydrogenase | Hs99999905_m1 |
| RPLP0 | Ribosomal protein, large, P0 | Hs99999902_m1 |
| NLRP1 | NACHT, LRR and PYD domains-containing protein 1 | Hs00248187_m1 |
| NLRP3 | NACHT, LRR and PYD domains-containing protein 3 | Hs00918082_m1 |
| NLRC4 | NLR family CARD domain-containing protein 4 | Hs00368367_m1 |
| NOD1 | Nucleotide-binding oligomerization domain-containing protein 1 | Hs00196075_m1 |
| NOD2 | Nucleotide-binding oligomerization domain-containing protein 2 | Hs00223394_m1 |
| AIM2 | Absent in melanoma 2 | Hs00915710_m1 |
| CASP1 | Caspase-1 / Interleukin-1 converting enzyme | Hs00354836_m1 |
| CASP4 | Caspase-4 | Hs01031947_m1 |
| IL1B | Interleukin-1 beta | Hs00174097_m1 |
| IL18 | Interleukin-18 | Hs01038788_m1 |
Chorioamniotic membrane tissue lysates
Fragments of snap-frozen chorioamniotic membranes (TNL and TIL; n=10 each) were homogenized using a mechanical tissue homogenizer (T-25 Ultra-Turrax®, IKA® Works, Inc., Wilmington, NC, USA) in 2ml of 1X PBS containing a complete protease inhibitor cocktail (Cat. No. 11697498001; Roche Applied Science, Mannheim, Germany). Tissue lysates were centrifuged at 15700 × g for 5 min at 4°C, and the supernatant was collected and stored at -80°C. The protein concentration of the lysates was determined using the Quick Start™ Bradford Protein Assay Kit (Bio-Rad, Hercules, CA, USA). Triplicate cell lysates were obtained from the membranes of each patient.
Chorioamniotic membrane tissue supernatants
Chorioamniotic membrane samples were collected from each group of women (TNL, n=7, and TIL, n=4) and processed the same day. Tissue samples were washed with 1X PBS (Invitrogen) and cut into 2cm × 2cm pieces. These tissue explants were transferred into 6-well tissue culture plates containing 2ml of Dulbecco’s Modified Eagle Medium (DMEM) (Invitrogen) per well supplemented with 10% Fetal Bovine Serum (FBS) (Invitrogen) and 1% Penicillin/Streptomycin (P/S) (Invitrogen). Tissue samples were placed in a humidified 5% CO2 incubator at 37°C overnight, and then tissue culture supernatants were collected and stored at -80°C. Triplicate supernatants were obtained from the membranes of each patient.
Enzyme-linked immunosorbent assays
The concentrations of NLRP1, NLRP3, AIM2, NOD2, CASP-1, CASP-4, IL18, pro-IL-1β, and IL-1β were measured in the chorioamniotic membrane tissue lysates or supernatants using specific and sensitive 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, TX, USA; pro-IL-1β and IL-1β ELISA kits from R&D Systems, Minneapolis, MN, USA; IL-18 ELISA kits from MBL International Corporation, Woburn, MA, USA), following the manufacturers’ instructions. Briefly, recombinant human standards and the samples were incubated in duplicate wells of the 96-well microplates pre-coated with monoclonal antibodies specific for target analytes. During incubation, immobilized antibodies in the microplates bound to the target proteins present in the standard and sample groups. After washing the unbound substances, enzyme-conjugated antibodies bound to the target analytes were added to the wells. After the incubation, assay plates were washed to remove the unbound antibodies, followed by the addition of a substrate solution that developed color proportional to the amount of target protein bound in the initial step. Finally, the color development was stopped by the addition of a sulfuric acid solution, and the microplates were read using a programmable spectrophotometer (SpectraMax M5 Multi-Mode Microplate Reader, Molecular Devices, Sunnyvale, CA, USA). The sensitivities of the assays were <4.68 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 measures approximately 10% of the pro-IL-1β. The immunoassays for NLRC4 and NOD1 ELISA did not meet our criteria for validation; instead, immunoblotting was performed.
Immunohistochemistry
Samples of chorioamniotic membranes collected from each study group (TNL and TIL; n=10 each) were included. Five-μm-thick sections of formalin-fixed, paraffin-embedded chorioamniotic membrane tissues were placed on silanized slides. Immunostainings for NLRP1, NLRP3, NLRC4, AIM2, NOD1, NOD2, CASP-1, CASP-4, IL-1β, and IL-18 were performed using a Leica Bond Max automatic staining system (Leica Microsystems, Wetzlar, Germany), and the Bond™ Polymer Refine Detection Kit (Leica Microsystems) was used to detect the chromogenic reaction of horseradish peroxidase. Primary antibodies and a description of the immunostaining conditions are presented in Table III. Mouse IgG (Invitrogen) and rabbit IgG (Invitrogen) were used as negative controls. A PerkinElmer Pannoramic MIDI slide scanner (PerkinElmer, Waltham, MA, USA) was used to assess the intensity of staining (a semi-quantitative method of analysis).
Table III.
Primary antibodies and immunostaining conditions
| Primary antibody | Vendor | Host species /clonality | Dilution | Incubation time |
|---|---|---|---|---|
| Anti-AIM2 | Santa Cruz Biotechnology (Dallas, TX, USA) | Rabbit / polyclonal | (1:100) | 15 min |
| Anti-CARD7 (NLRP1) | Acris Antibodies (Herford, Germany) | Rabbit / polyclonal | (1:500) | 15 min |
| Anti-CARD12 (NLRC4) | Abcam Inc. (Cambridge, MA, USA) | Rabbit / polyclonal | (1:1500) | 15 min |
| Anti-CARD15 (NOD2) | Abcam Inc. (Cambridge, MA, USA) | Rabbit / polyclonal | (1:200) | 15 min |
| Anti-CASP1 | R&D Systems, Inc. (Minneapolis, MN, USA) | Mouse / monoclonal | (1:2000) | 15 min |
| Anti-CASP4 | Abcam Inc. (Cambridge, MA, USA) | Rabbit / polyclonal | (1:100) | 15 min |
| Anti-CIAS/NALP3 (NLRP3) | Millipore Corporation (Temecula, CA, USA) | Rabbit / polyclonal | (1:100) | 15 min |
| Anti-NOD1 | Lifespan Bio (Seattle, WA, USA) | Rabbit / polyclonal | (1:200) | 15 min |
| IL-• | Novus Biologicals (Littleton, CO, USA) | Rabbit / polyclonal | (1:50) | 15 min |
| IL-18 | Lifespan Bio (Seattle, WA, USA) | Rabbit / polyclonal | (1:10) | 15 min |
Chorioamniotic membrane tissue extracts
Chorioamniotic membrane samples from the two study groups (TNL and TIL; n=4 each) were collected and processed on the same day. Ten or 12 tissue explants were obtained from each membrane using a dermatological punch (12mm Acu-Punch, Acuderm Inc., Fort Lauderdale, FL, USA). Tissue explants were placed at 37°C in a humidified 5% CO2 incubator for 24h in 500μL of DMEM (4.5 g/L glucose, L-glutamine, sodium pyruvate, and 1% antibiotics; Gibco®, Life Technologies) in a 24-well plate. Following incubation, tissue explants were homogenized in their conditioned medium using a mechanical tissue homogenizer (T-25 Ultra-Turrax, IKA Works, Inc.). Tissue extracts were centrifuged at 14,000 g for 3-5 min at 4°C, and the supernatant was collected and filtered using a syringe filter (Millex-GV Syringe Filter Unit, 0.22μm, PVDF, 33mm, gamma-sterilized, EMD Millipore, Billerica, MA, USA). Tissue extracts were stored at -80°C until use.
Immunoblotting
Chorioamniotic membrane tissue extracts (40μg for the caspases and 100μg for IL-18 per well) or tissue lysates (20μg for NOD1 and NLRC4 per well) were subjected to 4-12% SDS-polyacrylamide gel electrophoresis (Invitrogen). After electrophoresis, separated proteins were transferred onto nitrocellulose membranes (Bio-Rad), and the membranes were blocked with 5% non-fat dry milk in Tris-buffered saline containing 0.1% Tween-20 (BioRad) and probed overnight at 4°C with specific human antibodies (mouse anti-CASP-1 monoclonal antibody [R&D Systems], rabbit anti-CASP-4 polyclonal antibody [Abcam, Cambridge, MA, USA], rabbit anti-IL-18 polyclonal antibody [Santa Cruz Biotechnology, Dallas, TX, USA], rabbit anti-NOD1 polyclonal antibody [Enzo Life Sciences, Farmingdale, NY, USA],165 or mouse anti-NLRC4 (IPAF) antibody [BioLegend, San Diego, CA, USA]). Nitrocellulose membranes were then stripped with Restore™ Plus Western Blot Stripping Buffer (Pierce Biotechnology, Thermo Fisher Scientific Inc., Rockford, IL, USA) for 15 min, washed with PBS, blocked, and probed for 1h at room temperature with a mouse anti-GAPDH monoclonal antibody (Santa Cruz Biotechnology) or a mouse anti-ACTB monoclonal antibody (Sigma-Aldrich Co., Saint Louis, MO, USA). A horseradish peroxidase-conjugated anti-mouse or anti-rabbit IgG (Cell Signaling, Boston, MA, USA) was used as a secondary antibody. Signals were detected by chemiluminescence with ChemiGlow West Reagents (Protein Simple, Santa Clara, CA, USA). Images were acquired using the FUJIFILM LAS-4000 Imaging System (FUJIFILM North America Corporation, Valhalla, NY, USA).
Statistical analyses
Demographic and clinical data were analyzed using SPSS v.19.0 (SPSS Inc., Chicago, IL, USA). Comparisons among the groups were performed using the Chi-square and Fisher’s exact tests for proportions, as well as the Mann-Whitney U-test for non-normally distributed continuous variables. All other data were analyzed in the R statistical language and environment (www.r-project.org). The qRT-PCR dataset, gene expressions relative to ACTB/GAPDH/RPLP0 were calculated, and the fold-changes between the groups were estimated using a linear model in which the -ΔCt value of a gene (surrogate for log2 gene expression) was the dependent variable and the group was the independent variable. ELISA, IHC, and immunoblotting data were analyzed using linear models in which the protein concentration or intensity was the dependent variable. A fold-change of expression >1.5 and a P value of <0.05 were regarded as statistically significant.
RESULTS
NLRP3 and NOD2 concentrations increase in the chorioamniotic membranes in spontaneous labor at term
We first investigated whether major inflammasome components (NLRP1, NLRP3, NLRC4, and AIM2), and NOD proteins (NOD1 and NOD2) were expressed by the chorioamniotic membranes from women who had undergone spontaneous labor at term. All genes encoding for these proteins were expressed at a detectable level in the chorioamniotic membranes from women with or without spontaneous labor at term. However, no differences were observed in the expression of the evaluated genes, except NOD1, between these two groups of women (Figure 1A, Figures 2A and 2D).
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 at term with (TIL, n=28) or without labor (TNL, n=29). Relative gene expressions are presented as -ΔCt values. (B) Protein concentrations of inflammasome components and NOD2 in chorioamniotic membrane tissue lysates (n=10 each). (C) Representative immunostainings for inflammasome components and NOD2 in the chorioamniotic membranes (n=10 each), 200× magnification.
Figure 2. NOD1 and NLRC4 in the chorioamniotic membranes.

(A and D) mRNA abundance of NOD1 and NLRC4 in the chorioamniotic membranes from women at term with (TIL, n=28) or without labor (TNL, n=29). Relative gene expressions are presented as -ΔCt values. (B and E) Protein quantity of NOD1 and NLRC4 in chorioamniotic membrane tissue lysates (n=8 each). (C and F) Intensity of the immunostainings for NOD1 and NLRC4 in the chorioamniotic membranes (n=10 each) and representative immunostainings, 200× magnification.
NLRP3 and NOD2 concentrations were higher in the chorioamniotic membrane tissue lysates from women who underwent spontaneous labor at term than in those without labor (Figure 1B). However, no differences between these two groups of women were observed in the protein concentrations of the other evaluated NLRs (Figure 1B, Figures 2B and 2E). Immunostaining on formalin-fixed, paraffin-embedded tissue sections collected from the same samples showed that all these proteins are expressed in the chorioamniotic membranes. In general, most of the proteins showed immunoreactivity in the chorionic trophoblast cells as well as in the decidual stromal cells, while some showed weak immunoreactivity in the amniotic mesodermal and epithelial cells (Figure 1C, Figures 2C and 2F).
Collectively, these results suggest that inflammasomes –may participate in the physiologic sterile inflammatory process of spontaneous parturition at term. Additionally, the constitutive expression of the investigated inflammasome components and NOD proteins in the chorioamniotic membranes suggests that inflammasomes may participate in pathological innate immune activation leading to preterm labor and term labor in the setting of intra-amniotic infection or sterile intra-amniotic inflammation.
Activation of CASP-1 in the chorioamniotic membranes in spontaneous labor at term
Activation of the inflammasome leads to the activation of the inflammatory caspases, i.e., CASP-1 and CASP-4.76, 110, 128, 177, 178 We previously demonstrated that amniotic fluid concentration of CASP-1 is higher in women in spontaneous labor at term than in those wihout labor at term.75 Therefore, we investigated whether the higher NLRP3 and NOD2 concentrations were associated with the activation of CASP-1 and CASP-4 in the chorioamniotic membranes in spontaneous labor at term. Genes encoding for CASP-1 and CASP-4 were expressed at detectable levels in the chorioamniotic membranes from women with or without spontaneous labor at term; however, the mRNA abundance and protein concentration were not significantly different between these two groups (Figures 3A and 3B). Semi-quantitative analysis of immunostaining indicated that the intensity of both CASP-1 and CASP-4 was higher in the chorioamniotic membranes from women who underwent spontaneous labor at term than in those without labor (Figure 3C). We also found that the immunoreactivity of the active form of CASP-1 (p20) was greater in the chorioamniotic membranes from women who underwent spontaneous labor at term than in women who did not undergo labor. The active form of CASP-4 was undetectable by immunoblotting (Figure 3D). These data suggest that spontaneous labor at term involves the participation of inflammasomes which, in turn, could activate CASP-1 in the chorioamniotic membranes.
Figure 3. Inflammatory caspases in the chorioamniotic membranes.

(A) mRNA abundance of CASP-1 and CASP-4 in the chorioamniotic membranes from women at term with (TIL, n=28) or without labor (TNL, n=29). Relative gene expressions are presented as -ΔCt values. (B) Protein concentrations of CASP-1 and CASP-4 in chorioamniotic membrane tissue lysates (n=10 each). (C) Intensity of the immunostainings for CASP-1 and CASP-4 in the chorioamniotic membranes (n=10 each) and representative immunostainings, 200× magnification. (D) Immunoblotting of CASP-1, CASP-4, and GAPDH in the chorioamniotic membranes and their quantifications (n=4 each).
Increased mRNA abundance and protein concentration of IL-1β in the chorioamniotic membranes in spontaneous labor at term
Activated CASP-1 subunits (p10 and p20) are able to convert inactive pro-IL-1β into its bioactive and secreted form.112-114 We previously reported that the IL-1β concentration in amniotic fluid (and IL-1 bioactivity) was higher among women in labor than in those wihout labor.7-9 Therefore, we investigated whether the activation of CASP-1 was associated with the release of mature IL-1β in the chorioamniotic membranes in women who underwent spontaneous labor at term. mRNA abundance of IL1B in the chorioamniotic membranes was higher in women who had undergone spontaneous labor at term than in those from women without labor (Figure 4A). The concentrations of mature IL-1β and its pro-form were also higher in the chorioamniotic membranes from women who underwent labor than in those from women without labor (Figure 4B). IL-1β immunoreactivity appeared to be greater in the chorioamniotic membranes from women with labor than in those without labor (Figure 4C). These data suggest that the active forms of CASP-1 may participate in the release of the mature form of IL-1β by the chorioamniotic membranes in spontaneous labor at term.
Figure 4. IL-1β in the chorioamniotic membranes.

(A) mRNA abundance of IL1β in the chorioamniotic membranes from women at term with (TIL, n=28) or without labor (TNL, n=29). Relative gene expressions are presented as -ΔCt values. (B) Protein concentrations of mature and pro-form IL-1β in chorioamniotic membrane supernatants (TNL, n=7, and TIL, n=4). (C) Intensity of the immunostainings for IL-1β in the chorioamniotic membranes (n=10 each) and representative immunostainings, 200× magnification.
IL-18 expression does not increase in the chorioamniotic membranes in spontaneous labor at term
Activated CASP-1 subunits (p10 and p20) are also able to convert inactive pro-IL-18 into its bioactive and secreted form.115 We previously demonstrated that during term pregnancies the IL-18 amniotic fluid concentration tends to be higher in women who undergo spontaneous labor than in women without labor; yet, this increase was not statistically significant.173 We therefore investigated whether activation of CASP-1 was associated with the release of mature IL-18 in the chorioamniotic membranes of women who underwent spontaneous labor at term. Consistent with our published findings,173 we found that the mRNA abundance and protein concentration of IL-18 in the chorioamniotic membranes were not different between women with and without spontaneous labor at term (Figure 5A-C). Indeed, mRNA expression of IL-18 was lower in the chorioamniotic membranes from women who underwent labor at term than in those without labor (Figure 5A). No differences were observed in the abundance of the mature form of IL-18 in the chorioamniotic membranes between women with and without spontaneous labor at term (Figure 5D). These data suggest that IL-18 does not participate in the physiologic sterile inflammatory process of spontaneous parturition at term.
Figure 5. IL-18 in the chorioamniotic membranes.

(A) mRNA abundance of IL-18 in the chorioamniotic membranes from women at term with (TIL, n=28) or without labor (TNL, n=29). Relative gene expressions are presented as -ΔCt values. (B) Protein concentrations of IL-18 in chorioamniotic membrane tissue lysates (n=10 each). (C) Intensity of the immunostainings for IL-18 in the chorioamniotic membranes (n=10 each) and representative immunostainings, 200× magnification. (D) Immunoblotting of IL-18 and GAPDH in the chorioamniotic membranes (n=4 each).
DISCUSSION
Principal findings of the study
1) Inflammasome components and NOD proteins were expressed in the chorioamniotic membranes from women with normal term pregnancies; 2) NLRP3 and NOD2 protein concentrations were greater in the chorioamniotic membrane tissue lysates from women who underwent labor at term than in those from women who did not undergo labor; 3) the immunoreactivity of CASP-1 and CASP-4 in the chorioamniotic membranes was higher among women in labor at term than in those without labor; 4) the active form of CASP-1 (p20) was higher in the chorioamniotic membranes from women in labor at term than in women without labor; 5) mRNA abundance and protein expression profiles of IL-1β were greater in the chorioamniotic membranes from women in spontaneous labor at term than in those without labor; and 6) mRNA abundance and protein expression profiles of IL-18 did not increase in the chorioamniotic membranes from women who underwent spontaneous labor at term. Collectively, these data support a role for the inflammasome (NLRP1, NLRP3, AIM2 or NLRC4) in the activation of CASP-1 and the consequent release of mature IL-1β by the chorioamniotic membranes of patients who underwent spontaneous labor at term.
The expression of inflammasome components and NOD proteins in the chorioamniotic membranes at term
We first demonstrated that inflammasome components NLRP1, NLRP3, NLRC4, and AIM2, as well as NOD1 and NOD2 proteins, were expressed at the mRNA and protein levels by the chorioamniotic membranes from women at term. This is consistent with previous studies showing that the human placenta constitutively expresses inflammasome components including NOD1, NOD2, NOD3, NOD4, NALP1, NALP2, NALP4, NALP7, NALP10, NALP12, and NAIP, as well as CASP-1, CASP-4, and CASP-5,179 and that the chorioamniotic membranes express NOD1, NOD2, NLRP1, NLRP3, and ASC.165 The expression of inflammasome components and inflammatory caspases in the chorioamniotic membranes may be an important feature of innate immune mechanisms at the maternal-fetal interface, which will ensure the rapid activation of an immune response when exogenous and/or endogenous signal(s) are recognized.
Increased concentrations of NLRP3 and NOD2 in the chorioamniotic membranes in spontaneous labor at term
A role for the inflammasomein the physiological proinflammatory processes of spontaneous labor at term was initially suggested by our group.73, 75 Herein, we provide evidence to support this hypothesis, and demonstrate for the first time that NLRP3 (also known as cryopyrin), the PRR component of the NLRP3 inflammasome, is increased in the chorioamniotic membranes during spontaneous labor at term. Besides cryopyrin, the NLRP3 inflammasome contains the adaptor molecule ASC containing two death-fold domains, one pyrin domain and one CARD, and pro-caspase-1.104, 131, 180, 181 Activation of the NLRP3 inflammasome can be triggered by several stimuli,182-188 chemically and structurally different, including crystalline material,184, 189 extracellular ATP released from dying cells,190 peptide aggregates such as vaccine adjuvant,191-195 phospholipid cardiolipin and mitochondrial DNA,196-198 bacterial toxins 190, 199, 200 [i.e., nigericin (Streptomyces hygroscopicus), listeriolysin O (Listeria monoccytogenes), aerolysin (Aeromonas), β-and-γ hemolysins (Staphylococcus aureus)], DAMPs (damage-associated molecular patterns),186, 187 and PAMPs (pathogen-associated molecular patterns) 80, 201-211. Activation of the NLRP3 inflammasome requires two steps: priming and assembly of the inflammasome complex.212, 213 The priming step is initiated by PRRs, cytokine receptors, or any other factor able to induce activation of NF-κB, which results in the up-regulation of NLRP3 to a functional level and pro-IL-1β expression.212-214 The second step is post-transcriptional and allows the assembly of the NLRP3 inflammasome complex.212, 213 Taken together, these data suggest that during spontaneous labor at term the chorioamniotic membranes increase the production of NLRP3 as an initial step for inflammasome activation. Whether there is assembly of the NLRP3 inflammasome complex in the chorioamniotic membranes during spontaneous labor at term requires further investigation.
We also found that the chorioamniotic membranes from women who underwent spontaneous labor had greater concentrations of NOD2 than those from women without labor, and such an increase was not associated with an up-regulation of NOD2. Recently, it was demonstrated that the mRNA expression of NOD1 and NOD2 in the myometrium is higher in women at term with labor than in those at term without labor.215 NOD2 is an intracellular receptor that recognizes bacterial muramyl dipeptide (MDP) and activates NF-κB and MAPK pathways.216, 217 In dendritic cells, NOD2 can act synergistically with the NLRP3 inflammasome in response to MDP and uric acid.159 These results led us to suggest that the chorioamniotic membranes overproduce NOD2 protein which, in turn, could synergistically participate with the NLRP3 inflammasome in the physiological proinflammatory processes of spontaneous labor at term.
Activation of CASP-1, but not CASP-4, in the chorioamniotic membranes in spontaneous labor at term
Oligomerization of the inflammasome leads to the recruitment of ASC, which binds and activates pro-caspase-1 via its CARD.104, 218 We therefore hypothesized that the active forms of CASP-1, p10 and p20, would be increased in the chorioamniotic membranes from women who underwent spontaneous labor at term. We previously demonstrated that CASP-1 concentration in the amniotic fluid from women in spontaneous labor at term is higher than in women wihout labor.75 In the current study, we provide evidence to support our initial observation: CASP-1 immunoreactivity and its active form p20 are increased in the chorioamniotic membranes from women who underwent spontaneous labor at term. Recently, it was also found that the mRNA expression and active form p10 of CASP-1 are increased in the zone of rupture of the chorioamniotic membranes from women who undergo spontaneous labor at term.219 Together, these data suggest that during spontaneous labor at term, the chorioamniotic membranes release active forms of CASP1.
Unlike Lappas,219 we did not find differences between the mRNA expression of CASP-1 in the chorioamniotic membranes from women who underwent labor and non-labor deliveries. This discrepancy could be attributed to differences in sampling, number of observations, and inclusion of decidua in our experiments. Specifically, we sampled the middle portion of the chorioamniotic membranes and did not restrict ourselves to sampling the zone of rupture.66 Moreover, we utilized full-thickness, unaltered chorioamniotic membranes containing decidua parietalis since this type of sample includes immune cells that participate in labor;220, 221 and our sample size (n=28-29) was considerably larger than that included in the aforementioned study (n=8).
CASP-11 (human homologue CASP-4) is necessary for the activation of CASP-1, which results in the non-canonical NLRP3 inflammasome activation in response to Gram-negative bacteria such as Citrobacter rodentium, Escherichia coli, Vibrio cholerae, and Salmonella typhimurium.210, 222, 223 Although we found that CASP-4 immunoreactivity was higher in the chorioamniotic membranes from women who underwent spontaneous labor at term than in those without labor, we did not find the active form of CASP-4 in these tissues. These results demonstrate that the activation of CASP-4 is not implicated in the physiological proinflammatory processes of spontaneous labor at term. Our results also suggest that the chorioamniotic membranes can synthesize CASP-4 in the event of infection, where Gram-negative bacteria may cause non-canonical activation of inflammasomes.
