DOI QR코드

DOI QR Code

Severe choline deficiency induces alternative splicing aberrance in optimized duck primary hepatocyte cultures

  • Zhao, Lulu (Institute of Feed Research, Chinese Academy of Agricultural Sciences) ;
  • Cai, Hongying (Institute of Feed Research, Chinese Academy of Agricultural Sciences) ;
  • Wu, Yongbao (Institute of Feed Research, Chinese Academy of Agricultural Sciences) ;
  • Tian, Changfu (State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University) ;
  • Wen, Zhiguo (Institute of Feed Research, Chinese Academy of Agricultural Sciences) ;
  • Yang, Peilong (Institute of Feed Research, Chinese Academy of Agricultural Sciences)
  • Received : 2022.02.03
  • Accepted : 2022.04.18
  • Published : 2022.11.01

Abstract

Objective: Choline deficiency, one main trigger for nonalcoholic fatty liver disease (NAFLD), is closely related to lipid metabolism disorder. Previous study in a choline-deficient model has largely focused on gene expression rather than gene structure, especially sparse are studies regarding to alternative splicing (AS). In modern life science research, primary hepatocytes culture technology facilitates such studies, which can accurately imitate liver activity in vitro and show unique superiority. Whereas limitations to traditional hepatocytes culture technology exist in terms of efficiency and operability. This study pursued an optimization culture method for duck primary hepatocytes to explore AS in choline-deficient model. Methods: We performed an optimization culture method for duck primary hepatocytes with multi-step digestion procedure from Pekin duck embryos. Subsequently a NAFLD model was constructed with choline-free medium. RNA-seq and further analysis by rMATS were performed to identify AS events alterations in choline-deficency duck primary hepatocytes. Results: The results showed E13 (embryonic day 13) to E15 is suitable to obtain hepatocytes, and the viability reached over 95% by trypan blue exclusion assay. Primary hepatocyte retained their biological function as well identified by Periodic Acid-Schiff staining method and Glucose-6-phosphate dehydrogenase activity assay, respectively. Meanwhile, genes of alb and afp and specific protein of albumin were detected to verify cultured hepatocytes. Immunofluorescence was used to evaluate purity of hepatocytes, presenting up to 90%. On this base, choline-deficient model was constructed and displayed significantly increase of intracellular triglyceride and cholesterol as reported previously. Intriguingly, our data suggested that AS events in choline-deficient model were implicated in pivotal biological processes as an aberrant transcriptional regulator, of which 16 genes were involved in lipid metabolism and highly enriched in glycerophospholipid metabolism. Conclusion: An effective and rapid protocol for obtaining duck primary hepatocytes was established, by which our findings manifested choline deficiency could induce the accumulation of lipid and result in aberrant AS events in hepatocytes, providing a novel insight into various AS in the metabolism role of choline.

Keywords

Acknowledgement

Conceptualization, Lulu Zhao, Zhiguo Wen, and Peilong Yang; Data curation, Lulu Zhao and Changfu Tian; Funding acquisition, Zhiguo Wen and Peilong Yang; Investigation, Lulu Zhao; Methodology, Lulu Zhao, Hongying Cai and Yongbao Wu; Project administration, Zhiguo Wen and Peilong Yang; Resources, Hongying Cai, Yongbao Wu and Changfu Tian; Writing - original draft, Lulu Zhao; Writing - review & editing, Zhiguo Wen and Peilong Yang. We acknowledge the editors and anonymous reviewers for insightful suggestions on this work.

References

  1. Samuel VT, Shulman GI. Nonalcoholic fatty liver disease as a nexus of metabolic and hepatic diseases. Cell Metab 2018; 27:22-41. https://doi.org/10.1016/j.cmet.2017.08.002
  2. Iqbal U, Perumpail BA, Akhtar D, Kim D, Ahmed A. The epidemiology, risk profiling and diagnostic challenges of nonalcoholic fatty liver disease. Medicines 2019;6:41. https://doi.org/10.3390/medicines6010041
  3. Saadoun A, Leclercq B. In vivo lipogenesis in genetically fat and lean chickens of various ages. Comp Biochem Physiol B 1986;83:607-11. https://doi.org/10.1016/0305-0491(86)90304-4
  4. Shahriari A, Tabatabaie SRF, Jafari RA, Ghorbanzadeh B. Modulation of serum and liver triglyceride and abdominal fat pad weight by dietary garlic in male broilers. Int J Vet Res 2009;3: 101-5.
  5. Lu L, Chen Y, Wang Z, et al. The goose genome sequence leads to insights into the evolution of waterfowl and susceptibility to fatty liver. Genome Biol 2015;16:89. https://doi.org/10.1186/s13059-015-0652-y
  6. Shi Q, Aida K, Vandeberg JL, Wang XL. Passage-dependent changes in baboon endothelial cells-relevance to in vitro aging. DNA Cell Biol 2004;23:502-9. https://doi.org/10.1089/1044549041562294
  7. Takeba Y, Matsumoto N, Takenoshita-Nakaya S, et al. Comparative study of culture conditions for maintaining CYP3A4 and ATP-binding cassette transporters activity in primary cultured human hepatocytes. J Pharmacol Sci 2011;115: 516-24. https://doi.org/10.1254/jphs.10215FP
  8. Rambor E, Craene BD, Kock JD, et al. Strategies for immortalization of primary hepatocytes. J Hepatol 2014;61:925-43. https://doi.org/10.1016/j.jhep.2014.05.046
  9. Gomez-Lechon M, Tolosa L, Conde I, Donato MT. Competency of different cell models to predict human hepatotoxic drugs. Expert Opin Drug Metab Toxicol 2014;10:1553-68. https://doi.org/10.1517/17425255.2014.967680
  10. Selgen PO. Preparation of isolated rat liver cells. Methods Cell Biol 1976;13:29-83. https://doi.org/10.1016/S0091-679X(08)61797-5
  11. Alpini G, Phillips JO, Vroman B, Larusso NF. Recent advances in the isolation of liver cells. Hepatology 1994;20:494-514. https://doi.org/10.1002/hep.1840200231
  12. Ng IC, Zhang L, Shen N, et al. Isolation of primary rat hepatocytes with multiparameter perfusion control. J Vis Exp 2021;5:1-23. https://doi.org/10.3791/62289
  13. Simon J, Nunez-Garcia M, Fernandez-Tussy P, et al. Targeting hepatic glutaminase 1 ameliorates non-alcoholic steatohepatitis by restoring very-low-density lipoprotein triglyceride assembly. Cell Metab 2020;31:605-22. https://doi.org/10.1016/j.cmet. 2020.01.013
  14. Tatsuya Y, Daisuke M, Yoshihiro A, et al. Mitochondrial stasis reveals p62-mediated ubiquitination in parkin-independent mitophagy and mitigates nonalcoholic fatty liver disease. Cell Metab 2018;28:588-604. https://doi.org/10.1016/j.cmet.2018.06.014
  15. Yao L, Zhou J, Dai H, et al. Hepatic leukocyte immunoglobulin-like receptor B4 (LILRB4) attenuates nonalcoholic fatty liver disease via SHP1-TRAF6 pathway. Hepatology 2018;67:1303-19. https://doi.org/10.1002/hep.29633
  16. Ming L, Tan J, He Z, et al. Inhibitory effect of blue honeysuckle extract on high-fat-diet-induced fatty liver in mice. Anim Nutr 2018;4:288-93. https://doi.org/10.1016/j.aninu.2018.06.001
  17. Zeisel SH, Da Costa KA, Franklin PD, et al. Choline, an essential nutrient for humans. FASEB J 1991;5:2093-8. https://doi.org/10.1096/fasebj.5.7.2010061
  18. Hegsted DM, Mills CR, Elvehjem CA, Hart EB. Choline in the nutrition of chicks. J Biol Chem 1941;138:459-66. https://doi.org/10.1111/j.1469-185X.1941.tb01098.x
  19. Sashin N. Improved growth of mammalian and insect cells in media containing increased levels of choline. Appl Microbiol 1969;17:318-9. https://doi.org/10.1128/AEM.17.2.318-319.1969
  20. Olson RE. Scientific contributions of Wendell H. Griffith to our understanding of the function of choline. Fed Proc 1971; 30:131-8.
  21. Demirdjian L, Xu Y, Bahrami-Samani E, et al. Detecting allele-specific alternative splicing from population-scale RNA-Seq data. Am J Hum Genet 2020;107:461-72. https://doi.org/10.1016/j.ajhg.2020.07.005
  22. Hu X, Wang H, Li K, Liu X, Huang C. Genome-wide alternative splicing variation and its potential contribution to maize immature-ear heterosis. Crop J 2020;9:476-86. https://doi.org/10.1016/j.cj.2020.09.003
  23. Ryu J, Galan A, Xin X, et al. APPL1 Potentiates Insulin Sensitivity by Facilitating the Binding of IRS1/2 to the insulin receptor. Cell Rep 2014;7:1227-38. https://doi.org/10.1016/j.celrep.2014.04.006
  24. Galan-Davila AK, Ryu J, Dong K, et al. Alternative splicing variant of the scaffold protein APPL1 suppresses hepatic adiponectin signaling and function. J Biol Chem 2018;293: 6064-74. https://doi.org/10.1074/jbc.RA118.002162
  25. Timothy SW, Weatheritt R, Best AJ, Ha KCH, Blencowe BJ. Efficient and accurate quantitative profiling of alternative splicing patterns of any complexity on a laptop. Mol Cell 2018;72:187-200. https://doi.org/10.1016/j.molcel.2018.08.018
  26. Zheng YA, Kunlong B. Use of MTT assay for the determination of cell viability and proliferation. Immunol J 1992;8:266-9.
  27. Zhao X, Sun K, Lan Z, et al. Tenofovir and adefovir downregulate mitochondrial chaperone TRAP1 and succinate dehydrogenase subunit B to metabolically reprogram glucose metabolism and induce nephrotoxicity. Sci Rep 2017;7: 46344. https://doi.org/10.1038/srep46344
  28. Kim D, Langmead B, Salzberg SL. HISAT: A fast spliced aligner with low memory requirements. Nat Methods 2015; 12:357-60. https://doi.org/10.1038/nmeth.3317
  29. Pertea M, Pertea GM, Antonescu CM, Chang TC, Mendell JT, Salzberg SL. StringTie enables improved reconstruction of a transcriptome from RNA-seq reads. Nat Biotechnol 2015;33:290-5. https://doi.org/10.1038/nbt.3122
  30. Shen S, Park JW, Lu Z, et al. rMATS: Robust and flexible detection of differential alternative splicing from replicate RNA-Seq data. Proc Natl Acad Sci USA 2014;111:5593-601. https://doi.org/10.1073/pnas.1419161111
  31. Scotti MM, Swanson MS. RNA mis-splicing in disease. Nat Rev Genet 2016;17:19-32. https://doi.org/10.1038/nrg.2015.3
  32. Tuttleman JS, Pugh JC, Summers JW. In vitro experimental infection of primary duck hepatocyte cultures with duck hepatitis B virus. J Virol 1986;58:17-25. https://doi.org/10.1016/0166-0934(86)90075-3
  33. Figueiredo N, Matos B, Diniz M, Branco V, Martins M. Marine fish primary hepatocyte isolation and culture: new insights to enzymatic dissociation pancreatin digestion. Int J Environ Res Public Health 2021;18:1380. https://doi.org/10.3390/ijerph18041380
  34. Dobie R, Wilson-Kanamori JR, Henderson BEP, et al. Singlecell transcriptomics uncovers zonation of function in the mesenchyme during liver fibrosis. Cell Rep 2019;29:1832-47. https://doi.org/10.1016/j.celrep.2019.10.024
  35. Kitto LJ, Henderson NC. Hepatic Stellate cell regulation of liver regeneration and repair. Hepatol Commun 2021;5: 358-70. https://doi.org/10.1002/hep4.1628
  36. Chen H, Rui J. Histological and histochemical studies on the liver development of the beijing duck embryo. J Fudan Univ 1992;4:369-74.
  37. Coulais D, Panterne C, Fonteneau JF, Gregoire M. Purification of circulating plasmacytoid dendritic cells using counterflow centrifugal elutriation and immunomagnetic beads. Cytotherapy 2012;14:887-96. https://doi.org/10.3109/14653249.2012.689129
  38. Kim J, Park JE. Porcine epidemic diarrhea virus purification using low-speed centrifugation. J Prev Vet Med 2020;44:28-32. https://doi.org/10.13041/jpvm.2020.44.1.28
  39. Kong QF, Luo FH, Sachula WU, Bao JJ, Liu DY, Ying JW. Enrichment and identification of spermatogonial stem cells from goat testis. China J Anim Vet Sci 2013;44:1554-60. https://doi.org/10.11843/j.issn.0366-6964.2013.10.007
  40. Cho CH, Berthiaume F, Tilles AW, Yarmush ML. A new technique for primary hepatocyte expansion in vitro. Sci Transl Med 2008;101:345-356. https://doi.org/10.1002/bit.21911
  41. Corbin KD, Zeisel SH. Choline metabolism provides novel insights into nonalcoholic fatty liver disease and its progression. Curr Opin Gastroenterol 2012;28:159-65. https://doi.org/10. 1097/MOG.0b013e32834e7b4b https://doi.org/10.1097/MOG.0b013e32834e7b4b
  42. Sanders LM, Zeisel SH. Choline: dietary requirements and role in brain development. Nutr Today 2007;42:181-6. https://doi.org/10.1097/01.NT.0000286155.55343.fa
  43. Celia J, Araujo O, Valentim IB, Andrade K, Goulart M, Longevity C. Choline and cystine deficient diets in animal models with hepatocellular injury: evaluation of oxidative stress and expression of RAGE, TNF-α, and IL-1β. Oxid Med Cell Longev 2015;2015:121925. https://doi.org/10.1155/2015/121925
  44. Kanuri G, Bergheim I. In vitro and in vivo models of nonalcoholic fatty liver disease (NAFLD). Int J Mol Sci 2013;14: 11963-80. https://doi.org/10.3390/ijms140611963
  45. Jie Z, Yang W, Tang Q, Leng Y, Wei C. The effects of choline on hepatic lipid metabolism, mitochondrial function and antioxidative status in human hepatic C3A cells exposed to excessive energy substrates. Nutrients 2014;6:2552-71. https://doi.org/10.3390/nu6072552
  46. Climente-Gonzalez H, Porta-Pardo E, Godzik A, Eyras E. The functional impact of alternative splicing in cancer. Cell Rep 2017;20:2215-26. https://doi.org/10.1101/076653
  47. Huang R, Guo J, Yan P, et al. The construction of bone metastasis-specific prognostic model and co-expressed network of alternative splicing in breast cancer. Front Cell Dev Biol 2020;8:790. https://doi.org/10.3389/fcell.2020.00790
  48. Popli P, Richters MM, Chadchan SB, et al. Splicing factor SF3B1 promotes endometrial cancer progression via regulating KSR2 RNA maturation. Cell Death Dis 2020;11:842. https://doi.org/10.1038/s41419-020-03055-y
  49. Shen SM, Yan J, Cheng Z, et al. Nuclear PTEN safeguards pre-mRNA splicing to link Golgi apparatus for its tumor suppressive role. Nat Commun 2018;9:2392. https://doi.org/10.1038/s41467-018-04760-1
  50. Reddy A, Marquez Y, Kalyna M, Barta K. Complexity of the alternative splicing landscape in plants. Plant Cell 2013;25: 3657-83. https://doi.org/10.1105/tpc.113.117523
  51. Airoldi CA, Bergonzi S, Coen D. Single amino acid change alters the ability to specify male or female organ identity. Proc Natl Acad Sci USA 2010;107:18898-902. https://doi.org/10.1073/pnas.1009050107
  52. Severing EI, Dijk A, Morabito G, Busscher-Lange J, Immink R, Ham R. Predicting the impact of alternative splicing on plant MADS domain protein function. PLoS One 2012;7: e30524. https://doi.org/10.1371/journal.pone.0030524
  53. Chao Y, Jiang Y, Zhong M, Wei K, Zou C. Regulatory roles and mechanisms of alternative RNA splicing in adipogenesis and human metabolic health. Cell Biosci 2021;11:66. https://doi.org/10.1186/s13578-021-00581-w
  54. Bai YL, Zhang F, Zhang HY, Xu C, Xia C. Follicular fluid metabolite changes in dairy cows with inactive ovary identified using untargeted metabolomics. Biomed Res Int 2020;2020: 9837543. https://doi.org/10.1155/2020/9837543
  55. Luco RF, Allo M, Schor IE, Kornblihtt AR, Misteli T. Epigenetics in alternative pre-mRNA Splicing. Cell 2011;144:16-26. https://doi.org/10.1016/j.cell.2010.11.056