1
|
Mi X, Chen C, Feng C, Qin Y, Chen ZJ, Yang Y, Zhao S. The Functions and Application Prospects of Hepatocyte Growth Factor in Reproduction. Curr Gene Ther 2024; 24:347-355. [PMID: 39005061 DOI: 10.2174/0115665232291010240221104445] [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: 11/05/2023] [Revised: 02/01/2024] [Accepted: 02/13/2024] [Indexed: 07/16/2024]
Abstract
Hepatocyte growth factor (HGF) is expressed in multiple systems and mediates a variety of biological activities, such as mitosis, motility, and morphogenesis. A growing number of studies have revealed the expression patterns and functions of HGF in ovarian and testicular physiology from the prenatal to the adult stage. HGF regulates folliculogenesis and steroidogenesis by modulating the functions of theca cells and granulosa cells in the ovary. It also mediates somatic cell proliferation and steroidogenesis, thereby affecting spermatogenesis in males. In addition to its physiological effects on the reproductive system, HGF has shown advantages in preclinical studies over recent years for the treatment of male and female infertility, particularly in women with premature ovarian insufficiency. This review aims to summarize the pleiotropic functions of HGF in the reproductive system and to provide prospects for its clinical application.
Collapse
Affiliation(s)
- Xin Mi
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, Shandong, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, Shandong, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
| | - Caiyi Chen
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, Shandong, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, Shandong, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
| | - Chen Feng
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, Shandong, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, Shandong, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
| | - Yingying Qin
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, Shandong, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, Shandong, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
| | - Zi-Jiang Chen
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, Shandong, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, Shandong, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
- Research Unit of Gametogenesis and Health of ART-Offspring (No.2021RU001), Chinese Academy of Medical Sciences, Jinan, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yajuan Yang
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, Shandong, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, Shandong, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
| | - Shidou Zhao
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, Shandong, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, Shandong, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
| |
Collapse
|
2
|
Park H, Lee DH, You JH, Seok J, Lim JY, Kim GJ. Increased Hepatocyte Growth Factor Secretion by Placenta-Derived Mesenchymal Stem Cells Improves Ovarian Function in an Ovariectomized Rat Model via Vascular Remodeling by Wnt Signaling Activation. Cells 2023; 12:2708. [PMID: 38067136 PMCID: PMC10705748 DOI: 10.3390/cells12232708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/16/2023] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
The vascular network contributes to the development of follicles. However, the therapeutic mechanism between vascular remodeling and ovarian functions is still unclear. Therefore, we demonstrated whether increased HGF by placenta-derived mesenchymal stem cells (PD-MSCs) improves ovarian function in an ovariectomized rat model via vascular remodeling by Wnt signaling activation. We established a half-ovariectomized rat model in which damaged ovaries were induced by ovariectomy of half of each ovary, and PD-MSCs (5 × 105 cells) were transplanted by intravenous injection. Three weeks after transplantation, rats in all groups were sacrificed. We examined the secretion of HGF by PD-MSCs through culture medium. The vascular structure in injured ovarian tissues was restored to a greater extent in the PD-MSC transplantation (Tx) group than in the nontransplantation (NTx) group (* p < 0.05). The expression of genes related to Wnt signaling (e.g., LRP6, GSK3β, β-catenin) was significantly increased in the Tx group compared to the NTx group (* p < 0.05). However, the expression of genes related to vascular permeability (e.g., Asef, ERG3) was significantly decreased in the Tx group compared to the NTx group (* p < 0.05). Follicular development was improved in the Tx group compared to the NTx group (* p < 0.05). Furthermore, to evaluate vascular function, we cocultivated PD-MSCs after human umbilical vein endothelial cells (HUVECs) with lipopolysaccharide (LPS), and we analyzed the vascular formation assay and dextran assay in HUVECs. Cocultivation of PD-MSCs with injured HUVECs enhanced vascular formation and decreased endothelial cell permeability (* p < 0.05). Also, cocultivation of PD-MSCs with explanted ovarian tissues improved follicular maturation compared to cocultivation of the Wnt inhibitor-treated PD-MSCs with explanted ovarian tissues. Therefore, HGF secreted by PD-MSCs improved ovarian function in rats with ovarian dysfunction by decreasing vascular permeability via Wnt signaling.
Collapse
Affiliation(s)
- Hyeri Park
- Department of Bioinspired Science, CHA University, Seongnam-si 13488, Gyeonggi-do, Republic of Korea
- PLABiologics Co., Ltd., Seongnam-si 13522, Gyeonggi-do, Republic of Korea
| | - Dae Hyun Lee
- Department of Bioinspired Science, CHA University, Seongnam-si 13488, Gyeonggi-do, Republic of Korea
- PLABiologics Co., Ltd., Seongnam-si 13522, Gyeonggi-do, Republic of Korea
| | - Jun Hyeong You
- Department of Bioinspired Science, CHA University, Seongnam-si 13488, Gyeonggi-do, Republic of Korea
| | - Jin Seok
- Department of Bioinspired Science, CHA University, Seongnam-si 13488, Gyeonggi-do, Republic of Korea
| | - Ja-Yun Lim
- Department of Clinical Laboratory Science, Hyejeon College, Hongsung-gun 32244, Chungnam-do, Republic of Korea
| | - Gi Jin Kim
- Department of Bioinspired Science, CHA University, Seongnam-si 13488, Gyeonggi-do, Republic of Korea
- PLABiologics Co., Ltd., Seongnam-si 13522, Gyeonggi-do, Republic of Korea
| |
Collapse
|
3
|
Jiao W, Mi X, Yang Y, Liu R, Liu Q, Yan T, Chen ZJ, Qin Y, Zhao S. Mesenchymal stem cells combined with autocrosslinked hyaluronic acid improve mouse ovarian function by activating the PI3K-AKT pathway in a paracrine manner. Stem Cell Res Ther 2022; 13:49. [PMID: 35109928 PMCID: PMC8812195 DOI: 10.1186/s13287-022-02724-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 12/27/2021] [Indexed: 12/14/2022] Open
Abstract
Background Declining ovarian function in advance-aged women and in premature ovarian insufficiency (POI) patients seriously affects quality of life, and there is currently no effective treatment to rescue ovarian function in clinic. Stem cell transplantation is a promising therapeutic strategy for ovarian aging, but its clinical application is limited due to the low efficiency and unclear mechanism. Here, a novel combination of umbilical cord-mesenchymal stem cells (UC-MSCs) and autocrosslinked hyaluronic acid (HA) gel is explored to rescue ovarian reserve and fecundity in POI and naturally aging mice. Methods To investigate HA prolonged the survival after UC-MSCs transplantation, PCR and immunofluorescence were performed to track the cells on day 1, 3, 7 and 14 after transplantation. The effects of HA on UC-MSCs were analyzed by CCK8 assay, RNA-sequencing and 440 cytokine array. In vivo experiments were conducted to evaluate the therapeutic effects of UC-MSCs combined with HA transplantation in 4-vinylcyclohexene diepoxide (VCD)-induced POI mice and naturally aging mice model. Ovarian function was analyzed by ovarian morphology, follicle counts, estrous cycle, hormone levels and fertility ability. To investigate the mechanisms of stem cell therapy, conditioned medium was collected from UC-MSCs and fibroblast. Both in vitro ovarian culture model and 440 cytokine array were applied to assess the paracrine effect and determine the underlying mechanism. Hepatocyte growth factor (HGF) was identified as an effective factor and verified by HGF cytokine/neutralization antibody supplementation into ovarian culture system. Results HA not only prolongs the retention of UC-MSCs in the ovary, but also boosts their secretory function, and UC-MSCs promote follicular survival by activating the PI3K-AKT pathway through a paracrine mechanism both in vitro and in vivo. More importantly, HGF is identified as the key functional cytokine secreted by MSCs. Conclusions The results show that HA is an excellent cell scaffold to improve the treatment efficiency of UC-MSCs for ovarian aging under both physiological and pathological conditions, and the therapeutic mechanism is through activation of the PI3K-AKT pathway via HGF. These findings will facilitate the clinical application of MSCs transplantation for ovarian disorders. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02724-3.
Collapse
Affiliation(s)
- Wenlin Jiao
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China.,Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Xin Mi
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China.,Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Yajuan Yang
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China.,Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Ran Liu
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China.,Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Qiang Liu
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China.,Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Tao Yan
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China.,Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China.,Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, China.,Center for Reproductive Medicine, School of Medicine, Ren Ji Hospital, Shanghai Jiao Tong University, 200135, Shanghai, China
| | - Yingying Qin
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China.,Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Shidou Zhao
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China. .,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China. .,Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China. .,Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China. .,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
4
|
Mi X, Jiao W, Yang Y, Qin Y, Chen ZJ, Zhao S. HGF Secreted by Mesenchymal Stromal Cells Promotes Primordial Follicle Activation by Increasing the Activity of the PI3K-AKT Signaling Pathway. Stem Cell Rev Rep 2022; 18:1834-1850. [PMID: 35089464 PMCID: PMC9209380 DOI: 10.1007/s12015-022-10335-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/14/2022] [Indexed: 01/08/2023]
Abstract
Primordial follicle activation is fundamental for folliculogenesis and for the maintenance of fertility. An effective therapeutic strategy for patients with premature ovarian insufficiency (POI) is to promote the activation of residual primordial follicles. The secretome of human umbilical cord mesenchymal stromal cells (hUC-MSC-sec) contains several components that might promote the activation of primordial follicles. In the present study, we revealed that treatment with the hUC-MSC-sec significantly increased the proportion of activated primordial follicles in mouse ovaries both in vitro and in vivo. The activating effects of hUC-MSC-sec on primordial follicles were attributed to the activation of the PI3K-AKT signaling pathway by hepatocyte growth factor (HGF). While the effect of the hUC-MSC-sec was attenuated by the neutralizing antibodies against HGF, application of exogenous HGF alone also promoted the activation of primordial follicles. Furthermore, we demonstrated that HGF promoted the expression of KITL in granulosa cells by binding with the HGF receptor c-Met, thereby increasing the activity of the PI3K-AKT signaling pathway to activate primordial follicles. Taken together, our findings demonstrate that hUC-MSC-sec promotes primordial follicle activation through the functional component HGF to increase the PI3K-AKT signaling activity, highlighting the application of the hUC-MSC-sec or HGF for the treatment of POI patients.
Collapse
Affiliation(s)
- Xin Mi
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China.,Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Wenlin Jiao
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China.,Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Yajuan Yang
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China.,Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Yingying Qin
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China.,Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China.,Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, China.,Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, China
| | - Shidou Zhao
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, Shandong, China. .,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China. .,Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China. .,Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China. .,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
5
|
Abstract
Advanced maternal age is associated with the natural oocyte depletion, leading to low oocyte yield, high infertility treatment cancellation rates, and eventual decreases in pregnancy rates. Various innovative interventions have been introduced to improve the outcome of infertility treatment for aging patients. Numerous published data demonstrated that early follicle development was regulated by intraovarian growth factors through autocrine or paracrine mechanisms. Platelet-rich plasma (PRP), a plasma fraction of peripheral blood with a high concentration of platelets, has been implemented in regenerative medicine in the last decade. The plasma contains a variety of growth factors that were suggested to be able to enhance angiogenesis regeneration and the cell proliferation process. The initial report showed that an intraovarian injection of PRP improved the hormonal profile and increased the number of retrieved oocytes in patients with diminished ovarian reserve. Subsequently, several studies with larger sample sizes have reported that this approach resulted in several healthy live births with no apparent complications. However, the use of ovarian PRP treatment needs to be fully investigated, because no randomized controlled trial has yet been performed to confirm its efficacy.
Collapse
|
6
|
Wang DH, Ren J, Zhou CJ, Han Z, Wang L, Liang CG. Supplementation with CTGF, SDF1, NGF, and HGF promotes ovine in vitro oocyte maturation and early embryo development. Domest Anim Endocrinol 2018; 65:38-48. [PMID: 29890304 DOI: 10.1016/j.domaniend.2018.05.003] [Citation(s) in RCA: 12] [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: 09/23/2017] [Revised: 04/14/2018] [Accepted: 05/02/2018] [Indexed: 02/03/2023]
Abstract
The strategies for improving the in vitro maturation (IVM) of domestic animal oocytes focus on promoting nuclear and cytoplasmic maturation. The identification of paracrine factors and their supplementation in the culture medium represent effective approaches for oocyte maturation and embryo development. This study investigated the effects of paracrine factor supplementation including connective tissue growth factor (CTGF), nerve growth factor (NGF), hepatocyte growth factor (HGF), and stromal derived factor 1 (SDF1) on ovine oocytes and early parthenogenetic embryos using an in vitro culture system. First, we identified the optimal concentrations of CTGF (30 ng/mL), SDF1 (10 ng/mL), NGF (3 ng/mL), and HGF (100 ng/mL) for promoting oocyte maturation, which combined, induced nuclear maturation in 94.19% of oocytes. This combination also promoted cumulus cell expansion and inhibited oocyte/cumulus apoptosis, while enabling a larger proportion (33.04%) of embryos to develop into blastocysts than in the controls and prevented embryo apoptosis. These novel findings demonstrate that the paracrine factors CTGF, SDF1, NGF, and HGF facilitate ovine oocyte and early parthenogenetic embryo development in vitro. Thus, supplementation with these factors may help optimize the IVM of ovine oocytes and early parthenogenetic embryo development strategies.
Collapse
Affiliation(s)
- D H Wang
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, The Research Center for Laboratory Animal Science, College of Life Science, Inner Mongolia University, Hohhot, Inner Mongolia, People's Republic of China
| | - J Ren
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, The Research Center for Laboratory Animal Science, College of Life Science, Inner Mongolia University, Hohhot, Inner Mongolia, People's Republic of China
| | - C J Zhou
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, The Research Center for Laboratory Animal Science, College of Life Science, Inner Mongolia University, Hohhot, Inner Mongolia, People's Republic of China
| | - Z Han
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, The Research Center for Laboratory Animal Science, College of Life Science, Inner Mongolia University, Hohhot, Inner Mongolia, People's Republic of China
| | - L Wang
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, The Research Center for Laboratory Animal Science, College of Life Science, Inner Mongolia University, Hohhot, Inner Mongolia, People's Republic of China
| | - C G Liang
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, The Research Center for Laboratory Animal Science, College of Life Science, Inner Mongolia University, Hohhot, Inner Mongolia, People's Republic of China.
| |
Collapse
|
7
|
Mesenchymal Stem Cells in Restoration of Fertility at Experimental Pelvic Inflammatory Disease. Stem Cells Int 2017; 2017:2014132. [PMID: 28928773 PMCID: PMC5591961 DOI: 10.1155/2017/2014132] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 06/15/2017] [Accepted: 07/17/2017] [Indexed: 12/17/2022] Open
Abstract
Inflammatory disorders account for a significant percentage of gynecologic diseases, particularly in women of reproductive age. It is known that stem cells have anti-inflammatory and regenerative properties. Based on this, we investigated the effect of intravenous administration of cryopreserved mesenchymal stem cells (cMSCs) of bone marrow on experimental chronic inflammation of the ovaries. The paper shows that on the 21st day after cMSC therapy, leukocyte infiltration of ovaries was slightly relative to the control group without treatment, and the ratio of developing and atretic follicles in the animals with cMSC injection dramatically increased, while in the control, it still remained on the side of atretic forms. The number of apoptotic oocytes after stimulation of superovulation in the control group was significantly higher (85.3 ± 5.2%) than that in the animals with therapy (5.7 ± 0.8%). Relative number of fertilized eggs in the group with cMSC therapy was higher by 40% compare to that in the control. Pregnancy rate in natural estrous cycle after cell administration increased by 20%, and average number of litters in this group was two times significantly higher than that in the control. So the intravenous injection of cMSCs has the restorative effect on the fertility at experimental pelvic inflammatory disease.
Collapse
|
8
|
Li J, Mao Q, He J, She H, Zhang Z, Yin C. Human umbilical cord mesenchymal stem cells improve the reserve function of perimenopausal ovary via a paracrine mechanism. Stem Cell Res Ther 2017; 8:55. [PMID: 28279229 PMCID: PMC5345137 DOI: 10.1186/s13287-017-0514-5] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 01/05/2017] [Accepted: 02/18/2017] [Indexed: 01/20/2023] Open
Abstract
Background Human umbilical cord mesenchymal stem cells (hUCMSCs) are a type of pluripotent stem cell which are isolated from the umbilical cord of newborns. hUCMSCs have great therapeutic potential. We designed this experimental study in order to investigate whether the transplantation of hUCMSCs can improve the ovarian reserve function of perimenopausal rats and delay ovarian senescence. Method We selected naturally aging rats confirmed by vaginal smears as models of perimenopausal rats, divided into the control group and the treatment group, and selected young fertile female rats as normal controls. hUCMSCs were transplanted into rats of the treatment group through tail veins. Enzyme-linked immunosorbent assay (ELISA) detected serum levels of sex hormones, H&E staining showed ovarian tissue structure and allowed follicle counting, immunohistochemistry and western blot analysis revealed ovarian expression of hepatocyte growth factor (HGF), vascular endothelial cell growth factor (VEGF), and insulin-like growth factor-1 (IGF-1), polymerase chain reaction (PCR) and western blot analysis revealed hUCMSCs expression of HGF, VEGF, and IGF-1. Results At time points of 14, 21, and 28 days after hUCMSCs transplantation, estradiol (E2) and anti-Müllerian hormone (AMH) increased while follicle-stimulating hormone (FSH) decreased; ovarian structure improved and follicle number increased; ovarian expression of HGF, VEGF, and IGF-1 protein elevated significantly. Meanwhile, PCR and western blot analysis indicated hUCMSCs have the capacity of secreting HGF, VEGF, and IGF-1 cytokines. Conclusions Our results suggest that hUCMSCs can promote ovarian expression of HGF, VEGF, and IGF-1 through secreting those cytokines, resulting in improving ovarian reserve function and withstanding ovarian senescence.
Collapse
Affiliation(s)
- Jia Li
- Department of Obstetrics and Gynecology, Graduate College, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China.,Department of Obstetrics and Gynecology, Guangdong No.2 Provincial People's Hospital, NO.466 Xingangdong Road, Guangzhou, 510317, China
| | - QiuXian Mao
- Department of Obstetrics and Gynecology, Guangdong No.2 Provincial People's Hospital, NO.466 Xingangdong Road, Guangzhou, 510317, China
| | - JingJun He
- Department of Physical Examination, Guangdong No.2 Provincial People's Hospital, NO.466 Xingangdong Road, Guangzhou, 510317, China
| | - HaoQing She
- Department of Obstetrics and Gynecology, Medical College, NanHua University, Hengyang, Hunan, 421001, China
| | - Zhi Zhang
- Department of Laboratory Medicine, Guangdong No.2 Provincial People's Hospital, NO.466 Xingangdong Road, Guangzhou, 510317, China.
| | - ChunYan Yin
- Department of Obstetrics and Gynecology, Guangdong No.2 Provincial People's Hospital, NO.466 Xingangdong Road, Guangzhou, 510317, China.
| |
Collapse
|
9
|
Nivet AL, Léveillé MC, Leader A, Sirard MA. Transcriptional characteristics of different sized follicles in relation to embryo transferability: potential role of hepatocyte growth factor signalling. Mol Hum Reprod 2016; 22:475-84. [PMID: 27126491 DOI: 10.1093/molehr/gaw029] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 04/25/2016] [Indexed: 01/29/2023] Open
Abstract
STUDY HYPOTHESIS We hypothesized that a better discrimination between follicles containing oocytes with high developmental competence and those containing oocytes with low competence, based on a combination of a follicle's size and transcriptomic signature, will provide a reliable method to predict embryonic outcome of IVF. STUDY FINDING This study provides new insights on the impact of follicular size on oocyte quality as measured by embryonic development and demonstrates that medium follicles yield a better percentage of transferable embryos. WHAT IS KNOWN ALREADY Although it is generally accepted that large ovarian follicles contain better eggs, other studies report that a better follicular size subdivision and a better characterization are needed. STUDY DESIGN, SAMPLES/MATERIALS, METHODS Individual follicles (n = 136), from a total of 33 women undergoing IVF, were aspirated and categorized on the basis of their follicular liquid volume (small, medium or large) and the embryonic outcome of the enclosed oocyte: poor or good development. Comprehensive gene expression analysis between cells from the different sized follicles was performed using microarrays and quantitative RT-PCR to find molecular markers associated with follicular maturity and oocyte developmental competence. MAIN RESULTS AND THE ROLE OF CHANCE The analysis of embryonic outcome in relation to follicular size indicates that the medium-sized follicles category yielded more transferable embryos (35%) compared with the largest follicles (30%) (NS). Gene expression analysis revealed expression markers with significant (P < 0.05) discrimination between the poor development groups for all three follicle sizes, and good development medium-size follicles, including up-regulation of thrombomodulin, transforming growth factor, beta receptor II and chondrolecti, and those associated with hyaluronan synthesis, coagulation and hepatocyte growth factor signalling. LIMITATIONS, REASONS FOR CAUTION These analyses were performed in a single cohort of patients coming from a single clinic and the biomarkers generated will require validation in different geographical and biological contexts to ensure their global applicability. WIDER IMPLICATIONS OF THE FINDINGS Medium-size follicles seem to be the optimal size for a positive embryonic outcome and are associated with competence markers that may help in understanding the ideal differentiation status during late folliculogenesis. LARGE SCALE DATA The data discussed in this publication have been deposited in The National Center for Biotechnology Information Gene Expression Omnibus database and are accessible through GEO Series accession number GSE52851. STUDY FUNDING AND COMPETING INTERESTS This study was supported by Canadian Institutes of Health Research (CIHR) and Natural Sciences and Engineering Research Council of Canada (NSERC) to M.A.S. There are no competing interests to declare.
