1
|
Wang H. The role of granulosa cells in oocyte development and aging: Mechanisms and therapeutic opportunities. Semin Cell Dev Biol 2025; 171:103614. [PMID: 40300217 DOI: 10.1016/j.semcdb.2025.103614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/20/2025] [Accepted: 04/14/2025] [Indexed: 05/01/2025]
Abstract
Granulosa cells (GCs) are essential for oocyte maturation, providing metabolic support, hormonal signaling, and structural integrity critical to successful follicular development. However, advancing age disrupts these functions, driven by factors such as increased oxidative stress, mitochondrial dysfunction, and transcriptomic and proteomic alterations. These age-related changes in GCs contribute to compromised oocyte quality, diminished follicular support, and a decline in fertility, particularly in women of advanced maternal age. This review highlights recent progress in understanding the pivotal roles of GCs in maintaining oocyte health, with a focus on the mechanisms underlying their aging-related dysfunction. Furthermore, we explore promising therapeutic strategies, including antioxidant therapies, metabolic modulators, and GC-based rejuvenation techniques, aimed at mitigating the impacts of reproductive aging. By consolidating and analyzing existing research, this review provides valuable perspectives on fertility preservation and factors shaping reproductive outcomes in women of advanced maternal age.
Collapse
Affiliation(s)
- HaiYang Wang
- Institute of Modern Biology, Nanjing University, Nanjing 210008, China; Mechanobiology Institute, National University of Singapore, 5A Engineering Drive 1, 117411, Singapore.
| |
Collapse
|
2
|
Klabnik JL, Beever JE, Payton RR, Lamour KH, Schrick FN, Edwards JL. A Step Toward Understanding Direct Impacts of a Higher Estrus-Associated Temperature (HEAT): Transcript Level Changes in Cumulus-Oocyte Complexes Directly Exposed to Acute Elevated Temperature. Animals (Basel) 2025; 15:517. [PMID: 40003000 PMCID: PMC11851612 DOI: 10.3390/ani15040517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/31/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Elevated body temperature (HEAT) in sexually receptive females is a normal part of the periovulatory microenvironment. The objective was to identify direct (first 6 h) and delayed (4 h or 18 h of recovery) effects at 41 °C exposure during in vitro maturation (IVM) on transcripts involved in steroidogenesis, oocyte maturation, or previously impacted by elevated temperature using targeted RNA-sequencing. Most transcripts (72.3%) were impacted in the first 2 to 4 hIVM. Twelve of the fifteen transcripts first impacted at 4 hIVM had a higher abundance and three had a lower abundance. Direct exposure to 41 °C impacted the transcripts related to progesterone production and signaling, germinal vesicle breakdown, oocyte meiotic progression, transcriptional activity and/or alternative splicing, cell cycle, cumulus expansion, and/or ovulation. Three transcripts demonstrated a delayed impact; changes were not seen until the COCs recovered for 4 h. The use of multidimensional scaling plots to 'visualize' samples highlights that oocytes exposed to an acute elevation in temperature are more advanced at the molecular level during the initial stages of maturation. Described efforts represent important steps towards providing a novel insight into the dynamic physiology of the COC in the estrual female bovid, during HEAT and after body temperature returns to baseline.
Collapse
Affiliation(s)
- Jessica L. Klabnik
- Department of Animal Science, University of Tennessee Institute of Agriculture, Knoxville, TN 37996, USA; (J.E.B.); (R.R.P.); (F.N.S.)
| | - Jonathan E. Beever
- Department of Animal Science, University of Tennessee Institute of Agriculture, Knoxville, TN 37996, USA; (J.E.B.); (R.R.P.); (F.N.S.)
- Department of Large Animal Clinical Sciences, University of Tennessee Institute of Agriculture, Knoxville, TN 37996, USA
| | - Rebecca R. Payton
- Department of Animal Science, University of Tennessee Institute of Agriculture, Knoxville, TN 37996, USA; (J.E.B.); (R.R.P.); (F.N.S.)
| | - Kurt H. Lamour
- Department of Entomology and Plant Pathology, University of Tennessee Institute of Agriculture, Knoxville, TN 37996, USA;
| | - F. Neal Schrick
- Department of Animal Science, University of Tennessee Institute of Agriculture, Knoxville, TN 37996, USA; (J.E.B.); (R.R.P.); (F.N.S.)
| | - J. Lannett Edwards
- Department of Animal Science, University of Tennessee Institute of Agriculture, Knoxville, TN 37996, USA; (J.E.B.); (R.R.P.); (F.N.S.)
| |
Collapse
|
3
|
Owen CM, Jaffe LA. Luteinizing hormone-induced changes in the structure of mammalian preovulatory follicles. Curr Top Dev Biol 2024; 162:259-282. [PMID: 40180511 DOI: 10.1016/bs.ctdb.2024.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
Ovulation of a mammalian oocyte from its follicle, which occurs in response to luteinizing hormone (LH), requires complex restructuring of the ∼20 layers of surrounding somatic cells. This chapter describes the cellular architecture of preovulatory follicles, the localization of the receptors for LH, and the LH-induced changes in follicular structure, focusing on mice and other small mammals. The multiple interrelated processes that result in ovulation include breakdown of existing extracellular matrix, generation of new extracellular matrix, thinning of the follicular apex where the oocyte will be released, invagination of the follicular surface, and responses of the vascular system to support these dynamic changes. However, much remains unknown about how these events function together to release a fertilizable egg.
Collapse
Affiliation(s)
- Corie M Owen
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT, United States.
| | - Laurinda A Jaffe
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT, United States.
| |
Collapse
|
4
|
Thomas C, Marx TL, Penir SM, Schuh M. Ex vivo imaging reveals the spatiotemporal control of ovulation. Nat Cell Biol 2024; 26:1997-2008. [PMID: 39415041 PMCID: PMC11567896 DOI: 10.1038/s41556-024-01524-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 09/10/2024] [Indexed: 10/18/2024]
Abstract
During ovulation, an egg is released from an ovarian follicle, ready for fertilization. Ovulation occurs inside the body, impeding direct studies of its progression. Therefore, the exact mechanisms that control ovulation have remained unclear. Here we devised live imaging methods to study the entire process of ovulation in isolated mouse ovarian follicles. We show that ovulation proceeds through three distinct phases, follicle expansion (I), contraction (II) and rupture (III), culminating in the release of the egg. Follicle expansion is driven by hyaluronic acid secretion and an osmotic gradient-directed fluid influx into the follicle. Then, smooth muscle cells in the outer follicle drive follicle contraction. Follicle rupture begins with stigma formation, followed by the exit of follicular fluid and cumulus cells and the rapid release of the egg. These results establish a mechanistic framework for ovulation, a process of fundamental importance for reproduction.
Collapse
Affiliation(s)
- Christopher Thomas
- Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- IBDM, CNRS - UMR 7288, Aix-Marseille Université, Marseille, France
| | - Tabea Lilian Marx
- Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Promotionskolleg für Medizinstudierende, University Medical Center Göttingen, Göttingen, Germany
| | - Sarah Mae Penir
- Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Melina Schuh
- Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
- Cluster of Excellence 'Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells', University of Göttingen, Göttingen, Germany.
| |
Collapse
|
5
|
Zhu Y, Zhu H, Wu P. Gap junctions in polycystic ovary syndrome: Implications for follicular arrest. Dev Dyn 2024; 253:882-894. [PMID: 38501340 DOI: 10.1002/dvdy.706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 02/24/2024] [Accepted: 02/28/2024] [Indexed: 03/20/2024] Open
Abstract
Gap junctions are specialized intercellular conduits that provide a direct pathway between neighboring cells, which are involved in numerous physiological processes, such as cellular differentiation, cell growth, and metabolic coordination. The effect of gap junctional hemichannels in folliculogenesis is particularly obvious, and the down-regulation of connexins is related to abnormal follicle growth. Polycystic ovary syndrome (PCOS) is a ubiquitous endocrine disorder of the reproductive system, affecting the fertility of adult women due to anovulation. Exciting evidence shows that gap junction is involved in the pathological process related to PCOS and affects the development of follicles in women with PCOS. In this review, we examine the expression of connexins in follicular cells of PCOS and figure out whether such communication could have consequences for PCOS women. While along with results from clinical and related animal studies, we summarize the mechanism of connexins involved in the pathogenesis of PCOS.
Collapse
Affiliation(s)
- Ying Zhu
- Department of Gynaecology, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Hongqiu Zhu
- Department of Gynaecology, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Peijuan Wu
- Department of Gynaecology, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
6
|
Machaty Z. The signal that stimulates mammalian embryo development. Front Cell Dev Biol 2024; 12:1474009. [PMID: 39355121 PMCID: PMC11442298 DOI: 10.3389/fcell.2024.1474009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 09/05/2024] [Indexed: 10/03/2024] Open
Abstract
Embryo development is stimulated by calcium (Ca2+) signals that are generated in the egg cytoplasm by the fertilizing sperm. Eggs are formed via oogenesis. They go through a cell division known as meiosis, during which their diploid chromosome number is halved and new genetic combinations are created by crossing over. During formation the eggs also acquire cellular components that are necessary to produce the Ca2+ signal and also, to support development of the newly formed embryo. Ionized calcium is a universal second messenger used by cells in a plethora of biological processes and the eggs develop a "toolkit", a set of molecules needed for signaling. Meiosis stops twice and these arrests are controlled by a complex interaction of regulatory proteins. The first meiotic arrest lasts until after puberty, when a luteinizing hormone surge stimulates meiotic resumption. The cell cycle proceeds to stop again in the middle of the second meiotic division, right before ovulation. The union of the female and male gametes takes place in the oviduct. Following gamete fusion, the sperm triggers the release of Ca2+ from the egg's intracellular stores which in mammals is followed by repetitive Ca2+ spikes known as Ca2+ oscillations in the cytosol that last for several hours. Downstream sensor proteins help decoding the signal and stimulate other molecules whose actions are required for proper development including those that help to prevent the fusion of additional sperm cells to the egg and those that assist in the release from the second meiotic arrest, completion of meiosis and entering the first mitotic cell division. Here I review the major steps of egg formation, discuss the signaling toolkit that is essential to generate the Ca2+ signal and describe the steps of the signal transduction mechanism that activates the egg's developmental program and turns it into an embryo.
Collapse
Affiliation(s)
- Zoltan Machaty
- Department of Animal Sciences Purdue University West Lafayette, West Lafayette, IN, United States
| |
Collapse
|
7
|
Rulli SB, Ahtiainen P, Ratner LD, Jonas K, Calandra RS, Poutanen M, Huhtaniemi I. Elevated chorionic gonadotropic hormone in transgenic mice induces parthenogenetic activation and ovarian teratomas. Mol Cell Endocrinol 2024; 587:112214. [PMID: 38537882 DOI: 10.1016/j.mce.2024.112214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 02/25/2024] [Accepted: 03/19/2024] [Indexed: 04/13/2024]
Abstract
Both male and female reproductive functions are impacted by altered gonadotrophin secretion and action, which may also influence the development of endocrine tumours. To ascertain if chronic hypersecretion of human chorionic gonadotropin (hCG) contributes to the development of gonadal tumours, double transgenic (TG) mice that overexpress hCGα- and β-subunits were analysed. By the age of two months, ovarian tumours with characteristics of teratomas developed with 100% penetrance. Teratomas were also seen in wild-type ovaries orthotopically transplanted into TG mice, demonstrating an endocrine/paracrine mechanism for the hCG-induced ovarian tumorigenesis. Both in vitro and in vivo experiments showed oocyte parthenogenetic activation in TG females. In addition, ovaries showed reduced ovulatory gene expression, inhibited ERK1/2 phosphorylation, and impaired cumulus cell expansion. Hence, persistently high endocrine hCG activity causes parthenogenetic activation and development of ovarian teratomas, along with altered follicle development and impaired ERK1/2 signalling, offering a novel mechanism associated with the molecular pathogenesis of ovarian teratomas.
Collapse
Affiliation(s)
- Susana B Rulli
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Kiinamyllynkatu 10, FIN-20520, Turku, Finland; Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, 1428, Buenos Aires, Argentina; Centro de Estudios Biomédicos Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Hidalgo 775, C1405BCK, Ciudad Autónoma de Buenos Aires, Argentina.
| | - Petteri Ahtiainen
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Kiinamyllynkatu 10, FIN-20520, Turku, Finland
| | - Laura D Ratner
- Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, 1428, Buenos Aires, Argentina
| | - Kim Jonas
- Department of Digestion, Metabolism and Reproduction, Institute of Reproductive and Developmental Biology, Hammersmith Campus, Imperial College London, London, W12 0NN, UK; Department of Women and Children's Health, School of Population and Life Course Sciences, King's College London, London, SE1 1UL, UK
| | - Ricardo S Calandra
- Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, 1428, Buenos Aires, Argentina
| | - Matti Poutanen
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Kiinamyllynkatu 10, FIN-20520, Turku, Finland; Turku Center for Disease Modeling, University of Turku, Kiinamyllynkatu 10, FIN-20520, Turku, Finland
| | - Ilpo Huhtaniemi
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Kiinamyllynkatu 10, FIN-20520, Turku, Finland; Department of Digestion, Metabolism and Reproduction, Institute of Reproductive and Developmental Biology, Hammersmith Campus, Imperial College London, London, W12 0NN, UK
| |
Collapse
|
8
|
Zhao M, Subudeng G, Zhao Y, Hao S, Li H. Effect of Cyclic Adenosine Monophosphate on Connexin 37 Expression in Sheep Cumulus-Oocyte Complexes. J Dev Biol 2024; 12:10. [PMID: 38651455 PMCID: PMC11036199 DOI: 10.3390/jdb12020010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 03/10/2024] [Accepted: 03/26/2024] [Indexed: 04/25/2024] Open
Abstract
Gap junctional connection (GJC) in the cumulus-oocyte complex (COC) provides necessary support for message communication and nutrient transmission required for mammalian oocyte maturation. Cyclic adenosine monophosphate (cAMP) is not only a prerequisite for regulating oocyte meiosis, but also the key intercellular factor for affecting GJC function in COCs. However, there are no reports on whether cAMP regulates connexin 37 (Cx37) expression, one of the main connexin proteins, in sheep COCs. In this study, the expression of Cx37 protein and gene in immature sheep COC was detected using immunohistochemistry and PCR. Subsequently, the effect of cAMP on Cx37 expression in sheep COCs cultured in a gonadotropin-free culture system for 10 min or 60 min was evaluated using competitive ELISA, real-time fluorescent quantitative PCR (RT-qPCR), and Western blot. The results showed that the Cx37 protein was present in sheep oocytes and cumulus cells; the same results were found with respect to GJA4 gene expression. In the gonadotropin-free culture system, compared to the control, significantly higher levels of cAMP as well as Cx37 gene and protein expression were found in sheep COCs following treatment in vitro with Forskolin and IBMX (100 μM and 500 μM)) for 10 min (p < 0.05). Compared to the controls (at 10 or 60 min), cAMP levels in sheep COCs were significantly elevated as a result of Forskolin and IBMX treatment (p < 0.05). Following culturing in vitro for 10 min or 60 min, Forskolin and IBMX treatment can significantly promote Cx37 expression in sheep COCs (p < 0.05), a phenomenon which can be counteracted when the culture media is supplemented with RP-cAMP, a cAMP-specific competitive inhibitor operating through suppression of the protein kinase A (PKA). In summary, this study reports the preliminary regulatory mechanism of cAMP involved in Cx37 expression for the first time, and provides a novel explanation for the interaction between cAMP and GJC communication during sheep COC culturing in vitro.
Collapse
Affiliation(s)
- Mengyao Zhao
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China; (M.Z.); (G.S.); (Y.Z.); (S.H.)
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Inner Mongolia Agricultural University, Hohhot 010018, China
- Key Laboratory of Animal Embryo and Development Engineering of Autonomous Region Universities, Hohhot 010018, China
| | - Gerile Subudeng
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China; (M.Z.); (G.S.); (Y.Z.); (S.H.)
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Yufen Zhao
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China; (M.Z.); (G.S.); (Y.Z.); (S.H.)
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Shaoyu Hao
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China; (M.Z.); (G.S.); (Y.Z.); (S.H.)
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Haijun Li
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China; (M.Z.); (G.S.); (Y.Z.); (S.H.)
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Inner Mongolia Agricultural University, Hohhot 010018, China
- Key Laboratory of Animal Embryo and Development Engineering of Autonomous Region Universities, Hohhot 010018, China
| |
Collapse
|
9
|
Owen CM, Jaffe LA. Luteinizing hormone stimulates ingression of mural granulosa cells within the mouse preovulatory follicle†. Biol Reprod 2024; 110:288-299. [PMID: 37847612 PMCID: PMC10873281 DOI: 10.1093/biolre/ioad142] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/25/2023] [Accepted: 10/13/2023] [Indexed: 10/19/2023] Open
Abstract
Luteinizing hormone (LH) induces ovulation by acting on its receptors in the mural granulosa cells that surround a mammalian oocyte in an ovarian follicle. However, much remains unknown about how activation of the LH receptor modifies the structure of the follicle such that the oocyte is released and the follicle remnants are transformed into the corpus luteum. The present study shows that the preovulatory surge of LH stimulates LH receptor-expressing granulosa cells, initially located almost entirely in the outer layers of the mural granulosa, to rapidly extend inwards, intercalating between other cells. The cellular ingression begins within 30 min of the peak of the LH surge, and the proportion of LH receptor-expressing cell bodies in the inner half of the mural granulosa layer increases until the time of ovulation, which occurs at about 10 h after the LH peak. During this time, many of the initially flask-shaped cells appear to detach from the basal lamina, acquiring a rounder shape with multiple filipodia. Starting at about 4 h after the LH peak, the mural granulosa layer at the apical surface of the follicle where ovulation will occur begins to thin, and the basolateral surface develops invaginations and constrictions. Our findings raise the question of whether LH stimulation of granulosa cell ingression may contribute to these changes in the follicular structure that enable ovulation.
Collapse
Affiliation(s)
- Corie M Owen
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT, USA
| | - Laurinda A Jaffe
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT, USA
| |
Collapse
|
10
|
Sidell N, Rajakumar A. Retinoic Acid Action in Cumulus Cells: Implications for Oocyte Development and In Vitro Fertilization. Int J Mol Sci 2024; 25:1709. [PMID: 38338985 PMCID: PMC10855907 DOI: 10.3390/ijms25031709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/15/2024] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
In the field of human in vitro fertilization (IVF), selecting the best oocyte for freezing or embryo for transfer remains an important focus of clinical practice. Although several techniques are and have been used for this goal, results have generally not been favorable and/or are invasive such that damage to some embryos occurs, resulting in a reduced number of healthy births. Therefore, the search continues for non-invasive oocyte and embryo quality markers that signal the development of high-quality embryos. Multiple studies indicate the important positive effects of retinoic acid (RA) on oocyte maturation and function. We previously showed that a high follicular fluid (FF) RA concentration at the time of oocyte retrieval in IVF protocols was associated with oocytes, giving rise to the highest quality embryos, and that cumulus granulosa cells (CGCs) are the primary source of follicle RA synthesis. Data also demonstrated that connexin-43 (Cx43), the main connexin that forms gap junctions in CGCs, is regulated by RA and that RA induces a rapid increase in gap junction communication. Here, we hypothesize that CGC RA plays a causal role in oocyte competency through its action on Cx43 and, as such, may serve as a biomarker of oocyte competence. Multiple studies have demonstrated the requirement for Cx43 in CGCs for the normal progression of folliculogenesis, and that the increased expression of this connexin is linked to the improved developmental competence of the oocyte. The data have shown that RA can up-regulate gap junction intercellular communication (GJIC) in the cumulus-oocyte complex via a non-genomic mechanism that results in the dephosphorylation of Cx43 and enhanced GJIC. Recognizing the positive role played by gap junctions in CGCs in oocyte development and the regulation of Cx43 by RA, the findings have highlighted the possibility that CGC RA levels may serve as a non-invasive indicator for selecting high-quality oocytes for IVF procedures. In addition, the data suggest that the manipulation of Cx43 with retinoid compounds could provide new pharmacological approaches to improve IVF outcomes in cases of failed implantation, recurrent miscarriage, or in certain diseases that are characterized by reduced fecundity, such as endometriosis.
Collapse
Affiliation(s)
- Neil Sidell
- Department of Gynecology & Obstetrics, Emory University School of Medicine, Atlanta, GA 30322, USA;
| | | |
Collapse
|
11
|
Norris RP, Jaffe LA. Granulosa Cells Alone, Without Theca Cells, Can Mediate LH-induced Oocyte Meiotic Resumption. Endocrinology 2024; 165:bqad200. [PMID: 38180498 PMCID: PMC10805430 DOI: 10.1210/endocr/bqad200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/26/2023] [Accepted: 12/29/2023] [Indexed: 01/06/2024]
Abstract
Signaling in the granulosa cells of mammalian ovarian follicles is necessary for maintaining prophase arrest in the oocyte and for mediating the resumption of meiosis in response to luteinizing hormone (LH). However, the follicle also includes an outer layer of theca cells, some of which express receptors for LH. To investigate whether theca cells are required for maintaining meiotic arrest and reinitiating meiosis in response to LH, we mechanically separated the granulosa cells and oocyte from the theca and basal lamina. This was accomplished by cutting a slit in the outer surface of isolated follicles such that the mural granulosa cells and cumulus-oocyte complex were extruded from the theca shell, forming a lawn of cells on an organotypic membrane. The remnant of theca cells and basal lamina was then removed. The separation of the granulosa cells from the theca cells and basal lamina was demonstrated by immunofluorescence localization of endomucin (blood vessels of the theca) and laminin gamma (basal lamina). Cells comprising these granulosa cell-oocyte complexes expressed LH receptors and were connected by gap junctions. Oocytes within these granulosa cell complexes maintained meiotic arrest and resumed meiosis in response to LH, showing that the granulosa cells alone, without theca cells, transduce these signals. This semi-intact and mostly 2-dimensional preparation could facilitate imaging studies of follicle physiology.
Collapse
Affiliation(s)
- Rachael P Norris
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Laurinda A Jaffe
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT 06030, USA
| |
Collapse
|
12
|
Cheng M, Chen X, Han M, Luo X, Yu Y, Lv Y, Han Y, Cao L, Zhang J, Wang M, Jin Y. miR-155-5p improves oocyte maturation in porcine cumulus cells through connexin 43-mediated regulation of MPF activity. Theriogenology 2024; 214:124-133. [PMID: 37866301 DOI: 10.1016/j.theriogenology.2023.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 09/21/2023] [Accepted: 10/12/2023] [Indexed: 10/24/2023]
Abstract
In this study, we aimed to investigate the effect of the expression of miR-155-5p and its target genes on oocyte maturation. We analyzed the expression of miR-155-5p and its target genes in cumulus cells and oocytes using quantitative real-time reverse-transcription polymerase chain reaction. Using carboxyfluorescein, porcine cumulus cells were transfected with mimics and inhibitors of ssc-miR-155-5p to induce in vitro maturation, and subsequently, cumulus expansion, oocyte maturation, and cleavage rate were measured. We found that miR-155-5p expression in cumulus cells at the metaphase II stage was significantly higher than that at the germinal vesicle (GV) stage, whereas Cx43 expression was significantly lower than that at the GV stage (P < 0.05). Compared with those in the negative control group, the cumulus diffusion area of cumulus oocyte complexes; oocyte maturation rate; cleavage rate; HAS2, PTGS2, CD44, PTX3, and TNFAIP6 expression in cumulus cells; and GDF9, BMP15, CyclinB1, and CDK1 expression in oocytes were significantly increased in the miR-155-5p mimics group (P < 0.05), whereas the mRNA and protein expression of CX43 were significantly decreased (P < 0.05). Compared with that in the negative control group, the protein expression of CyclinB1 and p-CDK1 (Thr14, Tyr15) in the miR-155-5p mimics group was significantly increased (P < 0.05). These results suggest that miR-155-5p regulates maturation promoting factor activity by targeting Cx43, which improves the in vitro maturation and cleavage rate of porcine oocytes.
Collapse
Affiliation(s)
- Mimi Cheng
- Yanbian University, Jilin, Yanji, 133000, China
| | - Xuan Chen
- Yanbian University, Jilin, Yanji, 133000, China
| | - Mingzi Han
- Animal Disease Prevention and Control Center of Yanbian Korean Autonomous Prefecture, Jilin, Yanji, 133000, China
| | - Xiaotong Luo
- Institute of Animal Biotechnology, Jilin Academy of Agricultural Sciences, Jilin, Gongzhuling, 136100, China
| | - Yongsheng Yu
- Institute of Animal Biotechnology, Jilin Academy of Agricultural Sciences, Jilin, Gongzhuling, 136100, China
| | - Yanqiu Lv
- Yanbian University, Jilin, Yanji, 133000, China
| | - Yue Han
- Yanbian University, Jilin, Yanji, 133000, China
| | - Lipeng Cao
- Yanbian University, Jilin, Yanji, 133000, China
| | | | | | - Yi Jin
- Yanbian University, Jilin, Yanji, 133000, China.
| |
Collapse
|
13
|
Egbert JR, Silbern I, Uliasz TF, Lowther KM, Yee SP, Urlaub H, Jaffe LA. Phosphatases modified by LH signaling in ovarian follicles: testing their role in regulating the NPR2 guanylyl cyclase†. Biol Reprod 2024; 110:102-115. [PMID: 37774352 PMCID: PMC10790345 DOI: 10.1093/biolre/ioad130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2023] Open
Abstract
In response to luteinizing hormone (LH), multiple proteins in rat and mouse granulosa cells are rapidly dephosphorylated, but the responsible phosphatases remain to be identified. Because the phosphorylation state of phosphatases can regulate their interaction with substrates, we searched for phosphatases that might function in LH signaling by using quantitative mass spectrometry. We identified all proteins in rat ovarian follicles whose phosphorylation state changed detectably in response to a 30-min exposure to LH, and within this list, identified protein phosphatases or phosphatase regulatory subunits that showed changes in phosphorylation. Phosphatases in the phosphoprotein phosphatase (PPP) family were of particular interest because of their requirement for dephosphorylating the natriuretic peptide receptor 2 (NPR2) guanylyl cyclase in the granulosa cells, which triggers oocyte meiotic resumption. Among the PPP family regulatory subunits, PPP1R12A and PPP2R5D showed the largest increases in phosphorylation, with 4-10 fold increases in signal intensity on several sites. Although follicles from mice in which these phosphorylations were prevented by serine-to-alanine mutations in either Ppp1r12a or Ppp2r5d showed normal LH-induced NPR2 dephosphorylation, these regulatory subunits and others could act redundantly to dephosphorylate NPR2. Our identification of phosphatases and other proteins whose phosphorylation state is rapidly modified by LH provides clues about multiple signaling pathways in ovarian follicles.
Collapse
Affiliation(s)
- Jeremy R Egbert
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT, USA
| | - Ivan Silbern
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, Goettingen, Germany
- Institute of Clinical Chemistry, University Medical Center Goettingen, Goettingen, Germany
| | - Tracy F Uliasz
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT, USA
| | - Katie M Lowther
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT, USA
- Center for Mouse Genome Modification, University of Connecticut Health Center, Farmington CT, USA
| | - Siu-Pok Yee
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT, USA
- Center for Mouse Genome Modification, University of Connecticut Health Center, Farmington CT, USA
| | - Henning Urlaub
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, Goettingen, Germany
- Institute of Clinical Chemistry, University Medical Center Goettingen, Goettingen, Germany
- Cluster of Excellence “Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells” (MBExC), University of Göttingen, Göttingen, Germany
| | - Laurinda A Jaffe
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT, USA
| |
Collapse
|
14
|
Ezoe K, Takahashi T, Miki T, Kato K. Developmental perturbation in human embryos: Clinical and biological significance learned from time-lapse images. Reprod Med Biol 2024; 23:e12593. [PMID: 38983691 PMCID: PMC11232294 DOI: 10.1002/rmb2.12593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 05/30/2024] [Accepted: 06/07/2024] [Indexed: 07/11/2024] Open
Abstract
Background Time-lapse technology (TLT) has gained widespread adoption worldwide. In addition to facilitating the undisturbed culture of embryos, TLT offers the unique capability of continuously monitoring embryos to detect spatiotemporal changes. Although these observed phenomena play a role in optimal embryo selection/deselection, the clinical advantages of introducing TLT remain unclear. However, manual annotation of embryo perturbation could facilitate a comprehensive assessment of developmental competence. This process requires a thorough understanding of embryo observation and the biological significance associated with developmental dogma and variation. This review elucidates the typical behavior and variation of each phenomenon, exploring their clinical significance and research perspectives. Methods The MEDLINE database was searched using PubMed for peer-reviewed English-language original articles concerning human embryo development. Main findings TLT allows the observation of consecutive changes in embryo morphology, serving as potential biomarkers for embryo assessment. In assisted reproductive technology laboratories, several phenomena have not revealed their mechanism, posing difficulties such as fertilization deficiency and morula arrest. Conclusion A profound understanding of the biological mechanisms and significance of each phenomenon is crucial. Further collaborative efforts between the clinical and molecular fields following translational studies are required to advance embryonic outcomes and assessment.
Collapse
|
15
|
Das M, Son WY. In vitro maturation (IVM) of human immature oocytes: is it still relevant? Reprod Biol Endocrinol 2023; 21:110. [PMID: 37993914 PMCID: PMC10664544 DOI: 10.1186/s12958-023-01162-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 11/16/2023] [Indexed: 11/24/2023] Open
Abstract
In vitro maturation (IVM) of human immature oocytes has been shown to be a viable option for patients at risk of ovarian hyperstimulation syndrome (OHSS), those seeking urgent fertility preservation and in circumstances where controlled ovarian stimulation is not feasible. Moreover, IVM techniques can be combined with ovarian tissue cryobanking to increase the chances of conception in cancer survivors. The clinical applications of IVM in the field of reproductive medicine are rapidly expanding and the technique is now classified as non-experimental. In contrast to conventional IVF (in vitro fertilization), IVM offers several advantages, such as reduced gonadotropin stimulation, minimal risk of ovarian hyperstimulation syndrome (OHSS), reduced treatment times and lower costs. However, the technical expertise involved in performing IVM and its lower success rates compared to traditional IVF cycles, still pose significant challenges. Despite recent advances, such as innovative biphasic IVM systems, IVM is still an evolving technique and research is ongoing to refine protocols and identify techniques to improve its efficiency and effectiveness. A comprehensive understanding of the distinct mechanisms of oocyte maturation is crucial for obtaining more viable oocytes through in vitro methods, which will in turn lead to significantly improved success rates. In this review, the present state of human IVM programs and future research directions will be discussed, aiming to promote a better understanding of IVM and identify potential strategies to improve the overall efficiency and success rates of IVM programs, which will in turn lead to better clinical outcomes.
Collapse
Affiliation(s)
- Mausumi Das
- Department of Reproductive Medicine, Queen Charlotte and Hammersmith Hospitals, Imperial College Healthcare NHS Trust, London, UK
- Chelsea and Westminster Hospital NHS Foundation Trust, London, UK
| | | |
Collapse
|
16
|
Amargant F, Zhou LT, Yuan Y, Nahar A, Krisher RL, Spate LD, Roberts RM, Prather RS, Rowell EE, Laronda MM, Duncan FE. FGF2, LIF, and IGF1 (FLI) supplementation during human in vitro maturation enhances markers of gamete competence. Hum Reprod 2023; 38:1938-1951. [PMID: 37608600 DOI: 10.1093/humrep/dead162] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 07/05/2023] [Indexed: 08/24/2023] Open
Abstract
STUDY QUESTION Does a chemically defined maturation medium supplemented with FGF2, LIF, and IGF1 (FLI) improve in vitro maturation (IVM) of cumulus-oocyte complexes (COCs) obtained from children, adolescents, and young adults undergoing ovarian tissue cryopreservation (OTC)? SUMMARY ANSWER Although FLI supplementation did not increase the incidence of oocyte meiotic maturation during human IVM, it significantly improved quality outcomes, including increased cumulus cell expansion and mitogen-activated protein kinase (MAPK) expression as well as enhanced transzonal projection retraction. WHAT IS KNOWN ALREADY During OTC, COCs, and denuded oocytes from small antral follicles are released into the processing media. Recovery and IVM of these COCs is emerging as a complementary technique to maximize the fertility preservation potential of the tissue. However, the success of IVM is low, especially in the pediatric population. Supplementation of IVM medium with FLI quadruples the efficiency of pig production through improved oocyte maturation, but whether a similar benefit occurs in humans has not been investigated. STUDY DESIGN, SIZE, DURATION This study enrolled 75 participants between January 2018 and December 2021 undergoing clinical fertility preservation through the Fertility & Hormone Preservation & Restoration Program at the Ann & Robert H. Lurie Children's Hospital of Chicago. Participants donated OTC media, accumulated during tissue processing, for research. PARTICIPANTS/MATERIALS, SETTING, METHODS Participants who underwent OTC and include a pediatric population that encompassed children, adolescents, and young adults ≤22 years old. All participant COCs and denuded oocytes were recovered from media following ovarian tissue processing. IVM was then performed in either a standard medium (oocyte maturation medium) or one supplemented with FLI (FGF2; 40 ng/ml, LIF; 20 ng/ml, and IGF1; 20 ng/ml). IVM outcomes included meiotic progression, cumulus cell expansion, transzonal projection retraction, and detection of MAPK protein expression. MAIN RESULTS AND THE ROLE OF CHANCE The median age of participants was 6.3 years, with 65% of them classified as prepubertal by Tanner staging. Approximately 60% of participants had been exposed to chemotherapy and/or radiation prior to OTC. On average 4.7 ± 1 COCs and/or denuded oocytes per participant were recovered from the OTC media. COCs (N = 41) and denuded oocytes (N = 29) were used for IVM (42 h) in a standard or FLI-supplemented maturation medium. The incidence of meiotic maturation was similar between cohorts (COCs: 25.0% vs 28.6% metaphase II arrested eggs in Control vs FLI; denuded oocytes: 0% vs 5.3% in Control vs FLI). However, cumulus cell expansion was 1.9-fold greater in COCs matured in FLI-containing medium relative to Controls and transzonal projection retraction was more pronounced (2.45 ± 0.50 vs 1.16 ± 0.78 projections in Control vs FLIat 16 h). Additionally, MAPK expression was significantly higher in cumulus cells obtained from COCs matured in FLI medium for 16-18 h (chemiluminescence corrected area 621,678 vs 2,019,575 a.u., P = 0.03). LIMITATIONS, REASONS FOR CAUTION Our samples are from human participants who exhibited heterogeneity with respect to age, diagnosis, and previous treatment history. Future studies with larger sample sizes, including adult participants, are warranted to determine the mechanism by which FLI induces MAPK expression and activation. Moreover, studies that evaluate the developmental competence of eggs derived from FLI treatment, including assessment of embryos as outcome measures, will be required prior to clinical translation. WIDER IMPLICATIONS OF THE FINDINGS FLI supplementation may have a conserved beneficial effect on IVM for children, adolescents, and young adults spanning the agricultural setting to clinical fertility preservation. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by Department of Obstetrics and Gynecology startup funds (F.E.D.), Department of Surgery Faculty Practice Plan Grant and the Fertility & Hormone Preservation & Restoration Program at the Ann & Robert H. Lurie Children's Hospital of Chicago (M.M.L. and E.E.R.). M.M.L. is a Gesualdo Foundation Research Scholar. Y.Y.'s research is supported by the internal research funds provided by Colorado Center of Reproductive Medicine. Y.Y., L.D.S., R.M.R., and R.S.P. have a patent pending for FLI. The remaining authors have no conflict of interest. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Farners Amargant
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Luhan T Zhou
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Ye Yuan
- Colorado Center for Reproductive Medicine, Lone Tree, CO, USA
| | - Asrafun Nahar
- Colorado Center for Reproductive Medicine, Lone Tree, CO, USA
| | | | - Lee D Spate
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - R Michael Roberts
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - Randall S Prather
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - Erin E Rowell
- Division of Pediatric Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Monica M Laronda
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Francesca E Duncan
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
17
|
Telfer EE, Grosbois J, Odey YL, Rosario R, Anderson RA. Making a good egg: human oocyte health, aging, and in vitro development. Physiol Rev 2023; 103:2623-2677. [PMID: 37171807 PMCID: PMC10625843 DOI: 10.1152/physrev.00032.2022] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 05/03/2023] [Accepted: 05/06/2023] [Indexed: 05/13/2023] Open
Abstract
Mammalian eggs (oocytes) are formed during fetal life and establish associations with somatic cells to form primordial follicles that create a store of germ cells (the primordial pool). The size of this pool is influenced by key events during the formation of germ cells and by factors that influence the subsequent activation of follicle growth. These regulatory pathways must ensure that the reserve of oocytes within primordial follicles in humans lasts for up to 50 years, yet only approximately 0.1% will ever be ovulated with the rest undergoing degeneration. This review outlines the mechanisms and regulatory pathways that govern the processes of oocyte and follicle formation and later growth, within the ovarian stroma, through to ovulation with particular reference to human oocytes/follicles. In addition, the effects of aging on female reproductive capacity through changes in oocyte number and quality are emphasized, with both the cellular mechanisms and clinical implications discussed. Finally, the details of current developments in culture systems that support all stages of follicle growth to generate mature oocytes in vitro and emerging prospects for making new oocytes from stem cells are outlined.
Collapse
Affiliation(s)
- Evelyn E Telfer
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Johanne Grosbois
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Yvonne L Odey
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Roseanne Rosario
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Richard A Anderson
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
18
|
Owen CM, Jaffe LA. Luteinizing hormone stimulates ingression of mural granulosa cells within the mouse preovulatory follicle. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.21.537855. [PMID: 37131774 PMCID: PMC10153244 DOI: 10.1101/2023.04.21.537855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Luteinizing hormone (LH) induces ovulation by acting on its receptors in the mural granulosa cells that surround a mammalian oocyte in an ovarian follicle. However, much remains unknown about how activation of the LH receptor modifies the structure of the follicle such that the oocyte is released and the follicle remnants are transformed into the corpus luteum. The present study shows that the preovulatory surge of LH stimulates LH receptor-expressing granulosa cells, initially located almost entirely in the outer layers of the mural granulosa, to rapidly extend inwards, intercalating between other cells. The cellular ingression begins within 30 minutes of the peak of the LH surge, and the proportion of LH receptor-expressing cell bodies in the inner half of the mural granulosa layer increases until the time of ovulation, which occurs at about 10 hours after the LH peak. During this time, many of the initially flask-shaped cells appear to detach from the basal lamina, acquiring a rounder shape with multiple filipodia. Starting at about 4 hours after the LH peak, the mural granulosa layer at the apical surface of the follicle where ovulation will occur begins to thin, and the basolateral surface develops invaginations and constrictions. Our findings raise the question of whether LH stimulation of granulosa cell ingression may contribute to these changes in the follicular structure that enable ovulation.
Collapse
Affiliation(s)
- Corie M. Owen
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT 06030 USA
| | - Laurinda A. Jaffe
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT 06030 USA
| |
Collapse
|
19
|
Egbert JR, Silbern I, Uliasz TF, Lowther KM, Yee SP, Urlaub H, Jaffe LA. Phosphatases modified by LH signaling in ovarian follicles: testing their role in regulating the NPR2 guanylyl cyclase. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.12.544636. [PMID: 37333193 PMCID: PMC10274890 DOI: 10.1101/2023.06.12.544636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
In response to luteinizing hormone, multiple proteins in rat and mouse granulosa cells are rapidly dephosphorylated, but the responsible phosphatases remain to be identified. Because the phosphorylation state of phosphatases can regulate their interaction with substrates, we searched for phosphatases that might function in LH signaling by using quantitative mass spectrometry. We identified all proteins in rat ovarian follicles whose phosphorylation state changed detectably in response to a 30-minute exposure to LH, and within this list, identified protein phosphatases or phosphatase regulatory subunits that showed changes in phosphorylation. Phosphatases in the PPP family were of particular interest because of their requirement for dephosphorylating the natriuretic peptide receptor 2 (NPR2) guanylyl cyclase in the granulosa cells, which triggers oocyte meiotic resumption. Among the PPP family regulatory subunits, PPP1R12A and PPP2R5D showed the largest increases in phosphorylation, with 4-10 fold increases in signal intensity on several sites. Although follicles from mice in which these phosphorylations were prevented by serine-to-alanine mutations in either Ppp1r12a or Ppp2r5d showed normal LH-induced NPR2 dephosphorylation, these regulatory subunits and others could act redundantly to dephosphorylate NPR2. Our identification of phosphatases and other proteins whose phosphorylation state is rapidly modified by LH provides clues about multiple signaling pathways in ovarian follicles. Summary sentence Quantitative mass spectrometric analysis of phosphatases whose phosphorylation state is rapidly modified by luteinizing hormone provides clues about how LH signaling dephosphorylates NPR2 as well as a resource for future studies.
Collapse
Affiliation(s)
- Jeremy R. Egbert
- Department of Cell Biology, University of Connecticut Health Center, Farmington CT 06030 USA
| | - Ivan Silbern
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, 37077 Goettingen, Germany
- Institute of Clinical Chemistry, University Medical Center Goettingen, 37075 Goettingen, Germany
| | - Tracy F. Uliasz
- Department of Cell Biology, University of Connecticut Health Center, Farmington CT 06030 USA
| | - Katie M. Lowther
- Department of Cell Biology, University of Connecticut Health Center, Farmington CT 06030 USA
- Center for Mouse Genome Modification, University of Connecticut Health Center, Farmington CT 06030 USA
| | - Siu-Pok Yee
- Department of Cell Biology, University of Connecticut Health Center, Farmington CT 06030 USA
- Center for Mouse Genome Modification, University of Connecticut Health Center, Farmington CT 06030 USA
| | - Henning Urlaub
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, 37077 Goettingen, Germany
- Institute of Clinical Chemistry, University Medical Center Goettingen, 37075 Goettingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, 37075 Göttingen, Germany
| | - Laurinda A. Jaffe
- Department of Cell Biology, University of Connecticut Health Center, Farmington CT 06030 USA
| |
Collapse
|
20
|
de la Fuente A, Scoggin C, Bradecamp E, Martin-Pelaez S, van Heule M, Troedsson M, Daels P, Meyers S, Dini P. Transcriptome Signature of Immature and In Vitro-Matured Equine Cumulus-Oocytes Complex. Int J Mol Sci 2023; 24:13718. [PMID: 37762020 PMCID: PMC10531358 DOI: 10.3390/ijms241813718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 08/31/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Maturation is a critical step in the development of an oocyte, and it is during this time that the oocyte advances to metaphase II (MII) of the meiotic cycle and acquires developmental competence to be fertilized and become an embryo. However, in vitro maturation (IVM) remains one of the limiting steps in the in vitro production of embryos (IVP), with a variable percentage of oocytes reaching the MII stage and unpredictable levels of developmental competence. Understanding the dynamics of oocyte maturation is essential for the optimization of IVM culture conditions and subsequent IVP outcomes. Thus, the aim of this study was to elucidate the transcriptome dynamics of oocyte maturation by comparing transcriptomic changes during in vitro maturation in both oocytes and their surrounding cumulus cells. Cumulus-oocyte complexes were obtained from antral follicles and divided into two groups: immature and in vitro-matured (MII). RNA was extracted separately from oocytes (OC) and cumulus cells (CC), followed by library preparation and RNA sequencing. A total of 13,918 gene transcripts were identified in OC, with 538 differentially expressed genes (DEG) between immature OC and in vitro-matured OC. In CC, 13,104 genes were expressed with 871 DEG. Gene ontology (GO) analysis showed an association between the DEGs and pathways relating to nuclear maturation in OC and GTPase activity, extracellular matrix organization, and collagen trimers in CC. Additionally, the follicle-stimulating hormone receptor gene (FSHR) and luteinizing hormone/choriogonadotropin receptor gene (LHCGR) showed differential expressions between CC-MII and immature CC samples. Overall, these results serve as a foundation to further investigate the biological pathways relevant to oocyte maturation in horses and pave the road to improve the IVP outcomes and the overall clinical management of equine assisted reproductive technologies (ART).
Collapse
Affiliation(s)
- Alejandro de la Fuente
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Charles Scoggin
- LeBlanc Reproduction Center, Rood and Riddle Equine Hospital, Lexington, KY 40511, USA
| | - Etta Bradecamp
- LeBlanc Reproduction Center, Rood and Riddle Equine Hospital, Lexington, KY 40511, USA
| | - Soledad Martin-Pelaez
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Machteld van Heule
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
- Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, University of Ghent, 9820 Merelbeke, Belgium
| | - Mats Troedsson
- Gluck Equine Research Center, University of Kentucky, Lexington, KY 40506, USA
| | - Peter Daels
- Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, University of Ghent, 9820 Merelbeke, Belgium
| | - Stuart Meyers
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Pouya Dini
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| |
Collapse
|
21
|
Kim HM, Kang MK, Seong SY, Jo JH, Kim MJ, Shin EK, Lee CG, Han SJ. Meiotic Cell Cycle Progression in Mouse Oocytes: Role of Cyclins. Int J Mol Sci 2023; 24:13659. [PMID: 37686466 PMCID: PMC10487953 DOI: 10.3390/ijms241713659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 08/31/2023] [Accepted: 09/01/2023] [Indexed: 09/10/2023] Open
Abstract
All eukaryotic cells, including oocytes, utilize an engine called cyclin-dependent kinase (Cdk) to drive the cell cycle. Cdks are activated by a co-factor called cyclin, which regulates their activity. The key Cdk-cyclin complex that regulates the oocyte cell cycle is known as Cdk1-cyclin B1. Recent studies have elucidated the roles of other cyclins, such as B2, B3, A2, and O, in oocyte cell cycle regulation. This review aims to discuss the recently discovered roles of various cyclins in mouse oocyte cell cycle regulation in accordance with the sequential progression of the cell cycle. In addition, this review addresses the translation and degradation of cyclins to modulate the activity of Cdks. Overall, the literature indicates that each cyclin performs unique and redundant functions at various stages of the cell cycle, while their expression and degradation are tightly regulated. Taken together, this review provides new insights into the regulatory role and function of cyclins in oocyte cell cycle progression.
Collapse
Affiliation(s)
- Hye Min Kim
- Department of Biological Science, Inje University, Gimhae 50834, Republic of Korea; (H.M.K.); (E.K.S.)
- Department of Research Center, Dongnam Institute of Radiological and Medical Sciences, Busan 46033, Republic of Korea; (M.K.K.); (C.G.L.)
| | - Min Kook Kang
- Department of Research Center, Dongnam Institute of Radiological and Medical Sciences, Busan 46033, Republic of Korea; (M.K.K.); (C.G.L.)
| | - Se Yoon Seong
- Institute for Digital Antiaging Healthcare, Inje University, Gimhae 50834, Republic of Korea; (S.Y.S.); (J.H.J.); (M.J.K.)
| | - Jun Hyeon Jo
- Institute for Digital Antiaging Healthcare, Inje University, Gimhae 50834, Republic of Korea; (S.Y.S.); (J.H.J.); (M.J.K.)
| | - Min Ju Kim
- Institute for Digital Antiaging Healthcare, Inje University, Gimhae 50834, Republic of Korea; (S.Y.S.); (J.H.J.); (M.J.K.)
| | - Eun Kyeong Shin
- Department of Biological Science, Inje University, Gimhae 50834, Republic of Korea; (H.M.K.); (E.K.S.)
- Department of Research Center, Dongnam Institute of Radiological and Medical Sciences, Busan 46033, Republic of Korea; (M.K.K.); (C.G.L.)
| | - Chang Geun Lee
- Department of Research Center, Dongnam Institute of Radiological and Medical Sciences, Busan 46033, Republic of Korea; (M.K.K.); (C.G.L.)
| | - Seung Jin Han
- Department of Biological Science, Inje University, Gimhae 50834, Republic of Korea; (H.M.K.); (E.K.S.)
- Institute for Digital Antiaging Healthcare, Inje University, Gimhae 50834, Republic of Korea; (S.Y.S.); (J.H.J.); (M.J.K.)
- Department of Medical Biotechnology, Inje University, Gimhae 50834, Republic of Korea
- Institute of Basic Science, Inje University, Gimhae 50834, Republic of Korea
| |
Collapse
|
22
|
Buratini J, Dellaqua TT, de Lima PF, Renzini MM, Canto MD, Price CA. Oocyte secreted factors control genes regulating FSH signaling and the maturation cascade in cumulus cells: the oocyte is not in a hurry. J Assist Reprod Genet 2023; 40:1961-1971. [PMID: 37204638 PMCID: PMC10371970 DOI: 10.1007/s10815-023-02822-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 04/29/2023] [Indexed: 05/20/2023] Open
Abstract
PURPOSE To assess the effects of the oocyte on mRNA abundance of FSHR, AMH and major genes of the maturation cascade (AREG, EREG, ADAM17, EGFR, PTGS2, TNFAIP6, PTX3, and HAS2) in bovine cumulus cells. METHODS (1) Intact cumulus-oocyte complexes, (2) microsurgically oocytectomized cumulus-oolema complexes (OOX), and (3) OOX + denuded oocytes (OOX+DO) were subjected to in vitro maturation (IVM) stimulated with FSH for 22 h or with AREG for 4 and 22 h. After IVM, cumulus cells were separated and relative mRNA abundance was measured by RT-qPCR. RESULTS After 22 h of FSH-stimulated IVM, oocytectomy increased FSHR mRNA levels (p=0.005) while decreasing those of AMH (p=0.0004). In parallel, oocytectomy increased mRNA abundance of AREG, EREG, ADAM17, PTGS2, TNFAIP6, and PTX3, while decreasing that of HAS2 (p<0.02). All these effects were abrogated in OOX+DO. Oocytectomy also reduced EGFR mRNA levels (p=0.009), which was not reverted in OOX+DO. The stimulatory effect of oocytectomy on AREG mRNA abundance (p=0.01) and its neutralization in OOX+DO was again observed after 4 h of AREG-stimulated IVM. After 22 h of AREG-stimulated IVM, oocytectomy and addition of DOs to OOX caused the same effects on gene expression observed after 22 h of FSH-stimulated IVM, except for ADAM17 (p<0.025). CONCLUSION These findings suggest that oocyte-secreted factors inhibit FSH signaling and the expression of major genes of the maturation cascade in cumulus cells. These may be important actions of the oocyte favoring its communication with cumulus cells and preventing premature activation of the maturation cascade.
Collapse
Affiliation(s)
- Jose Buratini
- Biogenesi, Reproductive Medicine Centre, Monza, Italy
- Clinica EUGIN, Milan, Italy
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP Brazil
| | - Thaisy Tino Dellaqua
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP Brazil
| | - Paula Fernanda de Lima
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University, Botucatu, SP Brazil
| | | | | | - Christopher A. Price
- Centre de Recherche en Reproduction Animale, Faculté de Médecine Vétérinaire, Université de Montréal, St-Hyacinthe, Canada
| |
Collapse
|
23
|
Camp OG, Bembenek JN, Goud PT, Awonuga AO, Abu-Soud HM. The Implications of Insufficient Zinc on the Generation of Oxidative Stress Leading to Decreased Oocyte Quality. Reprod Sci 2023; 30:2069-2078. [PMID: 36920672 PMCID: PMC11047769 DOI: 10.1007/s43032-023-01212-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 03/07/2023] [Indexed: 03/16/2023]
Abstract
Zinc is a transition metal that displays wide physiological implications ranging from participation in hundreds of enzymes and proteins to normal growth and development. In the reproductive tract of both sexes, zinc maintains a functional role in spermatogenesis, ovulation, fertilization, normal pregnancy, fetal development, and parturition. In this work, we review evidence to date regarding the importance of zinc in oocyte maturation and development, with emphasis on the role of key zinc-binding proteins, as well as examine the effects of zinc and reactive oxygen species (ROS) on oocyte quality and female fertility. We summarize our current knowledge about the participation of zinc in the developing oocyte bound to zinc finger proteins as well as loosely bound zinc ion in the intracellular and extracellular environments. These include aspects related to (1) the impact of zinc deficiency and overwhelming production of ROS under inflammatory conditions on the offset of the physiological antioxidant machinery disturbing biomolecules, proteins, and cellular processes, and their role in contributing to further oxidative stress; (2) the role of ROS in modulating damage to proteins containing zinc, such as zinc finger proteins and nitric oxide synthases (NOS), and expelling the zinc resulting in loss of protein function; and (3) clarify the different role of oxidative stress and zinc deficiency in the pathophysiology of infertility diseases with special emphasis on endometriosis-associated infertility.
Collapse
Affiliation(s)
- Olivia G Camp
- Departments of Obstetrics and Gynecology and Biochemistry and Molecular Biology, The C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, 275 E. Hancock, Detroit, MI, 48201, USA
| | - Joshua N Bembenek
- Departments of Obstetrics and Gynecology and Biochemistry and Molecular Biology, The C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, 275 E. Hancock, Detroit, MI, 48201, USA
| | - Pravin T Goud
- Division of Reproductive Endocrinology and Infertility & California IVF Fertility Center, Department of Obstetrics and Gynecology, University of California Davis, Sacramento, CA, 95833, USA
- Laurel Fertility Care, San Francisco, CA, 94109, USA
- California Northstate University Medical College, Elk Grove, CA, 95757, USA
| | - Awoniyi O Awonuga
- Departments of Obstetrics and Gynecology and Biochemistry and Molecular Biology, The C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, 275 E. Hancock, Detroit, MI, 48201, USA
| | - Husam M Abu-Soud
- Departments of Obstetrics and Gynecology and Biochemistry and Molecular Biology, The C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, 275 E. Hancock, Detroit, MI, 48201, USA.
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, 48201, USA.
- Department of Microbiology, Immunology and Biochemistry, Wayne State University School of Medicine, Detroit, MI, 48201, USA.
| |
Collapse
|
24
|
Li Y, Chang HM, Sung YW, Zhu H, Leung PCK, Sun YP. Betacellulin regulates gap junction intercellular communication by inducing the phosphorylation of connexin 43 in human granulosa-lutein cells. J Ovarian Res 2023; 16:103. [PMID: 37231448 DOI: 10.1186/s13048-023-01185-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 05/06/2023] [Indexed: 05/27/2023] Open
Abstract
BACKGROUND The gap junction protein, connexin 43 (Cx43) is highly expressed in human granulosa-lutein (hGL) cells. The phosphorylation of certain amino acid residues in the Cx43 protein has been shown to be related to a decline in gap junction intercellular communication (GJIC), which subsequently affects oocyte meiotic resumption. As a member of the epidermal growth factor (EGF) family, betacellulin (BTC) mediates luteinizing hormone (LH)-induced oocyte maturation and cumulus cell expansion in mammalian follicles. Whether BTC can regulate Cx43 phosphorylation, which further reduces Cx43-coupled GJIC activity in hGL cells remains to be determined. METHODS Immortalized human granulosa cells (SVOG cells) and primary human granulosa-lutein cells obtained from women undergoing in vitro fertilization in an academic research center were used as the study models. The expression levels of Cx43 and phosphorylated Cx43 were examined following cell incubation with BTC at different time points. Several kinase inhibitors (sotrastaurin, AG1478, and U0126) and small interfering RNAs targeting EGF receptor (EGFR) and receptor tyrosine-protein kinase 4 (ErbB4) were used to verify the specificity of the effects and to investigate the molecular mechanisms. Real-time-quantitative PCR and western blot analysis were used to detect the specific mRNA and protein levels, respectively. GJIC between SVOG cells were evaluated using a scrape loading and dye transfer assay. Results were analyzed by one-way analysis of variance. RESULTS The results showed that BTC induced the rapid phosphorylation of Cx43 at serine368 without altering the expression of Cx43 in primary and immortalized hGL cells. Additionally, using a dual inhibition approach (kinase inhibitors and siRNA-based expression knockdown), we demonstrated that this effect was mainly mediated by the EGFR but not the ErbB4 receptor. Furthermore, using a protein kinase C (PKC) kinase assay and a scrape-loading and dye transfer assay, we revealed that PKC signaling is the downstream signaling pathway that mediates the increase in Cx43 phosphorylation and subsequent decrease in GJIC activity in response to BTC treatment in hGL cells. CONCLUSIONS BTC promptly induced the phosphorylation of connexin 43 at Ser368, leading to decreased GJIC activity in hGL cells. The BTC-induced cellular activities were most likely driven by the EGFR-mediated PKC-dependent signaling pathway. Our findings shed light on the detailed molecular mechanisms by which BTC regulates the process of oocyte meiotic resumption.
Collapse
Affiliation(s)
- Yuxi Li
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, 40, Daxue Road, Zhengzhou, 450052, Henan, China
- Department of Obstetrics and Gynecology, BC Children's Hospital Research Institute, University of British Columbia, Room 317, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hsun-Ming Chang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung, Taiwan
| | - Yu-Wen Sung
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung, Taiwan
| | - Hua Zhu
- Department of Obstetrics and Gynecology, BC Children's Hospital Research Institute, University of British Columbia, Room 317, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada
| | - Peter C K Leung
- Department of Obstetrics and Gynecology, BC Children's Hospital Research Institute, University of British Columbia, Room 317, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada.
| | - Ying-Pu Sun
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, 40, Daxue Road, Zhengzhou, 450052, Henan, China.
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
25
|
Xie J, Xu X, Liu S. Intercellular communication in the cumulus-oocyte complex during folliculogenesis: A review. Front Cell Dev Biol 2023; 11:1087612. [PMID: 36743407 PMCID: PMC9893509 DOI: 10.3389/fcell.2023.1087612] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 01/10/2023] [Indexed: 01/20/2023] Open
Abstract
During folliculogenesis, the oocyte and surrounding cumulus cells form an ensemble called the cumulus-oocyte complex (COC). Due to their interdependence, research on the COC has been a hot issue in the past few decades. A growing body of literature has revealed that intercellular communication is critical in determining oocyte quality and ovulation. This review provides an update on the current knowledge of COC intercellular communication, morphology, and functions. Transzonal projections (TZPs) and gap junctions are the most described structures of the COC. They provide basic metabolic and nutrient support, and abundant molecules for signaling pathways and regulations. Oocyte-secreted factors (OSFs) such as growth differentiation factor 9 and bone morphogenetic protein 15 have been linked with follicular homeostasis, suggesting that the communications are bidirectional. Using advanced techniques, new evidence has highlighted the existence of other structures that participate in intercellular communication. Extracellular vesicles can carry transcripts and signaling molecules. Microvilli on the oocyte can induce the formation of TZPs and secrete OSFs. Cell membrane fusion between the oocyte and cumulus cells can lead to sharing of cytoplasm, in a way making the COC a true whole. These findings give us new insights into related reproductive diseases like polycystic ovary syndrome and primary ovarian insufficiency and how to improve the outcomes of assisted reproduction.
Collapse
Affiliation(s)
- Jun Xie
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiao Xu
- Department of Obstetrics and Gynecology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Suying Liu
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, China,*Correspondence: Suying Liu,
| |
Collapse
|
26
|
Clarke HJ. Transzonal projections: Essential structures mediating intercellular communication in the mammalian ovarian follicle. Mol Reprod Dev 2022; 89:509-525. [PMID: 36112806 DOI: 10.1002/mrd.23645] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/22/2022] [Accepted: 08/31/2022] [Indexed: 12/25/2022]
Abstract
The development of germ cells relies on contact and communication with neighboring somatic cells that provide metabolic support and regulatory signals. In females, contact is achieved through thin cytoplasmic processes that project from follicle cells surrounding the oocyte, extend through an extracellular matrix (ECM) that lies between them, and reach its surface. In mammals, the ECM is termed the zona pellucida and the follicular cell processes are termed transzonal projections (TZPs). TZPs become detectable when the zona pellucida is laid down during early folliculogenesis and subsequently increase in number as oocyte growth progresses. They then rapidly disappear at the time of ovulation, permanently breaking germ-soma contact. Here we review the life cycle and functions of the TZPs. We begin with an overview of the morphology and cytoskeletal structure of TZPs, in the context of actin- and tubulin-based cytoplasmic processes in other cell types. Next, we review the roles played by TZPs in mediating progression through successive stages of oocyte development. We then discuss two mechanisms that may generate TZPs-stretching at pre-existing points of granulosa cell-oocyte contact and elaboration of new processes that push through the zona pellucida-as well as gene products implicated in their formation or function. Finally, we describe the signaling pathways that cause TZPs to be retracted in response to signals that also trigger meiotic maturation and ovulation of the oocyte. The principles and mechanisms that govern TZP behavior may be relevant to understanding communication between physically separated cells in other physiological contexts.
Collapse
Affiliation(s)
- Hugh J Clarke
- Program in Child Health and Human Development, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.,Department of Obstetrics and Gynecology, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
27
|
Dellaqua TT, Vígaro RA, Janini LCZ, Dal Canto M, Renzini MM, Lodde V, Luciano AM, Buratini J. Neuregulin 1 (NRG1) modulates oocyte nuclear maturation during IVM and improves post-IVF embryo development. Theriogenology 2022; 195:209-216. [DOI: 10.1016/j.theriogenology.2022.10.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/31/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022]
|
28
|
Das D, Arur S. Regulation of oocyte maturation: Role of conserved ERK signaling. Mol Reprod Dev 2022; 89:353-374. [PMID: 35908193 PMCID: PMC9492652 DOI: 10.1002/mrd.23637] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 07/12/2022] [Accepted: 07/18/2022] [Indexed: 11/11/2022]
Abstract
During oogenesis, oocytes arrest at meiotic prophase I to acquire competencies for resuming meiosis, fertilization, and early embryonic development. Following this arrested period, oocytes resume meiosis in response to species-specific hormones, a process known as oocyte maturation, that precedes ovulation and fertilization. Involvement of endocrine and autocrine/paracrine factors and signaling events during maintenance of prophase I arrest, and resumption of meiosis is an area of active research. Studies in vertebrate and invertebrate model organisms have delineated the molecular determinants and signaling pathways that regulate oocyte maturation. Cell cycle regulators, such as cyclin-dependent kinase (CDK1), polo-like kinase (PLK1), Wee1/Myt1 kinase, and the phosphatase CDC25 play conserved roles during meiotic resumption. Extracellular signal-regulated kinase (ERK), on the other hand, while activated during oocyte maturation in all species, regulates both species-specific, as well as conserved events among different organisms. In this review, we synthesize the general signaling mechanisms and focus on conserved and distinct functions of ERK signaling pathway during oocyte maturation in mammals, non-mammalian vertebrates, and invertebrates such as Drosophila and Caenorhabditis elegans.
Collapse
Affiliation(s)
- Debabrata Das
- Department of Genetics, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Swathi Arur
- Department of Genetics, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
29
|
Godin P, Tsoi MF, Morin M, Gévry N, Boerboom D. The granulosa cell response to luteinizing hormone is partly mediated by YAP1-dependent induction of amphiregulin. Cell Commun Signal 2022; 20:72. [PMID: 35619099 PMCID: PMC9137176 DOI: 10.1186/s12964-022-00843-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 02/08/2022] [Indexed: 12/23/2022] Open
Abstract
Background The LH surge is a pivotal event that triggers multiple key ovarian processes including oocyte maturation, cumulus expansion, follicular wall rupture and luteinization of mural granulosa and theca cells. Recently, LH-dependent activation of the Hippo signaling pathway has been shown to be required for the differentiation of granulosa cells into luteal cells. Still, the precise interactions between Hippo and LH signaling in murine granulosa cells remain to be elucidated. Methods To detect the expression of effectors of the Hippo pathway, western blot, immunohistochemical and RT-qPCR analyses were performed on granulosa cells treated with LH in vitro or isolated from immature mice treated with eCG and hCG. Cultured granulosa cells were pretreated with pharmacologic inhibitors to identify the signaling pathways involved in Hippo regulation by LH. To study the roles of Yap1 and Taz in the regulation of the LH signaling cascade, RT-qPCR and microarray analyses were done on granulosa cells from Yap1f/f;Tazf/f mice treated with an adenovirus to drive cre expression. RT-qPCR was performed to evaluate YAP1 binding to the Areg promoter following chromatin immunoprecipitation of granulosa cells collected from mice prior to or 60 min following hCG treatment. Results Granulosa cells showed a transient increase in LATS1, YAP1 and TAZ phosphorylation levels in response to the ovulatory signal. This Hippo activation by LH was mediated by protein kinase A. Furthermore, Yap1 and Taz are required for the induction of several LH target genes such as Areg, Pgr and Ptgs2, and for the activation of the ERK1/2 pathway. Consistent with these results, there was a substantial overlap between genes that are upregulated by LH and those that are downregulated following loss of Yap1/Taz, highlighting a major role for Hippo in mediating LH actions in the ovulation process. Finally, we showed that there is a marked recruitment of YAP1 to the Areg promoter of granulosa cells in response to hCG stimulation. Conclusions Overall, these results indicate that Hippo collaborates with the cAMP/PKA and ERK1/2 pathways to participate in the precise regulation of the LH cascade, and that Areg, as a direct transcriptional target of YAP1, is involved in mediating its actions in the ovary. Video Abstract
Supplementary Information The online version contains supplementary material available at 10.1186/s12964-022-00843-1.
Collapse
Affiliation(s)
- Philippe Godin
- Centre de Recherche en Reproduction et Fertilité (CRRF), Université de Montréal, Saint-Hyacinthe, QC, Canada
| | - Mayra F Tsoi
- Centre de Recherche en Reproduction et Fertilité (CRRF), Université de Montréal, Saint-Hyacinthe, QC, Canada
| | - Martin Morin
- Department of Biology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Nicolas Gévry
- Department of Biology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Derek Boerboom
- Centre de Recherche en Reproduction et Fertilité (CRRF), Université de Montréal, Saint-Hyacinthe, QC, Canada.
| |
Collapse
|
30
|
Ozturk S. Molecular determinants of the meiotic arrests in mammalian oocytes at different stages of maturation. Cell Cycle 2022; 21:547-571. [PMID: 35072590 PMCID: PMC8942507 DOI: 10.1080/15384101.2022.2026704] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 12/21/2021] [Accepted: 01/03/2022] [Indexed: 01/26/2023] Open
Abstract
Mammalian oocytes undergo two rounds of developmental arrest during maturation: at the diplotene of the first meiotic prophase and metaphase of the second meiosis. These arrests are strictly regulated by follicular cells temporally producing the secondary messengers, cAMP and cGMP, and other factors to regulate maturation promoting factor (composed of cyclin B1 and cyclin-dependent kinase 1) levels in the oocytes. Out of these normally appearing developmental arrests, permanent arrests may occur in the oocytes at germinal vesicle (GV), metaphase I (MI), or metaphase II (MII) stage. This issue may arise from absence or altered expression of the oocyte-related genes playing key roles in nuclear and cytoplasmic maturation. Additionally, the assisted reproductive technology (ART) applications such as ovarian stimulation and in vitro culture conditions both of which harbor various types of chemical agents may contribute to forming the permanent arrests. In this review, the molecular determinants of developmental and permanent arrests occurring in the mammalian oocytes are comprehensively evaluated in the light of current knowledge. As number of permanently arrested oocytes at different stages is increasing in ART centers, potential approaches for inducing permanent arrests to obtain competent oocytes are discussed.
Collapse
Affiliation(s)
- Saffet Ozturk
- Department of Histology and Embryology, Akdeniz University School of Medicine, Antalya, Turkey
| |
Collapse
|
31
|
Strączyńska P, Papis K, Morawiec E, Czerwiński M, Gajewski Z, Olejek A, Bednarska-Czerwińska A. Signaling mechanisms and their regulation during in vivo or in vitro maturation of mammalian oocytes. Reprod Biol Endocrinol 2022; 20:37. [PMID: 35209923 PMCID: PMC8867761 DOI: 10.1186/s12958-022-00906-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 02/06/2022] [Indexed: 12/18/2022] Open
Abstract
In vitro fertilization (IVF) is currently one of the most effective methods of infertility treatment. An alternative to commonly used ovarian hyperstimulation can become extracorporeal maturation of oocytes (in vitro maturation; IVM). Fertilization and normal development of the embryo depends on the cytoplasmic, nuclear and genomic maturity of the oocyte. The microenvironment of the ovarian follicle and maternal signals, which mediate bidirectional communication between granulosa, cumulus and oocyte cells, influence the growth, maturation and acquisition of oocyte development capability. During oogenesis in mammals, the meiosis is inhibited in the oocyte at the prophase I of the meiotic division due to the high cAMP level. This level is maintained by the activity of C-type natriuretic peptide (CNP, NPPC) produced by granulosa cells. The CNP binds to the NPR2 receptor in cumulus cells and is responsible for the production of cyclic guanosine monophosphate (cGMP). The cGMP penetrating into the oocyte through gap junctions inhibits phosphodiesterase 3A (PDE3A), preventing cAMP hydrolysis responsible for low MPF activity. The LH surge during the reproductive cycle reduces the activity of the CNP/NPR2 complex, which results in a decrease in cGMP levels in cumulus cells and consequently in the oocyte. Reduced cGMP concentration unblocks the hydrolytic activity of PDE3A, which decreases cAMP level inside the oocyte. This leads to the activation of MPF and resumption of meiosis. The latest IVM methods called SPOM, NFSOM or CAPA IVM consist of two steps: prematuration and maturation itself. Taking into account the role of cAMP in inhibiting and then unblocking the maturation of oocytes, they have led to a significant progress in terms of the percentage of mature oocytes in vitro and the proportion of properly developed embryos in both animals and humans.
Collapse
Affiliation(s)
- Patrycja Strączyńska
- Department of Gynecology, Obstetrics and Oncological Gynecology in Bytom, Medical University of Silesia, Katowice, Poland
- Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
- Gyncentrum Fertility Clinic, Katowice, Poland
| | - Krzysztof Papis
- Center for Translational Medicine, Warsaw University of Life Sciences, Warsaw, Poland.
- nOvum Fertility Clinic, Warsaw, Poland.
| | - Emilia Morawiec
- Gyncentrum Fertility Clinic, Katowice, Poland
- Department of Microbiology, Faculty of Medicine in Zabrze, University of Technology in Katowice, Katowice, Poland
| | | | - Zdzisław Gajewski
- Center for Translational Medicine, Warsaw University of Life Sciences, Warsaw, Poland
| | - Anita Olejek
- Department of Gynecology, Obstetrics and Oncological Gynecology in Bytom, Medical University of Silesia, Katowice, Poland
| | | |
Collapse
|
32
|
Yang Q, Zhu L, Wang M, Huang B, Li Z, Hu J, Xi Q, Liu J, Jin L. Analysis of maturation dynamics and developmental competence of in vitro matured oocytes under time-lapse monitoring. Reprod Biol Endocrinol 2021; 19:183. [PMID: 34893069 PMCID: PMC8662918 DOI: 10.1186/s12958-021-00868-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/29/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND To improve the developmental competence of in vitro cultured oocytes, extensive literature focused on maturation rate improvement with different additives in culture medium, while studies investigating the maturation dynamics of oocytes during in vitro maturation (IVM) and the influencing factors on oocyte viability are scarce. METHODS The study involved a retrospective observation by time-lapse monitoring of the IVM process of 157 donated GV oocytes from 59 infertile couples receiving ICSI in 2019, in Tongji Hospital, Wuhan, China. The GV oocytes derived from controlled ovarian hyperstimulation (COH) cycles underwent rescue IVM (R-IVM), and the maturation dynamics, including GVBD time (GV-MI), time from GVBD to maturation (MI-MII), maturation time (GV-MII), and MII arrest duration (MII-ICSI), were recorded by time-lapse monitoring. The matured oocytes were inseminated at different MII arrest points and subsequent embryo developments were assessed. The effects of baseline clinical characteristics, oocyte diameters, and maturation dynamics on the developmental competence of the oocytes were also analyzed. RESULTS Totally, 157 GV oocytes were collected. GVBD happened in 111 oocytes, with a median GV-MI duration of 3.7 h. The median MI-MII duration was 15.6 h and the median GV-MII duration was 19.5 h. The maturation rate reached 56.7% at 24 h and 66.9% at 48 h, and the clinical factors, including patient age, FSH level, AMH level, ovarian stimulation protocol, and serum estradiol and progesterone levels on hCG trigger day, showed no effects on the 24-h maturation rate. The normal fertilization rate of oocytes resuming meiosis within 8 h and matured within 24 h was significantly higher than that of oocytes resuming meiosis after 8 h and matured after 24 h. Furthermore, among those oocytes matured within 24 h, the high-quality embryo formation rate of oocytes resuming meiosis within 4.5 h and matured within 19 h was significantly higher. All stated time was measured from the start point of IVM. Additionally, for oocytes from patients with serum progesterone levels less than 1 ng/ml on hCG trigger day, the high-quality embryo formation rate was significantly increased. CONCLUSION R-IVM technology could increase the available embryos for patients in routine COH cycles, but excessive culture beyond 24 h is not recommended. GV-MI duration of the oocyte, recorded by time-lapse system, and serum progesterone levels of patients on hCG trigger day can significantly affect the developmental potential of the IVM oocytes.
Collapse
Affiliation(s)
- Qiyu Yang
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jiefang Road, Wuhan, 430030, China
| | - Lixia Zhu
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jiefang Road, Wuhan, 430030, China
| | - Meng Wang
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jiefang Road, Wuhan, 430030, China
| | - Bo Huang
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jiefang Road, Wuhan, 430030, China
| | - Zhou Li
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jiefang Road, Wuhan, 430030, China
| | - Juan Hu
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jiefang Road, Wuhan, 430030, China
| | - Qingsong Xi
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jiefang Road, Wuhan, 430030, China.
| | - Jing Liu
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jiefang Road, Wuhan, 430030, China.
| | - Lei Jin
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jiefang Road, Wuhan, 430030, China.
| |
Collapse
|
33
|
Duan J, Chen H, Li Y, Xu D, Li X, Zhang Z, Cheng J, Yang L, Li Q. 17β-Estradiol Enhances Porcine Meiosis Resumption from Autophagy-Induced Gap Junction Intercellular Communications and Connexin 43 Phosphorylation via the MEK/ERK Signaling Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:11847-11855. [PMID: 34609142 DOI: 10.1021/acs.jafc.1c04212] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Estrogen and its analogues are ubiquitous in agricultural environments, with large biological functions of oocyte development. Gap junction intercellular communications (GJICs) are the structural basis in cumulus-oocyte complexes (COCs) and regulate oocyte maturation and developmental material transport through a number of pathways. This study mainly determines the effect and potential mechanism of estrogen (17β-estradiol) in regulating GJICs in porcine COCs. In our study, 17β-estradiol increased porcine nuclear maturation in a time-dependent manner. The analysis revealed that 17β-estradiol upregulated the autophagy in COCs during in vitro maturation. In contrast with the control, 17β-estradiol decreased GJICs in a time-dependent manner between cumulus cells and oocytes, while it was consistent with the control group at 24 h. Carbenoxolone (CBX) blocks GJICs as a negative control group used in our system. Autophagy inhibitor autophinib decreased oocyte maturation, and the reduced nuclear maturation treated with autophinib was abolished by 17β-estradiol. Besides, the upregulation effect of autophinib on GJICs and transzonal projections (TZPs) was decreased by 17β-estradiol. 17β-Estradiol could reduce serine 368 phosphorylation of connexin 43 (Cx43) protein by autophinib in porcine COCs. These results were dependent upon the MEK/ERK signaling pathway. Furthermore, 17β-estradiol-induced GJICs and Cx43 phosphorylation were inhibited by autophinib or the MEK/ERK pathway inhibitors (Trametinib and FR 180204), indicating that 17β-estradiol regulated GJICs through the MEK/ERK signaling pathway. In conclusion, 17β-estradiol improves the autophagy-mediated nuclear maturation with downregulating GJICs and TZPs in porcine COCs. Such an effect occurs by phosphorylation of Cx43, which was regulated via the MEK/ERK signaling pathway.
Collapse
Affiliation(s)
- Jiaxin Duan
- College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang, Shaanxi 712100, People's Republic of China
| | - Huali Chen
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan 621000, People's Republic of China
| | - Yuan Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang, Shaanxi 712100, People's Republic of China
| | - Dejun Xu
- College of Animal Science and Technology, Southwest University, Chongqing, Sichuan 400000, People's Republic of China
| | - Xiaoya Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang, Shaanxi 712100, People's Republic of China
| | - Zelin Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang, Shaanxi 712100, People's Republic of China
| | - Jianyong Cheng
- College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang, Shaanxi 712100, People's Republic of China
| | - Li Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang, Shaanxi 712100, People's Republic of China
| | - Qingwang Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang, Shaanxi 712100, People's Republic of China
| |
Collapse
|
34
|
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: 26] [Impact Index Per Article: 6.5] [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
|
35
|
Daei-Farshbaf N, Aflatoonian R, Amjadi FS, Nikniyaz H, Taleahmad S, Bakhtiyari M. Identification of calcineurin as a predictor of oocyte quality and fertilization competence based on microarray data. Comput Biol Chem 2021; 94:107561. [PMID: 34461466 DOI: 10.1016/j.compbiolchem.2021.107561] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/26/2021] [Accepted: 08/09/2021] [Indexed: 11/19/2022]
Abstract
OBJECTIVE The aim of our study was to detect a biomarker for selection of competent oocytes with acceptable fertilization potential. Calcium ion fluctuation play the most critical role of modulating intercellular signaling pathways in oocyte maturation, egg activation and the egg-to-embryo transition. Since, the stimulatory action of calcium ion is mediated by binding to certain proteins, the calcium/calmodulin-binding genes (CBGs), as the main calcium binding group, was analyzed in detail. METHODS In this work, bioinformatics analysis was conducted on the CBGs of human cumulus cells (CCs) to elucidate a reliable biomarker for fertile oocyte selection. Calcineurin (CaN) or protein phosphatase 3 (PPP3) was selected which consists of a catalytic subunit A with PPP3CA (Aα), PPP3CB (Aβ), and PPP3CC (Aγ) isoforms and a regulatory subunit B. Whereas CaN A regulates calcium ion function, our study gives insights to probable role of related isoforms within human oogenesis process. The presence of CaN A in CCs surrounding growing and mature oocytes was confirmed by western blotting and the expression patterns of related isoforms were examined by reverse transcription-quantitative PCR (RT-qPCR). RESULTS Our results indicated the increased expression of the catalytic subunit of CaN protein in the CCs of metaphase (M) II oocytes. The expression level of PPP3CB was significantly elevated in CCs of fertile MII compared with those in the germinal vesicle (GV), MI and unfertilized MII oocytes (P ≤ 0.05). CONCLUSION Elevated level of PPP3CB isoform in the CCs of fertile MII oocyte could be a reliable indication of oocyte fertilization potential. However, further researches are required to introduce CaN Aβ as an appropriate biomarker for oocyte selection in assisted reproduction technique (ART) programs.
Collapse
Affiliation(s)
- Neda Daei-Farshbaf
- Department of Anatomy, Faculty of Medicine, Iran University of Medical Sciences, P.O. Box: 14155-5983, Tehran, Iran
| | - Reza Aflatoonian
- Department of Endocrinology and Female Infertility, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, Academic Center for Education, Culture and Research, P.O. Box: 16635-148, Tehran, Iran
| | - Fatemeh-Sadat Amjadi
- Department of Anatomy, Faculty of Medicine, Iran University of Medical Sciences, P.O. Box: 14155-5983, Tehran, Iran; Cellular and Molecular Research Center, Faculty of Medicine, Iran University of Medical Sciences, P.O. Box:14155-5983, Tehran, Iran
| | - Hossein Nikniyaz
- Department of Anatomy, Faculty of Medicine, Iran University of Medical Sciences, P.O. Box: 14155-5983, Tehran, Iran
| | - Sara Taleahmad
- Department of Molecular Systems Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology (RI-SCBT), Academic Center for Education, Culture and Research, P.O. Box: 16635-148, Tehran, Iran
| | - Mehrdad Bakhtiyari
- Department of Anatomy, Faculty of Medicine, Iran University of Medical Sciences, P.O. Box: 14155-5983, Tehran, Iran; Cellular and Molecular Research Center, Faculty of Medicine, Iran University of Medical Sciences, P.O. Box:14155-5983, Tehran, Iran.
| |
Collapse
|
36
|
Juengel JL, Cushman RA, Dupont J, Fabre S, Lea RG, Martin GB, Mossa F, Pitman JL, Price CA, Smith P. The ovarian follicle of ruminants: the path from conceptus to adult. Reprod Fertil Dev 2021; 33:621-642. [PMID: 34210385 DOI: 10.1071/rd21086] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 06/06/2021] [Indexed: 11/23/2022] Open
Abstract
This review resulted from an international workshop and presents a consensus view of critical advances over the past decade in our understanding of follicle function in ruminants. The major concepts covered include: (1) the value of major genes; (2) the dynamics of fetal ovarian development and its sensitivity to nutritional and environmental influences; (3) the concept of an ovarian follicle reserve, aligned with the rise of anti-Müllerian hormone as a controller of ovarian processes; (4) renewed recognition of the diverse and important roles of theca cells; (5) the importance of follicular fluid as a microenvironment that determines oocyte quality; (6) the 'adipokinome' as a key concept linking metabolic inputs with follicle development; and (7) the contribution of follicle development to the success of conception. These concepts are important because, in sheep and cattle, ovulation rate is tightly regulated and, as the primary determinant of litter size, it is a major component of reproductive efficiency and therefore productivity. Nowadays, reproductive efficiency is also a target for improving the 'methane efficiency' of livestock enterprises, increasing the need to understand the processes of ovarian development and folliculogenesis, while avoiding detrimental trade-offs as greater performance is sought.
Collapse
Affiliation(s)
- Jennifer L Juengel
- AgResearch Ltd, Invermay Agricultural Centre, Mosgiel, New Zealand; and Corresponding author
| | - Robert A Cushman
- Livestock Biosystems Research Unit, US Department of Agriculture, Agricultural Research Service, US Meat Animal Research Center, Clay Center, NE, USA
| | - Joëlle Dupont
- INRAE Institute UMR85 Physiologie de la Reproduction et des Comportements, Tours University, France
| | - Stéphane Fabre
- GenPhySE, Université de Toulouse, Institut national de recherche pour l'agriculture, l'alimentation et l'environnement, Institut national polytechnique de Toulouse, Ecole nationale vétérinaire de Toulouse, Castanet Tolosan, France
| | - Richard G Lea
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington, UK
| | - Graeme B Martin
- UWA Institute of Agriculture, University of Western Australia, Perth, WA, Australia
| | - Francesca Mossa
- Dipartimento di Medicina Veterinaria, Università degli Studi di Sassari, Italy
| | - Janet L Pitman
- School of Biological Sciences, Victoria University of Wellington, New Zealand
| | - Christopher A Price
- Faculty of Veterinary Medicine, Université de Montréal, Montréal, QC, Canada
| | - Peter Smith
- AgResearch Ltd, Invermay Agricultural Centre, Mosgiel, New Zealand
| |
Collapse
|
37
|
Frost ER, Ford EA, Peters AE, Reed NL, McLaughlin EA, Baker MA, Lovell-Badge R, Sutherland JM. Signal transducer and activator of transcription (STAT) 1 and STAT3 are expressed in the human ovary and have Janus kinase 1-independent functions in the COV434 human granulosa cell line. Reprod Fertil Dev 2021; 32:1027-1039. [PMID: 32758351 DOI: 10.1071/rd20098] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 06/29/2020] [Indexed: 01/03/2023] Open
Abstract
Ovarian granulosa cells are fundamental for oocyte maintenance and maturation. Recent studies have demonstrated the importance of members of the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signalling pathway in the granulosa cell population of mouse and horse ovaries, with perturbation of JAK1 signalling in the mouse shown to impair oocyte maintenance and accelerate primordial follicle activation. The presence and role of the JAK/STAT pathway in human granulosa cells has yet to be elucidated. In this study, expression of JAK1, STAT1 and STAT3 was detected in oocytes and granulosa cells of human ovarian sections from fetal (40 weeks gestation) and premenopausal ovaries (34-41 years of age; n=3). To determine the effects of JAK1 signalling in granulosa cells, the human granulosa-like cell line COV434 was used, with JAK1 inhibition using ruxolitinib. Chemical inhibition of JAK1 in COV434 cells with 100nM ruxolitinib for 72h resulted in significant increases in STAT3 mRNA (P=0.034) and p-Y701-STAT1 protein (P=0.0117), demonstrating a role for JAK1 in modulating STAT in granulosa cells. This study implicates a conserved role for JAK/STAT signalling in human ovary development, warranting further investigation of this pathway in human granulosa cell function.
Collapse
Affiliation(s)
- E R Frost
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science and Pharmacy and Environmental and Life Sciences, University of Newcastle, University Drive, Callaghan, NSW 2308, Australia; and Hunter Medical Research Institute, Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia; and Stem Cell Biology and Developmental Genetics Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK; and Corresponding author.
| | - E A Ford
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science and Pharmacy and Environmental and Life Sciences, University of Newcastle, University Drive, Callaghan, NSW 2308, Australia; and Hunter Medical Research Institute, Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia
| | - A E Peters
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science and Pharmacy and Environmental and Life Sciences, University of Newcastle, University Drive, Callaghan, NSW 2308, Australia; and Hunter Medical Research Institute, Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia
| | - N L Reed
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science and Pharmacy and Environmental and Life Sciences, University of Newcastle, University Drive, Callaghan, NSW 2308, Australia
| | - E A McLaughlin
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science and Pharmacy and Environmental and Life Sciences, University of Newcastle, University Drive, Callaghan, NSW 2308, Australia; and School of Science, Western Sydney University, Penrith, NSW 2751, Australia; and School of Biological Sciences, Faculty of Science, University of Auckland, Auckland 1142, New Zealand
| | - M A Baker
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science and Pharmacy and Environmental and Life Sciences, University of Newcastle, University Drive, Callaghan, NSW 2308, Australia; and Hunter Medical Research Institute, Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia
| | - R Lovell-Badge
- Stem Cell Biology and Developmental Genetics Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - J M Sutherland
- Priority Research Centre for Reproductive Science, Schools of Biomedical Science and Pharmacy and Environmental and Life Sciences, University of Newcastle, University Drive, Callaghan, NSW 2308, Australia; and Hunter Medical Research Institute, Kookaburra Circuit, New Lambton Heights, NSW 2305, Australia
| |
Collapse
|
38
|
Garner TB, Hester JM, Carothers A, Diaz FJ. Role of zinc in female reproduction. Biol Reprod 2021; 104:976-994. [PMID: 33598687 PMCID: PMC8599883 DOI: 10.1093/biolre/ioab023] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 01/09/2021] [Accepted: 02/15/2021] [Indexed: 11/14/2022] Open
Abstract
Zinc is a critical component in a number of conserved processes that regulate female germ cell growth, fertility, and pregnancy. During follicle development, a sufficient intracellular concentration of zinc in the oocyte maintains meiotic arrest at prophase I until the germ cell is ready to undergo maturation. An adequate supply of zinc is necessary for the oocyte to form a fertilization-competent egg as dietary zinc deficiency or chelation of zinc disrupts maturation and reduces the oocyte quality. Following sperm fusion to the egg to initiate the acrosomal reaction, a quick release of zinc, known as the zinc spark, induces egg activation in addition to facilitating zona pellucida hardening and reducing sperm motility to prevent polyspermy. Symmetric division, proliferation, and differentiation of the preimplantation embryo rely on zinc availability, both during the oocyte development and post-fertilization. Further, the fetal contribution to the placenta, fetal limb growth, and neural tube development are hindered in females challenged with zinc deficiency during pregnancy. In this review, we discuss the role of zinc in germ cell development, fertilization, and pregnancy with a focus on recent studies in mammalian females. We further detail the fundamental zinc-mediated reproductive processes that have only been explored in non-mammalian species and speculate on the role of zinc in similar mechanisms of female mammals. The evidence collected over the last decade highlights the necessity of zinc for normal fertility and healthy pregnancy outcomes, which suggests zinc supplementation should be considered for reproductive age women at risk of zinc deficiency.
Collapse
Affiliation(s)
- Tyler Bruce Garner
- Huck Institutes of the Life Sciences, Integrative and Biomedical Physiology Program, The Pennsylvania State University, University Park, PA, USA
| | - James Malcolm Hester
- Huck Institutes of the Life Sciences, Integrative and Biomedical Physiology Program, The Pennsylvania State University, University Park, PA, USA
| | - Allison Carothers
- Huck Institutes of the Life Sciences, Integrative and Biomedical Physiology Program, The Pennsylvania State University, University Park, PA, USA
| | - Francisco J Diaz
- Huck Institutes of the Life Sciences, Integrative and Biomedical Physiology Program, The Pennsylvania State University, University Park, PA, USA
- Department of Animal Science, The Pennsylvania State University, University Park, PA, USA
| |
Collapse
|
39
|
Poulsen LC, Bøtkjær JA, Østrup O, Petersen KB, Andersen CY, Grøndahl ML, Englund ALM. Two waves of transcriptomic changes in periovulatory human granulosa cells. Hum Reprod 2021; 35:1230-1245. [PMID: 32378719 DOI: 10.1093/humrep/deaa043] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 02/05/2020] [Indexed: 11/14/2022] Open
Abstract
STUDY QUESTION How does the human granulosa cell (GC) transcriptome change during ovulation? SUMMARY ANSWER Two transcriptional peaks were observed at 12 h and at 36 h after induction of ovulation, both dominated by genes and pathways known from the inflammatory system. WHAT IS KNOWN ALREADY The crosstalk between GCs and the oocyte, which is essential for ovulation and oocyte maturation, can be assessed through transcriptomic profiling of GCs. Detailed transcriptional changes during ovulation have not previously been assessed in humans. STUDY DESIGN, SIZE, DURATION This prospective cohort study comprised 50 women undergoing fertility treatment in a standard antagonist protocol at a university hospital-affiliated fertility clinic in 2016-2018. PARTICIPANTS/MATERIALS, SETTING, METHODS From each woman, one sample of GCs was collected by transvaginal ultrasound-guided follicle aspiration either before or 12 h, 17 h or 32 h after ovulation induction (OI). A second sample was collected at oocyte retrieval, 36 h after OI. Total RNA was isolated from GCs and analyzed by microarray. Gene expression differences between the five time points were assessed by ANOVA with a random factor accounting for the pairing of samples, and seven clusters of protein-coding genes representing distinct expression profiles were identified. These were used as input for subsequent bioinformatic analyses to identify enriched pathways and suggest upstream regulators. Subsets of genes were assessed to explore specific ovulatory functions. MAIN RESULTS AND THE ROLE OF CHANCE We identified 13 345 differentially expressed transcripts across the five time points (false discovery rate, <0.01) of which 58% were protein-coding genes. Two clusters of mainly downregulated genes represented cell cycle pathways and DNA repair. Upregulated genes showed one peak at 12 h that resembled the initiation of an inflammatory response, and one peak at 36 h that resembled the effector functions of inflammation such as vasodilation, angiogenesis, coagulation, chemotaxis and tissue remodelling. Genes involved in cell-matrix interactions as a part of cytoskeletal rearrangement and cell motility were also upregulated at 36 h. Predicted activated upstream regulators of ovulation included FSH, LH, transforming growth factor B1, tumour necrosis factor, nuclear factor kappa-light-chain-enhancer of activated B cells, coagulation factor 2, fibroblast growth factor 2, interleukin 1 and cortisol, among others. The results confirmed early regulation of several previously described factors in a cascade inducing meiotic resumption and suggested new factors involved in cumulus expansion and follicle rupture through co-regulation with previously described factors. LARGE SCALE DATA The microarray data were deposited to the Gene Expression Omnibus (www.ncbi.nlm.nih.gov/gds/, accession number: GSE133868). LIMITATIONS, REASONS FOR CAUTION The study included women undergoing ovarian stimulation and the findings may therefore differ from a natural cycle. However, the results confirm significant regulation of many well-established ovulatory genes from a series of previous studies such as amphiregulin, epiregulin, tumour necrosis factor alfa induced protein 6, tissue inhibitor of metallopeptidases 1 and plasminogen activator inhibitor 1, which support the relevance of the results. WIDER IMPLICATIONS OF THE FINDINGS The study increases our understanding of human ovarian function during ovulation, and the publicly available dataset is a valuable resource for future investigations. Suggested upstream regulators and highly differentially expressed genes may be potential pharmaceutical targets in fertility treatment and gynaecology. STUDY FUNDING/COMPETING INTEREST(S) The study was funded by EU Interreg ÔKS V through ReproUnion (www.reprounion.eu) and by a grant from the Region Zealand Research Foundation. None of the authors have any conflicts of interest to declare.
Collapse
Affiliation(s)
- L C Poulsen
- Fertility Clinic, Zealand University Hospital, Lykkebækvej 14, 4600 Køge, Denmark
| | - J A Bøtkjær
- Laboratory of Reproductive Biology, University Hospital of Copenhagen, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen Ø, Denmark
| | - O Østrup
- Center for Genomic Medicine, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen Ø, Denmark
| | - K B Petersen
- Fertility Clinic, Zealand University Hospital, Lykkebækvej 14, 4600 Køge, Denmark
| | - C Yding Andersen
- Laboratory of Reproductive Biology, University Hospital of Copenhagen, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen Ø, Denmark
| | - M L Grøndahl
- Fertility Clinic, University Hospital of Copenhagen, Herlev and Gentofte Hospital, Herlev Ringvej 75, 2730 Herlev, Denmark
| | - A L M Englund
- Fertility Clinic, Zealand University Hospital, Lykkebækvej 14, 4600 Køge, Denmark
| |
Collapse
|
40
|
Dozortsev DI, Diamond MP. Luteinizing hormone-independent rise of progesterone as the physiological trigger of the ovulatory gonadotropins surge in the human. Fertil Steril 2021; 114:191-199. [PMID: 32741458 DOI: 10.1016/j.fertnstert.2020.06.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 06/08/2020] [Accepted: 06/09/2020] [Indexed: 02/09/2023]
Abstract
The current ovarian cycle paradigm postulates that ovulation is triggered by a critically sustained elevation of estradiol. However, an in-depth look into the published data reveals considerable uncertainty about the relative roles of progesterone and estradiol in the ovulation process.This review provides compelling evidences that the role of estradiol in ovulation has been misinterpreted and that the true physiological trigger of ovulation is a luteinizing hormone-independent preovulatory progesterone surge in the circulation to approximately 0.5 ng/mL. Furthermore, the current work reconciles the ability of progesterone to trigger ovulation, with its well-established ability to block ovulation during pregnancy, or when administered in the form of a synthetic progestin in birth control formulations and with experimental data that estradiol benzoate triggers ovulation in the complete absence of progesterone.
Collapse
|
41
|
Palomino J, Flores J, Ramirez G, Parraguez VH, De los Reyes M. Expression Profiles of the Progesterone Receptor, Cyclooxygenase-2, Growth Differentiation Factor 9, and Bone Morphogenetic Protein 15 Transcripts in the Canine Oviducts during the Oestrous Cycle. Animals (Basel) 2021; 11:454. [PMID: 33572466 PMCID: PMC7916196 DOI: 10.3390/ani11020454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/30/2021] [Accepted: 02/05/2021] [Indexed: 11/29/2022] Open
Abstract
The gene expression in the canine oviduct, where oocyte maturation, fertilization, and early embryonic development occur, is still elusive. This study determined the oviductal expression of (PR), cyclooxygenase-2 (COX-2), growth differentiation factor 9 (GDF-9), and bone morphogenetic protein 15 (BMP-15) during the canine oestrous cycle. Samples were collected from bitches at anoestrus (9), proestrus (7), oestrus (8), and dioestrus (11), after routine ovariohysterectomy and the ovarian surface structures and plasma progesterone concentration evaluated the physiological status of each donor. The oviductal cells were isolated and pooled. Total RNA was isolated, and gene expression was assessed by qPCR followed by analysis using the t-test and ANOVA. The PR mRNA increased (P < 0.05) from the anoestrus to dioestrus with the plasma progesterone concentration (r = 0.8). COX-2 mRNA expression was low in the anoestrus and proestrus, and negligible in the oestrus, while it was around 10-fold higher (P < 0.05) in the dioestrus. The GDF-9 mRNA was expressed during all phases of the oestrous cycle and was most abundant (P < 0.05) during oestrus phase. The BMP-15 mRNA decreased (P < 0.05) in the anoestrus and proestrus phases. Thus, the transcripts were differentially expressed in a stage-dependent manner, suggesting the importance of oestrous cycle regulation for successful reproduction in dogs.
Collapse
Affiliation(s)
- Jaime Palomino
- Laboratory of Animal Reproduction, Department of Animal Production, Faculty of Veterinary Sciences, University of Chile, Santiago 8820000, Chile; (J.P.); (J.F.); (G.R.)
| | - Javiera Flores
- Laboratory of Animal Reproduction, Department of Animal Production, Faculty of Veterinary Sciences, University of Chile, Santiago 8820000, Chile; (J.P.); (J.F.); (G.R.)
| | - Georges Ramirez
- Laboratory of Animal Reproduction, Department of Animal Production, Faculty of Veterinary Sciences, University of Chile, Santiago 8820000, Chile; (J.P.); (J.F.); (G.R.)
| | - Victor H. Parraguez
- Laboratory of Animal Physiology, Department of Biological Sciences, Faculty of Veterinary Sciences, University of Chile, Santiago 8820000, Chile;
| | - Monica De los Reyes
- Laboratory of Animal Reproduction, Department of Animal Production, Faculty of Veterinary Sciences, University of Chile, Santiago 8820000, Chile; (J.P.); (J.F.); (G.R.)
| |
Collapse
|
42
|
Yang F, Liu Q, Chen Y, Ye H, Wang H, Zeng S. Integrative Proteomic and Phosphoproteomic Analyses of Granulosa Cells During Follicular Atresia in Porcine. Front Cell Dev Biol 2021; 8:624985. [PMID: 33520998 PMCID: PMC7843964 DOI: 10.3389/fcell.2020.624985] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 12/09/2020] [Indexed: 12/16/2022] Open
Abstract
Ovarian follicular atresia is a natural physiological process; however, the mechanism is not fully understood. In this study, quantitative proteomic and phosphoproteomic analyses of granulosa cells (GCs) in healthy (H), slightly atretic (SA), and atretic follicles (A) of porcine were performed by TMT labeling, enrichment of phosphopeptides, and liquid chromatography with tandem mass spectrometry (LC-MS/MS) analysis. In total, 6,201 proteins were quantified, and 4,723 phosphorylation sites of 1,760 proteins were quantified. In total, 24 (11 up, 13 down) and 50 (29 up, 21 down) proteins with a fold change (FC) > 5 were identified in H/SA and H/A, respectively. In addition, there were 20 (H/SA, up) and 39 (H/A, up) phosphosites with an FC > 7 that could serve as potential biomarkers for distinguishing different quality categories of follicles. Western blotting and immunofluorescence confirmed the reliability of the proteomic analysis. Some key proteins (e.g., MIF, beta catenin, integrin β2), phosphosites (e.g., S76 of caspase6, S22 and S636 of lamin A/C), pathways (e.g., apoptosis, regulation of actin cytoskeleton pathway), transcription factors (e.g., STAT5A, FOXO1, and BCLAF1), and kinases (e.g., PBK, CDK5, CDK12, and AKT3) involved in the atresia process were revealed via further analysis of the differentially expressed proteins (DEPs) and phosphorylated proteins (DEPPs). Further study showed that mutant caspase6 Ser76 to Ala increased the ratios of cleaved caspase6/caspase6 and cleaved caspase3/caspase3 and dephosphorylation of caspase6 at Ser76 increased cell apoptotic rate, a new potential pathway of follicular atresia. Collectively, the proteomic and phosphoproteomic profiling and functional research in the current study comprehensively analyzed the dynamic changes in protein expression and phosphorylation during follicular atresia and provided some new explanations regarding the regulation of this process.
Collapse
Affiliation(s)
- Feng Yang
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Qiang Liu
- Beijing Advanced Innovation Center for Genomics, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Yanhong Chen
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Huizhen Ye
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Han Wang
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shenming Zeng
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
43
|
Norris RP, Terasaki M. Gap junction internalization and processing in vivo: a 3D immuno-electron microscopy study. J Cell Sci 2021; 134:jcs252726. [PMID: 33277382 DOI: 10.1242/jcs.252726] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 11/26/2020] [Indexed: 12/15/2022] Open
Abstract
Gap junctions have well-established roles in cell-cell communication by way of forming permeable intercellular channels. Less is understood about their internalization, which forms double membrane vesicles containing cytosol and membranes from another cell called connexosomes or annular gap junctions. Here, we systematically investigated the fate of connexosomes in intact ovarian follicles. High-pressure frozen, serial-sectioned tissue was immunogold labeled for connexin 43 (Cx43, also known as GJA1). Within a volume corresponding to ∼35 cells, every labeled structure was categorized and had its surface area measured. Measurements support the concept that multiple connexosomes form from larger invaginated gap junctions. Subsequently, the inner and outer membranes separate, Cx43 immunogenicity is lost from the outer membrane, and the inner membrane appears to undergo fission. One pathway for processing involves lysosomes, based on localization of cathepsin B to some processed connexosomes. In summary, this study demonstrates new technology for high-resolution analyses of gap junction processing.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Rachael P Norris
- Department of Cell Biology, UConn Health, 263 Farmington Ave, Farmington, CT 06030, USA
| | - Mark Terasaki
- Department of Cell Biology, UConn Health, 263 Farmington Ave, Farmington, CT 06030, USA
| |
Collapse
|
44
|
Gingrich J, Pu Y, Upham BL, Hulse M, Pearl S, Martin D, Avery A, Veiga-Lopez A. Bisphenol S enhances gap junction intercellular communication in ovarian theca cells. CHEMOSPHERE 2021; 263:128304. [PMID: 33155548 PMCID: PMC7726030 DOI: 10.1016/j.chemosphere.2020.128304] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 09/04/2020] [Accepted: 09/10/2020] [Indexed: 05/08/2023]
Abstract
Gap junction intercellular communication (GJIC) is necessary for ovarian function, and it is temporospatially regulated during follicular development and ovulation. At outermost layer of the antral follicle, theca cells provide structural, steroidogenic, and vascular support. Inter- and extra-thecal GJIC is required for intrafollicular trafficking of signaling molecules. Because GJIC can be altered by hormones and endocrine disrupting chemicals (EDCs), we tested if any of five common EDCs (bisphenol A (BPA), bisphenol S (BPS), bisphenol F (BPF), perfluorooctanesulfonic acid (PFOS), and triphenyltin chloride (TPT)) can interfere with theca cell GJIC. Since most chemicals are reported to repress GJIC, we hypothesized that all chemicals tested, within environmentally relevant human exposure concentrations, will inhibit theca cell GJICs. To evaluate this hypothesis, we used a scrape loading/dye transfer assay. BPS, but no other chemical tested, enhanced GJIC in a dose- and time-dependent manner in ovine primary theca cells. A signal-protein inhibitor approach was used to explore the GJIC-modulatory pathways involved. Phospholipase C and mitogen-activated protein kinase (MAPK) inhibitors significantly attenuated BPS-induced enhanced GJIC. Human theca cells were used to evaluate translational relevance of these findings. Human primary theca cells had a ∼40% increase in GJIC in response to BPS, which was attenuated with a MAPK inhibitor, suggestive of a conserved mechanism. Upregulation of GJIC could result in hyperplasia of the theca cell layer or prevent ovulation by holding the oocyte in meiotic arrest. Further studies are necessary to understand in vitro to in vivo translatability of these findings on follicle development and fertility outcomes.
Collapse
Affiliation(s)
- Jeremy Gingrich
- Department of Animal Science, Michigan State University, East Lansing, MI, 48824, USA; Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, 48824, USA
| | - Yong Pu
- Department of Animal Science, Michigan State University, East Lansing, MI, 48824, USA
| | - Brad L Upham
- Department of Pediatrics and Human Development, Michigan State University, East Lansing, MI, 48824, USA
| | - Madeline Hulse
- Department of Obstetrics and Gynecology, Sparrow Health System, Lansing, MI, 48912, USA
| | - Sarah Pearl
- Department of Obstetrics and Gynecology, Sparrow Health System, Lansing, MI, 48912, USA
| | - Denny Martin
- Department of Obstetrics and Gynecology, Sparrow Health System, Lansing, MI, 48912, USA
| | - Anita Avery
- Department of Obstetrics and Gynecology, Sparrow Health System, Lansing, MI, 48912, USA; Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, East Lansing, MI, 48824, USA
| | - Almudena Veiga-Lopez
- Department of Animal Science, Michigan State University, East Lansing, MI, 48824, USA; Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
45
|
Tokmakov AA, Stefanov VE, Sato KI. Dissection of the Ovulatory Process Using ex vivo Approaches. Front Cell Dev Biol 2020; 8:605379. [PMID: 33363163 PMCID: PMC7755606 DOI: 10.3389/fcell.2020.605379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 11/19/2020] [Indexed: 12/23/2022] Open
Abstract
Ovulation is a unique physiological phenomenon that is essential for sexual reproduction. It refers to the entire process of ovarian follicle responses to hormonal stimulation resulting in the release of mature fertilization-competent oocytes from the follicles and ovaries. Remarkably, ovulation in different species can be reproduced out-of-body with high fidelity. Moreover, most of the molecular mechanisms and signaling pathways engaged in this process have been delineated using in vitro ovulation models. Here, we provide an overview of the major molecular and cytological events of ovulation observed in frogs, primarily in the African clawed frog Xenopus laevis, using mainly ex vivo approaches, with the focus on meiotic oocyte maturation and follicle rupture. For the purpose of comparison and generalization, we also refer extensively to ovulation in other biological species, most notoriously, in mammals.
Collapse
Affiliation(s)
| | - Vasily E Stefanov
- Department of Biochemistry, Saint Petersburg State University, Saint Petersburg, Russia
| | - Ken-Ichi Sato
- Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, Japan
| |
Collapse
|
46
|
Zhang H, Lu S, Xu R, Tang Y, Liu J, Li C, Wei J, Yao R, Zhao X, Wei Q, Ma B. Mechanisms of Estradiol-induced EGF-like Factor Expression and Oocyte Maturation via G Protein-coupled Estrogen Receptor. Endocrinology 2020; 161:5929646. [PMID: 33068422 DOI: 10.1210/endocr/bqaa190] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Indexed: 12/18/2022]
Abstract
Estrogen is an important modulator of reproductive activity through nuclear receptors and G protein-coupled estrogen receptor (GPER). Here, we observed that both estradiol and the GPER-specific agonist G1 rapidly induced cyclic adenosine monophosphate (cAMP) production in cumulus cells, leading to transient stimulation of phosphorylated cAMP response element binding protein (CREB), which was conducive to the transcription of epidermal growth factor (EGF)-like factors, amphiregulin, epiregulin, and betacellulin. Inhibition of GPER by G15 significantly reduced estradiol-induced CREB phosphorylation and EGF-like factor gene expression. Consistently, the silencing of GPER expression in cultured cumulus cells abrogated the estradiol-induced CREB phosphorylation and EGF-like factor transcription. In addition, the increase in EGF-like factor expression in the cumulus cells is associated with EGF receptor (EFGR) tyrosine kinase phosphorylation and extracellular signal-regulated kinase 1/2 (ERK1/2) activation. Furthermore, we demonstrated that GPER-mediated phosphorylation of EGFR and ERK1/2 was involved in reduced gap junction communication, cumulus expansion, increased oocyte mitochondrial activity and first polar body extrusion. Overall, our study identified a novel function for estrogen in regulating EGFR activation via GPER in cumulus cells during oocyte maturation.
Collapse
Affiliation(s)
- 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
| | - 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
| | - 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
| | - 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
| | - 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
| | - 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
| | - 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
| | - 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
| | - 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
|
47
|
Casillas F, Ducolomb Y, López A, Betancourt M. Effect of porcine immature oocyte vitrification on oocyte-cumulus cell gap junctional intercellular communication. Porcine Health Manag 2020; 6:37. [PMID: 33292603 PMCID: PMC7687833 DOI: 10.1186/s40813-020-00175-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 11/06/2020] [Indexed: 01/27/2023] Open
Abstract
Vitrification may severely affect cumulus cells and oocyte morphology and viability, limiting their maturation and developmental potential. The aim of this study was to evaluate the gap junction intercellular communication (GJIC) integrity after the vitrification of porcine immature cumulus-oocyte complexes (COCs). Fresh COCs were randomly distributed in three groups: untreated (control), toxicity (cryoprotectants exposure), and vitrification, then subjected to in vitro maturation (IVM). Oocyte viability and IVM were measured in all groups. The evaluation of GJIC was expressed as relative fluorescence intensity (RFI). Vitrification significantly decreased oocyte viability and maturation after 44 h of culture compared to control. Also, significantly reduced RFI was observed in vitrified COCs during the first hours of culture (4-8 h) compared to control. This study demonstrates that porcine oocyte viability and maturation after 44 h of culture decreased after vitrification. GJIC was also affected during the first hours of culture after the vitrification of immature oocytes, being one of the possible mechanisms by which oocyte maturation decreased.
Collapse
Affiliation(s)
- Fahiel Casillas
- Departamento de Biología de la Reproducción, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, 09340, CDMX, México.
| | - Yvonne Ducolomb
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, 09340, CDMX, México
| | - Alma López
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, 09340, CDMX, México
| | - Miguel Betancourt
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, 09340, CDMX, México
| |
Collapse
|
48
|
Silvestri G, Rathje CC, Harvey SC, Gould RL, Walling GA, Ellis PJ, Harvey KE, Griffin DK. Identification of optimal assisted aspiration conditions of oocytes for use in porcine in vitro maturation: A re‐evaluation of the relationship between the cumulus oocyte complex and oocyte quality. Vet Med Sci 2020. [PMCID: PMC8323607 DOI: 10.1002/vms3.378] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The quality of porcine oocytes for use in IVF is commonly graded according to the number of layers of cumulus cells (CCs) surrounding the oocyte; together these form the cumulus oocyte complex (COC). At least three compact layers of CCs is regarded as important for efficient IVP. To test this, oocytes were scored according to cumulus investment, with grade A representing COCs with three or more cumulus layers including granulosa cell‐cumulus oocyte complexes, grade B those with an intact corona radiata surrounded by another layer of cumulus cells and grades C and D representing COCs with lower CC investment. These oocytes were then monitored for in vitro maturation (IVM), as assessed by tubulin immunostaining for meiotic progression, the development of a cortical granule ring, and by glutathione levels. Results indicate that grading correlates closely with nuclear maturation and cytoplasmic maturation, suggesting that grading oocytes by cumulus investment is a reliable method to predict IVM success. Importantly, Grade A and B oocytes showed no significant differences in any measure and hence using a cut‐off of two or more CC layers may be optimal. We also determined the effect of assisted aspiration for oocyte retrieval, comparing the effect of needle size and applied pressure on the retrieval rate. These data indicated that both variables affected oocyte recovery rates and the quality of recovered oocytes. In combination, these experiments indicate that grade A and B oocytes have a similar developmental potential and that the recovery of oocytes of these grades is maximised by use of an 18‐gauge needle and 50 mmHg aspiration pressure.
Collapse
Affiliation(s)
| | | | - Simon C. Harvey
- School of Psychology and Life Sciences Canterbury Christ Church University Canterbury UK
| | | | | | | | - Katie E. Harvey
- School of Psychology and Life Sciences Canterbury Christ Church University Canterbury UK
| | | |
Collapse
|
49
|
Kowsar R, Mansouri A, Sadeghi N, Abadi MHA, Ghoreishi SM, Sadeghi K, Miyamoto A. A multilevel analysis identifies the different relationships between amino acids and the competence of oocytes matured individually or in groups. Sci Rep 2020; 10:16082. [PMID: 32999417 PMCID: PMC7528030 DOI: 10.1038/s41598-020-73225-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 09/11/2020] [Indexed: 11/03/2022] Open
Abstract
High-protein diets contribute to an increase in urea follicular concentrations associated with decreased fertility. Urea has been shown to interfere with the epidermal growth factor (EGF)/EGFR system, which has been shown to have a beneficial effect during in vitro maturation (IVM) of oocytes. Of note, the number of cumulus-oocyte complexes (COCs) in the maturation medium can change the maturation and the developmental competence of COCs. Therefore, it was hypothesized that, the presence of urea and EGF may have a differential effect on the depletion/appearance of AAs and competence of COCs matured individually (I-IVM system) or in groups (G-IVM system). In the G-IVM system, COCs increased consumption (depletion) of AAs compared with other groups in the presence of high-level urea (40 mg/dl) + EGF (10 ng/ml). In the I-IVM system, the non-cleaved COCs depleted more AAs than the cleaved COCs, in particular in the presence of urea. The combination of urea and EGF increased the depletion of AAs in the G-IVM system. However, the EGF abrogated the urea-induced depletion of AAs by the I-IVM COCs. The use of N-acetyl-L-cysteine as an EGFR inhibitor canceled urea-induced depletion of AAs. This shows the inhibiting effect of urea over the EGF/EGFR system. In the presence of urea + EGF, COCs had a lower degree of developmental competence than control in both I- and G-IVM systems. Arginine had the best predictive power to identify highly competent COCs in the G-IVM system, while glutamine was the best predictor of the cleavage in the I-IVM system. In conclusion, this multi-level study shows that COCs matured individually or in groups may have different association with AAs metabolism. These findings provide new insights into the relationships between AA metabolism and the subsequent developmental competence of COCs.
Collapse
Affiliation(s)
- Rasoul Kowsar
- Department of Animal Sciences, College of Agriculture, Isfahan University of Technology, 84156-83111, Isfahan, Iran. .,Global Agromedicine Research Center (GAMRC), Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, 080-8555, Japan.
| | - Alireza Mansouri
- Global Agromedicine Research Center (GAMRC), Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, 080-8555, Japan
| | - Nima Sadeghi
- FKA, Animal Husbandry and Agriculture Co, Isfahan, Iran
| | - Mohammad Heidaran Ali Abadi
- Department of Animal Sciences, College of Agriculture, Isfahan University of Technology, 84156-83111, Isfahan, Iran
| | - Seyed Mehdi Ghoreishi
- Department of Animal Sciences, College of Agriculture, Shiraz University, Shiraz, Iran
| | - Khaled Sadeghi
- Department of Animal Sciences, College of Agriculture, Isfahan University of Technology, 84156-83111, Isfahan, Iran
| | - Akio Miyamoto
- Global Agromedicine Research Center (GAMRC), Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, 080-8555, Japan
| |
Collapse
|
50
|
Mehlmann LM, Uliasz TF, Lowther KM. SNAP23 is required for constitutive and regulated exocytosis in mouse oocytes†. Biol Reprod 2020; 101:338-346. [PMID: 31201423 DOI: 10.1093/biolre/ioz106] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 04/17/2019] [Accepted: 06/10/2019] [Indexed: 12/25/2022] Open
Abstract
Mammalian oocytes are stored in the ovary for prolonged periods, and arrested in meiotic prophase. During this period, their plasma membranes are constantly being recycled by endocytosis and exocytosis. However, the function of this membrane turnover is unknown. Here, we investigated the requirement for exocytosis in the maintenance of meiotic arrest. Using Trim-away, a newly developed method for rapidly and specifically depleting proteins in oocytes, we have identified the SNARE protein, SNAP23, to be required for meiotic arrest. Degradation of SNAP23 causes premature meiotic resumption in follicle-enclosed oocytes. The reduction in SNAP23 is associated with loss of gap junction communication between the oocyte and surrounding follicle cells. Reduction of SNAP23 protein also inhibits regulated exocytosis in response to a Ca2+ stimulus (cortical granule exocytosis), as measured by lectin staining and cleavage of ZP2. Our results show an essential role for SNAP23 in two key processes that occur in mouse oocytes and eggs.
Collapse
Affiliation(s)
- Lisa M Mehlmann
- Department of Cell Biology, UConn Health, Farmington, Connecticut, USA
| | - Tracy F Uliasz
- Department of Cell Biology, UConn Health, Farmington, Connecticut, USA
| | - Katie M Lowther
- Department of Cell Biology, UConn Health, Farmington, Connecticut, USA
| |
Collapse
|