1
|
Ndandala CB, Guo Y, Ju Z, Fachri M, Mwemi HM, Chen H. Integrated lncRNA and mRNA Transcriptome Analyses of IGF1 and IGF2 Stimulated Ovaries Reveal Genes and Pathways Potentially Associated with Ovarian Development and Oocyte Maturation in Golden Pompano ( Trachinotus ovatus). Animals (Basel) 2025; 15:1134. [PMID: 40281968 PMCID: PMC12024298 DOI: 10.3390/ani15081134] [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: 03/12/2025] [Revised: 04/09/2025] [Accepted: 04/12/2025] [Indexed: 04/29/2025] Open
Abstract
Insulin-like growth factors (IGFs) play crucial roles in the regulation of animal growth and reproduction. However, the functional and regulatory mechanisms underlying ovarian growth and oocyte maturation in teleosts remain unclear. In this study, the expression profiles of lncRNAs and mRNAs were analyzed in the ovaries of golden pompano (Trachinotus ovatus) treated with IGF1 and IGF2 proteins to gain insights into the role of these two IGF ligands in the regulation of ovarian development and maturation. A total of 1494 lncRNAs and 8728 mRNAs were differentially expressed following IGF1 treatment compared with the control group. A total of 101 lncRNAs and 377 mRNAs were differentially expressed after IGF2 treatment compared to those in the control group. The results revealed that KEGG pathways enriched by target genes of the DE lncRNAs overlapped significantly with those enriched by the DE mRNAs in both the IGF1 and IGF2 groups. The key overlapping enriched pathways included ECM receptor interaction, gap junction, Hedgehog signaling pathway, Ras signaling pathway, Rap1 signaling pathway, TGF beta signaling pathway, Wnt signaling pathway, GnRH signaling pathway, progesterone-mediated oocyte maturation, oocyte meiosis, cell cycle, and MAPK signaling pathway. The differentially expressed genes (DEGs) involved in ovarian development and oocyte maturation were cyp17a1, cyp19a1, star, hsd17b3, hsd17b7, adam23, slc26a6, htr2b, h2ax, nanos3, krt18, pgr, and inhbb, following IGF1 and IGF2 treatment. Furthermore, four lncRNAs (MSTRG.66521.1, MSTRG.49969.1, MSTRG.59923.1, and MSTRG.13767.1) for IGF1 and two (MSTRG.20896.2 and MSTRG.58123.2) for IGF2 within the lncRNA-mRNA network were found to target DEGs related to ovarian development and maturation. This suggests that IGFs may affect reproductive processes by regulating the expression of lncRNAs and mRNAs. RT-qPCR analysis revealed that these six lncRNAs showed high expression levels in the brain, pituitary, liver, and gonad tissues, indicating their potential involvement in regulating ovarian growth and development. This study elucidates the lncRNA-mRNA regulatory mechanism in response to IGF1 and IGF2 treatment during stage III of ovarian development in golden pompano, thereby deepening our understanding of its functional role.
Collapse
Affiliation(s)
- Charles Brighton Ndandala
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; (C.B.N.); (Y.G.); (Z.J.); (M.F.); (H.M.M.)
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang 524025, China
| | - Yuwen Guo
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; (C.B.N.); (Y.G.); (Z.J.); (M.F.); (H.M.M.)
| | - Zhimin Ju
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; (C.B.N.); (Y.G.); (Z.J.); (M.F.); (H.M.M.)
| | - Muhammad Fachri
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; (C.B.N.); (Y.G.); (Z.J.); (M.F.); (H.M.M.)
| | - Happiness Moses Mwemi
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; (C.B.N.); (Y.G.); (Z.J.); (M.F.); (H.M.M.)
| | - Huapu Chen
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; (C.B.N.); (Y.G.); (Z.J.); (M.F.); (H.M.M.)
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang 524025, China
| |
Collapse
|
2
|
Fattahi A, Zarezadeh R, Rastgar Rezaei Y, Mettler L, Nouri M, Schmutzler AG, Salmassi A. Expression of interleukin-1β and its receptor in human granulosa cells and their association with steroidogenesis. Tissue Cell 2023; 85:102230. [PMID: 37820554 DOI: 10.1016/j.tice.2023.102230] [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: 03/27/2023] [Revised: 09/28/2023] [Accepted: 09/28/2023] [Indexed: 10/13/2023]
Abstract
This study aimed to investigate whether interleukin 1β (IL-1β) and soluble IL-1 receptor 2 (sIL-1R2) are expressed in human granulosa cells (GCs) and relate to ovarian steroidogenesis. Ninety-six women undergoing in vitro fertilization (IVF) were recruited. RT-PCR and immunocytochemistry were used to detect mRNAs and proteins of IL-1β and IL-1R2, respectively. The steroidogenesis of primary cultured GCs was evaluated following treatment with either IL-1β alone or IL-1β and FSH in combination. There were positive correlations between serum IL-1β and serum progesterone (r = 0.220, p = 0.032) and follicular fluid (FF) estradiol (r = 0.242, p = 0.018). Additionally, serum and FF sIL-1R2 were negatively and positively correlated with FF estradiol (r = -0.376, p = 0.005) and FF progesterone (r = 0.434, p = 0.001), respectively. The mRNA and protein expression of IL-1β and IL-1R2 became evident in GCs. IL-1β alone significantly increased estradiol secretion from GCs, but in the presence of FSH, it could notably promote progesterone secretion in addition to estradiol. In conclusion, IL-1β and sIL-1R2 are expressed in human GCs and substantially contribute to ovarian steroidogenesis, suggesting that the IL-1β system may be a potential target for optimizing ovarian hyperstimulation and steroidogenesis in IVF cycles.
Collapse
Affiliation(s)
- Amir Fattahi
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Zarezadeh
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yeganeh Rastgar Rezaei
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Liselotte Mettler
- Department of Gynecology & Obstetrics, University Hospitals Schleswig-Holstein, Campus Kiel, Germany
| | - Mohammad Nouri
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Andreas G Schmutzler
- Department of Gynecology & Obstetrics, University Hospitals Schleswig-Holstein, Campus Kiel, Germany.
| | - Ali Salmassi
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Gynecology & Obstetrics, University Hospitals Schleswig-Holstein, Campus Kiel, Germany.
| |
Collapse
|
3
|
Martínez-Moro Á, González-Brusi L, Querejeta-Fernández A, Padilla-Ruiz E, García-Blanco J, Bermejo-Álvarez P. Metabolomics analysis of human cumulus cells obtained from cumulus-oocyte complexes with different developmental potential. Hum Reprod 2023; 38:2187-2195. [PMID: 37697661 PMCID: PMC10628504 DOI: 10.1093/humrep/dead181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 08/11/2023] [Indexed: 09/13/2023] Open
Abstract
STUDY QUESTION Is the abundance of certain biochemical compounds in human cumulus cells (CCs) related to oocyte quality? SUMMARY ANSWER Malonate, 5-oxyproline, and erythronate were positively associated with pregnancy potential. WHAT IS KNOWN ALREADY CCs are removed and discarded prior to ICSI, thereby constituting an interesting biological material on which to perform molecular analysis aimed to predict oocyte developmental competence. Mitochondrial DNA content and transcriptional analyses in CC have been shown to provide a poor predictive value of oocyte competence, but the untargeted analysis of biochemical compounds (metabolomics) has been unexplored. STUDY DESIGN, SIZE, DURATION CCs were obtained from three groups of cumulus-oocyte complexes (COCs) of known developmental potential: oocytes not developing to blastocyst following ICSI (Bl-); oocytes developing to blastocyst but failing to establish pregnancy following embryo transfer (P-); and oocytes developing to blastocyst able to establish a pregnancy (P+). Metabolomics analyses were performed on 12 samples per group, each sample comprising the CC recovered from a single COC. PARTICIPANTS/MATERIALS, SETTING, METHODS Human CC samples were obtained from IVF treatments. Only unfrozen oocytes and embryos not submitted to preimplantation genetic testing were included in the analysis. Metabolomics analysis was performed by ultra-high performance liquid chromatography-tandem mass spectroscopy. MAIN RESULTS AND THE ROLE OF CHANCE The analysis identified 98 compounds, five of which were differentially abundant (P < 0.05) between groups: asparagine, proline, and malonate were less abundant in P- compared to Bl-, malonate and 5-oxoproline were less abundant in P- group compared to P+, and erythronate was less abundant in Bl- group compared to P+. No significant association between the abundance of the compounds identified and donor age or BMI was noted. LIMITATIONS, REASONS FOR CAUTION Data dispersion and the lack of coherence between developmental groups preclude the direct use of metabolic markers in clinical practice, where the uterine environment plays a major role in pregnancy outcome. The abundance of other compounds not detected by the analysis may be associated with oocyte competence. As donors were lean (only two with BMI > 30 kg/m2) and young (<34 years old), a possible effect of obesity or advanced age on the CC metabolome could not be determined. WIDER IMPLICATIONS OF THE FINDINGS The abundance of malonate, 5-oxyproline, and erythronate in CC was significantly higher in COCs ultimately establishing pregnancy, providing clues on the pathways required for oocyte competence. The untargeted analysis uncovered the presence of compounds that were not expected in CC, such as β-citrylglutamate and the neurotransmitter N-acetyl-aspartyl-glutamate, which may play roles in chromatin remodeling and signaling, respectively. STUDY FUNDING/COMPETING INTEREST(S) Research was supported by the Industrial Doctorate Project IND2017/BIO-7748 funded by Madrid Region Government. The authors declare no competing interest. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Álvaro Martínez-Moro
- Animal Reproduction Department, INIA, CSIC, Madrid, Spain
- IVF Spain, Madrid, Spain
| | | | - Ana Querejeta-Fernández
- Departamento de Química Inorgánica, Facultad de Ciencias Químicas, Universidad Complutense, Madrid, Spain
| | | | | | | |
Collapse
|
4
|
Wagner S, Whiteley SL, Castelli M, Patel HR, Deveson IW, Blackburn J, Holleley CE, Marshall Graves JA, Georges A. Gene expression of male pathway genes sox9 and amh during early sex differentiation in a reptile departs from the classical amniote model. BMC Genomics 2023; 24:243. [PMID: 37147622 PMCID: PMC10163765 DOI: 10.1186/s12864-023-09334-0] [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: 06/20/2022] [Accepted: 04/25/2023] [Indexed: 05/07/2023] Open
Abstract
BACKGROUND Sex determination is the process whereby the bipotential embryonic gonads become committed to differentiate into testes or ovaries. In genetic sex determination (GSD), the sex determining trigger is encoded by a gene on the sex chromosomes, which activates a network of downstream genes; in mammals these include SOX9, AMH and DMRT1 in the male pathway, and FOXL2 in the female pathway. Although mammalian and avian GSD systems have been well studied, few data are available for reptilian GSD systems. RESULTS We conducted an unbiased transcriptome-wide analysis of gonad development throughout differentiation in central bearded dragon (Pogona vitticeps) embryos with GSD. We found that sex differentiation of transcriptomic profiles occurs at a very early stage, before the gonad consolidates as a body distinct from the gonad-kidney complex. The male pathway genes dmrt1 and amh and the female pathway gene foxl2 play a key role in early sex differentiation in P. vitticeps, but the central player of the mammalian male trajectory, sox9, is not differentially expressed in P. vitticeps at the bipotential stage. The most striking difference from GSD systems of other amniotes is the high expression of the male pathway genes amh and sox9 in female gonads during development. We propose that a default male trajectory progresses if not repressed by a W-linked dominant gene that tips the balance of gene expression towards the female trajectory. Further, weighted gene expression correlation network analysis revealed novel candidates for male and female sex differentiation. CONCLUSION Our data reveal that interpretation of putative mechanisms of GSD in reptiles cannot solely depend on lessons drawn from mammals.
Collapse
Affiliation(s)
- Susan Wagner
- Institute for Applied Ecology, University of Canberra, Bruce, ACT, Australia
| | - Sarah L Whiteley
- Institute for Applied Ecology, University of Canberra, Bruce, ACT, Australia
- Australian National Wildlife Collection CSIRO, National Research Collections Australia, Crace, ACT, Australia
| | - Meghan Castelli
- Institute for Applied Ecology, University of Canberra, Bruce, ACT, Australia
- Australian National Wildlife Collection CSIRO, National Research Collections Australia, Crace, ACT, Australia
| | - Hardip R Patel
- Genome Sciences Department. John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Ira W Deveson
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- Faculty of Medicine, St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - James Blackburn
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- Faculty of Medicine, St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Clare E Holleley
- Institute for Applied Ecology, University of Canberra, Bruce, ACT, Australia
- Australian National Wildlife Collection CSIRO, National Research Collections Australia, Crace, ACT, Australia
| | - Jennifer A Marshall Graves
- Institute for Applied Ecology, University of Canberra, Bruce, ACT, Australia
- School of Life Sciences, La Trobe University, Bundoora, VIC, Australia
| | - Arthur Georges
- Institute for Applied Ecology, University of Canberra, Bruce, ACT, Australia.
| |
Collapse
|
5
|
Wei W, He J, Yaqoob MA, Gui L, Ren J, Li J, Li M. Integrated mRNA and miRNA Expression Profile Analysis of Female and Male Gonads in Acrossocheilus fasciatus. BIOLOGY 2022; 11:1296. [PMID: 36138775 PMCID: PMC9495813 DOI: 10.3390/biology11091296] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/18/2022] [Accepted: 08/26/2022] [Indexed: 06/16/2023]
Abstract
MicroRNAs (miRNAs) are regarded as key regulators in gonadal development and sex determination in diverse organisms. However, the functions of miRNAs in gonads of Acrossocheilus fasciatus, an economically important freshwater species in the south of China, are still unclear. Here, high-throughput sequencing was performed to investigate the mRNA and miRNAs on gonads of A. fasciatus. In total, 49,447 unigenes were obtained, including 11,635 differentially expressed genes (DEGs), among which 4147 upregulated genes and 7488 downregulated genes in the testis compared to the ovary, while 300 (237 known, and 63 novel) miRNAs with 36 differentially expressed miRNAs (DEMs) were identified, from which 17 upregulated and 19 downregulated DEMs. GO and KEGG enrichment analysis were performed to analyze the potential biological functions of DEGs and DEMs. Using qRT-PCR, 9 sex-related genes and 9 miRNAs were selected to verify the sequencing data. By dual-luciferase reporter assay, miR-22a-5p and miR-22b-5p interaction with piwil1, and miR-10d-5p interaction with piwil2 were identified. These findings could provide a reference for miRNA-regulated sex control of A. fasciatus and may reveal new insights into aquaculture and breeding concepts.