Collapse
Affiliation(s)
- A L Nivet
- Département des Sciences Animales, Centre de Recherche en Biologie de la Reproduction, Institut sur la Nutrition et les Aliments fonctionnels, Université Laval, 2440 Boulevard Hochelaga, Quebec, QC G1V 0A6, Canada
| | - M C Léveillé
- Ottawa Fertility Clinic, 100-955 Green Valley Crescent, Ottawa, ON K2C 3V4, Canada
| | - A Leader
- Ottawa Fertility Clinic, 100-955 Green Valley Crescent, Ottawa, ON K2C 3V4, Canada
| | - M A Sirard
- Département des Sciences Animales, Centre de Recherche en Biologie de la Reproduction, Institut sur la Nutrition et les Aliments fonctionnels, Université Laval, 2440 Boulevard Hochelaga, Quebec, QC G1V 0A6, Canada
| |
Collapse
|
10
|
Field SL, Dasgupta T, Cummings M, Orsi NM. Cytokines in ovarian folliculogenesis, oocyte maturation and luteinisation. Mol Reprod Dev 2013; 81:284-314. [DOI: 10.1002/mrd.22285] [Citation(s) in RCA: 142] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 11/18/2013] [Indexed: 01/22/2023]
Affiliation(s)
- Sarah L Field
- Women's Health Research Group; Leeds Institute of Cancer; Anatomy and Pathology; Wellcome Trust Brenner Building; St James's University Hospital; Leeds UK
| | - Tathagata Dasgupta
- Department of Systems Biology; Harvard Medical School; 200 Longwood Avenue Boston Massachusetts
| | - Michele Cummings
- Women's Health Research Group; Leeds Institute of Cancer; Anatomy and Pathology; Wellcome Trust Brenner Building; St James's University Hospital; Leeds UK
| | - Nicolas M. Orsi
- Women's Health Research Group; Leeds Institute of Cancer; Anatomy and Pathology; Wellcome Trust Brenner Building; St James's University Hospital; Leeds UK
| |
Collapse
|
11
|
Guo JQ, Gao X, Lin ZJ, Wu WZ, Huang LH, Dong HY, Chen J, Lu J, Fu YF, Wang J, Ma YJ, Chen XW, Wu ZX, He FQ, Yang SL, Liao LM, Zheng F, Tan JM. BMSCs reduce rat granulosa cell apoptosis induced by cisplatin and perimenopause. BMC Cell Biol 2013; 14:18. [PMID: 23510080 PMCID: PMC3640998 DOI: 10.1186/1471-2121-14-18] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Accepted: 02/20/2013] [Indexed: 11/10/2022] Open
Abstract
Background The objective of this study was to evaluate the effect of bone marrow mesenchymal stem cells (BMSCs) on the apoptosis of granulosa cells (GCs) in rats. BMSCs and GCs were isolated from rats. GCs were separated into one of the following three groups: an untreated control group (control), a cisplatin (5 mg/L) treatment group (cisplatin), and group co-cultured with BMSCs and treated with cisplatin (BMSC). GC apoptosis was analyzed by annexin V staining and real-time PCR analysis for apoptosis-related genes. The effect of BMSCs was also determined in 9 to 10 month-old perimenopausal rats that were separated into the following groups: saline control, BMSC transplantation (1–2 × 106 cells), and estrogen treatment (0.158 mg/kg/d) groups. A young group consisting of 3 to 4 month-old rats that were treated with saline was also evaluated as a control. After 1 and 3 months, GC apoptosis was evaluated by TUNEL analysis. Results Cisplatin increased GC apoptosis from 0.59% to 13.04% in the control and cisplatin treatment groups, respectively, which was significantly reduced upon co-culture with BMSCs to 4.84%. Cisplatin treatment increased p21 and bax and decreased c-myc mRNA expression, which was reversed upon co-culture with BMSCs. As compared to young rats, increased apoptosis was observed in the perimenopausal rats (P < 0.001). After 3 months, the apoptosis rate in the BMSC group was significantly lower than that of the control group (P = 0.007). Conclusions BMSC therapy may protect against GC apoptosis induced by cisplatin and perimenopause. Further studies are necessary to evaluate therapeutic efficacy of BMSCs.
Collapse
|
12
|
Effects of melatonin on ovarian follicles. Eur J Obstet Gynecol Reprod Biol 2012; 166:178-84. [PMID: 23102587 DOI: 10.1016/j.ejogrb.2012.10.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Revised: 08/24/2012] [Accepted: 10/03/2012] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To evaluate the histomorphometry and expression of Ki-67 and c-kit in ovarian follicles of pinealectomized or melatonin-treated pinealectomized rats. STUDY DESIGN Forty adult rats were randomly divided into four groups of 10 animals: Group I - control; Group II - sham-pinealectomized; Group III - pinealectomized (Px), and Group IV - Px treated with melatonin (10μg/night, per animal). After two months' treatment, on the night of proestrous, the animals were placed in metabolic cages for night urine collection and subsequent measurement of 6-sulfatoxymelatonin (6-SMT). The rats were anesthetized, blood samples were taken for estrogen and progesterone determinations, and they were then euthanized. The ovaries were dissected out for further histological and immunohistochemical analyses. Data were first submitted to analysis of variance (ANOVA) complemented with the Tukey-Kramer test for multiple comparisons (P<0.05). RESULTS The urinary levels of 6-SMT and serum progesterone were lower in the Px group (GIII). Exogenous melatonin treatment restored both blood melatonin and 6-SMT urinary levels. The histomorphometric data in Group III revealed a significant increase of degenerating antral and non-antral follicles with regard to the other groups. In addition no corpora lutea were observed in this group. No significant differences were noticed regarding the number of corpora lutea among the other groups (I, II and IV), but the number of cells and the thickness of the theca interna of Px animals (Group III) were higher than in the other groups. Conversely, the density of progesterone receptors (fmol/g) in the ovaries of Group III was significantly lower than in the other groups. CONCLUSION Our data indicate that melatonin exerts a role on the maintenance of a proper follicular function, and is thus important for ovulation and progesterone production.
Collapse
|
13
|
Şahin N, Toylu A, Gülekli B, Doğan E, Kovali M, Atabey N. The levels of hepatocyte growth factor in serum and follicular fluid and the expression of c-Met in granulosa cells in patients with polycystic ovary syndrome. Fertil Steril 2012; 99:264-269.e3. [PMID: 23036804 DOI: 10.1016/j.fertnstert.2012.08.059] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2012] [Revised: 08/28/2012] [Accepted: 08/28/2012] [Indexed: 11/29/2022]
Abstract
OBJECTIVE To evaluate the levels of hepatocyte growth factor (HGF) in follicular fluid (FF) and the expression of c-Met in granulosa cells (GCs) with respect to the quality of the oocyte and embryo both in patients with polycystic ovary syndrome (PCOS) and in the normal ovary during controlled ovarian hyperstimulation cycles. DESIGN Prospective controlled study. SETTING University hospital. PATIENT(S) Fifty-nine women undergoing IVF treatment (of whom 21 had PCOS and 38 were in the control group). INTERVENTION(S) A total of 168 FF samples were collected at the time of oocyte retrieval. The HGF levels were measured by ELISA, and the mRNA expression of c-Met in GCs was detected by real-time polymerase chain reaction. MAIN OUTCOME MEASURE(S) The predictive values of HGF levels in serum and FF and the mRNA expression of c-Met in GCs for successful fertilization and oocyte-embryo quality. RESULT(S) The levels of HGF in serum and FF and the c-Met expression in GCs were similar between the PCOS and control groups. Granulosa cells of fertilized oocytes (2PN) had a significantly higher level of c-Met expression than that in oocytes that failed to fertilize. The mean HGF level in FF was significantly higher in the grade 1 embryos than in the grades 2-4 embryos. CONCLUSION(S) This study suggests that HGF/c-Met signaling may be a crucial determinant of fertilization success.
Collapse
Affiliation(s)
- Nur Şahin
- Department of Obstetrics and Gynecology, Dokuz Eylül University Medical School Hospital, İzmir, Turkey.
| | - Aslı Toylu
- Department of Medical Biology and Genetics, Dokuz Eylül University Medical School Hospital, İzmir, Turkey
| | - Bülent Gülekli
- Department of Obstetrics and Gynecology, Dokuz Eylül University Medical School Hospital, İzmir, Turkey
| | - Erbil Doğan
- Department of Obstetrics and Gynecology, Dokuz Eylül University Medical School Hospital, İzmir, Turkey
| | - Müge Kovali
- Department of Obstetrics and Gynecology, Dokuz Eylül University Medical School Hospital, İzmir, Turkey
| | - Neşe Atabey
- Department of Medical Biology and Genetics, Dokuz Eylül University Medical School Hospital, İzmir, Turkey
| |
Collapse
|
14
|
Merkwitz C, Lochhead P, Tsikolia N, Koch D, Sygnecka K, Sakurai M, Spanel-Borowski K, Ricken AM. Expression of KIT in the ovary, and the role of somatic precursor cells. ACTA ACUST UNITED AC 2011; 46:131-84. [DOI: 10.1016/j.proghi.2011.09.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
15
|
Guglielmo MC, Ricci G, Catizone A, Barberi M, Galdieri M, Stefanini M, Canipari R. The effect of hepatocyte growth factor on the initial stages of mouse follicle development. J Cell Physiol 2011; 226:520-9. [PMID: 20683913 DOI: 10.1002/jcp.22361] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Interactions between theca and granulosa cells of the follicle are critical for the coordination of ovarian follicle development. The cell-cell interactions are mediated through the local production and actions of a variety of factors. The current study is designed to investigate the expression of Hgf and its receptor, c-Met, in the mouse ovary during in vivo folliculogenesis. We found that Hgf and c-Met mRNAs were already expressed in 2-day-old ovaries, and that, while c-Met levels remained constant until 22-day-old, Hgf levels slightly but not significantly increased with age. The expression of Hgf mRNA in theca/interstitial cells was higher than in granulosa cells in 22-day-old ovaries. Immunohistochemistry analysis confirmed the expression pattern demonstrated by RT-PCR. We investigated the role of hepatocyte growth factor (HGF) at the beginning of mouse folliculogenesis and its possible interaction with kit ligand (KL). Interestingly, both KL and HGF were able to increase the expression of each other, creating a positive feedback loop. In the presence of HGF, we observed an increase of granulosa cell proliferation and an increase in the number of pre-antral and early antral follicles in ovary organ cultures. We also observed a significant increase in the diameters of follicles in individual follicle cultures. Moreover, HGF stimulated the expression of the FSH receptors, both in the whole ovary and in isolated pre-antral follicle cultures. Based on the data presented, we concluded that HGF exerts multiple levels of control over follicular cell functions, which collectively enable the progression of follicular development.
Collapse
Affiliation(s)
- M C Guglielmo
- Department of Histology and Medical Embryology, La Sapienza University of Rome, Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
16
|
Gougeon A. Human ovarian follicular development: from activation of resting follicles to preovulatory maturation. ANNALES D'ENDOCRINOLOGIE 2010; 71:132-43. [PMID: 20362973 DOI: 10.1016/j.ando.2010.02.021] [Citation(s) in RCA: 151] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2010] [Revised: 02/12/2010] [Accepted: 02/15/2010] [Indexed: 11/25/2022]
Abstract
By integrating morphometrical and endocrinological data, as well as biological effects of various molecules synthesized by the human follicle, we propose a dynamic view of the follicle growth within the human ovary. Folliculogenesis starts with entry of resting follicles into the growth phase, a process where the kit system plays a key role. Several months are required for a new growing follicle to reach the preantral stage (0.15mm), then 70 additional days to reach the size of 2mm. Early growing follicle growth is regulated by subtle interactions between follicle-stimulating hormone (FSH) and local factors produced by theca and granulosa cells (GCs), as well as the oocyte. From the time they enter the selectable stage during the late luteal phase, follicles become sensitive to cyclic changes of FSH in terms of granulosa cell proliferation. During the early follicular phase, the early selected follicle grows very quickly and estradiol is present in the follicular fluid. However, the total steroid production remains moderate. From the mid-follicular phase, the preovulatory follicle synthesizes high quantities of estradiol, then after the mid-cycle gonadotropin surge, very large amounts of progesterone. At this stage of development, the responsiveness of the follicle to gonadotropins is maximum, especially to luteinizing hormone (LH) that triggers granulosa wall dissociation and cumulus expansion as well as oocyte nuclear maturation. Thus, as the follicle develops, its responsiveness to gonadotropins progressively increases under the control of local factors acting in an autocrine/paracrine fashion.
Collapse
Affiliation(s)
- A Gougeon
- Inserm U865, Anipath, faculté de médecine Laënnec, 7, rue Guillaume-Paradin, 69372 Lyon cedex 08, France.
| |
Collapse
|
17
|
Fu X, He Y, Xie C, Liu W. Bone marrow mesenchymal stem cell transplantation improves ovarian function and structure in rats with chemotherapy-induced ovarian damage. Cytotherapy 2008; 10:353-63. [PMID: 18574768 DOI: 10.1080/14653240802035926] [Citation(s) in RCA: 140] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Many investigations have reported that mesenchymal stem cell (MSC) transplantation can ameliorate the structure and function of injured tissues. The purpose of this study was to explore the therapeutic potency of MSC transplantation for chemotherapy-induced ovarian damage. METHODS MSC were isolated and cultured in vitro. The cytokines, including vascular endothelial growth factor (VEGF), hepatocyte growth factor (HGF) and insulin-like growth factor-1 (IGF-1), were detected in the MSC cultures using enzyme-linked immunosorbent assay (ELISA). Phosphoramide mustard (PM) was added to the media of granulosa cells (GC) cultured alone or co-cultured with MSC. GC apoptosis was assayed by Annexin-V and DNA fragmentation analysis. Chemotherapy-induced ovarian damage was induced in rats by intraperitoneal injection of cyclophosphamide (CTX). After the injection, MSC labeled with green fluorescent protein (GFP) were transplanted directly into bilateral ovaries. The rats were killed at 2, 4, 6 and 8 weeks after transplantation. Ovarian function was evaluated by estrous cycle changes and sexual hormone levels. The follicle number was counted, and GC apoptosis was analyzed by TUNEL. The expressions of Bcl-2 and Bax proteins were detected by Western blotting. RESULTS MSC released VEGF, HGF and IGF-1 in vitro. The GC apoptosis was diminished by co-culture with MSC, which also resulted in increased Bcl-2 expression. The ovarian function of the rats exposed to CTX injection was improved after MSC transplantation. MSC reduced apoptosis of GC and induced up-regulation of Bcl-2 in vivo. DISCUSSION MSC transplantation can improve ovarian function and structure damaged by chemotherapy. The paracrine mediators secreted by MSC might be involved in the repair of damaged ovaries.
Collapse
Affiliation(s)
- X Fu
- Department of Obstetrics and Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China
| | | | | | | |
Collapse
|
18
|
Expression, mutational analysis and in vitro response of imatinib mesylate and nilotinib target genes in ovarian granulosa cell tumors. Gynecol Oncol 2007; 108:182-90. [PMID: 18028988 DOI: 10.1016/j.ygyno.2007.09.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2007] [Revised: 09/12/2007] [Accepted: 09/13/2007] [Indexed: 10/22/2022]
Abstract
OBJECTIVES Granulosa cell tumors of the ovary (GCT) represent approximately 5% of malignant ovarian tumors. Surgery remains the primary modality of therapy and treatment options for advanced disease are limited. The molecular pathogenesis of GCT is not known but is likely to involve activation of tyrosine kinase-mediated cell signaling pathways. A recent case report of a patient with advanced recurrent GCT responding to the tyrosine kinase inhibitor, imatinib mesylate prompted us to explore a role for these therapies in GCT. METHODS The expression of the imatinib-sensitive tyrosine kinases, c-kit, c-Abl, PDGFR-alpha and PDGFR-beta, was determined using RT-PCR in a panel of GCT. Activating mutations of c-kit and PDGFR-alpha were also sought. The functional response was examined in two human-derived GCT cell lines. RESULTS All four kinases were expressed but at levels lower than those observed in pre-menopausal ovarian samples. Mutations in c-kit and PDGFR-alpha were not found. Both cell lines responded to imatinib and to the second generation, tyrosine kinase inhibitor, nilotinib, with dose-dependent decreases in cell proliferation and viability. These responses paralleled the imatinib-sensitive, K562 cell line but at approximately 240- and approximately 1000-fold higher concentrations of imatinib and nilotinib, respectively. CONCLUSIONS Our study suggests that human GCT, in general, are unlikely to respond to imatinib or nilotinib therapy. The response of the cell lines at high concentrations implies an "off-target" effect, which suggests that a tyrosine kinase inhibitor, of appropriate specificity, may represent a therapeutic option in GCT.
Collapse
|
19
|
Posadas EM, Kwitkowski V, Kotz HL, Espina V, Minasian L, Tchabo N, Premkumar A, Hussain MM, Chang R, Steinberg SM, Kohn EC. A prospective analysis of imatinib-induced c-KIT modulation in ovarian cancer: a phase II clinical study with proteomic profiling. Cancer 2007; 110:309-17. [PMID: 17559139 DOI: 10.1002/cncr.22757] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND c-Kit and platelet-derived growth factor receptor (PDGFR) are potential molecular targets in epithelial ovarian cancer (EOC). Imatinib inhibits the kinase domain and subsequent downstream signaling of these receptor tyrosine kinases. The objective of this study was to investigate biochemical and biologic effects of imatinib on EOC. METHODS Patients with recurrent EOC who had received no more than 4 prior regimens and who had good end-organ function were eligible. Imatinib was administered orally at a dose of 400 mg twice daily in continuous, 28-day cycles with reassessment imaging studies obtained every other cycle. Tumor core biopsies were obtained prior to and at 4 weeks into therapy; microdissected tumor and stroma were subjected to protein lysate array analysis. Blood samples were obtained monthly for cytokine measurements. RESULTS Twenty-three patients were enrolled, including 16 patients who received imatinib 600 mg daily because of gastrointestinal (GI) toxicity and fluid accumulation at the starting dose. The median time to disease progression was 2 months (range, 2-14 months). Common grade 3 toxicities included edema/ascites/pleural effusions in 11 patients (48%), GI complaints in 8 patients (35%), fatigue in 3 patients (13%), and grade 2 and 3 cytopenias in 10 patients and 3 patients (43% and 13%), respectively. Increased circulating levels of interleukin 6 were associated with grade >/=2 fluid collection (P = .02). A statistically significant trend was observed between pretreatment phosphorylated-kit levels in microdissected tumor and stroma and GI toxicity (P < .01), between tumor levels of epidermal growth factor receptor (EGFR) and PDGFR with grade of fatigue (P </= .005), and EGFR and phosphorylated-AKT levels with grade of ascites and edema (P </= .01). CONCLUSIONS The results of this study indicated imatinib had minimal activity as a single agent in EOC. Its ability to modulate its molecular targets suggests that it may be considered in combinatorial therapy.
Collapse
Affiliation(s)
- Edwin M Posadas
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892-1500, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Taniguchi F, Harada T, Iwabe T, Ohama Y, Takenaka Y, Terakawa N. Aberrant expression of keratinocyte growth factor receptor in ovarian surface epithelial cells of endometrioma. Fertil Steril 2007; 89:478-80. [PMID: 17482184 DOI: 10.1016/j.fertnstert.2007.02.060] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2006] [Revised: 02/23/2007] [Accepted: 02/23/2007] [Indexed: 10/23/2022]
Abstract
Ovarian surface epithelial cells (OSEs) are considered to be the common source of endometrioma and epithelial ovarian cancer. The present study reveals that keratinocyte growth factor receptor (KGFR) messenger RNA was expressed in OSEs of endometriomas but not in those of normal ovaries, suggesting that autocrine KGF/KGFR and paracrine fibroblast growth factor 10/KGFR signaling loops may be involved with the proliferation in OSEs of endometrioma.
Collapse
Affiliation(s)
- Fuminori Taniguchi
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, Yonago, Japan.
| | | | | | | | | | | |
Collapse
|
21
|
Uzumcu M, Pan Z, Chu Y, Kuhn PE, Zachow R. Immunolocalization of the hepatocyte growth factor (HGF) system in the rat ovary and the anti-apoptotic effect of HGF in rat ovarian granulosa cells in vitro. Reproduction 2006; 132:291-9. [PMID: 16885537 DOI: 10.1530/rep.1.00989] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Hepatocyte growth factor (HGF) regulates granulosa cell (GC) steroidogenesis and suppresses apoptosis in non-ovarian cells. The hypothesis was thus developed that intraovarian HGF supports folliculogenesis by mediating steroidogenesis and suppressing apoptosis. To investigate the latter, the anti-apoptotic actions of HGF were tested in GCs and follicles isolated from immature rats. Results showed that HGF suppressed apoptosis in GC and follicle cultures as visualized using apoptosis indicator dye, YO-PRO-1. Immunohistochemistry was used to investigate the distribution of HGF, c-met, and HGF activator (HGFA) protein during folliculogenesis in equine chorionic gonadotropin (eCG)-primed rats. Immunoreactive HGF content was the greatest in GCs within preantral follicles. Following eCG, large antral follicles showed elevated HGF staining in theca and interstitial cells when compared with GCs. Intense c-met staining was observed in GCs within non-primed small preantral follicles; following eCG, the level of c-met was diminished in GCs, but increased within theca and interstitial cells. Theca, interstitium, and GCs in non-primed and primed ovaries contained HGFA. Following eCG, HGFA was more apparent in theca cells and the interstitium when compared to that in GCs within large antral follicles. The presence of HGF, c-met, and HGFA in preantral follicles would potentially enable the anti-apoptotic effects of HGF that were observed in vitro to occur in vivo. Advanced folliculogenesis led to a change in the cellular distribution of the HGF, c-met, and HGFA, suggesting that the ovarian HGF system is hormonally regulated in vivo.
Collapse
Affiliation(s)
- Mehmet Uzumcu
- Department of Animal Sciences, Rutgers University, New Brunswick, New Jersey 08901, USA
| | | | | | | | | |
Collapse
|
22
|
Shimizu T, Iijima K, Sasada H, Sato E. Messenger ribonucleic acid expressions of hepatocyte growth factor, angiopoietins and their receptors during follicular development in gilts. J Reprod Dev 2006; 49:203-11. [PMID: 14967929 DOI: 10.1262/jrd.49.203] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Angiogenic factors are associated with angiogenesis during follicular development in the mammalian ovary. The aim of the present study was to determine the relationships between the vascular network and mRNA expressions of angiopoietins (Ang)-1, Ang-2 and hepatocyte growth factor (HGF), and their receptors in follicles at different developmental stages during follicular development. Ovaries in gilts were collected 72 h after equine chorionic gonadotropin (eCG, 1250 IU) treatment for histological observation of the capillary network. Granulosa cells and thecal tissues in small (<4 mm), medium (4-5 mm) or large (>5 mm) individual follicles were collected for detection of mRNA expression of HGF, Ang-1 and Ang-2 in granulosa cells, and HGF receptor (HGF-R) and Tie-2 in the theca cells by semi-quantitative RT-PCR. The number of capillaries in the thecal cell layer increased significantly in healthy follicles at all developmental stages in the eCG group compared with those in controls. The expression of Ang-1 mRNA declined in granulosa cells of medium and large follicles and the level of Ang-2 mRNA increased in granulosa cells of small follicles after eCG treatment. The ratio of Ang-2/Ang-1 increased in small, medium and large follicles from ovaries after eCG treatment, but Tie-2 mRNA expression in the theca cells did not change. The level of HGF mRNA increased in granulosa cells of small follicles after eCG treatment but HGF-R in theca cells was not increased by eCG. These data suggested that the angiopoietins might be associated with thecal angiogenesis during follicular development in eCG-treated gilts.
Collapse
Affiliation(s)
- Takashi Shimizu
- Laboratory of Animal Reproduction, Graduate School of Agricultural Science, Tohoku University, Aoku, Japan.
| | | | | | | |
Collapse
|
23
|
Carlsson IB, Laitinen MPE, Scott JE, Louhio H, Velentzis L, Tuuri T, Aaltonen J, Ritvos O, Winston RML, Hovatta O. Kit ligand and c-Kit are expressed during early human ovarian follicular development and their interaction is required for the survival of follicles in long-term culture. Reproduction 2006; 131:641-9. [PMID: 16595715 DOI: 10.1530/rep.1.00868] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The receptor tyrosine c-Kit and its cognate ligand, c-Kit ligand (KL, stem cell factor, SCF), are involved in ovarian follicular development in several animal species. We studied the expression of KL and c-Kit usingin situhybridization and immunohistochemistry in donated human ovarian cortical tissue. The KL transcripts were expressed in granulosa cells of primary follicles, whereas the expression of c-Kit was confined to the oocyte and granulosa cells in primary and secondary follicles. We employed an ovarian organ culture using firstly serum-containing and then serum-free medium to study the effects of KL and an anti-c-Kit antibody, ACK2, on the development and survival of ovarian folliclesin vitro. Culture of ovarian cortical slices for 7 days resulted in a 37% increase in the number of primary follicles and a 6% increase in secondary follicles. The proportion of viable follicles decreased in all cultures. The addition of KL (1, 10 and 100 ng/ml) into the culture media did not affect the developmental stages of the follicles or the proportion of atretic follicles. Inclusion of ACK2 (800 ng/ml) in the culture medium significantly increased the proportion of atretic follicles on days 7 (49 vs 28% in control cultures) and 14 (62 vs 38%) of culture. In conclusion, c-Kit and KL are expressed in human ovaries during follicular development. Blocking the c-Kit receptor induces follicular atresia. The KL/c-Kit signaling system is likely to control the survival of human ovarian follicles during early follicular development.
Collapse
Affiliation(s)
- Inger B Carlsson
- Karolinska Institutet, Department of Clinical Science, Intervention and Technology. Karolinska University Hospital, Stockholm, Sweden.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Wickenheisser JK, Nelson-DeGrave VL, McAllister JM. Human ovarian theca cells in culture. Trends Endocrinol Metab 2006; 17:65-71. [PMID: 16460956 DOI: 10.1016/j.tem.2006.01.003] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2005] [Revised: 11/30/2005] [Accepted: 01/19/2006] [Indexed: 12/23/2022]
Abstract
Elucidating the regulation of androgen biosynthesis in ovarian theca cells is not only important for determining the mechanisms of regulation of estrogen biosynthesis throughout the menstrual cycle, but is also essential for understanding the pathogenesis of excess androgen biosynthesis and polycystic ovary syndrome (PCOS). Human theca cells in primary and long-term culture have provided model systems for examining theca cell differentiation as well as the mechanisms underlying basal and cAMP-regulated steroid biosynthesis at both the transcriptional and post-transcriptional level in normal and PCOS ovaries. Results of these studies are expected to lead to the identification of novel targets for clinical treatment of infertility and PCOS.
Collapse
Affiliation(s)
- Jessica K Wickenheisser
- Departments of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA
| | | | | |
Collapse
|
25
|
Lim JC, Wojcik EM. Fine-needle aspiration cytology of papillary renal cell carcinoma: The association with concomitant secondary malignancies. Diagn Cytopathol 2006; 34:797-800. [PMID: 17115441 DOI: 10.1002/dc.20546] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Papillary renal cell carcinoma is a rare type of renal malignancy. Cytogenetic findings characteristic for this tumor have been described as well as mutations of the proto-oncogene c-met. Secondary malignancies occurring together with papillary renal cell carcinomas are rare, and are often of genitourinary tract origin. We describe two cases of papillary renal cell carcinoma occurring in association with two other visceral malignancies, gastrointestinal stromal tumor and colon adenocarcinoma.Two cases of papillary renal cell carcinoma diagnosed by fine-needle aspiration (FNA), occurring in association with gastrointestinal malignancies were reviewed. Both aspirates showed cytologic features characteristic for papillary renal cell carcinoma, namely papillary structures, foamy histiocytes, intracytoplasmic hemosiderin, and nuclear grooves. Subsequent histology and immunohistochemical stains supported the cytologic diagnosis. The histologic diagnosis of gastrointestinal stromal tumor and colon adenocarcinoma were confirmed. Papillary renal cell carcinoma is a type of renal carcinoma that can be often accurately diagnosed by FNA. The occurrence of associated visceral malignancies has never been reported. The possible role of the protooncogene c-met in the development of these tumors was explored.
Collapse
Affiliation(s)
- Jennifer C Lim
- Department of Pathology, New York University, New York, NY, USA
| | | |
Collapse
|
26
|
Taniguchi F, Harada T, Nara M, Deura I, Mitsunari M, Terakawa N. Coculture with a human granulosa cell line enhanced the development of murine preimplantation embryos via SCF/c-kit system. J Assist Reprod Genet 2004; 21:223-8. [PMID: 15526978 PMCID: PMC3455229 DOI: 10.1023/b:jarg.0000040238.61586.86] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
PURPOSE To evaluate the effect on the coculture of murine embryos with a human ovarian granulosa tumor derived cell line (KGN cells). METHODS We observed microscopically the growth of murine preimplantation embryos in the coculture system with KGN cells or in the presence with exogenous stem cell factor (SCF). The reverse transcriptase-polymerase chain reaction (RT-PCR) method was used to analyze the gene expression of SCF in KGN cells cocultured with murine embryos. RESULTS The coculture system with KGN cells significantly increased the rate of embryo development to late blastocyst and to hatching stage. We also found that coculture with murine embryos enhanced the gene expression of SCF in KGN cells. Adding human recombinant SCF to the medium significantly enhanced embryo development to late blastocyst and hatching stage. CONCLUSIONS KGN cells may facilitate preimplantion embryo development through SCF/c-kit paracrine system.
Collapse
Affiliation(s)
- Fuminori Taniguchi
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, 36-1 Nishimachi, Yonago 683-8504, Japan.
| | | | | | | | | | | |
Collapse
|
27
|
Hammadeh ME, Fischer-Hammadeh C, Hoffmeister H, Herrmann W, Rosenbaum P, Schmidt W. Relationship between cytokine concentrations (FGF, sICAM-1 and SCF) in serum, follicular fluid and ICSI outcome. Am J Reprod Immunol 2004; 51:81-5. [PMID: 14725569 DOI: 10.1046/j.8755-8920.2003.00121.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
OBJECTIVE The aim of this study was (i) to investigate the existence of fibroblast growth factor (FGF), soluble intracellular adhesion molecule-1 (sICAM-1), and stem cell factor (SCF) in serum and human follicular fluid (FF) of intracytoplasmic sperm injection (ICSI) patients, and (ii) to determine the relationship between these parameters and ICSI outcome. MATERIAL AND METHOD Seventy-five patients undergoing controlled ovarian hyperstimulation with human menopausal gonadotropin (hMG) after down-regulation with GnRHa were included in this study. The concentrations of FGF, SCF, and sICAM-1 were measured by using commercially available enzyme-linked immunosorbent assay test kits. RESULTS The FGF, sICAM-1, and SCF concentrations in the serum of women who become pregnant (group I) were 8.5 +/- 1.5 pg/mL, 235.8 +/- 81.1 ng/mL, and 597.7 +/- 139.9 pg/mL, and the corresponding concentrations of women who did not (group II) were 6.4 +/- 3.6 pg/mL, 230.6 +/- 66.5 ng/mL, and 569.6 +/- 91.4 pg/mL respectively. No significant difference was observed between the two investigated groups with regard to the number of hMG ampoules administered for controlled ovarian hyperstimulation, estradiol concentration on the day of human chorionic gonadotropin (hCG) injection, number of retrieved oocytes and fertilization rate. CONCLUSION The concentration of FGF, sICAM-1, and SCF did not differ significantly between the two groups in serum or in FF. Besides, the ICSI outcome was not related to their concentrations in serum or FF. Therefore, these parameters could not be used as a prognostic factor in ICSI program.
Collapse
Affiliation(s)
- Mohamad Eid Hammadeh
- Department of Obstetrics and Gynecology, The University of Saarland, Homburg/Saar, Germany.
| | | | | | | | | | | |
Collapse
|
28
|
Taniguchi F, Harada T, Deura I, Iwabe T, Tsukihara S, Terakawa N. Hepatocyte growth factor promotes cell proliferation and inhibits progesterone secretion via PKA and MAPK pathways in a human granulosa cell line. Mol Reprod Dev 2004; 68:335-44. [PMID: 15112327 DOI: 10.1002/mrd.20076] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Hepatocyte growth factor (HGF) is a mesenchymal-derived paracrine factor that acts through a c-met receptor. The activated c-met receptor recruits various signal proteins. We used a steroidogenic human granulosa-like tumor cell line (KGN cells) to analyze the biological function of HGF in human ovary cells. First, we designed a method to analyze local production and action of HGF in the human ovary. Although c-met mRNA is expressed in KGN cells, granulosa lutein, theca, and ovarian stroma cells, we observed HGF mRNA only in theca and stroma cells. Adding HGF to the medium enhanced mitogenic activity in KGN cells. We next examined the activation of intracellular signal transduction molecules induced by HGF in KGN cells. Here, we showed that HGF activated the distinct phosphorylation of Raf-1, MEK1/2, and ERK1/2, but did not induce phosphorylation of Akt. HGF enhanced the phosphorylation of Elk-1 and c-Jun as nuclear transcription factors. U0126, a MEK1/2 inhibitor, completely abrogated the phosphorylation of ERK1/2 and the cell proliferation in response to HGF. In contrast, H-89, a protein kinase A inhibitor, further enhanced the HGF-induced phosphorylation of ERK1/2 and cell proliferation. In addition, we revealed that HGF suppressed progesterone synthesis in KGN cells. Adding HGF suppressed the forskolin-induced steroidogenic acute regulatory protein (StAR) expression, which is a key regulator in progesterone synthesis. Crosstalk signals between PKA and the mitogen-activated protein kinase (MAPK) pathway were mutually inhibitory. These results demonstrated for the first time that theca cell-derived HGF may be capable of stimulating the proliferation of granulosa cells and suppressing progesterone synthesis via an activating MAPK pathway.
Collapse
Affiliation(s)
- Fuminori Taniguchi
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, Yonago, Japan.
| | | | | | | | | | | |
Collapse
|
29
|
Nilsson EE, Skinner MK. Kit ligand and basic fibroblast growth factor interactions in the induction of ovarian primordial to primary follicle transition. Mol Cell Endocrinol 2004; 214:19-25. [PMID: 15062541 DOI: 10.1016/j.mce.2003.12.001] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2003] [Revised: 11/25/2003] [Accepted: 12/02/2003] [Indexed: 10/26/2022]
Abstract
Ovulated eggs during a female's reproductive life are derived from a pool of primordial follicles arrested in prophase of the first meiotic division. When follicles leave the resting pool they undergo a primordial to primary follicle transition and will grow and develop until either ovulation occurs or follicles undergo atresia. Several growth factors have been implicated as acting locally within the ovary to regulate the primordial to primary follicle transition. How these growth factors may interact and cooperate to perform this vital function remains to be elucidated. The objective of the current study is to investigate interactions between kit ligand (KL) (i.e. stem cell factor) and basic fibroblast growth factor (bFGF) that promote the primordial to primary follicle transition in rat ovaries. Ovaries were removed from 4-day-old rat pups and cultured for 2 weeks with KL alone or with KL and a neutralizing antibody against bFGF. The ability of KL treatment to increase primordial follicle transition was blocked with a bFGF neutralizing antibody. In addition, ovary cultures were treated with bFGF alone or with bFGF and an anti-c-kit receptor antibody which blocks KL signaling. The ability of bFGF treatment to increase primordial follicle transition was blocked with an anti-c-kit receptor antibody. Observations indicate that both KL and bFGF must be active in order to optimally promote the changes that occur in oocytes, granulosa cells, and stromal/interstitial cells when primordial follicles initiate development. Cultured ovaries were treated with either KL or bFGF for 3 days and then bFGF and KL mRNA expression levels in the whole ovary were measured. KL was not found to regulate bFGF expression. In contrast, bFGF treatment was found to increase KL mRNA expression in cultured ovaries. These observations suggest that one function of the oocyte-derived bFGF is to increase the granulosa derived KL expression and that both KL and bFGF are required to optimally promote primordial to primary follicle transition. Elucidating the cell-cell interactions that mediate this network of specific locally derived growth factors is critical to understanding the physiology of the primordial to primary follicle transition.
Collapse
Affiliation(s)
- Eric E Nilsson
- Center for Reproductive Biology, School of Molecular Biosciences, Washington State University, Pullman, WA 99164-4231, USA
| | | |
Collapse
|
30
|
Fujii A, Harada T, Yamauchi N, Iwabe T, Nishi Y, Yanase T, Nawata H, Terakawa N. Interleukin-8 gene and protein expression are up-regulated by interleukin-1beta in normal human ovarian cells and a granulosa tumor cell line. Fertil Steril 2003; 79:151-7. [PMID: 12524080 DOI: 10.1016/s0015-0282(02)04408-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To evaluate the expression, regulation, and role of interleukin (IL)-8 in human ovary. DESIGN Prospective study. SETTING University hospital. PATIENT(S) Sixteen premenopausal women. INTERVENTION Follicular fluid and granulosa lutein cells (GLCs) were collected during IVF cycles. Ovarian stromal and theca cells were obtained from women underwent surgery. KGN cells, the human granulosa cell tumor cell line, were also used. MAIN OUTCOME MEASURES The levels of IL-8 and IL-1beta in follicular fluid and IL-8 protein production were determined using ELISA. Interleukin-8 and IL-8 receptor gene expression in ovarian cells and the effect of IL-8 on the proliferation of stromal cells were determined. The expression of pIkappaB was evaluated by Western blot, and the effect of NF-kappaB inhibitor APDC was examined by Northern blot analysis and ELISA in KGN cells. The levels of IL-8 and IL-1beta in follicular fluid; each concentration and the volume showed a positive correlation. Reverse transcription polymerase chain reaction showed the presence of IL-8 mRNA in all ovarian cells. In contrast, IL-8 receptor mRNA was only detected in stromal cells. The expression of IL-8 in GLCs and KGN cells was increased by addition of IL-1beta and TNFalpha. Interleukin-8 increased the proliferation of ovarian stromal cells. The expression of pIkappaB in KGN cells was induced by IL-1beta, and the effects were reduced by APDC. CONCLUSION(S) Interleukin 8 induced by IL-1beta via activation of NF-kappaB in granulosa cells may have a role in the periovulatory period of follicular maturation.
Collapse
Affiliation(s)
- Akiko Fujii
- Department of Obstetrics and Gynecology, Tottori University School of Medicine, Yonago, Japan
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Zachow RJ, Woolery JK. Effects of hepatocyte growth factor on cyclic nucleotide-dependent signaling and steroidogenesis in rat ovarian granulosa cells in vitro. Biol Reprod 2002; 67:454-9. [PMID: 12135881 DOI: 10.1095/biolreprod67.2.454] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Hepatocyte growth factor (HGF) down-modulates FSH-dependent estradiol-17beta (E(2)) production in ovarian granulosa cells in vitro. The mechanisms of action underlying the antiestrogenic effects of HGF are vague, although evidence indicates that HGF may affect cAMP signal transduction in rat granulosa cells. The present study investigated the effects of HGF on FSH-induced steroidogenesis in the presence and absence of insulin-like growth factor I (IGF-I), as well as the actions of HGF within cyclic nucleotide-dependent signal transduction cascades in granulosa cells. Immature rat granulosa cells were incubated with FSH, IGF-I, and HGF. HGF impaired the production of FSH-stimulated and FSH + IGF-I-stimulated E(2) synthesis, as well as FSH + IGF-I-dependent estrone production. Progesterone synthesis was not altered by HGF. HGF suppressed FSH-dependent cAMP content at 24 h, but not at 36 h; cGMP content was stimulated by HGF with and without FSH at 24 h. In the presence of the cyclic nucleotide phosphodiesterase (PDE) inhibitor, 3-isobutyl-1-methylxanthine (IBMX), FSH-dependent cAMP accumulation was not affected by HGF. The suppressive effect of HGF on FSH-dependent E(2) production was alleviated by IBMX, whereas the HGF-dependent block in FSH + IGF-I-supported E(2) production was not prevented by IBMX. The effects of HGF on cyclic nucleotide PDE activities were manifested in a time-dependent and hormone-dependent manner. FSH-induced cAMP PDE was suppressed by HGF at 24 h but not at 36 h, whereas FSH-dependent cGMP PDE was impaired at 36 h, but not at 24 h. HGF prevented the IGF-I-dependent reduction in FSH-stimulated cAMP-PDE activity at 24 and 36 h, and lowered FSH + IGF-I-stimulated cGMP-PDE activity at 36 h, concomitant with an HGF-dependent increase in cGMP content at 24 h. These data indicate that HGF affects cAMP-directed and cGMP-directed signaling pathways at multiple sites in granulosa cells. These HGF-dependent effects may provide insight for mechanisms of action whereby HGF reduces E(2) secretion by granulosa cells.
Collapse
Affiliation(s)
- Rob J Zachow
- Department of Applied Dental Medicine, School of Dental Medicine, Southern Illinois University, 2800 College Avenue, Alton, IL 62002, USA.
| | | |
Collapse
|