Next Article in Journal
Cellular and Molecular Network Characteristics of TARM1-Related Genes in Mycobacterium tuberculosis Infections
Previous Article in Journal
Integrated miRNA and mRNA Transcriptome Analysis Reveals Regulatory Mechanisms in the Response of Winter Brassica rapa to Drought Stress
Previous Article in Special Issue
The Underlying Molecular Mechanisms of the Placenta Accreta Spectrum: A Narrative Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Hidden Relationship between Intestinal Microbiota and Immunological Modifications in Preeclampsia Pathogenesis

by
Enrica Zambella
1,
Beatrice Peruffo
1,
Alice Guarano
2,
Annalisa Inversetti
1,3 and
Nicoletta Di Simone
1,3,*
1
Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy
2
Humanitas San Pio X, Via Francesco Nava 31, 20159 Milan, Italy
3
IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(18), 10099; https://doi.org/10.3390/ijms251810099
Submission received: 15 August 2024 / Revised: 16 September 2024 / Accepted: 18 September 2024 / Published: 20 September 2024
(This article belongs to the Special Issue Physiology and Pathophysiology of Placenta 2.0)

Abstract

:
Preeclampsia is a multifactorial gestational syndrome characterized by increased blood pressure during pregnancy associated with multiorgan involvement. The impact of this disease on maternal and neonatal health is significant, as it can lead to various fetal comorbidities and contribute to the development of maternal comorbidities later in life. Consistent evidence has shown that the microbiota acts as a regulator of the immune system, and it may, therefore, influence the development of preeclampsia by modulating immune factors. This narrative review aims to investigate the role of the immune system in the pathogenesis of preeclampsia and to summarize the most recent literature on the possible link between preeclampsia and alterations in the intestinal microbiota. To this end, we conducted a literature search, aiming to perform a narrative review, on PubMed and Embase from January 1990 to March 2024, focusing on the latest studies that highlight the main differences in microbial composition between patients with and without preeclampsia, as well as the effects of microbial metabolites on the immune system. From the review of 28 studies assessing the intestinal microbiota in preeclamptic women, preeclampsia could be associated with a state of dysbiosis. Moreover, these patients showed higher plasmatic levels of endotoxin, pro-inflammatory cytokines, and T helper 17 cells; however, the findings on specific microbes and metabolites that could cause immune imbalances in preeclampsia are still preliminary.

1. Introduction

Hypertensive disorders of pregnancy (HDPs) represent an important topic in the obstetrical field. HDPs affect a variable number of pregnancies worldwide, with an incidence range that goes from 3% to 8–10% [1]. These conditions lead to a higher risk of adverse pregnancy outcomes like pre-term birth, intrauterine growth restriction (IUGR) [2], defined as estimated fetal weight (EFW)/fetal abdominal circumference (AC) below the 3rd percentile or as a drop in AC or EFW of >2 quartiles or >50 percentiles and abnormal Doppler velocimetry of the uteroplacental and fetoplacental circulations [3], and lastly maternal and/or perinatal death [1,4]. Indeed, HDPs still represent the sixth cause of maternal death in the United States (6.3% in the period 2017–2019) [5] and the fifth in Italy [6]. In the group of HDPs, preeclampsia (PE) is the most severe disease, characterized by de novo hypertension after 20 weeks’ gestation (systolic blood pressure (SBP) of ≥140 mmHg and/or diastolic (DBP) of ≥90 mmHg on two measurements executed at least 4 h apart, or SBP of ≥160 mmHg and/or DBP of ≥100 mmHg collected on a single measurement) and proteinuria (>300 mg/24 h or protein/creatinine ratio of ≥0.3 mg/dL). In the absence of proteinuria, at least one of these new-onset signs or symptoms should be identified in addition to hypertension: thrombocytopenia, liver dysfunction, renal insufficiency, pulmonary edema, and headache unresponsive to medication and not justified by alternative diagnoses or visual disturbances [1,7]. The International Society for the Study of Hypertension in Pregnancy (ISSHP) also includes uteroplacental dysfunction (e.g., placental abruption, fetal growth restriction defined as an estimated fetal weight <10th percentile, abnormal umbilical artery Doppler, or intrauterine fetal death) as a diagnostic criterion for PE [8]. Preeclampsia represents the second cause of maternal death after postpartum hemorrhage in developing countries [9] and, according to Ending Preventable Maternal Mortality (EPMM) strategies stated by the WHO, it is one of the highly preventable causes of obstetric adverse outcomes [10]. For this reason, extending our knowledge of the possible risk factors and developing a proper screening strategy is essential to identify and treat this condition in order to prevent most related deaths [11]. Moreover, over the past decade, PE has been associated with a higher occurrence of chronic diseases later on in both the mother’s and offspring’s lives [12,13]. There is increasing evidence about the possible role of altered immune regulation in preeclampsia, related to defective maternal–fetal immune tolerance and higher prevalence in patients affected by autoimmune diseases [14]. Hence, more systemic and complex mechanisms involved in preeclampsia pathogenesis need to be studied.
The gut microbiota is a complex association of microorganisms’ ecosystems, including bacteria, fungi, archaea, viruses, and protozoa, able to resist unfavorable gastrointestinal conditions in a strict symbiotic relationship with the host [15]. These microorganisms and their collective genomes (microbiome) have several essential metabolic functions (metabolome); therefore, the gut microbiota is also defined as the postnatally acquired “metabolic organ” [16]. The gut microbiota has an important influence on many developmental processes and functions of the host, in particular on the immune system. It is well known that the microbiota enhances the integrity of epithelial cells as a barrier against pathogens, and it is also involved in priming immune cells through the production of immunomodulatory metabolites [17,18,19]. In this regard, short-chain fatty acids (SCFAs) are metabolites produced by gut bacteria during fermentation of dietary fiber, and it has been demonstrated that they can influence the balance of pro-inflammatory and anti-inflammatory cytokines and can have a potential role in Treg cell development in offspring [20]. Moreover, considering that microbiota-derived metabolites, like lipopolysaccharide (LPS) and peptidoglycans, can cross the syncytiotrophoblast barriers, the development of immune cells starts in the womb, and this process can be influenced by the maternal microbiota [21]. In a study using 16S rRNA sequencing on human fetal meconium at 18–23 weeks of gestation, the presence of Micrococcaceae was linked to the development of specific intestinal memory T cells and processes like Toll-like receptor signaling, phagolysosome function, immune cell chemoattraction, and macrophage inhibition [22]. Similarly, another study demonstrated the presence of bacterial cocci within the fetal gut lumen at 14 weeks of gestation, able to elicit a memory response by the mesenteric lymph node T cells [23]. Structural changes in commensal microbial communities lead to dysbiosis, recently associated with several systemic inflammatory syndromes, such as inflammatory bowel diseases (IBDs) or asthma, and chronic diseases like hypertension, obesity, diabetes mellitus and metabolic syndrome [24]. These chronic diseases are well-known risk factors for PE development, and they also account for many of the long-term outcomes of PE in women’s lives [25]. In early pregnant obese and overweight women, specific gut microbiome compositions, such as higher levels of Collinsella and Coprococcus, have been linked to increased insulin resistance and abnormal lipid metabolism [26].
In this delicate balance, the microbiota may play a role in the pathogenesis of immune diseases, since a dysregulation occurring at any level of this complex process could potentially cause an immunological disorder. The impact of microbiota on the maternal immune system [26,27] as well as the interaction between intestinal dysbiosis and metabolic diseases leads to the hypothesis of the key role of microbiota in PE pathogenesis [28]. The principal aim of this narrative review is to describe the potential association between dysbiosis and preeclampsia compared to controls, the role of metabolomic analyses, and the clinical impact of microbial dysbiosis on maternal health.

2. Methods

We conducted a literature search from January 1990 to March 2024 across two electronic databases: PubMed and Embase. We used various combinations of the following keywords to analyze relevant articles: “gut microbiota” or “intestinal microbiota” or “gut metabolome” or “intestinal metabolome” and “preeclampsia” or “gestational hypertension” or “hypertensive pregnancy disorders”. We included randomized controlled trials, cohort studies, case–control studies, and cross-sectional studies in our review. Additionally, reference lists of retrieved reports, review articles, and meta-analyses were evaluated to identify potential additional studies. We excluded case reports, letters and non-English language publications. Two authors (E.Z. and B.P.) independently conducted electronic searches and analyzed a total of 264 studies.

3. Preeclampsia and Its Pathogenesis

The primary mechanisms behind PE are still partially unknown, even if some steps towards understanding its pathophysiology have been made in the last decade [29,30,31].
Placentation has a key role: cytotrophoblast cells invade the uterine myometrium (invasive phenotype) and shape efficient vascular anastomoses between the spiral arteries to support the placental circulation [32,33]. It has been demonstrated that in HDP/PE disease, cytotrophoblasts do not acquire the physiological invasive phenotype due to trophoblast immaturity, leading to an inadequate vascular invasion and narrow blood vessels. Therefore, the development of abnormal vessels results in endothelial damage and leads to insufficient oxygen transfer, placental ischemia, and consequent increased reactive oxygen species (ROS) production [34]. These alterations cause an imbalance in the immune tolerance mechanisms, causing the secretion of inflammatory molecules and an alteration between angiogenic and anti-angiogenic factor proportions. The increase in pro-inflammatory and anti-angiogenic proteins in maternal blood circulation represents the basis of the clinical syndrome, with its typical systemic signs and symptoms [35].
PE is commonly classified into two different subtypes according to a worldwide consensus, which differ in incidence, pathogenesis, and maternal–fetal outcomes: early-onset preeclampsia (EOPE), before 34 weeks of gestation, which accounts for a minority of all cases (5–20%) and late-onset preeclampsia (LOPE), starting after 34 weeks (80% of all cases) [36,37].
The variability in symptom severity, the effectiveness of preventive treatments, and long-term outcomes differ between the two subtypes as the result of different pathogenic mechanisms [38]. EOPE’s etiology has been directly linked to early defects in placental development, leading to reduced uteroplacental perfusion, hypoxia, and oxidative stress [39].
Many studies demonstrated that an adverse uterine environment leads to an increased risk for IUGR, low birth weight, neonatal death, congenital heart defects, and more [37,40,41,42].
On the other hand, LOPE is identified as “maternal preeclampsia” due to maternal endothelial dysfunction [43]: this condition leads to an imbalance between angiogenic and anti-angiogenic proteins [44] and it is related to a later maternal predisposition to metabolic disorders, such as dyslipidemia, long-term diabetes [13,38], and cardiovascular disorders, like essential hypertension and coronary heart disease [45,46]. In a reciprocal way, environmental factors like chronic hypertension, obesity, and diabetes significantly increase the likelihood of developing preeclampsia [47,48].
Placental growth factor (PIGF), a pro-angiogenic molecule, and soluble FMS-like tyrosine kinase-1 (sFlt1), an anti-angiogenic factor, are two of the most-studied biomarkers involved in PE [49,50,51]. Low levels of circulating PIGF and high levels of sFlt-1 are normally found in patients with PE [52,53], leading to an elevated ratio, recently studied as a predictive biomarker of PE [54].
Moreover, there are a few more candidate biomarkers for PE prediction: first-trimester pregnancy-associated plasma protein A (PAPPA-A) [55], Pentraxin 3 [56], microRNAs [57] and some circulating HDP-associated peptides (P-2081, P-2091, P-2127, P-2209, P-2378, P-2858, and P-3156) [58,59].

