Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.259
Filter
1.
J Ethnopharmacol ; 336: 118724, 2025 Jan 10.
Article in English | MEDLINE | ID: mdl-39181283

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Wenshen Xiaozheng Tang (WXT), a traditional Chinese medicine (TCM) decoction, is effective for treating endometriosis. However, the effect of WXT on endometrium-derived mesenchymal stem cells (eMSCs) which play a key role in the fibrogenesis of endometriosis requires further elucidation. AIMS OF THE STUDY: The aim of this study was to clarify the potential mechanism of WXT in improving fibrosis in endometriosis by investigating the regulation of WXT on differentiation and paracrine of eMSCs. MATERIALS AND METHODS: The nude mice with endometriosis were randomly divided into model group, WXT group and mifepristone group. After 21 days of treatment, the lesion volume was calculated. Fibrosis in the lesions was evaluated by Masson staining and expression of fibrotic proteins. The differentiation of eMSCs in vivo was explored using a fate-tracking experiment. To further clarify the regulation of WXT on eMSCs, primary eMSCs from the ectopic lesions of endometriosis patients were isolated and characterized. The effect of WXT on the proliferation and differentiation of ectopic eMSCs was examined. To evaluate the role of WXT on the paracrine activity of ectopic eMSCs, the conditioned medium (CM) from ectopic eMSCs pretreated with WXT was collected and applied to treat ectopic endometrial stromal cells (ESCs), after which the expression of fibrotic proteins in ectopic ESCs was assessed. In addition, transcriptome sequencing was used to investigate the regulatory mechanism of WXT on ectopic eMSCs, and western blot and ELISA were employed to determine the key mediator. RESULTS: WXT impeded the growth of ectopic lesions in nude mice with endometriosis and reduced collagen deposition and the expression of fibrotic proteins fibronectin, collagen I, α-SMA and CTGF in the endometriotic lesions. The fate-tracking experiment showed that WXT prevented human eMSCs from differentiating into myofibroblasts in the nude mice. We successfully isolated eMSCs from the lesions of patients with endometriosis and demonstrated that WXT suppressed proliferation and myofibroblast differentiation of ectopic eMSCs. Moreover, the expression of α-SMA, collagen I, fibronectin and CTGF in ectopic ESCs was significantly down-regulated by the CM of ectopic MSCs pretreated with WXT. Combining the results of RNA sequencing, western blot and ELISA, we found that WXT not only reduced thrombospondin 4 expression in ectopic eMSCs, but also decreased thrombospondin 4 secretion from ectopic eMSCs. Thrombospondin 4 concentration-dependently upregulated the expression of collagen I, fibronectin, α-SMA and CTGF in ectopic ESCs, indicating that thrombospondin 4 was a key mediator of WXT in inhibiting the fibrotic process in endometriosis. CONCLUSION: WXT improved fibrosis in endometriosis by regulating differentiation and paracrine signaling of eMSCs. Thrombospondin 4, whose release from ectopic eMSCs is inhibited by WXT, may be a potential target for the treatment of endometriosis.


Subject(s)
Cell Differentiation , Drugs, Chinese Herbal , Endometriosis , Endometrium , Fibrosis , Mesenchymal Stem Cells , Mice, Nude , Paracrine Communication , Endometriosis/drug therapy , Endometriosis/pathology , Endometriosis/metabolism , Female , Animals , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Paracrine Communication/drug effects , Humans , Cell Differentiation/drug effects , Endometrium/drug effects , Endometrium/metabolism , Endometrium/pathology , Mice , Cells, Cultured , Adult , Disease Models, Animal
2.
J Tradit Chin Med ; 44(5): 991-999, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39380230

ABSTRACT

OBJECTIVE: To investigate the effects and mechanisms of the Jiawei Pentongling formula (, JWPTL) in treating endometriosis-related pain using network pharmacology study and experimental validation. METHODS: Active ingredients and relevant targets of JWPTL, as well as genes for endometriosis-related pain, were collected from public databases. Prediction of core targets and pathways of JWPTL against pain associated with endometriosis by protein-protein interaction (PPI) network work, gene ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) functional enrichment analysis. The Sprague-Dawley rat endo-metriosis model was constructed using the autologous endosomal transplantation method, and the successfully modeled rats were randomly divided into the model group and the JWPTL group, with 8 rats in each group. Another 8 rats were set up in the sham group. Rats in the JWPTL group used the rectal delivery method with the addition of JWPTL (7.77 g·kg-1·d-1) once a day for 28 d. Rats in the model and sham-operated groups were given equal amounts of saline using the same administration method. The 50% paw withdrawal threshold (PWT) of the rats was measured at different time points. After the intervention, the expression of phosphatidylinositol 3-kinase (PI3K) and protein kinase B (Akt) proteins and mRNA in endometriotic tissues was detected by immune-ohistochemistry and reverse transcription-polymerase chain reaction. RESULTS: From GeneCards, Online Mendelian Inheritance in Man and other databases, a total of 964 endometriosis (EMs) -related pain targets were screened, 142 active ingredients of JWPTL, 605 targets, and 221 potential targets were obtained by intersection of Venn diagram; 44 core targets were identified by constructing PPI network. KEGG enrichment analysis showed that JWPTL mainly involves the PI3K-Akt signaling pathway, estrogen signaling pathway, hypoxia inducible factor-1 signaling pathway, tumour necrotizing factor signaling pathway, and other signaling pathways in the treatment of EMs-related pain. Animal experiments showed that JWPTL could up-regulate the mechanical pain threshold and reduce the expression of PI3K and Akt proteins and mRNA in ectopic endometrial tissues of model rats. CONCLUSIONS: The present study preliminarily analyzed the pharmacological mechanism of the formula, and molecular docking and animal experiments showed the feasibility of this study, suggesting that the formula may inhibit the release of inflammatory factors and reverse the pain associated with EMs by downregulating the PI3K/Akt signaling pathway.


