1
|
Su G, Wei Z, Bai C, Li D, Zhao X, Liu X, Song L, Zhang L, Li G, Yang L. Generation of Codon-Optimized Fad3 Gene Transgenic Bovine That Produce More n-3 Polyunsaturated Fatty Acids. Animals (Basel) 2025; 15:93. [PMID: 39795036 PMCID: PMC11718938 DOI: 10.3390/ani15010093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 01/01/2025] [Accepted: 01/02/2025] [Indexed: 01/13/2025] Open
Abstract
Polyunsaturated fatty acids (PUFAs) such as linoleic acid (18:2, n-6) and α-linolenic acid (18:3, n-3) are essential for the growth, development, and well-being of mammals. However, most mammals, including humans, cannot synthesize n-3 and n-6 PUFAs and these must be obtained through diet. The beneficial effect of converting n-6 polyunsaturated fatty acids (n-6 PUFAs) into n-3 polyunsaturated fatty acids (n-3 PUFAs) has led to extensive research on the flax fatty acid desaturase 3 (Fad3) gene, which encodes fatty acid desaturase. Still, the plant-derived Fad3 gene is used much less in transgenic animals than the Fat-1 gene from Caenorhabditis elegans. To address this problem, we used somatic cell nuclear transfer (SCNT) technology to create codon-optimized Fad3 transgenic cattle. Gas chromatographic analysis showed that the n-3 PUFA content of transgenic cattle increased significantly, and the ratio of n-6 PUFAs to n-3 PUFAs decreased from 3.484 ± 0.46 to about 2.78 ± 0.14 (p < 0.05). In conclusion, Fad3 gene knock-in cattle are expected to improve the nutritional value of beef and can be used as an animal model to study the therapeutic effects of n-3 PUFAs in various diseases.
Collapse
Affiliation(s)
- Guanghua Su
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock (R2BGL), Inner Mongolia University, 24 Zhaojun Rd., Hohhot 010070, China; (Z.W.); (C.B.); (D.L.); (X.Z.); (X.L.); (L.S.); (L.Z.); (G.L.)
- College of Life Sciences, Inner Mongolia University, 24 Zhaojun Rd., Hohhot 010070, China
| | - Zhuying Wei
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock (R2BGL), Inner Mongolia University, 24 Zhaojun Rd., Hohhot 010070, China; (Z.W.); (C.B.); (D.L.); (X.Z.); (X.L.); (L.S.); (L.Z.); (G.L.)
- College of Life Sciences, Inner Mongolia University, 24 Zhaojun Rd., Hohhot 010070, China
| | - Chunling Bai
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock (R2BGL), Inner Mongolia University, 24 Zhaojun Rd., Hohhot 010070, China; (Z.W.); (C.B.); (D.L.); (X.Z.); (X.L.); (L.S.); (L.Z.); (G.L.)
- College of Life Sciences, Inner Mongolia University, 24 Zhaojun Rd., Hohhot 010070, China
| | - Danyi Li
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock (R2BGL), Inner Mongolia University, 24 Zhaojun Rd., Hohhot 010070, China; (Z.W.); (C.B.); (D.L.); (X.Z.); (X.L.); (L.S.); (L.Z.); (G.L.)
- College of Life Sciences, Inner Mongolia University, 24 Zhaojun Rd., Hohhot 010070, China
| | - Xiaoyu Zhao
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock (R2BGL), Inner Mongolia University, 24 Zhaojun Rd., Hohhot 010070, China; (Z.W.); (C.B.); (D.L.); (X.Z.); (X.L.); (L.S.); (L.Z.); (G.L.)
- College of Life Sciences, Inner Mongolia University, 24 Zhaojun Rd., Hohhot 010070, China
| | - Xuefei Liu
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock (R2BGL), Inner Mongolia University, 24 Zhaojun Rd., Hohhot 010070, China; (Z.W.); (C.B.); (D.L.); (X.Z.); (X.L.); (L.S.); (L.Z.); (G.L.)
- College of Life Sciences, Inner Mongolia University, 24 Zhaojun Rd., Hohhot 010070, China
| | - Lishuang Song
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock (R2BGL), Inner Mongolia University, 24 Zhaojun Rd., Hohhot 010070, China; (Z.W.); (C.B.); (D.L.); (X.Z.); (X.L.); (L.S.); (L.Z.); (G.L.)
- College of Life Sciences, Inner Mongolia University, 24 Zhaojun Rd., Hohhot 010070, China
| | - Li Zhang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock (R2BGL), Inner Mongolia University, 24 Zhaojun Rd., Hohhot 010070, China; (Z.W.); (C.B.); (D.L.); (X.Z.); (X.L.); (L.S.); (L.Z.); (G.L.)
- College of Life Sciences, Inner Mongolia University, 24 Zhaojun Rd., Hohhot 010070, China
| | - Guangpeng Li
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock (R2BGL), Inner Mongolia University, 24 Zhaojun Rd., Hohhot 010070, China; (Z.W.); (C.B.); (D.L.); (X.Z.); (X.L.); (L.S.); (L.Z.); (G.L.)
- College of Life Sciences, Inner Mongolia University, 24 Zhaojun Rd., Hohhot 010070, China
| | - Lei Yang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock (R2BGL), Inner Mongolia University, 24 Zhaojun Rd., Hohhot 010070, China; (Z.W.); (C.B.); (D.L.); (X.Z.); (X.L.); (L.S.); (L.Z.); (G.L.)
- College of Life Sciences, Inner Mongolia University, 24 Zhaojun Rd., Hohhot 010070, China
| |
Collapse
|
2
|
Montgomery T, Uh K, Lee K. TET enzyme driven epigenetic reprogramming in early embryos and its implication on long-term health. Front Cell Dev Biol 2024; 12:1358649. [PMID: 39149518 PMCID: PMC11324557 DOI: 10.3389/fcell.2024.1358649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 07/23/2024] [Indexed: 08/17/2024] Open
Abstract
Mammalian embryo development is initiated by the union of paternal and maternal gametes. Upon fertilization, their epigenome landscape is transformed through a series of finely orchestrated mechanisms that are crucial for survival and successful embryogenesis. Specifically, maternal or oocyte-specific reprogramming factors modulate germ cell specific epigenetic marks into their embryonic states. Rapid and dynamic changes in epigenetic marks such as DNA methylation and histone modifications are observed during early embryo development. These changes govern the structure of embryonic genome prior to zygotic genome activation. Differential changes in epigenetic marks are observed between paternal and maternal genomes because the structure of the parental genomes allows interaction with specific oocyte reprogramming factors. For instance, the paternal genome is targeted by the TET family of enzymes which oxidize the 5-methylcytosine (5mC) epigenetic mark into 5-hydroxymethylcytosine (5hmC) to lower the level of DNA methylation. The maternal genome is mainly protected from TET3-mediated oxidation by the maternal factor, STELLA. The TET3-mediated DNA demethylation occurs at the global level and is clearly observed in many mammalian species. Other epigenetic modulating enzymes, such as DNA methyltransferases, provide fine tuning of the DNA methylation level by initiating de novo methylation. The mechanisms which initiate the epigenetic reprogramming of gametes are critical for proper activation of embryonic genome and subsequent establishment of pluripotency and normal development. Clinical cases or diseases linked to mutations in reprogramming modulators exist, emphasizing the need to understand mechanistic actions of these modulators. In addition, embryos generated via in vitro embryo production system often present epigenetic abnormalities. Understanding mechanistic actions of the epigenetic modulators will potentially improve the well-being of individuals suffering from these epigenetic disorders and correct epigenetic abnormalities in embryos produced in vitro. This review will summarize the current understanding of epigenetic reprogramming by TET enzymes during early embryogenesis and highlight its clinical relevance and potential implication for assisted reproductive technologies.
Collapse
Affiliation(s)
- Ty Montgomery
- Division of Animal Sciences, University of Missouri, Columbia, MO, United States
| | - Kyungjun Uh
- Futuristic Animal Resource and Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si, Republic of Korea
| | - Kiho Lee
- Division of Animal Sciences, University of Missouri, Columbia, MO, United States
| |
Collapse
|
3
|
Sindhu P, Magotra A, Sindhu V, Chaudhary P. Unravelling the impact of epigenetic mechanisms on offspring growth, production, reproduction and disease susceptibility. ZYGOTE 2024; 32:190-206. [PMID: 39291610 DOI: 10.1017/s0967199424000224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Epigenetic mechanisms, such as DNA methylation, histone modifications and non-coding RNA molecules, play a critical role in gene expression and regulation in livestock species, influencing development, reproduction and disease resistance. DNA methylation patterns silence gene expression by blocking transcription factor binding, while histone modifications alter chromatin structure and affect DNA accessibility. Livestock-specific histone modifications contribute to gene expression and genome stability. Non-coding RNAs, including miRNAs, piRNAs, siRNAs, snoRNAs, lncRNAs and circRNAs, regulate gene expression post-transcriptionally. Transgenerational epigenetic inheritance occurs in livestock, with environmental factors impacting epigenetic modifications and phenotypic traits across generations. Epigenetic regulation revealed significant effect on gene expression profiling that can be exploited for various targeted traits like muscle hypertrophy, puberty onset, growth, metabolism, disease resistance and milk production in livestock and poultry breeds. Epigenetic regulation of imprinted genes affects cattle growth and metabolism while epigenetic modifications play a role in disease resistance and mastitis in dairy cattle, as well as milk protein gene regulation during lactation. Nutri-epigenomics research also reveals the influence of maternal nutrition on offspring's epigenetic regulation of metabolic homeostasis in cattle, sheep, goat and poultry. Integrating cyto-genomics approaches enhances understanding of epigenetic mechanisms in livestock breeding, providing insights into chromosomal structure, rearrangements and their impact on gene regulation and phenotypic traits. This review presents potential research areas to enhance production potential and deepen our understanding of epigenetic changes in livestock, offering opportunities for genetic improvement, reproductive management, disease control and milk production in diverse livestock species.
Collapse
Affiliation(s)
- Pushpa Sindhu
- Department of Animal Genetics and Breeding, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, Haryana, India
| | - Ankit Magotra
- Department of Animal Genetics and Breeding, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, Haryana, India
| | - Vikas Sindhu
- Department of Animal Nutrition, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, Haryana, India
| | - Pradeep Chaudhary
- Department of Animal Genetics and Breeding, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, Haryana, India
| |
Collapse
|
4
|
Ren X, Tong Y, Yang T, Huang S, Xu T, Xue Q, Shi D, Li X. Overexpression of BRG1 improves early development of porcine somatic cell nuclear transfer embryos. Theriogenology 2024; 217:51-63. [PMID: 38245973 DOI: 10.1016/j.theriogenology.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/15/2023] [Accepted: 01/04/2024] [Indexed: 01/23/2024]
Abstract
The epigenetic modification levels of donor cells directly affect the developmental potential of somatic cell nuclear transfer (SCNT) embryos. BRG1, as an epigenetic modifying enzyme, has not yet been studied in donor cells and SCNT embryos. In this study, BRG1 was overexpressed in porcine fetal fibroblasts (PFFs), its effect on chromatin openness and gene transcription was examined, subsequently, the development potential of porcine SCNT embryos was investigated. The results showed that compared with the control group, the percentage of G1 phase cells was significantly increased (32.3 % ± 0.87 vs 25.7 % ± 0.81, P < 0.05) in the experimental group. The qRT-PCR results showed that the expression of H3K9me3-related genes was significantly decreased (P < 0.05), HAT1 was significantly increased (P < 0.05). Assay of Transposase Accessible Chromatin sequencing (ATAC-seq) results revealed that SMARCA4、NANOG、SOX2、MAP2K6 and HIF1A loci had more open chromatin peaks in the experimental group. The RNA-seq results showed that the upregulated genes were mainly enriched in PI3K/AKT and WNT signaling pathways, and the downregulated genes were largely focused on disease development. Interestingly, the developmental rate of porcine SCNT embryos was improved (27.33 % ± 1.40 vs 17.83 % ± 2.02, P < 0.05), the expression of zygotic gene activation-related genes in 4-cell embryos, and embryonic development-related genes in blastocysts was significantly upregulated in the experimental group (P < 0.05). These results suggest that overexpression of BRG1 in donor cells is benefit for the developmental potential of porcine SCNT embryos.
Collapse
Affiliation(s)
- Xuan Ren
- Guangxi Key Laboratory of Animal Breeding and Disease Control, Guangxi University, Nanning, 530005, China
| | - Yi Tong
- Guangxi Key Laboratory of Animal Breeding and Disease Control, Guangxi University, Nanning, 530005, China
| | - Ting Yang
- Guangxi Key Laboratory of Animal Breeding and Disease Control, Guangxi University, Nanning, 530005, China
| | - Shihai Huang
- College of Life Science and Technology, Guangxi University, Nanning, China
| | - Tairan Xu
- Guangxi Key Laboratory of Animal Breeding and Disease Control, Guangxi University, Nanning, 530005, China
| | - Qingsong Xue
- Guangxi Key Laboratory of Animal Breeding and Disease Control, Guangxi University, Nanning, 530005, China
| | - Deshun Shi
- Guangxi Key Laboratory of Animal Breeding and Disease Control, Guangxi University, Nanning, 530005, China
| | - Xiangping Li
- Guangxi Key Laboratory of Animal Breeding and Disease Control, Guangxi University, Nanning, 530005, China.
| |
Collapse
|
5
|
Fatima N, Saif Ur Rahman M, Qasim M, Ali Ashfaq U, Ahmed U, Masoud MS. Transcriptional Factors Mediated Reprogramming to Pluripotency. Curr Stem Cell Res Ther 2024; 19:367-388. [PMID: 37073151 DOI: 10.2174/1574888x18666230417084518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 02/01/2023] [Accepted: 02/06/2023] [Indexed: 04/20/2023]
Abstract
A unique kind of pluripotent cell, i.e., Induced pluripotent stem cells (iPSCs), now being targeted for iPSC synthesis, are produced by reprogramming animal and human differentiated cells (with no change in genetic makeup for the sake of high efficacy iPSCs formation). The conversion of specific cells to iPSCs has revolutionized stem cell research by making pluripotent cells more controllable for regenerative therapy. For the past 15 years, somatic cell reprogramming to pluripotency with force expression of specified factors has been a fascinating field of biomedical study. For that technological primary viewpoint reprogramming method, a cocktail of four transcription factors (TF) has required: Kruppel-like factor 4 (KLF4), four-octamer binding protein 34 (OCT3/4), MYC and SOX2 (together referred to as OSKM) and host cells. IPS cells have great potential for future tissue replacement treatments because of their ability to self-renew and specialize in all adult cell types, although factor-mediated reprogramming mechanisms are still poorly understood medically. This technique has dramatically improved performance and efficiency, making it more useful in drug discovery, disease remodeling, and regenerative medicine. Moreover, in these four TF cocktails, more than 30 reprogramming combinations were proposed, but for reprogramming effectiveness, only a few numbers have been demonstrated for the somatic cells of humans and mice. Stoichiometry, a combination of reprogramming agents and chromatin remodeling compounds, impacts kinetics, quality, and efficiency in stem cell research.
