1
|
Zhang C, Guo Y, Liu Y, Liu K, Hu W, Wang H. Sperm miR-142-3p Reprogramming Mediates Paternal Pre-Pregnancy Caffeine Exposure-Induced Non-Alcoholic Steatohepatitis in Male Offspring Rats. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405592. [PMID: 39291441 PMCID: PMC11558112 DOI: 10.1002/advs.202405592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/28/2024] [Indexed: 09/19/2024]
Abstract
Numerous studies have suggested a strong association between paternal adverse environmental exposure and increased disease susceptibility in offspring. However, the impact of paternal pre-pregnant caffeine exposure (PPCE) on offspring health remains unexplored. This study elucidates the sperm reprogramming mechanism and potential intervention targets for PPCE-induced non-alcoholic steatohepatitis (NASH) in offspring. Here, male rats are administrated caffeine (15-60 mg kg-1/d) by gavage for 8 weeks and then mated with females to produce offspring. This study finds that NASH with transgenerational inheritance occurred in PPCE adult offspring. Mechanistically, a reduction of miR-142-3p is implicated in the occurrence of NASH, characterized by hepatic lipid metabolism dysfunction and chronic inflammation through an increase in ACSL4. Conversely, overexpression of miR-142-3p mitigated these manifestations. The origin of reduced miR-142-3p levels is traced to hypermethylation in the miR-142-3p promoter region of parental sperm, induced by elevated corticosterone levels rather than by caffeine per se. Similar outcomes are confirmed in offspring conceived via in vitro fertilization using miR-142-3pKO sperm. Overall, this study provides the first evidence of transgenerational inheritance of NASH in PPCE offspring and identifies miR-142-3p as a potential therapeutic target for NASH induced by paternal environmental adversities.
Collapse
Affiliation(s)
- Cong Zhang
- Department of PharmacologySchool of Basic Medical SciencesWuhan UniversityWuhan430071China
| | - Yu Guo
- Department of PharmacologySchool of Basic Medical SciencesWuhan UniversityWuhan430071China
- Hubei Provincial Key Laboratory of Developmentally Originated DiseaseWuhan430071China
| | - Yi Liu
- Department of PharmacologySchool of Basic Medical SciencesWuhan UniversityWuhan430071China
| | - Kexin Liu
- Department of PharmacologySchool of Basic Medical SciencesWuhan UniversityWuhan430071China
- Hubei Provincial Key Laboratory of Developmentally Originated DiseaseWuhan430071China
| | - Wen Hu
- Hubei Provincial Key Laboratory of Developmentally Originated DiseaseWuhan430071China
- Department of PharmacyZhongnan Hospital of Wuhan UniversityWuhan430072China
| | - Hui Wang
- Department of PharmacologySchool of Basic Medical SciencesWuhan UniversityWuhan430071China
- Hubei Provincial Key Laboratory of Developmentally Originated DiseaseWuhan430071China
| |
Collapse
|
2
|
Ban M, Feng W, Hou M, Zhang Z, Cui L. IVF exposure induced intergenerational effects on metabolic phenotype in mice. Reprod Biomed Online 2024; 49:103992. [PMID: 38889592 DOI: 10.1016/j.rbmo.2024.103992] [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: 03/20/2024] [Accepted: 04/09/2024] [Indexed: 06/20/2024]
Abstract
RESEARCH QUESTION What is the potential transmission of metabolic phenotype from IVF offspring to the subsequent generation? DESIGN An IVF mouse model was established. The F1 generation mice were produced though IVF or natural mating and the F2 generation was obtained through the mating of F1 generation males with normal females. Their metabolic phenotype, including systemic and hepatic glucolipid metabolism, was examined. RESULTS It was found that IVF F1 males exhibited metabolic changes. Compared with the control group, the IVF F1 generation showed increased body weight, elevated fasting glucose and insulin, and increased serum triglyceride concentrations. IVF F1 mice also showed an increased expression of hepatic lipogenesis and autophagy genes. Moreover, IVF F1 males transmitted some metabolic changes to their own male progeny (IVF F2) in the absence of a dietary challenge. IVF F2 mice had increased peri-epididymal and subcutaneous fat and decreased insulin sensitivity. Under the 'second hit' of a high-fat diet, IVF F2 mice further showed increased hepatic lipid deposition with unaltered autophagy levels. CONCLUSION This research demonstrates the impact of IVF on hepatic glucose-lipid metabolism in two successive generations of offspring, highlighting the need for additional investigation. Enhanced understanding of the mechanisms underlying the transmission of multigenerational effects induced by IVF could potentially lead to the advancement of therapeutic interventions for individuals experiencing infertility.
Collapse
Affiliation(s)
- Miaomiao Ban
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Shandong, China.; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Shandong, China.; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Shandong, China.; Shandong Technology Innovation Center for Reproductive Health, Shandong, China.; Shandong Provincial Clinical Research Center for Reproductive Health, Shandong, China.; Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.; Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Shandong, China
| | - Wanbing Feng
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Shandong, China.; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Shandong, China.; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Shandong, China.; Shandong Technology Innovation Center for Reproductive Health, Shandong, China.; Shandong Provincial Clinical Research Center for Reproductive Health, Shandong, China.; Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.; Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Shandong, China
| | - Min Hou
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Shandong, China.; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Shandong, China.; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Shandong, China.; Shandong Technology Innovation Center for Reproductive Health, Shandong, China.; Shandong Provincial Clinical Research Center for Reproductive Health, Shandong, China.; Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.; Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Shandong, China
| | - Zhirong Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Shandong, China.; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Shandong, China.; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Shandong, China.; Shandong Technology Innovation Center for Reproductive Health, Shandong, China.; Shandong Provincial Clinical Research Center for Reproductive Health, Shandong, China.; Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.; Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Shandong, China
| | - Linlin Cui
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Shandong, China.; Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Shandong, China.; Shandong Technology Innovation Center for Reproductive Health, Shandong, China.; Shandong Provincial Clinical Research Center for Reproductive Health, Shandong, China.; Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.; Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Shandong, China.; State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, The Second Hospital, Shandong University, Shandong, China..