3.1. Placental Dysfunction and the “Two-Stage Model”

Placental dysfunction, and, in particular, the “two-stage theory”, has been one of the most studied hypotheses behind PE, although the comprehension of PE pathogenesis is still limited. Here, we report an overview of the “two-stage theory”:
  • Stage 1 (placental stage): The initial stage involves poor placentation, where the extravillous trophoblasts fail to properly invade and remodel maternal spiral arteries [60]. This vascular transformation is often incomplete or absent, particularly in the junctional zone of the myometrium, in women who subsequently develop PE. Consequently, this results in inadequate blood flow to the placenta and ischemia, triggering the release of anti-angiogenic factors such as sFlt-1 and PlGF [61,62] but also of immune mediators and ROS [47,63].
  • Stage 2 (maternal symptoms): These placental factors enter the maternal bloodstream, leading to systemic endothelial dysfunction and the onset of clinical symptoms, such as hypertension and proteinuria [64,65].

3.2. Angiogenic Imbalance

One of the hallmarks of preeclampsia is an imbalance between pro-angiogenic and anti-angiogenic factors. In healthy pregnancies, pro-angiogenic factors like VEGF and PlGF promote blood vessel formation and placental growth. On the other hand, PlGF levels are significantly reduced in PE, while anti-angiogenic factors such as sFlt-1 and soluble endoglin (sEng) are elevated. This imbalance disrupts normal vascular function, contributing to widespread endothelial damage and the development of systemic hypertension [64,66]. The sFlt-1/PlGF ratio is increasingly popular in clinical settings to predict the onset and severity of preeclampsia [48,63,67]. Furthermore, therapeutic strategies targeting this imbalance, such as the use of VEGF agonists or neutralizing antibodies against sFlt-1, are being explored as potential treatments [47].

3.3. Endothelial Dysfunction and Systemic Oxidative Stress

Another pathogenetic hypothesis involves endothelial dysfunction. The endothelium is damaged by the combination of oxidative stress, inflammation, and anti-angiogenic factors [60]. This results in impaired vascular tone regulation [63,66], contributing to the development of a PE-associated prothrombotic state [68].

3.4. Genetic and Epigenetic Aspects

The genetic side has been investigated to assess whether there is a predisposition to PE. Variants in genes related to angiogenesis, immune regulation, and placental development have been associated with an increased risk of the disorder [64]. Among them, polymorphisms in the FLT1 gene (which encodes sFlt-1) are linked to higher levels of sFlt-1 and a higher risk of developing PE [69].
Moreover, epigenetic modifications such as altered microRNA (miRNA) expression also influence transcriptional activity related to trophoblast invasion and vascular function [70]. For example, chromosome 19 microRNA cluster (C19MC) is supposed to be heavily involved in PE. Dysregulation of miR-519d, miR-517-5p, and others from this cluster affects angiogenesis: miR-519d suppresses trophoblast invasion, while miR-517-5p impairs syncytiotrophoblast differentiation, leading to a decreased ability of cells to invade and proliferate [70,71].
Also, several long non-coding RNAs (lncRNAs) have been associated with PE. For instance, IGFBP1 is linked to placental insufficiency, while EGFR-AS1 influences cell proliferation by modulating the JAK-STAT signaling pathway [72,73].
Lastly, circular RNA (circRNA) has been suggested to participate in the progression of EOPE. CircRNA_06354 was recently identified by Zhang et al. as playing a role in angiogenesis via the hsa-miR-92a-3p/VEGF-A pathway; therefore, a dysregulated expression impairs trophoblast invasion into spiral arteries, contributing to EOPE [74].
These aspects highlight a complex interaction between genetic and environmental factors in determining the pathogenetic mechanisms underlying PE [64].

4. Immune System and Immune Dysfunctions in Preeclampsia

Pregnancy can physiologically develop when proper immune tolerance between mother and fetus occurs, in order to prevent fetal rejection [51,75]. Natural killer cells (NK cells) and T cells located in the decidua play a crucial role in the maternal immune tolerance to the fetus through well-known strategies of antigen presentation, inhibitory cytokine production, and cytolysis of targeted cells [76,77,78]. The conflict between maternal and paternal genes leads to higher NK cell activity, decreased regulatory T-cell (Treg) number, and other changes in immune mediators’ production [79]. The dysregulation of all these mechanisms prevents the establishment of a pregnancy-favorable environment and leads to various pregnancy disorders, such as preeclampsia and recurrent spontaneous abortion [78,80].
Indeed, according to recent studies, an abnormal shift to T helper (Th1) cell phenotype, higher doses of pro-inflammatory molecules and higher levels of NK cells were found in PE patients [76,81]. This inflammatory environment supports the production of pro-inflammatory cytokines, autoantibodies, and ROS, with a worsening effect on the ongoing ischemic process, as previously described [51]. A schematic representation of these changes is shown in Figure 1.
The inflammatory response to oxidative stress is self-perpetuating and leads to a chronic condition fundamental for PE pathogenesis. This compromised self-regulation process resembles the immune dysfunctions in autoimmunity and is also involved in some other pregnancy complications, such as gestational diabetes mellitus (GDM) and pre-term delivery [13]. The existing connection between pregnancy and autoimmune diseases is extremely complex: in patients affected by autoimmune diseases, the abnormal activation of maternal innate and adaptive immune systems can interfere with placentation and tolerance of the fetal semi-allograft, with a higher risk of PE, IUGR, prematurity, congenital heart block, and autoimmune diseases in the offspring and even stillbirth [14,82,83,84,85]. It is well known that systemic lupus erythematosus (SLE), antiphospholipid syndrome (APS) (mostly the active disease), and hypothyroidism are related to an increased risk of PE [86,87,88]. Furthermore, PE patients showed higher levels of antibodies against angiotensin II type 1 receptor (AT1-AAs) [89]: these antibodies show a persistent effect on vasoconstriction and lead to endothelial damage and insufficient trophoblast invasion, typical conditions of PE pathogenesis [90]. Furthermore, these autoantibodies could increase sFlt-1 secretion, but it is still uncertain whether these anomalies are pathogenic or epiphenomena of PE [91]. Finally, prior exposure to paternal/fetal antigens could have a protective role for PE: the maternal immune system’s progressive exposure to paternal semen leads to a persistent tolerance toward the fetal semi-allograft until pregnancy term [2,92,93]. For this reason, long inter-pregnancy interval (>10 years), pregnancies with different paternity, and nulliparity could represent possible PE risk factors [93,94,95]. It has also been noticed that PE incidence is higher in pregnancy conceived via intra-cytoplasmic sperm injection (ICSI) [96,97,98] or with an egg and/or sperm donor [95,99].

5. Gut Microbiota and Preeclampsia

In consideration of the increasing evidence of the microbiota’s impact on immune regulation, the hypothesis on the possible effect of intestinal dysbiosis and consequent altered immune modulation on PE development could be an interesting and new point of view. Indeed, this represents a very recent field of research, and the current literature on this topic is constantly growing. We found 28 relevant reports assessing the differences among PE patients and controls of gut flora composition, including bacteria, viruses, and fungi (i.e., mycobiome) (Table 1) and its metabolites, and lastly the possible causative role of the microbiota, as described in the following paragraphs.

5.1. Differences in Gut Microbiota Composition in PE Patients

One of the first studies evaluating microbial composition in PE patients was conducted by Liu et al., who demonstrated in PE patients an overall increase in pathogenic bacteria, Clostridium perfringens and Bulleidia moorei, and a reduction in propionic-producing bacteria, known as probiotic bacteria, such as Coprococcus catus [100]. Similar results on PE-induced dysbiosis were found in another cohort of patients, showing an inverse correlation between Lactobacillus abundance and SBP, DBP, and proteinuria [101]. Furthermore, a recent pilot study confirmed a lower abundance of Coprococcus in HDP patients, while a higher abundance of pathogenic bacteria (Rothia, Actinomyces, and Enterococcus) and Escherichia as the dominant bacterium were found in the case group [102]. Other studies demonstrated significant alterations in microbial composition during the third trimester compared to the second trimester in PE patients, with a reduction in Firmicutes and an increase in Bacteroidetes, Proteobacteria, Blautia, and Faecalibacterium [103,104]. Similarly, Wu et al. evaluated the longitudinal variation of gut microbiota in pregnant patients, and the group who would later develop PE or gestational hypertension was significantly enriched for Polycyclovorans and Pelomonas at the end of the first trimester and at term of pregnancy [105]. Then, Lv et al. found a significant enrichment of Blautia, Ruminococcus2, Bilophila, and Fusobacterium and a significant depletion of Faecalibacterium, Gemmiger, Akkermansia, Dialister, and Methanobrevibacter in antenatal PE samples; these taxonomic alterations were similar to postpartum samples [106]. Similarly to another study, Blautia and Ruminococcus2 were positively correlated with obesity, dyslipidemia, glycosyltransferases, biotin metabolism, and oxidative phosphorylation pathways [106,107]. Afterwards, the same group found an enrichment of other species belonging to Pauljensenia and Collinsella in women with early-onset PE through deep shotgun metagenomic sequencing, showing an improvement in the accuracy of species identification with this technique [108]. Different results were obtained by Zhao et al., who found lower levels of Bacteroidetes, Ruminococcus, and Oscillospira and an increasing ratio of Firmicutes and Bacteroidetes in the PE group compared to controls [109]. Although fungi account for about 0.1% of the gut microbiome, they still represent a substantial part of the microbiome biomass. For this reason, the mycobiome, the fungal component of the gut flora, was also evaluated. Meijer et al. found a similar bacterial microbiome composition in the PE group compared to previous studies, but, analyzing the mycobiome, they found no significant differences in the abundance of Saccharomycetes, Dothideomycetes and Sordariomycetes classes between the case and control groups, except for ASV-36, corresponding to the genus Peniophora [110]. In conclusion, alpha diversity (i.e., observed richness of taxa of an average sample) is significantly lower in PE patients, while, according to a recent meta-analysis, a comparison of relative abundance of Bacteroidetes, Firmicutes, Actinobacteria, and Proteobacteria showed no significant results [111].

