1
|
Milosevic A, Lavrnja I, Savic D, Milosevic K, Skuljec J, Bjelobaba I, Janjic MM. Rat Ovarian Function Is Impaired during Experimental Autoimmune Encephalomyelitis. Cells 2023; 12:cells12071045. [PMID: 37048118 PMCID: PMC10093247 DOI: 10.3390/cells12071045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/21/2023] [Accepted: 03/28/2023] [Indexed: 04/14/2023] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease affecting the CNS and occurring far more prevalently in women than in men. In both MS and its animal models, sex hormones play important immunomodulatory roles. We have previously shown that experimental autoimmune encephalomyelitis (EAE) affects the hypothalamic-pituitary-gonadal axis in rats of both sexes and induces an arrest in the estrous cycle in females. To investigate the gonadal status in female rats with EAE, we explored ovarian morphometric parameters, circulating and intraovarian sex steroid levels, and the expression of steroidogenic machinery components in the ovarian tissue. A prolonged state of diestrus was recorded during the peak of EAE, with maintenance of the corpora lutea, elevated intraovarian progesterone levels, and increased gene and protein expression of StAR, similar to the state of pseudopregnancy. The decrease in CYP17A1 protein expression was followed by a decrease in ovarian testosterone and estradiol levels. On the contrary, serum testosterone levels were slightly increased. With unchanged serum estradiol levels, these results point at extra-gonadal sites of sex steroid biosynthesis and catabolism as important regulators of their circulating levels. Our study suggests alterations in the function of the female reproductive system during central autoimmunity and highlights the bidirectional relationships between hormonal status and EAE.
Collapse
Affiliation(s)
- Ana Milosevic
- Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
| | - Irena Lavrnja
- Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
| | - Danijela Savic
- Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
| | - Katarina Milosevic
- Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
| | - Jelena Skuljec
- Department of Neurology, University Medicine Essen, 45147 Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, 45147 Essen, Germany
| | - Ivana Bjelobaba
- Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
| | - Marija M Janjic
- Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
| |
Collapse
|
2
|
Zhao S, Gong J, Wang Y, Heng N, Wang H, Hu Z, Wang H, Zhang H, Zhu H. Sirtuin 3 regulation: a target to alleviate β-hydroxybutyric acid-induced mitochondrial dysfunction in bovine granulosa cells. J Anim Sci Biotechnol 2023; 14:18. [PMID: 36788581 PMCID: PMC9926763 DOI: 10.1186/s40104-022-00825-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 12/11/2022] [Indexed: 02/16/2023] Open
Abstract
BACKGROUND During the transition period, the insufficient dry matter intake and a sharply increased in energy consumption to produce large quantities of milk, high yielding cows would enter a negative energy balance (NEB) that causes an increase in ketone bodies (KBs) and decrease in reproduction efficiency. The excess concentrations of circulating KBs, represented by β-hydroxybutyric acid (BHBA), could lead to oxidative damage, which potentially cause injury to follicular granulosa cells (fGCs) and delayed follicular development. Sirtuin 3 (Sirt3) regulates mitochondria reactive oxygen species (mitoROS) homeostasis in a beneficial manner; however, the molecular mechanisms underlying its involvement in the BHBA-induced injury of fGCs is poorly understood. The aim of this study was to explore the protection effects and underlying mechanisms of Sirt3 against BHBA overload-induced damage of fGCs. RESULTS Our findings demonstrated that 2.4 mmol/L of BHBA stress increased the levels of mitoROS in bovine fGCs. Further investigations identified the subsequent mitochondrial dysfunction, including an increased abnormal rate of mitochondrial architecture, mitochondrial permeability transition pore (MPTP) opening, reductions in mitochondrial membrane potential (MMP) and Ca2+ release; these dysfunctions then triggered the caspase cascade reaction of apoptosis in fGCs. Notably, the overexpression of Sirt3 prior to treatment enhanced mitochondrial autophagy by increasing the expression levels of Beclin-1, thus preventing BHBA-induced mitochondrial oxidative stress and mitochondrial dysfunction in fGCs. Furthermore, our data suggested that the AMPK-mTOR-Beclin-1 pathway may be involved in the protective mechanism of Sirt3 against cellular injury triggered by BHBA stimulation. CONCLUSIONS These findings indicate that Sirt3 protects fGCs from BHBA-triggered injury by enhancing autophagy, attenuating oxidative stress and mitochondrial damage. This study provides new strategies to mitigate the fGCs injury caused by excessive BHBA stress in dairy cows with ketosis.
Collapse
Affiliation(s)
- Shanjiang Zhao
- grid.410727.70000 0001 0526 1937State Key Laboratory of Animal Nutrition, Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jianfei Gong
- grid.410727.70000 0001 0526 1937State Key Laboratory of Animal Nutrition, Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yi Wang
- grid.410727.70000 0001 0526 1937State Key Laboratory of Animal Nutrition, Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Nuo Heng
- grid.410727.70000 0001 0526 1937State Key Laboratory of Animal Nutrition, Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Huan Wang
- grid.410727.70000 0001 0526 1937State Key Laboratory of Animal Nutrition, Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Zhihui Hu
- grid.410727.70000 0001 0526 1937State Key Laboratory of Animal Nutrition, Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Haoyu Wang
- grid.410727.70000 0001 0526 1937State Key Laboratory of Animal Nutrition, Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Haobo Zhang
- grid.410727.70000 0001 0526 1937State Key Laboratory of Animal Nutrition, Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Huabin Zhu
- State Key Laboratory of Animal Nutrition, Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China.
| |
Collapse
|
3
|
Shi J, Liu C, Chen M, Yan J, Wang C, Zuo Z, He C. The interference effects of bisphenol A on the synthesis of steroid hormones in human ovarian granulosa cells. ENVIRONMENTAL TOXICOLOGY 2021; 36:665-674. [PMID: 33258555 DOI: 10.1002/tox.23070] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 11/20/2020] [Indexed: 06/12/2023]
Abstract
Numerous studies have shown that endocrine-disrupting chemicals are one of the important pathogenic factors in women with polycystic ovary syndrome. Our previous study has revealed that bisphenol A (BPA) can cause steroid hormone imbalance, polycystic ovary, and estrus cycle disorder. In this study, we aimed to explore the effect of BPA, a typical environmental estrogen, on the synthesis of steroid hormones in human ovarian granulosa KGN cells. Exposure of KGN cells to BPA (0.5, 5, 50, and 500 μg/L) resulted in the decrease of progesterone (P), estradiol (E2), and the ratio of estradiol to testosterone (E2/T). BPA affected the expression of genes related to steroid hormone synthesis in KGN cells, including the decreased expression of the steroidogenic acute regulatory protein, ferredoxin, and ferredoxin reductase genes during progesterone synthesis; upregulating the expression of cytochrome p450 oxidoreductase gene associated with E2 and T synthesis; and the downregulated cytochrome P450 family 1 subfamily A member 1 and cytochrome P450 family 1 subfamily B member 1 in E2 degradation. BPA also reduced the expression of stimulatory G proteins (GS) in follicle-stimulating hormone receptor (FSHR)/GS/adenylate cyclase (AC) signaling pathway. In summary, our research has demonstrated that environment-relevant level of BPA exposure leads to steroid hormone synthesis disorder in human ovarian granulosa cells, which might cause the reduction of gene expression in hormone synthesis and the suppression of the FSHR/GS/AC signaling pathway.
Collapse
Affiliation(s)
- Junxia Shi
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Chenyu Liu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Mingyue Chen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Jinhui Yan
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Chonggang Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- State Key Laboratory of Marine Environmental Science, Xiamen University, Xiamen, Fujian, China
| | - Zhenghong Zuo
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- State Key Laboratory of Marine Environmental Science, Xiamen University, Xiamen, Fujian, China
| | - Chengyong He
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
4
|
Bakhshalizadeh S, Amidi F, Shirazi R, Shabani Nashtaei M. Vitamin D3 regulates steroidogenesis in granulosa cells through AMP-activated protein kinase (AMPK) activation in a mouse model of polycystic ovary syndrome. Cell Biochem Funct 2018; 36:183-193. [PMID: 29676471 DOI: 10.1002/cbf.3330] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Revised: 02/20/2018] [Accepted: 03/04/2018] [Indexed: 01/11/2023]
Abstract
Polycystic ovary syndrome (PCOS) is an endocrine and metabolic disorder in reproductive-aged women. Hormonal abnormality caused by steroidogenesis disturbances appears to be the main culprit of the clinical picture in PCOS. Vitamin D3 could regulate steroidogenesis in granulosa cells, but the mechanism of action of vitamin D3 on steroidogenesis remains unknown. AMP-activated protein kinase (AMPK) has a modulating role in steroid hormone production. We investigated the effect of vitamin D3 on steroidogenesis in cultured granulosa cells of dehydroepiandrosterone-induced PCOS mice and studied the involvement of AMPK signalling pathway in the current process. Immunoblotting assay showed that vitamin D3 could increase phosphorylation of AMPK alpha and acetyl-CoA carboxylase, main substrate of AMPK. Vitamin D3 and 5-aminoimidazole-4-carboxamide-1-β-D-riboside or Aicar (AMPK activator) not only reduced gene expression of steroidogenic enzymes (P450scc or Cyp11a1, StAR, Cyp19a1 and 3B-HSD), but also reduced production of progesterone and 17B-estradiol assessed by radioimmunoassay. Pretreatment with compound C (AMPK inhibitor) decreased APMK phosphorylation and eliminated the effects of vitamin D3 and Aicar on steroidogenic enzymes expression and estradiol and progesterone production. This study showed that vitamin D3 has the main role in regulating of steroidogenesis in granulosa cells of mouse polycystic ovary through activation of the AMPK signalling pathway. SIGNIFICANCE OF THE STUDY Polycystic ovarian syndrome (PCOS) is an endocrine disorder of women in reproductive age. This disorder is partly related to disruption in steroidogenesis pathway and dysregulation of estradiol and progesterone production in granulosa cells of polycystic ovaries. Previously, we have shown that vitamin D3 could modulate steroidogenesis pathway in PCOS granulosa cells. In this study, we investigate the molecular mechanism of vitamin D3 in regulation of steroidogenesis pathway. We have shown that vitamin D3 has a modulating role in steroidogenesis pathway of granulosa cells by regulation of AMP-activated protein kinase (AMPK) as an underlying molecular mechanism in mouse polycystic ovary.
Collapse
Affiliation(s)
- Shabnam Bakhshalizadeh
- Department of Anatomical Sciences, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Fardin Amidi
- Department of Anatomy, School of medicine, Tehran University of Medical Sciences, Tehran, Iran.,Department of Infertility, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Shirazi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Anatomical Sciences, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Shabani Nashtaei
- Department of Anatomy, School of medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Bird AD, Greatorex S, Reser D, Lavery GG, Cole TJ. Hydroxysteroid dehydrogenase HSD1L is localised to the pituitary-gonadal axis of primates. Endocr Connect 2017; 6:489-499. [PMID: 28871060 PMCID: PMC5592779 DOI: 10.1530/ec-17-0119] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 08/07/2017] [Indexed: 01/03/2023]
Abstract
Steroid hormones play clinically important and specific regulatory roles in the development, growth, metabolism, reproduction and brain function in human. The type 1 and 2 11-beta hydroxysteroid dehydrogenase enzymes (11β-HSD1 and 2) have key roles in the pre-receptor modification of glucocorticoids allowing aldosterone regulation of blood pressure, control of systemic fluid and electrolyte homeostasis and modulation of integrated metabolism and brain function. Although the activity and function of 11β-HSDs is thought to be understood, there exists an open reading frame for a distinct 11βHSD-like gene; HSD11B1L, which is present in human, non-human primate, sheep, pig and many other higher organisms, whereas an orthologue is absent in the genomes of mouse, rat and rabbit. We have now characterised this novel HSD11B1L gene as encoded by 9 exons and analysis of EST library transcripts indicated the use of two alternate ATG start sites in exons 2 and 3, and alternate splicing in exon 9. Relatively strong HSD11B1L gene expression was detected in human, non-human primate and sheep tissue samples from the brain, ovary and testis. Analysis in non-human primates and sheep by immunohistochemistry localised HSD11B1L protein to the cytoplasm of ovarian granulosa cells, testis Leydig cells, and gonadatroph cells in the anterior pituitary. Intracellular localisation analysis in transfected human HEK293 cells showed HSD1L protein within the endoplasmic reticulum and sequence analysis suggests that similar to 11βHSD1 it is membrane bound. The endogenous substrate of this third HSD enzyme remains elusive with localisation and expression data suggesting a reproductive hormone as a likely substrate.
Collapse
Affiliation(s)
- A Daniel Bird
- Department of Biochemistry and Molecular BiologyMonash University, Melbourne, Victoria, Australia
| | - Spencer Greatorex
- Department of Biochemistry and Molecular BiologyMonash University, Melbourne, Victoria, Australia
| | - David Reser
- Department of PhysiologyMonash University, Melbourne, Victoria, Australia
| | - Gareth G Lavery
- Institute of Metabolism and Systems Research2nd Floor IBR Tower, University of Birmingham, Birmingham, UK
- Centre for EndocrinologyDiabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
| | - Timothy J Cole
- Department of Biochemistry and Molecular BiologyMonash University, Melbourne, Victoria, Australia
| |
Collapse
|
6
|
Johnson A, Lee J. Granulosa cell responsiveness to follicle stimulating hormone during early growth of hen ovarian follicles. Poult Sci 2016; 95:108-14. [DOI: 10.3382/ps/pev318] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2015] [Indexed: 11/20/2022] Open
|
7
|
Effect of Anti-Müllerian hormone (AMH) and bone morphogenetic protein 15 (BMP-15) on steroidogenesis in primary-cultured human luteinizing granulosa cells through Smad5 signalling. J Assist Reprod Genet 2015; 32:1079-88. [PMID: 26003656 DOI: 10.1007/s10815-015-0494-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 05/04/2015] [Indexed: 12/18/2022] Open
Abstract
PURPOSE To determine if there is any effect of AMH and BMP-15 on estradiol and progesterone production from primary-cultured human luteinizing granulosa cells, to delineate what is the effect of FSH on their actions and which are the possible mechanisms involved. METHODS Luteinizing granulosa cells (GCs), obtained from follicular fluid of 30 women undergoing in vitro fertilization, were cultured, after a short 24-h preincubation period, in serum-free medium for 24 or/and 48 h in the presence/absence of various concentrations of AMH, BMP-15 and FSH alone or in combinations. Estradiol and progesterone production, SMAD5 phosphorylation and StAR expression were studied in parallel. Steroids were measured in culture-supernatant using enzyme-immunoassays, while Smad5-signaling pathway activation and StAR protein expression were assessed immunocytochemically. RESULT(S) We found that the treatment of AMH in GCs for 24/48 h attenuated FSH-induced estradiol production (p < 0.001), had no effect on basal estradiol levels, decreased basal progesterone production (p < 0.001) and FSH-induced StAR expression (p < 0.001). On the other hand, BMP-15 decreased basal estradiol levels (p < 0.001) and attenuated FSH-induced estradiol production (p < 0.001). Furthermore, BMP-15 reduced progesterone basal secretion (p < 0.001), an effect that was partially reversed by FSH (p < 0.01), probably via increasing StAR expression (p < 0.001). FSH-induced StAR expression was also attenuated by BMP-15 (p < 0.001). FSH, AMH and BMP-15 activated Smad-signaling pathway, as confirmed by the increase of phospo-Smad5 protein levels (p < 0.001 compared to control). CONCLUSION(S) AMH and BMP-15 by interacting with FSH affect the production of estradiol and progesterone from cultured luteinizing-granulosa cells possibly via Smad5-protein phosphorylation.
Collapse
|
8
|
Casarini L, Moriondo V, Marino M, Adversi F, Capodanno F, Grisolia C, La Marca A, La Sala GB, Simoni M. FSHR polymorphism p.N680S mediates different responses to FSH in vitro. Mol Cell Endocrinol 2014; 393:83-91. [PMID: 24970684 DOI: 10.1016/j.mce.2014.06.013] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 06/16/2014] [Accepted: 06/16/2014] [Indexed: 02/07/2023]
Abstract
The single nucleotide polymorphism p.N680S of the follicle-stimulating hormone (FSH) receptor (FSHR) is a discrete marker of ovarian response but previous in vitro studies failed to demonstrate differences in the response to FSH between N and S carrier cells. Here we demonstrate that p.N680S mediates different kinetics of the response to FSH in vitro. Intracellular cAMP production is faster in p.N680S N than in S homozygous human granulosa cells (45 versus 90 min to achieve the plateau, respectively; Mann-Whitney's U-test; p < 0.005; n = 4). Reflecting the cAMP kinetics, phospho-ERK1/2 and -CREB activation, AREG and STARD1 gene expressions and progesterone production were qualitatively and quantitatively different in N versus S homozygous cells. Finally, the blockade of ERK pathway by U0126 abolishes the genotype-mediated different effects on gene expression and progesterone production (Mann-Whitney's U-test; p ≥ 0.005; n = 3).
Collapse
Affiliation(s)
- Livio Casarini
- Unit of Endocrinology, Dept. of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy; Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy.
| | - Valeria Moriondo
- Unit of Endocrinology, Dept. of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy; Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy
| | - Marco Marino
- Unit of Endocrinology, Dept. of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy; Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy
| | - Francesca Adversi
- Unit of Obstetrics and Gynecology, IRCCS-Arcispedale Santa Maria Nuova, Reggio Emilia, Italy
| | - Francesco Capodanno
- Unit of Obstetrics and Gynecology, IRCCS-Arcispedale Santa Maria Nuova, Reggio Emilia, Italy
| | - Chiarina Grisolia
- Dept. of Laboratory Medicine and Pathology, Laboratory of Chemical-Clinical Analysis, University Hospital of Modena, Modena, Italy
| | - Antonio La Marca
- Dept. of Medical and Mother-Infant and Adult Surgical Sciences, Unit of Obstetrics and Gynecology, University Hospital of Modena, Modena, Italy
| | - Giovanni Battista La Sala
- Unit of Obstetrics and Gynecology, IRCCS-Arcispedale Santa Maria Nuova, Reggio Emilia, Italy; Dept. of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Reggio Emilia, Italy
| | - Manuela Simoni
- Unit of Endocrinology, Dept. of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy; Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy; Azienda USL, Modena, Italy
| |
Collapse
|
9
|
Abarikwu SO, Pant AB, Farombi EO. The protective effects of quercetin on the cytotoxicity of atrazine on rat Sertoli-germ cell co-culture. ACTA ACUST UNITED AC 2012; 35:590-600. [PMID: 22372587 DOI: 10.1111/j.1365-2605.2011.01239.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
To evaluate the direct effect of atrazine (ATZ) and the protective effect of quercetin (QT) on testicular cells, we used primary cultures of rat Sertoli-germ cells (SGCs). ATZ (232 μm) up-regulated the mRNA expression of GATA-4, androgen receptor (AR), androgen-binding protein (ABP), steroidogenic acute regulatory protein (StAR), cytochrome P450 side-chain cleavage enzyme (CYP11A1), cyclooxygenase-2 (COX-2) and NF-κappaB (NF-κB) and down-regulated the expression of stem cell factor (SCF) mRNA. There was no change on the mRNA expression of oestrogen receptor-alpha (ER-α). Simultaneous supplementation of QT in the culture normalizes the expression of these genes. The stimulatory action of follicle stimulating hormone (10 ng/mL) on ATZ-induced StAR and CYP11A1 mRNA levels were also prevented by QT. Furthermore, ATZ-stimulatory action on AR mRNA was opposed in a dose-dependent manner in the presence of increasing concentrations of QT (10-50 μm).The dislodgement of germ cells from the Sertoli cells monolayer and decrease in SGCs viability was prevented by QT. To show whether or not the disrupted interactions of Sertoli and germ cells impaired spermatogenesis, adult male rats exposed in vivo to ATZ (50 mg/kg b.wt) for 1 week had their daily spermatozoa production (DSP) per gram testis lowered by 30%. DSP was significantly increased in the QT(10 mg/kg) + ATZ-treated rats as compared with the ATZ-treated rats. Taken together, ATZ can alter SGCs expression of spermatogenesis- and steroiodogenesis-related genes resulting in a decrease in sperm production in the testis as well as cell viability. QT might block these molecular events-induced by ATZ thereby protecting testicular Sertoli-germ cells from ATZ-induced toxicity.
Collapse
Affiliation(s)
- S O Abarikwu
- Department of Chemical Sciences, College of Natural Sciences, Redeemer's University, Redemption City, Ogun State, Nigeria.
| | | | | |
Collapse
|
10
|
Perlman S, Bouquin T, van den Hazel B, Jensen TH, Schambye HT, Knudsen S, Okkels JS. Transcriptome analysis of FSH and FSH variant stimulation in granulosa cells from IVM patients reveals novel regulated genes. ACTA ACUST UNITED AC 2006; 12:135-44. [PMID: 16556681 DOI: 10.1093/molehr/gah247] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
FSH is crucial for oocyte maturation and fertility and is the main component in infertility treatment in assisted reproduction. The granulosa cells expressing the FSH receptor interact with the oocyte and provide nourishing substrates controlling the oocyte maturation. Thus, transcriptome analysis of granulosa cells stimulated by FSH is of major importance in understanding the communication between oocytes and granulosa cells. In this study, gene expression profiles were assessed in human granulosa cells from normal cycling in vitro maturation (IVM) patients using oligonucleotide gene chips. Granulosa cells were stimulated for 2 h with either FSH or a previously generated glycosylated FSH variant (FSH1208) that exhibited increased in vivo activity because of prolonged half-life. The analysis identified 74 significantly FSH/FSH1208 regulated genes. Amongst these were well known FSH regulated genes as well as genes not previously described to be important in the FSH signalling pathway. These novel FSH regulated genes include transcription factors [cAMP responsive element modulator (CREM)/inducible cAMP early repressors (ICER), GATA 6, ZFN 361, Bcl11a, CITED1 and TCF 8] and other regulatory proteins and enzymes (IGF-BP3, syntaxin and PCK1) possibly important for oocyte/granulosa cell interaction and function. Array data were validated for 13 genes by northern blots or RT-PCR. Furthermore, no significant differences in gene regulation were detected between the two FSH analogs. This work uncovers novel data important for understanding the folliculogenesis. Furthermore, the results suggest that FSH1208 has a gene expression profile like FSH and thus, in the light of known prolonged in vivo activity, might be a candidate for improved infertility treatment.
Collapse
Affiliation(s)
- S Perlman
- Maxygen, Hørsholm, Technical University of Denmark, Lyngby
| | | | | | | | | | | | | |
Collapse
|
11
|
Myllymäki SA, Karjalainen M, Haavisto TE, Toppari J, Paranko J. Infantile 4-tert-octylphenol exposure transiently inhibits rat ovarian steroidogenesis and steroidogenic acute regulatory protein (StAR) expression. Toxicol Appl Pharmacol 2005; 207:59-68. [PMID: 16054900 DOI: 10.1016/j.taap.2004.12.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2004] [Revised: 11/29/2004] [Accepted: 12/01/2004] [Indexed: 11/24/2022]
Abstract
Phenolic compounds, such as 4-tert-octylphenol (OP), have been shown to interfere with rat ovarian steroidogenesis. However, little is known about steroidogenic effects of infantile OP exposure on immature ovary. The aim of the present study was to investigate the effects of infantile OP exposure on plasma FSH, LH, estradiol, and progesterone levels in 14-day-old female rats. The effect on ovarian steroidogenic acute regulatory protein (StAR) and FSH receptor (FSHr) expression was analyzed by Western blotting. Ex vivo analysis was carried out for follicular estradiol, progesterone, testosterone, and cAMP production. Sprague-Dawley rats were given OP (0, 10, 50, or 100 mg/kg) subcutaneously on postnatal days 6, 8, 10, and 12. On postnatal day 14, plasma FSH was decreased and progesterone increased significantly at a dose of 100 mg OP/kg. In addition, the highest OP dose advanced the time of vaginal opening in puberty. OP had no effect on infantile LH and estradiol levels or ovarian FSHr content. Ovarian StAR protein content and ex vivo hormone and cAMP production were decreased at all OP doses compared to controls. However, hormone levels recovered independent on FSH and even increased above the control level during a prolonged culture. On postnatal day 35, no statistically significant differences were seen between control and OP-exposed animals in plasma FSH, LH, estradiol, and progesterone levels, or in ovarian StAR protein content. The results indicate that the effect of OP on the infantile ovary is reversible, while more permanent effects in the hypothalamus and pituitary, as described earlier, are involved in the reduction of circulating FSH levels and premature vaginal opening.
Collapse
Affiliation(s)
- S A Myllymäki
- Department of Biology, Laboratory of Animal Physiology, 20014 University of Turku, Finland.
| | | | | | | | | |
Collapse
|
12
|
Rusovici R, Hui YY, Lavoie HA. Epidermal growth factor-mediated inhibition of follicle-stimulating hormone-stimulated StAR gene expression in porcine granulosa cells is associated with reduced histone H3 acetylation. Biol Reprod 2004; 72:862-71. [PMID: 15590903 DOI: 10.1095/biolreprod.104.034298] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Steroidogenic acute regulatory protein (StAR) mediates cholesterol transport into the mitochondria and is essential for ovarian steroidogenesis. Epidermal growth factor (EGF) has been reported to inhibit FSH-stimulated differentiation in porcine granulosa cells. Previous studies have demonstrated FSH stimulates StAR mRNA accumulation and gene promoter activation in granulosa cells. Treatment of granulosa cells with FSH (5 ng/ml, 6 h) increased StAR mRNA, whereas coaddition of EGF (10 ng/ ml) significantly reduced (P < 0.05) FSH-stimulated mRNA accumulation by 62.7% +/- 13.9%. Under these same conditions, FSH-stimulated cAMP accumulation in cultures was unaltered by coincubation with EGF. RNA stability studies showed that cotreatment with FSH and EGF did not alter the StAR mRNA half-life compared with FSH alone, t(1/2) = 1.9-3.8 and 2.7-4.1 h, respectively. EGF significantly inhibited (P < 0.05) FSH-stimulated StAR heterogeneous nuclear RNA levels by 47.6% +/- 6.8 %, implicating a repressive effect on transcription. Surprisingly, EGF (1-50 ng/ml) did not affect FSH stimulation of a 1423-base pair StAR gene promoter-luciferase construct in transient transfection assays in porcine granulosa cells. To evaluate FSH and EGF effects on the endogenous StAR gene, chromatin immunoprecipitation assays were performed in combination with real-time polymerase chain reaction. FSH increased histone H3 acetylation (lysines 9, 14) within the proximal region of the StAR gene promoter and coincubation with EGF blocked this effect. Dimethylation (lysine 9) of histone H3 was not influenced by treatments. In conclusion, EGF repression of FSH-stimulated StAR transcription in porcine granulosa cells is accompanied by reductions in histone H3 acetylation associated with the StAR gene promoter.
