Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.367
Filtrar
1.
Aging (Albany NY) ; 16(7): 6273-6289, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38568100

RESUMO

OBJECTIVE: The purpose of this study was to explore the therapeutic characteristics of mesenchymal stem cells generated from human umbilical cord (hUC-MSCs) when utilized in conjunction with auto-crosslinked hyaluronic acid gel (HA-gel) for the management of intrauterine adhesion (IUA). The goal was to see how this novel therapy could enhance healing and improve outcomes for IUA patients. METHODS: In this study, models of intrauterine adhesion (IUA) were established in Sprague-Dawley (SD) rats, which were then organized and divided into hUC-MSCs groups. The groups involved: hUC-MSCs/HA-gel group, control group, and HA-gel group. Following treatment, the researchers examined the uterine cavities and performed detailed analyses of the endometrial tissues to determine the effectiveness of the interventions. RESULTS: The results indicated that in comparison with to the control group, both HA-gel, hUC-MSCs, and hUC-MSCs/HA-gel groups showed partial repair of IUA. However, in a more notable fashion transplantation of hUC-MSCs/HA-gel complex demonstrated significant dual repair effects. Significant outcomes were observed in the group treated with hUC-MSCs and HA-gel, they showed thicker endometrial layers, less fibrotic tissue, and a higher number of endometrial glands. This treatment strategy also resulted in a significant improvement in fertility restoration, indicating a profound therapeutic effect. CONCLUSIONS: The findings of this study suggest that both HA-gel, hUC-MSCs, and hUC-MSCs/HA-gel complexes have the potential for partial repair of IUA and fertility restoration caused by endometrium mechanical injury. Nonetheless, the transplantation of the hUC-MSCs/HA-gel complex displayed exceptional dual healing effects, combining effective anti-adhesive properties with endometrial regeneration stimuli.


Assuntos
Ácido Hialurônico , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Ratos Sprague-Dawley , Cordão Umbilical , Doenças Uterinas , Ácido Hialurônico/farmacologia , Ácido Hialurônico/química , Feminino , Animais , Transplante de Células-Tronco Mesenquimais/métodos , Humanos , Ratos , Aderências Teciduais , Cordão Umbilical/citologia , Doenças Uterinas/terapia , Géis , Endométrio/efeitos dos fármacos , Endométrio/citologia , Modelos Animais de Doenças
2.
Am J Reprod Immunol ; 91(4): e13842, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38650366

RESUMO

PROBLEM: Although endometrial receptivity is a key factor in influencing implantation in both naturally conceived and assisted reproductive technology (ART) cycles, very little is known about the endometrium milieu around the time of implantation. Previous studies have demonstrated the presence of several cytokines in the endometrium that affect implantation. However, there is lacking data about the presence of immune cell subtypes within the endometrium and in the uterine cavity at the time of implantation. METHOD OF STUDY: This study was approved by the Institutional Review Board (# 225589). The study was designed as a prospective observational cohort study between May 2021 and December 2022 at a single academic-based fertility center. All patients underwent at least one In Vitro Fertilization (IVF) cycle and have frozen embryos. Twenty-four participants were recruited for this study which was conducted during the frozen embryo transfer (FET) cycle regardless of the outcome of previous cycles. Two samples were acquired from each subject, denoted as lower and upper. A trial transfer catheter was introduced under ultrasound guidance into the lower uterine segment. Upon removal, the tip was rinsed in IMDM medium containing 10% FBS (lower uterus). A transfer catheter was then loaded with the embryo that was placed in the upper uterus under ultrasound guidance. The tip of the transfer catheter was rinsed in separate aliquot of the above media (upper uterus). After centrifugation, pelleted cells were stained for the following surface markers: CD45, CD3, CD19, CD4, CD8, gamma delta TCR, CD25, CD127, CD66b, CD14, CD16, CD56 and acquired on Sony SP6800 Spectral Analyzer. RESULTS: Upon staining the pelleted cells, we were able to identify viable leukocytes from samples obtained from both, upper and lower uterus (0.125 × 106 cells ± SD 0.32), (0.123 × 106 cells ± SD 0.12), respectively. Among total viable cells, there was no significant difference in both percent and number of CD45+ cells between the upper and lower uterus (9.88% ± 6.98 SD, 13.67% ± 9.79 SD, p = .198) respectively. However, there was significantly higher expression of CD3+ (p = .006), CD19+ (p = .032) and CD14+ (p = .019) cells in samples collected from upper compared to lower uterus. Within all CD3+ cells, we found that gamma delta T cells (GDT) were the major population of T cells in both upper and lower uterus. In contrast, CD8+ T cells were significantly higher in the lower uterus when compared to the upper uterus (p = .009). There was no statistically significant difference in the expression of CD4+ T cells, T regulatory cells (CD4+CD25+CD127-), NK cells (CD56+), neutrophils (CD66b+) and FcγRIII+ cells (CD16+) between upper and lower uterus. CONCLUSIONS: We believe the immune milieu at the time of embryo transfer will affect implantation. Understanding the composition of immune cells will guide further research in identifying optimal immune milieus that favor implantation. Comprehensive analysis of endometrium is expected to lead to new diagnostic and therapeutic approaches to improve IVF outcomes.