Subject(s)
Drugs, Chinese Herbal , Endometriosis , Network Pharmacology , Rats, Sprague-Dawley , Female , Animals , Endometriosis/drug therapy , Endometriosis/metabolism , Endometriosis/genetics , Drugs, Chinese Herbal/administration & dosage , Rats , Humans , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/genetics , Pain/drug therapy , Pain/metabolism , Pain/genetics , Protein Interaction Maps , Signal Transduction/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/genetics
3.
Int J Mol Sci ; 25(18)2024 Sep 21.
Article in English | MEDLINE | ID: mdl-39337624

ABSTRACT

Endometriosis, a debilitating condition, affects one in ten women of reproductive age. Its pathophysiology remains unclear, though deficiencies in immune surveillance are thought to create an environment conducive to the evasion of ectopic endometrial cells from the immune system. Our research explores the immunological impact of endometriosis both locally and systemically, emphasizing natural killer (NK) and T cell subpopulations. We incorporated 62 female patients who underwent laparoscopic surgery; of those, 47 had endometriosis, and 15 were controls. We collected peritoneal fluid (PF) and peripheral blood (PB) samples which were tagged with monoclonal antibodies and subsequently scrutinized using flow cytometry. Our findings revealed significant differences in immunological profiles based on demographic factors and symptomatology. In the endometriosis cohort, there was an increase in PB CD56HiCD16dim and PF CD8+ CD56dimCD16Hi NK cells. CD16+ CD4 T cell levels were significantly lower in the PB of endometriosis patients who smoke. Individuals with more severe disease displayed significantly higher levels of PB CD16+ CD8 T cells, which also increased in those with non-menstrual pelvic pain. Dysmenorrhea severity correlated with a progressive increase in PF CD8+ CD56dimCD16Hi NK cells. These variations in specific lymphocyte subsets, namely, within NK and T cells, suggest potential immunological mechanisms in the evolution and clinical presentation of endometriosis.


Subject(s)
Endometriosis , Killer Cells, Natural , Humans , Endometriosis/immunology , Endometriosis/pathology , Endometriosis/metabolism , Female , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Adult , Ascitic Fluid/immunology , Ascitic Fluid/metabolism , Ascitic Fluid/pathology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD56 Antigen/metabolism , Flow Cytometry
4.
Rev Assoc Med Bras (1992) ; 70(9): e20240485, 2024.
Article in English | MEDLINE | ID: mdl-39292088

ABSTRACT

OBJECTIVE: Endometriosis is a disease in which stromal cells and endometrial glands extend outside of the uterine cavity. Nevertheless, treatment failure and recurrence cause difficulties in management. This study aimed to evaluate the receptor-level components of bilateral endometriomas in the recurrence state. METHODS: Our retrospective cohort study was conducted with patients who underwent surgery for bilateral endometriomas between 2015 and 2021. In total, 113 patients were allocated. A total of 76 patients did not meet the eligibility criteria, and the data of 37 patients were evaluated. Medical treatments, recurrences, and postoperative follow-up data were collected. In archived tissue samples, measurements of progesterone receptor A and progesterone receptor B, histoscores and immunoreactivity scores, and their ratios were calculated in the group that received no postoperative medical treatment. Criteria for recurrence were a repeat operation and/or the detection of a new endometrioma>2 cm at the follow-up examination. RESULTS: No recurrence was observed in 73.0% (n=27) of the cases, whereas recurrence was observed in 27.0% (n=10) of the participants. Patients without recurrence had significantly higher progesterone receptor B histoscore/progesterone receptor A histoscore and progesterone receptor B immunoreactivity score/progesterone receptor A immunoreactivity score results (p=0.01). Nevertheless, when the histoscores and immunoreactivity scores for both receptors were contrasted separately, there was no appreciable difference between them. CONCLUSION: The dominance of progesterone receptor B over progesterone receptor A was inversely proportional to the recurrence status in bilateral endometriomas. Furthermore, our study revealed that assessing receptor levels alone did not result in a significant difference in recurrence.


Subject(s)
Endometriosis , Receptors, Progesterone , Humans , Female , Endometriosis/surgery , Endometriosis/metabolism , Endometriosis/pathology , Receptors, Progesterone/metabolism , Receptors, Progesterone/analysis , Adult , Retrospective Studies , Recurrence , Middle Aged , Immunohistochemistry
5.
J Ovarian Res ; 17(1): 183, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39267109

ABSTRACT

Ferroptosis is a novel type of programmed cell death dependent on iron and characterized by the accumulation of lipid peroxides in cells and is closely related to various diseases. Female infertility is a global health concern, which is associated with a variety of factors. The etiology remains unknown in many women with infertility. With further investigation into the pathogenesis of infertility, a growing number of studies have demonstrated the close connections between infertility and ferroptosis. Through a literature review, it is found that ferroptosis is closely involved in endometriosis- and polycystic ovarian syndrome (PCOS)-associated infertility and tubal factor infertility. Iron overload increases the resistance to ferroptosis, and ferroptosis in some cells accelerates endometrial lesion growth. Moreover, iron overload may be hazardous to oocytes. This review may shed some light on the diagnosis and treatment of female infertility.


