1
|
Chen L, Wu Y, Lv T, Tuo R, Xiao Y. Mesenchymal stem cells enchanced by salidroside to inhibit ferroptosis and improve premature ovarian insufficiency via Keap1/Nrf2/GPX4 signaling. Redox Rep 2025; 30:2455914. [PMID: 39874130 PMCID: PMC11776066 DOI: 10.1080/13510002.2025.2455914] [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] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Regenerative medicine researches have shown that mesenchymal stem cells (MSCs) may be an effective treatment method for premature ovarian insufficiency (POI). However, the efficacy of MSCs is still limited. PURPOSE This study aims to explain whether salidroside and MSCs combination is a therapeutic strategy to POI and to explore salidroside-enhanced MSCs inhibiting ferroptosis via Keap1/Nrf2/GPX4 signaling. METHODS The effect of salidroside and MSCs on ovarian granular cells (GCs) was analyzed. After treatment, hormone levels and -fertility of rats were measured. Lipid peroxidation levels, iron deposition and mitochondrial morphology were detected. The genes and proteins of Keap1/Nrf2/GPX4 signaling were examined. RESULTS Salidroside and MSCs were found to inhibit cell death of GCs by reducing peroxidation and intracellular ferrous. Salidroside promotes the proliferation of MSCs and supports cell survival in ovary. Salidroside combined with MSCs therapy restored ovarian function, which was better than MSCs monotherapy. Salidroside-enhanced MSCs to inhibit ferroptosis. The results showed activation of the Keap1/Nrf2/GPX4 signaling and an increase in anti-ferroptosis molecule. CONCLUSIONS Salidroside-enhanced MSCs as a ferroptosis inhibitor and provide new therapeutic strategies for POI. The possible mechanisms of MSCs were related to maintaining redox homeostasis via a Keap1/Nrf2/GPX4 signaling.
Collapse
Affiliation(s)
- Lixuan Chen
- Department of Hematology, Shenzhen Qianhai Shekou Pilot Free Trade Zone Hospital, Shenzhen, People’s Republic of China
| | - Yingnan Wu
- Translational Medicine Center, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Tiying Lv
- Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Rui Tuo
- Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Yang Xiao
- Department of Hematology, Shenzhen Qianhai Shekou Pilot Free Trade Zone Hospital, Shenzhen, People’s Republic of China
- Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| |
Collapse
|
2
|
Mao K, Yan HZ, Yang JQ, Yin ZX, Gao XQ, Liu HF, Su H, Geng YY, Ge P, Zhou DX. The role of ATG7-mediated ferroptosis in formaldehyde-induced ovarian injury in rats. Reprod Toxicol 2025; 134:108902. [PMID: 40164397 DOI: 10.1016/j.reprotox.2025.108902] [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: 02/18/2025] [Revised: 03/26/2025] [Accepted: 03/27/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND To investigate the mechanisms of toxicological damage of formaldehyde to the female reproductive system METHODS: A Mendelian randomization method was used to predict a causal relationship between ferroptosis genes and female infertility. Changes in the structure and function of rat ovaries were observed by HE staining and transmission electron microscopy. Combined with molecular experiments, we verified the alterations of GSH, iron ions, MDA levels, and the expression of ferroptosis genes. RESULTS Mendelian randomization suggested a significant causal relationship between ATG7 and female infertility. Animal experiments showed that formaldehyde caused abnormal follicular maturation and ovarian mitochondrial alterations. Meanwhile, formaldehyde induced elevated levels of iron ions and MDA and decreased GSH levels. Further detection by Western blotting and qRT-PCR revealed that the expression of the ferroptosis marker gene GPX4 decreased and ATG7 increased in the ovary after formaldehyde stimulation. CONCLUSIONS Formaldehyde affected the structure and physiological function of rat ovary by activating the expression level of ATG7 and inhibiting the activity of GPX4, which caused iron overload and elevated levels of peroxides, leading to the occurrence of ferroptosis in ovarian tissue.
Collapse
Affiliation(s)
- Kun Mao
- Zonglian college, Medical School, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Hao-Zhe Yan
- Zonglian college, Medical School, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Jia-Qi Yang
- Zonglian college, Medical School, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Zi-Xin Yin
- Zonglian college, Medical School, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Xin-Qi Gao
- Zonglian college, Medical School, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Hao-Fu Liu
- Zonglian college, Medical School, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Heng Su
- Zonglian college, Medical School, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yuan-Yuan Geng
- Department of Pathology, The Second Afliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Pan Ge
- Department of Pathology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Institute of Genetics and Developmental Biology, Medical School, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi 710061, China.
| | - Dang-Xia Zhou
- Department of Pathology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Institute of Genetics and Developmental Biology, Medical School, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi 710061, China.
| |
Collapse
|
3
|
Hu Z, Gao J, Long P, Quan R, Huang F, Jiang J, Zhang J, Chen J, Xiao H, Huang H. CKAP5 deficiency induces premature ovarian insufficiency. EBioMedicine 2025; 115:105718. [PMID: 40252251 DOI: 10.1016/j.ebiom.2025.105718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 04/07/2025] [Accepted: 04/07/2025] [Indexed: 04/21/2025] Open
Abstract
BACKGROUND Premature ovarian insufficiency (POI) is characterized by ovarian dysfunction that develops from diminished ovarian reserve (DOR). The exact aetiology of POI remains poorly understood. This study aims to elucidate the role of CKAP5 in the regulation of ovarian function and fertility. METHODS Bulk RNA sequencing of granulosa cells was conducted in the control group and in the patients with DOR to screen for candidate genes, which were further validated by gene burden analysis in a next-generation sequencing cohort of POI and control individuals. Additionally, ovarian reserve was evaluated in heterozygous Ckap5 knockout mice, alongside the ovarian and oocyte single-cell transcriptome analysis. The regulatory mechanism of CKAP5 was studied through in vivo and in vitro experiments. FINDINGS CKAP5 was identified as a key hub gene associated with ovarian ageing. Heterozygous Ckap5 knockout mice exhibited a POI-like phenotype, characterized by a reduced primordial follicle pool and accelerated follicular atresia. CKAP5 promotes autophagy via ATG7 and simultaneously supports DNA damage repair through the ATM. Finally, a variant in CKAP5 (NM_0001008938.4, c.630 + 7_630 + 11delCAAAA) was identified in patients with POI, resulting in protein truncation and loss of function. INTERPRETATION CKAP5 deficiency induces premature ovarian insufficiency in both humans and mice. FUNDING The National Key R&D Program of China (2017YFC1001100), the National Natural Science Foundation of China (81501248, 81471453 and 81801295), the Health Research Project of Hunan Provincial Health Commission (W20243018), the Science and Technology Innovation Program of Hunan Province (2021RC3031), the National Natural Science Foundation of Hunan Province (2022JJ30066), the Scientific Research Program of Hunan Provincial Health Commission (202205033471 and 21B0058), the Open Research Fund of Hunan Provincial Key Laboratory of Regional Hereditary Birth Defects Prevention and Control (HPKL2023013).
Collapse
Affiliation(s)
- Zihao Hu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Institute of Reproductive & Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Jingping Gao
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Panpan Long
- Institute of Reproductive & Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Ruping Quan
- Institute of Reproductive & Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Fei Huang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jixuan Jiang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jing Zhang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jianlin Chen
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Hongmei Xiao
- Institute of Reproductive & Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, China.
| | - Hualin Huang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
4
|
Ruan H, Xiang H, Liu Y, Wang P, Dong L, Cao Y, Liang D, Ding Z. FSP1 regulates ferroptosis and mitochondrial function during mouse oocyte maturation. Exp Cell Res 2025; 447:114524. [PMID: 40113029 DOI: 10.1016/j.yexcr.2025.114524] [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: 02/09/2025] [Revised: 03/17/2025] [Accepted: 03/17/2025] [Indexed: 03/22/2025]
Abstract
Oocyte quality plays a fundamental role in fertilization and embryonic development. Emerging evidence indicates that ferroptosis may impair oocyte quality. Ferroptosis suppressor protein 1 (FSP1), a known ferroptosis inhibitor, has an uncharacterized function in regulating oocyte quality during meiotic maturation. This study identified FSP1 expression across all stages of meiotic maturation with localization to the cytoplasm of mouse oocytes. Aged mice exhibited a marked reduction in FSP1 expression within the ovaries and oocytes. Pharmacological inhibition of FSP1 disrupted germinal vesicle breakdown and polar body emission, leading to spindle defects and chromosome misalignment. Additionally, FSP1 inhibition persistently activated the spindle assembly checkpoint, resulting in meiotic arrest. At the mechanistic level, inhibition of FSP1 led to an increase in intracellular Fe2+ levels, enhanced dihydroethidium fluorescence, excessive accumulation of reactive oxygen species, and intensified lipid peroxidation. Disruptions in ferroptosis-associated gene expression further indicated that oocytes underwent ferroptosis. Moreover, mitochondrial dysfunction was evident following FSP1 inhibition, as reflected by aberrant mitochondrial distribution, diminished ATP production, and an elevated mitochondrial membrane potential. Collectively, these results establish FSP1 as a key regulator of oocyte meiotic maturation by modulating iron homeostasis and mitochondrial function, while its inhibition triggers ferroptosis-dependent meiotic failure.
Collapse
Affiliation(s)
- Hongzhen Ruan
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Hefei, 230022, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No.81 Meishan Road, Hefei, 230032, China; Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, No.81 Meishan Road, Hefei, 230032, China
| | - Huifen Xiang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Hefei, 230022, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No.81 Meishan Road, Hefei, 230032, China; Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, No.81 Meishan Road, Hefei, 230032, China
| | - Yajing Liu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Hefei, 230022, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No.81 Meishan Road, Hefei, 230032, China; Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, No.81 Meishan Road, Hefei, 230032, China
| | - Peiwen Wang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Hefei, 230022, China
| | - Liuliu Dong
- Bengbu Medical University, No. 2600 Donghai Avenue, Bengbu, 233030, Anhui, China
| | - Yunxia Cao
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Hefei, 230022, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No.81 Meishan Road, Hefei, 230032, China; Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, No.81 Meishan Road, Hefei, 230032, China.
| | - Dan Liang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Hefei, 230022, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No.81 Meishan Road, Hefei, 230032, China; Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, No.81 Meishan Road, Hefei, 230032, China.
| | - Zhiming Ding
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No.81 Meishan Road, Hefei, 230032, China; Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, No.81 Meishan Road, Hefei, 230032, China.
| |
Collapse
|
5
|
Sun X, Teng R, Xu N, Sun Y, Zhang E, Chen X, Guo Q, Li S. PFOS exposure impairs porcine oocyte maturation and embryo development via mitochondria-dependent ferroptosis. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 374:126185. [PMID: 40189092 DOI: 10.1016/j.envpol.2025.126185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/30/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025]
Abstract
Perfluorooctane sulfonate (PFOS) is a widely utilized chemical known for its exceptional environmental stability over extended periods, its significant potential to bioaccumulate in living organisms, and its considerable risks to both health and the environment. Several studies have suggested that PFOS may pose reproductive risks in mammals; however, the exact mechanisms driving these effects are not well understood. In this study, we explored the possible mechanisms by which PFOS toxicity affects the maturation of mammalian oocytes and the embryonic development employing porcine oocytes as a model system. SMART-seq results suggested that PFOS may affect oocyte maturation through mechanisms involving ferroptosis, autophagy, and alterations in membrane structure. Our results suggest that PFOS exposure adversely affects mitochondrial function and structure, thereby influencing peroxisome biogenesis and contributing to oxidative stress. Most importantly, we found that exposure to PFOS significantly elevated Fe2+ levels, an indicator associated with ferroptosis in oocytes. Furthermore, malondialdehyde (MDA) levels in the PFOS group were significantly higher than those in the control group. Additionally, the mRNA expression levels of PCBP1 and PCBP2, which are related to ferroptosis, as well as the expression level of P53, were significantly reduced in the PFOS group. Overall, exposure to PFOS in vitro results in mitochondrial damage in porcine oocytes, which induces lipid peroxidation and subsequently leads to the occurrence of ferroptosis.
Collapse
Affiliation(s)
- Xiaoqing Sun
- Jilin Agricultural University, Xincheng Street 2888, Changchun, 130118, China
| | - Ran Teng
- Jilin Agricultural University, Xincheng Street 2888, Changchun, 130118, China
| | - Ning Xu
- Jilin Agricultural University, Xincheng Street 2888, Changchun, 130118, China
| | - Yutong Sun
- Affiliated Middle School to Jilin University, Changchun, 130000, China
| | - Enbo Zhang
- Jilin Agricultural University, Xincheng Street 2888, Changchun, 130118, China
| | - Xingfu Chen
- Jilin Agricultural University, Xincheng Street 2888, Changchun, 130118, China
| | - Qing Guo
- College of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, 163319, China
| | - Suo Li
- Jilin Agricultural University, Xincheng Street 2888, Changchun, 130118, China.
| |
Collapse
|
6
|
Weng Y, Zhang W, Qu F, Deng Z, Zhang X, Liu S, Wei H, Hao T, Gao L, Zhang M, Chen Y. Human platelet-rich plasma promotes primordial follicle activation via the PI3K/Akt signaling pathway. Mol Hum Reprod 2025; 31:gaaf007. [PMID: 40088933 DOI: 10.1093/molehr/gaaf007] [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: 05/15/2024] [Revised: 01/18/2025] [Indexed: 03/17/2025] Open
Abstract
The activation of dormant primordial follicles is a promising method to improve the fertility of premature ovarian insufficiency (POI) patients. Many experiments from both human and animal studies suggest that human platelet-rich plasma (hPRP) may restore ovarian function and promote follicle growth. However, the underlying mechanisms remain unclear. In the current study, our results demonstrate that hPRP significantly increased the number of growing follicles and promoted the proliferation of granulosa cells in cultured mouse ovaries. hPRP also significantly increased the protein levels of phosphorylated protein kinase B (p-Akt) and forkhead box O3a (p-FOXO3a), as well as the number of oocytes with FOXO3a nuclear export in cultured mouse ovaries. Immunofluorescence results showed that in vitro treatment with hPRP significantly increased the fluorescence intensity of p-Akt in oocytes. The inhibition of the phosphatidylinositol 3-kinase (PI3K)/Akt pathway by LY294002 blocked the hPRP-induced increase in the number of growing follicles in cultured mouse ovaries. Furthermore, hPRP injected i.p. or added to the medium significantly increased the number of growing follicles and the protein levels of p-Akt in the ovaries of newborn mice and in cultured human ovarian tissues. Taken together, our findings from mouse and human experiments indicate that hPRP promotes the activation of primordial follicles through the PI3K/Akt signaling pathway in oocytes.
Collapse
Affiliation(s)
- Yashuang Weng
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China
- Center for Reproductive Medicine, Zhongshan City People's Hospital, Zhongshan, China
| | - Wenbo Zhang
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China
| | - Fan Qu
- Center for Reproductive Medicine, Zhongshan City People's Hospital, Zhongshan, China
| | - Zehua Deng
- Center for Reproductive Medicine, Zhongshan City People's Hospital, Zhongshan, China
| | - Xiaodan Zhang
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China
| | - Shuang Liu
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China
| | - Hongwei Wei
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China
| | - Tiantian Hao
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China
| | - Longwei Gao
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China
| | - Meijia Zhang
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yuezhou Chen
- Center for Reproductive Medicine, Zhongshan City People's Hospital, Zhongshan, China
| |
Collapse
|
7
|
Wu Y, Zhang Z, Xu Y, Zhang Y, Chen L, Zhang Y, Hou K, Yang M, Jin Z, Cai Y, Zhao J, Sun S. A high-resolution N-glycoproteome landscape of aging mouse ovary. Redox Biol 2025; 81:103584. [PMID: 40073759 PMCID: PMC11938160 DOI: 10.1016/j.redox.2025.103584] [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: 01/16/2025] [Revised: 02/19/2025] [Accepted: 03/04/2025] [Indexed: 03/14/2025] Open
Abstract
Ovarian aging typically precedes the decline of other organ systems, yet its molecular mechanisms remain poorly understood. Glycosylation as one of the most important protein modifications has been especially unexplored in this context. Here, we present the first high-resolution glycoproteomic landscape of aging mouse ovaries, uncovering site-specific N-glycan signatures across subcellular components such as high proportions of complex glycans, core fucosylation, and LacdiNAc branches at the zone pellucida. We report three major glycosylation alterations in aged ovaries: the frequently changed core-fucosylation associated with cell adhesion and immune responses, the decreased LacdiNAc glycans on zona pellucida (ZP) responsible for fertility decline, and the increased sialylated glycans modified by Neu5Ac and Neu5Gc playing different roles in immune activation and responses. Integrated multi-omic analyses further highlight the unique role of glycosylation, distinct from phosphorylation, in regulating key signaling pathways, antigen processing and presentation, complement coagulation cascades, ROS biosynthetic and metabolic processes, as well as cell death. This study offers a novel glycobiological perspective on ovarian aging, broadening our understanding of its molecular mechanisms beyond traditional multi-omic approaches.
Collapse
Affiliation(s)
- Yongqi Wu
- Laboratory for Disease Glycoproteomics, College of Life Sciences, Northwest University, Xi'an, 710069, PR China
| | - Zhida Zhang
- Laboratory for Disease Glycoproteomics, College of Life Sciences, Northwest University, Xi'an, 710069, PR China
| | - Yongchao Xu
- Laboratory for Disease Glycoproteomics, College of Life Sciences, Northwest University, Xi'an, 710069, PR China
| | - Yingjie Zhang
- Laboratory for Disease Glycoproteomics, College of Life Sciences, Northwest University, Xi'an, 710069, PR China
| | - Lin Chen
- Laboratory for Disease Glycoproteomics, College of Life Sciences, Northwest University, Xi'an, 710069, PR China
| | - Yiwen Zhang
- Laboratory for Disease Glycoproteomics, College of Life Sciences, Northwest University, Xi'an, 710069, PR China
| | - Ke Hou
- Laboratory for Disease Glycoproteomics, College of Life Sciences, Northwest University, Xi'an, 710069, PR China
| | - Muyao Yang
- Laboratory for Disease Glycoproteomics, College of Life Sciences, Northwest University, Xi'an, 710069, PR China
| | - Zhehui Jin
- Laboratory for Disease Glycoproteomics, College of Life Sciences, Northwest University, Xi'an, 710069, PR China
| | - Yinli Cai
- Laboratory for Disease Glycoproteomics, College of Life Sciences, Northwest University, Xi'an, 710069, PR China
| | - Jiayu Zhao
- Laboratory for Disease Glycoproteomics, College of Life Sciences, Northwest University, Xi'an, 710069, PR China
| | - Shisheng Sun
- Laboratory for Disease Glycoproteomics, College of Life Sciences, Northwest University, Xi'an, 710069, PR China.
| |
Collapse
|
8
|
Chen L, Zeng L, Pan S, Zu L, Pan H, Fan L. β-sitosterol in Yijing Hugui decoction prevents cyclophosphamide-induced premature ovarian insufficiency via the AKT1/Nrf2 pathway. Cytotechnology 2025; 77:76. [PMID: 40078376 PMCID: PMC11893954 DOI: 10.1007/s10616-025-00740-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
Premature ovarian insufficiency (POI) is a condition marked by premature depletion of ovarian function, affecting a significant portion of women. The objective of this study is to assess the therapeutic efficacy of Yijing Hugui decoction (YJHGD) in the treatment of POI and to elucidate its pharmacological mechanisms. In this study, network pharmacology was used to identify key bioactive compounds in YJHGD, and the components were characterized using LC-MS. In vitro, we used KGN cells treated with cyclophosphamide (CP) to model POI. In vivo, a CP-induced POI mouse model was established. The in vitro therapeutic effects of β-sitosterol on CP-treated KGN cells were evaluated through various parameters. These parameters encompass cell viability, oxidative markers, antioxidant indexes, ATP concentration, intracellular ROS levels, apoptosis rate, and apoptosis-related protein expression. The in vivo therapeutic effects of β-sitosterol in POI mice were assessed through H&E staining, circulating reproductive hormone level detection, reproductive hormone receptor expression measurement, oxidative stress profile, and apoptosis assay. The potential protein target of β-sitosterol was identified utilizing molecular docking in conjunction with drug affinity responsive target stability (DARTS). β-sitosterol was identified as a major active component of YJHGD contributing to its therapeutic effects. In β-sitosterol-treated KGN human granulosa cells, oxidative stress and apoptosis were significantly reduced (P < 0.05). The interaction between β-sitosterol and AKT1 was verified. Furthermore, β-sitosterol significantly activated the AKT1/Nrf2 signaling pathway in vivo and in vitro (P < 0.05). AKT1 activator insulin significantly alleviated CP-induced oxidative stress (P < 0.05). Our results suggest that β-sitosterol inhibits oxidative stress and apoptosis by targeting AKT1 and activating the Keap1/Nrf2/HO-1 signaling. In vivo studies demonstrated that β-sitosterol significantly restored ovarian tissue damage in mice, reduced the circulating levels of reproductive hormones, downregulated the expression of reproductive hormone receptors, alleviated oxidative stress and ROS generation, and improved apoptosis (P < 0.05), which was achieved through the AKT1/Nrf2 pathway. In Conclusion, YJHGD possesses therapeutic potential for the treatment of POI. The active compound, β-sitosterol, demonstrated significant anti-POI effects through its interaction with AKT1, leading to the activation of AKT1/Nrf2 signaling pathway. This interaction contributes to the reduction of oxidative stress and the prevention of cellular apoptosis. Our results suggest that β-sitosterol may represent a novel therapeutic approach. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-025-00740-8.
Collapse
Affiliation(s)
- Li Chen
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, No. 16, Airport Road, Baiyun District, Guangzhou, 510405 Guangdong China
| | - Li Zeng
- Gynecology Department, The Seventh Clinical College of Guangzhou University of Chinese Medicine, No. 25, Yu’an Road, Xin’an Street, Bao’an District, Shenzhen, 518100 Guangdong China
| | - Shuyu Pan
- Gynecology Department, The Seventh Clinical College of Guangzhou University of Chinese Medicine, No. 25, Yu’an Road, Xin’an Street, Bao’an District, Shenzhen, 518100 Guangdong China
| | - Li Zu
- Gynecology Department, The Seventh Clinical College of Guangzhou University of Chinese Medicine, No. 25, Yu’an Road, Xin’an Street, Bao’an District, Shenzhen, 518100 Guangdong China
| | - Hongyan Pan
- Gynecology Department, The Seventh Clinical College of Guangzhou University of Chinese Medicine, No. 25, Yu’an Road, Xin’an Street, Bao’an District, Shenzhen, 518100 Guangdong China
| | - Li Fan
- Gynecology Department, The Seventh Clinical College of Guangzhou University of Chinese Medicine, No. 25, Yu’an Road, Xin’an Street, Bao’an District, Shenzhen, 518100 Guangdong China
| |
Collapse
|
9
|
Ma Q, Wu L, Wu J, Ni B, Wang J, Song S. The Mechanistic Study of Mitochondrial Autophagy and Ferroptosis in the Progression of Decreased Ovarian Reserve. Reprod Sci 2025; 32:1202-1212. [PMID: 39948331 DOI: 10.1007/s43032-025-01811-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: 12/03/2024] [Accepted: 01/27/2025] [Indexed: 04/10/2025]
Abstract
OBJECTIVE To investigate the mechanism in the progression of decreased ovarian reserve (DOR). METHODS Three-month-old female SD rats were employed and randomly divided into the model group and the normal group. The model group was intraperitoneally injected with 4-vinylcyclohexene diepoxide (VCD). Thereafter, blood sample from the abdominal aorta was taken, and rats were sacrificed, and ovarian tissues were obtained by laparotomy. RESULTS HE staining results revealed that the model group exhibited significantly reduced ovarian volume, increased follicular atresia, and decreased quantities of growing follicles and corpus luteum, thereby indicating degraded reserve function of ovarian. TEM images revealed that prominent autophagic vacuoles could be observed in the model group, accompanied by the mitochondria shrinkage and generation of the autophagosome. The expression of Pink1, Parkin, BNIP3L and LC3II genes in ovaries of the model group was significantly higher than those of the normal group (P < 0.05). In addition, the protein expression of Pink1, Parkin, BNIP3L and LC3II in ovaries of the model group were higher than those of the normal group (P < 0.05). The expression of Fe2+ and GSH in oocytes of the model group was higher than those of the normal group (P < 0.05). The expression of FTH1 and GPX4 in oocytes of the model group was significantly higher than those of the normal group (P < 0.05). CONCLUSIONS Mitochondrial autophagy and ferroptosis may participate in the progression of decreased ovarian reserve (DOR).
Collapse
Affiliation(s)
- Qianwen Ma
- TCM Department, Hangzhou Ninth People's Hospital, Number 98, Yilong Road, Yipeng Street, Qiantang District, Hangzhou, 311225, Zhejiang, China.
| | - Lifei Wu
- Sports Department, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jianfei Wu
- TCM Department, Hangzhou Ninth People's Hospital, Number 98, Yilong Road, Yipeng Street, Qiantang District, Hangzhou, 311225, Zhejiang, China
| | - Binfei Ni
- TCM Department, Hangzhou Ninth People's Hospital, Number 98, Yilong Road, Yipeng Street, Qiantang District, Hangzhou, 311225, Zhejiang, China
| | - Jiajia Wang
- TCM Department, Hospital of Taizhou, Number 150, Ximen Street, Taizhou, 317000, Zhejiang, China.
| | - Shiyan Song
- Gynecology Department, Nanjing Hospital of Chinese Medicine, Number 157, Daming Road, Qinhuai District, Nanjing, 210000, Jiangsu, China.
| |
Collapse
|
10
|
Hu J, Wang H, Fang J, Jiang R, Kong Y, Zhang T, Yang G, Jin H, Shi S, Song N, Qi L, Huang X, Wu Z, Yao G. Ovarian aging-associated downregulation of GPX4 expression regulates ovarian follicular development by affecting granulosa cell functions and oocyte quality. FASEB J 2025; 39:e70469. [PMID: 40100097 DOI: 10.1096/fj.202401580rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 02/17/2025] [Accepted: 03/07/2025] [Indexed: 03/20/2025]
Abstract
In the physiological state, female fertility declines with age, as evidenced by a steady decline in oocyte quantity and quality. Aging of the first organ, the ovary, is accompanied by increased oxidative stress levels in the ovary, causing a decline in the ovarian reserve and follicular atresia. Ferroptosis is a novel mode of programmed cell death discovered in recent years and is involved in the onset and development of various diseases. To investigate whether ferroptosis regulates ovarian aging, we first examined granulosa cells from patients with a normal ovarian reserve, decreased ovarian reserve (DOR), and advanced age (Aged). GPX4, a key gene involved in ferroptosis, was identified. The marker of its activity, glutathione (GSH), was significantly downregulated in granulosa cells from the DOR and Aged groups. Transmission electron microscopy confirmed abnormal changes in mitochondrial morphology in granulosa cells from the DOR and Aged groups. In vitro, granulosa cell culture results showed that ferroptosis inducers inhibited cell growth by downregulating GPX4 expression. In contrast, ferroptosis inhibitors reversed the inhibitory effects of ferroptosis on granulosa cell growth by upregulating GPX4 expression. The results of mice in vivo experiments showed that the expression level of GPX4 was significantly decreased in the oocytes of aged mice and that Fer-1, an inhibitor of ferroptosis, reversed the decrease in the number of oocytes retrieved and the quality of oocytes in aged mice. Cyclophosphamide (CTX) was used to generate a mouse model of premature ovarian failure. The results showed that Fer-1 treatment significantly restored the inhibitory effect of CTX on GPX4 expression in the cumulus cells and partially reversed the adverse effects of CTX on the follicular reserve in the ovaries, the number of oocytes retrieved, and the quality of the oocytes in mice. The study findings suggest that ferroptosis is involved in regulating ovarian aging and that GPX4 is a key gene in regulating ovarian follicle development and ferroptosis and a potential key target for treating ovarian aging.
Collapse
Affiliation(s)
- Jingyi Hu
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huihui Wang
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Junnan Fang
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ran Jiang
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yue Kong
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Tongwei Zhang
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guang Yang
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Haixia Jin
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Senlin Shi
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ning Song
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lin Qi
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xianju Huang
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhaoting Wu
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guidong Yao
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
11
|
Yuan Y, Yang X, Zhao Y, Flores JJ, Huang L, Gu L, Li R, Zhang X, Zhu S, Dong S, Kanamaru H, He Q, Tao Y, Yi K, Han M, Chen X, Wu L, Zhang JH, Xie Z, Tang J. Mitochondrial ferritin upregulation by deferiprone reduced neuronal ferroptosis and improved neurological deficits via NDRG1/Yap pathway in a neonatal rat model of germinal matrix hemorrhage. J Cereb Blood Flow Metab 2025; 45:510-527. [PMID: 39318194 PMCID: PMC11563512 DOI: 10.1177/0271678x241252110] [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: 05/15/2023] [Revised: 03/19/2024] [Accepted: 04/05/2024] [Indexed: 09/26/2024]
Abstract
Ferroptosis contributes to brain injury after germinal matrix hemorrhage (GMH). Mitochondrial ferritin (FTMT), a novel mitochondrial outer membrane protein, reduces oxidative stress in neurodegenerative diseases. In vitro, Deferiprone has been shown to upregulate FTMT. However, the effects of FTMT upregulation by Deferiprone on neuronal ferroptosis after GMH and its underlying mechanism has not been investigated. In our study, 389 Sprague-Dawley rat pups of postnatal day 7 were used to establish a collagenase-induced GMH model and an iron-overload model of intracerebral FeCl2 injection. The brain expressions of FTMT, N-myc downstream-regulated gene-1 (NDGR1), Yes-associated protein (YAP), ferroptosis-related molecules including transferrin receptor (TFR) and acyl-CoA synthase long-chain family member 4 (ACSL4) were increased after GMH. FTMT agonist Deferiprone improved neurological deficits and hydrocephalus after GMH. Deferiprone or Adenovirus-FTMT enhanced YAP phosphorylation at the Ser127 site and attenuated ferroptosis, which was reversed by NDRG1 CRISPR Knockout. Iron overload induced neuronal ferroptosis and neurological deficits, which were improved by YAP CRISPR Knockout. Collectively, FTMT upregulation by Deferiprone reduced neuronal ferroptosis and neurological deficits via the NDRG1/YAP signaling pathway after GMH. Deferiprone may serve as a potential non-invasive treatment for GMH patients.
Collapse
Affiliation(s)
- Ye Yuan
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Xiao Yang
- Department of Obstetrics and Gynecology, University‐Town Hospital of Chongqing Medical University, Chongqing, China
| | - Yutong Zhao
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Jerry J Flores
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Lei Huang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, California, USA
- Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Lingui Gu
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT Alliance, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ruihao Li
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Xingyu Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Shiyi Zhu
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Siyuan Dong
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Hideki Kanamaru
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Qiuguang He
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Yihao Tao
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Kun Yi
- Chongqing Key Laboratory of Ophthalmology and Chongqing Eye Institute, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Mingyang Han
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Xionghui Chen
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Lei Wu
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, California, USA
- Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, California, USA
- Departments of Anesthesiology and Neurology, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Zongyi Xie
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, California, USA
| |
Collapse
|
12
|
Yu Z, Zhang X, Nong Y, Ding H, Fu X, Li F, Liu L, Li M, Peng W, Wu H, Liu F. Analysis of post-transcriptional regulatory signatures and immune cell subsets in premature ovarian insufficiency based on full-length transcriptome. Sci Rep 2025; 15:5533. [PMID: 39953072 PMCID: PMC11829046 DOI: 10.1038/s41598-025-89391-5] [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: 08/27/2024] [Accepted: 02/05/2025] [Indexed: 02/17/2025] Open
Abstract
Premature ovarian insufficiency (POI) is a reproductive endocrine disorder characterized by infertility and the perimenopausal syndrome. Its genetic etiology is highly heterogeneous and not yet fully understood. Limited by short-read sequencing, the profile and structural variation of the full-length transcript for POI have remained elusive. Therefore, this study included peripheral blood samples from 5 POI patients and 5 controls, characterizing full-length transcripts of POI using Oxford Nanopore sequencing firstly. Ultimately, we identified 26,122 transcripts, including 7,724 novel gene loci and 13,593 novel transcripts. A total of 382 differentially expressed transcripts were identified, including 366 down-regulated and 16 up-regulated transcripts. Based on transcript structure variant analysis, 8,834 alternative splicing events, 65,254 alternative polyadenylation sites and 32 motifs were further identified, revealing the diversity sources of transcript isoforms, proteins and genetic complexity. Enrichment analysis of differentially AS genes suggested that the ferroptosis pathway may play an important role in the pathogenesis of POI.Additionally, 494 high-confidence lncRNAs, 1,768 transcription factors, and novel gene-coding regions were predicted based on full-length transcript sequence. Analysis of immune cell subtypes revealed the expression of CD8 + T cells and monocytes were down-regulated in POI, which was significantly positively correlated with AMH, suggesting that CD8 + T cells and monocytes could serve as potential diagnostic markers and immunotherapy targets for POI. Conclusively, this study provides new perspectives on the pathogenesis, post-transcriptional regulation mechanisms, and immune targets of POI.
Collapse
Affiliation(s)
- Zhaoyang Yu
- Guangdong Women and Children Hospital, Guangzhou, China
- Guangxi Medical University, Nanning, China
| | - Xiqian Zhang
- Guangdong Women and Children Hospital, Guangzhou, China
| | - Yingqi Nong
- Guangdong Women and Children Hospital, Guangzhou, China
| | - Hongfan Ding
- Guangxi Medical University, Nanning, China
- Shenzhen Baoan distric SongGang People's Hospital, Shenzhen, China
| | - Xiaoqian Fu
- Reproductive Medicine Research Center, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Feiwen Li
- Reproductive Medicine Research Center, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Lidan Liu
- Reproductive Medicine Research Center, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Mujun Li
- Reproductive Medicine Research Center, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Weilong Peng
- School of Computer Science and Cyber Engineering, Guangzhou University, Guangzhou, China
| | - Huimei Wu
- Reproductive Medicine Research Center, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.
| | - Fenghua Liu
- Guangdong Women and Children Hospital, Guangzhou, China.
| |
Collapse
|
13
|
Mu C, Li X, Yang J, Tian GG, Bai H, Lin W, Wang L, Wu J. Spatial Transcriptome and Single Nucleus Transcriptome Sequencing Reveals Tetrahydroxy Stilbene Glucoside Promotes Ovarian Organoids Development Through the Vegfa-Ephb2 Pair. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410098. [PMID: 39629971 PMCID: PMC11775562 DOI: 10.1002/advs.202410098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/03/2024] [Indexed: 01/30/2025]
Abstract
Ovarian dysfunction is a major factor leading to female infertility. Understanding how to improve or reshape ovarian function has become an important entry point for preventing and treating female infertility caused by ovarian dysfunction. Here, plant-derived compounds are screened for in vitro activity upon ovarian organoids derived from feeder-free female germline stem cells. Tetrahydroxy stilbene glucoside (TSG) is found to promote the development of ovarian organoids. Single nucleus transcriptome sequencing and spatial transcriptome sequencing are used to establish a comprehensive spatiotemporal map to elucidate the role of TSG in ovarian organoid development, encompassing cell types and subtypes, transcription factors, pseudo-time sequence, and cell communication dynamics. This analysis indicates that TSG promotes ovarian organoid development through the vascular endothelial growth factor A-Eph receptor B2 ligand-receptor pair between granulosa cells and oocytes. This study has enhanced the understanding of the mechanisms of ovarian organoid development, establishes a technical platform for screening compounds for treating infertility and related diseases, and lays a foundation for clinically applying plant-derived compounds.
