51
|
Grossman H, Shalgi R. A Role of MicroRNAs in Cell Differentiation During Gonad Development. Results Probl Cell Differ 2016; 58:309-36. [PMID: 27300184 DOI: 10.1007/978-3-319-31973-5_12] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
MicroRNAs (miRNAs) are a group of small noncoding RNA molecules that play a major role in posttranscriptional regulation of gene expression and are expressed in an organ-specific manner. One miRNA can potentially regulate the expression of several genes, depending on cell type and differentiation stage. miRNAs are differentially expressed in the male and female gonads and have an organ-specific reproductive function. Exerting their affect through germ cells and gonadal somatic cells, miRNAs regulate key proteins necessary for gonad development. The role of miRNAs in the testes is only starting to emerge though they have been shown to be required for adequate spermatogenesis. Widely explored in the ovary, miRNAs were suggested to play a fundamental role in follicles' assembly, growth, differentiation, and ovulation. In this chapter, we focus on data obtained from mice in which distinct proteins that participate in the biosynthesis of miRNAs were conditionally knocked out from germ cells (spermatogonial cells or oocytes) or gonadal somatic cells (Sertoli or granulosa cells). We detail recent advances in identification of particular miRNAs and their significance in the development and function of male and female gonads. miRNAs can serve as biomarkers and therapeutic agents of pathological conditions; thus, elucidating the branched and complex network of reproduction-related miRNAs will aid understanding of gonads' physiology and managing reproduction disorders.
Collapse
Affiliation(s)
- Hadas Grossman
- Department of Cell Biology and Development, Tel Aviv University, Ramat Aviv, Israel
| | - Ruth Shalgi
- Department of Cell Biology and Development, Tel Aviv University, Ramat Aviv, Israel.
| |
Collapse
|
52
|
Wang D, Chen J, Li R, Wu G, Sun Z, Wang Z, Zhai Z, Fang F, Guo Y, Zhong Y, Jiang M, Xu H, Chen M, Shen G, Sun J, Yan B, Yu C, Tian Z, Xiao W. PAX5 interacts with RIP2 to promote NF-κB activation and drug-resistance of B-lymphoproliferative disorders. J Cell Sci 2016; 129:2261-72. [DOI: 10.1242/jcs.183889] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 04/11/2016] [Indexed: 12/17/2022] Open
Abstract
Paired box protein 5 (PAX5) plays a lineage determination role in B-cell development. However, high expression of PAX5 has been also found in various malignant diseases including B-lymphoproliferative disorders (B-LPDs), but its functions and mechanisms in these diseases are still unclear. Here, we show that PAX5 induces drug-resistance through association and activation of receptor-interacting serine/threonine-protein kinase2 (RIP2) and subsequent activation of NF-κB signaling and anti-apoptosis genes expression in B-lymphoproliferative cells. Furthermore, PAX5 is able to interact with RIP1-3, modulating both RIP1- mediated TNFR and RIP2-mediated NOD1 and NOD2 pathways. Our findings describe a novel function of PAX5 in regulating RIP1 and RIP2 activation, which is at least involved in chemo drug-resistance in B-LPDs.
Collapse
Affiliation(s)
- Dong Wang
- Key Laboratory of Innate Immunity and Chronic Disease of CAS, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
- Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
- Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, China
| | - Jingyu Chen
- Key Laboratory of Innate Immunity and Chronic Disease of CAS, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
- Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
- Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, China
| | - Rui Li
- Key Laboratory of Innate Immunity and Chronic Disease of CAS, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
- Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
- Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, China
| | - Guolin Wu
- Department of Hematology, Anhui Provincial Hospital, 17 Lujiang Road, Hefei, Anhui Province 230001, China
| | - Zimin Sun
- Department of Hematology, Anhui Provincial Hospital, 17 Lujiang Road, Hefei, Anhui Province 230001, China
| | - Zhitao Wang
- Department of Hematology, The Second Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui Province 230601, China
| | - Zhimin Zhai
- Department of Hematology, The Second Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui Province 230601, China
| | - Fang Fang
- Key Laboratory of Innate Immunity and Chronic Disease of CAS, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
- Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
- Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, China
| | - Yugang Guo
- Key Laboratory of Innate Immunity and Chronic Disease of CAS, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
- Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
- Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, China
| | - Yongjun Zhong
- Key Laboratory of Innate Immunity and Chronic Disease of CAS, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
- Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
- Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, China
| | - Ming Jiang
- Key Laboratory of Innate Immunity and Chronic Disease of CAS, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
- Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
- Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, China
| | - Huan Xu
- Key Laboratory of Innate Immunity and Chronic Disease of CAS, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
- Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
- Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, China
| | - Minhua Chen
- Key Laboratory of Innate Immunity and Chronic Disease of CAS, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
- Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
- Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, China
| | - Guodong Shen
- Key Laboratory of Innate Immunity and Chronic Disease of CAS, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
- Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
- Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, China
| | - Jie Sun
- Key Laboratory of Innate Immunity and Chronic Disease of CAS, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
- Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
- Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, China
| | - Bailing Yan
- Emergency Department, the First Hospital of Jilin Univesity, Changchun 130021, China
| | - Chundong Yu
- Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, China
| | - Zhigang Tian
- Key Laboratory of Innate Immunity and Chronic Disease of CAS, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
- Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
- Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, China
| | - Weihua Xiao
- Key Laboratory of Innate Immunity and Chronic Disease of CAS, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
- Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
- Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, China
| |
Collapse
|
53
|
Li JR, Wang W, Shi FX. Induction of follicular luteinization by equine chorionic gonadotropin in cyclic guinea pigs. J Zhejiang Univ Sci B 2015; 16:980-90. [PMID: 26642181 DOI: 10.1631/jzus.b1500046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The effects of equine chorionic gonadotropin (eCG) on follicular development and ovulation in cyclic guinea pigs were investigated by histological and immunohistochemical analyses. Three groups of guinea pigs (n=12) were administrated subcutaneously with saline, 20 or 50 IU of eCG, respectively, on cyclic Day 12 (Day 1=vaginal openings). Ovaries were collected at 4 and 8 d after administration (6 animals per group each time). The eCG administration induced significant and distinct morphological changes in the ovaries, as it promoted the luteinization of granulosa cells, but not follicular development. In addition, proliferating cell nuclear antigen (PCNA) and steroidogenic acute regulatory protein (StAR) were immunolocalized specifically in luteinized follicles. Our experiments together indicate that eCG administration can induce follicular luteinization but not superovulation in guinea pigs. The eCG in cyclic guinea pigs functions similar to that of luteinizing hormone (LH), but not follicle-stimulating hormone (FSH).
Collapse
Affiliation(s)
- Jun-rong Li
- Laboratory of Animal Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.,College of Agriculture and Bio-engineering, Jinhua Polytechnic, Jinhua 321017, China
| | - Wei Wang
- Laboratory of Animal Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Fang-xiong Shi
- Laboratory of Animal Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
54
|
Maalouf SW, Liu WS, Pate JL. MicroRNA in ovarian function. Cell Tissue Res 2015; 363:7-18. [PMID: 26558383 DOI: 10.1007/s00441-015-2307-4] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 09/29/2015] [Indexed: 01/14/2023]
Abstract
The mammalian ovary is a dynamic organ. The coordination of follicle recruitment, selection, and ovulation and the timely development and regression of the corpus luteum are essential for a functional ovary and fertility. Deregulation of any of these processes results in ovarian dysfunction and potential infertility. MicroRNA (miRNA) are short noncoding RNA that regulate developmental processes and time-sensitive functions. The expression of miRNA in the ovary varies with cell type, function, and stage of the estrous cycle. miRNA are involved in the formation of primordial follicles, follicular recruitment and selection, follicular atresia, oocyte-cumulus cell interaction, granulosal cell function, and luteinization. miRNA are differentially expressed in luteal cells at the various stages of the estrous cycle and during maternal recognition of pregnancy, suggesting a role in luteal development, maintenance, and regression. An understanding of the patterns of expression and functions of miRNA in the ovary will lead to novel therapeutics to treat ovarian dysfunction and improve fertility and, potentially, to the development of better contraceptives.
Collapse
Affiliation(s)
- S W Maalouf
- Department of Animal Science, Center for Reproductive Biology and Health, The Pennsylvania State University, 324 Henning Building, University Park, PA 16802, USA
| | - W S Liu
- Department of Animal Science, Center for Reproductive Biology and Health, The Pennsylvania State University, 324 Henning Building, University Park, PA 16802, USA
| | - J L Pate
- Department of Animal Science, Center for Reproductive Biology and Health, The Pennsylvania State University, 324 Henning Building, University Park, PA 16802, USA.
| |
Collapse
|
55
|
Growth Hormone Ameliorates the Radiotherapy-Induced Ovarian Follicular Loss in Rats: Impact on Oxidative Stress, Apoptosis and IGF-1/IGF-1R Axis. PLoS One 2015; 10:e0140055. [PMID: 26465611 PMCID: PMC4605641 DOI: 10.1371/journal.pone.0140055] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 09/21/2015] [Indexed: 01/25/2023] Open
Abstract
Radiotherapy is one of the standard cytotoxic therapies for cancer. However, it has a profound impact on ovarian function leading to premature ovarian failure and infertility. Since none of the currently available methods for fertility preservation guarantees future fertility, the need for an effective radioprotective agent is highly intensified. The present study investigated the mechanisms of the potential radioprotective effect of growth hormone (GH) on γ irradiation-induced ovarian failure and the impact of the insulin like growth factor 1 (IGF-1) in the underlying protection. Immature female Sprague-Dawley rats were either exposed to single whole body irradiation (3.2 Gy) and/or treated with GH (1 mg/kg s.c). Experimental γ-irradiation produced an array of ovarian dysfunction that was evident by assessment of hormonal changes, follicular development, proliferation marker (PCNA), oxidative stress as well as apoptotic markers. In addition, IGF-1/IGF-1R axis expression was assessed using real-time PCR and immunolocalization techniques. Furthermore, after full maturity, fertility assessment was performed. GH significantly enhanced follicular development and restored anti-Mullerian hormone serum level as compared with the irradiated group. In addition, GH significantly ameliorated the deleterious effects of irradiation on oxidative status, PCNA and apoptosis. Interestingly, GH was shown to enhance the ovarian IGF-1 at transcription and translation levels, a property that contributes significantly to its radioprotective effect. Finally, GH regained the fertility that was lost following irradiation. In conclusion, GH showed a radioprotective effect and rescued the ovarian reserve through increasing local IGF-1 level and counteracting the oxidative stress-mediated apoptosis.
Collapse
|
56
|
Melo RMC, Martins YS, Luz RK, Rizzo E, Bazzoli N. PCNA and apoptosis during post-spawning ovarian remodeling in the teleost Oreochromis niloticus. Tissue Cell 2015; 47:541-9. [PMID: 26542933 DOI: 10.1016/j.tice.2015.10.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 09/22/2015] [Accepted: 10/07/2015] [Indexed: 01/06/2023]
Abstract
The balance between cell proliferation and apoptosis is crucial for tissue development and homeostasis. The present study investigated the contribution of proliferating cell nuclear antigen (PCNA) and apoptosis during ovarian remodeling after spawning in the Nile tilapia Oreochromis niloticus. Breeding females were kept in controlled conditions and ovary samples were collected weekly for TUNEL assay, immunohistochemistry for PCNA and caspase-3 and morphometric analysis. During the follicular growth, PCNA labeled mainly the nuclei of oocytes and follicular cells in a high proportion of follicles especially in primary growth, while a low occurrence of apoptosis in follicular and theca cells was detected. At 0-3 days post-spawning, post-ovulatory follicles showed no proliferative activity, however the follicular cells exhibited high rates of apoptosis. At 7-10 days, PCNA labeled the thecal cells in a low proportion of post-ovulatory follicles, which showed follicular cells with lower rates of apoptosis. PCNA labeled mainly the theca in the advanced and late stages of atretic follicles, while the follicular cells exhibited a significant increase of apoptosis along follicular atresia. We concluded that PCNA and apoptosis work cooperatively to ensuring the success of follicle development and maintaining of tissue homeostasis during follicular growth. PCNA and apoptosis are also essential mechanisms in the follicular regression during post-spawning ovarian recovery in the Nile tilapia.
Collapse
Affiliation(s)
- Rafael M C Melo
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Yuri S Martins
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Ronald K Luz
- Laboratório de Aquacultura, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Elizete Rizzo
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Nilo Bazzoli
- Programa de Pós-graduação em Zoologia de Vertebrados, Pontifícia Universidade Católica de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
57
|
Li J, Shen T, Wu G, Wei Q, Mao D, Shi F. Potential roles of matrix metalloproteinases and characteristics of ovarian development in neonatal guinea pigs. Tissue Cell 2015; 47:478-88. [DOI: 10.1016/j.tice.2015.07.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 07/01/2015] [Accepted: 07/20/2015] [Indexed: 12/31/2022]
|
58
|
Xie S, Zhang X, Chen W, Xie C, Chen W, Cheng P, Zhou Y, Chen B. Developmental Status: Impact of Short-Term Ischemia on Follicular Survival of Whole Ovarian Transplantation in a Rabbit Model. PLoS One 2015; 10:e0135049. [PMID: 26271079 PMCID: PMC4536045 DOI: 10.1371/journal.pone.0135049] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2014] [Accepted: 07/16/2015] [Indexed: 12/24/2022] Open
Abstract
Ischemia is the first mechanism that provokes the loss of follicles in ovarian grafts over the long term. In whole ovarian transplantation, it remains unknown, however, how changes in follicular development are influenced by short-term ischemia. Fresh whole ovarian orthotopic auto-transplantation was performed in rabbits with 45 min ischemia, and the impact of ischemia on follicular survival and development status was evaluated at different time-points (1 day, 3 days, 1 week, 2 weeks and 1 month). Assessment of follicular quantity and morphology was carried out via histologic analysis. Follicle proliferating status was evidenced by immunostaining with proliferating cell nuclear antigen (PCNA), and the Hedgehog signaling pathway (Patched and Gli); was verified via TUNEL assay. Quantitative PCR was carried out to quantify the mRNA of target genes including PCNA, Patched, Gli, Caspase 3, Bax, and Bcl-2. Compared with its contralateral fresh controls, the morphology, proliferation and apoptosis of the follicles in the grafts showed no significant differences and most primordial follicles were quiescent. However, morphology and proliferation status were significantly decreased 1 week after grafting, in comparison with the longitudinal grafting time. Patched and Gli in the Hedgehog signaling pathway were activated in only the follicles of the grafts. Short-term ischemia slightly impacts follicular survival and development status in whole ovarian grafting. Receiving intervention in the first week post-transplantation might be helpful.
Collapse
Affiliation(s)
- Shuangshuang Xie
- Obstetrical Department, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Xing Zhang
- Surgical Laboratory, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Wenming Chen
- Centre for Reproductive Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Chichi Xie
- Centre for Reproductive Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Wenwei Chen
- Transplantation Centre, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Pu Cheng
- Oncological Surgery, the Second Affiliated Hospital of Zhejiang University School of Medcine, Hangzhou, Zhejiang, 310000, China
| | - Ying Zhou
- Centre for Reproductive Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- * E-mail: (YZ); (BC)
| | - Bicheng Chen
- Transplantation Centre, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- * E-mail: (YZ); (BC)
| |
Collapse
|
59
|
Lin Y, Sun Z. In vivo pancreatic β-cell-specific expression of antiaging gene Klotho: a novel approach for preserving β-cells in type 2 diabetes. Diabetes 2015; 64:1444-58. [PMID: 25377875 PMCID: PMC4375073 DOI: 10.2337/db14-0632] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Protein expression of an antiaging gene, Klotho, was depleted in pancreatic islets in patients with type 2 diabetes mellitus (T2DM) and in db/db mice, an animal model of T2DM. The objective of this study was to investigate whether in vivo expression of Klotho would preserve pancreatic β-cell function in db/db mice. We report for the first time that β-cell-specific expression of Klotho attenuated the development of diabetes in db/db mice. β-Cell-specific expression of Klotho decreased hyperglycemia and enhanced glucose tolerance. The beneficial effects of Klotho were associated with significant improvements in T2DM-induced decreases in number of β-cells, insulin storage levels in pancreatic islets, and glucose-stimulated insulin secretion from pancreatic islets, which led to increased blood insulin levels in diabetic mice. In addition, β-cell-specific expression of Klotho decreased intracellular superoxide levels, oxidative damage, apoptosis, and DNAJC3 (a marker for endoplasmic reticulum stress) in pancreatic islets. Furthermore, β-cell-specific expression of Klotho increased expression levels of Pdx-1 (insulin transcription factor), PCNA (a marker of cell proliferation), and LC3 (a marker of autophagy) in pancreatic islets in db/db mice. These results reveal that β-cell-specific expression of Klotho improves β-cell function and attenuates the development of T2DM. Therefore, in vivo expression of Klotho may offer a novel strategy for protecting β-cells in T2DM.
Collapse
Affiliation(s)
- Yi Lin
- Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Zhongjie Sun
- Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| |
Collapse
|
60
|
Xu B, Zhang YW, Tong XH, Liu YS. Characterization of microRNA profile in human cumulus granulosa cells: Identification of microRNAs that regulate Notch signaling and are associated with PCOS. Mol Cell Endocrinol 2015; 404:26-36. [PMID: 25622783 DOI: 10.1016/j.mce.2015.01.030] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Revised: 01/03/2015] [Accepted: 01/20/2015] [Indexed: 12/26/2022]
Abstract
CONTEXT Polycystic ovary syndrome (PCOS), a complex and heterogeneous endocrine condition, is characterized by polycystic ovaries, hyperandrogenism, insulin resistance and chronic anovulation. Cumulus granulosa cells surrounding the oocyte are involved in different aspects of PCOS pathology. Several studies suggested that miRNAs play an important regulatory role at the post-transcriptional level in cumulus granulosa cells. OBJECTIVE Our objective was to describe the altered miRNA expression profiles and miRNA targeted signaling pathways in PCOS. DESIGN Case-control study that involved 21 women with PCOS and 20 women without the disease (controls). The miRNA expression profiles of human cumulus granulosa cells were determined using next generation sequencing by Illumina Hiseq 2000. The differentially expressed miRNAs and novel miRNAs were validated by quantitative real-time PCR. The Notch3 and MAPK3 were demonstrated to be targeted by miR-483-5p based on quantitative real-time PCR, western blot and luciferase activity assay. RESULTS Compared with controls, a total of 59 known miRNA were identified that differentially expressed in PCOS cumulus granulosa cells, including 21 miRNAs increase and 38 miRNAs decrease. Moreover, the novel miRNAs were predicted in PCOS and control cumulus granulosa cells. The potential regulating roles of miRNA in pathophysiology of PCOS were analyzed by GO and KEGG pathway annotation, and several important processes were identified to be targeted by the differentially expressed miRNAs, such as Notch signaling, regulation of hormone, and energy metabolism. Furthermore, Notch3 and MAPK3, the members of Notch signaling and ERK-MAPK pathway, were demonstrated to be regulated by miR-483-5p based on negative expression correlation validation and detection of Notch3/MAPK3 expression after miR-483-5p mimics transfection. Dual luciferase activity assay suggested that Notch3 and MAPK3 were directly targeted by miR-483-5p. CONCLUSION Our data suggested that miRNAs and their targeted pathways (e.g. Notch signaling pathway) play important roles in the etiology and pathophysiology of PCOS, and provides novel candidates for molecular biomarkers or treatment targets in the research of female infertility associated with PCOS.
Collapse
Affiliation(s)
- Bo Xu
- Center for Reproductive Medicine, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei 230001, China.
| | - Yuan-Wei Zhang
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Xian-Hong Tong
- Center for Reproductive Medicine, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei 230001, China
| | - Yu-Sheng Liu
- Center for Reproductive Medicine, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei 230001, China
| |
Collapse
|
61
|
Li C, Guo S, Zhang M, Gao J, Guo Y. DNA methylation and histone modification patterns during the late embryonic and early postnatal development of chickens. Poult Sci 2015; 94:706-21. [PMID: 25691759 DOI: 10.3382/ps/pev016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Early mammalian embryonic cells have been proven to be essential for embryonic development and the health of neonates. A series of epigenetic reprogramming events, including DNA methylation and histone modifications, occur during early embryonic development. However, epigenetic marks in late embryos and neonates are not well understood, especially in avian species. To investigate the epigenetic patterns of developing embryos and posthatched chicks, embryos at embryonic day 5 (E5), E8, E11, E14, E17, and E20 and newly hatched chicks on day of life 1 (D1), D7, D14, D21 were collected. The levels of global DNA methylation and histone H3 at lysine 9 residue (H3K9) modifications were measured in samples of liver, jejunum, and breast skeletal muscles by Western blotting and immunofluorescence staining. According to our data, decreased levels of proliferating cell nuclear antigen expression were found in the liver and a V-shaped pattern of proliferating cell nuclear antigen expression was found in the jejunum. The level of proliferating cell nuclear antigen in muscle was relatively stable. Caspase 3 expression gradually decreased over time in liver, was stable in the jejunum, and increased in muscle. Levels of DNA methylation and H3K9 acetylation decreased in liver over time, while the pattern was N-shaped in jejunal tissue and W-shaped in pectoral muscles, and these changes were accompanied by dynamic changes of DNA methyltransferases, histone acetyltransferases 1, and histone deacetylase 2. Moreover, dimethylation, trimethylation, and acetylation of H3K9 were expressed in a time- and tissue-dependent manner. After birth, epigenetic marks were relatively stable and found at lower levels. These results indicate that spatiotemporal specific epigenetic alterations could be critical for the late development of chick embryos and neonates.
Collapse
Affiliation(s)
- Changwu Li
- State Key Laboratory of Animal Nutrition, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China
| | - Shuangshuang Guo
- State Key Laboratory of Animal Nutrition, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China
| | - Ming Zhang
- State Key Laboratory of Animal Nutrition, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China
| | - Jing Gao
- State Key Laboratory of Animal Nutrition, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China
| | - Yuming Guo
- State Key Laboratory of Animal Nutrition, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China
| |
Collapse
|
62
|
Guzmán JM, Luckenbach JA, Yamamoto Y, Swanson P. Expression profiles of Fsh-regulated ovarian genes during oogenesis in coho salmon. PLoS One 2014; 9:e114176. [PMID: 25485989 PMCID: PMC4259363 DOI: 10.1371/journal.pone.0114176] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 11/05/2014] [Indexed: 01/17/2023] Open
Abstract
The function of follicle-stimulating hormone (Fsh) during oogenesis in fishes is poorly understood. Using coho salmon as a fish model, we recently identified a suite of genes regulated by Fsh in vitro and involved in ovarian processes mostly unexplored in fishes, like cell proliferation, differentiation, survival or extracellular matrix (ECM) remodeling. To better understand the role of these Fsh-regulated genes during oocyte growth in fishes, we characterized their mRNA levels at discrete stages of the ovarian development in coho salmon. While most of the transcripts were expressed at low levels during primary growth (perinucleolus stage), high expression of genes associated with cell proliferation (pim1, pcna, and mcm4) and survival (ddit4l) was found in follicles at this stage. The transition to secondary oocyte growth (cortical alveolus and lipid droplet stage ovarian follicles) was characterized by a marked increase in the expression of genes related to cell survival (clu1, clu2 and ivns1abpa). Expression of genes associated with cell differentiation and growth (wt2l and adh8l), growth factor signaling (inha), steroidogenesis (cyp19a1a) and the ECM (col1a1, col1a2 and dcn) peaked in vitellogenic follicles, showing a strong and positive correlation with transcripts for fshr. Other genes regulated by Fsh and associated with ECM function (ctgf, wapl and fn1) and growth factor signaling (bmp16 and smad5l) peaked in maturing follicles, along with increases in steroidogenesis-related gene transcripts. In conclusion, ovarian genes regulated by Fsh showed marked differences in their expression patterns during oogenesis in coho salmon. Our results suggest that Fsh regulates different ovarian processes at specific stages of development, likely through interaction with other intra- or extra-ovarian factors.
Collapse
Affiliation(s)
- José M. Guzmán
- Environmental and Fisheries Sciences Division, Northwest Fisheries Science Center, National Marine Fisheries Service, National Oceanic and Atmospheric Administration, Seattle, Washington, 98112, United States of America
- * E-mail:
| | - J. Adam Luckenbach
- Environmental and Fisheries Sciences Division, Northwest Fisheries Science Center, National Marine Fisheries Service, National Oceanic and Atmospheric Administration, Seattle, Washington, 98112, United States of America
- Center for Reproductive Biology, Washington State University, Pullman, Washington, 99164, United States of America
| | - Yoji Yamamoto
- Environmental and Fisheries Sciences Division, Northwest Fisheries Science Center, National Marine Fisheries Service, National Oceanic and Atmospheric Administration, Seattle, Washington, 98112, United States of America
- School of Aquatic and Fishery Sciences, University of Washington, Seattle, Washington, 98195, United States of America
- Department of Marine Biosciences, Tokyo University of Marine Science and Technology, 4-5-7 Konan, Minato-ku, Tokyo, 108-8477, Japan
| | - Penny Swanson
- Environmental and Fisheries Sciences Division, Northwest Fisheries Science Center, National Marine Fisheries Service, National Oceanic and Atmospheric Administration, Seattle, Washington, 98112, United States of America
- Center for Reproductive Biology, Washington State University, Pullman, Washington, 99164, United States of America
| |
Collapse
|
63
|
Kwak DH, Park JH, Lee HS, Moon JS, Lee S. Aristolochic Acid I Induces Ovarian Toxicity by Inhibition of Akt Phosphorylation. Chem Res Toxicol 2014; 27:2128-35. [DOI: 10.1021/tx5003854] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Dong Hoon Kwak
- Institute
for Glycoscience, Wonkwang University, Iksan 570-749, Republic of Korea
| | - Ji-Hye Park
- Department
of Pharmacology, Wonkwang University School of Medicine and Wonkwang Brain Research Institute, Iksan 570-749, Republic of Korea
| | - Hak-Seung Lee
- Department
of Pharmacology, Wonkwang University School of Medicine and Wonkwang Brain Research Institute, Iksan 570-749, Republic of Korea
| | - Ji-Sook Moon
- Department
of Pharmacology, Wonkwang University School of Medicine and Wonkwang Brain Research Institute, Iksan 570-749, Republic of Korea
| | - Seoul Lee
- Department
of Pharmacology, Wonkwang University School of Medicine and Wonkwang Brain Research Institute, Iksan 570-749, Republic of Korea
| |
Collapse
|
64
|
Sui S, Jia Y, He B, Li R, Li X, Cai D, Song H, Zhang R, Zhao R. Maternal Low-protein Diet Alters Ovarian Expression of Folliculogenic and Steroidogenic Genes and Their Regulatory MicroRNAs in Neonatal Piglets. ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES 2014; 27:1695-704. [PMID: 25358362 PMCID: PMC4213680 DOI: 10.5713/ajas.2014.14335] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 07/25/2014] [Accepted: 08/23/2014] [Indexed: 11/27/2022]
Abstract
Maternal malnutrition during pregnancy may give rise to female offspring with disrupted ovary functions in adult age. Neonatal ovary development predisposes adult ovary function, yet the effect of maternal nutrition on the neonatal ovary has not been described. Therefore, here we show the impact of maternal protein restriction on the expression of folliculogenic and steroidogenic genes, their regulatory microRNAs and promoter DNA methylation in the ovary of neonatal piglets. Sows were fed either standard-protein (SP, 15% crude protein) or low-protein (LP, 7.5% crude protein) diets throughout gestation. Female piglets born to LP sows showed significantly decreased ovary weight relative to body weight (p<0.05) at birth, which was accompanied with an increased serum estradiol level (p<0.05). The LP piglets demonstrated higher ratio of bcl-2 associated X protein/B cell lymphoma/leukemia-2 mRNA (p<0.01), which was associated with up-regulated mRNA expression of bone morphogenic protein 4 (BMP4) (p<0.05) and proliferating cell nuclear antigen (PCNA) (p<0.05). The steroidogenic gene, cytochrome P450 aromatase (CYP19A1) was significantly down-regulated (p<0.05) in LP piglets. The alterations in ovarian gene expression were associated with a significant down-regulation of follicle-stimulating hormone receptor mRNA expression (p<0.05) in LP piglets. Moreover, three microRNAs, including miR-423-5p targeting both CYP19A1 and PCNA, miR-378 targeting CYP19A1 and miR-210 targeting BMP4, were significantly down-regulated (p<0.05) in the ovary of LP piglets. These results suggest that microRNAs are involved in mediating the effect of maternal protein restriction on ovarian function through regulating the expression of folliculogenic and steroidogenic genes in newborn piglets.
Collapse
Affiliation(s)
- Shiyan Sui
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Nanjing 210095, China ; The Department of Agriculture and Biological Sciences, Dali University of Yunnan Province, Dali 671003, China
| | - Yimin Jia
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Nanjing 210095, China
| | - Bin He
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Nanjing 210095, China
| | - Runsheng Li
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Nanjing 210095, China
| | - Xian Li
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Nanjing 210095, China
| | - Demin Cai
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Nanjing 210095, China
| | - Haogang Song
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Nanjing 210095, China
| | - Rongkui Zhang
- Shanghai Farm of Bright Food (Group) Co., Ltd, Dafeng 224100, China
| | - Ruqian Zhao
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
65
|
Sui S, He B, Jia Y, Li R, Cai D, Li X, Song H, Jia L, Zhao R. Maternal protein restriction during gestation and lactation programs offspring ovarian steroidogenesis and folliculogenesis in the prepubertal gilts. J Steroid Biochem Mol Biol 2014; 143:267-76. [PMID: 24787658 DOI: 10.1016/j.jsbmb.2014.04.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 04/17/2014] [Accepted: 04/20/2014] [Indexed: 01/28/2023]
Abstract
Maternal malnutrition may disrupt ovarian functions in adult offspring. Steroidogenesis and folliculogenesis in the offspring ovary appear to be the major targets of nutritional programming. Nevertheless, the mechanism by which maternal low-protein diet affects the offspring steroidogenesis and folliculogenesis, and the possible pathway linking these two processes remain unclear. In this study, Landrace×Yorkshire crossbred sows were fed either standard (SP) or low-protein (LP, 50% of the SP) diets throughout gestation and lactation. Female offspring were fed the same diet after weaning until 6 months of age. LP offspring had higher serum 17β-estradiol level (P<0.01), which was accompanied by lower mRNA (P<0.05) but higher protein (P<0.05) expression of cytochrome P450 aromatase (CYP19A1) in the ovary. CYP19A1 protein up-regulation was associated with lower ovarian expression of drosha (P<0.05) and miRNAs targeting CYP19A1 (P<0.05). LP offspring had less graafian follicles with more apoptotic granulosa cells (P<0.05), as well as higher caspase 3 activity (P<0.05) and FasL expression (P<0.05) in the ovary. FasL gene up-regulation was associated with higher ERα protein expression (P<0.05) and binding to FasL gene promoter. These results suggest that a maternal LP diet in pregnancy and lactation elevated serum 17β-estradiol level by activating CYP19A1 through miRNA-mediated mechanism, and induced granulosa apoptosis in graafian follicles through ER-activated Fas/FasL-caspase 3 pathway.
Collapse
Affiliation(s)
- Shiyan Sui
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Nanjing 210095, China
| | - Bin He
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Nanjing 210095, China
| | - Yimin Jia
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Nanjing 210095, China
| | - Runsheng Li
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Nanjing 210095, China
| | - Demin Cai
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Nanjing 210095, China
| | - Xi Li
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Nanjing 210095, China
| | - Haogang Song
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Nanjing 210095, China
| | - Longfei Jia
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Nanjing 210095, China
| | - Ruqian Zhao
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
66
|
Zhang H, Jiang X, Zhang Y, Xu B, Hua J, Ma T, Zheng W, Sun R, Shen W, Cooke HJ, Hao Q, Qiao J, Shi Q. microRNA 376a regulates follicle assembly by targeting Pcna in fetal and neonatal mouse ovaries. Reproduction 2014; 148:43-54. [DOI: 10.1530/rep-13-0508] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
In mammals, the primordial follicle pool, providing all oocytes available to a female throughout her reproductive life, is established perinatally. Dysregulation of primordial follicle assembly results in female reproductive diseases, such as premature ovarian insufficiency and infertility. Female mice lackingDicer1(Dicer), a gene required for biogenesis of microRNAs, show abnormal morphology of follicles and infertility. However, the contribution of individual microRNAs to primordial follicle assembly remains largely unknown. Here, we report that microRNA 376a (miR-376a) regulates primordial follicle assembly by modulating the expression of proliferating cell nuclear antigen (Pcna), a gene we previously reported to regulate primordial follicle assembly by regulating oocyte apoptosis in mouse ovaries. miR-376a was shown to be negatively correlated withPcnamRNA expression in fetal and neonatal mouse ovaries and to directly bind toPcnamRNA 3′ untranslated region. Cultured 18.5 days postcoitum mouse ovaries transfected with miR-376a exhibited decreasedPcnaexpression both in protein and mRNA levels. Moreover, miR-376a overexpression significantly increased primordial follicles and reduced apoptosis of oocytes, which was very similar to those in ovaries co-transfected with miR-376a and siRNAs targetingPcna. Taken together, our results demonstrate that miR-376a regulates primordial follicle assembly by modulating the expression ofPcna. To our knowledge, this is the first microRNA–target mRNA pair that has been reported to regulate mammalian primordial follicle assembly and further our understanding of the regulation of primordial follicle assembly.
Collapse
|
67
|
Abstract
The female germline comprises a reserve population of primordial (non-growing) follicles containing diplotene oocytes arrested in the first meiotic prophase. By convention, the reserve is established when all individual oocytes are enclosed by granulosa cells. This commonly occurs prior to or around birth, according to species. Histologically, the ‘reserve’ is the number of primordial follicles in the ovary at any given age and is ultimately depleted by degeneration and progression through folliculogenesis until exhausted. How and when the reserve reaches its peak number of follicles is determined by ovarian morphogenesis and germ cell dynamics involving i) oogonial proliferation and entry into meiosis producing an oversupply of oocytes and ii) large-scale germ cell death resulting in markedly reduced numbers surviving as the primordial follicle reserve. Our understanding of the processes maintaining the reserve comes primarily from genetically engineered mouse models, experimental activation or destruction of oocytes, and quantitative histological analysis. As the source of ovulated oocytes in postnatal life, the primordial follicle reserve requires regulation of i) its survival or maintenance, ii) suppression of development (dormancy), and iii) activation for growth and entry into folliculogenesis. The mechanisms influencing these alternate and complex inter-related phenomena remain to be fully elucidated. Drawing upon direct and indirect evidence, we discuss the controversial concept of postnatal oogenesis. This posits a rare population of oogonial stem cells that contribute new oocytes to partially compensate for the age-related decline in the primordial follicle reserve.
Collapse
|
68
|
Wang F, Wang L, Shi Z, Liang G. Comparative N-glycoproteomic and phosphoproteomic profiling of human placental plasma membrane between normal and preeclampsia pregnancies with high-resolution mass spectrometry. PLoS One 2013; 8:e80480. [PMID: 24260401 PMCID: PMC3829899 DOI: 10.1371/journal.pone.0080480] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Accepted: 10/03/2013] [Indexed: 11/22/2022] Open
Abstract
Preeclampsia is a serious complication of pregnancy, which affects 2–8% of all pregnancies and is one of the leading causes of maternal and perinatal mortality and morbidity worldwide. To better understand the molecular mechanisms involved in pathological development of placenta in preeclampsia, we used high-resolution LC-MS/MS technologies to construct a comparative N-glycoproteomic and phosphoproteomic profiling of human placental plasma membrane in normal and preeclamptic pregnancies. A total of 1027 N-glyco- and 2094 phospho- sites were detected in human placental plasma membrane, and 5 N-glyco- and 38 phospho- proteins, respectively, with differentially expression were definitively identified between control and preeclamptic placental plasma membrane. Further bioinformatics analysis indicated that these differentially expressed proteins correlate with several specific cellular processes occurring during pathological changes of preeclamptic placental plasma membrane.
Collapse
Affiliation(s)
- Fuqiang Wang
- CAS Key Laboratory of Soft Matter Chemistry, Department of Chemistry, University of Science and Technology of China, Anhui, China ; State Key Laboratory of Reproductive Medicine, Analysis Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | | | | | | |
Collapse
|
69
|
Wang F, Shi Z, Wang P, You W, Liang G. Comparative proteome profile of human placenta from normal and preeclamptic pregnancies. PLoS One 2013; 8:e78025. [PMID: 24205073 PMCID: PMC3799759 DOI: 10.1371/journal.pone.0078025] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 09/09/2013] [Indexed: 12/17/2022] Open
Abstract
To better understand the molecular mechanisms involved in pathological development of placenta in preeclampsia, we used LC-MS/MS to construct a large-scale comparative proteome profile of human placentas from normal and preeclamptic pregnancies. A total of 2636 proteins were detected in human placentas, and 171 different proteins were definitively identified between control and preeclamptic placentas. Further bioinformatics analysis indicated that these differentially expressed proteins correlate with several specific cellular processes which occur during pathological changes of preeclamptic placenta. 6 proteins were randomly selected to verify their expression patterns with Western blotting. Of which, 3 proteins’ cellular localizations were validated with immunohistochemistry. Elucidation of how protein-expression changes coordinate the pathological development would provide researchers with a better understanding of the critical biological processes of preeclampsia and potential targets for therapeutic agents to regulate placenta function, and eventually benefit the treatment of preeclampsia.
Collapse
Affiliation(s)
- Fuqiang Wang
- CAS Key Laboratory of Soft Matter Chemistry, Department of Chemistry, University of Science and Technology of China, Hefei, Anhui, China
- State Key Laboratory of Reproductive Medicine, Analysis Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhonghua Shi
- State Key Laboratory of Reproductive Medicine, Analysis Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ping Wang
- Key Laboratory of Living Donor Liver Transplantation, Ministry of Public Health, Department of Liver Transplantation Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China;
| | - Wei You
- Key Laboratory of Living Donor Liver Transplantation, Ministry of Public Health, Department of Liver Transplantation Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China;
- * E-mails: (GL); (WY)
| | - Gaolin Liang
- CAS Key Laboratory of Soft Matter Chemistry, Department of Chemistry, University of Science and Technology of China, Hefei, Anhui, China
- * E-mails: (GL); (WY)
| |
Collapse
|
70
|
Mahran YF, El-Demerdash E, Nada AS, Ali AA, Abdel-Naim AB. Insights into the protective mechanisms of tamoxifen in radiotherapy-induced ovarian follicular loss: impact on insulin-like growth factor 1. Endocrinology 2013; 154:3888-99. [PMID: 23798597 DOI: 10.1210/en.2013-1214] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Radiotherapy is one of the most common and effective cancer treatments. However, it has a profound impact on ovarian function, leading to premature ovarian failure. With the hope of preserving fertility in cancer survivors, the need for an effective radioprotective therapy is evident. The present study investigated the mechanism of the potential radioprotective effect of tamoxifen (TAM) on γ-irradiation-induced ovarian failure on experimental rats and the impact of the IGF-1 in the underlying protective mechanisms. Female Sprague Dawley rats were either exposed to single whole-body irradiation (3.2 Gy; lethal dose [LD₂₀]) and/or treated with TAM (1 mg/kg). γ-Irradiation caused an array of ovarian dysfunction that was evident by assessment of hormonal changes, follicular development, proliferation marker (proliferating cell nuclear antigen), and oxidative stress as well as apoptotic markers. In addition, IGF-1/IGF-1 receptor axis expression was assessed using real-time RT-PCR and immunolocalization techniques. Furthermore, fertility assessment was performed. TAM significantly enhanced follicular development and restored the anti-Mullerian hormone level. Moreover, it ameliorated the deleterious effects of irradiation on oxidative stress, proliferating cell nuclear antigen expression, and apoptosis. Interestingly, TAM was shown to enhance the ovarian IGF-1 but not IGF-1 receptor, a property that contributed significantly to its radioprotective mechanisms. Finally, TAM regained the fertility that was lost after irradiation. In conclusion, TAM showed a radioprotective effect and saved the ovarian reserve and fertility through increasing anti-Mullerian hormone and the local IGF-1 level and counteracting the oxidative stress-mediated apoptosis.
Collapse
Affiliation(s)
- Yasmen F Mahran
- Professor of Pharmacology and Toxicology, Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Abasia, Cairo, Egypt. or
| | | | | | | | | |
Collapse
|
71
|
Abd-Allah SH, Shalaby SM, Pasha HF, El-Shal AS, Raafat N, Shabrawy SM, Awad HA, Amer MG, Gharib MA, El Gendy EA, Raslan AA, El-Kelawy HM. Mechanistic action of mesenchymal stem cell injection in the treatment of chemically induced ovarian failure in rabbits. Cytotherapy 2013; 15:64-75. [PMID: 23260087 DOI: 10.1016/j.jcyt.2012.08.001] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Accepted: 08/16/2012] [Indexed: 01/02/2023]
Abstract
BACKGROUND No curative treatment is known for primary ovarian failure; however, mesenchymal stem cells (MSCs), through self-renewal and regeneration, push the trial to evaluate their role in the treatment of ovarian failure. The aim of this study was to explore the impact of MSCs on cyclophosphamide (CTX)-induced ovarian failure in rabbits and to clarify the mechanism(s) by which MSCs exert their action. METHODS Thirty-five adult female rabbits were injected with CTX to induce ovarian failure. Five rabbits were euthanized after the last injection of CTX for histological examination. The others (30 rabbits) were further subdivided into two groups: group 1 (ovarian failure group, 15 rabbits) received no treatment; group 2 (ovarian failure and MSC recipient group, 15 rabbits) received MSCs isolated from extracted bone marrow of male rabbits. RESULTS A decrease of follicle-stimulating hormone and an increase of estrogen and vascular endothelial growth factor (VEGF) levels in the MSC recipient group versus the ovarian failure group were found. Weak caspase-3 expression and +ve proliferating cell nuclear antigen staining after MSC injection were detected. Cytological and histological examinations showed increased follicle numbers with apparent normal structure of ovarian follicles in the MSC recipient group. Moreover, Y chromosome-containing cells from male donors were present within the ovarian tissues in group 2. CONCLUSIONS The current study suggests that intravenous injection of MSCs into rabbits with chemotherapy-induced ovarian damage improved ovarian function. MSCs accomplish this function by direct differentiation into specific cellular phenotypes and by secretion of VEGF, which influence the regeneration of the ovary.
Collapse
Affiliation(s)
- Somia H Abd-Allah
- Medical Biochemistry Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
72
|
Nteeba J, Ross JW, Perfield JW, Keating AF. High fat diet induced obesity alters ovarian phosphatidylinositol-3 kinase signaling gene expression. Reprod Toxicol 2013; 42:68-77. [PMID: 23954404 DOI: 10.1016/j.reprotox.2013.07.026] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 07/29/2013] [Accepted: 07/31/2013] [Indexed: 12/17/2022]
Abstract
Insulin regulates ovarian phosphatidylinositol-3-kinase (PI3 K) signaling, important for primordial follicle viability and growth activation. This study investigated diet-induced obesity impacts on: (1) insulin receptor (Insr) and insulin receptor substrate 1 (Irs1); (2) PI3K components (Kit ligand (Kitlg), kit (c-Kit), protein kinase B alpha (Akt1) and forkhead transcription factor subfamily 3 (Foxo3a)); (3) xenobiotic biotransformation (microsomal epoxide hydrolase (Ephx1), Cytochrome P450 isoform 2E1 (Cyp2e1), Glutathione S-transferase (Gst) isoforms mu (Gstm) and pi (Gstp)) and (4) microRNA's 184, 205, 103 and 21 gene expression. INSR, GSTM and GSTP protein levels were also measured. Obese mouse ovaries had decreased Irs1, Foxo3a, Cyp2e1, MiR-103, and MiR-21 but increased Kitlg, Akt1, and miR-184 levels relative to lean littermates. These results support that diet-induced obesity potentially impairs ovarian function through aberrant gene expression.
Collapse
Affiliation(s)
- J Nteeba
- Department of Animal Science, Iowa State University, Ames, IA 50011, USA.
| | | | | | | |
Collapse
|
73
|
Role of Mael in early oogenesis and during germ-cell differentiation from embryonic stem cells in mice in vitro. ZYGOTE 2013; 22:513-20. [DOI: 10.1017/s0967199412000743] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
SummaryIn a previous study, we have identified a set of conserved spermatogenic genes whose expression is restricted to testis and ovary and that are developmentally regulated. One of these genes, the transcription factor Mael, has been reported to play an essential role in mouse spermatogenesis. Nevertheless, the role of Mael in mouse oogenesis has not been defined. In order to analyse the role of Mael in mouse oogenesis, the expression of this gene was blocked during early oogenesis in mouse in vitro using RNAi technology. In addition, the role of Mael during differentiation of embryonic stem cells (ESC) into germ cells in vitro was analysed. Results show that downregulation of Mael by a specific short interfering RNA disrupted fetal oocyte growth and differentiation in fetal ovary explants in culture and the expression of several germ-cell markers in ESC during their differentiation. These results suggest that there is an important role for Mael in early oogenesis and during germ-cell differentiation from embryonic stem cells in mouse in vitro.
Collapse
|
74
|
Sato A, Shibuya H. WNK signaling is involved in neural development via Lhx8/Awh expression. PLoS One 2013; 8:e55301. [PMID: 23383144 PMCID: PMC3559379 DOI: 10.1371/journal.pone.0055301] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2012] [Accepted: 12/20/2012] [Indexed: 02/07/2023] Open
Abstract
WNK kinase family is conserved among many species and regulates SPAK/OSR1 and ion co-transporters. Some mutations in human WNK1 or WNK4 are associated with Pseudohypoaldosteronism type II, a form of hypertension. WNK is also involved in developmental and cellular processes, but the molecular mechanisms underlying its regulation in these processes remain unknown. Here, we identify a new target gene in WNK signaling, Arrowhead and Lhx8, which is a mammalian homologue of Drosophila Arrowhead. In Drosophila, WNK was shown to genetically interact with Arrowhead. In Wnk1 knockout mice, levels of Lhx8 expression were reduced. Ectopic expression of WNK1, WNK4 or Osr1 in mammalian cells induced the expression of the Lhx8. Moreover, neural specification was inhibited by the knockdown of both Wnk1 and Wnk4 or Lhx8. Drosophila WNK mutant caused defects in axon guidance during embryogenesis. These results suggest that WNK signaling is involved in the morphological and neural development via Lhx8/Arrowhead.
Collapse
Affiliation(s)
- Atsushi Sato
- Department of Molecular Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Hiroshi Shibuya
- Department of Molecular Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
- * E-mail:
| |
Collapse
|
75
|
Sun X, Tong H, Zhang M, Wang XH. Rosuvastatin inhibits the smooth muscle cell proliferation by targeting TNFα mediated Rho kinase pathway. J Geriatr Cardiol 2012; 9:180-4. [PMID: 22916066 PMCID: PMC3418909 DOI: 10.3724/sp.j.1263.2012.03301] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Revised: 05/21/2012] [Accepted: 05/28/2012] [Indexed: 01/28/2023] Open
Abstract
Objective To investigate whether Tumor Necrosis Factor-alpha (TNFα) is capable of activating Rho kinase pathway which leads to smooth muscle cell proliferation and the intervention function of Rosuvastatin, and clarify the mechanism and intervention manner of anti-atherosclerosis by Rosuvastatin. Methods Wistar neonate rat smooth muscle cells were cultured, and the activity of cell proliferation was determined by methyl thiazolyl tetrazolium (MTT). The expression of Rho kinase genes after the stimulation of TNFα was evaluated by RT-PCR. Western blot method was used to measure the protein expression of proliferating cell nuclear antigen (PCNA) after TNFα stimulation and Rosuvastatin intervention in smooth muscle cell. Results The TNFα stimulation significantly enhanced the expression of Rho kinase and increased the expression of PCNA protein in smooth muscle cells (P < 0.05). These effects were positively correlated with prolonged treatment whereas additional Rosuvastatin administration inhibited the above-mentioned effects (P < 0.05). Conclusions The activation of TNFα mediated Rho kinase signaling pathway can significantly promote smooth muscle cell proliferation, and Rosuvastatin can not only inhibit this pathway but also the induced proliferation.
Collapse
Affiliation(s)
- Xiao Sun
- Department of Cardiology, Fengtian Hospital affiliated to Shenyang Medical College, Shenyang 110024, Liaoning Province, China
| | | | | | | |
Collapse
|
76
|
Salvetti NR, Ortega HH, Veiga-Lopez A, Padmanabhan V. Developmental programming: impact of prenatal testosterone excess on ovarian cell proliferation and apoptotic factors in sheep. Biol Reprod 2012; 87:22, 1-10. [PMID: 22539681 DOI: 10.1095/biolreprod.112.100024] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Prenatal testosterone (T) excess leads to reproductive dysfunctions in sheep, which include increased ovarian follicular recruitment and persistence. To test the hypothesis that follicular disruptions in T sheep stem from changes in the developmental ontogeny of ovarian proliferation and apoptotic factors, pregnant Suffolk sheep were injected twice weekly with T propionate or dihydrotestosterone propionate (DHT; a nonaromatizable androgen) from Days 30 to 90 of gestation. Changes in developmental expression of proliferating cell nuclear antigen (PCNA), BCL2, BAX, activated CASP3, and FAS/FASLG were determined at Fetal Days 90 and 140, 22 wk, 10 mo, and 21 mo of age by immunocytochemisty. Prenatal T treatment induced changes in expression of proliferative and apoptotic markers in a follicle-, age-, and steroid-specific manner. Changes in BAX were evident only during fetal life and PCNA, BCL2, and CASP3 only postnatally. Prenatal T and not DHT increased PCNA and decreased BCL2 in granulosa/theca cells of antral follicles at 10 and 21 mo but decreased CASP3 in granulosa/theca cells of antral follicles at 22 wk (prepubertal) and 10 and 21 mo. Both treatments decreased BAX immunostaining in granulosa cells of Fetal Day 90 primordial/primary follicles. Neither treatment affected FAS expression at any developmental time point in any follicular compartment. Effects on BAX appear to be programmed by androgenic actions and PCNA, BCL2, and CASP3 by estrogenic actions of T. Overall, the findings demonstrate that fetal exposure to excess T disrupts the ovarian proliferation/apoptosis balance, thus providing a basis for the follicular disruptions evidenced in these females.
Collapse
Affiliation(s)
- Natalia R Salvetti
- Department of Morphological Sciences, Faculty of Veterinary Sciences, National University of Litoral, Esperanza, Argentina
| | | | | | | |
Collapse
|
77
|
Abstract
SummaryOogenesis is a highly complex process that requires the exquisite temporal and spatial regulation of gene expression at multiple levels. Skin–embryo–brain–oocyte homeobox (Sebox) gene encodes a transcription factor that is highly expressed in germinal vesicle stage oocytes and that plays an essential role in early embryogenesis at the 2-cell stage in the mouse. AsSeboxis also expressed in mouse fetal ovaries, the aim of the present study was to study its role during the early oogenesisin vitro. Expression ofSeboxwas low in 15.5 to 17.5 days post coitum (dpc) ovaries, showed a peak at 18.5 dpc and then its expression decreased dramatically in newborn ovaries.Seboxexpression was efficiently knocked down (>80%) in fetal mouse ovary explants in culture using RNAi technology. When fetal ovary explants were transfected withSebox-specific RNAi, the number of oocytes at germinal vesicle stage and showing a diameter of 40–70 μm was decreased significantly to 75% after 7 days of culture relative to the negative control, and to 22.4% after 10 days of culture, thus indicating thatSeboxplays an important role in the early oogenesis in mice.
Collapse
|
78
|
Zhang LJ, Pan B, Chen B, Zhang XF, Liang GJ, Feng YN, Wang LQ, Ma JM, Li L, Shen W. Expression and epigenetic dynamics of transcription regulator Lhx8 during mouse oogenesis. Gene 2012; 506:1-9. [PMID: 22796561 DOI: 10.1016/j.gene.2012.06.093] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Revised: 06/23/2012] [Accepted: 06/28/2012] [Indexed: 12/22/2022]
Abstract
The spatial and temporal specific activation and inhibition of numerous genes are required for successful oogenesis which is precisely regulated by germ cell-related transcription factors, and appropriate epigenetic modifications, including DNA methylation, histone modification and other mechanisms that closely regulate the functional exertion of these transcription factors. In this study, we characterized the correlation between the expression and epigenetic dynamics of Lhx8, a germ cell specific transcription factor during mouse oogenesis. Immunohistochemistry, quantitative PCR and western blots were performed to localize and quantify the expressional characteristics of Lhx8 in oocytes of 13.5 dpc (day post coitum), 17.5 dpc, 0 dpp (day post partum), 3 dpp, 7 dpp and 14 dpp. The results showed that LHX8 protein was located in the nucleus of oocytes, and increasingly expressed during primordial follicle activation. Sequencing of bisulfite-converted genomic DNAs revealed that the methylation dynamics of Lhx8-3' was highly changeable but almost no change occurred in Lhx8-5'. ChIP-QPCR analysis showed that histone H3 acetylation of Lhx8 was also increased during primordial follicle assembly and activation. In conclusion, Lhx8 expression is related with the activation of primordial follicles, which is highly correlated with the demethylation of Lhx8-3' untranslated region and the high acetylation of histone H3.
Collapse
Affiliation(s)
- Lian-Jun Zhang
- Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Li W, Zhou N. URG4 upregulation is associated with tumor growth and poor survival in epithelial ovarian cancer. Arch Gynecol Obstet 2012; 286:209-15. [PMID: 22395861 DOI: 10.1007/s00404-012-2269-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Accepted: 02/20/2012] [Indexed: 11/28/2022]
Abstract
PURPOSE Up-regulated gene 4 (URG4) has been demonstrated to be involved in progression of various human cancers. This study investigated the clinicopathological significance of URG4 in epithelial ovarian cancer (EOC). METHODS Immunohistochemistry was applied to investigate the expression of URG4 in ovarian tissues of 116 patients. The correlation of URG4 with proliferating cell nuclear antigen index (PCNA) was analyzed and the prognostic value of URG4 in patients was also investigated. Pearson Chi-square test, Spearman correlation coefficient, univariate analysis, multivariate analysis, and Kaplan-Meier method were adopted. RESULTS The positive rate of URG4 in EOC was higher than that in borderline and benign tumors (P = 0.001). URG4 was positively correlated with PCNA (r = 0.86, P = 0.006). In addition, univariate analysis showed URG4 expression level, clinical stage, pathologic grade, lymphatic metastasis, chemotherapy, and ascites influenced survival time (all P < 0.05). In Cox multivariate analysis, all the aforementioned factors were found to be independent prognostic factors except pathologic grade and ascites (all P < 0.05). CONCLUSIONS Our results suggest for the first time that the URG4 might be involved in the progression of EOC. URG4 was a new target to assess the prognosis of EOC.
Collapse
Affiliation(s)
- Weiping Li
- Department of Gynaecology and Obstetrics, General Hospital of PLA, Beijing, China
| | | |
Collapse
|
80
|
Sarraj MA, Drummond AE. Mammalian foetal ovarian development: consequences for health and disease. Reproduction 2012; 143:151-63. [DOI: 10.1530/rep-11-0247] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The development of a normal ovary during foetal life is essential for the production and ovulation of a high-quality oocyte in adult life. Early in embryogenesis, the primordial germ cells (PGCs) migrate to and colonise the genital ridges. Once the PGCs reach the bipotential gonad, the absence of the sex-determining region on the Y chromosome (SRY) gene and the presence of female-specific genes ensure that the indifferent gonad takes the female pathway and an ovary forms. PGCs enter into meiosis, transform into oogonia and ultimately give rise to oocytes that are later surrounded by granulosa cells to form primordial follicles. Various genes and signals are implicated in germ and somatic cell development, leading to successful follicle formation and normal ovarian development. This review focuses on the differentiation events, cellular processes and molecular mechanisms essential for foetal ovarian development in the mice and humans. A better understanding of these early cellular and morphological events will facilitate further study into the regulation of oocyte development, manifestation of ovarian disease and basis of female infertility.
Collapse
|
81
|
Chen D, Zhang Y, Yi Q, Huang Y, Hou H, Zhang Y, Hao Q, Cooke HJ, Li L, Sun Q, Shi Q. Regulation of asymmetrical cytokinesis by cAMP during meiosis I in mouse oocytes. PLoS One 2012; 7:e29735. [PMID: 22253767 PMCID: PMC3256179 DOI: 10.1371/journal.pone.0029735] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Accepted: 12/04/2011] [Indexed: 01/24/2023] Open
Abstract
Mammalian oocytes undergo an asymmetrical first meiotic division, extruding half of their chromosomes in a small polar body to preserve maternal resources for embryonic development. To divide asymmetrically, mammalian oocytes relocate chromosomes from the center of the cell to the cortex, but little is known about the underlying mechanisms. Here, we show that upon the elevation of intracellular cAMP level, mouse oocytes produced two daughter cells with similar sizes. This symmetrical cell division could be rescued by the inhibition of PKA, a cAMP-dependent protein kinase. Live cell imaging revealed that a symmetrically localized cleavage furrow resulted in symmetrical cell division. Detailed analyses demonstrated that symmetrically localized cleavage furrows were caused by the inappropriate central positioning of chromosome clusters at anaphase onset, indicating that chromosome cluster migration was impaired. Notably, high intracellular cAMP reduced myosin II activity, and the microinjection of phospho-myosin II antibody into the oocytes impeded chromosome migration and promoted symmetrical cell division. Our results support the hypothesis that cAMP plays a role in regulating asymmetrical cell division by modulating myosin II activity during mouse oocyte meiosis I, providing a novel insight into the regulation of female gamete formation in mammals.
Collapse
Affiliation(s)
- Dawei Chen
- School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Yuanwei Zhang
- School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Qiyi Yi
- School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Yun Huang
- School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Heli Hou
- School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Yingyin Zhang
- School of Life Sciences, University of Science and Technology of China, Hefei, China
- Hefei National Laboratory for Physical Sciences at Microscale, Hefei, China
| | - Qiaomei Hao
- School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Howard J. Cooke
- School of Life Sciences, University of Science and Technology of China, Hefei, China
- Hefei National Laboratory for Physical Sciences at Microscale, Hefei, China
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, United Kingdom
| | - Lei Li
- Chinese Academy of Sciences, Beijing, China
| | | | - Qinghua Shi
- School of Life Sciences, University of Science and Technology of China, Hefei, China
- Hefei National Laboratory for Physical Sciences at Microscale, Hefei, China
- * E-mail:
| |
Collapse
|