5.2. The Causative Role of Microbiota in PE Pathogenesis

It is well known that there is a different intestinal microbial composition in patients affected by hypertension. Indeed, several cross-sectional studies in humans found higher abundances of Gram-negative bacteria, including Klebsiella, Parabacteroides, Desulfovibrio, and Prevotella, and lower abundances of SCFA-producing bacteria, including Ruminococcaceae, Roseburia, and Faecalibacterium spp. [112]. Similarly, in PE patients, SBP and DBP significantly acted on microbial diversity and structure compared to other clinical parameters such as age, body mass index (BMI), gestational age, fasting glucose, and glycosylated hemoglobin [108]. To understand the causative role of the microbiota in the pathogenesis of this disease, Chen et al. performed a fecal microbiota transplantation (FMT) in an antibiotic-treated mouse model: they showed an increase in SBP, proteinuria, histopathological structural alterations in placenta, Th17, and higher intestinal permeability in mice transplanted with PE microbiota [25]. Similar data were found in other studies, with an increase in peripheral Th17 cells, increased serum levels of tumor necrosis factor α (TNF-α), interleukin 17 (IL-17), IL-1β, IL-6, and LPS in PE patients and, similarly, in a preeclamptic mouse model after FMT [28,109,113]. Lastly, four different studies used mendelian randomization (MR) to understand the effect of microbial changes on PE. Three of these studies showed a protective role against HDPs and PE of Bifidobacterium, Collinsella, Enterorhabdus, Eubacterium, Lachnospiraceae, Tyzzerella 3, and Methanobrevibacter, while LachnospiraceaeUCG010, Olsenella, RuminococcaceaeUCG009, and Ruminococcus2 were related to a significantly increased risk for gestational hypertension [105,114,115]. Similarly, another study showed a potential protective role of Streptococcus by affecting TNF-α, a protective effect of the Enterobacteriales order, and a pathological role of Olsenella, through modulation of unsaturated fatty acid biosynthesis with secondary effects on vasoconstriction, oxidative stress, inflammation, and coagulation [28].

5.3. Metabolomic Analysis of Intestinal Microbiota PE Patients

There is increasing evidence on the effects of microbial metabolites on the immune and endocrine systems, and these variations may be associated with obesity, elevated blood pressure, and other comorbidities [116]. Recently, Cui et al. reviewed seven human cohort-controlled trials, six of them comparing gut microbiota composition and its association with SCFAs between healthy pregnant women and patients affected by PE [117]. These studies showed that SCFAs are strictly related to gut microbiota abundance; specifically, serum acetate levels seemed to be protective against PE. Moreover, PE patients had significantly decreased serum acetate, propionate, and butyrate concentrations, related to the concomitant reduction in Coprococcus [20,118,119]. It was recently demonstrated that Firmicutes had a lower abundance in PE patients, and SCFAs produced by this phylum may influence blood pressure and renal function [120]. Another recent study evaluated the variation of SCFAs in serum samples of 38 PE patients and 29 healthy pregnant women after 28 gestational weeks: significantly increased levels of acetic acid, propionic acid, isobutyric acid, and valeric acid were found in the PE group, and these metabolites were positively correlated to blood pressure and urea levels and negatively correlated with the gestational week and fetal birth weight [121]. Additionally, according to the results of ROC curve analyses, the authors demonstrated the possible use of these SCFAs as serum biomarkers for the diagnosis of PE, especially isobutyric acid, which showed the highest sensibility and specificity [121]. On the other hand, bioactive metabolites derived from the gut microbiota, such as trimethylamine N-oxide (TMAO), were found to be related to higher levels of carnitine and choline, sterol and cholesterol metabolism, and atherosclerotic-like lesions in the spiral arteries in placentas obtained from PE patients [122,123]. Furthermore, lower plasmatic levels of betaine and higher plasmatic levels of choline were associated with higher HDP risk in the early second trimester of pregnancy [124].
Table 1. The intestinal microbial composition in patients affected by preeclampsia (PE) according to the most recent literature.
Table 1. The intestinal microbial composition in patients affected by preeclampsia (PE) according to the most recent literature.
Author, YearCountryStudy DesignMethodsNumber and Type of PatientsNumber of Stool SamplesPhylum LevelsClinical Impact on Maternal and Neonatal Health
Liu et al., 2017 [100]ChinaCase–control16S rDNA gene amplicon sequencing26 PE patients (11 mild symptoms and 15 severe symptoms) vs. 3 controls100Similar composition in the dominant phyla Increased level of Cyanbacteria in PE groupN/A
Lv et al., 2019 [106]ChinaCase–control16S
rRNA gene amplicon sequencing
78 PE patients vs. 72 controlsAntepartum: 48 PE vs. 51 controls; 1 week postpartum: 35 PE vs. 17 controls; 6 weeks postpartum: 18 PE vs. 11 controlsSimilar composition in the dominant phylaPositive correlation between DBP and SBP levels and Anaerococcus, Ruminococcus2, Fusobacterium,
and Oribacterium, positive correlation among birth weight and PE-depleted genera.
Wang et al., 2020
[103]
ChinaNested case–control16S rRNA gene amplicon sequencing25 PE patients vs. 25 controls20–24 weeks gestation: 25 PE vs. 25 controls; 32–34 weeks gestation: 15 PE vs. 22 controlsIn T2, no significant differences between PE group and controls; in T3, significantly lower levels of Firmicutes, significantly higher levels of Bacteroidetes and Proteobacteria in PE group vs. controlsN/A
Huang et al., 2021
[101]
ChinaCase–control16S rRNA gene amplicon sequencing26 PE patients, 25 APG § patients vs. 28 controls and 21 healthy non-pregnant patientsN/ASimilar composition in the dominant phyla Significantly increased percentage of TM7 in the APG groupSignificant correlation of g_1_68,
g_Porphyromonas, g_Mobiluncus,
and g_Lactococcus
with SBP and DBP.
Significant inverse correlation among g_Lactobacillus and SBP, DBP and UP.
Miao et al., 2021
[107]
ChinaCase–control16S rRNA gene amplicon sequencing12 PE patients vs. 8 controls20Slightly increased levels of Firmicutes and significantly decreased levels of Actinobacteria in PE groupPositive correlation between Blautia and maternal age, pregestational weight, HCT, PCR, TGs, and LDL. Positive correlation between Ruminococcus and pregestational weight, pregestational BMI, antepartum weight, antepartum BMI, LBP, and TGs. Negative correlation between Bifidobacterium and SBP, DBP, cholesterol, AST, and TGs.
Lv et al., 2022
[108]
ChinaCase–controlShotgun metagenomic sequencing40 PE patients vs. 37 controls77Major phyla included Firmicutes, Actinobacteria, and Bacteroidetes in both groupsN/A
Geldenhuys et al., 2022
[104]
South AfricaCase–control16S rRNA gene amplicon sequencing10 PE patients vs. 11 controls6 PE vs. 8 controlsNo significant difference between two groupsN/A
Wu et al., 2023
[105]
ChinaNested case–control16S rRNA gene amplicon sequencing29 GH patients and 17 PE patients vs. 30 controls17 PE and 29 GH vs.
30 controls
No significant difference between GH, PE, and control groups during the early, middle, and late trimesterIn the early trimester, positive correlation among Polycyclovorans and DBP. In the middle trimester, negative correlation between Acidibacter, Amphiplicatus, and Shewanella and DBP. In the late trimester, Pelomonas positively correlated with SBP, Thermomonas negatively correlated with DBP.
Wu et al., 2023
[102]
ChinaCase–control16S rRNA gene amplicon sequencing14 PE patients vs. 9 controlsN/ADetection of Euryarchaeota only in controlsN/A
Zhao et al., 2023
[109]
ChinaCase–control16S rRNA gene amplicon sequencing41 PE patients vs. 45 controls86Significantly lower relative abundance of Bacteroidetes and significantly higher relative abundance of Proteobacteria in PE groupN/A
Abbreviations: rRNA: ribosomal RNA; PE: preeclampsia; N/A: not available; T2/T3: second/third trimester; APG: abnormal placental growth; GH: gestational hypertension; DBP: diastolic blood pressure; SBP: systolic blood pressure; UP: urinary protein concentration; HCT: hematocrit; PCR: C-reactive protein; TGs: triglycerides; LDL: low-density lipoprotein cholesterol: LBP: lipopolysaccharide-binding protein; BMI: body mass index; AST: aspartate aminotransferase. § Abnormal placental growth diagnosis: abnormal PIGF Multiple of the Median (MoM) values in early and middle pregnancy.

