Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 6 de 6
1.
Adv Sci (Weinh) ; 11(21): e2309002, 2024 Jun.
Article En | MEDLINE | ID: mdl-38569496

Preeclampsia (PE) is considered as a disease of placental origin. However, the specific mechanism of placental abnormalities remains elusive. This study identified thrombospondin-1 (THBS1) is downregulated in preeclamptic placentae and negatively correlated with blood pressure. Functional studies show that THBS1 knockdown inhibits proliferation, migration, and invasion and increases the cycle arrest and apoptosis rate of HTR8/SVneo cells. Importantly, THBS1 silencing induces necroptosis in HTR8/SVneo cells, accompanied by the release of damage-associated molecular patterns (DAMPs). Necroptosis inhibitors necrostatin-1 and GSK'872 restore the trophoblast survival while pan-caspase inhibitor Z-VAD-FMK has no effect. Mechanistically, the results show that THBS1 interacts with transforming growth factor B-activated kinase 1 (TAK1), which is a central modulator of necroptosis quiescence and affects its stability. Moreover, THBS1 silencing up-regulates the expression of neuronal precursor cell-expressed developmentally down-regulated 4 (NEDD4), which acts as an E3 ligase of TAK1 and catalyzes K48-linked ubiquitination of TAK1 in HTR8/SVneo cells. Besides, THBS1 attenuates PE phenotypes and improves the placental necroptosis in vivo. Taken together, the down-regulation of THBS1 destabilizes TAK1 by activating NEDD4-mediated, K48-linked TAK1 ubiquitination and promotes necroptosis and DAMPs release in trophoblast cells, thus participating in the pathogenesis of PE.


MAP Kinase Kinase Kinases , Necroptosis , Nedd4 Ubiquitin Protein Ligases , Pre-Eclampsia , Thrombospondin 1 , Trophoblasts , Ubiquitination , Humans , Pre-Eclampsia/metabolism , Pre-Eclampsia/genetics , Female , Pregnancy , Trophoblasts/metabolism , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Kinase Kinases/genetics , Necroptosis/genetics , Nedd4 Ubiquitin Protein Ligases/metabolism , Nedd4 Ubiquitin Protein Ligases/genetics , Thrombospondin 1/metabolism , Thrombospondin 1/genetics , Adult , Placenta/metabolism
2.
Microbiol Spectr ; 11(3): e0007623, 2023 06 15.
Article En | MEDLINE | ID: mdl-37199635

Fetuses diagnosed with fetal growth restriction (FGR) are at an elevated risk of stillbirth and adulthood morbidity. Gut dysbiosis has emerged as one of the impacts of placental insufficiency, which is the main cause of FGR. This study aimed to characterize the relationships among the intestinal microbiome, metabolites, and FGR. Characterization was conducted on the gut microbiome, fecal metabolome, and human phenotypes in a cohort of 35 patients with FGR and 35 normal pregnancies (NP). The serum metabolome was analyzed in 19 patients with FGR and 31 normal pregnant women. Multidimensional data was integrated to reveal the links between data sets. A fecal microbiota transplantation mouse model was used to determine the effects of the intestinal microbiome on fetal growth and placental phenotypes. The diversity and composition of the gut microbiota were altered in patients with FGR. A group of microbial species altered in FGR closely correlated with fetal measurements and maternal clinical variables. Fecal and serum metabolism profiles were distinct in FGR patients compared to those in the NP group. Altered metabolites were identified and associated with clinical phenotypes. Integrated multi-omics analysis revealed the interactions among gut microbiota, metabolites, and clinical measurements. Microbiota from FGR gravida transplanted to mice progestationally induced FGR and placental dysfunction, including impaired spiral artery remodeling and insufficient trophoblast cell invasion. Taken together, the integration of microbiome and metabolite profiles from the human cohort indicates that patients with FGR endure gut dysbiosis and metabolic disorders, which contribute to disease pathogenesis. IMPORTANCE Downstream of the primary cause of fetal growth restriction are placental insufficiency and fetal malnutrition. Gut microbiota and metabolites appear to play an important role in the progression of gestation, while dysbiosis induces maternal and fetal complications. Our study elaborates the significant differences in microbiota profiles and metabolome characteristics between women with FGR and normal pregnancies. This is the first attempt so far that reveals the mechanistic links in multi-omics in FGR, providing a novel insight into host-microbe interaction in placenta-derived diseases.


Fetal Growth Retardation , Gastrointestinal Microbiome , Animals , Female , Humans , Mice , Pregnancy , Dysbiosis , Fetal Growth Retardation/metabolism , Fetal Growth Retardation/microbiology , Placenta/pathology , Cohort Studies , Feces/microbiology , RNA, Ribosomal, 16S/genetics , Bacteria/classification , Bacteria/genetics , Bacteria/metabolism , Adult , Biodiversity , Serum/metabolism
3.
J Extracell Vesicles ; 12(5): e12328, 2023 05.
Article En | MEDLINE | ID: mdl-37165987

Preeclampsia (PE) is a multisystem disorder with high maternal morbidity and mortality rates. Currently, no practical therapeutic approach is available to prevent PE progression, except for early delivery. Gut dysbiosis is associated with PE development. Previous data showed that the abundance of Akkermansia muciniphila (Am) was lower in patients with PE than in normotensive pregnant women. Here, in this study, decreased abundance of Am was observed in a PE mouse model. Also, we found that administration with Am could significantly attenuate systolic blood pressure, promote foetal growth and improve the placental pathology in mice with PE. Moreover, Am-derived extracellular vesicles (AmEVs) were transferred from the gastrointestinal (GI) tract to the placenta and mitigated pre-eclamptic symptoms in PE mice. These beneficial effects of AmEVs were mediated by enhanced trophoblast invasion of the spiral artery (SpA) and SpA remodelling through activation of the epidermal growth factor receptor (EGFR)-phosphatidylinositol-3-kinase (PI3K)-protein kinase B (AKT) signalling pathway. Collectively, our findings revealed the potential benefit of using AmEVs for PE treatment and highlighted important host-microbiota interactions.


Extracellular Vesicles , Pre-Eclampsia , Pregnancy , Female , Mice , Humans , Animals , Placentation , Placenta/metabolism , Placenta/pathology , Pre-Eclampsia/drug therapy , Pre-Eclampsia/prevention & control , Extracellular Vesicles/metabolism
4.
Mol Cell Endocrinol ; 548: 111614, 2022 05 15.
Article En | MEDLINE | ID: mdl-35304192

We previously reported that cyclophilin A (CyPA) production is upregulated in preeclampsia (PE). Moreover, CyPA is known to induce PE-like features in pregnant mice and impair trophoblast invasiveness. In this study, we further illustrated the role of CyPA in PE. RNA-seq analysis, RT-qPCR, immunohistochemical (IHC) staining, and western blotting of mouse placentae revealed that CyPA increased the levels of extracellular matrix (ECM) proteins, such as collagen I and fibronectin, and activated the TGF-ß/Smad3 signaling pathway. Additionally, CyPA inhibited the expression of genes involved in epithelial-mesenchymal transition (EMT) (e.g., E-cadherin, N-cadherin, and vimentin) in mouse placentae. We then constructed stable overexpressing and knock-down CyPA cell models (using HTR8/SVneo cells) to clarify the molecular mechanism. We found that CyPA regulated the levels of ECM-related proteins and the EMT process through the TGF-ß/Smad3 pathway. We also identified SERPINH1 as a putative CyPA-binding protein, using liquid chromatography-electrospray mass spectrometry (LC-MS)/MS. SERPINH1 was found to be upregulated in the placentae of PE. Silencing SERPINH1 expression reversed the upregulation of ECM proteins and inhibition of the EMT process induced by the overexpression of CyPA. These findings revealed the functions of CyPA in the impaired invasiveness of trophoblasts in PE and indicated that CyPA and SERPINH1 may represent promising targets for the treatment of PE.


Cyclophilin A , Epithelial-Mesenchymal Transition , HSP47 Heat-Shock Proteins , Pre-Eclampsia , Trophoblasts , Animals , Cell Movement/genetics , Cyclophilin A/pharmacology , Epithelial-Mesenchymal Transition/genetics , Extracellular Matrix/metabolism , Female , HSP47 Heat-Shock Proteins/metabolism , Humans , Mice , Pre-Eclampsia/genetics , Pre-Eclampsia/metabolism , Pregnancy , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism , Trophoblasts/metabolism
5.
Placenta ; 117: 95-108, 2022 01.
Article En | MEDLINE | ID: mdl-34785431

INTRODUCTION: Abnormal extravillous trophoblast (EVT) function is closely related to preeclampsia (PE) and may be caused by inadequate autophagy, apoptosis, and senescence. Cyclosporin A (CsA) is an effective immunosuppressant that has been reported to stimulate autophagy and exert benign biological effects on EVTs. Therefore, we hypothesized that CsA may display therapeutic efficacy against PE by activating autophagy. METHODS: We established the nitro-l-arginine methyl ester (l-NAME)-induced preeclamptic mice model and a hypoxia-reoxygenation (H/R) model in vitro. The effects of CsA on autophagy were evaluated by western blotting (WB). The effects of CsA on apoptosis were analyzed by Hematoxylin-eosin (H&E) staining, cell apoptosis assay and WB. Senescence-associated ß-galactosidase (SA-ß-gal) staining, RT-qPCR and WB were used to examine the senescence level. RT-qPCR were used to detect the senescence-associated secretory phenotype (SASP) level. DCFH-DA fluorescent probe, dihydroethidium (DHE) staining and mitochondrial membrane potential (ΔΨm) were used to detect senescence-associated mitochondrial dysfunction (SAMD). RESULTS: CsA alleviated PE-like symptoms and reduced placental necrosis and senescence in mice injected with l-NAME. CsA ameliorated placental SASP and SAMD level induced by l-NAME. CsA also upregulated the expression of autophagic proteins in mouse placentas disrupted using l-NAME. In vitro, we found that CsA reversed H/R-induced apoptosis and senescence, as well as decreasing SASP and SAMD levels and upregulating autophagic proteins levels. Notably, 3-methyladenine (3-MA), an early phase inhibitor of autophagosome formation, abolished the protective effects of CsA against H/R. DISCUSSION: CsA may display some therapeutic effects against PE by activating autophagy in vivo and in vitro.


Autophagy/drug effects , Cyclosporine/therapeutic use , Immunosuppressive Agents/therapeutic use , Pre-Eclampsia/drug therapy , Trophoblasts/drug effects , Animals , Apoptosis/drug effects , Cellular Senescence/drug effects , Cyclosporine/pharmacology , Drug Evaluation, Preclinical , Female , Immunosuppressive Agents/pharmacology , Mice , NG-Nitroarginine Methyl Ester , Placenta/pathology , Pre-Eclampsia/pathology , Pregnancy , Senescence-Associated Secretory Phenotype
6.
Nan Fang Yi Ke Da Xue Xue Bao ; 38(9): 1131-1134, 2018 Aug 30.
Article Zh | MEDLINE | ID: mdl-30377109

OBJECTIVE: To investigate the association between maternal serum neutrophil-lymphocyte ratio (NLR) and placental inflammatory response (PIR) in late pregnancy. METHODS: We retrospectively analyzed the clinical and follow-up data of 478 pregnant women undergoing routine prenatal examination and delivery in our hospital in the year 2016. According to the placental pathological results, the women were divided into PIR group (238 cases) and control group (240 cases). The levels of serum inflammatory makers including leukocytes, neutrophils, lymphocytes, C-reactive protein (CRP) and NLR were compared between the two groups to analyze the association of these markers with PIR. Multivariate analysis was performed to identify the independent risk factors of PIR. Logistic regression model was established and the area under the receiver operating characteristic curve (ROC) was used for analyzing the prognostic value of these makers in late pregnancy. RESULTS: The areas under the curve (AUC) of leukocytes, neutrophils, lymphocytes, CRP and NLR were 0.698 (95%CI: 0.485-0.766), 0.716 (95%CI: 0.453-0.783), 0.329 (95%CI: 0.228-0.431), 0.725 (95%CI: 0.677-0.765) and 0.801 (95%CI: 0.742-0.856), respectively. After adjusting the confounders, multivariate logistic regression analysis showed that preterm labor (OR=2.446, 95%CI: 1.003-4.590), premature rupture of membranes (OR=2.304, 95%CI: 1.049-4.161), NLR > 7 (OR=3.268, 95%CI: 2.071-6.920), and CRP > 15 mg/L (OR=2.137, 95%CI: 1.412-8.236) were independent risk factors for PIR. CONCLUSIONS: An increased NLR in late pregnancy can serve as an effective indicator for predicting the risk of PIR.


Chorioamnionitis/blood , Lymphocytes/cytology , Neutrophils/cytology , Biomarkers/blood , C-Reactive Protein/analysis , Case-Control Studies , Female , Humans , Leukocyte Count , Lymphocyte Count , Pregnancy , Prognosis , ROC Curve , Retrospective Studies
...