Increased mRNA abundance and protein concentration of IL-1β in the chorioamniotic membranes in spontaneous labor at term
The produced active forms of CASP-1 (p10 and p20) assemble to form hetero-tetramers that convert inactive pro-IL-1β into its bioactive and secreted form.113, 224-230 Therefore, we investigated whether the chorioamniotic membranes express IL1B and release its bioactive form. We found that the mRNA abundance and release of the pro- and mature forms of IL-1β were higher in the chorioamniotic membranes from women who had undergone spontaneous labor at term than in those without labor. Elevated amniotic fluid IL-1β in spontaneous labor at term was demonstrated more than two decades ago.7-9 IL-1β actively participates in the process of labor by inducing: 1) the biosynthesis of prostaglandin E2 by the human amnion231 and myometrial cells232, 233, 2) the expression of cyclooxygenase-2 in human myometrial cells,234 and 3) the expression of matrix-metabolizing enzymes (MMP-1, -3, -9, and cathepsin S) in human cervical smooth muscle cells.235 Indeed, systemic administration of IL-1β causes preterm birth in mice10, 236 and monkeys,237-244 confirming a central role for IL-1β in the process of labor, and this effect can be abrogated by the administration of the IL-1 receptor antagonist.10 In the study herein, we demonstrated that the chorioamniotic membranes release mature IL-1β, which is most likely mediated by active CASP-1. This mature form of IL-1β will then participate in the physiological process of spontaneous labor at term.
mRNA abundance and protein concentration of IL-18 in the chorioamniotic membranes do not increase in spontaneous labor at term
In addition to cleaving IL-1β, active CASP-1 converts pro-IL-18 into its mature form.115-118, 124 Yet, contrary to IL-1β, IL-18 concentration in the amniotic fluid does not significantly increase during term and preterm parturition.173 This is consistent with our results since no increase was observed in the concentration of IL-18 in the chorioamniotic membrane extracts from women who underwent spontaneous labor at term when compared to those women without labor. Indeed, the mRNA abundance of IL-18 was lower in the chorioamniotic membranes from women who underwent labor than in those without labor, and the mature form of IL-18 was observed mainly in non-laboring tissues at term. IL-18 is a major IFN-γ inducing factor that activates Th-1 responses in T cells and NK cells,245-252 and its concentration in the amniotic fluid increased in response to intra-amniotic infection.173, 253 It is possible that IL-18 participates in host defense against pathogens in the chorioamniotic membranes rather than in the physiologic inflammatory process of spontaneous labor at term.
In summary, the study herein provides evidence that supports a role for the inflammasome (NLRP1, NLRP3, AIM2, or NLRC4) in the activation of CASP-1 and the consequent release of mature IL-1β by the chorioamniotic membranes in spontaneous labor at term. Up-regulation of NLRP3 and NOD2 proteins suggests that these NLRs are implicated in the physiological proinflammatory process of spontaneous labor at term; yet, further research is needed in order to prove inflammasome assembly and activation in the chorioamniotic membranes.
Acknowledgments
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, U. S. Department of Health and Human Services (NICHD/NIH/DHHS), and, in part, with federal funds from the NICHD/NIH/DHHS under Contract No. HHSN275201300006C. We gratefully acknowledge Amy-Eunice Furcron, Lorri McLuckie, Rona Wang, Yang Jiang and Gerardo Rodriguez for their contributions to the execution of this study. We thank the physicians and nurses from the Center for Advanced Obstetrical Care and Research, and the research assistants from the PRB Clinical Laboratory, for their help in collecting human samples. We also thank staff members of the PRB Histology and Pathology Units for their examination of the pathological sections, and Maureen McGerty for her critical readings of the manuscript.
Footnotes
DISCLOSURE/CONFLICT OF INTEREST
The authors disclose no conflicts of interest.
References
- 1.Romero R, Espinoza J, Goncalves LF, Kusanovic JP, Friel LA, Nien JK. Inflammation in preterm and term labour and delivery. Semin Fetal Neonatal Med. 2006;11:317–326. doi: 10.1016/j.siny.2006.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Haddad R, Tromp G, Kuivaniemi H, Chaiworapongsa T, Kim YM, Mazor M, Romero R. Human spontaneous labor without histologic chorioamnionitis is characterized by an acute inflammation gene expression signature. Am J Obstet Gynecol. 2006;195:394 e391–324. doi: 10.1016/j.ajog.2005.08.057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Hassan SS, Romero R, Haddad R, Hendler I, Khalek N, Tromp G, Diamond MP, Sorokin Y, Malone J., Jr The transcriptome of the uterine cervix before and after spontaneous term parturition. Am J Obstet Gynecol. 2006;195:778–786. doi: 10.1016/j.ajog.2006.06.021. [DOI] [PubMed] [Google Scholar]
- 4.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:S194–202. doi: 10.1111/j.1753-4887.2007.tb00362.x. [DOI] [PubMed] [Google Scholar]
- 5.Norman JE, Bollapragada S, Yuan M, Nelson SM. Inflammatory pathways in the mechanism of parturition. BMC Pregnancy Childbirth. 2007;7(Suppl 1):S7. doi: 10.1186/1471-2393-7-S1-S7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Mittal P, Romero R, Tarca AL, Gonzalez J, Draghici S, Xu Y, Dong Z, Nhan-Chang CL, Chaiworapongsa T, Lye S, Kusanovic JP, Lipovich L, Mazaki-Tovi S, Hassan SS, Mesiano S, Kim CJ. Characterization of the myometrial transcriptome and biological pathways of spontaneous human labor at term. J Perinat Med. 2010;38:617–643. doi: 10.1515/JPM.2010.097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Romero R, Brody DT, Oyarzun E, Mazor M, Wu YK, Hobbins JC, Durum SK. Infection and labor. III. Interleukin-1: a signal for the onset of parturition. Am J Obstet Gynecol. 1989;160:1117–1123. doi: 10.1016/0002-9378(89)90172-5. [DOI] [PubMed] [Google Scholar]
- 8.Romero R, Parvizi ST, Oyarzun E, Mazor M, Wu YK, Avila C, Athanassiadis AP, Mitchell MD. Amniotic fluid interleukin-1 in spontaneous labor at term. J Reprod Med. 1990;35:235–238. [PubMed] [Google Scholar]
- 9.Romero R, Mazor M, Brandt F, Sepulveda W, Avila C, Cotton DB, Dinarello CA. Interleukin-1 alpha and interleukin-1 beta in preterm and term human parturition. Am J Reprod Immunol. 1992;27:117–123. doi: 10.1111/j.1600-0897.1992.tb00737.x. [DOI] [PubMed] [Google Scholar]
- 10.Romero R, Sepulveda W, Mazor M, Brandt F, Cotton DB, Dinarello CA, Mitchell MD. The natural interleukin-1 receptor antagonist in term and preterm parturition. Am J Obstet Gynecol. 1992;167:863–872. doi: 10.1016/s0002-9378(12)80003-2. [DOI] [PubMed] [Google Scholar]
- 11.Romero R, Mazor M, Sepulveda W, Avila C, Copeland D, Williams J. Tumor necrosis factor in preterm and term labor. Am J Obstet Gynecol. 1992;166:1576–1587. doi: 10.1016/0002-9378(92)91636-o. [DOI] [PubMed] [Google Scholar]
- 12.Saito S, Kasahara T, Kato Y, Ishihara Y, Ichijo M. Elevation of amniotic fluid interleukin 6 (IL-6), IL-8 and granulocyte colony stimulating factor (G-CSF) in term and preterm parturition. Cytokine. 1993;5:81–88. doi: 10.1016/1043-4666(93)90027-3. [DOI] [PubMed] [Google Scholar]
- 13.Opsjln SL, Wathen NC, Tingulstad S, Wiedswang G, Sundan A, Waage A, Austgulen R. Tumor necrosis factor, interleukin-1, and interleukin-6 in normal human pregnancy. Am J Obstet Gynecol. 1993;169:397–404. doi: 10.1016/0002-9378(93)90096-2. [DOI] [PubMed] [Google Scholar]
- 14.Romero R, Gomez R, Galasso M, Mazor M, Berry SM, Quintero RA, Cotton DB. The natural interleukin-1 receptor antagonist in the fetal, maternal, and amniotic fluid compartments: the effect of gestational age, fetal gender, and intrauterine infection. Am J Obstet Gynecol. 1994;171:912–921. doi: 10.1016/s0002-9378(94)70058-3. [DOI] [PubMed] [Google Scholar]
- 15.Andrews WW, Hauth JC, Goldenberg RL, Gomez R, Romero R, Cassell GH. Amniotic fluid interleukin-6: correlation with upper genital tract microbial colonization and gestational age in women delivered after spontaneous labor versus indicated delivery. Am J Obstet Gynecol. 1995;173:606–612. doi: 10.1016/0002-9378(95)90290-2. [DOI] [PubMed] [Google Scholar]
- 16.Olah KS, Vince GS, Neilson JP, Deniz G, Johnson PM. Interleukin-6, interferon-gamma, interleukin-8, and granulocyte-macrophage colony stimulating factor levels in human amniotic fluid at term. J Reprod Immunol. 1996;32:89–98. doi: 10.1016/s0165-0378(96)00990-4. [DOI] [PubMed] [Google Scholar]
- 17.Maymon E, Ghezzi F, Edwin SS, Mazor M, Yoon BH, Gomez R, Romero R. The tumor necrosis factor alpha and its soluble receptor profile in term and preterm parturition. Am J Obstet Gynecol. 1999;181:1142–1148. doi: 10.1016/s0002-9378(99)70097-9. [DOI] [PubMed] [Google Scholar]
- 18.Bowen JM, Chamley L, Keelan JA, Mitchell MD. Cytokines of the placenta and extra-placental membranes: roles and regulation during human pregnancy and parturition. Placenta. 2002;23:257–273. doi: 10.1053/plac.2001.0782. [DOI] [PubMed] [Google Scholar]
- 19.Kemp B, Winkler M, Maas A, Maul H, Ruck P, Reineke T, Rath W. Cytokine concentrations in the amniotic fluid during parturition at term: correlation to lower uterine segment values and to labor. Acta Obstet Gynecol Scand. 2002;81:938–942. doi: 10.1034/j.1600-0412.2002.811007.x. [DOI] [PubMed] [Google Scholar]
- 20.Keelan JA, Blumenstein M, Helliwell RJ, Sato TA, Marvin KW, Mitchell MD. Cytokines, prostaglandins and parturition--a review. Placenta. 2003;24(Suppl A):S33–46. doi: 10.1053/plac.2002.0948. [DOI] [PubMed] [Google Scholar]
- 21.Gotsch F, Romero R, Kusanovic JP, Erez O, Espinoza J, Kim CJ, Vaisbuch E, Than NG, Mazaki-Tovi S, Chaiworapongsa T, Mazor M, Yoon BH, Edwin S, Gomez R, Mittal P, Hassan SS, Sharma S. The anti-inflammatory limb of the immune response in preterm labor, intra-amniotic infection/inflammation, and spontaneous parturition at term: a role for interleukin-10. J Matern Fetal Neonatal Med. 2008;21:529–547. doi: 10.1080/14767050802127349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Romero R, Ceska M, Avila C, Mazor M, Behnke E, Lindley I. Neutrophil attractant/activating peptide-1/interleukin-8 in term and preterm parturition. Am J Obstet Gynecol. 1991;165:813–820. doi: 10.1016/0002-9378(91)90422-n. [DOI] [PubMed] [Google Scholar]
- 23.Romero R, Gomez R, Galasso M, Munoz H, Acosta L, Yoon BH, Svinarich D, Cotton DB. Macrophage inflammatory protein-1 alpha in term and preterm parturition: effect of microbial invasion of the amniotic cavity. Am J Reprod Immunol. 1994;32:108–113. doi: 10.1111/j.1600-0897.1994.tb01101.x. [DOI] [PubMed] [Google Scholar]
- 24.Dudley DJ, Hunter C, Mitchell MD, Varner MW. Elevations of amniotic fluid macrophage inflammatory protein-1 alpha concentrations in women during term and preterm labor. Obstet Gynecol. 1996;87:94–98. doi: 10.1016/0029-7844(95)00366-5. [DOI] [PubMed] [Google Scholar]
- 25.Athayde N, Romero R, Maymon E, Gomez R, Pacora P, Araneda H, Yoon BH. A role for the novel cytokine RANTES in pregnancy and parturition. Am J Obstet Gynecol. 1999;181:989–994. doi: 10.1016/s0002-9378(99)70337-6. [DOI] [PubMed] [Google Scholar]
