Skip to content
Publicly Available Published by De Gruyter August 22, 2017

Chronic inflammatory lesions of the placenta are associated with an up-regulation of amniotic fluid CXCR3: A marker of allograft rejection

  • Eli Maymon , Roberto Romero EMAIL logo , Gaurav Bhatti , Piya Chaemsaithong , Nardhy Gomez-Lopez , Bogdan Panaitescu , Noppadol Chaiyasit , Percy Pacora , Zhong Dong , Sonia S. Hassan and Offer Erez EMAIL logo

Abstract

Objective:

The objective of this study is to determine whether the amniotic fluid (AF) concentration of soluble CXCR3 and its ligands CXCL9 and CXCL10 changes in patients whose placentas show evidence of chronic chorioamnionitis or other placental lesions consistent with maternal anti-fetal rejection.

Methods:

This retrospective case-control study included 425 women with (1) preterm delivery (n=92); (2) term in labor (n=68); and (3) term not in labor (n=265). Amniotic fluid CXCR3, CXCL9 and CXCL10 concentrations were determined by ELISA.

Results:

(1) Amniotic fluid concentrations of CXCR3 and its ligands CXCL9 and CXCL10 are higher in patients with preterm labor and maternal anti-fetal rejection lesions than in those without these lesions [CXCR3: preterm labor and delivery with maternal anti-fetal rejection placental lesions (median, 17.24 ng/mL; IQR, 6.79–26.68) vs. preterm labor and delivery without these placental lesions (median 8.79 ng/mL; IQR, 4.98–14.7; P=0.028)]; (2) patients with preterm labor and chronic chorioamnionitis had higher AF concentrations of CXCL9 and CXCL10, but not CXCR3, than those without this lesion [CXCR3: preterm labor with chronic chorioamnionitis (median, 17.02 ng/mL; IQR, 5.57–26.68) vs. preterm labor without chronic chorioamnionitis (median, 10.37 ng/mL; IQR 5.01–17.81; P=0.283)]; (3) patients with preterm labor had a significantly higher AF concentration of CXCR3 than those in labor at term regardless of the presence or absence of placental lesions.

Conclusion:

Our findings support a role for maternal anti-fetal rejection in a subset of patients with preterm labor.

Introduction

The fetus and placenta are semi-allografts that express paternal antigens, and tolerance of such antigens is considered a central mechanism for reproductive success [1], [2], [3], [4], [5], [6], [7], [8], [9]. A breakdown of maternal-fetal tolerance has been implicated in the mechanisms of disease responsible for a subset of recurrent pregnancy loss [10], fetal death [11], [12], preterm labor [13], [14], [15], [16], [17], [18], preterm prelabor rupture of the membranes (preterm PROM) [15] and other obstetrical complications [19], [20], [21], [22], [23], [24].

The most common placental pathologic lesion in late spontaneous preterm labor and birth is chronic chorioamnionitis [22], [25], which is considered a manifestation of maternal anti-fetal rejection. In this lesion, maternal CD8+ T cells infiltrate the chorioamniotic membranes; thus, lymphocytes from the host (i.e. mother) infiltrate the semi-allograft fetus. There is evidence that CD8+ cytotoxic T cells can establish direct contact with trophoblast cells in the chorion laeve and induce apoptosis [11], [25]. Damage of the trophoblast in the chorion laeve is considered a mechanism of disease for preterm labor and preterm PROM [25], which reflects a clinical manifestation of rejection.

The proposed mechanisms whereby maternal CD8+ T cells infiltrate the chorioamniotic membranes involve the generation of a T-cell chemokine gradient from the amniotic cavity [13]. Increased concentrations of the T-cell chemokines CXCL9, CXCL10 and CXCL11 are thought to be responsible for the chemotaxis of T cells from the peripheral circulation into the decidua and, subsequently, the chorioamniotic membranes [13], [25]. The receptor for CXCL9, CXCL10 and CXCL11 is CXCR3 [26], [27], [28], [29], [30]. The importance of CXCR3 in the mechanisms of allograft rejection is demonstrated by the observation that either gene deletion or protein neutralization of the receptor protects against the pancreatic islets and cardiac allograft rejection [31], [32].

CXCR3 is involved in the chemotaxis of activated T cells, dendritic cells and natural killer (NK) cells [33], [34] as well as angiogenesis [34], [35], [36], [37], [38], [39]. This chemokine receptor is mainly expressed on T helper (Th)1 cells [40], [41], [42], [43] and is up-regulated in activated lymphocytes recruited to the site of inflammation [34], [37]. The expression of CXCR3 and its ligands (CXCL9, CXCL10 and CXCL11) is increased during allograft rejection [44], [45], [46], [47], [48], [49], [50], [51], [52], [53], [54], [55], [56], [57], [58], [59], [60], [61], [62]. CXCR3 is also expressed by the placenta (villous cytotrophoblasts and syncytiotrophoblasts) [63], [64], the fetal membranes, and the choriodecidua of women who deliver term or preterm [64], [65]. In addition, CXCL10 concentration in the amniotic fluid is greater in women who undergo spontaneous preterm labor with chronic chorioamnionitis than in those without this placental lesion [13], [66]. CXCL10 concentration also increases in the amniotic fluid during the mid-trimester of pregnancy in patients who undergo spontaneous preterm birth after 32 weeks of gestation compared to those who deliver at term [67]. Therefore, it is likely that CXCR3 plays a role in the recruitment of T cells in chronic inflammatory lesions of the placenta, which represent maternal anti-fetal rejection.

The objective of this study is to determine whether the amniotic fluid concentration of CXCR3 and its ligands changes in patients whose placentas had chronic chorioamnionitis or other placental lesions consistent with maternal anti-fetal rejection (villitis of unknown etiology and chronic deciduitis with plasma cells).

Materials and methods

Characteristics of the study population

The retrospective case-control study included 425 patients in the following groups: (1) spontaneous preterm labor and delivery (n=92); (2) term in labor (n=68); and (3) term not in labor (n=265). All samples were obtained from the Bank of Biological Materials at Wayne State University, the Detroit Medical Center, and the Perinatology Research Branch of the Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD) (Detroit, MI, USA). The inclusion criteria were: (1) singleton gestation; (2) transabdominal amniocentesis performed for microbiological studies in patients with the diagnosis of preterm labor at less than 36 weeks of gestation [68]; (3) intraoperative transabdominal amniocentesis performed for research purposes in patients at term with and without labor; (4) a live-born fetus with available neonatal outcomes; and (5) an available placental pathology report. Patients were excluded from this study if chromosomal or structural fetal anomalies or placenta previa was present.

All patients provided written informed consent, and the use of biological specimens and clinical data for research purposes was approved by the Institutional Review Boards of Wayne State University and NICHD.

Biological samples and analysis

Amniotic fluid samples were transported to the clinical laboratory in capped sterile syringes and cultured for aerobic and anaerobic bacteria, including genital Mycoplasmas. Evaluation of the white blood cell (WBC) count, glucose concentration and Gram stain of the amniotic fluid samples was performed shortly after collection. Amniotic fluid samples were centrifuged at 1300× g for 10 min at 4°C shortly after collection and stored at −70°C until analysis. Amniotic fluid concentrations of CXCL9, CXCL10 and CXCR3 were measured using enzyme-linked immunosorbent assays (ELISA; CXCL9 and CXCL10 ELISA kits from R&D Systems, Minneapolis, MN, USA, and a CXCR3 ELISA kit from Cloud Clone, Houston, TX, USA). Immunoassays were performed following the manufacturers’ instructions. CXCL9 and CXCL10 were captured by specific pre-coated monoclonal antibodies and then detected by enzyme-linked polyclonal antibodies that were also specific to the same targets, respectively. The color developed from the substrate was proportional to the amount of target, and the target concentrations (pg/mL) in the samples were interpolated from the standard curve. The CXCR3 kit utilized a pre-coated antibody and a biotin-conjugated antibody specific to CXCR3. The CXCR3 concentrations (ng/mL) in the samples were interpolated from the standard curve. Sensitivity and intra- and inter-assay coefficients of variations for each assay are displayed in Table 1.

Table 1:

Assay characteristics for amniotic fluid CXCL3, CXCL9 and CXCL10.

AnalytesSensitivity (pg/mL)Intra-assay coefficients of variations (%)Inter-assay coefficients of variations (%)
CXCR31467.9519.3
CXCL914.95.344.29
CXCL103.295.965.50

Clinical definitions

Gestational age was determined by the last menstrual period and confirmed by ultrasound examination, or by ultrasound examination alone, if the sonographic determination of gestational age was not consistent with menstrual dating. Preterm labor was diagnosed by the presence of at least two regular uterine contractions every 10 min in association with cervical changes in patients with a gestational age between 20 and 36 6/7 weeks, which led to preterm delivery (defined as birth prior to the 37th week of gestation).

Study groups

Patients with spontaneous preterm labor and delivery and term delivery with and without labor were classified according to the presence or absence of chronic chorioamnionitis into the following study groups: (1) without chronic chorioamnionitis [spontaneous preterm labor (n=51), term in labor (n=48), and term not in labor (n=187)]; and (2) with chronic chorioamnionitis [preterm delivery (n=41), term in labor (n=20), and term not in labor (n=78)].

