DOI QR코드

DOI QR Code

Maternal betaine supplementation ameliorates fatty liver disease in offspring mice by inhibiting hepatic NLRP3 inflammasome activation

  • Lun Li (Department of Delivery Room, Guangzhou Women and Children's Medical Center) ;
  • Liuqiao Sun (Department of Maternal, Child and Adolescent Health, School of Medicine, Jinan University) ;
  • Xiaoping Liang (Department of Nutrition, School of Medicine, Jinan University) ;
  • Qian Ou (Department of Nutrition, School of Medicine, Jinan University) ;
  • Xuying Tan (Department of Child Health Care, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health) ;
  • Fangyuan Li (Department of Nutrition, School of Medicine, Jinan University) ;
  • Zhiwei Lai (Department of Nutrition, School of Medicine, Jinan University) ;
  • Chenghe Ding (Department of Nutrition, School of Medicine, Jinan University) ;
  • Hangjun Chen (Department of Nutrition, School of Medicine, Jinan University) ;
  • Xinxue Yu (Department of Nutrition, School of Medicine, Jinan University) ;
  • Qiongmei Wu (Department of Nutrition, School of Medicine, Jinan University) ;
  • Jun Wei (Department of Science and Technology, Guangzhou Customs) ;
  • Feng Wu (Department of Science and Technology, Guangzhou Customs) ;
  • Lijun Wang (Department of Nutrition, School of Medicine, Jinan University)
  • Received : 2023.04.14
  • Accepted : 2023.08.14
  • Published : 2023.12.01

Abstract

BACKGROUND/OBJECTIVES: Previous research has shown maternal betaine supplementation alleviates fetal-derived hepatic steatosis. Therefore, this study examined the anti-inflammatory effect of maternal betaine intake in offspring mice and its mechanism. MATERIALS/METHODS: Female C57BL/6J mice and their offspring were randomly divided into 3 groups according to the treatment received during gestation and lactation: control diet (CD), fatty liver disease (FLD), and fatty liver disease + 1% betaine (FLD-BET). The FLD group was given a high-fat diet and streptozotocin (HFD + STZ), and the FLD-BET group was treated with HFD + STZ + 1% betaine. After weaning, the offspring mice were given a normal diet for 5 weeks and then dissected to measure the relevant indexes. RESULTS: Compared to the CD group, the offspring mice in the FLD group revealed obvious hepatic steatosis and increased serum levels of alanine aminotransferase, interleukin (IL)-6, and tumor necrosis factor (TNF)-α; maternal betaine supplementation reversed these changes. The hepatic mRNA expression levels of IL-6, IL-18, and Caspase-1 were significantly higher in the FLD group than in the CD group. Maternal betaine supplementation reduced the expression of IL-1β, IL-6, IL-18, and apoptosis-associated speck-like protein containing C-terminal caspase recruitment domain (ASC). Maternal betaine supplementation also reversed the increasing protein expressions of nitric oxide dioxygenase-like receptor family pyrin domain containing 3 (NLRP3), ASC, Caspase-1, IL-1β, and IL-18 in offspring mice exposed to HFD + STZ. Maternal betaine supplementation decreased the homocysteine (Hcy) and s-adenosine homocysteine (SAH) levels significantly in the livers. Furthermore, the hepatic Hcy concentrations showed significant inverse relationships with the mRNA expression of TNF-α, NLRP3, ASC, and IL-18. The hepatic SAH concentration was inversely associated with the IL-1β mRNA expression. CONCLUSIONS: The lipotropic and anti-inflammatory effect of maternal betaine supplementation may be associated with the inhibition of NLRP3 inflammasome in the livers of the offspring mice.

Keywords

Acknowledgement

This research was funded by the Guangdong Basic and Applied Basic Research Foundation (Grant number: 2023A1515030168) and the Scientific Research Fund of China Nutrition Society (Grant numbers: CNS-HPNK2021-44). The funder did not participate in our article.

References

  1. Saiman Y, Duarte-Rojo A, Rinella ME. Fatty liver disease: diagnosis and stratification. Annu Rev Med 2022;73:529-44. https://doi.org/10.1146/annurev-med-042220-020407
  2. Campisano S, La Colla A, Echarte SM, Chisari AN. Interplay between early-life malnutrition, epigenetic modulation of the immune function and liver diseases. Nutr Res Rev 2019;32:128-45. https://doi.org/10.1017/S0954422418000239
  3. Li M, Reynolds CM, Segovia SA, Gray C, Vickers MH. Developmental programming of nonalcoholic fatty liver disease: the effect of early life nutrition on susceptibility and disease severity in later life. BioMed Res Int 2015;2015:437107.
  4. Hagstrom H, Simon TG, Roelstraete B, Stephansson O, Soderling J, Ludvigsson JF. Maternal obesity increases the risk and severity of NAFLD in offspring. J Hepatol 2021;75:1042-8. https://doi.org/10.1016/j.jhep.2021.06.045
  5. Brumbaugh DE, Tearse P, Cree-Green M, Fenton LZ, Brown M, Scherzinger A, Reynolds R, Alston M, Hoffman C, Pan Z, et al. Intrahepatic fat is increased in the neonatal offspring of obese women with gestational diabetes. J Pediatr 2013;162:930-6.e1. https://doi.org/10.1016/j.jpeds.2012.11.017
  6. Henao-Mejia J, Elinav E, Jin C, Hao L, Mehal WZ, Strowig T, Thaiss CA, Kau AL, Eisenbarth SC, Jurczak MJ, et al. Inflammasome-mediated dysbiosis regulates progression of NAFLD and obesity. Nature 2012;482:179-85. https://doi.org/10.1038/nature10809
  7. Wree A, Eguchi A, McGeough MD, Pena CA, Johnson CD, Canbay A, Hoffman HM, Feldstein AE. NLRP3 inflammasome activation results in hepatocyte pyroptosis, liver inflammation, and fibrosis in mice. Hepatology 2014;59:898-910. https://doi.org/10.1002/hep.26592
  8. Dwivedi DK, Jena GB. NLRP3 inhibitor glibenclamide attenuates high-fat diet and streptozotocin-induced non-alcoholic fatty liver disease in rat: studies on oxidative stress, inflammation, DNA damage and insulin signalling pathway. Naunyn Schmiedebergs Arch Pharmacol 2020;393:705-16. https://doi.org/10.1007/s00210-019-01773-5
  9. Zhang Y, Wen J, Liu D, Qiu Z, Zhu Q, Li R, Zhang Y. Demethylenetetrahydroberberine alleviates nonalcoholic fatty liver disease by inhibiting the NLRP3 inflammasome and oxidative stress in mice. Life Sci 2021;281:119778.
  10. Wang LJ, Zhang HW, Zhou JY, Liu Y, Yang Y, Chen XL, Zhu CH, Zheng RD, Ling WH, Zhu HL. Betaine attenuates hepatic steatosis by reducing methylation of the MTTP promoter and elevating genomic methylation in mice fed a high-fat diet. J Nutr Biochem 2014;25:329-36. https://doi.org/10.1016/j.jnutbio.2013.11.007
  11. Wang L, Chen L, Tan Y, Wei J, Chang Y, Jin T, Zhu H. Betaine supplement alleviates hepatic triglyceride accumulation of apolipoprotein E deficient mice via reducing methylation of peroxisomal proliferator-activated receptor alpha promoter. Lipids Health Dis 2013;12:34.
  12. Sun L, Tan X, Liang X, Chen H, Ou Q, Wu Q, Yu X, Zhao H, Huang Q, Yi Z, et al. Maternal betaine supplementation mitigates maternal high fat diet-induced NAFLD in offspring mice through gut microbiota. Nutrients 2023;15:284.
  13. Zhao G, He F, Wu C, Li P, Li N, Deng J, Zhu G, Ren W, Peng Y. Betaine in inflammation: mechanistic aspects and applications. Front Immunol 2018;9:1070.
  14. Ge CX, Yu R, Xu MX, Li PQ, Fan CY, Li JM, Kong LD. Betaine prevented fructose-induced NAFLD by regulating LXRα/PPARα pathway and alleviating ER stress in rats. Eur J Pharmacol 2016;770:154-64. https://doi.org/10.1016/j.ejphar.2015.11.043
  15. Szkudelski T. The mechanism of alloxan and streptozotocin action in B cells of the rat pancreas. Physiol Res 2001;50:537-46.
  16. Dong X, Lin D, Sheng J, Xie Y. Intrauterine hyperglycemia induces liver inflammation in mouse male offspring. Int Immunopharmacol 2021;99:107974.
  17. Lo L, McLennan SV, Williams PF, Bonner J, Chowdhury S, McCaughan GW, Gorrell MD, Yue DK, Twigg SM. Diabetes is a progression factor for hepatic fibrosis in a high fat fed mouse obesity model of nonalcoholic steatohepatitis. J Hepatol 2011;55:435-44. https://doi.org/10.1016/j.jhep.2010.10.039
  18. Santhekadur PK, Kumar DP, Sanyal AJ. Preclinical models of non-alcoholic fatty liver disease. J Hepatol 2018;68:230-7. https://doi.org/10.1016/j.jhep.2017.10.031
  19. Kleiner DE, Brunt EM, Van Natta M, Behling C, Contos MJ, Cummings OW, Ferrell LD, Liu YC, Torbenson MS, Unalp-Arida A, et al. Design and validation of a histological scoring system for nonalcoholic fatty liver disease. Hepatology 2005;41:1313-21. https://doi.org/10.1002/hep.20701
  20. Hu Y, Sun Q, Hu Y, Hou Z, Zong Y, Omer NA, Abobaker H, Zhao R. Corticosterone-induced lipogenesis activation and lipophagy inhibition in chicken liver are alleviated by maternal betaine supplementation. J Nutr 2018;148:316-25. https://doi.org/10.1093/jn/nxx073
  21. Joselit Y, Nanobashvili K, Jack-Roberts C, Greenwald E, Malysheva OV, Caudill MA, Saxena A, Jiang X. Maternal betaine supplementation affects fetal growth and lipid metabolism of high-fat fed mice in a temporal-specific manner. Nutr Diabetes 2018;8:41.
  22. Zhao N, Yang S, Jia Y, Sun B, He B, Zhao R. Maternal betaine supplementation attenuates glucocorticoid-induced hepatic lipid accumulation through epigenetic modification in adult offspring rats. J Nutr Biochem 2018;54:105-12. https://doi.org/10.1016/j.jnutbio.2017.12.003
  23. Soderborg TK, Clark SE, Mulligan CE, Janssen RC, Babcock L, Ir D, Young B, Krebs N, Lemas DJ, Johnson LK, et al. The gut microbiota in infants of obese mothers increases inflammation and susceptibility to NAFLD. Nat Commun 2018;9:4462.
  24. Knorr J, Wree A, Tacke F, Feldstein AE. The NLRP3 inflammasome in alcoholic and nonalcoholic steatohepatitis. Semin Liver Dis 2020;40:298-306. https://doi.org/10.1055/s-0040-1708540
  25. Seo YY, Cho YK, Bae JC, Seo MH, Park SE, Rhee EJ, Park CY, Oh KW, Park SW, Lee WY. Tumor necrosis factor-α as a predictor for the development of nonalcoholic fatty liver disease: a 4-year follow-up study. Endocrinol Metab 2013;28:41-5. https://doi.org/10.3803/EnM.2013.28.1.41
  26. Wu Q, Bazzini AA. Translation and mRNA stability control. Annu Rev Biochem 2023;92:227-45. https://doi.org/10.1146/annurev-biochem-052621-091808
  27. Vogel C, Marcotte EM. Insights into the regulation of protein abundance from proteomic and transcriptomic analyses. Nat Rev Genet 2012;13:227-32. https://doi.org/10.1038/nrg3185
  28. Kim DH, Kim SM, Lee B, Lee EK, Chung KW, Moon KM, An HJ, Kim KM, Yu BP, Chung HY. Effect of betaine on hepatic insulin resistance through FOXO1-induced NLRP3 inflammasome. J Nutr Biochem 2017;45:104-14. https://doi.org/10.1016/j.jnutbio.2017.04.014
  29. Wei M, Wang L, Wu T, Xi J, Han Y, Yang X, Zhang D, Fang Q, Tang B. NLRP3 activation was regulated by DNA methylation modification during mycobacterium tuberculosis infection. BioMed Res Int 2016;2016:4323281.
  30. Wang R, Wang Y, Mu N, Lou X, Li W, Chen Y, Fan D, Tan H. Activation of NLRP3 inflammasomes contributes to hyperhomocysteinemia-aggravated inflammation and atherosclerosis in apoE-deficient mice. Lab Invest 2017;97:922-34. https://doi.org/10.1038/labinvest.2017.30
  31. Conley SM, Abais-Battad JM, Yuan X, Zhang Q, Boini KM, Li PL. Contribution of guanine nucleotide exchange factor Vav2 to NLRP3 inflammasome activation in mouse podocytes during hyperhomocysteinemia. Free Radic Biol Med 2017;106:236-44. https://doi.org/10.1016/j.freeradbiomed.2017.02.027
  32. Jacobsen DW. Hyperhomocysteinemia and oxidative stress: time for a reality check? Arterioscler Thromb Vasc Biol 2000;20:1182-4. https://doi.org/10.1161/01.ATV.20.5.1182
  33. Schroder K, Tschopp J. The inflammasomes. Cell 2010;140:821-32. https://doi.org/10.1016/j.cell.2010.01.040