Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 479
Filter
1.
J Transl Med ; 22(1): 834, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39261935

ABSTRACT

BACKGROUND: Premature ovarian insufficiency (POI) is a condition characterized by a substantial decline or loss of ovarian function in women before the age of 40. However, the pathogenesis of POI remains to be further elucidated, and specific targeted drugs which could delay or reverse ovarian reserve decline are urgently needed. Abnormal DNA damage repair (DDR) and cell senescence in granulosa cells are pathogenic mechanisms of POI. Ubiquitin-specific protease 14 (USP14) is a key enzyme that regulates the deubiquitylation of DDR-related proteins, but whether USP14 participates in the pathogenesis of POI remains unclear. METHODS: We measured USP14 mRNA expression in granulosa cells from biochemical POI (bPOI) patients. In KGN cells, we used IU1 and siRNA-USP14 to specifically inhibit USP14 and constructed a cell line stably overexpressing USP14 to examine its effects on DDR function and cellular senescence in granulosa cells. Next, we explored the therapeutic potential of IU1 in POI mouse models induced by D-galactose. RESULTS: USP14 expression in the granulosa cells of bPOI patients was significantly upregulated. In KGN cells, IU1 treatment and siUSP14 transfection decreased etoposide-induced DNA damage levels, promoted DDR function, and inhibited cell senescence. USP14 overexpression increased DNA damage, impaired DDR function, and promoted cell senescence. Moreover, IU1 treatment and siUSP14 transfection increased nonhomologous end joining (NHEJ), upregulated RNF168, Ku70, and DDB1, and increased ubiquitinated DDB1 levels in KGN cells. Conversely, USP14 overexpression had the opposite effects. Intraperitoneal IU1 injection alleviated etoposide-induced DNA damage in granulosa cells, ameliorated the D-galactose-induced POI phenotype, promoted DDR, and inhibited cell senescence in ovarian granulosa cells in vivo. CONCLUSIONS: Upregulated USP14 in ovarian granulosa cells may play a role in POI pathogenesis, and targeting USP14 may be a potential POI treatment strategy. Our study provides new insights into the pathogenesis of POI and a novel POI treatment strategy.


Subject(s)
Cellular Senescence , DNA Damage , DNA Repair , Granulosa Cells , Primary Ovarian Insufficiency , Ubiquitin Thiolesterase , Female , Primary Ovarian Insufficiency/pathology , Primary Ovarian Insufficiency/metabolism , Primary Ovarian Insufficiency/genetics , Granulosa Cells/metabolism , Granulosa Cells/drug effects , Granulosa Cells/pathology , Cellular Senescence/drug effects , Animals , Humans , Ubiquitin Thiolesterase/metabolism , Ubiquitin Thiolesterase/genetics , DNA Repair/drug effects , Mice , Adult , Mice, Inbred C57BL , Cell Line
2.
Cell Biochem Funct ; 42(7): e4122, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39256962

ABSTRACT

Premature ovarian failure (POF) is intricately linked to cellular fates such as senescence, apoptosis, and impaired granulosa cell (GC) differentiation, each of which contributes to ovarian dysfunction and follicular depletion. Autophagy is essential in preventing POF by maintaining cellular homeostasis through the degradation and recycling of damaged organelles and proteins, thereby preserving ovarian function and preventing follicular depletion. Recent studies have revealed that the targeted regulation and disruption of autophagy through various molecular mechanisms ultimately lead to the pathogenesis of POF. In this review, we provide a comprehensive analysis of the disruption in regulatory mechanisms of autophagy contributing to POF. Specifically, we elucidate the molecular mechanisms that can be targeted to restore autophagy homeostasis, offering therapeutic potential for the treatment of POF.


Subject(s)
Autophagy , Primary Ovarian Insufficiency , Humans , Primary Ovarian Insufficiency/metabolism , Primary Ovarian Insufficiency/pathology , Female , Animals , Granulosa Cells/metabolism , Granulosa Cells/pathology
3.
Mol Reprod Dev ; 91(8): e23768, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39155689

ABSTRACT

The pathologic mechanism of polycystic ovary syndrome (PCOS) is related to increased autophagy of granulosa cells. Both berberine and metformin have been shown to improve PCOS, but whether the combination of berberine and metformin can better improve PCOS by inhibiting autophagy remains unclear. PCOS models were constructed by injecting dehydroepiandrosterone into rats, and berberine, metformin or berberine combined with metformin was administered to rats after modeling. Rats' body weight and ovarian weight were measured before and after modeling. Histopathological examination of ovarian tissue and estrous cycle analysis of rats were performed. Insulin resistance, hormone levels, oxidative stress, and lipid metabolism in PCOS rats were assessed. Expression of the AMPK/AKT/mTOR pathway and autophagy-related proteins was analyzed by Western blot assays. Granulosa cells were isolated from rat ovarian tissue and identified by immunofluorescence staining followed by transmission electron microscopy analysis. Berberine combined with metformin reduced the body weight and ovarian weight of PCOS rats, increased the number of primordial and primary follicles, decreased the number of secondary and atretic follicles, normalized the estrous cycle, and improved insulin resistance, androgen biosynthesis, oxidative stress and lipid metabolism disorders, and increased estrogen production. In addition, berberine combined with metformin reduced the number of autophagosomes in granulosa cells, which may be related to AMPK/AKT/mTOR pathway activation, decreased Beclin1 and LC3II/LC3I levels, and increased p62 expression. Berberine combined with metformin could inhibit autophagy by activating the AMPK/AKT/mTOR pathway in PCOS, indicating that berberine combined with metformin is a potential treatment strategy for PCOS.