The presence of placental lesions associated with maternal anti-fetal rejection was examined as a secondary outcome in patients with spontaneous preterm delivery and term delivery with and without labor.

Sonographic assessment of the cervix

Transvaginal ultrasound examinations were performed using commercially available ultrasound systems (Acuson Sequoia, Siemens Medical Systems, Mountain View, CA, USA; Voluson 730 Expert™ or Voluson E8, GE Healthcare, Milwaukee, WI, USA) equipped with endovaginal transducers with frequency ranges of 5–7.5 MHz and 5–9 MHz, respectively. All sonographic examinations of the cervix were performed using a previously described technique [69], [70].

Placental examination

Placentas were collected in dry plastic containers with airtight lids and labeled by research personnel. The umbilical cord was cut at the place of insertion into the chorionic plate. Membrane rolls and pieces of the umbilical cord and placental disc (selected using a random-sequence generator for the purposes of placental sampling) were obtained within 30 min of delivery and placed in formalin. After overnight fixation in formalin, the tissue samples were embedded in paraffin and were further processed for histologic examination. A five micron-thick Hematoxylin and Eosin (H&E) section was taken from each paraffin block. The H&E sections of the chorioamniotic membrane roll (n=2), umbilical cord (n=2) and placental disc (n=2) were examined by pathologists who were blinded to the clinical diagnoses and outcomes.

The diagnosis of acute histologic chorioamnionitis was based on the presence of acute inflammatory changes in the extra-placental chorioamniotic membrane roll and/or the chorionic plate of the placenta, using previously described criteria [71], [72]. The grading and staging of placental lesions consistent with amniotic fluid infection is defined according to the Amniotic Fluid Infection Nosology Committee of the Perinatal Section of the Society for Pediatric Pathology as reported by Redline et al. [73]. Chronic placental inflammatory lesions included: (1) chronic chorioamnionitis; (2) villitis of unknown etiology; and (3) chronic deciduitis. Chronic chorioamnionitis was diagnosed when lymphocytic infiltration into the chorionic trophoblast layer or chorioamniotic connective tissue was observed [13], [25]. Villitis of unknown etiology was defined as the presence of lymphohistiocytic infiltration of varying proportions in the placental villous tree [19], [74], [75]. Chronic deciduitis was defined as the presence of lymphocytic infiltration into the decidua of the basal plate [76].

Placental lesions consistent with maternal anti-fetal rejection proposed by our group included chronic chorioamnionitis, villitis of unknown etiology or chronic deciduitis with plasma cells [14], [19].

Statistical analysis

All analyses were performed using the R software package [77]. The two-sided, two-sample Wilcoxon test (also known as the Mann-Whitney U test) was used to compare differences between the groups, and the obtained P-values were adjusted, for multiple comparisons, using the Holm-Bonferroni method. A P-value cut-off of 0.05 was used to determine statistical significance. The Fisher’s exact test was used for categorical variables. A linear model was used to compare the groups while adjusting for gestational age at amniocentesis.

Results

Clinical characteristics of the study population

The demographic and clinical characteristics are shown in Table 2. As expected, gestational age at amniocentesis and at delivery as well as birthweight were significantly lower in patients with spontaneous preterm delivery than in those who delivered at term (P<0.001 for each).

Table 2:

Demographic and clinical characteristics of the study population.

Spontaneous preterm labor (n=92)P-valueaTerm delivery without labor (n=265)Term delivery with labor (n=68)P-value
Maternal age (years)23 (21–28)0.1726 (23–30)25.5 (21.25–29.0)0.04
Nulliparity (%)26.1% (24)17.2% (19)25% (17)<0.001
Gestational age at amniocentesis (weeks)32.7 (29.4–34.3)<0.00139 (38.9–39.3)39 (38.4–40.3)0.38
Gestational age at delivery (weeks)33.4 (30.0–35.3)<0.00139 (38.9–39.3)39 (38.45–40.3)0.38
Birth weight (g)2003 (1243–2468)<0.0013375 (3130–3655)3323 (3110–3685)0.76
  1. Data are presented as median (interquartile range) or % (n);

  2. acomparison between spontaneous preterm labor and term delivery with labor.

In the preterm labor group, the median interval from amniocentesis to delivery is 1 day (range: 0–21 days).

Amniotic fluid CXCR3, CXCL9 and CXCL10 concentrations are higher in spontaneous preterm deliveries with placental lesions consistent with maternal anti-fetal rejection

Among patients presenting with spontaneous preterm labor who delivered preterm, the median of amniotic fluid CXCR3, CXCL9 and CXCL10 concentrations is significantly higher in those who have placental lesions consistent with maternal anti-fetal rejection compared to those without these lesions [CXCR3, CXCL9 and CXCL10; preterm delivery with placental lesions consistent with maternal anti-fetal rejection: median (IQR) 17.24 (ng/mL) (6.79–26.68); 0.35 (ng/mL) (0.16–0.86); and 3.12 (ng/mL) (1.46–4.4) vs. preterm delivery without placental lesions consistent with maternal anti-fetal rejection: median (IQR) 8.79 (ng/mL) (4.98–14.7); 0.10 (ng/mL) (0.07–0.18); and 1.12 (ng/mL) (0.65–1.47); P=0.028, P<0.001 and P<0.001, respectively] (Figure 1).

Figure 1: Amniotic fluid CXCR3, CXCL9 and CXCL10 concentrations (ng/mL) in patients with spontaneous preterm labor according to the absence or presence of placental lesions consistent with maternal anti-fetal rejection.Red line represents the median.
Figure 1:

Amniotic fluid CXCR3, CXCL9 and CXCL10 concentrations (ng/mL) in patients with spontaneous preterm labor according to the absence or presence of placental lesions consistent with maternal anti-fetal rejection.

Red line represents the median.

Amniotic fluid CXCL9 and CXCL10, but not CXCR3, concentrations are higher in spontaneous preterm deliveries with chronic chorioamnionitis

The median amniotic fluid CXCL9 and CXCL10, but not CXCR3, concentrations were significantly higher in patients with spontaneous preterm labor and delivery whose placentas had chronic chorioamnionitis than in those without chronic chorioamnionitis [CXCR3, CXCL9 and CXCL10 (median and interquartile range; IQR): preterm delivery with chronic chorioamnionitis: 17.02 (ng/mL) (5.57–26.68); 0.34 (ng/mL) (0.16–0.67); and 2.94 (ng/mL) (1.52–4.37) vs. preterm delivery without chronic chorioamnionitis: 10.37 (ng/mL) (5.01–17.81); 0.12 (ng/mL) (0.07–0.27); and 1.19 (ng/mL) (0.66–1.63); P=0.283, P=0.006 and P<0.001, respectively] (Figure 2).

Figure 2: Amniotic fluid CXCR3, CXCL9 and CXCL10 concentrations (ng/mL) in patients with spontaneous preterm labor according to the absence or presence of chronic chorioamnionitis.Red line represents the median.
Figure 2:

Amniotic fluid CXCR3, CXCL9 and CXCL10 concentrations (ng/mL) in patients with spontaneous preterm labor according to the absence or presence of chronic chorioamnionitis.

Red line represents the median.

CXCR3, CXCL9 and CXCL10 concentrations in the amniotic fluid of patients in labor at term

Median CXCR3, CXCL9 and CXCL10 concentrations in the amniotic fluid were not significantly different in patients with labor at term whose placenta had chronic chorioamnionitis compared to those without chronic chorioamnionitis, those whose placental lesions were consistent with maternal anti-fetal rejection and those without placental lesions consistent with maternal anti-fetal rejection (Table 3).

Table 3:

Amniotic fluid CXCR3, CXCL9 and CXCL10 concentrations in patients with spontaneous term labor according to the presence or absence of chronic chorioamnionitis/placental lesions consistent with maternal anti-fetal rejection.

AnalytesTerm Labor
Without chronic chorioamnionitis (n=48)With chronic chorioamnionitis (n=20 )P-valueWithout placental lesions consistent with maternal anti-fetal rejection (n=39 )With placental lesions consistent with maternal anti-fetal rejection (n=29)P-value
Median (IQR) (ng/mL)Median (IQR) (ng/mL)Median (IQR) (ng/mL)Median (IQR) (ng/mL)
CXCR31.39 (0.51–3.27)a2.57 (0.37–5.38)10.99 (0.51–2.98)2.57 (0.37–5.48)a1
CXCL90.1 (0.007–0.15)0.16 (0.03–0.27)10.1 (0.06–0.15)0.13 (0.06–0.25)1
CXCL100.5 (0.27–0.74)1.55 (0.25–2.14)0.3340.44 (0.26–0.75)0.76 (0.32–1.85)0.463
  1. aData are not available for one case; IQR=interquartile range.

Amniotic fluid CXCR3 concentration is higher in patients with spontaneous preterm labor and chronic chorioamnionitis

Among patients with chronic chorioamnionitis, the median amniotic fluid concentration of CXCR3, but not of CXCL10 or CXCL9, is significantly higher in patients with spontaneous preterm labor compared to those with labor at term [CXCR3, CXCL10, CXCL9, median (IQR); preterm delivery with chronic chorioamnionitis: 17.02 (ng/mL) (5.57–26.68); 2.94 (ng/mL) (1.52–4.37); and 0.34 (ng/mL) (0.16–0.67) vs. term in labor with chronic chorioamnionitis 2.57 (ng/mL) (0.37–5.38); 1.55 (ng/mL) (0.25–2.14); 0.16 (ng/mL) (0.03–0.27); P<0.001 for CXCR3 and not significant for CXCL10 and for CXCL9 after adjusting for gestational age] (Figure 3A).

Figure 3A: Amniotic fluid CXCR3, CXCL9 and CXCL10 concentrations (ng/mL) in patients with spontaneous preterm or term labor and the presence of chronic chorioamnionitis.Red line represents the median.
Figure 3A:

Amniotic fluid CXCR3, CXCL9 and CXCL10 concentrations (ng/mL) in patients with spontaneous preterm or term labor and the presence of chronic chorioamnionitis.

Red line represents the median.

Similar results were found in patients whose placentas were without chronic chorioamnionitis [CXCR3, CXCL10, CXCL9, median (IQR); spontaneous preterm labor and delivery without chronic chorioamnionitis 10.37 (ng/mL) (5.01–17.81); 1.19 (ng/mL) (0.66–1.63); and 0.12 (ng/mL) (0.07–0.27) vs. term in labor without chronic chorioamnionitis: 1.39 (ng/mL) (0.51–3.27); 0.5 (ng/mL) (0.27–0.74); and 0.1 (ng/mL) (0.07–0.15); P<0.01 for CXCR3 and not significant for CXCL9 and CXCL10 after adjusting for gestational age] (Figure 3B).

Figure 3B: Amniotic fluid CXCR3, CXCL9 and CXCL10 concentrations (ng/mL) in patients with spontaneous preterm or term labor and the absence of chronic chorioamnionitis.Red line represents the median.
Figure 3B:

Amniotic fluid CXCR3, CXCL9 and CXCL10 concentrations (ng/mL) in patients with spontaneous preterm or term labor and the absence of chronic chorioamnionitis.

Red line represents the median.

Amniotic fluid CXCR3 and CXCL9 concentrations in the amniotic fluid are higher in patients with spontaneous preterm labor and placental lesions consistent with maternal anti-fetal rejection

Among patients presenting with spontaneous preterm labor who subsequently delivered preterm, the median amniotic fluid concentrations of CXCR3 and CXCL9, but not CXCL10, were significantly higher in those who had placental lesions consistent with maternal anti-fetal rejection than those with labor at term who had similar lesions; preterm labor and delivery with placental lesions consistent with maternal anti-fetal rejection: median (IQR) 17.24 (ng/mL) (6.79–26.68); 0.35 (ng/mL) (0.16–0.86); 3.12 (ng/mL) (1.46–4.4) vs. term in labor with these placental lesions: median (IQR) 2.57 (ng/mL) (0.37–5.48); 0.13 (ng/mL) (0.06–0.25); and 0.76 (ng/mL) (0.32–1.85); P<0.001 for CXCR3 and P=0.003 for CXCL9 and not significant for CXCL10 after adjusting for gestational age] (Figure 4A).

Figure 4A: Amniotic fluid CXCR3, CXCL9 and CXCL10 concentrations (ng/mL) in patients with spontaneous preterm or term labor with placental lesions consistent with maternal anti-fetal rejection.Red line represents the median.
Figure 4A:

Amniotic fluid CXCR3, CXCL9 and CXCL10 concentrations (ng/mL) in patients with spontaneous preterm or term labor with placental lesions consistent with maternal anti-fetal rejection.

Red line represents the median.

Similarly, among patients presenting with spontaneous preterm labor who subsequently had a preterm delivery, the median amniotic fluid CXCR3, but not CXCL9 or CXCL10, concentration is significantly higher in those who did not have placental lesions consistent with maternal anti-fetal rejection than in patients with labor at term without these lesions [CXCR3, CXCL9 and CXCL10; preterm labor and delivery without placental lesions consistent with maternal anti-fetal rejection: median (IQR) 8.79 (ng/mL) (4.98–14.7); 0.10 (ng/mL) (0.07–0.18); 1.12 (ng/mL) (0.65–1.47) vs. term in labor without these placental lesions: median (IQR) 0.99 (ng/mL) (0.51–2.98); 0.1 (ng/mL) (0.06–0.15); and 0.44 (ng/mL) (0.26–0.75); P<0.001 for CXCR3 and not significant for CXCL9 and CXCL10 after adjusting for gestational age] (Figure 4B).

Figure 4B: Amniotic fluid CXCR3, CXCL9 and CXCL10 concentrations (ng/mL) in patients with spontaneous preterm or term labor without placental lesions consistent with maternal anti-fetal rejection.Red line represents the median.
Figure 4B:

Amniotic fluid CXCR3, CXCL9 and CXCL10 concentrations (ng/mL) in patients with spontaneous preterm or term labor without placental lesions consistent with maternal anti-fetal rejection.

Red line represents the median.

CXCR3, CXCL9 and CXCL10 concentrations in the amniotic fluid of patients at term with and without labor

Median CXCR3, CXCL9 and CXCL10 concentrations in the amniotic fluid were not significantly different between patients at term with and without labor, regardless of the presence or absence of chronic chorioamnionitis and placental lesions consistent with maternal anti-fetal rejection.

Discussion

Principal findings of the study

(1) CXCR3 is present in human amniotic fluid at the third trimester of pregnancy; (2) patients with spontaneous preterm labor who deliver preterm and who have placental lesions consistent with maternal anti-fetal rejection have higher median amniotic fluid CXCR3, CXCL9 and CXCL10 concentrations than those without these placental lesions; (3) among patients with placental lesions consistent with maternal anti-fetal rejection, those with spontaneous preterm labor and delivery have higher median CXCR3 and CXCL9 concentrations in the amniotic fluid compared to those with labor at term; (4) among patients with the absence of placental lesions consistent with maternal anti-fetal rejection, those with spontaneous preterm labor and delivery have a higher median CXCR3 concentration in the amniotic fluid than those with labor at term. These findings strongly suggest that the concentrations of the chemokine ligands and their soluble receptors reflect pathologic chronic inflammation; and (5) spontaneous labor at term is not associated with a change in the amniotic fluid concentrations of CXCR3, CXCL9 and CXCL10, indicating that physiologic labor at term does not increase the concentration of these chemokines or their receptors in the amniotic cavity.

CXCR3 functions and its ligands

The CXCR3 receptor is a transmembrane G protein-coupled receptor that belongs to the CXC chemokine family [34], [37], and its gene is located on chromosome Xq13 [78]. This receptor is expressed on several immune cells, including activated T cells [40], [41], [42], [43], dendritic cells [79], [80] and NK cells [81], but not on resting T cells, B cells, monocytes, or granulocytes [34]. Non-immune cells such as fibroblasts, endothelial, epithelial and smooth-muscle cells also express this receptor [38], [82].

CXCR3-deficient mice show a normal phenotype [32], [83], [84]; however, these mice display a diminished recruitment of mononuclear cells and CD8+ T cells in the meninges in a model of lymphocytic choriomeningitis virus infection [83]. Similar results have been observed in a model of bleomycin-induced lung injury where CXCR3-deficient mice showed a decreased recruitment of CD8+ T cells, NK cells and natural killer T (NKT) cells in the lung and liver [85]. These observations suggest that CXCR3 is required for the inflammation-induced recruitment of CD8+ T cells, NK cells and NKT cells.

CXCR3 has three splice variants: (1) CXCR3A, (2) CXCR3B and (3) CXCR3-alt [34], [38]. CXCR3A is mainly expressed on most cells, including leukocytes [33], [78], while endothelial cells express CXCR3B [63]. There are five chemokines known as CXCR3 ligands: (1) CXCL4 (platelet factor 4), (2) CXCL4L1 (platelet factor 4 variant), (3) CXCL9 (monokine induced by IFN-γ or MIG), (4) CXCL10 (IFN-γ-induced protein 10 or IP-10) and (5) CXCL11 (IFN-γ-inducible T-cell α chemoattractant or I-TAC) [34]. CXCL9, CXCL10 and CXCL11 are also recognized as IFN-γ-inducible CXCR3 ligands, as they are induced by this cytokine and others, such as tumor necrosis factor (TNF)-α [27], [29], [86], [87]. In contrast, CXCL4 and CXCL4L1 are produced by activated platelets and are not induced by IFN-γ [88], [89]. All of these chemokines bind to both CXCR3A and CXCR3B, with the exception of CXCL11, which binds to CXCR3-alt [27], [33], [63], [80], [90].

