1
|
Wang J, Liu L, Li Z, Wang H, Ren Y, Wang K, Liu Y, Tao X, Zheng L. JMJD3 regulate H3K27me3 modification via interacting directly with TET1 to affect spermatogonia self-renewal and proliferation. BMC Genomics 2024; 25:225. [PMID: 38424516 PMCID: PMC10905883 DOI: 10.1186/s12864-024-10120-9] [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: 10/20/2023] [Accepted: 02/13/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND In epigenetic modification, histone modification and DNA methylation coordinate the regulation of spermatogonium. Not only can methylcytosine dioxygenase 1 (TET1) function as a DNA demethylase, converting 5-methylcytosine to 5-hydroxymethylcytosine, it can also form complexes with other proteins to regulate gene expression. H3K27me3, one of the common histone modifications, is involved in the regulation of stem cell maintenance and tumorigenesis by inhibiting gene transcription. METHODS we examined JMJD3 at both mRNA and protein levels and performed Chip-seq sequencing of H3K27me3 in TET1 overexpressing cells to search for target genes and signaling pathways of its action. RESULTS This study has found that JMJD3 plays a leading role in spermatogonia self-renewal and proliferation: at one extreme, the expression of the self-renewal gene GFRA1 and the proliferation-promoting gene PCNA was upregulated following the overexpression of JMJD3 in spermatogonia; at the other end of the spectrum, the expression of differentiation-promoting gene DAZL was down-regulated. Furthermore, the fact that TET1 and JMJD3 can form a protein complex to interact with H3K27me3 has also been fully proven. Then, through analyzing the sequencing results of CHIP-Seq, we found that TET1 targeted Pramel3 when it interacted with H3K27me3. Besides, TET1 overexpression not only reduced H3K27me3 deposition at Pramel3, but promoted its transcriptional activation as well, and the up-regulation of Pramel3 expression was verified in JMJD3-overexpressing spermatogonia. CONCLUSION In summary, our study identified a novel link between TET1 and H3K27me3 and established a Tet1-JMJD3-H3K27me3-Pramel3 axis to regulate spermatogonia self-renewal and proliferation. Judging from the evidence offered above, we can safely conclude that this study provides new ideas for further research regarding the mechanism of spermatogenesis and spermatogenesis disorders on an apparent spectrum.
Collapse
Affiliation(s)
- Jin Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Lingling Liu
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Zebin Li
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Haoyu Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yuanyuan Ren
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Kaisheng Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yang Liu
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Xinjie Tao
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Liming Zheng
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China.
| |
Collapse
|
2
|
Silva AFB, Morais ANP, Lima LF, Ferreira ACA, Silva RF, Sá NAR, Kumar S, Oliveira AC, Alves BG, Rodrigues APR, Gastal EL, Bordignon V, Figueiredo JR. Trimethylation profile of histones H3 lysine 4 and 9 in late preantral and early antral caprine follicles grown in vivo versus in vitro in the presence of anethole. Mol Reprod Dev 2023; 90:810-823. [PMID: 37671983 DOI: 10.1002/mrd.23700] [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: 11/21/2022] [Revised: 06/13/2023] [Accepted: 07/19/2023] [Indexed: 09/07/2023]
Abstract
This study assessed the histones methylation profile (H3K4me3 and H3K9me3) in late preantral (PA) and early antral (EA) caprine follicles grown in vivo and in vitro, and the anethole effect during in vitro culture of PA follicles. Uncultured in vivo-grown follicles (PA, n = 64; EA, n = 73) were used as controls to assess the methylation profile and genes' expression related to apoptosis cascade (BAX, proapoptotic; BCL2, antiapoptotic), steroidogenesis (CYP17, CYP19A1), and demethylation (KDM1AX1, KDM1AX2, KDM3A). The isolated PA follicles (n = 174) were cultured in vitro for 6 days in α-MEM+ in either absence (control) or presence of anethole. After culture, EA follicles were evaluated for methylation, mRNA abundance, and morphometry. Follicle diameter increased after culture, regardless of treatment. The methylation profile and the mRNA abundance were similar between in vivo-grown PA and EA follicles. Anethole treatment led to higher H3K4me3 fluorescence intensity in EA follicles. The mRNA abundances of BAX, CYP17, and CYP19A1 were higher, and BCL2 and KDM3A were lower in in vitro-grown EA follicles than in vivo-grown follicles. In conclusion, in vitro follicle culture affected H3K4me3 fluorescence intensity, mRNA abundance of apoptotic genes, and steroidogenic and demethylase enzymes compared with in vivo-grown follicles.
Collapse
Affiliation(s)
- Ana F B Silva
- Laboratory of Manipulation of Oocytes and Preantral Follicles, Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, Ceará, Brazil
| | - Ana N P Morais
- Laboratory of Manipulation of Oocytes and Preantral Follicles, Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, Ceará, Brazil
| | - Laritza F Lima
- Laboratory of Manipulation of Oocytes and Preantral Follicles, Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, Ceará, Brazil
| | - Anna C A Ferreira
- Laboratory of Manipulation of Oocytes and Preantral Follicles, Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, Ceará, Brazil
| | - Renato F Silva
- Laboratory of Manipulation of Oocytes and Preantral Follicles, Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, Ceará, Brazil
| | - Naiza A R Sá
- Laboratory of Manipulation of Oocytes and Preantral Follicles, Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, Ceará, Brazil
| | - Satish Kumar
- Postgraduate Program in Veterinary Sciences, Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, Ceará, Brazil
| | - Ariclécio C Oliveira
- Superior Institute of Biomedical Science, State University of Ceará, Fortaleza, Ceará, Brazil
| | - Benner G Alves
- Postgraduate Program in Animal Bioscience, Federal University of Goiás, Jataí, Goiás, Brazil
| | - Ana P R Rodrigues
- Laboratory of Manipulation of Oocytes and Preantral Follicles, Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, Ceará, Brazil
| | - Eduardo L Gastal
- Animal Science, School of Agricultural Sciences, Southern Illinois University, Carbondale, Illinois, USA
| | - Vilceu Bordignon
- Department of Animal Science, McGill University, Sainte-Anne-de-Bellevue, Quebec, Canada
| | - José R Figueiredo
- Laboratory of Manipulation of Oocytes and Preantral Follicles, Faculty of Veterinary Medicine, State University of Ceará, Fortaleza, Ceará, Brazil
| |
Collapse
|
3
|
Yang Y, Cai Y, Guo J, Dai K, Liu L, Chen Z, Wang F, Deng M. Knockdown of KDM5B Leads to DNA Damage and Cell Cycle Arrest in Granulosa Cells via MTF1. Curr Issues Mol Biol 2023; 45:3219-3237. [PMID: 37185734 PMCID: PMC10136914 DOI: 10.3390/cimb45040210] [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: 03/01/2023] [Revised: 03/26/2023] [Accepted: 03/28/2023] [Indexed: 05/17/2023] Open
Abstract
KDM5B is essential for early embryo development, which is under the control of maternal factors in oocytes. Granulosa cells (GCs) play a critical role during oocyte mature. However, the role of KDM5B in GCs remains to be elucidated. In the current study, we found that KDM5B expressed highly in the ovaries and located in goat GCs. Using an RNA sequence, we identified 1353 differentially expressed genes in the KDM5B knockdown GCs, which were mainly enriched in cell cycle, cell division, DNA replication and the cellular oxidative phosphorylation regulation pathway. Moreover, we reported a decrease in the percentage of proliferated cells but an increase in the percentage of apoptotic cells in the KDM5B knockdown GCs. In addition, in the KDM5B knockdown GCs, the percentage of GCs blocked at the S phase was increased compared to the NC group, suggesting a critical role of KDM5B in the cell cycle. Moreover, in the KDM5B knockdown GCs, the reactive oxygen species level, the mitochondrial depolarization ratio, and the expression of intracellular phosphorylated histone H2AX (γH2AX) increased, suggesting that knockdown of KDM5B leads to DNA damage, primarily in the form of DNA double-strand breaks (DSBs). Interestingly, we found a down-regulation of MTF1 in the KDM5B knockdown GCs, and the level of cell proliferation, as well as the cell cycle block in the S phase, was improved. In contrast, in the group with both KDM5B knockdown and MTF1 overexpression, the level of ROS, the expression of γH2AX and the number of DNA DSB sites decreased. Taken together, our results suggest that KDM5B inhibits DNA damage and promotes the cell cycle in GCs, which might occur through the up-regulation of MTF1.
Collapse
Affiliation(s)
- Yingnan Yang
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing 210095, China
| | - Yu Cai
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing 210095, China
| | - Jinjing Guo
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing 210095, China
| | - Keke Dai
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing 210095, China
| | - Liang Liu
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing 210095, China
| | - Zili Chen
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing 210095, China
| | - Feng Wang
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing 210095, China
| | - Mingtian Deng
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
4
|
Mo J, Liu X, Huang Y, He R, Zhang Y, Huang H. Developmental origins of adult diseases. MEDICAL REVIEW (BERLIN, GERMANY) 2022; 2:450-470. [PMID: 37724166 PMCID: PMC10388800 DOI: 10.1515/mr-2022-0027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 10/11/2022] [Indexed: 09/20/2023]
Abstract
The occurrence and mechanisms of developmental adult diseases have gradually attracted attention in recent years. Exposure of gametes and embryos to adverse environments, especially during plastic development, can alter the expression of certain tissue-specific genes, leading to increased susceptibility to certain diseases in adulthood, such as diabetes, cardiovascular disease, neuropsychiatric, and reproductive system diseases, etc. The occurrence of chronic disease in adulthood is partly due to genetic factors, and the remaining risk is partly due to environmental-dependent epigenetic information alteration, including DNA methylation, histone modifications, and noncoding RNAs. Changes in this epigenetic information potentially damage our health, which has also been supported by numerous epidemiological and animal studies in recent years. Environmental factors functionally affect embryo development through epimutation, transmitting diseases to offspring and even later generations. This review mainly elaborated on the concept of developmental origins of adult diseases, and revealed the epigenetic mechanisms underlying these events, discussed the theoretical basis for the prevention and treatment of related diseases.
Collapse
Affiliation(s)
- Jiaying Mo
- Department of Obstetrics and Gynecology, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, Zhejiang Province, China
- The Key Laboratory of Reproductive Genetics (Zhejiang University), Ministry of Education, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Xuanqi Liu
- The Key Laboratory of Reproductive Genetics (Zhejiang University), Ministry of Education, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Yutong Huang
- The Key Laboratory of Reproductive Genetics (Zhejiang University), Ministry of Education, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Renke He
- Department of Obstetrics and Gynecology, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, Zhejiang Province, China
- The Key Laboratory of Reproductive Genetics (Zhejiang University), Ministry of Education, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Yu Zhang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Hefeng Huang
- Department of Obstetrics and Gynecology, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, Zhejiang Province, China
- The Key Laboratory of Reproductive Genetics (Zhejiang University), Ministry of Education, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China
| |
Collapse
|
5
|
Whitworth KM, Green JA, Redel BK, Geisert RD, Lee K, Telugu BP, Wells KD, Prather RS. Improvements in pig agriculture through gene editing. CABI AGRICULTURE AND BIOSCIENCE 2022; 3:41. [PMID: 35755158 PMCID: PMC9209828 DOI: 10.1186/s43170-022-00111-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 06/12/2022] [Indexed: 05/06/2023]
Abstract
Genetic modification of animals via selective breeding is the basis for modern agriculture. The current breeding paradigm however has limitations, chief among them is the requirement for the beneficial trait to exist within the population. Desirable alleles in geographically isolated breeds, or breeds selected for a different conformation and commercial application, and more importantly animals from different genera or species cannot be introgressed into the population via selective breeding. Additionally, linkage disequilibrium results in low heritability and necessitates breeding over successive generations to fix a beneficial trait within a population. Given the need to sustainably improve animal production to feed an anticipated 9 billion global population by 2030 against a backdrop of infectious diseases and a looming threat from climate change, there is a pressing need for responsive, precise, and agile breeding strategies. The availability of genome editing tools that allow for the introduction of precise genetic modification at a single nucleotide resolution, while also facilitating large transgene integration in the target population, offers a solution. Concordant with the developments in genomic sequencing approaches, progress among germline editing efforts is expected to reach feverish pace. The current manuscript reviews past and current developments in germline engineering in pigs, and the many advantages they confer for advancing animal agriculture.
Collapse
Affiliation(s)
- Kristin M. Whitworth
- Division of Animal Science, College of Agriculture Food and Natural Resources, University of Missouri, 920 East Campus Drive, Columbia, MO 65211 USA
| | - Jonathan A. Green
- Division of Animal Science, College of Agriculture Food and Natural Resources, University of Missouri, 920 East Campus Drive, Columbia, MO 65211 USA
| | - Bethany K. Redel
- United States Department of Agriculture – Agriculture Research Service, Plant Genetics Research Unit, Columbia, MO 65211 USA
| | - Rodney D. Geisert
- Division of Animal Science, College of Agriculture Food and Natural Resources, University of Missouri, 920 East Campus Drive, Columbia, MO 65211 USA
| | - Kiho Lee
- Division of Animal Science, College of Agriculture Food and Natural Resources, University of Missouri, 920 East Campus Drive, Columbia, MO 65211 USA
| | - Bhanu P. Telugu
- Division of Animal Science, College of Agriculture Food and Natural Resources, University of Missouri, 920 East Campus Drive, Columbia, MO 65211 USA
| | - Kevin D. Wells
- Division of Animal Science, College of Agriculture Food and Natural Resources, University of Missouri, 920 East Campus Drive, Columbia, MO 65211 USA
| | - Randall S. Prather
- Division of Animal Science, College of Agriculture Food and Natural Resources, University of Missouri, 920 East Campus Drive, Columbia, MO 65211 USA
| |
Collapse
|
6
|
Bai D, Sun J, Chen C, Jia Y, Li Y, Liu K, Zhang Y, Yin J, Liu Y, Han X, Ruan J, Kou X, Zhao Y, Wang H, Wang Z, Chen M, Teng X, Jiang C, Gao S, Liu W. Aberrant H3K4me3 modification of epiblast genes of extraembryonic tissue causes placental defects and implantation failure in mouse IVF embryos. Cell Rep 2022; 39:110784. [PMID: 35508139 DOI: 10.1016/j.celrep.2022.110784] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 02/22/2022] [Accepted: 04/13/2022] [Indexed: 12/13/2022] Open
Abstract
Assisted reproductive technology has been widely applied in the treatment of human infertility. However, accumulating evidence indicates that in vitro fertilization (IVF) is associated with a low pregnancy rate, placental defects, and metabolic diseases in offspring. Here, we find that IVF manipulation notably disrupts extraembryonic tissue-specific gene expression, and 334 epiblast (Epi)-specific genes and 24 Epi-specific transcription factors are abnormally expressed in extraembryonic ectoderm (ExE) of IVF embryos at embryonic day 7.5. Combined histone modification analysis reveals that aberrant H3K4me3 modification at the Epi active promoters results in increased expression of these genes in ExE. Importantly, we demonstrate that knockdown of the H3K4me3-recruited regulator Kmt2e, which is highly expressed in IVF embryos, greatly improves the development of IVF embryos and reduces abnormal gene expression in ExE. Our study therefore identifies that abnormal H3K4me3 modification in extraembryonic tissue is a major cause of implantation failure and abnormal placental development of IVF embryos.
Collapse
Affiliation(s)
- Dandan Bai
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Jin Sun
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Chuan Chen
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Yanping Jia
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Yanhe Li
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Kuisheng Liu
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Yalin Zhang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Jiqing Yin
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Yingdong Liu
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Xiaoxiao Han
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Jingling Ruan
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Xiaochen Kou
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Yanhong Zhao
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Hong Wang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Zheng Wang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Miaoxin Chen
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Xiaoming Teng
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Cizhong Jiang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China.
| | - Shaorong Gao
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China.
| | - Wenqiang Liu
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China.
| |
Collapse
|
7
|
Cheng H, Han Y, Zhang J, Zhang S, Zhai Y, An X, Li Q, Duan J, Zhang X, Li Z, Tang B, Shen H. Effects of dimethyl sulfoxide (DMSO) on DNA methylation and histone modification in parthenogenetically activated porcine embryos. Reprod Fertil Dev 2022; 34:598-607. [PMID: 35397781 DOI: 10.1071/rd21083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 12/20/2021] [Indexed: 12/18/2022] Open
Abstract
Epigenetic mechanisms play an important role in oogenesis and early embryo development in mammals. Dimethyl sulfoxide (DMSO) is frequently used as a solvent in biological studies and as a vehicle for drug therapy. Recent studies suggest that DMSO detrimentally affects porcine embryonic development, yet the mechanism of the process in parthenogenetically activated porcine embryos has not been reported. In this study, we found that treatment of embryos with 1.5% DMSO significantly decreased the cleavage and blastocyst rates, total cell number of blastocysts and the anti-apoptotic gene BCL-2 transcription level; however, the percentage of apoptotic cells and the expression levels of the pro-apoptotic gene BAX were not changed. Treatment with DMSO significantly decreased the expression levels of DNMT1 , DNMT3a , DNMT3b , TET1 , TET2 , TET3 , KMT2C , MLL2 and SETD3 in most of the stages of embryonic development and increased 5-mC signals, while the staining intensity for 5-hmC had no change in porcine preimplantation embryos from 2-cell to the blastocyst stages. Meanwhile, DMSO decreased the level of H3K4me3 during the development of parthenogenetically activated porcine embryos. After treatment with DMSO, expression levels of the pluripotency-related genes POU5F1 and NANOG decreased significantly (P <0.01), whereas the imprinted gene H19 did not change (P >0.05). In conclusion, these results suggest that DMSO can affect genome-wide DNA methylation and histone modification by regulating the expression of epigenetic modification enzymes, and DMSO also influences the expression level of pluripotent genes. These dysregulations lead to defects in embryonic development.
Collapse
Affiliation(s)
- Hui Cheng
- College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, China
| | - Yu Han
- College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, China
| | - Jian Zhang
- College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, China
| | - Sheng Zhang
- Academy of Translational Medicine, First Hospital, Jilin University, Changchun, Jilin 130021, China
| | - Yanhui Zhai
- College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, China
| | - Xinglan An
- Academy of Translational Medicine, First Hospital, Jilin University, Changchun, Jilin 130021, China
| | - Qi Li
- Academy of Translational Medicine, First Hospital, Jilin University, Changchun, Jilin 130021, China
| | - Jiahui Duan
- College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, China
| | - Xueming Zhang
- College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, China
| | - Ziyi Li
- Academy of Translational Medicine, First Hospital, Jilin University, Changchun, Jilin 130021, China
| | - Bo Tang
- College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, China
| | - Haiqing Shen
- College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, China
| |
Collapse
|
8
|
Dang Y, Luo L, Shi Y, Li S, Wang S, Zhang K. KDM5-mediated redistribution of H3K4me3 is required for oocyte-to-embryonic transition in cattle. Biol Reprod 2022; 106:1059-1071. [PMID: 35243485 DOI: 10.1093/biolre/ioac047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 11/12/2022] Open
Abstract
Reprogramming of histone modifications is critical to safeguard correct gene expression profile during preimplantation development. Of interest, trimethylation of lysine 4 on histone 3 (H3K4me3) exhibits a unique and dynamic landscape with a potential species-specific feature. Here, we address how it is reprogrammed and its functional significance during oocyte maturation and early embryonic development in cows. Notably, the overall signal of H3K4me3 decreased sharply during embryonic genome activation (EGA). By using low input ChIP-seq, we find widespread broad H3K4me3 domains in oocytes and early cleaved embryos. The broad domains are gradually removed after fertilization, which is obviously seen during EGA. Meanwhile, H3K4me3 become enriched at promoter regions after the removal of broad H3K4me3. Interestingly, the gene expression level displays a positive correlation with the relative H3K4me3 signal of their promoters when embryos reach 16-cell stage. Importantly, disruption of H3K4me3 demethylases KDM5 increases H3K4me3 level, decreases the embryonic developmental rate and results in dysregulation of over a thousand genes. Meanwhile, KDM5 deficiency causes a redistribution of H3K4me3 across genome. In particular, H3K4me3 in gene body or intergenic regions can't be removed and H3K4me3 in promoter regions is aberrantly reduced. Besides, the positive correlation between promoter H3K4me3 enrichment and gene expression level disappear. Overall, we describe the genomic reprogramming of H3K4me3 with a greater resolution during bovine preimplantation development and propose that KDM5-mediated redistribution of H3K4me3 plays an important role in modulating oocyte-to-embryonic transition.
Collapse
Affiliation(s)
- Yanna Dang
- Laboratory of Mammalian Molecular Embryology, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Lei Luo
- Laboratory of Mammalian Molecular Embryology, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Yan Shi
- Laboratory of Mammalian Molecular Embryology, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Shuang Li
- Laboratory of Mammalian Molecular Embryology, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Shaohua Wang
- Laboratory of Mammalian Molecular Embryology, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Kun Zhang
- Laboratory of Mammalian Molecular Embryology, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| |
Collapse
|
9
|
Lu X, Zhang Y, Wang L, Wang L, Wang H, Xu Q, Xiang Y, Chen C, Kong F, Xia W, Lin Z, Ma S, Liu L, Wang X, Ni H, Li W, Guo Y, Xie W. Evolutionary epigenomic analyses in mammalian early embryos reveal species-specific innovations and conserved principles of imprinting. SCIENCE ADVANCES 2021; 7:eabi6178. [PMID: 34818044 PMCID: PMC8612685 DOI: 10.1126/sciadv.abi6178] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 10/06/2021] [Indexed: 05/24/2023]
Abstract
While mouse remains the most popular model, the conservation of parental-to-embryonic epigenetic transition across mammals is poorly defined. Through analysis of oocytes and early embryos in human, bovine, porcine, rat, and mouse, we revealed remarkable species-specific innovations as no single animal model fully recapitulates the human epigenetic transition. In rodent oocytes, transcription-dependent DNA methylation allows methylation of maternal imprints but not intergenic paternal imprints. Unexpectedly, prevalent DNA hypermethylation, paralleled by H3K36me2/3, also occurs in nontranscribed regions in porcine and bovine oocytes, except for megabase-long “CpG continents (CGCs)” where imprinting control regions preferentially reside. Broad H3K4me3 and H3K27me3 domains exist in nonhuman oocytes, yet only rodent H3K27me3 survives beyond genome activation. Coincidently, regulatory elements preferentially evade H3K27me3 in rodent oocytes, and failure to do so causes aberrant embryonic gene repression. Hence, the diverse mammalian innovations of parental-to-embryonic transition center on a delicate “to-methylate-or-not” balance in establishing imprints while protecting other regulatory regions.
Collapse
Affiliation(s)
- Xukun Lu
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
- Center for Stem Cell Biology and Regenerative Medicine, MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yu Zhang
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
- Center for Stem Cell Biology and Regenerative Medicine, MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Lijuan Wang
- Center for Stem Cell Biology and Regenerative Medicine, MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing 100084, China
- College of Animal Science and Technology, Beijing University of Agriculture, Beijing 102206, China
| | - Leyun Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| | - Huili Wang
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Qianhua Xu
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
- Center for Stem Cell Biology and Regenerative Medicine, MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yunlong Xiang
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
- Center for Stem Cell Biology and Regenerative Medicine, MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Chaolei Chen
- College of Animal Science and Technology, Beijing University of Agriculture, Beijing 102206, China
| | - Feng Kong
- Center for Stem Cell Biology and Regenerative Medicine, MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Weikun Xia
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
- Center for Stem Cell Biology and Regenerative Medicine, MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Zili Lin
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
- Center for Stem Cell Biology and Regenerative Medicine, MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Sinan Ma
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Ling Liu
- Center for Stem Cell Biology and Regenerative Medicine, MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xiangguo Wang
- College of Animal Science and Technology, Beijing University of Agriculture, Beijing 102206, China
| | - Hemin Ni
- College of Animal Science and Technology, Beijing University of Agriculture, Beijing 102206, China
| | - Wei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| | - Yong Guo
- College of Animal Science and Technology, Beijing University of Agriculture, Beijing 102206, China
| | - Wei Xie
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
- Center for Stem Cell Biology and Regenerative Medicine, MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
10
|
Bilmez Y, Talibova G, Ozturk S. Dynamic changes of histone methylation in mammalian oocytes and early embryos. Histochem Cell Biol 2021; 157:7-25. [PMID: 34599660 DOI: 10.1007/s00418-021-02036-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/15/2021] [Indexed: 12/18/2022]
Abstract
Histone methylation is a key epigenetic mechanism and plays a major role in regulating gene expression during oocyte maturation and early embryogenesis. This mechanism can be briefly defined as the process by which methyl groups are transferred to lysine and arginine residues of histone tails extending from nucleosomes. While methylation of the lysine residues is catalyzed by histone lysine methyltransferases (KMTs), protein arginine methyltransferases (PRMTs) add methyl groups to the arginine residues. When necessary, the added methyl groups can be reversibly removed by histone demethylases (HDMs) by a process called histone demethylation. The spatiotemporal regulation of methylation and demethylation in histones contributes to modulating the expression of genes required for proper oocyte maturation and early embryonic development. In this review, we comprehensively evaluate and discuss the functional importance of dynamic histone methylation in mammalian oocytes and early embryos, regulated by KMTs, PRMTs, and HDMs.
Collapse
Affiliation(s)
- Yesim Bilmez
- Department of Histology and Embryology, Akdeniz University School of Medicine, Campus, 07070, Antalya, Turkey
| | - Gunel Talibova
- Department of Histology and Embryology, Akdeniz University School of Medicine, Campus, 07070, Antalya, Turkey
| | - Saffet Ozturk
- Department of Histology and Embryology, Akdeniz University School of Medicine, Campus, 07070, Antalya, Turkey.
| |
Collapse
|
11
|
Comparison of Histone H3K4me3 between IVF and ICSI Technologies and between Boy and Girl Offspring. Int J Mol Sci 2021; 22:ijms22168574. [PMID: 34445278 PMCID: PMC8395251 DOI: 10.3390/ijms22168574] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/03/2021] [Accepted: 08/06/2021] [Indexed: 01/04/2023] Open
Abstract
Epigenetics play a vital role in early embryo development. Offspring conceived via assisted reproductive technologies (ARTs) have a three times higher risk of epigenetic diseases than naturally conceived children. However, investigations into ART-associated placental histone modifications or sex-stratified analyses of ART-associated histone modifications remain limited. In the current study, we carried out immunohistochemistry, chip-sequence analysis, and a series of in vitro experiments. Our results demonstrated that placentas from intra-cytoplasmic sperm injection (ICSI), but not in vitro fertilization (IVF), showed global tri-methylated-histone-H3-lysine-4 (H3K4me3) alteration compared to those from natural conception. However, for acetylated-histone-H3-lysine-9 (H3K9ac) and acetylated-histone-H3-lysine-27 (H3K27ac), no significant differences between groups could be found. Further, sex -stratified analysis found that, compared with the same-gender newborn cord blood mononuclear cell (CBMC) from natural conceptions, CBMC from ICSI-boys presented more genes with differentially enriched H3K4me3 (n = 198) than those from ICSI-girls (n = 79), IVF-girls (n = 5), and IVF-boys (n = 2). We also found that varying oxygen conditions, RNA polymerase II subunit A (Polr2A), and lysine demethylase 5A (KDM5A) regulated H3K4me3. These findings revealed a difference between IVF and ICSI and a difference between boys and girls in H3K4me3 modification, providing greater insight into ART-associated epigenetic alteration.
Collapse
|
12
|
Bi D, Yao J, Wang Y, Qin G, Zhang Y, Wang Y, Zhao J. CRISPR/Cas13d-mediated efficient KDM5B mRNA knockdown in porcine somatic cells and parthenogenetic embryos. Reproduction 2021; 162:149-160. [PMID: 34096883 PMCID: PMC8284906 DOI: 10.1530/rep-21-0053] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 06/07/2021] [Indexed: 12/26/2022]
Abstract
An efficient mRNA knockdown strategy is needed to explore gene function in cells and embryos, especially to understand the process of maternal mRNA decay during early embryo development. Cas13, a novel RNA-targeting CRISPR effector protein, could bind and cleave complementary single-strand RNA, which has been employed for mRNA knockdown in mouse and human cells and RNA-virus interference in plants. Cas13 has not yet been reported to be used in pigs. In the current study, we explored the feasibility of CRISPR/Cas13d-mediated endogenous RNA knockdown in pigs. KDM5B, a histone demethylase of H3K4me3, was downregulated at the transcriptional level by 50% with CRISPR/Cas13d in porcine fibroblast cells. Knockdown of KDM5B-induced H3K4me3 expression and decreased the abundance of H3K27me3, H3K9me3, H3K4ac, H4K8ac, and H4K12ac. These changes affected cell proliferation and cell cycle. Furthermore, stable integration of the CRISPR/Cas13d system into the porcine genome resulted in the continuous expression of Cas13d and persistent knockdown of KDM5B. Finally, the RNA-targeting potential of Cas13d was further validated in porcine parthenogenetic embryos. By microinjection of Cas13d mRNA and gRNA targeting KDM5B into porcine oocytes, the expression of KDM5B was downregulated, the abundance of H3K4me3 increased as expected, and the expression of embryonic development-related genes was changed accordingly. These results indicate that CRISPR/Cas13d provides an easily programmable platform for spatiotemporal transcriptional manipulation in pigs.
Collapse
Affiliation(s)
- Dengfeng Bi
- School of Life Sciences, University of Science and Technology of China, Hefei, China.,State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Jing Yao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yu Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Guosong Qin
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Yunting Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yanfang Wang
- Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jianguo Zhao
- School of Life Sciences, University of Science and Technology of China, Hefei, China.,State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
13
|
Dynamic reprogramming and function of RNA N 6-methyladenosine modification during porcine early embryonic development. ZYGOTE 2021; 29:417-426. [PMID: 33890562 DOI: 10.1017/s0967199420000799] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
N6-Methyladenosine (m6A) regulates oocyte-to-embryo transition and the reprogramming of somatic cells into induced pluripotent stem cells. However, the role of m6A methylation in porcine early embryonic development and its reprogramming characteristics in somatic cell nuclear transfer (SCNT) embryos are yet to be known. Here, we showed that m6A methylation was essential for normal early embryonic development and its aberrant reprogramming in SCNT embryos. We identified a persistent occurrence of m6A methylation in embryos between 1-cell to blastocyst stages and m6A levels abruptly increased during the morula-to-blastocyst transition. Cycloleucine (methylation inhibitor, 20 mM) treatment efficiently reduced m6A levels, significantly decreased the rates of 4-cell embryos and blastocysts, and disrupted normal lineage allocation. Moreover, cycloleucine treatment also led to higher levels in both apoptosis and autophagy in blastocysts. Furthermore, m6A levels in SCNT embryos at the 4-cell and 8-cell stages were significantly lower than that in parthenogenetic activation (PA) embryos, suggesting an abnormal reprogramming of m6A methylation in SCNT embryos. Correspondingly, expression levels of m6A writers (METTL3 and METTL14) and eraser (FTO) were apparently higher in SCNT 8-cell embryos compared with their PA counterparts. Taken together, these results indicated that aberrant nuclear transfer-mediated reprogramming of m6A methylation was involved in regulating porcine early embryonic development.
Collapse
|
14
|
Zhu L, Marjani SL, Jiang Z. The Epigenetics of Gametes and Early Embryos and Potential Long-Range Consequences in Livestock Species-Filling in the Picture With Epigenomic Analyses. Front Genet 2021; 12:557934. [PMID: 33747031 PMCID: PMC7966815 DOI: 10.3389/fgene.2021.557934] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 02/04/2021] [Indexed: 12/31/2022] Open
Abstract
The epigenome is dynamic and forged by epigenetic mechanisms, such as DNA methylation, histone modifications, chromatin remodeling, and non-coding RNA species. Increasing lines of evidence support the concept that certain acquired traits are derived from environmental exposure during early embryonic and fetal development, i.e., fetal programming, and can even be "memorized" in the germline as epigenetic information and transmitted to future generations. Advances in technology are now driving the global profiling and precise editing of germline and embryonic epigenomes, thereby improving our understanding of epigenetic regulation and inheritance. These achievements open new avenues for the development of technologies or potential management interventions to counteract adverse conditions or improve performance in livestock species. In this article, we review the epigenetic analyses (DNA methylation, histone modification, chromatin remodeling, and non-coding RNAs) of germ cells and embryos in mammalian livestock species (cattle, sheep, goats, and pigs) and the epigenetic determinants of gamete and embryo viability. We also discuss the effects of parental environmental exposures on the epigenetics of gametes and the early embryo, and evidence for transgenerational inheritance in livestock.
Collapse
Affiliation(s)
- Linkai Zhu
- AgCenter, School of Animal Sciences, Louisiana State University, Baton Rouge, LA, United States
| | - Sadie L. Marjani
- Department of Biology, Central Connecticut State University, New Britain, CT, United States
| | - Zongliang Jiang
- AgCenter, School of Animal Sciences, Louisiana State University, Baton Rouge, LA, United States
| |
Collapse
|
15
|
Deng M, Wan Y, Chen B, Dai X, Liu Z, Yang Y, Cai Y, Zhang Y, Wang F. Long non-coding RNA lnc_3712 impedes nuclear reprogramming via repressing Kdm5b. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 24:54-66. [PMID: 33738138 PMCID: PMC7940708 DOI: 10.1016/j.omtn.2021.02.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 02/15/2021] [Indexed: 12/20/2022]
Abstract
Long non-coding RNAs (lncRNAs) are involved in shaping chromosome conformation and regulation of preimplantation development. However, the role of lncRNA during somatic cell nuclear transfer (SCNT) reprogramming remains largely unknown. In the present study, we identified 114 upregulated lncRNAs in the 8-cell SCNT embryos as candidate key molecules involved in nuclear reprogramming in goat. We found that H3K4me3 was an epigenetic barrier in goat nuclear reprogramming that and injection of Kdm5b mRNA greatly improved SCNT embryos development through removal of H3K4me3. We further reported that knockdown of lnc_3712 increased the expression of Kdm5b, which led to H3K4me3 demethylation. Of note, the development of goat SCNT embryos was improved when lnc_3712 was knocked down, whereas the blastocyst rate showed no difference in lnc_3712 and Kdm5b double knockdown SCNT embryos compared with the negative control SCNT embryos. Specifically, in lnc_3712 knockdown SCNT embryos, partial of the transcriptional activity and the expression of critical embryonic genes (Wee1, Ctsb, and Ybx1) were similar with that of in vitro fertilization embryos. Therefore, our results elucidate the critical role of lnc_3712 in regulating the development of goat SCNT embryos via repressing Kdm5b, which advances our current understanding of the role of lncRNAs during nuclear reprogramming.
Collapse
Affiliation(s)
- Mingtian Deng
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yongjie Wan
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Baobao Chen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiangpeng Dai
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, Jilin 130021, China
| | - Zifei Liu
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yingnan Yang
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yu Cai
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yanli Zhang
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Feng Wang
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
16
|
Fu YD, Huang MJ, Guo JW, You YZ, Liu HM, Huang LH, Yu B. Targeting histone demethylase KDM5B for cancer treatment. Eur J Med Chem 2020; 208:112760. [PMID: 32883639 DOI: 10.1016/j.ejmech.2020.112760] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/12/2020] [Accepted: 08/13/2020] [Indexed: 02/07/2023]
Abstract
KDM5B (Lysine-Specific Demethylase 5B) erases the methyl group from H3K4me2/3, which performs wide regulatory effects on chromatin structure, and represses the transcriptional function of genes. KDM5B functions as an oncogene and associates with human cancers closely. Targeting KDM5B has been a promising direction for curing cancer since the emergence of potent KDM5B inhibitor CPI-455. In this area, most reported KDM5B inhibitors are Fe (Ⅱ) chelators, which also compete with the cofactor 2-OG in the active pockets. Besides, Some KDM5B inhibitors have been identified through high throughput screening or biochemical screening. In this reviewing article, we summarized the pioneering progress in KDM5B to provide a comprehensive realization, including crystal structure, transcriptional regulation function, cancer-related functions, development of inhibitors, and SAR studies. We hope to provide a comprehensive overview of KDM5B and the development of KDM5B inhibitors.
Collapse
Affiliation(s)
- Yun-Dong Fu
- Green Catalysis Center, And College of Chemistry, Zhengzhou University, Zhengzhou, 450001, China
| | - Ming-Jie Huang
- Green Catalysis Center, And College of Chemistry, Zhengzhou University, Zhengzhou, 450001, China
| | - Jia-Wen Guo
- Green Catalysis Center, And College of Chemistry, Zhengzhou University, Zhengzhou, 450001, China
| | - Ya-Zhen You
- Green Catalysis Center, And College of Chemistry, Zhengzhou University, Zhengzhou, 450001, China
| | - Hong-Min Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Li-Hua Huang
- Green Catalysis Center, And College of Chemistry, Zhengzhou University, Zhengzhou, 450001, China.
| | - Bin Yu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
17
|
Li F, Wang D, Song R, Cao C, Zhang Z, Wang Y, Li X, Huang J, Liu Q, Hou N, Xu B, Li X, Gao X, Jia Y, Zhao J, Wang Y. The asynchronous establishment of chromatin 3D architecture between in vitro fertilized and uniparental preimplantation pig embryos. Genome Biol 2020; 21:203. [PMID: 32778155 PMCID: PMC7418210 DOI: 10.1186/s13059-020-02095-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 07/07/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Pigs are important animals for agricultural and biomedical research, and improvement is needed for use of the assisted reproductive technologies. Determining underlying mechanisms of epigenetic reprogramming in the early stage of preimplantation embryos derived from in vitro fertilization (IVF), parthenogenesis, and androgenesis will not only contribute to assisted reproductive technologies of pigs but also will shed light into early human development. However, the reprogramming of three-dimensional architecture of chromatin in this process in pigs is poorly understood. RESULTS We generate three-dimensional chromatin profiles for pig somatic cells, IVF, parthenogenesis, and androgenesis preimplantation embryos. We find that the chromosomes in the pig preimplantation embryos are enriched for superdomains, which are more rare in mice. However, p(s) curves, compartments, and topologically associated domains (TADs) are largely conserved in somatic cells and are gradually established during preimplantation embryogenesis in both mammals. In the uniparental pig embryos, the establishment of chromatin architecture is highly asynchronized at all levels from IVF embryos, and a remarkably strong decompartmentalization is observed during zygotic genome activation (ZGA). Finally, chromosomes originating from oocytes always establish TADs faster than chromosomes originating from sperm, both before and during ZGA. CONCLUSIONS Our data highlight a potential unique 3D chromatin pattern of enriched superdomains in pig preimplantation embryos, an unusual decompartmentalization process during ZGA in the uniparental embryos, and an asynchronized TAD reprogramming between maternal and paternal genomes, implying a severe dysregulation of ZGA in the uniparental embryos in pigs.
Collapse
Affiliation(s)
- Feifei Li
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, and China National Center for Bioinformation, Beijing, 100101 China
| | - Danyang Wang
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, and China National Center for Bioinformation, Beijing, 100101 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Ruigao Song
- University of Chinese Academy of Sciences, Beijing, 100049 China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Chunwei Cao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China
| | - Zhihua Zhang
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, and China National Center for Bioinformation, Beijing, 100101 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Yu Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193 China
| | - Xiaoli Li
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, and China National Center for Bioinformation, Beijing, 100101 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Jiaojiao Huang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Qiang Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Naipeng Hou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193 China
| | - Bingxiang Xu
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, and China National Center for Bioinformation, Beijing, 100101 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Xiao Li
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, and China National Center for Bioinformation, Beijing, 100101 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Xiaomeng Gao
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, and China National Center for Bioinformation, Beijing, 100101 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Yan Jia
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, and China National Center for Bioinformation, Beijing, 100101 China
| | - Jianguo Zhao
- University of Chinese Academy of Sciences, Beijing, 100049 China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yanfang Wang
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193 China
| |
Collapse
|
18
|
White MD, Plachta N. Specification of the First Mammalian Cell Lineages In Vivo and In Vitro. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a035634. [PMID: 31615786 DOI: 10.1101/cshperspect.a035634] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Our understanding of how the first mammalian cell lineages arise has been shaped largely by studies of the preimplantation mouse embryo. Painstaking work over many decades has begun to reveal how a single totipotent cell is transformed into a multilayered structure representing the foundations of the body plan. Here, we review how the first lineage decision is initiated by epigenetic regulation but consolidated by the integration of morphological features and transcription factor activity. The establishment of pluripotent and multipotent stem cell lines has enabled deeper analysis of molecular and epigenetic regulation of cell fate decisions. The capability to assemble these stem cells into artificial embryos is an exciting new avenue of research that offers a long-awaited window into cell fate specification in the human embryo. Together, these approaches are poised to profoundly increase our understanding of how the first lineage decisions are made during mammalian embryonic development.
Collapse
Affiliation(s)
- Melanie D White
- Institute of Molecular and Cell Biology, A*STAR, Singapore 138673
| | - Nicolas Plachta
- Institute of Molecular and Cell Biology, A*STAR, Singapore 138673
| |
Collapse
|
19
|
Glanzner WG, Gutierrez K, Rissi VB, de Macedo MP, Lopez R, Currin L, Dicks N, Baldassarre H, Agellon LB, Bordignon V. Histone Lysine Demethylases KDM5B and KDM5C Modulate Genome Activation and Stability in Porcine Embryos. Front Cell Dev Biol 2020; 8:151. [PMID: 32211412 PMCID: PMC7076052 DOI: 10.3389/fcell.2020.00151] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 02/24/2020] [Indexed: 12/16/2022] Open
Abstract
The lysine demethylases KDM5B and KDM5C are highly, but transiently, expressed in porcine embryos around the genome activation stage. Attenuation of KDM5B and KDM5C mRNA hampered embryo development to the blastocyst stage in fertilized, parthenogenetically activated and nuclear transfer embryos. While KDM5B attenuation increased H3K4me2-3 levels on D3 embryos and H3K4me1-2-3 on D5 embryos, KDM5C attenuation increased H3K9me1 on D3 embryos, and H3K9me1 and H3K4me1 on D5 embryos. The relative mRNA abundance of EIF1AX and EIF2A on D3 embryos, and the proportion of D4 embryos presenting a fluorescent signal for uridine incorporation were severely reduced in both KDM5B- and KDM5C-attenuated compared to control embryos, which indicate a delay in the initiation of the embryo transcriptional activity. Moreover, KDM5B and KDM5C attenuation affected DNA damage response and increased DNA double-strand breaks (DSBs), and decreased development of UV-irradiated embryos. Findings from this study revealed that both KDM5B and KDM5C are important regulators of early development in porcine embryos as their attenuation altered H3K4 and H3K9 methylation patterns, perturbed embryo genome activation, and decreased DNA damage repair capacity.
Collapse
Affiliation(s)
- Werner Giehl Glanzner
- Department of Animal Science, McGill University, Sainte-Anne-de-Bellevue, QC, Canada
| | - Karina Gutierrez
- Department of Animal Science, McGill University, Sainte-Anne-de-Bellevue, QC, Canada
| | - Vitor Braga Rissi
- Laboratory of Biotechnology and Animal Reproduction - BioRep, Federal University of Santa Maria (UFSM), Santa Maria, Brazil
| | | | - Rosalba Lopez
- Department of Animal Science, McGill University, Sainte-Anne-de-Bellevue, QC, Canada
| | - Luke Currin
- Department of Animal Science, McGill University, Sainte-Anne-de-Bellevue, QC, Canada
| | - Naomi Dicks
- Department of Animal Science, McGill University, Sainte-Anne-de-Bellevue, QC, Canada
| | - Hernan Baldassarre
- Department of Animal Science, McGill University, Sainte-Anne-de-Bellevue, QC, Canada
| | - Luis B Agellon
- School of Human Nutrition, McGill University, Sainte-Anne-de-Bellevue, QC, Canada
| | - Vilceu Bordignon
- Department of Animal Science, McGill University, Sainte-Anne-de-Bellevue, QC, Canada
| |
Collapse
|
20
|
Deng M, Liu Z, Chen B, Wan Y, Yang H, Zhang Y, Cai Y, Zhou J, Wang F. Aberrant DNA and histone methylation during zygotic genome activation in goat cloned embryos. Theriogenology 2020; 148:27-36. [PMID: 32126393 DOI: 10.1016/j.theriogenology.2020.02.036] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 02/17/2020] [Accepted: 02/22/2020] [Indexed: 01/23/2023]
Abstract
In somatic cell nuclear transfer (SCNT) embryos, developmental defects first appear at the time of zygotic genome activation (ZGA), a process that is under the control of DNA and histone methylation. However, dynamics of 5-mC and 5-hmC during ZGA differ between porcine and bovine SCNT embryos, and histone methylation during ZGA in goat SCNT embryos remains poorly understood. Therefore, in the present study, we investigated the dynamic changes of 5-mC, 5-hmC, H3K4me2/3, and H3K9me3, as well as the expression of key genes related to these epigenetic modifications, during ZGA in goat cloned embryos. Compared with the IVF embryos, the 5-mC signal intensity was significantly increased at the 2- and 4-cell stage SCNT embryos, and the H3K4me3 and H3K9me3 signal intensity was significantly increased at 2- to 8-cell stage SCNT embryos, while the 5-hmC and H3K4me2 signal intensity was significantly lower at the 4- and 8-cell stage SCNT embryos. Of note, the H3K9me3 level was also significantly higher, whereas H3K4me3 signal intensity showed no statistical difference in the pronuclear stage SCNT embryos. Moreover, the expression of TET2, DNMT3B, KDM4A, SUV39H1, G9A, and SETDB1 was significantly increased, while the expression of UHRF1, PCNA, KDM4B, KDM4D, KDM5A, KDM5B, and KDM5C was significantly decreased at the 8-cell stage SCNT embryos. Our data revealed aberrant DNA and histone methylation during ZGA in goat cloned embryos. We further inferred that the abnormally higher level of 5-mC, H3K4me3, and H3K9me3 might serve as epigenetic barriers of the reprogramming and modifying these aberrant modifications might be a promising strategy to improve cloning efficiency in goat.
Collapse
Affiliation(s)
- Mingtian Deng
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China; College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Zifei Liu
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China; College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Baobao Chen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yongjie Wan
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China; College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Hua Yang
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China; College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yanli Zhang
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China; College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yu Cai
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China; College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Jianguo Zhou
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China; National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing, 210095, China
| | - Feng Wang
- Jiangsu Livestock Embryo Engineering Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China; College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
21
|
Zhang L, Yu M, Xu H, Wei X, Liu Y, Huang C, Chen H, Guo Z. RNA sequencing revealed the abnormal transcriptional profile in cloned bovine embryos. Int J Biol Macromol 2020; 150:492-500. [PMID: 32035150 DOI: 10.1016/j.ijbiomac.2020.02.026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 02/04/2020] [Accepted: 02/04/2020] [Indexed: 12/11/2022]
Abstract
Somatic cell nuclear transfer (SCNT) has potential applications in agriculture and biomedicine, but the efficiency of cloning is still low. In this study, the transcriptional profiles in cloned and fertilized embryos were measured and compared by RNA sequencing. The 2-cell embryos were detected to identify the earliest transcriptional differences between embryos derived through IVF and SCNT. As a result, 364 genes showed decreased expression in cloned 2-cell embryos and were enriched in "intracellular protein transport" and "ubiquitin mediated proteolysis". In blastocysts, 593 genes showed decreased expression in cloned blastocysts and were enriched in "RNA binding", "nucleotide binding", "embryo development", and "adherens junction". We identified 14 development related genes that were not activated in the cloned embryos. Then, 68 and 245 long non-coding RNAs were recognized abnormally expressed in cloned 2-cell embryos and cloned blastocysts, respectively. Furthermore, we found that incomplete RNA-editing occurred in cloned embryos and might be caused by decreased ADAR expression. In conclusion, our study revealed the abnormal transcripts and deficient RNA-editing sites in cloned embryos and provided new data for further mechanistic studies of somatic nuclear reprogramming.
Collapse
Affiliation(s)
- Lei Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Yangling, Shaanxi Province 712100, China.
| | - Mengying Yu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Yangling, Shaanxi Province 712100, China.
| | - Hongyu Xu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Yangling, Shaanxi Province 712100, China.
| | - Xing Wei
- College of Veterinary Medicine, Northwest A&F University, Yangling, China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Yangling, Shaanxi Province 712100, China.
| | - Yingxiang Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Yangling, Shaanxi Province 712100, China.
| | - Chenyang Huang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Yangling, Shaanxi Province 712100, China.
| | - Huanhuan Chen
- College of Veterinary Medicine, Northwest A&F University, Yangling, China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Yangling, Shaanxi Province 712100, China.
| | - Zekun Guo
- College of Veterinary Medicine, Northwest A&F University, Yangling, China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Yangling, Shaanxi Province 712100, China.
| |
Collapse
|
22
|
Weng XG, Cai MM, Zhang YT, Liu Y, Liu C, Liu ZH. Improvement in the in vitro development of cloned pig embryos after kdm4a overexpression and an H3K9me3 methyltransferase inhibitor treatment. Theriogenology 2019; 146:162-170. [PMID: 31791612 DOI: 10.1016/j.theriogenology.2019.11.027] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 11/06/2019] [Accepted: 11/24/2019] [Indexed: 01/26/2023]
Abstract
Aberrant epigenetic reprogramming is a major cause of the developmental failure of embryos after somatic cell nuclear transfer (SCNT). Histone H3 lysine 9 trimethylation (H3K9me3), a histone marker of transcriptional repression, is considered a key barrier to the development of cloned embryos. In the present study, H3K9me3 levels were much higher in SCNT embryos than IVF embryos at the 4-cell and 2-cell stages. The microinjection of the kdm4a mRNA encoding an H3K9me3 demethylase significantly increased the developmental efficiency of cloned porcine embryos. Moreover, we evaluated the effect of chaetocin, an inhibitor of histone methyltransferases suv39h1/2, on SCNT embryo development. Chaetocin did not suppress the H3K9me3 modification in porcine embryonic fibroblast (PEF) but downregulated the expression of suv39h1, suv39h2, and kdm4d. However, 10 nM chaetocin treatment efficiently decreased the H3K9me3 level in cloned embryos. Importantly, a chaetocin treatment at the 4-cell stage for 6 h significantly increased the blastocyst rate and total cell numbers. Furthermore, the inhibitor treatment upregulated the expression of related developmental genes. In summary, both overexpression of kdm4a and treatment with a suv39h1/2 inhibitor improve the epigenetic reprogramming of cloned embryos and further improve the developmental competence in vitro.
Collapse
Affiliation(s)
- Xiao-Gang Weng
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, 150030, Heilongjiang, PR China
| | - Ming-Ming Cai
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, 150030, Heilongjiang, PR China
| | - Yu-Ting Zhang
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, 150030, Heilongjiang, PR China
| | - Yan Liu
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, 150030, Heilongjiang, PR China
| | - Cong Liu
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, 150030, Heilongjiang, PR China
| | - Zhong-Hua Liu
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, 150030, Heilongjiang, PR China.
| |
Collapse
|
23
|
Detection of coding sequence, mRNA expression and three insertions/deletions (indels) of KDM6A gene in male pig. Theriogenology 2019; 133:10-21. [DOI: 10.1016/j.theriogenology.2019.04.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 03/31/2019] [Accepted: 04/18/2019] [Indexed: 01/20/2023]
|
24
|
Rissi VB, Glanzner WG, De Macedo MP, Gutierrez K, Baldassarre H, Gonçalves PBD, Bordignon V. The histone lysine demethylase KDM7A is required for normal development and first cell lineage specification in porcine embryos. Epigenetics 2019; 14:1088-1101. [PMID: 31216927 DOI: 10.1080/15592294.2019.1633864] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
There is growing evidence that histone lysine demethylases (KDMs) play critical roles in the regulation of embryo development. This study investigated if KDM7A, a lysine demethylase known to act on mono-(me1) and di-(me2) methylation of H3K9 and H3K27, participates in the regulation of early embryo development. Knockdown of KDM7A mRNA reduced blastocyst formation by 69.2% in in vitro fertilized (IVF), 48.4% in parthenogenetically activated (PA), and 48.1% in somatic cell nuclear transfer (SCNT) embryos compared to controls. Global immunofluorescence (IF) signal in KDM7A knockdown compared to control embryos was increased for H3K27me1 on D7, for H3K27me2 on D3 and D5, for H3K9me1 on D5 and D7, and for H3K9me2 on D5 embryos, but decreased for H3K9me1, me2 and me3 on D3. Moreover, KDM7A knockdown altered mRNA expression, including the downregulation of KDM3C on D3, NANOG on D5 and D7, and OCT4 on D7 embryos, and the upregulation of CDX2, KDM4B and KDM6B on D5 embryos. On D3 and D5 embryos, total cell number and mRNA expression of embryo genome activation (EGA) markers (EIF1AX and PPP1R15B) were not affected by KDM7A knockdown. However, the ratio of inner cell mass (ICM)/total number of cells in D7 blastocysts was reduced by 45.5% in KDM7A knockdown compared to control embryos. These findings support a critical role for KDM7A in the regulation of early development and cell lineage specification in porcine embryos, which is likely mediated through the modulation of H3K9me1/me2 and H3K27me1/me2 levels, and changes in the expression of other KDMs and pluripotency genes.
Collapse
Affiliation(s)
- Vitor Braga Rissi
- Laboratory of Biotechnology and Animal Reproduction - BioRep, Federal University of Santa Maria (UFSM) , Santa Maria , RS , Brazil
| | - Werner Giehl Glanzner
- Department of Animal Science, McGill University , Sainte Anne de Bellevue , QC , Canada
| | | | - Karina Gutierrez
- Department of Animal Science, McGill University , Sainte Anne de Bellevue , QC , Canada
| | - Hernan Baldassarre
- Department of Animal Science, McGill University , Sainte Anne de Bellevue , QC , Canada
| | - Paulo Bayard Dias Gonçalves
- Laboratory of Biotechnology and Animal Reproduction - BioRep, Federal University of Santa Maria (UFSM) , Santa Maria , RS , Brazil
| | - Vilceu Bordignon
- Department of Animal Science, McGill University , Sainte Anne de Bellevue , QC , Canada
| |
Collapse
|
25
|
Cai Q, Niu H, Zhang B, Shi X, Liao M, Chen Z, Mo D, He Z, Chen Y, Cong P. Effect of EZH2 knockdown on preimplantation development of porcine parthenogenetic embryos. Theriogenology 2019; 132:95-105. [PMID: 31004879 DOI: 10.1016/j.theriogenology.2019.04.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 03/11/2019] [Accepted: 04/06/2019] [Indexed: 10/27/2022]
Abstract
The EZH2 protein endows the polycomb repressive complex 2 (PRC2) with histone lysine methyltransferase activity that is associated with transcriptional repression. Recent investigations have documented crucial roles for EZH2 in mediating X-inactivation, stem cell pluripotency and cancer metastasis. However, there is little evidence demonstrating the maternal effect of EZH2 on porcine preimplantation development. Here, we took parthenogenetic activation embryos to eliminate the confounding paternal influence. We showed that the dynamic expression of EZH2 during early development was accompanied by changes in H3K27me3 levels. Depletion of EZH2 in MII oocytes by small interfering RNA not only impaired embryonic development at the blastocyst stage (P < 0.05), but also disrupted the equilibrium of H3K4me3 and H3K27me3 in the embryo. Interestingly, the expression of TET1, a member of Ten-Eleven Translocation gene family for converting 5-methylcytosine (5 mC) to 5-hydroxymethylcytosine (5hmC), was decreased after EZH2 knockdown, in contrast to the increase of the other two members, TET2 and TET3 (P < 0.05). These results indicate a correlation between histone methylation and DNA methylation, and between EZH2 and TET1. Along with the downregulation of TET1, the expression of the pluripotency gene NANOG was decreased (P < 0.05), which is consistent with a previous finding in mouse ES cells. Meanwhile, the abundance of OCT4 and SOX2 were also down-regulated. Moreover, EZH2 knockdown reduced the capacity of cells in the blastocysts to resist apoptosis. Taken together, our data suggest that EZH2 is integral to the developmental program of porcine parthenogenetic embryos and exerts its function by regulating pluripotency, differentiation and apoptosis.
Collapse
Affiliation(s)
- Qingqing Cai
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China
| | - Huiran Niu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China
| | - Bingyue Zhang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China
| | - Xuan Shi
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China
| | - Mengqin Liao
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China
| | - Zihao Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China
| | - Delin Mo
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China
| | - Zuyong He
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China
| | - Yaosheng Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China
| | - Peiqing Cong
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China.
| |
Collapse
|
26
|
Abstract
Epigenetic mechanisms allow the establishment and maintenance of multiple cellular phenotypes from a single genomic code. At the initiation of development, the oocyte and spermatozoa provide their fully differentiated chromatin that soon after fertilization undergo extensive remodeling, resulting in a totipotent state that can then drive cellular differentiation towards all cell types. These remodeling involves different epigenetic modifications, including DNA methylation, post-translational modifications of histones, non-coding RNAs, and large-scale chromatin conformation changes. Moreover, epigenetic remodeling is responsible for reprogramming somatic cells to totipotency upon somatic cell nuclear transfer/cloning, which is often incomplete and inefficient. Given that environmental factors, such as assisted reproductive techniques (ARTs), can affect epigenetic remodeling, there is interest in understanding the mechanisms driving these changes. We describe and discuss our current understanding of mechanisms responsible for the epigenetic remodeling that ensues during preimplantation development of mammals, presenting findings from studies of mouse embryos and when available comparing them to what is known for human and cattle embryos.
Collapse
Affiliation(s)
- Pablo J Ross
- Department of Animal Science, University of California Davis, Davis, CA, United States
| | - Rafael V Sampaio
- Department of Animal Science, University of California Davis, Davis, CA, United States.,Department of Animal Science, University of California Davis, Davis, CA, United States
| |
Collapse
|
27
|
Cui Y, Zhang Y, Wei Z, Gao J, Yu T, Chen R, Lv X, Pan C. Pig KDM5B: mRNA expression profiles of different tissues and testicular cells and association analyses with testicular morphology traits. Gene 2018; 650:27-33. [DOI: 10.1016/j.gene.2018.01.092] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 01/18/2018] [Accepted: 01/29/2018] [Indexed: 12/31/2022]
|
28
|
Histone demethylase lysine demethylase 5B in development and cancer. Oncotarget 2018; 8:8980-8991. [PMID: 27974677 PMCID: PMC5352456 DOI: 10.18632/oncotarget.13858] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 11/08/2016] [Indexed: 12/25/2022] Open
Abstract
Histone methylation is one of the most important chromatin posttranslational modifications. It has a range of influences on nuclear functions including epigenetic inheritance, transcriptional regulation and the maintenance of genome integrity. Changes in histone methylation status take part in various physiological and pathological processes. KDM5B (lysine demethylase 5B, also called JARID1B or PLU-1) encodes the histone H3 lysine4 (H3K4) demethylase and exhibits a strong transcriptional repression activity. KDM5B plays a role in cell differentiation, stem cell self-renewal and other developmental progresses. Recent studies showed that KDM5B expression was increased in breast, bladder, lung, prostate and many other tumors and promotes tumor initiation, invasion and metastasis. Given its association with tumor progression and prognosis of cancer patients, KDM5B was proposed to be a novel target for the prevention and treatment of human cancers. In this review, we will summarize recent advances in our understanding of the regulation and function of KDM5B in development and cancer.
Collapse
|
29
|
Zhai Y, Li W, Zhang Z, Cao Y, Wang Z, Zhang S, Li Z. Epigenetic states of donor cells significantly affect the development of somatic cell nuclear transfer (SCNT) embryos in pigs. Mol Reprod Dev 2017; 85:26-37. [PMID: 29205617 DOI: 10.1002/mrd.22935] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 11/29/2017] [Indexed: 01/31/2023]
Affiliation(s)
- Yanhui Zhai
- College of Veterinary Medicine; Jilin University; Changchun China
- First Hospital; Jilin University; Changchun China
| | - Wei Li
- First Hospital; Jilin University; Changchun China
| | - Zhiren Zhang
- College of Animal Science; Jilin University; Changchun China
| | - Yunqing Cao
- College of Veterinary Medicine; Jilin University; Changchun China
| | | | - Sheng Zhang
- First Hospital; Jilin University; Changchun China
| | - Ziyi Li
- First Hospital; Jilin University; Changchun China
| |
Collapse
|
30
|
Xu Q, Xie W. Epigenome in Early Mammalian Development: Inheritance, Reprogramming and Establishment. Trends Cell Biol 2017; 28:237-253. [PMID: 29217127 DOI: 10.1016/j.tcb.2017.10.008] [Citation(s) in RCA: 152] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 10/25/2017] [Accepted: 10/27/2017] [Indexed: 01/17/2023]
Abstract
Drastic epigenetic reprogramming takes place during preimplantation development, leading to the conversion of terminally differentiated gametes to a totipotent embryo. Deficiencies in remodeling of the epigenomes can cause severe developmental defects, including embryonic lethality. However, how chromatin modifications and chromatin organization are reprogrammed upon fertilization in mammals has long remained elusive. Here, we review recent progress in understanding how the epigenome is dynamically regulated during early mammalian development. The latest studies, including many from genome-wide perspectives, have revealed unusual principles of reprogramming for histone modifications, chromatin accessibility, and 3D chromatin architecture. These advances have shed light on the regulatory network controlling the earliest development and maternal-zygotic transition.
Collapse
Affiliation(s)
- Qianhua Xu
- Center for Stem Cell Biology and Regenerative Medicine, MOE Key Laboratory of Bioinformatics, THU-PKU Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Wei Xie
- Center for Stem Cell Biology and Regenerative Medicine, MOE Key Laboratory of Bioinformatics, THU-PKU Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
31
|
Chen Z, Zuo X, Li H, Hong R, Ding B, Liu C, Gao D, Shang H, Cao Z, Huang W, Zhang X, Zhang Y. Effects of melatonin on maturation, histone acetylation, autophagy of porcine oocytes and subsequent embryonic development. Anim Sci J 2017; 88:1298-1310. [PMID: 28349625 DOI: 10.1111/asj.12779] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Revised: 12/05/2016] [Accepted: 12/12/2016] [Indexed: 12/12/2022]
Abstract
Melatonin (MLT) is an endogenous hormone with roles in animal germ cell development. However, the effect of MLT on porcine oocyte maturation and its underlying mechanisms remain largely unknown. Here, we investigated the effects of exogenous MLT on oocyte maturation, histone acetylation, autophagy and subsequent embryonic development. We found that 1 nmol/L MLT supplemented in maturation medium was the optimal concentration to promote porcine oocyte maturation and subsequent developmental competence and quality of parthenogenetic embryos. Interestingly, the beneficial effects of 1 nmol/L MLT treatment on porcine oocyte maturation and embryo development were mainly attributed to the first half period of in vitro maturation. Simultaneously, MLT treatment could also improve maturation of small follicle-derived oocytes, morphologically poor (cumulus cell layer ≤1) and even artificially denuded oocytes and their subsequent embryo development. Furthermore, MLT treatment not only could decrease the levels of H3K27ac and H4K16ac in metaphase II (MII) oocytes, but also could increase the expression abundances of genes associated with cumulus cell expansion, meiotic maturation, histone acetylation and autophagy in cumulus cells or MII oocytes. These results indicate that MLT treatment can facilitate porcine oocyte maturation and subsequent embryonic development probably, through improvements in histone acetylation and autophagy in oocytes.
Collapse
Affiliation(s)
- Zhen Chen
- Anhui Provincial Laboratory of Local Livestock and Poultry Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui province, China
| | - Xiaoyuan Zuo
- Anhui Provincial Laboratory of Local Livestock and Poultry Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui province, China
| | - Hui Li
- Anhui Provincial Laboratory of Local Livestock and Poultry Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui province, China
| | - Renyun Hong
- Anhui Provincial Laboratory of Local Livestock and Poultry Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui province, China
| | - Biao Ding
- Anhui Provincial Laboratory of Local Livestock and Poultry Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui province, China
| | - Chengxue Liu
- Anhui Provincial Laboratory of Local Livestock and Poultry Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui province, China
| | - Di Gao
- Anhui Provincial Laboratory of Local Livestock and Poultry Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui province, China
| | - Hui Shang
- Anhui Provincial Laboratory of Local Livestock and Poultry Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui province, China
| | - Zubing Cao
- Anhui Provincial Laboratory of Local Livestock and Poultry Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui province, China
| | - Weiping Huang
- Anhui Provincial Laboratory of Local Livestock and Poultry Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui province, China
| | - Xiaorong Zhang
- Anhui Provincial Laboratory of Local Livestock and Poultry Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui province, China
| | - Yunhai Zhang
- Anhui Provincial Laboratory of Local Livestock and Poultry Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui province, China
| |
Collapse
|
32
|
Ross PJ, Canovas S. Mechanisms of epigenetic remodelling during preimplantation development. Reprod Fertil Dev 2017; 28:25-40. [PMID: 27062872 DOI: 10.1071/rd15365] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Epigenetics involves mechanisms independent of modifications in the DNA sequence that result in changes in gene expression and are maintained through cell divisions. Because all cells in the organism contain the same genetic blueprint, epigenetics allows for cells to assume different phenotypes and maintain them upon cell replication. As such, during the life cycle, there are moments in which the epigenetic information needs to be reset for the initiation of a new organism. In mammals, the resetting of epigenetic marks occurs at two different moments, which both happen to be during gestation, and include primordial germ cells (PGCs) and early preimplantation embryos. Because epigenetic information is reversible and sensitive to environmental changes, it is probably no coincidence that both these extensive periods of epigenetic remodelling happen in the female reproductive tract, under a finely controlled maternal environment. It is becoming evident that perturbations during the extensive epigenetic remodelling in PGCs and embryos can lead to permanent and inheritable changes to the epigenome that can result in long-term changes to the offspring derived from them, as indicated by the Developmental Origins of Health and Disease (DOHaD) hypothesis and recent demonstration of inter- and trans-generational epigenetic alterations. In this context, an understanding of the mechanisms of epigenetic remodelling during early embryo development is important to assess the potential for gametic epigenetic mutations to contribute to the offspring and for new epimutations to be established during embryo manipulations that could affect a large number of cells in the offspring. It is of particular interest to understand whether and how epigenetic information can be passed on from the gametes to the embryo or offspring, and whether abnormalities in this process could lead to transgenerationally inheritable phenotypes. The aim of this review is to highlight recent progress made in understanding the nature and mechanisms of epigenetic remodelling that ensue after fertilisation.
Collapse
Affiliation(s)
- Pablo Juan Ross
- Department of Animal Science, University of California, Davis, CA 95616 USA
| | - Sebastian Canovas
- LARCEL (Laboratorio Andaluz de Reprogramación Celular), BIONAND, Centro Andaluz de Nanomedicina y Biotecnología Campanillas, Malaga 29590, Spain
| |
Collapse
|
33
|
Distinct features of H3K4me3 and H3K27me3 chromatin domains in pre-implantation embryos. Nature 2016; 537:558-562. [DOI: 10.1038/nature19362] [Citation(s) in RCA: 410] [Impact Index Per Article: 45.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2015] [Accepted: 08/17/2016] [Indexed: 12/31/2022]
|
34
|
Cao Z, Li Y, Chen Z, Wang H, Zhang M, Zhou N, Wu R, Ling Y, Fang F, Li N, Zhang Y. Genome-Wide Dynamic Profiling of Histone Methylation during Nuclear Transfer-Mediated Porcine Somatic Cell Reprogramming. PLoS One 2015; 10:e0144897. [PMID: 26683029 PMCID: PMC4687693 DOI: 10.1371/journal.pone.0144897] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 11/24/2015] [Indexed: 01/01/2023] Open
Abstract
The low full-term developmental efficiency of porcine somatic cell nuclear transfer (SCNT) embryos is mainly attributed to imperfect epigenetic reprogramming in the early embryos. However, dynamic expression patterns of histone methylation involved in epigenetic reprogramming progression during porcine SCNT embryo early development remain to be unknown. In this study, we characterized and compared the expression patterns of multiple histone methylation markers including transcriptionally repressive (H3K9me2, H3K9me3, H3K27me2, H3K27me3, H4K20me2 and H4K20me3) and active modifications (H3K4me2, H3K4me3, H3K36me2, H3K36me3, H3K79me2 and H3K79me3) in SCNT early embryos from different developmental stages with that from in vitro fertilization (IVF) counterparts. We found that the expression level of H3K9me2, H3K9me3 and H4K20me3 of SCNT embryos from 1-cell to 4-cell stages was significantly higher than that in the IVF embryos. We also detected a symmetric distribution pattern of H3K9me2 between inner cell mass (ICM) and trophectoderm (TE) in SCNT blastocysts. The expression level of H3K9me2 in both lineages from SCNT expanded blastocyst onwards was significantly higher than that in IVF counterparts. The expression level of H4K20me2 was significantly lower in SCNT embryos from morula to blastocyst stage compared with IVF embryos. However, no aberrant dynamic reprogramming of H3K27me2/3 occurred during early developmental stages of SCNT embryos. The expression of H3K4me3 was higher in SCNT embryos at 4-cell stage than that of IVF embryos. H3K4me2 expression in SCNT embryos from 8-cell stage to blastocyst stage was lower than that in the IVF embryos. Dynamic patterns of other active histone methylation markers were similar between SCNT and IVF embryos. Taken together, histone methylation exhibited developmentally stage-specific abnormal expression patterns in porcine SCNT early embryos.
Collapse
Affiliation(s)
- Zubing Cao
- Anhui Provincial Laboratory of Local Livestock and Poultry Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei City, Anhui Province, China
- State Key Laboratory for Agrobiotechnology, College of Biological Science, China Agricultural University, Haidian District, Beijing, China
| | - Yunsheng Li
- Anhui Provincial Laboratory of Local Livestock and Poultry Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei City, Anhui Province, China
| | - Zhen Chen
- Anhui Provincial Laboratory of Local Livestock and Poultry Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei City, Anhui Province, China
| | - Heng Wang
- Anhui Provincial Laboratory of Local Livestock and Poultry Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei City, Anhui Province, China
| | - Meiling Zhang
- Anhui Provincial Laboratory of Local Livestock and Poultry Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei City, Anhui Province, China
| | - Naru Zhou
- Anhui Provincial Laboratory of Local Livestock and Poultry Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei City, Anhui Province, China
| | - Ronghua Wu
- Anhui Provincial Laboratory of Local Livestock and Poultry Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei City, Anhui Province, China
| | - Yinghui Ling
- Anhui Provincial Laboratory of Local Livestock and Poultry Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei City, Anhui Province, China
| | - Fugui Fang
- Anhui Provincial Laboratory of Local Livestock and Poultry Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei City, Anhui Province, China
| | - Ning Li
- State Key Laboratory for Agrobiotechnology, College of Biological Science, China Agricultural University, Haidian District, Beijing, China
- * E-mail: (YHZ); (NL)
| | - Yunhai Zhang
- Anhui Provincial Laboratory of Local Livestock and Poultry Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei City, Anhui Province, China
- * E-mail: (YHZ); (NL)
| |
Collapse
|
35
|
Marcho C, Cui W, Mager J. Epigenetic dynamics during preimplantation development. Reproduction 2015; 150:R109-20. [PMID: 26031750 DOI: 10.1530/rep-15-0180] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 06/01/2015] [Indexed: 12/31/2022]
Abstract
Successful mammalian development requires descendants of single-cell zygotes to differentiate into diverse cell types even though they contain the same genetic material. Preimplantation dynamics are first driven by the necessity of reprogramming haploid parental epigenomes to reach a totipotent state. This process requires extensive erasure of epigenetic marks shortly after fertilization. During the few short days after formation of the zygote, epigenetic programs are established and are essential for the first lineage decisions and differentiation. Here we review the current understanding of DNA methylation and histone modification dynamics responsible for these early changes during mammalian preimplantation development. In particular, we highlight insights that have been gained through next-generation sequencing technologies comparing human embryos to other models as well as the recent discoveries of active DNA demethylation mechanisms at play during preimplantation.
Collapse
Affiliation(s)
- Chelsea Marcho
- Department of Veterinary and Animal ScienceUniversity of Massachusetts Amherst, 661 North Pleasant Street, Amherst, Massachusetts 01003, USA
| | - Wei Cui
- Department of Veterinary and Animal ScienceUniversity of Massachusetts Amherst, 661 North Pleasant Street, Amherst, Massachusetts 01003, USA
| | - Jesse Mager
- Department of Veterinary and Animal ScienceUniversity of Massachusetts Amherst, 661 North Pleasant Street, Amherst, Massachusetts 01003, USA
| |
Collapse
|
36
|
Fork C, Gu L, Hitzel J, Josipovic I, Hu J, SzeKa Wong M, Ponomareva Y, Albert M, Schmitz SU, Uchida S, Fleming I, Helin K, Steinhilber D, Leisegang MS, Brandes RP. Epigenetic Regulation of Angiogenesis by JARID1B-Induced Repression of HOXA5. Arterioscler Thromb Vasc Biol 2015; 35:1645-52. [PMID: 26023081 DOI: 10.1161/atvbaha.115.305561] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 05/17/2015] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Altering endothelial biology through epigenetic modifiers is an attractive novel concept, which is, however, just in its beginnings. We therefore set out to identify chromatin modifiers important for endothelial gene expression and contributing to angiogenesis. APPROACH AND RESULTS To identify chromatin modifying enzymes in endothelial cells, histone demethylases were screened by microarray and polymerase chain reaction. The histone 3 lysine 4 demethylase JARID1B was identified as a highly expressed enzyme at the mRNA and protein levels. Knockdown of JARID1B by shRNA in human umbilical vein endothelial cells attenuated cell migration, angiogenic sprouting, and tube formation. Similarly, pharmacological inhibition and overexpression of a catalytic inactive JARID1B mutant reduced the angiogenic capacity of human umbilical vein endothelial cells. To identify the in vivo relevance of JARID1B in the vascular system, Jarid1b knockout mice were studied. As global knockout results in increased mortality and developmental defects, tamoxifen-inducible and endothelial-specific knockout mice were generated. Acute knockout of Jarid1b attenuated retinal angiogenesis and endothelial sprout outgrowth from aortic segments. To identify the underlying mechanism, a microarray experiment was performed, which led to the identification of the antiangiogenic transcription factor HOXA5 to be suppressed by JARID1B. Importantly, downregulation or inhibition of JARID1B, but not of JARID1A and JARID1C, induced HOXA5 expression in human umbilical vein endothelial cells. Consistently, chromatin immunoprecipitation revealed that JARID1B occupies and reduces the histone 3 lysine 4 methylation levels at the HOXA5 promoter, demonstrating a direct function of JARID1B in endothelial HOXA5 gene regulation. CONCLUSIONS JARID1B, by suppressing HOXA5, maintains the endothelial angiogenic capacity in a demethylase-dependent manner.
Collapse
Affiliation(s)
- Christian Fork
- From the Institute for Cardiovascular Physiology, Medical Faculty (C.F., L.G., J.H., I.J., M.S.W., M.S.L., R.P.B.), Institutes of Vascular Signalling (J.H., I.F.) and Cardiovascular Regeneration (Y.P., S.U.), Centre for Molecular Medicine, and Institute of Pharmaceutical Chemistry/ZAFES (D.S.), Goethe-University Frankfurt, Frankfurt am Main, Germany; Biotech Research and Innovation Centre (BRIC) (M.A., S.U.S., K.H.), Centre for Epigenetics (M.A., S.U.S., K.H.), University of Copenhagen, Copenhagen, Denmark; and German Center for Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany (C.F., L.G., J.H., I.J., M.S.W., Y.P., S.U., I.F., M.S.L., R.P.B.).
| | - Lunda Gu
- From the Institute for Cardiovascular Physiology, Medical Faculty (C.F., L.G., J.H., I.J., M.S.W., M.S.L., R.P.B.), Institutes of Vascular Signalling (J.H., I.F.) and Cardiovascular Regeneration (Y.P., S.U.), Centre for Molecular Medicine, and Institute of Pharmaceutical Chemistry/ZAFES (D.S.), Goethe-University Frankfurt, Frankfurt am Main, Germany; Biotech Research and Innovation Centre (BRIC) (M.A., S.U.S., K.H.), Centre for Epigenetics (M.A., S.U.S., K.H.), University of Copenhagen, Copenhagen, Denmark; and German Center for Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany (C.F., L.G., J.H., I.J., M.S.W., Y.P., S.U., I.F., M.S.L., R.P.B.)
| | - Juliane Hitzel
- From the Institute for Cardiovascular Physiology, Medical Faculty (C.F., L.G., J.H., I.J., M.S.W., M.S.L., R.P.B.), Institutes of Vascular Signalling (J.H., I.F.) and Cardiovascular Regeneration (Y.P., S.U.), Centre for Molecular Medicine, and Institute of Pharmaceutical Chemistry/ZAFES (D.S.), Goethe-University Frankfurt, Frankfurt am Main, Germany; Biotech Research and Innovation Centre (BRIC) (M.A., S.U.S., K.H.), Centre for Epigenetics (M.A., S.U.S., K.H.), University of Copenhagen, Copenhagen, Denmark; and German Center for Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany (C.F., L.G., J.H., I.J., M.S.W., Y.P., S.U., I.F., M.S.L., R.P.B.)
| | - Ivana Josipovic
- From the Institute for Cardiovascular Physiology, Medical Faculty (C.F., L.G., J.H., I.J., M.S.W., M.S.L., R.P.B.), Institutes of Vascular Signalling (J.H., I.F.) and Cardiovascular Regeneration (Y.P., S.U.), Centre for Molecular Medicine, and Institute of Pharmaceutical Chemistry/ZAFES (D.S.), Goethe-University Frankfurt, Frankfurt am Main, Germany; Biotech Research and Innovation Centre (BRIC) (M.A., S.U.S., K.H.), Centre for Epigenetics (M.A., S.U.S., K.H.), University of Copenhagen, Copenhagen, Denmark; and German Center for Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany (C.F., L.G., J.H., I.J., M.S.W., Y.P., S.U., I.F., M.S.L., R.P.B.)
| | - Jiong Hu
- From the Institute for Cardiovascular Physiology, Medical Faculty (C.F., L.G., J.H., I.J., M.S.W., M.S.L., R.P.B.), Institutes of Vascular Signalling (J.H., I.F.) and Cardiovascular Regeneration (Y.P., S.U.), Centre for Molecular Medicine, and Institute of Pharmaceutical Chemistry/ZAFES (D.S.), Goethe-University Frankfurt, Frankfurt am Main, Germany; Biotech Research and Innovation Centre (BRIC) (M.A., S.U.S., K.H.), Centre for Epigenetics (M.A., S.U.S., K.H.), University of Copenhagen, Copenhagen, Denmark; and German Center for Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany (C.F., L.G., J.H., I.J., M.S.W., Y.P., S.U., I.F., M.S.L., R.P.B.)
| | - Michael SzeKa Wong
- From the Institute for Cardiovascular Physiology, Medical Faculty (C.F., L.G., J.H., I.J., M.S.W., M.S.L., R.P.B.), Institutes of Vascular Signalling (J.H., I.F.) and Cardiovascular Regeneration (Y.P., S.U.), Centre for Molecular Medicine, and Institute of Pharmaceutical Chemistry/ZAFES (D.S.), Goethe-University Frankfurt, Frankfurt am Main, Germany; Biotech Research and Innovation Centre (BRIC) (M.A., S.U.S., K.H.), Centre for Epigenetics (M.A., S.U.S., K.H.), University of Copenhagen, Copenhagen, Denmark; and German Center for Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany (C.F., L.G., J.H., I.J., M.S.W., Y.P., S.U., I.F., M.S.L., R.P.B.)
| | - Yuliya Ponomareva
- From the Institute for Cardiovascular Physiology, Medical Faculty (C.F., L.G., J.H., I.J., M.S.W., M.S.L., R.P.B.), Institutes of Vascular Signalling (J.H., I.F.) and Cardiovascular Regeneration (Y.P., S.U.), Centre for Molecular Medicine, and Institute of Pharmaceutical Chemistry/ZAFES (D.S.), Goethe-University Frankfurt, Frankfurt am Main, Germany; Biotech Research and Innovation Centre (BRIC) (M.A., S.U.S., K.H.), Centre for Epigenetics (M.A., S.U.S., K.H.), University of Copenhagen, Copenhagen, Denmark; and German Center for Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany (C.F., L.G., J.H., I.J., M.S.W., Y.P., S.U., I.F., M.S.L., R.P.B.)
| | - Mareike Albert
- From the Institute for Cardiovascular Physiology, Medical Faculty (C.F., L.G., J.H., I.J., M.S.W., M.S.L., R.P.B.), Institutes of Vascular Signalling (J.H., I.F.) and Cardiovascular Regeneration (Y.P., S.U.), Centre for Molecular Medicine, and Institute of Pharmaceutical Chemistry/ZAFES (D.S.), Goethe-University Frankfurt, Frankfurt am Main, Germany; Biotech Research and Innovation Centre (BRIC) (M.A., S.U.S., K.H.), Centre for Epigenetics (M.A., S.U.S., K.H.), University of Copenhagen, Copenhagen, Denmark; and German Center for Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany (C.F., L.G., J.H., I.J., M.S.W., Y.P., S.U., I.F., M.S.L., R.P.B.)
| | - Sandra U Schmitz
- From the Institute for Cardiovascular Physiology, Medical Faculty (C.F., L.G., J.H., I.J., M.S.W., M.S.L., R.P.B.), Institutes of Vascular Signalling (J.H., I.F.) and Cardiovascular Regeneration (Y.P., S.U.), Centre for Molecular Medicine, and Institute of Pharmaceutical Chemistry/ZAFES (D.S.), Goethe-University Frankfurt, Frankfurt am Main, Germany; Biotech Research and Innovation Centre (BRIC) (M.A., S.U.S., K.H.), Centre for Epigenetics (M.A., S.U.S., K.H.), University of Copenhagen, Copenhagen, Denmark; and German Center for Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany (C.F., L.G., J.H., I.J., M.S.W., Y.P., S.U., I.F., M.S.L., R.P.B.)
| | - Shizuka Uchida
- From the Institute for Cardiovascular Physiology, Medical Faculty (C.F., L.G., J.H., I.J., M.S.W., M.S.L., R.P.B.), Institutes of Vascular Signalling (J.H., I.F.) and Cardiovascular Regeneration (Y.P., S.U.), Centre for Molecular Medicine, and Institute of Pharmaceutical Chemistry/ZAFES (D.S.), Goethe-University Frankfurt, Frankfurt am Main, Germany; Biotech Research and Innovation Centre (BRIC) (M.A., S.U.S., K.H.), Centre for Epigenetics (M.A., S.U.S., K.H.), University of Copenhagen, Copenhagen, Denmark; and German Center for Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany (C.F., L.G., J.H., I.J., M.S.W., Y.P., S.U., I.F., M.S.L., R.P.B.)
| | - Ingrid Fleming
- From the Institute for Cardiovascular Physiology, Medical Faculty (C.F., L.G., J.H., I.J., M.S.W., M.S.L., R.P.B.), Institutes of Vascular Signalling (J.H., I.F.) and Cardiovascular Regeneration (Y.P., S.U.), Centre for Molecular Medicine, and Institute of Pharmaceutical Chemistry/ZAFES (D.S.), Goethe-University Frankfurt, Frankfurt am Main, Germany; Biotech Research and Innovation Centre (BRIC) (M.A., S.U.S., K.H.), Centre for Epigenetics (M.A., S.U.S., K.H.), University of Copenhagen, Copenhagen, Denmark; and German Center for Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany (C.F., L.G., J.H., I.J., M.S.W., Y.P., S.U., I.F., M.S.L., R.P.B.)
| | - Kristian Helin
- From the Institute for Cardiovascular Physiology, Medical Faculty (C.F., L.G., J.H., I.J., M.S.W., M.S.L., R.P.B.), Institutes of Vascular Signalling (J.H., I.F.) and Cardiovascular Regeneration (Y.P., S.U.), Centre for Molecular Medicine, and Institute of Pharmaceutical Chemistry/ZAFES (D.S.), Goethe-University Frankfurt, Frankfurt am Main, Germany; Biotech Research and Innovation Centre (BRIC) (M.A., S.U.S., K.H.), Centre for Epigenetics (M.A., S.U.S., K.H.), University of Copenhagen, Copenhagen, Denmark; and German Center for Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany (C.F., L.G., J.H., I.J., M.S.W., Y.P., S.U., I.F., M.S.L., R.P.B.)
| | - Dieter Steinhilber
- From the Institute for Cardiovascular Physiology, Medical Faculty (C.F., L.G., J.H., I.J., M.S.W., M.S.L., R.P.B.), Institutes of Vascular Signalling (J.H., I.F.) and Cardiovascular Regeneration (Y.P., S.U.), Centre for Molecular Medicine, and Institute of Pharmaceutical Chemistry/ZAFES (D.S.), Goethe-University Frankfurt, Frankfurt am Main, Germany; Biotech Research and Innovation Centre (BRIC) (M.A., S.U.S., K.H.), Centre for Epigenetics (M.A., S.U.S., K.H.), University of Copenhagen, Copenhagen, Denmark; and German Center for Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany (C.F., L.G., J.H., I.J., M.S.W., Y.P., S.U., I.F., M.S.L., R.P.B.)
| | - Matthias S Leisegang
- From the Institute for Cardiovascular Physiology, Medical Faculty (C.F., L.G., J.H., I.J., M.S.W., M.S.L., R.P.B.), Institutes of Vascular Signalling (J.H., I.F.) and Cardiovascular Regeneration (Y.P., S.U.), Centre for Molecular Medicine, and Institute of Pharmaceutical Chemistry/ZAFES (D.S.), Goethe-University Frankfurt, Frankfurt am Main, Germany; Biotech Research and Innovation Centre (BRIC) (M.A., S.U.S., K.H.), Centre for Epigenetics (M.A., S.U.S., K.H.), University of Copenhagen, Copenhagen, Denmark; and German Center for Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany (C.F., L.G., J.H., I.J., M.S.W., Y.P., S.U., I.F., M.S.L., R.P.B.)
| | - Ralf P Brandes
- From the Institute for Cardiovascular Physiology, Medical Faculty (C.F., L.G., J.H., I.J., M.S.W., M.S.L., R.P.B.), Institutes of Vascular Signalling (J.H., I.F.) and Cardiovascular Regeneration (Y.P., S.U.), Centre for Molecular Medicine, and Institute of Pharmaceutical Chemistry/ZAFES (D.S.), Goethe-University Frankfurt, Frankfurt am Main, Germany; Biotech Research and Innovation Centre (BRIC) (M.A., S.U.S., K.H.), Centre for Epigenetics (M.A., S.U.S., K.H.), University of Copenhagen, Copenhagen, Denmark; and German Center for Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany (C.F., L.G., J.H., I.J., M.S.W., Y.P., S.U., I.F., M.S.L., R.P.B.)
| |
Collapse
|