Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros








Intervalo de ano de publicação
1.
Front Immunol ; 15: 1378863, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38765018

RESUMO

Background: At menstruation, the functional layer of the human endometrium sheds off due to the trigger of the release of inflammatory factors, including interleukin 6 (IL-6), as a result of a sharp decline in progesterone levels, leading to tissue breakdown and bleeding. The endometrial mesenchymal stem-like cells (CD140b+CD146+ eMSC) located in the basalis are responsible for the cyclical regeneration of the endometrium after menstruation. Endometrial cells from the menstruation phase have been proven to secrete a higher amount of IL-6 and further enhance the self-renewal and clonogenic activity of eMSC. However, the IL-6-responsive mechanism remains unknown. Thus, we hypothesized that IL-6 secreted from niche cells during menstruation regulates the proliferation and self-renewal of eMSC through the WNT/ß-catenin signaling pathway. Methods: In this study, the content of IL-6 across the menstrual phases was first evaluated. Coexpression of stem cell markers (CD140b and CD146) with interleukin 6 receptor (IL-6R) was confirmed by immunofluorescent staining. In vitro functional assays were conducted to investigate the effect of IL-6 on the cell activities of eMSC, and the therapeutic role of these IL-6- and WNT5A-pretreated eMSC on the repair of injured endometrium was observed using an established mouse model. Results: The endometrial cells secrete a high amount of IL-6 under hypoxic conditions, which mimic the physiological microenvironment in the menstruation phase. Also, the expression of IL-6 receptors was confirmed in our eMSC, indicating their capacity to respond to IL-6 in the microenvironment. Exogenous IL-6 can significantly enhance the self-renewal, proliferation, and migrating capacity of eMSC. Activation of the WNT/ß-catenin signaling pathway was observed upon IL-6 treatment, while suppression of the WNT/ß-catenin signaling impaired the stimulatory role of IL-6 on eMSC activities. IL-6- and WNT5A-pretreated eMSC showed better performance during the regeneration of the injured mouse endometrium. Conclusion: We demonstrate that the high level of IL-6 produced by endometrial cells at menstruation can induce the stem cells in the human endometrium to proliferate and migrate through the activation of the WNT/ß-catenin pathway. Treatment of eMSC with IL-6 and WNT5A might enhance their therapeutic potential in the regeneration of injured endometrium.


Assuntos
Proliferação de Células , Endométrio , Interleucina-6 , Menstruação , Células-Tronco Mesenquimais , Via de Sinalização Wnt , Feminino , Células-Tronco Mesenquimais/metabolismo , Humanos , Interleucina-6/metabolismo , Endométrio/metabolismo , Endométrio/citologia , Animais , Camundongos , Adulto , Células Cultivadas , Autorrenovação Celular
2.
Biol Res ; 57(1): 6, 2024 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-38347646

RESUMO

BACKGROUND: The monthly regeneration of human endometrial tissue is maintained by the presence of human endometrial mesenchymal stromal/stem cells (eMSC), a cell population co-expressing the perivascular markers CD140b and CD146. Endometrial regeneration is impaired in the presence of intrauterine adhesions, leading to infertility, recurrent pregnancy loss and placental abnormalities. Several types of somatic stem cells have been used to repair the damaged endometrium in animal models, reporting successful pregnancy. However, the ability of endometrial stem cells to repair the damaged endometrium remains unknown. METHODS: Electrocoagulation was applied to the left uterine horn of NOD/SCID mice causing endometrial injury. Human eMSC or PBS was then injected into the left injured horn while the right normal horn served as controls. Mice were sacrificed at different timepoints (Day 3, 7 and 14) and the endometrial morphological changes as well as the degree of endometrial injury and repair were observed by histological staining. Gene expression of various inflammatory markers was assessed using qPCR. The functionality of the repaired endometrium was evaluated by fertility test. RESULTS: Human eMSC successfully incorporated into the injured uterine horn, which displayed significant morphological restoration. Also, endometrium in the eMSC group showed better cell proliferation and glands formation than the PBS group. Although the number of blood vessels were similar between the two groups, gene expression of VEGF-α significantly increased in the eMSC group. Moreover, eMSC had a positive impact on the regeneration of both stromal and epithelial components of the mouse endometrium, indicated by significantly higher vimentin and CK19 protein expression. Reduced endometrial fibrosis and down-regulation of fibrosis markers were also observed in the eMSC group. The eMSC group had a significantly higher gene expression of anti-inflammatory factor Il-10 and lower mRNA level of pro-inflammatory factors Ifng and Il-2, indicating the role of eMSC in regulation of inflammatory reactions. The eMSC group showed higher implantation sites than the PBS group, suggesting better endometrial receptivity with the presence of newly emerged endometrial lining. CONCLUSIONS: Our findings suggest eMSC improves regeneration of injured endometrium in mice.


