1
|
Liu Q, Sun Z, Liu Y, He X, Ren C, Wang X, Di R, Zhao Y, Zhang Z, Chu M. Whole transcriptome analysis in oviduct provides insight into microRNAs and ceRNA regulative networks that targeted reproduction of goat (Capra hircus). BMC Genomics 2025; 26:250. [PMID: 40087554 PMCID: PMC11907954 DOI: 10.1186/s12864-025-11438-8] [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: 09/18/2024] [Accepted: 03/04/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Reproduction traits are crucial for livestock breeding and represent key economic indicators in the domestic goat (Capra hircus) industry. The oviduct, a critical organ in female mammals, plays a pivotal role in several reproductive processes; however, its molecular mechanisms remain largely unknown. Non-coding RNA and mRNAs are essential regulatory elements in reproductive processes; yet their specific roles and regulatory networks in goat oviducts remain unclear. RESULTS In this study, we conducted small RNA sequencing of the oviduct in high- and low-fecundity goats during the follicular (FH and FL groups, n = 10) and luteal (LH and LL groups, n = 10) phase, profiling 20 tissue samples. Combinatorial whole-transcriptome expression profiles were applied to the same samples to uncover the competitive endogenous RNA (ceRNA) regulation network associated with goat fecundity. RT-qPCR was employed to validate the miRNA profiling results, and ceRNA regulatory networks were analyzed through luciferase assay. Gene set enrichment analysis (GSEA) confirmed that the cytokine-cytokine receptor interaction and TGF-β signaling pathway, both related to embryonic development, were enriched in DEM target genes. Additionally, miR-328-3p, a core miRNA, targets SMAD3 and BOP1, which are involved in the negative regulation of cell growth and embryonic development. TOB1 and TOB2, targeted by miR-204-3p, regulate cell proliferation via the protein kinase C-activating G-protein coupled receptor signaling pathway. Analyses of ceRNA regulatory networks revealed that LNC_005981 - miR-328-3p - SMAD3 and circ_0021923 - miR-204-3p - DOT1L may affect goats' reproduction, findings that were validated using luciferase assay. CONCLUSION Analysis of whole-transcriptome profiling of goat oviducts identified several key miRNAs and ceRNAs that may regulate oocyte maturation, embryo development, and the interactions between the oviduct and gametes/early embryos, providing insights into the molecular mechanisms of reproductive regulatory networks.
Collapse
Affiliation(s)
- Qingqing Liu
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), No. 2 Yuanmingyuan West Rd, Beijing, 100193, China
- College of Animal Science and Technology, Anhui Agricultural University, No. 130 Changjiang West Rd, Hefei, 230036, China
| | - Zhipeng Sun
- College of Animal Science and Technology, Anhui Agricultural University, No. 130 Changjiang West Rd, Hefei, 230036, China
- College of Animal Science and Technology, Chongqing Key Laboratory of Herbivore Science, Southwest University, Chongqing, 400715, China
| | - Yufang Liu
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), No. 2 Yuanmingyuan West Rd, Beijing, 100193, China
| | - Xiaoyun He
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), No. 2 Yuanmingyuan West Rd, Beijing, 100193, China
| | - Chunhuan Ren
- College of Animal Science and Technology, Anhui Agricultural University, No. 130 Changjiang West Rd, Hefei, 230036, China
| | - Xiangyu Wang
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), No. 2 Yuanmingyuan West Rd, Beijing, 100193, China
| | - Ran Di
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), No. 2 Yuanmingyuan West Rd, Beijing, 100193, China
| | - Yongju Zhao
- College of Animal Science and Technology, Chongqing Key Laboratory of Herbivore Science, Southwest University, Chongqing, 400715, China
| | - Zijun Zhang
- College of Animal Science and Technology, Anhui Agricultural University, No. 130 Changjiang West Rd, Hefei, 230036, China.
| | - Mingxing Chu
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), No. 2 Yuanmingyuan West Rd, Beijing, 100193, China.
| |
Collapse
|
2
|
Fu X, Liu Q, Song Y, Wang Y, Zhou Y, Wang S, Liu T, Zhuang F, Hu J, Sun Y. The molecule events expression of TGF-β/Smad signaling pathway in morphological and structural developmental characteristics of gonads in goose embryos. Poult Sci 2025; 104:104808. [PMID: 39823842 PMCID: PMC11786765 DOI: 10.1016/j.psj.2025.104808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 01/10/2025] [Indexed: 01/20/2025] Open
Abstract
China is the largest producer and consumer of geese with significant social and economic value in agriculture. The Jilin White Goose, known for its excellent egg-laying and reproductive characteristics, is a prominent breeding breed in the northeast of China widely used for cross-breeding.Gonad development is a complex process, which will differentiate into testes or ovaries, thus affecting sex determination. Since the level of gonadal development in geese of different sexes is also related to their productivity, the study of gonadal function in geese is of great theoretical and practical importance. In this study, we used the embryonic gonads of Jilin white geese as the research object to reveal the morphologic and histologic developmental characteristics and process of the gonads of goose embryos from the 9th day to the 28th day and to investigate the mechanisms regulating the differentiation of the gonads. The results of ALP-PAS staining by histomorphological observations showed that the primordial germ cells in the female gonads were fully differentiated at embryonic day 19 (E19) and in the male gonads at embryonic day 18 (E18).Previous studies indicate that the TGF-β /Smad signaling pathway is crucial for sex differentiation and gamete formation in animals. In our study, we quantitatively analyzed key genes and proteins in this pathway within goose embryos. We found that TGF-βRI/II receptors and Smad2/3 are involved in gonadal development early in differentiation. Smad2 protein levels peaked at E18 in male gonads and E19 in female gonads, while Smad3 peaked at E10 for both. It has been suggested that there is a novel interaction between TGF-β and Wnt/β-catenin signaling pathway. In this study, the expression of LEF1, a protein downstream of the Wnt/β-catenin signaling pathway, peaked at E19 in the female gonads and at E18 in the male gonads, and the protein expression level of β-catenin was significantly increased in the female gonads at E19 and E28 and peaked at E28 in the male gonads. β-catenin expression levels were significantly higher in the male gonads than in the other periods and reached a minimum at E28. We injected the TMAO drug, which activates the TGF-β /Smad pathway, into goose embryos using the embryo injection technique to study how it affects gene and protein expression in this signaling pathway. We found that in female gonads, the levels of Smad2/3 proteins were lower than in the control group at E18, but higher than the control group at E19. In male gonads, the results for Smad3 proteins matched those of the females, while the results for Smad2 proteins were the opposite. This shows that TMAO activation of the TGF-β /Smad signaling pathway may impact gonadal development in goose embryos. In conclusion, this study suggest that the TGF-β /Smad and Wnt signaling pathways may enhance gonadal tissue development in goose embryos. The study identified key time points in gonadogenesis, providing a new theoretical basis for further research on the molecular mechanisms of gonadal development.
Collapse
Affiliation(s)
- Xianou Fu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Qiuyuan Liu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Yupu Song
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Yudong Wang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Yuxuan Zhou
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Sihui Wang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Tuoya Liu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Fangming Zhuang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Jingtao Hu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Yongfeng Sun
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China; Key Laboratory of Animal Production, Product Quality and Security, Jilin Agricultural University, Ministry of Education, Changchun, 130118, China; Joint Laboratory of Modern Agricultural Technology International Cooperation, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China.
| |
Collapse
|
3
|
Xu X, Jiang H, Wang D, Rehman SU, Li Z, Song X, Cui K, Luo X, Yang C, Liu Q. Exploration of transcriptional regulation network between buffalo oocytes and granulosa cells and its impact on different diameter follicles. BMC Genomics 2024; 25:1004. [PMID: 39462339 PMCID: PMC11515274 DOI: 10.1186/s12864-024-10912-z] [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: 01/06/2024] [Accepted: 10/16/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND Buffalo is a globally important livestock species, but its reproductive performance is relatively low than cattles. At present, dominant follicle development specific process and mechanistic role of follicular growth related genes in water buffaloes are not well understood. Therefore, we comprehensively performed transcriptomics of granulosa cells and oocytes from different-sized follicles in water buffalo to identify key candidate genes that influence follicle development and diameter, and further explored the potential regulatory mechanisms of granulosa cells and oocytes in the process of water buffalo follicle development. RESULTS In this study, we found918 granulosa cell transcripts and 1401 oocyte transcripts were correlated in follicles of different diameters, and the expression differences were significant. Subsequent enrichment analysis of the co-expressed differentially expressed transcripts identified several genes targeted by long non-coding RNAs (lncRNAs) and associated with follicular development. Notably, the upregulation of BUB1 regulated by MSTRG.41325.4 and interactive action of SMAD2 and SMAD7 might have key regulatory role in follicular development. Additionally, we also detected key differentially expressed genes that potentially influence follicular hormone metabolism and growth, like ID2, CHRD, TGIF2 and MAD2L1, and constructed an interaction network between lncRNA transcripts and mRNAs. CONCLUSIONS In summary, this study preliminarily revealed the differences in gene expression patterns among buffalo follicles of different sizes and their potential molecular regulatory mechanisms. It provides a new perspective for exploring the mechanism of buffalo follicular dominance and improving buffalo reproductive performance.
Collapse
Affiliation(s)
- Xiaoxian Xu
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, 528225, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
| | - Hancai Jiang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
| | - Dong Wang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
| | - Saif Ur Rehman
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
| | - Zhipeng Li
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
| | - Xinhui Song
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
| | - Kuiqing Cui
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, 528225, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
| | - Xier Luo
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, 528225, China
| | - Chunyan Yang
- Buffalo Research Institute, Chinese Academy of Agricultural Sciences, Nanning, 530001, China.
| | - Qingyou Liu
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, 528225, China.
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China.
| |
Collapse
|
4
|
Latham KE. Early Cell Lineage Formation in Mammals: Complexity, Species Diversity, and Susceptibility to Disruptions Impacting Embryo Viability. Mol Reprod Dev 2024; 91:e70002. [PMID: 39463042 DOI: 10.1002/mrd.70002] [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: 08/25/2024] [Revised: 09/24/2024] [Accepted: 10/07/2024] [Indexed: 10/29/2024]
Abstract
The emergence of the earliest cell lineages in mammalian embryos is a complex process that utilizes an extensive network of chromatin regulators, transcription factors, cell polarity regulators, and cellular signaling pathways. These factors and pathways operate over a protracted period of time as embryos cleave, undergo compaction, and form blastocysts. The first cell fate specification event separates the pluripotent inner cell mass from the trophectoderm lineage. The second event separates pluripotent epiblast from hypoblast. This review summarizes over 50 years of study of these early lineage forming events, addressing the complexity of the network of interacting molecules, cellular functions and pathways that drive them, interspecies differences, and aspects of these mechanisms that likely underlie their high susceptibility to disruption by numerous environmental factors that can compromise embryo viability, such as maternal health and diet, environmental toxins, and other stressors.