- 26.Esplin MS, Romero R, Chaiworapongsa T, Kim YM, Edwin S, Gomez R, Gonzalez R, Adashi EY. Amniotic fluid levels of immunoreactive monocyte chemotactic protein-1 increase during term parturition. J Matern Fetal Neonatal Med. 2003;14:51–56. doi: 10.1080/jmf.14.1.51.56. [DOI] [PubMed] [Google Scholar]
- 27.Hamill N, Romero R, Gotsch F, Kusanovic JP, Edwin S, Erez O, Than NG, Mittal P, Espinoza J, Friel LA, Vaisbuch E, Mazaki-Tovi S, Hassan SS. Exodus-1 (CCL20): evidence for the participation of this chemokine in spontaneous labor at term, preterm labor, and intrauterine infection. J Perinat Med. 2008;36:217–227. doi: 10.1515/JPM.2008.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Yellon SM, Mackler AM, Kirby MA. The role of leukocyte traffic and activation in parturition. J Soc Gynecol Investig. 2003;10:323–338. doi: 10.1016/s1071-5576(03)00116-3. [DOI] [PubMed] [Google Scholar]
- 29.Unal ER, Cierny JT, Roedner C, Newman R, Goetzl L. Maternal inflammation in spontaneous term labor. Am J Obstet Gynecol. 2011;204:223 e221–225. doi: 10.1016/j.ajog.2011.01.002. [DOI] [PubMed] [Google Scholar]
- 30.Cierny JT, Unal ER, Flood P, Rhee KY, Praktish A, Olson TH, Goetzl L. Maternal inflammatory markers and term labor performance. Am J Obstet Gynecol. 2014;210:447 e441–446. doi: 10.1016/j.ajog.2013.11.038. [DOI] [PubMed] [Google Scholar]
- 31.Neal JL, Lamp JM, Lowe NK, Gillespie SL, Sinnott LT, McCarthy DO. Differences in inflammatory markers between nulliparous women admitted to hospitals in preactive vs active labor. Am J Obstet Gynecol. 2015;212:68 e61–68. doi: 10.1016/j.ajog.2014.07.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Taniguchi T, Matsuzaki N, Kameda T, Shimoya K, Jo T, Saji F, Tanizawa O. The enhanced production of placental interleukin-1 during labor and intrauterine infection. Am J Obstet Gynecol. 1991;165:131–137. doi: 10.1016/0002-9378(91)90241-i. [DOI] [PubMed] [Google Scholar]
- 33.Fidel PL, Jr, Romero R, Ramirez M, Cutright J, Edwin SS, LaMarche S, Cotton DB, Mitchell MD. Interleukin-1 receptor antagonist (IL-1ra) production by human amnion, chorion, and decidua. Am J Reprod Immunol. 1994;32:1–7. doi: 10.1111/j.1600-0897.1994.tb00872.x. [DOI] [PubMed] [Google Scholar]
- 34.Dudley DJ, Collmer D, Mitchell MD, Trautman MS. Inflammatory cytokine mRNA in human gestational tissues: implications for term and preterm labor. J Soc Gynecol Investig. 1996;3:328–335. [PubMed] [Google Scholar]
- 35.Ammala M, Nyman T, Salmi A, Rutanen EM. The interleukin-1 system in gestational tissues at term: effect of labour. Placenta. 1997;18:717–723. doi: 10.1016/s0143-4004(97)90014-x. [DOI] [PubMed] [Google Scholar]
- 36.Keelan JA, Marvin KW, Sato TA, Coleman M, McCowan LM, Mitchell MD. Cytokine abundance in placental tissues: evidence of inflammatory activation in gestational membranes with term and preterm parturition. Am J Obstet Gynecol. 1999;181:1530–1536. doi: 10.1016/s0002-9378(99)70400-x. [DOI] [PubMed] [Google Scholar]
- 37.Young A, Thomson AJ, Ledingham M, Jordan F, Greer IA, Norman JE. Immunolocalization of proinflammatory cytokines in myometrium, cervix, and fetal membranes during human parturition at term. Biol Reprod. 2002;66:445–449. doi: 10.1095/biolreprod66.2.445. [DOI] [PubMed] [Google Scholar]
- 38.Lonergan M, Aponso D, Marvin KW, Helliwell RJ, Sato TA, Mitchell MD, Chaiwaropongsa T, Romero R, Keelan JA. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), TRAIL receptors, and the soluble receptor osteoprotegerin in human gestational membranes and amniotic fluid during pregnancy and labor at term and preterm. J Clin Endocrinol Metab. 2003;88:3835–3844. doi: 10.1210/jc.2002-021905. [DOI] [PubMed] [Google Scholar]
- 39.Osman I, Young A, Ledingham MA, Thomson AJ, Jordan F, Greer IA, Norman JE. Leukocyte density and pro-inflammatory cytokine expression in human fetal membranes, decidua, cervix and myometrium before and during labour at term. Mol Hum Reprod. 2003;9:41–45. doi: 10.1093/molehr/gag001. [DOI] [PubMed] [Google Scholar]
- 40.Esplin MS, Peltier MR, Hamblin S, Smith S, Fausett MB, Dildy GA, Branch DW, Silver RM, Adashi EY. Monocyte chemotactic protein-1 expression is increased in human gestational tissues during term and preterm labor. Placenta. 2005;26:661–671. doi: 10.1016/j.placenta.2004.09.012. [DOI] [PubMed] [Google Scholar]
- 41.Kim GJ, Romero R, Kuivaniemi H, Tromp G, Haddad R, Kim YM, Kim MR, Nien JK, Hong JS, Espinoza J, Santolaya J, Yoon BH, Mazor M, Kim CJ. Expression of bone morphogenetic protein 2 in normal spontaneous labor at term, preterm labor, and preterm premature rupture of membranes. Am J Obstet Gynecol. 2005;193:1137–1143. doi: 10.1016/j.ajog.2005.06.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Kim YM, Romero R, Chaiworapongsa T, Kim GJ, Kim MR, Kuivaniemi H, Tromp G, Espinoza J, Bujold E, Abrahams VM, Mor G. Toll-like receptor-2 and -4 in the chorioamniotic membranes in spontaneous labor at term and in preterm parturition that are associated with chorioamnionitis. Am J Obstet Gynecol. 2004;191:1346–1355. doi: 10.1016/j.ajog.2004.07.009. [DOI] [PubMed] [Google Scholar]
- 43.Koga K, Mor G. Toll-like receptors at the maternal-fetal interface in normal pregnancy and pregnancy disorders. Am J Reprod Immunol. 2010;63:587–600. doi: 10.1111/j.1600-0897.2010.00848.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Koga K, Izumi G, Mor G, Fujii T, Osuga Y. Toll-like receptors at the maternal-fetal interface in normal pregnancy and pregnancy complications. Am J Reprod Immunol. 2014;72:192–205. doi: 10.1111/aji.12258. [DOI] [PubMed] [Google Scholar]
- 45.Stephen GL, Lui S, Hamilton SA, Tower CL, Harris LK, Stevens A, Jones RL. Transcriptomic profiling of human choriodecidua during term labor: inflammation as a key driver of labor. Am J Reprod Immunol. 2015;73:36–55. doi: 10.1111/aji.12328. [DOI] [PubMed] [Google Scholar]
- 46.Gomez-Lopez N, Tong WC, Arenas-Hernandez M, Tanaka S, Hajar O, Olson DM, Taggart MJ, Mitchell BF. Chemotactic activity of gestational tissues through late pregnancy, term labor, and RU486-induced preterm labor in Guinea pigs. Am J Reprod Immunol. 2015;73:341–352. doi: 10.1111/aji.12333. [DOI] [PubMed] [Google Scholar]
- 47.Liggins G. Cervical ripening as an inflammatory reaction. In: Ellwood E, Anderson A, editors. The cervix in pregnancy and labor: clinical and biochemical investigations. Edinburgh: Churchill Livingstone; 1981. pp. 1–9. [Google Scholar]
- 48.Bokstrom H, Brannstrom M, Alexandersson M, Norstrom A. Leukocyte subpopulations in the human uterine cervical stroma at early and term pregnancy. Hum Reprod. 1997;12:586–590. doi: 10.1093/humrep/12.3.586. [DOI] [PubMed] [Google Scholar]
- 49.Mackler AM, Iezza G, Akin MR, McMillan P, Yellon SM. Macrophage trafficking in the uterus and cervix precedes parturition in the mouse. Biol Reprod. 1999;61:879–883. doi: 10.1095/biolreprod61.4.879. [DOI] [PubMed] [Google Scholar]
- 50.Kelly RW. Inflammatory mediators and cervical ripening. J Reprod Immunol. 2002;57:217–224. doi: 10.1016/s0165-0378(02)00007-4. [DOI] [PubMed] [Google Scholar]
- 51.Sakamoto Y, Moran P, Bulmer JN, Searle RF, Robson SC. Macrophages and not granulocytes are involved in cervical ripening. J Reprod Immunol. 2005;66:161–173. doi: 10.1016/j.jri.2005.04.005. [DOI] [PubMed] [Google Scholar]
- 52.Yellon SM, Ebner CA, Sugimoto Y. Parturition and recruitment of macrophages in cervix of mice lacking the prostaglandin F receptor. Biol Reprod. 2008;78:438–444. doi: 10.1095/biolreprod.107.063404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Yellon SM, Oshiro BT, Chhaya TY, Lechuga TJ, Dias RM, Burns AE, Force L, Apostolakis EM. Remodeling of the cervix and parturition in mice lacking the progesterone receptor B isoform. Biol Reprod. 2011;85:498–502. doi: 10.1095/biolreprod.111.091983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Clyde LA, Lechuga TJ, Ebner CA, Burns AE, Kirby MA, Yellon SM. Transection of the pelvic or vagus nerve forestalls ripening of the cervix and delays birth in rats. Biol Reprod. 2011;84:587–594. doi: 10.1095/biolreprod.110.086207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Payne KJ, Clyde LA, Weldon AJ, Milford TA, Yellon SM. Residency and activation of myeloid cells during remodeling of the prepartum murine cervix. Biol Reprod. 2012;87:106. doi: 10.1095/biolreprod.112.101840. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Myers DA. The recruitment and activation of leukocytes into the immune cervix: further support that cervical remodeling involves an immune and inflammatory mechanism. Biol Reprod. 2012;87:107. doi: 10.1095/biolreprod.112.105049. [DOI] [PubMed] [Google Scholar]
- 57.Thomson AJ, Telfer JF, Young A, Campbell S, Stewart CJ, Cameron IT, Greer IA, Norman JE. Leukocytes infiltrate the myometrium during human parturition: further evidence that labour is an inflammatory process. Hum Reprod. 1999;14:229–236. [PubMed] [Google Scholar]
- 58.Shynlova O, Tsui P, Dorogin A, Lye SJ. Monocyte chemoattractant protein-1 (CCL-2) integrates mechanical and endocrine signals that mediate term and preterm labor. J Immunol. 2008;181:1470–1479. doi: 10.4049/jimmunol.181.2.1470. [DOI] [PubMed] [Google Scholar]
- 59.Shynlova O, Tsui P, Jaffer S, Lye SJ. Integration of endocrine and mechanical signals in the regulation of myometrial functions during pregnancy and labour. Eur J Obstet Gynecol Reprod Biol. 2009;144(Suppl 1):S2–10. doi: 10.1016/j.ejogrb.2009.02.044. [DOI] [PubMed] [Google Scholar]
- 60.Hamilton S, Oomomian Y, Stephen G, Shynlova O, Tower CL, Garrod A, Lye SJ, Jones RL. Macrophages infiltrate the human and rat decidua during term and preterm labor: evidence that decidual inflammation precedes labor. Biol Reprod. 2012;86:39. doi: 10.1095/biolreprod.111.095505. [DOI] [PubMed] [Google Scholar]
- 61.Shynlova O, Nedd-Roderique T, Li Y, Dorogin A, Nguyen T, Lye SJ. Infiltration of myeloid cells into decidua is a critical early event in the labour cascade and post-partum uterine remodelling. J Cell Mol Med. 2013;17:311–324. doi: 10.1111/jcmm.12012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Shynlova O, Lee YH, Srikhajon K, Lye SJ. Physiologic uterine inflammation and labor onset: integration of endocrine and mechanical signals. Reprod Sci. 2013;20:154–167. doi: 10.1177/1933719112446084. [DOI] [PubMed] [Google Scholar]
- 63.Arenas-Hernandez M, Romero R, St Louis D, Hassan SS, Kaye EB, Gomez-Lopez N. An imbalance between innate and adaptive immune cells at the maternal-fetal interface occurs prior to endotoxin-induced preterm birth. Cell Mol Immunol. 2015 doi: 10.1038/cmi.2015.22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Osman I, Young A, Jordan F, Greer IA, Norman JE. Leukocyte density and proinflammatory mediator expression in regional human fetal membranes and decidua before and during labor at term. J Soc Gynecol Investig. 2006;13:97–103. doi: 10.1016/j.jsgi.2005.12.002. [DOI] [PubMed] [Google Scholar]
- 65.Gomez-Lopez N, Estrada-Gutierrez G, Jimenez-Zamudio L, Vega-Sanchez R, Vadillo-Ortega F. Fetal membranes exhibit selective leukocyte chemotaxic activity during human labor. J Reprod Immunol. 2009;80:122–131. doi: 10.1016/j.jri.2009.01.002. [DOI] [PubMed] [Google Scholar]
