1
|
Li M, Liang W, Luo Y, Wang J, Liu X, Li S, Hao Z. Transforming growth factor-β1 mediates the SMAD4/BMF pathway to regulate ovarian granulosa cell apoptosis in small tail Han sheep. Theriogenology 2024; 214:360-369. [PMID: 37979327 DOI: 10.1016/j.theriogenology.2023.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/23/2023] [Accepted: 11/08/2023] [Indexed: 11/20/2023]
Abstract
Transforming growth factor (TGF)-β1 is an important multifunctional cytokine in the TGF-β signaling pathway, which is involved in the molecular regulation of multiple activities, including follicle development and ovulation in female reproductive physiology. However, the biological function of TGF-β1 in follicular development and in regulating the proliferation or apoptosis of granulosa cells in small tail Han sheep remain unclear. In this study, we analyzed the expression levels of TGF-β1 in the ovary at the follicular stage in small tail Han sheep. We further examined the effects of TGF-β1 on the viability, proliferation, and apoptosis of granulosa cells. Differential expression of TGF-β1 at the mRNA and protein levels was detected in the ovaries between the beginning of estrus and at preovulation. Cell Counting Kit-8, 5-ethynyl-2'-deoxyuridine (EdU) labeling, and flow cytometry assays showed that adding 5 and 10 ng/mL TGF-β1 could improve the viability and growth rate, reduce the apoptosis rate, and reduce the expression level of the pro-apoptotic factor Bcl-2-modifying factor (BMF) in granulosa cells. Treatment of 10 ng/mL TGF-β1 at all time points (except 72 h) significantly increased the positive rate of EdU labeling compared to that of the control group. RNA interference of SMAD4 reversed the decreased apoptosis rate caused by stimulation with 10 ng/mL TGF-β1, accompanied by a corresponding increase in the BMF expression level. Collectively, these results indicate that TGF-β1 plays a role in the ovarian follicular-phase activity of small tail Han sheep by inhibiting the apoptosis of sheep granulosa cells through the SMAD4/BMF pathway to promote proliferation and vitality. This study provides new insight into the molecular mechanism underlying TGF-β1 function regulation in granulosa cells, suggests a new target for the regulation of follicle development, and expands the new field of animal reproduction regulation technology.
Collapse
Affiliation(s)
- Mingna Li
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China.
| | - Weiwei Liang
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Yuzhu Luo
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Jiqing Wang
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China.
| | - Xiu Liu
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Shaobin Li
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Zhiyun Hao
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| |
Collapse
|
2
|
Upregulation of miR-33 Exacerbates Heat-Stress-Induced Apoptosis in Granulosa Cell and Follicular Atresia of Nile Tilapia (Oreochromis niloticus) by Targeting TGFβ1I1. Genes (Basel) 2022; 13:genes13061009. [PMID: 35741771 PMCID: PMC9222912 DOI: 10.3390/genes13061009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/18/2022] [Accepted: 05/23/2022] [Indexed: 02/04/2023] Open
Abstract
High temperature affects egg quality and increases follicular atresia in teleosts. The present study aimed to explore the regulated mechanism of ovary syndrome of Nile tilapia (Oreochromis niloticus) exposed to heat stress. To this end, we conducted histological and biochemical analyses and integrated miRNA-target gene analyses. The histochemical analyses confirmed that heat stress promoted the apoptosis of granulosa cell and therefore resulted in increased follicular atresia in the ovary. Heat stress led to the differential expression of multiple miRNAs (miR-27e, -27b-3p, -33, -34a -133a-5p, and -301b-5p). In a luciferase activity assay, miR-33 bound to the 3′-untranslated region (UTR) of the TGFβ1I1 (transforming growth factor-β1-induced transcript 1) gene and inhibited its expression. A TGFβ1I1 gene signal was detected in the granulosa cells of Nile tilapia by immunohistochemical analysis. Up-regulation of the miR-33 of tilapia at 6 d and 12 d exposed to heat (34.5 °C ± 0.5 °C) had significant down-regulation of the TGFβ1I1 expression of the gene and protein in tilapia ovaries. An miRNA-target gene integrated analysis revealed that miR-33 and TGFβ1I1 function in an apoptosis-related signal pathway. The signal transduction of the vascular endothelial growth factor (VEGF) family members VEGFA and its receptor (KDR) in the heat-stressed group decreased significantly compared with the control group. Transcript-levels of the Bax and Caspase-3 as apoptotic promotors were activated and Bcl-2 and Caspase-8 as apoptotic inhibitors were suppressed in the heat-stressed tilapia. These results suggest that heat stress increases the expression of miR-33, which targets TGFβ1I1 and inhibits its expression, resulting in decreased levels of follicle-stimulating hormone and 17β-estradiol and increased apoptosis by suppressing VEGF signaling, eventually inducing follicular atresia. In conclusion, our results show that the miR-33/TGFβ1I1 axis of Nile tilapia is involved in the follicular development of broodstock, and can suppress VEGF signaling to accelerate follicular atresia. Our findings demonstrate the suppressive role of miR-33 during oocyte development in Nile tilapia.
Collapse
|
3
|
Ni N, Fang X, Mullens DA, Cai JJ, Ivanov I, Bartholin L, Li Q. Transcriptomic Profiling of Gene Expression Associated with Granulosa Cell Tumor Development in a Mouse Model. Cancers (Basel) 2022; 14:2184. [PMID: 35565312 PMCID: PMC9105549 DOI: 10.3390/cancers14092184] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/05/2022] [Accepted: 04/24/2022] [Indexed: 11/16/2022] Open
Abstract
Ovarian granulosa cell tumors (GCTs) are rare sex cord-stromal tumors, accounting for ~5% ovarian tumors. The etiology of GCTs remains poorly defined. Genetically engineered mouse models are potentially valuable for understanding the pathogenesis of GCTs. Mice harboring constitutively active TGFβ signaling (TGFBR1-CA) develop ovarian GCTs that phenocopy several hormonal and molecular characteristics of human GCTs. To determine molecular alterations in the ovary upon TGFβ signaling activation, we performed transcriptomic profiling of gene expression associated with GCT development using ovaries from 1-month-old TGFBR1-CA mice and age-matched controls. RNA-sequencing and bioinformatics analysis coupled with the validation of select target genes revealed dysregulations of multiple cellular events and signaling molecules/pathways. The differentially expressed genes are enriched not only for known GCT-related pathways and tumorigenic events but also for signaling events potentially mediated by neuroactive ligand-receptor interaction, relaxin signaling, insulin signaling, and complements in TGFBR1-CA ovaries. Additionally, a comparative analysis of our data in mice with genes dysregulated in human GCTs or granulosa cells overexpressing a mutant FOXL2, the genetic hallmark of adult GCTs, identified some common genes altered in both conditions. In summary, this study has revealed the molecular signature of ovarian GCTs in a mouse model that harbors the constitutive activation of TGFBR1. The findings may be further exploited to understand the pathogenesis of a class of poorly defined ovarian tumors.
Collapse
Affiliation(s)
- Nan Ni
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843, USA; (N.N.); (X.F.); (J.J.C.)
| | - Xin Fang
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843, USA; (N.N.); (X.F.); (J.J.C.)
| | - Destiny A. Mullens
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA; (D.A.M.); (I.I.)
| | - James J. Cai
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843, USA; (N.N.); (X.F.); (J.J.C.)
| | - Ivan Ivanov
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA; (D.A.M.); (I.I.)
| | - Laurent Bartholin
- INSERM U1052, CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Université Lyon 1, F-69000 Lyon, France;
- Centre Léon Bérard, F-69008 Lyon, France
| | - Qinglei Li
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843, USA; (N.N.); (X.F.); (J.J.C.)
| |
Collapse
|
4
|
Li M, Liang W, Zhu C, Qin S. Smad4 mediates Bmf involvement in sheep granulosa cell apoptosis. Gene 2022; 817:146231. [PMID: 35063577 DOI: 10.1016/j.gene.2022.146231] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 12/23/2021] [Accepted: 01/14/2022] [Indexed: 12/23/2022]
Abstract
Bcl-2-modifying factor (Bmf) functions to mediate follicular atresia and oocyte growth in mice. It has been proven that TGF-β can induce Bmf expression via the Smad4 pathway in a variety of cells, and then induce cell apoptosis. Based on this, we hypothesized that Smad4 and Bmf may play important roles in the apoptosis of granulosa cells (GCs) in domestic animals. This study used small-tailed Han sheep follicular GCs cultured in vitro as a model system, and overexpression or interference experiments, to explore the biological roles of Bmf and reveal the preliminary regulatory mechanisms between Smad4 and Bmf in the process of GCs' apoptosis. We found that the proliferation rate of sheep GCs was significantly increased after the knockdown of Bmf, whereas overexpressing Bmf increased the apoptosis rate of GCs, results also verified by the expression patterns of PCNA, Bcl-2, and Bax genes. After the Smad4 knockdown, the apoptosis rate of GCs was increased, while the mRNA and protein expression of Bmf was significantly up-regulated. A rescue experiment verified that the Bmf knockdown could alleviate GCs' apoptosis induced by Smad4 knockdown. In conclusion, our study not only elucidated an important role for Bmf in the apoptosis of sheep GCs but also revealed a new regulatory pathway between Smad4 and Bmf in this process.
Collapse
Affiliation(s)
- Mingna Li
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, Faculty of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China..
| | - Weiwei Liang
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, Faculty of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Caiye Zhu
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Shizhen Qin
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| |
Collapse
|
5
|
Li Q, Du X, Wang L, Shi K, Li Q. TGF-β1 controls porcine granulosa cell states: A miRNA-mRNA network view. Theriogenology 2020; 160:50-60. [PMID: 33181481 DOI: 10.1016/j.theriogenology.2020.11.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 10/31/2020] [Accepted: 11/03/2020] [Indexed: 12/14/2022]
Abstract
TGF-β1, an important multi-functional cytokine of the TGF-β signaling pathway, has been reported to be crucial for ovarian granulosa cell (GC) states and female fertility. However, the molecular mechanism underlying TGF-β1 regulation of GC states remains largely unknown. Here, we provide a comprehensive transcriptomic view on TGF-β1 regulation of cell states in porcine GCs. We first confirmed that TGF-β1 can control GC states (apoptosis and proliferation) in pig ovary. RNA-seq showed that 909 differentially expressed genes (DEGs), including 890 DEmRNAs and 19 DEmiRNAs, were identified in TGF-β1-treated porcine GCs. Functional annotation showed that these DEGs were mainly involved in regulating cell states. In addition, multiple hub genes were identified by constructing the protein-protein interaction network, DEmiRNA-DEmRNAs regulatory network, and gene-pathway-function co-expression networks, which were further found to be enriched in FoxO, TGF-β, Wnt, PIK3-Akt, p53 and Ras signaling pathways that play important roles in regulating cell states, cell cycle, proliferation, stress-responses and inflammation. The current research deeply reveals the effects of TGF-β1 on porcine GCs, and also identifies potential therapeutic RNA molecules for inhibiting and rescuing female infertility.
Collapse
Affiliation(s)
- Qiqi Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Xing Du
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Lingfang Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Kerong Shi
- College of Animal Science and Technology, Shandong Agricultural University, Taian, 271018, China
| | - Qifa Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
6
|
Shi X, Tang T, Lin Q, Liu H, Qin Y, Liang X, Cong P, Mo D, Liu X, Chen Y, He Z. Efficient generation of bone morphogenetic protein 15-edited Yorkshire pigs using CRISPR/Cas9†. Biol Reprod 2020; 103:1054-1068. [PMID: 32761111 DOI: 10.1093/biolre/ioaa138] [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: 02/18/2019] [Revised: 06/25/2020] [Accepted: 08/06/2020] [Indexed: 12/15/2022] Open
Abstract
Bone morphogenetic protein 15 (BMP15), a member of the transforming growth factor beta superfamily, plays an essential role in ovarian follicular development in mono-ovulatory mammalian species. Studies using a biallelic knockout mouse model revealed that BMP15 potentially has just a minimal impact on female fertility and ovarian follicular development in polyovulatory species. In contrast, our previous study demonstrated that in vivo knockdown of BMP15 significantly affected porcine female fertility, as evidenced by the dysplastic ovaries containing significantly decreased numbers of follicles and an increased number of abnormal follicles. This finding implied that BMP15 plays an important role in the regulation of female fertility and ovarian follicular development in polyovulatory species. To further investigate the regulatory role of BMP15 in porcine ovarian and follicular development, here, we describe the efficient generation of BMP15-edited Yorkshire pigs using CRISPR/Cas9. Using artificial insemination experiments, we found that the biallelically edited gilts were all infertile, regardless of different genotypes. One monoallelically edited gilt #4 (Δ66 bp/WT) was fertile and could deliver offspring with a litter size comparable to that of wild-type gilts. Further analysis established that the infertility of biallelically edited gilts was caused by the arrest of follicular development at preantral stages, with formation of numerous structurally abnormal follicles, resulting in streaky ovaries and the absence of obvious estrous cycles. Our results strongly suggest that the role of BMP15 in nonrodent polyovulatory species may be as important as that in mono-ovulatory species.
Collapse
Affiliation(s)
- Xuan Shi
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P.R. China
| | - Tao Tang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P.R. China
| | - Qiyuan Lin
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P.R. China
| | - Hongbo Liu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P.R. China
| | - Yufeng Qin
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P.R. China
| | - Xinyu Liang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P.R. China
| | - Peiqing Cong
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P.R. China
| | - Delin Mo
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P.R. China
| | - Xiaohong Liu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P.R. China
| | - Yaosheng Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P.R. China
| | - Zuyong He
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P.R. China
| |
Collapse
|
7
|
miR-130a/TGF-β1 axis is involved in sow fertility by controlling granulosa cell apoptosis. Theriogenology 2020; 157:407-417. [PMID: 32871445 DOI: 10.1016/j.theriogenology.2020.08.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 08/05/2020] [Accepted: 08/12/2020] [Indexed: 12/16/2022]
Abstract
TGF-β1 is a ligand of the TGF-β superfamily and an important cytokine that regulates ovarian functions including follicular development, steroid production, ovulation, luteinization, and female fertility. However, little is known about the regulation of TGF-β1 expression in ovary. Here, we identified that TGF-β1 is a functional target of miR-130a in porcine ovarian granulosa cells (GCs). The 3'-UTR sequence of TGF-β1 gene (1137 bp in length) in Large White (LW) pig was isolated, and multiple RNA regulatory elements (RREs), including several binding motifs of different miRNAs, were identified in this region. Luciferase activity assay showed that miR-130a dramatically suppresses the 3'-UTR luciferase activity of TGF-β1 gene, and further inhibits the expression of TGF-β1 in porcine GCs. FACS revealed that miR-130a acts as a pro-apoptotic factor and promotes GC apoptosis by inhibiting TGF-β1. Two novel linked mutations (-573G > A and -540T > C) were identified in the promoter region of ssc-miR-130a, but their polymorphisms are not associated with sow reproductive traits. Importantly, combined genotype analysis with a known mutation (c.1583 A > G) in the 3'-UTR of porcine TGF-β1 gene showed a significant association with reproductive performance in LW sow population. Overall, our findings defined a novel regulatory axis, miR-130a/TGF-β1 axis, which is involved in regulating sow fertility.
Collapse
|
8
|
MiR-126* is a novel functional target of transcription factor SMAD4 in ovarian granulosa cells. Gene 2019; 711:143953. [PMID: 31269463 DOI: 10.1016/j.gene.2019.143953] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 06/28/2019] [Accepted: 06/28/2019] [Indexed: 01/06/2023]
Abstract
Both SMAD4 and miR-126* have been proven to be involved in granulosa cell (GC) apoptosis and even follicular atresia, through commonly regulating follicle-stimulating hormone receptor (FSHR), the FSH-specific transmembrane receptor of GCs. However, the regulatory relationship between them in GCs is still unknown. In this study, we report that SMAD4 suppresses the expression of miR-126* and impairs its function in GCs of the porcine ovary by acting as a transcription factor. A classic SMAD4-binding element (SBE) site was found in the promoter of miR-126* by using in silico methods. Luciferase assay, qRT-PCR, and ChIP assay proved that SMAD4 serves as a transcriptional repressor and directly binds to SBE site within miR-126* gene promoter, which further reduces miR-126* gene expression and inhibits its transcriptional activity in GCs. Furthermore, SMAD4 also controls miR-126*-mediated expression of FSHR (a direct target of miR-126* in GCs). In addition, we prove that SMAD4 induces CYP19A1 expression (encodes aromatase, the key enzyme for oestrogen biosynthesis) and inhibits GC apoptosis through the miR-126*/FSHR axis. Taken together, our findings not only established a direct link between SMAD4 and miRNA-126*, two key factors of GC apoptosis, but also revealed an important way in which the SMAD4 regulates GC function, the miRNA-126*/FSHR axis.
Collapse
|
9
|
Prognostic Impact of Canonical TGF-β Signaling in Urothelial Bladder Cancer. ACTA ACUST UNITED AC 2019; 55:medicina55060302. [PMID: 31238579 PMCID: PMC6630377 DOI: 10.3390/medicina55060302] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 06/12/2019] [Accepted: 06/20/2019] [Indexed: 01/12/2023]
Abstract
Background and objectives: Dysregulation of TGF-β signaling plays multiple roles in cancer development and progression. In the canonical TGF-β pathway, TGF-β regulates the expression of hundreds of target genes via interaction with Smads, signal transducers and transcriptional modulators. We evaluated the association of TGF-β1, Smad2, and Smad4, the key components of canonical TGFβ pathway, with clinicopathologic characteristics of urothelial bladder cancer, and assessed their prognostic value in prediction of patients’ outcome. Materials and Methods: Immunohistochemical analysis of TGF-β1, Smad2, and Smad4 expression was performed on 404 urothelial bladder cancer samples, incorporated in tissue microarrays. Expression status was correlated with clinicopathological and follow-up data. The median follow-up was 61 months. Results: High expression of TGF-β1, Smad2, and Smad4 was detected in 68.1%, 31.7% and 45.2% of the tumors, respectively. TGF-β1 overexpression was significantly associated with high tumor grade, and advanced pathologic stage (p < 0.001, respectively). Conversely, high Smad2 and Smad4 expression was linked to low tumor grade (p = 0,003, p = 0.048, respectively), and low tumor stage (p < 0.001, p = 0.003, respectively). Smad2 showed an inverse correlation with variant morphology and divergent differentiation of urothelial tumors (p = 0.014). High TGF-β1 correlated directly, while Smad2 and Smad4 correlated inversely to cancer-specific death (p = 0.043, p = 0.003, and p = 0.022, respectively). There was a strong relationship between Smad2 and Smad4 expression (p < 0.001). Survival analyses showed that high Smad2 and Smad4 expression was associated with longer overall survival (p = 0.003, p = 0.034, respectively), while in multivariate regression analysis TGF-β1 manifested as an independent predictor of poor outcome. Conclusions: Unraveling the complex roles and significance of TGF-β signaling in urothelial bladder cancer might have important implications for therapy of this disease. Assessment of TGF-β pathway status in patients with urothelial bladder cancer may provide useful prognostic information, and identify patients that could have the most benefit from therapy targeting TGF-β signaling cascade.
Collapse
|
10
|
Zare H, Shafabakhsh R, Reiter RJ, Asemi Z. Melatonin is a potential inhibitor of ovarian cancer: molecular aspects. J Ovarian Res 2019; 12:26. [PMID: 30914056 PMCID: PMC6434863 DOI: 10.1186/s13048-019-0502-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 03/18/2019] [Indexed: 12/13/2022] Open
Abstract
Ovarian cancer is one of the most common causes of morbidity related to gynecologic malignancies. Possible risk factors are including hereditary ovarian cancer, obesity, diabetes mellitus, alcohol consumption, aging, and smoking. Various molecular signaling pathways including inflammation, oxidative stress, apoptosis and angiogenesis are involved in this progression of ovarian cancer. Standard treatments for recently diagnosed patients are Surgery and chemotherapy such as co-treatment with other drugs such that the exploitation of neoadjuvant chemotherapy is expanding. Melatonin (N-acetyl-5-methoxy-tryptamine), an endogenous agent secreted from the pineal gland, has anti-carcinogenic features, such as regulation of estradiol production, cell cycle modulation, stimulation of apoptosis as well as anti-angiogenetic properties, anti-inflammatory activities, significant antioxidant effects and modulation of various immune system cells and cytokines. Multiple studies have shown the significant beneficial roles of melatonin in various types of cancers including ovarian cancer. This paper aims to shed light on the roles of melatonin in ovarian cancer treatment from the standpoint of the molecular aspects.
Collapse
Affiliation(s)
- Hadis Zare
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R, Iran
| | - Rana Shafabakhsh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R, Iran
| | - Russel J Reiter
- Department of Cellular and Structural Biology, University of Texas Health Science, Center, San Antonio, TX, USA
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R, Iran.
| |
Collapse
|
11
|
Rimon-Dahari N, Heinemann-Yerushalmi L, Hadas R, Kalich-Philosoph L, Ketter D, Nevo N, Galiani D, Dekel N. Vasorin: a newly identified regulator of ovarian folliculogenesis. FASEB J 2018; 32:2124-2136. [PMID: 29259033 DOI: 10.1096/fj.201700057rrr] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Members of the TGF-β superfamily take part in the control of folliculogenesis. Vasorin (Vasn) is a newly identified negative regulator of TGF-β signaling whose possible involvement in ovarian physiology has never been studied. Here, we demonstrate that Vasn is expressed in the ovary by somatic cells of follicles, and that its expression is up-regulated by LH. We established a conditional knockout (cKO) mouse model in which Vasn is deleted specifically in granulosa cells of growing follicles from the secondary stage onwards. Using this model, we show that, upon hormonal stimulation, follicle ovulation size is almost 2-fold higher. This enhanced ovulatory response is associated with overactivation of the TGF-β signaling pathway and a lower number of atretic antral follicles. Of importance, we demonstrate that the number of primordial follicles is reduced in prepubertal cKO mouse ovaries, which suggests that the production of VASN by growing follicles protects the ovarian reserve. Finally, analysis of systemic KO mice revealed that the ovarian reserve is almost 2.5-fold higher, which implies that Vasn may also play a role in primordial follicle formation. Overall, our findings reveal that Vasn is a new regulator that exerts an effect on several key ovarian functions, including folliculogenesis, maintenance of the ovarian reserve, and ovulation.-Rimon-Dahari, N., Heinemann-Yerushalmi, L., Hadas, R., Kalich-Philosoph, L., Ketter, D., Nevo, N., Galiani, D., Dekel, N. Vasorin: a newly identified regulator of ovarian folliculogenesis.
Collapse
Affiliation(s)
- Nitzan Rimon-Dahari
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | | | - Ron Hadas
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | | | - Dafna Ketter
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Nava Nevo
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Dalia Galiani
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Nava Dekel
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
12
|
Li J, Bao R, Peng S, Zhang C. The molecular mechanism of ovarian granulosa cell tumors. J Ovarian Res 2018; 11:13. [PMID: 29409506 PMCID: PMC5802052 DOI: 10.1186/s13048-018-0384-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 01/30/2018] [Indexed: 12/12/2022] Open
Abstract
Over these years, more and more sex cord-stromal tumors have been reported. Granulosa cell tumor (GCT) is a rare tumor in ovaries, accounts for 2% to 5% of ovarian cancers. The main different feature of GCTs from other ovarian cancers is that GCTs can lead to abnormally secreted hormones (estrogen, inhibin and Müllerian inhibiting substance). The GCT is divided into two categories according to the age of patients, namely AGCT (adult granulosa cell tumor) and JGCT (Juvenile granulosa cell tumor). AGCT patients accounts for 95%. Although the pathogenesis is not clear, FOXL2 (Forkhead box L2) mutation was considered as the most critical factor in AGCT development. The current treatment is dominated by surgery. Target therapy remains in the adjuvant therapy stage, such as hormone therapy. During these years, other pathogenic factors were also explored, such as PI3K/AKT (phosphatidylinositol-3-kinase; serine/threonine kinase), TGF-β (Transforming growth factor beta) signaling pathway, Notch signaling pathway, GATA4 and VEGF (vascular endothelial growth factor). These factors and signaling pathway play important roles in GCT cell proliferation, apoptosis, or angiogenesis. The purpose of this review is to summarize the possible pathogenic factors and signaling pathways, which may shed lights on developing potential therapeutic targets for GCT.
Collapse
Affiliation(s)
- Jiaheng Li
- Joint programme of Nanchang University and Queen Mary University of London, Nanchang, China
| | - Riqiang Bao
- Joint programme of Nanchang University and Queen Mary University of London, Nanchang, China
| | - Shiwei Peng
- Department of Gynecology and Obstetrics, Jiangxi Provincial People's Hospital, Nanchang, China
| | - Chunping Zhang
- Department of Cell Biology, School of Medicine, Nanchang University, Nanchang, Jiangxi, 330006, People's Republic of China.
| |
Collapse
|
13
|
Wang M, Sun J, Xu B, Chrusciel M, Gao J, Bazert M, Stelmaszewska J, Xu Y, Zhang H, Pawelczyk L, Sun F, Tsang SY, Rahman N, Wolczynski S, Li X. Functional Characterization of MicroRNA-27a-3p Expression in Human Polycystic Ovary Syndrome. Endocrinology 2018; 159:297-309. [PMID: 29029022 DOI: 10.1210/en.2017-00219] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 09/14/2017] [Indexed: 11/19/2022]
Abstract
The goal of this study was to characterize the function of microRNA-27a-3p (miR-27a-3p) in polycystic ovary syndrome (PCOS). miR-27a-3p expression was analyzed in excised granulosa cells (GCs) from 21 patients with PCOS and 12 normal patients undergoing in vitro fertilization cycle treatments and in 17 nontreated cuneiform ovarian resection PCOS samples and 13 control ovarian samples from patients without PCOS. We found that the expression of miR-27a-3p was significantly increased in both excised GCs and the ovaries of patients with PCOS compared with the controls. Insulin treatment of the human granulosa-like tumor cell line (KGN) resulted in decreased downregulated expression of miR-27a-3p, and this effect appeared to be mediated by signal transducer and activator of transcription STAT1 and STAT3. The overexpression of miR-27a-3p in KGN cells inhibited SMAD5, which in turn decreased cell proliferation and promoted cell apoptosis. After KGN cells were stimulated with insulin for 48 hours, there was increased expression of SMAD5 protein and decreased apoptosis. Additionally, knockdown/overexpression of SMAD5 in KGN cells reduced/increased cell number and promoted/inhibited cell apoptosis. Insulin-stimulated primary GCs isolated from patients with PCOS, in contrast to normal GCs or KGN cells, did not exhibit decreased miR-27a-3p expression. The differences in the expression levels in KGN cells and human PCOS GCs are likely explained by increased miR-27a-3p expression in the GCs caused by insulin resistance in PCOS. Taken together, our data provided evidence for a functional role of miR-27a-3p in the GCs' dysfunction that occurs in patients with PCOS.
Collapse
Affiliation(s)
- Mingming Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Jing Sun
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Bo Xu
- Center for Reproductive Medicine, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei, Anhui, China
| | - Marcin Chrusciel
- Institute of Biomedicine, Department of Physiology, University of Turku, Turku, Finland
| | - Jun Gao
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Maciej Bazert
- Department of Infertility and Reproductive Endocrinology, Poznan University of Medical Sciences, Poznan, Poland
| | - Joanna Stelmaszewska
- Department of Reproduction and Gynecological Endocrinology, Medical University of Bialystok, Bialystok, Poland
| | - Yunyun Xu
- Department of General Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Hongwen Zhang
- Department of General Surgery, 306th Hospital of People's Liberation Army of China, Beijing, China
| | - Leszek Pawelczyk
- Department of Infertility and Reproductive Endocrinology, Poznan University of Medical Sciences, Poznan, Poland
| | - Fei Sun
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Suk Ying Tsang
- School of Life Science and State Key Laboratory of Agro-Biotechnology, Chinese University of Hong Kong, Hong Kong 999077, China
| | - Nafis Rahman
- Institute of Biomedicine, Department of Physiology, University of Turku, Turku, Finland
- Department of Reproduction and Gynecological Endocrinology, Medical University of Bialystok, Bialystok, Poland
| | - Slawomir Wolczynski
- Department of Reproduction and Gynecological Endocrinology, Medical University of Bialystok, Bialystok, Poland
| | - Xiangdong Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| |
Collapse
|
14
|
Gao Y, Fang X, Vincent DF, Threadgill DW, Bartholin L, Li Q. Disruption of postnatal folliculogenesis and development of ovarian tumor in a mouse model with aberrant transforming growth factor beta signaling. Reprod Biol Endocrinol 2017; 15:94. [PMID: 29221447 PMCID: PMC5723096 DOI: 10.1186/s12958-017-0312-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 11/26/2017] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Transforming growth factor beta (TGFB) superfamily signaling is implicated in the development of sex cord-stromal tumors, a category of poorly defined gonadal tumors. The aim of this study was to determine potential effects of dysregulated TGFB signaling in the ovary using Cre recombinase driven by growth differentiation factor 9 (Gdf9) promoter known to be expressed in oocytes. METHODS A mouse model containing constitutively active TGFBR1 (TGFBR1CA) using Gdf9-iCre (termed TGFBR1-CAG9Cre) was generated. Hematoxylin and eosin (H & E) staining, follicle counting, and immunohistochemistry and immunofluorescence analyses using antibodies directed to Ki67, forkhead box L2 (FOXL2), forkhead box O1 (FOXO1), inhibin alpha (INHA), and SRY (sex determining region Y)-box 9 were performed to determine the characteristics of the TGFBR1-CAG9Cre ovary. Terminal deoxynucleotidyl transferase (TdT) labeling of 3'-OH ends of DNA fragments, real-time PCR, and western blotting were used to examine apoptosis, select gene expression, and TGFBR1 activation. RNAscope in situ hybridization was used to localize the expression of GLI-Kruppel family member GLI1 (Gli1) in ovarian tumor tissues. RESULTS TGFBR1-CAG9Cre females were sterile. Sustained activation of TGFBR1 led to altered granulosa cell proliferation evidenced by high expression of Ki67. At an early age, these mice demonstrated follicular defects and development of ovarian granulosa cell tumors, which were immunoreactive for granulosa cell markers including FOXL2, FOXO1, and INHA. Further histochemical and molecular analyses provided evidence of overactivation of TGFBR1 in the granulosa cell compartment during ovarian pathogenesis in TGFBR1-CAG9Cre mice, along with upregulation of Gli1 and Gli2 and downregulation of Tgfbr3 in ovarian tumor tissues. CONCLUSIONS These results reinforce the role of constitutively active TGFBR1 in promoting ovarian tumorigenesis in mice. The mouse model created in this study may be further exploited to define the cellular and molecular mechanisms of TGFB/activin downstream signaling in granulosa cell tumor development. Future studies are needed to test whether activation of TGFB/activin signaling contributes to the development of human granulosa cell tumors.
Collapse
Affiliation(s)
- Yang Gao
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, 77843, USA
| | - Xin Fang
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, 77843, USA
| | - David F Vincent
- Cancer Research UK Beatson Institute, Garscube Estate, G61 1BD, Glasgow, UK
| | - David W Threadgill
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, 77843, USA
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, Texas A&M University, College Station, TX, 77843, USA
| | - Laurent Bartholin
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR5286, Université Lyon 1, Centre Léon Bérard, F-69000, Lyon, France
| | - Qinglei Li
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, 77843, USA.
| |
Collapse
|
15
|
Monsivais D, Matzuk MM, Pangas SA. The TGF-β Family in the Reproductive Tract. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a022251. [PMID: 28193725 DOI: 10.1101/cshperspect.a022251] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The transforming growth factor β (TGF-β) family has a profound impact on the reproductive function of various organisms. In this review, we discuss how highly conserved members of the TGF-β family influence the reproductive function across several species. We briefly discuss how TGF-β-related proteins balance germ-cell proliferation and differentiation as well as dauer entry and exit in Caenorhabditis elegans. In Drosophila melanogaster, TGF-β-related proteins maintain germ stem-cell identity and eggshell patterning. We then provide an in-depth analysis of landmark studies performed using transgenic mouse models and discuss how these data have uncovered basic developmental aspects of male and female reproductive development. In particular, we discuss the roles of the various TGF-β family ligands and receptors in primordial germ-cell development, sexual differentiation, and gonadal cell development. We also discuss how mutant mouse studies showed the contribution of TGF-β family signaling to embryonic and postnatal testis and ovarian development. We conclude the review by describing data obtained from human studies, which highlight the importance of the TGF-β family in normal female reproductive function during pregnancy and in various gynecologic pathologies.
Collapse
Affiliation(s)
- Diana Monsivais
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas 77030.,Center for Drug Discovery, Baylor College of Medicine, Houston, Texas 77030
| | - Martin M Matzuk
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas 77030.,Center for Drug Discovery, Baylor College of Medicine, Houston, Texas 77030.,Department of Molecular and Cellular Biology, Baylor College of Medicine Houston, Texas 77030.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030.,Department of Pharmacology, Baylor College of Medicine, Houston, Texas 77030
| | - Stephanie A Pangas
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas 77030.,Center for Drug Discovery, Baylor College of Medicine, Houston, Texas 77030.,Department of Molecular and Cellular Biology, Baylor College of Medicine Houston, Texas 77030
| |
Collapse
|
16
|
Fullerton PT, Monsivais D, Kommagani R, Matzuk MM. Follistatin is critical for mouse uterine receptivity and decidualization. Proc Natl Acad Sci U S A 2017; 114:E4772-E4781. [PMID: 28559342 PMCID: PMC5474784 DOI: 10.1073/pnas.1620903114] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Embryo implantation remains a significant challenge for assisted reproductive technology, with implantation failure occurring in ∼50% of in vitro fertilization attempts. Understanding the molecular mechanisms underlying uterine receptivity will enable the development of new interventions and biomarkers. TGFβ family signaling in the uterus is critical for establishing and maintaining pregnancy. Follistatin (FST) regulates TGFβ family signaling by selectively binding TGFβ family ligands and sequestering them. In humans, FST is up-regulated in the decidua during early pregnancy, and women with recurrent miscarriage have lower endometrial expression of FST during the luteal phase. Because global knockout of Fst is perinatal lethal in mice, we generated a conditional knockout (cKO) of Fst in the uterus using progesterone receptor-cre to study the roles of uterine Fst during pregnancy. Uterine Fst-cKO mice demonstrate severe fertility defects and deliver only 2% of the number of pups delivered by control females. In Fst-cKO mice, the uterine luminal epithelium does not respond properly to estrogen and progesterone signals and remains unreceptive to embryo attachment by continuing to proliferate and failing to differentiate. The uterine stroma of Fst-cKO mice also responds poorly to artificial decidualization, with lower levels of proliferation and differentiation. In the absence of uterine FST, activin B expression and signaling are up-regulated, and bone morphogenetic protein (BMP) signals are impaired. Our findings support a model in which repression of activin signaling by FST enables uterine receptivity by preserving critical BMP signaling.
Collapse
Affiliation(s)
- Paul T Fullerton
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030
- Center for Reproductive Medicine, Baylor College of Medicine, Houston, TX 77030
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX 77030
| | - Diana Monsivais
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030
- Center for Reproductive Medicine, Baylor College of Medicine, Houston, TX 77030
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX 77030
| | - Ramakrishna Kommagani
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| | - Martin M Matzuk
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030;
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030
- Center for Reproductive Medicine, Baylor College of Medicine, Houston, TX 77030
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
- Department of Pharmacology, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
17
|
Sun W, Xu Y, Zhao C, Hao F, Chen D, Guan J, Zhang K. Targeting TGF-β1 suppresses survival of and invasion by anaplastic thyroid carcinoma cells. Am J Transl Res 2017; 9:1418-1425. [PMID: 28386367 PMCID: PMC5376032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 02/14/2017] [Indexed: 06/07/2023]
Abstract
BACKGROUND AND AIMS Overexpression of transforming growth factor (TGF)-β1 has been implicated in promoting cell survival, migration and invasion in many cancers, including anaplastic thyroid cancer (ATC). In the present study, we studied the effect of suppressing TGF-β1 by RNA silencing on the survival, invasion and metastasis of ATC cells. METHODS Small interfering RNA (siRNA) constructs targeting TGF-β1 were validated and used to develop clonal derivatives of the ATC cell line, 8505C. The cells were used in several in vitro assays, including migration, invasion, survival rate, colony formation and apoptosis. A wound healing assay was used to determine the migration of cells in culture and a Boyden chamber transwell assay was used for invasion. Further, clones were used in an in vivo mouse model to study the kinetics of tumor growth and metastatic growth in lungs. RESULTS Targeting TGF-β1 expression in 8505C cells caused a 70% decrease in migration and a 78% decrease in invasion, as well as a 68% decrease in proliferation and a 19% increase in apoptosis in vitro. The growth of primary tumors in vivo was also inhibited when compared with parental 8505C cells; however, the number of mice bearing lung metastases was not significantly decreased. CONCLUSIONS Targeting TGF-β1 may be effective in inhibiting primary tumor formation, but not metastasis, by ATC cells. TGF-β1 inhibition in combination with other tumor-targeted therapies may be more effective in inhibiting ATC.
Collapse
Affiliation(s)
- Wenhai Sun
- Department of Thyroid Surgery, The Affiliated Hospital of Qingdao UniversityQingdao, China
| | - Yanyan Xu
- Department of Clinical Laboratory, People’s Hospital of ChengyangQingdao, China
| | - Cheng Zhao
- Department of Ultrasonic Diagnosis, The Affiliated Hospital of Qingdao UniversityQingdao, China
| | - Fengyun Hao
- Department of Pathology, The Affiliated Hospital of Qingdao UniversityQingdao, China
| | - Dong Chen
- Department of Pathology, The Affiliated Hospital of Qingdao UniversityQingdao, China
| | - Jinping Guan
- Department of General Surgery, The Affiliated Hospital of Qingdao UniversityQingdao, China
| | - Kejun Zhang
- Department of Thyroid Surgery, The Affiliated Hospital of Qingdao UniversityQingdao, China
| |
Collapse
|
18
|
Fang X, Gao Y, Li Q. SMAD3 Activation: A Converging Point of Dysregulated TGF-Beta Superfamily Signaling and Genetic Aberrations in Granulosa Cell Tumor Development? Biol Reprod 2016; 95:105. [PMID: 27683263 PMCID: PMC5178148 DOI: 10.1095/biolreprod.116.143412] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 08/10/2016] [Accepted: 09/21/2016] [Indexed: 12/30/2022] Open
Abstract
Ovarian granulosa cell tumors (GCTs) are rare gynecologic tumors in women. Due to the rarity and limited research efforts invested, the etiology of GCTs remains poorly defined. A landmark study has discovered the mutation of forkhead box L2 (FOXL2) as a genetic hallmark of adult GCTs in the human. However, our understanding of the role of cell signaling in GCT development is far from complete. Increasing lines of evidence highlight the importance of TGF-beta (TGFB) superfamily signaling in the pathogenesis of GCTs. This review draws on findings using genetically modified mouse models and human patient specimens and cell lines to reveal SMAD3 activation as a potentially key converging point of dysregulated TGFB superfamily signaling and genetic aberrations in GCT development. It is anticipated that deciphering the role of TGFB superfamily signaling cascades in ovarian tumorigenesis will help develop new therapeutic approaches for GCTs by targeting core signaling elements essential for tumor initiation, growth, and progression.
Collapse
Affiliation(s)
- Xin Fang
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas
| | - Yang Gao
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas
| | - Qinglei Li
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas
| |
Collapse
|
19
|
Gao Y, Vincent DF, Davis AJ, Sansom OJ, Bartholin L, Li Q. Constitutively active transforming growth factor β receptor 1 in the mouse ovary promotes tumorigenesis. Oncotarget 2016; 7:40904-40918. [PMID: 27344183 PMCID: PMC5173031 DOI: 10.18632/oncotarget.10149] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 06/06/2016] [Indexed: 12/11/2022] Open
Abstract
Despite the well-established tumor suppressive role of TGFβ proteins, depletion of key TGFβ signaling components in the mouse ovary does not induce a growth advantage. To define the role of TGFβ signaling in ovarian tumorigenesis, we created a mouse model expressing a constitutively active TGFβ receptor 1 (TGFBR1) in ovarian somatic cells using conditional gain-of-function approach. Remarkably, these mice developed ovarian sex cord-stromal tumors with complete penetrance, leading to reproductive failure and mortality. The tumors expressed multiple granulosa cell markers and caused elevated serum inhibin and estradiol levels, reminiscent of granulosa cell tumors. Consistent with the tumorigenic effect, overactivation of TGFBR1 altered tumor microenvironment by promoting angiogenesis and enhanced ovarian cell proliferation, accompanied by impaired cell differentiation and dysregulated expression of critical genes in ovarian function. By further exploiting complementary genetic models, we substantiated our finding that constitutively active TGFBR1 is a potent oncogenic switch in mouse granulosa cells. In summary, overactivation of TGFBR1 drives gonadal tumor development. The TGFBR1 constitutively active mouse model phenocopies a number of morphological, hormonal, and molecular features of human granulosa cell tumors and are potentially valuable for preclinical testing of targeted therapies to treat granulosa cell tumors, a class of poorly defined ovarian malignancies.
Collapse
Affiliation(s)
- Yang Gao
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - David F. Vincent
- Cancer Research UK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| | - Anna Jane Davis
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Owen J. Sansom
- Cancer Research UK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| | - Laurent Bartholin
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR5286, Lyon, France
| | - Qinglei Li
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| |
Collapse
|
20
|
Rodriguez A, Tripurani SK, Burton JC, Clementi C, Larina I, Pangas SA. SMAD Signaling Is Required for Structural Integrity of the Female Reproductive Tract and Uterine Function During Early Pregnancy in Mice. Biol Reprod 2016; 95:44. [PMID: 27335065 PMCID: PMC5029477 DOI: 10.1095/biolreprod.116.139477] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 06/10/2016] [Indexed: 12/20/2022] Open
Abstract
Pregnancy is a complex physiological process tightly controlled by the interplay among hormones, morphogens, transcription factors, and signaling pathways. Although recent studies using genetically engineered mouse models have revealed that ligands and receptors of transforming growth factor beta (TGFbeta) and bone morphogenetic protein (BMP) signaling pathways are essential for multiple reproductive events during pregnancy, the functional role of SMAD transcription factors, which serve as the canonical signaling platform for the TGFbeta/BMP pathways, in the oviduct and uterus is undefined. Here, we used a mouse model containing triple conditional deletion of the BMP receptor signaling Smads (Smad1 and Smad5) and Smad4, the central mediator of both TGFbeta and BMP signaling, to investigate the role of the SMADs in reproductive tract structure and function in cells from the Amhr2 lineage. Unlike the respective single- or double-knockouts, female Smad1(flox/flox) Smad5(flox/flox) Smad4(flox/flox) Amhr2(cre/+)conditional knockout (i.e., Smad1/5/4-Amhr2-cre KO) mice are sterile. We discovered that Smad1/5/4-Amhr2-cre KO females have malformed oviducts that subsequently develop oviductal diverticuli. These oviducts showed dysregulation of multiple genes essential for oviduct and smooth muscle development. In addition, uteri from Smad1/5/4-Amhr2-cre KO females exhibit multiple defects in stroma, epithelium, and smooth muscle layers and fail to assemble a closed uterine lumen upon embryo implantation, with defective uterine decidualization that led to pregnancy loss at early to mid-gestation. Taken together, our study uncovers a new role for the SMAD transcription factors in maintaining the structural and functional integrity of oviduct and uterus, required for establishment and maintenance of pregnancy.
Collapse
Affiliation(s)
- Amanda Rodriguez
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas Graduate Program in Molecular and Cell Biology, Baylor College of Medicine, Houston, Texas
| | - Swamy K Tripurani
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas
| | - Jason C Burton
- Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, Texas
| | - Caterina Clementi
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, Texas
| | - Irina Larina
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas
| | - Stephanie A Pangas
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas Center for Reproductive Medicine, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
21
|
Kim SY. Insights into granulosa cell tumors using spontaneous or genetically engineered mouse models. Clin Exp Reprod Med 2016; 43:1-8. [PMID: 27104151 PMCID: PMC4838576 DOI: 10.5653/cerm.2016.43.1.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 02/24/2016] [Accepted: 03/04/2016] [Indexed: 12/26/2022] Open
Abstract
Granulosa cell tumors (GCTs) are rare sex cord-stromal tumors that have been studied for decades. However, their infrequency has delayed efforts to research their etiology. Recently, mutations in human GCTs have been discovered, which has led to further research aimed at determining the molecular mechanisms underlying the disease. Mouse models have been important tools for studying GCTs, and have provided means to develop and improve diagnostics and therapeutics. Thus far, several genetically modified mouse models, along with one spontaneous mouse model, have been reported. This review summarizes the phenotypes of these mouse models and their applicability in elucidating the mechanisms of granulosa cell tumor development.
Collapse
Affiliation(s)
- So-Youn Kim
- Division of Reproductive Science in Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
22
|
Wang X, Chen E, Tang M, Yang X, Wang Y, Quan Z, Wu X, Luo C. The SMAD2/3 pathway is involved in hepaCAM-induced apoptosis by inhibiting the nuclear translocation of SMAD2/3 in bladder cancer cells. Tumour Biol 2016; 37:10731-43. [DOI: 10.1007/s13277-016-4821-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Accepted: 01/08/2016] [Indexed: 11/25/2022] Open
|
23
|
Liu Z, Ren YA, Pangas SA, Adams J, Zhou W, Castrillon DH, Wilhelm D, Richards JS. FOXO1/3 and PTEN Depletion in Granulosa Cells Promotes Ovarian Granulosa Cell Tumor Development. Mol Endocrinol 2015; 29:1006-24. [PMID: 26061565 DOI: 10.1210/me.2015-1103] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The forkhead box (FOX), FOXO1 and FOXO3, transcription factors regulate multiple functions in mammalian cells. Selective inactivation of the Foxo1 and Foxo3 genes in murine ovarian granulosa cells severely impairs follicular development and apoptosis causing infertility, and as shown here, granulosa cell tumor (GCT) formation. Coordinate depletion of the tumor suppressor Pten gene in the Foxo1/3 strain enhanced the penetrance and onset of GCT formation. Immunostaining and Western blot analyses confirmed FOXO1 and phosphatase and tensin homolog (PTEN) depletion, maintenance of globin transcription factor (GATA) 4 and nuclear localization of FOXL2 and phosphorylated small mothers against decapentaplegic (SMAD) 2/3 in the tumor cells, recapitulating results we observed in human adult GCTs. Microarray and quantitative PCR analyses of mouse GCTs further confirmed expression of specific genes (Foxl2, Gata4, and Wnt4) controlling granulosa cell fate specification and proliferation, whereas others (Emx2, Nr0b1, Rspo1, and Wt1) were suppressed. Key genes (Amh, Bmp2, and Fshr) controlling follicle growth, apoptosis, and differentiation were also suppressed. Inhbb and Grem1 were selectively elevated, whereas reduction of Inha provided additional evidence that activin signaling and small mothers against decapentaplegic (SMAD) 2/3 phosphorylation impact GCT formation. Unexpectedly, markers of Sertoli/epithelial cells (SRY [sex determining region Y]-box 9/keratin 8) and alternatively activated macrophages (chitinase 3-like 3) were elevated in discrete subpopulations within the mouse GCTs, indicating that Foxo1/3/Pten depletion not only leads to GCTs but also to altered granulosa cell fate decisions and immune responses. Thus, analyses of the Foxo1/3/Pten mouse GCTs and human adult GCTs provide strong evidence that impaired functions of the FOXO1/3/PTEN pathways lead to dramatic changes in the molecular program within granulosa cells, chronic activin signaling in the presence of FOXL2 and GATA4, and tumor formation.
Collapse
Affiliation(s)
- Zhilin Liu
- Departments of Molecular and Cellular Biology (Z.L., Y.A.R., S.A.P., J.A., J.S.R.), Pathology and Immunology (S.A.P.), and Obstetrics and Gynecology (J.A.), Baylor College of Medicine, and Department of Experimental Radiation Oncology (W.Z.), The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030; Department of Pathology (D.H.C.), The University of Texas Southwestern Medical School, Dallas, Texas 75390; and Department of Anatomy and Developmental Biology (D.W.), Monash University, Clayton VIC 3800, Australia
| | - Yi A Ren
- Departments of Molecular and Cellular Biology (Z.L., Y.A.R., S.A.P., J.A., J.S.R.), Pathology and Immunology (S.A.P.), and Obstetrics and Gynecology (J.A.), Baylor College of Medicine, and Department of Experimental Radiation Oncology (W.Z.), The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030; Department of Pathology (D.H.C.), The University of Texas Southwestern Medical School, Dallas, Texas 75390; and Department of Anatomy and Developmental Biology (D.W.), Monash University, Clayton VIC 3800, Australia
| | - Stephanie A Pangas
- Departments of Molecular and Cellular Biology (Z.L., Y.A.R., S.A.P., J.A., J.S.R.), Pathology and Immunology (S.A.P.), and Obstetrics and Gynecology (J.A.), Baylor College of Medicine, and Department of Experimental Radiation Oncology (W.Z.), The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030; Department of Pathology (D.H.C.), The University of Texas Southwestern Medical School, Dallas, Texas 75390; and Department of Anatomy and Developmental Biology (D.W.), Monash University, Clayton VIC 3800, Australia
| | - Jaye Adams
- Departments of Molecular and Cellular Biology (Z.L., Y.A.R., S.A.P., J.A., J.S.R.), Pathology and Immunology (S.A.P.), and Obstetrics and Gynecology (J.A.), Baylor College of Medicine, and Department of Experimental Radiation Oncology (W.Z.), The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030; Department of Pathology (D.H.C.), The University of Texas Southwestern Medical School, Dallas, Texas 75390; and Department of Anatomy and Developmental Biology (D.W.), Monash University, Clayton VIC 3800, Australia
| | - Wei Zhou
- Departments of Molecular and Cellular Biology (Z.L., Y.A.R., S.A.P., J.A., J.S.R.), Pathology and Immunology (S.A.P.), and Obstetrics and Gynecology (J.A.), Baylor College of Medicine, and Department of Experimental Radiation Oncology (W.Z.), The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030; Department of Pathology (D.H.C.), The University of Texas Southwestern Medical School, Dallas, Texas 75390; and Department of Anatomy and Developmental Biology (D.W.), Monash University, Clayton VIC 3800, Australia
| | - Diego H Castrillon
- Departments of Molecular and Cellular Biology (Z.L., Y.A.R., S.A.P., J.A., J.S.R.), Pathology and Immunology (S.A.P.), and Obstetrics and Gynecology (J.A.), Baylor College of Medicine, and Department of Experimental Radiation Oncology (W.Z.), The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030; Department of Pathology (D.H.C.), The University of Texas Southwestern Medical School, Dallas, Texas 75390; and Department of Anatomy and Developmental Biology (D.W.), Monash University, Clayton VIC 3800, Australia
| | - Dagmar Wilhelm
- Departments of Molecular and Cellular Biology (Z.L., Y.A.R., S.A.P., J.A., J.S.R.), Pathology and Immunology (S.A.P.), and Obstetrics and Gynecology (J.A.), Baylor College of Medicine, and Department of Experimental Radiation Oncology (W.Z.), The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030; Department of Pathology (D.H.C.), The University of Texas Southwestern Medical School, Dallas, Texas 75390; and Department of Anatomy and Developmental Biology (D.W.), Monash University, Clayton VIC 3800, Australia
| | - JoAnne S Richards
- Departments of Molecular and Cellular Biology (Z.L., Y.A.R., S.A.P., J.A., J.S.R.), Pathology and Immunology (S.A.P.), and Obstetrics and Gynecology (J.A.), Baylor College of Medicine, and Department of Experimental Radiation Oncology (W.Z.), The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030; Department of Pathology (D.H.C.), The University of Texas Southwestern Medical School, Dallas, Texas 75390; and Department of Anatomy and Developmental Biology (D.W.), Monash University, Clayton VIC 3800, Australia
| |
Collapse
|