| |
Collapse
|
3
|
Cao Y, Tian GG, Hong X, Lu Q, Wei T, Chen HF, Wu J. Reproductive chemical database: a curated database of chemicals that modulate protein targets regulating important reproductive biological processes. Cell Biosci 2024; 14:73. [PMID: 38845051 PMCID: PMC11157792 DOI: 10.1186/s13578-024-01261-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 05/31/2024] [Indexed: 06/09/2024] Open
Abstract
Recent studies have shifted the spotlight from adult disease to gametogenesis and embryo developmental events, and these are greatly affected by various environmental chemicals, such as drugs, metabolites, pollutants, and others. Growing research has highlighted the critical importance of identifying and understanding the roles of chemicals in reproductive biology. However, the functions and mechanisms of chemicals in reproductive processes remain incomplete. We developed a comprehensive database called the Reproductive Chemical Database (RCDB) ( https://yu.life.sjtu.edu.cn/ChenLab/RCDB ) to facilitate research on chemicals in reproductive biology. This resource is founded on rigorous manual literature extraction and precise protein target prediction methodologies. This database focuses on the delineation of chemicals associated with phenotypes, diseases, or endpoints intricately associated with four important reproductive processes: female and male gamete generation, fertilization, and embryo development in human and mouse. The RCDB encompasses 93 sub-GO processes, and it revealed 1447 intricate chemical-biological process interactions. To date, the RCDB has meticulously cataloged and annotated 830 distinct chemicals, while also predicting 614 target proteins from a selection of 3800 potential candidates. Additionally, the RCDB offers an online predictive tool that empowers researchers to ascertain whether specific chemicals play discernible functional roles in these reproductive processes. The RCDB is an exhaustive, cross-platform, manually curated database, which provides a user-friendly interface to search, browse, and use reproductive processes modulators and their comprehensive related information. The RCDB will help researchers to understand the whole reproductive process and related diseases and it has the potential to promote reproduction research in the pharmacological and pathophysiological areas.
Collapse
Affiliation(s)
- Yuedi Cao
- Key Laboratory for the Genetics of Development & Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Geng G Tian
- Key Laboratory for the Genetics of Development & Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200240, China
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiaokun Hong
- State Key Laboratory of Microbial Metabolism and Joint International Research Laboratory of Metabolic & Developmental Sciences, National Experimental Teaching Center for Life Sciences and Biotechnology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Qing Lu
- Key Laboratory for the Genetics of Development & Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Ting Wei
- State Key Laboratory of Microbial Metabolism and Joint International Research Laboratory of Metabolic & Developmental Sciences, National Experimental Teaching Center for Life Sciences and Biotechnology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Hai-Feng Chen
- State Key Laboratory of Microbial Metabolism and Joint International Research Laboratory of Metabolic & Developmental Sciences, National Experimental Teaching Center for Life Sciences and Biotechnology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Ji Wu
- Key Laboratory for the Genetics of Development & Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200240, China.
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, 750004, China.
| |
Collapse
|
4
|
Chen H, Luo S, Deng X, Li S, Mao Y, Yan J, Cheng Y, Liu X, Pan J, Huang H. Pre-eclamptic foetal programming predisposes offspring to hepatic steatosis via DNA methylation. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167189. [PMID: 38648899 DOI: 10.1016/j.bbadis.2024.167189] [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: 11/23/2023] [Revised: 04/01/2024] [Accepted: 04/16/2024] [Indexed: 04/25/2024]
Abstract
OBJECTIVES Gamete and embryo-foetal origins of adult diseases hypothesis proposes that adulthood chronic disorders are associated with adverse foetal and early life traits. Our study aimed to characterise developmental changes and underlying mechanisms of metabolic disorders in offspring of pre-eclampsia (PE) programmed pregnancy. METHODS Nω-Nitro-l-arginine methyl ester hydrochloride (L-NAME) induced pre-eclampsia-like C57BL/6J mouse model was used. Lipid profiling, histological morphology, indirect calorimetry, mRNA sequencing, and pyrosequencing were performed on PE offspring of both young and elderly ages. RESULTS PE offspring exhibited increased postnatal weight gain, hepatic lipid accumulation, enlarged adipocytes, and impaired energy balance that continued to adulthood. Integrated RNA sequencing of foetal and 52-week-old livers revealed that the differentially expressed genes were mainly enriched in lipid metabolism, including glycerol-3-phosphate acyl-transferase 3 (Gpat3), a key enzyme for de novo synthesis of triglycerides (TG), and carnitine palmitoyltransferase-1a (Cpt1a), a key transmembrane enzyme that mediates fatty acid degradation. Pyrosequencing of livers from PE offspring identified hypomethylated and hypermethylated regions in Gpat3 and Cpt1a promoters, which were associated with upregulated and downregulated expressions of Gpat3 and Cpt1a, respectively. These epigenetic alterations are persistent and consistent from the foetal stage to adulthood in PE offspring. CONCLUSION These findings suggest a methylation-mediated epigenetic mechanism for PE-induced intergenerational lipid accumulation, impaired energy balance and obesity in offspring, and indicate the potential benefits of early interventions in offspring exposed to maternal PE to reduce their susceptibility to metabolic disorder in their later life.
Collapse
Affiliation(s)
- Huixi Chen
- The International Peace Maternal and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200000, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai 200011, China; Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai 200030, China; Key Laboratory of Reproductive Genetics (Ministry of Education), Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China; State Key Laboratory of Cardiology, Shanghai 200000, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China
| | - Sisi Luo
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai 200011, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai 200011, China
| | - Xiuyu Deng
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200000, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai 200011, China
| | - Sisi Li
- Shanghai Key Laboratory of Reproduction and Development, Shanghai 200011, China; Reproductive Medicine Center, International Institutes of Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, China
| | - Yiting Mao
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai 200011, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai 200011, China
| | - Jing Yan
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai 200011, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai 200011, China
| | - Yi Cheng
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai 200011, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai 200011, China
| | - Xia Liu
- The International Peace Maternal and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200000, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai 200011, China
| | - Jiexue Pan
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai 200011, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai 200011, China; Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai 200030, China.
| | - Hefeng Huang
- The International Peace Maternal and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200000, China; Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai 200011, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai 200011, China; Reproductive Medicine Center, International Institutes of Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, China; Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai 200030, China; Key Laboratory of Reproductive Genetics (Ministry of Education), Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China; State Key Laboratory of Cardiology, Shanghai 200000, China.
| |
Collapse
|
5
|
胡 沛, 潘 洁, 刘 欣, 黄 荷. [Reproductive Health-Oriented Development in Assisted Reproductive Technologies]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2024; 55:501-506. [PMID: 38948299 PMCID: PMC11211779 DOI: 10.12182/20240560401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Indexed: 07/02/2024]
Abstract
Assisted reproductive technologies (ARTs) are core components of the field of reproductive medicine, encompassing multiple pivotal stages of early development from gamete maturation and fertilization to embryo development. Against the backdrop of a deteriorating trend of global decline in fertility rates, patients with infertility problems increasingly turn to ARTs to realize their dreams of parenthood. However, concomitant with this trend is a growing apprehension regarding the potential adverse effects of ARTs. Herein, we endeavor to discuss several common ARTs procedures utilized in clinical settings and the relevant cutting-edge advancements. The ARTs discussed in the article include in vitro fertilization (IVF), intracytoplasmic sperm injection (ICSI), biphasic in vitro maturation (biphasic IVM), frozen embryo transfer (FET), preimplantation genetic testing (PGT), non-invasive PGT (niPGT), etc. In addition, we reevaluated their roles within the broader context of assisted reproduction aimed at promoting reproductive health. Additionally, we will delve into the impact of ARTs on the reproductive health of the offspring. By prioritizing the reproductive well-being of both patients and their offspring, the ongoing development and improvement of ARTs to enhance their efficacy and safety will contribute significantly to the advancement of human reproductive health.
Collapse
Affiliation(s)
- 沛然 胡
- 复旦大学附属妇产科医院 生殖与发育研究院 (上海 200030)Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai 200030, China
| | - 洁雪 潘
- 复旦大学附属妇产科医院 生殖与发育研究院 (上海 200030)Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai 200030, China
| | - 欣梅 刘
- 复旦大学附属妇产科医院 生殖与发育研究院 (上海 200030)Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai 200030, China
| | - 荷凤 黄
- 复旦大学附属妇产科医院 生殖与发育研究院 (上海 200030)Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai 200030, China
- 上海市生殖与发育重点实验室 (上海 200030)Shanghai Key Laboratory of Reproduction and Development, Shanghai 200030, China
- 中国医学科学院 胚胎源性疾病研究创新单元2019RU056 (上海 200030)Research Units of Diseases of Embryonic Origin (No. 2019RU056), Chinese Academy of Medical Sciences, Shanghai 200030, China
- 浙江大学医学院附属妇产科医院 生殖遗传教育部重点实验室 (杭州 310030)Key Laboratory of Reproductive Genetics of the Ministry of Education, Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310030, China
| |
Collapse
|
6
|
Tan LL, Xiong YW, Zhang J, Li DX, Huang Y, Wang H. Like father, like daughter:Paternal cadmium exposure causes hepatic glucose metabolic disorder and phospholipids accumulation in adult female offspring. CHEMOSPHERE 2023; 338:139437. [PMID: 37451636 DOI: 10.1016/j.chemosphere.2023.139437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/21/2023] [Accepted: 07/05/2023] [Indexed: 07/18/2023]
Abstract
Cadmium (Cd), is a well-known reproductive toxicant. The impacts of paternal Cd exposure on offspring glucose and lipid metabolism remain unclear, despite the abundance of adverse reports following early exposure from the mother. Here, we assessed paternally acquired metabolic derailment using a mouse model. LC-MS/MS, transcriptomics and molecular experimental techniques were subsequently applied in this study to explore the potential mechanism. We found that paternal Cd exposure caused glucose intolerance, lower insulin sensitivity and abnormal hepatic glycogen storage in adult female offspring, but not in males. LC-MS/MS data showed that hepatic phospholipids accumulation was also only observed in adult female offspring after paternal Cd exposure. Gene expression data showed that the level of insulin signaling and lipid transport-related genes was decreased in Cd-treated adult female offspring livers. Meanwhile, AHR, a transcription factor that combines with phospholipids to promote insulin resistance, was increased in Cd-treated adult female offspring livers. In addition, the escalation of the afore-mentioned lipid metabolites in the liver occurred as early as fetal stages in the female pups following paternal Cd exposure, suggesting the potential for these lipid species to be selected as early markers of disease for metabolic derailment later in life. Altogether, paternal Cd exposure causes offspring glucose metabolism disorder and phospholipids accumulation in a sex-dependent manner. This study provides a theoretical framework for future understanding of paternal-originated metabolic diseases.
Collapse
Affiliation(s)
- Lu-Lu Tan
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Yong-Wei Xiong
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Jin Zhang
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Dai-Xin Li
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Yichao Huang
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, China.
| | - Hua Wang
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, China.
| |
Collapse
|
7
|
Chen P, Gu M, Wan S, Jiang X, Zhang F, Li Y, Zhou Q, Lu Y, Li L, Wang X. Gestational Diabetes Mellitus Impedes Fetal Lung Development Through Exosome-Dependent Crosstalk Between Trophoblasts and Lung Epithelial Cells. Int J Nanomedicine 2023; 18:641-657. [PMID: 36789391 PMCID: PMC9922507 DOI: 10.2147/ijn.s396194] [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: 11/12/2022] [Accepted: 02/02/2023] [Indexed: 02/10/2023] Open
Abstract
Background Fetal lung underdevelopment (FLUD) is associated with neonatal and childhood severe respiratory diseases, among which gestational diabetes mellitus (GDM) play crucial roles as revealed by recent prevalence studies, yet mechanism underlying GDM-induced FLUD, especially the role of trophoblasts, is not all known. Methods From the perspective of trophoblast-derived exosomes, we established in vitro, ex vivo, in vivo and GDM trophoblast models. Utilizing placenta-derived exosomes (NUB-exos and GDMUB-exos) isolated from normal and GDM umbilical cord blood plasma and trophoblast-derived exosomes (NC-exos and HG-exos) isolated from HTR8/SVneo trophoblasts medium with/without high glucose treatment, we examined their effects on fetal lung development and biological functions. Results We found that, compared with the NUB-exos group, the exosome concentration increased in GDMUB-exos group, and the content of exosomes also changed evidenced by 61 dysregulated miRNAs. After applying these exosomes to A549 alveolar type II epithelial cells, the proliferation and biological functions were suppressed while the proportion of apoptotic cells was increased as compared to the control. In ex vivo studies, we found that GDMUB-exos showed significant suppression on the growth of the fetal lung explants, where the number of terminal buds and the area of explant surface decreased and shrank. Besides, the expression of Fgf10, Vegfa, Flt-1, Kdr and surfactant proteins A, B, C, and D was downregulated in GDMUB-exos group, whilst Sox9 was upregulated. For in vivo studies, we found significant suppression of fetal lung development in GDMUB-exos group. Importantly, we found consistent alterations when we used NC-exos and HG-exos, suggesting a dominant role of trophoblasts in placenta-derived exosome-induced FLUD. Conclusion In conclusion, GDM can adversely affect trophoblasts and alter exosome contents, causing crosstalk disorder between trophoblasts and fetal lung epithelial cells and finally leading to FLUD. Findings of this study will shine insight into the theoretical explanation for the pathogenesis of FLUD.
Collapse
Affiliation(s)
- Pengzheng Chen
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital, Shandong University, Jinan, People’s Republic of China
| | - Mengqi Gu
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital, Shandong University, Jinan, People’s Republic of China
| | - Shuting Wan
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital, Shandong University, Jinan, People’s Republic of China
| | - Xiaotong Jiang
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital, Shandong University, Jinan, People’s Republic of China
| | - Fengyuan Zhang
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People’s Republic of China
| | - Yuchen Li
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People’s Republic of China
| | - Qian Zhou
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People’s Republic of China
| | - Yuan Lu
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People’s Republic of China
| | - Lei Li
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital, Shandong University, Jinan, People’s Republic of China,Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People’s Republic of China,Laboratory of Medical Science and Technology Innovation Center (Institute of Translational Medicine), Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, People’s Republic of China,Correspondence: Lei Li; Xietong Wang, Department of Obstetrics and Gynaecology, Shandong Provincial Hospital, Shandong University, Jinan, People’s Republic of China, Tel +8615168889200; +8615168888928, Email ;
| | - Xietong Wang
- Department of Obstetrics and Gynaecology, Shandong Provincial Hospital, Shandong University, Jinan, People’s Republic of China,Department of Obstetrics and Gynaecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People’s Republic of China,Laboratory of Medical Science and Technology Innovation Center (Institute of Translational Medicine), Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, People’s Republic of China,Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital, Jinan, People’s Republic of China
| |
Collapse
|
8
|
Hua L, Ju L, Xu H, Li C, Sun S, Zhang Q, Cao J, Ding R. Outdoor air pollution exposure and the risk of asthma and wheezing in the offspring. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:14165-14189. [PMID: 36149565 DOI: 10.1007/s11356-022-23094-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 09/14/2022] [Indexed: 06/16/2023]
Abstract
According to the "fetal origin of disease" hypothesis, air pollution exposure in pregnancy may play an important role in stimulating the early programming of asthma and allergies. However, previous studies reported inconsistent findings. The aim of this meta-analysis was to provide higher grade evidence and quantitatively analyze the link between prenatal exposure to outdoor air pollutants and childhood asthma and wheezing. Databases (Web of Science and PubMed) were extensively searched for articles published from the start of the database to September 15, 2021. Either random-effect model or fixed-effect model was used to estimate the disease-specific relative risks (RR) with the corresponding 95% confidence intervals (CIs) to estimate the association. Newcastle-Ottawa Quality Score (NOS) was used to assess the quality of studies. This study finally included 13 cohort studies, and the findings showed that NO2 and SO2 exposure during entire pregnancy was significantly associated with wheezing (RR = 1.032, 95% CI: 1.000, 1.066) and asthma (RR = 1.114, 95% CI: 1.066, 1.164), respectively. Further analyses showed that PM2.5 were positively associated with asthma in the second (RR = 1.194, 95% CI: 1.143, 1.247) and third trimester (RR = 1.050, 95% CI: 1.007, 1.094), while NO2 (RR = 1.060, 95% CI: 1.021, 1.101) and SO2 (RR = 1.067, 95% CI: 1.013, 1.123) were shown positively associated with asthma only in the second trimester. The relationship between wheezing and outdoor air pollutants was not significant in any of the pregnancy subgroups. This study suggests that prenatal exposure of outdoor air pollution may increase the asthma and wheezing risk in the offspring and that the second trimester may be a sensitive period for air pollution exposure. But the interpretation of the causal association is hampered by limited number of studies on dose response.
Collapse
Affiliation(s)
- Lei Hua
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Liangliang Ju
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Hanbing Xu
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Changlian Li
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Shu Sun
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Qi Zhang
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Jiyu Cao
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China
- Department of Teaching Center for Preventive Medicine, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Rui Ding
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China.
| |
Collapse
|
9
|
Lu YJ, Li Q, Chen LX, Tian T, Kang J, Hao YX, Zhou JS, Wang YY, Yan LY, Li R, Chang L, Qiao J. Association between maternal MTHFR C677T/A1298C combination polymorphisms and IVF/ICSI outcomes: a retrospective cohort study. Hum Reprod Open 2022; 2023:hoac055. [PMID: 36531663 PMCID: PMC9749479 DOI: 10.1093/hropen/hoac055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 10/10/2022] [Indexed: 10/11/2023] Open
Abstract
STUDY QUESTION What are the roles of maternal 5,10-methylenetetrahydrofolate reductase (MTHFR) C677T/A1298C combination polymorphisms on the embryological and clinical outcomes of IVF/ICSI? SUMMARY ANSWER Our study reveals for the first time that the oocyte maturation potential gradually decreases with a reduction of maternal MTHFR activity determined by combined C677T/A1298C polymorphisms, while embryo quality was worse in women with intermediate MTHFR activity. WHAT IS KNOWN ALREADY Although many previous studies have explored the association between MTHFR polymorphisms and IVF/ICSI outcomes, the results remain contradictory due to inadequate samples, no adjustment for potential confounders and/or the study of C677T and A1298C separately. Few studies have systematically investigated the exact role of MTHFR activity determined by combined C677T/A1298C polymorphisms on the embryological and clinical outcomes of IVF/ICSI. STUDY DESIGN SIZE DURATION This is a retrospective cohort study investigating 1160 women who were referred for MTHFR genotyping and IVF/ICSI treatment at Peking University Third Hospital from May 2017 to May 2020. PARTICIPANTS/MATERIALS SETTING METHODS Women who were referred for MTHFR genotyping and their first IVF/ICSI treatment at our hospital were included and those undergoing preimplantation genetic testing cycles were excluded. The included women were divided into different cohorts according to their C677T, A1298C and combined C677T/A1298C genotypes. The embryological outcomes, including oocytes retrieved, metaphase II oocytes, oocyte maturation rate, normal fertilization rate and transplantable embryo rate, were evaluated by generalized linear regression models. The clinical outcomes, including biochemical pregnancy rate, clinical pregnancy rate and live birth rate, were evaluated by log-binomial regression models. All outcomes were adjusted for potential confounders. MAIN RESULTS AND THE ROLE OF CHANCE Women with the combined 677TT/1298AA genotype (hereafter abbreviated as TT/AA, as with other combined genotypes), whose enzyme activity was the lowest, had a lower oocyte maturation rate compared with those with the wild-type genotype (P = 0.007). Moreover, the oocyte maturation rate decreased linearly with the decline in MTHFR enzyme activity determined by combined C677T/A1298C genotypes (P-trend = 0.001). The combined CC/AC, CC/CC&CT/AA and CT/AC genotypes with intermediate enzyme activity were associated with a lower transplantable embryo rate (P = 0.013, 0.030 and 0.039, respectively). The differences in clinical outcomes between women with wild-type genotype and combined C677T/A1298C variant genotypes were not significant. LIMITATIONS REASONS FOR CAUTION Our study population had comparable embryological outcomes but worse clinical outcomes than other women undergoing IVF/ICSI treatment at our hospital. Therefore, the results related to the clinical outcomes should be generalized with caution. In addition, we did not detect the folate concentration of each patient during pregnancy. However, this might not have much influence on our results because almost all of our study participants took sufficient folic acid around pregnancy. WIDER IMPLICATIONS OF THE FINDINGS We provide a holistic view of the effect of MTHFR C677T and A1298C polymorphisms on the IVF/ICSI outcomes, which can contribute to providing reasonable folic acid supplementation suggestions for women with different MTHFR genotypes, especially for those with a low oocyte maturation rate and/or low embryo quality. STUDY FUNDING/COMPETING INTERESTS This work was funded by the National Natural Science Foundation of China (31871447, and 82101677), the National Key Research and Development Program (2019YFA0801400) and the Natural Science Foundation of Beijing Municipality (7202226). The authors declare that they have no competing interests. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Yong-Jie Lu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Qin Li
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Li-Xue Chen
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Tian Tian
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Jia Kang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Yong-Xiu Hao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Jian-Suo Zhou
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| | - Yuan-Yuan Wang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Li-Ying Yan
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Rong Li
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Liang Chang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Jie Qiao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| |
Collapse
|
10
|
Borišek J, Aupič J, Magistrato A. Establishing the catalytic and regulatory mechanism of
RNA
‐based machineries. WIRES COMPUTATIONAL MOLECULAR SCIENCE 2022. [DOI: 10.1002/wcms.1643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Jure Borišek
- Theory Department National Institute of Chemistry Ljubljana Slovenia
| | | | | |
Collapse
|
11
|
Li XY, Pan JX, Zhu H, Ding GL, Huang HF. Environmental epigenetic interaction of gametes and early embryos. Biol Reprod 2022; 107:196-204. [PMID: 35323884 DOI: 10.1093/biolre/ioac051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/18/2022] [Accepted: 02/24/2022] [Indexed: 11/14/2022] Open
Abstract
In recent years, the developmental origins of diseases have been increasingly recognized and accepted. As such, it has been suggested that most adulthood chronic diseases such as diabetes, obesity, cardiovascular disease, and even tumors may develop at a very early stage. In addition to intrauterine environmental exposure, germ cells carry an important inheritance role as the primary link between the two generations. Adverse external influences during differentiation and development can cause damage to germ cells, which may then increase the risk of chronic disease development later in life. Here, we further elucidate and clarify the concept of gamete and embryo origins of adult diseases by focusing on the environmental insults on germ cells, from differentiation to maturation and fertilization.
Collapse
Affiliation(s)
- Xin-Yuan Li
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China.,Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences
| | - Jie-Xue Pan
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China.,Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences
| | - Hong Zhu
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China.,Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences
| | - Guo-Lian Ding
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China.,Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - He-Feng Huang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China.,Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China.,The Key Laboratory of Reproductive Genetics (Zhejiang University), Ministry of Education, Hangzhou, China
| |
Collapse
|
12
|
D’Esposito RJ, Myers CA, Chen AA, Vangaveti S. Challenges with Simulating Modified RNA: Insights into Role and Reciprocity of Experimental and Computational Approaches. Genes (Basel) 2022; 13:540. [PMID: 35328093 PMCID: PMC8949676 DOI: 10.3390/genes13030540] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/15/2022] [Accepted: 03/16/2022] [Indexed: 01/12/2023] Open
Abstract
RNA is critical to a broad spectrum of biological and viral processes. This functional diversity is a result of their dynamic nature; the variety of three-dimensional structures that they can fold into; and a host of post-transcriptional chemical modifications. While there are many experimental techniques to study the structural dynamics of biomolecules, molecular dynamics simulations (MDS) play a significant role in complementing experimental data and providing mechanistic insights. The accuracy of the results obtained from MDS is determined by the underlying physical models i.e., the force-fields, that steer the simulations. Though RNA force-fields have received a lot of attention in the last decade, they still lag compared to their protein counterparts. The chemical diversity imparted by the RNA modifications adds another layer of complexity to an already challenging problem. Insight into the effect of RNA modifications upon RNA folding and dynamics is lacking due to the insufficiency or absence of relevant experimental data. This review provides an overview of the state of MDS of modified RNA, focusing on the challenges in parameterization of RNA modifications as well as insights into relevant reference experiments necessary for their calibration.
Collapse
Affiliation(s)
- Rebecca J. D’Esposito
- Department of Chemistry, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, USA; (R.J.D.); (A.A.C.)
| | - Christopher A. Myers
- Department of Physics, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, USA;
| | - Alan A. Chen
- Department of Chemistry, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, USA; (R.J.D.); (A.A.C.)
- The RNA Institute, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, USA
| | - Sweta Vangaveti
- The RNA Institute, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, USA
| |
Collapse
|
13
|
Pan HT, Shi XL, Fang M, Sun XM, Chen PP, Ding JL, Xia GY, Yu B, Zhang T, Zhu HD. Profiling of exosomal microRNAs expression in umbilical cord blood from normal and preeclampsia patients. BMC Pregnancy Childbirth 2022; 22:124. [PMID: 35152894 PMCID: PMC8842963 DOI: 10.1186/s12884-022-04449-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 02/03/2022] [Indexed: 02/08/2023] Open
Abstract
Background Epidemiological and experimental studies suggest that preeclampsia has a negative impact on maternity and offspring health. Previous studies report that dysregulation in utero-environment increases risk for elderly disease such as cardiovascular disease. However, the underlying mechanisms remain elusive. Specific microRNAs (miRNAs) are packaged in exosomes may regulate microvascular dysfunction in offspring of mothers with preeclampsia. The present study aimed to identify the differential expression profiles of microRNAs in the serum exosomes between patients with preeclampsia and normal pregnancies. Methods A comprehensive miRNA sequence-based approach was performed to compare exosomes carry miRNAs (Exo-miRNAs) expression levels in umbilical serum between normal and preeclampsia patients. Exosomes were isolated using the ExoQuick precipitation kit. Serum exosomes were then viewed under electron microscopy, and their characteristics determined by western blotting and nanoparticle-tracking analysis. Illumina platform was used to perform sequencing. Bioinformatics analysis was used to explore differentially expressed Exo-miRNAs in umbilical serum. Results Based on sequence similarity, 1733 known miRNAs were retrieved. Furthermore, 157 mature miRNAs in serum exosomes were significantly differential expressed between PE and those control groups (P<0.05, log2|FC| > 1). Out, of the 157 miRNAs, 96 were upregulated miRNAs whereas 61 miRNAs were downregulated. The 157 differentially expressed miRNAs targeted 51,424 differentially expressed genes. Functional analysis through KEGG pathway and Gene Ontology results uncovered that target genes of miRNAs with differential expression were significantly linked to several pathways and biological processes. Conclusion The findings of this study showed differential expression of umbilical serum Exo-miRNAs in normal compared with PE patients, implying that these Exo-miRNAs may associate with microvascular dysfunction in offspring of mothers with preeclampsia. Supplementary Information The online version contains supplementary material available at 10.1186/s12884-022-04449-w.
Collapse
Affiliation(s)
- Hai-Tao Pan
- Shaoxing Maternity and Child Health Care Hospital, Shaoxing, 312000, China.,Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, China.,The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Xiao-Liang Shi
- Shaoxing Maternity and Child Health Care Hospital, Shaoxing, 312000, China.,Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, China
| | - Min Fang
- Shaoxing Maternity and Child Health Care Hospital, Shaoxing, 312000, China.,Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, China
| | - Xiang-Mei Sun
- Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, China
| | - Pan-Pan Chen
- Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, China
| | - Jin-Long Ding
- Shaoxing Maternity and Child Health Care Hospital, Shaoxing, 312000, China.,Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, China
| | - Gui-Yu Xia
- Shaoxing Maternity and Child Health Care Hospital, Shaoxing, 312000, China.,Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, China
| | - Bin Yu
- Shaoxing Maternity and Child Health Care Hospital, Shaoxing, 312000, China.,Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, China
| | - Tao Zhang
- Shaoxing Maternity and Child Health Care Hospital, Shaoxing, 312000, China. .,Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, China.
| | - Hong-Dan Zhu
- Shaoxing Maternity and Child Health Care Hospital, Shaoxing, 312000, China. .,Obstetrics and Gynecology Hospital of Shaoxing University, Shaoxing, China.
| |
Collapse
|
14
|
Mao Y, Yiran Z, Sisi L, Huixi C, Xia L, Ting W, Guolian D, Xinmei L, Sheng J, Meng Y, Huang H. Advanced paternal age increased metabolic risks in mice offspring. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166355. [DOI: 10.1016/j.bbadis.2022.166355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 01/07/2022] [Accepted: 01/25/2022] [Indexed: 10/19/2022]
|
15
|
Merrill SM, Moore SR, Gladish N, Giesbrecht GF, Dewey D, Konwar C, MacIssac JL, Kobor MS, Letourneau NL. Paternal adverse childhood experiences: Associations with infant DNA methylation. Dev Psychobiol 2021; 63:e22174. [PMID: 34333774 DOI: 10.1002/dev.22174] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 06/17/2021] [Accepted: 06/24/2021] [Indexed: 12/14/2022]
Abstract
Adverse childhood experiences (ACEs), or cumulative childhood stress exposures, such as abuse, neglect, and household dysfunction, predict later health problems in both the exposed individuals and their offspring. One potential explanation suggests exposure to early adversity predicts epigenetic modification, especially DNA methylation (DNAm), linked to later health. Stress experienced preconception by mothers may associate with DNAm in the next generation. We hypothesized that fathers' exposure to ACEs also associates with their offspring DNAm, which, to our knowledge, has not been previously explored. An epigenome-wide association study (EWAS) of blood DNAm (n = 45) from 3-month-old infants was regressed onto fathers' retrospective ACEs at multiple Cytosine-phosphate-Guanosine (CpG) sites to discover associations. This accounted for infants' sex, age, ethnicity, cell type proportion, and genetic variability. Higher ACE scores associated with methylation values at eight CpGs. Post-hoc analysis found no contribution of paternal education, income, marital status, and parental postpartum depression, but did with paternal smoking and BMI along with infant sleep latency. These same CpGs also contributed to the association between paternal ACEs and offspring attention problems at 3 years. Collectively, these findings suggested there were biological associations with paternal early life adversity and offspring DNAm in infancy, potentially affecting offspring later childhood outcomes.
Collapse
Affiliation(s)
- Sarah M Merrill
- BC Children's Hospital Research Institute Vancouver, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Molecular Medicine and Therapeutics, Vancouver, British Columbia, Canada
| | - Sarah R Moore
- BC Children's Hospital Research Institute Vancouver, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Molecular Medicine and Therapeutics, Vancouver, British Columbia, Canada
| | - Nicole Gladish
- BC Children's Hospital Research Institute Vancouver, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Molecular Medicine and Therapeutics, Vancouver, British Columbia, Canada
| | - Gerald F Giesbrecht
- Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada.,Owerko Centre at the Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Deborah Dewey
- Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada.,Owerko Centre at the Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada.,Department of Community Health Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Chaini Konwar
- BC Children's Hospital Research Institute Vancouver, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Molecular Medicine and Therapeutics, Vancouver, British Columbia, Canada
| | - Julia L MacIssac
- BC Children's Hospital Research Institute Vancouver, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Molecular Medicine and Therapeutics, Vancouver, British Columbia, Canada
| | - Michael S Kobor
- BC Children's Hospital Research Institute Vancouver, Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Molecular Medicine and Therapeutics, Vancouver, British Columbia, Canada.,Program in Child and Brain Development, CIFAR, Toronto, Ontario, Canada
| | - Nicole L Letourneau
- Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada.,Owerko Centre at the Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada.,Department of Psychiatry, University of Calgary, Calgary, Alberta, Canada.,Faculty of Nursing, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
16
|
Qin N, Zhou Z, Zhao W, Zou K, Shi W, Yu C, Liu X, Dong Z, Mao Y, Liu X, Sheng J, Ding G, Wu Y, Huang H. Abnormal Glucose Metabolism in Male Mice Offspring Conceived by in vitro Fertilization and Frozen-Thawed Embryo Transfer. Front Cell Dev Biol 2021; 9:637781. [PMID: 33634140 PMCID: PMC7900417 DOI: 10.3389/fcell.2021.637781] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 01/20/2021] [Indexed: 12/16/2022] Open
Abstract
Frozen and thawed embryo transfer (FET) is currently widely applied in routine assisted reproductive technology (ART) procedure. It is of great necessity to assess the safety of FET and investigate the long-term effect including glucose metabolism on FET-conceived offspring. The mouse model is a highly efficient method to figure out the relationship between the process of FET and offspring health. In this study, we obtained mouse offspring of natural conception (NC), in vitro fertilization (IVF), and FET. Glucose and insulin tolerance test (GTT/ITT) were performed on both chow fed or high fat diet (HFD) fed offspring to examine the glucose metabolism status. We detected hepatic PI3K/AKT pathway by western blotting and transcriptome status by RNA-sequencing. Impaired glucose tolerance (IGT) and decreased insulin tolerance were occurred in FET conceived male offspring. After challenged with the HFD-fed, male offspring in FET group performed earlier and severer IGT than IVF group. Furthermore, higher HOMA-IR index and higher serum insulin level post glucose injected in FET-chow group suggested the insulin resistance status. The PI3K/AKT signaling pathway, the major pathway of insulin in the liver, were also disrupted in FET group. Transcriptomics of the liver reveals significantly downregulated in glucose metabolic process and insulin resistance in the FET-chow group. In our study, FET-conceived male mouse offspring presented glucose metabolism dysfunction mainly manifesting insulin resistance. The hepatic insulin signaling pathway were in concordance with reduced glycogen synthesis, increased glycolysis and enhanced gluconeogenesis status in FET-conceived male offspring.
Collapse
Affiliation(s)
- Ningxin Qin
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai Jiao Tong University, Shanghai, China
| | - Zhiyang Zhou
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai Jiao Tong University, Shanghai, China
| | - Wenlong Zhao
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai Jiao Tong University, Shanghai, China
| | - Kexin Zou
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai Jiao Tong University, Shanghai, China
| | - Weihui Shi
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai Jiao Tong University, Shanghai, China
| | - Chuanjin Yu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai Jiao Tong University, Shanghai, China
| | - Xia Liu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai Jiao Tong University, Shanghai, China
| | - Zehan Dong
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai Jiao Tong University, Shanghai, China
| | - Yiting Mao
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai Jiao Tong University, Shanghai, China
| | - Xinmei Liu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai Jiao Tong University, Shanghai, China.,Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Jianzhong Sheng
- Department of Pathology and Pathophysiology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Guolian Ding
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai Jiao Tong University, Shanghai, China.,Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Yanting Wu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai Jiao Tong University, Shanghai, China.,Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Hefeng Huang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai Jiao Tong University, Shanghai, China.,Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China.,The Key Laboratory of Reproductive Genetics, Ministry of Education, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
17
|
Epigenetic Mechanisms of Paternal Stress in Offspring Development and Diseases. Int J Genomics 2021; 2021:6632719. [PMID: 33532485 PMCID: PMC7837765 DOI: 10.1155/2021/6632719] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/28/2020] [Accepted: 01/08/2021] [Indexed: 01/10/2023] Open
Abstract
The major biological function of the sperm cell is to transmit the paternal genetic and epigenetic information to the embryo as well as the following offspring. Sperm has a unique epigenome. An increasing body of epidemiological study supports that paternal stress induced by environmental exposures and lifestyle can modulate the sperm epigenome (including histone modification, DNA methylation, and noncoding RNA expression), sperm-egg fusion, embryo development, and offspring health. Based on the existing literature, we have summarized the paternal exposure on sperm epigenome along with the representative phenotypes of offspring and the possible mechanism involved.
Collapse
|
18
|
Hu M, Lou Y, Liu S, Mao Y, Le F, Wang L, Li L, Wang Q, Li H, Lou H, Wang N, Jin F. Altered expression of DNA damage repair genes in the brain tissue of mice conceived by in vitro fertilization. Mol Hum Reprod 2020; 26:141-153. [PMID: 32003796 DOI: 10.1093/molehr/gaaa010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 12/16/2019] [Accepted: 01/08/2020] [Indexed: 12/22/2022] Open
Abstract
Our previous study revealed a higher incidence of gene dynamic mutation in newborns conceived by IVF, highlighting that IVF may be disruptive to the DNA stability of IVF offspring. However, the underlying mechanisms remain unclear. The DNA damage repair system plays an essential role in gene dynamic mutation and neurodegenerative disease. To evaluate the long-term impact of IVF on DNA damage repair genes, we established an IVF mouse model and analyzed gene and protein expression levels of MSH2, MSH3, MSH6, MLH1, PMS2, OGG1, APEX1, XPA and RPA1 and also the amount of H2AX phosphorylation of serine 139 which is highly suggestive of DNA double-strand break (γH2AX expression level) in the brain tissue of IVF conceived mice and their DNA methylation status using quantitative real-time PCR, western blotting and pyrosequencing. Furthermore, we assessed the capacity of two specific non-physiological factors in IVF procedures during preimplantation development. The results demonstrated that the expression and methylation levels of some DNA damage repair genes in the brain tissue of IVF mice were significantly changed at 3 weeks, 10 weeks and 1.5 years of age, when compared with the in vivo control group. In support of mouse model findings, oxygen concentration of in vitro culture environment was shown to have the capacity to modulate gene expression and DNA methylation levels of some DNA damage repair genes. In summary, our study indicated that IVF could bring about long-term alterations of gene and protein expression and DNA methylation levels of some DNA damage repair genes in the brain tissue and these alterations might be resulted from the different oxygen concentration of culture environment, providing valuable perspectives to improve the safety and efficiency of IVF at early embryonic stage and also throughout different life stages.
Collapse
Affiliation(s)
- Minhao Hu
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, 1 Xueshi Road, Hangzhou 310006, China
| | - Yiyun Lou
- Department of Gynaecology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou 310007, China
| | - Shuyuan Liu
- Department of Gynaecology, Weifang Maternal and Child Health Hospital, Weifang 261000, China
| | - Yuchan Mao
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, 1 Xueshi Road, Hangzhou 310006, China
| | - Fang Le
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, 1 Xueshi Road, Hangzhou 310006, China
| | - Liya Wang
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, 1 Xueshi Road, Hangzhou 310006, China
| | - Lejun Li
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, 1 Xueshi Road, Hangzhou 310006, China
| | - Qijing Wang
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, 1 Xueshi Road, Hangzhou 310006, China
| | - Hongping Li
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, 1 Xueshi Road, Hangzhou 310006, China
| | - Hangying Lou
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, 1 Xueshi Road, Hangzhou 310006, China
| | - Ning Wang
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, 1 Xueshi Road, Hangzhou 310006, China
| | - Fan Jin
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, 1 Xueshi Road, Hangzhou 310006, China.,Women's Reproductive Health Laboratory of Zhejiang Province, Key Laboratory of Reproductive Genetics, Ministry of Education, Hangzhou 310006, China
| |
Collapse
|
19
|
Health status and nutritional development of adopted Ethiopian children living in southern Spain: A prospective cohort study. Nutrition 2019; 71:110611. [PMID: 31785516 DOI: 10.1016/j.nut.2019.110611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 08/12/2019] [Accepted: 09/01/2019] [Indexed: 11/23/2022]
Abstract
OBJECTIVE The first aim of this study was to evaluate the health status and anthropometrical development of adopted children from Ethiopia living in southern Spain. A second aim was to evaluate the association between these parameters and adherence to the Mediterranean dietary pattern. METHODS The study sample included 53 adopted children from Ethiopia and a matched sample of 54 native-born children. A physical examination of the children, including height and weight, was conducted in Ethiopia at the time of entry into the adoption process. Height and weight were re-measured at the first day of adoption and 6, 12, and 24 mo after adoption. After 2 y of follow-up, another physical examination was performed, including the KIDMED test, to measure adherence to the Mediterranean diet. RESULTS Skin and digestive conditions were the most prevalent disorders in Ethiopian children before adoption and at the end of follow-up. Baseline anthropometric characteristics indicated a low wasting prevalence (7.5%); however, stunted growth was more prevalent (35.8%). After 6 mo, the weight-for-age of Ethiopian children was restored (change from baseline P < 0.001), and not significantly different from the Spanish children at 1-y after adoption. Height-for-age also increased from baseline (P < 0.001. A higher KIDMED score was associated with increased weight-for-age (r = 0.279; P = 0.045) and height-for-age (r = 0.385; P = 0.004). CONCLUSIONS This prospective study of adopted Ethiopian children confirmed a rapid growth development that occurred from the beginning of the adoption process and continued after the 2-y of follow-up. A higher adherence to the Mediterranean diet was associated with better growth development, which reinforces the importance of a balanced and adequate diet in growing children.
Collapse
|
20
|
Zhao L, Zheng X, Liu J, Zheng R, Yang R, Wang Y, Sun L. PPAR signaling pathway in the first trimester placenta from in vitro fertilization and embryo transfer. Biomed Pharmacother 2019; 118:109251. [PMID: 31351426 DOI: 10.1016/j.biopha.2019.109251] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 07/16/2019] [Accepted: 07/18/2019] [Indexed: 01/27/2023] Open
Abstract
Placenta is a temporary critical organ related to fetal development and pregnancy outcomes. And epidemiologic data demonstrate an increased risk of placental abnormality after in vitro fertilization and embryo transfer (IVF-ET). This study aims to explore the molecular mechanism for PPAR signaling pathway in placenta subjected to IVF-ET in the first trimester. Four first trimester placenta samples from double chorionic twins to single reduction in IVF-ET only because of oviducts factors. The other four control placenta samples from double chorionic twin were derived from those unplanned spontaneously conceived pregnancy after the legal termination. Affymetrix HG-U133 Plus 2.0 Array was performed to evaluate the global gene expressions. We confirmed microarray results from 10 significant differential genes using RT-qPCR. And 10 deregulated gene products were stained in the first trimester placenta by immunohistochemistry. These differentially expressed genes in IVF-ET placentas were submitted to functional annotation of clustering tools of bioinformatics resources and gene ontology enrichment analysis. Schematic representation of placental PPAR signaling pathway was labelled by Kyoto Encyclopedia of Genes and Genomes (KEGG). Analysis results of early placental PPAR signaling pathway gene expression from 8 women demonstrated 34 genes with a significant change in expression between IVF-ET and control group, 25 up-regulated; 9 down-regulated. KEGG pathway analysis indicated that IVF-ET manipulation extensively over-activated PPAR signaling pathway. Immune tolerance, trophoblast invasion, syncytia formation, lipid and glucose metabolism, inflammatory response and other complex biological functions were disturbed. RT-qPCR results and proteins staining intensity were consisted with microarray. Placental gene expressions and functions in PPAR signaling pathway were affected by IVF-ET treatment in the first trimester, which may offer a potential mechanism for the pathogenesis of various adverse outcomes during the perinatal period.
Collapse
Affiliation(s)
- Liang Zhao
- Department of Obstetrics and Gynecology, Beijing Jishuitan Hospital, No. 31, Xinjiekou East Street, Xicheng District, Beijing, 100035, China
| | - Xiuli Zheng
- Department of Obstetrics and Gynecology, Beijing Jishuitan Hospital, No. 31, Xinjiekou East Street, Xicheng District, Beijing, 100035, China
| | - Jingfang Liu
- Department of Obstetrics and Gynecology, Beijing Jishuitan Hospital, No. 31, Xinjiekou East Street, Xicheng District, Beijing, 100035, China
| | - Rong Zheng
- Department of Obstetrics and Gynecology, Beijing Jishuitan Hospital, No. 31, Xinjiekou East Street, Xicheng District, Beijing, 100035, China
| | - Rui Yang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, No. 49, Huayuan North Road, Haidian District, Beijing, 100191, China
| | - Ying Wang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, No. 49, Huayuan North Road, Haidian District, Beijing, 100191, China
| | - Lifang Sun
- Department of Obstetrics and Gynecology, Beijing Jishuitan Hospital, No. 31, Xinjiekou East Street, Xicheng District, Beijing, 100035, China.
| |
Collapse
|
21
|
Zhao L, Zheng X, Liu J, Zheng R, Yang R, Wang Y, Sun L. The placental transcriptome of the first-trimester placenta is affected by in vitro fertilization and embryo transfer. Reprod Biol Endocrinol 2019; 17:50. [PMID: 31262321 PMCID: PMC6604150 DOI: 10.1186/s12958-019-0494-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 06/17/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The placenta is a highly specialized temporary organ that is related to fetal development and pregnancy outcomes, and epidemiological data demonstrate an increased risk of placental abnormality after in vitro fertilization and embryo transfer (IVF-ET). METHODS This study examines alterations in the transcriptome profile of first-trimester placentas from IVF-ET pregnancies and analyzes the potential mechanisms that play a role in the adverse perinatal outcomes associated with IVF-ET procedures. Four human placental villi from first-trimester samples were obtained through fetal bud aspiration from patients subjected to IVF-ET due to oviductal factors. An additional four control human placental villi were derived from a group of subjects who spontaneously conceived a twin pregnancy. We analyzed their transcriptomes by microarray. Then, RT-qPCR and immunohistochemistry were utilized to analyze several dysregulated genes to validate the microarray results. Biological functions and pathways were analyzed with bioinformatics tools. RESULTS A total of 3405 differentially regulated genes were identified as significantly dysregulated (> 2-fold change; P < 0.05) in the IVF-ET placenta in the first trimester: 1910 upregulated and 1495 downregulated genes. Functional enrichment analysis of the differentially regulated genes demonstrated that the genes were involved in more than 50 biological processes and pathways that have been shown to play important roles in the first trimester in vivo. These pathways can be clustered into coagulation cascades, immune response, transmembrane signaling, metabolism, cell cycle, stress control, invasion and vascularization. Nearly the same number of up- and downregulated genes participate in the same biological processes related to placental development and maintenance. Procedures utilized in IVF-ET altered the expression of first-trimester placental genes that are critical to these biological processes and triggered a compensatory mechanism during early implantation in vivo. CONCLUSION These data provide a potential basis for further analysis of the higher frequency of adverse perinatal outcomes following IVF-ET, with the ultimate goal of developing safer IVF-ET protocols.
Collapse
Affiliation(s)
- Liang Zhao
- Department of Obstetrics and Gynecology, Beijing Jishuitan, Hospital, No. 31, Xinjiekou East Street, Xicheng District, Beijing, 100035, People's Republic of China
| | - Xiuli Zheng
- Department of Obstetrics and Gynecology, Beijing Jishuitan, Hospital, No. 31, Xinjiekou East Street, Xicheng District, Beijing, 100035, People's Republic of China
| | - Jingfang Liu
- Department of Obstetrics and Gynecology, Beijing Jishuitan, Hospital, No. 31, Xinjiekou East Street, Xicheng District, Beijing, 100035, People's Republic of China
| | - Rong Zheng
- Department of Obstetrics and Gynecology, Beijing Jishuitan, Hospital, No. 31, Xinjiekou East Street, Xicheng District, Beijing, 100035, People's Republic of China
| | - Rui Yang
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University Third Hospital, No. 49, Huayuan North Road, Haidian District, Beijing, 100191, People's Republic of China
| | - Ying Wang
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University Third Hospital, No. 49, Huayuan North Road, Haidian District, Beijing, 100191, People's Republic of China
| | - Lifang Sun
- Department of Obstetrics and Gynecology, Beijing Jishuitan, Hospital, No. 31, Xinjiekou East Street, Xicheng District, Beijing, 100035, People's Republic of China.
| |
Collapse
|