Assuntos
Transferência Embrionária , Endométrio , Útero , Humanos , Feminino , Adulto , Transferência Embrionária/métodos , Útero/imunologia , Endométrio/imunologia , Endométrio/citologia , Estudos Prospectivos , Implantação do Embrião/imunologia , Fertilização In Vitro , Gravidez , Líquidos Corporais/imunologia
3.
Reproduction ; 166(2): 161-174, 2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-37252830

RESUMO

In brief: Endometrial stromal cell motility is fundamental to regeneration and repair of this tissue and crucial for successful reproduction. This paper shows a role for the mesenchymal stem cell (MSC) secretome in enhancing endometrial stromal cell motility. Abstract: Cyclic regeneration and repair of the endometrium are crucial for successful reproduction. Mesenchymal stem cells (MSCs) derived from bone marrow (BM-MSC) and umbilical cord (UC-MSC) facilitate tissue repair via their secretome, which contains growth factors and cytokines that promote wound healing. Despite the implication of MSCs in endometrial regeneration and repair, mechanisms remain unclear. This study tested the hypothesis that the BM-MSC and UC-MSC secretomes upregulate human endometrial stromal cell (HESC) proliferation, migration, and invasion and activate pathways to increase HESC motility. BM-MSCs were purchased from ATCC and cultured from the BM aspirate of three healthy female donors. UC-MSCs were cultured from umbilical cords of two healthy male term infants. Using indirect co-culture of MSCs and hTERT-immortalized HESCs via a transwell system, we demonstrated that co-culture of HESCs with BM-MSCs or UC-MSCs from all donors significantly increased HESC migration and invasion, whereas effects on HESC proliferation varied among BM-MSC and UC-MSC donors. Analysis of gene expression by mRNA sequencing and RT-qPCR showed that expression of CCL2 and HGF was upregulated in HESCs that had been cocultured with BM-MSCs or UC-MSCs. Validation studies revealed that exposure to recombinant CCL2 for 48 h significantly increased HESC migration and invasion. Increased HESC motility by the BM-MSC and UC-MSC secretome appears to be mediated in part by upregulated HESC CCL2 expression. Our data support the potential for leveraging MSC secretome as a novel cell-free therapy to treat disorders of endometrial regeneration.


Assuntos
Endométrio , Células-Tronco Mesenquimais , Secretoma , Células Estromais , Feminino , Humanos , Masculino , Diferenciação Celular , Movimento Celular/genética , Movimento Celular/fisiologia , Proliferação de Células , Técnicas de Cocultura , Endométrio/citologia , Endométrio/metabolismo , Células Epiteliais , Expressão Gênica , Células-Tronco Mesenquimais/metabolismo , Secretoma/metabolismo , Células Estromais/metabolismo , Células Estromais/fisiologia , Regulação para Cima , Células da Medula Óssea/fisiologia , Cordão Umbilical/citologia , Cordão Umbilical/fisiologia
4.
Stem Cells ; 41(2): 153-168, 2023 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-36573461

RESUMO

Mesenchymal stem cells (MSCs) have been demonstrated to protect against fatty liver diseases, but the mechanism is still not clear. Menstrual blood-derived endometrial stem cells (MenSCs) are a substantial population of MSCs that can be obtained in a noninvasive manner. In the present study, we investigated the therapeutic effects and underlying mechanisms of MenSC transplantation in mouse models of diet-induced nonalcoholic fatty liver disease (NAFLD). The results revealed that MenSCs markedly promoted hepatic glycogen storage and attenuated lipid accumulation after transplantation. We further identified Rnf186 as a novel regulator involved in MenSC-based therapy for NAFLD mice. Rnf186 deficiency substantially inhibited high-fat diet-induced insulin resistance and abnormal hepatic glucose and lipid metabolism in mice. Mechanistically, Rnf186 regulated glucose and lipid metabolism through the AMPK-mTOR pathway. More importantly, hepatocyte growth factor (HGF) is identified as the key functional cytokine secreted by MenSCs and decreases the expression of hepatic Rnf186. HGF deficient MenSCs cannot attenuate glucose and lipid accumulation after transplantation in NAFLD mice. Collectively, our results provide preliminary evidence for the protective roles of HGF secreted by MenSCs in fatty liver diseases through downregulation of hepatic Rnf186 and suggest that MenSCs or Rnf186 may be an alternative therapeutic approach/target for the treatment of NAFLD.


Assuntos
Endométrio , Fator de Crescimento de Hepatócito , Células-Tronco Mesenquimais , Hepatopatia Gordurosa não Alcoólica , Animais , Camundongos , Proliferação de Células , Regulação para Baixo , Glucose/metabolismo , Fator de Crescimento de Hepatócito/genética , Fator de Crescimento de Hepatócito/metabolismo , Lipídeos , Fígado/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/terapia , Menstruação/sangue , Menstruação/genética , Menstruação/metabolismo , Endométrio/citologia , Endométrio/metabolismo
5.
F S Sci ; 3(3): 255-268, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35717521

RESUMO

OBJECTIVE: To determine whether bone marrow (BM)-derived cells engrafting the murine endometrium express the glucocorticoid receptor (GR) and androgen receptor (AR). Recent data demonstrate that BM is a long-term source of multiple hematopoietic and nonhematopoietic endometrial cell types. Important roles for glucocorticoids and androgens in regulating endometrial functions, including decidualization and early embryo attachment/invasion, have very recently emerged. Whether endometrial cells of BM origin express glucocorticoid or ARs has not been previously studied. DESIGN: Animal study. SETTING: Basic science laboratory. ANIMAL(S): Wild-type C57BL/6J male mice expressing enhanced green fluorescent protein (GFP) and syngeneic wild-type C57BL/6J female mice aged 6-9 weeks. INTERVENTION(S): Murine bone marrow transplant. MAIN OUTCOME MEASURE(S): Bone marrow cells were harvested from adult wild-type C57BL/6 mice and subjected to flow cytometry to identify the percentage of hematopoietic and nonhematopoietic cells expressing GR or AR. Uterine tissue sections from lethally irradiated syngeneic adult female C57BL/6 mice that had been recipients of BM transplants from adult male transgenic donor mice ubiquitously expressing GFP were studied. Immunohistochemistry was performed in the uterine tissue sections of the recipient mice at 5, 9, and 12 months after transplant using specific anti-GR, anti-AR, anti-GFP, anti-CD45 (pan leukocyte marker), and anti-F4/80 (murine macrophage marker) primary antibodies. Confocal laser microscopy was used to localize and quantitate BM-derived (GFP+) cell types in the endometrial stromal and epithelial compartments and determine whether BM-derived cell types in the murine endometrium express GR or AR. RESULT(S): Hematopoietic cells comprised 93.6%-96.6% of all cells in the BM, of which 98.1% ± 0.2% expressed GR and 92.2% ± 4.4% expressed AR. Nonhematopoietic cells comprised 0.4%-1.3% of BM, of which 52.8% ± 5.9% expressed GR and 48.9% ± 3.4% expressed AR. After BM transplant, the proportion of cells originating from BM in the endometrial stromal compartment increased over time, reaching 13.5% ± 2.3% at 12 months after transplant. In the epithelial compartments, <1% of the cells were of BM origin at 12 months after transplant. Most (60%-72%) GR+ and/or AR+ BM-derived cells in the stroma were hematopoietic (CD45+) cells, of which 37%-51% were macrophages. Nonetheless, 28%-33% of GR+ cells, and 28%-40% of AR+ BM-derived cells, were nonhematopoietic (CD45-) stromal cells of BM origin. CONCLUSION(S): A substantial number of BM-derived cells express GR and AR, suggesting a role for these cells in both glucocorticoid-regulated and androgen-regulated endometrial functions, such as proliferation and/or decidualization.


Assuntos
Endométrio , Células-Tronco Hematopoéticas , Receptores Androgênicos , Receptores de Glucocorticoides , Animais , Medula Óssea , Endométrio/citologia , Feminino , Células-Tronco Hematopoéticas/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores Androgênicos/genética , Receptores de Glucocorticoides/genética
6.
Biomed Res Int ; 2022: 7663379, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35378789

RESUMO

EMs is a kind of benign disease with certain malignant behaviors. The adhesion, invasive growth, and angiogenesis of ectopic endometrial cells are the pathological basis of EMs occurrence, but its etiology and pathogenesis have not been completely illustrated yet. In our research, we aim to investigate the role of miR-363 in the pathogenesis of endometriosis. Real-time quantitative PCR was used to detect the expression of miR-363 before and after ESC/NSC transfection. CCK-8, flow cytometry, and transwell assay were used to detect the effect of the miR-363 expression on cell proliferation, apoptosis, and invasion. The effects of the miR-363 expression on the contents of Fas/APO-1 and ICAM-1 in cell culture supernatant were detected by ELISA. qRT-PCR and WB assay were used to detect the effects of the miR-363 expression on the mRNA and protein expression levels of ICAM-1, MMP-7, and VEGF in ESC. The increased expression of miR-363 could inhibit the proliferation and invasion of ESC, promote apoptosis, and inhibit the secretion of FAS/APO-1 and ICAM-1. The knockdown expression of miR-363 promoted proliferation and invasion of NSC, inhibited apoptosis, and promoted secretion of FAS/APO-1 and ICAM-1. VCAM-1, VEGF, and MMP-7 were detected in ESCs before transfection. The protein expression level was higher than that of NSCs. Compared with pretransfection, the protein levels of VCAM-1, VEGF, and MMP-7 in the M-363 group were significantly downregulated. The downregulated expression of miR-363 was associated with a stronger cell proliferation ability, a lower cell apoptosis rate, and a stronger ESC. Migration is associated with invasiveness, proliferation, angiogenesis, and immune escape. The low expression of miR-363 promotes endogenesis through posttranscriptional regulation of target genes VCAM-1, MMP-7, and VEGF. The differential expression of miR-363 between ESC and NSC may be an important factor in the many biological differences between ESC and NSC.


Assuntos
Endometriose , Células Epiteliais , MicroRNAs , Movimento Celular/genética , Proliferação de Células/genética , Endometriose/patologia , Endométrio/citologia , Endométrio/patologia , Células Epiteliais/metabolismo , Feminino , Humanos , MicroRNAs/metabolismo , Células Estromais/patologia
7.
Reprod Biol Endocrinol ; 20(1): 29, 2022 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-35120552

RESUMO

BACKGROUND: Several surveys have reported that patients treated with gonadotropin-releasing hormone antagonist (GnRH-ant) protocol showed a significantly lower rate of implantation and clinical pregnancy compared to GnRH agonist (GnRH-a) protocol during in vitro fertilization-fresh embryo transfer. Subsequent studies imputed this poor outcome to the negative effects of GnRH-ant on endometrial receptive. However, the mechanisms were not fully understood. METHODS: The clinical data of 2815 patients undergoing fresh embryo transfer in our center were analyzed. Human endometrial stromal cells (ESCs) from healthy women undergoing elective pregnancy termination of a normal pregnancy at 8-10 weeks gestation were treated with GnRH-analogs or imatinib (c-kit receptor inhibitor). CCK8 and Flow cytometry were used to investigated the growth ability of ESCs. Immunofluorescence staining and western blot was used to detected the target proteins. RESULTS: The clinical data showed that the endometrial thickness on HCG Day were significantly lower in GnRH-ant group. Although no difference of embryo quality in these two groups, GnRH-ant group showed remarkably decreased rate of HCG positive, embryo implantation and pregnancy. Moreover, GnRH-ant significantly reduced the proliferation and induced the apoptosis of ESCs. Furthermore, the expression and activation of c-kit receptor, which played pivotal roles during embryo implantation, were observably decreased by GnRH-ant. Inhibiting the activation of c-kit by imatinib remarkably suppressed the proliferation and promoted the apoptosis of ESCs. Additionally, the phosphorylation of AKT and expression of Cyclin D1, which were closely related with cellular growth, were distinctly lessened after treating with imatinib. CONCLUSIONS: In summary, our study showed that GnRH-ant weakened the activization of c-kit receptor by decreasing its expression, causing the impaired growth ability of ESCs. Our findings provided a new insight into the effects of GnRH-ant on endometrium.


Assuntos
Endométrio/efeitos dos fármacos , Antagonistas de Hormônios/farmacologia , Células Estromais/efeitos dos fármacos , Adulto , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Células Cultivadas , Implantação do Embrião/efeitos dos fármacos , Implantação do Embrião/fisiologia , Transferência Embrionária , Endométrio/citologia , Feminino , Fertilização In Vitro/métodos , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Humanos , Recém-Nascido , Masculino , Indução da Ovulação/efeitos adversos , Indução da Ovulação/métodos , Gravidez , Cultura Primária de Células , Proteínas Proto-Oncogênicas c-kit/genética , Proteínas Proto-Oncogênicas c-kit/metabolismo , Estudos Retrospectivos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Células Estromais/fisiologia
8.
BMC Cancer ; 22(1): 184, 2022 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-35177036

RESUMO

BACKGROUND: S100A1 expression is deregulated in a variety of human malignancies, but its role in normal and malignant endometrial cells is unclear. METHODS: We used endometrial carcinoma (Em Ca) cell lines to evaluate the physical and functional interaction of S100A1 with p53 and its negative regulator, mouse double minute 2 (MDM2). We also evaluated the expression of S100A1, p53, and MDM2 in clinical samples consisting of 89 normal endometrial and 189 Em Ca tissues. RESULTS: S100A1 interacted with MDM2 but not p53 in Em Ca cell lines. Treatment of cells stably overexpressing S100A1 with Nutlin-3A, an inhibitor of the p53/MDM2 interaction, increased expression of p53-target genes including p21waf1 and BAX. S100A1 overexpression enhanced cellular migration, but also sensitized cells to the antiproliferative and proapoptotic effects of Adriamycin, a genotoxic agent; these phenotypes were abrogated when S100A1 was knocked down using shRNA. In clinical samples from normal endometrium, S100A1 expression was significantly higher in endometrial glandular cells of the middle/late secretory and menstrual stages when compared to cells in the proliferative phases; high S100A1 was also positively correlated with expression of MDM2 and p21waf1 and apoptotic status, and inversely correlated with Ki-67 scores. However, such correlations were absent in Em Ca tissues. CONCLUSION: The interaction between S100A1 and MDM2 may modulate proliferation, susceptibility to apoptosis, and migration through alterations in p53 signaling in normal- but not malignant-endometrial cells.


Assuntos
Neoplasias do Endométrio/genética , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteínas S100/metabolismo , Transdução de Sinais/genética , Proteína Supressora de Tumor p53/metabolismo , Animais , Apoptose , Linhagem Celular Tumoral , Proliferação de Células/genética , Endométrio/citologia , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Camundongos
9.
Sci Rep ; 12(1): 2618, 2022 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-35173188

RESUMO

Endometriosis (EMs) is one of the most frequent diseases of reproductive-age women and is characterized by the growth of endometrial tissues beyond the uterus. The enhanced proliferative and migratory potential of endometrial stromal cells (ESCs) plays an important role in the progression of EMs. Mounting studies have demonstrated that long noncoding RNAs (lncRNAs) exert an important role in regulating the development and progression of EMs. Given the aberrant expression of lncRNA ADAMTS9-AS1 in ectopic endometrium (ecEM), we investigated the biological effect of ADAMTS9-AS1 on ESC proliferation and migration and explored the underlying mechanism. The current data showed that ADAMTS9-AS1 expression was significantly upregulated in ecEM compared with eutopic endometrium (euEM) in patients with EMs and in a murine model of EMs. Functionally, ADAMTS9-AS1 knockdown in ectopic ESCs (EESCs) decreased cell viability and migration, whereas ADAMTS9-AS1 overexpression in normal ESCs (NESCs) enhanced cell viability and migration. More importantly, the effect of ADAMTS9-AS1 inhibition on decreasing ESC viability was significantly blocked by ferrostatin-1 (Fer-1, a ferroptosis inhibitor), and ADAMTS9-AS1 overexpression repressed erastin (a ferroptosis activator)-induced cell death. Furthermore, the regulatory role of ADAMTS9-AS1 in ferroptosis was defined and evidenced by increased reactive oxygen species (ROS) levels and malonyl dialdehyde (MDA) content and decreased expression of glutathione peroxidase 4 (GPX4) after ADAMTS9-AS1 inhibition. Mechanistically, ADAMTS9-AS1 functioned as a competing endogenous RNA (ceRNA) by sponging miR-6516-5p to derepress the expression of GPX4, the critical repressor of ferroptosis. Taken together, these results demonstrate that upregulated ADAMTS9-AS1 accelerates ESC proliferation and migration by regulating miR-6516-5p/GPX4-dependent ferroptosis and may be a potential target for the treatment of EMs.


Assuntos
Proteína ADAMTS9/fisiologia , Endometriose/genética , Endometriose/fisiopatologia , Endométrio/citologia , Endométrio/fisiologia , Ferroptose/genética , Ferroptose/fisiologia , Expressão Gênica/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/genética , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/metabolismo , RNA Longo não Codificante/fisiologia , Células Estromais/fisiologia , Animais , Movimento Celular/genética , Proliferação de Células/genética , Células Cultivadas , Modelos Animais de Doenças , Endometriose/patologia , Endometriose/terapia , Feminino , Humanos , Camundongos Endogâmicos BALB C , Terapia de Alvo Molecular
10.
Int J Mol Sci ; 23(3)2022 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-35163159

RESUMO

BACKGROUND: Gestagens are the most widely used therapy in anestrus type II. The aim of this research is to evaluate the effectiveness of the vaginal progesterone inserts therapy in anestrus type II in cows. METHODS: The study was conducted on 33 cows. Progesterone (PR) and estrogen (ER) receptors expression in endometrium was assessed on a molecular level based on mRNA tissue expression. Additionally, blood 17ß-estradiol and progesterone levels were evaluated. RESULTS: A decrease in mRNA expression of A and B PR and ER α was noted in treated and untreated animals. In the treated group, an increase of ERß mRNA expression was observed, while a decreased was found in untreated animals. There was increased PR, ERα and ß expression in endometrial tissue in treated cows, and decreased expression of these factors in untreated cows. In the treated group, recurrence of ovarian cyclicity was noted in 52% of animals and pregnancy was obtained in 34.8% of them, while in the untreated group, recurrence did not occur. In the control group, spontaneous recurrence of ovarian cyclicity was not observed. An increase of PR expression was correlated with increased proliferation of endometrial cells. CONCLUSIONS: It seems likely that the endometrium is well developed and ready for placentation after removing the exogenous source of progesterone and preventing the recurrence of cyclicity of ovaries.


Assuntos
Anestro , Endométrio/citologia , Estradiol/administração & dosagem , Regulação da Expressão Gênica/efeitos dos fármacos , Progesterona/administração & dosagem , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo , Administração Intravaginal , Animais , Bovinos , Endométrio/efeitos dos fármacos , Endométrio/metabolismo , Estradiol/sangue , Estrogênios/administração & dosagem , Estrogênios/sangue , Feminino , Progesterona/sangue , Progestinas/administração & dosagem , Progestinas/sangue
11.
Int J Mol Sci ; 23(4)2022 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-35216052

RESUMO

The cyclic regeneration of human endometrium is guaranteed by the proliferative capacity of endometrial mesenchymal stromal cells (E-MSCs). Due to this, the autologous infusion of E-MSCs has been proposed to support endometrial growth in a wide range of gynecological diseases. We aimed to compare two different endometrial sampling methods, surgical curettage and vacuum aspiration biopsy random assay (VABRA), and to validate a novel xeno-free method to culture human E-MSCs. Six E-MSCs cell samples were isolated after mechanical tissue homogenization and cultured using human platelet lysate. E-MSCs were characterized for the colony formation capacity, proliferative potential, and multilineage differentiation. The expression of mesenchymal and stemness markers were tested by FACS analysis and real-time PCR, respectively. Chromosomal alterations were evaluated by karyotype analysis, whereas tumorigenic capacity and invasiveness were tested by soft agar assay. Both endometrial sampling techniques allowed efficient isolation and expansion of E-MSCs using a xeno-free method, preserving their mesenchymal and stemness phenotype, proliferative potential, and limited multi-lineage differentiation ability during the culture. No chromosomal alterations and invasive/tumorigenic capacity were observed. Herein, we report the first evidence of efficient E-MSCs isolation and culture in Good Manufacturing Practice compliance conditions, suggesting VABRA endometrial sampling as alternative to surgical curettage.


Assuntos
Diferenciação Celular/fisiologia , Endométrio/citologia , Células-Tronco Mesenquimais/citologia , Adulto , Biomarcadores/metabolismo , Células da Medula Óssea/citologia , Técnicas de Cultura de Células/métodos , Proliferação de Células/fisiologia , Células Cultivadas , Endométrio/metabolismo , Feminino , Humanos , Adulto Jovem
12.
Molecules ; 27(3)2022 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-35164046

RESUMO

Endometriosis presents high prevalence and its physiopathology involves hyperactivation of endometrial and vaginal cells, especially by bacteria. The disease has no cure and therapies aiming to inhibit its development are highly desirable. Therefore, this study investigated whether MiodesinTM (10 µg/mL = IC80; 200 µg/mL = IC50), a natural compound constituted by Uncaria tomentosa, Endopleura uchi, and astaxanthin, could exert anti-inflammatory and anti-proliferative effects against Lipopolysaccharides (LPS) stimulation in endometrial and Candida albicans vaginal cell lines. VK2 E6/E7 (vaginal) and KLE (epithelial) cell lines were stimulated with Candida albicans (1 × 107 to 5 × 107/mL) and LPS (1 µg/mL), respectively. MiodesinTM inhibited mRNA expression for Nuclear factor kappa B (NF-κB), ciclo-oxigenase 1 (COX-1), and phospholipase A2 (PLA2), beyond the C-C motif chemokine ligand 2 (CCL2), CCL3, and CCL5 in VK2 E6/E7 cells (p < 0.05). In addition, the inhibitory effects of both doses of MiodesinTM (10 µg/mL and 200 µg/mL) resulted in reduced secretion of interleukin-1ß (IL-1ß), IL-6, IL-8, tumor necrosis factor α (TNF-α) (24 h, 48 h, and 72 h) and CCL2, CCL3, and CLL5 (p < 0.05) by VK2 E6/E7 cells. In the same way, COX-1 MiodesinTM inhibited LPS-induced hyperactivation of KLE cells, as demonstrated by reduced secretion of IL-1ß, IL-6, IL-8, TNF-α (24 h, 48 h, and 72 h) and CCL2, CCL3, and CLL5 (p < 0.05). Furthermore, MiodesinTM also inhibited mRNA expression and secretion of matrix metalloproteinase-2 (MMP-2), MMP-9, and vascular endothelial growth factor (VEGF), which are key regulators of invasion of endometrial cells. Thus, the study concludes that MiodesinTM presents beneficial effects in the context of endometriosis, positively affecting the inflammatory and proliferative response.


Assuntos
Produtos Biológicos/farmacologia , Endométrio/imunologia , Vagina/imunologia , Candida albicans/fisiologia , Quimiocinas/metabolismo , Citocinas/metabolismo , Endométrio/citologia , Feminino , Humanos , Lipopolissacarídeos/farmacologia , Fosfolipases A2/metabolismo , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/genética , Vagina/citologia , Vagina/microbiologia
13.
Gene ; 822: 146337, 2022 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-35182676

RESUMO

The extracellular vesicles (EVs) in uterine fluids play a vital role in embryo implantation by mediating intrauterine communication between conceptus and maternal endometrium in pigs. However, the regulatory mechanism of EVs in uterine fluids is largely unclear. In order to understand the effect of EVs in uterine flushing fluids (UFs) during embryo implantation on endometrial epithelial cells (EECs) and embryonic trophoblast cells (PTr2 cells). The UFs-EVs on day 13 of pregnancy (D13) were added to the culture medium of EECs and PTr2 cells. It was found that PKH-67 labeled UFs-EVs could be taken up in EECs and PTr2 cells. Transcriptome sequencing analysis showed that a total of 1793 and 6279 genes were differentially expressed in the EECs and PTr2 cells after the treatment of UFs-EVs on D13, respectively. Among these genes, real-time quantitative PCR (RT-qPCR) results indicated that ID2, ITGA5, CXCL10 and CXCL11 genes were differentially expressed in both EECs and PTr2 cells after treatment. Bioinformatics analysis showed that the differentially expressed (DE) genes in EECs and PTr2 cells after treatment are involved in immune regulation, cell migration, cell adhesion and the secretion and uptake of EVs. Our research offers novel insight into the regulation mechanism of UFs-EVs on D13 in EECs and PTr2 cells.


Assuntos
Endométrio/citologia , Vesículas Extracelulares/transplante , Perfilação da Expressão Gênica/veterinária , Redes Reguladoras de Genes , Trofoblastos/citologia , Animais , Adesão Celular , Técnicas de Cultura de Células , Movimento Celular , Células Cultivadas , Implantação do Embrião , Endométrio/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Feminino , Regulação da Expressão Gênica , Gravidez , Análise de Sequência de RNA , Suínos , Trofoblastos/metabolismo
14.
Reprod Sci ; 29(3): 723-733, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34981457

RESUMO

The cause of endometriosis, which is characterized by the existence of functional endometrial tissue outside the uterine cavity, is poorly understood. Seminal plasma (SP) is rich in multiple cytokines that may promote endometrial tissue survival. Here, we evaluated the effect of SP on growth of endometrial mesenchymal stem cells (MSCs) from women with endometriosis (E-MSCs) and women without endometriosis (NE-MSCs). Proliferation, cell foci formation, cell cycle progression, and growth marker expression of E- and NE-MSCs were promoted by SP. These effects may be mediated through activation of transforming growth factor beta 1 (TGF-ß1), Akt, and p42/44 signaling, which enhances CDK2 and CDK6 expression and accelerates cell cycle progression. Xenografts exposed to SP exhibited a three-fold increase in volume and four-fold increase in weight after 14 days. Our findings demonstrate that TGF-ß1 in SP may promote endometrial tissue survival which will allow us to understand the pathogenesis and develop novel approaches for prevention and therapies of endometriosis.


Assuntos
Endometriose/metabolismo , Endométrio/citologia , Células-Tronco Mesenquimais/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Sêmen/química , Fator de Crescimento Transformador beta1/metabolismo , Adulto , Animais , Biomarcadores/metabolismo , Ciclo Celular/fisiologia , Proliferação de Células , Células Cultivadas , Citocinas/metabolismo , Endometriose/patologia , Feminino , Humanos , Camundongos , Transdução de Sinais
15.
Reprod Biol Endocrinol ; 20(1): 3, 2022 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-34980157

RESUMO

BACKGROUND: Successful implantation is dependent on coordination between maternal endometrium and embryo, and the role of EVs in the required cross-talk cell-to-cell has been recently established. In this regard, it has been reported that EVs secreted by the maternal endometrium can be internalized by human trophoblastic cells transferring their contents and enhancing their adhesive and invasive capacity. This is the first study to comprehensively evaluate three EV isolation methods on human endometrial epithelial cells in culture and to describe the proteomic content of EVs secreted by pHEECs from fertile women. METHODS: Ishikawa cells and pHEECs were in vitro cultured and hormonally treated; subsequently, conditioned medium was collected and EVs isolated. Ishikawa cells were used for the comparison of EVs isolation methods ultracentrifugation, ExoQuick-TC and Norgen Cell Culture Media Exosome Purification Kit (n = 3 replicates/isolation method). pHEECs were isolated from endometrial biopsies (n = 8/replicate; 3 replicates) collected from healthy oocyte donors with confirmed fertility, and protein content of EVs isolated by the most efficient methodology was analysed using liquid chromatography-tandem mass spectrometry. EV concentration and size were analyzed by nanoparticle tracking analysis, EV morphology visualized by transmission electron microscopy and protein marker expression was determined by Western blotting. RESULTS: Ultracentrifugation was the most efficient methodology for EV isolation from medium of endometrial epithelial cells. EVs secreted by pHEECs and isolated by ultracentrifugation were heterogeneous in size and expressed EV protein markers HSP70, TSG101, CD9, and CD81. Proteomic analysis identified 218 proteins contained in these EVs enriched in biological processes involved in embryo implantation, including cell adhesion, differentiation, communication, migration, extracellular matrix organization, vasculature development, and reproductive processes. From these proteins, 82 were selected based on their functional relevance in implantation success as possible implantation biomarkers. CONCLUSIONS: EV protein cargos are implicated in biological processes related to endometrial receptivity, embryo implantation, and early embryo development, supporting the concept of a communication system between the embryo and the maternal endometrium via EVs. Identified proteins may define new biomarkers of endometrial receptivity and implantation success.


Assuntos
Implantação do Embrião/fisiologia , Endométrio/metabolismo , Vesículas Extracelulares/metabolismo , Proteoma/análise , Adolescente , Adulto , Células Cultivadas , Endométrio/citologia , Endométrio/patologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Vesículas Extracelulares/patologia , Feminino , Fertilidade/fisiologia , Humanos , Proteoma/metabolismo , Proteômica , Adulto Jovem
16.
Bioengineered ; 13(1): 800-809, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34964708

RESUMO

Hypoxia-induced damage in endometrial stromal cells (ESCs) is an important event in the pathological progression of Endometriosis. It is reported that significant inflammation is induced by hypoxia in ESCs, mediated by serval inflammatory progressions, pathways, or factors. Sitagliptin, an important member of the dipeptidyl peptidase-4 (DPP-4) inhibitors family and has been widely used for the management of type 2 diabetes. It has been recently reported to exert significant anti-inflammatory effects. Here, we aim to assess whether Sitagliptin possesses a protective effect against hypoxia-induced damages in ESCs. Our findings indicate that exposure to hypoxia significantly increased oxidative stress in ESCs by increasing the production of reactive oxygen species (ROS) and decreasing the levels of reduced glutathione (GSH), which was ameliorated by Sitagliptin. Additionally, the excessively produced inflammatory mediators, including tumor necrosis factor (TNF)-α, interleukin (IL)-6, monocyte chemoattractant protein-1 (MCP-1), cyclooxygenase-2 (COX-2), prostaglandin E2 (PGE2), and high mobility group box (HMGB)-1, in hypoxia-treated HESCs were pronouncedly repressed by Sitagliptin. The activated p38 mitogen-activated protein kinases (MAPK) pathway was observed in hypoxia-stimulated HESCs, then greatly inhibited by the introduction of Sitagliptin. Lastly, hypoxia-induced phosphorylation and degradation of IκBα, as well as the upregulation of nuclear factor kappa-B (NF-κB) p65 and increased transcriptional activity of NF-κB, were dramatically abolished by Sitagliptin. Collectively, Sitagliptin ameliorated hypoxia-induced damages in ESCs by suppressing the inflammation.


Assuntos
Anti-Inflamatórios/farmacologia , Endometriose/metabolismo , Endométrio/citologia , Fosfato de Sitagliptina/farmacologia , Hipóxia Celular/efeitos dos fármacos , Células Cultivadas , Progressão da Doença , Endometriose/tratamento farmacológico , Endométrio/efeitos dos fármacos , Endométrio/metabolismo , Feminino , Glutationa/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Células Estromais/citologia , Células Estromais/enzimologia , Células Estromais/metabolismo
17.
Mol Med Rep ; 25(2)2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34913072

RESUMO

Endometriosis (EM), the presence of functional endometrial glands and stroma outside the uterine cavity, is a common gynecological disorder. At present, the pathogenesis of EM has not been fully elucidated, so there is still a lack of effective therapy. The present study aimed to explore the role of C­C motif chemokine ligand 28 (CCL28) and its underlying mechanism in endometrial stromal cells to propose a novel therapy for EM treatment. The expression of CCL28 and CC chemokine receptor 10 (CCR10) were examined. After CCL28 knockdown or overexpression by lentivirus infection, cell proliferation and invasion were measured. It was revealed that compared with normal, the expression levels of CCL28 and CCR10 were significantly elevated in endometrial tissues of patients with EM. Knockdown of CCL28 in endometrial stromal cells significantly suppressed cell proliferation and invasion, and this was accompanied by significantly reduced expression levels of CCR10, MMP2, MMP9, integrin ß1 (ITGB1) and phosphorylated (p)­ERK/ERK ratio. The addition of the CCL28 recombinant protein had an opposite effect to CCL28 downregulation. Furthermore, the ERK inhibitor, PD98059, reduced CCL28­induced cell proliferation and invasion, as well as the expression levels of MMP2, MMP9, ITGB1 and p­ERK. Therefore, the present study indicated that CCL28 may contribute to the progression of EM by regulating MMP2, MMP9 and ITGB1 expression and function via the activation of the ERK signaling pathway.


Assuntos
Quimiocinas CC/metabolismo , Endometriose/patologia , Endométrio/patologia , Células Estromais/patologia , Adulto , Movimento Celular , Proliferação de Células , Células Cultivadas , Quimiocinas CC/genética , Endometriose/cirurgia , Endométrio/citologia , Endométrio/cirurgia , Feminino , Técnicas de Silenciamento de Genes , Humanos , Laparoscopia , Sistema de Sinalização das MAP Quinases , Pessoa de Meia-Idade , Cultura Primária de Células , Receptores CCR10/metabolismo
18.
Physiol Genomics ; 54(2): 71-85, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34890509

RESUMO

In cattle, starting 4-5 days after estrus, preimplantation embryonic development occurs in the confinement of the uterine lumen. Cells in the endometrial epithelial layer control the molecular traffic to and from the lumen and, thereby determine luminal composition. Starting early postestrus, endometrial function is regulated by sex steroids, but the effects of progesterone on luminal cells transcription have not been measured in vivo. The first objective was to determine the extent to which progesterone controls transcription in luminal epithelial cells 4 days (D4) after estrus. The second objective was to discover luminal transcripts that predict pregnancy outcomes when the effect of progesterone is controlled. Endometrial luminal epithelial cells were collected from embryo transfer recipients on D4 using a cytological brush and their transcriptome was determined by RNASeq. Pregnancy by embryo transfer was measured on D30 (25 pregnant and 18 nonpregnant). Progesterone concentration on D4 was associated positively (n = 182) and negatively (n = 58) with gene expression. Progesterone-modulated transcription indicated an increase in oxidative phosphorylation, biosynthetic activity, and proliferation of epithelial cells. When these effects of progesterone were controlled, different genes affected positively (n = 22) and negatively (n = 292) odds of pregnancy. These set of genes indicated that a receptive uterine environment was characterized by the inhibition of phosphoinositide signaling and innate immune system responses. A panel of 25 genes predicted the pregnancy outcome with sensitivity and specificity ranging from 64%-96% and 44%-83%, respectively. In conclusion, in the early diestrus, both progesterone-dependent and progesterone-independent mechanisms regulate luminal epithelial transcription associated with pregnancy outcomes in cattle.


Assuntos
Endométrio/metabolismo , Células Epiteliais/metabolismo , Progesterona/metabolismo , Transcriptoma/genética , Útero/metabolismo , Animais , Bovinos , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Análise por Conglomerados , Transferência Embrionária , Desenvolvimento Embrionário , Endométrio/citologia , Estro/genética , Feminino , Perfilação da Expressão Gênica/métodos , Gravidez , Progesterona/farmacologia , RNA-Seq/métodos , Transdução de Sinais/genética , Transcriptoma/efeitos dos fármacos , Útero/citologia
19.
Reprod Sci ; 29(1): 133-142, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34773204

RESUMO

Decidualization is a substantive differentiation process experienced by endometrium to prepare for pregnancy. During this process, the endometrial stromal cells are transformed to endometrial epithelial cells. The receptivity of endometrium is necessary for the decidualization and successful implantation of endometrium, while the main hormones coordinating this process are estrogen and progesterone (P). In our study, the immunofluorescence, qPCR, and western blot experiments were conducted on different types of clinical endometrial tissue samples. The experimental results show that in the endometrium of normal subjects during the luteal phase, the protein level and serum P4 level of the orphan nuclear receptor NR4A1 messenger RNA were all significantly higher than those of patients with endometriosis or primary infertility, and the two levels presented positive correlation. Through decidualization induction of the human endometrial stromal cells (hESCs) cultured in vitro and additional P treatment, the results of chromatin immunoprecipitation and other experiments show that the P treatment could upregulate the expression of NR4A1 in hESCs, and this process was mediated under the direct effect of progesterone receptor (PR) and NR4A1. When the NR4A1 in hESCs was silenced, the promotion of hESC proliferation by P was inhibited. P and overexpressed NR4A1 increased the expression of epithelial cell marker in decidual hESCs, and qPCR showed that NR4A1's response to P was earlier than that of the epithelial cell marker. The results of spheroid adhesion assay show that the silent NR4A1 had reduced the adhesion of decidual hESCs induced in vitro to embryo. To sum it up, NR4A1 participated in the decidualization process by responding to the P regulation via and by promoting the hESCs' mesenchymal-epithelial transition, so as to further influence the receptivity of endometrium.


Assuntos
Endométrio/metabolismo , Transição Epitelial-Mesenquimal/fisiologia , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Células Estromais/metabolismo , Adulto , Linhagem Celular , Endométrio/citologia , Feminino , Regulação da Expressão Gênica , Humanos , Progesterona/sangue , Células Estromais/citologia , Adulto Jovem
20.
Biol Reprod ; 106(1): 155-172, 2022 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-34591094

RESUMO

The decidua is a hallmark of reproduction in many placental mammals. Differentiation of decidual stromal cells is known to be induced by progesterone and the cyclic AMP/protein kinase A (cAMP/PKA) pathway. Several candidates have been identified as the physiological stimulus for adenylyl cyclase activation, but their relative importance remains unclear. To bypass this uncertainty, the standard approach for in vitro experiments uses membrane-permeable cAMP and progestin. We phylogenetically infer that prostaglandin E2 (PGE2) likely was the signal that ancestrally induced decidualization in conjunction with progesterone. This suggests that PGE2 and progestin should be able to activate the core gene regulatory network of decidual cells. To test this prediction, we performed a genome-wide study of gene expression in human endometrial fibroblasts decidualized with PGE2 and progestin. Comparison to a cAMP-based protocol revealed shared activation of core decidual genes and decreased induction of senescence-associated genes. Single-cell transcriptomics of PGE2-mediated decidualization revealed a distinct, early-activated state transitioning to a differentiated decidual state. PGE2-mediated decidualization was found to depend upon progestin-dependent induction of PGE2 receptor 2 (PTGER2) which in turn leads to PKA activation upon PGE2 stimulation. Progesterone-dependent induction of PTGER2 is absent in opossum, an outgroup taxon of placental mammals which is incapable of decidualization. Together, these findings suggest that the origin of decidualization involved the evolution of progesterone-dependent activation of the PGE2/PTGER2/PKA axis, facilitating entry into a PKA-dominant rather than AKT-dominant cellular state. We propose the use of PGE2 for in vitro decidualization as an alternative to 8-Br-cAMP.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Decídua/citologia , Dinoprostona/farmacologia , Linhagem Celular Transformada , Células Cultivadas , AMP Cíclico/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Decídua/fisiologia , Endométrio/citologia , Endométrio/metabolismo , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/fisiologia , Expressão Gênica , Estudo de Associação Genômica Ampla , Humanos , Acetato de Medroxiprogesterona/farmacologia , Gravidez , Receptores de Prostaglandina E Subtipo EP2/metabolismo , Análise de Sequência de RNA , Análise de Célula Única
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...