Collapse
Affiliation(s)
- Chunlan Mu
- School of Basic Medical SciencesKey Laboratory of Fertility Preservation and Maintenance of Ministry of EducationNingxia Medical UniversityYinchuan750004China
- Key Laboratory for the Genetics of Developmental & Neuropsychiatric Disorders (Ministry of Education)Bio‐X InstitutesShanghai Jiao Tong UniversityShanghai200240China
| | - Xiaoyong Li
- Key Laboratory for the Genetics of Developmental & Neuropsychiatric Disorders (Ministry of Education)Bio‐X InstitutesShanghai Jiao Tong UniversityShanghai200240China
| | - Jiamei Yang
- School of Basic Medical SciencesKey Laboratory of Fertility Preservation and Maintenance of Ministry of EducationNingxia Medical UniversityYinchuan750004China
- School of PharmacologyNingxia Medical UniversityYinchuan750004China
| | - Geng G. Tian
- School of Agriculture and BiologyShanghai Jiao Tong UniversityShanghai200240China
| | - Hepeng Bai
- Key Laboratory for the Genetics of Developmental & Neuropsychiatric Disorders (Ministry of Education)Bio‐X InstitutesShanghai Jiao Tong UniversityShanghai200240China
| | - Wenhui Lin
- School of Life Sciences and BiotechnologyJoint International Research Laboratory of Metabolic and Developmental SciencesShanghai Jiao Tong UniversityShanghai200240China
| | - Linhui Wang
- Department of Urology, Changhai HospitalSecond Military Medical University (Naval Medical University)Shanghai200433China
- Shanghai Key Laboratory of Cell EngineeringShanghai200433China
| | - Ji Wu
- School of Basic Medical SciencesKey Laboratory of Fertility Preservation and Maintenance of Ministry of EducationNingxia Medical UniversityYinchuan750004China
- Key Laboratory for the Genetics of Developmental & Neuropsychiatric Disorders (Ministry of Education)Bio‐X InstitutesShanghai Jiao Tong UniversityShanghai200240China
| |
Collapse
|
14
|
Qin S, Chi X, Zhu Z, Chen C, Zhang T, He M, Gao M, Zhao T, Zhang J, Zhang L, Zheng W, Chen Z, Wang W, Zhou B, Xia G, Wang C. Oocytes maintain low ROS levels to support the dormancy of primordial follicles. Aging Cell 2025; 24:e14338. [PMID: 39297300 PMCID: PMC11709087 DOI: 10.1111/acel.14338] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 08/05/2024] [Accepted: 08/28/2024] [Indexed: 01/11/2025] Open
Abstract
Primordial follicles (PFs) function as the long-term reserve for female reproduction, remaining dormant in the ovaries and becoming progressively depleted with age. Oxidative stress plays an important role in promoting female reproductive senescence during aging, but the underlying mechanisms remain unclear. Here, we find that low levels of reactive oxygen species (ROS) are essential for sustaining PF dormancy. Compared to growing follicles, oocytes within PFs were shown to be more susceptible to ROS, which accumulates and damages PFs to promote reproductive senescence. Mechanistically, oocytes within PFs were shown to express high levels of the intracellular antioxidant enzyme superoxide dismutase 1 (SOD1), counteracting ROS accumulation. Decreased SOD1 expression, as a result of aging or through the experimental deletion of the Sod1 gene in oocytes, resulted in increased oxidative stress and triggered ferroptosis within PFs. In conclusion, this study identified antioxidant defense mechanisms protecting PFs in mouse ovaries and characterized cell death mechanisms of oxidative stress-induced PF death.
Collapse
Affiliation(s)
- Shaogang Qin
- State Key Laboratory of Farm Animal Biotech BreedingCollege of Biological Sciences, China Agricultural UniversityBeijingChina
| | - Xinyue Chi
- State Key Laboratory of Farm Animal Biotech BreedingCollege of Biological Sciences, China Agricultural UniversityBeijingChina
| | - Zijian Zhu
- State Key Laboratory of Farm Animal Biotech BreedingCollege of Biological Sciences, China Agricultural UniversityBeijingChina
| | - Chuanhe Chen
- State Key Laboratory of Animal NutritionInstitute of Animal Science, Chinese Academy of Agricultural SciencesBeijingChina
| | - Tuo Zhang
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Department of PhysiologyCollege of Basic Medicine, Guizhou Medical UniversityGuiyangGuizhou ProvinceChina
| | - Meina He
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Department of PhysiologyCollege of Basic Medicine, Guizhou Medical UniversityGuiyangGuizhou ProvinceChina
| | - Meng Gao
- State Key Laboratory of Farm Animal Biotech BreedingCollege of Biological Sciences, China Agricultural UniversityBeijingChina
| | - Ting Zhao
- State Key Laboratory of Farm Animal Biotech BreedingCollege of Biological Sciences, China Agricultural UniversityBeijingChina
| | - Jingwen Zhang
- State Key Laboratory of Farm Animal Biotech BreedingCollege of Biological Sciences, China Agricultural UniversityBeijingChina
| | - Lifan Zhang
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western ChinaCollege of Life Science, Ningxia UniversityYinchuanChina
| | - Wenying Zheng
- State Key Laboratory of Farm Animal Biotech BreedingCollege of Biological Sciences, China Agricultural UniversityBeijingChina
| | - Ziqi Chen
- State Key Laboratory of Farm Animal Biotech BreedingCollege of Biological Sciences, China Agricultural UniversityBeijingChina
| | - Wenji Wang
- School of Life Science, Taizhou UniversityTaizhouChina
| | - Bo Zhou
- State Key Laboratory of Farm Animal Biotech BreedingCollege of Biological Sciences, China Agricultural UniversityBeijingChina
| | - Guoliang Xia
- State Key Laboratory of Farm Animal Biotech BreedingCollege of Biological Sciences, China Agricultural UniversityBeijingChina
| | - Chao Wang
- State Key Laboratory of Farm Animal Biotech BreedingCollege of Biological Sciences, China Agricultural UniversityBeijingChina
| |
Collapse
|
15
|
Hu M, Qin Y, Jiao X. Can ferroptosis be a target for reproductive health? Trends Endocrinol Metab 2024:S1043-2760(24)00318-7. [PMID: 39706760 DOI: 10.1016/j.tem.2024.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 11/21/2024] [Accepted: 11/25/2024] [Indexed: 12/23/2024]
Abstract
Ferroptosis has been implicated in several reproductive disorders, but the underlying mechanisms remain unknown; thus, interventions targeting this pathway are lacking. Here we summarize the emerging findings on ferroptosis in reproductive biology and corresponding disorders, and highlight perspectives and challenges on future ferroptosis research with potential clinical applications.
Collapse
Affiliation(s)
- Mengchun Hu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children, and Reproductive Health, Shandong University, 250012, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, 250012, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong, 250012, China; Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, 250012, China; Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, 250012, China; Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong, 250012, China; Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, Shandong, 250012, China
| | - Yingying Qin
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children, and Reproductive Health, Shandong University, 250012, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, 250012, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong, 250012, China; Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, 250012, China; Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, 250012, China; Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong, 250012, China; Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, Shandong, 250012, China
| | - Xue Jiao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children, and Reproductive Health, Shandong University, 250012, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, 250012, China; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong, 250012, China; Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, 250012, China; Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, 250012, China; Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong, 250012, China; Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, Shandong, 250012, China.
| |
Collapse
|
16
|
Yang S, Ye Z, Chen W, Wang P, Zhao S, Zhou X, Li W, Cheng F. BMAL1 alleviates sepsis-induced AKI by inhibiting ferroptosis. Int Immunopharmacol 2024; 142:113159. [PMID: 39303541 DOI: 10.1016/j.intimp.2024.113159] [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: 07/01/2024] [Revised: 09/08/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND The role of BMAL1 in various diseases remains unclear, particularly its impact on sepsis-induced acute kidney injury (AKI). This study aims to investigate the role of BMAL1 in sepsis-induced AKI and its potential effects on cell ferroptosis. Initially, we assessed BMAL1 expression levels in mice treated with sepsis-induced AKI (via LPS injection) and in LPS-stimulated renal tubular epithelial cells. Subsequently, we explored the correlation between BMAL1 and ferroptosis using sequencing technology, validating our findings throughout experimental approaches. To further elucidate BMAL1's specific effects on AKI-related ferroptosis, we constructed BMAL1 overexpression models in mice and cells, analysing its impact on AKI and ferroptosis both in vivo and in vitro. Furthermore, using transcriptome sequencing technology, we identified key BMAL1-regulated genes and their associated biological pathways, validating these findings through in vivo and in vitro experiments. RESULTS Our findings indicate decreased BMAL1 expression in sepsis-induced AKI. BMAL1 overexpression effectively mitigated renal tubular injury by reducing ferroptosis levels in renal tubular epithelial cells. Using transcriptome sequencing and ChIP-qPCR technology, we identified YAP as a target of BMAL1. The overexpression of BMAL1 significantly reduced the transcriptional activity of YAP and inhibited the Hippo signalling pathway. Treatment with the Hippo inhibitor Verteporfin (VP) reversed the BMAL1-downregulation-induced damage. Additionally, our study revealed that YAP positively regulates ACSL4 gene expression and its downstream signalling pathways. CONCLUSION This study demonstrates that BMAL1 overexpression alleviates renal tubular epithelial cell injury and ferroptosis by inhibiting YAP expression and the Hippo pathway, thereby exerting protective effects in sepsis-induced AKI. These findings underscore the therapeutic potential of targeting BMAL1 in managing sepsis-induced AKI.
Collapse
Affiliation(s)
- Songyuan Yang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zehua Ye
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Wu Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Peihan Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Shen Zhao
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xiangjun Zhou
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Wei Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Fan Cheng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| |
Collapse
|
17
|
Wang R, Song J, Cai M, Xue Y, Liu J, Zuo N, De Felici M, Wang J, Shen W, Sun X. Gut microbiota modulation by L-Fucose as a strategy to alleviate Ochratoxin A toxicity on primordial follicle formation. JOURNAL OF HAZARDOUS MATERIALS 2024; 480:136469. [PMID: 39536347 DOI: 10.1016/j.jhazmat.2024.136469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/16/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
In this study, we investigated the potential benefits of L-Fucose administration to pregnant mice exposed to Ochratoxin A (OTA), a widespread mycotoxin, producing ovarian damage in offspring. The results showed that administration of 3.5 μg/d OTA induced alterations in intestinal tissues and gut microbiota of pregnant mice, leading to heightened local and systemic inflammation. This inflammatory affected the ovaries of their 3 dpp offspring, in which elevated levels of LPS and ROS were found associated to significant decreased oocyte count and impaired primordial follicle assembly. Moreover, mRNA-Seq analysis showed significant changes in ovarian transcriptomes linked to various GO terms and KEGG pathways, notably ferroptosis, a recognized form of cell death observed. Interestingly, administration of 0.3 g/kg b. w. L-Fucose following OTA exposure mitigated these effects on intestinal tissues and gut microbiota in mothers and on the offspring's ovaries. Similar benefits were obtained by gut microbiota transplantation from L-Fucose-treated pregnant females into OTA-exposed mothers. These findings suggest that inflammatory impact of OTA on maternal intestine/gut can pass to the fetus causing offspring ovary defects and support the use of L-Fucose as adjuvant to counteract the adverse effects of mycotoxins on the gut microbiota, particularly reference to those affecting reproductive organs.
Collapse
Affiliation(s)
- Ruiting Wang
- College of Life Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, China
| | - Jie Song
- College of Life Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, China
| | - Muyu Cai
- College of Life Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, China
| | - Yuan Xue
- College of Life Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, China
| | - Jing Liu
- Central laboratory of Qingdao Agricultural University, Qingdao 266109, China
| | - Ning Zuo
- College of Life Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, China
| | - Massimo De Felici
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome 00133, Italy
| | - Junjie Wang
- College of Life Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, China
| | - Wei Shen
- College of Life Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, China
| | - Xiaofeng Sun
- College of Life Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, China.
| |
Collapse
|
18
|
Zhang T, Zhao L, Tang X. Down-regulated BNC1 promotes glioma by inhibiting ferroptosis via TCF21/PI3K signaling pathway BNC1TCF21PI3K. Tissue Cell 2024; 91:102633. [PMID: 39603023 DOI: 10.1016/j.tice.2024.102633] [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: 08/28/2024] [Revised: 11/14/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024]
Abstract
We elucidate the role of the BNC1 gene in glioma and its potential mechanism. The expression levels of BNC1 in patients with glioma or corresponding cell lines were down-regulated. High BNC1 expression increased survival rate in patients with glioma. BNC1 gene reduced cell proliferation, and enhanced ferroptosis of glioma cells through the induction of TCF21/PI3K signaling pathway. Meanwhile, BNC1 gene could decline tumor proliferation in mice model of glioma. The ferroptosis inhibitor alleviated the impact of BNC1 on glioma ferroptosis, while the ferroptosis agonist weakened the effect of BNC1 on glioma ferroptosis. SiTCF21 also declined the effects of BNC1 on ferroptosis of glioma. The enhanced expression of TCF21 also inhibited the effect of BNC1 on ferroptosis of glioma. BNC1 protein interlinked with TCF21 protein, and bioluminescence imaging demonstrated that BNC1 enhanced TCF21 expression in the brain tissue of the mouse model of glioma. In conclusion, BNC1 reduced cell proliferation, and increased ferroptosis of glioma cells by TCF21/PI3K signaling pathway, may be a feasible strategy to treat glioma.
Collapse
Affiliation(s)
- Tao Zhang
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China
| | - Long Zhao
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China
| | - Xiaoping Tang
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China.
| |
Collapse
|
19
|
Wang R, Geng J. The melatonin-FTO-ATF4 signaling pathway protects granulosa cells from cisplatin-induced chemotherapeutic toxicity by suppressing ferroptosis. J Assist Reprod Genet 2024; 41:3503-3516. [PMID: 39388020 PMCID: PMC11707222 DOI: 10.1007/s10815-024-03276-6] [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: 07/09/2024] [Accepted: 09/22/2024] [Indexed: 10/15/2024] Open
Abstract
PURPOSE In cisplatin-induced premature ovarian failure (POF) mice, granulosa cells showed a high level of ferroptosis. Previous research has indicated that the fat mass and obesity-associated protein/activating transcription factor 4 (FTO/ATF4) axis was involved in the regulation of ferroptosis. The purpose of this study was to explore the role of the FTO/ATF4 axis in cisplatin-induced ferroptosis in granulosa cell. METHODS The extent of ferroptosis was assessed by transmission electron microscopy (TEM) and ROS, GPX, GSH, and MDA assays. Western blotting was used to evaluate the protein expression levels of ferroptosis-related molecules. Ferroptosis activator and inhibitor were also used. RESULTS We found that ferroptosis increased in a concentration-dependent manner in cisplatin-induced injured granulosa cells, accompanied by the downregulation of FTO. In addition, gain- and loss-of-function studies showed that FTO affects ferroptosis in injured cells by regulating ATF4 expression. Ferrostatin-1 inhibited the effect of FTO downregulation on injured granulosa cells ferroptosis, and erastin reversed the protective effect of FTO on ferroptosis in injured granulosa cells. Finally, melatonin was used, and we found that melatonin reduced ferroptosis in cisplatin-induced injured granulosa cells by upregulating FTO expression. CONCLUSION Our study demonstrated that cisplatin induced granulosa cell ferroptosis by downregulating the expression of FTO. ATF4 was identified as a downstream target of FTO, and overexpression of ATF4 reversed the effects of decreased FTO on ferroptosis. Additionally, melatonin mitigates the cytotoxic effects of cisplatin by upregulating FTO expression. The melatonin-FTO-ATF4 signaling pathway plays a vital role in the treatment of cisplatin-induced POF.
Collapse
Affiliation(s)
- Rongli Wang
- 1Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, 100044, China.
| | - Jing Geng
- 1Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, 100044, China.
| |
Collapse
|
20
|
Li Y, Li X, Han Z, Yang R, Zhou W, Peng Y, He J, Liu S. Population structure and selective signature analysis of local sheep breeds in Xinjiang, China based on high-density SNP chip. Sci Rep 2024; 14:28133. [PMID: 39548146 PMCID: PMC11568293 DOI: 10.1038/s41598-024-76573-w] [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: 04/03/2024] [Accepted: 10/15/2024] [Indexed: 11/17/2024] Open
Abstract
The frigid and droughty climate of Xinjiang in China has given rise to unique indigenous sheep breeds with robust adaptability and resistance. To investigate the genetic mechanism of adaptability of Xinjiang sheep to the local extreme environment, we conducted population genetic structure analyses for three native Xinjiang sheep breeds: Altay sheep (ALT), Bashbay Sheep (BSBC), and Duolang sheep (DLC), as well as two foreign sheep breeds: Suffolk and Dorset, using the Ovine Infinium HD SNP BeadChip(680 K). Our findings revealed distinct genetic and evolutionary histories between Xinjiang and foreign sheep breeds. Principal Component Analysis (PCA) and phylogenetic tree effectively differentiate these five sheep breeds based on their geographical origins, and the domestication level of Xinjiang sheep is comparatively lower than that of foreign sheep breeds. Furthermore, by utilizing three selective signature methods, namely Fixation Index (Fst), Cross Population Extended Haplotype Homozygosity Test (XP-EHH), and Nucleotide Diversity (π), we have successfully identified 22 potential candidate genes. Among these genes, there are TBXT, PDGFD, and VEGFA, which are closely related to tail type and lipid metabolism; VIL1, SLC11A1, and ZBTB46, which are associated with immune function; and candidate genes such as BNC1, HDAC1, and BMP5, which impact sheep reproductive traits. This study establishes a foundation for conserving and utilizing local sheep germplasm resources in Xinjiang and provides molecular insights into the genetic mechanisms governing sheep adaptation to extreme cold and arid environments.
Collapse
Affiliation(s)
- Yanhao Li
- College of Animal Science and Technology, Tarim University, Alar, 843300, Xinjiang, China
- Key Laboratory of Tarim Animal Husbandry Science and Technology, Xinjiang Production and Construction Corps, Alar, 843300, xinjiang, China
| | - Xiaopeng Li
- College of Animal Science and Technology, Tarim University, Alar, 843300, Xinjiang, China
- Key Laboratory of Tarim Animal Husbandry Science and Technology, Xinjiang Production and Construction Corps, Alar, 843300, xinjiang, China
| | - Zhipeng Han
- College of Animal Science and Technology, Tarim University, Alar, 843300, Xinjiang, China
- Key Laboratory of Tarim Animal Husbandry Science and Technology, Xinjiang Production and Construction Corps, Alar, 843300, xinjiang, China
| | - Ruizhi Yang
- Key Laboratory of Tarim Animal Husbandry Science and Technology, Xinjiang Production and Construction Corps, Alar, 843300, xinjiang, China
- College of Life Science and Technology, Tarim University, Alar, 843300, Xinjiang, China
| | - Wen Zhou
- College of Animal Science and Technology, Tarim University, Alar, 843300, Xinjiang, China
- Key Laboratory of Tarim Animal Husbandry Science and Technology, Xinjiang Production and Construction Corps, Alar, 843300, xinjiang, China
| | - Yuwei Peng
- College of Animal Science and Technology, Tarim University, Alar, 843300, Xinjiang, China
- Key Laboratory of Tarim Animal Husbandry Science and Technology, Xinjiang Production and Construction Corps, Alar, 843300, xinjiang, China
| | - Jianzhong He
- College of Animal Science and Technology, Tarim University, Alar, 843300, Xinjiang, China.
- Key Laboratory of Tarim Animal Husbandry Science and Technology, Xinjiang Production and Construction Corps, Alar, 843300, xinjiang, China.
| | - Shudong Liu
- College of Animal Science and Technology, Tarim University, Alar, 843300, Xinjiang, China.
- Key Laboratory of Tarim Animal Husbandry Science and Technology, Xinjiang Production and Construction Corps, Alar, 843300, xinjiang, China.
| |
Collapse
|
21
|
Tang S, Guo T, Song C, Wang L, Zhang J, Rajkovic A, Lin X, Chen S, Liu Y, Tian W, Wu B, Wang S, Wang W, Lai Y, Wang A, Xu S, Jin L, Ke H, Zhao S, Li Y, Qin Y, Zhang F, Chen ZJ. MGA loss-of-function variants cause premature ovarian insufficiency. J Clin Invest 2024; 134:e183758. [PMID: 39545409 PMCID: PMC11563689 DOI: 10.1172/jci183758] [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: 06/11/2024] [Accepted: 09/20/2024] [Indexed: 11/17/2024] Open
Abstract
Although premature ovarian insufficiency (POI), a common cause of female infertility and subfertility, has a well-established hereditary component, the genetic factors currently implicated in POI account for only a limited proportion of cases. Here, using an exome-wide, gene-based case-control analysis in a discovery cohort comprising 1,027 POI cases and 2,733 ethnically matched women controls from China, we found that heterozygous loss-of-function (LoF) variants of MAX dimerization protein (MGA) were significantly enriched in the discovery cohort, accounting for 2.6% of POI cases, while no MGA LoF variants were found in the matched control females. Further exome screening was conducted in 4 additional POI cohorts (2 from China and 2 from the United States) for replication studies, and we identified heterozygous MGA LoF variants in 1.0%, 1.4%, 1.0%, and 1.0% of POI cases, respectively. Overall, a total of 37 distinct heterozygous MGA LoF variants were discovered in 38 POI cases, accounting for approximately 2.0% of the total 1,910 POI cases analyzed in this study. Accordingly, Mga+/- female mice were subfertile, exhibiting shorter reproductive lifespan and decreased follicle number compared with WT, mimicking the observed phenotype in humans. Our findings highlight the essential role of MGA deficiency for impaired female reproductive ability.
Collapse
Affiliation(s)
- Shuyan Tang
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, Institute of Medical Genetics and Genomics, Fudan University, Shanghai, China
| | - Ting Guo
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, China
| | - Chengcheng Song
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, Institute of Medical Genetics and Genomics, Fudan University, Shanghai, China
| | - Lingbo Wang
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, Institute of Medical Genetics and Genomics, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Jun Zhang
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Aleksandar Rajkovic
- Department of Pathology, Obstetrics, Gynecology and Reproductive Sciences, University of California San Francisco, San Francisco, California, USA
| | - Xiaoqi Lin
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, Institute of Medical Genetics and Genomics, Fudan University, Shanghai, China
| | - Shiling Chen
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yujun Liu
- Human Phenome Institute, Zhangjiang Fudan International Innovation Center and
| | - Weidong Tian
- School of Life Sciences, Fudan University, Shanghai, China
| | - Bangguo Wu
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenwen Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yunhui Lai
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ao Wang
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shuhua Xu
- Human Phenome Institute, Zhangjiang Fudan International Innovation Center and
- School of Life Sciences, Fudan University, Shanghai, China
| | - Li Jin
- Human Phenome Institute, Zhangjiang Fudan International Innovation Center and
- School of Life Sciences, Fudan University, Shanghai, China
| | - Hanni Ke
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, Institute of Medical Genetics and Genomics, Fudan University, Shanghai, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, China
| | - Shidou Zhao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, China
| | - Yan Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingying Qin
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, China
| | - Feng Zhang
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, Institute of Medical Genetics and Genomics, Fudan University, Shanghai, China
- Human Phenome Institute, Zhangjiang Fudan International Innovation Center and
- Shanghai Key Laboratory of Embryo Original Diseases, Soong Ching Ling Institute of Maternity and Child Health, International Peace Maternity and Child Health Hospital of China Welfare Institute, Shanghai, China
| | - Zi-Jiang Chen
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (no. 2021RU001), Jinan, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| |
Collapse
|
22
|
Guo Z, Zeng Q, Li Q, Shan B, Huo Y, Shi X, Li Q, Du X. LncRNA NORFA promotes the synthesis of estradiol and inhibits the apoptosis of sow ovarian granulosa cells through SF-1/CYP11A1 axis. Biol Direct 2024; 19:107. [PMID: 39523350 PMCID: PMC11552157 DOI: 10.1186/s13062-024-00563-1] [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] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Biosynthesis of 17β-estradiol (E2) is a crucial ovarian function in mammals, which is essential for follicular development and pregnancy outcome. Exploring the epigenetic regulation of E2 synthesis is beneficial for maintaining ovary health and the optimal reproductive traits. NORFA is the first validated sow fertility-associated long non-coding RNA (lncRNA). However, its role on steroidogenesis is elusive. The aim of this study is to investigate the regulation and underlying mechanism of NORFA to E2 synthesis in sow granulosa cells (GCs). RESULTS Through Pearson correlation analysis and comparative detection, we found that NORFA expression was positively correlated with the levels of pregnenolone (PREG) and E2 in follicles, which also exhibited similar alteration patterns during follicular atresia. ELISA was conducted and indicated for the first time that NORFA induced the synthesis of PREG and E2 in sow GCs in a dose- and time-dependent manner. RNA-seq, GSEA and quantitative analyses results validated that CYP11A1, the coding gene of P450SCC which is the first step rate-limiting enzyme of E2 synthesis, was a positive functional target of NORFA. Mechanistically, NORFA promotes SF-1 expression by stabilizing NR5A1 mRNA through directly interacting with its 3'-UTR, and also tethers SF-1 to shuttle into nucleus. Additionally, SF-1 in the nucleus activates CYP11A1 transcription by directly binding to its promoter, which ultimately induces E2 synthesis and inhibits GC apoptosis. CONCLUSION Our findings highlight that NORFA, a multifunctional lncRNA, induces E2 synthesis and inhibits GC apoptosis through the SF-1/CYP11A1 axis in a ceRNA-independent manner, which provide valuable clues and potential targets for follicular atresia inhibition and female fertility improvement.
Collapse
Affiliation(s)
- Zhennan Guo
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| | - Qiang Zeng
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| | - Qiqi Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
- College of Animal Husbandry and Veterinary Medicine, Jiangsu Vocational College of Agriculture and Forestry, Zhenjiang, Jiangsu, 212400, China
| | - Baosen Shan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| | - Yangan Huo
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| | - Xiaoli Shi
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
- National Experimental Teaching Demonstration Center for Animal Science, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| | - Qifa Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China
| | - Xing Du
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, China.
| |
Collapse
|
23
|
Chen J, He Z, Xu W, Kang Y, Zhu F, Tang H, Wang J, Zhong F. Human umbilical cord mesenchymal stem cells restore chemotherapy-induced premature ovarian failure by inhibiting ferroptosis in vitro ovarian culture system. Reprod Biol Endocrinol 2024; 22:137. [PMID: 39511578 PMCID: PMC11542367 DOI: 10.1186/s12958-024-01310-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 10/30/2024] [Indexed: 11/15/2024] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) have shown potential in repairing chemotherapy-induced premature ovarian failure (POF). However, challenges such as stem cell loss and immune phagocytosis post-transplantation hinder their application. Due to easy and safe handling, in vitro ovarian culture is widely available for drug screening, pathophysiological research, and in vitro fertilization. MSCs could exhibit therapeutic capacity for ovarian injury, and avoid stem cell loss and immune phagocytosis in vitro tissue culture system. Therefore, this study utilizes an in vitro ovarian culture system to investigate the reparative potential of human umbilical cord mesenchymal stem cells (hUCMSCs) and their mechanism. METHODS In this study, a chemotherapy-induced POF model was established by introducing cisplatin in vitro ovarian culture system. The reparative effects of hUCMSCs on damaged ovarian tissue were validated through Transwell chambers. Tissue histology examination, immunohistochemical staining, Western blotting, and RT-PCR were employed to evaluate the expression effects of hUCMSCs on ferroptosis and fibrosis-related genes during the process of repairing cisplatin-induced POF. RESULTS Cisplatin was found to activate ovarian follicles in vitro POF model. Transcriptomic sequencing analysis revealed that cisplatin could activate genes associated with ferroptosis. hUCMSCs alleviated cisplatin-induced POF by suppressing the expression of ferroptosis. Moreover, inhibiting ferroptosis by hUCMSCs also ameliorated ovarian hormone levels and reduced the expression of fibrosis-related factors α-SMA and COL-I in the ovaries. CONCLUSIONS This study confirms that cisplatin-induced ovarian damage via ferroptosis in vitro POF model, and hUCMSCs repair ovarian injury by inhibiting the ferroptosis pathway and suppressing fibrosis. This research contributes to evaluating the effectiveness of hUCMSCs in treating chemotherapy-induced POF by inhibiting ferroptosis in an in vitro ovarian culture system and provides a potential therapeutic strategy for chemotherapy-induced POF.
Collapse
Affiliation(s)
- Jiaqi Chen
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, 230032, China
| | - Zhuoying He
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, 230032, China
| | - Wenjuan Xu
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, 230032, China
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, Anhui, 230022, China
| | - Yumiao Kang
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, 230032, China
| | - Fengyu Zhu
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, 230032, China
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, Anhui, 230022, China
| | - Heng Tang
- Wanbei Coal Electric Group General Hospital, Suzhou, Anhui Province, 234011, China.
| | - Jianye Wang
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, 230032, China.
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, Anhui, 230022, China.
| | - Fei Zhong
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China.
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, 230032, China.
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, Anhui, 230022, China.
| |
Collapse
|
24
|
Lafraoui I, Heddar A, Cantalloube A, Braham I, Peigné M, Beneteau C, Gricourt S, Poirsier C, Legrand S, Stoeva R, Metayer-Amelot L, Lobersztajn A, Lebrun S, Gruchy N, Abdennebi I, Cedrin-Durnerin I, Fernandez H, Luton D, Torre A, Zagdoun L, Chevalier N, Khrouf M, Mahmoud K, Epelboin S, Catteau-Jonard S, Misrahi M. Genetic Landscape of a Cohort of 120 Patients with Diminished Ovarian Reserve: Correlation with Infertility. Int J Mol Sci 2024; 25:11915. [PMID: 39595984 PMCID: PMC11593603 DOI: 10.3390/ijms252211915] [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: 09/23/2024] [Revised: 10/28/2024] [Accepted: 10/31/2024] [Indexed: 11/28/2024] Open
Abstract
Diminished ovarian reserve (DOR) and primary ovarian insufficiency (POI) are major causes of female infertility. We recently found a monogenic etiology in 29.3% of POI, leading to personalized medicine. The genetic landscape of DOR is unknown. A prospective study (2018-2023) of an international cohort of 120 patients with unexplained DOR was performed using a large custom targeted next-generation sequencing panel including all known POI-causing genes. The diagnostic yield, based on the American College of Medical Genetics, was 24, 2%. Genes belong to different pathways: metabolism and mitochondria (29.7%), follicular growth (24.3%), DNA repair/meiosis (18.9%), aging (16.2%), ovarian development (8.1%), and autophagy (2.7%). Five genes were recurrently found: LMNA, ERCC6, SOX8, POLG, and BMPR1B. Six genes identified in single families with POI were involved in DOR, GNAS, TGFBR3, XPNPEP2, EXO1, BNC1, ATG, highlighting their role in maintaining ovarian reserve. In our cohort, 26 pregnancies were recorded, but no pregnancy was observed when meiosis/DNA repair genes were involved, suggesting severely impaired oocyte quality. Additional studies should confirm these preliminary results. This study with a large NGS panel defines the genetic landscape of a large cohort of DOR. It supports routine genetic diagnosis. Genetics could be a biomarker predicting infertility and progression to POI.
Collapse
Affiliation(s)
- Imène Lafraoui
- Unité de Génétique Moléculaire des Maladies Métaboliques et de la Reproduction, Hôpital Bicêtre, Faculté de Médecine Paris Saclay, INSERM U1193, 94275 Le Kremlin-Bicêtre, France; (I.L.); (A.H.)
- Laboratoire de Biologie Moléculaire National de Référence-LBMR Pour les Infertilités Génétiques Chez la Femme et l’Homme, Hôpitaux Universitaires Paris Saclay, 94275 Le Kremlin Bicêtre, France
| | - Abdelkader Heddar
- Unité de Génétique Moléculaire des Maladies Métaboliques et de la Reproduction, Hôpital Bicêtre, Faculté de Médecine Paris Saclay, INSERM U1193, 94275 Le Kremlin-Bicêtre, France; (I.L.); (A.H.)
- Laboratoire de Biologie Moléculaire National de Référence-LBMR Pour les Infertilités Génétiques Chez la Femme et l’Homme, Hôpitaux Universitaires Paris Saclay, 94275 Le Kremlin Bicêtre, France
| | - Adèle Cantalloube
- Service de Gynécologie-Obstétrique, Hôpital Tenon, Hôpitaux Universitaires Paris Centre, 75014 Paris, France; (A.C.); (S.G.); (S.E.)
| | - Inès Braham
- Service d’Endocrinologie, Diabétologie et Médecine de la Reproduction, CHU de Nice, 06000 Nice, France; (I.B.); (N.C.)
| | - Maëliss Peigné
- Service de Médecine de la Reproduction et Préservation de la Fertilité, Hôpital Jean-Verdier, Université Sorbonne Paris Nord, 93430 Bondy, France; (M.P.); (I.C.-D.)
| | - Claire Beneteau
- Service de Génétique Médicale, CHU de Nantes, 44000 Nantes, France;
| | - Solenne Gricourt
- Service de Gynécologie-Obstétrique, Hôpital Tenon, Hôpitaux Universitaires Paris Centre, 75014 Paris, France; (A.C.); (S.G.); (S.E.)
| | - Claire Poirsier
- Departement de Genetique, Centre Hospitalier Universitaire de Reims, 51092 Reims, France;
| | - Stéphanie Legrand
- Centre de Fertilité, Clinique de l’Atlantique, 17138 La Rochelle, France;
| | - Radka Stoeva
- Laboratoire de Génétique Médicale et Cytogénétique, CH Le Mans, 72037 Le Mans, France;
| | - Laure Metayer-Amelot
- Service d’Endocrinologie et Médecine de la Reproduction, CH Le Mans, 72037 Le Mans, France;
| | - Annina Lobersztajn
- Centre de la Fertilité—Paris Est, Nogent sur Marne, 94130 Nogent-sur-Marne, France;
| | - Soizic Lebrun
- Service de Génétique, FHU GenOMedS, CHRU de Tours, 37000 Tours, France;
| | - Nicolas Gruchy
- EA 7450 BioTARGen, FHU G4 Genomics, Service de Génétique Clinique, Departement de Genetique, CHU Côte de Nacre, Université de Caen Normandie UNICAEN, 14000 Caen, France;
| | - Inès Abdennebi
- Centre d’Aide Médicale à la Procréation, Centre Hospitalier Intercommunal de Créteil, 94000 Créteil, France;
| | - Isabelle Cedrin-Durnerin
- Service de Médecine de la Reproduction et Préservation de la Fertilité, Hôpital Jean-Verdier, Université Sorbonne Paris Nord, 93430 Bondy, France; (M.P.); (I.C.-D.)
| | - Hervé Fernandez
- Service de Gynécologie-Obstétrique, Hôpital Bicêtre, Université Paris Saclay, 94270 Le Kremlin-Bicêtre, France; (H.F.); (D.L.)
| | - Dominique Luton
- Service de Gynécologie-Obstétrique, Hôpital Bicêtre, Université Paris Saclay, 94270 Le Kremlin-Bicêtre, France; (H.F.); (D.L.)
| | - Antoine Torre
- Centre d’Assistance Médicale à la Procréation Clinico-Biologique, Centre Hospitalier Sud Francilien Corbeil-Essonnes, 91100 Corbeil-Essonnes, France;
| | - Léonore Zagdoun
- Service de Diabétologie et Endocrinologie, Centre Hospitalier de Mont de Marsan et Pays des Sources, 40024 Mont de Marsan, France;
| | - Nicolas Chevalier
- Service d’Endocrinologie, Diabétologie et Médecine de la Reproduction, CHU de Nice, 06000 Nice, France; (I.B.); (N.C.)
| | - Mohamed Khrouf
- Centre FERTILLIA de Médecine de la Reproduction-Clinique la Rose, Tunis 1053, Tunisia; (M.K.); (K.M.)
| | - Khaled Mahmoud
- Centre FERTILLIA de Médecine de la Reproduction-Clinique la Rose, Tunis 1053, Tunisia; (M.K.); (K.M.)
| | - Sylvie Epelboin
- Service de Gynécologie-Obstétrique, Hôpital Tenon, Hôpitaux Universitaires Paris Centre, 75014 Paris, France; (A.C.); (S.G.); (S.E.)
| | - Sophie Catteau-Jonard
- Service de Gynécologie Endocrinienne, CHU de Lille, Hôpital Jeanne-de-Flandre, 59000 Lille, France;
| | - Micheline Misrahi
- Unité de Génétique Moléculaire des Maladies Métaboliques et de la Reproduction, Hôpital Bicêtre, Faculté de Médecine Paris Saclay, INSERM U1193, 94275 Le Kremlin-Bicêtre, France; (I.L.); (A.H.)
- Laboratoire de Biologie Moléculaire National de Référence-LBMR Pour les Infertilités Génétiques Chez la Femme et l’Homme, Hôpitaux Universitaires Paris Saclay, 94275 Le Kremlin Bicêtre, France
| |
Collapse
|
25
|
Han S, Yu C, Qiu M, Xiong X, Peng H, Song X, Hu C, Zhang Z, Xia B, Yang L, Chen J, Zhu S, Li W, Yang C. USP13 regulates ferroptosis in chicken follicle granulosa cells by deubiquitinating ATG7. Poult Sci 2024; 103:104209. [PMID: 39214053 PMCID: PMC11402030 DOI: 10.1016/j.psj.2024.104209] [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: 06/27/2024] [Revised: 08/07/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024] Open
Abstract
The development and maturation of follicles are intricately linked to egg production and reproductive performance of chickens. Granulosa cells death directly affects the development and maturation of follicles, thereby impacting the reproductive performance of hens. Ferroptosis is a new type of cell death, it is unknown how it affects the growth and development of chicken follicles. In this study, RNA-seq analysis revealed significant differences in the expression of ferroptosis-related genes between normal follicles and atretic follicles, suggesting a potential role for ferroptosis in follicle growth and development. In addition, we found that ubiquitin-specific protease 13 (USP13) was significantly upregulated in atrophic follicles. Overexpression of USP13 results in depletion of glutathione (GSH), peroxidation of lipids, accumulation of iron, and activation of ferroptosis in chicken granulosa cells. In contrast, USP13 knockdown significantly inhibited ferroptosis events. Mechanistically, USP13 prevents the degradation of autophagy related 7 (ATG7) by deubiquitinating it, thereby enhancing the stability of ATG7 protein and ultimately promoting ferroptosis. In conclusion, this study elucidates the crucial role of the USP13-ATG7 axis in regulating ferroptosis in chicken follicle granulosa cells, thereby presenting a novel avenue for molecular breeding research in chickens.
Collapse
Affiliation(s)
- Shunshun Han
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Chunlin Yu
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Mohan Qiu
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Xia Xiong
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Han Peng
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Xiaoyan Song
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Chenming Hu
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Zengrong Zhang
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Bo Xia
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Li Yang
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Jialei Chen
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Shiliang Zhu
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Wen Li
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Chaowu Yang
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China.
| |
Collapse
|
26
|
Ji R, Wang S, Chen X, Yang Z, Zhang Z, Bao S, Xiao Z, Zhang Y, Yin T, Yang J. Platycodin D ameliorates polycystic ovary syndrome-induced ovarian damage by upregulating CD44 to attenuate ferroptosis. Free Radic Biol Med 2024; 224:707-722. [PMID: 39321891 DOI: 10.1016/j.freeradbiomed.2024.09.033] [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: 07/15/2024] [Revised: 09/10/2024] [Accepted: 09/22/2024] [Indexed: 09/27/2024]
Abstract
Recently, the potential association between polycystic ovary syndrome (PCOS) development and progression and ferroptosis has garnered attention. Increasing evidence suggests that targeting ferroptosis may be an effective strategy for treating PCOS. First, we observed that the expression of the ferroptosis regulatory molecules SLC7A11, GPX4, and FTH1 was decreased in the granulosa cells (GCs) of patients with PCOS and ovarian tissues of rats with PCOS; in contrast, TFR1 expression was increased. This suggests that GC ferroptosis is involved in PCOS pathogenesis. Furthermore, bioinformatics analysis of GC datasets from patients with PCOS and PCOS clinical samples and animal model analysis revealed CD44 as a key molecule regulating ferroptosis in PCOS, which was down-regulated in GCs of PCOS patients and rats. Subsequently, molecular docking was performed to screen existing natural compounds for inhibiting ferroptosis. Dynamic simulation and cellular thermal shift assay identified platycodin D as a natural plant extract for inhibiting ferroptosis by targeting CD44 in GCs. Subsequently, a series of functional experiments revealed that platycodin D ameliorated ovarian damage in rats with PCOS. This was primarily owing to the protective effects achieved by promoting glutathione production, attenuating lipid accumulation and lipid peroxidation in GCs, inhibiting iron overload, and scavenging reactive oxygen species. In addition, western blotting and immunofluorescence staining revealed that platycodin D upregulated the expression of CD44 and SLC7A11 in GCs. Furthermore, by knocking down CD44 and SLC7A11 in vivo and in vitro, respectively, the ameliorative effect of platycodin D on ferroptosis in the GCs of rats with PCOS was reversed. Collectively, these findings suggest that platycodin D attenuates ferroptosis in GCs by activating CD44/SLC7A11 axis, thereby upregulating system Xc-. In conclusion, platycodin D can attenuate ferroptosis in GCs by activating CD44, potentially ameliorating ovarian damage in PCOS.
Collapse
Affiliation(s)
- Rui Ji
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, China; Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, China
| | - Shujun Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xin Chen
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, China; Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, China
| | - Zhe Yang
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, China; Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, China
| | - Zhimo Zhang
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, China; Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, China
| | - Shenglan Bao
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, China; Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, China
| | - Zhuoni Xiao
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, China; Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, China.
| | - Yan Zhang
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, WuHan, China.
| | - Tailang Yin
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, China; Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, China.
| | - Jing Yang
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, China; Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, China.
| |
Collapse
|
27
|
Yang Z, Wei Y, Fu Y, Wang X, Shen W, Shi A, Zhang H, Li H, Song X, Wang J, Jin M, Zheng H, Tao J, Wang Y. Folic acids promote in vitro maturation of bovine oocytes by inhibition of ferroptosis via upregulated glutathione and downregulated Fe 2+ accumulation. Anim Reprod Sci 2024; 270:107605. [PMID: 39362062 DOI: 10.1016/j.anireprosci.2024.107605] [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: 05/10/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 10/05/2024]
Abstract
Bovine embryos by in vitro fertilization have become the primary source of commercial embryo transfers globally. However, the developmental capacity of in vitro maturation (IVM) oocytes is considerably lower than that of in vivo maturation (IVO) oocytes, owing to the production of reactive oxygen species (ROS) via mitochondrial metabolism, which was higher in IVM oocytes than in IVO oocytes. To avoid the negative effects of ROS on embryo quality, folic acid (FA) was supplemented directly into the IVM medium to antagonize ROS production, however, the mechanisms remain unknown. In the present study, five levels of FA (0, 25, 50, 100, and 200 µM) were supplemented into the bovine oocyte culture medium. The maturation, cleavage, and blastocyst formation rates increased by 8.95 %, 6.94 %, and 4.36 %, respectively, in the 50 µM group compared to the 0 µM group. Moreover, 7904 differential genes were identified between 0 µM and 50 µM groups by transcriptome sequencing, and they were mainly enriched in 8 pathways. The glutathione, ROS, and Fe2+ levels in oocytes were found to be associated with ferroptosis. Our results revealed that 50 µM FA promoted the IVM of bovine oocytes and affected the expression of genes involved in the ferroptosis pathway. The downregulation of TFR1 and STEAP3 led to a decrease in intracellular Fe2+ accumulation, and the upregulation of GCL increased oocyte GSH levels, thereby reducing the production of ROS in the ferroptosis pathway. Our study provides a new insight into the molecular mechanisms by which FA promotes bovine oocyte development in vitro.
Collapse
Affiliation(s)
- Zhuo Yang
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China
| | - Yaochang Wei
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Yu Fu
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China
| | - Xiaoyan Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Wenjuan Shen
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China
| | - An Shi
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China
| | - Han Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Heqiang Li
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Xuexiao Song
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Jie Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Mengdong Jin
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Hao Zheng
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China
| | - Jinzhong Tao
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China.
| | - Yongsheng Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China.
| |
Collapse
|
28
|
Hong H, Xiao C, Weng L, Wang Q, Lai D. The effect of norepinephrine on ovarian dysfunction by mediating ferroptosis in mice model. Acta Biochim Biophys Sin (Shanghai) 2024; 57:542-553. [PMID: 39439417 DOI: 10.3724/abbs.2024187] [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] [Indexed: 10/25/2024] Open
Abstract
Studies have shown that stress is associated with ovarian dysfunction. Norepinephrine (NE), a classic stress hormone involved in the stress response, is less recognized for its role in ovarian function. In this study, an NE-treated mouse model is induced by intraperitoneal injection of NE for 4 weeks. Compared with normal control mice, NE-treated mice show disturbances in the estrous cycle, decreased levels of anti-Mullerian hormone (AMH) and estradiol (E2), and increased level of follicle-stimulating hormone (FSH). Additionally, the numbers of primordial follicles, primary follicles, secondary follicles, and antral follicles are decreased, whereas the number of atretic follicles is increased in NE-treated mice, indicating NE-induced ovarian dysfunction. RNA sequencing further reveals that genes associated with ferroptosis are significantly enriched in NE-treated ovarian tissues. Concurrently, the levels of reactive oxygen species (ROS), ferrous ions, and malondialdehyde (MDA) are increased, whereas the expression level of glutathione peroxidase 4 (GPX4) is decreased. To elucidate the mechanism of NE-induced ferroptosis in ovaries and the potential reversal by Coenzyme Q10 (CoQ10), an antioxidant, we conduct both in vitro and in vivo experiments. In vitro, the granulosa cell line KGN, when treated with NE, shows decreased cell viability, reduced expression of GPX4, elevated levels of ferrous ion and ROS, and increased MDA level. However, these NE-induced changes are reversed by the addition of CoQ10. Compared with the NE group, the NE-treated mice supplemented with CoQ10 present increased GPX4 level and decreased iron, ROS, and MDA levels. Moreover, the differential expression of genes associated with ferroptosis induced by NE is ameliorated by CoQ10 in NE-treated mice. Additionally, CoQ10 improves ovarian function, as evidenced by increased ovarian weight, more regular estrous cycles, and an increase in follicles at various stages of growth in NE-treated mice. In conclusion, NE induces ovarian dysfunction by triggering ferroptosis in ovarian tissues, and CoQ10 represents a promising approach for protecting reproductive function by inhibiting ferroptosis.
Collapse
Affiliation(s)
- Hanqing Hong
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai 200030, China
| | - Chengqi Xiao
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai 200030, China
| | - Lichun Weng
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai 200030, China
| | - Qian Wang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai 200030, China
| | - Dongmei Lai
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai 200030, China
| |
Collapse
|
29
|
Liu Y, Zhang W, Wang H, Liu H, Yu Q, Luo X, Feng X, Yang P. Fine particulate matter potentiates Th17-cell pathogenicity in experimental autoimmune uveitis via ferroptosis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 284:116979. [PMID: 39232294 DOI: 10.1016/j.ecoenv.2024.116979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 09/06/2024]
Abstract
The effect of fine particulate matter (PM2.5) on the development of uveitis remains unclear. Therefore, this study was designed to investigate the role of PM2.5 in experimental autoimmune uveitis (EAU) and its potential mechanism. Our results showed that PM2.5 could exacerbate the activity of EAU, as evidenced by severer clinical and pathological changes, correlated with elevated Th17 cells frequency and IL-17A expression. Proteomic analysis revealed ferroptosis was the most significant pathway. In vivo, the levels of Fe2+, ROS, lipid ROS, and malondialdehyde, as well as the expression of TFRC, HMOX1, FTH1, and FTL1 in CD4+ T cells were increased, while GSH/GSSG ratio and the expression of ACSL1 and GPX4 were decreased after PM2.5 exposure. In vitro, the expression of TFRC and HMOX1 were increased, while the expression FTH1, FTL1, ACSL1, and GPX4 were decreased after PM2.5 exposure. Ferrostatin-1 effectively alleviated PM2.5-induced intraocular inflammation and suppressed the frequency of Th17 cells. These results suggest that PM2.5 could aggravate intraocular inflammation and immune response in EAU mice through ferroptosis. Ferroptosis could be a potential marker for the prevention and treatment of uveitis.
Collapse
Affiliation(s)
- Yaning Liu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wanyun Zhang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongmiao Wang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hui Liu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qiuyue Yu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiang Luo
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaojie Feng
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Peizeng Yang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
30
|
Peiyin F, Yuxian W, Jiali Z, Jian X. Research progress of ferroptosis in female infertility. J Ovarian Res 2024; 17:183. [PMID: 39267109 PMCID: PMC11391650 DOI: 10.1186/s13048-024-01508-y] [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: 03/13/2024] [Accepted: 08/30/2024] [Indexed: 09/14/2024] Open
Abstract
Ferroptosis is a novel type of programmed cell death dependent on iron and characterized by the accumulation of lipid peroxides in cells and is closely related to various diseases. Female infertility is a global health concern, which is associated with a variety of factors. The etiology remains unknown in many women with infertility. With further investigation into the pathogenesis of infertility, a growing number of studies have demonstrated the close connections between infertility and ferroptosis. Through a literature review, it is found that ferroptosis is closely involved in endometriosis- and polycystic ovarian syndrome (PCOS)-associated infertility and tubal factor infertility. Iron overload increases the resistance to ferroptosis, and ferroptosis in some cells accelerates endometrial lesion growth. Moreover, iron overload may be hazardous to oocytes. This review may shed some light on the diagnosis and treatment of female infertility.
