1
|
Munisha M, Huang R, Khan J, Schimenti JC. Chronic replication stress-mediated genomic instability disrupts placenta development in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.28.640689. [PMID: 40093167 PMCID: PMC11908151 DOI: 10.1101/2025.02.28.640689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Abnormal placentation drives many pregnancy-related pathologies and poor fetal outcomes, but the underlying molecular causes are understudied. Here, we show that persistent replication stress due to mutations in the MCM2-7 replicative helicase disrupts placentation and reduces embryo viability in mice. MCM-deficient embryos exhibited normal morphology but their placentae had a drastically diminished junctional zone (JZ). Whereas cell proliferation in the labyrinth zone (LZ) remained unaffected, JZ cell proliferation was reduced during development. MCM2-7 deficient trophoblast stem cells (TSCs) failed to maintain stemness, suggesting that replication stress affects the initial trophoblast progenitor pool in a manner that preferentially impacts the developing JZ. In contrast, pluripotency of mouse embryonic stem cells with MCM2-7 deficiency were not affected. Developing female mice deficient for FANCM, a protein involved in replication-associated DNA repair, also had placentae with a diminished JZ. These findings indicate that replication stress-induced genomic instability compromises embryo outcomes by impairing placentation.
Collapse
|
2
|
Sun X, Feng Y, Xie L, Wu Y, Mao J, Zhang Y, Zhang L, Yuan X, Ni J, Xiao X, Liu H, Dai L, Ma F. Maternal plasma extracellular vesicles tsRNA as potential biomarkers for assessing preterm labor risk. BMC Pregnancy Childbirth 2025; 25:553. [PMID: 40348952 PMCID: PMC12065249 DOI: 10.1186/s12884-025-07672-3] [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: 01/15/2025] [Accepted: 04/29/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND Spontaneous preterm labor (PTL) accounts for approximately 70% of preterm births, posing significant risks to both maternal and neonatal health. Current predictive biomarkers lack sufficient reliability, underscoring the need for non-invasive and dependable indicators. Emerging research indicates that tRNA-derived small RNAs (tsRNAs) are involved in various diseases; however, their potential association with PTL remains underexplored. METHODS Bioinformatics analyses of public GEO datasets (PRJNA415953 and PRJNA428989) were conducted to identify tsRNAs associated with PTL. Validation was performed using plasma extracellular vesicles samples collected at 12 weeks of gestation from PTL patients (n = 45) and healthy controls (n = 38). Functional assays were used to assess the impact of tsRNA1 (tRNA-Gly-GCC-5p-tRF-921) on extravillous trophoblast (EVT) function, including apoptosis, migration, invasion, and endothelial-like tube formation in HTR8/SVneo cells. Transcriptomic sequencing was conducted to identify tsRNA1-mediated pathways, and DNA methylation patterns were predicted based on the transcriptomic data. Statistical significance was determined using Student's t-test. RESULTS Two tsRNAs, tsRNA1 and tsRNA3 (tRNA-Gly-GCC-5p-tR-half-368), were significantly upregulated in PTL patient samples compared to controls. Overexpression of tsRNA1 impaired EVT function, increased apoptosis, and altered DNA methylation profiles, implicating its critical role in PTL mechanisms. CONCLUSIONS This study identifies tsRNA1 as a key regulator of EVT dysfunction and placental pathology in PTL. The findings provide novel insights into the mechanistic role of tsRNAs in PTL and highlight tsRNA1 as a promising biomarker for early risk stratification and prediction of the condition. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Xinrui Sun
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), West China Second University Hospital, Ministry of Education, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Ying Feng
- Department of Histology, Embryology and Neurobiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Liang Xie
- The Joint Laboratory for Pulmonary Development and Related Diseases, West China Second University Hospital, West China Institute of Women and Children's Health, Sichuan University, Chengdu, Sichuan, China
| | - Yilun Wu
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), West China Second University Hospital, Ministry of Education, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jia Mao
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, State Key Laboratory of Biotherapy and Cancer Center, College of Life Sciences, Sichuan University, Chengdu, 610064, China
| | - Yi Zhang
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), West China Second University Hospital, Ministry of Education, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Linyu Zhang
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), West China Second University Hospital, Ministry of Education, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xin Yuan
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), West China Second University Hospital, Ministry of Education, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jiangping Ni
- College of Horticulture, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Xue Xiao
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China.
| | - Hanmin Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China.
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China.
| | - Li Dai
- National Center for Birth Defects Monitoring, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China.
- National Health Commission Key Laboratory of Chronobiology (Sichuan University), Chengdu, Sichuan, China.
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China.
| | - Fang Ma
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), West China Second University Hospital, Ministry of Education, Sichuan University, Chengdu, Sichuan, 610041, P. R. China.
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
3
|
Cai S, Xue B, Li S, Wang X, Zeng X, Zhu Z, Fan X, Zou Y, Yu H, Qiao S, Zeng X. Methionine regulates maternal-fetal immune tolerance and endometrial receptivity by enhancing embryonic IL-5 secretion. Cell Rep 2025; 44:115291. [PMID: 39937648 DOI: 10.1016/j.celrep.2025.115291] [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: 01/30/2024] [Revised: 07/25/2024] [Accepted: 01/17/2025] [Indexed: 02/14/2025] Open
Abstract
Endometrial receptivity and maternal-fetal immune tolerance are two crucial processes for a successful pregnancy. However, the molecular mechanisms of nutrition involved are largely unexplored. Here, we showed that maternal methionine supply significantly improved pregnancy outcomes, which was closely related to interleukin-5 (IL-5) concentration. Mechanistically, methionine induced embryonic IL-5 secretion, which enhanced the conversion of CD4+ T cells to IL-5+ Th2 cells in the uterus, thereby improving maternal-fetal immune tolerance. Meanwhile, methionine-mediated IL-5 secretion activated the nuclear factor κB (NF-κB) pathway and enhanced integrin αvβ3 expression in endometrial cells, which improved endometrial receptivity. Further, methionine strongly influenced the DNA methylation and transcription levels of the transcription factor eomesodermin (Eomes), which bound directly to the IL-5 promoter region and inhibited IL-5 transcription. Methionine modulated IL-5 transcription, maternal-fetal immune tolerance, and endometrial receptivity via its effects on Eomes. This study reveals the crucial functions of methionine and IL-5 and offers a potential nutritional strategy for successful pregnancy.
Collapse
Affiliation(s)
- Shuang Cai
- State Key Laboratory of Animal Nutrition and Feeding, Ministry of Agriculture and Rural Affairs Feed Industry Center, China Agricultural University, Beijing, China; Frontier Technology Research Institute of China Agricultural University in Shenzhen, Shenzhen, China
| | - Bangxin Xue
- State Key Laboratory of Animal Nutrition and Feeding, Ministry of Agriculture and Rural Affairs Feed Industry Center, China Agricultural University, Beijing, China; Frontier Technology Research Institute of China Agricultural University in Shenzhen, Shenzhen, China
| | - Siyu Li
- State Key Laboratory of Animal Nutrition and Feeding, Ministry of Agriculture and Rural Affairs Feed Industry Center, China Agricultural University, Beijing, China; Frontier Technology Research Institute of China Agricultural University in Shenzhen, Shenzhen, China
| | - Xinyu Wang
- State Key Laboratory of Animal Nutrition and Feeding, Ministry of Agriculture and Rural Affairs Feed Industry Center, China Agricultural University, Beijing, China; Frontier Technology Research Institute of China Agricultural University in Shenzhen, Shenzhen, China
| | - Xiangzhou Zeng
- State Key Laboratory of Animal Nutrition and Feeding, Ministry of Agriculture and Rural Affairs Feed Industry Center, China Agricultural University, Beijing, China; Frontier Technology Research Institute of China Agricultural University in Shenzhen, Shenzhen, China
| | - Zhekun Zhu
- State Key Laboratory of Animal Nutrition and Feeding, Ministry of Agriculture and Rural Affairs Feed Industry Center, China Agricultural University, Beijing, China; Frontier Technology Research Institute of China Agricultural University in Shenzhen, Shenzhen, China
| | - Xinyin Fan
- State Key Laboratory of Animal Nutrition and Feeding, Ministry of Agriculture and Rural Affairs Feed Industry Center, China Agricultural University, Beijing, China; Frontier Technology Research Institute of China Agricultural University in Shenzhen, Shenzhen, China
| | - Yijin Zou
- State Key Laboratory of Animal Nutrition and Feeding, Ministry of Agriculture and Rural Affairs Feed Industry Center, China Agricultural University, Beijing, China; Frontier Technology Research Institute of China Agricultural University in Shenzhen, Shenzhen, China
| | - Haitao Yu
- State Key Laboratory of Animal Nutrition and Feeding, Ministry of Agriculture and Rural Affairs Feed Industry Center, China Agricultural University, Beijing, China; Frontier Technology Research Institute of China Agricultural University in Shenzhen, Shenzhen, China
| | - Shiyan Qiao
- State Key Laboratory of Animal Nutrition and Feeding, Ministry of Agriculture and Rural Affairs Feed Industry Center, China Agricultural University, Beijing, China; Frontier Technology Research Institute of China Agricultural University in Shenzhen, Shenzhen, China
| | - Xiangfang Zeng
- State Key Laboratory of Animal Nutrition and Feeding, Ministry of Agriculture and Rural Affairs Feed Industry Center, China Agricultural University, Beijing, China; Frontier Technology Research Institute of China Agricultural University in Shenzhen, Shenzhen, China.
| |
Collapse
|
4
|
Liu L, Tang L, Chen S, Zheng L, Ma X. Decoding the molecular pathways governing trophoblast migration and placental development; a literature review. Front Endocrinol (Lausanne) 2024; 15:1486608. [PMID: 39665023 PMCID: PMC11631628 DOI: 10.3389/fendo.2024.1486608] [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: 08/26/2024] [Accepted: 11/08/2024] [Indexed: 12/13/2024] Open
Abstract
Placental development is a multifaceted process critical for a fruitful pregnancy, reinforced by a complex network of molecular pathways that synchronize trophoblast migration, differentiation, and overall placental function. This review provides an in-depth analysis of the key signaling pathways, such as Wnt, Notch, TGF-β, and VEGF, which play fundamental roles in trophoblast proliferation, invasion, and the complicated process of placental vascular development. For instance, the Wnt signaling pathway is essential to balance trophoblast stem cell proliferation and differentiation, while Notch signaling stimulates cell fate decisions and invasive behavior. TGF-β signaling plays a critical role in trophoblast invasion and differentiation, predominantly in response to the low oxygen environment of early pregnancy, regulated by hypoxia-inducible factors (HIFs). These factors promote trophoblast adaptation, ensure proper placental attachment and vascularization, and facilitate adequate fetal-maternal exchange. Further, we explore the epigenetic and post-transcriptional regulatory mechanisms that regulate trophoblast function, including DNA methylation and the contribution of non-coding RNAs, which contribute to the fine-tuning of gene expression during placental development. Dysregulation of these pathways is associated with severe pregnancy complications, such as preeclampsia, intrauterine growth restriction, and recurrent miscarriage, emphasizing the critical need for targeted therapeutic strategies. Finally, emerging technologies like trophoblast organoids, single-cell RNA sequencing, and placenta-on-chip models are discussed as innovative tools that hold promise for advancing our understanding of placental biology and developing novel interventions to improve pregnancy outcomes. This review emphasizes the importance of understanding these molecular mechanisms to better address placental dysfunctions and associated pregnancy disorders.
Collapse
Affiliation(s)
- Lianlian Liu
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Lin Tang
- Obstetrics Department, Foshan Maternity and Child Health Care Hospital, Foshan, China
| | - Shuai Chen
- Pathology Department, The Second Hospital of Jilin University, Changchun, China
| | - Lianwen Zheng
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Xiaoyan Ma
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
5
|
Lai Y, Fu Z, Gao Y, Ma N, Li L. Hypoxia-inducible factors (HIFs) in early pregnancy: implications for miscarriage†. Biol Reprod 2024; 111:987-999. [PMID: 39325972 DOI: 10.1093/biolre/ioae139] [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: 05/09/2024] [Revised: 09/07/2024] [Accepted: 09/25/2024] [Indexed: 09/28/2024] Open
Abstract
Miscarriage poses a significant threat to both maternal and fetal health. Its etiology remains unknown, and there are no established effective identification or prevention strategies. A low-oxygen environment in early pregnancy is a physiological necessity for embryonic and placental growth. Hypoxia-inducible factors are a family of classic hypoxia signaling molecules whose expression level may fluctuate abnormally because of an imbalance in oxygen levels. Its unusual fluctuations initiate multiple signaling pathways at the maternal womb. Hypoxia-inducible factors are a family of classic hypoxia-signaling molecules and immune tolerance. Notably, aberrant regulation of these processes may lead to miscarriage. This review aims to clarify how the hypoxia-inducible factor-1α mediates the aberrant regulation of biological processes, including autophagy, metabolic reprogramming, et al., and how these effects impact trophoblasts and other cells at the maternal-fetal interface. These findings provide new insights into potential therapeutic and preventive strategies for miscarriage.
Collapse
Affiliation(s)
- Yuxuan Lai
- Department of Social Medicine and Health Care Management, School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Zhiyu Fu
- Department of Social Medicine and Health Care Management, School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Yaxin Gao
- Department of Social Medicine and Health Care Management, School of Public Health, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Environment and Population Health, School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Ning Ma
- Department of Social Medicine and Health Care Management, School of Public Health, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Environment and Population Health, School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Lu Li
- Department of Social Medicine and Health Care Management, School of Public Health, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Environment and Population Health, School of Public Health, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
6
|
Fang S, Chang KW, Lefebvre L. Roles of endogenous retroviral elements in the establishment and maintenance of imprinted gene expression. Front Cell Dev Biol 2024; 12:1369751. [PMID: 38505259 PMCID: PMC10948482 DOI: 10.3389/fcell.2024.1369751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 02/26/2024] [Indexed: 03/21/2024] Open
Abstract
DNA methylation (DNAme) has long been recognized as a host defense mechanism, both in the restriction modification systems of prokaryotes as well as in the transcriptional silencing of repetitive elements in mammals. When DNAme was shown to be implicated as a key epigenetic mechanism in the regulation of imprinted genes in mammals, a parallel with host defense mechanisms was drawn, suggesting perhaps a common evolutionary origin. Here we review recent work related to this hypothesis on two different aspects of the developmental imprinting cycle in mammals that has revealed unexpected roles for long terminal repeat (LTR) retroelements in imprinting, both canonical and noncanonical. These two different forms of genomic imprinting depend on different epigenetic marks inherited from the mature gametes, DNAme and histone H3 lysine 27 trimethylation (H3K27me3), respectively. DNAme establishment in the maternal germline is guided by transcription during oocyte growth. Specific families of LTRs, evading silencing mechanisms, have been implicated in this process for specific imprinted genes. In noncanonical imprinting, maternally inherited histone marks play transient roles in transcriptional silencing during preimplantation development. These marks are ultimately translated into DNAme, notably over LTR elements, for the maintenance of silencing of the maternal alleles in the extraembryonic trophoblast lineage. Therefore, LTR retroelements play important roles in both establishment and maintenance of different epigenetic pathways leading to imprinted expression during development. Because such elements are mobile and highly polymorphic among different species, they can be coopted for the evolution of new species-specific imprinted genes.
Collapse
Affiliation(s)
| | | | - Louis Lefebvre
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
7
|
Lu X, Mao J, Qian C, Lei H, Mu F, Sun H, Yan S, Fang Z, Lu J, Xu Q, Dong J, Su D, Wang J, Jin N, Chen S, Wang X. High estrogen during ovarian stimulation induced loss of maternal imprinted methylation that is essential for placental development via overexpression of TET2 in mouse oocytes. Cell Commun Signal 2024; 22:135. [PMID: 38374066 PMCID: PMC10875811 DOI: 10.1186/s12964-024-01516-x] [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: 12/17/2023] [Accepted: 02/07/2024] [Indexed: 02/21/2024] Open
Abstract
BACKGROUND Ovarian stimulation (OS) during assisted reproductive technology (ART) appears to be an independent factor influencing the risk of low birth weight (LBW). Previous studies identified the association between LBW and placenta deterioration, potentially resulting from disturbed genomic DNA methylation in oocytes caused by OS. However, the mechanisms by which OS leads to aberrant DNA methylation patterns in oocytes remains unclear. METHODS Mouse oocytes and mouse parthenogenetic embryonic stem cells (pESCs) were used to investigate the roles of OS in oocyte DNA methylation. Global 5-methylcytosine (5mC) and 5-hydroxymethylcytosine (5hmC) levels were evaluated using immunofluorescence or colorimetry. Genome-wide DNA methylation was quantified using an Agilent SureSelectXT mouse Methyl-Seq. The DNA methylation status of mesoderm-specific transcript homologue (Mest) promoter region was analyzed using bisulfite sequencing polymerase chain reaction (BSP). The regulatory network between estrogen receptor alpha (ERα, ESR1) and DNA methylation status of Mest promoter region was further detected following the knockdown of ERα or ten-eleven translocation 2 (Tet2). RESULTS OS resulted in a significant decrease in global 5mC levels and an increase in global 5hmC levels in oocytes. Further investigation revealed that supraphysiological β-estradiol (E2) during OS induced a notable decrease in DNA 5mC and an increase in 5hmC in both oocytes and pESCs of mice, whereas inhibition of estrogen signaling abolished such induction. Moreover, Tet2 may be a direct transcriptional target gene of ERα, and through the ERα-TET2 axis, supraphysiological E2 resulted in the reduced global levels of DNA 5mC. Furthermore, we identified that MEST, a maternal imprinted gene essential for placental development, lost its imprinted methylation in parthenogenetic placentas originating from OS, and ERα and TET2 combined together to form a protein complex that may promote Mest demethylation. CONCLUSIONS In this study, a possible mechanism of loss of DNA methylation in oocyte caused by OS was revealed, which may help increase safety and reduce epigenetic abnormalities in ART procedures.
Collapse
Affiliation(s)
- Xueyan Lu
- Reproductive Medicine Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Air Force Medical University, No.1, Xinsi Road, Baqiao District, Xi'an, 710000, Shaanxi Province, China
| | - Jiaqin Mao
- Reproductive Medicine Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Air Force Medical University, No.1, Xinsi Road, Baqiao District, Xi'an, 710000, Shaanxi Province, China
| | - Chenxi Qian
- Reproductive Medicine Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Air Force Medical University, No.1, Xinsi Road, Baqiao District, Xi'an, 710000, Shaanxi Province, China
| | - Hui Lei
- Reproductive Medicine Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Air Force Medical University, No.1, Xinsi Road, Baqiao District, Xi'an, 710000, Shaanxi Province, China
| | - Fei Mu
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| | - Huijun Sun
- Reproductive Medicine Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Air Force Medical University, No.1, Xinsi Road, Baqiao District, Xi'an, 710000, Shaanxi Province, China
| | - Song Yan
- Reproductive Medicine Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Air Force Medical University, No.1, Xinsi Road, Baqiao District, Xi'an, 710000, Shaanxi Province, China
| | - Zheng Fang
- Reproductive Medicine Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Air Force Medical University, No.1, Xinsi Road, Baqiao District, Xi'an, 710000, Shaanxi Province, China
| | - Jie Lu
- Reproductive Medicine Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Air Force Medical University, No.1, Xinsi Road, Baqiao District, Xi'an, 710000, Shaanxi Province, China
| | - Qian Xu
- Reproductive Medicine Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Air Force Medical University, No.1, Xinsi Road, Baqiao District, Xi'an, 710000, Shaanxi Province, China
| | - Jie Dong
- Reproductive Medicine Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Air Force Medical University, No.1, Xinsi Road, Baqiao District, Xi'an, 710000, Shaanxi Province, China
| | - Danjie Su
- Reproductive Medicine Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Air Force Medical University, No.1, Xinsi Road, Baqiao District, Xi'an, 710000, Shaanxi Province, China
| | - Jingjing Wang
- Reproductive Medicine Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Air Force Medical University, No.1, Xinsi Road, Baqiao District, Xi'an, 710000, Shaanxi Province, China
| | - Ni Jin
- Reproductive Medicine Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Air Force Medical University, No.1, Xinsi Road, Baqiao District, Xi'an, 710000, Shaanxi Province, China
| | - Shuqiang Chen
- Reproductive Medicine Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Air Force Medical University, No.1, Xinsi Road, Baqiao District, Xi'an, 710000, Shaanxi Province, China.
| | - Xiaohong Wang
- Reproductive Medicine Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Air Force Medical University, No.1, Xinsi Road, Baqiao District, Xi'an, 710000, Shaanxi Province, China.
| |
Collapse
|
8
|
Mostafa Domiaty D, Ibrahim Al-Hazani TM, Alshehri E, Zamil aldajani H, Fahad Alqassim NA, Mohammed Al-balawi A, Abdullah AlQassim F, Abdullah Alduwish M, Saeed Al-Qahtani W. SARS-CoV-2 impact on ACE2 expression in NSCLC: mRNA and protein insights COVID-19 associated (ACE2) expression in non-small cell lung cancer (NSCLC). Heliyon 2024; 10:e23926. [PMID: 38261909 PMCID: PMC10796980 DOI: 10.1016/j.heliyon.2023.e23926] [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: 07/01/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 01/25/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) is a pervasive and challenging global health concern. This research delves into the intricate relationship between NSCLC and ACE2 expression, exploring the potential impact of COVID-19 history on this interaction. Tissue samples were meticulously gathered from a cohort of 32 NSCLC patients, 18 of whom had a documented history of COVID-19 infection. The methodology included extensive investigations, such as cell dissociation, histopathological analysis, immunohistochemistry, cell culture, adhesion assays, immunocytochemistry, RNA isolation, and RT-PCR analysis. The results of this comprehensive study unearthed intriguing findings regarding ACE2 expression patterns within NSCLC tissues. Notably, variations were observed in ACE2 profiles between individuals with and without a prior record of COVID-19 infection, hinting at a dynamic interplay. These discoveries carry profound implications for both the understanding of NSCLC progression and the response to COVID-19 in patients with pre-existing NSCLC. The interrelationship between ACE2 expression, NSCLC, and COVID-19, as revealed in this study, may significantly influence patient outcomes and, potentially, therapeutic strategies. In summary, this research serves as an essential contribution to the growing body of knowledge on NSCLC, offering unique insights into the intricate connections between ACE2, COVID-19, and NSCLC. This information may open new avenues for tailored treatment approaches and clinical management strategies, ultimately benefiting patients grappling with NSCLC in the background of the current COVID-19 pandemic.
Collapse
Affiliation(s)
- Dalia Mostafa Domiaty
- College of Science, Department of Biological Sciences, University of Jeddah, P.O. BOX 13151, Jeddah, 21493, Jeddah, Saudi Arabia
| | - Tahani Mohamed Ibrahim Al-Hazani
- Department of Biology, College of Sciences and Humanities, Prince Sattam Bin Abdulaziz University, P.O. Box 83, Al-Kharj, 11940, Saudi Arabia
| | - Eman Alshehri
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Haya Zamil aldajani
- King Abdulaziz Medical City, Ministry of National Guard Affairs, Riyadh, Saudi Arabia
| | | | | | | | - Manal Abdullah Alduwish
- Department of Biology, College of Sciences and Humanities, Prince Sattam Bin Abdulaziz University, P.O. Box 83, Al-Kharj, 11940, Saudi Arabia
| | - Wedad Saeed Al-Qahtani
- Department of Forensic Sciences, College of Criminal Justice, Naif Arab University for Security Sciences, P.O. Box 6830, 11452, Riyadh, Saudi Arabia
| |
Collapse
|
9
|
Doria-Borrell P, Moya-Navamuel M, Hemberger M, Pérez-García V. Generation of Knockout Mouse Trophoblast Stem Cells by CRISPR/Cas9. Methods Mol Biol 2024; 2781:81-91. [PMID: 38502445 DOI: 10.1007/978-1-0716-3746-3_8] [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: 03/21/2024]
Abstract
The placenta is the organ that dictates the reproductive outcome of mammalian pregnancy by supplying nutrients and oxygen to the developing fetus to sustain its normal growth. During early mammalian development, trophoblast cells are the earliest cell type to differentiate with multipotent capacity to generate the trophoblast components of the placenta. The isolation and use of mouse trophoblast stem cells (mTSCs) to model in vitro trophoblast differentiation, in combination with CRISPR/Cas9 genome editing technology, has provided tremendous insight into the molecular mechanisms governing early mouse placentation. By knocking out a specific gene of interest in mTSCs, researchers are shedding light onto the molecular pathways involved in normal placental development and pregnancy disorders associated with abnormal placentation. In this chapter, we provide a detailed protocol for the genetic modification of mTSCs by using CRISPR/Cas9 genome editing system.
Collapse
Affiliation(s)
- Paula Doria-Borrell
- Centro de Investigación Príncipe Felipe, Eduardo Primo Yúfera, Valencia, Spain
| | - Maria Moya-Navamuel
- Centro de Investigación Príncipe Felipe, Eduardo Primo Yúfera, Valencia, Spain
| | - Myriam Hemberger
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | | |
Collapse
|
10
|
Alotaibi MA, Al-Hazani TMI, Alwaili MA, Jalal AS, Alshaya DS, Safhi FA, Alamoudi MO, Alarifi S, Saeed Al-Qahtani W. SARS-CoV-2 virus associated angiotensin converting enzyme 2 expression modulation in colorectal cancer: Insights from mRNA and protein analysis COVID-19 associated (ACE2) expression in colorectal cancer. Microb Pathog 2023; 185:106389. [PMID: 37839761 DOI: 10.1016/j.micpath.2023.106389] [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: 08/19/2023] [Revised: 10/08/2023] [Accepted: 10/09/2023] [Indexed: 10/17/2023]
Abstract
The SARS-CoV-2 virus gains entry into human cells by exploiting the angiotensin-converting enzyme 2 (ACE2), a key component known as the spike protein (S), as a point of entry. Initially, SARS-CoV-2 suppresses the natural function of ACE2, leading to a gradual decline in cell health. Additionally, individuals with cancer are considered more susceptible to COVID-19. This study investigates the expression patterns of ACE2 in colorectal cancer (CRC) patients with and without a history of COVID-19 infection. RT-PCR was used to analyze samples from both cancerous and adjacent non-affected colorectal tissues of 47 CRC patients, comprising two groups: 24 CRC patients with no history of COVID-19 and 23 CRC patients with a recent history of COVID-19 infection. Epithelial CR cells were isolated from both types of tissues and cultured to evaluate cell adhesion. Immunohistochemistry analyses were conducted to examine ACE2 protein expression using various ACE2 antibodies for both cell types. The study revealed ACE2 mRNA expression in all CRC tissues of patients with and without a history of COVID-19. ACE2 expression was significantly higher in CRC patients without a history of COVID-19. Notably, the non-affected colorectal cancer (NACRC) tissues of patients without a history of COVID-19 also showed ACE2 expression, whereas no ACE2 expression was detected in the biopsies of CRC patients with a positive COVID-19 history. ACE2 antibodies were employed to validate ACE2 protein expression at the mRNA level. COVID-19 appears to downregulate ACE2 expression in both CRC and NACRC tissues of CRC patients with a positive history of COVID-19 infection.
Collapse
Affiliation(s)
| | - Tahani Mohamed Ibrahim Al-Hazani
- Department of Biology, College of Sciences and Humanities, Prince Sattam Bin Abdulaziz University, P.O. Box 83, Al-Kharj, 11940, Saudi Arabia.
| | - Maha Abdulla Alwaili
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh, 11671, Saudi Arabia
| | - Areej Saud Jalal
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh, 11671, Saudi Arabia
| | - Dalal S Alshaya
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh, 11671, Saudi Arabia
| | - Fatmah Ahmed Safhi
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh, 11671, Saudi Arabia
| | - Muna O Alamoudi
- Department of Biology, Faculty of Science, University of Hail, Hail, 81411, Saudi Arabia
| | - Saud Alarifi
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Wedad Saeed Al-Qahtani
- Department of Forensic Sciences, College of Criminal Justice, Naif Arab University for Security Sciences, P.O. Box 6830, Riyadh, 11452, Saudi Arabia.
| |
Collapse
|
11
|
Weigert R, Hetzel S, Bailly N, Haggerty C, Ilik IA, Yung PYK, Navarro C, Bolondi A, Kumar AS, Anania C, Brändl B, Meierhofer D, Lupiáñez DG, Müller FJ, Aktas T, Elsässer SJ, Kretzmer H, Smith ZD, Meissner A. Dynamic antagonism between key repressive pathways maintains the placental epigenome. Nat Cell Biol 2023; 25:579-591. [PMID: 37024684 PMCID: PMC10104784 DOI: 10.1038/s41556-023-01114-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 02/21/2023] [Indexed: 04/08/2023]
Abstract
DNA and Histone 3 Lysine 27 methylation typically function as repressive modifications and operate within distinct genomic compartments. In mammals, the majority of the genome is kept in a DNA methylated state, whereas the Polycomb repressive complexes regulate the unmethylated CpG-rich promoters of developmental genes. In contrast to this general framework, the extra-embryonic lineages display non-canonical, globally intermediate DNA methylation levels, including disruption of local Polycomb domains. Here, to better understand this unusual landscape's molecular properties, we genetically and chemically perturbed major epigenetic pathways in mouse trophoblast stem cells. We find that the extra-embryonic epigenome reflects ongoing and dynamic de novo methyltransferase recruitment, which is continuously antagonized by Polycomb to maintain intermediate, locally disordered methylation. Despite its disorganized molecular appearance, our data point to a highly controlled equilibrium between counteracting repressors within extra-embryonic cells, one that can seemingly persist indefinitely without bistable features typically seen for embryonic forms of epigenetic regulation.
Collapse
Affiliation(s)
- Raha Weigert
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany
- Department of Medical Biotechnology, Technische Universität Berlin, Berlin, Germany
| | - Sara Hetzel
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Nina Bailly
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany
- Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Chuck Haggerty
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany
- Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Ibrahim A Ilik
- Otto Warburg Laboratories, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Philip Yuk Kwong Yung
- Science for Life Laboratory, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- Ming Wai Lau Centre for Reparative Medicine, Stockholm node, Karolinska Institutet, Stockholm, Sweden
| | - Carmen Navarro
- Science for Life Laboratory, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- Ming Wai Lau Centre for Reparative Medicine, Stockholm node, Karolinska Institutet, Stockholm, Sweden
| | - Adriano Bolondi
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany
- Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Abhishek Sampath Kumar
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Chiara Anania
- Epigenetics and Sex Development Group, Berlin Institute for Medical Systems Biology, Max-Delbrück Center for Molecular Medicine, Berlin-Buch, Germany
| | - Björn Brändl
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany
- Universitätsklinikum Schleswig-Holstein Campus Kiel, Zentrum für Integrative Psychiatrie gGmbH, Kiel, Germany
| | - David Meierhofer
- Mass Spectrometry Joint Facilities Scientific Service, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Darío G Lupiáñez
- Epigenetics and Sex Development Group, Berlin Institute for Medical Systems Biology, Max-Delbrück Center for Molecular Medicine, Berlin-Buch, Germany
| | - Franz-Josef Müller
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany
- Universitätsklinikum Schleswig-Holstein Campus Kiel, Zentrum für Integrative Psychiatrie gGmbH, Kiel, Germany
| | - Tugce Aktas
- Otto Warburg Laboratories, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Simon J Elsässer
- Science for Life Laboratory, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- Ming Wai Lau Centre for Reparative Medicine, Stockholm node, Karolinska Institutet, Stockholm, Sweden
| | - Helene Kretzmer
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Zachary D Smith
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT, USA.
| | - Alexander Meissner
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany.
- Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, Berlin, Germany.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, US.
| |
Collapse
|
12
|
Andrews S, Krueger C, Mellado-Lopez M, Hemberger M, Dean W, Perez-Garcia V, Hanna CW. Mechanisms and function of de novo DNA methylation in placental development reveals an essential role for DNMT3B. Nat Commun 2023; 14:371. [PMID: 36690623 PMCID: PMC9870994 DOI: 10.1038/s41467-023-36019-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 01/11/2023] [Indexed: 01/24/2023] Open
Abstract
DNA methylation is a repressive epigenetic modification that is essential for development, exemplified by the embryonic and perinatal lethality observed in mice lacking de novo DNA methyltransferases (DNMTs). Here we characterise the role for DNMT3A, 3B and 3L in gene regulation and development of the mouse placenta. We find that each DNMT establishes unique aspects of the placental methylome through targeting to distinct chromatin features. Loss of Dnmt3b results in de-repression of germline genes in trophoblast lineages and impaired formation of the maternal-foetal interface in the placental labyrinth. Using Sox2-Cre to delete Dnmt3b in the embryo, leaving expression intact in placental cells, the placental phenotype was rescued and, consequently, the embryonic lethality, as Dnmt3b null embryos could now survive to birth. We conclude that de novo DNA methylation by DNMT3B during embryogenesis is principally required to regulate placental development and function, which in turn is critical for embryo survival.
Collapse
Affiliation(s)
- Simon Andrews
- Bioinformatics Programme, Babraham Institute, Cambridge, UK
| | - Christel Krueger
- Bioinformatics Programme, Babraham Institute, Cambridge, UK
- Epigenetics Programme, Babraham Institute, Cambridge, UK
- Bioinformatics Innovation Hub, Altos Labs Cambridge Institute, Cambridge, UK
| | | | - Myriam Hemberger
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Wendy Dean
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada
| | | | - Courtney W Hanna
- Epigenetics Programme, Babraham Institute, Cambridge, UK.
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
| |
Collapse
|
13
|
Genome-wide assessment of DNA methylation alterations induced by superovulation, sexual immaturity and in vitro follicle growth in mouse blastocysts. Clin Epigenetics 2023; 15:9. [PMID: 36647174 PMCID: PMC9843966 DOI: 10.1186/s13148-023-01421-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 01/03/2023] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND In their attempt to fulfill the wish of having children, women who suffer from fertility issues often undergo assisted reproductive technologies such as ovarian stimulation, which has been associated with adverse health outcomes and imprinting disorders in children. However, given the crucial role of exogenous hormone stimulation in improving human infertility treatments, a more comprehensive analysis of the potential impacts on DNA methylation in embryos following ovarian stimulation is needed. Here, we provide genome-wide DNA methylation profiles of blastocysts generated after superovulation of prepubertal or adult mice, compared with blastocysts derived from non-stimulated adult mice. Additionally, we assessed the impact of the in vitro growth and maturation of oocytes on methylation in blastocysts. RESULTS Neither hormone stimulation nor sexual maturity had an impact on the low global methylation levels characteristic of the blastocyst stage or was associated with extensive DNA methylation alterations. However, we found hormone- and age-associated changes at specific positions but dispersed throughout the genome. In particular, we detected anomalous methylation at a limited number of CpG islands. Additionally, superovulation in adult mice was associated with alterations at the Sgce and Zfp777 imprinted genes. On the other hand, in vitro culture of follicles from the early pre-antral stage was associated with globally reduced methylation and increased variability at imprinted loci in blastocysts. CONCLUSIONS Our results indicate a minimal effect of ovarian stimulation of adult and prepubertal mice on the DNA methylation landscape attained at the blastocyst stage, but potentially greater impacts of in vitro growth and maturation of oocytes. These findings have potential significance for the improvement of assisted reproductive techniques, in particular for those related to treatments in prepubertal females, which could be crucial for improving human fertility preservation strategies.
Collapse
|
14
|
Zhang X, Wang C, He D, Cheng Y, Yu L, Qi D, Li B, Zheng F. Identification of DNA methylation-regulated genes as potential biomarkers for coronary heart disease via machine learning in the Framingham Heart Study. Clin Epigenetics 2022; 14:122. [PMID: 36180886 PMCID: PMC9526342 DOI: 10.1186/s13148-022-01343-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 09/25/2022] [Indexed: 11/10/2022] Open
Abstract
Background DNA methylation-regulated genes have been demonstrated as the crucial participants in the occurrence of coronary heart disease (CHD). The machine learning based on DNA methylation-regulated genes has tremendous potential for mining non-invasive predictive biomarkers and exploring underlying new mechanisms of CHD. Results First, the 2085 age-gender-matched individuals in Framingham Heart Study (FHS) were randomly divided into training set and validation set. We then integrated methylome and transcriptome data of peripheral blood leukocytes (PBLs) from the training set to probe into the methylation and expression patterns of CHD-related genes. A total of five hub DNA methylation-regulated genes were identified in CHD through dimensionality reduction, including ATG7, BACH2, CDKN1B, DHCR24 and MPO. Subsequently, methylation and expression features of the hub DNA methylation-regulated genes were used to construct machine learning models for CHD prediction by LightGBM, XGBoost and Random Forest. The optimal model established by LightGBM exhibited favorable predictive capacity, whose AUC, sensitivity, and specificity were 0.834, 0.672, 0.864 in the validation set, respectively. Furthermore, the methylation and expression statuses of the hub genes were verified in monocytes using methylation microarray and transcriptome sequencing. The methylation statuses of ATG7, DHCR24 and MPO and the expression statuses of ATG7, BACH2 and DHCR24 in monocytes of our study population were consistent with those in PBLs from FHS. Conclusions We identified five DNA methylation-regulated genes based on a predictive model for CHD using machine learning, which may clue the new epigenetic mechanism for CHD. Supplementary Information The online version contains supplementary material available at 10.1186/s13148-022-01343-2.
Collapse
Affiliation(s)
- Xiaokang Zhang
- Center for Gene Diagnosis and Department of Clinical Laboratory Medicine, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, China
| | - Chen Wang
- Center for Gene Diagnosis and Department of Clinical Laboratory Medicine, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, China
| | - Dingdong He
- Center for Gene Diagnosis and Department of Clinical Laboratory Medicine, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, China.,Department of Clinical Laboratory Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yating Cheng
- Center for Gene Diagnosis and Department of Clinical Laboratory Medicine, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, China
| | - Li Yu
- Center for Gene Diagnosis and Department of Clinical Laboratory Medicine, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, China
| | - Daoxi Qi
- Center for Gene Diagnosis and Department of Clinical Laboratory Medicine, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, China
| | - Boyu Li
- Center for Gene Diagnosis and Department of Clinical Laboratory Medicine, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, China
| | - Fang Zheng
- Center for Gene Diagnosis and Department of Clinical Laboratory Medicine, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, China.
| |
Collapse
|
15
|
Dvoran M, Nemcova L, Kalous J. An Interplay between Epigenetics and Translation in Oocyte Maturation and Embryo Development: Assisted Reproduction Perspective. Biomedicines 2022; 10:biomedicines10071689. [PMID: 35884994 PMCID: PMC9313063 DOI: 10.3390/biomedicines10071689] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/25/2022] [Accepted: 06/28/2022] [Indexed: 12/11/2022] Open
Abstract
Germ cell quality is a key prerequisite for successful fertilization and early embryo development. The quality is determined by the fine regulation of transcriptomic and proteomic profiles, which are prone to alteration by assisted reproduction technology (ART)-introduced in vitro methods. Gaining evidence shows the ART can influence preset epigenetic modifications within cultured oocytes or early embryos and affect their developmental competency. The aim of this review is to describe ART-determined epigenetic changes related to the oogenesis, early embryogenesis, and further in utero development. We confront the latest epigenetic, related epitranscriptomic, and translational regulation findings with the processes of meiotic maturation, fertilization, and early embryogenesis that impact the developmental competency and embryo quality. Post-ART embryo transfer, in utero implantation, and development (placentation, fetal development) are influenced by environmental and lifestyle factors. The review is emphasizing their epigenetic and ART contribution to fetal development. An epigenetic parallel among mouse, porcine, and bovine animal models and human ART is drawn to illustrate possible future mechanisms of infertility management as well as increase the awareness of the underlying mechanisms governing oocyte and embryo developmental complexity under ART conditions.
Collapse
|
16
|
Sandovici I, Fernandez-Twinn DS, Hufnagel A, Constância M, Ozanne SE. Sex differences in the intergenerational inheritance of metabolic traits. Nat Metab 2022; 4:507-523. [PMID: 35637347 DOI: 10.1038/s42255-022-00570-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 04/05/2022] [Indexed: 02/02/2023]
Abstract
Strong evidence suggests that early-life exposures to suboptimal environmental factors, including those in utero, influence our long-term metabolic health. This has been termed developmental programming. Mounting evidence suggests that the growth and metabolism of male and female fetuses differ. Therefore, sexual dimorphism in response to pre-conception or early-life exposures could contribute to known sex differences in susceptibility to poor metabolic health in adulthood. However, until recently, many studies, especially those in animal models, focused on a single sex, or, often in the case of studies performed during intrauterine development, did not report the sex of the animal at all. In this review, we (a) summarize the evidence that male and females respond differently to a suboptimal pre-conceptional or in utero environment, (b) explore the potential biological mechanisms that underlie these differences and (c) review the consequences of these differences for long-term metabolic health, including that of subsequent generations.
Collapse
Affiliation(s)
- Ionel Sandovici
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Denise S Fernandez-Twinn
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Antonia Hufnagel
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Miguel Constância
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK.
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
| | - Susan E Ozanne
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
| |
Collapse
|
17
|
Abstract
The metaphase II (MII) oocyte is the mature female gamete, produced from a complex maturation process called oogenesis that starts in the first weeks of embryogenesis in the female embryo tract, continues during puberty, and is completed at fertilization with the spermatozoon. Oogenesis is closely related to folliculogenesis. In assisted reproduction techniques, oocytes are retrieved in cumulus-oocyte complexes after ovarian stimulation. Before being used for in vitro fertilization or cryopreservation, the metaphase (MII) oocytes can be classified according to different morphological traits and by the presence/absence of the meiotic spindle. Except for a few and rare morphological characteristics that make the oocyte discarded, none of the morphological characteristics is predictive of oocyte competence in giving a viable embryo. On the other side, specific key performance indicators based on MII oocytes test the efficacy of in vitro treatments. Molecular, cellular, or genetic abnormalities in the oocytes have observable consequences on the embryo development dynamics and its genetic content. Besides what can be seen in vitro, several intrinsic and extrinsic factors related to the patient are responsible for the oocyte quality. The clinician and the patient herself must be aware of these factors to preserve the reproductive functions as much as possible. In the present review, we have revised oogenesis and the role of mature oocytes in supporting the fertilization process and early embryo development; we have also listed the oocyte morphological traits and key performance indicators related to the oocyte quality and studied the intrinsic and extrinsic factors that irreversibly impact female fertility.
Collapse
Affiliation(s)
- Sandrine Chamayou
- Unit of Reproductive Medicine, HERA Center, Sant'Agata Li Battiati, Catania, Italy -
| |
Collapse
|
18
|
Liu H, Wang LL, Xu QH, Wang J, Zhang YJ, Luo J, Liao AH. UHRF1 shapes both the trophoblast invasion and decidual macrophage differentiation in early pregnancy. FASEB J 2022; 36:e22247. [PMID: 35262949 DOI: 10.1096/fj.202101647rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 02/04/2022] [Accepted: 02/23/2022] [Indexed: 11/11/2022]
Abstract
Trophoblasts play critical roles in establishment and maintenance of a normal pregnancy. Their dysfunction in early pregnancy is closely related to pregnancy-related diseases, including recurrent pregnancy loss (RPL). Epigenetic modifications dynamically change during pregnancy; however, the role of the epigenetic modifier UHRF1 in trophoblast regulation remains unknown. This is the first study to show that UHRF1 expression was localized in the cytoplasm of cytotrophoblasts, syncytiotrophoblasts, and villi columns, and decreased in the villi of patients with RPL. The invasion and cell viability in a UHRF1 knockdown trophoblast cell line were significantly decreased. In addition, the mRNA expression profiles of Swan71 cells were partially altered by UHRF1 knockdown. The altered immune-related genes were screened out and the pro-inflammatory TH1-type chemokine/cytokines CXCL2 and IL-1β were identified as the most promising targets of UHRF1 in the trophoblasts, which were significantly increased in the UHRF1 knockdown Swan71 cells, villi, and serum from patients with RPL. The macrophages treated with the supernatants of UHRF1 knockdown Swan71 cells were polarized to the M1 phenotype and secreted high levels of pro-inflammatory cytokines, which might be driven by the activated MyD88/NF-κB signaling pathway and mediated by the increased expression of CXCR2 and IL-1R1 (CXCL2 and IL-1β receptors, respectively). In addition, the supernatants of UHRF1 knockdown Swan71 cells showed stronger chemotaxis to macrophages than those from the controls. Our findings highlight the previously unknown roles of UHRF1 as one of the key regulators on the trophoblasts and their cross-talk with local immune cells, and demonstrate a potential approach for RPL intervention.
Collapse
Affiliation(s)
- Hong Liu
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Li-Ling Wang
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Qian-Han Xu
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Jing Wang
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Yu-Jing Zhang
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Jing Luo
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Ai-Hua Liao
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| |
Collapse
|
19
|
Sobočan N, Himelreich-Perić M, Katušić-Bojanac A, Krasić J, Sinčić N, Majić Ž, Jurić-Lekić G, Šerman L, Marić A, Ježek D, Bulić-Jakuš F. Extended Prophylactic Effect of N-tert-Butyl-α-phenylnitron against Oxidative/Nitrosative Damage Caused by the DNA-Hypomethylating Drug 5-Azacytidine in the Rat Placenta. Int J Mol Sci 2022; 23:603. [PMID: 35054786 PMCID: PMC8775603 DOI: 10.3390/ijms23020603] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/02/2022] [Accepted: 01/04/2022] [Indexed: 12/10/2022] Open
Abstract
Antioxidant N-tert-Butyl-α-phenylnitron (PBN) partly protected embryos from the negative effects of a DNA demethylating drug 5-azacytidine during pregnancy. Our aim was to investigate PBN's impact on the placenta. Fischer rat dams were treated on gestation days (GD) 12 and 13 by PBN (40 mg/kg), followed by 5azaC (5 mg/kg) after one hour. Global methylation was assessed by pyrosequencing. Numerical density was calculated from immunohistochemical expression in single cells for proliferating (PCNA), oxidative (oxoguanosine) and nitrosative (nitrotyrosine) activity. Results were compared with the PBN-treated and control rats. PBN-pretreatment significantly increased placental weight at GD15 and GD20, diminished by 5azaC, and diminished apoptosis in GD 20 placentas caused by 5azaC. Oxoguanosine expression in placentas of 5azaC-treated dams was especially high in the placental labyrinth on GD 15, while PBN-pretreatment lowered its expression on GD 15 and GD 20 in both the labyrinth and basal layer. 5azaC enhanced nitrotyrosine level in the labyrinth of both gestational stages, while PBN-pretreatment lowered it. We conclude that PBN exerted its prophylactic activity against DNA hypomethylating agent 5azaC in the placenta through free radical scavenging, especially in the labyrinthine part of the placenta until the last day of pregnancy.
Collapse
Affiliation(s)
- Nikola Sobočan
- Scientific Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (N.S.); (A.K.-B.); (J.K.); (N.S.); (G.J.-L.); (L.Š.); (D.J.); (F.B.-J.)
- Department of Gastroenterology, University Hospital Merkur, 10000 Zagreb, Croatia
| | - Marta Himelreich-Perić
- Scientific Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (N.S.); (A.K.-B.); (J.K.); (N.S.); (G.J.-L.); (L.Š.); (D.J.); (F.B.-J.)
- Department of Biology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia;
| | - Ana Katušić-Bojanac
- Scientific Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (N.S.); (A.K.-B.); (J.K.); (N.S.); (G.J.-L.); (L.Š.); (D.J.); (F.B.-J.)
- Department of Biology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia;
| | - Jure Krasić
- Scientific Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (N.S.); (A.K.-B.); (J.K.); (N.S.); (G.J.-L.); (L.Š.); (D.J.); (F.B.-J.)
- Department of Biology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia;
| | - Nino Sinčić
- Scientific Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (N.S.); (A.K.-B.); (J.K.); (N.S.); (G.J.-L.); (L.Š.); (D.J.); (F.B.-J.)
- Department of Biology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia;
| | - Željka Majić
- Department of Biology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia;
| | - Gordana Jurić-Lekić
- Scientific Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (N.S.); (A.K.-B.); (J.K.); (N.S.); (G.J.-L.); (L.Š.); (D.J.); (F.B.-J.)
- Department of Histology and Embryology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Ljiljana Šerman
- Scientific Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (N.S.); (A.K.-B.); (J.K.); (N.S.); (G.J.-L.); (L.Š.); (D.J.); (F.B.-J.)
- Department of Biology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia;
| | - Andreja Marić
- Department of Internal Medicine, County Hospital Čakovec, 40000 Čakovec, Croatia;
| | - Davor Ježek
- Scientific Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (N.S.); (A.K.-B.); (J.K.); (N.S.); (G.J.-L.); (L.Š.); (D.J.); (F.B.-J.)
- Department of Histology and Embryology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Floriana Bulić-Jakuš
- Scientific Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (N.S.); (A.K.-B.); (J.K.); (N.S.); (G.J.-L.); (L.Š.); (D.J.); (F.B.-J.)
- Department of Biology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia;
| |
Collapse
|
20
|
Pastor WA, Kwon SY. Distinctive aspects of the placental epigenome and theories as to how they arise. Cell Mol Life Sci 2022; 79:569. [PMID: 36287261 PMCID: PMC9606139 DOI: 10.1007/s00018-022-04568-9] [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: 03/05/2022] [Revised: 08/18/2022] [Accepted: 09/21/2022] [Indexed: 11/26/2022]
Abstract
The placenta has a methylome dramatically unlike that of any somatic cell type. Among other distinctions, it features low global DNA methylation, extensive “partially methylated domains” packed in dense heterochromatin and methylation of hundreds of CpG islands important in somatic development. These features attract interest in part because a substantial fraction of human cancers feature the exact same phenomena, suggesting parallels between epigenome formation in placentation and cancer. Placenta also features an expanded set of imprinted genes, some of which come about by distinctive developmental pathways. Recent discoveries, some from far outside the placental field, shed new light on how the unusual placental epigenetic state may arise. Nonetheless, key questions remain unresolved.
Collapse
Affiliation(s)
- William A Pastor
- Department of Biochemistry, McGill University, Montreal, QC, H3G 1Y6, Canada.
- Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, H3A 1A3, Canada.
| | - Sin Young Kwon
- Department of Biochemistry, McGill University, Montreal, QC, H3G 1Y6, Canada
| |
Collapse
|
21
|
Li N, Hou R, Liu C, Yang T, Qiao C, Wei J. Integration of transcriptome and proteome profiles in placenta accreta reveals trophoblast over-migration as the underlying pathogenesis. Clin Proteomics 2021; 18:31. [PMID: 34963445 PMCID: PMC8903580 DOI: 10.1186/s12014-021-09336-8] [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: 04/22/2021] [Accepted: 12/09/2021] [Indexed: 12/03/2022] Open
Abstract
Background Placenta accreta (PA) is a major cause of maternal morbidity and mortality in modern obstetrics, few studies have explored the underlying molecular mechanisms. Methods In our study, transcriptome and proteome profiling were performed in placental tissues from ten participants including five cases each in the PA and control groups to clarify the pathogenesis of PA. Results We identified differential expression of 37,743 transcripts and 160 proteins between the PA and control groups with an overlap rate of 0.09%. The 33 most-significant transcripts and proteins were found and further screened and analyzed. Adhesion-related signature, chemotaxis related signatures and immune related signature were found in the PA group and played a certain role. Sum up two points, three significant indicators, methyl-CpG-binding domain protein 2 (MeCP2), podocin (PODN), and apolipoprotein D (ApoD), which participate in “negative regulation of cell migration”, were downregulated at the mRNA and protein levels in PA group. Furthermore, transwell migration and invasion assay of HTR-8/SVneo cell indicated the all of them impaired the migration and invasion of trophoblast. Conclusion A poor correlation was observed between the transcriptome and proteome data and MeCP2, PODN, and ApoD decreased in transcriptome and proteome profiling, resulting in increased migration of trophoblasts in the PA group, which clarify the mechanism of PA and might be the biomarkers or therapy targets in the future. Supplementary Information The online version contains supplementary material available at 10.1186/s12014-021-09336-8.
Collapse
Affiliation(s)
- Na Li
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China.,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province; Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Benxi, China
| | - Rui Hou
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China.,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province; Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Benxi, China
| | - Caixia Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China.,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province; Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Benxi, China
| | - Tian Yang
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Chong Qiao
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China. .,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province; Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Benxi, China.
| | - Jun Wei
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China. .,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province; Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Benxi, China.
| |
Collapse
|
22
|
Wu D, Shi L, Chen F, Lin Q, Kong J. Methylation Status of the miR-141-3p Promoter Regulates miR-141-3p Expression, Inflammasome Formation, and the Invasiveness of HTR-8/SVneo Cells. Cytogenet Genome Res 2021; 161:501-513. [PMID: 34879371 DOI: 10.1159/000519740] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 09/19/2021] [Indexed: 11/19/2022] Open
Abstract
MicroRNA-141 (miR-141-3p) is upregulated in preeclampsia. This study investigated the effect of methylation of the miR-141-3p promoter on cell viability, invasion capability, and inflammasomes in vitro. The expression of miR-141-3p and methylation status of the miR-141-3p promoter were examined by RT-qPCR and pyrosequencing in villus tissues of women with spontaneous delivery (VTsd), villus tissues of women with preeclampsia (VTpe), and also in HTR-8/SVneo cells treated with a miR-141-3p inhibitor and 20 μmol/L 5-aza-2'-deoxycytidine (5-Aza), a DNA methyltransferase inhibitor. Cell viability and invasion were evaluated by CCK-8 and transwell assays. In addition, the levels of CXCL12, CXCR4, CXCR2, MMPs, NLRP3, and ASC expression were assessed by western blotting, and IL-1β and IL-18 concentrations were assayed by ELISA. miR-141-3p expression was upregulated, and the levels of miR-141-3p promoter methylation and CXCL12, CXCR4, and CXCR2 expression were decreased in VTpe relative to VTsd. In HTR-8/SVneo cells, hypomethylation caused by 5-Aza treatment increased miR-141-3p expression, while DNA methyltransferase 3 (DNMT3) transfection decreased miR-141-3p expression. miRNA-141-3p induced NLRP3, IL-1β, and IL-18 production, decreased CXCR4, MMP, and MMP2 production, and suppressed cell growth and invasion. Furthermore, we observed that NLRP3 plays an important mediatory role in the effects of miR-141-3p described above. Decreased methylation of the miR-141-3p promoter increases miR-141-3p expression, which in turn increases NLRP3 expression, resulting in higher IL-1β and IL-18 levels and lower levels of MMP2/9 and CXCR4. We conclude that modification of the miR-141-3p promoter might be a curial mediator in preeclampsia.
Collapse
Affiliation(s)
- Dongcai Wu
- Department of Obstetrics, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Li Shi
- Department of Medical Ultrasonics, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Fangrong Chen
- Department of Obstetrics, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Qing Lin
- Department of Obstetrics, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Jiao Kong
- Department of Obstetrics, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| |
Collapse
|
23
|
Mortimer R, James K, Bormann CL, Harris AL, Yeh J, Toth TL, Souter I, Roberts DJ, Sacha CR. Male factor infertility and placental pathology in singleton live births conceived with in vitro fertilization. J Assist Reprod Genet 2021; 38:3223-3232. [PMID: 34704166 PMCID: PMC8666390 DOI: 10.1007/s10815-021-02344-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 10/15/2021] [Indexed: 12/23/2022] Open
Abstract
PURPOSE We sought to determine whether pregnancies conceived in those with male factor infertility have unique placental pathology profiles compared to those undergoing infertility treatments for other indications. METHODS This was a retrospective cohort study of placental pathology from 464 live births conceived from autologous fresh IVF cycles at an academic fertility center from 2004 to 2017. Placental pathology was compared between live births arising from patients with male factor infertility alone and those with another infertility diagnosis. Placental outcomes were compared with parametric or non-parametric tests; logistic regression was performed to account for potential confounders. RESULTS Compared to cycles performed for a non-male factor diagnosis, male factor infertility cycles had a higher mean paternal age (38.2 years vs. 36.5 years, p < 0.001), a higher female mean BMI (24.3 vs. 23.3 kg/m2, p = 0.01), and a lower day 3 follicle stimulating hormone (FSH) level (6.8 vs. 7.3 IU/mL, p = 0.02). The mean numbers of embryos transferred, and day of transfer were similar between groups, and more cycles used ICSI in the male factor infertility group (90.6% vs. 22.5%, p < 0.001). Placental pathology in our adjusted model was similar between the male factor and non-male factor groups. In our unadjusted subgroup analysis, cycles for male factor using ICSI appeared to lead to more small placentas by weight compared to cycles performed with conventional insemination (45.8% < 10th percentile vs. 18.8%, p = 0.04). CONCLUSION Male factor infertility is not associated with significantly different placental pathology compared to other infertility diagnoses.
Collapse
Affiliation(s)
- Roisin Mortimer
- Department of OB/GYN, Massachusetts General Hospital and Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA.
| | - K James
- Center for Outcomes Research, Department of OB/GYN, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - C L Bormann
- Massachusetts General Hospital Fertility Center and Harvard Medical School, Boston, MA, USA
| | - A L Harris
- Department of Women's Health, Wright Patterson Air Force Base, Wright Patterson AFB, Dayton, OH, USA
- Department of OB/GYN, Boonshoft School of Medicine, Wright State University, Dayton, OH, USA
| | - J Yeh
- Division of Reprod Endo & Infertil, UMass Medical, Worcester, MA, USA
| | - T L Toth
- Boston IVF, Department of OB/GYN, Harvard Medical School and Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - I Souter
- Massachusetts General Hospital Fertility Center and Harvard Medical School, Boston, MA, USA
| | - D J Roberts
- Department of Pathology, Harvard Medical School and Massachusetts General, Boston Hospital, Boston, MA, USA
| | - C R Sacha
- Massachusetts General Hospital Fertility Center and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
24
|
Allen MC, Moog NK, Buss C, Yen E, Gustafsson HC, Sullivan EL, Graham AM. Co-occurrence of preconception maternal childhood adversity and opioid use during pregnancy: Implications for offspring brain development. Neurotoxicol Teratol 2021; 88:107033. [PMID: 34601061 PMCID: PMC8578395 DOI: 10.1016/j.ntt.2021.107033] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 09/20/2021] [Accepted: 09/24/2021] [Indexed: 12/11/2022]
Abstract
Understanding of the effects of in utero opioid exposure on neurodevelopment is a priority given the recent dramatic increase in opioid use among pregnant individuals. However, opioid abuse does not occur in isolation-pregnant individuals abusing opioids often have a significant history of adverse experiences in childhood, among other co-occurring factors. Understanding the specific pathways in which these frequently co-occurring factors may interact and cumulatively influence offspring brain development in utero represents a priority for future research in this area. We highlight maternal history of childhood adversity (CA) as one such co-occurring factor that is more prevalent among individuals using opioids during pregnancy and which is increasingly shown to affect offspring neurodevelopment through mechanisms beginning in utero. Despite the high incidence of CA history in pregnant individuals using opioids, we understand very little about the effects of comorbid prenatal opioid exposure and maternal CA history on fetal brain development. Here, we first provide an overview of current knowledge regarding effects of opioid exposure and maternal CA on offspring neurodevelopment that may occur during gestation. We then outline potential mechanistic pathways through which these factors might have interactive and cumulative influences on offspring neurodevelopment as a foundation for future research in this area.
Collapse
Affiliation(s)
- Madeleine C Allen
- Department of Psychiatry, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239, United States
| | - Nora K Moog
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Psychology, Luisenstrasse 57, 10117 Berlin, Germany
| | - Claudia Buss
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Psychology, Luisenstrasse 57, 10117 Berlin, Germany; Development, Health and Disease Research Program, University of California, Irvine, 837 Health Sciences Drive, Irvine, California 92697, United States
| | - Elizabeth Yen
- Department of Pediatrics, Tufts Medical Center, Boston, MA 02111, United States
| | - Hanna C Gustafsson
- Department of Psychiatry, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239, United States
| | - Elinor L Sullivan
- Department of Psychiatry, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239, United States; Division of Neuroscience, Oregon National Primate Research Center, 505 NW 185(th) Ave., Beaverton, OR 97006, United States; Department of Behavioral Neuroscience, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239, United States
| | - Alice M Graham
- Department of Psychiatry, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239, United States.
| |
Collapse
|
25
|
The Impact of Hypoxia in Early Pregnancy on Placental Cells. Int J Mol Sci 2021; 22:ijms22189675. [PMID: 34575844 PMCID: PMC8466283 DOI: 10.3390/ijms22189675] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 09/04/2021] [Accepted: 09/05/2021] [Indexed: 12/14/2022] Open
Abstract
Oxygen levels in the placental microenvironment throughout gestation are not constant, with severe hypoxic conditions present during the first trimester. This hypoxic phase overlaps with the most critical stages of placental development, i.e., blastocyst implantation, cytotrophoblast invasion, and spiral artery remodeling initiation. Dysregulation of any of these steps in early gestation can result in pregnancy loss and/or adverse pregnancy outcomes. Hypoxia has been shown to regulate not only the self-renewal, proliferation, and differentiation of trophoblast stem cells and progenitor cells, but also the recruitment, phenotype, and function of maternal immune cells. In this review, we will summarize how oxygen levels in early placental development determine the survival, fate, and function of several important cell types, e.g., trophoblast stem cells, extravillous trophoblasts, syncytiotrophoblasts, uterine natural killer cells, Hofbauer cells, and decidual macrophages. We will also discuss the cellular mechanisms used to cope with low oxygen tensions, such as the induction of hypoxia-inducible factor (HIF) or mammalian target of rapamycin (mTOR) signals, regulation of the metabolic pathway, and adaptation to autophagy. Understanding the beneficial roles of hypoxia in early placental development will provide insights into the root cause(s) of some pregnancy disorders, such as spontaneous abortion, preeclampsia, and intrauterine growth restriction.
Collapse
|
26
|
Human HAND1 Inhibits the Conversion of Cholesterol to Steroids in Trophoblasts. J Genet Genomics 2021; 49:350-363. [PMID: 34391879 DOI: 10.1016/j.jgg.2021.07.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/11/2021] [Accepted: 07/17/2021] [Indexed: 11/24/2022]
Abstract
Steroidogenesis from cholesterol in placental trophoblasts is fundamentally involved in the establishment and maintenance of pregnancy. The transcription factor gene Heart And Neural crest Derivatives expressed 1 (Hand1) promotes differentiation of mouse trophoblast giant cells. However, the role of HAND1 in human trophoblasts remains unknown. Here, we report that HAND1 inhibits human trophoblastic progesterone (P4) and estradiol (E2) from cholesterol through down-regulation of the expression of steroidogenic enzymes including aromatase, P450 cholesterol side-chain cleavage enzyme (P450scc) and 3β-hydroxysteroid dehydrogenase type 1 (3β-HSD1). Mechanically, while HAND1 inhibits transcription of aromatase by directly binding to aromatase gene promoter, it restrains transcription of P450scc by up-regulation of the methylation status of P450scc gene promoter through its binding to ALKBH1, a demethylase. Unlike aromatase and P450scc, HAND1 decreases 3β-HSD1 mRNA levels by reduction of its RNA stability through binding to and subsequent destabilization of protein HuR. Finally, HAND1 suppresses circulating P4 and E2 levels derived from JEG-3 xenograft, and attenuates uterine response to P4 and E2. Thus, our results uncover a hitherto uncharacterized role of HAND1 in regulation of cholesterol metabolism in human trophoblasts, which may help pinpoint the underlying mechanisms involved in supporting the development and physiological function of the human placenta.
Collapse
|
27
|
Bebbere D, Albertini DF, Coticchio G, Borini A, Ledda S. The subcortical maternal complex: emerging roles and novel perspectives. Mol Hum Reprod 2021; 27:6311673. [PMID: 34191027 DOI: 10.1093/molehr/gaab043] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/19/2021] [Indexed: 11/13/2022] Open
Abstract
Since its recent discovery, the subcortical maternal complex (SCMC) is emerging as a maternally inherited and crucial biological structure for the initial stages of embryogenesis in mammals. Uniquely expressed in oocytes and preimplantation embryos, where it localizes to the cell subcortex, this multiprotein complex is essential for early embryo development in the mouse and is functionally conserved across mammalian species, including humans. The complex has been linked to key processes leading the transition from oocyte to embryo, including meiotic spindle formation and positioning, regulation of translation, organelle redistribution, and epigenetic reprogramming. Yet, the underlying molecular mechanisms for these diverse functions are just beginning to be understood, hindered by unresolved interplay of SCMC components and variations in early lethal phenotypes. Here we review recent advances confirming involvement of the SCMC in human infertility, revealing an unexpected relationship with offspring health. Moreover, SCMC organization is being further revealed in terms of novel components and interactions with additional cell constituents. Collectively, this evidence prompts new avenues of investigation into possible roles during the process of oogenesis and the regulation of maternal transcript turnover during the oocyte to embryo transition.
Collapse
Affiliation(s)
- Daniela Bebbere
- Department of Veterinary Medicine, University of Sassari, Sassari, Italy
| | | | | | | | - Sergio Ledda
- Department of Veterinary Medicine, University of Sassari, Sassari, Italy
| |
Collapse
|
28
|
Perez-Garcia V, Lea G, Lopez-Jimenez P, Okkenhaug H, Burton GJ, Moffett A, Turco MY, Hemberger M. BAP1/ASXL complex modulation regulates epithelial-mesenchymal transition during trophoblast differentiation and invasion. eLife 2021; 10:63254. [PMID: 34170818 PMCID: PMC8233037 DOI: 10.7554/elife.63254] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 06/14/2021] [Indexed: 12/28/2022] Open
Abstract
Normal function of the placenta depends on the earliest developmental stages when trophoblast cells differentiate and invade into the endometrium to establish the definitive maternal-fetal interface. Previously, we identified the ubiquitously expressed tumour suppressor BRCA1-associated protein 1 (BAP1) as a central factor of a novel molecular node controlling early mouse placentation. However, functional insights into how BAP1 regulates trophoblast biology are still missing. Using CRISPR/Cas9 knockout and overexpression technology in mouse trophoblast stem cells, here we demonstrate that the downregulation of BAP1 protein is essential to trigger epithelial-mesenchymal transition (EMT) during trophoblast differentiation associated with a gain of invasiveness. Moreover, we show that the function of BAP1 in suppressing EMT progression is dependent on the binding of BAP1 to additional sex comb-like (ASXL1/2) proteins to form the polycomb repressive deubiquitinase (PR-DUB) complex. Finally, both endogenous expression patterns and BAP1 overexpression experiments in human trophoblast stem cells suggest that the molecular function of BAP1 in regulating trophoblast differentiation and EMT progression is conserved in mice and humans. Our results reveal that the physiological modulation of BAP1 determines the invasive properties of the trophoblast, delineating a new role of the BAP1 PR-DUB complex in regulating early placentation.
Collapse
Affiliation(s)
- Vicente Perez-Garcia
- Epigenetics Programme, The Babraham Institute, Babraham Research Campus, Cambridge, United Kingdom.,Centre for Trophoblast Research, Department of Physiology, Development and Neurosicence, University of Cambridge, Cambridge, United Kingdom.,Centro de Investigación Príncipe Felipe, Eduardo Primo Yúfera, Valencia, Spain.,Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Georgia Lea
- Epigenetics Programme, The Babraham Institute, Babraham Research Campus, Cambridge, United Kingdom
| | | | - Hanneke Okkenhaug
- Epigenetics Programme, The Babraham Institute, Babraham Research Campus, Cambridge, United Kingdom
| | - Graham J Burton
- Centre for Trophoblast Research, Department of Physiology, Development and Neurosicence, University of Cambridge, Cambridge, United Kingdom
| | - Ashley Moffett
- Centre for Trophoblast Research, Department of Physiology, Development and Neurosicence, University of Cambridge, Cambridge, United Kingdom.,Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Margherita Y Turco
- Centre for Trophoblast Research, Department of Physiology, Development and Neurosicence, University of Cambridge, Cambridge, United Kingdom.,Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Myriam Hemberger
- Epigenetics Programme, The Babraham Institute, Babraham Research Campus, Cambridge, United Kingdom.,Centre for Trophoblast Research, Department of Physiology, Development and Neurosicence, University of Cambridge, Cambridge, United Kingdom.,Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Canada
| |
Collapse
|
29
|
Defective folate metabolism causes germline epigenetic instability and distinguishes Hira as a phenotype inheritance biomarker. Nat Commun 2021; 12:3714. [PMID: 34140513 PMCID: PMC8211854 DOI: 10.1038/s41467-021-24036-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 05/29/2021] [Indexed: 02/02/2023] Open
Abstract
The mechanism behind transgenerational epigenetic inheritance is unclear, particularly through the maternal grandparental line. We previously showed that disruption of folate metabolism in mice by the Mtrr hypomorphic mutation results in transgenerational epigenetic inheritance of congenital malformations. Either maternal grandparent can initiate this phenomenon, which persists for at least four wildtype generations. Here, we use genome-wide approaches to reveal genetic stability in the Mtrr model and genome-wide differential DNA methylation in the germline of Mtrr mutant maternal grandfathers. We observe that, while epigenetic reprogramming occurs, wildtype grandprogeny and great grandprogeny exhibit transcriptional changes that correlate with germline methylation defects. One region encompasses the Hira gene, which is misexpressed in embryos for at least three wildtype generations in a manner that distinguishes Hira transcript expression as a biomarker of maternal phenotypic inheritance.
Collapse
|
30
|
Maternal n-3 PUFA deficiency alters uterine artery remodeling and placental epigenome in the mice. J Nutr Biochem 2021; 96:108784. [PMID: 34062269 DOI: 10.1016/j.jnutbio.2021.108784] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 02/20/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023]
Abstract
The maternal n-3 polyunsaturated fatty acid (PUFA) deficiency on decidual vascular structure and angiogenesis in mice placenta was investigated. Namely, we studied uterine artery remodeling, fatty acid metabolism, and placental epigenetic methylation in this animal model. Weanling female Swiss albino mice were fed either alpha-linolenic acid (18:3 n-3, ALA) deficient diets (0.13% energy from ALA) or a sufficient diet (2.26% energy from ALA) throughout the study. The dietary n-3 PUFA deficiency altered uteroplacental morphology and vasculature by reversing luminal to vessel area and increased luminal wall thickness at 8.5-12.5gD. Further, placentas (F0 and F1) showed a significant decrease in the expression of VCAM1, HLAG proteins and an increase in MMP9, KDR expression. The conversion of ALA to long-chain (LC) n-3 PUFAs was significantly decreased in plasma and placenta during the n-3 deficiency state. Reduced n-3 LCPUFAs increased the placental expression of intracellular proteins FABP3, FABP4, and ADRP to compensate decreased availability of these fatty acids in the n-3 deficient mice. The N-3 PUFA deficiency significantly increased the 5-methylcytosine levels in the placenta but not in the liver. The alteration in DNA methylation continued to the next generation in the placental epigenome with augmented expression of DNMT3A and DNMT3B. Our study showed that maternal n-3 PUFA deficiency alters placental vascular architecture and induces epigenetic changes suggesting the importance of n-3 PUFA intake during the development of the fetus. Moreover, the study shows that the placenta is the susceptible target for epigenetic alteration in maternal deficiency n-3 fatty acids.
Collapse
|
31
|
Unraveling the Balance between Genes, Microbes, Lifestyle and the Environment to Improve Healthy Reproduction. Genes (Basel) 2021; 12:genes12040605. [PMID: 33924000 PMCID: PMC8073673 DOI: 10.3390/genes12040605] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/08/2021] [Accepted: 04/17/2021] [Indexed: 12/16/2022] Open
Abstract
Humans’ health is the result of a complex and balanced interplay between genetic factors, environmental stimuli, lifestyle habits, and the microbiota composition. The knowledge about their single contributions, as well as the complex network linking each to the others, is pivotal to understand the mechanisms underlying the onset of many diseases and can provide key information for their prevention, diagnosis and therapy. This applies also to reproduction. Reproduction, involving almost 10% of our genetic code, is one of the most critical human’s functions and is a key element to assess the well-being of a population. The last decades revealed a progressive decline of reproductive outcomes worldwide. As a consequence, there is a growing interest in unveiling the role of the different factors involved in human reproduction and great efforts have been carried out to improve its outcomes. As for many other diseases, it is now clear that the interplay between the underlying genetics, our commensal microbiome, the lifestyle habits and the environment we live in can either exacerbate the outcome or mitigate the adverse effects. Here, we aim to analyze how each of these factors contribute to reproduction highlighting their individual contribution and providing supporting evidence of how to modify their impact and overall contribution to a healthy reproductive status.
Collapse
|
32
|
The regulation mechanisms and the Lamarckian inheritance property of DNA methylation in animals. Mamm Genome 2021; 32:135-152. [PMID: 33860357 DOI: 10.1007/s00335-021-09870-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 04/05/2021] [Indexed: 12/19/2022]
Abstract
DNA methylation is a stable and heritable epigenetic mechanism, of which the main functions are stabilizing the transcription of genes and promoting genetic conservation. In animals, the direct molecular inducers of DNA methylation mainly include histone covalent modification and non-coding RNA, whereas the fundamental regulators of DNA methylation are genetic and environmental factors. As is well known, competition is present everywhere in life systems, and will finally strike a balance that is optimal for the animal's survival and reproduction. The same goes for the regulation of DNA methylation. Genetic and environmental factors, respectively, are responsible for the programmed and plasticity changes of DNA methylation, and keen competition exists between genetically influenced procedural remodeling and environmentally influenced plastic alteration. In this process, genetic and environmental factors collaboratively decide the methylation patterns of corresponding loci. DNA methylation alterations induced by environmental factors can be transgenerationally inherited, and exhibit the characteristic of Lamarckian inheritance. Further research on regulatory mechanisms and the environmental plasticity of DNA methylation will provide strong support for understanding the biological function and evolutionary effects of DNA methylation.
Collapse
|
33
|
Sun J, Zheng W, Liu W, Kou X, Zhao Y, Liang Z, Wang L, Zhang Z, Xiao J, Gao R, Gao S, Jiang C. Differential Transcriptomes and Methylomes of Trophoblast Stem Cells From Naturally-Fertilized and Somatic Cell Nuclear-Transferred Embryos. Front Cell Dev Biol 2021; 9:664178. [PMID: 33869230 PMCID: PMC8047118 DOI: 10.3389/fcell.2021.664178] [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: 02/04/2021] [Accepted: 02/25/2021] [Indexed: 12/05/2022] Open
Abstract
Trophoblast stem cells (TSCs) are critical to mammalian embryogenesis by providing the cell source of the placenta. TSCs can be derived from trophoblast cells. However, the efficiency of TSC derivation from somatic cell nuclear transfer (NT) blastocysts is low. The regulatory mechanisms underlying transcription dynamics and epigenetic landscape remodeling during TSC derivation remain elusive. Here, we derived TSCs from the blastocysts by natural fertilization (NF), NT, and a histone deacetylase inhibitor Scriptaid-treated NT (SNT). Profiling of the transcriptomes across the stages of TSC derivation revealed that fibroblast growth factor 4 (FGF4) treatment resulted in many differentially expressed genes (DEGs) at outgrowth and initiated transcription program for TSC formation. We identified 75 transcription factors (TFs) that are continuously upregulated during NF TSC derivation, whose transcription profiles can infer the time course of NF not NT TSC derivation. Most DEGs in NT outgrowth are rescued in SNT outgrowth. The correct time course of SNT TSC derivation is inferred accordingly. Moreover, these TFs comprise an interaction network important to TSC stemness. Profiling of DNA methylation dynamics showed an extremely low level before FGF4 treatment and gradual increases afterward. FGF4 treatment results in a distinct DNA methylation remodeling process committed to TSC formation. We further identified 1,293 CpG islands (CGIs) whose DNA methylation difference is more than 0.25 during NF TSC derivation. The majority of these CGIs become highly methylated upon FGF4 treatment and remain in high levels. This may create a barrier for lineage commitment to restrict embryonic development, and ensure TSC formation. There exist hundreds of aberrantly methylated CGIs during NT TSC derivation, most of which are corrected during SNT TSC derivation. More than half of the aberrantly methylated CGIs before NT TSC formation are inherited from the donor genome. In contrast, the aberrantly methylated CGIs upon TSC formation are mainly from the highly methylated CGIs induced by FGF4 treatment. Functional annotation indicates that the aberrantly highly methylated CGIs play a role in repressing placenta development genes, etc., related to post-implantation development and maintaining TSC pluripotency. Collectively, our findings provide novel insights into the transcription dynamics, DNA methylation remodeling, and the role of FGF4 during TSC derivation.
Collapse
Affiliation(s)
- Jin Sun
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Weisheng Zheng
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Wenqiang Liu
- Clinical and Translation Research Center of Shanghai First Maternity and Infant Hospital, Tongji University, Shanghai, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, China
| | - Xiaochen Kou
- Clinical and Translation Research Center of Shanghai First Maternity and Infant Hospital, Tongji University, Shanghai, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, China
| | - Yanhong Zhao
- Clinical and Translation Research Center of Shanghai First Maternity and Infant Hospital, Tongji University, Shanghai, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, China
| | - Zehang Liang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Lu Wang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Zihao Zhang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Jing Xiao
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Rui Gao
- Clinical and Translation Research Center of Shanghai First Maternity and Infant Hospital, Tongji University, Shanghai, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, China
| | - Shaorong Gao
- Clinical and Translation Research Center of Shanghai First Maternity and Infant Hospital, Tongji University, Shanghai, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, China
| | - Cizhong Jiang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| |
Collapse
|
34
|
Xia W, Xie W. Rebooting the Epigenomes during Mammalian Early Embryogenesis. Stem Cell Reports 2020; 15:1158-1175. [PMID: 33035464 PMCID: PMC7724468 DOI: 10.1016/j.stemcr.2020.09.005] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/10/2020] [Accepted: 09/11/2020] [Indexed: 02/08/2023] Open
Abstract
Upon fertilization, terminally differentiated gametes are transformed to a totipotent zygote, which gives rise to an embryo. How parental epigenetic memories are inherited and reprogrammed to accommodate parental-to-zygotic transition remains a fundamental question in developmental biology, epigenetics, and stem cell biology. With the rapid advancement of ultra-sensitive or single-cell epigenome analysis methods, unusual principles of epigenetic reprogramming begin to be unveiled. Emerging data reveal that in many species, the parental epigenome undergoes dramatic reprogramming followed by subsequent re-establishment of the embryo epigenome, leading to epigenetic "rebooting." Here, we discuss recent progress in understanding epigenetic reprogramming and their functions during mammalian early development. We also highlight the conserved and species-specific principles underlying diverse regulation of the epigenome in early embryos during evolution.
Collapse
Affiliation(s)
- Weikun Xia
- Center for Stem Cell Biology and Regenerative Medicine, MOE Key Laboratory of Bioinformatics, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Wei Xie
- Center for Stem Cell Biology and Regenerative Medicine, MOE Key Laboratory of Bioinformatics, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China.
| |
Collapse
|
35
|
Menezo Y, Clement P, Clement A, Elder K. Methylation: An Ineluctable Biochemical and Physiological Process Essential to the Transmission of Life. Int J Mol Sci 2020; 21:ijms21239311. [PMID: 33297303 PMCID: PMC7730869 DOI: 10.3390/ijms21239311] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/28/2020] [Accepted: 12/02/2020] [Indexed: 02/07/2023] Open
Abstract
Methylation is a universal biochemical process which covalently adds methyl groups to a variety of molecular targets. It plays a critical role in two major global regulatory mechanisms, epigenetic modifications and imprinting, via methyl tagging on histones and DNA. During reproduction, the two genomes that unite to create a new individual are complementary but not equivalent. Methylation determines the complementary regulatory characteristics of male and female genomes. DNA methylation is executed by methyltransferases that transfer a methyl group from S-adenosylmethionine, the universal methyl donor, to cytosine residues of CG (also designated CpG). Histones are methylated mainly on lysine and arginine residues. The methylation processes regulate the main steps in reproductive physiology: gametogenesis, and early and late embryo development. A focus will be made on the impact of assisted reproductive technology and on the impact of endocrine disruptors (EDCs) via generation of oxidative stress.
Collapse
Affiliation(s)
- Yves Menezo
- Laboratoire CLEMENT, Avenue d’Eylau, 75016 Paris, France; (P.C.); (A.C.)
- Correspondence:
| | - Patrice Clement
- Laboratoire CLEMENT, Avenue d’Eylau, 75016 Paris, France; (P.C.); (A.C.)
| | - Arthur Clement
- Laboratoire CLEMENT, Avenue d’Eylau, 75016 Paris, France; (P.C.); (A.C.)
| | - Kay Elder
- Bourn Hall Clinic, Bourn, Cambridge CB232TN, UK;
| |
Collapse
|
36
|
Uechi L, Jalali M, Wilbur JD, French JL, Jumbe NL, Meaney MJ, Gluckman PD, Karnani N, Sakhanenko NA, Galas DJ. Complex genetic dependencies among growth and neurological phenotypes in healthy children: Towards deciphering developmental mechanisms. PLoS One 2020; 15:e0242684. [PMID: 33270668 PMCID: PMC7714163 DOI: 10.1371/journal.pone.0242684] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 11/09/2020] [Indexed: 11/18/2022] Open
Abstract
The genetic mechanisms of childhood development in its many facets remain largely undeciphered. In the population of healthy infants studied in the Growing Up in Singapore Towards Healthy Outcomes (GUSTO) program, we have identified a range of dependencies among the observed phenotypes of fetal and early childhood growth, neurological development, and a number of genetic variants. We have quantified these dependencies using our information theory-based methods. The genetic variants show dependencies with single phenotypes as well as pleiotropic effects on more than one phenotype and thereby point to a large number of brain-specific and brain-expressed gene candidates. These dependencies provide a basis for connecting a range of variants with a spectrum of phenotypes (pleiotropy) as well as with each other. A broad survey of known regulatory expression characteristics, and other function-related information from the literature for these sets of candidate genes allowed us to assemble an integrated body of evidence, including a partial regulatory network, that points towards the biological basis of these general dependencies. Notable among the implicated loci are RAB11FIP4 (next to NF1), MTMR7 and PLD5, all highly expressed in the brain; DNMT1 (DNA methyl transferase), highly expressed in the placenta; and PPP1R12B and DMD (dystrophin), known to be important growth and development genes. While we cannot specify and decipher the mechanisms responsible for the phenotypes in this study, a number of connections for further investigation of fetal and early childhood growth and neurological development are indicated. These results and this approach open the door to new explorations of early human development.
Collapse
Affiliation(s)
- Lisa Uechi
- Pacific Northwest Research Institute, Seattle, Washington, United States of America
| | - Mahjoubeh Jalali
- Pacific Northwest Research Institute, Seattle, Washington, United States of America
| | - Jayson D. Wilbur
- Metrum Research Group, Tariffville, CT, United States of America
| | | | - N. L. Jumbe
- Pharmactuarials LLC, Mountain View, CA, United States of America
| | - Michael J. Meaney
- Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Child and Brain Development Program, Canadian Institute for Advanced Research (CIFAR) Institute, Toronto, Canada
| | - Peter D. Gluckman
- Centre for Human Evolution, Adaptation and Disease, Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Neerja Karnani
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Brenner Centre for Molecular Medicine, National University of Singapore, Singapore, Singapore
| | - Nikita A. Sakhanenko
- Pacific Northwest Research Institute, Seattle, Washington, United States of America
- * E-mail: (DJG); (NAS)
| | - David J. Galas
- Pacific Northwest Research Institute, Seattle, Washington, United States of America
- * E-mail: (DJG); (NAS)
| | | |
Collapse
|
37
|
Castillo‐Fernandez J, Herrera‐Puerta E, Demond H, Clark SJ, Hanna CW, Hemberger M, Kelsey G. Increased transcriptome variation and localised DNA methylation changes in oocytes from aged mice revealed by parallel single-cell analysis. Aging Cell 2020; 19:e13278. [PMID: 33201571 PMCID: PMC7744954 DOI: 10.1111/acel.13278] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/27/2020] [Accepted: 10/18/2020] [Indexed: 01/08/2023] Open
Abstract
Advancing maternal age causes a progressive reduction in fertility. The decline in developmental competence of the oocyte with age is likely to be a consequence of multiple contributory factors. Loss of epigenetic quality of the oocyte could impair early developmental events or programme adverse outcomes in offspring that manifest only later in life. Here, we undertake joint profiling of the transcriptome and DNA methylome of individual oocytes from reproductively young and old mice undergoing natural ovulation. We find reduced complexity as well as increased variance in the transcriptome of oocytes from aged females. This transcriptome heterogeneity is reflected in the identification of discrete sub-populations. Oocytes with a transcriptome characteristic of immature chromatin configuration (NSN) clustered into two groups: one with reduced developmental competence, as indicated by lower expression of maternal effect genes, and one with a young-like transcriptome. Oocytes from older females had on average reduced CpG methylation, but the characteristic bimodal methylation landscape of the oocyte was preserved. Germline differentially methylated regions of imprinted genes were appropriately methylated irrespective of age. For the majority of differentially expressed transcripts, the absence of correlated methylation changes suggests a post-transcriptional basis for most age-related effects on the transcriptome. However, we did find differences in gene body methylation at which there were corresponding changes in gene expression, indicating age-related effects on transcription that translate into methylation differences. Interestingly, oocytes varied in expression and methylation of these genes, which could contribute to variable competence of oocytes or penetrance of maternal age-related phenotypes in offspring.
Collapse
Affiliation(s)
| | - Erika Herrera‐Puerta
- Epigenetics ProgrammeBabraham InstituteCambridgeUK
- Science and Biotechnology Faculty, Biology ProgramCES UniversityMedellinColombia
| | | | | | - Courtney W. Hanna
- Epigenetics ProgrammeBabraham InstituteCambridgeUK
- Centre for Trophoblast ResearchUniversity of CambridgeCambridgeUK
| | - Myriam Hemberger
- Epigenetics ProgrammeBabraham InstituteCambridgeUK
- Centre for Trophoblast ResearchUniversity of CambridgeCambridgeUK
- Departments of Biochemistry & Molecular Biology and Medical GeneticsCumming School of MedicineUniversity of CalgaryCalgaryALCanada
- Alberta Children’s Hospital Research InstituteUniversity of CalgaryCalgaryALCanada
| | - Gavin Kelsey
- Epigenetics ProgrammeBabraham InstituteCambridgeUK
- Centre for Trophoblast ResearchUniversity of CambridgeCambridgeUK
| |
Collapse
|
38
|
Hernández-Vargas P, Muñoz M, Domínguez F. Identifying biomarkers for predicting successful embryo implantation: applying single to multi-OMICs to improve reproductive outcomes. Hum Reprod Update 2020; 26:264-301. [PMID: 32096829 DOI: 10.1093/humupd/dmz042] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 10/08/2019] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Successful embryo implantation is a complex process that requires the coordination of a series of events, involving both the embryo and the maternal endometrium. Key to this process is the intricate cascade of molecular mechanisms regulated by endocrine, paracrine and autocrine modulators of embryonic and maternal origin. Despite significant progress in ART, implantation failure still affects numerous infertile couples worldwide and fewer than 10% of embryos successfully implant. Improved selection of both the viable embryos and the optimal endometrial phenotype for transfer remains crucial to enhancing implantation chances. However, both classical morphological embryo selection and new strategies incorporated into clinical practice, such as embryonic genetic analysis, morphokinetics or ultrasound endometrial dating, remain insufficient to predict successful implantation. Additionally, no techniques are widely applied to analyse molecular signals involved in the embryo-uterine interaction. More reliable biological markers to predict embryo and uterine reproductive competence are needed to improve pregnancy outcomes. Recent years have seen a trend towards 'omics' methods, which enable the assessment of complete endometrial and embryonic molecular profiles during implantation. Omics have advanced our knowledge of the implantation process, identifying potential but rarely implemented biomarkers of successful implantation. OBJECTIVE AND RATIONALE Differences between the findings of published omics studies, and perhaps because embryonic and endometrial molecular signatures were often not investigated jointly, have prevented firm conclusions being reached. A timely review summarizing omics studies on the molecular determinants of human implantation in both the embryo and the endometrium will help facilitate integrative and reliable omics approaches to enhance ART outcomes. SEARCH METHODS In order to provide a comprehensive review of the literature published up to September 2019, Medline databases were searched using keywords pertaining to omics, including 'transcriptome', 'proteome', 'secretome', 'metabolome' and 'expression profiles', combined with terms related to implantation, such as 'endometrial receptivity', 'embryo viability' and 'embryo implantation'. No language restrictions were imposed. References from articles were also used for additional literature. OUTCOMES Here we provide a complete summary of the major achievements in human implantation research supplied by omics approaches, highlighting their potential to improve reproductive outcomes while fully elucidating the implantation mechanism. The review highlights the existence of discrepancies among the postulated biomarkers from studies on embryo viability or endometrial receptivity, even using the same omic analysis. WIDER IMPLICATIONS Despite the huge amount of biomarker information provided by omics, we still do not have enough evidence to link data from all omics with an implantation outcome. However, in the foreseeable future, application of minimally or non-invasive omics tools, together with a more integrative interpretation of uniformly collected data, will help to overcome the difficulties for clinical implementation of omics tools. Omics assays of the embryo and endometrium are being proposed or already being used as diagnostic tools for personalised single-embryo transfer in the most favourable endometrial environment, avoiding the risk of multiple pregnancies and ensuring better pregnancy rates.
Collapse
Affiliation(s)
- Purificación Hernández-Vargas
- IVI-RMA Alicante, Innovation. Avda. de Denia 111, 03015 Alicante, Spain.,Fundación IVI, Innovation-IIS La Fe, Avda. Fernando Abril Martorell 106, Torre A, 1° 1.23, 46026 Valencia, Spain
| | - Manuel Muñoz
- IVI-RMA Alicante, Innovation. Avda. de Denia 111, 03015 Alicante, Spain.,Fundación IVI, Innovation-IIS La Fe, Avda. Fernando Abril Martorell 106, Torre A, 1° 1.23, 46026 Valencia, Spain
| | - Francisco Domínguez
- Fundación IVI, Innovation-IIS La Fe, Avda. Fernando Abril Martorell 106, Torre A, 1° 1.23, 46026 Valencia, Spain
| |
Collapse
|
39
|
Leseva MN, Binder AM, Ponsonby AL, Vuillermin P, Saffery R, Michels KB. Differential gene expression and limited epigenetic dysregulation at the materno-fetal interface in preeclampsia. Hum Mol Genet 2020; 29:335-350. [PMID: 31868881 DOI: 10.1093/hmg/ddz287] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 11/26/2019] [Indexed: 12/31/2022] Open
Abstract
Despite the many advances made in the diagnosis and management of preeclampsia, this syndrome remains a leading cause of maternal mortality and life-long morbidity, as well as adverse fetal outcomes. Successful prediction and therapeutic intervention require an improved understanding of the molecular mechanisms, which underlie preeclampsia pathophysiology. We have used an integrated approach to discover placental genetic and epigenetic markers of preeclampsia and validated our findings in an independent cohort of women. We observed the microRNA, MIR138, to be upregulated in singleton preeclamptic placentas; however, this appears to be a female infant sex-specific effect. We did not identify any significant differentially methylated positions (DMPs) in singleton pregnancies, indicating that DNA methylation changes in mild forms of the disease are likely limited. However, we identified infant sex-specific preeclampsia-associated differentially methylated regions among singletons. Disease-associated DMPs were more obvious in a limited sampling of twin pregnancies. Interestingly, 2 out of the 10 most significant changes in methylation over larger regions overlap between singletons and twins and correspond to NAPRT1 and ZNF417.
Collapse
Affiliation(s)
- Milena N Leseva
- Institute for Prevention and Cancer Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg 79110, Germany
| | - Alexandra M Binder
- Department of Epidemiology, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Anne-Louise Ponsonby
- Discovery Theme, Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia.,Cell Biology Theme, The Murdoch Children's Research Institute, Royal Children's Hospital, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Peter Vuillermin
- Cell Biology Theme, The Murdoch Children's Research Institute, Royal Children's Hospital, University of Melbourne, Parkville, Victoria 3052, Australia.,School of Medicine, Deakin University, Geelong, Victoria 3220, Australia.,Child Health Research Unit, Barwon Health, Geelong, Victoria 3220, Australia
| | - Richard Saffery
- Cell Biology Theme, The Murdoch Children's Research Institute, Royal Children's Hospital, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Karin B Michels
- Institute for Prevention and Cancer Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg 79110, Germany.,Department of Epidemiology, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA 90095, USA
| | | |
Collapse
|
40
|
Morgan HL, Aljumah A, Rouillon C, Watkins AJ. Paternal low protein diet and the supplementation of methyl-donors impact fetal growth and placental development in mice. Placenta 2020; 103:124-133. [PMID: 33120048 PMCID: PMC7907633 DOI: 10.1016/j.placenta.2020.10.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 10/14/2020] [Accepted: 10/16/2020] [Indexed: 12/24/2022]
Abstract
Introduction Paternal low-protein diet can alter sperm methylation status, fetal growth and program offspring ill-health, however its impact on the placenta remains poorly defined. Here we examine the influence paternal low-protein diet has on fetal and placental development and the additional impact of supplementary methyl-donors on fetoplacental physiology. Methods Male C57BL/6J mice were fed a control normal protein diet (NPD; 18% protein), a low-protein diet (LPD; 9% protein) or LPD with methyl-donor supplementation (MD-LPD; choline chloride, betaine, methionine, folic acid, vitamin B12) for a minimum of 8 weeks. Males were mated with 8–11 week old female C57BL/6J mice and fetal and placental tissue collected on embryonic day 17.5. Results Paternal LPD was associated with increased fetal weights compared to NPD and MD-LPD with 22% fetuses being above the 90th centile for fetal weight. However, LPD and MD-LPD placental weights were reduced when compared to NPD. Placentas from LPD fathers demonstrated a reduced junctional zone area and reduced free-fatty acid content. MD-LPD placentas did not mirror these finding, demonstrating an increased chorion area, a reduction in junctional-specific glycogen staining and reduced placental Dnmt3bexpression, none of which were apparent in either NPD or LPD placentas. Discussion A sub-optimal paternal diet can influence fetal growth and placental development, and dietary methyl-donor supplementation alters placental morphology and gene expression differentially to that observed with LPD alone. Understanding how paternal diet and micro-nutrient supplementation influence placental development is crucial for determining connections between paternal well-being and future offspring health. Paternal low protein diet (LPD) increased late gestation fetal weight. Supplementing the LPD with methyl donors (MD-LPD) normalised fetal weight. Placental weight and morphology are altered by both LPD and MD-LPD. Placental metabolite content and gene expression were perturbed by paternal LPD and MD-LPD.
Collapse
Affiliation(s)
- Hannah L Morgan
- Division of Child Health, Obstetrics and Gynaecology, Faculty of Medicine, University of Nottingham, Nottingham, NG7 2UH, UK.
| | - Arwa Aljumah
- Division of Child Health, Obstetrics and Gynaecology, Faculty of Medicine, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Charlène Rouillon
- INRAE, Fish Physiology and Genomics, Bat 16A, Campus de Beaulieu, Rennes, France
| | - Adam J Watkins
- Division of Child Health, Obstetrics and Gynaecology, Faculty of Medicine, University of Nottingham, Nottingham, NG7 2UH, UK; Aston Research Centre for Healthy Ageing, School of Life and Health Sciences, Aston University, Birmingham, B4 7ET, UK(1)
| |
Collapse
|
41
|
Jia Y, Xie H, Zhang J, Ying H. Induction of TGF-β receptor I expression in a DNA methylation-independent manner mediated by DNMT3A downregulation is involved in early-onset severe preeclampsia. FASEB J 2020; 34:13224-13238. [PMID: 32794622 DOI: 10.1096/fj.202000253rr] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 07/09/2020] [Accepted: 07/09/2020] [Indexed: 02/06/2023]
Abstract
Preeclampsia, especially early-onset severe preeclampsia is one of the leading causes of maternal and fetal morbidity and mortality. Although it has been well known that the pathophysiology of early-onset severe preeclampsia begins with abnormal placentation and aberrant activation of TGF-β signaling inhibits trophoblast cell invasion, the mechanisms underlying dysregulation of TGF-β signaling in early-onset severe preeclampsia remain elusive to date. Here, we revealed that induction of TGFBR1/TGF-β signaling mediated by DNMT3A downregulation plays a critical role in early-onset severe preeclampsia. Our results show that DNMT3A downregulation elevates TGFBR1 expression in trophoblast cells. Moreover, inhibition of TGFBR1 and TGF-β/Smad signaling can rescue the deficiencies of trophoblast cell migration and invasion caused by DNMT3A knockdown. Mechanistically, DNMT3A suppresses the transcription of TGFBR1 through recruiting EZH2 to its promoter but not changing DNA methylation of TGFBR1 promoter. In human samples, we detected lowly expressed DNMT3A, highly expressed TGFBR1 and hyperactivation of TGF-β/Smad signaling in decidua-embedded extravillous trophoblasts in early-onset severe preeclampsia, which provides the clinical evidence for the correlation between DNMT3A and TGFBR1. Collectively, our findings demonstrate that DNA methylation-independent induction of TGFBR1 mediated by DNMT3A downregulation is relevant to the development of early-onset severe preeclampsia.
Collapse
Affiliation(s)
- Yuanhui Jia
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Han Xie
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jiqin Zhang
- Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Hao Ying
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
42
|
The role and mechanisms of DNA methylation in the oocyte. Essays Biochem 2020; 63:691-705. [PMID: 31782490 PMCID: PMC6923320 DOI: 10.1042/ebc20190043] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/29/2019] [Accepted: 10/29/2019] [Indexed: 12/27/2022]
Abstract
Epigenetic information in the mammalian oocyte has the potential to be transmitted to the next generation and influence gene expression; this occurs naturally in the case of imprinted genes. Therefore, it is important to understand how epigenetic information is patterned during oocyte development and growth. Here, we review the current state of knowledge of de novo DNA methylation mechanisms in the oocyte: how a distinctive gene-body methylation pattern is created, and the extent to which the DNA methylation machinery reads chromatin states. Recent epigenomic studies building on advances in ultra-low input chromatin profiling methods, coupled with genetic studies, have started to allow a detailed interrogation of the interplay between DNA methylation establishment and chromatin states; however, a full mechanistic description awaits.
Collapse
|
43
|
Huo Y, Yan ZQ, Yuan P, Qin M, Kuo Y, Li R, Yan LY, Feng HL, Qiao J. Single-cell DNA methylation sequencing reveals epigenetic alterations in mouse oocytes superovulated with different dosages of gonadotropins. Clin Epigenetics 2020; 12:75. [PMID: 32487258 PMCID: PMC7268365 DOI: 10.1186/s13148-020-00866-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 05/19/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Epigenetic abnormalities caused by superovulation have recently attracted increasing attention. Superovulation with exogenous hormones may prevent oocytes from establishing an appropriate epigenetic state, and this effect may extend to the methylation programming in preimplantation embryos, as de novo DNA methylation is a function of developmental stage of follicles and oocyte size. Follicle-stimulating hormone (FSH) and human menopausal gonadotropin (hMG) are common gonadotropins used for superovulation, and appropriate concentrations of these gonadotropins might be necessary. However, no systematic study on the effects of DNA methylation alterations in oocytes associated with superovulation with different dosages of FSH/hMG at the single-cell level has yet been reported. In the current study, different dosages of FSH/hMG combined with human chorionic gonadotropin (hCG) were used in female mice to generate experimental groups, while naturally matured oocytes and oocytes superovulated with only hCG were respectively used as controls. Single-cell level DNA methylation sequencing was carried out on all these matured oocytes. RESULTS In this study, we revealed that the genome-wide methylation pattern and CG methylation level of the maternal imprinting control regions of all mature oocytes were globally conserved and stable. However, methylation alterations associated with superovulation were found at a specific set of loci, and the differentially methylated regions (DMRs) mainly occurred in regions other than promoters. Furthermore, some of the annotated genes in the DMRs were involved in biological processes such as glucose metabolism, nervous system development, cell cycle, cell proliferation, and embryo implantation and were altered in all dosages of FSH/hMG group (for example, Gfod2 and SYF2). Other genes were impaired only after high gonadotropin dosages (for instance, Sox17 and Phactr4). CONCLUSIONS In conclusion, the current study addressed the effects of superovulation on DNA methylation from the perspective of different dosages of gonadotropins at the single-cell level. We found that the genome-wide DNA methylation landscape was globally preserved irrespective of superovulation or of the kind and dosage of gonadotropins used, whereas the methylation alterations associated with superovulation occurred at a specific set of loci. These observed effects reflect that superovulation recruits oocytes that would not normally be ovulated or that have not undergone complete epigenetic maturation. Our results provide an important reference for the safety assessment of superovulation with different dosages of gonadotropins. However, it should be noted that this study has some limitations, as the sample number and library coverage of analyzed oocytes were relatively low. Future studies with larger sample sizes and high-coverage libraries that examine the effects of superovulation on embryo development and offspring health as well as the underlying mechanisms are still needed.
Collapse
Affiliation(s)
- Ying Huo
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, No. 49 North HuaYuan Road, Hai Dian District, Beijing, 100191, China.,Key Laboratory of Assisted Reproduction, Ministry of Education, No. 49 North HuaYuan Road, Hai Dian District, Beijing, 100191, China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, No. 49 North HuaYuan Road, Hai Dian District, Beijing, 100191, China.,Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, No. 38 XueYuan Road, Haidian District, Beijing, 100191, China
| | - Zhi Qiang Yan
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, No. 49 North HuaYuan Road, Hai Dian District, Beijing, 100191, China.,Key Laboratory of Assisted Reproduction, Ministry of Education, No. 49 North HuaYuan Road, Hai Dian District, Beijing, 100191, China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, No. 49 North HuaYuan Road, Hai Dian District, Beijing, 100191, China.,Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
| | - Peng Yuan
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, No. 49 North HuaYuan Road, Hai Dian District, Beijing, 100191, China.,Key Laboratory of Assisted Reproduction, Ministry of Education, No. 49 North HuaYuan Road, Hai Dian District, Beijing, 100191, China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, No. 49 North HuaYuan Road, Hai Dian District, Beijing, 100191, China
| | - Meng Qin
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, No. 49 North HuaYuan Road, Hai Dian District, Beijing, 100191, China.,Key Laboratory of Assisted Reproduction, Ministry of Education, No. 49 North HuaYuan Road, Hai Dian District, Beijing, 100191, China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, No. 49 North HuaYuan Road, Hai Dian District, Beijing, 100191, China
| | - Ying Kuo
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, No. 49 North HuaYuan Road, Hai Dian District, Beijing, 100191, China.,Key Laboratory of Assisted Reproduction, Ministry of Education, No. 49 North HuaYuan Road, Hai Dian District, Beijing, 100191, China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, No. 49 North HuaYuan Road, Hai Dian District, Beijing, 100191, China
| | - Rong Li
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, No. 49 North HuaYuan Road, Hai Dian District, Beijing, 100191, China.,Key Laboratory of Assisted Reproduction, Ministry of Education, No. 49 North HuaYuan Road, Hai Dian District, Beijing, 100191, China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, No. 49 North HuaYuan Road, Hai Dian District, Beijing, 100191, China.,National Clinical Research Center of Obstetrics and Gynecology, No. 49 North HuaYuan Road, Hai Dian District, Beijing, 100191, China
| | - Li Ying Yan
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, No. 49 North HuaYuan Road, Hai Dian District, Beijing, 100191, China.,Key Laboratory of Assisted Reproduction, Ministry of Education, No. 49 North HuaYuan Road, Hai Dian District, Beijing, 100191, China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, No. 49 North HuaYuan Road, Hai Dian District, Beijing, 100191, China.,National Clinical Research Center of Obstetrics and Gynecology, No. 49 North HuaYuan Road, Hai Dian District, Beijing, 100191, China
| | - Huai Liang Feng
- The New York Fertility Center, New York Hospital Queens, Weill Medical College of Cornell University, New York, NY, USA.
| | - Jie Qiao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, No. 49 North HuaYuan Road, Hai Dian District, Beijing, 100191, China. .,Key Laboratory of Assisted Reproduction, Ministry of Education, No. 49 North HuaYuan Road, Hai Dian District, Beijing, 100191, China. .,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, No. 49 North HuaYuan Road, Hai Dian District, Beijing, 100191, China. .,Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, No. 38 XueYuan Road, Haidian District, Beijing, 100191, China. .,National Clinical Research Center of Obstetrics and Gynecology, No. 49 North HuaYuan Road, Hai Dian District, Beijing, 100191, China.
| |
Collapse
|
44
|
Senner CE, Chrysanthou S, Burge S, Lin HY, Branco MR, Hemberger M. TET1 and 5-Hydroxymethylation Preserve the Stem Cell State of Mouse Trophoblast. Stem Cell Reports 2020; 15:1301-1316. [PMID: 32442533 PMCID: PMC7724466 DOI: 10.1016/j.stemcr.2020.04.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 02/06/2023] Open
Abstract
The ten-eleven translocation factor TET1 and its conferred epigenetic modification 5-hydroxymethylcytosine (5hmC) have important roles in maintaining the pluripotent state of embryonic stem cells (ESCs). We previously showed that TET1 is also essential to maintain the stem cell state of trophoblast stem cells (TSCs). Here, we establish an integrated panel of absolute 5hmC levels, genome-wide DNA methylation and hydroxymethylation patterns, transcriptomes, and TET1 chromatin occupancy in TSCs and differentiated trophoblast cells. We show that the combined presence of 5-methylcytosine (5mC) and 5hmC correlates with transcriptional activity of associated genes. Hypoxia can slow down the global loss of 5hmC that occurs upon differentiation of TSCs. Notably, unlike in ESCs and epiblast cells, most TET1-bound regions overlap with active chromatin marks and TFAP2C binding sites and demarcate putative trophoblast enhancer regions. These chromatin modification and occupancy patterns are highly informative to identify novel candidate regulators of the TSC state. 5hmC to 5mC ratios correlate with gene activity in TS cells TS cell differentiation-associated loss of 5hmC is slowed down in hypoxia TET1 binding in TS cells forms long-range interactions with key trophoblast genes Intergenic TET1 binding sites in TS cells demarcate putative trophoblast enhancers
Collapse
Affiliation(s)
- Claire E Senner
- Epigenetics Programme, Babraham Institute, Cambridge CB22 3AT, UK; Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Tennis Court Road, Cambridge CB2 3EG, UK.
| | - Stephanie Chrysanthou
- Epigenetics Programme, Babraham Institute, Cambridge CB22 3AT, UK; Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Sarah Burge
- Epigenetics Programme, Babraham Institute, Cambridge CB22 3AT, UK
| | - Hai-Yan Lin
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Miguel R Branco
- Blizard Institute, Barts and the London School of Medicine and Dentistry, QMUL, London E1 2AT, UK
| | - Myriam Hemberger
- Epigenetics Programme, Babraham Institute, Cambridge CB22 3AT, UK; Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Tennis Court Road, Cambridge CB2 3EG, UK; Departments of Biochemistry & Molecular Biology and Medical Genetics, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive N.W., Calgary, Alberta T2N 4N1, Canada; Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive N.W., Calgary, Alberta, T2N 4N1, Canada.
| |
Collapse
|
45
|
Tong J, Niu Y, Chen ZJ, Zhang C. Comparison of the transcriptional profile in the decidua of early-onset and late-onset pre-eclampsia. J Obstet Gynaecol Res 2020; 46:1055-1066. [PMID: 32281216 DOI: 10.1111/jog.14257] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 02/28/2020] [Accepted: 03/16/2020] [Indexed: 12/14/2022]
Abstract
AIM To compare early-onset pre-eclampsia (EOPE) and late-onset pre-eclampsia (LOPE) and provide insight into the pathophysiology of pre-eclampsia (PE). METHODS Our recent work compared the transcriptomics in decidua of EOPE, LOPE and normal pregnancies (NP). RESULTS We found there are a significant number of genes uniquely expressed in the decidua of EOPE and LOPE comparing with NP. Moreover, EOPE and LOPE have their distinct profiles. Unique EOPE-associated genes were mainly involved in apoptosis related pathways such as 'apoptosis' and 'Ras signaling pathway'. PIK3CB and BCL-2 are the core regulatory genes in EOPE decidua, their abnormal expression caused decidual abnormal apoptosis which is relevant to the pathogenesis of EOPE. Whereas, LOPE is a more complicated entity which has more special LOPE-associated genes involved in decidua differentiation, especially in 'gap junction pathway', 'vascular smooth muscle contraction' and 'long-term depression'. PIK3CB, FLT1, CBLC and ITGA7 are the core regulatory genes differentially expressed in EOPE decidua comparing with LOPE. CONCLUSION In brief, the different decidual transcriptomics of EOPE and LOPE may correlate with their different etiology. These findings highlight the complex pathophysiology of PE and provide potential targets for a new treatment strategy in patients with PE.
Collapse
Affiliation(s)
- Jing Tong
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Yichao Niu
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Cong Zhang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China.,Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Ji'nan, China
| |
Collapse
|
46
|
Abstract
The mammalian genome experiences profound setting and resetting of epigenetic patterns during the life-course. This is understood best for DNA methylation: the specification of germ cells, gametogenesis, and early embryo development are characterised by phases of widespread erasure and rewriting of methylation. While mitigating against intergenerational transmission of epigenetic information, these processes must also ensure correct genomic imprinting that depends on faithful and long-term memory of gamete-derived methylation states in the next generation. This underscores the importance of understanding the mechanisms of methylation programming in the germline.
De novo methylation in the oocyte is of particular interest because of its intimate association with transcription, which results in a bimodal methylome unique amongst mammalian cells. Moreover, this methylation landscape is entirely set up in a non-dividing cell, making the oocyte a fascinating model system in which to explore mechanistic determinants of methylation. Here, we summarise current knowledge on the oocyte DNA methylome and how it is established, focussing on recent insights from knockout models in the mouse that explore the interplay between methylation and chromatin states. We also highlight some remaining paradoxes and enigmas, in particular the involvement of non-nuclear factors for correct
de novo methylation.
Collapse
Affiliation(s)
- Hannah Demond
- Epigenetics Programme, The Babraham Institute, Cambridge, UK
| | - Gavin Kelsey
- Epigenetics Programme, The Babraham Institute, Cambridge, UK.,Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| |
Collapse
|
47
|
Saenz-de-Juano MD, Ivanova E, Billooye K, Herta AC, Smitz J, Kelsey G, Anckaert E. Genome-wide assessment of DNA methylation in mouse oocytes reveals effects associated with in vitro growth, superovulation, and sexual maturity. Clin Epigenetics 2019; 11:197. [PMID: 31856890 PMCID: PMC6923880 DOI: 10.1186/s13148-019-0794-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 12/03/2019] [Indexed: 12/27/2022] Open
Abstract
Background In vitro follicle culture (IFC), as applied in the mouse system, allows the growth and maturation of a large number of immature preantral follicles to become mature and competent oocytes. In the human oncofertility clinic, there is increasing interest in developing this technique as an alternative to ovarian cortical tissue transplantation and to preserve the fertility of prepubertal cancer patients. However, the effect of IFC and hormonal stimulation on DNA methylation in the oocyte is not fully known, and there is legitimate concern over epigenetic abnormalities that could be induced by procedures applied during assisted reproductive technology (ART). Results In this study, we present the first genome-wide analysis of DNA methylation in MII oocytes obtained after natural ovulation, after IFC and after superovulation. We also performed a comparison between prepubertal and adult hormonally stimulated oocytes. Globally, the distinctive methylation landscape of oocytes, comprising alternating hyper- and hypomethylated domains, is preserved irrespective of the procedure. The conservation of methylation extends to the germline differential methylated regions (DMRs) of imprinted genes, necessary for their monoallelic expression in the embryo. However, we do detect specific, consistent, and coherent differences in DNA methylation in IFC oocytes, and between oocytes obtained after superovulation from prepubertal compared with sexually mature females. Several methylation differences span entire transcription units. Among these, we found alterations in Tcf4, Sox5, Zfp521, and other genes related to nervous system development. Conclusions Our observations show that IFC is associated with altered methylation at specific set of loci. DNA methylation of superovulated prepubertal oocytes differs from that of superovulated adult oocytes, whereas oocytes from superovulated adult females differ very little from naturally ovulated oocytes. Importantly, we show that regions other than imprinted gDMRs are susceptible to methylation changes associated with superovulation, IFC, and/or sexual immaturity in mouse oocytes. Our results provide an important reference for the use of in vitro growth and maturation of oocytes, particularly from prepubertal females, in assisted reproductive treatments or fertility preservation.
Collapse
Affiliation(s)
- Maria Desemparats Saenz-de-Juano
- Follicle Biology Laboratory (FOBI), UZ Brussel, Vrije Universiteit Brussel, Laarbeeklaan, Brussels, Belgium.,Present Address: Animal Physiology, Institute of Agricultural Sciences, ETH Zurich, Zurich, Switzerland
| | - Elena Ivanova
- Epigenetics Programme, Babraham Institute, Cambridge, CB22 3AT, UK
| | - Katy Billooye
- Follicle Biology Laboratory (FOBI), UZ Brussel, Vrije Universiteit Brussel, Laarbeeklaan, Brussels, Belgium
| | - Anamaria-Cristina Herta
- Follicle Biology Laboratory (FOBI), UZ Brussel, Vrije Universiteit Brussel, Laarbeeklaan, Brussels, Belgium
| | - Johan Smitz
- Follicle Biology Laboratory (FOBI), UZ Brussel, Vrije Universiteit Brussel, Laarbeeklaan, Brussels, Belgium
| | - Gavin Kelsey
- Epigenetics Programme, Babraham Institute, Cambridge, CB22 3AT, UK.,Centre for Trophoblast Research, University of Cambridge, Cambridge, CB2 3EG, UK
| | - Ellen Anckaert
- Follicle Biology Laboratory (FOBI), UZ Brussel, Vrije Universiteit Brussel, Laarbeeklaan, Brussels, Belgium.
| |
Collapse
|
48
|
Hemberger M, Hanna CW, Dean W. Mechanisms of early placental development in mouse and humans. Nat Rev Genet 2019; 21:27-43. [PMID: 31534202 DOI: 10.1038/s41576-019-0169-4] [Citation(s) in RCA: 298] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/09/2019] [Indexed: 02/08/2023]
Abstract
The importance of the placenta in supporting mammalian development has long been recognized, but our knowledge of the molecular, genetic and epigenetic requirements that underpin normal placentation has remained remarkably under-appreciated. Both the in vivo mouse model and in vitro-derived murine trophoblast stem cells have been invaluable research tools for gaining insights into these aspects of placental development and function, with recent studies starting to reshape our view of how a unique epigenetic environment contributes to trophoblast differentiation and placenta formation. These advances, together with recent successes in deriving human trophoblast stem cells, open up new and exciting prospects in basic and clinical settings that will help deepen our understanding of placental development and associated disorders of pregnancy.
Collapse
Affiliation(s)
- Myriam Hemberger
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Canada. .,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Canada. .,Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Canada. .,Epigenetics Programme, The Babraham Institute, Babraham Research Campus, Cambridge, UK. .,Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
| | - Courtney W Hanna
- Epigenetics Programme, The Babraham Institute, Babraham Research Campus, Cambridge, UK.,Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Wendy Dean
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Canada. .,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Canada. .,Epigenetics Programme, The Babraham Institute, Babraham Research Campus, Cambridge, UK. .,Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, Canada.
| |
Collapse
|
49
|
5-Methyltetrahydrofolate reduces blood homocysteine level significantly in C677T methyltetrahydrofolate reductase single-nucleotide polymorphism carriers consulting for infertility. J Gynecol Obstet Hum Reprod 2019; 49:101622. [PMID: 31446167 DOI: 10.1016/j.jogoh.2019.08.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 05/27/2019] [Accepted: 08/20/2019] [Indexed: 10/26/2022]
Abstract
PURPOSE Methyltetrahydrofolate reductase (MTHFR) C677T (ala222Val) is a single-nucleotide polymorphism (SNP) that affects the formation of 5-methyltetrahydrofolate (5-MTHF), the active folate that allows the recycling of homocysteine (Hcy) to Methionine. Hcy is at the epicentre of oxidative stress and DNA methylation errors. This SNP often increases the circulating Hcy levels and consequently reduces the methylation process, which is involved in the epigenesis and imprinting of markings in gametes. This study aimed to investigate decreases in Hcy levels in MTHFR SNP carriers. PROCEDURE Eighty-nine couples with fertility problems for at least 3 years were included in this program. The women were systematically tested for the MTHFR C 677T isoform. If the woman tested positive, testing of the male partner was proposed. All the carriers had well-controlled blood Hcy levels before and after treatment (600μg of 5-MTHF/day, with a backup of one carbon cycle during at least 3 months). FINDINGS As expected, the circulating Hcy level was higher in the homozygous patients than in the heterozygous and wild-type patients. The treatments caused a significant decrease of the circulating Hcy in the SNP carriers group. CONCLUSIONS Couples with a long history of infertility should be analysed for MTHFR SNP and homocysteine and should be treated with physiological doses of 5-MTHF instead of high doses of folic acid.
Collapse
|
50
|
Li R, Liang F, Li M, Zou D, Sun S, Zhao Y, Zhao W, Bao Y, Xiao J, Zhang Z. MethBank 3.0: a database of DNA methylomes across a variety of species. Nucleic Acids Res 2019; 46:D288-D295. [PMID: 29161430 PMCID: PMC5753180 DOI: 10.1093/nar/gkx1139] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 10/30/2017] [Indexed: 12/11/2022] Open
Abstract
MethBank (http://bigd.big.ac.cn/methbank) is a database that integrates high-quality DNA methylomes across a variety of species and provides an interactive browser for visualization of methylation data. Here, we present an updated implementation of MethBank (version 3.0) by incorporating more DNA methylomes from multiple species and equipping with more enhanced functionalities for data annotation and more friendly web interfaces for data presentation, search and visualization. MethBank 3.0 features large-scale integration of high-quality methylomes, involving 34 consensus reference methylomes derived from a large number of human samples, 336 single-base resolution methylomes from different developmental stages and/or tissues of five plants, and 18 single-base resolution methylomes from gametes and early embryos at multiple stages of two animals. Additionally, it is enhanced by improving the functionalities for data annotation, which accordingly enables systematic identification of methylation sites closely associated with age, sites with constant methylation levels across different ages, differentially methylated promoters, age-specific differentially methylated cytosines/regions, and methylated CpG islands. Moreover, MethBank provides tools to estimate human methylation age online and to identify differentially methylated promoters, respectively. Taken together, MethBank is upgraded with significant improvements and advances over the previous version, which is of great help for deciphering DNA methylation regulatory mechanisms for epigenetic studies.
Collapse
Affiliation(s)
- Rujiao Li
- BIG Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Fang Liang
- BIG Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Mengwei Li
- BIG Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China.,CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Dong Zou
- BIG Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China.,CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Shixiang Sun
- BIG Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China.,CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yongbing Zhao
- Lymphocyte Nuclear Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wenming Zhao
- BIG Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yiming Bao
- BIG Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China.,CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jingfa Xiao
- BIG Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China.,CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100049, China.,Collaborative Innovation Center of Genetics and Development, Fudan University, Shanghai 200438, China
| | - Zhang Zhang
- BIG Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China.,CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100049, China.,Collaborative Innovation Center of Genetics and Development, Fudan University, Shanghai 200438, China
| |
Collapse
|