Impact of Transgenerational Nutrition on Nonalcoholic Fatty Liver Disease Development: Interplay between Gut Microbiota, Epigenetics and Immunity
Abstract
:1. Introduction
2. Immune-Mediated Pathogenesis of Nonalcoholic Fatty Liver Disease (NAFLD)
2.1. Development of NASH
2.1.1. Role of CD8+ T Cells
2.1.2. Activation of CD8+ T Cells during NASH
2.1.3. Role of CD4+ T Cells
2.1.4. Activation of CD4+ T Cells during NASH
3. Effects of Maternal Nutrition on the Development of Offspring NAFLD
3.1. Hepatic Lipogenesis
3.2. Insulin Resistance
3.3. Maternal Under-Nutrition
4. Interplay of Early-Life Nutrition, Gut Microbiome, and Hepatic Inflammation
4.1. Gut Microbiome in Early Life
4.2. Microbial Composition
4.3. Gut-Liver Axis
4.4. Gut Bacteria and NAFLD Pathogenesis
4.5. Metabolites of Gut Bacteria and NAFLD Pathogenesis
4.6. Bile Acids
5. Effects of Maternal Nutrition on Epigenetic Remodeling in Offspring
5.1. DNA Methylation
5.2. microRNA and Histone Modifications
6. Impacts of Maternal Diet-Induced Epigenetic Remodeling on Immune Regulation
7. Therapeutic Intervention
8. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Welsh, J.A.; Karpen, S.; Vos, M.B. Increasing prevalence of nonalcoholic fatty liver disease among United States adolescents, 1988–1994 to 2007–2010. J. Pediatr. 2013, 162, 496–500.e1. [Google Scholar] [CrossRef]
- Hirsova, P.; Bamidele, A.O.; Wang, H.; Povero, D.; Revelo, X.S. Emerging Roles of T Cells in the Pathogenesis of Nonalcoholic Steatohepatitis and Hepatocellular Carcinoma. Front. Endocrinol. 2021, 12, 760860. [Google Scholar] [CrossRef] [PubMed]
- Huang, D.Q.; El-Serag, H.B.; Loomba, R. Global epidemiology of NAFLD-related HCC: Trends, predictions, risk factors and prevention. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 223–238. [Google Scholar] [CrossRef] [PubMed]
- Younossi, Z.M.; Golabi, P.; de Avila, L.; Paik, J.M.; Srishord, M.; Fukui, N.; Qiu, Y.; Burns, L.; Afendy, A.; Nader, F. The global epidemiology of NAFLD and NASH in patients with type 2 diabetes: A systematic review and meta-analysis. J. Hepatol. 2019, 71, 793–801. [Google Scholar] [CrossRef] [PubMed]
- Buzzetti, E.; Pinzani, M.; Tsochatzis, E.A. The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD). Metabolism 2016, 65, 1038–1048. [Google Scholar] [CrossRef]
- Barker, D.J. The developmental origins of chronic adult disease. Acta Paediatr. Suppl. 2004, 93, 26–33. [Google Scholar] [CrossRef]
- Hsu, C.N.; Tain, Y.L. The Good, the Bad, and the Ugly of Pregnancy Nutrients and Developmental Programming of Adult Disease. Nutrients 2019, 11, 894. [Google Scholar] [CrossRef]
- Parthasarathy, G.; Revelo, X.; Malhi, H. Pathogenesis of Nonalcoholic Steatohepatitis: An Overview. Hepatol. Commun. 2020, 4, 478–492. [Google Scholar] [CrossRef]
- Kazankov, K.; Jorgensen, S.M.D.; Thomsen, K.L.; Moller, H.J.; Vilstrup, H.; George, J.; Schuppan, D.; Gronbaek, H. The role of macrophages in nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 145–159. [Google Scholar] [CrossRef]
- Imajo, K.; Fujita, K.; Yoneda, M.; Nozaki, Y.; Ogawa, Y.; Shinohara, Y.; Kato, S.; Mawatari, H.; Shibata, W.; Kitani, H.; et al. Hyperresponsivity to low-dose endotoxin during progression to nonalcoholic steatohepatitis is regulated by leptin-mediated signaling. Cell Metab. 2012, 16, 44–54. [Google Scholar] [CrossRef]
- Pan, J.; Ou, Z.; Cai, C.; Li, P.; Gong, J.; Ruan, X.Z.; He, K. Fatty acid activates NLRP3 inflammasomes in mouse Kupffer cells through mitochondrial DNA release. Cell Immunol. 2018, 332, 111–120. [Google Scholar] [CrossRef] [PubMed]
- Deczkowska, A.; David, E.; Ramadori, P.; Pfister, D.; Safran, M.; At The, B.; Giladi, A.; Jaitin, D.A.; Barboy, O.; Cohen, M.; et al. XCR1(+) type 1 conventional dendritic cells drive liver pathology in non-alcoholic steatohepatitis. Nat. Med. 2021, 27, 1043–1054. [Google Scholar] [CrossRef] [PubMed]
- Ramadori, P.; Kam, S.; Heikenwalder, M. T cells: Friends and foes in NASH pathogenesis and hepatocarcinogenesis. Hepatology 2022, 75, 1038–1049. [Google Scholar] [CrossRef] [PubMed]
- Wen, Y.; Lambrecht, J.; Ju, C.; Tacke, F. Hepatic macrophages in liver homeostasis and diseases-diversity, plasticity and therapeutic opportunities. Cell. Mol. Immunol. 2021, 18, 45–56. [Google Scholar] [CrossRef]
- Malehmir, M.; Pfister, D.; Gallage, S.; Szydlowska, M.; Inverso, D.; Kotsiliti, E.; Leone, V.; Peiseler, M.; Surewaard, B.G.J.; Rath, D.; et al. Platelet GPIbalpha is a mediator and potential interventional target for NASH and subsequent liver cancer. Nat. Med. 2019, 25, 641–655. [Google Scholar] [CrossRef] [PubMed]
- Sutti, S.; Albano, E. Adaptive immunity: An emerging player in the progression of NAFLD. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 81–92. [Google Scholar] [CrossRef] [PubMed]
- Dudek, M.; Pfister, D.; Donakonda, S.; Filpe, P.; Schneider, A.; Laschinger, M.; Hartmann, D.; Huser, N.; Meiser, P.; Bayerl, F.; et al. Auto-aggressive CXCR6(+) CD8 T cells cause liver immune pathology in NASH. Nature 2021, 592, 444–449. [Google Scholar] [CrossRef] [PubMed]
- Breuer, D.A.; Pacheco, M.C.; Washington, M.K.; Montgomery, S.A.; Hasty, A.H.; Kennedy, A.J. CD8(+) T cells regulate liver injury in obesity-related nonalcoholic fatty liver disease. Am. J. Physiol. Gastrointest. Liver Physiol. 2020, 318, G211–G224. [Google Scholar] [CrossRef] [PubMed]
- Bhattacharjee, J.; Kirby, M.; Softic, S.; Miles, L.; Salazar-Gonzalez, R.M.; Shivakumar, P.; Kohli, R. Hepatic Natural Killer T-cell and CD8+ T-cell Signatures in Mice with Nonalcoholic Steatohepatitis. Hepatol. Commun. 2017, 1, 299–310. [Google Scholar] [CrossRef]
- Van Herck, M.A.; Vonghia, L.; Kwanten, W.J.; Jule, Y.; Vanwolleghem, T.; Ebo, D.G.; Michielsen, P.P.; De Man, J.G.; Gama, L.; De Winter, B.Y.; et al. Diet Reversal and Immune Modulation Show Key Role for Liver and Adipose Tissue T Cells in Murine Nonalcoholic Steatohepatitis. Cell. Mol. Gastroenterol. Hepatol. 2020, 10, 467–490. [Google Scholar] [CrossRef]
- Wolf, M.J.; Adili, A.; Piotrowitz, K.; Abdullah, Z.; Boege, Y.; Stemmer, K.; Ringelhan, M.; Simonavicius, N.; Egger, M.; Wohlleber, D.; et al. Metabolic activation of intrahepatic CD8+ T cells and NKT cells causes nonalcoholic steatohepatitis and liver cancer via cross-talk with hepatocytes. Cancer Cell 2014, 26, 549–564. [Google Scholar] [CrossRef] [PubMed]
- Her, Z.; Tan, J.H.L.; Lim, Y.S.; Tan, S.Y.; Chan, X.Y.; Tan, W.W.S.; Liu, M.; Yong, K.S.M.; Lai, F.; Ceccarello, E.; et al. CD4(+) T Cells Mediate the Development of Liver Fibrosis in High Fat Diet-Induced NAFLD in Humanized Mice. Front. Immunol. 2020, 11, 580968. [Google Scholar] [CrossRef] [PubMed]
- Rau, M.; Schilling, A.K.; Meertens, J.; Hering, I.; Weiss, J.; Jurowich, C.; Kudlich, T.; Hermanns, H.M.; Bantel, H.; Beyersdorf, N.; et al. Progression from Nonalcoholic Fatty Liver to Nonalcoholic Steatohepatitis Is Marked by a Higher Frequency of Th17 Cells in the Liver and an Increased Th17/Resting Regulatory T Cell Ratio in Peripheral Blood and in the Liver. J. Immunol. 2016, 196, 97–105. [Google Scholar] [CrossRef] [PubMed]
- Luo, X.Y.; Takahara, T.; Kawai, K.; Fujino, M.; Sugiyama, T.; Tsuneyama, K.; Tsukada, K.; Nakae, S.; Zhong, L.; Li, X.K. IFN-gamma deficiency attenuates hepatic inflammation and fibrosis in a steatohepatitis model induced by a methionine- and choline-deficient high-fat diet. Am. J. Physiol. Gastrointest. Liver Physiol. 2013, 305, G891–G899. [Google Scholar] [CrossRef] [PubMed]
- Inzaugarat, M.E.; Ferreyra Solari, N.E.; Billordo, L.A.; Abecasis, R.; Gadano, A.C.; Chernavsky, A.C. Altered phenotype and functionality of circulating immune cells characterize adult patients with nonalcoholic steatohepatitis. J. Clin. Immunol. 2011, 31, 1120–1130. [Google Scholar] [CrossRef]
- Sun, G.; Jin, H.; Zhang, C.; Meng, H.; Zhao, X.; Wei, D.; Ou, X.; Wang, Q.; Li, S.; Wang, T.; et al. OX40 Regulates Both Innate and Adaptive Immunity and Promotes Nonalcoholic Steatohepatitis. Cell Rep. 2018, 25, 3786–3799.e4. [Google Scholar] [CrossRef] [PubMed]
- Zhu, J.; Yamane, H.; Cote-Sierra, J.; Guo, L.; Paul, W.E. GATA-3 promotes Th2 responses through three different mechanisms: Induction of Th2 cytokine production, selective growth of Th2 cells and inhibition of Th1 cell-specific factors. Cell Res. 2006, 16, 3–10. [Google Scholar] [CrossRef]
- Su, L.; Wu, Z.; Chi, Y.; Song, Y.; Xu, J.; Tan, J.; Cong, X.; Liu, Y. Mesenteric lymph node CD4(+) T lymphocytes migrate to liver and contribute to non-alcoholic fatty liver disease. Cell. Immunol. 2019, 337, 33–41. [Google Scholar] [CrossRef] [PubMed]
- Ross, T.T.; Crowley, C.; Kelly, K.L.; Rinaldi, A.; Beebe, D.A.; Lech, M.P.; Martinez, R.V.; Carvajal-Gonzalez, S.; Boucher, M.; Hirenallur-Shanthappa, D.; et al. Acetyl-CoA Carboxylase Inhibition Improves Multiple Dimensions of NASH Pathogenesis in Model Systems. Cell. Mol. Gastroenterol. Hepatol. 2020, 10, 829–851. [Google Scholar] [CrossRef]
- Purcell, A.R.; Rodrigo, N.; Cao, Q.; Joseph, O.; Gill, A.J.; Saad, S.; Pollock, C.A.; Glastras, S.J. Maternal Weight Intervention in the Perinatal Period Improves Liver Health in the Offspring of Mothers with Obesity. Nutrients 2023, 16, 109. [Google Scholar] [CrossRef]
- Cohen, C.C.; Perng, W.; Sauder, K.A.; Shapiro, A.L.B.; Starling, A.P.; Friedman, C.; Felix, J.F.; Kupers, L.K.; Moore, B.F.; Hebert, J.R.; et al. Maternal Diet Quality During Pregnancy and Offspring Hepatic Fat in Early Childhood: The Healthy Start Study. J. Nutr. 2023, 153, 1122–1132. [Google Scholar] [CrossRef] [PubMed]
- Peng, H.; Li, J.; Xu, H.; Wang, X.; He, L.; McCauley, N.; Zhang, K.K.; Xie, L. Offspring NAFLD liver phospholipid profiles are differentially programmed by maternal high-fat diet and maternal one carbon supplement. J. Nutr. Biochem. 2023, 111, 109187. [Google Scholar] [CrossRef] [PubMed]
- Quek, S.X.Z.; Tan, E.X.; Ren, Y.P.; Muthiah, M.; Loo, E.X.L.; Tham, E.H.; Siah, K.T.H. Factors early in life associated with hepatic steatosis. World J. Hepatol. 2022, 14, 1235–1247. [Google Scholar] [CrossRef] [PubMed]
- Chirala, S.S.; Chang, H.; Matzuk, M.; Abu-Elheiga, L.; Mao, J.; Mahon, K.; Finegold, M.; Wakil, S.J. Fatty acid synthesis is essential in embryonic development: Fatty acid synthase null mutants and most of the heterozygotes die in utero. Proc. Natl. Acad. Sci. USA 2003, 100, 6358–6363. [Google Scholar] [CrossRef] [PubMed]
- McCurdy, C.E.; Bishop, J.M.; Williams, S.M.; Grayson, B.E.; Smith, M.S.; Friedman, J.E.; Grove, K.L. Maternal high-fat diet triggers lipotoxicity in the fetal livers of nonhuman primates. J. Clin. Investig. 2009, 119, 323–335. [Google Scholar] [CrossRef]
- Thorn, S.R.; Baquero, K.C.; Newsom, S.A.; El Kasmi, K.C.; Bergman, B.C.; Shulman, G.I.; Grove, K.L.; Friedman, J.E. Early life exposure to maternal insulin resistance has persistent effects on hepatic NAFLD in juvenile nonhuman primates. Diabetes 2014, 63, 2702–2713. [Google Scholar] [CrossRef] [PubMed]
- Raffaella, C.; Francesca, B.; Italia, F.; Marina, P.; Giovanna, L.; Susanna, I. Alterations in hepatic mitochondrial compartment in a model of obesity and insulin resistance. Obesity 2008, 16, 958–964. [Google Scholar] [CrossRef] [PubMed]
- Bruce, K.D.; Cagampang, F.R.; Argenton, M.; Zhang, J.; Ethirajan, P.L.; Burdge, G.C.; Bateman, A.C.; Clough, G.F.; Poston, L.; Hanson, M.A.; et al. Maternal high-fat feeding primes steatohepatitis in adult mice offspring, involving mitochondrial dysfunction and altered lipogenesis gene expression. Hepatology 2009, 50, 1796–1808. [Google Scholar] [CrossRef] [PubMed]
- Alfaradhi, M.Z.; Fernandez-Twinn, D.S.; Martin-Gronert, M.S.; Musial, B.; Fowden, A.; Ozanne, S.E. Oxidative stress and altered lipid homeostasis in the programming of offspring fatty liver by maternal obesity. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2014, 307, R26–R34. [Google Scholar] [CrossRef]
- Burgueno, A.L.; Cabrerizo, R.; Gonzales Mansilla, N.; Sookoian, S.; Pirola, C.J. Maternal high-fat intake during pregnancy programs metabolic-syndrome-related phenotypes through liver mitochondrial DNA copy number and transcriptional activity of liver PPARGC1A. J. Nutr. Biochem. 2013, 24, 6–13. [Google Scholar] [CrossRef]
- Stefan, N.; Kantartzis, K.; Haring, H.U. Causes and metabolic consequences of Fatty liver. Endocr. Rev. 2008, 29, 939–960. [Google Scholar] [CrossRef]
- Cerf, M.E.; Chapman, C.S.; Muller, C.J.; Louw, J. Gestational high-fat programming impairs insulin release and reduces Pdx-1 and glucokinase immunoreactivity in neonatal Wistar rats. Metabolism 2009, 58, 1787–1792. [Google Scholar] [CrossRef]
- Cerf, M.E.; Chapman, C.S.; Louw, J. High-fat programming of hyperglycemia, hyperinsulinemia, insulin resistance, hyperleptinemia, and altered islet architecture in 3-month-old wistar rats. ISRN Endocrinol. 2012, 2012, 627270. [Google Scholar] [CrossRef] [PubMed]
- Dumortier, O.; Blondeau, B.; Duvillie, B.; Reusens, B.; Breant, B.; Remacle, C. Different mechanisms operating during different critical time-windows reduce rat fetal beta cell mass due to a maternal low-protein or low-energy diet. Diabetologia 2007, 50, 2495–2503. [Google Scholar] [CrossRef] [PubMed]
- Parlee, S.D.; MacDougald, O.A. Maternal nutrition and risk of obesity in offspring: The Trojan horse of developmental plasticity. Biochim. Biophys. Acta 2014, 1842, 495–506. [Google Scholar] [CrossRef] [PubMed]
- Dumolt, J.H.; Powell, T.L.; Jansson, T. Placental Function and the Development of Fetal Overgrowth and Fetal Growth Restriction. Obstet. Gynecol. Clin. N. Am. 2021, 48, 247–266. [Google Scholar] [CrossRef] [PubMed]
- Magee, T.R.; Han, G.; Cherian, B.; Khorram, O.; Ross, M.G.; Desai, M. Down-regulation of transcription factor peroxisome proliferator-activated receptor in programmed hepatic lipid dysregulation and inflammation in intrauterine growth-restricted offspring. Am. J. Obstet. Gynecol. 2008, 199, 271.e1–271.e5. [Google Scholar] [CrossRef] [PubMed]
- Souza-Mello, V.; Mandarim-de-Lacerda, C.A.; Aguila, M.B. Hepatic structural alteration in adult programmed offspring (severe maternal protein restriction) is aggravated by post-weaning high-fat diet. Br. J. Nutr. 2007, 98, 1159–1169. [Google Scholar] [CrossRef] [PubMed]
- Amadou, C.; Nabi, O.; Serfaty, L.; Lacombe, K.; Boursier, J.; Mathurin, P.; Ribet, C.; de Ledinghen, V.; Zins, M.; Charles, M.A. Association between birth weight, preterm birth, and nonalcoholic fatty liver disease in a community-based cohort. Hepatology 2022, 76, 1438–1451. [Google Scholar] [CrossRef]
- Arifuzzaman, M.; Collins, N.; Guo, C.J.; Artis, D. Nutritional regulation of microbiota-derived metabolites: Implications for immunity and inflammation. Immunity 2024, 57, 14–27. [Google Scholar] [CrossRef]
- Mueller, N.T.; Bakacs, E.; Combellick, J.; Grigoryan, Z.; Dominguez-Bello, M.G. The infant microbiome development: Mom matters. Trends Mol. Med. 2015, 21, 109–117. [Google Scholar] [CrossRef] [PubMed]
- Koren, O.; Goodrich, J.K.; Cullender, T.C.; Spor, A.; Laitinen, K.; Backhed, H.K.; Gonzalez, A.; Werner, J.J.; Angenent, L.T.; Knight, R.; et al. Host remodeling of the gut microbiome and metabolic changes during pregnancy. Cell 2012, 150, 470–480. [Google Scholar] [CrossRef] [PubMed]
- Yatsunenko, T.; Rey, F.E.; Manary, M.J.; Trehan, I.; Dominguez-Bello, M.G.; Contreras, M.; Magris, M.; Hidalgo, G.; Baldassano, R.N.; Anokhin, A.P.; et al. Human gut microbiome viewed across age and geography. Nature 2012, 486, 222–227. [Google Scholar] [CrossRef] [PubMed]
- Ma, J.; Prince, A.L.; Bader, D.; Hu, M.; Ganu, R.; Baquero, K.; Blundell, P.; Alan Harris, R.; Frias, A.E.; Grove, K.L.; et al. High-fat maternal diet during pregnancy persistently alters the offspring microbiome in a primate model. Nat. Commun. 2014, 5, 3889. [Google Scholar] [CrossRef] [PubMed]
- Dominguez-Bello, M.G.; Godoy-Vitorino, F.; Knight, R.; Blaser, M.J. Role of the microbiome in human development. Gut 2019, 68, 1108–1114. [Google Scholar] [CrossRef]
- Laterza, L.; Rizzatti, G.; Gaetani, E.; Chiusolo, P.; Gasbarrini, A. The Gut Microbiota and Immune System Relationship in Human Graft-versus-Host Disease. Mediterr. J. Hematol. Infect. Dis. 2016, 8, e2016025. [Google Scholar] [CrossRef]
- Tamburini, S.; Shen, N.; Wu, H.C.; Clemente, J.C. The microbiome in early life: Implications for health outcomes. Nat. Med. 2016, 22, 713–722. [Google Scholar] [CrossRef] [PubMed]
- Soderborg, T.K.; Clark, S.E.; Mulligan, C.E.; Janssen, R.C.; Babcock, L.; Ir, D.; Young, B.; Krebs, N.; Lemas, D.J.; Johnson, L.K.; et al. The gut microbiota in infants of obese mothers increases inflammation and susceptibility to NAFLD. Nat. Commun. 2018, 9, 4462. [Google Scholar] [CrossRef] [PubMed]
- Zhong, C.; Wang, Q.; He, Y.; Zhao, X.; Wang, Y.; He, L.; Wei, H.; Tao, X. Maternal supplementation with human milk-derived Lactiplantibacillus plantarum WLPL04 affects the immunity and gut microbiota of offspring rats. Food Funct. 2023, 14, 5326–5341. [Google Scholar] [CrossRef]
- Jee, J.J.; Yang, L.; Shivakumar, P.; Xu, P.P.; Mourya, R.; Thanekar, U.; Yu, P.; Zhu, Y.; Pan, Y.; Wang, H.; et al. Maternal regulation of biliary disease in neonates via gut microbial metabolites. Nat. Commun. 2022, 13, 18. [Google Scholar] [CrossRef]
- Le, H.H.; Lee, M.T.; Besler, K.R.; Johnson, E.L. Host hepatic metabolism is modulated by gut microbiota-derived sphingolipids. Cell Host Microbe 2022, 30, 798–808.e7. [Google Scholar] [CrossRef] [PubMed]
- Zhang, H.; Wang, Y.; Zhang, X.; Zhang, L.; Zhao, X.; Xu, Y.; Wang, P.; Liang, X.; Xue, M.; Liang, H. Maternal Folic Acid Supplementation during Pregnancy Prevents Hepatic Steatosis in Male Offspring of Rat Dams Fed High-Fat Diet, Which Is Associated with the Regulation of Gut Microbiota. Nutrients 2023, 15, 4726. [Google Scholar] [CrossRef] [PubMed]
- Tripathi, A.; Debelius, J.; Brenner, D.A.; Karin, M.; Loomba, R.; Schnabl, B.; Knight, R. The gut-liver axis and the intersection with the microbiome. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 397–411. [Google Scholar] [CrossRef] [PubMed]
- Mouries, J.; Brescia, P.; Silvestri, A.; Spadoni, I.; Sorribas, M.; Wiest, R.; Mileti, E.; Galbiati, M.; Invernizzi, P.; Adorini, L.; et al. Microbiota-driven gut vascular barrier disruption is a prerequisite for non-alcoholic steatohepatitis development. J. Hepatol. 2019, 71, 1216–1228. [Google Scholar] [CrossRef] [PubMed]
- Rahman, K.; Desai, C.; Iyer, S.S.; Thorn, N.E.; Kumar, P.; Liu, Y.; Smith, T.; Neish, A.S.; Li, H.; Tan, S.; et al. Loss of Junctional Adhesion Molecule A Promotes Severe Steatohepatitis in Mice on a Diet High in Saturated Fat, Fructose, and Cholesterol. Gastroenterology 2016, 151, 733–746.e12. [Google Scholar] [CrossRef]
- Hild, B.; Dreier, M.S.; Oh, J.H.; McCulloch, J.A.; Badger, J.H.; Guo, J.; Thefaine, C.E.; Umarova, R.; Hall, K.D.; Gavrilova, O.; et al. Neonatal exposure to a wild-derived microbiome protects mice against diet-induced obesity. Nat. Metab. 2021, 3, 1042–1057. [Google Scholar] [CrossRef]
- Le Roy, T.; Llopis, M.; Lepage, P.; Bruneau, A.; Rabot, S.; Bevilacqua, C.; Martin, P.; Philippe, C.; Walker, F.; Bado, A.; et al. Intestinal microbiota determines development of non-alcoholic fatty liver disease in mice. Gut 2013, 62, 1787–1794. [Google Scholar] [CrossRef] [PubMed]
- Soderborg, T.K.; Friedman, J.E. Imbalance in gut microbes from babies born to obese mothers increases gut permeability and myeloid cell adaptations that provoke obesity and NAFLD. Microb. Cell 2018, 6, 102–104. [Google Scholar] [CrossRef]
- Yu, H.R.; Sheen, J.M.; Hou, C.Y.; Lin, I.C.; Huang, L.T.; Tain, Y.L.; Cheng, H.H.; Lai, Y.J.; Lin, Y.J.; Tiao, M.M.; et al. Effects of Maternal Gut Microbiota-Targeted Therapy on the Programming of Nonalcoholic Fatty Liver Disease in Dams and Fetuses, Related to a Prenatal High-Fat Diet. Nutrients 2022, 14, 4004. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef]
- Dai, X.; Guo, Z.; Chen, D.; Li, L.; Song, X.; Liu, T.; Jin, G.; Li, Y.; Liu, Y.; Ajiguli, A.; et al. Maternal sucralose intake alters gut microbiota of offspring and exacerbates hepatic steatosis in adulthood. Gut Microbes 2020, 11, 1043–1063. [Google Scholar] [CrossRef] [PubMed]
- Zhang, N.; Qu, Y.; Qin, B. Sodium butyrate ameliorates non-alcoholic fatty liver disease by upregulating miR-150 to suppress CXCR4 expression. Clin. Exp. Pharmacol. Physiol. 2021, 48, 1125–1136. [Google Scholar] [CrossRef] [PubMed]
- Du, J.; Zhang, P.; Luo, J.; Shen, L.; Zhang, S.; Gu, H.; He, J.; Wang, L.; Zhao, X.; Gan, M.; et al. Dietary betaine prevents obesity through gut microbiota-drived microRNA-378a family. Gut Microbes 2021, 13, 1–19. [Google Scholar] [CrossRef] [PubMed]
- Hsu, C.L.; Schnabl, B. The gut-liver axis and gut microbiota in health and liver disease. Nat. Rev. Microbiol. 2023, 21, 719–733. [Google Scholar] [CrossRef] [PubMed]
- Jiao, N.; Baker, S.S.; Chapa-Rodriguez, A.; Liu, W.; Nugent, C.A.; Tsompana, M.; Mastrandrea, L.; Buck, M.J.; Baker, R.D.; Genco, R.J.; et al. Suppressed hepatic bile acid signalling despite elevated production of primary and secondary bile acids in NAFLD. Gut 2018, 67, 1881–1891. [Google Scholar] [CrossRef] [PubMed]
- Fang, S.; Suh, J.M.; Reilly, S.M.; Yu, E.; Osborn, O.; Lackey, D.; Yoshihara, E.; Perino, A.; Jacinto, S.; Lukasheva, Y.; et al. Intestinal FXR agonism promotes adipose tissue browning and reduces obesity and insulin resistance. Nat. Med. 2015, 21, 159–165. [Google Scholar] [CrossRef]
- Jiang, C.; Xie, C.; Li, F.; Zhang, L.; Nichols, R.G.; Krausz, K.W.; Cai, J.; Qi, Y.; Fang, Z.Z.; Takahashi, S.; et al. Intestinal farnesoid X receptor signaling promotes nonalcoholic fatty liver disease. J. Clin. Investig. 2015, 125, 386–402. [Google Scholar] [CrossRef]
- Hang, S.; Paik, D.; Yao, L.; Kim, E.; Trinath, J.; Lu, J.; Ha, S.; Nelson, B.N.; Kelly, S.P.; Wu, L.; et al. Bile acid metabolites control T(H)17 and T(reg) cell differentiation. Nature 2019, 576, 143–148. [Google Scholar] [CrossRef] [PubMed]
- Thompson, M.D.; Derse, A.; Ferey, J.; Reid, M.; Xie, Y.; Christ, M.; Chatterjee, D.; Nguyen, C.; Harasymowicz, N.; Guilak, F.; et al. Transgenerational impact of maternal obesogenic diet on offspring bile acid homeostasis and nonalcoholic fatty liver disease. Am. J. Physiol. Endocrinol. Metab. 2019, 316, E674–E686. [Google Scholar] [CrossRef]
- Eslam, M.; George, J. Genetic contributions to NAFLD: Leveraging shared genetics to uncover systems biology. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 40–52. [Google Scholar] [CrossRef]
- Suter, M.A.; Ma, J.; Vuguin, P.M.; Hartil, K.; Fiallo, A.; Harris, R.A.; Charron, M.J.; Aagaard, K.M. In utero exposure to a maternal high-fat diet alters the epigenetic histone code in a murine model. Am. J. Obstet. Gynecol. 2014, 210, 463.e1–463.e11. [Google Scholar] [CrossRef] [PubMed]
- Ge, Z.J.; Luo, S.M.; Lin, F.; Liang, Q.X.; Huang, L.; Wei, Y.C.; Hou, Y.; Han, Z.M.; Schatten, H.; Sun, Q.Y. DNA methylation in oocytes and liver of female mice and their offspring: Effects of high-fat-diet-induced obesity. Environ. Health Perspect. 2014, 122, 159–164. [Google Scholar] [CrossRef] [PubMed]
- Fu, Q.; Cheung, W.A.; Majnik, A.V.; Ke, X.; Pastinen, T.; Lane, R.H. Adverse Maternal Environments Perturb Hepatic DNA Methylome and Transcriptome Prior to the Adult-Onset Non-Alcoholic Fatty Liver Disease in Mouse Offspring. Nutrients 2023, 15, 2167. [Google Scholar] [CrossRef] [PubMed]
- Ando, Y.; Munetsuna, E.; Yamada, H.; Ikeya, M.; Teshigawara, A.; Kageyama, I.; Nouchi, Y.; Wakasugi, T.; Yamazaki, M.; Mizuno, G.; et al. Impact of maternal fructose intake on liver stem/progenitor cells in offspring: Insights into developmental origins of health and disease. Life Sci. 2024, 336, 122315. [Google Scholar] [CrossRef] [PubMed]
- Chen, S.Y.; Chen, Y.Z.; Lee, Y.J.; Jiang, C.L.; Lu, S.C.; Lin, F.J. Maternal hypercholesterolemia exacerbates atherosclerosis lesions in female offspring through potentiating macrophage polarization toward an inflammatory M1 phenotype. J. Nutr. Biochem. 2021, 90, 108575. [Google Scholar] [CrossRef] [PubMed]
- Lai, Z.; Chen, J.; Ding, C.; Wong, K.; Chen, X.; Pu, L.; Huang, Q.; Chen, X.; Cheng, Z.; Liu, Y.; et al. Association of Hepatic Global DNA Methylation and Serum One-Carbon Metabolites with Histological Severity in Patients with NAFLD. Obesity 2020, 28, 197–205. [Google Scholar] [CrossRef] [PubMed]
- Murphy, S.K.; Yang, H.; Moylan, C.A.; Pang, H.; Dellinger, A.; Abdelmalek, M.F.; Garrett, M.E.; Ashley-Koch, A.; Suzuki, A.; Tillmann, H.L.; et al. Relationship between methylome and transcriptome in patients with nonalcoholic fatty liver disease. Gastroenterology 2013, 145, 1076–1087. [Google Scholar] [CrossRef] [PubMed]
- Marchlewicz, E.H.; Dolinoy, D.C.; Tang, L.; Milewski, S.; Jones, T.R.; Goodrich, J.M.; Soni, T.; Domino, S.E.; Song, P.X.; Burant, C.F.; et al. Lipid metabolism is associated with developmental epigenetic programming. Sci. Rep. 2016, 6, 34857. [Google Scholar] [CrossRef]
- Hanlon, M.A.; Gulati, R.; Johnston, M.; Fleifil, Y.; Rivas, M.; Timchenko, N.A. Genetic Ablation of C/EBPalpha-p300 Pathway Blocks Development of Obese Pregnancy Associated Liver Disorders in Offspring. Cell. Mol. Gastroenterol. Hepatol. 2024, 17, 347–360. [Google Scholar] [CrossRef]
- Suter, M.A.; Chen, A.; Burdine, M.S.; Choudhury, M.; Harris, R.A.; Lane, R.H.; Friedman, J.E.; Grove, K.L.; Tackett, A.J.; Aagaard, K.M. A maternal high-fat diet modulates fetal SIRT1 histone and protein deacetylase activity in nonhuman primates. FASEB J. 2012, 26, 5106–5114. [Google Scholar] [CrossRef]
- Li, J.; Huang, J.; Li, J.S.; Chen, H.; Huang, K.; Zheng, L. Accumulation of endoplasmic reticulum stress and lipogenesis in the liver through generational effects of high fat diets. J. Hepatol. 2012, 56, 900–907. [Google Scholar] [CrossRef] [PubMed]
- Pirola, C.J.; Fernandez Gianotti, T.; Castano, G.O.; Mallardi, P.; San Martino, J.; Mora Gonzalez Lopez Ledesma, M.; Flichman, D.; Mirshahi, F.; Sanyal, A.J.; Sookoian, S. Circulating microRNA signature in non-alcoholic fatty liver disease: From serum non-coding RNAs to liver histology and disease pathogenesis. Gut 2015, 64, 800–812. [Google Scholar] [CrossRef] [PubMed]
- Cheung, O.; Puri, P.; Eicken, C.; Contos, M.J.; Mirshahi, F.; Maher, J.W.; Kellum, J.M.; Min, H.; Luketic, V.A.; Sanyal, A.J. Nonalcoholic steatohepatitis is associated with altered hepatic MicroRNA expression. Hepatology 2008, 48, 1810–1820. [Google Scholar] [CrossRef] [PubMed]
- Zhang, J.; Zhang, F.; Didelot, X.; Bruce, K.D.; Cagampang, F.R.; Vatish, M.; Hanson, M.; Lehnert, H.; Ceriello, A.; Byrne, C.D. Maternal high fat diet during pregnancy and lactation alters hepatic expression of insulin like growth factor-2 and key microRNAs in the adult offspring. BMC Genom. 2009, 10, 478. [Google Scholar] [CrossRef] [PubMed]
- Hsu, S.H.; Wang, B.; Kota, J.; Yu, J.; Costinean, S.; Kutay, H.; Yu, L.; Bai, S.; La Perle, K.; Chivukula, R.R.; et al. Essential metabolic, anti-inflammatory, and anti-tumorigenic functions of miR-122 in liver. J. Clin. Investig. 2012, 122, 2871–2883. [Google Scholar] [CrossRef]
- Campisano, S.; La Colla, A.; Echarte, S.M.; Chisari, A.N. Interplay between early-life malnutrition, epigenetic modulation of the immune function and liver diseases. Nutr. Res. Rev. 2019, 32, 128–145. [Google Scholar] [CrossRef]
- Enninga, E.A.L.; Jang, J.S.; Hur, B.; Johnson, E.L.; Wick, M.J.; Sung, J.; Chakraborty, R. Maternal obesity is associated with phenotypic alterations in fetal immune cells by single-cell mass cytometry. Am. J. Reprod. Immunol. 2021, 85, e13358. [Google Scholar] [CrossRef] [PubMed]
- Gonzalez-Espinosa, L.O.; Montiel-Cervantes, L.A.; Guerra-Marquez, A.; Penaflor-Juarez, K.; Reyes-Maldonado, E.; Vela-Ojeda, J. Maternal obesity associated with increase in natural killer T cells and CD8+ regulatory T cells in cord blood units. Transfusion 2016, 56, 1075–1081. [Google Scholar] [CrossRef]
- Cifuentes-Zuniga, F.; Arroyo-Jousse, V.; Soto-Carrasco, G.; Casanello, P.; Uauy, R.; Krause, B.J.; Castro-Rodriguez, J.A. IL-10 expression in macrophages from neonates born from obese mothers is suppressed by IL-4 and LPS/INFgamma. J. Cell Physiol. 2017, 232, 3693–3701. [Google Scholar] [CrossRef]
- Sureshchandra, S.; Chan, C.N.; Robino, J.J.; Parmelee, L.K.; Nash, M.J.; Wesolowski, S.R.; Pietras, E.M.; Friedman, J.E.; Takahashi, D.; Shen, W.; et al. Maternal Western-style diet remodels the transcriptional landscape of fetal hematopoietic stem and progenitor cells in rhesus macaques. Stem Cell Rep. 2022, 17, 2595–2609. [Google Scholar] [CrossRef]
- Nash, M.J.; Dobrinskikh, E.; Soderborg, T.K.; Janssen, R.C.; Takahashi, D.L.; Dean, T.A.; Varlamov, O.; Hennebold, J.D.; Gannon, M.; Aagaard, K.M.; et al. Maternal diet alters long-term innate immune cell memory in fetal and juvenile hematopoietic stem and progenitor cells in nonhuman primate offspring. Cell Rep. 2023, 42, 112393. [Google Scholar] [CrossRef] [PubMed]
- Apostol, A.C.; Jensen, K.D.C.; Beaudin, A.E. Training the Fetal Immune System Through Maternal Inflammation-A Layered Hygiene Hypothesis. Front. Immunol. 2020, 11, 123. [Google Scholar] [CrossRef] [PubMed]
- Aluvihare, V.R.; Kallikourdis, M.; Betz, A.G. Regulatory T cells mediate maternal tolerance to the fetus. Nat. Immunol. 2004, 5, 266–271. [Google Scholar] [CrossRef] [PubMed]
- Craven, L.; Rahman, A.; Nair Parvathy, S.; Beaton, M.; Silverman, J.; Qumosani, K.; Hramiak, I.; Hegele, R.; Joy, T.; Meddings, J.; et al. Allogenic Fecal Microbiota Transplantation in Patients With Nonalcoholic Fatty Liver Disease Improves Abnormal Small Intestinal Permeability: A Randomized Control Trial. Am. J. Gastroenterol. 2020, 115, 1055–1065. [Google Scholar] [CrossRef] [PubMed]
- Stols-Goncalves, D.; Mak, A.L.; Madsen, M.S.; van der Vossen, E.W.J.; Bruinstroop, E.; Henneman, P.; Mol, F.; Scheithauer, T.P.M.; Smits, L.; Witjes, J.; et al. Faecal Microbiota transplantation affects liver DNA methylation in Non-alcoholic fatty liver disease: A multi-omics approach. Gut Microbes 2023, 15, 2223330. [Google Scholar] [CrossRef] [PubMed]
- Bajaj, J.S.; Kakiyama, G.; Savidge, T.; Takei, H.; Kassam, Z.A.; Fagan, A.; Gavis, E.A.; Pandak, W.M.; Nittono, H.; Hylemon, P.B.; et al. Antibiotic-Associated Disruption of Microbiota Composition and Function in Cirrhosis Is Restored by Fecal Transplant. Hepatology 2018, 68, 1549–1558. [Google Scholar] [CrossRef]
