1
|
Li D, Zhou L, Liu Z, Zhang Z, Mao W, Shi W, Zhu M, Wang F, Wan Y. FTO demethylates regulates cell-cycle progression by controlling CCND1 expression in luteinizing goat granulosa cells. Theriogenology 2024; 216:20-29. [PMID: 38154203 DOI: 10.1016/j.theriogenology.2023.12.029] [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: 09/09/2023] [Revised: 12/21/2023] [Accepted: 12/22/2023] [Indexed: 12/30/2023]
Abstract
In mammals, N6-methyladenosine (m6A) stands out as one of the most abundant internal mRNA modifications and plays a crucial role in follicular development. Nonetheless, the precise mechanism by which the demethylase FTO regulates the progression of the goat luteinizing granulosa cells (LGCs) cycle remains to be elucidated. In our study, we primarily assessed the protein and mRNA expression levels of genes using Western blotting and quantitative real-time polymerase chain reaction (qRT-PCR), cell proliferation via EdU, cell viability with CCK-8, and apoptosis and cell cycle progression through flow cytometry. Here, the results demonstrated that knockdown of FTO significantly enhanced apoptosis, impeded cell proliferation, and increased autophagy levels in goat LGCs. Furthermore, the silencing of FTO substantially reduced cyclin D1 (CCND1) expression through the recognition and degradation of YTHDF2, consequently prolonging the cell cycle progression. This study sheds light on the mechanism by which FTO demethylation governs cell cycle progression by controlling the expression of CCND1 in goat LGCs, underscoring the dynamic role of m6A modification in the regulation of cell cycle progression.
Collapse
Affiliation(s)
- Dongxu Li
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Lei Zhou
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Zifei Liu
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Zhen Zhang
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Weijia Mao
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Wangwang Shi
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Minghui Zhu
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Feng Wang
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yongjie Wan
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
2
|
Münzel T, Hahad O, Daiber A. Coding Hypertension by Nicotine: Unraveling the Sex-Specific Role Of m6A Demethylase for NOX2 RNA-Protein Connection. Hypertension 2024; 81:252-254. [PMID: 38232145 DOI: 10.1161/hypertensionaha.123.22086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Affiliation(s)
- Thomas Münzel
- Department of Cardiology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Omar Hahad
- Department of Cardiology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Andreas Daiber
- Department of Cardiology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
3
|
Wang Y, Chen C, Yan W, Fu Y. Epigenetic modification of m 6A methylation: Regulatory factors, functions and mechanism in inflammatory bowel disease. Int J Biochem Cell Biol 2024; 166:106502. [PMID: 38030117 DOI: 10.1016/j.biocel.2023.106502] [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] [Revised: 11/19/2023] [Accepted: 11/21/2023] [Indexed: 12/01/2023]
Abstract
Although the exact cause of inflammatory bowel disease (IBD) is still unknown, there is a lot of evidence to support the notion that it results from a combination of environmental factors, immune system issues, gut microbial changes, and genetic susceptibility. In recent years, the role of epigenetics in the pathogenesis of IBD has drawn increasing attention. The regulation of IBD-related immunity, the preservation of the intestinal epithelial barrier, and autophagy are all significantly influenced by epigenetic factors. The most extensive epigenetic methylation modification of mammalian mRNA among them is N6-methyladenosine (m6A). It summarizes the general structure and function of the m6A regulating factors, as well as their complex effects on IBD by regulating the intestinal mucous barrier, intestine mucosal immunity, epidermal cell death, and intestinal microorganisms.This paper provides key insights for the future identification of potential new targets for the diagnosis and treatment of IBD.
Collapse
Affiliation(s)
- Yanping Wang
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chaoyue Chen
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Yan
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yu Fu
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
4
|
Zhou J, Han Y, Hou R. Potential role of N6-methyladenosine modification in the development of Parkinson's disease. Front Cell Dev Biol 2023; 11:1321995. [PMID: 38155838 PMCID: PMC10753761 DOI: 10.3389/fcell.2023.1321995] [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: 10/15/2023] [Accepted: 12/04/2023] [Indexed: 12/30/2023] Open
Abstract
N6-methyladenosine (m6A) represents the most abundant modification of messenger RNA (mRNA) and is regulated by methyltransferases (writers), demethylases (erasers), and m6A-binding proteins (readers). A dynamic modification process is implicated in nearly every critical stage of RNA metabolism, including mRNA stability, transcription, translation, splicing, nuclear export, and decay. Notably, m6A methylation is significantly enriched in the brain and has recently been shown to be associated with neurodevelopmental disorders and the development of Parkinson's disease (PD). In this review, we summarize the proteins involved in the process of m6A modification and elucidate the emerging role of m6A modification in PD, which could illuminate alternative strategies for the prevention and treatment of PD.
Collapse
Affiliation(s)
- Jiale Zhou
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yang Han
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Ruizhe Hou
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
5
|
Song YP, Lv JW, Zhang ZC, Qian QH, Fan YJ, Chen DZ, Zhang H, Xu FX, Zhang C, Huang Y, Wang H, Wei W, Xu DX. Effects of Gestational Arsenic Exposures on Placental and Fetal Development in Mice: The Role of Cyr61 m6A. ENVIRONMENTAL HEALTH PERSPECTIVES 2023; 131:97004. [PMID: 37682722 PMCID: PMC10489955 DOI: 10.1289/ehp12207] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 06/13/2023] [Accepted: 08/08/2023] [Indexed: 09/10/2023]
Abstract
BACKGROUND Several epidemiological investigations demonstrated that maternal arsenic (As) exposure elevated risk of fetal growth restriction (FGR), but the mechanism remains unclear. OBJECTIVES This study aimed to investigate the effects of gestational As exposure on placental and fetal development and its underlying mechanism. METHODS Dams were exposed to 0.15, 1.5, and 15 mg / L NaAsO 2 throughout pregnancy via drinking water. Sizes of fetuses and placentas, placental histopathology, and glycogen content were measured. Placental RNA sequencing was conducted. Human trophoblasts were exposed to NaAsO 2 (2 μ M ) to establish an in vitro model of As exposure. The mRNA stability and protein level of genes identified through RNA sequencing were measured. N 6 -Methyladenosine (m 6 A ) modification was detected by methylated RNA immunoprecipitation-quantitative real-time polymerase chain reason (qPCR). The binding ability of insulin-like growth factor 2 binding protein 2 to the gene of interest was detected by RNA-binding protein immunoprecipitation-qPCR. Intracellular S-adenosylmethionine (SAM) and methyltransferase activity were determined by liquid chromatography-tandem mass spectrometry (LC-MS/MS) and colorimetry, respectively. In vitro As + 3 methyltransferase (As3MT) knockdown or SAM supplementation and in vivo folic acid (FA) supplementation were used to evaluate the protective effect. A case-control study verified the findings. RESULTS Sizes of fetuses (exposed to 1.5 and 15 mg / L NaAsO 2 ) and placentas (exposed to 15 mg / L NaAsO 2 ) were lower in As-exposed mice. More glycogen + trophoblasts accumulated and the expression of markers of interstitial invasion was lower in the 15 mg / L NaAsO 2 -exposed mouse group in comparison with control. Placental RNA sequencing identified cysteine-rich angiogenic inducer 61 (Cyr61) as a candidate gene of interest. Mechanistically, mice and cells exposed to As had lower protein expression of CYR61, and this was attributed to a lower incidence of Cyr61 m 6 A . Furthermore, cells exposed to As had lower methyltransferase activity, suggesting that this could be the mechanism by which Cyr61 m 6 A was affected. Depletion of intracellular SAM, a cofactor for m 6 A methyltransferase catalytic domain, partially contributed to As-induced methyltransferase activity reduction. Either As3MT knockdown or SAM supplementation attenuated As-induced Cyr61 m 6 A down-regulation. In mice, FA supplementation rescued As-induced defective trophoblastic invasion and FGR. In humans, a negative correlation between maternal urinary As and plasma CYR61 was observed in infants who were small for gestational age. DISCUSSION Using in vitro and in vivo models, we found that intracellular SAM depletion-mediated Cyr61 m 6 A down-regulation partially contributed to As-induced defective trophoblastic invasion and FGR. https://doi.org/10.1289/EHP12207.
Collapse
Affiliation(s)
- Ya-Ping Song
- Department of Toxicology, Anhui Medical University, Hefei, Anhui, China
| | - Jin-Wei Lv
- Department of Toxicology, Anhui Medical University, Hefei, Anhui, China
| | - Zhi-Cheng Zhang
- Department of Toxicology, Anhui Medical University, Hefei, Anhui, China
| | - Qing-Hua Qian
- Department of Toxicology, Anhui Medical University, Hefei, Anhui, China
| | - Yi-Jun Fan
- Department of Toxicology, Anhui Medical University, Hefei, Anhui, China
- Second Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
| | - Dao-Zhen Chen
- Wuxi Maternity and Child Health Care Hospital, Wuxi, Jiangsu, China
| | - Heng Zhang
- Wuxi Maternity and Child Health Care Hospital, Wuxi, Jiangsu, China
| | - Fei-Xiang Xu
- Department of Toxicology, Anhui Medical University, Hefei, Anhui, China
| | - Cheng Zhang
- Department of Toxicology, Anhui Medical University, Hefei, Anhui, China
| | - Yichao Huang
- Department of Toxicology, Anhui Medical University, Hefei, Anhui, China
| | - Hua Wang
- Department of Toxicology, Anhui Medical University, Hefei, Anhui, China
| | - Wei Wei
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Education Ministry of China, Anhui Medical University, Hefei, Anhui, China
| | - De-Xiang Xu
- Department of Toxicology, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
6
|
Zhang M, Nie J, Chen Y, Li X, Chen H. Connecting the Dots: N6-Methyladenosine (m 6 A) Modification in Spermatogenesis. Adv Biol (Weinh) 2023; 7:e2300068. [PMID: 37353958 DOI: 10.1002/adbi.202300068] [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: 02/09/2023] [Revised: 05/20/2023] [Indexed: 06/25/2023]
Abstract
N6-methyladenosine (m6 A) is the most common RNA modification found in eukaryotes and is involved in multiple biological processes, including neuronal development, tumorigenesis, and gametogenesis. It is well known that methylation-modifying enzymes (classified into writers, erasers, and readers) mediate catalysis, clearance, and recognition of m6 A. Recent studies suggest that these genes may be associated with spermatogenesis. Numerous studies have revealed the m6 A role during spermatogenesis. However, the expression patterns and relationships of these m6 A enzymes during various stages of spermatogenesis remain unknown. In this review, it is aimed to provide an overview of m6 A enzyme functions and elucidate their potential mechanisms and regulatory relationships at a specific phase during spermatogenesis, providing new insights into the m6 A modification underlying the spermatogenesis process.
Collapse
Affiliation(s)
- Mengya Zhang
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, 226000, China
| | - Junyu Nie
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, 226000, China
| | - Yufei Chen
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, 226000, China
| | - Xiaofeng Li
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Lianhua Road No. 1120, Futian District, Shenzhen, Guangdong Province, 518000, P. R. China
| | - Hao Chen
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, 226000, China
| |
Collapse
|
7
|
Li JZ, Zhou SM, Yuan WB, Chen HQ, Zeng Y, Fan J, Zhang Z, Wang N, Cao J, Liu WB. RNA binding protein YTHDF1 mediates bisphenol S-induced Leydig cell damage by regulating the mitochondrial pathway of BCL2 and the expression of CDK2-CyclinE1. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 321:121144. [PMID: 36702435 DOI: 10.1016/j.envpol.2023.121144] [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: 12/05/2022] [Revised: 01/18/2023] [Accepted: 01/22/2023] [Indexed: 06/18/2023]
Abstract
Bisphenol S (BPS) causes reproductive adverse effects on humans and animals. However, the detailed mechanism is still unclear. This research aimed to clarify the role of RNA binding protein YTHDF1 in Leydig cell damage induced by BPS. The mouse TM3 Leydig cells were exposed to BPS of 0, 20, 40, and 80 μmol/L for 72 h. Results showed that TM3 Leydig cells apoptosis rate markedly increased in BPS exposure group. Meanwhile, the apoptosis-related molecule BCL2 protein level decreased significantly, and Caspase9, Caspase3, and BAX increased significantly. Moreover, the cell cycle was blocked in the G1/S phase, CDK2 and CyclinE1 were considerably down-regulated in BPS exposure groups, and the protein level of RNA binding protein YTHDF1 decreased sharply. Furthermore, after overexpression of YTHDF1, the cell viability significantly increased, and the apoptosis rate significantly decreased in TM3 Leydig cells. In the meantime, BCL2, CDK2, and CyclinE1 were significantly up-regulated, and BAX, Caspase9, and Caspase3 were significantly down-regulated. Conversely, interference with YTHDF1 decreased cell proliferation and promoted apoptosis. Importantly, overexpression of YTHDF1 alleviated the cell viability decrease induced by BPS, and interference with YTHDF1 exacerbated the situation. RIP assays showed that the binding of YTHDF1 to CDK2, CyclinE1, and BCL2 significantly increased after overexpressing YTHDF1. Collectively, our study suggested that YTHDF1 plays an essential role in BPS-induced TM3 Leydig cell damage by regulating CDK2-CyclinE1 and BCL2 mitochondrial pathway at the translational level.
Collapse
Affiliation(s)
- Jing-Zhi Li
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Shi-Meng Zhou
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China; School of Public Health, China Medical University, Shenyang, Liaoning, 110122, China
| | - Wen-Bo Yuan
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Hong-Qiang Chen
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Department of Environmental Health, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yong Zeng
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Department of Environmental Health, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jun Fan
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Zhe Zhang
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Na Wang
- Department of Environmental Health, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China; School of Public Health, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - Jia Cao
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Wen-Bin Liu
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Department of Environmental Health, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| |
Collapse
|
8
|
Rader MA, Jaime OG, Abarca VO, Young KA. Photoperiod alters testicular methyltransferase complex mRNA expression in Siberian hamsters. Gen Comp Endocrinol 2023; 333:114186. [PMID: 36521516 PMCID: PMC10575611 DOI: 10.1016/j.ygcen.2022.114186] [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: 05/02/2022] [Revised: 11/06/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022]
Abstract
Exposure to long photoperiods stimulates, whereas exposure to short photoperiods transiently inhibit testicular function in Siberian hamsters via well-described neuroendocrine mechanisms. However, less is known about the intra-testicular regulation of these photoperiod-mediated changes. N6-methyladenosine (m6A) is one of the most common mRNA modifications in eukaryotes, with alterations in m6A mRNA methylation affecting testis function and fertility. We hypothesized that genes controlling m6A methylation such as methyltransferase-like-3 (Mettl3) and -14 (Mettl14) and Wilms' tumor-1 associated protein (Wtap), part of an mRNA methylating methyl-transferase complex, or the fat-mass-and-obesity-associated (Fto) and the α-ketoglutarate-dependent dioxygenase alkB homolog-5 (Alkbh5) genes responsible for m6A demethylation, may be differentially regulated by photoperiod in the testis. Male hamsters were exposed to long (LD, control) photoperiod for 14-weeks, short (SD) photoperiod for 2, 5, 8, 11 and 14-weeks to induce regression, or SD for 14-weeks followed by transfer to LD for 1, 2, 4 or 8-weeks to induce recrudescence (post-transfer, PT). SD exposure significantly reduced body, testis, and epididymal masses compared to all other groups. Spermatogenic index, seminiferous tubule diameters and testosterone concentrations significantly decreased in SD as compared to LD, returning to levels no different than LD in post-transfer groups. SD exposure significantly decreased Wtap, Fto, Alkbh5, but increased Mettl14 mRNA expression as compared to LD, with values in PT groups restored to LD levels. Mettl3 mRNA expression did not change. These results suggest that testicular recovery induced by stimulatory photoperiod is relatively rapid, and that the methyltransferase complex may play a role during photostimulated testicular recrudescence.
Collapse
Affiliation(s)
- Melanie A Rader
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA 90840, USA
| | - Olga G Jaime
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA 90840, USA
| | - Victor O Abarca
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA 90840, USA
| | - Kelly A Young
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA 90840, USA.
| |
Collapse
|
9
|
Liu H, Zheng J, Liao A. The regulation and potential roles of m6A modifications in early embryonic development and immune tolerance at the maternal-fetal interface. Front Immunol 2022; 13:988130. [PMID: 36225914 PMCID: PMC9549360 DOI: 10.3389/fimmu.2022.988130] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 09/09/2022] [Indexed: 12/16/2022] Open
Abstract
The immune microenvironment at the maternal-fetal interface was determined by the crosstalk between the trophoblast and maternal-derived cells, which dynamically changed during the whole gestation. Trophoblasts act as innate immune cells and dialogue with maternal-derived cells to ensure early embryonic development, depending on the local immune microenvironment. Therefore, dysfunctions in trophoblasts and maternal decidual cells contribute to pregnancy complications, especially recurrent pregnancy loss in early pregnancy. Since many unknown regulatory factors still affect the complex immune status, exploring new potential aspects that could influence early pregnancy is essential. RNA methylation plays an important role in contributing to the transcriptional regulation of various cells. Sufficient studies have shown the crucial roles of N6-methyladenosine (m6A)- and m6A-associated- regulators in embryogenesis during implantation. They are also essential in regulating innate and adaptive immune cells and the immune response and shaping the local and systemic immune microenvironment. However, the function of m6A modifications at the maternal-fetal interface still lacks wide research. This review highlights the critical functions of m6A in early embryonic development, summarizes the reported research on m6A in regulating immune cells and tumor immune microenvironment, and identifies the potential value of m6A modifications in shaping trophoblasts, decidual immune cells, and the microenvironment at the maternal-fetal interface. The m6A modifications are more likely to contribute to embryogenesis, placentation and shape the immune microenvironment at the maternal-fetal interface. Uncovering these crucial regulatory mechanisms could provide novel therapeutic targets for RNA methylation in early pregnancy.
Collapse
Affiliation(s)
- Hong Liu
- Department of Reproduction, Maternal and Child Health Hospital of Hubei Province, Affiliated in Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Zheng
- Department of Reproduction, Maternal and Child Health Hospital of Hubei Province, Affiliated in Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Jie Zheng, ; Aihua Liao,
| | - Aihua Liao
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Jie Zheng, ; Aihua Liao,
| |
Collapse
|
10
|
Sun X, Lu J, Li H, Huang B. The Role of m 6A on Female Reproduction and Fertility: From Gonad Development to Ovarian Aging. Front Cell Dev Biol 2022; 10:884295. [PMID: 35712673 PMCID: PMC9197073 DOI: 10.3389/fcell.2022.884295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 04/21/2022] [Indexed: 11/20/2022] Open
Abstract
The growth and maturation of oocyte is accompanied by the accumulation of abundant RNAs and posttranscriptional regulation. N6-methyladenosine (m6A) is the most prevalent epigenetic modification in mRNA, and precisely regulates the RNA metabolism as well as gene expression in diverse physiological processes. Recent studies showed that m6A modification and regulators were essential for the process of ovarian development and its aberrant manifestation could result in ovarian aging. Moreover, the specific deficiency of m6A regulators caused oocyte maturation disorder and female infertility with defective meiotic initiation, subsequently the oocyte failed to undergo germinal vesicle breakdown and consequently lost the ability to resume meiosis by disrupting spindle organization as well as chromosome alignment. Accumulating evidence showed that dysregulated m6A modification contributed to ovarian diseases including polycystic ovarian syndrome (PCOS), primary ovarian insufficiency (POI), ovarian aging and other ovarian function disorders. However, the complex and subtle mechanism of m6A modification involved in female reproduction and fertility is still unknown. In this review, we have summarized the current findings of the RNA m6A modification and its regulators in ovarian life cycle and female ovarian diseases. And we also discussed the role and potential clinical application of the RNA m6A modification in promoting oocyte maturation and delaying the reproduction aging.
Collapse
Affiliation(s)
- Xiaoyan Sun
- State Key Laboratory of Reproductive Medicine, Gusu School, Suzhou Municipal Hospital, Suzhou Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Suzhou, China
| | - Jiafeng Lu
- State Key Laboratory of Reproductive Medicine, Gusu School, Suzhou Municipal Hospital, Suzhou Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Suzhou, China
| | - Hong Li
- State Key Laboratory of Reproductive Medicine, Gusu School, Suzhou Municipal Hospital, Suzhou Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Suzhou, China
| | - Boxian Huang
- State Key Laboratory of Reproductive Medicine, Gusu School, Suzhou Municipal Hospital, Suzhou Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Suzhou, China
| |
Collapse
|
11
|
Chang Y, Yi M, Wang J, Cao Z, Zhou T, Ge W, Muhammad Z, Zhang Z, Feng Y, Yan Z, Felici MD, Shen W, Cao H. Genetic Regulation of N6-Methyladenosine-RNA in Mammalian Gametogenesis and Embryonic Development. Front Cell Dev Biol 2022; 10:819044. [PMID: 35359444 PMCID: PMC8964082 DOI: 10.3389/fcell.2022.819044] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 02/21/2022] [Indexed: 01/20/2023] Open
Abstract
Emerging evidence shows that m6A is the most abundant modification in eukaryotic RNA molecules. It has only recently been found that this epigenetic modification plays an important role in many physiological and pathological processes, such as cell fate commitment, immune response, obesity, tumorigenesis, and relevant for the present review, gametogenesis. Notably the RNA metabolism process mediated by m6A is controlled and regulated by a series of proteins termed writers, readers and erasers that are highly expressed in germ cells and somatic cells of gonads. Here, we review and discuss the expression and the functional emerging roles of m6A in gametogenesis and early embryogenesis of mammals. Besides updated references about such new topics, readers might find in the present work inspiration and clues to elucidate epigenetic molecular mechanisms of reproductive dysfunction and perspectives for future research.
Collapse
Affiliation(s)
- Yuguang Chang
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Mingliang Yi
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Jing Wang
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Zhikun Cao
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Tingting Zhou
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Wei Ge
- Key Laboratory of Animal Reproduction and Biotechnology in Universities of Shandong, College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | - Zafir Muhammad
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Zijun Zhang
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Yanqin Feng
- Key Laboratory of Animal Reproduction and Biotechnology in Universities of Shandong, College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | - Zihui Yan
- Key Laboratory of Animal Reproduction and Biotechnology in Universities of Shandong, College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | - Massimo De Felici
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- *Correspondence: Massimo De Felici, ; Wei Shen, ; Hongguo Cao,
| | - Wei Shen
- Key Laboratory of Animal Reproduction and Biotechnology in Universities of Shandong, College of Life Sciences, Qingdao Agricultural University, Qingdao, China
- *Correspondence: Massimo De Felici, ; Wei Shen, ; Hongguo Cao,
| | - Hongguo Cao
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
- *Correspondence: Massimo De Felici, ; Wei Shen, ; Hongguo Cao,
| |
Collapse
|
12
|
Yuan X, Li T, Shi L, Miao J, Guo Y, Chen Y. Human umbilical cord mesenchymal stem cells deliver exogenous miR-26a-5p via exosomes to inhibit nucleus pulposus cell pyroptosis through METTL14/NLRP3. Mol Med 2021; 27:91. [PMID: 34412584 PMCID: PMC8375162 DOI: 10.1186/s10020-021-00355-7] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 08/11/2021] [Indexed: 12/26/2022] Open
Abstract
Background Intervertebral disc degeneration (IVDD) is the breakdown of the discs supporting the vertebrae. It is one of the most frequent causes of back pain worldwide. Currently, the clinical interventions for IVDD are mainly focused on symptom releases. Thus, new therapeutic options are needed. Methods Nucleus pulposus (NP) samples were obtained from 20 patients experiencing IVDD and 10 healthy volunteers compared for mRNA N6-methyladenosine (m6A) mRNA modification as well as methyltransferase (METT) like METTL3, METTL14, and Wilms’ tumor 1-associated protein mRNA and protein abundance following exosomes exposure from mesenchymal stem cells. In addition, microRNA expressions were also compared. The correlation between the NLR family pyrin domain containing 3 (NLRP3) and METTL14 was measured by luciferase reporter assay. Cytokines were evaluated using an enzyme-linked immunosorbent assay. METTL14, NLRP3, and insulin-like growth factor 2 mRNA-binding protein 2 mRNAs were measured via a quantitative reverse transcription-polymerase chain reaction. Protein was assayed using western blots. Cell death was assessed by propidium iodide staining, lactate dehydrogenase release, gasdermin-N domain abundance, and caspase-1 activation. Results The human umbilical cord mesenchymal stem cell (hucMSC) exosomes were found to effectively improve the viability of NP cells and protect them from pyroptosis through targeting METTL14, with a methyltransferase catalyzing m6A modification. METTL14 was highly present in NP cells from IVDD patients, which stabilize NLRP3 mRNA in an IGFBP2-dependent manner. The elevated NLRP3 levels result in the increase of interleukin 1β (IL-1β) and IL-18 levels and trigger pyroptotic NP cell death. Such pathogenic axis could be blocked by hucMSC exosomes, which directly degrade METTL14 through exosomal miR-26a-5p. Conclusions The results of the current study revealed the beneficial effects of hucMSC exosomes on NP cells and determined a potential mechanism inducing IVDD. Supplementary Information The online version contains supplementary material available at 10.1186/s10020-021-00355-7.
Collapse
Affiliation(s)
- Xiaoqiu Yuan
- Spine Center, Department of Orthopaedics, Changzheng Hospital, Naval Medical University, No 415 Fengyang Road, Shanghai, 200003, China
| | - Tiefeng Li
- Spine Center, Department of Orthopaedics, Changzheng Hospital, Naval Medical University, No 415 Fengyang Road, Shanghai, 200003, China
| | - Lei Shi
- Spine Center, Department of Orthopaedics, Changzheng Hospital, Naval Medical University, No 415 Fengyang Road, Shanghai, 200003, China
| | - Jinhao Miao
- Spine Center, Department of Orthopaedics, Changzheng Hospital, Naval Medical University, No 415 Fengyang Road, Shanghai, 200003, China
| | - Yongfei Guo
- Spine Center, Department of Orthopaedics, Changzheng Hospital, Naval Medical University, No 415 Fengyang Road, Shanghai, 200003, China
| | - Yu Chen
- Spine Center, Department of Orthopaedics, Changzheng Hospital, Naval Medical University, No 415 Fengyang Road, Shanghai, 200003, China.
| |
Collapse
|
13
|
Wilson SL, Wallingford M. Epigenetic regulation of reproduction in human and in animal models. Mol Hum Reprod 2021; 27:6329199. [PMID: 34318322 DOI: 10.1093/molehr/gaab041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/07/2021] [Indexed: 12/24/2022] Open
Affiliation(s)
- Samantha L Wilson
- Princess Margaret Cancer Centre, University Health Network, Toronto Medical Discovery Tower, Toronto, ON, Canada
| | - Mary Wallingford
- Mother Infant Research Institute, Tufts Medical Center, Boston, MA, USA.,Division of Obstetrics and Gynecology, Tufts University School of Medicine, Boston, MA, USA
| |
Collapse
|