Assuntos
Células-Tronco Mesenquimais , Doenças Uterinas , Camundongos , Feminino , Humanos , Gravidez , Animais , Camundongos Endogâmicos NOD , Camundongos SCID , Placenta/patologia , Endométrio/metabolismo , Endométrio/patologia , Doenças Uterinas/terapia , Doenças Uterinas/metabolismo , Doenças Uterinas/patologia , Fibrose
3.
Biol. Res ; 57: 6-6, 2024. ilus, graf
Artigo em Inglês | LILACS | ID: biblio-1550061

RESUMO

BACKGROUND: The monthly regeneration of human endometrial tissue is maintained by the presence of human endometrial mesenchymal stromal/stem cells (eMSC), a cell population co-expressing the perivascular markers CD140b and CD146. Endometrial regeneration is impaired in the presence of intrauterine adhesions, leading to infertility, recurrent pregnancy loss and placental abnormalities. Several types of somatic stem cells have been used to repair the damaged endometrium in animal models, reporting successful pregnancy. However, the ability of endometrial stem cells to repair the damaged endometrium remains unknown. METHODS: Electrocoagulation was applied to the left uterine horn of NOD/SCID mice causing endometrial injury. Human eMSC or PBS was then injected into the left injured horn while the right normal horn served as controls. Mice were sacrificed at different timepoints (Day 3, 7 and 14) and the endometrial morphological changes as well as the degree of endometrial injury and repair were observed by histological staining. Gene expression of various inflammatory markers was assessed using qPCR. The functionality of the repaired endometrium was evaluated by fertility test. RESULTS: Human eMSC successfully incorporated into the injured uterine horn, which displayed significant morphological restoration. Also, endometrium in the eMSC group showed better cell proliferation and glands formation than the PBS group. Although the number of blood vessels were similar between the two groups, gene expression of VEGF-α significantly increased in the eMSC group. Moreover, eMSC had a positive impact on the regeneration of both stromal and epithelial components of the mouse endometrium, indicated by significantly higher vimentin and CK19 protein expression. Reduced endometrial fibrosis and down-regulation of fibrosis markers were also observed in the eMSC group. The eMSC group had a significantly higher gene expression of anti-inflammatory factor Il-10 and lower mRNA level of pro-inflammatory factors Ifng and Il-2, indicating the role of eMSC in regulation of inflammatory reactions. The eMSC group showed higher implantation sites than the PBS group, suggesting better endometrial receptivity with the presence of newly emerged endometrial lining. CONCLUSIONS: Our findings suggest eMSC improves regeneration of injured endometrium in mice.


Assuntos
Humanos , Animais , Feminino , Gravidez , Camundongos , Doenças Uterinas/metabolismo , Doenças Uterinas/patologia , Doenças Uterinas/terapia , Células-Tronco Mesenquimais , Placenta/patologia , Fibrose , Camundongos SCID , Camundongos Endogâmicos NOD , Endométrio/metabolismo , Endométrio/patologia
4.
Front Cell Dev Biol ; 10: 1060298, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36561369

RESUMO

The placenta is important for fetal development in mammals, and spatial transcriptomic profiling of placenta helps to resolve its structure and function. In this study, we described the landscape of spatial transcriptome of human placental villi obtained from two pregnant women at the first trimester using the modified Stereo-seq method applied for paraformaldehyde (PFA) fixation samples. The PFA fixation of human placenta villi was better than fresh villi embedded in optimum cutting temperature (OCT) compound, since it greatly improved tissue morphology and the specificity of RNA signals. The main cell types in chorionic villi such as syncytiotrophoblasts (SCT), villous cytotrophoblasts (VCT), fibroblasts (FB), and extravillous trophoblasts (EVT) were identified with the spatial transcriptome data, whereas the minor cell types of Hofbauer cells (HB) and endothelial cells (Endo) were spatially located by deconvolution of scRNA-seq data. We demonstrated that the Stereo-seq data of human villi could be used for sophisticated analyses such as spatial cell-communication and regulatory activity. We found that the SCT and VCT exhibited the most ligand-receptor pairs that could increase differentiation of the SCT, and that the spatial localization of specific regulons in different cell types was associated with the pathways related to hormones transport and secretion, regulation of mitotic cell cycle, and nutrient transport pathway in SCT. In EVT, regulatory pathways such as the epithelial to mesenchyme transition, epithelial development and differentiation, and extracellular matrix organization were identified. Finally, viral receptors and drug transporters were identified in villi according to the pathway analysis, which could help to explain the vertical transmission of several infectious diseases and drug metabolism efficacy. Our study provides a valuable resource for further investigation of the placenta development, physiology and pathology in a spatial context.

5.
Commun Biol ; 5(1): 1064, 2022 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-36207605

RESUMO

Human endometrium undergoes cycles of regeneration in women of reproductive age. The endometrial mesenchymal stromal/stem cells (eMSC) contribute to this process. Notch signaling is essential for homeostasis of somatic stem cells. However, its role in eMSC remains unclear. We show with gain- and loss-of-function experiments that activation of Notch signaling promotes eMSC maintenance, while inhibition induces opposite effect. The activation of Notch signaling better maintains eMSC in a quiescent state. However, these quiescent eMSC can re-enter the cell cycle depending on the Notch and Wnt activities in the microenvironment, suggesting a crosstalk between the two signaling pathways. We further show that the Notch signaling is involved in endometrial remodeling event in a mouse menstrual-like model. Suppression of Notch signaling reduces the proliferation of Notch1+ label-retaining stromal cells and delays endometrial repair. Our data demonstrate the importance of Notch signaling in regulating the endometrial stem/progenitor cells in vitro and in vivo.


Assuntos
Endométrio , Células-Tronco Mesenquimais , Animais , Endométrio/metabolismo , Feminino , Humanos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Transdução de Sinais , Células-Tronco , Células Estromais
6.
Reprod Biol Endocrinol ; 20(1): 120, 2022 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-35964080

RESUMO

During implantation, a symphony of interaction between the trophoblast originated from the trophectoderm of the implanting blastocyst and the endometrium leads to a successful pregnancy. Defective interaction between the trophoblast and endometrium often results in implantation failure, pregnancy loss, and a number of pregnancy complications. Owing to ethical concerns of using in vivo approaches to study human embryo implantation, various in vitro culture models of endometrium were established in the past decade ranging from two-dimensional cell-based to three-dimensional extracellular matrix (ECM)/tissue-based culture systems. Advanced organoid systems have also been established for recapitulation of different cellular components of the maternal-fetal interface, including the endometrial glandular organoids, trophoblast organoids and blastoids. However, there is no single ideal model to study the whole implantation process leaving more research to be done pursuing the establishment of a comprehensive in vitro model that can recapitulate the biology of trophoblast-endometrium interaction during early pregnancy. This would allow us to have better understanding of the physiological and pathological process of trophoblast-endometrium interaction during implantation.


