1
|
Zhao T, He M, Zhu Z, Zhang T, Zheng W, Qin S, Gao M, Wang W, Chen Z, Han J, Liu L, Zhou B, Wang H, Zhang H, Xia G, Wang J, Wang F, Wang C. P62 promotes FSH-induced antral follicle formation by directing degradation of ubiquitinated WT1. Cell Mol Life Sci 2024; 81:221. [PMID: 38763964 PMCID: PMC11102895 DOI: 10.1007/s00018-024-05251-x] [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: 12/23/2023] [Revised: 04/15/2024] [Accepted: 04/22/2024] [Indexed: 05/21/2024]
Abstract
In females, the pathophysiological mechanism of poor ovarian response (POR) is not fully understood. Considering the expression level of p62 was significantly reduced in the granulosa cells (GCs) of POR patients, this study focused on identifying the role of the selective autophagy receptor p62 in conducting the effect of follicle-stimulating hormone (FSH) on antral follicles (AFs) formation in female mice. The results showed that p62 in GCs was FSH responsive and that its level increased to a peak and then decreased time-dependently either in ovaries or in GCs after gonadotropin induction in vivo. GC-specific deletion of p62 resulted in subfertility, a significantly reduced number of AFs and irregular estrous cycles, which were same as pathophysiological symptom of POR. By conducting mass spectrum analysis, we found the ubiquitination of proteins was decreased, and autophagic flux was blocked in GCs. Specifically, the level of nonubiquitinated Wilms tumor 1 homolog (WT1), a transcription factor and negative controller of GC differentiation, increased steadily. Co-IP results showed that p62 deletion increased the level of ubiquitin-specific peptidase 5 (USP5), which blocked the ubiquitination of WT1. Furthermore, a joint analysis of RNA-seq and the spatial transcriptome sequencing data showed the expression of steroid metabolic genes and FSH receptors pivotal for GCs differentiation decreased unanimously. Accordingly, the accumulation of WT1 in GCs deficient of p62 decreased steroid hormone levels and reduced FSH responsiveness, while the availability of p62 in GCs simultaneously ensured the degradation of WT1 through the ubiquitin‒proteasome system and autophagolysosomal system. Therefore, p62 in GCs participates in GC differentiation and AF formation in FSH induction by dynamically controlling the degradation of WT1. The findings of the study contributes to further study the pathology of POR.
Collapse
Affiliation(s)
- Ting Zhao
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Meina He
- College of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou Province, 550025, China
| | - Zijian Zhu
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Tuo Zhang
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Department of Physiology, College of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou Province, 550025, China
| | - Wenying Zheng
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Shaogang Qin
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Meng Gao
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Wenji Wang
- School of Life Science, Taizhou University, Taizhou, 318000, China
| | - Ziqi Chen
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Jun Han
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Longping Liu
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Bo Zhou
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Haibin Wang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, School of Medicine, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, Fujian Province, 361005, China
| | - Hua Zhang
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Guoliang Xia
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, College of Life Science, Ningxia University, Yinchuan, 750021, China
| | - Jianbin Wang
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Fengchao Wang
- Transgenic Animal Center, National Institute of Biological Sciences, Beijing, 102206, China
| | - Chao Wang
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, 100193, China.
- China Agricultural University, No.2 Yuan Ming Yuan West Road, Haidian District, Beijing, 100193, China.
| |
Collapse
|
2
|
Pandey AN, Yadav PK, Premkumar KV, Tiwari M, Pandey AK, Chaube SK. Reactive oxygen species signalling in the deterioration of quality of mammalian oocytes cultured in vitro: Protective effect of antioxidants. Cell Signal 2024; 117:111103. [PMID: 38367792 DOI: 10.1016/j.cellsig.2024.111103] [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: 11/12/2023] [Revised: 02/13/2024] [Accepted: 02/14/2024] [Indexed: 02/19/2024]
Abstract
The in vitro fertilization (IVF) is the first choice of infertile couples worldwide to plan for conception. Besides having a significant advancement in IVF procedure, the success rate is still poor. Although several approaches have been tested to improve IVF protocol, minor changes in culture conditions, physical factors and/or drug treatment generate reactive oxygen species (ROS) in oocytes. Due to large size and huge number of mitochondria, oocyte is more susceptible towards ROS-mediated signalling under in vitro culture conditions. Elevation of ROS levels destabilize maturation promoting factor (MPF) that results in meiotic exit from diplotene as well as metaphase-II (M-II) arrest in vitro. Once meiotic exit occurs, these oocytes get further arrested at metaphase-I (M-I) stage or metaphase-III (M-III)-like stage under in vitro culture conditions. The M-I as well as M-III arrested oocytes are not fit for fertilization and limits IVF outcome. Further, the generation of excess levels of ROS cause oxidative stress (OS) that initiate downstream signalling to initiate various death pathways such as apoptosis, autophagy, necroptosis and deteriorates oocyte quality under in vitro culture conditions. The increase of cellular enzymatic antioxidants and/or supplementation of exogenous antioxidants in culture medium protect ROS-induced deterioration of oocyte quality in vitro. Although a growing body of evidence suggests the ROS and OS-mediated deterioration of oocyte quality in vitro, their downstream signalling and related mechanisms remain poorly understood. Hence, this review article summarizes the existing evidences concerning ROS and OS-mediated downstream signalling during deterioration of oocyte quality in vitro. The use of various antioxidants against ROS and OS-mediated impairment of oocyte quality in vitro has also been explored in order to increase the success rate of IVF during assisted reproductive health management.
Collapse
Affiliation(s)
- Ashutosh N Pandey
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Pramod K Yadav
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Karuppanan V Premkumar
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Meenakshi Tiwari
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Ajai K Pandey
- Department of Kayachikitsa, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Shail K Chaube
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India.
| |
Collapse
|
3
|
Chesnokov MS, Mamedova AR, Zhivotovsky B, Kopeina GS. A matter of new life and cell death: programmed cell death in the mammalian ovary. J Biomed Sci 2024; 31:31. [PMID: 38509545 PMCID: PMC10956231 DOI: 10.1186/s12929-024-01017-6] [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: 09/29/2023] [Accepted: 02/27/2024] [Indexed: 03/22/2024] Open
Abstract
BACKGROUND The mammalian ovary is a unique organ that displays a distinctive feature of cyclic changes throughout the entire reproductive period. The estrous/menstrual cycles are associated with drastic functional and morphological rearrangements of ovarian tissue, including follicular development and degeneration, and the formation and subsequent atrophy of the corpus luteum. The flawless execution of these reiterative processes is impossible without the involvement of programmed cell death (PCD). MAIN TEXT PCD is crucial for efficient and careful clearance of excessive, depleted, or obsolete ovarian structures for ovarian cycling. Moreover, PCD facilitates selection of high-quality oocytes and formation of the ovarian reserve during embryonic and juvenile development. Disruption of PCD regulation can heavily impact the ovarian functions and is associated with various pathologies, from a moderate decrease in fertility to severe hormonal disturbance, complete loss of reproductive function, and tumorigenesis. This comprehensive review aims to provide updated information on the role of PCD in various processes occurring in normal and pathologic ovaries. Three major events of PCD in the ovary-progenitor germ cell depletion, follicular atresia, and corpus luteum degradation-are described, alongside the detailed information on molecular regulation of these processes, highlighting the contribution of apoptosis, autophagy, necroptosis, and ferroptosis. Ultimately, the current knowledge of PCD aberrations associated with pathologies, such as polycystic ovarian syndrome, premature ovarian insufficiency, and tumors of ovarian origin, is outlined. CONCLUSION PCD is an essential element in ovarian development, functions and pathologies. A thorough understanding of molecular mechanisms regulating PCD events is required for future advances in the diagnosis and management of various disorders of the ovary and the female reproductive system in general.
Collapse
Affiliation(s)
- Mikhail S Chesnokov
- Faculty of Medicine, MV Lomonosov Moscow State University, Moscow, Russia
- Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Aygun R Mamedova
- Faculty of Medicine, MV Lomonosov Moscow State University, Moscow, Russia
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Boris Zhivotovsky
- Faculty of Medicine, MV Lomonosov Moscow State University, Moscow, Russia.
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.
- Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden.
| | - Gelina S Kopeina
- Faculty of Medicine, MV Lomonosov Moscow State University, Moscow, Russia.
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.
| |
Collapse
|
4
|
Zhu JR, Zhu WJ, Li T, Ou JP. Autophagy activity is increased in the cumulus cells of women with poor ovarian response. Taiwan J Obstet Gynecol 2024; 63:205-213. [PMID: 38485316 DOI: 10.1016/j.tjog.2024.01.016] [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] [Accepted: 09/18/2023] [Indexed: 03/19/2024] Open
Abstract
OBJECTIVE To evaluate the autophagy status of cumulus cells (CCs) in women with poor ovarian response (POR). MATERIALS AND METHODS CCs were divided into normal ovarian response (NOR) group and POR group. The ultrastructure of autophagy was analyzed by transmission electron microscopy (NOR: n = 18, POR: n = 26). The mRNA and protein of autophagy markers were detected by Quantitative real-time polymerase chain reaction (NOR: n = 15, POR: n = 19) and Western blotting (NOR: n = 41, POR: n = 38), respectively. RESULTS Transmission electron microscopy demonstrated abundant autophagosomes and even autophagic death in the POR group. There were no differences in LC3 and P62 mRNA expression between the two groups (p > 0.05). The BCL2 mRNA expression was lower in the POR group (p < 0.05). Moreover, the LC3 II/I ratio and the P62 protein expression were significantly higher in the POR group (p < 0.05). CONCLUSIONS Autophagy in CCs of POR women is activated and the autophagic flux is blocked. The up-regulation of autophagy in CCs may be related to the pathogenesis of POR.
Collapse
Affiliation(s)
- Jie-Ru Zhu
- Center for Reproductive Medicine, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Wei-Jie Zhu
- College of Life Science and Technology, Jinan University, Guangzhou, People's Republic of China
| | - Tao Li
- Center for Reproductive Medicine, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jian-Ping Ou
- Center for Reproductive Medicine, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China.
| |
Collapse
|
5
|
Kandouz M. Cell Death, by Any Other Name…. Cells 2024; 13:325. [PMID: 38391938 PMCID: PMC10886887 DOI: 10.3390/cells13040325] [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: 12/31/2023] [Revised: 02/04/2024] [Accepted: 02/06/2024] [Indexed: 02/24/2024] Open
Abstract
Studies trying to understand cell death, this ultimate biological process, can be traced back to a century ago. Yet, unlike many other fashionable research interests, research on cell death is more alive than ever. New modes of cell death are discovered in specific contexts, as are new molecular pathways. But what is "cell death", really? This question has not found a definitive answer yet. Nevertheless, part of the answer is irreversibility, whereby cells can no longer recover from stress or injury. Here, we identify the most distinctive features of different modes of cell death, focusing on the executive final stages. In addition to the final stages, these modes can differ in their triggering stimulus, thus referring to the initial stages. Within this framework, we use a few illustrative examples to examine how intercellular communication factors in the demise of cells. First, we discuss the interplay between cell-cell communication and cell death during a few steps in the early development of multicellular organisms. Next, we will discuss this interplay in a fully developed and functional tissue, the gut, which is among the most rapidly renewing tissues in the body and, therefore, makes extensive use of cell death. Furthermore, we will discuss how the balance between cell death and communication is modified during a pathological condition, i.e., colon tumorigenesis, and how it could shed light on resistance to cancer therapy. Finally, we briefly review data on the role of cell-cell communication modes in the propagation of cell death signals and how this has been considered as a potential therapeutic approach. Far from vainly trying to provide a comprehensive review, we launch an invitation to ponder over the significance of cell death diversity and how it provides multiple opportunities for the contribution of various modes of intercellular communication.
Collapse
Affiliation(s)
- Mustapha Kandouz
- Department of Pathology, School of Medicine, Wayne State University, 540 East Canfield Avenue, Detroit, MI 48201, USA;
- Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA
| |
Collapse
|
6
|
Telfer EE, Grosbois J, Odey YL, Rosario R, Anderson RA. Making a good egg: human oocyte health, aging, and in vitro development. Physiol Rev 2023; 103:2623-2677. [PMID: 37171807 PMCID: PMC10625843 DOI: 10.1152/physrev.00032.2022] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 05/03/2023] [Accepted: 05/06/2023] [Indexed: 05/13/2023] Open
Abstract
Mammalian eggs (oocytes) are formed during fetal life and establish associations with somatic cells to form primordial follicles that create a store of germ cells (the primordial pool). The size of this pool is influenced by key events during the formation of germ cells and by factors that influence the subsequent activation of follicle growth. These regulatory pathways must ensure that the reserve of oocytes within primordial follicles in humans lasts for up to 50 years, yet only approximately 0.1% will ever be ovulated with the rest undergoing degeneration. This review outlines the mechanisms and regulatory pathways that govern the processes of oocyte and follicle formation and later growth, within the ovarian stroma, through to ovulation with particular reference to human oocytes/follicles. In addition, the effects of aging on female reproductive capacity through changes in oocyte number and quality are emphasized, with both the cellular mechanisms and clinical implications discussed. Finally, the details of current developments in culture systems that support all stages of follicle growth to generate mature oocytes in vitro and emerging prospects for making new oocytes from stem cells are outlined.
Collapse
Affiliation(s)
- Evelyn E Telfer
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Johanne Grosbois
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Yvonne L Odey
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Roseanne Rosario
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Richard A Anderson
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
7
|
Stringer JM, Alesi LR, Winship AL, Hutt KJ. Beyond apoptosis: evidence of other regulated cell death pathways in the ovary throughout development and life. Hum Reprod Update 2023; 29:434-456. [PMID: 36857094 PMCID: PMC10320496 DOI: 10.1093/humupd/dmad005] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 12/06/2022] [Indexed: 03/02/2023] Open
Abstract
BACKGROUND Regulated cell death is a fundamental component of numerous physiological processes; spanning from organogenesis in utero, to normal cell turnover during adulthood, as well as the elimination of infected or damaged cells throughout life. Quality control through regulation of cell death pathways is particularly important in the germline, which is responsible for the generation of offspring. Women are born with their entire supply of germ cells, housed in functional units known as follicles. Follicles contain an oocyte, as well as specialized somatic granulosa cells essential for oocyte survival. Follicle loss-via regulated cell death-occurs throughout follicle development and life, and can be accelerated following exposure to various environmental and lifestyle factors. It is thought that the elimination of damaged follicles is necessary to ensure that only the best quality oocytes are available for reproduction. OBJECTIVE AND RATIONALE Understanding the precise factors involved in triggering and executing follicle death is crucial to uncovering how follicle endowment is initially determined, as well as how follicle number is maintained throughout puberty, reproductive life, and ovarian ageing in women. Apoptosis is established as essential for ovarian homeostasis at all stages of development and life. However, involvement of other cell death pathways in the ovary is less established. This review aims to summarize the most recent literature on cell death regulators in the ovary, with a particular focus on non-apoptotic pathways and their functions throughout the discrete stages of ovarian development and reproductive life. SEARCH METHODS Comprehensive literature searches were carried out using PubMed and Google Scholar for human, animal, and cellular studies published until August 2022 using the following search terms: oogenesis, follicle formation, follicle atresia, oocyte loss, oocyte apoptosis, regulated cell death in the ovary, non-apoptotic cell death in the ovary, premature ovarian insufficiency, primordial follicles, oocyte quality control, granulosa cell death, autophagy in the ovary, autophagy in oocytes, necroptosis in the ovary, necroptosis in oocytes, pyroptosis in the ovary, pyroptosis in oocytes, parthanatos in the ovary, and parthanatos in oocytes. OUTCOMES Numerous regulated cell death pathways operate in mammalian cells, including apoptosis, autophagic cell death, necroptosis, and pyroptosis. However, our understanding of the distinct cell death mediators in each ovarian cell type and follicle class across the different stages of life remains the source of ongoing investigation. Here, we highlight recent evidence for the contribution of non-apoptotic pathways to ovarian development and function. In particular, we discuss the involvement of autophagy during follicle formation and the role of autophagic cell death, necroptosis, pyroptosis, and parthanatos during follicle atresia, particularly in response to physiological stressors (e.g. oxidative stress). WIDER IMPLICATIONS Improved knowledge of the roles of each regulated cell death pathway in the ovary is vital for understanding ovarian development, as well as maintenance of ovarian function throughout the lifespan. This information is pertinent not only to our understanding of endocrine health, reproductive health, and fertility in women but also to enable identification of novel fertility preservation targets.
Collapse
Affiliation(s)
- Jessica M Stringer
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Lauren R Alesi
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Amy L Winship
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Karla J Hutt
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| |
Collapse
|
8
|
Li Z, Ruan Z, Feng Y, Wang Y, Zhang J, Lu C, Shi D, Lu F. METTL3-mediated m6A methylation regulates granulosa cells autophagy during follicular atresia in pig ovaries. Theriogenology 2023; 201:83-94. [PMID: 36857977 DOI: 10.1016/j.theriogenology.2023.02.021] [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: 01/06/2023] [Revised: 02/16/2023] [Accepted: 02/18/2023] [Indexed: 02/24/2023]
Abstract
Follicular atresia is a normal physiological event in mammals, yet its mechanism remains to be studied. Granulosa cell (GC) autophagy is closely associated with follicular atresia. The N6-methyladenosine (m6A) modification is the most common post-transcriptional modification in eukaryotes, but its role in follicular atresia is still unknown. In this study, the possible relationship amongst follicular atresia, GC autophagy and m6A modification was studied. Our results showed that the level of autophagy in GCs increased with the degree of follicle atresia, whereas the overall m6A level decreased. Rapamycin treatment induced atresia in vitro cultured follicles, whereas 3-Methyladenine inhibited follicular atresia. Progressed atretic follicle (PAF) GCs had significantly lower METTL3 levels and significantly higher FTO levels than healthy follicle (HF) GCs. Differential gene expression analysis of GCs in PAF and HF by RNA sequencing was showed that the autophagy-related genes ULK1, ULK2, ATG2A, and ATG2B were significantly elevated in the PAF. In cultured GCs, overexpression of METTL3 significantly decreased the mRNA level of ULK1, as well as the autophagy level, whereas knockdown of METTL3 by RNAi significantly increased the mRNA level of ULK1, as well as the autophagy level. Our results indicate that m6A modification can regulate autophagy in GCs and play a role in the process of porcine follicular atresia.
Collapse
Affiliation(s)
- Zhengda Li
- Animal Reproduction Institute, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China; Reproductive Medical and Genetic Center, The People's Hospital of Guangxi Zhuang Autonoumous Region, Nanning, Guangxi, 530021, China
| | - Ziyun Ruan
- Animal Reproduction Institute, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China; School of Basic Medicine, Guangxi University of Traditional Chinese Medicine, Nanning, Guangxi, 530001, China
| | - Yun Feng
- Animal Reproduction Institute, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
| | - Yanxin Wang
- Animal Reproduction Institute, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
| | - Jun Zhang
- Animal Reproduction Institute, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
| | - Canqiang Lu
- Animal Reproduction Institute, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
| | - Deshun Shi
- Animal Reproduction Institute, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China.
| | - Fenghua Lu
- Animal Reproduction Institute, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China.
| |
Collapse
|
9
|
Castro-Cruz A, Echeverría OM, Sánchez-Sánchez L, Muñoz-Velasco I, Juárez-Chavero S, Torres-Ramírez N, Vázquez-Nin GH, Escobar ML. Dissection of the autophagic route in oocytes from atretic follicles. Biol Cell 2023; 115:e2200046. [PMID: 36571578 DOI: 10.1111/boc.202200046] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 12/19/2022] [Accepted: 12/21/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND INFORMATION Autophagy is a conserved process that functions as a cytoprotective mechanism; it may function as a cell death process called programmed cell death type II. There is considerable evidence for the presence of autophagic cell death during oocyte elimination in prepubertal rats. However, the mechanisms involved in this process have not been deciphered. RESULTS Our observations revealed autophagic cell death in oocytes with increased labeling of the autophagic proteins Beclin 1, light chain 3 A (LC3 A), and lysosomal-associated membrane protein 1 (Lamp1). Furthermore, mTOR and phosphorylated (p)-mTOR (S2448) proteins were significantly decreased in oocytes with increased levels of autophagic proteins, indicating autophagic activation. Moreover, phosphorylated protein kinase B (p-AKT) was not expressed by oocytes, but mitogen-activated protein kinase/extracellular signalregulated kinase (MAPK/ERK) signaling was observed. Additionally, selective and elevated mitochondrial degradation was identified in altered oocytes. CONCLUSIONS All these results suggest that mTOR downregulation, which promotes autophagy, could be mediated by low energy levels and sustained starvation involving the phosphoinositide 3-kinase (PI3K)/AKT/mTOR and MAPK/ERK pathways. SIGNIFICANCE In this work, we analyzed the manner in which autophagy is carried out in oocytes undergoing autophagic cell death by studying the behavior of proteins involved in different steps of the autophagic pathway.
Collapse
Affiliation(s)
- Abraham Castro-Cruz
- Laboratorio de Microscopía Electrónica, Departamento de Biología Celular, Facultad de Ciencias, Universidad Nacional Autónoma de México,Ciudad Universitaria, Col. Universidad Nacional Autónoma de México, Coyoacán, Ciudad de México, México
| | - Olga M Echeverría
- Laboratorio de Microscopía Electrónica, Departamento de Biología Celular, Facultad de Ciencias, Universidad Nacional Autónoma de México,Ciudad Universitaria, Col. Universidad Nacional Autónoma de México, Coyoacán, Ciudad de México, México
| | - Luis Sánchez-Sánchez
- Laboratorio de Biología Molecular del Cáncer, Lab. 6, 2do piso, UMIEZ, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, México, Ciudad de México, Iztapalapa, México
| | - Israel Muñoz-Velasco
- Laboratorio de Microscopía Electrónica, Departamento de Biología Celular, Facultad de Ciencias, Universidad Nacional Autónoma de México,Ciudad Universitaria, Col. Universidad Nacional Autónoma de México, Coyoacán, Ciudad de México, México
| | - Silvia Juárez-Chavero
- Laboratorio de Microscopía Electrónica, Departamento de Biología Celular, Facultad de Ciencias, Universidad Nacional Autónoma de México,Ciudad Universitaria, Col. Universidad Nacional Autónoma de México, Coyoacán, Ciudad de México, México
| | - Nayeli Torres-Ramírez
- Laboratorio de Microscopía Electrónica, Departamento de Biología Celular, Facultad de Ciencias, Universidad Nacional Autónoma de México,Ciudad Universitaria, Col. Universidad Nacional Autónoma de México, Coyoacán, Ciudad de México, México
| | - Gerardo H Vázquez-Nin
- Laboratorio de Microscopía Electrónica, Departamento de Biología Celular, Facultad de Ciencias, Universidad Nacional Autónoma de México,Ciudad Universitaria, Col. Universidad Nacional Autónoma de México, Coyoacán, Ciudad de México, México
| | - María Luisa Escobar
- Laboratorio de Microscopía Electrónica, Departamento de Biología Celular, Facultad de Ciencias, Universidad Nacional Autónoma de México,Ciudad Universitaria, Col. Universidad Nacional Autónoma de México, Coyoacán, Ciudad de México, México
| |
Collapse
|
10
|
Cacciottola L, Camboni A, Cernogoraz A, Donnez J, Dolmans MM. Role of apoptosis and autophagy in ovarian follicle pool decline in children and women diagnosed with benign or malignant extra-ovarian conditions. Hum Reprod 2023; 38:75-88. [PMID: 36346333 DOI: 10.1093/humrep/deac237] [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: 03/23/2022] [Revised: 09/26/2022] [Indexed: 11/09/2022] Open
Abstract
STUDY QUESTION Which biological mechanisms are responsible for physiological ovarian reserve decline owing to aging, or pathological follicle depletion triggered by inflammation or a pro-oxidant environment throughout a woman's lifetime? SUMMARY ANSWER Ovarian follicle pool size is modulated by both apoptosis and autophagy, the first responsible for its physiological decline over time and increasing in the event of prior chemotherapy in children, and the latter playing a major role in physiological ovarian follicle pool diminution before puberty. WHAT IS KNOWN ALREADY Among the different pathways of controlled cell death, apoptosis and autophagy are implicated in follicle loss. Apoptosis participates in eliminating damaged follicles, such as those impaired by chemotherapy (CHT), but its involvement in physiological age-related follicle decline is less well understood. Autophagy has proved crucial in follicle quiescence maintenance in murine models, but its contribution to human follicle pool modulation is still unclear. STUDY DESIGN, SIZE, DURATION This retrospective study included 84 patients with benign or malignant extra-ovarian conditions aged between 1 and 35 years, with ovarian tissue stored for histological analyses at the time of cryopreservation (between 2012 and 2021) at a tertiary care center. PARTICIPANTS/MATERIALS, SETTING, METHODS Ovarian fragments were used for the following analyses: hematoxylin and eosin staining for follicle count and classification; cleaved caspase-3 immunostaining to identify follicle apoptosis; and microtubule-associated proteins 1A/1B light chain 3B immunolabeling to detect follicle autophagy. Transmission electron microscopy was also carried out to investigate ultrastructural features of oocytes and granulosa cells. All analyses stratified patients by age, menarchal status (premenarchal = 32; postmenarchal = 52), potentially gonadotoxic CHT before cryopreservation (n = 14), presence of endometriosis and use of hormonal treatment. MAIN RESULTS AND THE ROLE OF CHANCE Premenarchal patients had a larger follicle pool in terms of total follicle density [mean, range 4979.98 (342.2-21789) versus 918.8 (26.18-3983), P < 0.001], but higher rates of morphologically abnormal [8.52 (0-25.37)% versus 3.54 (0-17.5)%, P < 0.001] and atretic [15.8 (0‒31.85)% versus 10.6 (0-33.33)%, P < 0.01] follicles than postmenarchal subjects. Apoptosis rates did not change with increasing age [27.94 (0-93.2)% in prepubertal subjects and 29.5 (0-100)% in postpubertal subjects], but autophagic follicles were around 10 times more common in premenarchal than postmenarchal subjects [10.21 (0-62.3)% versus 1.34 (0-25)%, P < 0.001], playing a crucial role in age-related follicle decline and elimination of 'abnormal' follicles, that are rarely seen after menarche. The impact of diagnosis and previous CHT varied according to age. In premenarchal patients with previous CHT, significantly more apoptotic [40.22 (0-100)% versus 26.79 (0-87)%, P < 0.05] and fewer abnormal [3.84 (0-10-76)% versus 9.83 (0-25.37)%, P < 0.01] follicles were detected than in subjects with no CHT prior to ovarian tissue cryopreservation, suggesting a direct effect on follicle elimination, especially of those with abnormalities. In postmenarchal subjects with previous CHT, quiescent follicle rates were lower than in patients with no CHT before tissue freezing [71.57 (0-100)% versus 85.89 (50-100)%, P < 0.05], suggesting accelerated follicle activation and growth. Moreover, increased autophagic activity was observed in the event of a cancer diagnosis compared to benign conditions after puberty [26.27 (0-100)% versus 9.48 (0-29.41)%, respectively, P < 0.05]. LIMITATIONS, REASONS FOR CAUTION The impact of specific CHT protocols could not be investigated since the group of patients with previous CHT was highly heterogeneous. WIDER IMPLICATIONS OF THE FINDINGS This study yields a deeper understanding of regulation of the follicle pool decline, showing for the first time that both apoptosis and autophagy pathways are involved in physiological follicle depletion, the latter being crucial before puberty. Moreover, our data showed a different response to non-physiological damage according to age, with higher apoptosis rates only in premenarchal subjects with previous CHT, confirming that this pathway is activated by drugs known to induce DNA damage in oocytes, such as alkylating agents, but not by cancer itself. STUDY FUNDING/COMPETING INTEREST(S) This study was supported by grants from the Fonds National de la Recherche Scientifique de Belgique (F.R.S.-FNRS/FRIA FC29657 awarded to L.C., CDR J.0063.20 and grant 5/4/150/5 awarded to M.M.D.), grants from the Fondation contre le Cancer (grant 2018-042 awarded to A.Ca.), the Fondazione Comunitaria del Varesotto and Provincia di Varese ('Amalia Griffini' Fellowship in Gynecology and Obstetrics awarded to A.Ce.), Fonds Spéciaux de Recherche, Fondation St Luc and donations from the Ferrero family. The authors have no competing interests to declare. TRIAL REGISTRAION NUMBER N/A.
Collapse
Affiliation(s)
- L Cacciottola
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - A Camboni
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium.,Department of Anatomopathology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - A Cernogoraz
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium.,Department of Gynecology and Obstetrics, F. Del Ponte Hospital, University of Insubria, Varese, Italy
| | - J Donnez
- Society for Research into Infertility, Brussels, Belgium.,Professor EM, Université Catholique de Louvain, Brussels, Belgium
| | - M M Dolmans
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium.,Department of Gynecology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
11
|
Clark ZL, Ruebel ML, Schall PZ, Karl KR, Ireland JJ, Latham KE. Follicular Hyperstimulation Dysgenesis: New Explanation for Adverse Effects of Excessive FSH in Ovarian Stimulation. Endocrinology 2022; 163:bqac100. [PMID: 35833461 PMCID: PMC9342683 DOI: 10.1210/endocr/bqac100] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Indexed: 11/19/2022]
Abstract
High follicle-stimulating hormone (FSH) doses during ovarian stimulation protocols for assisted reproductive technologies (ART) are detrimental to ovulatory follicle function and oocyte quality. However, the mechanisms are unclear. In a small ovarian reserve heifer model, excessive FSH doses lead to phenotypic heterogeneity of ovulatory size follicles, with most follicles displaying signs of premature luteinization and a range in severity of abnormalities. By performing whole transcriptome analyses of granulosa cells, cumulus cells, and oocytes from individual follicles of animals given standard or excessive FSH doses, we identified progressive changes in the transcriptomes of the 3 cell types, with increasing severity of follicular abnormality with the excessive doses. The granulosa and cumulus cells each diverged progressively from their normal phenotypes and became highly similar to each other in the more severely affected follicles. Pathway analysis indicates a possible dysregulation of the final stages of folliculogenesis, with processes characteristic of ovulation and luteinization occurring concurrently rather than sequentially in the most severely affected follicles. These changes were associated with disruptions in key pathways in granulosa and cumulus cells, which may account for previously reported reduced estradiol production, enhanced progesterone and oxytocin production and diminished ovulation rates. Predicted deficiencies in oocyte survival, stress response, and fertilization suggest likely reductions in oocyte health, which could further compromise oocyte quality and ART outcomes.
Collapse
Affiliation(s)
- Zaramasina L Clark
- Reproductive and Developmental Sciences Program and the Department of Animal Science, Michigan State University, East Lansing, MI 48824, USA
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
| | - Meghan L Ruebel
- Reproductive and Developmental Sciences Program and the Department of Animal Science, Michigan State University, East Lansing, MI 48824, USA
- USDA-ARS Arkansas Children’s Nutrition Center 15 Children’s Way Little Rock, AR 72202, USA
| | - Peter Z Schall
- Reproductive and Developmental Sciences Program and the Department of Animal Science, Michigan State University, East Lansing, MI 48824, USA
- University of Michigan Medical School, Department of Human Genetics, Ann Arbor, Michigan, USA
| | - Kaitlin R Karl
- Reproductive and Developmental Sciences Program and the Department of Animal Science, Michigan State University, East Lansing, MI 48824, USA
| | - James J Ireland
- Reproductive and Developmental Sciences Program and the Department of Animal Science, Michigan State University, East Lansing, MI 48824, USA
| | - Keith E Latham
- Reproductive and Developmental Sciences Program and the Department of Animal Science, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
12
|
Feng YQ, Wang JJ, Li MH, Tian Y, Zhao AH, Li L, De Felici M, Shen W. Impaired primordial follicle assembly in offspring ovaries from zearalenone-exposed mothers involves reduced mitochondrial activity and altered epigenetics in oocytes. Cell Mol Life Sci 2022; 79:258. [PMID: 35469021 PMCID: PMC11071983 DOI: 10.1007/s00018-022-04288-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/31/2022] [Accepted: 04/04/2022] [Indexed: 01/18/2023]
Abstract
Previous works have shown that zearalenone (ZEA), as an estrogenic pollutant, has adverse effects on mammalian folliculogenesis. In the present study, we found that prolonged exposure of female mice to ZEA around the end of pregnancy caused severe impairment of primordial follicle formation in the ovaries of newborn mice and altered the expression of many genes in oocytes as revealed by single-cell RNA sequencing (scRNA-seq). These changes were associated with morphological and molecular alterations of mitochondria, increased autophagic markers in oocytes, and epigenetic changes in the ovaries of newborn mice from ZEA-exposed mothers. The latter increased expression of HDAC2 deacetylases was leading to decreased levels of H3K9ac and H4K12ac. Most of these modifications were relieved when the expression of Hdac2 in newborn ovaries was reduced by RNA interference during in vitro culture in the presence of ZEA. Such changes were also alleviated in offspring ovaries from mothers treated with both ZEA and the coenzyme Q10 (CoQ10), which is known to be able to restore mitochondrial activities. We concluded that impaired mitochondrial activities in oocytes caused by ZEA are at the origin of metabolic alterations that modify the expression of genes controlling autophagy and primordial follicle assembly through changes in epigenetic histones.
Collapse
Affiliation(s)
- Yan-Qin Feng
- College of Life Sciences, Key Laboratory of Animal Reproduction and Biotechnology in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, China
| | - Jun-Jie Wang
- College of Life Sciences, Key Laboratory of Animal Reproduction and Biotechnology in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, China
| | - Ming-Hao Li
- College of Life Sciences, Key Laboratory of Animal Reproduction and Biotechnology in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, China
| | - Yu Tian
- College of Life Sciences, Key Laboratory of Animal Reproduction and Biotechnology in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, China
| | - Ai-Hong Zhao
- Qingdao Academy of Agricultural Sciences, Qingdao, 266100, China
| | - Lan Li
- College of Life Sciences, Key Laboratory of Animal Reproduction and Biotechnology in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, China
| | - Massimo De Felici
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Wei Shen
- College of Life Sciences, Key Laboratory of Animal Reproduction and Biotechnology in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, China.
| |
Collapse
|
13
|
Przygrodzka E, Monaco CF, Plewes MR, Li G, Wood JR, Cupp AS, Davis JS. Protein Kinase A and 5' AMP-Activated Protein Kinase Signaling Pathways Exert Opposite Effects on Induction of Autophagy in Luteal Cells. Front Cell Dev Biol 2021; 9:723563. [PMID: 34820368 PMCID: PMC8607825 DOI: 10.3389/fcell.2021.723563] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 10/22/2021] [Indexed: 12/21/2022] Open
Abstract
In the absence of pregnancy the ovarian corpus luteum undergoes regression, a process characterized by decreased production of progesterone and structural luteolysis involving apoptosis. Autophagy has been observed in the corpus luteum during luteal regression. Autophagy is a self-degradative process important for balancing sources of cellular energy at critical times in development and in response to nutrient stress, but it can also lead to apoptosis. Mechanistic target of rapamycin (MTOR) and 5′ AMP-activated protein kinase (AMPK), key players in autophagy, are known to inhibit or activate autophagy, respectively. Here, we analyzed the signaling pathways regulating the initiation of autophagy in bovine luteal cells. In vivo studies showed increased activating phosphorylation of AMPKα (Thr172) and elevated content of LC3B, a known marker of autophagy, in luteal tissue during PGF2α-induced luteolysis. In vitro, AMPK activators 1) stimulated phosphorylation of regulatory associated protein of MTOR (RPTOR) leading to decreased activity of MTOR, 2) increased phosphorylation of Unc-51-Like Kinase 1 (ULK1) and Beclin 1 (BECN1), at sites specific for AMPK and required for autophagy initiation, 3) increased levels of LC3B, and 4) enhanced colocalization of autophagosomes with lysosomes indicating elevated autophagy. In contrast, LH/PKA signaling in luteal cells 1) reduced activation of AMPKα and phosphorylation of RPTOR, 2) elevated MTOR activity, 3) stimulated phosphorylation of ULK1 at site required for ULK1 inactivation, and 4) inhibited autophagosome formation as reflected by reduced content of LC3B-II. Pretreatment with AICAR, a pharmacological activator of AMPK, inhibited LH-mediated effects on RPTOR, ULK1 and BECN1. Our results indicate that luteotrophic signaling via LH/PKA/MTOR inhibits, while luteolytic signaling via PGF2α/Ca2+/AMPK activates key signaling pathways involved in luteal cell autophagy.
Collapse
Affiliation(s)
- Emilia Przygrodzka
- Department of Obstetrics and Gynecology, Olson Center for Women's Health, University of Nebraska Medical Center, Omaha, NE, United States
| | - Corrine F Monaco
- Department of Obstetrics and Gynecology, Olson Center for Women's Health, University of Nebraska Medical Center, Omaha, NE, United States.,Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Michele R Plewes
- Department of Obstetrics and Gynecology, Olson Center for Women's Health, University of Nebraska Medical Center, Omaha, NE, United States.,Veterans Affairs Nebraska Western Iowa Health Care System, Omaha, NE, United States
| | - Guojuan Li
- Department of Obstetrics and Gynecology, Olson Center for Women's Health, University of Nebraska Medical Center, Omaha, NE, United States
| | - Jennifer R Wood
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Andrea S Cupp
- Department of Obstetrics and Gynecology, Olson Center for Women's Health, University of Nebraska Medical Center, Omaha, NE, United States.,Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - John S Davis
- Department of Obstetrics and Gynecology, Olson Center for Women's Health, University of Nebraska Medical Center, Omaha, NE, United States.,Veterans Affairs Nebraska Western Iowa Health Care System, Omaha, NE, United States
| |
Collapse
|
14
|
Przygrodzka E, Plewes MR, Davis JS. Luteinizing Hormone Regulation of Inter-Organelle Communication and Fate of the Corpus Luteum. Int J Mol Sci 2021; 22:9972. [PMID: 34576135 PMCID: PMC8470545 DOI: 10.3390/ijms22189972] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/10/2021] [Accepted: 09/12/2021] [Indexed: 12/18/2022] Open
Abstract
The corpus luteum is an endocrine gland that synthesizes the steroid hormone progesterone. luteinizing hormone (LH) is a key luteotropic hormone that stimulates ovulation, luteal development, progesterone biosynthesis, and maintenance of the corpus luteum. Luteotropic and luteolytic factors precisely regulate luteal structure and function; yet, despite recent scientific progress within the past few years, the exact mechanisms remain largely unknown. In the present review, we summarize the recent progress towards understanding cellular changes induced by LH in steroidogenic luteal cells. Herein, we will focus on the effects of LH on inter-organelle communication and steroid biosynthesis, and how LH regulates key protein kinases (i.e., AMPK and MTOR) responsible for controlling steroidogenesis and autophagy in luteal cells.
Collapse
Affiliation(s)
- Emilia Przygrodzka
- Olson Center for Women’s Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Nebraska Medical Center, Omaha, NE 68198-3255, USA; (E.P.); (M.R.P.)
| | - Michele R. Plewes
- Olson Center for Women’s Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Nebraska Medical Center, Omaha, NE 68198-3255, USA; (E.P.); (M.R.P.)
- Veterans Affairs Nebraska Western Iowa Health Care System, 4101 Woolworth Ave, Omaha, NE 68105, USA
| | - John S. Davis
- Olson Center for Women’s Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Nebraska Medical Center, Omaha, NE 68198-3255, USA; (E.P.); (M.R.P.)
- Veterans Affairs Nebraska Western Iowa Health Care System, 4101 Woolworth Ave, Omaha, NE 68105, USA
| |
Collapse
|
15
|
Peters AE, Caban SJ, McLaughlin EA, Roman SD, Bromfield EG, Nixon B, Sutherland JM. The Impact of Aging on Macroautophagy in the Pre-ovulatory Mouse Oocyte. Front Cell Dev Biol 2021; 9:691826. [PMID: 34268312 PMCID: PMC8277196 DOI: 10.3389/fcell.2021.691826] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/04/2021] [Indexed: 12/13/2022] Open
Abstract
Accompanying the precipitous age-related decline in human female fertility is an increase in the proportion of poor-quality oocytes within the ovary. The macroautophagy pathway, an essential protein degradation mechanism responsible for maintaining cell health, has not yet been thoroughly investigated in this phenomenon. The aim of this study was to characterize the macroautophagy pathway in an established mouse model of oocyte aging using in-depth image analysis-based methods and to determine mechanisms that account for the observed changes. Three autophagy pathway markers were selected for assessment of gene and protein expression in this model: Beclin 1; an initiator of autophagosome formation, Microtubule-associated protein 1 light chain 3B; a constituent of the autophagosome membrane, and lysosomal-associated membrane protein 1; a constituent of the lysosome membrane. Through quantitative image analysis of immunolabeled oocytes, this study revealed impairment of the macroautophagy pathway in the aged oocyte with an attenuation of both autophagosome and lysosome number. Additionally, an accumulation of amphisomes greater than 10 μm2 in area were observed in aging oocytes, and this accumulation was mimicked in oocytes treated with lysosomal inhibitor chloroquine. Overall, these findings implicate lysosomal dysfunction as a prominent mechanism by which these age-related changes may occur and highlight the importance of macroautophagy in maintaining mouse pre-ovulatory oocyte quality. This provides a basis for further investigation of dysfunctional autophagy in poor oocyte quality and for the development of therapeutic or preventative strategies to aid in the maintenance of pre-ovulatory oocyte health.
Collapse
Affiliation(s)
- Alexandra E Peters
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science & Pharmacy and Environmental & Life Sciences, University of Newcastle, Callaghan, NSW, Australia.,Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Shandelle J Caban
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science & Pharmacy and Environmental & Life Sciences, University of Newcastle, Callaghan, NSW, Australia.,Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Eileen A McLaughlin
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science & Pharmacy and Environmental & Life Sciences, University of Newcastle, Callaghan, NSW, Australia.,Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Science, Western Sydney University, Penrith, NSW, Australia.,School of Biological Sciences, Faculty of Science, The University of Auckland, Auckland, New Zealand
| | - Shaun D Roman
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science & Pharmacy and Environmental & Life Sciences, University of Newcastle, Callaghan, NSW, Australia.,Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,Priority Research Centre for Drug Development, The University of Newcastle, Callaghan, NSW, Australia
| | - Elizabeth G Bromfield
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science & Pharmacy and Environmental & Life Sciences, University of Newcastle, Callaghan, NSW, Australia.,Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Brett Nixon
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science & Pharmacy and Environmental & Life Sciences, University of Newcastle, Callaghan, NSW, Australia.,Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Jessie M Sutherland
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science & Pharmacy and Environmental & Life Sciences, University of Newcastle, Callaghan, NSW, Australia.,Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| |
Collapse
|
16
|
Xu F, Lawson MS, Campbell SP, Tkachenko OY, Park BS, Bishop CV, Xu J. Stage-dependent actions of antimüllerian hormone in regulating granulosa cell proliferation and follicular function in the primate ovary. F&S SCIENCE 2020; 1:161-171. [PMID: 34355206 PMCID: PMC8329754 DOI: 10.1016/j.xfss.2020.10.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To study the direct action and physiological role of antimüllerian hormone (AMH) in regulating ovarian follicular development and function in vivo in primates. DESIGN Animals were assigned to six treatment sequences in a crossover design study. Intraovarian infusion was performed during the follicular phase of the menstrual cycle with agents including: control vehicle; recombinant human AMH (rhAMH); and neutralizing anti-human AMH antibody (AMHAb). Before ovariectomy after the final treatment, the animals received intravenous injections of bromodeoxyuridine (BrdU). SETTING National primate research center. ANIMALS Adult female rhesus macaques (Macaca mulatta). INTERVENTIONS None. MAIN OUTCOME MEASURES Cycle length, follicle cohorts, and serum steroid levels were assessed. Ovarian histology, as well as granulosa cell (GC) proliferation and oocyte viability, were evaluated. RESULTS In vehicle-infused ovaries, a dominant follicle was observed at midcycle E2 peak. However, rhAMH-treated ovaries exhibited an increased number of small antral follicles, whereas AMHAb-treated ovaries developed multiple large antral follicles. Serum E2 levels in the follicular phase decreased after rhAMH infusion and increased after AMHAb infusion. The rhAMH infusion increased serum T levels. Whereas early-growing follicles of rhAMH-treated ovaries contained BrdU-positive GCs, antral follicles containing BrdU-positive GCs were identified in AMHAb-treated ovaries. Autophagy was observed in oocytes of early-growing and antral follicles exposed to AMHAb and rhAMH, respectively. CONCLUSIONS AMH enhanced early-stage follicle growth, but prevented antral follicle development and function via its stage-dependent regulation of GC proliferation and oocyte viability. This study provides information relevant to the pathophysiology of ovarian dysfunction and the treatment of infertility.
Collapse
Affiliation(s)
- Fuhua Xu
- Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology, School of Medicine, Oregon Health & Science University, Portland
| | - Maralee S. Lawson
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton
| | - Shawn P. Campbell
- Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology, School of Medicine, Oregon Health & Science University, Portland
| | - Olena Y. Tkachenko
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton
| | - Byung S. Park
- OHSU-PSU School of Public Health, Oregon Health & Science University, Portland
| | - Cecily V. Bishop
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton
- Department of Animal and Rangeland Sciences, College of Agriculture, Oregon State University, Corvallis, Oregon
| | - Jing Xu
- Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology, School of Medicine, Oregon Health & Science University, Portland
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton
| |
Collapse
|
17
|
Wang Z, El Zowalaty AE, Li Y, Andersen CL, Ye X. Association of luteal cell degeneration and progesterone deficiency with lysosomal storage disorder mucolipidosis type IV in Mcoln1-/- mouse model†. Biol Reprod 2020; 101:782-790. [PMID: 31317194 DOI: 10.1093/biolre/ioz126] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 04/13/2019] [Accepted: 07/11/2019] [Indexed: 12/20/2022] Open
Abstract
Transient receptor potential cation channel, mucolipin subfamily, member 1 (TRPML1) (MCOLN1/Mcoln1) is a lysosomal counter ion channel. Mutations in MCOLN1 cause mucolipidosis type IV (MLIV), a progressive and severe lysosomal storage disorder with a slow onset. Mcoln1-/- mice recapitulate typical MLIV phenotypes but roles of TRPML1 in female reproduction are unknown. Despite normal mating activities, Mcoln1-/- female mice had reduced fertility at 2 months old and quickly became infertile at 5 months old. Progesterone deficiency was detected on 4.5 days post coitum/gestation day 4.5 (D4.5). Immunohistochemistry revealed TRPML1 expression in luteal cells of wild type corpus luteum (CL). Corpus luteum formation was not impaired in 5-6 months old Mcoln1-/- females indicated by comparable CL numbers in control and Mcoln1-/- ovaries on both D1.5 and D4.5. In the 5-6 months old Mcoln1-/- ovaries, histology revealed less defined corpus luteal cord formation, extensive luteal cell vacuolization and degeneration; immunofluorescence revealed disorganized staining of collagen IV, a basal lamina marker for endothelial cells; Nile Red staining detected lipid droplet accumulation, a typical phenotype of MLIV; immunofluorescence of heat shock protein 60 (HSP60, a mitochondrial marker) and in situ hybridization of steroidogenic acute regulatory protein (StAR, for the rate-limiting step of steroidogenesis) showed reduced expression of HSP60 and StAR, indicating impaired mitochondrial functions. Luteal cell degeneration and impaired mitochondrial functions can both contribute to progesterone deficiency in the Mcoln1-/- mice. This study demonstrates a novel function of TRPML1 in maintaining CL luteal cell integrity and function.
Collapse
Affiliation(s)
- Zidao Wang
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA.,Interdisciplinary Toxicology Program, University of Georgia, Athens, Georgia, USA
| | - Ahmed E El Zowalaty
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA.,Interdisciplinary Toxicology Program, University of Georgia, Athens, Georgia, USA
| | - Yuehuan Li
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Christian L Andersen
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA.,Interdisciplinary Toxicology Program, University of Georgia, Athens, Georgia, USA
| | - Xiaoqin Ye
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA.,Interdisciplinary Toxicology Program, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
18
|
A dual death/survival role of autophagy in the adult ovary of Lagostomus maximus (Mammalia- Rodentia). PLoS One 2020; 15:e0232819. [PMID: 32469908 PMCID: PMC7259749 DOI: 10.1371/journal.pone.0232819] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 04/22/2020] [Indexed: 12/16/2022] Open
Abstract
Follicular atresia is a cell death event that occurs in the great majority of follicles before ovulation in the mature mammalian ovary. Germ cell loss has been mainly associated to apoptosis although autophagy also seems to be at play. Aimed to increase our understanding on the possible cooperating role of autophagy and apoptosis in follicular atresia and/or follicular survival, we analyzed both programmed cell death mechanisms in a rodent model, the South American plains vizcacha, Lagostomus maximus. Female vizcacha shows highly suppressed apoptosis-dependent follicular atresia in the adult ovary, with continuous folliculogenesis and massive polyovulation. This strategy of massive ovulation requires a permanent remodeling of the ovarian architecture to maintain the availability of quiescent primordial follicles throughout the individual's reproductive lifespan. We report here our analysis of autophagy (BECN1, LAMP1 and LC3B-I/II) and apoptosis (BCL2 and ACTIVE CASPASE-3) markers which revealed interactive behaviors between both processes, with autophagy promoting survival or cell death depending on the ovarian structure. Strong BECN1, LC3B-II and LAMP1 staining was observed in atretic follicles and degenerating corpora lutea that also expressed nuclear ACTIVE CASPASE-3. Healthy follicles showed a slight expression of autophagy proteins but a strong expression of BCL2 and no detectable ACTIVE CASPASE-3. Transmission electron microscopy revealed a high formation of autophagosomes, autolysosomes and lysosomes in atretic follicles and degenerating corpora lutea and a low number of autophagic vesicles in normal follicles. The co-expression of LC3B-BECN1, LC3B-LAMP1 and LC3B-ACTIVE CASPASE-3 was only detected in atretic follicles and degenerating corpora lutea, while co-expression of BCL2-BECN1 was only observed in normal follicles. We propose that autophagy could act as a mechanism to eliminate altered follicles and remnant corpora lutea providing the necessary space for maturation of primordial follicles that continuously enter the growing follicular pool to sustain massive ovulation.
Collapse
|
19
|
Escobar ML, Echeverria OM, Palacios-Martínez S, Juárez-Chavero S, Sánchez-Sánchez L, Vázquez-Nin GH. Beclin 1 Interacts With Active Caspase-3 and Bax in Oocytes From Atretic Follicles in the Rat Ovary. J Histochem Cytochem 2019; 67:873-889. [PMID: 31583941 PMCID: PMC6882064 DOI: 10.1369/0022155419881127] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Oocyte cell death is a normal process in the mammalian ovary during follicular growth. Recent reports have demonstrated the presence of pro-apoptotic and pro-autophagic proteins during oocyte elimination. The goal of this study was to identify the interactions between proteins involved in different types of programmed cell death in the same oocyte during follicular atresia. We evaluated the presence of Beclin 1 and its interaction with the pro-apoptotic proteins active caspase-3, Bax, and Bak by means of histochemical observations, ultrastructural immunodetection, and immunoprecipitation techniques in ovaries from prepubertal (28- and 33-day-old), juvenile (40-day-old), and young adult (90-day-old) rats. In this study, we identified that oocyte elimination occurred with a high quantity of pro-autophagic protein Beclin 1 and increased the presence of the pro-apoptotic proteins active caspase-3, Bax, and Bak. Conversely, the antiapoptotic protein Bcl-2 was reduced in oocytes from atretic follicles. In addition, Beclin 1 was shown to interact with active caspase-3 and Bax. Our results suggest that the increase in Beclin 1 is directly related to the rise of pro-apoptotic proteins, which could promote the apoptotic process during oocyte elimination.
Collapse
Affiliation(s)
- María L. Escobar
- María L. Escobar, Lab. Microscopía Electrónica, Departamento de Biología Celular, Facultad de Ciencias, Universidad Nacional Autónoma de México, Av. Universidad 3000, Ciudad Universitaria, 04510 Ciudad de México, México. E-mail:
| | - Olga M. Echeverria
- Lab. Microscopía Electrónica, Departamento de Biología Celular, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Sebastián Palacios-Martínez
- Lab. Microscopía Electrónica, Departamento de Biología Celular, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Silvia Juárez-Chavero
- Lab. Microscopía Electrónica, Departamento de Biología Celular, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Luis Sánchez-Sánchez
- Lab. Biología Molecular del Cáncer, Laboratorio 6, 2º piso. UMIEZ, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Gerardo H. Vázquez-Nin
- Lab. Microscopía Electrónica, Departamento de Biología Celular, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, México
| |
Collapse
|
20
|
Yadav AK, Yadav PK, Chaudhary GR, Tiwari M, Gupta A, Sharma A, Pandey AN, Pandey AK, Chaube SK. Autophagy in hypoxic ovary. Cell Mol Life Sci 2019; 76:3311-3322. [PMID: 31062072 PMCID: PMC11105528 DOI: 10.1007/s00018-019-03122-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 03/30/2019] [Accepted: 04/29/2019] [Indexed: 12/19/2022]
Abstract
Oxygen deprivation affects human health by modulating system as well as cellular physiology. Hypoxia generates reactive oxygen species (ROS), causes oxidative stress and affects female reproductive health by altering ovarian as well as oocyte physiology in mammals. Hypoxic conditions lead to several degenerative changes by inducing various cell death pathways like autophagy, apoptosis and necrosis in the follicle of mammalian ovary. The encircling somatic cell death interrupts supply of nutrients to the oocyte and nutrient deprivation may result in the generation of ROS. Increased level of ROS could induce granulosa cells as well as oocyte autophagy. Although autophagy removes damaged proteins and subcellular organelles to maintain the cell survival, irreparable damages could induce cell death within intra-follicular microenvironment. Hypoxia-induced autophagy is operated through 5' AMP activated protein kinase-mammalian target of rapamycin, endoplasmic reticulum stress/unfolded protein response and protein kinase C delta-c-junN terminal kinase 1 pathways in a wide variety of somatic cell types. Similar to somatic cells, we propose that hypoxia may induce granulosa cell as well as oocyte autophagy and it could be responsible at least in part for germ cell elimination from mammalian ovary. Hypoxia-mediated germ cell depletion may cause several reproductive impairments including early menopause in mammals.
Collapse
Affiliation(s)
- Anil Kumar Yadav
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Pramod K Yadav
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Govind R Chaudhary
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Meenakshi Tiwari
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Anumegha Gupta
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Alka Sharma
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Ashutosh N Pandey
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Ajai K Pandey
- Department of Kayachikitsa, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Shail K Chaube
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
21
|
Sperm quality in fish: Determinants and affecting factors. Theriogenology 2019; 135:94-108. [PMID: 31203093 DOI: 10.1016/j.theriogenology.2019.06.009] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 06/05/2019] [Indexed: 11/23/2022]
Abstract
Fish sperm quality assessment is helpful for optimizing production and for monitoring the environmental state. Sperm can be monitored relatively easy and, to date, various analyses have been applied and proven to be helpful in this task. Among them, sperm motility parameters such as sperm speed are one of the main performance traits during assisted fish reproduction. Apart from motility the sperm concentration, volume, and seminal plasma pH and osmolality are also frequently evaluated and are the main sperm quality indicators measured in fish sperm. However, other parameters also determine sperm fertilization potential. Recent knowledge reveals several additional parameters of high importance for sperm function. Among them are DNA integration, membrane stability, mitochondria status and enzymatic activity. Measuring all these parameters in fish sperm provides complex knowledge regarding male fertility and helps to improve broodstock maintenance protocols as well as gamete handling and fertilization processes. This review focuses on the presentation of the sperm quality measures for freshwater and marine species of the fish and provides information regarding recent methods of sperm quality evaluation.
Collapse
|
22
|
Maternal β-hydroxy-β-methylbutyrate (HMB) supplementation during pregnancy affects early folliculogenesis in the ovary of newborn piglets. Theriogenology 2019; 128:91-100. [PMID: 30743108 DOI: 10.1016/j.theriogenology.2019.02.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 01/15/2019] [Accepted: 02/01/2019] [Indexed: 12/22/2022]
Abstract
Beta-hydroxy-beta-methylbutyrate (HMB) is a leucine metabolite with protein anabolic effects. This study was designed to determine whether prenatal HMB treatment has an effect on oogenesis and folliculogenesis in the ovary of newborn piglets. HMB decreased the number of egg nests and primordial follicles and increased the pool of developing follicles compared to the control group. Although the percentage of TUNEL-positive oocytes within the egg nests was higher in HMB-treated group no increase in the Bax/Bcl-2 ratio and active caspase-3 expression was observed. Moreover, the granulosa cell proliferation index and StAR protein expression were higher in HMB-treated group. In contrast to the control group, the expression of E-cadherins was reduced after the HMB treatment. In addition, a significant increase in the serum level of gonadotropins and steroid hormones was detected in HMB-treated piglets. In conclusion, prenatal HMB treatment dysregulates hormonal homeostasis which impairs early folliculogenesis in piglets.
Collapse
|
23
|
Zhou J, Peng X, Mei S. Autophagy in Ovarian Follicular Development and Atresia. Int J Biol Sci 2019; 15:726-737. [PMID: 30906205 PMCID: PMC6429023 DOI: 10.7150/ijbs.30369] [Citation(s) in RCA: 166] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 12/15/2018] [Indexed: 12/24/2022] Open
Abstract
Autophagy is a mechanism that exists in all eukaryotes under a variety of physiological and pathological conditions. In the mammalian ovaries, less than 1% of follicles ovulate, whereas the remaining 99% undergo follicular atresia. Autophagy and apoptosis have been previously found to be involved in the regulation of both primordial follicular development as well as atresia. The relationship between autophagy, follicular development, and atresia have been summarized in this review with the aim to obtain a more comprehensive understanding of the role played by autophagy in follicular development and atresia.
Collapse
Affiliation(s)
- Jiawei Zhou
- Institute of Animal Science and Veterinary Medicine, Hubei Academy of Agricultural Sciences, Wuhan 430064, China.,Hubei Key Lab for Animal Embryo Engineering and Molecular Breeding, Wuhan 430064, China
| | - Xianwen Peng
- Institute of Animal Science and Veterinary Medicine, Hubei Academy of Agricultural Sciences, Wuhan 430064, China.,Hubei Key Lab for Animal Embryo Engineering and Molecular Breeding, Wuhan 430064, China
| | - Shuqi Mei
- Institute of Animal Science and Veterinary Medicine, Hubei Academy of Agricultural Sciences, Wuhan 430064, China.,Hubei Key Lab for Animal Embryo Engineering and Molecular Breeding, Wuhan 430064, China
| |
Collapse
|
24
|
Pajokh M, Talaei-Khozani T, Bordbar H, Mesbah F. Apoptosis, Autophagy, and Necrosis in Murine Embryonic Gonadal Ridges and Neonatal Ovaries: An Animal Model. IRANIAN JOURNAL OF MEDICAL SCIENCES 2019; 44:35-43. [PMID: 30666074 PMCID: PMC6330520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND In mammalian ovaries, loss of over two-thirds of germ cells happens due to cell death. Nonetheless, the exact mechanism of cell death has yet to be determined. The present basic practical study was designed to detect 3 types of programmed cell death, namely apoptosis, autophagy, and necrosis, in murine embryonic gonadal ridges and neonatal ovaries. METHODS Twenty gonadal ridges and ovaries from female mouse embryos 13.5 days post coitum and newborn mice 1 day postnatal were collected. The TUNEL assay was performed to evaluate apoptosis. The interplay of autophagy was evaluated by immunohistochemistry for beclin-1. Necrotic cell death was analyzed by propidium iodide (PI) staining. The count and percentage of the labeled oocytes in the gonadal ridges and ovaries were evaluated and compared using the independent t test and one-way ANOVA. A P value less than 0.05 was considered statistically significant. RESULTS We detected TUNEL-positive reaction in the embryonic germ cells and in the small and large oocytes of the neonatal ovaries. The germ cells and small oocytes reacted to beclin-1. PI absorption was detected in the embryonic germ cells and the large oocytes of the neonatal ovaries, but not in the small oocytes. The percentage of the TUNEL-positive and PI-labeled oocytes in the gonadal ridges was significantly higher than that in the neonatal ovaries (P<0.01 and P=0.01). In the neonatal ovaries, the percentage of the beclin-1-labeled oocytes was significantly higher than that in the embryonic phase (P<0.01). CONCLUSION We showed that all 3 types of programmed cell death, namely apoptosis, autophagy, and necrosis, accounted for embryonic and neonatal germ-cell loss. Our observations demonstrated a potential role for necrosis, particularly in the embryonic gonadal ridge in comparison to the neonatal ovary, in mice.
Collapse
|
25
|
Zhang Y, Mu X, Gao R, Geng Y, Liu X, Chen X, Wang Y, Ding Y, Wang Y, He J. Foetal-neonatal exposure of Di (2-ethylhexyl) phthalate disrupts ovarian development in mice by inducing autophagy. JOURNAL OF HAZARDOUS MATERIALS 2018; 358:101-112. [PMID: 29990796 DOI: 10.1016/j.jhazmat.2018.06.042] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 05/28/2018] [Accepted: 06/19/2018] [Indexed: 05/21/2023]
Abstract
The female reproductive lifespan is largely determined by the size of the primordial follicle pool, which is established early in life. We previously reported that Di (2-ethylhexyl) phthalate (DEHP), an environmental endocrine disruptor and a widely-spreading plasticizer, impairs primordial folliculogenesis. In the present study, we found DEHP significantly altered the number and sex ratio of the offspring of neonatal-exposed mice. Furthermore, by a neonatal exposure model and an ovary culture model, it showed that DEHP activated autophagy in the ovary, with increased autophagy-related gene expression and recognizable autophagosomes, while inhibition of autophagy by 3-MA attenuated the adverse impact of DEHP on primordial folliculogenesis. Moreover, key components of AMPK-SKP2-CARM1 signalling were up-regulated by DEHP in the ovary, and AMPK inhibitor Compound C reduced autophagy-related gene expression and partially recovered primordial follicle assembly. Collectively, this study demonstrates that DEHP induces autophagy by activating AMPK-SKP2-CARM1 signalling in mice perinatal ovaries, which results in disrupted primordial folliculogenesis and reduced female fertility.
Collapse
Affiliation(s)
- Yan Zhang
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing, 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China
| | - Xinyi Mu
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing, 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China
| | - Rufei Gao
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing, 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China
| | - Yanqing Geng
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing, 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China
| | - Xueqing Liu
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing, 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China
| | - Xuemei Chen
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing, 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China
| | - Yuheng Wang
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing, 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China
| | - Yubin Ding
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing, 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China
| | - Yingxiong Wang
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing, 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China
| | - Junlin He
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, Chongqing, 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, PR China.
| |
Collapse
|
26
|
Hale BJ, Hager CL, Seibert JT, Selsby JT, Baumgard LH, Keating AF, Ross JW. Heat stress induces autophagy in pig ovaries during follicular development. Biol Reprod 2018; 97:426-437. [PMID: 29025092 DOI: 10.1093/biolre/iox097] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Accepted: 08/24/2017] [Indexed: 12/23/2022] Open
Abstract
Hyperthermia or heat stress (HS) occurs when heat dissipation mechanisms are overwhelmed by external and internal heat production. Hyperthermia negatively affects reproduction and potentially compromises oocyte integrity and reduces developmental competence of ensuing embryos. Autophagy is the process by which cells recycle energy through the reutilization of cellular components and is activated by a variety of stressors. Study objectives were to characterize autophagy-related proteins in the ovary following cyclical HS during the follicular phase. Twelve gilts were synchronized and subjected to cyclical HS (n = 6) or thermal neutral (n = 6) conditions for 5 days during the follicular phase. Ovarian protein abundance of Beclin 1 and microtubule associated protein light chain 3 beta II were each elevated as a result of HS (P = 0.001 and 0.003, respectively). The abundance of the autophagy related (ATG)12-ATG5 complex was decreased as a result of HS (P = 0.002). Regulation of autophagy and apoptosis occurs in tight coordination, and B-cell lymphoma (BCL)2 and BCL2L1 are involved in regulating both processes. BCL2L1 protein abundance, as detected via immunofluorescence, was increased in both the oocyte (∼1.6-fold; P < 0.01) and granulosa cells of primary follicles (∼1.4-fold P < 0.05) of HS ovaries. These results suggest that ovarian autophagy induction occurs in response to HS during the follicular phase, and that HS increases anti-apoptotic signaling in oocytes and early follicles. These data contribute to the biological understanding of how HS acts as an environmental stress to affect follicular development and negatively impact reproduction.
Collapse
Affiliation(s)
- Benjamin J Hale
- Department of Animal Science, Iowa State University, Ames, Iowa, USA
| | - Candice L Hager
- Department of Animal Science, Iowa State University, Ames, Iowa, USA
| | - Jacob T Seibert
- Department of Animal Science, Iowa State University, Ames, Iowa, USA
| | - Joshua T Selsby
- Department of Animal Science, Iowa State University, Ames, Iowa, USA
| | - Lance H Baumgard
- Department of Animal Science, Iowa State University, Ames, Iowa, USA
| | - Aileen F Keating
- Department of Animal Science, Iowa State University, Ames, Iowa, USA
| | - Jason W Ross
- Department of Animal Science, Iowa State University, Ames, Iowa, USA
| |
Collapse
|
27
|
Yadav PK, Tiwari M, Gupta A, Sharma A, Prasad S, Pandey AN, Chaube SK. Germ cell depletion from mammalian ovary: possible involvement of apoptosis and autophagy. J Biomed Sci 2018; 25:36. [PMID: 29681242 PMCID: PMC5911955 DOI: 10.1186/s12929-018-0438-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 04/17/2018] [Indexed: 12/19/2022] Open
Abstract
Mammalian ovary contains millions of germ cells during embryonic life but only few of them are culminated into oocytes that achieve meiotic competency just prior to ovulation. The majority of germ cells are depleted from ovary through several pathways. Follicular atresia is one of the major events that eliminate germ cells from ovary by engaging apoptotic as well as non-apoptotic pathways of programmed cell death. Apoptosis is characterized by several morphological changes that include cell shrinkage, nuclear condensation, membrane blebbing and cytoplasmic fragmentation by both mitochondria- as well as death receptor-mediated pathways in encircling granulosa cells and oocyte. Although necroapoptosis have been implicated in germ cell depletion, autophagy seems to play an active role in the life and death decisions of ovarian follicles. Autophagy is morphologically characterized by intracellular reorganization of membranes and increased number of autophagic vesicles that engulf bulk cytoplasm as well as organelles. Autophagy begins with the encapsulation of cytoplasmic constituents in a membrane sac known as autophagosomes. The autophagic vesicles are then destroyed by the lysosomal enzymes such as hydrolases that results in follicular atresia. It seems that apoptosis as well as autophagy could play active roles in germ cells depletion from ovary. Hence, it is important to prevent these two pathways in order to retain the germ cells in ovary of several mammalian species that are either threatened or at the verge of extinction. The involvement of apoptosis and autophagy in germ cell depletion from mammalian ovary is reviewed and possible pathways have been proposed.
Collapse
Affiliation(s)
- Pramod K Yadav
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, UP, India
| | - Meenakshi Tiwari
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, UP, India
| | - Anumegha Gupta
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, UP, India
| | - Alka Sharma
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, UP, India
| | - Shilpa Prasad
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, UP, India
| | - Ashutosh N Pandey
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, UP, India
| | - Shail K Chaube
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, UP, India.
| |
Collapse
|
28
|
Mao Z, Yang L, Lu X, Tan A, Wang Y, Ding F, Xiao L, Qi X, Yu Y. C1QTNF3 in the murine ovary and its function in folliculogenesis. Reproduction 2018; 155:333-346. [DOI: 10.1530/rep-17-0783] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 02/01/2018] [Indexed: 12/27/2022]
Abstract
C1q/tumor necrosis factor-related protein 3 (C1QTNF3) is a novel adipokine with modulating effects on metabolism, inflammation and the cardiovascular system. C1QTNF3 expression levels in the sera and omental adipose tissues of women with PCOS are low compared to control subjects. However, the expression and function of C1QTNF3 in the ovary has not previously been examined. Here, we assessed the expression patterns of C1qtnf3 in the ovary and explored its role in folliculogenesis. The C1qtnf3 transcript abundance was higher in large follicles than in small follicles and was under the influence of gonadotropin. C1QTNF3 was detected mainly in the granulosa cells and oocytes of growing follicles and modestly in the granulosa cells of atretic follicles and in luteal cells. Excess androgen significantly decreased C1QTNF3 expression in the ovaries in vivo and in granulosa cells in vitro. Recombinant C1QTNF3 protein accelerated the weight gain of ovarian explants and the growth of preantral follicles induced by follicle stimulating hormone (FSH) in vitro. The stimulatory effect of C1QTNF3 on ovarian growth was accompanied by the initiation of AKT, mTOR, p70S6K and 4EBP1 phosphorylation, an increase in CCND2 expression and a reduction in cleaved CASP3 levels. Moreover, the addition of C1QTNF3 accelerated proliferation and reduced activated CASP3/7 activity in granulosa cells. In vivo, the ovarian intrabursal administration of the C1QTNF3 antibody delayed gonadotropin-induced antral follicle development. Taken together, our data demonstrate that C1QTNF3 is an intraovarian factor that promotes follicle growth by accelerating proliferation, decelerating apoptosis and promoting AKT/mTOR phosphorylation.
Collapse
|
29
|
Liu Y, Wang YL, He SW, Chen MH, Zhang Z, Fu XP, Fu BB, Liao BQ, Lin YH, Qi ZQ, Wang HL. Protective effects of resveratrol against mancozeb induced apoptosis damage in mouse oocytes. Oncotarget 2018; 8:6233-6245. [PMID: 28031523 PMCID: PMC5351627 DOI: 10.18632/oncotarget.14056] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 12/13/2016] [Indexed: 11/25/2022] Open
Abstract
Mancozeb, a mixture of ethylene-bis-dithiocarbamate manganese and zinc salts, is one of the most widely used fungicides in agriculture. Mancozeb could lead to mitochondria dysfunction, cellular anti-oxidation enzymes depletion and apoptotic pathways activation. Previous studies indicated the exposure of mancozeb through mother would lead to irregular estrous cycles, decreased progesterone levels, reduced litter sizes, and more frequent delivery of dead fetuses. In this study, we investigated mancozeb inducing reproductive toxicity, especially focusing on its apoptotic effect and epigenetic modifications. We also showed that resveratrol, a kind of phytoalexin found in peanuts and grapes, can alleviate mancozeb's adverse effects, such as declined fertility, decreased ovary weight and primary follicles. Besides, mancozeb treated oocytes displayed suboptimal developmental competence and this can also be improved by treatment of resveratrol. More detailed investigation of these processes revealed that mancozeb increased reactive oxygen species, causing cell apoptosis and abnormal epigenetic modifications, and resveratrol can block these cytotoxic changes. Collectively, our results showed that resveratrol can alleviate mancozeb induced infertility and this was mainly through the correction of apoptotic tendency and the abnormity of cellular epigenetic modification.
Collapse
Affiliation(s)
- Yu Liu
- Organ Transplantation Institute, Medical College, Xiamen University, Xiamen City, Fujian Province, China.,Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen City, Fujian Province, China
| | - Ya-Long Wang
- Organ Transplantation Institute, Medical College, Xiamen University, Xiamen City, Fujian Province, China.,Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen City, Fujian Province, China
| | - Shu-Wen He
- Organ Transplantation Institute, Medical College, Xiamen University, Xiamen City, Fujian Province, China.,Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen City, Fujian Province, China
| | - Ming-Huang Chen
- Department of Gynaecology and Obstetrics, Zhongshan Hospital, Xiamen University, Xiamen City, Fujian Province, China
| | - Zhen Zhang
- Xiamen Institute for Food and Drug Quality Control, Xiamen City, Fujian Province, China
| | - Xian-Pei Fu
- Organ Transplantation Institute, Medical College, Xiamen University, Xiamen City, Fujian Province, China.,Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen City, Fujian Province, China
| | - Bin-Bin Fu
- Organ Transplantation Institute, Medical College, Xiamen University, Xiamen City, Fujian Province, China.,Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen City, Fujian Province, China
| | - Bao-Qiong Liao
- Organ Transplantation Institute, Medical College, Xiamen University, Xiamen City, Fujian Province, China.,Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen City, Fujian Province, China
| | - Yan-Hong Lin
- Department of Gynecology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Zhong-Quan Qi
- Organ Transplantation Institute, Medical College, Xiamen University, Xiamen City, Fujian Province, China.,Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen City, Fujian Province, China
| | - Hai-Long Wang
- Organ Transplantation Institute, Medical College, Xiamen University, Xiamen City, Fujian Province, China.,Fujian Key Laboratory of Organ and Tissue Regeneration, Xiamen City, Fujian Province, China
| |
Collapse
|
30
|
Sun YC, Wang YY, Sun XF, Cheng SF, Li L, Zhao Y, Shen W, Chen H. The role of autophagy during murine primordial follicle assembly. Aging (Albany NY) 2018; 10:197-211. [PMID: 29410391 PMCID: PMC5842841 DOI: 10.18632/aging.101376] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 01/30/2018] [Indexed: 11/25/2022]
Abstract
It is generally accepted that significant germ cell loss occurs during the establishment of the primordial follicle pool in most mammalian ovaries around the time of birth. However, the underlying mechanisms responsible for these processes remain largely unknown. In this investigation, we explored the role of autophagy during the establishment of the primordial follicle pool and found that autophagy was active in this process. Our data suggested that 17.5 dpc ovaries treated with rapamycin displayed a delay in germ cell cyst breakdown resulting in more oocytes at day 5 of treatment, while, ovaries that treated with 3-MA showed the opposite effect. We found that rapamycin treatment promoted autophagy and depressed cell apoptosis increasing the number of NOBOX positive oocytes. Furthermore, our results also revealed that epigenetic regulator, Sirt1, plays a role in germ cell loss. An epigenetic inhibitor or RNAi treatment of Sirt1, showed an increased level of H4K16ac and a decreased level of autophagy. Thus, these data indicate that autophagy prevents germ cell over loss during the establishment of primordial follicle pool, and this process may be influenced by Sirt1-invovled epigenetic regulation.
Collapse
Affiliation(s)
- Yuan-Chao Sun
- College of Animal Science and Technology, Northwest A&F University, Shaanxi Key Laboratory of Molecular Biology for Agriculture, Yangling Shaanxi, China
- Institute of Reproductive Sciences, College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | - Yong-Yong Wang
- Department of Reproductive Medicine, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, China
| | - Xiao-Feng Sun
- Institute of Reproductive Sciences, College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | - Shun-Feng Cheng
- Institute of Reproductive Sciences, College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | - Lan Li
- Institute of Reproductive Sciences, College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | - Yong Zhao
- Institute of Reproductive Sciences, College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | - Wei Shen
- Institute of Reproductive Sciences, College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | - Hong Chen
- College of Animal Science and Technology, Northwest A&F University, Shaanxi Key Laboratory of Molecular Biology for Agriculture, Yangling Shaanxi, China
| |
Collapse
|
31
|
Cejko BI, Sarosiek B, Krejszeff S, Kowalski RK. Multiple collections of common carp Cyprinus carpio L. semen during the reproductive period and its effects on sperm quality. Anim Reprod Sci 2018; 188:178-188. [DOI: 10.1016/j.anireprosci.2017.12.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 12/01/2017] [Accepted: 12/03/2017] [Indexed: 11/30/2022]
|
32
|
Johansson HKL, Svingen T, Fowler PA, Vinggaard AM, Boberg J. Environmental influences on ovarian dysgenesis - developmental windows sensitive to chemical exposures. Nat Rev Endocrinol 2017; 13:400-414. [PMID: 28450750 DOI: 10.1038/nrendo.2017.36] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
A woman's reproductive health and ability to have children directly affect numerous aspects of her life, from personal well-being and socioeconomic standing, to morbidity and lifespan. In turn, reproductive health depends on the development of correctly functioning ovaries, a process that starts early during fetal life. Early disruption to ovarian programming can have long-lasting consequences, potentially manifesting as disease much later in adulthood. A growing body of evidence suggests that exposure to chemicals early in life, including endocrine-disrupting chemicals, can cause a range of disorders later in life, such as those described in the ovarian dysgenesis syndrome hypothesis. In this Review, we discuss four specific time windows during which the ovary is particularly sensitive to disruption by exogenous insults: gonadal sex determination, meiotic division, follicle assembly and the first wave of follicle recruitment. To date, most evidence points towards the germ cell lineage being the most vulnerable to chemical exposure, particularly meiotic division and follicle assembly. Environmental chemicals and pharmaceuticals, such as bisphenols or mild analgesics (including paracetamol), can also affect the somatic cell lineages. This Review summarizes our current knowledge pertaining to environmental chemicals and pharmaceuticals, and their potential contributions to the development of ovarian dysgenesis syndrome. We also highlight knowledge gaps that need addressing to safeguard female reproductive health.
Collapse
Affiliation(s)
- Hanna Katarina Lilith Johansson
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Kemitorvet, Building 202, DK-2800 Kgs. Lyngby, Denmark
| | - Terje Svingen
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Kemitorvet, Building 202, DK-2800 Kgs. Lyngby, Denmark
| | - Paul A Fowler
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Anne Marie Vinggaard
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Kemitorvet, Building 202, DK-2800 Kgs. Lyngby, Denmark
| | - Julie Boberg
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Kemitorvet, Building 202, DK-2800 Kgs. Lyngby, Denmark
| |
Collapse
|
33
|
Shen M, Jiang Y, Guan Z, Cao Y, Li L, Liu H, Sun SC. Protective mechanism of FSH against oxidative damage in mouse ovarian granulosa cells by repressing autophagy. Autophagy 2017; 13:1364-1385. [PMID: 28598230 DOI: 10.1080/15548627.2017.1327941] [Citation(s) in RCA: 150] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Oxidative stress-induced granulosa cell (GCs) death represents a common reason for follicular atresia. Follicle-stimulating hormone (FSH) has been shown to prevent GCs from oxidative injury, although the underlying mechanism remains to be elucidated. Here we first report that the suppression of autophagic cell death via some novel signaling effectors is engaged in FSH-mediated GCs protection against oxidative damage. The decline in GCs viability caused by oxidant injury was remarkably reduced following FSH treatment, along with impaired macroautophagic/autophagic flux under conditions of oxidative stress both in vivo and in vitro. Blocking of autophagy displayed similar levels of suppression in oxidant-induced cell death compared with FSH treatment, but FSH did not further improve survival of GCs pretreated with autophagy inhibitors. Further investigations revealed that activation of the phosphoinositide 3-kinase (PI3K)-AKT-MTOR (mechanistic target of rapamycin [serine/threonine kinase]) signaling pathway was required for FSH-mediated GCs survival from oxidative stress-induced autophagy. Additionally, the FSH-PI3K-AKT axis also downregulated the autophagic response by targeting FOXO1, whereas constitutive activation of FOXO1 in GCs not only abolished the protection from FSH, but also emancipated the autophagic process, from the protein level of MAP1LC3B-II to autophagic gene expression. Furthermore, FSH inhibited the production of acetylated FOXO1 and its interaction with Atg proteins, followed by a decreased level of autophagic cell death upon oxidative stress. Taken together, our findings suggest a new mechanism involving FSH-FOXO1 signaling in defense against oxidative damage to GCs by restraining autophagy, which may be a potential avenue for the clinical treatment of anovulatory disorders.
Collapse
Affiliation(s)
- Ming Shen
- a College of Animal Science and Technology , Nanjing Agricultural University , Nanjing , China
| | - Yi Jiang
- a College of Animal Science and Technology , Nanjing Agricultural University , Nanjing , China
| | - Zhiqiang Guan
- a College of Animal Science and Technology , Nanjing Agricultural University , Nanjing , China
| | - Yan Cao
- a College of Animal Science and Technology , Nanjing Agricultural University , Nanjing , China
| | - Liechuan Li
- a College of Animal Science and Technology , Nanjing Agricultural University , Nanjing , China
| | - Honglin Liu
- a College of Animal Science and Technology , Nanjing Agricultural University , Nanjing , China
| | - Shao-Chen Sun
- a College of Animal Science and Technology , Nanjing Agricultural University , Nanjing , China
| |
Collapse
|
34
|
Danilović Luković J, Korać A, Milošević I, Lužajić T, Milanović Z, Kovačević Filipović M, Radovanović A. Z-cells and oogonia/oocytes in the advanced process of autophagy are the dominant altered cells in the ovaries of hypothyroid newborn rats. ACTA VET-BEOGRAD 2017. [DOI: 10.1515/acve-2017-0009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Abstract
Induced prenatal hypothyroidism in rat pups leads to accelerated primordial follicle assembly and premature follicular atresia with ovary failure. This work investigates the influence of maternal hypothyroidism induced with 6-n-propyl-2-thyouracil (PTU) on the number and morphology of oogonia/oocytes in newborn rat pups with light and transmission electron microscopy. Expression of apoptosis and autophagy markers in oogonia/oocytes were examined using immunohistochemistry. Hypothyroid newborn pups had a decreased number of mitotic and resting oogonia, while the number of altered oogonia/oocytes was increased. Ultrastructural observations revealed the increased presence of degenerated pachytene oocytes (Z-cells) and oogonia/oocytes undergoing autophagy, apoptosis and combined apoptosis and autophagy, in this group. The most abundant altered oogonia/oocytes in the hypothyroid group were those with morphological features of advanced autophagy and Z-cells. The percentage of TUNEL (terminal deoxynucleotidyl transferase dUTP nick end labeling) positive oogonia/oocytes was significantly lower in the hypothyroid group. No significant difference was recorded in the expression of caspase-3, ATG7 and LC3 possibly reflecting that these proteins were not involved in the oogonia/oocyte alteration process during prenatal rat hypothyroidism. The obtained results indicate that developmental hypothyroidism in the offspring enhances the number of Z-cells and oogonia/oocytes altered with the advanced process of autophagy.
Collapse
Affiliation(s)
- Jelena Danilović Luković
- State University of Novi Pazar, Department of Biomedical Sciences, Vuka Karadžića bb, 36300 Novi Pazar, Serbia
| | - Aleksandra Korać
- University of Belgrade, Center for Electron Microscopy, Faculty of Biology, Studentski trg 16, 11000 Belgrade, Serbia
| | - Ivan Milošević
- University of Belgrade, Faculty of Veterinary Medicine, Bulevar oslobođenja 18, 11000 Belgrade, Serbia
| | - Tijana Lužajić
- University of Belgrade, Faculty of Veterinary Medicine, Bulevar oslobođenja 18, 11000 Belgrade, Serbia
| | - Zorana Milanović
- University of Belgrade, Faculty of Veterinary Medicine, Bulevar oslobođenja 18, 11000 Belgrade, Serbia
| | | | - Anita Radovanović
- University of Belgrade, Faculty of Veterinary Medicine, Bulevar oslobođenja 18, 11000 Belgrade, Serbia
| |
Collapse
|
35
|
Sun YC, Sun XF, Dyce PW, Shen W, Chen H. The role of germ cell loss during primordial follicle assembly: a review of current advances. Int J Biol Sci 2017; 13:449-457. [PMID: 28529453 PMCID: PMC5436565 DOI: 10.7150/ijbs.18836] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 01/25/2017] [Indexed: 12/27/2022] Open
Abstract
In most female mammals, early germline development begins with the appearance of primordial germ cells (PGCs), and develops to form mature oocytes following several vital processes. It remains well accepted that significant germ cell apoptosis and oocyte loss takes place around the time of birth. The transition of the ovarian environment from fetal to neonatal, coincides with the loss of germ cells and the timing of follicle formation. All told it is common to lose approximately two thirds of germ cells during this transition period. The current consensus is that germ cell loss can be attributed, at least in part, to programmed cell death (PCD). Recently, autophagy has been implicated as playing a part in germ cell loss during the time of parturition. In this review, we discuss the major opinions and mechanisms of mammalian ovarian PCD during the process of germ cell loss. We also pay close attention to the function of autophagy in germ cell loss, and speculate that autophagy may also serve as a critical and necessary process during the establishment of primordial follicle pool.
Collapse
Affiliation(s)
- Yuan-Chao Sun
- College of Animal Science and Technology, Northwest A&F University, Shaanxi Key Laboratory of Molecular Biology for Agriculture, Yangling Shaanxi 712100, China
| | - Xiao-Feng Sun
- Institute of Reproductive Sciences, College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, China
| | - Paul W Dyce
- Department of Animal Sciences, Auburn University, Auburn, AL 36849, USA
| | - Wei Shen
- Institute of Reproductive Sciences, College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, China
| | - Hong Chen
- College of Animal Science and Technology, Northwest A&F University, Shaanxi Key Laboratory of Molecular Biology for Agriculture, Yangling Shaanxi 712100, China
| |
Collapse
|
36
|
Madden JA, Thomas PQ, Keating AF. Phosphoramide mustard induces autophagy markers and mTOR inhibition prevents follicle loss due to phosphoramide mustard exposure. Reprod Toxicol 2016; 67:65-78. [PMID: 27888070 DOI: 10.1016/j.reprotox.2016.11.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 11/15/2016] [Accepted: 11/21/2016] [Indexed: 01/07/2023]
Abstract
Phosphoramide mustard (PM) is an ovotoxic metabolite of cyclophosphamide. Postnatal day 4 Fisher 344 rat ovaries were exposed to vehicle control (1% DMSO) or PM (60μM)±LY294002 or rapamycin for 2 or 4 d. Transmission election microscopy revealed abnormally large golgi apparatus and electron dense mitochondria in PM-exposed ovaries prior to and at the time of follicle depletion. PM exposure increased (P<0.05) mRNA abundance of Bbc3, Cdkn1a, Ctfr, Edn1, Gstp1, Nqo1, Tlr4, Tnfrsfla, Txnrd1 and decreased (P<0.05) Casp1 and Il1b after 4d. PM exposure increased (P<0.1) BECN1 and LAMP, decreased (P<0.1) ABCB1 and did not alter ABCC1 protein. LY294002 did not impact PM-induced ovotoxicity, but decreased ABCC1 and ABCB1 protein. Rapamycin prevented PM-induced follicle loss. These data suggest that the mammalian target of rapamycin, mTOR, may be a gatekeeper of PM-induced follicle loss.
Collapse
Affiliation(s)
- Jill A Madden
- Department of Animal Science, Iowa State University, Ames, IA, 50011, United States
| | - Porsha Q Thomas
- Department of Animal Science, Iowa State University, Ames, IA, 50011, United States
| | - Aileen F Keating
- Department of Animal Science, Iowa State University, Ames, IA, 50011, United States.
| |
Collapse
|
37
|
Hułas-Stasiak M, Dobrowolski P, Tomaszewska E. Prenatally administered dexamethasone impairs folliculogenesis in spiny mouse offspring. Reprod Fertil Dev 2016; 28:1038-1048. [PMID: 25562684 DOI: 10.1071/rd14224] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 11/21/2014] [Indexed: 01/07/2023] Open
Abstract
This study was designed to determine whether prenatal dexamethasone treatment has an effect on follicular development and atresia in the ovary of spiny mouse (Acomys cahirinus) offspring. Dexamethasone (125µg kg-1 bodyweight per day) was administered to pregnant spiny mice from Day 20 of gestation to parturition. The processes of follicle loss were analysed using classical markers of apoptosis (terminal deoxynucleotidyl transferase-mediated dUTP nick end-labelling reaction, active caspase-3) and autophagy (Lamp1). The present study indicated that dexamethasone reduced the pool of healthy primordial follicles. Moreover, the oocytes from these follicles showed intensive caspase-3 and Lamp1 staining. Surprisingly, dexamethasone caused an increase in the number of secondary follicles; however, most of these follicles were characterised by extensive degeneration of the oocyte and caspase-3 and Lamp1 labelling. Western-blot analysis indicated that the glucocorticoid receptor as well as apoptosis and autophagy markers were more strongly expressed in the DEX-treated group than in the control. On the basis of these findings, we have concluded that dexamethasone impairs spiny mouse folliculogenesis and enhances follicular atresia through induction of autophagy or combined autophagy and apoptosis.
Collapse
Affiliation(s)
- Monika Hułas-Stasiak
- Department of Comparative Anatomy and Anthropology, Maria Curie-Sklodowska University, Akademicka St.19, 20-033 Lublin, Poland
| | - Piotr Dobrowolski
- Department of Comparative Anatomy and Anthropology, Maria Curie-Sklodowska University, Akademicka St.19, 20-033 Lublin, Poland
| | - Ewa Tomaszewska
- Department of Animal Biochemistry and Physiology, Faculty of Veterinary Medicine, University of Life Sciences, Akademicka St. 12, 20-950 Lublin, Poland
| |
Collapse
|
38
|
Yuan J, Zhang Y, Sheng Y, Fu X, Cheng H, Zhou R. MYBL2 guides autophagy suppressor VDAC2 in the developing ovary to inhibit autophagy through a complex of VDAC2-BECN1-BCL2L1 in mammals. Autophagy 2016; 11:1081-98. [PMID: 26060891 PMCID: PMC4590641 DOI: 10.1080/15548627.2015.1040970] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Oogenesis is essential for female gamete production in mammals. The total number of ovarian follicles is determined early in life and production of ovarian oocytes is thought to stop during the lifetime. However, the molecular mechanisms underling oogenesis, particularly autophagy regulation in the ovary, remain largely unknown. Here, we reveal an important MYBL2-VDAC2-BECN1-BCL2L1 pathway linking autophagy suppression in the developing ovary. The transcription factors GATA1 and MYBL2 can bind to and activate the Vdac2 promoter. MYBL2 regulates the spatiotemporal expression of VDAC2 in the developing ovary. Strikingly, in the VDAC2 transgenic pigs (Sus scrofa/Ss), VDAC2 exerts its function by inhibiting autophagy in the ovary. In contrast, Vdac2 knockout promotes autophagy. Moreover, VDAC2-mediated autophagy suppression is dependent on its interactions with both BECN1 and BCL2L1 to stabilize the BECN1 and BCL2L1 complex, suggesting VDAC2 as an autophagy suppressor in the pathway. Our findings provide a functional connection among the VDAC2, MYBL2, the BECN1-BCL2L1 pathway and autophagy suppression in the developing ovary, which is implicated in improving female fecundity.
Collapse
Key Words
- ATG12, autophagy-related 12
- ATG16L1, autophagy-related 16-like 1
- ATG5, autophagy-related 5
- BAK, BCL2-antagonist/killer 1
- BCL2, B-cell CLL/lymphoma 2
- BCL2L1, BCL2-like 1
- BECN1, Beclin 1, autophagy related
- Beclin1
- CDS, coding DNA sequence
- Dpp, days postpartum
- GATA1, GATA binding protein 1 (globin transcription factor 1)
- GATA2, GATA binding protein 2
- LC3B, microtubule-associated protein 1 light chain 3 beta
- MBS, MYBL2 binding site
- MYBL2
- MYBL2, v-myb avian myeloblastosis viral oncogene homolog-like 2
- SP1, Sp1 transcription factor
- VDAC, voltage-dependent anion channel
- VDAC2
- ovary
- reproduction
- transcription regulation
- wt, wild type
Collapse
Affiliation(s)
- Jia Yuan
- a Department of Genetics; College of Life Sciences; Wuhan University ; Wuhan , China
| | | | | | | | | | | |
Collapse
|
39
|
Bonnet A, Servin B, Mulsant P, Mandon-Pepin B. Spatio-Temporal Gene Expression Profiling during In Vivo Early Ovarian Folliculogenesis: Integrated Transcriptomic Study and Molecular Signature of Early Follicular Growth. PLoS One 2015; 10:e0141482. [PMID: 26540452 PMCID: PMC4634757 DOI: 10.1371/journal.pone.0141482] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 10/08/2015] [Indexed: 11/19/2022] Open
Abstract
Background The successful achievement of early ovarian folliculogenesis is important for fertility and reproductive life span. This complex biological process requires the appropriate expression of numerous genes at each developmental stage, in each follicular compartment. Relatively little is known at present about the molecular mechanisms that drive this process, and most gene expression studies have been performed in rodents and without considering the different follicular compartments. Results We used RNA-seq technology to explore the sheep transcriptome during early ovarian follicular development in the two main compartments: oocytes and granulosa cells. We documented the differential expression of 3,015 genes during this phase and described the gene expression dynamic specific to these compartments. We showed that important steps occurred during primary/secondary transition in sheep. We also described the in vivo molecular course of a number of pathways. In oocytes, these pathways documented the chronology of the acquisition of meiotic competence, migration and cellular organization, while in granulosa cells they concerned adhesion, the formation of cytoplasmic projections and steroid synthesis. This study proposes the involvement in this process of several members of the integrin and BMP families. The expression of genes such as Kruppel-like factor 9 (KLF9) and BMP binding endothelial regulator (BMPER) was highlighted for the first time during early follicular development, and their proteins were also predicted to be involved in gene regulation. Finally, we selected a data set of 24 biomarkers that enabled the discrimination of early follicular stages and thus offer a molecular signature of early follicular growth. This set of biomarkers includes known genes such as SPO11 meiotic protein covalently bound to DSB (SPO11), bone morphogenetic protein 15 (BMP15) and WEE1 homolog 2 (S. pombe)(WEE2) which play critical roles in follicular development but other biomarkers are also likely to play significant roles in this process. Conclusions To our knowledge, this is the first in vivo spatio-temporal exploration of transcriptomes derived from early follicles in sheep.
Collapse
Affiliation(s)
- Agnes Bonnet
- INRA, UMR 1388 GenPhySE (Génétique, Physiologie et Systèmes d’Elevage), F-31326 Castanet-Tolosan, France
- Université de Toulouse, INP, ENSAT, GenPhySE (Génétique, Physiologie et Systèmes d’Elevage), F-31326 Castanet-Tolosan, France
- Université de Toulouse, INP, ENVT, GenPhySE (Génétique, Physiologie et Systèmes d’Elevage), F-31076 Toulouse, France
- * E-mail:
| | - Bertrand Servin
- INRA, UMR 1388 GenPhySE (Génétique, Physiologie et Systèmes d’Elevage), F-31326 Castanet-Tolosan, France
- Université de Toulouse, INP, ENSAT, GenPhySE (Génétique, Physiologie et Systèmes d’Elevage), F-31326 Castanet-Tolosan, France
- Université de Toulouse, INP, ENVT, GenPhySE (Génétique, Physiologie et Systèmes d’Elevage), F-31076 Toulouse, France
| | - Philippe Mulsant
- INRA, UMR 1388 GenPhySE (Génétique, Physiologie et Systèmes d’Elevage), F-31326 Castanet-Tolosan, France
- Université de Toulouse, INP, ENSAT, GenPhySE (Génétique, Physiologie et Systèmes d’Elevage), F-31326 Castanet-Tolosan, France
- Université de Toulouse, INP, ENVT, GenPhySE (Génétique, Physiologie et Systèmes d’Elevage), F-31076 Toulouse, France
| | - Beatrice Mandon-Pepin
- INRA, UMR1198 Biologie du Développement et de la Reproduction, F-78350 Jouy-en-Josas, France
| |
Collapse
|
40
|
Escobar ML, Echeverría OM, García G, Ortíz R, Vázquez-Nin GH. Immunohistochemical and ultrastructural study of the lamellae of oocytes in atretic follicles in relation to different processes of cell death. Eur J Histochem 2015; 59:2535. [PMID: 26428888 PMCID: PMC4598600 DOI: 10.4081/ejh.2015.2535] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 06/24/2015] [Indexed: 11/23/2022] Open
Abstract
Atresia is the process through which non-selectable oocytes are eliminated; it involves apoptosis and/or autophagy. This study used immunohistochemical and ultrastructural techniques to characterize the lamellae present in the cytoplasm of oocytes in follicles in the process of atresia in prepubertal and adult Wistar rats. The results indicate that the lamellae are positive to tubulin and myosin immunodetection under light and electron microscopy. Labeling is greater with anti-tubulin and lesser with anti-myosin. Our observations indicate that lamellae are present in oocytes at the initial antral stage in prepubertal rats; that is, from day 14 post-birth to adult age. We were able to determine that the increase in altered lamellae principally occurs in the apoptotic cells rather than in the autophagic cells.
Collapse
|
41
|
Omari S, Waters M, Naranian T, Kim K, Perumalsamy AL, Chi M, Greenblatt E, Moley KH, Opferman JT, Jurisicova A. Mcl-1 is a key regulator of the ovarian reserve. Cell Death Dis 2015; 6:e1755. [PMID: 25950485 PMCID: PMC4669721 DOI: 10.1038/cddis.2015.95] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 02/07/2015] [Accepted: 02/18/2015] [Indexed: 02/05/2023]
Abstract
A majority of ovarian follicles are lost to natural death, but the disruption of factors involved in maintenance of the oocyte pool results in a further untimely follicular depletion known as premature ovarian failure. The anti-apoptotic B-cell lymphoma 2 (Bcl-2) family member myeloid cell leukemia-1 (MCL-1) has a pro-survival role in various cell types; however, its contribution to oocyte survival is unconfirmed. We present a phenotypic characterization of oocytes deficient in Mcl-1, and establish its role in maintenance of the primordial follicle (PMF) pool, growing oocyte survival and oocyte quality. Mcl-1 depletion resulted in the premature exhaustion of the ovarian reserve, characterized by early PMF loss because of activation of apoptosis. The increasingly diminished surviving cohort of growing oocytes displayed elevated markers of autophagy and mitochondrial dysfunction. Mcl-1-deficient ovulated oocytes demonstrated an increased susceptibility to cellular fragmentation with activation of the apoptotic cascade. Concomitant deletion of the pro-apoptotic Bcl-2 member Bcl-2-associated X protein (Bax) rescued the PMF phenotype and ovulated oocyte death, but did not prevent the mitochondrial dysfunction associated with Mcl-1 deficiency and could not rescue long-term breeding performance. We thus recognize MCL-1 as the essential survival factor required for conservation of the postnatal PMF pool, growing follicle survival and effective oocyte mitochondrial function.
Collapse
Affiliation(s)
- S Omari
- 1] Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, 25 Orde Street, Toronto, Ontario M5T 3H7, Canada [2] Department of Physiology, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - M Waters
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, 25 Orde Street, Toronto, Ontario M5T 3H7, Canada
| | - T Naranian
- 1] Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, 25 Orde Street, Toronto, Ontario M5T 3H7, Canada [2] Department of Physiology, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - K Kim
- 1] Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, 25 Orde Street, Toronto, Ontario M5T 3H7, Canada [2] Department of Physiology, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - A L Perumalsamy
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, 25 Orde Street, Toronto, Ontario M5T 3H7, Canada
| | - M Chi
- Department of Obstetrics and Gynecology, Washington University in St. Louis, 660S Euclid Avenue, St. Louis, MO 63110, USA
| | - E Greenblatt
- 1] Centre for Fertility and Reproductive Health, Mount Sinai Hospital, 250 Dundas Street, Toronto, Ontario M5T 2Z5, Canada [2] Department of Obstetrics and Gynecology, University of Toronto, 92 College Street, Toronto, Ontario M5G 1L4, Canada
| | - K H Moley
- Department of Obstetrics and Gynecology, Washington University in St. Louis, 660S Euclid Avenue, St. Louis, MO 63110, USA
| | - J T Opferman
- Department of Biochemistry, St. Jude Children's Research Hospital, MS 340, Room D4063D, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - A Jurisicova
- 1] Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, 25 Orde Street, Toronto, Ontario M5T 3H7, Canada [2] Department of Physiology, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada [3] Department of Obstetrics and Gynecology, University of Toronto, 92 College Street, Toronto, Ontario M5G 1L4, Canada
| |
Collapse
|
42
|
Tumor necrosis factor alpha inhibits ovulation and induces granulosa cell death in rat ovaries. Reprod Med Biol 2014; 14:107-115. [PMID: 26161038 PMCID: PMC4490172 DOI: 10.1007/s12522-014-0201-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 11/26/2014] [Indexed: 12/05/2022] Open
Abstract
Purpose We evaluated the role of tumor necrosis factor alpha (TNFα) in rat ovulation and granulosa cell death of ovarian follicles during the periovulatory stage. Methods Immature rats primed with pregnant mare serum gonadotropin were injected intraperitoneally with human chorionic gonadotropin (hCG), and TNFα was injected into the bursa 48 h later. The total number of released oocytes was counted. Apoptosis was measured with terminal deoxynucleotidyl transferase‐mediated dUTP nick end labeling (TUNEL) and the expression of cleaved caspase 3 and Bax/Bcl‐2. Autophagy was assessed by the expression of light chain protein 3 (LC3) and autophagosomes under transmission electron microscopy. Results TNFα significantly decreased the number of released oocytes, and many unruptured follicles were observed. TUNEL analysis revealed a larger number of apoptotic cells, and the cleaved caspase 3 and Bax/Bcl‐2 increased more than that of the control 12 h after hCG administration. Furthermore, the expression of LC3 wwas significantly higher than that of the control, and autophagosomes were observed in the cytoplasm. Conclusions Our data indicated that TNFα is an important mediator of ovulation in terms of decreasing the number of released oocytes and inducing granulosa cell death of unruptured follicles via apoptosis and autophagy for remodeling ovarian tissues.
Collapse
|
43
|
McGinnis LK, Pelech S, Kinsey WH. Post-ovulatory aging of oocytes disrupts kinase signaling pathways and lysosome biogenesis. Mol Reprod Dev 2014; 81:928-45. [PMID: 25242074 DOI: 10.1002/mrd.22413] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 08/07/2014] [Indexed: 12/21/2022]
Abstract
Post-ovulatory aging of oocytes results in the progressive loss of fertilization and developmental competence. This degradation of oocyte quality has been the object of numerous investigations, primarily focused on individual signaling pathways which provide limited insight into the status of global signaling events. The purpose of the present investigation was to comprehensively assess broad patterns of signaling pathway activity during in vitro aging as an initial step in defining control points that can be targeted to prevent the reduction in oocyte quality during prolonged culture. An antibody microarray-based phospho-proteome analysis performed on oocytes before and after eight hours of culture revealed significant changes in the abundance or activation state of 43 proteins that function in a wide variety of protein kinase-mediated signaling pathways. Several of the most significantly affected kinases were studied by Western blot and confocal immunofluorescence to corroborate the array results. Prolonged culture resulted in global changes in the abundance and activity of protein kinases that regulate the response to calcium, stress, and cell-cycle control. Examination of intracellular structures revealed a previously unrecognized increase in the abundance of large autophogagic lysosomes, which correlates with changes in protein kinase pathways. These results provide insight into the stresses experienced by oocytes during culture and the diversity of responses that results from them. The observed increase in autophagy-related activity, together with the disruptions in calcium signaling, cell-cycle, and stress-response pathways, have the potential to negatively impact oocyte quality by interfering with the normal sequence of biochemical changes that constitute egg activation following fertilization.
Collapse
Affiliation(s)
- Lynda K McGinnis
- Department Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas; Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas
| | | | | |
Collapse
|
44
|
Padua MB, Fox SC, Jiang T, Morse DA, Tevosian SG. Simultaneous gene deletion of gata4 and gata6 leads to early disruption of follicular development and germ cell loss in the murine ovary. Biol Reprod 2014; 91:24. [PMID: 24899573 DOI: 10.1095/biolreprod.113.117002] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Granulosa cell formation and subsequent follicular assembly are important for ovarian development and function. Two members of the GATA family of transcription factors, GATA4 and GATA6, are expressed in ovarian somatic cells early in development, and their importance in adult ovarian function has been recently highlighted. In this study, we demonstrated that the embryonic loss of Gata4 and Gata6 expression within the ovary results in a strong down-regulation of genes involved in the ovarian developmental pathway (Fst and Irx3) as well as diminished expression of the pregranulosa and granulosa cell markers SPRR2 and FOXL2, respectively. Postnatal ovaries deficient in both Gata genes show impaired somatic cell proliferation and arrested follicular development at the primordial stage, where oocytes are either enclosed by one layer of squamous granulosa cells or remain in germ cell nests/clusters. Furthermore, germ cell nests and primordial follicles are predominantly localized to the central region of the Sf1Cre; Gata4(flox/flox) Gata6(flox/flox) ovaries, where the boundary between the medulla and cortex is almost nonexistent. Lastly, most of the oocytes are lost early in development in conditional double mutant ovaries, which confirms the importance of normally differentiated granulosa cells as supporting cells for oocyte survival. Thus, both GATA4 and GATA6 proteins are fundamental regulators of granulosa cell differentiation and proliferation, and consequently of proper follicular assembly during normal ovarian development and function.
Collapse
Affiliation(s)
- Maria B Padua
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| | - Shawna C Fox
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| | - Tianyu Jiang
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| | - Deborah A Morse
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| | - Sergei G Tevosian
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| |
Collapse
|
45
|
Liew SH, Vaithiyanathan K, Cook M, Bouillet P, Scott CL, Kerr JB, Strasser A, Findlay JK, Hutt KJ. Loss of the proapoptotic BH3-only protein BCL-2 modifying factor prolongs the fertile life span in female mice. Biol Reprod 2014; 90:77. [PMID: 24571986 DOI: 10.1095/biolreprod.113.116947] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The duration of the female fertile life span is influenced by the number of oocytes stored in the ovary as primordial follicles. Cell death, both during ovarian development in the embryo and in the postnatal ovary, plays a critical role in determining how many primordial follicles are established and maintained within the ovary. However, the roles of individual apoptotic regulators in mediating cell death within the ovary have not yet been characterized. In this study, gene targeted mice were used to investigate the role of BCL-2-modifying factor (BMF), a proapoptotic protein belonging to the BH3-only subgroup of the BCL-2 family, in determining the number of primordial follicles maintained in the adult ovary and the length of the fertile life span. Stereological analysis of ovaries showed that Bmf(-/-) mice had significantly more primordial follicles than wild-type (WT) control animals at Postnatal Days 100, 200, 300, and 400 but not at Day 20. No differences were observed between WT and Bmf(-/-) mice in the number of ova shed following ovulatory stimulation with exogenous gonadotropins. Bmf(-/-) females were fertile and produced the same number pups/litter as WT females, but Bmf(-/-) females produced litters more frequently and consequently more offspring than WT females over a 6-mo period. Furthermore, the fertile life span of Bmf(-/-) females was significantly extended compared to WT females. Our findings support an important role for BMF in determining the number of primordial follicles maintained in the ovary throughout adult reproductive life and thus indicate that the length of female fertility may be extended by increasing the number of primordial follicles maintained within the ovary through inhibition of BMF.
Collapse
Affiliation(s)
- Seng H Liew
- MIMR-PHI Institute of Medical Research, Monash University, Clayton, Victoria, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Holt JE, Pye V, Boon E, Stewart JL, García-Higuera I, Moreno S, Rodríguez R, Jones KT, McLaughlin EA. The APC/C activator FZR1 is essential for meiotic prophase I in mice. Development 2014; 141:1354-65. [PMID: 24553289 DOI: 10.1242/dev.104828] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Fizzy-related 1 (FZR1) is an activator of the Anaphase promoting complex/cyclosome (APC/C) and an important regulator of the mitotic cell division cycle. Using a germ-cell-specific conditional knockout model we examined its role in entry into meiosis and early meiotic events in both sexes. Loss of APC/C(FZR1) activity in the male germline led to both a mitotic and a meiotic testicular defect resulting in infertility due to the absence of mature spermatozoa. Spermatogonia in the prepubertal testes of such mice had abnormal proliferation and delayed entry into meiosis. Although early recombination events were initiated, male germ cells failed to progress beyond zygotene and underwent apoptosis. Loss of APC/C(FZR1) activity was associated with raised cyclin B1 levels, suggesting that CDK1 may trigger apoptosis. By contrast, female FZR1Δ mice were subfertile, with premature onset of ovarian failure by 5 months of age. Germ cell loss occurred embryonically in the ovary, around the time of the zygotene-pachytene transition, similar to that observed in males. In addition, the transition of primordial follicles into the growing follicle pool in the neonatal ovary was abnormal, such that the primordial follicles were prematurely depleted. We conclude that APC/C(FZR1) is an essential regulator of spermatogonial proliferation and early meiotic prophase I in both male and female germ cells and is therefore important in establishing the reproductive health of adult male and female mammals.
Collapse
Affiliation(s)
- Janet E Holt
- School of Biomedical Sciences & Pharmacy, University of Newcastle, Callaghan, NSW 2308, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Chalmey C, Giton F, Chalmel F, Fiet J, Jégou B, Mazaud-Guittot S. Systemic compensatory response to neonatal estradiol exposure does not prevent depletion of the oocyte pool in the rat. PLoS One 2013; 8:e82175. [PMID: 24358151 PMCID: PMC3864944 DOI: 10.1371/journal.pone.0082175] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Accepted: 10/21/2013] [Indexed: 11/19/2022] Open
Abstract
The formation of ovarian follicles is a finely tuned process that takes place within a narrow time-window in rodents. Multiple factors and pathways have been proposed to contribute to the mechanisms triggering this process but the role of endocrine factors, especially estrogens, remains elusive. It is currently hypothesized that removal from the maternal hormonal environment permits follicle formation at birth. However, experimentally-induced maintenance of high 17β-estradiol (E2) levels leads to subtle, distinct, immediate effects on follicle formation and oocyte survival depending on the species and dose. In this study, we examined the immediate effects of neonatal E2 exposure from post-natal day (PND) 0 to PND2 on the whole organism and on ovarian follicle formation in rats. Measurements of plasma E2, estrone and their sulfate conjugates after E2 exposure showed that neonatal female rats rapidly acquire the capability to metabolize and clear excessive E2 levels. Concomitant modifications to the mRNA content of genes encoding selected E2 metabolism enzymes in the liver and the ovary in response to E2 exposure indicate that E2 may modify the neonatal maturation of these organs. In the liver, E2 treatment was associated with lower acquisition of the capability to metabolize E2. In the ovary, E2 depleted the oocyte pool in a dose dependent manner by PND3. In 10 µg/day E2-treated ovaries, apoptotic oocytes were observed in newly formed follicles in addition to areas of ovarian cord remodeling. At PND6, follicles without any visible oocyte were present and multi-oocyte follicles were not observed. Our study reveals a major species-difference. Indeed, neonatal exposure to E2 depletes the oocyte pool in the rat ovary, whereas in the mouse it is well known to increase oocyte survival.
Collapse
Affiliation(s)
- Clémentine Chalmey
- Institut National de la Santé et de la Recherche Médicale, Unité 1085 Institut de Recherche en Santé Environnement et Travail, Institut Fédératif de Recherche 140, Université de Rennes 1, Rennes, France
| | - Franck Giton
- AP-HP, Hôpital H. Mondor - A. Chenevier, service de Biochimie et de Génétique, Créteil, France
- Institut National de la Santé et de la Recherche Médicale, U955 Équipe 07, Créteil, France
| | - Frédéric Chalmel
- Institut National de la Santé et de la Recherche Médicale, Unité 1085 Institut de Recherche en Santé Environnement et Travail, Institut Fédératif de Recherche 140, Université de Rennes 1, Rennes, France
| | - Jean Fiet
- Institut National de la Santé et de la Recherche Médicale, U955 Équipe 07, Créteil, France
| | - Bernard Jégou
- Institut National de la Santé et de la Recherche Médicale, Unité 1085 Institut de Recherche en Santé Environnement et Travail, Institut Fédératif de Recherche 140, Université de Rennes 1, Rennes, France
- Ecole des Hautes Études en Santé Publique, Rennes, France
| | - Séverine Mazaud-Guittot
- Institut National de la Santé et de la Recherche Médicale, Unité 1085 Institut de Recherche en Santé Environnement et Travail, Institut Fédératif de Recherche 140, Université de Rennes 1, Rennes, France
- * E-mail:
| |
Collapse
|
48
|
Liu L, Jin H, Ou J, Jiang J, Pi J, Ke C, Yang F, Qiao D, Cai H, Cai J. Cinobufacini-induced HeLa cell apoptosis enhanced by curcumin. ACTA ACUST UNITED AC 2013. [DOI: 10.1007/s11434-013-5739-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
49
|
Escobar Sánchez ML, Echeverría Martínez OM, Vázquez-Nin GH. Immunohistochemical and ultrastructural visualization of different routes of oocyte elimination in adult rats. Eur J Histochem 2012; 56:e17. [PMID: 22688298 PMCID: PMC3428966 DOI: 10.4081/ejh.2012.17] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Revised: 02/04/2012] [Accepted: 02/08/2012] [Indexed: 01/07/2023] Open
Abstract
Cell death is a process for maintaining homeostasis in tissues and organs. In the ovary, apoptotic cell death has been implicated in follicular atresia; in the elimination of the follicles that are not ovulated during adult life. Recent studies indicate that apoptosis and autophagy are two programmed processes of cell death. Apoptosis is performed by proteases called caspases and leads to such morphological traits as DNA fragmentation. Autophagy, in turn, is characterized by the exacerbated formation of autophagosomes; a process in which the amount of the LC3 and Lamp 1 proteins increases. In this study, oocytes from all stages of the estrous cycle of Wistar rats were analyzed. The apoptosis process was identified by immunodetecting active Caspase-3 and locating DNA fragmentation using the TUNEL technique. Autophagy was evaluated through immunodetection of the LC3 and Lamp 1 proteins, and by ultrastructural localization of autophagic vesicle formation. All techniques were conducted using the same oocytes. Results show that all phases of the estrous cycle contain dying oocytes that test positive simultaneously for apoptosis and autophagy markers. The highest level of apoptosis was found during estrus; while the proestrous stage had the highest level of autophagy. The diestrous and metestrous phases were characterized by a high frequency of the presence of markers of apoptosis and autophagy in the same oocyte. Our results demonstrate that during oocyte elimination in adult rats the proteins involved in both processes, apoptosis and autophagy, are present in the same cell at the same time.
Collapse
Affiliation(s)
- M L Escobar Sánchez
- Departamento de Biología Celular, Facultad de Ciencias, Universidad Nacional Autónoma de México (UNAM), Coyoacán, México
| | | | | |
Collapse
|
50
|
Thomson TC, Schneemann A, Johnson J. Oocyte destruction is activated during viral infection. Genesis 2012; 50:453-65. [PMID: 22173880 DOI: 10.1002/dvg.22004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2010] [Revised: 11/30/2011] [Accepted: 12/11/2011] [Indexed: 11/10/2022]
Abstract
Viral infection has been associated with a starvation-like state in Drosophila melanogaster. Because starvation and inhibiting TOR kinase activity in vivo result in blocked oocyte production, we hypothesized that viral infection would also result in compromised oogenesis. Wild-type flies were injected with flock house virus (FHV) and survival and embryo production were monitored. Infected flies had a dose-responsive loss of fecundity that corresponded to a global reduction in Akt/TOR signaling. Highly penetrant egg chamber destruction mid-way through oogenesis was noted and FHV coat protein was detected within developing egg chambers. As seen with in vivo TOR inhibition, oogenesis was partially rescued in loss of function discs large and merlin mutants. As expected, mutants in genes known to be involved in virus internalization and trafficking [Clathrin heavy chain (chc) and synaptotagmin] survive longer during infection. However, oogenesis was rescued only in chc mutants. This suggests that viral response mechanisms that control fly survival and egg chamber survival are separable. The genetic and signaling requirements for oocyte destruction delineated here represent a novel host-virus interaction with implications for the control of both fly and virus populations.
Collapse
Affiliation(s)
- Travis C Thomson
- Department of Obstetrics, Gynecology, and Reproductive Sciences/Division of Reproductive Endocrinology and Infertility, Yale School of Medicine, New Haven, Connecticut, USA
| | | | | |
Collapse
|