Subject(s)
Autophagy , Berberine , Metformin , Polycystic Ovary Syndrome , Proto-Oncogene Proteins c-akt , Signal Transduction , TOR Serine-Threonine Kinases , Female , Animals , Metformin/pharmacology , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/drug therapy , Polycystic Ovary Syndrome/pathology , Autophagy/drug effects , Berberine/pharmacology , Rats , TOR Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , AMP-Activated Protein Kinases/metabolism , Rats, Sprague-Dawley , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Granulosa Cells/pathology , Insulin Resistance , Ovary/drug effects , Ovary/metabolism , Ovary/pathology , Drug Therapy, Combination , Oxidative Stress/drug effects
4.
Cell Signal ; 122: 111334, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39102927

ABSTRACT

OBJECTIVE: Chronic psychological stress is associated with impaired follicular development and ovarian dysfunction. Many aspects of this dysfunction and the underlying mechanisms remain unclear. Using a chronic unpredictable mild stress (CUMS) mouse model, we investigate the influence of chronic stress on ovarian function and explore potential mechanisms. METHODS: A CUMS mouse model was constructed over eight months, covering the period from sexual maturity to the onset of declining fertility in mice. At the end of the 2nd, 4th, 6th, and 8th months of exposure to CUMS, behavioral and physiological assays, including the sucrose preference test, tail suspension test, and serum corticosterone levels, were conducted to validate the effectiveness of the stress model. Fertility and ovarian function were assessed by analyzing the estrous cycle, number of offspring, sex hormone levels, follicle counts, granulosa cell proliferation and apoptosis, and the expression levels of fibrosis markers. Furthermore, proteomic analyses were performed on the ovaries to investigate the molecular mechanisms of ovarian fibrosis induced by CUMS. RESULTS: With continued CUMS exposure, there was a gradual decline in both the ovary-to-body weight ratio and the number of offspring. Moreover, the percentage of atretic follicles was notably higher in the CUMS-exposed groups compared to the control groups. It is noticeable that CUMS triggered granulosa cell apoptosis and halted proliferation. Additionally, increased expression of α-SMA and Collagen I in the ovaries of CUMS-exposed mice indicated that CUMS could induce ovarian fibrosis. Proteomic analysis provided insights into the activation of specific biological processes and molecules associated with fibrosis induced by chronic stress. CONCLUSIONS: Our results strongly suggest that exposure to CUMS induces ovarian fibrosis, which influences follicular development and ultimately contributes to fertility decline. These findings offer novel perspectives on the impact of chronic stress on ovarian dysfunction.


Subject(s)
Fertility , Fibrosis , Ovary , Stress, Psychological , Animals , Female , Mice , Stress, Psychological/complications , Ovary/pathology , Ovary/metabolism , Apoptosis , Granulosa Cells/metabolism , Granulosa Cells/pathology , Disease Models, Animal , Ovarian Follicle/metabolism , Ovarian Follicle/pathology , Cell Proliferation
5.
Biochim Biophys Acta Mol Cell Res ; 1871(7): 119822, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39159685

ABSTRACT

Polycystic ovary syndrome (PCOS) is a common endocrine disorder characterized by chronic ovulation dysfunction and overproduction of androgens. Women with PCOS are commonly accompanied by insulin resistance (IR), which can impair insulin sensitivity and elevate blood glucose levels. IR promotes ovarian cysts, ovulatory dysfunction, and menstrual irregularities in women patients, leading to the pathogenesis of PCOS. Secreted frizzled-related protein 4 (SFRP4), a secreted glycoprotein, exhibits significantly elevated expression levels in obese individuals with IR and PCOS. Whereas, whether it plays a role in regulating IR-induced PCOS still has yet to be understood. In this study, we respectively established in vitro IR-induced hyperandrogenism in human ovarian granular cells and in vivo IR-induced PCOS models in mice to investigate the action mechanisms of SFRP4 in modulating IR-induced PCOS. Here, we revealed that SFRP4 expression levels in both mRNA and protein were remarkably upregulated in the IR-induced hyperandrogenism with elevated testosterone in the human ovarian granulosa cell line KGN. Under normal conditions without hyperandrogenism, overexpressing SFRP4 triggered the remarkable elevation of testosterone along with the increased nuclear translocation of ß-catenin, cell apoptosis and proinflammatory cytokine IL-6. Furthermore, we found that phytopharmaceutical disruption of SFRP4 by genistein ameliorated IR-induced increase in testosterone in ovarian granular cells, and IR-induced PCOS in high-fat diet obese mice. Our study reveals that SFRP4 contributes to IR-induced PCOS by triggering ovarian granulosa cell hyperandrogenism and apoptosis through the nuclear ß-catenin/IL-6 signaling axis. Elucidating the role of SFRP4 in PCOS may provide a novel therapeutic strategy for IR-related PCOS therapy.


Subject(s)
Apoptosis , Granulosa Cells , Hyperandrogenism , Insulin Resistance , Interleukin-6 , Polycystic Ovary Syndrome , Signal Transduction , beta Catenin , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/pathology , Polycystic Ovary Syndrome/genetics , Female , Animals , Humans , Hyperandrogenism/metabolism , Hyperandrogenism/genetics , Hyperandrogenism/pathology , Granulosa Cells/metabolism , Granulosa Cells/pathology , beta Catenin/metabolism , beta Catenin/genetics , Mice , Interleukin-6/metabolism , Interleukin-6/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , Mice, Inbred C57BL , Cell Line
6.
Biochim Biophys Acta Mol Basis Dis ; 1870(7): 167334, 2024 10.
Article in English | MEDLINE | ID: mdl-38971505

