Next Article in Journal
Transthyretin as a Biomarker to Predict and Monitor Major Depressive Disorder Identified by Whole-Genome Transcriptomic Analysis in Mouse Models
Next Article in Special Issue
Versatile Role of Prokineticins and Prokineticin Receptors in Neuroinflammation
Previous Article in Journal
Modified Vaccinia Virus Ankara as a Viral Vector for Vaccine Candidates against Chikungunya Virus
Previous Article in Special Issue
IL-17A Damages the Blood–Retinal Barrier through Activating the Janus Kinase 1 Pathway
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pathophysiological Implication of Pattern Recognition Receptors in Fetal Membranes Rupture: RAGE and NLRP Inflammasome

1
CNRS, INSERM, GReD, Université Clermont Auvergne, 63000 Clermont-Ferrand, France
2
CHU de Clermont-Ferrand, Biochemistry and Molecular Genetic Department, 63000 Clermont-Ferrand, France
*
Author to whom correspondence should be addressed.
Biomedicines 2021, 9(9), 1123; https://doi.org/10.3390/biomedicines9091123
Submission received: 30 July 2021 / Revised: 25 August 2021 / Accepted: 26 August 2021 / Published: 31 August 2021
(This article belongs to the Special Issue Angiogenesis and Inflammation in Biological Barriers 2.0)

Abstract

:
Preterm prelabor ruptures of fetal membranes (pPROM) are a pregnancy complication responsible for 30% of all preterm births. This pathology currently appears more as a consequence of early and uncontrolled process runaway activation, which is usually implicated in the physiologic rupture at term: inflammation. This phenomenon can be septic but also sterile. In this latter case, the inflammation depends on some specific molecules called “alarmins” or “damage-associated molecular patterns” (DAMPs) that are recognized by pattern recognition receptors (PRRs), leading to a microbial-free inflammatory response. Recent data clarify how this activation works and which receptor translates this inflammatory signaling into fetal membranes (FM) to manage a successful rupture after 37 weeks of gestation. In this context, this review focused on two PRRs: the receptor for advanced glycation end-products (RAGE) and the NLRP7 inflammasome.

1. Fetal Membranes: A Human Barrier Essential to Childbirth

During pregnancy, the fetus must be in a peaceful and homeostatic environment, hence being protected against external aggression so that it can grow. Fetal membranes (FM), an adjustable biological barrier, are essential to this protection.

1.1. Structure of Fetal Membranes

The FM, such as the placenta, are extra-embryonic transient tissues expelled during delivery. The placenta is located on the surface of the uterus wall, while the FMs are attached to the placenta and cover the inner wall of the amniotic cavity containing the fetus and amniotic fluid (AF). With a surface area of around 1500 cm2 and a thickness between 200 and 300 µm at term, FM interact with the placenta and maternal decidua. Together, the placenta and FM ensure materno-fetal interactions, constituting a real interface and complex barrier between the mother and her future child throughout pregnancy. Fetal membranes are composed of two layers: the amnion, which is directly in contact with the AF, and the chorion, which is attached to the maternal decidua [1,2,3,4].

1.2. A Multifunctional Barrier: Different Biological Roles of FM during Pregnancy

FM accomplishes several important physiological functions throughout pregnancy until parturition. First, thanks to their elastic resistance strength, they constitute a physical barrier. This is an absolute requirement for maintaining the integrity of the amniotic cavity in the semirigid uterus, while the movements of the AF, which are caused by the development of the fetus, exert permanent pressure in all directions. This resistance and elasticity of the FM are conferred mainly by the extracellular matrix of the amnion [5,6,7].
In addition, FMs are also involved in the regulation of AF volume and homeostasis. Indeed, they allow for bidirectional transfers of AF between the amniotic cavity and maternal circulation thanks, for example, to aquaporins [8,9,10].
They also constitute a barrier against infections and toxins by releasing some antimicrobial molecules (defensins, lactoferrin, elafin, etc.) [11,12,13]. Furthermore, it has been also described that amniotic tight junctions decrease the spread of microbes in pregnancy, whereas inflammatory mediators can disrupt this barrier, especially in the case of chorioamnionitis [14,15].
At the end of this nine-month period, this barrier breaks and the parturition starts, opening the way for the fetus. This phenomenon, called FM rupture, is complex and requires the involvement of many molecular and cellular mechanisms, leading to progressive FM weakening (Figure 1).

2. Inflammation or How Does the FM Rupture Occur Right on Time or Too Early?

2.1. Happy Ending: Well-Balanced Inflammation for a Well-Prepared Rupture of the FM at Term

Most of the time, FM rupture occurs at the beginning of labor. However, in 10% of term deliveries, it happens before labor, meaning that a rupture is not limited to only being a consequence of labor contractions [16,17]. Most of time, this breakage takes place in a particular zone of the FM facing the cervix called the “zone of altered morphology” (ZAM), which is in opposition to the “zone of intact morphology” (ZIM) (note that sometimes the rupture site is aside the ZAM) [18]. Anatomically, the ZAM presents a thinner structure than the ZIM because of the combination of exacerbated mechanisms in this zone, such as oxidative stress, hormone production, apoptosis, senescence, epithelio-mesenchymal transition, inflammation and extracellular matrix (ECM) degradation [3,18,19,20,21,22]. In addition, these mechanisms are associated with stretching and the appearance of microfractures of the barrier, which become weaker throughout pregnancy [4,23].
Recently, more and more studies have particularly focused on the establishment and implication of the inflammation process. Indeed, several results have highlighted a higher inflammation phenomenon in the ZAM. For example, at term, chorion-derived trophoblasts from the ZAM modify their phenotype, inducing maternal immune cell activation. In addition, a transcriptome realized by the Nhan-Chang group revealed a differential expression of inflammatory genes between chorionic ZAM and ZIM [24,25]. More generally, in parallel with this ZAM tissue-specific inflammation, the AF levels of cytokines, such as IL1β, IL6 and TNFα, are increased during labor [26,27]. Furthermore, the release of TNFα induces apoptosis in FM, and IL1β and TNFα lead to downstream chemokine production (IL8, CXCL1, CCL2) associated with the recruitment of leukocytes toward the amnion and chorion [28]. In addition, these pro-inflammatory mediators increase the gelatinase (metalloproteases 2, 9) activity, leading to the degradation of the ECM in the FM (via the nuclear factor-kappa B (NFκB) pathway) [22,29,30,31,32].
Thus, inflammation appears to be central in the physiological weakening of FM, and this response is set up sequentially during pregnancy. Furthermore, this end-of-pregnancy inflammation is mainly microbial free and, thus, is called sterile and results from the recognition of some specific molecules, the alarmins or DAMPs (damage-associated molecular patterns). Several types of DAMPs (high-mobility group box 1 (HMGB1), uric acid, cell-free DNA, etc.) are released by senescent FM cells the closer the pregnancy comes to term; they spread throughout adjacent tissues as a consequence of cell apoptosis or are encapsulated in small extracellular vesicles called exosomes, amplifying both inflammation and senescence. In fact, for example, a gradual increase in the maternal plasma exosomes containing pro-inflammatory agents was discovered during mouse gestation [33]. They have been reported in gestational tissues during labor and described as responsible for the activation of various pro-inflammatory pathways, such as the p38 MAPK or NFκB pathways [34,35]. To act as “inflammation promoters,” DAMPs are recognized by the receptors of innate immunity: the PRRs (pattern recognition receptors). It is worth noting that PAMPs/MAMPs (pathogen-associated molecular patterns/micro-organism associated molecular patterns) are recognized by the same receptors in the case of a microbial response. Two classes of PRRs have been described: the membrane PRRs and cytosolic PRRs, allowing for a recognition of a danger signal outside and inside the target cells, respectively. Among the membrane-bound PRRs are the CLRs (C-type Lectin receptors), TLRs (toll-like receptors) and, more recently, RAGE (receptor for advanced glycation end products). Alternatively, NLRs (nucleotide-binding and oligomerization domain (NOD)-like receptors), RLRs (retinoic acid-inducible gene-I (RIG-I)-like receptors), ALRs (absent-in-melanoma (AIM)-like receptors) and the pyrins are cytosolic PRRs [36,37] (Figure 2).
As mentioned above, inflammation is an essential component of the physiological process of FM weakening. Nevertheless, as Paracelsus said, “The dose makes the poison” (or should we say, DAMPs make the way out). Indeed, even if this physiological well-balanced inflammation leads to a healthy birth right on time, when it is overly exacerbated or not well-regulated, it can result in preterm prelabor rupture of the FM and an unfortunate preterm birth.

2.2. Preterm Prelabor Rupture of Fetal Membranes (pPROM)

2.2.1. Definition and Consequences of the pPROM

In obstetrics, pPROM is defined as a rupture of the FM before the onset of labor. It can happen at any gestational age. pPROM affects 2–4% of singleton pregnancies and 7–20% of twin pregnancies [16]. This complication is responsible for one-third of preterm births (PTB) and is associated with an elevation of perinatal and maternal mortality [16,38,39]. Indeed, pPROM increases the risk of placental abruption, postpartum endometritis and chorioamnionitis [40,41]. Such early inflammation of the placenta and FM can be caused by the rise of genital germs or by contamination following invasive medical procedures. The incidence of chorioamnionitis is directly related to the duration of the latency period between FM rupture and childbirth [42,43]. Finally, pPROM leads to a decrease in AF volume, which is potentially associated with umbilical cord compressions or prolapses. pPROM can also lead to neonatal infection, though its main outcome is PTB, whose consequences vary depending on the gestational age (developmental disorders of lungs, intestines, sensorial organs, nervous system, etc.) [44].

2.2.2. Pathophysiology of pPROM and Inflammation

As previously described, membrane rupture results from a complex and multifactorial chain, leading to a progressive weakening of the FM. pPROM results from this same chain but in an earlier and accelerated way. We can imagine that while the FM normally takes the time on the slow lane, those of mothers undergoing a pPROM burn the speed limits, arriving too early at the final destination: their decay. This pathology is described as being the consequence of a myriad of risk factors whose origin can be maternal (smoking, drug consumption, ethnic polymorphisms, nutrition), feto-utero-placental (multiple pregnancies, infection, premature labor), environmental (pollutants, lead, etc.) or iatrogenic (amniocentesis, etc.) [45,46,47].
Indeed, sterile or septic inflammation is considered essential in the pathogenesis of pPROM. Several inflammatory markers have been found to increase in AF, maternal blood, the placenta and the FM in cases of pPROM [48,49,50,51,52,53]. These higher levels of pro-inflammatory cytokines have been correlated with the recruitment of leukocytes (neutrophils, monocytes, macrophages, T cells) within the AF of pPROM patients, especially those with a positive bacterial culture [49,54]. Conversely, higher levels of anti-inflammatory cytokines (IL1RA) are associated with a longer latency (>7 days) to delivery in women with pPROM [55]. Thus, many interleukins have been studied as the potential biomarkers of pPROM, such as IL6 [56,57,58]. If it is well known that an infectious stimulation provokes an inflammatory response, in particular at the chorion level (first target of vaginal pathogens); here, some teams have started to study sterile inflammation (potentially caused by smoking, nutritional deficiencies, pollutants, etc.) in the context of pPROM [59,60]. Indeed, its role is not negligible. A study showed that 58% of patients with sterile intra-amniotic inflammation have histological chorioamnionitis features (infiltration of polynuclears at the level of the fetal membranes) [61]. Moreover, higher concentrations of alarmins such as HMGB1, S100β, heat shock protein 70, cell-free DNA and S100 proteins have been found within AF, maternal blood or even gestational tissues (placenta, FM) in cases of pPROM or chorioamnionitis [62,63,64,65,66,67,68,69]; these increased amounts of DAMPs, which can lead to an inflammatory stimulation by interacting with PRRs, might be the consequence of oxidative stress or senescence [70,71]. Finally, a study revealed that the in vivo inhibition (in a murine model) of the inflammatory pathway NFκB causes delays in lipopolysaccharide (LPS)-induced PTB, clearly showing the importance of inflammation in parturition [72].
In the following parts of this review, special attention will be paid to the importance of the RAGE membrane receptor and to an intracytoplasmic receptor family that can form an inflammatory platform: the inflammasome.

