1
|
Niinuma SA, Habib H, Takemoto ASN, Das P, Sathyapalan T, Atkin SL, Butler AE. A Cross-Sectional Exploratory Study of Rat Sarcoid (Ras) Activation in Women with and Without Polycystic Ovary Syndrome. Cells 2025; 14:377. [PMID: 40072105 PMCID: PMC11898917 DOI: 10.3390/cells14050377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/01/2025] [Accepted: 03/04/2025] [Indexed: 03/15/2025] Open
Abstract
Objective: Rat sarcoma (Ras) proteins, Kirsten, Harvey, and Neuroblastoma rat sarcoma viral oncogene homolog (KRAS, HRAS, and NRAS, respectively), are a family of GTPases, which are key regulators of cellular growth, differentiation, and apoptosis through signal transduction pathways modulated by growth factors that have been recognized to be dysregulated in PCOS. This study explores Ras signaling proteins and growth factor-related proteins in polycystic ovary syndrome (PCOS). Methods: In a well-validated PCOS database of 147 PCOS and 97 control women, plasma was batch analyzed using Somascan proteomic analysis for circulating KRas, Ras GTPase-activating protein-1 (RASA1), and 45 growth factor-related proteins. The cohort was subsequently stratified for BMI (body mass index), testosterone, and insulin resistance (HOMA-IR) for subset analysis. Results: Circulating KRas, and RASA1 did not differ between PCOS and control women (p > 0.05). EGF1, EGFR, and EGFRvIII were decreased in PCOS (p = 0.04, p = 0.04 and p < 0.001, respectively). FGF8, FGF9, and FGF17 were increased in PCOS (p = 0.02, p = 0.03 and p = 0.04, respectively), and FGFR1 was decreased in PCOS (p < 0.001). VEGF-D (p < 0.001), IGF1 (p < 0.001), IGF-1sR (p = 0.02), and PDGFRA (p < 0.001) were decreased in PCOS compared to controls. After stratifying for BMI ≤ 29.9 kg/m2, EGFR FGF8, FGFR1 VEGF-D, IGF1, and IGF-1sR differed (p < 0.05) though EGF1, EGFRvIII, FGF8, FGFR1, and VEGF-D no longer differed; after subsequently stratifying for HOMA-IR, only FGFR1, VEGF-D, IGF1, and IGF-1sR differed between groups (p < 0.05). Conclusions: Several growth factors that activate Ras differ between women with and without PCOS, and when stratified for BMI and HOMA-IR, only FGFR1, VEGF-D, IGF1, and IGF-1sR differed; these appear to be inherent features of the pathophysiology of PCOS.
Collapse
Affiliation(s)
- Sara Anjum Niinuma
- Research Department, Royal College of Surgeons of Ireland, Busaiteen, Adliya P.O. Box 15503, Bahrain; (S.A.N.); (H.H.); (A.S.-N.T.); (P.D.); (S.L.A.)
| | - Haniya Habib
- Research Department, Royal College of Surgeons of Ireland, Busaiteen, Adliya P.O. Box 15503, Bahrain; (S.A.N.); (H.H.); (A.S.-N.T.); (P.D.); (S.L.A.)
| | - Ashleigh Suzu-Nishio Takemoto
- Research Department, Royal College of Surgeons of Ireland, Busaiteen, Adliya P.O. Box 15503, Bahrain; (S.A.N.); (H.H.); (A.S.-N.T.); (P.D.); (S.L.A.)
| | - Priya Das
- Research Department, Royal College of Surgeons of Ireland, Busaiteen, Adliya P.O. Box 15503, Bahrain; (S.A.N.); (H.H.); (A.S.-N.T.); (P.D.); (S.L.A.)
| | - Thozhukat Sathyapalan
- Academic Endocrinology, Diabetes and Metabolism, Hull York Medical School, Hull HU6 7RX, UK;
| | - Stephen L. Atkin
- Research Department, Royal College of Surgeons of Ireland, Busaiteen, Adliya P.O. Box 15503, Bahrain; (S.A.N.); (H.H.); (A.S.-N.T.); (P.D.); (S.L.A.)
| | - Alexandra E. Butler
- Research Department, Royal College of Surgeons of Ireland, Busaiteen, Adliya P.O. Box 15503, Bahrain; (S.A.N.); (H.H.); (A.S.-N.T.); (P.D.); (S.L.A.)
| |
Collapse
|
2
|
Habel A, Weili X, Hadj Ahmed M, Stayoussef M, Bouaziz H, Ayadi M, Mezlini A, Larbi A, Yaacoubi-Loueslati B. Immune checkpoints as potential theragnostic biomarkers for epithelial ovarian cancer. Int J Biol Markers 2023; 38:203-213. [PMID: 37518940 DOI: 10.1177/03936155231186163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/01/2023]
Abstract
BACKGROUND Epithelial ovarian cancer (EOC) is the leading cause of death associated with gynecologic tumors. EOC is asymptomatic in early stages, so most patients are not diagnosed until late stages, highlighting the need to develop new diagnostic biomarkers. Mediators of the tumoral microenvironment may influence EOC progression and resistance to treatment. AIM To analyze immune checkpoints to evaluate them as theranostic biomarkers for EOC. PATIENTS AND METHODS Serum levels of 16 immune checkpoints were determined in EOC patients and healthy controls using the MILLIPLEX MAP® Human Immuno-Oncology Checkpoint Protein Magnetic Bead Panel. RESULTS Seven receptors: BTLA, CD40, CD80/B7-1, GITRL, LAG-3, TIM-3, TLR-2 are differentially expressed between EOC and healthy controls. Serum levels of immune checkpoints in EOC patients are positively significantly correlated with levels of their ligands, with a higher significant correlation between CD80 and CTLA4 than between CD28 and CD80. Four receptors, CD40, HVEM, PD-1, and PD-L1, are positively associated with the development of resistance to Taxol-platinum-based chemotherapy. All of them have an acceptable area under the curve (>0.7). CONCLUSION This study has yielded a first panel of seven immune checkpoints (BTLA, CD40, CD80/B7-1, GITRL, LAG-3, TIM-3, TLR-2) associated with a higher risk of EOC and a second panel of four immune checkpoints (CD40, HVEM, PD-1, PD-L1) that may help physicians to identify EOC patients who are at high risk of developing resistance to EOC chemotherapy.
Collapse
Affiliation(s)
- Azza Habel
- Laboratory of Mycology, Pathologies, and Biomarkers (LR16ES05), Faculty of Sciences of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Xu Weili
- Singapore Immunology Network, Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| | - Mariem Hadj Ahmed
- Laboratory of Mycology, Pathologies, and Biomarkers (LR16ES05), Faculty of Sciences of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Mouna Stayoussef
- Laboratory of Mycology, Pathologies, and Biomarkers (LR16ES05), Faculty of Sciences of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | | | - Mouna Ayadi
- Salah Azaiez Oncology Institute, Tunis, Tunisia
| | | | - Anis Larbi
- Singapore Immunology Network, Agency for Science Technology and Research (A*STAR), Singapore, Singapore
- Beckman Coulter Life Sciences, Villepinte, France
| | - Basma Yaacoubi-Loueslati
- Laboratory of Mycology, Pathologies, and Biomarkers (LR16ES05), Faculty of Sciences of Tunis, University of Tunis El Manar, Tunis, Tunisia
| |
Collapse
|
3
|
Sun P, Wang H, Liu L, Guo K, Li X, Cao Y, Ko C, Lan ZJ, Lei Z. Aberrant activation of KRAS in mouse theca-interstitial cells results in female infertility. Front Physiol 2022; 13:991719. [PMID: 36060690 PMCID: PMC9437434 DOI: 10.3389/fphys.2022.991719] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 07/28/2022] [Indexed: 11/13/2022] Open
Abstract
KRAS plays critical roles in regulating a range of normal cellular events as well as pathological processes in many tissues mediated through a variety of signaling pathways, including ERK1/2 and AKT signaling, in a cell-, context- and development-dependent manner. The in vivo function of KRAS and its downstream targets in gonadal steroidogenic cells for the development and homeostasis of reproductive functions remain to be determined. To understand the functions of KRAS signaling in gonadal theca and interstitial cells, we generated a Kras mutant (tKrasMT) mouse line that selectively expressed a constitutively active KrasG12D in these cells. KrasG12D expression in ovarian theca cells did not block follicle development to the preovulatory stage. However, tKrasMT females failed to ovulate and thus were infertile. The phosphorylated ERK1/2 and forkhead box O1 (FOXO1) and total FOXO1 protein levels were markedly reduced in tKrasMT theca cells. KrasG12D expression in theca cells also curtailed the phosphorylation of ERK1/2 and altered the expression of several ovulation-related genes in gonadotropin-primed granulosa cells. To uncover downstream targets of KRAS/FOXO1 signaling in theca cells, we found that the expression of bone morphogenic protein 7 (Bmp7), a theca-specific factor involved in ovulation, was significantly elevated in tKrasMT theca cells. Chromosome immunoprecipitation assays demonstrated that FOXO1 interacted with the Bmp7 promoter containing forkhead response elements and that the binding activity was attenuated in tKrasMT theca cells. Moreover, Foxo1 knockdown caused an elevation, whereas Foxo1 overexpression resulted in an inhibition of Bmp7 expression, suggesting that KRAS signaling regulates FOXO1 protein levels to control Bmp7 expression in theca cells. Thus, the anovulation phenotype observed in tKrasMT mice may be attributed to aberrant KRAS/FOXO1/BMP7 signaling in theca cells. Our work provides the first in vivo evidence that maintaining normal KRAS activity in ovarian theca cells is crucial for ovulation and female fertility.
Collapse
Affiliation(s)
- Penghao Sun
- Department of Andrology, The First Hospital of Jilin University, Changchun, China
| | - Hongliang Wang
- Department of Andrology, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Zhenmin Lei, ; Hongliang Wang,
| | - Lingyun Liu
- Department of Andrology, The First Hospital of Jilin University, Changchun, China
| | - Kaimin Guo
- Department of Andrology, The First Hospital of Jilin University, Changchun, China
| | - Xian Li
- Department of OB/GYN, University of Louisville School of Medicine, Louisville, KY, United States
| | - Yin Cao
- Department of Andrology, The First Hospital of Jilin University, Changchun, China
| | - Chemyong Ko
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Zi-Jian Lan
- Birth Defects Center, University of Louisville School of Dentistry, Louisville, KY, United States
| | - Zhenmin Lei
- Department of OB/GYN, University of Louisville School of Medicine, Louisville, KY, United States
- *Correspondence: Zhenmin Lei, ; Hongliang Wang,
| |
Collapse
|
4
|
Jumaa MG. PATTERN OF KRAS GENE EXPRESSION IN IRAQI WOMEN OVARIAN CARCINOMA. WIADOMOSCI LEKARSKIE (WARSAW, POLAND : 1960) 2022; 75:765-769. [PMID: 35633344 DOI: 10.36740/wlek202204103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
OBJECTIVE The aim: The goal of this study was to detect if KRAS gene and levels of had any clinical significance in the ovarian cancer by measuring levels of KRAS mRNA. PATIENTS AND METHODS Materials and methods: The investigation was conducted on 84 tissue samples from newly diagnosed patients with ovarian cancer. Twenty-eight tissue sections with benign ovarian tumors were used as a control group. The qRT- PCR technique was used for measuring and analyzing levels of KRAS mRNA. RESULTS Results: Relative increasing of KRAS mRNA level in cancer samples was statistically significant (P<0.01) when compared to benign tumors. Statistically no significant differences were found between KRAS mRNA levels and menopausal status or family history. Gene expression has been substantially connected with age groups as the highest levels of KRAS mRNA was recorded in patients with age 50-74 years (P<0.01). Endometrium tumors exhibited significant correlations (P<0.01) across histopathological tumor types. In correlation with tumor stages, stage I was substantially linked compared to stage I (P<0.01). CONCLUSION Conclusions: It was concluded that over expression of the KRAS gene is linked to early stages of ovarian cancer, which implying that mRNA levels could be used as a diagnostic and predictive factor for ovarian cancer. More research with larger groups of ovarian cancer specimens in both primary and advanced stages is needed.
Collapse
|
5
|
Changes in Transcriptomic Profiles in Different Reproductive Periods in Yaks. BIOLOGY 2021; 10:biology10121229. [PMID: 34943144 PMCID: PMC8698885 DOI: 10.3390/biology10121229] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/21/2021] [Accepted: 11/22/2021] [Indexed: 11/21/2022]
Abstract
Simple Summary The molecular regulation mechanism of yak ovarian activity has attracted extensive attention. This study investigated the global gene expression profiles in different reproductive stages (anestrus, estrus, and pregnancy) by RNA-seq technology. Enrichment analysis revealed that DEGs were involved in the process of follicular growth, ovulation, and hormone metabolism. This study explored the regulation mechanism of the yak ovary in the reproductive cycle and laid a theoretical foundation for further understanding the reproductive characteristics of yak. Abstract Yak reproductive characteristics have received extensive attention, though the molecular regulation mechanism of its ovarian activity remains to be explored. Therefore, this study initially conducted a comparative analysis of yak ovarian activities in anestrus, estrus, and pregnancy regarding their morphology and histology, followed by implementing RNA sequencing (RNA-seq) technology to detect the overall gene expression and biological mechanism in different reproductive stages. H&E staining showed that there were more growing follicles and mature follicles in ovarian tissue sections during estrus than ovarian tissues during non-estrus. The RNA-seq analysis of yak ovary tissues in three periods showed that DEGs related to follicular development and hormone metabolism were screened in the three comparison groups, such as COL1A2, NR4A1, THBS2, PTGS2, SCARB1, STAR, and WNT2B. Bioinformatics analysis showed that these DEGs are involved in ion binding, cell development, metabolic processes, enriched in ECM–receptor interactions, steroid biosynthesis, together with aldosterone generation/discharge and Wnt/PI3K-Akt signaling pathways. In addition, we speculate alternate splice development events to have important role/s in regulating ovarian functional genomic expression profiles. These results provide essential knowledge aimed at scrutinizing pivotal biomarkers for yak ovarian activity, together with paving the way for enhancing researchers’ focus on improving yak reproductive performance.
Collapse
|
6
|
Bernabò N, Di Berardino C, Capacchietti G, Peserico A, Buoncuore G, Tosi U, Crociati M, Monaci M, Barboni B. In Vitro Folliculogenesis in Mammalian Models: A Computational Biology Study. Front Mol Biosci 2021; 8:737912. [PMID: 34859047 PMCID: PMC8630647 DOI: 10.3389/fmolb.2021.737912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 10/04/2021] [Indexed: 11/27/2022] Open
Abstract
In vitro folliculogenesis (ivF) has been proposed as an emerging technology to support follicle growth and oocyte development. It holds a great deal of attraction from preserving human fertility to improving animal reproductive biotechnology. Despite the mice model, where live offspring have been achieved,in medium-sized mammals, ivF has not been validated yet. Thus, the employment of a network theory approach has been proposed for interpreting the large amount of ivF information collected to date in different mammalian models in order to identify the controllers of the in vitro system. The WoS-derived data generated a scale-free network, easily navigable including 641 nodes and 2089 links. A limited number of controllers (7.2%) are responsible for network robustness by preserving it against random damage. The network nodes were stratified in a coherent biological manner on three layers: the input was composed of systemic hormones and somatic-oocyte paracrine factors; the intermediate one recognized mainly key signaling molecules such as PI3K, KL, JAK-STAT, SMAD4, and cAMP; and the output layer molecules were related to functional ivF endpoints such as the FSH receptor and steroidogenesis. Notably, the phenotypes of knock-out mice previously developed for hub.BN indirectly corroborate their biological relevance in early folliculogenesis. Finally, taking advantage of the STRING analysis approach, further controllers belonging to the metabolic axis backbone were identified, such as mTOR/FOXO, FOXO3/SIRT1, and VEGF, which have been poorly considered in ivF to date. Overall, this in silico study identifies new metabolic sensor molecules controlling ivF serving as a basis for designing innovative diagnostic and treatment methods to preserve female fertility.
Collapse
Affiliation(s)
- Nicola Bernabò
- Unit of Basic and Applied Biosciences, University of Teramo, Teramo, Italy
- National Research Council, Institute of Biochemistry and Cell Biology, Rome, Italy
| | | | | | - Alessia Peserico
- Unit of Basic and Applied Biosciences, University of Teramo, Teramo, Italy
| | - Giorgia Buoncuore
- Unit of Basic and Applied Biosciences, University of Teramo, Teramo, Italy
| | - Umberto Tosi
- Unit of Basic and Applied Biosciences, University of Teramo, Teramo, Italy
| | - Martina Crociati
- Department of Veterinary Medicine, University of Perugia, Perugia, Italy
- Centre for Perinatal and Reproductive Medicine, University of Perugia, Perugia, Italy
| | - Maurizio Monaci
- Department of Veterinary Medicine, University of Perugia, Perugia, Italy
- Centre for Perinatal and Reproductive Medicine, University of Perugia, Perugia, Italy
| | - Barbara Barboni
- Unit of Basic and Applied Biosciences, University of Teramo, Teramo, Italy
| |
Collapse
|
7
|
Nguyen MLT, Toan NL, Bozko M, Bui KC, Bozko P. Cholangiocarcinoma Therapeutics: An Update. Curr Cancer Drug Targets 2021; 21:457-475. [PMID: 33563168 DOI: 10.2174/1568009621666210204152028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/12/2020] [Accepted: 11/20/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Cholangiocarcinoma (CCA) is the second most common hepatobiliary cancer and associated with a poor prognosis. Only one-third of CCA cases are diagnosed at operable stages. However, a high rate of relapse has been observed postoperatively. Besides screening for operable individuals, efficacious therapeutic for recurrent and advanced CCA is urgently needed. The treatment outcome of available therapeutics is important to clarify clinical indication and facilitate the development of treatment strategies. OBJECTIVE This review aims to compare the treatment outcome of different therapeutics based on both overall survival and progression-free survival. METHODS Over one hundred peer-reviewed articles were examined. We compared the treatment outcome between different treatment methods, including tumor resection with or without postoperative systematic therapy, chemotherapies including FOFLOX, and targeted therapies, such as IDH1, K-RAS, and FGFR inhibitors. Notably, the scientific basis and outcome of available treatment methods were compared with the standard first-line therapy. RESULTS CCAs at early stages should firstly undergo tumor resection surgery, followed by postoperative treatment with Capecitabine. Chemotherapy can be considered as a preoperative option for unresectable CCAs. Inoperable CCAs with genetic aberrances like FGFR alterations, IDH1, and KRAS mutations should be considered with targeted therapies. Fluoropyrimidine prodrug (S-1)/Gemcitabine/Cisplatin and nab-Paclitaxel/Gemcitabine/Cisplatin show favorable outcome which hints at the triplet regimen to be superior to Gemcitabine/Cisplatin on CCA. The triplet chemotherapeutic should be tested further compared to Gemcitabine/Cisplatin among CCAs without genetic alterations. Gemcitabine plus S-1 was recently suggested as the convenient and equivalent standard first-line for advanced/recurrent biliary tract cancer. CONCLUSION This review provides a comparative outcome between novel targeted therapies and currently available therapeutics.
Collapse
Affiliation(s)
- Mai Ly Thi Nguyen
- Department of Internal Medicine I, Universitätsklinikum Tübingen, Tübingen, Germany
| | - Nguyen Linh Toan
- Department of Pathophysiology, Vietnam Military Medical University, Hanoi, Vietnam
| | - Maria Bozko
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Poland
| | - Khac Cuong Bui
- Department of Internal Medicine I, Universitätsklinikum Tübingen, Tübingen, Germany
| | - Przemyslaw Bozko
- Department of Internal Medicine I, Universitätsklinikum Tübingen, Tübingen, Germany
| |
Collapse
|
8
|
Billhaq DH, Lee S. The Role of the Guanosine Nucleotide-Binding Protein in the Corpus Luteum. Animals (Basel) 2021; 11:1524. [PMID: 34073800 PMCID: PMC8225084 DOI: 10.3390/ani11061524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/21/2021] [Accepted: 05/23/2021] [Indexed: 12/22/2022] Open
Abstract
The corpus luteum is a temporary endocrine gland in the ovary. In the ovarian cycle, repeated patterns of specific cellular proliferation, differentiation, and transformation occur that accompany the formation and regression of the corpus luteum. Molecular mechanism events in the ovarian microenvironment, such as angiogenesis and apoptosis, are complex. Recently, we focused on the role of RAS protein in the ovarian corpus luteum. RAS protein plays a vital role in the modulation of cell survival, proliferation, and differentiation by molecular pathway signaling. Additionally, reproductive hormones regulate RAS activity in the cellular physiological function of ovarian follicles during pre-ovulatory maturation and ovulation. Thus, we have reviewed the role of RAS protein related to the biological events of the corpus luteum in the ovary.
Collapse
Affiliation(s)
| | - Seunghyung Lee
- College of Animal Life Sciences, Kangwon National University, Chuncheon 24341, Korea;
| |
Collapse
|
9
|
Sha QQ, Zhu YZ, Xiang Y, Yu JL, Fan XY, Li YC, Wu YW, Shen L, Fan HY. Role of CxxC-finger protein 1 in establishing mouse oocyte epigenetic landscapes. Nucleic Acids Res 2021; 49:2569-2582. [PMID: 33621320 PMCID: PMC7969028 DOI: 10.1093/nar/gkab107] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 02/04/2021] [Accepted: 02/08/2021] [Indexed: 12/15/2022] Open
Abstract
During oogenesis, oocytes gain competence and subsequently undergo meiotic maturation and prepare for embryonic development; trimethylated histone H3 on lysine-4 (H3K4me3) mediates a wide range of nuclear events during these processes. Oocyte-specific knockout of CxxC-finger protein 1 (CXXC1, also known as CFP1) impairs H3K4me3 accumulation and causes changes in chromatin configurations. This study investigated the changes in genomic H3K4me3 landscapes in oocytes with Cxxc1 knockout and the effects on other epigenetic factors such as the DNA methylation, H3K27me3, H2AK119ub1 and H3K36me3. H3K4me3 is overall decreased after knocking out Cxxc1, including both the promoter region and the gene body. CXXC1 and MLL2, which is another histone H3 methyltransferase, have nonoverlapping roles in mediating H3K4 trimethylation during oogenesis. Cxxc1 deletion caused a decrease in DNA methylation levels and affected H3K27me3 and H2AK119ub1 distributions, particularly at regions with high DNA methylation levels. The changes in epigenetic networks implicated by Cxxc1 deletion were correlated with the transcriptional changes in genes in the corresponding genomic regions. This study elucidates the epigenetic changes underlying the phenotypes and molecular defects in oocytes with deleted Cxxc1 and highlights the role of CXXC1 in orchestrating multiple factors that are involved in establishing the appropriate epigenetic states of maternal genome.
Collapse
Affiliation(s)
- Qian-Qian Sha
- Fertility Preservation Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou 510317, China
| | - Ye-Zhang Zhu
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Yunlong Xiang
- Center for Stem Cell Biology and Regenerative Medicine, MOE Key Laboratory of Bioinformatics, THU-PKU Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Jia-Li Yu
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Xiao-Ying Fan
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health GuangDong Laboratory), Guangzhou 510005, China
| | - Yan-Chu Li
- Fertility Preservation Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou 510317, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Yun-Wen Wu
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Li Shen
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Heng-Yu Fan
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
10
|
Metformin Prevents Follicular Atresia in Aging Laying Chickens through Activation of PI3K/AKT and Calcium Signaling Pathways. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:3648040. [PMID: 33294120 PMCID: PMC7718058 DOI: 10.1155/2020/3648040] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 08/05/2020] [Accepted: 09/17/2020] [Indexed: 12/18/2022]
Abstract
Increased follicular atresia occurs with aging and results in reduced fecundity in laying chickens. Therefore, relieving follicular atresia of aging poultry is a crucial measure to maintain sustained high laying performance. As an antiaging agent, metformin was reported to play important roles in preventing aging in diverse animals. In this study, the physiological state of the prehierarchical follicles in the peak-laying hens (D280) and aged hens (D580) was compared, followed with exploration for the possible capacity of metformin in delaying atresia of the prehierarchical follicles in the aged D580 hens. Results showed that the capacity of yolk deposition within follicles declined with aging, and the point of endoplasmic reticulum- (ER-) mitochondrion contact decreased in the ultrastructure of the follicular cells. Meanwhile, the expression of apoptosis signaling genes was increased in the atretic small white follicles. Subsequently, the H2O2-induced follicular atresia model was established to evaluate the enhancing capacity of metformin on yolk deposition and inhibition of apoptosis in the atretic small white follicles. Metformin inhibited apoptosis through regulating cooperation of the mitochondrion-associated ER membranes and the insulin (PI3K/AKT) signaling pathway. Furthermore, metformin regulated calcium ion homeostasis to relieve ER-stress and inhibited release of mitochondrion apoptosis factors (BAD and caspase). Additionally, metformin activated PI3K/AKT that suppressed activation of BAD (downstream of the insulin signaling pathway) in the atretic follicles. Further, serum estrogen level and liver estrogen receptor-α expression were increased after dietary metformin supplementation in D580 hens. These results indicated that administration of dietary metformin activated the PI3K/AKT and calcium signaling pathway and enhanced yolk deposition to prevent chicken follicular atresia.
Collapse
|
11
|
da Silva LFI, Da Broi MG, da Luz CM, da Silva LECM, Ferriani RA, Meola J, Navarro PA. miR-532-3p: a possible altered miRNA in cumulus cells of infertile women with advanced endometriosis. Reprod Biomed Online 2020; 42:579-588. [PMID: 33358886 DOI: 10.1016/j.rbmo.2020.10.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/14/2020] [Accepted: 10/15/2020] [Indexed: 12/20/2022]
Abstract
RESEARCH QUESTION Is the profile of microRNA (miRNA) altered in cumulus cells of infertile women with early (EI/II) and advanced (EIII/IV) endometriosis? DESIGN In this prospective case-control study, a miRNA profile including 754 targets was evaluated in samples of cumulus cells from infertile women with endometriosis (5 EI/II, 5 EIII/IV) and infertile controls (5, male and/or tubal factor) undergoing ovarian stimulation for intracytoplasmic sperm injection, using TaqMan® Array Human MicroRNA Cards A and B. The groups were compared with Kruskal-Wallis test, followed by Benjamini-Hochberg correction and Dunn's post hoc test. An in silico enrichment analysis was performed to list the possibly altered pathways in which the altered miRNA target genes are involved. RESULTS Only the miRNA miR-532-3p showed significant differences among the analysed groups, being down-regulated in the EIII/IV group compared with the infertile control group, as well as compared with the EI/II group. The enrichment analysis showed that some genes regulated by this miRNA are involved in important pathways for the acquisition of oocyte competence, such as the oxytocin, calcium, Wnt, FoxO, ErbB and Ras signalling pathways, as well as the oocyte meiosis pathway. CONCLUSION The present findings bring new perspectives to understanding the follicular microenvironment of infertile women with different stages of endometriosis. It is suggested that the dysregulation of miR-532-3p may be a potential mechanism involved in the aetiopathogenesis of endometriosis-related infertility. Further studies are needed to evaluate these pathways in cumulus cells of infertile women with the disease, as well as their impact on the acquisition of oocyte competence.
Collapse
Affiliation(s)
- Liliane Fabio Isidoro da Silva
- Division of Human Reproduction, Department of Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto - USP, São Paulo, Brazil
| | - Michele Gomes Da Broi
- Division of Human Reproduction, Department of Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto - USP, São Paulo, Brazil; National Institute of Hormones and Women's Health - CNPq, Porto Alegre, Brazil
| | - Caroline Mantovani da Luz
- Division of Human Reproduction, Department of Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto - USP, São Paulo, Brazil
| | - Lilian Eslaine Costa Mendes da Silva
- Division of Human Reproduction, Department of Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto - USP, São Paulo, Brazil
| | - Rui Alberto Ferriani
- Division of Human Reproduction, Department of Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto - USP, São Paulo, Brazil; National Institute of Hormones and Women's Health - CNPq, Porto Alegre, Brazil
| | - Juliana Meola
- Division of Human Reproduction, Department of Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto - USP, São Paulo, Brazil; National Institute of Hormones and Women's Health - CNPq, Porto Alegre, Brazil
| | - Paula Andrea Navarro
- Division of Human Reproduction, Department of Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto - USP, São Paulo, Brazil; National Institute of Hormones and Women's Health - CNPq, Porto Alegre, Brazil.
| |
Collapse
|
12
|
Karaer A, Tuncay G, Dogan B, Tecellioglu N, Cigremis Y. Microarray analysis of cumulus cells in women with ovarian endometriosis undergoing intracytoplasmic sperm injection. JOURNAL OF ENDOMETRIOSIS AND PELVIC PAIN DISORDERS 2020. [DOI: 10.1177/2284026520906070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective: The aim of this study was to find the significantly altered genes in cumulus cells of women with ovarian endometriosis by using microarray and quantitative polymerase chain reaction analysis. Methods: Thirty women with ovarian endometriosis and 30 age–body mass index matched controls (women with infertility as a result of pure male factor) were enrolled in this study. Cumulus cells from study participants who underwent controlled ovarian hyperstimulation were isolated mechanically. Microarray comparative genomic hybridization was used to compare the transcriptome of cumulus cells from women with ovarian endometriosis and controls. According to the different expression levels in the microarrays and their putative functions, KRAS, ZNF322, and SDHA were selected and analyzed by real-time quantitative polymerase chain reaction. Results: There was no significant difference in the basal conditions between patients with endometriosis and controls, such as age, body mass index, basal follicle stimulating hormone and estradiol levels, and total gonadotrophin dosage. The gene expression profile of cumulus cells from patients with endometriosis was significantly different from that of controls. A total of 295 genes were significantly up- or down-regulated (p-value < 0.05 and absolute fold change > 1.5). For all of the genes adjusted p-value was found to be 0.999. Polymerase chain reaction analysis showed that KRAS and ZNF322 mRNA levels in the cumulus cells of patients with ovarian endometriosis were significantly up-regulated compared to controls (fold changes: 3.05 and 3.22, respectively). Conclusion: KRAS and ZNF322 mRNA levels in the cumulus cells of patients with ovarian endometriosis were significantly up-regulated.
Collapse
Affiliation(s)
- Abdullah Karaer
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, School of Medicine, Inonu University, Malatya, Turkey
| | - Gorkem Tuncay
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, School of Medicine, Inonu University, Malatya, Turkey
| | - Berat Dogan
- Department of Biomedical Engineering, School of Engineering, Inonu University, Malatya, Turkey
| | - Nihan Tecellioglu
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, School of Medicine, Inonu University, Malatya, Turkey
| | - Yilmaz Cigremis
- Department of Medical Biology and Genetics, School of Medicine, Inonu University, Malatya, Turkey
| |
Collapse
|
13
|
Zhou X, He Y, Jiang Y, He B, Deng X, Zhang Z, Yuan X, Li J. MiR-126-3p inhibits apoptosis and promotes proliferation by targeting phosphatidylinositol 3-kinase regulatory subunit 2 in porcine ovarian granulosa cells. ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES 2019; 33:879-887. [PMID: 31480138 PMCID: PMC7206374 DOI: 10.5713/ajas.19.0290] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 08/14/2019] [Indexed: 12/14/2022]
Abstract
Objective Numerous studies have indicated that the apoptosis and proliferation of granulosa cells (GCs) are closely related to the normal growth and development of follicles and ovaries. Previous evidence has suggested that miR-126-3p might get involved in the apoptosis and proliferation of GCs, and phosphatidylinositol 3-kinase regulatory subunit 2 (PIK3R2) gene has been predicted as one target of miR-126-3p. However, the molecular regulation of miR-126-3p on PIK3R2 and the effects of PIK3R2 on porcine GCs apoptosis and proliferation remain virtually unexplored. Methods In this study, using porcine GCs as a cellular model, luciferase report assay, mutation and deletion were applied to verify the targeting relationship between miR-126-3p and PIK3R2. Annexin-V/PI staining and 5-ethynyl-2′-deoxyuridine assay were applied to explore the effect of PIK3R2 on GCs apoptosis and proliferation, respectively. Real-time quantitative polymerase chain reaction and Western Blot were applied to explore the regulation of miR-126-3p on PIK3R2 expression. Results We found that miR-126-3p targeted at PIK3R2 and inhibited its mRNA and protein expression. Knockdown of PIK3R2 significantly inhibited the apoptosis and promoted the proliferation of porcine GCs, and significantly down-regulated the mRNA expression of several key genes of PI3K pathway such as insulin-like growth factor 1 receptor (IGF1R), insulin receptor (INSR), pyruvate dehydrogenase kinase 1 (PDK1), and serine/threonine kinase 1 (AKT1). Conclusion MiR-126-3p might target and inhibit the mRNA and protein expressions of PIK3R2, thereby inhibiting GC apoptosis and promoting GC proliferation by down-regulating several key genes of the PI3K pathway, IGF1R, INSR, PDK1, and AKT1. These findings would provide great insight into further exploring the molecular regulation of miR-126-3p and PIK3R2 on the functions of GCs during the folliculogenesis in female mammals.
Collapse
Affiliation(s)
- Xiaofeng Zhou
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, National Engineering Research Centre for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Yingting He
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, National Engineering Research Centre for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Yao Jiang
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, National Engineering Research Centre for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Bo He
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, National Engineering Research Centre for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Xi Deng
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, National Engineering Research Centre for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Zhe Zhang
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, National Engineering Research Centre for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Xiaolong Yuan
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, National Engineering Research Centre for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Jiaqi Li
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, National Engineering Research Centre for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| |
Collapse
|
14
|
Kahnamouyi S, Nouri M, Farzadi L, Darabi M, Hosseini V, Mehdizadeh A. The role of mitogen-activated protein kinase-extracellular receptor kinase pathway in female fertility outcomes: a focus on pituitary gonadotropins regulation. Ther Adv Endocrinol Metab 2018; 9:209-215. [PMID: 29977499 PMCID: PMC6022971 DOI: 10.1177/2042018818772775] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 03/30/2018] [Indexed: 11/16/2022] Open
Abstract
Mammalian reproduction systems are largely regulated by the secretion of two gonadotropins, that is, luteinizing hormone (LH) and follicle-stimulating hormone (FSH). The main action of LH and FSH on the ovary is to stimulate secretion of estradiol and progesterone, which play an important role in the ovarian function and reproductive cycle control. FSH and LH secretions are strictly controlled by the gonadotropin-releasing hormone (GnRH), which is secreted from the hypothalamus into the pituitary vascular system. Maintaining normal secretion of LH and FSH is dependent on pulsatile secretion of GnRH. Extracellular signal-regulated kinase (ERK) proteins, as the main components of mitogen-activated protein kinase (MAPK) signaling pathways, are involved in the primary regulation of GnRH-stimulated transcription of the gonadotropins' α subunit in the pituitary cells. However, GnRH-stimulated expression of the β subunit has not yet been reported. Furthermore, GnRH-mediated stimulation of ERK1 and ERK2 leads to several important events such as cell proliferation and differentiation. In this review, we briefly introduce the relationship between ERK signaling and gonadotropin secretion, and its importance in female infertility.
Collapse
Affiliation(s)
- Samira Kahnamouyi
- Stem cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Laya Farzadi
- Women Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoud Darabi
- Department of Biochemistry and Clinical Laboratories, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahid Hosseini
- Department of Biochemistry and Clinical Laboratories, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
15
|
Roof AK, Gutierrez-Hartmann A. Consider the context: Ras/ERK and PI3K/AKT/mTOR signaling outcomes are pituitary cell type-specific. Mol Cell Endocrinol 2018; 463:87-96. [PMID: 28445712 DOI: 10.1016/j.mce.2017.04.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 04/20/2017] [Accepted: 04/21/2017] [Indexed: 12/11/2022]
Abstract
Conserved signaling pathways are critical regulators of pituitary homeostasis and, when dysregulated, contribute to adenoma formation. Pituitary adenomas are typically benign and rarely progress to malignant cancer. Pituitary and other neuroendocrine cell types often display non-proliferative responses to ERK and PI3K, in contrast to non-endocrine cell types which typically proliferate in response to ERK and PI3K activation. These differences likely contribute to the infrequent progression to malignancy in many endocrine tumors. In this review, we highlight the Ras/ERK and PI3K/AKT/mTOR signaling pathways in each pituitary cell type, as well as in other endocrine tissues. Furthermore, we provide evidence that a balance of ERK and PI3K signaling is required to maintain pituitary homeostasis. It is unlikely that one sole oncogene will be identified as being responsible for sporadic pituitary adenoma formation. This review emphasizes the necessity to consider endocrine cell-specific contexts and the interplay of signaling pathways to define the mechanisms underlying pituitary tumorigenesis.
Collapse
Affiliation(s)
- Allyson K Roof
- Program in Integrated Physiology and Reproductive Sciences, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, United States
| | - Arthur Gutierrez-Hartmann
- Program in Integrated Physiology and Reproductive Sciences, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, United States; Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, United States; Department of Biochemistry and Molecular Genetics, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, United States.
| |
Collapse
|
16
|
Ji SY, Liu XM, Li BT, Zhang YL, Liu HB, Zhang YC, Chen ZJ, Liu J, Fan HY. The polycystic ovary syndrome-associated gene Yap1 is regulated by gonadotropins and sex steroid hormones in hyperandrogenism-induced oligo-ovulation in mouse. Mol Hum Reprod 2017; 23:698-707. [PMID: 28961951 DOI: 10.1093/molehr/gax046] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 08/04/2017] [Indexed: 11/13/2022] Open
Affiliation(s)
- Shu-Yan Ji
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Xiao-Man Liu
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
- Center for Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China
- The Key Laboratory for Reproductive Endocrinology of the Ministry of Education, Jinan, Shandong 250001, China
| | - Bo-Tai Li
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Yin-Li Zhang
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Hong-Bin Liu
- Center for Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China
- The Key Laboratory for Reproductive Endocrinology of the Ministry of Education, Jinan, Shandong 250001, China
| | - Yu-Chao Zhang
- Center for Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China
- The Key Laboratory for Reproductive Endocrinology of the Ministry of Education, Jinan, Shandong 250001, China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China
- The Key Laboratory for Reproductive Endocrinology of the Ministry of Education, Jinan, Shandong 250001, China
| | - Junping Liu
- Institute of Aging Research, Hangzhou Normal University, Hangzhou 310058, China
| | - Heng-Yu Fan
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
17
|
Gilchrist RB, Luciano AM, Richani D, Zeng HT, Wang X, Vos MD, Sugimura S, Smitz J, Richard FJ, Thompson JG. Oocyte maturation and quality: role of cyclic nucleotides. Reproduction 2016; 152:R143-57. [PMID: 27422885 DOI: 10.1530/rep-15-0606] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 07/15/2016] [Indexed: 12/12/2022]
Abstract
The cyclic nucleotides, cAMP and cGMP, are the key molecules controlling mammalian oocyte meiosis. Their roles in oocyte biology have been at the forefront of oocyte research for decades, and many of the long-standing controversies in relation to the regulation of oocyte meiotic maturation are now resolved. It is now clear that the follicle prevents meiotic resumption through the actions of natriuretic peptides and cGMP - inhibiting the hydrolysis of intra-oocyte cAMP - and that the pre-ovulatory gonadotrophin surge reverses these processes. The gonadotrophin surge also leads to a transient spike in cAMP in the somatic compartment of the follicle. Research over the past two decades has conclusively demonstrated that this surge in cAMP is important for the subsequent developmental capacity of the oocyte. This is important, as oocyte in vitro maturation (IVM) systems practised clinically do not recapitulate this cAMP surge in vitro, possibly accounting for the lower efficiency of IVM compared with clinical IVF. This review particularly focuses on this latter aspect - the role of cAMP/cGMP in the regulation of oocyte quality. We conclude that clinical practice of IVM should reflect this new understanding of the role of cyclic nucleotides, thereby creating a new generation of ART and fertility treatment options.
Collapse
Affiliation(s)
- R B Gilchrist
- Discipline of Obstetrics and GynaecologySchool of Women's and Children's Health, University of New South Wales, Sydney, New South Wales, Australia
| | - A M Luciano
- Reproductive and Developmental Biology LaboratoryDepartment of Health, Animal Science and Food Safety, University of Milan, Milano, Italy
| | - D Richani
- Discipline of Obstetrics and GynaecologySchool of Women's and Children's Health, University of New South Wales, Sydney, New South Wales, Australia
| | - H T Zeng
- Center for Reproductive MedicineSixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - X Wang
- Discipline of Obstetrics and GynaecologySchool of Women's and Children's Health, University of New South Wales, Sydney, New South Wales, Australia Department of Obstetrics and GynaecologySt George Public Hospital, Sydney, Australia
| | - M De Vos
- Follicle Biology LaboratoryUniversity Hospital UZBrussel, Medical School, Vrije Universiteit Brussel, Brussels, Belgium
| | - S Sugimura
- Institute of AgricultureDepartment of Biological Production, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - J Smitz
- Follicle Biology LaboratoryUniversity Hospital UZBrussel, Medical School, Vrije Universiteit Brussel, Brussels, Belgium
| | - F J Richard
- Centre de Recherche en Biologie de la ReproductionDépartement des Sciences Animales, Faculté des sciences de l'agriculture et de l'alimentation, Université Laval, Québec, Canada
| | - J G Thompson
- School of MedicineRobinson Research Institute and ARC Centre of Excellence for Nanoscale BioPhotonics, The University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
18
|
Shimada M, Umehara T, Hoshino Y. Roles of epidermal growth factor (EGF)-like factor in the ovulation process. Reprod Med Biol 2016; 15:201-216. [PMID: 29259438 DOI: 10.1007/s12522-016-0236-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 02/01/2016] [Indexed: 12/22/2022] Open
Abstract
Luteinizing hormone (LH) surge stimulates preovulatory follicles to induce the ovulation process, including oocyte maturation, cumulus expansion, and granulosa cell luteinization. The matured oocytes surrounded by an expanded cumulus cell layer are released from follicles to the oviduct. However, LH receptors are dominantly expressed in granulosa cells, but less in cumulus cells and are not expressed in oocytes, indicating that the secondary factors expressed and secreted from LH-stimulated granulosa cells are required for the induction of the ovulation process. Prostaglandin and progesterone are well-known factors that are produced in granulosa cells and then stimulate in both granulosa and cumulus cells. The mutant mice of prostaglandin synthase (Ptgs2KO mice) or progesterone receptor (PRKO mice) revealed that the functions were essential to accomplish the ovulation process, but not to induce the ovulation process. To identify the factors initiating the transfer of the stimuli of LH surge from granulosa cells to cumulus cells, M. Conti's lab and our group performed microarray analysis of granulosa cells and identified the epidermal growth factor (EGF)-like factor, amphiregulin (AREG), epiregulin (EREG), and β-cellulin (BTC) that act on EGF receptor (EGFR) and then induce the ERK1/2 and Ca2+-PLC pathways in cumulus cells. When each of the pathways was down-regulated using a pharmacological approach or gene targeting study, the induction of cumulus expansion and oocyte maturation were dramatically suppressed, indicating that both pathways are inducers of the ovulation process. However, an in vitro culture study also revealed that the EGFR-induced unphysiological activation of PKC in cumulus cells accelerated oocyte maturation with low cytostatic activity. Thus, the matured oocytes are not arrested at the metaphase II (MII) stage and then spontaneously form pronuclei. The expression of another type of EGF-like factor, neuregulin 1 (NRG1), that does not act on EGFR, but selectively binds to ErbB3 is observed in granulosa cells after the LH surge. NRG1 supports EGFR-induced ERK1/2 phosphorylation, but reduces PKC activity to physiological level in the cumulus cells, which delays the timing of meiotic maturation of oocytes to adjust the timing of ovulation. Thus, both types of EGF-like factor are rapidly induced by LH surge and then stimulate cumulus cells to control ERK1/2 and PKC pathways, which results in the release of matured oocytes with a fertilization competence.
Collapse
Affiliation(s)
- Masayuki Shimada
- Laboratory of Reproductive Endocrinology, Graduate School of Biosphere Science Hiroshima University Higashi-Hiroshima 739-8528 Hiroshima Japan
| | - Takashi Umehara
- Laboratory of Reproductive Endocrinology, Graduate School of Biosphere Science Hiroshima University Higashi-Hiroshima 739-8528 Hiroshima Japan
| | - Yumi Hoshino
- Laboratory of Reproductive Endocrinology, Graduate School of Biosphere Science Hiroshima University Higashi-Hiroshima 739-8528 Hiroshima Japan
| |
Collapse
|
19
|
Abstract
In mammals, ovulation is a multistep physiological process that includes preovulatory follicle growth, oocyte meiotic maturation, cumulus-oocyte complex (COC) expansion, follicle rupture, and luteinization. TGF-β signaling pathway has multiple functions in mammalian ovary, as its complexity in ovarian function has been demonstrated by mouse models with knockouts of TGF-β receptors and SMADs. We describe the protocol that we use to study functions of TGF-β signaling pathway in follicle development and ovulation. Because total knockout of TGF-β pathway components often causes embryonic lethality, which prevents further investigation of these genes in ovarian functions, people have generated ovarian cell type-specific knockout mouse strains for TGF-β signaling pathway genes. These mouse models are also described.
Collapse
Affiliation(s)
- Chao Yu
- Life Science Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, 310058, China
| | - Jian-Jie Zhou
- Life Science Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, 310058, China
| | - Heng-Yu Fan
- Life Science Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
20
|
Booth A, Trudeau T, Gomez C, Lucia MS, Gutierrez-Hartmann A. Persistent ERK/MAPK activation promotes lactotrope differentiation and diminishes tumorigenic phenotype. Mol Endocrinol 2015; 28:1999-2011. [PMID: 25361391 DOI: 10.1210/me.2014-1168] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The signaling pathways that govern the lactotrope-specific differentiated phenotype, and those that control lactotrope proliferation in both physiological and pathological lactotrope expansion, are poorly understood. Moreover, the specific role of MAPK signaling in lactotrope proliferation vs differentiation, whether activated phosphorylated MAPK is sufficient for prolactinoma tumor formation remain unknown. Given that oncogenic Ras mutations and persistently activated phosphorylated MAPK are found in human tumors, including prolactinomas and other pituitary tumors, a better understanding of the role of MAPK in lactotrope biology is required. Here we directly examined the role of persistent Ras/MAPK signaling in differentiation, proliferation, and tumorigenesis of rat pituitary somatolactotrope GH4 cells. We stimulated Ras/MAPK signaling in a persistent, long-term manner (over 6 d) in GH4 cells using two distinct approaches: 1) a doxycycline-inducible, oncogenic V12Ras expression system; and 2) continuous addition of exogenous epidermal growth factor. We find that long-term activation of the Ras/MAPK pathway over 6 days promotes differentiation of the bihormonal somatolactotrope GH4 precursor cell into a prolactin-secreting, lactotrope cell phenotype in vitro and in vivo with GH4 cell xenograft tumors. Furthermore, we show that persistent activation of the Ras/MAPK pathway not only fails to promote cell proliferation, but also diminishes tumorigenic characteristics in GH4 cells in vitro and in vivo. These data demonstrate that activated MAPK promotes differentiation and is not sufficient to drive tumorigenesis, suggesting that pituitary lactotrope tumor cells have the ability to evade the tumorigenic fate that is often associated with Ras/MAPK activation.
Collapse
Affiliation(s)
- Allyson Booth
- Program in Reproductive Sciences and Integrated Physiology (A.B., A.G.-H.) and Departments of Medicine and of Biochemistry and Molecular Genetics (T.T., C.G., A.G.-H.) and Pathology (M.S.L.), University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado 80045
| | | | | | | | | |
Collapse
|
21
|
Booth AK, Gutierrez-Hartmann A. Signaling pathways regulating pituitary lactotrope homeostasis and tumorigenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 846:37-59. [PMID: 25472533 DOI: 10.1007/978-3-319-12114-7_2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Dysregulation of the signaling pathways that govern lactotrope biology contributes to tumorigenesis of prolactin (PRL)-secreting adenomas, or prolactinomas, leading to a state of pathological hyperprolactinemia. Prolactinomas cause hypogonadism, infertility, osteoporosis, and tumor mass effects, and are the most common type of neuroendocrine tumor. In this review, we highlight signaling pathways involved in lactotrope development, homeostasis, and physiology of pregnancy, as well as implications for signaling pathways in pathophysiology of prolactinoma. We also review mutations found in human prolactinoma and briefly discuss animal models that are useful in studying pituitary adenoma, many of which emphasize the fact that alterations in signaling pathways are common in prolactinomas. Although individual mutations have been proposed as possible driving forces for prolactinoma tumorigenesis in humans, no single mutation has been clinically identified as a causative factor for the majority of prolactinomas. A better understanding of lactotrope-specific responses to intracellular signaling pathways is needed to explain the mechanism of tumorigenesis in prolactinoma.
Collapse
Affiliation(s)
- Allyson K Booth
- Program in Reproductive Sciences and Integrated Physiology, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado, USA
| | | |
Collapse
|
22
|
Mejia R, Waite C, Ascoli M. Activation of Gq/11 in the mouse corpus luteum is required for parturition. Mol Endocrinol 2014; 29:238-46. [PMID: 25495873 DOI: 10.1210/me.2014-1324] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Mice with a deletion of Gα(q/11) in granulosa cells were previously shown to be subfertile. They also have a reduced ovulatory response due to a deficiency in the ability of the activated LH receptor to fully induce the granulosa cell progesterone receptor. Because this conditional deletion of Gα(q/11) will interfere with the actions of any G protein-coupled receptor that activates G(q/11) in granulosa or luteal cells, we sought to determine whether the actions of other hormones that contribute to fertility were also impaired. We focused our attention on prostaglandin F2 (PGF2)α, because this hormone is known to activate phospholipase C (a prominent Gα(q/11) effector) in luteal cells and because the action of PGF2α on luteal cells is the first step in the murine parturition pathway. Our data show that the conditional deletion of Gα(q/11) from granulosa cells prevents the ability of PGF2α to induce Akr1c18 in luteal cells. Akr1c18 codes for 20α-hydroxysteroid dehydrogenase, an enzyme that inactivates progesterone. The PGF2α-mediated induction of this enzyme towards the end of pregnancy increases the inactivation of progesterone and precipitates parturition in mice. Thus, the conditional deletion of Gαq/11 from granulosa/luteal cells prevents the progesterone withdrawal that occurs at the end of pregnancy and impairs parturition. This novel molecular defect contributes to the subfertile phenotype of the mice with a deletion of Gα(q/11) from granulosa cells.
Collapse
Affiliation(s)
- Rachel Mejia
- Department of Obstetrics and Gynecology (R.M., M.A.), Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242; and Department of Pharmacology (C.W., M.A.), Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242
| | | | | |
Collapse
|
23
|
Zhang YL, Xia Y, Yu C, Richards JS, Liu J, Fan HY. CBP-CITED4 is required for luteinizing hormone-triggered target gene expression during ovulation. Mol Hum Reprod 2014; 20:850-60. [PMID: 24878634 DOI: 10.1093/molehr/gau040] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Pituitary-secreted luteinizing hormone (LH) induces ovulation by activating an extracellular regulated kinase 1/2 (ERK1/2) cascade. However, little is known regarding the ERK1/2 downstream effectors that are involved in regulating rapid, transient expression of LH-target gene in ovulatory follicles. By comparing the gene expression profiles of LH-stimulated wild type with ERK1/2-deleted ovarian granulosa cells (GCs), we identified Cited4 as a previously unknown LH target gene during ovulation. LH induced Cited4 expression in pre-ovulatory follicles in an ERK1/2-dependent manner. CITED4 formed an endogenous protein complex and docked on the promoters of LH and ERK1/2 target genes. Both CITED4 expression and CBP acetyltransferase activity leading to histone acetylation were indispensable for LH-induced ovulation-related events. LH induced dynamic histone acetylation changes in pre-ovulatory GCs, including the acetylation of histone H2B (Lys5) and H3 (Lys9). This was essential for the rapid responses and dramatic increases of LH target gene expressions by the ordered activation of ERK1/2 and CITED4-CBP. In addition, histone deacetylases (HDACs) antagonized CITED4-CBP to turn off expression of these genes after exposure to LH. Thus, we determined that CITED4 was a novel LH and ERK1/2 target for triggering ovulation. These results support the proposition that LH induces rapid, significant gene expression in pre-ovulatory follicles by modulating histone acetylation status.
Collapse
Affiliation(s)
- Yin-Li Zhang
- Life Sciences Institute and Innovation Center for Cell Biology, Zhejiang University, Hangzhou, China
| | - Yan Xia
- Life Sciences Institute and Innovation Center for Cell Biology, Zhejiang University, Hangzhou, China
| | - Chao Yu
- Life Sciences Institute and Innovation Center for Cell Biology, Zhejiang University, Hangzhou, China
| | - JoAnne S Richards
- Department of Cellular and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Junping Liu
- Institute of Aging Research, Hangzhou Normal University, Hangzhou, China
| | - Heng-Yu Fan
- Life Sciences Institute and Innovation Center for Cell Biology, Zhejiang University, Hangzhou, China
| |
Collapse
|
24
|
Rosario R, Cohen PA, Shelling AN. The role of FOXL2 in the pathogenesis of adult ovarian granulosa cell tumours. Gynecol Oncol 2014; 133:382-7. [DOI: 10.1016/j.ygyno.2013.12.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 12/05/2013] [Accepted: 12/09/2013] [Indexed: 12/12/2022]
|
25
|
Maurya VK, Sangappa C, Kumar V, Mahfooz S, Singh A, Rajender S, Jha RK. Expression and activity of Rac1 is negatively affected in the dehydroepiandrosterone induced polycystic ovary of mouse. J Ovarian Res 2014; 7:32. [PMID: 24628852 PMCID: PMC3995551 DOI: 10.1186/1757-2215-7-32] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 02/22/2014] [Indexed: 02/07/2023] Open
Abstract
Background Polycystic ovarian syndrome (PCOS) is characterized by the presence of multiple follicular cysts, giving rise to infertility due to anovulation. This syndrome affects about 10% of women, worldwide. The exact molecular mechanism leading to PCOS remains obscure. RhoGTPase has been associated with oogenesis, but its role in PCOS remains unexplored. Therefore, we attempted to elucidate the Vav-Rac1 signaling in PCOS mice model. Methods We generated a PCOS mice model by injecting dehydroepiandrosterone (DHEA) for a period of 20 days. The expression levels of Rac1, pRac1, Vav, pVav and Caveolin1 were analyzed by employing immuno-blotting and densitometry. The association between Vav and Rac1 proteins were studied by immuno-precipitation. Furthermore, we analyzed the activity of Rac1 and levels of inhibin B and 17β-estradiol in ovary using biochemical assays. Results The presence of multiple follicular cysts in ovary were confirmed by histology. The activity of Rac1 (GTP bound state) was significantly reduced in the PCOS ovary. Similarly, the expression levels of Rac1 and its phosphorylated form (pRac1) were decreased in PCOS in comparison to the sham ovary. The expression level and activity (phosphorylated form) of guanine nucleotide exchanger of Rac1, Vav, was moderately down-regulated. We observed comparatively increased expressions of Caveolin1, 17β-estradiol, and inhibin B in the polycystic ovary. Conclusion We conclude that hyperandrogenization (PCOS) by DHEA diminishes ovarian Rac1 and Vav expression and activity along with an increase in expression of Caveolin1. This is accompanied by an increase in the intra-ovarian level of '17 β-estradiol and inhibin B.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Rajesh Kumar Jha
- Division of Endocrinology, Life Science North 111B/101, CSIR-Central Drug Research Institute, B,S, 10/1, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India.
| |
Collapse
|
26
|
Sominsky L, Sobinoff AP, Jobling MS, Pye V, McLaughlin EA, Hodgson DM. Immune regulation of ovarian development: programming by neonatal immune challenge. Front Neurosci 2013; 7:100. [PMID: 23781169 PMCID: PMC3679471 DOI: 10.3389/fnins.2013.00100] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Accepted: 05/22/2013] [Indexed: 12/20/2022] Open
Abstract
Neonatal immune challenge by administration of lipopolysaccharide (LPS) produces enduring alterations in the development and activity of neuroendocrine, immune and other physiological systems. We have recently reported that neonatal exposure to an immune challenge by administration of LPS results in altered reproductive development in the female Wistar rat. Specifically, LPS-treated animals exhibited diminished ovarian reserve and altered reproductive lifespan. In the current study, we examined the cellular mechanisms that lead to the previously documented impaired ovulation and reduced follicular pool. Rats were administered intraperitoneally either 0.05 mg/kg of LPS (Salmonella Enteritidis) or an equivalent volume of non-pyrogenic saline on postnatal days (PNDs) 3 and 5, and ovaries were obtained on PND 7. Microarray analysis revealed a significant upregulation in transcript expression (2-fold change; p < 0.05) for a substantial number of genes in the ovaries of LPS-treated animals, implicated in immune cell signaling, inflammatory responses, reproductive system development and disease. Several canonical pathways involved in immune recognition were affected by LPS treatment, such as nuclear factor-κB (NF-κB) activation and LPS-stimulated mitogen-activated protein kinase (MAPK) signaling. Quantitative Real-time PCR analysis supported the microarray results. Protein expression analysis of several components of the MAPK signaling pathway revealed a significant upregulation in the expression of Toll-like receptor 4 (TLR4) in the neonatal ovary of LPS-treated animals. These results indicate that neonatal immune challenge by administration of LPS has a direct effect on the ovary during the sensitive period of follicular formation. Given the pivotal role of inflammatory processes in the regulation of reproductive health, our findings suggest that early life immune activation via TLR signaling may have significant implications for the programming of ovarian development and fertility.
Collapse
Affiliation(s)
- Luba Sominsky
- Laboratory of Neuroimmunology, Faculty of Science and IT, School of Psychology, The University of Newcastle Callaghan, NSW, Australia
| | | | | | | | | | | |
Collapse
|
27
|
Yu C, Zhang YL, Fan HY. Selective Smad4 knockout in ovarian preovulatory follicles results in multiple defects in ovulation. Mol Endocrinol 2013; 27:966-78. [PMID: 23592428 DOI: 10.1210/me.2012-1364] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The TGF-β signaling pathway is involved with multiple processes in the mammalian ovary, including primordial follicle formation, granulosa cell (GC) proliferation, follicle atresia, ovulation, and feedback regulation between the pituitary and ovary. The transcriptional factor SMAD4 (Sma- and Mad-related protein 4) is the central component of the canonical TGF-β signaling pathway. Smad4 knockout (KO) using Amhr2-Cre, which is expressed in GCs of immature developing follicles, causes premature luteinization. In this study, we specifically depleted Smad4 in GCs of preovulatory follicles using Cyp19-Cre mice. As different from results with Smad4(fl/fl);Amhr2-Cre mice, Smad4 depletion in preovulatory follicles did not cause premature luteinization or suppress GC proliferation; rather, it increased follicle atresia. In addition, Nppc and Npr2 expressions were reduced by Smad4 depletion; thus, their effect of maintaining oocyte meiotic arrest was weakened in Smad4 conditional KO mice. Smad4(fl/fl);Cyp19-Cre female mice were subfertile and had irregular estrous cycles and ovulation defects. Smad4 KO also blocked LH-induced cumulus expansion and follicle rupture, but not oocyte meiotic resumption. Our results also indicated that SMAD4 was required for LH-stimulated activation of ERK1/2 and the expressions of ovulation-related genes. The defects arising from SMAD4 depletion could not be rescued by intraovarian mediators of LH actions, such as epidermal growth factor-like factors and prostaglandin E2. Furthermore, corpus lutea did not form in Smad4(fl/fl);Cyp19-Cre female mice, indicating that SMAD4 was crucial for GCs terminal differentiation. Thus, by characterizing the ovarian phenotypes of preovulatory follicle-specific Smad4 KO mice, we identified the developmental stage-specific functions of the canonical TGF-β signaling pathway in ovulation and luteinization.
Collapse
Affiliation(s)
- Chao Yu
- Life Sciences Institute, Zhejiang University, 866 Yu Hang Tang Road, Hangzhou, China 310058
| | | | | |
Collapse
|
28
|
Mielczarek-Palacz A, Sikora J, Kondera-Anasz Z, Hauza G. Imbalance in serum soluble CD30/CD30L and CD40/CD40L systems are associated with ovarian tumors. Hum Immunol 2013; 74:70-4. [DOI: 10.1016/j.humimm.2012.10.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Revised: 09/04/2012] [Accepted: 10/03/2012] [Indexed: 01/12/2023]
|
29
|
Fan HY, Liu Z, Mullany LK, Richards JS. Consequences of RAS and MAPK activation in the ovary: the good, the bad and the ugly. Mol Cell Endocrinol 2012; 356:74-9. [PMID: 22197887 PMCID: PMC3327778 DOI: 10.1016/j.mce.2011.12.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Revised: 12/05/2011] [Accepted: 12/06/2011] [Indexed: 11/29/2022]
Abstract
This review summarizes studies providing evidence (1) that endogenous RAS activation regulates important physiological events during ovulation and luteinization (2) that expression of the mutant, active KRAS(G12D) in granulosa cells in vivo causes abnormal follicle growth arrest leading to premature ovarian failure and (3) that KRAS(G12D) expression in ovarian surface epithelial (OSE) cells renders them susceptible to the pathological outcome of transformation and tumor formation. These diverse effects of RAS highlight how critical its activation is linked to cell- and stage-specific events in the ovary that control normal processes and that can also lead to altered granulosa cell and OSE cell fates.
Collapse
Affiliation(s)
- Heng-Yu Fan
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, United States
| | | | | | | |
Collapse
|
30
|
Zheng W, Nagaraju G, Liu Z, Liu K. Functional roles of the phosphatidylinositol 3-kinases (PI3Ks) signaling in the mammalian ovary. Mol Cell Endocrinol 2012; 356:24-30. [PMID: 21684319 DOI: 10.1016/j.mce.2011.05.027] [Citation(s) in RCA: 127] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Accepted: 05/10/2011] [Indexed: 12/31/2022]
Abstract
Phosphatidylinositol 3-kinase (PI3K) signaling is a fundamental pathway for the regulation of cell proliferation, survival, migration, and metabolism in a variety of physiological and pathological processes. In recent years information provided by genetically modified mouse models has revealed that PI3K signaling plays vital roles in oogenesis, folliculogenesis, ovulation, and carcinogenesis in mouse ovary. In this review, we summarize (1) the physiological function of intra-oocyte PI3K signaling in regulation of primordial follicle survival and activation; (2) intra-granulosa cell PI3K signaling in regulation of cyclic follicular recruitment and ovulation; (3) intra-oocyte PI3K signaling in regulation of meiosis resumption and early embryogenesis; and also (4) the pathological function of PI3K signaling in ovarian diseases such as premature ovarian failure, granulosa cell tumors, and ovarian surface epithelium carcinomas. This updated info hopefully will lead to a better understanding of the human ovary and provide potential therapies for treating human infertility.
Collapse
Affiliation(s)
- Wenjing Zheng
- Department of Cell and Molecular Biology, University of Gothenburg, Gothenburg SE-40530, Sweden.
| | | | | | | |
Collapse
|
31
|
Shimada M. Regulation of oocyte meiotic maturation by somatic cells. Reprod Med Biol 2012; 11:177-184. [PMID: 29662364 DOI: 10.1007/s12522-012-0130-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2012] [Accepted: 04/26/2012] [Indexed: 01/21/2023] Open
Abstract
In preovulatory follicles, each oocyte is surrounded by numerous layers of cumulus cells, forming the cumulus cell-oocyte complex. An LH surge induces meiotic resumption of the oocyte to progress to metaphase II. Because the expression of LH receptors is not detected in the oocyte and is minimal (negligible) in cumulus cells as compared with granulosa cells, secondary factors from granulosa cells are required to induce the ovulation process. One of the key factors secreted from granulosa cells is an EGF-like factor that activates the EGFR-ERK1/2 pathway in cumulus cells. The activated ERK1/2 pathway is not only involved in gene expression but also essential for the close of gap-junctional communication among cumulus cells and between cumulus cells and the oocyte. Closing gap-junctional communication decreases the amount of cGMP and/or cAMP to transfer into the oocyte, which requires activation of phosphodiesterase type III (PDE3) in the oocyte. PDE3 brakes down cAMP to decrease PKA activity in the oocyte. This decrease in PKA activity induces activation of CDK1 to resume meiosis from the germinal vesicle stage. Thus, the functions of cumulus cells that are regulated by granulosa cell-secreted factors are essential for oocyte meiotic resumption and maturation with developmental competence.
Collapse
Affiliation(s)
- Masayuki Shimada
- Laboratory of Reproductive Endocrinology, Graduate School of Biosphere Science Hiroshima University Higashi 739-8528 Hiroshima Hiroshima Japan
| |
Collapse
|
32
|
Richards JS, Fan HY, Liu Z, Tsoi M, Laguë MN, Boyer A, Boerboom D. Either Kras activation or Pten loss similarly enhance the dominant-stable CTNNB1-induced genetic program to promote granulosa cell tumor development in the ovary and testis. Oncogene 2012; 31:1504-20. [PMID: 21860425 PMCID: PMC3223552 DOI: 10.1038/onc.2011.341] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Revised: 06/22/2011] [Accepted: 07/03/2011] [Indexed: 12/16/2022]
Abstract
WNT, RAS or phosphoinositide 3-kinase signaling pathways control specific stages of ovarian follicular development. To analyze the functional interactions of these pathways in granulosa cells during follicular development in vivo, we generated specific mutant mouse models. Stable activation of the WNT signaling effector β-catenin (CTNNB1) in granulosa cells results in the formation of premalignant lesions that develop into granulosa cell tumors (GCTs) spontaneously later in life or following targeted deletion of the tumor suppressor gene Pten. Conversely, expression of oncogenic KRAS(G12D) dramatically arrests proliferation, differentiation and apoptosis in granulosa cells, and consequently, small abnormal follicle-like structures devoid of oocytes accumulate in the ovary. Because of the potent anti-proliferative effects of KRAS(G12D) in granulosa cells, we sought to determine whether KRAS(G12D) would block precancerous lesion and tumor formation in follicles of the CTNNB1-mutant mice. Unexpectedly, transgenic Ctnnb1;Kras-mutant mice exhibited increased GC proliferation, decreased apoptosis and impaired differentiation and developed early-onset GCTs leading to premature death in a manner similar to the Ctnnb1;Pten-mutant mice. Microarray and reverse transcription-PCR analyses revealed that gene regulatory processes induced by CTNNB1 were mostly enhanced by either KRAS activation or Pten loss in remarkably similar patterns and degree. The concomitant activation of CTNNB1 and KRAS in Sertoli cells also caused testicular granulosa cell tumors that showed gene expression patterns that partially overlapped those observed in GCTs of the ovary. Although the mutations analyzed herein have not yet been linked to adult GCTs in humans, they may be related to juvenile GCTs or to tumors in other tissues where CTNNB1 is mutated. Importantly, the results provide strong evidence that CTNNB1 is the driver in these contexts and that KRAS(G12D) and Pten loss promote the program set in motion by the CTNNB1.
Collapse
Affiliation(s)
- J S Richards
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| | | | | | | | | | | | | |
Collapse
|
33
|
Hu MW, Wang ZB, Schatten H, Sun QY. New understandings on folliculogenesis/oogenesis regulation in mouse as revealed by conditional knockout. J Genet Genomics 2012; 39:61-8. [PMID: 22361505 DOI: 10.1016/j.jgg.2012.01.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2011] [Revised: 12/28/2011] [Accepted: 01/05/2012] [Indexed: 01/27/2023]
Abstract
In comparison to conventional knockout technology and in vitro research methods, conditional gene knockout has remarkable advantages. In the past decade, especially during the past five years, conditional knockout approaches have been used to study the regulation of folliculogenesis, follicle growth, oocyte maturation and other major reproductive events. In this review, we summarize the recent findings about folliculogenesis/oogenesis regulation, including the functions of four signaling cascades or glycoprotein domains that have been extensively studied by conditional gene deletion. Several other still fragmented areas of related work are introduced which are awaiting clarification. We have also discussed the future potential of this technology in clarifying gene functions in reproductive biology.
Collapse
Affiliation(s)
- Meng-Wen Hu
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | | | | | | |
Collapse
|
34
|
Abstract
Adult granulosa cell tumor is often a hormonally active stromal cell neoplasm of the ovary with malignant potential. Intra-operative pathological assessment is a valuable tool in guiding optimal surgical treatment in patients. Of the various intra-operative cytological diagnostic modalities, scrape smear cytology is an effective, economical, simple, fast and reliable method with results comparable with frozen section diagnosis. We describe a case of adult granulosa cell tumor in a 30-years-old lady diagnosed on intra-operative scrape cytology, and further reconfirmed on frozen section and histopathology.
Collapse
Affiliation(s)
- Prabal Deb
- Department of Pathology, Armed Forces Medical College, India
| | | | | |
Collapse
|
35
|
Shimada M, Yamashita Y. The Key Signaling Cascades in Granulosa Cells During Follicular Development and Ovulation Process. ACTA ACUST UNITED AC 2011. [DOI: 10.1274/jmor.28.25] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
36
|
Noma N, Kawashima I, Fan HY, Fujita Y, Kawai T, Tomoda Y, Mihara T, Richards JS, Shimada M. LH-induced neuregulin 1 (NRG1) type III transcripts control granulosa cell differentiation and oocyte maturation. Mol Endocrinol 2010; 25:104-16. [PMID: 21047912 DOI: 10.1210/me.2010-0225] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Epidermal growth factor (EGF)-like factors [amphiregulin (AREG), betacellulin, and epiregulin] are induced by LH and activate the EGF receptor (ERBB1)/ERK1/2 pathway in granulosa cells and cumulus cells of preovulatory follicles to impact ovulation. However, the expression and roles of other ERBB family members and their ligands have not been explored in detail. Herein, we document that two transcripts of the neuregulin (Nrg1) gene are expressed in granulosa cells, and that the type III Nrg1 is induced during ovulation in an ERK1/2 and C/EBPβ-dependent manner. Western blotting shows that intact (75 kDa) and secreted (45 kDa) forms of neuregulin 1 (NRG1) are present in the ovary. NRG1 likely binds to ERBB3/ERBB2 complexes that are expressed in granulosa cells and cumulus cells. In cultured granulosa cells, NRG1 selectively stimulates the phosphorylation of AKT/PKB compared to ERK1/2. However, when granulosa cells were cultured with NRG1 and AREG, the phosphorylation of ERK1/2 was markedly enhanced as compared with that by AREG alone. Cotreatment with NRG1 and AREG also increased progesterone production. When cumulus-oocyte complexes (COCs) were cultured with both NRG1 and AREG, the matured oocytes exhibited significantly higher developmental competence as compared with that of oocytes cultured with AREG alone. Collectively, these results document that the expression of type III NRG1 is induced in granulosa cells during ovulation and that NRG1 enhances AREG-induced ERK1/2 phosphorylation in both granulosa cells and cumulus cells. The NRG1 pathway has two roles: one is to enhance AREG-induced progesterone production in granulosa cells, and the other is to regulate oocyte maturation by a cumulus cell-dependent mechanism.
Collapse
Affiliation(s)
- Noritaka Noma
- Laboratory of Reproductive Endocrinology, Graduate School of Biosphere Science, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Luo J, McGinnis LK, Kinsey WH. Role of Fyn kinase in oocyte developmental potential. Reprod Fertil Dev 2010; 22:966-76. [PMID: 20591331 DOI: 10.1071/rd09311] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2009] [Accepted: 01/20/2010] [Indexed: 12/22/2022] Open
Abstract
Fyn kinase is highly expressed in oocytes, with inhibitor and dominant-negative studies suggesting a role in the signal transduction events during egg activation. The purpose of the present investigation was to test the hypothesis that Fyn is required for calcium signalling, meiosis resumption and pronuclear congression using the Fyn-knockout mouse as a model. Accelerated breeding studies revealed that Fyn-null females produced smaller litter sizes at longer intervals and exhibited a rapid decline in pup production with increasing age. Fyn-null females produced a similar number of oocytes, but the frequency of immature oocytes and mature oocytes with spindle chromosome abnormalities was significantly higher than in controls. Fertilised Fyn-null oocytes frequently (24%) failed to undergo pronuclear congression and remained at the one-cell stage. Stimulation with gonadotropins increased the number of oocytes ovulated, but did not overcome the above defects. Fyn-null oocytes overexpressed Yes kinase in an apparent effort to compensate for the loss of Fyn, yet still exhibited an altered pattern of protein tyrosine phosphorylation. In summary, Fyn-null female mice exhibit reduced fertility that appears to result from actin cytoskeletal defects rather than calcium signalling. These defects cause developmental arrest during oocyte maturation and pronuclear congression.
Collapse
Affiliation(s)
- Jinping Luo
- Center for Reproductive Sciences, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | | | | |
Collapse
|