Skip to content
Publicly Available Published by De Gruyter September 1, 2017

Fetal death: an extreme manifestation of maternal anti-fetal rejection

  • Kia Lannaman , Roberto Romero EMAIL logo , Tinnakorn Chaiworapongsa , Yeon Mee Kim , Steven J. Korzeniewski , Eli Maymon , Nardhy Gomez-Lopez , Bogdan Panaitescu , Sonia S. Hassan , Lami Yeo , Bo Hyun Yoon , Chong Jai Kim and Offer Erez EMAIL logo

Abstract

Objective:

The aim of this study was to determine the association between chronic placental inflammation and amniotic fluid (AF) markers of maternal anti-fetal rejection as well as the presence of microorganisms in the AF fluid of patients with fetal death.

Study Design:

This cohort study included 40 patients with fetal death whose placentas were examined for chronic inflammatory lesions and whose AF chemokine ligand (CXCL)10 and interleukin (IL)-6 concentrations were determined by immunoassays. AF was processed for bacteria, mycoplasmas and viruses using cultivation and molecular microbiologic techniques (i.e. PCR-ESI/MS).

Results:

(1) The most prevalent placental findings were maternal vascular underperfusion (63.2%, 24/38), followed by chronic inflammatory lesions (57.9%, 22/38); (2) chronic chorioamnionitis (18/38) was three times more frequent than villitis of unknown etiology (6/38); (3) an elevated AF CXCL10 concentration (above the 95th centile) was present in 60% of the cases, and a receiver operating characteristics (ROC)-derived cut-off of 2.9 ng/mL had a sensitivity of 73% and a specificity of 75% in the identification of chronic placental inflammatory lesions; (4) only five cases had microbial invasion of the amniotic cavity, and the presence of microorganisms did not correlate with chronic placental inflammation.

Conclusion:

In women with unexplained fetal death, there is an association between elevated AF CXCL10 and chronic placental inflammatory lesions. Therefore, we conclude that a subset of patients with fetal death may have endured a breakdown of maternal-fetal tolerance, which cannot be attributed to microorganisms in the amniotic cavity.

Introduction

Fetal death, characterized by the cessation of heart activity after 20 weeks of gestation [1], is considered one of the “great obstetrical syndromes” [2], [3], [4] caused by multiple etiologies, including failure of the placenta to provide oxygen/nutrients to support fetal growth [5], [6], [7], [8], [9], [10], [11], [12], [13], [14]; infections such as syphilis [15], [16], [17], [18], [19], malaria [16], [20], [21], [22], Zika virus [23], [24], [25] and Listeria monocytogenes [26], [27], [28], [29], [30]; fetal cardiac arrhythmias [31], [32]; congenital anomalies [6], [33], [34], [35], [36]; umbilical cord accidents (i.e. “true” knots of the umbilical cord) [10], [35], [36], [37], [38], [39] and complications of monochorial placentation in multiple gestation pregnancies [40], [41], [42].

Affecting more than three million pregnancies per year, fetal death is a clinical challenge; at present, there are no good methods to assess the magnitude of risk or to prevent this devastating complication of pregnancy [43], [44], [45]. Although several classification systems have been proposed and are used to attempt to identify co-morbid events associated with fetal death [36], [46], [47], [48], [49], [50], [51], [52], [53], [54], [55], [56], [57], [58], [59], [60], [61], [62], [63], a firm causal link between the events labeled in these classifications as “causes” of fetal death and the actual cause of death is extremely difficult to prove [39], [49], [58], [64], [65], [66]. Therefore, these classification systems provide a general idea of the context in which fetal death occurred, rather than providing unequivocal proof of the cause of death (e.g. total placental abruption [67], [68] or maternal trauma [69], [70], [71]).

The fetus and placenta are semi-allografts to the maternal host, as 50% of their genomes are paternally derived [72], [73], [74], [75], [76]. The placenta is widely considered to be the most successful transplant in nature, a biological adaptation required for viviparity, and accomplished by immune tolerance [72], [76], [77], [78], [79], [80], [81], [82], [83], [84], [85], [86]. Sir Peter Medawar, the founder of the field of Reproductive Immunology, crafted the fundamental question of the field [87], [88]:

The immunological problem of pregnancy may be formulated thus: how does the pregnant mother contrive to nourish within itself, for many weeks or months, a foetus that is an antigenically foreign body?”

In other words, why are the fetus and placenta not rejected?

Tolerance, “the active state of antigen-specific non-responsiveness,” leading to diminished reactivity to paternal antigens expressed by the placenta and/or fetus, is considered key for a successful pregnancy [75], [76], [77], [78], [83], [84], [85], [86], [89], [90], [91], [92], [93], [94], [95], [96], [97], [98], [99], [100], [101], [102], [103], [104]. The mechanisms responsible for tolerance in pregnancy have been reviewed by Elizabeth Bonney [90], [91], [93], [104], [105], [106], Sing-Sing Way [107], [108], and Adrian Erlebacher [72], [86], [109], [110], [111], [112].

A fundamental question, largely overlooked, is whether semi-allograft rejection mediated by a breakdown of tolerance is a mechanism of disease responsible for adverse pregnancy outcome. Transplant rejection is characterized by the infiltration of donor CD8+ (cytotoxic) T cells into the graft and by organ dysfunction or failure [113], [114], [115]. Renal transplant rejection is diagnosed in patients who have a drop in the glomerular filtration rate (i.e. creatinine clearance) and an infiltration of lymphocytes from the donor into the transplanted organ [116], [117]. The presence of maternal lymphocytes at the maternofetal interfaces, such as the chorioamniotic membranes and villous tree, as seen in chronic chorioamnionitis and villitis of unknown etiology (VUE), respectively, demonstrates a breakdown in maternal (host) tolerance of the fetus (graft) [118], [119], [120], [121], [122], [123]. In parallel fashion, chemokine markers of chronic inflammation, such as chemokine ligand 10 (CXCL10) were elevated in the amniotic fluid (AF) when signs of rejection were identified by placental histology [118], [121], [123], [124].

We have provided evidence that maternal anti-fetal rejection is operative in a subset of patients with spontaneous preterm labor [118], [119], [120], [122], [124], [125], [126], preterm prelabor rupture of the membranes (PPROM) [118], maternal floor infarction [127], and other obstetrical syndromes [121], [122], [126]. Three placental pathology lesions have been considered manifestations of maternal anti-fetal rejection: (1) chronic chorioamnionitis [118], [119], [120], [121], [122], [124], [126], [128], [129]; (2) VUE [122], [123], [126], [130], [131]; and (3) chronic deciduitis [122], [126], [129]. In these lesions, maternal lymphocytes infiltrate the fetal tissue (the chorioamniotic membranes in chronic chorioamnionitis, the villous tree in VUE, and the basal plate of the placenta in chronic deciduitis) [118], [123], [126], [130], [131], [132], [133], [134].

The ultimate form of transplant rejection is a total failure of the transplanted organ, which manifests itself as renal failure [117], cardiac failure [135], etc. Fetal death in the context of maternal anti-fetal rejection can be considered as the most severe expression of such a rejection. We previously reported that chronic chorioamnionitis is frequent in patients with fetal death [121], [126]; however, a pervasive question is whether some cases of chronic chorioamnionitis may be due to subclinical infection rather than to maternal anti-fetal rejection. Therefore, we conducted this study to determine whether fetal death associated with placental lesions consistent with maternal anti-fetal rejection is related to intra-amniotic infection. This clinical condition was diagnosed by the presence of bacteria and/or viruses using cultivation and molecular microbiologic techniques.

Materials and methods

Patient population and study materials

This cohort study was performed at Hutzel Women’s Hospital, Detroit, MI, USA. Subjects included patients who presented with unexplained fetal death (n=40) defined as spontaneous death of a fetus at or after 20 weeks of gestation and not attributed to an identifiable cause. Patients with a multifetal gestation, pre-gestational diabetes, and prenatal diagnoses of fetal anomalies as well as chromosomal abnormalities were excluded from the study.

Patients diagnosed with fetal death by ultrasound were offered amniocentesis for karyotype determination and assessment for the presence of intra-amniotic infection. We selected cases in which AF samples were available from the Bank of Biological Materials of the Perinatology Research Branch, Wayne State University, and the Detroit Medical Center. Each participant provided written informed consent for the use of their information and biological specimens for research. The Institutional Review Boards of Wayne State University and the Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), approved the collection of clinical information and use of biological materials for research purposes.

Small-for-gestational age was defined as birthweight less than the 10th percentile for gestational age [136]. Chronic maternal condition was defined as a chronic medical illness affecting an organ system that may require medication (e.g. asthma, lupus). Prior preterm birth was defined as a pregnancy delivering at or after 20+0 and before 37+0 weeks of gestation. Preeclampsia was defined as the presence of new-onset hypertension and proteinuria at or beyond 20+0 weeks of gestation [137]. Mean arterial blood pressure [138] was defined as diastolic blood pressure + (1/3*pulse pressure). The use of illicit drugs was determined by urine drug screening. Microbial invasion of the amniotic cavity was defined as a positive culture or positive result from molecular microbiologic techniques [139]. Clinical chorioamnionitis and placental abruption were diagnosed by physicians in the clinical setting.

Histopathologic examination

Histopathologic assessment of placental lesions was performed by examination of hematoxylin and eosin (H&E)-stained sections of the chorioamniotic membranes, placental disc and umbilical cord in each case. Diagnoses of findings were made by perinatal pathologists, according to previously published criteria [118], [126], [140], [141], [142], [143], [144]. The following categories of placental findings were characterized: findings consistent with amniotic fluid infection (AFI), fetal vascular thrombo-occlusive disease, maternal vascular underperfusion, and chronic inflammatory lesions. Sub-classes of chronic inflammatory lesions included in this study were chronic chorioamnionitis, VUE and chronic deciduitis. Chronic chorioamnionitis was diagnosed when lymphocytic infiltrates were observed in the chorionic trophoblast layer or chorioamniotic connective tissue [118], [126] (Figure 1). The severity of inflammation was scored on grade and stage. The extent of inflammation was graded as 0 (no inflammation), 1 (more than two foci or patchy inflammation), or 2 (diffuse inflammation). The stage of inflammation was scored as stage 1 if amniotropic lymphocytic infiltration was limited to the chorionic trophoblast layer sparing the chorioamniotic connective tissue, and stage 2 if lymphocytic infiltration into the chorioamniotic connective tissue was noted. Villitis of unknown etiology was defined as the presence of lymphohistiocytic infiltrates in the chorionic villi [126], [144], [145]. Grading of VUE is based on the number of affected villi and the extent of involvement. Low-grade lesions affect less than 10 villi; high-grade lesions affect more than 10 villi. VUE can be characterized by whether the pattern of involvement is distal (terminal and intermediate villi), proximal (stem villi), or basal (anchoring villi). In this study, the pattern of VUE was characterized as either proximal or basal, according to the protocol at our institution. Chronic deciduitis was diagnosed by the presence of both lymphocytes and plasma cells in the basal plate of the placenta [133].

Figure 1: Histology of the extraplacental chorioamniotic membranes.(A) Normal chorioamniotic membranes with no evidence of chronic inflammatory cell infiltration in the amnion and choriodecidua. (B) Chronic chorioamnionitis, stage 2: chorioamniotic membranes from FD, gestational age 31.4 weeks. Chronic inflammatory cells (arrows) infiltrate into the chorioamniotic connective tissues beyond the chorionic trophoblast layer. FD=Fetal death.
Figure 1:

Histology of the extraplacental chorioamniotic membranes.

(A) Normal chorioamniotic membranes with no evidence of chronic inflammatory cell infiltration in the amnion and choriodecidua. (B) Chronic chorioamnionitis, stage 2: chorioamniotic membranes from FD, gestational age 31.4 weeks. Chronic inflammatory cells (arrows) infiltrate into the chorioamniotic connective tissues beyond the chorionic trophoblast layer. FD=Fetal death.

Enzyme-linked immunosorbent assay for CXCL10 and IL-6

Amniotic fluid concentrations of CXCL10 and interleukin (IL)-6 were measured by specific ELISA assays (R&D Systems, Minneapolis, MN, USA), according to the manufacturer’s instructions. The sensitivity of the assay for AF CXCL10 and IL-6 was 6.82 pg/mL and 2.24 pg/mL, respectively. The inter-assay coefficient of variation was 2.75% for CXCL10 and 2.84% for IL-6; the intra-assay coefficient of variation was 3.87% and 3.68%, respectively. Gervasi et al. [146] reported reference ranges for AF CXCL10 sampled in the mid-trimester for pregnancies later delivered at term without complications; the 95th percentile for CXCL10 was defined as 2.2 ng/mL and this cut-off was used in our study [146]. IL-6 is an inflammation-related cytokine associated with systemic inflammation in the maternal [147], fetal [148], [149], and placental compartments [150] as well as with intra-amniotic infection and intra-amniotic inflammation [146], [151], [152], [153], [154], [155], [156], [157], [158], [159], [160], [161], [162], [163], [164], [165], [166], [167]. We used a cut-off of 2.6 ng/mL for the identification of intra-amniotic inflammation [157], [168], [169].

Microbiologic evaluation

Amniotic fluid was transported in a capped sterile syringe to the clinical laboratory where it was cultured for aerobic and anaerobic bacteria, including genital Mycoplasmas and viruses. Determinations of the AF white blood cell count [170], glucose concentration [171] and Gram stain [172] were performed first. The AF was then centrifuged at 1300× g for 10 min at 4°C and then stored at −80°C until analyzed by immunoassay. The analysis for the presence of bacteria, genital mycoplasmas and/or viruses was performed by cultivation and broad-range polymerase chain reaction (PCR) coupled with electrospray ionization mass spectrometry (PCR/ESI-MS) (Ibis® Technology, Athogen, Carlsbad, CA, USA) [139], [173], [174], [175], [176], [177], [178], [179], [180], [181].

Statistical analysis

Distributions of continuous variables were examined for skewness and normality. The Shapiro-Wilk test was performed to assess deviations of arithmetic data from normality. Bivariate analysis was performed with the Mann-Whitney U and Kruskal-Wallis tests for the assessment of non-parametric continuous variables (e.g. CXCL10, IL-6). Spearman’s rho was used to assess correlations. Odds ratios and 95% confidence intervals (CI) were calculated to assess magnitudes of association. Statistical analysis was performed using SPSS Version 19 (SPSS Inc., Chicago, IL, USA). A two-tailed P value of <0.05 was considered significant.

Results

Study population

Forty patients with a fetal death met the inclusion criteria. Table 1 provides the clinical characteristics, obstetrical histories and laboratory results of the AF samples. Patients of African-American origin comprised 87.5% (35/40) of the study population. Only one patient had a prior fetal demise. The median gestational age at presentation was 30.9 weeks, indicating that most fetal deaths occurred in the third trimester, and 27.5% of fetal deaths occurred in small-for-gestational-age cases. The median interval from diagnosis to delivery was 0.1 weeks [range 0–0.5 weeks], one patient delivered spontaneously, 37 had induction of labor, and two were delivered by cesarean section. A Kleihauer-Betke test was performed in 33 patients and only one had a positive result.

Table 1:

Clinical, obstetrical and laboratory results of patients with fetal death.

Stillbirths (n=40)
Maternal age (years)25.0 (±4.6)
Race/ethnicity
African American87.5% (35/40)
Caucasian2.5% (1/40)
Hispanic5% (2/40)
Other5% (2/40)
Body mass index (kg/m2)30.2 (±8.7)
Mean arterial pressure (mm Hg)95 (±13)
Nulliparity37.5% (15/40)
Prior preterm birth17.5% (7/40)
Maternal medical comorbiditiesa37.5% (15/40)
Active tobacco smoker20% (8/40)
Drug use during index pregnancy (positive urine drug screen)28.2% (11/39)
Gestational age at diagnosis and amniocentesis (weeks)30.9 [25.2–35.7]
Interval from diagnosis to delivery (weeks)0.1 [0–0.5]
Induction of labor92.5% (37/40)
Stillborn birthweight (g)1403 [625–2283]
Birthweight <10th percentile27.5% (11/40)
Male fetus57.5% (23/40)
Kleihauer-Betke positive3% (1/33)
Amniotic fluid
White blood cell count (cells/mL3)1 [0–13]
Glucose (mg/dL)18 [10–22]
CXCL-10 (ng/mL)2.9 [1.7–7. 6]
IL-6 (ng/mL)2.2 [0.5–5.8]
Positive bacterial culture2.5% (1/40)
Positive viral culture2.9% (1/34)
Positive bacterial molecular study2.5% (1/40)
Positive viral molecular study10% (4/40)
  1. Data presented as median [IQR] or mean (±SD) and % (n/N). aMedical comorbidities included (n): asthma (7), cerebral palsy (1), chronic hypertension (3), Crohn’s disease (1), drug abuse (2), gastroesophageal reflux disease (1), liver disease (1), systemic lupus erythematosus (1), psychiatric mood disorder (1), Wiskott-Aldridge carrier (1).

Placental lesions

The most frequent lesions were those consistent with maternal vascular underperfusion (63.2%; 24/38), followed by chronic inflammatory lesions (57.9%; 22/38), acute inflammatory lesions consistent with AFI (28.2%; 11/39), and fetal vascular thrombo-occlusive disease (23.1%; 9/39) (Figure 2). More than one type of lesion was diagnosed in 61.5% (24/39) of the placentas. The most frequent combination of lesions (14/22) included those consistent with maternal vascular underperfusion and chronic placental inflammation (Figure 3).

Figure 2: Frequency of placental lesions in fetal death.The classification of placental lesions was based on those categorized by the Society for Pediatric Pathology – Chronic inflammatory lesions encompassed chronic chorioamnionitis, villitis of unknown etiology and chronic deciduitis. Chronic inflammatory lesions were the second most predominant histopathological lesion observed in unexplained fetal death.
Figure 2:

Frequency of placental lesions in fetal death.

The classification of placental lesions was based on those categorized by the Society for Pediatric Pathology – Chronic inflammatory lesions encompassed chronic chorioamnionitis, villitis of unknown etiology and chronic deciduitis. Chronic inflammatory lesions were the second most predominant histopathological lesion observed in unexplained fetal death.

Figure 3: Frequency of isolated and mixed placental lesions in unexplained fetal death.The placental examination demonstrated lesions from multiple histological classes (mixed) more frequently than those confined to one class (isolated). The most abundant findings were lesions of maternal vascular underperfusion and chronic inflammation. Some placentas even had lesions from 3 different classes. The frequency of isolated lesions is represented in solid colors and the frequency of mixed lesions in colors derived from the blend of the classes involved. AFI=Amniotic fluid infection, CIL=chronic inflammatory lesion, FVTO=fetal vascular thrombo-occlusive disease, MVU=maternal vascular underperfusion.
Figure 3:

Frequency of isolated and mixed placental lesions in unexplained fetal death.

The placental examination demonstrated lesions from multiple histological classes (mixed) more frequently than those confined to one class (isolated). The most abundant findings were lesions of maternal vascular underperfusion and chronic inflammation. Some placentas even had lesions from 3 different classes. The frequency of isolated lesions is represented in solid colors and the frequency of mixed lesions in colors derived from the blend of the classes involved. AFI=Amniotic fluid infection, CIL=chronic inflammatory lesion, FVTO=fetal vascular thrombo-occlusive disease, MVU=maternal vascular underperfusion.

Chronic placental inflammatory lesions consisted of 18 cases with chronic chorioamnionitis (17/18 cases were preterm gestation), six cases with VUE (5/6 cases were preterm gestation) and seven cases with chronic deciduitis. In regard to the staging and grading of chronic chorioamnionitis, 61.1% (11/18) were stage 2 (involvement of the chorionic connective tissue) (Figure 1), 38.9% (7/18) were grade 2 (a diffuse, rather than localized, inflammatory process), and 33.3% (6/18) also involved the chorionic plate. In four of the cases, chronic deciduitis was also present.

All six cases of VUE had basal, low-grade lesions (five cases were preterm gestation). In all six cases, the AF was negative for bacteria and viruses. In one-half of the VUE cases (3/6), there was evidence of chronic chorioamnionitis, and equally in 3/6 cases, there was chronic deciduitis.

Amniotic fluid concentrations of CXCL10 and interleukin-6 in fetal death

The median AF concentration of CXCL10 (a chemokine elevated in cases of allograft rejection) was 2.9 ng/mL [IQR 1.7–7.6] – this is higher than the 95th centile of CXCL10 measured in the mid-trimester AF (2.2 ng/mL) of patients with a normal pregnancy outcome (see Supplemental Material Figure 1). An elevated AF CXCL10 concentration was present in 60% (24/40) of patients with a fetal death.

The AF CXCL10 concentration was elevated for 17 of 22 (77.2%) patients with chronic placental inflammatory lesions, including two cases with both chronic chorioamnionitis and VUE – this included 13 cases with chronic chorioamnionitis and five with VUE. There was a positive correlation between the concentration of CXCL10 and the occurrence of chronic placental inflammatory lesions (r=0.37, P=0.02). We found no correlation between gestational age and CXCL10 for the entire study population (r=−0.23; P=0.16).

The median AF concentration of CXCL10 in patients with and without chronic placental inflammatory lesions was [5.2 (2.2–9.2) vs. 2.1 (1.6–3.3), P=0.01], respectively. In the current study, a receiver operating characteristics (ROC) curve was plotted using sensitivity and 1-specificity; an AF CXCL10 concentration of 2.9 ng/mL had a sensitivity of 73%, a specificity of 75%, a positive likelihood ratio of 2.9, and a negative likelihood ratio of 0.4 with an area under the curve of 0.72 (P=0.02) for the presence of chronic placental inflammatory lesions. Of note, using the ROC curve, the previously reported cut-off for the 95th percentile of CXCL10 (2.2 ng/mL) [146] would have had a sensitivity of 77%, a specificity of 56%, a positive likelihood ratio of 1.8, and a negative likelihood ratio of 0.4 for the presence of placental lesions consistent with chronic inflammation.

Forty-five percent (18/40) of patients had intra-amniotic inflammation (defined as an AF IL-6 concentration ≥2.6 ng/ml) [157], [168], [169]. The median concentration of AF IL-6 was 2.2 [0.5–5.8] ng/ml; neither acute inflammatory lesions of the placenta nor microbial invasion of the amniotic cavity was associated with AF IL-6 (P=0.14, P=0.26, respectively).

In 13 of 40 (32.5%) patients, both AF markers of acute (IL-6) and chronic (CXCL10) inflammation were greater than the 95th percentile. The odds of co-occurrence of elevated AF CXCL10 and IL-6 were nine times higher among patients who had mixed placental histologic lesions compared to those with an isolated lesion or no lesion (OR 9; 95% CI 1.2–62); adjustment for gestational age at diagnosis slightly increased the odds ratio (OR 11; 1.4–83).

Microbial invasion of the amniotic cavity

A positive AF result for microorganisms was present in 12.5% (5/40) of the study population (Table 1). In most cases, the organisms were viruses (i.e. parvovirus); in one case, it was Lactobacillus sakei (Table 2). The organisms, clinical characteristics, results of AF analysis, IL-6, and CXCL10, as well as the results of placental pathologic findings, are described in Table 2.

Table 2:

Microbial invasion of the amniotic cavity in fetal death: microorganisms, amniotic fluid analysis and placental findings.

OrganismCulturePCRGenome/wellGA diagnosis/amniocentesis (weeks)AF WBC (cells/mL3)AF glucose (mg/dL)AF IL-6 (ng/mL)AF CXCL10 (ng/mL)Placental histological classes
AFICILFVTOMVU
Parvovirus B-19NS+921261113.81.7+b+
Parvovirus B-19NS+93139260220.31.2+b
Cytomegalovirus++685270180.42.0
Human enterovirusa+23334101.21.9+
Coagulase negative staphylococcia+NA
Lactobacillus sakei+15291090.57. 7+b+
  1. NS=no specimen, NA=not applicable, PCR=polymerase chain reaction, GA=gestational age, AF=amniotic fluid, WBC=white blood cell, AFI=amniotic fluid infection, CIL=chronic inflammatory lesion, FVTO=fetal vascular thromboocclusive disease, MVU=maternal vascular underperfusion. aThese microorganisms were recovered from the same AF specimen, bchronic chorioamnionitis was the only chronic inflammatory lesion present – there were no chronic villitis cases.

Standard cultivation techniques identified one bacteria and one virus. Cytomegalovirus was identified by culture and confirmed with molecular microbiology. One culture of AF was positive for coagulase negative staphylococci – it is noteworthy that this patient had only four AF white blood cells per cubic milliliter, a glucose of 10 mg/dL, and a normal IL-6/CXCL10, and no evidence of acute inflammatory lesions of the placenta (acute chorioamnionitis, funisitis, or chorionic villitis). Therefore, it is possible that this represents contamination of the specimen, given that this organism is frequently present on the skin. Molecular microbiologic techniques identified two cases of parvovirus B-19, one of Lactobacillus sakei and one of human enterovirus.

None of the cases with microbial invasion of the amniotic cavity had acute inflammatory lesions of the placenta. Indeed, if pathological lesions were present, these cases had either chronic inflammatory lesions or maternal underperfusion or sometimes both. Three cases with chronic inflammatory lesions had microbial invasion: two with parvovirus B-19 and one case with Lactobacillus sakei.

Discussion

Principal findings of the study: (1) chronic placental inflammatory lesions were present in 57.9% (22/38) of cases; (2) chronic chorioamnionitis was the most frequent chronic inflammatory lesion of the placenta, followed by chronic deciduitis with plasma cells and VUE; (3) one-half of the VUE cases (3/6) also demonstrated chronic chorioamnionitis; (4) most cases of chronic chorioamnionitis had no evidence of microbial invasion of the amniotic cavity, except for three (two with parvovirus B-19, one with Lactobacillus sakei). However, these cases did not have acute inflammatory lesions, indicating the absence of either a maternal inflammatory response (acute chorioamnionitis) or a fetal inflammatory response (acute chorionic vasculitis or funisitis); (5) an elevated AF concentration of CXCL10 (above the 95th centile) was present in 60% of cases with fetal death; (6) an elevated AF CXCL10 concentration of 2.9 ng/mL had a sensitivity of 73% and a specificity of 75% in the identification of chronic placental inflammatory lesions; (7) microbial invasion of the amniotic cavity, detected by a combination of cultivation and molecular microbiologic techniques, was present in only 12.5% of fetal death cases. Importantly, most of the organisms were viruses (parvovirus B-19, Cytomegalovirus and enterovirus); (8) acute placental inflammatory lesions were infrequent in cases of fetal death; and (9) the placentas of pregnancies complicated with fetal death often had more than one lesion – the most frequent combination included maternal vascular lesions of underperfusion and chronic inflammatory lesions; 10) the odds of both an elevation in acute (IL-6) and chronic (CXCL10) inflammation markers were nine times greater when more than one class of placental lesion was diagnosed compared to a single class or when no lesion was found. Collectively, this evidence suggests that maternal anti-fetal rejection plays a role in unexplained fetal death. This is supported in patients with a fetal death by a high frequency of chronic placental inflammatory lesions, a high concentration of CXCL10, and a low frequency of microbial invasion of the amniotic cavity.

Fetal death: a manifestation of allograft failure

Fetal death is syndromic in nature and can be caused by maternal insults (i.e. maternal trauma leading to placental abruption [182], [183], a metabolic disorder such as diabetic ketoacidosis [184], [185], [186], maternal sepsis [187], [188], etc.), placental disease (i.e. observed in patients who have absent or reversed end-diastolic velocities of the umbilical artery [189], [190], massive perivillous fibrin deposition [191], [192], [193], etc.), or fetal disorders (such as cardiac arrhythmias [31], [32], hydrops fetalis [194], [195], critical congenital anomalies [33], [34] and some chromosomal abnormalities [196], [197]).

Although the fetus and placenta are known to be semi-allografts [87] and immune disorders can cause fetal death (i.e. Rh alloimmunization [198], [199]), the role of maternal anti-fetal rejection as a mechanism of disease responsible for fetal death has been considered only recently [121], [126].

Allograft rejection is a dynamic process that occurs frequently in transplanted organs [200], [201]. Infiltration of lymphocytes from the recipient into the donor organ is not uncommon [113], [114], [115], as is the deposition of complement by antibody-mediated rejection [202], [203]. However, histopathologic evidence of an immune response in the graft is not sufficient to define rejection – the latter requires organ dysfunction, which is clinically manifested by a functional disorder of the transplanted organ, such as renal failure, congestive heart failure, pulmonary insufficiency, etc. [204], [205], [206], [207].

The placenta is a multi-functional organ that has respiratory, nutritive, excretory, endocrine and immune functions, among others [208], [209], [210]. Therefore, severe placental dysfunction could manifest in a number of ways, such as inadequate transport of oxygen, leading to fetal myocardial failure and subsequent death [5]. Nutritive failure of the placenta could lead to fetal growth restriction, a risk factor of fetal death; however, this in and of itself would not be sufficient to cause death unless placental respiratory failure also occurs. Other forms of placental dysfunction may be more subtle, e.g. placental sulfatase deficiency can lead to an enzymatic inability to hydrolyze the sulfate group from DHA sulfate (DHA-S) and 16a-hydroxy-DHA sulfate. This condition is associated with low levels of estrogens and is clinically manifested with post-term pregnancy [211].

The immune functions of the placenta are complex and involve host defense against microorganisms that may gain access to the uterus via an ascending pathway or through a hematogenous route [73], [212], [213], [214], [215], [216], [217], [218], [219], [220]. Some infections are known to be a cause of fetal death (i.e. syphilis [15], [16], [17], [18], [19]). Another major immunological function is the successful establishment and maintenance of maternal-fetal tolerance [72], [75], [83], [84], [92], [94], [99], [104], [111], the breakdown of which may lead to maternal anti-fetal rejection. The clinical spectrum of maternal anti-fetal rejection may include subclinical semi-allograft rejection detected only at the time of examination of the placenta (by identification of chronic chorioamnionitis [118], [119], [120], [121], [122], [124], [126], [128], [129], VUE [122], [123], [126], [130], [131], or chronic deciduitis [122], [126], [129]), preterm labor with intact membranes [118], [119], [120], [122], [124], [125], [126], PPROM [118], maternal perivillous fibrin deposition [127], and other obstetrical syndromes [121], [122], [126]. However, the extreme form of rejection would include a functional failure of the placenta of such magnitude that it would lead to fetal death [121], [126].

The precise mechanisms whereby rejection may lead to fetal death are not yet known; however, we have described a new entity, the fetal inflammatory response syndrome type 2 (FIRS type 2), which occurs in fetuses whose placentas have lesions consistent with maternal anti-fetal rejection [122], [126]. FIRS type 2 is characterized by a unique change in the profile of cytokines in the peripheral blood, with an elevation of CXCL10, and by a stereotypic transcriptome [122]. Gene ontology analysis of differentially expressed genes in fetal white blood cells with FIRS type 2 indicates enrichment for genes that participate in immune-related processes [122]. In contrast, in FIRS type 1, the systemic inflammatory response associated with microorganisms or “danger signals” can lead to a situation that resembles septic shock in the fetus and newborn, and some reports of fetal death [221], [222], [223], [224], [225]. However, the natural history of FIRS type 2 has not been elucidated; thus, it is unclear how maternal anti-fetal rejection and FIRS type 2 can progress toward fetal death. However, the data presented herein show a clear association between fetal death, placental lesions of maternal anti-fetal rejection, and an elevation of CXCL10 in the AF, as well as the absence of microbial invasion of the amniotic cavity in most cases. Therefore, the immune disorder detected in the placenta and AF cannot be attributed to a viral or bacterial infection.

Placental pathology in fetal death

The most common lesion associated with fetal death was maternal vascular lesions of underperfusion [6], [7], [61], [144], [226], [227], [228], [229], [230], [231], [232], [233]. These lesions fall into two categories: villous changes and vascular lesions. At the most recent meeting of the Amsterdam Placental Workshop, experts recommended to re-label this category of lesions into “maternal vascular malperfusion” [234]. It is believed that an inadequate supply of nutrients and oxygen would occur in patients who have lesions such as massive perivillous fibrin deposition and massive placental infarction [235].

An important finding of this study is that chronic placental inflammatory lesions were observed in 57.9% (22/38) of cases. Chronic inflammatory lesions included chronic chorioamnionitis, VUE, and chronic deciduitis with plasma cells. Chronic placental inflammatory lesions were associated in 63.6% (14/22) of cases with lesions of maternal vascular underperfusion. The precise mechanism responsible for the association between chronic placental inflammatory lesions and maternal vascular lesions of underperfusion is unknown. It is noteworthy that in one condition, massive perivillous fibrin deposition, there is evidence of antibody-mediated maternal anti-fetal rejection (documented by a mismatch of HLA antigens between the mother and the fetus, maternal sensitization demonstrated by the presence of anti-HLA antibodies, and complement fixation of the umbilical vein shown as C4a deposit) [127]. This condition has also been found to be associated with an abnormal, severe angiogenic/anti-angiogenic profile which begins in early pregnancy [236] and has been successfully treated with pravastatin [237]. Further studies are required to determine the extent of the association between chronic placental inflammation and maternal vascular lesions of underperfusion in other obstetrical syndromes.

Acute placental inflammatory lesions were present in 28.2% of cases. Although placental pathologists have considered that acute chorioamnionitis, funisitis, and chorionic vasculitis are manifestations of an amniotic fluid infection syndrome, it is now known that these lesions can occur in the absence of bacteria or viruses in the amniotic cavity and that these processes have been attributed to danger signals that can initiate an inflammatory response in the absence of microbial products [139], [173], [174], [175], [176]. These danger signals are considered to be alarmins [238] (e.g. high-mobility group box-1 (HMGB1) [169], [239] and IL-1α [240]), which are present in the AF. It is noteworthy that only 12.5% of patients in our study had bacteria or viruses in the AF. Therefore, an infection-related etiology appears to be rare in high-income countries. In low-income countries, infection appears to play a more important role [21]. The frequency of the acute placental inflammatory lesions (28.2%) is at odds with the frequency of microbial invasion of the amniotic cavity, which was 12.5%. A possible explanation is that some patients may have developed acute inflammatory lesions and, specifically, acute chorioamnionitis during the induction of labor because of fetal death. The induction process may result in the administration of agents such as prostaglandins [241], [242], [243], [244], [245], [246], [247], [248], [249], [250], [251], [252], insertion of a Foley catheter [253], or any other method of induction of labor that may predispose to inflammation. Additionally, labor per se is an inflammatory state associated with the infiltration of inflammatory cells into the cervix [254], [255], [256], [257], [258], [259], [260], [261], [262], [263], [264], myometrium [256], [265], [266], [267], [268], [269], [270], [271] and chorioamniotic membranes [272], [273], [274], [275], [276], [277]. It should also be considered that amniocenteses were performed at the time of diagnosis and placentas were examined after delivery. While there is a temporal dissociation between the assessment of the microbial state of the amniotic cavity and placental examination, the overwhelming majority (92.5%) of patients in our study were induced and the median interval from diagnosis to delivery was short [0.1 (0–0.5) weeks]. Therefore, a smaller fraction of those with acute inflammation may not be fully explained by the process of labor induction.

The least common of the four major categories of lesions recognized by the Society of Pediatric Pathology was fetal vascular thrombo-occlusive disease – this was present in 23.1% of cases and associated with maternal vascular lesions of underperfusion in two-thirds of cases. Whether this represents simultaneous activation of the coagulation system in the mother and fetus, due to acquired or congenital thrombophilic states, remains to be determined. Some investigators have suggested that thrombophilia is a risk factor for fetal death; however, a systematic review and meta-analysis have not confirmed the initial reports [278], [279]. This may represent a lack of association or an incomplete knowledge of thrombophilic mutations at the present time.

In only 10.3% of cases did placental pathology not reveal a lesion belonging to the four major categories recognized by the Society of Pediatric Pathology. The circumstances of these fetal deaths were examined and there were no explanations for these occurrences (i.e. cord accident, maternal fetal hemorrhage, etc.).

Amniotic fluid cytokines in fetal death

The current study focuses on the examination of the concentration of two cytokines: IL-6, a marker of acute inflammatory lesions of the placenta [150], and CXCL10, a marker of chronic placental inflammatory lesions [126]. The widely used cut-off for intra-amniotic inflammation is an AF IL-6 concentration of 2.6 ng/mL [157], [168], [169]. Only one patient with microbial invasion of the amniotic cavity had an elevated AF IL-6 concentration – parvovirus B19 was detected and the IL-6 concentration was 13.7 ng/mL. This patient did not have evidence of acute chorioamnionitis but rather of chronic placental inflammatory lesions and, specifically, chronic chorioamnionitis, as well as maternal vascular lesions of underperfusion. Parvovirus B19 is typically transmitted transplacentally [280] and this may be the reason for the absence of acute chorioamnionitis.

Mixed histologic lesions were the most frequently observed placental finding, occurring in 61.5% (24/39) of placentas. The odds of a woman having a combined elevation of AF IL-6 and CXCL10 were nine times greater when mixed placental lesions were present than when no lesion or only a solitary lesion class was diagnosed. The most common classes of placental lesions found when mixed lesions were present were those of maternal vascular underperfusion and chronic inflammation. Microbial invasion of the amniotic cavity does not fully explain the elevation of IL-6 seen in unexplained fetal death. This finding raises the question of whether dysregulation of inflammatory pathways (both acute and chronic) is underway as mixed lesions are developing in the placenta. More investigation into the overlap of acute and chronic inflammation may help to answer this question.

The association between chronic placental inflammatory lesions, especially chronic chorioamnionitis, and fetal death has been previously reported by our group [121], [126]. The novel aspect of this report was the use of PCR techniques to evaluate for the presence of microorganisms in the AF. Using these advanced microbiological techniques, we determined that chronic inflammatory lesions, particularly VUE, could not be attributed to intra-amniotic infection by bacteria or viruses.

The diagnostic performance of CXCL10 in the identification of patients with chronic inflammatory lesions indicted a sensitivity of 73%, a specificity of 75%, a positive likelihood ratio of 2.9, and a negative likelihood ratio of 0.4. Further studies are required to determine whether examination of the behavior of the inflammatory/cytokine network can enhance the diagnostic performance of CXCL10. CXCL10, as well as CXCL9 and CXCL11, are T-cell chemokines, which may play a role in the rejection of the semi-allograft fetus [118], [121], [124]. Similarly, the receptor for these T-cell chemokines is CXCR3 and its soluble form is increased in the AF of women who underwent spontaneous preterm labor and whose placentas presented lesions of maternal anti-fetal rejection [281]. Thus, it is possible that a combination of chemokines and their receptors may be of help in identifying the patients with maternal anti-fetal rejection. The same can be said for the determination of these factors in maternal and fetal blood.

Strengths and limitations

A strength of this study is that we investigated placental pathology, AF cytokines, and microbial invasion of the amniotic cavity in a cohort of patients with fetal death. One of the limitations of the study is the time interval between amniocentesis and delivery, even though the majority of patients had labor induced following the diagnosis of fetal death at admission. Another limitation is the uncertainty about the relationship between the frequency of acute inflammatory lesions of the placenta, microbial invasion of the amniotic cavity, and the interval to delivery. This relationship questions whether acute inflammatory lesions may be due to the process of induction of labor and labor per se rather than a pre-existing pathologic process related to fetal death.

Conclusions

This cohort study of patients with unexplained fetal death suggests that placental lesions associated with chronic placental inflammatory lesions are second only to the prevalence of lesions consistent with maternal underperfusion; and there is a strong correlation with the AF concentration of CXCL10, a chemokine involved in allograft rejection and fetal death. The mechanisms whereby maternal anti-fetal rejection may lead to fetal death require further investigation and may be related to the deployment of FIRS type 2.


Corresponding authors: Roberto Romero, MD, D.Med.Sci., Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD and Detroit, MI, USA, Tel.: +(313) 993-2700, Fax: +(313) 993-2694; and Offer Erez, MD, Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD and Detroit, MI, USA

Author’s statement

  1. Conflict of interest: Authors state no conflict of interest.

  2. Material and methods: Informed consent: Informed consent has been obtained from all individuals included in this study.

  3. Ethical approval: The research related to human subject use has complied with all the relevant national regulations, and institutional policies, and is in accordance with the tenets of the Helsinki Declaration, and has been approved by the authors’ institutional review board or equivalent committee.

  4. Funding: This research was supported, in part, by the Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS); and, in part, with Federal funds from NICHD/NIH/DHHS under Contract No. HHSN275201300006C.

References

[1] Blencowe H, Cousens S, Jassir FB, Say L, Chou D, Mathers C, et al. National, regional, and worldwide estimates of stillbirth rates in 2015, with trends from 2000: a systematic analysis. Lancet Glob Health. 2016;4:e98–108.10.1016/S2214-109X(15)00275-2Search in Google Scholar

[2] Romero R. Prenatal medicine: the child is the father of the man. Prenat Neonat Med. 1996;1:8–11.10.1080/14767050902784171Search in Google Scholar PubMed

[3] Di Renzo GC. The great obstetrical syndromes. J Matern Fetal Neonatal Med. 2009;22:633–5.10.1080/14767050902866804Search in Google Scholar PubMed

[4] Brosens I, Pijnenborg R, Vercruysse L, Romero R. The “Great Obstetrical Syndromes” are associated with disorders of deep placentation. Am J Obstet Gynecol. 2011;204:193–201.10.1016/j.ajog.2010.08.009Search in Google Scholar PubMed PubMed Central

[5] Ananth CV, Berkowitz GS, Savitz DA, Lapinski RH. Placental abruption and adverse perinatal outcomes. J Am Med Assoc. 1999;282:1646–51.10.1001/jama.282.17.1646Search in Google Scholar PubMed

[6] Kidron D, Bernheim J, Aviram R. Placental findings contributing to fetal death, a study of 120 stillbirths between 23 and 40 weeks gestation. Placenta. 2009;30:700–4.10.1016/j.placenta.2009.05.009Search in Google Scholar PubMed

[7] Korteweg FJ, Erwich JJ, Holm JP, Ravisé JM, van der Meer J, Veeger NJ, et al. Diverse placental pathologies as the main causes of fetal death. Obstet Gynecol. 2009;114:809–17.10.1097/AOG.0b013e3181b72ebeSearch in Google Scholar PubMed

[8] Stillbirth Collaborative Research Network Writing Group. Causes of death among stillbirths. J Am Med Assoc. 2011;306:2459–68.10.1001/jama.2011.1823Search in Google Scholar PubMed PubMed Central

[9] Ananth CV, Lavery JA, Vintzileos AM, Skupski DW, Varner M, Saade G, et al. Severe placental abruption: clinical definition and associations with maternal complications. Am J Obstet Gynecol. 2016;214:272 e271–9.10.1016/j.ajog.2015.09.069Search in Google Scholar PubMed

[10] Bendon RW. Review of some causes of stillbirth. Pediatr Dev Pathol. 2001;4:517–31.10.1007/s10024001-0084-4Search in Google Scholar PubMed

[11] Hoyert DL, Gregory EC. Cause of fetal death: data from the fetal death report, 2014. Natl Vital Stat Rep. 2016;65:1–25.Search in Google Scholar

[12] Korzeniewski SJ, Romero R, Chaiworapongsa T, Chaemsaithong P, Kim CJ, Kim YM, et al. Maternal plasma angiogenic index-1 (placental growth factor/soluble vascular endothelial growth factor receptor-1) is a biomarker for the burden of placental lesions consistent with uteroplacental underperfusion: a longitudinal case-cohort study. Am J Obstet Gynecol. 2016;214:629.e1–629.e617.10.1016/j.ajog.2015.11.015Search in Google Scholar

[13] Kim YM, Chaemsaithong P, Romero R, Shaman M, Kim CJ, Kim JS, et al. Placental lesions associated with acute atherosis. J Matern Fetal Neonatal Med. 2015;28:1554–62.10.3109/14767058.2014.960835Search in Google Scholar

[14] Ptacek I, Sebire NJ, Man JA, Brownbill P, Heazell AE. Systematic review of placental pathology reported in association with stillbirth. Placenta. 2014;35:552–62.10.1016/j.placenta.2014.05.011Search in Google Scholar

[15] Sheffield JS, Sanchez PJ, Wendel GD, Jr., Fong DW, Margraf LR, Zeray F, et al. Placental histopathology of congenital syphilis. Obstet Gynecol. 2002;100:126–33.10.1097/00006250-200207000-00020Search in Google Scholar

[16] Yakoob MY, Lawn JE, Darmstadt GL, Bhutta ZA. Stillbirths: epidemiology, evidence, and priorities for action. Semin Perinatol. 2010;34:387–94.10.1053/j.semperi.2010.09.010Search in Google Scholar

[17] Hawkes S, Matin N, Broutet N, Low N. Effectiveness of interventions to improve screening for syphilis in pregnancy: a systematic review and meta-analysis. Lancet Infect Dis. 2011;11:684–91.10.1016/S1473-3099(11)70104-9Search in Google Scholar

[18] Qin J, Yang T, Xiao S, Tan H, Feng T, Fu H. Reported estimates of adverse pregnancy outcomes among women with and without syphilis: a systematic review and meta-analysis. PLoS One. 2014;9:e102203.10.1371/journal.pone.0102203Search in Google Scholar

[19] Rac MW, Revell PA, Eppes CS. Syphilis during pregnancy: a preventable threat to maternal-fetal health. Am J Obstet Gynecol. 2017;216:352–63.10.1016/j.ajog.2016.11.1052Search in Google Scholar

[20] Nosten F, McGready R, Simpson JA, Thwai KL, Balkan S, Cho T, et al. Effects of Plasmodium vivax malaria in pregnancy. Lancet. 1999;354:546–9.10.1016/S0140-6736(98)09247-2Search in Google Scholar

[21] Goldenberg RL, McClure EM, Saleem S, Reddy UM. Infection-related stillbirths. Lancet. 2010;375:1482–90.10.1016/S0140-6736(09)61712-8Search in Google Scholar

[22] Bhutta ZA, Yakoob MY, Lawn JE, Rizvi A, Friberg IK, Weissman E, et al. Stillbirths: what difference can we make and at what cost? Lancet. 2011;377:1523–38.10.1016/S0140-6736(10)62269-6Search in Google Scholar

[23] Sarno M, Sacramento GA, Khouri R, do Rosário MS, Costa F, Archanjo G, et al. Zika Virus infection and stillbirths: a case of hydrops fetalis, hydranencephaly and fetal demise. PLoS Negl Trop Dis. 2016;10:e0004517.10.1371/journal.pntd.0004517Search in Google Scholar

[24] Brasil P, Pereira JP, Jr., Moreira ME, Ribeiro Nogueira RM, Damasceno L, Wakimoto M, et al. Zika virus infection in pregnant women in Rio de Janeiro. N Engl J Med. 2016;375:2321–34.10.1056/NEJMoa1602412Search in Google Scholar

[25] Citil Dogan A, Wayne S, Bauer S, Ogunyemi D, Kulkharni SK, Maulik D, et al. The Zika virus and pregnancy: evidence, management, and prevention. J Matern Fetal Neonatal Med. 2017;30:386–96.10.3109/14767058.2016.1174210Search in Google Scholar

[26] Goldenberg RL, Thompson C. The infectious origins of stillbirth. Am J Obstet Gynecol. 2003;189:861–73.10.1067/S0002-9378(03)00470-8Search in Google Scholar

[27] Lamont RF, Sobel J, Mazaki-Tovi S, Kusanovic JP, Vaisbuch E, Kim SK, et al. Listeriosis in human pregnancy: a systematic review. J Perinat Med. 2011;39:227–36.10.1515/jpm.2011.035Search in Google Scholar PubMed PubMed Central

[28] Committee on Obstetric Practice American College of Obstetricians Gynecologists. Committee Opinion No. 614: Management of pregnant women with presumptive exposure to Listeria monocytogenes. Obstet Gynecol. 2014;124:1241–4.10.1097/01.AOG.0000457501.73326.6cSearch in Google Scholar PubMed

[29] McLauchlin J. Human listeriosis in Britain, 1967–85, a summary of 722 cases. 1. Listeriosis during pregnancy and in the newborn. Epidemiol Infect. 1990;104:181–9.10.1017/S0950268800059343Search in Google Scholar PubMed PubMed Central

[30] Plaza MC, Gilbert-Barness E. Fetal death in utero secondary to Listeria monocytogenes placental infection. Pediatr Pathol Mol Med. 2001;20:433–7.10.3109/15513810109168624Search in Google Scholar

[31] Crotti L, Tester DJ, White WM, Bartos DC, Insolia R, Besana A, et al. Long QT syndrome-associated mutations in intrauterine fetal death. J Am Med Assoc. 2013;309:1473–82.10.1001/jama.2013.3219Search in Google Scholar PubMed PubMed Central

[32] Pusiol T, Roncati L, Lavezzi AM, Taddei F, Piscioli F, Ottaviani G. Sudden fetal death due to dualism of the sino-atrial node. Cardiovasc Pathol. 2016;25:325–8.10.1016/j.carpath.2016.04.001Search in Google Scholar PubMed

[33] Boyle B, McConkey R, Garne E, Loane M, Addor MC, Bakker MK, et al. Trends in the prevalence, risk and pregnancy outcome of multiple births with congenital anomaly: a registry-based study in 14 European countries 1984–2007. Br J Obstet Gynaecol. 2013;120:707–16.10.1111/1471-0528.12146Search in Google Scholar PubMed

[34] Frey HA, Odibo AO, Dicke JM, Shanks AL, Macones GA, Cahill AG. Stillbirth risk among fetuses with ultrasound-detected isolated congenital anomalies. Obstet Gynecol. 2014;124:91–8.10.1097/AOG.0000000000000335Search in Google Scholar PubMed PubMed Central

[35] Getahun D, Ananth CV, Kinzler WL. Risk factors for antepartum and intrapartum stillbirth: a population-based study. Am J Obstet Gynecol. 2007;196:499–507.10.1016/j.ajog.2006.09.017Search in Google Scholar PubMed

[36] Vergani P, Cozzolino S, Pozzi E, Cuttin MS, Greco M, Ornaghi S, et al. Identifying the causes of stillbirth: a comparison of four classification systems. Am J Obstet Gynecol. 2008;199:319.e1–4.10.1016/j.ajog.2008.06.098Search in Google Scholar PubMed

[37] Collins JH. Umbilical cord accidents: human studies. Semin Perinatol. 2002;26:79–82.10.1053/sper.2002.29860Search in Google Scholar PubMed

[38] Sherer DM, Dalloul M, Ward K, Nakagawa J, Joseph I, Grube S. Coexisting true umbilical cord knot and nuchal cord: possible cumulative increased risk for adverse perinatal outcome. Ultrasound Obstet Gynecol. 2016.10.1002/uog.17389Search in Google Scholar PubMed

[39] Silver RM, Varner MW, Reddy U, Goldenberg R, Pinar H, Conway D, et al. Work-up of stillbirth: a review of the evidence. Am J Obstet Gynecol. 2007;196:433–44.10.1016/j.ajog.2006.11.041Search in Google Scholar PubMed PubMed Central

[40] Victoria A, Mora G, Arias F. Perinatal outcome, placental pathology, and severity of discordance in monochorionic and dichorionic twins. Obstet Gynecol. 2001;97:310–5.10.1097/00006250-200102000-00027Search in Google Scholar

[41] Kaufman HK, Hume RF, Jr., Calhoun BC, Carlson N, Yorke V, Elliott D, et al. Natural history of twin gestation complicated by in utero fetal demise: associations of chorionicity, prematurity, and maternal morbidity. Fetal Diagn Ther. 2003;18:442–6.10.1159/000073140Search in Google Scholar PubMed

[42] Lewi L, Gucciardo L, Van Mieghem T, de Koninck P, Beck V, Medek H, et al. Monochorionic diamniotic twin pregnancies: natural history and risk stratification. Fetal Diagn Ther. 2010;27:121–33.10.1159/000313300Search in Google Scholar PubMed

[43] Fretts R. Stillbirth epidemiology, risk factors, and opportunities for stillbirth prevention. Clin Obstet Gynecol. 2010;53:588–96.10.1097/GRF.0b013e3181eb63fcSearch in Google Scholar PubMed

[44] Smith GC. Screening and prevention of stillbirth. Best Pract Res Clin Obstet Gynaecol. 2017;38:71–82.10.1016/j.bpobgyn.2016.08.002Search in Google Scholar

[45] Reddy UM. Prediction and prevention of recurrent stillbirth. Obstet Gynecol. 2007;110:1151–64.10.3109/9781841847191.005Search in Google Scholar

[46] de Galan-Roosen AE, Kuijpers JC, van der Straaten PJ, Merkus JM. Fundamental classification of perinatal death. Validation of a new classification system of perinatal death. Eur J Obstet Gynecol Reprod Biol. 2002;103:30–6.10.1016/S0301-2115(02)00023-4Search in Google Scholar

[47] Gardosi J, Kady SM, McGeown P, Francis A, Tonks A. Classification of stillbirth by relevant condition at death (ReCoDe): population based cohort study. Br Med J. 2005;331:1113–7.10.1136/bmj.38629.587639.7CSearch in Google Scholar PubMed PubMed Central

[48] Korteweg FJ, Gordijn SJ, Timmer A, Erwich JJ, Bergman KA, Bouman K, et al. The Tulip classification of perinatal mortality: introduction and multidisciplinary inter-rater agreement. Br J Obstet Gynaecol. 2006;113:393–401.10.1111/j.1471-0528.2006.00881.xSearch in Google Scholar PubMed

[49] Flenady V, Froen JF, Pinar H, Torabi R, Saastad E, Guyon G, et al. An evaluation of classification systems for stillbirth. BMC Pregnancy Childbirth. 2009;9:24.10.1186/1471-2393-9-24Search in Google Scholar PubMed PubMed Central

[50] Gordijn SJ, Korteweg FJ, Erwich JJ, Holm JP, van Diem MT, Bergman KA, et al. A multilayered approach for the analysis of perinatal mortality using different classification systems. Eur J Obstet Gynecol Reprod Biol. 2009;144:99–104.10.1016/j.ejogrb.2009.01.012Search in Google Scholar PubMed

[51] Reddy UM, Goldenberg R, Silver R, Smith GC, Pauli RM, Wapner RJ, et al. Stillbirth classification–developing an international consensus for research: executive summary of a National Institute of Child Health and Human Development workshop. Obstet Gynecol. 2009;114:901–14.10.1097/AOG.0b013e3181b8f6e4Search in Google Scholar PubMed PubMed Central

[52] Boyd TK, Wright CA, Odendaal H, Elliott AJ, Sens MA, Dunn Folkerth R, et al. The stillbirth classification system for the safe passage study: incorporating mechanism, etiology, and recurrence. Pediatr Dev Pathol. 2016. [Epub ahead of print].10.1177/1093526616686251Search in Google Scholar PubMed

[53] Hey EN, Lloyd DJ, Wigglesworth JS. Classifying perinatal death: fetal and neonatal factors. Br J Obstet Gynaecol. 1986;93:1213–23.10.1111/j.1471-0528.1986.tb07854.xSearch in Google Scholar PubMed

[54] Wigglesworth JS. Monitoring perinatal mortality. A pathophysiological approach. Lancet. 1980;2:684–6.10.1016/S0140-6736(80)92717-8Search in Google Scholar

[55] Rayburn W, Sander C, Barr M, Jr., Rygiel R. The stillborn fetus: placental histologic examination in determining a cause. Obstet Gynecol. 1985;65:637–41.Search in Google Scholar

[56] VanderWielen B, Zaleski C, Cold C, McPherson E. Wisconsin stillbirth services program: a multifocal approach to stillbirth analysis. Am J Med Genet A. 2011;155A:1073–80.10.1002/ajmg.a.34016Search in Google Scholar

[57] Leisher SH, Teoh Z, Reinebrant H, Allanson E, Blencowe H, Erwich JJ, et al. Seeking order amidst chaos: a systematic review of classification systems for causes of stillbirth and neonatal death, 2009–2014. BMC Pregnancy Childbirth. 2016;16:295.10.1186/s12884-016-1071-0Search in Google Scholar

[58] Horn LC, Langner A, Stiehl P, Wittekind C, Faber R. Identification of the causes of intrauterine death during 310 consecutive autopsies. Eur J Obstet Gynecol Reprod Biol. 2004;113:134–8.10.1016/S0301-2115(03)00371-3Search in Google Scholar

[59] Dudley DJ, Goldenberg R, Conway D, Silver RM, Saade GR, Varner MW, et al. A new system for determining the causes of stillbirth. Obstet Gynecol. 2010;116(2 Pt 1):254–60.10.1097/AOG.0b013e3181e7d975Search in Google Scholar PubMed PubMed Central

[60] Baird D, Walker J, Thomson AM. The causes and prevention of stillbirths and first week deaths. III. A classification of deaths by clinical cause; the effect of age, parity and length of gestation on death rates by cause. J Obstet Gynaecol Br Emp. 1954;61:433–48.10.1111/j.1471-0528.1954.tb07507.xSearch in Google Scholar PubMed

[61] Hovatta O, Lipasti A, Rapola J, Karjalainen O. Causes of stillbirth: a clinicopathological study of 243 patients. Br J Obstet Gynaecol. 1983;90:691–6.10.1111/j.1471-0528.1983.tb09296.xSearch in Google Scholar PubMed

[62] Cole SK, Hey EN, Thomson AM. Classifying perinatal death: an obstetric approach. Br J Obstet Gynaecol. 1986;93:1204–12.10.1111/j.1471-0528.1986.tb07853.xSearch in Google Scholar PubMed

[63] Chan A, King JF, Flenady V, Haslam RH, Tudehope DI. Classification of perinatal deaths: development of the Australian and New Zealand classifications. J Paediatr Child Health. 2004;40:340–7.10.1111/j.1440-1754.2004.00398.xSearch in Google Scholar PubMed

[64] Nappi L, Trezza F, Bufo P, Riezzo I, Turillazzi E, Borghi C, et al. Classification of stillbirths is an ongoing dilemma. J Perinat Med. 2016;44:837–43.10.1515/jpm-2015-0318Search in Google Scholar PubMed

[65] Laury A, Sanchez-Lara PA, Pepkowitz S, Graham JM, Jr. A study of 534 fetal pathology cases from prenatal diagnosis referrals analyzed from 1989 through 2000. Am J Med Genet A. 2007;143A:3107–20.10.1002/ajmg.a.32094Search in Google Scholar

[66] Korteweg FJ, Erwich JJ, Timmer A, van der Meer J, Ravisé JM, Veeger NJ, et al. Evaluation of 1025 fetal deaths: proposed diagnostic workup. Am J Obstet Gynecol. 2012;206: 53.e1–53.e12.10.1016/j.ajog.2011.10.026Search in Google Scholar

[67] Kasai M, Aoki S, Ogawa M, Kurasawa K, Takahashi T, Hirahara F. Prediction of perinatal outcomes based on primary symptoms in women with placental abruption. J Obstet Gynaecol Res. 2015;41:850–6.10.1111/jog.12637Search in Google Scholar

[68] Nkwabong E, Tiomela Goula G. Placenta abruption surface and perinatal outcome. J Matern Fetal Neonatal Med. 2017;30:1456–9.10.1080/14767058.2016.1219988Search in Google Scholar

[69] Kvarnstrand L, Milsom I, Lekander T, Druid H, Jacobsson B. Maternal fatalities, fetal and neonatal deaths related to motor vehicle crashes during pregnancy: a national population-based study. Acta Obstet Gynecol Scand. 2008;87:946–52.10.1080/00016340802302184Search in Google Scholar

[70] Klinich KD, Flannagan CA, Rupp JD, Sochor M, Schneider LW, Pearlman MD. Fetal outcome in motor-vehicle crashes: effects of crash characteristics and maternal restraint. Am J Obstet Gynecol. 2008;198:450 e451–9.10.1016/j.ajog.2008.02.009Search in Google Scholar

[71] McClure EM, Garces A, Saleem S, Moore JL, Bose CL, Esamai F, et al. Global Network for Women’s and Children’s Health Research: probable causes of stillbirth in low- and middle-income countries using a prospectively defined classification system. Br J Obstet Gynaecol. 2017. [Epub ahead of print].10.1111/1471-0528.14493Search in Google Scholar

[72] Erlebacher A. Why isn’t the fetus rejected? Curr Opin Immunol. 2001;13:590–3.10.1016/S0952-7915(00)00264-8Search in Google Scholar

[73] Mor G, Cardenas I. The immune system in pregnancy: a unique complexity. Am J Reprod Immunol. 2010;63:425–33.10.1111/j.1600-0897.2010.00836.xSearch in Google Scholar PubMed PubMed Central

[74] Chaouat G, Petitbarat M, Dubanchet S, Rahmati M, Ledee N. Tolerance to the foetal allograft? Am J Reprod Immunol. 2010;63:624–36.10.1111/j.1600-0897.2010.00832.xSearch in Google Scholar PubMed

[75] PrabhuDas M, Bonney E, Caron K, Dey S, Erlebacher A, Fazleabas A, et al. Immune mechanisms at the maternal-fetal interface: perspectives and challenges. Nat Immunol. 2015;16:328–34.10.1038/ni.3131Search in Google Scholar PubMed PubMed Central

[76] Trowsdale J, Betz AG. Mother’s little helpers: mechanisms of maternal-fetal tolerance. Nat Immunol. 2006;7:241–6.10.1038/ni1317Search in Google Scholar PubMed

[77] Munoz-Suano A, Hamilton AB, Betz AG. Gimme shelter: the immune system during pregnancy. Immunol Rev. 2011;241:20–38.10.1111/j.1600-065X.2011.01002.xSearch in Google Scholar PubMed

[78] Samstein RM, Josefowicz SZ, Arvey A, Treuting PM, Rudensky AY. Extrathymic generation of regulatory T cells in placental mammals mitigates maternal-fetal conflict. Cell. 2012;150:29–38.10.1016/j.cell.2012.05.031Search in Google Scholar PubMed PubMed Central

[79] Leslie M. Immunology. Fetal immune system hushes attacks on maternal cells. Science. 2008;322:1450–1.10.1126/science.322.5907.1450bSearch in Google Scholar PubMed

[80] Mold JE, Michaelsson J, Burt TD, Muench MO, Beckerman KP, Busch MP, et al. Maternal alloantigens promote the development of tolerogenic fetal regulatory T cells in utero. Science. 2008;322:1562–5.10.1126/science.1164511Search in Google Scholar PubMed PubMed Central

[81] Burlingham WJ. A lesson in tolerance–maternal instruction to fetal cells. N Engl J Med. 2009;360:1355–7.10.1056/NEJMcibr0810752Search in Google Scholar PubMed PubMed Central

[82] Bluestone JA. Mechanisms of tolerance. Immunol Rev. 2011;241:5–19.10.1111/j.1600-065X.2011.01019.xSearch in Google Scholar PubMed

[83] Betz AG. Immunology: Tolerating pregnancy. Nature. 2012;490:47–8.10.1038/490047aSearch in Google Scholar PubMed

[84] Rowe JH, Ertelt JM, Xin L, Way SS. Pregnancy imprints regulatory memory that sustains anergy to fetal antigen. Nature. 2012;490:102–6.10.1038/nature11462Search in Google Scholar PubMed PubMed Central

[85] Williams Z. Inducing tolerance to pregnancy. N Engl J Med. 2012;367:1159–61.10.1056/NEJMcibr1207279Search in Google Scholar

[86] Erlebacher A. Mechanisms of T cell tolerance towards the allogeneic fetus. Nat Rev Immunol. 2013;13:23–33.10.1038/nri3361Search in Google Scholar

[87] Medawar PB. Some immunological and endocrinological problems raised by the evolution of viviparity in vertebrates. Soc Exp Biol. 1953:320–38.Search in Google Scholar

[88] Billington WD. The immunological problem of pregnancy: 50 years with the hope of progress. A tribute to Peter Medawar. J Reprod Immunol. 2003;60:1–11.10.1016/S0165-0378(03)00083-4Search in Google Scholar

[89] Munn DH, Zhou M, Attwood JT, Bondarev I, Conway SJ, Marshall B, et al. Prevention of allogeneic fetal rejection by tryptophan catabolism. Science. 1998;281:1191–3.10.1126/science.281.5380.1191Search in Google Scholar

[90] Bonney EA, Matzinger P. Much IDO about pregnancy. Nat Med. 1998;4:1128–9.10.1038/2624Search in Google Scholar

[91] Bonney EA. Maternal tolerance is not critically dependent on interleukin-4. Immunology. 2001;103:382–9.10.1046/j.1365-2567.2001.01239.xSearch in Google Scholar

[92] Szekeres-Bartho J. Immunological relationship between the mother and the fetus. Int Rev Immunol. 2002;21:471–95.10.1080/08830180215017Search in Google Scholar

[93] Bonney EA, Onyekwuluje J. Maternal tolerance to H-Y is independent of IL-10. Immunol Invest. 2004;33:385–95.10.1081/IMM-200032732Search in Google Scholar

[94] Aluvihare VR, Kallikourdis M, Betz AG. Regulatory T cells mediate maternal tolerance to the fetus. Nat Immunol. 2004;5:266–71.10.1038/ni1037Search in Google Scholar

[95] Moffett A, Loke YW. The immunological paradox of pregnancy: a reappraisal. Placenta. 2004;25:1–8.10.1016/S0143-4004(03)00167-XSearch in Google Scholar

[96] Piccinni MP. T cell tolerance towards the fetal allograft. J Reprod Immunol. 2010;85:71–5.10.1016/j.jri.2010.01.006Search in Google Scholar PubMed

[97] Nancy P, Tagliani E, Tay CS, Asp P, Levy DE, Erlebacher A. Chemokine gene silencing in decidual stromal cells limits T cell access to the maternal-fetal interface. Science. 2012;336:1317–21.10.1126/science.1220030Search in Google Scholar PubMed PubMed Central

[98] Shepard MT, Bonney EA. PD-1 regulates T cell proliferation in a tissue and subset-specific manner during normal mouse pregnancy. Immunol Invest. 2013;42:385–408.10.3109/08820139.2013.782317Search in Google Scholar PubMed PubMed Central

[99] Hemberger M. Immune balance at the foeto-maternal interface as the fulcrum of reproductive success. J Reprod Immunol. 2013;97:36–42.10.1016/j.jri.2012.10.006Search in Google Scholar PubMed

[100] Xin L, Ertelt JM, Rowe JH, Jiang TT, Kinder JM, Chaturvedi V, et al. Cutting edge: committed Th1 CD4+ T cell differentiation blocks pregnancy-induced Foxp3 expression with antigen-specific fetal loss. J Immunol. 2014;192:2970–4.10.4049/jimmunol.1302678Search in Google Scholar PubMed PubMed Central

[101] Racicot K, Kwon JY, Aldo P, Silasi M, Mor G. Understanding the complexity of the immune system during pregnancy. Am J Reprod Immunol. 2014;72:107–16.10.1111/aji.12289Search in Google Scholar PubMed PubMed Central

[102] Kinder JM, Jiang TT, Ertelt JM, Xin L, Strong BS, Shaaban AF, et al. Cross-generational reproductive fitness enforced by microchimeric maternal cells. Cell. 2015;162:505–15.10.1016/j.cell.2015.07.006Search in Google Scholar PubMed PubMed Central

[103] Chaturvedi V, Ertelt JM, Jiang TT, Kinder JM, Xin L, Owens KJ, et al. CXCR3 blockade protects against Listeria monocytogenes infection-induced fetal wastage. J Clin Invest. 2015;125:1713–25.10.1172/JCI78578Search in Google Scholar PubMed PubMed Central

[104] Bonney EA. Immune regulation in pregnancy: a matter of perspective? Obstet Gynecol Clin North Am. 2016;43:679–98.10.1016/j.ogc.2016.07.004Search in Google Scholar PubMed PubMed Central

[105] Bonney EA, Onyekwuluje J. The H-Y response in mid-gestation and long after delivery in mice primed before pregnancy. Immunol Invest. 2003;32:71–81.10.1081/IMM-120019209Search in Google Scholar

[106] Norton MT, Fortner KA, Oppenheimer KH, Bonney EA. Evidence that CD8 T-cell homeostasis and function remain intact during murine pregnancy. Immunology. 2010;131:426–37.10.1111/j.1365-2567.2010.03316.xSearch in Google Scholar PubMed PubMed Central

[107] Rosenblum MD, Way SS, Abbas AK. Regulatory T cell memory. Nat Rev Immunol. 2016;16:90–101.10.1038/nri.2015.1Search in Google Scholar PubMed PubMed Central

[108] Jiang TT, Chaturvedi V, Ertelt JM, Kinder JM, Clark DR, Valent AM, et al. Regulatory T cells: new keys for further unlocking the enigma of fetal tolerance and pregnancy complications. J Immunol. 2014;192:4949–56.10.4049/jimmunol.1400498Search in Google Scholar PubMed PubMed Central

[109] Erlebacher A. Immune surveillance of the maternal/fetal interface: controversies and implications. Trends Endocrinol Metab. 2010;21:428–34.10.1016/j.tem.2010.02.003Search in Google Scholar PubMed PubMed Central

[110] Tagliani E, Erlebacher A. Dendritic cell function at the maternal-fetal interface. Expert Rev Clin Immunol. 2011;7:593–602.10.1586/eci.11.52Search in Google Scholar PubMed PubMed Central

[111] Erlebacher A. Immunology of the maternal-fetal interface. Annu Rev Immunol. 2013;31:387–411.10.1146/annurev-immunol-032712-100003Search in Google Scholar PubMed

[112] Nancy P, Erlebacher A. T cell behavior at the maternal-fetal interface. Int J Dev Biol. 2014;58:189–98.10.1387/ijdb.140054aeSearch in Google Scholar PubMed PubMed Central

[113] Strom TB, Tilney NL, Paradysz JM, Bancewicz J, Carpenter CB. Cellular components of allograft rejection: identity, specificity, and cytotoxic function of cells infiltrating acutely rejecting allografts. J Immunol. 1977;118:2020–6.10.4049/jimmunol.118.6.2020Search in Google Scholar

[114] Chan SY, DeBruyne LA, Goodman RE, Eichwald EJ, Bishop DK. In vivo depletion of CD8+ T cells results in Th2 cytokine production and alternate mechanisms of allograft rejection. Transplantation. 1995;59:1155–61.10.1097/00007890-199504270-00014Search in Google Scholar

[115] Valujskikh A, Lantz O, Celli S, Matzinger P, Heeger PS. Cross-primed CD8(+) T cells mediate graft rejection via a distinct effector pathway. Nat Immunol. 2002;3:844–51.10.1038/ni831Search in Google Scholar PubMed

[116] Nankivell BJ, Borrows RJ, Fung CL, O’Connell PJ, Allen RD, Chapman JR. The natural history of chronic allograft nephropathy. N Engl J Med. 2003;349:2326–33.10.1056/NEJMoa020009Search in Google Scholar PubMed

[117] Nankivell BJ, Alexander SI. Rejection of the kidney allograft. N Engl J Med. 2010;363:1451–62.10.1056/NEJMra0902927Search in Google Scholar PubMed

[118] Kim CJ, Romero R, Kusanovic JP, Yoo W, Dong Z, Topping V, et al. The frequency, clinical significance, and pathological features of chronic chorioamnionitis: a lesion associated with spontaneous preterm birth. Mod Pathol. 2010;23:1000–11.10.1038/modpathol.2010.73Search in Google Scholar PubMed PubMed Central

[119] Lee J, Romero R, Xu Y, Kim JS, Topping V, Yoo W, et al. A signature of maternal anti-fetal rejection in spontaneous preterm birth: chronic chorioamnionitis, anti-human leukocyte antigen antibodies, and C4d. PLoS One. 2011;6:e16806.10.1371/journal.pone.0016806Search in Google Scholar PubMed PubMed Central

[120] Lee J, Romero R, Xu Y, Kim JS, Park JY, Kusanovic JP, et al. Maternal HLA panel-reactive antibodies in early gestation positively correlate with chronic chorioamnionitis: evidence in support of the chronic nature of maternal anti-fetal rejection. Am J Reprod Immunol. 2011;66:510–26.10.1111/j.1600-0897.2011.01066.xSearch in Google Scholar PubMed PubMed Central

[121] Lee J, Romero R, Dong Z, Xu Y, Qureshi F, Jacques S, et al. Unexplained fetal death has a biological signature of maternal anti-fetal rejection: chronic chorioamnionitis and alloimmune anti-human leucocyte antigen antibodies. Histopathology. 2011;59:928–38.10.1111/j.1365-2559.2011.04038.xSearch in Google Scholar PubMed PubMed Central

[122] Lee J, Romero R, Chaiworapongsa T, Dong Z, Tarca AL, Xu Y, et al. Characterization of the fetal blood transcriptome and proteome in maternal anti-fetal rejection: evidence of a distinct and novel type of human fetal systemic inflammatory response. Am J Reprod Immunol. 2013;70:265–84.10.1111/aji.12142Search in Google Scholar PubMed PubMed Central

[123] Kim MJ, Romero R, Kim CJ, Tarca AL, Chhauy S, LaJeunesse C, et al. Villitis of unknown etiology is associated with a distinct pattern of chemokine up-regulation in the feto-maternal and placental compartments: implications for conjoint maternal allograft rejection and maternal anti-fetal graft-versus-host disease. J Immunol. 2009;182:3919–27.10.4049/jimmunol.0803834Search in Google Scholar PubMed PubMed Central

[124] Ogge G, Romero R, Lee DC, Gotsch F, Than NG, Lee J, et al. Chronic chorioamnionitis displays distinct alterations of the amniotic fluid proteome. J Pathol. 2011;223:553–65.10.1002/path.2825Search in Google Scholar PubMed PubMed Central

[125] Lee J, Romero R, Xu Y, Miranda J, Yoo W, Chaemsaithong P, et al. Detection of anti-HLA antibodies in maternal blood in the second trimester to identify patients at risk of antibody-mediated maternal anti-fetal rejection and spontaneous preterm delivery. Am J Reprod Immunol. 2013;70:162–75.10.1111/aji.12141Search in Google Scholar PubMed PubMed Central

[126] Kim CJ, Romero R, Chaemsaithong P, Kim JS. Chronic inflammation of the placenta: definition, classification, pathogenesis, and clinical significance. Am J Obstet Gynecol. 2015;213(4 Suppl):S53–69.10.1016/j.ajog.2015.08.041Search in Google Scholar PubMed PubMed Central

[127] Romero R, Whitten A, Korzeniewski SJ, Than NG, Chaemsaithong P, Miranda J, et al. Maternal floor infarction/massive perivillous fibrin deposition: a manifestation of maternal antifetal rejection? Am J Reprod Immunol. 2013;70:285–98.10.1111/aji.12143Search in Google Scholar

[128] Lee J, Kim JS, Park JW, Park CW, Park JS, Jun JK, et al. Chronic chorioamnionitis is the most common placental lesion in late preterm birth. Placenta. 2013;34:681–9.10.1016/j.placenta.2013.04.014Search in Google Scholar

[129] Bang H, Bae GE, Park HY, Kim YM, Choi SJ, Oh SY, et al. Chronic Placental Inflammation in Twin Pregnancies. J Pathol Transl Med. 2015;49:489–96.10.4132/jptm.2015.09.09Search in Google Scholar

[130] Redline RW, Patterson P. Villitis of unknown etiology is associated with major infiltration of fetal tissue by maternal inflammatory cells. Am J Pathol. 1993;143:473–9.Search in Google Scholar

[131] Kim JS, Romero R, Kim MR, Kim YM, Friel L, Espinoza J, et al. Involvement of Hofbauer cells and maternal T cells in villitis of unknown aetiology. Histopathology. 2008;52:457–64.10.1111/j.1365-2559.2008.02964.xSearch in Google Scholar

[132] Jacques SM, Qureshi F. Chronic chorioamnionitis: a clinicopathologic and immunohistochemical study. Hum Pathol. 1998;29:1457–61.10.1016/S0046-8177(98)90016-8Search in Google Scholar

[133] Khong TY, Bendon RW, Qureshi F, Redline RW, Gould S, Stallmach T, et al. Chronic deciduitis in the placental basal plate: definition and interobserver reliability. Hum Pathol. 2000;31:292–5.10.1016/S0046-8177(00)80241-5Search in Google Scholar

[134] Myerson D, Parkin RK, Benirschke K, Tschetter CN, Hyde SR. The pathogenesis of villitis of unknown etiology: analysis with a new conjoint immunohistochemistry-in situ hybridization procedure to identify specific maternal and fetal cells. Pediatr Dev Pathol. 2006;9:257–65.10.2350/08-05-0103.1Search in Google Scholar

[135] Duquesnoy RJ, Demetris AJ. Immunopathology of cardiac transplant rejection. Curr Opin Cardiol. 1995;10:193–206.10.1097/00001573-199503000-00014Search in Google Scholar

[136] 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

[137] ACOG practice bulletin. Diagnosis and management of preeclampsia and eclampsia. Number 33, January 2002. Obstet Gynecol. 2002;99:159–67.10.1097/00006250-200201000-00028Search in Google Scholar

[138] Smulyan H, Safar ME. Blood pressure measurement: retrospective and prospective views. Am J Hypertens. 2011;24:628–34.10.1038/ajh.2011.22Search in Google Scholar PubMed

[139] 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

[140] Redline RW, Faye-Petersen O, Heller D, Qureshi F, Savell V, Vogler C, et al. Amniotic infection syndrome: nosology and reproducibility of placental reaction patterns. Pediatr Dev Pathol. 2003;6:435–48.10.1007/s10024-003-7070-ySearch in Google Scholar PubMed

[141] Redline RW, Ariel I, Baergen RN, Desa DJ, Kraus FT, Roberts DJ, et al. Fetal vascular obstructive lesions: nosology and reproducibility of placental reaction patterns. Pediatr Dev Pathol. 2004;7:443–52.10.1007/s10024-004-2020-xSearch in Google Scholar PubMed

[142] Redline RW, Boyd T, Campbell V, Hyde S, Kaplan C, Khong TY, et al. Maternal vascular underperfusion: nosology and reproducibility of placental reaction patterns. Pediatr Dev Pathol. 2004;7:237–49.10.1007/s10024-003-8083-2Search in Google Scholar PubMed

[143] 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

[144] Redline RW. Classification of placental lesions. Am J Obstet Gynecol. 2015;213(4 Suppl):S21–8.10.1016/j.ajog.2015.05.056Search in Google Scholar PubMed

[145] Redline RW. Villitis of unknown etiology: noninfectious chronic villitis in the placenta. Hum Pathol. 2007;38:1439–46.10.1016/j.humpath.2007.05.025Search in Google Scholar PubMed

[146] 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

[147] 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 PubMed

[148] Salafia CM, Sherer DM, Spong CY, Lencki S, Eglinton GS, Parkash V, et al. Fetal but not maternal serum cytokine levels correlate with histologic acute placental inflammation. Am J Perinatol. 1997;14:419–22.10.1055/s-2007-994172Search in Google Scholar PubMed

[149] 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

[150] Kim CJ, Romero R, Chaemsaithong P, Chaiyasit N, Yoon BH, Kim YM. Acute chorioamnionitis and funisitis: definition, pathologic features, and clinical significance. Am J Obstet Gynecol. 2015;213(4 Suppl):S29–52.10.1016/j.ajog.2015.08.040Search 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, 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

[153] 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

[154] Gomez R, Romero R, Galasso M, Behnke E, Insunza A, Cotton DB. The value of amniotic fluid interleukin-6, white blood cell count, and gram stain in the diagnosis of microbial invasion `of the amniotic cavity in patients at term. Am J Reprod Immunol. 1994;32:200–10.10.1111/j.1600-0897.1994.tb01115.xSearch in Google Scholar

[155] Halgunset J, Johnsen H, Kjollesdal AM, Qvigstad E, Espevik T, Austgulen R. Cytokine levels in amniotic fluid and inflammatory changes in the placenta from normal deliveries at term. Eur J Obstet Gynecol Reprod Biol. 1994;56:153–60.10.1016/0028-2243(94)90162-7Search in Google Scholar

[156] 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

[157] 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

[158] Hsu CD, Meaddough E, Hong SF, Aversa K, Lu LC, Copel JA. Elevated amniotic fluid nitric oxide metabolites and interleukin-6 in intra-amniotic infection. J Soc Gynecol Investig. 1998;5:21–4.10.1016/S1071-5576(97)00099-3Search in Google Scholar

[159] 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

[160] Bashiri A, Horowitz S, Huleihel M, Hackmon R, Dukler D, Mazor M. Elevated concentrations of interleukin-6 in intra-amniotic infection with Ureaplasma urealyticum in asymptomatic women during genetic amniocentesis. Acta Obstet Gynecol Scand. 1999;78:379–82.10.1080/j.1600-0412.1999.780506.xSearch in Google Scholar

[161] Baud O, Emilie D, Pelletier E, Lacaze-Masmonteil T, Zupan V, Fernandez H, et al. Amniotic fluid concentrations of interleukin-1beta, interleukin-6 and TNF-alpha in chorioamnionitis before 32 weeks of gestation: histological associations and neonatal outcome. Br J Obstet Gynaecol. 1999;106:72–7.10.1111/j.1471-0528.1999.tb08088.xSearch in Google Scholar

[162] Hsu CD, Aversa K, Meaddough E. The role of amniotic fluid interleukin-6, and cell adhesion molecules, intercellular adhesion molecule-1 and leukocyte adhesion molecule-1, in intra-amniotic infection. Am J Reprod Immunol. 2000;43:251–4.10.1111/j.8755-8920.2000.430501.xSearch in Google Scholar

[163] Harirah H, Donia SE, Hsu CD. Amniotic fluid matrix metalloproteinase-9 and interleukin-6 in predicting intra-amniotic infection. Obstet Gynecol. 2002;99:80–4.10.1097/00006250-200201000-00016Search in Google Scholar

[164] Oh KJ, Park KH, Kim SN, Jeong EH, Lee SY, Yoon HY. Predictive value of intra-amniotic and serum markers for inflammatory lesions of preterm placenta. Placenta. 2011;32:732–6.10.1016/j.placenta.2011.07.080Search in Google Scholar

[165] 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.e1–125.e15.10.1016/j.ajog.2013.11.032Search in Google Scholar

[166] 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. 2015;28:1510–9.10.3109/14767058.2014.961417Search in Google Scholar

[167] 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 membranes: a step toward the early treatment of acute intra-amniotic inflammation/infection. J Matern Fetal Neonatal Med. 2016;29:360–7.10.3109/14767058.2015.1006621Search in Google Scholar

[168] 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

[169] 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

[170] Romero R, Quintero R, Nores J, Avila C, Mazor M, Hanaoka S, et al. Amniotic fluid white blood cell count: a rapid and simple test to diagnose microbial invasion of the amniotic cavity and predict preterm delivery. Am J Obstet Gynecol. 1991; 165(4 Pt 1):821–30.10.1016/0002-9378(91)90423-OSearch in Google Scholar

[171] Romero R, Jimenez C, Lohda AK, Nores J, Hanaoka S, Avila C, et al. Amniotic fluid glucose concentration: a rapid and simple method for the detection of intraamniotic infection in preterm labor. Am J Obstet Gynecol. 1990;163:968–74.10.1016/0002-9378(90)91106-MSearch in Google Scholar

[172] Romero R, Emamian M, Quintero R, Wan M, Hobbins JC, Mazor M, et al. The value and limitations of the Gram stain examination in the diagnosis of intraamniotic infection. Am J Obstet Gynecol. 1988;159:114–9.10.1016/0002-9378(88)90503-0Search in Google Scholar

[173] 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

[174] 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. 2014:1–17.10.3109/14767058.2014.954243Search in Google Scholar PubMed PubMed Central

[175] 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. 2015;28:1394–409.10.3109/14767058.2014.958463Search in Google Scholar PubMed PubMed Central

[176] 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

[177] Romero R, Chaemsaithong P, Korzeniewski SJ, Tarca AL, Bhatti G, Xu Z, et al. Clinical chorioamnionitis at term II: the intra-amniotic inflammatory response. J Perinat Med. 2016;44:5–22.10.1515/jpm-2015-0045Search in Google Scholar PubMed PubMed Central

[178] Romero R, Chaemsaithong P, Korzeniewski SJ, Kusanovic JP, Docheva N, Martinez-Varea A, 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;44:23–32.10.1515/jpm-2015-0044Search in Google Scholar PubMed PubMed Central

[179] Romero R, Chaemsaithong P, Docheva N, Korzeniewski SJ, Tarca AL, Bhatti G, et al. Clinical chorioamnionitis at term IV: the maternal plasma cytokine profile. J Perinat Med. 2016;44:77–98.10.1515/jpm-2015-0103Search in Google Scholar PubMed PubMed Central

[180] Romero R, Chaemsaithong P, Docheva N, Korzeniewski SJ, Tarca AL, Bhatti G, et al. Clinical chorioamnionitis at term V: umbilical cord plasma cytokine profile in the context of a systemic maternal inflammatory response. J Perinat Med. 2016;44:53–76.10.1515/jpm-2015-0121Search in Google Scholar PubMed PubMed Central

[181] Romero R, Chaemsaithong P, Docheva N, Korzeniewski SJ, Kusanovic JP, Yoon BH, et al. Clinical chorioamnionitis at term VI: acute chorioamnionitis and funisitis according to the presence or absence of microorganisms and inflammation in the amniotic cavity. J Perinat Med. 2016;44:33–51.10.1515/jpm-2015-0119Search in Google Scholar

[182] Weiss HB, Songer TJ, Fabio A. Fetal deaths related to maternal injury. J Am Med Assoc. 2001;286:1863–8.10.1001/jama.286.15.1863Search in Google Scholar

[183] Chibber R, Al-Harmi J, Fouda M, El-Saleh E. Motor-vehicle injury in pregnancy and subsequent feto-maternal outcomes: of grave concern. J Matern Fetal Neonatal Med. 2015;28:399–402.10.3109/14767058.2014.918094Search in Google Scholar

[184] Lufkin EG, Nelson RL, Hill LM, Melton LJ, 3rd, O’Fallon WM, Evans AT, 3rd. An analysis of diabetic pregnancies at Mayo Clinic, 1950–79. Diabetes Care. 1984;7:539–47.10.2337/diacare.7.6.539Search in Google Scholar

[185] Montoro MN, Myers VP, Mestman JH, Xu Y, Anderson BG, Golde SH. Outcome of pregnancy in diabetic ketoacidosis. Am J Perinatol. 1993;10:17–20.10.1055/s-2007-994692Search in Google Scholar

[186] Pinto ME, Villena JE. Diabetic ketoacidosis during gestational diabetes. A case report. Diabetes Res Clin Pract. 2011;93:e92–4.10.1016/j.diabres.2011.05.014Search in Google Scholar

[187] White BA, Labhsetwar SA, Mian AN. Streptococcus bovis bacteremia and fetal death. Obstet Gynecol. 2002; 100(5 Pt 2):1126–9.10.1097/00006250-200211001-00029Search in Google Scholar

[188] Barth T, Broscheit J, Bussen S, Dietl J. Maternal sepsis and intrauterine fetal death resulting from Candida tropicalis chorioamnionitis in a woman with a retained intrauterine contraceptive device. Acta Obstet Gynecol Scand. 2002;81:981–2.10.1034/j.1600-0412.2002.811014.xSearch in Google Scholar

[189] Malcolm G, Ellwood D, Devonald K, Beilby R, Henderson-Smart D. Absent or reversed end diastolic flow velocity in the umbilical artery and necrotising enterocolitis. Arch Dis Child. 1991;66(7 Spec No):805–7.10.1136/adc.66.7_Spec_No.805Search in Google Scholar

[190] Valcamonico A, Danti L, Frusca T, Soregaroli M, Zucca S, Abrami F, et al. Absent end-diastolic velocity in umbilical artery: risk of neonatal morbidity and brain damage. Am J Obstet Gynecol. 1994;170:796–801.10.1016/S0002-9378(94)70285-3Search in Google Scholar

[191] Naeye RL. Maternal floor infarction. Hum Pathol. 1985;16:823–8.10.1016/S0046-8177(85)80254-9Search in Google Scholar

[192] Andres RL, Kuyper W, Resnik R, Piacquadio KM, Benirschke K. The association of maternal floor infarction of the placenta with adverse perinatal outcome. Am J Obstet Gynecol. 1990;163:935–8.10.1016/0002-9378(90)91100-QSearch in Google Scholar

[193] Mandsager NT, Bendon R, Mostello D, Rosenn B, Miodovnik M, Siddiqi TA. Maternal floor infarction of the placenta: prenatal diagnosis and clinical significance. Obstet Gynecol. 1994;83(5 Pt 1):750–4.Search in Google Scholar

[194] Yeom W, Paik ES, An JJ, Oh SY, Suk-Joo Choi SJ, Roh CR, et al. Clinical characteristics and perinatal outcome of fetal hydrops. Obstet Gynecol Sci. 2015;58:90–7.10.5468/ogs.2015.58.2.90Search in Google Scholar

[195] Kim SA, Lee SM, Hong JS, Lee J, Park CW, Kim BJ, et al. Ultrasonographic severity scoring of non-immune hydrops: a predictor of perinatal mortality. J Perinat Med. 2015;43:53–9.10.1515/jpm-2013-0208Search in Google Scholar

[196] Warburton D. Chromosomal causes of fetal death. Clin Obstet Gynecol. 1987;30:268–77.10.1097/00003081-198706000-00006Search in Google Scholar

[197] Wapner RJ. Genetics of stillbirth. Clin Obstet Gynecol. 2010;53:628–34.10.1097/GRF.0b013e3181ee2793Search in Google Scholar

[198] Bowman JM. Alloimmunization in twin pregnancies. Am J Obstet Gynecol. 1985;153:7–13.10.1016/0002-9378(85)90581-2Search in Google Scholar

[199] Branch DW. Immunologic disease and fetal death. Clin Obstet Gynecol. 1987;30:295–311.10.1097/00003081-198706000-00009Search in Google Scholar PubMed

[200] Hara S. Current pathological perspectives on chronic rejection in renal allografts. Clin Exp Nephrol. 2016. [Epub ahead of print].10.1007/s10157-016-1361-xSearch in Google Scholar PubMed

[201] Neuberger J. Incidence, timing, and risk factors for acute and chronic rejection. Liver Transpl Surg. 1999;5(4 Suppl 1):S30–36.Search in Google Scholar

[202] Stites E, Le Quintrec M, Thurman JM. The Complement System and Antibody-Mediated Transplant Rejection. J Immunol. 2015;195:5525–31.10.4049/jimmunol.1501686Search in Google Scholar PubMed PubMed Central

[203] Stegall MD, Chedid MF, Cornell LD. The role of complement in antibody-mediated rejection in kidney transplantation. Nat Rev Nephrol. 2012;8:670–8.10.1038/nrneph.2012.212Search in Google Scholar

[204] Arcasoy SM, Kotloff RM. Lung transplantation. N Engl J Med. 1999;340:1081–91.10.1056/NEJM199904083401406Search in Google Scholar

[205] Nankivell BJ, Chapman JR. Chronic allograft nephropathy: current concepts and future directions. Transplantation. 2006;81:643–54.10.1097/01.tp.0000190423.82154.01Search in Google Scholar

[206] Chang DH, Kobashigawa JA. Current diagnostic and treatment strategies for cardiac allograft vasculopathy. Expert Rev Cardiovasc Ther. 2015;13:1147–54.10.1586/14779072.2015.1087312Search in Google Scholar

[207] Verleden GM, Vos R, Vanaudenaerde B, Dupont L, Yserbyt J, Van Raemdonck D, et al. Current views on chronic rejection after lung transplantation. Transpl Int. 2015;28:1131–9.10.1111/tri.12579Search in Google Scholar

[208] Burton GJ, Jauniaux E. What is the placenta? Am J Obstet Gynecol. 2015;213(4 Suppl):S6 e1, S6–8.10.1016/j.ajog.2015.07.050Search in Google Scholar

[209] Nelson DM. How the placenta affects your life, from womb to tomb. Am J Obstet Gynecol. 2015;213(4 Suppl):S12–13.10.1016/j.ajog.2015.08.015Search in Google Scholar

[210] Thornburg KL, Marshall N. The placenta is the center of the chronic disease universe. Am J Obstet Gynecol. 2015; 213(4 Suppl):S14–20.10.1016/j.ajog.2015.08.030Search in Google Scholar

[211] Bedin M, Alsat E, Tanguy G, Cedard L. Placental sulfatase deficiency: clinical and biochemical study of 16 cases. Eur J Obstet Gynecol Reprod Biol. 1980;10:21–34.10.1016/0028-2243(80)90033-7Search in Google Scholar

[212] Abrahams VM, Bole-Aldo P, Kim YM, Straszewski-Chavez SL, Chaiworapongsa T, Romero R, et al. Divergent trophoblast responses to bacterial products mediated by TLRs. J Immunol. 2004;173:4286–96.10.4049/jimmunol.173.7.4286Search in Google Scholar PubMed

[213] Mor G. Inflammation and pregnancy: the role of toll-like receptors in trophoblast-immune interaction. Ann N Y Acad Sci. 2008;1127:121–8.10.1196/annals.1434.006Search in Google Scholar

[214] Abrahams VM, Aldo PB, Murphy SP, Visintin I, Koga K, Wilson G, et al. TLR6 modulates first trimester trophoblast responses to peptidoglycan. J Immunol. 2008;180:6035–43.10.4049/jimmunol.180.9.6035Search in Google Scholar

[215] Cardenas I, Means RE, Aldo P, Koga K, Lang SM, Booth C, et al. Viral infection of the placenta leads to fetal inflammation and sensitization to bacterial products predisposing to preterm labor. J Immunol. 2010;185:1248–57.10.4049/jimmunol.1000289Search in Google Scholar

[216] Cardenas I, Mor G, Aldo P, Lang SM, Stabach P, Sharp A, et al. Placental viral infection sensitizes to endotoxin-induced pre-term labor: a double hit hypothesis. Am J Reprod Immunol. 2011;65:110–7.10.1111/j.1600-0897.2010.00908.xSearch in Google Scholar

[217] Liu J, Feng Y, Wang J, Li X, Lei C, Jin D, et al. An “immune barrier” is formed in the placenta by hepatitis B immunoglobulin to protect the fetus from hepatitis B virus infection from the mother. Hum Vaccin Immunother. 2015;11:2068–76.10.1080/21645515.2015.1010890Search in Google Scholar

[218] Mor G, Kwon JY. Trophoblast-microbiome interaction: a new paradigm on immune regulation. Am J Obstet Gynecol. 2015;213(4 Suppl):S131–7.10.1016/j.ajog.2015.06.039Search in Google Scholar

[219] Racicot K, Kwon JY, Aldo P, Abrahams V, El-Guindy A, Romero R, et al. Type I interferon regulates the placental inflammatory response to bacteria and is targeted by virus: mechanism of polymicrobial infection-induced preterm birth. Am J Reprod Immunol. 2016;75:451–60.10.1111/aji.12501Search in Google Scholar

[220] Mor G. Placental inflammatory response to zika virus may affect fetal brain development. Am J Reprod Immunol. 2016;75:421–2.10.1111/aji.12505Search in Google Scholar

[221] 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

[222] 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

[223] Romero R, Maymon E, Pacora P, Gomez R, Mazor M, Yoon BH, et al. Further observations on the fetal inflammatory response syndrome: a potential homeostatic role for the soluble receptors of tumor necrosis factor alpha. Am J Obstet Gynecol. 2000;183:1070–7.10.1067/mob.2000.108885Search in Google Scholar

[224] 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

[225] 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;40:19–32.10.1515/JPM.2011.100Search in Google Scholar

[226] Heazell AE, Martindale EA. Can post-mortem examination of the placenta help determine the cause of stillbirth? J Obstet Gynaecol. 2009;29:225–8.10.1080/01443610802716042Search in Google Scholar

[227] Amir H, Weintraub A, Aricha-Tamir B, Apel-Sarid L, Holcberg G, Sheiner E. A piece in the puzzle of intrauterine fetal death: pathological findings in placentas from term and preterm intrauterine fetal death pregnancies. J Matern Fetal Neonatal Med. 2009;22:759–64.10.3109/14767050902929396Search in Google Scholar

[228] Tellefsen CH, Vogt C. How important is placental examination in cases of perinatal deaths? Pediatr Dev Pathol. 2011;14:99–104.10.2350/10-07-0870-OA.1Search in Google Scholar

[229] Bonetti LR, Ferrari P, Trani N, Maccio L, Laura S, Giuliana S, et al. The role of fetal autopsy and placental examination in the causes of fetal death: a retrospective study of 132 cases of stillbirths. Arch Gynecol Obstet. 2011;283:231–41.10.1007/s00404-009-1317-4Search in Google Scholar

[230] Flenady V, Middleton P, Smith GC, Wes Duke W, Erwich JJ, Khong TY, et al. Stillbirths: the way forward in high-income countries. Lancet. 2011;377:1703–17.10.1016/S0140-6736(11)60064-0Search in Google Scholar

[231] Kim YM, Chaemsaithong P, Romero R, Shaman M, Kim CJ, Kim JS, et al. The frequency of acute atherosis in normal pregnancy and preterm labor, preeclampsia, small-for-gestational age, fetal death and midtrimester spontaneous abortion. J Matern Fetal Neonatal Med. 2015;28:2001–9.10.3109/14767058.2014.976198Search in Google Scholar PubMed PubMed Central

[232] Pinar H, Goldenberg RL, Koch MA, Heim-Hall J, Hawkins HK, Shehata B, et al. Placental findings in singleton stillbirths. Obstet Gynecol. 2014;123(2 Pt 1):325–36.10.1097/AOG.0000000000000100Search in Google Scholar PubMed PubMed Central

[233] Roescher AM, Timmer A, Erwich JJ, Bos AF. Placental pathology, perinatal death, neonatal outcome, and neurological development: a systematic review. PLoS One. 2014;9:e89419.10.1371/journal.pone.0089419Search in Google Scholar

[234] Khong TY, Mooney EE, Ariel I, Balmus NC, Boyd TK, Brundler MA, et al. Sampling and Definitions of Placental Lesions: Amsterdam Placental Workshop Group Consensus Statement. Arch Pathol Lab Med. 2016;140:698–713.10.5858/arpa.2015-0225-CCSearch in Google Scholar

[235] Katzman PJ, Genest DR. Maternal floor infarction and massive perivillous fibrin deposition: histological definitions, association with intrauterine fetal growth restriction, and risk of recurrence. Pediatr Dev Pathol. 2002;5:159–64.10.1007/s10024001-0195-ySearch in Google Scholar

[236] Whitten AE, Romero R, Korzeniewski SJ, Tarca AL, Schwartz AG, Yeo L, et al. Evidence of an imbalance of angiogenic/antiangiogenic factors in massive perivillous fibrin deposition (maternal floor infarction): a placental lesion associated with recurrent miscarriage and fetal death. Am J Obstet Gynecol. 2013;208:310.e1–310.e311.10.1016/j.ajog.2013.01.017Search in Google Scholar

[237] Chaiworapongsa T, Romero R, Korzeniewski SJ, Chaemsaithong P, Hernandez-Andrade E, Segars JH, et al. Pravastatin to prevent recurrent fetal death in massive perivillous fibrin deposition of the placenta (MPFD). J Matern Fetal Neonatal Med. 2016;29:855–62.10.3109/14767058.2015.1022864Search in Google Scholar

[238] Oppenheim JJ, Yang D. Alarmins: chemotactic activators of immune responses. Curr Opin Immunol. 2005;17:359–65.10.1016/j.coi.2005.06.002Search in Google Scholar

[239] 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

[240] 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

[241] Romero R, Emamian M, Quintero R, Wan M, Hobbins JC, Mitchell MD. Amniotic fluid prostaglandin levels and intra-amniotic infections. Lancet. 1986;1:1380.10.1016/S0140-6736(86)91685-5Search in Google Scholar

[242] 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

[243] 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

[244] Romero R, Wu YK, Sirtori M, Oyarzun E, Mazor M, Hobbins JC, et al. Amniotic fluid concentrations of prostaglandin F2 alpha, 13,14-dihydro-15-keto-prostaglandin F2 alpha (PGFM) and 11-deoxy-13,14-dihydro-15-keto-11, 16-cyclo-prostaglandin E2 (PGEM-LL) in preterm labor. Prostaglandins. 1989;37:149–61.10.1016/0090-6980(89)90038-5Search in Google Scholar

[245] 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

[246] Bry K, Hallman M. Prostaglandins, inflammation, and preterm labor. J Perinatol. 1989;9:60–5.Search in Google Scholar

[247] 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

[248] 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 PubMed

[249] 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

[250] 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

[251] Park JY, Romero R, Lee J, Chaemsaithong P, Chaiyasit N, Yoon BH. An elevated amniotic fluid prostaglandin F2alpha concentration is associated with intra-amniotic inflammation/infection, and clinical and histologic chorioamnionitis, as well as impending preterm delivery in patients with preterm labor and intact membranes. J Matern Fetal Neonatal Med. 2016;29:2563–72.10.3109/14767058.2015.1094794Search in Google Scholar

[252] Maddipati KR, Romero R, Chaiworapongsa T, Chaemsaithong P, Zhou SL, Zhonghui X, et al. Lipidomic analysis of patients with microbial invasion of the amniotic cavity reveals up-regulation of leukotriene B4. FASEB J. 2016;30:3296–307.10.1096/fj.201600583RSearch in Google Scholar PubMed PubMed Central

[253] Lim SY, Kim YH, Kim CH, Cho MK, Kim JW, Kang WD, et al. The effect of a Foley catheter balloon on cervical ripening. J Obstet Gynaecol. 2013;33:830–8.10.3109/01443615.2013.831043Search in Google Scholar PubMed

[254] Liggins G. Cervical ripening as an inflammatory reaction. In: Ellwood E, Anderson A, editors. The cervix in pregnancy and labor: clinical and biochemical investigations. Edinburgh: Churchill Livingstone; 1981. p. 1–9.Search in Google Scholar

[255] Bokstrom H, Brannstrom M, Alexandersson M, Norstrom A. Leukocyte subpopulations in the human uterine cervical stroma at early and term pregnancy. Hum Reprod. 1997;12:586–90.10.1093/humrep/12.3.586Search in Google Scholar PubMed

[256] Mackler AM, Iezza G, Akin MR, McMillan P, Yellon SM. Macrophage trafficking in the uterus and cervix precedes parturition in the mouse. Biol Reprod. 1999;61:879–83.10.1095/biolreprod61.4.879Search in Google Scholar PubMed

[257] Kelly RW. Inflammatory mediators and cervical ripening. J Reprod Immunol. 2002;57:217–24.10.1016/S0165-0378(02)00007-4Search in Google Scholar

[258] Osman I, Young A, Ledingham MA, Thomson AJ, Jordan F, Greer IA, et al. Leukocyte density and pro-inflammatory cytokine expression in human fetal membranes, decidua, cervix and myometrium before and during labour at term. Mol Hum Reprod. 2003;9:41–5.10.1093/molehr/gag001Search in Google Scholar PubMed

[259] Sakamoto Y, Moran P, Bulmer JN, Searle RF, Robson SC. Macrophages and not granulocytes are involved in cervical ripening. J Reprod Immunol. 2005;66:161–73.10.1016/j.jri.2005.04.005Search in Google Scholar PubMed

[260] Yellon SM, Ebner CA, Sugimoto Y. Parturition and recruitment of macrophages in cervix of mice lacking the prostaglandin F receptor. Biol Reprod. 2008;78:438–44.10.1095/biolreprod.107.063404Search in Google Scholar PubMed PubMed Central

[261] Yellon SM, Oshiro BT, Chhaya TY, Lechuga TJ, Dias RM, Burns AE, et al. Remodeling of the cervix and parturition in mice lacking the progesterone receptor B isoform. Biol Reprod. 2011;85:498–502.10.1095/biolreprod.111.091983Search in Google Scholar PubMed PubMed Central

[262] Clyde LA, Lechuga TJ, Ebner CA, Burns AE, Kirby MA, Yellon SM. Transection of the pelvic or vagus nerve forestalls ripening of the cervix and delays birth in rats. Biol Reprod. 2011;84:587–94.10.1095/biolreprod.110.086207Search in Google Scholar PubMed PubMed Central

[263] Payne KJ, Clyde LA, Weldon AJ, Milford TA, Yellon SM. Residency and activation of myeloid cells during remodeling of the prepartum murine cervix. Biol Reprod. 2012;87:106.10.1095/biolreprod.112.101840Search in Google Scholar PubMed PubMed Central

[264] Myers DA. The recruitment and activation of leukocytes into the immune cervix: further support that cervical remodeling involves an immune and inflammatory mechanism. Biol Reprod. 2012;87:107.10.1095/biolreprod.112.105049Search in Google Scholar PubMed

[265] Thomson AJ, Telfer JF, Young A, Campbell S, Stewart CJ, Cameron IT, et al. Leukocytes infiltrate the myometrium during human parturition: further evidence that labour is an inflammatory process. Hum Reprod. 1999;14:229–36.10.1093/humrep/14.1.229Search in Google Scholar

[266] Shynlova O, Tsui P, Dorogin A, Lye SJ. Monocyte chemoattractant protein-1 (CCL-2) integrates mechanical and endocrine signals that mediate term and preterm labor. J Immunol. 2008;181:1470–9.10.4049/jimmunol.181.2.1470Search in Google Scholar PubMed

[267] Shynlova O, Tsui P, Jaffer S, Lye SJ. Integration of endocrine and mechanical signals in the regulation of myometrial functions during pregnancy and labour. Eur J Obstet Gynecol Reprod Biol. 2009;144(Suppl 1):S2–10.10.1016/j.ejogrb.2009.02.044Search in Google Scholar

[268] Hamilton S, Oomomian Y, Stephen G, Shynlova O, Tower CL, Garrod A, et al. Macrophages infiltrate the human and rat decidua during term and preterm labor: evidence that decidual inflammation precedes labor. Biol Reprod. 2012;86:39.10.1095/biolreprod.111.095505Search in Google Scholar

[269] Shynlova O, Nedd-Roderique T, Li Y, Dorogin A, Nguyen T, Lye SJ. Infiltration of myeloid cells into decidua is a critical early event in the labour cascade and post-partum uterine remodelling. J Cell Mol Med. 2013;17:311–24.10.1111/jcmm.12012Search in Google Scholar

[270] Shynlova O, Lee YH, Srikhajon K, Lye SJ. Physiologic uterine inflammation and labor onset: integration of endocrine and mechanical signals. Reprod Sci. 2013;20:154–67.10.1177/1933719112446084Search in Google Scholar

[271] Arenas-Hernandez M, Romero R, St Louis D, Hassan SS, Kaye EB, Gomez-Lopez N. An imbalance between innate and adaptive immune cells at the maternal-fetal interface occurs prior to endotoxin-induced preterm birth. Cell Mol Immunol. 2016;13:462–73.10.1038/cmi.2015.22Search in Google Scholar

[272] Ammala M, Nyman T, Salmi A, Rutanen EM. The interleukin-1 system in gestational tissues at term: effect of labour. Placenta. 1997;18:717–23.10.1016/S0143-4004(97)90014-XSearch in Google Scholar

[273] Osman I, Young A, Jordan F, Greer IA, Norman JE. Leukocyte density and proinflammatory mediator expression in regional human fetal membranes and decidua before and during labor at term. J Soc Gynecol Investig. 2006;13:97–103.10.1016/j.jsgi.2005.12.002Search in Google Scholar PubMed

[274] Gomez-Lopez N, Estrada-Gutierrez G, Jimenez-Zamudio L, Vega-Sanchez R, Vadillo-Ortega F. Fetal membranes exhibit selective leukocyte chemotaxic activity during human labor. J Reprod Immunol. 2009;80:122–31.10.1016/j.jri.2009.01.002Search in Google Scholar PubMed

[275] Gomez-Lopez N, Vadillo-Perez L, Hernandez-Carbajal A, Godines-Enriquez M, Olson DM, Vadillo-Ortega F. Specific inflammatory microenvironments in the zones of the fetal membranes at term delivery. Am J Obstet Gynecol. 2011;205:235.e15–24.10.1016/j.ajog.2011.04.019Search in Google Scholar PubMed

[276] Gomez-Lopez N, Vadillo-Perez L, Nessim S, Olson DM, Vadillo-Ortega F. Choriodecidua and amnion exhibit selective leukocyte chemotaxis during term human labor. Am J Obstet Gynecol. 2011;204:364.e9–16.10.1016/j.ajog.2010.11.010Search in Google Scholar PubMed

[277] Gomez-Lopez N, Vega-Sanchez R, Castillo-Castrejon M, Romero R, Cubeiro-Arreola K, Vadillo-Ortega F. Evidence for a role for the adaptive immune response in human term parturition. Am J Reprod Immunol. 2013;69:212–30.10.1111/aji.12074Search in Google Scholar

[278] Alonso A, Soto I, Urgelles MF, Corte JR, Rodriguez MJ, Pinto CR. Acquired and inherited thrombophilia in women with unexplained fetal losses. Am J Obstet Gynecol. 2002;187:1337–42.10.1067/mob.2002.126849Search in Google Scholar

[279] Rey E, Kahn SR, David M, Shrier I. Thrombophilic disorders and fetal loss: a meta-analysis. Lancet. 2003;361:901–8.10.1016/S0140-6736(03)12771-7Search in Google Scholar

[280] Lamont RF, Sobel JD, Vaisbuch E, Kusanovic JP, Mazaki-Tovi S, Kim SK, et al. Parvovirus B19 infection in human pregnancy. Br J Obstet Gynaecol. 2011;118:175–86.10.1111/j.1471-0528.2010.02749.xSearch in Google Scholar PubMed PubMed Central

[281] Maymon E, Romero R, Bhatti G, Chaemsaithong P, Gomez-Lopez N, Panaitescu B, et al. Chronic inflammatory lesions of the placenta are associated with an up-regulation of amniotic fluid CXCR3: a marker of allograft rejection. J Perinatal Med. 2017. Accepted; in press.10.1515/jpm-2017-0042Search in Google Scholar PubMed PubMed Central


Supplemental Material:

The online version of this article (DOI https://doi.org/10.1515/jpm-2017-0073) offers supplementary material, available to authorized users.


Received: 2017-3-3
Accepted: 2017-3-8
Published Online: 2017-9-1
Published in Print: 2017-10-26

©2017 Walter de Gruyter GmbH, Berlin/Boston

Downloaded on 2.6.2024 from https://www.degruyter.com/document/doi/10.1515/jpm-2017-0073/html
Scroll to top button