3. The Receptor for Advanced Glycation End Products (RAGE)

3.1. RAGE Protein Structure

Discovered in 1992 by Michael Neeper, the RAGE protein (molecular weight of about 46 kDa) is a membrane receptor belonging to the immunoglobulin superfamily and induces downstream different intracellular signals. Globally, it is expressed at low levels in all human tissue, except in the lungs, where its expression is more important [73,74]. Structurally, the full-length RAGE (fl-RAGE) is composed of three domains: an extracellular domain (itself subdivided into three parts involved in ligand recognition: V, C1 and C2), a transmembrane domain and a cytoplasmic portion required for signal transduction by its interaction with adaptor proteins (Myeloid differentiation primary response 88 (MyD88), Diaphanous-1 (Dia-1) and the Toll-interleukin 1 Receptor (TIR) domain containing adaptor protein (TIRAP)) [75,76,77,78].
RAGE also exists in truncated forms. Two of them are soluble: sRAGE (soluble RAGE), which is derived from a proteolytic cleavage of fl-RAGE by specific metalloproteases, and esRAGE (endogenous secretory RAGE), which is derived from one specific splicing transcript [79,80]. Functionally, these two forms, which lack the transmembrane and intracellular domains, act as inhibitors of signal transduction by capturing RAGE ligands in the extracellular environment before their recognition by the functional fl-RAGE. There is also an isoform named DN-RAGE (dominant negative), which is addressed to the membrane but does not have an intracellular domain. Finally, Nt-RAGE (N-terminal truncated), resulting from alternative splicing, is V-domain lacking, so it cannot bind to the usual ligands (Figure 3) [74].

3.2. RAGE, Ligands and Signaling

Thanks to its multiple extracellular domains (V, C1, C2), RAGE can interact with a considerable panel of ligands, mainly endogenous ligands that are accumulated in tissues during aging, inflammation and other stresses: the DAMPs are also called alarmins [81,82]. Among these ligands, RAGE can interact with the HMGB1, an S100 protein family, advanced glycation end-products (AGEs) derived from the Maillard reaction of glycation, cell-free DNA and RNA, uric acid... This diversity of ligands, which is associated with other factors such as cell type, ligand or receptor concentration at the membrane or type of adaptors, allows RAGE to induce numerous signaling pathways, leading to various cell responses. For example, RAGE has been described in cell migration and proliferation but also in apoptosis [83,84]. A recent study also demonstrated that RAGE can induce autophagy [85]. Finally, one of the main roles of RAGE is its ability to initiate an inflammatory response [75].

3.3. RAGE and Inflammation

In the literature, RAGE exhibits high levels of expression in the inflammatory lesions associated with many types of pathologies. On the other hand, blocking RAGE delays the inflammatory response [86]. For example, RAGE knockout mice are protected against inflammatory disorders (sepsis, smoking-induced lung inflammation, etc.) [87,88]. In addition, RAGE is expressed in many immune cells, and an accumulation of its ligands is often observed at inflammation sites [89,90,91]. The stimulation of RAGE initiates various pro-inflammatory pathways. For example, the NFκB pathway was the first signal described as activated by AGEs [92]. This transcription factor regulates the expression of several genes, including pro-inflammatory mediators (TNFα, IL1β, IL6, cyclooxygenase 2, etc.) and RAGE itself, amplifying chronic inflammation [93]. Activation of RAGE by its ligands can also lead to the induction of MAP kinase signaling cascades, such as ERK, JNK or p38-MAPK (Figure 4) [94,95].

3.4. RAGE and Fetal Membranes Rupture

Despite the well-described role of RAGE in inflammatory pathologies, only a few studies have investigated its potential implication in FM inflammation and their subsequent rupture. Romero et al. demonstrated higher AF levels of sRAGE and esRAGE in the presence of intra-amniotic inflammation and infection [96]. Nevertheless, under the same conditions, another study demonstrated that these levels are not impacted by the presence of intra-amniotic inflammation/infection, instead revealing the expression of RAGE within the amnion and chorion [97]. Later, other studies reported an increase in plasmatic esRAGE levels in pPROM but also in placental RAGE levels [63,98]. However, more recently, in 2020, another study found no significant difference in plasmatic sRAGE levels between pPROM and term rupture [99].
Concerning its ligands, DAMPs are found in AF and gestational tissues and are often dysregulated in cases of pregnancy pathologies such as pPROM. For example, overexpression of HMGB1 in the sera and placentas of mothers undergoing pPROM has been described [63,100]. In addition, a preliminary study supported the idea of using AGEs as an early (first trimester) serum marker of preterm labor and pPROM [101]. Furthermore, a direct link was demonstrated between the risk factors of pPROM (e.g., smoking) and the release or production of alarmins; for example, it has been well described that smoking induces alarmin release [102].
Nevertheless, a recent study clarified the implication of RAGE in FM weakening because of sterile inflammation [103]. Indeed, the ontogeny of the RAGE axis in FM obtained from nonpathological deliveries at term by cesarean produced the first data about the expression of its adaptors in these tissues, which is essential to RAGE activity. It was elucidated that the FM expressed RAGE and HMGB1 but also two intracellular adaptors (Dia-1 and MyD88) during the three trimesters of human pregnancy. Furthermore, the RAGE protein was identified in the amniotic epithelium, the first cells in contact with AF alarmins and in the chorion too [103]. A comparison of the RAGE actors’ expressions between the two layers of FM, but also between the ZAM (rupture zone, the weaker) and ZIM, revealed that RAGE and HMGB1 expression is significantly higher in the amnion compared with chorion. Even better, an overexpression of RAGE was found in the ZAM compared with the ZIM for the amnion (layer considered the stronger in FM) [103]. This suggests an overactivation of RAGE in the amnion around the zone of rupture, which is probably involved in its weakening. Romero et al. described an HMGB1-associated sterile inflammation on the fetal side of the FM [66]. Moreover, the overexpression of Dia-1 was revealed in amnion, whereas MyD88 was found overexpressed in chorion [103]. These differences could be considered a clue for layer-specific signaling, here depending on the recruited adaptor. Such consideration is supported by the fact that it is already well known that MyD88 is shared with TLR2/4 and TLR4 is highly expressed in the FM as well, opening the way to study the hypothesis of cooperation between RAGE and TLR4 in FM weakening [104].
In parallel, AGEs and HMGB1, which are both alarmins found to be increased in the case of pPROM, can induce the sterile inflammation of FM. Indeed, some studies have demonstrated that they induce cytokine release (IL8, IL6, TNFα, IL1β) in the FM without a distinction between amnion and chorion [35,105]. Later, these first data were complemented by revealing that amnion and choriodecidua did not respond in the same way to the stimuli from alarmins. Both layers respond to alarmins by the release of TNFα and IL1β, but only the amnion produced IL6 following the treatment. Above all, RAGE was found to be required in the FM layers to induce a pro-inflammatory signal after AGEs or HMGB1 exposure. Indeed, a total abortion of HMGB1-induced inflammation by the RAGE inhibitor was observed, but only partially decreased AGEs-induced inflammation [103]. These differences remind us of the complexity of RAGE activity, depending on the ligand type and ligand concentration. To conclude, recent data support the importance of RAGE engagement in FM sterile inflammation because of alarmins. Obviously, further studies are required to describe this RAGE activation in the FM, the adaptor recruitment and specificity of actions, here investigating downstream cell pathways (MAPK, NFκB?), the type of cell responding in the tissue and possible cooperation with other inflammation actors, such as the inflammasome platforms described below.

4. Inflammasomes

4.1. What Are Inflammasomes?

The first inflammasome was discovered in 2002 by the biochemist Jürg Tschopp [106]. This molecular platform can recognize, as a PRR, the DAMPs and PAMPs and induce inflammatory processes, especially pyroptosis, which is a recently described inflammation-specific cell death [107,108]. In fact, an inflammasome is an intracellular complex composed of three actors: a sensor, an effector (the pro-caspase 1) and an adaptor between the two previous parts. These inflammasomes are formed by cytosolic PRRs, such as NLRs; however, membrane PRR activation can stimulate the downstream formation of inflammasomes [109,110]. In this review, we focus on one subfamily of NLRs, the NLRPs (NLR and Pyrin domain containing), which includes 14 members: NLRP1 to NLRP14.

4.1.1. The NLRP Inflammasome: The Trio NLRP/ASC/pro-Caspase 1

For this group, the inflammasome includes one NLRP as a sensor linked to the procaspase 1, here thanks to the ASC adaptor (apoptosis-associated speck-like protein containing a caspase recruitment domain (CARD)). More precisely, a NLRP (except for NLRP1 and 10) is generally composed of these domains: a pyrin domain (PYD) followed by a central domain NACHT (NAIP, CIITA, HET-E, TP1 proteins) involved in oligomerization; a domain associated with NACHT (NAD); and, finally, a carboxy terminal domain made of Leucine-rich repeats (LRR) [111,112,113]. During the assembly of the inflammasome, the NLRP interacts with the adaptor ASC because of the PYD domain of each actor. the ASC also contains a CARD domain that binds the CARD domain of pro-caspase 1 [114,115]. Caspases are divided into two classes: pro-apoptotic and pro-inflammatory caspases. Caspase 1 belongs to the second one and is included in the inflammasome as a pro-caspase 1 protein composed of three domains: CARD, p20 and p10 (Figure 5). Following such an association, inflammasomes assemble together, leading to the formation of a specific structure called specks [116,117,118].

4.1.2. NLRP Inflammasome Activation

Based on studies about NLRP3, two activation pathways have been described. To form a functional NLRP inflammasome, the canonical pathway requires two steps: (i) a priming pro-inflammatory actor recruitment or transcription, here following DAMP/PAMP recognition by a PRR, and (ii) induction of the assembly of an active multiprotein complex. Priming can allow for the transcription of NLRPs, ASC, pro-caspase 1 and pro-interleukins 18 and 1β. Furthermore, non-transcriptional priming can occur and modulate the post-translational modifications required for inflammasome availability [119,120,121,122]. Once the inflammasome is assembled, the pro-caspase 1 is maturated in caspase 1, which cleaves pro-IL18 and pro-IL1β in active forms (triggering inflammation), and a protein named gasdermin D leads to the generation of an active N-terminal portion. Such final active portions assemble to form pores at the cell membrane, inducing pyroptosis (i.e., cell lysis) and inflammation through the consequent release of intracellular contents, including IL18, IL1β and inflammasomes specks, provoking an amplification of the inflammatory signal and immune cell recruitment [123,124,125] (Figure 6).
Thus, regarding their inflammatory action, NLRP inflammasomes are already implicated in different pathogeneses, as is the case more often in pathological pregnancies [127,128,129,130,131,132,133].

4.2. NLRP Inflammasomes, FM Diseases and pPROM

Studies on the pathological implication of NLRP inflammasomes (especially NLRP3) have focused on preeclampsia, an obstetrical disease where the uric acid levels are particularly increased in the blood [127,134,135]. Indeed, uric acid has been found to provoke an ASC-dependent activation of caspase 1 and IL1β release in vitro, which is associated with an overexpression of NLRP3 in human first trimester trophoblasts [136,137].
Furthermore, in the case of chorioamnionitis, a risk factor of pPROM, FM exhibits higher expression of inflammasome actors. In addition, one study demonstrated that AF from mothers with intra-amniotic inflammation (IAI) or infection contains more important ASC levels [138,139]. These data implicate NLRP inflammasomes in septic or sterile inflammation occurring during chorioamnionitis, a disease affecting FM barrier integrity.
In preterm labor, the concentration of caspase-1 in the AF is higher if labor is associated with infection and/or IAI when compared with a situation of preterm labor without infection and/or inflammation [140]. Gasdermin D has also been detected in the AF and FM in preterm labor, suggesting that pyroptosis may be involved in preterm labor and PTB [141]. Therefore, preterm labor seems to involve inflammasomes, especially in the case of infection or IAI [142]. In addition, Motomura et al. demonstrated in vivo that IL1α induces PTB through the NLRP3 inflammasome [143]. This NLRP3 implication is confirmed by Faro’s team, who induced PTB in an animal model by LPS-induced IAI, a phenomenon that was reduced by the use of a specific inhibitor of NLRP3 [144].
Finally, one study shows that in pPROM, the expression of transcripts and proteins related to the inflammasome signaling pathway (Nucleotide-binding oligomerization domain-containing protein 1 (NOD1) and NFκB) are increased in the placenta and FM [145]. In addition, Theis et al. examined the AF in mothers undergoing pPROM. The results revealed a positive correlation between microbial burden and ASC and IL6 concentrations [146]. Recently, another study highlighted higher expression levels of NLRP3, NLRP1 inflammasomes, IL18 and IL1β into the placenta and FM in the case of pPROM [147]. To conclude, all of these data demonstrate a clear link between NLRP inflammasomes and pPROM.

4.3. Focus on NLRP7

As detailed before, the NLRP family is composed of 14 members. Personal unpublished data identified the NLRP members expressed in the human amnion and chorion (collected at term) but also the inflammasome actors ASC and caspase-1 (Table 1).
In the obstetrical sphere, studies have been more focused on NLRP3, sometimes NLRP1, but some new data have recently emerged on NLRP7. In a more functional view, the NLRP7 inflammasome has been described as being involved in the activation of caspase-1 and IL1β release following infection of THP-1 cells (monocytes cell line) by Mycobacterium bovis. This activation of the NLRP7 inflammasome induced cell death, which the authors suggested to be pyroptosis [148]. Another team focused on the post-translational modifications regulating NLRP7. Indeed, the NLRP7 protein is constitutively ubiquitinated and recruited to the endolysosome to be degraded. Upon stimulation by a pro-inflammatory ligand, the STAMBP (Signal transducing adaptor molecule binding protein) enzyme deubiquitinates NLRP7, preventing its degradation and allowing the formation of an active inflammasome that activates IL1β [149]. Finally, Abi Nahed et al. recently demonstrated that the activation of the NLRP7 inflammasome in a trophoblastic cell line induced the secretion of IL1β but not IL18 [150]. Numerous studies have described the involvement of the NLRP7 inflammasome in some obstetrical complications, such as the recurrent hydatidiform mole (a disease characterized by an excessive trophoblastic proliferation without embryonic development), especially those related to mutations of NLRP7 [151,152,153,154,155]. In addition, the NLRP7 inflammasome is highly expressed by placental trophoblast cells and is deregulated in fetal growth restrictions, a disorder importantly related to abnormal placental development [150]. In addition, Tsai et al. demonstrated that NLRP7 relates to decidualization and macrophage differentiation, supporting their hypothesis of a multitasking gene implicated in endometrial hemostasis and reproductive success [156].
To conclude, NLRP7 seems important for inflammation response and pregnancy success. This is why, in the last part of this review, special emphasis will be placed on the emerging data about its implication in FM inflammation establishment.

4.4. Does NLRP7 Play a Role in FM Rupture?

The presence of the three NLRP7 inflammasome actors (ASC, caspase 1 and NLRP7) has been characterized in the amnion and chorion but also in primary amniotic epithelial cells (AECs) that were collected from the FM of women with term deliveries. It suggests that this NLRP7 inflammasome can assemble in the FM at all stages of pregnancy [157]. According to the literature, only one ligand is known and has been used to specifically activate the NLRP7 inflammasome: fibroblast stimulating lipopeptide (FSL-1) [158]. FSL-1 is a synthetic lipopeptide derived from the lipoprotein of Mycoplasma salivarium, representing the N-terminal portion of the LP-44 lipoprotein of this mycoplasma [159]. It is strongly similar to another lipopeptide, MALP-2 (Macrophage-activating lipopeptide-2), which is derived from the lipoprotein of Mycoplasma fermentans [160]. Numerous studies have used FSL-1 on FM, amnion cells or myometrial cells [22,161,162,163,164,165,166,167,168,169,170]; they allowed for the characterization of the signaling pathway induced by FSL-1, and they demonstrated an induction of the synthesis of pro-inflammatory mediators. The NLRP7 inflammasome, though specifically activated by this ligand, has been studied in the FM context in only one study [157].
Thus, Lavergne et al. demonstrated that AECs can form a functional NLRP7 inflammasome. More interestingly, the quantification of the expression of these three actors in the FM revealed an overexpression of ASC (in both layers of FM) at term with labor compared with term FM without labor [157]. This observation confirms the study of Gomez-Lopez et al., who described an increase of ASC/Caspase-1 complexes in the FM with labor [138]. No difference was found for NLRP7, whereas Romero et al. demonstrated an overexpression of NLRP3 protein in the FM with labor [171].
Furthermore, as previously mentioned, NLRP7 is activated by FSL-1, which is derived from M. salivarium, and similar to a lipoprotein from M. fermentans. Mycoplasmas have been studied extensively for their involvement in pPROM pathogenesis. Numerous mycoplasmas have been identified in the genital tract (M. genitalium, M. hominis, M. fermentans, M. penetrans, Ureaplasma spp., etc.), and some of them have been found to be related to pPROM or chorioamnionitis [172,173,174,175,176,177,178,179]. Moreover, Lavergne et al. found the presence of M. fermentans and M. salivarium in healthy human FM after vaginal deliveries but also after cesarean deliveries, proving that bacteria detection was not because of contamination after crossing the genital tract. These observations imply a genital tract ascension, even in healthy carriers. However, it can be supposed that overproliferation might lead to a pathological outcome, such as for the other mycoplasmas described in the literature.
In the aforementioned study, the M. salivarium-derived lipopeptide FSL-1 not only induced the expression of NLRP7 and caspase-1 in the AECs but also allowed the colocalization of ASC and NLRP7 around the nuclei, proving inflammasome assembly. In parallel, FSL-1 leads to an increase of IL1β transcription but not IL18, bringing to the fore the study of Abi Nahed et al. on trophoblasts, where only an induction of IL1β was found in response to NLRP7 inflammasome [150]. In addition, FSL-1 induces the cleavage of gasdermin D into its pyroptotic fragment. These experiments prove the importance of the NLRP family members, which are directly linked to cell death, inflammation and DAMP release, all of which are mechanisms involved in FM rupture.

5. Conclusion

This review has summarized recent data about the implication of inflammation in FM weakening. Furthermore, two new actors seem particularly interesting to explore for their inflammatory ability: RAGE and NLRP7. Both might be overactivated in risk contexts (smoking, germs rising in excess, chorioamnionitis) and be involved in pPROM pathogenesis. More studies are required to know in detail the implication of RAGE and its adaptors, but also of NLRP inflammasome pathophysiological weakening of FM to elucidate the link between the materno-fetal exposome, the PAMP/DAMPome and pPROM.

Author Contributions

H.C. wrote the manuscript with the help of M.L.; C.B., K.C., C.D.S.D.O., L.B. and V.S. revised the manuscript. All authors have read and approved the published version of the manuscript.

Funding

H.C. was funded by a French ministerial grant. M.L. was supported by Cancéropôle Lyon Auvergne Rhône-Alpes and the Région Auvergne Rhône-Alpes.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Thanks to Nadia Alfaidy for editing the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Naeye, R.L.; Peters, E.C. Causes and Consequences of Premature Rupture of Fetal Membranes. Lancet 1980, 1, 192–194. [Google Scholar] [CrossRef]
  2. Romero, R.; Espinoza, J.; Kusanovic, J.P.; Gotsch, F.; Hassan, S.; Erez, O.; Chaiworapongsa, T.; Mazor, M. The Preterm Parturition Syndrome. BJOG 2006, 113 (Suppl. 3), 17–42. [Google Scholar] [CrossRef]
  3. Menon, R. Human Fetal Membranes at Term: Dead Tissue or Signalers of Parturition? Placenta 2016, 44, 1–5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Richardson, L.S.; Vargas, G.; Brown, T.; Ochoa, L.; Sheller-Miller, S.; Saade, G.R.; Taylor, R.N.; Menon, R. Discovery and Characterization of Human Amniochorionic Membrane Microfractures. Am. J. Pathol. 2017, 187, 2821–2830. [Google Scholar] [CrossRef] [Green Version]
  5. Lavery, J.P.; Miller, C.E. The Viscoelastic Nature of Chorioamniotic Membranes. Obstet. Gynecol. 1977, 50, 467–472. [Google Scholar]
  6. Oyen, M.L.; Calvin, S.E.; Landers, D.V. Premature Rupture of the Fetal Membranes: Is the Amnion the Major Determinant? Am. J. Obstet. Gynecol. 2006, 195, 510–515. [Google Scholar] [CrossRef] [PubMed]
  7. Verbruggen, S.W.; Oyen, M.L.; Phillips, A.T.M.; Nowlan, N.C. Function and Failure of the Fetal Membrane: Modelling the Mechanics of the Chorion and Amnion. PLoS ONE 2017, 12, e0171588. [Google Scholar] [CrossRef] [Green Version]
  8. Prat, C.; Blanchon, L.; Borel, V.; Gallot, D.; Herbet, A.; Bouvier, D.; Marceau, G.; Sapin, V. Ontogeny of Aquaporins in Human Fetal Membranes. Biol. Reprod. 2012, 86, 48. [Google Scholar] [CrossRef]
  9. Prat, C.; Bouvier, D.; Comptour, A.; Marceau, G.; Belville, C.; Clairefond, G.; Blanc, P.; Gallot, D.; Blanchon, L.; Sapin, V. All-Trans-Retinoic Acid Regulates Aquaporin-3 Expression and Related Cellular Membrane Permeability in the Human Amniotic Environment. Placenta 2015, 36, 881–887. [Google Scholar] [CrossRef]
  10. Kobayashi, K.; Yasui, M. Cellular and Subcellular Localization of Aquaporins 1, 3, 8, and 9 in Amniotic Membranes during Pregnancy in Mice. Cell Tissue Res. 2010, 342, 307–316. [Google Scholar] [CrossRef]
  11. Ashraf, H.; Font, K.; Powell, C.; Schurr, M. Antimicrobial Activity of an Amnion-Chorion Membrane to Oral Microbes. Int. J. Dent. 2019, 2019, 1269534. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Szukiewicz, D.; Alkhalayla, H.; Pyzlak, M.; Watroba, M.; Szewczyk, G.; Wejman, J. Human Beta-Defensin 1, 2 and 3 Production by Amniotic Epithelial Cells with Respect to Human Papillomavirus (HPV) Infection, HPV Oncogenic Potential and the Mode of Delivery. Microb. Pathog. 2016, 97, 154–165. [Google Scholar] [CrossRef]
  13. Zare-Bidaki, M.; Sadrinia, S.; Erfani, S.; Afkar, E.; Ghanbarzade, N. Antimicrobial Properties of Amniotic and Chorionic Membranes: A Comparative Study of Two Human Fetal Sacs. J. Reprod. Infertil. 2017, 18, 218–224. [Google Scholar]
  14. Kobayashi, K.; Miwa, H.; Yasui, M. Inflammatory Mediators Weaken the Amniotic Membrane Barrier through Disruption of Tight Junctions. J. Physiol. 2010, 588, 4859–4869. [Google Scholar] [CrossRef]
  15. Kobayashi, K.; Kadohira, I.; Tanaka, M.; Yoshimura, Y.; Ikeda, K.; Yasui, M. Expression and Distribution of Tight Junction Proteins in Human Amnion during Late Pregnancy. Placenta 2010, 31, 158–162. [Google Scholar] [CrossRef]
  16. Caughey, A.B.; Robinson, J.N.; Norwitz, E.R. Contemporary Diagnosis and Management of Preterm Premature Rupture of Membranes. Rev. Obstet. Gynecol. 2008, 1, 11–22. [Google Scholar] [PubMed]
  17. Moore, R.M.; Mansour, J.M.; Redline, R.W.; Mercer, B.M.; Moore, J.J. The Physiology of Fetal Membrane Rupture: Insight Gained from the Determination of Physical Properties. Placenta 2006, 27, 1037–1051. [Google Scholar] [CrossRef] [PubMed]
  18. El Khwad, M.; Stetzer, B.; Moore, R.M.; Kumar, D.; Mercer, B.; Arikat, S.; Redline, R.W.; Mansour, J.M.; Moore, J.J. Term Human Fetal Membranes Have a Weak Zone Overlying the Lower Uterine Pole and Cervix Before Onset of Labor1. Biol. Reprod. 2005, 72, 720–726. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Chai, M.; Barker, G.; Menon, R.; Lappas, M. Increased Oxidative Stress in Human Fetal Membranes Overlying the Cervix from Term Non-Labouring and Post Labour Deliveries. Placenta 2012, 33, 604–610. [Google Scholar] [CrossRef]
  20. Chai, M.; Walker, S.P.; Riley, C.; Rice, G.E.; Permezel, M.; Lappas, M. Effect of Supracervical Apposition and Spontaneous Labour on Apoptosis and Matrix Metalloproteinases in Human Fetal Membranes. Biomed. Res. Int. 2013, 2013, 316146. [Google Scholar] [CrossRef] [Green Version]
  21. Colatto, B.N.; de Souza, I.F.; Schinke, L.A.A.; Noda-Nicolau, N.M.; da Silva, M.G.; Morceli, G.; Menon, R.; Polettini, J. Telomere Length and Telomerase Activity in Foetal Membranes from Term and Spontaneous Preterm Births. Reprod. Sci. 2020, 27, 411–417. [Google Scholar] [CrossRef] [PubMed]
  22. Lim, R.; Barker, G.; Lappas, M. SMAD7 Regulates Proinflammatory and Prolabor Mediators in Amnion and Myometrium. Biol. Reprod. 2017, 97, 288–301. [Google Scholar] [CrossRef] [Green Version]
  23. Richardson, L.S.; Taylor, R.N.; Menon, R. Reversible EMT and MET Mediate Amnion Remodeling during Pregnancy and Labor. Sci. Signal. 2020, 13, eaay1486. [Google Scholar] [CrossRef] [PubMed]
  24. Marcellin, L.; Schmitz, T.; Messaoudene, M.; Chader, D.; Parizot, C.; Jacques, S.; Delaire, J.; Gogusev, J.; Schmitt, A.; Lesaffre, C.; et al. Immune Modifications in Fetal Membranes Overlying the Cervix Precede Parturition in Humans. J. Immunol. 2017, 198, 1345–1356. [Google Scholar] [CrossRef]
  25. Nhan-Chang, C.-L.; Romero, R.; Tarca, A.L.; Mittal, P.; Kusanovic, J.P.; Erez, O.; Mazaki-Tovi, S.; Chaiworapongsa, T.; Hotra, J.; Than, N.G.; 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.e1–462.e41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Cox, S.M.; Casey, M.L.; MacDonald, P.C. Accumulation of Interleukin-1beta and Interleukin-6 in Amniotic Fluid: A Sequela of Labour at Term and Preterm. Hum. Reprod. Update 1997, 3, 517–527. [Google Scholar] [CrossRef]
  27. Maymon, E.; Ghezzi, F.; Edwin, S.S.; Mazor, M.; Yoon, B.H.; Gomez, R.; Romero, R. The Tumor Necrosis Factor Alpha and Its Soluble Receptor Profile in Term and Preterm Parturition. Am. J. Obstet. Gynecol. 1999, 181, 1142–1148. [Google Scholar] [CrossRef]
  28. Menon, R.; Lombardi, S.J.; Fortunato, S.J. TNF-Alpha Promotes Caspase Activation and Apoptosis in Human Fetal Membranes. J. Assist. Reprod. Genet. 2002, 19, 201–204. [Google Scholar] [CrossRef]
  29. Ito, A.; Nakamura, T.; Uchiyama, T.; Hirose, K.; Hirakawa, S.; Sasaguri, Y.; Mori, Y. Stimulation of the Biosynthesis of Interleukin 8 by Interleukin 1 and Tumor Necrosis Factor Alpha in Cultured Human Chorionic Cells. Biol. Pharm. Bull. 1994, 17, 1463–1467. [Google Scholar] [CrossRef] [Green Version]
  30. Trautman, M.S.; Dudley, D.J.; Edwin, S.S.; Collmer, D.; Mitchell, M.D. Amnion Cell Biosynthesis of Interleukin-8: Regulation by Inflammatory Cytokines. J. Cell Physiol. 1992, 153, 38–43. [Google Scholar] [CrossRef]
  31. Kumar, D.; Fung, W.; Moore, R.M.; Pandey, V.; Fox, J.; Stetzer, B.; Mansour, J.M.; Mercer, B.M.; Redline, R.W.; Moore, J.J. Proinflammatory Cytokines Found in Amniotic Fluid Induce Collagen Remodeling, Apoptosis, and Biophysical Weakening of Cultured Human Fetal Membranes1. Biol. Reprod. 2006, 74, 29–34. [Google Scholar] [CrossRef] [Green Version]
  32. Kumar, D.; Schatz, F.; Moore, R.; Mercer, B.; Rangaswamy, N.; Mansour, J.; Lockwood, C.; Moore, J. The Effects of Thrombin and Cytokines upon the Biomechanics and Remodeling of Isolated Amnion Membrane, In Vitro. Placenta 2011, 32, 206–213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Sheller-Miller, S.; Trivedi, J.; Yellon, S.M.; Menon, R. Exosomes Cause Preterm Birth in Mice: Evidence for Paracrine Signaling in Pregnancy. Sci. Rep. 2019, 9, 608. [Google Scholar] [CrossRef] [Green Version]
  34. Bredeson, S.; Papaconstantinou, J.; Deford, J.H.; Kechichian, T.; Syed, T.A.; Saade, G.R.; Menon, R. HMGB1 Promotes a P38MAPK Associated Non-Infectious Inflammatory Response Pathway in Human Fetal Membranes. PLoS ONE 2014, 9, e113799. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Lappas, M.; Permezel, M.; Rice, G.E. Advanced Glycation Endproducts Mediate Pro-Inflammatory Actions in Human Gestational Tissues via Nuclear Factor- B and Extracellular Signal-Regulated Kinase 1/2. J. Endocrinol. 2007, 193, 269–277. [Google Scholar] [CrossRef]
  36. Olive, C. Pattern Recognition Receptors: Sentinels in Innate Immunity and Targets of New Vaccine Adjuvants. Expert Rev. Vaccines 2012, 11, 237–256. [Google Scholar] [CrossRef]
  37. Hangai, S.; Kimura, Y.; Taniguchi, T.; Yanai, H. Signal Transducing Innate Receptors in Tumor Immunity. Cancer Sci. 2021, 112, 2578–2591. [Google Scholar] [CrossRef] [PubMed]
  38. Blanchon, L.; Accoceberry, M.; Belville, C.; Delabaere, A.; Prat, C.; Lemery, D.; Sapin, V.; Gallot, D. Rupture of membranes: Pathophysiology, diagnosis, consequences and management. J. Gynecol. Obstet. Biol. Reprod. 2013, 42, 105–116. [Google Scholar] [CrossRef]
  39. Menon, R.; Richardson, L.S. Preterm Prelabor Rupture of the Membranes: A Disease of the Fetal Membranes. Semin. Perinatol. 2017, 41, 409–419. [Google Scholar] [CrossRef]
  40. Furman, B.; Shoham-Vardi, I.; Bashiri, A.; Erez, O.; Mazor, M. Clinical Significance and Outcome of Preterm Prelabor Rupture of Membranes: Population-Based Study. Eur. J. Obstet. Gynecol. Reprod. Biol. 2000, 92, 209–216. [Google Scholar] [CrossRef]
  41. Ananth, C.V.; Oyelese, Y.; Srinivas, N.; Yeo, L.; Vintzileos, A.M. Preterm Premature Rupture of Membranes, Intrauterine Infection, and Oligohydramnios: Risk Factors for Placental Abruption. Obstet. Gynecol. 2004, 104, 71–77. [Google Scholar] [CrossRef] [PubMed]
  42. Middleton, P.; Shepherd, E.; Flenady, V.; McBain, R.D.; Crowther, C.A. Planned Early Birth versus Expectant Management (Waiting) for Prelabour Rupture of Membranes at Term (37 Weeks or More). Cochrane Database Syst. Rev. 2017, 1, CD005302. [Google Scholar] [CrossRef]
  43. Newton, E.R. Preterm Labor, Preterm Premature Rupture of Membranes, and Chorioamnionitis. Clin. Perinatol. 2005, 32, 571–600. [Google Scholar] [CrossRef]
  44. Platt, M.J. Outcomes in Preterm Infants. Public Health 2014, 128, 399–403. [Google Scholar] [CrossRef] [PubMed]
  45. Bouvier, D.; Forest, J.-C.; Blanchon, L.; Bujold, E.; Pereira, B.; Bernard, N.; Gallot, D.; Sapin, V.; Giguère, Y. Risk Factors and Outcomes of Preterm Premature Rupture of Membranes in a Cohort of 6968 Pregnant Women Prospectively Recruited. J. Clin. Med. 2019, 8, 1987. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Forde, B.; Habli, M. Unique Considerations: Preterm Prelabor Rupture of Membranes in the Setting of Fetal Surgery and Higher Order Pregnancies. Obstet. Gynecol. Clin. N. Am. 2020, 47, 653–669. [Google Scholar] [CrossRef] [PubMed]
  47. Modi, B.P.; Teves, M.E.; Pearson, L.N.; Parikh, H.I.; Haymond-Thornburg, H.; Tucker, J.L.; Chaemsaithong, P.; Gomez-Lopez, N.; York, T.P.; Romero, R.; et al. Mutations in Fetal Genes Involved in Innate Immunity and Host Defense against Microbes Increase Risk of Preterm Premature Rupture of Membranes (PPROM). Mol. Genet. Genom. Med. 2017, 5, 720–729. [Google Scholar] [CrossRef] [Green Version]
  48. Dorfeuille, N.; Morin, V.; Tétu, A.; Demers, S.; Laforest, G.; Gouin, K.; Piedboeuf, B.; Bujold, E. Vaginal Fluid Inflammatory Biomarkers and the Risk of Adverse Neonatal Outcomes in Women with PPROM. Am. J. Perinatol. 2016, 33, 1003–1007. [Google Scholar] [CrossRef]
  49. Galaz, J.; Romero, R.; Slutsky, R.; Xu, Y.; Motomura, K.; Para, R.; Pacora, P.; Panaitescu, B.; Hsu, C.-D.; Kacerovsky, M.; et al. Cellular Immune Responses in Amniotic Fluid of Women with Preterm Prelabor Rupture of Membranes. J. Perinat. Med. 2020, 48, 222–233. [Google Scholar] [CrossRef]
  50. Romero, R.; Miranda, J.; Chaemsaithong, P.; Chaiworapongsa, T.; Kusanovic, J.P.; Dong, Z.; Ahmed, A.I.; Shaman, M.; Lannaman, K.; Yoon, B.H.; et al. Sterile and Microbial-Associated Intra-Amniotic Inflammation in Preterm Prelabor Rupture of Membranes. J. Matern.-Fetal Neonatal Med. 2015, 28, 1394–1409. [Google Scholar] [CrossRef] [PubMed]
  51. Menon, R.; Lombardi, S.J.; Fortunato, S.J. IL-18, a Product of Choriodecidual Cells, Increases during Premature Rupture of Membranes but Fails to Turn on the Fas-FasL-Mediated Apoptosis Pathway. J. Assist. Reprod. Genet. 2001, 18, 276–284. [Google Scholar] [CrossRef] [PubMed]
  52. Shobokshi, A.; Shaarawy, M. Maternal Serum and Amniotic Fluid Cytokines in Patients with Preterm Premature Rupture of Membranes with and without Intrauterine Infection. Int. J. Gynaecol. Obstet. 2002, 79, 209–215. [Google Scholar] [CrossRef]
  53. Vajrychová, M.; Stráník, J.; Pimková, K.; Barman, M.; Kukla, R.; Zedníková, P.; Bolehovská, R.; Plíšková, L.; Hornychová, H.; Andrýs, C.; et al. Comprehensive Proteomic Investigation of Infectious and Inflammatory Changes in Late Preterm Prelabour Rupture of Membranes. Sci. Rep. 2020, 10, 17696. [Google Scholar] [CrossRef] [PubMed]
  54. Gomez-Lopez, N.; Romero, R.; Leng, Y.; Xu, Y.; Slutsky, R.; Levenson, D.; Pacora, P.; Jung, E.; Panaitescu, B.; Hsu, C.-D. The Origin of Amniotic Fluid Monocytes/Macrophages in Women with Intra-Amniotic Inflammation or Infection. J. Perinat. Med. 2019, 47, 822–840. [Google Scholar] [CrossRef]
  55. Ronzoni, S.; Steckle, V.; D’Souza, R.; Murphy, K.E.; Lye, S.; Shynlova, O. Cytokine Changes in Maternal Peripheral Blood Correlate With Time-to-Delivery in Pregnancies Complicated by Premature Prelabor Rupture of the Membranes. Reprod. Sci. 2019, 26, 1266–1276. [Google Scholar] [CrossRef] [PubMed]
  56. Raba, G.; Kacerovsky, M.; Laudański, P. Eotaxin-2 as a Potential Marker of Preterm Premature Rupture of Membranes: A Prospective, Cohort, Multicenter Study. Adv. Clin. Exp. Med. 2021, 30, 197–202. [Google Scholar] [CrossRef]
  57. Seliger, G.; Bergner, M.; Haase, R.; Stepan, H.; Schleußner, E.; Zöllkau, J.; Seeger, S.; Kraus, F.B.; Hiller, G.G.R.; Wienke, A.; et al. Daily Monitoring of Vaginal Interleukin 6 as a Predictor of Intraamniotic Inflammation after Preterm Premature Rupture of Membranes—A New Method of Sampling Studied in a Prospective Multicenter Trial. J. Perinat. Med. 2021, 49, 572–582. [Google Scholar] [CrossRef]
  58. Li, W.; Zhao, X.; Li, S.; Chen, X.; Cui, H.; Chang, Y.; Zhang, R. Upregulation of TNF-α and IL-6 Induces Preterm Premature Rupture of Membranes by Activation of ADAMTS-9 in Embryonic Membrane Cells. Life Sci. 2020, 260, 118237. [Google Scholar] [CrossRef]
  59. Zaga, V.; Estrada-Gutierrez, G.; Beltran-Montoya, J.; Maida-Claros, R.; Lopez-Vancell, R.; Vadillo-Ortega, F. Secretions of Interleukin-1beta and Tumor Necrosis Factor Alpha by Whole Fetal Membranes Depend on Initial Interactions of Amnion or Choriodecidua with Lipopolysaccharides or Group B Streptococci. Biol. Reprod. 2004, 71, 1296–1302. [Google Scholar] [CrossRef]
  60. Zaga-Clavellina, V.; Garcia-Lopez, G.; Flores-Herrera, H.; Espejel-Nuñez, A.; Flores-Pliego, A.; Soriano-Becerril, D.; Maida-Claros, R.; Merchant-Larios, H.; Vadillo-Ortega, F. In Vitro Secretion Profiles of Interleukin (IL)-1beta, IL-6, IL-8, IL-10, and TNF Alpha after Selective Infection with Escherichia Coli in Human Fetal Membranes. Reprod. Biol. Endocrinol. 2007, 5, 46. [Google Scholar] [CrossRef] [Green Version]
  61. Romero, R.; Miranda, J.; Chaiworapongsa, T.; Korzeniewski, S.J.; Chaemsaithong, P.; Gotsch, F.; Dong, Z.; Ahmed, A.I.; Yoon, B.H.; Hassan, S.S.; 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, 458–474. [Google Scholar] [CrossRef] [Green Version]
  62. Baumbusch, M.A.; Buhimschi, C.S.; Oliver, E.A.; Zhao, G.; Thung, S.; Rood, K.; Buhimschi, I.A. High Mobility Group-Box 1 (HMGB1) Levels Are Increased in Amniotic Fluid of Women with Intra-Amniotic Inflammation-Determined Preterm Birth, and the Source May Be the Damaged Fetal Membranes. Cytokine 2016, 81, 82–87. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Yan, H.; Zhu, L.; Zhang, Z.; Li, H.; Li, P.; Wang, Y.; Leng, M. HMGB1-RAGE Signaling Pathway in PPROM. Taiwan. J. Obstet. Gynecol. 2018, 57, 211–216. [Google Scholar] [CrossRef]
  64. Chaiworapongsa, T.; Erez, O.; Kusanovic, J.P.; Vaisbuch, E.; Mazaki-Tovi, S.; Gotsch, F.; Than, N.G.; Mittal, P.; Kim, Y.M.; Camacho, N.; et al. Amniotic Fluid Heat Shock Protein 70 Concentration in Histologic Chorioamnionitis, Term and Preterm Parturition. J. Matern. Fetal. Neonatal Med. 2008, 21, 449–461. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Friel, L.A.; Romero, R.; Edwin, S.; Nien, J.K.; Gomez, R.; Chaiworapongsa, T.; Kusanovic, J.P.; Tolosa, J.E.; Hassan, S.S.; Espinoza, J. The Calcium Binding Protein, S100B, Is Increased in the Amniotic Fluid of Women with Intra-Amniotic Infection/Inflammation and Preterm Labor with Intact or Ruptured Membranes. J. Perinat. Med. 2007, 35, 385–393. [Google Scholar] [CrossRef] [PubMed]
  66. Romero, R.; Chaiworapongsa, T.; Savasan, Z.A.; Xu, Y.; Hussein, Y.; Dong, Z.; Kusanovic, J.P.; Kim, C.J.; Hassan, S.S. Damage-Associated Molecular Patterns (DAMPs) in Preterm Labor with Intact Membranes and Preterm PROM: A Study of the Alarmin HMGB1. J. Matern. Fetal. Neonatal Med. 2011, 24, 1444–1455. [Google Scholar] [CrossRef]
  67. van Boeckel, S.R.; Davidson, D.J.; Norman, J.E.; Stock, S.J. Cell-Free Fetal DNA and Spontaneous Preterm Birth. Reproduction 2018, 155, R137–R145. [Google Scholar] [CrossRef] [Green Version]
  68. Golubinskaya, V.; Puttonen, H.; Fyhr, I.-M.; Rydbeck, H.; Hellström, A.; Jacobsson, B.; Nilsson, H.; Mallard, C.; Sävman, K. Expression of S100A Alarmins in Cord Blood Monocytes Is Highly Associated With Chorioamnionitis and Fetal Inflammation in Preterm Infants. Front. Immunol. 2020, 11, 1194. [Google Scholar] [CrossRef] [PubMed]
  69. Kacerovsky, M.; Vlkova, B.; Musilova, I.; Andrys, C.; Pliskova, L.; Zemlickova, H.; Stranik, J.; Halada, P.; Jacobsson, B.; Celec, P. Amniotic Fluid Cell-Free DNA in Preterm Prelabor Rupture of Membranes. Prenat. Diagn. 2018, 38, 1086–1095. [Google Scholar] [CrossRef]
  70. Menon, R.; Behnia, F.; Polettini, J.; Saade, G.R.; Campisi, J.; Velarde, M. Placental Membrane Aging and HMGB1 Signaling Associated with Human Parturition. Aging 2016, 8, 216–230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Padron, J.G.; Saito Reis, C.A.; Kendal-Wright, C.E. The Role of Danger Associated Molecular Patterns in Human Fetal Membrane Weakening. Front. Physiol. 2020, 11, 602. [Google Scholar] [CrossRef]
  72. Sheller-Miller, S.; Radnaa, E.; Yoo, J.-K.; Kim, E.; Choi, K.; Kim, Y.; Kim, Y.N.; Richardson, L.; Choi, C.; Menon, R. Exosomal Delivery of NF-ΚB Inhibitor Delays LPS-Induced Preterm Birth and Modulates Fetal Immune Cell Profile in Mouse Models. Sci. Adv. 2021, 7, eabd3865. [Google Scholar] [CrossRef]
  73. Bongarzone, S.; Savickas, V.; Luzi, F.; Gee, A.D. Targeting the Receptor for Advanced Glycation Endproducts (RAGE): A Medicinal Chemistry Perspective. J. Med. Chem. 2017, 60, 7213–7232. [Google Scholar] [CrossRef] [Green Version]
  74. Barbezier, N.; Tessier, F.J.; Chango, A. Receptor of advanced glycation endproducts RAGE/AGER: An integrative view for clinical applications. Ann. Biol. Clin. 2014, 72, 669–680. [Google Scholar] [CrossRef]
  75. Kierdorf, K.; Fritz, G. RAGE Regulation and Signaling in Inflammation and Beyond. J. Leukoc. Biol. 2013, 94, 55–68. [Google Scholar] [CrossRef]
  76. Sorci, G.; Riuzzi, F.; Giambanco, I.; Donato, R. RAGE in Tissue Homeostasis, Repair and Regeneration. Biochim. Biophys. Acta (BBA)-Mol. Cell Res. 2013, 1833, 101–109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Hudson, B.I.; Kalea, A.Z.; Del Mar Arriero, M.; Harja, E.; Boulanger, E.; D’Agati, V.; Schmidt, A.M. Interaction of the RAGE Cytoplasmic Domain with Diaphanous-1 Is Required for Ligand-Stimulated Cellular Migration through Activation of Rac1 and Cdc42. J. Biol. Chem. 2008, 283, 34457–34468. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Sakaguchi, M.; Murata, H.; Yamamoto, K.; Ono, T.; Sakaguchi, Y.; Motoyama, A.; Hibino, T.; Kataoka, K.; Huh, N. TIRAP, an Adaptor Protein for TLR2/4, Transduces a Signal from RAGE Phosphorylated upon Ligand Binding. PLoS ONE 2011, 6, e23132. [Google Scholar] [CrossRef] [Green Version]
  79. Raucci, A.; Cugusi, S.; Antonelli, A.; Barabino, S.M.; Monti, L.; Bierhaus, A.; Reiss, K.; Saftig, P.; Bianchi, M.E. A Soluble Form of the Receptor for Advanced Glycation Endproducts (RAGE) Is Produced by Proteolytic Cleavage of the Membrane-Bound Form by the Sheddase a Disintegrin and Metalloprotease 10 (ADAM10). FASEB J. 2008, 22, 3716–3727. [Google Scholar] [CrossRef]
  80. Zhang, L.; Bukulin, M.; Kojro, E.; Roth, A.; Metz, V.V.; Fahrenholz, F.; Nawroth, P.P.; Bierhaus, A.; Postina, R. Receptor for Advanced Glycation End Products Is Subjected to Protein Ectodomain Shedding by Metalloproteinases. J. Biol. Chem. 2008, 283, 35507–35516. [Google Scholar] [CrossRef] [Green Version]
  81. Teissier, T.; Boulanger, É. The Receptor for Advanced Glycation End-Products (RAGE) Is an Important Pattern Recognition Receptor (PRR) for Inflammaging. Biogerontology 2019, 20, 279–301. [Google Scholar] [CrossRef]
  82. Schmidt, A.M.; Yan, S.D.; Yan, S.F.; Stern, D.M. The Biology of the Receptor for Advanced Glycation End Products and Its Ligands. Biochim. Biophys. Acta 2000, 1498, 99–111. [Google Scholar] [CrossRef] [Green Version]
  83. Sakaguchi, M.; Kinoshita, R.; Putranto, E.W.; Ruma, I.M.W.; Sumardika, I.W.; Youyi, C.; Tomonobu, N.; Yamamoto, K.-I.; Murata, H. Signal Diversity of Receptor for Advanced Glycation End Products. Acta Med. Okayama 2017, 71, 459–465. [Google Scholar] [CrossRef]
  84. Tsoporis, J.N.; Izhar, S.; Leong-Poi, H.; Desjardins, J.-F.; Huttunen, H.J.; Parker, T.G. S100B Interaction With the Receptor for Advanced Glycation End Products (RAGE): A Novel Receptor-Mediated Mechanism for Myocyte Apoptosis Postinfarction. Circ. Res. 2010, 106, 93–101. [Google Scholar] [CrossRef] [PubMed]
  85. Zhang, Q.; Raoof, M.; Chen, Y.; Sumi, Y.; Sursal, T.; Junger, W.; Brohi, K.; Itagaki, K.; Hauser, C.J. Circulating Mitochondrial DAMPs Cause Inflammatory Responses to Injury. Nature 2010, 464, 104–107. [Google Scholar] [CrossRef] [Green Version]
