Skip to content
Publicly Available Published by De Gruyter August 4, 2015

Clinical chorioamnionitis at term IV: the maternal plasma cytokine profile

  • Roberto Romero EMAIL logo , Piya Chaemsaithong , Nikolina Docheva , Steven J. Korzeniewski , Adi L. Tarca , Gaurav Bhatti , Zhonghui Xu , Juan P. Kusanovic , Zhong Dong , Noppadol Chaiyasit , Ahmed I. Ahmed , Bo Hyun Yoon , Sonia S. Hassan , Tinnakorn Chaiworapongsa and Lami Yeo

Abstract

Introduction: Fever is a major criterion for clinical chorioamnionitis; yet, many patients with intrapartum fever do not have demonstrable intra-amniotic infection. Some cytokines, such as interleukin (IL)-1, IL-6, interferon-gamma (IFN-γ), and tumor necrosis factor alpha (TNF-α), can induce a fever. The objective of this study was to determine whether maternal plasma concentrations of cytokines could be of value in the identification of patients with the diagnosis of clinical chorioamnionitis at term who have microbial-associated intra-amniotic inflammation.

Methods: A retrospective cross-sectional study was conducted, including patients with clinical chorioamnionitis at term (n=41; cases) and women in spontaneous labor at term without clinical chorioamnionitis (n=77; controls). Women with clinical chorioamnionitis were classified into three groups according to the results of amniotic fluid culture, broad-range polymerase chain reaction coupled with electrospray ionization mass spectrometry (PCR/ESI-MS), and amniotic fluid IL-6 concentration: 1) no intra-amniotic inflammation; 2) intra-amniotic inflammation without detectable microorganisms; or 3) microbial-associated intra-amniotic inflammation. The maternal plasma concentrations of 29 cytokines were determined with sensitive and specific V-PLEX immunoassays. Nonparametric statistical methods were used for analysis, adjusting for a false discovery rate of 5%.

Results: 1) The maternal plasma concentrations of pyrogenic cytokines (IL-1β, IL-2, IL-6, IFN-γ, and TNF-α) were significantly higher in patients with clinical chorioamnionitis at term than in those with spontaneous term labor without clinical chorioamnionitis; 2) the maternal plasma concentrations of cytokines were not significantly different among the three subgroups of patients with clinical chorioamnionitis (intra-amniotic inflammation with and without detectable bacteria and those without intra-amniotic inflammation); and 3) among women with the diagnosis of clinical chorioamnionitis, but without evidence of intra-amniotic inflammation, the maternal plasma concentrations of pyrogenic cytokines were significantly higher than in patients with spontaneous labor at term. These observations suggest that a fever can be mediated by increased circulating concentrations of these cytokines, despite the absence of a local intra-amniotic inflammatory response.

Conclusions: 1) The maternal plasma concentrations of pyrogenic cytokines (e.g. IL-1β, IL-2, IL-6, IFN-γ, and TNF-α) are higher in patients with intra-partum fever and the diagnosis of clinical chorioamnionitis at term than in those in spontaneous labor at term without a fever; and 2) maternal plasma cytokine concentrations have limited value in the identification of patients with bacteria in the amniotic cavity. Accurate assessment of the presence of intra-amniotic infection requires amniotic fluid analysis.

Introduction

Clinical chorioamnionitis is characterized by maternal fever accompanied by at least two of the following signs: maternal or fetal tachycardia, maternal leukocytosis, uterine tenderness, or foul-smelling amniotic fluid [1–13]. This pregnancy complication is associated with adverse maternal [14–19], fetal, and neonatal/infant outcomes [20–40]. Most of the signs of clinical chorioamnionitis (except for fetal tachycardia and foul-smelling amniotic fluid) are thought to reflect a maternal inflammatory response to microbial invasion of the amniotic cavity (MIAC) [1, 7]. Fever, the cardinal sign of clinical chorioamnionitis, is mediated by the effect of pyrogenic cytokines [41–43], which include interleukin (IL)-1 [43–51], IL-2 [52–54], IL-6 [55–59], interferon-gamma (IFN-γ) [60, 61], and tumor necrosis factor-alpha (TNF-α) [54, 62–65]. Some of these cytokines have primary pyrogenic effects (i.e., IL-1 [43–51]), or TNF-α [54, 62–65], while others induce fever indirectly (i.e., IL-2 [52–54]). Fever can be generated independently of IL-1 and TNF-α by the engagement of Toll-like receptors [66–70]. These different pathways converge in the production of cyclooxygenase-2 [69–73] and prostaglandin E2 [74–80], which bind to prostanoid receptors in the hypothalamus [43, 70]. Thermosensitive neurons in the thermoregulatory center then raise the hypothalamic thermostatic set point [69, 81, 82].

Many cases of intra-partum fever are not related to infection [9, 12, 83–91]. We recently reported that only 54% of women with clinical chorioamnionitis (all had fever) at term have microorganisms isolated from the amniotic cavity; 24% had intra-amniotic inflammation without demonstrable bacteria and 22% had no intra-amniotic inflammation [9]. Epidural analgesia can cause maternal hyperthermia in the absence of intra-amniotic infection [83, 86, 92–107]. Therefore, when patients have a fever, the accurate diagnosis of infection is challenging, but important. We recently reported that the signs used in the diagnosis of clinical chorioamnionitis do not accurately identify patients with proven intra-amniotic infection [4].

The objective of this study was to determine if the maternal plasma concentration of cytokines could distinguish patients with clinical chorioamnionitis with and without proven intra-amniotic infection.

Materials and methods

Study population

A retrospective cross-sectional case-control study was conducted by searching the clinical database and Bank of Biological Samples of Wayne State University, the Detroit Medical Center, and the Perinatology Research Branch (NICHD/NIH/DHHS). The inclusion criteria were: 1) singleton gestation; 2) gestational age of ≥37 weeks; and 3) the absence of fetal malformations.

Cases consisted of women with clinical chorioamnionitis at term (n=41). These women had also been included in a prior study that provided a detailed description of sample collection, microbiological studies, and determination of amniotic fluid IL-6 concentrations using a sensitive and specific enzyme-linked immunosorbent assay [9]. Controls were women with spontaneous term labor without clinical and histological chorioamnionitis (n=77; gestational age of 37–42 weeks).

All patients provided written informed consent, and the use of biological specimens as well as clinical and ultrasound data for research purposes was approved by the Institutional Review Boards of NICHD, Wayne State University, and Sótero del Río Hospital, Santiago, Chile.

Clinical definitions

MIAC was defined according to the results of amniotic fluid culture and broad-range polymerase chain reaction coupled with electrospray ionization mass spectrometry (PCR/ESI-MS), (Ibis® Technology, Athogen, Carlsbad, CA, USA) [108–111]. Intra-amniotic inflammation was diagnosed when the amniotic fluid IL-6 concentration was ≥2.6 ng/mL [10, 112–125]. Based on the results of amniotic fluid cultures, PCR/ESI-MS, and amniotic fluid concentrations of IL-6, patients with clinical chorioamnionitis at term were classified as having: 1) no intra-amniotic inflammation; 2) intra-amniotic inflammation without detectable bacteria (an elevated amniotic fluid IL-6 concentration without evidence of bacteria using cultivation and molecular methods); or 3) microbial-associated intra-amniotic inflammation (combination of MIAC and intra-amniotic inflammation). Clinical chorioamnionitis was diagnosed by the presence of maternal fever (temperature >37.8°C) accompanied by two or more of the following criteria: 1) maternal tachycardia (heart rate >100 beats/min); 2) uterine tenderness; 3) foul-smelling amniotic fluid; 4) fetal tachycardia (heart rate >160 beats/min); and 5) maternal leukocytosis (leukocyte count >15,000 cells/mm3) [3, 4, 7, 8, 126, 127].

Controls were women with spontaneous labor without clinical or histological chorioamnionitis, who delivered a normal term (≥37 weeks) infant whose birthweight was appropriate for the gestational age (10th–90th percentile) [128, 129].

Acute histologic chorioamnionitis was diagnosed based on the presence of inflammatory cells in the chorionic plate and/or chorioamniotic membranes [130–139]. Acute funisitis was diagnosed based on the presence of neutrophils in the wall of the umbilical vessels and/or Wharton’s jelly, using criteria previously described [130, 140–145]. Acute inflammatory lesions of the placenta or placental lesions consistent with amniotic fluid infection were defined by the presence of acute histologic chorioamnionitis and/or acute funisitis [130].

Sample collection and cytokines/chemokines immunoassays

Maternal blood samples were collected at admission during labor in both cases and controls. These samples were placed into tubes containing EDTA, then centrifuged for 10 min at 4°C and stored at −70°C. Laboratory personnel were blinded to clinical diagnosis. The maternal plasma concentrations of the following 29 cytokines/chemokines were determined with sensitive and specific V-PLEX immunoassays (Meso Scale Discovery, Gaithersburg, MD, USA) [Pro-inflammatory cytokines: IFN-γ, IL-1α, IL-1β, IL-2, IL-6, IL-7, IL-12p70, IL-12/IL-23p40, IL-15, IL-16, IL-17α, TNF-α, TNF-β, vascular endothelial growth factor (VEGF), granulocyte macrophage colony-stimulating factor (GM-CSF); anti-inflammatory cytokines: IL-4, IL-5, IL-10, IL-13; and chemokines: IL-8, thymus and activation-regulated chemokine (TARC), eotaxin, eotaxin-3, macrophage-derived chemokine (MDC), macrophage inflammatory protein (MIP)-1α, MIP-1β, monocyte chemoattractant protein (MCP)-1, MCP-4, C-X-C motif chemokine 10 (CXCL-10) or interferon gamma-induced protein 10 (IP-10)]. Briefly, 50 μL of a maternal blood sample or calibrator were dispensed into separate wells of the plates and incubated for 2 h with vigorous shaking at room temperature. The samples and calibrators were discarded, and the plates were washed three times with phosphate-buffered saline and 0.05% Tween-20, followed by an addition of 25 μL of the 1×detection antibody solution into each well. Plates were then incubated for 2 h with vigorous shaking at room temperature. The detection antibody was removed, and the plates were washed three times. One hundred and fifty microliters of 2×Read Buffer T were added to each well, and the signals were read by the SECTOR® Imager 2400 (Meso Scale Discovery). Standard curves were generated, and the assay values of the samples were interpolated from the curves. The assay characteristics are described in the Supplementary Table. The coefficient of variation was ≤14.6% for 20 of the 29 analytes. For samples with concentrations below the limits of detection, missing values were replaced with 99% of the lowest detectable concentration.

Statistical analysis

For demographics data analysis: the Kolmogorov-Smirnov test was used to test whether the distribution of continuous variables was normal. Chi-square and Fisher’s exact tests were used for comparisons of proportions. Kruskal-Wallis and the Mann-Whitney U-tests were used to compare median concentrations of analytes between and among groups. Statistical analysis of demographics data was performed using SPSS 19 (IBM Corp, Armonk, NY, USA). A P-value <0.05 was considered statistically significant.

Comparison of analyte concentrations determined by multiplex assay was restricted to the analytes that were detected in a number of samples larger than one-half of the size of the smallest group. Statistical analysis was performed using the Wilcoxon rank-sum test and R statistical environment [146]. Nominal P-values were adjusted using the Benjamini and Hochberg method [147], controlling the false discovery rate at 5%.

Results

Characteristics of the study population

Clinical characteristics of the study population are displayed in Table 1. Patients with clinical chorioamnionitis had a significantly lower median maternal age than those with spontaneous term labor without clinical chorioamnionitis (P=0.007). Otherwise, there were no significant differences in the clinical characteristics between these two groups. All patients with clinical chorioamnionitis received epidural analgesia.

Table 1

Clinical characteristics of the study population.

Term in labor (n=77)Clinical chorioamnionitis at term (n=41)P-value
Maternal age (years)25 (19.5–30)23.7 (21.6–24.6)0.007
Body mass index (kg/m2)23.9 (21.3–26.1)23.7 (21.6–24.6)0.53
Amniotic fluid glucose (mg/dL)N/A9 (9–9)N/A
Amniotic fluid white blood cell count (cell/mm3)N/A58 (5–695)N/A
Gestational age at amniocentesis (weeks)N/A39.6 (38.9–40.7)N/A
Gestational age at delivery (weeks)39.6 (38.9–40.7)40 (39.1–40.7)0.33
Birthweight (g)3450 (3200–3665)3550 (3200–3780)0.39

Data presented as median (interquartile range). N/A=not applicable.

When classified according to the presence or absence of both intra-amniotic inflammation and microorganisms (by amniotic fluid culture and PCR/ESI-MS), 58.5% (24/41) of the cases had microbial-associated intra-amniotic inflammation, 22% (9/41) had intra-amniotic inflammation without demonstrable bacteria, and 19.5% (8/41) had no evidence of intra-amniotic inflammation. The microorganisms identified in the amniotic fluid were previously reported [9]. A placental histopathology report was not available for one patient with clinical chorioamnionitis. Acute inflammatory lesions of the placenta were found in 62.5% (25/40) of patients with clinical chorioamnionitis. Among cases with intra-amniotic inflammation with and without demonstrable microorganisms, 79.2% (19/24) and 62.5% (5/8) had acute inflammatory lesions of the placenta, respectively. Only 12.5% (1/8) of cases without intra-amniotic inflammation had acute inflammatory lesions of the placenta.

Maternal plasma cytokine and chemokine concentrations in clinical chorioamnionitis at term and spontaneous term labor without clinical chorioamnionitis

Clinical chorioamnionitis vs. spontaneous labor at term without clinical chorioamnionitis

The maternal plasma cytokine and chemokine concentrations of women with spontaneous labor at term and those with clinical chorioamnionitis at term are described in Table 2. Women with clinical chorioamnionitis had significantly higher median maternal plasma concentrations of 10/19 inflammation-related cytokines than controls (after correcting for false discovery). The fold difference in median maternal plasma concentrations of IL-2, IL-6, IL-1β, IL-17α, IFN-γ, TNF-β, TNF-α, IL-15, IL-12/IL-23p40, IL-10, and IL-5 ranged from 1.17 to 13.13. IL-2 had the greatest fold change of 13.13 (Table 2). Importantly, the maternal plasma concentrations of several pyrogenic cytokines (IL-6, IL-2, TNF-α, and IL-1β) were higher in women with clinical chorioamnionitis (all with a fever) than in those with spontaneous labor at term without clinical chorioamnionitis.

Table 2

Concentrations of maternal plasma cytokines and chemokines in term in labor vs. clinical chorioamnionitis at term.

Analytes (pg/mL)Term in labor median (IQR) (n=77)Clinical chorioamnionitis at term median (IQR) (n=41)Fold changeAdjusted P-value
Pro-inflammatory cytokines
 IL-20.01 (0.01–0.12)0.13 (0.06–0.22)13.130.001
 IL-62.53 (1.64–4.08)10.94 (5.36–22.08)4.32<0.0001
 IL-1β0.12 (0.01–0.31)0.21 (0.11–0.34)1.750.03
 IL-17α0.55 (0.34–0.71)0.8 (0.63–1.32)1.450.002
 TNF-β0.07 (0.02–0.11)0.1 (0.05–0.15)1.430.04
 TNF-α2.06 (1.62–2.48)2.84 (1.9–3.89)1.380.008
 IL-152.05 (1.71–2.42)2.66 (2.25–3.67)1.30<0.0001
 IL-12/IL-23p4076.89 (62.43–98.69)89.81 (70.86–112.47)1.170.046
 IFN-γ5.24 (3.26–8.34)6.15 (3.66–19.81)1.170.09
 IL-1α2.61 (1.65–4.27)2.59 (1.94–3.52)0.990.78
 IL-12p700.07 (0.01–0.13)0.06 (0.01–0.19)0.860.24
 GM-CSF0.02 (0.02–0.09)0.02 (0.02–0.12)1.000.88
 IL-16158.99 (129.21–224.54)173.57 (127.9–220.27)1.100.93
 IL-73.51 (2.04–6.51)3.41 (2.15–7.64)0.970.88
 VEGF14.36 (10.67–23.71)13.48 (9.41–17.25)0.940.24
Anti-inflammatory cytokines
 IL-100.41 (0.25–0.86)2.04 (0.32–5)4.970.0006
 IL-50.36 (0.21–0.57)0.48 (0.27–0.83)1.330.04
 IL-130.65 (0.28–1.01)0.65 (0.28–1.04)1.000.88
 IL-40.03 (0.01–0.03)0.03 (0.01–0.04)1.000.97
Chemokines
 MCP-157.38 (51.48–70.38)73.61 (53.78–104.7)1.280.03
 MIP-1α10.41 (7.35–12.25)13.27 (9.29–17.36)1.270.02
 IL-83.0 (1.9–5.09)3.67 (3.29–7.47)1.220.03
 Eotaxin56.8 (43.72–77.74)46.93 (31.29–56.09)0.830.008
 MCP-443.79 (29.91–73.38)30.48 (24.06–43.03)0.700.02
 Eotaxin-319.36 (14.89–25.59)4.67 (0.64–13.1)0.24<0.0001
 MDC520.65 (443.02–644.53)515.51 (403.33–637.29)0.990.76
 CXCL-10 or IP-10238.32 (172.2–327.48)242.38 (179.13–485.75)1.020.37
 TARC34.99 (18.74–52.3)35.59 (22.81–52.87)1.020.78
 MIP-1β59.25 (45.93–78.56)59.33 (41.53–80.4)1.0010.93

IQR=interquartile range, IL=interleukin, TNF=tumor necrosis factor, IFN-γ=interferon gamma, GM-CSF=granulocyte macrophage colony-stimulating factor, VEGF=vascular endothelial growth factor, MCP=monocyte chemoattractant protein, MIP=macrophage inflammatory protein, MDC=macrophage-derived chemokine, CXCL-10=C-X-C motif chemokine 10, IP-10=interferon gamma-induced protein 10, TARC=thymus and activation-regulated chemokine. The units of all analytes are pg/mL.

Patients with clinical chorioamnionitis at term had significantly higher median maternal plasma concentrations of three of the ten chemokines (MCP-1, MIP-1α, and IL-8) than controls, with a fold difference in the median of approximately 1.2 for the three of them. Eotaxin, MCP-4, and eotaxin-3 were significantly lower in women with clinical chorioamnionitis than in controls, and fold differences in the median were 0.83, 0.70, and 0.24, respectively (Table 2).

