Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Pathol ; 260(3): 248-260, 2023 07.
Article in English | MEDLINE | ID: mdl-36992523

ABSTRACT

Endometriosis is a common gynecological disorder that causes female infertility. Our recent research found that excessive oxidative stress in ovaries of endometriosis patients induced senescence of cumulus granulosa cells. Here, we analyzed the transcriptomic and metabolomics profiles of follicles in a mouse model of endometriosis and in patients with endometriosis and investigated the potential function of changed metabolites in granulosa cells. RNA-sequencing indicated that both endometriosis lesions and oxidative stress in mice induced abnormalities of reactive oxidative stress, steroid hormone biosynthesis, and lipid metabolism. The mouse model and women with endometriosis showed altered lipid metabolism. Nontargeted metabolite profiling of follicular fluid from endometriosis and male-factor infertility patients by liquid chromatography mass spectrometry identified 55 upregulated and 67 downregulated metabolites. These differential metabolites were mainly involved in steroid hormone biosynthesis and glycerophospholipid metabolism. Phosphatidylinositol (PI 16:0/18:2) was significantly elevated in follicular fluid from endometriosis patients compared with controls (p < 0.05), while lysophosphatidylinositol (LPI 18:2, 20:2, 18:1, 20:3 and 18:3) was reduced (p < 0.05). Upregulated PI and downregulated LPI correlated with oocyte retrieval number and mature oocyte number. LPI inhibited cellular reactive oxidative stress induced by hemin in granulosa cells. Cell proliferation inhibition, senescence, and apoptosis induced by hemin were partially reversed by LPI. Moreover, LPI administration rescued hemin blocking of cumulus-oocyte complex expansion and stimulated expression of ovulation-related genes. Transcriptomic Switching mechanism at 5' end of the RNA transcript sequencing and western blot revealed that LPI effects on granulosa cells were associated with its regulation of MAPK-ERK1/2 signaling, which was suppressed in the presence of hemin. In conclusion, our results revealed the dysregulation of lipid metabolism in endometriotic follicles. LPI may represent a novel agent for in vitro follicular culture that reverses the excessive oxidative stress from endometriotic lesions. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
Endometriosis , Infertility , Humans , Female , Male , Animals , Mice , Endometriosis/metabolism , Transcriptome , Hemin/metabolism , Metabolomics , Infertility/complications , Lipid Metabolism , RNA/metabolism , Steroids , Hormones
2.
Endocrinology ; 164(2)2022 12 19.
Article in English | MEDLINE | ID: mdl-36524678

ABSTRACT

The mechanism by which endometriosis, a common gynecological disease characterized by chronic pelvic pain and infertility, causes infertility remains elusive. Luteinized unruptured follicle syndrome, the most common type of ovulatory dysfunction, is a cause of endometriosis-associated infertility involving reduced numbers of retrieved and mature oocytes. Ovulation is controlled by luteinizing hormone and paracrine signals produced within the follicle microenvironment. Generally, interleukin (IL)-1ß is elevated in endometriosis follicular fluid, whereby it amplifies ovulation signals by activating extracellular-regulated kinase 1/2 and CCAAT/enhancer binding protein ß pathways. However, this amplification of ovulation by IL-1ß does not occur in patients with endometriosis. To illuminate the mechanism of ovulatory dysfunction in endometriosis, we analyzed the effect of oxidative stress and IL-1ß expression on endometriosis follicles. We found that oxidative stress decreased EZH2 expression and reduced H3K27Me3 levels in endometriosis ovarian granulosa cells (GCs). Selective Ezh2 depletion in mice ovarian GCs reduced fertility by disturbing cumulus-oocyte complex expansion and reducing epidermal growth factor-like factor expression. Gene expression and H3K27Me3 ChIP-sequencing (ChIP-Seq) of GCs revealed IL-1 receptor 2 (IL-1R2), a high-affinity IL-1ß-receptor that suppresses IL-1ß-mediated inflammatory cascades during ovulation, as a crucial target gene of the EZH2-H3K27Me3 axis. Moreover, IL-1ß addition did not restore ovulation upon Ezh2 knockdown, indicating a vital function of IL-1R2 in endometriosis. Thus, our findings show that reducing EZH2 and H3K27Me3 in GCs suppressed ovulatory signals by increasing IL-1R2 expression, which may ultimately contribute to endometriosis-associated infertility.


