Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters











Database
Language
Publication year range
1.
Reprod Biol Endocrinol ; 20(1): 4, 2022 Jan 03.
Article in English | MEDLINE | ID: mdl-34980155

ABSTRACT

BACKGROUND: Insulin resistance (IR) contributes to ovarian dysfunctions in polycystic ovarian syndrome (PCOS) patients. Serum amyloid A1 (SAA1) is an acute phase protein produced primarily by the liver in response to inflammation. In addition to its role in inflammation, SAA1 may participate in IR development in peripheral tissues. Yet, expressional regulation of SAA1 in the ovary and its role in the pathogenesis of ovarian IR in PCOS remain elusive. METHODS: Follicular fluid, granulosa cells and peripheral venous blood were collected from PCOS and non-PCOS patients with and without IR to measure SAA1 abundance for analysis of its correlation with IR status. The effects of SAA1 on its own expression and insulin signaling pathway were investigated in cultured primary granulosa cells. RESULTS: Ovarian granulosa cells were capable of producing SAA1, which could be induced by SAA1 per se. Moreover, the abundance of SAA1 significantly increased in granulosa cells and follicular fluid in PCOS patients with IR. SAA1 treatment significantly attenuated insulin-stimulated membrane translocation of glucose transporter 4 and glucose uptake in granulosa cells through induction of phosphatase and tensin homolog deleted on chromosome 10 (PTEN) expression with subsequent inhibition of Akt phosphorylation. These effects of SAA1 could be blocked by inhibitors for toll-like receptors 2/4 (TLR 2/4) and nuclear factor kappa light chain enhancer of activated B (NF-κB). CONCLUSIONS: Human granulosa cells are capable of feedforward production of SAA1, which significantly increased in PCOS patients with IR. Excessive SAA1 reduces insulin sensitivity in granulosa cells via induction of PTEN and subsequent inhibition of Akt phosphorylation upon activation of TLR2/4 and NF-κB pathway. These findings highlight that elevation of SAA1 in the ovary promotes the development of IR in granulosa cells of PCOS patients.


Subject(s)
Granulosa Cells/metabolism , Insulin Resistance/genetics , Polycystic Ovary Syndrome/genetics , Serum Amyloid A Protein/physiology , Adult , Case-Control Studies , Cells, Cultured , Female , Follicular Fluid/chemistry , Follicular Fluid/metabolism , Granulosa Cells/drug effects , Humans , Ovary/drug effects , Ovary/metabolism , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/metabolism , Serum Amyloid A Protein/genetics , Serum Amyloid A Protein/metabolism , Serum Amyloid A Protein/pharmacology
2.
Transl Res ; 227: 15-29, 2021 01.
Article in English | MEDLINE | ID: mdl-32640290

ABSTRACT

Endometriosis is a common gynecological disease in which ovarian dysfunction can be an important cause of infertility. Elevated progesterone (P4) levels during the follicular phase is possibly associated with impaired oocyte quality and pregnancy outcome in endometriosis. Beclin-1 (BECN1), an essential mediator of autophagy, has been shown to be related to the development and progression of endometriosis. This study aimed to investigate the autophagic activity in ovarian granulosa cells (GCs) of patients with endometriosis and to clarify the role of BECN1 in preovulatory P4 elevation. Our results demonstrated that serum P4/estradiol (E2) ratio and P4-to-follicle index (the average P4 secretion per follicle) on the day of human chorionic gonadotropin administration were elevated in women with ovarian endometriosis. Increased expression of BECN1 and enhanced autophagy were observed in GCs of patients with ovarian endometriomas. In cultured GCs, BECN1 knockdown reduced P4 secretion and the expression of key steroidogenic enzymes; whereas overexpression of BECN1 resulted in induced P4 production with activated biosynthesis pathway. Moreover, inhibition of autophagy by BECN1 knockdown significantly attenuated low-density lipoprotein (LDL)-induced P4 synthesis. These findings provide new insights into the role of BECN1 in late follicular P4 elevation in patients with endometriosis by promoting the degradation pathway of LDL for P4 biosynthesis via lysosome activation in GCs, and have potential therapeutic implications for the improvement of oocyte quality in women affected by endometriosis.