Assuntos
Implantação do Embrião , Trofoblastos , Blastocisto , Implantação do Embrião/fisiologia , Embrião de Mamíferos , Endométrio , Feminino , Humanos , Gravidez , Trofoblastos/fisiologia
8.
Int J Mol Sci ; 23(9)2022 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-35563003

RESUMO

Human endometrium is an incredibly dynamic tissue undergoing cyclic regeneration and shedding during a woman's reproductive life. Endometrial mesenchymal stromal/stem-like cells (eMSC) contribute to this process. A hypoxic niche with low oxygen levels has been reported in multiple somatic stem cell types. However, the knowledge of hypoxia on eMSC remains limited. In mice, stromal stem/progenitor cells can be identified by the label-retaining technique. We examined the relationship between the label-retaining stromal cells (LRSC) and hypoxia during tissue breakdown in a mouse model of simulated menses. Our results demonstrated that LRSC resided in a hypoxic microenvironment during endometrial breakdown and early repair. Immunofluorescence staining revealed that the hypoxic-located LRSC underwent proliferation and was highly colocalized with Notch1. In vitro studies illustrated that hypoxia activated Notch signaling in eMSC, leading to enhanced self-renewal, clonogenicity and proliferation of cells. More importantly, HIF-1α played an essential role in the hypoxia-mediated maintenance of eMSC through the activation of Notch signaling. In conclusion, our findings show that some endometrial stem/progenitor cells reside in a hypoxic niche during menstruation, and hypoxia can regulate the self-renewal activity of eMSC via Notch signaling.


Assuntos
Hipóxia Celular , Endométrio , Células-Tronco Mesenquimais , Animais , Hipóxia Celular/fisiologia , Endométrio/metabolismo , Feminino , Hipóxia/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Receptores Notch/metabolismo , Transdução de Sinais , Células-Tronco/metabolismo
10.
Front Cell Dev Biol ; 10: 837827, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35295855

RESUMO

Endometrial mesenchymal stem-like cells (eMSC) reside in the basal layer of the endometrium and are responsible for cyclic regeneration during the reproductive lives of women. Myometrial cells act as a component of the niche and regulate the stem cell fate through the activation of WNT/ß-catenin signaling via WNT5A. Since WNT5A-responsive mechanisms on eMSC are still uncertain, we hypothesize that the WNT ligand-WNT5A works to activate WNT/ß-catenin signaling through binding to Frizzled receptors (FZDs) and co-receptor low-density lipoprotein receptor-related protein 5 (LRP5). Among the various receptors that have been reported to interact with WNT5A, we found FZD5 abundantly expressed by eMSC when compared to unfractionated stromal cells. Neutralizing the protein expression by using anti-FZD5 antibody suppressed the stimulatory effects on phenotypic expression and the clonogenicity of eMSC in a myometrial cell-eMSC co-culture system as well as in an L-Wnt5a conditioned medium. Gene silencing of FZD5 not only reduced the binding of WNT5A to eMSC but also decreased the TCF/LEF transcriptional activities and expression of active ß-catenin. Inhibition of LRP coreceptors with recombinant Dickkopf-1 protein significantly reduced the binding affinity of eMSC to WNT5A as well as the proliferation and self-renewal activity. During postpartum remodeling in mouse endometrium, active ß-catenin (ABC) was detected in label-retaining stromal cells (LRSCs), and these ABC+ LRSCs express FZD5 and LRP5, suggesting the activation of WNT/ß-catenin signaling. In conclusion, our findings demonstrate the interaction of WNT5A, FZD5, and LRP5 in regulating the proliferation and self-renewal of eMSC through WNT/ß-catenin signaling.

11.
Stem Cell Res Ther ; 12(1): 306, 2021 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-34051872

RESUMO

BACKGROUND: Endometrial mesenchymal-like stromal/stem cells (eMSCs) have been proposed as adult stem cells contributing to endometrial regeneration. One set of perivascular markers (CD140b&CD146) has been widely used to enrich eMSCs. Although eMSCs are easily accessible for regenerative medicine and have long been studied, their cellular heterogeneity, relationship to primary counterpart, remains largely unclear. METHODS: In this study, we applied 10X genomics single-cell RNA sequencing (scRNA-seq) to cultured human CD140b+CD146+ endometrial perivascular cells (ePCs) from menstrual and secretory endometrium. We also analyzed publicly available scRNA-seq data of primary endometrium and performed transcriptome comparison between cultured ePCs and primary ePCs at single-cell level. RESULTS: Transcriptomic expression-based clustering revealed limited heterogeneity within cultured menstrual and secretory ePCs. A main subpopulation and a small stress-induced subpopulation were identified in secretory and menstrual ePCs. Cell identity analysis demonstrated the similar cellular composition in secretory and menstrual ePCs. Marker gene expression analysis showed that the main subpopulations identified from cultured secretory and menstrual ePCs simultaneously expressed genes marking mesenchymal stem cell (MSC), perivascular cell, smooth muscle cell, and stromal fibroblast. GO enrichment analysis revealed that genes upregulated in the main subpopulation enriched in actin filament organization, cellular division, etc., while genes upregulated in the small subpopulation enriched in extracellular matrix disassembly, stress response, etc. By comparing subpopulations of cultured ePCs to the publicly available primary endometrial cells, it was found that the main subpopulation identified from cultured ePCs was culture-unique which was unlike primary ePCs or primary endometrial stromal fibroblast cells. CONCLUSION: In summary, these data for the first time provides a single-cell atlas of the cultured human CD140b+CD146+ ePCs. The identification of culture-unique relatively homogenous cell population of CD140b+CD146+ ePCs underscores the importance of in vivo microenvironment in maintaining cellular identity.


Assuntos
Endométrio , Pericitos , Adulto , Antígeno CD146/genética , Diferenciação Celular , Células Cultivadas , Feminino , Humanos , Análise de Sequência de RNA
12.
Stem Cell Res Ther ; 11(1): 239, 2020 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-32552749

RESUMO

BACKGROUND: The identification of endometrial stem/progenitor cells in a high turnover rate tissue suggests that a well-orchestrated underlying network controls the behaviour of these stem cells. The thickness of the endometrium can grow from 0.5-1 mm to 5-7 mm within a week indicating the need of stem cells for self-renewal and differentiation during this period. The cyclical regeneration of the endometrium suggests specific signals can activate the stem cells during or shortly after menstruation. METHODS: Endometrial mesenchymal stem-like cells (eMSCs) were cocultured with endometrial epithelial or stromal cells from different phases of the menstrual cycle; the clonogenicity and the phenotypic expression of eMSC markers (CD140b and CD146) were assessed. The functional role of WNT/ß-catenin signalling on eMSC was determined by western blot analysis, immunofluorescent staining, flow cytometry, quantitative real-time PCR and small interfering RNA. The cytokine levels in the conditioned medium of epithelial or stromal cells cocultured with eMSCs were evaluated by enzyme-linked immunosorbent assays. RESULTS: Coculture of endometrial cells (epithelial or stromal) from the menstrual phase enhanced the clonogenicity and self-renewal activities of eMSCs. Such phenomenon was not observed in niche cells from the proliferative phase. Coculture with endometrial cells from the menstrual phase confirmed an increase in expression of active ß-catenin in the eMSCs. Treatment with IWP-2, a WNT inhibitor, suppressed the observed effects. Anti-R-spondin-1 antibody reduced the stimulatory action of endometrial niche cells on WNT/ß-catenin activation in the T cell factor/lymphoid enhancer-binding factor luciferase reporter assay. Moreover, the mRNA level and protein immunoreactivities of leucine-rich repeat-containing G-protein coupled receptor 5 were higher in eMSCs than unfractionated stromal cells. Conditioned media of endometrial niche cells cocultured with eMSCs contained increased levels of C-X-C motif ligand 1 (CXCL1), CXCL5 and interleukin 6. Treatment with these cytokines increased the clonogenic activity and phenotypic expression of eMSCs. CONCLUSIONS: Our findings indicate a role of WNT/ß-catenin signalling in regulating activities of endometrial stem/progenitor cells during menstruation. Certain cytokines at menstruation can stimulate the proliferation and self-renewal activities of eMSCs. Understanding the mechanism in the regulation of eMSCs may contribute to treatments of endometrial proliferative disorders such as Asherman's syndrome.