ABSTRACT

Ovarian aging, a complex and challenging concern within the realm of reproductive medicine, is associated with reduced fertility, menopausal symptoms and long-term health risks. Our previous investigation revealed a correlation between Peroxiredoxin 4 (PRDX4) and human ovarian aging. The purpose of this research was to substantiate the protective role of PRDX4 against ovarian aging and elucidate the underlying molecular mechanism in mice. In this study, a Prdx4-/- mouse model was established and it was observed that the deficiency of PRDX4 led to only an accelerated decline of ovarian function in comparison to wild-type (WT) mice. The impaired ovarian function observed in this study can be attributed to an imbalance in protein homeostasis, an exacerbation of endoplasmic reticulum stress (ER stress), and ultimately an increase in apoptosis of granulosa cells. Furthermore, our research reveals a noteworthy decline in the expression of Follicle-stimulating hormone receptor (FSHR) in aging Prdx4-/- mice, especially the functional trimer, due to impaired disulfide bond formation. Contrarily, the overexpression of PRDX4 facilitated the maintenance of protein homeostasis, mitigated ER stress, and consequently elevated E2 levels in a simulated KGN cell aging model. Additionally, the overexpression of PRDX4 restored the expression of the correct spatial conformation of FSHR, the functional trimer. In summary, our research reveals the significant contribution of PRDX4 in delaying ovarian aging, presenting a novel and promising therapeutic target for ovarian aging from the perspective of endoplasmic reticulum protein homeostasis.


Subject(s)
Aging , Endoplasmic Reticulum Stress , Granulosa Cells , Mice, Knockout , Ovary , Peroxiredoxins , Proteostasis , Animals , Female , Peroxiredoxins/metabolism , Peroxiredoxins/genetics , Granulosa Cells/metabolism , Granulosa Cells/pathology , Mice , Aging/metabolism , Aging/pathology , Ovary/metabolism , Ovary/pathology , Humans , Apoptosis , Receptors, FSH/metabolism , Receptors, FSH/genetics
7.
Free Radic Biol Med ; 222: 72-84, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38825211

ABSTRACT

Premature ovarian insufficiency (POI) is a clinical syndrome characterised by a decline in ovarian function in women before 40 years of age and is associated with oestradiol deficiency and a complex pathogenesis. However, the aetiology of POI is still unclear and effective preventative and treatment strategies are still lacking. Methyltransferase like 3 (METTL3) is an RNA methyltransferase that is involved in spermatogenesis, oocyte development and maturation, early embryonic development, and embryonic stem cell differentiation and formation, but its role in POI is unknown. In the present study, METTL3 deficiency in follicular theca cells was found to lead to reduced fertility in female mice, with a POI-like phenotype, and METTL3 knockout promoted ovarian inflammation. Further, a reduction in METTL3 in follicular theca cells led to a decrease in the m6A modification of pri-miR-21, which further reduced pri-miR-21 recognition and binding by DGCR8 proteins, leading to a decrease in the synthesis of mature miR-21-5p. Decrease of miR-21-5p promoted the secretion of interleukin-1ß (IL-1ß) from follicular theca cells. Acting in a paracrine manner, IL-1ß inhibited the cAMP-PKA pathway and activated the NF-κB pathway in follicular granulosa cells. This activation increased the levels of reactive oxygen species in granulosa cells, causing disturbances in the intracellular Ca2+ balance and mitochondrial damage. These cellular events ultimately led to granulosa cell apoptosis and a decrease in oestradiol synthesis, resulting in POI development. Collectively, these findings reveal how METTL3 deficiency promotes the expression and secretion of IL-1ß in theca cells, which regulates ovarian functions, and proposes a new theory for the development of POI disease.


Subject(s)
Interleukin-1beta , Methyltransferases , Primary Ovarian Insufficiency , Theca Cells , Animals , Female , Humans , Mice , Gene Expression Regulation , Granulosa Cells/metabolism , Granulosa Cells/pathology , Interleukin-1beta/metabolism , Interleukin-1beta/genetics , Methyltransferases/metabolism , Methyltransferases/genetics , Mice, Knockout , MicroRNAs/genetics , MicroRNAs/metabolism , Primary Ovarian Insufficiency/pathology , Primary Ovarian Insufficiency/genetics , Primary Ovarian Insufficiency/metabolism , Signal Transduction , Theca Cells/metabolism , Theca Cells/pathology
8.
Reproduction ; 168(2)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38912966

ABSTRACT

In brief: This study reveals that orthotopic transplantation of 3D hUC-MSC spheroids is more effective than monolayer-cultured hUC-MSCs in improving POF and distinctly reducing oxidative stress through the paracrine effect, thereby preventing apoptosis and autophagy of GCs. Abstract: Premature ovarian failure (POF) is a common reproductive disease in women younger than 40 years old, and studies have demonstrated that the application of human umbilical cord mesenchymal stem cells (hUC-MSCs) is a promising therapy strategy for POF. Given the previously established therapeutic advantages of 3D MSC spheroids, and to evaluate their effectiveness, both 3D hUC-MSC spheroids and monolayer-cultured hUC-MSCs were employed to treat a cyclophosphamide-induced POF rat model through orthotopic transplantation. The effects of these two forms on POF were subsequently assessed by examining apoptosis, autophagy, and oxidative damage in ovarian granulosa cells (GCs). The results indicated that hUC-MSC spheroids exhibited superior treatment effects on resisting autophagy, apoptosis, and oxidative damage in GCs compared to monolayer-cultured hUC-MSCs. To further elucidate the impact of hUC-MSC spheroids in vitro, a H2O2-induced KGN cells model was established and co-cultured with both forms of hUC-MSCs. As expected, the hUC-MSC spheroids also exhibited superior effects in resisting apoptosis and autophagy caused by oxidative damage. Therefore, this study demonstrates that 3D hUC-MSC spheroids have potential advantages in POF therapy; however, the detailed mechanisms need to be further investigated. Furthermore, this study will provide a reference for the clinical treatment strategy of POF.


Subject(s)
Apoptosis , Autophagy , Disease Models, Animal , Granulosa Cells , Mesenchymal Stem Cells , Oxidative Stress , Primary Ovarian Insufficiency , Spheroids, Cellular , Female , Animals , Rats , Granulosa Cells/pathology , Granulosa Cells/cytology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Primary Ovarian Insufficiency/pathology , Primary Ovarian Insufficiency/therapy , Primary Ovarian Insufficiency/chemically induced , Humans , Mesenchymal Stem Cell Transplantation , Rats, Sprague-Dawley , Umbilical Cord/cytology , Cells, Cultured
9.
Biol Reprod ; 111(3): 625-639, 2024 Sep 14.
Article in English | MEDLINE | ID: mdl-38874314

ABSTRACT

The morbidity of polycystic ovary syndrome (PCOS) is in highly increasing rate nowadays. PCOS not only affects the fertility in women, but also threatens the health of whole life. Hence, to find the prognostic risk factors is of great value. However, the effective predictors in clinical practice of PCOS are still in blackness. In this study, we found Klotho (KL) was increased in follicular fluid (FF) and primary luteinized granulosa cells (GCs) from PCOS patients with hyperandrogenism. Furthermore, we found follicular KL was negatively correlated with numbers of mature oocytes, and positively correlated with serum testosterone, LH, and LH/FSH levels menstrual cycle and number of total antral follicles in PCOS patients. In primary luteinized GCs, the increased KL was accompanied with upregulation of cell apoptosis and inflammation-related genes. In ovaries of PCOS mice and cultured human KGN cell line, KL was up-regulated and accompanied by apoptosis, inflammation, and mitochondrial dysfunction. Therefore, our findings suggest new mechanisms for granulosa cell injury and revealed to target inhibit KL maybe a new therapeutic strategy for treatment of PCOS.


Subject(s)
Apoptosis , Glucuronidase , Granulosa Cells , Inflammation , Klotho Proteins , Polycystic Ovary Syndrome , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/pathology , Female , Granulosa Cells/metabolism , Granulosa Cells/pathology , Apoptosis/physiology , Humans , Mice , Animals , Klotho Proteins/metabolism , Inflammation/metabolism , Inflammation/pathology , Glucuronidase/metabolism , Glucuronidase/genetics , Adult , Disease Progression , Follicular Fluid/metabolism
10.
Mol Cell Endocrinol ; 592: 112322, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38942281

ABSTRACT

Polycystic ovary syndrome (PCOS) is a type of follicular dysplasia with an unclear pathogenesis, posing certain challenges in its diagnosis and treatment. Cancer susceptibility candidate 15 (CASC15), a long non-coding RNA closely associated with tumour development, has been implicated in PCOS onset and development. Therefore, this study aimed to investigate the molecular mechanisms underlying PCOS by downregulating CASC15 expression in both in vitro and in vivo models. We explored the potential regulatory relationship between CASC15 expression and PCOS by examining cell proliferation, cell cycle dynamics, cell autophagy, steroid hormone secretion capacity, and overall ovarian function in mice. We found that CASC15 expression in granulosa cells derived from patients with PCOS was significantly higher than those of the normal group (P < 0.001). In vitro experiments revealed that downregulating CASC15 significantly inhibited cell proliferation, promoted apoptosis, induced G1-phase cell cycle arrest, and influenced cellular autophagy levels. Moreover, downregulating CASC15 affected the follicular development process in newborn mouse ovaries. In vivo studies in mice demonstrated that disrupting CASC15 expression improved PCOS-related symptoms such as polycystic changes and hyperandrogenism, and significantly affected ovulation induction and embryo implantation in pregnant mice. Overall, CASC15 was highly expressed in granulosa cells of patients with PCOS and its downregulation improved PCOS-related symptoms by influencing granulosa cell function and follicular development in mice.


Subject(s)
Apoptosis , Autophagy , Cell Proliferation , Down-Regulation , Granulosa Cells , Ovarian Follicle , Polycystic Ovary Syndrome , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/pathology , Polycystic Ovary Syndrome/genetics , Female , Granulosa Cells/metabolism , Granulosa Cells/pathology , Animals , Cell Proliferation/genetics , Humans , Down-Regulation/genetics , Ovarian Follicle/metabolism , Ovarian Follicle/pathology , Autophagy/genetics , Mice , Apoptosis/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Adult , Pregnancy
11.
Sci Rep ; 14(1): 13628, 2024 06 13.
Article in English | MEDLINE | ID: mdl-38871783

ABSTRACT

This study intended to investigate if gynecological cancers compromise ovarian function and reduce the success of assisted reproduction techniques (ART). No clinical and molecular data together is available on this issue for gynecological or other organ cancers. Steroidogenic pathways and DNA damage response characteristics of the granulosa cells retrieved from the 39 gynecological cancer patients were analyzed together with their clinical ART characteristics in comparison to 31 control ART patients. Patients with gynecological malignancies were similar to the control IVF patients for the number of mature oocytes retrieved, fertilization rates and embryo development competency. Molecular analyses of the granulosa cells retrieved from these cancer patients did not detect any perturbations in gonadotropin receptor expression and response, sex steroid production, cholesterol utilization/storage and, DNA damage response pattern in comparison to control IVF patients without cancer. This study provides the first reassuring clinical and molecular combined data set that the presence of gynecological malignancy does not appear to have any detrimental effect on clinical IVF cycle characteristics and ovarian functioning at molecular level.


