Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.446
Filter
Add more filters

Publication year range
1.
EMBO J ; 41(7): e108747, 2022 04 04.
Article in English | MEDLINE | ID: mdl-35266581

ABSTRACT

Mesoderm arises at gastrulation and contributes to both the mouse embryo proper and its extra-embryonic membranes. Two-photon live imaging of embryos bearing a keratin reporter allowed recording filament nucleation and elongation in the extra-embryonic region. Upon separation of amniotic and exocoelomic cavities, keratin 8 formed apical cables co-aligned across multiple cells in the amnion, allantois, and blood islands. An influence of substrate rigidity and composition on cell behavior and keratin content was observed in mesoderm explants. Embryos lacking all keratin filaments displayed a deflated extra-embryonic cavity, a narrow thick amnion, and a short allantois. Single-cell RNA sequencing of sorted mesoderm cells and micro-dissected amnion, chorion, and allantois, provided an atlas of transcriptomes with germ layer and regional information. It defined the cytoskeleton and adhesion expression profile of mesoderm-derived keratin 8-enriched cells lining the exocoelomic cavity. Those findings indicate a novel role for keratin filaments in the expansion of extra-embryonic structures and suggest mechanisms of mesoderm adaptation to the environment.


Subject(s)
Gastrulation , Mesoderm , Animals , Embryo, Mammalian , Extraembryonic Membranes , Keratins/genetics , Keratins/metabolism , Mesoderm/metabolism , Mice
2.
Immunol Rev ; 308(1): 149-167, 2022 07.
Article in English | MEDLINE | ID: mdl-35285967

ABSTRACT

Human parturition at term and preterm is an inflammatory process synchronously executed by both fetomaternal tissues to transition them from a quiescent state t an active state of labor to ensure delivery. The initiators of the inflammatory signaling mechanism can be both maternal and fetal. The placental (fetal)-maternal immune and endocrine mediated homeostatic imbalances and inflammation are well reported. However, the fetal inflammatory response (FIR) theories initiated by the fetal membranes (amniochorion) at the choriodecidual interface are not well established. Although immune cell migration, activation, and production of proparturition cytokines to the fetal membranes are reported, cellular level events that can generate a unique set of inflammation are not well discussed. This review discusses derangements to fetal membrane cells (physiologically and pathologically at term and preterm, respectively) in response to both endogenous and exogenous factors to generate inflammatory signals. In addition, the mechanisms of inflammatory signal propagation (fetal signaling of parturition) and how these signals cause immune imbalances at the choriodecidual interface are discussed. In addition to maternal inflammation, this review projects FIR as an additional mediator of inflammatory overload required to promote parturition.


Subject(s)
Labor, Obstetric , Placenta , Extraembryonic Membranes/metabolism , Female , Humans , Infant, Newborn , Inflammation/metabolism , Labor, Obstetric/metabolism , Parturition/metabolism , Placenta/metabolism , Pregnancy
3.
PLoS Pathog ; 19(8): e1011274, 2023 08.
Article in English | MEDLINE | ID: mdl-37549143

ABSTRACT

Zika virus (ZIKV) can be transmitted vertically from mother to fetus during pregnancy, resulting in a range of outcomes including severe birth defects and fetal/infant death. Potential pathways of vertical transmission in utero have been proposed but remain undefined. Identifying the timing and routes of vertical transmission of ZIKV may help us identify when interventions would be most effective. Furthermore, understanding what barriers ZIKV overcomes to effect vertical transmission may help improve models for evaluating infection by other pathogens during pregnancy. To determine the pathways of vertical transmission, we inoculated 12 pregnant rhesus macaques with an African-lineage ZIKV at gestational day 30 (term is 165 days). Eight pregnancies were surgically terminated at either seven or 14 days post-maternal infection. Maternal-fetal interface and fetal tissues and fluids were collected and evaluated for ZIKV using RT-qPCR, in situ hybridization, immunohistochemistry, and plaque assays. Four additional pregnant macaques were inoculated and terminally perfused with 4% paraformaldehyde at three, six, nine, or ten days post-maternal inoculation. For these four cases, the entire fixed pregnant uterus was evaluated with in situ hybridization for ZIKV RNA. We determined that ZIKV can reach the MFI by six days after infection and infect the fetus by ten days. Infection of the chorionic membrane and the extraembryonic coelomic fluid preceded infection of the fetus and the mesenchymal tissue of the placental villi. We did not find evidence to support a transplacental route of ZIKV vertical transmission via infection of syncytiotrophoblasts or villous cytotrophoblasts. The pattern of infection observed in the maternal-fetal interface provides evidence of paraplacental vertical ZIKV transmission through the chorionic membrane, the outer layer of the fetal membranes.


Subject(s)
Pregnancy Complications, Infectious , Zika Virus Infection , Zika Virus , Humans , Animals , Pregnancy , Female , Zika Virus/genetics , Macaca mulatta , Placenta , Pregnancy Complications, Infectious/metabolism , Fetal Death , Infectious Disease Transmission, Vertical , Extraembryonic Membranes/metabolism
4.
Am J Pathol ; 194(5): 684-692, 2024 05.
Article in English | MEDLINE | ID: mdl-38320630

ABSTRACT

Preterm, prelabor rupture of the human fetal membranes (pPROM) is involved in 40% of spontaneous preterm births worldwide. Cellular-level disturbances and inflammation are effectors of membrane degradation, weakening, and rupture. Maternal risk factors induce oxidative stress (OS), senescence, and senescence-associated inflammation of the fetal membranes as reported mechanisms related to pPROM. Inflammation can also arise in fetal membrane cells (amnion/chorion) due to OS-induced autophagy and epithelial-mesenchymal transition (EMT). Autophagy, EMT, and their correlation in pPROM, along with OS-induced autophagy-related changes in amnion and chorion cells in vitro, were investigated. Immunocytochemistry staining of cytokeratin-18 (epithelial marker)/vimentin (mesenchymal marker) and proautophagy-inducing factor LC3B were performed in fetal membranes from pPROM, term not in labor, and term labor. Ultrastructural changes associated with autophagy were verified by transmission electron microscopy of the fetal membranes and in cells exposed to cigarette smoke extract (an OS inducer). EMT and LC3B staining was compared in the chorion from pPROM versus term not in labor. Transmission electron microscopy confirmed autophagosome formation in pPROM amnion and chorion. In cell culture, autophagosomes were formed in the amnion with OS treatment, while autophagosomes were accumulated in both cell types with autophagy inhibition. This study documents the association between pPROMs and amniochorion autophagy and EMT, and supports a role for OS in inducing dysfunctional cells that increase inflammation, predisposing membranes to rupture.


Subject(s)
Extraembryonic Membranes , Fetal Membranes, Premature Rupture , Female , Infant, Newborn , Humans , Extraembryonic Membranes/metabolism , Fetal Membranes, Premature Rupture/metabolism , Inflammation/pathology , Epithelial-Mesenchymal Transition , Autophagy
5.
J Pathol ; 262(2): 240-253, 2024 02.
Article in English | MEDLINE | ID: mdl-38018407

ABSTRACT

Preterm labor/birth is the leading cause of perinatal mortality and morbidity worldwide. Previous studies demonstrated that T cells were crucial for maintaining maternal-fetal immune tolerance during the first trimester of pregnancy; however, their phenotypes and functions in labor and delivery remain largely unknown. We recruited three cohorts of women at delivery for T-cell immunophenotyping in the placentas, fetal membranes, umbilical cord blood, and maternal peripheral blood. Our data showed a differential enrichment of T cells during the third trimester of human pregnancy, with CD4+ T cells being more observable within the umbilical cord blood, whereas CD8+ T cells became relatively more abundant in fetal membranes. CD4+ and CD8+ T cells derived from fetal membranes were dominated by effector memory T cells and exhibited extensive expression of activation markers but decreased expression of homing receptor. In comparison with term births, fetal membrane CD8+ T cells, especially the central memory subset, were significantly increased in frequency and showed more profound activation in spontaneous preterm birth patients. Finally, using an allogeneic mouse model, we found that T-cell-activation-induced preterm birth could be alleviated by the depletion of CD8+ T but not CD4+ T cells in vivo. Collectively, we showed that CD8+ T cells in fetal membranes displayed a unique phenotype, and their activation was involved in the pathophysiology of spontaneous preterm birth, which provides novel insights into the immune mechanisms of preterm birth and potential targets for the prevention of this syndrome. © 2023 The Pathological Society of Great Britain and Ireland.


Subject(s)
Obstetric Labor, Premature , Premature Birth , Pregnancy , Animals , Mice , Humans , Female , Infant, Newborn , Premature Birth/chemically induced , Premature Birth/prevention & control , CD8-Positive T-Lymphocytes , Extraembryonic Membranes , Phenotype
6.
Immunology ; 172(4): 577-587, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38631842

ABSTRACT

Preterm birth is the largest contributor to neonatal morbidity and is often associated with chorioamnionitis, defined as inflammation/infection of the fetal membranes (FMs). Chorioamnionitis is characterised by neutrophil infiltration of the FMs and is associated with elevated levels of the neutrophil chemoattractant, interleukin (IL)-8 and the proinflammatory cytokine, IL-1ß. While FMs can respond to infections through innate immune sensors, such as toll-like receptors (TLRs), the downstream mechanisms by which chorioamnionitis arises are not fully understood. A novel group of non-classical microRNAs (miR-21a, miR-29a, miR-146a-3p, Let-7b) function as endogenous danger signals by activating the ssRNA viral sensors TLR7 and TLR8. In this study, the pro-inflammatory roles of TLR7/TLR8-activating miRs were examined as mediators of FM inflammation in response to bacterial lipopolysaccharide (LPS) using an in vitro human FM explant system, an in vivo mouse model of pregnancy, and human clinical samples. Following LPS exposure, miR-146a-3p was significantly increased in both human FM explants and wild-type mouse FMs. Expression of miR-146a-3p was also significantly elevated in FMs from women with preterm birth and chorioamnionitis. FM IL-8 and inflammasome-mediated IL-1ß production in response to LPS was dependent on miR-146a-3p and TLR8 downstream of TLR4 activation. In wild-type mice, LPS exposure increased FM IL-8 and IL-1ß production and induced preterm birth. In TLR7-/-/TLR8-/- mice, LPS exposure was able to initiate but not sustain preterm birth, and FM inflammation was reduced. Together, we demonstrate a novel signalling mechanism at the maternal-fetal interface in which TLR8-activating miR-146a-3p acts as an intermediate danger signal to drive FM inflammasome-dependent and -independent mechanisms of inflammation and, thus, may play a role in chorioamnionitis and subsequent preterm birth.


Subject(s)
Chorioamnionitis , Extraembryonic Membranes , Lipopolysaccharides , MicroRNAs , Toll-Like Receptor 8 , Animals , Female , Humans , Mice , Pregnancy , Chorioamnionitis/immunology , Chorioamnionitis/metabolism , Extraembryonic Membranes/metabolism , Extraembryonic Membranes/immunology , Inflammation/immunology , Inflammation/metabolism , Interleukin-1beta/metabolism , Interleukin-8/metabolism , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , MicroRNAs/metabolism , Premature Birth/immunology , Signal Transduction , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 7/genetics , Toll-Like Receptor 8/metabolism , Toll-Like Receptor 8/genetics
7.
Biol Reprod ; 110(5): 971-984, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38335245

ABSTRACT

Intrauterine infection is a significant cause of neonatal morbidity and mortality. Ureaplasma parvum is a microorganism commonly isolated from cases of preterm birth and preterm premature rupture of membranes (pPROM). However, the mechanisms of early stage ascending reproductive tract infection remain poorly understood. To examine inflammation in fetal (chorioamnionic) membranes we utilized a non-human primate (NHP) model of choriodecidual U. parvum infection. Eight chronically catheterized pregnant rhesus macaques underwent maternal-fetal catheterization surgery at ~105-112 days gestation and choriodecidual inoculation with U. parvum (105 CFU/mL, n =4) or sterile media (controls; n = 4) starting at 115-119 days, repeated at 5-day intervals until C-section at 136-140 days (term=167 days). The average inoculation to delivery interval was 21 days, and Ureaplasma infection of the amniotic fluid (AF) was undetectable in all animals. Choriodecidual Ureaplasma infection resulted in increased fetal membrane expression of MMP-9 and PTGS2, but did not result in preterm labor or increased concentrations of AF pro-inflammatory cytokines. However, membrane expression of inflammasome sensors, NLRP3, NLRC4, AIM2, and NOD2, and adaptor ASC (PYCARD) gene expression were significantly increased. Gene expression of IL-1ß, IL-18, IL-18R1  , CASPASE-1, and pro-CASPASE-1 protein increased with Ureaplasma infection. Downstream inflammatory genes MYD88 and NFκB (Nuclear factor kappa-light-chain-enhancer of activated B cells) were also significantly upregulated. These results demonstrate that choriodecidual Ureaplasma infection, can cause activation of inflammasome complexes and pathways associated with pPROM and preterm labor prior to microbes being detectable in the AF.


Subject(s)
Inflammasomes , Macaca mulatta , Ureaplasma Infections , Ureaplasma , Animals , Female , Pregnancy , Inflammasomes/metabolism , Disease Models, Animal , Chorion/metabolism , Extraembryonic Membranes/metabolism , Extraembryonic Membranes/microbiology , Decidua/metabolism , Decidua/microbiology , Pregnancy Complications, Infectious/microbiology
8.
Biomed Microdevices ; 26(3): 32, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38963644

ABSTRACT

Fetal membrane (amniochorion), the innermost lining of the intrauterine cavity, surround the fetus and enclose amniotic fluid. Unlike unidirectional blood flow, amniotic fluid subtly rocks back and forth, and thus, the innermost amnion epithelial cells are continuously exposed to low levels of shear stress from fluid undulation. Here, we tested the impact of fluid motion on amnion epithelial cells (AECs) as a bearer of force impact and their potential vulnerability to cytopathologic changes that can destabilize fetal membrane functions. A previously developed amnion membrane (AM) organ-on-chip (OOC) was utilized but with dynamic flow to culture human fetal amnion membrane cells. The applied flow was modulated to perfuse culture media back and forth for 48 h to mimic fluid motion. A static culture condition was used as a negative control, and oxidative stress (OS) condition was used as a positive control representing pathophysiological changes. The impacts of fluidic motion were evaluated by measuring cell viability, cellular transition, and inflammation. Additionally, scanning electron microscopy (SEM) imaging was performed to observe microvilli formation. The results show that regardless of the applied flow rate, AECs and AMCs maintained their viability, morphology, innate meta-state, and low production of pro-inflammatory cytokines. E-cadherin expression and microvilli formation in the AECs were upregulated in a flow rate-dependent fashion; however, this did not impact cellular morphology or cellular transition or inflammation. OS treatment induced a mesenchymal morphology, significantly higher vimentin to cytokeratin 18 (CK-18) ratio, and pro-inflammatory cytokine production in AECs, whereas AMCs did not respond in any significant manner. Fluid motion and shear stress, if any, did not impact AEC cell function and did not cause inflammation. Thus, when using an amnion membrane OOC model, the inclusion of a dynamic flow environment is not necessary to mimic in utero physiologic cellular conditions of an amnion membrane.


Subject(s)
Amniotic Fluid , Extraembryonic Membranes , Lab-On-A-Chip Devices , Humans , Amniotic Fluid/cytology , Extraembryonic Membranes/cytology , Extraembryonic Membranes/metabolism , Amnion/cytology , Amnion/metabolism , Cell Survival , Epithelial Cells/cytology , Epithelial Cells/metabolism , Motion , Oxidative Stress , Models, Biological , Microphysiological Systems
9.
Ultrasound Obstet Gynecol ; 64(3): 374-380, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38514967

ABSTRACT

OBJECTIVE: To assess the feasibility of using a novel device designed for minimally invasive suturing to anchor fetal membranes to the uterine wall and to close surgical defects after fetoscopy. METHODS: We tested the WestStitch™ suturing device both ex vivo and in vivo. In the ex-vivo studies, 12-Fr trocar defects were created with a fetoscope in five specimens of human uterine tissue with fetal membranes attached. Specimens were examined for integrity of the anchoring stitch. For the in-vivo studies, trocar defects were created in the two uterine horns of three pregnant ewes, each carrying twins at approximately 79-90 days' gestation. One trocar defect in each ewe was repaired using the suture device, and the other was left unrepaired as a control. The repair sites were examined for membrane-anchoring integrity when the defect was created and at delivery. RESULTS: Fetal membranes were anchored successfully to the uterine myometrium using the suture-delivery device in all five experiments performed ex vivo. The in-vivo experiments also revealed successful membrane anchoring compared with controls, both at the time of device deployment and 1-9 weeks after the procedure. CONCLUSIONS: We successfully anchored amniotic membranes to the underlying myometrium using a suturing device, both ex vivo and in vivo. Further studies are needed to evaluate the efficacy of the device and to determine whether it can successfully anchor fetal membranes percutaneously in human patients. © 2024 International Society of Ultrasound in Obstetrics and Gynecology.


Subject(s)
Extraembryonic Membranes , Fetal Membranes, Premature Rupture , Fetoscopy , Suture Techniques , Female , Pregnancy , Fetal Membranes, Premature Rupture/prevention & control , Animals , Humans , Fetoscopy/methods , Sheep , Extraembryonic Membranes/diagnostic imaging , Feasibility Studies
10.
Prenat Diagn ; 44(3): 317-324, 2024 03.
Article in English | MEDLINE | ID: mdl-38168862

ABSTRACT

OBJECTIVE: To compare the biomechanical properties of fetal preterm membranes (20 + 0 weeks to 30 + 0 weeks) to those of the term (37 + 0 to 41 + 0 weeks). METHOD: Amnion and chorion were manually separated and samples were cut to the required geometry. Rectangular samples with (mode 1) and without (uniaxial) a notch, were tested for tearing energy, critical elongation, and tangent stiffness. Suture retention and inter-suture distance testing investigated the effect of suture placement. RESULTS: From the 15 preterm and 10 term placentas studied, no notable differences were observed in uniaxial testing. Mode 1 fracture testing showed a difference in tearing energy between the preterm and term chorion (0.025 ± 0.005 vs. 0.017 ± 0.005 J/m-1 ; p = 0.027) but not in the amnion (0.030 ± 0.017 vs. 0.029 ± 0.009 J/m-1 ; p = 0.895). Both preterm amnion and chorion showed a higher critical elongation compared with term (1.229 ± 0.057 vs. 1.166 ± 0.046; p = 0.019 and 1.307 ± 0.049 vs. 1.218 ± 0.058; p = 0.012). Preterm amnion had a higher suture retention strength than its term counterpart (0.189 ± 0.065 vs. 0.121 ± 0.031 N; p = 0.023). In inter-suture distance tests, no significant interaction was observed beyond 3 mm, but the preterm chorion showed less interaction at 1-2 mm distances. CONCLUSION: Preterm membranes have equivalent or superior tensile properties to term membranes. The chorion appears to contribute to the mechanical integrity of fetal membranes, particularly in preterm stages.


Subject(s)
Amnion , Extraembryonic Membranes , Humans , Pregnancy , Female , Infant, Newborn , Chorion , Placenta
11.
Arch Gynecol Obstet ; 309(5): 1919-1923, 2024 May.
Article in English | MEDLINE | ID: mdl-37184578

ABSTRACT

PURPOSE: The fetal membranes are essential for the maintenance of pregnancy, and their integrity until parturition is critical for both fetal and maternal health. Preterm premature rupture of the membranes (pPROM) is known to be an indicator of preterm birth, but the underlying architectural and mechanical changes that lead to fetal membrane failure are not yet fully understood. The aim of this study was to gain new insights into the anatomy of the fetal membrane and to establish a tissue processing and staining protocol suitable for future prospective cohort studies. METHODS: In this proof of principle study, we collected fetal membranes from women undergoing vaginal delivery or cesarean section. Small membrane sections were then fixed, stained for nucleic acids, actin, and collagen using fluorescent probes, and subsequently imaged in three dimensions using a spinning disk confocal microscope. RESULTS: Four fetal membranes of different types were successfully processed and imaged after establishing a suitable protocol. Cellular and nuclear outlines are clearly visible in all cases, especially in the uppermost membrane layer. Focal membrane (micro) fractures could be identified in several samples. CONCLUSION: The presented method proves to be well suited to determine whether and how the occurrence of membrane (micro) fractures and cellular jamming correlate with the timing of membrane rupture and the mode of delivery. In future measurements, this method could be combined with mechanical probing techniques to compare optical and mechanical sample information.


Subject(s)
Fetal Membranes, Premature Rupture , Premature Birth , Female , Infant, Newborn , Pregnancy , Humans , Cesarean Section , Prospective Studies , Chorion , Extraembryonic Membranes , Microscopy, Confocal
12.
Reprod Domest Anim ; 59(7): e14676, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39021341

ABSTRACT

Studies investigating physiological deviations from normality in newborn calves derived from in vitro fertilization procedures remain important for the understanding of factors that reduce calf survival after birth. The aim of this study was to investigate parameters affecting health and welfare of newborn Flemish calves derived from in vitro embryo production (IVP) in the first hours of life in comparison to in vivo-derived calves. Physical traits of newborn calves and fetal membranes (FM) were recorded soon after birth. Newborn venous blood samples were collected at several time points within the first 24 h of life for analyses of energy substrates, electrolytes, blood gases, acid-base balance, blood chemistry, and haematology. A liver biopsy was taken within the first hour after birth for analysis of gene expression of key enzymes of the fructolytic and glycolytic pathways. Newborn IVP calves were heavier and larger at birth, which was associated with heavier FM. At several time points during the first 24 h of life, IVP-derived calves had altered rectal temperature, blood gases, electrolyte concentrations, blood parameters for liver, kidney and muscle function, and acid-base balance, plasma lipid metabolism, and hemogram parameters. The relative mRNA abundances for triokinase and lactate dehydrogenase-B were greater in IVP calves. In summary, IVP-derived newborn calves were at higher risk of clinical problems after birth, which was markedly greater in heavier and larger calves. Such animals take longer to adapt to extrauterine life and should receive a special attention during the immediate neonatal period.


Subject(s)
Animals, Newborn , Energy Metabolism , Animals , Cattle/physiology , Liver/metabolism , Female , Fertilization in Vitro/veterinary , Extraembryonic Membranes/metabolism , Male , Acid-Base Equilibrium
13.
Int J Mol Sci ; 25(10)2024 May 09.
Article in English | MEDLINE | ID: mdl-38791199

ABSTRACT

Danger-associated molecular patterns (DAMPs) are elevated within the amniotic cavity, and their increases correlate with advancing gestational age, chorioamnionitis, and labor. Although the specific triggers for their release in utero remain unclear, it is thought that they may contribute to the initiation of parturition by influencing cellular stress mechanisms that make the fetal membranes (FMs) more susceptible to rupture. DAMPs induce inflammation in many different tissue types. Indeed, they precipitate the subsequent release of several proinflammatory cytokines that are known to be key for the weakening of FMs. Previously, we have shown that in vitro stretch of human amnion epithelial cells (hAECs) induces a cellular stress response that increases high-mobility group box-1 (HMGB1) secretion. We have also shown that cell-free fetal DNA (cffDNA) induces a cytokine response in FM explants that is fetal sex-specific. Therefore, the aim of this work was to further investigate the link between stretch and the DAMPs HMGB1 and cffDNA in the FM. These data show that stretch increases the level of cffDNA released from hAECs. It also confirms the importance of the sex of the fetus by demonstrating that female cffDNA induced more cellular stress than male fetuses. Our data treating hAECs and human amnion mesenchymal cells with HMGB1 show that it has a differential effect on the ability of the cells of the amnion to upregulate the proinflammatory cytokines and propagate a proinflammatory signal through the FM that may weaken it. Finally, our data show that sulforaphane (SFN), a potent activator of Nrf2, is able to mitigate the proinflammatory effects of stretch by decreasing the levels of HMGB1 release and ROS generation after stretch and modulating the increase of key cytokines after cell stress. HMGB1 and cffDNA are two of the few DAMPs that are known to induce cytokine release and matrix metalloproteinase (MMP) activation in the FMs; thus, these data support the general thesis that they can function as potential central players in the normal mechanisms of FM weakening during the normal distension of this tissue at the end of a normal pregnancy.


Subject(s)
Extraembryonic Membranes , HMGB1 Protein , Inflammation , Humans , HMGB1 Protein/metabolism , HMGB1 Protein/genetics , Female , Pregnancy , Inflammation/metabolism , Inflammation/pathology , Extraembryonic Membranes/metabolism , Cell-Free Nucleic Acids/metabolism , Male , Amnion/metabolism , Cytokines/metabolism , Epithelial Cells/metabolism , Cells, Cultured , Alarmins/metabolism
14.
Biol Reprod ; 109(3): 330-339, 2023 09 12.
Article in English | MEDLINE | ID: mdl-37427976

ABSTRACT

Preterm premature rupture of membranes (pPROM) is a major cause of preterm birth and neonatal mortality. Reactive oxygen species (ROS) have been identified as a critical factor in the development of pPROM. Mitochondria are known to be the primary source of ROS and play a vital role in maintaining cellular function. The Nuclear erythroid 2-related factor 2 (NRF2) has been demonstrated to play a crucial role in regulating mitochondrial function. However, research exploring the impact of NRF2-regulated mitochondria on pPROM is limited. Therefore, we collected fetal membrane tissues from pPROM and spontaneous preterm labor (sPTL) puerpera, measured the expression level of NRF2, and evaluated the degree of mitochondrial damage in both groups. In addition, we isolated human amniotic epithelial cells (hAECs) from the fetal membranes and used small interfering RNA (siRNA) to suppress NRF2 expression, enabling us to evaluate the impact of NRF2 on mitochondrial damage and ROS production. Our findings indicated that the expression level of NRF2 in pPROM fetal membranes was significantly lower than in sPTL fetal membranes, accompanied by increased mitochondrial damage. Furthermore, after the inhibition of NRF2 in hAECs, the degree of mitochondrial damage was significantly exacerbated, along with a marked increase in both cellular and mitochondrial ROS levels. The regulation of the mitochondrial metabolic process via NRF2 in fetal membranes has the potential to influence ROS production.


Subject(s)
Fetal Membranes, Premature Rupture , Premature Birth , Female , Humans , Infant, Newborn , Extraembryonic Membranes/metabolism , Fetal Membranes, Premature Rupture/metabolism , Mitochondria/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Premature Birth/metabolism , Reactive Oxygen Species/metabolism
15.
Inflamm Res ; 72(4): 797-812, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36879064

ABSTRACT

OBJECTIVES: Sterile inflammation of fetal membranes is an indispensable event of normal parturition. However, triggers of sterile inflammation are not fully resolved. Serum amyloid A1 (SAA1) is an acute phase protein produced primarily by the liver. Fetal membranes can also synthesize SAA1 but its functions are not well defined. Given the role of SAA1 in the acute phase response to inflammation, we postulated that SAA1 synthesized in the fetal membranes may be a trigger of local inflammation at parturition. METHODS: The changes of SAA1 abundance in parturition were studied in the amnion of human fetal membranes. The role of SAA1 in chemokine expression and leukocyte chemotaxis was examined in cultured human amnion tissue explants as well as primary human amnion fibroblasts. The effects of SAA1 on monocytes, macrophages and dendritic cells were investigated in cells derived from a human leukemia monocytic cell line (THP-1). RESULTS: SAA1 synthesis increased significantly in human amnion at parturition. SAA1 evoked multiple chemotaxis pathways in human amnion fibroblasts along with upregulation of a series of chemokines via both toll-like receptor 4 (TLR4) and formyl peptide receptor 2 (FPR2). Moreover, SAA1-conditioned medium of cultured amnion fibroblasts was capable of chemoattracting virtually all types of mononuclear leukocytes, particularly monocytes and dendritic cells, which reconciled with the chemotactic activity of conditioned medium of cultured amnion tissue explants collected from spontaneous labor. Furthermore, SAA1 could induce the expression of genes associated with inflammation and extracellular matrix remodeling in monocytes, macrophages and dendritic cells derived from THP-1. CONCLUSIONS: SAA1 is a trigger of sterile inflammation of the fetal membranes at parturition.


