1
|
Liu L, Hao M, Zhang J, Chen Z, Zhou J, Wang C, Zhang H, Wang J. FSHR-mTOR-HIF1 signaling alleviates mouse follicles from AMPK-induced atresia. Cell Rep 2023; 42:113158. [PMID: 37733588 DOI: 10.1016/j.celrep.2023.113158] [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: 11/21/2022] [Revised: 07/24/2023] [Accepted: 09/05/2023] [Indexed: 09/23/2023] Open
Abstract
The majority of activated ovarian follicles undergo atresia during reproductive life in mammals, and only a small number of follicles are ovulated. Though hormone treatment has been widely used to promote folliculogenesis, the molecular mechanism behind follicle selection and atresia remains under debate due to inconsistency among investigation models. Using a high-throughput molecular pathology strategy, we depicted a transcriptional atlas of mouse follicular granulosa cells (GCs) under physiological condition and obtained molecular signatures in healthy and atresia GCs during development. Functional results revealed hypoxia-inducible factor 1 (HIF1) as a major effector downstream of follicle-stimulating hormone (FSH), and HIF1 activation is essential for follicle growth. Energy shortage leads to prevalent AMP-activated protein kinase (AMPK) activation and drives follicular atresia. FSHR-mTOR-HIF1 signaling helps follicles escape from the atresia fate, while energy stress persists. Our work provides a comprehensive understanding of the molecular network behind follicle selection and atresia under physiological condition.
Collapse
Affiliation(s)
- Longping Liu
- School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Ming Hao
- School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Jianyun Zhang
- Department of Oral Pathology, Peking University School, Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials, Digital Medical Devices, Beijing 100081, P.R. China
| | - Ziqi Chen
- State Key Laboratory of Farm Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Jiaqi Zhou
- State Key Laboratory of Farm Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Chao Wang
- State Key Laboratory of Farm Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Hua Zhang
- State Key Laboratory of Farm Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Jianbin Wang
- School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
2
|
Lei Z, Ali I, Yang M, Yang C, Li Y, Li L. Non-Esterified Fatty Acid-Induced Apoptosis in Bovine Granulosa Cells via ROS-Activated PI3K/AKT/FoxO1 Pathway. Antioxidants (Basel) 2023; 12:antiox12020434. [PMID: 36829992 PMCID: PMC9952034 DOI: 10.3390/antiox12020434] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 02/05/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
Non-esterified fatty acid (NEFA), one of negative energy balance (NEB)'s most well-known products, has a significant impact on cows' reproductive potential. Our study used an in vitro model to investigate the deleterious effects of NEFA on bovine granulosa cells (BGCs) and its underlying molecular mechanism. The results showed that high levels of NEFA led to the accumulation of reactive oxygen species (ROS), increased the expression of apoptosis-related factors such as Bcl2-Associated X/B-cell lymphoma-2 (Bax/Bcl-2) and Caspase-3, and down-regulated steroid synthesis-related genes such as sterol regulatory element binding protein 1 (SREBP-1), cytochrome P450c17 (CYP17), and cytochrome P450 aromatase (CYP19), to promote oxidative stress, cell apoptosis, and steroid hormone synthesis disorders in BGCs. In addition, NEFA significantly inhibited phosphatidylinositol 3-kinase (PI3K) and phosphorylated protein kinase B (p-AKT) activity and increased forkhead box O1 (FoxO1) expression. To further explore the role of the PI3K/AKT/FoxO1 signaling pathway in NEFA, we found that pretreatment with AKT-specific activator SC79 (5 mg/mL) for 2 h or transfection with FoxO1 knockdown siRNA in BGCs could alleviate the negative effects of NEFA treatment by decreasing Bax/Bcl-2 ratio and Caspase-3 expression, and upregulating SREBP-1, CYP17, and CYP19 expression. Meanwhile, SC79 significantly inhibited NEFA-induced dephosphorylation and massive nuclear translocation of FoxO1. Taken together, the NEFA induced oxidative stress, apoptosis, and steroid hormone synthesis disorders in BGCs by inhibiting the PI3K/AKT pathway that regulates FoxO1 phosphorylation and nuclear translocation. Our findings help to clarify the molecular mechanisms underlying the negative effects of high levels of NEFA on BGCs.
Collapse
Affiliation(s)
| | | | | | | | | | - Lian Li
- Correspondence: ; Tel.: +86-25-84395314
| |
Collapse
|
3
|
Aslanian-Kalkhoran L, Esparvarinha M, Nickho H, Aghebati-Maleki L, Heris JA, Danaii S, Yousefi M. Understanding main pregnancy complications through animal models. J Reprod Immunol 2022; 153:103676. [PMID: 35914401 DOI: 10.1016/j.jri.2022.103676] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/27/2022] [Accepted: 07/21/2022] [Indexed: 10/16/2022]
Abstract
Since human pregnancy is an inefficient process, achieving desired and pleasant outcome of pregnancy - the birth of a healthy and fit baby - is the main goal in any pregnancy. Spontaneous pregnancy failure is actually the most common complication of pregnancy and Most of these pregnancy losses are not known. Animal models have been utilized widely to investigate the system of natural biological adaptation to pregnancy along with increasing our comprehension of the most important hereditary and non-hereditary factors that contribute to pregnancy disorders. We use model organisms because their complexity better reproduces the human condition. A useful animal model for the disease should be pathologically similar to the disease conditions in humans. Animal models deserve a place in research because of the ethical limitations that apply to pregnant women's experiments. The present review provides insights into the overall risk factors involved in recurrent miscarriage, recurrent implant failure and preeclampsia and animal models developed to help researchers identify the source of miscarriage and the best research and treatment strategy for women with Repeated miscarriage and implant failure.
Collapse
Affiliation(s)
- Lida Aslanian-Kalkhoran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran
| | - Mojgan Esparvarinha
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran
| | - Hamid Nickho
- Department of Immuunology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Islamic Republic of Iran; Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Leili Aghebati-Maleki
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran
| | - Javad Ahmadian Heris
- Department of Allergy and Clinical Immunology, Pediatric Hospital, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran
| | - Shahla Danaii
- Gynecology Department, Eastern Azerbaijan ACECR ART Centre, Eastern Azerbaijan Branch of ACECR, Tabriz, Islamic Republic of Iran
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran.
| |
Collapse
|
4
|
Sudhakaran G, Guru A, Hari Deva Muthu B, Murugan R, Arshad A, Arockiaraj J. Evidence-based hormonal, mutational, and endocrine-disrupting chemical-induced zebrafish as an alternative model to study PCOS condition similar to mammalian PCOS model. Life Sci 2022; 291:120276. [PMID: 34990650 DOI: 10.1016/j.lfs.2021.120276] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 12/15/2021] [Accepted: 12/24/2021] [Indexed: 12/13/2022]
Abstract
Polycystic ovarian syndrome (PCOS) causes swollen ovaries in women at reproductive age due to hormonal disorder with small cysts on the outer edges. The cause of the disorder is still yet to be found. Multiple factors have increased PCOS prevalence, hyperandrogenism, oxidative stress, inflammation, and insulin resistance. Various animal PCOS models have been developed to imitate the pathophysiology of PCOS in humans. Zebrafish is one of the most versatile animal experimental models because of the transparency of the embryos, small size, and rapid growth. The zebrafish similarity to higher vertebrates made it a useful non-mammalian model for PCOS drug testing and screening. This review provides an insight into the usage of zebrafish, a non-mammalian model for PCOS, as an opportunity for evaluating future initiatives in such a research domain.
Collapse
Affiliation(s)
- Gokul Sudhakaran
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chennai 603 203, Tamil Nadu, India
| | - Ajay Guru
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chennai 603 203, Tamil Nadu, India
| | - B Hari Deva Muthu
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chennai 603 203, Tamil Nadu, India
| | - Raghul Murugan
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chennai 603 203, Tamil Nadu, India
| | - Aziz Arshad
- International Institute of Aquaculture and Aquatic Sciences (I-AQUAS), Universiti Putra Malaysia, 71050 Port Dickson, Negeri Sembilan, Malaysia
| | - Jesu Arockiaraj
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chennai 603 203, Tamil Nadu, India; Foundation for Aquaculture Innovations and Technology Transfer (FAITT), Thoraipakkam, Chennai 600 097, Tamil Nadu, India.
| |
Collapse
|
5
|
Do trends of adiposity and metabolic parameters vary in women with different ovarian reserve status? A population-based cohort study. ACTA ACUST UNITED AC 2021; 27:684-692. [PMID: 32301892 DOI: 10.1097/gme.0000000000001513] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To investigate whether trends of adiposity and glucose metabolism parameters in women with low ovarian reserve status based on their anti-Mullerian hormone (AMH) levels differ from those with high ovarian reserve. METHODS In this population-based prospective study, eligible women, aged 20 to 50 years, were selected from among participants of the Tehran Lipid and Glucose Study (TLGS). Generalized estimating equation (GEE) models were applied to compare changes in various adiposity and metabolic parameters across time between women in the first and fourth quartiles of age-specific AMH, after adjustment for confounders. Pooled logistic regression was used to compare progression of prediabetes mellitus (pre-DM) and diabetes mellitus (DM) between the women of these two age-specific AMH quartiles. RESULTS In this study of a total of 1,015 participants and with a median follow-up of 16 years, we observed that over time, both groups of women in the first and fourth quartiles of age-specific AMH experienced significant positive trends in their adiposity indices including central obesity, waist-to-hip ratio (WHR), waist-to-height ratio (WHtR), a body shape index (ABSI), and a negative trend in visceral adiposity index (VAI), whereas there was no significant difference in these parameters between the two groups. This study revealed that odds ratios of diabetes and prediabetes in women in the first quartile of age-specific AMH were not significantly different, compared with those in the fourth quartile. CONCLUSION Women with lower ovarian reserve do not experience different over time trends of adiposity and glucose metabolism parameters during their reproductive life span.