  86. Hofmann, M.A.; Drury, S.; Fu, C.; Qu, W.; Taguchi, A.; Lu, Y.; Avila, C.; Kambham, N.; Bierhaus, A.; Nawroth, P.; et al. RAGE Mediates a Novel Proinflammatory Axis: A Central Cell Surface Receptor for S100/Calgranulin Polypeptides. Cell 1999, 97, 889–901. [Google Scholar]
  87. Xie, J.; Méndez, J.D.; Méndez-Valenzuela, V.; Aguilar-Hernández, M.M. Cellular Signalling of the Receptor for Advanced Glycation End Products (RAGE). Cell. Signal. 2013, 25, 2185–2197. [Google Scholar] [CrossRef]
  88. Liliensiek, B.; Weigand, M.A.; Bierhaus, A.; Nicklas, W.; Kasper, M.; Hofer, S.; Plachky, J.; Gröne, H.-J.; Kurschus, F.C.; Schmidt, A.M.; et al. Receptor for Advanced Glycation End Products (RAGE) Regulates Sepsis but Not the Adaptive Immune Response. J. Clin. Investig. 2004, 113, 1641–1650. [Google Scholar] [CrossRef]
  89. Chen, Y.; Akirav, E.M.; Chen, W.; Henegariu, O.; Moser, B.; Desai, D.; Shen, J.M.; Webster, J.C.; Andrews, R.C.; Mjalli, A.M.; et al. RAGE Ligation Affects T Cell Activation and Controls T Cell Differentiation. J. Immunol. 2008, 181, 4272–4278. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Chuah, Y.K.; Basir, R.; Talib, H.; Tie, T.H.; Nordin, N. Receptor for Advanced Glycation End Products and Its Involvement in Inflammatory Diseases. Int. J. Inflamm. 2013, 2013, 403460. [Google Scholar] [CrossRef] [Green Version]
  91. Collison, K.S.; Parhar, R.S.; Saleh, S.S.; Meyer, B.F.; Kwaasi, A.A.; Hammami, M.M.; Schmidt, A.M.; Stern, D.M.; Al-Mohanna, F.A. RAGE-Mediated Neutrophil Dysfunction Is Evoked by Advanced Glycation End Products (AGEs). J. Leukoc. Biol. 2002, 71, 433–444. [Google Scholar] [PubMed]
  92. Yan, S.D.; Schmidt, A.M.; Anderson, G.M.; Zhang, J.; Brett, J.; Zou, Y.S.; Pinsky, D.; Stern, D. Enhanced Cellular Oxidant Stress by the Interaction of Advanced Glycation End Products with Their Receptors/Binding Proteins. J. Biol. Chem. 1994, 269, 9889–9897. [Google Scholar] [CrossRef]
  93. Li, J.; Schmidt, A.M. Characterization and Functional Analysis of the Promoter of RAGE, the Receptor for Advanced Glycation End Products. J. Biol. Chem. 1997, 272, 16498–16506. [Google Scholar] [CrossRef] [Green Version]
  94. Bianchi, M.E. DAMPs, PAMPs and Alarmins: All We Need to Know about Danger. J. Leukoc. Biol. 2007, 81, 1–5. [Google Scholar] [CrossRef]
  95. Bopp, C.; Bierhaus, A.; Hofer, S.; Bouchon, A.; Nawroth, P.P.; Martin, E.; Weigand, M.A. Bench-to-Bedside Review: The Inflammation-Perpetuating Pattern-Recognition Receptor RAGE as a Therapeutic Target in Sepsis. Crit. Care 2007, 12, 201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Romero, R.; Espinoza, J.; Hassan, S.; Gotsch, F.; Kusanovic, J.P.; Avila, C.; Erez, O.; Edwin, S.; Schmidt, A.M. Soluble Receptor for Advanced Glycation End Products (SRAGE) and Endogenous Secretory RAGE (EsRAGE) in Amniotic Fluid: Modulation by Infection and Inflammation. J. Perinat. Med. 2008, 36, 388–398. [Google Scholar] [CrossRef] [PubMed]
  97. Buhimschi, I.A.; Zhao, G.; Pettker, C.M.; Bahtiyar, M.O.; Magloire, L.K.; Thung, S.; Fairchild, T.; Buhimschi, C.S. The Receptor for Advanced Glycation End Products (RAGE) System in Women with Intraamniotic Infection and Inflammation. Am. J. Obstet. Gynecol. 2007, 196, 181.e1–181.e13. [Google Scholar] [CrossRef]
  98. Rzepka, R.; Dołęgowska, B.; Sałata, D.; Rajewska, A.; Budkowska, M.; Domański, L.; Kwiatkowski, S.; Mikołajek-Bedner, W.; Torbé, A. Soluble Receptors for Advanced Glycation End Products and Receptor Activator of NF-ΚB Ligand Serum Levels as Markers of Premature Labor. BMC Pregnancy Childbirth 2015, 15, 134. [Google Scholar] [CrossRef] [Green Version]
  99. Bouvier, D.; Giguère, Y.; Blanchon, L.; Bujold, E.; Pereira, B.; Bernard, N.; Gallot, D.; Sapin, V.; Forest, J.-C. Study of SRAGE, HMGB1, AGE, and S100A8/A9 Concentrations in Plasma and in Serum-Extracted Extracellular Vesicles of Pregnant Women With Preterm Premature Rupture of Membranes. Front. Physiol. 2020, 11, 609. [Google Scholar] [CrossRef] [PubMed]
  100. Qiu, X.-Y.; Sun, L.; Han, X.-L.; Chang, Y.; Cheng, L.; Yin, L.-R. Alarmin High Mobility Group Box-1 in Maternal Serum as a Potential Biomarker of Chorioamnionitis-Associated Preterm Birth. Gynecol. Endocrinol. 2017, 33, 128–131. [Google Scholar] [CrossRef] [PubMed]
  101. Kansu-Celik, H.; Tasci, Y.; Karakaya, B.K.; Cinar, M.; Candar, T.; Caglar, G.S. Maternal Serum Advanced Glycation End Products Level as an Early Marker for Predicting Preterm Labor/PPROM: A Prospective Preliminary Study. J. Matern. Fetal. Neonatal Med. 2019, 32, 2758–2762. [Google Scholar] [CrossRef] [PubMed]
  102. Pouwels, S.D.; Hesse, L.; Faiz, A.; Lubbers, J.; Bodha, P.K.; ten Hacken, N.H.T.; van Oosterhout, A.J.M.; Nawijn, M.C.; Heijink, I.H. Susceptibility for Cigarette Smoke-Induced DAMP Release and DAMP-Induced Inflammation in COPD. Am. J. Physiol. -Lung Cell. Mol. Physiol. 2016, 311, L881–L892. [Google Scholar] [CrossRef] [Green Version]
  103. Choltus, H.; Lavergne, M.; Belville, C.; Gallot, D.; Minet-Quinard, R.; Durif, J.; Blanchon, L.; Sapin, V. Occurrence of a RAGE-Mediated Inflammatory Response in Human Fetal Membranes. Front. Physiol. 2020, 11, 581. [Google Scholar] [CrossRef] [PubMed]
  104. Robertson, S.A.; Hutchinson, M.R.; Rice, K.C.; Chin, P.; Moldenhauer, L.M.; Stark, M.J.; Olson, D.M.; Keelan, J.A. Targeting Toll-like Receptor-4 to Tackle Preterm Birth and Fetal Inflammatory Injury. Clin. Transl. Immunol. 2020, 9, e1121. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Plazyo, O.; Romero, R.; Unkel, R.; Balancio, A.; Mial, T.N.; Xu, Y.; Dong, Z.; Hassan, S.S.; Gomez-Lopez, N. HMGB1 Induces an Inflammatory Response in the Chorioamniotic Membranes That Is Partially Mediated by the Inflammasome. Biol. Reprod. 2016, 95, 130. [Google Scholar] [CrossRef]
  106. Martinon, F.; Burns, K.; Tschopp, J. The Inflammasome: A Molecular Platform Triggering Activation of Inflammatory Caspases and Processing of ProIL-Beta. Mol. Cell 2002, 10, 417–426. [Google Scholar] [CrossRef]
  107. Gomez, A.; Serrano, A.; Salero, E.; Tovar, A.; Amescua, G.; Galor, A.; Keane, R.W.; Pablo de Rivero Vaccari, J.; Sabater, A.L. Tumor Necrosis Factor-Alpha and Interferon-Gamma Induce Inflammasome-Mediated Corneal Endothelial Cell Death. Exp. Eye Res. 2021, 207, 108574. [Google Scholar] [CrossRef]
  108. Paik, S.; Kim, J.K.; Silwal, P.; Sasakawa, C.; Jo, E.-K. An Update on the Regulatory Mechanisms of NLRP3 Inflammasome Activation. Cell Mol. Immunol. 2021, 18, 1141–1160. [Google Scholar] [CrossRef]
  109. de Vasconcelos, N.M.; Lamkanfi, M. Recent Insights on Inflammasomes, Gasdermin Pores, and Pyroptosis. Cold Spring Harb Perspect. Biol. 2020, 12, a036392. [Google Scholar] [CrossRef] [PubMed]
  110. Dubois, H.; Wullaert, A.; Lamkanfi, M. General Strategies in Inflammasome Biology. Curr Top. Microbiol. Immunol. 2016, 397, 1–22. [Google Scholar] [CrossRef] [PubMed]
  111. Singer, H.; Biswas, A.; Zimmer, N.; Messaed, C.; Oldenburg, J.; Slim, R.; El-Maarri, O. NLRP7 Inter-Domain Interactions: The NACHT-Associated Domain Is the Physical Mediator for Oligomeric Assembly. Mol. Hum. Reprod 2014, 20, 990–1001. [Google Scholar] [CrossRef] [Green Version]
  112. Istomin, A.Y.; Godzik, A. Understanding Diversity of Human Innate Immunity Receptors: Analysis of Surface Features of Leucine-Rich Repeat Domains in NLRs and TLRs. BMC Immunol. 2009, 10, 48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Yeretssian, G. Effector Functions of NLRs in the Intestine: Innate Sensing, Cell Death, and Disease. Immunol. Res. 2012, 54, 25–36. [Google Scholar] [CrossRef] [PubMed]
  114. Malik, A.; Kanneganti, T.-D. Inflammasome Activation and Assembly at a Glance. J. Cell Sci. 2017, 130, 3955–3963. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Dick, M.S.; Sborgi, L.; Rühl, S.; Hiller, S.; Broz, P. ASC Filament Formation Serves as a Signal Amplification Mechanism for Inflammasomes. Nat. Commun. 2016, 7, 11929. [Google Scholar] [CrossRef] [Green Version]
  116. Pop, C.; Salvesen, G.S. Human Caspases: Activation, Specificity, and Regulation. J. Biol. Chem. 2009, 284, 21777–21781. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Boucher, D.; Monteleone, M.; Coll, R.C.; Chen, K.W.; Ross, C.M.; Teo, J.L.; Gomez, G.A.; Holley, C.L.; Bierschenk, D.; Stacey, K.J.; et al. Caspase-1 Self-Cleavage Is an Intrinsic Mechanism to Terminate Inflammasome Activity. J. Exp. Med. 2018, 215, 827–840. [Google Scholar] [CrossRef]
  118. Kesavardhana, S.; Malireddi, R.K.S.; Kanneganti, T.-D. Caspases in Cell Death, Inflammation, and Pyroptosis. Annu. Rev. Immunol. 2020, 38, 567–595. [Google Scholar] [CrossRef] [Green Version]
  119. Groslambert, M.; Py, B.F. Spotlight on the NLRP3 Inflammasome Pathway. J. Inflamm Res. 2018, 11, 359–374. [Google Scholar] [CrossRef] [Green Version]
  120. Xiang, P.; Chen, T.; Mou, Y.; Wu, H.; Xie, P.; Lu, G.; Gong, X.; Hu, Q.; Zhang, Y.; Ji, H. NZ Suppresses TLR4/NF-ΚB Signalings and NLRP3 Inflammasome Activation in LPS-Induced RAW264.7 Macrophages. Inflamm. Res. 2015, 64, 799–808. [Google Scholar] [CrossRef]
  121. Juliana, C.; Fernandes-Alnemri, T.; Kang, S.; Farias, A.; Qin, F.; Alnemri, E.S. Non-Transcriptional Priming and Deubiquitination Regulate NLRP3 Inflammasome Activation. J. Biol. Chem. 2012, 287, 36617–36622. [Google Scholar] [CrossRef] [Green Version]
  122. Kesavardhana, S.; Kanneganti, T.-D. Mechanisms Governing Inflammasome Activation, Assembly and Pyroptosis Induction. Int. Immunol. 2017, 29, 201–210. [Google Scholar] [CrossRef]
  123. Evavold, C.L.; Ruan, J.; Tan, Y.; Xia, S.; Wu, H.; Kagan, J.C. The Pore-Forming Protein Gasdermin D Regulates Interleukin-1 Secretion from Living Macrophages. Immunity 2018, 48, 35–44.e6. [Google Scholar] [CrossRef] [Green Version]
  124. Monteleone, M.; Stanley, A.C.; Chen, K.W.; Brown, D.L.; Bezbradica, J.S.; von Pein, J.B.; Holley, C.L.; Boucher, D.; Shakespear, M.R.; Kapetanovic, R.; et al. Interleukin-1β Maturation Triggers Its Relocation to the Plasma Membrane for Gasdermin-D-Dependent and -Independent Secretion. Cell Rep. 2018, 24, 1425–1433. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. He, W.; Wan, H.; Hu, L.; Chen, P.; Wang, X.; Huang, Z.; Yang, Z.-H.; Zhong, C.-Q.; Han, J. Gasdermin D Is an Executor of Pyroptosis and Required for Interleukin-1β Secretion. Cell Res. 2015, 25, 1285–1298. [Google Scholar] [CrossRef]
  126. Chan, A.H.; Schroder, K. Inflammasome Signaling and Regulation of Interleukin-1 Family Cytokines. J. Exp. Med. 2019, 217, e20190314. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Zhou, F.; Li, C.; Zhang, S.-Y. NLRP3 Inflammasome: A New Therapeutic Target for High-Risk Reproductive Disorders? Chin. Med. J. 2020, 134, 20–27. [Google Scholar] [CrossRef]
  128. Lahooti, B.; Chhibber, T.; Bagchi, S.; Varahachalam, S.P.; Jayant, R.D. Therapeutic Role of Inflammasome Inhibitors in Neurodegenerative Disorders. Brain. Behav. Immun. 2021, 91, 771–783. [Google Scholar] [CrossRef]
  129. Sarmah, D.; Datta, A.; Raut, S.; Sarkar, A.; Shah, B.; Bohra, M.; Singh, U.; Jagtap, P.; Baidya, F.; Kalia, K.; et al. The Role of Inflammasomes in Atherosclerosis and Stroke Pathogenesis. Curr. Pharm. Des. 2020, 26, 4234–4245. [Google Scholar] [CrossRef]
  130. Moloudizargari, M.; Moradkhani, F.; Asghari, N.; Fallah, M.; Asghari, M.H.; Moghadamnia, A.A.; Abdollahi, M. NLRP Inflammasome as a Key Role Player in the Pathogenesis of Environmental Toxicants. Life Sci. 2019, 231, 116585. [Google Scholar] [CrossRef] [PubMed]
  131. Lee, C.; Do, H.T.T.; Her, J.; Kim, Y.; Seo, D.; Rhee, I. Inflammasome as a Promising Therapeutic Target for Cancer. Life Sci. 2019, 231, 116593. [Google Scholar] [CrossRef] [PubMed]
  132. Soellner, L.; Kopp, K.M.; Mütze, S.; Meyer, R.; Begemann, M.; Rudnik, S.; Rath, W.; Eggermann, T.; Zerres, K. NLRP Genes and Their Role in Preeclampsia and Multi-Locus Imprinting Disorders. J. Perinat. Med. 2018, 46, 169–173. [Google Scholar] [CrossRef]
  133. Stødle, G.S.; Silva, G.B.; Tangerås, L.H.; Gierman, L.M.; Nervik, I.; Dahlberg, U.E.; Sun, C.; Aune, M.H.; Thomsen, L.C.V.; Bjørge, L.; et al. Placental Inflammation in Pre-Eclampsia by Nod-like Receptor Protein (NLRP)3 Inflammasome Activation in Trophoblasts. Clin. Exp. Immunol. 2018, 193, 84–94. [Google Scholar] [CrossRef] [Green Version]
  134. Murthi, P.; Pinar, A.A.; Dimitriadis, E.; Samuel, C.S. Inflammasomes—A Molecular Link for Altered Immunoregulation and Inflammation Mediated Vascular Dysfunction in Preeclampsia. Int. J. Mol. Sci. 2020, 21, 1406. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Weel, I.C.; Romão-Veiga, M.; Matias, M.L.; Fioratti, E.G.; Peraçoli, J.C.; Borges, V.T.; Araujo, J.P.; Peraçoli, M.T. Increased Expression of NLRP3 Inflammasome in Placentas from Pregnant Women with Severe Preeclampsia. J. Reprod. Immunol. 2017, 123, 40–47. [Google Scholar] [CrossRef] [Green Version]
  136. Mulla, M.J.; Myrtolli, K.; Potter, J.; Boeras, C.; Kavathas, P.B.; Sfakianaki, A.K.; Tadesse, S.; Norwitz, E.R.; Guller, S.; Abrahams, V.M. Uric Acid Induces Trophoblast IL-1β Production via the Inflammasome: Implications for the Pathogenesis of Preeclampsia. Am. J. Reprod. Immunol. 2011, 65, 542–548. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Mulla, M.J.; Salmon, J.E.; Chamley, L.W.; Brosens, J.J.; Boeras, C.M.; Kavathas, P.B.; Abrahams, V.M. A Role for Uric Acid and the Nalp3 Inflammasome in Antiphospholipid Antibody-Induced IL-1β Production by Human First Trimester Trophoblast. PLoS ONE 2013, 8, e65237. [Google Scholar] [CrossRef]
  138. Gomez-Lopez, N.; Romero, R.; Xu, Y.; Garcia-Flores, V.; Leng, Y.; Panaitescu, B.; Miller, D.; Abrahams, V.M.; Hassan, S.S. Inflammasome Assembly in the Chorioamniotic Membranes during Spontaneous Labor at Term. Am. J. Reprod. Immunol. 2017, 77, e12648. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Gomez-Lopez, N.; Romero, R.; Xu, Y.; Plazyo, O.; Unkel, R.; Leng, Y.; Than, N.G.; Chaiworapongsa, T.; Panaitescu, B.; Dong, Z.; et al. A Role for the Inflammasome in Spontaneous Preterm Labor With Acute Histologic Chorioamnionitis. Reprod. Sci. 2017, 24, 1382–1401. [Google Scholar] [CrossRef] [PubMed]
  140. Gotsch, F.; Romero, R.; Chaiworapongsa, T.; Erez, O.; Vaisbuch, E.; Espinoza, J.; Kusanovic, J.P.; Mittal, P.; Mazaki-Tovi, S.; Kim, C.J.; et al. Evidence of the Involvement of Caspase-1 under Physiologic and Pathologic Cellular Stress during Human Pregnancy: A Link between the Inflammasome and Parturition. J. Matern. Fetal. Neonatal Med. 2008, 21, 605–616. [Google Scholar] [CrossRef] [Green Version]
  141. Gomez-Lopez, N.; Romero, R.; Tarca, A.L.; Miller, D.; Panaitescu, B.; Schwenkel, G.; Gudicha, D.W.; Hassan, S.S.; Pacora, P.; Jung, E.; 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. [Google Scholar] [CrossRef]
  142. Gomez-Lopez, N.; Romero, R.; Garcia-Flores, V.; Leng, Y.; Miller, D.; Hassan, S.S.; Hsu, C.-D.; Panaitescu, B. Inhibition of the NLRP3 Inflammasome Can Prevent Sterile Intra-Amniotic Inflammation, Preterm Labor/Birth, and Adverse Neonatal Outcomes. Biol. Reprod. 2019, 100, 1306–1318. [Google Scholar] [CrossRef] [PubMed]
  143. Motomura, K.; Romero, R.; Garcia-Flores, V.; Leng, Y.; Xu, Y.; Galaz, J.; Slutsky, R.; Levenson, D.; Gomez-Lopez, N. The Alarmin Interleukin-1α Causes Preterm Birth through the NLRP3 Inflammasome. Mol. Hum. Reprod. 2020, 26, 712–726. [Google Scholar] [CrossRef] [PubMed]
  144. Faro, J.; Romero, R.; Schwenkel, G.; Garcia-Flores, V.; Arenas-Hernandez, M.; Leng, Y.; Xu, Y.; Miller, D.; Hassan, S.S.; Gomez-Lopez, N. Intra-Amniotic Inflammation Induces Preterm Birth by Activating the NLRP3 Inflammasome. Biol. Reprod. 2019, 100, 1290–1305. [Google Scholar] [CrossRef]
  145. Zhu, L.; Wang, Y.; Zhu, Y.; Zhang, W.; Zhu, J. Expression of NOD1 and Downstream Factors in Placenta, Fetal Membrane and Plasma from Pregnancies with Premature Rupture of Membranes and Their Significance. Int. J. Clin. Exp. Pathol. 2018, 11, 5745–5754. [Google Scholar]
  146. Theis, K.R.; Romero, R.; Motomura, K.; Galaz, J.; Winters, A.D.; Pacora, P.; Miller, D.; Slutsky, R.; Florova, V.; Levenson, D.; et al. Microbial Burden and Inflammasome Activation in Amniotic Fluid of Patients with Preterm Prelabor Rupture of Membranes. J. Perinat. Med. 2020, 48, 115–131. [Google Scholar] [CrossRef] [Green Version]
  147. Zhu, J.; Ma, C.; Luan, X.; Li, J.; Peng, F.; Huang, L. Inflammasome Components and ADAMTS4 in Premature Rupture of Membranes. Mol. Med. Rep. 2021, 23, 101. [Google Scholar] [CrossRef] [PubMed]
  148. Zhou, Y.; Shah, S.Z.A.; Yang, L.; Zhang, Z.; Zhou, X.; Zhao, D. Virulent Mycobacterium Bovis Beijing Strain Activates the NLRP7 Inflammasome in THP-1 Macrophages. PLoS ONE 2016, 11, e0152853. [Google Scholar] [CrossRef] [Green Version]
  149. Bednash, J.S.; Weathington, N.; Londino, J.; Rojas, M.; Gulick, D.L.; Fort, R.; Han, S.; McKelvey, A.C.; Chen, B.B.; Mallampalli, R.K. Targeting the Deubiquitinase STAMBP Inhibits NALP7 Inflammasome Activity. Nat. Commun. 2017, 8, 15203. [Google Scholar] [CrossRef] [Green Version]
  150. Abi Nahed, R.; Reynaud, D.; Borg, A.J.; Traboulsi, W.; Wetzel, A.; Sapin, V.; Brouillet, S.; Dieudonné, M.N.; Dakouane-Giudicelli, M.; Benharouga, M.; et al. NLRP7 Is Increased in Human Idiopathic Fetal Growth Restriction and Plays a Critical Role in Trophoblast Differentiation. J. Mol. Med. 2019, 97, 355–367. [Google Scholar] [CrossRef] [PubMed]
  151. Wang, R.-Y.; Li, Y.-J.; Zhen, L.; Jiang, F.; Gu, C.-M.; Li, D.-Z. Detection of Parental Contribution to Molar Genome Leads to Diagnosis of Recurrent Hydatidiform Mole in a Family with NLRP7 Variants. Fetal Pediatr Pathol 2020. [Google Scholar] [CrossRef]
  152. Rezaei, M.; Suresh, B.; Bereke, E.; Hadipour, Z.; Aguinaga, M.; Qian, J.; Bagga, R.; Fardaei, M.; Hemida, R.; Jagadeesh, S.; et al. Novel Pathogenic Variants in NLRP7, NLRP5, and PADI6 in Patients with Recurrent Hydatidiform Moles and Reproductive Failure. Clin. Genet. 2021, 99, 823–828. [Google Scholar] [CrossRef]
  153. Qian, J.; Nguyen, N.M.P.; Rezaei, M.; Huang, B.; Tao, Y.; Zhang, X.; Cheng, Q.; Yang, H.; Asangla, A.; Majewski, J.; et al. Biallelic PADI6 Variants Linking Infertility, Miscarriages, and Hydatidiform Moles. Eur. J. Hum. Genet. 2018, 26, 1007–1013. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Zhang, P.; Zhu, X.; Yu, X.; Huang, B.; Jiang, T.; Zhang, X.; Yang, H.; Qian, J. Abnormal Processing of IL-1β in NLRP7-Mutated Monocytes in Hydatidiform Mole Patients. Clin. Exp. Immunol. 2020, 202, 72–79. [Google Scholar] [CrossRef]
  155. Fallahi, J.; Razban, V.; Momtahan, M.; Akbarzadeh-Jahromi, M.; Namavar-Jahromi, B.; Anvar, Z.; Fardaei, M. A Novel Mutation in NLRP7 Related to Recurrent Hydatidiform Mole and Reproductive Failure. Int. J. Fertil Steril 2019, 13, 135–138. [Google Scholar] [CrossRef] [PubMed]
  156. Tsai, P.-Y.; Chen, K.-R.; Li, Y.-C.; Kuo, P.-L. NLRP7 Is Involved in the Differentiation of the Decidual Macrophages. Int. J. Mol. Sci. 2019, 20, 5994. [Google Scholar] [CrossRef] [Green Version]
  157. Lavergne, M.; Belville, C.; Choltus, H.; Gross, C.; Minet-Quinard, R.; Gallot, D.; Sapin, V.; Blanchon, L. Human Amnion Epithelial Cells (AECs) Respond to the FSL-1 Lipopeptide by Engaging the NLRP7 Inflammasome. Front. Immunol. 2020, 11, 1645. [Google Scholar] [CrossRef] [PubMed]
  158. Khare, S.; Dorfleutner, A.; Bryan, N.B.; Yun, C.; Radian, A.D.; de Almeida, L.; Rojanasakul, Y.; Stehlik, C. An NLRP7-Containing Inflammasome Mediates Recognition of Microbial Lipopeptides in Human Macrophages. Immunity 2012, 36, 464–476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Shibata, K.; Hasebe, A.; Into, T.; Yamada, M.; Watanabe, T. The N-Terminal Lipopeptide of a 44-KDa Membrane-Bound Lipoprotein of Mycoplasma Salivarium Is Responsible for the Expression of Intercellular Adhesion Molecule-1 on the Cell Surface of Normal Human Gingival Fibroblasts. J. Immunol. 2000, 165, 6538–6544. [Google Scholar] [CrossRef] [Green Version]
  160. Mühlradt, P.F.; Kiess, M.; Meyer, H.; Süssmuth, R.; Jung, G. Isolation, Structure Elucidation, and Synthesis of a Macrophage Stimulatory Lipopeptide from Mycoplasma Fermentans Acting at Picomolar Concentration. J. Exp. Med. 1997, 185, 1951–1958. [Google Scholar] [CrossRef] [Green Version]
  161. Lappas, M. A20, an Essential Component of the Ubiquitin-Editing Protein Complex, Is a Negative Regulator of Inflammation in Human Myometrium and Foetal Membranes. Mol. Hum. Reprod. 2017, 23, 628–645. [Google Scholar] [CrossRef]
  162. Lim, R.; Barker, G.; Liong, S.; Nguyen-Ngo, C.; Tong, S.; Kaitu’u-Lino, T.; Lappas, M. ATF3 Is a Negative Regulator of Inflammation in Human Fetal Membranes. Placenta 2016, 47, 63–72. [Google Scholar] [CrossRef] [PubMed]
  163. Lim, R.; Barker, G.; Lappas, M. Pellino 1 Is a Novel Regulator of TNF and TLR Signalling in Human Myometrial and Amnion Cells. J. Reprod. Immunol. 2018, 127, 24–35. [Google Scholar] [CrossRef]
  164. Goradia, P.; Lim, R.; Lappas, M. DREAM Is Involved in the Genesis of Inflammation-Induced Prolabour Mediators in Human Myometrial and Amnion Cells. Biomed. Res. Int. 2018, 2018, 8237087. [Google Scholar] [CrossRef] [Green Version]
  165. Lim, R.; Barker, G.; Lappas, M. PARK7 Regulates Inflammation-Induced pro-Labour Mediators in Myometrial and Amnion Cells. Reproduction 2018, 155, 207–218. [Google Scholar] [CrossRef] [PubMed]
  166. Lim, R.; Barker, G.; Lappas, M. The TLR2 Ligand FSL-1 and the TLR5 Ligand Flagellin Mediate pro-Inflammatory and pro-Labour Response via MyD88/TRAF6/NF-ΚB-Dependent Signalling. Am. J. Reprod. Immunol. 2014, 71, 401–417. [Google Scholar] [CrossRef]
  167. Ariza, M.-E.; Glaser, R.; Kaumaya, P.T.P.; Jones, C.; Williams, M.V. The EBV-Encoded DUTPase Activates NF-Kappa B through the TLR2 and MyD88-Dependent Signaling Pathway. J. Immunol. 2009, 182, 851–859. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Lim, R.; Tran, H.T.; Liong, S.; Barker, G.; Lappas, M. The Transcription Factor Interferon Regulatory Factor-1 (IRF1) Plays a Key Role in the Terminal Effector Pathways of Human Preterm Labor. Biol. Reprod. 2016, 94, 32. [Google Scholar] [CrossRef] [PubMed]
  169. Lim, R.; Lappas, M. A Novel Role for GSK3 in the Regulation of the Processes of Human Labour. Reproduction 2015, 149, 189–202. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Lappas, M. Effect of Spontaneous Term Labour on the Expression of the NR4A Receptors Nuclear Receptor Related 1 Protein (Nurr1), Neuron-Derived Clone 77 (Nur77) and Neuron-Derived Orphan Receptor 1 (NOR1) in Human Fetal Membranes and Myometrium. Reprod. Fertil Dev. 2016, 28, 893–906. [Google Scholar] [CrossRef] [PubMed]
