51
|
Takahashi T, Ogiwara K. Signal pathway of LH-induced expression of nuclear progestin receptor in vertebrate ovulation. Gen Comp Endocrinol 2022; 321-322:114025. [PMID: 35292264 DOI: 10.1016/j.ygcen.2022.114025] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 02/18/2022] [Accepted: 03/10/2022] [Indexed: 11/19/2022]
Abstract
Nuclear progestin receptor (PGR), which is induced in the follicles destined to undergo ovulation, is believed to be obligatory for rupture of the follicles during ovulation in vertebrates. Studies in some mammals and teleost medaka have revealed the outline of the central signaling pathway that leads to the PGR expression in the preovulatory follicles at ovulation. In this review, we summarize the current knowledge on what signaling mediators are involved in the LH-induced follicular expression of PGR at ovulation in these animals. LH-inducibility of follicular PGR expression is conserved. In both group of animals, activation of the LH receptor on the granulosa cell surface with LH commonly results in the increase of intracellular cAMP levels, while the downstream signaling cascades activated by high level of cAMP are totally different between mice and medaka. PGR is currently presumed to be induced via PKA/CREB-mediated transactivation and ERK1/2-dependent signaling in mice, but the receptor is induced via EPAC/RAP and AKT/CREB pathways in the teleost medaka. The differences and similarities in the signaling pathways for PGR expression between them is discussed from comparative and evolutionary aspects. We also discussed questions concerning PGR expression and its regulation needed to be investigated in future.
Collapse
Affiliation(s)
- Takayuki Takahashi
- Laboratory of Reproductive and Developmental Biology, Faculty of Science, Hokkaido University, Sapporo 060-0810, Japan.
| | - Katsueki Ogiwara
- Laboratory of Reproductive and Developmental Biology, Faculty of Science, Hokkaido University, Sapporo 060-0810, Japan
| |
Collapse
|
52
|
Marchais M, Gilbert I, Bastien A, Macaulay A, Robert C. Mammalian cumulus-oocyte complex communication: a dialog through long and short distance messaging. J Assist Reprod Genet 2022; 39:1011-1025. [PMID: 35499777 PMCID: PMC9107539 DOI: 10.1007/s10815-022-02438-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 02/13/2022] [Indexed: 12/13/2022] Open
Abstract
Communications are crucial to ovarian follicle development and to ovulation, and while both folliculogenesis and oogenesis are distinct processes, they share highly interdependent signaling pathways. Signals from distant organs such as the brain must be processed and compartments within the follicle have to be synchronized. The hypothalamic–pituitary–gonadal (HPG) axis relies on long-distance signalling analogous to wireless communication by which data is disseminated in the environment and cells equipped with the appropriate receptors receive and interpret the messages. In contrast, direct cell-to-cell transfer of molecules is a very targeted, short distance messaging system. Numerous signalling pathways have been identified and proven to be essential for the production of a developmentally competent egg. The development of the cumulus-oocyte complex relies largely on short distance communications or direct transfer type via extensions of corona radiata cells through the zona pellucida. The type of information transmitted through these transzonal projections is still largely uncharacterized. This review provides an overview of current understanding of the mechanisms by which the gamete receives and transmits information within the follicle. Moreover, it highlights the fact that in addition to the well-known systemic long-distance based communications from the HPG axis, these mechanisms acting more locally should also be considered as important targets for controlling/optimizing oocyte quality.
Collapse
Affiliation(s)
- Mathilde Marchais
- Département des sciences animales, Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle (CRDSI), Réseau Québécois en Reproduction (RQR), Pavillon Paul Comtois, Université Laval, Québec, QC, Canada
| | - Isabelle Gilbert
- Département des sciences animales, Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle (CRDSI), Réseau Québécois en Reproduction (RQR), Pavillon Paul Comtois, Université Laval, Québec, QC, Canada
| | - Alexandre Bastien
- Département des sciences animales, Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle (CRDSI), Réseau Québécois en Reproduction (RQR), Pavillon Paul Comtois, Université Laval, Québec, QC, Canada
| | - Angus Macaulay
- Département des sciences animales, Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle (CRDSI), Réseau Québécois en Reproduction (RQR), Pavillon Paul Comtois, Université Laval, Québec, QC, Canada
| | - Claude Robert
- Département des sciences animales, Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle (CRDSI), Réseau Québécois en Reproduction (RQR), Pavillon Paul Comtois, Université Laval, Québec, QC, Canada.
| |
Collapse
|
53
|
The Molecular Quality and Mitochondrial Activity of Porcine Cumulus-Oocyte Complexes Are Affected by Their Exposure to Three Endocrine-Active Compounds under 3D In Vitro Maturation Conditions. Int J Mol Sci 2022; 23:ijms23094572. [PMID: 35562963 PMCID: PMC9100547 DOI: 10.3390/ijms23094572] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/12/2022] [Accepted: 04/19/2022] [Indexed: 12/15/2022] Open
Abstract
Thus far, the potential short- and long-term detrimental effects of a variety of environmental chemicals designated as endocrine-active compounds (EACs) have been found to interfere with histo- and anatomo-physiological functions of the reproductive system in humans and wildlife species. For those reasons, this study sought to examine whether selected EACs, which encompass the fungicide vinclozolin (Vnz), the androgenic anabolic steroid nandrolone (Ndn) and the immunosuppressant cyclosporin A (CsA), affect the developmental competence and molecular quality (MQ) of porcine cumulus–oocyte complexes (COCs) subjected to in vitro maturation (IVM) under 3D culture conditions. The COCs underwent 3D-IVM in the presence of Vnz, Ndn or CsA for 48 h. To explore whether the selected EACs induce internucleosomal DNA fragmentation in cumulus cells (CCs), TUNEL-assisted detection of late apoptotic cells was performed. Additionally, for the detailed evaluation of pro- and antiapoptotic pathways in COCs, apoptosis proteome profiler arrays were used. To determine changes in intracellular metabolism in COCs, comprehensive assessments of mitochondrial ultrastructure and activity were carried out. Moreover, the relative abundances (RAs) of mRNAs transcribed from genes that are involved in scavenging reactive oxygen species (ROS), such as SIRT3 and FOXO3, and intramitochondrial bioenergetic balance, such as ATP synthase subunit (ATP5A1), were ascertained. Finally, to investigate the extent of progression of oocyte maturation, the intraooplasmic levels of cAMP and the RAs of mRNA transcripts encoding regulatory and biocatalytic subunits of a heterodimeric meiosis-promoting factor, termed cyclin B1 (CCNB1) and cyclin-dependent kinase 1 (CDC2), were also estimated. The obtained results provide, for the first time, strong evidence that both Vnz and Ndn decrease the developmental competence of oocytes and stimulate apoptosis processes in CCs. The present study is also the first to highlight that Vnz accelerates the maturation process in immature oocytes due to both increased ROS production and the augmented RA of the CCNB1 gene. Furthermore, Vnz was proven to trigger proapoptotic events in CCs by prompting the activity of the FOXO3 transcription factor, which regulates the mitochondrial apoptosis pathway. In turn, Ndn was shown to inhibit oocyte maturation by inducing molecular events that ultimately lead to an increase in the intraooplasmic cAMP concentration. However, due to the simultaneous enhancement of the expression of TNF-β and HSP27 proteins in CCs, Ndn might be responsible for the onset of their neoplastic transformation. Finally, our current investigation is the first to clearly demonstrate that although CsA did not interfere with the nuclear and cytoplasmic maturation of oocytes, by inducing mitophagy in CCs, it disrupted oocyte metabolism, consequently attenuating the parameters related to the MQ of COCs. Summing up, Vnz, Ndn and CsA reduced not only the processes of growth and IVM but also the MQ of porcine COCs, which might make them unsuitable for assisted reproductive technologies (ARTs) such as in vitro fertilization by either gamete co-incubation or intracytoplasmic sperm injection (ICSI) and cloning by somatic cell nuclear transfer (SCNT).
Collapse
|
54
|
Molecular determinants regulating the release of the egg during ovulation: Perspectives in piscine models. AQUACULTURE AND FISHERIES 2022. [DOI: 10.1016/j.aaf.2022.03.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
55
|
Yamazaki W, Tan SL, Taketo T. Role of the X and Y Chromosomes in the Female Germ Cell Line Development in the Mouse (Mus musculus). Sex Dev 2022:1-10. [PMID: 35235936 DOI: 10.1159/000521151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 11/18/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND In eutherian mammals, the sex chromosome complement, XX and XY, determines sexual differentiation of gonadal primordia into testes and ovaries, which in turn direct differentiation of germ cells into haploid sperm and oocytes, respectively. When gonadal sex is reversed, however, the germ cell sex becomes discordant with the chromosomal sex. XY females in humans are infertile, while XY females in the mouse (Mus musculus) are subfertile or infertile dependent on the cause of sex reversal and the genetic background. This article reviews publications to understand how the sex chromosome complement affects the fertility of XY oocytes by comparing with XX and monosomy X (XO) oocytes. SUMMARY The results highlight 2 folds disadvantage of XY oocytes over XX oocytes: (1) the X and Y chromosomes fail to pair during the meiotic prophase I, resulting in sex chromosome aneuploidy at the first meiotic division and (2) expression of the Y-linked genes during oocyte growth affects the transcriptome landscape and renders the ooplasmic component incompetent for embryonic development. Key Message: The XX chromosome complement gives the oocyte the highest competence for embryonic development.
Collapse
Affiliation(s)
- Wataru Yamazaki
- Department of Surgery, McGill University, Montreal, Québec, Canada.,Research Institute of McGill University Health Centre, Montreal, Québec, Canada
| | - Seang Lin Tan
- Department of Obstetrics and Gynecology, McGill University, Montreal, Québec, Canada.,Research Institute of McGill University Health Centre, Montreal, Québec, Canada.,OriginElle Fertility Clinic and Women's Health Centre, Montreal, Québec, Canada
| | - Teruko Taketo
- Department of Surgery, McGill University, Montreal, Québec, Canada.,Department of Obstetrics and Gynecology, McGill University, Montreal, Québec, Canada.,Department of Biology, McGill University, Montreal, Québec, Canada.,Research Institute of McGill University Health Centre, Montreal, Québec, Canada
| |
Collapse
|
56
|
Buratini J, Soares ACS, Barros RG, Dellaqua TT, Lodde V, Franciosi F, Dal Canto M, Renzini MM, Luciano AM. Physiological parameters related to oocyte nuclear differentiation for the improvement of IVM/IVF outcomes in women and cattle. Reprod Fertil Dev 2022; 34:27-35. [PMID: 35231269 DOI: 10.1071/rd21278] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
In vitro maturation (IVM) has been applied in numerous different contexts and strategies in humans and animals, but in both cases it represents a challenge still far from being overcome. Despite the large dataset produced over the last two decades on the mechanisms that govern antral follicular development and oocyte metabolism and differentiation, IVM outcomes are still unsatisfactory. This review specifically focuses on data concerning the potential consequences of using supraphysiological levels of FSH during IVM, as well as on the regulation of oocyte chromatin dynamics and its utility as a potential marker of oocyte developmental competence. Taken together, the data revisited herein indicate that a significant improvement in IVM efficacy may be provided by the integration of pre-OPU patient-specific protocols preparing the oocyte population for IVM and more physiological culture systems mimicking more precisely the follicular environment that would be experienced by the recovered oocytes until completion of metaphase II.
Collapse
Affiliation(s)
- Jose Buratini
- Biogenesi Reproductive Medicine Centre - Eugin Group, Istituti Clinici Zucchi, Monza, Italy; and Department of Structural and Functional Biology, Sao Paulo State University, Botucatu, Brazil
| | | | - Rodrigo Garcia Barros
- Reproductive and Developmental Biology Laboratory, Department of Health, Animal Science and Food Safety, University of Milan, Milan, Italy
| | - Thaisy Tino Dellaqua
- Department of Structural and Functional Biology, Sao Paulo State University, Botucatu, Brazil
| | - Valentina Lodde
- Reproductive and Developmental Biology Laboratory, Department of Health, Animal Science and Food Safety, University of Milan, Milan, Italy
| | - Federica Franciosi
- Reproductive and Developmental Biology Laboratory, Department of Health, Animal Science and Food Safety, University of Milan, Milan, Italy
| | | | - Mario Mignini Renzini
- Biogenesi Reproductive Medicine Centre - Eugin Group, Istituti Clinici Zucchi, Monza, Italy
| | - Alberto Maria Luciano
- Reproductive and Developmental Biology Laboratory, Department of Health, Animal Science and Food Safety, University of Milan, Milan, Italy
| |
Collapse
|
57
|
IMD/ADM2 operates as a secretory factor that controls cumulus-oocyte complexes (COCs) conformation for oocytes in vitro maturation. In Vitro Cell Dev Biol Anim 2022; 58:149-168. [PMID: 35102467 DOI: 10.1007/s11626-022-00647-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 12/28/2021] [Indexed: 12/13/2022]
Abstract
During in vitro maturation (IVM), the compact structure of cumulus-oocyte complexes (COCs) is vital for oocyte competence acquisition. Intermedin/Adrenomedullin-2 (IMD/ADM2) binds to the receptor RAMP (1, 2, or 3):CLR. Recently, it was demonstrated that IMD/ADM2 stimulates oocyte competence and improves bovine embryo quality. Therefore, this study aimed to examine the IMD/ADM2 as a secretory factor controlling COCs conformation for oocyte maturation. The results showed that traditional M-CDM medium induced in COCs the Imd/Adm2 gene expression during IVM and produced IMD/ADM2 peptide secretion. Furthermore, after IVM, in the oocytes, the expression of ramps (1, 2, or 3) and clr was demolished, and RAMPs and CLR proteins were decreased, with a negative Pearson correlation. These results suggest that RAMPs and CLR are synthesized and stored during oocyte maturation. Supplementing the M-CDM with α-RAMP1 or α-IMD/ADM2 antibodies elicits a negative effect (P < 0.05) in COCs compaction. Blocking the IMD/ADM2 signaling pathway with any α-RAMPs or α-CLR antibodies produces a similar lower yield of oocytes in metaphase II (P > 0.05) but was lower than control culture medium (P < 0.05). In conclusion, when COCs are cultured with M-CDM, the IMD/ADM2 becomes expressed and secreted. In turn, it acts as a ligand preferentially to RAMP1:CLR or RAMP3:CLR, present in cumulus cells and oocytes. Sequentially, COCs compact structure is conformed to promote an adequate bidirectional communication that conduces the oocytes' maturation.
Collapse
|
58
|
Krisher RL. Present state and future outlook for the application of in vitro oocyte maturation (IVM) in human infertility treatment. Biol Reprod 2022; 106:235-242. [DOI: 10.1093/biolre/ioac010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Abstract
In vitro oocyte maturation (IVM) is an assisted reproductive technology in which a meiotically immature oocyte (prophase I or germinal vesicle stage) is recovered from an antral follicle and matured in vitro prior to fertilization. This technology, although in widespread use in domestic livestock, is not typically implemented during human IVF cycles. This review examines how IVM is currently used in the clinical setting, including the various ways IVM is defined in practice. The role of IVM in patient care, and the major challenges for implementation are described. Efficiency and safety are critically explored. The role of IVM in oncofertility will also be discussed. Finally, the outlook for the future of clinical IVM is considered.
Collapse
|
59
|
Buratini J, Dellaqua TT, Dal Canto M, La Marca A, Carone D, Mignini Renzini M, Webb R. The putative roles of FSH and AMH in the regulation of oocyte developmental competence: from fertility prognosis to mechanisms underlying age-related subfertility. Hum Reprod Update 2021; 28:232-254. [PMID: 34969065 DOI: 10.1093/humupd/dmab044] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 11/18/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Fertility loss during female ageing is associated with increasing basal FSH and decreasing anti-Müllerian hormone (AMH) concentrations, together with compromised oocyte quality, presumably due to increased oxidative stress (OS) and DNA damage, as well as reduced metabolic and meiotic competences. Basal FSH and AMH circulatory concentrations have been broadly utilized as IVF success predictors, regardless of fluctuations in prognostic accuracy; basal FSH and AMH perform better in pre-advanced maternal age (AMA: >35 years) and AMA patients, respectively. The relationships between FSH and AMH intrafollicular levels and IVF outcomes suggest, nevertheless, that both hormones regulate oocyte competence, supporting the hypothesis that changes in FSH/AMH levels cause, at least in part, oocyte quality degradation during ageing. To understand the reasons behind the fluctuations in FSH and AMH prognostic accuracies and to clarify their participation in mechanisms determining oocyte competence and age-related subfertility, a deeper knowledge of the regulation of FSH and AMH intrafollicular signalling during the female reproductive lifespan, and of their effects on the cumulus-oocyte complex, is required. OBJECTIVE AND RATIONALE An extensive body of information on the regulation of FSH and AMH intrafollicular availability and signalling, as well as on the control of folliculogenesis and oocyte metabolism, has been accumulated. However, these datasets have been explored within the relatively narrow boundaries of their specific subjects. Given the aforementioned gaps in knowledge and their clinical relevance, herein we integrate clinical and basic data, within a wide biological perspective, aiming to shed light on (i) the reasons for the variability in the accuracy of serum FSH and AMH as fertility markers, and on (ii) the potential roles of these hormones in mechanisms regulating oocyte quality, particularly those associated with ageing. SEARCH METHODS The PubMed database encompassing the period between 1960 and 2021 was searched. Principal search terms were FSH, FSH receptor, AMH, oocyte, maternal age, cumulus, transzonal projections (TZPs), actin, OS, redox, reactive oxygen species, mitochondria, DNA damage, DNA repair, aneuploidy, spindle, meiosis, gene expression, transcription, translation, oocyte secreted factors (OSFs), cAMP, cyclic guanosine monophosphate, natriuretic peptide C, growth differentiation factor 9, bone morphogenetic protein 15 and fibroblast growth factor. OUTCOMES Our analysis suggests that variations in the accuracy of fertility prognosis reflect a modest association between circulatory AMH levels and oocyte quality as well as increasing basal FSH inter-cycle variability with age. In addition, the basic and clinical data articulated herein support the hypothesis that increased intrafollicular FSH levels, as maternal age advances, may override the physiological protective influences of AMH and OSFs against excessive FSH signalling in cumulus cells. This would result in the disruption of oocyte homeostasis via reduced TZP-mediated transfer of cumulus-derived molecules essential for meiotic competence, gene expression, redox activity and DNA repair. WIDER IMPLICATIONS In-depth data analysis, encompassing a wide biological perspective has revealed potential causative mechanisms of age-related subfertility triggered by alterations in FSH/AMH signalling during the female reproductive life. Insights from new mechanistic models arising from this analysis should contribute to advancing our comprehension of oocyte biology in humans and serve as a valuable reference for novel AMA subfertility treatments aimed at improving oocyte quality through the modulation of AMH/FSH action.
Collapse
Affiliation(s)
- Jose Buratini
- Biogenesi Reproductive Medicine Centre-Eugin Group, Istituti Clinici Zucchi, Monza, Italy.,Clinica Eugin Modena, Modena, Italy.,Department of Structural and Functional Biology, Sao Paulo State University, Botucatu, Brazil
| | - Thaisy Tino Dellaqua
- Department of Structural and Functional Biology, Sao Paulo State University, Botucatu, Brazil
| | - Mariabeatrice Dal Canto
- Biogenesi Reproductive Medicine Centre-Eugin Group, Istituti Clinici Zucchi, Monza, Italy.,Clinica Eugin Modena, Modena, Italy
| | - Antonio La Marca
- Clinica Eugin Modena, Modena, Italy.,Department of Medical and Surgical Sciences of the Mother, Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Mario Mignini Renzini
- Biogenesi Reproductive Medicine Centre-Eugin Group, Istituti Clinici Zucchi, Monza, Italy.,Clinica Eugin Modena, Modena, Italy
| | - Robert Webb
- Division of Animal Sciences, School of Biosciences, University of Nottingham, Nottinghamshire, UK
| |
Collapse
|
60
|
Sitruk-Ware R. [New frontiers in contraception research]. Med Sci (Paris) 2021; 37:1014-1020. [PMID: 34851278 DOI: 10.1051/medsci/2021163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Improving current contraceptives and discover novel methods easy to use with added health benefits would meet the needs of couples who seek alternatives to current methods. New delivery systems target user-controlled, longer-acting options to provide choice, user's autonomy and improve compliance. Self-injections, microarray patches, pod rings able to deliver several molecules aim to prevent both pregnancies and sexually transmitted infections. Improved intrauterine systems and non-surgical permanent methods are also on the research agenda. The search for novel methods must continue, to curb maternal mortality led by multiple pregnancies and unsafe abortion, still a burden in many countries.
Collapse
Affiliation(s)
- Régine Sitruk-Ware
- The Population Council, Center for Biomedical Research, 1230 York Avenue, New York, NY 10065, États-Unis
| |
Collapse
|
61
|
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
|
62
|
Tesfaye D, Menjivar N, Gebremedhn S. Current knowledge and the future potential of extracellular vesicles in mammalian reproduction. Reprod Fertil Dev 2021; 34:174-189. [PMID: 35231266 DOI: 10.1071/rd21277] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Extracellular vesicles (EVs), which contain various functional classes of vesicles, namely exosomes, microvesicles, and apoptotic bodies, represent the major nano-shuttle to transfer bioactive molecules from donor to recipient cells to facilitate cell-to-cell communication in the follicular, oviduct, and uterine microenvironments. In addition to transferring various molecular cargos in the form of miRNAs, mRNAs, proteins, lipids, and DNA molecules, the relative proportion of those molecular cargos in the reproductive fluids can be associated with the physiological and pathological condition of the host animal. Inside the follicle, EV-mediated circulation of miRNAs has been reported to be associated with the growth status of the enclosed oocytes, the metabolic status, and the advanced maternal aging of the animal. Importantly, EVs have the potential to protect their cargo molecules from extracellular degradation or modification while travelling to the recipient cells. This fact together with the enormous availability in almost all biological fluids and spent culture media make them attractive in the search for biomarkers of oocyte/embryo developmental competence, receptive maternal environment and a multitude of reproductive pathophysiological conditions. One of the key factors that have contributed to the lower efficiency of assisted reproductive technologies (ART) is the absence of several maternal in vivo factors in the ART procedures. For this, several studies have been conducted to supplement various components present in the follicular and oviductal fluids into the existing ART procedures and significant positive impacts have been observed in terms of embryo cleavage rate, blastocyst rate, resistance to stress, and survival after cryopreservation. The potential of EVs in shuttling protective messages against environmental and physiological stressors has been evidenced. The effective use of the EV-coupled molecular signals against stress-associated conditions has the potential to pave the path for the application of these protective signals against oxidative stress-associated pathological conditions including PCOS, ageing, and endometritis. In this review, we provide current knowledge and potential future use of EVs as remedies in reproductive pathophysiological conditions, mainly in follicular and oviductal microenvironments.
Collapse
Affiliation(s)
- Dawit Tesfaye
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, Colorado State University, 3051 Rampart Road, Fort Collins, CO 80521, USA
| | - Nico Menjivar
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, Colorado State University, 3051 Rampart Road, Fort Collins, CO 80521, USA
| | | |
Collapse
|
63
|
Compromised Cumulus-Oocyte Complex Matrix Organization and Expansion in Women with PCOS. Reprod Sci 2021; 29:836-848. [PMID: 34748173 DOI: 10.1007/s43032-021-00775-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 10/14/2021] [Indexed: 10/19/2022]
Abstract
The cumulus-oocyte complex (COC) matrix plays a critical role in the ovulation and fertilization process and a major predictor of oocyte quality. Proteomics studies of follicular fluid showed differential expression of COC matrix proteins in women with polycystic ovary syndrome (PCOS), indicating altered COC matrix in these women. In the present study, we aimed to understand COC matrix gene induction in humans and its probable dysfunction in women with PCOS. Animal studies have shown that amphiregulin (AREG) and growth differentiation factor-9 (GDF-9) are important in the induction of COC matrix genes which are involved in cumulus expansion. The effects of AREG and GDF-9 on expression of tumor necrosis factor alpha induced protein 6 (TNFAIP6) and hyaluronan synthase 2 (HAS2) on human cumulus granulosa cells (CGCs) and murine COC expansion were evaluated. Further time-dependent effects of growth factor supplementation on these gene expressions in CGCs from PCOS and control women were compared. Follicular fluid from PCOS showed reduced COC matrix expansion capacity, using murine COCs. Expression of COC matrix genes TNFAIP6 and HAS2 were significantly reduced in CGCs of PCOS. Treatment of CGCs with AREG and GDF-9 together induced expression of both these genes in controls and could only restore HAS2 but not TNFAIP6 expression in PCOS. Our results suggest that the reduced potential of follicular fluid to support COC expansion, altered expression of structural constituents, and intrinsic defects in granulosa cells of women with PCOS may contribute to the aberrant COC organization and expansion in PCOS, thus affecting fertilization.
Collapse
|
64
|
Uzbekova S, Teixeira-Gomes AP, Marestaing A, Jarrier-Gaillard P, Papillier P, Shedova EN, Singina GN, Uzbekov R, Labas V. Protein Palmitoylation in Bovine Ovarian Follicle. Int J Mol Sci 2021; 22:ijms222111757. [PMID: 34769186 PMCID: PMC8583988 DOI: 10.3390/ijms222111757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 11/16/2022] Open
Abstract
Protein palmitoylation is a reversible post-translational modification by fatty acids (FA), mainly a palmitate (C16:0). Palmitoylation allows protein shuttling between the plasma membrane and cytosol to regulate protein stability, sorting and signaling activity and its deficiency leads to diseases. We aimed to characterize the palmitoyl-proteome of ovarian follicular cells and molecular machinery regulating protein palmitoylation within the follicle. For the first time, 84 palmitoylated proteins were identified from bovine granulosa cells (GC), cumulus cells (CC) and oocytes by acyl-biotin exchange proteomics. Of these, 32 were transmembrane proteins and 27 proteins were detected in bovine follicular fluid extracellular vesicles (ffEVs). Expression of palmitoylation and depalmitoylation enzymes as palmitoyltransferases (ZDHHCs), acylthioesterases (LYPLA1 and LYPLA2) and palmitoylthioesterases (PPT1 and PPT2) were analysed using transcriptome and proteome data in oocytes, CC and GC. By immunofluorescence, ZDHHC16, PPT1, PPT2 and LYPLA2 proteins were localized in GC, CC and oocyte. In oocyte and CC, abundance of palmitoylation-related enzymes significantly varied during oocyte maturation. These variations and the involvement of identified palmitoyl-proteins in oxidation-reduction processes, energy metabolism, protein localization, vesicle-mediated transport, response to stress, G-protein mediated and other signaling pathways suggests that protein palmitoylation may play important roles in oocyte maturation and ffEV-mediated communications within the follicle.
Collapse
Affiliation(s)
- Svetlana Uzbekova
- CNRS, IFCE, INRAE, Université de Tours, PRC, 37380 Nouzilly, France; (A.M.); (P.J.-G.); (P.P.); (V.L.)
- Correspondence: ; Tel.: +33-247-427-951
| | | | - Aurélie Marestaing
- CNRS, IFCE, INRAE, Université de Tours, PRC, 37380 Nouzilly, France; (A.M.); (P.J.-G.); (P.P.); (V.L.)
| | - Peggy Jarrier-Gaillard
- CNRS, IFCE, INRAE, Université de Tours, PRC, 37380 Nouzilly, France; (A.M.); (P.J.-G.); (P.P.); (V.L.)
| | - Pascal Papillier
- CNRS, IFCE, INRAE, Université de Tours, PRC, 37380 Nouzilly, France; (A.M.); (P.J.-G.); (P.P.); (V.L.)
| | - Ekaterina N. Shedova
- L.K. Ernst Federal Research Center for Animal Husbandry, Dubrovitzy 60, 142132 Podolsk, Russia; (E.N.S.); (G.N.S.)
| | - Galina N. Singina
- L.K. Ernst Federal Research Center for Animal Husbandry, Dubrovitzy 60, 142132 Podolsk, Russia; (E.N.S.); (G.N.S.)
| | - Rustem Uzbekov
- Laboratoire Biologie Cellulaire et Microscopie Électronique, Faculté de Médecine, Université de Tours, 37032 Tours, France;
| | - Valerie Labas
- CNRS, IFCE, INRAE, Université de Tours, PRC, 37380 Nouzilly, France; (A.M.); (P.J.-G.); (P.P.); (V.L.)
| |
Collapse
|
65
|
The Role of MAPK3/1 and AKT in the Acquisition of High Meiotic and Developmental Competence of Porcine Oocytes Cultured In Vitro in FLI Medium. Int J Mol Sci 2021; 22:ijms222011148. [PMID: 34681809 PMCID: PMC8537457 DOI: 10.3390/ijms222011148] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/12/2021] [Accepted: 10/14/2021] [Indexed: 12/27/2022] Open
Abstract
The developmental potential of porcine oocytes cultured in vitro was remarkably enhanced in a medium containing FGF2, LIF and IGF1 (FLI) when compared to a medium supplemented with gonadotropins and EGF (control). We analyzed the molecular background of the enhanced oocyte quality by comparing the time course of MAPK3/1 and AKT activation, and the expression of genes controlled by these kinases in cumulus-oocyte complexes (COCs) cultured in FLI and the control medium. The pattern of MAPK3/1 activation in COCs was very similar in both media, except for a robust increase in MAPK3/1 phosphorylation during the first hour of culture in the FLI medium. The COCs cultured in the FLI medium exhibited significantly higher activity of AKT than in the control medium from the beginning up to 16 h of culture; afterwards a deregulation of AKT activity occurred in the FLI medium, which was not observed in the control medium. The expression of cumulus cell genes controlled by both kinases was also modulated in the FLI medium, and in particular the genes related to cumulus-expansion, signaling, apoptosis, antioxidants, cell-to-cell communication, proliferation, and translation were significantly overexpressed. Collectively, these data indicate that both MAPK3/1 and AKT are implicated in the enhanced quality of oocytes cultured in FLI medium.
Collapse
|
66
|
Lu S, Zhang H, Tang Y, Xu R, Liu J, Yao R, Wei J, Li C, Zhao X, Wei Q, Ma B. G protein-coupled estrogen receptor signaling dependent epidermal growth-like factor expression is required for NPR2 inhibition and meiotic resumption in goat oocytes. Theriogenology 2021; 176:35-42. [PMID: 34571396 DOI: 10.1016/j.theriogenology.2021.09.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 09/13/2021] [Accepted: 09/15/2021] [Indexed: 01/12/2023]
Abstract
G protein-coupled estrogen receptor (GPER), which is different from traditional estrogen nuclear receptors (ERs), mediates the rapid transduction of nongenomic signals in cells, and works by regulating transcription and intracellular second messengers. Studies have shown that GPER may regulate oocyte maturation, but the relevant mechanism is not entirely clear. Here, goat cumulus-oocyte complexes (COCs) were used as a model to explore the regulation and mechanism of GPER on oocyte maturation. Our study showed that 17β-estradiol (E2) significantly reduced cyclic guanosine monophosphate (cGMP) synthesis in COCs and accelerated the meiotic resumption of goat oocytes via GPER. Further investigation found that GPER mediated the downregulation of natriuretic peptide receptor 2 (NPR2) protein expression in goat cumulus cells by E2. In addition, we found that E2 significantly upregulated the mRNA levels of epidermal growth (EGF)-like factors in goat cumulus cells through GPER, and activated the downstream EGF receptor (EGFR) and extracellular signal-regulated kinase 1/2 (ERK1/2) signaling pathways. Both AG1478 (EGFR inhibitor) and U0126 (ERK1/2 inhibitor) abolished the inhibitory effect of E2 on the protein expression of NPR2. These results indicate that, through GPER, E2 upregulates the mRNA levels of EGF-like factors in goat cumulus cells and activates the downstream EGF signaling network to suppress the expression of NPR2 protein, which results in a decrease in cGMP synthesis and acceleration of meiotic resumption in goat oocytes.
Collapse
Affiliation(s)
- Sihai Lu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, People's Republic of China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, People's Republic of China
| | - Hui Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, People's Republic of China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, People's Republic of China
| | - Yaju Tang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, People's Republic of China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, People's Republic of China
| | - Rui Xu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, People's Republic of China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, People's Republic of China
| | - Jie Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, People's Republic of China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, People's Republic of China
| | - Ru Yao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, People's Republic of China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, People's Republic of China
| | - Juncai Wei
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, People's Republic of China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, People's Republic of China
| | - Chan Li
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, People's Republic of China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, People's Republic of China
| | - Xiaoe Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, People's Republic of China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, People's Republic of China
| | - Qiang Wei
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, People's Republic of China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, People's Republic of China
| | - Baohua Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, People's Republic of China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, People's Republic of China.
| |
Collapse
|
67
|
Richani D, Gilchrist RB. Approaches to oocyte meiotic arrest in vitro and impact on oocyte developmental competence. Biol Reprod 2021; 106:243-252. [PMID: 34534265 DOI: 10.1093/biolre/ioab176] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 09/16/2021] [Indexed: 01/07/2023] Open
Abstract
Oocytes are maintained in a state of meiotic arrest following the first meiotic division until ovulation is triggered. Within the antral follicle, meiotic arrest is actively suppressed in a process facilitated by the cyclic nucleotides cGMP and cAMP. If removed from this inhibitory follicular environment and cultured in vitro, mammalian oocytes undergo spontaneous meiotic resumption in the absence of the usual stimulatory follicular stimuli, leading to asynchronicity with oocyte cytoplasmic maturation and lower developmental competence. For more than 50 years, pharmacological agents have been used to attenuate oocyte germinal vesicle (GV) breakdown in vitro. Agents which increase intra-oocyte cAMP or prevent its degradation have been predominantly used, however agents such as kinase and protein synthesis inhibitors have also been trialled. Twenty years of research demonstrates that maintaining GV arrest for a period before in vitro maturation (IVM) improves oocyte developmental competence, and is likely attributed to maintenance of bidirectional communication with cumulus cells leading to improved oocyte metabolic function. However, outcomes are influenced by various factors including the mode of action of the modulators, dose, treatment duration, species, and the degree of hormonal priming of the oocyte donor. Cyclic GMP and/or cAMP modulation in a prematuration step (called pre-IVM) prior to IVM has shown the greatest consistency in improving oocyte developmental competence, whereas kinase and protein synthesis inhibitors have proven less effective at improving IVM outcomes. Such pre-IVM approaches have shown potential to alter current use of artificial reproductive technologies in medical and veterinary practice.
Collapse
Affiliation(s)
- Dulama Richani
- Fertility & Research Centre, School of Women's & Children's Health, University of New South Wales Sydney, NSW 2052, Australia
| | - Robert B Gilchrist
- Fertility & Research Centre, School of Women's & Children's Health, University of New South Wales Sydney, NSW 2052, Australia
| |
Collapse
|
68
|
Sirait B, Wiweko B, Jusuf AA, Iftitah D, Muharam R. Oocyte Competence Biomarkers Associated With Oocyte Maturation: A Review. Front Cell Dev Biol 2021; 9:710292. [PMID: 34527670 PMCID: PMC8435600 DOI: 10.3389/fcell.2021.710292] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 08/10/2021] [Indexed: 01/20/2023] Open
Abstract
Oocyte developmental competence is one of the determining factors that influence the outcomes of an IVF cycle regarding the ability of a female gamete to reach maturation, be fertilized, and uphold an embryonic development up until the blastocyst stage. The current approach of assessing the competency of an oocyte is confined to an ambiguous and subjective oocyte morphological evaluation. Over the years, a myriad of biomarkers in the cumulus-oocyte-complex has been identified that could potentially function as molecular predictors for IVF program prognosis. This review aims to describe the predictive significance of several cumulus-oocyte complex (COC) biomarkers in evaluating oocyte developmental competence. A total of eight acclaimed cumulus biomarkers are examined in the study. RT-PCR and microarray analysis were extensively used to assess the significance of these biomarkers in foreseeing oocyte developmental competence. Notably, these biomarkers regulate vital processes associated with oocyte maturation and were found to be differentially expressed in COC encapsulating oocytes of different maturity. The biomarkers were reviewed according to the respective oocyte maturation events namely: nuclear maturation, apoptosis, and extracellular matrix remodeling, and steroid metabolism. Although substantial in vitro evidence was presented to justify the potential use of cumulus biomarkers in predicting oocyte competency and IVF outcomes, the feasibility of assessing these biomarkers as an add-on prognostic procedure in IVF is still restricted due to study challenges.
Collapse
Affiliation(s)
- Batara Sirait
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universitas Kristen Indonesia, Jakarta, Indonesia.,Morula IVF Jakarta Clinic, Jakarta, Indonesia
| | - Budi Wiweko
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.,Yasmin IVF Clinic, Dr. Cipto Mangunkusumo General Hospital, Jakarta, Indonesia.,Human Reproductive, Infertility, and Family Planning Research Cluster, Indonesia Medical Education and Research Institute (IMERI), Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Ahmad Aulia Jusuf
- Department of Histology, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Dein Iftitah
- Human Reproductive, Infertility, and Family Planning Research Cluster, Indonesia Medical Education and Research Institute (IMERI), Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - R Muharam
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.,Yasmin IVF Clinic, Dr. Cipto Mangunkusumo General Hospital, Jakarta, Indonesia.,Human Reproductive, Infertility, and Family Planning Research Cluster, Indonesia Medical Education and Research Institute (IMERI), Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| |
Collapse
|
69
|
Zheng S, Chen Y, Ma M, Li M. Mechanism of quercetin on the improvement of ovulation disorder and regulation of ovarian CNP/NPR2 in PCOS model rats. J Formos Med Assoc 2021; 121:1081-1092. [PMID: 34538551 DOI: 10.1016/j.jfma.2021.08.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 05/26/2021] [Accepted: 08/12/2021] [Indexed: 01/09/2023] Open
Abstract
PURPOSE To investigate the effects of quercetin on ovulation disorder and expression of androgen receptor (AR) and C-type natriuretic peptide (CNP) / Natriuretic Peptide Receptor 2 (NPR2) in dehydroepiandrosterone (DHEA)-induced polycystic ovary syndrome (PCOS) rat model. METHODS DHEA was used to construct the PCOS rat model. After intervention with quercetin, metformin, and AR, the estrous cycle, ovarian and uterine weight of rats were measured. The morphological changes of ovarian and uterine were detected by hematoxylin-eosin staining (HE staining). Luteinizing hormone (LH) and follicle-stimulating hormone (FSH) were measured by Enzyme linked immunosorbent assay (ELISA). Immunohistochemical detection of interleukin-6 (IL-6), interleukin-1β (IL-1β), tumor necrosis factor-alpha (TNF-α), B-cell lymphoma-2 (BCL-2), BCL2-Associated X (Bax) and AR expression in ovarian. Determination of the expression of CNP and NPR2 mRNA by qRT-PCR. Chromatin immunocoprecipitation (ChIP) was used to detect the ability of AR to bind to CNP or NPR2 promoter. RESULTS The results showed that quercetin could significantly reduce the expression of Testosterone (T) , Estradiol (E2) , LH, Bax, IL-1β, IL-6 and TNF-α, increase the expression of FSH and Bcl-2, inhibit the expression of AR, regulate the expression of CNP / NPR2 gene and protein by affecting the combination of AR with the specific sequence of CNP and NPR2 gene promoters, restore the maturation of oocyte and ovulation. CONCLUSION The results suggest that quercetin can alleviate the hormone, metabolic and ovulatory aberrations caused by PCOS, and provide experimental basis for the clinical application of quercetin in PCOS.
Collapse
Affiliation(s)
- Shaoyan Zheng
- Pharmacy Department, Foshan Women And Children Hospital Affiliated to Southern Medical University, China.
| | - Yanxia Chen
- Pharmacy Department, The First Hospital of Guangzhou University of Traditional Chinese Medicine, China
| | - Mingying Ma
- Pharmacy Department, Nanfang Hospital Affiliated to Southern Medical University, China
| | - Miaoxia Li
- Comprehensive Laboratory, Guangdong Huiqun Chinese Traditional Medicine CO., Ltd, China
| |
Collapse
|
70
|
Li J, Liu Z, Kang T, Li M, Wang D, Cheng CHK. Igf3: a novel player in fish reproduction†. Biol Reprod 2021; 104:1194-1204. [PMID: 33693502 DOI: 10.1093/biolre/ioab042] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/26/2021] [Accepted: 03/12/2021] [Indexed: 11/13/2022] Open
Abstract
As in other vertebrates, fish reproduction is tightly controlled by gonadotropin signaling. One of the most perplexing aspects of gonadotropin action on germ cell biology is the restricted expression of gonadotropin receptors in somatic cells of the gonads. Therefore, the identification of factors conveying the action of gonadotropins on germ cells is particularly important for understanding the mechanism of reproduction. Insulin-like growth factors (Igfs) are recognized as key factors in regulating reproduction by triggering a series of physiological processes in vertebrates. Recently, a novel member of Igfs called Igf3 has been identified in teleost. Different from the conventional Igf1 and Igf2 that are ubiquitously expressed in a majority of tissues, Igf3 is solely or highly expressed in the fish gonads. The role of Igf3 in mediating the action of gonadotropin through Igf type 1 receptor on several aspects of oogenesis and spermatogenesis have been demonstrated in several fish species. In this review, we will summarize existing data on Igf3. This new information obtained from Igf3 provides insight into elucidating the molecular mechanism of fish reproduction, and also highlights the importance of Igf system in mediating the action of gonadotropin signaling on animal reproduction.
Collapse
Affiliation(s)
- Jianzhen Li
- College of Life Sciences, Northwest Normal University, Lanzhou, Gansu, China
| | - Zhiquan Liu
- College of Life Sciences, Northwest Normal University, Lanzhou, Gansu, China
| | - Tao Kang
- College of Life Sciences, Northwest Normal University, Lanzhou, Gansu, China
| | - Minghui Li
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, China
| | - Deshou Wang
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, China
| | - Christopher H K Cheng
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| |
Collapse
|
71
|
Johnson GP, Jonas KC. Mechanistic insight into how gonadotropin hormone receptor complexes direct signaling†. Biol Reprod 2021; 102:773-783. [PMID: 31882999 DOI: 10.1093/biolre/ioz228] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/09/2019] [Accepted: 12/17/2019] [Indexed: 12/29/2022] Open
Abstract
Gonadotropin hormones and their receptors play a central role in the control of male and female reproduction. In recent years, there has been growing evidence surrounding the complexity of gonadotropin hormone/receptor signaling, with it increasingly apparent that the Gαs/cAMP/PKA pathway is not the sole signaling pathway that confers their biological actions. Here we review recent literature on the different receptor-receptor, receptor-scaffold, and receptor-signaling molecule complexes formed and how these modulate and direct gonadotropin hormone-dependent intracellular signal activation. We will touch upon the more controversial issue of extragonadal expression of FSHR and the differential signal pathways activated in these tissues, and lastly, highlight the open questions surrounding the role these gonadotropin hormone receptor complexes and how this will shape future research directions.
Collapse
Affiliation(s)
| | - Kim Carol Jonas
- Department of Women and Children's Health, School of Life Course Sciences, King's College London, London, UK
| |
Collapse
|
72
|
Li S, Wang J, Zhang H, Ma D, Zhao M, Li N, Men Y, Zhang Y, Chu H, Lei C, Shen W, Othman OEM, Zhao Y, Min L. Transcriptome profile of goat folliculogenesis reveals the interaction of oocyte and granulosa cell in correlation with different fertility population. Sci Rep 2021; 11:15698. [PMID: 34344973 PMCID: PMC8333342 DOI: 10.1038/s41598-021-95215-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 07/15/2021] [Indexed: 11/28/2022] Open
Abstract
To understand the molecular and genetic mechanisms related to the litter size in one species of two different populations (high litter size and low litter size), we performed RNA-seq for the oocytes and granulosa cells (GCs) at different developmental stages of follicle, and identified the interaction of genes from both sides of follicle (oocyte and GCs) and the ligand-receptor pairs from these two sides. Our data were very comprehensive to uncover the difference between these two populations regarding the folliculogenesis. First, we identified a set of potential genes in oocyte and GCs as the marker genes which can be used to determine the goat fertility capability and ovarian reserve ability. The data showed that GRHPR, GPR84, CYB5A and ERAL1 were highly expressed in oocyte while JUNB, SCN2A, MEGE8, ZEB2, EGR1and PRRC2A were highly expressed in GCs. We found more functional genes were expressed in oocytes and GCs in high fertility group (HL) than that in low fertility group (LL). We uncovered that ligand-receptor pairs in Notch signaling pathway and transforming growth factor-β (TGF-β) superfamily pathways played important roles in goat folliculogenesis for the different fertility population. Moreover, we discovered that the correlations of the gene expression in oocytes and GCs at different stages in the two populations HL and LL were different, too. All the data reflected the gene expression landscape in oocytes and GCs which was correlated well with the fertility capability.
Collapse
Affiliation(s)
- Shen Li
- College of Animal Sciences and Technology, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China
| | - Junjie Wang
- College of Life Sciences, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China
| | - Hongfu Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, People's Republic of China
| | - Dongxue Ma
- College of Animal Sciences and Technology, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China
| | - Minghui Zhao
- College of Life Sciences, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China
| | - Na Li
- College of Life Sciences, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China
| | - Yuhao Men
- College of Animal Sciences and Technology, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China
| | - Yuan Zhang
- Jining Animal Husbandry Development Center, Jining, People's Republic of China
| | - Huimin Chu
- Jining Agricultural Science Institute, Jining, People's Republic of China
| | - Chuzhao Lei
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, People's Republic of China
| | - Wei Shen
- College of Life Sciences, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China
| | | | - Yong Zhao
- College of Life Sciences, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China. .,State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, People's Republic of China.
| | - Lingjiang Min
- College of Animal Sciences and Technology, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China.
| |
Collapse
|
73
|
Tahir MS, Porto-Neto LR, Gondro C, Shittu OB, Wockner K, Tan AWL, Smith HR, Gouveia GC, Kour J, Fortes MRS. Meta-Analysis of Heifer Traits Identified Reproductive Pathways in Bos indicus Cattle. Genes (Basel) 2021; 12:768. [PMID: 34069992 PMCID: PMC8157873 DOI: 10.3390/genes12050768] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/12/2021] [Accepted: 05/13/2021] [Indexed: 12/12/2022] Open
Abstract
Fertility traits measured early in life define the reproductive potential of heifers. Knowledge of genetics and biology can help devise genomic selection methods to improve heifer fertility. In this study, we used ~2400 Brahman cattle to perform GWAS and multi-trait meta-analysis to determine genomic regions associated with heifer fertility. Heifer traits measured were pregnancy at first mating opportunity (PREG1, a binary trait), first conception score (FCS, score 1 to 3) and rebreeding score (REB, score 1 to 3.5). The heritability estimates were 0.17 (0.03) for PREG1, 0.11 (0.05) for FCS and 0.28 (0.05) for REB. The three traits were highly genetically correlated (0.75-0.83) as expected. Meta-analysis was performed using SNP effects estimated for each of the three traits, adjusted for standard error. We identified 1359 significant SNPs (p-value < 9.9 × 10-6 at FDR < 0.0001) in the multi-trait meta-analysis. Genomic regions of 0.5 Mb around each significant SNP from the meta-analysis were annotated to create a list of 2560 positional candidate genes. The most significant SNP was in the vicinity of a genomic region on chromosome 8, encompassing the genes SLC44A1, FSD1L, FKTN, TAL2 and TMEM38B. The genomic region in humans that contains homologs of these genes is associated with age at puberty in girls. Top significant SNPs pointed to additional fertility-related genes, again within a 0.5 Mb region, including ESR2, ITPR1, GNG2, RGS9BP, ANKRD27, TDRD12, GRM1, MTHFD1, PTGDR and NTNG1. Functional pathway enrichment analysis resulted in many positional candidate genes relating to known fertility pathways, including GnRH signaling, estrogen signaling, progesterone mediated oocyte maturation, cAMP signaling, calcium signaling, glutamatergic signaling, focal adhesion, PI3K-AKT signaling and ovarian steroidogenesis pathway. The comparison of results from this study with previous transcriptomics and proteomics studies on puberty of the same cattle breed (Brahman) but in a different population identified 392 genes in common from which some genes-BRAF, GABRA2, GABR1B, GAD1, FSHR, CNGA3, PDE10A, SNAP25, ESR2, GRIA2, ORAI1, EGFR, CHRNA5, VDAC2, ACVR2B, ORAI3, CYP11A1, GRIN2A, ATP2B3, CAMK2A, PLA2G, CAMK2D and MAPK3-are also part of the above-mentioned pathways. The biological functions of the positional candidate genes and their annotation to known pathways allowed integrating the results into a bigger picture of molecular mechanisms related to puberty in the hypothalamus-pituitary-ovarian axis. A reasonable number of genes, common between previous puberty studies and this study on early reproductive traits, corroborates the proposed molecular mechanisms. This study identified the polymorphism associated with early reproductive traits, and candidate genes that provided a visualization of the proposed mechanisms, coordinating the hypothalamic, pituitary, and ovarian functions for reproductive performance in Brahman cattle.
Collapse
Affiliation(s)
- Muhammad S. Tahir
- School of Chemistry and Molecular Bioscience, The University of Queensland Australia, Brisbane, QLD 4072, Australia; (M.S.T.); (O.B.S.); (K.W.); (A.W.L.T.); (H.R.S.); (J.K.)
| | - Laercio R. Porto-Neto
- Commonwealth Scientific and Industrial Research Organization, Brisbane, QLD 4072, Australia;
| | - Cedric Gondro
- Department of Animal Science, Michigan State University, East Lansing, MI 48824, USA;
| | - Olasege B. Shittu
- School of Chemistry and Molecular Bioscience, The University of Queensland Australia, Brisbane, QLD 4072, Australia; (M.S.T.); (O.B.S.); (K.W.); (A.W.L.T.); (H.R.S.); (J.K.)
| | - Kimberley Wockner
- School of Chemistry and Molecular Bioscience, The University of Queensland Australia, Brisbane, QLD 4072, Australia; (M.S.T.); (O.B.S.); (K.W.); (A.W.L.T.); (H.R.S.); (J.K.)
| | - Andre W. L. Tan
- School of Chemistry and Molecular Bioscience, The University of Queensland Australia, Brisbane, QLD 4072, Australia; (M.S.T.); (O.B.S.); (K.W.); (A.W.L.T.); (H.R.S.); (J.K.)
| | - Hugo R. Smith
- School of Chemistry and Molecular Bioscience, The University of Queensland Australia, Brisbane, QLD 4072, Australia; (M.S.T.); (O.B.S.); (K.W.); (A.W.L.T.); (H.R.S.); (J.K.)
| | - Gabriela C. Gouveia
- Animal Science Department, Veterinary School, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil;
| | - Jagish Kour
- School of Chemistry and Molecular Bioscience, The University of Queensland Australia, Brisbane, QLD 4072, Australia; (M.S.T.); (O.B.S.); (K.W.); (A.W.L.T.); (H.R.S.); (J.K.)
| | - Marina R. S. Fortes
- School of Chemistry and Molecular Bioscience, The University of Queensland Australia, Brisbane, QLD 4072, Australia; (M.S.T.); (O.B.S.); (K.W.); (A.W.L.T.); (H.R.S.); (J.K.)
| |
Collapse
|
74
|
Mu R, Yu YY, Gegen T, Wen D, Wang F, Chen Z, Xu WB. Transcriptome analysis of ovary tissues from low- and high-yielding Changshun green-shell laying hens. BMC Genomics 2021; 22:349. [PMID: 33990173 PMCID: PMC8122536 DOI: 10.1186/s12864-021-07688-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 04/28/2021] [Indexed: 02/18/2023] Open
Abstract
Background Changshun green-shell laying hens are unique to Guizhou Province, China, and have high egg quality. Improving egg production performance has become an important breeding task, and in recent years, the development of high-throughput sequencing technology provides a fast and exact method for genetic selection. Therefore, we aimed to use this technology to analyze the differences between the ovarian mRNA transcriptome of low and high-yield Changshun green-shell layer hens, identify critical pathways and candidate genes involved in controlling the egg production rate, and provide basic data for layer breeding. Results The egg production rates of the low egg production group (LP) and the high egg production group (HP) were 68.00 ± 5.56 % and 93.67 ± 7.09 %, with significant differences between the groups (p < 0.01). Moreover, the egg weight, shell thickness, strength and layer weight of the LP were significantly greater than those of the HP (p < 0.05). More than 41 million clean reads per sample were obtained, and more than 90 % of the clean reads were mapped to the Gallus gallus genome. Further analysis identified 142 differentially expressed genes (DEGs), and among them, 55 were upregulated and 87 were downregulated in the ovaries. KEGG pathway enrichment analysis identified 9 significantly enriched pathways, with the neuroactive ligand-receptor interaction pathway being the most enriched. GO enrichment analysis indicated that the GO term transmembrane receptor protein tyrosine kinase activity, and the DEGs identified in this GO term, including PRLR, NRP1, IL15, BANK1, NTRK1, CCK, and HGF may be associated with crucial roles in the regulation of egg production. Conclusions The above-mentioned DEGs may be relevant for the molecular breeding of Changshun green-shell laying hens. Moreover, enrichment analysis indicated that the neuroactive ligand-receptor interaction pathway and receptor protein tyrosine kinases may play crucial roles in the regulation of ovarian function and egg production. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-07688-x.
Collapse
Affiliation(s)
- Ren Mu
- College of Biological Science and Agriculture, Qiannan Normal University for Nationalities Duyun, Jianjiang Road 5, 558000, Duyun, China
| | - Yi-Yin Yu
- College of Biological Science and Agriculture, Qiannan Normal University for Nationalities Duyun, Jianjiang Road 5, 558000, Duyun, China
| | - Tuya Gegen
- Library, Qiannan Normal University for Nationalities, 558000, Duyun, China
| | - Di Wen
- College of Biological Science and Agriculture, Qiannan Normal University for Nationalities Duyun, Jianjiang Road 5, 558000, Duyun, China
| | - Fen Wang
- College of Biological Science and Agriculture, Qiannan Normal University for Nationalities Duyun, Jianjiang Road 5, 558000, Duyun, China
| | - Zhi Chen
- College of Biological Science and Agriculture, Qiannan Normal University for Nationalities Duyun, Jianjiang Road 5, 558000, Duyun, China.
| | - Wen-Bin Xu
- College of Animal Sciences, Zhejiang University, 310058, Hangzhou, China. .,School of Marine Sciences, Ningbo University, 315211, Ningbo, China.
| |
Collapse
|
75
|
Abstract
Meiosis is a highly conserved and essential process in gametogenesis in sexually reproducing organisms. However, there are substantial sex-specific differences within individual species with respect to meiosis-related chromatin reorganization, recombination, and tolerance for meiotic defects. A wide range of murine models have been developed over the past two decades to study the complex regulatory processes governing mammalian meiosis. The present review article thus provides a comprehensive overview of the knockout mice that have been employed to study meiosis, with a particular focus on gene- and gametogenesis-related sexual dimorphism observed in these model animals. In so doing, we aim to provide a firm foundation for the future study of sex-specific differences in meiosis at the molecular level.
Collapse
Affiliation(s)
- Rong Hua
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Mingxi Liu
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| |
Collapse
|
76
|
Giles J, Alama P, Gamiz P, Vidal C, Badia P, Pellicer A, Bosch E. Medroxyprogesterone acetate is a useful alternative to a gonadotropin-releasing hormone antagonist in oocyte donation: a randomized, controlled trial. Fertil Steril 2021; 116:404-412. [PMID: 33814126 DOI: 10.1016/j.fertnstert.2021.02.036] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/21/2021] [Accepted: 02/23/2021] [Indexed: 10/21/2022]
Abstract
OBJECTIVE To compare ovarian response and reproductive outcomes in oocyte donors undergoing pituitary suppression with medroxyprogesterone acetate (MPA) versus those undergoing conventional treatment with a gonadotropin-releasing hormone (GnRH) antagonist. DESIGN A prospective, randomized, controlled trial of cycles was conducted from October 2017 to June 2019 to evaluate ovarian response in terms of the number of oocytes. The reproductive outcomes of the recipients were retrospectively analyzed later. SETTING A university-affiliated private in vitro fertilization center. PATIENT(S) We randomly divided 318 donors into 2 groups in a 1:1 ratio. The oocytes obtained were assigned to 364 recipients. One hundred sixty-one donors were treated with a daily dose of 10 mg of MPA administered orally from the beginning of ovarian stimulation (OS), and 156 were treated with a GnRH antagonist (initiated once the leading follicle reached a diameter of 13 mm). Transvaginal ultrasound was performed, and serum estradiol, luteinizing hormone, and progesterone levels were recorded during monitoring. The following additional parameters were analyzed: endocrine profile (in follicular fluid), number of metaphase II oocytes, and pregnancy outcome. INTERVENTION(S) The donors included in the study group were stimulated using recombinant follicle-stimulating hormone and MPA at 10 mg/day, simultaneously begun on cycle day 2 or 3. Ovulation was induced using a GnRH agonist when dominant follicles matured. A short protocol with ganirelix at 0.25 mg/day was used for the control group. Oocytes were assigned to the recipients, followed by routine in vitro fertilization procedures in which 1 embryo was usually transferred. MAIN OUTCOME MEASURE(S) The primary outcome measure was the numbers of oocytes and metaphase II oocytes retrieved. The secondary outcomes were the incidence of premature luteinizing hormone surge, serum and follicular fluid hormone profiles, and clinical pregnancy outcomes in the recipient group. RESULT(S) The number of oocytes retrieved was 21.4 ± 11.7 in the MPA group and 21.2 ± 9.2 in the antagonist group (mean difference 0.14; 95% confidence interval -2.233, 2.517). The total dose of recombinant follicle-stimulating hormone, duration of OS, and endocrine profiles of the serum and follicular fluids were comparable in the 2 groups. No early ovulation was observed in either group. No statistically significant differences with respect to implantation rate (68.1% in the MPA group vs. 62% in the antagonist group), clinical pregnancy rate (64.5% in the MPA group vs. 57.8 in the antagonist group), ongoing pregnancy rate (55.4% in the MPA group vs. 48.5% in the antagonist group), live birth rate (55.1% in the MPA group vs. 48.5% in the antagonist group), or cumulative live birth rate (73.8% in the MPA group vs. 70.7% in the antagonist group) were observed between the groups. CONCLUSION(S) The administration of MPA resulted in oocyte retrieval rates, endocrine profiles, viable embryo numbers, and pregnancy outcomes similar to those achieved with the GnRH antagonist. Therefore, MPA can be recommended for OS in oocyte donation because it permits a more patient-friendly approach. CLINICAL TRIAL REGISTRATION NUMBER NCT03300960.
Collapse
Affiliation(s)
- Juan Giles
- IVI-RMA, Valencia, Spain; IVI Foundation, Instituto de Investigación Sanitaria Hospital Universitario y Politécnico La Fe, Valencia, Spain.
| | - Pilar Alama
- IVI-RMA, Valencia, Spain; IVI Foundation, Instituto de Investigación Sanitaria Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Pilar Gamiz
- IVI-RMA, Valencia, Spain; IVI Foundation, Instituto de Investigación Sanitaria Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Carmen Vidal
- IVI-RMA, Valencia, Spain; IVI Foundation, Instituto de Investigación Sanitaria Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | | | - Antonio Pellicer
- IVI Foundation, Instituto de Investigación Sanitaria Hospital Universitario y Politécnico La Fe, Valencia, Spain; IVI-RMA, Rome, Italy; Department of Pediatrics, Obstetrics and Gynecology, University of Valencia, Valencia, Spain; Department of Genomic and Systems Reproductive Medicine, IVI-RMA IVI Foundation, Valencia, Spain
| | - Ernesto Bosch
- IVI-RMA, Valencia, Spain; IVI Foundation, Instituto de Investigación Sanitaria Hospital Universitario y Politécnico La Fe, Valencia, Spain
| |
Collapse
|
77
|
Buratini J, Dal Canto M, De Ponti E, Brambillasca F, Brigante C, Gippone S, Mignini Renzini M, La Marca A. Maternal age affects the relationship of basal FSH and anti-Müllerian hormone concentrations with post-ICSI/IVF live birth. Reprod Biomed Online 2021; 42:748-756. [PMID: 33653653 DOI: 10.1016/j.rbmo.2020.12.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 12/04/2020] [Accepted: 12/08/2020] [Indexed: 10/22/2022]
Abstract
RESEARCH QUESTION Does the association of basal FSH and anti-Müllerian hormone (AMH) concentrations with post-IVF/intracytoplasmic sperm injection (ICSI) live birth change with maternal age? DESIGN A total of 2003 IVF/ICSI patients were stratified according to basal FSH/AMH in concordant favourable (CF; AMH >1 ng/ml and FSH ≤10 IU/l), concordant unfavourable (CU; AMH ≤1 ng/ml and FSH >10 IU/l), discordant with favourable AMH (DFA) and discordant with favourable FSH (DFF) groups, as well as according to age in pre-advanced maternal age (pre-AMA; <35), AMA-1 (≥35, ≤37), AMA-2 (>37, ≤40) and AMA-3 (>40). IVF/ICSI outcomes were compared among CF, CU, DFA and DFF groups, and the association of basal FSH and AMH concentrations with live birth was tested by univariate and multivariate analysis in total, pre-AMA and AMA groups, separately. RESULTS Different outcome patterns were observed in discordant AMH/FSH groups from different age categories; favourable basal FSH concentrations were associated with higher delivery rates in pre-AMA patients, but with lower delivery rates in AMA groups. Within pre-AMA patients, DFF patients presented higher delivery rates but lower oocyte yield compared with DFA patients. In the univariate analysis, favourable AMH (P < 0.02) and oocyte yield (P < 0.002) were positively associated with live birth in all AMA groups. The multivariate analysis revealed that favourable basal FSH, but not AMH or oocyte yield, is associated with live birth in pre-AMA patients independently of other variables (P = 0.012). CONCLUSIONS The relationship of basal FSH and AMH with IVF/ICSI success changes with maternal age; basal FSH better reflects clinical outcomes probably determined by oocyte quality in pre-AMA patients, while AMH better suits AMA patients.
Collapse
Affiliation(s)
- Jose Buratini
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, Brazil; Biogenesi, Reproductive Medicine Centre, Monza, Italy.
| | | | | | | | | | | | | | - Antonio La Marca
- Clinica Eugin, Modena, Italy; Department of Medical and Surgical Sciences of the Mother, Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
78
|
He M, Zhang T, Yang Y, Wang C. Mechanisms of Oocyte Maturation and Related Epigenetic Regulation. Front Cell Dev Biol 2021; 9:654028. [PMID: 33842483 PMCID: PMC8025927 DOI: 10.3389/fcell.2021.654028] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 02/25/2021] [Indexed: 12/16/2022] Open
Abstract
Meiosis is the basis of sexual reproduction. In female mammals, meiosis of oocytes starts before birth and sustains at the dictyate stage of meiotic prophase I before gonadotropins-induced ovulation happens. Once meiosis gets started, the oocytes undergo the leptotene, zygotene, and pachytene stages, and then arrest at the dictyate stage. During each estrus cycle in mammals, or menstrual cycle in humans, a small portion of oocytes within preovulatory follicles may resume meiosis. It is crucial for females to supply high quality mature oocytes for sustaining fertility, which is generally achieved by fine-tuning oocyte meiotic arrest and resumption progression. Anything that disturbs the process may result in failure of oogenesis and seriously affect both the fertility and the health of females. Therefore, uncovering the regulatory network of oocyte meiosis progression illuminates not only how the foundations of mammalian reproduction are laid, but how mis-regulation of these steps result in infertility. In order to provide an overview of the recently uncovered cellular and molecular mechanism during oocyte maturation, especially epigenetic modification, the progress of the regulatory network of oocyte meiosis progression including meiosis arrest and meiosis resumption induced by gonadotropins is summarized. Then, advances in the epigenetic aspects, such as histone acetylation, phosphorylation, methylation, glycosylation, ubiquitination, and SUMOylation related to the quality of oocyte maturation are reviewed.
Collapse
Affiliation(s)
- Meina He
- Department of Biology, School of Basic Medical Science, Guizhou Medical University, Guiyang, China
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, College of Life Science, Ningxia University, Yinchuan, China
| | - Tuo Zhang
- Department of Physiology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, College of Life Science, Ningxia University, Yinchuan, China
| | - Yi Yang
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, College of Life Science, Ningxia University, Yinchuan, China
| | - Chao Wang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, College of Life Science, Ningxia University, Yinchuan, China
| |
Collapse
|
79
|
Abbassi L, El-Hayek S, Carvalho KF, Wang W, Yang Q, Granados-Aparici S, Mondadori R, Bordignon V, Clarke HJ. Epidermal growth factor receptor signaling uncouples germ cells from the somatic follicular compartment at ovulation. Nat Commun 2021; 12:1438. [PMID: 33664246 PMCID: PMC7933413 DOI: 10.1038/s41467-021-21644-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 02/07/2021] [Indexed: 01/31/2023] Open
Abstract
Germ cells are physically coupled to somatic support cells of the gonad during differentiation, but this coupling must be disrupted when they are mature, freeing them to participate in fertilization. In mammalian females, coupling occurs via specialized filopodia that project from the ovarian follicular granulosa cells to the oocyte. Here, we show that signaling through the epidermal growth factor receptor (EGFR) in the granulosa, which becomes activated at ovulation, uncouples the germ and somatic cells by triggering a massive and temporally synchronized retraction of the filopodia. Although EGFR signaling triggers meiotic maturation of the oocyte, filopodial retraction is independent of the germ cell state, being regulated solely within the somatic compartment, where it requires ERK-dependent calpain-mediated loss of filopodia-oocyte adhesion followed by Arp2/3-mediated filopodial shortening. By uncovering the mechanism regulating germ-soma uncoupling at ovulation, our results open a path to improving oocyte quality in human and animal reproduction.
Collapse
Affiliation(s)
- Laleh Abbassi
- Research Institute of the McGill University Health Centre, Montreal, Canada
- Division of Experimental Medicine, McGill University, Montreal, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Stephany El-Hayek
- Research Institute of the McGill University Health Centre, Montreal, Canada
- Department of Biology, McGill University, Montreal, Canada
- Centre for Arab Genomic Studies, Dubai, United Arab Emirates
| | - Karen Freire Carvalho
- Research Institute of the McGill University Health Centre, Montreal, Canada
- Division of Experimental Medicine, McGill University, Montreal, Canada
| | - Wusu Wang
- Research Institute of the McGill University Health Centre, Montreal, Canada
- College of Animal Science and Technology, Northwest A&F University, Shaanxi, PR China
| | - Qin Yang
- Research Institute of the McGill University Health Centre, Montreal, Canada
| | | | - Rafael Mondadori
- Department of Animal Science, McGill University, Montreal, Canada
| | - Vilceu Bordignon
- Department of Animal Science, McGill University, Montreal, Canada
| | - Hugh J Clarke
- Research Institute of the McGill University Health Centre, Montreal, Canada.
- Division of Experimental Medicine, McGill University, Montreal, Canada.
- Department of Biology, McGill University, Montreal, Canada.
- Department of Obstetrics and Gynecology, McGill University, Montreal, Canada.
| |
Collapse
|
80
|
Machtinger R, Baccarelli AA, Wu H. Extracellular vesicles and female reproduction. J Assist Reprod Genet 2021; 38:549-557. [PMID: 33471231 PMCID: PMC7910356 DOI: 10.1007/s10815-020-02048-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 12/21/2020] [Indexed: 01/28/2023] Open
Abstract
Extracellular vesicles (EVs) are nano-sized membrane bound complexes that have been identified as a mean for intercellular communication between cells and tissues both in physiological and pathological conditions. These vesicles contain numerous molecules involved in signal transduction including microRNAs, mRNAs, DNA, proteins, lipids, and cytokines and can affect the behavior of recipient cells. Female reproduction is dependent on extremely fine-tuned endocrine regulation, and EVs may represent an added layer that contributes to this regulation. This narrative review article provides an update on the research of the role of EVs in female reproduction including folliculogenesis, fertilization, embryo quality, and implantation. We also highlight potential pitfalls in typical EV studies and discuss gaps in the current literature.
Collapse
Affiliation(s)
- Ronit Machtinger
- Sheba Medical Center, Ramat Gan and Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
- Infertility and IVF Unit, Department of Obstetrics and Gynecology, Chaim Sheba Medical Center, 52621, Tel Hashomer, Israel.
| | - Andrea A Baccarelli
- Environmental Precision Biosciences Laboratory, Columbia University, Mailman School of Public Health, New York, NY, USA
| | - Haotian Wu
- Environmental Precision Biosciences Laboratory, Columbia University, Mailman School of Public Health, New York, NY, USA
| |
Collapse
|
81
|
In vitro growth culture in a medium with reduced sodium chloride improves maturation and developmental competence of pig oocytes derived from small antral follicles. Theriogenology 2021; 165:37-43. [PMID: 33639366 DOI: 10.1016/j.theriogenology.2020.12.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/11/2020] [Accepted: 12/14/2020] [Indexed: 11/24/2022]
Abstract
The objective of this study was to evaluate the effects of reducing the sodium chloride content in in vitro growth (IVG) medium to 61.6 mM on in vitro maturation (IVM) and embryonic development of pig oocytes derived from small antral follicles (SAF) less than 3 mm in diameter. SAF oocytes were cultured for 2 days to induce IVG in alpha-minimal essential medium with 108 mM NaCl (αMEM-108) or porcine zygote medium (PZM) containing 61.6 mM (PZM-61.6) or 108 mM (PZM-108) NaCl. These media were further supplemented with 1 mM dibutyryl cyclic adenosine monophosphate (dbcAMP) and 10% (v/v) fetal bovine serum. After IVG culture, oocytes were matured for 44 h in our standard IVM medium. The IVG culture in PZM-61.6 significantly increased nuclear maturation (88.0 ± 2.2%) of SAF oocytes compared to that in PZM-108 (77.3 ± 3.9%) or αMEM-108 (75.9 ± 3.8%). After parthenogenesis (PA), the proportions of blastocysts, based on the number of metaphase II (MII) oocytes, induced for PA were not different among IVG oocytes cultured in PZM-61.6 (50.2 ± 3.0%), PZM-108 (46.8 ± 2.9%), or αMEM-108 (45.6 ± 2.9%). The IVM oocytes derived from IVG in PZM-61.6 showed increased perivitelline space (PVS) (12.1 ± 0.6 μm) and intra-oocyte glutathione (GSH) content (1.19 ± 0.04 pixels/oocyte) compared to PVS (8.0 ± 0.5 and 7.4 ± 0.4 μm) and GSH (1.03 ± 0.04 and 1.00 ± 0.04 pixels/oocyte) of oocytes derived from PZM-108 and αMEM-108, respectively. The IVG culture in PZM-61.6 stimulated meiotic resumption after IVG and faster nuclear progression after IVM than that in αMEM-108. After somatic cell nuclear transfer (SCNT), the blastocyst formation of SAF oocytes grown in PZM-61.6 (17.8 ± 3.3%) was higher than that of oocytes grown in PZM-108 (7.5 ± 2.7%) but not different from that of oocytes in αMEM-108 (11.4 ± 3.4%). Regardless of the different osmotic pressures, nuclear maturation was significantly increased by IVG culture in PZM with reduced NaCl (86.8 ± 2.3 and 84.9 ± 4.2% in PZM-61.6 and PZM-61.6 with sorbitol, respectively) than in PZM-108 (70.5 ± 3.4%). Blastocyst formation was not affected by the differences in NaCl content and osmotic pressure of the IVG medium, whereas the mean number of cells in blastocysts was significantly higher following IVG culture in PZM-61.6 than in the other groups. In conclusion, the results demonstrate that, following SCNT in pigs, IVG culture of SAF oocytes in a medium with a reduced NaCl concentration stimulates oocyte maturation and improves subsequent embryonic development.
Collapse
|
82
|
Jang YJ, Kim JS, Yun PR, Seo YW, Lee TH, Park JI, Chun SY. Involvement of peroxiredoxin 2 in cumulus expansion and oocyte maturation in mice. Reprod Fertil Dev 2021; 32:783-791. [PMID: 32389179 DOI: 10.1071/rd19310] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 11/30/2019] [Indexed: 01/11/2023] Open
Abstract
Peroxiredoxin 2 (Prdx2), an antioxidant enzyme, is expressed in the ovary during the ovulatory process. The aim of the present study was to examine the physiological role of Prdx2 during ovulation using Prdx2-knockout mice and mouse cumulus-oocyte complex (COC) from WT mice. Two days of treatment of immature mice (21-23 days old) with equine chorionic gonadotrophin and followed by treatment with human chorionic gonadotrophin greatly impaired cumulus expansion and oocyte maturation in Prdx2-knockout but not wild-type mice. Treatment of COCs in culture with conoidin A (50µM), a 2-cys Prdx inhibitor, abolished epiregulin (EPI)-induced cumulus expansion. Conoidin A treatment also inhibited EPI-stimulated signal molecules, including signal transducer and activator of transcription-3, AKT and mitogen-activated protein kinase 1/2. Conoidin A treatment also reduced the gene expression of EPI-stimulated expansion-inducing factors (hyaluronan synthase 2 (Has2), pentraxin 3 (Ptx3), TNF-α induced protein 6 (Tnfaip6) and prostaglandin-endoperoxide synthase 2 (Ptgs2)) and oocyte-derived factors (growth differentiation factor 9 (Gdf9) and bone morphogenetic protein 15 (Bmp15)). Furthermore, conoidin A inhibited EPI-induced oocyte maturation and the activity of connexins 43 and 37. Together, these results demonstrate that Prdx2 plays a role in regulating cumulus expansion and oocyte maturation during the ovulatory process in mice, probably by modulating epidermal growth factor receptor signalling.
Collapse
Affiliation(s)
- You-Jee Jang
- Animal Facility of Aging Science, Korea Basic Science Institute, Gwangju 61186, Republic of Korea
| | - Jin-Seon Kim
- School of Biological Sciences and Biotechnology, Faculty of Life Science, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Pu-Reum Yun
- School of Biological Sciences and Biotechnology, Faculty of Life Science, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Young-Woo Seo
- Animal Facility of Aging Science, Korea Basic Science Institute, Gwangju 61186, Republic of Korea
| | - Tae-Hoon Lee
- Department of Oral Biochemistry, College of Dentistry, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Jae-Il Park
- Animal Facility of Aging Science, Korea Basic Science Institute, Gwangju 61186, Republic of Korea. Corresponding authors.
| | - Sang-Young Chun
- School of Biological Sciences and Biotechnology, Faculty of Life Science, Chonnam National University, Gwangju 61186, Republic of Korea. Corresponding authors.
| |
Collapse
|
83
|
Cui C, Wang J, Han X, Wang Q, Zhang S, Liang S, Li H, Meng L, Zhang C, Chen H. Identification of small extracellular vesicle-linked miRNA specifically derived from intrafollicular cells in women with polycystic ovary syndrome. Reprod Biomed Online 2021; 42:870-880. [PMID: 33840620 DOI: 10.1016/j.rbmo.2021.02.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 01/06/2021] [Accepted: 02/04/2021] [Indexed: 01/04/2023]
Abstract
RESEARCH QUESTION This study aimed to identify small extracellular vesicle (sEV)-linked microRNAs (miRNA) specifically derived from intrafollicular cells in women with polycystic ovary syndrome (PCOS) and to investigate their biological functions. DESIGN A total of 120 women were recruited from September 2017 to October 2018. To investigate miRNA profiles in sEV derived from follicular fluid and serum, 30 women with PCOS and 30 without PCOS were included for a miRNA microarray containing probes interrogating 2549 human miRNA. To study the expression levels of differentially expressed miRNA, sEV in follicular fluid obtained from another 30 PCOS and 30 non-PCOS patients were used for quantitative real-time polymerase chain reaction analysis. RESULTS A total of 281 sEV-linked miRNA specifically derived from intrafollicular cells were identified, 179 of which were expressed in both the PCOS and non-PCOS groups. Twenty-six of the 179 intrafollicle-specific sEV-linked miRNA were predicted to target 1537 genes. Functional analysis suggested that these genes were involved in pathways related to folliculogenesis, including the MAPK, and PI3K-Akt signalling pathways. Quantitative real-time polymerase chain reaction analysis showed that the expression of seven intrafollicle-specific sEV-linked miRNA was significantly higher in follicular fluid-derived sEV in women with PCOS than in women without it. These miRNA and their corresponding target genes were identified as being involved in the MAPK signalling pathway and oocyte meiosis. CONCLUSIONS The data suggest that the aberrantly expressed miRNA and their target genes might be associated with PCOS, providing novel insights into the molecular mechanisms underlying regulation of folliculogenesis and oocyte maturation in PCOS.
Collapse
Affiliation(s)
- Chenchen Cui
- Department of Reproductive Medicine Center, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital of Henan University, Zhengzhou, Henan, China; Henan Joint International Research Laboratory of Reproductive Bioengineering, Zhengzhou Henan Province, China
| | - Jie Wang
- Women & Infant Hospital of Zhengzhou, Zhengzhou, Henan, China
| | - Xiao Han
- Department of Reproductive Medicine Center, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital of Henan University, Zhengzhou, Henan, China; Henan Joint International Research Laboratory of Reproductive Bioengineering, Zhengzhou Henan Province, China
| | - Qian Wang
- Department of Reproductive Medicine Center, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital of Henan University, Zhengzhou, Henan, China; Henan Joint International Research Laboratory of Reproductive Bioengineering, Zhengzhou Henan Province, China
| | - Shan Zhang
- Department of Reproductive Medicine Center, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital of Henan University, Zhengzhou, Henan, China; Department of Reproductive Medicine Center, Henan Provincial People's Hospital Affiliated to Xinxiang Medical College, Zhengzhou, Henan, China
| | - Shoujing Liang
- Department of Reproductive Medicine Center, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital of Henan University, Zhengzhou, Henan, China; Henan Joint International Research Laboratory of Reproductive Bioengineering, Zhengzhou Henan Province, China
| | - Huan Li
- Department of Reproductive Medicine Center, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital of Henan University, Zhengzhou, Henan, China; Henan Joint International Research Laboratory of Reproductive Bioengineering, Zhengzhou Henan Province, China
| | - Li Meng
- Department of Reproductive Medicine Center, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital of Henan University, Zhengzhou, Henan, China; Henan Joint International Research Laboratory of Reproductive Bioengineering, Zhengzhou Henan Province, China
| | - Cuilian Zhang
- Department of Reproductive Medicine Center, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital of Henan University, Zhengzhou, Henan, China; Henan Joint International Research Laboratory of Reproductive Bioengineering, Zhengzhou Henan Province, China.
| | - Huanhuan Chen
- Department of Reproductive Medicine Center, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital of Henan University, Zhengzhou, Henan, China; Henan Joint International Research Laboratory of Reproductive Bioengineering, Zhengzhou Henan Province, China.
| |
Collapse
|
84
|
Miyazaki T, Uenoyama R, Matsuzaki T, Yamashita T, Hirata TI, Miyazaki M. Detection of urinary luteinizing hormone in Japanese black cows after administration of gonadotropin-releasing hormone. J Vet Med Sci 2021; 83:431-434. [PMID: 33455959 PMCID: PMC8025431 DOI: 10.1292/jvms.20-0506] [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] [Indexed: 11/22/2022] Open
Abstract
The blood luteinizing hormone (LH) surge in cows is well studied. However, little is known about urinary LH in cows. This study examined urinary LH concentrations after administration of gonadotropin-releasing hormone (GnRH) in six Japanese black cows to induce LH secretion from the pituitary gland into the bloodstream. Abrupt rises in plasma and urinary LH were observed after GnRH administration. Plasma and urinary LH peaked at 2 and 5 hr, respectively. A positive correlation was observed between plasma LH concentrations and urinary LH amounts. Ovulation was confirmed in the cows after 48 hr of GnRH administration. These data strongly suggest that urinary LH is derived from plasma LH, which triggers ovulation in cows.
Collapse
Affiliation(s)
- Tamako Miyazaki
- Faculty of Agriculture, Iwate University, 3-18-8 Ueda, Morioka, Iwate 020-8550, Japan
| | - Reiko Uenoyama
- Faculty of Agriculture, Iwate University, 3-18-8 Ueda, Morioka, Iwate 020-8550, Japan
| | - Takashi Matsuzaki
- Faculty of Agriculture, Iwate University, 3-18-8 Ueda, Morioka, Iwate 020-8550, Japan
| | - Tetsuro Yamashita
- Faculty of Agriculture, Iwate University, 3-18-8 Ueda, Morioka, Iwate 020-8550, Japan
| | - Toh-Ichi Hirata
- Faculty of Agriculture, Iwate University, 3-18-8 Ueda, Morioka, Iwate 020-8550, Japan
| | - Masao Miyazaki
- Faculty of Agriculture, Iwate University, 3-18-8 Ueda, Morioka, Iwate 020-8550, Japan
| |
Collapse
|
85
|
Abstract
Infertility is described as unexplained when pregnancy does not occur despite ovulation, patent Fallopian tubes, and normal semen parameters. Oocyte developmental competence (or quality) is rate-limiting for pregnancy success as oocytes provide virtually all the cellular building blocks including mitochondria required during embryogenesis. However, available tests estimate oocyte numbers (anti-Müllerian hormone, follicle-stimulating hormone and antral follicle count) and ovulation (luteal phase serum progesterone) but not the third, and most pivotal, oocyte-specific parameter, quality. Severe depletion of the follicular reserve manifests as premature ovarian insufficiency and is an obvious cause of anovulation with overt symptoms and clear diagnostic criteria. In contrast, there are no biomarkers of poor oocyte quality other than through in vitro fertilization when readouts of oocyte quality such as preimplantation embryo development can be assessed. The most common cause of poor oocyte quality is natural aging, which is strongly tied to reduced oocyte mitochondrial efficiency and increased oxidative stress. In younger women, quality may also be impaired due to accelerated aging or sporadic genetic mutations which cause severe defects during oocyte and embryo development. Thus, poor oocyte quality often provides an explanation for infertility, but because it cannot be measured using conventional tests, many cases of infertility are often incorrectly labeled "unexplained." Since female age remains the best predictor of oocyte quality, age over 37 years should be considered an independent diagnostic criterion.
Collapse
Affiliation(s)
- Hayden Anthony Homer
- Christopher Chen Oocyte Biology Research Laboratory, UQ Centre for Clinical Research, The University of Queensland, Brisbane, Herston 4029, Queensland, Australia
| |
Collapse
|
86
|
Kawagoe Y, Kawashima I, Sato Y, Okamoto N, Matsubara K, Kawamura K. CXCL5-CXCR2 signaling is a senescence-associated secretory phenotype in preimplantation embryos. Aging Cell 2020; 19:e13240. [PMID: 32959976 PMCID: PMC7576282 DOI: 10.1111/acel.13240] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 08/15/2020] [Accepted: 08/22/2020] [Indexed: 12/12/2022] Open
Abstract
Pregnancy rate of women decreases with age due to declining quality of oocytes and embryos. However, there is no established method to improve pregnancy rate in aging women. In this study, we identified a senescence-associated secretory phenotype (SASP) factor partially responsible for the decline in embryo implantation potential. Based on microarray analysis using young and aging human embryos at the same morphological grade, 702 genes showed >fivefold increases in aging human blastocysts. Among these genes, C-X-C motif chemokine 5 (CXCL5) showed 7.7-fold increases in aging human blastocysts. However, no-age-dependent changes in expression of the CXCR2, the cognate receptor for CXCL5, were found. In aging mice, Cxcl5 transcript levels were also increased in oocytes and embryos. Treatment of young mouse embryos with CXCL5 decreased implantation rates, together with increased expression of aging markers (P53, P21, Pai-1, and Il-6). Moreover, CXCL5 treatment suppressed trophoblast outgrowth in young mouse blastocysts. Conversely, suppression of CXCL5-CXCR2 signaling in aging mouse embryos using neutralizing antibodies and a receptor antagonist improved the implantation rate, leading to increases in pregnancy and delivery of normal pups. The gene expression pattern of these embryos was comparable to that in young mouse embryos showing enriched cell proliferation-related pathways. In conclusion, we identified CXCL5 as a SASP factor in human and mouse embryos and suppression of CXCL5-CXCR2 signaling during embryo culture improved pregnancy success in aging mice. Future analysis on CXCL5-CXCR2 signaling suppression in human embryos could be the basis to improve embryo development and pregnancy outcome in middle-aged infertile patients.
Collapse
Affiliation(s)
- Yuta Kawagoe
- Department of Obstetrics and Gynecology Advanced Reproduction Research Center International University of Health and Welfare School of Medicine Narita Japan
- The United Graduate School of Agriculture Sciences Iwate University Morioka Japan
| | - Ikko Kawashima
- Institute of Advanced BioMedical Engineering and Science Tokyo Women's Medical University Shinjuku‐ku Japan
| | - Yorino Sato
- Department of Obstetrics and Gynecology Advanced Reproduction Research Center International University of Health and Welfare School of Medicine Narita Japan
| | - Naoki Okamoto
- Department of Obstetrics and Gynecology Advanced Reproduction Research Center International University of Health and Welfare School of Medicine Narita Japan
| | - Kazuei Matsubara
- The United Graduate School of Agriculture Sciences Iwate University Morioka Japan
| | - Kazuhiro Kawamura
- Department of Obstetrics and Gynecology Advanced Reproduction Research Center International University of Health and Welfare School of Medicine Narita Japan
| |
Collapse
|
87
|
Qiu J, McGaughey SA, Groszmann M, Tyerman SD, Byrt CS. Phosphorylation influences water and ion channel function of AtPIP2;1. PLANT, CELL & ENVIRONMENT 2020; 43:2428-2442. [PMID: 32678928 DOI: 10.1111/pce.13851] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 07/09/2020] [Accepted: 07/09/2020] [Indexed: 05/24/2023]
Abstract
The phosphorylation state of two serine residues within the C-terminal domain of AtPIP2;1 (S280, S283) regulates its plasma membrane localization in response to salt and osmotic stress. Here, we investigated whether the phosphorylation state of S280 and S283 also influence AtPIP2;1 facilitated water and cation transport. A series of single and double S280 and S283 phosphomimic and phosphonull AtPIP2;1 mutants were tested in heterologous systems. In Xenopus laevis oocytes, phosphomimic mutants AtPIP2;1 S280D, S283D, and S280D/S283D had significantly greater ion conductance for Na+ and K+ , whereas the S280A single phosphonull mutant had greater water permeability. We observed a phosphorylation-dependent inverse relationship between AtPIP2;1 water and ion transport with a 10-fold change in both. The results revealed that phosphorylation of S280 and S283 influences the preferential facilitation of ion or water transport by AtPIP2;1. The results also hint that other regulatory sites play roles that are yet to be elucidated. Expression of the AtPIP2;1 phosphorylation mutants in Saccharomyces cerevisiae confirmed that phosphorylation influences plasma membrane localization, and revealed higher Na+ accumulation for S280A and S283D mutants. Collectively, the results show that phosphorylation in the C-terminal domain of AtPIP2;1 influences its subcellular localization and cation transport capacity.
Collapse
Affiliation(s)
- Jiaen Qiu
- ARC Centre of Excellence in Plant Energy Biology, School of Agriculture, Food and Wine, The University of Adelaide, Adelaide, Australia
| | - Samantha A McGaughey
- ARC Centre of Excellence in Plant Energy Biology, School of Agriculture, Food and Wine, The University of Adelaide, Adelaide, Australia
- ARC Centre of Excellence for Translational Photosynthesis, Division of Plant Sciences, Research School of Biology, Australian National University, Acton, Australia
| | - Michael Groszmann
- ARC Centre of Excellence for Translational Photosynthesis, Division of Plant Sciences, Research School of Biology, Australian National University, Acton, Australia
| | - Stephen D Tyerman
- ARC Centre of Excellence in Plant Energy Biology, School of Agriculture, Food and Wine, The University of Adelaide, Adelaide, Australia
| | - Caitlin S Byrt
- ARC Centre of Excellence in Plant Energy Biology, School of Agriculture, Food and Wine, The University of Adelaide, Adelaide, Australia
- ARC Centre of Excellence for Translational Photosynthesis, Division of Plant Sciences, Research School of Biology, Australian National University, Acton, Australia
| |
Collapse
|
88
|
Lodde V, Colleoni S, Tessaro I, Corbani D, Lazzari G, Luciano AM, Galli C, Franciosi F. A prematuration approach to equine IVM: considering cumulus morphology, seasonality, follicle of origin, gap junction coupling and large-scale chromatin configuration in the germinal vesicle. Reprod Fertil Dev 2020; 31:1793-1804. [PMID: 31630726 DOI: 10.1071/rd19230] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 09/13/2019] [Indexed: 12/21/2022] Open
Abstract
Several studies report that a two-step culture where mammalian oocytes are first kept under meiosis-arresting conditions (prematuration) followed by IVM is beneficial to embryo development. The most promising results were obtained by stratifying the oocyte population using morphological criteria and allocating them to different culture conditions to best meet their metabolic needs. In this study, horse oocytes were characterised to identify subpopulations that may benefit from prematuration. We investigated gap-junction (GJ) coupling, large-scale chromatin configuration and meiotic competence in compact and expanded cumulus-oocyte complexes (COCs) according to follicle size (<1, 1-2, >2cm) and season. Then we tested the effect of cilostamide-based prematuration in compact COCs collected from follicles <1 and 1-2cm in diameter on embryo development. Meiotic competence was not affected by prematuration, whereas COCs from follicles 1-2cm in diameter yielded embryos with a higher number of cells per blastocyst than oocytes that underwent direct IVM (P<0.01, unpaired Mann-Whitney test), suggesting improved developmental competence. Oocytes collected from follicles <1cm in diameter were not affected by prematuration. This study represents an extensive characterisation of the functional properties of immature horse oocytes and is the first report of the effects of cilostamide-based prematuration in horse oocyte IVM on embryo development.
Collapse
Affiliation(s)
- Valentina Lodde
- Dipartimento di Scienze Veterinarie per la Salute la Produzione Animale e la Sicurezza Alimentare 'Carlo Cantoni', Reproductive and Developmental Biology Lab, Università degli Studi di Milano, via Celoria, 10 20133 Milano, Italy
| | - Silvia Colleoni
- Laboratory of Reproductive Technologies, Avantea, Cremona, Via Porcellasco, 7f 26100 Cremona, Italy
| | - Irene Tessaro
- Dipartimento di Scienze Veterinarie per la Salute la Produzione Animale e la Sicurezza Alimentare 'Carlo Cantoni', Reproductive and Developmental Biology Lab, Università degli Studi di Milano, via Celoria, 10 20133 Milano, Italy
| | - Davide Corbani
- Dipartimento di Scienze Veterinarie per la Salute la Produzione Animale e la Sicurezza Alimentare 'Carlo Cantoni', Reproductive and Developmental Biology Lab, Università degli Studi di Milano, via Celoria, 10 20133 Milano, Italy
| | - Giovanna Lazzari
- Laboratory of Reproductive Technologies, Avantea, Cremona, Via Porcellasco, 7f 26100 Cremona, Italy; and Fondazione Avantea, Via Porcellasco, 7f 26100 Cremona, Italy
| | - Alberto M Luciano
- Dipartimento di Scienze Veterinarie per la Salute la Produzione Animale e la Sicurezza Alimentare 'Carlo Cantoni', Reproductive and Developmental Biology Lab, Università degli Studi di Milano, via Celoria, 10 20133 Milano, Italy
| | - Cesare Galli
- Laboratory of Reproductive Technologies, Avantea, Cremona, Via Porcellasco, 7f 26100 Cremona, Italy; and Fondazione Avantea, Via Porcellasco, 7f 26100 Cremona, Italy
| | - Federica Franciosi
- Dipartimento di Scienze Veterinarie per la Salute la Produzione Animale e la Sicurezza Alimentare 'Carlo Cantoni', Reproductive and Developmental Biology Lab, Università degli Studi di Milano, via Celoria, 10 20133 Milano, Italy; and Corresponding author.
| |
Collapse
|
89
|
Leal GR, Graciosa MAG, Monteiro CAS, Pasolini R, Dos Reis Camargo AJ, Oliveira CS, de Paula Vasconcelos CO, Garcia Nogueira LA, Reis Ferreira AM, Serapião RV. The SPOM-adapted IVM system improves in vitro production of bovine embryos. Theriogenology 2020; 158:277-282. [PMID: 33002771 DOI: 10.1016/j.theriogenology.2020.09.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 09/14/2020] [Accepted: 09/16/2020] [Indexed: 10/23/2022]
Abstract
This study aimed to test the effects of an IVM SPOM adaptation (SPOM-adapted IVM) on the production, total number of cells (TNC), apoptosis, and cryotolerance (post-warming survival and cytoskeleton actin integrity) of bovine IVP embryos. Two experiments were conducted with two experimental groups based on IVM treatment: A control group (TCM 199 without FCS) and an SPOM-adapted group (TCM 199 with forskolin and IBMX in pre-IVM and IVM with cilostamide). The first experiment evaluated embryo in vitro production, TNC, and apoptosis rate on D9 of development. In the second experiment, embryos were vitrified/warmed at D7 (control fresh and vitrified; SPOM-adapted fresh and vitrified) and assessed regarding post-warming survival rates and cytoskeleton actin integrity. Statistical analysis was performed using GraphPad INSTAT software at a significance level of 5%. An increase (p < 0.05) in blastocyst production was observed in the SPOM-adapted group comparing to the control group. There was no difference (p > 0.05) in the TNC or apoptosis rate between the groups. Regarding cryopreservation, no differences were found (p > 0.05) in actin integrity or post-warming survival rates between the vitrified groups. In both vitrified groups, we observed a significantly lower uninjured pattern of actin integrity compared to the fresh groups (p < 0.05). We conclude that the SPOM-adapted IVM system is beneficial for blastocyst production and does not affect the quality and cryotolerance of the produced embryos.
Collapse
Affiliation(s)
- Gabriela Ramos Leal
- Universidade Federal Fluminense (UFF), Department of Veterinary Medicine, Vital Brazil Filho St., 64, 24230-340, Niteroi, Rio de Janeiro, Brazil.
| | - Maria Alice Guimarães Graciosa
- Empresa de Pesquisa Agropecuária do Estado do Rio de Janeiro (PESAGRO RIO), São Boa Ventura Av., 770, 24120-19, Fonseca, Niteroi, Rio de Janeiro, Brazil
| | - Clara Ana Santos Monteiro
- Universidade Federal Fluminense (UFF), Department of Veterinary Medicine, Vital Brazil Filho St., 64, 24230-340, Niteroi, Rio de Janeiro, Brazil
| | - Renata Pasolini
- Empresa de Pesquisa Agropecuária do Estado do Rio de Janeiro (PESAGRO RIO), São Boa Ventura Av., 770, 24120-19, Fonseca, Niteroi, Rio de Janeiro, Brazil
| | - Agostinho Jorge Dos Reis Camargo
- Empresa de Pesquisa Agropecuária do Estado do Rio de Janeiro (PESAGRO RIO), São Boa Ventura Av., 770, 24120-19, Fonseca, Niteroi, Rio de Janeiro, Brazil
| | - Clara Slade Oliveira
- Embrapa Gado de Leite, Laboratory of Animal Reproduction, Santa Monica Experimental Field (LRA-CESM) - Santa Monica Rd., 27640-000, Valença, Rio de Janeiro, Brazil
| | - Carlos Otávio de Paula Vasconcelos
- Universidade Federal Fluminense (UFF), Department of Veterinary Medicine, Vital Brazil Filho St., 64, 24230-340, Niteroi, Rio de Janeiro, Brazil
| | - Luiz Altamiro Garcia Nogueira
- Universidade Federal Fluminense (UFF), Department of Veterinary Medicine, Vital Brazil Filho St., 64, 24230-340, Niteroi, Rio de Janeiro, Brazil
| | - Ana Maria Reis Ferreira
- Universidade Federal Fluminense (UFF), Department of Veterinary Medicine, Vital Brazil Filho St., 64, 24230-340, Niteroi, Rio de Janeiro, Brazil
| | - Raquel Varella Serapião
- Empresa de Pesquisa Agropecuária do Estado do Rio de Janeiro (PESAGRO RIO), São Boa Ventura Av., 770, 24120-19, Fonseca, Niteroi, Rio de Janeiro, Brazil; Embrapa Gado de Leite, Laboratory of Animal Reproduction, Santa Monica Experimental Field (LRA-CESM) - Santa Monica Rd., 27640-000, Valença, Rio de Janeiro, Brazil
| |
Collapse
|
90
|
Fu A, Koth ML, Brown RM, Shaw SA, Wang L, Krentz KJ, Zhang X, Hui CC, Jorgensen JS. IRX3 and IRX5 collaborate during ovary development and follicle formation to establish responsive granulosa cells in the adult mouse†. Biol Reprod 2020; 103:620-629. [PMID: 32507881 PMCID: PMC7822710 DOI: 10.1093/biolre/ioaa100] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 05/27/2020] [Accepted: 06/05/2020] [Indexed: 12/13/2022] Open
Abstract
Healthy development of ovarian follicles depends on appropriate interactions and function between oocytes and their surrounding granulosa cells. Previously, we showed that double knockout of Irx3 and Irx5 (Irx3/5 DKO) in mice resulted in abnormal follicle morphology and follicle death. Further, female mouse models of individual Irx3 or Irx5 knockouts were both subfertile but with distinct defects. Notably, the expression profile of each gene suggests independent roles for each; first, they are colocalized in pre-granulosa cells during development that then progresses to include oocyte expression during germline nest breakdown and primordial follicle formation. Thereafter, their expression patterns diverge between oocytes and granulosa cells coinciding with the formulation and maturation of intimate oocyte-granulosa cell interactions. The objective of this study was to investigate the contributions of Irx5 and somatic cell-specific expression of Irx3 during ovarian development. Our results show that Irx3 and Irx5 contribute to female fertility through different mechanisms and that Irx3 expression in somatic cells is important for oocyte quality and survival. Based on evaluation of a series of genetically modified mouse models, we conclude that IRX3 and IRX5 collaborate in the same cells and then in neighboring cells to foster a healthy and responsive follicle. Long after these two factors have extinguished, their legacy enables these intercellular connections to mature and respond to extracellular signals to promote follicle maturation and ovulation.
Collapse
Affiliation(s)
- Anqi Fu
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin–Madison, Madison, WI, USA
| | - Megan L Koth
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin–Madison, Madison, WI, USA
| | - Ryan M Brown
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin–Madison, Madison, WI, USA
| | - Sarah A Shaw
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin–Madison, Madison, WI, USA
| | - Linda Wang
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin–Madison, Madison, WI, USA
| | - Kathleen J Krentz
- Genome Editing and Animal Models Core, Biotechnology Center, University of Wisconsin–Madison, Madison, WI, USA
| | - Xiaoyun Zhang
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children and Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Chi-chung Hui
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children and Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Joan S Jorgensen
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin–Madison, Madison, WI, USA
| |
Collapse
|
91
|
Wang JJ, Niu MH, Zhang T, Shen W, Cao HG. Genome-Wide Network of lncRNA-mRNA During Ovine Oocyte Development From Germinal Vesicle to Metaphase II in vitro. Front Physiol 2020; 11:1019. [PMID: 32973554 PMCID: PMC7461901 DOI: 10.3389/fphys.2020.01019] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/24/2020] [Indexed: 12/19/2022] Open
Abstract
Long non-coding RNA (lncRNA) is involved in many biological processes, and it has been closely investigated. However, research into the role of lncRNA in ovine ovarian development is scant and poorly understood, particularly in relation to the molecular mechanisms of ovine oocyte maturation. In the current study, RNA sequencing was performed with germinal vesicle (GV) and in vitro matured metaphase II (MII) stage oocytes, isolated from ewes. Through the use of bioinformatic analysis, abundant candidate lncRNAs in stage-specific ovine oocytes were identified, and their trans- and cis-regulatory effects were deeply dissected using computational prediction software. Functional enrichment analysis of these lncRNAs revealed that they were involved in the regulation of many key signaling pathways during ovine oocyte development, which was reflected by their targeted genes. From this study, multiple lncRNA-mRNA networks were presumed to be involved in key signaling pathways regarding ovine oocyte maturation and meiotic resumption. In particular, one novel lncRNA (MSTRG.17927) appeared to mediate the regulation of phosphatidylinositol 3-kinase signaling (PI3K) signaling during ovine oocyte maturation. Therefore, this research offers novel insights into the molecular mechanisms underlying ovine oocyte meiotic maturation regulated by lncRNA-mRNA networks from a genome-wide perspective.
Collapse
Affiliation(s)
- Jun-Jie Wang
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, China.,Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | - Meng-Han Niu
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Teng Zhang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Wei Shen
- Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | - Hong-Guo Cao
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| |
Collapse
|
92
|
Pereira JL, Curcio AG, Barroso LM, Mogollón-Waltero EM, Gomes HF, Maia RC, Viana KS, Caldas Bussiere MC, Marin DFD, Dias AJB. Modulation of phosphatidylinositol 3-kinase activity during in vitro oocyte maturation increases the production of bovine blastocysts. ZYGOTE 2020; 28:1-6. [PMID: 32744197 DOI: 10.1017/s0967199420000209] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
This study aimed to evaluate the effect of regulating phosphatidylinositol 3-kinase (PI3K) activity on the kinetics of oocyte nuclear maturation and the blastocyst rate. To evaluate oocyte viability, nuclear maturation rate and in vitro embryo production, cumulus-oocyte complexes (COCs) were maintained for 0, 10 min, 6 h or 22 h in TCM 199 medium supplemented with 20 nM wortmannin, an inhibitor of PI3K. After each period, COCs were transferred to the same medium without wortmannin and kept under the same conditions until completion of 22 h of in vitro maturation (IVM). To evaluate the effect of time on progression of nuclear maturation, COCs cultivated with 20 nM wortmannin was maintained for 22, 28 or 34 h of IVM. To determine the effect of wortmannin on the activity of maturation-promoting factor (MPF), COCs were kept under IVM conditions in the presence of the inhibitor for 0, 1, 3, 6, or 8 h. Exposure of COCs to wortmannin decreased (P < 0.05) the percentage of oocytes that reached metaphase II (MII) up to 22 h, MPF activity and reduced PI3K activity by 30%. However, after 28 and 34 h, 70% of oocytes reached the MII stage in the presence of inhibitor Moreover, COCs matured in the presence of wortmannin showed an increase (P < 0.05) in the blastocyst rate. These findings suggested that the regulation of the PI3K activity during IVM of bovine COCs interfered with the meiotic progression due to control of MPF activity, positively affecting the blastocyst rate.
Collapse
Affiliation(s)
- Janaína Leite Pereira
- Laboratório de Reprodução e Melhoramento Genético Animal, Centro de Ciências e Tecnologias Agropecuárias, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Avenida Alberto Lamego, 2000, CEP: 28013-602. Campos dos Goytacazes, Rio de Janeiro, Brazil
| | - Alinne Glória Curcio
- Laboratório de Reprodução e Melhoramento Genético Animal, Centro de Ciências e Tecnologias Agropecuárias, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Avenida Alberto Lamego, 2000, CEP: 28013-602. Campos dos Goytacazes, Rio de Janeiro, Brazil
| | - Laura Mathias Barroso
- Laboratório de Reprodução e Melhoramento Genético Animal, Centro de Ciências e Tecnologias Agropecuárias, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Avenida Alberto Lamego, 2000, CEP: 28013-602. Campos dos Goytacazes, Rio de Janeiro, Brazil
| | - Edgar Mauricio Mogollón-Waltero
- Laboratório de Reprodução e Melhoramento Genético Animal, Centro de Ciências e Tecnologias Agropecuárias, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Avenida Alberto Lamego, 2000, CEP: 28013-602. Campos dos Goytacazes, Rio de Janeiro, Brazil
| | - Helga Fernandes Gomes
- Laboratório de Reprodução e Melhoramento Genético Animal, Centro de Ciências e Tecnologias Agropecuárias, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Avenida Alberto Lamego, 2000, CEP: 28013-602. Campos dos Goytacazes, Rio de Janeiro, Brazil
| | - Roger Cardoso Maia
- Laboratório de Reprodução e Melhoramento Genético Animal, Centro de Ciências e Tecnologias Agropecuárias, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Avenida Alberto Lamego, 2000, CEP: 28013-602. Campos dos Goytacazes, Rio de Janeiro, Brazil
| | - Kelen Salaroli Viana
- Laboratório de Reprodução e Melhoramento Genético Animal, Centro de Ciências e Tecnologias Agropecuárias, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Avenida Alberto Lamego, 2000, CEP: 28013-602. Campos dos Goytacazes, Rio de Janeiro, Brazil
| | - Maria Clara Caldas Bussiere
- Laboratório de Reprodução e Melhoramento Genético Animal, Centro de Ciências e Tecnologias Agropecuárias, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Avenida Alberto Lamego, 2000, CEP: 28013-602. Campos dos Goytacazes, Rio de Janeiro, Brazil
| | - Diego Fernando Dubeibe Marin
- Laboratório de Reprodução e Melhoramento Genético Animal, Centro de Ciências e Tecnologias Agropecuárias, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Avenida Alberto Lamego, 2000, CEP: 28013-602. Campos dos Goytacazes, Rio de Janeiro, Brazil
| | - Angelo José Burla Dias
- Laboratório de Reprodução e Melhoramento Genético Animal, Centro de Ciências e Tecnologias Agropecuárias, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Avenida Alberto Lamego, 2000, CEP: 28013-602. Campos dos Goytacazes, Rio de Janeiro, Brazil
| |
Collapse
|
93
|
Qasemi M, Amidi F. Extracellular microRNA profiling in human follicular fluid: new biomarkers in female reproductive potential. J Assist Reprod Genet 2020; 37:1769-1780. [PMID: 32642870 PMCID: PMC7468023 DOI: 10.1007/s10815-020-01860-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 06/10/2020] [Indexed: 12/11/2022] Open
Abstract
MicroRNAs (miRNAs) are small, about 22 nucleotides, non-coding RNAs which regulate a wide range of gene expression during post-transcriptional stage. They are released into intra- and extracellular microenvironments and play vital roles in different physiological and pathological pathways. Due to easy accessibility, detection of extracellular miRNAs in body fluids, e.g. serum, plasma, cerebrospinal fluid, and follicular fluid, has been explored in recent years. Since miRNAs are stable at unsuitable conditions, scientists have been investigating to use them as biomarkers in different fields of medicines. It goes without saying that experienced biomarkers would be required in reproductive medicine as well. Biomarkers can help clinicians and embryologists to diagnose disorders and assess the embryo quality via molecular pattern which is more reliable than nowadays routine methods. Follicular fluid as a noninvasive fluid in assisted reproductive techniques (ART) has attracted researchers as a rich pool for biomarkers, and miRNAs are not exception. Although miRNA biomarkers in reproduction field are located on their initial stage and there is a long path to move forward, several meticulous studies have been performed and discovered their associations with various conditions. In this regard, we summarize the reported miRNAs in follicular fluid and their correlations with female infertility and ART success rate, while subsequent investigations are required.
Collapse
Affiliation(s)
- Maryam Qasemi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fardin Amidi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Infertility, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
94
|
Idrees M, Oh SH, Muhammad T, El-Sheikh M, Song SH, Lee KL, Kong IK. Growth Factors, and Cytokines; Understanding the Role of Tyrosine Phosphatase SHP2 in Gametogenesis and Early Embryo Development. Cells 2020; 9:cells9081798. [PMID: 32751109 PMCID: PMC7465981 DOI: 10.3390/cells9081798] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/28/2020] [Accepted: 07/28/2020] [Indexed: 12/19/2022] Open
Abstract
Growth factors and cytokines have vital roles in germ cell development, gamete maturation, and early embryo development. Cell surface receptors are present for growth factors and cytokines to integrate with and trigger protein signaling in the germ and embryo intracellular milieu. Src-homology-2-containing phosphotyrosine phosphatase (SHP2) is a ubiquitously expressed, multifunctional protein that plays a central role in the signaling pathways involved in growth factor receptors, cytokine receptors, integrins, and G protein-coupled receptors. Over recent decades, researchers have recapitulated the protein signaling networks that influence gamete progenitor specification as well as gamete differentiation and maturation. SHP2 plays an indispensable role in cellular growth, survival, proliferation, differentiation, and migration, as well as the basic events in gametogenesis and early embryo development. SHP2, a classic cytosolic protein and a key regulator of signal transduction, displays unconventional nuclear expression in the genital organs. Several observations provided shreds of evidence that this behavior is essential for fertility. The growth factor and cytokine-dependent roles of SHP2 and its nuclear/cytoplasmic presence during gamete maturation, early embryonic development and embryo implantation are fascinating and complex subjects. This review is intended to summarize the previous and recent knowledge about the SHP2 functions in gametogenesis and early embryo development.
Collapse
Affiliation(s)
- Muhammad Idrees
- Division of Applied Life Science (BK21 Plus), Gyeongsang National University, Jinju 52828, Korea; (M.I.); (S.-H.O.); (M.E.-S.)
| | - Seon-Hwa Oh
- Division of Applied Life Science (BK21 Plus), Gyeongsang National University, Jinju 52828, Korea; (M.I.); (S.-H.O.); (M.E.-S.)
| | - Tahir Muhammad
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada;
| | - Marwa El-Sheikh
- Division of Applied Life Science (BK21 Plus), Gyeongsang National University, Jinju 52828, Korea; (M.I.); (S.-H.O.); (M.E.-S.)
- Department of Microbial Biotechnology, Genetic Engineering and Biotechnology Division, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Seok-Hwan Song
- The King Kong Ltd., Gyeongsang National University, Jinju 52828, Korea; (S.-H.S.); (K.-L.L.)
| | - Kyeong-Lim Lee
- The King Kong Ltd., Gyeongsang National University, Jinju 52828, Korea; (S.-H.S.); (K.-L.L.)
| | - Il-Keun Kong
- Division of Applied Life Science (BK21 Plus), Gyeongsang National University, Jinju 52828, Korea; (M.I.); (S.-H.O.); (M.E.-S.)
- The King Kong Ltd., Gyeongsang National University, Jinju 52828, Korea; (S.-H.S.); (K.-L.L.)
- Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Gyeongnam Province, Korea
- Correspondence: ; Tel.: +82-55-772-1942
| |
Collapse
|
95
|
Luo Y, Zhang R, Gao J, Wang Y, Zhang W, Qing S. The localization and expression of epidermal growth factor and epidermal growth factor receptor in bovine ovary during oestrous cycle. Reprod Domest Anim 2020; 55:822-832. [PMID: 32330337 DOI: 10.1111/rda.13690] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 04/14/2020] [Indexed: 12/12/2022]
Abstract
Epidermal growth factor (EGF) is one of the important regulatory factors of EGF family. EGF has been indicated to effectively inhibit the apoptosis of follicular cells, to promote the proliferation of granulosa cells and the maturation of oocytes, and to induce ovulation process via binding to epidermal growth factor receptor (EGFR). However, little is known about the distribution and expression of EGF and EGFR in cattle ovary especially during oestrous cycle. In this study, the localization and expression rule of EGF and EGFR in cattle ovaries of follicular phase and luteal phase at different time points in oestrous cycle were investigated by using IHC and real-time qPCR. The results showed that EGF and EGFR in cattle ovary were mainly expressed in granulosa cells, cumulus cells, oocytes, zona pellucida, follicular fluid and theca folliculi externa of follicles. The protein and mRNA expression of EGF/EGFR in follicles changed regularly with the follicular growth wave both in follicular and in luteal phase ovaries. In follicular phase ovaries, the protein expression of EGF and EGFR was higher in antral follicles than that of those in other follicles during follicular growth stage, and the mRNA expression of EGFR was also increased in stage of dominant follicle selection. However, in luteal phase ovaries, the growth of follicles was impeded during corpus luteum development under the action of progesterone secreted by granular lutein cell. The mRNA and protein expressions of EGF and EGFR in ovarian follicles during oestrous cycle indicate that they play a role in promoting follicular development in follicular growth waves and mediating the selection process of dominant follicles.
Collapse
Affiliation(s)
- Yuru Luo
- College of Veterinary Medicine, Northwest A&F University, Yangling, China.,Beijing Shunxin Xinyuan Research Institute of Cattle Breeding, Beijing, China
| | - Ruiqi Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Jing Gao
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Yali Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Weimin Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Suzhu Qing
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| |
Collapse
|
96
|
Snanoudj S, Molin A, Colson C, Coudray N, Paulien S, Mittre H, Gérard M, Schaefer E, Goldenberg A, Bacchetta J, Odent S, Naudion S, Demeer B, Faivre L, Gruchy N, Kottler ML, Richard N. Maternal Transmission Ratio Distortion of GNAS Loss-of-Function Mutations. J Bone Miner Res 2020; 35:913-919. [PMID: 31886927 DOI: 10.1002/jbmr.3948] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 12/09/2019] [Accepted: 12/14/2019] [Indexed: 12/14/2022]
Abstract
Pseudohypoparathyroidism type 1A (PHP1A) and pseudopseudohypoparathyroidism (PPHP) are two rare autosomal dominant disorders caused by loss-of-function mutations in the imprinted Guanine Nucleotide Binding Protein, Alpha Stimulating Activity (GNAS) gene, coding Gs α. PHP1A is caused by mutations in the maternal allele and results in Albright's hereditary osteodystrophy (AHO) and hormonal resistance, mainly to the parathormone (PTH), whereas PPHP, with AHO features and no hormonal resistance, is linked to mutations in the paternal allele. This study sought to investigate parental transmission of GNAS mutations. We conducted a retrospective study in a population of 204 families with 361 patients harboring GNAS mutations. To prevent ascertainment bias toward a higher proportion of affected children due to the way in which data were collected, we excluded from transmission analysis all probands in the ascertained sibships. After bias correction, the distribution ratio of the mutated alleles was calculated from the observed genotypes of the offspring of nuclear families and was compared to the expected ratio of 50% according to Mendelian inheritance (one-sample Z-test). Sex ratio, phenotype of the transmitting parent, and transmission depending on the severity of the mutation were also analyzed. Transmission analysis was performed in 114 nuclear families and included 250 descendants. The fertility rates were similar between male and female patients. We showed an excess of transmission from mother to offspring of mutated alleles (59%, p = .022), which was greater when the mutations were severe (61.7%, p = .023). Similarly, an excess of transmission was found when the mother had a PHP1A phenotype (64.7%, p = .036). By contrast, a Mendelian distribution was observed when the mutations were paternally inherited. Higher numbers of females within the carriers, but not in noncarriers, were also observed. The mother-specific transmission ratio distortion (TRD) and the sex-ratio imbalance associated to PHP1A point to a role of Gs α in oocyte biology or embryogenesis, with implications for genetic counseling. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Sarah Snanoudj
- Normandie Université, UNICAEN, CHU de Caen Normandie, Department of Genetics, Reference Center for Rare Diseases of Calcium and Phosphorus Metabolism, EA7450 BioTARGen, Caen, France
| | - Arnaud Molin
- Normandie Université, UNICAEN, CHU de Caen Normandie, Department of Genetics, Reference Center for Rare Diseases of Calcium and Phosphorus Metabolism, EA7450 BioTARGen, Caen, France
| | - Cindy Colson
- Normandie Université, UNICAEN, CHU de Caen Normandie, Department of Genetics, Reference Center for Rare Diseases of Calcium and Phosphorus Metabolism, EA7450 BioTARGen, Caen, France
| | - Nadia Coudray
- Normandie Université, UNICAEN, CHU de Caen Normandie, Department of Genetics, Reference Center for Rare Diseases of Calcium and Phosphorus Metabolism, EA7450 BioTARGen, Caen, France
| | - Sylvie Paulien
- Normandie Université, UNICAEN, CHU de Caen Normandie, Department of Genetics, Reference Center for Rare Diseases of Calcium and Phosphorus Metabolism, EA7450 BioTARGen, Caen, France
| | - Hervé Mittre
- Normandie Université, UNICAEN, CHU de Caen Normandie, Department of Genetics, Reference Center for Rare Diseases of Calcium and Phosphorus Metabolism, EA7450 BioTARGen, Caen, France
| | - Marion Gérard
- Normandie Université, UNICAEN, CHU de Caen Normandie, Department of Genetics, Reference Center for Rare Diseases of Calcium and Phosphorus Metabolism, EA7450 BioTARGen, Caen, France
| | - Elise Schaefer
- Department of Genetics, CHU de Strasbourg, Strasbourg, France
| | | | - Justine Bacchetta
- Department of Pediatric Nephrology, Rheumatology and Dermatology, CHU de Lyon, Bron, France
| | - Sylvie Odent
- Department of Genetics, CHU de Rennes, Rennes, France
| | - Sophie Naudion
- Department of Genetics, CHU de Bordeaux, Bordeaux, France
| | | | | | - Nicolas Gruchy
- Normandie Université, UNICAEN, CHU de Caen Normandie, Department of Genetics, Reference Center for Rare Diseases of Calcium and Phosphorus Metabolism, EA7450 BioTARGen, Caen, France
| | - Marie-Laure Kottler
- Normandie Université, UNICAEN, CHU de Caen Normandie, Department of Genetics, Reference Center for Rare Diseases of Calcium and Phosphorus Metabolism, EA7450 BioTARGen, Caen, France
| | - Nicolas Richard
- Normandie Université, UNICAEN, CHU de Caen Normandie, Department of Genetics, Reference Center for Rare Diseases of Calcium and Phosphorus Metabolism, EA7450 BioTARGen, Caen, France
| |
Collapse
|
97
|
Zhang H, McClatchie T, Baltz JM. l-Serine transport in growing and maturing mouse oocytes. J Cell Physiol 2020; 235:8585-8600. [PMID: 32329057 DOI: 10.1002/jcp.29702] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/27/2020] [Accepted: 03/28/2020] [Indexed: 01/24/2023]
Abstract
Serine has roles in cell metabolism besides protein synthesis including providing one-carbon units to the folate cycle. Since growing mouse oocytes undergo a burst of folate accumulation as they near full size, we have investigated whether oocytes transport serine. Substantial serine transport appeared in oocytes near the end of their growth. Serine transport continued when oocytes resumed meiosis but ceased partway through first meiotic metaphase, remaining quiescent in mature eggs in second meiotic metaphase. The serine transporter was sodium dependent and inhibited by alanine, cysteine, leucine, or histidine, and had a Michaelis-Menten constant (Km ) for serine of 200 µM. Unexpectedly, exposing cumulus cell-enclosed oocytes to the physiological mediator of meiotic arrest, natriuretic peptide precursor Type C, substantially stimulated serine transport by the enclosed oocyte. Finally, in addition to transport by the oocyte itself, cumulus cells also supply serine to the enclosed oocyte via gap junctions within intact cumulus-oocyte complexes.
Collapse
Affiliation(s)
- Han Zhang
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Division of Reproductive Medicine, Department of Obstetrics and Gynecology, University of Ottawa Faculty of Medicine, Ottawa, Ontario, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa Faculty of Medicine, Ottawa, Ontario, Canada
| | - Taylor McClatchie
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Division of Reproductive Medicine, Department of Obstetrics and Gynecology, University of Ottawa Faculty of Medicine, Ottawa, Ontario, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa Faculty of Medicine, Ottawa, Ontario, Canada
| | - Jay M Baltz
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Division of Reproductive Medicine, Department of Obstetrics and Gynecology, University of Ottawa Faculty of Medicine, Ottawa, Ontario, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa Faculty of Medicine, Ottawa, Ontario, Canada
| |
Collapse
|
98
|
Gupta A, Chaube SK. Cilostamide and rolipram prevent spontaneous meiotic resumption from diplotene arrest in rat oocytes cultured in vitro. Eur J Pharmacol 2020; 878:173115. [PMID: 32302597 DOI: 10.1016/j.ejphar.2020.173115] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 03/29/2020] [Accepted: 04/09/2020] [Indexed: 11/17/2022]
Abstract
The involvement of specific phosphodiesterases (PDEs) in the modulation of cAMP and thereby spontaneous meiotic resumption remains poorly understood. This work aims to evaluate the effects of cilostamide and rolipram (PDE 3A and PDE 4D inhibitors) on spontaneous meiotic resumption from diplotene arrest in rat oocytes cultured in vitro. For this purpose, diplotene-arrested cumulus oocyte complexes (COCs) were collected from rat ovary. The COCs and denuded oocytes were exposed to various concentrations of cilostamide (0.0, 2.5, 5.0 and 10 μM) and rolipram (0, 10, 50 and 100 μM) for various times (0, 3, 5, 7, 14, 16, 18, 20, 22 and 24 h). Cilostamide inhibited spontaneous meiotic resumption in a concentration- and time-dependent manner in COCs and denuded oocytes. Although rolipram showed inhibition of spontaneous meiotic resumption up to some extent, cilostamide was more potent to prevent spontaneous meiotic resumption in both COCs and denuded oocytes. Cilostamide significantly reduced PDE 3A expression, increased cAMP level and prevented spontaneous meiotic resumption in COCs and denuded oocytes. Although rolipram inhibited PDE 4D expression in cumulus cells, increased cAMP level but was not sufficient to prevent spontaneous meiotic resumption. We conclude that both drugs prevent spontaneous resumption from diplotene-arrest through PDE 3A/PDE 4D-cAMP mediated pathway. However, as compare to rolipram, cilostamide was more potent in preventing spontaneous resumption from diplotene-arrest in rat oocytes cultured in vitro. Thus, cilostamide could be used as a potential candidate for the development of female contraceptive drug in future.
Collapse
Affiliation(s)
- Anumegha Gupta
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India
| | - Shail K Chaube
- Cell Physiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India.
| |
Collapse
|
99
|
Tesfaye D, Hailay T, Salilew-Wondim D, Hoelker M, Bitseha S, Gebremedhn S. Extracellular vesicle mediated molecular signaling in ovarian follicle: Implication for oocyte developmental competence. Theriogenology 2020; 150:70-74. [PMID: 32088041 DOI: 10.1016/j.theriogenology.2020.01.075] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 01/29/2020] [Indexed: 12/18/2022]
Abstract
The bidirectional communication between the oocyte and the companion somatic cells in the follicular environment is known to be mediated by either a direct communication via gap junction or transzonal projections or indirectly through endocrine, paracrine and autocrine signaling factors. Extracellular vesicles (EVs), which are found in various biological fluids, including follicular fluid (FF) are known to play important roles in mediating the communication between the oocyte and the surrounding somatic cells through shuttling bioactive molecules to facilitate follicular growth and oocyte maturation. As vesicles in the extracellular space are known to reflect the physiological status of the donor or the releasing cells, molecules carried by the EVs in the follicular environment could be markers of the internal and external stressors. EVs exhibit greater degree of heterogeneity in their size, biogenesis and the bioactive molecule they carry. The process of biogenesis of EVs is known to be regulated by several proteins associated with the endosomal sorting complex required for transport (ESCRT) proteins. The type of EVs and surface proteins markers vary according to the type of protein involved in their biogenesis. EVs are recently reported to play indispensable role in promoting cell-to-cell communication during follicular growth. Recent advancements in EV research opened the possibilities to load EVs with specific molecules like miRNA, siRNA, CRISPR-cas9 complex and protein, which showed a new horizon for their application in therapeutics. The present review explores the biogenesis, the role and the future prospects of EVs with a special emphasis given to follicular growth and oocyte maturation.
Collapse
Affiliation(s)
- Dawit Tesfaye
- Department of Biomedical Sciences, Animal Reproduction and Biotechnology Laboratory (ARBL), Colorado State University, Fort Collins, CO, USA.
| | - Tsige Hailay
- Institute of Animal Sciences, Department of Animal Breeding and Husbandry, University of Bonn, Bonn, Germany
| | - Dessie Salilew-Wondim
- Institute of Animal Sciences, Department of Animal Breeding and Husbandry, University of Bonn, Bonn, Germany
| | - Michael Hoelker
- Institute of Animal Sciences, Department of Animal Breeding and Husbandry, University of Bonn, Bonn, Germany
| | - Simret Bitseha
- Hawassa University, College of Agriculture, Department of Animal Sciences, Hawassa, Ethiopia
| | - Samuel Gebremedhn
- Department of Biomedical Sciences, Animal Reproduction and Biotechnology Laboratory (ARBL), Colorado State University, Fort Collins, CO, USA
| |
Collapse
|
100
|
Braga DPDAF, Zanetti BF, Setti AS, Iaconelli A, Borges E. Immature oocyte incidence: Contributing factors and effects on mature sibling oocytes in intracytoplasmic sperm injection cycles. JBRA Assist Reprod 2020; 24:70-76. [PMID: 31589389 PMCID: PMC6993156 DOI: 10.5935/1518-0557.20190056] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Objective The aim of this study was to investigate which factors contribute to the incidence of immature oocytes (germinal vesicle -GV- and metaphase I -MI-) and how they impact the intracytoplasmic sperm injection (ICSI) outcomes of sibling mature oocytes. Methods Data from 3,920 cycles performed from June/2010 to August/2016 in a private university-affiliated IVF center were evaluated for the influence of controlled ovarian stimulation protocol (COS) on immature oocytes incidence and its effects on ICSI outcomes. Results MI (p=0.004) and GV (p=0.029) number were negatively correlated with gonadotropin dose. Patients stimulated by rFSH had increased GV/oocyte rate in both GnRH agonists (p<0.001) and antagonist (p=0.042) protocols, in comparison to rFSH associated with rLH protocol. MI and GV/oocyte rates were negatively correlated to fertilization (p<0.001), high-quality embryo on da p<0.001; GV/oocyte p=0.033) and pregnancy (MI/oocyte p=0.002; GV/oocyte p=0.013) rates. Cycles above a 10.5% MI/oocyte cut-off were correlated to higher response to ovarian stimulation, poor embryo development and almost two times lower pregnancy rate. Immature oocyte incidence is affected by COS and impacts on ICSI outcomes. Conclusion Our evidence suggests that oocytes derived from a cohort with high incidence of maturation fail may have detrimental clinical outcomes.
Collapse
Affiliation(s)
- Daniela Paes de Almeida Ferreira Braga
- Fertility Medical Group, São Paulo, SP - Brazil.,Instituto Sapientiae - Centro de Estudos e Pesquisa em Reprodução Humana Assistida, São Paulo, SP - Brazil
| | - Bianca Ferrarini Zanetti
- Fertility Medical Group, São Paulo, SP - Brazil.,Instituto Sapientiae - Centro de Estudos e Pesquisa em Reprodução Humana Assistida, São Paulo, SP - Brazil
| | - Amanda Souza Setti
- Fertility Medical Group, São Paulo, SP - Brazil.,Instituto Sapientiae - Centro de Estudos e Pesquisa em Reprodução Humana Assistida, São Paulo, SP - Brazil
| | - Assumpto Iaconelli
- Fertility Medical Group, São Paulo, SP - Brazil.,Instituto Sapientiae - Centro de Estudos e Pesquisa em Reprodução Humana Assistida, São Paulo, SP - Brazil
| | - Edson Borges
- Fertility Medical Group, São Paulo, SP - Brazil.,Instituto Sapientiae - Centro de Estudos e Pesquisa em Reprodução Humana Assistida, São Paulo, SP - Brazil
| |
Collapse
|