Subject(s)
Endometriosis , Infertility, Female , Animals , Female , Mice , Endometriosis/complications , Endometriosis/genetics , Endometriosis/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Granulosa Cells/metabolism , Histones/metabolism , Infertility, Female/genetics , Infertility, Female/metabolism , Receptors, Interleukin-1 Type II/genetics , Receptors, Interleukin-1 Type II/metabolism , Humans
3.
J Reprod Immunol ; 152: 103649, 2022 08.
Article in English | MEDLINE | ID: mdl-35714422

ABSTRACT

Endometriosis (EMs) is a common gynecological disease whose pathogenesis remains unclear. Immunological factors have been a key hotspot in recent years. Peritoneal fluid samples from women with EMs show defectively activated macrophages (MΦs) and strong NOD-like receptor family pyrin domain containing 3 (NLRP3) expression. Activated MΦs secrete interleukin 1ß, which stimulates migration of endometrial stromal cells (ESCs) and promotes accumulation of extracellular matrix. Levels of interleukin 1ß in peritoneal fluid were significantly higher in patients with stage III-IV EMs compared with stage I-II EMs. We also found that the size and weight of endometrial lesions in NLRP3-/- mice were significantly lower than those of wild-type mice, and this phenomenon was reversed by intraperitoneally injecting peritoneal MΦs derived from wild-type mice. Moreover, we observed that the NLRP3 inflammasome was activated in MΦs by crosstalk between MΦs and ESCs. Targeted inhibition of NLRP3 significantly reduced lesion development in vivo and suppressed the migration ability of ESCs in vitro. Collectively, these findings suggest that the occurrence of EMs may be associated with the interaction between MΦs and ESCs.


Subject(s)
Endometriosis , Animals , Female , Interleukin-1beta/metabolism , Macrophages/metabolism , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Signal Transduction , Stromal Cells
4.
Mol Med Rep ; 24(3)2021 Sep.
Article in English | MEDLINE | ID: mdl-34278456

ABSTRACT

The pathological expression and function of lactate dehydrogenase A (LDHA), a key enzyme that converts pyruvate into lactic acid during glycolysis, remains unknown in endometriosis. In the present study, LDHA expression in tissue samples was determined by immunohistochemistry. To examine whether LDHA was induced by hypoxia, primary cultured endometrial stromal cells (ESCs) and glandular epithelial Ishikawa cells were exposed to 1% O2 (hypoxia) or 21% O2 (normoxia). Cellular functions were assessed by flow cytometry, Transwell and Cell Counting Kit­8 assays in LDHA­silenced ESCs and Ishikawa cells. Mitochondrial functions were evaluated using mitochondrial membrane potential JC­1 staining, reactive oxygen species flow cytometric analysis and ATP detection. Additionally, lactic acid production was examined and western blotting was used to evaluate the expression levels of proteins associated with apoptosis, cell cycle and glycolysis, as well as regulatory proteins involved in epithelial­mesenchymal transformation and glycolytic pathways. LDHA was localized to endometrial glandular cells and stromal cells. However, LDHA protein expression was higher in endometriotic lesions compared with that in normal and eutopic endometria. LDHA expression levels in ectopic glandular cells were higher during the proliferative stage compared with during the secretory stage. Hypoxia treatment of Ishikawa cells and ESCs markedly induced the mRNA and protein expression of LDHA. Silencing of LDHA expression in Ishikawa cells and THESC cells significantly promoted impaired mitochondrial function and apoptosis while inhibiting migration and glycolysis. However, it had no obvious effect on proliferation. In conclusion, the present study revealed that LDHA was highly expressed in endometriotic tissues, where it may serve a notable role in the occurrence and development of endometriosis.


Subject(s)
Apoptosis/drug effects , Endometriosis/drug therapy , Hypoxia/chemically induced , Lactate Dehydrogenase 5/metabolism , Lactate Dehydrogenase 5/pharmacology , Protective Agents/pharmacology , Adult , Cell Proliferation , Endometriosis/pathology , Endometrium/metabolism , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Female , Glycolysis , Humans , L-Lactate Dehydrogenase/metabolism , Lactate Dehydrogenase 5/genetics , Lactic Acid/metabolism , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Stromal Cells/metabolism
5.
Redox Biol ; 30: 101431, 2020 02.
Article in English | MEDLINE | ID: mdl-31972508

ABSTRACT

Endometriosis an important cause of female infertility and seriously impact physical and psychological health of patients. Endometriosis is now considered to be a public health problem that deserves in-depth investigation, especially the etiopathogenesis of endometriosis-associated infertility. We aimed to illuminate the etiopathogenesis of endometriosis-associated infertility that involve excessive oxidative stress (OS) induced pathological changes of ovary cumulus granulosa cell (GCs). Senescence-associated ß-galactosidase (SA ß-gal) activity in GCs from endometriosis patients, soluble isoform of advanced glycation end products receptor (sRAGE) expression in follicular fluid from endometriosis patients and differentially expressed senescence-associated secretory phenotype factors (IL-1ß, MMP-9, KGF and FGF basic protein) are all useful indexes to evaluate oocyte retrieval number and mature oocyte number. RNA-sequencing and bioinformatics analysis indicated senescent phenotype of endometriosis GCs and aggravated endoplasmic reticulum (ER) stress in endometriosis GCs. Targeting ER stress significantly alleviated OS-induced GCs senescence as well as mitochondrial membrane potential (MMP) and adenosine triphosphate (ATP) reduction in GCs. Moreover, melatonin administration rescued OS-enhanced ER stress, cellular senescence, and MMP and ATP abnormities of endometriosis GCs in vitro and in vivo. In conclusion, our results indicated excessive reactive oxygen species induces senescence of endometriosis GCs via arouse ER stress, which finally contributes to endometriosis-associated infertility, and melatonin may represent a novel adjuvant therapy strategy for endometriosis-associated infertility.


Subject(s)
Antigens, Neoplasm/genetics , Cumulus Cells/cytology , Endometriosis/drug therapy , Infertility, Female/drug therapy , Melatonin/administration & dosage , Mitogen-Activated Protein Kinases/genetics , Oxidative Stress/drug effects , Animals , Cell Line , Cellular Senescence/drug effects , Cumulus Cells/metabolism , Disease Models, Animal , Endometriosis/complications , Endometriosis/genetics , Female , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Infertility, Female/etiology , Infertility, Female/genetics , Melatonin/pharmacology , Mice , Ovulation Induction , Sequence Analysis, RNA
6.
Cell Death Dis ; 10(2): 144, 2019 02 13.
Article in English | MEDLINE | ID: mdl-30760709

ABSTRACT

Endometriosis is associated with benign but adversely developed cysts in the extrauterine environment. The oxidative imbalanced environment induces DNA damage and affects cell cycle progression of endometrial stromal cells (ESCs) and endometrial epithelial cells, but how endometriotic cells maintain proliferation in the presence of oxidative stress is not clear. Growing evidence has indicated that the ectopic hypoxic microenvironment and oxidative stress can stimulate the growth of endometriotic cells, which is mainly due to the increase of HIF-1α. We found that the master hypoxia-associated miRNA miR-210-3p was increased in stromal and glandular cells of ectopic lesions compared with that of eutopic and normal endometria and was consistent with the expression of HIF-1α and the local oxidative stress-induced DNA damage predictor 8-OHdG. Moreover, miR-210-3p was upregulated in ESCs and Ishikawa cells under hypoxic conditions but not in normoxic culture. Knockdown of miR-210-3p induced a G2/M arrest of ESCs and Ishikawa cells under hypoxia, while no effect was found under normoxia. BARD1 was identified as a target of miR-210-3p. BARD1 expression was decreased in endometriotic tissues compared with eutopic and normal endometria and negatively correlated with the expression of miR-210-3p. Multivariate regression analysis showed that BARD1 downregulation could serve as an indicator for endometriotic severity. Our results suggest that miR-210-3p attenuates the G2/M cell cycle checkpoint by inactivating BRCA1 complex function in response to DNA damage under hypoxia via targeting the 3' untranslated region of BARD1 mRNA. Endometriotic mouse model experiments showed that intraperitoneal injection of the miR-210-3p inhibitor or vitamin C suppressed the growth of endometriotic lesions. Together, our results demonstrate that endometriotic cells inhibit BARD1/BRCA1 function by upregulating miR-210-3p, which might be the underlying mechanism for endometriotic cell maintenance of growth in oxidative stress. Furthermore, inhibition of miR-210-3p and administration of vitamin C are promising approaches for the treatment of endometriosis.


Subject(s)
Endometriosis/genetics , Endometriosis/metabolism , MicroRNAs/metabolism , Oxidative Stress/physiology , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , 8-Hydroxy-2'-Deoxyguanosine/metabolism , Animals , Cell Cycle Checkpoints/genetics , Disease Models, Animal , Endometriosis/pathology , Female , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Oxidative Stress/genetics , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...