  171. Romero, R.; Xu, Y.; Plazyo, O.; Chaemsaithong, P.; Chaiworapongsa, T.; Unkel, R.; Than, N.G.; Chiang, P.J.; Dong, Z.; Xu, Z.; et al. A Role for the Inflammasome in Spontaneous Labor at Term. Am. J. Reprod. Immunol. 2018, 79, e12440. [Google Scholar] [CrossRef]
  172. Kacerovsky, M.; Pliskova, L.; Bolehovska, R.; Musilova, I.; Hornychova, H.; Tambor, V.; Jacobsson, B. The Microbial Load with Genital Mycoplasmas Correlates with the Degree of Histologic Chorioamnionitis in Preterm PROM. Am. J. Obstet. Gynecol. 2011, 205, 213.E1–213.E7. [Google Scholar] [CrossRef] [PubMed]
  173. Kacerovský, M.; Pavlovský, M.; Tosner, J. Preterm Premature Rupture of the Membranes and Genital Mycoplasmas. Acta Med. 2009, 52, 117–120. [Google Scholar] [CrossRef] [Green Version]
  174. Paramel Jayaprakash, T.; Wagner, E.C.; van Schalkwyk, J.; Albert, A.Y.K.; Hill, J.E.; Money, D.M.; PPROM Study Group. High Diversity and Variability in the Vaginal Microbiome in Women Following Preterm Premature Rupture of Membranes (PPROM): A Prospective Cohort Study. PLoS ONE 2016, 11, e0166794. [Google Scholar] [CrossRef]
  175. Musilova, I.; Pliskova, L.; Kutova, R.; Hornychova, H.; Jacobsson, B.; Kacerovsky, M. Ureaplasma Species and Mycoplasma Hominis in Cervical Fluid of Pregnancies Complicated by Preterm Prelabor Rupture of Membranes. J. Matern. Fetal Neonatal Med. 2016, 29, 1–7. [Google Scholar] [CrossRef] [PubMed]
  176. Latino, M.A.; Botta, G.; Badino, C.; Maria, D.D.; Petrozziello, A.; Sensini, A.; Leli, C. Association between Genital Mycoplasmas, Acute Chorioamnionitis and Fetal Pneumonia in Spontaneous Abortions. J. Perinat. Med. 2018, 46, 503–508. [Google Scholar] [CrossRef] [PubMed]
  177. Kacerovský, M.; Boudys, L. Preterm premature rupture of membranes and Ureaplasma urealyticum. Ceska Gynekol 2008, 73, 154–159. [Google Scholar]
  178. Kwak, D.-W.; Hwang, H.-S.; Kwon, J.-Y.; Park, Y.-W.; Kim, Y.-H. Co-Infection with Vaginal Ureaplasma Urealyticum and Mycoplasma Hominis Increases Adverse Pregnancy Outcomes in Patients with Preterm Labor or Preterm Premature Rupture of Membranes. J. Matern. Fetal Neonatal Med. 2014, 27, 333–337. [Google Scholar] [CrossRef]
  179. Cox, C.; Saxena, N.; Watt, A.P.; Gannon, C.; McKenna, J.P.; Fairley, D.J.; Sweet, D.; Shields, M.D.; Cosby, S.L.; Coyle, P.V. The Common Vaginal Commensal Bacterium Ureaplasma Parvum Is Associated with Chorioamnionitis in Extreme Preterm Labor. J. Matern. Fetal Neonatal Med. 2016, 29, 3646–3651. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Physiological functions of the FM barrier. FMs are a nine-month organ required for the progress of the pregnancy. Indeed, they exercise different essential functions. First, they constitute an adjustable tank resisting internal and external pressure where the fetus can move and grow. FM also allows the amniotic fluid (AF) to achieve homeostasis by letting ions, water and other compounds pass, notably through aquaporins. On the other hand, they physically and chemically limit the ascension of microbial agents. Finally, they are implicated in parturition by their planned and controlled rupture at term. SMART Servier Medical Art device was used from the creation of the figure.
Figure 1. Physiological functions of the FM barrier. FMs are a nine-month organ required for the progress of the pregnancy. Indeed, they exercise different essential functions. First, they constitute an adjustable tank resisting internal and external pressure where the fetus can move and grow. FM also allows the amniotic fluid (AF) to achieve homeostasis by letting ions, water and other compounds pass, notably through aquaporins. On the other hand, they physically and chemically limit the ascension of microbial agents. Finally, they are implicated in parturition by their planned and controlled rupture at term. SMART Servier Medical Art device was used from the creation of the figure.
Biomedicines 09 01123 g001
Figure 2. Two types of inflammation. This figure distinguishes the two types of inflammation: (i) the septic one implies the recognition of PAMPs/MAMPs derived from micro-organisms (viruses, bacteria or fungi); (ii) the sterile one is triggered by the recognition of DAMPs by the members of both PRRs (cytosolic or membrane-addressed). Both the recognition of PAMPs/MAMPs and DAMPs lead to a pro-inflammatory response.
Figure 2. Two types of inflammation. This figure distinguishes the two types of inflammation: (i) the septic one implies the recognition of PAMPs/MAMPs derived from micro-organisms (viruses, bacteria or fungi); (ii) the sterile one is triggered by the recognition of DAMPs by the members of both PRRs (cytosolic or membrane-addressed). Both the recognition of PAMPs/MAMPs and DAMPs lead to a pro-inflammatory response.
Biomedicines 09 01123 g002
Figure 3. RAGE and its isoforms. This figure illustrates the different protein isoforms of RAGE. Only the full-length (fl-RAGE) version can exercise a complete signal transduction by first interacting with ligands with its extracellular domain (V: variable; C: constant) similar to immunoglobulins and then recruiting protein adaptors using its intracellular domain. Other isoforms, especially soluble ones (esRAGE, directly translated from a transcript and sRAGE, resulting from an enzymatic cleavage of fl-RAGE), are implicated in the negative feedback of RAGE activity by competition. Finally, Nt-RAGE and DN-RAGE have also been briefly described in the literature.
Figure 3. RAGE and its isoforms. This figure illustrates the different protein isoforms of RAGE. Only the full-length (fl-RAGE) version can exercise a complete signal transduction by first interacting with ligands with its extracellular domain (V: variable; C: constant) similar to immunoglobulins and then recruiting protein adaptors using its intracellular domain. Other isoforms, especially soluble ones (esRAGE, directly translated from a transcript and sRAGE, resulting from an enzymatic cleavage of fl-RAGE), are implicated in the negative feedback of RAGE activity by competition. Finally, Nt-RAGE and DN-RAGE have also been briefly described in the literature.
Biomedicines 09 01123 g003
Figure 4. RAGE signaling pathway. Following the binding of its ligands, RAGE can induce different proinflammatory pathways, implicating some adaptor intracellular proteins (TIRAP, MyD88, Dia-1). This activation results in the induction of proinflammatory gene transcription (TNFα, cyclooxygenase 2, IL1β, IL6, IL8), thus maintaining the inflammation phenomenon.
Figure 4. RAGE signaling pathway. Following the binding of its ligands, RAGE can induce different proinflammatory pathways, implicating some adaptor intracellular proteins (TIRAP, MyD88, Dia-1). This activation results in the induction of proinflammatory gene transcription (TNFα, cyclooxygenase 2, IL1β, IL6, IL8), thus maintaining the inflammation phenomenon.
Biomedicines 09 01123 g004
Figure 5. NLRP inflammasome structure. A NLRP inflammasome is composed of three major elements. First, there is a sensor, the receptor NLRP, which itself is generally composed of different domains: PYD (Pyrin domain), NACHT (acronym for NAIP, CIITA, HET-E, TP1), NAD (NACHT-associated domain) and LRR (Leucine-rich repeats). This LRR domain allows for an interaction with the adaptor ASC by the PYD domains. Then, ASC can recruit pro-caspase 1 thanks to the CARD (caspase recruitment domain) of each actor. In addition, pro-caspase 1 also contains a central p20 domain (including the catalytic site) and a p10 domain.
Figure 5. NLRP inflammasome structure. A NLRP inflammasome is composed of three major elements. First, there is a sensor, the receptor NLRP, which itself is generally composed of different domains: PYD (Pyrin domain), NACHT (acronym for NAIP, CIITA, HET-E, TP1), NAD (NACHT-associated domain) and LRR (Leucine-rich repeats). This LRR domain allows for an interaction with the adaptor ASC by the PYD domains. Then, ASC can recruit pro-caspase 1 thanks to the CARD (caspase recruitment domain) of each actor. In addition, pro-caspase 1 also contains a central p20 domain (including the catalytic site) and a p10 domain.
Biomedicines 09 01123 g005
Figure 6. NLRP inflammasome activation. Following the priming step (transcriptional and/or not), the activated NLRP can form the inflammasome with ASC and pro-caspase 1. This functional NLRP inflammasome leads to the autolytic cleavage of pro-caspase 1 into caspase-1, which is the active enzyme. Downstream, caspase-1 activates the interleukins IL18 and IL1β but also releases the N-terminal fragment of the gasdermin D from the inhibitory C-terminal fragment. Finally, a formation of pores by the N-terminal gasdermin D at the plasma membrane induces pyroptosis and propagation of the inflammatory signal [117,124,126].
Figure 6. NLRP inflammasome activation. Following the priming step (transcriptional and/or not), the activated NLRP can form the inflammasome with ASC and pro-caspase 1. This functional NLRP inflammasome leads to the autolytic cleavage of pro-caspase 1 into caspase-1, which is the active enzyme. Downstream, caspase-1 activates the interleukins IL18 and IL1β but also releases the N-terminal fragment of the gasdermin D from the inhibitory C-terminal fragment. Finally, a formation of pores by the N-terminal gasdermin D at the plasma membrane induces pyroptosis and propagation of the inflammatory signal [117,124,126].
Biomedicines 09 01123 g006
Table 1. Expression of the NRLP family in human FM. The mRNA expression of the 14 NLRPs, ASC and caspase-1 was investigated by RT-qPCR in terms of human amnion and chorion. “+” indicates the presence of the transcripts, and “Ø” indicates the absence of RNA expression.
Table 1. Expression of the NRLP family in human FM. The mRNA expression of the 14 NLRPs, ASC and caspase-1 was investigated by RT-qPCR in terms of human amnion and chorion. “+” indicates the presence of the transcripts, and “Ø” indicates the absence of RNA expression.
NLRPASCCaspase-1
1234567891011121214
Term Amnion+++ØØ++Ø++ØØØ+++
Term Chorion+++ØØ++Ø++Ø+Ø+++
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Choltus, H.; Lavergne, M.; De Sousa Do Outeiro, C.; Coste, K.; Belville, C.; Blanchon, L.; Sapin, V. Pathophysiological Implication of Pattern Recognition Receptors in Fetal Membranes Rupture: RAGE and NLRP Inflammasome. Biomedicines 2021, 9, 1123. https://doi.org/10.3390/biomedicines9091123

AMA Style

Choltus H, Lavergne M, De Sousa Do Outeiro C, Coste K, Belville C, Blanchon L, Sapin V. Pathophysiological Implication of Pattern Recognition Receptors in Fetal Membranes Rupture: RAGE and NLRP Inflammasome. Biomedicines. 2021; 9(9):1123. https://doi.org/10.3390/biomedicines9091123

Chicago/Turabian Style

Choltus, Helena, Marilyne Lavergne, Coraline De Sousa Do Outeiro, Karen Coste, Corinne Belville, Loïc Blanchon, and Vincent Sapin. 2021. "Pathophysiological Implication of Pattern Recognition Receptors in Fetal Membranes Rupture: RAGE and NLRP Inflammasome" Biomedicines 9, no. 9: 1123. https://doi.org/10.3390/biomedicines9091123

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