Maternal plasma cytokine concentrations in patients with clinical chorioamnionitis according to the presence of inflammation and bacteria in the amniotic fluid

In this study, six of the ten pro-inflammatory cytokines (IL-6, IL-2, IFN-γ, TNF-α, IL-17α, and IL-15) had significantly higher concentrations in cases without intra-amniotic inflammation than in controls, and fold differences in the median ranged from 1.82 to 18.69 (Table 3). IL-2 and IFN-γ had fold changes >15 when comparing cases without intra-amniotic inflammation to women in spontaneous labor at term (Figures 1 and 2). IL-6 was the only pro-inflammatory cytokine that was significantly higher in cases with intra-amniotic inflammation without demonstrable microorganisms than in those with spontaneous labor at term (Figure 3).

Table 3

Concentrations of maternal plasma cytokines and chemokines in the subgroups of clinical chorioamnionitis and term in labor.

Analytes (pg/mL)Term in labor (controls) (n=77)Clinical chorioamnnionitis at term (n=41)
Without intra-amniotic inflammation (n=8)With intra-amniotic inflammation without demonstrable bacteria (n=9)With microbial-associated intra-amniotic inflammation (n=24)
Median (IQR)Median (IQR)Fold change (compared to term in labor)Adjusted P-value (compared to term in labor)Median (IQR)Fold change (compared to term in labor)Adjusted P-value (compared to term in labor)Adjusted P-value (compared to without intra-amniotic inflammation)Median (IQR)Fold change (compared to term in labor)Adjusted P-value (compared to term in labor)Adjusted P-value (compared to without intra-amniotic inflammation)Adjusted P-value (compared to with intra-amniotic inflammation without demonstrable bacteria)
Pro-inflammatory cytokines
 IL-62.53 (1.64–4.08)6.72 (5.05–12.91)2.660.0089.75 (6.14–22.92)3.850.0090.7815.58 (5.54–22.34)6.16<0.00010.700.92
 IL-20.01 (0.01–0.12)0.18 (0.14–0.24)18.690.0080.12 (0.05–0.14)12.120.270.740.13 (0.02–0.21)13.130.040.400.92
 IFN-γ5.24 (3.26–8.34)88.73 (13.78–135.99)16.930.0086.15 (3.83–20.57)1.170.450.745.04 (3.31–7.92)0.960.820.080.87
 TNF-α2.06 (1.62–2.48)4.46 (3.2–5.79)2.170.042.98 (2.2–3.48)1.450.050.742.42 (1.55–3.3)1.170.260.260.87
 IL-17α0.55 (0.34–0.71)1.32 (0.71–3.87)2.400.040.83 (0.68–1.32)1.510.050.740.74 (0.48–1.07)1.350.090.400.87
 IL-152.05 (1.71–2.42)3.73 (2.93–4.51)1.820.0082.75 (2.25–3.33)1.340.070.742.58 (2.23–3.18)1.260.0020.400.97
 TNF-β0.07 (0.02–0.11)0.1 (0.07–0.11)1.430.510.13 (0.08–0.15)1.860.270.740.1 (0.05–0.15)1.430.110.840.92
 IL-12/IL-23p4076.89 (62.43–98.69)112.2 (87.39–147.02)1.460.0796.63 (58.76–128.84)1.260.300.7483.45 (70.45–101.04)1.090.370.310.87
 IL-16158.99 (129.21–224.54)118.8 (89.15–155.07)0.750.13133.63 (128.67–191.54)0.840.530.74187.65 (159.96–229.7)1.180.310.260.87
 IL-1α2.61 (1.65–4.27)2.65 (1.81–3.68)1.020.912.7 (2.25–5.08)1.030.530.742.57 (2.07–3.19)0.980.821.000.87
 GM-CSF0.02 (0.02–0.09)0.02 (0.02–0.13)1.000.830.02 (0.02–0.17)1.000.530.880.02 (0.02–0.06)1.000.880.900.87
 IL-73.51 (2.04–6.51)7.79 (3.83–9.14)2.220.154.58 (3.68–7.64)1.30.270.742.72 (1.55–5.07)0.780.270.260.46
 IL-1β0.12 (0.01–0.31)0.21 (0.1–0.4)1.790.320.31 (0.12–0.35)2.580.270.740.21 (0.11–0.28)1.710.111.000.87
 IL-12p700.07 (0.01–0.13)0.13 (0.04–0.24)1.930.270.13 (0.04–0.21)1.860.150.960.05 (0.01–0.19)0.640.930.300.87
 VEGF14.36 (10.67–23.71)13.98 (10.43–17.51)0.970.6116.8 (12.11–26.54)1.170.980.7412.21 (9.25–15.12)0.850.190.840.87
Anti-inflammatory cytokines
 IL-100.41 (0.25–0.86)1.9 (1.3–5.42)4.630.023.19 (0.92–3.8)7.780.050.961.75 (0.3–5.49)4.270.050.700.95
 IL-50.36 (0.21–0.57)0.28 (0.2–0.47)0.790.830.38 (0.27–0.54)1.060.610.740.59 (0.4–0.91)1.640.010.260.87
 IL-40.03 (0.01–0.03)0.02 (0.01–0.02)0.670.360.02 (0.01–0.03)0.670.810.740.03 (0.01–0.04)1.000.610.400.87
 IL-130.65 (0.28–1.01)0.28 (0.28–0.41)0.430.070.28 (0.28–0.78)0.430.450.740.93 (0.28–1.37)1.420.270.190.87
Chemokines
 CXCL-10 or IP-10238.32 (172.2–327.48)503.96 (284.8–998.46)2.110.02242.6 (179.13–557.89)1.020.460.74209.99 (169.03–296.8)0.880.700.080.87
 Eotaxin-319.36 (14.89–25.59)10.12 (0.64–22.92)0.520.224.67 (0.64–9.54)0.240.0080.743.93 (0.64–12.97)0.200.00030.840.92
 MCP-443.79 (29.91–73.38)33.11 (25.28–52.48)0.760.5038.85 (34.99–46.92)0.890.980.7426.05 (22.15–37.84)0.590.0030.400.41
 MCP-157.38 (51.48–70.38)94.93 (65.69–130.46)1.650.0773.01 (55.47–91.16)1.270.270.7469.27 (52.17–99.48)1.210.260.470.92
 MIP-1β59.25 (45.93–78.56)68.62 (52.61–165.9)1.160.2957.28 (44.39–69.42)0.970.810.7460.51 (38.55–80.72)1.020.820.400.92
 MIP-1α10.41 (7.35–12.25)20.64 (9.81–25.13)1.980.1011.84 (8.95–13.77)1.140.410.7412.63 (9.4–15.2)1.210.110.400.92
 IL-83 (1.9–5.09)4 (2.77–7.97)1.330.443.58 (3.43–4.71)1.190.270.963.61 (3.14–8.6)1.200.091.000.92
 TARC34.99 (18.74–52.3)26.89 (21.74–34.94)0.770.5042.57 (20.45–73.51)1.220.530.7436.75 (24.22–55.2)1.050.610.400.92
 MDC520.65 (443.02–644.53)526.94 (471.77–590.9)1.010.73463.75 (421.72–732.89)0.890.980.78512.05 (396.98–639.64)0.980.681.000.87
 Eotaxin56.8 (43.72–77.74)47.95 (41.02–58.23)0.840.2740.04 (31.29–54.04)0.700.100.7446.68 (32.87–57.96)0.820.081.000.92

IQR=interquartile range, IL=interleukin, TNF=tumor necrosis factor, IFN-γ=interferon gamma, GM-CSF=granulocyte macrophage colony-stimulating factor, VEGF=vascular endothelial growth factor, MCP=monocyte chemoattractant protein, MIP=macrophage inflammatory protein, MDC=macrophage-derived chemokine, CXCL-10=C-X-C motif chemokine 10, IP-10=interferon gamma-induced protein 10, TARC=thymus and activation-regulated chemokine. The units of all analytes are pg/mL.

Figure 1: The maternal plasma concentrations of interleukin (IL)-2 in patients with spontaneous term labor without clinical chorioamnionitis (n=77, controls), clinical chorioamnionitis without intra-amniotic inflammation (n=8), clinical chorioamnionitis with intra-amniotic inflammation without demonstrable bacteria (n=9), and clinical chorioamnionitis with microbial-associated intra-amniotic inflammation (n=24). The median (interquartile range) maternal plasma concentrations of IL-2 are 0.01 (0.01–0.12) pg/mL (term in labor), 0.18 (0.14–0.24) pg/mL (clinical chorioamnionitis without intra-amniotic inflammation), 0.12 (0.05–0.14) pg/mL (clinical chorioamnionitis with intra-amniotic inflammation without demonstrable bacteria), and 0.13 (0.02–0.21) pg/mL (clinical chorioamnionitis with microbial-associated intra-amniotic inflammation). LOD=limit of detection.
Figure 1:

The maternal plasma concentrations of interleukin (IL)-2 in patients with spontaneous term labor without clinical chorioamnionitis (n=77, controls), clinical chorioamnionitis without intra-amniotic inflammation (n=8), clinical chorioamnionitis with intra-amniotic inflammation without demonstrable bacteria (n=9), and clinical chorioamnionitis with microbial-associated intra-amniotic inflammation (n=24). The median (interquartile range) maternal plasma concentrations of IL-2 are 0.01 (0.01–0.12) pg/mL (term in labor), 0.18 (0.14–0.24) pg/mL (clinical chorioamnionitis without intra-amniotic inflammation), 0.12 (0.05–0.14) pg/mL (clinical chorioamnionitis with intra-amniotic inflammation without demonstrable bacteria), and 0.13 (0.02–0.21) pg/mL (clinical chorioamnionitis with microbial-associated intra-amniotic inflammation). LOD=limit of detection.

Figure 2: The maternal plasma concentrations of interferon (IFN)-γ in patients with spontaneous term labor without clinical chorioamnionitis (n=77, controls), clinical chorioamnionitis without intra-amniotic inflammation (n=8), clinical chorioamnionitis with intra-amniotic inflammation without demonstrable bacteria (n=9), and clinical chorioamnionitis with microbial-associated intra-amniotic inflammation (n=24). The median (interquartile range) maternal plasma concentrations of IFN-γ are 5.24 (3.26–8.34) pg/mL (term in labor), 88.73 (13.78–135.99) pg/mL (clinical chorioamnionitis without intra-amniotic inflammation), 6.15 (3.83–20.57) pg/mL (clinical chorioamnionitis with intra-amniotic inflammation without demonstrable bacteria), and 5.04 (3.31–7.92) pg/mL (clinical chorioamnionitis with microbial-associated intra-amniotic inflammation).
Figure 2:

The maternal plasma concentrations of interferon (IFN)-γ in patients with spontaneous term labor without clinical chorioamnionitis (n=77, controls), clinical chorioamnionitis without intra-amniotic inflammation (n=8), clinical chorioamnionitis with intra-amniotic inflammation without demonstrable bacteria (n=9), and clinical chorioamnionitis with microbial-associated intra-amniotic inflammation (n=24). The median (interquartile range) maternal plasma concentrations of IFN-γ are 5.24 (3.26–8.34) pg/mL (term in labor), 88.73 (13.78–135.99) pg/mL (clinical chorioamnionitis without intra-amniotic inflammation), 6.15 (3.83–20.57) pg/mL (clinical chorioamnionitis with intra-amniotic inflammation without demonstrable bacteria), and 5.04 (3.31–7.92) pg/mL (clinical chorioamnionitis with microbial-associated intra-amniotic inflammation).

Figure 3: The maternal plasma concentrations of interleukin (IL)-6 in patients with spontaneous term labor without clinical chorioamnionitis (n=77, controls), clinical chorioamnionitis without intra-amniotic inflammation (n=8), clinical chorioamnionitis with intra-amniotic inflammation without demonstrable bacteria (n=9), and clinical chorioamnionitis with microbial-associated intra-amniotic inflammation (n=24). The median (interquartile range) maternal plasma concentrations of IL-6 are 2.53 (1.64–4.08) pg/mL (term in labor), 6.72 (5.05–12.91) pg/mL (clinical chorioamnionitis without intra-amniotic inflammation), 9.75 (6.14–22.92) pg/mL (clinical chorioamnionitis with intra-amniotic inflammation without demonstrable bacteria), and 15.58 (5.54–22.34) pg/mL (clinical chorioamnionitis with microbial-associated intra-amniotic inflammation).
Figure 3:

The maternal plasma concentrations of interleukin (IL)-6 in patients with spontaneous term labor without clinical chorioamnionitis (n=77, controls), clinical chorioamnionitis without intra-amniotic inflammation (n=8), clinical chorioamnionitis with intra-amniotic inflammation without demonstrable bacteria (n=9), and clinical chorioamnionitis with microbial-associated intra-amniotic inflammation (n=24). The median (interquartile range) maternal plasma concentrations of IL-6 are 2.53 (1.64–4.08) pg/mL (term in labor), 6.72 (5.05–12.91) pg/mL (clinical chorioamnionitis without intra-amniotic inflammation), 9.75 (6.14–22.92) pg/mL (clinical chorioamnionitis with intra-amniotic inflammation without demonstrable bacteria), and 15.58 (5.54–22.34) pg/mL (clinical chorioamnionitis with microbial-associated intra-amniotic inflammation).

Patients with clinical chorioamnionitis and microbial-associated intra-amniotic inflammation had significantly higher median maternal plasma concentrations of IL-2, IL-6, and IL-15 than those with spontaneous labor at term, with fold changes of 13.13, 6.16, and 1.26, respectively (Table 3). IFN-γ had a fold change >15, and the difference was detected only in the subgroup of clinical chorioamnionitis without intra-amniotic inflammation, but not in those with intra-amniotic inflammation, when compared to controls. In contrast, maternal IL-6 concentrations were significantly higher in all subgroups with clinical chorioamnionitis than in the control group (i.e., clinical chorioamnionitis without intra-amniotic inflammation: fold change 2.66, P=0.008; clinical chorioamnionitis with intra-amniotic inflammation without demonstrable bacteria: fold change 3.85, P=0.009; clinical chorioamnionitis with microbial-associated intra-amniotic inflammation: fold change 6.16, P=0.0000025; Table 3; Figure 3).

Among the four anti-inflammatory cytokines, the median maternal plasma IL-10 concentration was four times greater in women with chorioamnionitis without intra-amniotic inflammation than in controls (Table 3). However, the difference did not reach statistical significance in cases with intra-amniotic inflammation with or without detectable microorganisms, when each one was compared to controls (Table 3).

Patients with clinical chorioamnionitis without intra-amniotic inflammation had significantly higher maternal plasma CXCL-10 (or IP-10) concentrations than controls (fold change=2.11; P=0.02) (Table 3). Eotaxin-3 is the only chemokine that had a significantly lower concentration in cases of intra-amniotic inflammation without demonstrable microorganisms than in controls (fold change=0.24; P=0.008). The concentrations of MCP-4 and eotaxin-3 were significantly lower in cases with microbial-associated intra-amniotic inflammation than in controls (eotaxin-3: fold change=0.20, P=0.003; MCP-4: fold change=0.59, P=0.003; Table 3). Overall, no differences could be detected in the maternal plasma concentrations of cytokines among the subgroups of patients with clinical chorioamnionitis at term (Table 3).

The supplementary material contains the scatterplots of maternal plasma concentrations of cytokines/chemokines among the different groups (Supplementary Figures 1–3).

Discussion

Principal findings of this study

1) The maternal plasma concentrations of pyrogenic cytokines (IL-1β, IL-2, IL-6, IFN-γ, and TNF-α) were significantly higher in patients with clinical chorioamnionitis at term than in those with spontaneous term labor without clinical chorioamnionitis; 2) the maternal plasma concentrations of cytokines were not significantly different among the three subgroups of patients with clinical chorioamnionitis (intra-amniotic inflammation with and without detectable bacteria, and those without intra-amniotic inflammation); and 3) among women with the diagnosis of clinical chorioamnionitis but without evidence of intra-amniotic inflammation, the maternal plasma concentrations of pyrogenic cytokines were significantly higher than in women with spontaneous labor at term. These observations suggest that the intra-amniotic inflammatory response observed in some patients with clinical chorioamnionitis is not reflected in the maternal circulation.

Maternal plasma cytokines in patients with clinical chorioamnionitis

Intra-amniotic inflammation and bacteria are frequently present in patients with clinical chorioamnionitis at term [1, 3, 7, 9, 12] and other complications of pregnancy, such as preterm labor [13, 116, 119, 148–164], preterm prelabor rupture of the membranes (PPROM) [109, 117, 121, 135, 165–176], cervical insufficiency [177–179], and in patients with an asymptomatic short cervix [118, 180–185]. Microorganisms and their products can initiate an inflammatory response by the engagement of pattern recognition receptors [163, 172, 186–200] and the production of cytokines [112, 122, 151, 201–224], chemokines [213, 215, 216, 219, 223, 225–244], prostaglandins [245–261], and proteases [142, 145, 262–277]. If microorganisms in the amniotic cavity reach the fetus, this can result in a fetal systemic inflammatory response [278, 279], which is associated with multi-organ involvement [141, 278, 280–289] and preterm delivery [290].

We report herein, for the first time, a systematic study of the maternal cytokine profile using multiplex immunoassays in patients with clinical chorioamnionitis at term stratified according to the status of the amniotic cavity (presence/absence of inflammation and bacteria). The results showed that clinical chorioamnionitis at term is associated with higher maternal plasma pro- and anti-inflammatory cytokines, as well as some chemokines, than in spontaneous labor at term without a fever. The greatest fold change was observed in the maternal plasma concentration of IL-2, followed by IL-6 (13.13 vs. 4.32, respectively). Other cytokines and chemokines had fold changes of a lesser magnitude (<5).

IL-2 is a lymphocyte growth factor involved in the differentiation of CD4+ and CD8+ cells, as well as immune tolerance [291–296], and it has been implicated in the pathogenesis of sepsis [297–300]. Therapeutic administration of IL-2 to patients with cancer can lead to a sepsis-like syndrome [301–303]. In vitro and in vivo studies have shown that IL-2 up-regulates the production of IL-1β and TNF-α in peripheral blood mononuclear cells [52, 304], which are also involved in the pathogenesis of sepsis [305, 306]. Moreover, IL-2 administration is associated with the activation of complement [307–311], neutrophils [311, 312], and the coagulation system [313]. Circulating IL-2 receptor concentrations can also predict the likelihood of sepsis in both newborns [314] and adults [315–317].