Collapse
|
6
|
Ryu Y, Kim YJ, Kim YY, Kim J, Kim SW, Kim H, Ku SY. Consecutive Low Doses of Streptozotocin Induce Polycystic Ovary Syndrome Features in Mice. Int J Mol Sci 2021; 22:1299. [PMID: 33525535 PMCID: PMC7865447 DOI: 10.3390/ijms22031299] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/21/2021] [Accepted: 01/26/2021] [Indexed: 02/07/2023] Open
Abstract
Polycystic ovarian syndrome (PCOS) is a common reproductive endocrine disorder in reproductive-age women. Due to its various pathophysiological properties and clinical heterophenotypes, the mechanism of PCOS pathogenesis is still unclear. Several animal models have been used to study PCOS and allow the exploration of the specific mechanism underlying PCOS. We focused on streptozotocin (STZ) to develop a non-steroidal and non-diabetic PCOS model. We administered multiple STZ injections to female C57BL/6 mice (3-4 weeks old) at different concentrations: STZ-15 (15 mg/kg), STZ-30 (30 mg/kg), and STZ-60 (60 mg/kg) treatments. During the experimental period, we analyzed body weight, blood glucose levels, and estrous cycle pattern. Furthermore, five weeks after STZ administration, we examined hormone levels and the morphology of ovarian tissues. Mice in the STZ-15 group did not show differences in body weights, blood glucose level, insulin level, and insulin tolerance compared to wild-type and control groups whereas those in the STZ-60 group presented a typical diabetes phenotype. In the case of the STZ-30 group, only increased blood glucose level was observed. Total testosterone levels were significantly elevated in STZ-15 and STZ-30 groups. Luteinizing hormone (LH) and estradiol levels were not significantly changed in the STZ-treated groups. The number of ovarian antral follicles and atretic follicles significantly increased in the ovary of mice in the STZ-15 and STZ-30 groups. All STZ-treated groups manifested irregular estrus cycles. However, the patterns of estrous cycles were different between mice treated with different STZ concentrations. We found that PI3K-AKT and IRS-1 signaling in the ovary was enhanced by low doses of STZ treatment. Taken together, our finding indicates that multiple injections of STZ at low doses induce PCOS features in mice without induction of diabetes features.
Collapse
Affiliation(s)
- Youngjae Ryu
- Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Korea; (Y.R.); (Y.Y.K.)
| | - Yong Jin Kim
- Department of Obstetrics and Gynecology, Korea University College of Medicine, Seoul 02841, Korea;
| | - Yoon Young Kim
- Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Korea; (Y.R.); (Y.Y.K.)
- Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University College of Medicine, Seoul 03080, Korea;
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul 03080, Korea; (S.W.K.); (H.K.)
| | - Jungwoo Kim
- Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University College of Medicine, Seoul 03080, Korea;
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul 03080, Korea; (S.W.K.); (H.K.)
| | - Sung Woo Kim
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul 03080, Korea; (S.W.K.); (H.K.)
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Hoon Kim
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul 03080, Korea; (S.W.K.); (H.K.)
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Seung Yup Ku
- Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University College of Medicine, Seoul 03080, Korea;
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul 03080, Korea; (S.W.K.); (H.K.)
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
7
|
Rong Y, Mo Y, Liu Y, Deng Y, Hu S, Li L, Hu J, Hu B, He H, Wang J. MiR-181a-5p inhibits goose granulosa cell viability by targeting SIRT1. Br Poult Sci 2021; 62:373-378. [PMID: 33415990 DOI: 10.1080/00071668.2020.1870660] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
1. Granulosa cells (GCs) are involved in folliculogenesis, follicular development, and atresia. Previous studies have shown that microRNA-181a-5p (miR-181a-5p) and sirtuin 1 (SIRT1) are involved in GC proliferation and apoptosis, and SIRT1 has been predicted as one target of miR-181a-5p. However, there are few studies with poultry.2. Quantitative real-time PCR (qRT-PCR) was used to detect the expression level of miR-181a-5p in granulosa layers during geese ovarian follicular development. A methyl thiazolyl tetrazolium (MTT) assay was performed to assess the viability of geese granulosa cells treated with miR-181a-5p mimic or inhibitor. The binding sites between the SIRT1 3'-UTR region and miR-181a-5p were evaluated using a luciferase reporter assay system. SIRT1 mRNA levels were detected using qRT-PCR after transfection with miR-181a-5p mimic and inhibitor.3. The miR-181a-5p suppressed geese GC viability and regulated the mRNA expression of viability-related genes in geese GCs. SIRT1 was a target gene of miR-181a-5p and miR-181a-5p suppressed its mRNA expression.4. The miR-181a-5p may target and inhibit SIRT1 expression, thus suppressing GC viability by regulating viability-related key genes.
Collapse
Affiliation(s)
- Y Rong
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, P. R. China
| | - Y Mo
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, P. R. China
| | - Y Liu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, P. R. China
| | - Y Deng
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, P. R. China
| | - S Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, P. R. China
| | - L Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, P. R. China
| | - J Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, P. R. China
| | - B Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, P. R. China
| | - H He
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, P. R. China
| | - J Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, P. R. China
| |
Collapse
|
8
|
Huang Q, Liu Y, Yang Z, Xie Y, Mo Z. The Effects of Cholesterol Metabolism on Follicular Development and Ovarian Function. Curr Mol Med 2019; 19:719-730. [PMID: 31526349 DOI: 10.2174/1566524019666190916155004] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 08/21/2019] [Accepted: 09/03/2019] [Indexed: 12/23/2022]
Abstract
Cholesterol is an important substrate for the synthesis of ovarian sex hormones and has an important influence on follicular development. The cholesterol in follicular fluid is mainly derived from plasma. High-density lipoprotein (HDL) and lowdensity lipoprotein (LDL) play important roles in ovarian cholesterol transport. The knockout of related receptors in the mammalian HDL and LDL pathways results in the reduction or absence of fertility, leading us to support the importance of cholesterol homeostasis in the ovary. However, little is known about ovarian cholesterol metabolism and the complex regulation of its homeostasis. Here, we reviewed the cholesterol metabolism in the ovary and speculated that regardless of the functioning of cholesterol metabolism in the system or the ovarian microenvironment, an imbalance in cholesterol homeostasis is likely to have an adverse effect on ovarian structure and function.
Collapse
Affiliation(s)
- Qin Huang
- Clinical Anatomy & Reproductive Medicine Application Institute, Department of Histology and Embryology, Hengyang Medical school, University of South China, Hengyang 421001, China
| | - Yannan Liu
- Nursing School, Hunan University of Medicine, Huaihua 418000, China
| | - Zhen Yang
- Clinical Anatomy & Reproductive Medicine Application Institute, Department of Histology and Embryology, Hengyang Medical school, University of South China, Hengyang 421001, China
| | - Yuanjie Xie
- Clinical Anatomy & Reproductive Medicine Application Institute, Department of Histology and Embryology, Hengyang Medical school, University of South China, Hengyang 421001, China
| | - Zhongcheng Mo
- Clinical Anatomy & Reproductive Medicine Application Institute, Department of Histology and Embryology, Hengyang Medical school, University of South China, Hengyang 421001, China
| |
Collapse
|
9
|
Ryu Y, Kim SW, Kim YY, Ku SY. Animal Models for Human Polycystic Ovary Syndrome (PCOS) Focused on the Use of Indirect Hormonal Perturbations: A Review of the Literature. Int J Mol Sci 2019; 20:2720. [PMID: 31163591 PMCID: PMC6600358 DOI: 10.3390/ijms20112720] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 05/14/2019] [Accepted: 05/23/2019] [Indexed: 02/07/2023] Open
Abstract
Hormonal disturbances, such as hyperandrogenism, are considered important for developing polycystic ovary syndrome (PCOS) in humans. Accordingly, directly hormone-regulated animal models are widely used for studying PCOS, as they replicate several key PCOS features. However, the pathogenesis and treatment of PCOS are still unclear. In this review, we aimed to investigate animal PCOS models and PCOS-like phenotypes in animal experiments without direct hormonal interventions and determine the underlying mechanisms for a better understanding of PCOS. We summarized animal PCOS models that used indirect hormonal interventions and suggested or discussed pathogenesis of PCOS-like features in animals and PCOS-like phenotypes generated in other animals. We presented integrated physiological insights and shared cellular pathways underlying the pathogenesis of PCOS in reviewed animal models. Our review indicates that the hormonal and metabolic changes could be due to molecular dysregulations, such as upregulated PI3K-Akt and extracellular signal-regulated kinase (ERK) signalling, that potentially cause PCOS-like phenotypes in the animal models. This review will be helpful for considering alternative animal PCOS models to determine the cellular/molecular mechanisms underlying PCOS symptoms. The efforts to determine the specific cellular mechanisms of PCOS will contribute to novel treatments and control methods for this complex syndrome.
Collapse
Affiliation(s)
- Youngjae Ryu
- Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Korea.
| | - Sung Woo Kim
- Department of Obstetrics and Gynecology, College of Medicine, Seoul National University, Seoul 03080, Korea.
| | - Yoon Young Kim
- Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Korea.
- Department of Obstetrics and Gynecology, College of Medicine, Seoul National University, Seoul 03080, Korea.
| | - Seung-Yup Ku
- Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Korea.
- Department of Obstetrics and Gynecology, College of Medicine, Seoul National University, Seoul 03080, Korea.
| |
Collapse
|
10
|
El-Sayyad HIH, El-Shershaby EMF, El-Mansi AA, El-Ashry NE. Anti-hypercholesterolemic impacts of barley and date palm fruits on the ovary of Wistar albino rats and their offspring. Reprod Biol 2018; 18:236-251. [PMID: 30005909 DOI: 10.1016/j.repbio.2018.07.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 05/25/2018] [Accepted: 07/05/2018] [Indexed: 12/23/2022]
Abstract
A high cholesterol diet is related to ovarian dysfunction and infertility which has been increased among young ages consuming processed food products. The present study was conducted to evaluate the role of a high cholesterol diet on the ovaries of young female rats via assessments of histopathology, immunohistochemistry, oxidative stress and apoptic markers. Also, mating of hypercholesterolemic female rats was carried out to measure the fertility and numbers of their offspring. At the same time, phytotherapy was carried out through supplementing the diet with barley and/ or date palm fruits (10%) during the experiment to assess the phyto-therapeutic impacts in attenuation of drastic hypercholesterolemic effects. Hypercholesterolemic diet-fed rats exhibited damage of the ovarian follicles and increased follicular atresia. Furthermore, expression of cleaved caspase-3 was upregulated, while PCNA was downregulated in granulosa, theca and stroma cells. Hypercholesterolemic female rats showed marked depletion of antioxidative enzymes, increased lipid peroxidation and apoptotic markers. Alterations to the female serum hormones were detected. Offspring maternally fed on hypercholesterolemic diet showed a significant decrease of body weight and altered sex ratio. However, concomitant supplementation of barley and or date fruits to hypercholesterolemic groups revealed marked improvement of ovarian structure and function. On the basis of these evidences, it is believed that the enhanced synergistic effects of barley and/or date palm fruits in the amelioration of ovarian structure and functions were elicited by the potential antioxidant activity of their phytomicronutrients, polyphenols, β-glucan and trace elements. These materials scavenge free radicals from inflamed cells that can be used to establish an effective and novel therapeutic strategy for activating ovarian cell regeneration.
Collapse
Affiliation(s)
| | | | - Ahmed A El-Mansi
- Zoology Dept., Faculty of Science, Mansoura University, Mansoura, Egypt; Biology Dept., Faculty of Science, King Khalid University, Abha, Saudi Arabia.
| | | |
Collapse
|
11
|
Zhang M, Zhang Q, Hu Y, Xu L, Jiang Y, Zhang C, Ding L, Jiang R, Sun J, Sun H, Yan G. miR-181a increases FoxO1 acetylation and promotes granulosa cell apoptosis via SIRT1 downregulation. Cell Death Dis 2017; 8:e3088. [PMID: 28981116 PMCID: PMC5680589 DOI: 10.1038/cddis.2017.467] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 08/06/2017] [Accepted: 08/17/2017] [Indexed: 12/23/2022]
Abstract
Oxidative stress impairs follicular development by inducing granulosa cell (GC) apoptosis, which involves enhancement of the transcriptional activity of the pro-apoptotic factor Forkhead box O1 (FoxO1). However, the mechanism by which oxidative stress promotes FoxO1 activity is still unclear. Here, we found that miR-181a was upregulated in hydrogen peroxide (H2O2)-treated GCs and a 3-nitropropionic acid (NP)-induced in vivo model of ovarian oxidative stress. miR-181a overexpression promoted GC apoptosis, whereas knockdown of endogenous miR-181a blocked H2O2-induced cell apoptosis. Moreover, we identified that Sirtuin 1 (SIRT1), a deacetylase that suppresses FoxO1 acetylation in GCs, was downregulated by miR-181a and reversed the promoting effects of H2O2 and miR-181a on FoxO1 acetylation and GC apoptosis. Importantly, decreased miR-181a expression in the in vivo ovarian oxidative stress model inhibited apoptosis by upregulating SIRT1 expression and FoxO1 deacetylation. Together, our results suggest that miR-181a mediates oxidative stress-induced FoxO1 acetylation and GC apoptosis by targeting SIRT1 both in vitro and in vivo.
Collapse
Affiliation(s)
- Mei Zhang
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, People’s Republic of China
| | - Qun Zhang
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, People’s Republic of China
| | - Yali Hu
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, People’s Republic of China
| | - Lu Xu
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, People’s Republic of China
| | - Yue Jiang
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, People’s Republic of China
| | - Chunxue Zhang
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, People’s Republic of China
| | - Lijun Ding
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, People’s Republic of China
| | - Ruiwei Jiang
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, People’s Republic of China
| | - Jianxin Sun
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Haixiang Sun
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, People’s Republic of China
| | - Guijun Yan
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, People’s Republic of China
| |
Collapse
|
12
|
Bastu E, Zeybek U, Gurel Gurevin E, Yüksel Ozgor B, Celik F, Okumus N, Demiral I, Dural O, Celik C, Bulut H, Ilkay Armutak E, Baysal B, Buyru F, Yeh J. Effects of Irisin and Exercise on Metabolic Parameters and Reproductive Hormone Levels in High-Fat Diet-Induced Obese Female Mice. Reprod Sci 2017; 25:281-291. [DOI: 10.1177/1933719117711264] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Ercan Bastu
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Istanbul University School of Medicine, Istanbul, Turkey
| | - Umit Zeybek
- Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Ebru Gurel Gurevin
- Department of Biology, Faculty of Science, Istanbul University, Istanbul, Turkey
| | - Bahar Yüksel Ozgor
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Istanbul University School of Medicine, Istanbul, Turkey
| | - Faruk Celik
- Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Nazli Okumus
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Istanbul University School of Medicine, Istanbul, Turkey
| | - Irem Demiral
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Istanbul University School of Medicine, Istanbul, Turkey
| | - Ozlem Dural
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Istanbul University School of Medicine, Istanbul, Turkey
| | - Cem Celik
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Istanbul University School of Medicine, Istanbul, Turkey
| | - Huri Bulut
- Department of Biology, Faculty of Science, Istanbul University, Istanbul, Turkey
| | - Elif Ilkay Armutak
- Department of Histology and Embryology, Faculty of Veterinary, Istanbul University, Istanbul, Turkey
| | - Bulent Baysal
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Istanbul University School of Medicine, Istanbul, Turkey
| | - Faruk Buyru
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Istanbul University School of Medicine, Istanbul, Turkey
| | - John Yeh
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
13
|
Zhou J, Yao W, Liu K, Wen Q, Wu W, Liu H, Li Q. MicroRNA let-7g regulates mouse granulosa cell autophagy by targeting insulin-like growth factor 1 receptor. Int J Biochem Cell Biol 2016; 78:130-140. [PMID: 27417237 DOI: 10.1016/j.biocel.2016.07.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Revised: 07/05/2016] [Accepted: 07/11/2016] [Indexed: 01/07/2023]
Abstract
As an important type of somatic cell, granulosa cells play a major role in deciding the fate of follicles. Therefore, analyses of granulosa cell apoptosis and follicular atresia have become hotspots of animal research. Autophagy is a cellular catabolic mechanism that protects cells from stress conditions, including starvation, hypoxia, and accumulation of misfolded proteins. However, the relationship between autophagy and apoptosis in granulosa cells is not well known. Here, we demonstrate that let-7g regulates the mouse granulosa cell autophagy signaling pathway by inhibiting insulin-like growth factor 1 receptor expression and affecting the phosphorylation of protein kinase B/mammalian target of rapamycin. Small interference-mediated knockdown of insulin-like growth factor 1 receptor significantly promoted autophagy signaling of mouse granulosa cells. In contrast, overexpression of insulin-like growth factor 1 receptor in mouse granulosa cells attenuated autophagy activity in the presence of let-7g. In addition, overexpression of let-7g increased the apoptosis rate, as indicated by an increased number of terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling-positive cells. Finally, 3-methyladenine as well as the lysosomal enzyme inhibitor chloroquine partially blocked apoptosis. In summary, this study demonstrates that let-7g regulates autophagy in mouse granulosa cells by targeting insulin-like growth factor 1 receptor and downregulating protein kinase B/mammalian target of rapamycin signaling, and that mouse granulosa cell autophagy induced by let-7g participates in apoptosis.
Collapse
Affiliation(s)
- Jilong Zhou
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Wang Yao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Kaiqing Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Qiannan Wen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Wangjun Wu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Honglin Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| | - Qifa Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
14
|
Velázquez MA. [Impact of maternal overnutrition on the periconceptional period]. ACTA ACUST UNITED AC 2015; 62:246-53. [PMID: 25733194 DOI: 10.1016/j.endonu.2015.01.004] [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: 12/03/2014] [Revised: 01/13/2015] [Accepted: 01/16/2015] [Indexed: 10/23/2022]
Abstract
Overnutrition may lead to obesity. Maternal obesity may affect fertility not only via anovulation, but also through direct effects on oocytes and preimplantation embryos, indicating that the periconceptional period is sensitive to conditions of overnutrition. The periconceptional period includes from folliculogenesis to implantation. Animal model studies suggest that oocytes derived from obese females usually have a small size and mitochondrial abnormalities. These disruptions are probably induced by changes in the components of the ovarian follicular fluid. Experimental evidence also suggests that obesity may affect the microenvironment in oviducts and uterus, resulting in development of preimplantation embryos with reduced cell numbers and up-regulation of proinflammatory genes. However, further research is needed for in-depth characterization of the effects of maternal obesity during the periconceptional period.
Collapse
Affiliation(s)
- Miguel Abraham Velázquez
- Centre for Biological Sciences, University of Southampton, Southampton General Hospital , Southampton, Reino Unido.
| |
Collapse
|
15
|
Nakagami H, Kajihara T, Kamei Y, Ishihara O, Kayano H, Sasaki A, Itakura A. Amniotic components in the uterine vasculature and their role in amniotic fluid embolism. J Obstet Gynaecol Res 2014; 41:870-5. [DOI: 10.1111/jog.12650] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 10/23/2014] [Indexed: 11/29/2022]
Affiliation(s)
- Hiroko Nakagami
- Department of Obstetrics and Gynecology; Saitama Medical University; Saitama Japan
| | - Takeshi Kajihara
- Department of Obstetrics and Gynecology; Saitama Medical University; Saitama Japan
| | - Yoshimasa Kamei
- Department of Obstetrics and Gynecology; Saitama Medical University; Saitama Japan
| | - Osamu Ishihara
- Department of Obstetrics and Gynecology; Saitama Medical University; Saitama Japan
| | - Hidekazu Kayano
- Department of Pathology; Saitama Medical University; Saitama Japan
| | - Atsushi Sasaki
- Department of Pathology; Saitama Medical University; Saitama Japan
| | - Atsuo Itakura
- Department of Obstetrics and Gynecology; Juntendo University; Tokyo Japan
| |
Collapse
|
16
|
Maternal high-fat diet induces follicular atresia but does not affect fertility in adult rabbit offspring. J Dev Orig Health Dis 2014; 5:88-97. [DOI: 10.1017/s2040174414000014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Alterations to the metabolic environment in utero can have an impact on subsequent female reproductive performance. Here, we used a model of rabbits receiving a high-fat diet (H diet; 7.7% fat and 0.2% cholesterol) or a control diet (C diet; 1.8% fat, no cholesterol) from 10 weeks of age up to mating at 27 weeks and throughout gestation and lactation. At weaning at 5 weeks of age, F1 female offspring were placed on either C or H diet, resulting in a total of four groups C/C, C/H, H/C and H/H diet. Female offspring were mated between 18 and 22 weeks of age and euthanized at 28 days of gestation. A few days before mating and/or just before euthanasia, F1 female rabbits were fasted overnight, weighed, and blood sampled for steroids and biochemistry. Organs were weighed at euthanasia and the ovaries were collected. C/H and H/H F1 offspring had higher cholesterol and high-density lipoprotein plasma concentrations, together with a higher fat mass compared with C/C does, reflecting the effect of the postnatal diet; however, no effect of the antenatal diet was observed on most parameters. The number of primordial, primary and secondary follicles were not different between the groups, but a significantly higher number of atretic follicles was observed in the C/H (P<0.001) and in the H/C (P<0.001) compared with control C/C ovaries, demonstrating both an effect of prenatal and postnatal maternal nutrition. These data indicated that both maternal and postnatal high-fat diet may induce follicular apoptosis; however, in this model, the reproduction was not affected.
Collapse
|
17
|
Cordier AG, Léveillé P, Dupont C, Tarrade A, Picone O, Larcher T, Dahirel M, Poumerol E, Mandon-Pepin B, Lévy R, Chavatte-Palmer P. Dietary lipid and cholesterol induce ovarian dysfunction and abnormal LH response to stimulation in rabbits. PLoS One 2013; 8:e63101. [PMID: 23690983 PMCID: PMC3653923 DOI: 10.1371/journal.pone.0063101] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Accepted: 03/28/2013] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND/AIM Excess of fat intake is dramatically increasing in women of childbearing age and results in numerous health complications, including reproductive disorders. Using rabbit does as a biomedical model, the aim of this study was to evaluate onset of puberty, endocrine responses to stimulation and ovarian follicular maturation in females fed a high fat high cholesterol diet (HH diet) from 10 weeks of age (i.e., 2 weeks before normal onset of puberty) or a control diet (C diet). METHODOLOGY/PRINCIPAL FINDINGS Three experiments were performed, each including 8 treated (HH group) and 8 control (C group) does. In experiment 1, the endocrine response to Gonadotropin releasing hormone (GnRH) was evaluated at 13, 18 and 22 weeks of age. In experiment 2, the follicular population was counted in ovaries of adult females (18 weeks of age). In experiment 3, the LH response to mating and steroid profiles throughout gestation were evaluated at 18 weeks of age. Fetal growth was monitored by ultrasound and offspring birth weight was recorded. Data showed a significantly higher Luteinizing hormone (LH) response after induction of ovulation at 13 weeks of age in the HH group. There was no difference at 18 weeks, but at 22 weeks, the LH response to GnRH was significantly reduced in the HH group. The number of atretic follicles was significantly increased and the number of antral follicles significantly reduced in HH does vs. controls. During gestation, the HH diet induced intra-uterine growth retardation (IUGR). CONCLUSION The HH diet administered from before puberty onwards affected onset of puberty, follicular growth, hormonal responses to breeding and GnRH stimulation in relation to age and lead to fetal IUGR.
Collapse
Affiliation(s)
- Anne-Gaël Cordier
- INRA, UMR1198 Biologie du Développement et Reproduction, Jouy-en-Josas, France
- APHP, Hosp Antoine Béclère, Service de Gynécologie-Obstétrique et Médecine de la Reproduction, Clamart, France
| | - Pauline Léveillé
- INRA, UMR1198 Biologie du Développement et Reproduction, Jouy-en-Josas, France
- Université Paris 13, Sorbonne Paris Cité, Unité de Recherche en Epidémiologie Nutritionnelle (UREN), Bobigny, France
- APHP, Hôpital Jean-Verdier, Bondy, France
| | - Charlotte Dupont
- INRA, UMR1198 Biologie du Développement et Reproduction, Jouy-en-Josas, France
- Université Paris 13, Sorbonne Paris Cité, Unité de Recherche en Epidémiologie Nutritionnelle (UREN), Bobigny, France
- APHP, Hôpital Jean-Verdier, Bondy, France
| | - Anne Tarrade
- INRA, UMR1198 Biologie du Développement et Reproduction, Jouy-en-Josas, France
- PremUp foundation, Paris, France
| | - Olivier Picone
- INRA, UMR1198 Biologie du Développement et Reproduction, Jouy-en-Josas, France
| | | | - Michèle Dahirel
- INRA, UMR1198 Biologie du Développement et Reproduction, Jouy-en-Josas, France
- PremUp foundation, Paris, France
| | - Elodie Poumerol
- INRA, UMR1198 Biologie du Développement et Reproduction, Jouy-en-Josas, France
| | | | - Rachel Lévy
- Université Paris 13, Sorbonne Paris Cité, Unité de Recherche en Epidémiologie Nutritionnelle (UREN), Bobigny, France
- APHP, Hôpital Jean-Verdier, Bondy, France
| | - Pascale Chavatte-Palmer
- INRA, UMR1198 Biologie du Développement et Reproduction, Jouy-en-Josas, France
- PremUp foundation, Paris, France
| |
Collapse
|
18
|
Kajihara T, Brosens JJ, Ishihara O. The role of FOXO1 in the decidual transformation of the endometrium and early pregnancy. Med Mol Morphol 2013; 46:61-8. [PMID: 23381604 DOI: 10.1007/s00795-013-0018-z] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Accepted: 07/17/2012] [Indexed: 12/25/2022]
Abstract
Successful pregnancy requires coordination of embryo development, decidualization of endometrium, and placenta formation. Decidualization denotes the transformation of endometrial stromal cells into specialized secretory cells, a process further characterized with influx of specialized immune cells into stroma, predominantly uterine natural killer cells and macrophages, and vascular remodeling. This differentiation process depends on the convergence of the cyclic adenosine monophosphate and progesterone signaling pathways. The decidual process is indispensable for the formation of a functional feto-maternal interface as it controls tissue homeostasis during endovascular trophoblast invasion and bestows tissue resistance to environmental stress signals, including protection against oxidative cell death. FOXO proteins have emerged as key mediators of cell fate because of their ability to regulate either pro-apoptotic genes or genes involved in differentiation, cell cycle arrest, oxidative defenses, and DNA repair. In the endometrium, FOXO1 is of particular importance as a critical regulator of progesterone-dependent differentiation, menstrual shedding, and protection of the feto-maternal against oxidative damage during pregnancy.
Collapse
Affiliation(s)
- Takeshi Kajihara
- Department of Obstetrics and Gynecology, Saitama Medical University, 38 Morohongo, Moroyama, Iruma-gun, Saitama, Japan.
| | | | | |
Collapse
|
19
|
Elucidation of regulatory mechanisms revealed by human promoter sequence analysis of genes co-expressed in forskolin-treated theca cells in PCOS. Arch Gynecol Obstet 2012; 287:477-85. [DOI: 10.1007/s00404-012-2580-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Accepted: 09/20/2012] [Indexed: 10/27/2022]
|
20
|
Shen M, Lin F, Zhang J, Tang Y, Chen WK, Liu H. Involvement of the up-regulated FoxO1 expression in follicular granulosa cell apoptosis induced by oxidative stress. J Biol Chem 2012; 287:25727-40. [PMID: 22669940 DOI: 10.1074/jbc.m112.349902] [Citation(s) in RCA: 151] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Follicular atresia is common in female mammalian ovaries, where most follicles undergo degeneration at any stage of growth and development. Oxidative stress gives rise to triggering granulosa cell apoptosis, which has been suggested as a major cause of follicular atresia. However, the underlying mechanism by which the oxidative stress induces follicular atresia remains unclear. FoxO transcription factors are known as critical mediators in the regulation of oxidative stress and apoptosis. In this study, the involvement of FoxO1 in oxidative stress-induced apoptosis of mouse follicular granulosa cells (MGCs) was investigated in vivo and in vitro. It was observed that increased apoptotic signals correlated with elevated expression of FoxO1 in MGCs when mice were treated with the oxidant. Correspondingly, the expressions of FoxO1 target genes, such as proapoptotic genes and antioxidative genes, were also up-regulated. In primary cultured MGCs, treatment with H(2)O(2) led to FoxO1 nuclear translocation. Further studies with overexpression and knockdown of FoxO1 demonstrated the critical role of FoxO1 in the induction of MGC apoptosis by oxidative stress. Finally, inactivation of FoxO1 by insulin treatment confirmed that FoxO1 induced by oxidative stress played a pivotal role in up-regulating the expression of downstream apoptosis-related genes in MGCs. Our results suggest that up-regulation of FoxO1 by oxidative stress leads to apoptosis of granulosa cells, which eventually results in follicular atresia in mice.
Collapse
Affiliation(s)
- Ming Shen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | | | | | | | | | | |
Collapse
|
21
|
Israel DD, Sheffer-Babila S, de Luca C, Jo YH, Liu SM, Xia Q, Spergel DJ, Dun SL, Dun NJ, Chua SC. Effects of leptin and melanocortin signaling interactions on pubertal development and reproduction. Endocrinology 2012; 153:2408-19. [PMID: 22408174 PMCID: PMC3381095 DOI: 10.1210/en.2011-1822] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Leptin and melanocortin signaling control ingestive behavior, energy balance, and substrate utilization, but only leptin signaling defects cause hypothalamic hypogonadism and infertility. Although GnRH neurons do not express leptin receptors, leptin influences GnRH neuron activity via regulation of immediate downstream mediators including the neuropeptides neuropeptide Y and the melanocortin agonist and antagonist, α-MSH, agouti-related peptide, respectively. Here we show that modulation of melanocortin signaling in female db/db mice through ablation of agouti-related peptide, or heterozygosity of melanocortin 4 receptor, restores the timing of pubertal onset, fertility, and lactation. Additionally, melanocortin 4 receptor activation increases action potential firing and induces c-Fos expression in GnRH neurons, providing further evidence that melanocortin signaling influences GnRH neuron activity. These studies thus establish melanocortin signaling as an important component in the leptin-mediated regulation of GnRH neuron activity, initiation of puberty and fertility.
Collapse
Affiliation(s)
- Davelene D Israel
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|