Collapse
Affiliation(s)
- Keith E Latham
- Department of Animal Science, Michigan State University, Lansing, Michigan, USA
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Lansing, Michigan, USA
- Reproductive and Developmental Sciences Program, Michigan State University, Lansing, Michigan, USA
| |
Collapse
|
5
|
Deng T, Li K, Du L, Liang M, Qian L, Xue Q, Qiu S, Xu L, Zhang L, Gao X, Lan X, Li J, Gao H. Genome-Wide Gene-Environment Interaction Analysis Identifies Novel Candidate Variants for Growth Traits in Beef Cattle. Animals (Basel) 2024; 14:1695. [PMID: 38891742 PMCID: PMC11171348 DOI: 10.3390/ani14111695] [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: 04/16/2024] [Revised: 05/24/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
Complex traits are widely considered to be the result of a compound regulation of genes, environmental factors, and genotype-by-environment interaction (G × E). The inclusion of G × E in genome-wide association analyses is essential to understand animal environmental adaptations and improve the efficiency of breeding decisions. Here, we systematically investigated the G × E of growth traits (including weaning weight, yearling weight, 18-month body weight, and 24-month body weight) with environmental factors (farm and temperature) using genome-wide genotype-by-environment interaction association studies (GWEIS) with a dataset of 1350 cattle. We validated the robust estimator's effectiveness in GWEIS and detected 29 independent interacting SNPs with a significance threshold of 1.67 × 10-6, indicating that these SNPs, which do not show main effects in traditional genome-wide association studies (GWAS), may have non-additive effects across genotypes but are obliterated by environmental means. The gene-based analysis using MAGMA identified three genes that overlapped with the GEWIS results exhibiting G × E, namely SMAD2, PALMD, and MECOM. Further, the results of functional exploration in gene-set analysis revealed the bio-mechanisms of how cattle growth responds to environmental changes, such as mitotic or cytokinesis, fatty acid β-oxidation, neurotransmitter activity, gap junction, and keratan sulfate degradation. This study not only reveals novel genetic loci and underlying mechanisms influencing growth traits but also transforms our understanding of environmental adaptation in beef cattle, thereby paving the way for more targeted and efficient breeding strategies.
Collapse
Affiliation(s)
- Tianyu Deng
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (T.D.); (K.L.); (L.D.); (M.L.); (L.Q.); (Q.X.); (S.Q.); (L.X.); (L.Z.); (X.G.)
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang 712100, China;
| | - Keanning Li
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (T.D.); (K.L.); (L.D.); (M.L.); (L.Q.); (Q.X.); (S.Q.); (L.X.); (L.Z.); (X.G.)
| | - Lili Du
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (T.D.); (K.L.); (L.D.); (M.L.); (L.Q.); (Q.X.); (S.Q.); (L.X.); (L.Z.); (X.G.)
| | - Mang Liang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (T.D.); (K.L.); (L.D.); (M.L.); (L.Q.); (Q.X.); (S.Q.); (L.X.); (L.Z.); (X.G.)
| | - Li Qian
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (T.D.); (K.L.); (L.D.); (M.L.); (L.Q.); (Q.X.); (S.Q.); (L.X.); (L.Z.); (X.G.)
| | - Qingqing Xue
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (T.D.); (K.L.); (L.D.); (M.L.); (L.Q.); (Q.X.); (S.Q.); (L.X.); (L.Z.); (X.G.)
| | - Shiyuan Qiu
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (T.D.); (K.L.); (L.D.); (M.L.); (L.Q.); (Q.X.); (S.Q.); (L.X.); (L.Z.); (X.G.)
| | - Lingyang Xu
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (T.D.); (K.L.); (L.D.); (M.L.); (L.Q.); (Q.X.); (S.Q.); (L.X.); (L.Z.); (X.G.)
| | - Lupei Zhang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (T.D.); (K.L.); (L.D.); (M.L.); (L.Q.); (Q.X.); (S.Q.); (L.X.); (L.Z.); (X.G.)
| | - Xue Gao
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (T.D.); (K.L.); (L.D.); (M.L.); (L.Q.); (Q.X.); (S.Q.); (L.X.); (L.Z.); (X.G.)
| | - Xianyong Lan
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang 712100, China;
| | - Junya Li
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (T.D.); (K.L.); (L.D.); (M.L.); (L.Q.); (Q.X.); (S.Q.); (L.X.); (L.Z.); (X.G.)
| | - Huijiang Gao
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (T.D.); (K.L.); (L.D.); (M.L.); (L.Q.); (Q.X.); (S.Q.); (L.X.); (L.Z.); (X.G.)
| |
Collapse
|
6
|
Wijayanti D, Luo Y, Bai Y, Pan C, Qu L, Guo Z, Lan X. New insight into copy number variations of goat SMAD2 gene and their associations with litter size and semen quality. Theriogenology 2023; 206:114-122. [PMID: 37229957 DOI: 10.1016/j.theriogenology.2023.05.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 05/10/2023] [Accepted: 05/12/2023] [Indexed: 05/27/2023]
Abstract
Copy number variations (CNV) contribute significantly to genetic variations. Numerous studies have shown that CNV affects phenotypic traits in livestock. The SMAD family member 2 (SMAD2) is a leading candidate gene in reproduction and has a crucial effect on litter size. Additionally, SMAD2 is also required for male reproduction and influences male germ cell development. However, there are no reports on investigating the effect of CNVs in the SMAD2 gene on reproductive traits in goat. Therefore, the goal of this study was to explore associations between CNV of the SMAD2 gene and litter size and semen quality in Shaanbei white cashmere (SBWC) goats. In this study, two CNVs within the SMAD2 were identified in 352 SBWC goats (50 males and 302 females). The association analysis revealed that only CNV2 was significantly associated with female goat first-born litter size (P = 3.59 × 10-4), male semen concentration (P < 0.01), ejaculation volume, live sperm count, and sperm deformity rate (P < 0.05). In terms of phenotypic performance, the individuals with loss genotypes outperformed those with other genotypes. CNV1 and CNV2 genotype combinations containing their dominant genotypes were also associated with goat litter size (P = 1.7 × 10-5), but no differences in semen quality were found. In summary, CNV2 of the SMAD2 gene is useful for molecular marker-assisted selection breeding, as it is associated with essential goat reproductive traits.
Collapse
Affiliation(s)
- Dwi Wijayanti
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China; Department of Animal Science, Perjuangan University of Tasikmalaya, Tasikmalaya, West Java, 46115, Indonesia.
| | - Yunyun Luo
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| | - Yangyang Bai
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| | - Chuanying Pan
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| | - Lei Qu
- Shaanxi Provincial Engineering and Technology Research Center of Cashmere Goats, Yulin University, Yulin, Shaanxi, 719000, PR China; Life Science Research Center, Yulin University, Yulin, Shaanxi, 719000, PR China.
| | - Zhengang Guo
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China; Institute of Animal Husbandry and Veterinary Science of Bijie City, Guizhou, 551700, China.
| | - Xianyong Lan
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| |
Collapse
|
7
|
Liu J, Qi N, Xing W, Li M, Qian Y, Luo G, Yu S. The TGF-β/SMAD Signaling Pathway Prevents Follicular Atresia by Upregulating MORC2. Int J Mol Sci 2022; 23:ijms231810657. [PMID: 36142569 PMCID: PMC9505042 DOI: 10.3390/ijms231810657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/02/2022] [Accepted: 09/03/2022] [Indexed: 11/16/2022] Open
Abstract
In mammals, female fertility is determined by the outcome of follicular development (ovulation or atresia). The TGF-β/SMAD signaling pathway is an important regulator of this outcome. However, the molecular mechanism by which the TGF-β/SMAD signaling pathway regulates porcine follicular atresia has not been fully elucidated. Microrchidia family CW-type zinc finger 2 (MORC2) is anovel epigenetic regulatory protein widely expressed in plants, nematodes, and mammals. Our previous studies showed that MORC2 is a potential downstream target gene of the TGF-β/SMAD signaling pathway. However, the role of MORC2 in porcine follicular atresia is unknown. To investigate this, qRT-PCR, western blotting, and TdT-mediated dUTP nick-end labeling were performed. Additionally, the luciferase activity assay was conductedto confirm that the TGF-β/SMAD signaling pathway regulates MORC2. Our results demonstrate that MORC2 is animportant anti-apoptotic molecule that prevents porcine follicular atresia via a pathway involving mitochondrial apoptosis, not DNA repair. Notably, this studyrevealsthat the TGF-β/SMAD signaling pathway inhibits porcine granulosa cell apoptosis by up-regulating MORC2. The transcription factor SMAD4 regulated the expression of MORC2 by binding to its promoter. Our results will help to reveal the mechanism underlying porcine follicular atresia and improve the reproductive efficiency of sows.
Collapse
Affiliation(s)
- Jiying Liu
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212018, China
| | - Nannan Qi
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212018, China
| | - Wenwen Xing
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212018, China
| | - Mengxuan Li
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212018, China
| | - Yonghang Qian
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212018, China
| | - Gang Luo
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212018, China
| | - Shali Yu
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
- Correspondence:
| |
Collapse
|
8
|
Wooldridge LK, Keane JA, Rhoads ML, Ealy AD. Bioactive supplements influencing bovine in vitro embryo development. J Anim Sci 2022; 100:6620796. [PMID: 35772761 DOI: 10.1093/jas/skac091] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 03/21/2022] [Indexed: 12/12/2022] Open
Abstract
Ovum pickup and in vitro production (IVP) of bovine embryos are replacing traditional multiple ovulation embryo transfer (MOET) as the primary means for generating transferable embryos from genetically elite sires and dams. However, inefficiencies in the IVP process limit the opportunities to produce large numbers of transferable embryos. Also, the post-transfer competency of IVP embryos is inferior to embryos produced by artificial insemination or MOET. Numerous maternal, paternal, embryonic, and culture-related factors can have adverse effects on IVP success. This review will explore the various efforts made on describing how IVP embryo development and post-transfer competency may be improved by supplementing hormones, growth factors, cytokines, steroids and other bioactive factors found in the oviduct and uterus during early pregnancy. More than 40 of these factors, collectively termed as embryokines, are reviewed here. Several embryokines contain abilities to promote embryo development, including improving embryo survivability, improving blastomere cell numbers, and altering the distribution of blastomere cell types in blastocysts. A select few embryokines also can benefit pregnancy retention after IVP embryo transfer and improve neonatal calf health and performance, although very few embryokine-supplemented embryo transfer studies have been completed. Also, supplementing several embryokines at the same time holds promise for improving IVP embryo development and competency. However, more work is needed to explore the post-transfer consequences of adding these putative embryokines for any adverse outcomes, such as large offspring syndrome and poor postnatal health, and to specify the specific embryokine combinations that will best represent the ideal conditions found in the oviduct and uterus.
Collapse
Affiliation(s)
- Lydia K Wooldridge
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Jessica A Keane
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Michelle L Rhoads
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Alan D Ealy
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| |
Collapse
|
9
|
Daigneault BW, de Agostini Losano JD. Tributyltin chloride exposure to post-ejaculatory sperm reduces motility, mitochondrial function and subsequent embryo development. Reprod Fertil Dev 2022; 34:833-843. [PMID: 35610772 DOI: 10.1071/rd21371] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 04/26/2022] [Indexed: 11/23/2022] Open
Abstract
CONTEXT Male exposure to environmental toxicants can disrupt spermatogenesis and sperm function. However, consequences of environmentally relevant organotin exposure to post-ejaculatory mammalian spermatozoa on fertility are poorly understood. AIMS Determine the consequences of tributyltin chloride (TBT) exposure on post-ejaculatory sperm function and subsequent embryo development. METHODS Frozen-thawed bovine sperm were exposed to TBT (0.1-100nM) for 90min (acute) and 6h (short-term) followed by quantification of multiple sperm kinematics via computer aided sperm analysis. JC-1 dye was used to measure mitochondrial membrane potential. Sperm were then exposed to TBT for 90min in non-capacitating conditions, washed several times by centrifugation and applied to gamete co-incubation for in vitro embryo production to the blastocyst stage. KEY RESULTS 100nM TBT decreased total motility (88 vs 79%), progressive motility (80 vs 70%) curvilinear velocity and beat-cross frequency for 90min with similar phenotypes at 6h (P<0.05). Sperm mitochondrial membrane potential was lower in 10 and 100nM groups after 6h (P≤0.05). Embryos fertilised from TBT-exposed sperm had reduced cleavage rate (80 vs 62%) and 8-16 cell morula development (55 vs 24%) compared to development from unexposed sperm. CONCLUSIONS Exposure of post-ejaculatory mammalian sperm to TBT alters sperm function through lowered motility and mitochondrial membrane potential. Fertilisation of oocytes with TBT-exposed sperm reduces embryo development through mechanisms of paternal origin. IMPLICATIONS Acute and short-term environmental exposure of post-ejaculatory sperm to organotins and endocrine disrupting chemicals such as TBT contribute to idiopathic subfertility and early embryo loss.
Collapse
|
10
|
Liu X, Mai H, Chen P, Zhang Z, Wu T, Chen J, Sun P, Zhou C, Liang X, Huang R. Comparative analyses in transcriptome of human granulosa cells and follicular fluid micro-environment between poor ovarian responders with conventional controlled ovarian or mild ovarian stimulations. Reprod Biol Endocrinol 2022; 20:54. [PMID: 35313911 PMCID: PMC8935846 DOI: 10.1186/s12958-022-00926-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/07/2022] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Both mild and conventional controlled ovarian stimulation are the frequently used protocols for poor ovarian responders. However, there are some debates about which treatment is better. Moreover, little is known about the follicular physiology after the two ovarian stimulation protocols. This study was intended to investigate the features in granulosa cells and follicular fluid micro-environment after the two different ovarian stimulation protocols in poor responders. METHODS Granulosa cells RNA were sequenced using Illumina Hiseq technology. Specific differently expressed genes and proteins were verified by real-time quantitative PCR and Western blot analysis. Moreover, hormone and cytokine concentrations in the follicular fluid were measured by electrochemiluminescence immunoassay and enzyme-linked immunoabsorbent assay. The correlation between the results of molecular experiments and the laboratory outcomes were analyzed by Spearman correlation analysis. RESULTS The differentially expressed genes between the two groups were involved in 4 signaling pathways related to the follicular development; three proteins pertinent to the TGF-β signaling pathway were expressed differently in granulosa cells between the two, and the constituents in the follicular fluid were also different. Further, a correlation between the TGF-β signaling pathway and the good-quality embryo was observed. CONCLUSIONS The present study made a comparison for the first time in the transcriptome of human granulosa cells and the follicular fluid micro-environment between poor responders with the conventional controlled ovarian stimulation or the mild ovarian stimulation, showing that the TGF-β signaling pathway may correlate with the good-quality of embryos in the mild group, which may be instrumental to the choice of optimal management for IVF patients.
Collapse
Affiliation(s)
- Xiaoping Liu
- Reproductive Medicine Research Center, the Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Huisi Mai
- Reproductive Medicine Research Center, the Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Panyu Chen
- Reproductive Medicine Research Center, the Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zhiqiang Zhang
- Reproductive Medicine Research Center, the Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Taibao Wu
- Reproductive Medicine Research Center, the Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jianhui Chen
- Reproductive Medicine Research Center, the Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Peng Sun
- Reproductive Medicine Research Center, the Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Chuanchuan Zhou
- Reproductive Medicine Research Center, the Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiaoyan Liang
- Reproductive Medicine Research Center, the Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Rui Huang
- Reproductive Medicine Research Center, the Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
11
|
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
|
12
|
Identification and Expression Characterization of the Smad3 Gene and SNPs Associated with Growth Traits in the Hard Clam (Meretrix meretrix). FISHES 2021. [DOI: 10.3390/fishes6040083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
It has been demonstrated that the sekelsky mothers against decapentaplegic homolog 3 (Smad3) plays an important role in the growth and development of vertebrates. However, little is known about the association between the Smad3 gene and the growth traits of mollusks. In this study, Smad3 from the hard clam Meretrix meretrix (Mm-Smad3) was cloned, characterized, and screened for growth-related single nucleotide polymorphisms (SNPs) in its exons. The full-length cDNA of Mm-Smad3 was 1938 bp, encoding a protein with 428 amino acid residues. The protein sequence included an MH1 (27–135 aa) and MH2 domain (233–404 aa). Promoter analysis showed that the promoter sequence of Mm-Smad3 was 2548 bp, and the binding sites of Pit-1a, Antp, Hb, and other transcription factors are related to the growth and development of hard clams. The phylogenetic tree was divided into two major clusters, including mollusks and vertebrate. The expression level of Mm-Smad3 was predominantly detected in the mantle and foot, while extremely less expression was observed in the digestive gland. The low expression level of Mm-Smad3 was detected at the stages of unfertilized mature eggs, fertilized eggs, four-cell embryos, blastula, gastrulae, trochophore, and D-shaped larvae, whereas an opposite trend was observed regarding the highest expression at the umbo larvae stage (p < 0.05). In the mantle repair experiment, the time-course expression profiles showed that compared to the expression level at 0 h, Mm-Smad3 significantly decreased at 6 h (p < 0.05) but increased at 12 and 48 h. Further, the association analysis identified 11 SNPs in the exons of Mm-Smad3, of which three loci (c.597 C > T, c.660 C > T, c.792 A > T) were significantly related to the growth traits of clam (p < 0.05). Overall, our findings indicated that Mm-Smad3 is a growth-related gene and the detected SNP sites provide growth-related markers for molecular marker-assisted breeding of this species.
Collapse
|
13
|
Rajput SK, Yang C, Ashry M, Folger JK, Knott JG, Smith GW. Role of bone morphogenetic protein signaling in bovine early embryonic development and stage specific embryotropic actions of follistatin†. Biol Reprod 2021; 102:795-805. [PMID: 31965149 DOI: 10.1093/biolre/ioz235] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 11/21/2019] [Accepted: 01/14/2020] [Indexed: 01/10/2023] Open
Abstract
Characterization of the molecular factors regulating early embryonic development and their functional mechanisms is critical for understanding the causes of early pregnancy loss in monotocous species (cattle, human). We previously characterized a stage specific functional role of follistatin, a TGF-beta superfamily binding protein, in promoting early embryonic development in cattle. The mechanism by which follistatin mediates this embryotropic effect is not precisely known as follistatin actions in cattle embryos are independent of its classically known activin inhibition activity. Apart from activin, follistatin is known to bind and modulate the activity of the bone morphogenetic proteins (BMPs), which signal through SMAD1/5 pathway and regulate several aspects of early embryogenesis in other mammalian species. Present study was designed to characterize the activity and functional requirement of BMP signaling during bovine early embryonic development and to investigate if follistatin involves BMP signaling for its stage specific embryotropic actions. Immunostaining and western blot analysis demonstrated that SMAD1/5 signaling is activated after embryonic genome activation in bovine embryos. However, days 1-3 follistatin treatment reduced the abundance of phosphorylated SMAD1/5 in cultured embryos. Inhibition of active SMAD1/5 signaling (8-16 cell to blastocyst) using pharmacological inhibitors and/or lentiviral-mediated inhibitory SMAD6 overexpression showed that SMAD1/5 signaling is required for blastocyst production, first cell lineage determination as well as mRNA and protein regulation of TE (CDX2) cell markers. SMAD1/5 signaling was also found to be essential for embryotropic actions of follistatin during days 4-7 but not days 1-3 of embryo development suggesting a role for follistatin in regulation of SMAD1/5 signaling in bovine embryos.