Collapse
Affiliation(s)
- Raluca Rusovici
- Department of Cell and Developmental Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina 29208, USA
| | | | | |
Collapse
|
13
|
Johnson AL, Bridgham JT, Woods DC. Cellular Mechanisms and Modulation of Activin A- and Transforming Growth Factor β-Mediated Differentiation in Cultured Hen Granulosa Cells1. Biol Reprod 2004; 71:1844-51. [PMID: 15269104 DOI: 10.1095/biolreprod.104.032573] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Undifferentiated granulosa cells from prehierarchal (6- to 8-mm-diameter) hen follicles express very low to undetectable levels of LH receptor (LH-R) mRNA, P450 cholesterol side chain cleavage (P450scc) enzyme activity, and steroidogenic acute regulatory (StAR) protein, and produce negligible progesterone, in vitro, following an acute (3-h) challenge with either FSH or LH. It has previously been established that culturing such cells with FSH for 18-20 h induces LH-R, P450scc, and StAR expression, which enables the initiation of progesterone production. The present studies were conducted to characterize the ability of activin and transforming growth factor (TGF) beta, both alone and in combination with FSH, to promote hen granulosa cell differentiation, in vitro. A 20-h culture of prehierarchal follicle granulosa cells with activin A or transforming growth factor beta (TGFbeta)1 increased LH-R mRNA levels compared with control cultured cells. Activin A and TGFbeta1 also promoted FSH-receptor (FSH-R) mRNA expression when combined with FSH treatment. Neither activin A nor TGFbeta1 alone stimulated progesterone production after 20 h culture. However, preculture with either factor for 20 h (to induce gonadotropin receptor mRNA expression) followed by a 3-h challenge with FSH or LH potentiated StAR expression and progesterone production compared with cells challenged with gonadotropin in the absence of activin A or TGFbeta1 preculture. Significantly, activation of the mitogen-activated protein (MAP) kinase pathway with transforming growth factor alpha (TGFalpha) (monitored by Erk phosphorylation) blocked TGFbeta1-induced LH-R expression, and this effect was associated with the inhibition of Smad2 phosphorylation. We conclude that a primary differentiation-inducing action of activin A and TGFbeta1 on hen granulosa cells from prehierarchal follicles is directed toward LH-R expression. Enhanced LH-R levels subsequently sensitize granulosa cells to LH, which in turn promotes StAR plus P450scc expression and subsequently an increase in P4 production. Significantly, the finding that TGFbeta signaling is negatively regulated by MAP kinase signaling is proposed to represent a mechanism that prevents premature differentiation of granulosa cells.
Collapse
Affiliation(s)
- A L Johnson
- Department of Biological Sciences and Walther Cancer Center, The University of Notre Dame, Notre Dame, Indiana 46556, USA.
| | | | | |
Collapse
|
14
|
Ke FC, Chuang LC, Lee MT, Chen YJ, Lin SW, Wang PS, Stocco DM, Hwang JJ. The Modulatory Role of Transforming Growth Factor β1 and Androstenedione on Follicle-Stimulating Hormone-Induced Gelatinase Secretion and Steroidogenesis in Rat Granulosa Cells1. Biol Reprod 2004; 70:1292-8. [PMID: 14711789 DOI: 10.1095/biolreprod.103.023531] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
To investigate the potential roles of matrix metalloproteinases (MMPs) in ovarian granulosa cell differentiation, we studied the interactive effects of FSH and local ovarian factors, transforming growth factor beta1 (TGFbeta1) and androstenedione, on gelatinase secretion and progesterone production in rat ovarian granulosa cells. Granulosa cells of eCG-primed immature rats were treated once with various doses of FSH and TGFbeta1 and androstenedione alone or in combinations for 2 days. Conditioned media were analyzed for gelatinase activity using gelatin-zymography/densitometry and progesterone levels using enzyme immunoassay. Cell lysates were analyzed for steroidogenic acute regulatory (StAR) and cholesterol side-chain-cleavage (P450scc) enzyme protein levels. This study demonstrates for the first time that FSH dose-dependently increased the secretion of a major 63-kDa gelatinase and minor 92- and 67-kDa gelatinases. TGFbeta1 also dose-dependently increased the secretion of 63-kDa gelatinase, while androstenedione alone had no effect. The 92-kDa gelatinase was identified as the pro-MMP9 that could be cleaved by aminophenylmercuric acetate into the 83-kDa active form. Importantly, we show that TGFbeta1 and androgen act in an additive manner to enhance FSH stimulatory effects both on the secretion of gelatinases and the production of progesterone. We further show by immunoblotting that the enhancing effect of TGFbeta1 and androstenedione on FSH-stimulated steroidogenesis is partly mediated through the increased level of StAR protein and/or P450scc enzyme. In conclusion, this study indicates that, during antral follicle development, TGFbeta1 and androgen act to enhance FSH promotion of granulosa cell differentiation and that the process may involve the interplay of modulating cell- to-matrix/cell-to-cell interaction and steroidogenic activity.
Collapse
Affiliation(s)
- Ferng-Chun Ke
- Institute of Molecular and Cellular Biology, National Taiwan University, Taipei 106, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Tsuchiya M, Inoue K, Matsuda H, Nakamura K, Mizutani T, Miyamoto K, Minegishi T. Expression of steroidogenic acute regulatory protein (StAR) and LH receptor in MA-10 cells. Life Sci 2003; 73:2855-63. [PMID: 14511770 DOI: 10.1016/s0024-3205(03)00698-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
LH stimulation is mediated by its own receptor at the first step of the cascade, after which intracellular cAMP increases to stimulate the transcription of steroidogenic acute regulatory protein (StAR) in mouse MA-10 Leydig tumor cells. StAR mediates the rate-limiting step of steroidogenesis, which is the transfer of cholesterol to the inner mitochondrial membrane. Northern blot analysis consistently revealed two major transcripts, of about 3.6 kb and 1.6 kb, that hybridized with rat StAR mRNA. In this culture, treatment with hCG led StAR mRNA levels to rapidly and strongly increase by 3 h. Parallel increases were observed in transcripts of both sizes. Compared to StAR mRNA expression, LH receptor mRNA levels gradually decreased and declined to 50% of control values between 6 and 12 h incubation. Compared to the control, StAR mRNA levels increased and LH receptor mRNA levels decreased in a dose-dependent manner in the presence of increasing concentrations of hCG (3-100 ng/ml) and of increasing concentrations of 8-Br-cAMP (0.2-2 mM) after 4 h incubation. Since the over production of steroid hormones might be toxic to the own cells, the LH signal transduction that stimulates steroidogenesis might concomitantly decrease the responsiveness of steroidogenesis to LH stimulation at the receptor level. This result should be further investigated to clarify the mechanism of LH receptor regulation and steroidogenesis.
Collapse
Affiliation(s)
- Megumi Tsuchiya
- Department of Obstetrics and Gynecology, School of Medicine, Gunma University, Maebashi, Gunma 371-8511, Japan
| | | | | | | | | | | | | |
Collapse
|
16
|
Mizutani T, Yamada K, Yazawa T, Okada T, Minegishi T, Miyamoto K. Cloning and characterization of gonadotropin-inducible ovarian transcription factors (GIOT1 and -2) that are novel members of the (Cys)(2)-(His)(2)-type zinc finger protein family. Mol Endocrinol 2001; 15:1693-705. [PMID: 11579202 DOI: 10.1210/mend.15.10.0718] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Gonadotropins are essential for ovarian follicular development and differentiation. To identify genes that are rapidly induced by gonadotropin in the immature rat ovary, ovarian genes were screened by a subtraction cloning procedure. cDNA clones encoding novel members of the (Cys)(2)-(His)(2)-type zinc finger protein family GIOT1 and -2 (gonadotropin-inducible transcription factor 1 and 2), were identified. Two isoforms of GIOT2 (GIOT2 alpha and 2 beta), which are probably produced by alternative splicing, also exist. Nucleotide sequence analysis revealed that GIOT1, but not GIOT2, contains the krüppel-associated box-A domain at the NH(2) terminus. RNA analyses revealed that these mRNAs were rapidly and temporarily induced by gonadotropins in the rat testis as well as in the ovary. In situ hybridization study revealed that expression of GIOT1 was induced in theca interna cells in the ovary and Leydig cells in the testis. Interestingly, the gene expression of GIOT1 is restricted to the pituitary, adrenal, testis, and ovary, while GIOT2 gene is expressed ubiquitously. A functional analysis of GIOT1 and -2 by a GAL4-based mammalian one-hybrid system revealed that GIOT1, but not GIOT2, is a transcriptional repressor and that the krüppel-associated box-A domain of GIOT1 is responsible for the transcriptional repressor activity. A GAL4-based yeast two-hybrid system was also used to identify proteins that interact with the rat GIOT1. We cloned genes encoding rat homologs of human I-mfa domain containing protein and transcriptional intermediary factor 1 beta, both of which are transcription-regulatory proteins. Interaction of these proteins with GIOT1 was directly demonstrated by GST pull-down assay. Our data strongly suggest that GIOT1 may function as a novel transcriptional repressor by working with rat homologs of human I-mfa domain containing protein and transcriptional intermediary factor 1 beta proteins and may play a significant role at the transcription level in the folliculogenesis.
Collapse
Affiliation(s)
- T Mizutani
- Department of Biochemistry, Fukui Medical University, Matsuoka, Fukui 910-1193, Japan
| | | | | | | | | | | |
Collapse
|