Collapse
Affiliation(s)
- Wenbo Wei
- Key Laboratory of Integrated Rice-Fish Farming, Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai 201306, China
| | - Jiamei He
- Key Laboratory of Integrated Rice-Fish Farming, Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai 201306, China
| | - Muhammad Amjad Yaqoob
- Key Laboratory of Integrated Rice-Fish Farming, Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai 201306, China
| | - Lang Gui
- Key Laboratory of Integrated Rice-Fish Farming, Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai 201306, China
| | - Jianfeng Ren
- Key Laboratory of Freshwater Aquatic Genetic Resources Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai 201306, China
| | - Jiale Li
- Key Laboratory of Freshwater Aquatic Genetic Resources Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai 201306, China
| | - Mingyou Li
- Key Laboratory of Integrated Rice-Fish Farming, Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai 201306, China
| |
Collapse
|
6
|
Di Renzo L, De Lorenzo A, Fontanari M, Gualtieri P, Monsignore D, Schifano G, Alfano V, Marchetti M. Immunonutrients involved in the regulation of the inflammatory and oxidative processes: implication for gamete competence. J Assist Reprod Genet 2022; 39:817-846. [PMID: 35353297 PMCID: PMC9050992 DOI: 10.1007/s10815-022-02472-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 03/16/2022] [Indexed: 11/30/2022] Open
Abstract
Purpose The purpose of this umbrella review is to bring together the most recent reviews concerning the role of immunonutrients for male and female infertility. Methods Regarding immunonutrients and fertility, the authors have analyzed reviews, systematic reviews, and meta-analyses published between 2011 and June 2021. All reviews on animal or in vitro studies were excluded. Relevant keywords to term micronutrients were analyzed alone or in association with other terms such as “gamete competence,” “male OR female fertility,” “male OR female infertility,” “fertile, “folliculogenesis,” “spermatogenesis,” “immunomodulation,” “immune system,” “oxidative stress.” Results The primary research has included 108 results, and after screening by title, abstract. and not topic-related, 41 studies have been included by full texts. The results show the molecular mechanisms and the immunonutrients related impact on gamete formation, development. and competence. In particular, this review focused on arginine, glutamine, vitamin C, vitamin D, vitamin E, omega-3, selenium, and zinc. Conclusions Inflammation and oxidative stress significantly impact human reproduction. For this reason, immunonutrients may play an important role in the treatment of infertile patients. However, due to the lack of consistent clinical trials, their application is limited. Therefore, the development of clinical trials is necessary to define the correct supplementation, in case of deficiency.
Collapse
Affiliation(s)
- Laura Di Renzo
- Section of Clinical Nutrition and Nutrigenomic, Department of Biomedicine and Prevention, University of Tor Vergata, Via Montpellier 1, 00133, Rome, Italy.,School of Specialization in Food Sciences, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy.,Italian University Network for Sustainable Development (RUS), Food Working Group, University of Tor Vergata, Via Cracovia, 00133, Rome, Italy
| | - Antonino De Lorenzo
- Section of Clinical Nutrition and Nutrigenomic, Department of Biomedicine and Prevention, University of Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
| | - Marco Fontanari
- School of Specialization in Food Sciences, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
| | - Paola Gualtieri
- Section of Clinical Nutrition and Nutrigenomic, Department of Biomedicine and Prevention, University of Tor Vergata, Via Montpellier 1, 00133, Rome, Italy.
| | - Diego Monsignore
- School of Specialization in Food Sciences, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
| | - Giulia Schifano
- School of Specialization in Food Sciences, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
| | - Valentina Alfano
- School of Specialization in Food Sciences, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
| | - Marco Marchetti
- PhD School of Applied Medical-Surgical Sciences, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
| | | |
Collapse
|
7
|
Rehnitz J, Messmer B, Bender U, Nguyen XP, Germeyer A, Hinderhofer K, Strowitzki T, Capp E. Activation of AKT/mammalian target of rapamycin signaling in the peripheral blood of women with premature ovarian insufficiency and its correlation with FMR1 expression. Reprod Biol Endocrinol 2022; 20:44. [PMID: 35248053 PMCID: PMC8898473 DOI: 10.1186/s12958-022-00919-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 02/27/2022] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND The protein kinase B (AKT)/mammalian target of rapamycin (mTOR) signaling pathway regulates early follicular activation and follicular pool maintenance in female germline cells. Fragile X mental retardation 1 (FMR1) regulates folliculogenesis and it is variably expressed in patients with Premature Ovary Insufficiency. FMR1 expression is supposed to be linked to AKT/mTOR signaling in an ovarian response dependent manner as demonstrated in recent in vitro and in vivo studies in the female germline in vitro and in vivo. METHODS We evaluated changes in the expression of AKT/mTOR signaling pathway genes by real time PCR in the peripheral blood of 74 patients with Premature Ovarian Insufficiency and 56 fertile controls and correlated their expression with FMR1 expression. RESULTS Expression of the genes AKT1, TSC2, mTOR, and S6K was significantly more abundant in patients with POI than in the controls. For AKT1, TSC2 and mTOR, gene expression was not affected by FMR1-CGG repeat number in the 5´-untranslated region. FMR1 and S6K expression levels, however, were significantly upregulated in patients with POI and an FMR1 premutation. Independent of a premutation, expression of mTOR, S6K, and TSC2 was significantly correlated with that of FMR1 in all patients. Furthermore, when grouped according to ovarian reserve, this effect remained significant only for mTOR and S6K, with higher significance note in patients with Premature Ovarian Insufficiency than in the controls. CONCLUSIONS In Premature ovarian insufficiency patients, activation of AKT/mTOR signaling pathway is remarkable and putatively pathognomonic. Additionally, it seems to be triggered by an FMR1/mTOR/S6K linkage mechanism, most relevant in premutation carriers.
Collapse
Affiliation(s)
- Julia Rehnitz
- University Women's Hospital Heidelberg, Department of Gynecological Endocrinology and Fertility Disorders, Heidelberg, Germany.
| | - Birgitta Messmer
- University Women's Hospital Heidelberg, Department of Gynecological Endocrinology and Fertility Disorders, Heidelberg, Germany
| | - Ulrike Bender
- University Women's Hospital Heidelberg, Department of Gynecological Endocrinology and Fertility Disorders, Heidelberg, Germany
| | - Xuan Phuoc Nguyen
- University Women's Hospital Heidelberg, Department of Gynecological Endocrinology and Fertility Disorders, Heidelberg, Germany
| | - Ariane Germeyer
- University Women's Hospital Heidelberg, Department of Gynecological Endocrinology and Fertility Disorders, Heidelberg, Germany
| | - Katrin Hinderhofer
- Institute of Human Genetics, University Heidelberg, Laboratory of Molecular Genetics, Heidelberg, Germany
| | - Thomas Strowitzki
- University Women's Hospital Heidelberg, Department of Gynecological Endocrinology and Fertility Disorders, Heidelberg, Germany
| | - Edison Capp
- University Women's Hospital Heidelberg, Department of Gynecological Endocrinology and Fertility Disorders, Heidelberg, Germany
- Department of Obstetrics and Gynecology, Medicine School, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| |
Collapse
|
8
|
Ghezelayagh Z, Khoshdel-Rad N, Ebrahimi B. Human ovarian tissue in-vitro culture: primordial follicle activation as a new strategy for female fertility preservation. Cytotechnology 2022; 74:1-15. [PMID: 35185282 PMCID: PMC8816997 DOI: 10.1007/s10616-021-00510-2] [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: 05/29/2021] [Accepted: 11/18/2021] [Indexed: 02/03/2023] Open
Abstract
Cryopreservation and transplantation of ovarian tissue is the only fertility preservation option used for prepubertal girls and women who don't have a chance for embryo or oocyte vitrification. For women with aggressive cancer, hormone-responsive malignancies, autoimmune diseases, etc. ovary transplantation cannot be performed so an alternative technology called in-vitro follicle activation is thinkable. In this method, dormant primordial follicles are activated from the resting primordial pool by in-vitro culture and enter their growth phase. Different in-vitro culture media and supplements in addition to various culturing methods have been conducted for activating these dormant follicles. Furthermore, several signaling pathways such as Hippo, phosphatidylinositol-3-kinase, and mTOR influence follicle activation. Therefore, the addition of different activators of these signaling pathways can beneficially regulate this culture system. This review summarizes the findings on different aspects of human ovarian tissue culture strategies for in-vitro follicular activation, their medium, and different factors involved in this activation. Afterward, signaling pathways important for follicle activation and their clinical applications towards improving activation in culture are also reviewed.
Collapse
Affiliation(s)
- Zeinab Ghezelayagh
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
- Department of Developmental Biology, Faculty of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, ACECR, Tehran, Iran
| | - Niloofar Khoshdel-Rad
- Department of Developmental Biology, Faculty of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, ACECR, Tehran, Iran
- Department of Stem Cell and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Bita Ebrahimi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| |
Collapse
|
9
|
Regulation of mitochondrial morphology and metabolism by Jak-STAT pathway. JOURNAL OF ANIMAL REPRODUCTION AND BIOTECHNOLOGY 2021. [DOI: 10.12750/jarb.36.4.189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
10
|
Uzbekova S, Bertevello PS, Dalbies-Tran R, Elis S, Labas V, Monget P, Teixeira-Gomes AP. Metabolic exchanges between the oocyte and its environment: focus on lipids. Reprod Fertil Dev 2021; 34:1-26. [PMID: 35231385 DOI: 10.1071/rd21249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Finely regulated fatty acid (FA) metabolism within ovarian follicles is crucial to follicular development and influences the quality of the enclosed oocyte, which relies on the surrounding intra-follicular environment for its growth and maturation. A growing number of studies have examined the association between the lipid composition of follicular compartments and oocyte quality. In this review, we focus on lipids, their possible exchanges between compartments within the ovarian follicle and their involvement in different pathways during oocyte final growth and maturation. Lipidomics provides a detailed snapshot of the global lipid profiles and identified lipids, clearly discriminating the cells or fluid from follicles at distinct physiological stages. Follicular fluid appears as a main mediator of lipid exchanges between follicular somatic cells and the oocyte, through vesicle-mediated and non-vesicular transport of esterified and free FA. A variety of expression data allowed the identification of common and cell-type-specific actors of lipid metabolism in theca cells, granulosa cells, cumulus cells and oocytes, including key regulators of FA uptake, FA transport, lipid transformation, lipoprotein synthesis and protein palmitoylation. They act in harmony to accompany follicular development, and maintain intra-follicular homeostasis to allow the oocyte to accumulate energy and membrane lipids for subsequent meiotic divisions and first embryo cleavages.
Collapse
Affiliation(s)
- Svetlana Uzbekova
- CNRS, IFCE, INRAE, Université de Tours, PRC, F-37380 Nouzilly, France; and LK Ernst Federal Science Centre for Animal Husbandry, Podolsk, Russia
| | | | | | - Sebastien Elis
- CNRS, IFCE, INRAE, Université de Tours, PRC, F-37380 Nouzilly, France
| | - Valerie Labas
- CNRS, IFCE, INRAE, Université de Tours, PRC, F-37380 Nouzilly, France; and INRAE, Université de Tours, CHRU Tours, Plate-Forme PIXANIM, F-37380 Nouzilly, France
| | - Philippe Monget
- CNRS, IFCE, INRAE, Université de Tours, PRC, F-37380 Nouzilly, France
| | - Ana-Paula Teixeira-Gomes
- CNRS, IFCE, INRAE, Université de Tours, PRC, F-37380 Nouzilly, France; and INRAE, Université de Tours, CHRU Tours, Plate-Forme PIXANIM, F-37380 Nouzilly, France
| |
Collapse
|
11
|
Rehnitz J, Capp E, Messmer B, Nguyen XP, Germeyer A, Freis A, Dietrich JE, Hinderhofer K, Strowitzki T, Vogt PH. FMR1 and AKT/mTOR Signaling in Human Granulosa Cells: Functional Interaction and Impact on Ovarian Response. J Clin Med 2021; 10:jcm10173892. [PMID: 34501340 PMCID: PMC8432207 DOI: 10.3390/jcm10173892] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 08/20/2021] [Accepted: 08/25/2021] [Indexed: 01/12/2023] Open
Abstract
We aimed to determine whether a functional link with impact on female ovarian reserve exists between FMR1 expression and expression ratios of AKT/mTOR signaling genes in human granulosa cells in vivo, as suggested from prior in vitro data. Three hundred and nine women, who were classified as normal (NOR; n = 225) and poor (POR; n = 84) responders based on their ovarian reserve, were recruited during stimulation for assisted reproductive techniques. Expressions of FMR1 and of key genes of the AKT/mTOR and AKT/FOXO1/3 signaling pathways were comparatively analyzed in their granulosa cells. FMR1 expression in granulosa cells of NOR and POR correlated significantly with AKT1, TSC2, mTOR, and S6K expression. No correlation was found between FMR1 and FOXO1 in all, and FOXO3 expression in POR, patients. AKT1 expression was significantly higher and FOXO1 expression lower in POR samples, whereas AKT1 expression was lower and FOXO1 expression was higher in NOR samples. In human native granulosa cells, FMR1 expression significantly correlated with the expression of key genes of the AKT/mTOR signaling pathway, but not with the FOXO1/3 signaling pathway. Our data point to a functional link between FMR1 expression and expression of the AKT/mTOR signaling pathway genes controlling human follicular maturation.
Collapse
Affiliation(s)
- Julia Rehnitz
- Division of Reproductive Genetics, Department of Gynecological Endocrinology and Fertility Disorders, University Women’s Hospital Heidelberg, 69120 Heidelberg, Germany; (B.M.); (X.P.N.); (P.H.V.)
- Department of Gynecological Endocrinology and Fertility Disorders, University Women’s Hospital Heidelberg, 69120 Heidelberg, Germany; (E.C.); (A.G.); (A.F.); (J.E.D.); (T.S.)
- Correspondence: ; Tel.: +49-6221-56-7910
| | - Edison Capp
- Department of Gynecological Endocrinology and Fertility Disorders, University Women’s Hospital Heidelberg, 69120 Heidelberg, Germany; (E.C.); (A.G.); (A.F.); (J.E.D.); (T.S.)
- Department of Obstetrics and Gynecology, Medicine School, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-003, Brazil
| | - Birgitta Messmer
- Division of Reproductive Genetics, Department of Gynecological Endocrinology and Fertility Disorders, University Women’s Hospital Heidelberg, 69120 Heidelberg, Germany; (B.M.); (X.P.N.); (P.H.V.)
| | - Xuan Phuoc Nguyen
- Division of Reproductive Genetics, Department of Gynecological Endocrinology and Fertility Disorders, University Women’s Hospital Heidelberg, 69120 Heidelberg, Germany; (B.M.); (X.P.N.); (P.H.V.)
| | - Ariane Germeyer
- Department of Gynecological Endocrinology and Fertility Disorders, University Women’s Hospital Heidelberg, 69120 Heidelberg, Germany; (E.C.); (A.G.); (A.F.); (J.E.D.); (T.S.)
| | - Alexander Freis
- Department of Gynecological Endocrinology and Fertility Disorders, University Women’s Hospital Heidelberg, 69120 Heidelberg, Germany; (E.C.); (A.G.); (A.F.); (J.E.D.); (T.S.)
| | - Jens Erik Dietrich
- Department of Gynecological Endocrinology and Fertility Disorders, University Women’s Hospital Heidelberg, 69120 Heidelberg, Germany; (E.C.); (A.G.); (A.F.); (J.E.D.); (T.S.)
| | - Karin Hinderhofer
- Laboratory of Molecular Genetics, Institute of Human Genetics, University Heidelberg, 69120 Heidelberg, Germany;
| | - Thomas Strowitzki
- Department of Gynecological Endocrinology and Fertility Disorders, University Women’s Hospital Heidelberg, 69120 Heidelberg, Germany; (E.C.); (A.G.); (A.F.); (J.E.D.); (T.S.)
| | - Peter H. Vogt
- Division of Reproductive Genetics, Department of Gynecological Endocrinology and Fertility Disorders, University Women’s Hospital Heidelberg, 69120 Heidelberg, Germany; (B.M.); (X.P.N.); (P.H.V.)
| |
Collapse
|
12
|
Ferrer-Buitrago M, Tilleman L, Thys V, Hachem A, Boel A, Van Nieuwerburgh F, Deforce D, Leybaert L, De Sutter P, Parrington J, Heindryckx B. Comparative study of preimplantation development following distinct assisted oocyte activation protocols in a PLC-zeta knockout mouse model. Mol Hum Reprod 2021; 26:801-815. [PMID: 32898251 DOI: 10.1093/molehr/gaaa060] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/06/2020] [Indexed: 12/13/2022] Open
Abstract
Mammalian fertilization encompasses a series of Ca2+ oscillations initiated by the sperm factor phospholipase C zeta (PLCζ). Some studies have shown that altering the Ca2+ oscillatory regime at fertilization affects preimplantation blastocyst development. However, assisted oocyte activation (AOA) protocols can induce oocyte activation in a manner that diverges profoundly from the physiological Ca2+ profiling. In our study, we used the newly developed PLCζ-null sperm to investigate the independent effect of AOA on mouse preimplantation embryogenesis. Based on previous findings, we hypothesized that AOA protocols with Ca2+ oscillatory responses might improve blastocyst formation rates and differing Ca2+ profiles might alter blastocyst transcriptomes. A total of 326 MII B6D2F1-oocytes were used to describe Ca2+ profiles and to compare embryonic development and individual blastocyst transcriptomes between four control conditions: C1 (in-vivo fertilization), C2 (ICSI control sperm), C3 (parthenogenesis) and C4 (ICSI-PLCζ-KO sperm) and four AOA groups: AOA1 (human recombinant PLCζ), AOA2 (Sr2+), AOA3 (ionomycin) and AOA4 (TPEN). All groups revealed remarkable variations in their Ca2+ profiles; however, oocyte activation rates were comparable between the controls (91.1% ± 13.8%) and AOA (86.9% ± 11.1%) groups. AOA methods which enable Ca2+ oscillatory responses (AOA1: 41% and AOA2: 75%) or single Ca2+ transients (AOA3: 50%) showed no significantly different blastocyst rates compared to ICSI control group (C2: 70%). In contrast, we observed a significant decrease in compaction (53% vs. 83%) and blastocyst rates (41% vs. 70%) in the absence of an initial Ca2+ trigger (AOA4) compared with the C2 group. Transcription profiles did not identify significant differences in gene expression levels between the ICSI control group (C2) and the four AOA groups.
Collapse
Affiliation(s)
- M Ferrer-Buitrago
- Ghent-Fertility and Stem Cell Team (G-FAST), Department for Reproductive Medicine, Ghent University Hospital, Ghent, Belgium.,CREA. Medicina de la Reproducción S.L. Calle San Martín, 4 - 46003 (Valencia, Spain)
| | - L Tilleman
- Laboratory for Pharmaceutical Biotechnology, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - V Thys
- Ghent-Fertility and Stem Cell Team (G-FAST), Department for Reproductive Medicine, Ghent University Hospital, Ghent, Belgium
| | - A Hachem
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK.,Department of Anatomy, College of Veterinary Medicine, University of Al-Qadisiyah, Diwaniyah City, Iraq
| | - A Boel
- Ghent-Fertility and Stem Cell Team (G-FAST), Department for Reproductive Medicine, Ghent University Hospital, Ghent, Belgium
| | - F Van Nieuwerburgh
- Laboratory for Pharmaceutical Biotechnology, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - D Deforce
- Laboratory for Pharmaceutical Biotechnology, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - L Leybaert
- Physiology Group, Department of Basic Medical Sciences, Ghent University, Ghent, Belgium
| | - P De Sutter
- Ghent-Fertility and Stem Cell Team (G-FAST), Department for Reproductive Medicine, Ghent University Hospital, Ghent, Belgium
| | - J Parrington
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - B Heindryckx
- Ghent-Fertility and Stem Cell Team (G-FAST), Department for Reproductive Medicine, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
13
|
Li Y, Liang Z, Liang Z, Yang X, Xia H, Yu H. Abnormal PIWI-interacting RNA profile and its association with the deformed extracellular matrix of oocytes from recurrent oocyte maturation arrest patients. Fertil Steril 2021; 115:1318-1326. [PMID: 33622565 DOI: 10.1016/j.fertnstert.2020.11.037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 11/19/2020] [Accepted: 11/30/2020] [Indexed: 01/01/2023]
Abstract
OBJECTIVE To depict the PIWI-interacting RNA (piRNA) profile in oocytes from patients with recurrent oocyte maturation arrest (ROMA) and explore the piRNA candidates associated with the disease. DESIGN An observational study. SETTING Academic research unit. PATIENT(S) Sixteen ROMA patients who provided 140 immature oocytes that arrested at metaphase I, and 146 control patients who provided 420 oocytes for in vitro culture that were collected at the stages of germinal vesicle (GV), metaphase I (MI), and MII. INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) Expression profiles of piRNA and quantitative reverse-transcription polymerase chain reaction (qRT-PCR) validating data of piR-hsa-17139 and its target genes. RESULT(S) After the piRNA profile was established using piRNA sequencing and hierarchical clustering, the target genes of the piRNA were predicted by bioinformatics databases and matched with mRNA sequencing data. The piRNA expression profiles showed a greater quantity of differentially expressed piRNAs in the older-stage oocytes compared with the early-stage oocytes. The piRNA and mRNA sequencing data indicated that the most affected genes were mainly concentrated in the extracellular matrix (ECM) pathway. Based on the comparison of the piRNA and mRNA sequencing data, four differentially expressed piRNAs were associated with modulation of those ECM pathway genes. The qRT-PCR validation confirmed that piR-hsa-17139 was the only up-regulated piRNA, and its target ECM genes were suppressed in ROMA oocytes. The expression level of piR-hsa-17139 declined slightly while the expression of its target ECM genes plunged dramatically during the development of normal oocytes. CONCLUSION(S) As the important genome monitors in gametogenesis, abnormally expressed piRNAs may affect the expression of ECM modulating genes, which subsequently contributes to ROMA.
Collapse
Affiliation(s)
- Yi Li
- Center for Reproductive Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China.
| | - Zhenjie Liang
- Center for Reproductive Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Zhongkun Liang
- Center for Reproductive Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xi Yang
- Center for Reproductive Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Huayang Xia
- Center for Reproductive Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Hao Yu
- Urological Surgery Department, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| |
Collapse
|
14
|
Bertevello PS, Teixeira-Gomes AP, Labas V, Cordeiro L, Blache MC, Papillier P, Singina G, Uzbekov R, Maillard V, Uzbekova S. MALDI-TOF Mass Spectrometry Revealed Significant Lipid Variations in Follicular Fluid and Somatic Follicular Cells but Not in Enclosed Oocytes between the Large Dominant and Small Subordinate Follicles in Bovine Ovary. Int J Mol Sci 2020; 21:E6661. [PMID: 32932995 PMCID: PMC7554725 DOI: 10.3390/ijms21186661] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/07/2020] [Accepted: 09/09/2020] [Indexed: 12/13/2022] Open
Abstract
Lipid metabolism in ovarian follicular cells supports the preparation of an enclosed oocyte to ovulation. We aimed to compare lipid composition of a dominant large follicle (LF) and subordinated small follicles (SFs) within the same ovaries. Mass spectrometry imaging displayed the differences in the distribution of several lipid features between the different follicles. Comparison of lipid fingerprints between LF and SF by Matrix Assisted Laser Desorption/Ionisation Time-Of-Flight (MALDI-TOF) mass spectrometry revealed that in the oocytes, only 8 out of 468 detected lipids (1.7%) significantly changed their abundance (p < 0.05, fold change > 2). In contrast, follicular fluid (FF), granulosa, theca and cumulus cells demonstrated 55.5%, 14.9%, 5.3% and 9.8% of significantly varied features between LF and SF, respectively. In total, 25.2% of differential lipids were identified and indicated potential changes in membrane and signaling lipids. Tremendous changes in FF lipid composition were likely due to the stage specific secretions from somatic follicular cells that was in line with the differences observed from FF extracellular vesicles and gene expression of candidate genes in granulosa and theca cells between LF and SF. In addition, lipid storage in granulosa and theca cells varied in relation to follicular size and atresia. Differences in follicular cells lipid profiles between LF and SF may probably reflect follicle atresia degree and/or accumulation of appropriate lipids for post-ovulation processes as formation of corpus luteum. In contrast, the enclosed oocyte seems to be protected during final follicular growth, likely due in part to significant lipid transformations in surrounding cumulus cells. Therefore, the enclosed oocyte could likely keep lipid building blocks and energy resources to support further maturation and early embryo development.
Collapse
Affiliation(s)
- Priscila Silvana Bertevello
- CNRS, IFCE, INRAE, Université de Tours, PRC, 37380 Nouzilly, France; (P.S.B.); (V.L.); (L.C.); (M.-C.B.); (P.P.); (V.M.)
| | - Ana-Paula Teixeira-Gomes
- INRAE, Université de Tours, ISP, 37380 Nouzilly, France;
- CHU de Tours, INRAE, Université de Tours, PRC, CIRE, 37380 Nouzilly, France
| | - Valerie Labas
- CNRS, IFCE, INRAE, Université de Tours, PRC, 37380 Nouzilly, France; (P.S.B.); (V.L.); (L.C.); (M.-C.B.); (P.P.); (V.M.)
- CHU de Tours, INRAE, Université de Tours, PRC, CIRE, 37380 Nouzilly, France
| | - Luiz Cordeiro
- CNRS, IFCE, INRAE, Université de Tours, PRC, 37380 Nouzilly, France; (P.S.B.); (V.L.); (L.C.); (M.-C.B.); (P.P.); (V.M.)
| | - Marie-Claire Blache
- CNRS, IFCE, INRAE, Université de Tours, PRC, 37380 Nouzilly, France; (P.S.B.); (V.L.); (L.C.); (M.-C.B.); (P.P.); (V.M.)
| | - Pascal Papillier
- CNRS, IFCE, INRAE, Université de Tours, PRC, 37380 Nouzilly, France; (P.S.B.); (V.L.); (L.C.); (M.-C.B.); (P.P.); (V.M.)
| | - Galina Singina
- L.K. Ernst Institute of Animal Husbandry, Dubrovitzy 60, Podolsk, 142132 Moscow, Russia;
| | - Rustem Uzbekov
- Laboratoire Biologie Cellulaire et Microscopie Électronique, Faculté de Médecine, Université de Tours, 10, bd Tonnellé, 37032 Tours, France;
| | - Virginie Maillard
- CNRS, IFCE, INRAE, Université de Tours, PRC, 37380 Nouzilly, France; (P.S.B.); (V.L.); (L.C.); (M.-C.B.); (P.P.); (V.M.)
- Laboratoire Biologie Cellulaire et Microscopie Électronique, Faculté de Médecine, Université de Tours, 10, bd Tonnellé, 37032 Tours, France;
| | - Svetlana Uzbekova
- CNRS, IFCE, INRAE, Université de Tours, PRC, 37380 Nouzilly, France; (P.S.B.); (V.L.); (L.C.); (M.-C.B.); (P.P.); (V.M.)
- CHU de Tours, INRAE, Université de Tours, PRC, CIRE, 37380 Nouzilly, France
| |
Collapse
|
15
|
Liao Q, Feng X, Li X, Chen G, Chen J, Yang B, Li K, Ai J. Lapatinib‑induced inhibition of ovarian function is counteracted by the STAT3 pathway both in vivo and in vitro. Oncol Rep 2020; 44:1127-1135. [PMID: 32582968 PMCID: PMC7388577 DOI: 10.3892/or.2020.7660] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 06/09/2020] [Indexed: 12/22/2022] Open
Abstract
The present study was designed to ascertain whether lapatinib, a tyrosine kinase inhibitor of epidermal growth factor receptor (EGFR), affects ovarian reserve and fertility potential in a mouse model. Female C57BL/6 mice were treated with either vehicle or lapatinib (100 or 200 mg/kg/day orally) for 4 weeks, after which body weight, vaginal smears, follicle numbers, serum anti‑Müllerian hormone (AMH) levels and mating outcomes were analyzed to assess the ovarian reserve and reproductive function. Slices from the ovaries of 4‑week‑old mice were cultured with lapatinib (0, 5 or 10 µM) for 24 and 48 h, and protein expression levels were assessed to validate the changes in signaling pathways. The results indicated that mice treated with 200 mg/kg lapatinib showed a slight decrease in body weight compared to those treated with vehicle or 100 mg/kg lapatinib. There was no statistical difference in estrous cyclicity among the three groups. No significant difference was observed in follicle numbers, AMH levels, histological morphologies of the ovaries or mating outcomes in the three groups of mice. Western blotting and immunohistochemical staining of the EGF receptor and its main downstream signaling pathways showed decreased phosphorylation of EGFR and mitogen‑activated protein kinase (MAPK)3/1 and increased phosphorylation of signal transducers and activators of transcription (STAT)3 in the lapatinib‑treated groups compared to the control group. Our study suggests that lapatinib has little effect on ovarian reserve and reproductive function in a mouse model. This lack of effect of lapatinib on ovarian function may be due to the activation of the STAT3 signaling pathway that counteracts the inhibitory effects of lapatinib on EGF receptors.
Collapse
Affiliation(s)
- Qiuyue Liao
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Xue Feng
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Xi Li
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Ge Chen
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Jing Chen
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Bin Yang
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Kezhen Li
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Jihui Ai
- Department of Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
16
|
Gonadal development and sex determination in mouse. Reprod Biol 2020; 20:115-126. [DOI: 10.1016/j.repbio.2020.01.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 01/21/2020] [Accepted: 01/25/2020] [Indexed: 12/18/2022]
|
17
|
Ochiai A, Kuroda K. Preconception resveratrol intake against infertility: Friend or foe? Reprod Med Biol 2020; 19:107-113. [PMID: 32273814 PMCID: PMC7138940 DOI: 10.1002/rmb2.12303] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 09/20/2019] [Accepted: 09/25/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Resveratrol is an antiaging, antioxidant, anti-inflammatory, and insulin-sensitizing natural polyphenolic compound. Growing evidence indicates that resveratrol has potential therapeutic effects in infertile women with diminished ovarian function, polycystic ovary syndrome (PCOS), or endometriosis. However, only one clinical trial in women undergoing in vitro fertilization (IVF) cycles using resveratrol has ever been reported. This review focuses on the potential therapeutic effects of resveratrol on pregnancy and on its advantages and disadvantages in pregnancy outcomes during infertility treatment. METHODS We performed a literature review to describe the known impacts of resveratrol on the ovary and endometrium. RESULTS Resveratrol upregulates sirtuin (SIRT)1 expression in ovaries, which is associated with protection against oxidative stress. It leads to the activation of telomerase activity and mitochondrial function, improving ovarian function. In the endometrium, resveratrol downregulates the CRABP2-RAR pathway leading to suppressing decidual and senescent changes of endometrial cells, which is essential for embryo implantation and placentation. Moreover, resveratrol may also induce deacetylation of important decidual-related genes. CONCLUSIONS Resveratrol has potential therapeutic effects for improving ovarian function; however, it also has anti-deciduogenic actions in uterine endometrium. In addition, its teratogenicity has not yet been ruled out; thus, resveratrol should be avoided during the luteal phase and pregnancy.
Collapse
Affiliation(s)
- Asako Ochiai
- Department of Obstetrics and GynecologyFaculty of MedicineJuntendo UniversityTokyoJapan
| | - Keiji Kuroda
- Department of Obstetrics and GynecologyFaculty of MedicineJuntendo UniversityTokyoJapan
- Center for Reproductive Medicine and Implantation ResearchSugiyama Clinic ShinjukuTokyoJapan
| |
Collapse
|
18
|
Zhang D, Zhang Y, Gao Y, Chai X, Pi R, Chan G, Hu Y. Translating traditional herbal formulas into modern drugs: a network-based analysis of Xiaoyao decoction. Chin Med 2020; 15:25. [PMID: 32190105 PMCID: PMC7075005 DOI: 10.1186/s13020-020-00302-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 02/12/2020] [Indexed: 12/22/2022] Open
Abstract
Background Traditional Chinese medicine (TCM) encompasses numerous herbal formulas which play critical therapeutic roles through "multi-components, multi-targets and multi-pathways" mechanisms. Exploring the interaction among these mechanisms can certainly help to depict the core therapeutic function of herbal formulas. Xiaoyao decoction (XYD) is one of the most well-known traditional Chinese medicine formulas which has been widely applied to treat various diseases. In this study, taking XYD as an example, we proposed a network pharmacology-based method to identify the main therapeutic targets of this herbal concoctions. Methods Chemical data of XYD were retrieved from the Traditional Chinese Medicine Systems Pharmacology Database (TCMSP), Traditional Chinese Medicines Integrated Database (TCMID) and Compound Reference Database (CRD) and screened oral bioavailability attributes from SwissADME using Veber's filter. Targets of sample chemicals were identified using the online tool similarity ensemble approach (SEA), and pathways were enriched using STRING database. On the basis of targets-pathways interactions from the enrichment, a "targets-pathways-targets" (TPT) network was constructed. In the TPT network, the importance of each target was calculated by the declining value of network efficiency, which represents the influential strength of a specific set-off target on the whole network. Network-based predictive results were statistically validated with existing experimental evidence. Results The TPT network was comprised of 279 nodes and 6549 edges. The declining value of network efficiency of the sample targets was significantly correlated with their involvement frequency in existing studies of XYD using Spearman's test (p < 0.001). The top 10% of candidate targets, such as AKT1, PIK3R1, NFKB1 and RELA, etc., were chosen as XYD's main therapeutic targets, which further show pharmacological functions synergistically through 11 main pathways. These pathways are responsible for endocrine, nutritional or metabolic diseases, neoplasms and diseases of the nervous system, etc. Conclusions The network pharmacology-based approach in the present study shows promising potential for identifying the main therapeutic targets from TCM formulas. This study provides valuable information for TCM researchers and clinicians for better understanding the main therapeutic targets and therapeutic roles of herbal decoctions in clinical settings.
Collapse
Affiliation(s)
- Daiyan Zhang
- 1State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Yun Zhang
- 1State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Yan Gao
- 1State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Xingyun Chai
- 2Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029 China
| | - Rongbiao Pi
- 3School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006 China
| | - Ging Chan
- 1State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Yuanjia Hu
- 1State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| |
Collapse
|
19
|
Hall SE, Upton RMO, McLaughlin EA, Sutherland JM. Phosphoinositide 3-kinase/protein kinase B (PI3K/AKT) and Janus kinase/signal transducer and activator of transcription (JAK/STAT) follicular signalling is conserved in the mare ovary. Reprod Fertil Dev 2019; 30:624-633. [PMID: 28945982 DOI: 10.1071/rd17024] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 09/03/2017] [Indexed: 01/02/2023] Open
Abstract
The mare ovary is unique in its anatomical structure; however, the signalling pathways responsible for physiological processes, such as follicular activation, remain uncharacterised. This provided us with the impetus to explore whether signalling molecules from important folliculogenesis pathways, phosphoinositide 3-kinase/protein kinase B (PI3K/AKT) and Janus kinase/signal transducer and activator of transcription (JAK/STAT), are conserved in the mare ovary. Messenger RNA expression of six genes important in follicle development was measured using quantitative polymerase chain reaction and protein localisation of key pathway members (PI3K, AKT1, phosphatase and tensin homologue (PTEN), JAK1, STAT3 and suppressor of cytokine signalling 4 (SOCS4)) was compared in tissue from fetal and adult mare ovaries. Tissue from adult ovaries exhibited significantly increased levels of mRNA expression of PI3K, AKT1, PTEN, JAK1, STAT3 and SOCS4 compared with tissue from fetal ovaries. PI3K, AKT1, JAK1 and STAT3 demonstrated redistributed localisation, from pregranulosa cells in fetal development, to both the oocyte and granulosa cells of follicles in the adult ovary, whilst negative feedback molecules PTEN and SOCS4 were only localised to the granulosa cells in the adult ovary. These findings suggest that the PI3K/AKT and JAK/STAT signalling pathways are utilised during folliculogenesis in the mare, similarly to previously studied mammalian species, and may serve as useful biomarkers for assessment of ovary development in the horse.
Collapse
Affiliation(s)
- Sally E Hall
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Life Sciences Building, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Rose M O Upton
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Life Sciences Building, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Eileen A McLaughlin
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Life Sciences Building, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Jessie M Sutherland
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Life Sciences Building, University of Newcastle, Callaghan, NSW 2308, Australia
| |
Collapse
|
20
|
Sutherland JM, Frost ER, Ford EA, Peters AE, Reed NL, Seldon AN, Mihalas BP, Russel DL, Dunning KR, McLaughlin EA. Janus kinase JAK1 maintains the ovarian reserve of primordial follicles in the mouse ovary. Mol Hum Reprod 2019; 24:533-542. [PMID: 30247637 DOI: 10.1093/molehr/gay041] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 09/24/2018] [Indexed: 01/13/2023] Open
Abstract
STUDY QUESTION Is the Janus kinase and signal transducer and activator of transcription (JAK-STAT) signalling pathway involved in ovarian follicle development and primordial follicle activation? SUMMARY ANSWER JAK1 is a key factor involved in the regulation of primordial follicle activation and maintenance of the ovarian reserve. WHAT IS KNOWN ALREADY A series of integrated, intrinsic signalling pathways (including PI3K/AKT, mTOR and KITL) are responsible for regulating the ovarian reserve of non-growing primordial follicles and ultimately female fertility. The JAK-STAT signal transduction pathway is highly conserved with established roles in cell division and differentiation. Key pathway members (specifically JAK1, STAT3 and SOCS4) have been previously implicated in early follicle development. STUDY DESIGN, SIZE, DURATION A laboratory animal study was undertaken using the C57Bl/6 inbred mouse strain as a model for human ovarian follicle development. To determine which Jak genes were most abundantly expressed during primordial follicle activation, mRNA expression was analysed across a developmental time-course, with ovaries collected from female mice at post-natal days 1 (PND1), 4 (PND4), 8 (PND8), as well as at 6 weeks (6WK) and 7 months (7MTH) (n ≥ 4). Functional analysis of JAK1 was performed on PND2 mouse ovaries subjected to in vitro explant culture treated with 12.5 μM Ruxolitinib (JAK inhibitor) or vehicle control (DMSO) for 48 h prior to histological assessment (n ≥ 4). PARTICIPANTS/MATERIALS, SETTING, METHODS The expression and localization of the JAK family during ovarian follicle development in the C57Bl/6 inbred mouse strain were evaluated using quantitative PCR, immunoblotting and immunolocalisation. Functional studies were undertaken using the JAK inhibitor Ruxolitinib to investigate the underpinning cellular mechanisms via biochemical in vitro inhibition and histological assessment of intact neonate ovaries. All experiments were replicated at least three times using tissue from different mice unless otherwise stated. MAIN RESULTS AND THE ROLE OF CHANCE Jak1 is the predominant Jak mRNA expressed in the C57Bl/6 mouse ovary across all developmental time-points assessed (P ≤ 0.05). Forty-eight hour inhibition of JAK1 with Ruxolitinib of PND2 ovaries in vitro demonstrated concomitant acceleration of primordial follicle activation and apoptosis (P ≤ 0.001) and upregulation of downstream JAK-STAT pathway members STAT3 and suppressors of cytokine signalling 4 (SOCS4). LARGE-SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION Results are shown in one species, the C57Bl/6 mouse strain as an established model of human ovary development. Ruxolitinib also inhibits JAK2, with decreased efficacy. However, Jak2 mRNA had limited expression in the mouse ovary, particularly at the neonatal stages of follicle development, thus any effect of Ruxolitinib on primordial follicle activation was unlikely to be mediated via this isoform. WIDER IMPLICATIONS OF THE FINDINGS This study supports a key role for JAK1 in the maintenance and activation of primordial follicles, with potential for targeting the JAK-STAT pathway as a method of regulating the ovarian reserve and female fertility. STUDY FUNDING AND COMPETING INTEREST(S) This project has been funded by the Australian National Health and Medical Research Council (G1600095) and The Hunter Medical Research Institute Bob and Terry Kennedy Children's Research Project Grant in Pregnancy & Reproduction (G1501433). All authors declare no conflict of interests.
Collapse
Affiliation(s)
- Jessie M Sutherland
- School of Biomedical Science & Pharmacy, University of Newcastle, Callaghan, NSW, Australia
| | - Emily R Frost
- School of Biomedical Science & Pharmacy, University of Newcastle, Callaghan, NSW, Australia
| | - Emmalee A Ford
- School of Biomedical Science & Pharmacy, University of Newcastle, Callaghan, NSW, Australia
| | - Alexandra E Peters
- School of Biomedical Science & Pharmacy, University of Newcastle, Callaghan, NSW, Australia
| | - Natalie L Reed
- School of Biomedical Science & Pharmacy, University of Newcastle, Callaghan, NSW, Australia
| | - Alexandra N Seldon
- School of Biomedical Science & Pharmacy, University of Newcastle, Callaghan, NSW, Australia
| | - Bettina P Mihalas
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW, Australia
| | - Darryl L Russel
- Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Kylie R Dunning
- Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia.,Australian Research Council, Centre of Excellence for Nanoscale BioPhotonics
| | - Eileen A McLaughlin
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW, Australia.,School of Biological Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
21
|
SIRT1 participates in the response to methylglyoxal-dependent glycative stress in mouse oocytes and ovary. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1389-1401. [PMID: 30771486 DOI: 10.1016/j.bbadis.2019.02.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 01/18/2019] [Accepted: 02/11/2019] [Indexed: 12/21/2022]
Abstract
Methylglyoxal (MG), a highly reactive dicarbonyl derived from metabolic processes, is the most powerful precursor of advanced glycation end products (AGEs). Glycative stress has been recently associated with ovarian dysfunctions in aging and PCOS syndrome. We have investigated the role of the NAD+-dependent Class III deacetylase SIRT1 in the adaptive response to MG in mouse oocytes and ovary. In mouse oocytes, MG induced up-expression of glyoxalase 1 (Glo1) and glyoxalase 2 (Glo2) genes, components of the main MG detoxification system, whereas inhibition of SIRT1 by Ex527 or sirtinol reduced this response. In addition, the inhibition of SIRT1 worsened the effects of MG on oocyte maturation rates, while SIRT1 activation by resveratrol counteracted MG insult. Ovaries from female mice receiving 100 mg/kg MG by gastric administration for 28 days (MG mice) exhibited increased levels of SIRT1 along with over-expression of catalase, superoxide dismutase 2, SIRT3, PGC1α and mtTFA. Similar levels of MG-derived AGEs were observed in the ovaries from MG and control groups, along with enhanced protein expression of glyoxalase 1 in MG mice. Oocytes ovulated by MG mice exhibited atypical meiotic spindles, a condition predisposing to embryo aneuploidy. Our results from mouse oocytes revealed for the first time that SIRT1 could modulate MG scavenging by promoting expression of glyoxalases. The finding that up-regulation of glyoxalase 1 is associated with that of components of a SIRT1 functional network in the ovaries of MG mice provides strong evidence that SIRT1 participates in the response to methylglyoxal-dependent glycative stress in the female gonad.
Collapse
|
22
|
Anifandis G, Michopoulos A, Daponte A, Chatzimeletiou K, Simopoulou M, Messini CI, Polyzos NP, Vassiou K, Dafopoulos K, Goulis DG. Artificial oocyte activation: physiological, pathophysiological and ethical aspects. Syst Biol Reprod Med 2018; 65:3-11. [DOI: 10.1080/19396368.2018.1516000] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- George Anifandis
- Department of Obstetrics and Gynecology, ART Unit, University of Thessaly, School of Health Sciences, Faculty of Medicine, Larisa, Greece
| | - Alexandros Michopoulos
- Unit of Reproductive Endocrinology, 1st Department of Obstetrics and Gynecology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Alexandros Daponte
- Department of Obstetrics and Gynecology, ART Unit, University of Thessaly, School of Health Sciences, Faculty of Medicine, Larisa, Greece
| | - Katerina Chatzimeletiou
- Unit of Human Reproduction, 1st Department of Obstetrics and Gynecology, Aristotle University Medical School, Papageorgiou General Hospital, Thessaloniki, Greece
| | - Mara Simopoulou
- Department of Physiology, Kapodistrian University of Athens, School of Health Sciences, Faculty of Medicine, Athens, Greece
| | - Christina I. Messini
- Department of Obstetrics and Gynecology, ART Unit, University of Thessaly, School of Health Sciences, Faculty of Medicine, Larisa, Greece
| | - Nikolas P. Polyzos
- Vrije Universiteit Brussel, Brussels, Belgium
- Universitair Ziekenhuis Brussel, Brussels, Belgium
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Katerina Vassiou
- Department of Anatomy, University of Thessaly, School of Health Sciences, Faculty of Medicine, Larisa, Greece
| | - Konstantinos Dafopoulos
- Department of Obstetrics and Gynecology, ART Unit, University of Thessaly, School of Health Sciences, Faculty of Medicine, Larisa, Greece
| | - Dimitrios G. Goulis
- Unit of Reproductive Endocrinology, 1st Department of Obstetrics and Gynecology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
23
|
Shah JS, Sabouni R, Cayton Vaught KC, Owen CM, Albertini DF, Segars JH. Biomechanics and mechanical signaling in the ovary: a systematic review. J Assist Reprod Genet 2018; 35:1135-1148. [PMID: 29691711 PMCID: PMC6063820 DOI: 10.1007/s10815-018-1180-y] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 04/05/2018] [Indexed: 01/19/2023] Open
Abstract
PURPOSE Mammalian oogenesis and folliculogenesis share a dynamic connection that is critical for gamete development. For maintenance of quiescence or follicular activation, follicles must respond to soluble signals (growth factors and hormones) and physical stresses, including mechanical forces and osmotic shifts. Likewise, mechanical processes are involved in cortical tension and cell polarity in oocytes. Our objective was to examine the contribution and influence of biomechanical signaling in female mammalian gametogenesis. METHODS We performed a systematic review to assess and summarize the effects of mechanical signaling and mechanotransduction in oocyte maturation and folliculogenesis and to explore possible clinical applications. The review identified 2568 publications of which 122 met the inclusion criteria. RESULTS The integration of mechanical and cell signaling pathways in gametogenesis is complex. Follicular activation or quiescence are influenced by mechanical signaling through the Hippo and Akt pathways involving the yes-associated protein (YAP), transcriptional coactivator with PDZ-binding motif (TAZ), phosphatase and tensin homolog deleted from chromosome 10 (PTEN) gene, the mammalian target of rapamycin (mTOR), and forkhead box O3 (FOXO3) gene. CONCLUSIONS There is overwhelming evidence that mechanical signaling plays a crucial role in development of the ovary, follicle, and oocyte throughout gametogenesis. Emerging data suggest the complexities of mechanotransduction and the biomechanics of oocytes and follicles are integral to understanding of primary ovarian insufficiency, ovarian aging, polycystic ovary syndrome, and applications of fertility preservation.
Collapse
Affiliation(s)
- Jaimin S Shah
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Texas at Houston Health Science Center, Houston, TX, USA
| | - Reem Sabouni
- Jones Institute for Reproductive Medicine, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Kamaria C Cayton Vaught
- Howard W. and Georgeanna Seegar Jones Division of Reproductive Sciences and Women's Health Research, Baltimore, MD, USA
| | - Carter M Owen
- Program in Reproductive and Adult Endocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | | | - James H Segars
- Howard W. and Georgeanna Seegar Jones Division of Reproductive Sciences and Women's Health Research, Baltimore, MD, USA.
- Gynecology and Obstetrics, 720 Rutland Avenue/Ross 624, Baltimore, MD, 21205, USA.
| |
Collapse
|
24
|
Mao Z, Yang L, Lu X, Tan A, Wang Y, Ding F, Xiao L, Qi X, Yu Y. C1QTNF3 in the murine ovary and its function in folliculogenesis. Reproduction 2018; 155:333-346. [DOI: 10.1530/rep-17-0783] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 02/01/2018] [Indexed: 12/27/2022]
Abstract
C1q/tumor necrosis factor-related protein 3 (C1QTNF3) is a novel adipokine with modulating effects on metabolism, inflammation and the cardiovascular system. C1QTNF3 expression levels in the sera and omental adipose tissues of women with PCOS are low compared to control subjects. However, the expression and function of C1QTNF3 in the ovary has not previously been examined. Here, we assessed the expression patterns of C1qtnf3 in the ovary and explored its role in folliculogenesis. The C1qtnf3 transcript abundance was higher in large follicles than in small follicles and was under the influence of gonadotropin. C1QTNF3 was detected mainly in the granulosa cells and oocytes of growing follicles and modestly in the granulosa cells of atretic follicles and in luteal cells. Excess androgen significantly decreased C1QTNF3 expression in the ovaries in vivo and in granulosa cells in vitro. Recombinant C1QTNF3 protein accelerated the weight gain of ovarian explants and the growth of preantral follicles induced by follicle stimulating hormone (FSH) in vitro. The stimulatory effect of C1QTNF3 on ovarian growth was accompanied by the initiation of AKT, mTOR, p70S6K and 4EBP1 phosphorylation, an increase in CCND2 expression and a reduction in cleaved CASP3 levels. Moreover, the addition of C1QTNF3 accelerated proliferation and reduced activated CASP3/7 activity in granulosa cells. In vivo, the ovarian intrabursal administration of the C1QTNF3 antibody delayed gonadotropin-induced antral follicle development. Taken together, our data demonstrate that C1QTNF3 is an intraovarian factor that promotes follicle growth by accelerating proliferation, decelerating apoptosis and promoting AKT/mTOR phosphorylation.
Collapse
|
25
|
Wang L, Jiang Z, Huang D, Duan J, Huang C, Sullivan S, Vali K, Yin Y, Zhang M, Wegrzyn J, Tian X(C, Tang Y. JAK/STAT3 regulated global gene expression dynamics during late-stage reprogramming process. BMC Genomics 2018; 19:183. [PMID: 29510661 PMCID: PMC5840728 DOI: 10.1186/s12864-018-4507-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 01/29/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The generation of induced pluripotent stem cells (iPSCs) has underdefined mechanisms. In addition, leukemia inhibitory factor (LIF) activated Janus kinase/signal transducer and activator of transcription 3 (JAK/STAT3) pathway is the master regulator for naïve-state pluripotency achievement and maintenance. However, the regulatory process to attain naïve pluripotent iPSCs is not well understood. RESULTS We performed transcriptome analysis to dissect the genomic expression during mouse iPSC induction, with or without blocking the JAK/STAT3 activity. We describe JAK/STAT3 signaling-specific biological events such as gametogenesis, meiotic/mitotic cell cycle, and DNA repair, and JAK/STAT3-dependent expression of key transcription factors such as the naïve pluripotency-specific genes, developmental pluripotency associated (Dppa) family, along with histone modifiers and non-coding RNAs in reprogramming. We discover that JAK/STAT3 activity does not affect early phase mesenchymal to epithelial transition (MET) but is necessary for proper imprinting of the Dlk1-Dio3 region, an essential event for pluripotency achievement at late-reprogramming stage. This correlates with the JAK/STAT3-dependent stimulation of Dppa3 and Polycomb repressive complex 2 (PRC2) genes. We further demonstrate that JAK/STAT3 activity is essential for DNA demethylation of pluripotent loci including Oct4, Nanog, and the Dlk1-Dio3 regions. These findings correlate well with the previously identified STAT3 direct targets. We further propose a model of pluripotency achievement regulated by JAK/STAT3 signaling during the reprogramming process. CONCLUSIONS Our study illustrates novel insights for JAK/STAT3 promoted pluripotency establishment, which are valuable for further improving the naïve-pluripotent iPSC generation across different species including humans.
Collapse
Affiliation(s)
- Ling Wang
- Department of Animal Science, Institute for Systems Genomics, University of Connecticut, Storrs, CT USA
| | - Zongliang Jiang
- Department of Animal Science, Institute for Systems Genomics, University of Connecticut, Storrs, CT USA
- Present address: School of Animal Science, Louisiana State University, Baton Rouge, LA USA
| | - Delun Huang
- Department of Animal Science, Institute for Systems Genomics, University of Connecticut, Storrs, CT USA
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Animal Reproduction Institute, Guangxi University, Nanning, Guangxi People’s Republic of China
| | - Jingyue Duan
- Department of Animal Science, Institute for Systems Genomics, University of Connecticut, Storrs, CT USA
| | - Chang Huang
- Department of Animal Science, Institute for Systems Genomics, University of Connecticut, Storrs, CT USA
| | - Shannon Sullivan
- Department of Animal Science, Institute for Systems Genomics, University of Connecticut, Storrs, CT USA
| | - Kaneha Vali
- Department of Animal Science, Institute for Systems Genomics, University of Connecticut, Storrs, CT USA
| | - Yexuan Yin
- Department of Animal Science, Institute for Systems Genomics, University of Connecticut, Storrs, CT USA
| | - Ming Zhang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Animal Reproduction Institute, Guangxi University, Nanning, Guangxi People’s Republic of China
| | - Jill Wegrzyn
- Department of Ecology and Evolutionary Biology, Computational Biology Core, Institute for Systems Genomics, University of Connecticut, Storrs, CT USA
| | - Xiuchun ( Cindy) Tian
- Department of Animal Science, Institute for Systems Genomics, University of Connecticut, Storrs, CT USA
| | - Young Tang
- Department of Animal Science, Institute for Systems Genomics, University of Connecticut, Storrs, CT USA
| |
Collapse
|
26
|
Puttabyatappa M, Padmanabhan V. Developmental Programming of Ovarian Functions and Dysfunctions. VITAMINS AND HORMONES 2018; 107:377-422. [PMID: 29544638 PMCID: PMC6119353 DOI: 10.1016/bs.vh.2018.01.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The pathophysiological mechanisms underlying the origin of several ovarian pathologies remain unclear. In addition to the genetic basis, developmental insults are gaining attention as a basis for the origin of these pathologies. Such early insults include maternal over or under nutrition, stress, and exposure to environmental chemicals. This chapter reviews the development and physiological function of the ovary, the known ovarian pathologies, the developmental check points of ovarian differentiation impacted by developmental insults, the role played by steroidal and metabolic factors as mediaries, the epigenetic mechanisms via which these mediaries induce their effects, and the knowledge gaps for targeting future studies to ultimately aid in the development of improved treatments.
Collapse
|
27
|
TMCO1 is essential for ovarian follicle development by regulating ER Ca 2+ store of granulosa cells. Cell Death Differ 2018; 25:1686-1701. [PMID: 29467381 PMCID: PMC6143536 DOI: 10.1038/s41418-018-0067-x] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 01/15/2018] [Accepted: 01/16/2018] [Indexed: 01/10/2023] Open
Abstract
TMCO1 (transmembrane and coiled-coil domains 1) is an endoplasmic reticulum (ER) transmembrane protein that actively prevents Ca2+ stores from overfilling. To characterize its physiological function(s), we generated Tmco1−/− knockout (KO) mice. In addition to the main clinical features of human cerebrofaciothoracic (CFT) dysplasia spectrum, Tmco1−/− females manifest gradual loss of ovarian follicles, impaired ovarian follicle development, and subfertility with a phenotype analogous to the premature ovarian failure (POF) in women. In line with the role of TMCO1 as a Ca2+ load-activated Ca2+ channel, we have detected a supernormal Ca2+ signaling in Tmco1−/− granulosa cells (GCs). Interestingly, although spontaneous Ca2+ oscillation pattern was altered, ER Ca2+ stores of germinal vesicle (GV) stage oocytes and metaphase II (MII) arrested eggs were normal upon Tmco1 ablation. Combined with RNA-sequencing analysis, we also detected increased ER stress-mediated apoptosis and enhanced reactive oxygen species (ROS) level in Tmco1−/− GCs, indicating the dysfunctions of GCs upon TMCO1 deficiency. Taken together, these results reveal that TMCO1 is essential for ovarian follicle development and female fertility by maintaining ER Ca2+ homeostasis of GCs, disruption of which causes ER stress-mediated apoptosis and increased cellular ROS level in GCs and thus leads to impaired ovarian follicle development.
Collapse
|
28
|
Michaelis M, Sobczak A, Koczan D, Langhammer M, Reinsch N, Schoen J, Weitzel JM. Selection for female traits of high fertility affects male reproductive performance and alters the testicular transcriptional profile. BMC Genomics 2017; 18:889. [PMID: 29157197 PMCID: PMC5697431 DOI: 10.1186/s12864-017-4288-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 11/08/2017] [Indexed: 02/08/2023] Open
Abstract
Background Many genes important for reproductive performance are shared by both sexes. However, fecundity indices are primarily based on female parameters such as litter size. We examined a fertility mouse line (FL2), which has a considerably increased number of offspring and a total litter weight of 180% compared to a randomly bred control line (Ctrl) after more than 170 generations of breeding. In the present study, we investigated whether there might be a parallel evolution in males after more than 40 years of breeding in this outbred mouse model. Results Males of the fertility mouse line FL2 showed reduced sperm motility performance in a 5 h thermal stress experiment and reduced birth rate in the outbred mouse line. Transcriptional analysis of the FL2 testis showed the differential expression of genes associated with steroid metabolic processes (Cyp1b1, Cyp19a1, Hsd3b6, and Cyp21a1) and female fecundity (Gdf9), accompanied by 150% elevated serum progesterone levels in the FL2 males. Cluster analysis revealed the downregulation of genes of the kallikrein-related peptidases (KLK) cluster located on chromosome 7 in addition to alterations in gene expression with serine peptidase activity, e.g., angiotensinogen (Agt), of the renin-angiotensin system essential for ovulation. Although a majority of functional annotations map to female reproduction and ovulation, these genes are differentially expressed in FL2 testis. Conclusions These data indicate that selection for primary female traits of increased litter size not only affects sperm characteristics but also manifests as transcriptional alterations of the male side likely with direct long-term consequences for the reproductive performance of the mouse line. Electronic supplementary material The online version of this article (10.1186/s12864-017-4288-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marten Michaelis
- Institute of Reproductive Biology, University of Rostock, Rostock, Germany. .,Leibniz Institute for Farm Animal Biology (FBN), Institute of Reproductive Biology, FBN Dummerstorf, Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany.
| | - Alexander Sobczak
- Institute of Reproductive Biology, University of Rostock, Rostock, Germany
| | - Dirk Koczan
- Institute of Immunology, University of Rostock, Rostock, Germany
| | - Martina Langhammer
- Institute of Genetics and Biometry, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Norbert Reinsch
- Institute of Genetics and Biometry, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Jennifer Schoen
- Institute of Reproductive Biology, University of Rostock, Rostock, Germany
| | - Joachim M Weitzel
- Institute of Reproductive Biology, University of Rostock, Rostock, Germany. .,Leibniz Institute for Farm Animal Biology (FBN), Institute of Reproductive Biology, FBN Dummerstorf, Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany.
| |
Collapse
|
29
|
Differential Expression of miR-93 and miR-21 in Granulosa Cells and Follicular Fluid of Polycystic Ovary Syndrome Associating with Different Phenotypes. Sci Rep 2017; 7:14671. [PMID: 29116087 PMCID: PMC5676684 DOI: 10.1038/s41598-017-13250-1] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 08/31/2017] [Indexed: 12/14/2022] Open
Abstract
The heterogeneous and multifactorial essence of polycystic ovary syndrome (PCOS) renders a remarkable significance to microRNAs (miRNAs). Normo-androgenic (NA) and hyperandrogenic (HA) PCOS patients were compared with matched healthy women. Expression of miRNAs and TGFβ signaling genes was studied by qRT-PCR and western blotting. Effect of androgen on expression of miR-93 and miR-21 and involvement of androgen receptor were appraised. In granulosa cells (GCs), miR-93 and miR-21 showed significantly increased levels in HA patients compared to NA patients. On the contrary, follicular fluid (FF) levels of both miRNAs were significantly decreased in HA group compared to control women. No significant change in the expression of miRNAs in serum samples was detected. Furthermore, mRNA levels of SMAD7 and TGFBR2 were significantly downregulated in GCs of HA group compared to NA and control subjects. TGFBR2 protein level was significantly decreased in HA patients compared to controls. Free testosterone and free androgen index were positively correlated with expression of miR-93 and miR-21 in GCs of PCOS group. Our findings show distinct molecular signature of different subtypes of PCOS. Intermediary position of miRNAs as androgen responsive factors may play critical role in the pathogenesis of PCOS in hyperandrogenic condition.
Collapse
|
30
|
Saccon TD, Moreira F, Cruz LA, Mondadori RG, Fang Y, Barros CC, Spinel L, Bartke A, Masternak MM, Schneider A. Ovarian aging and the activation of the primordial follicle reserve in the long-lived Ames dwarf and the short-lived bGH transgenic mice. Mol Cell Endocrinol 2017; 455:23-32. [PMID: 27771355 PMCID: PMC5397383 DOI: 10.1016/j.mce.2016.10.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 06/22/2016] [Accepted: 10/15/2016] [Indexed: 12/09/2022]
Abstract
The aim of this study was to evaluate the effect of growth hormone (GH) in the maintenance of the ovarian primordial follicle reserve. Ovaries from 16 mo old GH-deficient Ames Dwarf (df/df) and Normal (N/df) mice were used. A subgroup of df/df and N mice received GH or saline injections for six weeks starting at 14 mo of age. In addition, ovaries from 12 mo old mice overexpressing bovine GH (bGH) and controls were used. df/df mice had higher number of primordial and total follicles than N/df mice (p < 0.05), while GH treatment decreased follicle counts in both genotypes (p < 0.05). In addition, bGH mice had lower number of primordial and total follicles than the controls (p < 0.05). pFoxO3a levels were higher in mice treated with GH and in bGH mice (p < 0.05) when comparing with age match controls. These results indicate that increased circulating GH is associated with a reduced ovarian primordial follicle reserve and increased pFoxO3a content in oocytes.
Collapse
Affiliation(s)
- Tatiana D Saccon
- Faculdade de Nutrição, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Fabiana Moreira
- Campus Araquari, Instituto Federal Catarinense, Araquari, SC, Brazil
| | - Luis A Cruz
- Instituto de Biologia, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Rafael G Mondadori
- Instituto de Biologia, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Yimin Fang
- Departments of Internal Medicine and Physiology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Carlos C Barros
- Faculdade de Nutrição, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - L Spinel
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, USA
| | - A Bartke
- Departments of Internal Medicine and Physiology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Michal M Masternak
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, USA; Department of Head and Neck Surgery, The Greater Poland Cancer Centre, Poznan, Poland
| | - A Schneider
- Faculdade de Nutrição, Universidade Federal de Pelotas, Pelotas, RS, Brazil; College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, USA.
| |
Collapse
|
31
|
FMR1 and AKT/mTOR signalling pathways: potential functional interactions controlling folliculogenesis in human granulosa cells. Reprod Biomed Online 2017; 35:485-493. [DOI: 10.1016/j.rbmo.2017.07.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 07/11/2017] [Accepted: 07/26/2017] [Indexed: 11/20/2022]
|
32
|
Molecular Mechanisms Responsible for Increased Vulnerability of the Ageing Oocyte to Oxidative Damage. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:4015874. [PMID: 29312475 PMCID: PMC5664291 DOI: 10.1155/2017/4015874] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 08/03/2017] [Indexed: 12/23/2022]
Abstract
In their midthirties, women experience a decline in fertility, coupled to a pronounced increase in the risk of aneuploidy, miscarriage, and birth defects. Although the aetiology of such pathologies are complex, a causative relationship between the age-related decline in oocyte quality and oxidative stress (OS) is now well established. What remains less certain are the molecular mechanisms governing the increased vulnerability of the aged oocyte to oxidative damage. In this review, we explore the reduced capacity of the ageing oocyte to mitigate macromolecular damage arising from oxidative insults and highlight the dramatic consequences for oocyte quality and female fertility. Indeed, while oocytes are typically endowed with a comprehensive suite of molecular mechanisms to moderate oxidative damage and thus ensure the fidelity of the germline, there is increasing recognition that the efficacy of such protective mechanisms undergoes an age-related decline. For instance, impaired reactive oxygen species metabolism, decreased DNA repair, reduced sensitivity of the spindle assembly checkpoint, and decreased capacity for protein repair and degradation collectively render the aged oocyte acutely vulnerable to OS and limits their capacity to recover from exposure to such insults. We also highlight the inadequacies of our current armoury of assisted reproductive technologies to combat age-related female infertility, emphasising the need for further research into mechanisms underpinning the functional deterioration of the ageing oocyte.
Collapse
|
33
|
Sun HY, Li Q, Liu YY, Wei XH, Pan CS, Fan JY, Han JY. Xiao-Yao-San, a Chinese Medicine Formula, Ameliorates Chronic Unpredictable Mild Stress Induced Polycystic Ovary in Rat. Front Physiol 2017; 8:729. [PMID: 29018356 PMCID: PMC5614964 DOI: 10.3389/fphys.2017.00729] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 09/07/2017] [Indexed: 12/16/2022] Open
Abstract
Chronic stress induces endocrine disturbance, which contributes to the development of polycystic ovary syndrome (PCOS), a condition that remains a challenge for clinicians to cope with. The present study investigated the effect of Xiao-Yao-San (XYS), a traditional Chinese medicine formula used for treatment of gynecological disease, on the chronic stress-induced polycystic ovary and its underlying mechanism. Female Sprague-Dwaley rats underwent a 3 weeks chronic unpredictable mild stress (CUMS) procedure to establish the PCOS model, followed by 4 weeks treatment with XYS (0.505 g/kg or 1.01 g/kg) by gavage. Granulosa cells were exposed to noradrenaline (1 mM) in vitro for 24 h, followed by incubation with or without XYS-treated rat serum for 24 h. Post-treatment with XYS ameliorated CUMS-induced irregular estrous cycles and follicles development abnormalities, decrease of estradiol and progesterone level as well as increase of luteinizing hormone in serum, reduced cystic follicles formation and the apoptosis and autophagy of granulosa cells, attenuated the increase in dopamine beta hydroxylase and c-fos level in locus coeruleus, the noradrenaline level in serum and ovarian tissue, and the expression of beta 2 adrenergic receptor in ovarian tissue. Besides, XYS alleviated the reduction of phosphorylation of ribosomal protein S6 kinase polypeptide I and protein kinase B, as well as the increase of microtubule-associated protein light chain 3-I to microtubule-associated protein light chain 3-II conversion both in vivo and in vitro. This study demonstrated XYS as a potential strategy for CUMS induced polycystic ovary, and suggested that the beneficial role of XYS was correlated with the regulation of the sympathetic nerve activity.
Collapse
Affiliation(s)
- Hao-Yu Sun
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking UniversityBeijing, China.,Tasly Microcirculation Research Center, Peking University Health Science CenterBeijing, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of ChinaBeijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of ChinaBeijing, China
| | - Quan Li
- Tasly Microcirculation Research Center, Peking University Health Science CenterBeijing, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of ChinaBeijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of ChinaBeijing, China.,State Key Laboratory of Core Technology in Innovative Chinese MedicineBeijing, China
| | - Yu-Ying Liu
- Tasly Microcirculation Research Center, Peking University Health Science CenterBeijing, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of ChinaBeijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of ChinaBeijing, China.,State Key Laboratory of Core Technology in Innovative Chinese MedicineBeijing, China
| | - Xiao-Hong Wei
- Tasly Microcirculation Research Center, Peking University Health Science CenterBeijing, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of ChinaBeijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of ChinaBeijing, China.,State Key Laboratory of Core Technology in Innovative Chinese MedicineBeijing, China
| | - Chun-Shui Pan
- Tasly Microcirculation Research Center, Peking University Health Science CenterBeijing, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of ChinaBeijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of ChinaBeijing, China.,State Key Laboratory of Core Technology in Innovative Chinese MedicineBeijing, China
| | - Jing-Yu Fan
- Tasly Microcirculation Research Center, Peking University Health Science CenterBeijing, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of ChinaBeijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of ChinaBeijing, China.,State Key Laboratory of Core Technology in Innovative Chinese MedicineBeijing, China
| | - Jing-Yan Han
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking UniversityBeijing, China.,Tasly Microcirculation Research Center, Peking University Health Science CenterBeijing, China.,Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of ChinaBeijing, China.,Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of ChinaBeijing, China.,State Key Laboratory of Core Technology in Innovative Chinese MedicineBeijing, China
| |
Collapse
|
34
|
Hachem A, Godwin J, Ruas M, Lee HC, Ferrer Buitrago M, Ardestani G, Bassett A, Fox S, Navarrete F, de Sutter P, Heindryckx B, Fissore R, Parrington J. PLCζ is the physiological trigger of the Ca 2+ oscillations that induce embryogenesis in mammals but conception can occur in its absence. Development 2017; 144:2914-2924. [PMID: 28694258 DOI: 10.1242/dev.150227] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 07/01/2017] [Indexed: 12/21/2022]
Abstract
Activation of the egg by the sperm is the first, vital stage of embryogenesis. The sperm protein PLCζ has been proposed as the physiological agent that triggers the Ca2+ oscillations that normally initiate embryogenesis. Consistent with this, recombinant PLCζ induces Ca2+ oscillations in eggs and debilitating mutations in the PLCZ1 gene are associated with infertility in men. However, there has been no evidence that knockout of the gene encoding PLCζ abolishes the ability of sperm to induce Ca2+ oscillations in eggs. Here, we show that sperm derived from Plcz1-/- male mice fail to trigger Ca2+ oscillations in eggs, cause polyspermy and thus demonstrate that PLCζ is the physiological trigger of these Ca2+ oscillations. Remarkably, some eggs fertilized by PLCζ-null sperm can develop, albeit at greatly reduced efficiency, and after a significant time-delay. In addition, Plcz1-/- males are subfertile but not sterile, suggesting that in the absence of PLCζ, spontaneous egg activation can eventually occur via an alternative route. This is the first demonstration that in vivo fertilization without the normal physiological trigger of egg activation can result in offspring. PLCζ-null sperm now make it possible to resolve long-standing questions in fertilization biology, and to test the efficacy and safety of procedures used to treat human infertility.
Collapse
Affiliation(s)
- Alaa Hachem
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Jonathan Godwin
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Margarida Ruas
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Hoi Chang Lee
- Department of Veterinary and Animal Sciences, University of Massachusetts, 661 North Pleasant Street, Amherst, MA 01003-9286, USA
| | - Minerva Ferrer Buitrago
- Department for Reproductive Medicine, Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium
| | - Goli Ardestani
- Department of Veterinary and Animal Sciences, University of Massachusetts, 661 North Pleasant Street, Amherst, MA 01003-9286, USA
| | - Andrew Bassett
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Sebastian Fox
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Felipe Navarrete
- Department of Veterinary and Animal Sciences, University of Massachusetts, 661 North Pleasant Street, Amherst, MA 01003-9286, USA
| | - Petra de Sutter
- Department for Reproductive Medicine, Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium
| | - Björn Heindryckx
- Department for Reproductive Medicine, Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium
| | - Rafael Fissore
- Department of Veterinary and Animal Sciences, University of Massachusetts, 661 North Pleasant Street, Amherst, MA 01003-9286, USA
| | - John Parrington
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| |
Collapse
|
35
|
Hart RJ. Physiological Aspects of Female Fertility: Role of the Environment, Modern Lifestyle, and Genetics. Physiol Rev 2016; 96:873-909. [DOI: 10.1152/physrev.00023.2015] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Across the Western World there is an increasing trend to postpone childbearing. Consequently, the negative influence of age on oocyte quality may lead to a difficulty in conceiving for many couples. Furthermore, lifestyle factors may exacerbate a couple's difficulty in conceiving due mainly to the metabolic influence of obesity; however, the negative impacts of low peripheral body fat, excessive exercise, the increasing prevalence of sexually transmitted diseases, and smoking all have significant negative effects on fertility. Other factors that impede conception are the perceived increasing prevalence of the polycystic ovary syndrome, which is further exacerbated by obesity, and the presence of uterine fibroids and endometriosis (a progressive pelvic inflammatory disorder) which are more prevalent in older women. A tendency for an earlier sexual debut and to have more sexual partners has led to an increase in sexually transmitted diseases. In addition, there are several genetic influences that may limit the number of oocytes within the ovary; consequently, by postponing attempts at childbearing, a limitation of oocyte number may become evident, whereas in previous generations with earlier conception this potentially reduced reproductive life span did not manifest in infertility. Environmental influences on reproduction are under increasing scrutiny. Although firm evidence is lacking however, dioxin exposure may be linked to endometriosis, phthalate exposure may influence ovarian reserve, and bisphenol A may interfere with oocyte development and maturation. However, chemotherapy or radiotherapy is recognized to lead to ovarian damage and predispose the woman to ovarian failure.
Collapse
Affiliation(s)
- Roger J. Hart
- School of Women's and Infants Health, University of Western Australia & Fertility Specialists of Western Australia, Subiaco, Perth Western Australia
| |
Collapse
|
36
|
Ilha GF, Rovani MT, Gasperin BG, Antoniazzi AQ, Gonçalves PBD, Bordignon V, Duggavathi R. Lack of FSH support enhances LIF-STAT3 signaling in granulosa cells of atretic follicles in cattle. Reproduction 2016; 150:395-403. [PMID: 26336147 DOI: 10.1530/rep-15-0026] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Subordinate follicles (SFs) of bovine follicular waves undergo atresia due to declining FSH concentrations; however, the signalling mechanisms have not been fully deciphered. We used an FSH-induced co-dominance model to determine the effect of FSH on signalling pathways in granulosa cells of the second-largest follicles (SF in control cows and co-dominant follicle (co-DF2) in FSH-treated cows). The SF was smaller than DF in control cows while diameters of co-DF1 and co-DF2 in FSH-treated cows were similar. The presence of cleaved CASP3 protein confirmed that granulosa cells of SFs, but not of DFs and co-DFs, were apoptotic. To determine the effect of FSH on molecular characteristics of the second-largest follicles, we generated relative variables for the second largest follicle in each cow. For this, variables of SF or co-DF2 were divided by the variables of the largest follicle DF or co-DF1 in each cow. There was higher transcript abundance of MAPK1/3 and AKT1/2/3 but lower abundance of phosphorylated MAPK3/1 in SF than co-DF2 granulosa cells. Abundance of mRNA and phosphorylated protein of STAT3 was higher in granulosa cells of control SF than FSH-treated co-DF2. SF granulosa cells had higher levels of LIFR and IL6ST transcripts, the two receptors involved in STAT3 activation. Further, lower transcript abundance of interleukin 6 receptor (IL6R), another receptor involved in STAT3 activation, indicated that STAT3 activation in SF granulosa cells could be mainly due to leukemia inhibitory factor (LIF) signalling. These results indicate that atresia due to lack of FSH is associated with activated LIF-STAT3 signalling in SF granulosa cells, as FSH treatment reversed such activation.
Collapse
Affiliation(s)
- Gustavo Freitas Ilha
- Laboratory of Biotechnology and Animal Reproduction - BioRepVeterinary Hospital, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, BrazilLaboratory of Animal Reproduction - ReproPELFederal University of Pelotas, Capão do Leão, Rio Grande do Sul, BrazilDepartment of Animal ScienceMcGill University, 21111 Lakeshore Road, Sainte Anne de Bellevue, Quebec, Canada H9X 3V9
| | - Monique T Rovani
- Laboratory of Biotechnology and Animal Reproduction - BioRepVeterinary Hospital, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, BrazilLaboratory of Animal Reproduction - ReproPELFederal University of Pelotas, Capão do Leão, Rio Grande do Sul, BrazilDepartment of Animal ScienceMcGill University, 21111 Lakeshore Road, Sainte Anne de Bellevue, Quebec, Canada H9X 3V9
| | - Bernardo G Gasperin
- Laboratory of Biotechnology and Animal Reproduction - BioRepVeterinary Hospital, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, BrazilLaboratory of Animal Reproduction - ReproPELFederal University of Pelotas, Capão do Leão, Rio Grande do Sul, BrazilDepartment of Animal ScienceMcGill University, 21111 Lakeshore Road, Sainte Anne de Bellevue, Quebec, Canada H9X 3V9
| | - Alfredo Quites Antoniazzi
- Laboratory of Biotechnology and Animal Reproduction - BioRepVeterinary Hospital, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, BrazilLaboratory of Animal Reproduction - ReproPELFederal University of Pelotas, Capão do Leão, Rio Grande do Sul, BrazilDepartment of Animal ScienceMcGill University, 21111 Lakeshore Road, Sainte Anne de Bellevue, Quebec, Canada H9X 3V9
| | - Paulo Bayard Dias Gonçalves
- Laboratory of Biotechnology and Animal Reproduction - BioRepVeterinary Hospital, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, BrazilLaboratory of Animal Reproduction - ReproPELFederal University of Pelotas, Capão do Leão, Rio Grande do Sul, BrazilDepartment of Animal ScienceMcGill University, 21111 Lakeshore Road, Sainte Anne de Bellevue, Quebec, Canada H9X 3V9
| | - Vilceu Bordignon
- Laboratory of Biotechnology and Animal Reproduction - BioRepVeterinary Hospital, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, BrazilLaboratory of Animal Reproduction - ReproPELFederal University of Pelotas, Capão do Leão, Rio Grande do Sul, BrazilDepartment of Animal ScienceMcGill University, 21111 Lakeshore Road, Sainte Anne de Bellevue, Quebec, Canada H9X 3V9
| | - Raj Duggavathi
- Laboratory of Biotechnology and Animal Reproduction - BioRepVeterinary Hospital, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, BrazilLaboratory of Animal Reproduction - ReproPELFederal University of Pelotas, Capão do Leão, Rio Grande do Sul, BrazilDepartment of Animal ScienceMcGill University, 21111 Lakeshore Road, Sainte Anne de Bellevue, Quebec, Canada H9X 3V9
| |
Collapse
|
37
|
Clarke HJ, Vieux KF. Epigenetic inheritance through the female germ-line: The known, the unknown, and the possible. Semin Cell Dev Biol 2015; 43:106-116. [DOI: 10.1016/j.semcdb.2015.07.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 07/04/2015] [Accepted: 07/06/2015] [Indexed: 02/06/2023]
|
38
|
Sirtuin Functions in Female Fertility: Possible Role in Oxidative Stress and Aging. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:659687. [PMID: 26075037 PMCID: PMC4436464 DOI: 10.1155/2015/659687] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Revised: 10/13/2014] [Accepted: 11/04/2014] [Indexed: 12/15/2022]
Abstract
In search for strategies aimed at preventing oxidative threat to female fertility, a possible role of sirtuins has emerged. Sirtuins (silent information regulator 2 (Sir2) proteins), NAD(+) dependent enzymes with deacetylase and/or mono-ADP-ribosyltransferase activity, are emerging as key antiaging molecules and regulators in many diseases. Recently, a crucial role for SIRT1 and SIRT3, the main components of sirtuin family, as sensors and guardians of the redox state in oocytes, granulosa cells, and early embryos has emerged. In this context, the aim of the present review is to summarize current knowledge from research papers on the role of sirtuins in female fertility with particular emphasis on the impairment of SIRT1 signalling with oocyte aging. On this basis, the authors wish to build up a framework to promote research on the possible role of sirtuins as targets for future strategies for female fertility preservation.
Collapse
|
39
|
Zamah AM, Hassis ME, Albertolle ME, Williams KE. Proteomic analysis of human follicular fluid from fertile women. Clin Proteomics 2015; 12:5. [PMID: 25838815 PMCID: PMC4357057 DOI: 10.1186/s12014-015-9077-6] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 02/09/2015] [Indexed: 01/08/2023] Open
Abstract
Background Follicular fluid is a unique biological fluid in which the critical events of oocyte and follicular maturation and somatic cell-germ cell communication occur. Because of the intimate proximity of follicular fluid to the maturing oocyte, this fluid provides a unique window into the processes occurring during follicular maturation. A thorough identification of the specific components within follicular fluid may provide a better understanding of intrafollicular signaling, as well as reveal potential biomarkers of oocyte health for women undergoing assisted reproductive treatment. In this study, we used high and low pH HPLC peptide separations followed by mass spectrometry to perform a comprehensive proteomic analysis of human follicular fluid from healthy ovum donors. Next, using samples from a second set of patients, an isobaric mass tagging strategy for quantitative analysis was used to identify proteins with altered abundances after hCG treatment. Results A total of 742 follicular fluid proteins were identified in healthy ovum donors, including 413 that have not been previously reported. The proteins belong to diverse functional groups including insulin growth factor and insulin growth factor binding protein families, growth factor and related proteins, receptor signaling, defense/immunity, anti-apoptotic proteins, matrix metalloprotease related proteins, and complement activity. In a quantitative analysis, follicular fluid samples from age-matched women undergoing in vitro fertilization oocyte retrieval were compared and 17 follicular fluid proteins were found at significantly altered levels (p < 0.05) between pre-hCG and post-hCG samples. These proteins belong to a variety of functional processes, including protease inhibition, inflammation, and cell adhesion. Conclusions This database of FF proteins significantly extends the known protein components present during the peri-ovulatory period and provides a useful basis for future studies comparing follicular fluid proteomes in various fertility, disease, and environmental exposure conditions. We identified 17 differentially expressed proteins after hCG treatment and together these data showed the feasibility for defining biomarkers that illuminate how the ovarian follicle microenvironment is altered in various infertility-related conditions. Electronic supplementary material The online version of this article (doi:10.1186/s12014-015-9077-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alberuni M Zamah
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, University of Illinois at Chicago College of Medicine, Chicago, IL 60612 USA
| | - Maria E Hassis
- Sandler-Moore Mass Spectrometry Core Facility, University of California at San Francisco, San Francisco, CA 94143 USA
| | - Matthew E Albertolle
- Sandler-Moore Mass Spectrometry Core Facility, University of California at San Francisco, San Francisco, CA 94143 USA
| | - Katherine E Williams
- Sandler-Moore Mass Spectrometry Core Facility, University of California at San Francisco, San Francisco, CA 94143 USA ; Center for Reproductive Sciences and the Department of Obstetrics and Gynecology, University of California at San Francisco, San Francisco, CA 94143 USA
| |
Collapse
|
40
|
Cardoso RC, Puttabyatappa M, Padmanabhan V. Steroidogenic versus Metabolic Programming of Reproductive Neuroendocrine, Ovarian and Metabolic Dysfunctions. Neuroendocrinology 2015; 102:226-37. [PMID: 25832114 PMCID: PMC4591099 DOI: 10.1159/000381830] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 03/21/2015] [Indexed: 12/12/2022]
Abstract
The susceptibility of the reproductive system to early exposure to steroid hormones has become a major concern in our modern societies. Human fetuses are at risk of abnormal programming via exposure to endocrine disrupting chemicals, inadvertent use of contraceptive pills during pregnancy, as well as from excess exposure to steroids due to disease states. Animal models provide an unparalleled resource to understand the developmental origin of diseases. In female sheep, prenatal exposure to testosterone excess results in an array of adult reproductive disorders that recapitulate those seen in women with polycystic ovary syndrome (PCOS), including disrupted neuroendocrine feedback mechanisms, increased pituitary sensitivity to gonadotropin-releasing hormone, luteinizing hormone excess, functional hyperandrogenism, and multifollicular ovarian morphology culminating in early reproductive failure. Prenatal testosterone treatment also leads to fetal growth retardation, insulin resistance, and hypertension. Mounting evidence suggests that developmental exposure to an improper steroidal/metabolic environment may mediate the programming of adult disorders in prenatal testosterone-treated females, and these defects are maintained or amplified by the postnatal sex steroid and metabolic milieu. This review addresses the steroidal and metabolic contributions to the development and maintenance of the PCOS phenotype in the prenatal testosterone-treated sheep model, including the effects of prenatal and postnatal treatment with an androgen antagonist or insulin sensitizer as potential strategies to prevent/ameliorate these dysfunctions. Insights obtained from these intervention strategies on the mechanisms underlying these defects are likely to have translational relevance to human PCOS.
Collapse
Affiliation(s)
- Rodolfo C Cardoso
- Department of Pediatrics, University of Michigan, Ann Arbor, Mich., USA
| | | | | |
Collapse
|
41
|
Yesilaltay A, Dokshin GA, Busso D, Wang L, Galiani D, Chavarria T, Vasile E, Quilaqueo L, Orellana JA, Walzer D, Shalgi R, Dekel N, Albertini DF, Rigotti A, Page DC, Krieger M. Excess cholesterol induces mouse egg activation and may cause female infertility. Proc Natl Acad Sci U S A 2014; 111:E4972-80. [PMID: 25368174 PMCID: PMC4246315 DOI: 10.1073/pnas.1418954111] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The HDL receptor scavenger receptor, class B type I (SR-BI) controls the structure and fate of plasma HDL. Female SR-BI KO mice are infertile, apparently because of their abnormal cholesterol-enriched HDL particles. We examined the growth and meiotic progression of SR-BI KO oocytes and found that they underwent normal germinal vesicle breakdown; however, SR-BI KO eggs, which had accumulated excess cholesterol in vivo, spontaneously activated, and they escaped metaphase II (MII) arrest and progressed to pronuclear, MIII, and anaphase/telophase III stages. Eggs from fertile WT mice were activated when loaded in vitro with excess cholesterol by a cholesterol/methyl-β-cyclodextrin complex, phenocopying SR-BI KO oocytes. In vitro cholesterol loading of eggs induced reduction in maturation promoting factor and MAPK activities, elevation of intracellular calcium, extrusion of a second polar body, and progression to meiotic stages beyond MII. These results suggest that the infertility of SR-BI KO females is caused, at least in part, by excess cholesterol in eggs inducing premature activation and that cholesterol can activate WT mouse eggs to escape from MII arrest. Analysis of SR-BI KO female infertility raises the possibility that abnormalities in cholesterol metabolism might underlie some cases of human female infertility of unknown etiology.
Collapse
Affiliation(s)
| | - Gregoriy A Dokshin
- Departments of Biology and Howard Hughes Medical Institute, Whitehead Institute, Cambridge, MA 02142
| | - Dolores Busso
- Departamento de Nutrición, Diabetes y Metabolismo and
| | | | - Dalia Galiani
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot 76100, Israel
| | | | - Eliza Vasile
- The Koch Institute, Massachusetts Institute of Technology, Cambridge, MA 02139
| | | | - Juan Andrés Orellana
- Departamento de Neurología, Pontificia Universidad Católica de Chile, 8330024 Santiago, Chile
| | | | - Ruth Shalgi
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; and
| | - Nava Dekel
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - David F Albertini
- Department of Molecular and Integrative Physiology, Institute for Reproductive Health and Regenerative Medicine, University of Kansas Medical Center, Kansas City, KS 66160
| | | | - David C Page
- Departments of Biology and Howard Hughes Medical Institute, Whitehead Institute, Cambridge, MA 02142
| | | |
Collapse
|
42
|
Gu L, Liu H, Gu X, Boots C, Moley KH, Wang Q. Metabolic control of oocyte development: linking maternal nutrition and reproductive outcomes. Cell Mol Life Sci 2014; 72:251-71. [PMID: 25280482 DOI: 10.1007/s00018-014-1739-4] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2014] [Revised: 09/12/2014] [Accepted: 09/22/2014] [Indexed: 02/01/2023]
Abstract
Obesity, diabetes, and related metabolic disorders are major health issues worldwide. As the epidemic of metabolic disorders continues, the associated medical co-morbidities, including the detrimental impact on reproduction, increase as well. Emerging evidence suggests that the effects of maternal nutrition on reproductive outcomes are likely to be mediated, at least in part, by oocyte metabolism. Well-balanced and timed energy metabolism is critical for optimal development of oocytes. To date, much of our understanding of oocyte metabolism comes from the effects of extrinsic nutrients on oocyte maturation. In contrast, intrinsic regulation of oocyte development by metabolic enzymes, intracellular mediators, and transport systems is less characterized. Specifically, decreased acid transport proteins levels, increased glucose/lipid content and elevated reactive oxygen species in oocytes have been implicated in meiotic defects, organelle dysfunction and epigenetic alteration. Therefore, metabolic disturbances in oocytes may contribute to the diminished reproductive potential experienced by women with metabolic disorders. In-depth research is needed to further explore the underlying mechanisms. This review also discusses several approaches for metabolic analysis. Metabolomic profiling of oocytes, the surrounding granulosa cells, and follicular fluid will uncover the metabolic networks regulating oocyte development, potentially leading to the identification of oocyte quality markers and prevention of reproductive disease and poor outcomes in offspring.
Collapse
Affiliation(s)
- Ling Gu
- College of Animal Science and Technology, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, Jiangsu, China,
| | | | | | | | | | | |
Collapse
|
43
|
Bar-Joseph H, Ben-Ami I, Ron-El R, Shalgi R, Chuderland D. Pigment epithelium–derived factor exerts antioxidative effects in granulosa cells. Fertil Steril 2014; 102:891-898.e3. [DOI: 10.1016/j.fertnstert.2014.06.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 06/10/2014] [Accepted: 06/10/2014] [Indexed: 01/07/2023]
|
44
|
McLaughlin M, Kinnell HL, Anderson RA, Telfer EE. Inhibition of phosphatase and tensin homologue (PTEN) in human ovary in vitro results in increased activation of primordial follicles but compromises development of growing follicles. Mol Hum Reprod 2014; 20:736-744. [PMID: 24830779 PMCID: PMC4106636 DOI: 10.1093/molehr/gau037] [Citation(s) in RCA: 135] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 05/01/2014] [Accepted: 05/09/2014] [Indexed: 12/22/2022] Open
Abstract
In the mammalian ovary a small number of follicles are steadily recruited from the quiescent pool to undergo development. Follicle loss, maintenance and growth are strictly controlled by complex molecular interactions including the phosphoinositide 3-kinase (PI3K)-protein kinase B (Akt) signalling pathway. Stimulation of PI3K promotes phosphorylation of Akt resulting in follicle survival and activation of growth whereas this pathway is suppressed by the actions of the phosphatase and tensin homologue (PTEN). The aim of this study was to determine the effect of dipotassium bisperoxo(5-hydroxypyridine-2-carboxyl)oxovanadate (bpV), a reversible inhibitor of PTEN, on the activation, survival and development of human ovarian follicles in vitro. Biopsied ovarian tissue fragments were obtained from 17 women aged 23-46 years and exposed to 1 µM bpV(HOpic) (n = 146) or control medium (n = 128) for 24 h. Media were then replaced with control medium and all tissue incubated for a further 5 days. Ovarian tissue from each treatment group was fixed after the initial 24 h culture period and phosphorylated Akt was quantified by western blotting. After 6 days incubation all tissue fragments were inspected under light microscopy and any secondary follicles ≥100 µm isolated. Isolated follicles were cultured individually in control medium supplemented with 100 ng/ml recombinant human activin A. Tissue fragments without follicles suitable for isolation were fixed and processed for histological and immunohistochemical analysis. During 6 days culture, follicle activation occurred in tissue samples from both treatment groups but with significantly more follicles progressing to the secondary stage of development in the presence of 1 µM bpV(HOpic) compared with control (31 versus 16%; P < 0.05). Increased activation was associated with increased Akt phosphorylation and increased nuclear export of FOXO3. However isolated and cultured follicles that had been exposed to bpV(HOpic) showed limited growth and reduced survival compared with follicles from control fragments (P < 0.05). This study demonstrates that inhibition of PTEN with bpV(HOpic) affects human ovarian follicle development by promoting the initiation of follicle growth and development to the secondary stage, as in rodent species, but severely compromises the survival of isolated secondary follicles.
Collapse
Affiliation(s)
- Marie McLaughlin
- Institute of Cell Biology and Centre for Integrative Physiology, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| | - Hazel L Kinnell
- Medical Research Council Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Richard A Anderson
- Medical Research Council Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Evelyn E Telfer
- Institute of Cell Biology and Centre for Integrative Physiology, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| |
Collapse
|
45
|
Identification of sites of STAT3 action in the female reproductive tract through conditional gene deletion. PLoS One 2014; 9:e101182. [PMID: 24983622 PMCID: PMC4077744 DOI: 10.1371/journal.pone.0101182] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 06/03/2014] [Indexed: 12/26/2022] Open
Abstract
The STAT3 transcription factor is a pleiotropic transducer of signalling by hormones, growth factors and cytokines that has been identified in the female reproductive tract from oocytes and granulosa cells of the ovary to uterine epithelial and stromal cells. In the present study we used transgenic models to investigate the importance of STAT3 for reproductive performance in these different tissues. The Cre-LoxP system was used to delete STAT3 in oocytes by crossing Stat3fl/fl with Zp3-cre+ mice, or in ovarian granulosa cells and uterine stroma by crossing with Amhr2-Cre+ mice. Surprisingly, deletion of STAT3 in oocytes had no effect on fertility indicating that the abundance of STAT3 protein in maturing oocytes and fertilized zygotes is not essential to these developmental stages. In Stat3fl/fl;Amhr2-cre+ females impaired fertility was observed through significantly fewer litters and smaller litter size. Ovulation rate, oocyte fertilization and development to blastocyst were unaffected in this line; however, poor recombination efficiency in granulosa cells had yielded no net change in STAT3 protein abundance. In contrast, uteri from these mice showed STAT3 protein depletion selectively from the stomal compartment. A significant reduction in number of viable fetuses on gestational day 18, increased fetal resorptions and disrupted placental morphology were evident causes of the reduced fertility. In conclusion, this study defines an important role for STAT3 in uterine stromal cells during embryo implantation and the development of a functional placenta.
Collapse
|
46
|
Di Emidio G, Falone S, Vitti M, D'Alessandro AM, Vento M, Di Pietro C, Amicarelli F, Tatone C. SIRT1 signalling protects mouse oocytes against oxidative stress and is deregulated during aging. Hum Reprod 2014; 29:2006-17. [PMID: 24963165 DOI: 10.1093/humrep/deu160] [Citation(s) in RCA: 144] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
STUDY QUESTION Is SIRT1 involved in the oxidative stress (OS) response in mouse oocytes? SUMMARY ANSWER SIRT1 plays a pivotal role in the adaptive response of mouse germinal vesicle (GV) oocytes to OS and promotes a signalling cascade leading to up-regulation of the MnSod gene. WHAT IS KNOWN ALREADY OS is known to continuously threaten acquisition and maintenance of oocyte developmental potential during in vivo processes and in vitro manipulations. Previous studies in somatic cells have provided strong evidence for the role of SIRT1 as a sensor of the cell redox state and a protector against OS and aging. STUDY DESIGN, SIZE, DURATION GV oocytes obtained from young (4-8 weeks) and reproductively old (48-52 weeks) CD1 mice were blocked in the prophase stage by 0.5 µM cilostamide. Groups of 30 oocytes were exposed to 25 µM H2O2 and processed following different times for the analysis of intracellular localization of SIRT1 and FOXO3A, and evaluation of Sirt1, miRNA-132, FoxO3a and MnSod gene expression. Another set of oocytes was cultured in the presence or absence of the SIRT1-specific inhibitor Ex527, and exposed to H2O2 in order to assess the involvement of SIRT1 in the activation of a FoxO3a-MnSod axis and ROS detoxification. In the last part of this study, GV oocytes were maturated in vitro in the presence of different Ex527 concentrations (0, 2.5, 5, 10, 20 µM) and assessed for maturation rates following 16 h. Effects of Ex527 on spindle morphology and ROS levels were also evaluated. PARTICIPANTS/MATERIALS, SETTING, METHODS SIRT1 and FOXO3A intracellular distribution in response to OS was investigated by immunocytochemistry. Real-time RT-PCR was employed to analyse Sirt1, miR-132, FoxO3a and MnSod gene expression. Reactive oxygen species (ROS) production was evaluated by in vivo measurement of carboxy-H2DCF diacetate labelling. Spindle and chromosomal distribution in in vitro matured oocytes were analysed by immunocytochemistry and DNA fluorescent labelling, respectively. MAIN RESULTS AND THE ROLE OF CHANCE Specific changes in the intracellular localization of SIRT1 and up-regulation of Sirt1 gene were detected in mouse oocytes in response to OS. Moreover, increased intracellular ROS were observed when SIRT1 activity was inhibited by Ex527. In aged oocytes Sirt1 was expressed more than in young oocytes but SIRT1 protein was undetectable. Upon OS, significant changes in miR-132 micro-RNA, a validated Sirt1 modulator, were observed. A negative correlation between Sirt1 mRNA and miR-132 levels was observed when young oocytes exposed to OS were compared with young control oocytes, and when aged oocytes were compared with young control oocytes. FoxO3a and MnSod transcripts were increased upon OS with the same kinetics as Sirt1 transcripts, and up-regulation of MnSod gene was prevented by oocyte treatment with Ex527, indicating that SIRT1 acts upstream to the FoxO3a-MnSod axis. Finally, the results of the in vitro maturation assay suggested that SIRT1 might be involved in oocyte maturation by regulating the redox state and ensuring normal spindle assembly. LIMITATIONS, REASONS FOR CAUTION The main limitation of this study was the absence of direct quantification of SIRT1 enzymatic activity due to the lack of an appropriately sensitive method. WIDER IMPLICATIONS OF THE FINDINGS The present findings may provide a valuable background for studying the regulation of SIRT1 during oogenesis and its relevance as a sensor of oocyte redox state and energy status. The antioxidant response orchestrated by SIRT1 in oocytes seems to decrease with aging. This suggests that SIRT1 could be an excellent pharmacological target for improving oocyte quality and IVF outcome in aging or aging-like diseases. STUDY FUNDING/COMPETING INTERESTS The work was supported by the Ministero dell'Università e della Ricerca Scientifica (MIUR) to C.T., F.A., C.D., A.M.D. The authors declare no conflict of interest.
Collapse
Affiliation(s)
- Giovanna Di Emidio
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Via Vetoio, 67100 L'Aquila, Italy
| | - Stefano Falone
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Via Vetoio, 67100 L'Aquila, Italy
| | - Maurizio Vitti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Via Vetoio, 67100 L'Aquila, Italy
| | - Anna Maria D'Alessandro
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Via Vetoio, 67100 L'Aquila, Italy
| | - Marilena Vento
- Servizio di PMA, Azienda Ospedaliera Cannizzaro, Catania, Italy
| | - Cinzia Di Pietro
- Dipartimento Gian Filippo Ingrassia, Sezione di Biologia, Genetica, Genomica Cellulare e Molecolare Giovanni Sichel, Università degli Studi di Catania, Catania, Italy
| | - Fernanda Amicarelli
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Via Vetoio, 67100 L'Aquila, Italy
| | - Carla Tatone
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Via Vetoio, 67100 L'Aquila, Italy Infertility Service, San Salvatore Hospital, Via Vetoio, 67100 L'Aquila, Italy
| |
Collapse
|
47
|
Kashir J, Deguchi R, Jones C, Coward K, Stricker SA. Comparative biology of sperm factors and fertilization-induced calcium signals across the animal kingdom. Mol Reprod Dev 2013; 80:787-815. [PMID: 23900730 DOI: 10.1002/mrd.22222] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 07/23/2013] [Indexed: 11/08/2022]
Abstract
Fertilization causes mature oocytes or eggs to increase their concentrations of intracellular calcium ions (Ca²⁺) in all animals that have been examined, and such Ca²⁺ elevations, in turn, provide key activating signals that are required for non-parthenogenetic development. Several lines of evidence indicate that the Ca²⁺ transients produced during fertilization in mammals and other taxa are triggered by soluble factors that sperm deliver into oocytes after gamete fusion. Thus, for a broad-based analysis of Ca²⁺ dynamics during fertilization in animals, this article begins by summarizing data on soluble sperm factors in non-mammalian species, and subsequently reviews various topics related to a sperm-specific phospholipase C, called PLCζ, which is believed to be the predominant activator of mammalian oocytes. After characterizing initiation processes that involve sperm factors or alternative triggering mechanisms, the spatiotemporal patterns of Ca²⁺ signals in fertilized oocytes or eggs are compared in a taxon-by-taxon manner, and broadly classified as either a single major transient or a series of repetitive oscillations. Both solitary and oscillatory types of fertilization-induced Ca²⁺ signals are typically propagated as global waves that depend on Ca²⁺ release from the endoplasmic reticulum in response to increased concentrations of inositol 1,4,5-trisphosphate (IP₃). Thus, for taxa where relevant data are available, upstream pathways that elevate intraoocytic IP3 levels during fertilization are described, while other less-common modes of producing Ca²⁺ transients are also examined. In addition, the importance of fertilization-induced Ca²⁺ signals for activating development is underscored by noting some major downstream effects of these signals in various animals.
Collapse
Affiliation(s)
- Junaid Kashir
- Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Level 3, Women's Centre, John Radcliffe Hospital, Headington, Oxford, UK
| | | | | | | | | |
Collapse
|
48
|
Ma JY, Li M, Luo YB, Song S, Tian D, Yang J, Zhang B, Hou Y, Schatten H, Liu Z, Sun QY. Maternal factors required for oocyte developmental competence in mice: transcriptome analysis of non-surrounded nucleolus (NSN) and surrounded nucleolus (SN) oocytes. Cell Cycle 2013; 12:1928-38. [PMID: 23673344 DOI: 10.4161/cc.24991] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
During mouse antral follicle development, the oocyte chromatin gradually transforms from a less condensed state with no Hoechst-positive rim surrounding the nucleolus (NSN) to a fully condensed chromatin state with a Hoechst-positive rim surrounding the nucleolus (SN). Compared with SN oocytes, NSN oocytes display a higher gene transcription activity and a lower rate of meiosis resumption (G2/M transition), and they are mostly arrested at the two-cell stage after in vitro fertilization. To explore the differences between NSN and SN oocytes, and the maternal factors required for oocyte developmental competence, we compared the whole-transcriptome profiles between NSN and SN oocytes. First, we found that the NSN and SN oocytes were different in their metabolic pathways. In the phosphatidylinositol signaling pathway, the SN oocytes tend to produce diacylglycerol, whereas the NSN oocytes tend to produce phosphatidylinositol (3,4,5)-trisphosphate. For energy production, the SN oocytes and NSN oocytes differed in the gluconeogenesis and in the synthesis processes. Second, we also found that the key genes associated with oocyte meiosis and/or preimplantation embryo development were differently expressed in the NSN and SN oocytes. Our results illustrate that during the NSN-SN transition, the oocytes change their metabolic activities and accumulate maternal factors for further oocyte maturation and post-fertilization embryo development.
Collapse
Affiliation(s)
- Jun-Yu Ma
- College of Life Science, Northeast Agricultural University, Harbin, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|