1
|
Du X, Fu Y, Tian Z, Liu H, Xin H, Fu X, Wang F, Zhang H, Zeng X. Microcystin-LR accelerates follicular atresia in mice via JNK-mediated adherent junction damage of ovarian granulosa cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 252:114592. [PMID: 36731181 DOI: 10.1016/j.ecoenv.2023.114592] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 01/25/2023] [Accepted: 01/29/2023] [Indexed: 06/18/2023]
Abstract
Microcystin-LR (MC-LR), one of aquatic environmental contaminants with reproductive toxicity produced by cyanobacterial blooms, but its toxic effects and mechanisms on the ovary are not fully understood. Here, proteomic techniques and molecular biology experiments were performed to study the potential mechanism of MC-LR-caused ovarian toxicity. Results showed that protein expression profile of ovarian granulosa cells (KK-1) was changed by 17 μg/mL MC-LR exposure. Comparing with the control group, 118 upregulated proteins as well as 97 downregulated proteins were identified in MC-LR group. Function of differentially expressed proteins was found to be enriched in pathways related to adherent junction, such as cadherin binding, cell-cell junction, cell adhesion and focal adherens. Furthermore, in vitro experiments, MC-LR significantly downregulated the expression levels of proteins associated with adherent junction (β-catenin, N-cadherin, and α-catenin) as well as caused cytoskeletal disruption in KK-1 cells (P < 0.05), indicating that the adherent junction was damaged. Results of in vivo experiments have shown that after 14 days of acute MC-LR exposure (40 μg/kg), damaged adherent junction and an increased number of atretic follicles were observed in mouse ovaries. Moreover, MC-LR activated JNK, an upstream regulator of adherent junction proteins, in KK-1 cells and mouse ovarian tissues. In contrast, JNK inhibition alleviated MC-LR-induced adherent junction damage in vivo and in vitro, as well as the number of atretic follicles. Taken together, findings from the present study indicated that JNK is involved in MC-LR-induced granulosa cell adherent junction damage, which accelerated follicular atresia. Our study clarified a novel mechanism of MC-LR-caused ovarian toxicity, providing a theoretical foundation for protecting female reproductive health from environmental pollutants.
Collapse
Affiliation(s)
- Xingde Du
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Yu Fu
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Zhihui Tian
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Haohao Liu
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Hongxia Xin
- Henan Eye Institute, Henan Eye Hospital, Henan Provincial People's Hospital, Zhengzhou 450003, China
| | - Xiaoli Fu
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Fufang Wang
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Huizhen Zhang
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China.
| | - Xin Zeng
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
2
|
Chakraborty P, Anderson RL, Roy SK. Bone morphogenetic protein 2- and estradiol-17β-induced changes in ovarian transcriptome during primordial follicle formation†. Biol Reprod 2022; 107:800-812. [PMID: 35639639 PMCID: PMC9767675 DOI: 10.1093/biolre/ioac111] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 03/22/2022] [Accepted: 05/14/2022] [Indexed: 11/13/2022] Open
Abstract
Estradiol-17β has been shown to promote primordial follicle formation and to involve bone morphogenetic protein 2 (BMP2) as a downstream effector to promote primordial follicle in hamsters. However, the molecular mechanism whereby these factors regulate ovarian somatic cells to pre-granulosa cells transition leading to primordial follicle formation remains unclear. The objective of this study was to determine whether BMP2 and/or estradiol-17β would regulate the expression of specific ovarian transcriptome during pre-granulosa cells transition and primordial follicle formation in the mouse ovary. BMP2 mRNA level increased during the period of primordial follicle formation with the concurrent presence of BMP2 protein in ovarian somatic cells. Estradiol-17β but not BMP2 exposure led to increased expression of ovarian BMP2 messenger RNA (mRNA), and the effect of estradiol-17β could not be suppressed by 4-[6-[4-(1-Piperazinyl)phenyl]pyrazolo[1,5-a]pyrimidin-3-yl]quinoline dihydrochloride (LDN) 193189. BMP2 or estradiol-17β stimulated primordial follicle formation without inducing apoptosis. Ribonucleic acid-sequence analysis (RNA-seq) of ovaries exposed to exogenous BMP2 or estradiol-17β revealed differential expression of several thousand genes. Most of the differentially expressed genes, which were common between BMP2 or estradiol-17β treatment demonstrated concordant changes, suggesting that estradiol-17β and BMP2 affected the same set of genes during primordial follicle formation. Further, we have identified that estradiol-17β, in cooperation with BMP2, could affect the expression of three major transcription factors, GATA binding protein 2, GATA binding protein 4 and Early growth response 2, and one serine protease, hepsin, in pre-granulosa cells during primordial follicle formation. Taken together, results of this study suggest that estradiol-17β and BMP2 may regulate ovarian gene expression that promote somatic cells to pre-granulosa cells transition and primordial follicle formation in the mouse ovary.
Collapse
Affiliation(s)
- Prabuddha Chakraborty
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Rebecca L Anderson
- Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Shyamal K Roy
- Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE, USA.,Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
3
|
Huang C, Lv X, Chen P, Liu J, He C, Chen L, Wang H, Moness ML, Dong J, Rueda BR, Davis JS, Wang C. Human papillomavirus targets the YAP1-LATS2 feedback loop to drive cervical cancer development. Oncogene 2022; 41:3761-3777. [PMID: 35761037 PMCID: PMC10399300 DOI: 10.1038/s41388-022-02390-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 06/13/2022] [Accepted: 06/15/2022] [Indexed: 12/12/2022]
Abstract
Human papillomavirus (HPV) infection is very common in sexually active women, but cervical cancer only develops in a small fraction of HPV-infected women, suggesting that unknown intrinsic factors associated with the unique genetic/genomic background of the high-risk population play a critical role in cervical carcinogenesis. Although our previous studies have identified the hyperactivated YAP1 oncogene as a critical contributor to cervical cancer, the molecular mechanism by which YAP1 drives cervical cancer is unknown. In the present study, we found that although the hyperactivated YAP1 caused a malignant transformation of immortalized cervical epithelial cells, it induced cellular senescence in cultures of primary human cervical epithelial cells (HCvECs). However, the hyperactivated YAP1 induced malignant transformation of HCvECs in the presence of high-risk HPV E6/E7 proteins, suggesting that the hyperactivated YAP1 synergizes with HPV to initiate cervical cancer development. Our mechanistic studies demonstrate that YAP1, via up-regulating LATS2, formed a YAP1-LATS2 negative feedback loop in cervical epithelial cells to maintain homeostasis of cervical tissue. Intriguingly, we found that high-risk HPV targets LATS2 to disrupt the feedback loop leading to the malignant transformation of cervical epithelial cells. Finally, we report that mitomycin C, an FDA-approved drug that could upregulate LATS2 and drive cellular senescence in vitro and in vivo, induced a regression of cervical cancer in a pre-clinial animal model. Thus, high-risk HPV targeting the YAP1-LATS2 feedback loop represents a new mechanism of cervical cancer development.
Collapse
Affiliation(s)
- Cong Huang
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA.,Department of Dermatology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, China
| | - Xiangmin Lv
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Peichao Chen
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA.,College of Life and Environmental Science, Wenzhou University, Wenzhou, Zhejiang, 325035, China
| | - Jiyuan Liu
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Chunbo He
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA.,Fred & Pamela Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Li Chen
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA.,Key Laboratory of Animal Ecology and Conservation Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Hongbo Wang
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Madelyn L Moness
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Jixin Dong
- Fred & Pamela Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Bo R Rueda
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - John S Davis
- Fred & Pamela Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA.,Olson Center for Women's Health, Department of Obstetrics & Gynecology, University of Nebraska Medical Center, Omaha, NE, 68198, USA.,Veterans Administration Nebraska-Western Iowa Healthcare System, Omaha, NE, 68105, USA
| | - Cheng Wang
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
4
|
Epithelial and Neural Cadherin in Mammalian Fertilization: Studies in the Mouse Model. Cells 2021; 11:cells11010102. [PMID: 35011663 PMCID: PMC8750299 DOI: 10.3390/cells11010102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/20/2021] [Accepted: 12/24/2021] [Indexed: 12/18/2022] Open
Abstract
Successful mammalian fertilization requires a well-orchestrated sequence of molecular events leading to gamete fusion. Since this interaction involves Ca2+-dependent adhesion events, the participation of the Ca+2-dependent cell-cell adhesion proteins Epithelial (E-cad) and Neural (N-cad) cadherin is envisaged. We have previously reported the expression of E-cad and N-cad in human gametes and showed evidence of their involvement in sperm-oocyte adhesion events leading to fertilization. To overcome ethical limitations associated with the use of human gametes in fertilization-related studies, the mouse has been selected worldwide as the experimental model for over 4 decades. Herein, we report a detailed study aimed at characterizing the expression of E-cad and N-cad in murine gametes and their involvement in murine fertilization using specific antibodies and blocking peptides towards both adhesion proteins. E-cad and N-cad protein forms, as well as other members of the adhesion complex, specifically β-catenin and actin, were identified in spermatozoa, cumulus cells and oocytes protein extracts by means of Western immunoblotting. In addition, subcellular localization of these proteins was determined in whole cells using optical fluorescent microscopy. Gamete pre-incubation with anti-E-cad (ECCD-1) or N-cad (H-63) antibodies resulted in decreased (p < 0.05) In Vitro Fertilization (IVF) rates, when using both cumulus-oocytes complexes and cumulus-free oocytes. Moreover, IVF assays done with denuded oocytes and either antibodies or blocking peptides against E-cad and N-cad led to lower (p < 0.05) fertilization rates. When assessing each step, penetration of the cumulus mass was lower (p < 0.05) when spermatozoa were pre-incubated with ECCD-1 or blocking peptides towards E-cad or towards both E- and N-cad. Moreover, sperm-oolemma binding was impaired (p < 0.0005) after sperm pre-incubation with E-cad antibody or blocking peptide towards E-cad, N-cad or both proteins. Finally, sperm-oocyte fusion was lower (p < 0.05) after sperm pre-incubation with either antibody or blocking peptide against E-cad or N-cad. Our studies demonstrate the expression of members of the adherent complex in the murine model, and the use of antibodies and specific peptides revealed E-cad and N-cad participation in mammalian fertilization.
Collapse
|
5
|
Burton JJN, Luke AJ, Pepling ME. Regulation of mouse primordial follicle formation by signaling through the PI3K pathway. Biol Reprod 2021; 106:515-525. [PMID: 34725674 DOI: 10.1093/biolre/ioab204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 10/04/2021] [Accepted: 10/27/2021] [Indexed: 11/13/2022] Open
Abstract
Cell signaling mediated by the KIT receptor is critical for many aspects of oogenesis including the proliferation and migration of primordial germ cells, as well as the survival, growth, and maturation of ovarian follicles. We previously showed that KIT regulates cyst breakdown and primordial follicle formation, and in this study, have investigated the mechanisms downstream of the receptor by modulating the activity of two downstream signaling cascades: the phosphoinositide 3-kinase (PI3K) and the mitogen-activated protein kinase (MAPK) pathways. E17.5 ovaries were cultured for five days with a daily dose of media supplemented with either the PI3K inhibitor LY294002, the MEK inhibitor U0126, or a DMSO vehicle control. Our histological observations aligned with the established role of PI3K in oocyte growth and primordial follicle activation but also revealed that LY294002 treatment delayed the processes of cyst breakdown and primordial follicle formation. U0126 treatment also led to a reduction in oocyte growth and follicle development but did not appear to affect cyst breakdown. The delay in cyst breakdown was mitigated when ovaries were dually dosed with LY294002 and KITL, suggesting that while KIT may signal through PI3K to promote cyst breakdown, other signaling networks downstream of the receptor could compensate. These observations unearth a role for PI3K signaling in the establishment of the ovarian reserve and suggest that PI3K might be the primary mediator of KIT-induced cyst breakdown and primordial follicle formation in the mouse ovary.
Collapse
Affiliation(s)
| | - Amanda J Luke
- Department of Biology, Syracuse University, Syracuse, New York
| | | |
Collapse
|
6
|
Słowińska M, Paukszto Ł, Pardyak L, Jastrzębski JP, Liszewska E, Wiśniewska J, Kozłowski K, Jankowski J, Bilińska B, Ciereszko A. Transcriptome and Proteome Analysis Revealed Key Pathways Regulating Final Stage of Oocyte Maturation of the Turkey ( Meleagris gallopavo). Int J Mol Sci 2021; 22:ijms221910589. [PMID: 34638931 PMCID: PMC8508634 DOI: 10.3390/ijms221910589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 09/22/2021] [Accepted: 09/27/2021] [Indexed: 11/18/2022] Open
Abstract
In birds, the zona pellucida (ZP) matrix that surrounds the ovulated oocyte—called the inner perivitelline layer—is involved in sperm–zona interaction and successful fertilization. To identify the important genes and proteins connected with the final step of egg development, next-generation sequencing and two-dimensional electrophoresis, combined with mass spectrometry, were used for the analysis of mature oocytes at the F1 developmental stage. A total of 8161 genes and 228 proteins were annotated. Six subfamilies of genes, with codes ZP, ZP1–4, ZPD, and ZPAX, were identified, with the dominant expression of ZPD. The main expression site for ZP1 was the liver; however, granulosa cells may also participate in local ZP1 secretion. A ubiquitination system was identified in mature oocytes, where ZP1 was found to be the main ubiquitinated protein. Analysis of transcripts classified in estrogen receptor (ESR) signaling indicated the presence of ESR1 and ESR2, as well as a set of estrogen-dependent genes involved in both genomic and nongenomic mechanisms for the regulation of gene expression by estrogen. Oxidative phosphorylation was found to be a possible source of adenosine triphosphate, and the nuclear factor erythroid 2-related factor 2 signaling pathway could be involved in the response against oxidative stress. Oocyte–granulosa cell communication by tight, adherens, and gap junctions seems to be essential for the final step of oocyte maturation.
Collapse
Affiliation(s)
- Mariola Słowińska
- Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences in Olsztyn, 10-748 Olsztyn, Poland; (E.L.); (A.C.)
- Correspondence: ; Tel.: +48-89-539-3173
| | - Łukasz Paukszto
- Department of Plant Physiology, Genetics and Biotechnology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland; (Ł.P.); (J.P.J.)
| | - Laura Pardyak
- Center of Experimental and Innovative Medicine, University of Agriculture in Krakow, 30-248 Kraków, Poland;
| | - Jan P. Jastrzębski
- Department of Plant Physiology, Genetics and Biotechnology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland; (Ł.P.); (J.P.J.)
| | - Ewa Liszewska
- Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences in Olsztyn, 10-748 Olsztyn, Poland; (E.L.); (A.C.)
| | - Joanna Wiśniewska
- Department of Biological Function of Food, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences in Olsztyn, 10-748 Olsztyn, Poland;
| | - Krzysztof Kozłowski
- Department of Poultry Science, Faculty of Animal Bioengineering, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland; (K.K.); (J.J.)
| | - Jan Jankowski
- Department of Poultry Science, Faculty of Animal Bioengineering, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland; (K.K.); (J.J.)
| | - Barbara Bilińska
- Department of Endocrinology, Institute of Zoology, Jagiellonian University, 30-387 Kraków, Poland;
| | - Andrzej Ciereszko
- Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences in Olsztyn, 10-748 Olsztyn, Poland; (E.L.); (A.C.)
| |
Collapse
|
7
|
Abbassi L, El-Hayek S, Carvalho KF, Wang W, Yang Q, Granados-Aparici S, Mondadori R, Bordignon V, Clarke HJ. Epidermal growth factor receptor signaling uncouples germ cells from the somatic follicular compartment at ovulation. Nat Commun 2021; 12:1438. [PMID: 33664246 PMCID: PMC7933413 DOI: 10.1038/s41467-021-21644-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 02/07/2021] [Indexed: 01/31/2023] Open
Abstract
Germ cells are physically coupled to somatic support cells of the gonad during differentiation, but this coupling must be disrupted when they are mature, freeing them to participate in fertilization. In mammalian females, coupling occurs via specialized filopodia that project from the ovarian follicular granulosa cells to the oocyte. Here, we show that signaling through the epidermal growth factor receptor (EGFR) in the granulosa, which becomes activated at ovulation, uncouples the germ and somatic cells by triggering a massive and temporally synchronized retraction of the filopodia. Although EGFR signaling triggers meiotic maturation of the oocyte, filopodial retraction is independent of the germ cell state, being regulated solely within the somatic compartment, where it requires ERK-dependent calpain-mediated loss of filopodia-oocyte adhesion followed by Arp2/3-mediated filopodial shortening. By uncovering the mechanism regulating germ-soma uncoupling at ovulation, our results open a path to improving oocyte quality in human and animal reproduction.
Collapse
Affiliation(s)
- Laleh Abbassi
- Research Institute of the McGill University Health Centre, Montreal, Canada
- Division of Experimental Medicine, McGill University, Montreal, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Stephany El-Hayek
- Research Institute of the McGill University Health Centre, Montreal, Canada
- Department of Biology, McGill University, Montreal, Canada
- Centre for Arab Genomic Studies, Dubai, United Arab Emirates
| | - Karen Freire Carvalho
- Research Institute of the McGill University Health Centre, Montreal, Canada
- Division of Experimental Medicine, McGill University, Montreal, Canada
| | - Wusu Wang
- Research Institute of the McGill University Health Centre, Montreal, Canada
- College of Animal Science and Technology, Northwest A&F University, Shaanxi, PR China
| | - Qin Yang
- Research Institute of the McGill University Health Centre, Montreal, Canada
| | | | - Rafael Mondadori
- Department of Animal Science, McGill University, Montreal, Canada
| | - Vilceu Bordignon
- Department of Animal Science, McGill University, Montreal, Canada
| | - Hugh J Clarke
- Research Institute of the McGill University Health Centre, Montreal, Canada.
- Division of Experimental Medicine, McGill University, Montreal, Canada.
- Department of Biology, McGill University, Montreal, Canada.
- Department of Obstetrics and Gynecology, McGill University, Montreal, Canada.
| |
Collapse
|
8
|
Piprek RP, Kloc M, Mizia P, Kubiak JZ. The Central Role of Cadherins in Gonad Development, Reproduction, and Fertility. Int J Mol Sci 2020; 21:E8264. [PMID: 33158211 PMCID: PMC7663743 DOI: 10.3390/ijms21218264] [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] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/25/2020] [Accepted: 10/26/2020] [Indexed: 12/15/2022] Open
Abstract
Cadherins are a group of membrane proteins responsible for cell adhesion. They are crucial for cell sorting and recognition during the morphogenesis, but they also play many other roles such as assuring tissue integrity and resistance to stretching, mechanotransduction, cell signaling, regulation of cell proliferation, apoptosis, survival, carcinogenesis, etc. Within the cadherin superfamily, E- and N-cadherin have been especially well studied. They are involved in many aspects of sexual development and reproduction, such as germline development and gametogenesis, gonad development and functioning, and fertilization. E-cadherin is expressed in the primordial germ cells (PGCs) and also participates in PGC migration to the developing gonads where they become enclosed by the N-cadherin-expressing somatic cells. The differential expression of cadherins is also responsible for the establishment of the testis or ovary structure. In the adult testes, N-cadherin is responsible for the integrity of the seminiferous epithelium, regulation of sperm production, and the establishment of the blood-testis barrier. Sex hormones regulate the expression and turnover of N-cadherin influencing the course of spermatogenesis. In the adult ovaries, E- and N-cadherin assure the integrity of ovarian follicles and the formation of corpora lutea. Cadherins are expressed in the mature gametes and facilitate the capacitation of sperm in the female reproductive tract and gamete contact during fertilization. The germ cells and accompanying somatic cells express a series of different cadherins; however, their role in gonads and reproduction is still unknown. In this review, we show what is known and unknown about the role of cadherins in the germline and gonad development, and we suggest topics for future research.
Collapse
Affiliation(s)
- Rafał P. Piprek
- Department of Comparative Anatomy, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387 Krakow, Poland;
| | - Malgorzata Kloc
- The Houston Methodist Research Institute, Houston, TX 77030, USA;
- Department of Surgery, The Houston Methodist Hospital, Houston, TX 77030, USA
- MD Anderson Cancer Center, University of Texas, Houston, TX 77030, USA
| | - Paulina Mizia
- Department of Comparative Anatomy, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387 Krakow, Poland;
| | - Jacek Z. Kubiak
- Cycle Group, Institute of Genetics and Development of Rennes, Faculty of Medicine, UnivRennes, UMR 6290 CNRS/UR1, F-35000 Rennes, France
- Department of Regenerative Medicine and Cell Biology, Military Institute of Hygiene and Epidemiology (WIHE), 01-163 Warsaw, Poland
| |
Collapse
|
9
|
He C, Lv X, Huang C, Hua G, Ma B, Chen X, Angeletti PC, Dong J, Zhou J, Wang Z, Rueda BR, Davis JS, Wang C. YAP1-LATS2 feedback loop dictates senescent or malignant cell fate to maintain tissue homeostasis. EMBO Rep 2019; 20:e44948. [PMID: 30755404 PMCID: PMC6399607 DOI: 10.15252/embr.201744948] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 01/09/2019] [Accepted: 01/10/2019] [Indexed: 12/16/2022] Open
Abstract
Dysfunction of the homeostasis-maintaining systems in specific cell types or tissues renders the organism susceptible to a range of diseases, including cancers. One of the emerging mechanisms for maintaining tissue homeostasis is cellular senescence. Here, we report that the Hippo pathway plays a critical role in controlling the fate of ovarian cells. Hyperactivation of Yes-associated protein 1 (YAP1), the major effector of the Hippo pathway, induces senescence in cultured primary human ovarian surface epithelial cells (hOSEs). Large tumor suppressor 2 (LATS2), the primary upstream negative regulator of YAP1, is elevated in both YAP1-induced and natural replicative-triggered senescence. Deletion of LATS2 in hOSEs prevents these cells from natural replicative and YAP1-induced senescence. Most importantly, loss of LATS2 switches ovarian cells from YAP-induced senescence to malignant transformation. Our results demonstrate that LATS2 and YAP1, two major components of the Hippo/YAP signaling pathway, form a negative feedback loop to control YAP1 activity and prevent ovarian cells from malignant transformation. Human cancer genomic data extracted from TCGA datasets further confirm the clinical relevance of our finding.
Collapse
Affiliation(s)
- Chunbo He
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA, USA
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
- Olson Center for Women's Health, Department of Obstetrics & Gynecology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Xiangmin Lv
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA, USA
- Olson Center for Women's Health, Department of Obstetrics & Gynecology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Cong Huang
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA, USA
- Olson Center for Women's Health, Department of Obstetrics & Gynecology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Guohua Hua
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Bowen Ma
- Olson Center for Women's Health, Department of Obstetrics & Gynecology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Xingcheng Chen
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Peter C Angeletti
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Jixin Dong
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jin Zhou
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
- Department of Obstetrics and gynecology, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Zhengfeng Wang
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Bo R Rueda
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA, USA
| | - John S Davis
- Olson Center for Women's Health, Department of Obstetrics & Gynecology, University of Nebraska Medical Center, Omaha, NE, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
- Omaha Veterans Affairs Medical Center, Omaha, NE, USA
| | - Cheng Wang
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA, USA
- Olson Center for Women's Health, Department of Obstetrics & Gynecology, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
10
|
Yan H, Wen J, Zhang T, Zheng W, He M, Huang K, Guo Q, Chen Q, Yang Y, Deng G, Xu J, Wei Z, Zhang H, Xia G, Wang C. Oocyte-derived E-cadherin acts as a multiple functional factor maintaining the primordial follicle pool in mice. Cell Death Dis 2019; 10:160. [PMID: 30770786 PMCID: PMC6377673 DOI: 10.1038/s41419-018-1208-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 10/31/2018] [Accepted: 11/05/2018] [Indexed: 01/06/2023]
Abstract
In mammals, female fecundity is determined by the size of the primordial follicle (PF) pool, which is established during the perinatal period. As a non-renewable resource, the preservation of dormant PFs is crucial for sustaining female reproduction throughout life. Although studies have revealed that several oocyte-derived functional genes and pathways, such as newborn ovary homeobox (NOBOX) and 3-phosphoinositide-dependent protein kinase-1, participate in maintaining the PF pool, our understanding of the underlying molecular mechanisms is still incomplete. Here, we demonstrate that E-cadherin (E-cad) plays a crucial role in the maintenance of PFs in mice. E-cad is specifically localized to the cytomembrane of oocytes in PFs. Knockdown of E-cad in neonatal ovaries resulted in significant PF loss owing to oocyte apoptosis. In addition, the expression pattern of NOBOX is similar to that of E-cad. Knockdown of E-cad resulted in a decreased NOBOX level, whereas overexpression of Nobox partially rescued the follicle loss induced by silencing E-cad. Furthermore, E-cad governed NOBOX expression by regulating the shuttle protein, β-catenin, which acts as a transcriptional co-activator. Notably, E-cad, which is a transmembrane protein expressed in the oocytes, was also responsible for maintaining the PF structure by facilitating cell–cell adhesive contacts with surrounding pregranulosa cells. In conclusion, E-cad in oocytes of PFs plays an indispensable role in the maintenance of the PF pool by facilitating follicular structural stability and regulating NOBOX expression. These findings shed light on the physiology of sustaining female reproduction.
Collapse
Affiliation(s)
- Hao Yan
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, College of Life Science, NingXia University, Yinchuan, Ningxia, 750021, China.,State Key Laboratory of Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing, 100193, China
| | - Jia Wen
- State Key Laboratory of Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing, 100193, China
| | - Tuo Zhang
- State Key Laboratory of Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing, 100193, China
| | - Wenying Zheng
- State Key Laboratory of Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing, 100193, China
| | - Meina He
- State Key Laboratory of Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing, 100193, China
| | - Kun Huang
- State Key Laboratory of Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing, 100193, China
| | - Qirui Guo
- State Key Laboratory of Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing, 100193, China
| | - Qian Chen
- State Key Laboratory of Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing, 100193, China
| | - Yi Yang
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, College of Life Science, NingXia University, Yinchuan, Ningxia, 750021, China
| | - Guangcun Deng
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, College of Life Science, NingXia University, Yinchuan, Ningxia, 750021, China
| | - Jinrui Xu
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, College of Life Science, NingXia University, Yinchuan, Ningxia, 750021, China
| | - Zhiqing Wei
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, College of Life Science, NingXia University, Yinchuan, Ningxia, 750021, China
| | - Hua Zhang
- State Key Laboratory of Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing, 100193, China.
| | - Guoliang Xia
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, College of Life Science, NingXia University, Yinchuan, Ningxia, 750021, China. .,State Key Laboratory of Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing, 100193, China.
| | - Chao Wang
- State Key Laboratory of Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
11
|
Maternal β-hydroxy-β-methylbutyrate (HMB) supplementation during pregnancy affects early folliculogenesis in the ovary of newborn piglets. Theriogenology 2019; 128:91-100. [PMID: 30743108 DOI: 10.1016/j.theriogenology.2019.02.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 01/15/2019] [Accepted: 02/01/2019] [Indexed: 12/22/2022]
Abstract
Beta-hydroxy-beta-methylbutyrate (HMB) is a leucine metabolite with protein anabolic effects. This study was designed to determine whether prenatal HMB treatment has an effect on oogenesis and folliculogenesis in the ovary of newborn piglets. HMB decreased the number of egg nests and primordial follicles and increased the pool of developing follicles compared to the control group. Although the percentage of TUNEL-positive oocytes within the egg nests was higher in HMB-treated group no increase in the Bax/Bcl-2 ratio and active caspase-3 expression was observed. Moreover, the granulosa cell proliferation index and StAR protein expression were higher in HMB-treated group. In contrast to the control group, the expression of E-cadherins was reduced after the HMB treatment. In addition, a significant increase in the serum level of gonadotropins and steroid hormones was detected in HMB-treated piglets. In conclusion, prenatal HMB treatment dysregulates hormonal homeostasis which impairs early folliculogenesis in piglets.
Collapse
|
12
|
Guo C, Liu G, Zhao D, Mi Y, Zhang C, Li J. Interaction of Follicle-Stimulating Hormone and Stem Cell Factor to Promote Primordial Follicle Assembly in the Chicken. Front Endocrinol (Lausanne) 2019; 10:91. [PMID: 30837955 PMCID: PMC6389603 DOI: 10.3389/fendo.2019.00091] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 01/31/2019] [Indexed: 12/12/2022] Open
Abstract
Follicle-stimulating hormone (FSH) and KIT signaling are required for ovarian development. In this study the interactive effect of FSH and stem cell factor (SCF) on folliculogenesis was investigated in the chicken. Correlated changes between the FSH receptor and the expression of KIT signaling genes were seen to be involved in the formation of the chicken primordial follicles. Follicle-stimulating hormone and SCF displayed a reciprocal stimulating effect in the promotion of folliculogenesis involving elevated phosphorylation of mitogen-activated protein kinases (MAPK) and protein kinase B (AKT) proteins. Knockdown of c-KIT or SCF reduced the stimulatory effect of FSH on KIT signaling as well as upon MAPK and AKT phosphorylation. Treatment of FSH and SCF in combination enhanced ovarian cell proliferation and N-cadherin expression, but inhibited cell apoptosis and E-cadherin expression. Overall, the reciprocal stimulating effect of FSH and SCF in promoting chicken follicle assembly involving accelerated ovarian cell proliferation, N-cadherin expression, inhibited cell apoptosis, and E-cadherin expression is demonstrated.
Collapse
|
13
|
Genes responsible for proliferation, differentiation, and junction adhesion are significantly up-regulated in human ovarian granulosa cells during a long-term primary in vitro culture. Histochem Cell Biol 2018; 151:125-143. [PMID: 30382374 PMCID: PMC6394675 DOI: 10.1007/s00418-018-1750-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2018] [Indexed: 01/10/2023]
Abstract
The human ovarian granulosa cells (GCs) surround the oocyte and form the proper architecture of the ovarian follicle. The ability of GCs to proliferate and differentiate in the conditions of in vitro culture has been proven. However, there is still a large field for extensive investigation of molecular basics, as well as marker genes, responsible for these processes. This study aimed to find the new marker genes, encoding proteins that regulate human GCs in vitro capability for proliferation and differentiation during long-term primary culture. The human follicular GCs were collected from hyper-stimulated ovarian follicles during IVF procedures and transferred to a long-term in vitro culture. The culture lasted for 30 days, with RNA samples isolated at days 1, 7, 15, 30. Transcriptomic analysis was then performed with the use of Affymetrix microarray. Obtained results were then subjected to bioinformatical evaluation and sorting. After subjecting the datasets to KEGG analysis, three differentially expressed ontology groups "cell differentiation" (GO:0030154), "cell proliferation" (GO:0008283) and "cell-cell junction organization" (GO:0045216) were chosen for further investigation. All three of those ontology groups are involved in human GCs' in vitro lifespan, proliferation potential, and survival capability. Changes in expression of genes of interest belonging to the chosen GOs were validated with the use of RT-qPCR. In this manuscript, we suggest that VCL, PARVA, FZD2, NCS1, and COL5A1 may be recognized as new markers of GC in vitro differentiation, while KAT2B may be a new marker of their proliferation. Additionally, SKI, GLI2, FERMT2, and CDH2 could also be involved in GC in vitro proliferation and differentiation processes. We demonstrated that, in long-term in vitro culture, GCs exhibit markers that suggest their ability to differentiate into different cells types. Therefore, the higher expression profile of these genes may also be associated with the induction of cellular differentiation processes that take place beyond the long-term primary in vitro culture.
Collapse
|
14
|
Significant Down-Regulation of "Biological Adhesion" Genes in Porcine Oocytes after IVM. Int J Mol Sci 2017; 18:ijms18122685. [PMID: 29232894 PMCID: PMC5751287 DOI: 10.3390/ijms18122685] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 11/10/2017] [Accepted: 12/07/2017] [Indexed: 01/06/2023] Open
Abstract
Proper maturation of the mammalian oocyte is a compound processes determining successful monospermic fertilization, however the number of fully mature porcine oocytes is still unsatisfactory. Since oocytes’ maturation and fertilization involve cellular adhesion and membranous contact, the aim was to investigate cell adhesion ontology group in porcine oocytes. The oocytes were collected from ovaries of 45 pubertal crossbred Landrace gilts and subjected to two BCB tests. After the first test, only granulosa cell-free BCB+ oocytes were directly exposed to microarray assays and RT-qPCR (“before IVM” group), or first in vitro matured and then if classified as BCB+ passed to molecular analyses (“after IVM” group). As a result, we have discovered substantial down-regulation of genes involved in adhesion processes, such as: organization of actin cytoskeleton, migration, proliferation, differentiation, apoptosis, survival or angiogenesis in porcine oocytes after IVM, compared to oocytes analyzed before IVM. In conclusion, we found that biological adhesion may be recognized as the process involved in porcine oocytes’ successful IVM. Down-regulation of genes included in this ontology group in immature oocytes after IVM points to their unique function in oocyte’s achievement of fully mature stages. Thus, results indicated new molecular markers involved in porcine oocyte IVM, displaying essential roles in biological adhesion processes.
Collapse
|
15
|
Wang C, Zhou B, Xia G. Mechanisms controlling germline cyst breakdown and primordial follicle formation. Cell Mol Life Sci 2017; 74:2547-2566. [PMID: 28197668 PMCID: PMC11107689 DOI: 10.1007/s00018-017-2480-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 01/20/2017] [Accepted: 01/30/2017] [Indexed: 12/11/2022]
Abstract
In fetal females, oogonia proliferate immediately after sex determination. The progress of mitosis in oogonia proceeds so rapidly that the incompletely divided cytoplasm of the sister cells forms cysts. The oogonia will then initiate meiosis and arrest at the diplotene stage of meiosis I, becoming oocytes. Within each germline cyst, oocytes with Balbiani bodies will survive after cyst breakdown (CBD). After CBD, each oocyte is enclosed by pre-granulosa cells to form a primordial follicle (PF). Notably, the PF pool formed perinatally will be the sole lifelong oocyte source of a female. Thus, elucidating the mechanisms of CBD and PF formation is not only meaningful for solving mysteries related to ovarian development but also contributes to the preservation of reproduction. However, the mechanisms that regulate these phenomena are largely unknown. This review summarizes the progress of cellular and molecular research on these processes in mice and humans.
Collapse
Affiliation(s)
- Chao Wang
- State Key Laboratory for Agro-Biotechnology, College of Biological Science, China Agricultural University, Beijing, 100193, China
| | - Bo Zhou
- State Key Laboratory for Agro-Biotechnology, College of Biological Science, China Agricultural University, Beijing, 100193, China
| | - Guoliang Xia
- State Key Laboratory for Agro-Biotechnology, College of Biological Science, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
16
|
Zhao D, Lv C, Liu G, Mi Y, Zhang C. Effect of estrogen on chick primordial follicle development and activation. Cell Biol Int 2017; 41:630-638. [PMID: 28328180 DOI: 10.1002/cbin.10766] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 03/19/2017] [Indexed: 11/12/2022]
Abstract
Steroid hormones and their receptors play pivotal roles in avian reproduction and development. 17β-estradiol (E2 ) is an essential mediator in ovarian development. In this study, Day 3 (D3) chicks were injected with E2 (0.025, 0.25 or 2.5 mg/kg body weight) for 4 days to assess the effects of estrogen on avian follicle development. Morphological analysis showed that treatment of E2 at 2.5 mg/kg increased the number of growing follicles by 28. 5% and activated follicles by 8.9%, compared with the group. Treatment of E2 also led to increased diameter and area of three kinds of follicles and oocytes, respectively. However, this stimulating effect of E2 was inhibited by combined treatment of the estrogen receptor antagonist tamoxifen, resulting in decreased number of the growing follicles by 14.3% and the activated follicles by 5.8%, respectively. Immunohistochemical staining of the proliferating cell nuclear antigen (PCNA) revealed that the ratios of the PCNA-positive granulosa and interstitial cells were increased by 14.0% and 21.5%, respectively. Western blot analysis of PCNA confirmed this stimulating effect of E2 . Expression of ERα mRNAs was elevated by administration of E2 in vivo and in vitro. Furthermore, E- and N-cadherin displayed increased expression after E2 treatment in vivo and in vitro. In conclusion, E2 can promote chicken primordial follicle development and activation involving its increased receptor and cadherin production at the early stage of ovarian development.
Collapse
Affiliation(s)
- Dan Zhao
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Caifeng Lv
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Guang Liu
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yuling Mi
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Caiqiao Zhang
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| |
Collapse
|
17
|
Li F, Miao X, Chen Y, Curry TE. CXADR-like membrane protein (CLMP) in the rat ovary: stimulation by human chorionic gonadotrophin during the periovulatory period. Reprod Fertil Dev 2017; 28:742-9. [PMID: 25400132 DOI: 10.1071/rd14201] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 09/17/2014] [Indexed: 01/20/2023] Open
Abstract
CXADR-like membrane protein (CLMP) is a novel cell-cell adhesion molecule. The present study investigated the spatiotemporal expression pattern of CLMP and its regulation in the rat ovary during the periovulatory period. Real-time polymerase chain reaction analysis revealed that Clmp mRNA was rapidly stimulated in intact ovaries by 4h after human chorionic gonadotrophin (hCG) treatment. In situ hybridisation analysis demonstrated that Clmp mRNA expression was stimulated in theca cells at 4h after hCG and remained elevated until 12h. Clmp mRNA was also upregulated in granulosa cells and was present in forming corpora lutea. Our data indicate that the protein kinase A but not the protein kinase C pathway regulates the expression of Clmp mRNA in granulosa cells. Phosphatidylinositol 3 kinase and p38 kinase are also involved in regulating Clmp mRNA expression. The stimulation of Clmp mRNA by hCG requires new protein synthesis. Furthermore, inhibition of epidermal growth factor receptor activation significantly inhibited Clmp mRNA expression, whereas inhibition of prostaglandin synthesis or progesterone action had no effect. The stimulation of CLMP in the rat ovary may be important in cell adhesion events during ovulation and luteal formation such as maintaining the structure and communication of ovarian follicular and luteal cells.
Collapse
Affiliation(s)
- Feixue Li
- Zhejiang Key Laboratory of Organ Development and Regeneration, Institute of Developmental and Regenerative Biology, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 310036, People's Republic of China
| | - Xiaoping Miao
- Zhejiang Key Laboratory of Organ Development and Regeneration, Institute of Developmental and Regenerative Biology, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 310036, People's Republic of China
| | - Yonglong Chen
- Zhejiang Key Laboratory of Organ Development and Regeneration, Institute of Developmental and Regenerative Biology, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 310036, People's Republic of China
| | - Thomas E Curry
- Department of Obstetrics and Gynecology, Chandler Medical Center, 800 Rose Street, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
18
|
GGPP-Mediated Protein Geranylgeranylation in Oocyte Is Essential for the Establishment of Oocyte-Granulosa Cell Communication and Primary-Secondary Follicle Transition in Mouse Ovary. PLoS Genet 2017; 13:e1006535. [PMID: 28072828 PMCID: PMC5224981 DOI: 10.1371/journal.pgen.1006535] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 12/13/2016] [Indexed: 12/24/2022] Open
Abstract
Folliculogenesis is a progressive and highly regulated process, which is essential to provide ova for later reproductive life, requires the bidirectional communication between the oocyte and granulosa cells. This physical connection-mediated communication conveys not only the signals from the oocyte to granulosa cells that regulate their proliferation but also metabolites from the granulosa cells to the oocyte for biosynthesis. However, the underlying mechanism of establishing this communication is largely unknown. Here, we report that oocyte geranylgeranyl diphosphate (GGPP), a metabolic intermediate involved in protein geranylgeranylation, is required to establish the oocyte-granulosa cell communication. GGPP and geranylgeranyl diphosphate synthase (Ggpps) levels in oocytes increased during early follicular development. The selective depletion of GGPP in mouse oocytes impaired the proliferation of granulosa cells, primary-secondary follicle transition and female fertility. Mechanistically, GGPP depletion inhibited Rho GTPase geranylgeranylation and its GTPase activity, which was responsible for the accumulation of cell junction proteins in the oocyte cytoplasm and the failure to maintain physical connection between oocyte and granulosa cells. GGPP ablation also blocked Rab27a geranylgeranylation, which might account for the impaired secretion of oocyte materials such as Gdf9. Moreover, GGPP administration restored the defects in oocyte-granulosa cell contact, granulosa cell proliferation and primary-secondary follicle transition in Ggpps depletion mice. Our study provides the evidence that GGPP-mediated protein geranylgeranylation contributes to the establishment of oocyte-granulosa cell communication and then regulates the primary-secondary follicle transition, a key phase of folliculogenesis essential for female reproductive function. Folliculogenesis is a progressive and highly regulated process that requires the tight coordination of metabolism and bidirectional communication between the oocyte and granulosa cells. How this communication is established remains unclear. Here, we find that GGPP-mediated protein geranylgeranylation, a post-translational modification, is essential for the oocyte-granulosa cell communication. GGPP depletion in oocytes inhibits Rho GTPase geranylgeranylation-regulated cell adhesion and impairs Rab GTPase geranylgeranylation-directed cell secretion, which are responsible for the failure to maintain oocyte-granulosa cell communication. This communication defect is probably not able to support the proliferation of granulosa cells from one layer to multiple layers and ultimately results in the failure of the primary-secondary follicle transition and female subfertility. Our findings provide the evidence of GGPP-mediated protein geranylgeranylation involving in regulating primary-secondary follicle transition and establish a novel link between folliculogenesis and GGPP-regulated membrane dynamics.
Collapse
|
19
|
York JP, Ren YA, Zeng J, Bin Zhang, Wang F, Chen R, Liu J, Xia X, Zhang P. Growth Arrest Specific 2 (GAS2) is a Critical Mediator of Germ Cell Cyst Breakdown and Folliculogenesis in Mice. Sci Rep 2016; 6:34956. [PMID: 27734842 PMCID: PMC5062118 DOI: 10.1038/srep34956] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 09/21/2016] [Indexed: 01/25/2023] Open
Abstract
In the mouse ovary, the primordial follicle pool is established through a diverse array of signaling pathways and tissue remodeling events. Growth arrest specific gene two (GAS2) is a highly conserved cytoskeleton-associated protein whose in vivo function remains unclear. In Drosophila, loss of the GAS2 homolog, Pigs, results in infertility. We demonstrate herein that, in the mouse ovary, GAS2 is expressed in the stromal cells surrounding the oocyte cysts on 16.5 dpc, and in stromal cells surrounding growing follicles during juvenile and adult life. We have generated genetically engineered mice with inactivated Gas2. Gas2 homozygous mutant mice are viable but have severely impaired fertility in females, in which oocyte cyst breakdown is disrupted and follicle growth is impaired, with significantly reduced numbers of large antral follicles and corpora lutea. In these mutant mice, the organization of the basal lamina surrounding developing follicles is severely defective at multiple stages of folliculogenesis. We also found that Notch signaling activity was altered in ovaries from Gas2 null mice around the time of birth and during follicular development later in life. These results indicate that GAS2 is a critical and novel regulator of tissue remodeling in the ovary during oocyte cyst breakdown and folliculogenesis.
Collapse
Affiliation(s)
- J Philippe York
- Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX 77030, USA.,Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yi Athena Ren
- Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jie Zeng
- The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510150, China
| | - Bin Zhang
- Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX 77030, USA
| | - Fang Wang
- The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510150, China
| | - Rui Chen
- The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510150, China
| | - Jianqiao Liu
- The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510150, China
| | - Xuefeng Xia
- The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510150, China.,Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX 77030, USA.,Biochemistry and Molecular Biology, Baylor College of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Pumin Zhang
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA.,Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA.,Biochemistry and Molecular Biology, Baylor College of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| |
Collapse
|
20
|
Importância das comunicações intercelulares para o desenvolvimento de folículos ovarianos. ACTA ACUST UNITED AC 2016. [DOI: 10.1016/j.recli.2015.12.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
21
|
Niu W, Wang Y, Wang Z, Xin Q, Wang Y, Feng L, Zhao L, Wen J, Zhang H, Wang C, Xia G. JNK signaling regulates E-cadherin junctions in germline cysts and determines primordial follicle formation in mice. Development 2016; 143:1778-87. [PMID: 27013242 DOI: 10.1242/dev.132175] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Accepted: 03/11/2016] [Indexed: 12/23/2022]
Abstract
Physiologically, the size of the primordial follicle pool determines the reproductive lifespan of female mammals, while its establishment largely depends on a process of germline cyst breakdown during the perinatal period. The mechanisms regulating this process are poorly understood. Here we demonstrate that c-Jun amino-terminal kinase (JNK) signaling is crucial for germline cyst breakdown and primordial follicle formation. JNK was specifically localized in oocytes and its activity increased as germline cyst breakdown progressed. Importantly, disruption of JNK signaling with a specific inhibitor (SP600125) or knockdown technology (Lenti-JNK-shRNAs) resulted in significantly suppressed cyst breakdown and primordial follicle formation in cultured mouse ovaries. Our results show that E-cadherin is intensely expressed in germline cysts, and that its decline is necessary for oocyte release from the cyst. However, inhibition of JNK signaling leads to aberrantly enhanced localization of E-cadherin at oocyte-oocyte contact sites. WNT4 expression is upregulated after SP600125 treatment. Additionally, similar to the effect of SP600125 treatment, WNT4 overexpression delays cyst breakdown and is accompanied by abnormal E-cadherin expression patterns. In conclusion, our results suggest that JNK signaling, which is inversely correlated with WNT4, plays an important role in perinatal germline cyst breakdown and primordial follicle formation by regulating E-cadherin junctions between oocytes in mouse ovaries.
Collapse
Affiliation(s)
- Wanbao Niu
- State Key Laboratory of Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing 100193, China
| | - Ye Wang
- State Key Laboratory of Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing 100193, China
| | - Zhengpin Wang
- Laboratory of Cellular and Developmental Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Qiliang Xin
- State Key Laboratory of Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing 100193, China
| | - Yijing Wang
- National Institute of Biological Sciences, Zhongguancun Life Science Park, Changping, Beijing 102206, China
| | - Lizhao Feng
- State Key Laboratory of Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing 100193, China
| | - Lihua Zhao
- State Key Laboratory of Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing 100193, China
| | - Jia Wen
- State Key Laboratory of Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing 100193, China
| | - Hua Zhang
- State Key Laboratory of Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing 100193, China
| | - Chao Wang
- State Key Laboratory of Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing 100193, China
| | - Guoliang Xia
- State Key Laboratory of Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing 100193, China
| |
Collapse
|
22
|
Wang B, Zhao J, Zhang P. Gene signatures in osteoarthritic acetabular labrum using microarray analysis. Int J Rheum Dis 2016; 20:1927-1934. [PMID: 26833791 DOI: 10.1111/1756-185x.12810] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Osteoarthritis (OA) is the most common chronic joint disease. This study aimed to uncover underlying mechanisms of OA pathogenesis and explore the potential biomarkers of osteoarthritic acetabular labrum. METHODS The microarray data GSE60762 was utilized, containing five OA acetabular labrum samples and three healthy control samples. Data were preprocessed by oligo package and the differentially expressed genes (DEGs) were identified using limma package with predefined criteria, followed by functional enrichment analysis by the GoFunction in R Bioconductor, and protein-protein interaction (PPI) network analysis. RESULTS As a result, 141 DEGs (44 were up-regulated and 97 were down-regulated) were identified between OA and healthy acetabular labrum cells. Up-regulated genes including CDH2 and WNT5A were significantly enriched in intracellular signal transduction function, while down-regulated genes such as KDR, FLT1 and CDH5 were remarkably correlated with cardiovascular system development. FLT1, KDR, CDH2 and CDH5 were the striking nodes in the PPI network. CONCLUSION CDH2, WNT5A, KDR, FLT1 and CDH5 might serve as the biomarkers of OA prognosis. Intracellular signal transduction and cardiovascular system development might play significant roles in the destruction of labrum during OA progression. However, more experimental validations are warranted to confirm our findings.
Collapse
Affiliation(s)
- Beiyue Wang
- Department of Orthopedics, Jinling Hospital, Nanjing, China
| | - Jianning Zhao
- Department of Orthopedics, Jinling Hospital, Nanjing, China
| | - Peng Zhang
- Department of Orthopedics, Jiangsu Province Geriatric Institute, Nanjing, Jiangsu, China
| |
Collapse
|
23
|
Hua G, Lv X, He C, Remmenga SW, Rodabough KJ, Dong J, Yang L, Lele SM, Yang P, Zhou J, Karst A, Drapkin RI, Davis JS, Wang C. YAP induces high-grade serous carcinoma in fallopian tube secretory epithelial cells. Oncogene 2015; 35:2247-65. [PMID: 26364602 PMCID: PMC4791205 DOI: 10.1038/onc.2015.288] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 06/11/2015] [Accepted: 07/06/2015] [Indexed: 12/12/2022]
Abstract
Accumulating evidence indicates that ovarian high-grade serous carcinoma (HGSC) originates from fallopian tube secretory epithelial cells (FTSECs). However, the molecular mechanisms underlying the initiation and progression of HGSC derived from FTSECs remains unclear. In this study, we found that the Hippo/Yes-associated protein (YAP) signaling pathway has a critical role in the initiation and progression of fallopian tube and ovarian HGSC. Importantly, YAP was overexpressed in inflammatory and cancerous fallopian tube tissues. Further, overexpression of wild-type YAP, or constitutively active YAP in immortalized FTSECs, induced cell proliferation, migration, colony formation and tumorigenesis. Moreover, the Hippo/YAP and the fibroblast growth factor (FGF) signaling pathways formed an autocrine/paracrine-positive feedback loop to drive the progression of the FTSEC-derived HGSC. Evidence in this study strongly suggests that combined therapy with inhibitors of YAP (such as verteporfin) and FGF receptors (such as BGJ398) can provide a novel therapeutic strategy to treat fallopian tube and ovarian HGSC.
Collapse
Affiliation(s)
- G Hua
- Olson Center for Women's Health, Department of Obstetrics/Gynecology, University of Nebraska Medical Center, Omaha, NE, USA.,The Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - X Lv
- Olson Center for Women's Health, Department of Obstetrics/Gynecology, University of Nebraska Medical Center, Omaha, NE, USA
| | - C He
- Olson Center for Women's Health, Department of Obstetrics/Gynecology, University of Nebraska Medical Center, Omaha, NE, USA.,The Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - S W Remmenga
- Olson Center for Women's Health, Department of Obstetrics/Gynecology, University of Nebraska Medical Center, Omaha, NE, USA
| | - K J Rodabough
- Olson Center for Women's Health, Department of Obstetrics/Gynecology, University of Nebraska Medical Center, Omaha, NE, USA
| | - J Dong
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - L Yang
- The Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - S M Lele
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - P Yang
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - J Zhou
- Department of Obstetrics and Gynecology, Urumuqi General Hospital of Lanzhou Military Region, Urumuqi, China
| | - A Karst
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - R I Drapkin
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - J S Davis
- Olson Center for Women's Health, Department of Obstetrics/Gynecology, University of Nebraska Medical Center, Omaha, NE, USA.,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.,Omaha Veterans Affairs Medical Center, Omaha, NE, USA
| | - C Wang
- Olson Center for Women's Health, Department of Obstetrics/Gynecology, University of Nebraska Medical Center, Omaha, NE, USA.,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
24
|
Li J, Zhao D, Guo C, Li J, Mi Y, Zhang C. Involvement of Notch signaling in early chick ovarian follicle development. Cell Biol Int 2015; 40:65-73. [DOI: 10.1002/cbin.10538] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 08/15/2015] [Indexed: 11/11/2022]
Affiliation(s)
- Jun Li
- Department of Veterinary Medicine, College of Animal Sciences; Zhejiang University; No. 866 Yuhangtang Road Hangzhou 310058 People's Republic of China
| | - Dan Zhao
- Department of Veterinary Medicine, College of Animal Sciences; Zhejiang University; No. 866 Yuhangtang Road Hangzhou 310058 People's Republic of China
| | - Changquan Guo
- Department of Veterinary Medicine, College of Animal Sciences; Zhejiang University; No. 866 Yuhangtang Road Hangzhou 310058 People's Republic of China
| | - Jian Li
- Department of Veterinary Medicine, College of Animal Sciences; Zhejiang University; No. 866 Yuhangtang Road Hangzhou 310058 People's Republic of China
| | - Yuling Mi
- Department of Veterinary Medicine, College of Animal Sciences; Zhejiang University; No. 866 Yuhangtang Road Hangzhou 310058 People's Republic of China
| | - Caiqiao Zhang
- Department of Veterinary Medicine, College of Animal Sciences; Zhejiang University; No. 866 Yuhangtang Road Hangzhou 310058 People's Republic of China
| |
Collapse
|
25
|
Vazquez-Levin MH, Marín-Briggiler CI, Caballero JN, Veiga MF. Epithelial and neural cadherin expression in the mammalian reproductive tract and gametes and their participation in fertilization-related events. Dev Biol 2015; 401:2-16. [DOI: 10.1016/j.ydbio.2014.12.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Revised: 12/23/2014] [Accepted: 12/28/2014] [Indexed: 01/10/2023]
|
26
|
YAP forms autocrine loops with the ERBB pathway to regulate ovarian cancer initiation and progression. Oncogene 2015; 34:6040-54. [PMID: 25798835 PMCID: PMC4580488 DOI: 10.1038/onc.2015.52] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 11/21/2014] [Accepted: 11/25/2014] [Indexed: 12/22/2022]
Abstract
Mechanisms underlying ovarian cancer initiation and progression are unclear. Herein, we report that the Yes-associated protein (YAP), a major effector of the Hippo tumor suppressor pathway, interacts with ERBB signaling pathways to regulate the initiation and progression of ovarian cancer. Immunohistochemistry studies indicate that YAP expression is associated with poor clinical outcomes in patients. Overexpression or constitutive activation of YAP leads to transformation and tumorigenesis in human ovarian surface epithelial cells, and promotes growth of cancer cells in vivo and in vitro. YAP induces the expression of epidermal growth factor (EGF) receptors (EGFR, ERBB3) and production of EGF-like ligands (HBEGF, NRG1 and NRG2). HBEGF or NRG1, in turn, activates YAP and stimulates cancer cell growth. Knockdown of ERBB3 or HBEGF eliminates YAP effects on cell growth and transformation, whereas knockdown of YAP abrogates NRG1- and HBEGF-stimulated cell proliferation. Collectively, our study demonstrates the existence of HBEGF & NRGs/ERBBs/YAP/HBEGF & NRGs autocrine loop that controls ovarian cell tumorigenesis and cancer progression.
Collapse
|
27
|
Highly robust protein production by co-culture of CHO spheroids layered on feeder cells in serum-free medium. Colloid Polym Sci 2014. [DOI: 10.1007/s00396-013-3093-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
28
|
Tagler D, Makanji Y, Tu T, Bernabé BP, Lee R, Zhu J, Kniazeva E, Hornick JE, Woodruff TK, Shea LD. Promoting extracellular matrix remodeling via ascorbic acid enhances the survival of primary ovarian follicles encapsulated in alginate hydrogels. Biotechnol Bioeng 2014; 111:1417-29. [PMID: 24375265 DOI: 10.1002/bit.25181] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 12/18/2013] [Accepted: 12/23/2013] [Indexed: 01/12/2023]
Abstract
The in vitro growth of ovarian follicles is an emerging technology for fertility preservation. Various strategies support the culture of secondary and multilayer follicles from various species including mice, non-human primate, and human; however, the culture of early stage (primary and primordial) follicles, which are more abundant in the ovary and survive cryopreservation, has been limited. Hydrogel-encapsulating follicle culture systems that employed feeder cells, such as mouse embryonic fibroblasts (MEFs), stimulated the growth of primary follicles (70-80 µm); yet, survival was low and smaller follicles (<70 µm) rapidly lost structure and degenerated. These morphologic changes were associated with a breakdown of the follicular basement membrane; hence, this study investigated ascorbic acid based on its role in extracellular matrix (ECM) deposition/remodeling for other applications. The selection of ascorbic acid was further supported by a microarray analysis that suggested a decrease in mRNA levels of enzymes within the ascorbate pathway between primordial, primary, and secondary follicles. The supplementation of ascorbic acid (50 µg/mL) significantly enhanced the survival of primary follicles (<80 µm) cultured in alginate hydrogels, which coincided with improved structural integrity. Follicles developed antral cavities and increased to diameters exceeding 250 µm. Consistent with improved structural integrity, the gene/protein expression of ECM and cell adhesion molecules was significantly changed. This research supports the notion that modifying the culture environment (medium components) can substantially enhance the survival and growth of early stage follicles.
Collapse
Affiliation(s)
- David Tagler
- Department of Chemical and Biological Engineering, McCormick School of Engineering, Northwestern University, Technological Institute E-136, 2145 Sheridan Road, Evanston, Illinois, 60208; Institute for BioNanotechnology in Medicine (IBNAM), Northwestern University, 303 E. Superior Street, Suite 11-131, Chicago, Illinois, 60611
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Prunskaite-Hyyryläinen R, Shan J, Railo A, Heinonen KM, Miinalainen I, Yan W, Shen B, Perreault C, Vainio SJ. Wnt4, a pleiotropic signal for controlling cell polarity, basement membrane integrity, and antimüllerian hormone expression during oocyte maturation in the female follicle. FASEB J 2013; 28:1568-81. [PMID: 24371124 DOI: 10.1096/fj.13-233247] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Wnt4 is a key signal that channels the developmental fate of the indifferent mammalian gonad toward the ovary, but whether Wnt4 has later roles during ovary development remains unknown. To investigate this, we inactivated the Wnt4 gene by crossing Amhr2Cre and doxycycline-inducible Rosa(rtTA)-knock-in Cre mice with mice carrying a floxed Wnt4 allele and used a novel Wnt4(mCherry)-knock-in mouse. In these models, ovarian folliculogenesis was compromised, and female fertility was severely reduced, and Wnt4 deficiency eventually led to premature ovarian failure. These anomalies were associated with cell polarity defects in the follicle. Within the follicle, laminin and type IV collagen assembled ectopic basement membrane-like structures, the cell adherens junction components N-cadherin and β-catenin lost their polarized expression pattern, and expression of the gap junction protein connexin 43 was reduced by ~30% when compared with that of the controls. Besides these changes, expression of antimüllerian hormone (Amh) was inhibited in the absence of Wnt4 signaling in vivo. Consistent with this, Wnt4 signaling up-regulated Amh gene expression in KK1 cells in vitro. Thus, Wnt4 signaling is necessary during maturation of the ovarian follicles, where it coordinates expression of Amh, cell survival, and polarized organization of the follicular cells.
Collapse
Affiliation(s)
- Renata Prunskaite-Hyyryläinen
- 1Laboratory of Developmental Biology, Department of Medical Biochemistry and Molecular Biology, University of Oulu and Biocenter Oulu, Aapistie 5, P. O. Box 5000, FIN-90220, Oulu, Finland.
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Knapczyk-Stwora K, Durlej-Grzesiak M, Ciereszko RE, Koziorowski M, Slomczynska M. Antiandrogen flutamide affects folliculogenesis during fetal development in pigs. Reproduction 2013; 145:265-76. [PMID: 23580948 DOI: 10.1530/rep-12-0236] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Androgen deficiency during prenatal development may affect the expression of genes involved in the folliculogenesis regulation. In order to study the effect of antiandrogen on fetal ovarian development, pregnant gilts were injected with flutamide (for 7 days, 50 mg/kg bodyweight per day) or corn oil (control groups) starting on gestation days 43 (GD50), 83 (GD90), or 101 (GD108). The obtained fetal ovaries were fixed for histology and immunohistochemistry or frozen for real-time PCR. Morphological evaluation, TUNEL assay, and expression of selected factors (Ki-67, GATA binding transcription factor 4 (GATA4), E-Cadherin and tumor necrosis factor a (TNFa)) were performed. On GD90 and GD108, ovaries following flutamide administration showed a higher number of egg nests and lower number off ollicles than those in respective control groups. An increased mRNA and protein expression of Ki-67 was observed in flutamide-treated groups compared with controls on GD50 and GD108 but decreased expression was found on GD90. In comparison to control groups a higher percentage of TUNEL-positive cells was shown after flutamide exposure on GD50 and GD90 and a lower percentage of apoptotic cells was observed on GD108. These data were consistent with changes in TNF (TNFa) mRNA expression, which increased on GD90 and decreased on GD108. E-cadherin mRNA and protein expression was upregulated on GD50 and downregulated on GD90 and GD108. In conclusion diminished androgen action in porcine fetal ovaries during mid- and late gestation leads to changes in the expression of genes crucial for follicle formation. Consequently, delayed folliculogenesis was observed on GD90 and GD108. It seems however that androgens exhibit diverse biological effects depending on the gestational period.
Collapse
Affiliation(s)
- Katarzyna Knapczyk-Stwora
- Department of Endocrinology, Institute of Zoology, Jagiellonian University, Gronostajowa 9, 30-387 Krakow, Poland
| | | | | | | | | |
Collapse
|
31
|
Mora JM, Fenwick MA, Castle L, Baithun M, Ryder TA, Mobberley M, Carzaniga R, Franks S, Hardy K. Characterization and Significance of Adhesion and Junction-Related Proteins in Mouse Ovarian Follicles1. Biol Reprod 2012; 86:153, 1-14. [DOI: 10.1095/biolreprod.111.096156] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
32
|
Mukherjee A, Reisdorph N, Guda B, Pandey S, Roy SK. Changes in ovarian protein expression during primordial follicle formation in the hamster. Mol Cell Endocrinol 2012; 348:87-94. [PMID: 21821096 PMCID: PMC3418795 DOI: 10.1016/j.mce.2011.07.043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Revised: 07/19/2011] [Accepted: 07/20/2011] [Indexed: 01/01/2023]
Abstract
Although many proteins have been shown to affect the transition of primordial follicles to the primary stage, factors regulating the formation of primordial follicles remains sketchy at best. Differentiation of somatic cells into early granulosa cells during ovarian morphogenesis is the hallmark of primordial follicle formation; hence, critical changes are expected in protein expression. We wanted to identify proteins, the expression of which would correlate with the formation of primordial follicles as a first step to determine their biological function in folliculogenesis. Proteins were extracted from embryonic (E15) and 8-day-old (P8) hamster ovaries and fractionated by two-dimensional gel electrophoresis. Gels were stained with Proteosilver, and images of protein profiles corresponding to E15 and P8 ovaries were overlayed to identify protein spots showing altered expression. Some of the protein spots were extracted from SyproRuby-stained preparative gels, digested with trypsin, and analyzed by mass spectrometry. Both E15 and P8 ovaries had high molecular weight proteins at acidic, basic, and neutral ranges; however, we focused on small molecular weight proteins at 4-7 pH range. Many of those spots might represent post-translational modification. Mass spectrometric analysis revealed the identity of these proteins. The formation of primordial follicles on P8 correlated with many differentially and newly expressed proteins. Whereas Ebp1 expression was downregulated in ovarian somatic cells, Sfrs3 expression was specifically upregulated in newly formed granulosa cells of primordial follicles on P8. The results show for the first time that the morphogenesis of primordial follicles in the hamster coincides with altered and novel expression of proteins involved in cell proliferation, transcriptional regulation, and metabolism. Therefore, formation of primordial follicles is an active process requiring differentiation of somatic cells into early granulosa cells and their interaction with the oocytes.
Collapse
Affiliation(s)
- Anindit Mukherjee
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center 984515 Nebraska Medical Center, Omaha, NE 68198-4515
| | - Nichole Reisdorph
- Department of Immunology, National Jewish Medical and Research Center, 1400 Jackson St, K924, Denver, CO 80206
| | - Babu Guda
- Department of Genetics, Cell Biology and Anatomy, and Center for Bioinformatics and System Biology, University of Nebraska Medical Center 984515 Nebraska Medical Center, Omaha, NE 68198-4515
| | - Sanjit Pandey
- Department of Genetics, Cell Biology and Anatomy, and Center for Bioinformatics and System Biology, University of Nebraska Medical Center 984515 Nebraska Medical Center, Omaha, NE 68198-4515
| | - Shyamal K Roy
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center 984515 Nebraska Medical Center, Omaha, NE 68198-4515
- Department of Cellular and Integrative Physiology, Department of OB/GYN and Olson Center for Women's Health, and Eppley Institute for Cancer Research, University of Nebraska Medical Center 984515 Nebraska Medical Center, Omaha, NE 68198-4515
| |
Collapse
|
33
|
Günther J, Díaz V, Pedernera E, Méndez C. Gonadotropins regulate cadherin expression in the ovary of the chicken embryo (Gallus gallus). Gen Comp Endocrinol 2011; 174:344-7. [PMID: 22019531 DOI: 10.1016/j.ygcen.2011.09.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2011] [Revised: 08/22/2011] [Accepted: 09/23/2011] [Indexed: 11/17/2022]
Abstract
Cadherins are adhesion molecules that play a crucial role in tissue morphogenesis. Studies on N-cadherin and E-cadherin in the ovary of fetal hamster suggest that these adhesion molecules are involved in primordial follicle formation. In chicken embryo, present results demonstrate that N-cadherin is located on the surface epithelium and in the cortical cords of the ovary. Moreover, N-cadherin is identified in germ cells on day 14 of chicken embryo development. Quantification of mRNA of N-cadherin and E-cadherin demonstrates that treatments with follicle-stimulating hormone (FSH) or human chorionic gonadotropin (hCG), increase N-cadherin expression. Whereas, E-cadherin expression is decreased by gonadotropin treatments. The negative correlation between both cadherins expression is demonstrated after 18 h of hormonal treatment. Regulation of cadherin expression by gonadotropins and the presence of N-cadherin in the ovarian cortex suggest that these adhesion molecules are involved in ovarian morphogenesis in the chicken embryo.
Collapse
Affiliation(s)
- Jeannette Günther
- Departamento de Embriología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad Universitaria, México DF 04360, Mexico
| | | | | | | |
Collapse
|
34
|
Abstract
The differentiation of primordial germ cells (PGCs) into functional oocytes is important for the continuation of species. In mammals, PGCs begin to differentiate into oocytes during embryonic development. Oocytes develop in clusters called germ line cysts. During fetal or neonatal development, germ cell cysts break apart into single oocytes that become surrounded by pregranulosa cells to form primordial follicles. During the process of cyst breakdown, a subset of cells in each cyst undergoes cell death with only one-third of the initial number of oocytes surviving to form primordial follicles. The mechanisms that control cyst breakdown, oocyte survival, and follicle assembly are currently under investigation. This review describes the mechanisms that have been implicated in the control of primordial follicle formation, which include programmed cell death regulation, growth factor and other signaling pathways, regulation by transcription factors and hormones, meiotic progression, and changes in cell adhesion. Elucidation of mechanisms leading to formation of the primordial follicle pool will help research efforts in ovarian biology and improve treatments of female infertility, premature ovarian failure, and reproductive cancers.
Collapse
Affiliation(s)
- Melissa E Pepling
- Department of Biology, Syracuse University, Syracuse, New York 13244, USA.
| |
Collapse
|
35
|
Xu B, Hua J, Zhang Y, Jiang X, Zhang H, Ma T, Zheng W, Sun R, Shen W, Sha J, Cooke HJ, Shi Q. Proliferating cell nuclear antigen (PCNA) regulates primordial follicle assembly by promoting apoptosis of oocytes in fetal and neonatal mouse ovaries. PLoS One 2011; 6:e16046. [PMID: 21253613 PMCID: PMC3017099 DOI: 10.1371/journal.pone.0016046] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2010] [Accepted: 12/03/2010] [Indexed: 11/18/2022] Open
Abstract
Primordial follicles, providing all the oocytes available to a female throughout her reproductive life, assemble in perinatal ovaries with individual oocytes surrounded by granulosa cells. In mammals including the mouse, most oocytes die by apoptosis during primordial follicle assembly, but factors that regulate oocyte death remain largely unknown. Proliferating cell nuclear antigen (PCNA), a key regulator in many essential cellular processes, was shown to be differentially expressed during these processes in mouse ovaries using 2D-PAGE and MALDI-TOF/TOF methodology. A V-shaped expression pattern of PCNA in both oocytes and somatic cells was observed during the development of fetal and neonatal mouse ovaries, decreasing from 13.5 to 18.5 dpc and increasing from 18.5 dpc to 5 dpp. This was closely correlated with the meiotic prophase I progression from pre-leptotene to pachytene and from pachytene to diplotene when primordial follicles started to assemble. Inhibition of the increase of PCNA expression by RNA interference in cultured 18.5 dpc mouse ovaries strikingly reduced the apoptosis of oocytes, accompanied by down-regulation of known pro-apoptotic genes, e.g. Bax, caspase-3, and TNFα and TNFR2, and up-regulation of Bcl-2, a known anti-apoptotic gene. Moreover, reduced expression of PCNA was observed to significantly increase primordial follicle assembly, but these primordial follicles contained fewer granulosa cells. Similar results were obtained after down-regulation by RNA interference of Ing1b, a PCNA-binding protein in the UV-induced apoptosis regulation. Thus, our results demonstrate that PCNA regulates primordial follicle assembly by promoting apoptosis of oocytes in fetal and neonatal mouse ovaries.
Collapse
Affiliation(s)
- Bo Xu
- Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Juan Hua
- Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Yuanwei Zhang
- Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Xiaohua Jiang
- Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Huan Zhang
- Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Tieliang Ma
- Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Wei Zheng
- Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Rui Sun
- Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Wei Shen
- Department of Animal and Poultry Science, University of Guelph, Ontario, Canada
| | - Jiahao Sha
- Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Howard J. Cooke
- MRC Human Genetics Unit and Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, United Kingdom
| | - Qinghua Shi
- Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
- * E-mail:
| |
Collapse
|