Collapse
Affiliation(s)
- Sandeep K Rajput
- Laboratory of Mammalian Reproductive Biology and Genomics, Michigan State University, East Lansing, Michigan, USA.,Colorado Center for Reproductive Medicine (CCRM), Lone Tree, CO 80124, USA
| | - Chunyan Yang
- Guangxi Buffalo Research Institute, Chinese Academy of Agricultural Science, Nanning, P.R. China
| | - Mohamed Ashry
- Laboratory of Mammalian Reproductive Biology and Genomics, Michigan State University, East Lansing, Michigan, USA.,Department of Theriogenology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt and
| | - Joseph K Folger
- Laboratory of Mammalian Reproductive Biology and Genomics, Michigan State University, East Lansing, Michigan, USA
| | - Jason G Knott
- Developmental Epigenetics Laboratory, Michigan State University, East Lansing, Michigan, USA
| | - George W Smith
- Laboratory of Mammalian Reproductive Biology and Genomics, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
14
|
Llobat L. Pluripotency and Growth Factors in Early Embryonic Development of Mammals: A Comparative Approach. Vet Sci 2021; 8:vetsci8050078. [PMID: 34064445 PMCID: PMC8147802 DOI: 10.3390/vetsci8050078] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 04/27/2021] [Accepted: 05/02/2021] [Indexed: 12/24/2022] Open
Abstract
The regulation of early events in mammalian embryonic development is a complex process. In the early stages, pluripotency, cellular differentiation, and growth should occur at specific times and these events are regulated by different genes that are expressed at specific times and locations. The genes related to pluripotency and cellular differentiation, and growth factors that determine successful embryonic development are different (or differentially expressed) among mammalian species. Some genes are fundamental for controlling pluripotency in some species but less fundamental in others, for example, Oct4 is particularly relevant in bovine early embryonic development, whereas Oct4 inhibition does not affect ovine early embryonic development. In addition, some mechanisms that regulate cellular differentiation do not seem to be clear or evolutionarily conserved. After cellular differentiation, growth factors are relevant in early development, and their effects also differ among species, for example, insulin-like growth factor improves the blastocyst development rate in some species but does not have the same effect in mice. Some growth factors influence genes related to pluripotency, and therefore, their role in early embryo development is not limited to cell growth but could also involve the earliest stages of development. In this review, we summarize the differences among mammalian species regarding the regulation of pluripotency, cellular differentiation, and growth factors in the early stages of embryonic development.
Collapse
Affiliation(s)
- Lola Llobat
- Research Group Microbiological Agents Associated with Animal Reproduction (PROVAGINBIO), Department of Animal Production and Health, Veterinary Public Health and Food Science and Technology (PASAPTA) Facultad de Veterinaria, Universidad Cardenal Herrera-CEU, CEU Universities, 46113 Valencia, Spain
| |
Collapse
|
15
|
Shi Y, Zhao P, Dang Y, Li S, Luo L, Hu B, Wang S, Wang H, Zhang K. Functional roles of the chromatin remodeler SMARCA5 in mouse and bovine preimplantation embryos†. Biol Reprod 2021; 105:359-370. [PMID: 33899080 DOI: 10.1093/biolre/ioab081] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/07/2021] [Accepted: 04/18/2021] [Indexed: 12/30/2022] Open
Abstract
Upon fertilization, extensive chromatin reprogramming occurs during preimplantation development. Growing evidence reveals species-dependent regulations of this process in mammals. ATP-dependent chromatin remodeling factor SMARCA5 (also known as SNF2H) is required for peri-implantation development in mice. However, the specific functional role of SMARCA5 in preimplantation development and if it is conserved among species remain unclear. Herein, comparative analysis of public RNA-seq datasets reveals that SMARCA5 is universally expressed during oocyte maturation and preimplantation development in mice, cattle, humans, and pigs with species-specific patterns. Immunostaining analysis further describes the temporal and spatial changes of SMARCA5 in both mouse and bovine models. siRNA-mediated SMARCA5 depletion reduces the developmental capability and compromises the specification and differentiation of inner cell mass in mouse preimplantation embryos. Indeed, OCT4 is not restricted into the inner cell mass and the formation of epiblast and primitive endoderm disturbed with reduced NANOG and SOX17 in SMARCA5-deficient blastocysts. RNA-seq analysis shows SMARCA5 depletion causes limited effects on the transcriptomics at the morula stage, however, dysregulates 402 genes, including genes involved in transcription regulation and cell proliferation at the blastocyst stage in mice. By comparison, SMARCA5 depletion does not affect the development through the blastocyst stage but significantly compromises the blastocyst quality in cattle. Primitive endoderm formation is greatly disrupted with reduced GATA6 in bovine blastocysts. Overall, our studies demonstrate the importance of SMARCA5 in fostering the preimplantation development in mice and cattle while there are species-specific effects.
Collapse
Affiliation(s)
- Yan Shi
- Laboratory of Mammalian Molecular Embryology, College of Animal Sciences, Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Panpan Zhao
- Laboratory of Mammalian Molecular Embryology, College of Animal Sciences, Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Yanna Dang
- Laboratory of Mammalian Molecular Embryology, College of Animal Sciences, Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Shuang Li
- Laboratory of Mammalian Molecular Embryology, College of Animal Sciences, Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Lei Luo
- Laboratory of Mammalian Molecular Embryology, College of Animal Sciences, Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Bingjie Hu
- Laboratory of Mammalian Molecular Embryology, College of Animal Sciences, Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Shaohua Wang
- Laboratory of Mammalian Molecular Embryology, College of Animal Sciences, Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Huanan Wang
- Laboratory of Mammalian Molecular Embryology, College of Animal Sciences, Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Kun Zhang
- Laboratory of Mammalian Molecular Embryology, College of Animal Sciences, Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| |
Collapse
|
16
|
Li S, Shi Y, Dang Y, Luo L, Hu B, Wang S, Wang H, Zhang K. NOTCH signaling pathway is required for bovine early embryonic development†. Biol Reprod 2021; 105:332-344. [PMID: 33763686 DOI: 10.1093/biolre/ioab056] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/04/2020] [Accepted: 03/17/2021] [Indexed: 12/28/2022] Open
Abstract
The NOTCH signaling pathway plays an important role in regulating various biological processes, including lineage specification and apoptosis. Multiple components of the NOTCH pathway have been identified in mammalian preimplantation embryos. However, the precise role of the NOTCH pathway in early embryonic development is poorly understood, especially in large animals. Here, we show that the expression of genes encoding key transcripts of the NOTCH pathway is dynamic throughout early embryonic development. We also confirm the presence of active NOTCH1 and RBPJ. By using pharmacological and RNA interference tools, we demonstrate that the NOTCH pathway is required for the proper development of bovine early embryos. This functional consequence could be partly attributed to the major transcriptional mediator, Recombination Signal Binding Protein For Immunoglobulin Kappa J Region (RBPJ), whose deficiency also compromised the embryo quality. Indeed, both NOTCH1 and RBPJ knockdown cause a significant increase of histone H3 serine 10 phosphorylation (pH3S10, a mitosis marker) positive blastomeres, suggesting a cell cycle arrest at mitosis. Importantly, RNA sequencing analyses reveal that either NOTCH1 or RBPJ depletion triggers a reduction in H1FOO that encodes the oocyte-specific linker histone H1 variant. Interestingly, depleting H1FOO results in detrimental effects on the developmental competence of early embryos, similar with NOTCH1 inhibition. Overall, our results reveal a crucial role for NOTCH pathway in regulating bovine preimplantation development, likely by controlling cell proliferation and maintaining H1FOO expression.
Collapse
Affiliation(s)
- Shuang Li
- Laboratory of Mammalian Molecular Embryology, College of Animal Sciences, Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yan Shi
- Laboratory of Mammalian Molecular Embryology, College of Animal Sciences, Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yanna Dang
- Laboratory of Mammalian Molecular Embryology, College of Animal Sciences, Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Lei Luo
- Laboratory of Mammalian Molecular Embryology, College of Animal Sciences, Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Bingjie Hu
- Laboratory of Mammalian Molecular Embryology, College of Animal Sciences, Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Shaohua Wang
- Laboratory of Mammalian Molecular Embryology, College of Animal Sciences, Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Huanan Wang
- Laboratory of Mammalian Molecular Embryology, College of Animal Sciences, Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Kun Zhang
- Laboratory of Mammalian Molecular Embryology, College of Animal Sciences, Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
17
|
Akizawa H, Yanagawa Y, Nagano M, Bai H, Takahashi M, Kawahara M. Significance of CCN2 expression in bovine preimplantation development. Anim Sci J 2018; 90:49-54. [PMID: 30358017 DOI: 10.1111/asj.13126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 08/10/2018] [Accepted: 09/27/2018] [Indexed: 11/30/2022]
Abstract
In mammalian preimplantation development, the first cell lineage segregation occurs during the blastocyst stage, when the inner cell mass and trophectoderm (TE) differentiate. Species-specific analyses are essential to elucidate the molecular mechanisms that underlie this process, since they differ between various species. We previously showed that the reciprocal regulation of CCN2 and TEAD4 is required for proper TE differentiation in bovine blastocysts; however, the function of CCN2 during early embryogenesis has remained otherwise elusive. The present study assessed the spatiotemporal expression dynamics of CCN2 in bovine embryos, and evaluated how changes to CCN2 expression (using a CCN2 knockdown (KD) blastocyst model) regulate the expression of pluripotency-related genes such as OCT4 and NANOG. The conducted quantitative PCR analysis revealed that CCN2 mRNA was expressed in bovine oocytes (at the metaphase stage of their second meiosis) and embryos. Similarly, immunostaining detected both cytoplasmic and nuclear CCN2 at all analyzed oocyte and embryonic stages. Finally, both OCT4 and NANOG expression levels were shown to be significantly reduced in CCN2 KD blastocysts. Together, these results demonstrate that bovine CCN2 exhibits unique expression patterns during preimplantation development, and is required for the proper expression of key regulatory genes in bovine blastocysts.
Collapse
Affiliation(s)
- Hiroki Akizawa
- Laboratory of Animal Breeding and Reproduction, Research Faculty of Agriculture, Hokkaido University, Sapporo, Japan
| | - Yojiro Yanagawa
- Laboratory of Theriogenology, Department of Veterinary Clinical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Masashi Nagano
- Laboratory of Theriogenology, Department of Veterinary Clinical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Hanako Bai
- Laboratory of Animal Breeding and Reproduction, Research Faculty of Agriculture, Hokkaido University, Sapporo, Japan
| | - Masashi Takahashi
- Laboratory of Animal Breeding and Reproduction, Research Faculty of Agriculture, Hokkaido University, Sapporo, Japan
| | - Manabu Kawahara
- Laboratory of Animal Breeding and Reproduction, Research Faculty of Agriculture, Hokkaido University, Sapporo, Japan
| |
Collapse
|
18
|
Fonseca PADS, Id-Lahoucine S, Reverter A, Medrano JF, Fortes MS, Casellas J, Miglior F, Brito L, Carvalho MRS, Schenkel FS, Nguyen LT, Porto-Neto LR, Thomas MG, Cánovas A. Combining multi-OMICs information to identify key-regulator genes for pleiotropic effect on fertility and production traits in beef cattle. PLoS One 2018; 13:e0205295. [PMID: 30335783 PMCID: PMC6193631 DOI: 10.1371/journal.pone.0205295] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 09/21/2018] [Indexed: 12/21/2022] Open
Abstract
The identification of biological processes related to the regulation of complex traits is a difficult task. Commonly, complex traits are regulated through a multitude of genes contributing each to a small part of the total genetic variance. Additionally, some loci can simultaneously regulate several complex traits, a phenomenon defined as pleiotropy. The lack of understanding on the biological processes responsible for the regulation of these traits results in the decrease of selection efficiency and the selection of undesirable hitchhiking effects. The identification of pleiotropic key-regulator genes can assist in developing important tools for investigating biological processes underlying complex traits. A multi-breed and multi-OMICs approach was applied to study the pleiotropic effects of key-regulator genes using three independent beef cattle populations evaluated for fertility traits. A pleiotropic map for 32 traits related to growth, feed efficiency, carcass and meat quality, and reproduction was used to identify genes shared among the different populations and breeds in pleiotropic regions. Furthermore, data-mining analyses were performed using the Cattle QTL database (CattleQTLdb) to identify the QTL category annotated in the regions around the genes shared among breeds. This approach allowed the identification of a main gene network (composed of 38 genes) shared among breeds. This gene network was significantly associated with thyroid activity, among other biological processes, and displayed a high regulatory potential. In addition, it was possible to identify genes with pleiotropic effects related to crucial biological processes that regulate economically relevant traits associated with fertility, production and health, such as MYC, PPARG, GSK3B, TG and IYD genes. These genes will be further investigated to better understand the biological processes involved in the expression of complex traits and assist in the identification of functional variants associated with undesirable phenotypes, such as decreased fertility, poor feed efficiency and negative energetic balance.
Collapse
Affiliation(s)
- Pablo Augusto de Souza Fonseca
- University of Guelph, Department of Animal Biosciences, Centre for Genetic Improvement of Livestock, Guelph, Ontario, Canada
- Universidade Federal de Minas Gerais, Departamento de Biologia Geral, Belo Horizonte, Minas Gerais, Brazil
| | - Samir Id-Lahoucine
- University of Guelph, Department of Animal Biosciences, Centre for Genetic Improvement of Livestock, Guelph, Ontario, Canada
| | - Antonio Reverter
- CSIRO Agriculture and Food, Queensland Bioscience Precinct, Brisbane, Queensland, Australia
| | - Juan F. Medrano
- University of California-Davis, Department of Animal Science, Davis, California, United States of America
| | - Marina S. Fortes
- The University of Queensland, School of Chemistry and Molecular Biosciences, Brisbane, Queensland, Australia
| | - Joaquim Casellas
- Universitat Autònoma de Barcelona, Departament de Ciència Animal i dels Aliments, Barcelona, Bellaterra, Barcelona, Spain
| | - Filippo Miglior
- University of Guelph, Department of Animal Biosciences, Centre for Genetic Improvement of Livestock, Guelph, Ontario, Canada
- Canadian Dairy Network, Guelph, Ontario, Canada
| | - Luiz Brito
- University of Guelph, Department of Animal Biosciences, Centre for Genetic Improvement of Livestock, Guelph, Ontario, Canada
| | - Maria Raquel S. Carvalho
- Universidade Federal de Minas Gerais, Departamento de Biologia Geral, Belo Horizonte, Minas Gerais, Brazil
| | - Flávio S. Schenkel
- University of Guelph, Department of Animal Biosciences, Centre for Genetic Improvement of Livestock, Guelph, Ontario, Canada
| | - Loan T. Nguyen
- The University of Queensland, School of Chemistry and Molecular Biosciences, Brisbane, Queensland, Australia
| | - Laercio R. Porto-Neto
- CSIRO Agriculture and Food, Queensland Bioscience Precinct, Brisbane, Queensland, Australia
| | - Milton G. Thomas
- Colorado State University, Department of Animal Science, Fort-Colins, Colorado, United States of America
| | - Angela Cánovas
- University of Guelph, Department of Animal Biosciences, Centre for Genetic Improvement of Livestock, Guelph, Ontario, Canada
- * E-mail:
| |
Collapse
|
19
|
Godini R, Fallahi H. Dynamics changes in the transcription factors during early human embryonic development. J Cell Physiol 2018; 234:6489-6502. [PMID: 30246428 DOI: 10.1002/jcp.27386] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 08/17/2018] [Indexed: 12/30/2022]
Abstract
Development of an embryo from a single cell, zygote, to multicellular morulae requires activation of hundreds of genes that were mostly inactivated before fertilization. Inevitably, transcription factors (TFs) would be involved in modulating the drastic changes in gene expression pattern observed at all preimplantation stages. Despite many ongoing efforts to uncover the role of TFs at the early stages of embryogenesis, still many unanswered questions remained that need to be explored. This could be done by studying the expression pattern of multiple genes obtained by high-throughput techniques. In the current study, we have identified a set of TFs that are involved in the progression of the zygote to blastocyst. Global gene expression patterns of consecutive stages were compared and differences documented. Expectedly, at the early stages of development, only a few sets of TFs differentially expressed while at the later stages hundreds of TFs appear to be upregulated. Interestingly, the expression levels of many TFs show an oscillation pattern during development indicating a need for their precise expression. A significant shift in gene expression was observed during the transition from four- to eight-cell stages, an indication of zygote genome activation. Additionally, we have found 11 TFs that were common in all stages including ATF3, EN1, IFI16, IKZF3, KLF3, NPAS3, NR2F2, RUNX1, SOX2, ZBTB20, and ZSCAN4. However, their expression patterns did not follow similar trends in the steps studied. Besides, our findings showed that both upregulation and active downregulation of the TFs expression is required for successful embryogenesis. Furthermore, our detailed network analysis identified the hub TFs for each transition. We found that HNF4A, FOXA2, and EP300 are the three most important elements for the first division of zygote.
Collapse
Affiliation(s)
- Rasoul Godini
- Department of Biology, School of Sciences, Razi University, Kermanshah, Iran
| | - Hossein Fallahi
- Department of Biology, School of Sciences, Razi University, Kermanshah, Iran
| |
Collapse
|
20
|
Follistatin supplementation during in vitro embryo culture improves developmental competence of bovine embryos produced using sex-sorted semen. Reprod Biol 2018; 18:267-273. [PMID: 30196810 PMCID: PMC7747478 DOI: 10.1016/j.repbio.2018.06.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 05/29/2018] [Accepted: 06/22/2018] [Indexed: 11/20/2022]
Abstract
Using sex-sorted semen to produce offspring of desired sex is associated with reduced developmental competence in vitro and lower fertility rates in vivo. The objectives of the present study were to investigate the effects of exogenous follistatin supplementation on the developmental competence of bovine embryos produced with sex-sorted semen and possible link between TGF-β regulated pathways and embryotrophic actions of follistatin. Effects of follistatin on expression of cell lineage markers (CDX2 and Nanog) and downstream targets of SMAD signaling (CTGF, ID1, ID2 and ID3) and AKT phosphorylation were investigated. Follistatin was supplemented during the initial 72 h of embryo culture. Exogenous follistatin restored the in vitro developmental competence of embryos produced with sex-sorted semen to the levels of control embryos produced with unsorted semen, and comparable results were obtained using sorted semen from three different bulls. The mRNA abundance for SMAD signaling downstream target genes, CTGF (SMAD 2/3 pathway) and ID2 (SMAD 1/5 pathway), was lower in blastocysts produced using sex-sorted versus unsorted semen, but mRNA levels for CDX2, NANOG, ID1 and ID3 were similar in both groups. Follistatin supplementation restored CTGF and ID2 mRNA in blastocysts produced using sex-sorted semen to levels of control embryos. Moreover, levels of phosphorylated (p)AKT (Ser-473 and Thr-308) were similar in embryos derived from sex-sorted and unsorted semen, but follistatin treatment increased pAKT levels in both groups. Taken together, results demonstrated that follistatin improves in vitro development of embryos produced with sex-sorted semen and such effects are associated with enhanced indices of SMAD signaling.
Collapse
|
21
|
Effect of exogenous transforming growth factor β1 (TGF-β1) on early bovine embryo development. ZYGOTE 2018; 26:232-241. [DOI: 10.1017/s096719941800014x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
SummaryDuring preimplantation development, embryos are exposed and have the capacity to respond to different growth factors present in the maternal environment. Among these factors, transforming growth factor β1 (TGF-β1) is a well known modulator of embryonic growth and development. However, its action during the first stages of development, when the embryo transits through the oviduct, has not been yet elucidated. The objective of the present study was to examine the effect of early exposure to exogenous TGF-β1 on embryo development and expression of pluripotency (OCT4, NANOG) and DNA methylation (DNMT1, DNMT3A, DNMT3B) genes in bovine embryos produced in vitro. First, gene expression analysis of TGF-β receptors confirmed a stage-specific expression pattern, showing greater mRNA abundance of TGFBR1 and TGFBR2 from the 2- to the 8-cell stage, before embryonic genome activation. Second, embryo culture for the first 48 h in serum-free CR1aa medium supplemented with 50 or 100 ng/ml recombinant TGF-β1 did not affect the cleavage and blastocyst rate (days 7 and 8). However, RT-qPCR analysis showed a significant increase in the relative abundance of NANOG and DNMT3A in the 8-cell stage embryos and expanded blastocysts (day 8) derived from TGF-β1 treated embryos. These results suggest an early action of exogenous TGF-β1 on the bovine embryo, highlighting the importance to provide a more comprehensive understanding of the role of TGF-β signalling during early embryogenesis.
Collapse
|
22
|
Daigneault BW, Rajput S, Smith GW, Ross PJ. Embryonic POU5F1 is Required for Expanded Bovine Blastocyst Formation. Sci Rep 2018; 8:7753. [PMID: 29773834 PMCID: PMC5958112 DOI: 10.1038/s41598-018-25964-x] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 04/27/2018] [Indexed: 01/08/2023] Open
Abstract
POU5F1 is a transcription factor and master regulator of cell pluripotency with indispensable roles in early embryo development and cell lineage specification. The role of embryonic POU5F1 in blastocyst formation and cell lineage specification differs between mammalian species but remains completely unknown in cattle. The CRISPR/Cas9 system was utilized for targeted disruption of the POU5F1 gene by direct injection into zygotes. Disruption of the bovine POU5F1 locus prevented blastocyst formation and was associated with embryonic arrest at the morula stage. POU5F1 knockout morulas developed at a similar rate as control embryos and presented a similar number of blastomeres by day 5 of development. Initiation of SOX2 expression by day 5 of development was not affected by lack of POU5F1. On the other hand, CDX2 expression was aberrant in embryos lacking POU5F1. Notably, the phenotype observed in bovine POU5F1 knockout embryos reveals conserved functions associated with loss of human embryonic POU5F1 that differ from Pou5f1- null mice. The similarity observed in transcriptional regulation of early embryo development between cattle and humans combined with highly efficient gene editing techniques make the bovine a valuable model for human embryo biology with expanded applications in agriculture and assisted reproductive technologies.
Collapse
Affiliation(s)
- Bradford W Daigneault
- Department of Animal Science, Michigan State University, East-Lansing, Michigan, USA
| | - Sandeep Rajput
- Department of Animal Science, Michigan State University, East-Lansing, Michigan, USA
| | - George W Smith
- Department of Animal Science, Michigan State University, East-Lansing, Michigan, USA
| | - Pablo J Ross
- Department of Animal Science, University of California Davis, Davis, CA, USA.
| |
Collapse
|
23
|
Wang Y, Ding X, Tan Z, Xing K, Yang T, Wang Y, Sun D, Wang C. Genome-wide association study for reproductive traits in a Large White pig population. Anim Genet 2018; 49:127-131. [PMID: 29411893 PMCID: PMC5873431 DOI: 10.1111/age.12638] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/27/2017] [Indexed: 11/27/2022]
Abstract
Using the PorcineSNP80 BeadChip, we performed a genome‐wide association study for seven reproductive traits, including total number born, number born alive, litter birth weight, average birth weight, gestation length, age at first service and age at first farrowing, in a population of 1207 Large White pigs. In total, we detected 12 genome‐wide significant and 41 suggestive significant SNPs associated with six reproductive traits. The proportion of phenotypic variance explained by all significant SNPs for each trait ranged from 4.46% (number born alive) to 11.49% (gestation length). Among them, 29 significant SNPs were located within known QTL regions for swine reproductive traits, such as corpus luteum number, stillborn number and litter size, of which one QTL region associated with litter size contained the ALGA0098819 SNP for total number born. Subsequently, we found that 376 functional genes contained or were near these significant SNPs. Of these, 14 genes—BHLHA15, OCM2, IL1B2, GCK, SMAD2, HABP2, PAQR5, GRB10, PRELID2, DMKN, GPI, GPIHBP1, ADCY2 and ACVR2B—were considered important candidates for swine reproductive traits based on their critical roles in embryonic development, energy metabolism and growth development. Our findings contribute to the understanding of the genetic mechanisms for reproductive traits and could have a positive effect on pig breeding programs.
Collapse
Affiliation(s)
- Y Wang
- Key Laboratory of Animal Genetics and Breeding of Ministry of Agriculture, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100094, China
| | - X Ding
- Key Laboratory of Animal Genetics and Breeding of Ministry of Agriculture, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100094, China
| | - Z Tan
- Key Laboratory of Animal Genetics and Breeding of Ministry of Agriculture, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100094, China
| | - K Xing
- Key Laboratory of Animal Genetics and Breeding of Ministry of Agriculture, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100094, China
| | - T Yang
- Key Laboratory of Animal Genetics and Breeding of Ministry of Agriculture, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100094, China
| | - Y Wang
- Beijing Shunxin Agriculture Co., Ltd., Beijing, 101300, China
| | - D Sun
- Key Laboratory of Animal Genetics and Breeding of Ministry of Agriculture, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100094, China
| | - C Wang
- Key Laboratory of Animal Genetics and Breeding of Ministry of Agriculture, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100094, China
| |
Collapse
|
24
|
Zhang K, Wang H, Rajput SK, Folger JK, Smith GW. Characterization of H3.3 and HIRA expression and function in bovine early embryos. Mol Reprod Dev 2018; 85:106-116. [PMID: 29232016 DOI: 10.1002/mrd.22939] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 12/01/2017] [Indexed: 01/20/2023]
Abstract
Histone variant H3.3 is encoded by two distinct genes, H3F3A and H3F3B, that are closely associated with actively transcribed genes. H3.3 replacement is continuous and essential for maintaining correct chromatin structure during mouse oogenesis. Upon fertilization, H3.3 is incorporated to parental chromatin, and is required for blastocyst formation in mice. The H3.3 exchange process is facilitated by the chaperone HIRA, particularly during zygote development. We previously demonstrated that H3.3 is required for bovine early embryonic development; here, we explored the mechanisms of its functional requirement. H3F3A mRNA abundance is stable whereas H3F3B and HIRA mRNA are relatively dynamic during early embryonic development. H3F3B mRNA quantity is also considerably higher than H3F3A. Immunofluorescence analysis revealed an even distribution of H3.3 between paternal and maternal pronuclei in zygotes, and subsequent stage-specific localization of H3.3 in early bovine embryos. Knockdown of H3.3 by targeting both H3F3A and H3F3B dramatically decreased the expression of NANOG (a pluripotency marker) and CTGF (Connective tissue growth factor; a trophectoderm marker) in bovine blastocysts. Additionally, we noted that Histone H3 lysine 36 dimethylation and linker Histone H1 abundance is reduced in H3.3-deficient embryos, which was similar to effects following knockdown of CHD1 (Chromodomain helicase DNA-binding protein 1). By contrast, no difference was observed in the abundance of Histone H3 lysine 4 trimethylation, Histone H3 lysine 9 dimethylation, or Splicing factor 3 B1. Collectively, these results established that H3.3 is required for correct epigenetic modifications and H1 deposition, dysregulation of which likely mediate the poor development in H3.3-deficient embryos.
Collapse
Affiliation(s)
- Kun Zhang
- Laboratory of Mammalian Molecular Embryology, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Institute of Dairy Sciences, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Laboratory of Mammalian Reproductive Biology and Genomics, Michigan State University, East Lansing, Michigan
- Department of Animal Science, Michigan State University, East Lansing, Michigan
| | - Han Wang
- Laboratory of Mammalian Molecular Embryology, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Sandeep K Rajput
- Laboratory of Mammalian Reproductive Biology and Genomics, Michigan State University, East Lansing, Michigan
- Department of Animal Science, Michigan State University, East Lansing, Michigan
| | - Joseph K Folger
- Laboratory of Mammalian Reproductive Biology and Genomics, Michigan State University, East Lansing, Michigan
- Department of Animal Science, Michigan State University, East Lansing, Michigan
| | - George W Smith
- Laboratory of Mammalian Reproductive Biology and Genomics, Michigan State University, East Lansing, Michigan
- Department of Animal Science, Michigan State University, East Lansing, Michigan
| |
Collapse
|
25
|
Ashry M, Rajput SK, Folger JK, Knott JG, Hemeida NA, Kandil OM, Ragab RS, Smith GW. Functional role of AKT signaling in bovine early embryonic development: potential link to embryotrophic actions of follistatin. Reprod Biol Endocrinol 2018; 16:1. [PMID: 29310676 PMCID: PMC5759257 DOI: 10.1186/s12958-017-0318-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 12/25/2017] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND TGF-β signaling pathways regulate several crucial processes in female reproduction. AKT is a non-SMAD signaling pathway regulated by TGF-β ligands essential for oocyte maturation and early embryonic development in the mouse, but its regulatory role in bovine early embryonic development is not well established. Previously, we demonstrated a stimulatory role for follistatin (a binding protein for specific members of TGF-β superfamily) in early bovine embryonic development. The objectives of the present studies were to determine the functional role of AKT signaling in bovine early embryonic development and embryotrophic actions of follistatin. METHODS We used AKT inhibitors III and IV as pharmacological inhibitors of AKT signaling pathway during the first 72 h of in vitro embryo culture. Effects of AKT inhibition on early embryonic development and AKT phosphorylation were investigated in the presence or absence of exogenous follistatin. RESULTS Pharmacological inhibition of AKT signaling resulted in a significant reduction in early embryo cleavage, and development to the 8- to 16-cell and blastocyst stages (d7). Treatment with exogenous follistatin increased AKT phosphorylation and rescued the inhibitory effect of AKT inhibitors III and IV on AKT phosphorylation and early embryonic development. CONCLUSIONS Collectively, results suggest a potential requirement of AKT for bovine early embryonic development, and suggest a potential role for follistatin in regulation of AKT signaling in early bovine embryos.
Collapse
Affiliation(s)
- Mohamed Ashry
- Laboratory of Mammalian Reproductive Biology and Genomics, Michigan State University, East Lansing, MI 48824 USA
- Department of Animal Science, Michigan State University, East Lansing, MI 48824 USA
- Department of Theriogenology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Sandeep K. Rajput
- Laboratory of Mammalian Reproductive Biology and Genomics, Michigan State University, East Lansing, MI 48824 USA
- Department of Animal Science, Michigan State University, East Lansing, MI 48824 USA
| | - Joseph K. Folger
- Laboratory of Mammalian Reproductive Biology and Genomics, Michigan State University, East Lansing, MI 48824 USA
- Department of Animal Science, Michigan State University, East Lansing, MI 48824 USA
| | - Jason G. Knott
- Developmental Epigenetics Laboratory, Michigan State University, East Lansing, MI 48824 USA
- Department of Animal Science, Michigan State University, East Lansing, MI 48824 USA
| | - Nabil A. Hemeida
- Department of Theriogenology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Omaima M. Kandil
- Department of Animal Reproduction and Artificial Insemination, Veterinary Research Division, National Research Center, Giza, Egypt
| | - Refaat S. Ragab
- Department of Theriogenology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - George W. Smith
- Laboratory of Mammalian Reproductive Biology and Genomics, Michigan State University, East Lansing, MI 48824 USA
- Department of Animal Science, Michigan State University, East Lansing, MI 48824 USA
| |
Collapse
|
26
|
Profiling of proteins secreted in the bovine oviduct reveals diverse functions of this luminal microenvironment. PLoS One 2017; 12:e0188105. [PMID: 29155854 PMCID: PMC5695823 DOI: 10.1371/journal.pone.0188105] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 11/01/2017] [Indexed: 11/19/2022] Open
Abstract
The oviductal microenvironment is a site for key events that involve gamete maturation, fertilization and early embryo development. Secretions into the oviductal lumen by either the lining epithelium or by transudation of plasma constituents are known to contain elements conducive for reproductive success. Although previous studies have identified some of these factors involved in reproduction, knowledge of secreted proteins in the oviductal fluid remains rudimentary with limited definition of function even in extensively studied species like cattle. In this study, we used a shotgun proteomics approach followed by bioinformatics sequence prediction to identify secreted proteins present in the bovine oviductal fluid (ex vivo) and secretions from the bovine oviductal epithelial cells (in vitro). From a total of 2087 proteins identified, 266 proteins could be classified as secreted, 109 (41%) of which were common for both in vivo and in vitro conditions. Pathway analysis indicated different classes of proteins that included growth factors, metabolic regulators, immune modulators, enzymes, and extracellular matrix components. Functional analysis revealed mechanisms in the oviductal lumen linked to immune homeostasis, gamete maturation, fertilization and early embryo development. These results point to several novel components that work together with known elements mediating functional homeostasis, and highlight the diversity of machinery associated with oviductal physiology and early events in cattle fertility.
Collapse
|
27
|
Li Y, Liu X, Chen Z, Song D, Yang J, Zuo X, Cao Z, Liu Y, Zhang Y. Effect of follistatin on pre-implantational development of pig parthenogenetic embryos. Anim Sci J 2017; 89:316-327. [PMID: 29119699 DOI: 10.1111/asj.12936] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 08/25/2017] [Indexed: 11/30/2022]
Abstract
The present study was designed to explore effects of follistatin (FST) on pre-implantational development of parthenogenetically activated embryos (PAEs) in pigs. First, we investigated the FST messenger RNA expression level and dynamic FST protein expression patterns in porcine oocytes and PAEs. Then, PAEs were placed in embryo culture medium supplemented with 10 ng/mL of FST-288, FST-300, and FST-315. Next, PAEs were cultured with 0, 1, 10 and 100 ng/mL of FST-315 protein throughout the in vitro culture (IVC) duration. Further, 10 ng/mL of FST-300 was added from the start of IVC in which PAEs were treated for 30, 48 and 60 h. The results showed that 1 ng/mL FST-315 could significantly increase the total cell numbers of blastocyst and trophectoderm cell number in PAEs. Exogenous FST-300 supplementation could significantly promote the early cleavage divisions and improve the blastocyst formation rate of porcine embryos. FST-300 appeared to affect early embryonic development before activation of the embryonic genome. In all, the study confirmed for the first time that FST plays a role in promoting early embryonic development in pigs, which differed with different FST subtypes. FST-300 could facilitate the initial cleavage time and improve the blastocyst formation rate, and FST-315 could improve the blastocyst quality.
Collapse
Affiliation(s)
- 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, China
| | - Xing Liu
- Anhui Provincial Laboratory of Local Livestock and Poultry Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 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, China
| | - Dandan Song
- Anhui Provincial Laboratory of Local Livestock and Poultry Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Jie Yang
- Anhui Provincial Laboratory of Local Livestock and Poultry Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 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, 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, China
| | - Ya Liu
- Anhui Provincial Laboratory of Local Livestock and Poultry Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 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, China
| |
Collapse
|
28
|
Lee SH, Oh HJ, Kim MJ, Kim GA, Choi YB, Jo YK, Setyawan EMN, Lee BC. Effect of co-culture canine cumulus and oviduct cells with porcine oocytes during maturation and subsequent embryo development of parthenotes in vitro. Theriogenology 2017; 106:108-116. [PMID: 29049922 DOI: 10.1016/j.theriogenology.2017.09.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 09/04/2017] [Accepted: 09/09/2017] [Indexed: 10/18/2022]
Abstract
In the estrus stage, canine oocytes are surrounded by cumulus cells and undergo maturation in the oviduct for 2-3 days after ovulation. We hypothesized that canine oviduct cells (cOC) and canine cumulus cells (cCC) during this stage might affect the maturation of oocytes and thereby improve subsequent embryo development. Therefore, the objective of this study was to compare the effects of a cOC and cCC co-culture on oocyte in vitro maturation (IVM) and subsequent embryo development, and to analyze the gene expressions in a molecular fashion what co-culture actually gives the specific pathways in which the co-culture cells act to improve maturation and embryo development. The effect of co-culture using cOC and cCC on porcine oocyte IVM was investigated. Thereafter, oocytes were activated using electrical stimulation and embryo developmental competence was estimated. The expression of the genes related to oocyte maturation, embryo development and apoptosis were analyzed. Also, reactive oxygen species (ROS) levels after IVM was analyzed. The IVM rate and embryo development including cleavage, blastocyst formation rates, and total blastocyst cell numbers from cOC group were significantly higher than other groups (P < 0.05). The expression of SMAD2/3 and growth differentiation factor 9 (GDF9) was significantly increased in cOC and oocytes from the cOC group compared with other groups. Moreover, the levels of GDF9, prostaglandin-endoperoxide synthase 2 (PTGS2), WNT3A and matrix metalloproteinase 2 (MMP2) were significantly up-regulated in blastocysts from the cOC group. The concentration of ROS was significantly lower in the supernatant of cOC groups compared with other groups. Also, the expression of BCL2 was significantly increased in porcine cumulus cells and oocytes from cOC group. The present study demonstrated that co-culture with cOC improved in vitro porcine oocyte maturation and subsequent embryo development competence. Also, co-culture with cOC during IVM induces a suitable environment for oocyte maturation by enhancing the mRNA level of SMAD2/3 and GDF9, and for embryo development by elevating the expression level of PTGS2, WNT3A and MMP2. In addition, the decreased ROS level in cOC co-culture could have a beneficial influence on oocyte maturation.
Collapse
Affiliation(s)
- Seok Hee Lee
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Hyun Ju Oh
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Min Jung Kim
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Geon A Kim
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Yoo Bin Choi
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Young Kwang Jo
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Erif Maha Nugraha Setyawan
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Byeong Chun Lee
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
29
|
Lee BM, Chun JL, Lee JH, Kim EY, Park KS, Lee JH, Daigneault BW, Smith GW, Kim KJ, Chang KT, Lee SR, Kim SU, Choi SA, Lee KB, Kim MK. Follistatin Rescues Blastocyst Development of Poor Quality Porcine Cumulus-Oocyte Complexes by Delaying Meiotic Resumption With Decreased cGMP. Reprod Sci 2017; 25:759-772. [PMID: 28845753 DOI: 10.1177/1933719117725829] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Mammalian oocytes resume maturation when removed from follicles and cultured in vitro. During folliculogenesis, oocytes are bathed in follicular fluid (FF), which provides an important and specialized microenvironment for oocyte competence. Follistatin (FST) is one component of FF that may play a role in oocyte maturation and embryo development. This study was conducted to examine possible effects of FST on porcine oocyte competence and embryo development. Exogenous FST in oocyte maturation medium for 22 or 44 hours did not improve nuclear maturation and had no effect on good quality cumulus-oocyte complexes (COCs). However, FST improved blastocyst rates in embryos derived from oocytes with less than 2 layers of cumulus. Follistatin treatment of the poor quality COC group increased transcript levels for genes indicative of oocyte quality. Transcript levels were also altered for cumulus expansion-related genes in response to FST when measured during the germinal vesicle breakdown stage. Interestingly, high-quality oocytes remained at germinal vesicle stage much longer than low-quality oocytes, FST treatment induced temporary blockage of spontaneous meiotic resumption when added during culture of both good and poor quality COCs, and levels of cyclic guanosine monophosphate (cGMP) were higher in FST-treated versus untreated groups for both good and poor quality oocytes. In conclusion, FST treatment of porcine oocytes during in vitro maturation can rescue competency of poor quality oocytes to develop to blastocyst stage following in vitro fertilization. Beneficial effects of addition of FST to culture medium may be mediated by inhibiting degradation of cGMP and temporarily delaying nuclear maturation.
Collapse
Affiliation(s)
- Bo Myeong Lee
- 1 Department of Animal Science and Biotechnology, College of Agriculture and Life Science, Chungnam National University, Yuseong-gu, Daejeon, Republic of Korea
| | - Ju Lan Chun
- 1 Department of Animal Science and Biotechnology, College of Agriculture and Life Science, Chungnam National University, Yuseong-gu, Daejeon, Republic of Korea
| | - Ji Hye Lee
- 1 Department of Animal Science and Biotechnology, College of Agriculture and Life Science, Chungnam National University, Yuseong-gu, Daejeon, Republic of Korea
| | - Eun Young Kim
- 1 Department of Animal Science and Biotechnology, College of Agriculture and Life Science, Chungnam National University, Yuseong-gu, Daejeon, Republic of Korea
| | - Kang-Sun Park
- 1 Department of Animal Science and Biotechnology, College of Agriculture and Life Science, Chungnam National University, Yuseong-gu, Daejeon, Republic of Korea
| | - Jin-Hee Lee
- 1 Department of Animal Science and Biotechnology, College of Agriculture and Life Science, Chungnam National University, Yuseong-gu, Daejeon, Republic of Korea
| | - Bradford W Daigneault
- 2 Laboratory of Mammalian Reproductive Biology and Genomics, Michigan State University, East Lansing, MI, USA.,3 Department of Animal Science, Michigan State University, East Lansing, MI, USA
| | - George W Smith
- 2 Laboratory of Mammalian Reproductive Biology and Genomics, Michigan State University, East Lansing, MI, USA.,3 Department of Animal Science, Michigan State University, East Lansing, MI, USA.,4 Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Keun Jung Kim
- 5 Chungnam Livestock Research Institute, Cheongyang-Gun, Chungcheongnam-do, Korea
| | - Kyu-Tae Chang
- 6 National Primate Research Center (NPRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Chungcheongbuk-do, Republic of Korea
| | - Sang-Rae Lee
- 6 National Primate Research Center (NPRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Chungcheongbuk-do, Republic of Korea
| | - Sun-Uk Kim
- 6 National Primate Research Center (NPRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Chungcheongbuk-do, Republic of Korea.,7 Futuristic Animal Resource & Research Center (FARRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Chungcheongbuk-do, Republic of Korea
| | - Seon-A Choi
- 6 National Primate Research Center (NPRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Chungcheongbuk-do, Republic of Korea.,7 Futuristic Animal Resource & Research Center (FARRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Chungcheongbuk-do, Republic of Korea
| | - Kyung-Bon Lee
- 8 Department of Biology Education, College of Education, Chonnam National University, Gwangju, Republic of Korea
| | - Min Kyu Kim
- 1 Department of Animal Science and Biotechnology, College of Agriculture and Life Science, Chungnam National University, Yuseong-gu, Daejeon, Republic of Korea
| |
Collapse
|
30
|
Zhenhua G, Rajput SK, Folger JK, Di L, Knott JG, Smith GW. Pre- and Peri-/Post-Compaction Follistatin Treatment Increases In Vitro Production of Cattle Embryos. PLoS One 2017; 12:e0170808. [PMID: 28122009 PMCID: PMC5266319 DOI: 10.1371/journal.pone.0170808] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 01/11/2017] [Indexed: 12/23/2022] Open
Abstract
Our previous studies demonstrated that maternal (oocyte derived) follistatin (FST) expression is positively associated with bovine oocyte competence and exogenous follistatin treatment during the pre-compaction period of development (d 1–3 post insemination) is stimulatory to bovine early embryogenesis in vitro [blastocyst rates and cell numbers/allocation to trophectoderm (TE)]. In the present study, bovine embryos were treated with exogenous follistatin during d 1–3, d 4–7 and d 1–7 post insemination to test the hypothesis that embryotropic effects of exogenous follistatin are specific to the pre-compaction period (d 1–3) of early embryogenesis. Follistatin treatment during d 4–7 (peri-/post-compaction period) of embryo culture increased proportion of embryos reaching blastocyst and expanded blastocyst stage and total cell numbers compared to controls, but blastocyst rates and total cell numbers were lower than observed following d 1–3 (pre-compaction) follistatin treatment. Follistatin supplementation during d 1–7 of embryo culture increased development to blastocyst and expanded blastocyst stages and blastocyst total cell numbers compared to d 1–3 and d 4–7 follistatin treatment and untreated controls. A similar increase in blastocyst CDX2 mRNA and protein (TE cell marker) was observed in response to d 1–3, d 4–7 and d 1–7 follistatin treatment. However, an elevation in blastocyst BMP4 protein (TE cell regulator) was observed in response to d 1–3 and d 1–7, but not d 4–7 (peri-/post-compaction) follistatin treatment. In summary, our study revealed the potential utility of follistatin treatment for increasing the success rate of in vitro embryo production in cattle. Such results also expand our understanding of the embryotropic actions of follistatin and demonstrate that follistatin actions on blastocyst development and cell allocation to the TE layer are not specific to the pre-compaction period.
Collapse
Affiliation(s)
- Guo Zhenhua
- Animal Husbandry Research Institute of Heilongjiang Academy of Agricultural Sciences (HAAS), Harbin, P.R. China
- Laboratory of Mammalian Reproductive Biology and Genomics, Michigan State University, East Lansing, MI, United States of America
| | - Sandeep K. Rajput
- Laboratory of Mammalian Reproductive Biology and Genomics, Michigan State University, East Lansing, MI, United States of America
- Department of Animal Science, Michigan State University, East Lansing, MI, United States of America
| | - Joseph K. Folger
- Laboratory of Mammalian Reproductive Biology and Genomics, Michigan State University, East Lansing, MI, United States of America
- Department of Animal Science, Michigan State University, East Lansing, MI, United States of America
| | - Liu Di
- Animal Husbandry Research Institute of Heilongjiang Academy of Agricultural Sciences (HAAS), Harbin, P.R. China
- Laboratory of Mammalian Reproductive Biology and Genomics, Michigan State University, East Lansing, MI, United States of America
| | - Jason G. Knott
- Department of Animal Science, Michigan State University, East Lansing, MI, United States of America
- Developmental Epigenetics Laboratory, Michigan State University, East Lansing, MI, United States of America
| | - George W. Smith
- Laboratory of Mammalian Reproductive Biology and Genomics, Michigan State University, East Lansing, MI, United States of America
- Department of Animal Science, Michigan State University, East Lansing, MI, United States of America
- * E-mail:
| |
Collapse
|
31
|
Zhang K, Rajput SK, Wang S, Folger JK, Knott JG, Smith GW. CHD1 Regulates Deposition of Histone Variant H3.3 During Bovine Early Embryonic Development. Biol Reprod 2016; 94:140. [PMID: 27170440 PMCID: PMC4946808 DOI: 10.1095/biolreprod.116.138693] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 05/06/2016] [Indexed: 02/06/2023] Open
Abstract
The CHD family of proteins is characterized by the presence of chromodomains and SNF2-related helicase/ATPase domains, which alter gene expression by modification of chromatin structure. Chd1-null embryos arrest at the peri-implantation stage in mice. However, the functional role of CHD1 during preimplantation development remains unclear, given maternal-derived CHD1 may mask the essential role of CHD1 during this stage in traditional knockout models. The objective of this study was to characterize CHD1 expression and elucidate its functional role in preimplantation development using the bovine model. CHD1 mRNA was elevated after meiotic maturation and remained increased through the 16-cell stage, followed by a sharp decrease at morula to blastocyst stage. Similarly, immunoblot analysis indicated CHD1 protein level is increased after maturation, maintained at high level after fertilization and declined sharply afterwards. CHD1 mRNA level was partially decreased in response to alpha-amanitin (RNA polymerase II inhibitor) treatment, suggesting that CHD1 mRNA in eight-cell embryos is of both maternal and zygotic origin. Results of siRNA-mediated silencing of CHD1 in bovine early embryos demonstrated that the percentages of embryos developing to the 8- to 16-cell and blastocyst stages were both significantly reduced. However, expression of NANOG (inner cell mass marker) and CDX2 (trophectoderm marker) were not affected in CHD1 knockdown blastocysts. In addition, we found that histone variant H3.3 immunostaining is altered in CHD1 knockdown embryos. Knockdown of H3.3 using siRNA resulted in a similar phenotype to CHD1-ablated embryos. Collectively, our results demonstrate that CHD1 is required for bovine early development, and suggest that CHD1 may regulate H3.3 deposition during this period.
Collapse
Affiliation(s)
- Kun Zhang
- Laboratory of Mammalian Reproductive Biology and Genomics, Michigan State University, East Lansing, Michigan Department of Animal Science, Michigan State University, East Lansing, Michigan Laboratory of Mammalian Molecular Embryology, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Sandeep K Rajput
- Laboratory of Mammalian Reproductive Biology and Genomics, Michigan State University, East Lansing, Michigan Department of Animal Science, Michigan State University, East Lansing, Michigan
| | - Shaohua Wang
- Laboratory of Mammalian Molecular Embryology, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Joseph K Folger
- Laboratory of Mammalian Reproductive Biology and Genomics, Michigan State University, East Lansing, Michigan Department of Animal Science, Michigan State University, East Lansing, Michigan
| | - Jason G Knott
- Department of Animal Science, Michigan State University, East Lansing, Michigan
| | - George W Smith
- Laboratory of Mammalian Reproductive Biology and Genomics, Michigan State University, East Lansing, Michigan Department of Animal Science, Michigan State University, East Lansing, Michigan Department of Physiology, Michigan State University, East Lansing, Michigan
| |
Collapse
|
32
|
Kobayashi Y, Yoshimoto Y, Yamamoto Y, Kimura K, Okuda K. Roles of EDNs in regulating oviductal NO synthesis and smooth muscle motility in cows. Reproduction 2016; 151:615-22. [PMID: 26980806 DOI: 10.1530/rep-15-0586] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 03/15/2016] [Indexed: 12/12/2022]
Abstract
Endothelins (EDNs) participate in various physiological events including smooth muscle contraction, nitric oxide (NO) synthesis, and embryonic development. In this study, we investigated the regional roles of EDNs produced by bovine oviductal epithelial cells in NO synthesis and smooth muscle motility. Quantification of mRNA expressions indicated that expression of EDN receptor B (EDNRB) in the ampullary region was higher after ovulation than before ovulation, whereas expression of EDNRA in the isthmic region was higher after ovulation than before ovulation. Immunohistochemistry revealed that the EDN receptors (EDNRA and EDNRB) were expressed in the epithelium, whereas smooth muscle showed positive staining only for EDNRA. The expressionsPlease suggest whether 'NOS2' can be treated as the updated symbol for 'iNOS' as per gene nomenclature. of inducible NO synthase (iNOS) protein and its mRNA (NOS2) in cultured epithelial cells isolated from the ampulla were stimulated by EDN1, but not by EDN2 or EDN3, after 1h of incubation. In isthmic epithelial cells, none of the EDNs affected the expression of NOS2 Isometric contraction tests indicated that spontaneous waves were strong in the isthmic region but weak in the ampullary region. EDN1 modulated smooth muscle motility in both the regions. The overall findings suggest that EDN1 plays region-specific roles in smooth muscle motility and epithelial NO synthesis, providing an optimal oviductal microenvironment for transport of gametes, fertilization, and development/transport of early embryo.
Collapse
Affiliation(s)
- Yoshihiko Kobayashi
- Laboratory of Reproductive PhysiologyGraduate School of Environmental and Life Science, Okayama University, Okayama, Japan
| | - Yuka Yoshimoto
- Laboratory of Reproductive PhysiologyGraduate School of Environmental and Life Science, Okayama University, Okayama, Japan
| | - Yuki Yamamoto
- Laboratory of Reproductive PhysiologyGraduate School of Environmental and Life Science, Okayama University, Okayama, Japan
| | - Koji Kimura
- Laboratory of Reproductive PhysiologyGraduate School of Environmental and Life Science, Okayama University, Okayama, Japan
| | - Kiyoshi Okuda
- Laboratory of Reproductive PhysiologyGraduate School of Environmental and Life Science, Okayama University, Okayama, Japan Obihiro University of Agriculture and Veterinary MedicineHokkaido, Japan
| |
Collapse
|
33
|
Schubert C. Superfamily Supports Pregnancy. Biol Reprod 2015. [DOI: 10.1095/biolreprod.115.135756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|