- 66.Gomez-Lopez N, Vadillo-Perez L, Hernandez-Carbajal A, Godines-Enriquez M, Olson DM, Vadillo-Ortega F. Specific inflammatory microenvironments in the zones of the fetal membranes at term delivery. Am J Obstet Gynecol. 2011;205:235 e215–224. doi: 10.1016/j.ajog.2011.04.019. [DOI] [PubMed] [Google Scholar]
- 67.Gomez-Lopez N, Vadillo-Perez L, Nessim S, Olson DM, Vadillo-Ortega F. Choriodecidua and amnion exhibit selective leukocyte chemotaxis during term human labor. Am J Obstet Gynecol. 2011;204:364 e369–316. doi: 10.1016/j.ajog.2010.11.010. [DOI] [PubMed] [Google Scholar]
- 68.Gomez-Lopez N, Vega-Sanchez R, Castillo-Castrejon M, Romero R, Cubeiro-Arreola K, Vadillo-Ortega F. Evidence for a role for the adaptive immune response in human term parturition. Am J Reprod Immunol. 2013;69:212–230. doi: 10.1111/aji.12074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Romero R, Nores J, Mazor M, Sepulveda W, Oyarzun E, Parra M, Insunza A, Montiel F, Behnke E, Cassell GH. Microbial invasion of the amniotic cavity during term labor. Prevalence and clinical significance. J Reprod Med. 1993;38:543–548. [PubMed] [Google Scholar]
- 70.Bollapragada S, Youssef R, Jordan F, Greer I, Norman J, Nelson S. Term labor is associated with a core inflammatory response in human fetal membranes, myometrium, and cervix. Am J Obstet Gynecol. 2009;200:104 e101–111. doi: 10.1016/j.ajog.2008.08.032. [DOI] [PubMed] [Google Scholar]
- 71.Hassan SS, Romero R, Tarca AL, Nhan-Chang CL, Vaisbuch E, Erez O, Mittal P, Kusanovic JP, Mazaki-Tovi S, Yeo L, Draghici S, Kim JS, Uldbjerg N, Kim CJ. The transcriptome of cervical ripening in human pregnancy before the onset of labor at term: identification of novel molecular functions involved in this process. J Matern Fetal Neonatal Med. 2009;22:1183–1193. doi: 10.3109/14767050903353216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Nhan-Chang CL, Romero R, Tarca AL, Mittal P, Kusanovic JP, Erez O, Mazaki-Tovi S, Chaiworapongsa T, Hotra J, Than NG, Kim JS, Hassan SS, Kim CJ. Characterization of the transcriptome of chorioamniotic membranes at the site of rupture in spontaneous labor at term. Am J Obstet Gynecol. 2010;202:462 e461–441. doi: 10.1016/j.ajog.2010.02.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Pineles BL, Romero R, Montenegro D, Tarca AL, Than NG, Hassan S, Gotsch F, Draghici S, Espinoza J, Kim CJ. “The inflammasome” in human parturition. Reproductive Sciences. 2007;14:59A. [Google Scholar]
- 74.Montenegro D, Romero R, Pineles P, Tarca AL, Madsen-Bouterse SA, Hassan S, Kusanovic JP, Draghici S, Espinoza J, Kim CJ. Differential expression of the inflammasome components in the fetal inflammatory response syndrome. Reproductive Sciences. 2007;14:59A–60A. [Google Scholar]
- 75.Gotsch F, Romero R, Chaiworapongsa T, Erez O, Vaisbuch E, Espinoza J, Kusanovic JP, Mittal P, Mazaki-Tovi S, Kim CJ, Kim JS, Edwin S, Nhan-Chang CL, Hamill N, Friel L, Than NG, Mazor M, Yoon BH, Hassan SS. Evidence of the involvement of caspase-1 under physiologic and pathologic cellular stress during human pregnancy: a link between the inflammasome and parturition. J Matern Fetal Neonatal Med. 2008;21:605–616. doi: 10.1080/14767050802212109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Martinon F, Burns K, Tschopp J. The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol Cell. 2002;10:417–426. doi: 10.1016/s1097-2765(02)00599-3. [DOI] [PubMed] [Google Scholar]
- 77.Petrilli V, Papin S, Tschopp J. The inflammasome. Curr Biol. 2005;15:R581. doi: 10.1016/j.cub.2005.07.049. [DOI] [PubMed] [Google Scholar]
- 78.Ogura Y, Sutterwala FS, Flavell RA. The inflammasome: first line of the immune response to cell stress. Cell. 2006;126:659–662. doi: 10.1016/j.cell.2006.08.002. [DOI] [PubMed] [Google Scholar]
- 79.Sutterwala FS, Ogura Y, Flavell RA. The inflammasome in pathogen recognition and inflammation. J Leukoc Biol. 2007;82:259–264. doi: 10.1189/jlb.1206755. [DOI] [PubMed] [Google Scholar]
- 80.Mariathasan S, Monack DM. Inflammasome adaptors and sensors: intracellular regulators of infection and inflammation. Nat Rev Immunol. 2007;7:31–40. doi: 10.1038/nri1997. [DOI] [PubMed] [Google Scholar]
- 81.Stutz A, Golenbock DT, Latz E. Inflammasomes: too big to miss. J Clin Invest. 2009;119:3502–3511. doi: 10.1172/JCI40599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Franchi L, Eigenbrod T, Munoz-Planillo R, Nunez G. The inflammasome: a caspase-1-activation platform that regulates immune responses and disease pathogenesis. Nat Immunol. 2009;10:241–247. doi: 10.1038/ni.1703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Jha S, Ting JP. Inflammasome-associated nucleotide-binding domain, leucine-rich repeat proteins and inflammatory diseases. J Immunol. 2009;183:7623–7629. doi: 10.4049/jimmunol.0902425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Pedra JH, Cassel SL, Sutterwala FS. Sensing pathogens and danger signals by the inflammasome. Curr Opin Immunol. 2009;21:10–16. doi: 10.1016/j.coi.2009.01.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Lamkanfi M, Dixit VM. Inflammasomes: guardians of cytosolic sanctity. Immunol Rev. 2009;227:95–105. doi: 10.1111/j.1600-065X.2008.00730.x. [DOI] [PubMed] [Google Scholar]
- 86.Latz E. The inflammasomes: mechanisms of activation and function. Curr Opin Immunol. 2010;22:28–33. doi: 10.1016/j.coi.2009.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Schroder K, Tschopp J. The inflammasomes. Cell. 2010;140:821–832. doi: 10.1016/j.cell.2010.01.040. [DOI] [PubMed] [Google Scholar]
- 88.Franchi L, Munoz-Planillo R, Reimer T, Eigenbrod T, Nunez G. Inflammasomes as microbial sensors. Eur J Immunol. 2010;40:611–615. doi: 10.1002/eji.200940180. [DOI] [PubMed] [Google Scholar]
- 89.Bauernfeind F, Ablasser A, Bartok E, Kim S, Schmid-Burgk J, Cavlar T, Hornung V. Inflammasomes: current understanding and open questions. Cell Mol Life Sci. 2011;68:765–783. doi: 10.1007/s00018-010-0567-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Kersse K, Bertrand MJ, Lamkanfi M, Vandenabeele P. NOD-like receptors and the innate immune system: coping with danger, damage and death. Cytokine Growth Factor Rev. 2011;22:257–276. doi: 10.1016/j.cytogfr.2011.09.003. [DOI] [PubMed] [Google Scholar]
- 91.Gross O, Thomas CJ, Guarda G, Tschopp J. The inflammasome: an integrated view. Immunol Rev. 2011;243:136–151. doi: 10.1111/j.1600-065X.2011.01046.x. [DOI] [PubMed] [Google Scholar]
- 92.Lamkanfi M. Emerging inflammasome effector mechanisms. Nat Rev Immunol. 2011;11:213–220. doi: 10.1038/nri2936. [DOI] [PubMed] [Google Scholar]
- 93.Lamkanfi M, Dixit VM. Modulation of inflammasome pathways by bacterial and viral pathogens. J Immunol. 2011;187:597–602. doi: 10.4049/jimmunol.1100229. [DOI] [PubMed] [Google Scholar]
- 94.Broz P, Monack DM. Molecular mechanisms of inflammasome activation during microbial infections. Immunol Rev. 2011;243:174–190. doi: 10.1111/j.1600-065X.2011.01041.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Skeldon A, Saleh M. The inflammasomes: molecular effectors of host resistance against bacterial, viral, parasitic, and fungal infections. Front Microbiol. 2011;2:15. doi: 10.3389/fmicb.2011.00015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Horvath GL, Schrum JE, De Nardo CM, Latz E. Intracellular sensing of microbes and danger signals by the inflammasomes. Immunol Rev. 2011;243:119–135. doi: 10.1111/j.1600-065X.2011.01050.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.van de Veerdonk FL, Netea MG, Dinarello CA, Joosten LA. Inflammasome activation and IL-1beta and IL-18 processing during infection. Trends Immunol. 2011;32:110–116. doi: 10.1016/j.it.2011.01.003. [DOI] [PubMed] [Google Scholar]
- 98.Franchi L, Munoz-Planillo R, Nunez G. Sensing and reacting to microbes through the inflammasomes. Nat Immunol. 2012;13:325–332. doi: 10.1038/ni.2231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Dagenais M, Skeldon A, Saleh M. The inflammasome: in memory of Dr. Jurg Tschopp. Cell Death Differ. 2012;19:5–12. doi: 10.1038/cdd.2011.159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Ciraci C, Janczy JR, Sutterwala FS, Cassel SL. Control of innate and adaptive immunity by the inflammasome. Microbes Infect. 2012 doi: 10.1016/j.micinf.2012.07.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Rathinam VA, Vanaja SK, Fitzgerald KA. Regulation of inflammasome signaling. Nat Immunol. 2012;13:333–332. doi: 10.1038/ni.2237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Franchi L, Nunez G. Immunology. Orchestrating inflammasomes. Science. 2012;337:1299–1300. doi: 10.1126/science.1229010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Henao-Mejia J, Elinav E, Strowig T, Flavell RA. Inflammasomes: far beyond inflammation. Nat Immunol. 2012;13:321–324. doi: 10.1038/ni.2257. [DOI] [PubMed] [Google Scholar]
- 104.Latz E, Xiao TS, Stutz A. Activation and regulation of the inflammasomes. Nat Rev Immunol. 2013;13:397–411. doi: 10.1038/nri3452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Bauernfeind F, Hornung V. Of inflammasomes and pathogens--sensing of microbes by the inflammasome. EMBO Mol Med. 2013;5:814–826. doi: 10.1002/emmm.201201771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Vladimer GI, Marty-Roix R, Ghosh S, Weng D, Lien E. Inflammasomes and host defenses against bacterial infections. Curr Opin Microbiol. 2013 doi: 10.1016/j.mib.2012.11.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Lamkanfi M, Dixit VM. Mechanisms and functions of inflammasomes. Cell. 2014;157:1013–1022. doi: 10.1016/j.cell.2014.04.007. [DOI] [PubMed] [Google Scholar]
- 108.Ulland TK, Ferguson PJ, Sutterwala FS. Evasion of inflammasome activation by microbial pathogens. J Clin Invest. 2015;125:469–477. doi: 10.1172/JCI75254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Vanaja SK, Rathinam VA, Fitzgerald KA. Mechanisms of inflammasome activation: recent advances and novel insights. Trends Cell Biol. 2015 doi: 10.1016/j.tcb.2014.12.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Guo H, Callaway JB, Ting JP. Inflammasomes: mechanism of action, role in disease, and therapeutics. Nat Med. 2015;21:677–687. doi: 10.1038/nm.3893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Black RA, Kronheim SR, Merriam JE, March CJ, Hopp TP. A pre-aspartate-specific protease from human leukocytes that cleaves pro-interleukin-1 beta. J Biol Chem. 1989;264:5323–5326. [PubMed] [Google Scholar]
- 112.Kostura MJ, Tocci MJ, Limjuco G, Chin J, Cameron P, Hillman AG, Chartrain NA, Schmidt JA. Identification of a monocyte specific pre-interleukin 1 beta convertase activity. Proc Natl Acad Sci U S A. 1989;86:5227–5231. doi: 10.1073/pnas.86.14.5227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Thornberry NA, Bull HG, Calaycay JR, Chapman KT, Howard AD, Kostura MJ, Miller DK, Molineaux SM, Weidner JR, Aunins J, et al. A novel heterodimeric cysteine protease is required for interleukin-1 beta processing in monocytes. Nature. 1992;356:768–774. doi: 10.1038/356768a0. [DOI] [PubMed] [Google Scholar]
- 114.Cerretti DP, Kozlosky CJ, Mosley B, Nelson N, Van Ness K, Greenstreet TA, March CJ, Kronheim SR, Druck T, Cannizzaro LA, et al. Molecular cloning of the interleukin-1 beta converting enzyme. Science. 1992;256:97–100. doi: 10.1126/science.1373520. [DOI] [PubMed] [Google Scholar]
