Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 135
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Circulation ; 149(21): 1670-1688, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38314577

RESUMO

BACKGROUND: Preeclampsia is a serious disease of pregnancy that lacks early diagnosis methods or effective treatment, except delivery. Dysregulated uterine immune cells and spiral arteries are implicated in preeclampsia, but the mechanistic link remains unclear. METHODS: Single-cell RNA sequencing and spatial transcriptomics were used to identify immune cell subsets associated with preeclampsia. Cell-based studies and animal models including conditional knockout mice and a new preeclampsia mouse model induced by recombinant mouse galectin-9 were applied to validate the pathogenic role of a CD11chigh subpopulation of decidual macrophages (dMφ) and to determine its underlying regulatory mechanisms in preeclampsia. A retrospective preeclampsia cohort study was performed to determine the value of circulating galectin-9 in predicting preeclampsia. RESULTS: We discovered a distinct CD11chigh dMφ subset that inhibits spiral artery remodeling in preeclampsia. The proinflammatory CD11chigh dMφ exhibits perivascular enrichment in the decidua from patients with preeclampsia. We also showed that trophoblast-derived galectin-9 activates CD11chigh dMφ by means of CD44 binding to suppress spiral artery remodeling. In 3 independent preeclampsia mouse models, placental and plasma galectin-9 levels were elevated. Galectin-9 administration in mice induces preeclampsia-like phenotypes with increased CD11chigh dMφ and defective spiral arteries, whereas galectin-9 blockade or macrophage-specific CD44 deletion prevents such phenotypes. In pregnant women, increased circulating galectin-9 levels in the first trimester and at 16 to 20 gestational weeks can predict subsequent preeclampsia onset. CONCLUSIONS: These findings highlight a key role of a distinct perivascular inflammatory CD11chigh dMφ subpopulation in the pathogenesis of preeclampsia. CD11chigh dMφ activated by increased galectin-9 from trophoblasts suppresses uterine spiral artery remodeling, contributing to preeclampsia. Increased circulating galectin-9 may be a biomarker for preeclampsia prediction and intervention.


Assuntos
Decídua , Galectinas , Macrófagos , Pré-Eclâmpsia , Remodelação Vascular , Pré-Eclâmpsia/metabolismo , Pré-Eclâmpsia/imunologia , Gravidez , Feminino , Animais , Galectinas/metabolismo , Macrófagos/metabolismo , Macrófagos/imunologia , Macrófagos/patologia , Camundongos , Humanos , Decídua/metabolismo , Decídua/patologia , Camundongos Knockout , Útero/metabolismo , Útero/irrigação sanguínea , Modelos Animais de Doenças , Receptores de Hialuronatos/metabolismo , Receptores de Hialuronatos/genética , Estudos Retrospectivos , Camundongos Endogâmicos C57BL , Antígenos CD11
2.
Cell Mol Life Sci ; 81(1): 208, 2024 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-38710919

RESUMO

Trophoblast stem cells (TSCs) can be chemically converted from embryonic stem cells (ESCs) in vitro. Although several transcription factors (TFs) have been recognized as essential for TSC formation, it remains unclear how differentiation cues link elimination of stemness with the establishment of TSC identity. Here, we show that PRDM14, a critical pluripotent circuitry component, is reduced during the formation of TSCs. The reduction is further shown to be due to the activation of Wnt/ß-catenin signaling. The extinction of PRDM14 results in the erasure of H3K27me3 marks and chromatin opening in the gene loci of TSC TFs, including GATA3 and TFAP2C, which enables their expression and thus the initiation of the TSC formation process. Accordingly, PRDM14 reduction is proposed here as a critical event that couples elimination of stemness with the initiation of TSC formation. The present study provides novel insights into how induction signals initiate TSC formation.


Assuntos
Diferenciação Celular , Proteínas de Ligação a DNA , Fatores de Transcrição , Trofoblastos , Via de Sinalização Wnt , Trofoblastos/metabolismo , Trofoblastos/citologia , Animais , Camundongos , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Diferenciação Celular/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/genética , Fator de Transcrição GATA3/metabolismo , Fator de Transcrição GATA3/genética , Fator de Transcrição AP-2/metabolismo , Fator de Transcrição AP-2/genética , Células-Tronco/metabolismo , Células-Tronco/citologia , Proteínas de Ligação a RNA/metabolismo , Proteínas de Ligação a RNA/genética , Histonas/metabolismo , Histonas/genética
3.
Reproduction ; 167(1)2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-37947291

RESUMO

In brief: Corticotropin-releasing hormone binding protein (CRHBP) is fundamental to the stress response and plays an important role in parturition during pregnancy. This study shows that abnormal CRHBP expression could be an early warning sign of recurrent pregnancy loss and that CRHBP knockdown could suppress HTR8/SVneo cell invasion by the PKC signaling pathway via interacting with CRH receptor 2. Abstract: Trophoblast invasion is critical for placentation and pregnancy success. Trophoblast dysfunction results in many pregnancy complications, including recurrent pregnancy loss (RPL). Corticotropin-releasing hormone binding protein (CRHBP) is fundamental to the stress response and plays an important role in parturition during pregnancy via binding with CRH. To further characterize its function in early pregnancy, we explored the expression of CRHBP in villi during early pregnancy. Compared with normal pregnant women, we demonstrated that the expression of CRHBP decreased in the trophoblasts and villi in RPL patients and that knockdown of CRHBP expression could suppress HTR8/SVneo cell invasion significantly. Our further exploration indicated that the capacity of CRHBP for regulating trophoblast invasion was associated with the PKC signaling pathway via interacting with CRH receptor 2. These findings might provide a new fundamental mechanism for successful pregnancy and a new diagnostic and therapeutic target for RPL.


Assuntos
Aborto Habitual , Receptores de Hormônio Liberador da Corticotropina , Gravidez , Humanos , Feminino , Receptores de Hormônio Liberador da Corticotropina/genética , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Regulação para Baixo , Hormônio Liberador da Corticotropina/genética , Hormônio Liberador da Corticotropina/metabolismo , Linhagem Celular , Trofoblastos/metabolismo , Aborto Habitual/metabolismo , Movimento Celular
4.
Inflamm Res ; 72(7): 1341-1357, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37328599

RESUMO

OBJECTIVE AND DESIGN: To investigate the balancing mechanisms between decidualization-associated inflammation and pregnancy-related immunotolerance. MATERIAL OR SUBJECTS: Decidual samples from women with normal pregnancy (n = 58) or unexplained spontaneous miscarriage (n = 13), peripheral blood from normal pregnancy and endometria from non-pregnancy (n = 10) were collected. Primary endometrial stromal cells (ESCs), decidual stromal cells (DSCs), decidual immune cells (DICs) and peripheral blood mononuclear cells (PBMCs) were isolated. TREATMENT: The plasmid carrying neuropilin-1 (NRP1) gene was transfected into ESC for overexpression. To induce decidualization in vitro, ESCs were treated with a combination of 10 nM estradiol, 100 nM progesterone and 0.5 mM cAMP. Anti-Sema3a and anti-NRP1 neutralizing antibodies were applied to block the ligand-receptor interactions. METHODS: RNA-seq analysis was performed to identify differentially expressed genes in DSCs and DICs, and NRP1 expression was verified by Western blotting and flow cytometry. The secretion of inflammatory mediators was measured using a multifactor cytometric bead array. The effects of Sema3a-NRP1 pathway on DICs were determined by flow cytometry. Statistical differences between groups were compared using the T test and one way or two-way ANOVA. RESULTS: Combined with five RNA-seq datasets, NRP1 was the only immune checkpoint changing oppositely between DSCs and DICs. The decreased expression of NRP1 in DSCs allowed intrinsic inflammatory responses required for decidualization, while its increased expression in DICs enhanced tolerant phenotypes beneficial to pregnancy maintenance. DSC-secreted Sema3a promoted immunosuppression in DICs via NRP1 binding. In women with miscarriage, NRP1 was abnormally elevated in DSCs but diminished in decidual macrophages and NK cells. CONCLUSION: NRP1 is a multifunctional controller that balances the inflammatory states of DSCs and DICs in gravid uterus. Abnormal expression of NRP1 is implicated in miscarriage.


Assuntos
Aborto Espontâneo , Decídua , Humanos , Gravidez , Feminino , Decídua/metabolismo , Neuropilina-1/genética , Neuropilina-1/metabolismo , Leucócitos Mononucleares/metabolismo , Células Cultivadas , Células Estromais/metabolismo
5.
Int J Mol Sci ; 24(2)2023 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-36675047

RESUMO

T-cell immunoglobulin mucin-3 (Tim-3) is an important checkpoint that induces maternal-fetal tolerance in pregnancy. Macrophages (Mφs) play essential roles in maintaining maternal-fetal tolerance, remodeling spiral arteries, and regulating trophoblast biological behaviors. In the present study, the formation of the labyrinth zone showed striking defects in pregnant mice treated with Tim-3 neutralizing antibodies. The adoptive transfer of Tim-3+Mφs, rather than Tim-3-Mφs, reversed the murine placental dysplasia resulting from Mφ depletion. With the higher production of angiogenic growth factors (AGFs, including PDGF-AA, TGF-α, and VEGF), Tim-3+dMφs were more beneficial in promoting the invasion and tube formation ability of trophoblasts. The blockade of AGFs in Tim-3+Mφs led to the narrowing of the labyrinthine layer of the placenta, compromising maternal-fetal tolerance, and increasing the risk of fetal loss. Meanwhile, the AGFs-treated Tim-3-Mφs could resolve the placental dysplasia and fetal loss resulting from Mφ depletion. These findings emphasized the vital roles of Tim-3 in coordinating Mφs-extravillous trophoblasts interaction via AGFs to promote pregnancy maintenance and in extending the role of checkpoint signaling in placental development. The results obtained in our study also firmly demonstrated that careful consideration of reproductive safety should be taken when selecting immune checkpoint and AGF blockade therapies in real-world clinical care.


Assuntos
Comunicação Celular , Macrófagos , Placenta , Manutenção da Gravidez , Trofoblastos , Animais , Feminino , Camundongos , Gravidez , Decídua/metabolismo , Receptor Celular 2 do Vírus da Hepatite A/genética , Receptor Celular 2 do Vírus da Hepatite A/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Macrófagos/metabolismo , Placenta/metabolismo , Manutenção da Gravidez/genética , Manutenção da Gravidez/fisiologia , Trofoblastos/metabolismo , Comunicação Celular/genética , Comunicação Celular/fisiologia
6.
Int J Mol Sci ; 24(9)2023 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-37175543

RESUMO

Iron is necessary for various critical biological processes, but iron overload is also dangerous since labile iron is redox-active and toxic. We found that low serum iron and decidual local iron deposition existed simultaneously in recurrent pregnancy loss (RPL) patients. Mice fed with a low-iron diet (LID) also showed iron deposition in the decidua and adverse pregnancy outcomes. Decreased ferroportin (cellular iron exporter) expression that inhibited the iron export from decidual stromal cells (DSCs) might be the reason for local iron deposition in DSCs from low-serum-iron RPL patients and LID-fed mice. Iron supplementation reduced iron deposition in the decidua of spontaneous abortion models and improved pregnancy outcomes. Local iron overload caused ferroptosis of DSCs by downregulating glutathione (GSH) and glutathione peroxidase 4 levels. Both GSH and cystine (for the synthesis of GSH) supplementation reduced iron-induced lipid reactive oxygen species (ROS) and cell death in DSCs. Ferroptosis inhibitor, cysteine, and GSH supplementation all effectively attenuated DSC ferroptosis and reversed embryo loss in the spontaneous abortion model and LPS-induced abortion model, making ferroptosis mitigation a potential therapeutic target for RPL patients. Further study that improves our understanding of low-serum-iron-induced DSC ferroptosis is needed to inform further clinical evaluations of the safety and efficacy of iron supplementation in women during pregnancy.


Assuntos
Aborto Habitual , Ferroptose , Sobrecarga de Ferro , Gravidez , Humanos , Feminino , Animais , Camundongos , Ferro/metabolismo , Ferroptose/fisiologia , Aborto Habitual/metabolismo , Células Estromais/metabolismo , Sobrecarga de Ferro/metabolismo
7.
Reproduction ; 163(5): 241-250, 2022 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-35174787

RESUMO

Decidual stromal cells (DSCs) modulate the function of trophoblasts through various factors. Wnt signaling pathway is active at the maternal-fetal interface. Here, we isolated endometrial stromal cells (ESCs) from women of reproductive ages and DSCs from normal pregnancy during the first trimester (6-10 weeks). Real-time quantitative PCR and western blotting were used to screen out the most variable WNT ligands between ESCs and DSCs, which turned out to be WNT16. Both culture mediums from DSCs and recombinant protein of human WNT16 enhanced the survival and invasion of HTR8/SVneo trophoblastic cells. Furthermore, the regulation of DSCs on trophoblast was partly blockaded after we knocked down WNT16 in DSCs. Treating HTR8/SVneo trophoblastic cells with small molecular inhibitors and small interfering RNA (siRNA), we found that the activity of AKT/beta-catenin (CTNNB1) correlated with the effect of WNT16. The crosstalk of WNT16/AKT/beta-catenin between DSCs and trophoblasts was determined to be downregulated in unexplained recurrent spontaneous abortion. This study suggests that WNT16 from DSCs promotes HTR8/SVneo trophoblastic cells invasion and survival via AKT/beta-catenin pathway at the maternal-fetal interface in human early pregnancy. The disturbance of this crosstalk between DSCs and trophoblasts might cause pregnancy failure.


Assuntos
Aborto Habitual , Trofoblastos , Aborto Habitual/metabolismo , Movimento Celular , Feminino , Humanos , Gravidez , Primeiro Trimestre da Gravidez , Proteínas Proto-Oncogênicas c-akt/metabolismo , Células Estromais/metabolismo , Trofoblastos/metabolismo , Proteínas Wnt/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo
8.
FASEB J ; 35(8): e21754, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34191338

RESUMO

To obtain a successful pregnancy, the establishment of maternal-fetal tolerance and successful placentation are required to be established. Disruption of this immune balance and/or inadequate placental perfusion is believed to be associated with a lot of pregnancy-related complications, such as recurrent spontaneous abortion, pre-eclampsia, and fetal intrauterine growth restriction. Extravillous trophoblasts (EVTs) have the unique ability to instruct decidual immune cells (DICs) to develop a regulatory phenotype for fetal tolerance. Utilizing immortalized human first trimester extravillous trophoblast cells and primary EVTs, we found that DICs promote EVT function and placental development. We have previously shown that checkpoints T-cell immunoglobulin mucin-3 (Tim-3) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) are important for DIC function. In the present study, we showed that blockade of Tim-3 and CTLA-4 pathways leaded to the abnormal DICs-EVTs interaction, poor placental development, and increased fetal loss. Treatment with IL-4 and IL-10 could rescue the adverse effects of targeting Tim-3 and CTLA-4 on the pregnancy outcome. Hence, the reproductive safety must be a criterion considered in the assessment of immuno-therapeutic agents. In addition, IL-4 and IL-10 may represent novel therapeutic strategies to prevent pregnancy loss induced by checkpoint inhibition.


Assuntos
Antígeno CTLA-4/imunologia , Decídua/imunologia , Receptor Celular 2 do Vírus da Hepatite A/imunologia , Interleucina-10/imunologia , Interleucina-4/imunologia , Trofoblastos/imunologia , Animais , Antígeno CTLA-4/antagonistas & inibidores , Comunicação Celular/imunologia , Células Cultivadas , Decídua/citologia , Perda do Embrião/imunologia , Feminino , Receptor Celular 2 do Vírus da Hepatite A/antagonistas & inibidores , Humanos , Tolerância Imunológica , Interleucina-10/administração & dosagem , Interleucina-4/administração & dosagem , Masculino , Troca Materno-Fetal/imunologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos CBA , Camundongos Endogâmicos DBA , Modelos Imunológicos , Placentação/imunologia , Gravidez , Resultado da Gravidez , Transdução de Sinais/imunologia , Trofoblastos/citologia
9.
J Biomed Sci ; 29(1): 101, 2022 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-36419076

RESUMO

BACKGROUND: Sleep disturbance can cause adverse pregnancy outcomes by changing circadian gene expression. The potential mechanisms remain unclear. Decidualization is critical for the establishment and maintenance of normal pregnancy, which can be regulated by circadian genes. Whether Rev-erbα, a critical circadian gene, affects early pregnancy outcome by regulating decidualization needs to be explored. METHODS: QPCR, western blot and artificial decidualization mouse model were used to confirm the effect of sleep disturbance on Rev-erbα expression and decidualization. The regulatory mechanism of Rev-erbα on decidualization was assessed using QPCR, western blot, RNA-Seq, and Chip-PCR. Finally, sleep disturbance mouse model was used to investigate the effect of therapeutic methods targeting Rev-erbα and interleukin 6 (IL-6) on improving adverse pregnancy outcomes induced by sleep disturbance. RESULTS: Dysregulation of circadian rhythm due to sleep disturbance displayed abnormal expression profile of circadian genes in uterine including decreased level of Rev-erbα, accompanied by defective decidualization. Rev-erbα could regulate decidualization by directly repressing IL-6, which reduced the expression of CCAAT/enhancer-binding protein ß (C/EBPß) and its target insulin-like growth factor binding protein 1 (IGFBP1), the marker of decidualization, by inhibiting progesterone receptors (PR) expression. Moreover, deficient decidualization, higher abortion rate and lower implantation number were exhibited in the mouse models with sleep disturbance compared with those in normal mouse. Pharmacological activation of Rev-erbα or neutralization of IL-6 alleviated the adverse effect of sleep disturbance on pregnancy outcomes. CONCLUSIONS: Taken together, Rev-erbα regulated decidualization via IL-6-PR-C/EBPß axis and might be a connector between sleep and pregnancy outcome. Therapies targeting Rev-erbα and IL-6 might help improving adverse pregnancy outcomes induced by sleep disturbance.


Assuntos
Interleucina-6 , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares , Animais , Feminino , Camundongos , Gravidez , Ritmo Circadiano/genética , Interleucina-6/genética , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/genética , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/metabolismo , Receptores de Interleucina-6 , Sono , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo
10.
Reprod Biomed Online ; 44(5): 803-816, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35339367

RESUMO

RESEARCH QUESTION: Increased granulosa cell division is associated with abnormal folliculogenesis in polycystic ovary syndrome (PCOS). Lethal-7i microRNA (let-7i) may play an important role in the follicular development and granulosa cell growth; therefore is let-7i involved in PCOS pathogenesis? DESIGN: The expression of let-7i was measured in granulosa-luteal cells (GLC) from women with or without PCOS. A human granulosa cell line, KGN, was used for the functional study. Mimics and inhibitors of let-7i, lentiviruses expressing insulin-like growth factor 2 mRNA binding protein (IMP2), and small-interfering RNAs were transfected into KGN cells. KGN cell proliferation was determined by 5-ethynyl-2'-deoxyuridine (EdU) and Cell Counting Kit-8 (CCK-8) assays. The cell cycle and apoptosis were assessed by propidium iodide-annexin V (PI-A) staining and fluorescence-activated cell sorting. Oestradiol concentration was determined by enzyme-linked immunoassay. Bioinformatics analysis and luciferase reporter assay were applied to confirm the let-7i target genes. RESULTS: The study showed that let-7i was down-regulated in PCOS GLC (P = 0.001). Mimics of let-7i inhibited KGN proliferation (P = 0.001), and decreased aromatase expression (P = 0.030) and oestradiol production (P = 0.029), whereas let-7i inhibitors had the opposite effect. Bioinformatics analysis and quantitative real-time (qRT) PCR identified IMP2 as a target of let-7i (P = 0.021). qRT-PCR and western blot analysis indicated that IMP2 was up-regulated in GLC in women with PCOS (P = 0.001 and P = 0.044), and IMP2 expression was suppressed by let-7i in KGN cells (P < 0.001). Luciferase reporter assay results (P = 0.002), combined with the rescue assay, confirmed that let-7i inhibited KGN cell proliferation and reduced oestradiol concentration by directly targeting IMP2. CONCLUSIONS: let-7i was down-regulated in PCOS GLC. Overexpression of let-7i inhibited KGN cell proliferation and decreased oestradiol production in an IMP2-dependent manner, providing a new molecular mechanism for PCOS.


Assuntos
Células Lúteas , MicroRNAs , Síndrome do Ovário Policístico , Feminino , Humanos , Apoptose/fisiologia , Proliferação de Células/fisiologia , Estradiol/metabolismo , Células da Granulosa/metabolismo , Células Lúteas/metabolismo , Células Lúteas/patologia , MicroRNAs/genética , MicroRNAs/metabolismo , Síndrome do Ovário Policístico/metabolismo
11.
Proc Natl Acad Sci U S A ; 116(3): 890-899, 2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30593567

RESUMO

The AML1-ETO fusion protein, generated by the t(8;21) chromosomal translocation, is causally involved in nearly 20% of acute myeloid leukemia (AML) cases. In leukemic cells, AML1-ETO resides in and functions through a stable protein complex, AML1-ETO-containing transcription factor complex (AETFC), that contains multiple transcription (co)factors. Among these AETFC components, HEB and E2A, two members of the ubiquitously expressed E proteins, directly interact with AML1-ETO, confer new DNA-binding capacity to AETFC, and are essential for leukemogenesis. However, the third E protein, E2-2, is specifically silenced in AML1-ETO-expressing leukemic cells, suggesting E2-2 as a negative factor of leukemogenesis. Indeed, ectopic expression of E2-2 selectively inhibits the growth of AML1-ETO-expressing leukemic cells, and this inhibition requires the bHLH DNA-binding domain. RNA-seq and ChIP-seq analyses reveal that, despite some overlap, the three E proteins differentially regulate many target genes. In particular, studies show that E2-2 both redistributes AETFC to, and activates, some genes associated with dendritic cell differentiation and represses MYC target genes. In AML patients, the expression of E2-2 is relatively lower in the t(8;21) subtype, and an E2-2 target gene, THPO, is identified as a potential predictor of relapse. In a mouse model of human t(8;21) leukemia, E2-2 suppression accelerates leukemogenesis. Taken together, these results reveal that, in contrast to HEB and E2A, which facilitate AML1-ETO-mediated leukemogenesis, E2-2 compromises the function of AETFC and negatively regulates leukemogenesis. The three E proteins thus define a heterogeneity of AETFC, which improves our understanding of the precise mechanism of leukemogenesis and assists development of diagnostic/therapeutic strategies.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Leucemia Mieloide Aguda/etiologia , Proteínas de Fusão Oncogênica/metabolismo , Proteína 1 Parceira de Translocação de RUNX1/metabolismo , Proteína 2 Semelhante ao Fator 7 de Transcrição/metabolismo , Diferenciação Celular , Linhagem Celular Tumoral , Humanos , Leucemia Mieloide Aguda/metabolismo , Recidiva
12.
J Cell Mol Med ; 25(16): 7690-7708, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34145738

RESUMO

The maternal-foetal interface is an immune-privileged site where the semi-allogeneic embryo is protected from attacks by the maternal immune system. Uterine macrophages are key players in establishing and maintaining pregnancy, and the dysregulation of the M1-M2 subpopulation balance causes abortion. We separated two distinct mouse uterine macrophage subpopulations during early pregnancy, CD45+ F4/80+ CD206- M1-like (M1) and CD45+ F4/80+ CD206+ M2-like (M2) cells. The M1 preponderance was significantly exaggerated at 6 hours after lipopolysaccharide (LPS) treatment, and adoptive transfer of M2 macrophages partially rescued LPS-induced abortion. RNA sequencing analysis of mouse uterine M2 versus M1 revealed 1837 differentially expressed genes (DEGs), among which 629 was up-regulated and 1208 was down-regulated. Histone deacetylase 9 (Hdac9) was one of the DEGs and validated to be significantly up-regulated in uterine M2 as compared with M1. Remarkably, this differential expression profile between M1 and M2 was also evident in primary splenic macrophages and in vitro polarized murine peritoneal, bone marrow-derived and RAW 264.7 macrophages. In Hdac9/HDAC9 knockout RAW 264.7 and human THP-1-derived macrophages, the expression of M1 differentiation markers was unchanged or decreased whereas M2 markers were increased compared with the wild-type cells, and these effects were unrelated to compromised proliferation. Furthermore, Hdac9/HDAC9 ablation significantly enhanced the phagocytosis of fluorescent microspheres in M2 Raw 264.7 cells yet decreased the capacity of THP-1-derived M1 macrophages. The above results demonstrate that Hdac9/HDAC9 deficiency exaggerates M2 macrophage polarization in mouse and human macrophages, which may provide clues for our understanding of the epigenetic regulation on macrophage M1/M2 polarization in maternal-foetal tolerance.


Assuntos
Feto/imunologia , Histona Desacetilases/fisiologia , Lipopolissacarídeos/toxicidade , Macrófagos/imunologia , Proteínas Repressoras/fisiologia , Útero/imunologia , Animais , Células Cultivadas , Modelos Animais de Doenças , Feminino , Feto/efeitos dos fármacos , Feto/metabolismo , Perfilação da Expressão Gênica/métodos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Fagocitose , Útero/efeitos dos fármacos , Útero/metabolismo
13.
BMC Immunol ; 22(1): 57, 2021 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-34399700

RESUMO

BACKGROUND: Circadian rhythm is an important player for reproduction. Rev-erbα, a significant clock gene, is involved in regulating cell differentiation, inflammation and metabolism. Macrophage polarization plays crucial roles in immune tolerance at the maternal-fetus interface, which also modulates the initiation and resolution of inflammation. Alteration of macrophage polarization induces adverse pregnancy outcomes such as infertility, recurrent spontaneous abortion and preterm labor. RESULTS: Decidual macrophages from LPS-induced mice abortion model displayed M1-like bias, accompanied by decreased expression of Rev-erbα. SR9009, an agonist of Rev-erbα, may reduce lipopolysaccharide (LPS)-induced M1 polarization of macrophages via activation of PI3K but not NF-κB signaling pathway. Furthermore, SR9009 could reduce M1-like polarization of decidual macrophages induced by LPS and attenuate LPS-induced resorption rates in mice model. CONCLUSIONS: Both in vivo and in vitro experiments demonstrated that the pharmacological activation of Rev-erbα using SR9009 could attenuate the effect of LPS on macrophage polarization and protect pregnancy. This study may provide a potential therapeutic strategy for miscarriage induced by inflammation.


Assuntos
Aborto Espontâneo/prevenção & controle , Decídua/imunologia , Inflamação/imunologia , Macrófagos/imunologia , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/metabolismo , Animais , Diferenciação Celular , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Lipopolissacarídeos/imunologia , Camundongos , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/agonistas , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/genética , Gravidez , Pirrolidinas/farmacologia , Transdução de Sinais , Células Th1/imunologia , Tiofenos/farmacologia , Células U937
14.
Biol Reprod ; 104(2): 410-417, 2021 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-31329823

RESUMO

Normal pregnancy is associated with several immune adaptations in both systemic and local maternal-fetal interface to allow the growth of semi-allogeneic conceptus. A failure in maternal immune tolerance to the fetus may result in abnormal pregnancies, such as recurrent spontaneous abortion. The regulation of T-cell homeostasis during pregnancy has important implications for maternal tolerance and immunity. Cytotoxic T-lymphocyte antigen-4 (CTLA-4) and T-cell immunoglobulin mucin-3 (Tim-3) are important negative immune regulatory molecules involved in viral persistence and tumor metastasis. Here we described the lower frequency of splenic T cells co-expressing CTLA-4 and Tim-3 accompanied by higher levels of proinflammatory but lower anti-inflammatory cytokines production in abortion-prone mouse model. Blockade of CTLA-4 and Tim-3 pathways leaded to the dysfunction of splenic T cells. By the higher expression during normal pregnancy, CTLA-4 and Tim-3 co-expression on splenic T cells linked to immunosuppressive phenotype. As the spleen is an important site for peripheral immune activation, our data suggest potential noninvasive biomarkers and therapeutic targets for miscarriage.


Assuntos
Aborto Animal/patologia , Antígeno CTLA-4/metabolismo , Baço/metabolismo , Aborto Animal/genética , Animais , Antígeno CTLA-4/genética , Feminino , Regulação da Expressão Gênica , Imunoglobulina G , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos CBA , Gravidez , Subpopulações de Linfócitos T
15.
Reproduction ; 162(2): 161-170, 2021 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-34115609

RESUMO

Deficient decidualization of endometrial stromal cells (ESCs) can cause adverse pregnancy outcomes including miscarriage, intrauterine growth restriction, and pre-eclampsia. Decidualization is regulated by multiple factors such as hormones and circadian genes. Melatonin, a circadian-controlled hormone, is reported to be important for various reproductive processes, including oocyte maturation and placenta development. Its receptor, MT1, is considered to be related to intrauterine growth restriction and pre-eclampsia. However, the role of melatonin-MT1 signal in decidualization remains unknown. Here, we reported that decidual stromal cells from miscarriages displayed deficient decidualization with decreased MT1 expression. The expression level of MT1 is gradually increased with the process of decidualization induction in vitro. MT1 knockdown suppressed the decidualization level, while the overexpression of MT1 promoted the decidualization process. Moreover, changing MT1 level could regulate the expression of decidualization-related transcription factor FOXO1. Melatonin promoted decidualization and reversed the decidualization deficiency due to MT1 knockdown. Using in vitro and in vivo experiments, we further identified that lipopolysaccharide (LPS) could induce inflammation and decidualization resistance with downregulated MT1 expression, and melatonin could reverse the inflammation and decidualization resistance induced by LPS. These results suggested that the melatonin-MT1 signal might be essential for decidualization and might provide a novel therapeutic target for decidualization deficiency-associated pregnancy complications.


Assuntos
Aborto Espontâneo/patologia , Decídua/patologia , Endométrio/patologia , Regulação da Expressão Gênica , Melatonina/metabolismo , Receptor MT1 de Melatonina/metabolismo , Aborto Espontâneo/etiologia , Aborto Espontâneo/metabolismo , Adulto , Animais , Estudos de Casos e Controles , Células Cultivadas , Decídua/metabolismo , Endométrio/metabolismo , Feminino , Humanos , Camundongos , Gravidez , Adulto Jovem
16.
Reproduction ; 162(2): 107-115, 2021 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-33999841

RESUMO

The T-box transcription factor protein eomesodermin (Eomes) is known for both homeostasis and function of effector and memory CD8+T cells. However, much less is known about the functional regulation of Eomes on CD8+ T cells during pregnancy. In the present study, we concluded the higher Eomes expression dCD8+T cells during normal early pregnancy. The number of Eomes+dCD8+T cells decreased in miscarriage. This Eomes+dCD8+T cell subset also expressed less growth-promoting factors, shifted toward pro-inflammatory phenotype in miscarriage. Primary Trophoblasts and HTR8/SVneo cell line could increase Eomes expression of dCD8+T cells from both normal early pregnancy and miscarriage, which might provide a new strategy for therapy to promote maternal-fetal tolerance and prevent pregnancy loss. These findings indicated that Eomes might be promising early warming targets of miscarriage. In addition, this study suggested that the reproductive safety must be a criterion considered in modulating the dose and function of Eomes in CD8+T cells to reverse T cell exhaustion.


Assuntos
Aborto Espontâneo/patologia , Linfócitos T CD8-Positivos/imunologia , Diferenciação Celular , Tolerância Imunológica , Proteínas com Domínio T/metabolismo , Aborto Espontâneo/etiologia , Estudos de Casos e Controles , Feminino , Humanos , Gravidez , Proteínas com Domínio T/genética
17.
Reproduction ; 161(1): 1-10, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33112295

RESUMO

To obtain a successful pregnancy, trophoblasts must provide a physical barrier, suppress maternal reactivity, produce immunosuppressive hormones locally, and enhance the production of blocking factors that are able to bind to several antigenic sites. Inadequate placental perfusion has been closely associated with several pregnancy-associated diseases. Galectin-9 (Gal-9) has a wide variety of regulatory functions in innate and adaptive immunity during infection, tumor growth, and organ transplantation. We utilized immortalized human first-trimester extravillous trophoblast cells (HTR8/SVneo) for our functional study and examined the effects of Gal-9 on apoptosis, cytokine production and angiogenesis of HTR8/SVneo cells. Gal-9 inhibited the apoptosis and IFN-γ and IL-17A production, promoted IL-4 production, and coordinated the crosstalk between HTR8/SVneo cells and human umbilical vein endothelial cells via its interaction with Tim-3. Blockade of JNK signaling inhibited Gal-9 activities in HTR8/SVneo cells. In addition, we detected a correlation between low levels of Gal-9 and spontaneous abortion. So Gal-9 could inhibit the apoptosis and proinflammatory cytokine expression, and promote the angiogenesis and IL-4 production in HTR8/SVneo cells via Tim-3 in a JNK dependent manner to help the maintenance of normal pregnancy. These findings possibly identify Gal-9 as a key regulator of trophoblast cells and suggest its potential as a biomarker and target for the treatment of recurrent pregnancy loss.


Assuntos
Aborto Habitual/metabolismo , Galectinas/metabolismo , Sistema de Sinalização das MAP Quinases , Placentação , Trofoblastos/fisiologia , Feminino , Receptor Celular 2 do Vírus da Hepatite A/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Gravidez
18.
Reproduction ; 161(5): 573-580, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33780908

RESUMO

Decidualization is the functional transformation process of endometrium in response to ovarian steroids dedicated to support embryo development. Defective decidualization is closely associated with various pregnancy complications such as recurrent miscarriage (RM). Dual specificity MAPK phosphatases (MKPs) are a family of phosphatases specifically regulating mitogen-activated protein kinase (MAPK) signaling with dual specificity for threonine and tyrosine. Here, using RNA-seq,we found that dual specificity phosphatase 1 (DUSP1) expression was prominently elevated among the MKP family members in db-cAMP treated primary human endometrial stromal cells (ESCs). We verified that its induction by db-cAMP in ESCs was in a dose- and time-dependent manner and that primary human decidual stromal cells (DSCs) present higher expression of DUSP1 than ESCs. A protein kinase A (PKA) inhibitor H-89 abolished its induction in ESCs, but not ESI-09, an EPAC1/2 inhibitor. Knock-down of TORC2/3 but not CREB by siRNA in ESCs diminished its induction by db-cAMP. Furthermore, knock-down of DUSP1, as well as TORC2/3 by siRNA caused abnormal activation of JNK during db-cAMP induction in ESCs, accompanied by decreased IGFBP1 expression, an ESC decidualization indicator, which could be fully rescued by a JNK inhibitor SP600125. In addition, Western blot showed that DUSP1 expression was reduced in the DSCs of patients with RM, along with JNK overactivation and decreased IGFBP1 expression. In conclusion, our results demonstrated that TORC2/3-mediated DUSP1 upregulation in response to the cAMP/PKA signaling safeguards IGFBP1 expression via restraining JNK activity, indicating its involvement in ESC decidualization, and that aberrant expression of DUSP1 in DSCs might engage in the pathogenesis of RM.


Assuntos
Aborto Habitual/patologia , Decídua/patologia , Fosfatase 1 de Especificidade Dupla/metabolismo , Endométrio/patologia , Células Estromais/patologia , Fatores de Transcrição/metabolismo , Aborto Habitual/genética , Aborto Habitual/metabolismo , Estudos de Casos e Controles , Decídua/metabolismo , Fosfatase 1 de Especificidade Dupla/genética , Endométrio/metabolismo , Feminino , Humanos , Gravidez , RNA-Seq , Transdução de Sinais , Células Estromais/metabolismo , Fatores de Transcrição/genética
19.
Acta Biochim Biophys Sin (Shanghai) ; 53(9): 1207-1215, 2021 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-34259317

RESUMO

Endometritis is an inflammatory disease of the endometrium, which is responsible for endometrial dysfunction, decidualization failure, and increased incidence of early pregnancy loss. SCM-198, a synthetic form of leonurine, is well known to possess anti-inflammatory effects. SCM-198 has been reported to display beneficial effects on endometritis. However, the specific mechanisms of SCM-198 in preventing endometritis remain unknown. In this study, we focused on the molecular mechanism of SCM-198 in inhibiting endometritis. The anti-inflammatory effects and the related signaling pathways of SCM-198 were studied in vitro using human endometrial stromal cells (hESCs). Reverse transcriptase-polymerase chain reaction and western blot analysis results demonstrated that SCM-198 markedly inhibited lipopolysaccharide (LPS)-induced endometrial inflammatory response by suppressing the LPS-JNK-cJUN/cFOS-TLR4-NF-κB pathway. The preventive and therapeutic effects of SCM-198 on endometrial inflammation were explored by using a mouse model of LPS-induced endometritis. SCM-198 produced essentially the same effects when administered either post-treatment (after LPS) or pre-treatment (before LPS) via vaginal or intraperitoneal administration. In vivo results indicated that SCM-198 is a potential effective drug for the treatment of endometritis.


Assuntos
Anti-Inflamatórios/farmacologia , Endometrite/prevenção & controle , Ácido Gálico/análogos & derivados , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Adulto , Animais , Anti-Inflamatórios/uso terapêutico , Citocinas/metabolismo , Endometrite/induzido quimicamente , Endometrite/patologia , Endométrio/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Feminino , Ácido Gálico/farmacologia , Ácido Gálico/uso terapêutico , Humanos , Lipopolissacarídeos/toxicidade , Camundongos Endogâmicos C57BL , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Células Estromais/efeitos dos fármacos , Receptor 4 Toll-Like/antagonistas & inibidores , Receptor 4 Toll-Like/metabolismo
20.
Acta Biochim Biophys Sin (Shanghai) ; 53(10): 1333-1341, 2021 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-34343226

RESUMO

Endometrial dysfunction is an important factor for implantation failure. The function of the endometrium is regulated by multiple factors like sex hormones and circadian rhythms. Endometrial stromal cells (ESCs) are a major cellular component in the endometrium, which is essential for proper physiological activities of the endometrium and the establishment of pregnancy. Melatonin, as a circadian-controlled hormone, plays beneficial roles in the regulation of reproductive processes. MT1, a melatonin receptor, can regulate cell proliferation and apoptosis. Whether melatonin-MT1 signal affects biological function of ESCs remains unknown. Here, we showed that MT1 was expressed in human ESCs (hESCs), which could be regulated by estrogen and progesterone. MT1 knockdown inhibited proliferative activity and promoted apoptosis of hESCs by activating caspase-3 and upregulating the Bax/Bcl2 ratio. Melatonin could reverse the effect of MT1 knockdown on proliferative activity and apoptosis of hESCs. Melatonin could promote proliferative activity of hESCs via the JNK/P38 signal pathway and repress the apoptosis of hESCs via the JNK signal pathway. Moreover, in vivo experiments showed that MT1 expression was decreased in endometrial cells from mice with disrupted circadian rhythm, accompanied by increased apoptosis and suppressed proliferative activity, which could be alleviated by administration of melatonin. These results showed the regulatory effect of melatonin-MT1 signal on biological behaviors of ESCs, which might provide a novel therapeutic strategy for endometrial dysfunction induced by disrupted circadian rhythm.


Assuntos
Endométrio/metabolismo , Melatonina/farmacologia , Receptor MT1 de Melatonina/metabolismo , Células Estromais/metabolismo , Adulto , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Ritmo Circadiano , Modelos Animais de Doenças , Endométrio/citologia , Endométrio/efeitos dos fármacos , Estrogênios/fisiologia , Feminino , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Progesterona/fisiologia , Receptor MT1 de Melatonina/agonistas , Receptor MT1 de Melatonina/genética , Células Estromais/efeitos dos fármacos , Regulação para Cima/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA