1
|
Xu YP, Fu JC, Hong ZL, Zeng DF, Guo CQ, Li P, Wu JX. Psychological stressors involved in the pathogenesis of premature ovarian insufficiency and potential intervention measures. Gynecol Endocrinol 2024; 40:2360085. [PMID: 38813955 DOI: 10.1080/09513590.2024.2360085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 05/14/2024] [Indexed: 05/31/2024] Open
Abstract
Premature ovarian insufficiency (POI) is a common gynecological endocrine disease, which seriously affects women's physical and mental health and fertility, and its incidence is increasing year by year. With the development of social economy and technology, psychological stressors such as anxiety and depression caused by social, life and environmental factors may be one of the risk factors for POI. We used PubMed to search peer-reviewed original English manuscripts published over the last 10 years to identify established and experimental studies on the relationship between various types of stress and decreased ovarian function. Oxidative stress, follicular atresia, and excessive activation of oocytes, caused by Stress-associated factors may be the main causes of ovarian function damage. This article reviews the relationship between psychological stressors and hypoovarian function and the possible early intervention measures in order to provide new ideas for future clinical treatment and intervention.
Collapse
Affiliation(s)
- Ying-Pei Xu
- Department of Reproductive Medicine, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, Fujian Province, China
- Department of Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Ji-Chun Fu
- Department of Reproductive Medicine, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, Fujian Province, China
| | - Zhi-Lin Hong
- Clinical Laboratory Center, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - De-Fei Zeng
- Department of Reproductive Medicine, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, Fujian Province, China
| | - Chao-Qin Guo
- Department of Reproductive Medicine, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, Fujian Province, China
| | - Ping Li
- Department of Reproductive Medicine, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, Fujian Province, China
| | - Jin-Xiang Wu
- Department of Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
- Department of Reproductive Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| |
Collapse
|
2
|
Xu X, Jiang H, Wang D, Rehman SU, Li Z, Song X, Cui K, Luo X, Yang C, Liu Q. Exploration of transcriptional regulation network between buffalo oocytes and granulosa cells and its impact on different diameter follicles. BMC Genomics 2024; 25:1004. [PMID: 39462339 PMCID: PMC11515274 DOI: 10.1186/s12864-024-10912-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 10/16/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND Buffalo is a globally important livestock species, but its reproductive performance is relatively low than cattles. At present, dominant follicle development specific process and mechanistic role of follicular growth related genes in water buffaloes are not well understood. Therefore, we comprehensively performed transcriptomics of granulosa cells and oocytes from different-sized follicles in water buffalo to identify key candidate genes that influence follicle development and diameter, and further explored the potential regulatory mechanisms of granulosa cells and oocytes in the process of water buffalo follicle development. RESULTS In this study, we found918 granulosa cell transcripts and 1401 oocyte transcripts were correlated in follicles of different diameters, and the expression differences were significant. Subsequent enrichment analysis of the co-expressed differentially expressed transcripts identified several genes targeted by long non-coding RNAs (lncRNAs) and associated with follicular development. Notably, the upregulation of BUB1 regulated by MSTRG.41325.4 and interactive action of SMAD2 and SMAD7 might have key regulatory role in follicular development. Additionally, we also detected key differentially expressed genes that potentially influence follicular hormone metabolism and growth, like ID2, CHRD, TGIF2 and MAD2L1, and constructed an interaction network between lncRNA transcripts and mRNAs. CONCLUSIONS In summary, this study preliminarily revealed the differences in gene expression patterns among buffalo follicles of different sizes and their potential molecular regulatory mechanisms. It provides a new perspective for exploring the mechanism of buffalo follicular dominance and improving buffalo reproductive performance.
Collapse
Affiliation(s)
- Xiaoxian Xu
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, 528225, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
| | - Hancai Jiang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
| | - Dong Wang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
| | - Saif Ur Rehman
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
| | - Zhipeng Li
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
| | - Xinhui Song
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
| | - Kuiqing Cui
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, 528225, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
| | - Xier Luo
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, 528225, China
| | - Chunyan Yang
- Buffalo Research Institute, Chinese Academy of Agricultural Sciences, Nanning, 530001, China.
| | - Qingyou Liu
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, 528225, China.
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China.
| |
Collapse
|
3
|
Lee YZ, Cheng SH, Lin YF, Wu CC, Tsai YC. The Beneficial Effects of Lacticaseibacillus paracasei subsp. paracasei DSM 27449 in a Letrozole-Induced Polycystic Ovary Syndrome Rat Model. Int J Mol Sci 2024; 25:8706. [PMID: 39201391 PMCID: PMC11354393 DOI: 10.3390/ijms25168706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 09/02/2024] Open
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent endocrine disorder affecting women of reproductive age globally. Emerging evidence suggests that the dysregulation of microRNAs (miRNAs) and gut dysbiosis are linked to the development of PCOS. In this study, the effects of Lacticaseibacillus paracasei subsp. paracasei DSM 27449 (DSM 27449) were investigated in a rat model of PCOS induced by letrozole. The administration of DSM 27449 resulted in improved ovarian function, reduced cystic follicles, and lower serum testosterone levels. Alterations in miRNA expressions and increased levels of the pro-apoptotic protein Bax in ovarian tissues were observed in PCOS-like rats. Notably, the administration of DSM 27449 restored the expression of miRNAs, including miR-30a-5p, miR-93-5p, and miR-223-3p, leading to enhanced ovarian function through the downregulation of Bax expressions in ovarian tissues. Additionally, 16S rRNA sequencing showed changes in the gut microbiome composition after letrozole induction. The strong correlation between specific bacterial genera and PCOS-related parameters suggested that the modulation of the gut microbiome by DSM 27449 was associated with the improvement of PCOS symptoms. These findings demonstrate the beneficial effects of DSM 27449 in ameliorating PCOS symptoms in letrozole-induced PCOS-like rats, suggesting that DSM 27449 may serve as a beneficial dietary supplement with the therapeutic potential for alleviating PCOS.
Collapse
Affiliation(s)
- Yan Zhang Lee
- Biomedical Industry Ph.D. Program, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan;
| | - Shih-Hsuan Cheng
- Bened Biomedical Co., Ltd., Taipei 115011, Taiwan; (S.-H.C.); (Y.-F.L.); (C.-C.W.)
| | - Yu-Fen Lin
- Bened Biomedical Co., Ltd., Taipei 115011, Taiwan; (S.-H.C.); (Y.-F.L.); (C.-C.W.)
| | - Chien-Chen Wu
- Bened Biomedical Co., Ltd., Taipei 115011, Taiwan; (S.-H.C.); (Y.-F.L.); (C.-C.W.)
| | - Ying-Chieh Tsai
- Biomedical Industry Ph.D. Program, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan;
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| |
Collapse
|
4
|
Tang Y, Du E, Wang G, Qin F, Meng Z, Dai L, Wang Y, Ren S. A negative feedback loop centered on SMAD3 expression in transforming growth factor β1-induced corneal myofibroblast differentiation. Exp Eye Res 2023; 236:109654. [PMID: 37734427 DOI: 10.1016/j.exer.2023.109654] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 09/10/2023] [Accepted: 09/14/2023] [Indexed: 09/23/2023]
Abstract
SMAD3 downregulation is documented in transforming growth factor β1 (TGF-β1)-induced corneal fibroblasts differentiation to myofibroblasts ("fibroTOmyoDiff") or corneal wound healing. However, the exact regulatory mechanism of TGF-β1/SMAD3 pathway in this context remains unclear. Here, we investigated the role and related mechanism of SMAD3 down-regulation in TGF-β1-induced human corneal fibroTOmyoDiff. By detecting expression changes of SMAD family during this process, we demonstrated that SMAD3 protein expression was dramatically decreased in the process and the decrease occurred mainly in SMAD3 gene transcription. Furthermore, SMAD3 overexpression using lentivirus infection and knockdown using sgRNA lentivirus infection or siRNAs revealed that SMAD3 overexpression enhanced TGF-β1-induced corneal fibroTOmyoDiff and vice versa. In addition, specific siRNAs and inhibitors targeting particular signaling pathway were used to figure out the intracellular signaling pathway regulating SMAD3, and the result showed that the decease of SMAD3 induced by TGF-β1 stimulation in human corneal fibroblasts (HCFs) was strikingly prevented by SMAD4 knockdown or p38 signaling inhibitor SB203580 treatment. Collectively, these results demonstrate that, in TGF-β1 induced corneal fibroTOmyoDiff, down-regulation of SMAD3 expression regulated by SMAD4 and p38 signaling pathways forms a negative feedback loop of TGFβ signaling to avoid excessive activation of the signaling, which suggest that SMAD3 may be a key target for corneal fibrosis treatment.
Collapse
Affiliation(s)
- Yunlan Tang
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Enming Du
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Gang Wang
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Fangyuan Qin
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Zhihong Meng
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Lijuan Dai
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Yiqiang Wang
- Wisdom Lake Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, 215123, China.
| | - Shengwei Ren
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China.
| |
Collapse
|
5
|
Bai X, Wang S. Signaling pathway intervention in premature ovarian failure. Front Med (Lausanne) 2022; 9:999440. [PMID: 36507521 PMCID: PMC9733706 DOI: 10.3389/fmed.2022.999440] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 11/14/2022] [Indexed: 11/27/2022] Open
Abstract
Premature ovarian failure (POF) is a multifactorial disease that refers to the occurrence of secondary amenorrhea, estrogen decrease, and gonadotropin increase in women under the age of 40. The prevalence of POF is increasing year by year, and the existing instances can be categorized as primary or secondary cases. This disease has adverse effects on both the physiology and psychology of women. Hormone replacement therapy is the recommended treatment for POF, and a multidisciplinary strategy is required to enhance the quality of life of patients. According to recent studies, the primary mechanism of POF is the depletion of ovarian reserve function as a result of increased primordial follicular activation or primordial follicular insufficiency. Therefore, understanding the processes of primordial follicle activation and associated pathways and exploring effective interventions are important for the treatment of POF.
Collapse
|
6
|
Yang M, Jin C, Cheng X, Liu T, Ji Y, Meng F, Han X, Liang Q, Cao X, Huang L, Du X, Zeng X, Bu G. Corticosterone triggers anti-proliferative and apoptotic effects, and downregulates the ACVR1-SMAD1-ID3 cascade in chicken ovarian prehierarchical, but not preovulatory granulosa cells. Mol Cell Endocrinol 2022; 552:111675. [PMID: 35577112 DOI: 10.1016/j.mce.2022.111675] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/04/2022] [Accepted: 05/08/2022] [Indexed: 10/18/2022]
Abstract
The coordinated proliferation and apoptosis of granulosa cells plays a critical role in follicular development. To identify the exact mechanisms of how stress-driven glucocorticoid production suppresses reproduction, granulosa cells were isolated from chicken follicles at different developmental stages and then treated with corticosterone. Using CCK-8, EDU and TUNEL assays, we showed that corticosterone could trigger both anti-proliferative and pro-apoptotic effects in granulosa cells from 6 to 8 mm follicles only, while depicting no influence on granulosa cells from any preovulatory follicles. High-throughput transcriptomic analysis identified 1362 transcripts showing differential expression profiles in granulosa cells from 6 to 8 mm follicles after corticosterone treatment. Interestingly, Kyoto Encyclopedia of Genes and Genomes enrichment analysis revealed that 17 genes were enriched in the TGF-β signaling pathway, and 13 showed differential expression patterns consistent with corticosterone-induced effects. The differential expression profiles of these 13 genes were examined by quantitative real-time PCR in cultured chicken ovarian granulosa cells at diverse developmental stages following corticosterone challenge for a short (8 h) or long period (24 h). After 24 h of treatment, INHBB, FST, FMOD, NOG, ACVR1, SMAD1 and ID3 were the genes that responded consistently with corticosterone-induced proliferative and apoptotic events in all granulosa cells detected. However, only ACVR1, SMAD1 and ID3 could initiate coincident expression patterns after being treated for 8 h, suggesting their significance in corticosterone-mediated actions. Collectively, these findings indicate that corticosterone can inhibit proliferation and cause apoptosis in chicken ovarian prehierarchical, but not preovulatory granulosa cells, through impeding ACVR1-SMAD1-ID3 signaling presumptively.
Collapse
Affiliation(s)
- Ming Yang
- Isotope Research Laboratory, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China
| | - Chenchen Jin
- Department of Bio-engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China
| | - Xinyue Cheng
- Department of Bio-engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China
| | - Tuoyuan Liu
- Isotope Research Laboratory, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China
| | - Yu Ji
- Department of Bio-engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China
| | - Fengyan Meng
- Isotope Research Laboratory, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China; Department of Bio-engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China
| | - Xingfa Han
- Isotope Research Laboratory, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China; Department of Bio-engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China
| | - Qiuxia Liang
- Isotope Research Laboratory, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China; Department of Bio-engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China
| | - Xiaohan Cao
- Isotope Research Laboratory, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China; Department of Bio-engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China
| | - Linyan Huang
- Isotope Research Laboratory, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China; Department of Bio-engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China
| | - Xiaogang Du
- Isotope Research Laboratory, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China; Department of Bio-engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China
| | - Xianyin Zeng
- Isotope Research Laboratory, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China; Department of Bio-engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China.
| | - Guixian Bu
- Isotope Research Laboratory, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China; Department of Bio-engineering and Applied Biology, College of Life Science, Sichuan Agricultural University, Xinkang Road, Ya'an, 625014, PR China.
| |
Collapse
|
7
|
Widowati W, Prahastuti S, Tjokropranoto R, Onggowidjaja P, Kusuma HSW, Afifah E, Arumwardana S, Maulana MA, Rizal R. Quercetin prevents chronic kidney disease on mesangial cells model by regulating inflammation, oxidative stress, and TGF-β1/SMADs pathway. PeerJ 2022; 10:e13257. [PMID: 35673387 PMCID: PMC9167587 DOI: 10.7717/peerj.13257] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 03/22/2022] [Indexed: 01/13/2023] Open
Abstract
Background Chronic kidney disease (CKD) happens due to decreasing kidney function. Inflammation and oxidative stress have been shown to result in the progression of CKD. Quercetin is widely known to have various bioactivities including antioxidant, anticancer, and anti-inflammatory activities. Objective To evaluate the activity of quercetin to inhibit inflammation, stress oxidative, and fibrosis on CKD cells model (mouse mesangial cells induced by glucose). Methods and Material The SV40 MES 13 cells were plated in a 6-well plate with cell density at 5,000 cells/well. The medium had been substituted for 3 days with a glucose-induced medium with a concentration of 20 mM. Quercetin was added with 50, 10, and 5 µg/mL concentrations. The negative control was the untreated cell. The levels of TGF-β1, TNF-α, and MDA were determined using ELISA KIT. The gene expressions of the SMAD7, SMAD3, SMAD2, and SMAD4 were analyzed using qRT-PCR. Results Glucose can lead to an increase in inflammatory cytokines TNF-α, TGF-β1, MDA as well as the expressions of the SMAD2, SMAD3, SMAD4, and a decrease in SMAD7. Quercetin caused the reduction of TNF-α, TGF-β1, MDA as well as the expression of the SMAD2, SMAD3, SMAD4, and increased SMAD7. Conclusion Quercetin has anti-inflammation, antioxidant, antifibrosis activity in the CKD cells model. Thus, quercetin is a promising substance for CKD therapy and further research is needed to prove this in CKD animal model.
Collapse
Affiliation(s)
- Wahyu Widowati
- Faculty of Medicine, Maranatha Christian University, Bandung, West Java, Indonesia
| | - Sijani Prahastuti
- Faculty of Medicine, Maranatha Christian University, Bandung, West Java, Indonesia
| | - Rita Tjokropranoto
- Faculty of Medicine, Maranatha Christian University, Bandung, West Java, Indonesia
| | - Philips Onggowidjaja
- Faculty of Medicine, Maranatha Christian University, Bandung, West Java, Indonesia
| | - Hanna Sari Widya Kusuma
- Biomolecular and Biomedical Research Center, Aretha Medika Utama, Bandung, West Java, Indonesia
| | - Ervi Afifah
- Biomolecular and Biomedical Research Center, Aretha Medika Utama, Bandung, West Java, Indonesia
| | - Seila Arumwardana
- Biomolecular and Biomedical Research Center, Aretha Medika Utama, Bandung, West Java, Indonesia
| | - Muhamad Aldi Maulana
- Biomolecular and Biomedical Research Center, Aretha Medika Utama, Bandung, West Java, Indonesia
| | - Rizal Rizal
- Biomolecular and Biomedical Research Center, Aretha Medika Utama, Bandung, West Java, Indonesia,Biomedical Engineering, Department of Electrical Engineering, Faculty of Engineering, University of Indonesia, Depok, West Java, Indonesia
| |
Collapse
|
8
|
Wu L, Fang Q, Wang M, Wang Y, Zhu X, Fang Z, Lu F, Xu B, Jin R, Han H, Tong X. Effect of weight loss on pregnancy outcomes, neuronal-reproductive-metabolic hormones and gene expression profiles in granulosa cells in obese infertile PCOS patients undergoing IVF-ET. Front Endocrinol (Lausanne) 2022; 13:954428. [PMID: 36246893 PMCID: PMC9562768 DOI: 10.3389/fendo.2022.954428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE To investigate the effect of weight loss on pregnancy outcomes, PCOS related neuronal-reproductive-metabolic hormones and ovarian granulosa cell gene expression profiles in obese PCOS infertile patients undergoing in vitro fertilization-embryo transfer (IVF-ET). METHODS 75 patients undergoing IVF-ET due to tubal factors alone collected as the control group (group A), and 352 patients with obese PCOS infertility were divided into four groups according to the amount of weight loss before IVF: 0 kg (group B), 1-5 kg (group C), 5-10 kg (group D), and >10 kg (group E). Six cases of ovarian granulosa cells were collected randomly with the random number table method in each group for detecting mRNA profiling. Pathway networks and biological functions of the differentially expressed genes were analyzed. Validation by RT-PCR was performed. RESULTS (1) The levels of luteinizing hormone(LH), testosterone(T) and homeostasis model assessment insulin resistance(HOMA-IR) in group E were significantly lower than those in groups B and C (P<0.05). (2) Compared with groups A and E, groups B and C showed increased total gonadotropin (Gn) and days of Gn stimulation (P<0.05), and the E2 level on trigger day and number of oocytes obtained in group B was significantly less than that in group E (P<0.05 or 0.01). Embryo implantation rate, clinical pregnancy rate and live birth rate were increased and miscarriage rate was decreased in groups A, D and E compared with group B (P<0.05 or 0.01). (3) There were significant differences among the control group and PCOS groups in some genes that are involved in neuronal-reproductive-metabolic endocrine, transcriptional regulation, cell proliferation and differentiation, etc (P<0.05). RNA-Seq results were validated by real time PCR analysis for the expression of follicle stimulating hormone receptor (FSHR), drosophila mothers against decapentaplegic protein 7(Smad7) and glutathione peroxidase 3(GPX3) genes that are known to have an important role in follicular development. Functional alterations were confirmed by the improvement in the ovarian responsiveness to Gn and embryo quality. CONCLUSION Weight loss more than 5kg may regulate the neuroreproductive endocrine hormone secretion, insulin resistance and gene expression profiles of ovarian granulosa cells, so as to improve the ovarian responsiveness to Gn, the embryo quality, embryo implantation rate, clinical pregnancy rate, live birth rate, and reduce the spontaneous abortion rate in obese infertile PCOS patients undergoing IVF-ET. CLINICAL TRIAL REGISTRATION www.chictr.org.cn, identifier ChiCTR1800018298.
Collapse
Affiliation(s)
- Limin Wu
- Reproductive and genetic branch, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Qunying Fang
- Reproductive and genetic branch, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Mengli Wang
- Reproductive and genetic branch, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Graduate school, Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Yurui Wang
- Reproductive and genetic branch, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xinyi Zhu
- Reproductive and genetic branch, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhaohui Fang
- Endocrine Department, The First Affiliated Hospital, Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Fangting Lu
- Reproductive and genetic branch, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Bo Xu
- Reproductive and genetic branch, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Rentao Jin
- Reproductive and genetic branch, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- *Correspondence: Xianhong Tong, ; Hui Han, ; Rentao Jin,
| | - Hui Han
- Neurology Department, The First Affiliated Hospital, Anhui University of Traditional Chinese Medicine, Hefei, China
- *Correspondence: Xianhong Tong, ; Hui Han, ; Rentao Jin,
| | - Xianhong Tong
- Reproductive and genetic branch, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- *Correspondence: Xianhong Tong, ; Hui Han, ; Rentao Jin,
| |
Collapse
|
9
|
Amoushahi M, Lykke-Hartmann K. Distinct Signaling Pathways Distinguish in vivo From in vitro Growth in Murine Ovarian Follicle Activation and Maturation. Front Cell Dev Biol 2021; 9:708076. [PMID: 34368158 PMCID: PMC8346253 DOI: 10.3389/fcell.2021.708076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 06/18/2021] [Indexed: 11/24/2022] Open
Abstract
Women with cancer and low ovarian reserves face serious challenges in infertility treatment. Ovarian tissue cryopreservation is currently used for such patients to preserve fertility. One major challenge is the activation of dormant ovarian follicles, which is hampered by our limited biological understanding of molecular determinants that activate dormant follicles and help maintain healthy follicles during growth. Here, we investigated the transcriptomes of oocytes isolated from dormant (primordial) and activated (primary) follicles under in vivo and in vitro conditions. We compared the biological relevance of the initial molecular markers of mature metaphase II (MII) oocytes developed in vivo or in vitro. The expression levels of genes involved in the cell cycle, signal transduction, and Wnt signaling were highly enriched in oocytes from primary follicles and MII oocytes. Interestingly, we detected strong downregulation of the expression of genes involved in mitochondrial and reactive oxygen species (ROS) production in oocytes from primordial follicles, in contrast to oocytes from primary follicles and MII oocytes. Our results showed a dynamic pattern in mitochondrial and ROS production-related genes, emphasizing their important role(s) in primordial follicle activation and oocyte maturation. The transcriptome of MII oocytes showed a major divergence from that of oocytes of primordial and primary follicles.
Collapse
Affiliation(s)
| | - Karin Lykke-Hartmann
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Clinical Genetics, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
10
|
Labazi H, Nilsen M, MacLean MR. Sex-dependent right ventricular hypertrophic gene changes after methamphetamine treatment in mice. Eur J Pharmacol 2021; 900:174066. [PMID: 33789156 PMCID: PMC8111419 DOI: 10.1016/j.ejphar.2021.174066] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 03/17/2021] [Accepted: 03/22/2021] [Indexed: 11/19/2022]
Abstract
Methamphetamine (MA) abuse is associated with the development of pulmonary arterial hypertension (PAH) and subsequent right ventricular failure. A recent clinical study demonstrated that female sex is a major risk factor for MA-induced PAH. The mechanisms associated with increased prevalence and severity of MA-induced PAH in females are still unclear. We hypothesized that MA may promote changes in gene expression in the right ventricle contributing to the development and/or worsening of PAH in females. Male and female C57BL/6 mice were treated with either MA or vehicle. Right and left ventricular systolic pressures (RVSP and LVSP, respectively) were assessed and tissue samples were collected for gene expression and histology. LVSP and RVSP were not affected by MA in either males or females. Right ventricular hypertrophy was significantly increased by MA in females but it was not affected by MA in males. In the female mice, MA-induced right ventricular hypertrophy was associated with increased expression of brain natriuretic peptide gene and members of the TGF-β receptor signaling pathway such as TGF-β receptor-1, smad3 and smad7. In male mice, there were no changes in right ventricular gene expression. Our results suggest that MA caused right ventricular hypertrophy in female mice, but not in males and that this was associated with an increase in hypertrophic genes. The right ventricular hypertrophy was not dependent on increased RVSP suggesting a direct effect of MA on the right ventricle. If this translates to PAH patients, it might explain the poor outcome observed in MA-associated female PAH patients.
Collapse
Affiliation(s)
- Hicham Labazi
- Institute of Cardiovascular & Medical Sciences and College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK; Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK.
| | - Margaret Nilsen
- Institute of Cardiovascular & Medical Sciences and College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK; Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Margaret R MacLean
- Institute of Cardiovascular & Medical Sciences and College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK; Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| |
Collapse
|
11
|
Enhanced Intestinal TGF-β/SMAD-Dependent Signaling in Simian Immunodeficiency Virus Infected Rhesus Macaques. Cells 2021; 10:cells10040806. [PMID: 33916615 PMCID: PMC8066988 DOI: 10.3390/cells10040806] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/18/2021] [Accepted: 04/01/2021] [Indexed: 12/12/2022] Open
Abstract
Transforming growth factor-β signaling (TGF-β) maintains a balanced physiological function including cell growth, differentiation, and proliferation and regulation of immune system by modulating either SMAD2/3 and SMAD7 (SMAD-dependent) or SMAD-independent signaling pathways under normal conditions. Increased production of TGF-β promotes immunosuppression in Human Immunodeficiency Virus (HIV)/Simian Immunodeficiency Virus (SIV) infection. However, the cellular source and downstream events of increased TGF-β production that attributes to its pathological manifestations remain unknown. Here, we have shown increased production of TGF-β in a majority of intestinal CD3−CD20−CD68+ cells from acute and chronically SIV infected rhesus macaques, which negatively correlated with the frequency of jejunum CD4+ T cells. No significant changes in intestinal TGF-β receptor II expression were observed but increased production of the pSMAD2/3 protein and SMAD3 gene expression in jejunum tissues that were accompanied by a downregulation of SMAD7 protein and gene expression. Enhanced TGF-β production by intestinal CD3−CD20−CD68+ cells and increased TGF-β/SMAD-dependent signaling might be due to a disruption of a negative feedback loop mediated by SMAD7. This suggests that SIV infection impacts the SMAD-dependent signaling pathway of TGF-β and provides a potential framework for further study to understand the role of viral factor(s) in modulating TGF-β production and downregulating SMAD7 expression in SIV. Regulation of mucosal TGF-β expression by therapeutic TGF-β blockers may help to create effective antiviral mucosal immune responses.
Collapse
|
12
|
BustosRivera-Bahena G, López-Guerrero DV, Márquez-Bandala AH, Esquivel-Guadarrama FR, Montiel-Hernández JL. TGF-β1 signaling inhibit the in vitro apoptotic, infection and stimulatory cell response induced by influenza H1N1 virus infection on A549 cells. Virus Res 2021; 297:198337. [PMID: 33581185 DOI: 10.1016/j.virusres.2021.198337] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 01/18/2021] [Accepted: 02/02/2021] [Indexed: 12/12/2022]
Abstract
Influenza A virus (IAV) infection induces host cell responses that could derive in inflammatory and apoptotic response. In this respect, in multiple pathological situations, TGF-β1 has shown anti-inflammatory effect, but its role during IAV infection is poorly understood. Interestingly, recent profiling expression studies have suggested that the TGF-β1 pathway could be functionally related to the IAV infection's host response. To gain an understanding of the involvement of TGF-β1's signaling pathway during IAV infection, we compared different apoptotic proteins such as TNFR1, Fas ligand, XIAP, cIAP, among others proteins, and pro-inflammatory elements like IL-1β in the A549 cells during IAV infection (H1N1/NC/99), with and without 1 h of pre-treatment with TGF-β1. Pre-incubation with TGF-β1 significantly inhibited apoptosis and the presence of pro-apoptotic factors. Moreover, the relative abundance of immunodetected IAV M1 protein along 24 -h post-infection period was abridged, which correlated with a disminished infectious viral progeny Additionally, caspase 1 activation and increase of IL-1β induced by IAV infection was also reduced by TGF-β1 signaling activation. Whereas IAV infection increase of Smad-7 and, as consequence, partially inhibiting Smad2/3 phosphorylation, pre-treatment with TGF-β1 blocked IAV-dependent Smad7 induction and prevented Smad2/3 signaling shutdown. All these data suggest the role of TGF-β1 signaling pathway in the control of host cell response induced by the IAV infection and identify a potential clinical target to modulate acute cell death.
Collapse
Affiliation(s)
- Genoveva BustosRivera-Bahena
- Instituto de Biotecnología, UNAM, Cuernavaca, México; Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca, México
| | - Delia Vanessa López-Guerrero
- Facultad de Medicina, Universidad Autónoma del Estado de Morelos, Cuernavaca, México; Facultad de Nutrición, Universidad Autónoma del Estado de Morelos, Cuernavaca, México
| | - Alicia Helena Márquez-Bandala
- Instituto de Biotecnología, UNAM, Cuernavaca, México; Facultad de Medicina, Universidad Autónoma del Estado de Morelos, Cuernavaca, México
| | | | | |
Collapse
|
13
|
Abstract
Advanced maternal age is associated with the natural oocyte depletion, leading to low oocyte yield, high infertility treatment cancellation rates, and eventual decreases in pregnancy rates. Various innovative interventions have been introduced to improve the outcome of infertility treatment for aging patients. Numerous published data demonstrated that early follicle development was regulated by intraovarian growth factors through autocrine or paracrine mechanisms. Platelet-rich plasma (PRP), a plasma fraction of peripheral blood with a high concentration of platelets, has been implemented in regenerative medicine in the last decade. The plasma contains a variety of growth factors that were suggested to be able to enhance angiogenesis regeneration and the cell proliferation process. The initial report showed that an intraovarian injection of PRP improved the hormonal profile and increased the number of retrieved oocytes in patients with diminished ovarian reserve. Subsequently, several studies with larger sample sizes have reported that this approach resulted in several healthy live births with no apparent complications. However, the use of ovarian PRP treatment needs to be fully investigated, because no randomized controlled trial has yet been performed to confirm its efficacy.
Collapse
|
14
|
Transcriptome Analysis of Porcine Granulosa Cells in Healthy and Atretic Follicles: Role of Steroidogenesis and Oxidative Stress. Antioxidants (Basel) 2020; 10:antiox10010022. [PMID: 33379347 PMCID: PMC7824097 DOI: 10.3390/antiox10010022] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/20/2020] [Accepted: 12/22/2020] [Indexed: 12/14/2022] Open
Abstract
One of the main causes of female infertility is a deregulated antral follicular atresia, a process of which the underlying molecular mechanisms are largely unknown. Our objective was therefore to characterize the complex transcriptome changes in porcine granulosa cells of healthy antral (HA) and advanced antral atretic (AA) follicles, using ELISA and RNA-Seq followed by qRT-PCR and immunohistochemistry. Granulosa cell RNA-Seq data revealed 2160 differentially expressed genes, 1483 with higher and 677 with lower mRNA concentrations in AA follicles. Bioinformatic analysis showed that the upregulated genes in AA follicles were highly enriched in inflammation and apoptosis processes, while the downregulated transcripts were mainly highlighted in the steroid biosynthesis pathway and response to oxidative stress processes including antioxidant genes (e.g., GSTA1, GCLC, GCLM, IDH1, GPX8) involved in the glutathione metabolism pathway and other redox-related genes (e.g., RRM2B, NDUFS4). These observations were confirmed by RT-qPCR and immunohistochemistry. Additionally, the granulosa cells of AA follicles express significantly stronger 8-OHdG immunostaining, a marker of oxidative DNA damage, implicating that oxidative stress may participate in follicular atresia. We hypothesize that the decrease in anti-apoptotic factors and steroid hormones coincides with increased oxidative stress markers and the expression of pro-apoptotic factors, all contributing to antral follicular atresia.
Collapse
|
15
|
Mbatha B, Khathi A, Sibiya N, Booysen I, Mangundu P, Ngubane P. Cardio-protective effects of a dioxidovanadium(V) complex in male sprague-dawley rats with streptozotocin-induced diabetes. Biometals 2020; 34:161-173. [PMID: 33206308 DOI: 10.1007/s10534-020-00270-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 11/09/2020] [Indexed: 11/24/2022]
Abstract
Cardiovascular complications are among the leading causes of morbidity and mortality in diabetes mellitus (DM). Despite the anti-hyperglycemic effects of various anti-diabetic therapeutic agents like insulin, some of these drugs are implicated in precipitating cardiovascular dysfunction. There is therefore an imperative need to seek alternative drugs that may ameliorate these complications. Accordingly, the aim of the study was to investigate the effects of a dioxidovanadium (V) complex, cis-[VO2(obz)py]) on selected cardiovascular function markers in STZ-induced diabetic rats. The vanadium complex (40 mg kg) was administered orally twice every 3rd day 5 weeks, non-diabetic and diabetic control groups received distilled water whereas the insulin group received subcutaneous insulin injections twice daily for 5 weeks. Blood glucose concentrations, mean arterial pressure (MAP), heart rate, triglycerides (TG) and total cholesterol concentrations were monitored weekly for 5 weeks. Rats were then euthanised and blood and hearts were collected for biochemical analysis. There was a significant decrease in blood glucose, triglycerides, cholesterol concentrations as well as blood pressure of vanadium treated rats compared to the untreated diabetic animals. Vanadium treatment also attenuated cardiac oxidative stress and decreased the expression of transforming growth factor β1 (TGFβ1) and Smad7. Lastly, the administration of the vanadium complex significantly decreased C reactive protein (CRP) and cardiotropin 1(CT-1) concentrations in the plasma and heart tissues. The administration of the dioxidovanadium(V) complex to diabetic rats culminated into cardio-protective effects. Taken together, these observations suggest that this metal complex exhibit a significant potential as an alternative therapeutic drug for DM management.
Collapse
Affiliation(s)
- Bonisiwe Mbatha
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu Natal, Durban, South Africa. .,Department of Human Physiology, University of KwaZulu Natal, E-Block, Level 4, Room E4-402, University Road, Chiltern Hills, Westville Campus, 3629, Westville, Private Bag X54001, Durban, 4000, South Africa.
| | - Andile Khathi
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu Natal, Durban, South Africa
| | - Ntethelelo Sibiya
- Pharmacology Division, Faculty of Pharmacy, Rhodes University, Grahamstown, South Africa
| | - Irvin Booysen
- School of Chemistry and Physics, College of Agricultural and Environmental Sciences, University of KwaZulu-Natal, Pietermaritzburg, South Africa
| | - Patrick Mangundu
- School of Chemistry and Physics, College of Agricultural and Environmental Sciences, University of KwaZulu-Natal, Pietermaritzburg, South Africa
| | - Phikelelani Ngubane
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu Natal, Durban, South Africa
| |
Collapse
|
16
|
Zhang X, Chen Y, Yang M, Shang J, Xu Y, Zhang L, Wu X, Ding Y, Liu Y, Chu M, Yin Z. MiR-21-5p actions at the Smad7 gene during pig ovarian granulosa cell apoptosis. Anim Reprod Sci 2020; 223:106645. [PMID: 33217624 DOI: 10.1016/j.anireprosci.2020.106645] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 12/26/2022]
Abstract
MicroRNAs (miRNAs) are endogenous non-coding RNAs in eukaryotic cells that modulate apoptosis of ovarian granulosa cells (GCs), which is an important cause of mammalian follicular atresia. In the present study, associations were evaluated between miR-21-5p and the extent of Smad7 protein production in regulation of ovarian granulosa cell (pGC) apoptosis. There was detection of miR-21-5p and Smad7 primarily in the cytoplasm and nucleus of pGCs, respectively. When there was an enhanced abundance of miR-21-5p and decreased abundance of Smad7 there were similar effects in pGCs, including inducing proliferation, inhibiting apoptosis, increasing the number of cells in S and G2/M phases, increasing serum estradiol, and decreasing serum progesterone concentrations. Furthermore, the Smad7 mRNA transcript was identified as a target for miR-21-5p actions, with enhanced abundances of miR-21-5p being associated with a lesser abundance of Smad7 mRNA transcript and protein in pGCs. Overall, results from the present study indicate that miR-21-5p has actions on the Smad7 mRNA transcript during the process of ovarian granulosa cell apoptosis in pigs.
Collapse
Affiliation(s)
- Xiaodong Zhang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, PR China; Key Laboratory of Local Animal Genetic Resources Conservation and Bio-Breeding of Anhui Province, Hefei, 230036, PR China
| | - Yige Chen
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, PR China; Key Laboratory of Local Animal Genetic Resources Conservation and Bio-Breeding of Anhui Province, Hefei, 230036, PR China
| | - Min Yang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, PR China; Key Laboratory of Local Animal Genetic Resources Conservation and Bio-Breeding of Anhui Province, Hefei, 230036, PR China
| | - Jinnan Shang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, PR China; Key Laboratory of Local Animal Genetic Resources Conservation and Bio-Breeding of Anhui Province, Hefei, 230036, PR China
| | - Yiliang Xu
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, PR China; Key Laboratory of Local Animal Genetic Resources Conservation and Bio-Breeding of Anhui Province, Hefei, 230036, PR China
| | - Liang Zhang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, PR China; Key Laboratory of Local Animal Genetic Resources Conservation and Bio-Breeding of Anhui Province, Hefei, 230036, PR China
| | - Xudong Wu
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, PR China; Key Laboratory of Local Animal Genetic Resources Conservation and Bio-Breeding of Anhui Province, Hefei, 230036, PR China
| | - Yueyun Ding
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, PR China; Key Laboratory of Local Animal Genetic Resources Conservation and Bio-Breeding of Anhui Province, Hefei, 230036, PR China
| | - Yuanyuan Liu
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230031, PR China
| | - Mingxing Chu
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, PR China
| | - Zongjun Yin
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, PR China; Key Laboratory of Local Animal Genetic Resources Conservation and Bio-Breeding of Anhui Province, Hefei, 230036, PR China.
| |
Collapse
|
17
|
Ma M, Zhang J, Gao X, Yao W, Li Q, Pan Z. miR-361-5p Mediates SMAD4 to Promote Porcine Granulosa Cell Apoptosis through VEGFA. Biomolecules 2020; 10:biom10091281. [PMID: 32899767 PMCID: PMC7563248 DOI: 10.3390/biom10091281] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/01/2020] [Accepted: 09/03/2020] [Indexed: 12/16/2022] Open
Abstract
Follicular atresia is an inevitable degenerative process that occurs in mammalian ovarian follicles. The molecular events involved in atresia, particularly granulosa cell apoptosis, have long attracted researchers’ attention. Vascular endothelial growth factor A (VEGFA) is downregulated during follicular atresia in porcine ovaries and serves as an inhibitor of apoptosis in granulosa cells. In addition, transforming growth factor (TGF)-βsignaling has been considered a central trigger in granulosa cell apoptosis. However, the link between TGF-β signaling and VEGFA is unknown. We proved that miR-361-5p is significantly upregulated during the atresia process and that it promotes GC apoptosis by directly targeting the VEGFA 3′UTR. In addition, we revealed that the miR-361-5p coding gene MIR361 was significantly downregulated by SMAD4, the central intracellular mediator of TGF-β signaling, that bound to the MIR361 promoter. In conclusion, our findings expanded what is known about VEGFA posttranscriptional regulation and revealed a complete SMAD4/miR-361-5p/VEGFA regulatory network in ovarian granulosa cell apoptosis. These data provide useful references for follicular atresia and ovarian physiological function studies.
Collapse
|
18
|
Haddad YH, Said RS, Kamel R, Morsy EME, El-Demerdash E. Phytoestrogen genistein hinders ovarian oxidative damage and apoptotic cell death-induced by ionizing radiation: co-operative role of ER-β, TGF-β, and FOXL-2. Sci Rep 2020; 10:13551. [PMID: 32782329 PMCID: PMC7419553 DOI: 10.1038/s41598-020-70309-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 07/14/2020] [Indexed: 01/17/2023] Open
Abstract
Radiotherapy is a well-known cause of premature ovarian failure (POF). Therefore, we investigated the molecular influence of genistein (GEN) on the ovarian reserve of rats exposed to ϒ-radiation. Female Sprague Dawley rats were exposed to a 3.2 Gy γ-radiation to induce POF and/or treated with either GEN (5 mg/kg, i.p.) or Ethinyl estradiol (E2; 0.1 mg/kg, s.c.), once daily for 10 days. GEN was able to conserve primordial follicles stock and population of growing follicles accompanied with reduction in atretic follicles. GEN restored the circulating estradiol and anti-Müllerian hormone levels which were diminished after irradiation. GEN has potent antioxidant activity against radiation-mediated oxidative stress through upregulating endogenous glutathione levels and glutathione peroxidase activity. Mechanistically, GEN inhibited the intrinsic pathway of apoptosis by repressing Bax expression and augmenting Bcl-2 expression resulted in reduced Bax/Bcl-2 ratio with subsequent reduction in cytochrome c and caspase 3 expression. These promising effects of GEN are associated with improving granulosa cells proliferation. On the molecular basis, GEN reversed ovarian apoptosis through up-regulation of ER-β and FOXL-2 with downregulation of TGF-β expression, therefore inhibiting transition of primordial follicles to more growing follicles. GEN may constitute a novel therapeutic modality for safeguarding ovarian function of females' cancer survivors.
Collapse
Affiliation(s)
| | - Riham S Said
- Department of Drug Radiation Research, National Center for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| | - Rehab Kamel
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Engy M El Morsy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Ebtehal El-Demerdash
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
19
|
Gandhi CR. Pro- and Anti-fibrogenic Functions of Gram-Negative Bacterial Lipopolysaccharide in the Liver. Front Med (Lausanne) 2020; 7:130. [PMID: 32373617 PMCID: PMC7186417 DOI: 10.3389/fmed.2020.00130] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 03/24/2020] [Indexed: 12/14/2022] Open
Abstract
Extensive research performed over several decades has identified cells participating in the initiation and progression of fibrosis, and the numerous underlying inter- and intra-cellular signaling pathways. However, liver fibrosis continues to be a major clinical challenge as the precise targets of treatment are still elusive. Activation of physiologically quiescent perisinusoidal hepatic stellate cells (HSCs) to a myofibroblastic proliferating, contractile and fibrogenic phenotype is a critical event in the pathogenesis of chronic liver disease. Thus, elucidation of the mechanisms of the reversal to quiescence or inhibition of activated HSCs, and/or their elimination via apoptosis has been the focus of intense investigation. Lipopolysaccharide (LPS), a gut-resident Gram-negative bacterial endotoxin, is a powerful pro-inflammatory molecule implicated in hepatic injury, inflammation and fibrosis. In both acute and chronic liver injury, portal venous levels of LPS are elevated due to increased intestinal permeability. LPS, via CD14 and Toll-like receptor 4 (TLR4) and its adapter molecules, stimulates macrophages, neutrophils and several other cell types to produce inflammatory mediators as well as factors that can activate HSCs and stimulate their fibrogenic activity. LPS also stimulates synthesis of pro- and anti-inflammatory cytokines/chemokines, growth mediators and molecules of immune regulation by HSCs. However, LPS was found to arrest proliferation of activated HSCs and to convert them into non-fibrogenic phenotype. Interestingly, LPS can elicit responses in HSCs independent of CD14 and TLR4. Identifying and/or developing non-inflammatory but anti-fibrogenic mimetics of LPS could be relevant for treating liver fibrosis.
Collapse
Affiliation(s)
- Chandrashekhar R Gandhi
- Divisions of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Cincinnati VA Medical Center, Cincinnati, OH, United States
| |
Collapse
|
20
|
Chu YL, Xu YR, Yang WX, Sun Y. The role of FSH and TGF-β superfamily in follicle atresia. Aging (Albany NY) 2019; 10:305-321. [PMID: 29500332 PMCID: PMC5892684 DOI: 10.18632/aging.101391] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 02/23/2018] [Indexed: 01/02/2023]
Abstract
Most of the mammalian follicles undergo a degenerative process called “follicle atresia”. Apoptosis of granulosa cells is the main characteristic of follicle atresia. Follicle stimulating hormone (FSH) and the transforming growth factor β (TGF-β) superfamily have important regulatory functions in this process. FSH activates protein kinase A and cooperating with insulin receptor substrates, it promotes the PI3K/Akt pathway which weakens apoptosis. Both Smad or non-Smad signaling of the transforming growth factor β superfamily seem to be related to follicle atresia, and the effect of several important family members on follicle atresia is concluded in this article. FSH and TGF-β are likely to mutually influence each other and what we have already known about the possible underlying molecular mechanism is also discussed below.
Collapse
Affiliation(s)
- Yu-Lan Chu
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ya-Ru Xu
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wan-Xi Yang
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yi Sun
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
21
|
Yao W, Pan Z, Du X, Zhang J, Li Q. miR-181b-induced SMAD7 downregulation controls granulosa cell apoptosis through TGF-β signaling by interacting with the TGFBR1 promoter. J Cell Physiol 2018; 233:6807-6821. [PMID: 29319157 DOI: 10.1002/jcp.26431] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 01/05/2018] [Indexed: 12/25/2022]
Abstract
SMAD7 disrupts the TGF-β signaling pathway by influencing TGFBR1 stability and by blocking the binding of TGFBR1 to SMAD2/3. In this study, we showed that SMAD7 attenuated the TGF-β signaling pathway in ovarian granulosa cells (GCs) by regulating TGFBR1 transcriptional activity. To function as a transcription factor, SMAD7 downregulated the mRNA levels of TGFBR1 via direct binding to the SMAD-binding elements (SBEs) within the promoter region of pig TGFBR1. We also showed that SMAD7 enhanced porcine GC apoptosis by interrupting TGFBR1 and the TGF-β signaling pathway. Interestingly, miR-181b, a microRNA that is downregulated during porcine follicular atresia, was identified to be directly targeting SMAD7 at its 3'-UTR. By inhibiting SMAD7, miR-181b could inhibit GC apoptosis by activating the TGF-β signaling pathway. Our findings provide new insights into the mechanisms underlying the regulation of the TGF-β signaling pathway by SMAD7 and miR-181b.
Collapse
Affiliation(s)
- Wang Yao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Zengxiang Pan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Xing Du
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Jinbi Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Qifa Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
22
|
Differential Expression of miR-93 and miR-21 in Granulosa Cells and Follicular Fluid of Polycystic Ovary Syndrome Associating with Different Phenotypes. Sci Rep 2017; 7:14671. [PMID: 29116087 PMCID: PMC5676684 DOI: 10.1038/s41598-017-13250-1] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 08/31/2017] [Indexed: 12/14/2022] Open
Abstract
The heterogeneous and multifactorial essence of polycystic ovary syndrome (PCOS) renders a remarkable significance to microRNAs (miRNAs). Normo-androgenic (NA) and hyperandrogenic (HA) PCOS patients were compared with matched healthy women. Expression of miRNAs and TGFβ signaling genes was studied by qRT-PCR and western blotting. Effect of androgen on expression of miR-93 and miR-21 and involvement of androgen receptor were appraised. In granulosa cells (GCs), miR-93 and miR-21 showed significantly increased levels in HA patients compared to NA patients. On the contrary, follicular fluid (FF) levels of both miRNAs were significantly decreased in HA group compared to control women. No significant change in the expression of miRNAs in serum samples was detected. Furthermore, mRNA levels of SMAD7 and TGFBR2 were significantly downregulated in GCs of HA group compared to NA and control subjects. TGFBR2 protein level was significantly decreased in HA patients compared to controls. Free testosterone and free androgen index were positively correlated with expression of miR-93 and miR-21 in GCs of PCOS group. Our findings show distinct molecular signature of different subtypes of PCOS. Intermediary position of miRNAs as androgen responsive factors may play critical role in the pathogenesis of PCOS in hyperandrogenic condition.
Collapse
|
23
|
Zhang L, Gao J, Cui S. miR-21 is involved in norepinephrine-mediated rat granulosa cell apoptosis by targeting SMAD7. J Mol Endocrinol 2017; 58:199-210. [PMID: 28473352 DOI: 10.1530/jme-16-0248] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 03/29/2017] [Indexed: 12/12/2022]
Abstract
Substantive evidence has indicated that the sympathetic innervation contributes to the regulation and development of ovarian functions. Norepinephrine (NE) is one of the major neurotransmitters contained in the extrinsic ovarian sympathetic nerves and is thought to be a potent moderator of ovarian functions such as steroidogenesis and granulosa cell proliferation or apoptosis. However, the mechanisms of NE regulation of granulosa cell apoptosis in the rat ovary are rare. Real-time PCR and Western blot results show that NE regulates the expression of miR-21 in primary granulosa cells in a dose-dependent manner. Additionally, we found that miR-21 is involved in NE-mediated rat granulosa cells apoptosis and blocks granulosa cell apoptosis by targeting Smad7, a transforming growth factor-beta-inducible mediator of apoptosis in granulosa cells. In primary granulosa cells, a combined treatment of NE and TGF-β increased apoptosis and decreased miR-21 expression, but increased SMAD7 protein levels. We also demonstrated that NE regulates miR-21 by coupling to α1A-adrenergic receptor (α1A-AR). This is the first demonstration that NE controls the reproductive functions by modulating the expression of miR-21 and promoting TGF-β-induced granulosa cell apoptosis. Such NE-mediated effects could be potentially used for regulating the reproductive processes and for treating reproductive disorders.
Collapse
Affiliation(s)
| | | | - Sheng Cui
- State Key Laboratory of AgrobiotechnologyCollege of Biological Sciences, China Agricultural University, Beijing, China
| |
Collapse
|
24
|
Sharum IB, Granados-Aparici S, Warrander FC, Tournant FP, Fenwick MA. Serine threonine kinase receptor associated protein regulates early follicle development in the mouse ovary. Reproduction 2017; 153:221-231. [DOI: 10.1530/rep-16-0612] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 11/14/2016] [Accepted: 11/22/2016] [Indexed: 12/13/2022]
Abstract
The molecular mechanisms involved in regulating the development of small, gonadotrophin-independent follicles are poorly understood; however, many studies have highlighted an essential role for TGFB ligands. Canonical TGFB signalling is dependent upon intracellular SMAD proteins that regulate transcription. STRAP has been identified in other tissues as an inhibitor of the TGFB–SMAD signalling pathway. Therefore, in this study we aimed to determine the expression and role of STRAP in the context of early follicle development. Using qPCR, Strap, Smad3 and Smad7 revealed similar expression profiles in immature ovaries from mice aged 4–16 days containing different populations of early growing follicles. STRAP and SMAD2/3 proteins co-localised in granulosa cells of small follicles using immunofluorescence. Using an established culture model, neonatal mouse ovary fragments with a high density of small non-growing follicles were used to examine the effects of Strap knockdown using siRNA and STRAP protein inhibition by immuno-neutralisation. Both interventions caused a reduction in the proportion of small, non-growing follicles and an increase in the proportion and size of growing follicles in comparison to untreated controls, suggesting inhibition of STRAP facilitates follicle activation. Recombinant STRAP protein had no effect on small, non-growing follicles, but increased the mean oocyte size of growing follicles in the neonatal ovary model and also promoted the growth of isolated preantral follicles in vitro. Overall findings indicate STRAP is expressed in the mouse ovary and is capable of regulating development of small follicles in a stage-dependent manner.
Collapse
|
25
|
Li L, Zhou X, Wang X, Wang J, Zhang W, Wang B, Cao Y, Kee K. A dominant negative mutation at the ATP binding domain ofAMHR2is associated with a defective anti-Müllerian hormone signaling pathway. Mol Hum Reprod 2016; 22:669-78. [DOI: 10.1093/molehr/gaw040] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 06/07/2016] [Indexed: 11/12/2022] Open
|
26
|
Zhou J, Liu J, Pan Z, Du X, Li X, Ma B, Yao W, Li Q, Liu H. The let-7g microRNA promotes follicular granulosa cell apoptosis by targeting transforming growth factor-β type 1 receptor. Mol Cell Endocrinol 2015; 409:103-12. [PMID: 25817543 DOI: 10.1016/j.mce.2015.03.012] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 03/18/2015] [Accepted: 03/19/2015] [Indexed: 12/22/2022]
Abstract
The intronic microRNA let-7g controls cell differentiation and proliferation during angiogenesis and oncogenesis. Here, we demonstrate that let-7g regulates granulosa cell (GC) apoptosis and follicular atresia in the pig ovary. Bioinformatics analyses and luciferase reporter assays showed that transforming growth factor-β type 1 receptor (TGFBR1) is a let-7g target. Overexpression of let-7g induced apoptosis of porcine GCs in vitro and repressed the mRNA and protein levels of TGFBR1, as well as the level of phosphorylated SMAD3 (p-SMAD3) protein. RNA interference-mediated knockdown of TGFBR1 and inhibitor LY2157299-mediated blocking of TGFBR1 significantly increased the rate of apoptosis of GCs and Caspase-3 activity. In addition, treatment of porcine GCs with TGF-β1 reduced the level of let-7g and increased the levels of the TGFBR1 mRNA and proteins significantly. Overall, these results demonstrate that let-7g regulates the apoptosis of GCs in the pig ovary by targeting TGFBR1 and down-regulating the TGF-β signaling pathway.
Collapse
Affiliation(s)
- Jilong Zhou
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Jiying Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Zengxiang Pan
- 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
| | - Xinyu Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Baiquan Ma
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Wang Yao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Qifa Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Honglin Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
27
|
Liu J, Yao W, Yao Y, Du X, Zhou J, Ma B, Liu H, Li Q, Pan Z. MiR-92a inhibits porcine ovarian granulosa cell apoptosis by targeting Smad7 gene. FEBS Lett 2015; 588:4497-503. [PMID: 25448599 DOI: 10.1016/j.febslet.2014.10.021] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2014] [Revised: 09/30/2014] [Accepted: 10/19/2014] [Indexed: 02/06/2023]
Abstract
Smad7 has a key role in apoptosis of mammalian ovarian granulosa cells (GCs), as it antagonizes and fine-tunes transforming growth factor β (TGFβ) signaling. This study demonstrates that miR-92a regulates GC apoptosis in pig ovaries by targeting Smad7 directly. The expression level of miR-92a was down-regulated in atretic porcine follicles, whereas miR-92a expression led to inhibition of GC apoptosis. The Smad7 gene was identified as a direct target of miR-92a using a dual-luciferase reporter assay. Transfection of GCs with miR-92a mimics decreased Smad7 mRNA and protein levels, whereas expression of an miR-92a inhibitor in GCs had the opposite effect. In addition, knockdown of Smad7 prevented GC apoptosis in cells that expressed the miR-92a inhibitor.
Collapse
Affiliation(s)
- Jiying Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, People’s Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
Transforming growth factor beta (TGFB) superfamily signaling regulates essential reproductive functions. Dysregulation of TGFB signaling results in cellular and molecular deficiencies in the ovary, leading to reproductive diseases and cancer development. SMAD proteins are canonical TGFB signaling components consisting of receptor-regulated SMADs (SMAD1/2/3/5/9), a common SMAD (SMAD4), and inhibitory SMADs (SMAD6/7). Inhibitory SMADs are negative regulators of TGFB and bone morphogenetic protein signaling, and their reproductive functions are poorly defined. Emerging evidence supports that inhibitory SMADs are potential regulators of ovarian function. Further efforts and new genetic models are needed to unveil the role of inhibitory SMADs in the ovary.
Collapse
Affiliation(s)
- Qinglei Li
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas
| |
Collapse
|
29
|
Gao Y, Duran S, Lydon JP, DeMayo FJ, Burghardt RC, Bayless KJ, Bartholin L, Li Q. Constitutive activation of transforming growth factor Beta receptor 1 in the mouse uterus impairs uterine morphology and function. Biol Reprod 2014; 92:34. [PMID: 25505200 DOI: 10.1095/biolreprod.114.125146] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Despite increasing evidence pointing to the essential involvement of the transforming growth factor beta (TGFB) superfamily in reproduction, a definitive role of TGFB signaling in the uterus remains to be unveiled. In this study, we generated a gain-of-function mouse model harboring a constitutively active (CA) TGFB receptor 1 (TGFBR1), the expression of which was conditionally induced by the progesterone receptor (Pgr)-Cre recombinase. Overactivation of TGFB signaling was verified by enhanced phosphorylation of SMAD2 and increased expression of TGFB target genes in the uterus. TGFBR1 Pgr-Cre CA mice were sterile. Histological, cellular, and molecular analyses demonstrated that constitutive activation of TGFBR1 in the mouse uterus promoted formation of hypermuscled uteri. Accompanying this phenotype was the upregulation of a battery of smooth muscle genes in the uterus. Furthermore, TGFB ligands activated SMAD2/3 and stimulated the expression of a smooth muscle maker gene, alpha smooth muscle actin (ACTA2), in human uterine smooth muscle cells. Immunofluorescence microscopy identified a marked reduction of uterine glands in TGFBR1 Pgr-Cre CA mice within the endometrial compartment that contained myofibroblast-like cells. Thus, constitutive activation of TGFBR1 in the mouse uterus caused defects in uterine morphology and function, as evidenced by abnormal myometrial structure, dramatically reduced uterine glands, and impaired uterine decidualization. These results underscore the importance of a precisely controlled TGFB signaling system in establishing a uterine microenvironment conducive to normal development and function.
Collapse
Affiliation(s)
- Yang Gao
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, Texas
| | - Samantha Duran
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, Texas
| | - John P Lydon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Francesco J DeMayo
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Robert C Burghardt
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, Texas
| | - Kayla J Bayless
- Department of Molecular & Cellular Medicine, Texas A&M Health Science Center, Texas A&M University, College Station, Texas
| | - Laurent Bartholin
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR5286, Université Lyon 1, Centre Léon Bérard, Lyon, France
| | - Qinglei Li
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, Texas
| |
Collapse
|
30
|
Li Q. Transforming growth factor β signaling in uterine development and function. J Anim Sci Biotechnol 2014; 5:52. [PMID: 25478164 PMCID: PMC4255921 DOI: 10.1186/2049-1891-5-52] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 10/28/2014] [Indexed: 12/16/2022] Open
Abstract
Transforming growth factor β (TGFβ) superfamily is evolutionarily conserved and plays fundamental roles in cell growth and differentiation. Mounting evidence supports its important role in female reproduction and development. TGFBs1-3 are founding members of this growth factor family, however, the in vivo function of TGFβ signaling in the uterus remains poorly defined. By drawing on mouse and human studies as a main source, this review focuses on the recent progress on understanding TGFβ signaling in the uterus. The review also considers the involvement of dysregulated TGFβ signaling in pathological conditions that cause pregnancy loss and fertility problems in women.
Collapse
Affiliation(s)
- Qinglei Li
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843 USA
| |
Collapse
|
31
|
Wang Y, Wang XH, Fan DX, Zhang Y, Li MQ, Wu HX, Jin LP. PCSK6 regulated by LH inhibits the apoptosis of human granulosa cells via activin A and TGFβ2. J Endocrinol 2014; 222:151-60. [PMID: 24860148 DOI: 10.1530/joe-13-0592] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Mammalian proprotein convertases (PCs) play an important role in folliculogenesis, as they proteolytically activate a variety of substrates such as the transforming growth factor beta (TGFβ) superfamily. PC subtilism/kexin 6 (PCSK6) is a member of the PC family and is ubiquitously expressed and implicated in many physiological and pathological processes. However, in human granulosa cells, the expression of the PC family members, their hormonal regulation, and the function of PCs are not clear. In this study, we found that PCSK6 is the most highly expressed PC family member in granulosa cells. LH increased PCSK6 mRNA level and PCSK6 played an anti-apoptosis function in KGN cells. Knockdown of PCSK6 not only increased the secretion of activin A and TGFβ2 but also decreased the secretion of follistatin, estrogen, and the mRNA levels of FSH receptor (FSHR) and P450AROM (CYP19A1). We also found that, in the KGN human granulosa cell line, TGFβ2 and activin A could promote the apoptosis of KGN cells and LH could regulate the follistatin level. These data indicate that PCSK6, which is regulated by LH, is highly expressed in human primary granulosa cells of pre-ovulatory follicles and plays important roles in regulating a series of downstream molecules and apoptosis of KGN cells.
Collapse
Affiliation(s)
- Ying Wang
- Laboratory for Reproductive ImmunologyHospital and Institute of Obstetrics and Gynecology, Fudan University Shanghai Medical College, No. 413, Zhaozhou Road, Shanghai 200011, ChinaShanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghai 200011, ChinaDepartment of Reproductive MedicineShanghai First Maternity and Infant Hospital, Tongji University School of Medicine, No. 536, Changle Road, Shanghai 200040, ChinaLaboratory for Reproductive ImmunologyHospital and Institute of Obstetrics and Gynecology, Fudan University Shanghai Medical College, No. 413, Zhaozhou Road, Shanghai 200011, ChinaShanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghai 200011, ChinaDepartment of Reproductive MedicineShanghai First Maternity and Infant Hospital, Tongji University School of Medicine, No. 536, Changle Road, Shanghai 200040, China
| | - Xiao-Hui Wang
- Laboratory for Reproductive ImmunologyHospital and Institute of Obstetrics and Gynecology, Fudan University Shanghai Medical College, No. 413, Zhaozhou Road, Shanghai 200011, ChinaShanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghai 200011, ChinaDepartment of Reproductive MedicineShanghai First Maternity and Infant Hospital, Tongji University School of Medicine, No. 536, Changle Road, Shanghai 200040, ChinaLaboratory for Reproductive ImmunologyHospital and Institute of Obstetrics and Gynecology, Fudan University Shanghai Medical College, No. 413, Zhaozhou Road, Shanghai 200011, ChinaShanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghai 200011, ChinaDepartment of Reproductive MedicineShanghai First Maternity and Infant Hospital, Tongji University School of Medicine, No. 536, Changle Road, Shanghai 200040, China
| | - Deng-Xuan Fan
- Laboratory for Reproductive ImmunologyHospital and Institute of Obstetrics and Gynecology, Fudan University Shanghai Medical College, No. 413, Zhaozhou Road, Shanghai 200011, ChinaShanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghai 200011, ChinaDepartment of Reproductive MedicineShanghai First Maternity and Infant Hospital, Tongji University School of Medicine, No. 536, Changle Road, Shanghai 200040, ChinaLaboratory for Reproductive ImmunologyHospital and Institute of Obstetrics and Gynecology, Fudan University Shanghai Medical College, No. 413, Zhaozhou Road, Shanghai 200011, ChinaShanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghai 200011, ChinaDepartment of Reproductive MedicineShanghai First Maternity and Infant Hospital, Tongji University School of Medicine, No. 536, Changle Road, Shanghai 200040, China
| | - Yuan Zhang
- Laboratory for Reproductive ImmunologyHospital and Institute of Obstetrics and Gynecology, Fudan University Shanghai Medical College, No. 413, Zhaozhou Road, Shanghai 200011, ChinaShanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghai 200011, ChinaDepartment of Reproductive MedicineShanghai First Maternity and Infant Hospital, Tongji University School of Medicine, No. 536, Changle Road, Shanghai 200040, ChinaLaboratory for Reproductive ImmunologyHospital and Institute of Obstetrics and Gynecology, Fudan University Shanghai Medical College, No. 413, Zhaozhou Road, Shanghai 200011, ChinaShanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghai 200011, ChinaDepartment of Reproductive MedicineShanghai First Maternity and Infant Hospital, Tongji University School of Medicine, No. 536, Changle Road, Shanghai 200040, China
| | - Ming-Qing Li
- Laboratory for Reproductive ImmunologyHospital and Institute of Obstetrics and Gynecology, Fudan University Shanghai Medical College, No. 413, Zhaozhou Road, Shanghai 200011, ChinaShanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghai 200011, ChinaDepartment of Reproductive MedicineShanghai First Maternity and Infant Hospital, Tongji University School of Medicine, No. 536, Changle Road, Shanghai 200040, ChinaLaboratory for Reproductive ImmunologyHospital and Institute of Obstetrics and Gynecology, Fudan University Shanghai Medical College, No. 413, Zhaozhou Road, Shanghai 200011, ChinaShanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghai 200011, ChinaDepartment of Reproductive MedicineShanghai First Maternity and Infant Hospital, Tongji University School of Medicine, No. 536, Changle Road, Shanghai 200040, China
| | - Hai-Xia Wu
- Laboratory for Reproductive ImmunologyHospital and Institute of Obstetrics and Gynecology, Fudan University Shanghai Medical College, No. 413, Zhaozhou Road, Shanghai 200011, ChinaShanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghai 200011, ChinaDepartment of Reproductive MedicineShanghai First Maternity and Infant Hospital, Tongji University School of Medicine, No. 536, Changle Road, Shanghai 200040, China
| | - Li-Ping Jin
- Laboratory for Reproductive ImmunologyHospital and Institute of Obstetrics and Gynecology, Fudan University Shanghai Medical College, No. 413, Zhaozhou Road, Shanghai 200011, ChinaShanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghai 200011, ChinaDepartment of Reproductive MedicineShanghai First Maternity and Infant Hospital, Tongji University School of Medicine, No. 536, Changle Road, Shanghai 200040, ChinaLaboratory for Reproductive ImmunologyHospital and Institute of Obstetrics and Gynecology, Fudan University Shanghai Medical College, No. 413, Zhaozhou Road, Shanghai 200011, ChinaShanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghai 200011, ChinaDepartment of Reproductive MedicineShanghai First Maternity and Infant Hospital, Tongji University School of Medicine, No. 536, Changle Road, Shanghai 200040, China
| |
Collapse
|
32
|
Juanchich A, Le Cam A, Montfort J, Guiguen Y, Bobe J. Identification of Differentially Expressed miRNAs and Their Potential Targets During Fish Ovarian Development1. Biol Reprod 2013; 88:128. [DOI: 10.1095/biolreprod.112.105361] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
|
33
|
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
|