- 115.Gu Y, Kuida K, Tsutsui H, Ku G, Hsiao K, Fleming MA, Hayashi N, Higashino K, Okamura H, Nakanishi K, Kurimoto M, Tanimoto T, Flavell RA, Sato V, Harding MW, Livingston DJ, Su MS. Activation of interferon-gamma inducing factor mediated by interleukin-1beta converting enzyme. Science. 1997;275:206–209. doi: 10.1126/science.275.5297.206. [DOI] [PubMed] [Google Scholar]
- 116.Ghayur T, Banerjee S, Hugunin M, Butler D, Herzog L, Carter A, Quintal L, Sekut L, Talanian R, Paskind M, Wong W, Kamen R, Tracey D, Allen H. Caspase-1 processes IFN-gamma-inducing factor and regulates LPS-induced IFN-gamma production. Nature. 1997;386:619–623. doi: 10.1038/386619a0. [DOI] [PubMed] [Google Scholar]
- 117.Dinarello CA. Interleukin-1 beta, interleukin-18, and the interleukin-1 beta converting enzyme. Ann N Y Acad Sci. 1998;856:1–11. doi: 10.1111/j.1749-6632.1998.tb08307.x. [DOI] [PubMed] [Google Scholar]
- 118.Fantuzzi G, Dinarello CA. Interleukin-18 and interleukin-1 beta: two cytokine substrates for ICE (caspase-1) J Clin Immunol. 1999;19:1–11. doi: 10.1023/a:1020506300324. [DOI] [PubMed] [Google Scholar]
- 119.Sansonetti PJ, Phalipon A, Arondel J, Thirumalai K, Banerjee S, Akira S, Takeda K, Zychlinsky A. Caspase-1 activation of IL-1beta and IL-18 are essential for Shigella flexneri-induced inflammation. Immunity. 2000;12:581–590. doi: 10.1016/s1074-7613(00)80209-5. [DOI] [PubMed] [Google Scholar]
- 120.Kahlenberg JM, Lundberg KC, Kertesy SB, Qu Y, Dubyak GR. Potentiation of caspase-1 activation by the P2X7 receptor is dependent on TLR signals and requires NF-kappaB-driven protein synthesis. J Immunol. 2005;175:7611–7622. doi: 10.4049/jimmunol.175.11.7611. [DOI] [PubMed] [Google Scholar]
- 121.Cayrol C, Girard JP. The IL-1-like cytokine IL-33 is inactivated after maturation by caspase-1. Proc Natl Acad Sci U S A. 2009;106:9021–9026. doi: 10.1073/pnas.0812690106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Luthi AU, Cullen SP, McNeela EA, Duriez PJ, Afonina IS, Sheridan C, Brumatti G, Taylor RC, Kersse K, Vandenabeele P, Lavelle EC, Martin SJ. Suppression of interleukin-33 bioactivity through proteolysis by apoptotic caspases. Immunity. 2009;31:84–98. doi: 10.1016/j.immuni.2009.05.007. [DOI] [PubMed] [Google Scholar]
- 123.Talabot-Ayer D, Lamacchia C, Gabay C, Palmer G. Interleukin-33 is biologically active independently of caspase-1 cleavage. J Biol Chem. 2009;284:19420–19426. doi: 10.1074/jbc.M901744200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.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. 2014 doi: 10.1146/annurev-immunol-032414-112306. [DOI] [PubMed] [Google Scholar]
- 125.Cookson BT, Brennan MA. Pro-inflammatory programmed cell death. Trends Microbiol. 2001;9:113–114. doi: 10.1016/s0966-842x(00)01936-3. [DOI] [PubMed] [Google Scholar]
- 126.Miao EA, Rajan JV, Aderem A. Caspase-1-induced pyroptotic cell death. Immunol Rev. 2011;243:206–214. doi: 10.1111/j.1600-065X.2011.01044.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Shalini S, Dorstyn L, Dawar S, Kumar S. Old, new and emerging functions of caspases. Cell Death Differ. 2015;22:526–539. doi: 10.1038/cdd.2014.216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Sollberger G, Strittmatter GE, Kistowska M, French LE, Beer HD. Caspase-4 is required for activation of inflammasomes. J Immunol. 2012;188:1992–2000. doi: 10.4049/jimmunol.1101620. [DOI] [PubMed] [Google Scholar]
- 129.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;92:501–509. doi: 10.1016/s0092-8674(00)80943-5. [DOI] [PubMed] [Google Scholar]
- 130.Ting JP, Lovering RC, Alnemri ES, Bertin J, Boss JM, Davis BK, Flavell RA, Girardin SE, Godzik A, Harton JA, Hoffman HM, Hugot JP, Inohara N, Mackenzie A, Maltais LJ, Nunez G, Ogura Y, Otten LA, Philpott D, Reed JC, Reith W, Schreiber S, Steimle V, Ward PA. The NLR gene family: a standard nomenclature. Immunity. 2008;28:285–287. doi: 10.1016/j.immuni.2008.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.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;20:319–325. doi: 10.1016/s1074-7613(04)00046-9. [DOI] [PubMed] [Google Scholar]
- 132.Sutterwala FS, Flavell RA. NLRC4/IPAF: a CARD carrying member of the NLR family. Clin Immunol. 2009;130:2–6. doi: 10.1016/j.clim.2008.08.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Qu Y, Misaghi S, Izrael-Tomasevic A, Newton K, Gilmour LL, Lamkanfi M, Louie S, Kayagaki N, Liu J, Komuves L, Cupp JE, Arnott D, Monack D, Dixit VM. Phosphorylation of NLRC4 is critical for inflammasome activation. Nature. 2012;490:539–542. doi: 10.1038/nature11429. [DOI] [PubMed] [Google Scholar]
- 134.Kerur N, Veettil MV, Sharma-Walia N, Bottero V, Sadagopan S, Otageri P, Chandran B. IFI16 acts as a nuclear pathogen sensor to induce the inflammasome in response to Kaposi Sarcoma-associated herpesvirus infection. Cell Host Microbe. 2011;9:363–375. doi: 10.1016/j.chom.2011.04.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Burckstummer T, Baumann C, Bluml S, Dixit E, Durnberger G, Jahn H, Planyavsky M, Bilban M, Colinge J, Bennett KL, Superti-Furga G. An orthogonal proteomic-genomic screen identifies AIM2 as a cytoplasmic DNA sensor for the inflammasome. Nat Immunol. 2009;10:266–272. doi: 10.1038/ni.1702. [DOI] [PubMed] [Google Scholar]
- 136.Fernandes-Alnemri T, Yu JW, Datta P, Wu J, Alnemri ES. AIM2 activates the inflammasome and cell death in response to cytoplasmic DNA. Nature. 2009;458:509–513. doi: 10.1038/nature07710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Roberts TL, Idris A, Dunn JA, Kelly GM, Burnton CM, Hodgson S, Hardy LL, Garceau V, Sweet MJ, Ross IL, Hume DA, Stacey KJ. HIN-200 proteins regulate caspase activation in response to foreign cytoplasmic DNA. Science. 2009;323:1057–1060. doi: 10.1126/science.1169841. [DOI] [PubMed] [Google Scholar]
- 138.Hornung V, Ablasser A, Charrel-Dennis M, Bauernfeind F, Horvath G, Caffrey DR, Latz E, Fitzgerald KA. AIM2 recognizes cytosolic dsDNA and forms a caspase-1-activating inflammasome with ASC. Nature. 2009;458:514–518. doi: 10.1038/nature07725. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Fernandes-Alnemri T, Yu JW, Juliana C, Solorzano L, Kang S, Wu J, Datta P, McCormick M, Huang L, McDermott E, Eisenlohr L, Landel CP, Alnemri ES. The AIM2 inflammasome is critical for innate immunity to Francisella tularensis. Nat Immunol. 2010;11:385–393. doi: 10.1038/ni.1859. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Hornung V, Latz E. Intracellular DNA recognition. Nat Rev Immunol. 2010;10:123–130. doi: 10.1038/nri2690. [DOI] [PubMed] [Google Scholar]
- 141.Franchi L, Nunez G. AIM2 joins the gang of microbial sensors. Cell Host Microbe. 2010;7:340–341. doi: 10.1016/j.chom.2010.05.002. [DOI] [PubMed] [Google Scholar]
- 142.Krieg AM. AIMing 2 defend against intracellular pathogens. Nat Immunol. 2010;11:367–369. doi: 10.1038/ni0510-367. [DOI] [PubMed] [Google Scholar]
- 143.Bird L. Innate immunity: Ready, AIM, fire! Nat Rev Immunol. 2010;10:287. doi: 10.1038/nri2771. [DOI] [PubMed] [Google Scholar]
- 144.Khare S, Luc N, Dorfleutner A, Stehlik C. Inflammasomes and their activation. Crit Rev Immunol. 2010;30:463–487. doi: 10.1615/critrevimmunol.v30.i5.50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Koizumi Y, Toma C, Higa N, Nohara T, Nakasone N, Suzuki T. Inflammasome activation via intracellular NLRs triggered by bacterial infection. Cell Microbiol. 2012;14:149–154. doi: 10.1111/j.1462-5822.2011.01707.x. [DOI] [PubMed] [Google Scholar]
- 146.Kanneganti TD, Lamkanfi M, Nunez G. Intracellular NOD-like receptors in host defense and disease. Immunity. 2007;27:549–559. doi: 10.1016/j.immuni.2007.10.002. [DOI] [PubMed] [Google Scholar]
- 147.Costello MJ, Joyce SK, Abrahams VM. NOD protein expression and function in first trimester trophoblast cells. Am J Reprod Immunol. 2007;57:67–80. doi: 10.1111/j.1600-0897.2006.00447.x. [DOI] [PubMed] [Google Scholar]
- 148.Kim YG, Park JH, Shaw MH, Franchi L, Inohara N, Nunez G. The cytosolic sensors Nod1 and Nod2 are critical for bacterial recognition and host defense after exposure to Toll-like receptor ligands. Immunity. 2008;28:246–257. doi: 10.1016/j.immuni.2007.12.012. [DOI] [PubMed] [Google Scholar]
- 149.King AE, Horne AW, Hombach-Klonisch S, Mason JI, Critchley HO. Differential expression and regulation of nuclear oligomerization domain proteins NOD1 and NOD2 in human endometrium: a potential role in innate immune protection and menstruation. Mol Hum Reprod. 2009;15:311–319. doi: 10.1093/molehr/gap020. [DOI] [PubMed] [Google Scholar]
- 150.Mulla MJ, Yu AG, Cardenas I, Guller S, Panda B, Abrahams VM. Regulation of Nod1 and Nod2 in first trimester trophoblast cells. Am J Reprod Immunol. 2009;61:294–302. doi: 10.1111/j.1600-0897.2009.00694.x. [DOI] [PubMed] [Google Scholar]
- 151.Franchi L, Warner N, Viani K, Nunez G. Function of Nod-like receptors in microbial recognition and host defense. Immunol Rev. 2009;227:106–128. doi: 10.1111/j.1600-065X.2008.00734.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Cardenas I, Mulla MJ, Myrtolli K, Sfakianaki AK, Norwitz ER, Tadesse S, Guller S, Abrahams VM. Nod1 activation by bacterial iE-DAP induces maternal-fetal inflammation and preterm labor. J Immunol. 2011;187:980–986. doi: 10.4049/jimmunol.1100578. [DOI] [PubMed] [Google Scholar]
- 153.Shah A, Hirsch E. TRIF is an essential adaptor protein of TLR3 and NOD2 synergy in macrophages - a role for viral priming in inflammation-induced parturition. Am J Obstet Gynecol. 2014;210:S231–S232. [Google Scholar]
- 154.Dinarello CA. Unraveling the NALP-3/IL-1beta inflammasome: a big lesson from a small mutation. Immunity. 2004;20:243–244. doi: 10.1016/s1074-7613(04)00055-x. [DOI] [PubMed] [Google Scholar]
- 155.Netea MG, Simon A, van de Veerdonk F, Kullberg BJ, Van der Meer JW, Joosten LA. IL-1beta processing in host defense: beyond the inflammasomes. PLoS Pathog. 2010;6:e1000661. doi: 10.1371/journal.ppat.1000661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Dinarello CA. IL-1: discoveries, controversies and future directions. Eur J Immunol. 2010;40:599–606. doi: 10.1002/eji.201040319. [DOI] [PubMed] [Google Scholar]
- 157.Shaw PJ, Lamkanfi M, Kanneganti TD. NOD-like receptor (NLR) signaling beyond the inflammasome. Eur J Immunol. 2010;40:624–627. doi: 10.1002/eji.200940211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Garlanda C, Dinarello CA, Mantovani A. The interleukin-1 family: back to the future. Immunity. 2013;39:1003–1018. doi: 10.1016/j.immuni.2013.11.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Conforti-Andreoni C, Beretta O, Licandro G, Qian HL, Urbano M, Vitulli F, Ricciardi-Castagnoli P, Mortellaro A. Synergism of NOD2 and NLRP3 activators promotes a unique transcriptional profile in murine dendritic cells. J Leukoc Biol. 2010;88:1207–1216. doi: 10.1189/jlb.1009652. [DOI] [PubMed] [Google Scholar]
- 160.von Moltke J, Trinidad NJ, Moayeri M, Kintzer AF, Wang SB, van Rooijen N, Brown CR, Krantz BA, Leppla SH, Gronert K, Vance RE. Rapid induction of inflammatory lipid mediators by the inflammasome in vivo. Nature. 2012;490:107–111. doi: 10.1038/nature11351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Abrahams VM. The role of the Nod-like receptor family in trophoblast innate immune responses. J Reprod Immunol. 2011;88:112–117. doi: 10.1016/j.jri.2010.12.003. [DOI] [PubMed] [Google Scholar]
- 162.Mulla MJ, Myrtolli K, Potter J, Boeras C, Kavathas PB, Sfakianaki AK, Tadesse S, Norwitz ER, Guller S, Abrahams VM. Uric acid induces trophoblast IL-1beta production via the inflammasome: implications for the pathogenesis of preeclampsia. Am J Reprod Immunol. 2011;65:542–548. doi: 10.1111/j.1600-0897.2010.00960.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Mulla MJ, Salmon JE, Chamley LW, Brosens JJ, Boeras CM, Kavathas PB, Abrahams VM. A role for uric acid and the Nalp3 inflammasome in antiphospholipid antibody-induced IL-1beta production by human first trimester trophoblast. PLoS One. 2013;8:e65237. doi: 10.1371/journal.pone.0065237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Kavathas PB, Boeras CM, Mulla MJ, Abrahams VM. Nod1, but not the ASC inflammasome, contributes to induction of IL-1beta secretion in human trophoblasts after sensing of Chlamydia trachomatis. Mucosal Immunol. 2013;6:235–243. doi: 10.1038/mi.2012.63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Hoang M, Potter JA, Gysler SM, Han CS, Guller S, Norwitz ER, Abrahams VM. Human fetal membranes generate distinct cytokine profiles in response to bacterial Toll-like receptor and nod-like receptor agonists. Biol Reprod. 2014;90:39. doi: 10.1095/biolreprod.113.115428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Abrahams VM. Novel mechanisms of placenta inflammation in obstetrics antiphospholipid syndrome. Am J Reprod Immunol. 2014;71:25. [Google Scholar]
- 167.Hansen L, Kotla S, Mari G, Rao G. The role of NLRP3 inflammasome in preeclampsia - a translational approach. Am J Obstet Gynecol. 2014;210:S134. [Google Scholar]
- 168.Khan RN, Hay DP. A clear and present danger: inflammasomes DAMPing down disorders of pregnancy. Hum Reprod Update. 2015;21:388–405. doi: 10.1093/humupd/dmu059. [DOI] [PubMed] [Google Scholar]
- 169.Pontillo A, Reis EC, Bricher PN, Vianna P, Diniz S, Fernandes KS, Chies JA, Sandrim V. NLRP1 L155H Polymorphism is a Risk Factor for Preeclampsia Development. Am J Reprod Immunol. 2015;73:577–581. doi: 10.1111/aji.12353. [DOI] [PubMed] [Google Scholar]
- 170.Matias ML, Romao M, Weel IC, Ribeiro VR, Nunes PR, Borges VT, Araujo JP, Jr, Peracoli JC, de Oliveira L, Peracoli MT. Endogenous and Uric Acid-Induced Activation of NLRP3 Inflammasome in Pregnant Women with Preeclampsia. PLoS One. 2015;10:e0129095. doi: 10.1371/journal.pone.0129095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Maneta E, Warren AY, Hay DP, Khan RN. Caspase-1-mediated cytokine release from gestational tissues, placental, and cord blood. Front Physiol. 2015;6:186. doi: 10.3389/fphys.2015.00186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Srinivasula SM, Poyet JL, Razmara M, Datta P, Zhang Z, Alnemri ES. The PYRIN-CARD protein ASC is an activating adaptor for caspase-1. J Biol Chem. 2002;277:21119–21122. doi: 10.1074/jbc.C200179200. [DOI] [PubMed] [Google Scholar]
- 173.Pacora P, Romero R, Maymon E, Gervasi MT, Gomez R, Edwin SS, Yoon BH. Participation of the novel cytokine interleukin 18 in the host response to intra-amniotic infection. Am J Obstet Gynecol. 2000;183:1138–1143. doi: 10.1067/mob.2000.108881. [DOI] [PubMed] [Google Scholar]
- 174.ACOG Practice Bulletin Number 49, December 2003: Dystocia and augmentation of labor. Obstet Gynecol. 2003;102:1445–1454. doi: 10.1016/j.obstetgynecol.2003.10.011. [DOI] [PubMed] [Google Scholar]
- 175.Redline RW. Placental pathology: a systematic approach with clinical correlations. Placenta. 2008;29(Suppl A):S86–91. doi: 10.1016/j.placenta.2007.09.003. [DOI] [PubMed] [Google Scholar]
- 176.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 doi: 10.1016/j.ajog.2015.08.040. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Martinon F, Tschopp J. Inflammatory caspases: linking an intracellular innate immune system to autoinflammatory diseases. Cell. 2004;117:561–574. doi: 10.1016/j.cell.2004.05.004. [DOI] [PubMed] [Google Scholar]
- 178.Martinon F, Tschopp J. Inflammatory caspases and inflammasomes: master switches of inflammation. Cell Death Differ. 2007;14:10–22. doi: 10.1038/sj.cdd.4402038. [DOI] [PubMed] [Google Scholar]
- 179.Yin Y, Yan Y, Jiang X, Mai J, Chen NC, Wang H, Yang XF. Inflammasomes are differentially expressed in cardiovascular and other tissues. Int J Immunopathol Pharmacol. 2009;22:311–322. doi: 10.1177/039463200902200208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Sutterwala FS, Ogura Y, Zamboni DS, Roy CR, Flavell RA. NALP3: a key player in caspase-1 activation. J Endotoxin Res. 2006;12:251–256. doi: 10.1177/09680519060120040701. [DOI] [PubMed] [Google Scholar]
- 181.Chae JJ, Cho YH, Lee GS, Cheng J, Liu PP, Feigenbaum L, Katz SI, Kastner DL. Gain-of-function Pyrin mutations induce NLRP3 protein-independent interleukin-1beta activation and severe autoinflammation in mice. Immunity. 2011;34:755–768. doi: 10.1016/j.immuni.2011.02.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Dostert C, Petrilli V, Van Bruggen R, Steele C, Mossman BT, Tschopp J. Innate immune activation through Nalp3 inflammasome sensing of asbestos and silica. Science. 2008;320:674–677. doi: 10.1126/science.1156995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Cassel SL, Eisenbarth SC, Iyer SS, Sadler JJ, Colegio OR, Tephly LA, Carter AB, Rothman PB, Flavell RA, Sutterwala FS. The Nalp3 inflammasome is essential for the development of silicosis. Proc Natl Acad Sci U S A. 2008;105:9035–9040. doi: 10.1073/pnas.0803933105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Hornung V, Bauernfeind F, Halle A, Samstad EO, Kono H, Rock KL, Fitzgerald KA, Latz E. Silica crystals and aluminum salts activate the NALP3 inflammasome through phagosomal destabilization. Nat Immunol. 2008;9:847–856. doi: 10.1038/ni.1631. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Yamasaki K, Muto J, Taylor KR, Cogen AL, Audish D, Bertin J, Grant EP, Coyle AJ, Misaghi A, Hoffman HM, Gallo RL. NLRP3/cryopyrin is necessary for interleukin-1beta (IL-1beta) release in response to hyaluronan, an endogenous trigger of inflammation in response to injury. J Biol Chem. 2009;284:12762–12771. doi: 10.1074/jbc.M806084200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Cassel SL, Joly S, Sutterwala FS. The NLRP3 inflammasome: a sensor of immune danger signals. Semin Immunol. 2009;21:194–198. doi: 10.1016/j.smim.2009.05.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Cassel SL, Sutterwala FS. Sterile inflammatory responses mediated by the NLRP3 inflammasome. Eur J Immunol. 2010;40:607–611. doi: 10.1002/eji.200940207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Leemans JC, Cassel SL, Sutterwala FS. Sensing damage by the NLRP3 inflammasome. Immunol Rev. 2011;243:152–162. doi: 10.1111/j.1600-065X.2011.01043.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Martinon F, Petrilli V, Mayor A, Tardivel A, Tschopp J. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature. 2006;440:237–241. doi: 10.1038/nature04516. [DOI] [PubMed] [Google Scholar]
- 190.Mariathasan S, Weiss DS, Newton K, McBride J, O’Rourke K, Roose-Girma M, Lee WP, Weinrauch Y, Monack DM, Dixit VM. Cryopyrin activates the inflammasome in response to toxins and ATP. Nature. 2006;440:228–232. doi: 10.1038/nature04515. [DOI] [PubMed] [Google Scholar]
- 191.Kool M, Petrilli V, De Smedt T, Rolaz A, Hammad H, van Nimwegen M, Bergen IM, Castillo R, Lambrecht BN, Tschopp J. Cutting edge: alum adjuvant stimulates inflammatory dendritic cells through activation of the NALP3 inflammasome. J Immunol. 2008;181:3755–3759. doi: 10.4049/jimmunol.181.6.3755. [DOI] [PubMed] [Google Scholar]
- 192.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;181:17–21. doi: 10.4049/jimmunol.181.1.17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.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;453:1122–1126. doi: 10.1038/nature06939. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.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;38:2085–2089. doi: 10.1002/eji.200838549. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Demento SL, Eisenbarth SC, Foellmer HG, Platt C, Caplan MJ, Mark Saltzman W, Mellman I, Ledizet M, Fikrig E, Flavell RA, Fahmy TM. Inflammasome-activating nanoparticles as modular systems for optimizing vaccine efficacy. Vaccine. 2009;27:3013–3021. doi: 10.1016/j.vaccine.2009.03.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Nakahira K, Haspel JA, Rathinam VA, Lee SJ, Dolinay T, Lam HC, Englert JA, Rabinovitch M, Cernadas M, Kim HP, Fitzgerald KA, Ryter SW, Choi AM. Autophagy proteins regulate innate immune responses by inhibiting the release of mitochondrial DNA mediated by the NALP3 inflammasome. Nat Immunol. 2011;12:222–230. doi: 10.1038/ni.1980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Iyer SS, He Q, Janczy JR, Elliott EI, Zhong Z, Olivier AK, Sadler JJ, Knepper-Adrian V, Han R, Qiao L, Eisenbarth SC, Nauseef WM, Cassel SL, Sutterwala FS. Mitochondrial cardiolipin is required for Nlrp3 inflammasome activation. Immunity. 2013;39:311–323. doi: 10.1016/j.immuni.2013.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.O’Neill LA. Cardiolipin and the Nlrp3 inflammasome. Cell Metab. 2013;18:610–612. doi: 10.1016/j.cmet.2013.10.013. [DOI] [PubMed] [Google Scholar]
- 199.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;126:1135–1145. doi: 10.1016/j.cell.2006.07.033. [DOI] [PubMed] [Google Scholar]
- 200.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;183:3942–3948. doi: 10.4049/jimmunol.0900729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Kanneganti TD, Body-Malapel M, Amer A, Park JH, Whitfield J, Franchi L, Taraporewala ZF, Miller D, Patton JT, Inohara N, Nunez G. Critical role for Cryopyrin/Nalp3 in activation of caspase-1 in response to viral infection and double-stranded RNA. J Biol Chem. 2006;281:36560–36568. doi: 10.1074/jbc.M607594200. [DOI] [PubMed] [Google Scholar]
- 202.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;10:1866–1878. doi: 10.1111/j.1462-5822.2008.01177.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Muruve DA, Petrilli V, Zaiss AK, White LR, Clark SA, Ross PJ, Parks RJ, Tschopp J. The inflammasome recognizes cytosolic microbial and host DNA and triggers an innate immune response. Nature. 2008;452:103–107. doi: 10.1038/nature06664. [DOI] [PubMed] [Google Scholar]
