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

Complementary Medicines
Country/Region as subject
Publication year range
1.
J Ovarian Res ; 17(1): 29, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38302986

ABSTRACT

BACKGROUND: Polycystic ovary syndrome (PCOS) is a frequent and complicated endocrine disease that remains a major reason for infertility. Bushenhuoluo Decotion (BSHLD) has been validated to exhibit curative effects on PCOS. This study was aimed to explore the potential mechanism underlying the therapeutic action of BSHLD. METHODS: PCOS rat model was induced by dehydroepiandrosterone (DHEA). Serum hormone and cytokines levels and ovarian pathological alterations were measured to assess ovarian function. Exosomes (Exos) were identified by Transmission electron microscopy and Nanoparticle Tracking Analysis. RT-qPCR, Western blotting, immunohistochemical staining, and immunofluorescence staining were performed to detect molecule expressions. Proliferation and pyroptosis of granulosa cells (GCs) were evaluated by CCK-8 and flow cytometry, respectively. The binding relationship between miR-30a-5p and suppressor of cytokine signaling 3 (SOCS3) was verified by dual luciferase reporter and RIP assays. RESULTS: BSHLD treatment improved serum hormone abnormality, insulin sensitivity, and ovarian morphologic changes of PCOS rats. Moreover, BSHLD treatment restrained the excessive autophagy and pyroptosis in ovarian tissues of PCOS rats. Moreover, BSHLD reduced the expression of miR-30a-5p in serum, serum-derived Exos, and ovarian tissues, thus inhibiting autophagy and NLRP3-mediated pyroptosis in GCs. Mechanistically, SOCS3 was proved as a target of miR-30a-5p and could activate mTOR/P70S6K pathway to repress autophagy. The inhibitory effect of miR-30a-5p deficiency on autophagy and pyroptosis of GCs was attenuated by rapamycin. CONCLUSION: Collectively, BSHLD suppressed autophagy and pyroptosis to improve POCS by regulating exosomal miR-30a-5p/SOCS3/mTOR signaling.


Subject(s)
Drugs, Chinese Herbal , MicroRNAs , Plant Extracts , Polycystic Ovary Syndrome , Animals , Female , Humans , Rats , Autophagy , Hormones , MicroRNAs/genetics , MicroRNAs/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Polycystic Ovary Syndrome/pathology , Pyroptosis , Suppressor of Cytokine Signaling 3 Protein/genetics , Suppressor of Cytokine Signaling 3 Protein/metabolism , TOR Serine-Threonine Kinases/metabolism , Plant Extracts/therapeutic use , Drugs, Chinese Herbal/therapeutic use
2.
Gynecol Endocrinol ; 39(1): 2247098, 2023 Aug 09.
Article in English | MEDLINE | ID: mdl-37573873

ABSTRACT

BACKGROUND: Anti-Müllerian hormone (AMH) has recently emerged as a promising biomarker for the detection of polycystic ovarian morphology. In polycystic ovary syndrome (PCOS), an elevated level of AMH has been suggested to add value to the Rotterdam criteria in cases of diagnostic uncertainty. In this study, we evaluated the correlation between AMH and PCOS, and the potential role of AMH in PCOS diagnosis. METHODS: A case-control study was performed on a total of 200 females, 100 of which were diagnosed with PCOS as per Rotterdam revised criteria (2003) and 100 as the control (non-PCOS group). Patient medical records were therefore retrieved for clinical, biochemical and ultrasound markers for PCOS diagnosis. Sensitivity, specificity, area under receiver operating characteristic (AUROC) curve, and multivariate linear regression models were applied to analyze our data. RESULTS: Mean serum levels of LH and AMH, and LH/FSH ratio were significantly different between compared groups. In the PCOS group, the mean serum AMH level was 6.78 ng/mL and LH/FSH ratio was 1.53 while those of controls were 2.73 ng/mL and 0.53, respectively (p < .001). The most suitable compromise between 81% specificity and 79% sensitivity was obtained with a cutoff value of 3.75 ng/mL (26.78 pmol/L) serum AMH concentration for PCOS prediction, with an AUROC curve of 0.9691. CONCLUSION: Serum AMH cutoff level of 3.75 ng/mL was identified as a convenient gauge for the prediction of PCOS and an adjuvant to the Rotterdam criteria.


Subject(s)
Anti-Mullerian Hormone , Polycystic Ovary Syndrome , Adult , Female , Humans , Anti-Mullerian Hormone/blood , Follicle Stimulating Hormone/blood , Luteinizing Hormone/blood , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/diagnosis , Polycystic Ovary Syndrome/pathology , Prolactin/blood , Sensitivity and Specificity , Vitamin D/blood , Case-Control Studies , Menstruation Disturbances/pathology
3.
Minerva Endocrinol (Torino) ; 48(2): 160-171, 2023 06.
Article in English | MEDLINE | ID: mdl-33103870

ABSTRACT

BACKGROUND: Polycystic ovary syndrome (PCOS) is a complex endocrine disorder with well-established metabolic abnormalities. In the present study, untargeted metabolomics technology was applied to analyze the serum and follicular fluid samples from women with polycystic ovary syndrome and healthy controls using 1H nuclear magnetic resonance (NMR). METHODS: Seventy samples for PCOS analysis were collected in hospital of Shandong University of Traditional Chinese Medicine (Jinan, China), NMR was used as analytical technology and multivariate analysis was applied to analyze metabolomics difference in PCOS and healthy controls. RESULTS: Significant metabolic differences were found in both serum and follicular fluid samples with orthogonal partial least-squares discriminant analysis (OPLS-DA). Three discriminated metabolites (1-Methylhistidine, threonine and Citrate) in both serum and follicular fluid were altered in PCOS patients. Abnormal energy metabolism, lipid metabolism and amino acid metabolism were detected in PCOS patients. Furthermore, more significantly changed amino acids were discovered in follicular fluid samples. CONCLUSIONS: Our findings would provide a resource for further investigations on metabolic disturbance in PCOS patients.


Subject(s)
Polycystic Ovary Syndrome , Humans , Female , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/pathology , Follicular Fluid/metabolism , Metabolomics , Magnetic Resonance Spectroscopy , Amino Acids/metabolism
4.
Phytother Res ; 37(4): 1449-1461, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36450691

ABSTRACT

Polycystic ovarian syndrome (PCOS) is a hormonal disorder that causes enlargement of ovaries and follicular maturation arrest, which lacks efficient treatment. N2, a semi-natural triterpenoid from the neem family, was already reported to have antioxidant and antiinflammatory properties in our previous report. This study investigated the anti-androgenic property of N2 on testosterone-induced oxidative stress in Chinese Hamster Ovarian cells (CHO) and PCOS zebrafish model. The testosterone exposure disrupted the antioxidant enzymes and ROS level and enhanced the apoptosis in both CHO cells and PCOS zebrafish. However, N2 significantly protected the CHO cells from ROS and apoptosis. N2 improved the Gonado somatic index (GSI) and upregulated the expression of the SOD enzyme in zebrafish ovaries. Moreover, the testosterone-induced follicular maturation arrest was normalized by N2 treatment in histopathology studies. In addition, the gene expression studies of Tox3 and Denndla in zebrafish demonstrated that N2 could impair PCOS condition. Furthermore, to confirm the N2 activity, the in-silico studies were performed against PCOS susceptible genes Tox3 and Dennd1a using molecular docking and molecular dynamic simulations. The results suggested that N2 alleviated the oxidative stress and apoptosis in-vitro and in-vivo and altered the expression of PCOS key genes.


Subject(s)
Polycystic Ovary Syndrome , Female , Humans , Animals , Cricetinae , Polycystic Ovary Syndrome/pathology , Cricetulus , Zebrafish/metabolism , Reactive Oxygen Species/metabolism , Antioxidants/metabolism , CHO Cells , Molecular Docking Simulation , Signal Transduction , Testosterone , Oxidative Stress , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Death Domain Receptor Signaling Adaptor Proteins/genetics , Death Domain Receptor Signaling Adaptor Proteins/metabolism
5.
J Tradit Chin Med ; 42(5): 741-748, 2022 10.
Article in English | MEDLINE | ID: mdl-36083481

ABSTRACT

OBJECTIVE: To assess the protective effect of dark chocolate (DC) on the letrozole-induced rat model of polycystic ovary syndrome (PCOS). METHODS: In this experimental study, 32 female Wistar rats, weighing (200 ± 20) g, were randomly categorized into 4 groups including control, letrozole (1 mg·kg·d), metformin (500 mg·kg·d) along with letrozole, and DC (500 mg·kg·d) along with letrozole for 28 d by oral gavage. Twenty-four hours after the last supplementation, direct blood sampling was taken from the heart to obtain blood serum for evaluation of sex hormones and gonadotropins, oxidative parameters, inflammatory cytokines, and ovarian tissue was examined for histology. RESULTS: The DC treatment significantly improved PCOS signs, as demonstrated by the significant restoration of ovarian morphology and physiological functions as compared with the letrozole group. DC treatment also decreased ovarian interleukin-1ß and tumor necrosis factor-α levels and improved total oxidative/antioxidative status as compared with the letrozole group. CONCLUSIONS: Treating the animals with DC could alleviate the PCOS symptoms and reduced the toxic effects of letrozole in the ovary. This effect may mediate through antioxidant and antiinflammatory properties.


Subject(s)
Chocolate , Letrozole , Polycystic Ovary Syndrome , Animals , Antioxidants , Disease Models, Animal , Female , Letrozole/toxicity , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/drug therapy , Polycystic Ovary Syndrome/pathology , Rats , Rats, Wistar
6.
Biomed Res Int ; 2022: 7402598, 2022.
Article in English | MEDLINE | ID: mdl-35845946

ABSTRACT

Background: Polycystic ovary syndrome (PCOS) is the most common hormonal disorder in women of reproductive age, and the major cause of infertility. Today, using medicinal plants instead of chemical drugs could be an alternative treatment option for PCOS. The purpose of this study was to determine the effect of Calendula officinalis hydroalcoholic extract on PCOS in rats. Method: 60 female adult rats were randomly divided into six groups, including control, sham, PCOS group, and treated PCOS groups receiving hydroalcoholic extract of Calendula officinalis with different dosages of 200, 500, and 1000 mg/kg. PCOS was induced by subcutaneous injection of DHEA 6 mg/100 g bw for 35 days. For two weeks, the extract was taken orally. The serum glucose, insulin, sex hormone levels, and oxidative status were measured at the end of the experiment. The ovaries were dissected for histomorphometric and pathological analysis. Results: When compared to the control and sham groups, the PCOS group showed a significant increase in glucose, insulin, testosterone, and malondialdehyde (MDA) concentrations, cystic and atretic follicles, and thickness of the theca and tunica albuginea layers, and a significant decrease in LH concentration, total antioxidant capacity, corpus luteum, antral follicles, and oocyte diameter. The mean concentration of FSH, on the other hand, did not change significantly. A trend of improvement was found in the treated groups with high doses of Calendula officinalis extract. Conclusion: In rats with PCOS and nonovulation, Calendula officinalis hydroalcoholic extract improved oxidative stress, restored folliculogenesis, and increased ovulation.


Subject(s)
Calendula , Insulins , Polycystic Ovary Syndrome , Androgens , Animals , Disease Models, Animal , Female , Glucose/adverse effects , Humans , Insulins/therapeutic use , Plant Extracts/therapeutic use , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/drug therapy , Polycystic Ovary Syndrome/pathology , Rats
7.
J Ovarian Res ; 15(1): 11, 2022 Jan 20.
Article in English | MEDLINE | ID: mdl-35057828

ABSTRACT

BACKGROUND: Melatonin, as a free radical scavenger exhibiting genomic actions, regulates the antioxidant genes expression and apoptosis mechanisms. In polycystic ovary syndrome (PCOS) patients, an imbalance between free radicals and antioxidants in follicular fluid leads to oxidative stress, aberrant folliculogenesis, and intrinsic defects in PCOS oocytes. In this experimental mouse model study, oocytes of PCOS and the control groups were cultured in different melatonin concentrations (10- 5, 10- 6, and 10- 7 M) to investigate the expression of oocyte maturation-related genes (Gdf9/Bmp15), antioxidant-related genes (Gpx1/Sod1), apoptotic biomarkers (Bcl2/Bax) and total intracellular ROS levels. RESULTS: Gdf9 and Bmp15, Gpx1 and Sod1 were up-regulated in PCOS and control oocytes cultured in all melatonin concentrations compared to those cultured in IVM basal medium (P < 0.05). A significant decrease in the total ROS level was observed in all groups cultured in the supplemented cultures. Melatonin increased Bcl2 and decreased Bax gene expression in PCOS and control oocytes compared to non-treated oocytes. CONCLUSIONS: Melatonin increased antioxidant gene expression and regulated the apoptosis pathway, effectively reducing the adverse effects of culture conditions on PCOS oocytes. Furthermore, it influenced the expression of oocyte maturation-related genes in PCOS, providing valuable support during the IVM process.


Subject(s)
Antioxidants/metabolism , Melatonin/pharmacology , Oocytes/drug effects , Oogenesis/drug effects , Animals , Apoptosis/drug effects , Apoptosis/genetics , Bone Morphogenetic Protein 15/genetics , Dehydroepiandrosterone/toxicity , Disease Models, Animal , Female , Glutathione Peroxidase/genetics , Growth Differentiation Factor 9/genetics , In Vitro Oocyte Maturation Techniques , Mice , Oocytes/metabolism , Oogenesis/genetics , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/pathology , Proto-Oncogene Proteins c-bcl-2/genetics , Reactive Oxygen Species/metabolism , Superoxide Dismutase-1/genetics , bcl-2-Associated X Protein/genetics , Glutathione Peroxidase GPX1
8.
J Ethnopharmacol ; 292: 114923, 2022 Jun 28.
Article in English | MEDLINE | ID: mdl-34923086

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Polycystic ovary syndrome (PCOS) is a common and complex endocrine disorder that is also an important cause of infertility. Adverse psychological stress can aggravate the occurrence and development of PCOS. Bushen Jieyu Tiaochong Formula (BJTF), a prescription of Traditional Chinese Medicine (TCM), has been used in the treatment of PCOS and shown to be effective in reducing negative emotion. However, the therapeutic mechanism has yet to be clearly elucidated. In the current study, we investigated the potential mechanism of action of BJTF. AIM OF THE STUDY: To investigate the role of PERK-ATF4-CHOP signaling in the molecular mechanisms that mediate the effects of BJTF in a rat model of PCOS, with chronic stress induced by letrozole and a chronic unpredictable mild stress (CUMS) paradigm. MATERIALS AND METHODS: In addition to the normal control group, the PCOS combined with CUMS model rats were randomly assigned to a model group, a Diane-35 (ethinylestradiol 35 µg/cyproterone acetate 2 mg)-treated positive control group, or one of three BJTF-treated groups receiving a low, medium, or high dose. Behavioral testing, including the sucrose preference test and open field test, was conducted, and hematoxylin and eosin (H&E) staining was used to observe changes in the pathological morphology of ovarian tissue. Free testosterone (FT), luteinizing hormone (LH), and follicle-stimulating hormone (FSH) levels in serum were quantified by enzyme-linked immunosorbent assays (ELISA). The hippocampal levels of norepinephrine (NE), 5-hydroxytryptamine/serotonin (5-HT), and 5-hydroxyindoleacetic acid (5-HIAA) were measured using high-performance liquid chromatography-electrochemical detection (HPLC-ECD). Apoptotic granulosa cells were detected using terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining. Furthermore, quantitative reverse transcription polymerase chain reaction (qRT-PCR) and immunohistochemistry were used to detect the expression of glucose-regulated protein 78 (GRP78) and CHOP in the ovarian tissues. The expression levels of GRP78, CHOP, PERK, and ATF4 in ovarian tissues were also measured by western blotting. RESULTS: Treatment with either BJTF or Diane-35 ameliorated the abnormal cystic dilatation of follicles in the model rats and reduced the serum levels of FT and LH, and the LH/FSH ratio. BJTF treatment also attenuated chronic psychological stress-like behavior and regulated the expression and metabolism of cerebral monoamine neurotransmitters. The efficacy of BJTF was greater than that of Diane-35, with the optimal effects observed at the medium dose. BJTF also lowered the apoptotic index of ovarian granulosa cells and downregulated the expression of GRP78, CHOP, and ATF4. Although the expression level of PERK was not significantly altered by BJTF, the mean PERK expression level was the lowest in the medium-dose BJTF group. CONCLUSIONS: Administration of BJTF has the therapeutic potential to promote the homeostasis of the reproductive endocrine environment and to restore follicular development and ovulation, possibly through the inhibition of the PERK-ATF4-CHOP signaling pathway, leading to downregulation of GRP78 expression to further delay ovarian granule cell apoptosis mediated by endoplasmic reticulum stress (ERS). Moreover, BJTF could improve behavioral performance by regulating cerebral monoamine neurotransmitters in this rat model. These findings provide a new perspective for treating PCOS related to psychological stress using TCM.