Subject(s)
Ferroptosis , Infertility, Female , Humans , Female , Infertility, Female/metabolism , Infertility, Female/pathology , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/pathology , Endometriosis/pathology , Endometriosis/metabolism , Animals
6.
BMC Med ; 22(1): 351, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39218863

ABSTRACT

BACKGROUND: Endometriosis is well known as a chronic inflammatory disease. The development of endometriosis is heavily influenced by the estrogen receptor ß (ERß), while NOD-like receptors (NLRs) family CARD domain-containing 5 (NLRC5) exhibits anti-inflammatory properties during endometriosis. However, whether NLRC5-mediated anti-inflammation is involved in the ERß-mediated endometriosis is still uncertain. This study aimed to assess that relation. METHODS: Nine cases of eutopic endometrial tissue and ten cases of ectopic endometrial tissue were collected from patients with endometriosis, and endometrial samples from ten healthy fertile women were analyzed, and the expression levels of ERß were quantified using immunohistochemistry (IHC). Subsequently, we constructed mouse model of endometriosis by intraperitoneal injection. We detected the expression of ERß, NLRC5, tumor necrosis factor-alpha (TNF-α), interleukin (IL)-6, and IL-10 and measured the volume of ectopic lesions in mice with endometriosis. In vitro, human endometrial stromal cells (hESCs) were transfected respectively with ERß-overexpressing and NLRC5-overexpressing plasmids. We then assessed the expression of ERß and NLRC5 using quantitative real-time PCR (qRT-PCR) and western blot analysis. Furthermore, we measured the concentrations of TNF-α, IL-6, and IL-10 in the cell culture supernatant through enzyme-linked immunosorbent assay (ELISA). Additionally, we evaluated the migration and invasion ability of hESCs using transwell and wound healing assays. RESULTS: Inhibition of NLRC5 expression promotes the development of ectopic lesions in mice with endometriosis, upregulates the expression of pro-inflammatory factors TNF-α and IL-6, and downregulates the expression of anti-inflammatory factor IL-10. The high expression of NLRC5 in endometriosis depended on the ERß overexpression. And ERß promoted the migration of hESCs partially depend on inflammatory microenvironment. Lastly, NLRC5 overexpression inhibited ERß-mediated development and inflammatory response of endometriosis. CONCLUSIONS: Our results suggest that the innate immune molecule NLRC5-mediated anti-inflammation participates in ERß-mediated endometriosis development, and partly clarifies the pathological mechanism of endometriosis, expanding our knowledge of the specific molecules related to the inflammatory response involved in endometriosis and potentially providing a new therapeutic target for endometriosis.


Subject(s)
Endometriosis , Estrogen Receptor beta , Intracellular Signaling Peptides and Proteins , Adult , Animals , Female , Humans , Mice , Disease Models, Animal , Endometriosis/metabolism , Endometriosis/pathology , Endometriosis/genetics , Endometrium/metabolism , Endometrium/pathology , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Immunohistochemistry , Inflammation , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism
7.
Proc Natl Acad Sci U S A ; 121(38): e2405474121, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39255000

ABSTRACT

Endometriosis negatively impacts the health-related quality of life of 190 million women worldwide. Novel advances in nonhormonal treatments for this debilitating condition are desperately needed. Macrophages play a vital role in the pathophysiology of endometriosis and represent a promising therapeutic target. In the current study, we revealed the full transcriptomic complexity of endometriosis-associated macrophage subpopulations using single-cell analyses in a preclinical mouse model of experimental endometriosis. We have identified two key lesion-resident populations that resemble i) tumor-associated macrophages (characterized by expression of Folr2, Mrc1, Gas6, and Ccl8+) that promoted expression of Col1a1 and Tgfb1 in human endometrial stromal cells and increased angiogenic meshes in human umbilical vein endothelial cells, and ii) scar-associated macrophages (Mmp12, Cd9, Spp1, Trem2+) that exhibited a phenotype associated with fibrosis and matrix remodeling. We also described a population of proresolving large peritoneal macrophages that align with a lipid-associated macrophage phenotype (Apoe, Saa3, Pid1) concomitant with altered lipid metabolism and cholesterol efflux. Gain of function experiments using an Apoe mimetic resulted in decreased lesion size and fibrosis, and modification of peritoneal macrophage populations in the preclinical model. Using cross-species analysis of mouse and human single-cell datasets, we determined the concordance of peritoneal and lesion-resident macrophage subpopulations, identifying key similarities and differences in transcriptomic phenotypes. Ultimately, we envisage that these findings will inform the design and use of specific macrophage-targeted therapies and open broad avenues for the treatment of endometriosis.


Subject(s)
Endometriosis , Macrophages , Single-Cell Analysis , Female , Single-Cell Analysis/methods , Animals , Humans , Endometriosis/metabolism , Endometriosis/pathology , Endometriosis/genetics , Mice , Macrophages/metabolism , Phenotype , Endometrium/metabolism , Endometrium/pathology , Disease Models, Animal , Human Umbilical Vein Endothelial Cells/metabolism , Transcriptome
8.
J Obstet Gynaecol Res ; 50(10): 1771-1784, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39293995

ABSTRACT

AIM: Endometriosis is a complex, multifactorial disease. Recent advances in molecular biology underscore that somatic mutations within the epithelial component of the normal endometrium, alongside aberrant epigenetic alterations within endometrial stromal cells, may serve as stimulators for the proliferation of endometriotic tissue within the peritoneal cavity. Nevertheless, pivotal inquiries persist: the deterministic factors driving endometriosis development in certain women while sparing others, notwithstanding comparable experiences of retrograde menstruation. Within this review, we endeavor to synopsize the current understanding of diverse pathophysiologic mechanisms underlying the initiation and progression of endometriosis and delineate avenues for future research. METHODS: A literature search without time restriction was conducted utilizing PubMed and Google Scholar. RESULTS: Given that aberrant clonal expansion stemming from cancer-associated mutations is common in normal endometrial tissue, only endometrial cells harboring mutations imparting proliferative advantages may be selected for survival outside the uterus. Endometriotic cells capable of engendering metabolic plasticity and modulating mitochondrial dynamics, thereby orchestrating responses to hypoxia, oxidative stress, inflammation, hormonal stimuli, and immune surveillance, and adeptly acclimating to their harsh surroundings, stand a chance at viability. CONCLUSION: The genesis of endometriosis appears to reflect the evolutionary principles of mutation, selection, clonal expansion, and adaptation to the environment.


Subject(s)
Endometriosis , Epigenesis, Genetic , Endometriosis/genetics , Endometriosis/metabolism , Humans , Female , Gene-Environment Interaction
9.
Nat Genet ; 56(9): 1925-1937, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39198675

ABSTRACT

The complex and dynamic cellular composition of the human endometrium remains poorly understood. Previous endometrial single-cell atlases profiled few donors and lacked consensus in defining cell types. We introduce the Human Endometrial Cell Atlas (HECA), a high-resolution single-cell reference atlas (313,527 cells) combining published and new endometrial single-cell transcriptomics datasets of 63 women with and without endometriosis. HECA assigns consensus and identifies previously unreported cell types, mapped in situ using spatial transcriptomics and validated using a new independent single-nuclei dataset (312,246 nuclei, 63 donors). In the functionalis, we identify intricate stromal-epithelial cell coordination via transforming growth factor beta (TGFß) signaling. In the basalis, we define signaling between fibroblasts and an epithelial population expressing progenitor markers. Integration of HECA with large-scale endometriosis genome-wide association study data pinpoints decidualized stromal cells and macrophages as most likely dysregulated in endometriosis. The HECA is a valuable resource for studying endometrial physiology and disorders, and for guiding microphysiological in vitro systems development.


Subject(s)
Endometriosis , Endometrium , Single-Cell Analysis , Humans , Female , Endometrium/metabolism , Endometrium/cytology , Single-Cell Analysis/methods , Endometriosis/genetics , Endometriosis/pathology , Endometriosis/metabolism , Transcriptome , Stromal Cells/metabolism , Epithelial Cells/metabolism , Genome-Wide Association Study , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/genetics , Gene Expression Profiling/methods , Signal Transduction/genetics , Fibroblasts/metabolism
10.
Reprod Biol Endocrinol ; 22(1): 100, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39118090

ABSTRACT

BACKGROUND: Patients with endometriosis suffer with chronic pelvic pain and infertility, and from the lack of pharmacologic therapies that consistently halt disease progression. Differences in the endometrium of patients with endometriosis vs. unaffected controls are well-documented. Specifically, shed endometrial tissues (delivered to the pelvic cavity via retrograde menstruation) reveal that a subset of stromal cells exhibiting pro-inflammatory, pro-fibrotic, and pro-senescence-like phenotypes is enhanced in endometriosis patients compared to controls. Additionally, cultured biopsy-derived endometrial stromal cells from endometriosis patients exhibit impaired decidualization, a defined differentiation process required for human embryo implantation and pregnancy. Quercetin, a senolytic agent, shows therapeutic potential for pulmonary fibrosis, a disorder attributed to senescent pulmonary fibroblasts. In rodent models of endometriosis, quercetin shows promise, and quercetin improves decidualization in vitro. However, the exact mechanisms are not completely understood. Therefore, we investigated the effects of quercetin on menstrual effluent-derived endometrial stromal cells from endometriosis patients and unaffected controls to define the signaling pathways underlying quercetin's effects on endometrial stromal cells. METHODS: Menstrual effluent-derived endometrial stromal cells were collected and cultured from unaffected controls and endometriosis patients and then, low passage cells were treated with quercetin (25 µM) under basal or standard decidualization conditions. Decidualization responses were analyzed by measuring the production of IGFBP1 and PRL. Also, the effects of quercetin on intracellular cAMP levels and cellular oxidative stress responses were measured. Phosphokinase arrays, western blotting, and flow cytometry methods were performed to define the effects of quercetin on various signaling pathways and the potential mechanistic roles of quercetin. RESULTS: Quercetin significantly promotes decidualization of control- and endometriosis-endometrial stromal cells. Quercetin substantially reduces the phosphorylation of multiple signaling molecules in the AKT and ERK1/2 pathways, while enhancing the phosphorylation of p53 and total p53 levels. Furthermore, p53 inhibition blocks decidualization while p53 activation promotes decidualization. Finally, we provide evidence that quercetin increases apoptosis of endometrial stromal cells with a senescent-like phenotype. CONCLUSIONS: These data provide insight into the mechanisms of action of quercetin on endometrial stromal cells and warrant future clinical trials to test quercetin and other senolytics for treating endometriosis.


Subject(s)
Cellular Senescence , Endometriosis , Proto-Oncogene Proteins c-akt , Quercetin , Stromal Cells , Tumor Suppressor Protein p53 , Quercetin/pharmacology , Female , Humans , Endometriosis/metabolism , Endometriosis/pathology , Endometriosis/drug therapy , Proto-Oncogene Proteins c-akt/metabolism , Adult , Stromal Cells/drug effects , Stromal Cells/metabolism , Cellular Senescence/drug effects , Tumor Suppressor Protein p53/metabolism , Endometrium/drug effects , Endometrium/metabolism , Endometrium/pathology , Decidua/drug effects , Decidua/metabolism , Signal Transduction/drug effects , MAP Kinase Signaling System/drug effects , Cells, Cultured
11.
Int J Mol Sci ; 25(16)2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39201680

ABSTRACT

Endometriosis is one of the most common causes of chronic pelvic pain and infertility that affects 10% of women of reproductive age. It is currently defined as the presence of endometrial epithelial and stromal cells at ectopic sites; however, advances in endometriosis research have some authors believing that endometriosis should be re-defined as "a fibrotic condition in which endometrial stroma and epithelium can be identified". microRNAs (miRNAs) are regulatory molecules that potentially play a role in endometriotic lesion development. There is evidence that suggests that miRNAs, including microRNA-21 (miR-21), participate in fibrotic processes in different organs, including the heart, kidney, liver and lungs. The objective of this study was to understand the role of miR-21 and the mechanisms that can contribute to the development of fibrosis by determining how IL-6 regulates miR-21 expression and how this miRNA regulates the transforming growth factor beta (TGF-ß) signaling pathway to promote fibrosis. We investigated the expression of miR-21 in the baboon and mouse model of endometriosis and its correlation with fibrosis. We demonstrated that inflammation and fibrosis are present at a very early stage of endometriosis and that the inflammatory environment in the peritoneal cavity, which includes interleukin 6 (IL-6), can regulate the expression of miR-21 in vitro and in vivo.


Subject(s)
Endometriosis , Fibrosis , Interleukin-6 , MicroRNAs , MicroRNAs/genetics , MicroRNAs/metabolism , Female , Endometriosis/genetics , Endometriosis/metabolism , Endometriosis/pathology , Animals , Interleukin-6/metabolism , Interleukin-6/genetics , Mice , Humans , Gene Expression Regulation , Papio , Endometrium/metabolism , Endometrium/pathology , Signal Transduction , Disease Models, Animal , Transforming Growth Factor beta/metabolism
12.
J Reprod Immunol ; 165: 104312, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39094215

ABSTRACT

BACKGROUND: Endometriosis (EMs) is a chronic disease characterized by endometrial-like tissue present outside of the uterus. Macrophages have been confirmed to participate in the development of EMs. Integrin ß3 (ITGB3), a ß-subunit of the integrin family, is crucial in tumor progression. In this study, we investigated the pivotal role of ITGB3 in endometrial stromal cells (ESCs) and its influence on the development of EMs, particularly focusing on the regulatory impact of macrophages. METHODS: In this study, we used western blot, Real-time qPCR, Immunohistochemistry to detected the high expression of ITGB3 in ESCs. ITGB3-overexpression ESCs (ITGB3-OE) was constructed and detected by RNA-seq with normal ESCs. ATP and lactate expression assay, transwell migration assay, wound healing, cell adhesion assay and other molecular biology techniques were used to explore the potential mechanisms. In vivo, we constructed the EMs mouse model and injected with cilengitite to inhibit ITGB3. RESULTS: Here, we found ITGB3 highly expressed in ectopic lesions in EMs. The increasing ITGB3 resulted in activating the glycolysis, which produced more ATP and lactate in ITGB3-OE. After culturing with lactate, the migration, proliferation and invasion ability of ESCs were enhanced, while the result in 2-DG was reversed. In vivo, the results showed that after antagonizing ITGB3, the number of ectopic lesions was decrease. CONCLUSIONS: Our findings indicate that ITGB3 up-regulated by macrophages are able to regulate the glycolysis to promote the development of EMs and lactate enhances the ability of proliferation, migration, invasion and adhesion of EMs iv vivo and in vitro.


Subject(s)
Endometriosis , Glycolysis , Integrin beta3 , Lactic Acid , Animals , Female , Humans , Mice , Cell Movement , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Endometriosis/pathology , Endometriosis/metabolism , Endometrium/pathology , Endometrium/metabolism , Integrin beta3/metabolism , Integrin beta3/genetics , Lactic Acid/metabolism , Macrophages/metabolism , Macrophages/immunology , Stromal Cells/metabolism , Stromal Cells/pathology
13.
J Ethnopharmacol ; 335: 118659, 2024 Dec 05.
Article in English | MEDLINE | ID: mdl-39098622

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Endometriosis (EMS) is a common gynecological disease that causes dysmenorrhea, chronic pelvic pain and infertility. Luoshi Neiyi Prescription (LSNYP), a traditional Chinese medicine (TCM) formula, is used to relieve EMS in the clinic. AIMS: This study aimed to examine the active components of LSNYP and the possible mechanism involved in its treatment of EMS. MATERIALS AND METHODS: Ultrahigh-performance liquid chromatography quadrupole time-of-flight mass spectrometry (UPLC-Q/TOF-MS) was used to identify the chemical components of LSNYP. Human primary ectopic endometrial stromal cells (ecESCs) and eutopic endometrial stromal cells (euESCs) were isolated, and the expression levels of hypoxia inducible factor 1A (HIF1A), enhancer of zeste homolog 2 (EZH2) and steroidogenic factor 1 (SF-1) were detected by immunofluorescence and qPCR. Cobalt chloride (CoCl2) was utilized to construct an in vitro hypoxic environment, and lentiviruses were engineered to downregulate HIF1A and EZH2 and upregulate EZH2. Subsequently, the expression levels of HIF1A, EZH2, and SF-1 were measured using qPCR or western blotting. The binding of EZH2 to the SF-1 locus in ESCs was examined via ChIP. Furthermore, the effects of LSNYP on the HIF1A/EZH2/SF-1 pathway were evaluated both in vitro and in vivo. RESULTS: A total of 185 components were identified in LSNYP. The protein and gene expression levels of HIF1A and SF-1 were increased, whereas those of EZH2 were decreased in ecESCs. After treating euESCs with 50 µmol L-1 CoCl2 for 24 h, cell viability and estradiol (E2) production were enhanced. Hypoxia decreased EZH2 protein expression, while si-HIF1A increased it. SF-1 was increased when EZH2 was downregulated in normal and hypoxic environments, whereas the overexpression of EZH2 led to a decrease in SF-1 expression. ChIP revealed that hypoxia reduced EZH2 binding to the SF-1 locus in euESCs. In vitro, LSNYP-containing serum decreased E2 and prostaglandin E2 (PGE2) production, inhibited cell proliferation and invasion, and reduced the expression of HIF1A, SF-1, steroidogenic acute regulatory protein (StAR), and aromatase cytochrome P450 (P450arom). In vivo, LSNYP suppressed inflammation and adhesion and inhibited the HIF1A/EZH2/SF-1 pathway in endometriotic tissues. CONCLUSIONS: LSNYP may exert pharmacological effects on EMS by inhibiting E2 synthesis and inflammation through regulation of the HIF1A/EZH2/SF-1 pathway. These results suggest that LSNYP may be a promising candidate for the treatment of EMS.


Subject(s)
Drugs, Chinese Herbal , Endometriosis , Enhancer of Zeste Homolog 2 Protein , Estradiol , Hypoxia-Inducible Factor 1, alpha Subunit , Adult , Animals , Female , Humans , Mice , Cells, Cultured , Drugs, Chinese Herbal/pharmacology , Endometriosis/drug therapy , Endometriosis/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Estradiol/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Inflammation/drug therapy , Signal Transduction/drug effects , Steroidogenic Factor 1/genetics , Steroidogenic Factor 1/metabolism , Stromal Cells/drug effects , Stromal Cells/metabolism
14.
Eur J Histochem ; 68(3)2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39105608

ABSTRACT

Endometriosis is an estrogen-dependent inflammatory gynecological disease whose pathogenesis is unclear. C-C motif chemokine ligand 18 (CCL18), a chemokine, is involved in several inflammatory diseases. In this study, we aimed to investigate the role of CCL18 in endometriosis and its underlying mechanisms. Human endometrium and peritoneal fluid were obtained from women with and without endometriosis for molecular studies. The expression level of CCL18 in each tissue sample was examined by RNA sequencing analysis, quantitative PCR analysis and immunohistochemistry staining. The effects of CCL18 on cell migration, tube formation and neurite growth were investigated in vitro using primary endometrial cells, human umbilical vein endothelial cells (HUVECs) and dorsal root ganglion (DRG) neurons, respectively. Moreover, the development of endometriosis in mice was studied in vivo by blocking CCL18. CCL18 was shown to be overexpressed in endometrial foci and peritoneal fluid in women with endometriosis and was positively correlated with endometriosis pain. In vitro, CCL18 promoted the migration of ectopic endometrial cells, tube formation of HUVECs, and nerve outgrowth of DRG neurons. More importantly, inhibition of CCL18 significantly suppressed lesion development, angiogenesis, and nerve infiltration in a mouse model of endometriosis. In conclusion, CCL18 may play a role in the progression of endometriosis by increasing endometrial cell migration and promoting neuroangiogenesis.


Subject(s)
Cell Movement , Chemokines, CC , Endometriosis , Endometrium , Human Umbilical Vein Endothelial Cells , Neovascularization, Pathologic , Endometriosis/metabolism , Endometriosis/pathology , Female , Humans , Animals , Endometrium/metabolism , Endometrium/pathology , Human Umbilical Vein Endothelial Cells/metabolism , Mice , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Chemokines, CC/metabolism , Adult , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Ascitic Fluid/metabolism , Ascitic Fluid/pathology , Mice, Inbred C57BL
15.
Front Immunol ; 15: 1396000, 2024.
Article in English | MEDLINE | ID: mdl-39192982

ABSTRACT

Endometriosis is a chronic inflammatory disease that causes debilitating pelvic pain in women. Macrophages are considered to be key players in promoting disease progression, as abundant macrophages are present in ectopic lesions and elevated in the peritoneum. In the present study, we examined the role of GATA6+ peritoneal macrophages on endometriosis-associated hyperalgesia using mice with a specific myeloid deficiency of GATA6. Lesion induction induced the disappearance of TIM4hi MHCIIlo residential macrophages and the influx of increased Ly6C+ monocytes and TIM4lo MHCIIhi macrophages. The recruitment of MHCIIhi inflammatory macrophages was extensive in Mac Gata6 KO mice due to the severe disappearance of TIM4hi MHCIIlo residential macrophages. Ki67 expression confirmed GATA6-dependent proliferative ability, showing different proliferative phenotypes of TIM4+ residential macrophages in Gata6f/f and Mac Gata6 KO mice. Peritoneal proinflammatory cytokines were elevated after lesion induction. When cytokine levels were compared between Gata6f/f and Mac Gata6 KO mice, TNFα at day 21 in Gata6f/f mice was higher than in Mac Gata6 KO mice. Lesion induction increased both abdominal and hind paw sensitivities. Gata6f/f mice tended to show higher sensitivity in the abdomen after day 21. Elevated expression of TRPV1 and CGRP was observed in the dorsal root ganglia after ELL induction in Gata6f/f mice until days 21 and 42, respectively. These results support that peritoneal GATA6+ macrophages are involved in the recruitment and reprogramming of monocyte-derived macrophages. The extensive recruitment of monocyte-derived macrophages in Mac Gata6 KO mice might protect against inflammatory stimuli during the resolution phase, whereas GATA6 deficiency did not affect lesion initiation and establishment at the acute phase of inflammation. GATA6+ residential macrophages act to sustain local inflammation in the peritoneum and sensitivities in the neurons, reflecting endometriosis-associated hyperalgesia.


Subject(s)
Endometriosis , GATA6 Transcription Factor , Macrophages, Peritoneal , Animals , Female , Mice , Cytokines/metabolism , Disease Models, Animal , Endometriosis/immunology , Endometriosis/pathology , Endometriosis/metabolism , Ganglia, Spinal/metabolism , Ganglia, Spinal/immunology , GATA6 Transcription Factor/metabolism , GATA6 Transcription Factor/genetics , Hyperalgesia/etiology , Hyperalgesia/metabolism , Hyperalgesia/immunology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Mice, Inbred C57BL , Mice, Knockout , Peritoneum/pathology , Peritoneum/immunology , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics
16.
Genes (Basel) ; 15(8)2024 Aug 17.
Article in English | MEDLINE | ID: mdl-39202445

ABSTRACT

Endometriosis is a chronic hormone-dependent disease characterized by the spread of endometrial cells outside the uterus, which form endometriotic lesions and disrupt the functions of the affected organs. The etiopathogenesis of endometriosis is still unclear, and thus it is important to examine the genes that may contribute to the establishment of endometriotic lesions. The aim of this study was to investigate the expression of new potential candidate gene latexin (LXN), an inhibitor of carboxypeptidases, in endometrium and endometriotic lesions to elucidate its possible role in endometriosis development. LXN expression in tissues was assessed using quantitative reverse transcription PCR (qRT-PCR) analysis and immunohistochemical staining (IHC). The functions of LXN were examined using Transwell and MTT assays. qRT-PCR analysis revealed that LXN expression in endometrium was menstrual cycle-dependent, being lowest in the early-secretory phase and highest in the late-secretory phase and was significantly upregulated in endometriotic lesions. IHC confirmed LXN expression in endometrial stromal cells, and in vitro assays demonstrated that knockdown of LXN effectively reduced the migratory capacity of endometrial stromal cells while promoting cell viability. In conclusion, our results showed that LXN can be involved in the pathogenesis of endometriosis by regulating the proliferation and migration activity of endometriotic stromal cells.


Subject(s)
Endometriosis , Endometrium , Menstrual Cycle , Up-Regulation , Humans , Female , Endometriosis/genetics , Endometriosis/metabolism , Endometriosis/pathology , Endometrium/metabolism , Endometrium/pathology , Menstrual Cycle/genetics , Menstrual Cycle/metabolism , Adult , Stromal Cells/metabolism , Stromal Cells/pathology , Cell Movement/genetics , Cell Proliferation , Carboxypeptidases/genetics , Carboxypeptidases/metabolism
17.
Int J Mol Sci ; 25(15)2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39125713

ABSTRACT

Endometriosis, often associated with chronic pelvic pain, can lead to anxiety and depression. This study investigates the role and mechanism of Glycine receptor alpha 3 (Glrα3) in the central sensitization of pain in endometriosis, aiming to identify new therapeutic targets. Using a Glrα3 knockout mouse model of endometriosis, we employed behavioral tests, qPCR, immunofluorescence, Nissl staining, MRI, and Western blot to assess the involvement of Glrα3 in central pain sensitization. Our results indicate that endometriosis-induced hyperalgesia and anxiety-depressive-like behaviors are linked to increased Glrα3 expression. Chronic pain in endometriosis leads to gray matter changes in the sensory and insular cortices, with Glrα3 playing a significant role. The inhibition of Glrα3 alleviates pain, reduces neuronal abnormalities, and decreases glial cell activation. The absence of Glrα3 effectively regulates the central sensitization of pain in endometriosis by inhibiting glial cell activation and maintaining neuronal stability. This study offers new therapeutic avenues for the clinical treatment of endometriosis-related pain.


Subject(s)
Endometriosis , Mice, Knockout , Animals , Female , Mice , Anxiety , Chronic Pain/metabolism , Chronic Pain/etiology , Chronic Pain/pathology , Chronic Pain/genetics , Disease Models, Animal , Endometriosis/metabolism , Endometriosis/pathology , Endometriosis/complications , Endometriosis/genetics , Hyperalgesia/metabolism , Hyperalgesia/etiology , Mice, Inbred C57BL , Pelvic Pain/etiology , Pelvic Pain/metabolism , Receptors, Glycine/genetics , Receptors, Glycine/metabolism
18.
Int J Mol Sci ; 25(15)2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39125716

ABSTRACT

In patients with endometriosis, refluxed endometrial fragments evade host immunosurveillance, developing into endometriotic lesions. However, the mechanisms underlying this evasion have not been fully elucidated. N-Myc and STAT Interactor (NMI) have been identified as key players in host immunosurveillance, including interferon (IFN)-induced cell death signaling pathways. NMI levels are markedly reduced in the stromal cells of human endometriotic lesions due to modulation by the Estrogen Receptor beta/Histone Deacetylase 8 axis. Knocking down NMI in immortalized human endometrial stromal cells (IHESCs) led to elevated RNA levels of genes involved in cell-to-cell adhesion and extracellular matrix signaling following IFNA treatment. Furthermore, NMI knockdown inhibited IFN-regulated canonical signaling pathways, such as apoptosis mediated by Interferon Stimulated Gene Factor 3 and necroptosis upon IFNA treatment. In contrast, NMI knockdown with IFNA treatment activated non-canonical IFN-regulated signaling pathways that promote proliferation, including ß-Catenin and AKT signaling. Moreover, NMI knockdown in IHESCs stimulated ectopic lesions' growth in mouse endometriosis models. Therefore, NMI is a novel endometriosis suppressor, enhancing apoptosis and inhibiting proliferation and cell adhesion of endometrial cells upon IFN exposure.


Subject(s)
Apoptosis , Endometriosis , Signal Transduction , Animals , Female , Humans , Mice , Apoptosis/genetics , Cell Adhesion/genetics , Cell Proliferation , Endometriosis/metabolism , Endometriosis/pathology , Endometriosis/genetics , Endometrium/metabolism , Endometrium/pathology , Interferons/metabolism , Intracellular Signaling Peptides and Proteins , Stromal Cells/metabolism
19.
Front Endocrinol (Lausanne) ; 15: 1372221, 2024.
Article in English | MEDLINE | ID: mdl-39149122

ABSTRACT

Background: Endometriosis (EM) is a prevalent gynecological disorder frequently associated with irregular menstruation and infertility. Programmed cell death (PCD) is pivotal in the pathophysiological mechanisms underlying EM. Despite this, the precise pathogenesis of EM remains poorly understood, leading to diagnostic delays. Consequently, identifying biomarkers associated with PCD is critical for advancing the diagnosis and treatment of EM. Methods: This study used datasets from the Gene Expression Omnibus (GEO) to identify differentially expressed genes (DEGs) following preprocessing. By cross-referencing these DEGs with genes associated with PCD, differentially expressed PCD-related genes (DPGs) were identified. Enrichment analyses for KEGG and GO pathways were conducted on these DPGs. Additionally, Mendelian randomization and machine learning techniques were applied to identify biomarkers strongly associated with EM. Results: The study identified three pivotal biomarkers: TNFSF12, AP3M1, and PDK2, and established a diagnostic model for EM based on these genes. The results revealed a marked upregulation of TNFSF12 and PDK2 in EM samples, coupled with a significant downregulation of AP3M1. Single-cell analysis further underscored the potential of TNFSF12, AP3M1, and PDK2 as biomarkers for EM. Additionally, molecular docking studies demonstrated that these genes exhibit significant binding affinities with drugs currently utilized in clinical practice. Conclusion: This study systematically elucidated the molecular characteristics of PCD in EM and identified TNFSF12, AP3M1, and PDK2 as key biomarkers. These findings provide new directions for the early diagnosis and personalized treatment of EM.


Subject(s)
Biomarkers , Endometriosis , Machine Learning , Mendelian Randomization Analysis , Humans , Endometriosis/genetics , Endometriosis/diagnosis , Endometriosis/metabolism , Female , Biomarkers/metabolism , Apoptosis/genetics , Gene Expression Profiling , Molecular Docking Simulation , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/genetics , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism
20.
J Ovarian Res ; 17(1): 160, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39103867

ABSTRACT

Extracellular vesicles (EVs), particles enriched in bioactive molecules like proteins, nucleic acids, and lipids, are crucial mediators of intercellular communication and play key roles in various physiological and pathological processes. EVs have been shown to be involved in ovarian follicular function and to be altered in two prevalent gynecological disorders; polycystic ovarian syndrome (PCOS) and endometriosis.Ovarian follicles are complex microenvironments where folliculogenesis takes place with well-orchestrated interactions between granulosa cells, oocytes, and their surrounding stromal cells. Recent research unveiled the presence of EVs, including exosomes and microvesicles, in the follicular fluid (FFEVs), which constitutes part of the developing oocyte's microenvironment. In the context of PCOS, a multifaceted endocrine, reproductive, and metabolic disorder, studies have explored the dysregulation of these FFEVs and their cargo. Nine PCOS studies were included in this review and two miRNAs were commonly reported in two different studies, miR-379 and miR-200, both known to play a role in female reproduction. Studies have also demonstrated the potential use of EVs as diagnostic tools and treatment options.Endometriosis, another prevalent gynecological disorder characterized by ectopic growth of endometrial-like tissue, has also been linked to aberrant EV signaling. EVs in the peritoneal fluid of women with endometriosis carry molecules that modulate the immune response and promote the establishment and maintenance of endometriosis lesions. EVs derived from endometriosis lesions, serum and peritoneal fluid obtained from patients with endometriosis showed no commonly reported biomolecules between the eleven reviewed studies. Importantly, circulating EVs have been shown to be potential biomarkers, also reflecting the severity of the pathology.Understanding the interplay of EVs within human ovarian follicles may provide valuable insights into the pathophysiology of both PCOS and endometriosis. Targeting EV-mediated communication may open avenues for novel diagnostic and therapeutic approaches for these common gynecological disorders. More research is essential to unravel the mechanisms underlying EV involvement in folliculogenesis and its dysregulation in PCOS and endometriosis, ultimately leading to more effective and personalized interventions.


Subject(s)
Endometriosis , Extracellular Vesicles , Polycystic Ovary Syndrome , Humans , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/pathology , Endometriosis/metabolism , Endometriosis/pathology , Female , Extracellular Vesicles/metabolism , Follicular Fluid/metabolism , MicroRNAs/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL