Search Results
You are looking at 1 - 10 of 11 items for
- Author: Yue Zhao x
- Refine by access: All content x
Search for other papers by Yue Zhao in
Google Scholar
PubMed
Search for other papers by Haoran Liu in
Google Scholar
PubMed
Search for other papers by Yang Yang in
Google Scholar
PubMed
Search for other papers by Wenqian Huang in
Google Scholar
PubMed
Search for other papers by Lan Chao in
Google Scholar
PubMed
Abnormal sperm parameters such as oligospermia, asthenospermia, and teratozoospermia result in male factor infertility. Previous studies have shown that mitochondria play an important role in human spermatozoa motility. But the related pathogenesis is far from elucidated. The aim of this study was to investigate the association between gene associated with retinoid-interferon-induced mortality 19 (GRIM19) and asthenospermia. In this study, Grim19 knockout model (Grim19+/− mouse) was created through genome engineering. We showed that compared with WT mice, the sperm count and motility of Grim19+/− mice were significantly reduced. Grim19 may contribute to sperm count and vitality by influencing the mitochondrial membrane potential, intracellular reactive oxygen species production, and increasing cell apoptosis. The spermatogenic cells of all levels in the lumen of the seminiferous tubules were sparsely arranged, and the intercellular space became larger in the testis tissue of Grim19+/− mice. The serum testosterone concentration is significantly reduced in Grim19+/− mice. The expression of steroid synthesis-related proteins STAR, CYP11A1, and HSD3B was decreased in Grim19+/− mice. To further confirm whether changes in testosterone biosynthesis were due to Grim19 downregulation, we validated this result using Leydig cells and TM3 cells. We also found that Notch signaling pathway was involved in Grim19-mediated testosterone synthesis to some extent. In conclusion, we revealed a mechanism underlying Grim19 mediated spermatozoa motility and suggested that Grim19 affected the synthesis of testosterone and steroid hormones in male mouse partly through regulating Notch signal pathways.
Search for other papers by Chen Geng in
Google Scholar
PubMed
Search for other papers by Hao-ran Liu in
Google Scholar
PubMed
Search for other papers by Yue Zhao in
Google Scholar
PubMed
Search for other papers by Yang Yang in
Google Scholar
PubMed
Search for other papers by Lan Chao in
Google Scholar
PubMed
The epithelial-to-mesenchymal transition may play a role in adenomyosis. GRIM19 expression is downregulated in adenomyotic lesions, and the effects of this downregulation in adenomyosis remain relatively unclear. In this study, we aimed to explore whether aberrant GRIM19 expression is associated with the epithelial-to-mesenchymal transition in adenomyosis and found that the expression of both GRIM19 and WT1 was low, and epithelial-to-mesenchymal transition, which included significant changes in CDH1, CDH2 and KRT8 expression, occurred in adenomyotic lesions, as confirmed by Western blotting and quantitative real-time PCR. We provided novel insights into WT1 expression in adenomyosis, revealing that WT1 expression was increased in the endometrial glands of adenomyotic lesions by immunohistochemistry. In vitro, knockdown of GRIM19 expression by small interfering RNA (siRNA) promoted the proliferation, migration and invasion of Ishikawa cells, as measured by Cell Counting Kit-8, wound healing assay and Transwell assays. Western blotting and quantitative real-time PCR confirmed that WT1 expression increased and epithelial-to-mesenchymal transition was induced, including the upregulation of CDH2 and downregulation of CDH1 and KRT8after transfecting the GRIM19 siRNA to Ishikawa cells. Furthermore, Wt1 expression was upregulated and epithelial-to-mesenchymal transition was observed, including downregulation of Cdh1 and Krt8 in Grim19 gene-knockdown mice. Upregulation of Wt1 expression in the endometrial glands of Grim19 knockdown mice was also verified by immunohistochemistry. Taken together, these results reveal that low expression of GRIM19 in adenomyosis may upregulate WT1 expression and induce epithelial-to-mesenchymal transition in the endometria, providing new insights into the pathogenesis of adenomyosis.