Subject(s)
Polycystic Ovary Syndrome , Activating Transcription Factor 4/metabolism , Animals , Apoptosis , Female , Follicle Stimulating Hormone , Granulosa Cells/metabolism , Humans , Luteinizing Hormone , Polycystic Ovary Syndrome/pathology , Rats , Signal Transduction , Testosterone
9.
J Food Biochem ; 46(3): e13635, 2022 03.
Article in English | MEDLINE | ID: mdl-33555077

ABSTRACT

Polycystic ovary syndrome (PCOS) is the most common cause of infertility associated with metabolic complications. Several classes of pharmacological agents have been used to manage PCOS. These drugs have shown adverse effects. Various studies showed the bee pollen (BP) as a substance rich in phytoestrogens. This study aimed to investigate the effects of BP and metformin alone and in combination with proliferation and apoptosis of granulosa cells in the rat model of PCOS. In this experimental study, 54 Wistar rats (180-210 g), was injected 2 mg of estradiol valerate intramuscularly and six rats were considered as control. After 60 days, the rats were divided into control, sham, and experimental groups. The rats were treated with bee pollen (50, 100, and 200 mg/kg) and metformin (300 mg/kg), either individually or in combination. Ovarian histology assessment was examined by H&E staining. The serum levels of NO and TNF-α were evaluated. The expressions of P53 and Ki67 were measured by IHC. In the BP and metformin-treated PCOS group, the preantral and antral follicles increased, and cystic follicles significantly decreased (p < .01). The levels of TNF-α, NO, as well as the expressions of Ki67 were decreased in the treated groups compared to the PCOS group (p < .01). On the contrary, apoptosis increased in the groups treated with BP compared to the untreated group (p < .01). BP individually or synergistically with metformin improved the symptoms of PCOS.


Subject(s)
Metformin , Polycystic Ovary Syndrome , Animals , Apoptosis , Bees , Cell Proliferation , Female , Granulosa Cells/metabolism , Granulosa Cells/pathology , Humans , Ki-67 Antigen/metabolism , Metformin/pharmacology , Pollen/metabolism , Polycystic Ovary Syndrome/drug therapy , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/pathology , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/metabolism
10.
J Ovarian Res ; 14(1): 173, 2021 Dec 11.
Article in English | MEDLINE | ID: mdl-34895279

ABSTRACT

INTRODUCTION: The hypothalamic-pituitary-ovarian (HPO) axis is the principal regulator of the reproductive system. The neurons in the arcuate nucleus of the hypothalamus signal the basophilic cells of the anterior pituitary to release luteinizing hormone (LH) and follicle stimulating hormone (FSH), which bind to the granulosa and theca cells of a follicle in the ovary to promote healthy follicular development. Disruption of this process at any time can lead to polycystic ovaries and, if left untreated, can lead to Polycystic Ovarian Syndrome (PCOS), one of the leading causes of infertility. A novel treatment option using 150 kHz Intermediate Frequency (IF) Electromagnetic Radiation (EMR) has been proposed to monitor the effect of this frequency during cystic development. METHODS: To prove this, an experiment was conducted to study the effect of whole-body exposure to 150 kHz EMR for 8 weeks at receptor, cellular, tissue and hormonal levels on the HPO axis of 25 young cyclic female rats. RESULTS: The results showed that 150 kHz EMR did not affect the histoarchitecture of neurons of arcuate nucleus of the hypothalamus of PCO-induced rats. It was also found that the number of basophilic cells of the pituitary gland was increased and the immunoreactivity of LH and FSH secretion increased. This EMR field also decreased the development of follicular cysts in the ovary and possibly increased the immunoreactivity of the LH and FSH receptors as well on the theca and granulosa cells of follicles in the ovary. CONCLUSION: There are still many limitations to this study. If properly evaluated, the results of this experiment could help develop a new non-invasive treatment option for women with PCOS in the near future.


Subject(s)
Magnetic Field Therapy , Polycystic Ovary Syndrome/therapy , Animals , Disease Models, Animal , Electromagnetic Radiation , Estradiol , Female , Follicle Stimulating Hormone/blood , Follicle Stimulating Hormone/metabolism , Hypothalamus/metabolism , Hypothalamus/pathology , Luteinizing Hormone/blood , Luteinizing Hormone/metabolism , Neurons/cytology , Ovary/metabolism , Ovary/pathology , Pituitary Gland/metabolism , Pituitary Gland/pathology , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/pathology , Rats, Sprague-Dawley , Receptors, FSH/metabolism , Receptors, LH/metabolism
11.
BMC Complement Med Ther ; 21(1): 291, 2021 Nov 29.
Article in English | MEDLINE | ID: mdl-34844580

ABSTRACT

BACKGROUND: Insulin resistance and hormonal imbalances are key features in the pathophysiology of polycystic ovarian syndrome (PCOS). We have previously shown that Ficus deltoidea var. deltoidea Jack (Moraceae) can improve insulin sensitivity and hormonal profile in PCOS female rats. However, biological characteristics underpinning the therapeutic effects of F. deltoidea for treating PCOS remain to be clarified. This study aims to investigate the biochemical, hormonal, and histomorphometric changes in letrozole (LTZ)-induced PCOS female rats following treatment with F. deltoidea. METHODS: PCOS was induced in rats except for normal control by administering LTZ at 1 mg/kg/day for 21 days. Methanolic extract of F. deltoidea leaf was then orally administered to the PCOS rats at the dose of 250, 500, or 1000 mg/kg/day, respectively for 15 consecutive days. Lipid profile was measured enzymatically in serum. The circulating concentrations of reproductive hormone and antioxidant enzymes were determined by ELISA assays. Ovarian and uterus histomorphometric changes were further observed by hematoxylin and eosin (H&E) staining. RESULTS: The results showed that treatment with F. deltoidea at the dose of 500 and 1000 mg/kg/day reduced insulin resistance, obesity indices, total cholesterol, triglycerides, low-density lipoprotein cholesterol (LDL), malondialdehyde (MDA), testosterone, luteinizing hormone (LH), and follicle-stimulating hormone (FSH) to near-normal levels in PCOS rats. The levels of high-density lipoprotein cholesterol (HDL), estrogen, and superoxide dismutase (SOD) are also similar to those observed in normal control rats. Histomorphometric measurements confirmed that F. deltoidea increased the corpus luteum number and the endometrial thickness. CONCLUSIONS: F. deltoidea can reverse PCOS symptoms in female rats by improving insulin sensitivity, antioxidant activities, hormonal imbalance, and histological changes. These findings suggest the potential use of F. deltoidea as an adjuvant agent in the treatment program of PCOS.


Subject(s)
Antioxidants/therapeutic use , Ficus , Hormones/metabolism , Insulin Resistance , Lipids/blood , Phytotherapy , Polycystic Ovary Syndrome/drug therapy , Animals , Antioxidants/pharmacology , Blood Glucose/metabolism , Corpus Luteum/drug effects , Disease Models, Animal , Endometrium/drug effects , Female , Follicle Stimulating Hormone/blood , Insulin/metabolism , Letrozole , Luteinizing Hormone/blood , Malondialdehyde/blood , Obesity , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Plant Leaves , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/pathology , Rats, Sprague-Dawley , Testosterone/blood
12.
Int J Mol Sci ; 22(20)2021 Oct 14.
Article in English | MEDLINE | ID: mdl-34681721

ABSTRACT

Ghrelin and nesfatin-1 are enteroendocrine peptide hormones expressed in rat X/A-like and human P/D1cells of the gastric mucosa. Besides their effect on food intake, both peptides are also implicated in various other physiological systems. One of these is the reproductive system. This present review illustrates the distribution of ghrelin and nesfatin-1 along the hypothalamus-pituitary-gonadal (HPG) axis, their modulation by reproductive hormones, and effects on reproductive functions as well as highlighting gaps in current knowledge to foster further research.


Subject(s)
Ghrelin/metabolism , Nucleobindins/metabolism , Reproduction/genetics , Female , Ghrelin/blood , Ghrelin/genetics , Humans , Hypothalamus/metabolism , Nucleobindins/blood , Nucleobindins/genetics , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/pathology , Pre-Eclampsia/metabolism , Pre-Eclampsia/pathology , Pregnancy
13.
Biomed Pharmacother ; 144: 112288, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34653763

ABSTRACT

The main features of polycystic ovary syndrome (PCOS) are abnormal follicular development and ovulation dysfunction, which are caused by the excessive autophagy of ovarian granulosa cells. Acupuncture has been shown to improve ovulation dysfunction and abnormal follicular development in PCOS patients, but its mechanism is unclear. This study hypothesized that the beneficial effects of acupuncture are the result of LncMEG3-mediated effects on the PI3K/AKT/mTOR pathway. Acupuncture (CV-4, RN-3, CV-6, SP-6 and EX-CA 1) was used to treat a rat model of polycystic ovary syndrome. Hematoxylin-eosin staining was used to observe ovarian morphology and enzyme-linked immunosorbent assay, western blotting, immunohistochemistry and real-time PCR were used to detect LH, E2, FSH, T, AMH, LncMEG3, PI3K, AKT, mTOR, P62 and LC3II/I expression. The ovarian morphology of 90% of the rats in the acupuncture treatment group was significantly improved after 11 consecutive days of therapy. Acupuncture also resulted in a significant decrease in serum LH, FSH, T and AMH levels and a significant increase in E2 level (P<0.01). LncMEG3, PI3K, AKT, mTOR, P62 and LC3II/I expression was decreased in ovarian granulosa cells after acupuncture compared with PCOS and lentiviral Intervention Group (P<0.05), while the expression of follicle stimulating hormone receptor was increased (P<0.05). These results indicate that acupuncture can down-regulate the expression of LncMEG3 and thereby inhibit the PI3K/AKT/mTOR pathway, reducing granulosa cell autophagy and normalizing their proliferation. These factors ultimately remedy abnormal follicular development. These findings suggest that acupuncture has clinical potential as a safe treatment for PCOS ovulatory dysfunction.


Subject(s)
Acupuncture Therapy , Autophagy , Granulosa Cells/enzymology , Ovulation , Phosphatidylinositol 3-Kinase/metabolism , Polycystic Ovary Syndrome/therapy , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Autophagy-Related Proteins/metabolism , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Female , Gonadal Steroid Hormones/metabolism , Gonadotropins, Pituitary/metabolism , Granulosa Cells/pathology , Polycystic Ovary Syndrome/enzymology , Polycystic Ovary Syndrome/pathology , Polycystic Ovary Syndrome/physiopathology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Rats, Sprague-Dawley , Signal Transduction
14.
Int J Mol Sci ; 22(17)2021 Aug 30.
Article in English | MEDLINE | ID: mdl-34502321

ABSTRACT

We examined the vasoactive effect of estradiol in a rat model of early PCOS and the influence of vitamin D deficiency (VDD). We created a model of chronic hyperandrogenism and VDD in adolescent female Wistar rats (N = 46) with four experimental groups: vitamin D supplemented (T-D+), VDD (T-D-), hyperandrogenic and vitamin D supplemented (T+D+), and hyperandrogenic and VDD (T+D-). T+ groups received an 8-week-long transdermal Androgel treatment, D-animals were on vitamin D-reduced diet and D+ rats were supplemented orally with vitamin D3. Estrogen-induced vasorelaxation of thoracic aorta segments were measured with a wire myograph system with or without the inhibition of endothelial nitric oxide synthase (eNOS) or cyclooxygenase-2 (COX-2). The distribution of estrogen receptor (ER), eNOS and COX-2 in the aortic wall was assessed by immunohistochemistry. VDD aortas showed significantly lower estradiol-induced relaxation independently of androgenic status that was further decreased by COX-2 inhibition. COX-2 inhibition failed to alter vessel function in D+ rats. Inhibition of eNOS abolished the estradiol-induced relaxation in all groups. Changes in vascular function in VDD were accompanied by significantly decreased ER and eNOS staining. Short-term chronic hyperandrogenism failed to, but VDD induced vascular dysfunction, compromised estrogen-dependent vasodilatation and changes in ER and eNOS immunostaining.


Subject(s)
Cholecalciferol/pharmacology , Estradiol/pharmacology , Polycystic Ovary Syndrome/drug therapy , Vasodilation , Vitamin D Deficiency/complications , Animals , Aorta/drug effects , Disease Models, Animal , Estrogens/pharmacology , Female , Nitric Oxide Synthase Type III/metabolism , Polycystic Ovary Syndrome/etiology , Polycystic Ovary Syndrome/pathology , Rats , Rats, Wistar , Vitamins/pharmacology
15.
Reprod Biol Endocrinol ; 19(1): 147, 2021 Sep 23.
Article in English | MEDLINE | ID: mdl-34551795

ABSTRACT

BACKGROUND: Polycystic ovary syndrome (PCOS), as the most common endocrine disorder in reproductive-aged women, is characterized by oxidative stress and ovarian tissue inflammation. Green tea extract (GTE) potentially possesses therapeutic effects for PCOS because of the antioxidant and anti-inflammatory compounds. This systematic review evaluates the potential roles of GTE on metabolic variables, hormone levels, and ovarian function in PCOS. METHODS: A systematic review was conducted of published studies reporting the effects of GTE on PCOS. Several major databases, including PubMed, SCOPUS, and Google Scholar, were searched up from inception to April 2021. Clinical trials and animal studies that assessed the effects of GTE on PCOS were eligible for inclusion. RESULTS: Of 314 articles found in the search, four human studies and four animal studies were included. All studies in humans showed the effects of GTE on weight loss. GTE's effect on decreasing testosterone levels in humans and LH levels in animals were also reported. In addition, increases in FSH and progesterone levels in animal models were observed. Although GTE improved fasting blood sugar and insulin levels, the effect of GTE on inflammatory parameters, such as TNF-alpha and IL-6 and antioxidant status, was limited to animal studies. CONCLUSION: Therefore, this review suggests that GTE could be considered a potential agent to attenuate PCOS complications mainly due to its effect on weight loss and glycemic levels. However, more studies are needed to formulate conclusions about the effects and mechanisms of GTE in PCOS.


Subject(s)
Plant Extracts/pharmacology , Polycystic Ovary Syndrome/metabolism , Tea/chemistry , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Blood Glucose/drug effects , Blood Glucose/metabolism , Female , Humans , Insulin/blood , Insulin Resistance , Oxidative Stress/drug effects , Phytotherapy/methods , Plant Extracts/therapeutic use , Polycystic Ovary Syndrome/drug therapy , Polycystic Ovary Syndrome/pathology
16.
Biochim Biophys Acta Mol Basis Dis ; 1867(12): 166242, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34389474