Collapse
Affiliation(s)
- Nazira Fatima
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Muhammad Saif Ur Rahman
- Institute of Advanced Studies, Shenzhen University, Shenzhen, 518060, China
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Muhammad Qasim
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| | - Usman Ali Ashfaq
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| | - Uzair Ahmed
- EMBL Partnership Institute for Genome Editing Technologies, Vilnius University, Vilnius, 10257, Lithuania
| | - Muhammad Shareef Masoud
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| |
Collapse
|
6
|
Glanzner WG, de Macedo MP, Gutierrez K, Bordignon V. Enhancement of Chromatin and Epigenetic Reprogramming in Porcine SCNT Embryos—Progresses and Perspectives. Front Cell Dev Biol 2022; 10:940197. [PMID: 35898400 PMCID: PMC9309298 DOI: 10.3389/fcell.2022.940197] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 06/20/2022] [Indexed: 12/12/2022] Open
Abstract
Over the last 25 years, cloned animals have been produced by transferring somatic cell nuclei into enucleated oocytes (SCNT) in more than 20 mammalian species. Among domestic animals, pigs are likely the leading species in the number of clones produced by SCNT. The greater interest in pig cloning has two main reasons, its relevance for food production and as its use as a suitable model in biomedical applications. Recognized progress in animal cloning has been attained over time, but the overall efficiency of SCNT in pigs remains very low, based on the rate of healthy, live born piglets following embryo transfer. Accumulating evidence from studies in mice and other species indicate that new strategies for promoting chromatin and epigenetic reprogramming may represent the beginning of a new era for pig cloning.
Collapse
|
7
|
Strategies to Improve the Efficiency of Somatic Cell Nuclear Transfer. Int J Mol Sci 2022; 23:ijms23041969. [PMID: 35216087 PMCID: PMC8879641 DOI: 10.3390/ijms23041969] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 01/04/2023] Open
Abstract
Mammalian oocytes can reprogram differentiated somatic cells into a totipotent state through somatic cell nuclear transfer (SCNT), which is known as cloning. Although many mammalian species have been successfully cloned, the majority of cloned embryos failed to develop to term, resulting in the overall cloning efficiency being still low. There are many factors contributing to the cloning success. Aberrant epigenetic reprogramming is a major cause for the developmental failure of cloned embryos and abnormalities in the cloned offspring. Numerous research groups attempted multiple strategies to technically improve each step of the SCNT procedure and rescue abnormal epigenetic reprogramming by modulating DNA methylation and histone modifications, overexpression or repression of embryonic-related genes, etc. Here, we review the recent approaches for technical SCNT improvement and ameliorating epigenetic modifications in donor cells, oocytes, and cloned embryos in order to enhance cloning efficiency.
Collapse
|
8
|
Zhao K, Wang M, Gao S, Chen J. Chromatin architecture reorganization during somatic cell reprogramming. Curr Opin Genet Dev 2021; 70:104-114. [PMID: 34530248 DOI: 10.1016/j.gde.2021.07.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/25/2021] [Accepted: 07/08/2021] [Indexed: 01/14/2023]
Abstract
It has been nearly 60 years since Dr John Gurdon achieved the first cloning of Xenopus by somatic cell nuclear transfer (SCNT). Later, in 2006, Takahashi and Yamanaka published their landmark study demonstrating the application of four transcription factors to induce pluripotency. These two amazing discoveries both clearly established that cell identity can be reprogrammed and that mature cells still contain the information required for lineage specification. Considering that different cell types possess identical genomes, what orchestrates reprogramming has attracted wide interest. Epigenetics, including high-level chromatin structure, might provide some answers. Benefitting from the tremendous progress in high-throughput and multi-omics techniques, we here address the roles and interactions of genome architecture, chromatin modifications, and transcription regulation during somatic cell reprogramming that were previously beyond reach. In addition, we provide perspectives on recent technical advances that might help to overcome certain barriers in the field.
Collapse
Affiliation(s)
- Kun Zhao
- Clinical and Translation Research Center of Shanghai First Maternity & Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; Frontier Science Center for Stem Cell Research, Tongji University, Shanghai 200092, China
| | - Mingzhu Wang
- Clinical and Translation Research Center of Shanghai First Maternity & Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; Frontier Science Center for Stem Cell Research, Tongji University, Shanghai 200092, China
| | - Shaorong Gao
- Clinical and Translation Research Center of Shanghai First Maternity & Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; Frontier Science Center for Stem Cell Research, Tongji University, Shanghai 200092, China.
| | - Jiayu Chen
- Clinical and Translation Research Center of Shanghai First Maternity & Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; Frontier Science Center for Stem Cell Research, Tongji University, Shanghai 200092, China.
| |
Collapse
|
9
|
Skrzyszowska M, Samiec M. Generating Cloned Goats by Somatic Cell Nuclear Transfer-Molecular Determinants and Application to Transgenics and Biomedicine. Int J Mol Sci 2021; 22:ijms22147490. [PMID: 34299109 PMCID: PMC8306346 DOI: 10.3390/ijms22147490] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/06/2021] [Accepted: 07/09/2021] [Indexed: 12/12/2022] Open
Abstract
The domestic goat (Capra aegagrus hircus), a mammalian species with high genetic merit for production of milk and meat, can be a tremendously valuable tool for transgenic research. This research is focused on the production and multiplication of genetically engineered or genome-edited cloned specimens by applying somatic cell nuclear transfer (SCNT), which is a dynamically developing assisted reproductive technology (ART). The efficiency of generating the SCNT-derived embryos, conceptuses, and progeny in goats was found to be determined by a variety of factors controlling the biological, molecular, and epigenetic events. On the one hand, the pivotal objective of our paper was to demonstrate the progress and the state-of-the-art achievements related to the innovative and highly efficient solutions used for the creation of transgenic cloned does and bucks. On the other hand, this review seeks to highlight not only current goals and obstacles but also future challenges to be faced by the approaches applied to propagate genetically modified SCNT-derived goats for the purposes of pharmacology, biomedicine, nutritional biotechnology, the agri-food industry, and modern livestock breeding.
Collapse
|
10
|
Manipulating the Epigenome in Nuclear Transfer Cloning: Where, When and How. Int J Mol Sci 2020; 22:ijms22010236. [PMID: 33379395 PMCID: PMC7794987 DOI: 10.3390/ijms22010236] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 12/24/2020] [Accepted: 12/25/2020] [Indexed: 12/20/2022] Open
Abstract
The nucleus of a differentiated cell can be reprogrammed to a totipotent state by exposure to the cytoplasm of an enucleated oocyte, and the reconstructed nuclear transfer embryo can give rise to an entire organism. Somatic cell nuclear transfer (SCNT) has important implications in animal biotechnology and provides a unique model for studying epigenetic barriers to successful nuclear reprogramming and for testing novel concepts to overcome them. While initial strategies aimed at modulating the global DNA methylation level and states of various histone protein modifications, recent studies use evidence-based approaches to influence specific epigenetic mechanisms in a targeted manner. In this review, we describe-based on the growing number of reports published during recent decades-in detail where, when, and how manipulations of the epigenome of donor cells and reconstructed SCNT embryos can be performed to optimize the process of molecular reprogramming and the outcome of nuclear transfer cloning.
Collapse
|
11
|
Wang Y, Liu Q, Kang J, Zhang Y, Quan F. Overexpression of PGC7 in donor cells maintains the DNA methylation status of imprinted genes in goat embryos derived from somatic cell nuclear transfer technology. Theriogenology 2020; 151:86-94. [PMID: 32344274 DOI: 10.1016/j.theriogenology.2020.04.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 03/26/2020] [Accepted: 04/11/2020] [Indexed: 11/27/2022]
Abstract
Abnormal methylation of imprinted genes is commonly observed in the embryos cloned by somatic cell nuclear transfer (SCNT) procedure and is one of the primary reasons for their abnormal development and high mortality. Primordial germ cell 7 (PGC7), a developmentally regulated gene highly expressed in primordial germ cells, maintains the methylation level of imprinted genes by reducing the levels of 5-hydroxy-methylcytosine(5hmC) and increasing the levels of 5-methylcytosine(5 mC) during embryonic development. In this study, we explored the methylation status of H19 differentially methylated regions (DMRs) in the organs of SCNT-cloned goat fetuses. Our results showed abnormal methylation patterns of the imprinted genes in the lungs and placenta of dead cloned goat fetuses than those in normal goat fetuses. The Igf2r DMRs were hypomethylated in the heart, liver, spleen, lungs, kidneys, and placenta of dead cloned goat fetuses compared with normal goat fetuses (P < 0.05). In addition, imprinted gene Igf2r DMRs were hypomethylated in the early-stage SCNT embryos than the IVF embryos. In contrast, imprinted gene Xist DMRs were hypermethylated in SCNT embryos than the IVF embryos. Significantly, the use of PGC7 overexpressing donor cells corrected the abnormal methylation of imprinted genes Igf2r and Xist in SCNT embryos (P < 0.05). Our results suggested that PGC7 plays a vital role in maintaining the methylation of imprinted genes during goat early embryonic development. Moreover, PGC7 overexpression in donor cells may reduce the developmental abnormalities associated with the SCNT embryos, while significantly enhancing both the pregnancy and kids born rates (P < 0.05) thereby increasing SCNT efficiency in livestock.
Collapse
Affiliation(s)
- Yufei Wang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Yangling, 712100, Shaanxi, China; College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Qingqing Liu
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Yangling, 712100, Shaanxi, China; College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Jian Kang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Yangling, 712100, Shaanxi, China; College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Yong Zhang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Yangling, 712100, Shaanxi, China; College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| | - Fusheng Quan
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Yangling, 712100, Shaanxi, China; College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| |
Collapse
|
12
|
Wang X, Qu J, Li J, He H, Liu Z, Huan Y. Epigenetic Reprogramming During Somatic Cell Nuclear Transfer: Recent Progress and Future Directions. Front Genet 2020; 11:205. [PMID: 32256519 PMCID: PMC7093498 DOI: 10.3389/fgene.2020.00205] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 02/21/2020] [Indexed: 12/21/2022] Open
Abstract
Somatic cell nuclear transfer (SCNT) has broad applications but is limited by low cloning efficiency. In this review, we mainly focus on SCNT-mediated epigenetic reprogramming in livestock and also describe mice data for reference. This review presents the factors contributing to low cloning efficiency, demonstrates that incomplete epigenetic reprogramming leads to the low developmental potential of cloned embryos, and further describes the regulation of epigenetic reprogramming by long non-coding RNAs, which is a new research perspective in the field of SCNT-mediated epigenetic reprogramming. In conclusion, this review provides new insights into the epigenetic regulatory mechanism during SCNT-mediated nuclear reprogramming, which could have great implications for improving cloning efficiency.
Collapse
Affiliation(s)
- Xiangyu Wang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| | - Jiadan Qu
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| | - Jie Li
- Department of Cadre Health Care, Qingdao Municipal Hospital, Qingdao, China
| | - Hongbin He
- College of Life Sciences, Shandong Normal University, Jinan, China
| | - Zhonghua Liu
- College of Life Sciences, Northeast Agricultural University, Harbin, China
| | - Yanjun Huan
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| |
Collapse
|
13
|
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
|
14
|
Comparison of pregnancy rates with transfer of in vivo produced embryos derived using multiple ovulation and embryo transfer (MOET) with in vitro produced embryos by somatic cell nuclear transfer (SCNT) in the dromedary camel (Camelus dromedaries). Anim Reprod Sci 2019; 209:106132. [PMID: 31514928 DOI: 10.1016/j.anireprosci.2019.106132] [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] [Received: 03/18/2019] [Revised: 05/15/2019] [Accepted: 07/18/2019] [Indexed: 11/22/2022]
Abstract
In the present study, there was comparison of pregnancy rates with transfer of in vivo-produced embryos using multiple ovulation and embryo transfer (MOET) with in vitro-produced embryos by somatic cell nuclear transfer (SCNT) in dromedary camels. In vivo-produced embryos were collected from donors after super-stimulation of follicular development on day 7 after ovulation, while in vitro-derived embryos were produced using SCNT from in vivo-matured oocytes collected from camels after follicular development super-stimulation. As a result of estrous synchronization, all recipient camels for both groups were 1 day earlier in stage of estrous cycle than developmental status of embryos at the time of transfer. The animals into which embryos were transferred were monitored at 7-day intervals after embryo transfer for signs of pregnancy based on response to presence of a male and there was ultrasonic confirmation on days 35 and 60 subsequent to day of estrus in recipient animals. A greater proportion of recipients (P < 0.05) were considered pregnant based on response to male presence when there was transfer of MOET-(76.8 ± 3.2) compared with SCNT- (26.4 ± 2.4) derived embryos on day 14. There was no difference in pregnancy losses in subsequent weeks until day 60 between groups. There were also no differences in calving rates of females in which MOET- (91.7%) and SCNT- (93.3%) derived embryos were transferred. These results indicate pregnancies at day 60 with SCNT-derived embryos are sustained for the remainder of gestation periods similar to when there was transfer of MOET-derived embryos in dromedary camels.
Collapse
|
15
|
Han C, Cui C, Xing X, Lu Z, Zhang J, Liu J, Zhang Y. Functions of intrinsic disorder in proteins involved in DNA demethylation during pre-implantation embryonic development. Int J Biol Macromol 2019; 136:962-979. [PMID: 31229544 DOI: 10.1016/j.ijbiomac.2019.06.143] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 06/18/2019] [Accepted: 06/19/2019] [Indexed: 01/21/2023]
Abstract
DNA demethylation is involved in many biological processes during pre-implantation embryonic development in mammals. To date, the complicated mechanism of DNA demethylation is still not fully understood. Ten-eleven translocation family (TET3, TET1 and TET2), thymine DNA glycosylase (TDG) and DNA methyltransferase 1 (DNMT1) are considered the major protein enzymes of DNA demethylation in pre-implantation embryos. TET3, TET1, TET2, TDG, and DNMT1 contain abundant levels of intrinsically disordered protein regions (IDPRs), which contribute to increasing the functional diversity of proteins. Thus we tried to explore the complicated DNA demethylation in pre-implantation embryos from the intrinsic disorder perspective. These five biological macromolecules all have DNA demethylation-related functional domains. They can work together to fulfill DNA demethylation in pre-implantation embryos through complex protein-protein interaction networks. Intrinsic disorder analysis results showed these proteins were partial intrinsically disordered proteins. Many identifiable disorder-based DNA-binding sites, protein-binding sites and post-translational modification sites located in the intrinsically disordered regions, and DNA demethylation deficiency point mutations in the IDPRs could significantly change the local disorder propensity of these proteins. To the best of our knowledge, this work provides a new viewpoint for studying the mechanism of DNA methylation reprogramming during mammalian pre-implantation embryonic development.
Collapse
Affiliation(s)
- Chengquan Han
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Chenchen Cui
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Xupeng Xing
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Zhenzhen Lu
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Jingcheng Zhang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Jun Liu
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Yong Zhang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China.
| |
Collapse
|