Assuntos
Menstruação , Células-Tronco Mesenquimais , Diferenciação Celular , Endométrio , Feminino , Humanos , Células-Tronco
13.
Stem Cells ; 37(11): 1455-1466, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31414525

RESUMO

Human endometrium undergoes cycles of proliferation and differentiation throughout the reproductive years of women. The endometrial stem/progenitor cells contribute to this regenerative process. They lie in the basalis layer of the endometrium next to the myometrium. We hypothesized that human myometrial cells provide niche signals regulating the activities of endometrial mesenchymal stem-like cells (eMSCs). In vitro coculture of myometrial cells enhanced the colony-forming and self-renewal ability of eMSCs. The cocultured eMSCs retained their multipotent characteristic and exhibited a greater total cell output when compared with medium alone culture. The expression of active ß-catenin in eMSCs increased significantly after coculture with myometrial cells, suggesting activation of WNT/ß-catenin signaling. Secretory factors in spent medium from myometrial cell culture produced the same stimulatory effects on eMSCs. The involvement of WNT/ß-catenin signaling in self-renewal of eMSCs was confirmed with the use of WNT activator (Wnt3A conditioned medium) and WNT inhibitors (XAV939 and inhibitor of Wnt Production-2 [IWP-2]). The myometrial cells expressed more WNT5A than other WNT ligands. Recombinant WNT5A stimulated whereas anti-WNT5A antibody suppressed the colony formation, self-renewal, and T-cell factor/lymphoid enhancer-binding factor (TCF/LEF) transcriptional activities of eMSCs. Moreover, eMSCs expressed FZD4 and LRP5. WNT5A is known to activate the canonical WNT signaling in the presence of these receptor components. WNT antagonist, DKK1, binds to LRP5/6. Consistently, DKK1 treatment nullified the stimulatory effect of myometrial cell coculture. In conclusion, our findings show that the myometrial cells are niche components of eMSCs, modulating the self-renewal activity of eMSCs by WNT5A-dependent activation of WNT/ß-catenin signaling. Stem Cells 2019;37:1455-1466.


Assuntos
Cateninas/metabolismo , Endométrio/metabolismo , Células-Tronco Mesenquimais/metabolismo , Miométrio/metabolismo , Proteínas Wnt/metabolismo , Proteína Wnt-5a/metabolismo , Adulto , Cateninas/genética , Células Cultivadas , Endométrio/citologia , Endométrio/efeitos dos fármacos , Feminino , Citometria de Fluxo , Imunofluorescência , Receptores Frizzled/genética , Receptores Frizzled/metabolismo , Inativação Gênica/fisiologia , Compostos Heterocíclicos com 3 Anéis/farmacologia , Humanos , Proteína-5 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Proteína-5 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Fator 1 de Ligação ao Facilitador Linfoide/genética , Fator 1 de Ligação ao Facilitador Linfoide/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Pessoa de Meia-Idade , Miométrio/citologia , Miométrio/efeitos dos fármacos , Proteínas Wnt/genética , Via de Sinalização Wnt/efeitos dos fármacos , Via de Sinalização Wnt/genética , Proteína Wnt-5a/genética
14.
Cell Prolif ; 50(3)2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28071833

RESUMO

OBJECTIVE: To study the effect on endometrial and endometriotic cells after co-culture with macrophages, using clonogenic, invasion and self-renewal assays. MATERIALS AND METHODS: Peripheral blood samples, endometrium and endometriotic tissues were collected. Autologous macrophages were co-cultured with endometrial and endometriotic cells. The number of colony-forming units (CFU), invasiveness and self-renewal activity after co-culture with macrophages were determined. The cytokine level of colony-stimulating factor-1 (CSF-1) from macrophages with and without endometriosis was compared. RESULTS: Co-culture with macrophages significantly increased the clonogenic and invasion ability of endometriotic stromal cells in vitro. Colony-stimulating factor-1 (CSF-1) was up-regulated in endometriotic macrophages conditioned medium when compared to those without the disease. CONCLUSIONS: These data suggest that macrophages may increase the proliferation and invasion activity of stromal clonogenic cells in women with endometriosis.