Maternal inflammatory response in clinical chorioamnionitis without intra-amniotic inflammation

We have previously reported that a subset of patients with clinical chorioamnionitis at term did not have intra-amniotic inflammation [9], and that their amniotic fluid inflammatory response was similar to that of women with spontaneous labor at term without fever or complications [12]. In the current study, patients with clinical chorioamnionitis without intra-amniotic inflammation had higher concentrations of several maternal plasma cytokines (e.g., IL-2, IFN-γ, IL-6, TNF-α, IL-17α, and IL-15) than patients with spontaneous term labor. Of interest, the maternal plasma IL-2 and IFN-γ concentrations had a fold change >15 in patients with clinical chorioamnionitis without intra-amniotic inflammation when compared to controls. Of note, IFN-γ was significantly higher only in patients with clinical chorioamnionitis without intra-amniotic inflammation, but not in those with intra-amniotic inflammation either with or without demonstrable bacteria when each was compared to controls (spontaneous labor at term without a fever).

IFN-γ is a potent pro-inflammatory cytokine with anti-viral properties [318, 319]. The administration of IFN-γ in patients with cancer can induce a fever [320, 321]. However, the pyrogenic effect appears to be due to the induction of IL-1 and TNF-α [60, 322, 323]. The administration of IFN-γ to human subjects initiates fever 3–4 h after injection [53]. Therefore, the fever of some patients with the diagnosis of clinical chorioamnionitis may be mediated by IFN-γ or other pyrogenic cytokines.

Maternal plasma cytokines in patients with intra-amniotic inflammation with or without bacteria in the amniotic cavity

We observed a dramatic elevation in the concentration of maternal plasma IL-6 (fold change >5) in patients with bacteria in the amniotic cavity. This is not unexpected, as this cytokine is a major mediator of the host response to infection and tissue damage [324–328]. Bacterial products are known to up-regulate the expression of IL-6 [329, 330] and so are other inflammatory cytokines elevated in cases of intra-amniotic infection [329, 330]. Despite the large magnitude of the elevation in maternal plasma IL-6, we are not persuaded that this information can be used to distinguish between patients with and without infection in the amniotic cavity. This is in keeping with the observations of others [331, 332]. The most likely explanation for this is that IL-6 is a mediator of the acute phase response, and therefore, its concentration is elevated in the presence of an inflammatory insult unrelated to infection [87, 333, 334]. For example, the maternal plasma concentration of IL-6 is elevated in patients with spontaneous labor at term [335–344] without proven infection, which was assessed by amniotic fluid analysis.

In contrast to maternal plasma, amniotic fluid IL-6 concentrations are of value in the identification of the patient at risk for preterm delivery [112, 116–119, 122, 123, 169, 211, 212] or neonatal complications [112, 116, 119, 122, 123, 214, 345, 346], and are superior to amniotic fluid white blood cell counts, glucose concentrations, or Gram stains [331, 347–353]. Recent studies suggest that amniotic fluid IL-6 analysis is equivalent to other techniques, such as proteomic analysis of amniotic fluid [353], and rapid assays are now available which provide results within 20 min [120, 124, 125, 354–356]. These assays allow IL-6 to be used as a point-of-care test [120, 124, 125, 354–356].

Conclusion

We demonstrate, for the first time, that patients with clinical chorioamnionitis at term have higher maternal plasma concentrations of pyrogenic cytokines identified thus far (IL-1β, IL-2, IL-6, IFN-γ, and TNF-α) than patients in spontaneous labor at term without a fever. However, among patients with clinical chorioamnionitis, the absolute concentration of cytokines cannot be used to identify those who have bacteria in the amniotic fluid. This suggests that amniotic fluid assessment is required to identify intra-amniotic inflammation. Recently, a transcervical amniotic fluid collector was described that showed promising results in the analysis of the amniotic fluid of patients with preterm PROM [357]. This device may be of value to a subset of patients who will benefit from anti-inflammatory or antibiotic treatment.


Corresponding author: Roberto Romero, MD, D. Med. Sci, Perinatology Research Branch, NICHD/NIH/DHHS, Wayne State University/Hutzel Women’s Hospital, 3990 John R, Box 4, Detroit, MI 48201, USA, Tel.: +1 313 993 2700, Fax: +1 313 993 2694, E-mail: ; Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD and Detroit, MI, USA; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA; Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA; and Department of Molecular Obstetrics and Genetics, Wayne State University, Detroit, MI, USA

References

[1] Gibbs RS. Diagnosis of intra-amniotic infection. Semin Perinatol. 1977;1:71–7.Search in Google Scholar

[2] Gibbs RS, Castillo MS, Rodgers PJ. Management of acute chorioamnionitis. Am J Obstet Gynecol. 1980;136:709–13.10.1016/0002-9378(80)90445-7Search in Google Scholar

[3] Gibbs RS, Blanco JD, St Clair PJ, Castaneda YS. Quantitative bacteriology of amniotic fluid from women with clinical intraamniotic infection at term. J Infect Dis. 1982;145:1–8.10.1093/infdis/145.1.1Search in Google Scholar PubMed

[4] Romero R, Chaemsaithong P, Korzeniewski SJ, et al. Clinical chorioamnionitis at term III: how well do clinical criteria perform in the identification of proven intra-amniotic infection? J Perinat Med. 2016;44:23–32.Search in Google Scholar

[5] MacVicar J. Chorioamnionitis. Clin Obstet Gynecol. 1970;13:272–90.10.1097/00003081-197006000-00005Search in Google Scholar PubMed

[6] Hollander D. Diagnosis of chorioamnionitis. Clin Obstet Gynecol. 1986;29:816–25.10.1097/00003081-198612000-00008Search in Google Scholar PubMed

[7] Newton ER. Chorioamnionitis and intraamniotic infection. Clin Obstet Gynecol. 1993;36:795–808.10.1097/00003081-199312000-00004Search in Google Scholar PubMed

[8] Tita AT, Andrews WW. Diagnosis and management of clinical chorioamnionitis. Clin Perinatol. 2010;37:339–54.10.1016/j.clp.2010.02.003Search in Google Scholar PubMed PubMed Central

[9] Romero R, Miranda J, Kusanovic JP, Chaiworapongsa T, Chaemsaithong P, Martinez A, et al. Clinical chorioamnionitis at term I: microbiology of the amniotic cavity using cultivation and molecular techniques. J Perinat Med. 2015;43:19–36.10.1515/jpm-2014-0249Search in Google Scholar PubMed PubMed Central

[10] Romero R, Chaiworapongsa T, Savasan ZA, Hussein Y, Dong Z, Kusanovic JP, et al. Clinical chorioamnionitis is characterized by changes in the expression of the alarmin HMGB1 and one of its receptors, sRAGE. J Matern Fetal Neonatal Med. 2012;25:558–67.10.3109/14767058.2011.599083Search in Google Scholar PubMed PubMed Central

[11] Fishman SG, Gelber SE. Evidence for the clinical management of chorioamnionitis. Semin Fetal Neonatal Med. 2012;17:46–50.10.1016/j.siny.2011.09.002Search in Google Scholar PubMed

[12] Romero R, Chaemsaithong P, Korzeniewski SJ, et al. Clinical chorioamnionitis at term II: the intra-amniotic inflammatory response. J Perinat Med. 2016;44:5–22.Search in Google Scholar

[13] Romero R, Dey SK, Fisher SJ. Preterm labor: one syndrome, many causes. Science. 2014;345:760–5.10.1126/science.1251816Search in Google Scholar PubMed PubMed Central

[14] Duff P, Sanders R, Gibbs RS. The course of labor in term patients with chorioamnionitis. Am J Obstet Gynecol. 1983;147:391–5.10.1016/S0002-9378(16)32231-1Search in Google Scholar

[15] Hauth JC, Gilstrap LC 3rd, Hankins GD, Connor KD. Term maternal and neonatal complications of acute chorioamnionitis. Obstet Gynecol. 1985;66:59–62.Search in Google Scholar

[16] Satin AJ, Maberry MC, Leveno KJ, Sherman ML, Kline DM. Chorioamnionitis: a harbinger of dystocia. Obstet Gynecol. 1992;79:913–5.Search in Google Scholar

[17] Mark SP, Croughan-Minihane MS, Kilpatrick SJ. Chorioamnionitis and uterine function. Obstet Gynecol. 2000;95:909–12.Search in Google Scholar

[18] Rouse DJ, Landon M, Leveno KJ, Leindecker S, Varner MW, Caritis SN, et al. The maternal-fetal medicine units cesarean registry: chorioamnionitis at term and its duration-relationship to outcomes. Am J Obstet Gynecol. 2004;191:211–6.10.1016/j.ajog.2004.03.003Search in Google Scholar

[19] Edwards RK. Chorioamnionitis and labor. Obstet Gynecol Clin North Am. 2005;32:287–96.10.1016/j.ogc.2004.12.002Search in Google Scholar

[20] Yoder PR, Gibbs RS, Blanco JD, Castaneda YS, St Clair PJ. A prospective, controlled study of maternal and perinatal outcome after intra-amniotic infection at term. Am J Obstet Gynecol. 1983;145:695–701.10.1016/0002-9378(83)90575-6Search in Google Scholar

[21] Yancey MK, Duff P, Kubilis P, Clark P, Frentzen BH. Risk factors for neonatal sepsis. Obstet Gynecol. 1996;87:188–94.10.1016/0029-7844(95)00402-5Search in Google Scholar

[22] Grether JK, Nelson KB. Maternal infection and cerebral palsy in infants of normal birth weight. J Am Med Assoc. 1997;278:207–11.10.1001/jama.1997.03550030047032Search in Google Scholar

[23] Ladfors L, Tessin I, Mattsson LA, Eriksson M, Seeberg S, Fall O. Risk factors for neonatal sepsis in offspring of women with prelabor rupture of the membranes at 34–42 weeks. J Perinat Med. 1998;26:94–101.10.1515/jpme.1998.26.2.94Search in Google Scholar PubMed

[24] Alexander JM, McIntire DM, Leveno KJ. Chorioamnionitis and the prognosis for term infants. Obstet Gynecol. 1999;94:274–8.Search in Google Scholar

[25] Wu YW, Colford JM Jr. Chorioamnionitis as a risk factor for cerebral palsy: a meta-analysis. J Am Med Assoc. 2000;284:1417–24.10.1001/jama.284.11.1417Search in Google Scholar PubMed

[26] Nelson KB, Willoughby RE. Infection, inflammation and the risk of cerebral palsy. Curr Opin Neurol. 2000;13:133–9.10.1097/00019052-200004000-00004Search in Google Scholar PubMed

[27] Chaiworapongsa T, Romero R, Kim JC, Kim YM, Blackwell SC, Yoon BH, et al. Evidence for fetal involvement in the pathologic process of clinical chorioamnionitis. Am J Obstet Gynecol. 2002;186:1178–82.10.1067/mob.2002.124042Search in Google Scholar PubMed

[28] Hagberg H, Wennerholm UB, Savman K. Sequelae of chorioamnionitis. Curr Opin Infect Dis. 2002;15:301–6.10.1097/00001432-200206000-00014Search in Google Scholar

[29] Willoughby RE Jr, Nelson KB. Chorioamnionitis and brain injury. Clin Perinatol. 2002;29:603–21.10.1016/S0095-5108(02)00058-1Search in Google Scholar

[30] Wu YW, Escobar GJ, Grether JK, Croen LA, Greene JD, Newman TB. Chorioamnionitis and cerebral palsy in term and near-term infants. J Am Med Assoc. 2003;290:2677–84.10.1001/jama.290.20.2677Search in Google Scholar PubMed

[31] Cooke R. Chorioamnionitis, maternal fever, and neonatal encephalopathy. Dev Med Child Neurol. 2008;50:9.10.1111/j.1469-8749.2007.00009.xSearch in Google Scholar PubMed

[32] Blume HK, Li CI, Loch CM, Koepsell TD. Intrapartum fever and chorioamnionitis as risks for encephalopathy in term newborns: a case-control study. Dev Med Child Neurol. 2008;50:19–24.10.1111/j.1469-8749.2007.02007.xSearch in Google Scholar PubMed

[33] Soraisham AS, Singhal N, McMillan DD, Sauve RS, Lee SK, Canadian Neonatal Network. A multicenter study on the clinical outcome of chorioamnionitis in preterm infants. Am J Obstet Gynecol. 2009;200:372.e1–6.10.1016/j.ajog.2008.11.034Search in Google Scholar PubMed

[34] Shatrov JG, Birch SC, Lam LT, Quinlivan JA, McIntyre S, Mendz GL. Chorioamnionitis and cerebral palsy: a meta-analysis. Obstet Gynecol. 2010;116:387–92.10.1097/AOG.0b013e3181e90046Search in Google Scholar PubMed

[35] Martinelli P, Sarno L, Maruotti GM, Paludetto R. Chorioamnionitis and prematurity: a critical review. J Matern Fetal Neonatal Med. 2012;25(Suppl 4):29–31.10.3109/14767058.2012.714981Search in Google Scholar PubMed

[36] de Sam Lazaro S, Cheng Y, Snowden J, Pereira L, Aziz N, Caughey A. Does the neonatal impact of chorioamnionitis in the setting of PPROM vary depending on degree of prematurity? Am J Obstet Gynecol. 2013;208(Suppl):S314.10.1016/j.ajog.2012.10.084Search in Google Scholar

[37] Garcia-Munoz Rodrigo F, Galan Henriquez GM, Ospina CG. Morbidity and mortality among very-low-birth-weight infants born to mothers with clinical chorioamnionitis. Pediatr Neonatol. 2014;55:381–6.10.1016/j.pedneo.2013.12.007Search in Google Scholar PubMed

[38] Uyemura A, Ameel B, Caughey A. Outcomes of chorioamnionitis in term pregnancies. Am J Obstet Gynecol. 2014;210(Suppl):S215.10.1016/j.ajog.2013.10.458Search in Google Scholar

[39] Malloy MH. Chorioamnionitis: epidemiology of newborn management and outcome United States 2008. J Perinatol. 2014;34:611–5.10.1038/jp.2014.81Search in Google Scholar PubMed

[40] Mendez-Figueroa H, Abramovici A, O’Neil AE, Dahlke J, Pedroza C, Chauhan S. Chorioamnionitis without and with neonatal sepsis: newborn and infant outcomes. Am J Obstet Gynecol. 2015;212(Suppl):S318–9.10.1016/j.ajog.2014.10.853Search in Google Scholar

[41] Dinarello CA, Bunn PA Jr. Fever. Semin Oncol. 1997;24: 288–98.Search in Google Scholar

[42] Dinarello CA. Infection, fever, and exogenous and endogenous pyrogens: some concepts have changed. J Endotoxin Res. 2004;10:201–22.Search in Google Scholar

[43] Conti B, Tabarean I, Andrei C, Bartfai T. Cytokines and fever. Front Biosci. 2004;9:1433–49.10.2741/1341Search in Google Scholar PubMed

[44] Dinarello CA. Interleukin-1. Rev Infect Dis. 1984;6:51–95.10.1093/clinids/6.1.51Search in Google Scholar PubMed

[45] Sobrado J, Moldawer LL, Bistrian BR, Dinarello CA, Blackburn GL. Effect of ibuprofen on fever and metabolic changes induced by continuous infusion of leukocytic pyrogen (interleukin 1) or endotoxin. Infect Immun. 1983;42:997–1005.10.1128/iai.42.3.997-1005.1983Search in Google Scholar PubMed PubMed Central

[46] Opp MR, Krueger JM. Interleukin 1-receptor antagonist blocks interleukin 1-induced sleep and fever. Am J Physiol. 1991;260:R453–7.10.1152/ajpregu.1991.260.2.R453Search in Google Scholar PubMed

[47] Luheshi G, Miller AJ, Brouwer S, Dascombe MJ, Rothwell NJ, Hopkins SJ. Interleukin-1 receptor antagonist inhibits endotoxin fever and systemic interleukin-6 induction in the rat. Am J Physiol. 1996;270:E91–5.10.1152/ajpendo.1996.270.1.E91Search in Google Scholar PubMed

[48] Gourine AV, Rudolph K, Tesfaigzi J, Kluger MJ. Role of hypothalamic interleukin-1beta in fever induced by cecal ligation and puncture in rats. Am J Physiol. 1998;275:R754–61.10.1152/ajpregu.1998.275.3.R754Search in Google Scholar PubMed

[49] Lundkvist J, Sundgren-Andersson AK, Tingsborg S, Ostlund P, Engfors C, Alheim K, et al. Acute-phase responses in transgenic mice with CNS overexpression of IL-1 receptor antagonist. Am J Physiol. 1999;276:R644–51.10.1152/ajpregu.1999.276.3.R644Search in Google Scholar PubMed

[50] Boneberg EM, Hartung T. Febrile temperatures attenuate IL-1 beta release by inhibiting proteolytic processing of the proform and influence Th1/Th2 balance by favoring Th2 cytokines. J Immunol. 2003;171:664–8.10.4049/jimmunol.171.2.664Search in Google Scholar

[51] Garlanda C, Dinarello CA, Mantovani A. The interleukin-1 family: back to the future. Immunity. 2013;39:1003–18.10.1016/j.immuni.2013.11.010Search in Google Scholar

[52] Nedwin GE, Svedersky LP, Bringman TS, Palladino MA Jr, Goeddel DV. Effect of interleukin 2, interferon-gamma, and mitogens on the production of tumor necrosis factors alpha and beta. J Immunol. 1985;135:2492–7.10.4049/jimmunol.135.4.2492Search in Google Scholar

[53] Lotze MT, Matory YL, Rayner AA, Ettinghausen SE, Vetto JT, Seipp CA, et al. Clinical effects and toxicity of interleukin-2 in patients with cancer. Cancer. 1986;58:2764–72.10.1002/1097-0142(19861215)58:12<2764::AID-CNCR2820581235>3.0.CO;2-ZSearch in Google Scholar

[54] Mier JW, Vachino G, van der Meer JW, Numerof RP, Adams S, Cannon JG, et al. Induction of circulating tumor necrosis factor (TNF alpha) as the mechanism for the febrile response to interleukin-2 (IL-2) in cancer patients. J Clin Immunol. 1988;8:426–36.10.1007/BF00916947Search in Google Scholar

[55] Ueno Y, Takano N, Kanegane H, Yokoi T, Yachie A, Miyawaki T, et al. The acute phase nature of interleukin 6: studies in Kawasaki disease and other febrile illnesses. Clin Exp Immunol. 1989;76:337–42.Search in Google Scholar

[56] LeMay LG, Vander AJ, Kluger MJ. Role of interleukin 6 in fever in rats. Am J Physiol. 1990;258:R798–803.10.1152/ajpregu.1990.258.3.R798Search in Google Scholar

[57] Coceani F, Lees J, Mancilla J, Belizario J, Dinarello CA. Interleukin-6 and tumor necrosis factor in cerebrospinal fluid: changes during pyrogen fever. Brain Res. 1993;612:165–71.10.1016/0006-8993(93)91657-ESearch in Google Scholar

[58] Chai Z, Gatti S, Toniatti C, Poli V, Bartfai T. Interleukin (IL)-6 gene expression in the central nervous system is necessary for fever response to lipopolysaccharide or IL-1 beta: a study on IL-6-deficient mice. J Exp Med. 1996;183:311–6.10.1084/jem.183.1.311Search in Google Scholar

[59] Cartmell T, Poole S, Turnbull AV, Rothwell NJ, Luheshi GN. Circulating interleukin-6 mediates the febrile response to localised inflammation in rats. J Physiol. 2000;526:653–61.10.1111/j.1469-7793.2000.00653.xSearch in Google Scholar

[60] Dinarello CA, Bernheim HA, Duff GW, Le HV, Nagabhushan TL, Hamilton NC, et al. Mechanisms of fever induced by recombinant human interferon. J Clin Invest. 1984;74:906–13.10.1172/JCI111508Search in Google Scholar

[61] Luheshi GN. Cytokines and fever. Mechanisms and sites of action. Ann NY Acad Sci. 1998;856:83–9.10.1111/j.1749-6632.1998.tb08316.xSearch in Google Scholar

[62] Dinarello CA, Cannon JG, Wolff SM, Bernheim HA, Beutler B, Cerami A, et al. Tumor necrosis factor (cachectin) is an endogenous pyrogen and induces production of interleukin 1. J Exp Med. 1986;163:1433–50.10.1084/jem.163.6.1433Search in Google Scholar

[63] Nakamura H, Seto Y, Motoyoshi S, Kadokawa T, Sunahara N. Recombinant human tumor necrosis factor causes long-lasting and prostaglandin-mediated fever, with little tolerance, in rabbits. J Pharmacol Exp Ther. 1988;245:336–41.Search in Google Scholar

[64] Morimoto A, Sakata Y, Watanabe T, Murakami N. Characteristics of fever and acute-phase response induced in rabbits by IL-1 and TNF. Am J Physiol. 1989;256:R35–41.10.1152/ajpregu.1989.256.1.R35Search in Google Scholar

[65] Kawasaki H, Moriyama M, Ohtani Y, Naitoh M, Tanaka A, Nariuchi H. Analysis of endotoxin fever in rabbits by using a monoclonal antibody to tumor necrosis factor (cachectin). Infect Immun. 1989;57:3131–5.10.1128/iai.57.10.3131-3135.1989Search in Google Scholar

[66] Gay NJ, Keith FJ. Drosophila Toll and IL-1 receptor. Nature. 1991;351:355–6.10.1038/351355b0Search in Google Scholar

[67] Dalpke A, Heeg K. Signal integration following Toll-like receptor triggering. Crit Rev Immunol. 2002;22:217–50.10.1615/CritRevImmunol.v22.i3.40Search in Google Scholar

[68] Beutler B. Toll-like receptors: how they work and what they do. Curr Opin Hematol. 2002;9:2–10.10.1097/00062752-200201000-00002Search in Google Scholar

[69] Leon LR. Invited review: cytokine regulation of fever: studies using gene knockout mice. J Appl Physiol (1985). 2002;92:2648–55.10.1152/japplphysiol.01005.2001Search in Google Scholar

[70] Romanovsky AA, Almeida MC, Aronoff DM, Ivanov AI, Konsman JP, Steiner AA, et al. Fever and hypothermia in systemic inflammation: recent discoveries and revisions. Front Biosci. 2005;10:2193–216.10.2741/1690Search in Google Scholar

[71] Li S, Wang Y, Matsumura K, Ballou LR, Morham SG, Blatteis CM. The febrile response to lipopolysaccharide is blocked in cyclooxygenase-2(-/-), but not in cyclooxygenase-1(-/-) mice. Brain Res. 1999;825:86–94.10.1016/S0006-8993(99)01225-1Search in Google Scholar

[72] Li S, Ballou LR, Morham SG, Blatteis CM. Cyclooxygenase-2 mediates the febrile response of mice to interleukin-1beta. Brain Res. 2001;910:163–73.10.1016/S0006-8993(01)02707-XSearch in Google Scholar

[73] Blatteis CM, Li S, Li Z, Feleder C, Perlik V. Cytokines, PGE2 and endotoxic fever: a re-assessment. Prostaglandins Other Lipid Mediat. 2005;76:1–18.10.1016/j.prostaglandins.2005.01.001Search in Google Scholar

[74] Mier JW, Souza LM, Allegretta M, Boone T, Bernheim HA, Dinarello CA. Dissimilarities between purified human interleukin-1 and recombinant human interleukin-2 in the induction of fever, brain prostaglandin, and acute-phase protein synthesis. J Biol Response Mod. 1985;4:35–45.Search in Google Scholar

[75] Dinarello CA, Cannon JG, Mancilla J, Bishai I, Lees J, Coceani F. Interleukin-6 as an endogenous pyrogen: induction of prostaglandin E2 in brain but not in peripheral blood mononuclear cells. Brain Res. 1991;562:199–206.10.1016/0006-8993(91)90622-3Search in Google Scholar

[76] Ushikubi F, Segi E, Sugimoto Y, Murata T, Matsuoka T, Kobayashi T, et al. Impaired febrile response in mice lacking the prostaglandin E receptor subtype EP3. Nature. 1998;395:281–4.10.1038/26233Search in Google Scholar

[77] Dinarello CA, Gatti S, Bartfai T. Fever: links with an ancient receptor. Curr Biol. 1999;9:R147–50.10.1016/S0960-9822(99)80085-2Search in Google Scholar

[78] Li S, Goorha S, Ballou LR, Blatteis CM. Intracerebroventricular interleukin-6, macrophage inflammatory protein-1 beta and IL-18: pyrogenic and PGE(2)-mediated? Brain Res. 2003;992:76–84.10.1016/j.brainres.2003.08.033Search in Google Scholar PubMed

[79] Ivanov AI, Romanovsky AA. Prostaglandin E2 as a mediator of fever: synthesis and catabolism. Front Biosci. 2004;9:1977–93.10.2741/1383Search in Google Scholar PubMed

[80] Oka T. Prostaglandin E2 as a mediator of fever: the role of prostaglandin E (EP) receptors. Front Biosci. 2004;9:3046–57.10.2741/1458Search in Google Scholar PubMed

[81] Laburn HP, Rosendorff C, Willies G, Woolf C. Proceedings: a role for noradrenaline and cyclic AMP in prostaglandin E1 Fever. J Physiol. 1974;240:49P–50P.Search in Google Scholar

[82] Hou CC, Lin H, Chang CP, Huang WT, Lin MT. Oxidative stress and pyrogenic fever pathogenesis. Eur J Pharmacol. 2011;667:6–12.10.1016/j.ejphar.2011.05.075Search in Google Scholar PubMed

[83] Lieberman E, Lang JM, Frigoletto F Jr, Richardson DK, Ringer SA, Cohen A. Epidural analgesia, intrapartum fever, and neonatal sepsis evaluation. Pediatrics. 1997;99:415–9.10.1542/peds.99.3.415Search in Google Scholar PubMed

[84] Gonen R, Korobochka R, Degani S, Gaitini L. Association between epidural analgesia and intrapartum fever. Am J Perinatol. 2000;17:127–30.10.1055/s-2000-9283Search in Google Scholar PubMed

[85] Goetzl L, Cohen A, Frigoletto F Jr, Ringer SA, Lang JM, Lieberman E. Maternal epidural use and neonatal sepsis evaluation in afebrile mothers. Pediatrics. 2001;108:1099–102.10.1542/peds.108.5.1099Search in Google Scholar

[86] Yancey MK, Zhang J, Schwarz J, Dietrich CS 3rd, Klebanoff M. Labor epidural analgesia and intrapartum maternal hyperthermia. Obstet Gynecol. 2001;98:763–70.10.1097/00006250-200111000-00010Search in Google Scholar

[87] Smulian JC, Bhandari V, Vintzileos AM, Shen-Schwarz S, Quashie C, Lai-Lin YL, et al. Intrapartum fever at term: serum and histologic markers of inflammation. Am J Obstet Gynecol. 2003;188:269–74.10.1067/mob.2003.11Search in Google Scholar

[88] Evron S, Parameswaran R, Zipori D, Ezri T, Sadan O, Koren R. Activin betaA in term placenta and its correlation with placental inflammation in parturients having epidural or systemic meperidine analgesia: a randomized study. J Clin Anesth. 2007;19:168–74.10.1016/j.jclinane.2006.10.013Search in Google Scholar

[89] Riley LE, Celi AC, Onderdonk AB, Roberts DJ, Johnson LC, Tsen LC, et al. Association of epidural-related fever and noninfectious inflammation in term labor. Obstet Gynecol. 2011;117:588–95.10.1097/AOG.0b013e31820b0503Search in Google Scholar

[90] Kovo M, Schreiber L, Ben-Haroush A, Shor S, Golan A, Bar J. Intrapartum fever at term: clinical characteristics and placental pathology. J Matern Fetal Neonatal Med. 2012;25:1273–7.10.3109/14767058.2011.629248Search in Google Scholar

[91] Sharma SK, Rogers BB, Alexander JM, McIntire DD, Leveno KJ. A randomized trial of the effects of antibiotic prophylaxis on epidural-related fever in labor. Anesth Analg. 2014;118:604–10.10.1213/ANE.0b013e3182a5d539Search in Google Scholar

[92] Gleeson NC, Nolan KM, Ford MR. Temperature, labour, and epidural analgesia. Lancet. 1989;2:861–2.10.1016/S0140-6736(89)93020-1Search in Google Scholar

[93] Fusi L, Steer PJ, Maresh MJ, Beard RW. Maternal pyrexia associated with the use of epidural analgesia in labour. Lancet. 1989;1:1250–2.10.1016/S0140-6736(89)92341-6Search in Google Scholar

[94] Vinson DC, Thomas R, Kiser T. Association between epidural analgesia during labor and fever. J Fam Pract. 1993;36:617–22.Search in Google Scholar

[95] Ramin SM, Gambling DR, Lucas MJ, Sharma SK, Sidawi JE, Leveno KJ. Randomized trial of epidural versus intravenous analgesia during labor. Obstet Gynecol. 1995;86:783–9.10.1016/0029-7844(95)00269-WSearch in Google Scholar

[96] Mayer DC, Chescheir NC, Spielman FJ. Increased intrapartum antibiotic administration associated with epidural analgesia in labor. Am J Perinatol. 1997;14:83–6.10.1055/s-2007-994103Search in Google Scholar PubMed

[97] Sharma SK, Sidawi JE, Ramin SM, Lucas MJ, Leveno KJ, Cunningham FG. Cesarean delivery: a randomized trial of epidural versus patient-controlled meperidine analgesia during labor. Anesthesiology. 1997;87:487–94.10.1097/00000542-199709000-00006Search in Google Scholar PubMed

[98] Dashe JS, Rogers BB, McIntire DD, Leveno KJ. Epidural analgesia and intrapartum fever: placental findings. Obstet Gynecol. 1999;93:341–4.10.1097/00006250-199903000-00005Search in Google Scholar

[99] Philip J, Alexander JM, Sharma SK, Leveno KJ, McIntire DD, Wiley J. Epidural analgesia during labor and maternal fever. Anesthesiology. 1999;90:1271–5.10.1097/00000542-199905000-00008Search in Google Scholar PubMed

[100] Lucas MJ, Sharma SK, McIntire DD, Wiley J, Sidawi JE, Ramin SM, et al. A randomized trial of labor analgesia in women with pregnancy-induced hypertension. Am J Obstet Gynecol. 2001;185:970–5.10.1067/mob.2001.117970Search in Google Scholar PubMed

[101] Sharma SK, Alexander JM, Messick G, Bloom SL, McIntire DD, Wiley J, et al. Cesarean delivery: a randomized trial of epidural analgesia versus intravenous meperidine analgesia during labor in nulliparous women. Anesthesiology. 2002;96:546–51.10.1097/00000542-200203000-00007Search in Google Scholar PubMed

[102] Goetzl L, Rivers J, Zighelboim I, Wali A, Badell M, Suresh MS. Intrapartum epidural analgesia and maternal temperature regulation. Obstet Gynecol. 2007;109:687–90.10.1097/01.AOG.0000255976.14297.f6Search in Google Scholar PubMed

[103] Segal S. Labor epidural analgesia and maternal fever. Anesth Analg. 2010;111:1467–75.10.1213/ANE.0b013e3181f713d4Search in Google Scholar PubMed

[104] Goetzl L. Epidural analgesia and maternal fever: a clinical and research update. Curr Opin Anaesthesiol. 2012;25:292–9.10.1097/ACO.0b013e3283530d7cSearch in Google Scholar PubMed

[105] Greenwell EA, Wyshak G, Ringer SA, Johnson LC, Rivkin MJ, Lieberman E. Intrapartum temperature elevation, epidural use, and adverse outcome in term infants. Pediatrics. 2012;129:e447–54.10.1542/peds.2010-2301Search in Google Scholar PubMed PubMed Central

[106] Arendt KW, Segal BS. The association between epidural labor analgesia and maternal fever. Clin Perinatol. 2013;40:385–98.10.1016/j.clp.2013.06.002Search in Google Scholar PubMed

[107] Goetzl L. Epidural fever in obstetric patients: it’s a hot topic. Anesth Analg. 2014;118:494–5.10.1213/ANE.0000000000000112Search in Google Scholar PubMed

[108] DiGiulio DB, Romero R, Amogan HP, Kusanovic JP, Bik EM, Gotsch F, et al. Microbial prevalence, diversity and abundance in amniotic fluid during preterm labor: a molecular and culture-based investigation. PLoS One. 2008;3:e3056.10.1371/journal.pone.0003056Search in Google Scholar PubMed PubMed Central

[109] DiGiulio DB, Romero R, Kusanovic JP, Gómez R, Kim CJ, Seok KS, et al. Prevalence and diversity of microbes in the amniotic fluid, the fetal inflammatory response, and pregnancy outcome in women with preterm pre-labor rupture of membranes. Am J Reprod Immunol. 2010;64: 38–57.10.1111/j.1600-0897.2010.00830.xSearch in Google Scholar PubMed PubMed Central

[110] DiGiulio DB, Gervasi M, Romero R, Mazaki-Tovi S, Vaisbuch E, Kusanovic JP, et al. Microbial invasion of the amniotic cavity in pre-eclampsia as assessed by cultivation and sequence-based methods. J Perinat Med. 2010;38:503–13.10.1515/jpm.2010.078Search in Google Scholar PubMed PubMed Central

[111] DiGiulio DB, Gervasi MT, Romero R, Vaisbuch E, Mazaki-Tovi S, Kusanovic JP, et al. Microbial invasion of the amniotic cavity in pregnancies with small-for-gestational-age fetuses. J Perinat Med. 2010;38:495–502.10.1515/jpm.2010.076Search in Google Scholar PubMed PubMed Central

[112] Yoon BH, Romero R, Moon JB, Shim SS, Kim M, Kim G, et al. Clinical significance of intra-amniotic inflammation in patients with preterm labor and intact membranes. Am J Obstet Gynecol. 2001;185:1130–6.10.1067/mob.2001.117680Search in Google Scholar PubMed

[113] Romero R, Chaiworapongsa T, Alpay Savasan Z, Xu Y, Hussein Y, Dong Z, et al. Damage-associated molecular patterns (DAMPs) in preterm labor with intact membranes and preterm PROM: a study of the alarmin HMGB1. J Matern Fetal Neonatal Med. 2011;24:1444–55.10.3109/14767058.2011.591460Search in Google Scholar PubMed PubMed Central

[114] Gervasi MT, Romero R, Bracalente G, Erez O, Dong Z, Hassan SS, et al. Midtrimester amniotic fluid concentrations of interleukin-6 and interferon-gamma-inducible protein-10: evidence for heterogeneity of intra-amniotic inflammation and associations with spontaneous early (<32 weeks) and late (>32 weeks) preterm delivery. J Perinat Med. 2012;40:329–43.10.1515/jpm-2012-0034Search in Google Scholar PubMed PubMed Central

[115] Kim SK, Romero R, Savasan ZA, Xu Y, Dong Z, Lee DC, et al. Endoglin in amniotic fluid as a risk factor for the subsequent development of bronchopulmonary dysplasia. Am J Reprod Immunol. 2013;69:105–23.10.1111/aji.12046Search in Google Scholar PubMed PubMed Central

[116] Romero R, Miranda J, Chaiworapongsa T, Chaemsaithong P, Gotsch F, Dong Z, et al. A novel molecular microbiologic technique for the rapid diagnosis of microbial invasion of the amniotic cavity and intra-amniotic infection in preterm labor with intact membranes. Am J Reprod Immunol. 2014;71:330–58.10.1111/aji.12189Search in Google Scholar PubMed PubMed Central

[117] Romero R, Miranda J, Chaemsaithong P, Chaiworapongsa T, Kusanovic JP, Dong Z, et al. Sterile and microbial-associated intra-amniotic inflammation in preterm prelabor rupture of membranes. J Matern Fetal Neonatal Med. 2014;1–16. [Epub ahead of print].10.3109/14767058.2014.958463Search in Google Scholar PubMed PubMed Central

[118] Romero R, Miranda J, Chaiworapongsa T, Chaemsaithong P, Gotsch F, Dong Z, et al. Sterile intra-amniotic inflammation in asymptomatic patients with a sonographic short cervix: prevalence and clinical significance. J Matern Fetal Neonatal Med. 2014;1–17. [Epub ahead of print].10.3109/14767058.2014.954243Search in Google Scholar PubMed PubMed Central

[119] Romero R, Miranda J, Chaiworapongsa T, Korzeniewski SJ, Chaemsaithong P, Gotsch F, et al. Prevalence and clinical significance of sterile intra-amniotic inflammation in patients with preterm labor and intact membranes. Am J Reprod Immunol. 2014;72:458–74.10.1111/aji.12296Search in Google Scholar PubMed PubMed Central

[120] Chaemsaithong P, Romero R, Korzeniewski SJ, Dong Z, Yeo L, Hassan SS, et al. A point of care test for the determination of amniotic fluid interleukin-6 and the chemokine CXCL-10/IP-10. J Matern Fetal Neonatal Med. 2014;1–10. [Epub ahead of print].10.3109/14767058.2014.961417Search in Google Scholar PubMed PubMed Central

[121] Kacerovsky M, Musilova I, Andrys C, Hornychova H, Pliskova L, Kostal M, et al. Prelabor rupture of membranes between 34 and 37 weeks: the intraamniotic inflammatory response and neonatal outcomes. Am J Obstet Gynecol. 2014;210:325.e1–10.10.1016/j.ajog.2013.10.882Search in Google Scholar PubMed

[122] Combs CA, Gravett M, Garite TJ, Hickok DE, Lapidus J, Porreco R, et al. Amniotic fluid infection, inflammation, and colonization in preterm labor with intact membranes. Am J Obstet Gynecol. 2014;210:125.e1–15.10.1016/j.ajog.2013.11.032Search in Google Scholar PubMed

[123] Cobo T, Kacerovsky M, Jacobsson B. Amniotic fluid infection, inflammation, and colonization in preterm labor with intact membranes. Am J Obstet Gynecol. 2014;211:708.10.1016/j.ajog.2014.06.060Search in Google Scholar PubMed

[124] Chaemsaithong P, Romero R, Korzeniewski SJ, Martinez-Varea A, Dong Z, Yoon BH, et al. A rapid interleukin-6 bedside test for the identification of intra-amniotic inflammation in preterm labor with intact membranes. J Matern Fetal Neonatal Med. 2015;1–11. [Epub ahead of print].10.3109/14767058.2015.1006620Search in Google Scholar PubMed PubMed Central

[125] Chaemsaithong P, Romero R, Korzeniewski SJ, Martinez-Varea A, Dong Z, Yoon BH, et al. A point of care test for interleukin-6 in amniotic fluid in preterm prelabor rupture of membrances: a step toward the early treatment of acute intra-amniotic inflammation/infection. J Matern Fetal Neonatal Med. 2015;1–8. [Epub ahead of print].10.3109/14767058.2015.1006621Search in Google Scholar PubMed PubMed Central

[126] Gibbs RS, Dinsmoor MJ, Newton ER, Ramamurthy RS. A randomized trial of intrapartum versus immediate postpartum treatment of women with intra-amniotic infection. Obstet Gynecol. 1988;72:823–8.10.1097/00006250-198812000-00001Search in Google Scholar

[127] Gilstrap LC 3rd, Cox SM. Acute chorioamnionitis. Obstet Gynecol Clin North Am. 1989;16:373–9.10.1016/S0889-8545(21)00165-0Search in Google Scholar

[128] Seeds JW. Impaired fetal growth: definition and clinical diagnosis. Obstet Gynecol. 1984;64:303–10.Search in Google Scholar

[129] Alexander GR, Himes JH, Kaufman RB, Mor J, Kogan M. A United States national reference for fetal growth. Obstet Gynecol. 1996;87:163–8.10.1016/0029-7844(95)00386-XSearch in Google Scholar

[130] Redline RW, Heller D, Keating S, Kingdom J. Placental diagnostic criteria and clinical correlation – a workshop report. Placenta. 2005;26(Suppl A):S114–7.10.1016/j.placenta.2005.02.009Search in Google Scholar PubMed

[131] Redline RW. Inflammatory responses in the placenta and umbilical cord. Semin Fetal Neonatal Med. 2006;11:296–301.10.1016/j.siny.2006.02.011Search in Google Scholar PubMed

[132] Chaiworapongsa T, Erez O, Kusanovic JP, Vaisbuch E, Mazaki-Tovi S, Gotsch F, et al. Amniotic fluid heat shock protein 70 concentration in histologic chorioamnionitis, term and preterm parturition. J Matern Fetal Neonatal Med. 2008;21:449–61.10.1080/14767050802054550Search in Google Scholar PubMed PubMed Central

[133] Seong HS, Lee SE, Kang JH, Romero R, Yoon BH. The frequency of microbial invasion of the amniotic cavity and histologic chorioamnionitis in women at term with intact membranes in the presence or absence of labor. Am J Obstet Gynecol. 2008;199:375.e1–5.10.1016/j.ajog.2008.06.040Search in Google Scholar PubMed PubMed Central

[134] Park CW, Moon KC, Park JS, Jun JK, Romero R, Yoon BH. The involvement of human amnion in histologic chorioamnionitis is an indicator that a fetal and an intra-amniotic inflammatory response is more likely and severe: clinical implications. Placenta. 2009;30:56–61.10.1016/j.placenta.2008.09.017Search in Google Scholar PubMed PubMed Central

[135] Kacerovsky M, Pliskova L, Bolehovska R, Musilova I, Hornychova H, Tambor V, et al. The microbial load with genital mycoplasmas correlates with the degree of histologic chorioamnionitis in preterm PROM. Am J Obstet Gynecol. 2011;205:213.e1–7.10.1016/j.ajog.2011.04.028Search in Google Scholar PubMed

[136] Mi Lee S, Romero R, Lee KA, Jin Yang H, Joon Oh K, Park CW, et al. The frequency and risk factors of funisitis and histologic chorioamnionitis in pregnant women at term who delivered after the spontaneous onset of labor. J Matern Fetal Neonatal Med. 2011;24:37–42.10.3109/14767058.2010.482622Search in Google Scholar PubMed PubMed Central

[137] Tsiartas P, Kacerovsky M, Musilova I, Hornychova H, Cobo T, Sävman K, et al. The association between histological chorioamnionitis, funisitis and neonatal outcome in women with preterm prelabor rupture of membranes. J Matern Fetal Neonatal Med. 2013;26:1332–6.10.3109/14767058.2013.784741Search in Google Scholar PubMed

[138] Korzeniewski SJ, Romero R, Cortez J, Pappas A, Schwartz AG, Kim CJ, et al. A "multi-hit" model of neonatal white matter injury: cumulative contributions of chronic placental inflammation, acute fetal inflammation and postnatal inflammatory events. J Perinat Med. 2014;42:731–43.10.1515/jpm-2014-0250Search in Google Scholar PubMed PubMed Central

[139] Kim SM, Romero R, Park JW, Oh KJ, Jun JK, Yoon BH. The relationship between the intensity of intra-amniotic inflammation and the presence and severity of acute histologic chorioamnionitis in preterm gestation. J Matern Fetal Neonatal Med. 2014;1–10. doi:10.3109/14767058.2014.961009.10.3109/14767058.2014.961009Search in Google Scholar PubMed PubMed Central

[140] Pacora P, Chaiworapongsa T, Maymon E, Kim YM, Gomez R, Yoon BH, et al. Funisitis and chorionic vasculitis: the histological counterpart of the fetal inflammatory response syndrome. J Matern Fetal Neonatal Med. 2002;11:18–25.10.1080/jmf.11.1.18.25Search in Google Scholar PubMed

[141] Yoon BH, Romero R, Park JS, Kim M, Oh SY, Kim CJ, et al. The relationship among inflammatory lesions of the umbilical cord (funisitis), umbilical cord plasma interleukin 6 concentration, amniotic fluid infection, and neonatal sepsis. Am J Obstet Gynecol. 2000;183:1124–9.10.1067/mob.2000.109035Search in Google Scholar PubMed

[142] Park JS, Romero R, Yoon BH, Moon JB, Oh SY, Han SY, et al. The relationship between amniotic fluid matrix metalloproteinase-8 and funisitis. Am J Obstet Gynecol. 2001;185:1156–61.10.1067/mob.2001.117679Search in Google Scholar PubMed

[143] Yoon BH, Romero R, Shim JY, Shim SS, Kim CJ, Jun JK. C-reactive protein in umbilical cord blood: a simple and widely available clinical method to assess the risk of amniotic fluid infection and funisitis. J Matern Fetal Neonatal Med. 2003;14:85–90.10.1080/jmf.14.2.85.90Search in Google Scholar PubMed

[144] Lee SE, Romero R, Kim CJ, Shim SS, Yoon BH. Funisitis in term pregnancy is associated with microbial invasion of the amniotic cavity and intra-amniotic inflammation. J Matern Fetal Neonatal Med. 2006;19:693–7.10.1080/14767050600927353Search in Google Scholar PubMed

[145] Park CW, Lee SM, Park JS, Jun JK, Romero R, Yoon BH. The antenatal identification of funisitis with a rapid MMP-8 bedside test. J Perinat Med. 2008;36:497–502.10.1515/JPM.2008.079Search in Google Scholar PubMed PubMed Central

[146] R Core Team. A language and environment for statistical computing. 2013. R Foundation for Statistical Computing, Vienna, Austria. http://www.R-project.org/. Accessed in January 2015.Search in Google Scholar

[147] Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Series B Stat Methodol. 1995;57:289–300.10.1111/j.2517-6161.1995.tb02031.xSearch in Google Scholar

[148] Romero R, Mazor M, Wu YK, Sirtori M, Oyarzun E, Mitchell MD, et al. Infection in the pathogenesis of preterm labor. Semin Perinatol. 1988;12:262–79.Search in Google Scholar

[149] Romero R, Mazor M. Infection and preterm labor. Clin Obstet Gynecol. 1988;31:553–84.10.1097/00003081-198809000-00006Search in Google Scholar

[150] Romero R, Sirtori M, Oyarzun E, Avila C, Mazor M, Callahan R, et al. Infection and labor. V. Prevalence, microbiology, and clinical significance of intraamniotic infection in women with preterm labor and intact membranes. Am J Obstet Gynecol. 1989;161:817–24.10.1016/0002-9378(89)90409-2Search in Google Scholar

[151] Romero R, Avila C, Santhanam U, Sehgal PB. Amniotic fluid interleukin 6 in preterm labor. Association with infection. J Clin Invest. 1990;85:1392–400.10.1172/JCI114583Search in Google Scholar

[152] Romero R, Avila C, Brekus CA, Morotti R. The role of systemic and intrauterine infection in preterm parturition. Ann NY Acad Sci. 1991;622:355–75.10.1111/j.1749-6632.1991.tb37880.xSearch in Google Scholar

[153] Gibbs RS, Romero R, Hillier SL, Eschenbach DA, Sweet RL. A review of premature birth and subclinical infection. Am J Obstet Gynecol. 1992;166:1515–28.10.1016/0002-9378(92)91628-NSearch in Google Scholar

[154] Gomez R, Romero R, Edwin SS, David C. Pathogenesis of preterm labor and preterm premature rupture of membranes associated with intraamniotic infection. Infect Dis Clin North Am. 1997;11:135–76.10.1016/S0891-5520(05)70347-0Search in Google Scholar

[155] Romero R, Gomez R, Chaiworapongsa T, Conoscenti G, Kim JC, Kim YM. The role of infection in preterm labour and delivery. Paediatr Perinat Epidemiol. 2001;15(Suppl 2):41–56.10.1046/j.1365-3016.2001.00007.xSearch in Google Scholar PubMed

[156] Goncalves LF, Chaiworapongsa T, Romero R. Intrauterine infection and prematurity. Ment Retard Dev Disabil Res Rev. 2002;8:3–13.10.1002/mrdd.10008Search in Google Scholar PubMed

[157] Romero R, Espinoza J, Chaiworapongsa T, Kalache K. Infection and prematurity and the role of preventive strategies. Semin Neonatol. 2002;7:259–74.10.1053/siny.2002.0121Search in Google Scholar

[158] Romero R, Espinoza J, Goncalves LF, Kusanovic JP, Friel LA, Nien JK. Inflammation in preterm and term labour and delivery. Semin Fetal Neonatal Med. 2006;11:317–26.10.1016/j.siny.2006.05.001Search in Google Scholar PubMed PubMed Central

[159] Romero R, Espinoza J, Kusanovic JP, Gotsch F, Hassan S, Erez O, et al. The preterm parturition syndrome. BJOG. 2006;113(Suppl 3):17–42.10.1111/j.1471-0528.2006.01120.xSearch in Google Scholar

[160] 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:S194–202.10.1111/j.1753-4887.2007.tb00362.xSearch in Google Scholar

[161] Romero R, Lockwood CJ. Pathogenesis of spontaneous preterm labor. In: Creasy RK, Resnik R, Iams JD, Lockwood CJ, Moore TR, editors. Creasy and Resnik’s maternal-fetal medicine: principles and practice. 6th ed. Philadelphia: Saunders Elsevier; 2009. pp. 545–82.10.1016/B978-1-4160-4224-2.50032-6Search in Google Scholar

[162] Bastek JA, Gomez LM, Elovitz MA. The role of inflammation and infection in preterm birth. Clin Perinatol. 2011;38: 385–406.10.1016/j.clp.2011.06.003Search in Google Scholar

[163] Agrawal V, Hirsch E. Intrauterine infection and preterm labor. Semin Fetal Neonatal Med. 2012;17:12–9.10.1016/j.siny.2011.09.001Search in Google Scholar

[164] Allen-Daniels MJ, Serrano MG, Pflugner LP, Fettweis JM, Prestosa MA, Koparde VN, et al. Identification of a gene in Mycoplasma hominis associated with preterm birth and microbial burden in intra-amniotic infection. Am J Obstet Gynecol. 2015, [Epub ahead of print].Search in Google Scholar

[165] Zlatnik FJ, Cruikshank DP, Petzold CR, Galask RP. Amniocentesis in the identification of inapparent infection in preterm patients with premature rupture of the membranes. J Reprod Med. 1984;29:656–60.Search in Google Scholar

[166] Feinstein SJ, Vintzileos AM, Lodeiro JG, Campbell WA, Weinbaum PJ, Nochimson DJ. Amniocentesis with premature rupture of membranes. Obstet Gynecol. 1986;68:147–52.Search in Google Scholar

[167] Romero R, Quintero R, Oyarzun E, Wu YK, Sabo V, Mazor M, et al. Intraamniotic infection and the onset of labor in preterm premature rupture of the membranes. Am J Obstet Gynecol. 1988;159:661–6.10.1016/S0002-9378(88)80030-9Search in Google Scholar

[168] Mercer BM. Preterm premature rupture of the membranes. Obstet Gynecol. 2003;101:178–93.Search in Google Scholar

[169] Shim SS, Romero R, Hong JS, Park CW, Jun JK, Kim BI, et al. Clinical significance of intra-amniotic inflammation in patients with preterm premature rupture of membranes. Am J Obstet Gynecol. 2004;191:1339–45.10.1016/j.ajog.2004.06.085Search in Google Scholar PubMed

[170] Santolaya-Forgas J, Romero R, Espinoza J, Erez O, Friel LA, Kusanovic JP, et al. Prelabour rupture of the membranes. In: Reece EA, Hobbins JC, editors. Clinical obstetrics the fetus and mothers. 3rd ed. Malden: Blackwell; 2008. pp. 1130–88.10.1002/9780470753293.ch63Search in Google Scholar

[171] Romero R, Yeo L, Gotsch F, Soto E, Hassan S, Kusanovic JP, et al. Prelabor rupture of the membranes. In: Winn HN, Chervanak FA, Romero R, editors. Clinical maternal-fetal medicine online. 2nd ed. London: Informa Healthcare; 2011. pp. 1–24.10.1201/9781003222590-2Search in Google Scholar

[172] Kacerovsky M, Andrys C, Drahosova M, Musilova I, Hornychova H, Lesko D, et al. Soluble Toll-like receptor 1 family members in the amniotic fluid of women with preterm prelabor rupture of the membranes. J Matern Fetal Neonatal Med. 2012;25:1699–704.10.3109/14767058.2012.658463Search in Google Scholar PubMed

[173] Kacerovsky M, Musilova I, Khatibi A, Skogstrand K, Hougaard DM, Tambor V, et al. Intraamniotic inflammatory response to bacteria: analysis of multiple amniotic fluid proteins in women with preterm prelabor rupture of membranes. J Matern Fetal Neonatal Med. 2012;25:2014–9.10.3109/14767058.2012.671873Search in Google Scholar

[174] Ismail AQ, Lahiri S. Management of prelabour rupture of membranes (PROM) at term. J Perinat Med. 2013;41:647–9.Search in Google Scholar

[175] Eschenbach D. Reply to: Ismail AQT, Lahiri S. Management of prelabor rupture of membranes (PROM) at term J Perinat Med. 2013;41:653–5.Search in Google Scholar

[176] Grunebaum A. Reply to “Management of prelabour rupture of membranes (PROM) at term”. J Perinat Med. 2013;41:651–2.10.1515/jpm-2013-0127Search in Google Scholar

[177] Romero R, Gonzalez R, Sepulveda W, Brandt F, Ramirez M, Sorokin Y, et al. Infection and labor. VIII. Microbial invasion of the amniotic cavity in patients with suspected cervical incompetence: prevalence and clinical significance. Am J Obstet Gynecol. 1992;167:1086–91.10.1016/S0002-9378(12)80043-3Search in Google Scholar

[178] Lee SE, Romero R, Park CW, Jun JK, Yoon BH. The frequency and significance of intraamniotic inflammation in patients with cervical insufficiency. Am J Obstet Gynecol. 2008;198:633.e1–8.10.1016/j.ajog.2007.11.047Search in Google Scholar PubMed

[179] Oh KJ, Lee SE, Jung H, Kim G, Romero R, Yoon BH. Detection of ureaplasmas by the polymerase chain reaction in the amniotic fluid of patients with cervical insufficiency. J Perinat Med. 2010;38:261–8.Search in Google Scholar

[180] Hassan SS, Romero R, Berry SM, Dang K, Blackwell SC, Treadwell MC, et al. Patients with an ultrasonographic cervical length <or=15 mm have nearly a 50% risk of early spontaneous preterm delivery. Am J Obstet Gynecol. 2000;182:1458–67.10.1067/mob.2000.106851Search in Google Scholar PubMed

[181] Gomez R, Romero R, Nien JK, Chaiworapongsa T, Medina L, Kim YM, et al. A short cervix in women with preterm labor and intact membranes: a risk factor for microbial invasion of the amniotic cavity. Am J Obstet Gynecol. 2005;192:678–89.10.1016/j.ajog.2004.10.624Search in Google Scholar PubMed

[182] Hassan S, Romero R, Hendler I, Gomez R, Khalek N, Espinoza J, et al. A sonographic short cervix as the only clinical manifestation of intra-amniotic infection. J Perinat Med. 2006;34:13–9.10.1515/JPM.2006.002Search in Google Scholar PubMed PubMed Central

[183] Kiefer DG, Keeler SM, Rust OA, Wayock CP, Vintzileos AM, Hanna N. Is midtrimester short cervix a sign of intraamniotic inflammation? Am J Obstet Gynecol. 2009;200:374.e1–5.10.1016/j.ajog.2009.01.047Search in Google Scholar PubMed

[184] Vaisbuch E, Hassan SS, Mazaki-Tovi S, Nhan-Chang CL, Kusanovic JP, Chaiworapongsa T, et al. Patients with an asymptomatic short cervix (<or=15 mm) have a high rate of subclinical intraamniotic inflammation: implications for patient counseling. Am J Obstet Gynecol. 2010;202: 433.e1–8.10.1016/j.ajog.2010.02.007Search in Google Scholar PubMed PubMed Central

[185] Vaisbuch E, Romero R, Erez O, Kusanovic JP, Mazaki-Tovi S, Gotsch F, et al. Clinical significance of early (<20 weeks) vs. late (20–24 weeks) detection of sonographic short cervix in asymptomatic women in the mid-trimester. Ultrasound Obstet Gynecol. 2010;36:471–81.10.1002/uog.7673Search in Google Scholar PubMed PubMed Central

[186] Elovitz MA, Wang Z, Chien EK, Rychlik DF, Phillippe M. A new model for inflammation-induced preterm birth: the role of platelet-activating factor and Toll-like receptor-4. Am J Pathol. 2003;163:2103–11.10.1016/S0002-9440(10)63567-5Search in Google Scholar

[187] Wang H, Hirsch E. Bacterially-induced preterm labor and regulation of prostaglandin-metabolizing enzyme expression in mice: the role of toll-like receptor 4. Biol Reprod. 2003;69:1957–63.10.1095/biolreprod.103.019620Search in Google Scholar PubMed

[188] Kim YM, Romero R, Chaiworapongsa T, Kim GJ, Kim MR, Kuivaniemi H, et al. Toll-like receptor-2 and -4 in the chorioamniotic membranes in spontaneous labor at term and in preterm parturition that are associated with chorioamnionitis. Am J Obstet Gynecol. 2004;191:1346–55.10.1016/j.ajog.2004.07.009Search in Google Scholar PubMed

[189] Mor G, Romero R, Aldo PB, Abrahams VM. Is the trophoblast an immune regulator? The role of Toll-like receptors during pregnancy. Crit Rev Immunol. 2005;25:375–88.10.1615/CritRevImmunol.v25.i5.30Search in Google Scholar PubMed

[190] Koga K, Cardenas I, Aldo P, Abrahams VM, Peng B, Fill S, et al. Activation of TLR3 in the trophoblast is associated with preterm delivery. Am J Reprod Immunol. 2009;61:196–212.10.1111/j.1600-0897.2008.00682.xSearch in Google Scholar PubMed PubMed Central

[191] Kawai T, Akira S. The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors. Nat Immunol. 2010;11:373–84.10.1038/ni.1863Search in Google Scholar PubMed

[192] Ilievski V, Hirsch E. Synergy between viral and bacterial toll-like receptors leads to amplification of inflammatory responses and preterm labor in the mouse. Biol Reprod. 2010;83:767–73.10.1095/biolreprod.110.085464Search in Google Scholar PubMed PubMed Central

[193] Pawelczyk E, Nowicki BJ, Izban MG, Pratap S, Sashti NA, Sanderson M, et al. Spontaneous preterm labor is associated with an increase in the proinflammatory signal transducer TLR4 receptor on maternal blood monocytes. BMC Pregnancy Childbirth. 2010;10:66.10.1186/1471-2393-10-66Search in Google Scholar PubMed PubMed Central

[194] Li L, Kang J, Lei W. Role of Toll-like receptor 4 in inflammation-induced preterm delivery. Mol Hum Reprod. 2010;16:267–72.10.1093/molehr/gap106Search in Google Scholar PubMed

[195] Thaxton JE, Nevers TA, Sharma S. TLR-mediated preterm birth in response to pathogenic agents. Infect Dis Obstet Gynecol. 2010;2010:378472.10.1155/2010/378472Search in Google Scholar PubMed PubMed Central

[196] Breen K, Brown A, Burd I, Chai J, Friedman A, Elovitz MA. TLR-4-dependent and -independent mechanisms of fetal brain injury in the setting of preterm birth. Reprod Sci. 2012;19:839–50.10.1177/1933719112438439Search in Google Scholar

[197] Kacerovsky M, Andrys C, Hornychova H, Pliskova L, Lancz K, Musilova I, et al. Amniotic fluid soluble Toll-like receptor 4 in pregnancies complicated by preterm prelabor rupture of the membranes. J Matern Fetal Neonatal Med. 2012;25:1148–55.10.3109/14767058.2011.626821Search in Google Scholar

[198] Andrys C, Kacerovsky M, Drahosova M, Musilova I, Pliskova L, Hornychova H, et al. Amniotic fluid soluble Toll-like receptor 2 in pregnancies complicated by preterm prelabor rupture of membranes. J Matern Fetal Neonatal Med. 2013;26:520–7.10.3109/14767058.2012.741634Search in Google Scholar

[199] Abrahams VM, Potter JA, Bhat G, Peltier MR, Saade G, Menon R. Bacterial modulation of human fetal membrane Toll-like receptor expression. Am J Reprod Immunol. 2013;69:33–40.10.1111/aji.12016Search in Google Scholar

[200] 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;72:192–205.10.1111/aji.12258Search in Google Scholar

[201] Romero R, Durum SK, Dinarello CA. Interleukin-1: a signal for the initiation of labor in chorioamnionitis. 33rd Annual Meeting for the Society for Gynecologic Investigation. Toronto, Ontario; 1986.Search in Google Scholar

[202] Romero R, Brody DT, Oyarzun E, Mazor M, Wu YK, Hobbins JC, et al. Infection and labor. III. Interleukin-1: a signal for the onset of parturition. Am J Obstet Gynecol. 1989;160:1117–23.10.1016/0002-9378(89)90172-5Search in Google Scholar

[203] Romero R, Manogue KR, Mitchell MD, Wu YK, Oyarzun E, Hobbins JC, et al. Infection and labor. IV. Cachectin-tumor necrosis factor in the amniotic fluid of women with intraamniotic infection and preterm labor. Am J Obstet Gynecol. 1989;161:336–41.10.1016/0002-9378(89)90515-2Search in Google Scholar

[204] Casey ML, Cox SM, Beutler B, Milewich L, MacDonald PC. Cachectin/tumor necrosis factor-alpha formation in human decidua. Potential role of cytokines in infection-induced preterm labor. J Clin Invest. 1989;83:430–6.10.1172/JCI113901Search in Google Scholar

[205] Romero R, Parvizi ST, Oyarzun E, Mazor M, Wu YK, Avila C, et al. Amniotic fluid interleukin-1 in spontaneous labor at term. J Reprod Med. 1990;35:235–8.Search in Google Scholar

[206] Romero R, Mazor M, Brandt F, Sepulveda W, Avila C, Cotton DB, et al. Interleukin-1 alpha and interleukin-1 beta in preterm and term human parturition. Am J Reprod Immunol. 1992;27:117–23.10.1111/j.1600-0897.1992.tb00737.xSearch in Google Scholar

[207] Romero R, Tartakovsky B. The natural interleukin-1 receptor antagonist prevents interleukin-1-induced preterm delivery in mice. Am J Obstet Gynecol. 1992;167:1041–5.10.1016/S0002-9378(12)80035-4Search in Google Scholar

[208] Romero R, Sepulveda W, Kenney JS, Archer LE, Allison AC, Sehgal PB. Interleukin 6 determination in the detection of microbial invasion of the amniotic cavity. Ciba Found Symp. 1992;167:205–20; discussion 220–3.10.1002/9780470514269.ch13Search in Google Scholar

[209] Romero R, Mazor M, Sepulveda W, Avila C, Copeland D, Williams J. Tumor necrosis factor in preterm and term labor. Am J Obstet Gynecol. 1992;166:1576–87.10.1016/0002-9378(92)91636-OSearch in Google Scholar

[210] Hillier SL, Witkin SS, Krohn MA, Watts DH, Kiviat NB, Eschenbach DA. The relationship of amniotic fluid cytokines and preterm delivery, amniotic fluid infection, histologic chorioamnionitis, and chorioamnion infection. Obstet Gynecol. 1993;81:941–8.Search in Google Scholar

[211] Romero R, Yoon BH, Kenney JS, Gomez R, Allison AC, Sehgal PB. Amniotic fluid interleukin-6 determinations are of diagnostic and prognostic value in preterm labor. Am J Reprod Immunol. 1993;30:167–83.10.1111/j.1600-0897.1993.tb00618.xSearch in Google Scholar

[212] Romero R, Yoon BH, Mazor M, Gomez R, Diamond MP, Kenney JS, et al. The diagnostic and prognostic value of amniotic fluid white blood cell count, glucose, interleukin-6, and gram stain in patients with preterm labor and intact membranes. Am J Obstet Gynecol. 1993;169:805–16.10.1016/0002-9378(93)90009-8Search in Google Scholar

[213] Saito S, Kasahara T, Kato Y, Ishihara Y, Ichijo M. Elevation of amniotic fluid interleukin 6 (IL-6), IL-8 and granulocyte colony stimulating factor (G-CSF) in term and preterm parturition. Cytokine. 1993;5:81–8.10.1016/1043-4666(93)90027-3Search in Google Scholar

[214] Yoon BH, Romero R, Kim CJ, Jun JK, Gomez R, Choi JH, et al. Amniotic fluid interleukin-6: a sensitive test for antenatal diagnosis of acute inflammatory lesions of preterm placenta and prediction of perinatal morbidity. Am J Obstet Gynecol. 1995;172:960–70.10.1016/0002-9378(95)90028-4Search in Google Scholar

[215] Arntzen KJ, Kjollesdal AM, Halgunset J, Vatten L, Austgulen R. TNF, IL-1, IL-6, IL-8 and soluble TNF receptors in relation to chorioamnionitis and premature labor. J Perinat Med. 1998;26:17–26.10.1515/jpme.1998.26.1.17Search in Google Scholar

[216] Hsu CD, Meaddough E, Aversa K, Hong SF, Lu LC, Jones DC, et al. Elevated amniotic fluid levels of leukemia inhibitory factor, interleukin 6, and interleukin 8 in intra-amniotic infection. Am J Obstet Gynecol. 1998;179:1267–70.10.1016/S0002-9378(98)70144-9Search in Google Scholar

[217] Athayde N, Romero R, Maymon E, Gomez R, Pacora P, Yoon BH, et al. Interleukin 16 in pregnancy, parturition, rupture of fetal membranes, and microbial invasion of the amniotic cavity. Am J Obstet Gynecol. 2000;182:135–41.10.1016/S0002-9378(00)70502-3Search in Google Scholar

[218] Pacora P, Romero R, Maymon E, Gervasi MT, Gomez R, Edwin SS, et al. Participation of the novel cytokine interleukin 18 in the host response to intra-amniotic infection. Am J Obstet Gynecol. 2000;183:1138–43.10.1067/mob.2000.108881Search in Google Scholar PubMed

[219] Jacobsson B, Mattsby-Baltzer I, Hagberg H. Interleukin-6 and interleukin-8 in cervical and amniotic fluid: relationship to microbial invasion of the chorioamniotic membranes. BJOG. 2005;112:719–24.10.1111/j.1471-0528.2005.00536.xSearch in Google Scholar

[220] Holst RM, Laurini R, Jacobsson B, Samuelsson E, Sävman K, Doverhag C, et al. Expression of cytokines and chemokines in cervical and amniotic fluid: relationship to histological chorioamnionitis. J Matern Fetal Neonatal Med. 2007;20: 885–93.10.1080/14767050701752601Search in Google Scholar

[221] Gotsch F, Romero R, Kusanovic JP, Erez O, Espinoza J, Kim CJ, et al. The anti-inflammatory limb of the immune response in preterm labor, intra-amniotic infection/inflammation, and spontaneous parturition at term: a role for interleukin-10. J Matern Fetal Neonatal Med. 2008;21:529–47.10.1080/14767050802127349Search in Google Scholar

[222] Marconi C, de Andrade Ramos BR, Peracoli JC, Donders GG, da Silva MG. Amniotic fluid interleukin-1 beta and interleukin-6, but not interleukin-8 correlate with microbial invasion of the amniotic cavity in preterm labor. Am J Reprod Immunol. 2011;65:549–56.10.1111/j.1600-0897.2010.00940.xSearch in Google Scholar

[223] Kacerovsky M, Celec P, Vlkova B, Skogstrand K, Hougaard DM, Cobo T, et al. Amniotic fluid protein profiles of intraamniotic inflammatory response to Ureaplasma spp. and other bacteria. PLoS One. 2013;8:e60399.10.1371/journal.pone.0060399Search in Google Scholar

[224] Cobo T, Jacobsson B, Kacerovsky M, Hougaard DM, Skogstrand K, Gratacós E, et al. Systemic and local inflammatory response in women with preterm prelabor rupture of membranes. PLoS One. 2014;9:e85277.10.1371/journal.pone.0085277Search in Google Scholar

[225] Romero R, Ceska M, Avila C, Mazor M, Behnke E, Lindley I. Neutrophil attractant/activating peptide-1/interleukin-8 in term and preterm parturition. Am J Obstet Gynecol. 1991;165:813–20.10.1016/0002-9378(91)90422-NSearch in Google Scholar

[226] Cherouny PH, Pankuch GA, Romero R, Botti JJ, Kuhn DC, Demers LM, et al. Neutrophil attractant/activating peptide-1/interleukin-8: association with histologic chorioamnionitis, preterm delivery, and bioactive amniotic fluid leukoattractants. Am J Obstet Gynecol. 1993;169:1299–303.10.1016/0002-9378(93)90297-VSearch in Google Scholar

[227] Puchner T, Egarter C, Wimmer C, Lederhilger F, Weichselbraun I. Amniotic fluid interleukin-8 as a marker for intraamniotic infection. Arch Gynecol Obstet. 1993;253:9–14.10.1007/BF02770627Search in Google Scholar PubMed

[228] Dudley DJ, Trautman MS, Mitchell MD. Inflammatory mediators regulate interleukin-8 production by cultured gestational tissues: evidence for a cytokine network at the chorio-decidual interface. J Clin Endocrinol Metab. 1993;76:404–10.Search in Google Scholar

[229] Romero R, Gomez R, Galasso M, Munoz H, Acosta L, Yoon BH, et al. Macrophage inflammatory protein-1 alpha in term and preterm parturition: effect of microbial invasion of the amniotic cavity. Am J Reprod Immunol. 1994;32:108–13.10.1111/j.1600-0897.1994.tb01101.xSearch in Google Scholar PubMed

[230] Dudley DJ, Spencer S, Edwin S, Mitchell MD. Regulation of human decidual cell macrophage inflammatory protein-1 alpha (MIP-1 alpha) production by inflammatory cytokines. Am J Reprod Immunol. 1995;34:231–5.10.1111/j.1600-0897.1995.tb00946.xSearch in Google Scholar

[231] Cohen J, Ghezzi F, Romero R, Ghidini A, Mazor M, Tolosa JE, et al. GRO alpha in the fetomaternal and amniotic fluid compartments during pregnancy and parturition. Am J Reprod Immunol. 1996;35:23–9.10.1111/j.1600-0897.1996.tb00004.xSearch in Google Scholar

[232] Dudley DJ, Hunter C, Mitchell MD, Varner MW. Elevations of amniotic fluid macrophage inflammatory protein-1 alpha concentrations in women during term and preterm labor. Obstet Gynecol. 1996;87:94–8.10.1016/0029-7844(95)00366-5Search in Google Scholar

[233] Ghezzi F, Gomez R, Romero R, Yoon BH, Edwin SS, David C, et al. Elevated interleukin-8 concentrations in amniotic fluid of mothers whose neonates subsequently develop bronchopulmonary dysplasia. Eur J Obstet Gynecol Reprod Biol. 1998;78:5–10.10.1016/S0301-2115(97)00236-4Search in Google Scholar

[234] Hsu CD, Meaddough E, Aversa K, Copel JA. The role of amniotic fluid L-selectin, GRO-alpha, and interleukin-8 in the pathogenesis of intraamniotic infection. Am J Obstet Gynecol. 1998;178:428–32.10.1016/S0002-9378(98)70414-4Search in Google Scholar

[235] Athayde N, Romero R, Maymon E, Gomez R, Pacora P, Araneda H, et al. A role for the novel cytokine RANTES in pregnancy and parturition. Am J Obstet Gynecol. 1999;181:989–94.10.1016/S0002-9378(99)70337-6Search in Google Scholar

[236] Jacobsson B, Holst RM, Wennerholm UB, Andersson B, Lilja H, Hagberg H. Monocyte chemotactic protein-1 in cervical and amniotic fluid: relationship to microbial invasion of the amniotic cavity, intra-amniotic inflammation, and preterm delivery. Am J Obstet Gynecol. 2003;189:1161–7.10.1067/S0002-9378(03)00594-5Search in Google Scholar

[237] Keelan JA, Yang J, Romero RJ, Chaiworapongsa T, Marvin KW, Sato TA, et al. Epithelial cell-derived neutrophil-activating peptide-78 is present in fetal membranes and amniotic fluid at increased concentrations with intra-amniotic infection and preterm delivery. Biol Reprod. 2004;70:253–9.10.1095/biolreprod.103.016204Search in Google Scholar PubMed

[238] Chaiworapongsa T, Romero R, Espinoza J, Kim YM, Edwin S, Bujold E, et al. Macrophage migration inhibitory factor in patients with preterm parturition and microbial invasion of the amniotic cavity. J Matern Fetal Neonatal Med. 2005;18:405–16.10.1080/14767050500361703Search in Google Scholar PubMed PubMed Central

[239] Esplin MS, Romero R, Chaiworapongsa T, Kim YM, Edwin S, Gomez R, et al. Monocyte chemotactic protein-1 is increased in the amniotic fluid of women who deliver preterm in the presence or absence of intra-amniotic infection. J Matern Fetal Neonatal Med. 2005;17:365–73.10.1080/14767050500141329Search in Google Scholar PubMed

[240] Holst RM, Mattsby-Baltzer I, Wennerholm UB, Hagberg H, Jacobsson B. Interleukin-6 and interleukin-8 in cervical fluid in a population of Swedish women in preterm labor: relationship to microbial invasion of the amniotic fluid, intra-amniotic inflammation, and preterm delivery. Acta Obstet Gynecol Scand. 2005;84:551–7.10.1111/j.0001-6349.2005.00708.xSearch in Google Scholar

[241] Hamill N, Romero R, Gotsch F, Kusanovic JP, Edwin S, Erez O, et al. Exodus-1 (CCL20): evidence for the participation of this chemokine in spontaneous labor at term, preterm labor, and intrauterine infection. J Perinat Med. 2008;36:217–27.10.1515/JPM.2008.034Search in Google Scholar

[242] Cobo T, Kacerovsky M, Palacio M, Hornychova H, Hougaard DM, Skogstrand K, et al. A prediction model of histological chorioamnionitis and funisitis in preterm prelabor rupture of membranes: analyses of multiple proteins in the amniotic fluid. J Matern Fetal Neonatal Med. 2012;25:1995–2001.10.3109/14767058.2012.666592Search in Google Scholar

[243] Kacerovsky M, Musilova I, Jacobsson B, Drahosova M, Hornychova H, Janku P, et al. Cervical fluid IL-6 and IL-8 levels in pregnancies complicated by preterm prelabor rupture of membranes. J Matern Fetal Neonatal Med. 2015;28:134–40.10.3109/14767058.2014.908179Search in Google Scholar

[244] Kacerovsky M, Musilova I, Jacobsson B, Drahosova M, Hornychova H, Janku P, et al. Vaginal fluid IL-6 and IL-8 levels in pregnancies complicated by preterm prelabor membrane ruptures. J Matern Fetal Neonatal Med. 2015;28:392–8.10.3109/14767058.2014.917625Search in Google Scholar

[245] Romero R, Emamian M, Wan M, Quintero R, Hobbins JC, Mitchell MD. Prostaglandin concentrations in amniotic fluid of women with intra-amniotic infection and preterm labor. Am J Obstet Gynecol. 1987;157:1461–7.10.1016/S0002-9378(87)80245-4Search in Google Scholar

[246] Romero R, Wu YK, Mazor M, Hobbins JC, Mitchell MD. Amniotic fluid prostaglandin E2 in preterm labor. Prostaglandins Leukot Essent Fatty Acids. 1988;34:141–5.10.1016/0952-3278(88)90137-8Search in Google Scholar

[247] Romero R, Wu YK, Mazor M, Oyarzun E, Hobbins JC, Mitchell MD. Amniotic fluid arachidonate lipoxygenase metabolites in preterm labor. Prostaglandins Leukot Essent Fatty Acids. 1989;36:69–75.10.1016/0952-3278(89)90020-3Search in Google Scholar

[248] Romero R, Mazor M, Wu YK, Avila C, Oyarzun E, Mitchell MD. Bacterial endotoxin and tumor necrosis factor stimulate prostaglandin production by human decidua. Prostaglandins Leukot Essent Fatty Acids. 1989;37:183–6.10.1016/0952-3278(89)90083-5Search in Google Scholar

[249] Romero R, Wu YK, Hobbins JC, Mitchell MD. A monokine stimulates prostaglandin-E2 production by human amnion. Prostaglandins Leukot Essent Fatty Acids. 1990;41:67–9.10.1016/0952-3278(90)90133-6Search in Google Scholar

[250] Mazor M, Wiznitzer A, Maymon E, Leiberman JR, Cohen A. Changes in amniotic fluid concentrations of prostaglandins E2 and F2 alpha in women with preterm labor. Isr J Med Sci. 1990;26:425–8.Search in Google Scholar

[251] Mitchell MD, Romero RJ, Avila C, Foster JT, Edwin SS. Prostaglandin production by amnion and decidual cells in response to bacterial products. Prostaglandins Leukot Essent Fatty Acids. 1991;42:167–9.10.1016/0952-3278(91)90152-USearch in Google Scholar

[252] Mitchell MD, Romero RJ, Edwin SS, Trautman MS. Prostaglandins and parturition. Reprod Fertil Dev. 1995;7:623–32.10.1071/RD9950623Search in Google Scholar

[253] Hsu CD, Meaddough E, Aversa K, Hong SF, Lee IS, Bahodo-Singh RO, et al. Dual roles of amniotic fluid nitric oxide and prostaglandin E2 in preterm labor with intra-amniotic infection. Am J Perinatol. 1998;15:683–7.10.1055/s-2007-999302Search in Google Scholar

[254] Mikamo H, Kawazoe K, Sato Y, Imai A, Tamaya T. Preterm labor and bacterial intraamniotic infection: arachidonic acid liberation by phospholipase A2 of Fusobacterium nucleatum. Am J Obstet Gynecol. 1998;179:1579–82.10.1016/S0002-9378(98)70028-6Search in Google Scholar

[255] Hertelendy F, Rastogi P, Molnar M, Romero R. Interleukin-1beta-induced prostaglandin E2 production in human myometrial cells: role of a pertussis toxin-sensitive component. Am J Reprod Immunol. 2001;45:142–7.10.1111/j.8755-8920.2001.450304.xSearch in Google Scholar PubMed

[256] Hertelendy F, Molnar M, Romero R. Interferon gamma antagonizes interleukin-1beta-induced cyclooxygenase-2 expression and prostaglandin E(2) production in human myometrial cells. J Soc Gynecol Investig. 2002;9:215–9.Search in Google Scholar

[257] Mitchell MD, Chang MC, Chaiworapongsa T, Lan HY, Helliwell RJ, Romero R, et al. Identification of 9alpha,11beta-prostaglandin F2 in human amniotic fluid and characterization of its production by human gestational tissues. J Clin Endocrinol Metab. 2005;90:4244–8.10.1210/jc.2004-2496Search in Google Scholar PubMed

[258] Lee SE, Park IS, Romero R, Yoon BH. Amniotic fluid prostaglandin F2 increases even in sterile amniotic fluid and is an independent predictor of impending delivery in preterm premature rupture of membranes. J Matern Fetal Neonatal Med. 2009;22:880–6.10.1080/14767050902994648Search in Google Scholar PubMed PubMed Central

[259] Maddipati KR, Romero R, Chaiworapongsa T, Zhou SL, Xu Z, Tarca AL, et al. Eicosanomic profiling reveals dominance of the epoxygenase pathway in human amniotic fluid at term in spontaneous labor. FASEB J. 2014;28:4835–46.10.1096/fj.14-254383Search in Google Scholar PubMed PubMed Central

[260] Hong JS, Romero R, Lee DC, Than NG, Yeo L, Chaemsaithong P, et al. Umbilical cord prostaglandins in term and preterm parturition. J Matern Fetal Neonatal Med. 2015;1–9. [Epub ahead of print].Search in Google Scholar

[261] Park JY, Ahn TG, Lee J, Yoon BH. Elevated prostaglandin F2a concentration in amniotic fluid is an independent risk factor for intra-amniotic inflammation and adverse pregnancy outcome in patients with preterm labor. Am J Obstet Gynecol. 2015;212:S360.10.1016/j.ajog.2014.10.943Search in Google Scholar

[262] Athayde N, Romero R, Gomez R, Maymon E, Pacora P, Mazor M, et al. Matrix metalloproteinases-9 in preterm and term human parturition. J Matern Fetal Med. 1999;8:213–9.Search in Google Scholar

[263] Maymon E, Romero R, Pacora P, Gervasi MT, Bianco K, Ghezzi F, et al. Evidence for the participation of interstitial collagenase (matrix metalloproteinase 1) in preterm premature rupture of membranes. Am J Obstet Gynecol. 2000;183:914–20.10.1067/mob.2000.108879Search in Google Scholar PubMed

[264] Maymon E, Romero R, Pacora P, Gervasi MT, Gomez R, Edwin SS, et al. Evidence of in vivo differential bioavailability of the active forms of matrix metalloproteinases 9 and 2 in parturition, spontaneous rupture of membranes, and intra-amniotic infection. Am J Obstet Gynecol. 2000;183:887–94.10.1067/mob.2000.108878Search in Google Scholar

[265] Maymon E, Romero R, Pacora P, Gomez R, Athayde N, Edwin S, et al. Human neutrophil collagenase (matrix metalloproteinase 8) in parturition, premature rupture of the membranes, and intrauterine infection. Am J Obstet Gynecol. 2000;183:94–9.10.1016/S0002-9378(00)99072-0Search in Google Scholar

[266] Maymon E, Romero R, Pacora P, Gomez R, Mazor M, Edwin S, et al. A role for the 72 kDa gelatinase (MMP-2) and its inhibitor (TIMP-2) in human parturition, premature rupture of membranes and intraamniotic infection. J Perinat Med. 2001;29:308–16.10.1515/JPM.2001.044Search in Google Scholar PubMed

[267] Yoon BH, Oh SY, Romero R, Shim SS, Han SY, Park JS, et al. An elevated amniotic fluid matrix metalloproteinase-8 level at the time of mid-trimester genetic amniocentesis is a risk factor for spontaneous preterm delivery. Am J Obstet Gynecol. 2001;185:1162–7.10.1067/mob.2001.117678Search in Google Scholar PubMed

[268] Angus SR, Segel SY, Hsu CD, Locksmith GJ, Clark P, Sammel MD, et al. Amniotic fluid matrix metalloproteinase-8 indicates intra-amniotic infection. Am J Obstet Gynecol. 2001;185:1232–8.10.1067/mob.2001.118654Search in Google Scholar PubMed

[269] Romero R, Chaiworapongsa T, Espinoza J, Gomez R, Yoon BH, Edwin S, et al. Fetal plasma MMP-9 concentrations are elevated in preterm premature rupture of the membranes. Am J Obstet Gynecol. 2002;187:1125–30.10.1067/mob.2002.127312Search in Google Scholar PubMed

[270] Moon JB, Kim JC, Yoon BH, Romero R, Kim G, Oh SY, et al. Amniotic fluid matrix metalloproteinase-8 and the development of cerebral palsy. J Perinat Med. 2002;30:301–6.10.1515/JPM.2002.044Search in Google Scholar PubMed

[271] Helmig BR, Romero R, Espinoza J, Chaiworapongsa T, Bujold E, Gomez R, et al. Neutrophil elastase and secretory leukocyte protease inhibitor in prelabor rupture of membranes, parturition and intra-amniotic infection. J Matern Fetal Neonatal Med. 2002;12:237–46.10.1080/jmf.12.4.237.246Search in Google Scholar PubMed

[272] Park KH, Chaiworapongsa T, Kim YM, Espinoza J, Yoshimatsu J, Edwin S, et al. Matrix metalloproteinase 3 in parturition, premature rupture of the membranes, and microbial invasion of the amniotic cavity. J Perinat Med. 2003;31:12–22.10.1515/JPM.2003.002Search in Google Scholar PubMed

[273] Biggio JR Jr, Ramsey PS, Cliver SP, Lyon MD, Goldenberg RL, Wenstrom KD. Midtrimester amniotic fluid matrix metalloproteinase-8 (MMP-8) levels above the 90th percentile are a marker for subsequent preterm premature rupture of membranes. Am J Obstet Gynecol. 2005;192:109–13.10.1016/j.ajog.2004.06.103Search in Google Scholar PubMed PubMed Central

[274] Nien JK, Yoon BH, Espinoza J, Kusanovic JP, Erez O, Soto E, et al. A rapid MMP-8 bedside test for the detection of intra-amniotic inflammation identifies patients at risk for imminent preterm delivery. Am J Obstet Gynecol. 2006;195:1025–30.10.1016/j.ajog.2006.06.054Search in Google Scholar

[275] Kim KW, Romero R, Park HS, Park CW, Shim SS, Jun JK, et al. A rapid matrix metalloproteinase-8 bedside test for the detection of intraamniotic inflammation in women with preterm premature rupture of membranes. Am J Obstet Gynecol. 2007;197:292.e1–5.10.1016/j.ajog.2007.06.040Search in Google Scholar

[276] Kim SM, Lee J, Park C-W, Park JS, Jun JK, Yoon BH. One third of early spontaneous preterm delivery can be identified by a rapid matrix metalloproteinase-8 (MMP-8) bedside test at the time of mid-trimester genetic amniocentesis (abstract 556). Am J Obstet Gynecol. 2015;212(Suppl):S277.10.1016/j.ajog.2014.10.602Search in Google Scholar

[277] Park HS. The value of the genedia MMP-8 rapid test for diagnosing intraamniotic infection/inflammation and predicting adverse pregnancy outcomes in women with preterm premature rupture of membranes. Am J Obstet Gynecol. 2015;212(Suppl):S174.10.1016/j.ajog.2014.10.368Search in Google Scholar

[278] Gomez R, Romero R, Ghezzi F, Yoon BH, Mazor M, Berry SM. The fetal inflammatory response syndrome. Am J Obstet Gynecol. 1998;179:194–202.10.1016/S0002-9378(98)70272-8Search in Google Scholar

[279] Gotsch F, Romero R, Kusanovic JP, Mazaki-Tovi S, Pineles BL, Erez O, et al. The fetal inflammatory response syndrome. Clin Obstet Gynecol. 2007;50:652–83.10.1097/GRF.0b013e31811ebef6Search in Google Scholar

[280] Yoon BH, Romero R, Kim KS, Park JS, Ki SH, Kim BI, et al. A systemic fetal inflammatory response and the development of bronchopulmonary dysplasia. Am J Obstet Gynecol. 1999;181:773–9.10.1016/S0002-9378(99)70299-1Search in Google Scholar

[281] Dammann O, Leviton A. Role of the fetus in perinatal infection and neonatal brain damage. Curr Opin Pediatr. 2000;12:99–104.10.1097/00008480-200004000-00002Search in Google Scholar PubMed

[282] Dammann O, Kuban KC, Leviton A. Perinatal infection, fetal inflammatory response, white matter damage, and cognitive limitations in children born preterm. Ment Retard Dev Disabil Res Rev. 2002;8:46–50.10.1002/mrdd.10005Search in Google Scholar PubMed

[283] Romero R, Espinoza J, Goncalves LF, Gomez R, Medina L, Silva M, et al. Fetal cardiac dysfunction in preterm premature rupture of membranes. J Matern Fetal Neonatal Med. 2004;16:146–57.10.1080/jmf.16.3.146.157Search in Google Scholar

[284] Kim YM, Romero R, Chaiworapongsa T, Espinoza J, Mor G, Kim CJ. Dermatitis as a component of the fetal inflammatory response syndrome is associated with activation of Toll-like receptors in epidermal keratinocytes. Histopathology. 2006;49:506–14.10.1111/j.1365-2559.2006.02542.xSearch in Google Scholar

[285] Kim SK, Romero R, Chaiworapongsa T, Kusanovic JP, Mazaki-Tovi S, Mittal P, et al. Evidence of changes in the immunophenotype and metabolic characteristics (intracellular reactive oxygen radicals) of fetal, but not maternal, monocytes and granulocytes in the fetal inflammatory response syndrome. J Perinat Med. 2009;37:543–52.10.1515/JPM.2009.106Search in Google Scholar

[286] Malaeb S, Dammann O. Fetal inflammatory response and brain injury in the preterm newborn. J Child Neurol. 2009;24:1119–26.10.1177/0883073809338066Search in Google Scholar

[287] Romero R, Savasan ZA, Chaiworapongsa T, Berry SM, Kusanovic JP, Hassan SS, et al. Hematologic profile of the fetus with systemic inflammatory response syndrome. J Perinat Med. 2011;40:19–32.Search in Google Scholar

[288] Chaiworapongsa T, Romero R, Berry SM, Hassan SS, Yoon BH, Edwin S, et al. The role of granulocyte colony-stimulating factor in the neutrophilia observed in the fetal inflammatory response syndrome. J Perinat Med. 2011;39:653–66.10.1515/jpm.2011.072Search in Google Scholar

[289] Romero R, Soto E, Berry SM, Hassan SS, Kusanovic JP, Yoon BH, et al. Blood pH and gases in fetuses in preterm labor with and without systemic inflammatory response syndrome. J Matern Fetal Neonatal Med. 2012;25:1160–70.10.3109/14767058.2011.629247Search in Google Scholar

[290] Romero R, Gomez R, Ghezzi F, Yoon BH, Mazor M, Edwin SS, et al. A fetal systemic inflammatory response is followed by the spontaneous onset of preterm parturition. Am J Obstet Gynecol. 1998;179:186–93.10.1016/S0002-9378(98)70271-6Search in Google Scholar

[291] Malek TR, Bayer AL. Tolerance, not immunity, crucially depends on IL-2. Nat Rev Immunol. 2004;4:665–74.10.1038/nri1435Search in Google Scholar PubMed

[292] Boyman O, Cho JH, Sprent J. The role of interleukin-2 in memory CD8 cell differentiation. Adv Exp Med Biol. 2010;684:28–41.10.1007/978-1-4419-6451-9_3Search in Google Scholar PubMed

[293] Kalia V, Sarkar S, Subramaniam S, Haining WN, Smith KA, Ahmed R. Prolonged interleukin-2Ralpha expression on virus-specific CD8+ T cells favors terminal-effector differentiation in vivo. Immunity. 2010;32:91–103.10.1016/j.immuni.2009.11.010Search in Google Scholar PubMed

[294] Pipkin ME, Sacks JA, Cruz-Guilloty F, Lichtenheld MG, Bevan MJ, Rao A. Interleukin-2 and inflammation induce distinct transcriptional programs that promote the differentiation of effector cytolytic T cells. Immunity. 2010;32:79–90.10.1016/j.immuni.2009.11.012Search in Google Scholar PubMed PubMed Central

[295] Pepper M, Pagán AJ, Igyártó BZ, Taylor JJ, Jenkins MK. Opposing signals from the Bcl6 transcription factor and the interleukin-2 receptor generate T helper 1 central and effector memory cells. Immunity. 2011;35:583–95.10.1016/j.immuni.2011.09.009Search in Google Scholar PubMed PubMed Central

[296] Boyman O, Sprent J. The role of interleukin-2 during homeostasis and activation of the immune system. Nat Rev Immunol. 2012;12:180–90.10.1038/nri3156Search in Google Scholar PubMed

[297] Gaynor ER, Vitek L, Sticklin L, Creekmore SP, Ferraro ME, Thomas JX Jr, et al. The hemodynamic effects of treatment with interleukin-2 and lymphokine-activated killer cells. Ann Intern Med. 1988;109:953–8.10.7326/0003-4819-109-12-953Search in Google Scholar PubMed

[298] Ognibene FP, Rosenberg SA, Lotze M, Skibber J, Parker MM, Shelhamer JH, et al. Interleukin-2 administration causes reversible hemodynamic changes and left ventricular dysfunction similar to those seen in septic shock. Chest. 1988;94:750–4.10.1378/chest.94.4.750Search in Google Scholar PubMed

[299] Hack CE, Wagstaff J, Strack van Schijndel RJ, Eerenberg AJ, Pinedo HM, Thijs LG, et al. Studies on the contact system of coagulation during therapy with high doses of recombinant IL-2: implications for septic shock. Thromb Haemost. 1991;65:497–503.10.1055/s-0038-1648179Search in Google Scholar

[300] Hack CE, Ogilvie AC, Eisele B, Eerenberg AJ, Wagstaff J, Thijs LG. C1-inhibitor substitution therapy in septic shock and in the vascular leak syndrome induced by high doses of interleukin-2. Intensive Care Med. 1993;19(Suppl 1):S19–28.10.1007/BF01738946Search in Google Scholar PubMed

[301] Rosenstein M, Ettinghausen SE, Rosenberg SA. Extravasation of intravascular fluid mediated by the systemic administration of recombinant interleukin 2. J Immunol. 1986;137:1735–42.10.4049/jimmunol.137.5.1735Search in Google Scholar

[302] Rosenberg SA, Lotze MT, Muul LM, Chang AE, Avis FP, Leitman S, et al. A progress report on the treatment of 157 patients with advanced cancer using lymphokine-activated killer cells and interleukin-2 or high-dose interleukin-2 alone. N Engl J Med. 1987;316:889–97.10.1056/NEJM198704093161501Search in Google Scholar PubMed

[303] Rosenberg SA, Lotze MT, Mule JJ. NIH conference. New approaches to the immunotherapy of cancer using interleukin-2. Ann Intern Med. 1988;108:853–64.10.7326/0003-4819-108-6-853Search in Google Scholar PubMed

[304] Kovacs EJ, Brock B, Varesio L, Young HA. IL-2 induction of IL-1 beta mRNA expression in monocytes. Regulation by agents that block second messenger pathways. J Immunol. 1989;143:3532–7.10.4049/jimmunol.143.11.3532Search in Google Scholar

[305] Beutler B, Cerami A. Cachectin: more than a tumor necrosis factor. N Engl J Med. 1987;316:379–85.10.1056/NEJM198702123160705Search in Google Scholar PubMed

[306] Dinarello CA. The proinflammatory cytokines interleukin-1 and tumor necrosis factor and treatment of the septic shock syndrome. J Infect Dis. 1991;163:1177–84.10.1093/infdis/163.6.1177Search in Google Scholar PubMed

[307] Thijs LG, Hack CE, Strack van Schijndel RJ, Nuijens JH, Wolbink GJ, Eerenberg-Belmer AJ, et al. Complement activation and high-dose of interleukin-2. Lancet. 1989;2:395.10.1016/S0140-6736(89)90577-1Search in Google Scholar

[308] Thijs LG, Hack CE, Strack van Schijndel RJ, Nuijens JH, Wolbink GJ, Eerenberg-Belmer AJ, et al. Activation of the complement system during immunotherapy with recombinant IL-2. Relation to the development of side effects. J Immunol. 1990;144:2419–24.10.4049/jimmunol.144.6.2419Search in Google Scholar

[309] Vachino G, Gelfand JA, Atkins MB, Tamerius JD, Demchak P, Mier JW. Complement activation in cancer patients undergoing immunotherapy with interleukin-2 (IL-2): binding of complement and C-reactive protein by IL-2-activated lymphocytes. Blood. 1991;78:2505–13.10.1182/blood.V78.10.2505.2505Search in Google Scholar

[310] Moore FD Jr, Schoof DD, Rodrick M, Eberlein TJ. The systemic complement activation caused by interleukin-2/lymphokine-activated killer-cell therapy of cancer causes minimal systemic neutrophil activation. Int J Cancer. 1991;49:504–8.10.1002/ijc.2910490405Search in Google Scholar

[311] Baars JW, Hack CE, Wagstaff J, Eerenberg-Belmer AJ, Wolbink GJ, Thijs LG, et al. The activation of polymorphonuclear neutrophils and the complement system during immunotherapy with recombinant interleukin-2. Br J Cancer. 1992;65:96–101.10.1038/bjc.1992.18Search in Google Scholar

[312] Sagone AL Jr, Husney RM, Triozzi PL, Rinehart J. Interleukin-2 therapy enhances salicylate oxidation by blood granulocytes. Blood. 1991;78:2931–6.10.1182/blood.V78.11.2931.2931Search in Google Scholar

[313] Baars JW, de Boer JP, Wagstaff J, Roem D, Eerenberg-Belmer AJ, Nauta J, et al. Interleukin-2 induces activation of coagulation and fibrinolysis: resemblance to the changes seen during experimental endotoxaemia. Br J Haematol. 1992;82:295–301.10.1111/j.1365-2141.1992.tb06421.xSearch in Google Scholar

[314] Spear ML, Stefano JL, Fawcett P, Proujansky R. Soluble interleukin-2 receptor as a predictor of neonatal sepsis. J Pediatr. 1995;126:982–5.10.1016/S0022-3476(95)70228-8Search in Google Scholar

[315] Delogu G, Casula MA, Mancini P, Tellan G, Signore L. Serum neopterin and soluble interleukin-2 receptor for prediction of a shock state in gram-negative sepsis. J Crit Care. 1995;10:64–71.10.1016/0883-9441(95)90018-7Search in Google Scholar

[316] Pierrakos C, Vincent JL. Sepsis biomarkers: a review. Crit Care. 2010;14:R15.10.1186/cc8872Search in Google Scholar PubMed PubMed Central

[317] Fjell CD, Thair S, Hsu JL, Walley KR, Russell JA, Boyd J. Cytokines and signaling molecules predict clinical outcomes in sepsis. PLoS One. 2013;8:e79207.10.1371/journal.pone.0079207Search in Google Scholar PubMed PubMed Central

[318] Dalton DK, Pitts-Meek S, Keshav S, Figari IS, Bradley A, Stewart TA. Multiple defects of immune cell function in mice with disrupted interferon-gamma genes. Science. 1993;259:1739–42.10.1126/science.8456300Search in Google Scholar PubMed

[319] Goodbourn S, Didcock L, Randall RE. Interferons: cell signalling, immune modulation, antiviral response and virus countermeasures. J Gen Virol. 2000;81:2341–64.10.1099/0022-1317-81-10-2341Search in Google Scholar PubMed

[320] Lane HC, Fauci AS. Immunologic reconstitution in the acquired immunodeficiency syndrome. Ann Intern Med. 1985;103:714–8.10.7326/0003-4819-103-5-714Search in Google Scholar PubMed

[321] Kurzrock R, Quesada JR, Rosenblum MG, Sherwin SA, Gutterman JU. Phase I study of i.v. administered recombinant gamma interferon in cancer patients. Cancer Treat Rep. 1986;70:1357–64.Search in Google Scholar

[322] Horning SJ, Levine JF, Miller RA, Rosenberg SA, Merigan TC. Clinical and immunologic effects of recombinant leukocyte A interferon in eight patients with advanced cancer. J Am Med Assoc. 1982;247:1718–22.10.1001/jama.1982.03320370032025Search in Google Scholar

[323] Dinarello CA. Cytokines as endogenous pyrogens. J Infect Dis. 1999;179(Suppl 2):S294–304.10.1086/513856Search in Google Scholar PubMed

[324] Helfgott DC, Tatter SB, Santhanam U, Clarick RH, Bhardwaj N, May LT, et al. Multiple forms of IFN-beta 2/IL-6 in serum and body fluids during acute bacterial infection. J Immunol. 1989;142:948–53.10.4049/jimmunol.142.3.948Search in Google Scholar

[325] Fong Y, Moldawer LL, Shires GT, Lowry SF. The biologic characteristics of cytokines and their implication in surgical injury. Surg Gynecol Obstet. 1990;170:363–78.Search in Google Scholar

[326] Schluter B, Konig B, Bergmann U, Müller FE, König W. Interleukin 6 – a potential mediator of lethal sepsis after major thermal trauma: evidence for increased IL-6 production by peripheral blood mononuclear cells. J Trauma. 1991;31:1663–70.10.1097/00005373-199112000-00017Search in Google Scholar PubMed

[327] Akira S, Kishimoto T. IL-6 and NF-IL6 in acute-phase response and viral infection. Immunol Rev. 1992;127:25–50.10.1111/j.1600-065X.1992.tb01407.xSearch in Google Scholar

[328] Youn YK, LaLonde C, Demling R. The role of mediators in the response to thermal injury. World J Surg. 1992;16:30–6.10.1007/BF02067111Search in Google Scholar PubMed

[329] Pang G, Couch L, Batey R, Clancy R, Cripps A. GM-CSF, IL-1 alpha, IL-1 beta, IL-6, IL-8, IL-10, ICAM-1 and VCAM-1 gene expression and cytokine production in human duodenal fibroblasts stimulated with lipopolysaccharide, IL-1 alpha and TNF-alpha. Clin Exp Immunol. 1994;96:437–43.10.1111/j.1365-2249.1994.tb06048.xSearch in Google Scholar PubMed PubMed Central

[330] Jung HC, Eckmann L, Yang SK, Panja A, Fierer J, Morzycka-Wroblewska E, et al. A distinct array of proinflammatory cytokines is expressed in human colon epithelial cells in response to bacterial invasion. J Clin Invest. 1995;95:55–65.10.1172/JCI117676Search in Google Scholar PubMed PubMed Central

[331] Wei SQ, Fraser W, Luo ZC. Inflammatory cytokines and spontaneous preterm birth in asymptomatic women: a systematic review. Obstet Gynecol. 2010;116:393–401.10.1097/AOG.0b013e3181e6dbc0Search in Google Scholar

[332] Evers AC, Nijhuis L, Koster MP, Bont LJ, Visser GH. Intrapartum fever at term: diagnostic markers to individualize the risk of fetal infection: a review. Obstet Gynecol Surv. 2012;67:187–200.10.1097/OGX.0b013e31824bb5f1Search in Google Scholar

[333] Goetzl L, Evans T, Rivers J, Suresh MS, Lieberman E. Elevated maternal and fetal serum interleukin-6 levels are associated with epidural fever. Am J Obstet Gynecol. 2002;187:834–8.10.1067/mob.2002.127135Search in Google Scholar

[334] Wang LZ, Hu XX, Liu X, Qian P, Ge JM, Tang BL. Influence of epidural dexamethasone on maternal temperature and serum cytokine concentration after labor epidural analgesia. Int J Gynaecol Obstet. 2011;113:40–3.10.1016/j.ijgo.2010.10.026Search in Google Scholar

[335] Opsjln SL, Wathen NC, Tingulstad S, Wiedswang G, Sundan A, Waage A, et al. Tumor necrosis factor, interleukin-1, and interleukin-6 in normal human pregnancy. Am J Obstet Gynecol. 1993;169:397–404.10.1016/0002-9378(93)90096-2Search in Google Scholar

[336] Austgulen R, Lien E, Liabakk NB, Jacobsen G, Arntzen KJ. Increased levels of cytokines and cytokine activity modifiers in normal pregnancy. Eur J Obstet Gynecol Reprod Biol. 1994;57:149–55.10.1016/0028-2243(94)90291-7Search in Google Scholar

[337] Arntzen KJ, Lien E, Austgulen R. Maternal serum levels of interleukin-6 and clinical characteristics of normal delivery at term. Acta Obstet Gynecol Scand. 1997;76:55–60.10.3109/00016349709047785Search in Google Scholar

[338] Greig PC, Murtha AP, Jimmerson CJ, Herbert WN, Roitman-Johnson B, Allen J. Maternal serum interleukin-6 during pregnancy and during term and preterm labor. Obstet Gynecol. 1997;90:465–9.10.1016/S0029-7844(97)00294-9Search in Google Scholar

[339] Hebisch G, Grauaug AA, Neumaier-Wagner PM, Stallmach T, Huch A, Huch R. The relationship between cervical dilatation, interleukin-6 and interleukin-8 during term labor. Acta Obstet Gynecol Scand. 2001;80:840–8.10.1034/j.1600-0412.2001.080009840.xSearch in Google Scholar PubMed

[340] Malamitsi-Puchner A, Protonotariou E, Boutsikou T, Makrakis E, Sarandakou A, Creatsas G. The influence of the mode of delivery on circulating cytokine concentrations in the perinatal period. Early Hum Dev. 2005;81:387–92.10.1016/j.earlhumdev.2004.10.017Search in Google Scholar PubMed

[341] Christiaens I, Zaragoza DB, Guilbert L, Robertson SA, Mitchell BF, Olson DM. Inflammatory processes in preterm and term parturition. J Reprod Immunol. 2008;79:50–7.10.1016/j.jri.2008.04.002Search in Google Scholar

[342] Unal ER, Cierny JT, Roedner C, Newman R, Goetzl L. Maternal inflammation in spontaneous term labor. Am J Obstet Gynecol. 2011;204:223.e1–5.10.1016/j.ajog.2011.01.002Search in Google Scholar

[343] Cierny J, Unal E, Flood P, Rhee KY, Praktish A, Olson T, et al. Inflammatory cytokines, fever and term labor performance. Am J Obstet Gynecol. 2013;208:S144.10.1016/j.ajog.2012.10.487Search in Google Scholar

[344] Cierny JT, Unal ER, Flood P, Rhee KY, Praktish A, Olson TH, et al. Maternal inflammatory markers and term labor performance. Am J Obstet Gynecol. 2014;210:447.e1–6.10.1016/j.ajog.2013.11.038Search in Google Scholar

[345] Lee SE, Romero R, Jung H, Park CW, Park JS, Yoon BH. The intensity of the fetal inflammatory response in intraamniotic inflammation with and without microbial invasion of the amniotic cavity. Am J Obstet Gynecol. 2007;197:294.e1–6.10.1016/j.ajog.2007.07.006Search in Google Scholar

[346] Armstrong-Wells J, Donnelly M, Post MD, Manco-Johnson MJ, Winn VD, Sébire G. Inflammatory predictors of neurologic disability after preterm premature rupture of membranes. Am J Obstet Gynecol. 2015;212:212.e1–9.10.1016/j.ajog.2014.09.016Search in Google Scholar

[347] Romero R, Yoon BH, Mazor M, Gomez R, Gonzalez R, Diamond MP, et al. A comparative study of the diagnostic performance of amniotic fluid glucose, white blood cell count, interleukin-6, and gram stain in the detection of microbial invasion in patients with preterm premature rupture of membranes. Am J Obstet Gynecol. 1993;169:839–51.10.1016/0002-9378(93)90014-ASearch in Google Scholar

[348] Coultrip LL, Lien JM, Gomez R, Kapernick P, Khoury A, Grossman JH. The value of amniotic fluid interleukin-6 determination in patients with preterm labor and intact membranes in the detection of microbial invasion of the amniotic cavity. Am J Obstet Gynecol. 1994;171:901–11.10.1016/S0002-9378(94)70057-5Search in Google Scholar

[349] Greci LS, Gilson GJ, Nevils B, Izquierdo LA, Qualls CR, Curet LB. Is amniotic fluid analysis the key to preterm labor? A model using interleukin-6 for predicting rapid delivery. Am J Obstet Gynecol. 1998;179:172–8.10.1016/S0002-9378(98)70269-8Search in Google Scholar

[350] El-Bastawissi AY, Williams MA, Riley DE, Hitti J, Krieger JN. Amniotic fluid interleukin-6 and preterm delivery: a review. Obstet Gynecol. 2000;95:1056–64.10.1097/00006250-200006001-00032Search in Google Scholar

[351] Conde-Agudelo A, Papageorghiou AT, Kennedy SH, Villar J. Novel biomarkers for the prediction of the spontaneous preterm birth phenotype: a systematic review and meta-analysis. BJOG. 2011;118:1042–54.10.1111/j.1471-0528.2011.02923.xSearch in Google Scholar PubMed

[352] Cobo T, Kacerovsky M, Holst RM, Hougaard DM, Skogstrand K, Wennerholm UB, et al. Intra-amniotic inflammation predicts microbial invasion of the amniotic cavity but not spontaneous preterm delivery in preterm prelabor membrane rupture. Acta Obstet Gynecol Scand. 2012;91:930–5.10.1111/j.1600-0412.2012.01427.xSearch in Google Scholar PubMed

[353] Romero R, Kadar N, Miranda J, Korzeniewski SJ, Schwartz AG, Chaemsaithong P, et al. The diagnostic performance of the Mass Restricted (MR) score in the identification of microbial invasion of the amniotic cavity or intra-amniotic inflammation is not superior to amniotic fluid interleukin-6. J Matern Fetal Neonatal Med. 2014;27:757–69.10.3109/14767058.2013.844123Search in Google Scholar PubMed PubMed Central

[354] Vousden N, Chandiramani M, Seed P, Shennan A. Interleukin-6 bedside testing in women at high risk of preterm birth. J Matern Fetal Neonatal Med. 2011;24:1301–4.10.3109/14767058.2011.558954Search in Google Scholar PubMed

[355] Kacerovsky M, Musilova I, Hornychova H, Kutova R, Pliskova L, Kostal M, et al. Bedside assessment of amniotic fluid interleukin-6 in preterm prelabor rupture of membranes. Am J Obstet Gynecol. 2014;211:385.e1–9.10.1016/j.ajog.2014.03.069Search in Google Scholar PubMed

[356] Berthiaume M, Rousseau E, Rola-Pleszczynski M, Pasquier JC. Rapid evaluation of the absence of inflammation after rupture of membranes. J Matern Fetal Neonatal Med. 2014;27:865–9.10.3109/14767058.2013.829814Search in Google Scholar PubMed

[357] Lee SM, Romero R, Park JS, Chaemsaithong P, Jun JK, Yoon BH. A transcervical amniotic fluid collector: a new medical device for the assessment of amniotic fluid in patients with ruptured membranes. J Perinat Med. 2015;43:381–9.10.1515/jpm-2014-0276Search in Google Scholar PubMed PubMed Central

The authors stated that there are no conflicts of interest regarding the publication of this article.


Supplemental Material:

The online version of this article (DOI: 10.1515/jpm-2015-0103) offers supplementary material, available to authorized users.


Received: 2015-3-23
Accepted: 2015-4-17
Published Online: 2015-8-4
Published in Print: 2016-1-1

©2016 by De Gruyter

Downloaded on 27.4.2024 from https://www.degruyter.com/document/doi/10.1515/jpm-2015-0103/html
Scroll to top button