- 204.Thomas PG, Dash P, Aldridge JR, Jr, Ellebedy AH, Reynolds C, Funk AJ, Martin WJ, Lamkanfi M, Webby RJ, Boyd KL, Doherty PC, Kanneganti TD. The intracellular sensor NLRP3 mediates key innate and healing responses to influenza A virus via the regulation of caspase-1. Immunity. 2009;30:566–575. doi: 10.1016/j.immuni.2009.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Allen IC, Scull MA, Moore CB, Holl EK, McElvania-TeKippe E, Taxman DJ, Guthrie EH, Pickles RJ, Ting JP. The NLRP3 inflammasome mediates in vivo innate immunity to influenza A virus through recognition of viral RNA. Immunity. 2009;30:556–565. doi: 10.1016/j.immuni.2009.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Duncan JA, Gao X, Huang MT, O’Connor BP, Thomas CE, Willingham SB, Bergstralh DT, Jarvis GA, Sparling PF, Ting JP. Neisseria gonorrhoeae activates the proteinase cathepsin B to mediate the signaling activities of the NLRP3 and ASC-containing inflammasome. J Immunol. 2009;182:6460–6469. doi: 10.4049/jimmunol.0802696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.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;183:3578–3581. doi: 10.4049/jimmunol.0901323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.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;206:79–87. doi: 10.1084/jem.20081667. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Menu P, Vince JE. The NLRP3 inflammasome in health and disease: the good, the bad and the ugly. Clin Exp Immunol. 2011;166:1–15. doi: 10.1111/j.1365-2249.2011.04440.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Rathinam VA, Vanaja SK, Waggoner L, Sokolovska A, Becker C, Stuart LM, Leong JM, Fitzgerald KA. TRIF licenses caspase-11-dependent NLRP3 inflammasome activation by gram-negative bacteria. Cell. 2012;150:606–619. doi: 10.1016/j.cell.2012.07.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Clay GM, Sutterwala FS, Wilson ME. NLR proteins and parasitic disease. Immunol Res. 2014;59:142–152. doi: 10.1007/s12026-014-8544-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Bauernfeind FG, Horvath G, Stutz A, Alnemri ES, MacDonald K, Speert D, Fernandes-Alnemri T, Wu J, Monks BG, Fitzgerald KA, Hornung V, Latz E. Cutting edge: NF-kappaB activating pattern recognition and cytokine receptors license NLRP3 inflammasome activation by regulating NLRP3 expression. J Immunol. 2009;183:787–791. doi: 10.4049/jimmunol.0901363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Sutterwala FS, Haasken S, Cassel SL. Mechanism of NLRP3 inflammasome activation. Ann N Y Acad Sci. 2014;1319:82–95. doi: 10.1111/nyas.12458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.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;183:792–796. doi: 10.4049/jimmunol.0900173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Lappas M. NOD1 and NOD2 regulate proinflammatory and prolabor mediators in human fetal membranes and myometrium via nuclear factor-kappa B. Biol Reprod. 2013;89:14. doi: 10.1095/biolreprod.113.110056. [DOI] [PubMed] [Google Scholar]
- 216.Kobayashi KS, Chamaillard M, Ogura Y, Henegariu O, Inohara N, Nunez G, Flavell RA. Nod2-dependent regulation of innate and adaptive immunity in the intestinal tract. Science. 2005;307:731–734. doi: 10.1126/science.1104911. [DOI] [PubMed] [Google Scholar]
- 217.Murray PJ. Beyond peptidoglycan for Nod2. Nat Immunol. 2009;10:1053–1054. doi: 10.1038/ni1009-1053. [DOI] [PubMed] [Google Scholar]
- 218.Fernandes-Alnemri T, Wu J, Yu JW, Datta P, Miller B, Jankowski W, Rosenberg S, Zhang J, Alnemri ES. The pyroptosome: a supramolecular assembly of ASC dimers mediating inflammatory cell death via caspase-1 activation. Cell Death Differ. 2007;14:1590–1604. doi: 10.1038/sj.cdd.4402194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Lappas M. Caspase-1 activation is increased with human labour in foetal membranes and myometrium and mediates infection-induced interleukin-1beta secretion. Am J Reprod Immunol. 2014;71:189–201. doi: 10.1111/aji.12174. [DOI] [PubMed] [Google Scholar]
- 220.Gomez-Lopez N, Guilbert LJ, Olson DM. Invasion of the leukocytes into the fetal-maternal interface during pregnancy. J Leukoc Biol. 2010;88:625–633. doi: 10.1189/jlb.1209796. [DOI] [PubMed] [Google Scholar]
- 221.Gomez-Lopez N, StLouis D, Lehr MA, Sanchez-Rodriguez EN, Arenas-Hernandez M. Immune cells in term and preterm labor. Cell Mol Immunol. 2014;11:571–581. doi: 10.1038/cmi.2014.46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Kayagaki N, Warming S, Lamkanfi M, Vande Walle L, Louie S, Dong J, Newton K, Qu Y, Liu J, Heldens S, Zhang J, Lee WP, Roose-Girma M, Dixit VM. Non-canonical inflammasome activation targets caspase-11. Nature. 2011;479:117–121. doi: 10.1038/nature10558. [DOI] [PubMed] [Google Scholar]
- 223.Broz P, Ruby T, Belhocine K, Bouley DM, Kayagaki N, Dixit VM, Monack DM. Caspase-11 increases susceptibility to Salmonella infection in the absence of caspase-1. Nature. 2012;490:288–291. doi: 10.1038/nature11419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.Wilson KP, Black JA, Thomson JA, Kim EE, Griffith JP, Navia MA, Murcko MA, Chambers SP, Aldape RA, Raybuck SA, et al. Structure and mechanism of interleukin-1 beta converting enzyme. Nature. 1994;370:270–275. doi: 10.1038/370270a0. [DOI] [PubMed] [Google Scholar]
- 225.Dinarello CA. Interleukin-1. Adv Pharmacol. 1994;25:21–51. doi: 10.1016/s1054-3589(08)60429-9. [DOI] [PubMed] [Google Scholar]
- 226.Dinarello CA. The biological properties of interleukin-1. Eur Cytokine Netw. 1994;5:517–531. [PubMed] [Google Scholar]
- 227.Dinarello CA. The interleukin-1 family: 10 years of discovery. FASEB J. 1994;8:1314–1325. [PubMed] [Google Scholar]
- 228.Dinarello CA. Interleukin-1. Cytokine Growth Factor Rev. 1997;8:253–265. doi: 10.1016/s1359-6101(97)00023-3. [DOI] [PubMed] [Google Scholar]
- 229.Dinarello CA. Immunological and inflammatory functions of the interleukin-1 family. Annu Rev Immunol. 2009;27:519–550. doi: 10.1146/annurev.immunol.021908.132612. [DOI] [PubMed] [Google Scholar]
- 230.Creagh EM. Caspase crosstalk: integration of apoptotic and innate immune signalling pathways. Trends Immunol. 2014;35:631–640. doi: 10.1016/j.it.2014.10.004. [DOI] [PubMed] [Google Scholar]
- 231.Romero R, Durum S, Dinarello CA, Oyarzun E, Hobbins JC, Mitchell MD. Interleukin-1 stimulates prostaglandin biosynthesis by human amnion. Prostaglandins. 1989;37:13–22. doi: 10.1016/0090-6980(89)90028-2. [DOI] [PubMed] [Google Scholar]
- 232.Hertelendy F, Romero R, Molnar M, Todd H, Baldassare JJ. Cytokine-initiated signal transduction in human myometrial cells. Am J Reprod Immunol. 1993;30:49–57. doi: 10.1111/j.1600-0897.1993.tb00601.x. [DOI] [PubMed] [Google Scholar]
- 233.Hertelendy F, Rastogi P, Molnar M, Romero R. Interleukin-1beta-induced prostaglandin E2 production in human myometrial cells: role of a pertussis toxin-sensitive component. Am J Reprod Immunol. 2001;45:142–147. doi: 10.1111/j.8755-8920.2001.450304.x. [DOI] [PubMed] [Google Scholar]
- 234.Belt AR, Baldassare JJ, Molnar M, Romero R, Hertelendy F. The nuclear transcription factor NF-kappaB mediates interleukin-1beta-induced expression of cyclooxygenase-2 in human myometrial cells. Am J Obstet Gynecol. 1999;181:359–366. doi: 10.1016/s0002-9378(99)70562-4. [DOI] [PubMed] [Google Scholar]
- 235.Watari M, Watari H, DiSanto ME, Chacko S, Shi GP, Strauss JF., 3rd Pro-inflammatory cytokines induce expression of matrix-metabolizing enzymes in human cervical smooth muscle cells. Am J Pathol. 1999;154:1755–1762. doi: 10.1016/S0002-9440(10)65431-4. [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;165:969–971. doi: 10.1016/0002-9378(91)90450-6. [DOI] [PubMed] [Google Scholar]
- 237.Gravett MG, Witkin SS, Haluska GJ, Edwards JL, Cook MJ, Novy MJ. An experimental model for intraamniotic infection and preterm labor in rhesus monkeys. Am J Obstet Gynecol. 1994;171:1660–1667. doi: 10.1016/0002-9378(94)90418-9. [DOI] [PubMed] [Google Scholar]
- 238.Witkin SS, Gravett MG, Haluska GJ, Novy MJ. Induction of interleukin-1 receptor antagonist in rhesus monkeys after intraamniotic infection with group B streptococci or interleukin-1 infusion. Am J Obstet Gynecol. 1994;171:1668–1672. doi: 10.1016/0002-9378(94)90419-7. [DOI] [PubMed] [Google Scholar]
- 239.Baggia S, Gravett MG, Witkin SS, Haluska GJ, Novy MJ. Interleukin-1 beta intra-amniotic infusion induces tumor necrosis factor-alpha, prostaglandin production, and preterm contractions in pregnant rhesus monkeys. J Soc Gynecol Investig. 1996;3:121–126. doi: 10.1177/107155769600300304. [DOI] [PubMed] [Google Scholar]
- 240.Vadillo-Ortega F, Sadowsky DW, Haluska GJ, Hernandez-Guerrero C, Guevara-Silva R, Gravett MG, Novy MJ. Identification of matrix metalloproteinase-9 in amniotic fluid and amniochorion in spontaneous labor and after experimental intrauterine infection or interleukin-1 beta infusion in pregnant rhesus monkeys. Am J Obstet Gynecol. 2002;186:128–138. doi: 10.1067/mob.2002.118916. [DOI] [PubMed] [Google Scholar]
- 241.Sadowsky DW, Adams KM, Gravett MG, Witkin SS, Novy MJ. Preterm labor is induced by intraamniotic infusions of interleukin-1beta and tumor necrosis factor-alpha but not by interleukin-6 or interleukin-8 in a nonhuman primate model. Am J Obstet Gynecol. 2006;195:1578–1589. doi: 10.1016/j.ajog.2006.06.072. [DOI] [PubMed] [Google Scholar]
- 242.Aagaard K, Ganu R, Ma J, Hu M, Miller L, Jobe AH, Kallapur SG, Chougnet CA. Intraamniotic interleukin-1 (IL1β) induces histological choriamnionitis and alters the microbiome in a primate model of inflammatory preterm birth. Am J Obstet Gynecol. 2014;208:S218. [Google Scholar]
- 243.Prince A, Ma J, Miller L, Hu M, Jobe AH, Chougnet CA, Kallapur SG, Aagaard K. Chorioamnionitis induced by intraamniotic injection of IL1, LPS or Ureaplasma parvum is associated with an altered microbiome in a primate model of inflammatory preterm birth. Am J Obstet Gynecol. 2014;212:S153. [Google Scholar]
- 244.Presicce P, Senthamaraikannan P, Alvarez M, Rueda CM, Cappelletti M, Miller LA, Jobe AH, Chougnet CA, Kallapur SG. Neutrophil recruitment and activation in decidua with intra-amniotic IL-1beta in the preterm rhesus macaque. Biol Reprod. 2015;92:56. doi: 10.1095/biolreprod.114.124420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245.Okamura H, Tsutsi H, Komatsu T, Yutsudo M, Hakura A, Tanimoto T, Torigoe K, Okura T, Nukada Y, Hattori K, et al. Cloning of a new cytokine that induces IFN-gamma production by T cells. Nature. 1995;378:88–91. doi: 10.1038/378088a0. [DOI] [PubMed] [Google Scholar]
- 246.Ushio S, Namba M, Okura T, Hattori K, Nukada Y, Akita K, Tanabe F, Konishi K, Micallef M, Fujii M, Torigoe K, Tanimoto T, Fukuda S, Ikeda M, Okamura H, Kurimoto M. 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;156:4274–4279. [PubMed] [Google Scholar]
- 247.Takeda K, Tsutsui H, Yoshimoto T, Adachi O, Yoshida N, Kishimoto T, Okamura H, Nakanishi K, Akira S. Defective NK cell activity and Th1 response in IL-18-deficient mice. Immunity. 1998;8:383–390. doi: 10.1016/s1074-7613(00)80543-9. [DOI] [PubMed] [Google Scholar]
- 248.Dinarello CA, Novick D, Puren AJ, Fantuzzi G, Shapiro L, Muhl H, Yoon DY, Reznikov LL, Kim SH, Rubinstein M. Overview of interleukin-18: more than an interferon-gamma inducing factor. J Leukoc Biol. 1998;63:658–664. [PubMed] [Google Scholar]
- 249.Dinarello CA. IL-18: A TH1-inducing, proinflammatory cytokine and new member of the IL-1 family. J Allergy Clin Immunol. 1999;103:11–24. doi: 10.1016/s0091-6749(99)70518-x. [DOI] [PubMed] [Google Scholar]
- 250.Dinarello CA. Interleukin-18. Methods. 1999;19:121–132. doi: 10.1006/meth.1999.0837. [DOI] [PubMed] [Google Scholar]
- 251.Novick D, Kim S, Kaplanski G, Dinarello CA. Interleukin-18, more than a Th1 cytokine. Semin Immunol. 2013;25:439–448. doi: 10.1016/j.smim.2013.10.014. [DOI] [PubMed] [Google Scholar]
- 252.Dinarello CA, Novick D, Kim S, Kaplanski G. Interleukin-18 and IL-18 binding protein. Front Immunol. 2013;4:289. doi: 10.3389/fimmu.2013.00289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253.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;110:598–603. [PubMed] [Google Scholar]