The CXCR3 receptor-ligand system plays a central role in both the chemotaxis of immune cells [34], [37], [91] and angiogenesis [34], [35], [36], [37], [38], [39]. CXCR3A activation induces chemotactic and proliferative responses [38]; whereas, CXCR3B activation mediates anti-proliferative and angiostatic effects on endothelial cells [38]. The binding of IFN-γ-inducible CXCR3 ligands results in the regulation of T-cell trafficking [26], increased production of Th1 cytokines and diminished synthesis of Th2 cytokines [37], [41], [43], [92]. However, the binding of platelet-derived CXCR3 ligands induces angiostasis [34], [93]. Due to the pleiotropic nature of CXCR3, the involvement of this receptor has been described in inflammatory, autoimmune and angiogenesis-related disorders such as inflammatory arthritis [94], [95], [96], [97], systemic sclerosis [97], [98], [99], [100], type I diabetes [97], [101], [102], transplant rejection [44], [45], [46], [47], [48], [49], [50], [51], [52], [53], [54], [55], [56], [57], [58], [59], [60], [61], [62] and cancer [36], [38], [103], [104], [105].

The role of CXCR3 in transplant rejection

There is compelling evidence that CXCR3 and its ligands (CXCL9, CXCL10 and CXCL4) are implicated in the pathogenesis of transplant rejection [34], [36], [44], [45], [46], [47], [48], [49], [50], [51], [52], [53], [54], [55], [56], [57], [58], [59], [60], [61], [62], [106]. For example, an increase in the concentration of CXCR3 ligands and the infiltration of CXCR3+ T cells has been demonstrated in biopsies from rejected solid organ transplants, including lungs [44], kidneys [107], skin [54] and endomyocardial tissue [46]. Further, the significance of CXCR3 ligands (CXCL9 and CXCL10) and the recruitment of CXCR3+ T cells in transplant rejection has been shown in several in vivo models of allograft rejection, such as the lungs [49], heart [32] and small intestine [108]. Given the chemotactic roles of CXCR3 on T cells, the blockage of CXCR3 can reduce an inflammatory response and this could be of benefit in controlling allograft rejection. For example, in a model of cardiac transplantation, CXCR3-deficient mice showed an increased tolerance to the development of acute allograft rejection [32]. These findings are consistent with previous reports demonstrating that CXCR3 blockage can delay rejection time and inhibit acute and chronic allograft rejection in murine models of cardiac and pancreatic islet transplants [109], [110], [111]. In humans, during acute renal allograft rejection, the expression of CXCR3 on peripheral CD4+ T cells increases after 3 days and remains elevated for 2 weeks [112]. In addition, the mRNA abundance of CXCR3 and CXCL10 in the urinary cells is higher in patients with acute renal allograft rejection than in healthy individuals [113]. Therefore, it has been suggested that the expression of CXCR3 and CXCL10 could be used as a urinary biomarker to predict acute renal allograft rejection [113]. Collectively, CXCR3 and its ligands play a role in the development of transplant rejection. For a more comprehensive review on the role of CXCR3 ligands (CXCL9 and CXCL10) in transplant rejection, the reader is referred to specific reviews [51], [59], [106].

A role for CXCR3, CXCL9 and CXCL10 in the placental lesions consistent with maternal anti-fetal rejection

Herein, we report that the median amniotic fluid concentration of CXCR3 as well as its ligands (CXCL9 and CXCL10) are higher in patients with placental lesions consistent with maternal anti-fetal rejection (villitis of unknown etiology, chronic deciduitis and chronic chorioamnionitis) than in those without these placental lesions. Patients who undergo spontaneous preterm labor have an increase in these chemokines in the amniotic fluid compared to those with labor at term.

Chronic chorioamnionitis is characterized by the infiltration of maternal CD8+ T lymphocytes into the chorioamniotic membranes [25], [114], [115], [116], which can induce trophoblast apoptosis [11], [25]. The proposed chemotactic signals responsible for the migration of T cells from the decidua into the chorioamniotic membranes are the CXCR3 ligands, such as amniotic fluid CXCL10 and the up-regulation of CXCL9, CXCL10 and CXCL11 in the chorioamniotic membranes [13], [25], [66]. Therefore, the observations from the current study are consistent with previous studies demonstrating that these chemokines, including their receptor (CXCR3), play a role in the pathogenesis of chronic chorioamnionitis.

The placental lesions associated with maternal anti-fetal rejection are chronic chorioamnionitis [11], [13], [14], [20], [21], [22], [117], villitis of unknown etiology [19], [21], [23], [24], [118], [119] and chronic deciduitis with plasma cells [21] as evidenced by the presence of cell- and antibody-mediated immune responses. For an in-depth appraisal on this topic, please refer to the review by Kim et al. [25].

A role for CXCR3, CXCL9 and CXCL10 in preterm labor

CXCR3 is expressed by the placenta (villous cytotrophoblasts and syncytiotrophoblasts) [63], [64] and fetal membranes [64], [65]. This receptor has been proposed to play a role in preterm [64] and term [65] labor processes. Specifically, CXCR3A protein expression is high in spontaneous preterm labor; whereas, CXCR3B protein expression is up-regulated in spontaneous term delivery [64]. In addition, CXCR3 mRNA abundance in the choriodecidual leukocytes is increased in spontaneous term labor [65].

It has been reported that the activation of CXCR3 signaling may impair maternal-fetal tolerance and predispose to spontaneous preterm labor [64] and Listeria monocytogenes-induced fetal death [120]. Also, there is an association between a fetal CXCR3 polymorphism (rs2280964) and spontaneous preterm birth [odds ratio: 0.52; 95% confidence interval (CI): 0.32–0.86] [64]. This specific polymorphism is associated with increased CXCL9 concentrations in the umbilical cord blood of newborns who were delivered preterm [64]. In an intra-peritoneal lipopolysaccharide model of spontaneous preterm birth, CXCR3-deficient mice have shown a decrease in interleukin (IL)-6 and CCL2 (also known as MCP-1) concentrations in the amniotic fluid compared to wild-type mice [64]. In addition, mice that receive a CXCR3 blocking agent or are CXCR3-deficient were protected against Listeria monocytogenes-induced fetal death [120]. This effect is potentially mediated by a reduction in the expression of CXCL9 by innate immune cells and/or a diminished influx of fetal-specific CD8+ T cells into the maternal-fetal interface [120], [121]. These observations suggest that the CXCL9/CXCR3 pathway regulates the infiltration of maternal immune cells (fetal-specific cytotoxic T cells) into the maternal-fetal interface, which can participate in maternal-fetal tolerance during pregnancy. A disruption in this pathway may be implicated in the mechanisms that lead to fetal death and premature labor.

A role for T cells in pregnancy

During pregnancy, maternal T cells recognize fetal antigens through interactions with antigen-presenting cells [122], [123]. Fetal antigen-specific Tregs (regulatory T cells) maintain maternal-fetal tolerance throughout pregnancy [3], [4], [124]. In contrast, effector T cells infiltrate into the maternal-fetal interface and are implicated in the processes of term [65], [125], [126], [127] and preterm [17], [25], [128], [129], [130] parturition.

Recently, we provided evidence that effector CD4+ T cells are involved in the process of parturition at term [65], and that activation of T cells by injecting a monoclonal antibody against the CD3 complex can induce preterm labor and birth [130]. Effector T cells are preferentially recruited into the zone rupture of the fetal membranes in spontaneous labor at term, a process mediated by CXCL10 and CCL5 [65], [126], [131]. Specifically, we have provided evidence that decidual CD4+ T cells are more abundant in spontaneous labor at term than in preterm and term gestations without labor [65]. These T cells express CD45RO, but not CD45RA [65], which suggests that they are memory T cells generated early in pregnancy when fetal-antigen presentation occurs [4], [122], [132]. The fact that decidual CD4+ T cells express IL-1β, TNF-α and MMP-9 (i.e. labor mediators [133], [134], [135], [136], [137], [138], [139], [140], [141], [142]) during spontaneous labor at term [65], as well as activation markers such as CD25 [143], suggests that the adaptive limb of the immune system participates in the process of parturition.

Another effector CD4+ T-cell subset that infiltrates the human decidua is the Th17 cells [144]. The tissue density of Th17 cells is higher in cases with acute chorioamnionitis than in cases without this placental lesion [145]. This finding further supports our hypothesis that pro-inflammatory effector T cells at the maternal-fetal interface are implicated in the pathophysiology of chronic chorioamnionitis, a lesion associated with preterm labor/birth [25].

Cytotoxic T cells (CD8+ T cells or CTLs) are present at the maternal-fetal interface in term gestations in the absence of labor, where they express perforin and granzyme B [146], [147], [148]. In the placenta, CTLs are abundant in cases with villitis of unknown etiology and express T-cell chemokine receptors (CXCR3 and CCR5) [19]. In the peripheral circulation, CD300a+ CTLs have an effector-memory phenotype, and their proportions are higher in women with chronic chorioamnionitis than in women without this placental lesion [117]. Taken together, these data suggest that CTLs may participate in the chronic pathological inflammatory response associated with term and preterm labor.

Conclusion

CXCR3 is detectable in the amniotic fluid and elevated amniotic fluid CXCR3, CXCL9 and CXCL10 concentrations are associated with the presence of placental lesions consistent with maternal anti-fetal rejection. These findings indicate that CXCR3 concentrations in the amniotic fluid may serve as a potential marker of maternal anti-fetal rejection in a subset of patients with spontaneous preterm delivery.


Corresponding authors: Roberto Romero, MD, D.Med.Sci. Perinatology Research Branch, NICHD/NIH/DHHS, Hutzel Women’s Hospital, 3990 John R, Box 4, Detroit, MI 48201, USA, Tel.: +(313) 993-2700, Fax: +(313) 993-2694; and Offer Erez, MD, Perinatology Research Branch, NICHD/NIH/DHHS, Wayne State University/Hutzel Women’s Hospital, 3990 John R, Box 4, Detroit, MI 48201, USA, Tel.: +(313) 993-2700, Fax: +(313) 993-2694

Acknowledgment

This research was supported, in part, by the Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services (NICHD/NIH/DHHS); and, in part, with Federal funds from NICHD/NIH/DHHS under Contract No. HHSN275201300006C.

  1. Author’s statement

  2. Conflict of interest: Authors state no conflicts of interest.

  3. Material and Methods: Informed consent: Informed consent was obtained from all individuals included in this study.

  4. 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.

References

[1] 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

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

[3] 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 PubMed

[4] 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

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

[6] 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

[7] 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

[8] 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

[9] 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

[10] 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 PubMed PubMed Central

[11] 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

[12] Lannaman K, Romero R, Chaemsaithong P, Ahmed AI, Yeo L, Hassan S, et al. Abstract No.497 Fetal death: an extreme form of maternal anti-fetal rejection. Am J Obstet Gynecol. 2015;212:S251.10.1016/j.ajog.2014.10.543Search in Google Scholar

[13] 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

[14] 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

[15] 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

[16] Stout MJ, Cao B, Landeau M, French J, Macones GA, Mysorekar IU. Increased human leukocyte antigen-G expression at the maternal-fetal interface is associated with preterm birth. J Matern Fetal Neonatal Med. 2015;28:454–9.10.3109/14767058.2014.921152Search in Google Scholar PubMed PubMed Central

[17] 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 PubMed PubMed Central

[18] Areia AL, Vale-Pereira S, Vaz-Ambrosio A, Alves V, Rodrigues-Santos P, Rosa MS, et al. Does progesterone administration in preterm labor influence Treg cells? J Prenat Med. 2016;44:605–11.10.1515/jpm-2015-0134Search in Google Scholar PubMed

[19] 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

[20] 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

[21] 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

[22] 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

[23] Lee KA, Kim YW, Shim JY, Won HS, Lee PR, Kim A, et al. Distinct patterns of C4d immunoreactivity in placentas with villitis of unknown etiology, cytomegaloviral placentitis, and infarct. Placenta. 2013;34:432–5.10.1016/j.placenta.2013.02.003Search in Google Scholar

[24] Rudzinski E, Gilroy M, Newbill C, Morgan T. Positive C4d immunostaining of placental villous syncytiotrophoblasts supports host-versus-graft rejection in villitis of unknown etiology. Pediatr Dev Pathol. 2013;16:7–13.10.2350/12-05-1195-OA.1Search in Google Scholar

[25] 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

[26] Groom JR, Luster AD. CXCR3 ligands: redundant, collaborative and antagonistic functions. Immunol Cell Biol. 2011; 89:207–15.10.1038/icb.2010.158Search in Google Scholar

[27] Cole KE, Strick CA, Paradis TJ, Ogborne KT, Loetscher M, Gladue RP, et al. Interferon-inducible T cell alpha chemoattractant (I-TAC): a novel non-ELR CXC chemokine with potent activity on activated T cells through selective high affinity binding to CXCR3. J Exp Med. 1998;187:2009–21.10.1084/jem.187.12.2009Search in Google Scholar

[28] Loos T, Dekeyzer L, Struyf S, Schutyser E, Gijsbers K, Gouwy M, et al. TLR ligands and cytokines induce CXCR3 ligands in endothelial cells: enhanced CXCL9 in autoimmune arthritis. Lab Invest. 2006;86:902–16.10.1038/labinvest.3700453Search in Google Scholar

[29] Proost P, Vynckier AK, Mahieu F, Put W, Grillet B, Struyf S, et al. Microbial Toll-like receptor ligands differentially regulate CXCL10/IP-10 expression in fibroblasts and mononuclear leukocytes in synergy with IFN-gamma and provide a mechanism for enhanced synovial chemokine levels in septic arthritis. Eur J Immunol. 2003;33:3146–53.10.1002/eji.200324136Search in Google Scholar

[30] Lu B, Humbles A, Bota D, Gerard C, Moser B, Soler D, et al. Structure and function of the murine chemokine receptor CXCR3. Eur J Immunol. 1999;29:3804–12.10.1002/(SICI)1521-4141(199911)29:11<3804::AID-IMMU3804>3.0.CO;2-9Search in Google Scholar

[31] Baker MS, Chen X, Rotramel AR, Nelson JJ, Lu B, Gerard C, et al. Genetic deletion of chemokine receptor CXCR3 or antibody blockade of its ligand IP-10 modulates posttransplantation graft-site lymphocytic infiltrates and prolongs functional graft survival in pancreatic islet allograft recipients. Surgery. 2003;134:126–33.10.1067/msy.2003.213Search in Google Scholar PubMed

[32] Hancock WW, Lu B, Gao W, Csizmadia V, Faia K, King JA, et al. Requirement of the chemokine receptor CXCR3 for acute allograft rejection. J Exp Med. 2000;192:1515–20.10.1084/jem.192.10.1515Search in Google Scholar PubMed PubMed Central

[33] Loetscher M, Gerber B, Loetscher P, Jones SA, Piali L, Clark-Lewis I, et al. Chemokine receptor specific for IP10 and mig: structure, function, and expression in activated T-lymphocytes. J Exp Med. 1996;184:963–9.10.1084/jem.184.3.963Search in Google Scholar PubMed PubMed Central

[34] Van Raemdonck K, Van den Steen PE, Liekens S, Van Damme J, Struyf S. CXCR3 ligands in disease and therapy. Cytokine Growth Factor Rev. 2015;26:311–27.10.1016/j.cytogfr.2014.11.009Search in Google Scholar PubMed

[35] Belperio JA, Keane MP, Arenberg DA, Addison CL, Ehlert JE, Burdick MD, et al. CXC chemokines in angiogenesis. J Leukoc Biol. 2000;68:1–8.10.1189/jlb.68.1.1Search in Google Scholar

[36] Romagnani P, Lasagni L, Annunziato F, Serio M, Romagnani S. CXC chemokines: the regulatory link between inflammation and angiogenesis. Trends Immunol. 2004;25:201–9.10.1016/j.it.2004.02.006Search in Google Scholar PubMed

[37] Lacotte S, Brun S, Muller S, Dumortier H. CXCR3, inflammation, and autoimmune diseases. Ann N Y Acad Sci. 2009;1173:310–7.10.1111/j.1749-6632.2009.04813.xSearch in Google Scholar PubMed

[38] Billottet C, Quemener C, Bikfalvi A. CXCR3, a double-edged sword in tumor progression and angiogenesis. Biochim Biophys Acta. 2013;1836:287–95.10.1016/j.bbcan.2013.08.002Search in Google Scholar PubMed

[39] Bosisio D, Salvi V, Gagliostro V, Sozzani S. Angiogenic and antiangiogenic chemokines. Chem Immunol Allergy. 2014;99:89–104.10.1159/000353317Search in Google Scholar PubMed

[40] Bonecchi R, Bianchi G, Bordignon PP, D’Ambrosio D, Lang R, Borsatti A, et al. Differential expression of chemokine receptors and chemotactic responsiveness of type 1 T helper cells (Th1s) and Th2s. J Exp Med. 1998;187:129–34.10.1084/jem.187.1.129Search in Google Scholar PubMed PubMed Central

[41] Rabin RL, Park MK, Liao F, Swofford R, Stephany D, Farber JM. Chemokine receptor responses on T cells are achieved through regulation of both receptor expression and signaling. J Immunol. 1999;162:3840–50.10.4049/jimmunol.162.7.3840Search in Google Scholar

[42] Koch MA, Tucker-Heard G, Perdue NR, Killebrew JR, Urdahl KB, Campbell DJ. The transcription factor T-bet controls regulatory T cell homeostasis and function during type 1 inflammation. Nat Immunol. 2009;10:595–602.10.1038/ni.1731Search in Google Scholar PubMed PubMed Central

[43] Groom JR, Luster AD. CXCR3 in T cell function. Exp Cell Res. 2011;317:620–31.10.1016/j.yexcr.2010.12.017Search in Google Scholar

[44] Agostini C, Calabrese F, Rea F, Facco M, Tosoni A, Loy M, et al. Cxcr3 and its ligand CXCL10 are expressed by inflammatory cells infiltrating lung allografts and mediate chemotaxis of T cells at sites of rejection. Am J Path. 2001;158:1703–11.10.1016/S0002-9440(10)64126-0Search in Google Scholar

[45] Miura M, Morita K, Kobayashi H, Hamilton TA, Burdick MD, Strieter RM, et al. Monokine induced by IFN-gamma is a dominant factor directing T cells into murine cardiac allografts during acute rejection. J Immunol. 2001;167:3494–504.10.4049/jimmunol.167.6.3494Search in Google Scholar

[46] Melter M, Exeni A, Reinders ME, Fang JC, McMahon G, Ganz P, et al. Expression of the chemokine receptor CXCR3 and its ligand IP-10 during human cardiac allograft rejection. Circulation. 2001;104:2558–64.10.1161/hc4601.098010Search in Google Scholar

[47] el-Sawy T, Fahmy NM, Fairchild RL. Chemokines: directing leukocyte infiltration into allografts. Curr Opin Immunol. 2002;14:562–8.10.1016/S0952-7915(02)00382-5Search in Google Scholar

[48] Zhao DX, Hu Y, Miller GG, Luster AD, Mitchell RN, Libby P. Differential expression of the IFN-gamma-inducible CXCR3-binding chemokines, IFN-inducible protein 10, monokine induced by IFN, and IFN-inducible T cell alpha chemoattractant in human cardiac allografts: association with cardiac allograft vasculopathy and acute rejection. J Immunol. 2002;169:1556–60.10.4049/jimmunol.169.3.1556Search in Google Scholar PubMed

[49] Belperio JA, Keane MP, Burdick MD, Lynch JP 3rd, Zisman DA, Xue YY, et al. Role of CXCL9/CXCR3 chemokine biology during pathogenesis of acute lung allograft rejection. J Immunol. 2003;171:4844–52.10.4049/jimmunol.171.9.4844Search in Google Scholar PubMed

[50] Hu H, Aizenstein BD, Puchalski A, Burmania JA, Hamawy MM, Knechtle SJ. Elevation of CXCR3-binding chemokines in urine indicates acute renal-allograft dysfunction. Am J Transplant. 2004;4:432–7.10.1111/j.1600-6143.2004.00354.xSearch in Google Scholar PubMed

[51] Tan J, Zhou G. Chemokine receptors and transplantation. Cell Mol Immunol. 2005;2:343–9.Search in Google Scholar

[52] Lazzeri E, Rotondi M, Mazzinghi B, Lasagni L, Buonamano A, Rosati A, et al. High CXCL10 expression in rejected kidneys and predictive role of pretransplant serum CXCL10 for acute rejection and chronic allograft nephropathy. Transplantation. 2005;79:1215–20.10.1097/01.TP.0000160759.85080.2ESearch in Google Scholar

[53] Romagnani P. From basic science to clinical practice: use of cytokines and chemokines as therapeutic targets in renal diseases. J Nephrol. 2005;18:229–33.Search in Google Scholar

[54] Piper KP, Horlock C, Curnow SJ, Arrazi J, Nicholls S, Mahendra P, et al. CXCL10-CXCR3 interactions play an important role in the pathogenesis of acute graft-versus-host disease in the skin following allogeneic stem-cell transplantation. Blood. 2007;110:3827–32.10.1182/blood-2006-12-061408Search in Google Scholar PubMed

[55] Crescioli C, Buonamano A, Scolletta S, Sottili M, Francalanci M, Giomarelli P, et al. Predictive role of pretransplant serum CXCL10 for cardiac acute rejection. Transplantation. 2009;87:249–55.10.1097/TP.0b013e3181919f5dSearch in Google Scholar PubMed

[56] Rotondi M, Netti GS, Lazzeri E, Stallone G, Bertoni E, Chiovato L, et al. High pretransplant serum levels of CXCL9 are associated with increased risk of acute rejection and graft failure in kidney graft recipients. Transpl Int. 2010;23:465–75.10.1111/j.1432-2277.2009.01006.xSearch in Google Scholar PubMed

[57] Heidt S, San Segundo D, Shankar S, Mittal S, Muthusamy AS, Friend PJ, et al. Peripheral blood sampling for the detection of allograft rejection: biomarker identification and validation. Transplantation. 2011;92:1–9.10.1097/TP.0b013e318218e978Search in Google Scholar PubMed

[58] Lo DJ, Weaver TA, Kleiner DE, Mannon RB, Jacobson LM, Becker BN, et al. Chemokines and their receptors in human renal allotransplantation. Transplantation. 2011;91:70–7.10.1097/TP.0b013e3181fe12fcSearch in Google Scholar PubMed PubMed Central

[59] Romagnani P, Crescioli C. CXCL10: a candidate biomarker in transplantation. Clin Chim Acta. 2012;413:1364–73.10.1016/j.cca.2012.02.009Search in Google Scholar PubMed

[60] Asaoka T, Marubashi S, Kobayashi S, Hama N, Eguchi H, Takeda Y, et al. Intragraft transcriptome level of CXCL9 as biomarker of acute cellular rejection after liver transplantation. J Surg Res. 2012;178:1003–14.10.1016/j.jss.2012.07.016Search in Google Scholar PubMed

[61] Zhang Q, Liu YF, Su ZX, Shi LP, Chen YH. Serum fractalkine and interferon-gamma inducible protein-10 concentrations are early detection markers for acute renal allograft rejection. Transplantation Proc. 2014;46:1420–5.10.1016/j.transproceed.2014.02.019Search in Google Scholar PubMed

[62] Croudace JE, Inman CF, Abbotts BE, Nagra S, Nunnick J, Mahendra P, et al. Chemokine-mediated tissue recruitment of CXCR3+ CD4+ T cells plays a major role in the pathogenesis of chronic GVHD. Blood. 2012;120:4246–55.10.1182/blood-2012-02-413260Search in Google Scholar PubMed

[63] Lasagni L, Francalanci M, Annunziato F, Lazzeri E, Giannini S, Cosmi L, et al. An alternatively spliced variant of CXCR3 mediates the inhibition of endothelial cell growth induced by IP-10, Mig, and I-TAC, and acts as functional receptor for platelet factor 4. J Exp Med. 2003;197:1537–49.10.1084/jem.20021897Search in Google Scholar PubMed PubMed Central

[64] Karjalainen MK, Ojaniemi M, Haapalainen AM, Mahlman M, Salminen A, Huusko JM, et al. CXCR3 polymorphism and expression associate with spontaneous preterm birth. J Immunol. 2015;195:2187–98.10.4049/jimmunol.1501174Search in Google Scholar PubMed

[65] 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

[66] 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

[67] 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 Prenat Med. 2012;40:329–43.10.1515/jpm-2012-0034Search in Google Scholar

[68] Lockwood CJ. Risk factors for preterm birth and new approaches to its early diagnosis. J Prenat Med. 2015;43:499–501.10.1515/jpm-2015-0261Search in Google Scholar

[69] Andersen HF, Nugent CE, Wanty SD, Hayashi RH. Prediction of risk for preterm delivery by ultrasonographic measurement of cervical length. Am J Obstet Gynecol. 1990;163:859–67.10.1016/0002-9378(90)91084-PSearch in Google Scholar

[70] Iams JD, Goldenberg RL, Meis PJ, Mercer BM, Moawad A, Das A, et al. The length of the cervix and the risk of spontaneous premature delivery. N Engl J Med. 1996;334:567–72.10.1056/NEJM199602293340904Search in Google Scholar PubMed

[71] 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 PubMed PubMed Central

[72] 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

[73] Redline RW, Faye-Petersen O, Heller D, Qureshi F, Savell V, Vogler C. 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

[74] 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 PubMed PubMed Central

[75] 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

[76] 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

[77] R Core Team. R: A language and environment for statistical computing. R Foundation for Statistical Computing, Vienna, Austria; 2016. URL https://www.R–project.org.Search in Google Scholar

[78] Loetscher M, Loetscher P, Brass N, Meese E, Moser B. Lymphocyte-specific chemokine receptor CXCR3: regulation, chemokine binding and gene localization. Eur J Immunol. 1998;28:3696–705.10.1002/(SICI)1521-4141(199811)28:11<3696::AID-IMMU3696>3.0.CO;2-WSearch in Google Scholar

[79] Garcia-Lopez MA, Sanchez-Madrid F, Rodriguez-Frade JM, Mellado M, Acevedo A, Garcia MI, et al. CXCR3 chemokine receptor distribution in normal and inflamed tissues: expression on activated lymphocytes, endothelial cells, and dendritic cells. Lab Invest. 2001;81:409–18.10.1038/labinvest.3780248Search in Google Scholar

[80] Struyf S, Salogni L, Burdick MD, Vandercappellen J, Gouwy M, Noppen S, et al. Angiostatic and chemotactic activities of the CXC chemokine CXCL4L1 (platelet factor-4 variant) are mediated by CXCR3. Blood. 2011;117:480–8.10.1182/blood-2009-11-253591Search in Google Scholar

[81] Lima M, Leander M, Santos M, Santos AH, Lau C, Queiros ML, et al. Chemokine receptor expression on normal blood CD56(+) NK-cells elucidates cell partners that comigrate during the innate and adaptive immune responses and identifies a transitional NK-cell population. J Immunol Res. 2015;2015:839684.10.1155/2015/839684Search in Google Scholar

[82] Strieter RM. Chemokines: not just leukocyte chemoattractants in the promotion of cancer. Nat Immunol. 2001;2:285–6.10.1038/86286Search in Google Scholar

[83] Christensen JE, Nansen A, Moos T, Lu B, Gerard C, Christensen JP, et al. Efficient T-cell surveillance of the CNS requires expression of the CXC chemokine receptor 3. J Neurosci. 2004;24:4849–58.10.1523/JNEUROSCI.0123-04.2004Search in Google Scholar

[84] Yates CC, Whaley D, Kulasekeran P, Hancock WW, Lu B, Bodnar R, et al. Delayed and deficient dermal maturation in mice lacking the CXCR3 ELR-negative CXC chemokine receptor. Am J Pathol. 2007;171:484–95.10.2353/ajpath.2007.061092Search in Google Scholar

[85] Jiang D, Liang J, Hodge J, Lu B, Zhu Z, Yu S, et al. Regulation of pulmonary fibrosis by chemokine receptor CXCR3. J Clin Invest. 2004;114:291–9.10.1172/JCI16861Search in Google Scholar

[86] Luster AD, Ravetch JV. Biochemical characterization of a gamma interferon-inducible cytokine (IP-10). J Exp Med. 1987;166:1084–97.10.1084/jem.166.4.1084Search in Google Scholar PubMed PubMed Central

[87] Farber JM. A macrophage mRNA selectively induced by gamma-interferon encodes a member of the platelet factor 4 family of cytokines. Proc Natl Acad Sci USA. 1990;87:5238–42.10.1073/pnas.87.14.5238Search in Google Scholar PubMed PubMed Central

[88] Struyf S, Burdick MD, Proost P, Van Damme J, Strieter RM. Platelets release CXCL4L1, a nonallelic variant of the chemokine platelet factor-4/CXCL4 and potent inhibitor of angiogenesis. Circ Res. 2004;95:855–7.10.1161/01.RES.0000146674.38319.07Search in Google Scholar PubMed

[89] Lasagni L, Grepin R, Mazzinghi B, Lazzeri E, Meini C, Sagrinati C, et al. PF-4/CXCL4 and CXCL4L1 exhibit distinct subcellular localization and a differentially regulated mechanism of secretion. Blood. 2007;109:4127–34.10.1182/blood-2006-10-052035Search in Google Scholar PubMed

[90] Ehlert JE, Addison CA, Burdick MD, Kunkel SL, Strieter RM. Identification and partial characterization of a variant of human CXCR3 generated by posttranscriptional exon skipping. J Immunol. 2004;173:6234–40.10.4049/jimmunol.173.10.6234Search in Google Scholar PubMed

[91] Trentin L, Agostini C, Facco M, Piazza F, Perin A, Siviero M, et al. The chemokine receptor CXCR3 is expressed on malignant B cells and mediates chemotaxis. J Clin Invest. 1999;104:115–21.10.1172/JCI7335Search in Google Scholar PubMed PubMed Central

[92] Gangur V, Simons FE, Hayglass KT. Human IP-10 selectively promotes dominance of polyclonally activated and environmental antigen-driven IFN-gamma over IL-4 responses. FASEB J. 1998;12:705–13.10.1096/fasebj.12.9.705Search in Google Scholar PubMed

[93] Gupta SK, Hassel T, Singh JP. A potent inhibitor of endothelial cell proliferation is generated by proteolytic cleavage of the chemokine platelet factor 4. Proc Natl Acad Sci USA. 1995;92:7799–803.10.1073/pnas.92.17.7799Search in Google Scholar PubMed PubMed Central

[94] Patel DD, Zachariah JP, Whichard LP. CXCR3 and CCR5 ligands in rheumatoid arthritis synovium. Clin Immunol. 2001;98:39–45.10.1006/clim.2000.4957Search in Google Scholar PubMed

[95] Ruschpler P, Lorenz P, Eichler W, Koczan D, Hanel C, Scholz R, et al. High CXCR3 expression in synovial mast cells associated with CXCL9 and CXCL10 expression in inflammatory synovial tissues of patients with rheumatoid arthritis. Arthritis Res Ther. 2003;5:R241–52.10.1186/ar783Search in Google Scholar PubMed PubMed Central

[96] Pharoah DS, Varsani H, Tatham RW, Newton KR, de Jager W, Prakken BJ, et al. Expression of the inflammatory chemokines CCL5, CCL3 and CXCL10 in juvenile idiopathic arthritis, and demonstration of CCL5 production by an atypical subset of CD8+ T cells. Arthritis Res Ther. 2006;8:R50.10.1186/ar1913Search in Google Scholar PubMed PubMed Central

[97] Antonelli A, Ferrari SM, Giuggioli D, Ferrannini E, Ferri C, Fallahi P. Chemokine (C-X-C motif) ligand (CXCL)10 in autoimmune diseases. Autoimmun Rev. 2014;13:272–80.10.1016/j.autrev.2013.10.010Search in Google Scholar PubMed

[98] Antonelli A, Ferri C, Fallahi P, Ferrari SM, Giuggioli D, Colaci M, et al. CXCL10 (alpha) and CCL2 (beta) chemokines in systemic sclerosis–a longitudinal study. Rheumatology. 2008;47:45–9.10.1093/rheumatology/kem313Search in Google Scholar PubMed

[99] Rabquer BJ, Tsou PS, Hou Y, Thirunavukkarasu E, Haines GK 3rd, Impens AJ, et al. Dysregulated expression of MIG/CXCL9, IP-10/CXCL10 and CXCL16 and their receptors in systemic sclerosis. Arthritis Res Ther. 2011;13:R18.10.1186/ar3242Search in Google Scholar PubMed PubMed Central

[100] van Bon L, Affandi AJ, Broen J, Christmann RB, Marijnissen RJ, Stawski L, et al. Proteome-wide analysis and CXCL4 as a biomarker in systemic sclerosis. N Engl J Med. 2014; 370:433–43.10.1056/NEJMoa1114576Search in Google Scholar PubMed PubMed Central

[101] Shimada A, Morimoto J, Kodama K, Suzuki R, Oikawa Y, Funae O, et al. Elevated serum IP-10 levels observed in type 1 diabetes. Diabetes Care. 2001;24:510–5.10.2337/diacare.24.3.510Search in Google Scholar PubMed

[102] Antonelli A, Ferrari SM, Corrado A, Ferrannini E, Fallahi P. CXCR3, CXCL10 and type 1 diabetes. Cytokine Growth Factor Rev. 2014;25:57–65.10.1016/j.cytogfr.2014.01.006Search in Google Scholar PubMed

[103] Vandercappellen J, Van Damme J, Struyf S. The role of CXC chemokines and their receptors in cancer. Cancer Lett. 2008;267:226–44.10.1016/j.canlet.2008.04.050Search in Google Scholar PubMed

[104] Vandercappellen J, Van Damme J, Struyf S. The role of the CXC chemokines platelet factor-4 (CXCL4/PF-4) and its variant (CXCL4L1/PF-4var) in inflammation, angiogenesis and cancer. Cytokine Growth Factor Rev. 2011;22:1–18.10.1016/j.cytogfr.2010.10.011Search in Google Scholar PubMed

[105] Ma B, Khazali A, Wells A. CXCR3 in carcinoma progression. Histol Histopathol. 2015;30:781–92.Search in Google Scholar

[106] Crescioli C. Chemokines and transplant outcome. Clin Biochem. 2016;49:355–62.10.1016/j.clinbiochem.2015.07.026Search in Google Scholar PubMed

[107] Panzer U, Reinking RR, Steinmetz OM, Zahner G, Sudbeck U, Fehr S, et al. CXCR3 and CCR5 positive T-cell recruitment in acute human renal allograft rejection. Transplantation. 2004;78:1341–50.10.1097/01.TP.0000140483.59664.64Search in Google Scholar

[108] Zhang Z, Kaptanoglu L, Tang Y, Ivancic D, Rao SM, Luster A, et al. IP-10-induced recruitment of CXCR3 host T cells is required for small bowel allograft rejection. Gastroenterology. 2004;126:809–18.10.1053/j.gastro.2003.12.014Search in Google Scholar

[109] Akashi S, Sho M, Kashizuka H, Hamada K, Ikeda N, Kuzumoto Y, et al. A novel small-molecule compound targeting CCR5 and CXCR3 prevents acute and chronic allograft rejection. Transplantation. 2005;80:378–84.10.1097/01.tp.0000166338.99933.e1Search in Google Scholar

[110] Schnickel GT, Hsieh GR, Garcia C, Shefizadeh A, Fishbein MC, Ardehali A. Role of CXCR3 and CCR5 in allograft rejection. Transplant Proc. 2006;38:3221–4.10.1016/j.transproceed.2006.10.164Search in Google Scholar

[111] Rosenblum JM, Zhang QW, Siu G, Collins TL, Sullivan T, Dairaghi DJ, et al. CXCR3 antagonism impairs the development of donor-reactive, IFN-gamma-producing effectors and prolongs allograft survival. Transplantation. 2009;87:360–9.10.1097/TP.0b013e31819574e9Search in Google Scholar

[112] Inston N, Drayson M, Ready A, Cockwell P. Serial changes in the expression of CXCR3 and CCR5 on peripheral blood lymphocytes following human renal transplantation. Exp Clin Transplant. 2007;5:638–42.Search in Google Scholar

[113] Tatapudi RR, Muthukumar T, Dadhania D, Ding R, Li B, Sharma VK, et al. Noninvasive detection of renal allograft inflammation by measurements of mRNA for IP-10 and CXCR3 in urine. Kidney Int. 2004;65:2390–7.10.1111/j.1523-1755.2004.00663.xSearch in Google Scholar

[114] Gersell DJ, Phillips NJ, Beckerman K. Chronic chorioamnionitis: a clinicopathologic study of 17 cases. Int J Gynecol Pathol. 1991;10:217–29.10.1097/00004347-199107000-00001Search in Google Scholar

[115] Gersell DJ. Chronic villitis, chronic chorioamnionitis, and maternal floor infarction. Semin Diagn Pathol. 1993;10: 251–66.Search in Google Scholar

[116] 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

[117] Xu Y, Tarquini F, Romero R, Kim CJ, Tarca AL, Bhatti G, et al. Peripheral CD300a+CD8+ T lymphocytes with a distinct cytotoxic molecular signature increase in pregnant women with chronic chorioamnionitis. Am J Reprod Immunol. 2012;67:184–97.10.1111/j.1600-0897.2011.01088.xSearch in Google Scholar PubMed PubMed Central

[118] Katzman PJ, Murphy SP, Oble DA. Immunohistochemical analysis reveals an influx of regulatory T cells and focal trophoblastic STAT-1 phosphorylation in chronic villitis of unknown etiology. Pediatr Dev Pathol. 2011;14:284–93.10.2350/10-09-0910-OA.1Search in Google Scholar PubMed

[119] Ito Y, Matsuoka K, Uesato T, Sago H, Okamoto A, Nakazawa A, et al. Increased expression of perforin, granzyme B, and C5b-9 in villitis of unknown etiology. Placenta. 2015;36:531–7.10.1016/j.placenta.2015.02.004Search in Google Scholar PubMed

[120] 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

[121] Bakardjiev A. Immunology: Stillbirth prevented by signal blockade. Nature. 2015;520:627–8.10.1038/520627aSearch in Google Scholar PubMed

[122] Erlebacher A, Vencato D, Price KA, Zhang D, Glimcher LH. Constraints in antigen presentation severely restrict T cell recognition of the allogeneic fetus. J Clin Invest. 2007;117: 1399–411.10.1172/JCI28214Search in Google Scholar PubMed PubMed Central

[123] Bizargity P, Bonney EA. Dendritic cells: a family portrait at mid-gestation. Immunology. 2009;126:565–78.10.1111/j.1365-2567.2008.02918.xSearch in Google Scholar PubMed PubMed Central

[124] 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

[125] 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

[126] 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;205:16.10.1016/j.ajog.2011.04.019Search in Google Scholar PubMed

[127] Gomez-Lopez N, Olson DM, Robertson SA. Interleukin-6 controls uterine Th9 cells and CD8(+) T regulatory cells to accelerate parturition in mice. Immunol Cell Biol. 2016;94:79–89.10.1038/icb.2015.63Search in Google Scholar PubMed

[128] Bizargity P, Del Rio R, Phillippe M, Teuscher C, Bonney EA. Resistance to lipopolysaccharide-induced preterm delivery mediated by regulatory T cell function in mice. Biol Reprod. 2009;80:874–81.10.1095/biolreprod.108.074294Search in Google Scholar PubMed PubMed Central

[129] St Louis D, Romero R, Plazyo O, Arenas-Hernandez M, Panaitescu B, Xu Y, et al. Invariant NKT cell activation induces late preterm birth that is attenuated by rosiglitazone. J Immunol. 2016;196:1044–59.10.4049/jimmunol.1501962Search in Google Scholar

[130] Gomez-Lopez N, Romero R, Arenas-Hernandez M, Ahn H, Panaitescu B, Vadillo-Ortega F, et al. In vivo T-cell activation by a monoclonal alphaCD3epsilon antibody induces preterm labor and birth. Am J Reprod Immunol. 2016;76:386–90.10.1111/aji.12562Search in Google Scholar

[131] Gomez-Lopez N, Hernandez-Santiago S, Lobb AP, Olson DM, Vadillo-Ortega F. Normal and premature rupture of fetal membranes at term delivery differ in regional chemotactic activity and related chemokine/cytokine production. Reprod Sci. 2013;20:276–84.10.1177/1933719112452473Search in Google Scholar

[132] Sharkey DJ, Tremellen KP, Jasper MJ, Gemzell-Danielsson K, Robertson SA. Seminal fluid induces leukocyte recruitment and cytokine and chemokine mRNA expression in the human cervix after coitus. J Immunol. 2012;188:2445–54.10.4049/jimmunol.1102736Search in Google Scholar

[133] Romero R, Brody DT, Oyarzun E, Mazor M, Wu YK, Hobbins JC, et al. Infection and labor. III. Interleukin-1: a signal for the onset of parturition. Am J Obstet Gynecol. 1989;160(5 Pt 1):1117–23.10.1016/0020-7292(90)90214-6Search in Google Scholar

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

[135] Romero R, Mazor M, Tartakovsky B. Systemic administration of interleukin-1 induces preterm parturition in mice. Am J Obstet Gynecol. 1991;165(4 Pt 1):969–71.10.1016/0002-9378(91)90450-6Search in Google Scholar

[136] 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

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

[138] Vadillo OF, Gonzalez AG, Furth EE, Lei H, Muschel RJ, Stetler-Stevenson WG, et al. 92-kd type IV collagenase (matrix metalloproteinase-9) activity in human amniochorion increases with labor. Am J Pathol. 1995;146:148–56.Search in Google Scholar

[139] Vadillo-Ortega F, Hernandez A, Gonzalez-Avila G, Bermejo L, Iwata K, Strauss JF 3rd. Increased matrix metalloproteinase activity and reduced tissue inhibitor of metalloproteinases-1 levels in amniotic fluids from pregnancies complicated by premature rupture of membranes. Am J Obstet Gynecol. 1996;174:1371–6.10.1016/S0002-9378(96)70687-7Search in Google Scholar

[140] Elliott CL, Loudon JA, Brown N, Slater DM, Bennett PR, Sullivan MH. IL-1beta and IL-8 in human fetal membranes: changes with gestational age, labor, and culture conditions. Am J Reprod Immunol. 2001;46:260–7.10.1034/j.1600-0897.2001.d01-11.xSearch in Google Scholar PubMed

[141] Xu P, Alfaidy N, Challis JR. Expression of matrix metalloproteinase (MMP)-2 and MMP-9 in human placenta and fetal membranes in relation to preterm and term labor. J Clin Endocrinol Metab. 2002;87:1353–61.10.1210/jcem.87.3.8320Search in Google Scholar PubMed

[142] 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

[143] Sindram-Trujillo AP, Scherjon SA, van Hulst-van Miert PP, Kanhai HH, Roelen DL, Claas FH. Comparison of decidual leukocytes following spontaneous vaginal delivery and elective cesarean section in uncomplicated human term pregnancy. J Reprod Immunol. 2004;62:125–37.10.1016/j.jri.2003.11.007Search in Google Scholar PubMed

[144] Nakashima A, Ito M, Yoneda S, Shiozaki A, Hidaka T, Saito S. Circulating and decidual Th17 cell levels in healthy pregnancy. Am J Reprod Immunol. 2010;63:104–9.10.1111/j.1600-0897.2009.00771.xSearch in Google Scholar PubMed

[145] Ito M, Nakashima A, Hidaka T, Okabe M, Bac ND, Ina S, et al. A role for IL-17 in induction of an inflammation at the fetomaternal interface in preterm labour. J Reprod Immunol. 2010;84:75–85.10.1016/j.jri.2009.09.005Search in Google Scholar PubMed

[146] Tilburgs T, Roelen DL, van der Mast BJ, van Schip JJ, Kleijburg C, de Groot-Swings GM, et al. Differential distribution of CD4(+)CD25(bright) and CD8(+)CD28(-) T-cells in decidua and maternal blood during human pregnancy. Placenta. 2006;27:25.10.1016/j.placenta.2005.11.008Search in Google Scholar PubMed

[147] Tilburgs T, Scherjon SA, Roelen DL, Claas FH. Decidual CD8+CD28- T cells express CD103 but not perforin. Hum Immunol. 2009;70:96–100.10.1016/j.humimm.2008.12.006Search in Google Scholar PubMed

[148] Tilburgs T, Schonkeren D, Eikmans M, Nagtzaam NM, Datema G, Swings GM, et al. Human decidual tissue contains differentiated CD8+ effector-memory T cells with unique properties. J Immunol. 2010;185:4470–7.10.4049/jimmunol.0903597Search in Google Scholar PubMed

Received: 2017-2-3
Accepted: 2017-4-19
Published Online: 2017-8-22
Published in Print: 2018-2-23

©2018 Walter de Gruyter GmbH, Berlin/Boston

Downloaded on 27.4.2024 from https://www.degruyter.com/document/doi/10.1515/jpm-2017-0042/html
Scroll to top button