6. Discussion

The intestinal microbiota is a highly complex system involved in various metabolic and immunological functions. It changes dynamically from childhood to adulthood, including during motherhood. During pregnancy, the gut microbiota shifts toward the end of gestation, with an increase in Proteobacteria and Actinobacteria, and a decrease in SCFAs. These changes appear to be related to physiological processes such as weight gain, low-grade inflammation, and insulin resistance [125]. The intestinal microbiota can also be influenced by factors like medication (e.g., antibiotics), diet, physical activity, and lifestyle, leading to the high heterogeneity of studies evaluating the microbiota in pregnant women. In this review of the most recent literature, preeclampsia (PE) was associated with intestinal dysbiosis compared to healthy controls. However, larger sample sizes and more standardized study protocols are needed to reduce the influence of confounding factors. Due to these limitations, a specific combination of microbes related to this disease has not been consistently reported in studies. Some of the selected studies evaluated the impact of these microbial changes on clinical parameters used for diagnosing preeclampsia [101,105,106,107], while others examined the correlation between gut dysbiosis and inflammatory or intestinal permeability markers [25,28,109,113]. Patients with PE showed higher plasmatic levels of LPS, elevated levels of pro-inflammatory Th17 cells, and increased pro-inflammatory cytokines. In a mouse model, LPS was involved in key pathways for blood pressure regulation, leading to decreased baroreflex sensitivity, increased Toll-like receptor (TLR) and TNF-alpha expression in the paraventricular nucleus, elevated heart rate, and increased norepinephrine levels [126]. Additionally, mice transplanted with PE microbiota exhibited lower expression of Claudin-4 and Occludin genes, and a marked reduction in zonula occludens-1 and -2, Claudin-4, and Occludin proteins in the colon [25]. These proteins form intestinal tight junctions, and zonulin, a protein secreted by the basal lamina of the intestinal epithelium, activates intracellular signaling pathways involved in paracellular permeability. Zonulin is one of the most-studied markers of intestinal permeability [127]. Increased gut permeability is linked to systemic translocation of metabolites and endotoxins, potentially amplifying intestinal permeability. Microbiota dysbiosis can induce low-grade chronic inflammation, impairing intercellular connections by downregulating tight junctions, and causing a “leaky gut” [128], a condition associated with increased endometrial inflammation in patients suffering from recurrent pregnancy loss [129]. According to the selected studies, preeclamptic patients exhibited gut microbial shifts, higher levels of inflammatory markers, and increased intestinal permeability, leading to the hypothesis that a leaky gut could be a potential pathological pathway involved in the development of preeclampsia. However, the current evidence is insufficient to establish a cause–effect relationship, and further studies are required to confirm this hypothesis. Lastly, several studies have annotated the functional characteristics of microbiota in PE using the KEGG (Kyoto Encyclopedia of Genes and Genomes) database [130]. Enzymes involved in transport systems (e.g., bacitracin/lantibiotics, rhamnose, and maltooligosaccharide transport), signal transduction, environmental adaptation, and cell motility were significantly upregulated in the PE group, suggesting that these mechanisms could contribute to intestinal dysbiosis [102]. From metabolomic analyses, the most consistent evidence identified isobutyric acid and trimethylamine N-oxide (TMAO) as early predictors of PE [121,123]. Trimethylamine (TMA) is mainly produced by the families Clostridia and Enterobacteriaceae, with the degradation of nutrients commonly found in dietary products like meat and eggs, and then it is oxidized by the hepatic enzymes. TMAO has various effects on inflammation (e.g., increasing TNF-α and IL-1B, decreasing IL-10), cholesterol metabolism, and thrombosis (e.g., inducing platelet hyperreactivity) [112]. These findings have been raised from few studies, so it is necessary to develop further associations between these metabolites and preeclampsia to introduce them into clinical practice.

7. Conclusions

The analysis of the intestinal microbiota in preeclampsia is a growing area of research. Over the past few decades, studies have increasingly demonstrated a connection between gut dysbiosis and this disease. However, it is essential to establish a microbial baseline in healthy pregnant women to allow for better comparisons between cases and controls and to reduce the heterogeneity of studies. Understanding the immunological impact of microbiota in preeclampsia could be a crucial step toward a more comprehensive study of its pathogenesis. Randomized clinical trials are necessary to provide stronger evidence on the relationship between microbiota and preeclampsia and to identify metabolic features as early diagnostic markers for PE. In this way, earlier preventive strategies could be developed to reduce the disease’s impact on maternal and fetal outcomes.

Author Contributions

Writing—original draft preparation, E.Z. and B.P.; writing—review and editing, A.I., E.Z., A.G. and B.P.; supervision, N.D.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created.

Acknowledgments

Figures were created with Biorender.com, accessed on 20 February 2024.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Gestational Hypertension and Preeclampsia: ACOG Practice Bulletin, Number 222. Obstet. Gynecol. 2020, 135, E237–E260. [CrossRef] [PubMed]
  2. Dimitriadis, E.; Rolnik, D.L.; Zhou, W.; Estrada-Gutierrez, G.; Koga, K.; Francisco, R.P.V.; Whitehead, C.; Hyett, J.; da Silva Costa, F.; Nicolaides, K.; et al. Pre-eclampsia. Nat. Rev. Dis. Primers 2023, 9, 8. [Google Scholar] [CrossRef] [PubMed]
  3. Lees, C.C.; Stampalija, T.; Baschat, A.A.; da Silva Costa, F.; Ferrazzi, E.; Figueras, F.; Hecher, K.; Poon, L.C.; Salomon, L.J.; Unterscheider, J. ISUOG Practice Guidelines: Diagnosis and management of small-for-gestational-age fetus and fetal growth restriction. Ultrasound Obstet. Gynecol. 2020, 56, 298–312. [Google Scholar] [CrossRef] [PubMed]
  4. Poon, L.C.; Shennan, A.; Hyett, J.A.; Kapur, A.; Hadar, E.; Divakar, H.; McAuliffe, F.; da Silva Costa, F.; von Dadelszen, P.; McIntyre, H.D.; et al. The International Federation of Gynecology and Obstetrics (FIGO) initiative on pre-eclampsia: A pragmatic guide for first-trimester screening and prevention. Int. J. Gynecol. Obstet. 2019, 145 (Suppl. S1), 1–33. [Google Scholar] [CrossRef] [PubMed]
  5. Hoyert, D.L. Maternal Mortality Rates in the United States, 2021; NCHS Health E-Stats: Hyattsville, MD, USA, 2023. [Google Scholar] [CrossRef]
  6. Dell’Oro, S.; Maraschini, A.; Lega, I.; D’Aloja, P.; Andreozzi, S.; Donati, S. Primo rapporto Itoss Sorveglianza della Mortalità Materna. Italian Obstetric Surveillance System (ItOSS). Not Ist Super Sanità 2019, 32 (Suppl. S1), 69. [Google Scholar]
  7. Ives, C.W.; Sinkey, R.; Rajapreyar, I.; Tita, A.T.N.; Oparil, S. Preeclampsia-Pathophysiology and Clinical Presentations: JACC State-of-the-Art Review. J. Am. Coll. Cardiol. 2020, 76, 1690–1702. [Google Scholar] [CrossRef]
  8. Magee, L.A.; Brown, M.A.; Hall, D.R.; Gupte, S.; Hennessy, A.; Karumanchi, S.A.; Kenny, L.C.; McCarthy, F.; Myers, J.; Poon, L.C.; et al. The 2021 International Society for the Study of Hypertension in Pregnancy classification, diagnosis & management recommendations for international practice. Pregnancy Hypertens 2022, 27, 148–169. [Google Scholar] [CrossRef]
  9. Lawrence, E.R.; Klein, T.J.; Beyuo, T.K. Maternal Mortality in Low and Middle-Income Countries. Obstet. Gynecol. Clin. 2022, 49, 713–733. [Google Scholar] [CrossRef]
  10. World Health Organization. Trends in Maternal Mortality 2000 to 2020 Estimates by WHO, UNICEF, UNFPA, World Bank Group and UNDESA/Population Division; World Health Organization: Geneva, Switzerland, 2023. [Google Scholar]
  11. Say, L.; Chou, D.; Gemmill, A.; Tunçalp, Ö.; Moller, A.B.; Daniels, J.; Gülmezoglu, A.M.; Temmerman, M.; Alkema, L. Global causes of maternal death: A WHO systematic analysis. Lancet Glob. Health 2024, 2, 2014. [Google Scholar] [CrossRef]
  12. Turbeville, H.R.; Sasser, J.M. Preeclampsia beyond pregnancy: Long- term consequences for mother and child. Am. J. Physiol. Ren. Physiol. 2020, 318, 1315–1326. [Google Scholar] [CrossRef]
  13. Bokuda, K.; Ichihara, A. Preeclampsia Up to Date—Whats Going on? Springer: Berlin/Heidelberg, Germany, 2023. [Google Scholar] [CrossRef]
  14. Canti, V.; Castiglioni, M.T.; Rosa, S.; Franchini, S.; Sabbadini, M.G.; Manfredi, A.A.; Rovere-Querini, P. Pregnancy outcomes in patients with systemic autoimmunity. Autoimmunity 2012, 45, 169–175. [Google Scholar] [CrossRef] [PubMed]
  15. Structure, function and diversity of the healthy human microbiome. Nature 2012, 486, 207–214. [CrossRef] [PubMed]
  16. Baquero, F.; Nombela, C. The microbiome as a human organ. Clin. Microbiol. Infect. 2012, 18, 2–4. [Google Scholar] [CrossRef] [PubMed]
  17. Cai, R.; Cheng, C.; Chen, J.; Xu, X.; Ding, C.; Gu, B. Interactions of commensal and pathogenic microorganisms with the mucus layer in the colon. Gut Microbes 2020, 11, 680–690. [Google Scholar] [CrossRef] [PubMed]
  18. Inversetti, A.; Zambella, E.; Guarano, A.; DellAvanzo, M.; Di Simone, N. Endometrial Microbiota and Immune Tolerance in Pregnancy. Int. J. Mol. Sci. 2023, 24, 2995. [Google Scholar] [CrossRef]
  19. DIppolito, S.; Di Nicuolo, F.; Pontecorvi, A.; Gratta, M.; Scambia, G.; Di Simone, N. Endometrial microbes and microbiome: Recent insights on the inflammatory and immune “players” of the human endometrium. Am. J. Reprod. Immunol. 2018, 80, e13065. [Google Scholar] [CrossRef]
  20. Hu, M.; Eviston, D.; Hsu, P.; Mariño, E.; Chidgey, A.; Santner-Nanan, B.; Wong, K.; Richards, J.L.; Yap, Y.A.; Collier, F.; et al. Decreased maternal serum acetate and impaired fetal thymic and regulatory T cell development in preeclampsia. Nat. Commun. 2019, 10, 3031. [Google Scholar] [CrossRef]
  21. Koren, O.; Konnikova, L.; Brodin, P.; Mysorekar, I.U.; Collado, M.C. The maternal gut microbiome in pregnancy: Implications for the developing immune system. Nat. Rev. Gastroenterol. Hepatol. 2024, 21, 35–45. [Google Scholar] [CrossRef]
  22. Rackaityte, E.; Halkias, J.; Fukui, E.M.; Mendoza, V.F.; Hayzelden, C.; Crawford, E.D.; Fujimura, K.E.; Burt, T.D.; Lynch, S.V. Viable bacterial colonization is highly limited in the human intestine in utero. Nat. Med. 2020, 26, 599–607. [Google Scholar] [CrossRef]
  23. Mishra, A.; Lai, G.C.; Yao, L.J.; Aung, T.T.; Shental, N.; Rotter-Maskowitz, A.; Shepherdson, E.; Singh, G.S.N.; Pai, R.; Shanti, A.; et al. Microbial exposure during early human development primes fetal immune cells. Cell 2021, 184, 3394. [Google Scholar] [CrossRef]
  24. Adak, A.; Khan, M.R. An insight into gut microbiota and its functionalities. Cell. Mol. Life Sci. 2019, 76, 473–493. [Google Scholar] [CrossRef] [PubMed]
  25. Chen, X.; Li, P.; Liu, M.; Zheng, H.; He, Y.; Chen, M.X.; Tang, W.; Yue, X.; Huang, Y.; Zhuang, L.; et al. Gut dysbiosis induces the development of pre-eclampsia through bacterial translocation. Gut 2020, 69, 513–522. [Google Scholar] [CrossRef] [PubMed]
  26. Gomez-Arango, L.F.; Barrett, H.L.; McIntyre, H.D.; Callaway, L.K.; Morrison, M.; Dekker Nitert, M.; SPRING Trial Group. Connections between the gut microbiome and metabolic hormones in early pregnancy in overweight and obese women. Diabetes 2016, 65, 2214–2223. [Google Scholar] [CrossRef] [PubMed]
  27. Edwards, S.M.; Cunningham, S.A.; Dunlop, A.L.; Corwin, E.J. The Maternal Gut Microbiome during Pregnancy. MCN Am. J. Matern. Child Nurs. 2017, 42, 310–316. [Google Scholar] [CrossRef] [PubMed]
  28. Xiong, Z.; Wang, Q.; Pei, S.; Zhu, Z. The causal role of intestinal microbiome in development of pre-eclampsia. Funct. Integr. Genom. 2023, 23, 127. [Google Scholar] [CrossRef]
  29. Deer, E.; Herrock, O.; Campbell, N.; Cornelius, D.; Fitzgerald, S.; Amaral, L.M.; LaMarca, B. The role of immune cells and mediators in preeclampsia. Nat. Rev. Nephrol. 2023, 19, 257–270. [Google Scholar] [CrossRef]
  30. Redman, C.W.; Sargent, I.L. Latest Advances in Understanding Preeclampsia. Science 1979, 308, 1592–1594. [Google Scholar] [CrossRef]
  31. Karumanchi, S.A. Two decades of advances in preeclampsia research: Molecular mechanisms and translational studies. J. Clin. Investig. 2024, 134, e184052. [Google Scholar] [CrossRef]
  32. Pijnenborg, R.; Vercruysse, L.; Hanssens, M. The Uterine Spiral Arteries In Human Pregnancy: Facts and Controversies. Placenta 2006, 27, 939–958. [Google Scholar] [CrossRef] [PubMed]
  33. Redman, C.W.G.; Staff, A.C.; Roberts, J.M. Syncytiotrophoblast stress in preeclampsia: The convergence point for multiple pathways. Am. J. Obstet. Gynecol. 2022, 226, S907–S927. [Google Scholar] [CrossRef]
  34. Ridder, A.; Giorgione, V.; Khalil, A.; Thilaganathan, B. Preeclampsia: The relationship between uterine artery blood flow and trophoblast function. Int. J. Mol. Sci. 2019, 20, 3263. [Google Scholar] [CrossRef]
  35. Tossetta, G.; Fantone, S.; Piani, F.; Crescimanno, C.; Ciavattini, A.; Giannubilo, S.R.; Marzioni, D. Modulation of NRF2/KEAP1 Signaling in Preeclampsia. Cells 2023, 12, 1545. [Google Scholar] [CrossRef]
  36. Robillard, P.Y.; Dekker, G.; Scioscia, M.; Bonsante, F.; Boukerrou, M.; Iacobelli, S.; Tran, P.L. Preeclampsia in 2023: Time for preventing early onset- and term preeclampsia: The paramount role of gestational weight gain. J. Reprod. Immunol. 2023, 158, 103968. [Google Scholar] [CrossRef] [PubMed]
  37. Karpova, N.S.; Dmitrenko, O.P.; Budykina, T.S. Literature Review: The Sflt1/Plgf Ratio and Pregestational Maternal Comorbidities: New Risk Factors to Predict Pre-Eclampsia. Int. J. Mol. Sci. 2023, 24, 6744. [Google Scholar] [CrossRef]
  38. Guo, F.; Zhang, B.; Yang, H.; Fu, Y.; Wang, Y.; Huang, J.; Cheng, M.; Li, X.; Shen, Z.; Li, L.; et al. Systemic transcriptome comparison between early- And late-onset pre-eclampsia shows distinct pathology and novel biomarkers. Cell Prolif. 2021, 54, e12968. [Google Scholar] [CrossRef]
  39. Burton, G.J.; Jauniaux, E. Pathophysiology of placental-derived fetal growth restriction. Am. J. Obstet. Gynecol. 2018, 218, S745–S761. [Google Scholar] [CrossRef]
  40. Li, F.; Wang, T.; Chen, L.; Zhang, S.; Chen, L.; Qin, J. Adverse pregnancy outcomes among mothers with hypertensive disorders in pregnancy: A meta-analysis of cohort studies. Pregnancy Hypertens 2021, 24, 107–117. [Google Scholar] [CrossRef]
  41. Kibret, K.T.; Chojenta, C.; Gresham, E.; Tegegne, T.K.; Loxton, D. Maternal dietary patterns and risk of adverse pregnancy (hypertensive disorders of pregnancy and gestational diabetes mellitus) and birth (preterm birth and low birth weight) outcomes: A systematic review and meta-analysis. Public Health Nutr. 2018, 22, 506–520. [Google Scholar] [CrossRef] [PubMed]
  42. Zhang, S.; Qiu, X.; Wang, T.; Chen, L.; Li, J.; Diao, J.; Li, Y.; Qin, J.; Chen, L.; Jiang, Y. Hypertensive Disorders in Pregnancy Are Associated With Congenital Heart Defects in Offspring: A Systematic Review and Meta-Analysis. Front. Cardiovasc. Med. 2022, 9, 842878. [Google Scholar] [CrossRef]
  43. Marín, R.; Chiarello, D.I.; Abad, C.; Rojas, D.; Toledo, F.; Sobrevia, L. Oxidative stress and mitochondrial dysfunction in early-onset and late-onset preeclampsia. Biochim. Biophys. Acta BBA Mol. Basis Dis. 2020, 1866, 165961. [Google Scholar] [CrossRef]
  44. Kluivers, A.C.M.; Biesbroek, A.; Visser, W.; Saleh, L.; Russcher, H.; Danser, A.H.J.; Neuman, R.I. Angiogenic imbalance in pre-eclampsia and fetal growth restriction: Enhanced soluble fms-like tyrosine kinase-1 binding or diminished production of placental growth factor? Ultrasound Obstet. Gynecol. 2023, 61, 466–473. [Google Scholar] [CrossRef] [PubMed]
  45. Atkinson, J.; Simpson, G.; Walker, S.P.; Tong, S.; Hastie, R.; Lindquist, A. The long-term risk of cardiovascular disease among women with a history of hypertensive disorders of pregnancy: A systematic review of clinical practice guidelines. BMC Cardiovasc. Disord. 2023, 23, 443. [Google Scholar] [CrossRef] [PubMed]
  46. Inversetti, A.; Pivato, C.A.; Cristodoro, M.; Latini, A.C.; Condorelli, G.; Di Simone, N.; Stefanini, G. Update on long-term cardiovascular risk after pre-eclampsia: A systematic review and meta-analysis. Eur. Heart J. Qual. Care Clin. Outcomes 2024, 10, 4–13. [Google Scholar] [CrossRef] [PubMed]
  47. Mészáros, B.; Kukor, Z.; Valent, S. Recent Advances in the Prevention and Screening of Preeclampsia. J. Clin. Med. 2023, 12, 6020. [Google Scholar] [CrossRef]
  48. Chang, K.J.; Seow, K.M.; Chen, K.H. Preeclampsia: Recent Advances in Predicting, Preventing, and Managing the Maternal and Fetal Life-Threatening Condition. Int. J. Environ. Res. Public Health 2023, 20, 2994. [Google Scholar] [CrossRef]
  49. Aneman, I.; Pienaar, D.; Suvakov, S.; Simic, T.P.; Garovic, V.D.; McClements, L. Mechanisms of Key Innate Immune Cells in Early- and Late-Onset Preeclampsia. Front. Immunol. 2020, 11, 1864. [Google Scholar] [CrossRef]
  50. Caillon, H.; Tardif, C.; Dumontet, E.; Winer, N.; Masson, D. Evaluation of sFlt-1/PlGF ratio for predicting and improving clinical management of pre-eclampsia: Experience in a specialized perinatal care center. Ann. Lab. Med. 2018, 38, 95–101. [Google Scholar] [CrossRef]
  51. Rana, S.; Lemoine, E.; Granger, J.; Karumanchi, S.A. Preeclampsia: Pathophysiology, Challenges and Perspectives. Circ. Res. 2019, 124, 1094–1112. [Google Scholar] [CrossRef]
  52. Kendall, R.L.; Thomas, K.A. Inhibition of vascular endothelial cell growth factor activity by an endogenously encoded soluble receptor (endothelial cells/mitogenic inhibitor/alternative transcription/angiogenesis). Proc. Natl. Acad. Sci. USA 1993, 90, 10705–10709. [Google Scholar] [CrossRef]
  53. Maynard, S.E.; Min, J.; Merchan, J.; Lim, K.H.; Li, J.; Mondal, S.; Libermann, T.A.; Morgan, J.P.; Sellke, F.W.; Stillman, I.E.; et al. Excess placental soluble fms-like tyrosine kinase 1 (sFlt1) may contribute to endothelial dysfunction, hypertension, and proteinuria in preeclampsia. J. Clin. Investig. 2003, 111, 649–658. [Google Scholar] [CrossRef]
  54. Cerdeira, A.S.; O’Sullivan, J.; Ohuma, E.O.; Harrington, D.; Szafranski, P.; Black, R.; Mackillop, L.; Impey, L.; Greenwood, C.; James, T.; et al. Randomized Interventional Study on Prediction of Preeclampsia/Eclampsia in Women with Suspected Preeclampsia: INSPIRE. Hypertension 2019, 74, 983–990. [Google Scholar] [CrossRef]
  55. Chen, Y.; Wang, X.; Hu, W.; Chen, Y.; Ning, W.; Lu, S.; He, P.; Li, L.; Zhang, Z. A risk model that combines MAP, PlGF, and PAPP-A in the first trimester of pregnancy to predict hypertensive disorders of pregnancy. J. Hum./Hypertens. 2022, 36, 184–191. [Google Scholar] [CrossRef]
  56. Colmenares-Mejía, C.C.; Quintero-Lesmes, D.C.; Bautista-Niño, P.K.; Guio Mahecha, E.; Beltrán Avendaño, M.; Díaz Martínez, L.A.; Ortiz Serrano, R.; Páez Leal, M.C.; Monterrosa Castro, Á.; Mesa Restrepo, C.M.; et al. Pentraxin-3 is a candidate biomarker on the spectrum of severity from pre-eclampsia to HELLP syndrome: GenPE study. Hypertens. Res. 2020, 43, 884–891. [Google Scholar] [CrossRef]
  57. Licini, C.; Avellini, C.; Picchiassi, E.; Mensà, E.; Fantone, S.; Ramini, D.; Tersigni, C.; Tossetta, G.; Castellucci, C.; Tarquini, F.; et al. Pre-eclampsia predictive ability of maternal miR-125b: A clinical and experimental study. Transl. Res. 2021, 228, 13–27. [Google Scholar] [CrossRef]
  58. Wakabayashi, I.; Yanagida, M.; Araki, Y. Associations of cardiovascular risk with circulating peptides related to hypertensive disorders of pregnancy. Hypertens. Res. 2021, 44, 1641–1651. [Google Scholar] [CrossRef]
  59. Araki, Y.; Yanagida, M. Hypertensive disorders of pregnancy: Strategy to develop clinical peptide biomarkers for more accurate evaluation of the pathophysiological status of this syndrome. Adv. Clin. Chem. 2020, 94, 1–30. [Google Scholar] [CrossRef]
  60. Mendes, S.; Timóteo-Ferreira, F.; Almeida, H.; Silva, E. New insights into the process of placentation and the role of oxidative uterine microenvironment. Oxidative Med. Cell. Longev. 2019, 2019, 9174521. [Google Scholar] [CrossRef]
  61. Hoffman, M.K. The great obstetrical syndromes and the placenta. BJOG Int. J. Obstet. Gynaecol. 2023, 130, 8–15. [Google Scholar] [CrossRef]
  62. 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. [Google Scholar] [CrossRef]
  63. Armaly, Z.; Jadaon, J.E.; Jabbour, A.; Abassi, Z.A. Preeclampsia: Novel mechanisms and potential therapeutic approaches. Front. Physiol. 2018, 9, 973. [Google Scholar] [CrossRef]
  64. Chiang, Y.T.; Seow, K.M.; Chen, K.H. The Pathophysiological, Genetic, and Hormonal Changes in Preeclampsia: A Systematic Review of the Molecular Mechanisms. Int. J. Mol. Sci. 2024, 25, 4532. [Google Scholar] [CrossRef]
  65. Staff, A.C. The two-stage placental model of preeclampsia: An update. J. Reprod. Immunol. 2019, 134, 1–10. [Google Scholar] [CrossRef] [PubMed]
  66. Torres-Torres, J.; Espino-y-Sosa, S.; Martinez-Portilla, R.; Borboa-Olivares, H.; Estrada-Gutierrez, G.; Acevedo-Gallegos, S.; Ruiz-Ramirez, E.; Velasco-Espin, M.; Cerda-Flores, P.; Ramirez-Gonzalez, A.; et al. A Narrative Review on the Pathophysiology of Preeclampsia. Int. J. Mol. Sci. 2024, 25, 7569. [Google Scholar] [CrossRef]
  67. Burwick, R.M.; Rodriguez, M.H. Angiogenic Biomarkers in Preeclampsia. Obstet. Gynecol. 2024, 143, 515–523. [Google Scholar] [CrossRef]
  68. Han, C.; Huang, P.; Lyu, M.; Dong, J. Oxidative stress and preeclampsia-associated prothrombotic state. Antioxidants 2020, 9, 1139. [Google Scholar] [CrossRef]
  69. McGinnis, R.; Steinthorsdottir, V.; Williams, N.O.; Thorleifsson, G.; Shooter, S.; Hjartardottir, S.; Bumpstead, S.; Stefansdottir, L.; Hildyard, L.; Sigurdsson, J.K.; et al. Variants in the fetal genome near FLT1 are associated with risk of preeclampsia. Nat. Genet. 2017, 49, 1255–1260. [Google Scholar] [CrossRef]
  70. Gu, Y.; Sun, J.; Groome, L.J.; Wang, Y. Differential miRNA expression profiles between the first and third trimester human placentas. J. Physiol. Endocrinol. Metab. 2013, 304, 836–843. [Google Scholar] [CrossRef]
  71. Munjas, J.; Sopić, M.; Stefanović, A.; Košir, R.; Ninić, A.; Joksić, I.; Antonić, T.; Spasojević-Kalimanovska, V.; Prosenc, Z. Non-coding RNAs in preeclampsia—Molecular mechanisms and diagnostic potential. Int. J. Mol. Sci. 2021, 22, 10652. [Google Scholar] [CrossRef]
  72. Dai, C.; Zhao, C.; Xu, M.; Sui, X.; Sun, L.; Liu, Y.; Su, M.; Wang, H.; Yuan, Y.; Zhang, S.; et al. Serum lncRNAs in early pregnancy as potential biomarkers for the prediction of pregnancy-induced hypertension, including preeclampsia. Mol. Ther. Nucleic Acids 2021, 24, 416–425. [Google Scholar] [CrossRef]
  73. Medina-Bastidas, D.; Guzmán-Huerta, M.; Borboa-Olivares, H.; Ruiz-Cruz, C.; Parra-Hernández, S.; Flores-Pliego, A.; Salido-Guadarrama, I.; Camargo-Marín, L.; Arambula-Meraz, E.; Estrada-Gutierrez, G. Placental microarray profiling reveals common mRNA and lncRNA expression patterns in preeclampsia and intrauterine growth restriction. Int. J. Mol. Sci. 2020, 21, 3597. [Google Scholar] [CrossRef]
  74. Zhang, Y.; Zhao, Y.; Yu, F.; Li, X.; Chen, X.; Zhu, D.; Sun, J.; Huang, Q.; Li, M.; Sun, M.; et al. CircRNA_06354 might promote early-onset preeclampsia in humans via hsa-miR-92a-3p/vascular endothelial growth factor-A. J. Hypertens. 2023, 41, 494–507. [Google Scholar] [CrossRef] [PubMed]
  75. Tersigni, C.; Meli, F.; Neri, C.; Iacoangeli, A.; Franco, R.; Lanzone, A.; Scambia, G.; Di Simone, N. Role of Human Leukocyte Antigens at the Feto-Maternal Interface in Normal and Pathological Pregnancy: An Update. Int. J. Mol. Sci. 2020, 21, 4756. [Google Scholar] [CrossRef]
  76. Guan, X.; Fu, Y.; Liu, Y.; Cui, M.; Zhang, C.; Zhang, Q.; Li, C.; Zhao, J.; Wang, C.; Song, J.; et al. The role of inflammatory biomarkers in the development and progression of pre-eclampsia: A systematic review and meta-analysis. Front. Immunol. 2023, 14, 1156039. [Google Scholar] [CrossRef]
  77. Sakaguchi, S.; Miyara, M.; Costantino, C.M.; Hafler, D.A. FOXP3 + regulatory T cells in the human immune system. Nat. Rev. Immunol. 2010, 10, 490–500. [Google Scholar] [CrossRef]
  78. Hosseini, A.; Dolati, S.; Hashemi, V.; Abdollahpour-Alitappeh, M.; Yousefi, M. Regulatory T and T helper 17 cells: Their Roles in Preeclampsia. J. Cell. Physiol. 2018, 233, 6561–6573. [Google Scholar] [CrossRef] [PubMed]
  79. Beneventi, F.; Badulli, C.; Locatelli, E.; Caporali, R.; Ramoni, V.; Cavagnoli, C.; Simonetta, M.; Garbin, G.; Tinelli, C.; Alpini, C.; et al. Soluble HLA-G in pregnancies complicated by autoimmune rheumatic diseases. J. Reprod. Immunol. 2015, 110, 67–73. [Google Scholar] [CrossRef]
  80. Zolfaghari, M.A.; Arefnezhad, R.; Parhizkar, F.; Hejazi, M.S.; Motavalli Khiavi, F.; Mahmoodpoor, A.; Yousefi, M. T lymphocytes and preeclampsia: The potential role of T-cell subsets and related MicroRNAs in the pathogenesis of preeclampsia. Am. J. Reprod. Immunol. 2021, 86, e13475. [Google Scholar] [CrossRef]
  81. Bisson, C.; Dautel, S.; Patel, E.; Suresh, S.; Dauer, P.; Rana, S. Preeclampsia pathophysiology and adverse outcomes during pregnancy and postpartum. Front. Med. 2023, 10, 1144170. [Google Scholar] [CrossRef]
  82. Gleicher, N. Maternal autoimmunity and adverse pregnancy outcomes. J. Autoimmun. 2014, 50, 83–86. [Google Scholar] [CrossRef]
  83. Andreoli, L.; Bazzani, C.; Taraborelli, M.; Reggia, R.; Lojacono, A.; Brucato, A.; Meroni, P.L.; Tincani, A. Pregnancy in autoimmune rheumatic diseases: The importance of counselling for old and new challenges. Autoimmun. Rev. 2010, 10, 51–54. [Google Scholar] [CrossRef]
  84. Østensen, M.; Brucato, A.; Carp, H.; Chambers, C.; Dolhain, R.J.; Doria, A.; Förger, F.; Gordon, C.; Hahn, S.; Khamashta, M.; et al. Pregnancy and reproduction in autoimmune rheumatic diseases. Rheumatology 2011, 50, 657–664. [Google Scholar] [CrossRef] [PubMed]
  85. Brucato, A.; Cimaz, R.; Caporali, R.; Ramoni, V.; Buyon, J. Pregnancy outcomes in patients with autoimmune diseases and anti-Ro/SSA antibodies. Clin. Rev. Allergy Immunol. 2011, 40, 27–41. [Google Scholar] [CrossRef] [PubMed]
  86. Wu, J.; Ma, J.; Zhang, W.H.; Di, W. Management and outcomes of pregnancy with or without lupus nephritis: A systematic review and meta-analysis. Ther. Clin. Risk Manag. 2018, 14, 885–901. [Google Scholar] [CrossRef] [PubMed]
  87. Walter, I.J.; Haneveld, M.J.K.; Lely, A.T.; Bloemenkamp, K.W.M.; Limper, M.; Kooiman, J. Pregnancy outcome predictors in antiphospholipid syndrome: A systematic review and meta-analysis. Autoimmun. Rev. 2021, 20, 102901. [Google Scholar] [CrossRef] [PubMed]
  88. Toloza, F.J.K.; Derakhshan, A.; Männistö, T.; Bliddal, S.; Popova, P.V.; Carty, D.M.; Chen, L.; Taylor, P.; Mosso, L.; Oken, E.; et al. Association between maternal thyroid function and risk of gestational hypertension and pre-eclampsia: A systematic review and individual-participant data meta-analysis. Lancet Diabetes Endocrinol. 2022, 10, 243–252. [Google Scholar] [CrossRef]
  89. Lei, J.; Li, Y.; Zhang, S.; Wu, Y.; Wang, P.; Liu, H. The Prognostic Role of Angiotensin Ii Type 1 Receptor Autoantibody in Non-Gravid Hypertension and Pre-Eclampsia. Medicine 2016, 95, e3494. [Google Scholar] [CrossRef]
  90. Thway, T.M.; Shlykov, S.G.; Day, M.C.; Sanborn, B.M.; Gilstrap, L.C., 3rd; Xia, Y.; Kellems, R.E. Antibodies from preeclamptic patients stimulate increased intracellular Ca2+ mobilization through angiotensin receptor activation. Circulation 2004, 110, 1612–1619. [Google Scholar] [CrossRef]
  91. Zhou, C.C.; Ahmad, S.; Mi, T.; Abbasi, S.; Xia, L.; Day, M.C.; Ramin, S.M.; Ahmed, A.; Kellems, R.E.; Xia, Y. Autoantibody from women with preeclampsia induces soluble Fms-like tyrosine kinase-1 production via angiotensin type 1 receptor and calcineurin/nuclear factor of activated T-cells signaling. Hypertension 2008, 51, 1010–1019. [Google Scholar] [CrossRef]
  92. Robertson, S.A.; Sharkey, D.J. Seminal fluid and fertility in women. Fertil. Steril. 2016, 106, 511–519. [Google Scholar] [CrossRef]
  93. Jung, E.; Romero, R.; Yeo, L.; Gomez-Lopez, N.; Chaemsaithong, P.; Jaovisidha, A.; Gotsch, F.; Erez, O. The etiology of preeclampsia. Am. J. Obstet. Gynecol. 2022, 226, S844–S866. [Google Scholar] [CrossRef]
  94. Rak, J.M.; Maestroni, L.; Balandraud, N.; Guis, S.; Boudinet, H.; Guzian, M.C.; Yan, Z.; Azzouz, D.; Auger, I.; Roudier, C.; et al. Transfer of the shared epitope through microchimerism in women with rheumatoid arthritis. Arthritis Rheum. 2009, 60, 73–80. [Google Scholar] [CrossRef] [PubMed]
  95. Martínez-Varea, A.; Pellicer, B.; Perales-Marín, A.; Pellicer, A. Relationship between maternal immunological response during pregnancy and onset of preeclampsia. J. Immunol. Res. 2014, 2014, 210241. [Google Scholar] [CrossRef]
  96. Wang, J.X.; Knottnerus, A.M.; Schuit, G.; Norman, R.J.; Chan, A.; Dekker, G.A. Surgically obtained sperm, and risk of gestational hypertension and pre-eclampsia. Lancet 2002, 359, 673–674. [Google Scholar] [CrossRef]
  97. Einarsson, J.I.; Sangi-Haghpeykar, H.; Gardner, M.O. Sperm exposure and development of preeclampsia. Am. J. Obs. Gynecol. 2003, 188, 1241–1243. [Google Scholar] [CrossRef] [PubMed]
  98. Hendin, N.; Meyer, R.; Peretz-Machluf, R.; Elbaz, L.; Maman, E.; Baum, M. Higher incidence of preeclampsia among participants undergoing in-vitro fertilization after fewer sperm exposures. Eur. J. Obstet. Gynecol. Reprod. Biol. 2023, 285, 12–16. [Google Scholar] [CrossRef] [PubMed]
  99. Pohjonen, E.M.; Söderström-Anttila, V.; Bergh, C.; Loft, A.; Magnusson, Å.; Pinborg, A.; Oldereid, N.B.; Petzold, M.; Romundstad, L.B.; Laivuori, H. Obstetric and perinatal risks after the use of donor sperm: A systematic review and meta-analysis. Eur. J. Obstet. Gynecol. Reprod. Biol. 2022, 274, 210–228. [Google Scholar] [CrossRef]
  100. Liu, J.; Yang, H.; Yin, Z.; Jiang, X.; Zhong, H.; Qiu, D.; Zhu, F.; Li, R. Remodeling of the gut microbiota and structural shifts in Preeclampsia patients in South China. Eur. J. Clin. Microbiol. Infect. Dis. 2017, 36, 713–719. [Google Scholar] [CrossRef]
  101. Huang, L.; Cai, M.; Li, L.; Zhang, X.; Xu, Y.; Xiao, J.; Huang, Q.; Luo, G.; Zeng, Z.; Jin, C.; et al. Gut microbiota changes in preeclampsia, abnormal placental growth and healthy pregnant women. BMC Microbiol. 2021, 21, 265. [Google Scholar] [CrossRef]
  102. Wu, Z.; Ge, M.; Liu, J.; Chen, X.; Cai, Z.; Huang, H. The gut microbiota composition and metabolites are different in women with hypertensive disorders of pregnancy and normotension: A pilot study. J. Obstet. Gynaecol. Res. 2023, 50, 334–341. [Google Scholar] [CrossRef]
  103. Wang, J.; Shi, Z.H.; Yang, J.; Wei, Y.; Wang, X.Y.; Zhao, Y.Y. Gut microbiota dysbiosis in preeclampsia patients in the second and third trimesters. Chin. Med. J. 2020, 133, 1057–1065. [Google Scholar] [CrossRef]
  104. Geldenhuys, J.; Redelinghuys, M.J.; Lombaard, H.A.; Ehlers, M.M.; Cowan, D.; Kock, M.M. Diversity of the gut, vaginal and oral microbiome among pregnant women in South Africa with and without pre-eclampsia. Front. Glob. Womens Health 2022, 3, 810673. [Google Scholar] [CrossRef] [PubMed]
  105. Wu, X.; Li, Q.; Cai, J.; Huang, H.; Ma, S.; Tan, H. Longitudinal change of gut microbiota in hypertensive disorders in pregnancy: A nested case–control and Mendelian randomization study. Sci. Rep. 2023, 13, 16986. [Google Scholar] [CrossRef]
  106. Lv, L.J.; Li, S.H.; Li, S.C.; Zhong, Z.C.; Duan, H.L.; Tian, C.; Li, H.; He, W.; Chen, M.C.; He, T.W.; et al. Early-onset preeclampsia is associated with gut microbial alterations in antepartum and postpartum women. Front. Cell. Infect. Microbiol. 2019, 9, 224. [Google Scholar] [CrossRef]
  107. Miao, T.; Yu, Y.; Sun, J.; Ma, A.; Yu, J.; Cui, M.; Yang, L.; Wang, H. Decrease in abundance of bacteria of the genus bifidobacterium in gut microbiota may be related to pre-eclampsia progression in women from east china. Food Nutr. Res. 2021, 65, 5781. [Google Scholar] [CrossRef] [PubMed]
  108. Lv, L.J.; Li, S.H.; Wen, J.Y.; Wang, G.Y.; Li, H.; He, T.W.; Lv, Q.B.; Xiao, M.C.; Duan, H.L.; Chen, M.C.; et al. Deep metagenomic characterization of gut microbial community and function in preeclampsia. Front. Cell. Infect. Microbiol. 2022, 12, 933523. [Google Scholar] [CrossRef] [PubMed]
  109. Zhao, Y.; Wang, B.; Zhao, X.; Cui, D.; Hou, S.; Zhang, H. The effect of gut microbiota dysbiosis on patients with preeclampsia. Front. Cell. Infect. Microbiol. 2023, 12, 1022857. [Google Scholar] [CrossRef]
  110. Meijer, S.; Pasquinelli, E.; Renzi, S.; Lavasani, S.; Nouri, M.; Erlandsson, L.; Cavalieri, D.; Hansson, S.R. Gut Micro- and Mycobiota in Preeclampsia: Bacterial Composition Differences Suggest Role in Pathophysiology. Biomolecules 2023, 13, 346. [Google Scholar] [CrossRef]
  111. Colonetti, T.; Limas Carmo Teixeira, D.; Grande, A.J.; Rodrigues Uggioni, M.L.; Generoso, J.; Harding, S.; Rodriguez-Mateos, A.; Rech, P.; Rosa Silva, F.; Toreti, I.; et al. The role of intestinal microbiota on pre-eclampsia: Systematic review and meta-analysis. Eur. J. Obstet. Gynecol. Reprod. Biol. 2023, 291, 49–58. [Google Scholar] [CrossRef]
  112. Verhaar, B.J.H.; Prodan, A.; Nieuwdorp, M.; Muller, M. Gut microbiota in hypertension and atherosclerosis: A review. Nutrients 2020, 12, 2982. [Google Scholar] [CrossRef]
  113. Jin, J.; Gao, L.; Zou, X.; Zhang, Y.; Zheng, Z.; Zhang, X.; Li, J.; Tian, Z.; Wang, X.; Gu, J.; et al. Gut Dysbiosis Promotes Preeclampsia by Regulating Macrophages and Trophoblasts. Circ. Res. 2022, 131, 492–506. [Google Scholar] [CrossRef]
  114. Li, P.; Wang, H.; Guo, L.; Gou, X.; Chen, G.; Lin, D.; Fan, D.; Guo, X.; Liu, Z. Association between gut microbiota and preeclampsia-eclampsia: A two-sample Mendelian randomization study. BMC Med. 2022, 20, 443. [Google Scholar] [CrossRef] [PubMed]
  115. Wu, X.; Li, Q.; Lin, D.; Cai, J.; Huang, H.; Tan, H. Gut microbiota and hypertensive disorders in pregnancy: Evidence from the Mendelian randomization study. Aging 2023, 15, 9105. [Google Scholar] [CrossRef] [PubMed]
  116. Ishimwe, J.A. Maternal microbiome in preeclampsia pathophysiology and implications on offspring health. Physiol. Rep. 2021, 9, e14875. [Google Scholar] [CrossRef] [PubMed]
  117. Cui, J.; Wang, J.; Wang, Y. The role of short-chain fatty acids produced by gut microbiota in the regulation of pre-eclampsia onset. Front. Cell. Infect. Microbiol. 2023, 13, 1177768. [Google Scholar] [CrossRef]
  118. Chang, Y.; Chen, Y.; Zhou, Q.; Wang, C.; Chen, L.; Di, W.; Zhang, Y. Short-chain fatty acids accompanying changes in the gut microbiome contribute to the development of hypertension in patients with preeclampsia. Clin. Sci. 2020, 134, 289–302. [Google Scholar] [CrossRef]
  119. Altemani, F.; Barrett, H.L.; Gomez-Arango, L.; Josh, P.; David McIntyre, H.; Callaway, L.K.; Morrison, M.; Tyson, G.W.; Dekker Nitert, M. Pregnant women who develop preeclampsia have lower abundance of the butyrate-producer Coprococcus in their gut microbiota. Pregnancy Hypertens 2021, 23, 211–219. [Google Scholar] [CrossRef]
  120. Qing, W.; Shi, Y.; Zhou, H.; Chen, M. Gut microbiota dysbiosis in patients with preeclampsia: A systematic review. Med. Microecol. 2021, 10, 100047. [Google Scholar] [CrossRef]
  121. Li, J.; Wang, L.; Chen, H.; Yang, Z.; Chen, S.; Wang, J.; Zhou, Y.; Xuan, R. The Diagnostic Potential of Gut Microbiota-Derived Short-Chain Fatty Acids in Preeclampsia. Front. Pediatr. 2022, 10, 878924. [Google Scholar] [CrossRef]
  122. Koeth, R.A.; Lam-Galvez, B.R.; Kirsop, J.; Wang, Z.; Levison, B.S.; Gu, X.; Copeland, M.F.; Bartlett, D.; Cody, D.B.; Dai, H.J.; et al. L-Carnitine in omnivorous diets induces an atherogenic gut microbial pathway in humans. J. Clin. Investig. 2019, 129, 373–387. [Google Scholar] [CrossRef]
  123. Wang, J.; Gu, X.; Yang, J.; Wei, Y.; Zhao, Y. Gut Microbiota Dysbiosis and Increased Plasma LPS and TMAO Levels in Patients With Preeclampsia. Front. Cell. Infect. Microbiol. 2019, 9, 409. [Google Scholar] [CrossRef]
  124. Xu, H.; Feng, P.; Sun, Y.; Wu, D.; Wang, D.; Wu, L.; Peng, H.; Li, H. Plasma trimethylamine N-oxide metabolites in the second trimester predict the risk of hypertensive disorders of pregnancy: A nested case-control study. Hypertens. Res. 2024, 47, 778–789. [Google Scholar] [CrossRef] [PubMed]
  125. Koren, O.; Goodrich, J.K.; Cullender, T.C.; Spor, A.; Laitinen, K.; Bäckhed, H.K.; Gonzalez, A.; Werner, J.J.; Angenent, L.T.; Knight, R.; et al. Host remodeling of the gut microbiome and metabolic changes during pregnancy. Cell 2012, 150, 470–480. [Google Scholar] [CrossRef] [PubMed]
  126. Masson, G.S.; Nair, A.R.; Dange, R.B.; Silva-Soares, P.P.; Michelini, L.C.; Francis, J. Toll-like receptor 4 Promotes Autonomic Dysfunction, Inflammation and microglia activation in the hypothalamic paraventricular nucleus: Role of endoplasmic reticulum stress. PLoS ONE 2015, 10, e0122850. [Google Scholar] [CrossRef] [PubMed]
  127. Sturgeon, C.; Fasano, A. Zonulin, a regulator of epithelial and endothelial barrier functions, and its involvement in chronic inflammatory diseases. Tissue Barriers 2016, 4, e1251384. [Google Scholar] [CrossRef] [PubMed]
  128. Brescia, P.; Rescigno, M. The gut vascular barrier: A new player in the gut–liver–brain axis. Trends Mol. Med. 2021, 27, 844–855. [Google Scholar] [CrossRef]
  129. Tersigni, C.; D’Ippolito, S.; Di Nicuolo, F.; Marana, R.; Valenza, V.; Masciullo, V.; Scaldaferri, F.; Malatacca, F.; de Waure, C.; Gasbarrini, A.; et al. Recurrent pregnancy loss is associated to leaky gut: A novel pathogenic model of endometrium inflammation? J. Transl. Med. 2018, 16, 102. [Google Scholar] [CrossRef]
  130. Kanehisa, M.; Sato, Y.; Morishima, K. BlastKOALA and GhostKOALA: KEGG Tools for Functional Characterization of Genome and Metagenome Sequences. J. Mol. Biol. 2016, 428, 726–731. [Google Scholar] [CrossRef]
Figure 1. Defective trophoblastic phenotype and related immunological dysfunctions in preeclampsia. On the left side, the physiological placentation processes are represented: the cytotrophoblast cells acquire an invasive phenotype to enhance efficient vascular anastomoses, and decidual T cells, NK cells, and M2 macrophages establish the maternal immune tolerance to the fetus. On the right side, there is a schematic representation of immunological changes in preeclampsia. The trophoblast without the invasive phenotype leads to the abnormal development of narrow vessels and endothelial damage (also due to higher levels of AT1-AAs) and increased ROS production. These changes cause an imbalance of angiogenic and anti-angiogenic factors (PIGF and sFlt-1) and a pro-inflammatory environment (Th1 shift, increasing levels of NK and M1 macrophages). Acronyms: Th1/Th2: T helper 1/2 cells; Treg: regulatory T cells; NK: natural killer cells; ROS: reactive oxygen species; AT1-AAs: antibodies against the angiotensin II type 1 receptor; PIGF: placental growth factor; sFlt-1: soluble FMS-like tyrosine kinase-1.
Figure 1. Defective trophoblastic phenotype and related immunological dysfunctions in preeclampsia. On the left side, the physiological placentation processes are represented: the cytotrophoblast cells acquire an invasive phenotype to enhance efficient vascular anastomoses, and decidual T cells, NK cells, and M2 macrophages establish the maternal immune tolerance to the fetus. On the right side, there is a schematic representation of immunological changes in preeclampsia. The trophoblast without the invasive phenotype leads to the abnormal development of narrow vessels and endothelial damage (also due to higher levels of AT1-AAs) and increased ROS production. These changes cause an imbalance of angiogenic and anti-angiogenic factors (PIGF and sFlt-1) and a pro-inflammatory environment (Th1 shift, increasing levels of NK and M1 macrophages). Acronyms: Th1/Th2: T helper 1/2 cells; Treg: regulatory T cells; NK: natural killer cells; ROS: reactive oxygen species; AT1-AAs: antibodies against the angiotensin II type 1 receptor; PIGF: placental growth factor; sFlt-1: soluble FMS-like tyrosine kinase-1.
Ijms 25 10099 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zambella, E.; Peruffo, B.; Guarano, A.; Inversetti, A.; Di Simone, N. The Hidden Relationship between Intestinal Microbiota and Immunological Modifications in Preeclampsia Pathogenesis. Int. J. Mol. Sci. 2024, 25, 10099. https://doi.org/10.3390/ijms251810099

AMA Style

Zambella E, Peruffo B, Guarano A, Inversetti A, Di Simone N. The Hidden Relationship between Intestinal Microbiota and Immunological Modifications in Preeclampsia Pathogenesis. International Journal of Molecular Sciences. 2024; 25(18):10099. https://doi.org/10.3390/ijms251810099

Chicago/Turabian Style

Zambella, Enrica, Beatrice Peruffo, Alice Guarano, Annalisa Inversetti, and Nicoletta Di Simone. 2024. "The Hidden Relationship between Intestinal Microbiota and Immunological Modifications in Preeclampsia Pathogenesis" International Journal of Molecular Sciences 25, no. 18: 10099. https://doi.org/10.3390/ijms251810099

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop