Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
Add more filters

Publication year range
1.
Biol Res ; 57(1): 13, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38561846

ABSTRACT

BACKGROUND: Endometrial fibrosis, a significant characteristic of intrauterine adhesion (IUA), is caused by the excessive differentiation and activation of endometrial stromal cells (ESCs). Glutaminolysis is the metabolic process of glutamine (Gln), which has been implicated in multiple types of organ fibrosis. So far, little is known about whether glutaminolysis plays a role in endometrial fibrosis. METHODS: The activation model of ESCs was constructed by TGF-ß1, followed by RNA-sequencing analysis. Changes in glutaminase1 (GLS1) expression at RNA and protein levels in activated ESCs were verified experimentally. Human IUA samples were collected to verify GLS1 expression in endometrial fibrosis. GLS1 inhibitor and glutamine deprivation were applied to ESCs models to investigate the biological functions and mechanisms of glutaminolysis in ESCs activation. The IUA mice model was established to explore the effect of glutaminolysis inhibition on endometrial fibrosis. RESULTS: We found that GLS1 expression was significantly increased in activated ESCs models and fibrotic endometrium. Glutaminolysis inhibition by GLS1 inhibitor bis-2-(5-phenylacetamido-1,2,4-thiadiazol-2-yl) ethyl sulfide (BPTES or glutamine deprivation treatment suppressed the expression of two fibrotic markers, α-SMA and collagen I, as well as the mitochondrial function and mTORC1 signaling in ESCs. Furthermore, inhibition of the mTORC1 signaling pathway by rapamycin suppressed ESCs activation. In IUA mice models, BPTES treatment significantly ameliorated endometrial fibrosis and improved pregnancy outcomes. CONCLUSION: Glutaminolysis and glutaminolysis-associated mTOR signaling play a role in the activation of ESCs and the pathogenesis of endometrial fibrosis through regulating mitochondrial function. Glutaminolysis inhibition suppresses the activation of ESCs, which might be a novel therapeutic strategy for IUA.


Subject(s)
Glutamine , Mitochondria , Female , Mice , Humans , Animals , Glutamine/metabolism , Fibrosis , Mitochondria/pathology , Mechanistic Target of Rapamycin Complex 1/metabolism , RNA/metabolism , Endometrium/metabolism , Endometrium/pathology
2.
J Assist Reprod Genet ; 41(6): 1645-1659, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38512656

ABSTRACT

PURPOSE: The window of implantation (WOI) is a brief period during which the endometrium is receptive to embryo implantation. This study investigated the relationship between miR-135a-5p and endometrial receptivity. METHODS: Peripheral blood was collected on the day of ovulation and the 5th day after ovulation for high-throughput sequencing from women who achieved clinical pregnancy through natural cycle frozen embryo transfer. RT-qPCR assessed miR-135a-5p expression in the endometrium tissue or cells during the mouse implantation window or decidualization. Scanning electron microscopy was utilized to observe pinopode morphology and quantity in mice overexpressing miR-135a-5p during the WOI. Human endometrial stromal cells (HESC) and artificial induction of mouse uterine decidualization were used to explore whether miR-135a-5p overexpression inhibits decidualization by regulating HOXA10 and BMPR2. Furthermore, the impact of miR-135a-5p on HESC proliferation and HTR8/SVneo invasion was explored. RESULTS: A total of 54 women were enrolled in the study. bioinformatics analysis and animal models demonstrated that miR-135a-5p was significantly downregulated during the WOI, and its high expression can lead to abnormal pregnancy outcomes. Overexpression of miR-135a-5p resulted in the absence of pinopode in mouse endometrial tissue during the WOI. High miR-135a-5p levels were found to potentially inhibit endometrial tissue decidualization by downregulating HOXA10 and BMPR2 expression. Finally, CEBPD was identified as a potential regulator of miR-135a-5p, which would explain the decreased miR-135a-5p expression during the WOI. CONCLUSION: MiR-135a-5p expression is significantly downregulated during the WOI. High miR-135a-5p levels suppress pinopode development and endometrial tissue decidualization through HOXA10 and BMPR2, contributing to inadequate endometrial receptivity.


Subject(s)
Decidua , Embryo Implantation , Endometrium , Homeobox A10 Proteins , MicroRNAs , Stromal Cells , Female , MicroRNAs/genetics , MicroRNAs/metabolism , Embryo Implantation/genetics , Humans , Mice , Stromal Cells/metabolism , Endometrium/metabolism , Animals , Pregnancy , Adult , Decidua/metabolism , Homeobox A10 Proteins/genetics , Homeobox A10 Proteins/metabolism , Embryo Transfer
3.
J Biol Chem ; 298(5): 101874, 2022 05.
Article in English | MEDLINE | ID: mdl-35358514

ABSTRACT

We previously reported that CCAAT/enhancer-binding protein beta (C/EBPß) is the pioneer factor inducing transcription enhancer mark H3K27 acetylation (H3K27ac) in the promoter and enhancer regions of genes encoding insulin-like growth factor-binding protein-1 (IGFBP-1) and prolactin (PRL) and that this contributes to decidualization of human endometrial stromal cells (ESCs). Peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α; PPARGC1A) is a transcriptional coactivator known to regulate H3K27ac. However, although PGC-1α is expressed in ESCs, the potential role of PGC-1α in mediating decidualization is unclear. Here, we investigated the involvement of PGC-1α in the regulation of decidualization. We incubated ESCs with cAMP to induce decidualization and knocked down PPARGC1A to inhibit cAMP-induced expression of IGFBP-1 and PRL. We found cAMP increased the recruitment of PGC-1α and p300 to C/EBPß-binding sites in the promoter and enhancer regions of IGFBP-1 and PRL, corresponding with increases in H3K27ac. Moreover, PGC-1α knockdown inhibited these increases, suggesting PGC-1α forms a histone-modifying complex with C/EBPß and p300 at these regions. To further investigate the regulation of PGC-1α, we focused on C/EBPß upstream of PGC-1α. We found cAMP increased C/EBPß recruitment to the novel enhancer regions of PPARGC1A. Deletion of these enhancers decreased PGC-1α expression, indicating that C/EBPß upregulates PGC-1α expression by binding to novel enhancer regions. In conclusion, PGC-1α is upregulated by C/EBPß recruitment to novel enhancers and contributes to decidualization by forming a histone-modifying complex with C/EBPß and p300, thereby inducing epigenomic changes in the promoters and enhancers of IGFBP-1 and PRL.


Subject(s)
Histones , Insulin-Like Growth Factor Binding Protein 1 , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Cyclic AMP/metabolism , Gene Expression Regulation , Histones/genetics , Histones/metabolism , Humans , Insulin-Like Growth Factor Binding Protein 1/genetics , Insulin-Like Growth Factor Binding Protein 1/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Prolactin/genetics , Prolactin/metabolism , Stromal Cells/metabolism
4.
FASEB J ; 36(5): e22319, 2022 05.
Article in English | MEDLINE | ID: mdl-35429060

ABSTRACT

Bone morphogenetic protein 2 (BMP2) has been shown to act as a critical regulator in the processes of embryo implantation and endometrial decidualization. The expression and production of pentraxin 3 (PTX3) is essential for successful pregnancy, and aberrant production of PTX3 is involved in the pathogenesis of several vascular complications during pregnancy. Studies have shown that several transforming growth factor ß superfamily members, including BMP2, can regulate female reproductive function by modulating the expression of PTX3 in human granulosa cells. However, to date, whether BMP2 can regulate the production of PTX3 during endometrial decidualization remains to be elucidated. In this study, we aimed to explore the effect of BMP2 on the expression and production of PTX3 and the underlying molecular mechanisms using immortalized human endometrial stromal cells (I-HESCs) and human decidual stromal cells (HDSCs). We demonstrated that treatment with exogenous BMP2 significantly suppressed PTX3 production by decreasing the mRNA level of PTX3 in both I-HESCs and HDSCs. The results also showed that BMP2 activated SMAD signaling by inducing an increase in the protein levels of phosphorylated SMAD1/5/8, and this effect could be abolished by pretreatment with the ALK2/3 inhibitor DMH-1 but not with the ALK1/4/7 inhibitor SB431542. Additionally, combined knockdown of ALK2 and ALK3 completely reversed the BMP2-induced suppressive effect on PTX3 expression, while concomitant knockdown of SMAD1 and SMAD5 or knockdown of SMAD4 completely reversed the BMP2-induced suppressive effect on PTX3 expression. Taken together, these results indicate that BMP2 suppressed PTX3 production by decreasing PTX expression, which is mediated by a canonical ALK2/3-mediated SMAD1/5-SMAD4-dependent signaling pathway. Our findings suggest that BMP2 may potentially regulate the process of endometrial decidualization by suppressing the production of PTX3 in humans.


Subject(s)
Bone Morphogenetic Protein 2 , Decidua , Serum Amyloid P-Component , Bone Morphogenetic Protein 2/metabolism , C-Reactive Protein/genetics , C-Reactive Protein/metabolism , Cells, Cultured , Decidua/metabolism , Female , Humans , Pregnancy , Serum Amyloid P-Component/genetics , Serum Amyloid P-Component/metabolism , Stromal Cells/metabolism
5.
J Assist Reprod Genet ; 40(11): 2739-2750, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37831348

ABSTRACT

PURPOSE: To investigate the expression and underlying mechanism of RPA2 in endometrium of patients with repeated implantation failure (RIF). METHODS: In this study, we retrieved the expression profiles from GEO databases and filtered the differentially expressed genes between RIF and the fertile control group. Ultimately, RPA2 was confirmed as a target gene. RPA2 expression in endometrial tissues of RIF patients, the control group, and different phases was detected by RT-qPCR, immunohistochemistry, and Western blotting. The role of RPA2 in endometrial decidualization was performed by in vitro decidualization inducing by 8-Br-cAMP and MPA. Furthermore, RT-qPCR was used to detect changes in the decidual biomarkers after transfection of RPA2 overexpression vector in human endometrium stromal cell (HESC). RESULTS: RPA2 was significantly upregulated in the mid-secretory endometrium of patients with RIF. As a proliferation-related gene, RPA2 was obviously higher expressed at proliferative phase during the normal menstrual cycles. Moreover, the downregulation of RPA2 was discovered during decidualization of HESC. Furthermore, RPA2 overexpression impaired decidualization by inhibiting the expression of prolactin (PRL) and insulin-like growth factor-binding protein 1 (IGFBP1). CONCLUSIONS: Our finding indicated that aberrant upregulation of RPA2 attenuated decidualization of HESC in RIF women and provided new potential therapeutic targets.


Subject(s)
Decidua , Endometrium , Humans , Female , Decidua/metabolism , Endometrium/metabolism , Fertility , Biomarkers/metabolism , Immunohistochemistry , Stromal Cells/metabolism , Embryo Implantation/genetics , Replication Protein A/metabolism
6.
J Biol Chem ; 297(4): 101150, 2021 10.
Article in English | MEDLINE | ID: mdl-34478711

ABSTRACT

Human endometrial stromal cells (ESCs) differentiate into decidual cells by the action of progesterone, which is essential for implantation and maintenance of pregnancy. We previously reported that glucose uptake by human ESCs increases during decidualization and that glucose is indispensable for decidualization. Although glucose transporter 1 (GLUT1) is upregulated during decidualization, it remains unclear whether it is involved in glucose uptake. Here, we attempted to determine the role of GLUT1 during decidualization as well as the factors underlying its upregulation. ESCs were incubated with cAMP to induce decidualization. Knockdown of GLUT1 suppressed cAMP-increased glucose uptake and the expressions of specific markers of decidualization, IGF-binding protein-1 (IGFBP-1), and prolactin (PRL). To investigate the regulation of GLUT1 expression, we focused on CCAAT enhancer-binding protein ß (C/EBPß) and Wilms' tumor 1 (WT1) as the upstream transcription factors regulating GLUT1 expression. Knockdown of either C/EBPß or WT1 suppressed cAMP-increased GLUT1 expression and glucose uptake. cAMP treatment also increased the recruitment of C/EBPß and WT1 to the GLUT1 promoter region. Interestingly, cAMP increased the H3K27 acetylation (H3K27ac) and p300 recruitment in the GLUT1 promoter region. Knockdown of C/EBPß or WT1 inhibited these events, indicating that both C/EBPß and WT1 contribute to the increase of H3K27ac by recruiting p300 to the GLUT1 promoter region during decidualization. These findings indicate that GLUT1 is involved in glucose uptake in ESCs during decidualization, thus facilitating the establishment of pregnancy.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Decidua/metabolism , Epigenesis, Genetic , Glucose Transporter Type 1/biosynthesis , Up-Regulation , WT1 Proteins/metabolism , Adult , CCAAT-Enhancer-Binding Protein-beta/genetics , Female , Glucose Transporter Type 1/genetics , Humans , Middle Aged , Stromal Cells , WT1 Proteins/genetics
7.
Exp Cell Res ; 404(2): 112659, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34022204

ABSTRACT

Endometriosis is a benign gynecological disease that is manifested by the presence and growth of endometrial cells and glands outside the uterine. Active angiogenesis, migration, and invasion of endometrial tissue outside the uterine are critical for the development of endometriosis and lead to the survival and growth of endometriotic lesions. Metformin, as an anti-diabetic agent, represents anti-angiogenic property. Here, we performed a study using human normal endometrial stromal cells (N-ESCs) from healthy endometrial tissue and human eutopic endometrial stromal cells (EU-ESCs) and ectopic endometrial stromal cells (ECT-ESCs) from endometriosis patients. ESCs were cultured and treated with different concentrations of Metformin (0-20 mmol/l) for 72 h to evaluate Metformin effect on cell viability, proliferation, migration was measured by methyl thiazolyl tetrazolium (MTT) assay and scratch test respectively as well as expression of angiogenesis and migration markers. The Metformin reduced cell migration, and proliferation of endometriotic stromal cells in a time and concentration dependently manner. Furthermore, Metformin attenuated the expression of angiogenic and inflammatory genes in human endometriotic stromal cells. The direct anti-proliferative effect on ECT-ESCs combined with the effects of Metformin on inflammatory and angiogenesis-related genes expression supports its therapeutic potential for endometriosis. Metformin could be used as an effective adjuvant in endometriosis treatment.


Subject(s)
Endometrium/drug effects , Metformin/pharmacology , Neovascularization, Pathologic/metabolism , Stromal Cells/metabolism , Cell Movement/drug effects , Cell Movement/physiology , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cells, Cultured , Endometriosis/drug therapy , Endometriosis/genetics , Endometrium/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Humans , Metformin/metabolism , Stromal Cells/drug effects
8.
Biochem Biophys Res Commun ; 571: 174-180, 2021 09 24.
Article in English | MEDLINE | ID: mdl-34330061

ABSTRACT

Decidualization - the differentiation of endometrial stromal cells (ESCs) into decidual cells - is a crucial step for successful embryo implantation and placentation that is initiated in the secretory phase of the menstrual cycle. During decidualization, ESCs undergo proliferation arrest and secrete inflammatory mediators, including senescence-associated secretory phenotype (SASP). Although several senolytic agents improve age-related diseases, their effects on cellular senescence in decidualizing ESCs has not been explored. To do this, we treated decidualized ESCs with the senolytic agents Quercetin (Que), Dasatinib (Das), and BPTES. Que decreased the number of senescence-associated ß-galactosidase (SA-ß-Gal) positive cells and expression of senescence markers in ESCs treated with the decidual stimulus (dibutyryl-cAMP plus progesterone: DP). Concomitantly, Que markedly increased the expression of the decidualization markers IGFBP1, PRL, and FOXO1, in decidualizing ESCs. Similar to Que, Das also stimulated decidualization. Treatment with a combination of Que and Das synergistically increased the expression of decidualization markers and senescence markers compared with treatment with Que or Das alone. However, BPTES did not enhance the expression of decidualization markers. These results imply that treatment with Que and/or Das can remove senescent decidual cells and enhance the decidualization of the rest of ESCs. Thus, senolytic modulation of abnormal ESC decidualization could alleviate infertility caused by dysfunctions of endometrial receptivity and embryo implantation.


Subject(s)
Dasatinib/pharmacology , Endometrium/drug effects , Quercetin/pharmacology , Stromal Cells/drug effects , Sulfides/pharmacology , Thiadiazoles/pharmacology , Cells, Cultured , Cellular Senescence/drug effects , Female , Humans
9.
Reprod Biol Endocrinol ; 19(1): 70, 2021 May 14.
Article in English | MEDLINE | ID: mdl-33990206

ABSTRACT

Adenomyosis (ADS) is an estrogen-dependent gynecological disease with unspecified etiopathogenesis. Local hyperestrogenism may serve a key role in contributing to the origin of ADS. Talin1 is mostly identified to be overexpressed and involved in the progression of numerous human carcinomas through mediating cell proliferation, adhesion and motility. Whether Talin1 exerts an oncogenic role in the pathogenesis of ADS and puts an extra impact on the efficacy of estrogen, no relevant data are available yet. Here we demonstrated that the adenomyotic eutopic and ectopic endometrial stromal cells (ADS_Eu_ESC and ADS_Ec_ESC) treated with ß-estradiol (ß-E2) presented stronger proliferative and pro-angiogenetic capacities, accompanied by increased expression of PCNA, Ki67, VEGFB and ANGPTL4 proteins. Meanwhile, these promoting effects were partially abrogated by Fulvestrant (ICI 182780, an estrogen-receptor antagonist). Aberrantly upregulation of Talin1 mRNA and protein level was observed in ADS endometrial specimens and stromal cells. Through performing functional experiments in vitro, we further determined that merely overexpression of Talin1 (OV-Talin1) also enhanced ADS stromal cell proliferation and pro-angiogenesis, while the most pronounced facilitating effects were found in the co-intervention group of OV-Talin1 plus ß-E2 treatment. Results from the xenograft nude mice model showed that the hypodermic endometrial lesions from co-intervention group had the highest mean weight and volume, compared with that of individual OV-Talin1 or ß-E2 treatment. The expression levels of PCNA, Ki67, VEGFB and ANGPTL4 in the lesions were correspondingly elevated the most in the co-intervention group. Our findings unveiled that overexpressed Talin1 might cooperate withß-E2 in stimulating ADS endometrial stromal cell proliferation and neovascularization, synergistically promoting the growth and survival of ectopic lesions. These results may be beneficial to provide a new insight for clarifying the pathogenesis of ADS.


Subject(s)
Adenomyosis/physiopathology , Endometrium/pathology , Stromal Cells/physiology , Talin/physiology , Adenocarcinoma , Adenomyosis/genetics , Adenomyosis/metabolism , Animals , Cell Division/drug effects , Cell Line, Tumor , Cells, Cultured , Colony-Forming Units Assay , Endometrial Neoplasms , Estradiol/pharmacology , Female , Gene Expression Regulation/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Myometrium/pathology , Neovascularization, Pathologic/physiopathology , Neovascularization, Physiologic/drug effects , RNA Interference , RNA, Small Interfering/genetics , Recombinant Proteins/metabolism , Specific Pathogen-Free Organisms , Stromal Cells/drug effects , Talin/biosynthesis , Talin/genetics , Up-Regulation
10.
Gynecol Endocrinol ; 37(2): 171-176, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32840162

ABSTRACT

AIMS: Human endometrium resists embryo implantation except during the window period. Currently, uterine HURP expression is known to be involved in endometrial stromal proliferation during embryo implantation of mice. Thus, we demonstrated hepatoma up-regulated protein (HURP) expression in the human endometrium during the menstrual cycle, as well as HURP regulation in endometrial stromal cells (ESCs). MATERIALS AND METHODS: We collected human endometrial samples from different menstrual cycle phases (early/late proliferative, and early/mid/late secretory), and then analyzed these samples by immunohistochemistry, reverse transcription-polymerase chain reaction, and Western blotting. We also assessed the effects of two sex-steroid hormones, 17ß-estradiol (E2) and 4-pregnene-3,20-dione (P4) on the cultured stromal cells. RESULTS: HURP protein was localized to the nucleus of the endometrial both epithelial and stromal cells in all stages. Also, HURP mRNA and protein in human endometrial tissue was significantly up-regulated during late-proliferative and secretory phase, compared with early-proliferative phase. In ESCs, HURP expression was regulated by E2, but not P4. CONCLUSIONS: We indicated that cyclic changes in HURP expression in human normal ESC strongly suggested up-regulation by estrogen. Taken together, since estrogen responses are fundamental in endometrial biology, uterine expression of HURP may be involved in female reproductive function during the menstrual cycle.


Subject(s)
Endometrium/metabolism , Menstrual Cycle/metabolism , Neoplasm Proteins/metabolism , Adult , Female , Humans , In Vitro Techniques
11.
Acta Biochim Biophys Sin (Shanghai) ; 53(3): 365-371, 2021 Mar 02.
Article in English | MEDLINE | ID: mdl-33462609

ABSTRACT

Homeobox A10 (HOXA10) is a transcription factor belonging to the homeobox gene family. It is highly expressed in endometrial stromal cells (ESCs) and plays essential roles in the proliferation and differentiation of endometrium, the establishment of endometrial receptivity and embryo implantation. However, little is known about the target genes and signaling pathways regulated by HOXA10 in ESCs. In this study, we identified 1830 transcripts regulated by HOXA10 in ESCs by RNA interference (RNAi) and RNA-sequencing (RNA-seq) analysis, of which 980 were positively regulated by HOXA10 and 850 were negatively regulated by HOXA10. Interestingly, matrix metallopeptidase-11 was downregulated by HOXA10 in stromal cells verified by quantitative real-time polymerase chain reaction and western blot analysis. Pathway analysis demonstrated that the target genes were enriched in various pathways, including cellular metabolism, DNA replication and repair, cell junction, and lysosome and signal transduction. The results of the present study provide novel insights into the mechanism underlying HOXA10 regulation in ESCs and may identify novel targets for the diagnosis and treatment of endometrium-related infertility.


Subject(s)
Endometrium/metabolism , Gene Expression Regulation , Homeobox A10 Proteins/metabolism , RNA-Seq , Signal Transduction , Female , Humans , Stromal Cells/metabolism
12.
BMC Genomics ; 21(1): 103, 2020 Jan 30.
Article in English | MEDLINE | ID: mdl-32000663

ABSTRACT

BACKGROUND: Luman is a member of CREB3 (cAMP responsive element-binding) subfamily of the basic leucine-zipper (bZIP) transcription factors. It may play an important regulatory role during the decidualization process since Luman was highly expressed in the decidual cells. However, the exact molecular mechanisms of how Luman regulating decidualization is unknown. RESULTS: Using an in vitro model, we prove that Luman knockdown significantly affects the decidualization process of mice endometrial stromal cells (ESCs) as the expression of two decidual markers PRL8a2 and PRL3c1 were repressed. We employed massively parallel RNA sequencing (RNA-Seq) to understand the changes in the transcriptional landscape associated with knockdown of Luman in ESCs during in vitro decidualization. We found significant dysregulation of genes related to protein processing in the endoplasmic reticulum (ER). Several genes involved in decidualization including bone morphogenetic proteins (e.g. BMP1, BMP4, BMP8A, BMP2, and BMP8B), growth factor-related genes (e.g. VEGFB, FGF10, and FGFR2), and transcription factors (IF4E, IF4A2, WNT4, WNT9A, ETS1, NOTCH1, IRX1, IDB1, IDB2, and IDB3), show altered expression. We also found that the knockdown of Luman is associated with increased expression of cell cycle-related genes including cycA1, cycB1, cycB2, CDK1, CDK2, and PLPK1, which resulted in an increased proportion of ESCs in the G1 phase. Differentially expressed genes (DEGs) were highly enriched on ECM-receptor interaction signaling, endoplasmic reticulum protein processing, focal adhesion, and PI3K-Akt signaling pathways. CONCLUSIONS: Luman knockdown results in widespread gene dysregulation during decidualization of ESCs. Genes involved in protein processing in ER, bone morphogenetic protein, growth factor, and cell cycle progression were identified as particularly important for explaining the decidual deficiency observed in this in vitro model. Therefore, this study provides clues as to the underlying mechanisms that may expand our understanding of gene regulation during decidualization.


Subject(s)
Cyclic AMP Response Element-Binding Protein/genetics , Decidua/metabolism , Gene Expression Profiling/veterinary , Gene Regulatory Networks , Animals , Cyclic AMP Response Element-Binding Protein/metabolism , Endometrium/metabolism , Female , Gene Expression Regulation , Gene Knockdown Techniques , High-Throughput Nucleotide Sequencing/veterinary , Mice , Sequence Analysis, RNA/veterinary
13.
Biol Reprod ; 103(1): 104-113, 2020 06 23.
Article in English | MEDLINE | ID: mdl-32285109

ABSTRACT

Successful assisted reproductive technology pregnancy depends on the viability of embryos and endometrial receptivity. However, the literature has neglected effects of the endometrial environment during the proliferative phase on implantation success or failure. Human endometrial stromal cells (hESCs) were isolated from endometrial tissues sampled at oocyte retrieval during the proliferative phase from women undergoing infertility treatment. Primary hESC cultures were used to investigate the relationship between stemness and senescence induction in this population and embryo receptivity. Patients were classified as receptive or non-receptive based on their pregnancy diagnosis after embryo transfer. Biomarkers of cellular senescence and somatic stem cells were compared between each sample. hESCs from non-receptive patients exhibited significantly higher (P < 0.01) proportions of senescent cells, mRNA expressions of CDKN2A and CDKN1A transcripts (P < 0.01), and expressions of genes encoding the senescence-associated secretory phenotype (P < 0.05). hESCs from receptive patients had significantly higher (P < 0.01) mRNA expressions of ABCG2 and ALDH1A1 transcripts. Our findings suggest that stemness is inversely associated with senescence induction in hESCs and, by extension, that implantation failure in infertility treatment may be attributable to a combination of senescence promotion and disruption of this maintenance function in this population during the proliferative phase of the menstrual cycle. This is a promising step towards potentially improving the embryo receptivity of endometrium. The specific mechanism by which implantation failure is prefigured by a loss of stemness among endometrial stem cells, and cellular senescence induction among hESCs, should be elucidated in detail in the future.


Subject(s)
Cellular Senescence/physiology , Embryo Implantation/physiology , Endometrium/cytology , Endometrium/physiology , Reproductive Techniques, Assisted , Stromal Cells/physiology , Adult , Biomarkers/analysis , Cell Cycle Checkpoints , Cells, Cultured , Cellular Senescence/genetics , Chemokines/analysis , Cytokines/analysis , Embryo Transfer , Female , Gene Expression , Humans , Infertility, Female/therapy , Middle Aged , Stem Cells/physiology , Stromal Cells/chemistry , Treatment Failure , beta-Galactosidase/analysis
14.
J Obstet Gynaecol Res ; 46(9): 1809-1818, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32643241

ABSTRACT

AIM: Human endometrial stromal cells (HESCs) were previously shown to be capable of discriminating embryos with different qualities. Here we aimed to compare the specific response of the HESC secretome to implanted blastocyst-conditioned medium (BCM) versus nonimplanted medium and identify cytokine candidates useful for the assessment of blastocyst implantation. METHODS: Cleavage embryos were individually cultured in one microdrop of medium for blastocyst formation. The BCM was collected after fresh blastocyst transfer on day 5 and used to supplement HESC culture medium. A high-throughput antibody array covering 440 cytokines was used to detect the secretory proteins of HESCs supplemented with implanted or nonimplanted BCM. RESULTS: A total of 22 differentially expressed proteins were found out of 440 cytokines in the supernatant of HESCs supplemented with BCM from the implanted group compared to the nonimplanted group, including seven upregulated and 15 downregulated proteins. Gene Ontology enrichment analysis showed that the differentially expressed proteins were mainly involved in cell chemotaxis and motility, and ERK1/2 cascade regulation. Kyoto Encyclopedia of Genes and Genomes analysis suggested that the mitogen-activated protein kinase and phosphatidylinositol 3 kinase/Akt pathways were mainly involved. CONCLUSION: HESCs specifically responded to BCM from different quality blastocysts, a finding that can be used to develop a novel approach for blastocyst quality assessment.


Subject(s)
Blastocyst , Embryo Implantation , Culture Media, Conditioned/pharmacology , Endometrium , Female , Humans , Stromal Cells
15.
Hum Reprod ; 34(4): 635-645, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30715393

ABSTRACT

STUDY QUESTION: Can menstrual stem cells (MenSCs) inhibit myofibroblast differentiation and reverse transforming growth factor ß (TGFß)-mediated activation of myofibroblast phenotypes in human endometrial stromal cells (ESCs)? SUMMARY ANSWER: MenSCs suppressed endometrial myofibroblast differentiation and reversed TGFß-mediated activation of myofibroblast phenotypes, which might be associated with activation of the Hippo/TAZ pathway. WHAT IS KNOWN ALREADY: The potential effect of MenSCs as a cell therapy include attenuation of intrauterine adhesions, but the underlying mechanisms by which MenSCs exerts these effects are not entirely understood. STUDY DESIGN, SIZE, DURATION: We evaluated the antagonistic effects of MenSCs on myofibroblast differentiation as well as the broader effect of the Hippo/TAZ signaling pathway on TGFß-mediated induction of myofibroblast gene expression. The study design was based on a cohort of clinical proliferative phase endometrial samples obtained from three healthy premenopausal females with regular menstrual cycles. PARTICIPANTS/MATERIALS, SETTING, METHODS: ESCs were cocultured with MenSCs or in MenSC-conditioned medium. Fibrotic markers (αSMA, collagen I, CTGF and fibronectin) as well as proliferation and wound-healing abilities were evaluated. Components of the Hippo/TAZ pathway (TAZ, p-TAZ, MOB1, p-MOB1, LATS1 and p-LATS1) were also investigated. Cell Counting Kit 8, wound healing assay, real-time PCR, western blotting, immunofluorescence and shRNA knockdown approaches were used to validate the findings. MAIN RESULTS AND THE ROLE OF CHANCE: MenSCs inhibited myofibroblast activation, resulting in more rapid proliferation of ESCs. MenSCs downregulated the expression of myofibroblast markers αSMA and collagen I and promoted endometrial wound healing. Coculture with MenSCs also attenuated the TGFß-mediated increase in expression of fibrotic marker genes αSMA, collagen I, CTGF and fibronectin, and restored the wound-healing ability inhibited by TGFß. MenSCs induced Hippo/TAZ pathway activation, resulting in nuclear export and cytoplasmic retention of TAZ. TAZ inhibition was demonstrated to have similar effects even in the absence of MenSCs, and inhibition of TAZ was sufficient to attenuate TGFß-mediated myofibroblast activation. LARGE SCALE DATA: N/A. LIMITATIONS, REASONS FOR CAUTION: This study included only in vitro experiments. Thus, additional data from in vivo experiments are needed in a future study. WIDER IMPLICATIONS OF THE FINDINGS: The Hippo/TAZ pathway may be an important therapeutic target for endometrial fibrosis. STUDY FUNDING/COMPETING INTEREST(S): This study was supported by the National Natural Science Foundation of China (No. 81601236) and Zhejiang Provincial Natural Science Foundation of China (LY19H040009). None of the authors has any competing interests to declare.


Subject(s)
Endometrium/cytology , Menstruation/metabolism , Myofibroblasts/metabolism , Protein Serine-Threonine Kinases/metabolism , Stem Cells/metabolism , Stromal Cells/metabolism , Trans-Activators/metabolism , Transforming Growth Factor beta/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Coculture Techniques , Culture Media, Conditioned/pharmacology , Female , Hippo Signaling Pathway , Humans , Signal Transduction/drug effects , Stromal Cells/drug effects , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Transforming Growth Factor beta/pharmacology
16.
Mol Reprod Dev ; 86(7): 908-921, 2019 07.
Article in English | MEDLINE | ID: mdl-31041824

ABSTRACT

Endoplasmic reticulum (ER) stress is involved in regulating cell metabolism, apoptosis, autophagy, and survival. However, there is not enough information about the role of ER stress in lipopolysaccharide (LPS)-induced apoptosis and inflammatory cytokine secretion in the uterus. In this study, we found that LPS induced apoptosis and inflammation in goat endometrial stromal cells (ESCs). LPS treatment inhibited cell viability and cell proliferation. In addition, the genes associated with proliferation, such as proliferating cell nuclear antigen and MKI67, were affected by LPS treatment. Moreover, LPS increased the secretion of interleukin (IL)-1ß and IL-8, promoting the levels of MYD88, caspase1, and TRL4. The 4-phenylbutyric acid pretreatment inhibited the expression of unfolded protein response proteins and the secretion of inflammatory cytokines in LPS-treated cells. However, blockage of inositol-requiring enzyme 1 and activating transcription factor 6 did not significantly reduce apoptosis and inflammatory cytokine secretion. Collectively, ER stress involved in LPS-induced apoptosis and inflammatory cytokine increased in goat ESCs. This study provides new insight into the function of ER stress in the pathological process.


Subject(s)
Apoptosis/drug effects , Endometritis/chemically induced , Endometrium/cytology , Endoplasmic Reticulum Stress , Goats/metabolism , Lipopolysaccharides/pharmacology , Stromal Cells/metabolism , Activating Transcription Factor 6/genetics , Animals , Apoptosis/genetics , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Cytokines/metabolism , Endometritis/metabolism , Endoribonucleases/genetics , Female , Gene Knockdown Techniques , Phenylbutyrates/pharmacology , Protein Serine-Threonine Kinases/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Transfection , Unfolded Protein Response/drug effects
17.
Reprod Biomed Online ; 38(4): 549-559, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30772194

ABSTRACT

Endometriosis is a complex and heterogeneous disorder of unknown aetiology. This benign disease possesses special biological behaviours that mimic those of malignant tumours. A pro-endometriotic niche established by an existing lesion is a supportive micro-environment for the progression of endometriosis. After the accumulation of cells by an existing lesion, these components display distinct characteristics that impair immune surveillance. Subsequent retrograde menstruation of endometrial stromal cells into the pro-endometriotic niche facilitates endometriotic progression from early initiation to an advanced lesion. This study aimed to highlight the innovative role of the pro-endometriotic niche in endometriosis, and to provide valuable treatment targets for endometriosis.


Subject(s)
Chemokines/immunology , Cytokines/immunology , Endometriosis/physiopathology , Endometrium/physiopathology , Menstruation Disturbances/physiopathology , Cell Adhesion , Cell Proliferation , Cells, Cultured , Dendritic Cells/cytology , Disease Progression , Endometriosis/blood , Endometriosis/diagnosis , Endometriosis/therapy , Endometrium/pathology , Epithelial Cells/cytology , Extracellular Matrix/metabolism , Female , Humans , Hypoxia , Immunosuppressive Agents , Inflammation , Killer Cells, Natural/cytology , Menstruation , Monitoring, Immunologic , Neoplasm Invasiveness , Neovascularization, Pathologic/pathology , Stromal Cells/cytology , Th17 Cells/cytology
18.
Reprod Biomed Online ; 39(4): 556-568, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31377021

ABSTRACT

RESEARCH QUESTION: Endometriosis is a common gynaecological disease defined by the presence of endometrium-like tissue outside the uterus. This complex disease, often accompanied by severe pain and infertility, causes a significant medical and socioeconomic burden; hence, novel strategies are being sought for the treatment of endometriosis. Here, we set out to explore the cytotoxic effects of a panel of compounds to find toxins with different efficiency in eutopic versus ectopic cells, thus highlighting alterations in the corresponding molecular pathways. DESIGN: The effect on cellular viability of 14 compounds was established in a cohort of paired eutopic and ectopic endometrial stromal cell samples from 11 patients. The biological targets covered by the panel included pro-survival enzymes, cytoskeleton proteins, the proteasome and the cell repair machinery. RESULTS: Protein kinase inhibitors GSK690693, ARC-775 and sorafenib, proteasome inhibitor bortezomib, and microtubule-depolymerizing toxin monomethyl auristatin E were more effective in eutopic cells. In contrast, 10 µmol/l of the anthracycline toxin doxorubicin caused cellular death in ectopic cells more effectively than in eutopic cells. The large-scale sequencing of mRNA isolated from doxorubicin-treated and control cells indicated different survival strategies in eutopic versus ectopic endometrium. CONCLUSIONS: Overall, the results confirm evidence of large-scale metabolic reprogramming in endometriotic cells, which underlies the observed differences in sensitivity towards toxins. The enhanced efficiency of doxorubicin interfering with redox equilibria and/or DNA repair mechanisms pinpoints key players that can be potentially used to selectively target ectopic lesions in endometriosis.


Subject(s)
Drug Resistance/physiology , Endometriosis/pathology , Endometrium/drug effects , Endometrium/pathology , Peritoneal Diseases/pathology , Adult , Aminobenzoates/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Caspase 3/drug effects , Caspase 3/metabolism , Cell Survival/drug effects , Cells, Cultured , Doxorubicin/pharmacology , Endometrium/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Humans , Necrosis/pathology , Oligopeptides/pharmacology , Oxadiazoles/pharmacology , Sorafenib/pharmacology , Stromal Cells/drug effects , Stromal Cells/metabolism , Stromal Cells/pathology , Young Adult
19.
Acta Biochim Biophys Sin (Shanghai) ; 51(6): 580-587, 2019 Jun 20.
Article in English | MEDLINE | ID: mdl-31032514

ABSTRACT

Increasing amounts of evidence demonstrated that accumulative reactive oxygen species (ROS) and apoptosis of human endometrial stromal cells (ESCs) are closely associated with endometrial dysfunction induced by oxidative stress, which plays an important role in the pathological process of multiple gynecological and reproduction-related diseases. SCM-198, an alkaloid active component of Leonurus japonicas Houtt, has been reported to have anti-oxidative activity. However, the specific mechanisms of SCM-198 in the prevention of endometrial damage remain unknown. In the present study, we assessed the effect of SCM-198 on hydrogen peroxide (H2O2)-induced oxidative injury in ESCs. ESCs were pretreated with SCM-198 for 4 h and then challenged with H2O2. Morphology changes, apoptosis rate, and intracellular ROS production were measured to assess the level of oxidative injury. Flow cytometry and western blot analysis were performed to detect the expression levels of Bax, Bcl-2, active-caspase-3, and mitogen-activated protein kinases pathways. Classic inflammation cytokines were measured by real-time polymerase chain reactions. Our results showed that SCM-198 attenuated apoptosis and ROS generation of ESCs induced by H2O2. H2O2 induced the apparent apoptotic characteristics, including fragmentation of DNA, upregulation of Bax/Bcl2, activation of caspase-3, and secretion of inflammation cytokines, which were all ameliorated by SCM-198. Furthermore, H2O2-induced apoptosis-related ERK1/2 pathway activation was restrained by SCM-198 pretreatment. These findings suggested that SCM-198 could protect ESCs from oxidative injury, mainly by inhibiting oxidative stress and reducing apoptosis.


Subject(s)
Gallic Acid/analogs & derivatives , MAP Kinase Signaling System/drug effects , Oxidative Stress , Proto-Oncogene Proteins c-bcl-2/metabolism , Stromal Cells/drug effects , bcl-2-Associated X Protein/metabolism , Apoptosis/drug effects , Cell Survival/drug effects , Cells, Cultured , Gallic Acid/pharmacology , Humans , Hydrogen Peroxide/pharmacology , Leonurus/chemistry , Models, Biological , Oxidants/pharmacology , Protective Agents/pharmacology , Stromal Cells/cytology , Stromal Cells/metabolism
20.
Article in English | MEDLINE | ID: mdl-29050963

ABSTRACT

Endometriosis is a disease characterized by regurgitated lesions which are invasive and migratory, embedding at ectopic, extra-uterine locations. Extracellular glucosylceramides (GlcCers), bioactive sphingolipids potentiating signals for cell migration, are found in elevated levels in endometriosis; however underlying mechanisms that result in cellular migration are poorly defined. Here, we demonstrated that internalized GlcCer induced migratory activity in immortalized human endometrial stromal cells (HESCs), with highest potency observed in long-chain GlcCer. Long-chain ceramide (Cer) similarly induced cellular migration and mass spectrometry results revealed that the migratory behavior was contributed through glycosylation of ceramides. Cells treated with GlcCer synthase inhibitor, or RNAi-mediated knockdown of glucosylceramide synthase (GCS), the enzyme catalyzing GlcCer production attenuated cell motility. Mechanistic studies showed that GlcCer acts through stromal cell-derived factor-1 alpha and its receptor, CXC chemokine receptor 4 (SDF-1α-CXCR4) signaling axis and is dependent on phosphorylation of LYN kinase at Tyr396, and dephosphorylation of Tyr507. Migration was prominently attenuated in cells exposed to CXCR4 antagonist, AMD3100, yet can be rescued with diprotin A, which prevents the degradation of SDF-1α. Furthermore, blocking of LYN kinase activity in the presence of SDF-1α and GlcCer reduced HESC migration, suggesting that LYN acts downstream of GlcCer-SDF-1α-CXCR4 axis as part of its intracellular signal transduction. Our results reveal a novel role of long-chain GlcCer and the dialog between GlcCer, LYNpTyr396 and SDF-1α-CXCR4 in inducing HESC migration. This finding may improve our understanding how endometriotic lesions invade to their ectopic sites, and the possibility of using GlcCer to modulate the SDF-1α-CXCR4-LYNpTyr396 axis in endometriosis.


Subject(s)
Cell Movement/drug effects , Endometrium/drug effects , Endometrium/physiology , Glucosylceramides/pharmacology , src-Family Kinases/physiology , Cell Movement/genetics , Cells, Cultured , Endometrium/cytology , Female , Glucosylceramides/chemistry , Glucosylceramides/metabolism , Humans , Receptor Cross-Talk/drug effects , Receptor Cross-Talk/physiology , Signal Transduction/drug effects , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL