1.Protective effect of follicle stimulating hormone on apoptosis of human epithelial ovarian cancer cell induced by cisplatin.
Chun-fang HUANG ; Dong-yuan LIU ; Wei-hua XU ; Keng SHEN
Acta Academiae Medicinae Sinicae 2003;25(4):443-446
OBJECTIVETo observe the protective effects of follicle stimulating hormone (FSH) on human epithelial ovarian cancer cell apoptosis induced by cisplatin (DDP).
METHODSOVCAR3-FSHR cell were treated with DDP and FSH at serials of concentrations, MTT assay was used to examine the growth inhibition of OVCAR3-FSHR cell after treatment with DDP and FSH. Flow cytometry was used to analyze the change of cell cycle and percentage of apoptosis.
RESULTSIt was revealed that FSH decreased the growth inhibition induced by DDP. We also demonstrated that FSH reduced the S-phase percentage compared with the DDP only groups after treatment for 24 hours and reduced apoptosis percentage after 48 hours treatment with DDP.
CONCLUSIONIt is suggested that FSH can protect the apoptosis induced by DDP. It also suggests that FSH may be an important chemoresistent reason for the chemotherapy of ovarian cancer.
Antineoplastic Agents ; pharmacology ; Apoptosis ; drug effects ; Cisplatin ; pharmacology ; Female ; Flow Cytometry ; Follicle Stimulating Hormone ; pharmacology ; Humans ; Ovarian Neoplasms ; chemistry ; pathology ; Receptors, FSH ; analysis ; Tumor Cells, Cultured
2.Effect of zuoguiyin on the expression of ovarian follicle-stimulating hormone receptor in rats during peri-menopausal period.
Wei ZHAO ; Hai-xia WEN ; Hui-li ZHENG
Chinese Journal of Integrated Traditional and Western Medicine 2010;30(3):286-290
OBJECTIVETo investigate the effect and mechanism of Zuoguiyin (ZGY, a Chinese medical remedy for regulating qi and nourishing yin) on expression of ovarian follicle-stimulating hormone receptor (FSHR) in rats during peri-menopausal period (PMP).
METHODSPMP rats were administered with low (13.78 g/kg), middle (20.67 g/kg) and high (31.00 g/kg) dose ZGY respectively by gastrogavage for eight weeks. FSHR mRNA and protein expressions were detected by RT-PCR and immunohistochemistry respectively. Besides, granulosa cells, collected from pregnant mare serum treated nonage rats, were incubated, and the level of FSHR mRNA expression in the cultured cells were detected after various disposals.
RESULTS(1) Ovarian levels of FSHR mRNA and protein expressions in PMP rats were significantly lower than those in young rats (P<0.01), but were up-regulated significantly by ZGY treatment (P<0.01). (2) As compared with the control, FSHR mRNA expression increased in cultured granulosa cells after treated by ZGY extract; the increasing effect of ZGY was enhanced by combined treatment with Forskolin and attenuated by Genistein (a tyrosine protein kinase inhibitor).
CONCLUSIONZGY could improve the ovarian functions during PMP by up-regulating the expression of FSHR and raise the ovarian responsibility to FSH, which may be possibly realized by activating cAMP-protein kinase and tyrosine protein kinase signal pathway.
Animals ; Cells, Cultured ; Drugs, Chinese Herbal ; pharmacology ; Female ; Ovary ; drug effects ; metabolism ; Perimenopause ; drug effects ; RNA, Messenger ; genetics ; Rats ; Rats, Wistar ; Receptors, FSH ; metabolism
3.Regulation of gonadotropin releasing hormone receptor mRNA expression in cultured rat granulosa cells.