Subject(s)
Autophagy/physiology , Beclin-1/physiology , Endometriosis/metabolism , Granulosa Cells/metabolism , Ovarian Follicle/metabolism , Progesterone/metabolism , Cholesterol/metabolism , Female , Humans , Pregnancy , Signal Transduction , Sperm Injections, Intracytoplasmic , Up-Regulation
3.
Transl Res ; 230: 55-67, 2021 04.
Article in English | MEDLINE | ID: mdl-33129993

ABSTRACT

Polycystic ovary syndrome (PCOS) is one of the most common endocrine disorders in reproductive-age women. Reduced progesterone levels are associated with luteal phase deficiency in women with PCOS. The levels of C-X-C motif chemokine ligand-14 (CXCL14) were previously reported to be decreased in human-luteinized granulosa (hGL) cells derived from PCOS patients. However, the function of CXCL14 in hGL cells and whether CXCL14 affects the synthesis of progesterone in hGL cells remain unclear. In the present study, the levels of CXCL14 were reduced in follicular fluid and hGL cells in PCOS patients, accompanied by decreased progesterone levels in follicular fluid and decreased steroidogenic acute regulatory (STAR) expression in hGL cells. CXCL14 administration partially reversed the low progesterone production and STAR expression in hGL cells obtained from PCOS patients. In primary hGL cells, CXCL14 upregulated STAR expression and progesterone production. CXCL14 activated the phosphorylation of cyclic adenosine monophosphate response element-binding protein (CREB) and CREB inhibitor attenuated the modulation of StAR expression by CXCL14. P38 and Jun N-terminal kinase (JNK) pathways were also activated by CXCL14 and inhibition of p38 and JNK attenuated the increase of phosphorylation of CREB, STAR expression and progesterone production caused by CXCL14. Our findings revealed the novel role of CXCL14 in upregulation of STAR expression and progesterone synthesis through CREB phosphorylation via activation of p38 and JNK pathways in hGL cells. This is likely contributing to the dysfunction in steroidogenesis in granulosa cells from PCOS patients.


Subject(s)
Chemokines, CXC/pharmacology , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Phosphoproteins/metabolism , Progesterone/biosynthesis , Adult , Anthracenes/pharmacology , Cells, Cultured , Cholesterol Side-Chain Cleavage Enzyme/genetics , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Enzyme Inhibitors/pharmacology , Female , Flavonoids/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , Imidazoles/pharmacology , Phosphoproteins/genetics , Polycystic Ovary Syndrome , Progesterone Reductase/genetics , Progesterone Reductase/metabolism , Pyridines/pharmacology
4.
EBioMedicine ; 52: 102635, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32028069

ABSTRACT

BACKGROUND: The ovulatory dysfunction mechanisms underlying polycystic ovary syndrome (PCOS) are not completely understood. There is no effective therapy for PCOS so far. METHODS: We measured the expression of four and a half LIM domain 2 (FHL2) and other related-genes in human granulosa cells (hGCs) from patients with and without PCOS. To minimise the heterogeneity of patients with PCOS, we only included PCOS patients meeting all three criteria according to the revised Rotterdam consensus. The in vitro effects of FHL2 on ovulatory genes and the underlying mechanisms were examined in KGN cells. The role of FHL2 in ovulation was investigated in vivo by overexpressing FHL2 in rat ovaries via intrabursal lentivirus injection. FINDINGS: Increased FHL2 and androgen receptor (AR) expression and decreased CCAAT/enhancer-binding protein ß (C/EBPß) expression were observed in hGCs from patients with PCOS. FHL2 inhibited the expression of ovulation-related genes, including phosphorylated ERK1/2, C/EBPß, COX2 and HAS2 in KGN cells. It was partially by interacting with AR to act as its co-regulator to inhibit C/EBPß expression and by binding to ERK1/2 to inhibit its phosphorylation. Moreover, FHL2 abundance in hGCs was positively correlated with the basal serum testosterone concentration of patients with PCOS, and dihydrotestosterone (DHT)-induced FHL2 upregulation was mediated by AR signalling in KGN cells. Additionally, lentiviral-mediated functional FHL2 overexpression in rat ovaries for 1 week contributed to an impaired superovulatory response, displaying decreased numbers of retrieved oocytes and a lower MII oocyte rate. 3-week FHL2 overexpression rat models without superovulation led to acyclicity and polycystic ovary morphology. INTERPRETATION: Our findings provide novel insights into the mechanisms underlying the pathogenesis of PCOS, suggesting that FHL2 could be a potential treatment target for ovulatory obstacles in PCOS. FUND: National Key Research and Development Program of China, National Natural Science Foundation, National Institutes of Health project and Shanghai Commission of Science and Technology.


Subject(s)
Gene Expression Regulation , LIM-Homeodomain Proteins/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Muscle Proteins/genetics , Ovulation/genetics , Polycystic Ovary Syndrome/etiology , Polycystic Ovary Syndrome/metabolism , Receptors, Androgen/metabolism , Transcription Factors/genetics , Animals , Biomarkers , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Disease Models, Animal , Female , Fluorescent Antibody Technique , Humans , LIM-Homeodomain Proteins/metabolism , MAP Kinase Signaling System , Muscle Proteins/metabolism , Protein Binding , Rats , Receptors, Androgen/genetics , Transcription Factors/metabolism
5.
Gynecol Endocrinol ; 36(8): 668-672, 2020 Aug.
Article in English | MEDLINE | ID: mdl-31902257

ABSTRACT

Our previous study have demonstrated the elevated cortisol concentration in the follicular fluid (FF) contributed to the insulin resistance of the granulosa cells (GCs) in PCOS, but the complicated cortisol generation mechanisms are still unknown. 11ß-hydroxysteroid type 1(11ß-HSD1) mainly functions as reductase in intact cells, converting cortisone to cortisol. Cortisol and IL-1ß are known to induce 11ß-HSD1 in number of tissues, but few results were obtained in ovarian GCs In this study, FF and GCs from PCOS and non-PCOS patients were collected to study the interaction of cortisol and IL-1ß in 11ß-HSD1 expression. The ELISA and qRT-PCR revealed that the cortisol and IL-1ß concentration in FF and 11ß-HSD1 abundance in GCs were elevated in PCOS patients. By using cultured GCs in vitro, we demonstrated that both cortisol and IL-1ß could stimulate 11ß-HSD1 expression. The induction of 11ß-HSD1 by IL-1ß was further inducted by cortisol, whereas the induction of IL-1ß and IL-6 expression by IL-1ß was completely inhibited by cortisol. In conclusion, cortisol and IL-1ß preformed a synergistically upregulation of 11ß-HSD1 expression in GCs, contributing to the accumulation of cortisol in FF of PCOS patients. This may lead to the metabolic disorders of the ovary.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , Granulosa Cells/metabolism , Polycystic Ovary Syndrome/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Adult , Case-Control Studies , Cells, Cultured , Female , Follicular Fluid/metabolism , Gene Expression Regulation, Enzymologic , Granulosa Cells/pathology , Humans , Hydrocortisone/metabolism , Insulin Resistance/physiology , Interleukin-1beta/metabolism , Ovary/metabolism , Ovary/pathology , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/pathology , Up-Regulation/genetics
6.
FASEB J ; 33(10): 11303-11313, 2019 10.
Article in English | MEDLINE | ID: mdl-31311314

ABSTRACT

The insulin resistance (IR) of ovarian granulosa cells from polycystic ovary syndrome (PCOS) aggravates the abnormalities in steroidogenesis and anovulation, and chemerin is an adipokine involved in regulating adipogenesis and glucose homeostasis. The role and underlying mechanism of chemerin in developing IR of the granulosa cells from PCOS remain unclear. Plasma, follicular fluid, and human granulosa-lutein cells (hGLs) were collected from non-PCOS and patients with PCOS with or without IR. The chemerin levels were elevated in both follicular fluid and hGL samples from patients with PCOS with IR, and the hGLs from patients with PCOS with IR showed decreased insulin sensitivity and impaired glucose uptake capacity. Moreover, treatment of chemerin attenuated insulin-stimulated glucose uptake by decreasing phosphorylation of insulin receptor substrate (IRS)1/2 Tyr612, phosphorylation of protein kinase B Ser473, and membrane translocation of glucose transporter type 4 through increasing Ser307 phosphorylation of IRS1 in cultured hGLs. These effects could be abolished by small interfering RNA-mediated knockdown of chemokine-like receptor 1. Furthermore, insulin induced the expression of chemerin in hGLs. Our findings demonstrate a novel role of chemerin in the metabolic dysfunction of PCOS, which suggested that chemerin and its receptor can be further implicated as potential therapeutic targets in the future treatment of PCOS.-Li, X., Zhu, Q., Wang, W., Qi, J., He, Y., Wang, Y., Lu, Y., Wu, H., Ding, Y., Sun, Y. Elevated chemerin induces insulin resistance in human granulosa-lutein cells from polycystic ovary syndrome patients.


Subject(s)
Chemokines/metabolism , Granulosa Cells/metabolism , Insulin Resistance/physiology , Insulin/metabolism , Luteal Cells/metabolism , Polycystic Ovary Syndrome/metabolism , Adipokines/metabolism , Adult , Female , Humans , Insulin Receptor Substrate Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Ovary/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Chemokine/metabolism
7.
Gynecol Endocrinol ; 35(8): 669-672, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31056990

ABSTRACT

Hyperandrogenism is one of the most common causes for anovulation in women and increases the risk for metabolic disorder in PCOS patients. Autophagy plays an important role in dysfunction of endocrine and anovulation. However, the relationship between hyperandrogenism and autophagy in human granulosa cells of PCOS patients remains unclear. By collecting granulosa cells from PCOS patients and non-PCOS patients, we found that the abundance of autophagy-related genes ATG5, ATG7, BECN1 mRNA and the ratio of autophagy marker protein light chain 3B II/I (LC3 II/I) were significantly increased whereas the abundance of the autophagy substrate SQSTM1/p62 was decreased in ovarian granulosa cells from PCOS patients. Furthermore, we demonstrated that BECN1 mRNA abundance in human granulosa cells positively correlated with the basal level of serum total testosterone and androgen up-regulated the abundance of BECN1 mRNA and the ratio of LC3II/LC3I in a dose-dependent manner in cultured granulosa cells. These observations indicated that androgen-induced activation of autophagy may play an important role in the development of PCOS and to explore the autophagy mechanisms involved in PCOS yield new insight into the pathophysiology and therapy of the disorder.


Subject(s)
Androgens/physiology , Autophagy/physiology , Granulosa Cells/physiology , Polycystic Ovary Syndrome/pathology , Adult , Androgens/metabolism , Androgens/pharmacology , Autophagy/drug effects , Autophagy/genetics , Case-Control Studies , Cells, Cultured , Female , Gene Expression Regulation/drug effects , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Granulosa Cells/pathology , Humans , Hyperandrogenism/complications , Hyperandrogenism/metabolism , Hyperandrogenism/pathology , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/metabolism , Primary Cell Culture , Young Adult
8.
Endocrinology ; 158(10): 3281-3291, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28651331

ABSTRACT

The proinflammatory cytokine interleukin-1ß (IL-1ß) may be involved in several ovulation-associated events, such as protease synthesis, prostaglandin production, and steroidogenesis in granulosa cells. However, the exact effect of IL-1ß on progesterone synthesis in granulosa cells and the underlying mechanism remain unclear. By using cultured granulosa-lutein cells collected from women undergoing in vitro fertilization or intracytoplasmic sperm injection, we found that IL-1ß upregulated steroidogenic acute regulatory protein (StAR) expression and progesterone synthesis in granulosa-lutein cells, which was comparable with luteinizing hormone effect and could be abolished by an IL-1 receptor antagonist. Moreover, IL-1ß activated the phosphorylation of cyclic adenosine monophosphate response element-binding protein (CREB), and knockdown of CREB attenuated the induction of StAR expression and progesterone synthesis by IL-1ß in granulosa-lutein cells. Furthermore, IL-1ß activated the extracellular signal-regulated kinase (ERK)1/2 and p38 pathways and inhibition of the ERK1/2 and p38 pathways attenuated the IL-1ß-induced phosphorylation of CREB, StAR expression, and progesterone synthesis in granulosa-lutein cells. In conclusion, IL-1ß could upregulate StAR expression and stimulate progesterone biosynthesis through increase in CREB phosphorylation via activating the ERK1/2 and p38 pathways in human granulosa-lutein cells.


Subject(s)
Interleukin-1beta/pharmacology , Luteal Cells/drug effects , Luteal Cells/metabolism , Phosphoproteins/biosynthesis , Progesterone/biosynthesis , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/physiology , Female , Gene Knockdown Techniques , Humans , Luteinizing Hormone/pharmacology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Phosphoproteins/genetics , Phosphorylation/drug effects , Receptors, Interleukin-1/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/physiology , Up-Regulation/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL