Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Sep 25.
Published in final edited form as: J Perinat Med. 2020 Sep 25;48(7):665–676. doi: 10.1515/jpm-2020-0037

CELLULAR IMMUNE RESPONSES IN AMNIOTIC FLUID OF WOMEN WITH A SONOGRAPHIC SHORT CERVIX

Jose Galaz 1,2,3, Roberto Romero 1,4,5,6,7,8, Yi Xu 1,2, Derek Miller 1,2, Dustyn Levenson 1,2, Robert Para 1,2, Aneesha Varrey 1,2, Richard Hsu 9, Anna Tong 9, Sonia S Hassan 2,10, Chaur-Dong Hsu 1,2,10, Nardhy Gomez-Lopez 1,2,11
PMCID: PMC8272936  NIHMSID: NIHMS1718259  PMID: 32716907

Abstract

Background:

A sonographic short cervix is one of the strongest predictors of a preterm delivery. However, the cellular immune composition of amniotic fluid in women with a short cervix has not yet been described. Herein, we determined cellular and soluble immune responses in amniotic fluid from pregnant women with a mid-trimester asymptomatic short cervix.

Methods:

Amniotic fluid samples (n=77) were collected from asymptomatic women with a cervical length between 15–25 mm (n=36, short cervix) or ≤15 mm (n=41, severely short cervix) diagnosed by ultrasound. Flow cytometry and multiplex measurement of cytokines/chemokines were performed.

Results:

1) the cellular immune composition of amniotic fluid was not different between women with a severely short cervix (≤15 mm) and those with a short cervix 15–25 mm; 2) amniotic fluid concentrations of multiple cytokines/chemokines were higher in women with a severely short cervix (≤15 mm) than in those with a short cervix 15–25 mm; 3) the cellular immune composition of amniotic fluid was not different between women with a severely short cervix (≤15 mm) who ultimately underwent preterm delivery compared to those who delivered at term; and 4) amniotic fluid concentrations of IL-2, but not other immune mediators, were increased in women with a severely short cervix (≤15 mm) who ultimately delivered preterm compared to those who delivered at term.

Conclusion:

Women with a severely short cervix (≤15 mm) have increased concentrations of pro-inflammatory mediators in the amniotic cavity; yet, this is not translated to changes in the cellular immune response.

Keywords: Amniotic fluid, cytokines, chemokines, immune cells, intra-amniotic inflammation, preterm

INTRODUCTION

Preterm birth is one of the great obstetrical syndromes (14) and a primary cause of perinatal morbidity and mortality worldwide (5, 6). Among the known risk factors that predispose to preterm birth, a sonographic short cervix is one of the strongest predictors (2, 723). Indeed, an asymptomatic woman with a mid-trimester cervical length of ≤15 mm has over a 50% chance of undergoing spontaneous preterm birth at 32 weeks or less (13, 24). Similarly, 50% of women diagnosed with a short cervix at 16–22 weeks will deliver at 32 weeks or less (25). Therefore, the shorter the sonographic cervical length, the higher is the risk of spontaneous preterm labor and birth (2, 719, 23).

A short cervix can be considered a syndrome caused by multiple pathological processes (1, 17, 26, 27), including prior cervical surgery (2835), congenital disorders (e.g. cervical hypoplasia after diethylstilbestrol exposure) (3640), genetic syndromes (e.g. Ehlers-Danlos syndrome) (41, 42), progesterone deficiency (17, 26, 27), vaginal microbiome composition (4345) and, more importantly, intra-amniotic inflammation and/or infection (19, 24, 25, 4651) - the well-known etiology for preterm birth (1, 2, 18, 5262). Recently, we used a combination of cultivation and molecular microbiological techniques to establish the nature of the inflammatory process in the amniotic cavity of women with a sonographic short cervix (19). Most women with a short cervix do not have detectable microorganisms or intra-amniotic inflammation [elevated concentrations of interleukin-6 ≥2.6 ng/mL (63)] (19). Yet, a fraction of these women have intra-amniotic inflammation in the absence of detectable microorganisms (19), a clinical condition that is referred to as sterile intra-amniotic inflammation (19, 61, 6471). Importantly, sterile intra-amniotic inflammation is more frequently observed than proven intra-amniotic infection in women with a short cervix (19, 25). Hence, the clinical condition of a short cervix can occur in the presence or absence of intra-amniotic inflammation/infection and, therefore, may involve diverse immune responses in the amniotic cavity.

The cellular immune responses in the amniotic cavity have been recently characterized during normal pregnancy (72) and in complications such as clinical chorioamnionitis at term (73), preterm labor with intact membranes (74), preterm clinical chorioamnionitis (75), and preterm prelabor rupture of membranes (PPROM) (76). However, the cellular immune composition of the amniotic fluid in women with a short cervix has not yet been described. Increased amniotic fluid concentrations of several cytokines and other soluble factors (e.g. complement) have been linked to a short cervix and/or cervical insufficiency (25, 49, 51, 77, 78). Indeed, the cytokine network of women with a short cervix who delivered preterm includes elevated concentrations of pro-inflammatory cytokines such as MIP-1α, MCP-1, and IL-6 (25). Hence, in the current study we determined both the cellular immune composition and cytokine/chemokine concentrations in amniotic fluid from women with a mid-trimester asymptomatic short cervix.

METHODS

Study design and population:

This cross-sectional study included patients who underwent amniocentesis due to clinical indications and amniotic fluid samples were immediately processed for immunophenotyping by flow cytometry. The collection of samples was approved by the Institutional Review Boards of the Detroit Medical Center, Wayne State University, and the Perinatology Research Branch, an intramural program of the Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (Detroit, MI, USA). All women provided written informed consent prior to the collection of amniotic fluid.

This study included 77 amniotic fluid samples collected from asymptomatic women with a short cervix diagnosed by ultrasound (see clinical definitions and amniotic fluid sample collection below). Women were divided into two groups: those with a cervical length 15–25 mm (n=36) (hereafter referred to as a short cervix) and those with ≤15 mm (n=41) (hereafter referred to as a severely short cervix) (Table 1). For all patients, the amniocentesis was performed at the time of diagnosis. The demographic and clinical characteristics of the study population are shown in Table 1. Patients with multiple pregnancies, preterm prelabor rupture of membranes (pPROM) or spontaneous preterm labor occurring prior to diagnosis with a short cervix, fetal malformations, or genetic disorders were excluded from this study. In addition, women with positive amniotic fluid bacterial cultures were also excluded.

Table 1.

Clinical and demographic characteristics of women with an asymptomatic short cervix.

>15 mm (n=36) ≤15 mm (n=41) p-value
Maternal age (years; median [IQR])a 25 (21.8–30) 26 (22–31) 0.4
Body mass index (kg/m2; median [IQR])a 27 (23.3–31.8)c 31.8 (27–34.9) 0.09
Primiparityb 11.1% (4/36) 9.8% (4/41) 1.0
Race/ethnicityb 0.4
 African-American 88.9% (32/36) 82.9% (34/41)
 White 2.8% (1/36) 9.8% (4/41)
 Other 8.3% (3/36) 7.3% (3/41)
Gestational age at amniocentesis (weeks; median [IQR])a 20.9 (20–24.3) 22.1 (20.7–23.1) 0.5
Sonographic cervical length at diagnosis (millimeters; median [IQR])a 21 (18–22.3) 6 (0–12) <0.001
IL-6 (ng/mL; median [IQR])a 0.3 (0.2–0.7) 0.7 (0.2–3.5) 0.02
Amniotic fluid glucose (mg/dl; median [IQR])a 29 (24–37)c 35.5 (24.3–41)c 0.3
Amniotic fluid WBC (cells/mm3; median [IQR])a 1 (0–4)e 2 (0–5.5)d 0.8
Intra-amniotic inflammationb 2.8% (1/36) 29.3% (12/41) 0.002
Treatmentb
 Cerclage 11.1% (4/36) 14.6% (6/41) 0.7
 Progesterone 27.8% (10/36) 29.3% (12/41) 1.0
 Cerclage and progesterone 41.7% (15/36) 34.1% (14/41) 0.6
 No treatment 19.4% (7/36) 22% (9/41) 1.0
Gestational age at delivery (weeks; median [IQR])a 38.4 (36.1–39.4) 35.9 (25.1–37.4) <0.001
Cesarean sectionb 16.7% (6/36) 39% (16/41) 0.04
Birthweight (grams; median [IQR])a 2935 (2425–3300) 2475 (740–3185) 0.04
Acute maternal inflammatory responseb
 Stage 1 (Early acute subchorionitis or chorionitis) 17.2% (5/29)g 10.8% (4/37)f 0.4
 Stage 2 (Acute chorioamnionitis) 17.2% (5/29)g 24.3% (9/37)f 0.55
 Stage 3 (Necrotizing chorioamnionitis) 3.4% (1/29)g 24.3% (9/37)f 0.03
Acute fetal inflammatory responseb
 Stage 1 (Chorionic vasculitis or umbilical phlebitis) 27.6% (8/29)g 18.9% (7/37)f 0.55
 Stage 2 (Umbilical arteritis) 10.3% (3/29)g 21.6% (8/37)f 0.3
 Stage 3 (Necrotizing funisitis) 0% (0/29)g 10.8% (4/37)f 0.1

Data are given as median (interquartile range, IQR) and percentage (n/N).

a

Mann-Whitney U test,

b

Fisher’s exact test,

c

One missing data,

d

Two missing data,

e

Three missing data,

f

Four missing data,

g

Seven missing data.

Abbreviations: WBC, white blood cells.

Clinical definitions

Gestational age was determined by the date of the last menstrual period and confirmed by ultrasound examination. The gestational age derived from sonographic fetal biometry was used if the estimation was inconsistent with menstrual dating. Spontaneous preterm labor was diagnosed by the presence of regular uterine contractions (at least two contractions every 10 minutes) associated with cervical changes in patients <37 weeks of gestation. Intra-amniotic inflammation was diagnosed when the interleukin-6 (IL-6) amniotic fluid concentration was ≥2.6 ng/mL (63, 79).

Placental histopathological examination

Placentas were examined histologically by perinatal pathologists blinded to clinical diagnoses and obstetrical outcomes according to standardized Perinatology Research Branch protocols (80, 81). Briefly, three to nine sections of the placenta were examined, and at least one full-thickness section was taken from the center of the placenta; others were taken randomly from the placental disc. Acute inflammatory lesions of the placenta (maternal inflammatory response and fetal inflammatory response) were diagnosed according to established criteria including staging and grading (80, 82). The proportions of patients whose placentas presented acute maternal and/or fetal inflammatory responses are displayed in Table 1.

Amniotic fluid sample collection

Amniotic fluid samples were collected to detect intra‐amniotic inflammation and/or infection in patients with a short cervix. Samples of amniotic fluid were transported to the laboratory in a sterile, capped syringe and immunophenotyping by flow cytometry was performed immediately. The rest of the sample was centrifuged at 1300 × g for 10 minutes at 4°C, and the supernatant was stored at −80°C until use. Additionally, an aliquot of amniotic fluid was transported to the clinical laboratory for culture of aerobic/anaerobic bacteria and genital mycoplasmas. The clinical laboratory also determined the amniotic fluid white blood cell (WBC) count (83), performed a Gram stain (84), and determined the amniotic fluid glucose concentration (85).

Determination of IL-6 concentration in amniotic fluid

Amniotic fluid concentrations of IL-6 were determined as previously established (63) using a sensitive and specific enzyme immunoassay obtained from R&D Systems (Minneapolis, MN, USA). The IL-6 concentrations were determined by interpolation from the standard curves. The inter- and intra-assay coefficients of variation for IL-6 were 8.7% and 4.6%, respectively. The sensitivity of the IL-6 assay was 0.09 pg/mL.

Multiplex determination of inflammatory-related proteins

Amniotic fluid samples were assessed using sensitive and specific V-PLEX immunoassays (Meso Scale Discovery, Gaithersburg, MD, USA) to measure amniotic fluid concentrations of several immune mediators: the Pro-inflammatory Panel 1 kit [Cat# K15049D (IL-1β, TNFα, IL-8, IL-12p70, IFNγ, IL-2, IL-4, IL-10, IL-13)], and Chemokine Panel 1 kit [Cat# K15047D; MCP-1 (CCL2), MIP-1α (CCL3), MIP-1β (CCL4), MCP-4 (CCL13), MDC (CCL22), IP-10 (CXCL10), TARC (CCL17), Eotaxin (CCL11), Eotaxin-3 (CCL26)], according to the manufacturer’s instructions. The plate signals were read using the QuickPlex SQ 120 (Meso Scale Discovery). Standard curves were generated and the assay values of the samples were interpolated from the curves. The detection limits of the assays were 0.05 pg/mL (IL-1β), 0.04 pg/mL (TNF‐α), 0.07 pg/mL (IL‐8), 0.11 pg/mL (IL‐12p70), 0.37 pg/mL (IFN‐γ), 0.09 pg/mL (IL‐2), 0.02 pg/mL (IL‐4), 0.04 pg/mL (IL‐10), 0.24 pg/mL (IL‐13), 0.09 pg/mL (MCP-1), 3.02 pg/mL (MIP-1α), 0.17 pg/mL (MIP-1β), 1.69 pg/mL (MCP-4), 1.22 pg/mL (MDC), 0.37 pg/mL (IP-10), 0.22 pg/mL (TARC), 3.26 pg/mL (Eotaxin), 1.77 pg/mL (Eotaxin-3).

Immunophenotyping by flow cytometry

Amniotic fluid samples (0.5‐1 mL) were centrifuged at 300 × g for 5 minutes at room temperature. The resulting amniotic fluid pellet was resuspended in 1 mL of 1X phosphate‐buffered saline (PBS) (Life Technologies, Grand Island, NY, USA) and stained with BD Horizon Fixable Viability Stain 510 dye (BD Biosciences, San Jose, CA, USA). Cells were washed in 1X PBS and incubated with 20 μL of human FcR blocking reagent (Miltenyi Biotec, San Diego, CA, USA) in 80 μL of stain buffer (BD Biosciences) for 10 minutes at 4°C. Next, cells were incubated with extracellular fluorochrome‐conjugated anti‐human monoclonal antibodies for 30 minutes at 4°C in the dark (Supplementary Table 1). Stained cells were then washed with 1X PBS, resuspended in 0.5 mL of stain buffer, and acquired using the BD LSR II or LSRFortessa Flow Cytometer (BD Bioscience) and BD FACSDiva 6.0 software (BD Bioscience). The analysis was performed and the figures were generated using the FlowJo version 10 software (FlowJo, Ashland, OR, USA). The absolute number of cells was determined using CountBright absolute counting beads (Molecular Probes, Eugene, OR, USA).

Statistical analysis

Statistical analyses were conducted using GraphPad Prism version 8.0.1 for Windows (GraphPad Software, San Diego, California, USA, www.graphpad.com). For patient demographics, the Mann-Whitney U-test was used to compare continuous variables and the Fisher’s exact test was used for nominal variables. The Mann-Whitney U-test was performed to compare the number of amniotic fluid immune cells and cytokine concentrations between the two study groups. A p‐value <0.05 was considered statistically significant for all tests.

RESULTS

Characteristics of the study population

The clinical and demographic characteristics of the study population are shown in Table 1. A total of 77 amniotic fluid samples were collected from asymptomatic women with a short cervix diagnosed using ultrasound. Thirty-six women had a cervical length 15–25 mm (hereafter referred to as a short cervix) and 41 had a cervical length ≤15 mm (hereafter referred to as a severely short cervix). Women with a severely short cervix had significantly higher amniotic fluid concentrations of IL-6 and consequently a higher rate of intra-amniotic inflammation than those with a short cervix (Table 1). In addition, women with a cervical length ≤15 mm delivered at an earlier gestational age and their newborns had a lower birthweight than those women who had a cervical length 15–25 mm (Table 1). No differences in maternal age, body mass index, race, gestational age at amniocentesis, amniotic fluid glucose concentrations, or WBC counts were found between the study groups. No differences in the treatment received by women with a short cervix were found between the study groups (Table 1). Patients with a cervical length ≤15 mm had a greater prevalence of severe maternal inflammatory responses in the placenta (i.e. stage 3) than those with a cervical length 15–25 mm (Table 1). Yet, no differences were observed in acute fetal inflammatory responses between the study groups (Table 1).

Amniotic fluid cellular and soluble immune responses in women with a short cervix

First, we determined the cellular immune composition of amniotic fluid using multi-color flow cytometry. Viable immune cell populations were identified using previously reported gating strategies (7276). The following immune cell types are reported: total leukocytes (CD45+ cells), neutrophils (CD45+CD15+CD14− cells), monocytes/macrophages (CD45+CD15-CD14+ cells), B cells (CD45+CD15-CD14-CD3-CD19+ cells), and T cells (CD45+CD15-CD14-CD3+CD19− cells). T cells were further identified as CD4+ T cells (CD3+CD4+CD8− cells) or CD8+ T cells (CD3+CD4-CD8+ cells).

No differences were found in the overall number of amniotic fluid leukocytes in women with a cervical length ≤15 mm compared to those with a cervical length 15–25 mm (Figure 1A). Similarly, no differences were found in the numbers of amniotic fluid innate immune cells (neutrophils and monocytes/macrophages) between the study groups (Figure 1B&C). Lastly, the numbers of adaptive immune cells (T cells and B cells) in amniotic fluid were not different between the study groups (Figure 1DG). These results show that women with a severely short cervix (≤15 mm) do not have a different cellular immune composition than those with a short cervix (15–25 mm).

Figure 1. Cellular immune composition of amniotic fluid.

Figure 1.

Numbers of (A) total leukocytes (CD45+ cells/mL), (B) neutrophils (CD15+ cells/mL), (C) monocytes/macrophages (CD14+ cells/mL), (D) B cells (CD19+ cells/mL), (E) total T cells (cells/mL), (F) CD4+ T cells (cells/mL) and (G) CD8+ T cells (cells/mL) in amniotic fluid from women with a short cervix with a cervical length 15–25 mm or ≤15 mm. N = 36–41 per group. Midlines = median, boxes = interquartile ranges, and whiskers = minimum/maximum ranges.

Given this negative result, we next evaluated whether the concentrations of cytokines/chemokines in amniotic fluid were different between women with a short cervix and those with a severely short cervix. Sixty-three amniotic fluid samples were available to evaluate inflammatory mediator concentrations. Amniotic fluid concentrations of IL-1β (Figure 2A), TNF-α (Figure 2B), IL-8 (Figure 2C), IL-12p70 (Figure 2D), IL-2 (Figure 2F), IL-4 (Figure 2G), IL-10 (Figure 2H), IL-13 (Figure 2I), MCP-1 (Figure 2J), MIP-1α (Figure 2K), MIP-1β (Figure 2L), MCP-4 (Figure 2M), and eotaxin (Figure 2Q) were higher in women with a cervical length ≤15 mm than in those with a cervical length 15–25 mm. No differences were found in the amniotic fluid concentrations of IFN-γ (Figure 2E), MDC (Figure 2N), IP-10 (Figure 2O), TARC (Figure 2P) and eotaxin-3 (Figure 2R) between the study groups. These findings demonstrate that a severely short cervix (≤15 mm) is accompanied by elevated concentrations of pro- and anti-inflammatory mediators in the amniotic cavity compared to those with a short cervix (15–25 mm).

Figure 2. Immune mediators in amniotic fluid.

Figure 2.

Amniotic fluid concentrations of cytokines [(A) IL-1β (pg/mL), (B) TNF-α (pg/mL), (C) IL-8 (pg/mL), (D) IL-12p70 (pg/mL), (E) IFN-γ (pg/mL), (F) IL-2 (pg/mL), (G) IL-4 (pg/mL), (H) IL-10 (pg/mL), (I) IL-13 (pg/mL)] and chemokines [(J) MCP-1 (pg/mL), (K) MIP-1α (pg/mL), (L) MIP-1 β (pg/mL), (M) MCP-4 (pg/mL), (N) MDC (pg/mL), (O) IP-10 (pg/mL), (P) TARC (pg/mL), (Q) eotaxin (pg/mL), and (R) eotaxin-3 (pg/mL)] in women with a short cervix with a cervical length 15–25 mm or ≤15 mm. N = 29–34 per group. Midlines = median, boxes = interquartile ranges, and whiskers = minimum/maximum ranges.

Amniotic fluid soluble and cellular immune responses in women with a severely short cervix (≤15 mm) who delivered preterm or at term

Thus far, we showed that women with a severely short cervix (≤15 mm) had elevated concentrations of pro-inflammatory mediators in amniotic fluid. Hence, we focused on this patient group and determined whether women who delivered preterm could be differentiated from those who delivered at term based on amniotic fluid cellular immune composition and/or soluble immune mediator concentrations. Interestingly, the amniotic fluid cellular immune composition was not different between women with a severely short cervix who ultimately delivered preterm compared to those who delivered at term (Figure 3AG). Among the 18 amniotic fluid cytokines/chemokines evaluated, only IL-2 concentrations were significantly elevated in women with a severely short cervix who ultimately delivered preterm compared to those who delivered at term (Figure 4F). No other differences were found in the amniotic fluid cytokine/chemokine concentrations between women with a severely short cervix who ultimately delivered preterm and those who delivered at term (Figure 4AE, GR). Taken together, these results show that the cellular and soluble immune responses in the amniotic cavity are not altered in women with a severely short cervix who are at high risk of preterm birth compared to those who will deliver at term.

Figure 3. Cellular immune composition of amniotic fluid from women with a severely short cervix.

Figure 3.

Numbers of (A) total leukocytes (CD45+ cells/mL), (B) neutrophils (CD15+ cells/mL), (C) monocytes/macrophages (CD14+ cells/mL), (D) B cells (CD19+ cells/mL), (E) total T cells (cells/mL), (F) CD4+ T cells (cells/mL) and (G) CD8+ T cells (cells/mL) in amniotic fluid from women with a cervical length ≤15 mm who delivered preterm or at term. N = 14–27 per group. Midlines = median, boxes = interquartile ranges, and whiskers = minimum/maximum ranges.

Figure 4. Immune mediators in amniotic fluid of women with a severely short cervix.

Figure 4.

Amniotic fluid concentrations of cytokines [(A) IL-1β (pg/mL), (B) TNF-α (pg/mL), (C) IL-8 (pg/mL), (D) IL-12p70 (pg/mL), (E) IFN-γ (pg/mL), (F) IL-2 (pg/mL), (G) IL-4 (pg/mL), (H) IL-10 (pg/mL), (I) IL-13 (pg/mL)] and chemokines [(J) MCP-1 (pg/mL), (K) MIP-1α (pg/mL), (L) MIP-1 β (pg/mL), (M) MCP-4 (pg/mL), (N) MDC (pg/mL), (O) IP-10 (pg/mL), (P) TARC (pg/mL), (Q) eotaxin (pg/mL), and (R) eotaxin-3 (pg/mL)] in women with a cervical length ≤15 mm who delivered preterm or at term. N =13–21 per group. Midlines = median, boxes = interquartile ranges, and whiskers = minimum/maximum ranges.

DISCUSSION

The principal findings of the current study are: 1) the cellular immune composition of amniotic fluid was not different between women with a severely short cervix (≤15 mm) and those with a short cervix 15–25 mm; 2) amniotic fluid concentrations of multiple cytokines/chemokines were higher in women with a severely short cervix (≤15 mm) than in those with a short cervix 15–25 mm; 3) the cellular immune composition of amniotic fluid was not different between women with a severely short cervix (≤15 mm) who ultimately underwent preterm delivery compared to those who delivered at term; and 4) amniotic fluid concentrations of IL-2, but not other immune mediators, were increased in women with a severely short cervix (≤15 mm) who ultimately delivered preterm compared to those who delivered at term. Collectively, these findings show that women with a severely short cervix (≤15 mm) have increased concentrations of immune mediators in the amniotic cavity compared to those with a short cervix 15–25 mm; yet, this is not translated to changes in the cellular immune composition.

Herein, we found that women with a severely short cervix (≤15 mm) had higher amniotic fluid concentrations of multiple soluble mediators than those with a short cervix 15–25 mm. This finding is consistent with previous reports focused on the cytokine responses in amniotic fluid of women with a short cervix (25, 49, 51, 77, 78). Yet, this elevated cytokine profile in the amniotic fluid was not accompanied by a concomitant increase in the cellular immune composition. This result has two potential explanations: 1) some women with a short cervix have elevated concentrations of IL-6 (some of these cases may be due to sterile intra-amniotic inflammation) and low white blood cell counts (19); or 2) some women with a short cervix have increased concentrations of specific inflammatory mediators (independent of increased IL-6 or related inflammatory mediators) and low white blood cell counts (86). These two scenarios occur in the absence of culturable microorganisms in amniotic fluid because we excluded cases with positive microbiological cultures; therefore, immune cells are increased when viable microbes invade the amniotic cavity. For example, an enhanced cellular immune response is commonly observed in cases of microbial invasion of the amniotic cavity accompanied by intra-amniotic inflammation (i.e. intra-amniotic infection) (7376). This intra-amniotic inflammatory response is characterized by the massive invasion of maternal and/or fetal immune cells such as neutrophils and monocytes/macrophages (87, 88), which enter the amniotic fluid to facilitate host defense against invaders. Neutrophils in the amniotic cavity possess an arsenal of weapons for combating microorganisms including the formation of neutrophil extracellular traps (NETS) (89), phagocytosis (90), production of reactive oxygen species (91), and by degranulation, which leads to the release of anti-microbial molecules such as elastase (9294), alpha-defensins (9497), lactoferrin (98), pentraxin-3 (99), myeloperoxidase (94, 100, 101), cathelicidin (94, 100) and cathepsin G (94, 102). Monocytes/macrophages in the amniotic cavity participate in host responses by releasing massive amounts of pro-inflammatory mediators such as IL-1β (73, 75). Moreover, macrophage extracellular traps (METs) have also been detected in the placental tissues upon infection with Streptococcus agalactiae (103). Our unpublished data also suggest that monocytes/macrophages orchestrate inflammasome activation in the amniotic cavity of women with intra-amniotic infection. In conclusion, increased cellular immune responses in the amniotic cavity are largely governed by the presence of viable microorganisms, and in the specific context of a sonographic short cervix inflammation is characterized by high concentrations of cytokines/chemokines without an exacerbated cellular immune response.

Interestingly, women with a severely short cervix (≤15 mm) who ultimately delivered preterm had elevated amniotic fluid concentrations of IL-2 compared to those who delivered at term. Interleukin-2 is a cytokine released upon T-cell activation (104) and has been previously shown to be increased in the amniotic cavity of women with a short cervix (25). This cytokine is also increased in amniotic fluid of women with intra-amniotic inflammation and/or infection who delivered preterm (86, 105, 106). Recently, we have also shown that fetal T cells found in amniotic fluid can express IL-2 upon stimulation (86). In addition, amniotic fluid concentrations of IL-2 tend to increase in women with idiopathic preterm labor and birth compared to those who had an episode of preterm labor but delivered at term (86). Taken together, these data suggest that IL-2, and potentially T-cell activation processes, are involved in the pathophysiology of preterm labor and birth in women with a severely short cervix.

Despite the absence of an augmented cellular immune response, women with a severely short cervix had an elevated pro-inflammatory cytokine/chemokine profile in the amniotic cavity. The most likely source of these cytokines is the fetal tissues. For example, it is likely that in women with a severely short cervix (≤15 mm) the chorioamniotic membranes are under greater tension than those of women with a short cervix 15–25 mm. This increased tensile force on the chorioamniotic membranes results in the release of several inflammatory mediators into the amniotic cavity (107). Indeed, it has been proposed that the cervical zone of the chorioamniotic membranes displays a unique inflammatory profile, which is different from that of the middle and peri-placental zones (108115). However, whether the chorioamniotic membranes from women with a severely short cervix (who are at higher risk of preterm delivery) display a different cytokine profile than those from women with a short cervix 15–25 mm requires further investigation.

It is worth mentioning this descriptive cross-sectional study does not allow us to determine causal links between intra-amniotic inflammation and a sonographic short cervix. It would be interesting to investigate whether inflammatory stimuli (e.g. bacteria) can induce cervical shortening or whether a short cervix leads to intra-amniotic inflammation. It is also likely that a short cervix is not accompanied by intra-amniotic inflammation, highlighting the syndromic nature of this clinical entity.

A limitation of this study is that we could not include gestational age-matched controls with a normal cervical length (>25 mm). This is due to the fact that cervical length measurement is not routinely performed in women who underwent genetic amniocentesis. However, the aim of this study was to compare cellular and soluble immune responses among women diagnosed with a sonographic short cervix.

CONCLUSION

In the current study, we report that the cellular immune composition of amniotic fluid was not different between women with a severely short cervix (≤15 mm) and those with a short cervix 15–25 mm. Yet, amniotic fluid concentrations of multiple cytokines/chemokines were higher in women with a severely short cervix (≤15 mm) than in those with a short cervix 15–25 mm. Interestingly, amniotic fluid concentrations of IL-2, but not other immune mediators, were increased in women with a severely short cervix (≤15 mm) who ultimately delivered preterm compared to those who delivered at term. These findings provide evidence that women with a severely short cervix (≤15 mm) have increased concentrations of pro-inflammatory mediators in the amniotic cavity; yet, this is not translated to changes in the cellular immune response.

Supplementary Material

Supplementary Table 1

ACKNOWLEDGEMENTS

We gratefully acknowledge Rona Wang and Hong Meng for her contributions to the execution of this study. We thank the physicians and nurses from the Center for Advanced Obstetrical Care and Research and the Intrapartum Unit for their help in collecting human samples. We also thank staff members of the PRB Clinical Laboratory and the PRB Histology/Pathology Unit for the processing and examination of the pathological sections.

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, 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. R. R. has contributed to this work as part of his official duties as an employee of the United States Federal Government. N. G.-L. was also supported by the Wayne State University Perinatal Initiative in Maternal, Perinatal and Child Health.

Footnotes

DECLARATION OF INTEREST

The authors declare no conflicts of interest.

REFERENCES

  • 1.Romero R, Espinoza J, Kusanovic JP, Gotsch F, Hassan S, Erez O, et al. The preterm parturition syndrome. BJOG. 2006;113 Suppl 3:17–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Goldenberg RL, Culhane JF, Iams JD, Romero R. Epidemiology and causes of preterm birth. Lancet. 2008;371:75–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Di Renzo GC. The great obstetrical syndromes. J Matern Fetal Neonatal Med. 2009;22:633–5. [DOI] [PubMed] [Google Scholar]
  • 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Liu L, Oza S, Hogan D, Perin J, Rudan I, Lawn JE, et al. Global, regional, and national causes of child mortality in 2000–13, with projections to inform post-2015 priorities: an updated systematic analysis. Lancet. 2015;385:430–40. [DOI] [PubMed] [Google Scholar]
  • 6.Chawanpaiboon S, Vogel JP, Moller AB, Lumbiganon P, Petzold M, Hogan D, et al. Global, regional, and national estimates of levels of preterm birth in 2014: a systematic review and modelling analysis. Lancet Glob Health. 2019;7:e37–e46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.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. [DOI] [PubMed] [Google Scholar]
  • 8.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. National Institute of Child Health and Human Development Maternal Fetal Medicine Unit Network. N Engl J Med. 1996;334:567–72. [DOI] [PubMed] [Google Scholar]
  • 9.Goldenberg RL, Iams JD, Mercer BM, Meis PJ, Moawad AH, Copper RL, et al. The preterm prediction study: the value of new vs standard risk factors in predicting early and all spontaneous preterm births. NICHD MFMU Network. Am J Public Health. 1998;88:233–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Heath VC, Southall TR, Souka AP, Elisseou A, Nicolaides KH. Cervical length at 23 weeks of gestation: prediction of spontaneous preterm delivery. Ultrasound Obstet Gynecol. 1998;12:312–7. [DOI] [PubMed] [Google Scholar]
  • 11.Watson WJ, Stevens D, Welter S, Day D. Observations on the sonographic measurement of cervical length and the risk of premature birth. J Matern Fetal Med. 1999;8:17–9. [DOI] [PubMed] [Google Scholar]
  • 12.Berghella V, Daly SF, Tolosa JE, DiVito MM, Chalmers R, Garg N, et al. Prediction of preterm delivery with transvaginal ultrasonography of the cervix in patients with high-risk pregnancies: does cerclage prevent prematurity? Am J Obstet Gynecol. 1999;181:809–15. [DOI] [PubMed] [Google Scholar]
  • 13.Hassan SS, Romero R, Berry SM, Dang K, Blackwell SC, Treadwell MC, et al. Patients with an ultrasonographic cervical length < or =15 mm have nearly a 50% risk of early spontaneous preterm delivery. Am J Obstet Gynecol. 2000;182:1458–67. [DOI] [PubMed] [Google Scholar]
  • 14.To MS, Skentou C, Liao AW, Cacho A, Nicolaides KH. Cervical length and funneling at 23 weeks of gestation in the prediction of spontaneous early preterm delivery. Ultrasound Obstet Gynecol. 2001;18:200–3. [DOI] [PubMed] [Google Scholar]
  • 15.Owen J, Yost N, Berghella V, Thom E, Swain M, Dildy GA 3rd, et al. Mid-trimester endovaginal sonography in women at high risk for spontaneous preterm birth. JAMA. 2001;286:1340–8. [DOI] [PubMed] [Google Scholar]
  • 16.Mazaki-Tovi S, Romero R, Kusanovic JP, Erez O, Pineles BL, Gotsch F, et al. Recurrent preterm birth. Semin Perinatol. 2007;31:142–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Romero R Prevention of spontaneous preterm birth: the role of sonographic cervical length in identifying patients who may benefit from progesterone treatment. Ultrasound Obstet Gynecol. 2007;30:675–86. [DOI] [PubMed] [Google Scholar]
  • 18.Romero R, Dey SK, Fisher SJ. Preterm labor: one syndrome, many causes. Science. 2014;345:760–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.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. 2015;28:1343–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Romero R, Conde-Agudelo A, Da Fonseca E, O’Brien JM, Cetingoz E, Creasy GW, et al. Vaginal progesterone for preventing preterm birth and adverse perinatal outcomes in singleton gestations with a short cervix: a meta-analysis of individual patient data. Am J Obstet Gynecol. 2018;218:161–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Hernandez-Andrade E, Maymon E, Luewan S, Bhatti G, Mehrmohammadi M, Erez O, et al. A soft cervix, categorized by shear-wave elastography, in women with short or with normal cervical length at 18–24 weeks is associated with a higher prevalence of spontaneous preterm delivery. J Perinat Med. 2018;46:489–501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Conde-Agudelo A, Romero R, Da Fonseca E, O’Brien JM, Cetingoz E, Creasy GW, et al. Vaginal progesterone is as effective as cervical cerclage to prevent preterm birth in women with a singleton gestation, previous spontaneous preterm birth, and a short cervix: updated indirect comparison meta-analysis. Am J Obstet Gynecol. 2018;219:10–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Liu CZ, Ho N, Nguyen AD, Lehner C, Sekar R, Amoako AA. The risk of preterm delivery and pregnancy outcomes in women with asymptomatic short cervix: a retrospective cohort study. J Matern Fetal Neonatal Med. 2019:1–7. [DOI] [PubMed] [Google Scholar]
  • 24.Vaisbuch E, Hassan SS, Mazaki-Tovi S, Nhan-Chang CL, Kusanovic JP, Chaiworapongsa T, et al. Patients with an asymptomatic short cervix (<or=15 mm) have a high rate of subclinical intraamniotic inflammation: implications for patient counseling. Am J Obstet Gynecol. 2010;202:433 e1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Tarca AL, Fitzgerald W, Chaemsaithong P, Xu Z, Hassan SS, Grivel JC, et al. The cytokine network in women with an asymptomatic short cervix and the risk of preterm delivery. Am J Reprod Immunol. 2017;78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Romero R, Espinoza J, Erez O, Hassan S. The role of cervical cerclage in obstetric practice: can the patient who could benefit from this procedure be identified? Am J Obstet Gynecol. 2006;194:1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Romero R, Yeo L, Miranda J, Hassan SS, Conde-Agudelo A, Chaiworapongsa T. A blueprint for the prevention of preterm birth: vaginal progesterone in women with a short cervix. J Perinat Med. 2013;41:27–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Moinian M, Andersch B. Does cervix conization increase the risk of complications in subsequent pregnancies? Acta Obstet Gynecol Scand. 1982;61:101–3. [DOI] [PubMed] [Google Scholar]
  • 29.Kristensen J, Langhoff-Roos J, Wittrup M, Bock JE. Cervical conization and preterm delivery/low birth weight. A systematic review of the literature. Acta Obstet Gynecol Scand. 1993;72:640–4. [DOI] [PubMed] [Google Scholar]
  • 30.Raio L, Ghezzi F, Di Naro E, Gomez R, Luscher KP. Duration of pregnancy after carbon dioxide laser conization of the cervix: influence of cone height. Obstet Gynecol. 1997;90:978–82. [DOI] [PubMed] [Google Scholar]
  • 31.Berghella V, Pereira L, Gariepy A, Simonazzi G. Prior cone biopsy: prediction of preterm birth by cervical ultrasound. Am J Obstet Gynecol. 2004;191:1393–7. [DOI] [PubMed] [Google Scholar]
  • 32.Bruinsma FJ, Quinn MA. The risk of preterm birth following treatment for precancerous changes in the cervix: a systematic review and meta-analysis. BJOG. 2011;118:1031–41. [DOI] [PubMed] [Google Scholar]
  • 33.Miller ES, Grobman WA. The association between cervical excisional procedures, midtrimester cervical length, and preterm birth. Am J Obstet Gynecol. 2014;211:242 e1–4. [DOI] [PubMed] [Google Scholar]
  • 34.Miller ES, Sakowicz A, Grobman WA. The association between cervical dysplasia, a short cervix, and preterm birth. Am J Obstet Gynecol. 2015;213:543 e1–4. [DOI] [PubMed] [Google Scholar]
  • 35.Gupta S, Chen S, Naqvi M, Saltzman DH, Rebarber A, Monteagudo A, et al. Change in cervical length and spontaneous preterm birth in nulliparous women with a history of loop electrosurgical excision procedure (*). J Matern Fetal Neonatal Med. 2019:1–5. [DOI] [PubMed] [Google Scholar]
  • 36.Singer MS, Hochman M. Incompetent cervix in a hormone-exposed offspring. Obstet Gynecol. 1978;51:625–6. [DOI] [PubMed] [Google Scholar]
  • 37.Goldstein DP. Incompetent cervix in offspring exposed to diethylstilbestrol in utero. Obstet Gynecol. 1978;52:73S–5S. [PubMed] [Google Scholar]
  • 38.Mangan CE, Borow L, Burtnett-Rubin MM, Egan V, Giuntoli RL, Mikuta JJ. Pregnancy outcome in 98 women exposed to diethylstilbestrol in utero, their mothers, and unexposed siblings. Obstet Gynecol. 1982;59:315–9. [PubMed] [Google Scholar]
  • 39.Ludmir J, Landon MB, Gabbe SG, Samuels P, Mennuti MT. Management of the diethylstilbestrol-exposed pregnant patient: a prospective study. Am J Obstet Gynecol. 1987;157:665–9. [DOI] [PubMed] [Google Scholar]
  • 40.Goldberg JM, Falcone T. Effect of diethylstilbestrol on reproductive function. Fertil Steril. 1999;72:1–7. [DOI] [PubMed] [Google Scholar]
  • 41.Hordnes K Ehlers-Danlos syndrome and delivery. Acta Obstet Gynecol Scand. 1994;73:671–3. [DOI] [PubMed] [Google Scholar]
  • 42.Ploeckinger B, Ulm MR, Chalubinski K. Ehlers-Danlos syndrome type II in pregnancy. Am J Perinatol. 1997;14:99–101. [DOI] [PubMed] [Google Scholar]
  • 43.Kindinger LM, Bennett PR, Lee YS, Marchesi JR, Smith A, Cacciatore S, et al. The interaction between vaginal microbiota, cervical length, and vaginal progesterone treatment for preterm birth risk. Microbiome. 2017;5:6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Witkin SS, Moron AF, Ridenhour BJ, Minis E, Hatanaka A, Sarmento SGP, et al. Vaginal Biomarkers That Predict Cervical Length and Dominant Bacteria in the Vaginal Microbiomes of Pregnant Women. mBio. 2019;10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Gerson KD, McCarthy C, Elovitz MA, Ravel J, Sammel MD, Burris HH. Cervicovaginal microbial communities deficient in Lactobacillus species are associated with second trimester short cervix. Am J Obstet Gynecol. 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Romero R, Gonzalez R, Sepulveda W, Brandt F, Ramirez M, Sorokin Y, et al. Infection and labor. VIII. Microbial invasion of the amniotic cavity in patients with suspected cervical incompetence: prevalence and clinical significance. Am J Obstet Gynecol. 1992;167:1086–91. [DOI] [PubMed] [Google Scholar]
  • 47.Hassan S, Romero R, Hendler I, Gomez R, Khalek N, Espinoza J, et al. A sonographic short cervix as the only clinical manifestation of intra-amniotic infection. J Perinat Med. 2006;34:13–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Lee SE, Romero R, Park CW, Jun JK, Yoon BH. The frequency and significance of intraamniotic inflammation in patients with cervical insufficiency. Am J Obstet Gynecol. 2008;198:633 e1–8. [DOI] [PubMed] [Google Scholar]
  • 49.Kiefer DG, Keeler SM, Rust OA, Wayock CP, Vintzileos AM, Hanna N. Is midtrimester short cervix a sign of intraamniotic inflammation? Am J Obstet Gynecol. 2009;200:374 e1–5. [DOI] [PubMed] [Google Scholar]
  • 50.Keeler SM, Kiefer DG, Rust OA, Vintzileos A, Atlas RO, Bornstein E, et al. Comprehensive amniotic fluid cytokine profile evaluation in women with a short cervix: which cytokine(s) correlates best with outcome? Am J Obstet Gynecol. 2009;201:276 e1–6. [DOI] [PubMed] [Google Scholar]
  • 51.Kiefer DG, Keeler SM, Rust O, Chow SS, Craig ME, Peltier MR, et al. Amniotic fluid inflammatory score is associated with pregnancy outcome in patients with mid trimester short cervix. Am J Obstet Gynecol. 2012;206:68 e1–6. [DOI] [PubMed] [Google Scholar]
  • 52.Romero R, Espinoza J, Chaiworapongsa T, Kalache K. Infection and prematurity and the role of preventive strategies. Semin Neonatol. 2002;7:259–74. [DOI] [PubMed] [Google Scholar]
  • 53.Goncalves LF, Chaiworapongsa T, Romero R. Intrauterine infection and prematurity. Ment Retard Dev Disabil Res Rev. 2002;8:3–13. [DOI] [PubMed] [Google Scholar]
  • 54.Romero R, Espinoza J, Goncalves LF, Kusanovic JP, Friel LA, Nien JK. Inflammation in preterm and term labour and delivery. Semin Fetal Neonatal Med. 2006;11:317–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Romero R, Espinoza J, Goncalves LF, Kusanovic JP, Friel L, Hassan S. The role of inflammation and infection in preterm birth. Semin Reprod Med. 2007;25:21–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Monckeberg M, Valdes R, Kusanovic JP, Schepeler M, Nien JK, Pertossi E, et al. Patients with acute cervical insufficiency without intra-amniotic infection/inflammation treated with cerclage have a good prognosis. J Perinat Med. 2019;47:500–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Romero R, Gomez-Lopez N, Winters AD, Jung E, Shaman M, Bieda J, et al. Evidence that intra-amniotic infections are often the result of an ascending invasion - a molecular microbiological study. J Perinat Med. 2019;47:915–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Oh KJ, Romero R, Park JY, Lee J, Conde-Agudelo A, Hong JS, et al. Evidence that antibiotic administration is effective in the treatment of a subset of patients with intra-amniotic infection/inflammation presenting with cervical insufficiency. Am J Obstet Gynecol. 2019;221:140 e1–e18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Oh KJ, Romero R, Park JY, Hong JS, Yoon BH. The earlier the gestational age, the greater the intensity of the intra-amniotic inflammatory response in women with preterm premature rupture of membranes and amniotic fluid infection by Ureaplasma species. J Perinat Med. 2019;47:516–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Yoon BH, Romero R, Park JY, Oh KJ, Lee J, Conde-Agudelo A, et al. Antibiotic administration can eradicate intra-amniotic infection or intra-amniotic inflammation in a subset of patients with preterm labor and intact membranes. Am J Obstet Gynecol. 2019;221:142 e1–e22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Theis KR, Romero R, Motomura K, Galaz J, Winters AD, Pacora P, et al. Microbial burden and inflammasome activation in amniotic fluid of patients with preterm prelabor rupture of membranes. J Perinat Med. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Oh KJ, Romero R, Park JY, Kang J, Hong JS, Yoon BH. A high concentration of fetal fibronectin in cervical secretions increases the risk of intra-amniotic infection and inflammation in patients with preterm labor and intact membranes. J Perinat Med. 2019;47:288–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.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. [DOI] [PubMed] [Google Scholar]
  • 64.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Gomez-Lopez N, Romero R, Panaitescu B, Leng Y, Xu Y, Tarca AL, et al. Inflammasome activation during spontaneous preterm labor with intra-amniotic infection or sterile intra-amniotic inflammation. Am J Reprod Immunol. 2018;80:e13049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Musilova I, Andrys C, Drahosova M, Soucek O, Pliskova L, Jacobsson B, et al. Cervical fluid interleukin 6 and intra-amniotic complications of preterm prelabor rupture of membranes. J Matern Fetal Neonatal Med. 2018;31:827–36. [DOI] [PubMed] [Google Scholar]
  • 68.Gomez-Lopez N, Romero R, Maymon E, Kusanovic JP, Panaitescu B, Miller D, et al. Clinical chorioamnionitis at term IX: in vivo evidence of intra-amniotic inflammasome activation. J Perinat Med. 2019;47:276–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Gomez-Lopez N, Romero R, Tarca AL, Miller D, Panaitescu B, Schwenkel G, et al. Gasdermin D: Evidence of pyroptosis in spontaneous preterm labor with sterile intra-amniotic inflammation or intra-amniotic infection. Am J Reprod Immunol. 2019;82:e13184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Peiris HN, Romero R, Vaswani K, Reed S, Gomez-Lopez N, Tarca AL, et al. Preterm labor is characterized by a high abundance of amniotic fluid prostaglandins in patients with intra-amniotic infection or sterile intra-amniotic inflammation. J Matern Fetal Neonatal Med. 2019:1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Musilova I, Kolackova M, Andrys C, Drahosova M, Baranova I, Chmelarova M, et al. Nicotinamide phosphoribosyltransferase and intra-amniotic inflammation in preterm prelabor rupture of membranes. J Matern Fetal Neonatal Med. 2019:1–11. [DOI] [PubMed] [Google Scholar]
  • 72.Gomez-Lopez N, Romero R, Xu Y, Miller D, Leng Y, Panaitescu B, et al. The immunophenotype of amniotic fluid leukocytes in normal and complicated pregnancies. Am J Reprod Immunol. 2018;79:e12827. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Martinez-Varea A, Romero R, Xu Y, Miller D, Ahmed AI, Chaemsaithong P, et al. Clinical chorioamnionitis at term VII: the amniotic fluid cellular immune response. J Perinat Med. 2017;45:523–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Gomez-Lopez N, Romero R, Galaz J, Xu Y, Panaitescu B, Slutsky R, et al. Cellular immune responses in amniotic fluid of women with preterm labor and intra-amniotic infection or intra-amniotic inflammation. Am J Reprod Immunol. 2019;82:e13171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Galaz J, Romero R, Xu Y, Miller D, Slutsky R, Levenson D, et al. Cellular immune responses in amniotic fluid of women with preterm clinical chorioamnionitis. Inflamm Res. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Galaz J, Romero R, Slutsky R, Xu Y, Motomura K, Para R, et al. Cellular immune responses in women with preterm prelabor rupture of membranes. Journal of Perinatal Medicine. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Son GH, You YA, Kwon EJ, Lee KY, Kim YJ. Comparative Analysis of Midtrimester Amniotic Fluid Cytokine Levels to Predict Spontaneous Very Pre-term Birth in Patients with Cervical Insufficiency. Am J Reprod Immunol. 2016;75:155–61. [DOI] [PubMed] [Google Scholar]
  • 78.Kim YM, Park KH, Park H, Yoo HN, Kook SY, Jeon SJ. Complement C3a, But Not C5a, Levels in Amniotic Fluid Are Associated with Intra-amniotic Infection and/or Inflammation and Preterm Delivery in Women with Cervical Insufficiency or an Asymptomatic Short Cervix (</= 25 mm). J Korean Med Sci. 2018;33:e220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Musilova I, Andrys C, Holeckova M, Kolarova V, Pliskova L, Drahosova M, et al. Interleukin-6 measured using the automated electrochemiluminescence immunoassay method for the identification of intra-amniotic inflammation in preterm prelabor rupture of membranes. J Matern Fetal Neonatal Med. 2018:1–131. [DOI] [PubMed] [Google Scholar]
  • 80.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:S29–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Romero R, Kim YM, Pacora P, Kim CJ, Benshalom-Tirosh N, Jaiman S, et al. The frequency and type of placental histologic lesions in term pregnancies with normal outcome. J Perinat Med. 2018;46:613–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Redline RW. Classification of placental lesions. Am J Obstet Gynecol. 2015;213:S21–8. [DOI] [PubMed] [Google Scholar]
  • 83.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;165:821–30. [DOI] [PubMed] [Google Scholar]
  • 84.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. [DOI] [PubMed] [Google Scholar]
  • 85.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. [DOI] [PubMed] [Google Scholar]
  • 86.Gomez-Lopez N, Romero R, Xu Y, Miller D, Arenas-Hernandez M, Garcia-Flores V, et al. Fetal T Cell Activation in the Amniotic Cavity during Preterm Labor: A Potential Mechanism for a Subset of Idiopathic Preterm Birth. J Immunol. 2019;203:1793–807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Gomez-Lopez N, Romero R, Xu Y, Leng Y, Garcia-Flores V, Miller D, et al. Are amniotic fluid neutrophils in women with intraamniotic infection and/or inflammation of fetal or maternal origin? Am J Obstet Gynecol. 2017;217:693 e1–e16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Gomez-Lopez N, Romero R, Leng Y, Xu Y, Slutsky R, Levenson D, et al. The origin of amniotic fluid monocytes/macrophages in women with intra-amniotic inflammation or infection. J Perinat Med. 2019;47:822–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Gomez-Lopez N, Romero R, Xu Y, Miller D, Unkel R, Shaman M, et al. Neutrophil Extracellular Traps in the Amniotic Cavity of Women with Intra-Amniotic Infection: A New Mechanism of Host Defense. Reprod Sci. 2017;24:1139–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Gomez-Lopez N, Romero R, Garcia-Flores V, Xu Y, Leng Y, Alhousseini A, et al. Amniotic fluid neutrophils can phagocytize bacteria: A mechanism for microbial killing in the amniotic cavity. Am J Reprod Immunol. 2017;78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Novakovic TR, Dolicanin ZC, Djordjevic NZ. Effects of maternal subclinical hypothyroidism on amniotic fluid cells oxidative status. Reprod Toxicol. 2018;78:97–101. [DOI] [PubMed] [Google Scholar]
  • 92.Rivero-Marcotegui A, Larranaga-Azcarate C, Ceres-Ruiz R, Garcia-Merlo S. Polymorphonuclear elastase and interleukin-6 in amniotic fluid in preterm labor. Clin Chem. 1997;43:857–9. [PubMed] [Google Scholar]
  • 93.Helmig BR, Romero R, Espinoza J, Chaiworapongsa T, Bujold E, Gomez R, et al. Neutrophil elastase and secretory leukocyte protease inhibitor in prelabor rupture of membranes, parturition and intra-amniotic infection. J Matern Fetal Neonatal Med. 2002;12:237–46. [DOI] [PubMed] [Google Scholar]
  • 94.Romero R, Kusanovic JP, Gotsch F, Erez O, Vaisbuch E, Mazaki-Tovi S, et al. Isobaric labeling and tandem mass spectrometry: a novel approach for profiling and quantifying proteins differentially expressed in amniotic fluid in preterm labor with and without intra-amniotic infection/inflammation. J Matern Fetal Neonatal Med. 2010;23:261–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Heine RP, Wiesenfeld H, Mortimer L, Greig PC. Amniotic fluid defensins: potential markers of subclinical intrauterine infection. Clin Infect Dis. 1998;27:513–8. [DOI] [PubMed] [Google Scholar]
  • 96.Espinoza J, Chaiworapongsa T, Romero R, Edwin S, Rathnasabapathy C, Gomez R, et al. Antimicrobial peptides in amniotic fluid: defensins, calprotectin and bacterial/permeability-increasing protein in patients with microbial invasion of the amniotic cavity, intra-amniotic inflammation, preterm labor and premature rupture of membranes. J Matern Fetal Neonatal Med. 2003;13:2–21. [DOI] [PubMed] [Google Scholar]
  • 97.Akinbi HT, Narendran V, Pass AK, Markart P, Hoath SB. Host defense proteins in vernix caseosa and amniotic fluid. Am J Obstet Gynecol. 2004;191:2090–6. [DOI] [PubMed] [Google Scholar]
  • 98.Pacora P, Maymon E, Gervasi MT, Gomez R, Edwin SS, Yoon BH, et al. Lactoferrin in intrauterine infection, human parturition, and rupture of fetal membranes. Am J Obstet Gynecol. 2000;183:904–10. [DOI] [PubMed] [Google Scholar]
  • 99.Musilova I, Andrys C, Krejsek J, Drahosova M, Zednikova B, Pliskova L, et al. Amniotic fluid pentraxins: Potential early markers for identifying intra-amniotic inflammatory complications in preterm pre-labor rupture of membranes. Am J Reprod Immunol. 2018;79:e12789. [DOI] [PubMed] [Google Scholar]
  • 100.Gravett MG, Novy MJ, Rosenfeld RG, Reddy AP, Jacob T, Turner M, et al. Diagnosis of intra-amniotic infection by proteomic profiling and identification of novel biomarkers. JAMA. 2004;292:462–9. [DOI] [PubMed] [Google Scholar]
  • 101.Myntti T, Rahkonen L, Nupponen I, Patari-Sampo A, Tikkanen M, Sorsa T, et al. Amniotic Fluid Infection in Preterm Pregnancies with Intact Membranes. Dis Markers. 2017;2017:8167276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Musilova I, Andrys C, Drahosova M, Soucek O, Pliskova L, Stepan M, et al. Amniotic fluid cathepsin-G in pregnancies complicated by the preterm prelabor rupture of membranes. J Matern Fetal Neonatal Med. 2017;30:2097–104. [DOI] [PubMed] [Google Scholar]
  • 103.Doster RS, Sutton JA, Rogers LM, Aronoff DM, Gaddy JA. Streptococcus agalactiae Induces Placental Macrophages To Release Extracellular Traps Loaded with Tissue Remodeling Enzymes via an Oxidative Burst-Dependent Mechanism. mBio. 2018;9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Tamura T, Nariuchi H. T cell activation through TCR/-CD3 complex. IL-2 production of T cell clones stimulated with anti-CD3 without cross-linkage. J Immunol. 1992;148:2370–7. [PubMed] [Google Scholar]
  • 105.Romero R, Grivel JC, Tarca AL, Chaemsaithong P, Xu Z, Fitzgerald W, et al. Evidence of perturbations of the cytokine network in preterm labor. Am J Obstet Gynecol. 2015;213:836 e1–e18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Bhatti G, Romero R, Rice GE, Fitzgerald W, Pacora P, Gomez-Lopez N, et al. Compartmentalized profiling of amniotic fluid cytokines in women with preterm labor. PLoS One. 2020;15:e0227881. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Adams Waldorf KM, Singh N, Mohan AR, Young RC, Ngo L, Das A, et al. Uterine overdistention induces preterm labor mediated by inflammation: observations in pregnant women and nonhuman primates. Am J Obstet Gynecol. 2015;213:830 e1–e19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Malak TM, Bell SC. Structural characteristics of term human fetal membranes: a novel zone of extreme morphological alteration within the rupture site. Br J Obstet Gynaecol. 1994;101:375–86. [DOI] [PubMed] [Google Scholar]
  • 109.McLaren J, Taylor DJ, Bell SC. Increased incidence of apoptosis in non-labour-affected cytotrophoblast cells in term fetal membranes overlying the cervix. Hum Reprod. 1999;14:2895–900. [DOI] [PubMed] [Google Scholar]
  • 110.El Khwad M, Stetzer B, Moore RM, Kumar D, Mercer B, Arikat S, et al. Term human fetal membranes have a weak zone overlying the lower uterine pole and cervix before onset of labor. Biol Reprod. 2005;72:720–6. [DOI] [PubMed] [Google Scholar]
  • 111.El Khwad M, Pandey V, Stetzer B, Mercer BM, Kumar D, Moore RM, et al. Fetal membranes from term vaginal deliveries have a zone of weakness exhibiting characteristics of apoptosis and remodeling. J Soc Gynecol Investig. 2006;13:191–5. [DOI] [PubMed] [Google Scholar]
  • 112.Nhan-Chang CL, Romero R, Tarca AL, Mittal P, Kusanovic JP, Erez O, et al. Characterization of the transcriptome of chorioamniotic membranes at the site of rupture in spontaneous labor at term. Am J Obstet Gynecol. 2010;202:462 e1–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.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. [DOI] [PubMed] [Google Scholar]
  • 114.Elfayomy AK, Almasry SM. Expression of tumor necrosis factor-alpha and vascular endothelial growth factor in different zones of fetal membranes: a possible relation to onset of labor. J Mol Histol. 2014;45:243–57. [DOI] [PubMed] [Google Scholar]
  • 115.Marcellin L, Schmitz T, Messaoudene M, Chader D, Parizot C, Jacques S, et al. Immune Modifications in Fetal Membranes Overlying the Cervix Precede Parturition in Humans. J Immunol. 2017;198:1345–56. [DOI] [PubMed] [Google Scholar]

Associated Data

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

Supplementary Materials

Supplementary Table 1

RESOURCES