1
|
Gan J, Wang Y, Zhang F, Zang X, Meng F, Gu T, Xu Z, Li Z, Cai G, Wu Z, Hong L. Analysis of differential transcriptome expression reveals that ISG15 provides support for embryo development by promoting angiogenesis in porcine mesometrium. Int J Biol Macromol 2025; 306:141601. [PMID: 40024418 DOI: 10.1016/j.ijbiomac.2025.141601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 02/26/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Mesometrium is the "gateway" through which the endometrium exchanges nutrients and substances with the outside world due to blood vessels entering endometrium across mesometrium. Therefore, dissecting the transcription atlas of the mesometrium will be a great help in understanding the role of mesometrium during implantation. In this study, we collected samples from the mesometrium and adjacent endometrium on the 12th day of estrous cycle and pregnancy. Transcription atlas of mesometrium and adjacent endometrium revealed that genes such as ISG15, which are related to the pathway of response to Interferon α and γ, were significantly enriched. The result of immunohistochemistry demonstrated that the core genes within these pathways were mainly located in the vascular endothelial cells both the endometrium and mesometrium. ISG15 interferon assay revealed the down-regulation of ISG15 induced proliferation, migration and tube formation. Taken together, we concluded that down-regulation of genes related to response of interferons promoted angiogenesis in the mesometrium and adjacent endometrium, allowing mesometrium to play an essential supportive role in pregnancy.
Collapse
Affiliation(s)
- Jianyu Gan
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Yongzhong Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Fan Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Xupeng Zang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Fanming Meng
- Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
| | - Ting Gu
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Zheng Xu
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Zicong Li
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Gengyuan Cai
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Zhenfang Wu
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Yunfu Subcenter of Guangdong Laboratory for Lingnan Modern Agriculture, Yunfu 527300, China.
| | - Linjun Hong
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; National Regional Gene Bank of Livestock (Gene Bank of Guangdong Livestock and Poultry), Guangzhou 510642, China.
| |
Collapse
|
2
|
Dlamini NH, Bridi A, da Silveira JC, Feugang JM. Unlocking Gamete Quality Through Extracellular Vesicles: Emerging Perspectives. BIOLOGY 2025; 14:198. [PMID: 40001966 PMCID: PMC11851576 DOI: 10.3390/biology14020198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 02/08/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025]
Abstract
Extracellular vesicles (EVs) are gaining recognition for their essential role in enhancing gamete quality and improving outcomes in assisted reproductive technologies. These nanosized particles, released by cells, carry proteins, lipids, and RNAs, facilitating critical cell communication and offering the potential to enhance gamete maturation and improve fertilization rates. Most research on males has concentrated on seminal plasma, a complex fluid produced by the testes and accessory glands vital in modulating sperm fertility potential. The components of seminal plasma significantly affect sperm functionality, embryo survival, and placental development, making this a prominent area of interest in reproductive biology. The EVs within seminal plasma contribute to maintaining sperm membrane stability, enhancing motility, and promoting capacitation, which may influence the female reproductive tract following mating. In females, EVs have been identified in both the follicular and uterine environments, where effective embryo-maternal communication is crucial. The oviduct epithelium supports gamete transport and early embryonic development, with EVs found in oviductal fluid playing a key role in reproductive processes. These EVs support the embryo's growth in the nutrient-rich uterine environment. These important studies underscore the significant role of EVs in transporting essential molecular compounds to gametes and embryos, leading to an enhanced understanding and potential manipulation of reproductive processes. This review aims to summarize the current research on the benefits of EVs in gamete manipulation and embryo development, highlighting their promising implications for reproductive health.
Collapse
Affiliation(s)
- Notsile H. Dlamini
- Department of Animal and Dairy Sciences, Mississippi State University, Starkville, MS 39759, USA;
| | - Alessandra Bridi
- University of the West of Santa Catarina, Xanxerê 89820-000, SC, Brazil;
| | | | - Jean M. Feugang
- Department of Animal and Dairy Sciences, Mississippi State University, Starkville, MS 39759, USA;
| |
Collapse
|
3
|
Shang Y, Zhang Q, Ding Y, Wang Y, Gu S, Zang X, Xu Z, Huang S, Li Z, Wu Z, Gu T, Hong L, Cai G. The Expression Pattern and Functional Analysis of Extracellular Vesicle Long Non-Coding RNAs from Uterine Fluid During Implantation in Pig. Animals (Basel) 2025; 15:245. [PMID: 39858245 PMCID: PMC11758334 DOI: 10.3390/ani15020245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 01/05/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Long non-coding RNAs (lncRNAs) act as competing endogenous RNAs and play significant roles in porcine embryo development. Extracellular vesicles (EVs) in the uterine fluid (UF) can target and deliver maternal endometrial signalling molecules to embryonic trophoblast cells, exerting crucial regulatory effects during embryo implantation. However, the specific roles of lncRNAs carried by UF-EVs during the embryo implantation period have not been thoroughly reported in the literature. In the present study, high-throughput sequencing and biological tools were applied to analyse lncRNAs in UF-EVs on days 9, 12, and 15 of pregnancy to identify key regulatory lncRNAs in UF-EVs during the porcine embryonic implantation period and to explore their expression patterns and functional roles. A total of 30,203 lncRNAs were identified and 7879 differentially expressed lncRNAs were screened, and qRT-PCR was used to verify the sequencing data. Days 9-12 of pregnancy represent a critical stage of embryo implantation characterised by substantial morphological changes in porcine embryos. During this period, we identified a total of 4348 differentially expressed lncRNAs. Through screening and validation, we discovered that LNC_026212 was highly expressed on day 12 of pregnancy and can promote the proliferation and migration of porcine trophoblast cells (PTr cells). These novel findings contribute to our understanding of the impact of lncRNAs on porcine reproductive processes, offering new research directions to improve the success rate of embryo implantation in pigs.
Collapse
Affiliation(s)
- Yijun Shang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou 510642, China
| | - Qiuping Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou 510642, China
| | - Yue Ding
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou 510642, China
| | - Yongzhong Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou 510642, China
| | - Shengchen Gu
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou 510642, China
| | - Xupeng Zang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou 510642, China
| | - Zheng Xu
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou 510642, China
| | - Sixiu Huang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou 510642, China
| | - Zicong Li
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou 510642, China
| | - Zhenfang Wu
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou 510642, China
- Yunfu Subcenter of Guangdong Laboratory for Lingnan Modern Agriculture, Yunfu 527300, China
- National Regional Gene Bank of Livestock and Poultry (Gene Bank of Guangdong Livestock and Poultry), Guangzhou 510642, China
| | - Ting Gu
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou 510642, China
- National Regional Gene Bank of Livestock and Poultry (Gene Bank of Guangdong Livestock and Poultry), Guangzhou 510642, China
| | - Linjun Hong
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou 510642, China
- National Regional Gene Bank of Livestock and Poultry (Gene Bank of Guangdong Livestock and Poultry), Guangzhou 510642, China
| | - Gengyuan Cai
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou 510642, China
- Yunfu Subcenter of Guangdong Laboratory for Lingnan Modern Agriculture, Yunfu 527300, China
- National Regional Gene Bank of Livestock and Poultry (Gene Bank of Guangdong Livestock and Poultry), Guangzhou 510642, China
| |
Collapse
|
4
|
Nabeel MA, Nowak RA. Extracellular Vesicles in Implantation: Cross-Talk Between the Embryo and Endometrium. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2025. [PMID: 39741215 DOI: 10.1007/102_2024_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
Abstract
Extracellular vesicles (EVs), including exosomes and microvesicles, have emerged as pivotal mediators of intercellular communication. Embryo implantation is a critical process in early pregnancy and requires communication between the embryo and maternal uterus. EVs are important in coordinating the communication between the embryo and maternal uterus. This review explores EV biogenesis, molecular composition, and functional roles during implantation. It emphasizes the dynamic role of EVs in modulating the maternal-embryo dialogue, which is critical for establishing a receptive endometrium and facilitating successful implantation. EVs secreted by the embryo and endometrial cells have been shown to carry a diverse cargo of proteins, lipids, and miRNAs, which collectively influence key physiological processes, including immune tolerance, endometrial receptivity, and trophoblast invasion. EVs can be potential candidates as non-invasive biomarkers to assess the quality of embryos and uterine receptivity to enhance reproductive success. By providing a comprehensive overview of the current understanding of EVs in implantation, this chapter aims to highlight the significance of EVs in reproductive biology and their potential applications in improving fertility rates.
Collapse
Affiliation(s)
- Muhammad Ashir Nabeel
- Department of Animal Sciences, College of Agricultural, Consumer and Environmental Sciences, University of Illinois, Urbana, IL, USA
| | - Romana A Nowak
- Department of Animal Sciences, College of Agricultural, Consumer and Environmental Sciences, University of Illinois, Urbana, IL, USA.
| |
Collapse
|
5
|
Chen W, Xie Y, Xu Z, Shang Y, Yang W, Wang P, Wu Z, Cai G, Hong L. Identification and Functional Analysis of miRNAs in Extracellular Vesicles of Semen Plasma from High- and Low-Fertility Boars. Animals (Basel) 2024; 15:40. [PMID: 39794983 PMCID: PMC11718777 DOI: 10.3390/ani15010040] [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: 11/19/2024] [Revised: 12/19/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
Artificial insemination (AI), as an efficient assisted reproduction technology, can help the livestock industry to improve livestock and poultry breeds, optimize production performance and improve reproductive efficiency. AI technology has been widely used in pig production in China, but boar fertility affects the effectiveness of AI, and more and more studies have shown that there are significant differences in the fertility of boars with similar semen quality indicators. Therefore, this study aimed to identify biomarker molecules that indicate the level of boar fertility, which is important for improving the efficiency of AI. In this study, we collected 40 mL of ejaculates per boar used for extracellular vesicle (EV) characterization in 20 boars and identified 53 differentially expressed miRNAs by small RNA sequencing, of which 44 miRNAs were up-regulated in the high-fertility seminal EVs compared with low-fertility seminal EVs, and nine miRNAs were down-regulated. miR-26a was most significantly down-regulated in the high-fertility group compared to the low-fertility group, and it was hypothesized that this miRNA could be used as a biomolecular marker of semen reproductive performance. To further determine the effect of miR-26a on sperm function, we successfully established a miR-26a overexpression model and found that miR-26a reduced sperm viability, motility, acrosome integrity, plasma membrane integrity and ATP levels. Bioinformatics analysis and dual luciferase reporter analysis revealed that miR-26a directly targets High mobility group A1 (HMGA1). In conclusion, miR-26a can be used as a biomarker to identify high and low fertility in boar semen.
Collapse
Affiliation(s)
- Weidong Chen
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (W.C.); (Y.X.); (Y.S.); (W.Y.); (P.W.); (Z.W.)
| | - Yanshe Xie
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (W.C.); (Y.X.); (Y.S.); (W.Y.); (P.W.); (Z.W.)
| | - Zhiqian Xu
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471023, China;
| | - Yijun Shang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (W.C.); (Y.X.); (Y.S.); (W.Y.); (P.W.); (Z.W.)
| | - Wenzheng Yang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (W.C.); (Y.X.); (Y.S.); (W.Y.); (P.W.); (Z.W.)
| | - Pengyao Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (W.C.); (Y.X.); (Y.S.); (W.Y.); (P.W.); (Z.W.)
| | - Zhenfang Wu
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (W.C.); (Y.X.); (Y.S.); (W.Y.); (P.W.); (Z.W.)
- Yunfu Subcenter of Guangdong Laboratory for Lingnan Modern Agriculture, Yunfu 527300, China
- Key Laboratory of South China Modern Biological Seed Industry, Ministry of Agriculture and Rural Affairs, Guangzhou 510520, China
| | - Gengyuan Cai
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (W.C.); (Y.X.); (Y.S.); (W.Y.); (P.W.); (Z.W.)
- Yunfu Subcenter of Guangdong Laboratory for Lingnan Modern Agriculture, Yunfu 527300, China
- Key Laboratory of South China Modern Biological Seed Industry, Ministry of Agriculture and Rural Affairs, Guangzhou 510520, China
- National Regional Gene Bank of Livestock and Poultry (Gene Bank of Guangdong Livestock and Poultry), Guangzhou 510642, China
| | - Linjun Hong
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (W.C.); (Y.X.); (Y.S.); (W.Y.); (P.W.); (Z.W.)
- Key Laboratory of South China Modern Biological Seed Industry, Ministry of Agriculture and Rural Affairs, Guangzhou 510520, China
- National Regional Gene Bank of Livestock and Poultry (Gene Bank of Guangdong Livestock and Poultry), Guangzhou 510642, China
| |
Collapse
|
6
|
He F, Li M, Chen F, Zhou R, Qi M, Fu B, Zhang H, Li Q, Bi Y, Huang T. Overexpression of miR-192 Inhibits In Vitro Porcine Embryo Development by Inducing Oxidative Stress Damage and Impairing Mitochondrial Function. Animals (Basel) 2024; 15:46. [PMID: 39794989 PMCID: PMC11718968 DOI: 10.3390/ani15010046] [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: 11/20/2024] [Revised: 12/19/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
Early embryonic development relies on intricately regulated gene expression, and miRNAs influence zygotic genome activation (ZGA), cleavage, and cell fate determination through post-transcriptional regulatory mechanisms. miR-192 is expressed in early pig embryos and participates in various reproductive processes. However, its role in pre-implantation pig embryo development remains poorly understood. In this study, we microinjected the miR-192 agonist (miR-192 agomir) into parthenogenetically activated pig embryos to evaluate its effects on early pig embryo development. Our findings indicate that compared to the control group (agomir NC), miR-192 agomir impairs the developmental capacity of parthenogenetic pig embryos to reach the 2-cell, 4-cell, and blastocyst stages. This impairment leads to imbalances in the oxidative-reductive system and abnormalities in mitochondrial function during the 4-cell stage, resulting in the significant accumulation of ROS, notable decreases in the expression of antioxidant enzymes CAT and SOD1 mRNA, reduction in mitochondrial membrane potential, and induction of apoptosis in pig blastocysts. Additionally, the overexpression of miR-192 inhibits the expression of its target genes YY1 and the pluripotency factor NANOG mRNA. In conclusion, this study reveals that the overexpression of miR-192 adversely affects early pig embryo development, providing new evidence for understanding the role miR-192 plays in reproduction.
Collapse
Affiliation(s)
- Fan He
- College of Animal Science and Technology, Shihezi University, Shihezi 832000, China; (F.H.); (M.L.); (M.Q.); (B.F.); (H.Z.); (Q.L.)
| | - Mingguo Li
- College of Animal Science and Technology, Shihezi University, Shihezi 832000, China; (F.H.); (M.L.); (M.Q.); (B.F.); (H.Z.); (Q.L.)
- Hubei Key Laboratory of Animal Embryo Engineering and Molecular Breeding, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430070, China;
| | - Fan Chen
- Hubei Key Laboratory of Animal Embryo Engineering and Molecular Breeding, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430070, China;
| | - Rong Zhou
- The State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China;
| | - Mengfan Qi
- College of Animal Science and Technology, Shihezi University, Shihezi 832000, China; (F.H.); (M.L.); (M.Q.); (B.F.); (H.Z.); (Q.L.)
| | - Binbin Fu
- College of Animal Science and Technology, Shihezi University, Shihezi 832000, China; (F.H.); (M.L.); (M.Q.); (B.F.); (H.Z.); (Q.L.)
| | - Huapeng Zhang
- College of Animal Science and Technology, Shihezi University, Shihezi 832000, China; (F.H.); (M.L.); (M.Q.); (B.F.); (H.Z.); (Q.L.)
| | - Qingchun Li
- College of Animal Science and Technology, Shihezi University, Shihezi 832000, China; (F.H.); (M.L.); (M.Q.); (B.F.); (H.Z.); (Q.L.)
| | - Yanzhen Bi
- Hubei Key Laboratory of Animal Embryo Engineering and Molecular Breeding, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430070, China;
| | - Tao Huang
- College of Animal Science and Technology, Shihezi University, Shihezi 832000, China; (F.H.); (M.L.); (M.Q.); (B.F.); (H.Z.); (Q.L.)
- Xinjiang Pig Breeding Engineering Technology Research Center, Xinjiang Tecon Husbandry S&T Co., Ltd., Changji 831100, China
| |
Collapse
|
7
|
Xu Z, Zhang K, Yang Y, Chang H, Wen F, Li X. The role of reproductive tract extracellular vesicles on boar sperm function. Theriogenology 2024; 230:278-284. [PMID: 39357166 DOI: 10.1016/j.theriogenology.2024.09.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 10/04/2024]
Abstract
Extracellular vesicles (EVs) are abundant in reproductive tract fluids and serve as important mediators of paracrine communication, influencing the function of gametes. Sperm undergo development in the male reproductive tract and exert their function within the female reproductive tract, engaging in interactions with various types of EVs present throughout the reproductive system. Previous studies have demonstrated that both male and female reproductive tract EVs can impact sperm function by transferring regulatory cargoes to them. Nevertheless, inconsistencies of previous research regarding the effects of EVs on sperm function, coupled with a lack of investigation into the influence of female reproductive tract EVs on sperm fertilization, have left the true role and underlying mechanisms of reproductive tract EVs on sperm function largely unexplored. Given that pigs represent significant economic livestock and serve as an ideal biomedical model for human diseases, this review aims to provide a comprehensive summary of the current knowledge regarding reproductive tract EVs and their influence on boar sperm function, while highlighting their potential roles. We anticipate that this review will facilitate future research on reproductive tract EVs and their impact on sperm function, contributing to improved animal reproductive efficiency and advancements in the treatment of male infertility.
Collapse
Affiliation(s)
- Zhiqian Xu
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471023, Henan, China
| | - Ke Zhang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471023, Henan, China
| | - Youbing Yang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471023, Henan, China
| | - Huixian Chang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471023, Henan, China
| | - Fengyun Wen
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471023, Henan, China.
| | - Xiaoxia Li
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471023, Henan, China.
| |
Collapse
|
8
|
Biase FH, Moorey SE, Schnuelle JG, Rodning S, Ortega MS, Spencer TE. Altered microRNA composition in the uterine lumen fluid in cattle (Bos taurus) pregnancies initiated by artificial insemination or transfer of an in vitro produced embryo. J Anim Sci Biotechnol 2024; 15:130. [PMID: 39267128 PMCID: PMC11397056 DOI: 10.1186/s40104-024-01083-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 07/29/2024] [Indexed: 09/14/2024] Open
Abstract
BACKGROUND MicroRNAs (miRNAs) are presented in the uterine lumen of many mammals, and in vitro experiments have determined that several miRNAs are important for the regulation of endometrial and trophoblast functions. Our aim was to identify and contrast the miRNAs present in extracellular vesicles (EVs) in the uterine lumen fluid (ULF) at the onset of attachment in cattle pregnancies (gestation d 18) initiated by artificial insemination (AI) or by the transfer of an in vitro-produced blastocyst (IVP-ET). A third group had no conceptus after the transfer of an IVP embryo. RESULTS The abundance of 263 annotated miRNAs was quantified in the EVs collected from ULF. There was an increase in the transcript abundance of 20 miRNAs in the ULF EVs from the AI pregnant group, while 4 miRNAs had a lower abundance relative to the group not containing a conceptus. Additionally, 4 miRNAs were more abundant in ULF EVs in the AI pregnant group relative to IVP-ET group (bta-mir-17, bta-mir-7-3, MIR7-1, MIR18A). Specific miRNAs in the ULF EVs were co-expressed with messenger RNAs expressed in extra-embryonic tissues and endometrium, including genes that are known to be their targets. CONCLUSIONS The results provide biological insights into the participation of miRNAs in the regulation of trophoblast proliferation and differentiation, as well as in endometrium receptivity. The knowledge that in vitro cultured embryos can contribute to the altered abundance of specific miRNAs in the uterine lumen can lead to the development of corrective approaches to reduce conceptus losses during the first month of pregnancy in cattle.
Collapse
Affiliation(s)
- Fernando H Biase
- School of Animal Sciences, Virginia Polytechnic Institute and State University, 175 W Campus Dr, Blacksburg, VA, 24061, USA.
| | - Sarah E Moorey
- Department of Animal Science, University of Tennessee, Knoxville, TN, 37996, USA
| | - Julie G Schnuelle
- Department of Clinical Sciences, Auburn University, Auburn, AL, 36849, USA
| | - Soren Rodning
- Department of Animal Science, Auburn University, Auburn, AL, 36849, USA
| | - Martha Sofia Ortega
- Department of Animal and Dairy Sciences, University of Wisconsin Madison, Madison, WI, 53706, USA
| | - Thomas E Spencer
- Division of Animal Sciences, University of Missouri, Columbia, MO, 65211, USA
| |
Collapse
|
9
|
Sunde J, Wasickanin M, Katz TA, Gillette L, Bidadi S, O’Neil D, Masand R, Burney RO, Pennington KA. The uterine secretome initiates growth of gynecologic tissues in ectopic locations. PLoS One 2024; 19:e0292978. [PMID: 38728307 PMCID: PMC11086859 DOI: 10.1371/journal.pone.0292978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 01/03/2024] [Indexed: 05/12/2024] Open
Abstract
Endosalpingiosis (ES) and endometriosis (EM) refer to the growth of tubal and endometrial epithelium respectively, outside of their site of origin. We hypothesize that uterine secretome factors drive ectopic growth. To test this, we developed a mouse model of ES and EM using tdTomato (tdT) transgenic fluorescent mice as donors. To block implantation factors, progesterone knockout (PKO) tdT mice were created. Fluorescent lesions were present after oviduct implantation with and without WT endometrium. Implantation was increased (p<0.05) when tdt oviductal tissue was implanted with endometrium compared to oviductal tissue alone. Implantation was reduced (p<0.0005) in animals implanted with minced tdT oviductal tissue with PKO tdT endometrium compared to WT endometrium. Finally, oviductal tissues was incubated with and without a known implantation factor, leukemia inhibitory factor (LIF) prior to and during implantation. LIF promoted lesion implantation. In conclusion, endometrial derived implantation factors, such as LIF, are necessary to initiate ectopic tissue growth. We have developed an animal model of ectopic growth of gynecologic tissues in a WT mouse which will potentially allow for development of new prevention and treatment modalities.
Collapse
Affiliation(s)
- Jan Sunde
- Department of Obstetrics and Gynecology, Madigan Army Medical Center, Tacoma, WA, United States of America
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Baylor College of Medicine, Houston, TX, United States of America
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston TX, United States of America
| | - Morgan Wasickanin
- Department of Obstetrics and Gynecology, Madigan Army Medical Center, Tacoma, WA, United States of America
| | - Tiffany A. Katz
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Baylor College of Medicine, Houston, TX, United States of America
| | - Laurel Gillette
- Department of Clinical Investigation, Madigan Army Medical Center, Tacoma, WA, United States of America
| | - Sanam Bidadi
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Baylor College of Medicine, Houston, TX, United States of America
| | - Derek O’Neil
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Baylor College of Medicine, Houston, TX, United States of America
| | - Ramya Masand
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston TX, United States of America
- Department of Pathology and Immunology, Baylor College of Medicine, Houston TX, United States of America
| | - Richard O. Burney
- Department of Obstetrics and Gynecology, Madigan Army Medical Center, Tacoma, WA, United States of America
- Department of Clinical Investigation, Madigan Army Medical Center, Tacoma, WA, United States of America
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Kathleen A. Pennington
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Baylor College of Medicine, Houston, TX, United States of America
- Department of Obstetrics and Gynecology, Basic Sciences Perinatology Research Laboratories, Baylor College of Medicine, Houston, TX, United States of America
| |
Collapse
|
10
|
Chang J, Pan X, Gao J, Zhuo Y, Jiang X, Che L, Lin Y, Fang Z, Feng B, Li J, Hua L, Zhao X, Zhang R, Wu D, Xu S. Revealing the mechanism of fiber promoting sow embryo implantation by altering the abundance of uterine fluid proteins: A proteomic perspective. J Proteomics 2024; 297:105123. [PMID: 38364904 DOI: 10.1016/j.jprot.2024.105123] [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: 11/18/2023] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 02/18/2024]
Abstract
Many studies have shown that fiber in the diet plays an important role in improving the reproductive performance of sows, but there is rarely research on the impact of fiber on early embryo implantation. This study used 4D-Label free technology to identify and analyze the effect of the fiber composition in the diet on the protein in the early pregnancy uterine fluid (UF) of sows. The results indicate that ratio of insoluble fibers to soluble fibers (ISF/SF) 4.89 can increase the concentration of progesterone (PROG) and reduce tumor necrosis factorα (TNF-α) concentration in sow UF. In addition, through 4D-Label free, we identified a total of 4248 proteins, 38 proteins abundance upregulated and 283 proteins abundance downregulated in UF. Through enrichment analysis of these differential abundance proteins (DAPs), it was found that these differential proteins are mainly related to the docking of extracellular vesicles, vesicular transport, inflammatory response, and insulin resistance. Therefore, the results of this study reveal the possible mechanism by which fiber improves the reproductive performance of sows, laying a theoretical foundation for future research on the effects of diet on reproduction. SIGNIFICANCE: This study demonstrates the importance of dietary fiber for early embryo implantation in sows. The effect of dietary ISF/SF on early embryo implantation in sows was elucidated from a proteomic perspective through 4D-Label free technology. This study not only has significant implications for improving sow reproductive efficiency, but also provides important theoretical references for studying early miscarriage and reproductive nutrition in human pregnancy.
Collapse
Affiliation(s)
- Junlei Chang
- Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, PR China.
| | - Xujing Pan
- Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, PR China.
| | - Junjie Gao
- Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, PR China.
| | - Yong Zhuo
- Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, PR China.
| | - Xuemei Jiang
- Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, PR China.
| | - Lianqiang Che
- Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, PR China
| | - Yan Lin
- Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, PR China.
| | - Zhengfeng Fang
- Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, PR China
| | - Bin Feng
- Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, PR China
| | - Jian Li
- Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, PR China
| | - Lun Hua
- Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, PR China.
| | - Xilun Zhao
- Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, PR China.
| | - Ruinan Zhang
- Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, PR China.
| | - De Wu
- Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, PR China.
| | - Shengyu Xu
- Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, PR China.
| |
Collapse
|
11
|
Wang Y, Cai S, Chen X, Sun Q, Yin T, Diao L. The role of extracellular vesicles from placenta and endometrium in pregnancy: Insights from tumor biology. J Reprod Immunol 2024; 162:104210. [PMID: 38359619 DOI: 10.1016/j.jri.2024.104210] [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: 04/17/2023] [Revised: 01/13/2024] [Accepted: 01/27/2024] [Indexed: 02/17/2024]
Abstract
Extracellular vesicles (EVs) are small membrane-bound particles secreted by various cell types that play a critical role in intercellular communication by packaging and delivering biomolecules. In recent years, EVs have emerged as essential messengers in mediating physiological and pathological processes in tumor biology. The tumor microenvironment (TME) plays a pivotal role in tumor generation, progression, and metastasis. In this review, we provide an overview of the impact of tumor-derived EVs on both tumor cells and the TME. Moreover, we draw parallels between tumor biology and pregnancy, as successful embryo implantation also requires intricate intercellular communication between the placental trophecepiblast and the endometrial epithelium. Additionally, we discuss the involvement of EVs in targeting immune responses, trophoblast invasion, migration, and angiogenesis, which are shared biological processes between tumors and pregnancy. Specifically, we highlight the effects of placenta-derived EVs on the fetal-maternal interface, placenta, endometrium, and maternal system, as well as the role of endometrium-derived EVs in embryo-endometrial communication. However, challenges still exist in EVs research, including the standardization of EVs isolation methods for diagnostic testing, which also apply to reproductive systems where EVs-mediated communication is proposed to take place. Through this review, we aim to deepen the understanding of EVs, particularly in the context of reproductive biology, and encourage further investigation in this field.
Collapse
Affiliation(s)
- Yanjun Wang
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
| | - Songchen Cai
- Shenzhen Key Laboratory for Reproductive Immunology of Peri-implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Shenzhen Zhongshan Obstetrics & Gynecology Hospital (formerly Shenzhen Zhongshan Urology Hospital), Shenzhen 518045, PR China
| | - Xian Chen
- Shenzhen Key Laboratory for Reproductive Immunology of Peri-implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Shenzhen Zhongshan Obstetrics & Gynecology Hospital (formerly Shenzhen Zhongshan Urology Hospital), Shenzhen 518045, PR China
| | - Qing Sun
- Shenzhen Key Laboratory for Reproductive Immunology of Peri-implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Shenzhen Zhongshan Obstetrics & Gynecology Hospital (formerly Shenzhen Zhongshan Urology Hospital), Shenzhen 518045, PR China
| | - Tailang Yin
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China.
| | - Lianghui Diao
- Shenzhen Key Laboratory for Reproductive Immunology of Peri-implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Shenzhen Zhongshan Obstetrics & Gynecology Hospital (formerly Shenzhen Zhongshan Urology Hospital), Shenzhen 518045, PR China; Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-implantation, Shenzhen 518045, PR China.
| |
Collapse
|
12
|
Beal JR, Ma Q, Bagchi IC, Bagchi MK. Role of Endometrial Extracellular Vesicles in Mediating Cell-to-Cell Communication in the Uterus: A Review. Cells 2023; 12:2584. [PMID: 37998319 PMCID: PMC10670844 DOI: 10.3390/cells12222584] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/26/2023] [Accepted: 10/29/2023] [Indexed: 11/25/2023] Open
Abstract
There are several critical events that occur in the uterus during early pregnancy which are necessary for the establishment and maintenance of pregnancy. These events include blastocyst implantation, uterine decidualization, uterine neoangiogenesis, differentiation of trophoblast stem cells into different trophoblast cell lineages, and formation of a placenta. These processes involve several different cell types within the pregnant uterus. Communication between these cell types must be intricately coordinated for successful embryo implantation and the formation of a functional maternal-fetal interface in the placenta. Understanding how this intricate coordination transpires has been a focus of researchers in the field for many years. It has long been understood that maternal endometrial tissue plays a key role in intercellular signaling during early pregnancy, sending signals to nearby tissues in a paracrine manner. Recently, insights have been obtained into the mechanisms by which these signaling events occur. Notably, the endometrium has been shown to secrete extracellular vesicles (EVs) that contain crucial cargo (proteins, lipids, RNA, miRNA) that are taken up by recipient cells to initiate a response leading to the occurrence of critical events during implantation and placentation. In this review, we aim to summarize the role that endometrium-derived EVs play in mediating cell-to-cell communications within the pregnant uterus to orchestrate the events that must occur to establish and maintain pregnancy. We will also discuss how aberrant endometrial EV signaling may lead to pathophysiological conditions, such as endometriosis and infertility.
Collapse
Affiliation(s)
- Jacob R. Beal
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Qiuyan Ma
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Indrani C. Bagchi
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Milan K. Bagchi
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
13
|
Toledo-Guardiola SM, Luongo C, Abril-Parreño L, Soriano-Úbeda C, Matás C. Different seminal ejaculated fractions in artificial insemination condition the protein cargo of oviductal and uterine extracellular vesicles in pig. Front Cell Dev Biol 2023; 11:1231755. [PMID: 37868907 PMCID: PMC10587466 DOI: 10.3389/fcell.2023.1231755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 09/21/2023] [Indexed: 10/24/2023] Open
Abstract
The seminal plasma (SP) is the liquid component of semen that facilitates sperm transport through the female genital tract. SP modulates the activity of the ovary, oviductal environment and uterine function during the periovulatory and early pregnancy period. Extracellular vesicles (EVs) secreted in the oviduct (oEVs) and uterus (uEVs) have been shown to influence the expression of endometrial genes that regulate fertilization and early embryo development. In some species, semen is composed of well-separated fractions that vary in concentration of spermatozoa and SP composition and volume. This study aimed to investigate the impact of different accumulative fractions of the porcine ejaculate (F1, composed of the sperm-rich fraction, SRF; F2, composed of F1 plus the intermediate fraction; F3, composed of F2 plus the post-SRF) on oEVs and uEVs protein cargo. Six days after the onset of estrus, we determined the oEVs and uEVs size and protein concentration in pregnant sows by artificial insemination (AI-sows) and in non-inseminated sows as control (C-sows). We also identified the main proteins in oEVs and uEVs, in AI-F1, AI-F2, AI-F3, and C-sows. Our results indicated that although the size of EVs is similar between AI- and C-sows, the protein concentration of both oEVs and uEVs was significantly lower in AI-sows (p < 0.05). Proteomic analysis identified 38 unique proteins in oEVs from AI-sows, mainly involved in protein stabilization, glycolytic and carbohydrate processes. The uEVs from AI-sows showed the presence of 43 unique proteins, including already-known fertility-related proteins (EZR, HSPAA901, PDS). We also demonstrated that the protein composition of oEVs and uEVs differed depending on the seminal fraction(s) inseminated (F1, F2, or F3). In conclusion, we found specific protein cargo in oEVs and uEVs according to the type of semen fraction the sow was inseminated with and whose functions these specific EVs proteins are closely associated with reproductive processes.
Collapse
Affiliation(s)
- S. M. Toledo-Guardiola
- Departamento de Fisiología, Facultad de Veterinaria, Campus de Excelencia Mare Nostrum Universidad de Murcia, Murcia, Spain
| | - C. Luongo
- Departamento de Fisiología, Facultad de Veterinaria, Campus de Excelencia Mare Nostrum Universidad de Murcia, Murcia, Spain
| | - L. Abril-Parreño
- Departamento de Fisiología, Facultad de Veterinaria, Campus de Excelencia Mare Nostrum Universidad de Murcia, Murcia, Spain
| | - C. Soriano-Úbeda
- Departamento de Medicina, Cirugía y Anatomía Veterinaria, Universidad de Léon, León, Spain
| | - C. Matás
- Departamento de Fisiología, Facultad de Veterinaria, Campus de Excelencia Mare Nostrum Universidad de Murcia, Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria Pascual Parrilla (IMIB-Arrixaca), Murcia, Spain
| |
Collapse
|
14
|
Guzewska MM, Witek KJ, Karnas E, Rawski M, Zuba-Surma E, Kaczmarek MM. miR-125b-5p impacts extracellular vesicle biogenesis, trafficking, and EV subpopulation release in the porcine trophoblast by regulating ESCRT-dependent pathway. FASEB J 2023; 37:e23054. [PMID: 37402070 DOI: 10.1096/fj.202300710r] [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: 04/11/2023] [Revised: 05/23/2023] [Accepted: 06/12/2023] [Indexed: 07/05/2023]
Abstract
Intercellular communication is a critical process that ensures cooperation between distinct cell types at the embryo-maternal interface. Extracellular vesicles (EVs) are considered to be potent mediators of this communication by transferring biological information in their cargo (e.g., miRNAs) to the recipient cells. miRNAs are small non-coding RNAs that affect the function and fate of neighboring and distant cells by regulating gene expression. Focusing on the maternal side of the dialog, we recently revealed the impact of embryonic signals, including miRNAs, on EV-mediated cell-to-cell communication. In this study, we show the regulatory mechanism of the miR-125b-5p ESCRT-mediated EV biogenesis pathway and the further secretion of EVs by trophoblasts at the time when the crucial steps of implantation are taking place. To test the ability of miR-125b-5p to influence the expression of genes involved in the generation and release of EV subpopulations in porcine conceptuses, we used an ex vivo approach. Next, in silico and in vitro analyses were performed to confirm miRNA-mRNA interactions. Finally, EV trafficking and release were assessed using several imaging and particle analysis tools. Our results indicated that conceptus development and implantation are accompanied by changes in the abundance of EV biogenesis and trafficking machinery. ESCRT-dependent EV biogenesis and the further secretion of EVs were modulated by miR-125b-5p, specifically impacting the ESCRT-II complex (via VPS36) and EV trafficking in primary porcine trophoblast cells. The identified miRNA-ESCRT interplay led to the generation and secretion of specific subpopulations of EVs. miRNA present at the embryo-maternal interface governs EV-mediated communication between the mother and the developing conceptus, leading to the generation, trafficking, and release of characteristic subpopulations of EVs.
Collapse
Affiliation(s)
- Maria M Guzewska
- Department of Hormonal Action Mechanisms, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Krzysztof J Witek
- Cell and Tissue Analysis and Imaging Laboratory, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Elżbieta Karnas
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Michał Rawski
- Małopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
| | - Ewa Zuba-Surma
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Monika M Kaczmarek
- Department of Hormonal Action Mechanisms, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
- Molecular Biology Laboratory, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| |
Collapse
|
15
|
Segura-Benítez M, Bas-Rivas A, Juárez-Barber E, Carbajo-García MC, Faus A, De Los Santos MJ, Pellicer A, Ferrero H. Human blastocysts uptake extracellular vesicles secreted by endometrial cells containing miRNAs related to implantation. Hum Reprod 2023:dead138. [PMID: 37407281 DOI: 10.1093/humrep/dead138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 06/06/2023] [Indexed: 07/07/2023] Open
Abstract
STUDY QUESTION Are the extracellular vesicles (EVs) secreted by the maternal endometrium uptaken by human embryos and is their miRNA cargo involved in implantation and embryo development? SUMMARY ANSWER Data suggest that EVs secreted by human endometrial epithelial cells are internalized by human blastocysts, and transport miRNAs to modulate biological processes related to implantation events and early embryo development. WHAT IS KNOWN ALREADY Successful implantation is dependent on coordination between maternal endometrium and embryo, and EVs role in the required cell-to-cell crosstalk has recently been established. In this regard, our group previously showed that protein cargo of EVs secreted by primary human endometrial epithelial cells (pHEECs) is implicated in biological processes related to endometrial receptivity, embryo implantation, and early embryo development. However, little is known about the regulation of these biological processes through EVs secreted by the endometrium at a transcriptomic level. STUDY DESIGN, SIZE, DURATION A prospective descriptive study was performed. Endometrial biopsies were collected from healthy oocyte donors with confirmed fertility on the day of oocyte retrieval, 36 h after the LH surge. pHEECs were isolated from endometrial biopsies (n = 8 in each pool) and cultured in vitro. Subsequently, conditioned medium was collected and EVs were isolated and characterized. Uptake of EVs by human blastocysts and miRNA cargo of these EVs (n = 3 pools) was analyzed. PARTICIPANTS/MATERIALS, SETTING, METHODS EVs were isolated from the conditioned culture media using ultracentrifugation, and characterization was performed using western blotting, nanoparticle tracking analysis, and transmission electron microscopy. EVs were fluorescently labeled with Bodipy-TR ceramide, and their uptake by human blastocysts was analyzed using confocal microscopy. Analysis of the miRNA cargo of EVs was performed using miRNA sequencing, target genes of the most expressed miRNA were annotated, and functional enrichment analysis was performed. MAIN RESULTS AND THE ROLE OF CHANCE EVs measured 100-300 nm in diameter, a concentration of 1.78 × 1011 ± 4.12 × 1010 (SD) particles/ml and expressed intraluminal protein markers Heat shock protein 70 (HSP70) and Tumor Susceptibility Gene 101 (TSG101), in addition to CD9 and CD81 transmembrane proteins. Human blastocysts efficiently internalized fluorescent EVs within 1-2 h, and more pronounced internalization was observed in the hatched pole of the embryos. miRNA-seq analysis featured 149 annotated miRNAs, of which 37 were deemed most relevant. The latter had 6592 reported gene targets, that in turn, have functional implications in several processes related to embryo development, oxygen metabolism, cell cycle, cell differentiation, apoptosis, metabolism, cellular organization, and gene expression. Among the relevant miRNAs contained in these EVs, we highlight hsa-miR-92a-3p, hsa-let-7b-5p, hsa-miR-30a-5p, hsa-miR-24-3p, hsa-miR-21-5p, and hsa-let-7a-5p as master regulators of the biological processes. LIMITATIONS, REASONS FOR CAUTION This is an in vitro study in which conditions of endometrial cell culture could not mimic the intrauterine environment. WIDER IMPLICATIONS OF THE FINDINGS This study defines potential biomarkers of endometrial receptivity and embryo competence that could be useful diagnostic and therapeutic targets for implantation success, as well as open insight further investigations to elucidate the molecular mechanisms implicated in a successful implantation. STUDY FUNDING/COMPETING INTEREST(S) This study was supported by the Spanish Ministry of Education through FPU awarded to M.S.-B. (FPU18/03735), the Health Institute Carlos III awarded to E.J.-B. (FI19/00110) and awarded to H.F. by the Miguel Servet Program 'Fondo Social Europeo «El FSE invierte en tu futuro»' (CP20/00120), and Generalitat Valenciana through VALi+d Programme awarded to M.C.C.-G. (ACIF/2019/139). The authors have no conflicts of interest to disclose. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Marina Segura-Benítez
- Fundación IVI, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
- Departamento de Pediatría, Obstetricia y Ginecología, Universidad de Valencia, Valencia, Spain
| | - Alba Bas-Rivas
- Fundación IVI, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | | | - María Cristina Carbajo-García
- Fundación IVI, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
- Departamento de Pediatría, Obstetricia y Ginecología, Universidad de Valencia, Valencia, Spain
| | - Amparo Faus
- Fundación IVI, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - María José De Los Santos
- Fundación IVI, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
- IVIRMA Valencia, Valencia, Spain
| | - Antonio Pellicer
- Fundación IVI, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
- IVIRMA Rome, Rome, Italy
| | - Hortensia Ferrero
- Fundación IVI, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| |
Collapse
|
16
|
Guzewska MM, Szuszkiewicz J, Kaczmarek MM. Extracellular vesicles: Focus on peri-implantation period of pregnancy in pigs. Mol Reprod Dev 2023; 90:634-645. [PMID: 36645872 DOI: 10.1002/mrd.23664] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 12/05/2022] [Accepted: 12/10/2022] [Indexed: 01/18/2023]
Abstract
The establishment of cell-to-cell communication between the endometrium and the developing embryo is the most important step in successful mammalian pregnancy. Close interaction between the uterine luminal epithelium and trophoblast cells requires triggering timely molecular dialog for successful maternal recognition of pregnancy, embryo implantation, and placenta development. Quite recently, extracellular vesicles (EVs) carrying unique molecular cargo emerged as evolutionarily conserved mediators of cell-to-cell communication during early pregnancy. To date, the presence of EVs at the embryo-maternal interface has been demonstrated in numerous mammals, including domestic livestock, such as pigs. However, few studies have focused on revealing the mechanism of EV-mediated crosstalk between developing early embryos and receptive endometrium. Over the past years, it has appeared that understanding the role of EVs in mammalian reproduction can substantially improve our understanding of the biological challenges of successful reproductive performance. This review describes current knowledge of EVs, specifically in relation to the peri-implantation period in pigs, characterized by common features of embryo implantation and high embryonic mortality in mammals.
Collapse
Affiliation(s)
- Maria M Guzewska
- Department of Hormonal Action Mechanisms, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Joanna Szuszkiewicz
- Department of Hormonal Action Mechanisms, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Monika M Kaczmarek
- Department of Hormonal Action Mechanisms, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
- Molecular Biology Laboratory, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| |
Collapse
|
17
|
Hong L, Zang X, Hu Q, He Y, Xu Z, Xie Y, Gu T, Yang H, Yang J, Shi J, Zheng E, Huang S, Xu Z, Liu D, Cai G, Li Z, Wu Z. Uterine luminal-derived extracellular vesicles: potential nanomaterials to improve embryo implantation. J Nanobiotechnology 2023; 21:79. [PMID: 36882792 PMCID: PMC9990359 DOI: 10.1186/s12951-023-01834-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 03/02/2023] [Indexed: 03/09/2023] Open
Abstract
Most pregnancy losses worldwide are caused by implantation failure for which there is a lack of effective therapeutics. Extracellular vesicles are considered potential endogenous nanomedicines because of their unique biological functions. However, the limited supply of ULF-EVs prevents their development and application in infertility diseases such as implantation failure. In this study, pigs were used as a human biomedical model, and ULF-EVs were isolated from the uterine luminal. We comprehensively characterized the proteins enriched in ULF-EVs and revealed their biological functions in promoting embryo implantation. By exogenously supplying ULF-EVs, we demonstrated that ULF-EVs improve embryo implantation, suggesting that ULF-EVs are a potential nanomaterial to treat implantation failure. Furthermore, we identified that MEP1B is important in improving embryo implantation by promoting trophoblast cell proliferation and migration. These results indicated that ULF-EVs can be a potential nanomaterial to improve embryo implantation.
Collapse
Affiliation(s)
- Linjun Hong
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, People's Republic of China. .,Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, People's Republic of China. .,Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, People's Republic of China.
| | - Xupeng Zang
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, People's Republic of China.,Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, People's Republic of China.,Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, People's Republic of China
| | - Qun Hu
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, People's Republic of China.,Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, People's Republic of China.,Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, People's Republic of China
| | - Yanjuan He
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, People's Republic of China.,Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, People's Republic of China.,Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, People's Republic of China
| | - Zhiqian Xu
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, People's Republic of China.,Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, People's Republic of China.,Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, People's Republic of China
| | - Yanshe Xie
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, People's Republic of China.,Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, People's Republic of China.,Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, People's Republic of China
| | - Ting Gu
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, People's Republic of China.,Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, People's Republic of China.,Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, People's Republic of China
| | - Huaqiang Yang
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, People's Republic of China.,Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, People's Republic of China.,Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, People's Republic of China
| | - Jie Yang
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, People's Republic of China.,Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, People's Republic of China.,Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, People's Republic of China
| | - Junsong Shi
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, People's Republic of China.,Yunfu Subcenter of Guangdong Laboratory for Lingnan Modern Agriculture, Yunfu, 527300, People's Republic of China
| | - Enqin Zheng
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, People's Republic of China.,Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, People's Republic of China.,Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, People's Republic of China
| | - Sixiu Huang
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, People's Republic of China.,Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, People's Republic of China.,Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, People's Republic of China
| | - Zheng Xu
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, People's Republic of China.,Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, People's Republic of China.,Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, People's Republic of China
| | - Dewu Liu
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, People's Republic of China.,Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, People's Republic of China.,Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, People's Republic of China
| | - Gengyuan Cai
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, People's Republic of China.,Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, People's Republic of China.,Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, People's Republic of China
| | - Zicong Li
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, People's Republic of China. .,Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, People's Republic of China. .,Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, People's Republic of China. .,State Key Laboratory of Livestock and Poultry Breeding, Guangzhou, 510642, People's Republic of China.
| | - Zhenfang Wu
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, People's Republic of China. .,Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, People's Republic of China. .,Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, People's Republic of China. .,State Key Laboratory of Livestock and Poultry Breeding, Guangzhou, 510642, People's Republic of China.
| |
Collapse
|
18
|
Interaction between Microbes and Host in Sow Vaginas in Early Pregnancy. mSystems 2023; 8:e0119222. [PMID: 36749039 PMCID: PMC10134864 DOI: 10.1128/msystems.01192-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Extensive research has explored the causes of embryo losses during early pregnancy by analyzing interaction mechanisms in sows' uterus, ignoring the importance of the lower reproductive tract in pregnancy development regulation. Despite recent progress in understanding the diversity of vaginal microbes under different physiological states, the dynamic of sows' vaginal microbiotas during pregnancy and the interaction between vaginal microbes and the host are poorly understood. Here, we performed a comprehensive analysis of sows' vaginal microbial communities in early pregnancy coupled with overall patterns of vaginal mucosal epithelium gene expression. The vaginal microbiota was analyzed by 16s rRNA or metagenome sequencing, and the vaginal mucosal epithelium transcriptome was analyzed by RNA sequencing, followed by integration of the data layers. We found that the sows' vaginal microbiotas in early pregnancy develop dynamically, and there is a homeostasis balance of Firmicutes and Proteobacteria. Subsequently, we identified two pregnancy-specific communities, which play diverse roles. The microbes in the vagina stimulate the epithelial cells, while vaginal epithelium changes its structure and functions in response to stimulation. These changes produce specific inflammation responses to promote pregnancy development. Our findings demonstrate the interaction between microbes and host in the sow vagina in early pregnancy to promote pregnancy development, meanwhile providing a reference data set for the study of targeted therapies of microbial homeostasis dysregulation in the female reproductive tract. IMPORTANCE This work sheds light on the dynamics of the sow vaginal microbiotas in early pregnancy and its roles in pregnancy development. Furthermore, this study provides insight into the functional mechanisms of reproductive tract microbes by outlining vaginal microbe-host interactions, which might identify new research and intervention targets for improving pregnancy development by modulating lower reproductive tract microbiota.
Collapse
|
19
|
Extracellular vesicles-encapsulated microRNA in mammalian reproduction: A review. Theriogenology 2023; 196:174-185. [PMID: 36423512 DOI: 10.1016/j.theriogenology.2022.11.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 11/08/2022] [Accepted: 11/12/2022] [Indexed: 11/16/2022]
Abstract
Extracellular vesicles (EVs) are nanoscale cell-derived lipid vesicles that participate in cell-cell communication by delivering cargo, including mRNAs, proteins and non-coding RNAs, to recipient cells. MicroRNA (miRNA), a non-coding RNA typically 22 nucleotides long, is crucial for nearly all developmental and pathophysiological processes in mammals by regulating recipient cells gene expression. Infertility is a worldwide health issue that affects 10-15% of couples during their reproductive years. Although assisted reproductive technology (ART) gives infertility couples hope, the failure of ART is mainly unknown. It is well accepted that EVs-encapsulated miRNAs have a role in different reproductive processes, implying that these EVs-encapsulated miRNAs could optimize ART, improve reproductive rate, and treat infertility. As a result, in this review, we describe the present understanding of EVs-encapsulated miRNAs in reproduction regulation.
Collapse
|
20
|
Fan W, Qi Y, Wang Y, Yan H, Li X, Zhang Y. Messenger roles of extracellular vesicles during fertilization of gametes, development and implantation: Recent advances. Front Cell Dev Biol 2023; 10:1079387. [PMID: 36684431 PMCID: PMC9849778 DOI: 10.3389/fcell.2022.1079387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 12/16/2022] [Indexed: 01/07/2023] Open
Abstract
Extracellular vesicles (EVs) have become a research hotspot in recent years because they act as messengers between cells in the physiological and pathological processes of the human body. It can be produced by the follicle, prostate, embryo, uterus, and oviduct in the reproductive field and exists in the extracellular environment as follicular fluid, semen, uterine cavity fluid, and oviduct fluid. Because extracellular vesicles are more stable at transmitting information, it allows all cells involved in the physiological processes of embryo formation, development, and implantation to communicate with one another. Extracellular vesicles carried miRNAs and proteins as mail, and when the messenger delivers the mail to the recipient cell, the recipient cell undergoes a series of changes. Current research begins with intercepting and decoding the information carried by extracellular vesicles. This information may help us gain a better understanding of the secrets of reproduction, as well as assist reproductive technology as an emerging marker and treatment.
Collapse
Affiliation(s)
- Weisen Fan
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yinghua Qi
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yaqian Wang
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Huiting Yan
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xuan Li
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yingjie Zhang
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan, China,*Correspondence: Yingjie Zhang,
| |
Collapse
|
21
|
Ssc-miR-92b-3p Regulates Porcine Trophoblast Cell Proliferation and Migration via the PFKM Gene. Int J Mol Sci 2022; 23:ijms232416138. [PMID: 36555776 PMCID: PMC9784024 DOI: 10.3390/ijms232416138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 12/13/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Embryo implantation, the pivotal stage of gestation, is fundamentally dependent on synchronous embryonic development and uterine receptivity. In the early gestation period, the uterus and conceptus secrete growth factors, cytokines, and hormones to promote implantation. Circulating exosomal miRNAs are potential indicators of normal or complicated gestation. Our previous study revealed that pregnant sows' serum exosomes had upregulated miR-92b-3p expression compared to non-pregnant sows, and that the expression level progressively increased during early gestation. The present study's findings indicate that, compared to the ninth day of the estrous cycle (C9), pregnant sows had upregulated miR-92b-3p expression in the endometrium and embryos during the implantation stage ranging from day 9 to day 15 of gestation. Additionally, our results demonstrate that miR-92b-3p promotes the proliferation and migration of Porcine Trophoblast Cells (PTr2). Dual-Luciferase Reporter (DLR) gene assay, real-time fluorescent quantitative PCR (RT-qPCR), and Western blotting (WB) confirmed the bioinformatics prediction that phosphofructokinase-M (PFKM) serves as a target gene of miR-92b-3p. Notably, interference of PFKM gene expression markedly promoted PTr2 proliferation and migration. Furthermore, mice with downregulated uterine miR-92b-3p expression had smaller rates of successful embryo implantation. In summary, miR-92b-3p putatively modulates embryo implantation by promoting PTr2 proliferation and migration via its target gene PFKM.
Collapse
|
22
|
Hua R, Liu Q, Lian W, Kang TT, Gao D, Huang C, Wang Y, Lei M. Extracellular vesicles derived from endometrial epithelial cells deliver exogenous miR-92b-3p to affect the function of embryonic trophoblast cells via targeting TSC1 and DKK3. Reprod Biol Endocrinol 2022; 20:152. [PMID: 36284344 PMCID: PMC9594956 DOI: 10.1186/s12958-022-01023-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 10/12/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) could mediate embryo-maternal communication to affect embryo implantation by delivering biology information, including microRNA (miRNA), protein, lipid. Our previous research shows that miR-92b-3p was differentially expressed in EVs of uterine flushing fluids during the embryo implantation period. However, the role of miR-92b-3p from EVs in embryo implantation remains elusive. MATERIALS AND METHODS EVs were isolated from porcine endometrial epithelial cells (EECs) by ultracentrifugation. MiR-92b-3p mimics and EVs were used to regulate the expression of miR-92b-3p in porcine trophoblast cells (PTr2 cells). Cell proliferation, migration and adhesion analyses were used to observe the phenotype. RT-qPCR, western blot and dual-luciferase reporter assay were used to assess the targets of miR-92b-3p. RESULTS In this study, EVs derived from porcine EECs were identified and could be taken up by PTr2 cells. We found that the EVs derived from EECs transfected with miR-92b-3p mimic (EVs-miR-92b-3p) significantly promoted the proliferation, migration and adhesion of PTr2 cells. We verified that Tuberous sclerosis complex subunit (TSC1) and Dickkopf 3 (DKK3) were the target genes of miR-92b-3p. Moreover, our study showed that miR-92b-3p plays a vital role in PTr2 cells via targeting TSC1 and DKK3. Furthermore, the 3'UTR vectors of TSC1 and DKK3 can rescue the effect of miR-92b-3p on PTr2 cells. CONCLUSIONS Taken together, this study reveals a novel mechanism that EVs derived from porcine EECs treated with miR-92b-3p crosstalk with trophoblasts by targeting TSC1 and DKK3, leading to an enhanced ability for implantation.
Collapse
Affiliation(s)
- Renwu Hua
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education and Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430000, China
- Shenzhen Key Laboratory of Fertility Regulation, Center of Assisted Reproduction and Embryology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518053, China
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology,Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Qiaorui Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education and Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430000, China
| | - Weisi Lian
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education and Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430000, China
| | - Ting Ting Kang
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518000, China
| | - Dengying Gao
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education and Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430000, China
| | - Cheng Huang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education and Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430000, China
| | - Yueying Wang
- Department of Reproductive Medicine, Jining No.1 People's Hospital, Jining, 272000, China.
| | - Minggang Lei
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education and Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430000, China.
| |
Collapse
|
23
|
Rudolf Vegas A, Hamdi M, Podico G, Bollwein H, Fröhlich T, Canisso IF, Bauersachs S, Almiñana C. Uterine extracellular vesicles as multi-signal messengers during maternal recognition of pregnancy in the mare. Sci Rep 2022; 12:15616. [PMID: 36114358 PMCID: PMC9481549 DOI: 10.1038/s41598-022-19958-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 09/07/2022] [Indexed: 11/09/2022] Open
Abstract
In contrast to other domestic mammals, the embryo-derived signal(s) leading to maternal recognition of pregnancy (MRP) are still unknow in the mare. We hypothesize that these embryonic signals could be packed into uterine extracellular vesicles (uEVs), acting as multi-signal messengers between the conceptus and the maternal tract, and contributing to MRP. To unveil these signals, the RNA and protein cargos of uEVs isolated from uterine lavages collected from pregnant mares (P; day 10, 11, 12 and 13 after ovulation) and cyclic control mares (C; day 10 and 13 after ovulation) were analyzed. Our results showed a fine-tuned regulation of the uEV cargo (RNAs and proteins), by the day of pregnancy, the estrous cycle, and even the size of the embryo. A particular RNA pattern was identified with specific increase on P12 related to immune system and hormonal response. Besides, a set of proteins as well as RNAs was highly enriched in EVs on P12 and P13. Differential abundance of miRNAs was also identified in P13-derived uEVs. Their target genes were linked to down- or upregulated genes in the embryo and the endometrium, exposing their potential origin. Our study identified for first time specific molecules packed in uEVs, which were previously associated to MRP in the mare, and thus bringing added value to the current knowledge. Further integrative and functional analyses will help to confirm the role of these molecules in uEVs during MRP in the mare.
Collapse
Affiliation(s)
- Alba Rudolf Vegas
- Functional Genomics Group, Institute of Veterinary Anatomy, Vetsuisse Faculty Zurich, University of Zurich, 8315, Lindau, ZH, Switzerland
| | - Meriem Hamdi
- Functional Genomics Group, Institute of Veterinary Anatomy, Vetsuisse Faculty Zurich, University of Zurich, 8315, Lindau, ZH, Switzerland
| | - Giorgia Podico
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61802, USA
| | - Heinrich Bollwein
- Clinic of Reproductive Medicine, Vetsuisse-Faculty, University of Zurich, 8315, Lindau, ZH, Switzerland
| | - Thomas Fröhlich
- Gene Center, Laboratory for Functional Genome Analysis, LMU Munich, 81377, Munich, Germany
| | - Igor F Canisso
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61802, USA
| | - Stefan Bauersachs
- Functional Genomics Group, Institute of Veterinary Anatomy, Vetsuisse Faculty Zurich, University of Zurich, 8315, Lindau, ZH, Switzerland
| | - Carmen Almiñana
- Functional Genomics Group, Institute of Veterinary Anatomy, Vetsuisse Faculty Zurich, University of Zurich, 8315, Lindau, ZH, Switzerland.
| |
Collapse
|
24
|
Szuszkiewicz J, Myszczynski K, Reliszko ZP, Heifetz Y, Kaczmarek MM. Early steps of embryo implantation are regulated by exchange of extracellular vesicles between the embryo and the endometrium. FASEB J 2022; 36:e22450. [PMID: 35848638 DOI: 10.1096/fj.202200677r] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/12/2022] [Accepted: 06/30/2022] [Indexed: 11/11/2022]
Abstract
In early pregnancy, as the embryo arrives in the uterus, intensive communication between the embryo and uterus begins. Hundreds of molecules are known to be involved, but despite numerous findings, full understanding of the complexity of the embryo-maternal dialog remains elusive. Recently, extracellular vesicles, nanoparticles able to transfer functionally active cargo between cells, have emerged as important players in cell-cell communication, and as such, they have gained great attention over the past decade also in reproductive biology. Here, we use a domestic animal model (Sus scrofa) with an epitheliochorial, superficial type of placentation because of its advantage in studding uterine luminal fluid extracellular vesicles. We show that during early pregnancy, the uterine lumen is abundant with extracellular vesicles that carry a plethora of miRNAs able to target genes involved in embryonic and organismal development. These extracellular vesicles, upon the delivery to primary trophoblast cells, affect genes governing development as well as cell-to-cell signaling and interactions, consequently having an impact on trophoblast cell proliferation, migration, and invasion. We conclude that the exchange of a unique population of extracellular vesicles and their molecular cargo at the maternal-embryo interface is the key to the success of embryo implantation and pregnancy.
Collapse
Affiliation(s)
- Joanna Szuszkiewicz
- Department of Hormonal Action Mechanisms, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Kamil Myszczynski
- Molecular Biology Laboratory, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Zaneta P Reliszko
- Department of Hormonal Action Mechanisms, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Yael Heifetz
- Department of Entomology, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Monika M Kaczmarek
- Department of Hormonal Action Mechanisms, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland.,Molecular Biology Laboratory, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| |
Collapse
|
25
|
He Y, Zang X, Kuang J, Yang H, Gu T, Yang J, Li Z, Zheng E, Xu Z, Cai G, Wu Z, Hong L. iTRAQ-based quantitative proteomic analysis of porcine uterine fluid during pre-implantation period of pregnancy. J Proteomics 2022; 261:104570. [DOI: 10.1016/j.jprot.2022.104570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 02/28/2022] [Accepted: 03/19/2022] [Indexed: 10/18/2022]
|