Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 409
Filter
1.
Cell Mol Biol (Noisy-le-grand) ; 70(5): 226-232, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38814209

ABSTRACT

This study aims to analyze the active components and mechanism of Bushen Huoxue (BSHX) formula on the autoimmune premature ovarian insufficiency (POI) by combining network pharmacology and Transcriptomics. The active components and targets of BSHXF were screened through Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP). POI-related targets were identified through Therapeutic Targets Database (TTD), DisGeNET and drugbank database. The Veen diagram was performed to obtain the action targets. The active compound-target network and Protein-Protein Interaction (PPI) network were built by using STRING database and Cytoscape software. Key targets and active compounds were further identified by topological analysis. Molecular docking shows that Kaempferol, Isorhamnetin and Anhydroicaritin have strong binding to AKT. Finally, a zp3-induced autoimmune ovarian function deficiency mouse model was used to explore the potential mechanism of POI. The potential pathways of BSHXF for the treatment of POI were identified by Transcriptomic analysis. PI3K-AKT and NF-kb pathways were the common pathways between network pharmacology and transcriptomics. Our results revealed that BSHXF could reduce the FSH expression levels and raise the E2, and AMH levels in the serum. Western bloting demonstrates that BSHXF could upregulate the expression of p-PI3K and p-AKT.


Subject(s)
Drugs, Chinese Herbal , Molecular Docking Simulation , Network Pharmacology , Phosphatidylinositol 3-Kinases , Primary Ovarian Insufficiency , Protein Interaction Maps , Proto-Oncogene Proteins c-akt , Signal Transduction , Primary Ovarian Insufficiency/drug therapy , Primary Ovarian Insufficiency/genetics , Primary Ovarian Insufficiency/metabolism , Female , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Animals , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , Protein Interaction Maps/drug effects , Mice , Gene Expression Profiling , Transcriptome/drug effects , Transcriptome/genetics , Disease Models, Animal , Humans
2.
Reprod Toxicol ; 126: 108608, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38735593

ABSTRACT

Tripterygium wilfordii (TW) preparations have been utilized in China for treating rheumatoid arthritis and autoimmune diseases. However, their clinical use is limited due to reproductive toxicity, notably premature ovarian failure (POF). Our study aimed to investigate the effect and mechanism of bergenin in attenuating POF induced by triptolide in mice. POF was induced in female ICR mice via oral triptolide administration (50 µg/kg) for 60 days. Mice received bergenin (25, 50, 100 mg/kg, i.g.) or estradiol valerate (EV) (0.1 mg/kg, i.g.) daily, 1 h before triptolide treatment. In vitro, ovarian granulosa cells (OGCs) were exposed to triptolide (100 nM) and bergenin (1, 3, 10 µM). Antioxidant enzyme activity, protein expression, apoptosis rate, and reactive oxygen species (ROS) levels were assessed. The results showed that triptolide-treated mice exhibited evident atrophy, along with an increase in atretic follicles. Bergenin (50, 100 mg/kg) and EV (0.1 mg/kg), orally administered, exerted significant anti-POF effect. Bergenin and EV also decreased apoptosis in mouse ovaries. In vitro, bergenin (1, 3, 10 µM) attenuated triptolide-induced OGCs apoptosis by reducing levels of apoptosis-related proteins. Additionally, bergenin reduced oxidative stress through downregulation of antioxidant enzymes activity and overall ROS levels. Moreover, the combined use with Sh-Nrf2 resulted in a reduced protection of bergenin against triptolide-induced apoptosis of OGCs. Together, bergenin counteracts triptolide-caused POF in mice by inhibiting Nrf2-mediated oxidative stress and preventing OGC apoptosis. Combining bergenin with TW preparations may effectively reduce the risk of POF.


Subject(s)
Antioxidants , Apoptosis , Benzopyrans , Diterpenes , Epoxy Compounds , Granulosa Cells , Mice, Inbred ICR , Phenanthrenes , Primary Ovarian Insufficiency , Reactive Oxygen Species , Animals , Female , Primary Ovarian Insufficiency/chemically induced , Primary Ovarian Insufficiency/drug therapy , Primary Ovarian Insufficiency/prevention & control , Diterpenes/pharmacology , Phenanthrenes/toxicity , Phenanthrenes/pharmacology , Epoxy Compounds/toxicity , Antioxidants/pharmacology , Apoptosis/drug effects , Benzopyrans/pharmacology , Benzopyrans/therapeutic use , Reactive Oxygen Species/metabolism , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Oxidative Stress/drug effects , NF-E2-Related Factor 2/metabolism , Mice , Cells, Cultured
3.
Obstet Gynecol ; 143(6): e149-e152, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38574363

ABSTRACT

BACKGROUND: Since the onset of the coronavirus disease (COVID-19) pandemic, a variety of long-COVID-19 symptoms and autoimmune complications have been recognized. CASES: We report three cases of autoimmune premature poor ovarian response in patients aged 30-37 years after mild to asymptomatic COVID-19 before vaccination, with nucleotide antibody confirmation. Two patients failed to respond to maximum-dose gonadotropins for more than 4 weeks, despite a recent history of response before having COVID-19. After a month of prednisone 30 mg, these two patients had normal follicle-stimulating hormone (FSH) levels, high oocyte yield, and blastocyst formation in successful in vitro fertilization cycles. All three patients have above-average anti-müllerian hormone levels that persisted throughout their clinical ovarian insufficiency. Two patients had elevated FSH levels, perhaps resulting from FSH receptor blockade. One patient, with a history of high response to gonadotropins 75 international units per day and below-normal FSH levels, had no ovarian response to more than a month of gonadotropins (525 international units daily), suggesting autoimmune block of the FSH glycoprotein and possible FSH receptor blockade. CONCLUSION: Auto-antibody production in response to COVID-19 before vaccination may be a rare cause of autoimmune poor ovarian response. Although vaccination is likely protective, further study will be required to evaluate the effect of vaccination and duration of autoimmune FSH or FSH receptor blockade.


Subject(s)
COVID-19 , Primary Ovarian Insufficiency , Receptors, FSH , SARS-CoV-2 , Humans , Female , COVID-19/immunology , COVID-19/complications , Primary Ovarian Insufficiency/immunology , Primary Ovarian Insufficiency/drug therapy , Adult , SARS-CoV-2/immunology , Pandemics , Follicle Stimulating Hormone/blood , Coronavirus Infections/immunology , Coronavirus Infections/complications , Coronavirus Infections/drug therapy , Pneumonia, Viral/immunology , Pneumonia, Viral/complications , Pneumonia, Viral/drug therapy , Betacoronavirus
4.
J Ovarian Res ; 17(1): 89, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38671471

ABSTRACT

BACKGROUND: Yu Linzhu (YLZ) is a classical Chinese traditional formula, which has been used for more than 600 years to regulate menstruation to help pregnancy. However, the mechanism of modern scientific action of YLZ needs to be further studied. METHODS: Thirty SD female rats were divided into three groups to prepare the blank serum and drug-containing serum, and then using UHPLC-QE-MS to identify the ingredients of YLZ and its drug-containing serum. Twenty-four SD female rats were divided into four groups, except the control group, 4-vinylcyclohexene dicycloxide (VCD) was intraperitoneally injected to establish a primary ovarian insufficiency (POI) model of all groups. Using vaginal smear to show that the estrous cycle of rats was disturbed after modeling, indicates that the POI model was successfully established. The ELISA test was used to measure the follicle-stimulating hormone (FSH), estradiol (E2), and anti-Mullerian hormone (AMH) levels in the serum of rats. HE stain was used to assess the morphology of ovarian tissue. The localization and relative expression levels of CX43 protein were detected by tissue immunofluorescence. Primary ovarian granulosa cells (GCs) were identified by cellular immunofluorescence. CCK8 was used to screen time and concentration of drug-containing serum and evaluate the proliferation effect of YLZ on VCD-induced GCs. ATP kit and Seahorse XFe24 were used to detect energy production and real-time glycolytic metabolism rate of GCs. mRNA and protein expression levels of HIF1α, CX43, PEK, LDH, HK1 were detected by RT-PCR and WB. RESULTS: UHPLC-QE-MS found 1702 ingredients of YLZ and 80 constituents migrating to blood. YLZ reduced the FSH while increasing the AMH and E2 levels. In ovarian tissues, YLZ improved ovarian morphology, follicle development, and the relative expression of CX43. In vitro studies, we found that YLZ increased the proliferative activity of GCs, ATP levels, glycolytic metabolic rate, HIF1α, CX43, PEK, HK1, LDH mRNA, and protein levels. CONCLUSIONS: The study indicated that YLZ increased the proliferation and glycolytic energy metabolism of GCs to improve follicular development further alleviating ovarian function.


Subject(s)
Cell Proliferation , Connexin 43 , Disease Models, Animal , Drugs, Chinese Herbal , Energy Metabolism , Granulosa Cells , Hypoxia-Inducible Factor 1, alpha Subunit , Primary Ovarian Insufficiency , Animals , Female , Granulosa Cells/metabolism , Granulosa Cells/drug effects , Primary Ovarian Insufficiency/metabolism , Primary Ovarian Insufficiency/chemically induced , Primary Ovarian Insufficiency/drug therapy , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Rats , Cell Proliferation/drug effects , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Energy Metabolism/drug effects , Connexin 43/metabolism , Connexin 43/genetics , Rats, Sprague-Dawley , Signal Transduction/drug effects
5.
Phytomedicine ; 129: 155630, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38678952

ABSTRACT

BACKGROUND: Premature ovarian insufficiency (POI) is a tricky puzzle in the field of female reproductive medicine. Bushen Huoxue recipe (BHR), a traditional Chinese medicine compound based on the combination of kidney-tonifying and blood-activating functions, has shown excellent efficacy in improving female irregular menstruation, POI, and infertility. However, the potential mechanism of BHR in POI treatment has not yet been elucidated. Bone marrow mesenchymal stem cells (BMSCs), a type of pluripotent stem cells, have received increasing attention for their significant role in improving ovarian function and restoring fertility in women with POI. PURPOSE: This study aimed to evaluate the therapeutic effect of BHR in POI mice and explore its potential mechanism. METHODS: A POI mouse model was established with a single intraperitoneal injection of 120 mg/kg cyclophosphamide (CTX). Distilled water, BHR, or dehydroepiandrosterone was administered via gavage for 28 consecutive days. The effect of BHR on ovarian function in POI mice was evaluated by assessing the estrous cycle, ovarian morphology, follicular development, hormone levels, and angiogenesis. The proportion of BMSCs in bone marrow, peripheral blood, and ovary was analyzed via flow cytometry, and the level of molecules mediating migration and homing in ovary was measured. Cell viability assays, scratch healing assays and transwell migration assays were performed to explore the effect of BHR on BMSCs proliferation and migration in vitro, and its potential mechanism was explored. RESULTS: BHR significantly ameliorated estrous cycle disorders, hormone disorders, ovarian morphology, ovarian microvascular formation, and ovarian reserve in POI mice. Meanwhile, the number of BMSCs number in the bone marrow, peripheral blood, and ovary was apparently increased. Of note, BHR increased the level of hepatocyte growth factor (HGF)/cellular mesenchymal epithelial transition factor (cMET) and stromal cell-derived factor-1(SDF-1)/CXC chemokine receptor 4 (CXCR4) in the ovaries of POI mice. Moreover, BHR treatment promoted BMSCs proliferation and migration in vitro, with a significant increase in the level of proliferating cell nuclear antigen, cMET, and CXCR4. CONCLUSIONS: BHR effectively restored ovarian reserve, ovarian function, and ovarian angiogenesis in CTX-induced POI mice. In addition, BHR promoted BMSCs proliferation, migration, and homing to the ovary, which was mediated by the SDF-1/CXCR4 and HGF/cMET signaling axis. Finally, the amelioration of ovarian reserve and ovarian function in CTX-induced POI mice by BHR may be related to its promotion of endogenous BMSCs proliferation and homing.


Subject(s)
Cell Proliferation , Disease Models, Animal , Drugs, Chinese Herbal , Mesenchymal Stem Cells , Ovary , Primary Ovarian Insufficiency , Animals , Female , Drugs, Chinese Herbal/pharmacology , Primary Ovarian Insufficiency/drug therapy , Mesenchymal Stem Cells/drug effects , Ovary/drug effects , Cell Proliferation/drug effects , Mice , Cyclophosphamide , Estrous Cycle/drug effects , Cell Movement/drug effects
6.
Aging (Albany NY) ; 16(5): 4541-4562, 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38428403

ABSTRACT

Ningxin-Tongyu-Zishen formula (NTZF) is a clinical experience formula for the treatment of premature ovarian insufficiency (POI) in traditional Chinese medicine (TCM), and the potential mechanism is unknown. For in vivo experiments, POI mouse models (C57BL/6 mice), were constructed by subcutaneous injection of D-galactose (D-gal, 200 mg/kg). After treatment of NTZF (10.14, 20.27, 40.54 g/kg;) or estradiol valerate (0.15 mg/kg), ovarian function, oxidative stress (OS) and protein expression of Sirt1/p53 were evaluated. For in vitro experiments, H2O2 (200 µM) was used to treat KGN to construct ovarian granulosa cells (OGCs) cell senescence model. Pretreatment with NTZF (1.06 mg/mL) or p53 inhibitor (Pifithrin-α, 1 µM) was performed before induction of senescence, and further evaluated the cell senescence, OS, mRNA and protein expression of Sirt1/p53. In vivo, NTZF improved ovarian function, alleviated OS and Sirt1/p53 signaling abnormalities in POI mice. In vitro experiments showed that NTZF reduced the level of OS and alleviated the senescence of H2O2-induced KGN. In addition, NTZF activated the protein expression of Sirt1, inhibited the mRNA transcription and protein expression of p53 and p21. Alleviating OGCs senescence and protecting ovarian function through Sirt1/p53 is one of the potential mechanisms of NTZF in the treatment of POI.


Subject(s)
Galactose , Primary Ovarian Insufficiency , Humans , Female , Mice , Animals , Galactose/toxicity , Sirtuin 1/genetics , Sirtuin 1/metabolism , Hydrogen Peroxide/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Mice, Inbred C57BL , Primary Ovarian Insufficiency/chemically induced , Primary Ovarian Insufficiency/drug therapy , Primary Ovarian Insufficiency/genetics , Granulosa Cells/metabolism , Cellular Senescence , RNA, Messenger/metabolism
7.
J Ethnopharmacol ; 326: 117944, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38382656

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Panax ginseng C. A. Mey., one of the most used herbs in the world, shows effective treatment in reproductive injury. Recent studies have proven that the processed product, red ginseng, which is more active than ginseng itself. Therefore, it is speculated that its main functional component, rare ginsenosides (heat-transformed saponin, HTS), may be effective in treating premature ovarian failure (POF), but its efficacy has not yet been experimentally confirmed. AIM OF THE STUDY: To evaluate whether HTS could attenuate cyclophosphamide-induced inflammation and oxidative damage in POF model rats and the human granulosa-like KGN cell line and protect granulosa cell proliferation. MATERIAL AND METHODS: HTS were isolated from ginsenosides and high performance liquid chromatography (HPLC) analysis was used to analyze the HTS components. Cyclophosphamide (CP) was used to establish a POF rat model and KGN cell injury model. Reactive oxygen species (ROS) and antioxidant enzyme production was determined using specific assays, while inflammatory cytokine secretion was measured by enzyme-linked immunosorbent assay (ELISA). The proliferative function of granulosa cells was assessed using high-content screening and immunohistochemistry to determine the Ki67 protein level. Protein expression in ovarian tissues and KGN cells was analyzed by Western blotting, quantitative real-time PCR (qRT-PCR) was used to determine the transcriptional changes in ovarian tissues and KGN cells. RESULTS: In CP-treated POF model rats, HTS significantly decreased malondialdehyde (MDA), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α) levels, increased glutathione oxidase (GSH) levels, and upregulated Ki67 expression in ovarian granulosa cells. In addition, HTS significantly increased cell survival and Ki67 expression levels in CP-treated cells, and superoxide dismutase (SOD) levels were significantly increased. HTS significantly downregulated IL-6, TNF-α, and interleukin-1ß (IL-1ß) mRNA expression and significantly inhibited nuclear factor kappa-B p65 (NF-κB p65) and p38 mitogen activated protein kinase (p38 MAPK) phosphorylation in POF model rats and KGN cells. Moreover, NF-κB p65 and p38 MAPK levels were significantly increased in ovarian granulosa cells. p65 and p38 protein and gene expression was significantly downregulated. CONCLUSION: HTS ameliorated CP-induced POF and human granulosa cell injury, possibly by inhibiting inflammation and oxidative damage mediated by the p38 MAPK/NF-κB p65 signaling pathway.


Subject(s)
Ginsenosides , Primary Ovarian Insufficiency , Rats , Humans , Animals , Female , NF-kappa B/metabolism , Ginsenosides/pharmacology , Ginsenosides/therapeutic use , Interleukin-6/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Primary Ovarian Insufficiency/chemically induced , Primary Ovarian Insufficiency/drug therapy , Ki-67 Antigen/metabolism , MAP Kinase Signaling System , Inflammation/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
8.
J Ethnopharmacol ; 326: 117955, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38395181

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Gui Shen Wan (GSW) stands out as a promising therapeutic approach for addressing Premature Ovarian Insufficiency (POI). With deep roots in traditional medicine, GSW highlights the ethnopharmacological significance of herbal interventions in addressing nuanced aspects of women's health, with a specific emphasis on ovarian functionality. Recognizing the importance of GSW in gynecological contexts resonates with a rich tradition of using botanical formulations to navigate the intricacies of reproductive health. Delving into GSW's potential for treating POI emphasizes the crucial role of ethnopharmacological insights in guiding modern research endeavors. AIM OF THE STUDY: GSW is extensively utilized in gynecological disorders and has recently emerged as a potential therapeutic approach for POI. The present investigation aimed to assess the efficacy of GSW in treating POI in rats and elucidate its underlying molecular mechanisms. MATERIALS AND METHODS: The study employed GSW for POI treatment in rats. GSW, prepared as pills, underwent HPLC fingerprinting for quality control. Reagents and drugs, including VCD and dehydroepiandrosterone (DHEA), were sourced from reputable providers. Eighty Sprague-Dawley rats were categorized into groups for POI induction and treatment. Ovarian tissue underwent HE staining, immunohistochemical staining, Western Blot, qRT-PCR, and vaginal secretion testing. ELISA was utilized for target molecule detection. This methodology ensures a robust and reliable experimental framework. RESULTS: The results highlight a robust collaborative improvement in POI among rats subjected to combined GSW and DHEA treatment. Particularly noteworthy is the substantial enhancement in the expression of vascular regeneration-related molecules-VDR-Klotho-VEGFR-accompanied by a significant elevation in autophagy levels. Post-GSW administration, rat ovarian morphology demonstrated increased stability, hormone levels exhibited more consistent maintenance, and there was a marked reduction in inflammatory response compared to other groups (p < 0.01). Furthermore, GSW intervention resulted in a more pronounced upregulation of ovarian autophagy (p < 0.05). CONCLUSION: By modulating VDR-Klotho signaling, GSW exerts regulatory control over ovarian autophagy and vascular regeneration, thereby mitigating the occurrence and progression of POI in rats.


Subject(s)
Menopause, Premature , Primary Ovarian Insufficiency , Humans , Rats , Female , Animals , Angiogenesis , Rats, Sprague-Dawley , Primary Ovarian Insufficiency/drug therapy , Primary Ovarian Insufficiency/metabolism , Dehydroepiandrosterone/therapeutic use , Receptors, Calcitriol
9.
Free Radic Res ; 58(2): 107-116, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38408280

ABSTRACT

BACKGROUND: Oxidative stress injury is an important pathological factor of premature ovarian failure (POF). Salidroside, extracted from the Chinese herb-Rhodiola rosea, has advantages in antioxidant characteristics. However, their therapeutic efficacy and mechanisms in POF have not been explored. PURPOSE: This study aims to assess the therapeutic effects of salidroside in chemotherapy-induced ovarian failure rats. METHODS: A POF rat model was established by injection of cyclophosphamide, followed by treatment with salidroside. The therapeutic effect of salidroside was evaluated based on hormone levels, follicle count, and reproductive ability. Oxidative stress injury was assessed by the detection of SOD enzyme activity and MDA levels. Differential gene expression of Keap1, Nrf2, HMOX1, NQO1, AMH, BMP15, and GDF9, were identified by qRT­PCR. The protein expression of Keap1, Nrf2, P53, and Bcl-2 were detected by western blot. RESULTS: Salidroside treatment markedly restored FSH, E2, and AMH hormone secretion levels, reduced follicular atresia, and increased antral follicle numbers in POF rats. In addition, salidroside improves fertility in POF rats, activates the Nrf2 signaling pathway, and reduces the level of oxidative stress. The recovery function of high dose salidroside (50 mg/kg) in a reproductive assay was significantly improved than that of lower dose salidroside (25 mg/kg). Meanwhile, the safety evaluation of salidroside treatment in rats showed that salidroside was safe for POF rats at doses of 25-50 mg/kg. CONCLUSIONS: Salidroside therapy improved premature ovarian failure significantly through antioxidant function and activating Nrf2 signaling.


Subject(s)
Glucosides , Phenols , Primary Ovarian Insufficiency , Humans , Rats , Female , Animals , Primary Ovarian Insufficiency/chemically induced , Primary Ovarian Insufficiency/drug therapy , Primary Ovarian Insufficiency/pathology , Kelch-Like ECH-Associated Protein 1 , Antioxidants/pharmacology , Antioxidants/therapeutic use , NF-E2-Related Factor 2 , Follicular Atresia , Cyclophosphamide/adverse effects , Hormones
10.
Mol Nutr Food Res ; 68(5): e2300784, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38314939

ABSTRACT

SCOPE: Premature ovarian insufficiency (POI) is a common female infertility problem, with its pathogenesis remains unknown. The NOD-like receptor family pyrin domain-containing 3 (NLRP3)-mediated pyroptosis has been proposed as a possible mechanism in POI. This study investigates the therapeutic effect of α-ketoglutarate (AKG) on ovarian reserve function in POI rats and further explores the potential molecular mechanisms. METHODS AND RESULTS: POI rats are caused by administration of cyclophosphamide (CTX) to determine whether AKG has a protective effect. AKG treatment increases the ovarian index, maintains both serum hormone levels and follicle number, and improves the ovarian reserve function in POI rats, as evidence by increased the level of lactate and the expression of rate-limiting enzymes of glycolysis in the ovaries, additionally reduced the expression of NLRP3, Gasdermin D (GSDMD), Caspase-1, Interleukin-18 (IL-18), and Interleukin-1 beta (IL-1ß). In vitro, KGN cells are treated with LPS and nigericin to mimic pyroptosis, then treated with AKG and MCC950. AKG inhibits inflammatory and pyroptosis factors such as NLRP3, restores the glycolysis process in vitro, meanwhile inhibition of NLRP3 has the same effect. CONCLUSION: AKG ameliorates CTX-induced POI by inhibiting NLRP3-mediated pyroptosis, which provides a new therapeutic strategy and drug target for clinical POI patients.


Subject(s)
Ovarian Reserve , Primary Ovarian Insufficiency , Humans , Rats , Female , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Ketoglutaric Acids/pharmacology , Primary Ovarian Insufficiency/chemically induced , Primary Ovarian Insufficiency/drug therapy , Pyroptosis , Granulosa Cells/metabolism , Inflammasomes/metabolism
11.
Adv Med Sci ; 69(1): 70-80, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38387407

ABSTRACT

PURPOSE: Metformin (MET), a first-line treatment for type 2 diabetes mellitus, restores ovarian function in women with polycystic ovary syndrome. MET has been shown to increase the rate of success for in vitro fertilization when utilized in assisted reproductive technologies. This study was designed to examine the impact of MET on ovarian function and fertility in a mouse model of galactose-induced premature ovarian insufficiency (POI). We further investigated the underlying mechanisms. MATERIALS AND METHODS: Female mice were divided into 4 groups: saline, d-galactose, d-galactose â€‹+ â€‹MET, and MET. Body weight, ovarian index, and fertility were assessed. The hormonal profile was done. Advanced glycation end products (AGEPs), receptor for advanced glycation end products (RAGE), phosphoinositide 3-kinase (PI3K), protein kinase B (Akt), forkhead box O3a (FOXO3a) expression were measured. Ovarian follicle counting and morphology were analyzed. Immunohistochemistry of cleaved caspase-3 expression was performed. RESULTS: Our findings demonstrated that MET reversed irregularities in the estrus cycle, enhanced the ovarian index, and improved the abnormal levels of hormones and AGEs induced by d-galactose. Furthermore, the expression levels of PI3K, Akt, FOXO3a, and RAGE were upregulated with d-galactose. However, MET attenuated their expression levels. The primordial follicles ratio was improved, whereas atretic follicles and apoptotic-related cleaved caspase-3 expression were decreased in the d-galactose â€‹+ â€‹MET group compared to the d-galactose group. CONCLUSION: This study demonstrates that MET partially rescued ovarian dysfunction and apoptosis induced by d-galactose via a mechanism involving PI3K-Akt-FOXO3a pathway. Our finding proposed that MET may be a promising alternative treatment for POI.


Subject(s)
Forkhead Box Protein O3 , Galactose , Metformin , Phosphatidylinositol 3-Kinases , Primary Ovarian Insufficiency , Proto-Oncogene Proteins c-akt , Signal Transduction , Female , Animals , Primary Ovarian Insufficiency/drug therapy , Primary Ovarian Insufficiency/chemically induced , Primary Ovarian Insufficiency/metabolism , Primary Ovarian Insufficiency/pathology , Forkhead Box Protein O3/metabolism , Mice , Metformin/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , Disease Models, Animal , Apoptosis/drug effects
12.
Gene ; 901: 148128, 2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38181927

ABSTRACT

Cyclophosphamide (CP), as an anti-cancer drug, is frequently used to treat various types of cancer. A decreased number of ovarian follicles impaired normal ovarian function, and subsequent premature ovarian failure (POF) presented as a side effect of cyclophosphamide usage. These events may eventually affect the fertility rate of individuals. The present study showed the effect of cyclophosphamide on ovarian reserves and the protective effect of L-carnitine (LC) as an antioxidant to prevent POF. To design the study, six to eight-week-old NMRI female mice were divided into three groups: control, cyclophosphamide (CP), and cyclophosphamide + L-carnitine (CP + LC). Mice received drugs intraperitoneally (IP) for 21 days. In the following 24 h after the last injection, both ovaries were used to evaluate the expression of Sohlh1 and Lhx8 genes by Real-time PCR. Furthermore, the alteration of Lhx8 promoter methylation was examined by Methylation-sensitive high-resolution melting analysis (MS-HRM). The present data showed the negative effect of CP on regulator genes of oogenesis including Sohlh1 and Lhx8. In addition, an examination of the epigenetic status of the Lhx8 gene showed a change in promoter methylation of this gene following cyclophosphamide injection. Although, L-carnitine is an effective antioxidant in relieving oxidative stress caused by cyclophosphamide and its damage, in the present study, however, the use of L-carnitine failed to protect the ovaries from changes caused by CP injection. So, using cyclophosphamide can alter the expression of folliculogenesis genes through its effects on epigenetic changes and may cause POF. The results of the present study showed that L-carnitine consumption can't protect the ovaries against the adverse effects of CP.


Subject(s)
Antioxidants , Primary Ovarian Insufficiency , Humans , Mice , Female , Animals , Antioxidants/pharmacology , Transcription Factors , Carnitine/pharmacology , Carnitine/therapeutic use , Cyclophosphamide/adverse effects , Primary Ovarian Insufficiency/chemically induced , Primary Ovarian Insufficiency/drug therapy , Primary Ovarian Insufficiency/genetics , Epigenesis, Genetic , Real-Time Polymerase Chain Reaction
13.
J Ethnopharmacol ; 323: 117718, 2024 Apr 06.
Article in English | MEDLINE | ID: mdl-38181933

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: It has been reported that apoptosis and oxidative stress are related to cyclophosphamide (CYC)-induced premature ovarian failure (POF). Therefore, anti-apoptotic and anti-oxidative stress treatments exhibit therapeutic efficacy in CYC-induced POF. Danggui Shaoyao San (DSS), which has been extensively used to treat gynecologic diseases, is found to inhibit apoptosis and reduce oxidative stress. However, the roles of DSS in regulating apoptosis and oxidative stress during CYC-induced POF, and its associated mechanisms are still unknown. AIM OF THE STUDY: This work aimed to investigate the roles and mechanisms of DSS in inhibiting apoptosis and oxidative stress in CYC-induced POF. MATERIALS AND METHODS: CYC (75 mg/kg) was intraperitoneally injected in mice to construct the POF mouse model for in vivo study. Thereafter, alterations of body weight, ovary morphology and estrous cycle were monitored to assess the ovarian protective properties of DSS. Serum LH and E2 levels were analyzed by enzyme-linked immunosorbent assay (ELISA). Hematoxylin-eosin (HE) staining was employed for examining ovarian pathological morphology and quantifying follicles in various stages. Meanwhile, TUNEL staining and apoptosis-related proteins were adopted for evaluating apoptosis. Oxidative stress was measured by the levels of ROS, MDA, and 4-HNE. Western blot (WB) assay was performed to detect proteins related to the SIRT1/p53 pathway. KGN cells were used for in vitro experiment. TBHP stimulation was carried out for establishing the oxidative stress-induced apoptosis cell model. Furthermore, MTT assay was employed for evaluating the protection of DSS from TBHP-induced oxidative stress. The anti-apoptotic ability of DSS was evaluated by hoechst/PI staining, JC-1 staining, and apoptosis-related proteins. Additionally, the anti-oxidative stress ability of DSS was measured by detecting the levels of ROS, MDA, and 4-HNE. Proteins related to SIRT1/p53 signaling pathway were also measured using WB and immunofluorescence (IF) staining. Besides, SIRT1 expression was suppressed by EX527 to further investigate the role of SIRT1 in the effects of DSS against apoptosis and oxidative stress. RESULTS: In the in vivo experiment, DSS dose-dependently exerted its anti-apoptotic, anti-oxidative stress, and ovarian protective effects. In addition, apoptosis, apoptosis-related protein and oxidative stress levels were inhibited by DSS treatment. DSS treatment up-regulated SIRT1 and down-regulated p53 expression. From in vitro experiment, it was found that DSS treatment protected KGN cells from TBHP-induced oxidative stress injury. Besides, DSS administration suppressed the apoptosis ratio, apoptosis-related protein levels, mitochondrial membrane potential damage, and oxidative stress. SIRT1 suppression by EX527 abolished the anti-apoptotic, anti-oxidative stress, and ovarian protective effects, as discovered from in vivo and in vitro experiments. CONCLUSIONS: DSS exerts the anti-apoptotic, anti-oxidative stress, and ovarian protective effects in POF mice, and suppresses the apoptosis and oxidative stress of KGN cells through activating SIRT1 and suppressing p53 pathway.


Subject(s)
Menopause, Premature , Primary Ovarian Insufficiency , Humans , Female , Mice , Animals , Primary Ovarian Insufficiency/chemically induced , Primary Ovarian Insufficiency/drug therapy , Primary Ovarian Insufficiency/prevention & control , Tumor Suppressor Protein p53/metabolism , Reactive Oxygen Species/metabolism , Sirtuin 1/metabolism , Oxidative Stress , Apoptosis , Cyclophosphamide/toxicity , Signal Transduction
14.
J Ovarian Res ; 17(1): 24, 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38273341

ABSTRACT

Premature ovarian failure (POF) is a leading cause of women's infertility without effective treatment. The purpose of this study was to investigate the protective effects of Luffa cylindrica fermentation liquid (LF) on cyclophosphamide (CTX) -induced POF in mice and to preliminarily investigate the underlying mechanisms. Thirty-two Balb/c mice were divided into four groups randomly. One group served as the control, while the other three received CTX injections to establish POF models. A 14-day gavage of either 5 or 10 µL/g LF was administered to two LF pretreatment groups. To analyze the effects of LF, the ovarian index, follicle number, the levels of serum sex hormones, superoxide dismutase (SOD), glutathione (GSH), malondialdehyde (MDA), inflammatory factors, and apoptosis of the ovarian cells were measured. The effects of LF pretreatment on the expression of TLR4/NF-κB and apoptosis pathways were also evaluated. We found that LF pretreatment increased the ovarian index and the number of primordial and antral follicles while decreasing those of atretic follicles. LF pretreatment also increased the serum levels of estradiol (E2) and anti-Müllerian hormone (AMH), while decreasing those of luteinizing hormone (LH) and follicle-stimulating hormone (FSH). Furthermore, LF pretreatment increased the levels of SOD and GSH in the ovaries, while decreasing those of MDA, tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß). LF administration reduced the amount of TUNEL+ ovarian cells and the levels of TLR4 and NF-κB P65 protein expression. In conclusion, LF has antioxidant, anti-inflammatory as well as anti-apoptotic effects against CTX-induced POF, and the inhibition of TLR4/NF-κB and apoptosis pathways may be involved in its mechanisms.


Subject(s)
Luffa , Menopause, Premature , Primary Ovarian Insufficiency , Humans , Female , Mice , Animals , Primary Ovarian Insufficiency/chemically induced , Primary Ovarian Insufficiency/drug therapy , Primary Ovarian Insufficiency/metabolism , Luffa/metabolism , NF-kappa B/metabolism , Fermentation , Toll-Like Receptor 4/metabolism , Cyclophosphamide/toxicity , Oxidative Stress , Apoptosis , Inflammation/chemically induced , Inflammation/drug therapy , Glutathione , Superoxide Dismutase/metabolism
15.
Sci Rep ; 14(1): 1447, 2024 01 16.
Article in English | MEDLINE | ID: mdl-38228655

ABSTRACT

Premature ovarian failure (POF) caused by chemotherapy is a growing concern for female reproductive health. The use of metformin (MET), which has anti-oxidative and anti-inflammatory effects, in the treatment of POF damaged by chemotherapy drugs remains unclear. In this study, we investigated the impact of MET on POF caused by cyclophosphamide (CTX) combined with busulfan (BUS) and M1 macrophages using POF model mice and primary granule cells (GCs). Our findings demonstrate that intragastric administration of MET ameliorates ovarian damage and alleviates hormonal disruption in chemotherapy-induced POF mice. This effect is achieved through the reduction of inflammatory and oxidative stress-related harm. Additionally, MET significantly relieves abnormal inflammatory response, ROS accumulation, and senescence in primary GCs co-cultured with M1 macrophages. We also observed that this protective role of MET is closely associated with the AMPK/PPAR-γ/SIRT1 pathway in cell models. In conclusion, our results suggest that MET can protect against chemotherapy-induced ovarian injury by inducing the expression of the AMPK pathway while reducing oxidative damage and inflammation.


Subject(s)
Antineoplastic Agents , Metformin , Primary Ovarian Insufficiency , Humans , Mice , Female , Animals , Primary Ovarian Insufficiency/chemically induced , Primary Ovarian Insufficiency/prevention & control , Primary Ovarian Insufficiency/drug therapy , AMP-Activated Protein Kinases/metabolism , Peroxisome Proliferator-Activated Receptors/metabolism , Sirtuin 1/genetics , Sirtuin 1/metabolism , Metformin/therapeutic use , Granulosa Cells/metabolism , Antineoplastic Agents/pharmacology
16.
Aging (Albany NY) ; 16(1): 844-856, 2024 Jan 10.
Article in English | MEDLINE | ID: mdl-38206302

ABSTRACT

Premature ovarian insufficiency (POI) is a diverse form of female infertility characterized by a decline in ovarian function before the age of 40. Melatonin (MT) is a potential clinical treatment for restoring or safeguarding ovarian function in POI. However, the specific therapeutic mechanism underlying this effect remains unclear. To address this, we conducted experiments using human granulosa cells (GCs) from both POI and normal patients. We examined the expression levels of autophagy-related genes and proteins in GCs through qRT-PCR and western blot analysis. Autophagy flux was monitored in GCs infected with GFP-LC3-adenovirus, and the regulatory function of MT in autophagy was investigated. Additionally, we employed pharmacological intervention of autophagy using 3-Methyladenine (3-MA) and RNA interference of Forkhead box O-3A (FOXO3A) to elucidate the mechanism of MT in the autophagy process. Compared to GCs from normal patients, GCs from POI patients exhibited irregular morphology, decreased proliferation, increased apoptosis, and elevated ROS levels. The expression of autophagy-related genes was downregulated in POI GCs, resulting in reduced autophagic activity. Furthermore, MT levels were decreased in POI GCs, but exogenous MT effectively activated autophagy. Mechanistically, melatonin treatment downregulated FOXO3A expression and induced phosphorylation in POI GCs. Importantly, silencing FOXO3A abolished the protective effect of melatonin on GCs. These findings indicate that autophagy is downregulated in POI GCs, accompanied by a deficiency in MT. Moreover, we demonstrated that supplementing MT can rescue autophagy levels and enhance GC viability through the activation of FOXO3A signaling. Thus, MT-FOXO3A may serve as a potential therapeutic target for POI treatment.


Subject(s)
Melatonin , Primary Ovarian Insufficiency , Female , Humans , Autophagy , Granulosa Cells/metabolism , Melatonin/pharmacology , Melatonin/therapeutic use , Primary Ovarian Insufficiency/drug therapy , Primary Ovarian Insufficiency/genetics , Primary Ovarian Insufficiency/metabolism , Signal Transduction
17.
Biochem Biophys Res Commun ; 696: 149506, 2024 Feb 12.
Article in English | MEDLINE | ID: mdl-38224665

ABSTRACT

AIM: To evaluate the synergistic effect of combined treatment with melatonin (MEL) and resveratrol (RES) in cisplatin (CIS)-induced premature ovarian failure (POF) model in rats and to elucidate the molecular mechanism of this therapeutic effect. MATERIAL & METHODS: Female Sprague Dawley rats were divided into 7 experimental groups as follows; CONT (Control), CIS, MEL, RES, POF + MEL, POF + RES, and POF + MEL + RES. H&E staining was performed to evaluate follicular cell vacuolization/degeneration, vascular congestion/hemorrhage, and inflammation, by using an ordinal scale from 0 to 4 to grade the severity of observed changes (0 = normal, 1 = mild, 2 = moderate, 3 = severe, 4 = very severe). Zona pellucida integrity and connective tissue amount in the ovarian tissue were detected using PAS & Masson Trichrome staining. The immunofluorescence method was used to determine the immune localizations of pH2Ax, SIRT1, FOXO3a, and BCL2. The connective tissue amounts and immunoreactivity staining intensities were measured using ImageJ. The gene expression of SIRT1, FOXO3a, and BCL2 was determined using RT-PCR. Serum estrogen hormone levels were measured by ELISA. Statistically, Bonferroni correction was performed, and p < 0.002 were considered significant. RESULTS: A significant difference was observed in the POF group compared to the CONT group in all parameters except tertiary follicle count and hemorrhage. The decrease in the number of atretic follicles in the POF + MEL + RES group was found significant compared to both POF + MEL and POF + RES groups. The expression of pH2Ax, SIRT1, FOXO3a, and BCL2 at the protein level and SIRT1 and BCL2 at the mRNA level were significant in the POF + MEL + RES group compared to the POF group. Between the single and combination treatment groups, the difference in protein level was found in pH2Ax, SIRT1, FOXO3a, and BCL2 expression. The POF + MEL + RES group exhibited significantly higher SIRT1 mRNA expression compared to the groups receiving single treatments. CONCLUSION: The present study provides evidence that MEL and RES have synergistic effects in preventing the decrease in follicle reserve and increase in DNA break (pH2Ax) and follicle atresia in POF ovaries. This therapeutic effect is mediated by SIRT1 overexpression and activation of the SIRT1/FOXO3a/BCL2 pathway.


Subject(s)
Melatonin , Primary Ovarian Insufficiency , Humans , Female , Rats , Animals , Primary Ovarian Insufficiency/drug therapy , Primary Ovarian Insufficiency/chemically induced , Resveratrol , Melatonin/pharmacology , Melatonin/therapeutic use , Sirtuin 1/genetics , Sirtuin 1/metabolism , Rats, Sprague-Dawley , Cisplatin/therapeutic use , Hemorrhage , RNA, Messenger , Proto-Oncogene Proteins c-bcl-2
18.
J Ethnopharmacol ; 324: 117782, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38272104

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: The Zishen Yutai pills (ZYP), a Chinese medicinal formulation derived from the Qing Dynasty prescription "Shou Tai pills", have been documented to exhibit beneficial effects in clinical observations treating premature ovarian failure (POF). However, the anti-POF effects and its comprehensive systemic mechanism have not yet been clarified. AIM OF THE REVIEW: Therapeutic effects and systemic mechanism of ZYP in POF were evaluated. MATERIALS AND METHODS: After pulverization, sieving, and stirring, ZYP was administered intragastrically to cisplatin-induced POF mice at a dose of 1.95 mg/kg/d for 14 days. The anti-POF effects of ZYP were investigated by assessing the number of ovarian follicles at different developmental stages, as well as measuring serum estradiol (E2) levels and ovarian-expressed anti-Müllerian hormone (AMH). Reproductive performance and offspring health were evaluated to predict fertility restoration. Furthermore, a combination of proteomic and metabolomic profiling was employed to elucidate the underlying molecular mechanism of ZYP in treating POF. Western blot (WB) analyses and real-time quantitative polymerase chain reaction (RT-qPCR) were conducted to explore the mechanisms through which ZYP exerted its anti-POF effects. RESULTS: We have demonstrated that oral administration of ZYP reversed the reduction in follicles at different developmental stages and stimulated the expressions of serum E2 and ovarian-expressed AMH in a cisplatin-induced POF model. Additionally, ZYP ameliorated follicle apoptosis in ovaries affected by cisplatin-induced POF. Furthermore, treatment with ZYP restored the quantity and quality of oocytes, as well as enhanced fertility. Our results revealed 62 differentially expressed proteins (DEPs) through proteomic analyses and identified 26 differentially expressed metabolites (DEMs) through metabolomic analyses. Both DEPs and DEMs were highly enriched in the arachidonic acid (AA) metabolism pathway. ZYP treatment effectively upregulated the protein and mRNA expression of critical targets in AA metabolism and the AKT pathway, including CYP17α1, HSD3ß1, LHR, STAR, and AKT, in cisplatin-induced POF mice. CONCLUSIONS: These results indicated that ZYP exerted protective effects against POF and restored fertility from cisplatin-induced apoptosis. ZYP could be a satisfying alternative treating POF.


Subject(s)
Drugs, Chinese Herbal , Menopause, Premature , Primary Ovarian Insufficiency , Female , Humans , Mice , Animals , Primary Ovarian Insufficiency/chemically induced , Primary Ovarian Insufficiency/drug therapy , Primary Ovarian Insufficiency/metabolism , Arachidonic Acid , Proto-Oncogene Proteins c-akt/metabolism , Cisplatin/adverse effects , Proteomics , Fertility , Anti-Mullerian Hormone
19.
Menopause ; 31(1): 65-67, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-38086010

ABSTRACT

Primary ovarian insufficiency (sometimes known as premature ovarian insufficiency) is a result of loss of ovarian follicular activity before the age of 40 years. It is an endocrine deficiency state in women, characterized by premature estrogen deprivation. In the absence of estrogen replacement, women experience bothersome menopause symptoms and a predisposition to accelerated aging and multimorbidity accumulation. Unless a true contraindication exists, estrogen therapy is recommended at least until the age of natural menopause. This Practice Pearl summarizes the clinical manifestations, diagnostic evaluation, and management of primary ovarian insufficiency.


Subject(s)
Menopause, Premature , Primary Ovarian Insufficiency , Female , Humans , Adult , Primary Ovarian Insufficiency/diagnosis , Primary Ovarian Insufficiency/genetics , Primary Ovarian Insufficiency/drug therapy , Estrogen Replacement Therapy , Menopause , Estrogens/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...