Subject(s)
Amnion , Parturition , Pregnancy , Female , Humans , Amnion/metabolism , Culture Media, Conditioned/metabolism , Culture Media, Conditioned/pharmacology , Parturition/genetics , Parturition/metabolism , Extraembryonic Membranes/metabolism , Chemokines/metabolism , Inflammation/metabolism , Serum Amyloid A Protein
16.
J Immunol ; 206(5): 1039-1045, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33472905

ABSTRACT

Preterm birth is associated with significant neonatal mortality and morbidity worldwide. Chorioamnionitis, inflammation of the fetal membranes (FMs), is a major risk factor and is characterized by neutrophil infiltration. However, the role of neutrophils at the FMs remains unclear. We recently reported that FMs exposed to bacterial LPS recruited more neutrophils compared with resting FMs and activated them to degranulate and release reactive oxygen species, chemokines/cytokines, and neutrophil extracellular traps. We posit that under resting conditions, neutrophils play a protective surveillance role, whereas during infection/inflammation, they induce FM tissue injury. To test this, human FM explants were exposed to neutrophil conditioned media (CM). We demonstrate that CM from neutrophils exposed to resting FM-CM did not affect FM viability or function. Conversely, CM from neutrophils activated by LPS-stimulated FM-CM significantly increased FM secretion of inflammatory IL-6, IL-8, GRO-α, and the markers of membrane weakening, MMP-9 and PGE2 This FM response was partially mediated by ERK signaling and neutrophil extracellular traps through the activation of the DNA sensor, TLR-9. Thus, neutrophils recruited by FMs during infection can propagate FM inflammation and weakening, acting in a feed-forward mechanism to propagate tissue injury at the maternal-fetal interface, increasing the risk of premature FM rupture and preterm birth in women with intrauterine infection.


Subject(s)
Extracellular Traps/immunology , Extraembryonic Membranes/immunology , Inflammation/immunology , MAP Kinase Signaling System/immunology , Neutrophil Activation/immunology , Neutrophils/immunology , Toll-Like Receptor 9/immunology , Chorioamnionitis/immunology , Cytokines/immunology , Female , Humans , Infant, Newborn , Maternal-Fetal Relations , Neutrophil Infiltration/immunology , Pregnancy , Premature Birth/immunology , Reactive Oxygen Species/immunology , Signal Transduction/immunology
17.
Adv Exp Med Biol ; 1398: 251-266, 2023.
Article in English | MEDLINE | ID: mdl-36717499

ABSTRACT

Water homeostasis is essential for fetal growth, and it depends on the successful development of the placenta. Many aquaporins (AQPs) were identified from blastocyst stages to term placenta. In the last years, cytokines, hormones, second messengers, intracellular pH, and membrane proteins were found to regulate their expression and function in the human placenta and fetal membranes. Accumulated data suggest that these proteins may be involved not only in the maintenance of the amniotic fluid volume homeostasis but also in the development of the placenta and fetal organs. In this sense, dysregulation of placental AQPs is associated with gestational disorders. Thus, current evidence shows that AQPs may collaborate in cellular events including trophoblast migration and apoptosis. In addition, aquaglyceroporins are involved in energy metabolism as well as urea elimination across the placenta. In the last year, the presence of AQP9 in trophoblast mitochondria opened new hypotheses about its role in pregnancy. However, much further work is needed to understand the importance of these proteins in human pregnancies.


Subject(s)
Aquaporins , Placenta , Pregnancy , Female , Humans , Placenta/metabolism , Aquaporins/genetics , Aquaporins/metabolism , Fetal Development , Amniotic Fluid , Extraembryonic Membranes
18.
Int J Mol Sci ; 24(4)2023 Feb 17.
Article in English | MEDLINE | ID: mdl-36835482

ABSTRACT

At the feto-maternal interface, fetal membranes (FM) play a crucial role throughout pregnancy. FM rupture at term implicates different sterile inflammation mechanisms including pathways activated by the transmembrane glycoprotein receptor for advanced glycation end-products (RAGE) belonging to the immunoglobulin superfamily. As the protein kinase CK2 is also implicated in the inflammation process, we aimed to characterize the expressions of RAGE and the protein kinase CK2 as a candidate regulator of RAGE expression. The amnion and choriodecidua were collected from FM explants and/or primary amniotic epithelial cells throughout pregnancy and at term in spontaneous labor (TIL) or term without labor (TNL). The mRNA and protein expressions of RAGE and the CK2α, CK2α', and CK2ß subunits were investigated using reverse transcription quantitative polymerase chain reaction and Western blot assays. Their cellular localizations were determined with microscopic analyses, and the CK2 activity level was measured. RAGE and the CK2α, CK2α', and CK2ß subunits were expressed in both FM layers throughout pregnancy. At term, RAGE was overexpressed in the amnion from the TNL samples, whereas the CK2 subunits were expressed at the same level in the different groups (amnion/choriodecidua/amniocytes, TIL/TNL), without modification of the CK2 activity level and immunolocalization. This work paves the way for future experiments regarding the regulation of RAGE expression by CK2 phosphorylation.


Subject(s)
Casein Kinase II , Extraembryonic Membranes , Protein Processing, Post-Translational , Receptor for Advanced Glycation End Products , Humans , Casein Kinase II/metabolism , Extraembryonic Membranes/metabolism , Phosphorylation , Receptor for Advanced Glycation End Products/metabolism
19.
Int J Mol Sci ; 24(13)2023 Jun 29.
Article in English | MEDLINE | ID: mdl-37446059

ABSTRACT

Inflammation of the fetal membranes is an indispensable event of parturition, with increasing prostaglandin E2 (PGE2) synthesis as one of the ultimate products that prime labor onset. In addition to PGE2, the fetal membranes also boast a large capacity for cortisol regeneration. It is intriguing how increased PGE2 synthesis is achieved in the presence of increasing amounts of classical anti-inflammatory glucocorticoids in the fetal membranes at parturition. 15(S)-hydroxyeicosatetraenoic acid (15(S)-HETE) synthesized by lipoxygenase 15/15B (ALOX15/15B) has been shown to enhance inflammation-induced PGE2 synthesis in amnion fibroblasts. Here, we examined whether glucocorticoids could induce ALOX15/15B expression and 15(S)-HETE production to promote PGE2 synthesis in amnion fibroblasts at parturition. We found that cortisol and 15(S)-HETE abundance increased parallelly in the amnion at parturition. Cortisol induced ALOX15/15B expression and 15(S)-HETE production paradoxically in amnion fibroblasts. Mechanism study revealed that this paradoxical induction was mediated by p300-mediated histone acetylation and interaction of glucocorticoid receptor with transcription factors CREB and STAT3. Conclusively, cortisol regenerated in the fetal membranes can paradoxically induce ALOX15/15B expression and 15(S)-HETE production in human amnion fibroblasts, which may further assist in the induction of PGE2 synthesis in the inflammatory responses of the fetal membranes for parturition.


Subject(s)
Amnion , Hydrocortisone , Pregnancy , Female , Humans , Hydrocortisone/metabolism , Amnion/metabolism , Glucocorticoids/metabolism , Dinoprostone/metabolism , Parturition , Extraembryonic Membranes/metabolism , Fibroblasts/metabolism , Inflammation/metabolism , Arachidonate 15-Lipoxygenase/metabolism
20.
Int J Mol Sci ; 24(21)2023 Oct 31.
Article in English | MEDLINE | ID: mdl-37958809

ABSTRACT

Clinically, unique markers in fetal membrane cells may contribute to the search for biomarkers for preterm prelabor rupture of the fetal membranes (pPROM) in maternal blood. pPROM is associated with overwhelming inflammation and premature cellular senescence causing "biological microfractures" of the fetal membranes. We hypothesize that these pathological processes are associated with the shedding of fetal membrane cells into the maternal circulation. The aim of this study was to identify markers expressed exclusively in fetal membrane cells to facilitate their isolation, characterization, and determination of biomarker potential in maternal blood. We have (1), by their transcriptomic profile, identified markers that are upregulated in amnion and chorion tissue compared to maternal white blood cells, and (2), by immunohistochemistry, confirmed the localization of the differentially expressed proteins in fetal membranes, placenta, and the placental bed of the uterus. RNA sequencing revealed 31 transcripts in the amnion and 42 transcripts in the chorion that were upregulated. Among these, 22 proteins were evaluated by immunohistochemistry. All but two transcripts were expressed both on mRNA and protein level in at least one fetal membrane cell type. Among these remaining 20 proteins, 9 proteins were not significantly expressed in the villous and extravillous trophoblasts of the placenta.


Subject(s)
Fetal Membranes, Premature Rupture , Placenta , Infant, Newborn , Humans , Female , Pregnancy , Placenta/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Fetal Membranes, Premature Rupture/genetics , Extraembryonic Membranes/metabolism , Biomarkers/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL