1
|
Chen Y, Bai X, Zhang Y, Zhao Y, Ma H, Yang Y, Wang M, Guo Y, Li X, Wu T, Zhang Y, Kong H, Zhao Y, Qu H. Zingiberis rhizoma-based carbon dots alter serum oestradiol and follicle-stimulating hormone levels in female mice. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2024; 52:12-22. [PMID: 37994799 DOI: 10.1080/21691401.2023.2276770] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 10/18/2023] [Indexed: 11/24/2023]
Abstract
Chinese herbs contain substances that regulate female hormones. Our study confirmed that Zingiberis rhizoma carbonisata contains Zingiberis rhizoma-based carbon dots (ZR-CDs), which exert regulatory effects on serum oestradiol and FSH in mice and show impacts on endometrial growth and follicular development that potentially affect the ability of female fertility. ZR-CDs were characterized to clarify the microstructure, optical features, and functional group characteristics. It shows that ZR-CDs are spherical carbon nanostructures ranging from 0.97 to 2.3 nm in diameter, with fluorescent properties and a surface rich in functional groups. We further investigated the impact of ZR-CDs on oestradiol and FSH in serum, growth, and the development of ovarian and uterine using normal female mice and exogenous oestradiol intervention model. It was observed that ZR-CDs accelerated oestrogen metabolism and attenuated oestradiol-induced endometrial hyperplasia. Simultaneously, ZR-CDs triggered an increase in FSH, even in the presence of high-serum oestradiol that inhibits FSH secretion. Our findings suggest that ZR-CDs could be a potential therapeutic treatment for anovulatory menstruation.
Collapse
Affiliation(s)
- Yumin Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xue Bai
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ying Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yafang Zhao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Huagen Ma
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yunbo Yang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Meijun Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yinghui Guo
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaopeng Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Tong Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yue Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Hui Kong
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yan Zhao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Huaihua Qu
- Centre of Scientific Experiment, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
2
|
Zhu L, Zhou X, Ma L, Hu Y. Effect of TSH on aromatase expression of ovarian granulosa cells in obese mice. Hormones (Athens) 2024; 23:821-829. [PMID: 38872063 DOI: 10.1007/s42000-024-00571-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 05/24/2024] [Indexed: 06/15/2024]
Abstract
PURPOSE Aromatase plays an important role in ovarian development, the normal progress of the menstrual cycle, and fertility status. Elevated aromatase activity is linked to obesity. There is a bidirectional relationship between obesity and thyroid function. Few studies have investigated the relationship between TSH and ovarian aromatase in obesity. Our aim was to investigate the effect of TSH on aromatase expression of ovarian granulosa cells in obese mice. METHODS Female mice pups were divided into an obesity group and a control group. Obese parameters and the time of pubertal onset were recorded. At the age of 5 weeks, blood and tissues were obtained. Serum aromatase and hormone concentrations were measured using ELISA. The granulosa cells were isolated and exposed to variable concentrations (0 μM, 1 μM, 10 μM, 100 μM) of TSH. The expression of CYP19A1 mRNA and protein were assessed via RT-qPCR and western blot. RESULTS In female mice, body weight, Lee's obesity index, and serum levels of E2, aromatase, and TSH were significantly higher in the obesity group compared to the control group, whereas the time of pubertal onset and serum T3 and T4 concentrations were significantly lower (all P < 0.001). In granulosa cells, the expression of CYP19A1 mRNA in the obesity group was lower than that in the control group at 1 μM and 100 μM concentrations of TSH (both P < 0.001). The expression of CYP19A1 protein in the obesity group was higher than that in the control group after TSH stimulation (P = 0.014, P < 0.001, and P = 0.004, respectively). With the increase of TSH concentrations, the expression of CYP19A1 mRNA and protein in the two groups significantly increased (all P < 0.001). CONCLUSION Early puberty and elevated serum aromatase and TSH levels were found in obese female mice. In the granulosa cells of obese mice, TSH directly regulates aromatase expression in a dose-dependent manner.
Collapse
Affiliation(s)
- Liping Zhu
- Department of Pediatrics, Linyi People's Hospital, Linyi, 276000, Shandong Province, China
| | - Xinhui Zhou
- Department of Pediatrics, Lanling County People's Hospital, Linyi, 277700, Shandong Province, China
| | - Ling Ma
- Department of Pediatrics, Pingyi Hospital of Traditional Chinese Medicine, Linyi, 273300, Shandong Province, China
| | - Yanyan Hu
- Department of Pediatrics, Linyi People's Hospital, Linyi, 276000, Shandong Province, China.
| |
Collapse
|
3
|
Vakili S, Koohpeyma F, Samare-Najaf M, Jahromi BN, Jafarinia M, Samareh A, Hashempur MH. The Effects of L-Tartaric Acid on Ovarian Histostereological and Serum Hormonal Analysis in an Animal Model of Polycystic Ovary Syndrome. Reprod Sci 2024; 31:3583-3594. [PMID: 39333435 DOI: 10.1007/s43032-024-01699-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 09/16/2024] [Indexed: 09/29/2024]
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrine-related reproductive disorder in women of reproductive age, accompanied by both the impairment of female fecundity and a risk of metabolic disorders. PCOS is emphasized as a worldwide concern due to its unknown etiology and lack of specific medications. The current study aimed to evaluate the effects of L-tartaric acid, an abundantly occurring compound in fruits, on the histostereological and hormonal changes caused by PCOS. Forty adult Sprague Dawley rats were randomly divided into four groups including controls (no intervention), Tartaric acid (40mg/Kg/day from day 21 onwards for 39 days), PCOS (21 days letrozole and then normal saline orally for 39 days), and PCOS + Tartaric acid. After treatments, the ovarian histostereological analysis as well as the level of reproductive hormones including luteinizing hormone (LH), follicle-stimulating hormone (FSH), estradiol, progesterone, and testosterone was measured. PCOS caused a significant decrease in the number of unilaminar, multilaminar, antral, and graafian follicles and increased follicular atresia (p-value < 0.001). Moreover, the weight and volume of ovarian tissue and related structures including cortex, medulla, and cysts increased significantly (p-value < 0.0001). However, corpus luteum volume was significantly decreased (p-value < 0.001). Although significant differences were found in some parameters with the control group (p-value < 0.05), the administration of tartaric acid restored the pathological effects of PCOS on the ovarian histostructure. Furthermore, tartaric acid improved the serum levels of LH, estradiol, progesterone, and testosterone (p-value < 0.05). The obtained findings may suggest tartaric acid as a novel strategy for PCOS management, although further studies are necessary.
Collapse
Affiliation(s)
- Sina Vakili
- Infertility Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farhad Koohpeyma
- Endocrinology and Metabolism Research Center, Shiraz University of Medical Science, Shiraz, Iran
| | - Mohammad Samare-Najaf
- 3Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Bahia Namavar Jahromi
- Infertility Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of OB-GYN, Division of Infertility and IVF, Shiraz School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Morteza Jafarinia
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Samareh
- Department of Clinical Biochemistry, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Applied Cellular and Molecular Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Hashem Hashempur
- Research Center for Traditional Medicine and History of Medicine, Department of Persian Medicine, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
4
|
Wadood AA, Xiquan Z. Unraveling the mysteries of chicken proteomics: Insights into follicle development and reproduction. J Proteomics 2024; 308:105281. [PMID: 39154802 DOI: 10.1016/j.jprot.2024.105281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/12/2024] [Accepted: 08/15/2024] [Indexed: 08/20/2024]
Abstract
Chicken proteomics is a valuable method for comprehending the many mechanisms involved in follicle growth and reproduction in birds. This study offers a thorough summary of the latest progress in chicken proteomics research, specifically highlighting the knowledge obtained regarding follicle development and reproductive physiology. Proteomic studies have revealed essential proteins and pathways that play a role in follicle development, including those that control oocyte size, maturation, and ovulation. Proteomic investigations have provided insight into the molecular pathways that govern reproductive processes. By utilizing advanced proteomic technologies, including mass spectrometry and protein microarray analysis, we have been able to identify and measure many proteins in chicken follicles at their different developmental stages. The utilization of proteomic methods has enabled the identification of previously unknown biomarkers for reproductive efficiency that expedited the creation of innovative diagnostic instruments for monitoring reproductive health in chicken. Chicken proteomics not only offers insights into follicle growth and reproduction but also uncovers the effects of environmental influences on reproductive function. This provides new opportunities for exploring the molecular pathways that cause these effects. The integration of current data with upcoming proteomic technologies offers the potential for innovative strategies to enhance chicken reproduction.
Collapse
Affiliation(s)
- Armughan Ahmed Wadood
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou, Guangdong, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou 510642, China
| | - Zhang Xiquan
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou, Guangdong, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
5
|
Shi XK, Peng T, Azimova B, Li XL, Li SS, Cao DY, Fu NJ, Zhang GL, Xiao WL, Wang F. Luteolin and its analog luteolin-7-methylether from Leonurus japonicus Houtt suppress aromatase-mediated estrogen biosynthesis to alleviate polycystic ovary syndrome by the inhibition of tumor progression locus 2. JOURNAL OF ETHNOPHARMACOLOGY 2024; 331:118279. [PMID: 38705425 DOI: 10.1016/j.jep.2024.118279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 04/19/2024] [Accepted: 04/29/2024] [Indexed: 05/07/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Leonurus japonicus Houtt (L. japonicus, Chinese motherwort), known as Yi Mu Cao which means "good for women", has long been widely used in China and other Asian countries to alleviate gynecological disorders, often characterized by estrogen dysregulation. It has been used for the treatment of polycystic ovary syndrome (PCOS), a common endocrine disorder in women but the underlying mechanism remains unknown. AIM OF THE STUDY The present study was designed to investigate the effect and mechanism of flavonoid luteolin and its analog luteolin-7-methylether contained in L. japonicus on aromatase, a rate-limiting enzyme that catalyzes the conversion of androgens to estrogens and a drug target to induce ovulation in PCOS patients. MATERIALS AND METHODS Estrogen biosynthesis in human ovarian granulosa cells was examined using ELISA. Western blots were used to explore the signaling pathways in the regulation of aromatase expression. Transcriptomic analysis was conducted to elucidate the potential mechanisms of action of compounds. Finally, animal models were used to assess the therapeutic potential of these compounds in PCOS. RESULTS Luteolin potently inhibited estrogen biosynthesis in human ovarian granulosa cells stimulated by follicle-stimulating hormone. This effect was achieved by decreasing cAMP response element-binding protein (CREB)-mediated expression of aromatase. Mechanistically, luteolin and luteolin-7-methylether targeted tumor progression locus 2 (TPL2) to suppress mitogen-activated protein kinase 3/6 (MKK3/6)-p38 MAPK-CREB pathway signaling. Transcriptional analysis showed that these compounds regulated the expression of different genes, with the MAPK signaling pathway being the most significantly affected. Furthermore, luteolin and luteolin-7-methylether effectively alleviated the symptoms of PCOS in mice. CONCLUSIONS This study demonstrates a previously unrecognized role of TPL2 in estrogen biosynthesis and suggests that luteolin and luteolin-7-methylether have potential as novel therapeutic agents for the treatment of PCOS. The results provide a foundation for further development of these compounds as effective and safe therapies for women with PCOS.
Collapse
Affiliation(s)
- Xiao-Ke Shi
- Center for Natural Products Research, Chinese Academy of Sciences, Chengdu 610041, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ting Peng
- Center for Natural Products Research, Chinese Academy of Sciences, Chengdu 610041, China; Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Bahtigul Azimova
- Department of Inorganic, Physical and Colloidal Chemistry, Tashkent Pharmaceutical Institute, 45 Aybek Street, 100015, Tashkent, Uzbekistan
| | - Xiao-Li Li
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Provincial Center for Research & Development of Natural Products, School of Pharmacy and School of Chemical Science and Technology, Yunnan University, Kunming, China.
| | - Shan-Shan Li
- Center for Natural Products Research, Chinese Academy of Sciences, Chengdu 610041, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Dong-Yi Cao
- Center for Natural Products Research, Chinese Academy of Sciences, Chengdu 610041, China; University of Chinese Academy of Sciences, Beijing 100049, China; Department of Pharmacy, Kunming Municipal Hospital of Traditional Chinese Medicine, Kunming 650500, China
| | - Nai-Jie Fu
- Center for Natural Products Research, Chinese Academy of Sciences, Chengdu 610041, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guo-Lin Zhang
- Center for Natural Products Research, Chinese Academy of Sciences, Chengdu 610041, China
| | - Wei-Lie Xiao
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Provincial Center for Research & Development of Natural Products, School of Pharmacy and School of Chemical Science and Technology, Yunnan University, Kunming, China
| | - Fei Wang
- Center for Natural Products Research, Chinese Academy of Sciences, Chengdu 610041, China.
| |
Collapse
|
6
|
Zhao R, Ran L, Yao H, He Y, Lu X, Zhu W, Zhang Y, Zhang T, Shi S, Luo Z, Zhang C. Moxibustion ameliorates ovarian function in premature ovarian insufficiency rats by activating cAMP/PKA/CREB to promote steroidogenesis in ovarian granulosa cells. J Steroid Biochem Mol Biol 2024; 242:106547. [PMID: 38754522 DOI: 10.1016/j.jsbmb.2024.106547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 05/05/2024] [Accepted: 05/12/2024] [Indexed: 05/18/2024]
Abstract
Premature ovarian insufficiency (POI) presents a substantial challenge to women's physiological and psychological well-being. Hormone replacement therapy, as the preferred therapeutic approach, involves solely exogenous supplementation of estrogen. Moxibustion, a traditional Chinese external treatment, has been investigated in our previous studies. It not only improves hormone levels and clinical symptoms in POI patients but also safeguards ovarian reserve. This study aims to explore the regulatory mechanisms by which moxibustion modulates hormone levels and restores ovarian function in POI. A POI rat model was established using cyclophosphamide, and moxibustion treatment was applied at acupoints "CV4" and "SP6" for a total of four courses. Subsequently, ovaries from each group were subjected to transcriptome sequencing (Bulk RNA-seq). Target pathways and key genes were selected through enrichment analysis and GSVA scoring, with validation using various techniques including electron microscopy, ELISA, Western blot, and immunohistochemistry. The results demonstrated that moxibustion restored the estrous cycle in POI rats, improved sex hormone levels, reduced the number of atretic follicles, and increased the count of dominant follicles (P<0.05). Bulk RNA-seq analysis revealed that moxibustion downregulated pathways associated with ovarian dysfunction, infertility, and immune responses, upregulated pathways related to follicular development and ovarian steroidogenesis. Furthermore, our data confirmed that moxibustion significantly increased the number of ovarian granulosa cells (GCs) and upregulated the expression of proteins related to steroidogenesis in GCs, including FSHR, P450 arom, cAMP, PKA, and CREB (P<0.05), with no significant effect observed on proteins related to steroidogenesis in theca cells. These outcomes aligned with the RNA-seq results. In conclusion, these findings propose that moxibustion enhances steroidogenesis in GCs through the activation of the cAMP/PKA/CREB pathway, consequently improving impaired ovarian function in POI rats. This study provides robust evidence supporting moxibustion as a targeted intervention for treating POI by specifically regulating steroidogenesis in GCs.
Collapse
Affiliation(s)
- Rui Zhao
- Department of Acupuncture and Moxibustion, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210022, China
| | - Lingxiang Ran
- Department of Acupuncture and Moxibustion, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210022, China; Department of Urology, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Hanyue Yao
- Department of Acupuncture and Moxibustion, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210022, China
| | - Yizhi He
- Department of Acupuncture and Moxibustion, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210022, China
| | - Xinru Lu
- Department of Acupuncture and Moxibustion, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210022, China
| | - Weina Zhu
- Central Laboratory, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210022, China; Department of Biobank, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210022, China
| | - Yajie Zhang
- Central Laboratory, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210022, China; Department of Biobank, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210022, China
| | - Tianyi Zhang
- Department of Acupuncture and Moxibustion, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210022, China
| | - Shijie Shi
- Department of Acupuncture and Moxibustion, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210022, China
| | - Zheng Luo
- Department of Acupuncture and Moxibustion, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210022, China
| | - Cairong Zhang
- Department of Acupuncture and Moxibustion, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210022, China.
| |
Collapse
|
7
|
Maurya S, Tripathi S, Arora T, Singh A. Adropin may regulate ovarian functions by improving antioxidant potential in adult mouse. J Steroid Biochem Mol Biol 2024; 242:106524. [PMID: 38670515 DOI: 10.1016/j.jsbmb.2024.106524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/14/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024]
Abstract
The corpus luteum (CL) is a temporary endocrine gland that synthesizes progesterone. The luteal progesterone plays a central role in the regulation of the estrous cycle as well as the implantation and maintenance of pregnancy. Our previous study showed the expression of adropin and its receptor, GPR19, in the luteal cells and its significant role in luteinization. The aim of the present study was to investigate the in vitro effect of adropin on hCG-induced ovarian functions in adult mice. We also evaluated the effect of exogenous treatment with adropin on ovarian steroidogenesis and anti-oxidant parameters, with special emphasis on CL function. Our results demonstrated that adropin acts synergistically with hCG to promote ovarian steroidogenesis and survival by increasing the expression of StAR, 3β-HSD, and aromatase proteins and decreasing the BAX/BCL2 ratio. Exogenous adropin treatment increased progesterone production by increasing the expression of GPR19, StAR and 3β-HSD enzymes in the mouse ovary. Also, adropin inhibited the luteal oxidative stress by increasing nuclear translocation of NRF-2 in CL, which resulted in increased HO-1 expression and SOD, catalase activity. Decreased oxidative stress might inhibit the translocation of NF-κB into the nucleus of luteal cells, resulting into increased survival and decreased apoptosis, as evident by decreased lipid peroxidation, BAX/BCL2 ratio, caspase 3, active caspase 3 expression, and TUNEL-positive cells in adropin treated mice. Our findings suggest that adropin can be a promising candidate that can enhance the survivability of the CL.
Collapse
Affiliation(s)
- Shweta Maurya
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Shashank Tripathi
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | | | - Ajit Singh
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India.
| |
Collapse
|
8
|
Bérubé M, Abedini A, Lapointe E, Gusscott S, Brind'Amour J, Zamberlam G, Boerboom D. SFRP4 promotes autophagy and blunts FSH responsiveness through inhibition of AKT signaling in ovarian granulosa cells. Cell Commun Signal 2024; 22:396. [PMID: 39138534 PMCID: PMC11323480 DOI: 10.1186/s12964-024-01736-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 07/04/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Secreted frizzled-related proteins (SFRPs) comprise a family of WNT signaling antagonists whose roles in the ovary are poorly understood. Sfrp4-null mice were previously found to be hyperfertile due to an enhanced granulosa cell response to gonadotropins, leading to decreased antral follicle atresia and enhanced ovulation rates. The present study aimed to elucidate the mechanisms whereby SFRP4 antagonizes FSH action. METHODS Primary cultures of granulosa cells from wild-type mice were treated with FSH and/or SFRP4, and effects of treatment on gene expression were evaluated by RT-qPCR and RNAseq. Bioinformatic analyses were conducted to analyse the effects of SFRP4 on the transcriptome, and compare them to those of FSH or a constitutively active mutant of FOXO1. Additional granulosa cell cultures from wild-type or Sfrp4-null mice, some pretreated with pharmacologic inhibitors of specific signaling effectors, were used to examine the effects of FSH and/or SFRP4 on signaling pathways, autophagy and apoptosis by western blotting and TUNEL. RESULTS Treatment of cultured granulosa cells with recombinant SFRP4 was found to decrease basal and FSH-stimulated mRNA levels of FSH target genes. Unexpectedly, this effect was found to occur neither via a canonical (CTNNB1-dependent) nor non-canonical WNT signaling mechanism, but was found to be GSK3β-dependent. Rather, SFRP4 was found to antognize AKT activity via a mechanism involving AMPK. This lead to the hypophosphorylation of FOXO1 and a decrease in the expression of a portion of the FSH and FOXO1 transcriptomes. Conversely, FSH-stimulated AMPK, AKT and FOXO1 phosphorylation levels were found to be increased in the granulosa cells of Sfrp4-null mice relative to wild-type controls. SFRP4 treatement of granulosa cells also induced autophagy by signaling via AKT-mTORC1-ULK1, as well as apoptosis. CONCLUSIONS This study identifies a novel GSK3β-AMPK-AKT signaling mechanism through which SFPR4 antagonizes FSH action, and further identifies SFRP4 as a novel regulator of granulosa cell autophagy. These findings provide a mechanistic basis for the phenotypic changes previously observed in Sfrp4-null mice, and broaden our understanding of the physiological roles of WNT signaling processes in the ovary.
Collapse
Affiliation(s)
- Michael Bérubé
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, 3200 rue Sicotte, St-Hyacinthe, QC, J2S 2M2, Canada
| | - Atefeh Abedini
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, 3200 rue Sicotte, St-Hyacinthe, QC, J2S 2M2, Canada
| | - Evelyne Lapointe
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, 3200 rue Sicotte, St-Hyacinthe, QC, J2S 2M2, Canada
| | - Samuel Gusscott
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, 3200 rue Sicotte, St-Hyacinthe, QC, J2S 2M2, Canada
| | - Julie Brind'Amour
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, 3200 rue Sicotte, St-Hyacinthe, QC, J2S 2M2, Canada
| | - Gustavo Zamberlam
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, 3200 rue Sicotte, St-Hyacinthe, QC, J2S 2M2, Canada
| | - Derek Boerboom
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, 3200 rue Sicotte, St-Hyacinthe, QC, J2S 2M2, Canada.
| |
Collapse
|
9
|
Ma X, Xu R, Chen J, Wang S, Hu P, Wu Y, Que Y, Du W, Cai X, Chen H, Guo J, Li TC, Ruan YC. The epithelial Na + channel (ENaC) in ovarian granulosa cells modulates Ca 2+ mobilization and gonadotrophin signaling for estrogen homeostasis and female fertility. Cell Commun Signal 2024; 22:398. [PMID: 39143495 PMCID: PMC11323461 DOI: 10.1186/s12964-024-01778-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 08/06/2024] [Indexed: 08/16/2024] Open
Abstract
Ovarian granulosa cells are essential to gonadotrophin-regulated estrogen production, female cycle maintenance and fertility. The epithelial Na+ channel (ENaC) is associated with female fertility; however, whether and how it plays a role in ovarian cell function(s) remained unexplored. Here, we report patch-clamp and Na+ imaging detection of ENaC expression and channel activity in both human and mouse ovarian granulosa cells, which are promoted by pituitary gonadotrophins, follicle stimulating hormone (FSH) or luteinizing hormone (LH). Cre-recombinase- and CRISPR-Cas9-based granulosa-specific knockout of ENaC α subunit (Scnn1a) in mice resulted in failed estrogen elevation at early estrus, reduced number of corpus luteum, abnormally extended estrus phase, reduced litter size and subfertility in adult female mice. Further analysis using technologies including RNA sequencing and Ca2+ imaging revealed that pharmacological inhibition, shRNA-based knockdown or the knockout of ENaC diminished spontaneous or stimulated Ca2+ oscillations, lowered the capacity of intracellular Ca2+ stores and impaired FSH/LH-stimulated transcriptome changes for estrogen production in mouse and/or human granulosa cells. Together, these results have revealed a previously undefined role of ENaC in modulating gonadotrophin signaling in granulosa cells for estrogen homeostasis and thus female fertility.
Collapse
Affiliation(s)
- Xiyang Ma
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Ruiyao Xu
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Junjiang Chen
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China
- Jinan University, Guangzhou, China
| | - Shan Wang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Peijie Hu
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Yong Wu
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Yanting Que
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Wanting Du
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Xiaojun Cai
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Hui Chen
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Jinghui Guo
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, China
| | - Tin Chiu Li
- Department of Obstetrics and Gynaecology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ye Chun Ruan
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China.
- Shenzhen Research Institute, The Hong Kong Polytechnic University, Shenzhen, China.
| |
Collapse
|
10
|
Zhang X, Zhang X, Zhang Z, Shi Y, Wang J, Ru S, Tian H. Bisphenol S causes excessive estrogen synthesis by activating FSHR and the downstream cAMP/PKA signaling pathway. Commun Biol 2024; 7:844. [PMID: 38987655 PMCID: PMC11237073 DOI: 10.1038/s42003-024-06449-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 06/12/2024] [Indexed: 07/12/2024] Open
Abstract
Estrogen excess in females has been linked to a diverse array of chronic and acute diseases. Emerging research shows that exposure to estrogen-like compounds such as bisphenol S leads to increases in 17β-estradiol levels, but the mechanism of action is unclear. The aim of this study was to reveal the underlying signaling pathway-mediated mechanisms, target site and target molecule of action of bisphenol S causing excessive estrogen synthesis. Human ovarian granulosa cells SVOG were exposed to bisphenol S at environmentally relevant concentrations (1 μg/L, 10 μg/L, and 100 μg/L) for 48 h. The results confirms that bisphenol S accumulates mainly on the cell membrane, binds to follicle stimulating hormone receptor (FSHR) located on the cell membrane, and subsequently activates the downstream cyclic adenosine monophosphate/protein kinase A (cAMP/PKA) signaling pathway, leading to enhanced conversion of testosterone to 17β-estradiol. This study deepens our knowledge of the mechanisms of environmental factors in pathogenesis of hyperestrogenism.
Collapse
Affiliation(s)
- Xiaorong Zhang
- College of Marine Life Sciences, Ocean University of China, 266003, Qingdao, Shandong Province, China
- Tai'an Agriculture and Rural Affairs Bureau, 271000, Tai'an, Shandong Province, China
| | - Xinda Zhang
- College of Marine Life Sciences, Ocean University of China, 266003, Qingdao, Shandong Province, China
| | - Zhenzhong Zhang
- College of Marine Life Sciences, Ocean University of China, 266003, Qingdao, Shandong Province, China
| | - Yijiao Shi
- College of Marine Life Sciences, Ocean University of China, 266003, Qingdao, Shandong Province, China
| | - Jun Wang
- College of Marine Life Sciences, Ocean University of China, 266003, Qingdao, Shandong Province, China
| | - Shaoguo Ru
- College of Marine Life Sciences, Ocean University of China, 266003, Qingdao, Shandong Province, China
| | - Hua Tian
- College of Marine Life Sciences, Ocean University of China, 266003, Qingdao, Shandong Province, China.
| |
Collapse
|
11
|
Xu Y, Zhang Z, Wang R, Xue S, Ying Q, Jin L. Roles of estrogen and its receptors in polycystic ovary syndrome. Front Cell Dev Biol 2024; 12:1395331. [PMID: 38961865 PMCID: PMC11219844 DOI: 10.3389/fcell.2024.1395331] [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: 03/03/2024] [Accepted: 05/21/2024] [Indexed: 07/05/2024] Open
Abstract
Polycystic ovary syndrome (PCOS) is an endocrine disorder characterized by abnormal steroid hormone levels in peripheral blood and poor-quality oocytes. In the ovary, androgen is produced by theca cells, and estrogen is produced by granulosa cells. Androgen is converted to estrogen in granulosa cells, with cytochrome P450 aromatase as the limiting enzyme during this process. Estrogen receptors (ER) include ER alpha, ER beta, and membrane receptor GPR30. Studies have demonstrated that the abnormal functions of estrogen and its receptors and estradiol synthesis-related enzymes are closely related to PCOS. In recent years, some estrogen-related drugs have made significant progress in clinical application for subfertility with PCOS, such as letrozole and clomiphene. This article will elaborate on the recent advances in PCOS caused by abnormal expression of estrogen and its receptors and the application of related targeted small molecule drugs in clinical research and treatment.
Collapse
Affiliation(s)
- Yao Xu
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Department of Assisted Reproduction, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ziyi Zhang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Department of Assisted Reproduction, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, China
| | - Rongxiang Wang
- Reproductive Medicine Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Songguo Xue
- Reproductive Medicine Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qian Ying
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Department of Assisted Reproduction, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Liping Jin
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Department of Assisted Reproduction, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, China
| |
Collapse
|
12
|
Vazakidou P, Evangelista S, Li T, Lecante LL, Rosenberg K, Koekkoek J, Salumets A, Velthut-Meikas A, Damdimopoulou P, Mazaud-Guittot S, Fowler PA, Leonards PEG, van Duursen MBM. The profile of steroid hormones in human fetal and adult ovaries. Reprod Biol Endocrinol 2024; 22:60. [PMID: 38778396 PMCID: PMC11110185 DOI: 10.1186/s12958-024-01233-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 05/15/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Reproduction in women is at risk due to exposure to chemicals that can disrupt the endocrine system during different windows of sensitivity throughout life. Steroid hormone levels are fundamental for the normal development and function of the human reproductive system, including the ovary. This study aims to elucidate steroidogenesis at different life-stages in human ovaries. METHODS We have developed a sensitive and specific LC-MS/MS method for 21 important steroid hormones and measured them at different life stages: in media from cultures of human fetal ovaries collected from elective terminations of normally progressing pregnancy and in media from adult ovaries from Caesarean section patients, and follicular fluid from women undergoing infertility treatment. Statistically significant differences in steroid hormone levels and their ratios were calculated with parametric tests. Principal component analysis (PCA) was applied to explore clustering of the ovarian-derived steroidogenic profiles. RESULTS Comparison of the 21 steroid hormones revealed clear differences between the various ovarian-derived steroid profiles. Interestingly, we found biosynthesis of both canonical and "backdoor" pathway steroid hormones and corticosteroids in first and second trimester fetal and adult ovarian tissue cultures. 17α-estradiol, a less potent naturally occurring isomer of 17β-estradiol, was detected only in follicular fluid. PCA of the ovarian-derived profiles revealed clusters from: adult ovarian tissue cultures with relatively high levels of androgens; first trimester and second trimester fetal ovarian tissue cultures with relatively low estrogen levels; follicular fluid with the lowest androgens, but highest corticosteroid, progestogen and estradiol levels. Furthermore, ratios of specific steroid hormones showed higher estradiol/ testosterone and estrone/androstenedione (indicating higher CYP19A1 activity, p < 0.01) and higher 17-hydroxyprogesterone/progesterone and dehydroepiandrosterone /androstenedione (indicating higher CYP17A1 activity, p < 0.01) in fetal compared to adult ovarian tissue cultures. CONCLUSIONS Human ovaries demonstrate de novo synthesis of non-canonical and "backdoor" pathway steroid hormones and corticosteroids. Elucidating the steroid profiles in human ovaries improves our understanding of physiological, life-stage dependent, steroidogenic capacity of ovaries and will inform mechanistic studies to identify endocrine disrupting chemicals that affect female reproduction.
Collapse
Affiliation(s)
- Paraskevi Vazakidou
- Section Environment and Health, Amsterdam Institute for Life and Environment, De Boelelaan 1085, Amsterdam, 1081 HV, The Netherlands.
| | - Sara Evangelista
- Section Environment and Health, Amsterdam Institute for Life and Environment, De Boelelaan 1085, Amsterdam, 1081 HV, The Netherlands
| | - Tianyi Li
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, SE-14186, Sweden
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, SE-14186, Sweden
| | - Laetitia L Lecante
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Kristine Rosenberg
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
- Nova Vita Clinic, Tallinn, Estonia
| | - Jacco Koekkoek
- Section Environment and Health, Amsterdam Institute for Life and Environment, De Boelelaan 1085, Amsterdam, 1081 HV, The Netherlands
| | - Andres Salumets
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, SE-14186, Sweden
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, SE-14186, Sweden
- Competence Center on Health Technologies, Tartu, Estonia
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Agne Velthut-Meikas
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Pauliina Damdimopoulou
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, SE-14186, Sweden
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, SE-14186, Sweden
| | - Séverine Mazaud-Guittot
- Univ Rennes, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Inserm, Rennes, F-35000, France
| | - Paul A Fowler
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Pim E G Leonards
- Section Environment and Health, Amsterdam Institute for Life and Environment, De Boelelaan 1085, Amsterdam, 1081 HV, The Netherlands
| | - Majorie B M van Duursen
- Section Environment and Health, Amsterdam Institute for Life and Environment, De Boelelaan 1085, Amsterdam, 1081 HV, The Netherlands
| |
Collapse
|
13
|
Moriwaki M, Liu L, James ER, Tolley N, O'Connora AM, Emery B, Aston KI, Campbell RA, Welt CK. Heterozygous Eif4nif1 Stop Gain Mice Replicate the Primary Ovarian Insufficiency Phenotype in Women. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.09.588694. [PMID: 38645151 PMCID: PMC11030307 DOI: 10.1101/2024.04.09.588694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
We created the c.1286C>G stop-gain mutation found in a family with primary ovarian insufficiency (POI) at age 30 years. The Eif4enif1 C57/Bl6 transgenic mouse model contained a floxed exon 10-19 cassette with a conditional knock-in cassette containing the c.1286C>G stop-gain mutation in exon 10. The hybrid offspring of CMV- Cre mice with Eif4enif1 WT/flx mice were designated Eif4enif1 WT/ Δ for simplicity. A subset of female heterozygotes ( Eif4enif1 WT/ Δ ) had no litters. In those with litters, the final litter was earlier (5.4±2.6 vs. 10.5±0.7 months; p=0.02). Heterozygous breeding pair ( Eif4enif1 WT/ Δ x Eif4enif1 WT/ Δ ) litter size was 60% of WT litter size (3.9±2.0 vs. 6.5±3.0 pups/litter; p <0.001). The genotypes were 35% Eif4enif1 WT/flx and 65% Eif4enif1 WT/ Δ , with no homozygotes. Homozygote embryos did not develop beyond the 4-8 cell stage. The number of follicles in ovaries from Eif4enif1 WT/ Δ mice was lower starting at the primordial (499±290 vs. 1445±381) and primary follicle stage (1069±346 vs. 1450±193) on day 10 (p<0.05). The preantral follicle number was lower starting on day 21 (213±86 vs. 522±227; p<0.01). Examination of ribosome protected mRNAs (RPR) demonstrated altered mRNA expression. The Eif4enif1 stop-gain mice replicate the POI phenotype in women. The unique mouse model provides a platform to study regulation of protein translation across oocyte and embryo development in mammals.
Collapse
|
14
|
Esteves SC, Humaidan P, Ubaldi FM, Alviggi C, Antonio L, Barratt CLR, Behre HM, Jørgensen N, Pacey AA, Simoni M, Santi D. APHRODITE criteria: addressing male patients with hypogonadism and/or infertility owing to altered idiopathic testicular function. Reprod Biomed Online 2024; 48:103647. [PMID: 38367592 DOI: 10.1016/j.rbmo.2023.103647] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 10/25/2023] [Accepted: 10/26/2023] [Indexed: 02/19/2024]
Abstract
RESEARCH QUESTION Can a novel classification system of the infertile male - 'APHRODITE' (Addressing male Patients with Hypogonadism and/or infeRtility Owing to altereD, Idiopathic TEsticular function) - stratify different subgroups of male infertility to help scientists to design clinical trials on the hormonal treatment of male infertility, and clinicians to counsel and treat the endocrinological imbalances in men and, ultimately, increase the chances of natural and assisted conception? DESIGN A collaboration between andrologists, reproductive urologists and gynaecologists, with specialization in reproductive medicine and expertise in male infertility, led to the development of the APHRODITE criteria through an iterative consensus process based on clinical patient descriptions and the results of routine laboratory tests, including semen analysis and hormonal testing. RESULTS Five patient groups were delineated according to the APHRODITE criteria; (1) Hypogonadotrophic hypogonadism (acquired and congenital); (2) Idiopathic male infertility with lowered semen analysis parameters, normal serum FSH and normal serum total testosterone concentrations; (3) A hypogonadal state with lowered semen analysis parameters, normal FSH and reduced total testosterone concentrations; (4) Lowered semen analysis parameters, elevated FSH concentrations and reduced or normal total testosterone concentrations; and (5) Unexplained male infertility in the context of unexplained couple infertility. CONCLUSION The APHRODITE criteria offer a novel and standardized patient stratification system for male infertility independent of aetiology and/or altered spermatogenesis, facilitating communication among clinicians, researchers and patients to improve reproductive outcomes following hormonal therapy. APHRODITE is proposed as a basis for future trials of the hormonal treatment of male infertility.
Collapse
Affiliation(s)
- Sandro C Esteves
- ANDROFERT, Andrology and Human Reproduction Clinic, Campinas, Brazil.; Department of Surgery (Division of Urology), University of Campinas (UNICAMP), Campinas, Brazil.; Faculty of Health, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark..
| | - Peter Humaidan
- Fertility Clinic at Skive Regional Hospital, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Filippo M Ubaldi
- IVIRMA Global Research Alliance, GENERA, Clinica Valle Giulia, Rome, Italy
| | - Carlo Alviggi
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Leen Antonio
- Department of Endocrinology, Universitair Ziekenhuis Leuven, Leuven, Belgium
| | | | - Hermann M Behre
- Center for Reproductive Medicine and Andrology, University Medicine Halle, Halle, Germany
| | - Niels Jørgensen
- Department of Growth and Reproduction and International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Allan A Pacey
- Faculty of Biology, Medicine and Health, Core Technology Facility, University of Manchester, Manchester, UK
| | - Manuela Simoni
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy.; Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero - Universitaria of Modena, Modena, Italy.; Unit of Andrology and Sexual Medicine of the Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero - Universitaria of Modena, Modena, Italy
| | - Daniele Santi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy.; Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero - Universitaria of Modena, Modena, Italy.; Unit of Andrology and Sexual Medicine of the Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero - Universitaria of Modena, Modena, Italy
| |
Collapse
|
15
|
Cáceres ARR, Cardone DA, Sanhueza MDLÁ, Bosch IM, Cuello-Carrión FD, Rodriguez GB, Scotti L, Parborell F, Halperin J, Laconi MR. Local effect of allopregnanolone in rat ovarian steroidogenesis, follicular and corpora lutea development. Sci Rep 2024; 14:6402. [PMID: 38493224 PMCID: PMC10944484 DOI: 10.1038/s41598-024-57102-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 03/14/2024] [Indexed: 03/18/2024] Open
Abstract
Allopregnanolone (ALLO) is a known neurosteroid and a progesterone metabolite synthesized in the ovary, CNS, PNS, adrenals and placenta. Its role in the neuroendocrine control of ovarian physiology has been studied, but its in situ ovarian effects are still largely unknown. The aims of this work were to characterize the effects of intrabursal ALLO administration on different ovarian parameters, and the probable mechanism of action. ALLO administration increased serum progesterone concentration and ovarian 3β-HSD2 while decreasing 20α-HSD mRNA expression. ALLO increased the number of atretic follicles and the number of positive TUNEL granulosa and theca cells, while decreasing positive PCNA immunostaining. On the other hand, there was an increase in corpora lutea diameter and PCNA immunostaining, whereas the count of TUNEL-positive luteal cells decreased. Ovarian angiogenesis and the immunohistochemical expression of GABAA receptor increased after ALLO treatment. To evaluate if the ovarian GABAA receptor was involved in these effects, we conducted a functional experiment with a specific antagonist, bicuculline. The administration of bicuculline restored the number of atretic follicles and the diameter of corpora lutea to normal values. These results show the actions of ALLO on the ovarian physiology of the female rat during the follicular phase, some of them through the GABAA receptor. Intrabursal ALLO administration alters several processes of the ovarian morpho-physiology of the female rat, related to fertility and oocyte quality.
Collapse
Affiliation(s)
- Antonella Rosario Ramona Cáceres
- Laboratorio de Fisiopatología Ovárica, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU - CONICET Mendoza), Av. Ruiz Leal s/n Parque General San Martín, CP 5500, Mendoza, Argentina
- Facultad de Ingeniería y Facultad de Ciencias Médicas, Universidad de Mendoza, Mendoza, Argentina
| | - Daniela Alejandra Cardone
- Laboratorio de Fisiopatología Ovárica, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU - CONICET Mendoza), Av. Ruiz Leal s/n Parque General San Martín, CP 5500, Mendoza, Argentina
| | - María de Los Ángeles Sanhueza
- Laboratorio de Fisiopatología Ovárica, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU - CONICET Mendoza), Av. Ruiz Leal s/n Parque General San Martín, CP 5500, Mendoza, Argentina
| | | | - Fernando Darío Cuello-Carrión
- Laboratorio de Oncología, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU - CONICET Mendoza), Mendoza, Argentina
| | | | - Leopoldina Scotti
- Ovarian Pathophysiology Studies Laboratory, Institute of Experimental Biology and Medicine (IByME) - CONICET, Buenos Aires, Argentina
| | - Fernanda Parborell
- Ovarian Pathophysiology Studies Laboratory, Institute of Experimental Biology and Medicine (IByME) - CONICET, Buenos Aires, Argentina
| | - Julia Halperin
- Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Ciudad Autónoma de Buenos Aires, Argentina
| | - Myriam Raquel Laconi
- Laboratorio de Fisiopatología Ovárica, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU - CONICET Mendoza), Av. Ruiz Leal s/n Parque General San Martín, CP 5500, Mendoza, Argentina.
- Facultad de Ingeniería y Facultad de Ciencias Médicas, Universidad de Mendoza, Mendoza, Argentina.
| |
Collapse
|
16
|
Li Y, Tan Z, Zuo P, Li M, Hou L, Wang X. Gestodene causes masculinization of the western mosquitofish (Gambusia affinis): Insights from ovary metabolomics. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 912:168693. [PMID: 38008334 DOI: 10.1016/j.scitotenv.2023.168693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/07/2023] [Accepted: 11/17/2023] [Indexed: 11/28/2023]
Abstract
Gestodene (GES) is a common synthetic progesterone frequently detected in aquatic environments. Chronic exposure to GES can cause masculinization of a variety of fish; however, whether metabolism is closely related to the masculinization has yet to be explored. Hence, the ovary metabolome of adult female western mosquitofish (Gambusia affinis) after exposing to GES (0.0, 5.0, 50.0, and 500.0 ng/L) for 40 days was analyzed by using high-performance liquid chromatography ionization with quadrupole time-of-flight tandem mass spectrometry (HPLC-QTOF-MS). The results showed that GES increased the levels of cysteine, taurine, ophthalmic acid and cAMP while decreased methionine, these metabolites changes may owing to the oxidative stress of the ovaries; while taurcholic acid and uric acid were decreased along with induced oocyte apopotosis. Steroids hormone metabolism was also significantly affected, with progesterone and cortisol being the most affected. Enzyme-linked immunoassay results showed that estradiol levels were decreased while testosterone levels were increased with GES exposure. In addition, correlation analysis showed that the differential metabolites of some amino acids (e.g. leucine) were strongly correlated with the levels of steroids hormones secreted by the pituitary gland. The results of this study suggest that GES affects ovarian metabolism via the hypothalamus-pituitary-gonad and hypothalamic-pituitary-adrenal axes, impair antioxidant capacity, induce apoptosis in the ovary of G. affinis, and finally caused masculinization.
Collapse
Affiliation(s)
- Yelin Li
- School of Life Sciences, Guangzhou University, Guangzhou 510006, China
| | - Zhiqing Tan
- School of Life Sciences, Guangzhou University, Guangzhou 510006, China; School of Life Sciences, Zhaoqing University, Zhaoqing 526000, China
| | - Peiyu Zuo
- School of Life Sciences, Guangzhou University, Guangzhou 510006, China
| | - Maorong Li
- School of Life Sciences, Guangzhou University, Guangzhou 510006, China
| | - Liping Hou
- School of Life Sciences, Guangzhou University, Guangzhou 510006, China.
| | - Xiaolan Wang
- School of Life Sciences, Guangzhou University, Guangzhou 510006, China.
| |
Collapse
|
17
|
Barcelos MN, Gonçalves-Santos E, Souza MA, Santos EC, Gonçalves RV, Castro-Gamero AM, Novaes RD. Prolonged testosterone 17β-cyclopentylpropionate exposition induces behavioral, ovarian, oviductal, uterine and reproductive disturbances in female mice. Life Sci 2024; 338:122408. [PMID: 38181852 DOI: 10.1016/j.lfs.2023.122408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/16/2023] [Accepted: 12/29/2023] [Indexed: 01/07/2024]
Abstract
Anabolic-androgenic steroids (AAS) abuse is often associated with metabolic disorders and infertility. However, the current evidence on AAS-induced reproductive toxicity is mainly based on male studies. Thus, AAS repercussions on female reproductive capacity remain poorly understood, despite scarce evidence that fertility determinants may be more severely impaired in females than males exposed to these drugs. Accordingly, this study used an integrated framework to investigate the impact of different testosterone 17β-cyclopentylpropionate (TC) doses on pain sensitivity, aggressiveness, anxiety, sexual behavior, ovarian, oviductal, uterine and reproductive morphofunctional and molecular outcomes. These parameters were used to explore the reproductive capacity in female mice exposed to this synthetic testosterone ester. The animals were untreated or intraperitoneally treated with 5, 10 and 20 mg/kg TC every 48 h for 12 weeks. Our findings indicated that testosterone was upregulated while the hormones luteinizing, follicle-stimulating, estrogen and progesterone were down-regulated by TC. This AAS also exerted deleterious effects on anxiety, aggressivity, nociception, exploratory and sexual behavior in female mice. Concurrently, TC attenuated ovarian follicle maturation, interrupted the estrous cycle, induced oviductal and uterine hypotrophy. Estrous cyclicity was reestablished 60 days after AAS treatment. However, TC-treated mice still exhibited impaired reproductive capacity, a disturbance potentially related to deficiency in folliculogenesis, sex hormones production, and endometrial receptivity mediate by ER-α, PR, HOXA-10 and LIF down-regulation. Taken together, our findings indicated that in addition to female behavior, reproductive organs microstructure and function are markedly impaired by TC in a dose-dependent manner, whose time-dependent reversibility remains to be clarified.
Collapse
Affiliation(s)
- Mônica N Barcelos
- Programa de Pós-Graduação em Biociências Aplicadas à Saúde, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil
| | - Elda Gonçalves-Santos
- Programa de Pós-Graduação em Biociências Aplicadas à Saúde, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil
| | - Matheus A Souza
- Programa de Pós-Graduação em Ciências Biológicas, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil
| | - Eliziária C Santos
- Faculdade de Medicina, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina 39100-000, Minas Gerais, Brazil
| | - Reggiani V Gonçalves
- Programa de Pós-Graduação em Biologia Animal, Departamento de Biologia Animal, Universidade Federal de Viçosa, Viçosa 36570-900, Minas Gerais, Brazil
| | - Angel Mauricio Castro-Gamero
- Programa de Pós-Graduação em Biociências Aplicadas à Saúde, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil; Instituto de Ciências da Natureza, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil
| | - Rômulo D Novaes
- Programa de Pós-Graduação em Biociências Aplicadas à Saúde, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil; Programa de Pós-Graduação em Ciências Biológicas, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil; Programa de Pós-Graduação em Biologia Animal, Departamento de Biologia Animal, Universidade Federal de Viçosa, Viçosa 36570-900, Minas Gerais, Brazil; Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Alfenas 37130-001, Minas Gerais, Brazil.
| |
Collapse
|
18
|
Nandi S, Kumar B S, Gupta PSP, Mondal S, Kumar VG. Influence of phenolic flavonols (Kaempferol, Querectin and Myricetin) on the survival and growth of ovine preantral follicles and granulosa cells cultured in vitro. Theriogenology 2024; 214:266-272. [PMID: 37948816 DOI: 10.1016/j.theriogenology.2023.10.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 09/01/2023] [Accepted: 10/16/2023] [Indexed: 11/12/2023]
Abstract
Study was carried out to examine the influence of plant bioactive compounds [Kaempferol (KAE), Querectin (QUE) and Myricetin (MYR)] on the survival and growth parameters of cultured ovine preantral follicles (PFs) granulosa cells (GCs) and expression of some key developmental genes. Ovine PFs were isolated from slaughterhouse derived ovaries and KAE, QUE and MYR were supplemented to the standard culture medium of GCs and PFs at concentrations of 0, 5, 10, 25, 50 and 100 μM and cultured for 5 and 7 days respectively. PFs morphological and functional parameters [follicle and enclosed oocyte growth rate, viability of follicles, antrum formation rate, oocyte maturation rate, estradiol concentration, reactive oxygen species (ROS) production] and GC growth parameters (metabolic activity, viability rate, cell number increment, ROS production) were measured after culture. Significantly higher PF growth, viability rate and estradiol concentration was observed at 10 μM, 25 μM and 10 μM concentration of KAE, MYR and QUE respectively compared to the control. ROS production was significantly decreased in the PF culture media treated with 10 μM KAE or MYR 25 μM or 10 μM QUE compared to those observed in the control group. Likewise, metabolic activity of GCs, viability rate and cell number increment cultured with KAE, MYR and QUE was significantly higher at 10, 25 and 10 μM concentrations respectively compared to those observed in control group. ROS production was significantly lower in the GC cultured with KAE, MYR and QUE at 10, 25 and 10 μM concentrations respectively compared to the control. Based on the results of the growth parameters, gene expression of PFs and GCs were studied by qPCR at selected concentrations (KAE, MYR and QUE at 10, 25 and 10 μM concentrations respectively) in the cultured PFs and GCs. Gene expression of GDF9, FGF2, CYP19A1 was significantly higher and Bax, Bcl2 expression was significantly lower in the PFs and GCs cultured with the KAE or QUE at 10 μM concentration. KAE, MYR and QUE have dose dependant responses on PFs and GCs morphological and functional parameters; however, KAE is more potent amongst the three in augmenting the ovarian functions.
Collapse
Affiliation(s)
- S Nandi
- ICAR-National Institute of Animal Nutrition and Physiology, Bangalore, India.
| | - Sampath Kumar B
- ICAR-National Institute of Animal Nutrition and Physiology, Bangalore, India; Veterinary College, Bangalore Campus, Hebbal, Bangalore, India
| | - P S P Gupta
- ICAR-National Institute of Animal Nutrition and Physiology, Bangalore, India
| | - S Mondal
- ICAR-National Institute of Animal Nutrition and Physiology, Bangalore, India
| | - V Girish Kumar
- Veterinary College, Bangalore Campus, Hebbal, Bangalore, India
| |
Collapse
|
19
|
Hayes E, Winston N, Stocco C. Molecular crosstalk between insulin-like growth factors and follicle-stimulating hormone in the regulation of granulosa cell function. Reprod Med Biol 2024; 23:e12575. [PMID: 38571513 PMCID: PMC10988955 DOI: 10.1002/rmb2.12575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 03/11/2024] [Accepted: 03/20/2024] [Indexed: 04/05/2024] Open
Abstract
Background The last phase of folliculogenesis is driven by follicle-stimulating hormone (FSH) and locally produced insulin-like growth factors (IGFs), both essential for forming preovulatory follicles. Methods This review discusses the molecular crosstalk of the FSH and IGF signaling pathways in regulating follicular granulosa cells (GCs) during the antral-to-preovulatory phase. Main findings IGFs were considered co-gonadotropins since they amplify FSH actions in GCs. However, this view is not compatible with data showing that FSH requires IGFs to stimulate GCs, that FSH renders GCs sensitive to IGFs, and that FSH signaling interacts with factors downstream of AKT to stimulate GCs. New evidence suggests that FSH and IGF signaling pathways intersect at several levels to regulate gene expression and GC function. Conclusion FSH and locally produced IGFs form a positive feedback loop essential for preovulatory follicle formation in all species. Understanding the mechanisms by which FSH and IGFs interact to control GC function will help design new interventions to optimize follicle maturation, perfect treatment of ovulatory defects, improve in vitro fertilization, and develop new contraceptive approaches.
Collapse
Affiliation(s)
- Emily Hayes
- Department of Physiology and BiophysicsUniversity of Illinois Chicago College of MedicineChicagoIllinoisUSA
| | - Nicola Winston
- Department of Obstetrics and GynecologyUniversity of Illinois Chicago College of MedicineChicagoIllinoisUSA
| | - Carlos Stocco
- Department of Physiology and BiophysicsUniversity of Illinois Chicago College of MedicineChicagoIllinoisUSA
- Department of Obstetrics and GynecologyUniversity of Illinois Chicago College of MedicineChicagoIllinoisUSA
| |
Collapse
|
20
|
Gao L, Zhang L, Zhang Y, Madaniyati M, Shi S, Huang L, Song X, Pang W, Chu G, Yang G. miR-10a-5p inhibits steroid hormone synthesis in porcine granulosa cells by targeting CREB1 and inhibiting cholesterol metabolism. Theriogenology 2023; 212:19-29. [PMID: 37683501 DOI: 10.1016/j.theriogenology.2023.08.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 08/12/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023]
Abstract
During growth, proliferation, differentiation, atresia, ovulation, and luteinization, the morphology and function of granulosa cells (GCs) change. Estrogen and progesterone are steroid hormones secreted by GCs that regulate the ovulation cycle of sows and help maintain pregnancy. miR-10a-5p is highly expressed in GCs and can inhibit GC proliferation. However, the role of miR-10a-5p in the steroid hormone synthesis of porcine GCs is unclear. In this study, miR-10a-5p agomir or antagomir was transfected into GCs. Overexpression of miR-10a-5p in GCs inhibited steroid hormone secretion and significantly downregulated steroid hormone synthesis via 3β-hydroxy steroid dehydrogenase and cytochrome P450 family 19 subfamily A member 1. Interference with miR-10a-5p had the opposite effect. Bodipy and Oil Red O staining showed that overexpression of miR-10a-5p significantly reduced the formation of lipid droplets. Overexpression significantly inhibited the content of total cholesterol esters in GCs. The mRNA and protein levels of 3-hydroxy-3-methylglutaryl-CoA reductase and scavenger receptor class B member 1 decreased significantly, and the opposite effects were seen by interference with miR-10a-5p. Bioinformatic analysis of potential targets identified cAMP-responsive element binding protein 1 as a potential target and dual-luciferase reporter system analysis confirmed that miR-10a-5p directly targets the 3' untranslated region. These findings suggest that miR-10a-5p inhibits the expression of 3β-hydroxy steroid dehydrogenase and cytochrome P450 family 19 subfamily A member 1 to inhibit the synthesis of steroid hormones in GCs. In addition, miR-10a-5p inhibits the cholesterol metabolism pathway of GCs to modulate steroid hormone synthesis.
Collapse
Affiliation(s)
- Lei Gao
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China; Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Lutong Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China; Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Yuli Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China; Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Mielie Madaniyati
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China; Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Shengjie Shi
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China; Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Liang Huang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China; Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Xiangrong Song
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China; Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Weijun Pang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China; Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Guiyan Chu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China; Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Gongshe Yang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China; Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China.
| |
Collapse
|
21
|
Awonuga AO, Camp OG, Abu-Soud HM. A review of nitric oxide and oxidative stress in typical ovulatory women and in the pathogenesis of ovulatory dysfunction in PCOS. Reprod Biol Endocrinol 2023; 21:111. [PMID: 37996893 PMCID: PMC10666387 DOI: 10.1186/s12958-023-01159-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 11/05/2023] [Indexed: 11/25/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is a heterogeneous functional endocrine disorder associated with a low-grade, chronic inflammatory state. Patients with PCOS present an increased risk of metabolic comorbidities and often menstrual dysregulation and infertility due to anovulation and/or poor oocyte quality. Multiple mechanisms including oxidative stress and low-grade inflammation are believed to be responsible for oocyte deterioration; however, the influence of nitric oxide (NO) insufficiency in oocyte quality and ovulatory dysfunction in PCOS is still a matter for debate. Higher production of superoxide (O2•-) mediated DNA damage and impaired antioxidant defense have been implicated as contributory factors for the development of PCOS, with reported alteration in superoxide dismutase (SOD) function, an imbalanced zinc/copper ratio, and increased catalase activity. These events may result in decreased hydrogen peroxide (H2O2) accumulation with increased lipid peroxidation events. A decrease in NO, potentially due to increased activity of NO synthase (NOS) inhibitors such as asymmetric dimethylarginine (ADMA), and imbalance in the distribution of reactive oxygen species (ROS), such as decreased H2O2 and increased O2•-, may offset the physiological processes surrounding follicular development, oocyte maturation, and ovulation contributing to the reproductive dysfunction in patients with PCOS. Thus, this proposal aims to evaluate the specific roles of NO, oxidative stress, ROS, and enzymatic and nonenzymatic elements in the pathogenesis of PCOS ovarian dysfunction, including oligo- anovulation and oocyte quality, with the intent to inspire better application of therapeutic options. The authors believe more consideration into the specific roles of oxidative stress, ROS, and enzymatic and nonenzymatic elements may allow for a more thorough understanding of PCOS. Future efforts elaborating on the role of NO in the preoptic nucleus to determine its influence on GnRH firing and follicle-stimulating hormone/Luteinizing hormone (FSH/LH) production with ovulation would be of benefit in PCOS. Consequently, treatment with an ADMA inhibitor or NO donor may prove beneficial to PCOS patients experiencing reproductive dysfunction and infertility.
Collapse
Affiliation(s)
- Awoniyi O Awonuga
- Departments of Obstetrics and Gynecology and Biochemistry and Molecular Biology, The C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, 275 E. Hancock Detroit, Detroit, MI, 48201, USA.
| | - Olivia G Camp
- Departments of Obstetrics and Gynecology and Biochemistry and Molecular Biology, The C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, 275 E. Hancock Detroit, Detroit, MI, 48201, USA
| | - Husam M Abu-Soud
- Departments of Obstetrics and Gynecology and Biochemistry and Molecular Biology, The C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, 275 E. Hancock Detroit, Detroit, MI, 48201, USA
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
- Department of Microbiology, Immunology and Biochemistry, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| |
Collapse
|
22
|
Maurya S, Tripathi S, Singh A. Ontogeny of adropin and its receptor expression during postnatal development and its pro-gonadal role in the ovary of pre-pubertal mouse. J Steroid Biochem Mol Biol 2023; 234:106404. [PMID: 37743028 DOI: 10.1016/j.jsbmb.2023.106404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/21/2023] [Accepted: 09/21/2023] [Indexed: 09/26/2023]
Abstract
Adropin, a highly conserved multifunctional peptide hormone, has a beneficial effect on the maintenance of gluco-lipid homeostasis, endothelial and cardiovascular functions. However, the expression and potential role of adropin in ovarian function are not fully elucidated. The present study aimed to investigate the expression of adropin and GPR19 in the mice ovary during various stages of postnatal development. This study also explored whether the treatment of adropin can modulate the timing of puberty, for which pre-pubertal mice were treated with adropin. The result showed the intense immunoreactivity of adropin in TICs, while GPR19 immunoreactivity was noted in GCs in infantile, pre-pubertal, and pubertal mice ovary. Also, adropin and GPR19 are highly expressed in the CL of the ovary of reproductively active mice. The fact that adropin expression in the ovary at different stages of postnatal development positively correlated with circulating progesterone and estradiol indicated that it has a role in the production of steroid hormones. Furthermore, the results of in vivo studies in pre-pubertal mice showed that adropin promotes early folliculogenesis by enhancing the proliferation (PCNA) of GCs of cortical ovarian follicles and promotes estradiol production by enhancing the expression of GPR19, StAR, CYP11A1 and aromatase proteins. Also, adropin treatment increases the Bax/Bcl2 ratio and expression of cleaved caspase-3 and ERα proteins, which may result in increased apoptosis of medullary follicles leading to the formation of a well-developed interstitium with interstitial glandular cells. Collectively, these findings indicate that adropin may be a factor that accelerates pubertal development in the ovary and could be utilized as a therapeutic approach for treating pubertal delay.
Collapse
Affiliation(s)
- Shweta Maurya
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Shashank Tripathi
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Ajit Singh
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India.
| |
Collapse
|
23
|
Widjiati W, Kuntjorodjakti S, Ananda AT, Sajida MVP, Ilmi AF, Adisti MZ, Chou D, Luqman EM. The Effect of Administering Forest Honey to Rats Exposed to Physical Stress on Corticosteroid Levels, Folliculogenesis and the Number of Corpus Luteum. BIOMEDICAL AND PHARMACOLOGY JOURNAL 2023; 16:1431-1440. [DOI: 10.13005/bpj/2721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Livestock that has stress releases glucocorticoids in response to it, and it causes inhibition of the hypothalamus-pituitary-gonadal axis (HPG) signaling pathway so that it reduces reproductive efficiency. Forest honey can reduce corticosteroid levels as a stress response from physical stress induction which is expected to increase reproductive efficiency including folliculogenesis and the formation of the corpus luteum. This study aims to determine the effect of forest honey on rats (Rattus novergicus) exposed to physical stress on corticosteroid levels, folliculogenesis, and the number of corpus luteum. This study is an experimental laboratory one using 32 rats which were divided into 4 treatment groups; control positive (C) treated with physical stress, treatment 1 (T1) treated with physical stress + honey 2 g/rat/day PO, treatment 2 (T2) treated with physical stress + honey 4 g/rat/day PO and treatment 3 (T3) treated with physical stress + honey 6 g/rat/day PO. All treatments were carried out for 14 days. The results showed that T1 had the lowest corticosteroid level compared to all treatment groups and the corticosteroid level of this group was significantly different (p <0.05) compared to that of C and T3. The folliculogenesis profile showed that the number of primary secondary, tertiary, and Graafian follicles of group T1 was significantly different (p<0.05) compared to that of C, T2, and T3. In terms of the number of corpus luteum, it showed that T1 had the highest number of corpus luteum, and the number of corpus luteum in this group was significantly different (p<0.05) from that of C, T2, and T3. It can be concluded that the administration of forest honey at a dose of 2g/rat/day could reduce corticosteroid levels, improve the folliculogenesis profile, and increase the number of corpus luteum in rats exposed to physical stress. The use of forest honey could reduce corticosteroid levels as a stress response from physical stress induction which was expected to increase reproductive efficiency.
Collapse
Affiliation(s)
- Widjiati Widjiati
- 1Department of Veterinary Science, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya 60115, Jawa Timur, Indonesia,
| | - Suryo Kuntjorodjakti
- 1Department of Veterinary Science, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya 60115, Jawa Timur, Indonesia,
| | - Aditya Tri Ananda
- 2Postgraduate Reproductive Biology, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya 60115, Jawa Timur, Indonesia
| | - Mey Vanda Pusparina Sajida
- 2Postgraduate Reproductive Biology, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya 60115, Jawa Timur, Indonesia
| | - Alivia Fairuz Ilmi
- 2Postgraduate Reproductive Biology, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya 60115, Jawa Timur, Indonesia
| | - Meisa Zalfa Adisti
- 3Graduate Student Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya 60115, Jawa Timur, Indonesia,
| | - Dean Chou
- 4Department of Biomedical Engineering National Cheng Kung University, No. 1, Dasyue Rd, East District, Tainan City, Taiwan
| | - Epy Muhammad Luqman
- 1Department of Veterinary Science, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya 60115, Jawa Timur, Indonesia,
| |
Collapse
|
24
|
Camajani E, Feraco A, Verde L, Moriconi E, Marchetti M, Colao A, Caprio M, Muscogiuri G, Barrea L. Ketogenic Diet as a Possible Non-pharmacological Therapy in Main Endocrine Diseases of the Female Reproductive System: A Practical Guide for Nutritionists. Curr Obes Rep 2023; 12:231-249. [PMID: 37405618 PMCID: PMC10482777 DOI: 10.1007/s13679-023-00516-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/20/2023] [Indexed: 07/06/2023]
Abstract
PURPOSEOF REVIEW This narrative review explored the role of ketogenic diets (KDs) in improving fertility outcomes, low-grade inflammation, body weight, visceral adipose tissue, and its potential use in certain types of cancer, through its favorable actions on mitochondrial function, reactive oxygen species generation, chronic inflammation, and tumor growth inhibition. RECENT FINDINGS : Nutrition is crucial to maintain the female reproductive system's health. Evidence on the association between diet and female reproductive system has greatly expanded over the last decade, leading to the identification of specific diet therapy, particularly KDs. KDs has been proved to be an effective weight-loss tool. To date, KDs is being increasingly used in the treatment of many diseases, such as obesity, type 2 diabetes mellitus. KDs is a dietary intervention capable of ameliorating the inflammatory state and oxidative stress through several mechanisms. Due to the increasing use of KDs beyond obesity, this literature review will provide the latest scientific evidence of its possible use in common disorders of the female endocrine-reproductive tract, and a practical guide to its use in these patients.
Collapse
Affiliation(s)
- Elisabetta Camajani
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166, Rome, Italy
| | - Alessandra Feraco
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166, Rome, Italy
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele, Rome, Italy
| | - Ludovica Verde
- Centro Italiano Per La Cura E Il Benessere del Paziente Con Obesità (C.I.B.O), Dipartimento Di Medicina Clinica E Chirurgia, Unit of Endocrinology, Università Degli Studi Di Napoli Federico II, Naples, Italy
- Department of Public Health, University "Federico II" of Naples, 80138, Naples, Italy
| | - Eleonora Moriconi
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele, Rome, Italy
| | - Marco Marchetti
- Section of Clinical Nutrition and Nutrigenomic, Department of Biomedicine and Prevention, University of Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
| | - Annamaria Colao
- Centro Italiano Per La Cura E Il Benessere del Paziente Con Obesità (C.I.B.O), Dipartimento Di Medicina Clinica E Chirurgia, Unit of Endocrinology, Università Degli Studi Di Napoli Federico II, Naples, Italy
- Dipartimento Di Medicina Clinica E Chirurgia, Unità Di Diabetologia E Andrologia, Università Degli Studi Di Napoli Federico II, Via Sergio Pansini 5, 80131Naples, , Endocrinologia, Italy
- Cattedra Unesco "Educazione Alla Salute E Allo Sviluppo Sostenibile", Università Degli Studi Di Napoli Federico II, Naples, Italy
| | - Massimiliano Caprio
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166, Rome, Italy
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele, Rome, Italy
| | - Giovanna Muscogiuri
- Centro Italiano Per La Cura E Il Benessere del Paziente Con Obesità (C.I.B.O), Dipartimento Di Medicina Clinica E Chirurgia, Unit of Endocrinology, Università Degli Studi Di Napoli Federico II, Naples, Italy.
- Dipartimento Di Medicina Clinica E Chirurgia, Unità Di Diabetologia E Andrologia, Università Degli Studi Di Napoli Federico II, Via Sergio Pansini 5, 80131Naples, , Endocrinologia, Italy.
- Cattedra Unesco "Educazione Alla Salute E Allo Sviluppo Sostenibile", Università Degli Studi Di Napoli Federico II, Naples, Italy.
| | - Luigi Barrea
- Centro Italiano Per La Cura E Il Benessere del Paziente Con Obesità (C.I.B.O), Dipartimento Di Medicina Clinica E Chirurgia, Unit of Endocrinology, Università Degli Studi Di Napoli Federico II, Naples, Italy
- Dipartimento Di Scienze Umanistiche, Centro Direzionale, Università Telematica Pegaso, Via Porzio Isola F2, 80143, Naples, Italy
| |
Collapse
|
25
|
Lai X, Peng S, Liu L, Zou Z, Cao L, Wang Y. Tissue-specific promoters regulate the transcription of cyp19a1 in the brain-pituitary-gonad axis of Japanese eel (Anguilla japonica). J Steroid Biochem Mol Biol 2023; 232:106334. [PMID: 37236374 DOI: 10.1016/j.jsbmb.2023.106334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 05/16/2023] [Accepted: 05/22/2023] [Indexed: 05/28/2023]
Abstract
Aromatase is a key enzyme that catalyzes the biosynthesis of estrogens. Previous study indicated that putative tissue-specific promoters of the one aromatase gene (cyp19a1) may drive the differential regulatory mechanisms of cyp19a1 expression in Anguilla japonica. In the present study, for elucidating the transcription characteristics and the function of putative tissue-specific promoters of cyp19a1 in the brain-pituitary-gonad (BPG) axis during vitellogenesis, we investigated the transcriptional regulation of cyp19a1 by 17β-estrogen (E2), testosterone (T), or human chorionic gonadotropin (HCG) in A. japonica. The expression of estrogen receptor (esra), androgen receptor (ara), or luteinizing hormone receptor (lhr) was up-regulated as cyp19a1 in response to E2, T, or HCG, respectively in the telencephalon, diencephalon, and pituitary. The expression of cyp19a1 was also upregulated in the ovary by HCG or T in a dose-dependent manner. Unlike in the brain and pituitary, the expression of esra and lhr, rather than ara, was upregulated by T in the ovary. Subsequently, four primary subtypes of 5'-untranslated terminal regions of cyp19a1 transcripts and the corresponding two 5' flanking regions (promoter P.I and P.II) were identified. The P.II existed in all BPG axis tissues, whereas the P.I with strong transcriptional activity was brain- and pituitary-specific. Furthermore, the transcriptional activity of promoters, the core promoter region, and the three putative hormone receptor response elements were validated. The transcriptional activity did not change when the HEK291T cells co-transfected with P.II and ar vector were exposed to T. These results suggested that the expression of cyp19a1 was upregulated indirectly through esra and lhr rather than ara by T in the ovary, whereas the expression of cyp19a1 was upregulated directly through androgen receptor and the downstream androgen response element of tissue-specific P.I in the brain and pituitary. The results of the study reveal the regulatory mechanisms of estrogen biosynthesis and provide a reference for optimizing the technology of artificially induced maturation in eels.
Collapse
Affiliation(s)
- Xiaojian Lai
- Fisheries College, Jimei University, Xiamen 361021, China; Engineering Research Center of the Modern Technology for Eel Industry, Ministry of Education P. R. China, Xiamen 361021, China.
| | - Shuai Peng
- Fisheries College, Jimei University, Xiamen 361021, China
| | - Liping Liu
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai 201306, China
| | - Zhihua Zou
- Fisheries College, Jimei University, Xiamen 361021, China
| | - Le Cao
- Fisheries College, Jimei University, Xiamen 361021, China; Engineering Research Center of the Modern Technology for Eel Industry, Ministry of Education P. R. China, Xiamen 361021, China
| | - Yilei Wang
- Fisheries College, Jimei University, Xiamen 361021, China.
| |
Collapse
|
26
|
Rashid R, Tripathi R, Singh A, Sarkar S, Kawale A, Bader GN, Gupta S, Gupta RK, Jha RK. Naringenin improves ovarian health by reducing the serum androgen and eliminating follicular cysts in letrozole-induced polycystic ovary syndrome in the Sprague Dawley rats. Phytother Res 2023; 37:4018-4041. [PMID: 37165686 DOI: 10.1002/ptr.7860] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 04/03/2023] [Accepted: 04/17/2023] [Indexed: 05/12/2023]
Abstract
Polycystic ovary syndrome (PCOS) is most common in women of reproductive age, giving rise to androgen excess and anovulation, leading to infertility and non-reproductive complications. We explored the ameliorating effect of naringenin in PCOS using the Sprague Dawley (SD) rat model and human granulosa cells. Letrozole-induced PCOS rats were given either naringenin (50 mg/kg/day) alone or in combination with metformin (300 mg/kg/day), followed by the estrous cycle, hormonal analysis, and glucose sensitivity test. To evaluate the effect of naringenin on granulosa cell (hGC) steroidogenesis, we treated cells with naringenin (2.5 μM) alone or in combination with metformin (1 mM) in the presence of forskolin (10 μM). To determine the steroidogenesis of CYP-17A1, -19A1, and 3βHSD2, the protein expression levels were examined. Treatment with naringenin in the PCOS animal groups increased ovulation potential and decreased cystic follicles and levels of androgens. The expression levels of CYP-17A1, -19A1, and 3βHSD2, were seen restored in the ovary of PCOS SD rats' model and in the human ovarian cells in response to the naringenin. We found an increased expression level of phosphorylated-AKT in the ovary and hGCs by naringenin. Naringenin improves ovulation and suppress androgens and cystic follicles, involving AKT activation.
Collapse
Affiliation(s)
- Rumaisa Rashid
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow, India
- Department of Pharmaceutical Sciences, University of Kashmir, Jammu and Kashmir, India
| | - Rupal Tripathi
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Akanksha Singh
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Sudarsan Sarkar
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Ajaykumar Kawale
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - G N Bader
- Department of Pharmaceutical Sciences, University of Kashmir, Jammu and Kashmir, India
| | - Satish Gupta
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Rakesh Kumar Gupta
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Rajesh Kumar Jha
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
27
|
Madsen JF, Amoushahi M, Choi CP, Bundgaard S, Heuck A, Lykke-Hartmann K. Inhibition of phosphodiesterase PDE8B reduces activation of primordial follicles in mouse ovaries. Mol Reprod Dev 2023; 90:378-388. [PMID: 37499226 DOI: 10.1002/mrd.23699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 06/12/2023] [Accepted: 07/12/2023] [Indexed: 07/29/2023]
Abstract
In the ovaries, cyclic adenosine 3',5'-monophosphate (cAMP) is a second messenger supporting the generation of steroids. Phosphodiesterases (PDEs) are regulators of intracellular cAMP, and therefore, potential regulators of ovarian function. Interestingly, the family of PDE genes are differentially expressed in human oocytes and granulosa cells from primordial and primary follicles, suggesting diverse roles. In this study, we addressed the functions of PDE3B and PDE8B in primordial follicle regulation using inhibitors of PDE3B and PDE8B in murine ovary primary in vitro cultures. Inhibition of PDE8B in ovarian cultures prevented primordial follicle activation, while inhibition of PDE3B had no effect on follicle distribution in the ovary, under the tested conditions. As cAMP levels may increase steroid levels, we assessed the protein levels of the steroidogenic acute regulatory protein (StAR) and aromatase enzymes, and found that inhibition of PDE3B reduced StAR protein levels, whereas inhibition of PDE8 did not alter StAR expression in our murine ovary culture system conditions. Our results showed that ketotifen-induced inhibition of PDE8B can decrease primordial follicle activation, whereas we observed no effect of follicle distribution, when PDE3B was inhibited. Expression of the StaR enzyme was not altered when PDE8B was inhibited, which might reflect not sufficient inhibition by ketotifen to induce StAR alterations, or redundant mechanisms.
Collapse
Affiliation(s)
| | | | | | - Stine Bundgaard
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Anders Heuck
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Karin Lykke-Hartmann
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Clinical Genetics, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
28
|
Sun Y, Liu Z, Zhang W, Lin H, Li Q, Liu C, Zhang C. Paternal genetic effects of cadmium exposure during pregnancy on hormone synthesis disorders in ovarian granulosa cells of offspring. J Ovarian Res 2023; 16:98. [PMID: 37194017 DOI: 10.1186/s13048-023-01175-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 04/25/2023] [Indexed: 05/18/2023] Open
Abstract
The aim of this study was to investigate the paternal genetic intergenerational and transgenerational genetic effects of cadmium (Cd) exposure during pregnancy on estradiol (E2) and progesterone (Pg) synthesis in the ovarian granulosa cells (GCs) of offspring. Pregnant SD rats were intragastrically exposed to CdCl2 (0, 0.5, 2.0, 8.0 mg/kg) from days 1 to 20 to produce the F1 generation, F1 males were mated with newly purchased females to produce the F2 generation, and the F3 generation was obtained in the same way. Using this model, Cd-induced hormone synthesis disorders in GCs of F1 have been observed [8]. In this study, altered serum E2 and Pg levels in both F2 and F3 generations showed a nonmonotonic dose‒response relationship. In addition, hormone synthesis-related genes (Star, Cyp11a1, Cyp17a1, Cyp19a1, Sf-1) and miRNAs were observed to be altered in both F2 and F3. No differential changes in DNA methylation modifications of hormone synthesis-related genes were observed, and only the Adcy7 was hypomethylated. In summary, paternal genetic intergenerational and transgenerational effects exist in ovarian GCs E2 and Pg synthesis disorders induced by Cd during pregnancy. In F2, the upregulation of StAR and CYP11A1, and changes in the miR-27a-3p, miR-27b-3p, and miR-146 families may be important, while changes in the miR-10b-5p and miR-146 families in F3 may be important.
Collapse
Affiliation(s)
- Yi Sun
- Department of Preventive Medicine, Fujian Provincial Key Laboratory of Environmental Factors and Cancer, Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China
- Key Laboratory of Environment and Female Reproductive Health, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Zhangpin Liu
- Department of Preventive Medicine, Fujian Provincial Key Laboratory of Environmental Factors and Cancer, Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China
| | - Wenchang Zhang
- Department of Preventive Medicine, Fujian Provincial Key Laboratory of Environmental Factors and Cancer, Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China.
| | - Hao Lin
- Fuzhou Center for Disease Control and Prevention, Fuzhou, 350005, Fujian Province, China
| | - Qingyu Li
- Department of Preventive Medicine, Fujian Provincial Key Laboratory of Environmental Factors and Cancer, Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China
| | - Chenchen Liu
- Department of Preventive Medicine, Fujian Provincial Key Laboratory of Environmental Factors and Cancer, Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian Province, China
| | - Chenyun Zhang
- School of Health Management, Fujian Medical University, Fuzhou, 350122, Fujian Province, China.
| |
Collapse
|
29
|
Lim J, Lee HL, Nguyen J, Shin J, Getze S, Quach C, Squire E, Jung KM, Mahler SV, Mackie K, Piomelli D, Luderer U. Adolescent exposure to low-dose Δ9-tetrahydrocannabinol depletes the ovarian reserve in female mice. Toxicol Sci 2023; 193:31-47. [PMID: 36912754 PMCID: PMC10176244 DOI: 10.1093/toxsci/kfad027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2023] Open
Abstract
Cannabis use by adolescents is widespread, but its effects on the ovaries remain largely unknown. Δ9-tetrahydrocannabinol (THC) exerts its pharmacological effects by activating, and in some conditions hijacking, cannabinoid receptors (CBRs). We hypothesized that adolescent exposure to THC affects ovarian function in adulthood. Peripubertal female C57BL/6N mice were given THC (5 mg/kg) or its vehicle, once daily by intraperitoneal injection. Some mice received THC from postnatal day (PND) 30-33 and their ovaries were harvested PND34; other mice received THC from PND30-43, and their ovaries were harvested PND70. Adolescent treatment with THC depleted ovarian primordial follicle numbers by 50% at PND70, 4 weeks after the last dose. The treatment produced primordial follicle activation, which persisted until PND70. THC administration also caused DNA damage in primary follicles and increased PUMA protein expression in oocytes of primordial and primary follicles. Both CB1R and CB2R were expressed in oocytes and theca cells of ovarian follicles. Enzymes involved in the formation (N-acylphosphatidylethanolamine phospholipase D) or deactivation (fatty acid amide hydrolase) of the endocannabinoid anandamide were expressed in granulosa cells of ovarian follicles and interstitial cells. Levels of mRNA for CBR1 were significantly increased in ovaries after adolescent THC exposure, and upregulation persisted for at least 4 weeks. Our results support that adolescent exposure to THC may cause aberrant activation of the ovarian endocannabinoid system in female mice, resulting in substantial loss of ovarian reserve in adulthood. Relevance of these findings to women who frequently used cannabis during adolescence warrants investigation.
Collapse
Affiliation(s)
- Jinhwan Lim
- Department of Environmental and Occupational Health, University of California Irvine, Irvine, California 92697, USA
- Dept. of Medicine, University of California Irvine, Irvine, California 92697, USA
| | - Hye-Lim Lee
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, California 92697, USA
| | - Julie Nguyen
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, California 92697, USA
| | - Joyce Shin
- Department of Environmental and Occupational Health, University of California Irvine, Irvine, California 92697, USA
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, California 92697, USA
| | - Samantha Getze
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, California 92697, USA
| | - Caitlin Quach
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, California 92697, USA
| | - Erica Squire
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, California 92697, USA
| | - Kwang-Mook Jung
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, California 92697, USA
| | - Stephen V Mahler
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, California 92697, USA
| | - Ken Mackie
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana 47405, USA
| | - Daniele Piomelli
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, California 92697, USA
| | - Ulrike Luderer
- Department of Environmental and Occupational Health, University of California Irvine, Irvine, California 92697, USA
- Dept. of Medicine, University of California Irvine, Irvine, California 92697, USA
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, California 92697, USA
| |
Collapse
|
30
|
Parker J. Pathophysiological Effects of Contemporary Lifestyle on Evolutionary-Conserved Survival Mechanisms in Polycystic Ovary Syndrome. Life (Basel) 2023; 13:life13041056. [PMID: 37109585 PMCID: PMC10145572 DOI: 10.3390/life13041056] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/15/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is increasingly being characterized as an evolutionary mismatch disorder that presents with a complex mixture of metabolic and endocrine symptoms. The Evolutionary Model proposes that PCOS arises from a collection of inherited polymorphisms that have been consistently demonstrated in a variety of ethnic groups and races. In utero developmental programming of susceptible genomic variants are thought to predispose the offspring to develop PCOS. Postnatal exposure to lifestyle and environmental risk factors results in epigenetic activation of developmentally programmed genes and disturbance of the hallmarks of health. The resulting pathophysiological changes represent the consequences of poor-quality diet, sedentary behaviour, endocrine disrupting chemicals, stress, circadian disruption, and other lifestyle factors. Emerging evidence suggests that lifestyle-induced gastrointestinal dysbiosis plays a central role in the pathogenesis of PCOS. Lifestyle and environmental exposures initiate changes that result in disturbance of the gastrointestinal microbiome (dysbiosis), immune dysregulation (chronic inflammation), altered metabolism (insulin resistance), endocrine and reproductive imbalance (hyperandrogenism), and central nervous system dysfunction (neuroendocrine and autonomic nervous system). PCOS can be a progressive metabolic condition that leads to obesity, gestational diabetes, type two diabetes, metabolic-associated fatty liver disease, metabolic syndrome, cardiovascular disease, and cancer. This review explores the mechanisms that underpin the evolutionary mismatch between ancient survival pathways and contemporary lifestyle factors involved in the pathogenesis and pathophysiology of PCOS.
Collapse
Affiliation(s)
- Jim Parker
- School of Medicine, University of Wollongong, Wollongong, NSW 2522, Australia
| |
Collapse
|
31
|
Krajnik K, Mietkiewska K, Skowronska A, Kordowitzki P, Skowronski MT. Oogenesis in Women: From Molecular Regulatory Pathways and Maternal Age to Stem Cells. Int J Mol Sci 2023; 24:ijms24076837. [PMID: 37047809 PMCID: PMC10095116 DOI: 10.3390/ijms24076837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/31/2023] [Accepted: 04/03/2023] [Indexed: 04/08/2023] Open
Abstract
It is a well-known fact that the reproductive organs in women, especially oocytes, are exposed to numerous regulatory pathways and environmental stimuli. The maternal age is one cornerstone that influences the process of oocyte fertilization. More precisely, the longer a given oocyte is in the waiting-line to be ovulated from menarche to menopause, the longer the duration from oogenesis to fertilization, and therefore, the lower the chances of success to form a viable embryo. The age of menarche in girls ranges from 10 to 16 years, and the age of menopause in women ranges from approximately 45 to 55 years. Researchers are paying attention to the regulatory pathways that are impacting the oocyte at the very beginning during oogenesis in fetal life to discover genes and proteins that could be crucial for the oocyte’s lifespan. Due to the general trend in industrialized countries in the last three decades, women are giving birth to their first child in their thirties. Therefore, maternal age has become an important factor impacting oocytes developmental competence, since the higher a woman’s age, the higher the chances of miscarriage due to several causes, such as aneuploidy. Meiotic failures during oogenesis, such as, for instance, chromosome segregation failures or chromosomal non-disjunction, are influencing the latter-mentioned aging-related phenomenon too. These errors early in life of women can lead to sub- or infertility. It cannot be neglected that oogenesis is a precisely orchestrated process, during which the oogonia and primary oocytes are formed, and RNA synthesis takes place. These RNAs are crucial for oocyte growth and maturation. In this review, we intend to describe the relevance of regulatory pathways during the oogenesis in women. Furthermore, we focus on molecular pathways of oocyte developmental competence with regard to maternal effects during embryogenesis. On the background of transcriptional mechanisms that enable the transition from a silenced oocyte to a transcriptionally active embryo, we will briefly discuss the potential of induced pluripotent stem cells.
Collapse
Affiliation(s)
- Kornelia Krajnik
- Department of Basic and Preclinical Sciences, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, 87-100 Torun, Poland
| | - Klaudia Mietkiewska
- Department of Basic and Preclinical Sciences, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, 87-100 Torun, Poland
| | - Agnieszka Skowronska
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-719 Olsztyn, Poland
| | - Pawel Kordowitzki
- Department of Basic and Preclinical Sciences, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, 87-100 Torun, Poland
| | - Mariusz T. Skowronski
- Department of Basic and Preclinical Sciences, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, 87-100 Torun, Poland
| |
Collapse
|
32
|
Fedeli V, Catizone A, Querqui A, Unfer V, Bizzarri M. The Role of Inositols in the Hyperandrogenic Phenotypes of PCOS: A Re-Reading of Larner’s Results. Int J Mol Sci 2023; 24:ijms24076296. [PMID: 37047265 PMCID: PMC10093919 DOI: 10.3390/ijms24076296] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/22/2023] [Accepted: 03/22/2023] [Indexed: 03/30/2023] Open
Abstract
Polycystic ovarian syndrome (PCOS) is the most common endocrinological disorder in women, in which, besides chronic anovulation/oligomenorrhea and ovarian cysts, hyperandrogenism plays a critical role in a large fraction of subjects. Inositol isomers—myo-Inositol and D-Chiro-Inositol—have recently been pharmacologically effective in managing many PCOS symptoms while rescuing ovarian fertility. However, some disappointing clinical results prompted the reconsideration of their specific biological functions. Surprisingly, D-Chiro-Ins stimulates androgen synthesis and decreases the ovarian estrogen pathway; on the contrary, myo-Ins activates FSH response and aromatase activity, finally mitigating ovarian hyperandrogenism. However, when the two isomers are given in association—according to the physiological ratio of 40:1—patients could benefit from myo-Ins enhanced FSH and estrogen responsiveness, while taking advantage of the insulin-sensitizing effects displayed mostly by D-Chiro-Ins. We need not postulate insulin resistance to explain PCOS pathogenesis, given that insulin hypersensitivity is likely a shared feature of PCOS ovaries. Indeed, even in the presence of physiological insulin stimulation, the PCOS ovary synthesizes D-Chiro-Ins four times more than that measured in control theca cells. The increased D-Chiro-Ins within the ovary is detrimental in preserving steroidogenic control, and this failure can easily explain why treatment strategies based upon high D-Chiro-Ins have been recognized as poorly effective. Within this perspective, two factors emerge as major determinants in PCOS: hyperandrogenism and reduced aromatase expression. Therefore, PCOS could no longer be considered a disease only due to increased androgen synthesis without considering the contemporary downregulation of aromatase and FSH receptors. Furthermore, these findings suggest that inositols can be specifically effective only for those PCOS phenotypes featured by hyperandrogenism.
Collapse
|
33
|
Belani MA, Shah P, Banker M, Gupta SS. Investigating the potential role of swertiamarin on insulin resistant and non-insulin resistant granulosa cells of poly cystic ovarian syndrome patients. J Ovarian Res 2023; 16:55. [PMID: 36932437 PMCID: PMC10024427 DOI: 10.1186/s13048-023-01126-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 02/23/2023] [Indexed: 03/19/2023] Open
Abstract
BACKGROUND AND AIM Conventional drugs have limitations due to prevalence of contraindications in PCOS patients. To explore the potential effects of swertiamarin, on abrupted insulin and steroidogenic signaling in human luteinized granulosa cells from PCOS patients with or without insulin resistance. EXPERIMENTAL PROCEDURE hLGCs from 8 controls and 16 PCOS patients were classified for insulin resistance based on down regulation of protein expression of insulin receptor-β (INSR- β) as shown in our previous paper. Cells were grouped as control, PCOS-IR and PCOS-NIR, treated with swertiamarin (66 µM) and metformin (1 mM). Expression of key molecules involved in insulin signaling, fat metabolism, IGF system and steroidogenesis were compared between groups. RESULTS Swertiamarin significantly (P < 0.05) reversed the expression of INSR-β, PI(3)K, p-Akt, PKC-ζ, PPARγ, (P < 0.01) IRS (Ser 307) and IGF system in PCOS-IR group and was equally potent to metformin. In the same group, candidate genes viz SREBP1c, FAS, ACC-1 and CPT-1 were down regulated by swertiamarin (P < 0.001) and metformin (P < 0.001). Significant upregulation was demonstrated in expression of StAR, CYP19A1, 17β-HSD and 3β-HSD when treated with swertiamarin (P < 0.01) and metformin (P < 0.01) in PCOS-IR followed by increase in 17β-HSD and 3β-HSD enzyme activity along with estradiol and progesterone secretions. However, swertiamarin did not reveal any effect on PCOS-NIR group as compared to metformin that significantly (P < 0.01) reversed all the parameters related to steroidogenesis and down regulated basal expression of insulin signaling genes. CONCLUSION Swertiamarin, presents itself as a potential fertility drug in hLGCs from PCOS-IR patients.
Collapse
Affiliation(s)
- Muskaan A. Belani
- Dr. Vikram Sarabhai Institute of Cell and Molecular Biology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat 390 002 India
| | - Preeti Shah
- Nova IVI Fertility, Behind Xavier’s Ladies Hostel, 108, Swastik Society Rd, Navrangpura, Ahmedabad, 390009 Gujarat India
| | - Manish Banker
- Nova IVI Fertility, Behind Xavier’s Ladies Hostel, 108, Swastik Society Rd, Navrangpura, Ahmedabad, 390009 Gujarat India
| | - Sarita S. Gupta
- Dr. Vikram Sarabhai Institute of Cell and Molecular Biology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat 390 002 India
| |
Collapse
|
34
|
Jiang XD, Liu Y, Wu JF, Gong SN, Ma Y, Zi XD. Regulation of proliferation, apoptosis, hormone secretion and gene expression by acetyl-L-carnitine in yak (Bos grunniens) granulosa cells. Theriogenology 2023; 203:61-68. [PMID: 36972666 DOI: 10.1016/j.theriogenology.2023.03.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 03/13/2023] [Accepted: 03/14/2023] [Indexed: 03/17/2023]
Abstract
Supplementation with acetyl-l-carnitine (ALC) during in vitro maturation significantly improves the rates of oocyte cleavage and morula and blastocyst formation in sheep and buffalo; however, the mode of action of ALC in improving oocyte competence is not completely understood. Therefore, the aim of this study was to investigate the effects of ALC on proliferation, antioxidant properties, lipid droplet accumulation and steroid hormone secretion in yak (Bos grunniens) granulosa cells (GCs). Yak GCs were identified using FSHR immunofluorescence. The cells were treated with different concentrations of ALC, cell proliferation was detected by cell counting kit-8, and the optimal concentration and treatment time were determined for subsequent experiments. Then, reactive oxygen species (ROS) were detected by a DCFH-DA probe, and lipid droplet accumulation was observed by oil red O staining. Estradiol (E2) and progesterone (P4) in the medium were detected by ELISA, and the expression of genes related to cell proliferation, apoptosis, the cell cycle, antioxidants and steroid synthesis was determined by RT‒qPCR. The results showed that 1 mM ALC treatment for 48 h was the optimum treatment. It significantly increased cell viability (P < 0.05), significantly decreased the amount of ROS and lipid droplet content, and promoted P4 and E2 secretion (P < 0.05) of yak GCs. RT‒qPCR results verified that GCs treated with 1 mM ALC for 48 h significantly increased the expression of genes related to anti-apoptosis and the cell cycle (BCL-2, PCNA, CCND1 and CCNB1), antioxidants (CAT, SOD2 and GPX1), and E2 and P4 secretion (StAR, CYP19A1 and HSD3B1) (P < 0.05), but it significantly decreased the expression of apoptosis genes (BAX and P53) (P < 0.05). In conclusion, ALC increased the viability of yak GCs, reduced the amount of ROS and lipid droplets, increased P4 and E2 synthesis and affected the expression of related genes in yak GCs.
Collapse
|
35
|
Romeo M, Spaggiari G, Nuzzo F, Granata ARM, Simoni M, Santi D. Follicle-stimulating hormone effectiveness in male idiopathic infertility: What happens in daily practice? Andrology 2023; 11:478-488. [PMID: 36424882 DOI: 10.1111/andr.13353] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 11/07/2022] [Accepted: 11/20/2022] [Indexed: 11/27/2022]
Abstract
OBJECTIVES To assess the effectiveness of follicle-stimulating hormone (FSH) administration in male idiopathic infertility in a clinical setting. METHODS A retrospective real-world study was carried out, including all consecutive FSH-treated infertile men attending the Andrology Unit of Modena (Italy) from June 2015 to May 2022. Medical history, physical and andrological examinations, hormonal and seminal parameters, therapeutic management and pregnancy data were collected. The primary endpoint was the number of pregnancies obtained after FSH administration, whereas semen parameters change was the secondary outcome. RESULTS A total of 194 of 362 (53.6%) infertile men, eligible according to the Italian Health System regulations, were treated with FSH (mean age 37.9 ± 6.1 years). Following FSH administration (mean therapy duration 9.1 ± 7.1 months), 43 pregnancies were recorded (27.6%), of which 22 occurred naturally and 21 after assisted reproduction. A significant increase in sperm concentration (9.9 ± 12.2 vs. 18.9 ± 38.9 million/mL, p = 0.045) was detected after treatment, together with a significant increase in normozoospermia (from 1.0% to 5.1%, p = .044) and a reduction in azoospermia rate (from 9.8% to 7%, p = 0.044). Dividing the cohort in FSH-responders and non-responders, in terms of pregnancy achieved, higher sperm concentrations (15.7 ± 26.6 vs. 22.2 ± 25.7 million/mL, p = 0.033) and progressive sperm motility (18.0 ± 18.2 vs. 27.3 ± 11.3, p = 0.044) were found in pregnancy group. CONCLUSION Our experience suggests that FSH, empirically administered to men with idiopathic infertility, leads to pregnancy in one out of four patients and increases sperm concentration. Although the expected limits because of a real-world data study, the number of FSH-treated patients required to achieve one pregnancy seems to be lower in clinical setting if compared to previously published data.
Collapse
Affiliation(s)
- Marilina Romeo
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy.,Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero - Universitaria of Modena, Modena, Italy
| | - Giorgia Spaggiari
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero - Universitaria of Modena, Modena, Italy.,Unit of Andrology and Sexual Medicine of the Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero - Universitaria of Modena, Modena, Italy
| | - Federico Nuzzo
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Antonio R M Granata
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero - Universitaria of Modena, Modena, Italy.,Unit of Andrology and Sexual Medicine of the Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero - Universitaria of Modena, Modena, Italy
| | - Manuela Simoni
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy.,Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero - Universitaria of Modena, Modena, Italy.,Unit of Andrology and Sexual Medicine of the Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero - Universitaria of Modena, Modena, Italy
| | - Daniele Santi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy.,Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero - Universitaria of Modena, Modena, Italy.,Unit of Andrology and Sexual Medicine of the Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero - Universitaria of Modena, Modena, Italy
| |
Collapse
|
36
|
Yamamoto N, Takeuchi H, Yamaoka M, Nakanishi T, Tonai S, Nishimura R, Morita T, Nagano M, Kameda S, Genda K, Kawase J, Yamashita Y. Lipopolysaccharide (LPS) suppresses follicle development marker expression and enhances cytokine expressions, which results in fail to granulosa cell proliferation in developing follicle in cows. Reprod Biol 2023; 23:100710. [PMID: 36470010 DOI: 10.1016/j.repbio.2022.100710] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 10/18/2022] [Accepted: 11/10/2022] [Indexed: 12/12/2022]
Abstract
Postpartum endometritis is known to be associated with ovarian dysfunction in cows. Lipopolysaccharide (LPS) generated by Gram-negative bacteria is recognized by toll-like receptor 4 (TLR4), which leads to an inflammatory response by the generation of cytokines such as tumor necrosis factor-α (TNF-α) and interleukins. In this study, we investigated the effect of endometrial LPS on granulosa cell functions during early follicular development in cows. Uteri and follicles were obtained from a slaughterhouse and classified into either clinical endometritis (CE) or normal groups by vaginal mucus test. TLR4 mRNA and protein in normal cows were expressed in granulosa cells collected from follicles measuring 1-3 and 4-7 mm in a diameter, respectively. LPS content in endometrium and follicular fluid of CE cows was significantly higher than that in normal cows. Compared to normal cows, CE cows showed lower expression of follicular development markers (FSHR, CYP19A1, CCND2, and LHCGR) in granulosa cells, lower estradiol-17β concentrations in follicular fluid, and lower granulosa cell proliferation. CE contraction significantly increased cytokine expressions (TNF, IL-1A, and IL-1B) in granulosa cells and suppressed apoptosis of granulosa cells compared to normal cows. LPS significantly suppressed the expression of follicular development markers and the production of estradiol-17β in granulosa cells and reduced granulosa cells proliferation compared to cells cultured without LPS. LPS significantly increased cytokine expressions and suppressed granulosa cell apoptosis. Thus, the present results suggest that the existence of LPS in developing follicles is one of the causes of ovarian quiescence in cows.
Collapse
Affiliation(s)
- Naoki Yamamoto
- Shimane Meat Sanitation Inspection Center, Shimane Prefecture, 1677-2 Asahiyama-cho Senyama, Ooda 699-2212, Japan
| | - Himeno Takeuchi
- Laboratory of Animal Reproductive Physiology, Department of Bioresource Sciences, Prefectural University of Hiroshima, 5562 Nanatuka-cho, Shobara 727-0023, Japan
| | - Manami Yamaoka
- Laboratory of Animal Reproductive Physiology, Graduate School of Comprehensive Scientific Research, Prefectural University of Hiroshima, 5562 Nanatuka-cho, Shobara 727-0023, Japan
| | - Tomoya Nakanishi
- Laboratory of Animal Reproductive Physiology, Graduate School of Comprehensive Scientific Research, Prefectural University of Hiroshima, 5562 Nanatuka-cho, Shobara 727-0023, Japan
| | - Shingo Tonai
- Laboratory of Animal Reproductive Physiology, Graduate School of Comprehensive Scientific Research, Prefectural University of Hiroshima, 5562 Nanatuka-cho, Shobara 727-0023, Japan
| | - Ryo Nishimura
- Laboratory of Theriogenology, Joint Department of Veterinary Medicine, Faculty of Agriculture, Tottori University, 4-101 Koyama-cho Minami, Tottori 680-8553, Japan
| | - Takehito Morita
- Laboratory of Veterinary Pathology, Joint Department of Veterinary Medicine, Faculty of Agriculture, Tottori University, 4-101 Koyama-cho Minami, Tottori 680-8553, Japan
| | - Masashi Nagano
- Laboratory of Animal Reproduction, Department of Animal Science, School of Veterinary Medicine, Kitasato University, Towada 034-8628, Japan
| | - Shingo Kameda
- Shimane Meat Sanitation Inspection Center, Shimane Prefecture, 1677-2 Asahiyama-cho Senyama, Ooda 699-2212, Japan
| | - Kaori Genda
- Shimane Meat Sanitation Inspection Center, Shimane Prefecture, 1677-2 Asahiyama-cho Senyama, Ooda 699-2212, Japan
| | - Jun Kawase
- Shimane Prefectural Institute of Public Health and Environment Science, 582-1 Nishihamasada-cho, Matsue, Shimane 690-0122, Japan
| | - Yasuhisa Yamashita
- Laboratory of Animal Reproductive Physiology, Department of Bioresource Sciences, Prefectural University of Hiroshima, 5562 Nanatuka-cho, Shobara 727-0023, Japan; Laboratory of Animal Reproductive Physiology, Graduate School of Comprehensive Scientific Research, Prefectural University of Hiroshima, 5562 Nanatuka-cho, Shobara 727-0023, Japan.
| |
Collapse
|
37
|
Su J, Yi S, Gao Z, Abbas K, Zhou X. DNA methylation mediates gonadal development via regulating the expression levels of cyp19a1a in loach Misgurnus anguillicaudatus. Int J Biol Macromol 2023; 235:123794. [PMID: 36828090 DOI: 10.1016/j.ijbiomac.2023.123794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 02/14/2023] [Accepted: 02/17/2023] [Indexed: 02/24/2023]
Abstract
DNA methylation-mediated transcriptional regulation has been considered to significantly impact some steroidogenic enzyme genes expression. To uncover the roles of DNA methylation on the regulation of aromatase gene expression during gametogenesis in Misgurnus anguillicaudatus, the expression profiles and cellular localization of cyp19a1a and cyp19a1b were analyzed, and the landscape of DNA methylation dynamics was investigated. We found that cyp19a1a was predominantly expressed in granulosa cells of oocytes, while cyp19a1b expression was enriched in radial glial cells of the forebrain. In ovary, cyp19a1a was highly expressed until the vitellogenesis stage. The average methylation levels, especially for two CpG sites within the cAMP response element, were negatively correlated with cyp19a1a expression levels, indicating that methylation could regulate cyp19a1a transcriptional activity by modulating the binding efficiency of cAMP to its response elements. Compared with in ovary, cyp19a1a showed lower expression in testis but was hypermethylated. Cyp19a1b in female brain weakly expressed before the vitellogenesis stage, but significantly elevated at the maturation stage. In both sexes, it maintained high methylation levels in brain despite the obvious fluctuation of the cyp19a1b expression. This study revealed that DNA methylation plays a key role in establishing cyp19a1a spatiotemporal expression patterns and thus mediates gonadal development in teleosts.
Collapse
Affiliation(s)
- Junxiao Su
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China.
| | - Shaokui Yi
- College of Life Sciences, Huzhou University, Huzhou 313000, China.
| | - Zexia Gao
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China.
| | - Khalid Abbas
- Aquaculture Biotechnology Lab, Department of Zoology, Wildlife and Fisheries, University of Agriculture, Faisalabad, Pakistan.
| | - Xiaoyun Zhou
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
38
|
A Cross-Species Analysis Reveals Dysthyroidism of the Ovaries as a Common Trait of Premature Ovarian Aging. Int J Mol Sci 2023; 24:ijms24033054. [PMID: 36769379 PMCID: PMC9918015 DOI: 10.3390/ijms24033054] [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: 12/06/2022] [Revised: 01/21/2023] [Accepted: 01/31/2023] [Indexed: 02/09/2023] Open
Abstract
Although the imbalance of circulating levels of Thyroid Hormones (THs) affects female fertility in vertebrates, its involvement in the promotion of Premature Ovarian Aging (POA) is debated. Therefore, altered synthesis of THs in both thyroid and ovary can be a trait of POA. We investigated the relationship between abnormal TH signaling, dysthyroidism, and POA in evolutionary distant vertebrates: from zebrafish to humans. Ovarian T3 signaling/metabolism was evaluated by measuring T3 levels, T3 responsive transcript, and protein levels along with transcripts governing T3 availability (deiodinases) and signaling (TH receptors) in distinct models of POA depending on genetic background and environmental exposures (e.g., diets, pesticides). Expression levels of well-known (Amh, Gdf9, and Inhibins) and novel (miR143/145 and Gas5) biomarkers of POA were assessed. Ovarian dysthyroidism was slightly influenced by genetics since very few differences were found between C57BL/6J and FVB/NJ females. However, diets exacerbated it in a strain-dependent manner. Similar findings were observed in zebrafish and mouse models of POA induced by developmental and long-life exposure to low-dose chlorpyrifos (CPF). Lastly, the T3 decrease in follicular fluids from women affected by diminished ovarian reserve, as well as of the transcripts modulating T3 signaling/availability in the cumulus cells, confirmed ovarian dysthyroidism as a common and evolutionary conserved trait of POA.
Collapse
|
39
|
Liu W, Chen M, Liu C, Wang L, Wei H, Zhang R, Ren Z, Chen Y, Luo M, Zhao J, Jiang H, Gao F, Li W. Epg5 deficiency leads to primary ovarian insufficiency due to WT1 accumulation in mouse granulosa cells. Autophagy 2023; 19:644-659. [PMID: 35786405 PMCID: PMC9851269 DOI: 10.1080/15548627.2022.2094671] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Primary ovarian insufficiency (POI), also known as premature ovarian failure, is an ovarian defect in humans characterized by the premature depletion of ovarian follicles before the age of 40. However, the mechanisms underlying POI remain largely unknown. Here, we show that knockout of Epg5 (ectopic P-granules autophagy protein 5 homolog (C. elegans)) results in subfertility in female mice, which exhibit a POI-like phenotype. Single-cell RNA sequencing analysis revealed that the knockout of Epg5 affected the differentiation of granulosa cells (GCs). Further investigation demonstrated that knockout of Epg5 blocks macroautophagic/autophagic flux, resulting in the accumulation of WT1 (WT1 transcription factor), an essential transcription factor for GCs, suggesting WT1 needs to be selectively degraded by the autophagy pathway. We found that the insufficient degradation of WT1 in the antral follicular stage contributes to reduced expression of steroidogenesis-related genes, thereby disrupting GC differentiation. Collectively, our studies show that EPG5 promotes WT1 degradation in GCs, indicating that the dysregulation of Epg5 in GCs can trigger POI pathogenesis.Abbreviations: 3-MA, 3-methyladenine; CHX, cycloheximide; CQ, chloroquine; EPG5, ectopic P-granules autophagy protein 5 homolog (C. elegans); GC, granulosa cell; MAP1LC3/LC3, microtubule-associated protein 1 light chain 3; MII, metaphase II; POI, primary ovarian insufficiency; PB1, polar body 1; SQSTM1/p62, sequestosome 1; WT1, WT1 transcription factor.
Collapse
Affiliation(s)
- Wenwen Liu
- Institute of Reproductive Health and Perinatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, P.R. China,College of Life Sciences, University of Science and Technology of China, Hefei, P.R. China,State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, P.R. China
| | - Min Chen
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, P.R. China,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P.R. China
| | - Chao Liu
- Institute of Reproductive Health and Perinatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, P.R. China,State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, P.R. China
| | - Liying Wang
- Institute of Reproductive Health and Perinatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, P.R. China,State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, P.R. China
| | - Huafang Wei
- Institute of Reproductive Health and Perinatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, P.R. China,State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, P.R. China
| | - Ruidan Zhang
- Institute of Reproductive Health and Perinatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, P.R. China,State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, P.R. China
| | - Zhengxing Ren
- Institute of Reproductive Health and Perinatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, P.R. China,State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, P.R. China
| | - Yinghong Chen
- Institute of Reproductive Health and Perinatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, P.R. China,State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, P.R. China
| | - Mengcheng Luo
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, P.R China
| | - Jianguo Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, P.R. China,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P.R. China
| | - Hongwei Jiang
- Department of Endocrinology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, P.R. China,National Center for Clinical Research of Metabolic Diseases, Luoyang Center for Endocrinology and Metabolism, Luoyang, P.R. China,CONTACT Hongwei Jiang Department of Endocrinology, The First Affiliated Hospital and Clinical Medicine College, Henan University of Science and Technology, Luoyang471003, P.R. China
| | - Fei Gao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, P.R. China,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P.R. China,Fei Gao State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing100101, P.R. China
| | - Wei Li
- Institute of Reproductive Health and Perinatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, P.R. China,State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, P.R. China,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P.R. China,Wei Li Institute of Reproductive Health and Perinatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou510623, P.R. China
| |
Collapse
|
40
|
Immunoendocrine abnormalities in the female reproductive system, and lung steroidogenesis during experimental pulmonary tuberculosis. Tuberculosis (Edinb) 2023; 138:102274. [PMID: 36463716 DOI: 10.1016/j.tube.2022.102274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 11/10/2022] [Accepted: 11/12/2022] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Tuberculosis (TB) caused by Mycobacterium tuberculosis mainly affects the lungs, but can spread to other organs. TB chronically activates the immune and endocrine systems producing remarkable functional changes.So far, it is unknown whether pulmonary non-disseminated TB cause changes in the female reproductive system and lung endocrinology. OBJECTIVE To investigate whether pulmonary TB produces immunoendocrine alterations of the female mice reproductive organs, and lung estradiol synthesis. METHODS BALB/c mice were infected intratracheally with Mycobacterium tuberculosis (Mtb) strain H37Rv. Groups of six non-infected and infected animals were euthanized on different days. Bacillary loads were determined in the lungs, ovaries and uterus. Immunohistochemistry and morphometry studies were performed in histological sections. Serum estradiol wasassayed, and supernatantfrom cultured lung cells was analyzed by Thin Layer Chromatography (TLC). RESULTS Mtb only grew in lung tissue. Histopathology revealed abnormal folliculogenesis and decreased corpora lutea. Altered ovarian expression of IL-6, IL-1β was found. The infection increased serum estradiol. Estradiol synthesis by infected lung cells triplicate after 30 pi days.Aromatase immunostaining was found in the alveolar and bronchial epithelium, being stronger in the infected lungs, mainly in macrophages. CONCLUSION Pulmonary TB affects the histophysiology of the female reproductive system in absence of its local infection, and disturbslung endocrinology.
Collapse
|
41
|
Devillers MM, François CM, Chester M, Corre R, Cluzet V, Giton F, Cohen-Tannoudji J, Guigon CJ. Androgen receptor signaling regulates follicular growth and steroidogenesis in interaction with gonadotropins in the ovary during mini-puberty in mice. Front Endocrinol (Lausanne) 2023; 14:1130681. [PMID: 37152943 PMCID: PMC10154677 DOI: 10.3389/fendo.2023.1130681] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 04/04/2023] [Indexed: 05/09/2023] Open
Abstract
In females, androgens contribute to ovarian diseases such as polycystic ovarian syndrome (PCOS), but their action is also crucial for ovarian physiology, i.e., follicular growth and estradiol (E2) synthesis during reproductive life, in interaction with the gonadotropins LH and FSH. However, it is unclear whether androgens already play a role in the ovary at mini-puberty, a phase of postnatal development with active follicular growth and high E2 levels. Therefore, we analyzed the potential actions of androgens on the ovary and their possible interaction with gonadotropins during this period in mice. We used molecular-based studies and pharmacological approaches in vivo and on cultured ovaries. We found that mini-pubertal ovaries produce significant amounts of testosterone and display androgen receptor (AR) expression in growing follicles, both under the control of LH. By blocking AR signaling either in vivo or in ovarian cultures, we found that this pathway may participate in the regulation of prepubertal E2 synthesis and follicular growth, possibly by regulating the expression of a number of key intra-ovarian regulators, including FSH receptor (Fshr), the aromatase enzyme converting androgens into estrogens (Cyp19a1) and the cell cycle inhibitor p27KIP1 (Cdkn1b). We further showed that AR may stimulate FSH-mediated regulation of Cyp19a1 through its action on Fshr mRNA abundance. Overall, this work supports the idea that AR signaling is already activated in mini-pubertal ovaries to regulate E2 synthesis and follicular growth, at the interplay with LH and FSH signaling. Its early action may, thus, contribute to the implementation of early ovarian function with possible impacts on reproductive function.
Collapse
Affiliation(s)
- Marie M. Devillers
- Université Paris-Cité, CNRS, Inserm, Biologie Fonctionnelle et Adaptative, Paris, France
| | - Charlotte M. François
- Université Paris-Cité, CNRS, Inserm, Biologie Fonctionnelle et Adaptative, Paris, France
| | - Mélanie Chester
- Université Paris-Cité, CNRS, Inserm, Biologie Fonctionnelle et Adaptative, Paris, France
| | - Raphaël Corre
- Université Paris-Cité, CNRS, Inserm, Biologie Fonctionnelle et Adaptative, Paris, France
| | - Victoria Cluzet
- Université Paris-Cité, CNRS, Inserm, Biologie Fonctionnelle et Adaptative, Paris, France
| | - Frank Giton
- AP-HP, Pôle biologie-Pathologie Henri Mondor, Inserm IMRB U955, Créteil, France
| | - Joëlle Cohen-Tannoudji
- Université Paris-Cité, CNRS, Inserm, Biologie Fonctionnelle et Adaptative, Paris, France
| | - Céline J. Guigon
- Université Paris-Cité, CNRS, Inserm, Biologie Fonctionnelle et Adaptative, Paris, France
- *Correspondence: Céline J. Guigon,
| |
Collapse
|
42
|
Selvaraj MK, Kaur J. Computational method for aromatase-related proteins using machine learning approach. PLoS One 2023; 18:e0283567. [PMID: 36989252 PMCID: PMC10057777 DOI: 10.1371/journal.pone.0283567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 03/12/2023] [Indexed: 03/30/2023] Open
Abstract
Human aromatase enzyme is a microsomal cytochrome P450 and catalyzes aromatization of androgens into estrogens during steroidogenesis. For breast cancer therapy, third-generation aromatase inhibitors (AIs) have proven to be effective; however patients acquire resistance to current AIs. Thus there is a need to predict aromatase-related proteins to develop efficacious AIs. A machine learning method was established to identify aromatase-related proteins using a five-fold cross validation technique. In this study, different SVM approach-based models were built using the following approaches like amino acid, dipeptide composition, hybrid and evolutionary profiles in the form of position-specific scoring matrix (PSSM); with maximum accuracy of 87.42%, 84.05%, 85.12%, and 92.02% respectively. Based on the primary sequence, the developed method is highly accurate to predict the aromatase-related proteins. Prediction scores graphs were developed using the known dataset to check the performance of the method. Based on the approach described above, a webserver for predicting aromatase-related proteins from primary sequence data was developed and implemented at https://bioinfo.imtech.res.in/servers/muthu/aromatase/home.html. We hope that the developed method will be useful for aromatase protein related research.
Collapse
Affiliation(s)
| | - Jasmeet Kaur
- Department of Biophysics, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| |
Collapse
|
43
|
Shi S, Hu Y, Song X, Huang L, Zhang L, Zhou X, Gao L, Pang W, Yang G, Chu G. Totipotency of miR-184 in porcine granulosa cells. Mol Cell Endocrinol 2022; 558:111765. [PMID: 36049599 DOI: 10.1016/j.mce.2022.111765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/21/2022] [Accepted: 08/24/2022] [Indexed: 12/15/2022]
Abstract
Estradiol (E2) synthesis, cell proliferation and the apoptosis of porcine granulosa cells (GCs) affect follicular growth and development. The miR-184 level in ovary tissues of Yorkshire × Landrace sows was significantly higher in high-yielding sows than that in low-yielding sows, which was the same as in Yorkshire sows. However, the roles of miR-184 on E2 granulosa cells (GCs) are still unclear. We found that miR-184 promoted E2 synthesis and proliferation but inhibited apoptosis in GCs by targeting nuclear receptor subfamily 1 group D member 1 (NR1D1), cyclin dependent kinase inhibitor 1A (P21,CDKN1A) and homeodomain interacting protein kinase 2 (HIPK2) respectively. These findings indicated that miR-184 is a novel key factor that regulates the physiological functions of GCs.
Collapse
Affiliation(s)
- Shengjie Shi
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, Yangling, 712100, China; Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Yamei Hu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, Yangling, 712100, China; Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Xiangrong Song
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, Yangling, 712100, China; Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Liang Huang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, Yangling, 712100, China; Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Lutong Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, Yangling, 712100, China; Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Xiaoge Zhou
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, Yangling, 712100, China; Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Lei Gao
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, Yangling, 712100, China; Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Weijun Pang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, Yangling, 712100, China; Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Gongshe Yang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, Yangling, 712100, China; Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China.
| | - Guiyan Chu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, Yangling, 712100, China; Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China.
| |
Collapse
|
44
|
Wei W, Komatsu K, Osuka S, Murase T, Bayasula B, Nakanishi N, Nakamura T, Goto M, Iwase A, Masubuchi S, Kajiyama H. Tamoxifen Activates Dormant Primordial Follicles in Mouse Ovaries. Reprod Sci 2022; 29:3404-3412. [PMID: 35212933 PMCID: PMC9734234 DOI: 10.1007/s43032-022-00896-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 02/16/2022] [Indexed: 12/14/2022]
Abstract
Our previous study found that 17β-estradiol (E2) suppresses primordial follicle activation and growth in cultured mouse ovaries. In this study, we administered tamoxifen, an estrogen receptor antagonist, into the abdominal cavity of mice to clarify the relationship between primordial follicle activation and the physiological concentration of E2 in mouse ovaries. The results showed that tamoxifen promoted primordial follicle activation. Administration of tamoxifen promoted degradation of the extracellular matrix surrounding primordial follicles in the ovaries. Furthermore, tamoxifen decreased the expression of stefin A, an inhibitor of cathepsins that digest some proteins and extracellular matrix, in the ovaries. Mechanical stress produced by the extracellular matrix reportedly suppresses the activation of primordial follicles. The collective results show that tamoxifen can promote primordial follicle activation through the degradation of the extracellular matrix surrounding primordial follicles. Our results indicate that E2 suppresses primordial follicle activation in vivo and that tamoxifen may be useful as a therapeutic agent against infertility.
Collapse
Affiliation(s)
- Wei Wei
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, Aichi, 466-8550, Japan
| | - Kouji Komatsu
- Department of Physiology, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan.
| | - Satoko Osuka
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, Aichi, 466-8550, Japan
- Department of Maternal and Perinatal Medicine, Nagoya University Hospital, 65 Tsurumai-Cho, Showa-Ku, Nagoya, Aichi, 466-8550, Japan
| | - Tomohiko Murase
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, Aichi, 466-8550, Japan
| | - Bayasula Bayasula
- Bell Research Center for Reproductive Health and Cancer, Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
| | - Natsuki Nakanishi
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, Aichi, 466-8550, Japan
| | - Tomoko Nakamura
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, Aichi, 466-8550, Japan
| | - Maki Goto
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, Aichi, 466-8550, Japan
| | - Akira Iwase
- Department of Obstetrics and Gynecology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, 371-8511, Japan
| | - Satoru Masubuchi
- Department of Physiology, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, Aichi, 466-8550, Japan
| |
Collapse
|
45
|
Kamal DAM, Ibrahim SF, Ugusman A, Zaid SSM, Mokhtar MH. Kelulut Honey Improves Folliculogenesis, Steroidogenic, and Aromatase Enzyme Profiles and Ovarian Histomorphology in Letrozole-Induced Polycystic Ovary Syndrome Rats. Nutrients 2022; 14:4364. [PMID: 36297046 PMCID: PMC9612175 DOI: 10.3390/nu14204364] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/09/2022] [Accepted: 10/14/2022] [Indexed: 11/16/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) has been linked to aberrant folliculogenesis and abnormalities in the aromatase enzyme (Cyp19a1) and the steroidogenic enzyme, 17-alpha-hydroxylase (Cyp17a1) expression. It has been demonstrated that Kelulut honey (KH) improves both female and male reproductive system anomalies in animal studies. Here, we examined the effects of isolated and combined KH, metformin, and clomiphene in improving folliculogenesis, aromatase, and steroidogenic enzyme profiles and ovarian histomorphology in letrozole-induced PCOS rats. Letrozole (1 mg/kg/day) was administered to female Sprague-Dawley (SD) rats for 21 days to induce PCOS. PCOS rats were subsequently divided into six experimental groups: untreated, treatment with metformin (500 mg/kg/day), clomiphene (2 mg/kg/day), KH (1 g/kg/day), combined KH (1 g/kg/day) and metformin (500 mg/kg/day), and combined KH (1 g/kg/day) and clomiphene (2 mg/kg/day). All treatments were given orally for 35 days. We found that KH was comparable with clomiphene and metformin in improving the expression of Cyp17a1 and Cyp19a1, apart from enhancing folliculogenesis both histologically and through the expression of folliculogenesis-related genes. Besides, the combination of KH with clomiphene was the most effective treatment in improving the ovarian histomorphology of PCOS rats. The effectiveness of KH in restoring altered folliculogenesis, steroidogenic, and aromatase enzyme profiles in PCOS warrants a future clinical trial to validate its therapeutic effect clinically.
Collapse
Affiliation(s)
- Datu Agasi Mohd Kamal
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu 88400, Malaysia
| | - Siti Fatimah Ibrahim
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Azizah Ugusman
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Siti Sarah Mohamad Zaid
- Department of Environment, Faculty of Forestry and Environment, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Mohd Helmy Mokhtar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
46
|
Chen P, Luo Q, Lin Y, Jin J, Hu KL, Wang F, Sun J, Chen R, Wei J, Chen G, Zhang D. Arsenic exposure during juvenile and puberty significantly affected reproductive system development of female SD rats. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 242:113857. [PMID: 35809398 DOI: 10.1016/j.ecoenv.2022.113857] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/29/2022] [Accepted: 07/03/2022] [Indexed: 06/15/2023]
Abstract
Infertility affects about 10-15% couples over the world, among which a large number of cases the underlying causes are still unclear. Recent studies suggest that environmental factors may play an important role in these idiopathic infertilities. Arsenic is a heavy metal found in drinking water over the world. Its effect on the development of female reproductive system at the environmental-relevant levels is still largely unknown. To test the hypothesis that arsenic exposure during juvenile and puberty may affect sex maturation and female reproductive system development, SD rats of 3 weeks of age were exposed to arsenic with environmental-relevant levels (0, 0.02, 0.2, or 2 mg/L, n = 16/group) through drinking water for about 44 days until the rats reached adulthood (65 days of age). Arsenic exposure significantly reduced the weights of both ovary and uterus without affecting the body weight. Also, arsenic exposure disturbed estrus cycles and reduced the numbers of primordial follicles and corpora lutea while increased atretic follicles. In addition, arsenic reduced serum levels of estradiol, progesterone and testosterone but increased LH and FSH levels in dose-dependent manners. QPCR and Western blot experiments indicated arsenic selectively down-regulated ovarian steroidogenic-related proteins FSHR, STAR, CYP17A1, HSD3B1 and CYP19A1 and signaling molecules PKA-ERK-JNK-cJUN, without affecting AKT and CREB. As about reproductive capacity, arsenic-exposed dams had smaller pups, reduced litter size and lower number of male pups without a change in female pups. In conclusion, juvenile and pubertal arsenic exposures at environmental-relevant levels significantly reduced reproductive functions and capacity by adult. Since the lowest effective dose is very close to the government safety standards, the relevancy of arsenic over exposure to reproductive defects in human deserves further study.
Collapse
Affiliation(s)
- Panpan Chen
- Key Laboratory of Women's Reproductive Health of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Qiong Luo
- Key Laboratory of Women's Reproductive Health of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yifeng Lin
- Key Laboratory of Women's Reproductive Health of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jiani Jin
- Key Laboratory of Women's Reproductive Health of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Kai-Lun Hu
- Key Laboratory of Women's Reproductive Health of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Feixia Wang
- Key Laboratory of Women's Reproductive Health of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jiwei Sun
- Key Laboratory of Women's Reproductive Health of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ruixue Chen
- Key Laboratory of Women's Reproductive Health of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Juan Wei
- Key Laboratory of Women's Reproductive Health of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Guangdi Chen
- Department of Public Health, and Department of Reproductive Endocrinology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dan Zhang
- Key Laboratory of Women's Reproductive Health of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
47
|
Mills G, Dahan MH. Gonadotropin releasing hormone (GnRH) antagonist administration to decrease the risk of ovarian hyperstimulation syndrome in GNRH agonist cycles triggered with human chorionic gonadotropin. Arch Gynecol Obstet 2022; 306:1731-1737. [PMID: 35932297 DOI: 10.1007/s00404-022-06717-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 07/18/2022] [Indexed: 12/01/2022]
Abstract
PURPOSE In Gonadotropin releasing hormone(GnRH) agonist IVF, after administration of human chorionic gonadotropin(HCG) triggering, there is a risk of ovarian hyperstimulation syndrome(OHSS). Few methods exist to prevent OHSS in these cases. Therefore, we investigated the use of a GnRH antagonist to decrease the risk of OHSS, due to its ability to decrease VEGF production and function. METHOD A retrospective cohort study of 171-IVF patients at risk for developing OHSS after a GnRH agonist cycle with HCG trigger was performed from 2011 to 2019. The patient population consisted of women with an unexpected exuberant response to stimulation based on ovarian reserve testing and were triggered with hCG. Women were converted to a freeze-all cycle and received either cabergoline 0.5 mg orally alone for 7 days from the collection(Group 1, n = 123) or received cabergoline 0.5 mg orally and ganirelix, 250 mcg SC for 7-10 days(Group 2, n = 48). RESULTS Group 1 had more cases of moderate and severe OHSS than group 2-(25% vs. 10% p = 0.03, and 52% vs. 25% p = 0.001 respectively). Group 1 reported more abdominal discomfort and bloating than group 2(91% vs. 65% p < 0.001) and the presence of free fluid was more frequent in group 1 than group 2(74% vs. 35% p < 0.001). Hemoconcentration and electrolyte disturbances were less severe in group 2 than in group 1 (p < 0.001 all cases). CONCLUSION In patients at high risk for developing OHSS after hCG trigger in a GnRH agonist cycle, the addition of GnRH antagonists in the luteal phase may reduce the risk of developing moderate and severe OHSS. The GnRH antagonist likely leads to more rapid luteolysis and down regulation of VEGF production and receptor response, thereby decreasing the hallmark increased vascular permeability.
Collapse
Affiliation(s)
- Ginevra Mills
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, MUHC Reproductive Center, McGill University, 888, Blvd. de Maisonneuve East, Suite 200, Montréal, QC, H2L 4S8, Canada
| | - Michael H Dahan
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, MUHC Reproductive Center, McGill University, 888, Blvd. de Maisonneuve East, Suite 200, Montréal, QC, H2L 4S8, Canada.
| |
Collapse
|
48
|
Li L, Zhang J, Lu C, Wang B, Guo J, Zhang H, Cui S. MicroRNA-7a2 Contributes to Estrogen Synthesis and Is Modulated by FSH via the JNK Signaling Pathway in Ovarian Granulosa Cells. Int J Mol Sci 2022; 23:ijms23158565. [PMID: 35955699 PMCID: PMC9369042 DOI: 10.3390/ijms23158565] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/28/2022] [Accepted: 07/30/2022] [Indexed: 11/22/2022] Open
Abstract
MicroRNA-7a2 (miR-7a2) plays fundamental roles in the female reproductive axis, and estrogen is indispensable for maintaining ovary function. However, the interaction between miR-7a2 and ovarian function is unclear. The present study aimed to determine whether and how miR-7a2 functions in estrogen synthesis. Firstly, the results verified that miR-7a was highly expressed in ovarian granulosa cells. The knockout (KO) of miR-7a2 caused infertility and abnormal ovarian function in mice. Concomitantly, the Cyp19a1 expression and estrogen synthesis were significantly inhibited, which was validated in primary granulosa cells. The mice transplanted with miR-7a2 KO ovaries showed similar results; however, estrogen supplementation reversed infertility. In the in vitro experiment, follicle-stimulating hormone (FSH) significantly improved the expression of miR-7a and Cyp19a1 and the synthesis of estrogen. However, the miR-7a2 KO markedly reversed the function of FSH. Also, FSH upregulated miR-7a by activating the (c-Jun N-terminal kinase) JNK signaling pathway. In addition, Golgi apparatus protein 1 (Glg1) was shown to be the target gene of miR-7a2. These findings indicated that miR-7a2 is essential for ovarian functions with respect to estrogen synthesis through the targeted inhibition of the expression of Glg1 and then promoting Cyp19a1 expression; the physiological process was positively regulated by FSH via the JNK signaling pathway in granulosa cells.
Collapse
Affiliation(s)
- Liuhui Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (L.L.); (C.L.); (B.W.); (J.G.); (H.Z.)
| | - Jinglin Zhang
- Institute of Reproduction and Metabolism, Yangzhou University, Yangzhou 225009, China;
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Chenyang Lu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (L.L.); (C.L.); (B.W.); (J.G.); (H.Z.)
| | - Bingjie Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (L.L.); (C.L.); (B.W.); (J.G.); (H.Z.)
| | - Jiajia Guo
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (L.L.); (C.L.); (B.W.); (J.G.); (H.Z.)
| | - Haitong Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (L.L.); (C.L.); (B.W.); (J.G.); (H.Z.)
| | - Sheng Cui
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (L.L.); (C.L.); (B.W.); (J.G.); (H.Z.)
- Institute of Reproduction and Metabolism, Yangzhou University, Yangzhou 225009, China;
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Correspondence:
| |
Collapse
|
49
|
Liu YN, Qin Y, Wu B, Peng H, Li M, Luo H, Liu LL. DNA Methylation in Polycystic Ovary Syndrome:Emerging Evidence and Challenges. Reprod Toxicol 2022; 111:11-19. [PMID: 35562068 DOI: 10.1016/j.reprotox.2022.04.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 04/20/2022] [Accepted: 04/29/2022] [Indexed: 12/09/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a disease related to reproductive endocrine abnormalities in women of reproductive age, often accompanied by metabolic diseases such as hyperandrogenemia, insulin resistance and dyslipidemia. However, the etiology and mechanism of PCOS are still unclear. In recent years, more and more studies have found that epigenetic factors play an important role in PCOS. DNA methylation is the most widely studied epigenetic modification. At present, changes of DNA methylation have been found in serum, ovarian, hypothalamus, skeletal muscle, adipose tissue of PCOS patients, and these changes are closely related to insulin resistance, lipid metabolism and follicular development of PCOS. Although the current research on DNA methylation in PCOS is not in-depth, it indicated up a good direction for future research on the etiology and mechanism of PCOS. This review discussed the relationship between DNA methylation and PCOS. It is expected to help accelerate the application of DNA methylation in the diagnosis and treatment of PCOS.
Collapse
Affiliation(s)
- Yan-Nan Liu
- Nursing School, Hunan University of Medicine, Huaihua 418000, Hunan, China
| | - Yi Qin
- Faculty of Nursing, Guangxi University of Chinese Medicine, Nanning,530200, Guangxi, China
| | - Bin Wu
- Nursing School, Hunan University of Medicine, Huaihua 418000, Hunan, China
| | - Hui Peng
- Nursing School, Hunan University of Medicine, Huaihua 418000, Hunan, China
| | - Ming Li
- School of Medicine, Hunan Provincial Key Laboratory for Synthetic Biology of Traditional Chinese Medicine,Hunan University of Medicine, Huaihua 418000, Hunan, China
| | - Hai Luo
- School of Medicine, Hunan Provincial Key Laboratory for Synthetic Biology of Traditional Chinese Medicine,Hunan University of Medicine, Huaihua 418000, Hunan, China.
| | - Lin-Lin Liu
- Faculty of Nursing, Guangxi University of Chinese Medicine, Nanning,530200, Guangxi, China.
| |
Collapse
|
50
|
Storman EM, Liu NJ, Gintzler AR. Relevance of c-Src and protein phosphatase 2A to aromatase activity: Evidence of an acute self-regulating oestrogenic signalling complex in rat central nervous system. J Neuroendocrinol 2022; 34:e13089. [PMID: 35043508 PMCID: PMC9038631 DOI: 10.1111/jne.13089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 12/30/2021] [Accepted: 12/30/2021] [Indexed: 12/27/2022]
Abstract
We previously reported that aromatase protein levels do not parallel aromatase enzyme activity. This suggests that oestrogenic signalling may be modulated via post-translational modification of aromatase protein. The tyrosine and serine phosphorylation state of aromatase are known to influence its activity. To investigate the possible relevance of aromatase phosphorylation to the incongruity observed between aromatase protein and its activity, we explored interactions between aromatase and the tyrosine kinase c-Src and the serine protein phosphatases 2A and 5 (PP2A and PP5), as well as the relationship between levels of tyrosine-phosphorylated aromatase and the extrapolated aromatase activity. We found that (a) hypothalamic aromatase was significantly more heavily tyrosine-phosphorylated than spinal aromatase; (b) aromatase was oligomerized with c-Src and PP2A/PP5, potentially activating aromatase via tyrosine-phosphorylation and serine-dephosphorylation; (c) the associations of c-Src and PP2A/PP5 with hypothalamic aromatase were substantially greater than with spinal aromatase; and (d) aromatase, oestrogen receptor α, PP2A, and c-Src were present in a common membrane oligomer. The existence of c-Src and PP2A in an oligomer that also contains aromatase and membrane oestrogen receptor α (and presumably other signalling molecules) indicates the presence in the CNS of a potentially self-regulating oestrogenic signalling unit. The degree to which such a complex operates autonomously and the regulatory factors thereof are likely to have substantial physiological implications and clinical relevance.
Collapse
Affiliation(s)
- Emiliya M Storman
- Department of Obstetrics and Gynecology, State University of New York, Downstate Health Sciences University, Brooklyn, New York, USA
| | - Nai-Jiang Liu
- Department of Obstetrics and Gynecology, State University of New York, Downstate Health Sciences University, Brooklyn, New York, USA
| | - Alan R Gintzler
- Department of Obstetrics and Gynecology, State University of New York, Downstate Health Sciences University, Brooklyn, New York, USA
| |
Collapse
|