1
|
Xie JK, Wang Q, Chen YH, Tang SB, Sun HY, Ge ZJ, Zhang CL. Effects of multisuperovulation on the transcription and genomic methylation of oocytes and offspring. Clin Epigenetics 2024; 16:135. [PMID: 39342274 PMCID: PMC11439255 DOI: 10.1186/s13148-024-01746-3] [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: 04/24/2024] [Accepted: 09/15/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Controlled ovarian stimulation is a common skill of assisted reproductive technologies (ARTs). In the clinic, some females would undergo more than one controlled ovarian stimulation cycle. However, few studies have focused on the influence of multi-superovulation on oocytes and offspring. RESULTS Here, we found that multi-superovulation disrupted the transcriptome of oocytes and that the differentially expressed genes (DEGs) were associated mainly with metabolism and fertilization. The disruption of mRNA degradation via poly (A) size and metabolism might be a reason for the reduced oocyte maturation rate induced by repeated superovulation. Multi-superovulation results in hypo-genomic methylation in oocytes. However, there was an increase in the methylation level of CGIs. The DMRs are not randomly distributed in genome elements. Genes with differentially methylated regions (DMRs) in promoters are enriched in metabolic pathways. With increasing of superovulation cycles, the glucose and insulin tolerance of offspring is also disturbed. CONCLUSIONS These results suggest that multi-superovulation has adverse effects on oocyte quality and offspring health.
Collapse
Affiliation(s)
- Juan-Ke Xie
- Reproductive Medicine Center, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Qian Wang
- Reproductive Medicine Center, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Yuan-Hui Chen
- Reproductive Medicine Center, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Shou-Bin Tang
- College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement, in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, China
| | - Hao-Yue Sun
- Reproductive Medicine Center, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Zhao-Jia Ge
- College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement, in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, China.
| | - Cui-Lian Zhang
- Reproductive Medicine Center, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, China.
| |
Collapse
|
2
|
Meulders B, Marei WFA, Xhonneux I, Loier L, Smits A, Leroy JLMR. Preconception Diet Interventions in Obese Outbred Mice and the Impact on Female Offspring Metabolic Health and Oocyte Quality. Int J Mol Sci 2024; 25:2236. [PMID: 38396912 PMCID: PMC10888670 DOI: 10.3390/ijms25042236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/07/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
Obese individuals often suffer from metabolic health disorders and reduced oocyte quality. Preconception diet interventions in obese outbred mice restore metabolic health and oocyte quality and mitochondrial ultrastructure. Also, studies in inbred mice have shown that maternal obesity induces metabolic alterations and reduces oocyte quality in offspring (F1). Until now, the effect of maternal high-fat diet on F1 metabolic health and oocyte quality and the potential beneficial effects of preconception dietary interventions have not been studied together in outbred mice. Therefore, we fed female mice a high-fat/high-sugar (HF/HS) diet for 7 weeks and switched them to a control (CONT) or caloric-restriction (CR) diet or maintained them on the HF/HS diet for 4 weeks before mating, resulting in three treatment groups: diet normalization (DN), CR, and HF/HS. In the fourth group, mice were fed CONT diet for 11 weeks (CONT). HF/HS mice were fed an HF/HS diet from conception until weaning, while all other groups were then fed a CONT diet. After weaning, offspring were kept on chow diet and sacrificed at 11 weeks. We observed significantly elevated serum insulin concentrations in female HF/HS offspring and a slightly increased percentage of mitochondrial ultrastructural abnormalities, mitochondrial size, and mitochondrial mean gray intensity in HF/HS F1 oocytes. Also, global DNA methylation was increased and cellular stress-related proteins were downregulated in HF/HS F1 oocytes. Mostly, these alterations were prevented in the DN group, while, in CR, this was only the case for a few parameters. In conclusion, this research has demonstrated for the first time that a maternal high-fat diet in outbred mice has a moderate impact on female F1 metabolic health and oocyte quality and that preconception DN is a better strategy to alleviate this compared to CR.
Collapse
Affiliation(s)
- Ben Meulders
- Gamete Research Centre, Laboratory of Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, 2610 Antwerp, Belgium; (B.M.); (W.F.A.M.); (I.X.); (L.L.); (A.S.)
| | - Waleed F. A. Marei
- Gamete Research Centre, Laboratory of Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, 2610 Antwerp, Belgium; (B.M.); (W.F.A.M.); (I.X.); (L.L.); (A.S.)
- Faculty of Veterinary Medicine, Department of Theriogenology, Cairo University, Giza 12211, Egypt
| | - Inne Xhonneux
- Gamete Research Centre, Laboratory of Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, 2610 Antwerp, Belgium; (B.M.); (W.F.A.M.); (I.X.); (L.L.); (A.S.)
| | - Lien Loier
- Gamete Research Centre, Laboratory of Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, 2610 Antwerp, Belgium; (B.M.); (W.F.A.M.); (I.X.); (L.L.); (A.S.)
| | - Anouk Smits
- Gamete Research Centre, Laboratory of Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, 2610 Antwerp, Belgium; (B.M.); (W.F.A.M.); (I.X.); (L.L.); (A.S.)
| | - Jo L. M. R. Leroy
- Gamete Research Centre, Laboratory of Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, 2610 Antwerp, Belgium; (B.M.); (W.F.A.M.); (I.X.); (L.L.); (A.S.)
| |
Collapse
|
3
|
Nandi S, Tripathi SK, Singh PK, Gupta PSP, Mondal S. Global DNA methylation, DNA methyltransferase and stress-related gene expression in ovine oocytes and embryos after exposure to metabolic stressors. Reprod Domest Anim 2023. [PMID: 36920043 DOI: 10.1111/rda.14341] [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: 01/25/2023] [Revised: 02/20/2023] [Accepted: 03/09/2023] [Indexed: 03/16/2023]
Abstract
DNA methylation, considered the most prominent epigenetic mark was important for the gene regulation in embryonic development. The present study aimed at evaluating the effects of metabolic stressors [Non-esterified fatty acid (NEFA), β-hydroxy-butyric acid (BHB), ammonia and urea] exposure during the in vitro ovine oocyte maturation, global DNA methylation, DNA methyltransferase and stress-related gene expression. Colorimetric analysis of global DNA methylation and the expression of the DNA methyltransferase genes (DNMT1, DNMT3A, and DNMT3B) were assessed in the matured oocytes, 2-cell embryos and blastocysts produced in vitro from oocytes exposed with the metabolic stressors during 24 h of the in vitro maturation (IVM). Further, the mRNA expression of the stress-related genes (SOD1, SOD2) in the matured oocytes, 2-cell embryos and blastocysts produced was assessed. Significant difference in global DNA methylation levels between all the treatments tested was observed when compared with control in oocytes, two-cell embryos and blastocysts. Elevated concentration of metabolic stressors resulted in increased expressions of several stress-related genes, i.e., SOD1, SOD2 and in mRNA expression of DNA methyltransferase genes. The present study is the first to report that the DNA methylation was sensitive to the effects of the metabolic stressors in ovine oocytes/embryos. The aberrant expressions of genes during oocyte development targeted in the present study can provide evidence for the early embryo developmental arrest and blastocysts quality. These results highlighted the sensitivity of the early embryogenesis and more precisely of the reprogramming period to metabolites challenges, in a realistic situation of elevated concentration of metabolic stressors.
Collapse
Affiliation(s)
- Sumanta Nandi
- Animal Biotechnology Laboratory, ICAR-National Institute of Animal Nutrition and Physiology, Bangalore, 560030, India
| | - Shiv K Tripathi
- Animal Biotechnology Laboratory, ICAR-National Institute of Animal Nutrition and Physiology, Bangalore, 560030, India
| | - Poonam K Singh
- Animal Biotechnology Laboratory, ICAR-National Institute of Animal Nutrition and Physiology, Bangalore, 560030, India
| | - Paluru S P Gupta
- Animal Biotechnology Laboratory, ICAR-National Institute of Animal Nutrition and Physiology, Bangalore, 560030, India
| | - Sukanta Mondal
- Animal Biotechnology Laboratory, ICAR-National Institute of Animal Nutrition and Physiology, Bangalore, 560030, India
| |
Collapse
|
4
|
Chao S, Li LJ, Lu J, Zhao SX, Zhao MH, Huang GA, Yin S, Shen W, Sun QY, Zhao Y, Ge ZJ, Zhao L. Epigallocatechin gallate improves the quality of diabetic oocytes. Biomed Pharmacother 2023; 159:114267. [PMID: 36669363 DOI: 10.1016/j.biopha.2023.114267] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/12/2023] [Accepted: 01/16/2023] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Maternal diabetes compromises the quality and developmental potential of oocytes. Therefore, it is important to study how to ameliorate the adverse effects of diabetes on oocyte quality. Epigallocatechin gallate (EGCG) has a variety of physiological activities, including anti-inflammatory, antioxidant, and anti-diabetes. In the present study, we evaluated the effect of EGCG on the maturation of diabetic oocytes in vitro. OBJECTIVE Investigating the role of EGCG in restoring the adverse effects of diabetes on oocyte quality. METHODS Diabetes mouse model was established by a single injection of streptozotocin (STZ). Oocytes were collected and matured in vitro with/without EGCG in M16 medium. RESULTS Compared with control, diabetic oocytes have a higher frequency of spindle defects and chromosome misalignment, but EGCG effectively reduces the incidence of oocytes with abnormal spindle assembly and chromosome mismatches. Moreover, the abnormal mitochondrial membrane potential (MMP) of diabetic oocytes is significantly alleviated by EGCG, and the reduced expression of genes regulating mitochondrial fusion (Mfn1 and Mfn2) and fission (Drp1) in diabetic oocytes is significantly increased while EGCG is added. EGCG also decreases the higher level of reactive oxygen species (ROS) in diabetic oocytes that may be regulated by the increased expression of superoxide dismutase 1 (Sod1) and superoxide dismutase 2 (Sod2). EGCG can also reduce the DNA damage of diabetic oocytes. CONCLUSIONS Our results suggest that EGCG, at least partially, improve the quality of diabetic oocytes.
Collapse
Affiliation(s)
- Shuo Chao
- College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, PR China
| | - Li-Jun Li
- College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, PR China
| | - Jun Lu
- College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, PR China
| | - Shu-Xian Zhao
- College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, PR China
| | - Ming-Hui Zhao
- College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, PR China
| | - Gui-An Huang
- College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, PR China
| | - Shen Yin
- College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, PR China
| | - Wei Shen
- College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, PR China
| | - Qing-Yuan Sun
- Fertility Preservation Lab and Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou 510317, PR China
| | - Yong Zhao
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, PR China
| | - Zhao-Jia Ge
- College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, PR China.
| | - Lei Zhao
- College of Horticulture, Qingdao Agricultural University, Qingdao 266109, PR China.
| |
Collapse
|
5
|
Tang SB, Zhang TT, Yin S, Shen W, Luo SM, Zhao Y, Zhang CL, Klinger FG, Sun QY, Ge ZJ. Inheritance of perturbed methylation and metabolism caused by uterine malnutrition via oocytes. BMC Biol 2023; 21:43. [PMID: 36829148 PMCID: PMC9960220 DOI: 10.1186/s12915-023-01545-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 02/13/2023] [Indexed: 02/26/2023] Open
Abstract
BACKGROUND Undernourishment in utero has deleterious effects on the metabolism of offspring, but the mechanism of the transgenerational transmission of metabolic disorders is not well known. In the present study, we found that undernourishment in utero resulted in metabolic disorders of female F1 and F2 in mouse model. RESULTS Undernutrition in utero induced metabolic disorders of F1 females, which was transmitted to F2 females. The global methylation in oocytes of F1 exposed to undernutrition in utero was decreased compared with the control. KEGG analysis showed that genes with differential methylation regions (DMRs) in promoters were significantly enriched in metabolic pathways. The altered methylation of some DMRs in F1 oocytes located at the promoters of metabolic-related genes were partially observed in F2 tissues, and the expressions of these genes were also changed. Meanwhile, the abnormal DNA methylation of the validated DMRs in F1 oocytes was also observed in F2 oocytes. CONCLUSIONS These results indicate that DNA methylation may mediate the transgenerational inheritance of metabolic disorders induced by undernourishment in utero via female germline.
Collapse
Affiliation(s)
- Shou-Bin Tang
- College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China
| | - Ting-Ting Zhang
- College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China
- Reproductive Medicine Center, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, 450003, People's Republic of China
| | - Shen Yin
- College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China
| | - Wei Shen
- College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China
| | - Shi-Ming Luo
- Fertility Preservation Lab and Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, People's Republic of China
| | - Yong Zhao
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, People's Republic of China
| | - Cui-Lian Zhang
- Reproductive Medicine Center, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, 450003, People's Republic of China
| | - Francesca Gioia Klinger
- Histology and Embryology, Saint Camillus International University of Health Sciences, Rome, Italy
| | - Qing-Yuan Sun
- Fertility Preservation Lab and Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, People's Republic of China.
| | - Zhao-Jia Ge
- College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China.
| |
Collapse
|
6
|
Overexpression of Tfap2a in Mouse Oocytes Impaired Spindle and Chromosome Organization. Int J Mol Sci 2022; 23:ijms232214376. [PMID: 36430853 PMCID: PMC9699359 DOI: 10.3390/ijms232214376] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/14/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
Transcription factor AP-2-alpha (Tfap2a) is an important sequence-specific DNA-binding protein that can regulate the transcription of multiple genes by collaborating with inducible viral and cellular enhancer elements. In this experiment, the expression, localization, and functions of Tfap2a were investigated in mouse oocytes during maturation. Overexpression via microinjection of Myc-Tfap2a mRNA into the ooplasm, immunofluorescence, and immunoblotting were used to study the role of Tfap2a in mouse oocyte meiosis. According to our results, Tfap2a plays a vital role in mouse oocyte maturation. Levels of Tfap2a in GV oocytes of mice suffering from type 2 diabetes increased considerably. Tfap2a was distributed in both the ooplasm and nucleoplasm, and its level gradually increased as meiosis resumption progressed. The overexpression of Tfap2a loosened the chromatin, accelerated germinal vesicle breakdown (GVBD), and blocked the first polar body extrusion 14 h after maturation in vitro. The width of the metaphase plate at metaphase I stage increased, and the spindle and chromosome organization at metaphase II stage were disrupted in the oocytes by overexpressed Tfap2a. Furthermore, Tfap2a overexpression dramatically boosted the expression of p300 in mouse GV oocytes. Additionally, the levels of pan histone lysine acetylation (Pan Kac), histone H4 lysine 12 acetylation (H4K12ac), and H4 lysine 16 acetylation (H4K16ac), as well as pan histone lysine lactylation (Pan Kla), histone H3 lysine18 lactylation (H3K18la), and H4 lysine12 lactylation (H4K12la), were all increased in GV oocytes after Tfap2a overexpression. Collectively, Tfap2a overexpression upregulated p300, increased the levels of histone acetylation and lactylation, impeded spindle assembly and chromosome alignment, and ultimately hindered mouse oocyte meiosis.
Collapse
|
7
|
Sheth VG, Sharma N, Kabeer SW, Tikoo K. Lactobacillus rhamnosus supplementation ameliorates high fat diet-induced epigenetic alterations and prevents its intergenerational inheritance. Life Sci 2022; 311:121151. [DOI: 10.1016/j.lfs.2022.121151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/20/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022]
|
8
|
Lu J, Zhao SX, Zhang MY, Ji PY, Chao S, Li LJ, Yin S, Zhao L, Zhao H, Sun QY, Ge ZJ. Tea polyphenols alleviate the adverse effects of diabetes on oocyte quality. Food Funct 2022; 13:5396-5405. [PMID: 35471225 DOI: 10.1039/d1fo03770f] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Maternal diabetes mellitus reduces oocyte quality, such as abnormalities of spindle assembly and chromosome segregation, mitochondrial dysfunction, decrease of fertilization rate, increase of ROS, and so on. So, it is important to research how to restore the decreased oocyte quality induced by maternal diabetes mellitus. Polyphenols are the most abundant bioactive components of green tea. It is reported that tea polyphenols have many health functions, for instance anti-oxidation, anti-inflammation, anti-obesity, and anti-diabetes. Thus, we hypothesize that tea polyphenols may play a crucial role in alleviating adverse effects of diabetes on oocyte quality. In the present study, we researched the effects of tea polyphenols on diabetic oocyte maturation in vitro. Compared with the control, oocytes from diabetic mice displayed a lower maturation rate and a higher frequency of spindle defects and chromosome misalignment. However, tea polyphenols significantly increased the oocyte maturation rate, and reduced the incidence of abnormal spindle assembly and chromosome segregation. Tea polyphenols also obviously decreased the reactive oxygen species (ROS) levels in diabetic oocytes, and increased the expression of antioxidant genes (Sod1 and Sod2). Abnormal mitochondrial membrane potential was also alleviated in diabetic oocytes, and the expression of genes regulating mitochondrial fusion (Opa1, Mfn1 and Mfn2) and fission (Drp1) was significantly increased while tea polyphenols were added. Meanwhile, tea polyphenols reduced DNA damage in diabetic oocytes which may be mediated by the increased expression of Rad51, related to DNA damage repair. Our results suggest that tea polyphenols would, at least partially, restore the adverse effects of diabetes mellitus on oocyte quality.
Collapse
Affiliation(s)
- Jun Lu
- College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, P. R. China.
| | - Shu-Xian Zhao
- College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, P. R. China.
| | - Man-Yu Zhang
- College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, P. R. China.
| | - Peng-Yuan Ji
- College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, P. R. China.
| | - Shuo Chao
- College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, P. R. China.
| | - Li-Jun Li
- College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, P. R. China.
| | - Shen Yin
- College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, P. R. China.
| | - Lei Zhao
- College of Horticulture, Qingdao Agricultural University, Qingdao 266109, P. R. China
| | - Hua Zhao
- Reproductive Medicine Center, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou 450003, P. R. China
| | - Qing-Yuan Sun
- Fertility Preservation Lab and Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou 510317, China.
| | - Zhao-Jia Ge
- College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, P. R. China.
| |
Collapse
|
9
|
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
|
10
|
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
|
11
|
Imprinted lncRNA Dio3os preprograms intergenerational brown fat development and obesity resistance. Nat Commun 2021; 12:6845. [PMID: 34824246 PMCID: PMC8617289 DOI: 10.1038/s41467-021-27171-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 11/08/2021] [Indexed: 12/17/2022] Open
Abstract
Maternal obesity (MO) predisposes offspring to obesity and metabolic disorders but little is known about the contribution of offspring brown adipose tissue (BAT). We find that MO impairs fetal BAT development, which persistently suppresses BAT thermogenesis and primes female offspring to metabolic dysfunction. In fetal BAT, MO enhances expression of Dio3, which encodes deiodinase 3 (D3) to catabolize triiodothyronine (T3), while a maternally imprinted long noncoding RNA, Dio3 antisense RNA (Dio3os), is inhibited, leading to intracellular T3 deficiency and suppression of BAT development. Gain and loss of function shows Dio3os reduces D3 content and enhances BAT thermogenesis, rendering female offspring resistant to high fat diet-induced obesity. Attributing to Dio3os inactivation, its promoter has higher DNA methylation in obese dam oocytes which persists in fetal and adult BAT, uncovering an oocyte origin of intergenerational obesity. Overall, our data uncover key features of Dio3os activation in BAT to prevent intergenerational obesity and metabolic dysfunctions. Maternal obesity predisposes offspring to obesity and metabolic disorders through incompletely understood mechanisms. Here the authors report that Dio3os is an imprinted long-coding RNA that modulates brown adipose tissue development and obesity resistance in the offspring.
Collapse
|
12
|
Abstract
Tea is the second most popular beverage in the world and beneficial to health. It has been demonstrated that tea polyphenols can reduce the risk of diseases, such as cancers, diabetes, obesity, Alzheimer's disease, etc. But the knowledge of tea extract on the female germline is limited. Folliculogenesis is a complicated process and prone to be affected by ROS. Tea polyphenols can reduce the accumulation of ROS in folliculogenesis and affect oocyte maturation. Tea extract also influences granulosa cell proliferation and expansion during oocyte growth and maturation. However, the studies about the benefits of tea extract on female germline are few, and the underlying mechanisms are obscure. In the present study, we will mainly discuss the effects of tea extract on ovarian function, oocyte maturation, and the underlying possible mechanisms, and according to the discussion, we suggest that tea extract may have benefits for oocytes at an appropriate dose.
Collapse
Affiliation(s)
- Lei Zhao
- College of Horticulture, Qingdao Agricultural University, Qingdao, Shandong, China
| | - Qing-Yuan Sun
- College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao, P.R. China.
- Fertility Preservation Lab and Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou 510317, China
| | - Zhao-Jia Ge
- College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao, P.R. China.
| |
Collapse
|
13
|
Dong MZ, Li QN, Fan LH, Li L, Shen W, Wang ZB, Sun QY. Diabetic Uterine Environment Leads to Disorders in Metabolism of Offspring. Front Cell Dev Biol 2021; 9:706879. [PMID: 34381787 PMCID: PMC8350518 DOI: 10.3389/fcell.2021.706879] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 06/28/2021] [Indexed: 12/11/2022] Open
Abstract
Aims Research evidence indicates that epigenetic modifications of gametes in obese or diabetic parents may contribute to metabolic disorders in offspring. In the present study, we sought to address the effect of diabetic uterine environment on the offspring metabolism. Methods Type 2 diabetes mouse model was induced by high-fat diet combined with streptozotocin (STZ) administration. We maintained other effect factors constant and changed uterine environment by zygote transfers, and then determined and compared the offspring numbers, symptoms, body weight trajectories, and metabolism indices from different groups. Result We found that maternal type 2 diabetes mice had lower fertility and a higher dystocia rate, accompanying the increased risk of offspring malformations and death. Compared to only a pre-gestational exposure to hyperglycemia, exposure to hyperglycemia both pre- and during pregnancy resulted in offspring growth restriction and impaired metabolism in adulthood. But there was no significant difference between a pre-gestational exposure group and a no exposure group. The deleterious effects, no matter bodyweight or glucose tolerance, could be rescued by transferring the embryos from diabetic mothers into normal uterine environment. Conclusion Our data demonstrate that uterine environment of maternal diabetes makes critical impact on the offspring health.
Collapse
Affiliation(s)
- Ming-Zhe Dong
- Institute of Reproductive Science, College of Life Sciences, Qingdao Agricultural University, Qingdao, China.,State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Qian-Nan Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Li-Hua Fan
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Li Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Wei Shen
- Institute of Reproductive Science, College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | - Zhen-Bo Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Qing-Yuan Sun
- Fertility Preservation Lab, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China
| |
Collapse
|
14
|
Bacon ER, Brinton RD. Epigenetics of the developing and aging brain: Mechanisms that regulate onset and outcomes of brain reorganization. Neurosci Biobehav Rev 2021; 125:503-516. [PMID: 33657435 DOI: 10.1016/j.neubiorev.2021.02.040] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 02/17/2021] [Accepted: 02/23/2021] [Indexed: 12/11/2022]
Abstract
Brain development is a life-long process that encompasses several critical periods of transition, during which significant cognitive changes occur. Embryonic development, puberty, and reproductive senescence are all periods of transition that are hypersensitive to environmental factors. Rather than isolated episodes, each transition builds upon the last and is influenced by consequential changes that occur in the transition before it. Epigenetic marks, such as DNA methylation and histone modifications, provide mechanisms by which early events can influence development, cognition, and health outcomes. For example, parental environment influences imprinting patterns in gamete cells, which ultimately impacts gene expression in the embryo which may result in hypersensitivity to poor maternal nutrition during pregnancy, raising the risks for cognitive impairment later in life. This review explores how epigenetics induce and regulate critical periods, and also discusses how early environmental interactions prime a system towards a particular health outcome and influence susceptibility to disease or cognitive impairment throughout life.
Collapse
Affiliation(s)
- Eliza R Bacon
- Department of Neuroscience, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA, 90089, USA; The Center for Precision Medicine, Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA
| | - Roberta Diaz Brinton
- Department of Neuroscience, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA, 90089, USA; Center for Innovation in Brain Science, School of Medicine, University of Arizona, Tucson, AZ, 85721, USA.
| |
Collapse
|
15
|
Villanueva-Hayes C, Millership SJ. Imprinted Genes Impact Upon Beta Cell Function in the Current (and Potentially Next) Generation. Front Endocrinol (Lausanne) 2021; 12:660532. [PMID: 33986727 PMCID: PMC8112240 DOI: 10.3389/fendo.2021.660532] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 04/01/2021] [Indexed: 11/23/2022] Open
Abstract
Beta cell failure lies at the centre of the aetiology and pathogenesis of type 2 diabetes and the epigenetic control of the expression of critical beta cell genes appears to play a major role in this decline. One such group of epigenetically-controlled genes, termed 'imprinted' genes, are characterised by transgenerational monoallelic expression due to differential allelic DNA methylation and play key functional roles within beta cells. Here, we review the evidence for this functional importance of imprinted genes in beta cells as well as their nutritional regulation by the diet and their altered methylation and/or expression in rodent models of diabetes and in type 2 diabetic islets. We also discuss imprinted genes in the context of the next generation, where dietary overnutrition in the parents can lead to their deregulation in the offspring, alongside beta cell dysfunction and defective glucose handling. Both the modulation of imprinted gene expression and the likelihood of developing type 2 diabetes in adulthood are susceptible to the impact of nutritional status in early life. Imprinted loci, therefore, represent an excellent opportunity with which to assess epigenomic changes in beta cells due to the diet in both the current and next generation.
Collapse
|
16
|
Alves JPM, Fernandes CCL, Calderón CEM, Rossetto R, Bertolini M, Rondina D. Short-term supplementation of diets rich in lipids or glycogen precursors can affect intra-follicular environment, oocyte mitochondrial gene expression, and embryo development following parthenogenesis in goat. Small Rumin Res 2021. [DOI: 10.1016/j.smallrumres.2020.106279] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
17
|
Xin Y, Jin Y, Ge J, Huang Z, Han L, Li C, Wang D, Zhu S, Wang Q. Involvement of SIRT3-GSK3β deacetylation pathway in the effects of maternal diabetes on oocyte meiosis. Cell Prolif 2020; 54:e12940. [PMID: 33107080 PMCID: PMC7791178 DOI: 10.1111/cpr.12940] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/16/2020] [Accepted: 10/03/2020] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVES It has been widely reported that maternal diabetes impairs oocyte quality. However, the responsible mechanisms remain to be explored. In the present study, we focused on whether SIRT3-GSK3β pathway mediates the meiotic defects in oocytes from diabetic mice. MATERIALS AND METHODS GSK3β functions in mouse oocyte meiosis were first detected by targeted siRNA knockdown. Spindle assembly and chromosome alignment were visualized by immunostaining and analysed under the confocal microscope. PCR-based site mutation of specific GSK3β lysine residues was used to confirm which lysine residues function in oocyte meiosis. siRNA knockdown coupled with cRNA overexpression was performed to detect SIRT3-GSK3β pathway functions in oocyte meiosis. Immunofluorescence was performed to detect ROS levels. T1DM mouse models were induced by a single intraperitoneal injection of streptozotocin. RESULTS In the present study, we found that specific depletion of GSK3β disrupts maturational progression and meiotic apparatus in mouse oocytes. By constructing site-specific mutants, we further revealed that acetylation state of lysine (K) 15 on GSK3β is essential for spindle assembly and chromosome alignment during oocyte meiosis. Moreover, non-acetylation-mimetic mutant GSK3β-K15R is capable of partly preventing the spindle/chromosome anomalies in oocytes with SIRT3 knockdown. A significant reduction in SIRT3 protein was detected in oocytes from diabetic mice. Of note, forced expression of GSK3β-K15R ameliorates maternal diabetes-associated meiotic defects in mouse oocytes, with no evident effects on oxidative stress. CONCLUSION Our data identify GSK3β as a cytoskeletal regulator that is required for the assembly of meiotic apparatus, and discover a beneficial effect of SIRT3-dependent GSK3β deacetylation on oocyte quality from diabetic mice.
Collapse
Affiliation(s)
- Yongan Xin
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, Nanjing, China
| | - Yifei Jin
- School of Nursing, Nanjing Medical University, Nanjing, China
| | - Juan Ge
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, Nanjing, China
| | - Zhenyue Huang
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, Nanjing, China
| | - Longsen Han
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, Nanjing, China
| | - Congyang Li
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, Nanjing, China
| | - Danni Wang
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, Nanjing, China
| | - Shuai Zhu
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, Nanjing, China
| | - Qiang Wang
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, Nanjing, China.,Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
18
|
He QL, Lyu TQ, Zhang YT, Wang HQ, Zhou Q, Zhang JM, Liu YY, Li JS, Jiang LG, Cheng D, Ge ZJ, Liu SZ. Effects of intrauterine exposure to 2,3',4,4',5-pentachlorobiphenyl on the reproductive system and sperm epigenetic imprinting of male offspring. J Appl Toxicol 2020; 40:1396-1409. [PMID: 32418265 DOI: 10.1002/jat.3992] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 03/30/2020] [Accepted: 04/04/2020] [Indexed: 12/11/2022]
Abstract
Polychlorinated biphenyls (PCBs) are a class of persistent organic environmental pollutants with a total of 209 homologs. The homolog 2,3',4,4',5-pentachlorobiphenyl (PCB118) is one of the most important dioxin-like PCBs and is highly toxic. PCB118 can accumulate in human tissues, serum and breast milk, which leads to direct exposure of the fetus during development. In the present study, pregnant mice were exposed to 0, 20 and 100 μg/kg/day of PCB118 during the stage of fetal primordial germ cell migration. Compared with the control group, we found morphological alterations of the seminiferous tubules and a higher sperm deformity rate in the male offspring in the treatment groups. Furthermore, the methylation patterns in the treatment groups of the imprinted genes H19 and Gtl2 in the sperm were altered in the male offspring. We also characterized the disturbance of the expression levels of DNA methyltransferase 1 (Dnmt1), Dnmt3a, Dnmt3b, Dnmt3l, and Uhrf1. The results indicated that intrauterine exposure to low doses of PCB118 could significantly damage the reproductive health of the male offspring. Therefore, attention should be paid to the adverse effects of PCB118 exposure during pregnancy on the reproductive system of male offspring.
Collapse
Affiliation(s)
- Qi-Long He
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Tian-Qi Lyu
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Yong-Tao Zhang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Hai-Quan Wang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Qian Zhou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Jian-Mei Zhang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Yuan-Yuan Liu
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Jia-Shuo Li
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Li-Gang Jiang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Dong Cheng
- Department of Toxicology, Shandong Center for Disease Control and Prevention, Jinan, China
| | - Zhao-Jia Ge
- College of Life Sciences, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao, China
| | - Shu-Zhen Liu
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, China
| |
Collapse
|
19
|
Li L, Jing Y, Dong MZ, Fan LH, Li QN, Wang ZB, Hou Y, Schatten H, Zhang CL, Sun QY. Type 1 diabetes affects zona pellucida and genome methylation in oocytes and granulosa cells. Mol Cell Endocrinol 2020; 500:110627. [PMID: 31639403 DOI: 10.1016/j.mce.2019.110627] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 10/17/2019] [Accepted: 10/17/2019] [Indexed: 11/17/2022]
Abstract
Diabetes affects oocyte nuclear and cytoplasmic quality. In this study, we generated a type 1 diabetes (T1D) mouse model by STZ injection to study the effects of T1D on zona pellucida and genomic DNA methylation of oocytes and granulosa cells. T1D mice showed fewer ovulated oocytes, reduced ovarian reserve, disrupted estrus cycle, and significantly ruptured zona pellucida in 2-cell in vivo embryos compared to controls. Notably, diabetic oocytes displayed thinner zona pellucida and treatment of oocytes with high concentration glucose reduced the zona pellucida thickness. Differential methylation genes in oocytes and granulosa cells were analyzed by methylation sequencing. These genes were significantly enriched in GO terms by GO analysis, and these GO terms were involved in multiple aspects of growth and development. Most notably, the abnormal methylation genes in oocytes may be related to oocyte zona pellucida changes in diabetic mice. These findings provide novel basic data for further understanding and elucidating dysgenesis and epigenetic changes in type 1 diabetes mellitus.
Collapse
Affiliation(s)
- Li Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Ying Jing
- Reproductive Medicine Center of People's Hospital of Zhengzhou University, Zhengzhou, 450003, Henan Province, PR China; Reproductive Medicine Center of Henan Provincial People's Hospital, Zhengzhou, 450003, Henan Province, PR China
| | - Ming-Zhe Dong
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Li-Hua Fan
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Qian-Nan Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Zhen-Bo Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Yi Hou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Heide Schatten
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, 65211, USA
| | - Cui-Lian Zhang
- Reproductive Medicine Center of People's Hospital of Zhengzhou University, Zhengzhou, 450003, Henan Province, PR China; Reproductive Medicine Center of Henan Provincial People's Hospital, Zhengzhou, 450003, Henan Province, PR China.
| | - Qing-Yuan Sun
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
20
|
Xu J, Huang J, Pan Q, Du M, Li Z, Dong H. Gestational diabetes promotes germ cell cCyst breakdown and primordial follicle formation in newborn mice via the AKT signaling pathway. PLoS One 2019; 14:e0215007. [PMID: 30973884 PMCID: PMC6459533 DOI: 10.1371/journal.pone.0215007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 03/25/2019] [Indexed: 01/23/2023] Open
Abstract
Type 1 diabetes (T1D) is a common disease in which pancreatic β cells are impaired due to auto-immunity, pregnancy in women with it is associated with increased risk of neonatal morbidity, mortality. However, the effects of gestational diabetes on the reproduction of newborn offspring are still poorly understood. Here, we determined the cyst breakdown and primordial follicle formation in neonatal offspring born by streptozotocin (STZ)-induced diabetic or non-diabetic female mice, and found that the germ cell cyst breakdown was promoted in neonatal offspring of STZ -induced diabetic mice at postnatal Day 1, which sequentially accelerated the primordial follicle formation. Further investigation revealed that, the expression level of PI3K and p-AKT were significantly increased in ovaries of offspring born by T1D mice. These results indicated that STZ -induced gestational diabetes promotes germ cell cyst breakdown and primordial follicle formation by regulating the PI3K/AKT signaling pathway in the newborn offspring. In addition, this effect can be rescued by an insulin supplement. Taken together, our results uncover the intergenerational effects of gestational diabetes on neonatal offspring folliculogenesis, and provide an experimental model for treating gestational diabetes and its complications in neonatal offspring.
Collapse
Affiliation(s)
- Junjun Xu
- Department of Animal Science and Technology, Qingdao Agricultural University, Qingdao, China
| | - Jiaojiao Huang
- Department of Animal Science and Technology, Qingdao Agricultural University, Qingdao, China
| | - Qingjie Pan
- Department of Animal Science and Technology, Qingdao Agricultural University, Qingdao, China
| | - Miao Du
- Department of Animal Science and Technology, Qingdao Agricultural University, Qingdao, China
| | - Zhen Li
- Department of Animal Science and Technology, Qingdao Agricultural University, Qingdao, China
| | - Huansheng Dong
- Department of Animal Science and Technology, Qingdao Agricultural University, Qingdao, China
- * E-mail:
| |
Collapse
|
21
|
Zou K, Ding G, Huang H. Advances in research into gamete and embryo-fetal origins of adult diseases. SCIENCE CHINA-LIFE SCIENCES 2019; 62:360-368. [PMID: 30685828 DOI: 10.1007/s11427-018-9427-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 09/19/2018] [Indexed: 12/11/2022]
Abstract
The fetal and infant origins of adult disease hypothesis proposed that the roots of adult chronic disease lie in the effects of adverse environments in fetal life and early infancy. In addition to the fetal period, fertilization and early embryonic stages, the critical time windows of epigenetic reprogramming, rapid cell differentiation and organogenesis, are the most sensitive stages to environmental disturbances. Compared with embryo and fetal development, gametogenesis and maturation take decades and are more vulnerable to potential damage for a longer exposure period. Therefore, we should shift the focus of adult disease occurrence and pathogenesis further back to gametogenesis and embryonic development events, which may result in intergenerational, even transgenerational, epigenetic re-programming with transmission of adverse traits and characteristics to offspring. Here, we focus on the research progress relating to diseases that originated from events in the gametes and early embryos and the potential epigenetic mechanisms involved.
Collapse
Affiliation(s)
- Kexin Zou
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China.,Institute of Embryo-Fetal Original Adult Disease, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Guolian Ding
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China.,Institute of Embryo-Fetal Original Adult Disease, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Hefeng Huang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China. .,Institute of Embryo-Fetal Original Adult Disease, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China.
| |
Collapse
|
22
|
Ou X, Zhu C, Sun S. Effects of obesity and diabetes on the epigenetic modification of mammalian gametes. J Cell Physiol 2018; 234:7847-7855. [DOI: 10.1002/jcp.27847] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 11/15/2018] [Indexed: 12/22/2022]
Affiliation(s)
- Xiang‐Hong Ou
- Fertility Preservation Lab, Reproductive Medicine Center, Guangdong Second Provincial General Hospital Guangzhou China
| | - Cheng‐Cheng Zhu
- College of Animal Science and Technology, Nanjing Agricultural University Nanjing China
- Nanjing Police Dog Institute of the Ministry of Public Security Nanjing China
| | - Shao‐Chen Sun
- College of Animal Science and Technology, Nanjing Agricultural University Nanjing China
| |
Collapse
|
23
|
Jing Y, Li L, Li Y, Ouyang Y, Sun Q, Zhang C, Li R. Embryo quality, and not chromosome nondiploidy, affects mitochondrial DNA content in mouse blastocysts. J Cell Physiol 2018; 234:10481-10488. [PMID: 30480815 DOI: 10.1002/jcp.27713] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Accepted: 10/16/2018] [Indexed: 01/08/2023]
Affiliation(s)
- Ying Jing
- Reproductive Medicine Center, The People's Hospital of Zhengzhou University Zhengzhou China
- Reproductive Medicine Center, The People's Hospital of Henan Province Zhengzhou China
- State Key Laboratory of Stem Cell and Reproductive Biology Institute of Zoology, Chinese Academy of Sciences Beijing China
| | - Li Li
- State Key Laboratory of Stem Cell and Reproductive Biology Institute of Zoology, Chinese Academy of Sciences Beijing China
| | - Yuan‐Yuan Li
- State Key Laboratory of Stem Cell and Reproductive Biology Institute of Zoology, Chinese Academy of Sciences Beijing China
| | - Ying‐Chun Ouyang
- State Key Laboratory of Stem Cell and Reproductive Biology Institute of Zoology, Chinese Academy of Sciences Beijing China
| | - Qing‐Yuan Sun
- State Key Laboratory of Stem Cell and Reproductive Biology Institute of Zoology, Chinese Academy of Sciences Beijing China
| | - Cui‐Lian Zhang
- Reproductive Medicine Center, The People's Hospital of Zhengzhou University Zhengzhou China
- Reproductive Medicine Center, The People's Hospital of Henan Province Zhengzhou China
| | - Rong Li
- Department of Obstetrics and Gynecology Center for Reproductive Medicine, Peking University Third Hospital Beijing China
| |
Collapse
|
24
|
Wang Q, Tang SB, Song XB, Deng TF, Zhang TT, Yin S, Luo SM, Shen W, Zhang CL, Ge ZJ. High-glucose concentrations change DNA methylation levels in human IVM oocytes. Hum Reprod 2018; 33:474-481. [PMID: 29377995 DOI: 10.1093/humrep/dey006] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 01/12/2018] [Indexed: 01/01/2023] Open
Abstract
STUDY QUESTION What are the effects of high-glucose concentrations on DNA methylation of human oocytes? SUMMARY ANSWER High-glucose concentrations altered DNA methylation levels of Peg3 and Adiponectin in human in vitro maturation oocytes. WHAT IS KNOWN ALREADY Maternal diabetes has a detrimental influence on oocyte quality including epigenetic modifications, as shown in non-human mammalian species. STUDY DESIGN, SIZE, DURATION Immature metaphase I (MI) stage oocytes of good quality were retrieved from patients who had normal ovarian potential and who underwent ICSI in the Reproductive Medicine Center of People's Hospital of Zhengzhou University. MI oocytes were cultured in medium with different glucose concentrations (control, 10 mM and 15 mM) in vitro and 48 h later, oocytes with first polar body extrusion were collected to check the DNA methylation levels. PARTICIPANTS/MATERIALS, SETTING, METHODS MI oocytes underwent in vitro maturation (IVM) at 37°C with 5% mixed gas for 48 h. Then the mature oocytes were treated with bisulfite buffer. Target sequences were amplified using nested or half-nested PCR and the DNA methylation status was tested using combined bisulfite restriction analysis (COBRA) and bisulfite sequencing (BS). MAIN RESULTS AND THE ROLE OF CHANCE High-glucose concentrations significantly decreased the first polar body extrusion rate. Compared to controls, the DNA methylation levels of Peg3 in human IVM oocytes were significantly higher in 10 mM (P < 0.001) and 15 mM (P < 0.001) concentrations of glucose. But the DNA methylation level of H19 was not affected by high-glucose concentrations in human IVM oocytes. We also found that there was a decrease in DNA methylation levels in the promoter of adiponectin in human IVM oocytes between controls and oocytes exposed to 10 mM glucose (P = 0.028). LARGE SCALE DATA N/A. LIMITATIONS REASONS FOR CAUTION It is not clear whether the alterations are beneficial or not for the embryo development and offspring health. The effects of high-glucose concentrations on the whole process of oocyte maturation are still not elucidated. Another issue is that the number of oocytes used in this study was limited. WIDER IMPLICATIONS OF THE FINDINGS This is the first time that the effects of high-glucose concentration on DNA methylation of human oocytes have been elucidated. Our result indicates that in humans, the high risk of chronic diseases in offspring from diabetic mothers may originate from abnormal DNA modifications in oocytes. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by the fund of National Natural Science Foundation of China (81401198) and Doctor Foundation of Qingdao Agricultural University (1116008).The authors declare that there are no potential conflicts of interest relevant to this article.
Collapse
Affiliation(s)
- Qian Wang
- Reproductive Medicine Center of People's Hospital of Zhengzhou University, Zhengzhou 450003, Henan Province, P.R. China
- Reproductive Medicine Center of Henan Provincial People's Hospital, Zhengzhou 450003, Henan Province, P.R. China
| | - Shou-Bin Tang
- Department of Biology, College of Life Sciences, Institute of Reproductive Sciences, Qingdao Agricultural University, 700# Changcheng Road, Chengyang District, Qingdao 266109, P.R. China
| | - Xiao-Bing Song
- Reproductive Medicine Center of People's Hospital of Zhengzhou University, Zhengzhou 450003, Henan Province, P.R. China
- Reproductive Medicine Center of Henan Provincial People's Hospital, Zhengzhou 450003, Henan Province, P.R. China
| | - Teng-Fei Deng
- Reproductive Medicine Center of People's Hospital of Zhengzhou University, Zhengzhou 450003, Henan Province, P.R. China
- Reproductive Medicine Center of Henan Provincial People's Hospital, Zhengzhou 450003, Henan Province, P.R. China
| | - Ting-Ting Zhang
- Reproductive Medicine Center of People's Hospital of Zhengzhou University, Zhengzhou 450003, Henan Province, P.R. China
- Reproductive Medicine Center of Henan Provincial People's Hospital, Zhengzhou 450003, Henan Province, P.R. China
| | - Shen Yin
- Department of Biology, College of Life Sciences, Institute of Reproductive Sciences, Qingdao Agricultural University, 700# Changcheng Road, Chengyang District, Qingdao 266109, P.R. China
| | - Shi-Ming Luo
- Department of Biology, College of Life Sciences, Institute of Reproductive Sciences, Qingdao Agricultural University, 700# Changcheng Road, Chengyang District, Qingdao 266109, P.R. China
| | - Wei Shen
- Department of Biology, College of Life Sciences, Institute of Reproductive Sciences, Qingdao Agricultural University, 700# Changcheng Road, Chengyang District, Qingdao 266109, P.R. China
| | - Cui-Lian Zhang
- Reproductive Medicine Center of People's Hospital of Zhengzhou University, Zhengzhou 450003, Henan Province, P.R. China
- Reproductive Medicine Center of Henan Provincial People's Hospital, Zhengzhou 450003, Henan Province, P.R. China
| | - Zhao-Jia Ge
- Department of Biology, College of Life Sciences, Institute of Reproductive Sciences, Qingdao Agricultural University, 700# Changcheng Road, Chengyang District, Qingdao 266109, P.R. China
| |
Collapse
|
25
|
Abstract
Background Developmental programming of the embryo is controlled by genetic information but also dictated by epigenetic information contained in spermatozoa. Lifestyle and environmental factors not only influence health in one individual but can also affect the phenotype of the following generations. This is mediated via epigenetic inheritance i.e., gametic transmission of environmentally-driven epigenetic information to the offspring. Evidence is accumulating that preconceptional exposure to certain lifestyle and environmental factors, such as diet, physical activity, and smoking, affects the phenotype of the next generation through remodeling of the epigenetic blueprint of spermatozoa. Scope of Review This review will summarize current knowledge about the different epigenetic signals in sperm that are responsive to environmental and lifestyle factors and are capable of affecting embryonic development and the phenotype of the offspring later in life. Major conclusions Like somatic cells, the epigenome of spermatozoa has proven to be dynamically reactive to a wide variety of environmental and lifestyle stressors. The functional consequence on embryogenesis and phenotype of the next generation remains largely unknown. However, strong evidence of environmentally-driven sperm-borne epigenetic factors, which are capable of altering the phenotype of the next generation, is emerging on a large scale.
Collapse
|
26
|
Tian S, Lin XH, Xiong YM, Liu ME, Yu TT, Lv M, Zhao W, Xu GF, Ding GL, Xu CM, Jin M, Feng C, Wu YT, Tan YJ, Gao Q, Zhang J, Li C, Ren J, Jin LY, Chen B, Zhu H, Zhang XY, Chen SC, Liu XM, Liu Y, Zhang JY, Wang L, Zhang P, Chen XJ, Jin L, Chen X, Meng YC, Wu DD, Lin H, Yang Q, Zhou CL, Li XZ, Wang YY, Xiang YQ, Liu ZW, Gao L, Chen LT, Pan HJ, Li R, Zhang FH, Xing LF, Zhu YM, Klausen C, Leung PCK, Li JX, Sun F, Sheng JZ, Huang HF. Prevalence of Prediabetes Risk in Offspring Born to Mothers with Hyperandrogenism. EBioMedicine 2017; 16:275-283. [PMID: 28111236 PMCID: PMC5474435 DOI: 10.1016/j.ebiom.2017.01.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 12/14/2016] [Accepted: 01/09/2017] [Indexed: 01/13/2023] Open
Abstract
Background Excessive androgen exposure during pregnancy has been suggested to induce diabetic phenotypes in offspring in animal models. The aim of this study was to investigate whether pregestational maternal hyperandrogenism in human influenced the glucose metabolism in offspring via epigenetic memory from mother's oocyte to child's somatic cells. Methods Of 1782 reproductive-aged women detected pregestational serum androgen, 1406 were pregnant between 2005 and 2010. Of 1198 women who delivered, 1116 eligible mothers (147 with hyperandrogenism and 969 normal) were recruited. 1216 children (156 children born to mothers with hyperandrogenism and 1060 born to normal mother) were followed up their glycometabolism in mean age of 5 years. Imprinting genes of oocyte from mothers and lymphocytes from children were examined. A pregestational hyperandrogenism rat model was also established. Findings Children born to women with hyperandrogenism showed increased serum fasting glucose and insulin levels, and were more prone to prediabetes (adjusted RR: 3.98 (95%CI 1.16–13.58)). Oocytes from women with hyperandrogenism showed increased insulin-like growth factor 2 (IGF2) expression. Lymphocytes from their children also showed increased IGF2 expression and decreased IGF2 methylation. Treatment of human oocytes with dihydrotestosterone upregulated IGF2 and downregulated DNMT3a levels. In rat, pregestational hyperandrogenism induced diabetic phenotypes and impaired insulin secretion in offspring. In consistent with the findings in human, hyperandrogenism also increased Igf2 expression and decreased DNMT3a in rat oocytes. Importantly, the same altered methylation signatures of Igf2 were identified in the offspring pancreatic islets. Interpretation Pregestational hyperandrogenism may predispose offspring to glucose metabolism disorder via epigenetic oocyte inheritance. Clinical trial registry no.: ChiCTR-OCC-14004537; www.chictr.org. Maternal hyperandrogenism may increase the risks of glucose metabolism disorder and prediabetes in their children. High androgen levels in women may directly increased IGF2 expression and decreased IGF2 methylation in oocytes Intergenerational inheritance of epigenetic alteration could be regarded important in determining development of diabetes.
Hyperandrogenemia can be observed in most patients with polycystic ovarian syndrome that is a common endocrine disorder in women of reproductive age, especially in subfertile women. We found that maternal hyperandrogenism may increase the risks of glucose metabolism disorder and prediabetes in their children. Also, Data from human and rat suggest that this glucose metabolism disorder may be mediated by DNA methylation modifications, and this kind of epigenetic modification may be transmitted from oocytes of mothers to somatic cells of offspring. Hence, intergenerational inheritance of epigenetic alteration should be regarded important in determining development of diabetes in the future.
Collapse
Affiliation(s)
- Shen Tian
- The Key Laboratory of Reproductive Genetics, Ministry of Education, Zhejiang University, 310006, China; Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China; Department of Reproductive Medicine, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Xian-Hua Lin
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Institute of Embryo-Fetal Original Adult Disease, Shanghai Key Laboratory for Reproductive Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yi-Meng Xiong
- The Key Laboratory of Reproductive Genetics, Ministry of Education, Zhejiang University, 310006, China; The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Miao-E Liu
- The Key Laboratory of Reproductive Genetics, Ministry of Education, Zhejiang University, 310006, China; Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Tian-Tian Yu
- The Key Laboratory of Reproductive Genetics, Ministry of Education, Zhejiang University, 310006, China; The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Min Lv
- The Key Laboratory of Reproductive Genetics, Ministry of Education, Zhejiang University, 310006, China; Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Wei Zhao
- The Key Laboratory of Reproductive Genetics, Ministry of Education, Zhejiang University, 310006, China; Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Gu-Feng Xu
- The Key Laboratory of Reproductive Genetics, Ministry of Education, Zhejiang University, 310006, China; Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Guo-Lian Ding
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Institute of Embryo-Fetal Original Adult Disease, Shanghai Key Laboratory for Reproductive Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Chen-Ming Xu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Institute of Embryo-Fetal Original Adult Disease, Shanghai Key Laboratory for Reproductive Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Min Jin
- Department of Reproductive Medicine, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Chun Feng
- Department of Reproductive Medicine, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Yan-Ting Wu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Institute of Embryo-Fetal Original Adult Disease, Shanghai Key Laboratory for Reproductive Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Ya-Jing Tan
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Institute of Embryo-Fetal Original Adult Disease, Shanghai Key Laboratory for Reproductive Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Qian Gao
- The Key Laboratory of Reproductive Genetics, Ministry of Education, Zhejiang University, 310006, China
| | - Jian Zhang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Institute of Embryo-Fetal Original Adult Disease, Shanghai Key Laboratory for Reproductive Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Cheng Li
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Institute of Embryo-Fetal Original Adult Disease, Shanghai Key Laboratory for Reproductive Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Jun Ren
- The Key Laboratory of Reproductive Genetics, Ministry of Education, Zhejiang University, 310006, China; Department of Pathology and Pathophysiology, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Lu-Yang Jin
- The Key Laboratory of Reproductive Genetics, Ministry of Education, Zhejiang University, 310006, China; Department of Pathology and Pathophysiology, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Bin Chen
- The Key Laboratory of Reproductive Genetics, Ministry of Education, Zhejiang University, 310006, China; Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Hong Zhu
- The Key Laboratory of Reproductive Genetics, Ministry of Education, Zhejiang University, 310006, China; Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Xue-Ying Zhang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Institute of Embryo-Fetal Original Adult Disease, Shanghai Key Laboratory for Reproductive Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Song-Chang Chen
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Institute of Embryo-Fetal Original Adult Disease, Shanghai Key Laboratory for Reproductive Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Xin-Mei Liu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Institute of Embryo-Fetal Original Adult Disease, Shanghai Key Laboratory for Reproductive Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Ye Liu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Institute of Embryo-Fetal Original Adult Disease, Shanghai Key Laboratory for Reproductive Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Jun-Yu Zhang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Institute of Embryo-Fetal Original Adult Disease, Shanghai Key Laboratory for Reproductive Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Li Wang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Institute of Embryo-Fetal Original Adult Disease, Shanghai Key Laboratory for Reproductive Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Ping Zhang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Institute of Embryo-Fetal Original Adult Disease, Shanghai Key Laboratory for Reproductive Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Xiao-Jun Chen
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Institute of Embryo-Fetal Original Adult Disease, Shanghai Key Laboratory for Reproductive Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Li Jin
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Institute of Embryo-Fetal Original Adult Disease, Shanghai Key Laboratory for Reproductive Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Xi Chen
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Institute of Embryo-Fetal Original Adult Disease, Shanghai Key Laboratory for Reproductive Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yi-Cong Meng
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Institute of Embryo-Fetal Original Adult Disease, Shanghai Key Laboratory for Reproductive Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Dan-Dan Wu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Institute of Embryo-Fetal Original Adult Disease, Shanghai Key Laboratory for Reproductive Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Hui Lin
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Institute of Embryo-Fetal Original Adult Disease, Shanghai Key Laboratory for Reproductive Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Qian Yang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Institute of Embryo-Fetal Original Adult Disease, Shanghai Key Laboratory for Reproductive Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Cheng-Liang Zhou
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Institute of Embryo-Fetal Original Adult Disease, Shanghai Key Laboratory for Reproductive Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Xin-Zhu Li
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Institute of Embryo-Fetal Original Adult Disease, Shanghai Key Laboratory for Reproductive Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yi-Yu Wang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Institute of Embryo-Fetal Original Adult Disease, Shanghai Key Laboratory for Reproductive Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yu-Qian Xiang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Institute of Embryo-Fetal Original Adult Disease, Shanghai Key Laboratory for Reproductive Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Zhi-Wei Liu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Institute of Embryo-Fetal Original Adult Disease, Shanghai Key Laboratory for Reproductive Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Ling Gao
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Institute of Embryo-Fetal Original Adult Disease, Shanghai Key Laboratory for Reproductive Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Lu-Ting Chen
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Institute of Embryo-Fetal Original Adult Disease, Shanghai Key Laboratory for Reproductive Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Hong-Jie Pan
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Institute of Embryo-Fetal Original Adult Disease, Shanghai Key Laboratory for Reproductive Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Rong Li
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Institute of Embryo-Fetal Original Adult Disease, Shanghai Key Laboratory for Reproductive Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Fang-Hong Zhang
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Lan-Feng Xing
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Yi-Min Zhu
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China
| | - Christian Klausen
- Department of Obstetrics and Gynecology, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| | - Peter C K Leung
- Department of Obstetrics and Gynecology, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| | - Ju-Xue Li
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Institute of Embryo-Fetal Original Adult Disease, Shanghai Key Laboratory for Reproductive Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Fei Sun
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Institute of Embryo-Fetal Original Adult Disease, Shanghai Key Laboratory for Reproductive Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Jian-Zhong Sheng
- The Key Laboratory of Reproductive Genetics, Ministry of Education, Zhejiang University, 310006, China; Department of Pathology and Pathophysiology, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - He-Feng Huang
- The Key Laboratory of Reproductive Genetics, Ministry of Education, Zhejiang University, 310006, China; The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China; Institute of Embryo-Fetal Original Adult Disease, Shanghai Key Laboratory for Reproductive Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China.
| |
Collapse
|
27
|
Desmet KLJ, Van Hoeck V, Gagné D, Fournier E, Thakur A, O'Doherty AM, Walsh CP, Sirard MA, Bols PEJ, Leroy JLMR. Exposure of bovine oocytes and embryos to elevated non-esterified fatty acid concentrations: integration of epigenetic and transcriptomic signatures in resultant blastocysts. BMC Genomics 2016; 17:1004. [PMID: 27931182 PMCID: PMC5146907 DOI: 10.1186/s12864-016-3366-y] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 12/02/2016] [Indexed: 12/30/2022] Open
Abstract
Background Metabolic stress associated with negative energy balance in high producing dairy cattle and obesity in women is a risk factor for decreased fertility. Non-esterified fatty acids (NEFA) are involved in this pathogenesis as they jeopardize oocyte and embryo development. Growing evidence indicates that maternal metabolic disorders can disturb epigenetic programming, such as DNA methylation, in the offspring. Oocyte maturation and early embryo development coincide with methylation changes and both are sensitive to adverse environments. Therefore, we investigated whether elevated NEFA concentrations affect establishment and maintenance of DNA methylation in oocytes and embryos, subsequently altering transcriptomic profiles and developmental competence of resultant blastocysts. Results Bovine oocytes and embryos were exposed to different NEFA concentrations in separate experiments. In the first experiment, oocytes were matured in vitro for 24 h in medium containing: 1) physiological (“BASAL”) concentrations of oleic (OA), palmitic (PA) and stearic (SA) acid or 2) pathophysiological (“HIGH COMBI”) concentrations of OA, PA and SA. In the second experiment, zygotes were cultivated in vitro for 6.5 days under BASAL or HIGH COMBI conditions. Developmental competence was evaluated by assessing cleavage and blastocyst rate. Overall gene expression and DNA methylation of resultant blastocysts were analyzed using microarray. DNA methylation data were re-evaluated by pyrosequencing. HIGH COMBI-exposed oocytes and embryos displayed a lower competence to develop into blastocysts compared to BASAL-exposed counterparts (19.3% compared to 23.2% and 18.2% compared to 25.3%, respectively) (P < 0.05). HIGH COMBI-exposed oocytes and embryos resulted in blastocysts with altered DNA methylation and transcriptomic fingerprints, compared to BASAL-exposed counterparts. Differences in gene expression and methylation were more pronounced after exposure during culture compared to maturation suggesting that zygotes are more susceptible to adverse environments. Main gene networks affected were related to lipid and carbohydrate metabolism, cell death, immune response and metabolic disorders. Conclusions Overall, high variation in methylation between blastocysts made it difficult to draw conclusions concerning methylation of individual genes, although a clear overview of affected pathways was obtained. This may offer clues regarding the high rate of embryonic loss and metabolic diseases during later life observed in offspring from mothers displaying lipolytic disorders.
Collapse
Affiliation(s)
- K L J Desmet
- Laboratory of Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium.
| | - V Van Hoeck
- Laboratory of Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - D Gagné
- Centre de Recherche en Biologie de la Reproduction (CRBR), Département des Sciences Animales, Université Laval, Québec, Canada
| | - E Fournier
- Centre de Recherche en Biologie de la Reproduction (CRBR), Département des Sciences Animales, Université Laval, Québec, Canada
| | - A Thakur
- British Columbia Cancer Agency, University of British Columbia, Vancouver, Canada
| | - A M O'Doherty
- School of Agriculture and Food Science, University College Dublin, Dublin, Ireland
| | - C P Walsh
- Centre for Molecular Biosciences, School of Biomedical Sciences, University of Ulster, Coleraine, UK
| | - M A Sirard
- Centre de Recherche en Biologie de la Reproduction (CRBR), Département des Sciences Animales, Université Laval, Québec, Canada
| | - P E J Bols
- Laboratory of Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - J L M R Leroy
- Laboratory of Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
28
|
Abstract
Diabetic embryopathy is a theoretical enigma and a clinical challenge. Both type 1 and type 2 diabetic pregnancy carry a significant risk for fetal maldevelopment, and the precise reasons for the diabetes-induced teratogenicity are not clearly identified. The experimental work in this field has revealed a partial, however complex, answer to the teratological question, and we will review some of the latest suggestions.
Collapse
Affiliation(s)
- Ulf J. Eriksson
- CONTACT Ulf J. Eriksson Department of Medical Cell Biology, Uppsala University, Biomedical Center, PO Box 571, SE-751 23 Uppsala, Sweden
| | | |
Collapse
|
29
|
McPherson NO, Bell VG, Zander-Fox DL, Fullston T, Wu LL, Robker RL, Lane M. When two obese parents are worse than one! Impacts on embryo and fetal development. Am J Physiol Endocrinol Metab 2015. [PMID: 26199280 DOI: 10.1152/ajpendo.00230.2015] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The prevalence of overweight and obesity in reproductive-age adults is increasing worldwide. While the effects of either paternal or maternal obesity on gamete health and subsequent fertility and pregnancy have been reported independently, the combination of having both parents overweight/obese on fecundity and offspring health has received minimal attention. Using a 2 × 2 study design in rodents we established the relative contributions of paternal and maternal obesity on fetal and embryo development and whether combined paternal and maternal obesity had an additive effect. Here, we show that parental obesity reduces fetal and placental weights without altering pregnancy establishment and is not dependent on an in utero exposure to a high-fat diet. Interestingly combined parental obesity seemed to accumulate both the negative influences of paternal and maternal obesity had alone on embryo and fetal health rather than an amplification, manifested as reduced embryo developmental competency, reduced blastocyst cell numbers, impaired mitochondrial function, and alterations to active and repressive embryonic chromatin marks, resulting in aberrant placental gene expression and reduced fetal liver mtDNA copy numbers. Further understanding both the maternal cytoplasmic and paternal genetic interactions during this early developmental time frame will be vital for understanding how developmental programming is regulated and for the proposition of interventions to mitigate their effects.
Collapse
Affiliation(s)
- N O McPherson
- School of Paediatrics and Reproductive Health, Robinson Research Institute, University of Adelaide, South Australia, Australia; Freemasons Centre for Men's Health, University of Adelaide, South Australia, Australia; Repromed, Dulwich, South Australia, Australia; and
| | - V G Bell
- School of Paediatrics and Reproductive Health, Robinson Research Institute, University of Adelaide, South Australia, Australia; Freemasons Centre for Men's Health, University of Adelaide, South Australia, Australia; Repromed, Dulwich, South Australia, Australia; and
| | - D L Zander-Fox
- School of Paediatrics and Reproductive Health, Robinson Research Institute, University of Adelaide, South Australia, Australia; Repromed, Dulwich, South Australia, Australia; and
| | - T Fullston
- School of Paediatrics and Reproductive Health, Robinson Research Institute, University of Adelaide, South Australia, Australia
| | - L L Wu
- School of Paediatrics and Reproductive Health, Robinson Research Institute, University of Adelaide, South Australia, Australia
| | - R L Robker
- School of Paediatrics and Reproductive Health, Robinson Research Institute, University of Adelaide, South Australia, Australia
| | - M Lane
- School of Paediatrics and Reproductive Health, Robinson Research Institute, University of Adelaide, South Australia, Australia; Monash In Vitro Fertilisation Group, Richmond, Australia
| |
Collapse
|
30
|
Environmental Impact on DNA Methylation in the Germline: State of the Art and Gaps of Knowledge. BIOMED RESEARCH INTERNATIONAL 2015; 2015:123484. [PMID: 26339587 PMCID: PMC4538313 DOI: 10.1155/2015/123484] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 05/03/2015] [Indexed: 12/19/2022]
Abstract
The epigenome consists of chemical changes in DNA and chromatin that without modifying the DNA sequence modulate gene expression and cellular phenotype. The epigenome is highly plastic and reacts to changing external conditions with modifications that can be inherited to daughter cells and across generations. Whereas this innate plasticity allows for adaptation to a changing environment, it also implies the potential of epigenetic derailment leading to so-called epimutations. DNA methylation is the most studied epigenetic mark. DNA methylation changes have been associated with cancer, infertility, cardiovascular, respiratory, metabolic, immunologic, and neurodegenerative pathologies. Experiments in rodents demonstrate that exposure to a variety of chemical stressors, occurring during the prenatal or the adult life, may induce DNA methylation changes in germ cells, which may be transmitted across generations with phenotypic consequences. An increasing number of human biomonitoring studies show environmentally related DNA methylation changes mainly in blood leukocytes, whereas very few data have been so far collected on possible epigenetic changes induced in the germline, even by the analysis of easily accessible sperm. In this paper, we review the state of the art on factors impinging on DNA methylation in the germline, highlight gaps of knowledge, and propose priorities for future studies.
Collapse
|
31
|
Edwards N, Farookhi R, Clarke HJ. Identification of a β-galactosidase transgene that provides a live-cell marker of transcriptional activity in growing oocytes and embryos. Mol Hum Reprod 2015; 21:583-93. [PMID: 25882542 PMCID: PMC4487448 DOI: 10.1093/molehr/gav020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 03/27/2015] [Accepted: 04/09/2015] [Indexed: 01/14/2023] Open
Abstract
Identifying the events and molecular mechanisms that regulate oocyte growth has emerged as a key objective of research in human fertility, fuelled by evidence from human and animal studies indicating that disease and environmental factors can act on oocytes to affect the health of the resulting individual and by efforts to grow oocytes in vitro to enable fertility preservation of cancer survivors. Techniques that monitor the development of growing oocytes would be valuable tools to assess the progression of growth under different conditions. Most methods used to assess oocytes grown in vitro are indirect, however, relying on characteristics of the somatic compartment of the follicle, or compromise the oocyte, preventing its subsequent culture or fertilization. We investigated the utility of T-cell factor/lymphoid enhancer-binding factor (TCF/Lef)-LacZ transgene expression as a predictor of global transcriptional activity in oocytes and early embryos. Using a fluorescent β-galactosidase substrate combined with live-cell imaging, we show that TCF/Lef-LacZ transgene expression is detectable in growing oocytes, lost in fully grown oocytes and resumes in late two-cell embryos. Transgene expression is likely regulated by a Wnt-independent mechanism. Using chromatin analysis, LacZ expression and methods to monitor and inhibit transcription, we show that TCF/Lef-LacZ expression mirrors transcriptional activity in oocytes and preimplantation embryos. Oocytes and preimplantation embryos that undergo live-cell imaging for TCF/Lef-LacZ expression are able to continue development in vitro. TCF/Lef-LacZ reporter expression in living oocytes and early embryos is thus a sensitive and faithful marker of transcriptional activity that can be used to monitor and optimize conditions for oocyte growth.
Collapse
Affiliation(s)
- Nicole Edwards
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC, Canada Department of Physiology, McGill University, Montreal, QC, Canada Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Riaz Farookhi
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC, Canada Department of Physiology, McGill University, Montreal, QC, Canada Research Institute of the McGill University Health Centre, Montreal, QC, Canada Department of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Hugh J Clarke
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC, Canada Research Institute of the McGill University Health Centre, Montreal, QC, Canada Department of Experimental Medicine, McGill University, Montreal, QC, Canada Department of Biology, McGill University, Montreal, QC, Canada
| |
Collapse
|
32
|
Clarke HJ, Vieux KF. Epigenetic inheritance through the female germ-line: The known, the unknown, and the possible. Semin Cell Dev Biol 2015; 43:106-116. [DOI: 10.1016/j.semcdb.2015.07.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 07/04/2015] [Accepted: 07/06/2015] [Indexed: 02/06/2023]
|
33
|
Ge ZJ, Schatten H, Zhang CL, Sun QY. Oocyte ageing and epigenetics. Reproduction 2015; 149:R103-R114. [PMID: 25391845 PMCID: PMC4397590 DOI: 10.1530/rep-14-0242] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Revised: 11/07/2014] [Accepted: 11/12/2014] [Indexed: 12/13/2022]
Abstract
It has become a current social trend for women to delay childbearing. However, the quality of oocytes from older females is compromised and the pregnancy rate of older women is lower. With the increased rate of delayed childbearing, it is becoming more and more crucial to understand the mechanisms underlying the compromised quality of oocytes from older women, including mitochondrial dysfunctions, aneuploidy and epigenetic changes. Establishing proper epigenetic modifications during oogenesis and early embryo development is an important aspect in reproduction. The reprogramming process may be influenced by external and internal factors that result in improper epigenetic changes in germ cells. Furthermore, germ cell epigenetic changes might be inherited by the next generations. In this review, we briefly summarise the effects of ageing on oocyte quality. We focus on discussing the relationship between ageing and epigenetic modifications, highlighting the epigenetic changes in oocytes from advanced-age females and in post-ovulatory aged oocytes as well as the possible underlying mechanisms.
Collapse
Affiliation(s)
- Zhao-Jia Ge
- Reproductive Medicine CenterHenan Provincial People's Hospital, #7 Weiwu Road, Jinshui District, Zhengzhou, Henan Province 450003, People's Republic of ChinaState Key Laboratory of Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, #1 Beichen West Road, Chaoyang District, Beijing 100101, People's Republic of ChinaReproductive Medicine CenterPeople's Hospital of Zhengzhou University, Zhengzhou, Henan Province 450003, People's Republic of ChinaDepartment of Veterinary PathobiologyUniversity of Missouri, Columbia, Missouri 65211, USA Reproductive Medicine CenterHenan Provincial People's Hospital, #7 Weiwu Road, Jinshui District, Zhengzhou, Henan Province 450003, People's Republic of ChinaState Key Laboratory of Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, #1 Beichen West Road, Chaoyang District, Beijing 100101, People's Republic of ChinaReproductive Medicine CenterPeople's Hospital of Zhengzhou University, Zhengzhou, Henan Province 450003, People's Republic of ChinaDepartment of Veterinary PathobiologyUniversity of Missouri, Columbia, Missouri 65211, USA Reproductive Medicine CenterHenan Provincial People's Hospital, #7 Weiwu Road, Jinshui District, Zhengzhou, Henan Province 450003, People's Republic of ChinaState Key Laboratory of Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, #1 Beichen West Road, Chaoyang District, Beijing 100101, People's Republic of ChinaReproductive Medicine CenterPeople's Hospital of Zhengzhou University, Zhengzhou, Henan Province 450003, People's Republic of ChinaDepartment of Veterinary PathobiologyUniversity of Missouri, Columbia, Missouri 65211, USA
| | - Heide Schatten
- Reproductive Medicine CenterHenan Provincial People's Hospital, #7 Weiwu Road, Jinshui District, Zhengzhou, Henan Province 450003, People's Republic of ChinaState Key Laboratory of Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, #1 Beichen West Road, Chaoyang District, Beijing 100101, People's Republic of ChinaReproductive Medicine CenterPeople's Hospital of Zhengzhou University, Zhengzhou, Henan Province 450003, People's Republic of ChinaDepartment of Veterinary PathobiologyUniversity of Missouri, Columbia, Missouri 65211, USA
| | - Cui-Lian Zhang
- Reproductive Medicine CenterHenan Provincial People's Hospital, #7 Weiwu Road, Jinshui District, Zhengzhou, Henan Province 450003, People's Republic of ChinaState Key Laboratory of Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, #1 Beichen West Road, Chaoyang District, Beijing 100101, People's Republic of ChinaReproductive Medicine CenterPeople's Hospital of Zhengzhou University, Zhengzhou, Henan Province 450003, People's Republic of ChinaDepartment of Veterinary PathobiologyUniversity of Missouri, Columbia, Missouri 65211, USA Reproductive Medicine CenterHenan Provincial People's Hospital, #7 Weiwu Road, Jinshui District, Zhengzhou, Henan Province 450003, People's Republic of ChinaState Key Laboratory of Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, #1 Beichen West Road, Chaoyang District, Beijing 100101, People's Republic of ChinaReproductive Medicine CenterPeople's Hospital of Zhengzhou University, Zhengzhou, Henan Province 450003, People's Republic of ChinaDepartment of Veterinary PathobiologyUniversity of Missouri, Columbia, Missouri 65211, USA
| | - Qing-Yuan Sun
- Reproductive Medicine CenterHenan Provincial People's Hospital, #7 Weiwu Road, Jinshui District, Zhengzhou, Henan Province 450003, People's Republic of ChinaState Key Laboratory of Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, #1 Beichen West Road, Chaoyang District, Beijing 100101, People's Republic of ChinaReproductive Medicine CenterPeople's Hospital of Zhengzhou University, Zhengzhou, Henan Province 450003, People's Republic of ChinaDepartment of Veterinary PathobiologyUniversity of Missouri, Columbia, Missouri 65211, USA
| |
Collapse
|
34
|
Lane M, Zander-Fox DL, Robker RL, McPherson NO. Peri-conception parental obesity, reproductive health, and transgenerational impacts. Trends Endocrinol Metab 2015; 26:84-90. [PMID: 25523615 DOI: 10.1016/j.tem.2014.11.005] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 11/16/2014] [Accepted: 11/20/2014] [Indexed: 12/20/2022]
Abstract
Maternal over-nutrition during pregnancy is a risk factor for pregnancy complications and is increasingly associated with adverse childhood outcomes such as increased propensity for obesity and metabolic disease. However, there is emerging evidence that parental lifestyle factors prior to and at conception have a powerful impact on the health of the offspring for more than one generation. Maternal and paternal obesity prior to conception alters the molecular composition of both oocytes and sperm, which can partly escape epigenetic reprogramming at fertilization, altering the developmental trajectory of the resultant embryo, ultimately increasing the incidence of obesity and metabolic disorders in offspring. Understanding the molecular underpinning of these changes may help create interventions to reduce the risk of disease in future generations.
Collapse
Affiliation(s)
- Michelle Lane
- Robinson Research Institute, School of Paediatrics and Reproductive Health, University of Adelaide, Adelaide, South Australia; Monash IVF Group, Richmond, Victoria, Australia; Freemasons Centre for Men's Health, University of Adelaide, Adelaide, South Australia.
| | - Deirdre L Zander-Fox
- Robinson Research Institute, School of Paediatrics and Reproductive Health, University of Adelaide, Adelaide, South Australia; Repromed, Dulwich, South Australia
| | - Rebecca L Robker
- Robinson Research Institute, School of Paediatrics and Reproductive Health, University of Adelaide, Adelaide, South Australia
| | - Nicole O McPherson
- Robinson Research Institute, School of Paediatrics and Reproductive Health, University of Adelaide, Adelaide, South Australia; Repromed, Dulwich, South Australia; Freemasons Centre for Men's Health, University of Adelaide, Adelaide, South Australia
| |
Collapse
|
35
|
Schatten H, Sun QY, Prather R. The impact of mitochondrial function/dysfunction on IVF and new treatment possibilities for infertility. Reprod Biol Endocrinol 2014; 12:111. [PMID: 25421171 PMCID: PMC4297407 DOI: 10.1186/1477-7827-12-111] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 09/04/2014] [Indexed: 11/12/2022] Open
Abstract
Mitochondria play vital roles in oocyte functions and they are critical indicators of oocyte quality which is important for fertilization and development into viable offspring. Quality-compromised oocytes are correlated with infertility, developmental disorders, reduced blastocyst cell number and embryo loss in which mitochondrial dysfunctions play a significant role. Increasingly, women affected by metabolic disorders such as diabetes or obesity and oocyte aging are seeking treatment in IVF clinics to overcome the effects of adverse metabolic conditions on mitochondrial functions and new treatments have become available to restore oocyte quality. The past decade has seen enormous advances in potential therapies to restore oocyte quality and includes dietary components and transfer of mitochondria from cells with mitochondrial integrity into mitochondria-impaired oocytes. New technologies have opened up new possibilities for therapeutic advances which will increase the success rates for IVF of oocytes from women with compromised oocyte quality.
Collapse
Affiliation(s)
- Heide Schatten
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO USA
| | - Qing-Yuan Sun
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100080 Beijing, China
| | - Randall Prather
- National Swine Resource and Research Center, University of Missouri, 65211 Columbia, USA
- Division of Animal Science, University of Missouri, 65211 Columbia, USA
| |
Collapse
|
36
|
Wei Y, Schatten H, Sun QY. Environmental epigenetic inheritance through gametes and implications for human reproduction. Hum Reprod Update 2014; 21:194-208. [PMID: 25416302 DOI: 10.1093/humupd/dmu061] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Traditional studies focused on DNA as the heritable information carrier that passes the phenotype from parents to offspring. However, increasing evidence suggests that information, that is independent of the DNA sequence, termed epigenetic information, can be inherited between generations. Recently, in our lab, we found that prediabetes in fathers increases the susceptibility to diabetes in offspring through gametic cytosine methylation changes. Paternal prediabetes changed overall methylation patterns in sperm, and a large portion of differentially methylated loci can be transmitted to pancreatic islets of offspring up to the second generation. In this review, we survey the extensive examples of environmentally induced epigenetic inheritance in various species, ranging from Caenorhabditis elegans to humans. We focus mainly on elucidating the molecular basis of environmental epigenetic inheritance through gametes, which is an emerging theme and has important implications for explaining the prevalence of obesity, type 2 diabetes and other chronic non-genetic diseases, which is also important for understanding the influence of environmental exposures on reproductive and overall health in offspring. METHODS For this review, we included relevant data and information obtained through a PubMed database search for all English language articles published up to August 2014 which included the term 'environmental epigenetic inheritance' and 'transgenerational epigenetic inheritance'. We focused on research papers using animal models including Drosophila, C. elegans, mouse and rat. Human data were also included. RESULTS Evidence from animal models suggests that environmental epigenetic inheritance through gametes exists in various species. Extensive molecular evidence suggests that epigenetic information carriers including DNA methylation, non-coding RNAs and chromatin proteins in gametes play important roles in the transmission of phenotypes from parents to offspring. CONCLUSIONS Given the large number of experimental evidence from various organisms, it is clear that parental environmental alterations can affect the phenotypes of offspring through gametic epigenetic alterations. This more recent thinking based on new data may have implications in explaining the prevalence of obesity, type 2 diabetes and other chronic non-genetic diseases. This also implies that, in the near future, epigenetic factors which are heritable should be regarded important in determining the risk of certain diseases. Moreover, identification of epigenetic markers in gametes (polar body or sperm) may hold great promise for predicting susceptibility to and preventing certain non-genetic diseases in offspring, as well as providing indications on parental environmental exposures.
Collapse
Affiliation(s)
- Yanchang Wei
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Heide Schatten
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA
| | - Qing-Yuan Sun
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
37
|
Jiang GJ, Ma JY, Zhang GL, Mo FF, Zhang DW, Gao SH, Li XL. Protein profiling the differences between diabetic and normal mouse cumulus cells. Mol Reprod Dev 2014; 81:1080-5. [DOI: 10.1002/mrd.22419] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 08/24/2014] [Indexed: 11/06/2022]
Affiliation(s)
- Guang-Jian Jiang
- Diabetes Research Center; Beijing University of Chinese Medicine; Beijing China
| | - Jun-Yu Ma
- Department of Animal Science and Technology; Qingdao Agricultural University; Qingdao China
| | - Guang-Li Zhang
- Diabetes Research Center; Beijing University of Chinese Medicine; Beijing China
| | - Fang-Fang Mo
- Diabetes Research Center; Beijing University of Chinese Medicine; Beijing China
| | - Dong-Wei Zhang
- Diabetes Research Center; Beijing University of Chinese Medicine; Beijing China
| | - Si-Hua Gao
- Diabetes Research Center; Beijing University of Chinese Medicine; Beijing China
| | - Xiu-Li Li
- Department of Obstetrics and Gynecology; Chinese PLA General Hospital; Hainan Branch; Sanya China
| |
Collapse
|
38
|
Ma JY, Zhao K, OuYang YC, Wang ZB, Luo YB, Hou Y, Schatten H, Shen W, Sun QY. Exogenous thymine DNA glycosylase regulates epigenetic modifications and meiotic cell cycle progression of mouse oocytes. ACTA ACUST UNITED AC 2014; 21:186-94. [DOI: 10.1093/molehr/gau094] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
39
|
Gu L, Liu H, Gu X, Boots C, Moley KH, Wang Q. Metabolic control of oocyte development: linking maternal nutrition and reproductive outcomes. Cell Mol Life Sci 2014; 72:251-71. [PMID: 25280482 DOI: 10.1007/s00018-014-1739-4] [Citation(s) in RCA: 140] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2014] [Revised: 09/12/2014] [Accepted: 09/22/2014] [Indexed: 02/01/2023]
Abstract
Obesity, diabetes, and related metabolic disorders are major health issues worldwide. As the epidemic of metabolic disorders continues, the associated medical co-morbidities, including the detrimental impact on reproduction, increase as well. Emerging evidence suggests that the effects of maternal nutrition on reproductive outcomes are likely to be mediated, at least in part, by oocyte metabolism. Well-balanced and timed energy metabolism is critical for optimal development of oocytes. To date, much of our understanding of oocyte metabolism comes from the effects of extrinsic nutrients on oocyte maturation. In contrast, intrinsic regulation of oocyte development by metabolic enzymes, intracellular mediators, and transport systems is less characterized. Specifically, decreased acid transport proteins levels, increased glucose/lipid content and elevated reactive oxygen species in oocytes have been implicated in meiotic defects, organelle dysfunction and epigenetic alteration. Therefore, metabolic disturbances in oocytes may contribute to the diminished reproductive potential experienced by women with metabolic disorders. In-depth research is needed to further explore the underlying mechanisms. This review also discusses several approaches for metabolic analysis. Metabolomic profiling of oocytes, the surrounding granulosa cells, and follicular fluid will uncover the metabolic networks regulating oocyte development, potentially leading to the identification of oocyte quality markers and prevention of reproductive disease and poor outcomes in offspring.
Collapse
Affiliation(s)
- Ling Gu
- College of Animal Science and Technology, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, Jiangsu, China,
| | | | | | | | | | | |
Collapse
|
40
|
Global genomic and transcriptomic analysis of human pancreatic islets reveals novel genes influencing glucose metabolism. Proc Natl Acad Sci U S A 2014; 111:13924-9. [PMID: 25201977 DOI: 10.1073/pnas.1402665111] [Citation(s) in RCA: 352] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Genetic variation can modulate gene expression, and thereby phenotypic variation and susceptibility to complex diseases such as type 2 diabetes (T2D). Here we harnessed the potential of DNA and RNA sequencing in human pancreatic islets from 89 deceased donors to identify genes of potential importance in the pathogenesis of T2D. We present a catalog of genetic variants regulating gene expression (eQTL) and exon use (sQTL), including many long noncoding RNAs, which are enriched in known T2D-associated loci. Of 35 eQTL genes, whose expression differed between normoglycemic and hyperglycemic individuals, siRNA of tetraspanin 33 (TSPAN33), 5'-nucleotidase, ecto (NT5E), transmembrane emp24 protein transport domain containing 6 (TMED6), and p21 protein activated kinase 7 (PAK7) in INS1 cells resulted in reduced glucose-stimulated insulin secretion. In addition, we provide a genome-wide catalog of allelic expression imbalance, which is also enriched in known T2D-associated loci. Notably, allelic imbalance in paternally expressed gene 3 (PEG3) was associated with its promoter methylation and T2D status. Finally, RNA editing events were less common in islets than previously suggested in other tissues. Taken together, this study provides new insights into the complexity of gene regulation in human pancreatic islets and better understanding of how genetic variation can influence glucose metabolism.
Collapse
|
41
|
El Hajj N, Schneider E, Lehnen H, Haaf T. Epigenetics and life-long consequences of an adverse nutritional and diabetic intrauterine environment. Reproduction 2014; 148:R111-20. [PMID: 25187623 PMCID: PMC4241689 DOI: 10.1530/rep-14-0334] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The phenomenon that adverse environmental exposures in early life are associated with increased susceptibilities for many adult, particularly metabolic diseases, is now referred to as ‘developmental origins of health and disease (DOHAD)’ or ‘Barker’ hypothesis. Fetal overnutrition and undernutrition have similar long-lasting effects on the setting of the neuroendocrine control systems, energy homeostasis, and metabolism, leading to life-long increased morbidity. There are sensitive time windows during early development, where environmental cues can program persistent epigenetic modifications which are generally assumed to mediate these gene–environment interactions. Most of our current knowledge on fetal programing comes from animal models and epidemiological studies in humans, in particular the Dutch famine birth cohort. In industrialized countries, there is more concern about adverse long-term consequences of fetal overnutrition, i.e. by exposure to gestational diabetes mellitus and/or maternal obesity which affect 10–20% of pregnancies. Epigenetic changes due to maternal diabetes/obesity may predispose the offspring to develop metabolic disease later in life and, thus, transmit the adverse environmental exposure to the next generation. This vicious cycle could contribute significantly to the worldwide metabolic disease epidemics. In this review article, we focus on the epigenetics of an adverse intrauterine environment, in particular gestational diabetes, and its implications for the prevention of complex disease.
Collapse
Affiliation(s)
- Nady El Hajj
- Institute of Human GeneticsJulius-Maximilians-Universität Würzburg, Biozentrum, Am Hubland, 97074 Würzburg, GermanyDepartment of Gynecology and ObstetricsStädtische Kliniken, 41239 Mönchengladbach, Germany
| | - Eberhard Schneider
- Institute of Human GeneticsJulius-Maximilians-Universität Würzburg, Biozentrum, Am Hubland, 97074 Würzburg, GermanyDepartment of Gynecology and ObstetricsStädtische Kliniken, 41239 Mönchengladbach, Germany
| | - Harald Lehnen
- Institute of Human GeneticsJulius-Maximilians-Universität Würzburg, Biozentrum, Am Hubland, 97074 Würzburg, GermanyDepartment of Gynecology and ObstetricsStädtische Kliniken, 41239 Mönchengladbach, Germany
| | - Thomas Haaf
- Institute of Human GeneticsJulius-Maximilians-Universität Würzburg, Biozentrum, Am Hubland, 97074 Würzburg, GermanyDepartment of Gynecology and ObstetricsStädtische Kliniken, 41239 Mönchengladbach, Germany
| |
Collapse
|
42
|
O'Doherty AM, O'Gorman A, al Naib A, Brennan L, Daly E, Duffy P, Fair T. Negative energy balance affects imprint stability in oocytes recovered from postpartum dairy cows. Genomics 2014; 104:177-85. [PMID: 25084396 DOI: 10.1016/j.ygeno.2014.07.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 07/14/2014] [Accepted: 07/15/2014] [Indexed: 12/27/2022]
Abstract
Ovarian follicle development in post-partum, high-producing dairy cows, occurs in a compromised endogenous metabolic environment (referred to as negative energy balance, NEB). Key events that occur during oocyte/follicle growth, such as the vital process of genomic imprinting, may be detrimentally affected by this altered ovarian environment. Imprinting is crucial for placental function and regulation of fetal growth, therefore failure to establish and maintain imprints during oocyte growth may contribute to early embryonic loss. Using ovum pick-up (OPU), oocytes and follicular fluid samples were recovered from cows between days 20 and 115 post-calving, encompassing the NEB period. In a complimentary study, cumulus oocyte complexes were in vitro matured under high non-esterified fatty acid (NEFA) concentrations and in the presence of the methyl-donor S-adenosylmethionine (SAM). Pyrosequencing revealed the loss of methylation at several imprinted loci in the OPU derived oocytes. The loss of DNA methylation was observed at the PLAGL1 locus in oocytes, following in vitro maturation (IVM) in the presence of elevated NEFAs and SAM. Finally, metabolomic analysis of postpartum follicular fluid samples revealed significant differences in several branched chain amino acids, with fatty acid profiles bearing similarities to those characteristic of lactating dairy cows. These results provide the first evidence that (1) the postpartum ovarian environment may affect maternal imprint acquisition and (2) elevated NEFAs during IVM can lead to the loss of imprinted gene methylation in bovine oocytes.
Collapse
Affiliation(s)
- Alan M O'Doherty
- School of Agriculture & Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Aoife O'Gorman
- School of Agriculture & Food Science, University College Dublin, Belfield, Dublin 4, Ireland; Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Abdullah al Naib
- Lyons Research Farm, University College Dublin, Belfield, Dublin 4, Ireland
| | - Lorraine Brennan
- School of Agriculture & Food Science, University College Dublin, Belfield, Dublin 4, Ireland; Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Edward Daly
- Lyons Research Farm, University College Dublin, Belfield, Dublin 4, Ireland
| | - Pat Duffy
- Lyons Research Farm, University College Dublin, Belfield, Dublin 4, Ireland
| | - Trudee Fair
- School of Agriculture & Food Science, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
43
|
Chaffin CL, Latham KE, Mtango NR, Midic U, VandeVoort CA. Dietary sugar in healthy female primates perturbs oocyte maturation and in vitro preimplantation embryo development. Endocrinology 2014; 155:2688-95. [PMID: 24731100 PMCID: PMC4060180 DOI: 10.1210/en.2014-1104] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The consumption of refined sugars continues to pose a significant health risk. However, nearly nothing is known about the effects of sugar intake by healthy women on the oocyte or embryo. Using rhesus monkeys, we show that low-dose sucrose intake over a 6-month period has an impact on the oocyte with subsequent effects on the early embryo. The ability of oocytes to resume meiosis was significantly impaired, although the differentiation of the somatic component of the ovarian follicle into progesterone-producing cells was not altered. Although the small subset of oocytes that did mature were able to be fertilized in vitro and develop into preimplantation blastocysts, there were >1100 changes in blastocyst gene expression. Because sucrose treatment ended before fertilization, the effects of sugar intake by healthy primates are concluded to be epigenetic modifications to the immature oocyte that are manifest in the preimplantation embryo.
Collapse
Affiliation(s)
- Charles L Chaffin
- Department of Obstetrics, Gynecology, and Reproductive Sciences (C.L.C.), University of Maryland School of Medicine, Baltimore, Maryland 21210; Department of Animal Science (K.E.L., U.M.), Michigan State University, East Lansing, Michigan 48824; The Fels Institute for Cancer Research and Molecular Biology and Department of Biochemistry (N.R.M.), Temple University School of Medicine, Philadelphia, Pennsylvania 19140; and California National Primate Research Center and Department of Obstetrics and Gynecology (C.A.V.), University of California, Davis, California 95616
| | | | | | | | | |
Collapse
|
44
|
Lu CL, Wang TR, Yan LY, Xia X, Zhu XH, Li R, Zhao HC, Yan J, Yin TL, Jin HY, Zhang Y, Zhang WX, Feng HL, Qiao J. Gonadotropin-mediated dynamic alterations during bovine oocyte maturation in vitro. Biol Reprod 2014; 91:44. [PMID: 24943039 DOI: 10.1095/biolreprod.114.117945] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Gonadotropins have been widely used in human-assisted reproduction and animal science for the past four decades. However, the effects of gonadotropins on oocyte maturation at the molecular and biochemical levels are poorly understood. To determine the effects of gonadotropins (recombinant follicle stimulating hormone and urinary human menopausal gonadotropin) on oocyte maturation, we used the bovine oocyte in vitro maturation model. First, we studied the effects of increasing gonadotropin concentrations on nuclear maturation and mitochondrial function in oocytes. Gonadotropins at concentrations of 0.075 and 0.75 IU/ml improved nuclear maturation and increased inner mitochondrial membrane potential and ATP levels; however, there were no beneficial effects at concentrations of 7.5 and 75 IU/ml. Second, we studied the effects of increasing gonadotropin concentrations on the status of methylation in matured (MII) oocytes. Aberrant methylation and demethylation of H19, SNRPN, and PEG3 genes were observed in MII oocytes at all concentrations except 0.075 IU/ml. The expression of genes that function in spindle formation, cell cycle control, and methylation was also downregulated by high gonadotropin concentrations. In conclusion, we established the optimal gonadotropin concentration (i.e., 0.075 IU/ml) to be used for bovine oocyte in vitro maturation studies. These results may provide a guide for clinical stimulation protocols and help to reduce the risks associated with gonadotropin administration during in vitro fertilization treatment.
Collapse
Affiliation(s)
- Cui-Ling Lu
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, China
| | - Tian-Ren Wang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, China
| | - Li-Ying Yan
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, China
| | - Xi Xia
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, China
| | - Xiao-Hui Zhu
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, China
| | - Rong Li
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, China
| | - Hong-Cui Zhao
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, China
| | - Jie Yan
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, China
| | - Tai-Lang Yin
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, China
| | - Hong-Yan Jin
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, China
| | - Yan Zhang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, China
| | - Wen-Xin Zhang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, China
| | - Huai-Liang Feng
- Department of Obstetrics and Gynecology, New York Hospital Queens-affiliated Weill Medical College of Cornell University, New York, New York
| | - Jie Qiao
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, China
| |
Collapse
|
45
|
Ge ZJ, Zhang CL, Schatten H, Sun QY. Maternal diabetes mellitus and the origin of non-communicable diseases in offspring: the role of epigenetics. Biol Reprod 2014; 90:139. [PMID: 24829025 DOI: 10.1095/biolreprod.114.118141] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2025] Open
Abstract
Offspring of diabetic mothers are susceptible to the onset of metabolic syndromes, such as type 2 diabetes and obesity at adulthood, and this trend can be inherited between generations. Genetics cannot fully explain how the noncommunicable disease in offspring of diabetic mothers is caused and inherited by the next generations. Many studies have confirmed that epigenetics may be crucial for the detrimental effects on offspring exposed to the hyperglycemic environment. Although the adverse effects on epigenetics in offspring of diabetic mothers may be the result of the poor intrauterine environment, epigenetic modifications in oocytes of diabetic mothers are also affected. Therefore, the present review is focused on the epigenetic alterations in oocytes and embryos of diabetic mothers. Furthermore, we also discuss initial mechanistic insight on maternal diabetes mellitus causing alterations of epigenetic modifications.
Collapse
Affiliation(s)
- Zhao-Jia Ge
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, P.R. China Reproductive Medicine Center, Henan Provincial People's Hospital, Zhengzhou, Henan Province, P.R. China Reproductive Medicine Center, People's Hospital of Zhengzhou University, Zhengzhou, Henan Province, P.R. China
| | - Cui-Lian Zhang
- Reproductive Medicine Center, Henan Provincial People's Hospital, Zhengzhou, Henan Province, P.R. China Reproductive Medicine Center, People's Hospital of Zhengzhou University, Zhengzhou, Henan Province, P.R. China
| | - Heide Schatten
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri
| | - Qing-Yuan Sun
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, P.R. China
| |
Collapse
|
46
|
Ge ZJ, Liang QX, Hou Y, Han ZM, Schatten H, Sun QY, Zhang CL. Maternal obesity and diabetes may cause DNA methylation alteration in the spermatozoa of offspring in mice. Reprod Biol Endocrinol 2014; 12:29. [PMID: 24721882 PMCID: PMC3984639 DOI: 10.1186/1477-7827-12-29] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Accepted: 04/03/2014] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND The adverse effects on offspring of diabetic and/or obese mothers can be passed to the next generation. However, the mechanisms behind this are still unclear. Epigenetics may play a key role during this process. METHODS To confirm the hypothesis, we investigated the DNA methylation of several imprinted genes in spermatozoa of offspring from diabetic and/or obese mothers utilizing streptozotocin (STZ)- and high-fat-diet (HFD)-induced mouse models. RESULTS We found that the DNA methylation of Peg3 was significantly increased in spermatozoa of offspring of obese mothers compared to that in spermatozoa of offspring of normal mothers. The DNA methylation of H19 was significantly higher in spermatozoa of offspring of diabetic mothers than that in spermatozoa of offspring of non-diabetic mothers. CONCLUSIONS These results indicate that pre-gestational diabetes and/or obesity can alter DNA methylation in offspring spermatozoa.
Collapse
Affiliation(s)
- Zhao-Jia Ge
- Reproductive Medicine Center, Henan Provincial People’s Hospital, Zhengzhou 450003, Henan Province, P.R. China
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, P.R. China
- Reproductive Medicine Center, People’s Hospital of Zhengzhou University, Zhengzhou 450003, Henan province, P.R. China
| | - Qiu-Xia Liang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, P.R. China
| | - Yi Hou
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, P.R. China
| | - Zhi-Ming Han
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, P.R. China
| | - Heide Schatten
- Department of Veterinary Pathobiology, University of Missouri, 65211 Columbia, MO, USA
| | - Qing-Yuan Sun
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, P.R. China
| | - Cui-Lian Zhang
- Reproductive Medicine Center, Henan Provincial People’s Hospital, Zhengzhou 450003, Henan Province, P.R. China
- Reproductive Medicine Center, People’s Hospital of Zhengzhou University, Zhengzhou 450003, Henan province, P.R. China
| |
Collapse
|
47
|
Ge ZJ, Luo SM, Lin F, Liang QX, Huang L, Wei YC, Hou Y, Han ZM, Schatten H, Sun QY. DNA methylation in oocytes and liver of female mice and their offspring: effects of high-fat-diet-induced obesity. ENVIRONMENTAL HEALTH PERSPECTIVES 2014; 122:159-164. [PMID: 24316659 PMCID: PMC3915265 DOI: 10.1289/ehp.1307047] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 12/05/2013] [Indexed: 05/25/2023]
Abstract
BACKGROUND Maternal obesity has adverse effects on oocyte quality, embryo development, and the health of the offspring. OBJECTIVES To understand the underlying mechanisms responsible for the negative effects of maternal obesity, we investigated the DNA methylation status of several imprinted genes and metabolism-related genes. METHODS Using a high-fat-diet (HFD)-induced mouse model of obesity, we analyzed the DNA methylation of several imprinted genes and metabolism-related genes in oocytes from control and obese dams and in oocytes and liver from their offspring. Analysis was performed using combined bisulfite restriction analysis (COBRA) and bisulfite sequencing. RESULTS DNA methylation of imprinted genes in oocytes was not altered in either obese dams or their offspring; however, DNA methylation of metabolism-related genes was changed. In oocytes of obese mice, the DNA methylation level of the leptin (Lep) promoter was significantly increased and that of the Ppar-α promoter was reduced. Increased methylation of Lep and decreased methylation of Ppar-α was also observed in the liver of female offspring from dams fed the high-fat diet (OHFD). mRNA expression of Lep and Ppar-α was also significantly altered in the liver of these OHFD. In OHFD oocytes, the DNA methylation level of Ppar-α promoter was increased. CONCLUSIONS Our results indicate that DNA methylation patterns of several metabolism-related genes are changed not only in oocytes of obese mice but also in oocytes and liver of their offspring. These data may contribute to the understanding of adverse effects of maternal obesity on reproduction and health of the offspring.
Collapse
Affiliation(s)
- Zhao-Jia Ge
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Ge ZJ, Liang QX, Luo SM, Wei YC, Han ZM, Schatten H, Sun QY, Zhang CL. Diabetic uterus environment may play a key role in alterations of DNA methylation of several imprinted genes at mid-gestation in mice. Reprod Biol Endocrinol 2013; 11:119. [PMID: 24378208 PMCID: PMC3896855 DOI: 10.1186/1477-7827-11-119] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 12/26/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Maternal diabetes mellitus not only has severe deleterious effects on fetal development, but also it affects transmission to the next generation. However, the underlying mechanisms for these effects are still not clear. METHODS We investigated the methylation patterns and expressions of the imprinted genes Peg3, Snrpn, and H19 in mid-gestational placental tissues and on the whole fetus utilizing the streptozotocin (STZ)-induced hyperglycemic mouse model for quantitative analysis of methylation by PCR and quantitative real-time PCR. The protein expression of Peg3 was evaluated by Western blot. RESULTS We found that the expression of H19 was significantly increased, while the expression of Peg3 was significantly decreased in dpc10.5 placentas of diabetic mice. We further found that the methylation level of Peg3 was increased and that of H19 was reduced in dpc10.5 placentas of diabetic mice. When pronuclear embryos of normal females were transferred to normal/diabetic (NN/ND) pseudopregnant females, the methylation and expression of Peg3 in placentas was also clearly altered in the ND group compared to the NN group. However, when the pronuclear embryos of diabetic female were transferred to normal pesudopregnant female mice (DN), the methylation and expression of Peg3 and H19 in dpc10.5 placentas was similar between the two groups. CONCLUSIONS We suggest that the effects of maternal diabetes on imprinted genes may primarily be caused by the adverse uterus environment.
Collapse
Affiliation(s)
- Zhao-Jia Ge
- Reproductive Medicine Center, Henan Provincial People’s Hospital, Zhengzhou 450003, Henan Province, P. R. China
- Reproductive Medicine Center, People’s Hospital of Zhengzhou University, Zhengzhou 450003, Henan Province, P. R. China
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, P. R. China
| | - Qiu-Xia Liang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, P. R. China
| | - Shi-Ming Luo
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, P. R. China
| | - Yan-Chang Wei
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, P. R. China
| | - Zhi-Ming Han
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, P. R. China
| | - Heide Schatten
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA
| | - Qing-Yuan Sun
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, P. R. China
| | - Cui-Lian Zhang
- Reproductive Medicine Center, Henan Provincial People’s Hospital, Zhengzhou 450003, Henan Province, P. R. China
| |
Collapse
|
49
|
Soubry A, Murphy SK, Wang F, Huang Z, Vidal AC, Fuemmeler BF, Kurtzberg J, Murtha A, Jirtle RL, Schildkraut JM, Hoyo C. Newborns of obese parents have altered DNA methylation patterns at imprinted genes. Int J Obes (Lond) 2013; 39:650-7. [PMID: 24158121 PMCID: PMC4048324 DOI: 10.1038/ijo.2013.193] [Citation(s) in RCA: 222] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 09/25/2013] [Accepted: 10/06/2013] [Indexed: 12/14/2022]
Abstract
Background: Several epidemiologic studies have demonstrated associations between periconceptional environmental exposures and health status of the offspring in later life. Although these environmentally related effects have been attributed to epigenetic changes, such as DNA methylation shifts at imprinted genes, little is known about the potential effects of maternal and paternal preconceptional overnutrition or obesity. Objective: We examined parental preconceptional obesity in relation to DNA methylation profiles at multiple human imprinted genes important in normal growth and development, such as: maternally expressed gene 3 (MEG3), mesoderm-specific transcript (MEST), paternally expressed gene 3 (PEG3), pleiomorphic adenoma gene-like 1 (PLAGL1), epsilon sarcoglycan and paternally expressed gene 10 (SGCE/PEG10) and neuronatin (NNAT). Methods: We measured methylation percentages at the differentially methylated regions (DMRs) by bisulfite pyrosequencing in DNA extracted from umbilical cord blood leukocytes of 92 newborns. Preconceptional obesity, defined as BMI ⩾30 kg m−2, was ascertained through standardized questionnaires. Results: After adjusting for potential confounders and cluster effects, paternal obesity was significantly associated with lower methylation levels at the MEST (β=−2.57; s.e.=0.95; P=0.008), PEG3 (β=−1.71; s.e.=0.61; P=0.005) and NNAT (β=−3.59; s.e.=1.76; P=0.04) DMRs. Changes related to maternal obesity detected at other loci were as follows: β-coefficient was +2.58 (s.e.=1.00; P=0.01) at the PLAGL1 DMR and −3.42 (s.e.=1.69; P=0.04) at the MEG3 DMR. Conclusion: We found altered methylation outcomes at multiple imprint regulatory regions in children born to obese parents, compared with children born to non-obese parents. In spite of the small sample size, our data suggest a preconceptional influence of parental life-style or overnutrition on the (re)programming of imprint marks during gametogenesis and early development. More specifically, the significant and independent association between paternal obesity and the offspring's methylation status suggests the susceptibility of the developing sperm for environmental insults. The acquired imprint instability may be carried onto the next generation and increase the risk for chronic diseases in adulthood.
Collapse
Affiliation(s)
- A Soubry
- 1] Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA [2] Epidemiology Research Group, Department of Public Health and Primary Care, Faculty of Medicine, KULeuven, Leuven, Belgium
| | - S K Murphy
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Duke University Medical Center, Durham, NC, USA
| | - F Wang
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - Z Huang
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Duke University Medical Center, Durham, NC, USA
| | - A C Vidal
- Department of Obstetrics and Gynecology, Division of Clinical and Epidemiologic Research and Cancer Prevention, Duke University Medical Center, Durham, NC, USA
| | - B F Fuemmeler
- Department of Community and Family Medicine, Duke University Medical Center, Durham, NC, USA
| | - J Kurtzberg
- Carolinas Cord Blood Bank, Robertson Cell and Translational Therapy Program, Duke Translational Research Institute, Duke University Medical Center, Durham, NC, USA
| | - A Murtha
- Department of Obstetrics and Gynecology, Division of Maternal and Fetal Medicine, Duke University Medical Center, Durham, NC, USA
| | - R L Jirtle
- Department of Oncology, McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, WI, USA
| | - J M Schildkraut
- 1] Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA [2] Department of Community and Family Medicine, Duke University Medical Center, Durham, NC, USA
| | - C Hoyo
- Department of Obstetrics and Gynecology, Division of Clinical and Epidemiologic Research and Cancer Prevention, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|