Yongbin PARK ; Kyungyoon KAM ; Minseok CHEON ; Kyungza RYU
Yonsei Medical Journal 2001;42(2):233-241
The homologous regulation of pituitary Gonadotropin Releasing Hormone Receptor (GnRH-R) mRNA expression by GnRH has been well demonstrated. However, the regulation of the ovarian GnRH-R is poorly understood. The present study was performed to demonstrate the presence of GnRH transcripts in addition to GnRH-R mRNA and the regulation of GnRH-R mRNA expression in the granulosa cells isolated from small antral follicles. The GnRH and GnRH-R mRNA levels were determined by a competitive reverse transcription-polymerase chain reaction (RT-PCR). The granulosa cells were obtained from immature rats implanted with diethylstilbestrol for 3 days. When GnRH transcript expression was examined in isolated granulosa cells by RT-PCR, the PCR products showed two bands. The larger band contained intronic sequences and the smaller band was a fully processed GnRH gene transcript identical to hypothalamic GnRH. This suggests that authentic GnRH gene transcripts are expressed in ovarian granulosa cells and may act on the granulosa cells in a paracrine or autocrine manner. Since GnRH action in the granulosa cells is mediated by specific GnRH-R, it is of interest to examine whether GnRH-R is synthesized in the granulosa cells. When the granulosa cells were cultured in media only, GnRH-R mRNA levels increased abruptly within 3 h and gradually decreased thereafter during the 24 h culture period. However, GnRH itself did not alter the GnRH-R mRNA expression levels in cultured granulosa cells. Interestingly, treatment with FSH decreased the GnRH-R mRNA levels in a dose-dependent manner. A time-course analysis revealed that the GnRH-R mRNA levels were significantly lower up to 9 h after FSH treatment, and returned to the basal level between 12 h-24 h. Activation of adenylate cyclase with forskolin also decreased the GnRH-R mRNA levels. It is therefore concluded that in the granulosa cells of the small antral follicles GnRH-R mRNA expression was not homologously regulated by GnRH, while FSH may negatively regulate GnRH-R mRNA expression in the granulosa cells possibly through a cAMP-protein kinase A pathway.
Animal
;
Cells, Cultured
;
FSH/pharmacology
;
Female
;
Gene Expression Regulation*
;
Gonadorelin/pharmacology
;
Granulosa Cells/metabolism*
;
Granulosa Cells/drug effects
;
RNA, Messenger/metabolism*
;
Rats
;
Rats, Sprague-Dawley
;
Receptors, LHRH/genetics*
;
Reverse Transcriptase Polymerase Chain Reaction
4.The molecular design and drug development of recombinant long-acting follicle stimulating hormone.
Acta Pharmaceutica Sinica 2012;47(4):421-426
Follicle-stimulating hormone (FSH) is a glycoprotein which regulates the development, growth, pubertal maturation and reproductive processes of the body. Exogenous FSH has been used to promote ovarian follicular growth and maturation in female and spermatogenesis in male. The relative short elimination half life and rapid metabolic clearance of current versions of FSH require a daily or twice-daily scheduled subcutaneous injection to maintain stable FSH level being not below the threshold during ovarian stimulation. The development of recombinant long-acting FSH with enhanced biological activities may be helpful for less injection therefore to improve patient compliance, while reducing patient stress and error rates. A number of technological strategies have been explored to develop recombinant longer-acting FSH. For examples, attachment of the C-terminal peptide (CTP) of the human chorionic gonadotropin beta subunit or a sequence containing potential glycosylation sites to either subunit of FSH, creation of a single chain containing the alpha and beta subunits of FSH combined with CTP or N-linked glycosylation signal sequence as a linker, or fusion of the Fc domain of IgGi to FSH. Based on the modifiable molecular structure and pharmacokinetic and pharmacodynamic properties of recombinant FSH, it is hopeful that more FSH drugs with prolonged half-life and increased bioactivity will be developed to meet the modern clinical demands.
Animals
;
Follicle Stimulating Hormone, Human
;
chemistry
;
genetics
;
metabolism
;
pharmacology
;
Glycosylation
;
Half-Life
;
Humans
;
Immunoglobulin Fc Fragments
;
chemistry
;
metabolism
;
Ovulation Induction
;
methods
;
Receptors, FSH
;
chemistry
;
metabolism
;
Recombinant Fusion Proteins
;
chemistry
;
genetics
;
metabolism
;
pharmacology
;
Reproduction
;
drug effects
5.Effects of Yangling Zhongyu Decoction on the secretion of ovarian granule cells in polycystic ovarian syndrome rat model.
Hong-Xia MA ; Jun XIE ; Mao-Hua LAI
Chinese Journal of Integrated Traditional and Western Medicine 2012;32(1):54-57
OBJECTIVETo observe the effects of Yangjing Zhongyu Decoction (YJZYD) on the serum estradiol (E2), testosterone (T), 17-hydroxyprogesterone (17-OHP), ovarian follicle stimulating hormone receptor (FSHR), insulin-like growth factor I (IGF-1), steroid hormone acute regulator protein (StAR) mRNA expressions in female rats.
METHODSFifty PCOS rats were equally divided into 5 groups, i. e., the control group (C, normal PNA rats), the model group (M), the low dose YJZYD group (Y1), the medium dose YJZYD group (Y2), and the high dose YJZYD group (Y3), 10 in each. The levels of serum hormones were detected using radioimmunoassay. The morphological changes of the ovary were observed using HE method. The expressions of FSHR, IGF-1, and StAR mRNA were detected using RT PCR.
RESULTSCompared with Group C, serum T and 17-OHP significantly increased (P < 0.01), E2 significantly decreased (P < 0.01), the expressions of FSHR, IGF-1, and StAR mRNA significantly decreased in Group M (P < 0.01). Compared with Group M, the serum T level significantly decreased (P < 0.01), 17-OHP decreased (P < 0.05), and E2 significantly increased in Group Y3 (P < 0.01). The expressions of FSHR, IGF-1, and StAR mRNA increased in Group Y1, Y2, and Y3. The increase was most obvious in Group Y3 (P < 0.01).
CONCLUSIONSYJZYD could lower the hyperandrogenemia of PCOS rats. It also could increase the ovarian expressions of FSHR, IGF-1, and StAR mRNA, improve the ovarian functions, and promote the follicular development.
17-alpha-Hydroxyprogesterone ; blood ; Androgens ; blood ; Animals ; Drugs, Chinese Herbal ; pharmacology ; Estradiol ; blood ; Female ; Insulin-Like Growth Factor I ; metabolism ; Ovary ; drug effects ; metabolism ; Polycystic Ovary Syndrome ; blood ; Rats ; Rats, Wistar ; Receptors, FSH ; blood ; Testosterone ; blood
6.An in vitro prototype of a porcine biomimetic testis-like cell culture system: a novel tool for the study of reassembled Sertoli and Leydig cells.
Iva ARATO ; Giovanni LUCA ; Francesca MANCUSO ; Catia BELLUCCI ; Cinzia LILLI ; Mario CALVITTI ; Barbara C HANSEN ; Domenico MILARDI ; Giuseppe GRANDE ; Riccardo CALAFIORE
Asian Journal of Andrology 2018;20(2):160-165
At present, there is no reliable in vitro assembled prepubertal testis-like biomimetic organ culture system designed to assess the functional effects of human gonadotropins on Sertoli and Leydig cells. Spermatogenesis is regulated by endocrine, paracrine, and juxtacrine factors (testicular cross-talk), mainly orchestrated by gonadotropins such as luteinizing hormone (LH) and follicle-stimulating hormone (FSH) that play a pivotal role by stimulating Leydig and Sertoli cells, respectively. The aim of our study was to set up an in vitro prepubertal porcine bioengineered construct as a new model for experimental studies on reassembled Sertoli and Leydig cells. We have evaluated Sertoli and Leydig cells obtained from 15- to 20-day-old neonatal pig testes in terms of purity and function. Subsequently, purified Sertoli and enriched Leydig cells were subjected to coincubation to obtain an in vitro prepubertal porcine testis-like culture system. We performed enzyme-linked immunosorbent assay (ELISA) for anti-Müllerian hormone (AMH), inhibin B, and testosterone secretion in the medium, and Real-Time PCR analysis of AMH, inhibin B, FSH-r, aromatase, LHr, and 3β-HSD mRNA expression levels. This in vitro testis-like system was highly responsive to the effects of human gonadotropins and testosterone. AMH mRNA expression and secretion declined, and inhibin-B increased, while FSH-receptor expression was downregulated upon FSH/LH exposure/treatment. Finally, the production of testosterone was increased selectively upon LH treatment. In summary, our proposed model could help to better determine the action of human gonadotropins on Sertoli and Leydig cells. The potential usefulness of the system for shedding light into male infertility-related issues is evident.
3-Hydroxysteroid Dehydrogenases/metabolism*
;
Animals
;
Animals, Newborn
;
Anti-Mullerian Hormone/metabolism*
;
Aromatase/metabolism*
;
Cell Culture Techniques
;
Enzyme-Linked Immunosorbent Assay
;
Follicle Stimulating Hormone/pharmacology*
;
Hormones/pharmacology*
;
In Vitro Techniques
;
Inhibins/metabolism*
;
Leydig Cells/metabolism*
;
Luteinizing Hormone/pharmacology*
;
Male
;
Models, Biological
;
Real-Time Polymerase Chain Reaction
;
Receptors, FSH/metabolism*
;
Receptors, LH/metabolism*
;
Sertoli Cells/metabolism*
;
Swine
;
Testis/metabolism*
;
Testosterone/metabolism*
7.High levels of testosterone inhibit ovarian follicle development by repressing the FSH signaling pathway.
Tao LIU ; Yu-qian CUI ; Han ZHAO ; Hong-bin LIU ; Shi-dou ZHAO ; Yuan GAO ; Xiao-li MU ; Fei GAO ; Zi-jiang CHEN
Journal of Huazhong University of Science and Technology (Medical Sciences) 2015;35(5):723-729
The effect of high concentrations of testosterone on ovarian follicle development was investigated. Primary follicles and granulosa cells were cultured in vitro in media supplemented with a testosterone concentration gradient. The combined effects of testosterone and follicle-stimulating hormone (FSH) on follicular growth and granulosa cell gonadotropin receptor mRNA expression were also investigated. Follicle growth in the presence of high testosterone concentrations was promoted at early stages (days 1-7), but inhibited at later stage (days 7-14) of in vitro culture. Interestingly, testosterone-induced follicle development arrest was rescued by treatment with high concentrations of FSH (400 mIU/mL). In addition, in cultured granulosa cells, high testosterone concentrations induced cell proliferation, and increased the mRNA expression level of FSH receptor (FSHR), and luteinized hormone/choriogonadotropin receptor. It was concluded that high concentrations of testosterone inhibited follicle development, most likely through regulation of the FSH signaling pathway, although independently from FSHR downregulation. These findings are an important step in further understanding the pathogenesis of polycystic ovary syndrome.
Androgens
;
pharmacology
;
Animals
;
Cell Proliferation
;
drug effects
;
Female
;
Follicle Stimulating Hormone
;
genetics
;
metabolism
;
pharmacology
;
Gene Expression Regulation, Developmental
;
Granulosa Cells
;
cytology
;
drug effects
;
metabolism
;
Mice
;
Ovarian Follicle
;
cytology
;
drug effects
;
growth & development
;
metabolism
;
Primary Cell Culture
;
RNA, Messenger
;
genetics
;
metabolism
;
Receptors, FSH
;
genetics
;
metabolism
;
Receptors, Gonadotropin
;
genetics
;
metabolism
;
Receptors, LH
;
genetics
;
metabolism
;
Signal Transduction
;
drug effects
;
genetics
;
Testosterone
;
antagonists & inhibitors
;
pharmacology
8.Effects of bushen tiaochong recipe containing serum on ovarian granulosa cell proliferation, steroidogenesis and associated gene expression in rats.
Tian XIA ; Song-ping LUO ; Yu FU ; Bing HAN
Chinese journal of integrative medicine 2007;13(3):200-205
OBJECTIVETo observe the effect of bushen tiaochong recipe (BSTCR) on rats' ovarian granulosa cell (GC) proliferation, steroidogenesis and follicle-stimulating hormone receptor (FSHR), and insulin-like growth factor-1 (IGF-1) mRNA expression using serum pharmacological method.
METHODSRats' GCs were incubated with 10% blank serum (as negative control group), follicle-stimulating hormone (FSH)-containing serum (S-FSH, as positive control group), or BSTCR (in different dosages) containing serum (S-BSTCR, as the BSTCR groups) for 48 h. 3H-TdR incorporation was then performed; DNA was measured to analyze the distribution of GCs in the cell cycle and their proliferation index (PI) using a flow cytometer; estradiol (E2) and progesterone (P) content in the culture fluid were examined by radioimmunoassay; and levels of FSHR and IGF-1 mRNA expression in GCs were measured by real-time RT-PCR.
RESULTSA dose-dependent increase of 3H-TdR incorporation in GC was shown in the BSTCR groups. Cells in G0/G1 phase had markedly less, while those in S phase had a significantly higher increase in the BSTCR groups compared with the negative control group. A high value of PI was also shown in the BSTCR groups, especially in the high dose group where the influence of cell proliferation was stronger than that in the positive control group. The levels of E2 and P in the BSTCR groups of all dosages were significantly higher than those in the negative control group, and did not show any significant difference compared with those in the positive control group. Levels of FSHR and IGF-1 mRNA expression in the BSTCR groups increased in a dose-dependent manner at levels higher than those in the negative control group.
CONCLUSIONS-BSTCR can obviously stimulate the proliferation and steroidogenesis of ovarian GCs. It is speculated that BSTCR could play a regulatory action on ovarian function through two different pathways of endocrine and autocrine by promoting FSHR and IGF-1 mRNA expression.
Animals ; Cell Cycle ; drug effects ; Cell Proliferation ; drug effects ; DNA ; biosynthesis ; Drugs, Chinese Herbal ; pharmacology ; Estradiol ; pharmacology ; Female ; Gene Expression Regulation ; drug effects ; Granulosa Cells ; cytology ; drug effects ; Humans ; Insulin-Like Growth Factor I ; genetics ; metabolism ; Progesterone ; pharmacology ; RNA, Messenger ; genetics ; metabolism ; Rats ; Rats, Sprague-Dawley ; Receptors, FSH ; genetics ; metabolism ; Steroids ; biosynthesis ; Tritium
9.Follicle stimulating hormone inhibits cisplatin induced apoptosis in ovarian cancer cells.
Chun-fang HUANG ; Dong-yuan LIU ; Keng SHEN
Acta Academiae Medicinae Sinicae 2003;25(4):447-450
OBJECTIVETo investigate whether the follicle stimulating hormone (FSH) can inhibit apoptosis in ovarian cancer cells induced by cisplatin (DDP) and its possible mechinism.
METHODSDNA fragmentation assay, (TdT-mediated dUTP nick end labling TUNEL), Western blot were used to analyze the changes in expression levels of Survivin and bcl-2 protein. The relative activity of caspase-3 was also determined.
RESULTS200 mIU/ml FSH could regulate down the percentage of apoptotic cells and DNA fragmentation induced by 5.0 micrograms/ml cisplatin, while 200 mIU/ml FSH increased Survivin protein expression but could't influence the expression of bcl-2 protein.
CONCLUSIONFSH can inhibit ovarian cancer cells apoptosis induced by cisplatin. The possible mechinism is up-regulation of Survivin expression and down-regulation of caspase activity.
Antineoplastic Agents ; antagonists & inhibitors ; pharmacology ; Apoptosis ; drug effects ; Caspase 3 ; Caspases ; metabolism ; Cisplatin ; antagonists & inhibitors ; pharmacology ; DNA Fragmentation ; Female ; Flow Cytometry ; Follicle Stimulating Hormone ; pharmacology ; Humans ; Inhibitor of Apoptosis Proteins ; Microtubule-Associated Proteins ; metabolism ; Neoplasm Proteins ; Ovarian Neoplasms ; metabolism ; pathology ; Proto-Oncogene Proteins c-bcl-2 ; metabolism ; Receptors, FSH ; metabolism ; Tumor Cells, Cultured
10.Effect of Yangjing Zhongyu Decoction on mRNA and protein expression of PCNA, StAR, and FSHR in ovarian granulosa cells cultured by excess androgen.
Yan-Hua ZHENG ; Tao DING ; Hong-Xia MA ; Dan-Feng YE ; Nian-Jun SU ; Xiao-Ke WU
Chinese Journal of Integrated Traditional and Western Medicine 2014;34(3):312-316
OBJECTIVETo observe the effect of Yangjing Zhongyu Decoction (YZD) on mRNA and protein expression of PCNA, StAR, and FSHR in ovarian granulose cells (GCs) cultured by excess androgen.
METHODSOvarian GCs from porcine follicles were isolated and cultured in vitro. Follicular stimulating hormone (FSH) or YZD was added in the GCs treated by excess testosterone propionate. Totally 48 h later mRNA and protein expression of PCNA, StAR, and FSHR were detected by RT-PCR and Western blot.
RESULTSExcess androgen inhibited mRNA and protein expression of PCNA, StAR, and FSHR of GCs. FSH and YZD could antagonize inhibition of excess androgens, and promote mRNA and protein expression of PCNA, StAR, and FSHR in GCs.
CONCLUSIONYZD could antagonize the inhibition of excess androgen on mRNA and protein expression of PCNA, StAR and FSHR in GCs. Thus, we inferred that YZD could improve the follicle dysplasia by promoting mRNA and protein expression of PCNA, StAR and FSHR in GCs.
Androgens ; pharmacology ; Animals ; Cells, Cultured ; Drugs, Chinese Herbal ; pharmacology ; Female ; Follicle Stimulating Hormone ; pharmacology ; Granulosa Cells ; cytology ; drug effects ; metabolism ; Membrane Transport Proteins ; genetics ; metabolism ; Ovarian Follicle ; cytology ; drug effects ; Proliferating Cell Nuclear Antigen ; genetics ; metabolism ; RNA, Messenger ; genetics ; Receptors, FSH ; genetics ; metabolism ; Swine