Subject(s)
DNA Damage , Fertilization in Vitro , Genital Neoplasms, Female , Humans , Female , Fertilization in Vitro/methods , Genital Neoplasms, Female/genetics , Genital Neoplasms, Female/pathology , Genital Neoplasms, Female/metabolism , Adult , Granulosa Cells/metabolism , Granulosa Cells/pathology , Pregnancy
12.
Mol Cell Endocrinol ; 592: 112292, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38830447

ABSTRACT

RESEARCH QUESTION: Granulosa cells (GCs) dysfunction plays a crucial role in the pathogenesis of polycystic ovary syndrome (PCOS). It is reported that YTH domain-containing family protein 2 (YTHDF2) is upregulated in mural GCs of PCOS patients. What effect does the differential expression of YTHDF2 have in PCOS patients? DESIGN: Mural GCs and cumulus GCs from 15 patients with PCOS and 15 ovulatory controls and 4 cases of pathological sections in each group were collected. Real-time PCR, Western Blot, immunohistochemistry, and immunofluorescence experiments were conducted to detect gene and protein expression. RNA immunoprecipitation assay was performed to evaluate the binding relationship between YTHDF2 and MSS51. Mitochondrial morphology, cellular ATP and ROS levels and glycolysis-related gene expression were detected after YTHDF2 overexpression or MSS51 inhibition. RESULTS: In the present study, we found that YTHDF2 was upregulated in GCs of PCOS patients while MSS51 was downregulated. YTHDF2 protein can bind to MSS51 mRNA and affect MSS51 expression. The reduction of MSS51 expression or the increase in YTHDF2 expression can lead to mitochondrial damage, reduced ATP levels, increased ROS levels and reduced expression of LDHA, PFKP and PKM. CONCLUSIONS: YTHDF2 may regulate the expression of MSS51, affecting the structure and function of mitochondria in GCs and interfering with cellular glycolysis, which may disturb the normal biological processes of GCs and follicle development in PCOS patients.


Subject(s)
Granulosa Cells , Mitochondria , Polycystic Ovary Syndrome , RNA-Binding Proteins , Humans , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/pathology , Female , Granulosa Cells/metabolism , Granulosa Cells/pathology , Mitochondria/metabolism , Mitochondria/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Adult , Reactive Oxygen Species/metabolism , Gene Expression Regulation , Adenosine Triphosphate/metabolism , Glycolysis/genetics , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
13.
Genes (Basel) ; 15(5)2024 05 12.
Article in English | MEDLINE | ID: mdl-38790245

ABSTRACT

Insulin receptor signaling promotes cell differentiation, proliferation, and growth which are essential for oocyte maturation, embryo implantation, endometrial decidualization, and placentation. The dysregulation of insulin signaling in women with metabolic syndromes including diabetes exhibits poor pregnancy outcomes that are poorly understood. We utilized the Cre/LoxP system to target the tissue-specific conditional ablation of insulin receptor (Insr) and insulin-like growth factor-1 receptor (Igf1r) using an anti-Mullerian hormone receptor 2 (Amhr2) Cre-driver which is active in ovarian granulosa and uterine stromal cells. Our long-term goal is to examine insulin-dependent molecular mechanisms that underlie diabetic pregnancy complications, and our conditional knockout models allow for such investigation without confounding effects of ligand identity, source and cross-reactivity, or global metabolic status within dams. Puberty occurred with normal timing in all conditional knockout models. Estrous cycles progressed normally in Insrd/d females but were briefly stalled in diestrus in Igf1rd/d and double receptor (DKO) mice. The expression of vital ovulatory genes (Lhcgr, Pgr, Ptgs2) was not significantly different in 12 h post-hCG superovulated ovaries in knockout mice. Antral follicles exhibited an elevated apoptosis of granulosa cells in Igf1rd/d and DKO mice. However, the distribution of ovarian follicle subtypes and subsequent ovulations was normal in all insulin receptor mutants compared to littermate controls. While ovulation was normal, all knockout lines were subfertile suggesting that the loss of insulin receptor signaling in the uterine stroma elicits implantation and decidualization defects responsible for subfertility in Amhr2-Cre-derived insulin receptor mutants.


Subject(s)
Ovary , Receptor, IGF Type 1 , Receptor, Insulin , Animals , Female , Mice , Pregnancy , Granulosa Cells/metabolism , Granulosa Cells/pathology , Infertility, Female/genetics , Infertility, Female/metabolism , Infertility, Female/pathology , Mice, Knockout , Ovary/metabolism , Ovary/pathology , Ovulation/genetics , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Signal Transduction/genetics
14.
Discov Med ; 36(184): 946-958, 2024 May.
Article in English | MEDLINE | ID: mdl-38798254

ABSTRACT

BACKGROUND: Polycystic ovary syndrome (PCOS) commonly impacts fertile females with potentially severe effects on fertility and metabolism. Blood ghrelin levels are lower in PCOS patients, and exogenous supplements have been proposed for their potential to trigger anti-inflammatory effects at the cellular level. This study aimed to investigate whether pretreatment with ghrelin reduced inflammation, insulin resistance, and reproductive abnormalities in PCOS and the underlying mechanism of this disorder. METHODS: Ghrelin supplementation was first tested in an inflammation model using human ovarian granulosa cells (KGN cells) that were built by treated with Lipolyaccharide. KGN cells were pretreated with ghrelin and exposed to lipopolysaccharide (LPS). Inflammatory gene expression and cytokine production were analyzed by Enzyme-linked immunosorbent assay (ELISA). Based on these results, the PCOS mice model was built with Dehydroepiandrosterone (DHEA) and a high-fat diet. The mRNA and protein expressions of inflammatory factors including Toll-like receptor 4 (TLR4), nuclear factor kappa-B-p65 (NF-κB-p65), Phospho-NF-κB-p65 (p-NF-κB-p65) and myeloid differentiation factor 88 (MYD88) related to the TLR4/NF-κB signaling pathway were evaluated in KGN cells and mouse ovarian tissues using Real-Time Quantitative Polymerase Chain Reaction (RT-qPCR) and western blot, respectively. Lipid metabolism was quantified via an automated biochemical analyzer. RESULTS: The mRNA and protein expressions of interleukin-6 (IL-6), interleukin-1 beta (IL-1ß), and tumor necrosis factor alpha (TNF-α) in ghrelin pretreated KGN cells were lower than the LPS group (p < 0.05). Protein expression was reduced for TLR4, NF-κB-p65, and MYD88 within KGN cells of ghrelin groups compared to the LPS group (p < 0.05). Ghrelin treatment restored the estrous cycle and slowed weight gain and abdominal fat weight of PCOS mice (p < 0.05). Ghrelin treatment decreased the serum concentrations of testosterone, luteinizing hormone, insulin, IL-6, IL-1ß, and TNF-α compared to the PCOS group (p < 0.05). Estradiol concentrations of mice treated with ghrelin were higher than the PCOS group (p < 0.05). The concentrations of low and high-density lipoprotein, triglyceride, and cholesterol in mice treated with ghrelin were lower than in the PCOS mice (p < 0.05). Inflammatory gene expression for IL-6, IL-1ß, TNF-α, TLR4, NF-κB-p65, and MYD88 decreased in the ovarian tissues of ghrelin-treated mice compared to the PCOS group (p < 0.05), along with reduced protein expression of TLR4, p-NF-κB-p65, and MYD88 (p < 0.05). CONCLUSIONS: In the present study, ghrelin treatment effectively reduced inflammation in vitro, and attenuated insulin resistance and reproductive abnormalities in PCOS mice through the TLR4/NF-κB signaling pathway, highlighting potential therapeutic avenues for future PCOS treatments and research directions.


Subject(s)
Ghrelin , Inflammation , Insulin Resistance , NF-kappa B , Polycystic Ovary Syndrome , Signal Transduction , Toll-Like Receptor 4 , Animals , Polycystic Ovary Syndrome/drug therapy , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/pathology , Female , Toll-Like Receptor 4/metabolism , Mice , Signal Transduction/drug effects , NF-kappa B/metabolism , Inflammation/metabolism , Humans , Disease Models, Animal , Granulosa Cells/metabolism , Granulosa Cells/drug effects , Granulosa Cells/pathology
15.
Mol Cell Endocrinol ; 589: 112248, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38663484

ABSTRACT

Young women undergoing anticancer treatment are at risk of premature ovarian failure (POF). Endometrial-derived stem cells (EnSCs) have demonstrated significant therapeutic potential for treating ovarian insufficiency, although the underlying mechanisms remain to be fully understood. This study aims to further investigate the therapeutic effects of EnSCs, particularly through the paracrine action of fibroblast growth factor 2 (FGF2), on POF. The findings show that exogenous FGF2 enhances the survival of ovarian granulosa cells damaged by cisplatin. FGF2 stimulates the proliferation of these damaged cells by suppressing the Hippo signaling pathway and activating YAP expression. In vivo experiments also revealed that FGF2 treatment significantly improves ovarian reserve and endocrine function in mice with POF. These results suggest that FGF2 can boost the proliferative capacity of damaged ovarian granulosa cells through the Hippo-YAP signaling pathway, providing a theoretical foundation for using EnSCs and FGF2 in clinical treatments for POF.


Subject(s)
Cell Proliferation , Fibroblast Growth Factor 2 , Granulosa Cells , Hippo Signaling Pathway , Primary Ovarian Insufficiency , Signal Transduction , YAP-Signaling Proteins , Primary Ovarian Insufficiency/metabolism , Primary Ovarian Insufficiency/pathology , Female , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factor 2/pharmacology , Granulosa Cells/metabolism , Granulosa Cells/drug effects , Granulosa Cells/pathology , Animals , Cell Proliferation/drug effects , Signal Transduction/drug effects , Humans , Mice , YAP-Signaling Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Cisplatin/pharmacology , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics
16.
Free Radic Biol Med ; 220: 1-14, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38677487

ABSTRACT

Primary ovarian insufficiency (POI) in younger women (under 40) manifests as irregular periods, high follicle-stimulating hormone (FSH), and low estradiol (E2), often triggered by chemotherapy. Though mesenchymal stem cell (MSC) therapy shows promise in treating POI, its exact mechanism remains unclear. This study reveals that human umbilical cord-derived MSCs (hUC-MSCs) can protect ovarian granulosa cells (GCs) from cyclophosphamide (CTX)-induced ferroptosis, a form of cell death driven by iron accumulation. CTX, commonly used to induce POI animal model, triggered ferroptosis in GCs, while hUC-MSCs treatment mitigated this effect, both in vivo and in vitro. Further investigations using ferroptosis and autophagy inhibitors suggest that hUC-MSCs act by suppressing ferroptosis in GCs. Interestingly, hUC-MSCs activate a protective antioxidant pathway in GCs via NRF2, a stress-response regulator. Overall, our findings suggest that hUC-MSCs improve ovarian function in CTX-induced POI by reducing ferroptosis in GCs. This study not only clarifies the mechanism behind the benefits of hUC-MSCs but also strengthens the case for their clinical use in treating POI. Additionally, it opens up a new avenue for protecting ovaries from chemotherapy-induced damage by regulating ferroptosis.


Subject(s)
Autophagy , Cyclophosphamide , Disease Models, Animal , Ferroptosis , Granulosa Cells , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Primary Ovarian Insufficiency , Umbilical Cord , Female , Primary Ovarian Insufficiency/chemically induced , Primary Ovarian Insufficiency/therapy , Primary Ovarian Insufficiency/metabolism , Primary Ovarian Insufficiency/pathology , Animals , Ferroptosis/drug effects , Granulosa Cells/metabolism , Granulosa Cells/drug effects , Granulosa Cells/pathology , Humans , Mice , Mesenchymal Stem Cells/metabolism , Umbilical Cord/cytology , Cyclophosphamide/adverse effects , Mesenchymal Stem Cell Transplantation/methods , Autophagy/drug effects , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Ferritins/metabolism
17.
Ecotoxicol Environ Saf ; 276: 116319, 2024 May.
Article in English | MEDLINE | ID: mdl-38615642

ABSTRACT

Di-hexyl phthalate (2-ethylhexyl) (DEHP) has been confirmed to cause female reproductive toxicity in humans and model animals by affecting the survival of ovarian granulosa cells (GCs), but the interrelationships between DEHP's on autophagy, apoptosis, and inflammation in GCs are not clear. Our previous study demonstrated that DEHP exposure resulted in the disturbance of intestinal flora associated with serum LPS release, which in turn led to impaired ovarian function. LPS has also been shown to determine cell fate by modulating cellular autophagy, apoptosis, and inflammation. Therefore, this study investigated the role and link between LPS and autophagy, apoptosis, and inflammation of GCs in DEHP-induced ovarian injury. Here, we constructed an in vivo injury model by continuous gavage of 0-1500 mg/kg of DEHP in female mice for 30 days and an in vitro injury model by treatment of human ovarian granulosa cells (KGN) cells with mono-2- ethylhexyl ester (MEHP, an active metabolite of DEHP in vivo). In addition, the expression of relevant pathway molecules was detected by immunohistochemistry, immunofluorescence, qRT-PCR, and Western blotting after the addition of the autophagy inhibitor 3-methyladenine (3-MA), the apoptosis inhibitor Z-VAD- FMK and the NF-κB inhibitor BAY11-7082. The current study found that autophagy and apoptosis were significantly activated in GCs of DEHP-induced atretic follicles in vivo and found that MEHP-induced KGN cells autophagy and apoptosis were independent and potentially cytotoxic of each other in vitro. Further studies confirmed that DEHP exposure resulted in LPS release from the intestinal tract and entering the ovary, thereby participating in DEHP-induced inflammation of GCs. In addition, we found that exogenous LPS synergized with MEHP could activate the NF-κB signaling pathway to induce inflammation and apoptosis of GCs in a relatively prolonged exposure condition. Meanwhile, inhibition of inflammatory activation could rescue apoptosis and estrogen secretion function of GCs induced by MEHP combined with LPS. These results indicated that the increased LPS influenced by DEHP might cooperate with MEHP to induce inflammatory apoptosis of GCs, an important cause of ovarian injury in mice.


Subject(s)
Apoptosis , Autophagy , Diethylhexyl Phthalate , Diethylhexyl Phthalate/analogs & derivatives , Granulosa Cells , Inflammation , Lipopolysaccharides , Female , Animals , Diethylhexyl Phthalate/toxicity , Autophagy/drug effects , Granulosa Cells/drug effects , Granulosa Cells/pathology , Lipopolysaccharides/toxicity , Apoptosis/drug effects , Mice , Inflammation/chemically induced , Inflammation/pathology , Reproduction/drug effects , Humans
18.
Mol Hum Reprod ; 30(5)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38603629

ABSTRACT

Polycystic ovary syndrome (PCOS) is one of the most common endocrine disorders in women of reproductive age, but its pathology has not been fully characterized and the optimal treatment strategy remains unclear. Cellular senescence is a permanent state of cell-cycle arrest that can be induced by multiple stresses. Senescent cells contribute to the pathogenesis of various diseases, owing to an alteration in secretory profile, termed 'senescence-associated secretory phenotype' (SASP), including with respect to pro-inflammatory cytokines. Senolytics, a class of drugs that selectively eliminate senescent cells, are now being used clinically, and a combination of dasatinib and quercetin (DQ) has been extensively used as a senolytic. We aimed to investigate whether cellular senescence is involved in the pathology of PCOS and whether DQ treatment has beneficial effects in patients with PCOS. We obtained ovaries from patients with or without PCOS, and established a mouse model of PCOS by injecting dehydroepiandrosterone. The expression of the senescence markers p16INK4a, p21, p53, γH2AX, and senescence-associated ß-galactosidase and the SASP-related factor interleukin-6 was significantly higher in the ovaries of patients with PCOS and PCOS mice than in controls. To evaluate the effects of hyperandrogenism and DQ on cellular senescence in vitro, we stimulated cultured human granulosa cells (GCs) with testosterone and treated them with DQ. The expression of markers of senescence and a SASP-related factor was increased by testosterone, and DQ reduced this increase. DQ reduced the expression of markers of senescence and a SASP-related factor in the ovaries of PCOS mice and improved their morphology. These results indicate that cellular senescence occurs in PCOS. Hyperandrogenism causes cellular senescence in GCs in PCOS, and senolytic treatment reduces the accumulation of senescent GCs and improves ovarian morphology under hyperandrogenism. Thus, DQ might represent a novel therapy for PCOS.


