DOI QR코드

DOI QR Code

Alfalfa xenomiR-162 targets G protein subunit gamma 11 to regulate milk protein synthesis in bovine mammary epithelial cells

  • Guizhi Meng (Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, College of Animal Science and Technology, Ningxia University) ;
  • Hongjuan Duan (Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, College of Animal Science and Technology, Ningxia University) ;
  • Jingying Jia (Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, College of Animal Science and Technology, Ningxia University) ;
  • Baobao Liu (Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, College of Animal Science and Technology, Ningxia University) ;
  • Yun Ma (Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, College of Animal Science and Technology, Ningxia University) ;
  • Xiaoyan Cai (Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, College of Animal Science and Technology, Ningxia University)
  • Received : 2023.09.15
  • Accepted : 2023.11.11
  • Published : 2024.03.01

Abstract

Objective: It was shown that microRNAs (miRNAs) play an important role in milk protein synthesis. However, the post-transcriptional regulation of casein expression by exogenous miRNA (xeno-miRNAs) in ruminants remains unclear. This study explores the regulatory roles of alfalfa xeno-miR162 on casein synthesis in bovine mammary epithelial cells (bMECs). Methods: The effects of alfalfa xenomiR-162 and G protein subunit gamma 11 (GNG11) on proliferation and milk protein metabolism of bMECs were detected by 5-Ethynyl-2'-Deoxyuridine (EdU) staining, flow cytometry, cell counting kit-8 (CCK-8), enzyme-linked immunosorbent assay, quantitative real-time polymerase chain reaction (qRT-PCR), and Western blot. Dual-luciferase reporter assay was used to verify the targeting relationship between GNG11 and xenomiR-162. Results: Results showed that over-expression of xenomiR-162 inhibited cell proliferation but promoted apoptosis, which also up-regulated the expression of several casein coding genes, including CSN1S1, CSN1S2, and CSN3, while decreasing the expression of CSN2. Furthermore, the targeting relationship between GNG11 and xenomiR-162 was determined, and it was confirmed that GNG11 silencing also inhibited cell proliferation but promoted apoptosis and reduced the expression of casein coding genes and genes related to the mammalian target of rapamycin (mTOR) pathway. Conclusion: Alfalfa xenomiR-162 appears to regulate bMECs proliferation and milk protein synthesis via GNG11 in the mTOR pathway, suggesting that this xeno-miRNA could be harnessed to modulate CSN3 expression in dairy cows, and increase κ-casein contents in milk.

Keywords

Acknowledgement

We thank LetPub (www.letpub.com) for linguistic assistance and pre-submission expert review. This work was funded by the National Natural Science Foundation of China (31960680); Outstanding Youth Program of Natural Science Foundation of Ningxia Hui Autonomous Region (2022AAC05012); Key Program of Natural Science Foundation of Ningxia Hui Autonomous Region (2023AAC02012).

References

  1. Liu J, Wang F, Song H, et al. Soybean-derived gma-miR159a alleviates colon tumorigenesis by suppressing TCF7/MYC in mice. J Nutr Biochem 2021; 92:108627. https://doi.org/10.1016/j.jnutbio.2021.108627 
  2. Lee RC, Feinbaum RL, Ambros V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 1993; 75:843-54. https://doi.org/10.1016/0092-8674(93)90529-y 
  3. Zhang L, Hou D, Chen X, et al. Exogenous plant MIR168a specifically targets mammalian LDLRAP1: evidence of cross-kingdom regulation by microRNA. Cell Res 2012; 22:107-26. https://doi.org/10.1038/cr.2011.158 
  4. Zhao Q, Liu Y, Zhang N, et al. Correction: Evidence for plant-derived xenomiRs based on a large-scale analysis of public small RNA sequencing data from human samples. PLoS One 2020;15:e230146. https://doi.org/10.1371/journal.pone.0230146 
  5. Jiao BL, Zhang XL, Wang SH, et al. MicroRNA-221 regulates proliferation of bovine mammary gland epithelial cells by targeting the STAT5a and IRS1 genes. J Dairy Sci 2019; 102:426-35. https://doi.org/10.3168/jds.2018-15108 
  6. Tian L, Zhang L, Cui Y, et al. miR-142-3p regulates milk synthesis and structure of murine mammary glands via PRLR-mediated multiple signaling pathways. J Agric Food Chem 2019; 67:9532-42. https://doi.org/10.1021/acs.jafc.9b03734 
  7. Chin AR, Fong MY, Somlo G, et al. Cross-kingdom inhibition of breast cancer growth by plant miR159. Cell Res 2016; 26:217-28. https://doi.org/10.1038/cr.2016.13 
  8. Jia J, Duan H, Liu B, Ma Y, Ma Y, Cai X. Alfalfa Xeno-miR168b target CPT1A to regulate milk fat synthesis in bovine mammary epithelial cells. Metabolites 2023; 13:76. https://doi.org/10.3390/metabo13010076 
  9. Severin S, Wenshui X. Milk biologically active components as nutraceuticals: review. Crit Rev Food Sci Nutr 2005;45:645-56. https://doi.org/10.1080/10408690490911756 
  10. Chen KL, Qian Y, Jiang LZ, et al. Regulation of SIRT7 on gene expression of milk protein, milk fat and lactose synthesis in dairy mammary epithelial cells. J Nanjing Agric Univ 2019;42:917-23. (In Chinese) 
  11. Li Y, Ren Q, Wang X, Luoreng Z, Wei D. Bta-miR-199a-3p inhibits LPS-induced inflammation in bovine mammary epithelial cells via the PI3K/AKT/NF-kappaB signaling pathway. Cells 2022; 11:3518. https://doi.org/10.3390/cells11213518 
  12. Herve L, Quesnel H, Veron M, et al. Milk yield loss in response to feed restriction is associated with mammary epithelial cell exfoliation in dairy cows. J Dairy Sci 2019; 102:2670-85. https://doi.org/10.3168/jds.2018-15398 
  13. Yu WY, Cai W, Ying HZ, Zhang WY, Zhang HH, Yu CH. Exogenous Plant gma-miR-159a, identified by miRNA library functional screening, ameliorated hepatic stellate cell activation and inflammation via inhibiting GSK-3beta-mediated pathways. J Inflamm Res 2021;14:2157-72. https://doi.org/10.2147/JIR.S304828 
  14. Luo Y, Wang P, Wang X, et al. Detection of dietetically absorbed maize-derived microRNAs in pigs. Sci Rep 2017; 7:645. https://doi.org/10.1038/s41598-017-00488-y 
  15. Liang H, Huang L, Cao J, Zen K, Chen X, Zhang CY. Regulation of mammalian gene expression by exogenous microRNAs. Wiley Interdiscip Rev RNA 2012; 3:733-42. https:// doi.org/10.1002/wrna.1127 
  16. Li QZ. Study on transmission of plant miRNAs between porcine mother and fetus. Yaan, Sichuan, China: Sichuan Agricultural University; 2013. 54 p. (In Chinese) 
  17. Jiang MM, Zhao F, Lou TT. Assessment of significant pathway signaling and prognostic value of gng11 in ovarian serous cystadenocarcinoma. Int J Gen Med 2021; 14:2329-41. https://doi.org/10.2147/IJGM.S314911 
  18. Gilman AG. G proteins: transducers of receptor-generated signals. Annu Rev Biochem 1987; 56:615-49. https://doi.org/10.1146/annurev.bi.56.070187.003151 
  19. Cheng C, Hua J, Tan J, Qian W, Zhang L, Hou X. Identification of differentially expressed genes, associated functional terms pathways, and candidate diagnostic biomarkers in inflammatory bowel diseases by bioinformatics analysis. Exp Ther Med 2019; 18:278-88. https://doi.org/10.3892/etm.2019.7541 
  20. Shi K, Li N, Yang M, Li W. Identification of key genes and pathways in female lung cancer patients who never smoked by a bioinformatics analysis. J Cancer 2019; 10:51-60. https://doi.org/10.7150/jca.26908 
  21. Zhou R, Zhou J, Muhuitijiang B, Tan W. Construction and experimental validation of a B cell senescence-related gene signature to evaluate prognosis and immunotherapeutic sensitivity in bladder cancer. Funct Integr Genomics 2022;23:3. https://doi.org/10.1007/s10142-022-00936-7 
  22. Patel A, Garcia-Closas M, Olshan AF, et al. Gene-level germline contributions to clinical risk of recurrence scores in black and white patients with breast cancer. Cancer Res 2022;82:25-35. https://doi.org/10.1158/0008-5472.CAN-21-1207
  23. Liu G, Liao Y, Sun B, et al. Effects of chronic heat stress on mRNA and miRNA expressions in dairy cows. Gene 2020;742:144550. https://doi.org/10.1016/j.gene.2020.144550 
  24. Herve L, Quesnel H, Veron M, et al. Milk yield loss in response to feed restriction is associated with mammary epithelial cell exfoliation in dairy cows. J Dairy Sci 2019; 102:2670-85. https://doi.org/10.3168/jds.2018-15398 
  25. Marzano F, Caratozzolo MF, Consiglio A, et al. Plant miRNAs reduce cancer cell proliferation by targeting MALAT1 and NEAT1: a beneficial cross-kingdom interaction. Front Genet 2020; 11:552490. https://doi.org/10.3389/fgene.2020.552490 
  26. Ditano JP, Sakurikar N, Eastman A. Activation of CDC25A phosphatase is limited by CDK2/cyclin A-mediated feedback inhibition. Cell Cycle 2021; 20:1308-19. https://doi.org/10.1080/15384101.2021.1938813 
  27. Tchakarska G, Sola B. The double dealing of cyclin D1. Cell Cycle 2020; 19:163-78. https://doi.org/10.1080/15384101.2019.1706903 
  28. Yu S, Qiao X, Song X, et al. The proliferating cell nuclear antigen (PCNA) is a potential proliferative marker in oyster crassostrea gigas. Fish Shellfish Immunol 2022; 122:306-15. https://doi.org/10.1016/j.fsi.2022.02.018 
  29. Chen K, Hou J, Song Y, et al. Chi-miR-3031 regulates beta-casein via the PI3K/AKT-mTOR signaling pathway in goat mammary epithelial cells (GMECs). BMC Vet Res 2018;14:369. https://doi.org/10.1186/s12917-018-1695-6 
  30. Liu Y, Hou J, Zhang M, et al. circ-016910 sponges miR-574-5p to regulate cell physiology and milk synthesis via MAPK and PI3K/AKT-mTOR pathways in GMECs. J Cell Physiol 2020; 235:4198-216. https://doi.org/10.1002/jcp.29370 
  31. Jaiswal L, Worku M. Recent perspective on cow's milk allergy and dairy nutrition. Crit Rev Food Sci Nutr 2022;62:7503-17. https://doi.org/10.1080/10408398.2021.1915241 
  32. Pereira PC. Milk nutritional composition and its role in human health. Nutrition 2014;30:619-27. https://doi.org/10.1016/j.nut.2013.10.011 
  33. Bian Y, Lei Y, Wang C, et al. Epigenetic regulation of miR-29s affects the lactation activity of dairy cow mammary epithelial cells. J Cell Physiol 2015; 230:2152-63. https://doi.org/10.1002/jcp.24944