Collapse
Affiliation(s)
- Fan Peiyin
- Reproductive Medicine Center, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, China
| | - Wang Yuxian
- Reproductive Medicine Center, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, China
| | - Zhang Jiali
- Reproductive Medicine Center, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, China
| | - Xu Jian
- Reproductive Medicine Center, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, China.
| |
Collapse
|
31
|
Ding H, Xiang Y, Zhu Q, Wu H, Xu T, Huang Z, Ge H. Endoplasmic reticulum stress-mediated ferroptosis in granulosa cells contributes to follicular dysfunction of polycystic ovary syndrome driven by hyperandrogenism. Reprod Biomed Online 2024; 49:104078. [PMID: 39024925 DOI: 10.1016/j.rbmo.2024.104078] [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: 01/11/2024] [Revised: 03/28/2024] [Accepted: 04/23/2024] [Indexed: 07/20/2024]
Abstract
RESEARCH QUESTION Does hyperandrogenaemia affect the function of ovarian granulosa cells by activating ferroptosis, and could this process be regulated by endoplasmic reticulum stress? DESIGN Levels of ferroptosis and endoplasmic reticulum stress in granulosa cells were detected in women with and without polycystic ovary syndrome (PCOS) undergoing IVF. Ferroptosis and endoplasmic reticulum stress levels of ovarian tissue and follicle development were detected in control mice and PCOS-like mice models, induced by dehydroepiandrosterone. An in-vitro PCOS model of KGN cells was constructed with testosterone and ferroptosis inhibitor Fer-1. Endoplasmic reticulum stress inhibitor, tauroursodeoxycholate (TUDCA), determined the potential mechanism associated with excessive induction of ferroptosis in granulosa cells related to PCOS, and levels of ferroptosis and endoplasmic reticulum stress were detected. RESULTS Activation of ferroptosis and endoplasmic reticulum stress occurred in granulosa cells of women with PCOS and the varies of PCOS-like mice. The findings in KGN cells demonstrated that testosterone treatment results in elevation of oxidative stress levels, particularly lipid peroxidation, and intracellular iron accumulation in granulosa cells. The expression of genes and proteins associated with factors related to ferroptosis, mitochondrial membrane potential and ultrastructure showed that testosterone activated ferroptosis, whereas Fer-1 reversed these alterations. During in-vitro experiments, activation of endoplasmic reticulum stress induced by testosterone treatment was detected in granulosa cells. In granulosa cells, TUDCA, an inhibitor of endoplasmic reticulum stress, significantly mitigated testosterone-induced ferroptosis. CONCLUSIONS Ferroptosis plays a part in reproductive injury mediated by hyperandrogens associated with PCOS, and may be regulated by endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Huimin Ding
- Department of Obstetrics and Gynecology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China; Graduate School, Nanjing University Of Chinese Medicine, Nanjing, China
| | - Yu Xiang
- Department of Obstetrics and Gynecology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China; Graduate School, Nanjing University Of Chinese Medicine, Nanjing, China
| | - Qi Zhu
- Department of Obstetrics and Gynecology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China; Graduate School, Nanjing Medical University, Nanjing, China
| | - Honghui Wu
- Department of Obstetrics and Gynecology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China; Graduate School, Dalian Medical University, Liaoning, China
| | - Tianyue Xu
- Department of Obstetrics and Gynecology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China; Graduate School, Nanjing University Of Chinese Medicine, Nanjing, China
| | - Zichao Huang
- Department of Obstetrics and Gynecology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China; Graduate School, Nanjing University Of Chinese Medicine, Nanjing, China
| | - Hongshan Ge
- Department of Obstetrics and Gynecology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China; Graduate School, Nanjing University Of Chinese Medicine, Nanjing, China; Graduate School, Nanjing Medical University, Nanjing, China; Graduate School, Dalian Medical University, Liaoning, China.
| |
Collapse
|
32
|
Wang S, Wu X, Yang J, Peng Y, Miao F, Li M, Zeng J. Sterigmatocystin declines mouse oocyte quality by inducing ferroptosis and asymmetric division defects. J Ovarian Res 2024; 17:175. [PMID: 39198920 PMCID: PMC11351269 DOI: 10.1186/s13048-024-01499-w] [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: 03/28/2024] [Accepted: 08/14/2024] [Indexed: 09/01/2024] Open
Abstract
BACKGROUND Sterigmatocystin (STE) is a mycotoxin widely found in contaminated food and foodstuffs, and excessive long-term exposure to STE is associated with several health issues, including infertility. However, there is little information available regarding the effects of STE toxin on the female reproductive system, particularly concerning oocyte maturation. METHODS In the present study, we investigated the toxic effects of STE on mouse oocyte maturation. We also used Western blot, immunofluorescence, and image quantification analyses to assess the impact of STE exposure on the oocyte maturation progression, mitochondrial distribution, oxidative stress, DNA damages, oocyte ferroptosis and asymmetric division defects. RESULTS Our results revealed that STE exposure disrupted mouse oocyte maturation progression. When we examined the cellular changes following 100 µM STE treatment, we found that STE adversely affected polar body extrusion and induced asymmetric division defects in oocytes. RNA-sequencing data showed that STE exposure affects the expression of several pathway-correlated genes during oocyte meiosis in mice, suggesting its toxicity to oocytes. Based on the RNA-seq data, we showed that STE exposure induced oxidative stress and caused DNA damage in oocytes. Besides, ferroptosis and α-tubulin acetylation were also found in STE-exposed oocytes. Moreover, we determined that STE exposure resulted in reduced RAF1 protein expression in mouse oocytes, and inhibition of RAF1 activity also causes defects in asymmetric division of mouse oocytes. CONCLUSIONS Collectively, our research provides novel insights into the molecular mechanisms whereby STE contributes to abnormal meiosis.
Collapse
Affiliation(s)
- Shiwei Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Heifei, Anhui, 230022, China
- College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Xuan Wu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Heifei, Anhui, 230022, China
- College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Juan Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Heifei, Anhui, 230022, China
| | - Yuwan Peng
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Heifei, Anhui, 230022, China
| | - Fulu Miao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Heifei, Anhui, 230022, China
| | - Min Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Heifei, Anhui, 230022, China
| | - Juan Zeng
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Heifei, Anhui, 230022, China.
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, China.
| |
Collapse
|
33
|
Bazzano MV, Köninger A, Solano ME. Beyond defence: Immune architects of ovarian health and disease. Semin Immunopathol 2024; 46:11. [PMID: 39134914 PMCID: PMC11319434 DOI: 10.1007/s00281-024-01021-w] [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: 03/14/2024] [Accepted: 07/23/2024] [Indexed: 08/15/2024]
Abstract
Throughout the individual's reproductive period of life the ovary undergoes continues changes, including cyclic processes of cell death, tissue regeneration, proliferation, and vascularization. Tissue-resident leucocytes particularly macrophages, play a crucial role in shaping ovarian function and maintaining homeostasis. Macrophages crucially promote angiogenesis in the follicles and corpora lutea, thereby supporting steroidogenesis. Recent research on macrophage origins and early tissue seeding has unveiled significant insights into their role in early organogenesis, e.g. in the testis. Here, we review evidence about the prenatal ovarian seeding of leucocytes, primarily macrophages with angiogenic profiles, and its connection to gametogenesis. In the prenatal ovary, germ cells proliferate, form cysts, and undergo changes that, following waves of apoptosis, give rice to the oocytes contained in primordial follicles. These follicles constitute the ovarian reserve that lasts throughout the female's reproductive life. Simultaneously, yolk-sac-derived primitive macrophages colonizing the early ovary are gradually replaced or outnumbered by monocyte-derived fetal macrophages. However, the cues indicating how macrophage colonization and follicle assembly are related are elusive. Macrophages may contribute to organogenesis by promoting early vasculogenesis. Whether macrophages contribute to ovarian lymphangiogenesis or innervation is still unknown. Ovarian organogenesis and gametogenesis are vulnerable to prenatal insults, potentially programming dysfunction in later life, as observed in polycystic ovary syndrome. Experimental and, more sparsely, epidemiological evidence suggest that adverse stimuli during pregnancy can program defective folliculogenesis or a diminished follicle reserve in the offspring. While the ovary is highly sensitive to inflammation, the involvement of local immune responses in programming ovarian health and disease remains to be thoroughly investigated.
Collapse
Affiliation(s)
- Maria Victoria Bazzano
- Laboratory of Translational Perinatology, University of Regensburg, Biopark 1-3, D-93053, Regensburg, Germany
| | - Angela Köninger
- University Department of Obstetrics and Gynecology, Clinic St. Hedwig of The Order of St. John, University of Regensburg, Steinmetzstr. 1-3, D-93049, Regensburg, Germany
| | - Maria Emilia Solano
- Laboratory of Translational Perinatology, University of Regensburg, Biopark 1-3, D-93053, Regensburg, Germany.
| |
Collapse
|
34
|
Wang M, Zhang BQ, Ma S, Xu Y, Zhao DH, Zhang JS, Li CJ, Zhou X, Zheng LW. Broadening horizons: the role of ferroptosis in polycystic ovary syndrome. Front Endocrinol (Lausanne) 2024; 15:1390013. [PMID: 39157678 PMCID: PMC11327064 DOI: 10.3389/fendo.2024.1390013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 07/22/2024] [Indexed: 08/20/2024] Open
Abstract
Polycystic ovarian syndrome (PCOS) is a common heterogeneous reproductive endocrine metabolic disorder in women of reproductive age characterized by clinical and biochemical hyperandrogenemia, ovulation disorders, and polycystic ovarian morphology. Ferroptosis is a novel type of cell death driven by iron accumulation and lipid peroxidation. Ferroptosis plays a role in maintaining redox balance, iron metabolism, lipid metabolism, amino acid metabolism, mitochondrial activity, and many other signaling pathways linked to diseases. Iron overload is closely related to insulin resistance, decreased glucose tolerance, and the occurrence of diabetes mellitus. There is limited research on the role of ferroptosis in PCOS. Patients with PCOS have elevated levels of ferritin and increased reactive oxygen species in ovarian GCs. Studying ferroptosis in PCOS patients is highly important for achieving personalized treatment. This article reviews the progress of research on ferroptosis in PCOS, introduces the potential connections between iron metabolism abnormalities and oxidative stress-mediated PCOS, and provides a theoretical basis for diagnosing and treating PCOS.
Collapse
Affiliation(s)
- Min Wang
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Bo-Qi Zhang
- College of Animal Sciences, Jilin University, Changchun, China
| | - Shuai Ma
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Ying Xu
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Dong-Hai Zhao
- Department of Pathology, Jilin Medical College, Jilin, China
| | - Jing-Shun Zhang
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Chun-Jin Li
- College of Animal Sciences, Jilin University, Changchun, China
| | - Xu Zhou
- College of Animal Sciences, Jilin University, Changchun, China
| | - Lian-Wen Zheng
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
35
|
Dai W, Xu B, Ding L, Zhang Z, Yang H, He T, Liu L, Pei X, Fu X. Human umbilical cord mesenchymal stem cells alleviate chemotherapy-induced premature ovarian insufficiency mouse model by suppressing ferritinophagy-mediated ferroptosis in granulosa cells. Free Radic Biol Med 2024; 220:1-14. [PMID: 38677487 DOI: 10.1016/j.freeradbiomed.2024.04.229] [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: 02/09/2024] [Revised: 04/08/2024] [Accepted: 04/16/2024] [Indexed: 04/29/2024]
Abstract
Primary ovarian insufficiency (POI) in younger women (under 40) manifests as irregular periods, high follicle-stimulating hormone (FSH), and low estradiol (E2), often triggered by chemotherapy. Though mesenchymal stem cell (MSC) therapy shows promise in treating POI, its exact mechanism remains unclear. This study reveals that human umbilical cord-derived MSCs (hUC-MSCs) can protect ovarian granulosa cells (GCs) from cyclophosphamide (CTX)-induced ferroptosis, a form of cell death driven by iron accumulation. CTX, commonly used to induce POI animal model, triggered ferroptosis in GCs, while hUC-MSCs treatment mitigated this effect, both in vivo and in vitro. Further investigations using ferroptosis and autophagy inhibitors suggest that hUC-MSCs act by suppressing ferroptosis in GCs. Interestingly, hUC-MSCs activate a protective antioxidant pathway in GCs via NRF2, a stress-response regulator. Overall, our findings suggest that hUC-MSCs improve ovarian function in CTX-induced POI by reducing ferroptosis in GCs. This study not only clarifies the mechanism behind the benefits of hUC-MSCs but also strengthens the case for their clinical use in treating POI. Additionally, it opens up a new avenue for protecting ovaries from chemotherapy-induced damage by regulating ferroptosis.
Collapse
Affiliation(s)
- Wenjie Dai
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
| | - Bo Xu
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
| | - Liyang Ding
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
| | - Zhen Zhang
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
| | - Hong Yang
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
| | - Tiantian He
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
| | - Ling Liu
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
| | - Xiuying Pei
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China.
| | - Xufeng Fu
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China.
| |
Collapse
|
36
|
Ye X, Lin Y, Ying Y, Shen X, Ni F, Wang F, Chen J, Zhao W, Yu X, Zhang D, Liu Y. Human Amniotic Epithelial Stem Cells Alleviate Autoimmune Premature Ovarian Insufficiency in Mice by Targeting Granulosa Cells via AKT/ERK Pathways. Stem Cell Rev Rep 2024; 20:1618-1635. [PMID: 38831179 PMCID: PMC11319531 DOI: 10.1007/s12015-024-10745-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] [Accepted: 05/29/2024] [Indexed: 06/05/2024]
Abstract
Autoimmune factors play an important role in premature ovarian insufficiency (POI). Human amniotic epithelial stem cells (hAESCs) have recently shown promising treatment effects on chemotherapy-induced POI. However, the therapeutic efficacy and underlying mechanisms of hAESCs in autoimmune POI remain to be investigated. In this study, we showed for the first time that intravenous transplantation of hAESCs could reside in the ovary of zona pellucida 3 peptide (pZP3) induced autoimmune POI mice model for at least 4 weeks. hAESCs could improve ovarian function and fertility, alleviate inflammation and reduce apoptosis of granulosa cells (GCs) in autoimmune POI mice. The transcriptome analysis of mice ovaries and in vitro co-cultivation experiments suggest that activation of the AKT and ERK pathways may be the key mechanism in the therapeutic effect of hAESCs. Our work provides the theoretical and experimental foundation for optimizing the administration of hAESCs, as well as the clinical application of hAESCs in autoimmune POI patients.
Collapse
Affiliation(s)
- Xiaohang Ye
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Zhejiang, 310006, China
| | - Yifeng Lin
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Zhejiang, 310006, China
| | - Yanyun Ying
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Zhejiang, 310006, China
| | - Xuezhi Shen
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Zhejiang, 310006, China
| | - Feida Ni
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Zhejiang, 310006, China
| | - Feixia Wang
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Zhejiang, 310006, China
| | - Jianpeng Chen
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Zhejiang, 310006, China
| | - Wei Zhao
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Zhejiang, 310006, China
| | - Xiaoming Yu
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Zhejiang, 310006, China
| | - Dan Zhang
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Zhejiang, 310006, China.
- Zhejiang Provincial Clinical Research Center for Child Health, Hangzhou, 310006, China.
| | - Yifeng Liu
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Zhejiang, 310006, China.
- Zhejiang Provincial Clinical Research Center for Child Health, Hangzhou, 310006, China.
| |
Collapse
|
37
|
Hu L, Hong T, He Y, Wang H, Cao J, Pu D, Gao L, Gao C, Cui Y, Wu J, Tan R. Chromosome Segregation-1-like Gene Participates in Ferroptosis in Human Ovarian Granulosa Cells via Nucleocytoplasmic Transport. Antioxidants (Basel) 2024; 13:911. [PMID: 39199156 PMCID: PMC11352033 DOI: 10.3390/antiox13080911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/22/2024] [Accepted: 07/26/2024] [Indexed: 09/01/2024] Open
Abstract
Premature ovarian insufficiency (POI) is defined as the depletion of ovarian function before the age of 40 years. The global prevalence of POI is 3.5%. To date, genetic factors account for 23.5% of the etiology of POI. Herein, a previously uncharacterized pathogenic homozygous variant of the chromosome segregation-1-like gene (CSE1L) was identified in POI patients via targeted panel sequencing. It is reported that dysregulated iron metabolism is involved in many reproductive endocrine disorders; however, its precise role in POI remains obscure. In this study, we identified CSE1L as a potential candidate gene that plays an important role in maintaining iron homeostasis. Deficiency of CSE1L led to ferroptosis in human granulosa cells, which was confirmed by transmission electron microscopy. Mechanistically, coimmunoprecipitation identified the direct interaction between CSE1L and FoxO1. Inhibition of CSE1L led to the excessive accumulation of FoxO1 in the nucleus via nucleocytoplasmic transport. Then, FoxO1 bound to the promoter region of NCOA4 and promoted its transcription, which was verified by a chromatin immunoprecipitation assay. Moreover, inhibition of CSE1L in cumulus cell monolayer could impede oocyte maturation, which might be associated with oxidative stress. Consequently, our study first revealed that CSE1L participated in ferroptosis in human ovarian granulosa cells via nucleocytoplasmic transportation, which might be helpful in revealing the molecular mechanism of CSE1L in the development of POI. Importantly, these findings might provide new insights into the application of ferroptosis inhibitors in the treatment of POI.
Collapse
Affiliation(s)
- Luanqian Hu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, China; (L.H.); (T.H.); (Y.H.); (H.W.); (J.C.); (D.P.); (L.G.); (C.G.); (Y.C.)
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 210000, China
| | - Tongtong Hong
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, China; (L.H.); (T.H.); (Y.H.); (H.W.); (J.C.); (D.P.); (L.G.); (C.G.); (Y.C.)
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 210000, China
| | - Yuheng He
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, China; (L.H.); (T.H.); (Y.H.); (H.W.); (J.C.); (D.P.); (L.G.); (C.G.); (Y.C.)
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 210000, China
| | - Huiyuan Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, China; (L.H.); (T.H.); (Y.H.); (H.W.); (J.C.); (D.P.); (L.G.); (C.G.); (Y.C.)
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 210000, China
| | - Jinxiang Cao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, China; (L.H.); (T.H.); (Y.H.); (H.W.); (J.C.); (D.P.); (L.G.); (C.G.); (Y.C.)
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 210000, China
| | - Danhua Pu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, China; (L.H.); (T.H.); (Y.H.); (H.W.); (J.C.); (D.P.); (L.G.); (C.G.); (Y.C.)
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 210000, China
| | - Li Gao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, China; (L.H.); (T.H.); (Y.H.); (H.W.); (J.C.); (D.P.); (L.G.); (C.G.); (Y.C.)
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 210000, China
| | - Chao Gao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, China; (L.H.); (T.H.); (Y.H.); (H.W.); (J.C.); (D.P.); (L.G.); (C.G.); (Y.C.)
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 210000, China
| | - Yugui Cui
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, China; (L.H.); (T.H.); (Y.H.); (H.W.); (J.C.); (D.P.); (L.G.); (C.G.); (Y.C.)
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 210000, China
| | - Jie Wu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, China; (L.H.); (T.H.); (Y.H.); (H.W.); (J.C.); (D.P.); (L.G.); (C.G.); (Y.C.)
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 210000, China
| | - Rongrong Tan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, China; (L.H.); (T.H.); (Y.H.); (H.W.); (J.C.); (D.P.); (L.G.); (C.G.); (Y.C.)
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing 210000, China
| |
Collapse
|
38
|
Li F, Guo L, Zhou M, Han L, Wu S, Wu L, Yang J. Cryptochrome 2 Suppresses Epithelial-Mesenchymal Transition by Promoting Trophoblastic Ferroptosis in Unexplained Recurrent Spontaneous Abortion. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:1197-1217. [PMID: 38537935 DOI: 10.1016/j.ajpath.2024.02.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 02/02/2024] [Accepted: 02/21/2024] [Indexed: 04/10/2024]
Abstract
Unexplained recurrent spontaneous abortion (URSA) is a serious reproductive issue that affects women of childbearing age. Studies have shown a close association between disrupted circadian rhythm and impaired epithelial-mesenchymal transition (EMT) in trophoblasts during URSA, although the underlying mechanism is not known. The current study investigated the regulatory relationship between circadian rhythm gene cryptochrome 2 (CRY2) and ferroptosis on the migratory ability of trophoblast cells. Cell proliferation experiments, wound-healing assays, and expression of related markers were conducted to study EMT. Trophoblastic ferroptosis was confirmed by the expressions of malondialdehyde, glutathione, mitochondrial membrane potential, divalent iron ions, and related genes. The results showed significant increased expression of CRY2 and decreased expression of brain and muscle aryl hydrocarbon receptor nuclear translocator-like protein 1 (BMAL1) in the URSA villous tissues, accompanied by iron-dependent oxidative changes and abnormal expression of ferroptosis-related proteins. CRY2 and BMAL1 were co-localized and functioned as a feedback loop, which regulated the dynamic changes of EMT-related markers in trophoblast cells. CRY2 promoted trophoblastic ferroptosis, whereas BMAL1 had the opposite effect. Particularly, the ferroptosis inhibitor (ferrostatin-1) effectively reversed the trophoblastic ferroptosis and EMT inhibition caused by CRY2 overexpression. Collectively, these results suggest that CRY2 regulates trophoblastic ferroptosis and hinders cellular EMT and migratory ability by suppressing BMAL1 expression.
Collapse
Affiliation(s)
- Faminzi Li
- Reproductive Medicine Center, Renmin Hospital of Wuhan University and Hubei Clinical Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, China
| | - Liantao Guo
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Mengqi Zhou
- Reproductive Medicine Center, Renmin Hospital of Wuhan University and Hubei Clinical Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, China
| | - Lu Han
- Reproductive Medicine Center, Renmin Hospital of Wuhan University and Hubei Clinical Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, China
| | - Shujuan Wu
- Reproductive Medicine Center, Renmin Hospital of Wuhan University and Hubei Clinical Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, China
| | - Lianzhi Wu
- Department of Obstetrics, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Jing Yang
- Reproductive Medicine Center, Renmin Hospital of Wuhan University and Hubei Clinical Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, China.
| |
Collapse
|
39
|
Zhu X, Li W, Lu M, Shang J, Zhou J, Lin L, Liu Y, Xing J, Zhang M, Zhao S, Lu J, Shi X. M 6A demethylase FTO-stabilized exosomal circBRCA1 alleviates oxidative stress-induced granulosa cell damage via the miR-642a-5p/FOXO1 axis. J Nanobiotechnology 2024; 22:367. [PMID: 38918838 PMCID: PMC11197183 DOI: 10.1186/s12951-024-02583-5] [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: 01/24/2024] [Accepted: 05/24/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND Premature ovarian insufficiency (POI) is an important cause of female infertility and seriously impacts the physical and psychological health of patients. Human umbilical cord mesenchymal stem cell-derived exosomes (HucMSCs-Exs, H-Exs) have exhibited protective effects on ovarian function with unclear mechanisms. METHODS A comprehensive analysis of the Gene Expression Omnibus (GEO) database were used to identify POI-associated circRNAs and miRNAs. The relationship between HucMSC-derived exosomal circBRCA1/miR-642a-5p/FOXO1 axis and POI was examined by RT-qPCR, Western blotting, reactive oxygen species (ROS) staining, senescence-associated β-gal (SA-β-gal) staining, JC-1 staining, TEM, oxygen consumption rate (OCR) measurements and ATP assay in vivo and in vitro. RT-qPCR detected the expression of circBRCA1 in GCs and serum of patients with normal ovarian reserve function (n = 50) and patients with POI (n = 50); then, the correlation of circBRCA1 with ovarian reserve function indexes was analyzed. RESULTS Herein, we found that circBRCA1 was decreased in the serum and ovarian granulosa cells (GCs) of patients with POI and was associated with decreased ovarian reserve. H-Exs improved the disorder of the estrous cycles and reproductive hormone levels, reduced the number of atretic follicles, and alleviated the apoptosis and senescence of GCs in rats with POI. Moreover, H-Exs mitigated mitochondrial damage and reversed the reduced circBRCA1 expression induced by oxidative stress in GCs. Mechanistically, FTO served as an eraser to increase the stability and expression of circBRCA1 by mediating the m6A demethylation of circBRCA1, and exosomal circBRCA1 sponged miR-642a-5p to block its interaction with FOXO1. CircBRCA1 insufficiency aggravated mitochondrial dysfunction, mimicking FTO or FOXO1 depletion effects, which was counteracted by miR-642a-5p inhibition. CONCLUSION H-Exs secreted circBRCA1 regulated by m6A modification, directly sponged miR-642a-5p to upregulate FOXO1, resisted oxidative stress injuries in GCs and protected ovarian function in rats with POI. Exosomal circBRCA1 supplementation may be a general prospect for the prevention and treatment of POI.
Collapse
Affiliation(s)
- Xiaolan Zhu
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20 Zhengdong Road, Zhenjiang, 212001, Jiangsu, China.
- Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, China.
- Reproductive Sciences Institute, Jiangsu University, Zhenjiang, China.
| | - Wenxin Li
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20 Zhengdong Road, Zhenjiang, 212001, Jiangsu, China
- Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Minjun Lu
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20 Zhengdong Road, Zhenjiang, 212001, Jiangsu, China
- Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Junyu Shang
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20 Zhengdong Road, Zhenjiang, 212001, Jiangsu, China
- Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Jiamin Zhou
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20 Zhengdong Road, Zhenjiang, 212001, Jiangsu, China
- Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Li Lin
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20 Zhengdong Road, Zhenjiang, 212001, Jiangsu, China
- Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yueqin Liu
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20 Zhengdong Road, Zhenjiang, 212001, Jiangsu, China
- Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Jie Xing
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20 Zhengdong Road, Zhenjiang, 212001, Jiangsu, China
- Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Mengxue Zhang
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20 Zhengdong Road, Zhenjiang, 212001, Jiangsu, China
- Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Shijie Zhao
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20 Zhengdong Road, Zhenjiang, 212001, Jiangsu, China
- Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Jingjing Lu
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20 Zhengdong Road, Zhenjiang, 212001, Jiangsu, China
- Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xuyan Shi
- Reproductive Medicine Center, The Fourth Affiliated Hospital of Jiangsu University, No. 20 Zhengdong Road, Zhenjiang, 212001, Jiangsu, China
- Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
40
|
Chen H, Nie P, Li J, Wu Y, Yao B, Yang Y, Lash GE, Li P. Cyclophosphamide induces ovarian granulosa cell ferroptosis via a mechanism associated with HO-1 and ROS-mediated mitochondrial dysfunction. J Ovarian Res 2024; 17:107. [PMID: 38762721 PMCID: PMC11102268 DOI: 10.1186/s13048-024-01434-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: 09/04/2023] [Accepted: 05/07/2024] [Indexed: 05/20/2024] Open
Abstract
Abnormal granulosa cell (GC) death contributes to cyclophosphamide (CTX) induced primary ovarian insufficiency (POI). To investigate the contribution of GCs to POI, gene profiles of GCs exposed to CTX were assessed using RNA-Seq and bioinformatics analysis. The results showed the differentially expressed genes (DEGs) were enriched in the ferroptosis-related pathway, which is correlated with upregulated heme oxygenase 1 (HO-1) and downregulated glutathione peroxidase-4 (GPX4). Using CTX-induced cell culture (COV434 and KGN cells), the levels of iron, reactive oxygen species (ROS), lipid peroxide, mitochondrial superoxide, mitochondrial morphology and mitochondrial membrane potential (MMP) were detected by DCFDA, MitoSOX, C11-BODIPY, MitoTracker, Nonylacridine Orange (NAO), JC-1 and transmission electron microscopy respectively. The results showed iron overload and disrupted ROS, including cytoROS, mtROS and lipROS homeostasis, were associated with upregulation of HO-1 and could induce ferroptosis via mitochondrial dysfunction in CTX-induced GCs. Moreover, HO-1 inhibition could suppress ferroptosis induced GPX4 depletion. This implies a role for ROS in CTX-induced ferroptosis and highlights the effect of HO-1 modulators in improving CTX-induced ovarian damage, which may provide a theoretical basis for preventing or restoring GC and ovarian function in patients with POI.
Collapse
Affiliation(s)
- Hui Chen
- Department of Pathology, Jinan University School of Medicine, Guangzhou, 510632, China
| | - Ping Nie
- Department of Pathology, Jinan University School of Medicine, Guangzhou, 510632, China
| | - Jingling Li
- Department of Pathology, Jinan University School of Medicine, Guangzhou, 510632, China
| | - Yongqi Wu
- Department of Pathology, Jinan University School of Medicine, Guangzhou, 510632, China
| | - Bo Yao
- Department of Pathology, Jinan University School of Medicine, Guangzhou, 510632, China
| | - Yabing Yang
- Guangdong Second Provincial General Hospital, Postdoctoral Research Station of Basic Medicine, Jinan University School of Medicine, Guangzhou, 510317, China
| | - Gendie E Lash
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Ping Li
- Department of Pathology, Jinan University School of Medicine, Guangzhou, 510632, China.
| |
Collapse
|
41
|
Pang Q, Tang Z, Luo L. The crosstalk between oncogenic signaling and ferroptosis in cancer. Crit Rev Oncol Hematol 2024; 197:104349. [PMID: 38626848 DOI: 10.1016/j.critrevonc.2024.104349] [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: 09/30/2023] [Revised: 03/13/2024] [Accepted: 04/10/2024] [Indexed: 04/19/2024] Open
Abstract
Ferroptosis, a novel form of cell death regulation, was identified in 2012. It is characterized by unique features that differentiate it from other types of cell death, including necrosis, apoptosis, autophagy, and pyroptosis. Ferroptosis is defined by an abundance of iron ions and lipid peroxidation, resulting in alterations in subcellular structures, an elevation in reactive oxygen species (ROS), a reduction in glutathione (GSH) levels, and an augmentation in Fe (II) cytokines. Ferroptosis, a regulated process, is controlled by an intricate network of signaling pathways, where multiple stimuli can either enhance or hinder the process. This review primarily examines the defensive mechanisms of ferroptosis and its interaction with the tumor microenvironment. The analysis focuses on the pathways that involve AMPK, p53, NF2, mTOR, System Xc-, Wnt, Hippo, Nrf2, and cGAS-STING. The text discusses the possibilities of employing a combination therapy that targets several pathways for the treatment of cancer. It emphasizes the necessity for additional study in this field.
Collapse
Affiliation(s)
- Qianghu Pang
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong 524023, China
| | - Zhirou Tang
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong 524023, China
| | - Lianxiang Luo
- The Marine Biomedical Research Institute of Guangdong Zhanjiang,School of Ocean and Tropical Medicine. Guangdong Medical University, Zhanjiang, Guangdong 524023, China.
| |
Collapse
|
42
|
Wang W, Zhou X, Kong L, Pan Z, Chen G. BUB1 Promotes Gemcitabine Resistance in Pancreatic Cancer Cells by Inhibiting Ferroptosis. Cancers (Basel) 2024; 16:1540. [PMID: 38672622 PMCID: PMC11048608 DOI: 10.3390/cancers16081540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 03/31/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
The development of chemotherapy resistance severely limits the therapeutic efficacy of gemcitabine (GEM) in pancreatic cancer (PC), and the dysregulation of ferroptosis is a crucial factor in the development of chemotherapy resistance. BUB1 Mitotic Checkpoint Serine/Threonine Kinase (BUB1) is highly overexpressed in PC patients and is closely associated with patient prognosis. However, none of the literature reports the connection between BUB1 and ferroptosis. The molecular mechanisms underlying GEM resistance are also not well understood. Therefore, this study first established the high expression levels of BUB1 in PC patients, then explored the role of BUB1 in the process of ferroptosis, and finally investigated the mechanisms by which BUB1 regulates ferroptosis and contributes to GEM resistance in PC cells. In this study, downregulation of BUB1 enhanced the sensitivity of PC cells to Erastin, and inhibited cell proliferation and migration. Mechanistically, BUB1 could inhibit the expression levels of Neurofibromin 2 (NF2) and MOB kinase activator 1 (MOB1), and promote Yes-associated protein (YAP) expression, thereby inhibiting ferroptosis and promoting GEM resistance in PC cells. Furthermore, the combination of BUB1 inhibition with GEM exhibited a synergistic therapeutic effect. These findings reveal the mechanisms underlying the development of GEM chemotherapy resistance based on ferroptosis and suggest that the combined use of BUB1 inhibitors may be an effective approach to enhance GEM efficacy.
Collapse
Affiliation(s)
- Weiming Wang
- Department of Hepato-Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; (W.W.); (L.K.); (Z.P.)
| | - Xiang Zhou
- Department of Breast Cancer, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China;
| | - Lingming Kong
- Department of Hepato-Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; (W.W.); (L.K.); (Z.P.)
| | - Zhenyan Pan
- Department of Hepato-Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; (W.W.); (L.K.); (Z.P.)
| | - Gang Chen
- Department of Hepato-Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; (W.W.); (L.K.); (Z.P.)
| |
Collapse
|
43
|
Zhou Y, Huang J, Zeng L, Yang Q, Bai F, Mai Q, Deng K. Human mesenchymal stem cells derived exosomes improve ovarian function in chemotherapy-induced premature ovarian insufficiency mice by inhibiting ferroptosis through Nrf2/GPX4 pathway. J Ovarian Res 2024; 17:80. [PMID: 38622725 PMCID: PMC11017636 DOI: 10.1186/s13048-024-01403-6] [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: 12/26/2023] [Accepted: 03/30/2024] [Indexed: 04/17/2024] Open
Abstract
BACKGROUND Chemotherapy exposure has become a main cause of premature ovarian insufficiency (POI). This study aimed to evaluate the role and molecular mechanism of human umbilical cord mesenchymal stem cell-derived exosomes (hUMSC-Exos) in ovarian function protection after chemotherapy. METHODS hUMSC-Exos were applied to cyclophosphamide-induced premature ovarian insufficiency mice and human ovarian granulosa tumor cells (KGN) to determine their effects on follicular development and granulosa cell apoptosis. Evaluation was done for iron ion and reactive oxygen species (ROS) production, lipid peroxidation levels, and changes in iron death-related molecules (nuclear factor (erythroid-derived 2)-like 2 (Nrf2), Glutathione Peroxidase enzyme 4 (GPX4), and Solute carrier family 7 member 11 cystine glutamate transporter (SLC7A11; xCT)). Furthermore, rescue experiments using an Nrf2 inhibitor were performed to assess the therapeutic effects of hUMSC-Exos on granulosa cells. RESULTS hUMSC-Exos promoted ovarian hormone levels and primary follicle development in POI mice and reduced granulosa cell apoptosis. After hUMSC-Exos treatment, the ROS production, free iron ions and lipid peroxidation levels of granulosa cells decreased, and the iron death marker proteins Nrf2, xCT and GPX4 also decreased. Furthermore, the Nrf2 inhibitor ML385 significantly attenuated the effects of hUMSC-Exos on granulosa cells. CONCLUSION hUMSC-Exos inhibit ferroptosis and protect against CTX-induced ovarian damage and granulosa cell apoptosis through the Nrf2/GPX4 signaling pathway, revealing a novel mechanism of hUMSC-Exos in POI therapy.
Collapse
Affiliation(s)
- Yuan Zhou
- Department of Gynecology, Shunde Hospital, Southern Medical University, Foshan, Guangdong, 528308, China
| | - Jinfa Huang
- Department of Gynecology, Shunde Hospital, Southern Medical University, Foshan, Guangdong, 528308, China
| | - Lingling Zeng
- Department of Gynecology, Shunde Hospital, Southern Medical University, Foshan, Guangdong, 528308, China
| | - Qian Yang
- Department of Gynecology, Shunde Hospital, Southern Medical University, Foshan, Guangdong, 528308, China
| | - Fangjuan Bai
- Department of Gynecology, Shunde Hospital, Southern Medical University, Foshan, Guangdong, 528308, China
| | - Qiqing Mai
- Department of Gynecology, Shunde Hospital, Southern Medical University, Foshan, Guangdong, 528308, China
| | - Kaixian Deng
- Department of Gynecology, Shunde Hospital, Southern Medical University, Foshan, Guangdong, 528308, China.
| |
Collapse
|
44
|
Pan R, Wang R, Cheng F, Wang L, Cui Z, She J, Yang X. Endometrial stem cells alleviate cisplatin-induced ferroptosis of granulosa cells by regulating Nrf2 expression. Reprod Biol Endocrinol 2024; 22:41. [PMID: 38605340 PMCID: PMC11008046 DOI: 10.1186/s12958-024-01208-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 03/19/2024] [Indexed: 04/13/2024] Open
Abstract
BACKGROUND Premature ovarian failure (POF) caused by cisplatin is a severe and intractable sequela for young women with cancer who received chemotherapy. Cisplatin causes the dysfunction of granulosa cells and mainly leads to but is not limited to its apoptosis and autophagy. Ferroptosis has been also reported to participate, while little is known about it. Our previous experiment has demonstrated that endometrial stem cells (EnSCs) can repair cisplatin-injured granulosa cells. However, it is still unclear whether EnSCs can play a repair role by acting on ferroptosis. METHODS Western blotting and quantitative reverse-transcription polymerase chain reaction (qRT-PCR) were applied to detect the expression levels of ferroptosis-related genes. CCK-8 and 5-Ethynyl-2'-deoxyuridine (EdU) assays were used to evaluate cell viability. Transmission electron microscopy (TEM) was performed to detect ferroptosis in morphology. And the extent of ferroptosis was assessed by ROS, GPx, GSSG and MDA indicators. In vivo, ovarian morphology was presented by HE staining and the protein expression in ovarian tissue was detected by immunohistochemistry. RESULTS Our results showed that ferroptosis could occur in cisplatin-injured granulosa cells. Ferroptosis inhibitor ferrostatin-1 (Fer-1) and EnSCs partly restored cell viability and mitigated the damage of cisplatin to granulosa cells by inhibiting ferroptosis. Moreover, the repair potential of EnSCs can be markedly blocked by ML385. CONCLUSION Our study demonstrated that cisplatin could induce ferroptosis in granulosa cells, while EnSCs could inhibit ferroptosis and thus exert repair effects on the cisplatin-induced injury model both in vivo and in vitro. Meanwhile, Nrf2 was validated to participate in this regulatory process and played an essential role.
Collapse
Affiliation(s)
- Rumeng Pan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, China
| | - Rongli Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, China
| | - Feiyan Cheng
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, China
| | - Lihui Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, China
| | - Zhiwei Cui
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, China
| | - Jing She
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, China
| | - Xinyuan Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, China.
| |
Collapse
|
45
|
Zhang G, Wang Q, Jiang B, Yao L, Wu W, Zhang X, Wan D, Gu Y. Progress of medicinal plants and their active metabolites in ischemia-reperfusion injury of stroke: a novel therapeutic strategy based on regulation of crosstalk between mitophagy and ferroptosis. Front Pharmacol 2024; 15:1374445. [PMID: 38650626 PMCID: PMC11033413 DOI: 10.3389/fphar.2024.1374445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 03/25/2024] [Indexed: 04/25/2024] Open
Abstract
The death of cells can occur through various pathways, including apoptosis, necroptosis, mitophagy, pyroptosis, endoplasmic reticulum stress, oxidative stress, ferroptosis, cuproptosis, and disulfide-driven necrosis. Increasing evidence suggests that mitophagy and ferroptosis play crucial regulatory roles in the development of stroke. In recent years, the incidence of stroke has been gradually increasing, posing a significant threat to human health. Hemorrhagic stroke accounts for only 15% of all strokes, while ischemic stroke is the predominant type, representing 85% of all stroke cases. Ischemic stroke refers to a clinical syndrome characterized by local ischemic-hypoxic necrosis of brain tissue due to various cerebrovascular disorders, leading to rapid onset of corresponding neurological deficits. Currently, specific therapeutic approaches targeting the pathophysiological mechanisms of ischemic brain tissue injury mainly include intravenous thrombolysis and endovascular intervention. Despite some clinical efficacy, these approaches inevitably lead to ischemia-reperfusion injury. Therefore, exploration of treatment options for ischemic stroke remains a challenging task. In light of this background, advancements in targeted therapy for cerebrovascular diseases through mitophagy and ferroptosis offer a new direction for the treatment of such diseases. In this review, we summarize the progress of mitophagy and ferroptosis in regulating ischemia-reperfusion injury in stroke and emphasize their potential molecular mechanisms in the pathogenesis. Importantly, we systematically elucidate the role of medicinal plants and their active metabolites in targeting mitophagy and ferroptosis in ischemia-reperfusion injury in stroke, providing new insights and perspectives for the clinical development of therapeutic drugs for these diseases.
Collapse
Affiliation(s)
- Guozhen Zhang
- College of the First Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China
- Department of Neurology, People’s Liberation Army Joint Logistics Support Force 940th Hospital, Lanzhou, Gansu, China
| | - Qiang Wang
- Department of Neurology, First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Bing Jiang
- Department of Integrated Chinese and Western Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu, China
| | - Lihe Yao
- Department of Neurology, First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Wenjuan Wu
- Department of Neurology, First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Xiaoyan Zhang
- Department of Neurology, People’s Liberation Army Joint Logistics Support Force 940th Hospital, Lanzhou, Gansu, China
| | - Dongjun Wan
- Department of Neurology, People’s Liberation Army Joint Logistics Support Force 940th Hospital, Lanzhou, Gansu, China
| | - Youquan Gu
- College of the First Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China
- Department of Neurology, First Hospital of Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
46
|
Wu M, Tang W, Chen Y, Xue L, Dai J, Li Y, Zhu X, Wu C, Xiong J, Zhang J, Wu T, Zhou S, Chen D, Sun C, Yu J, Li H, Guo Y, Huang Y, Zhu Q, Wei S, Zhou Z, Wu M, Li Y, Xiang T, Qiao H, Wang S. Spatiotemporal transcriptomic changes of human ovarian aging and the regulatory role of FOXP1. NATURE AGING 2024; 4:527-545. [PMID: 38594460 PMCID: PMC11031396 DOI: 10.1038/s43587-024-00607-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 03/05/2024] [Indexed: 04/11/2024]
Abstract
Limited understanding exists regarding how aging impacts the cellular and molecular aspects of the human ovary. This study combines single-cell RNA sequencing and spatial transcriptomics to systematically characterize human ovarian aging. Spatiotemporal molecular signatures of the eight types of ovarian cells during aging are observed. An analysis of age-associated changes in gene expression reveals that DNA damage response may be a key biological pathway in oocyte aging. Three granulosa cells subtypes and five theca and stromal cells subtypes, as well as their spatiotemporal transcriptomics changes during aging, are identified. FOXP1 emerges as a regulator of ovarian aging, declining with age and inhibiting CDKN1A transcription. Silencing FOXP1 results in premature ovarian insufficiency in mice. These findings offer a comprehensive understanding of spatiotemporal variability in human ovarian aging, aiding the prioritization of potential diagnostic biomarkers and therapeutic strategies.
Collapse
Affiliation(s)
- Meng Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China
| | - Weicheng Tang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China
| | - Ying Chen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China
| | - Liru Xue
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China
| | - Jun Dai
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China.
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China.
| | - Yan Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China.
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China.
| | - Xiaoran Zhu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China
| | - Chuqing Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China
| | - Jiaqiang Xiong
- Department of Obstetrics and Gynecology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Jinjin Zhang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China
| | - Tong Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China
| | - Su Zhou
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China
| | - Dan Chen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China
| | - Chaoyang Sun
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China
| | - Jing Yu
- Shanghai Health Commission Key Lab of Artificial Intelligence (AI)-Based Management of Inflammation and Chronic Diseases, Sino-French Cooperative Central Lab, Shanghai Pudong Gongli Hospital, Secondary Military Medical University, Shanghai, China
| | - Hongyi Li
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, China
| | - Yican Guo
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China
| | - Yibao Huang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China
| | - Qingqing Zhu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China
| | - Simin Wei
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China
| | - Ziliang Zhou
- College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Mingfu Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China
| | - Ya Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China
| | - Tao Xiang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China
| | | | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, China.
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, China.
| |
Collapse
|
47
|
Fan Y, Chen S, Chu C, Yin X, Jin J, Zhang L, Yan H, Cao Z, Liu R, Xin M, Li L, Yin C. TP63 truncating mutation causes increased cell apoptosis and premature ovarian insufficiency by enhanced transcriptional activation of CLCA2. J Ovarian Res 2024; 17:67. [PMID: 38528613 PMCID: PMC10962206 DOI: 10.1186/s13048-024-01396-2] [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: 08/31/2023] [Accepted: 03/18/2024] [Indexed: 03/27/2024] Open
Abstract
BACKGROUND Premature ovarian insufficiency (POI) is a severe disorder leading to female infertility. Genetic mutations are important factors causing POI. TP63-truncating mutation has been reported to cause POI by increasing germ cell apoptosis, however what factors mediate this apoptosis remains unclear. METHODS Ninety-three patients with POI were recruited from Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Whole-exome sequencing (WES) was performed for each patient. Sanger sequencing was used to confirm potential causative genetic variants. A minigene assay was performed to determine splicing effects of TP63 variants. A TP63-truncating plasmid was constructed. Real-time quantitative PCR, western blot analyses, dual luciferase reporter assays, immunofluorescence staining, and cell apoptosis assays were used to study the underlying mechanism of a TP63-truncating mutation causing POI. RESULTS By WES of 93 sporadic patients with POI, we found a 14-bp deletion covering the splice site in the TP63 gene. A minigene assay demonstrated that the 14-bp deletion variant led to exon 13 skipping during TP63 mRNA splicing, resulting in the generation of a truncated TP63 protein (TP63-mut). Overexpression of TP63-mut accelerated cell apoptosis. Mechanistically, the TP63-mut protein could bind to the promoter region of CLCA2 and activate the transcription of CLCA2 several times compared to that of the TP63 wild-type protein. Silencing CLCA2 using a specific small interfering RNA (siRNA) or inhibiting the Ataxia Telangiectasia Mutated (ATM) pathway using the KU55933 inhibitor attenuated cell apoptosis caused by TP63-mut protein expression. CONCLUSION Our findings revealed a crucial role for CLCA2 in mediating apoptosis in POI pathogenesis, and suggested that CLCA2 is a potential therapeutic target for POI.
Collapse
Affiliation(s)
- Yali Fan
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100006, China
| | - Shuya Chen
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100006, China
| | - Chunfang Chu
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
| | - Xiaodan Yin
- Department of Traditional Chinese Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
| | - Jing Jin
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
| | - Lingyan Zhang
- Department of Gynaecology and Obstetrics, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Huihui Yan
- Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
| | - Zheng Cao
- Department of Laboratory Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
| | - Ruixia Liu
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100006, China
| | - Mingwei Xin
- Department of Traditional Chinese Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China.
| | - Lin Li
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100006, China.
| | - Chenghong Yin
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100006, China.
| |
Collapse
|
48
|
Chesnokov MS, Mamedova AR, Zhivotovsky B, Kopeina GS. A matter of new life and cell death: programmed cell death in the mammalian ovary. J Biomed Sci 2024; 31:31. [PMID: 38509545 PMCID: PMC10956231 DOI: 10.1186/s12929-024-01017-6] [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: 09/29/2023] [Accepted: 02/27/2024] [Indexed: 03/22/2024] Open
Abstract
BACKGROUND The mammalian ovary is a unique organ that displays a distinctive feature of cyclic changes throughout the entire reproductive period. The estrous/menstrual cycles are associated with drastic functional and morphological rearrangements of ovarian tissue, including follicular development and degeneration, and the formation and subsequent atrophy of the corpus luteum. The flawless execution of these reiterative processes is impossible without the involvement of programmed cell death (PCD). MAIN TEXT PCD is crucial for efficient and careful clearance of excessive, depleted, or obsolete ovarian structures for ovarian cycling. Moreover, PCD facilitates selection of high-quality oocytes and formation of the ovarian reserve during embryonic and juvenile development. Disruption of PCD regulation can heavily impact the ovarian functions and is associated with various pathologies, from a moderate decrease in fertility to severe hormonal disturbance, complete loss of reproductive function, and tumorigenesis. This comprehensive review aims to provide updated information on the role of PCD in various processes occurring in normal and pathologic ovaries. Three major events of PCD in the ovary-progenitor germ cell depletion, follicular atresia, and corpus luteum degradation-are described, alongside the detailed information on molecular regulation of these processes, highlighting the contribution of apoptosis, autophagy, necroptosis, and ferroptosis. Ultimately, the current knowledge of PCD aberrations associated with pathologies, such as polycystic ovarian syndrome, premature ovarian insufficiency, and tumors of ovarian origin, is outlined. CONCLUSION PCD is an essential element in ovarian development, functions and pathologies. A thorough understanding of molecular mechanisms regulating PCD events is required for future advances in the diagnosis and management of various disorders of the ovary and the female reproductive system in general.
Collapse
Affiliation(s)
- Mikhail S Chesnokov
- Faculty of Medicine, MV Lomonosov Moscow State University, Moscow, Russia
- Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Aygun R Mamedova
- Faculty of Medicine, MV Lomonosov Moscow State University, Moscow, Russia
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Boris Zhivotovsky
- Faculty of Medicine, MV Lomonosov Moscow State University, Moscow, Russia.
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.
- Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden.
| | - Gelina S Kopeina
- Faculty of Medicine, MV Lomonosov Moscow State University, Moscow, Russia.
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.
| |
Collapse
|
49
|
Ni F, Wang F, Li J, Liu Y, Sun X, Chen J, Li J, Zhang Y, Jin J, Ye X, Tu M, Chen J, Chen C, Zhang D. BNC1 deficiency induces mitochondrial dysfunction-triggered spermatogonia apoptosis through the CREB/SIRT1/FOXO3 pathway: the therapeutic potential of nicotinamide riboside and metformin†. Biol Reprod 2024; 110:615-631. [PMID: 38079523 DOI: 10.1093/biolre/ioad168] [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: 05/17/2023] [Revised: 08/14/2023] [Accepted: 02/06/2023] [Indexed: 03/16/2024] Open
Abstract
Male infertility is a global health problem that disturbs numerous couples worldwide. Basonuclin 1 (BNC1) is a transcription factor mainly expressed in proliferative keratinocytes and germ cells. A frameshift mutation of BNC1 was identified in a large Chinese primary ovarian insufficiency pedigree. The expression of BNC1 was significantly decreased in the testis biopsies of infertile patients with nonobstructive azoospermia. Previous studies have revealed that mice with BNC1 deficiency are generally subfertile and undergo gradual spermatogenic failure. We observed that apoptosis of spermatogonia is tightly related to spermatogenic failure in mice with a Bnc1 truncation mutation. Such impairment is related to mitochondrial dysfunction causing lower mitochondrial membrane potential and higher reactive oxygen species. We showed that downregulation of CREB/SIRT1/FOXO3 signaling participates in the above impairment. Administration of nicotinamide riboside or metformin reversed mitochondrial dysfunction and inhibited apoptosis in Bnc1-knockdown spermatogonia by stimulating CREB/SIRT1/FOXO3 signaling. Dietary supplementation with nicotinamide riboside or metformin in mutated mice increased SIRT1 signaling, improved the architecture of spermatogenic tubules, inhibited apoptosis of the testis, and improved the fertility of mice with a Bnc1 truncation mutation. Our data establish that oral nicotinamide riboside or metformin can be useful for the treatment of spermatogenic failure induced by Bnc1 mutation.
Collapse
Affiliation(s)
- Feida Ni
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Feixia Wang
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jingyi Li
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yifeng Liu
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiao Sun
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jianpeng Chen
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jiaqun Li
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yanye Zhang
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jiani Jin
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiaohang Ye
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Mixue Tu
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jianhua Chen
- Department of Pathology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Chuan Chen
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Dan Zhang
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| |
Collapse
|
50
|
Zhang J, Su T, Fan Y, Cheng C, Xu L, LiTian. Spotlight on iron overload and ferroptosis: Research progress in female infertility. Life Sci 2024; 340:122370. [PMID: 38141854 DOI: 10.1016/j.lfs.2023.122370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/15/2023] [Accepted: 12/18/2023] [Indexed: 12/25/2023]
Abstract
Iron is an essential trace element for organisms. However, iron overload, which is common in haematological disorders (e.g. haemochromatosis, myelodysplastic syndromes, aplastic anaemia, and thalassaemia, blood transfusion-dependent or not), can promote reactive oxygen species generation and induce ferroptosis, a novel form of programmed cell death characterised by excess iron and lipid peroxidation, thus causing cell and tissue damage. Infertility is a global health concern. Recent evidence has indicated the emerging role of iron overload and ferroptosis in female infertility by inducing hypogonadism, causing ovary dysfunction, impairing preimplantation embryos, attenuating endometrial receptivity, and crosstalk between subfertility-related disorders, such as polycystic ovary syndrome and endometriosis. In addition, gut microbiota and their metabolites are involved in iron metabolism, ferroptosis, and female infertility. In this review, we systematically elaborate on the current research progress in female infertility with a novel focus on iron overload and ferroptosis and summarise promising therapies targeting iron overload and ferroptosis to recover fertility in women. In summary, our study provides new insights into female infertility and offers literature references for the clinical management of female infertility associated with iron overload and ferroptosis, which may be beneficial for females with haematopoietic disorders suffering from both iron overload and infertility.
Collapse
Affiliation(s)
- Jinghua Zhang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, China; Reproductive Medical Center, Peking University People's Hospital, Beijing 100044, China
| | - Tiantian Su
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, China; Reproductive Medical Center, Peking University People's Hospital, Beijing 100044, China
| | - Yuan Fan
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, China; Reproductive Medical Center, Peking University People's Hospital, Beijing 100044, China
| | - Cheng Cheng
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, China; Reproductive Medical Center, Peking University People's Hospital, Beijing 100044, China
| | - Lanping Xu
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People's Hospital & Institute of Hematology, No. 11 Xizhimen South Street, Xicheng District, Beijing 100044, China
| | - LiTian
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, China; Reproductive Medical Center, Peking University People's Hospital, Beijing 100044, China.
| |
Collapse
|