Subject(s)
Cellular Senescence , Granulosa Cells , Polycystic Ovary Syndrome , Quercetin , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/pathology , Female , Cellular Senescence/drug effects , Humans , Animals , Granulosa Cells/metabolism , Granulosa Cells/drug effects , Granulosa Cells/pathology , Quercetin/pharmacology , Mice , Senescence-Associated Secretory Phenotype , Adult , Dasatinib/pharmacology , Disease Models, Animal , Senotherapeutics/pharmacology , Hyperandrogenism/pathology , Hyperandrogenism/metabolism , Interleukin-6/metabolism , Dehydroepiandrosterone/pharmacology
19.
J Assist Reprod Genet ; 41(5): 1387-1401, 2024 May.
Article in English | MEDLINE | ID: mdl-38656738

ABSTRACT

OBJECTIVE: Women who are of reproductive age can suffer from polycystic ovary syndrome (PCOS), an endocrine disorder. Anovulatory infertility is mostly caused by aberrant follicular development, which is seen in PCOS patients. Due to the dysfunction of reproductive and endocrine function in PCOS patients, assisted reproduction treatment is one of the main means to obtain clinical pregnancy for PCOS patients. Long non-coding RNA (lncRNA) as a group of functional RNA molecules have been found to participate in the regulation of oocyte function, hormone metabolism, and proliferation and apoptosis of granulosa cells. In this study, we investigated the role of lncRNAs in follicular fluid-derived exosomes and the underlying mechanism of lncRNA LIPE-AS1. METHODS: We used RNA sequencing to analyze the lncRNA profiles of follicular fluid-derived exosomes in PCOS patients and controls. RT-qPCR was performed to detect the expression levels of these lncRNAs in control (n = 30) and PCOS (n = 30) FF exosome samples. Furthermore, we validated the performance of lncRNA LIPE-AS1 in oocyte maturation by in vitro maturation (IVM) experiments in mouse and steroid metabolism in granulosa cells. RESULTS: We found 501 lncRNAs were exclusively expressed in the control group and another 273 lncRNAs were found to be specifically expressed in the PCOS group. LncRNA LIPE-AS1, highly expressed in PCOS exosomes, was related to a poor oocyte maturation and embryo development in PCOS patients. Reduced number of MII oocytes were observed in the LIPE-AS1 group by in vitro maturation (IVM) experiments in mouse. LIPE-AS1 was also shown to modulate steroid metabolism and granulosa cell proliferation and apoptosis by LIPE-AS1/miR-4306/LHCGR axis. CONCLUSION: These findings suggested that the increased expression of LIPE-AS1, facilitated by follicular fluid exosomes, had a significant impact on both oocyte maturation and embryo development. We demonstrated the ceRNA mechanism involving LIPE-AS1, miR-4306, and LHCGR as a regulator of hormone production and metabolism. These findings indicate that LIPE-AS1 is essential in PCOS oocyte maturation and revealed a ceRNA network of LIPE-AS1 and provided new information on abnormal steroid metabolism and oocyte development in PCOS.


Subject(s)
Exosomes , Follicular Fluid , Granulosa Cells , Oocytes , Polycystic Ovary Syndrome , RNA, Long Noncoding , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/pathology , Polycystic Ovary Syndrome/metabolism , Female , Follicular Fluid/metabolism , RNA, Long Noncoding/genetics , Granulosa Cells/metabolism , Granulosa Cells/pathology , Humans , Exosomes/genetics , Exosomes/metabolism , Oocytes/metabolism , Oocytes/growth & development , Mice , Animals , In Vitro Oocyte Maturation Techniques , Adult , Steroids/metabolism , Oogenesis/genetics , Apoptosis/genetics , Cell Proliferation/genetics
20.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167182, 2024 06.
Article in English | MEDLINE | ID: mdl-38653359

ABSTRACT

OBJECTIVE: This work aimed to investigate the role of rhythm gene PER1 in mediating granulosa cell ferroptosis and lipid metabolism of polycystic ovary syndrome (PCOS). METHODS: We injected dehydroepiandrosterone and Ferrostatin-1 (Fer-1) into mice to explore the mechanism of ferroptosis in PCOS. The effect of PER1 on ferroptosis-like changes in granulosa cells was explored by overexpression of PER1 plasmid transfection and Fer-1 treatment. RESULTS: We found that Fer-1 ameliorated the characteristic polycystic ovary morphology, suppressed ferroptosis in the PCOS mice. PER1 and ALOX15 were highly expressed in PCOS, whereas SREBF2 was lowly expressed. Overexpression of PER1 decreased granulosa cell viability and inhibited proliferation. Meanwhile, overexpression of PER1 increased lipid reactive oxygen species, 4-Hydroxynonenal (4-HNE), Malondialdehyde (MDA), total Fe, and Fe2+ levels in granulosa cells and decreased Glutathione (GSH) content. Fer-1, SREBF2 overexpression, or ALOX15 silencing treatment reversed the effects of PER1 overexpression on granulosa cells. PER1 binds to the SREBF2 promoter and represses SREBF2 transcription. SREBF2 binds to the ALOX15 promoter and represses ALOX15 transcription. Correlation analysis of clinical trials showed that PER1 was positively correlated with total cholesterol, low-density lipoprotein cholesterol, luteinizing hormone, testosterone, 4-HNE, MDA, total Fe, Fe2+, and ALOX15. In contrast, PER1 was negatively correlated with SREBF2, high-density lipoprotein cholesterol, follicle-stimulating hormone, progesterone, and GSH. CONCLUSION: This study demonstrates that the rhythm gene PER1 promotes ferroptosis and dysfunctional lipid metabolism in granulosa cells in PCOS by inhibiting SREBF2/ALOX15 signaling.


Subject(s)
Ferroptosis , Granulosa Cells , Lipid Metabolism , Polycystic Ovary Syndrome , Animals , Female , Humans , Mice , Arachidonate 12-Lipoxygenase , Arachidonate 15-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/genetics , Cyclohexylamines/pharmacology , Dehydroepiandrosterone/metabolism , Ferroptosis/genetics , Granulosa Cells/metabolism , Granulosa Cells/pathology , Lipid Metabolism/genetics , Phenylenediamines/pharmacology , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/pathology , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL