Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 202
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
Phytomedicine ; 128: 155468, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38471315

ABSTRACT

BACKGROUND: Oxidative stress is considered the main cause of granulosa cell apoptosis in ovarian disease. Curcumin has various biological roles, but its potential role in protecting granulosa cells from oxidative damage remains unidentified. PURPOSE: The study revealed the protective effect of curcumin on granulosa cell survival under oxidative stress, and explored its mode of action. STUDY DESIGN: The protective effect of curcumin on oxidative stress-induced ovarian cell apoptosis was evaluated in vivo and in vitro, and the role of autophagy and AMPK/mTOR signaling pathway in this process was also demonstrated. METHODS: First, mice were injected to 3-nitropropionic acid (3-NPA, 20 mg/kg/day) for 14 consecutive days to establish the ovarian oxidative stress model, at same time, curcumin (50, 100, 200 mg/kg/day) was given orally. Thereafter, functional changes, cell apoptosis, and autophagy in ovarian tissue were evaluated by hematoxylin-eosin staining, enzyme-linked immunosorbent assay, western blotting, TUNEL assays, and transmission electron microscopy. Finally, oxidative stress model of granulosa cells was established with H2O2in vitro and treated with curcumin. The underlying mechanisms of curcumin to protect the apoptosis under oxidative stress in vitro were determined using western blotting and TUNEL assays. RESULTS: In our study, after curcumin treatment, the mouse ovarian function disorder under 3-nitropropionic acid-induced oxidative stress recovered significantly, and ovarian cell apoptosis decreased. H2O2 induced granulosa cell apoptosis in vitro, and curcumin antagonized this process. Autophagy contributes to tissue and cell survival under stress. We therefore examined the role of autophagy in this process. According to the in vivo and in vitro results, curcumin restored autophagy under oxidative stress. The autophagy inhibitor (chloroquine) exhibited the same effect as curcumin, whereas the autophagy activator (rapamycin) antagonized the effect of curcumin. In addition, the study found that the AMPK/mTOR pathway plays a crucial role in curcumin- mediated autophagy to protect against oxidative stress-induced apoptosis. CONCLUSION: Our findings for the first time systematically revealed a new mechanism through which curcumin protects ovarian granulosa cells from oxidative stress-induced damage through AMPK/mTOR-mediated autophagy and suggested that it can be a new therapeutic direction for female ovarian diseases.


Subject(s)
Autophagy , Curcumin , Ovary , Oxidative Stress , TOR Serine-Threonine Kinases , Animals , Female , Mice , AMP-Activated Protein Kinases/drug effects , AMP-Activated Protein Kinases/metabolism , Apoptosis/drug effects , Autophagy/drug effects , Curcumin/pharmacology , Granulosa Cells/drug effects , Hydrogen Peroxide/toxicity , Nitro Compounds , Ovary/drug effects , Oxidative Stress/drug effects , Propionates/pharmacology , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/drug effects , TOR Serine-Threonine Kinases/metabolism
2.
Phytomedicine ; 128: 155456, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38537446

ABSTRACT

BACKGROUND: Polycystic ovary syndrome (PCOS) is a heterogeneous metabolic and endocrine disorder that causes anovulatory infertility and abnormal folliculogenesis in women of reproductive age. Several studies have revealed inflammation in PCOS follicles, and recent evidence suggests that Berberine (BBR) effectively reduces inflammatory responses in PCOS, however, the underlying mechanisms remain unclear. PURPOSE: To determine the underlying mechanisms by which BBR alleviates inflammation in PCOS. STUDY DESIGN: Primary human GCs from healthy women and women with PCOS, and KGN cells were used for in vitro studies. ICR mice were used for in vivo studies. METHODS: Gene expression was measured using RT-qPCR. HAS2, inflammatory cytokines, and serum hormones were assayed by ELISA. Protein expression profiles were assayed by Western blot. Chronic low-grade inflammatory mouse models were developed by intraperitoneal injection with LPS, and PCOS mouse models were established by subcutaneous intraperitoneal injection of DHEA. BBR and 4-MU were administered by gavage. Ovarian morphologic changes were evaluated using H&E staining. HAS2 expression in the ovary was assayed using Western blot and immunohistochemistry. RESULTS: Our results confirmed that HAS2 expression and hyaluronan (HA) accumulation are closely associated with inflammatory responses in PCOS. Data obtained from in vitro studies showed that HAS2 and inflammatory genes (e.g., MCP-1, IL-1ß, and IL-6) are significantly upregulated in PCOS samples and LPS-induced KGN cells compared to their control groups. In addition, these effects were reversed by blocking HAS2 expression or HA synthesis using BBR or 4-MU, respectively. Furthermore, HAS2 overexpression induces the expression of inflammatory genes in PCOS. These results were further confirmed in LPS- and DHEA-induced mouse models, where inflammatory genes were reduced by BBR or 4-MU, and ovarian morphology was restored. CONCLUSIONS: Our results define previously unknown links between HAS2 and chronic low-grade inflammation in the follicles of women with PCOS. BBR exerts its anti-inflammatory effects by down-regulating HAS2. This study provides a novel therapeutic target for alleviating ovarian inflammation in women with PCOS.


Subject(s)
Berberine , Disease Models, Animal , Hyaluronan Synthases , Inflammation , Mice, Inbred ICR , Polycystic Ovary Syndrome , Polycystic Ovary Syndrome/drug therapy , Berberine/pharmacology , Female , Animals , Humans , Hyaluronan Synthases/metabolism , Inflammation/drug therapy , Mice , Hyaluronic Acid , Adult , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Dehydroepiandrosterone/pharmacology , Ovary/drug effects , Lipopolysaccharides , Cytokines/metabolism
3.
Appl Biochem Biotechnol ; 196(8): 5265-5282, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38153651

ABSTRACT

Polycystic ovarian syndrome (PCOS) is an endocrine syndrome in women of reproductive age. Berberine (BBR) is a Chinese herbal monomer that exhibits many pharmacological properties related to PCOS treatment. This study aims to analyze the effect of BBR on a cell model of PCOS and the underlying mechanism. Human ovarian granulosa (KGN) cells were treated with dihydrotestosterone (DHT) to mimic a PCOS cell model. The RNA expression of circ_0097636, miR-186-5p, and sirtuin3 (SIRT3) was determined by quantitative real-time polymerase chain reaction (qRT-PCR). Protein expression was detected by western blotting. Cell viability was analyzed by CCK-8 assay. Cell proliferation and apoptosis were investigated by 5-ethynyl-2'-deoxyuridine (EdU) assay and flow cytometry assay, respectively. The levels of interleukin-6 (IL-6), IL-1ß, and tumor necrosis factor-α (TNF-α) were analyzed by enzyme-linked immunosorbent assays (ELISAs). Fe2+ concentration was assessed by an iron assay kit. Oxidative stress was assessed by detecting reactive oxygen species (ROS) level and malondialdehyde (MDA) level using commercial kits. The association of miR-186-5p with circ_0097636 and SIRT3 was identified by dual-luciferase reporter assay and RNA pull-down assay. Circ_0097636 expression was downregulated in the follicular fluid of PCOS patients and DHT-treated KGN cells when compared with control groups. BBR treatment partially relieved the DHT-induced inhibitory effect on cell proliferation and promoted effects on cell apoptosis, inflammation, ferroptosis, and oxidative stress in KGN cells. Additionally, circ_0097636 bound to miR-186-5p, and SIRT3 was identified as a target gene of miR-186-5p in KGN cells. BBR treatment ameliorated DHT-induced KGN cell injury by upregulating circ_0097636 and SIRT3 expression and downregulating miR-186-5p expression. Moreover, circ_0097636 overexpression protected KGN cells from DHT-induced injury by increasing SIRT3 expression. BBR ameliorated DHT-induced KGN cell injury and ferroptosis by regulating the circ_0097636/miR-186-5p/SIRT3 pathway.


Subject(s)
Berberine , Dihydrotestosterone , Ferroptosis , Granulosa Cells , MicroRNAs , Polycystic Ovary Syndrome , RNA, Circular , Sirtuin 3 , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Dihydrotestosterone/pharmacology , Female , Ferroptosis/drug effects , Granulosa Cells/metabolism , Granulosa Cells/drug effects , Sirtuin 3/metabolism , Sirtuin 3/genetics , RNA, Circular/genetics , RNA, Circular/metabolism , Berberine/pharmacology , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/drug therapy , Polycystic Ovary Syndrome/genetics , Signal Transduction/drug effects , Cell Proliferation/drug effects , Apoptosis/drug effects
4.
Molecules ; 28(13)2023 Jun 23.
Article in English | MEDLINE | ID: mdl-37446601

ABSTRACT

Melatonin has profound antioxidant activity and numerous functions in humans as well as in livestock and poultry. Additionally, melatonin plays an important role in regulating the biological rhythms of animals. Combining melatonin with scientific breeding management has considerable potential for optimizing animal physiological functions, but this idea still faces significant challenges. In this review, we summarized the beneficial effects of melatonin supplementation on physiology and reproductive processes in cattle, including granulosa cells, oocytes, circadian rhythm, stress, inflammation, testicular function, spermatogenesis, and semen cryopreservation. There is much emerging evidence that melatonin can profoundly affect cattle. In the future, we hope that melatonin can not only be applied to cattle, but can also be used to safely and effectively improve the efficiency of animal husbandry.


Subject(s)
Animal Husbandry , Breeding , Cattle , Melatonin , Animals , Cattle/genetics , Cattle/growth & development , Cattle/physiology , Animal Husbandry/methods , Breeding/methods , Dietary Supplements , Granulosa Cells/drug effects , Granulosa Cells/physiology , Melatonin/pharmacology , Melatonin/physiology , Oocytes/drug effects , Oocytes/physiology , Reproduction/drug effects , Reproduction/physiology
5.
PeerJ ; 11: e14612, 2023.
Article in English | MEDLINE | ID: mdl-36684672

ABSTRACT

Background: Communication between oocytes and granulosa cells ultimately dictate follicle development or atresia. Melatonin is also involved in follicle development. This study aimed to investigate the effects of melatonin and its receptor antagonists on hormone secretion, as well as gene expression related to hormone synthesis, TGF-ß superfamily, and follicle development in bovine granulosa cells, and assess the effects of melatonin in the presence of 4-P-PDOT and luzindole. Methods: Bovine ovaries were collected from a local abattoir and follicular fluid (follicle diameter 5-8 mm) was collected for granulosa cell isolation and culture. Granulosa cells and culture medium were collected 48 h after treatment with melatonin at high dose concentrations (10-5 M) and low dose concentrations (10-9 M) in the absence/presence of 4-P-PDOT and luzindole (10-5 M or 10-9 M). Furthermore, the expression level of genes related to hormonal synthesis (CYP11A1, CYP19A1, StAR, and RUNX2), TGF-ß superfamily (BMP6, INHA, INHBA, INHBB, and TGFBR3), and development (EGFR, DNMT1A, and FSHR) were detected in each experimental group by real-time quantitative PCR. In addition, the level of hormones in culture medium were detected using ELISA. Results: Both 10-5 M and 10-9 M melatonin doses promoted the secretion of inhibin A and progesterone without affecting the production of inhibin B and estradiol. In addition, both promoted the gene expression of INHA, StAR, RUNX2, TGFBR3, EGFR, and DNMT1A, and inhibited the expression of BMP6, INHBB, CYP11A1, CYP19A1, and FSHR. When combined with different doses of 4-P-PDOT and luzindole, they exhibited different effects on the secretion of inhibin B, estradiol, inhibin A, and progesterone, and the expression of CYP19A1, RUNX2, BMP6, INHBB, EGFR, and DNMT1A induced by melatonin. Conclusion: High and low dose melatonin receptor antagonists exhibited different effects in regulating hormone secretion and the expression of various genes in response to melatonin. Therefore, concentration effects must be considered when using luzindole or 4-P-PDOT.


Subject(s)
Granulosa Cells , Melatonin , Animals , Cattle , Female , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Core Binding Factor Alpha 1 Subunit/metabolism , ErbB Receptors/metabolism , Estradiol/metabolism , Granulosa Cells/drug effects , Melatonin/pharmacology , Progesterone/metabolism
6.
Reprod Fertil Dev ; 34(18): 1128-1134, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36309990

ABSTRACT

CONTEXT: The role of metabolic hormones, medicinal plants and their interrelationships in the control of human reproductive processes are poorly understood. AIMS: To examine how leptin, obestatin and ginkgo (Ginkgo biloba L.) affect human ovarian hormone release. METHODS: We analysed the influence of leptin and obestatin alone and in combination with ginkgo extract on cultured human ovarian granulosa cells. The release of progesterone (P), insulin-like growth factor I (IGF-I), oxytocin (OT) and prostaglandin F (PGF) were analysed by enzyme immunoassay and enzyme-linked immunosorbent assay. KEY RESULTS: Leptin addition promoted the release of all the measured hormones. Obestatin stimulated the release of P, IGF-I and OT and inhibited PGF output. Ginkgo suppressed P, IGF-I and OT and promoted PGF release. Furthermore, ginkgo changed the stimulatory action of leptin on PGF to an inhibitory one. CONCLUSIONS: Leptin and obestatin are involved in the control of human ovarian hormone release and ginkgo influences their function. IMPLICATIONS: Leptin and obestatin could be useful as stimulators of human ovarian cell functions. The suppressive influence of ginkgo on ovarian function should lead to the development of ginkgo-containing drugs.


Subject(s)
Ghrelin , Ginkgo biloba , Granulosa Cells , Leptin , Plant Preparations , Female , Humans , Cells, Cultured , Ghrelin/pharmacology , Ginkgo biloba/chemistry , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Insulin-Like Growth Factor I/metabolism , Leptin/pharmacology , Progesterone/metabolism , Prostaglandins F/metabolism , Plant Preparations/pharmacology
7.
J Ethnopharmacol ; 288: 114862, 2022 Apr 24.
Article in English | MEDLINE | ID: mdl-34861362

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Bu-shen-zhu-yun decoction (BSZYD) is a traditional chinese herbal prescription is widely used in the treatment of infertility. AIM OF THE STUDY: We aimed to elucidate the impact of a traditional herbal prescription BSZYD on polycystic ovary syndrome (PCOS). MATERIALS AND METHODS: The candidate active compounds in BSZYD and their putative targets were investigated by bioinformatics analysis. A deydroepiandrosterone (DHEA)-induced PCOS rat model was then constructed using female Sprague-Dawley (SD) rats. Serum hormone levels were measured by enzyme-linked immunosorbent assay (ELISA). Pathological changes in ovarian tissues were analyzed with hematoxylin and eosin (H&E) staining. The expressions of estrogen receptor α (ER α)-mediated PI3K/AKT/mTOR pathway were measured by immunofluorescence and western blotting. RESULTS: Bioinformatics analysis showed that the putative targets of active compound candidates in BSZYD were enriched in PI3K/AKT and estrogen signaling pathways related to regulating ovarian ovulation. Animal experiments showed that BSZYD significantly alleviated pathological changes in the ovary, altered hormone levels of serum and reduced apoptosis rate of granulosa cells. In addition, BSZYD treatment notably upregulated the expressions of proteins in ER α-mediated PI3K/AKT/mTOR pathway and downregulated apoptosis-related proteins in PCOS rats. CONCLUSION: BSZYD can restore ovary lesions and ameliorate apoptosis through ER α-mediated PI3K/AKT/mTOR pathway, which might partly contribute to the treatment of PCOS.


Subject(s)
Apoptosis/drug effects , Drugs, Chinese Herbal/pharmacology , Granulosa Cells/drug effects , Polycystic Ovary Syndrome/drug therapy , Animals , Disease Models, Animal , Down-Regulation/drug effects , Estrogen Receptor alpha/metabolism , Female , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
8.
Physiol Res ; 70(5): 755-764, 2021 11 29.
Article in English | MEDLINE | ID: mdl-34505534

ABSTRACT

Beneficial effects of Sambucus nigra L. (black elder) as a traditional medicine have been associated with the phytoconstituents including polyphenols, terpenes and lectins. Various antioxidant rich natural products have also been implicated with improvement of reproductive health and fertility, however, the effect of Sambucus nigra on the ovarian cell functions has not been investigated yet. The objectives of the present study were to screen the polyphenols in the elderflower and elderberry extracts, and to examine the secretion activity of steroid hormones 17beta-estradiol and progesterone by human ovarian granulosa cells HGL5 after supplementation of the extracts at a concentration range of 12.5 to 100 microg.ml-1. Qualitative as well as quantitative screening of polyphenols by high-performance liquid chromatography with diode-array detector (HPLC-DAD) analysis revealed rutin to be the most abundant polyphenol in both elderflower and elderberry extracts. In culture, neither elderflower nor elderberry extract caused any significant impact (p>0.05) in cell viability as studied by AlamarBlue assay in comparison to control. However, a dose-dependent stimulation of 17beta-estradiol release was detected by ELISA after supplementation of elderflower (at 50 microg.ml-1; p<0.01) and elderberry (at 100 microg.ml-1; p<0.05) extracts at higher doses used in the study. On the other hand, both elderflower and elderberry extracts stimulated the secretion of progesterone by HGL5 cells at a lower dose (12.5 microg.ml-1; p<0.05), as compared to control. Therefore, elderflower and elderberry extracts may have the potential to regulate steroidogenesis in ovarian cells.


Subject(s)
Gonadal Steroid Hormones/metabolism , Granulosa Cells/drug effects , Plant Extracts/administration & dosage , Cell Line , Female , Granulosa Cells/metabolism , Humans , Plant Extracts/chemistry , Sambucus nigra/chemistry
9.
Food Funct ; 12(18): 8800-8811, 2021 Sep 20.
Article in English | MEDLINE | ID: mdl-34374402

ABSTRACT

Polycystic ovarian syndrome (PCOS) is a complex endocrinopathy in women of reproductive age and the main cause of female infertility, but there is no universal drug for PCOS therapy. As a predominant dietary isoflavone present in soybeans, genistein (GEN) possesses estrogenic and antioxidative properties, but limited information is available regarding its therapeutic potential and underlying molecular mechanism in PCOS. In this study, we found that GEN might restore the estrous cycle of PCOS mice and ameliorate the elevation of circulating T, AMH and LH levels as well as LH/FSH ratios along with reduced cystic follicles, indicating the importance of GEN in PCOS therapy. Meanwhile, GEN improved the ovarian secretion function of PCOS mice and attenuated oxidative damage of the ovary through enhancing its antioxidant capability dependent on ER. Supplementation of GEN improved the defect of the ATP level and mitochondrial membrane potential, indicating the significance of GEN in preventing mitochondrial dysfunction. Further analysis demonstrated that GEN via ER heightened the expression of Nrf2 and Foxo1 whose blockage antagonized the defence of GEN on the secretory and mitochondrial functions of ovarian granulosa cells followed by the limited antioxidant capability and increased intracellular ROS level. Moreover, nuclear translocation and transcriptional activity of Nrf2 presented a notable enhancement after exposure to GEN. Addition of the Nrf2 inhibitor ML385 hampered the GEN induction of Foxo1. Nrf2 might directly bind to the antioxidant response element of the Foxo1 promoter region. Collectively, GEN might exhibit therapeutic potential for PCOS mice via the ER-Nrf2-Foxo1-ROS pathway.


Subject(s)
Forkhead Box Protein O1/metabolism , Genistein/therapeutic use , NF-E2-Related Factor 2/metabolism , Polycystic Ovary Syndrome/drug therapy , Reactive Oxygen Species/metabolism , Receptors, Estrogen/metabolism , Animals , Antioxidants/metabolism , Dehydroepiandrosterone/pharmacology , Female , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Mice , Mice, Inbred ICR , Mitochondria/drug effects , Mitochondria/metabolism , Ovary/drug effects , Ovary/metabolism , Oxidative Stress , Polycystic Ovary Syndrome/metabolism
10.
Clin Transl Sci ; 14(5): 2043-2054, 2021 09.
Article in English | MEDLINE | ID: mdl-33982403

ABSTRACT

Evidence from clinical cases indicates an association between the low success rate of in vitro fertilization (IVF) and ovarian injury due to previous methotrexate (MTX) administration. Therefore, it is necessary to develop and propose reasonable clinical drug guidelines to improve the quality of oocytes and the development of embryos before pregnancy. In this study, we established a mouse model with previous MTX exposure to validate the effects of MTX on reproductive function in female mice. We observed that MTX administration could result in a decrease in the success rate of fertilization and an aberrant embryonic development in both natural fertilization and IVF, even after completion of five to six ovulation cycles after MTX withdrawal. Further research revealed senescence and apoptosis of follicular granulosa cells (GCs), accompanied by arrested follicle development and aberrant estradiol and anti-Mullerian hormone levels. Supportive evidence indicated that MTX administration induced senescence and apoptosis of human GCs in vitro, and the effects were consistent with the high levels of p21, p53, and oxidative stress. We further demonstrated that folic acid (FA) could improve oocyte function and embryonic development in vivo and in vitro by protecting GCs against apoptosis and senescence. Based on these findings, we propose the implementation of extended intervals between MTX exposure and conception or IVF and recommend FA as a special dietary supplement during this interval period; however, prospective inquiry in humans is necessary to further understand the relationship between MTX and FA recovery.


Subject(s)
Embryonic Development/drug effects , Fertilization in Vitro/methods , Granulosa Cells/drug effects , Methotrexate/adverse effects , Oocytes/drug effects , Animals , Apoptosis/drug effects , Cell Line , Cellular Senescence/drug effects , Female , Folic Acid/pharmacology , Folic Acid/therapeutic use , Granulosa Cells/pathology , Humans , Male , Maternal Exposure/adverse effects , Mice , Models, Animal , Oocytes/growth & development , Oocytes/pathology , Prospective Studies , Treatment Outcome
11.
Physiol Res ; 70(2): 237-243, 2021 04 30.
Article in English | MEDLINE | ID: mdl-33992047

ABSTRACT

The objective of this study was to examine the direct effects of the medicinal plant fennel (Foeniculumvulgare Mill.) on basic functions of ovarian cells, including proliferation, apoptosis, and response to the physiological hormonal stimulator, ghrelin. In the first series of experiments, porcine ovarian granulosa cells were cultured with (1, 10, 100 µg/ml) or without fennel extract. In the second series of experiments, cells were cultured with (1, 10, 100 ng/ml) or without ghrelin, alone or in combination with fennel extract (10 µg/ml). Expression of the proliferation marker, PCNA, and the apoptosis marker, bax, were analyzed via quantitative immunocytochemical methods. Fennel stimulated the accumulation of the proliferation marker, and suppressed the expression of the apoptosis marker. Ghrelin alone promoted proliferation and apoptosis of ovarian cells. The presence of fennel inhibited these ghrelin effects. These observations provide the first demonstration of (1) effects of fennel on farm animal reproduction, (2) direct effects of fennel on ovarian cells, (3) the ability of fennel to promote ovarian cell proliferation, to inhibit ovarian cell apoptosis, and to enhance the ovarian cell proliferation:apoptosis ratio. Furthermore, our results (4) confirm the involvement of ghrelin in the control of ovarian cell apoptosis and proliferation, and (5) demonstrate the ability of fennel to affect not only ovarian cell proliferation and apoptosis, but also to suppress the responses of ovarian cells to the upstream hormonal regulator ghrelin. Our results indicate the potential applicability of fennel as a bio-stimulator of farm animal reproduction.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Foeniculum , Ghrelin/pharmacology , Granulosa Cells/drug effects , Plant Extracts/pharmacology , Animals , Cells, Cultured , Female , Foeniculum/chemistry , Granulosa Cells/metabolism , Granulosa Cells/pathology , Plant Extracts/isolation & purification , Proliferating Cell Nuclear Antigen/metabolism , Sus scrofa , bcl-2-Associated X Protein/metabolism
12.
J Tradit Chin Med ; 41(2): 203-211, 2021 04.
Article in English | MEDLINE | ID: mdl-33825399

ABSTRACT

OBJECTIVE: To investigate whether Zichong granules (, ZCKL), a very effective herbal formula for treating infertility, have an impact on the differentiation of ovarian granulosa cells from human embryonic stem cells (hESCs) in vitro, and to explore the cellular mechanisms of its clinical effects. METHODS: Serum from ZCKL-medicated rats was prepared and used to treat mesoderm cells derived from hESCs for 6 d. Normal rat serum and a set of growth factors were used as negative and positive controls, respectively. RESULTS: ZCKL-medicated rat serum, but not normal rat serum, induced hESCs-derived mesoderm cells to differentiate into functional ovarian granulosa-like cells (OGLCs) in a similar manner to defined growth factors. The induced OGLCs resembled the morphology of native human granulosa cells, expressed granulosa cell-specific markers at both the mRNA and protein levels, produced high levels of estradiol and strongly responded to follicle-stimulating hormone stimulation. Furthermore, mRNA levels of follistatin, mothers against decapentaplegic homolog 8 and bone morphogenetic protein 6 were dynamically changed during the process. CONCLUSION: In the ZCKL treatment of infertility, one mechanism by which ZCKL may act is by influencing ovarian granulosa cell differentiation and development, possibly through the follistatin and BMP/SMAD signaling pathways.


Subject(s)
Cell Differentiation/drug effects , Drugs, Chinese Herbal/pharmacology , Fertility Agents, Female/pharmacology , Granulosa Cells/drug effects , Human Embryonic Stem Cells/drug effects , Infertility, Female/physiopathology , Animals , Cells, Cultured , Female , Granulosa Cells/cytology , Human Embryonic Stem Cells/cytology , Humans , Infertility, Female/drug therapy , Ovary/cytology , Ovary/drug effects , Rats
13.
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
14.
Mol Biol Rep ; 48(4): 3515-3527, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33881728

ABSTRACT

The role of copper and selenium on activation of estradiol synthesis pathways viz. PKA/AKT/WNT is not clearly elucidated. On this background we attempt to elcuiated the role of copper and selenium on mRNA expression of genes associated with estradiol synthesis in caprine ovarian granulose cell models. Ovarian granulosa cells from medium (3-5 mm) sized follicles were aspirated and distributed separately to different groups. Group I: control, Group II: cupric chloride (Cu: 0.5 mM), Group III: sodium selenite (Se: 100 ng/ml), Group IV: Cu + Se. The cells (105/well) were cultured in 96 well plate in the base culture medium of MEMα comprising of nonessential amino acids (1.1 mM), FSH (10 ng/mL), transferrin (5 µg/mL), IGF-I (2 ng/mL), androstenedione (10-6 M), penicillin (100 IU/mL), streptomycin (0.1 mg/mL) and fungizone (0.625 µl/mL) and insulin (1 ng/mL). The cells were incubated in a carbondioxide incubator (38 °C, 5% CO2, 95% RH). The medium was changed on alternate days and cells were harvested on day 6. Day 6 media was used for estimation of estradiol. The RNA isolated form harvested cells was used for qPCR assay. There was no significant (p > 0.05) difference in estradiol concentration between groups. The mRNA expression of AKT1, CYP19A1, WNT2 & 4, FZD6 and APC2 were significantly (p < 0.05) higher in Cu and Cu + Se groups compared to control. Whereas, the mRNA transcript of DVL1 and CSNK1 was significantly (p < 0.05) higher in Cu + Se group compared to control. Incontrast, no significant difference in mRNA expression of PRKAR1A and CTNNB1 was noticed. Our study support a key role of copper and selenium in activation of AKT and WNT signalling pathway that further lead to increase in the mRNA expression of CYP19A1.


Subject(s)
Aromatase/genetics , Copper/pharmacology , Granulosa Cells/metabolism , Selenium/pharmacology , Animals , Aromatase/metabolism , Cells, Cultured , Female , Goats , Granulosa Cells/drug effects , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Wnt Signaling Pathway
15.
Ecotoxicol Environ Saf ; 214: 112091, 2021 May.
Article in English | MEDLINE | ID: mdl-33706141

ABSTRACT

The occurrence of cadmium (Cd) in feed is a major problem in animal health and production. Studies have confirmed that Cd depresses egg production of laying hens, which is closely related to follicular atresia. This study aimed to assess the toxic impacts of Cd on the ovarian tissue, and to examine the mechanism of Cd-induced granulosa cell proliferation and apoptosis. Results from the nitric oxide (NO) and malondialdehyde (MDA) content, total superoxide dismutase (T-SOD), glutathione peroxide (GSH-Px), total nitric oxide synthase (T-NOS) and adenosine triphosphatase (ATPase) activities, terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL) assay, and hematoxylin-eosin (H & E) staining indicated that excess Cd induced oxidative stress, granulosa cell apoptosis and follicular atresia in the layer ovary. Low-dose Cd exposure (1 µM) induced the granulosa cell proliferation, upregulated the mRNA levels of RSK1 and RHEB, activated FoxO3a, AKT, ERK1/2, mTOR and p70S6K1 phosphorylation, and promoted cell cycle progression from phase G1 to S. However, high-dose Cd exposure (15 µM) induced reactive oxygen species (ROS) generation and cell apoptosis, upregulated the mRNA levels of the inflammatory factors, ASK1, JNK, p38 and TAK1, downregulated the expressions of RSK1 and RHEB genes, and inhibited the phosphorylation of ERK1/2, mTOR and p70S6K1 proteins, and the cell cycle progression. Rapamycin pre-treatment completely blocked the phosphorylation of mTOR and p70S6K1 proteins, and the cell cycle progression induced by 1 µM Cd, and accelerated 15 µM Cd-induced cell apoptosis and cell cycle arrest. The microRNA sequencing result showed that 15 µM Cd induced differential expression of microRNA genes, which may regulate AKT, ERK1/2 and mTOR signaling and cell cycle progression by regulating the activity of G proteins and cell cycle-related proteins. Conclusively, these results indicated that Cd can cause the ovarian damage and follicular atresia, and regulate cell cycle, cell proliferation or apoptosis of granulosa cells through MAPK, AKT/FoxO3a and mTOR pathways in laying hens.


Subject(s)
Cadmium/toxicity , Granulosa Cells/drug effects , Animals , Apoptosis , Cell Cycle , Cell Cycle Checkpoints , Cell Division , Cell Proliferation , Chickens/metabolism , Female , Follicular Atresia , Granulosa Cells/metabolism , In Situ Nick-End Labeling , Oxidative Stress , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
16.
Endocrinology ; 162(8)2021 08 01.
Article in English | MEDLINE | ID: mdl-33693742

ABSTRACT

Polycystic ovary syndrome (PCOS), the most common endocrine disorder in women of reproductive age, is characterized by hyperandrogenism and insulin resistance (IR); however, the pathogenesis of local ovarian IR in PCOS remains largely unclear. Humanin, a mitochondria-derived peptide, has been reported to be associated with IR. Our previous study confirmed that humanin is expressed in multiple cell types in the ovary and is present in follicular fluid. However, it remains unknown whether humanin participates in the pathogenesis of local ovarian IR or whether humanin supplementation can improve IR in PCOS patients. In this study, we compared humanin concentrations in follicular fluid from PCOS patients with and without IR. We further investigated the effect of humanin analogue (HNG) supplementation on IR in a rat model of dehydroepiandrosterone-induced PCOS. Humanin concentrations in the follicular fluid were found to be significantly lower in PCOS patients with IR than in those without IR. HNG supplementation attenuated both the increases in the levels of fasting plasma glucose and fasting insulin in rats with PCOS and the decreases in phosphorylation of IRS1, PI3K, AKT, and GLUT4 proteins in the granulosa cells of these rats. Combined supplementation with HNG and insulin significantly improved glucose consumption in normal and humanin-siRNA-transfected COV434 cells. In conclusion, downregulated humanin in the ovaries may be involved in the pathogenesis of IR in PCOS, and exogenous supplementation with HNG improved local ovarian IR through modulation of the IRS1/PI3K/Akt signaling pathway in a rat model. This finding supports the potential future use of HNG as a therapeutic drug for PCOS.


Subject(s)
Follicular Fluid/metabolism , Granulosa Cells/drug effects , Insulin Resistance , Intracellular Signaling Peptides and Proteins/blood , Polycystic Ovary Syndrome/blood , Adult , Animals , Case-Control Studies , Cell Line , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Glucose Transporter Type 4/metabolism , Granulosa Cells/metabolism , Humans , Insulin Receptor Substrate Proteins/metabolism , Intracellular Signaling Peptides and Proteins/pharmacology , Intracellular Signaling Peptides and Proteins/therapeutic use , Phosphatidylinositol 3-Kinases/metabolism , Polycystic Ovary Syndrome/drug therapy , Primary Cell Culture , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , Young Adult
17.
Endocr Regul ; 55(1): 52-60, 2021 Jan 29.
Article in English | MEDLINE | ID: mdl-33600664

ABSTRACT

Objectives. The application of nanoparticles is experiencing a rapid growth, but it faces a problem of their toxicity, especially adverse effects on female reproduction. Food and medicinal plants and their isoflavones can be protectors against environmental stressors, but their ability to abate the adverse effects of nanoparticles has not been studied yet. In the present study, we examined the effect of silver (AgNPs) and titanium dioxide (titania, TiO2NPs) nanoparticles alone or in combination with plant phytoestrogens/antioxidants (resveratrol, diosgenin, and quercetin) on accumulation of nanoparticles, and progesterone release by cultured porcine ovarian granulosa cells.Methods. Porcine granulosa cells were incubated in the presence of AgNPs or TiO2NPs (0.1, 1, 10 or 100 µg/ml) alone or in combination with resveratrol, diosgenin or quercetin (10 µg/ml) for 48 h. The accumulation of tested nanoparticles by granulosa cells was assessed under light microscope. Progesterone concentration in culture media was measured by ELISA kit.Results. Cells accumulated both AgNPs and TiO2NPs in a dose-dependent manner. AgNPs, but not TiO2NPs, at highest dose (100 µg/ml) resulted in a destruction of cell monolayer. Both Ag-NPs and TiO2NPs reduced progesterone release. Resveratrol, diosgenin, and quercetin promoted accumulation of both AgNPs and TiO2NPs in ovarian cells and inhibited the progesterone output. Furthermore, resveratrol and diosgenin, but not quercetin, prevented the suppressive action of both AgNPs, and TiO2NPs on progesterone release.Conclusions. These observations (1) demonstrate accumulation of AgNPs and TiO2NPs in ovarian cells, (2) confirm the toxic impact of AgNPs, and TiO2NPs on these cells, (3) confirm the inhibitory effects of plant polyphenols/phytoestrogens on ovarian steroidogenesis, (4) show the ability of these isoflavones to increase the accumulation of AgNPs and TiO2NPs, and (5) show their ability to reduce the suppressive effect of AgNPs and TiO2NPs on ovarian progesterone release. The suppressive effect of AgNPs and TiO2NPs on ovarian functions should be taken into account by their exposition. However, these adverse effects could be mitigated by some plant isoflavones.


Subject(s)
Granulosa Cells/metabolism , Isoflavones/pharmacology , Metal Nanoparticles/toxicity , Silver/metabolism , Titanium/metabolism , Animals , Cells, Cultured , Diosgenin/pharmacology , Female , Granulosa Cells/drug effects , Progesterone/metabolism , Quercetin/pharmacology , Resveratrol/pharmacology , Silver/toxicity , Swine , Titanium/toxicity
18.
Anim Reprod Sci ; 225: 106681, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33421819

ABSTRACT

The objective was to investigate effects of progesterone (P4) dose on abundance of luteinizing hormone receptor (LHCGR), aromatase (CYP19A1), 3ß-hydroxysteroid dehydrogenase (HSD3B1), and other steroidogenic mRNA transcripts in granulosa cells from dominant follicles. Nellore heifers were assigned to one of six groups: new, first-use controlled internal drug release device (CIDR1) inserted for 5 days (Large-P4-dose-D5; n = 7) or 6 days (Large-P4-dose-D6; n = 8), prostaglandin (PG)F2α administered on D0 and 1 previously-used CIDR (CIDR3) inserted for 5 days (Small- P4-dose-D5; n = 8) or 6 days (Small-P4-dose-D6; n = 8), CIDR1 inserted on D0 and removed plus PGF2α on D5 (Large-P4-dose-proestrus (PE); n = 7), and CIDR3 and PGF2α on D0 and 1, CIDR3 removed plus PGF2α on D5 (Small-P4-dose-PE; n = 7). Duration of P4 treatment (D5 compared to D6) affected abundances of CYP19A1 mRNA transcripts, with there being greater abundances on D6 than D5 (P ≤ 0.05). Heifers treated with the large dose of P4 had a smaller dominant follicle, less serum and intra-follicular estradiol (E2) concentrations (P ≤ 0.05) and lesser LHCGR, CYP19A1, and HSD3B1 transcript abundances (P ≤ 0.05). Heifers treated to induce PE had a larger follicle diameter (P = 0.09), greater intra-follicular E2 concentrations and larger abundances of CYP19A1 mRNA transcript (P ≤ 0.05) than heifers of the D6 group. Overall, treatment with larger doses of P4 resulted in lesser abundances of LHCGR, HSD3B1, and CYP19A1 mRNA transcripts; thus, potentially leading to development of smaller dominant follicles and lesser E2 concentrations.


Subject(s)
Cattle , Estrus Synchronization/drug effects , Progesterone/pharmacology , Receptors, LH/metabolism , Animals , Aromatase/genetics , Aromatase/metabolism , Cholesterol Side-Chain Cleavage Enzyme/genetics , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Dinoprost/administration & dosage , Dinoprost/analogs & derivatives , Dinoprost/pharmacology , Estradiol/administration & dosage , Estradiol/analogs & derivatives , Estradiol/pharmacology , Female , Gene Expression Regulation/drug effects , Granulosa Cells/drug effects , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Progesterone/administration & dosage , Progesterone Reductase/genetics , Progesterone Reductase/metabolism , Receptors, LH/genetics , Steroid Isomerases/genetics , Steroid Isomerases/metabolism
19.
J Ethnopharmacol ; 270: 113821, 2021 Apr 24.
Article in English | MEDLINE | ID: mdl-33460753

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Guizhi Fuling Wan (GFW) is a traditional Chinese medicine used to remove blood stasis and dissipate phlegm for treating gynecological diseases that was invented by Zhang Zhongjing in the Eastern Han dynasty. In recent years, GFW has been widely used to treat patients with polycystic ovary syndrome (PCOS). Clinical and animal studies have shown that it is effective in the treatment of PCOS, but its mechanism is unknown. Generally, it works by regulating autophagy via the PI3K/AKT/mTOR signaling pathway. AIM OF THE STUDY: This study investigated the effects and mechanism of GFW in PCOS rats with insulin resistance (IR) in order to provide better understanding of its observed clinical effects and a theoretical basis for the study of traditional Chinese medicine. MATERIALS AND METHODS: Eighty-four female Sprague-Dawley rats were randomly divided into seven groups (n = 12 per group): 1) control, 2) PCOS model, 3) low-dose GFW, 4) medium-dose GFW, 5) high-dose GFW, 6) metformin, and 7) medium-dose GFW plus LY294002. In all non-control groups, we induced PCOS through daily letrozole combined with intragastric high-fat emulsion for 21 days. After treatment, rats were sacrificed and serum follicle-stimulating hormone (FSH), testosterone (T), progesterone, luteinizing hormone (LH), 17ß-estradiol, fasting insulin (FINS), and fasting plasma glucose levels were measured by enzyme-linked immunosorbent assay (ELISA). The LH/FSH ratios and HOMA-IR values were calculated. Ovarian morphology was observed by hematoxylin and eosin staining, and all follicles were counted under a microscope. MDC-positive vesicles were used as markers to detect autophagy, and the expression levels of p62, Beclin1, and LC3-II were examined by immunostaining. Western blotting was used to measure PI3K/AKT/mTOR pathway activation, granulosa cell apoptosis, and autophagy. RESULTS: Compared with the PCOS model group, GFW-treated rats had less atretic and cystic follicles, and more mature follicles and corpus lutea. The GFW-treated rats had lower serum T, LH, and FINS levels than the PCOS model group, as well as lower LH/FSH ratios and HOMA-IR values. GFW treatment resulted in significantly reduced levels of cleaved-Caspase-3, cleaved-Caspase-9, BAX, Beclin1, Atg5, and LC3-II. Phosphorylation of PI3K, AKT, and mTOR was significantly higher in GFW-treated rats compared with the PCOS model group. The phosphorylation of PI3K, AKT, and mTOR was decreased with the use of a PI3K antagonist. CONCLUSIONS: Our results indicate that GFW inhibited granulosa cell autophagy and promoted follicular development to attenuate ovulation disorder in PCOS-IR rats. This was associated with activation of the PI3K/AKT/mTOR signaling pathway.


Subject(s)
Autophagy/drug effects , Drugs, Chinese Herbal/pharmacology , Granulosa Cells/drug effects , Polycystic Ovary Syndrome/drug therapy , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/metabolism , Beclin-1/genetics , Beclin-1/metabolism , Caspase 3/genetics , Caspase 3/metabolism , Caspase 9/genetics , Caspase 9/metabolism , Disease Models, Animal , Down-Regulation , Drugs, Chinese Herbal/therapeutic use , Female , Hormones/blood , Insulin Resistance , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Ovarian Follicle/drug effects , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Polycystic Ovary Syndrome/pathology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Up-Regulation , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
20.
J Ethnopharmacol ; 277: 113789, 2021 Sep 15.
Article in English | MEDLINE | ID: mdl-33422655

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Zihuai recipe (ZHR), a Chinese herbal prescription, is widely used for the clinical treatment of Diminished ovarian reserve (DOR) infertility. However, little is known regarding its underlying mechanisms of DOR treatment. AIM OF THE STUDY: This study aimed to investigate the beneficial effects of ZHR on the treatment of DOR and to reveal the underlying mechanisms. MATERIALS AND METHODS: Sixty female 8-week-old Sprague-Dawley rats were randomly divided into the following six groups (n=10 per group): control, DOR, low-dose(2.7 g/kg/day) ZHR (L-ZHR), medium-dose(5.4 g/kg/day), ZHR (M-ZHR), high-dose(10.8 g/kg/day) ZHR (H-ZHR), and hormone replacement therapy (HRT) treatment groups. The DOR model was established in all the groups, except the control group, by a single intraperitoneal injection of 90 mg/kg cyclophosphamide. After the induction of the DOR model, rats were weighed and administered either the relevant dose of ZHR or an equal volume of saline solution (in the control and DOR groups). Rats in the HRT group received estradiol valerate tablets (0.16 mg/kg/day), and with medroxyprogesterone acetate tablets (0.86 mg/kg/day) added on day 4. After 32 days of treatment, the rats were euthanized and the ovaries were collected for sampling. Ovarian morphology was observed by hematoxylin and eosin staining and the number of follicles was counted under a microscope. The serum levels of anti-Müllerian hormone (AMH), gonadotropin-releasing hormone (GnRH), follicle-stimulating hormone (FSH), luteinizing hormone (LH), and estradiol (E2) were quantified by ELISA. A TUNEL assay was used to analyze the level of apoptosis of the ovarian cells. The protein expressions of p-PI3K, p-AKT, PI3K, AKT, cleaved caspase-3, BAX, and Bcl-2 were measured by western blotting and immunohistochemistry. Data analysis was performed with SPSS 20.0 software. RESULTS: ZHR administration increased the ovarian index and the serum levels of AMH, GnRH, and E2, while lowering those of FSH and LH. ZHR treatment also increased the number of primordial, primary, secondary, and antral follicles, as well as the number of corpora lutea, but decreased the number of atretic follicles. Furthermore, ZHR administration decreased the percentage of TUNEL-positive ovarian cells. After treatment with ZHR, the protein expression levels of p-PI3K/PI3K, p-AKT/AKT, cleaved caspase-3 and BAX were decreased, whereas the level of Bcl-2 was increased. CONCLUSIONS: ZHR improved the ovarian reserve in CTX-induced DOR rats. The mechanisms of ZHR on DOR may be mediated through the regulation of gonadal hormones of the hypothalamic-pituitary-ovarian axis (HPOA), and the inhibition of PI3K/AKT-mediated apoptosis in granulosa cells.


Subject(s)
Cyclophosphamide/toxicity , Drugs, Chinese Herbal/pharmacology , Ovarian Reserve/drug effects , Animals , Antineoplastic Agents, Alkylating/toxicity , Apoptosis/drug effects , Dose-Response Relationship, Drug , Estradiol/administration & dosage , Female , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Hormone Replacement Therapy/methods , Hypothalamo-Hypophyseal System/metabolism , Ovarian Follicle/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL