1
|
Zhu Q, Du J, Li Y, Qin X, He R, Ma H, Liang X. Downregulation of glucose-energy metabolism via AMPK signaling pathway in granulosa cells of diminished ovarian reserve patients. Gene 2025; 933:148979. [PMID: 39366473 DOI: 10.1016/j.gene.2024.148979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 08/15/2024] [Accepted: 10/01/2024] [Indexed: 10/06/2024]
Abstract
Glucose metabolism plays a crucial role in the function of granulosa cells (GCs) and the development of follicles. In cases of diminished ovarian reserve (DOR), alterations in these processes can impact female fertility. This study aims to investigate changes in glucose-energy metabolism in GCs of young DOR patients aged 20 to 35 years and their correlation with the onset and progression of DOR. 72 DOR cases and 75 women with normal ovarian reserve (NOR) as controls were included based on the POSEIDON and Bologna criteria. Samples of GCs and follicular fluid (FF) were collected for a comprehensive analysis involving transcriptomics, metabolomics, RT-qPCR, JC-1 staining, and flow cytometry. The study identified differentially expressed genes and metabolites in GCs of DOR and NOR groups, revealing 7 common pathways related to glucose-energy metabolism, along with 11 downregulated genes and 14 metabolites. Key substances in the glucose-energy metabolism pathway, such as succinate, lactate, NADP, ATP, and ADP, showed decreased levels, with the DOR group exhibiting a reduced ADP/ATP ratio. Downregulation of genes involved in glycolysis (HK, PGK, LDH1), the TCA cycle (CS), and gluconeogenesis (PCK) was observed, along with reduced glucose content and expression of glucose transporter genes (GLUT1 and GLUT3) in DOR GCs. Additionally, decreased AMPK pathway activity and impaired mitochondrial function in DOR suggest a connection between mitochondrial dysfunction and disrupted energy metabolism. Above all, the decline in glucose-energy metabolism in DOR is closely associated with its onset and progression. Reduced glucose uptake and impaired mitochondrial function in DOR GCs lead to internal energy imbalances, hindering the AMPK signaling pathway, limiting energy production and supply, and ultimately impacting follicle development and maturation.
Collapse
Affiliation(s)
- Qinying Zhu
- Department of Obstetrics and Gynecology, Chinese Academy of Medical Sciences, Peking Union Medical College, National Clinical Research Center for Obstetric and Gynecologic Diseases, Peking Union Medical College Hospital, Beijing, China; The First Clinical Medical College of Lanzhou University, Lanzhou, China.
| | - Junhong Du
- The First Clinical Medical College of Lanzhou University, Lanzhou, China.
| | - Yi Li
- The First Clinical Medical College of Lanzhou University, Lanzhou, China.
| | - Xue Qin
- The First Clinical Medical College of Lanzhou University, Lanzhou, China.
| | - Ruifen He
- The First Clinical Medical College of Lanzhou University, Lanzhou, China.
| | - Hao Ma
- The First Clinical Medical College of Lanzhou University, Lanzhou, China.
| | - Xiaolei Liang
- Department of Obstetrics and Gynecology, the First Hospital of Lanzhou University, Gansu Provincial Clinical Research Center for Gynecological Oncology, Lanzhou, China.
| |
Collapse
|
2
|
Przygrodzka E, Bhinderwala F, Powers R, McFee RM, Cupp AS, Wood JR, Davis JS. Metabolic control of luteinizing hormone-responsive ovarian steroidogenesis. J Biol Chem 2024; 301:108042. [PMID: 39615688 DOI: 10.1016/j.jbc.2024.108042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 10/22/2024] [Accepted: 11/13/2024] [Indexed: 12/28/2024] Open
Abstract
The pituitary gonadotropin luteinizing hormone (LH) is the primary stimulus for ovulation, luteal formation, and progesterone synthesis, regardless of species. Despite increased awareness of intracellular signaling events initiating the massive production of progesterone during the reproductive cycle and pregnancy, critical gaps exist in our knowledge of the metabolic and lipidomic pathways required for initiating and maintaining luteal progesterone synthesis. Using untargeted metabolomics and metabolic flux analysis in primary steroidogenic luteal cells, evidence is provided for rapid LHCGR-stimulation of metabolic pathways leading to increased glycolysis and oxygen consumption. Treatment with LH stimulated posttranslational modifications of enzymes involved in de novo lipogenesis. Mechanistic studies implicated a crucial role for de novo fatty acid synthesis and fatty acid oxidation in energy homeostasis, LHCGR/PKA signaling, and, consequently, progesterone production. These findings reveal novel hormone-sensitive metabolic pathways essential for maintaining LHCGR/PKA signaling and steroidogenesis. Understanding hormonal control of metabolic pathways in steroidogenic cells may help elucidate approaches for improving ovarian function and successful reproduction or identifying metabolic targets for developing nonhormonal contraceptives.
Collapse
Affiliation(s)
- Emilia Przygrodzka
- Olson Center for Women's Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, Nebraska, USA.
| | - Fatema Bhinderwala
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Robert Powers
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, Nebraska, USA; Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Renee M McFee
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Andrea S Cupp
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Jennifer R Wood
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - John S Davis
- Olson Center for Women's Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, Nebraska, USA; VA Nebraska-Western Iowa Health Care System, Omaha, Nebraska, USA.
| |
Collapse
|
3
|
Zhang X, Ge J, Wang Y, Chen M, Guo X, Zhu S, Wang H, Wang Q. Integrative Omics Reveals the Metabolic Patterns During Oocyte Growth. Mol Cell Proteomics 2024; 23:100862. [PMID: 39414232 PMCID: PMC11585809 DOI: 10.1016/j.mcpro.2024.100862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/01/2024] [Accepted: 10/13/2024] [Indexed: 10/18/2024] Open
Abstract
Well-controlled metabolism is associated with high-quality oocytes and optimal development of a healthy embryo. However, the metabolic framework that controls mammalian oocyte growth remains unknown. In the present study, we comprehensively depict the temporal metabolic dynamics of mouse oocytes during in vivo growth through the integrated analysis of metabolomics and proteomics. Many novel metabolic features are discovered during this process. Of note, glycolysis is enhanced, and oxidative phosphorylation capacity is reduced in the growing oocytes, presenting a Warburg-like metabolic program. For nucleotide biosynthesis, the salvage pathway is markedly activated during oocyte growth, whereas the de novo pathway is evidently suppressed. Fatty acid synthesis and channeling into phosphoinositides are specifically elevated in oocytes accompanying primordial follicle activation; nevertheless, fatty acid oxidation is reduced in these oocytes simultaneously. Our data establish the metabolic landscape during in vivo oocyte growth and serve as a broad resource for probing mammalian oocyte metabolism.
Collapse
Affiliation(s)
- Xiang Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, China
| | - Juan Ge
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, China
| | - Yue Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, China
| | - Minjian Chen
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xuejiang Guo
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, China
| | - Shuai Zhu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, China.
| | - Hui Wang
- Department of Histology and Embryology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China.
| | - Qiang Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
4
|
Nahar A, Becker J, Pasquariello R, Herrick J, Rogers H, Zhang M, Schoolcraft W, Krisher RL, Yuan Y. FGF2, LIF, and IGF-1 supplementation improves mouse oocyte in vitro maturation via increased glucose metabolism†. Biol Reprod 2024; 110:672-683. [PMID: 38263524 DOI: 10.1093/biolre/ioae014] [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: 08/31/2023] [Revised: 12/22/2023] [Accepted: 01/15/2024] [Indexed: 01/25/2024] Open
Abstract
Chemically defined oocyte maturation media supplemented with FGF2, LIF, and IGF-1 (FLI medium) enabled significantly improved oocyte quality in multiple farm animals, yet the molecular mechanisms behind such benefits were poorly defined. Here, we first demonstrated that FLI medium enhanced mouse oocyte quality assessed by blastocyst formation after in vitro fertilization and implantation and fetal development after embryo transfer. We then analyzed the glucose concentrations in the spent media; reactive oxygen species concentrations; mitochondrial membrane potential; spindle morphology in oocytes; and the abundance of transcripts of endothelial growth factor-like factors, cumulus expansion factors, and glucose metabolism-related genes in cumulus cells. We found that FLI medium enabled increased glucose metabolism through glycolysis, pentose phosphate pathway, and hexosamine biosynthetic pathway, as well as more active endothelial growth factor-like factor expressions in cumulus cells, resulting in improved cumulus cell expansion, decreased spindle abnormality, and overall improvement in oocyte quality. In addition, the activities of MAPK1/3, PI3K/AKT, JAK/STAT3, and mTOR signaling pathways in cumulus cells were assessed by the phosphorylation of MAPK1/3, AKT, STAT3, and mTOR downstream target RPS6KB1. We demonstrated that FLI medium promoted activations of all these signaling pathways at multiple different time points during in vitro maturation.
Collapse
Affiliation(s)
- Asrafun Nahar
- Colorado Center for Reproductive Medicine, Lone Tree, CO, USA
| | - John Becker
- Colorado Center for Reproductive Medicine, Lone Tree, CO, USA
| | - Rolando Pasquariello
- Colorado Center for Reproductive Medicine, Lone Tree, CO, USA
- Department of Agricultural and Environmental Sciences, University of Milan, Milan, Italy
| | - Jason Herrick
- Colorado Center for Reproductive Medicine, Lone Tree, CO, USA
- Omaha's Henry Doorly Zoo and Aquarium, Omaha, NE, USA
| | - Heather Rogers
- Colorado Center for Reproductive Medicine, Lone Tree, CO, USA
| | - Mingxiang Zhang
- Colorado Center for Reproductive Medicine, Lone Tree, CO, USA
| | | | - Rebecca L Krisher
- Colorado Center for Reproductive Medicine, Lone Tree, CO, USA
- Genus Plc, DeForest, WI, USA
| | - Ye Yuan
- Colorado Center for Reproductive Medicine, Lone Tree, CO, USA
| |
Collapse
|
5
|
Xu Y, Zhou Z, Zhang G, Yang Z, Shi Y, Jiang Z, Liu Y, Chen H, Huang H, Zhang Y, Pan J. Metabolome implies increased fatty acid utilization and histone methylation in the follicles from hyperandrogenic PCOS women. J Nutr Biochem 2024; 125:109548. [PMID: 38104867 DOI: 10.1016/j.jnutbio.2023.109548] [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: 07/10/2023] [Revised: 11/21/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023]
Abstract
Well-balanced metabolism is essential for the high-quality of oocytes, and metabolic fluctuations of follicular microenvironment potentially encourage functional changes in follicle cells, ultimately impacting the developmental potential of oocytes. Here, the global metabolomic profiles of follicular fluid from PCOS women with ovarian hyperandrogenism and nonhyperandrogenism were depicted by untargeted metabolome and transcriptome. In parallel, functional methods were employed to evaluate the possible impact of dysregulated metabolites on oocyte and embryo development. Our findings demonstrated that PCOS women exhibited distinct metabolic features in follicles, such as the increase in fatty acid utilization and the downregulation in amino acid metabolism. And intrafollicular androgen levels were positively correlated with contents of multiple fatty acids, suggesting androgen as one of the contributing factors to the metabolic abnormalities in PCOS follicles. Moreover, we further demonstrated that elevated levels of α-linolenic acid and H3K27me3 could hinder oocyte maturation, fertilization, and early embryo development. Hopefully, our data serve as a broad resource on the metabolic abnormalities of PCOS follicles, and advances in the relevant knowledge will allow the identification of biomarkers that predict the progression of PCOS and its poor pregnancy outcomes.
Collapse
Affiliation(s)
- Yue Xu
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China; Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China
| | - Zhiyang Zhou
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China; Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China
| | - Gaochen Zhang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China; Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China
| | - Zuwei Yang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China; Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China; The International Peace Maternal and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yan Shi
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China; Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China
| | - Zhaoying Jiang
- Key Laboratory of Reproductive Genetics (Ministry of Education), Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ye Liu
- The International Peace Maternal and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Huixi Chen
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China; Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China; The International Peace Maternal and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hefeng Huang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China; Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China; The International Peace Maternal and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Key Laboratory of Reproductive Genetics (Ministry of Education), Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Yu Zhang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China; Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China.
| | - Jiexue Pan
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China; Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China; The International Peace Maternal and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Key Laboratory of Reproductive Genetics (Ministry of Education), Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
6
|
Przygrodzka E, Binderwala F, Powers R, McFee RM, Cupp AS, Wood JR, Davis JS. Central Role for Glycolysis and Fatty Acids in LH-responsive Progesterone Synthesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.14.580329. [PMID: 38405789 PMCID: PMC10888869 DOI: 10.1101/2024.02.14.580329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Progesterone production by the corpus luteum is fundamental for establishing and maintaining pregnancy. The pituitary gonadotropin luteinizing hormone (LH) is recognized as the primary stimulus for luteal formation and progesterone synthesis, regardless of species. Previous studies demonstrated an elevation in abundance of genes related to glucose and lipid metabolism during the follicular to luteal transition. However, the metabolic phenotype of these highly steroidogenic cells has not been studied. Herein, we determined acute metabolic changes induced by LH in primary luteal cells and defined pathways required for progesterone synthesis. Untargeted metabolomics analysis revealed that LH induces rapid changes in vital metabolic pathways, including glycolysis, tricarboxylic acid (TCA) cycle, pentose phosphate pathway, de novo lipogenesis, and hydrolysis of phospholipids. LH stimulated glucose uptake, enhanced glycolysis, and flux of [U- 13 C 6 ]-labeled glucose-derived carbons into metabolic branches associated with adenosine 5'-triphosphate (ATP) and NADH/NADPH production, synthesis of nucleotides, proteins, and lipids, glycosylation of proteins or lipids, and redox homeostasis. Selective use of small molecule inhibitors targeting the most significantly changed pathways, such as glycolysis, TCA cycle, and lipogenesis, uncovered cellular metabolic routes required for LH-stimulated steroidogenesis. Furthermore, LH via the protein kinase A (PKA) pathway triggered post- translational modification of acetyl-CoA carboxylase alpha (ACACA) and ATP citrate lyase (ACLY), enzymes involved in de novo synthesis of fatty acids. Inhibition of ACLY and fatty acid transport into mitochondria reduced LH-stimulated ATP, cAMP production, PKA activation, and progesterone synthesis. Taken together, these findings reveal novel hormone-sensitive metabolic pathways essential for maintaining LHCGR/PKA signaling and steroidogenesis in ovarian luteal cells. Significance The establishment and maintenance of pregnancy require a well-developed corpus luteum, an endocrine gland within the ovary that produces progesterone. Although there is increased awareness of intracellular signaling events initiating the massive production of progesterone during the reproductive cycle and pregnancy, there are critical gaps in our knowledge of the metabolic and lipidomic pathways required for initiating and maintaining luteal progesterone synthesis. Here, we describe rapid, hormonally triggered metabolic pathways, and define metabolic targets crucial for progesterone synthesis by ovarian steroidogenic cells. Understanding hormonal control of metabolic pathways may help elucidate approaches for improving ovarian function and successful reproduction or identifying metabolic targets for developing nonhormonal contraceptives.
Collapse
|
7
|
Dotania K, Tripathy M, Rai U. Ovarian nesfatin-1 in Hemidactylus flaviviridis: Reproductive phase-dependent expression, role and hormonal regulation. Comp Biochem Physiol A Mol Integr Physiol 2024; 288:111556. [PMID: 38016591 DOI: 10.1016/j.cbpa.2023.111556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 11/22/2023] [Accepted: 11/22/2023] [Indexed: 11/30/2023]
Abstract
Nesfatin-1 has recently emerged as a modulator of ovarian functions in mammals. Studies in non-mammalian vertebrates, though limited and majorly restricted to fishes, have evidenced a role of this peptide in the regulation of ovarian steroidogenesis and oocyte maturation. Interestingly, nesfatin-1 remains completely unexplored in reptiles. Therefore, the present study aimed to identify nesfatin-1 and elucidate its role and regulation in the ovary of Hemidactylus flaviviridis. Ovarian expression of nucb2/nesfatin-1 was highest during late recrudescence and breeding while it was lowest during regression. Follicular stage-dependent expression analysis showed significantly high expression of nucb2/nesfatin-1 in previtellogenic follicles. Further, in vitro treatment of recrudescent wall lizard ovaries with nesfatin-1 resulted in increased expression of anti-apoptotic gene, bcl-2, along with a concomitant decline in the pro-apoptotic gene, caspase-3. In addition, proliferation/differentiation markers like scf, c-kit, pcna, and bmp-15 were stimulated in ovaries incubated with the peptide. Ovarian steroidogenesis was also positively influenced by nesfatin-1 as treatment with the peptide resulted in heightened star expression as well as increased estradiol and progesterone production. Also, all concentrations of nesfatin-1 stimulated glucose uptake and metabolism in wall lizard ovary. Our observations provide the first evidence of ovarian functions of nesfatin-1 in a reptile. Further, ovarian nucb2/nesfatin-1 was differentially regulated by gonadotropin and sex steroids wherein its expression was stimulated by dihydrotestosterone (DHT) and 17β-estradiol (E2) but inhibited by follicle-stimulating hormone (FSH). In summary, this is the first report of the presence, reproductive stage-dependent expression, role, and regulation of ovarian nucb2/nesfatin-1 in H. flaviviridis.
Collapse
Affiliation(s)
| | - Mamta Tripathy
- Department of Zoology, University of Delhi, Delhi 110007, India.
| | - Umesh Rai
- University of Jammu, Jammu and Kashmir, 180006, India.
| |
Collapse
|
8
|
Elías-López AL, Vázquez-Mena O, Sferruzzi-Perri AN. Mitochondrial dysfunction in the offspring of obese mothers and it's transmission through damaged oocyte mitochondria: Integration of mechanisms. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166802. [PMID: 37414229 DOI: 10.1016/j.bbadis.2023.166802] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/23/2023] [Accepted: 06/29/2023] [Indexed: 07/08/2023]
Abstract
In vivo and in vitro studies demonstrate that mitochondria in the oocyte, are susceptible to damage by suboptimal pre/pregnancy conditions, such as obesity. These suboptimal conditions have been shown to induce mitochondrial dysfunction (MD) in multiple tissues of the offspring, suggesting that mitochondria of oocytes that pass from mother to offspring, can carry information that can programme mitochondrial and metabolic dysfunction of the next generation. They also suggest that transmission of MD could increase the risk of obesity and other metabolic diseases in the population inter- and trans-generationally. In this review, we examined whether MD observed in offspring tissues of high energetic demand, is the result of the transmission of damaged mitochondria from the oocytes of obese mothers to the offspring. The contribution of genome-independent mechanisms (namely mitophagy) in this transmission were also explored. Finally, potential interventions aimed at improving oocyte/embryo health were investigated, to see if they may provide an opportunity to halter the generational effects of MD.
Collapse
Affiliation(s)
- A L Elías-López
- Dirección de Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán", México.
| | | | - A N Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, UK.
| |
Collapse
|
9
|
Maucieri AM, Townson DH. Evaluating the impact of the hexosamine biosynthesis pathway and O-GlcNAcylation on glucose metabolism in bovine granulosa cells. Mol Cell Endocrinol 2023; 564:111863. [PMID: 36690170 DOI: 10.1016/j.mce.2023.111863] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 01/08/2023] [Accepted: 01/16/2023] [Indexed: 01/22/2023]
Abstract
Granulosa cells (GCs) of ovarian follicles prefer glucose as a metabolic substrate for growth and maturation. Disruption of glucose utilization via the hexosamine biosynthesis pathway (HBP) impairs O-linked N-acetylglucosaminylation (O-GlcNAcylation) and inhibits proliferation of bovine GCs of both small (3-5 mm) and large (>8.5 mm) antral follicles. Knowing that 2-5% of all glucose in cells is utilized via the HBP, the aim of this study was to characterize glucose metabolism in bovine GCs and determine the impact of the HBP and O-GlcNAcylation on metabolic activity. The GCs were initially cultured in serum-containing medium to confluency and then sub-cultured in serum-free medium in 96 well plates (n = 10 ovary pairs). The cells were exposed to vehicle and inhibitors of the HBP and O-GlcNAcylation for 24 h. Extracellular acidification rate (ECAR; an indicator of glycolysis) and oxygen consumption rate (OCR; an indicator of oxidative phosphorylation) of the GCs were measured using a Seahorse xFe96 Analyzer, including the implementation of glycolytic and mitochondrial stress tests. GCs from small antral follicles exhibited overall greater metabolic activity than GCs from large antral follicles as evidenced by increased ECAR and OCR. Inhibition of the HBP and O-GlcNAcylation had no effect on the metabolic activity of GCs from either type of follicle. The glycolytic stress test indicated that GCs from both types of follicles possessed additional glycolytic capacity; but again, inhibition of the HBP and O-GlcNAcylation did not affect this. Interestingly, inhibition of cellular respiration by 2-Deoxy-D-glucose impaired OCR only in GCs from small antral follicles, but exposure to the mitochondrial stress test had no effect. Conversely, in GCs from large antral follicles, oxidative metabolism was impaired by the mitochondrial stress test and was accompanied by a concomitant increase in glycolytic metabolism. Immunodetection of glycolytic enzymes revealed that phosphofructokinase expression is increased in GCs of small antral follicles compared to large follicles. Inhibition of O-GlcNAcylation impaired the expression of hexokinase only in GCs of small antral follicles. Inhibition of O-GlcNAcylation also impaired the expression of phosphofructokinase, pyruvate kinase and pyruvate dehydrogenase in GCs of both types of follicles, but had no effect on the expression of lactate dehydrogenase. The results indicate that GCs of small antral follicles possess greater aerobic glycolytic capacity than GCs from large antral follicles; but disruption of the HBP and O-GlcNAcylation has little to no impact on metabolic activity.
Collapse
Affiliation(s)
- Abigail M Maucieri
- Department of Animal and Veterinary Sciences, The University of Vermont, Burlington, VT, 05405, USA
| | - David H Townson
- Department of Animal and Veterinary Sciences, The University of Vermont, Burlington, VT, 05405, USA; Department of Fisheries, Animal and Veterinary Sciences, The University of Rhode Island, Kingston, RI, 02881, USA.
| |
Collapse
|
10
|
Zhang S, Mu L, Wang H, Xu X, Jia L, Niu S, Wang Y, Wang P, Li L, Chai J, Li Z, Zhang Y, Zhang H. Quantitative proteomic analysis uncovers protein-expression profiles during gonadotropin-dependent folliculogenesis in mice†. Biol Reprod 2023; 108:479-491. [PMID: 36477298 DOI: 10.1093/biolre/ioac217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 07/14/2022] [Accepted: 12/02/2022] [Indexed: 12/13/2022] Open
Abstract
Ovarian follicle is the basic functional unit of female reproduction, and is composed of oocyte and surrounding granulosa cells. In mammals, folliculogenesis strictly rely on gonadotropin regulations to determine the ovulation and the quality of eggs. However, the dynamic changes of protein-expressing profiles in follicles at different developmental stages remain largely unknown. By performing mass-spectrometry-based quantitative proteomic analysis of mouse follicles, we provide a proteomic database (~3000 proteins) that covers three key stages of gonadotropin-dependent folliculogenesis. By combining bioinformatics analysis with in situ expression validation, we showed that our proteomic data well reflected physiological changes during folliculogenesis, which provided potential to predict unknown regulators of folliculogenesis. Additionally, by using the oocyte structural protein zona pellucida protein 2 as the internal control, we showed the possibility of our database to predict the expression dynamics of oocyte-expressing proteins during folliculogenesis. Taken together, we provide a high-coverage proteomic database to study protein-expression dynamics during gonadotropin-dependent folliculogenesis in mammals.
Collapse
Affiliation(s)
- Shuo Zhang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Lu Mu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Haoran Wang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Xueqiang Xu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Longzhong Jia
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Shudong Niu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Yibo Wang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Peike Wang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Lingyu Li
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Junyi Chai
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Zhen Li
- State Key Laboratory of Plant Physiology and Biochemistry, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Yan Zhang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Hua Zhang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| |
Collapse
|
11
|
Sharma Y, Galvão AM. Maternal obesity and ovarian failure: is leptin the culprit? Anim Reprod 2023; 19:e20230007. [PMID: 36855701 PMCID: PMC9968511 DOI: 10.1590/1984-3143-ar2023-0007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 01/24/2023] [Indexed: 02/22/2023] Open
Abstract
At the time of its discovery and characterization in 1994, leptin was mostly considered a metabolic hormone able to regulate body weight and energy homeostasis. However, in recent years, a great deal of literature has revealed leptin's pleiotropic nature, through its involvement in numerous physiological contexts including the regulation of the female reproductive tract and ovarian function. Obesity has been largely associated with infertility, and leptin signalling is known to be dysregulated in the ovaries of obese females. Hence, the disruption of ovarian leptin signalling was shown to contribute to the pathophysiology of ovarian failure in obese females, affecting transcriptional programmes in the gamete and somatic cells. This review attempts to uncover the underlying mechanisms contributing to female infertility associated with obesity, as well as to shed light on the role of leptin in the metabolic dysregulation within the follicle, the effects on the oocyte epigenome, and the potential long-term consequence to embryo programming.
Collapse
Affiliation(s)
- Yashaswi Sharma
- Institute of Animal Reproduction and Food Research of PAS, Department of Reproductive Immunology and Pathology, Olsztyn, Poland
| | - António Miguel Galvão
- Institute of Animal Reproduction and Food Research of PAS, Department of Reproductive Immunology and Pathology, Olsztyn, Poland,Babraham Institute, Epigenetics Programme, Cambridge, United Kingdom UK,Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom UK,Corresponding author: ;
| |
Collapse
|
12
|
Ge ZJ, Gioia Klinger F, Taketo T. Editorial: Intra- and extra-environment and reproduction. Front Cell Dev Biol 2022; 10:1020470. [DOI: 10.3389/fcell.2022.1020470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022] Open
|
13
|
Dvoran M, Nemcova L, Kalous J. An Interplay between Epigenetics and Translation in Oocyte Maturation and Embryo Development: Assisted Reproduction Perspective. Biomedicines 2022; 10:biomedicines10071689. [PMID: 35884994 PMCID: PMC9313063 DOI: 10.3390/biomedicines10071689] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/25/2022] [Accepted: 06/28/2022] [Indexed: 12/11/2022] Open
Abstract
Germ cell quality is a key prerequisite for successful fertilization and early embryo development. The quality is determined by the fine regulation of transcriptomic and proteomic profiles, which are prone to alteration by assisted reproduction technology (ART)-introduced in vitro methods. Gaining evidence shows the ART can influence preset epigenetic modifications within cultured oocytes or early embryos and affect their developmental competency. The aim of this review is to describe ART-determined epigenetic changes related to the oogenesis, early embryogenesis, and further in utero development. We confront the latest epigenetic, related epitranscriptomic, and translational regulation findings with the processes of meiotic maturation, fertilization, and early embryogenesis that impact the developmental competency and embryo quality. Post-ART embryo transfer, in utero implantation, and development (placentation, fetal development) are influenced by environmental and lifestyle factors. The review is emphasizing their epigenetic and ART contribution to fetal development. An epigenetic parallel among mouse, porcine, and bovine animal models and human ART is drawn to illustrate possible future mechanisms of infertility management as well as increase the awareness of the underlying mechanisms governing oocyte and embryo developmental complexity under ART conditions.
Collapse
|
14
|
Herta AC, Mengden L, Akin N, Billooye K, Coucke W, Leersum J, Cava-Cami B, Saucedo-Cuevas L, Klamt F, Smitz J, Anckaert E. Characterization of carbohydrate metabolism in in vivo and in vitro grown and matured mouse antral follicles. Biol Reprod 2022; 107:998-1013. [PMID: 35717588 DOI: 10.1093/biolre/ioac124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 02/14/2022] [Accepted: 06/12/2022] [Indexed: 11/13/2022] Open
Abstract
Establishing an ideal human follicle culture system for oncofertility patients relies mainly on animal models since donor tissue is scarce and often of suboptimal quality. The in vitro system developed in our laboratory supports the growth of prepubertal mouse secondary follicles up to mature oocytes. Given the importance of glucose in preparing the oocyte for proper maturation, a baseline characterization of follicle metabolism both in the culture system and in vivo was carried out. Markers of glucose-related pathways (glycolysis, tricarboxylic acid (TCA) cycle, pentose phosphate pathway (PPP), polyol pathway, hexosamine biosynthesis pathway (HBP)) as well as for the antioxidant capacity were measured in the different follicle cell types by both enzymatic activities (spectrophotometric detection) and gene expression (qPCR). This study confirmed that in vivo the somatic cells, mainly granulosa, exhibit intense glycolytic activity, while oocytes perform PPP. Throughout the final maturation step, oocytes in vivo and in vitro showed steady levels for all the key enzymes and metabolites. On the other hand, ovulation triggers a boost of pyruvate and lactate uptake in cumulus cells in vivo, consumes reduced nicotinamide adenine dinucleotide phosphate (NADPH) and increases TCA cycle and small molecules antioxidant capacity (SMAC) activities, while in vitro, the metabolic upregulation in all the studied pathways is limited. This altered metabolic pattern might be a consequence of cell exhaustion because of culture conditions, impeding cumulus cells to fulfil their role in providing proper support for acquiring oocyte competence. SUMMARY SENTENCE: In vitro cultured mouse follicles exhibit altered glycolytic activity and redox metabolism in the somatic compartment during meiotic maturation.
Collapse
Affiliation(s)
- Anamaria-Cristina Herta
- Follicle Biology Laboratory (FOBI), Vrije Universiteit Brussel (VUB), Brussels, 1090, Belgium
| | - Lucia Mengden
- Laboratory of Cellular Biochemistry, Department of Biochemistry, ICBS, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre (RS), 90035003, Brazil
| | - Nazli Akin
- Follicle Biology Laboratory (FOBI), Vrije Universiteit Brussel (VUB), Brussels, 1090, Belgium
| | - Katy Billooye
- Follicle Biology Laboratory (FOBI), Vrije Universiteit Brussel (VUB), Brussels, 1090, Belgium
| | - Wim Coucke
- Freelance statistician, Brugstraat 107, 3001 Heverlee, Belgium
| | - Julia Leersum
- Follicle Biology Laboratory (FOBI), Vrije Universiteit Brussel (VUB), Brussels, 1090, Belgium
| | - Berta Cava-Cami
- Follicle Biology Laboratory (FOBI), Vrije Universiteit Brussel (VUB), Brussels, 1090, Belgium
| | - Laura Saucedo-Cuevas
- Follicle Biology Laboratory (FOBI), Vrije Universiteit Brussel (VUB), Brussels, 1090, Belgium
| | - Fábio Klamt
- Laboratory of Cellular Biochemistry, Department of Biochemistry, ICBS, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre (RS), 90035003, Brazil
| | - Johan Smitz
- Follicle Biology Laboratory (FOBI), Vrije Universiteit Brussel (VUB), Brussels, 1090, Belgium
| | - Ellen Anckaert
- Follicle Biology Laboratory (FOBI), Vrije Universiteit Brussel (VUB), Brussels, 1090, Belgium
| |
Collapse
|
15
|
Zhu S, Wang Q. Metabolic control of oocyte development. Biol Reprod 2022; 107:54-61. [PMID: 35470861 DOI: 10.1093/biolre/ioac082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 03/19/2022] [Accepted: 04/18/2022] [Indexed: 11/12/2022] Open
Abstract
Well balanced and timed metabolism is essential for oocyte development. The effects of extrinsic nutrients on oocyte maturation have been widely reported. In contrast, intrinsic control of oogenesis by intracellular metabolites and metabolic enzymes has received little attention. The comprehensive characterization of metabolic patterns could lead to more complete understanding of regulatory mechanisms underlying oocyte development. A cell's metabolic state is integrated with epigenetic regulation. Epigenetic modifications in germ cells are therefore sensitive to parental environmental exposures. Nevertheless, direct genetic evidence for metabolites involvement in epigenetic establishment during oocyte development is still lacking. Moreover, metabolic disorder-induced epigenetic perturbations during oogenesis might mediate the inter/transgenerational effects of environmental insults. The molecular mechanisms responsible for this deserve further investigation. Here, we summarize the findings on metabolic regulation in oocyte maturation, and how it contributes to oocyte epigenetic modification. Finally, we propose a mouse model that metabolic disorder in oocyte serves as a potential factor mediating the maternal environment effects on offspring health.
Collapse
Affiliation(s)
- Shuai Zhu
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, Nanjing 211166, China
| | - Qiang Wang
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, Nanjing 211166, China.,Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
16
|
Bjarkadottir BD, Walker CA, Fatum M, Lane S, Williams SA. Analysing culture methods of frozen human ovarian tissue to improve follicle survival. REPRODUCTION AND FERTILITY 2022; 2:59-68. [PMID: 35128433 PMCID: PMC8812444 DOI: 10.1530/raf-20-0058] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 02/25/2021] [Indexed: 11/18/2022] Open
Abstract
In vitro follicle growth is a potential fertility preservation method for patients for whom current methods are contraindicated. Currently, this method has only been successful using fresh ovarian tissue. Since many patients who may benefit from this treatment currently have cryopreserved ovarian tissue in storage, optimising in vitro follicle growth (IVG) for cryopreserved-thawed tissue is critical. This study sought to improve the first step of IVG by comparing different short-term culture systems for cryopreserved-thawed human ovarian tissue, in order to yield a higher number of healthy multilayer follicles. We compared two commonly used culture media (αMEM and McCoy’s 5A), and three plate conditions (300 µL, 1 mL on a polycarbonate membrane and 1 mL in a gas-permeable plate) on the health and development of follicles after 6 days of culture. A total of 5797 follicles from three post-pubertal patients (aged 21.3 ± 2.3 years) were analysed across six different culture conditions and non-cultured control. All culture systems supported follicle development and there was no difference in developmental progression between the different conditions tested. Differences in follicle morphology were evident with follicles cultured in low volume conditions having significantly greater odds of being graded as morphologically normal compared to other conditions. Furthermore, culture in a low volume of αMEM resulted in the highest proportion of morphologically normal primary and multilayer follicles (23.8% compared to 6.3-19.9% depending on condition). We, therefore, recommend culturing cryopreserved human ovarian tissue in a low volume of αMEM to support follicle health and development.
Collapse
Affiliation(s)
- Briet D Bjarkadottir
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, UK
| | - Charlotte A Walker
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, UK
| | - Muhammad Fatum
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, UK.,Department of Paediatric Oncology and Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Sheila Lane
- Oxford Fertility, Institute of Reproductive Sciences, Oxford, UK
| | - Suzannah A Williams
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, UK
| |
Collapse
|
17
|
Xu J, Zelinski MB. Oocyte quality following in vitro follicle development†. Biol Reprod 2021; 106:291-315. [PMID: 34962509 PMCID: PMC9004734 DOI: 10.1093/biolre/ioab242] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/15/2021] [Accepted: 12/24/2021] [Indexed: 12/30/2022] Open
Abstract
In vitro follicle development (IVFD) is an adequate model to obtain basic knowledge of folliculogenesis and provides a tool for ovarian toxicity screening. IVFD yielding competent oocytes may also offer an option for fertility and species preservation. To promote follicle growth and oocyte maturation in vitro, various culture systems are utilized for IVFD in rodents, domestic animals, wild animals, nonhuman primates, and humans. Follicle culture conditions have been improved by optimizing gonadotropin levels, regulatory factors, nutrient supplements, oxygen concentration, and culture matrices. This review summarizes quality assessment of oocytes generated from in vitro-developed antral follicles from the preantral stage, including oocyte epigenetic and genetic profile, cytoplasmic and nuclear maturation, preimplantation embryonic development following in vitro fertilization, as well as pregnancy and live offspring after embryo transfer. The limitations of oocyte quality evaluation following IVFD and the gaps in our knowledge of IVFD to support proper oocyte development are also discussed. The information may advance our understanding of the requirements for IVFD, with a goal of producing competent oocytes with genetic integrity to sustain embryonic development resulting in healthy offspring.
Collapse
Affiliation(s)
- Jing Xu
- Correspondence: Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR 97006, USA. Tel: +1 5033465411; Fax: +1 5033465585; E-mail:
| | - Mary B Zelinski
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA,Department of Obstetrics and Gynecology, School of Medicine, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
18
|
Pan Y, Zhu J, Lv Q, Shi D, Yang S, Xing Q, Zhang R, Cheng J, Deng Y. Follicle-stimulating hormone regulates glycolysis of water buffalo follicular granulosa cells through AMPK/SIRT1 signalling pathway. Reprod Domest Anim 2021; 57:185-195. [PMID: 34741362 DOI: 10.1111/rda.14039] [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: 06/29/2021] [Accepted: 11/02/2021] [Indexed: 01/04/2023]
Abstract
Glycolysis in follicular granulosa cells (GCs) is the primary source of energy metabolism substrate of oocytes and is closely related to follicular development in mammals. Many physiological functions of GCs are regulated by follicle-stimulating hormone (FSH). In contrast, whether FSH regulates the glycolysis of GCs and its mechanism remains unclear. This study explored the correlation between FSH concentration and glycolysis level of GCs from different diameters of water buffalo follicles, and further explored the mechanism of FSH regulation in glycolysis in vitro cultured GCs. Results showed the variation trend of lactic acid concentration in follicular fluid and the expression level of glycolysis-related genes in GCs were consistent with the variation trend of FSH concentration in follicular fluid from follicles with different diameters. When GCs were treated with FSH in vitro, the expression level of glycolysis-related genes, lactate production and glucose uptake increased correspondingly (p < .05). Furthermore, we found that expression trend of AMPK/Sirtuin1 (SIRT1) pathway-related genes in GCs was consistent with the expression trend of glycolysis-related genes and was positively correlated with FSH concentrations in vivo or cultured in vitro. Activation of SIRT1 increased the expression level of glycolytic key proteins and lactic acid production in GCs, while inhibition of SIRT1 showed the opposite effect. In general, glycolysis in water buffalo GCs in vivo or cultured in vitro was positively correlated with FSH concentration. AMPK/SIRT1 pathway plays an important role in the regulation of FSH on glycolysis in GCs. Our findings will enrich the understanding of FSH regulating the development of water buffalo follicles.
Collapse
Affiliation(s)
- Yu Pan
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Animal Reproduction Institute, Guangxi University, Nanning, China
| | - Jianzong Zhu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Animal Reproduction Institute, Guangxi University, Nanning, China
| | - Qiao Lv
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Animal Reproduction Institute, Guangxi University, Nanning, China
| | - Deshun Shi
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Animal Reproduction Institute, Guangxi University, Nanning, China
| | - Sufang Yang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Animal Reproduction Institute, Guangxi University, Nanning, China
| | - Qinghua Xing
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Animal Reproduction Institute, Guangxi University, Nanning, China
| | - Ruimen Zhang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Animal Reproduction Institute, Guangxi University, Nanning, China
| | - Juanru Cheng
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Animal Reproduction Institute, Guangxi University, Nanning, China
| | - Yanfei Deng
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Animal Reproduction Institute, Guangxi University, Nanning, China
| |
Collapse
|
19
|
Correlation between Pre-Ovulatory Follicle Diameter and Follicular Fluid Metabolome Profiles in Lactating Beef Cows. Metabolites 2021; 11:metabo11090623. [PMID: 34564438 PMCID: PMC8471867 DOI: 10.3390/metabo11090623] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/10/2021] [Accepted: 09/11/2021] [Indexed: 01/01/2023] Open
Abstract
Induced ovulation of small pre-ovulatory follicles reduced pregnancy rates, embryo survival, day seven embryo quality, and successful embryo cleavage in beef cows undergoing fixed-time artificial insemination. RNA-sequencing of oocytes and associated cumulus cells collected from pre-ovulatory follicles 23 h after gonadotropin-releasing hormone (GnRH) administration to induce the pre-ovulatory gonadotropin surge suggested reduced capacity for glucose metabolism in cumulus cells of follicles ≤11.7 mm. We hypothesized that the follicular fluid metabolome influences metabolic capacity of the cumulus-oocyte complex and contributes to reduced embryo cleavage and quality grade observed following induced ovulation of small follicles. Therefore, we performed a study to determine the correlation between pre-ovulatory follicle diameter and follicular fluid metabolome profiles in lactating beef cows (Angus, n = 130). We synchronized the development of a pre-ovulatory follicle and collected the follicular contents approximately 20 h after GnRH administration. We then performed ultra-high performance liquid chromatography-high resolution mass spectrometry (UHPLC-HRMS) metabolomic studies on 43 follicular fluid samples and identified 38 metabolites within pre-ovulatory follicles of increasing size. We detected 18 metabolites with a significant, positive correlation to follicle diameter. Individual and pathway enrichment analysis of significantly correlated metabolites suggest that altered glucose and amino acid metabolism likely contribute to reduced developmental competence of oocytes when small pre-ovulatory follicles undergo induced ovulation.
Collapse
|
20
|
Association of follicular fluid parameters at the time of ovum pick-up with in vitro blastocyst formation in Holstein heifers: a multi-level analysis. Livest Sci 2021. [DOI: 10.1016/j.livsci.2021.104647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
21
|
Cacciottola L, Donnez J, Dolmans MM. Ovarian tissue damage after grafting: systematic review of strategies to improve follicle outcomes. Reprod Biomed Online 2021; 43:351-369. [PMID: 34384692 DOI: 10.1016/j.rbmo.2021.06.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/14/2021] [Accepted: 06/21/2021] [Indexed: 12/17/2022]
Abstract
Frozen-thawed human ovarian tissue endures large-scale follicle loss in the early post-grafting period, characterized by hypoxia lasting around 7 days. Tissue revascularization occurs progressively through new vessel invasion from the host and neoangiogenesis from the graft. Such reoxygenation kinetics lead to further potential damage caused by oxidative stress. The aim of the present manuscript is to provide a systematic review of proangiogenic growth factors, hormones and various antioxidants administered in the event of ovarian tissue transplantation to protect the follicle pool from depletion by boosting revascularization or decreasing oxidative stress. Although almost all investigated studies revealed an advantage in terms of revascularization and reduction in oxidative stress, far fewer demonstrated a positive impact on follicle survival. As the cascade of events driven by ischaemia after transplantation is a complex process involving numerous players, it appears that acting on specific molecular mechanisms, such as concentrations of proangiogenic growth factors, is not enough to significantly mitigate tissue damage. Strategies exploiting the activated tissue response to ischaemia for tissue healing and remodelling purposes, such as the use of antiapoptotic drugs and adult stem cells, are also discussed in the present review, since they yielded promising results in terms of follicle pool protection.
Collapse
Affiliation(s)
- Luciana Cacciottola
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Jacques Donnez
- Prof. Emeritus, Université Catholique de Louvain, Brussels, Belgium
| | - Marie-Madeleine 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
|
22
|
Moorey SE, Monnig JM, Smith MF, Ortega MS, Green JA, Pohler KG, Bridges GA, Behura SK, Geary TW. Differential Transcript Profiles in Cumulus-Oocyte Complexes Originating from Pre-Ovulatory Follicles of Varied Physiological Maturity in Beef Cows. Genes (Basel) 2021; 12:genes12060893. [PMID: 34200628 PMCID: PMC8227736 DOI: 10.3390/genes12060893] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 05/27/2021] [Accepted: 05/27/2021] [Indexed: 12/13/2022] Open
Abstract
Small dominant follicle diameter at induced ovulation, but not at spontaneous ovulation, decreased pregnancy rate, fertilization rate, and day seven embryo quality in beef cows. We hypothesized that the physiological status of the follicle at GnRH-induced ovulation has a direct effect on the transcriptome of the Cumulus-Oocyte complex, thereby affecting oocyte competence and subsequent embryo development. The objective of this study was to determine if the transcriptome of oocytes and associated cumulus cells (CC) differed among small (≤11.7 mm) and large follicles (≥12.7 mm) exposed to a GnRH-induced gonadotropin surge and follicles (11.7–14.0 mm) exposed to an endogenous gonadotropin surge (spontaneous follicles). RNA sequencing data, from pools of four oocytes or their corresponding CC, revealed 69, 94, and 83 differentially expressed gene transcripts (DEG) among oocyte pools from small versus large, small versus spontaneous, and large versus spontaneous follicle classifications, respectively. An additional 128, 98, and 80 DEG were identified among small versus large, small versus spontaneous, and large versus spontaneous follicle CC pools, respectively. The biological pathway “oxidative phosphorylation” was significantly enriched with DEG from small versus spontaneous follicle oocyte pools (FDR < 0.01); whereas the glycolytic pathway was significantly enriched with DEG from CC pools obtained from large versus small follicles (FDR < 0.01). These findings collectively suggest that altered carbohydrate metabolism within the Cumulus-Oocyte complex likely contributes to the decreased competency of oocytes from small pre-ovulatory follicles exposed to an exogenous GnRH-induced gonadotropin surge.
Collapse
Affiliation(s)
- Sarah E. Moorey
- Department of Animal Science, University of Tennessee, Knoxville, TN 37996, USA
- Correspondence:
| | - Jenna M. Monnig
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA; (J.M.M.); (M.F.S.); (M.S.O.); (J.A.G.); (S.K.B.)
| | - Michael F. Smith
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA; (J.M.M.); (M.F.S.); (M.S.O.); (J.A.G.); (S.K.B.)
| | - M. Sofia Ortega
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA; (J.M.M.); (M.F.S.); (M.S.O.); (J.A.G.); (S.K.B.)
| | - Jonathan A. Green
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA; (J.M.M.); (M.F.S.); (M.S.O.); (J.A.G.); (S.K.B.)
| | - Ky G. Pohler
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA;
| | - G. Alan Bridges
- Department of Animal Science, University of Minnesota, St. Paul, MN 55108, USA;
| | - Susanta K. Behura
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA; (J.M.M.); (M.F.S.); (M.S.O.); (J.A.G.); (S.K.B.)
| | - Thomas W. Geary
- USDA-ARS Fort Keogh Livestock and Range Research Lab, Miles City, MT 59301, USA;
| |
Collapse
|
23
|
Supplementation of granulosa cells conditioned medium with pyruvate and testosterone could improve early follicular development in cultured 1-day-old mouse ovaries. ZYGOTE 2021; 29:468-475. [PMID: 33910666 DOI: 10.1017/s0967199421000174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
In vitro activation of primordial follicles could serve as a safe method to preserve fertility in patients with cancer subjected to ovarian tissue cryopreservation during oncotherapy, however the culture medium for this purpose requires to be optimized. Granulosa cell conditioned medium (GCCM) has been recognized to enhance primordial follicle activation and the present study was conducted to understand whether addition of pyruvate, a combination of insulin, transferrin and selenium (ITS) or testosterone to GCCM could improve its efficiency in this regard. To this end, 1-day-old mouse ovaries were cultured in four different media including CON (control; containing GGCM only), PYR (containing GCCM plus pyruvate), ITS (containing GCCM plus ITS) or TES (containing GCCM plus testosterone) for 11 days. Furthermore, follicular dynamics and gene expression of factors involved in follicular development were assessed using histological examination and RT-PCR, respectively, on days 5 and 11 of culture. Pyruvate decreased follicular activation, but it enhanced the progression of follicles to the primary stage. Moreover, it upregulated Bmp15 and Cx37 (P < 0.05). In the ITS group, activation of follicles was not affected and total number of follicles was reduced by day 11 of culture. Additionally, ITS downregulated Pi3k, Gdf9, Bmp15 and Cx37 (P < 0.05). Although testosterone did not affect primordial follicle activation, it enhanced the development of follicles up to the preantral stage (P < 0.05). Furthermore, testosterone inhibited the expression of Pten but stimulated the expression of Gdf9 and Cx37 (P < 0.05). In conclusion, the present study revealed that inclusion of pyruvate and testosterone into GCCM could enhance the early development of follicles in cultured 1-day-old mouse ovaries.
Collapse
|
24
|
Esmaeili-Fard SM, Gholizadeh M, Hafezian SH, Abdollahi-Arpanahi R. Genome-wide association study and pathway analysis identify NTRK2 as a novel candidate gene for litter size in sheep. PLoS One 2021; 16:e0244408. [PMID: 33481819 PMCID: PMC7822323 DOI: 10.1371/journal.pone.0244408] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 12/08/2020] [Indexed: 12/14/2022] Open
Abstract
Litter size is one of the most important economic traits in sheep. Identification of gene variants that are associated with the prolificacy rate is an important step in breeding program success and profitability of the farm. So, to identify genetic mechanisms underlying the variation in litter size in Iranian Baluchi sheep, a two-step genome-wide association study (GWAS) was performed. GWAS was conducted using genotype data from 91 Baluchi sheep. Estimated breeding values (EBVs) for litter size calculated for 3848 ewes and then used as the response variable. Besides, a pathway analysis using GO and KEGG databases were applied as a complementary approach. A total of three single nucleotide polymorphisms (SNPs) associated with litter size were identified, one each on OAR2, OAR10, and OAR25. The SNP on OAR2 is located within a novel putative candidate gene, Neurotrophic receptor tyrosine kinase 2. This gene product works as a receptor which is essential for follicular assembly, early follicular growth, and oocyte survival. The SNP on OAR25 is located within RAB4A which is involved in blood vessel formation and proliferation through angiogenesis. The SNP on OAR10 was not associated with any gene in the 1Mb span. Moreover, gene-set analysis using the KEGG database identified several pathways, such as Ovarian steroidogenesis, Steroid hormone biosynthesis, Calcium signaling pathway, and Chemokine signaling. Also, pathway analysis using the GO database revealed several functional terms, such as cellular carbohydrate metabolic, biological adhesion, cell adhesion, cell junction, and cell-cell adherens junction, among others. This is the first study that reports the NTRK2 gene affecting litter size in sheep and our study of this gene functions showed that this gene could be a good candidate for further analysis.
Collapse
Affiliation(s)
- Seyed Mehdi Esmaeili-Fard
- Department of Animal Sciences and Fisheries, Sari Agricultural Sciences and Natural Resources University (SANRU), Sari, Iran
- * E-mail:
| | - Mohsen Gholizadeh
- Department of Animal Sciences and Fisheries, Sari Agricultural Sciences and Natural Resources University (SANRU), Sari, Iran
| | - Seyed Hasan Hafezian
- Department of Animal Sciences and Fisheries, Sari Agricultural Sciences and Natural Resources University (SANRU), Sari, Iran
| | | |
Collapse
|
25
|
Grandhaye J, Hmadeh S, Plotton I, Levasseur F, Estienne A, LeGuevel R, Levern Y, Ramé C, Jeanpierre E, Guerif F, Dupont J, Froment P. The adiponectin agonist, AdipoRon, inhibits steroidogenesis and cell proliferation in human luteinized granulosa cells. Mol Cell Endocrinol 2021; 520:111080. [PMID: 33189865 DOI: 10.1016/j.mce.2020.111080] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 10/13/2020] [Accepted: 11/09/2020] [Indexed: 12/11/2022]
Abstract
During obesity, excess body weight is not only associated with an increased risk of type 2-diabetes, but also several other pathological processes, such as infertility. Adipose tissue is the largest endocrine organ of the body that produces adipokines, including adiponectin. Adiponectin has been reported to control fertility through the hypothalamic-pituitary-gonadal axis, and folliculogenesis in the ovaries. In this study, we focused on a recent adiponectin-like synthetic agonist called AdipoRon, and its action in human luteinized granulosa cells. We demonstrated that AdipoRon activated the adenosine monophosphate-activated protein kinase (AMPK) and peroxisome proliferator-activated receptor alpha (PPAR) signalling pathways in human luteinized granulosa cells. A 25 μM AdipoRon stimulation reduced granulosa cell proliferation by inducing cell cycle arrest in G1, associated with PTEN and p53 pathway activation. In addition, AdipoRon perturbed cell metabolism by decreasing mitochondrial activity and ATP production. In human luteinized granulosa cells, AdipoRon increased phosphodiesterase activity, leading to a drop in cyclic adenosine monophosphate (cAMP) production, aromatase expression and oestrogens secretion. In conclusion, AdipoRon impacted folliculogenesis by altering human luteinized granulosa cell function, via steroid production and cell proliferation. This agonist may have applications for improving ovarian function in metabolic disorders or granulosa cancers.
Collapse
Affiliation(s)
- Jérémy Grandhaye
- INRAE UMR85 Physiologie de la Reproduction et des Comportements, Nouzilly, France; CNRS UMR7247 Physiologie de la Reproduction et des Comportements, Nouzilly, France; Université de Tours, Tours, France; IFCE, Nouzilly, France
| | - Sandy Hmadeh
- INRAE UMR85 Physiologie de la Reproduction et des Comportements, Nouzilly, France; CNRS UMR7247 Physiologie de la Reproduction et des Comportements, Nouzilly, France; Université de Tours, Tours, France; IFCE, Nouzilly, France
| | - Ingrid Plotton
- Molecular Endocrinology and Rare Diseases, University Hospital, Claude Bernard Lyon 1 University, Bron, France
| | - Floriane Levasseur
- INRAE UMR85 Physiologie de la Reproduction et des Comportements, Nouzilly, France; CNRS UMR7247 Physiologie de la Reproduction et des Comportements, Nouzilly, France; Université de Tours, Tours, France; IFCE, Nouzilly, France
| | - Anthony Estienne
- INRAE UMR85 Physiologie de la Reproduction et des Comportements, Nouzilly, France; CNRS UMR7247 Physiologie de la Reproduction et des Comportements, Nouzilly, France; Université de Tours, Tours, France; IFCE, Nouzilly, France
| | - Rémy LeGuevel
- Plate-forme ImPACcell, Université de Rennes 1, France
| | - Yves Levern
- INRA UMR Infectiologie et Santé Publique, Service de Cytométrie, Nouzilly, France
| | - Christelle Ramé
- INRAE UMR85 Physiologie de la Reproduction et des Comportements, Nouzilly, France; CNRS UMR7247 Physiologie de la Reproduction et des Comportements, Nouzilly, France; Université de Tours, Tours, France; IFCE, Nouzilly, France
| | - Eric Jeanpierre
- INRAE UMR85 Physiologie de la Reproduction et des Comportements, Nouzilly, France; CNRS UMR7247 Physiologie de la Reproduction et des Comportements, Nouzilly, France; Université de Tours, Tours, France; IFCE, Nouzilly, France
| | | | - Joëlle Dupont
- INRAE UMR85 Physiologie de la Reproduction et des Comportements, Nouzilly, France; CNRS UMR7247 Physiologie de la Reproduction et des Comportements, Nouzilly, France; Université de Tours, Tours, France; IFCE, Nouzilly, France
| | - Pascal Froment
- INRAE UMR85 Physiologie de la Reproduction et des Comportements, Nouzilly, France; CNRS UMR7247 Physiologie de la Reproduction et des Comportements, Nouzilly, France; Université de Tours, Tours, France; IFCE, Nouzilly, France.
| |
Collapse
|
26
|
Vilela JDMV, Dolmans MM, Amorim CA. Ovarian tissue transportation: a systematic review. Reprod Biomed Online 2020; 42:351-365. [PMID: 33288476 DOI: 10.1016/j.rbmo.2020.11.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 11/06/2020] [Accepted: 11/06/2020] [Indexed: 12/20/2022]
Abstract
In recent years, some countries and fertility preservation networks have started adopting 24 h transportation for ovarian tissue, a practice that has the potential to spread very quickly due to the high costs and bureaucracy involved in the establishment of ovarian tissue cryobanks. While pregnancies and live births have been reported after such long periods of transportation, this, however, remains an empirical procedure. This review aims to prompt reflection on ovarian tissue transport, highlighting the lack of knowledge in humans by providing a counterpoint looking into more than 40 studies published in different animal models. By discussing these studies in animals, the findings of various models can be deciphered, and light shed on the patterns identified. Like the development of different assisted reproductive technology procedures, this is an important step in creating guidelines for future studies on human ovarian tissue transportation.
Collapse
Affiliation(s)
| | - Marie-Madeleine Dolmans
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium; Gynecology Department, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Christiani Andrade Amorim
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium.
| |
Collapse
|
27
|
Li L, Zhu S, Shu W, Guo Y, Guan Y, Zeng J, Wang H, Han L, Zhang J, Liu X, Li C, Hou X, Gao M, Ge J, Ren C, Zhang H, Schedl T, Guo X, Chen M, Wang Q. Characterization of Metabolic Patterns in Mouse Oocytes during Meiotic Maturation. Mol Cell 2020; 80:525-540.e9. [PMID: 33068521 DOI: 10.1016/j.molcel.2020.09.022] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 08/07/2020] [Accepted: 09/21/2020] [Indexed: 12/16/2022]
Abstract
Well-balanced and timed metabolism is essential for making a high-quality egg. However, the metabolic framework that supports oocyte development remains poorly understood. Here, we obtained the temporal metabolome profiles of mouse oocytes during in vivo maturation by isolating large number of cells at key stages. In parallel, quantitative proteomic analyses were conducted to bolster the metabolomic data, synergistically depicting the global metabolic patterns in oocytes. In particular, we discovered the metabolic features during meiotic maturation, such as the fall in polyunsaturated fatty acids (PUFAs) level and the active serine-glycine-one-carbon (SGOC) pathway. Using functional approaches, we further identified the key targets mediating the action of PUFA arachidonic acid (ARA) on meiotic maturation and demonstrated the control of epigenetic marks in maturing oocytes by SGOC network. Our data serve as a broad resource on the dynamics occurring in metabolome and proteome during oocyte maturation.
Collapse
Affiliation(s)
- Ling Li
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, Nanjing 211166, China
| | - Shuai Zhu
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, Nanjing 211166, China
| | - Wenjie Shu
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Yueshuai Guo
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, Nanjing 211166, China
| | - Yusheng Guan
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Juan Zeng
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, Nanjing 211166, China
| | - Haichao Wang
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, Nanjing 211166, China
| | - Longsen Han
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, Nanjing 211166, China
| | - Jiaqi Zhang
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, Nanjing 211166, China
| | - Xiaohui Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Chunling Li
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, Nanjing 211166, China
| | - Xiaojing Hou
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, Nanjing 211166, China
| | - Min Gao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Juan Ge
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, Nanjing 211166, China
| | - Chao Ren
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Hao Zhang
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, Nanjing 211166, China; Department of Histology and Embryology, Nanjing Medical University, Nanjing 211166, China
| | - Tim Schedl
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Xuejiang Guo
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, Nanjing 211166, China; Department of Histology and Embryology, Nanjing Medical University, Nanjing 211166, China.
| | - Minjian Chen
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China.
| | - Qiang Wang
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, Nanjing 211166, China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
28
|
Kim EH, Kim GA, Taweechaipaisankul A, Ridlo MR, Lee SH, Ra K, Ahn C, Lee BC. Phytanic acid-derived peroxisomal lipid metabolism in porcine oocytes. Theriogenology 2020; 157:276-285. [PMID: 32823023 DOI: 10.1016/j.theriogenology.2020.07.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 07/03/2020] [Accepted: 07/07/2020] [Indexed: 12/15/2022]
Abstract
Lipid metabolism plays an important role in oocyte maturation. The peroxisome is the fundamental mediator for this mechanism. In this study, we investigated the peroxisomal lipid metabolism in porcine oocytes. Phytanic acid (PA) was chosen as an activator of alpha-oxidation in peroxisomes. Oocyte maturation, embryo development, immunocytochemistry of peroxisomal lipid activities, and staining of mitochondrial potentials were assessed. We found that 40 μM PA not only increased porcine oocyte maturation and embryonic development, but also upregulated the expression of genes and proteins related to activities of the peroxisomal lipid metabolism (PHYH, PEX19, and PEX subfamilies) and mitochondrial potentials (NRF1 and PGC1α). Moreover, PA upregulated the lipid droplet and fatty acid content in the oocytes. Moreover, mitochondria were activated and the mitochondrial membrane potential was increased after PA treatment, resulting in the production of more ATPs in the oocytes. Our findings suggest that the degradation of PA via alpha-oxidation in the peroxisome may potentiate oocyte maturation processes, peroxisomal lipid oxidation, and mitochondria activities.
Collapse
Affiliation(s)
- Eui Hyun Kim
- Department of Theriogenology and Biotechnology, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Geon A Kim
- Department of Theriogenology and Biotechnology, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea; Department of Biomedical Laboratory Science, School of Medicine, Eulji University, Daejeon, Republic of Korea
| | - Anukul Taweechaipaisankul
- Department of Theriogenology and Biotechnology, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Muhammad Rosyid Ridlo
- Department of Theriogenology and Biotechnology, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea; Department of Bioresource Technology and Veterinary, Vocational College, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Seok Hee Lee
- Department of Theriogenology and Biotechnology, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Kihae Ra
- Department of Theriogenology and Biotechnology, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Curie Ahn
- Division of Nephrology, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Byeong Chun Lee
- Department of Theriogenology and Biotechnology, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
29
|
Xu S, Wu X, Dong Y, Xu M, Li Z, Chen S, Zhuo Y, Lin Y, Che L, Fang Z, Feng B, Li J, Wang J, Wu D, Ren Z. Glucose activates the primordial follicle through the AMPK/mTOR signaling pathway. Clin Transl Med 2020. [PMCID: PMC7418812 DOI: 10.1002/ctm2.122] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Background We have previously found that the energy level in sows affects the activation of primordial follicles. Glucose is the primary metabolic substrate of dietary energy and its effect and mechanism of action with regards to the activation and development of primordial follicle remain unclear. Studies utilizing several different animal cells have shown that energy stress, induced by glucose starvation, activates AMPK and participates in a variety of cellular processes by regulating the Hippo and mTOR signaling pathways. However, whether glucose can affect primordial follicle activation through the above pathways has not been reported. Methods We developed an in vitro culture system for mouse ovaries to investigate the effects of glucose on the primordial follicle activation. Protein expression of AMPK‐Hippo‐YAP and AMPK‐mTOR pathway was investigated under glucose starvation and optimal glucose level treatment. Then, ovaries were treated with AICAR or Compound C in vitro to explore the effect of AMPK activation or inhibition on primordial follicle activation, and the changes of AMPK‐Hippo‐YAP and AMPK‐mTOR signaling pathways. Finally, investigated the signaling pathways affected by glucose potentially affecting the primordial follicle activation in vivo. Results The glucose was an essential nutrient for primordial follicle activation and we identified 25 mM glucose as the optimal level (P < .05) for the primordial follicle activation in vitro. The glycolysis pathway was involved in primordial follicle activation (P < .05) of ovaries cultured in vitro. The glucose affected the activation of primordial follicles in vitro through AMPK/mTOR signaling pathway by AMPK activation or inhibition treatment and follicle ratio count (P < .05). Moreover, glucose affected the primordial follicle activation of ovary in vivo via mTOR signaling pathway. Conclusions This study demonstrates that glucose affects the primordial follicle activation through the AMPK/mTOR rather than the AMPK/Hippo signaling pathway.
Collapse
Affiliation(s)
- Shengyu Xu
- Animal Nutrition Institute, Sichuan Agricultural University; Key Laboratory of Animal Disease‐resistant Nutrition, Ministry of EducationMinistry of Agriculture and Rural Affairs, Sichuan Province Chengdu Sichuan P. R. China
| | - Xiaoling Wu
- Animal Nutrition Institute, Sichuan Agricultural University; Key Laboratory of Animal Disease‐resistant Nutrition, Ministry of EducationMinistry of Agriculture and Rural Affairs, Sichuan Province Chengdu Sichuan P. R. China
| | - Yanpeng Dong
- Animal Nutrition Institute, Sichuan Agricultural University; Key Laboratory of Animal Disease‐resistant Nutrition, Ministry of EducationMinistry of Agriculture and Rural Affairs, Sichuan Province Chengdu Sichuan P. R. China
| | - Mengmeng Xu
- Animal Nutrition Institute, Sichuan Agricultural University; Key Laboratory of Animal Disease‐resistant Nutrition, Ministry of EducationMinistry of Agriculture and Rural Affairs, Sichuan Province Chengdu Sichuan P. R. China
| | - Zimei Li
- Animal Nutrition Institute, Sichuan Agricultural University; Key Laboratory of Animal Disease‐resistant Nutrition, Ministry of EducationMinistry of Agriculture and Rural Affairs, Sichuan Province Chengdu Sichuan P. R. China
| | - Sirun Chen
- Animal Nutrition Institute, Sichuan Agricultural University; Key Laboratory of Animal Disease‐resistant Nutrition, Ministry of EducationMinistry of Agriculture and Rural Affairs, Sichuan Province Chengdu Sichuan P. R. China
| | - Yong Zhuo
- Animal Nutrition Institute, Sichuan Agricultural University; Key Laboratory of Animal Disease‐resistant Nutrition, Ministry of EducationMinistry of Agriculture and Rural Affairs, Sichuan Province Chengdu Sichuan P. R. China
| | - Yan Lin
- Animal Nutrition Institute, Sichuan Agricultural University; Key Laboratory of Animal Disease‐resistant Nutrition, Ministry of EducationMinistry of Agriculture and Rural Affairs, Sichuan Province Chengdu Sichuan P. R. China
| | - Lianqiang Che
- Animal Nutrition Institute, Sichuan Agricultural University; Key Laboratory of Animal Disease‐resistant Nutrition, Ministry of EducationMinistry of Agriculture and Rural Affairs, Sichuan Province Chengdu Sichuan P. R. China
| | - Zhengfeng Fang
- Animal Nutrition Institute, Sichuan Agricultural University; Key Laboratory of Animal Disease‐resistant Nutrition, Ministry of EducationMinistry of Agriculture and Rural Affairs, Sichuan Province Chengdu Sichuan P. R. China
| | - Bin Feng
- Animal Nutrition Institute, Sichuan Agricultural University; Key Laboratory of Animal Disease‐resistant Nutrition, Ministry of EducationMinistry of Agriculture and Rural Affairs, Sichuan Province Chengdu Sichuan P. R. China
| | - Jian Li
- Animal Nutrition Institute, Sichuan Agricultural University; Key Laboratory of Animal Disease‐resistant Nutrition, Ministry of EducationMinistry of Agriculture and Rural Affairs, Sichuan Province Chengdu Sichuan P. R. China
| | - Jianping Wang
- Animal Nutrition Institute, Sichuan Agricultural University; Key Laboratory of Animal Disease‐resistant Nutrition, Ministry of EducationMinistry of Agriculture and Rural Affairs, Sichuan Province Chengdu Sichuan P. R. China
| | - De Wu
- Animal Nutrition Institute, Sichuan Agricultural University; Key Laboratory of Animal Disease‐resistant Nutrition, Ministry of EducationMinistry of Agriculture and Rural Affairs, Sichuan Province Chengdu Sichuan P. R. China
| | - Zhihua Ren
- College of Veterinary Medicine, Sichuan Province Key Laboratory of Animal Disease and Human Health, Key Laboratory of Environmental Hazard and Human Health of Sichuan ProvinceSichuan Agricultural University Chengdu Sichuan P. R. China
| |
Collapse
|
30
|
Energy metabolism of the equine cumulus oocyte complex during in vitro maturation. Sci Rep 2020; 10:3493. [PMID: 32103136 PMCID: PMC7044441 DOI: 10.1038/s41598-020-60624-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 02/11/2020] [Indexed: 11/22/2022] Open
Abstract
Horses are one of the few species, beside humans, in which assisted reproductive technology has important clinical applications. Furthermore, the horse can serve as a valuable model for the study of comparative reproductive biology. Here we present the first comprehensive characterisation of energy metabolism and mitochondrial efficiency in equine cumulus-oocyte complexes (COCs) during in vitro maturation (IVM), as determined using a combination of non-invasive consumption and release assays and mitochondrial function analysis. These data reveal notable species-specific differences in the rate and kinetics of glucose consumption and glycolysis throughout IVM. Approximately 95% of glucose consumed was accounted for by lactate production; however, high concurrent oxygen consumption indicated a comparatively increased role for non-glycolytic oxidative phosphorylation. Up to 38% of equine COC oxygen consumption could be attributed to non-mitochondrial activities and there was a significant loss of spare respiratory capacity over the course of IVM. Notably, our data also revealed that current IVM protocols may be failing to satisfy the metabolic demands of the equine COC. Our findings constitute the first report on mitochondrial efficiency in the equine COC and provide new insight into comparative gamete biology as well as metabolism of the COC during in vitro maturation.
Collapse
|
31
|
Connolly JM, Kane MT, Quinlan LR, Hynes AC. Enhancing oxygen delivery to ovarian follicles by three different methods markedly improves growth in serum-containing culture medium. Reprod Fertil Dev 2019; 31:1339-1352. [PMID: 30975286 DOI: 10.1071/rd18286] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 02/07/2019] [Indexed: 12/12/2022] Open
Abstract
Invitro ovarian follicle culture systems are routinely used to study folliculogenesis and may provide solutions for infertility. Mouse follicles are typically cultured in standard gas-impermeable culture plates under gas phase oxygen concentrations of 5% or 20% (v/v). There is evidence that these conditions may not provide adequate oxygenation for follicles cultured as non-attached intact units in medium supplemented with serum and high levels of FSH. Three different methods of enhancing follicle oxygenation were investigated in this study: increasing the gas phase oxygen concentration, inverting the culture plates and using gas-permeable culture plates. Follicles cultured under 40% O2 were significantly larger (P P P 2 . These effects were associated with reduced secretion of vascular endothelial growth factor (P P P invivo -matured follicles (~500μm in diameter). Such follicular development is not possible under hypoxic conditions.
Collapse
Affiliation(s)
- J M Connolly
- Physiology, National University of Ireland Galway, University Road, Galway, Ireland; and Department of Physiology and Biochemistry, Ross University School of Medicine, Knoxville Campus, 9731 Cogdill Road, Knoxville, TN 37932, USA; and Corresponding author
| | - M T Kane
- Physiology, National University of Ireland Galway, University Road, Galway, Ireland
| | - L R Quinlan
- Physiology, National University of Ireland Galway, University Road, Galway, Ireland
| | - A C Hynes
- Physiology, National University of Ireland Galway, University Road, Galway, Ireland
| |
Collapse
|
32
|
Munakata Y, Shirasuna K, Kuwayama T, Iwata H. Cell-free DNA in medium is associated with the maturation ability of in vitro cultured oocytes. J Reprod Dev 2019; 65:171-175. [PMID: 30745495 PMCID: PMC6473116 DOI: 10.1262/jrd.2018-123] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Follicular fluid contains cell-free DNA (cfDNA), which may serve as a useful biomarker of oocyte ability. The present study evaluates whether nuclear and mitochondrial cfDNAs in conditioned oocyte growth medium determine the quality of oocytes cultured in vitro. Oocyte and granulosa cell complexes (OGCs) derived from early antral follicles of gilt ovaries were cultured for 14 days and the amount of cfDNA and lactate concentration in the conditioned culture medium were measured and compared to evaluate oocyte maturation ability. The amount of nuclear cfDNA, but not mitochondrial cfDNA, strongly correlated with the number of dead cells in OGCs. Furthermore, low mitochondrial cfDNA content and high lactate concentration in the medium was associated with high maturation ability of oocytes cultured in vitro. In conclusion, the amounts of nuclear and mitochondrial cfDNAs differentially reflect the conditions of OGCs, and low mitochondrial cfDNA, low glucose content, and high lactate concentration in the medium are associated with the proper maturation of oocytes.
Collapse
Affiliation(s)
- Yasuhisa Munakata
- Department of Animal Reproduction, Tokyo University of Agriculture, Kanagawa 243-0034, Japan
| | - Koumei Shirasuna
- Department of Animal Reproduction, Tokyo University of Agriculture, Kanagawa 243-0034, Japan
| | - Takehito Kuwayama
- Department of Animal Reproduction, Tokyo University of Agriculture, Kanagawa 243-0034, Japan
| | - Hisataka Iwata
- Department of Animal Reproduction, Tokyo University of Agriculture, Kanagawa 243-0034, Japan
| |
Collapse
|
33
|
Hoque SAM, Kawai T, Zhu Z, Shimada M. Mitochondrial Protein Turnover Is Critical for Granulosa Cell Proliferation and Differentiation in Antral Follicles. J Endocr Soc 2018; 3:324-339. [PMID: 30652133 PMCID: PMC6330174 DOI: 10.1210/js.2018-00329] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 11/27/2018] [Indexed: 12/28/2022] Open
Abstract
Granulosa cell (GC) proliferation is essential for follicular development. FSH is a key factor in GC proliferation, and a continuous supply of high levels of ATP is necessary for cell proliferation. However, genes encoding proteins of the glycolytic pathways are poorly expressed in GCs. Therefore, we hypothesized that mitochondrial gene expression and protein synthesis play a primary role in ATP production during GC proliferation. To test this hypothesis, we performed an in vivo study of GCs collected from 23-day-old mice ovaries with or without equine chorionic gonadotropin (eCG) priming. It was observed that mitochondrial activity with membrane potential, expression of protein-coding genes (Nd1-6, Cytb, Atpase6,8) and transcription-related genes (Polrmt, Tfam, Tfb2m), copy number of mitochondrial (mt-)DNA, and protein synthesis were increased in GCs after 24 hours of eCG injection and mostly maintained elevated up to 48 hours. Therefore, we performed in vitro culture of GCs in DMEM medium supplemented with FSH, testosterone, and serum and containing different glucose concentrations with or without d-chloramphenicol (CRP) for 24 hours. GC proliferation and ATP production were observed to be independent of glucose concentration. Furthermore, FSH-induced mitochondrial activity with membrane potential, ATP content, BrdU-incorporated cell proliferation, intensity of mt-ND1 and mt-ND6 proteins, and expressions of marker genes for proliferation and differentiation were significantly decreased by CRP treatment. These results revealed the crucial role of mitochondria in the supply of ATP and the necessity of mitochondrial gene expression and protein synthesis in not only the proliferation but also the differentiation of GCs during follicular development.
Collapse
Affiliation(s)
- S A Masudul Hoque
- Laboratory of Reproductive Endocrinology, Graduate School of Biosphere Science, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan.,Bangabandhu Sheikh Mujibur Rahman Agricultural University, Gazipur, Bangladesh
| | - Tomoko Kawai
- Laboratory of Reproductive Endocrinology, Graduate School of Biosphere Science, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan
| | - Zhendong Zhu
- Laboratory of Reproductive Endocrinology, Graduate School of Biosphere Science, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan.,College of Animal Science and Technology, Northwest A&F University, Shaanxi, China
| | - Masayuki Shimada
- Laboratory of Reproductive Endocrinology, Graduate School of Biosphere Science, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan
| |
Collapse
|
34
|
Baddela VS, Sharma A, Viergutz T, Koczan D, Vanselow J. Low Oxygen Levels Induce Early Luteinization Associated Changes in Bovine Granulosa Cells. Front Physiol 2018; 9:1066. [PMID: 30131718 PMCID: PMC6090175 DOI: 10.3389/fphys.2018.01066] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 07/17/2018] [Indexed: 11/29/2022] Open
Abstract
During follicle maturation, oxygen levels continuously decrease in the follicular fluid and reach lowest levels in the preovulatory follicle. The current study was designed to comprehensively understand effects of low oxygen levels on bovine granulosa cells (GC) using our established estrogen active GC culture model. As evident from flow cytometry analysis the viability of GC was not found to be affected at severely low oxygen condition (1% O2) compared to normal (atmospheric) oxygen condition (21% O2). Estimations of hormone concentrations using competitive radioimmunoassay revealed that the production of estradiol and progesterone was significantly reduced at low oxygen condition. To understand the genome-wide changes of gene expression, mRNA microarray analysis was performed using Affymetrix's Bovine Gene 1.0 ST Arrays. This resulted in the identification of 1104 differentially regulated genes of which 505 were up- and 599 down-regulated under low oxygen conditions. Pathway analysis using Ingenuity pathway analyzer (IPA) identified 36 significantly affected (p < 0.05) canonical pathways. Importantly, pathways like "Estrogen-mediated S-phase Entry" and "Cyclins and Cell Cycle Regulation" were found to be greatly down-regulated at low oxygen levels. This was experimentally validated using flow cytometry based cell cycle analysis. Up-regulation of critical genes associated with angiogenesis, inflammation, and glucose metabolism, and down-regulation of FSH signaling, steroidogenesis and cell proliferation indicated that low oxygen levels induced early luteinization associated changes in granulosa cells. Identification of unmethylated CpG sites in the CYP19A1 promoter region suggests that granulosa cells were not completely transformed into luteal cells under the present low oxygen in vitro condition. In addition, the comparison with earlier published in vivo microarray data indicated that 1107 genes showed a similar expression pattern in granulosa cells at low oxygen levels (in vitro) as found in preovulatory follicles after the LH surge (in vivo). Overall, our findings demonstrate for the first time that low oxygen levels in preovulatory follicles may play an important role in supporting early events of luteinization in granulosa cells.
Collapse
Affiliation(s)
- Vijay S. Baddela
- Reproductive Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Arpna Sharma
- Reproductive Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Torsten Viergutz
- Reproductive Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Dirk Koczan
- Institute of Immunology, University of Rostock, Rostock, Germany
| | - Jens Vanselow
- Reproductive Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| |
Collapse
|
35
|
Wang H, Cheng Q, Li X, Hu F, Han L, Zhang H, Li L, Ge J, Ying X, Guo X, Wang Q. Loss of TIGAR Induces Oxidative Stress and Meiotic Defects in Oocytes from Obese Mice. Mol Cell Proteomics 2018; 17:1354-1364. [PMID: 29776966 DOI: 10.1074/mcp.ra118.000620] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 04/25/2018] [Indexed: 12/26/2022] Open
Abstract
Maternal obesity has been reported to impair oocyte quality in mice, however, the underlying mechanisms remain unclear. In the present study, by conducting a comparative proteomic analysis, we identified a reduced expression of TIGAR (TP53-induced glycolysis and apoptosis regulator) protein in ovulated oocytes from high-fat diet (HFD)-fed mice. Specific depletion of TIGAR in mouse oocytes results in the marked elevation of reactive oxygen species (ROS) levels and the failure of meiotic apparatus assembly. Importantly, forced expression of TIGAR in HFD oocytes not only attenuates ROS production, but also partly prevents spindle disorganization and chromosome misalignment during meiosis. Meantime, we noted that TIGAR knockdown in oocytes induces a strong activation of autophagy, whereas overexpression of TIGAR significantly reduces the LC3 accumulation in HFD oocytes. By anti-oxidant treatment, we further demonstrated that such an autophagic response is dependent on the TIGAR-controlled ROS production. In summary, our data indicate a role for TIGAR in modulating redox homeostasis during oocyte maturation, and uncover that loss of TIGAR is a critical pathway mediating the effects of maternal obesity on oocyte quality.
Collapse
Affiliation(s)
- Haichao Wang
- From the ‡State Key Laboratory of Reproductive Medicine, Nanjing Medical University
| | - Qing Cheng
- §Departement of Obstetrics, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University
| | - Xiaoyan Li
- From the ‡State Key Laboratory of Reproductive Medicine, Nanjing Medical University.,¶College of Animal Science & Technology, Nanjing Agricultural University
| | - Feifei Hu
- ‖Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China, 210000
| | - Longsen Han
- From the ‡State Key Laboratory of Reproductive Medicine, Nanjing Medical University
| | - Hao Zhang
- From the ‡State Key Laboratory of Reproductive Medicine, Nanjing Medical University
| | - Ling Li
- From the ‡State Key Laboratory of Reproductive Medicine, Nanjing Medical University
| | - Juan Ge
- From the ‡State Key Laboratory of Reproductive Medicine, Nanjing Medical University
| | - Xiaoyan Ying
- ‖Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China, 210000
| | - Xuejiang Guo
- From the ‡State Key Laboratory of Reproductive Medicine, Nanjing Medical University;
| | - Qiang Wang
- From the ‡State Key Laboratory of Reproductive Medicine, Nanjing Medical University;
| |
Collapse
|
36
|
SINGH MANDEEP, HONPARKHE MRIGANK, KUMAR AJEET, SINGHAL SUMIT. Comparison of metabolites in the follicular fluid of bovine preovulatory and cystic ovarian follicles using nuclear magnetic resonance. THE INDIAN JOURNAL OF ANIMAL SCIENCES 2018. [DOI: 10.56093/ijans.v88i3.78261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Estimation of metabolites in cystic and normal preovulatory follicular fluid through proton Nuclear Magnetic Resonance (1H NMR) in cattle suffering from cystic ovarian follicle is highly desirable.The trans-vaginal ultrasound guided ablation was used to collect follicular fluid from cystic (15) and normally cycling (8) dairy cattle. NMR spectra of both fluids were recorded at a resonance frequency of 500.13 MHz on a Bruker Avance-500 spectrometer equipped with solid state probe (5 mm). Spectra were phased manually, baseline corrected, and calibrated against 3-(trimethylsilyl) propionic-2,2,3,3-d4 acid at 0.0 parts per million (ppm) using Prometab software running within MATLAB. The cystic ovarian follicle associated metabolites with variable importance in projection (VIP) scores >2 were lactate (1.98 ppm), UDP-G (5.62), pyruvate (2.34 and 2.38) and creatinine/creatine (3.14) in cystic and normal preovulatory follicular fluid.These metabolites showed identifiable peaks, and thus can be used as potential biomarkers for dairy cattle suffering from cystic ovarian follicle.
Collapse
|
37
|
Anastácio A, Rodriguez-Wallberg KA, Chardonnet S, Pionneau C, Fédérici C, Almeida Santos T, Poirot C. Protein profile of mouse ovarian follicles grown in vitro. Mol Hum Reprod 2017; 23:827-841. [PMID: 29069483 PMCID: PMC5909860 DOI: 10.1093/molehr/gax056] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 08/28/2017] [Accepted: 10/16/2017] [Indexed: 02/02/2023] Open
Abstract
STUDY QUESTION Could the follicle proteome be mapped by identifying specific proteins that are common or differ between three developmental stages from the secondary follicle (SF) to the antrum-like stage? SUMMARY ANSWER From a total of 1401 proteins identified in the follicles, 609 were common to the three developmental stages investigated and 444 were found uniquely at one of the stages. WHAT IS KNOWN ALREADY The importance of the follicle as a functional structure has been recognized; however, up-to-date the proteome of the whole follicle has not been described. A few studies using proteomics have previously reported on either isolated fully-grown oocytes before or after meiosis resumption or cumulus cells. STUDY DESIGN, SIZE, DURATION The experimental design included a validated mice model for isolation and individual culture of SFs. The system was chosen as it allows continuous evaluation of follicle growth and selection of follicles for analysis at pre-determined developmental stages: SF, complete Slavjanski membrane rupture (SMR) and antrum-like cavity (AF). The experiments were repeated 13 times independently to acquire the material that was analyzed by proteomics. PARTICIPANTS/MATERIALS, SETTING, METHODS SFs (n = 2166) were isolated from B6CBA/F1 female mice (n = 42), 12 days old, from 15 l. About half of the follicles isolated as SF were analyzed as such (n = 1143) and pooled to obtain 139 μg of extracted protein. Both SMR (n = 359) and AF (n = 124) were obtained after individual culture of 1023 follicles in a microdrop system under oil, selected for analysis and pooled, to obtain 339 μg and 170 μg of protein, respectively. The follicle proteome was analyzed combining isoelectric focusing (IEF) fractionation with 1D and 2D LC-MS/MS analysis to enhance protein identification. The three protein lists were submitted to the 'Compare gene list' tool in the PANTHER website to gain insights on the Gene Ontology Biological processes present and to Ingenuity Pathway Analysis to highlight protein networks. A label-free quantification was performed with 1D LC-MS/MS analyses to emphasize proteins with different expression profiles between the three follicular stages. Supplementary western blot analysis (using new biological replicates) was performed to confirm the expression variations of three proteins during follicle development in vitro. MAIN RESULTS AND THE ROLE OF CHANCE It was found that 609 out of 1401 identified proteins were common to the three follicle developmental stages investigated. Some proteins were identified uniquely at one stage: 71 of the 775 identified proteins in SF, 181 of 1092 in SMR and 192 of 1100 in AF. Additional qualitative and quantitative analysis highlighted 44 biological processes over-represented in our samples compared to the Mus musculus gene database. In particular, it was possible to identify proteins implicated in the cell cycle, calcium ion binding and glycolysis, with specific expressions and abundance, throughout in vitro follicle development. LARGE SCALE DATA Data are available via ProteomeXchange with identifier PXD006227. LIMITATIONS, REASONS FOR CAUTION The proteome analyses described in this study were performed after in vitro development. Despite fractionation of the samples before LC-MS/MS, proteomic approaches are not exhaustive, thus proteins that are not identified in a group are not necessarily absent from that group, although they are likely to be less abundant. WIDER IMPLICATIONS OF THE FINDINGS This study allowed a general view of proteins implicated in follicle development in vitro and it represents the most complete catalog of the whole follicle proteome available so far. Not only were well known proteins of the oocyte identified but also proteins that are probably expressed only in granulosa cells. STUDY FUNDING/COMPETING INTEREST(S) This study was supported by the Portuguese Foundation for Science and Technology, FCT (PhD fellowship SFRH/BD/65299/2009 to A.A.), the Swedish Childhood Cancer Foundation (PR 2014-0144 to K.A.R-.W.) and Stockholm County Council to K.A.R-.W. The authors of the study have no conflict of interest to report.
Collapse
Affiliation(s)
- Amandine Anastácio
- Université Paris VI (UPMC), Paris, France
- Department of Oncology-Pathology, Karolinska Institutet and Laboratory of Translational Fertility Preservation, Cancer Center Karolinska (CCK), Stockholm, Sweden
| | - Kenny A Rodriguez-Wallberg
- Department of Oncology-Pathology, Karolinska Institutet and Laboratory of Translational Fertility Preservation, Cancer Center Karolinska (CCK), Stockholm, Sweden
- Reproductive Medicine, Department of Gynecology and Reproduction, Karolinska University Hospital, Stockholm, Sweden
| | - Solenne Chardonnet
- Sorbonne Universités, UPMC Univ Paris 06, Inserm, UMS Omique, Plateforme P3S, Paris, France
| | - Cédric Pionneau
- Sorbonne Universités, UPMC Univ Paris 06, Inserm, UMS Omique, Plateforme P3S, Paris, France
| | | | - Teresa Almeida Santos
- Department of Human Reproduction, University Hospital of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Catherine Poirot
- Université Paris VI (UPMC), Paris, France
- Service d’Hématologie-Unité AJA, Hôpital Saint Louis, Paris, France
| |
Collapse
|
38
|
Baufeld A, Koczan D, Vanselow J. Induction of altered gene expression profiles in cultured bovine granulosa cells at high cell density. Reprod Biol Endocrinol 2017; 15:3. [PMID: 28056989 PMCID: PMC5217602 DOI: 10.1186/s12958-016-0221-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 12/13/2016] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND In previous studies it has been shown that bovine granulosa cells (GC) cultured at a high plating density dramatically change their physiological and molecular characteristics, thus resembling an early stage of luteinization. During the present study, these specific effects on the GC transcriptome were comprehensively analysed to clarify the underlying mechanisms. METHODS GC were cultured in serum free medium with FSH and IGF-1 stimulation at different initial plating density. The estradiol and progesterone production was determined by radioimmunoassays and the gene expression profiles were analysed by mRNA microarray analysis after 9 days. The data were statistically analysed and the abundance of selected, differentially expressed transcripts was re-evaluated by qPCR. Bioinformatic pathway analysis of density affected transcripts was done using Ingenuity Pathway Analysis. RESULTS The data showed that at high plating density the expression of 1510 annotated genes, represented by 1575 transcript clusters, showed highly altered expression levels. Nearly two-thirds were up- and one third down-regulated. Within the top up-regulated genes VNN2, RGS2 and PTX3 could be identified, as well as HBA or LOXL2. Down-regulated genes included important key genes of folliculogenesis like CYP19A1 and FSHR. Ingenuity pathway analysis identified "AMPK signaling" as well as "cAMP-mediated signaling" as major pathways affected by the alteration of the expression profile. Main putative upstream regulators were TGFB1 and VEGF, thus indicating a connection with cell differentiation and angiogenesis. A detailed cluster analysis revealed one single cluster that was highly associated with the upstream regulator beta-estradiol. Within this cluster key genes of steroid biosynthesis were not included, but instead, other genes importantly involved in follicular development, like OXT and VEGFA as well as the three most down-regulated genes TXNIP, PAG11 and ARRDC4 were identified. CONCLUSIONS From these data we hypothesize that high density conditions induce a stage of differentiation in cultured GC that is similar to early post-LH conditions in vivo. Furthermore we hypothesize that specific cell-cell-interactions led to this differentiation including transformations necessary to promote angiogenesis.
Collapse
Affiliation(s)
- Anja Baufeld
- Institute of Reproductive Biology, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| | - Dirk Koczan
- Institute for Immunology, University of Rostock, 18055 Rostock, Germany
| | - Jens Vanselow
- Institute of Reproductive Biology, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| |
Collapse
|
39
|
Shiratsuki S, Hara T, Munakata Y, Shirasuna K, Kuwayama T, Iwata H. Low oxygen level increases proliferation and metabolic changes in bovine granulosa cells. Mol Cell Endocrinol 2016; 437:75-85. [PMID: 27519633 DOI: 10.1016/j.mce.2016.08.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 08/06/2016] [Accepted: 08/08/2016] [Indexed: 01/04/2023]
Abstract
The present study addresses molecular backgrounds underlying low oxygen induced metabolic changes and 1.2-fold change in bovine granulosa cell (GCs) proliferation. RNA-seq revealed that low oxygen (5%) upregulated genes associated with HIF-1 and glycolysis and downregulated genes associated with mitochondrial respiration than that in high oxygen level (21%). Low oxygen level induced high glycolytic activity and low mitochondrial function and biogenesis. Low oxygen level enhanced GC proliferation with high expression levels of HIF-1, VEGF, AKT, mTOR, and S6RP, whereas addition of anti-VEGF antibody decreased cellular proliferation with low phosphorylated AKT and mTOR expression levels. Low oxygen level reduced SIRT1, whereas activation of SIRT1 by resveratrol increased mitochondrial replication and decreased cellular proliferation with reduction of phosphorylated mTOR. These results suggest that low oxygen level stimulates the HIF1-VEGF-AKT-mTOR pathway and up-regulates glycolysis, which contributes to GC proliferation, and downregulation of SIRT1 contributes to hypoxia-associated reduction of mitochondria and cellular proliferation.
Collapse
Affiliation(s)
- Shogo Shiratsuki
- Laboratory of Animal Reproduction, Department of Animal Science, Tokyo University of Agriculture, 1737 Funako, Atsugi, Kanagawa, 243-0034, Japan
| | - Tomotaka Hara
- Laboratory of Animal Reproduction, Department of Animal Science, Tokyo University of Agriculture, 1737 Funako, Atsugi, Kanagawa, 243-0034, Japan
| | - Yasuhisa Munakata
- Laboratory of Animal Reproduction, Department of Animal Science, Tokyo University of Agriculture, 1737 Funako, Atsugi, Kanagawa, 243-0034, Japan
| | - Koumei Shirasuna
- Laboratory of Animal Reproduction, Department of Animal Science, Tokyo University of Agriculture, 1737 Funako, Atsugi, Kanagawa, 243-0034, Japan
| | - Takehito Kuwayama
- Laboratory of Animal Reproduction, Department of Animal Science, Tokyo University of Agriculture, 1737 Funako, Atsugi, Kanagawa, 243-0034, Japan
| | - Hisataka Iwata
- Laboratory of Animal Reproduction, Department of Animal Science, Tokyo University of Agriculture, 1737 Funako, Atsugi, Kanagawa, 243-0034, Japan.
| |
Collapse
|
40
|
Cinco R, Digman MA, Gratton E, Luderer U. Spatial Characterization of Bioenergetics and Metabolism of Primordial to Preovulatory Follicles in Whole Ex Vivo Murine Ovary. Biol Reprod 2016; 95:129. [PMID: 27683265 PMCID: PMC5315427 DOI: 10.1095/biolreprod.116.142141] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 07/10/2016] [Accepted: 09/21/2016] [Indexed: 12/22/2022] Open
Abstract
Previous work characterizing ovarian bioenergetics has defined follicular metabolism by measuring metabolic by-products in culture media. However, culture conditions perturb the native state of the follicle, and these methods do not distinguish between metabolism occurring within oocytes or granulosa cells. We applied the phasor approach to fluorescence lifetime imaging microscopy (phasor FLIM) at 740-nm two-photon excitation to examine the spatial distribution of free and protein-bound nicotinamide adenine dinucleotide hydride (NADH) during primordial through preovulatory stages of follicular development in fresh ex vivo murine neonatal and gonadotropin stimulated prepubertal ovaries. We obtained subcellular resolution phasor FLIM images of primordial through primary follicles and quantified the free/bound NADH ratio (relative NADH/NAD+) separately for oocyte nucleus and oocyte cytoplasm. We found that dynamic changes in oocyte nucleus free/bound NADH paralleled the developmental maturation of primordial to primary follicles. Immunohistochemistry of NAD+-dependent deacetylase SIRTUIN 1 (SIRT1) in neonatal ovary revealed that increasing SIRT1 expression in oocyte nuclei was inversely related to decreasing free/bound NADH during the primordial to primary follicle transition. We characterized oocyte metabolism at these early stages to be NADH producing (glycolysis/Krebs). We extended the results of prior studies to show that cumulus and mural granulosa cell metabolism in secondary through preovulatory follicles is mainly NADH producing (glycolysis/Krebs cycle), while oocyte metabolism is mainly NADH consuming (oxidative phosphorylation). Taken together, our data characterize dynamic changes in free/bound NADH and SIRT1 expression during early follicular development and confirm results from previous studies defining antral and preovulatory follicle metabolism in culture.
Collapse
Affiliation(s)
- Rachel Cinco
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, California
| | - Michelle A Digman
- Laboratory for Fluorescence Dynamics, University of California Irvine, Irvine, California.,Department of Biomedical Engineering, University of California Irvine, Irvine, California
| | - Enrico Gratton
- Laboratory for Fluorescence Dynamics, University of California Irvine, Irvine, California.,Department of Biomedical Engineering, University of California Irvine, Irvine, California
| | - Ulrike Luderer
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, California .,Department of Medicine, University of California Irvine, Irvine, California.,Program in Public Health, University of California Irvine, Irvine, California
| |
Collapse
|
41
|
El-Hayek S, Clarke HJ. Control of Oocyte Growth and Development by Intercellular Communication Within the Follicular Niche. Results Probl Cell Differ 2016; 58:191-224. [PMID: 27300180 DOI: 10.1007/978-3-319-31973-5_8] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In the mammalian ovary, each oocyte grows and develops within its own structural and developmental niche-the follicle. Together with the female germ cell in the follicle are somatic granulosa cells, specialized companion cells that surround the oocyte and provide support to it, and an outer layer of thecal cells that serve crucial roles including steroid synthesis. These follicular compartments function as a single physiological unit whose purpose is to produce a healthy egg, which upon ovulation can be fertilized and give rise to a healthy embryo, thus enabling the female germ cell to fulfill its reproductive potential. Beginning from the initial stage of follicle formation and until terminal differentiation at ovulation, oocyte and follicle growth depend absolutely on cooperation between the different cellular compartments. This cooperation synchronizes the initiation of oocyte growth with follicle activation. During growth, it enables metabolic support for the follicle-enclosed oocyte and allows the follicle to fulfill its steroidogenic potential. Near the end of the growth period, intra-follicular interactions prevent the precocious meiotic resumption of the oocyte and ensure its nuclear differentiation. Finally, cooperation enables the events of ovulation, including meiotic maturation of the oocyte and expansion of the cumulus granulosa cells. In this chapter, we discuss the cellular interactions that enable the growing follicle to produce a healthy oocyte, focusing on the communication between the germ cell and the surrounding granulosa cells.
Collapse
Affiliation(s)
- Stephany El-Hayek
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC, Canada
- Department of Biology, McGill University, Montreal, QC, Canada
- Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Block E-M0.2218, Montreal, QC, Canada, H4A 3J1
| | - Hugh J Clarke
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC, Canada.
- Department of Biology, McGill University, Montreal, QC, Canada.
- Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Block E-M0.2218, Montreal, QC, Canada, H4A 3J1.
- Department of Experimental Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
42
|
Downs SM. Nutrient pathways regulating the nuclear maturation of mammalian oocytes. Reprod Fertil Dev 2015; 27:572-82. [DOI: 10.1071/rd14343] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 01/10/2015] [Indexed: 12/15/2022] Open
Abstract
Oocyte maturation is defined as that phase of development whereby a fully grown oocyte reinitiates meiotic maturation, completes one meiotic division with extrusion of a polar body, then arrests at MII until fertilisation. Completion of maturation depends on many different factors, not the least of which is the proper provision of energy substrates to fuel the process. Interaction of the oocyte and somatic compartment of the follicle is critical and involves numerous signals exchanged between the two cell types in both directions. One of the prominent functions of the cumulus cells is the channelling of metabolites and nutrients to the oocyte to help stimulate germinal vesicle breakdown and direct development to MII. This entails the careful integration and coordination of numerous metabolic pathways, as well as oocyte paracrine signals that direct certain aspects of cumulus cell metabolism. These forces collaborate to produce a mature oocyte that, along with accompanying physiological changes called cytoplasmic maturation, which impart subsequent developmental competence to the oocyte, can be fertilised and develop to term. This review focuses on nuclear maturation and the metabolic interplay that regulates it, with special emphasis on data generated in the mouse.
Collapse
|
43
|
Maruthini D, Harris SE, Barth JH, Balen AH, Campbell BK, Picton HM. The effect of metformin treatment in vivo on acute and long-term energy metabolism and progesterone production in vitro by granulosa cells from women with polycystic ovary syndrome. Hum Reprod 2014; 29:2302-16. [PMID: 25139174 PMCID: PMC4164147 DOI: 10.1093/humrep/deu187] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
STUDY QUESTION What are the consequences of polycystic ovary syndrome (PCOS) pathology and metformin-pretreatment in vivo in women with PCOS on the metabolism and steroid production of follicular phenotype- and long-term cultured-granulosa cells (GC)? SUMMARY ANSWER PCOS pathology significantly compromised glucose metabolism and the progesterone synthetic capacity of follicular- and long-term cultured-GCs and the metabolic impact of PCOS on GC function was alleviated by metformin-pretreatment in vivo. WHAT IS KNOWN ALREADY Granulosa cells from women with PCOS have been shown to have an impaired insulin-stimulated glucose uptake and lactate production in vitro. However, these results were obtained by placing GCs in unphysiological conditions in culture medium containing high glucose and insulin concentrations. Moreover, existing data on insulin-responsive steroid production in vitro by PCOS GCs vary. STUDY DESIGN, SIZE AND DURATION Case-control experimental research comparing glucose uptake, pyruvate and lactate production and progesterone production in vitro by GCs from three aetiological groups, all undergoing IVF; healthy control women (Control, n = 12), women with PCOS treated with metformin in vivo (Metformin, n = 8) and women with PCOS not exposed to metformin (PCOS, n = 8). The study was conducted over a period of 3 years between 2007 and 2010. PARTICIPANTS/MATERIALS, SETTING, METHODS Rotterdam criteria were used for the diagnosis of PCOS; all subjects were matched for age, BMI and baseline FSH. Individual patient cultures were undertaken with cells incubated in a validated, physiological, serum-free culture medium containing doses of 0–6 mM glucose and 0–100 ng/ml insulin for 6 h and 144 h to quantify the impact of treatments on acute and long-term metabolism, respectively, and progesterone production. The metabolite content of spent media was measured using spectrophotometric plate reader assay. The progesterone content of spent media was measured by enzyme-linked immunosorbent assay. Viable GC number was quantified after 144 h of culture by the vital dye Neutral Red uptake assay. MAIN RESULTS AND THE ROLE OF CHANCE Granulosa cells from women with PCOS pathology revealed reduced pyruvate production and preferential lactate production in addition to their reduced glucose uptake during cultures (P < 0.05). Metformin pretreatment alleviated this metabolic lesion (P < 0.05) and enhanced cell proliferation in vitro (P < 0.05), but cells retained a significantly reduced capacity for progesterone synthesis compared with controls (P < 0.05). LIMITATIONS, REASONS FOR CAUTION Although significant treatment effects were detected in this small cohort, further studies are required to underpin the molecular mechanisms of the effect of metformin on GCs. WIDER IMPLICATIONS OF THE FINDINGS The individual patient culture strategy combined with multifactorial experimental design strengthens the biological interpretation of the data. Collectively, these results support the notion that there is an inherent impairment in progesterone biosynthetic capacity of the GCs from women with PCOS. The positive, acute metabolic effect and the negative long-term steroidogenic effect on GCs following metformin exposure in vivo may have important implications for follicular development and luteinized GC function when the drug is used in clinical practice. STUDY FUNDING/COMPETING INTEREST(S) No competing interests. This work was supported by the UK Medical Research Council Grant Reference number G0800250.
Collapse
Affiliation(s)
- D Maruthini
- The Leeds Centre for Reproductive Medicine, Seacroft Hospital, York Road, Leeds LS14 6UH, UK
| | - S E Harris
- Division of Reproduction and Early Development, Leeds Institute for Genetics, Health and Therapeutics, University of Leeds, Clarendon Way, Leeds LS2 9JT, UK
| | - J H Barth
- Department of Clinical Biochemistry, Leeds Teaching Hospitals NHS Trust, Leeds General Infirmary, Great George Street, Leeds LS1 3EX, UK
| | - A H Balen
- The Leeds Centre for Reproductive Medicine, Seacroft Hospital, York Road, Leeds LS14 6UH, UK
| | - B K Campbell
- Division of Human Development at Nottingham University Hospitals NHS Trust, Department of Obstetrics and Gynaecology, Queen's Medical Centre Campus, Nottingham NG7 2UH, UK
| | - H M Picton
- Division of Reproduction and Early Development, Leeds Institute for Genetics, Health and Therapeutics, University of Leeds, Clarendon Way, Leeds LS2 9JT, UK
| |
Collapse
|
44
|
Zeng HT, Richani D, Sutton-McDowall ML, Ren Z, Smitz JEJ, Stokes Y, Gilchrist RB, Thompson JG. Prematuration with cyclic adenosine monophosphate modulators alters cumulus cell and oocyte metabolism and enhances developmental competence of in vitro-matured mouse oocytes. Biol Reprod 2014; 91:47. [PMID: 24966394 DOI: 10.1095/biolreprod.114.118471] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Oocyte in vitro maturation (IVM) is an important assisted reproductive technology and research tool. The adoption of IVM into routine clinical practice has been hindered by its significantly lower success rates compared to conventional in vitro fertilization. Cyclic AMP (cAMP) modulation and follicle-stimulating hormone (FSH), independently, have long been known to improve IVM oocyte developmental competence. This study comprehensively examined the effects of FSH and cAMP/cGMP modulation, alone and in combination, on IVM oocyte metabolism and developmental outcomes. Mouse cumulus-oocyte complexes (COCs) were subjected to a 1 h prematuration phase ± the cAMP modulator forskolin and cAMP/cGMP modulator 3-isobutyl-1-methylxanthine followed by IVM ± FSH. Prematuration with these cyclic nucleotide modulators or IVM with FSH significantly improved oocyte developmental competence and reduced spindle abnormalities compared to spontaneous IVM (no treatment); however, these two treatments in combination endowed even greater developmental competence (improved subsequent blastocyst rates and quality; P < 0.05), albeit blastocyst yield and quality remained significantly lower than that of oocytes matured in vivo. A significant additive effect of combined IVM treatments was evident as increased COC lactate production and oxygen consumption and enhanced oocyte oxidative metabolism, ATP production, ATP:ADP ratio, and glutathione levels (P < 0.05). Nevertheless, IVM increased reactive oxygen species production, particularly as a consequence of FSH addition, relative to in vivo matured oocytes. In conclusion, improvements in the embryo yield following IVM is associated with increased COC oxygen consumption and oocyte oxidative metabolism, but these remain metabolically and developmentally less competent relative to in vivo derived oocytes.
Collapse
Affiliation(s)
- Hai-Tao Zeng
- Research Centre for Reproductive Health, Robinson Institute, and Discipline of Obstetrics and Gynaecology, School of Paediatrics and Reproductive Health, The University of Adelaide, Adelaide, South Australia, Australia Center for Reproductive Medicine, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Dulama Richani
- Research Centre for Reproductive Health, Robinson Institute, and Discipline of Obstetrics and Gynaecology, School of Paediatrics and Reproductive Health, The University of Adelaide, Adelaide, South Australia, Australia
| | - Melanie L Sutton-McDowall
- Research Centre for Reproductive Health, Robinson Institute, and Discipline of Obstetrics and Gynaecology, School of Paediatrics and Reproductive Health, The University of Adelaide, Adelaide, South Australia, Australia
| | - Zi Ren
- Research Centre for Reproductive Health, Robinson Institute, and Discipline of Obstetrics and Gynaecology, School of Paediatrics and Reproductive Health, The University of Adelaide, Adelaide, South Australia, Australia Center for Reproductive Medicine, General Hospital of Guangdong, Guangzhou, People's Republic of China
| | - Johan E J Smitz
- Follicle Biology Laboratory, Center for Reproductive Medicine and Medical School, Free University Brussels (VUB), Brussels, Belgium
| | - Yvonne Stokes
- School of Mathematical Science, The University of Adelaide, Adelaide, South Australia, Australia
| | - Robert B Gilchrist
- Research Centre for Reproductive Health, Robinson Institute, and Discipline of Obstetrics and Gynaecology, School of Paediatrics and Reproductive Health, The University of Adelaide, Adelaide, South Australia, Australia
| | - Jeremy G Thompson
- Research Centre for Reproductive Health, Robinson Institute, and Discipline of Obstetrics and Gynaecology, School of Paediatrics and Reproductive Health, The University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
45
|
Makanji Y, Tagler D, Pahnke J, Shea LD, Woodruff TK. Hypoxia-mediated carbohydrate metabolism and transport promote early-stage murine follicle growth and survival. Am J Physiol Endocrinol Metab 2014; 306:E893-903. [PMID: 24569591 PMCID: PMC3989738 DOI: 10.1152/ajpendo.00484.2013] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Oxygen tension is critical for follicle growth and metabolism, especially for early-stage follicles, where vascularity is limited. Its role and underlying mechanism in the in vitro activation and maturation of immature to ovulatory follicles is largely unknown. In this study, early secondary (110 μm) murine follicles were isolated and encapsulated in alginate hydrogels to replicate the in vivo environment of the growing/maturing follicle. Encapsulated follicles were cultured for 8 days at either 2.5 or 20% O2. Survival (2.6-fold) and growth (1.2-fold) were significantly higher for follicles cultured at 2.5% compared with 20% O2. Using a mouse hypoxia-signaling pathway qRT-PCR array and GeneGo Metacore analysis, we found that direct target genes of the hypoxia-activated HIF1-complex were significantly upregulated in follicles cultured for 8 days at 2.5% compared with 20% O2, including the carbohydrate transport and metabolism genes Slc2a3, Vegfa, Slc2a1, Edn1, Pgk1, Ldha, and Hmox1. Other upregulated genes included carbohydrate transporters (Slc2a1, Slc2a3, and Slc16a3) and enzymes essential for glycolysis (Pgk1, Hmox1, Hk2, Gpi1, Pfkl, Pfkp, Aldoa, Gapdh, Pgam1, Eno1, Pkm2, and Ldha). For follicles cultured at 2.5% O2, a 7.2-fold upregulation of Vegfa correlated to an 18-fold increase in VEGFA levels, and a 3.2-fold upregulation of Ldha correlated to a 4.8-fold increase in lactate levels. Both VEGFA and lactate levels were significantly higher in follicles cultured at 2.5% compared with 20% O2. Therefore, enhanced hypoxia-mediated glycolysis is essential for growth and survival of early secondary follicles and provides vital insights into improving in vitro culture conditions.
Collapse
Affiliation(s)
- Yogeshwar Makanji
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, and
| | | | | | | | | |
Collapse
|
46
|
Dalton CM, Szabadkai G, Carroll J. Measurement of ATP in single oocytes: impact of maturation and cumulus cells on levels and consumption. J Cell Physiol 2014; 229:353-61. [PMID: 24002908 DOI: 10.1002/jcp.24457] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 08/16/2013] [Indexed: 11/07/2022]
Abstract
Mitochondria provide the primary source of ATP in the oocyte and early embryo and mitochondrial dysfunction and deficit of mitochondria-derived ATP has been linked to suboptimal developmental competence. We have undertaken a study of ATP in the maturing mouse oocyte using a novel recombinant FRET based probe, AT1.03. We show that AT1.03 can be successfully used to monitor cytosolic ATP levels in single live oocytes over extended time periods. We find that ATP levels undergo dynamic changes associated with specific maturational events and that oocytes display altered rates of ATP consumption at different stages of maturation. Cumulus enclosed oocytes have a higher ATP level during maturation than denuded oocytes and this can be abolished by inhibition of gap junctional communication between the oocyte and cumulus cells. Our work uses a new approach to shed light on regulation of ATP levels and ATP consumption during oocyte maturation.
Collapse
Affiliation(s)
- Caroline M Dalton
- Division of Biosciences, Department of Cell and Developmental Biology, UCL, London, UK
| | | | | |
Collapse
|
47
|
Kang B, Jiang DM, Bai L, He H, Ma R. Molecular characterisation and expression profiling of the ENO1 gene in the ovarian follicle of the Sichuan white goose. Mol Biol Rep 2014; 41:1927-35. [PMID: 24413993 DOI: 10.1007/s11033-014-3039-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 01/04/2014] [Indexed: 10/25/2022]
Abstract
The ENO1 gene encodes a multifunctional enzyme that has been identified as a key component of the glycolytic pathway. Our previous studies demonstrated that ENO1 gene expression was higher in the ovaries of laying geese compared with prelaying geese. However, the molecular characterisation and expression profiling of the ENO1 gene in geese tissues and ovarian follicles remain to be determined. In this study, ENO1 cDNA (1,445 bp long) of the Sichuan white goose was cloned and characterised. The ORF of ENO1 cDNA is 1,305 bp in length and encodes a 434 amino acid protein with a molecular weight of 47.27 kDa. ENO1 expression in all of the examined tissues was the highest in spleen and the lowest in breast muscle. High expression of ENO1 appeared in the kidney, liver, adrenal gland, and retina. With increasing follicle growth, ENO1 gene expression began to decrease from the small white follicle to F5, which was followed by a sharp increase in expression in F4 and then a gradual decrease in expression from F3 to F1. Furthermore, in the postovulatory follicles (POF), the levels of ENO1 gene expression decreased gradually from POF1 to POF4. In conclusion, the ENO1 transcript was widely distributed in various tissues of the Sichuan white goose, but ENO1 expression was tissue-specific. Furthermore, the results of the ENO1 expression profiling of ovarian follicles suggest that ENO1 may play an important dual role in the progress of follicular development, where ENO1 acts as a glycolytic enzyme and also mediates apoptosis.
Collapse
Affiliation(s)
- Bo Kang
- College of Animal Science and Technology, Sichuan Agricultural University, Ya'an, 625014, Sichuan, China
| | | | | | | | | |
Collapse
|
48
|
Gook DA, Edgar DH, Lewis K, Sheedy JR, Gardner DK. Impact of oxygen concentration on adult murine pre-antral follicle development in vitro and the corresponding metabolic profile. Mol Hum Reprod 2013; 20:31-41. [PMID: 24013158 DOI: 10.1093/molehr/gat062] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Oxygen concentration during in vitro culture has a significant effect on the physiology of embryos, altering metabolic profile and developmental outcome. Although atmospheric oxygen has been used routinely for the culture of ovarian follicles, oxygen concentration may also be critical for follicle growth but the optimal concentration has not been determined. In this study, mechanically isolated primary and secondary follicles (80-140 µm diameter) from adult mouse ovaries were cultured in serum-free conditions for 8 days in either 5 or 20% oxygen to determine growth (follicular diameter), morphology and viability. For each oxygen concentration, half of the medium was replaced on Days 2, 4 and 6 or on Day 4 only. In the latter group, metabolic analysis of spent follicular culture media was performed by (1)H-NMR. The proportion of viable, growing follicles was significantly (P < 0.0001) higher in 5% than in 20% oxygen (59% versus 8%). Reducing the frequency of medium replacement during culture in 5% oxygen resulted in significantly (P < 0.001) more viable follicles (79 versus 46%). In 20% oxygen, poor follicular viability was observed irrespective of the frequency of medium replacement (8 and 10% respectively). Metabolic profiles showed marked differences in amino acid and carbohydrate utilization with respect to both oxygen concentration and between Days 4 and 8 of development. Metabolites which significantly discriminated between oxygen concentration at both time points were glucose consumption, lactate utilization, alanine, alanyl-glutamine, leucine and proline. In conclusion, the poor in vitro follicular development previously observed in minimal culture conditions may reflect the use of 20% oxygen. Frequent medium replenishment is not necessary and does not overcome the detrimental effect of high oxygen on follicle viability. Further optimization of culture conditions would benefit from metabolic analyses and the use of 5% oxygen should be tested further for impact on functional aspects of follicle culture such as steroid production which is currently unknown.
Collapse
Affiliation(s)
- Debra A Gook
- Reproductive Services, Royal Women's Hospital, Grattan Street, Parkville, VIC 3052, Australia
| | | | | | | | | |
Collapse
|
49
|
Martínez F, Devesa M, Coroleu B, Tur R, González C, Boada M, Solé M, Veiga A, Barri PN. Cancer and fertility preservation: Barcelona consensus meeting. Gynecol Endocrinol 2013; 29:285-91. [PMID: 23347067 DOI: 10.3109/09513590.2012.743019] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Improvements in early diagnosis and treatment strategies in cancer patients have enabled younger women with cancer to survive. In addition to the stressful event of the diagnosis, patients with malignant diseases face the potential loss of the opportunity to have children. Preservation of fertility has become a challenging issue and it is still surrounded by controversies. On the basis of available evidence, a group of experts reached a consensus regarding the options for trying to preserve fertility in women with cancer: among established methods, in postpubertal women, oocyte cryopreservation is the preferred option, whereas ovarian tissue cryopreservation is the only possibility for prepubertal girls. Combining several strategies on an individual basis may improve the chances of success. Realistic information should be provided before any intervention is initiated. Counseling should offer support for patients and provide better care by understanding emotional needs, psychological predictors of distress and methods of coping. Early referral to the fertility specialist is essential as fertility preservation (FP) may improve quality of life in these patients. The information summarized here is intended to help specialists involved in the treatment of cancer and reproductive medicine to improve their understanding of procedures available for FP in young cancer patients.
Collapse
Affiliation(s)
- Francisca Martínez
- Departamento de Obstetricia, Ginecología y Medicina de Reproducción, Institut Universitari Dexeus, Universitat Autònoma de Barcelona, Barcelona, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Cotterill M, Harris SE, Collado Fernandez E, Lu J, Huntriss JD, Campbell BK, Picton HM. The activity and copy number of mitochondrial DNA in ovine oocytes throughout oogenesis in vivo and during oocyte maturation in vitro. Mol Hum Reprod 2013; 19:444-50. [PMID: 23468533 PMCID: PMC3690804 DOI: 10.1093/molehr/gat013] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Mitochondria are responsible for the production of ATP, which drives cellular metabolic and biosynthetic processes. This is the first study to quantify the mtDNA copy number across all stages of oogenesis in a large monovulatory species, it includes assessment of the activity of mitochondria in germinal vesicle (GV) and metaphase II (MII) oocytes through JC1 staining. Primordial to early antral follicles (n = 249) were isolated from the sheep ovarian cortex following digestion at 37°C for 1 h and all oocytes were disaggregated from their somatic cells. Germinal vesicle oocytes (n = 133) were aspirated from 3- to 5-mm diameter antral follicles, and mature MII oocytes (n = 71) were generated following in vitro maturation (IVM). The mtDNA copy number in each oocyte was quantified using real-time PCR and showed a progressive, but variable increase in the amount of mtDNA in oocytes from primordial follicles (605 ± 205, n = 8) to mature MII oocytes (744 633 ± 115 799, n = 13; P < 0.05). Mitochondrial activity (P > 0.05) was not altered during meiotic progression from GV to MII during IVM. The observed increase in the mtDNA copy number across oogenesis reflects the changing ATP demands needed to orchestrate cytoskeletal and cytoplasmic reorganization during oocyte growth and maturation and the need to fuel the resumption of meiosis in mature oocytes following the pre-ovulatory gonadotrophin surge.
Collapse
Affiliation(s)
- Matthew Cotterill
- Division of Reproduction and Early Development, Leeds Institute of Genetics Health and Therapeutics, University of Leeds, Clarendon Way, Leeds LS2 9JT, UK.
| | | | | | | | | | | | | |
Collapse
|