The PPARα/CYP4A14 bile acid pathway is associated with lipid metabolism disorders caused by low birth weight with high-fat diet

  • Fei Zhou Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China; Key Laboratory of Metabolic Diseases, Hebei General Hospital, Shijiazhuang, China
  • Linquan Yang Key Laboratory of Metabolic Diseases, Hebei General Hospital, Shijiazhuang, China
  • Wenwen Sun Department of Internal Medicine, North China University of Science and Technology, Tangshan, China
  • Xing Wang Key Laboratory of Metabolic Diseases, Hebei General Hospital, Shijiazhuang, China
  • Na Guo Key Laboratory of Metabolic Diseases, Hebei General Hospital, Shijiazhuang, China
  • Huijuan Ma Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China; Key Laboratory of Metabolic Diseases, Hebei General Hospital, Shijiazhuang, China; Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China
  • Linlin Yang Key Laboratory of Metabolic Diseases, Hebei General Hospital, Shijiazhuang, China
Keywords: LBW, lipid metabolism disorders, ACOX2, bile acid, LC-MS/MS

Abstract

Purpose: To investigate possible mechanisms underlying the greater susceptibility of lipid metabolism disorders in low birth weight (LBW) mice fed with high-fat diets (HFDs).

Methods: LBW mice model was established by using the pregnancy malnutrition method. Male pups were selected from LBW and normal-birth weight (NBW) offspring at random. After 3 weeks of weaning, all offspring mice were fed with HFD. Serum triglycerides (TGs), cholesterol (TC), low density lipoprotein (LDL-C), total bile acid (TAB), non-esterified fatty acid (NEFA), and mice fecal bile acid profiles were measured. Lipid deposition in liver sections was visualized by Oil Red O staining. The weight ratio of liver, muscle, and adiposity was calculated. Tandem mass tag (TMT) combined with LC-MS/MS was used to determine the differentially expressed proteins (DEPs) of liver tissue in two groups. Bioinformatics was used for further analysis of DEPs to screen key target proteins, and then Western Blot (WB) and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) were performed to validate the expressions of DEPs.

Results: LBW mice fed with HFD showed more severe lipid metabolism disorders in the childhood. In contrast to the NBW group, the serum bile acids and fecal ω-muricholic acid (ω-MCA) levels in the LBW group were significantly lower. LC-MS/MS analysis showed that downregulated proteins were associated with lipid metabolism, and further analysis found that these proteins are mainly concentrated in peroxisome proliferation-activated receptor (PPAR) and primary bile acid synthesis signaling pathways and are involved in cellular processes and metabolic processes through binding and catalytic functions. Bioinformatics analysis indicated that the level of Cytochrome P450 Family 46 Subfamily A Member 1 (CYP46A1), PPARα, key factors of cholesterol metabolism and bile acid synthesis, as well as downstream molecules Cytochrome P450 Family 4 Subfamily A Member 14 (CYP4A14), and Acyl-Coenzyme A Oxidase 2 (ACOX2) are markedly different in the liver of LBW individuals fed with HFD, and confirmed by WB and RT-qPCR.

Conclusion: LBW mice are more prone to dyslipidemia probably due to downregulated bile acid metabolism-related PPARα/CYP4A14 pathway, resulting in insufficient metabolism of cholesterol to bile acids, which, in turn, leads to elevated blood cholesterol.

Downloads

Download data is not yet available.

References


1.
Pauwels S, Ghosh M, Duca RC, Bekaert B, Freson K, Huybrechts I, et al. Dietary and supplemental maternal methyl-group donor intake and cord blood DNA methylation. Epigenetics 2017; 12(1): 1–10. doi: 10.1080/15592294.2016.1257450


2.
Bleu J, Massot M, Haussy C, Meylan S. Experimental litter size reduction reveals costs of gestation and delayed effects on offspring in a viviparous lizard. Proc Biol Sci 2012; 279(1728): 489–98. doi: 10.1098/rspb.2011.0966


3.
Desai M, Han G, Li T, Ross MG. Programmed epigenetic DNA methylation-mediated reduced neuroprogenitor cell proliferation and differentiation in small-for-gestational-age offspring. Neuroscience 2019; 412: 60–71. doi: 10.1016/j.neuroscience.2019.05.044


4.
Fabricius-Bjerre S, Jensen RB, Færch K, Larsen T, Mølgaard C, Michaelsen KF, et al. Impact of birth weight and early infant weight gain on insulin resistance and associated cardiovascular risk factors in adolescence. PLoS One 2011; 6(6): e20595.


5.
Ijzerman RG, Stehouwer CD, Van Weissenbruch MM, De Geus EJ, Boomsma DI. Evidence for genetic factors explaining the association between birth weight and low-density lipoprotein cholesterol and possible intrauterine factors influencing the association between birth weight and high-density lipoprotein cholesterol: analysis in twins. J Clin Endocrinol Metab 2001; 86(11): 5479–84. doi: 10.1210/jcem.86.11.7996


6.
Cirigliano A, Amelina A, Biferali B, Macone A, Mozzetta C, Bianchi MM, et al. Statins interfere with the attachment of S. cerevisiae mtDNA to the inner mitochondrial membrane. J Enzyme Inhib Med Chem 2020; 35(1): 129–37. doi: 10.1080/14756366.2019.1687461


7.
Wang S, Yao Y, Rao C, Zheng G, Chen W. 25-HC decreases the sensitivity of human gastric cancer cells to 5-fluorouracil and promotes cells invasion via the TLR2/NF-κB signaling pathway. Int J Oncol 2019; 54(3): 966–80. doi: 10.3892/ijo.2019.4684


8.
Nishitani S, Fukuhara A, Shin J, Okuno Y, Otsuki M, Shimomura I. Metabolomic and microarray analyses of adipose tissue of dapagliflozin-treated mice, and effects of 3-hydroxybutyrate on induction of adiponectin in adipocytes. Sci Rep 2018; 8(1): 8805. doi: 10.1038/s41598-018-27181-y


9.
Schramm G, Surmann EM, Wiesberg S, Oswald M, Reinelt G, Eils R, et al. Analyzing the regulation of metabolic pathways in human breast cancer. BMC Med Genom 2010; 3: 39. doi: 10.1186/1755-8794-3-39


10.
Dongfeng D, An C, Shujia P, Jikai Y, Tao Y, Rui D, et al. Explanation of colon cancer pathophysiology through analyzing the disrupted homeostasis of bile acids. Afr Health Sci 2014; 14(4): 925–8. doi: 10.4314/ahs.v14i4.22


11.
Wang Y, Gunewardena S, Li F, Matye DJ, Chen C, Chao X, et al. An FGF15/19-TFEB regulatory loop controls hepatic cholesterol and bile acid homeostasis. Nat Commun 2020; 11(1): 3612. doi: 10.1038/s41467-020-17363-6


12.
Sissung TM, Deeken J, Leibrand CR, Price DK, Ehrlich S, Steinberg SM, et al. Identification of novel SNPs associated with risk and prognosis in patients with castration-resistant prostate cancer. Pharmacogenomics 2016; 17(18): 1979–86. doi: 10.2217/pgs-2016-0134


13.
Du JE, You YA, Kwon EJ, Kim SM, Lee J, Han KH, et al. Maternal malnutrition affects hepatic metabolism through decreased hepatic taurine levels and changes in HNF4A methylation. Int J Mol Sci 2020; 21(23): 9060. doi: 10.3390/ijms21239060


14.
Fall CH, Sachdev HS, Osmond C, Lakshmy R, Biswas SD, Prabhakaran D, et al. Adult metabolic syndrome and impaired glucose tolerance are associated with different patterns of BMI gain during infancy: Data from the New Delhi Birth Cohort. Diabetes Care 2008; 31(12): 2349–56. doi: 10.2337/dc08-0911


15.
Patterson WB, Glasson J, Naik N, Jones RB, Berger PK, Plows JF, et al. Prenatal exposure to ambient air pollutants and early infant growth and adiposity in the Southern California Mother’s Milk Study. Environ Health 2021; 20(1): 67. doi: 10.1186/s12940-021-00753-8


16.
Rosario FJ, Nathanielsz PW, Powell TL, Jansson T. Maternal folate deficiency causes inhibition of mTOR signaling, down-regulation of placental amino acid transporters and fetal growth restriction in mice. Sci Rep 2017; 7(1): 3982. doi: 10.1038/s41598-017-03888-2


17.
Zhang L, Zhang J, Yan E, He J, Zhong X, Zhang L, et al. Dietary supplemented curcumin improves meat quality and antioxidant status of intrauterine growth retardation growing pigs via Nrf2 signal pathway. Animals 2020; 10(3): 539. doi: 10.3390/ani10030539


18.
Al Salmi I, Hannawi S. Birthweight and lipids in adult life: population-based cross sectional study. Lipids 2020; 55(4): 365–74. doi: 10.1002/lipd.12242


19.
Brøns C, Thuesen ACB, Elingaard-Larsen LO, Justesen L, Jensen RT, Henriksen NS, et al. Increased liver fat associates with severe metabolic perturbations in low birth weight men. Eur J Endocrinol 2022; 186(5): 511–21. doi: 10.1530/EJE-21-1221


20.
Küster A, Croyal M, Moyon T, Darmaun D, Ouguerram K, Ferchaud-Roucher V. Characterization of lipoproteins and associated lipidome in very preterm infants: a pilot study. Pediatr Res 2022; null(null): null. doi: 10.1038/s41390-022-02159-9


21.
Sarr O, Mathers KE, Vanderboor C, Wiggers K, Devgan A, Hardy DB, et al. Sex-specific alterations in hepatic cholesterol metabolism in low birth weight adult guinea pigs. Pediatr Res 2022; 91(5): 1078–89. doi: 10.1038/s41390-021-01491-w


22.
Nicklasson M, Sjöling Å, von Mentzer A, Qadri F, Svennerholm AM. Expression of colonization factor CS5 of enterotoxigenic Escherichia coli (ETEC) is enhanced in vivo and by the bile component Na glycocholate hydrate. PLoS One 2012; 7(4): e35827. doi: 10.1371/journal.pone.0035827


23.
Ushiroda C, Naito Y, Takagi T, Uchiyama K, Mizushima K, Higashimura Y, et al. Green tea polyphenol (epigallocatechin-3-gallate) improves gut dysbiosis and serum bile acids dysregulation in high-fat diet-fed mice. J Clin Biochem Nutr 2019; 65(1): 34–46. doi: 10.3164/jcbn.18-116


24.
Berger JM, Moon YA. Increased hepatic lipogenesis elevates liver cholesterol content. Mol Cells 2021; 44(2): 116–25. doi: 10.14348/molcells.2021.2147


25.
Lin YC, Huang HY, Shrestha S, Chou CH, Chen YH, Chen CR, et al. Multi-omics profiling reveals microRNA-mediated insulin signaling networks. BMC Bioinform 2020; 21(Suppl. 13): 389. doi: 10.1186/s12859-020-03678-0


26.
Moutinho M, Nunes MJ, Correia JC, Gama MJ, Castro-Caldas M, Cedazo-Minguez A, et al. Neuronal cholesterol metabolism increases dendritic outgrowth and synaptic markers via a concerted action of GGTase-I and Trk. Sci Rep 2016; 6: 30928. doi: 10.1038/srep30928


27.
Hernaiz-Llorens M, Roselló-Busquets C, Durisic N, Filip A, Ulloa F, Martínez-Mármol R, et al. Growth cone repulsion to Netrin-1 depends on lipid raft microdomains enriched in UNC5 receptors. Cell Mol Life Sci 2021; 78(6): 2797–820. doi: 10.1007/s00018-020-03663-z


28.
Gu M, Zhang Y, Fan S, Ding X, Ji G, Huang C. Extracts of Rhizoma polygonati odorati prevent high-fat diet-induced metabolic disorders in C57BL/6 mice. PLoS One 2013; 8(11): e81724. doi: 10.1371/journal.pone.0081724


29.
Matsushita Y, Ogawa D, Wada J, Yamamoto N, Shikata K, Sato C, et al. Activation of peroxisome proliferator-activated receptor delta inhibits streptozotocin-induced diabetic nephropathy through anti-inflammatory mechanisms in mice. Diabetes 2011; 60(3): 960–8. doi: 10.2337/db10-1361


30.
Choi SY, Kim TH, Hong MW, Park TS, Lee H, Lee SJ. Transcriptomic alterations induced by aflatoxin B1 and ochratoxin A in LMH cell line. Poult Sci 2020; 99(11): 5265–74. doi: 10.1016/j.psj.2020.05.058


31.
Chen Q, Qiu F, Zhou K, Matlock HG, Takahashi Y, Rajala RVS, et al. Pathogenic role of microRNA-21 in diabetic retinopathy through down regulation of PPARα. Diabetes 2017; 66(6): 1671–82. doi: 10.2337/db16-1246


32.
Jia Y, Wu C, Kim YS, Yang SO, Kim Y, Kim JS, et al. A dietary anthocyanin cyanidin-3-O-glucoside binds to PPARs to regulate glucose metabolism and insulin sensitivity in mice. Commun Biol 2020; 3(1): 514. doi: 10.1038/s42003-020-01231-6


33.
Nakagawa Y, Shimano H. CREBH regulates systemic glucose and lipid metabolism. Int J Mol Sci 2018; 19(5): 1396. doi: 10.3390/ijms19051396


34.
Huang J, Wang Y, Ying C, Liu L, Lou Z. Effects of mulberry leaf on experimental hyperlipidemia rats induced by high-fat diet. Exp Ther Med 2018; 16(2): 547–56. doi: 10.3892/etm.2018.6254


35.
Xin S, Zhan Q, Chen X, Xu J, Yu Y. Efficacy of serum miRNA test as a non-invasive method to diagnose nonalcoholic steatohepatitis: a systematic review and meta-analysis. BMC Gastroenterol 2020; 20(1): 186. doi: 10.1186/s12876-020-01334-8


36.
Zhao M, Zhao H, Deng J, Guo L, Wu B. Role of the CLOCK protein in liver detoxification. Br J Pharmacol 2019; 176(24): 4639–52. doi: 10.1111/bph.14828


37.
Selwyn FP, Cheng SL, Klaassen CD, Cui JY. Regulation of hepatic drug-metabolizing enzymes in germ-free mice by conventionalization and probiotics. Drug Metab Dispos 2016; 44(2): 262–74. doi: 10.1124/dmd.115.067504


38.
Li CY, Cheng SL, Bammler TK, Cui JY. Editor’s highlight: neonatal activation of the xenobiotic-sensors PXR and CAR results in acute and persistent down-regulation of PPARα-signaling in mouse liver. Toxicol Sci 2016; 153(2): 282–302. doi: 10.1093/toxsci/kfw127


39.
Zhang Q, Zhang Y, Sun S, Wang K, Qian J, Cui Z, et al. ACOX2 is a prognostic marker and impedes the progression of hepatocellular carcinoma via PPARα pathway. Cell Death Disease 2021; 12(1): 15. doi: 10.1038/s41419-020-03291-2


40.
Zhang Y, Chen Y, Zhang Z, Tao X, Xu S, Zhang X, et al. Acox2 is a regulator of lysine crotonylation that mediates hepatic metabolic homeostasis in mice. Cell Death Dis 2022; 13(3): 279. doi: 10.1038/s41419-022-04725-9


41.
Monte MJ, Alonso-Peña M, Briz O, Herraez E, Berasain C, Argemi J, et al. ACOX2 deficiency: An inborn error of bile acid synthesis identified in an adolescent with persistent hypertransaminasemia. J Hepatol 2017; 66(3): 581–8. doi: 10.1016/j.jhep.2016.11.005
Published
2023-01-24
How to Cite
Zhou F., Yang L., Sun W., Wang X., Guo N., Ma H., & Yang L. (2023). The PPARα/CYP4A14 bile acid pathway is associated with lipid metabolism disorders caused by low birth weight with high-fat diet. Food & Nutrition Research, 67. https://doi.org/10.29219/fnr.v67.8994
Section
Original Articles