Search for other papers by Yue-Mao Zheng in
Google Scholar
PubMed
Search for other papers by Hui-Ying Zhao in
Google Scholar
PubMed
Search for other papers by Xiao-E Zhao in
Google Scholar
PubMed
Search for other papers by Fu-Sheng Quan in
Google Scholar
PubMed
Search for other papers by Song Hua in
Google Scholar
PubMed
Search for other papers by Xiao-Ying He in
Google Scholar
PubMed
Search for other papers by Jun Liu in
Google Scholar
PubMed
Search for other papers by Xiao-Ning He in
Google Scholar
PubMed
Search for other papers by Hui Lin in
Google Scholar
PubMed
We assessed the developmental ability of embryos cloned from porcine neural stem (NS) cells, amniotic fluid-derived stem (AFS) cells, fetal fibroblast cells, adult fibroblast, and mammary gland epithelial cells. The five cell lines were transfected with enhanced green fluorescence protein gene respectively using lipofection. NS and AFS cells were induced to differentiate in vitro. Stem cells and their differentiated cells were harvested for analysis of the markers using RT-PCR. The five cell lines were used for nuclear transfer. The two-cell stage-cloned embryos derived from each cell line were transferred into the oviducts of surrogate mothers. The results showed that both NS and AFS cells expressed POU5F1, THY1 and SOX2, and they were both induced to differentiate into astrocyte (GFAP+), oligodendrocyte (GalC+), neuron (NF+, ENO2+, and MAP2+), adipocyte (LPL+ and PPARG-D+), osteoblast (osteonectin+ and osteocalcin+), myocyte (MYF6+ and MYOD+), and endothelium (PECAM1+, CD34+, CDH5+, and NOS3+) respectively. Seven cloned fetuses (28 days and 32 days) derived from stem cells were obtained. The in vitro developmental ability (morula–blastocyst rate was 28.26–30.07%) and in vivo developmental ability (pregnancy rate were 1.67–2.17%) of the embryos cloned from stem cells were higher (P<0.05) than that of the embryos cloned from somatic cells (morula–blastocyst rate was 16.27–19.28% and pregnancy rate was 0.00%), which suggests that the undifferentiated state of the donor cells increases cloning efficiency.
Search for other papers by Hong-Jie Yuan in
Google Scholar
PubMed
Search for other papers by Zhi-Bin Li in
Google Scholar
PubMed
Search for other papers by Xin-Yue Zhao in
Google Scholar
PubMed
Search for other papers by Guang-Yi Sun in
Google Scholar
PubMed
Search for other papers by Guo-Liang Wang in
Google Scholar
PubMed
Search for other papers by Ying-Qi Zhao in
Google Scholar
PubMed
Search for other papers by Min Zhang in
Google Scholar
PubMed
Search for other papers by Jing-He Tan in
Google Scholar
PubMed
Mechanisms by which female stress and particularly glucocorticoids impair oocyte competence are largely unclear. Although one study demonstrated that glucocorticoids triggered apoptosis in ovarian cells and oocytes by activating the FasL/Fas system, other studies suggested that they might induce apoptosis through activating other signaling pathways as well. In this study, both in vivo and in vitro experiments were conducted to test the hypothesis that glucocorticoids might trigger apoptosis in oocytes and ovarian cells through activating the TNF-α system. The results showed that cortisol injection of female mice (1.) impaired oocyte developmental potential and mitochondrial membrane potential with increased oxidative stress; (2.) induced apoptosis in mural granulosa cells (MGCs) with increased oxidative stress in the ovary; and (3.) activated the TNF-α system in both ovaries and oocytes. Culture with corticosterone induced apoptosis and activated the TNF-α system in MGCs. Knockdown or knockout of TNF-α significantly ameliorated the pro-apoptotic effects of glucocorticoids on oocytes and MGCs. However, culture with corticosterone downregulated TNF-α expression significantly in oviductal epithelial cells. Together, the results demonstrated that glucocorticoids impaired oocyte competence and triggered apoptosis in ovarian cells through activating the TNF-α system and that the effect of glucocorticoids on TNF-α expression might vary between cell types.
Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
Search for other papers by Ying Huang in
Google Scholar
PubMed
Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
Search for other papers by Jiang-Man Gao in
Google Scholar
PubMed
Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
Search for other papers by Chun-Mei Zhang in
Google Scholar
PubMed
Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
Search for other papers by Hong-Cui Zhao in
Google Scholar
PubMed
Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
Search for other papers by Yue Zhao in
Google Scholar
PubMed
Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
Search for other papers by Rong Li in
Google Scholar
PubMed
Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
Search for other papers by Yang Yu in
Google Scholar
PubMed
Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
Search for other papers by Jie Qiao in
Google Scholar
PubMed
Polycystic ovary syndrome (PCOS) is a common reproductive disorder that has many characteristic features including hyperandrogenemia, insulin resistance and obesity, which may have significant implications for pregnancy outcomes and long-term health of women. Daughters born to PCOS mothers constitute a high-risk group for metabolic and reproductive derangements, but no report has described potential growth and metabolic risk factors for such female offspring. Hence, we used a mouse model of dehydroepiandrosterone (DHEA)-induced PCOS to study the mechanisms underlying the pathology of PCOS by investigating the growth, developmental characteristics, metabolic indexes and expression profiles of key genes of offspring born to the models. We found that the average litter size was significantly smaller in the DHEA group, and female offspring had sustained higher body weight, increased body fat and triglyceride content in serum and liver; they also exhibited decreased energy expenditure, oxygen consumption and impaired glucose tolerance. Genes related to glucolipid metabolism such as Pparγ, Acot1/2, Fgf21, Pdk4 and Inhbb were upregulated in the liver of the offspring in DHEA group compared with those in controls, whereas Cyp17a1 expression was significantly decreased. However, the expression of these genes was not detected in male offspring. Our results show that female offspring in DHEA group exhibit perturbed growth and glucolipid metabolism that were not observed in male offspring.
Department of Obstetrics and Gynaecology, Key Laboratory of Assisted Reproduction, Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Neuroscience Research Institute, Center for Reproductive Medicine, Peking University Third Hospital, No. 49, North Huayuan Road, Haidian District, Beijing 100191, China
Department of Obstetrics and Gynaecology, Key Laboratory of Assisted Reproduction, Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Neuroscience Research Institute, Center for Reproductive Medicine, Peking University Third Hospital, No. 49, North Huayuan Road, Haidian District, Beijing 100191, China
Search for other papers by Haolin Zhang in
Google Scholar
PubMed
Search for other papers by Ming Yi in
Google Scholar
PubMed
Department of Obstetrics and Gynaecology, Key Laboratory of Assisted Reproduction, Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Neuroscience Research Institute, Center for Reproductive Medicine, Peking University Third Hospital, No. 49, North Huayuan Road, Haidian District, Beijing 100191, China
Search for other papers by Yan Zhang in
Google Scholar
PubMed
Department of Obstetrics and Gynaecology, Key Laboratory of Assisted Reproduction, Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Neuroscience Research Institute, Center for Reproductive Medicine, Peking University Third Hospital, No. 49, North Huayuan Road, Haidian District, Beijing 100191, China
Search for other papers by Hongyan Jin in
Google Scholar
PubMed
Department of Obstetrics and Gynaecology, Key Laboratory of Assisted Reproduction, Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Neuroscience Research Institute, Center for Reproductive Medicine, Peking University Third Hospital, No. 49, North Huayuan Road, Haidian District, Beijing 100191, China
Search for other papers by Wenxin Zhang in
Google Scholar
PubMed
Department of Obstetrics and Gynaecology, Key Laboratory of Assisted Reproduction, Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Neuroscience Research Institute, Center for Reproductive Medicine, Peking University Third Hospital, No. 49, North Huayuan Road, Haidian District, Beijing 100191, China
Search for other papers by Jingjing Yang in
Google Scholar
PubMed
Department of Obstetrics and Gynaecology, Key Laboratory of Assisted Reproduction, Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Neuroscience Research Institute, Center for Reproductive Medicine, Peking University Third Hospital, No. 49, North Huayuan Road, Haidian District, Beijing 100191, China
Search for other papers by Liying Yan in
Google Scholar
PubMed
Department of Obstetrics and Gynaecology, Key Laboratory of Assisted Reproduction, Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Neuroscience Research Institute, Center for Reproductive Medicine, Peking University Third Hospital, No. 49, North Huayuan Road, Haidian District, Beijing 100191, China
Search for other papers by Rong Li in
Google Scholar
PubMed
Department of Obstetrics and Gynaecology, Key Laboratory of Assisted Reproduction, Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Neuroscience Research Institute, Center for Reproductive Medicine, Peking University Third Hospital, No. 49, North Huayuan Road, Haidian District, Beijing 100191, China
Search for other papers by Yue Zhao in
Google Scholar
PubMed
Department of Obstetrics and Gynaecology, Key Laboratory of Assisted Reproduction, Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Neuroscience Research Institute, Center for Reproductive Medicine, Peking University Third Hospital, No. 49, North Huayuan Road, Haidian District, Beijing 100191, China
Department of Obstetrics and Gynaecology, Key Laboratory of Assisted Reproduction, Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Neuroscience Research Institute, Center for Reproductive Medicine, Peking University Third Hospital, No. 49, North Huayuan Road, Haidian District, Beijing 100191, China
Search for other papers by Jie Qiao in
Google Scholar
PubMed
Polycystic ovary syndrome (PCOS) is a complex endocrine and metabolic disorder with unclear etiology and unsatisfactory management. Effects of diets on the phenotype of PCOS were not fully understood. In the present study, we applied 45 and 60% high-fat diets (HFDs) on a rat model of PCOS induced by postnatal DHEA injection. We found that both DHEA and DHEA+HFDs rats exhibited reproductive abnormalities, including hyperandrogenism, irregular cycles and polycystic ovaries. The addition of HFDs, especially 60% HFDs, exaggerated morphological changes of ovaries and a number of metabolic changes, including increased body weight and body fat content, impaired glucose tolerance and increased serum insulin levels. Results from qPCR showed that DHEA-induced increased expression of hypothalamic androgen receptor and LH receptor were reversed by the addition of 60% HFDs. In contrast, the ovarian expression of LH receptor and insulin receptor mRNA was upregulated only with the addition of 60% HFDs. These findings indicated that DHEA and DHEA+HFDs might influence PCOS phenotypes through distinct mechanisms: DHEA affects the normal function of hypothalamus–pituitary–ovarian axis through LH, whereas the addition of HFDs exaggerated endocrine and metabolic dysfunction through ovarian responses to insulin-related mechanisms. We concluded that the addition of HFDs yielded distinct phenotypes of DHEA-induced PCOS and could be used for studies on both reproductive and metabolic features of the syndrome.
Search for other papers by Jing Cong in
Google Scholar
PubMed
Search for other papers by Hong-Lu Diao in
Google Scholar
PubMed
Search for other papers by Yue-Chao Zhao in
Google Scholar
PubMed
Search for other papers by Hua Ni in
Google Scholar
PubMed
Search for other papers by Yun-Qin Yan in
Google Scholar
PubMed
Search for other papers by Zeng-Ming Yang in
Google Scholar
PubMed
It has been shown that both prostaglandin I2 (PGI2) and PGE2 are essential for mouse implantation, whereas only PGE2 is required for hamster implantation. To date, the expression and regulation of cyclooxygenase (COX) and prostaglandin E synthase (PGES), which are responsible for PGE2 production, have not been reported in the rat. The aim of this study was to examine the expression pattern and regulation of COX-1, COX-2, membrane-associated PGES-1 (mPGES-1), mPGES-2 and cytosolic PGES (cPGES) in rat uterus during early pregnancy and pseudopregnancy, and under delayed implantation. At implantation site on day 6 of pregnancy, COX-1 immunostaining was highly visible in the luminal epithelium, and COX-2 immunostaining was clearly observed in the subluminal stroma. Both mPGES-1 mRNA and protein were only observed in the subluminal stroma surrounding the implanting blastocyst at the implantation site on day 6 of pregancy , but were not seen in the inter-implantation site on day 6 of pregnancy and on day 6 of pseudopregnancy. Our data suggest that the presence of an active blastocyst is required for mPGES-1 expression at the implantation site. When pregnant rats on day 5 were treated with nimesulide for 24 h, mPGES-1 protein expression was completely inhibited. cPGES immunostaining was clearly observed in the luminal epithelium and subluminal stromal cells immediately surrounding the implanting blastocyst on day 6 of pregnancy. mPGES-2 immunostaining was clearly seen in the luminal epithelium at the implantation site. Additionally, immunostaining for prostaglandin I synthase (PGIS) was also strongly detected at the implantation site. In conclusion, our results indicate that PGE2 and PGI2 should have a very important role in rat implantation.
Institute of Embryo-Fetal Original Adult Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
Search for other papers by Wei-Bin Wu in
Google Scholar
PubMed
Search for other papers by Yue-Ying Xu in
Google Scholar
PubMed
Search for other papers by Wei-Wei Cheng in
Google Scholar
PubMed
Search for other papers by Bo Yuan in
Google Scholar
PubMed
Institute of Embryo-Fetal Original Adult Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
Search for other papers by Jiu-Ru Zhao in
Google Scholar
PubMed
Search for other papers by Yan-Lin Wang in
Google Scholar
PubMed
Institute of Embryo-Fetal Original Adult Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
Search for other papers by Hui-Juan Zhang in
Google Scholar
PubMed
Fetal growth restriction (FGR) threatens perinatal health and is correlated with increased incidence of fetal original adult diseases. Most cases of FGR were idiopathic, which were supposed to be associated with placental abnormality. Decreased circulating placental growth factor (PGF) was recognized as an indication of placental deficiency in FGR. In this study, the epigenetic regulation of PGF in FGR placentas and the involvement of PGF in modulation of trophoblast activity were investigated. The expression level of PGF in placental tissues was determined by RT-qPCR, immunohistochemistry and ELISA. DNA methylation profile of PGF gene was analyzed by bisulfite sequencing. Trophoblastic cell lines were treated with ZM-306416, an inhibitor of PGF receptor FLT1, to observe the effect of PGF/FLT1 signaling on cell proliferation and migration. We demonstrated that PGF was downregulated in placentas from FGR pregnancies compared with normal controls. The villous expression of PGF was positively correlated with placental and fetal weight. The CpG island inside PGF promoter was hypomethylated without obvious difference in both normal and FGR placentas. However, the higher DNA methylation at another CpG island downstream exon 7 of PGF was demonstrated in FGR placentas. Additionally, we found FLT1 was expressed in trophoblast cells. Inhibition of PGF/FLT1 signaling by a selective inhibitor impaired trophoblast proliferation and migration. In conclusion, our data suggested that the PGF expression was dysregulated, and disrupted PGF/FLT1 signaling in trophoblast might contribute to placenta dysfunction in FGR. Thus, our results support the significant role of PGF in the pathogenesis of FGR.
Inner Mongolia Key Laboratory of Basic Veterinary Science, Inner Mongolia Agricultural University, Hohhot, China
Search for other papers by Yufen Zhao in
Google Scholar
PubMed
Search for other papers by Boyang Yu in
Google Scholar
PubMed
Inner Mongolia Key Laboratory of Basic Veterinary Science, Inner Mongolia Agricultural University, Hohhot, China
Search for other papers by Xinyu Liu in
Google Scholar
PubMed
Inner Mongolia Key Laboratory of Basic Veterinary Science, Inner Mongolia Agricultural University, Hohhot, China
Search for other papers by Jitu Hu in
Google Scholar
PubMed
Search for other papers by Yanyan Yang in
Google Scholar
PubMed
Inner Mongolia Key Laboratory of Basic Veterinary Science, Inner Mongolia Agricultural University, Hohhot, China
Search for other papers by Erge Namei in
Google Scholar
PubMed
Inner Mongolia Key Laboratory of Basic Veterinary Science, Inner Mongolia Agricultural University, Hohhot, China
Search for other papers by Bingxue Yang in
Google Scholar
PubMed
Inner Mongolia Key Laboratory of Basic Veterinary Science, Inner Mongolia Agricultural University, Hohhot, China
Search for other papers by Yue Bai in
Google Scholar
PubMed
Search for other papers by Yinghong Qian in
Google Scholar
PubMed
Inner Mongolia Key Laboratory of Basic Veterinary Science, Inner Mongolia Agricultural University, Hohhot, China
Search for other papers by Haijun Li in
Google Scholar
PubMed
Although urokinase-type plasminogen activator (PLAU) and urokinase-type plasminogen activator receptor (PLAUR) have been reported to play key roles in ovarian function, their precise contribution to mammalian follicular development remains unclear. In this study, we first observed that PLAU and PLAUR were present in bovine granulosa cells (GCs). Following culture of granulosa cells with PLAU (0.5 ng/mL) and PLAUR antibody (10 µg/mL) separately and together for 24 or 48 h, a proliferation assay showed that interaction between PLAU and PLAUR contributes to bovine GC proliferation. To study the potential pathways involved in PLAU/PLAUR-induced cell proliferation, ELISA and Western blotting were performed. We found that PLAU significantly increased the ratio of phosphorylated to non-phosphorylated ERK1/2 through PLAUR signaling. Further treatment with U0126, a specific ERK1/2 phosphorylation inhibitor, markedly suppressed PLAU/PLAUR-induced ERK1/2 phosphorylation and cell proliferation. In addition, we found that PLAU and PLAUR significantly increased the intracellular cAMP level and the use of Rp-cAMP, a specific PKA inhibitor, prevented PLAU/PLAUR from promoting activation of the ERK1/2 pathway and GC proliferation. Therefore, the interaction between PLAU and PLAUR may be involved in accumulating cAMP signals and enabling MAPK/ERK1/2 activation, affecting GC proliferation. Here, we provide new mechanistic insights into the roles of PLAU and PLAUR on promoting bovine GC proliferation. The finding that potential cross-points between PLAU/PLAUR-induced intracellular signals affect GC proliferation will help in understanding the mechanisms regulating early follicular development.
Search for other papers by Shengxian Li in
Google Scholar
PubMed
Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
Search for other papers by Jia Qi in
Google Scholar
PubMed
Search for other papers by Yongzhen Tao in
Google Scholar
PubMed
Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
Search for other papers by Qinling Zhu in
Google Scholar
PubMed
Search for other papers by Rong Huang in
Google Scholar
PubMed
Search for other papers by Yu Liao in
Google Scholar
PubMed
Search for other papers by Jiang Yue in
Google Scholar
PubMed
Search for other papers by Wei Liu in
Google Scholar
PubMed
Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
Search for other papers by Hanting Zhao in
Google Scholar
PubMed
School of Life Science and Technology, ShanghaiTech University, Shanghai, China
Key Laboratory of Food Safety Risk Assessment, Ministry of Health, Beijing, China
Search for other papers by Huiyong Yin in
Google Scholar
PubMed
Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
Search for other papers by Yun Sun in
Google Scholar
PubMed
Polycystic ovary syndrome (PCOS) is the most common endocrine disorder in reproductive-age women usually accompanied by lipid metabolic disorders. However, it remains unknown whether arachidonic acid (AA) and its metabolites in follicular fluid (FF) were altered in PCOS patients. This study was intended to measure the levels of AA and its metabolites in the FF of non-obese PCOS patients that underwent in vitro fertilization (IVF) and to explore the possible causes of the alterations. Thirty-nine non-obese women with PCOS and 30 non-obese women without PCOS were enrolled. AA and its metabolites were measured by liquid chromatography-mass spectrometry. The levels of AA metabolites generated via cyclooxygenase-2 (COX-2) pathway and cytochrome P450 epoxygenase pathway but not lipoxygenase (LOX) pathway were significantly higher in the FF of PCOS patients. The metabolites generated via COX-2 pathway were significantly correlated with levels of testosterone and fasting insulin in serum. The in vitro study further demonstrated that insulin but not testosterone could promote the IL-1β and hCG-induced COX-2 expression and prostaglandin E2 (PGE2) secretion in primary human granulosa cells. In conclusion, there was an elevation in AA metabolites in FF of PCOS patients. Insulin played a pivotal role in the increased AA metabolites generated via COX-2, which could be interpreted as another novel molecular pathophysiological mechanism of PCOS.