Assuntos
Movimento Celular , Células Clonais/citologia , Técnicas de Cocultura , Endométrio/citologia , Macrófagos/citologia , Células Estromais/citologia , Adulto , Feminino , Humanos , Pessoa de Meia-Idade
15.
Exp Cell Res ; 350(1): 184-189, 2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-27890645

RESUMO

The human endometrium is a highly dynamic tissue with the ability to cyclically regenerate during the reproductive life. Endometrial mesenchymal stem-like cells (eMSCs) located throughout the endometrium have shown to functionally contribute to endometrial regeneration. In this study we examine whether the menstrual cycle stage and the location in the endometrial bilayer (superficial and deep portions of the endometrium) has an effect on stem cell activities of eMSCs (CD140b+CD146+ cells). Here we show the percentage and clonogenic ability of eMSCs were constant in the various stages of the menstrual cycle (menstrual, proliferative and secretory). However, eMSCs from the menstrual endometrium underwent significantly more rounds of self-renewal and enabled a greater total cell output than those from the secretory phase. Significantly more eMSCs were detected in the deeper portion of the endometrium compared to the superficial layer but their clonogenic and self-renewal activities remained similar. Our findings suggest that eMSCs are activated in the menstrual phase for the cyclical regeneration of the endometrium.


Assuntos
Endométrio/citologia , Ciclo Menstrual/metabolismo , Células-Tronco Mesenquimais/citologia , Diferenciação Celular , Endométrio/ultraestrutura , Feminino , Humanos , Regeneração
16.
Stem Cells Dev ; 24(6): 768-80, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25386902

RESUMO

Human and mouse endometrium undergo dramatic cellular reorganization during pregnancy and postpartum. Somatic stem cells maintain homeostasis of the tissue by providing a cell reservoir for regeneration. We hypothesized that endometrial cells with quiescent properties (stem/progenitor cells) were involved in the regeneration of the endometrial tissue. Given that stem cells divide infrequently, they can retain the DNA synthesis label [bromodeoxyuridine (BrdU)] after a prolonged chase period. In this study, prepubertal mice were pulsed with BrdU and after a 6-week chase a small population of label-retaining stromal cells (LRSC) was located primarily beneath the luminal epithelium, adjacent to blood vessels, and near the endometrial-myometrial junction. Marker analyses suggested that they were of mesenchymal origin expressing CD44(+), CD90(+), CD140b(+), CD146(+), and Sca-1(+). During pregnancy, nonproliferating LRSC predominately resided at the interimplantation/placental loci of the gestational endometrium. Immediately after parturition, a significant portion of the LRSC underwent proliferation (BrdU(+)/Ki-67(+)) and expressed total and active ß-catenin. The ß-catenin expression in the LRSC was transiently elevated at postpartum day (PPD) 1. The proliferation of LRSC resulted in a significant decline in the proportion of LRSC in the postpartum uterus. The LRSC returned to dormancy at PPD7, and the percentage of LRSC remained stable thereafter until 11 weeks. This study demonstrated that LRSC can respond efficiently to physiological stimuli upon initiation of uterine involution and return to its quiescent state after postpartum repair.


Assuntos
Proliferação de Células , Endométrio/citologia , Células-Tronco Mesenquimais/citologia , Parto , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Endométrio/fisiologia , Feminino , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , beta Catenina/genética , beta Catenina/metabolismo
17.
Stem Cell Res Ther ; 5(4): 84, 2014 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-24996487

RESUMO

INTRODUCTION: Evidence suggests that the human endometrium contains stem or progenitor cells that are responsible for its remarkable regenerative capability. A common property of somatic stem cells is their quiescent state. It remains unclear whether slow-cycling cells exist in the human endometrium. We hypothesized that the human endometrium contains a subset of slow-cycling cells with somatic stem cell properties. Here, we established an in vitro stem cell assay to isolate human endometrial-derived mesenchymal stem-like cells (eMSC). METHODS: Single-cell stromal cultures were initially labeled with fluorescent nanoparticles and a small population of fluorescent persistent cells (FPC) remained after culture of 21 days. Two populations of stromal cells, namely FPC and non-FPC were sorted. RESULTS: Quantitative analysis of functional assays demonstrated that the FPC had higher colony forming ability, underwent more rounds of self-renewal and had greater enrichment of phenotypically defined prospective eMSC markers: CD146+/CD140b+ and W5C5+ than the non-FPC. They also differentiate into multiple mesenchymal lineages and the expression of lineage specific markers was lower than that of non-FPC. The FPC exhibit low proliferation activities. A proliferation dynamics study revealed that more FPC had a prolonged G1 phase. CONCLUSIONS: With this study we present an efficient method to label and isolate slow-proliferating cells obtained from human endometrial stromal cultures without genetic modifications. The FPC population could be easily maintained in vitro and are of interest for tissue-repair and engineering perspectives. In summary, nanoparticle labeling is a promising tool for the identification of putative somatic stem or progenitor cells when their surface markers are undefined.


Assuntos
Proliferação de Células , Endométrio/citologia , Adulto , Diferenciação Celular , Linhagem da Célula , Feminino , Citometria de Fluxo , Fase G1 , Humanos , Imuno-Histoquímica , Pessoa de Meia-Idade , Nanopartículas/análise , Células Estromais , Imagem com Lapso de Tempo
18.
Methods Mol Biol ; 945: 347-63, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23097117

RESUMO

The female reproductive system is a complex system. Epithelia of the female reproductive system including the ovaries, the oviduct, and the uterus are important sites for follicular development, ovulation, fertilization, implantation, and embryo development. They are also able to synthesize and secrete various hormones, growth factors, and cytokines, which are essential to women's health, sexuality, and reproduction. Conversely, their dysfunction has been implicated in disorders such as infertility, endometriosis, and many other gynecological diseases, as well as cancer. In this chapter, we describe detailed procedures for establishing and maintaining primary cultures of human ovarian surface epithelium, oviductal epithelium, and endometrium. We also provide protocols for cell immortalization, clonal isolation, and in coculture with stromal cells. These cultures can be useful models for investigating the molecular and cellular functions of these epithelia in both normal and pathological states.


Assuntos
Técnicas de Cultura de Células/métodos , Células Epiteliais/citologia , Genitália Feminina/citologia , Antígenos Transformantes de Poliomavirus/genética , Proliferação de Células , Separação Celular , Técnicas de Cocultura , Células Epiteliais/metabolismo , Feminino , Humanos , Células-Tronco/citologia , Células Estromais/citologia , Telomerase/genética
19.
Reprod Sci ; 19(1): 102-14, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22064386

RESUMO

Candidate stem/progenitor cells have been identified in mouse endometrium as label-retaining cells (LRCs). The role of endometrial stem/progenitor cells in initiating estrogen-stimulated endometrial growth in prepubertal and cycling mice was investigated following a single 17ß-estradiol (E2) injection in bromodeoxyuridine (BrdU)-labeled and -chased (LRC), ovariectomised mice. Proliferating (BrdU(+)/Ki-67(+)) and mitotic (BrdU(+)/PH3(+)) epithelial LRCs were first detected in prepubertal mice 8 hours following E2 treatment, initiating the proliferative response. In contrast, all epithelial LRCs and 16% of epithelial cells in cycling mice proliferated within 2 hours. In cycling mice, 12% of stromal LRCs initiated a proliferative response 8 hours after E2. Proliferating epithelial LRCs and most stromal LRCs (85%) lacked estrogen receptor-α (ESR1). These findings suggest that endometrial epithelial LRCs function as stem/progenitor cells by receiving proliferative signals from neighboring ESR1(+) niche cells to initiate the growth of the epithelium during development, while mature epithelial cells may undergo self-replication in cycling endometrium.


Assuntos
Endométrio/citologia , Endométrio/efeitos dos fármacos , Estrogênios/farmacologia , Regeneração/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Animais , Proliferação de Células/efeitos dos fármacos , Feminino , Camundongos , Distribuição Aleatória
20.
Fertil Steril ; 94(2): 769-71, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19945098
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA