Maternal Dietary Betaine Prevents High-Fat Diet-Induced Metabolic Disorders and Gut Microbiota Alterations in Mouse Dams and Offspring From Young to Adult

Jieying Liu, Lu Ding, Xiao Zhai, Dongmei Wang, Cheng Xiao, Xiangyi Hui, Tianshu Sun, Miao Yu, Qian Zhang, Ming Li, Xinhua Xiao, Jieying Liu, Lu Ding, Xiao Zhai, Dongmei Wang, Cheng Xiao, Xiangyi Hui, Tianshu Sun, Miao Yu, Qian Zhang, Ming Li, Xinhua Xiao

Abstract

Early life is a critical window for preventing the intergenerational transmission of metabolic diseases. Betaine has been proven to play a role in improving glucose and lipid metabolism disorders in animal models. However, whether maternal betaine supplementation plays a role in regulating gut microbiota in both dams and offspring remains unclear. In this study, C57BL/6 female mice were fed with control diet (Ctr), high-fat diet (HF), and high-fat with betaine supplementation (0.3% betaine in the diet, HFB) from 3 weeks prior to mating and lasted throughout pregnancy and lactation. After weaning, the offspring got free access to normal chow diet until 20 weeks of age. We found that maternal dietary betaine supplementation significantly improved glucose and insulin resistance, as well as reduced free fatty acid (FFA) concentration in dams and offspring from young to adult. When compared to the HF group, Intestinimonas and Acetatifactor were reduced by betaine supplementation in dams; Desulfovibrio was reduced in 4-week-old offspring of the HFB group; and Lachnoclostridium was enriched in 20-week-old offspring of the HFB group. Moreover, the persistent elevated genus Romboutsia in both dams and offspring in the HFB group was reported for the first time. Overall, maternal betaine could dramatically alleviate the detrimental effects of maternal overnutrition on metabolism in both dams and offspring. The persistent alterations in gut microbiota might play critical roles in uncovering the intergenerational metabolic benefits of maternal betaine, which highlights evidence for combating generational metabolic diseases.

Keywords: betaine; dams and offspring; glucose and lipid metabolism; gut microbiota; high-fat diet; intergeneration.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2022 Liu, Ding, Zhai, Wang, Xiao, Hui, Sun, Yu, Zhang, Li and Xiao.

Figures

FIGURE 1
FIGURE 1
Betaine improves glucose and lipid metabolism in high-fat diet dams. Dams were analyzed after weaning. (A) General picture of body composition; (B) body weight (left) and body composition (right) changes during the betaine treatment; (C) oral glucose tolerance test (left) and the area under the curve (right); (D) fasting plasma glucose; (E) fasting insulin level; (F) HOMA-IR; (G) FFAs. Ctr, standard control diet; HF, high-fat diet; HFB, high-fat diet with betaine; HOMA-IR, homeostasis model assessment of insulin resistance; FFAs, free fatty acids. Data are expressed as means ± S.E.M. (n = 4–6/group). One-way and two-way ANOVA; *p < 0.05 and **p < 0.01 vs. Ctr, #p < 0.05 and ##p < 0.01 vs. HF.
FIGURE 2
FIGURE 2
Betaine alters the gut microbiota in high-fat diet dams. The cecal contents were collected from dams after weaning. (A) Shannon index (left) and Simpson index (right); (B) PCoA plots of gut microbiome; (C) relative abundance of the top 10 species at the genus level; (D) relative abundance of Intestinimonas; (E) relative abundance of Acetatifactor; (F) relative abundance of Romboutsia; (G) distinct taxa identified in the three groups using LEfSe analysis. The colors represent the group in which the indicated taxa is more abundant compared to the other group. Ctr, standard control diet; HF, high-fat diet; HFB, high-fat diet with betaine. Data are expressed as means ± S.E.M. (n = 4–6/group). One-way ANOVA; **p < 0.01 vs. Ctr, ##p < 0.01 vs. HF.
FIGURE 3
FIGURE 3
Maternal betaine ameliorates glucose and lipid metabolism induced by early-life high-fat diet in 4-week-old offspring. Offspring mice were analyzed at 4 weeks of age. (A) General picture of body composition; (B) body weight (left) and body composition (right) changes; (C) oral glucose tolerance test (left) and the area under the curve (right); (D) insulin tolerance test (left) and the area under the curve (right); (E) fasting plasma glucose; (F) fasting insulin level; (G) HOMA-IR; (H) FFAs. Ctr.4, 4-week-old offspring of dams fed with the standard control diet; HF.4, 4-week-old offspring of dams fed with the high-fat diet; HFB.4, 4-week-old offspring of dams fed with the high-fat diet with betaine. HOMA-IR, homeostasis model assessment of insulin resistance; FFAs, free fatty acids. Data are expressed as means ± S.E.M. (n = 4–6/group). One-way and two-way ANOVA; *p < 0.05 and **p < 0.01 vs. Ctr, #p < 0.05 and ##p < 0.01 vs. HF.
FIGURE 4
FIGURE 4
Maternal betaine alters the gut microbiota in early-life high-fat diet offspring at 4 weeks of age. The cecal contents were collected from offspring at 4 weeks of age. (A) Shannon index (left) and Simpson index (right); (B) PCoA plots of gut microbiome; (C) relative abundance of the top 10 species at the genus level; (D) relative abundance of Desulfovibrio; (E) relative abundance of Romboutsia; (F) distinct taxa identified in the three groups using LEfSe analysis. The colors represent the group in which the indicated taxa is more abundant compared to the other group. Ctr.4, 4-week-old offspring of dams fed with the standard control diet; HF.4, 4-week-old offspring of dams fed with the high-fat diet; HFB.4, 4-week-old offspring of dams fed with the high-fat diet with betaine. Data are expressed as means ± S.E.M. (n = 6–8/group). One-way ANOVA; *p < 0.05 vs. Ctr, #p < 0.05 vs. HF.
FIGURE 5
FIGURE 5
Maternal betaine protects 20-week-old offspring from glucose and lipid metabolic disorders induced by early-life high-fat diet. Offspring mice were analyzed at 20 weeks of age. (A) General picture of body composition; (B) body weight (left) and body composition (right) changes; (C) oral glucose tolerance test (left) and the area under the curve (right); (D) insulin tolerance test (left) and the area under the curve (right); (E) fasting plasma glucose; (F) fasting insulin level; (G) HOMA-IR; (H) FFAs. Ctr.20, 20-week-old offspring of dams fed with the standard control diet; HF.20, 20-week-old offspring of dams fed with the high-fat diet; HFB.20, 20-week-old offspring of dams fed with the high-fat diet with betaine. HOMA-IR, homeostasis model assessment of insulin resistance; FFAs, free fatty acids. Data are expressed as means ± S.E.M. (n = 5–8/group). One-way and two-way ANOVA; **p < 0.01 vs. Ctr, #p < 0.05 vs. HF.
FIGURE 6
FIGURE 6
Maternal betaine alters the gut microbiota in early-life high-fat diet offspring at 20 weeks of age. The cecal contents were collected from offspring at 20 weeks of age. (A) Shannon index (left) and Simpson index (right); (B) PCoA plots of gut microbiome; (C) relative abundance of the top 10 species at the genus level; (D) relative abundance of Desulfovibrio; (E) relative abundance of Romboutsia; (F) distinct taxa identified in the three groups using LEfSe analysis. The colors represent the group in which the indicated taxa is more abundant compared to the other group. Ctr.20, 20-week-old offspring of dams fed with the standard control diet; HF.20, 20-week-old offspring of dams fed with the high-fat diet; HFB.20, 20-week-old offspring of dams fed with the high-fat diet with betaine. Data are expressed as means ± S.E.M. (n = 5–6/group). One-way ANOVA; **p < 0.01 vs. Ctr, #p < 0.05 vs. HF.
FIGURE 7
FIGURE 7
Heatmap of the correlation analysis between the altered genera and glucose and lipid metabolic parameters in dams and offspring. Correlation results in dams after weaning and in offspring at 4 and 20 weeks of age are shown. HOMA-IR, the homeostasis model assessment of insulin resistance; GLU, glucose; AST, aspartate aminotransferase; ALT, alanine transaminase; FFA, free fatty acid; HDL-C, high-density lipoprotein cholesterol; LDL-C, low-density lipoprotein cholesterol; TC, total cholesterol; TG, triglyceride. Values show a significant correlation between the genera and metabolic parameters: *p < 0.05 and **p < 0.01.

References

    1. Bojović K., Ignjatović ĐI., Bajić S., Milutinović D., Tomić M., Golić N., et al. (2020). Gut microbiota dysbiosis associated with altered production of short chain fatty acids in children with neurodevelopmental disorders. Front. Cell Infect. Microbiol. 10:223. 10.3389/fcimb.2020.00223
    1. Bokulich N. A., Subramanian S., Faith J. J., Gevers D., Gordon J. I., Knight R., et al. (2013). Quality-filtering vastly improves diversity estimates from Illumina amplicon sequencing. Nat. Methods 10 57–59. 10.1038/nmeth.2276
    1. Cai D., Jia Y., Lu J., Yuan M., Sui S., Song H., et al. (2014). Maternal dietary betaine supplementation modifies hepatic expression of cholesterol metabolic genes via epigenetic mechanisms in newborn piglets. Br. J. Nutr. 112 1459–1468. 10.1017/S0007114514002402
    1. Cai D., Yuan M., Liu H., Pan S., Ma W., Hong J., et al. (2016). Maternal betaine supplementation throughout gestation and lactation modifies hepatic cholesterol metabolic genes in weaning piglets via AMPK/LXR-Mediated pathway and histone modification. Nutrients 8:646. 10.3390/nu8100646
    1. Cano-Ibanez N., Martinez-Galiano J. M., Luque-Fernandez M. A., Martin-Pelaez S., Bueno-Cavanillas A., Delgado-Rodriguez M. (2020). Maternal dietary patterns during pregnancy and their association with gestational weight gain and nutrient adequacy. Int. J. Environ. Res. Public Health 17:7908. 10.3390/ijerph17217908
    1. Caporaso J. G., Kuczynski J., Stombaugh J., Bittinger K., Bushman F. D., Costello E. K., et al. (2010). QIIME allows analysis of high-throughput community sequencing data. Nat. Methods 7 335–336. 10.1038/nmeth.f.303
    1. Chen M., Xiao D., Liu W., Song Y., Zou B., Li L., et al. (2020). Intake of Ganoderma lucidum polysaccharides reverses the disturbed gut microbiota and metabolism in type 2 diabetic rats. Int. J. Biol. Macromol. 155 890–902. 10.1016/j.ijbiomac.2019.11.047
    1. Chen W., Zhang X., Xu M., Jiang L., Zhou M., Liu W., et al. (2021a). Betaine prevented high-fat diet-induced NAFLD by regulating the FGF10/AMPK signaling pathway in ApoE(-/-) mice. Eur. J. Nutr. 60 1655–1668. 10.1007/s00394-020-02362-6
    1. Chen Y. T., Hu Y., Yang Q. Y., Liu X. D., Son J. S., De Avila J. M., et al. (2021b). Embryonic exposure to hyper glucocorticoids suppresses brown fat development and thermogenesis via REDD1. Sci. Bull. (Beijing) 66 478–489. 10.1016/j.scib.2020.10.015
    1. Craig S. A. (2004). Betaine in human nutrition. Am. J. Clin. Nutr. 80 539–549. 10.1093/ajcn/80.3.539
    1. Diaz S. O., Pinto J., Graca G., Duarte I. F., Barros A. S., Galhano E., et al. (2011). Metabolic biomarkers of prenatal disorders: an exploratory NMR metabonomics study of second trimester maternal urine and blood plasma. J. Proteome Res. 10 3732–3742. 10.1021/pr200352m
    1. Du J., Li J., Liu X., Liu H., Obel C., Shen H., et al. (2021a). Association of maternal diabetes during pregnancy with high refractive error in offspring: a nationwide population-based cohort study. Diabetologia 64 2466–2477. 10.1007/s00125-021-05526-z
    1. Du J., Zhang P., Luo J., Shen L., Zhang S., Gu H., et al. (2021b). Dietary betaine prevents obesity through gut microbiota-drived microRNA-378a family. Gut Microbes 13 1–19. 10.1080/19490976.2020.1862612
    1. Edgar R. C. (2013). UPARSE: highly accurate OTU sequences from microbial amplicon reads. Nat. Methods 10 996–998. 10.1038/nmeth.2604
    1. Ejaz A., Martinez-Guino L., Goldfine A. B., Ribas-Aulinas F., De Nigris V., Ribó S., et al. (2016). Dietary betaine supplementation increases Fgf21 levels to improve glucose homeostasis and reduce hepatic lipid accumulation in mice. Diabetes Metab. Res. Rev. 65 902–912. 10.2337/db15-1094
    1. Fickert P., Fuchsbichler A., Marschall H. U., Wagner M., Zollner G., Krause R., et al. (2006). Lithocholic acid feeding induces segmental bile duct obstruction and destructive cholangitis in mice. Am. J. Pathol. 168 410–422. 10.2353/ajpath.2006.050404
    1. Fisher M. M., Magnusson R., Miyai K. (1971). Bile acid metabolism in mammals. I. Bile acid-induced intrahepatic cholestasis. Lab Invest. 25 88–91.
    1. Godfrey K. M., Reynolds R. M., Prescott S. L., Nyirenda M., Jaddoe V. W., Eriksson J. G., et al. (2017). Influence of maternal obesity on the long-term health of offspring. Lancet Diabetes Endocrinol. 5 53–64. 10.1016/S2213-8587(16)30107-3
    1. Harris M. I., Flegal K. M., Cowie C. C., Eberhardt M. S., Goldstein D. E., Little R. R., et al. (1998). Prevalence of diabetes, impaired fasting glucose, and impaired glucose tolerance in U.S. adults. the third national health and nutrition examination survey, 1988-1994. Diabetes Care 21 518–524. 10.2337/diacare.21.4.518
    1. Haugen A. C., Schug T. T., Collman G., Heindel J. J. (2015). Evolution of DOHaD: the impact of environmental health sciences. J. Dev. Orig. Health Dis. 6 55–64. 10.1017/S2040174414000580
    1. Inoue R., Ohue-Kitano R., Tsukahara T., Tanaka M., Masuda S., Inoue T., et al. (2017). Prediction of functional profiles of gut microbiota from 16S rRNA metagenomic data provides a more robust evaluation of gut dysbiosis occurring in Japanese type 2 diabetic patients. J. Clin. Biochem. Nutr. 61 217–221. 10.3164/jcbn.17-44
    1. Jiang M., Tang T., Liang X., Li J., Qiu Y., Liu S., et al. (2021). Maternal sevoflurane exposure induces temporary defects in interkinetic nuclear migration of radial glial progenitors in the fetal cerebral cortex through the Notch signalling pathway. Cell Prolif. 54:e13042. 10.1111/cpr.13042
    1. Kahn B. B., Flier J. S. (2000). Obesity and insulin resistance. J. Clin. Invest. 106 473–481.
    1. Kimura I., Miyamoto J., Ohue-Kitano R., Watanabe K., Yamada T., Onuki M., et al. (2020). Maternal gut microbiota in pregnancy influences offspring metabolic phenotype in mice. Science 367:eaaw8429. 10.1126/science.aaw8429
    1. Koistinen V. M., Kärkkäinen O., Borewicz K., Zarei I., Jokkala J., Micard V., et al. (2019). Contribution of gut microbiota to metabolism of dietary glycine betaine in mice and in vitro colonic fermentation. Microbiome 7:103. 10.1186/s40168-019-0718-2
    1. Kulkarni A., Dangat K., Kale A., Sable P., Chavan-Gautam P., Joshi S. (2011). Effects of altered maternal folic acid, vitamin B12 and docosahexaenoic acid on placental global DNA methylation patterns in Wistar rats. PLoS One 6:e17706. 10.1371/journal.pone.0017706
    1. Lever M., George P. M., Slow S., Bellamy D., Young J. M., Ho M., et al. (2014). Betaine and Trimethylamine-N-Oxide as predictors of cardiovascular outcomes show different patterns in diabetes mellitus: an observational study. PLoS One 9:e114969. 10.1371/journal.pone.0114969
    1. Lever M., Slow S., Mcgregor D. O., Dellow W. J., George P. M., Chambers S. T. (2012). Variability of plasma and urine betaine in diabetes mellitus and its relationship to methionine load test responses: an observational study. Cardiovasc. Diabetol. 11:34. 10.1186/1475-2840-11-34
    1. Lin H., An Y., Tang H., Wang Y. (2019). Alterations of bile acids and gut microbiota in obesity induced by high fat diet in rat model. J. Agric. Food Chem. 67 3624–3632. 10.1021/acs.jafc.9b00249
    1. Liu J., Yue S., Yang Z., Feng W., Meng X., Wang A., et al. (2018). Oral hydroxysafflor yellow A reduces obesity in mice by modulating the gut microbiota and serum metabolism. Pharmacol. Res. 134 40–50. 10.1016/j.phrs.2018.05.012
    1. Lorenzon F., Gregorio T., Niebisch F., Stolte R. C. K., Dos Santos G. J., Rafacho A., et al. (2021). Maternal vitamin D administration attenuates metabolic disturbances induced by prenatal exposure to dexamethasone in a sex-dependent manner. J. Steroid Biochem. Mol. Biol. 212:105941. 10.1016/j.jsbmb.2021.105941
    1. Mao G., Li S., Orfila C., Shen X., Zhou S., Linhardt R. J., et al. (2019). Depolymerized RG-I-enriched pectin from citrus segment membranes modulates gut microbiota, increases SCFA production, and promotes the growth of Bifidobacterium spp., Lactobacillus spp. and Faecalibaculum spp. Food Funct. 10 7828–7843. 10.1039/c9fo01534e
    1. Martínez-Cuesta M. C., Del Campo R., Garriga-García M., Peláez C., Requena T. (2021). Taxonomic characterization and short-chain fatty acids production of the obese microbiota. Front. Cell Infect. Microbiol. 11:598093. 10.3389/fcimb.2021.598093
    1. Mathews F., Yudkin P., Neil A. (2001). Maternal nutrition and pregnancy outcome. Arch. Dis. Child. 85:510.
    1. Mingrone G., Manco M., Mora M. E., Guidone C., Iaconelli A., Gniuli D., et al. (2008). Influence of maternal obesity on insulin sensitivity and secretion in offspring. Diabetes Care 31 1872–1876. 10.2337/dc08-0432
    1. Moeller A. H., Suzuki T. A., Phifer-Rixey M., Nachman M. W. (2018). Transmission modes of the mammalian gut microbiota. Science 362 453–457. 10.1126/science.aat7164
    1. Montazeri P., Vrijheid M., Martinez D., Basterrechea M., Fernandez-Somoano A., Guxens M., et al. (2018). Maternal metabolic health parameters during pregnancy in relation to early childhood BMI trajectories. Obesity (Silver Spring) 26 588–596. 10.1002/oby.22095
    1. Murri M., Leiva I., Gomez-Zumaquero J. M., Tinahones F. J., Cardona F., Soriguer F., et al. (2013). Gut microbiota in children with type 1 diabetes differs from that in healthy children: a case-control study. BMC Med. 11:46. 10.1186/1741-7015-11-46
    1. Pathak P., Xie C., Nichols R. G., Ferrell J. M., Boehme S., Krausz K. W., et al. (2018). Intestine farnesoid X receptor agonist and the gut microbiota activate G-protein bile acid receptor-1 signaling to improve metabolism. Hepatology 68 1574–1588. 10.1002/hep.29857
    1. Quast C., Pruesse E., Yilmaz P., Gerken J., Schweer T., Yarza P., et al. (2013). The SILVA ribosomal RNA gene database project: improved data processing and web-based tools. Nucleic Acids Res. 41 D590–D596. 10.1093/nar/gks1219
    1. Ribaroff G. A., Wastnedge E., Drake A. J., Sharpe R. M., Chambers T. J. G. (2017). Animal models of maternal high fat diet exposure and effects on metabolism in offspring: a meta-regression analysis. Obes. Rev. 18 673–686. 10.1111/obr.12524
    1. Ricaboni D., Mailhe M., Khelaifia S., Raoult D., Million M. (2016). Romboutsia timonensis, a new species isolated from human gut. New Microbes New Infect. 12 6–7. 10.1016/j.nmni.2016.04.001
    1. Ridlon J. M., Kang D. J., Hylemon P. B. (2006). Bile salt biotransformations by human intestinal bacteria. J. Lipid Res. 47 241–259. 10.1194/jlr.R500013-JLR200
    1. Segata N., Izard J., Waldron L., Gevers D., Miropolsky L., Garrett W. S., et al. (2011). Metagenomic biomarker discovery and explanation. Genome Biol. 12:R60. 10.1186/gb-2011-12-6-r60
    1. Sung M. M., Kim T. T., Denou E., Soltys C. M., Hamza S. M., Byrne N. J., et al. (2017). Improved glucose homeostasis in obese mice treated with resveratrol is associated with alterations in the gut microbiome. Diabetes Metab. Res. Rev. 66 418–425. 10.2337/db16-0680
    1. Tong X., Xu J., Lian F., Yu X., Zhao Y., Xu L., et al. (2018). Structural alteration of gut microbiota during the amelioration of human Type 2 diabetes with hyperlipidemia by metformin and a traditional chinese herbal formula: a multicenter, randomized, open label clinical trial. mBio 9:e02392-17. 10.1128/mBio.02392-17
    1. van Lee L., Tint M. T., Aris I. M., Quah P. L., Fortier M. V., Lee Y. S., et al. (2016). Prospective associations of maternal betaine status with offspring weight and body composition at birth: the Growing Up in Singapore Towards healthy Outcomes (GUSTO) cohort study. Am. J. Clin. Nutr. 104 1327–1333. 10.3945/ajcn.116.138818
    1. Vishvanath L., Gupta R. K. (2019). Contribution of adipogenesis to healthy adipose tissue expansion in obesity. J. Clin. Invest. 129 4022–4031. 10.1172/JCI129191
    1. Wang P., Li D., Ke W., Liang D., Hu X., Chen F. (2020). Resveratrol-induced gut microbiota reduces obesity in high-fat diet-fed mice. Int. J. Obes (Lond) 44 213–225. 10.1038/s41366-019-0332-1
    1. Wang Y., Song J., Zhai Y., Zhang C., Gerritsen J., Wang H., et al. (2015). Romboutsia sedimentorum sp. nov., isolated from an alkaline-saline lake sediment and emended description of the genus Romboutsia. Int. J. Syst. Evol. Microbiol. 65 1193–1198. 10.1099/ijs.0.000079
    1. Wu C. H., Ko J. L., Liao J. M., Huang S. S., Lin M. Y., Lee L. H., et al. (2019). D-methionine alleviates cisplatin-induced mucositis by restoring the gut microbiota structure and improving intestinal inflammation. Ther. Adv. Med. Oncol. 11:1758835918821021. 10.1177/1758835918821021
    1. Xiao S., Fei N., Pang X., Shen J., Wang L., Zhang B., et al. (2014). A gut microbiota-targeted dietary intervention for amelioration of chronic inflammation underlying metabolic syndrome. FEMS Microbiol. Ecol. 87 357–367. 10.1111/1574-6941.12228
    1. Yang S., Zhao N., Yang Y., Hu Y., Dong H., Zhao R. (2018). Mitotically stable modification of DNA methylation in IGF2/H19 imprinting control region is associated with activated hepatic igf2 expression in offspring rats from betaine-supplemented dams. J. Agric. Food Chem. 66 2704–2713. 10.1021/acs.jafc.7b05418
    1. Yao Y., Cai X., Ye Y., Wang F., Chen F., Zheng C. (2021). The role of microbiota in infant health: from early life to adulthood. Front. Immunol. 12:708472. 10.3389/fimmu.2021.708472
    1. Yoshida S., Takeuchi M., Kawakami C., Kawakami K., Ito S., Japan E., et al. (2020). Maternal multivitamin intake and orofacial clefts in offspring: Japan Environment and Children’s Study (JECS) cohort study. BMJ Open 10:e035817. 10.1136/bmjopen-2019-035817
    1. Yoshimoto S., Loo T. M., Atarashi K., Kanda H., Sato S., Oyadomari S., et al. (2013). Obesity-induced gut microbial metabolite promotes liver cancer through senescence secretome. Nature 499 97–101. 10.1038/nature12347
    1. Zhang X., Zhao Y., Xu J., Xue Z., Zhang M., Pang X., et al. (2015). Modulation of gut microbiota by berberine and metformin during the treatment of high-fat diet-induced obesity in rats. Sci. Rep. 5:14405. 10.1038/srep14405
    1. Zhao N., Yang S., Feng Y., Sun B., Zhao R. (2019). Enhanced hepatic cholesterol accumulation induced by maternal betaine exposure is associated with hypermethylation of CYP7A1 gene promoter. Endocrine 64 544–551. 10.1007/s12020-019-01906-z
    1. Zheng J., Xiao X., Zhang Q., Yu M., Xu J., Wang Z. (2014). Maternal high-fat diet modulates hepatic glucose, lipid homeostasis and gene expression in the PPAR pathway in the early life of offspring. Int. J. Mol. Sci. 15 14967–14983. 10.3390/ijms150914967
    1. Zhou L., Xiao X., Li M., Zhang Q., Yu M., Zheng J., et al. (2020). Maternal exercise improves high-fat diet-induced metabolic abnormalities and gut microbiota profiles in mouse dams and offspring. Front. Cell Infect. Microbiol. 10:292. 10.3389/fcimb.2020.00292
    1. Zhou L., Xiao X., Zhang Q., Zheng J., Deng M. (2019a). Maternal genistein intake mitigates the deleterious effects of high-fat diet on glucose and lipid metabolism and modulates gut microbiota in adult life of male mice. Front. Physiol. 10:985. 10.3389/fphys.2019.00985
    1. Zhou L., Xiao X., Zhang Q., Zheng J., Li M., Yu M., et al. (2019b). Dietary genistein could modulate hypothalamic circadian entrainment, reduce body weight, and improve glucose and lipid metabolism in female mice. Int. J. Endocrinol. 2019:2163838. 10.1155/2019/2163838
    1. Zhou L. Y., Xiao X. H., Zhang Q., Zheng J., Li M., Wang X. J., et al. (2019c). Gut microbiota might be a crucial factor in deciphering the metabolic benefits of perinatal genistein consumption in dams and adult female offspring. Food Funct. 10 4505–4521. 10.1039/c9fo01046g

Source: PubMed

3
Subscribe