ABSTRACT

Endoplasmic reticulum (ER) stress, with adaptive unfolded protein response (UPR), is a key link between obesity, insulin resistance and type 2 diabetes, all of which are often present in the most common endocrine-metabolic disorder in women of reproductive age, polycystic ovary syndrome (PCOS), which is characterized with hyperandrogenism. However, the link between excess androgen and endoplasmic reticulum (ER) stress/insulin resistance in patients with polycystic ovary syndrome (PCOS) is unknown. An unexpected role of kisspeptin was reported in the regulation of UPR pathways and its involvement in the androgen-induced ER stress in hypothalamic neuronal cells. To evaluate the relationship of kisspeptin and ER stress, we detected kisspeptin and other factors in blood plasm of PCOS patients, rat models and hypothalamic neuronal cells. We detected higher testosterone and lower kisspeptin levels in the plasma of PCOS than that in non-PCOS women. We established a PCOS rat model by dihydrotestosterone (DHT) chronic exposure, and observed significantly downregulated kisspeptin expression and activated UPR pathways in PCOS rat hypothalamus compared to that in controls. Inhibition or knockdown of kisspeptin completely mimicked the enhancing effect of DHT on UPR pathways in a hypothalamic neuronal cell line, GT1-7. Kp10, the most potent peptide of kisspeptin, effectively reversed or suppressed the activated UPR pathways induced by DHT or thapsigargin, an ER stress activator, in GT1-7 cells, as well as in the hypothalamus in PCOS rats. Similarly, kisspeptin attenuated thapsigargin-induced Ca2+ response and the DHT- induced insulin resistance in GT1-7 cells. Collectively, the present study has revealed an unexpected protective role of kisspeptin against ER stress and insulin resistance in the hypothalamus and has provided a new treatment strategy targeting hypothalamic ER stress and insulin resistance with kisspeptin as a potential therapeutic agent.


Subject(s)
Endoplasmic Reticulum Stress/genetics , Kisspeptins/blood , Neurons/metabolism , Polycystic Ovary Syndrome/genetics , Androgens/adverse effects , Animals , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Disease Models, Animal , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Female , Hypothalamus/metabolism , Hypothalamus/pathology , Insulin Resistance/genetics , Kisspeptins/genetics , Neurons/pathology , Obesity/metabolism , Obesity/pathology , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/pathology , Rats , Testosterone/blood , Unfolded Protein Response/genetics
17.
J Neuroendocrinol ; 33(8): e12999, 2021 08.
Article in English | MEDLINE | ID: mdl-34216402

ABSTRACT

Although polycystic ovary syndrome (PCOS) is the most common cause of anovulatory infertility worldwide, the aetiology of the disorder remains poorly defined. Animal-based evidence highlights the brain as a prime suspect in both the development and maintenance of PCOS. Prenatally androgenised (PNA) models of PCOS exhibit excessive GABAergic wiring associated with PCOS-like reproductive deficits in adulthood, with aberrant brain wiring detected as early as postnatal day (P) 25, prior to disease onset, in the PNA mouse. The mechanisms underlying this aberrant brain wiring remain unknown. Microglia, the immune cells of the brain, are regulators of neuronal wiring across development, mediating both the formation and removal of neuronal inputs. Here, we tested the hypothesis that microglia play a role in the excessive GABAergic wiring that leads to PCOS-like features in the PNA brain. Using specific immunolabelling, microglia number and morphology associated with activation states were analysed in PNA and vehicle-treated controls across developmental timepoints, including embryonic day 17.5, P0, P25 and P60 (n = 7-14 per group), and in two regions of the hypothalamus implicated in fertility regulation. At P0, fewer amoeboid microglia were observed in the rostral preoptic area (rPOA) of PNA mice. However, the greatest changes were observed at P25, with PNA mice exhibiting fewer total microglia, and specifically fewer "sculpting" microglia, in the rPOA. Based on these findings, we assessed microglia-mediated refinement of GABAergic synaptic terminals at two developmental stages of peak synaptic refinement: P7 and P15 (n = 7 per group). PNA mice showed a reduction in the uptake of GABAergic synaptic material at P15. These findings reveal time-specific changes in the microglia population and refinement of GABAergic inputs in a mouse model of PCOS driven by prenatal androgen excess and suggest a role for microglia in shaping the atypical brain wiring associated with the development of PCOS features.


Subject(s)
Brain/pathology , Microglia/physiology , Polycystic Ovary Syndrome/psychology , Animals , Disease Models, Animal , Female , GABAergic Neurons/physiology , Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/metabolism , Hypothalamus/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/metabolism , Polycystic Ovary Syndrome/pathology , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/pathology , Prenatal Exposure Delayed Effects/psychology
18.
J Reprod Immunol ; 146: 103345, 2021 08.
Article in English | MEDLINE | ID: mdl-34116484

ABSTRACT

Polycystic Ovary Syndrome (PCOS), a major endocrine disorder, affects the reproductive function of a woman, along with an association with metabolic conditions like insulin resistance and inflammation. The inflammatory nature of PCOS is much debated over, owing to numerous cases of elevation in cytokine levels. Studies have shown the beneficiary effect of Gamma-Linolenic acid (GLA) in reducing inflammation related to many conditions such as atopic dermatitis, rheumatoid arthritis, arterial disease, obesity, and even PCOS. The study aims at assessing the expression of inflammatory cytokines in the ovary and Peri-ovarian adipose tissue (POAT) of the Dehydroepiandrosterone (DHEA) induced PCOS rat model. Further, this study also evaluates the effect of γ-linolenic Acid (GLA) on these cytokines in POAT. Female Wistar rats were subcutaneously injected with 60 mg/kg DHEA daily for 28 days. These PCOS-induced rats were then orally administered with 50 mg/kg GLA for 14 days. The gene expression of cytokines was assessed by Real Time-PCR. The study showed an increase in the expression of cytokines in the ovary and POAT of the DHEA group. This suggests the role of ovarian adipose in adding to the pro-inflammatory state of PCOS. Moreover, the administration of GLA to the PCOS-induced rats resulted in a reduction of cytokine expression from the POAT, indicating that the compound was successful in reducing the associated inflammation. The study throws light on the possibility of using GLA as a supplementary or naturalistic alternative in ameliorating ovarian adipose-associated inflammation that accompanies PCOS.


Subject(s)
Adipose Tissue/drug effects , Cytokines/metabolism , Polycystic Ovary Syndrome/immunology , gamma-Linolenic Acid/pharmacology , Adipose Tissue/immunology , Adipose Tissue/pathology , Animals , Disease Models, Animal , Female , Humans , Inflammation/drug therapy , Inflammation/immunology , Inflammation/pathology , Ovary/drug effects , Ovary/immunology , Ovary/pathology , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/drug therapy , Polycystic Ovary Syndrome/pathology , Rats , gamma-Linolenic Acid/therapeutic use
19.
Reprod Sci ; 28(10): 2807-2815, 2021 10.
Article in English | MEDLINE | ID: mdl-33876387

ABSTRACT

Reactive oxygen species (ROS), involved in the pathogenesis of the polycystic ovary syndrome (PCOS), play a key role in the onset of apoptosis in follicles and granulosa cells (GCs). We aimed to investigate the antioxidant effects of AST and metformin separately and in combination on GCs using a PCOS mouse model. Forty-eight prepubertal female BALB C mice aged 25-30 days and weighing 12-14 g were studied. The PCOS model was created by subcutaneous injection of the dehydroepiandrosterone (DHEA) hormone in 8 mice of BALB C for 20 consecutive days. Apoptosis and the amount of ROS were evaluated in GCs of the ovaries via flow cytometry. The activity of AKT protein was measured by western blot, and the viability of GCs was investigated using spectrophotometry. Ovarian tissue sections were prepared, stained with H&E, and the morphology of the sections was examined. Statistical analysis was performed by SPSS v22.0 software using one-way ANOVA. We found that AST administration leads to a significant reduction in oxidative stress (P<0.01) and consequently a significant decrease in the rate of apoptosis (P<0.01). While the expression of AKT in the AST group revealed a significant increase (P<0.05), it decreased in the metformin group. However, it was still significantly higher than the control and PCOS groups. Ovulation was confirmed in both metformin and AST groups. Further studies are warranted to prove the efficacy of AST and to introduce it as a complementary therapeutic agent in PCOS.


Subject(s)
Granulosa Cells/drug effects , Metformin/therapeutic use , Oxidative Stress/drug effects , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/drug therapy , Animals , Dehydroepiandrosterone/toxicity , Female , Granulosa Cells/metabolism , Granulosa Cells/pathology , Metformin/pharmacology , Mice , Mice, Inbred BALB C , Oxidative Stress/physiology , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/pathology , Xanthophylls/pharmacology , Xanthophylls/therapeutic use
20.
Life Sci ; 276: 119456, 2021 Jul 01.
Article in English | MEDLINE | ID: mdl-33811895

ABSTRACT

BACKGROUND: In recent years, female infertility from Polycystic Ovary Syndrome (PCOS) has gained scientific interest. PCOS alters the metabolic and endocrine functioning in females. The elevation in androgens can damage the androgen receptors present on the kidney giving rise to renal disorders like Focal Segmental Glomerulosclerosis (FSGS). Transforming Growth Factor Beta (TGF-ß) in the ovary is activated by activin for Follicle Stimulating Hormone (FSH) secretion and in the kidney by thrombospondin 1 (TSP1) for cell growth and apoptosis. Studies show that gamma-linolenic acid (GLA) effectively treats breast cancer, eczema, inflammatory conditions and PCOS. AIM: The study aimed to find out the possibility of FSGS development in PCOS and to understand the effect of GLA on FSGS via the TGF-ß pathway. METHOD: To carry out the study, the dehydroepiandrosterone (DHEA) induced PCOS model was used. Three groups namely vehicle control, DHEA, and DHEA+GLA, were used with six animals in each. TGF-ß1, TGF-ß2, and TSP1 genes were studied using real-time PCR. RESULTS: The study showed an increase in the level of renal fibrosis biomarker, TSP1, in the DHEA group, which was further decreased by an anti-inflammatory agent, GLA. The TGF-ß1 and TGF-ß2 genes associated with the TGF-ß pathway were seen to be increased in DHEA-induced PCOS rats which showed a possible relation between the two conditions. CONCLUSION: The study shows a possible development of renal fibrosis in the DHEA-induced PCOS model. The GLA might act as a ligand to regulate TGF-ß signaling in glomerulosclerosis in a DHEA-induced PCOS model.


Subject(s)
Glomerulosclerosis, Focal Segmental/drug therapy , Polycystic Ovary Syndrome/complications , Transforming Growth Factor beta/metabolism , gamma-Linolenic Acid/pharmacology , Adjuvants, Immunologic/toxicity , Animals , Dehydroepiandrosterone/toxicity , Female , Glomerulosclerosis, Focal Segmental/etiology , Glomerulosclerosis, Focal Segmental/metabolism , Glomerulosclerosis, Focal Segmental/pathology , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/pathology , Rats , Rats, Wistar , Signal Transduction , Transforming Growth Factor beta/genetics
SELECTION OF CITATIONS
SEARCH DETAIL