1
|
Chen SY, Wang TE, Lee WY, Yang YY, Lai HC, Matsuda F, Kosek H, Chen YT, Li SH, Tsai PS. Cre-LoxP and tamoxifen-induced deletion of ovarian quiescin sulfhydryl oxidase 2 showed disruption of ovulatory activity in mice. J Ovarian Res 2024; 17:66. [PMID: 38504307 PMCID: PMC10949576 DOI: 10.1186/s13048-024-01388-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 03/08/2024] [Indexed: 03/21/2024] Open
Abstract
BACKGROUND Quiescin sulfhydryl oxidase 2 (QSOX2) is a flavin adenine dinucleotide-dependent sulfhydryl oxidase that is known to be involved in protein folding, cell growth regulation, and redox state modification through oxidative activities. Earlier studies demonstrated the tissue and cellular localization of QSOX2 in the male reproductive tract, as well as the highly-regulated mechanism of QSOX2 protein synthesis and expression through the coordinated action of testosterone and epididymal-enriched amino acid, glutamate. However, the presence and the functions of QSOX2 in female reproduction are unknown. In this study, we applied the Cre-loxP gene manipulation system to generate the heterozygous and homozygous Qsox2 knockout mice and examined its effects on ovarian function. RESULTS We demonstrated that QSOX2 was detected in the follicle-supporting cells (granulosa and cumulus cells) of ovarian follicles of all stages but was absent in the corpus luteum, suggesting its supportive role in folliculogenesis. In comparison with reproductive organogenesis in wild-type mice, there was no difference in testicular and epididymal structure in male Qsox2 knockout; however, Qsox2 knockout disrupted the regular ovulation process in female mice as a drastic decrease in the formation of the corpus luteum was detected, and no pregnancy was achieved when mating males with homozygous Qsox2 knockout females. RNAseq analyses further revealed that Qsox2 knockout altered critical signaling pathways and genes that are responsible for maintaining ovarian functions. CONCLUSION Our data demonstrated for the first time that Qsox2 is critical for ovarian function in mice.
Collapse
Affiliation(s)
- Shih-Yun Chen
- Department of Veterinary Medicine, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan
| | - Tse-En Wang
- Graduate Institute of Veterinary Medicine, National Taiwan University, 10617, Taipei, Taiwan
| | - Wei-Yun Lee
- Graduate Institute of Veterinary Medicine, National Taiwan University, 10617, Taipei, Taiwan
| | - Ya-Yi Yang
- Graduate Institute of Veterinary Medicine, National Taiwan University, 10617, Taipei, Taiwan
| | - Hong-Chun Lai
- Department of Veterinary Medicine, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan
| | - Fuko Matsuda
- Department of Veterinary Medical Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, 113-8657, Japan
| | - Haruhiko Kosek
- Center for Integrative Medical Sciences (IMS), RIEKN, Yokohama, Kanagawa, 230-0045, Japan
| | - You-Tzung Chen
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, 10617, Taipei, Taiwan
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, 10055, Taipei, Taiwan
| | - Sheng-Hsiang Li
- Department of Medical Research, MacKay Memorial Hospital, 25160, Tamsui, Taiwan
| | - Pei-Shiue Tsai
- Department of Veterinary Medicine, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan.
- Graduate Institute of Veterinary Medicine, National Taiwan University, 10617, Taipei, Taiwan.
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, 10617, Taipei, Taiwan.
| |
Collapse
|
2
|
Expression profile of genes related to pregnancy maintenance in Dromedary Camel during the first trimester. Anim Reprod Sci 2023; 251:107211. [PMID: 36990016 DOI: 10.1016/j.anireprosci.2023.107211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 02/27/2023] [Accepted: 03/06/2023] [Indexed: 03/18/2023]
Abstract
So far, few signals involved in embryo-maternal dialogue have been identified in pregnant she-camel. Our objective was to investigate expression profiles of genes relevant to uterine extracellular matrix remodeling (ITGB4, SLCO2A1, FOS, and JUN), uterine tissue vascularization, and placental formation (VEGFA, PGF, and PDGFA), embryonic growth and development (IGF1 and PTEN), plus cell death of uterine tissue (BCL2) in early pregnant versus non-pregnant she-camels. Forty genital tracts (20 pregnant and 20 non-pregnant) and blood samples were collected from abattoirs. Total RNA was extracted from uterine tissues and qRT-PCR was conducted for candidate genes. Serum concentrations of progesterone (P4) and estradiol17-β (E2) were measured. Expression of ITGB4, FOS, and PGF genes increased (P < 0.001) in the right uterine horn of pregnant versus non-pregnant she-camels. Moreover, JUN, SLCO2A1, VEGFA, and PTEN mRNAs were up-regulated (P < 0.001) in various segments of uterine tissues in pregnant groups. The PDGFA transcript was over-expressed (P < 0.001) in both uterine horns of pregnant groups. Additionally, IGF1 was higher (P < 0.001) in the right horn and the uterine body of pregnant groups, and expression of BCL2 was increased (P < 0.001) in the pregnant uterine body. Moreover, serum concentrations of P4 were higher (P < 0.001) and E2 lower (P < 0.05) in pregnant she-camels. Taken together, the fine-tuning of genes related to implantation, matrix formation, vascularization, and placental formation is highly required for successful pregnancy in she-camels.
Collapse
|
3
|
Pascuali N, Scotti L, Oubiña G, de Zúñiga I, Gomez Peña M, Pomilio C, Saravia F, Tesone M, Abramovich D, Parborell F. Platelet-derived growth factor B restores vascular barrier integrity and diminishes permeability in ovarian hyperstimulation syndrome. Mol Hum Reprod 2021; 26:585-600. [PMID: 32467982 DOI: 10.1093/molehr/gaaa038] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 04/08/2020] [Indexed: 01/29/2023] Open
Abstract
Although advances in the prediction and management of ovarian hyperstimulation syndrome (OHSS) have been introduced, complete prevention is not yet possible. Previously, we and other authors have shown that vascular endothelial growth factor, angiopoietins (ANGPTs) and sphingosine-1-phosphate are involved in OHSS etiology. In addition, we have demonstrated that ovarian protein levels of platelet-derived growth factor (PDGF) ligands -B and -D decrease in an OHSS rat model, whilst PDGFR-β and ANGPT2 remain unchanged. In the present work, we investigated the role of PDGF-B in OHSS by evaluating ligand protein levels in follicular fluid (FF) from women at risk of developing OHSS and by using an immature rat model of OHSS. We demonstrated that PDGF-B and PDGF-D are lower in FF from women at risk of developing OHSS compared to control patients (P < 0.05). In the OHSS rat model, PDGF-B (0.5 µg/ovary) administration decreased ovarian weight (P < 0.05), reduced serum progesterone (P < 0.05) and lowered the percentage of cysts (P < 0.05), compared to untreated OHSS rats, but had no effect on the proportion of follicles or corpora lutea (CL). PDGF-B treatment also restored the expression of steroidogenic acute regulatory protein (P < 0.05) and P450 cholesterol side-chain cleavage enzyme (P < 0.01) to control levels. In addition, PDGF-B increased the peri-endothelial cell area in CL and cystic structures, and reduced vascular permeability compared to untreated OHSS ovaries. Lastly, PDGF-B increased the levels of junction proteins claudin-5 (P < 0.05), occludin (P < 0.05) and β-catenin (P < 0.05), while boosting the extracellular deposition of collagen IV surrounding the ovarian vasculature (PP < 0.01), compared to OHSS alone. In conclusion, our findings indicate that PDGF-B could be another crucial mediator in the onset and development of OHSS, which may lead to the development of novel prediction markers and therapeutic strategies.
Collapse
Affiliation(s)
- Natalia Pascuali
- Instituto de Biología y Medicina Experimental (IByME) - CONICET, Buenos Aires, Argentina
| | - Leopoldina Scotti
- Instituto de Biología y Medicina Experimental (IByME) - CONICET, Buenos Aires, Argentina
| | - Gonzalo Oubiña
- Instituto de Biología y Medicina Experimental (IByME) - CONICET, Buenos Aires, Argentina
| | | | | | - Carlos Pomilio
- Instituto de Biología y Medicina Experimental (IByME) - CONICET, Buenos Aires, Argentina.,Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Flavia Saravia
- Instituto de Biología y Medicina Experimental (IByME) - CONICET, Buenos Aires, Argentina.,Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Marta Tesone
- Instituto de Biología y Medicina Experimental (IByME) - CONICET, Buenos Aires, Argentina.,Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Dalhia Abramovich
- Instituto de Biología y Medicina Experimental (IByME) - CONICET, Buenos Aires, Argentina
| | - Fernanda Parborell
- Instituto de Biología y Medicina Experimental (IByME) - CONICET, Buenos Aires, Argentina
| |
Collapse
|
4
|
Human Granulosa Cells-Stemness Properties, Molecular Cross-Talk and Follicular Angiogenesis. Cells 2021; 10:cells10061396. [PMID: 34198768 PMCID: PMC8229878 DOI: 10.3390/cells10061396] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/29/2021] [Accepted: 06/02/2021] [Indexed: 12/31/2022] Open
Abstract
The ovarian follicle is the basic functional unit of the ovary, comprising theca cells and granulosa cells (GCs). Two different types of GCs, mural GCs and cumulus cells (CCs), serve different functions during folliculogenesis. Mural GCs produce oestrogen during the follicular phase and progesterone after ovulation, while CCs surround the oocyte tightly and form the cumulus oophurus and corona radiata inner cell layer. CCs are also engaged in bi-directional metabolite exchange with the oocyte, as they form gap-junctions, which are crucial for both the oocyte’s proper maturation and GC proliferation. However, the function of both GCs and CCs is dependent on proper follicular angiogenesis. Aside from participating in complex molecular interplay with the oocyte, the ovarian follicular cells exhibit stem-like properties, characteristic of mesenchymal stem cells (MSCs). Both GCs and CCs remain under the influence of various miRNAs, and some of them may contribute to polycystic ovary syndrome (PCOS) or premature ovarian insufficiency (POI) occurrence. Considering increasing female fertility problems worldwide, it is of interest to develop new strategies enhancing assisted reproductive techniques. Therefore, it is important to carefully consider GCs as ovarian stem cells in terms of the cellular features and molecular pathways involved in their development and interactions as well as outline their possible application in translational medicine.
Collapse
|
5
|
Sominsky L, Younesi S, De Luca SN, Loone SM, Quinn KM, Spencer SJ. Ovarian follicles are resistant to monocyte perturbations-implications for ovarian health with immune disruption†. Biol Reprod 2021; 105:100-112. [PMID: 33709094 DOI: 10.1093/biolre/ioab049] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/12/2020] [Accepted: 03/10/2021] [Indexed: 02/03/2023] Open
Abstract
Monocytes and macrophages are the most abundant immune cell populations in the adult ovary, with well-known roles in ovulation and corpus luteum formation and regression. They are activated and proliferate in response to immune challenge and are suppressed by anti-inflammatory treatments. It is also likely they have a functional role in the healthy ovary in supporting the maturing follicle from the primordial through to the later stages; however, this role has been unexplored until now. Here, we utilized a Cx3cr1-Dtr transgenic Wistar rat model that allows a conditional depletion of circulating monocytes, to investigate their role in ovarian follicle health. Our findings show that circulating monocyte depletion leads to a significant depletion of ovarian monocytes and monocyte-derived macrophages. Depletion of monocytes was associated with a transient reduction in circulating anti-Müllerian hormone (AMH) at 5 days postdepletion. However, the 50-60% ovarian monocyte/macrophage depletion had no effect on ovarian follicle numbers, follicle atresia, or apoptosis, within 5-21 days postdepletion. These data reveal that the healthy adult ovary is remarkably resistant to perturbations of circulating and ovarian monocytes despite acute changes in AMH. These data suggest that short-term anti-inflammatory therapies that transiently impact on circulating monocytes are unlikely to disrupt ovarian follicle health, findings that have significant implications for fertility planning relative to the experience of an immune challenge or immunosuppression.
Collapse
Affiliation(s)
- Luba Sominsky
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
| | - Simin Younesi
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
| | - Simone N De Luca
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
| | - Sophie M Loone
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
| | - Kylie M Quinn
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
| | - Sarah J Spencer
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia.,ARC Centre of Excellence for Nanoscale Biophotonics, RMIT University, Melbourne, Victoria, Australia
| |
Collapse
|
6
|
Basini G, Bussolati S, Grolli S, Ramoni R, Conti V, Quintavalla F, Grasselli F. Platelets are involved in in vitro swine granulosa cell luteinization and angiogenesis. Anim Reprod Sci 2017; 188:51-56. [PMID: 29174088 DOI: 10.1016/j.anireprosci.2017.11.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 10/05/2017] [Accepted: 11/08/2017] [Indexed: 10/18/2022]
Abstract
During corpus luteum formation, impressive biological events take place to guarantee the transition from original follicular to luteal cells and to support required massive angiogenesis. It has been demonstrated that these phenomena resemble those essential for wound healing. After ovulation, blood vessels release their content in the antral cavity and coagulation takes place. Involvement of platelets in corpus luteum growth has been hypothesized both in human and in rat. On this basis, using platelet lysate (PL), a blood derivative with a higher platelet concentration, we aimed to assess a potential involvement of platelets in swine granulosa cell luteinization and on new blood vessel growth. Our results demonstrate, for the first time in the swine, that platelets could be directly involved in granulosa cell physiological luteinization, since the treatment with PL shifted steroid production from estradiol 17β to progesterone. Moreover, PL stimulated angiogenesis. Nitric oxide could be involved in these effects. These results are important to clarify complex intrafollicular molecular machinery. A better understanding of these mechanisms can be useful to develop more focused therapeutic strategies to contrast sow infertility. In addition, since the pig represents a model for translational studies, collected data could be of interest for human medicine because reproductive pathologies such as Polycystic Ovary Syndrome (PCOS) and endometriosis are often accompanied by platelet dysfunctions.
Collapse
Affiliation(s)
- Giuseppina Basini
- Dipartimento di Scienze Medico-Veterinarie, Università degli Studi di Parma, Via del Taglio 10, 43126 Parma, Italy.
| | - Simona Bussolati
- Dipartimento di Scienze Medico-Veterinarie, Università degli Studi di Parma, Via del Taglio 10, 43126 Parma, Italy
| | - Stefano Grolli
- Dipartimento di Scienze Medico-Veterinarie, Università degli Studi di Parma, Via del Taglio 10, 43126 Parma, Italy
| | - Roberto Ramoni
- Dipartimento di Scienze Medico-Veterinarie, Università degli Studi di Parma, Via del Taglio 10, 43126 Parma, Italy
| | - Virna Conti
- Dipartimento di Scienze Medico-Veterinarie, Università degli Studi di Parma, Via del Taglio 10, 43126 Parma, Italy
| | - Fausto Quintavalla
- Dipartimento di Scienze Medico-Veterinarie, Università degli Studi di Parma, Via del Taglio 10, 43126 Parma, Italy
| | - Francesca Grasselli
- Dipartimento di Scienze Medico-Veterinarie, Università degli Studi di Parma, Via del Taglio 10, 43126 Parma, Italy
| |
Collapse
|
7
|
Liu DT, Brewer MS, Chen S, Hong W, Zhu Y. Transcriptomic signatures for ovulation in vertebrates. Gen Comp Endocrinol 2017; 247:74-86. [PMID: 28111234 PMCID: PMC5410184 DOI: 10.1016/j.ygcen.2017.01.019] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 01/14/2017] [Accepted: 01/17/2017] [Indexed: 01/01/2023]
Abstract
The central roles of luteinizing hormone (LH), progestin and their receptors for initiating ovulation have been well established. However, signaling pathways and downstream targets such as proteases that are essential for the rupture of follicular cells are still unclear. Recently, we found anovulation in nuclear progestin receptor (Pgr) knockout (Pgr-KO) zebrafish, which offers a new model for examining genes and pathways that are important for ovulation and fertility. In this study, we examined expression of all transcripts using RNA-Seq in preovulatory follicular cells collected following the final oocyte maturation, but prior to ovulation, from wild-type (WT) or Pgr-KO fish. Differential expression analysis revealed 3567 genes significantly differentially expressed between WT and Pgr-KO fish (fold change⩾2, p<0.05). Among those, 1543 gene transcripts were significantly more expressed, while 2024 genes were significantly less expressed, in WT than those in Pgr-KO. We then retrieved and compared transcriptional data from online databases and further identified 661 conserved genes in fish, mice, and humans that showed similar levels of high (283 genes) or low (387) expression in animals that were ovulating compared to those with no ovulation. For the first time, ovulatory genes and their involved biological processes and pathways were also visualized using Enrichment Map and Cytoscape. Intriguingly, enrichment analysis indicated that the genes with higher expression were involved in multiple ovulatory pathways and processes such as inflammatory response, angiogenesis, cytokine production, cell migration, chemotaxis, MAPK, focal adhesion, and cytoskeleton reorganization. In contrast, the genes with lower expression were mainly involved in DNA replication, DNA repair, DNA methylation, RNA processing, telomere maintenance, spindle assembling, nuclear acid transport, catabolic processes, and nuclear and cell division. Our results indicate that a large set of genes (>3000) is differentially regulated in the follicular cells in zebrafish prior to ovulation, terminating programs such as growth and proliferation, and beginning processes including the inflammatory response and apoptosis. Further studies are required to establish relationships among these genes and an ovulatory circuit in the zebrafish model.
Collapse
Affiliation(s)
- Dong Teng Liu
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, Fujian Province 361102, People's Republic of China; Department of Biology, East Carolina University, Greenville, NC 27858, United States
| | - Michael S Brewer
- Department of Biology, East Carolina University, Greenville, NC 27858, United States
| | - Shixi Chen
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, Fujian Province 361102, People's Republic of China
| | - Wanshu Hong
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, Fujian Province 361102, People's Republic of China
| | - Yong Zhu
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, Fujian Province 361102, People's Republic of China; Department of Biology, East Carolina University, Greenville, NC 27858, United States.
| |
Collapse
|
8
|
|
9
|
Xu K, Chen X, Yang H, Xu Y, He Y, Wang C, Huang H, Liu B, Liu W, Li J, Kou X, Zhao Y, Zhao K, Zhang L, Hou Z, Wang H, Wang H, Li J, Fan H, Wang F, Gao Y, Zhang Y, Chen J, Gao S. Maternal Sall4 Is Indispensable for Epigenetic Maturation of Mouse Oocytes. J Biol Chem 2016; 292:1798-1807. [PMID: 28031467 DOI: 10.1074/jbc.m116.767061] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 12/09/2016] [Indexed: 12/24/2022] Open
Abstract
Sall4 (Splat-like 4) plays important roles in maintaining pluripotency of embryonic stem cells and in various developmental processes. Here, we find that Sall4 is highly expressed in oocytes and early embryos. To investigate the roles of SALL4 in oogenesis, we generated Sall4 maternal specific knock-out mice by using CRISPR/Cas9 system, and we find that the maternal deletion of Sall4 causes developmental arrest of oocytes at germinal vesicle stage with non-surrounded nucleus, and the subsequent meiosis resumption is prohibited. We further discover that the loss of maternal Sall4 causes failure in establishment of DNA methylation in oocytes. Furthermore, we find that Sall4 modulates H3K4me3 and H3K27me3 modifications by regulating the expression of key histone demethylases coding genes Kdm5b, Kdm6a, and Kdm6b in oocytes. Moreover, we demonstrate that the aberrant H3K4me3 and H3K27me3 cause mis-expression of genes that are critical for oocytes maturation and meiosis resumption. Taken together, our study explores a pivotal role of Sall4 in regulating epigenetic maturation of mouse oocytes.
Collapse
Affiliation(s)
- Kai Xu
- From the Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Xia Chen
- From the Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Hui Yang
- From the Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Yiwen Xu
- the Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China
| | - Yuanlin He
- the State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, Nanjing 210029, China
| | - Chenfei Wang
- From the Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Hua Huang
- the State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Baodong Liu
- the State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Wenqiang Liu
- From the Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Jingyi Li
- From the Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Xiaochen Kou
- From the Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Yanhong Zhao
- From the Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Kun Zhao
- From the Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Linfeng Zhang
- From the Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Zhenzhen Hou
- From the Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Hong Wang
- From the Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Hailin Wang
- the State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Jing Li
- the State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, Nanjing 210029, China
| | - Hengyu Fan
- the Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China
| | - Fengchao Wang
- National Institute of Biological Sciences, Beijing 102206, China
| | - Yawei Gao
- From the Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Yong Zhang
- From the Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Jiayu Chen
- From the Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China.
| | - Shaorong Gao
- From the Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China.
| |
Collapse
|
10
|
Di Pietro M, Scotti L, Irusta G, Tesone M, Parborell F, Abramovich D. Local administration of platelet-derived growth factor B (PDGFB) improves follicular development and ovarian angiogenesis in a rat model of Polycystic Ovary Syndrome. Mol Cell Endocrinol 2016; 433:47-55. [PMID: 27256152 DOI: 10.1016/j.mce.2016.05.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 05/04/2016] [Accepted: 05/27/2016] [Indexed: 11/18/2022]
Abstract
Alterations in ovarian angiogenesis are common features in Polycystic Ovary Syndrome (PCOS) patients; the most studied of these alterations is the increase in vascular endothelial growth factor (VEGF) production by ovarian cells. Platelet-derived growth factor B (PDGFB) and D (PDGFD) are decreased in follicular fluid of PCOS patients and in the ovaries of a rat model of PCOS. In the present study, we aimed to analyze the effects of local administration of PDGFB on ovarian angiogenesis, follicular development and ovulation in a DHEA-induced PCOS rat model. Ovarian PDGFB administration to PCOS rats partially restored follicular development, decreased the percentage of cysts, increased the percentage of corpora lutea, and decreased the production of anti-Müllerian hormone. In addition, PDGFB administration improved ovarian angiogenesis by reversing the increase in periendothelial cell area and restoring VEGF levels. Our results shed light into the mechanisms that lead to altered ovarian function in PCOS and provide new data for potential therapeutic strategies.
Collapse
Affiliation(s)
- Mariana Di Pietro
- Instituto de Biología y Medicina Experimental (IByME-CONICET), Vuelta de Obligado 2490, Buenos Aires, Argentina.
| | - Leopoldina Scotti
- Instituto de Biología y Medicina Experimental (IByME-CONICET), Vuelta de Obligado 2490, Buenos Aires, Argentina.
| | - Griselda Irusta
- Instituto de Biología y Medicina Experimental (IByME-CONICET), Vuelta de Obligado 2490, Buenos Aires, Argentina.
| | - Marta Tesone
- Instituto de Biología y Medicina Experimental (IByME-CONICET), Vuelta de Obligado 2490, Buenos Aires, Argentina; Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Argentina.
| | - Fernanda Parborell
- Instituto de Biología y Medicina Experimental (IByME-CONICET), Vuelta de Obligado 2490, Buenos Aires, Argentina.
| | - Dalhia Abramovich
- Instituto de Biología y Medicina Experimental (IByME-CONICET), Vuelta de Obligado 2490, Buenos Aires, Argentina.
| |
Collapse
|
11
|
Woad KJ, Robinson RS. Luteal angiogenesis and its control. Theriogenology 2016; 86:221-8. [PMID: 27177965 DOI: 10.1016/j.theriogenology.2016.04.035] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 02/11/2016] [Accepted: 03/14/2016] [Indexed: 12/17/2022]
Abstract
Angiogenesis, the formation of new blood vessels from preexisting ones, is critical to luteal structure and function. In addition, it is a complex and tightly regulated process. Not only does rapid and extensive angiogenesis occur to provide the corpus luteum with an unusually high blood flow and support its high metabolic rate, but in the absence of pregnancy, the luteal vasculature must rapidly regress to enable the next cycle of ovarian activity. This review describes a number of key endogenous stimulatory and inhibitory factors, which act in a delicate balance to regulate luteal angiogenesis and ultimately luteal function. In vitro luteal angiogenesis cultures have demonstrated critical roles for fibroblast growth factor 2 (FGF2) in endothelial cell proliferation and sprouting, although other factors such as vascular endothelial growth factor A (VEGFA) and platelet-derived growth factor were important modulators in the control of luteal angiogenesis. Post-transcriptional regulation by small non-coding microRNAs is also likely to play a central role in the regulation of luteal angiogenesis. Appropriate luteal angiogenesis requires the coordinated activity of numerous factors expressed by several cell types at different times, and this review will also describe the role of perivascular pericytes and the importance of vascular maturation and stability. It is hoped that a better understanding of the critical processes underlying the transition from follicle to corpus luteum and subsequent luteal development will benefit the management of luteal function in the future.
Collapse
Affiliation(s)
- Kathryn J Woad
- School of Veterinary Medicine and Science, Sutton Bonington Campus, University of Nottingham, Leicestershire, UK.
| | - Robert S Robinson
- School of Veterinary Medicine and Science, Sutton Bonington Campus, University of Nottingham, Leicestershire, UK
| |
Collapse
|
12
|
Hall AP, Ashton S, Horner J, Wilson Z, Reens J, Richmond GHP, Barry ST, Wedge SR. PDGFR Inhibition Results in Pericyte Depletion and Hemorrhage into the Corpus Luteum of the Rat Ovary. Toxicol Pathol 2015; 44:98-111. [PMID: 26534939 DOI: 10.1177/0192623315613452] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The growth plate, ovary, adrenal gland, and rodent incisor tooth are sentinel organs for antiangiogenic effects since they respond reliably, quantitatively, and sensitively to inhibition of the vascular endothelial growth factor receptor (VEGFR). Here we report that treatment of rats with platelet-derived growth factor receptor beta (PDGFRβ) inhibitors that target pericytes results in severe ovarian hemorrhage with degeneration and eventual rupture of the corpus luteum. Evaluation of the growth plate, adrenal gland, and incisor tooth that are typical target organs for antiangiogenic treatment in the rodent revealed no abnormalities. Histologically, the changes in the ovary were characterized by sinusoidal dilatation, increased vessel fragility, and hemorrhage into the corpus luteum. Immunocytochemical staining of vessels with alpha smooth muscle actin and CD31 that recognize pericytes and vascular endothelium, respectively, demonstrated that this effect was due to selective pericyte deficiency within corpora lutea. Further experiments in which rats were treated concurrently with both PDGFRβ and VEGFR inhibitors ablated the hemorrhagic response, resulting instead in corpus luteum necrosis. These changes are consistent with the notion that selective pericyte loss in the primitive capillary network resulted in increased vessel fragility and hemorrhage, whereas concomitant VEGFR inhibition resulted in vessel regression and reduced vascular perfusion that restricted development of the hemorrhagic vessels. These results also highlight the utility of the rodent ovary to respond differentially to VEGFR and PDGFR inhibitors, which may provide useful information during routine safety assessment for determining target organ toxicity.
Collapse
Affiliation(s)
- Anthony P Hall
- AstraZeneca, Drug Safety and Metabolism, Alderley Park, Macclesfield, Cheshire, UK
| | - Susan Ashton
- AstraZeneca, Oncology iMed, Alderley Park, Macclesfield, Cheshire, UK
| | - Judith Horner
- AstraZeneca, Drug Safety and Metabolism, Alderley Park, Macclesfield, Cheshire, UK
| | - Zena Wilson
- AstraZeneca, Oncology iMed, Alderley Park, Macclesfield, Cheshire, UK
| | - Jaimini Reens
- AstraZeneca, Drug Safety and Metabolism, Alderley Park, Macclesfield, Cheshire, UK
| | | | - Simon T Barry
- AstraZeneca, Oncology iMed, Alderley Park, Macclesfield, Cheshire, UK
| | - Steve R Wedge
- AstraZeneca, Oncology iMed, Alderley Park, Macclesfield, Cheshire, UK
| |
Collapse
|
13
|
Pascuali N, Scotti L, Abramovich D, Irusta G, Di Pietro M, Bas D, Tesone M, Parborell F. Inhibition of platelet-derived growth factor (PDGF) receptor affects follicular development and ovarian proliferation, apoptosis and angiogenesis in prepubertal eCG-treated rats. Mol Cell Endocrinol 2015; 412:148-58. [PMID: 25937181 DOI: 10.1016/j.mce.2015.04.021] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 04/24/2015] [Accepted: 04/24/2015] [Indexed: 11/21/2022]
Abstract
The platelet-derived growth factor (PDGF) system is crucial for blood vessel stability. In the present study, we evaluated whether PDGFs play a critical intraovarian survival role in gonadotropin-dependent folliculogenesis. We examined the effect of intrabursal administration of a selective platelet-derived growth factor receptor (PDGFR) inhibitor (AG1295) on follicular development, proliferation, apoptosis and blood vessel formation and stability in ovaries from rats treated with equine chorionic gonadotropin (eCG). The percentages of preantral follicles (PAFs) and early antral follicles (EAFs) were lower in AG1295-treated ovaries than in control ovaries (p < 0.01-0.05). The percentage of atretic follicles (AtrFs) increased in AG1295-treated ovaries compared to control (p < 0.05). The ovarian weight and estradiol concentrations were lower in AG1295-treated ovaries than in the control group (p < 0.01 and p < 0.05, respectively), whereas progesterone concentrations did not change. AG1295 decreased the proliferation index in EAFs (p < 0.05) and increased the percentage of nuclei positive for cleaved caspase-3 and apoptotic DNA fragmentation (p < 0.01-0.05). AG1295 increased the expression of Bax (p < 0.05) without changes in the expression of Bcl-2 protein. AG1295-treated ovaries increased the cleavage of caspase-8 (p < 0.05) and decreased AKT and BAD phosphorylation compared with control ovaries (p < 0.05). AG1295 caused a decrease not only in the endothelial cell area but also in the area of pericytes and vascular smooth muscle cells (VSMCs) in the ovary (p < 0.05). Our findings suggest that the local inhibition of PDGFs causes an increase in ovarian apoptosis through an imbalance in the ratio of antiapoptotic to proapoptotic proteins, thus leading a larger number of follicles to atresia. PDGFs could exert their mechanism of action through an autocrine/paracrine effect on granulosa and theca cells mediated by PDGFRs. In conclusion, these data clearly indicate that the PDGF system is necessary for follicular development induced by gonadotropins.
Collapse
Affiliation(s)
- Natalia Pascuali
- Instituto de Biología y Medicina Experimental (IByME), CONICET, Buenos Aires, Argentina
| | - Leopoldina Scotti
- Instituto de Biología y Medicina Experimental (IByME), CONICET, Buenos Aires, Argentina
| | - Dalhia Abramovich
- Instituto de Biología y Medicina Experimental (IByME), CONICET, Buenos Aires, Argentina
| | - Griselda Irusta
- Instituto de Biología y Medicina Experimental (IByME), CONICET, Buenos Aires, Argentina
| | - Mariana Di Pietro
- Instituto de Biología y Medicina Experimental (IByME), CONICET, Buenos Aires, Argentina
| | - Diana Bas
- Instituto de Biología y Medicina Experimental (IByME), CONICET, Buenos Aires, Argentina
| | - Marta Tesone
- Instituto de Biología y Medicina Experimental (IByME), CONICET, Buenos Aires, Argentina; Departamento de Química Biológica, Facultad de Ciencias Exactas, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Fernanda Parborell
- Instituto de Biología y Medicina Experimental (IByME), CONICET, Buenos Aires, Argentina.
| |
Collapse
|
14
|
miR-140-5p inhibits ovarian cancer growth partially by repression of PDGFRA. Biomed Pharmacother 2015; 75:117-22. [PMID: 26297547 DOI: 10.1016/j.biopha.2015.07.035] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 07/26/2015] [Indexed: 12/15/2022] Open
Abstract
Dysregulation of miRNAs is a common feature in human cancers, and miR-140-5p has been found to be down-regulated in cancer. However its role in ovarian cancer remains unclear. miR-140-5p was underexpressed in HCC tissues and cell lines compared with their normal controls. Additionally, PDGFRA was predicted the target gene of miR-140-5p. PDGFRA was inversely correlated with the expression of miR-140-5p in ovarian cancer cells. Importantly, we demonstrate that the over expression of miR-140-5p significantly inhibits ovarian cancer cell proliferation and induces apoptosis. Our results suggest the existence of a novel miR-140-5p-PDGFRA pathway and indicate that miR-140-5p acts as a tumor suppressor during ovarian carcinogenesis. These results may provide a promising alterative strategy for the therapeutic treatment of ovarian cancer.
Collapse
|
15
|
Brito IR, Sales AD, Rodrigues GQ, Lobo CH, Castro SV, Silva AWB, Moura AAA, Silva JRV, Rodrigues APR, Figueiredo JR. Differential gene expression and immunolocalization of platelet-derived growth factors and their receptors in caprine ovaries. Domest Anim Endocrinol 2015; 51:46-55. [PMID: 25498237 DOI: 10.1016/j.domaniend.2014.10.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 10/02/2014] [Accepted: 10/04/2014] [Indexed: 11/20/2022]
Abstract
This study evaluated the messenger RNA (mRNA) expression and immunolocalization of all members of the platelet-derived growth factor (PDGF) family in caprine ovaries by quantitative PCR and immunohistochemistry, respectively. Detectable levels of PDGF-A mRNA were not observed in primordial follicles. Higher levels of PDGF-B mRNA were observed in primary follicles than in primordial follicles (P < 0.05). PDGF-D mRNA levels were higher in secondary follicles than in the other preantral follicle categories (P < 0.05). PDGF-B mRNA expression was higher than PDGF-C mRNA expression in primary follicles (P < 0.05). In antral follicles, PDGF-A mRNA expression was higher in cumulus-oocyte complexes (COCs) from small antral follicles than in those from large antral follicles and their respective granulosa/theca (GT) cells (P < 0.05). Furthermore, in COCs from small and large antral follicles, PDGF-A mRNA expression was higher than that of the other PDGF isoforms (P < 0.05). The mRNA levels of PDGF-B and PDGF-D and PDGFR-α and PDGFR-β were higher in GT cells from large antral follicles than in GT cells from small antral follicles and in their respective COCs (P < 0.05). In COCs and GT cells from small antral follicles, the mRNA levels of PDGFR-α were higher than those of PDGFR-β (P < 0.05). All proteins were observed in the cytoplasm of oocytes from all follicular categories. In granulosa cells, all PDGFs and PDGFR-β were detected from starting at the secondary stage, and in theca cells, all proteins, except PDGF-C, were detected starting at the antral stage. In conclusion, PDGF and its receptors are differentially expressed in the oocytes and ovarian cells according to the stage of follicular development, suggesting their role in the regulation of folliculogenesis in goats.
Collapse
Affiliation(s)
- I R Brito
- Laboratory of Manipulation of Oocyte and Preantral Follicles (LAMOFOPA), Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, CE, Brazil.
| | - A D Sales
- Laboratory of Manipulation of Oocyte and Preantral Follicles (LAMOFOPA), Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, CE, Brazil
| | - G Q Rodrigues
- Laboratory of Manipulation of Oocyte and Preantral Follicles (LAMOFOPA), Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, CE, Brazil
| | - C H Lobo
- Laboratory of Animal Physiology, Department of Animal Science, Federal University of Ceará, Fortaleza, CE, Brazil
| | - S V Castro
- Laboratory of Manipulation of Oocyte and Preantral Follicles (LAMOFOPA), Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, CE, Brazil
| | - A W B Silva
- Biotechnology Nucleus of Sobral (NUBIS), Federal University of Ceará, Sobral, CE, Brazil
| | - A A A Moura
- Laboratory of Animal Physiology, Department of Animal Science, Federal University of Ceará, Fortaleza, CE, Brazil
| | - J R V Silva
- Biotechnology Nucleus of Sobral (NUBIS), Federal University of Ceará, Sobral, CE, Brazil
| | - A P R Rodrigues
- Laboratory of Manipulation of Oocyte and Preantral Follicles (LAMOFOPA), Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, CE, Brazil
| | - J R Figueiredo
- Laboratory of Manipulation of Oocyte and Preantral Follicles (LAMOFOPA), Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, CE, Brazil
| |
Collapse
|
16
|
Peri LE, Koh BH, Ward GK, Bayguinov Y, Hwang SJ, Gould TW, Mullan CJ, Sanders KM, Ward SM. A novel class of interstitial cells in the mouse and monkey female reproductive tracts. Biol Reprod 2015; 92:102. [PMID: 25788664 DOI: 10.1095/biolreprod.114.124388] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 03/12/2015] [Indexed: 01/14/2023] Open
Abstract
Growing evidence suggests important roles for specialized platelet-derived growth factor receptor alpha-positive (PDGFRalpha(+)) cells in regulating the behaviors of visceral smooth muscle organs. Examination of the female reproductive tracts of mice and monkeys showed that PDGFRalpha(+) cells form extensive networks in ovary, oviduct, and uterus. PDGFRalpha(+) cells were located in discrete locations within these organs, and their distribution and density were similar in rodents and primates. PDGFRalpha(+) cells were distinct from smooth muscle cells and interstitial cells of Cajal (ICC). This was demonstrated with immunohistochemical techniques and by performing molecular expression studies on PDGFRalpha(+) cells from mice with enhanced green fluorescent protein driven off of the endogenous promoter for Pdgfralpha. Significant differences in gene expression were found in PDGFRalpha(+) cells from ovary, oviduct, and uterus. Differences in gene expression were also detected in cells from different tissue regions within the same organ (e.g., uterine myometrium vs. endometrium). PDGFRalpha(+) cells are unlikely to provide pacemaker activity because they lack significant expression of key pacemaker genes found in ICC (Kit and Ano1). Gja1 encoding connexin 43 was expressed at relatively high levels in PDGFRalpha(+) cells (except in the ovary), suggesting these cells can form gap junctions to one another and neighboring smooth muscle cells. PDGFRalpha(+) cells also expressed the early response transcription factor and proto-oncogene Fos, particularly in the ovary. These data demonstrate extensive distribution of PDGFRalpha(+) cells throughout the female reproductive tract. These cells are a heterogeneous population of cells that are likely to contribute to different aspects of physiological regulation in the various anatomical niches they occupy.
Collapse
Affiliation(s)
- Lauren E Peri
- Department of Physiology & Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Byoung H Koh
- Department of Physiology & Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Grace K Ward
- Department of Physiology & Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Yulia Bayguinov
- Department of Physiology & Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Sung Jin Hwang
- Department of Physiology & Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Thomas W Gould
- Department of Physiology & Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Catrina J Mullan
- Department of Physiology & Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Kenton M Sanders
- Department of Physiology & Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Sean M Ward
- Department of Physiology & Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| |
Collapse
|
17
|
Somavilla AL, Sonstegard TS, Higa RH, Rosa AN, Siqueira F, Silva LOC, Torres Júnior RAA, Coutinho LL, Mudadu MA, Alencar MM, Regitano LCA. A genome-wide scan for selection signatures in Nellore cattle. Anim Genet 2014; 45:771-81. [DOI: 10.1111/age.12210] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/15/2014] [Indexed: 11/27/2022]
Affiliation(s)
- A. L. Somavilla
- Programa de Pós-Graduação em Genética e Melhoramento Animal; UNESP/FCAV; Jaboticabal Brasil
| | - T. S. Sonstegard
- Bovine Functional Genomics Laboratory; ANRI; USDA-ARS; Beltsville MD USA
| | - R. H. Higa
- Embrapa Informática Agropecuária; Campinas Brasil
| | - A. N. Rosa
- Embrapa Gado de Corte; Campo Grande Brasil
| | | | | | | | - L. L. Coutinho
- Escola Superior de Agricultura Luiz de Queiroz (Esalq); USP; Piracicaba Brasil
- CNPq Fellow; Brasília Distrito Federal Brasil
| | | | - M. M. Alencar
- Embrapa Pecuária Sudeste; São Carlos Brasil
- CNPq Fellow; Brasília Distrito Federal Brasil
| | - L. C. A. Regitano
- Embrapa Pecuária Sudeste; São Carlos Brasil
- CNPq Fellow; Brasília Distrito Federal Brasil
| |
Collapse
|
18
|
Macchiarelli G, Palmerini MG, Nottola SA, Cecconi S, Tanemura K, Sato E. Restoration of corpus luteum angiogenesis in immature hypothyroid rdw rats after thyroxine treatment: morphologic and molecular evidence. Theriogenology 2012; 79:116-26. [PMID: 23122683 DOI: 10.1016/j.theriogenology.2012.09.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Revised: 09/16/2012] [Accepted: 09/17/2012] [Indexed: 01/23/2023]
Abstract
Thyroxine (T4) plus gonadotropins might stimulate ovarian follicular angiogenesis in immature infertile hypothyroid rdw rats by upregulating mRNA expression of major angiogenic factors. Development of growing corpus luteum (CL) is strongly related to angiogenesis and to morphofunctional development of microcirculation. Our aim was to investigate if T4 is involved in CL angiogenesis and in the activation of capillary cells and angiogenic factors after ovulation in a spontaneous model of hypothyroidism, the rdw rat. Rdw rats were treated with T4 plus gonadotropins (equine chorionic gonadotropin plus human chorionic gonadotropin; eCG+hCG) or gonadotropins alone in order to evaluate the effects of T4 on early luteal angiogenesis, on microvascular cells and on expression of major growth factors which are involved in the regulation of angiogenesis. Wistar-Imamichi rats treated with gonadotropins were used as controls. The ovaries were collected 4 days after hCG administration and analyzed using morphologic and molecular approaches. Thyroxine plus gonadotropins stimulated the growth of CLs and follicles as in controls, differently from rdw rats treated only with gonadotropins, in which CLs were not found and only small follicles, often atretic, could be recognized. In T4 plus gonadotropin-treated rdw rats CLs showed increased microvasculature, numerous activated capillaries characterized by sprouting and other angiogenic figures, and associated pericytes. Quantitative analysis revealed that the number of pericytes in T4 plus gonadotropin-treated rdw rats was comparable with that found in control rats and was significantly higher than that found in gonadotropin-treated rdw rats. The mRNA expression of vascular endothelial growth factor and basic fibroblast growth factor was significantly higher in control rats and in T4 plus gonadotropin-treated rdw rats than in gonadotropin-treated rdw rats. mRNA expression of tumor necrosis factor α, transforming growth factor β, and epidermal growth factor did not show significant changes. Our data originally demonstrated that T4 promoted the growth of an active microcirculation in developing CLs of gonadotropin-primed hypothyroid rdw rats, mainly by inducing sprouting angiogenesis, pericyte recruitment, and upregulation of mRNA expression of vascular endothelial growth factor and basic fibroblast growth factor. In conclusion, we suggest that T4 plays a key role in restoring luteal angiogenesis in ovaries of immature hypothyroid rdw rats.
Collapse
Affiliation(s)
- Guido Macchiarelli
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy.
| | | | | | | | | | | |
Collapse
|
19
|
Coburn AM, Cappon GD, Bowman CJ, Stedman DB, Patyna S. Reproductive Toxicity Assessment of Sunitinib, A Multitargeted Receptor Tyrosine Kinase Inhibitor, in Male and Female Rats. ACTA ACUST UNITED AC 2012; 95:267-75. [DOI: 10.1002/bdrb.21012] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Accepted: 02/27/2012] [Indexed: 11/11/2022]
Affiliation(s)
| | | | | | | | - Shem Patyna
- Pfizer Worldwide Research and Development; San Diego; CA
| |
Collapse
|
20
|
Brito IR, Lima IMT, Saraiva MVA, Silva CMG, Magalhães-Padilha DM, Araújo VR, Barreto Luz V, Barbalho Silva AW, Campello CC, Silva JRV, Figueiredo JR. Expression Levels of mRNA-Encoding PDGF Receptors in Goat Ovaries and the Influence of PDGF on the In Vitro Development of Caprine Pre-Antral Follicles. Reprod Domest Anim 2011; 47:695-703. [DOI: 10.1111/j.1439-0531.2011.01946.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
21
|
Turner EC, Hughes J, Wilson H, Clay M, Mylonas KJ, Kipari T, Duncan WC, Fraser HM. Conditional ablation of macrophages disrupts ovarian vasculature. Reproduction 2011; 141:821-31. [PMID: 21393340 PMCID: PMC3101494 DOI: 10.1530/rep-10-0327] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2010] [Revised: 02/11/2011] [Accepted: 03/10/2011] [Indexed: 11/26/2022]
Abstract
Macrophages are the most abundant immune cell within the ovary. Their dynamic distribution throughout the ovarian cycle and heterogenic array of functions suggest the involvement in various ovarian processes, but their functional role has yet to be fully established. The aim was to induce conditional macrophage ablation to elucidate the putative role of macrophages in maintaining the integrity of ovarian vasculature. Using the CD11b-diphtheria toxin receptor (DTR) mouse, in which expression of human DTR is under the control of the macrophage-specific promoter sequence CD11b, ovarian macrophages were specifically ablated in adult females by injections of diphtheria toxin (DT). CD11b-DTR mice were given DT treatment or vehicle and ovaries collected at 2, 8, 16, 24 and 48 h. Histochemical stains were employed to characterise morphological changes, immunohistochemistry for F4/80 to identify macrophages and the endothelial cell marker CD31 used to quantify vascular changes. In normal ovaries, macrophages were detected in corpora lutea and in the theca layer of healthy and atretic follicles. As macrophage ablation progressed, increasing amounts of ovarian haemorrhage were observed affecting both luteal and thecal tissue associated with significant endothelial cell depletion, increased erythrocyte accumulation and increased follicular atresia by 16 h. These events were followed by necrosis and profound structural damage. Changes were limited to the ovary, as DT treatment does not disrupt the vasculature of other tissues likely reflecting the unique cyclical nature of the ovarian vasculature and heterogeneity between macrophages within different tissues. These results show that macrophages play a critical role in maintaining ovarian vascular integrity.
Collapse
Affiliation(s)
- Emily C Turner
- MRC Human Reproductive Sciences Unit, Queen's Institute of Medical Research, Centre for Reproductive BiologyUniversity of Edinburgh47 Little France Crescent, Edinburgh, EH16 4TJUK
| | - Jeremy Hughes
- MRC Centre for Inflammation ResearchUniversity of Edinburgh47 Little France Crescent, Edinburgh, EH16 4TJUK
| | - Helen Wilson
- MRC Human Reproductive Sciences Unit, Queen's Institute of Medical Research, Centre for Reproductive BiologyUniversity of Edinburgh47 Little France Crescent, Edinburgh, EH16 4TJUK
| | - Michael Clay
- MRC Centre for Inflammation ResearchUniversity of Edinburgh47 Little France Crescent, Edinburgh, EH16 4TJUK
| | - Katie J Mylonas
- MRC Centre for Inflammation ResearchUniversity of Edinburgh47 Little France Crescent, Edinburgh, EH16 4TJUK
| | - Tiina Kipari
- MRC Centre for Inflammation ResearchUniversity of Edinburgh47 Little France Crescent, Edinburgh, EH16 4TJUK
| | - W Colin Duncan
- Obstetrics and Gynaecology, Division of Reproductive and Developmental SciencesUniversity of Edinburgh47 Little France Crescent, Edinburgh, EH16 4TJUK
| | - Hamish M Fraser
- MRC Human Reproductive Sciences Unit, Queen's Institute of Medical Research, Centre for Reproductive BiologyUniversity of Edinburgh47 Little France Crescent, Edinburgh, EH16 4TJUK
| |
Collapse
|
22
|
Kuo SW, Ke FC, Chang GD, Lee MT, Hwang JJ. Potential role of follicle-stimulating hormone (FSH) and transforming growth factor (TGFβ1) in the regulation of ovarian angiogenesis. J Cell Physiol 2011; 226:1608-19. [DOI: 10.1002/jcp.22491] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
23
|
Hwu YM, Li SH, Lee RKK, Lin MH, Tsai YH, Yeh TS. Luteinizing hormone increases platelet-derived growth factor-D gene expression in human granulosa–luteal cells. Fertil Steril 2009; 92:2065-8. [DOI: 10.1016/j.fertnstert.2009.05.064] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2009] [Revised: 05/19/2009] [Accepted: 05/27/2009] [Indexed: 11/15/2022]
|
24
|
Robinson RS, Woad KJ, Hammond AJ, Laird M, Hunter MG, Mann GE. Angiogenesis and vascular function in the ovary. Reproduction 2009; 138:869-81. [DOI: 10.1530/rep-09-0283] [Citation(s) in RCA: 181] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Ovarian function is dependent on the establishment and continual remodelling of a complex vascular system. This enables the follicle and/or corpus luteum (CL) to receive the required supply of nutrients, oxygen and hormonal support as well as facilitating the release of steroids. Moreover, the inhibition of angiogenesis results in the attenuation of follicular growth, disruption of ovulation and drastic effects on the development and function of the CL. It appears that the production and action of vascular endothelial growth factor A (VEGFA) is necessary at all these stages of development. However, the expression of fibroblast growth factor 2 (FGF2) in the cow is more dynamic than that of VEGFA with a dramatic upregulation during the follicular–luteal transition. This upregulation is then likely to initiate intense angiogenesis in the presence of high VEGFA levels. Recently, we have developed a novel ovarian physiological angiogenesis culture system in which highly organised and intricate endothelial cell networks are formed. This system will enable us to elucidate the complex inter-play between FGF2 and VEGFA as well as other angiogenic factors in the regulation of luteal angiogenesis. Furthermore, recent evidence indicates that pericytes might play an active role in driving angiogenesis and highlights the importance of pericyte–endothelial interactions in this process. Finally, the targeted promotion of angiogenesis may lead to the development of novel strategies to alleviate luteal inadequacy and infertility.
Collapse
|
25
|
Woad KJ, Hammond AJ, Hunter M, Mann GE, Hunter MG, Robinson RS. FGF2 is crucial for the development of bovine luteal endothelial networks in vitro. Reproduction 2009; 138:581-8. [DOI: 10.1530/rep-09-0030] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The development of the corpus luteum requires angiogenesis, and involves the complex interplay between factors such as vascular endothelial growth factor A (VEGFA), fibroblast growth factor 2 (FGF2) and platelet-derived growth factor (PDGF). However, the relative role of these factors remains to be elucidated. This study used a new physiologically relevant mixed luteal cell culture system to test the hypotheses that: a) FGF2 and VEGFA are critical for bovine luteal angiogenesis; and b) local luteal PDGF signalling stimulates the formation of endothelial networks. Cells were treated with receptor tyrosine kinase inhibitors against VEGFA (SU1498), FGF2 (SU5402) or PDGF (AG1295) activity. After 9 days in culture, endothelial cells were immunostained for von Willebrand factor (VWF) and quantified by image analysis. Highly organised intricate endothelial networks were formed in the presence of exogenous VEGFA and FGF2. The inhibition of FGF2 activity reduced the total area of VWF staining versus controls (>95%; P<0.001). Inhibition of VEGF and PDGF activity reduced the endothelial network formation by more than 60 and 75% respectively (P<0.05). Progesterone production increased in all treatments from day 1 to 7 (P<0.001), and was unaffected by FGF2 or PDGF receptor kinase inhibition (P>0.05), but was reduced by the VEGF receptor inhibitor on days 5 and 7 (P<0.001). In conclusion, this study confirmed that VEGF signalling regulates both bovine luteal angiogenesis and progesterone production. However, FGF2 was crucial for luteal endothelial network formation. Also, for the first time, this study showed that local luteal PDGF activity regulates bovine luteal endothelial network formation in vitro.
Collapse
|
26
|
Fraser HM, Duncan WC. SRB Reproduction, Fertility and Development Award Lecture 2008. Regulation and manipulation of angiogenesis in the ovary and endometrium. Reprod Fertil Dev 2009; 21:377-92. [PMID: 19261215 DOI: 10.1071/rd08272] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2008] [Accepted: 01/19/2009] [Indexed: 12/27/2022] Open
Abstract
The marked cyclical physiological angiogenesis in the developing follicle, corpus luteum and endometrium implies a critical role in health and disease. Our approach to understanding its regulation has been to localise and quantify the temporal changes in putative angiogenic factors, and their receptors, in human and non-human primate tissue and to use antagonists to dissect their role by specific inhibition at defined periods during the ovulatory cycle in non-human primates in vivo. The course of angiogenesis throughout the cycle and the cellular and molecular effects of inhibitory treatments have been investigated in the marmoset ovary and uterus, whereas consequences on pituitary-ovarian function have been monitored in macaques. Inhibition of vascular endothelial growth factor (VEGF) at the time of follicle recruitment or selection prevents endothelial cell proliferation, leading to inhibition of follicular development. VEGF inhibition during the early luteal phase prevents angiogenesis and restricts development of the luteal microvasculature. Inhibition of angiogenesis at all stages of the cycle leads to profound suppression of ovarian function. Even during the 'post-angiogenic' period of the luteal phase, inhibition of VEGF precipitates a suppression of progesterone secretion, pointing to additional roles for VEGF in the ovary. In the endometrium, oestrogen drives endometrial angiogenesis through VEGF. Thus, oestrogen can restore angiogenesis after ovariectomy, but not in the presence of VEGF inhibitors. These investigations enhance our understanding of the regulation of angiogenesis in the ovary and uterus and inform studies on conditions with abnormal vascularisation, such as polycystic ovary syndrome, endometriosis, uterine fibroids and menstrual dysfunction.
Collapse
Affiliation(s)
- Hamish M Fraser
- MRC Human Reproductive Sciences Unit, Centre for Reproductive Biology, Queen's Institute of Medical Research, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.
| | | |
Collapse
|
27
|
YOSHIDA M, WATANABE G, SUZUKI T, INOUE K, TAKAHASHI M, MAEKAWA A, TAYA K, NISHIKAWA A. Long-Term Treatment with Bromocriptine Inhibits Endometrial Adenocarcinoma Development in Rats. J Reprod Dev 2009; 55:105-9. [DOI: 10.1262/jrd.20026] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Midori YOSHIDA
- Division of Pathology, National Institute of Health Sciences
| | - Gen WATANABE
- Veterinary Physiology, Tokyo University of Agriculture and Technology
| | | | - Kaoru INOUE
- Division of Pathology, National Institute of Health Sciences
| | - Miwa TAKAHASHI
- Division of Pathology, National Institute of Health Sciences
| | - Akihiko MAEKAWA
- Safety Assessment Division, Chemical Management Center, National Institute of Technology and Evaluation
| | - Kazuyoshi TAYA
- Veterinary Physiology, Tokyo University of Agriculture and Technology
| | | |
Collapse
|
28
|
Abstract
Angiogenesis is important for the formation and development of the corpus luteum and for maintenance of luteal function. Blood vessel regression is an important physiological phenomenon in the corpus luteum, which is associated with tissue involution during structural luteolysis. Angiogenesis actively occurs during the early luteal phase and is completed by the mid-luteal phase. Perivascular cells (pericytes) increase in number from the early luteal phase to the mid-luteal phase, suggesting that blood vessels are gradually stabilized until the mid-luteal phase. In the corpus luteum undergoing luteolysis, blood vessels and pericytes decrease in number, which is related to structural involution. In the corpus luteum of early pregnancy, the number of blood vessels with pericytes increases, suggesting that angiogenesis occurs again, accompanied by blood vessel stabilization. These changes in vasculature of the corpus luteum are regulated by the collaboration with vascular endothelial growth factor, which is involved in proliferation of vascular endothelial cells, and angiopoietins, which are involved in stabilization of blood vessels. This review focuses on angiogenesis, blood vessel stabilization and blood vessel regression during the divergent phases of luteal formation, luteal regression and luteal rescue by pregnancy. (Reprod Med Biol 2008; 7: 91-103).
Collapse
Affiliation(s)
- Norihiro Sugino
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Aki Matsuoka
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Ken Taniguchi
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Hiroshi Tamura
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Ube, Japan
| |
Collapse
|