- Ahn, S.B.; Jun, D.W.; Kang, B.K.; Lim, J.H.; Lim, S.; Chung, M.J. Randomized, Double-blind, Placebo-controlled Study of a Multispecies Probiotic Mixture in Nonalcoholic Fatty Liver Disease. Sci. Rep. 2019, 9, 5688. [Google Scholar] [CrossRef] [PubMed]
- Kobyliak, N.; Abenavoli, L.; Mykhalchyshyn, G.; Kononenko, L.; Boccuto, L.; Kyriienko, D.; Dynnyk, O. A Multi-strain Probiotic Reduces the Fatty Liver Index, Cytokines and Aminotransferase levels in NAFLD Patients: Evidence from a Randomized Clinical Trial. J. Gastrointest. Liver Dis. 2018, 27, 41–49. [Google Scholar] [CrossRef]
- Chambers, E.S.; Byrne, C.S.; Rugyendo, A.; Morrison, D.J.; Preston, T.; Tedford, C.; Bell, J.D.; Thomas, L.; Akbar, A.N.; Riddell, N.E.; et al. The effects of dietary supplementation with inulin and inulin-propionate ester on hepatic steatosis in adults with non-alcoholic fatty liver disease. Diabetes Obes. Metab. 2019, 21, 372–376. [Google Scholar] [CrossRef]
- Wahab, R.J.; Jaddoe, V.W.V.; Mezzoiuso, A.G.; Gaillard, R. Maternal polyunsaturated fatty acid concentrations during pregnancy and childhood liver fat accumulation. Clin. Nutr. 2022, 41, 847–854. [Google Scholar] [CrossRef]
- Fernandez-Hernando, C.; Ramirez, C.M.; Goedeke, L.; Suarez, Y. MicroRNAs in metabolic disease. Arterioscler. Thromb. Vasc. Biol. 2013, 33, 178–185. [Google Scholar] [CrossRef]
- He, X.; Li, Y.; Deng, X.; Xiao, X.; Zeng, J. Integrative evidence construction for resveratrol treatment of nonalcoholic fatty liver disease: Preclinical and clinical meta-analyses. Front. Pharmacol. 2023, 14, 1230783. [Google Scholar] [CrossRef] [PubMed]
- Zhu, K.; Kakkar, R.; Chahal, D.; Yoshida, E.M.; Hussaini, T. Efficacy and safety of semaglutide in non-alcoholic fatty liver disease. World J. Gastroenterol. 2023, 29, 5327–5338. [Google Scholar] [CrossRef] [PubMed]
- Armstrong, M.J.; Gaunt, P.; Aithal, G.P.; Barton, D.; Hull, D.; Parker, R.; Hazlehurst, J.M.; Guo, K.; LEAN Trial Team; Abouda, G.; et al. Liraglutide safety and efficacy in patients with non-alcoholic steatohepatitis (LEAN): A multicentre, double-blind, randomised, placebo-controlled phase 2 study. Lancet 2016, 387, 679–690. [Google Scholar] [CrossRef] [PubMed]
- Bridgeman, S.C.; Ellison, G.C.; Melton, P.E.; Newsholme, P.; Mamotte, C.D.S. Epigenetic effects of metformin: From molecular mechanisms to clinical implications. Diabetes Obes. Metab. 2018, 20, 1553–1562. [Google Scholar] [CrossRef] [PubMed]
- Huang, C.F.; Tiao, M.M.; Lin, I.C.; Huang, L.T.; Sheen, J.M.; Tain, Y.L.; Hsu, C.N.; Tsai, C.C.; Lin, Y.J.; Yu, H.R. Maternal Metformin Treatment Reprograms Maternal High-Fat Diet-Induced Hepatic Steatosis in Offspring Associated with Placental Glucose Transporter Modifications. Int. J. Mol. Sci. 2022, 23, 4239. [Google Scholar] [CrossRef] [PubMed]
- Schulthess, J.; Pandey, S.; Capitani, M.; Rue-Albrecht, K.C.; Arnold, I.; Franchini, F.; Chomka, A.; Ilott, N.E.; Johnston, D.G.W.; Pires, E.; et al. The Short Chain Fatty Acid Butyrate Imprints an Antimicrobial Program in Macrophages. Immunity 2019, 50, 432–445.e7. [Google Scholar] [CrossRef]
- Arpaia, N.; Campbell, C.; Fan, X.; Dikiy, S.; van der Veeken, J.; deRoos, P.; Liu, H.; Cross, J.R.; Pfeffer, K.; Coffer, P.J.; et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 2013, 504, 451–455. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Tzeng, H.-T.; Lee, W.-C. Impact of Transgenerational Nutrition on Nonalcoholic Fatty Liver Disease Development: Interplay between Gut Microbiota, Epigenetics and Immunity. Nutrients 2024, 16, 1388. https://doi.org/10.3390/nu16091388
Tzeng H-T, Lee W-C. Impact of Transgenerational Nutrition on Nonalcoholic Fatty Liver Disease Development: Interplay between Gut Microbiota, Epigenetics and Immunity. Nutrients. 2024; 16(9):1388. https://doi.org/10.3390/nu16091388
Chicago/Turabian StyleTzeng, Hong-Tai, and Wei-Chia Lee. 2024. "Impact of Transgenerational Nutrition on Nonalcoholic Fatty Liver Disease Development: Interplay between Gut Microbiota, Epigenetics and Immunity" Nutrients 16, no. 9: 1388. https://doi.org/10.3390/nu16091388
APA StyleTzeng, H. -T., & Lee, W. -C. (2024). Impact of Transgenerational Nutrition on Nonalcoholic Fatty Liver Disease Development: Interplay between Gut Microbiota, Epigenetics and Immunity. Nutrients, 16(9), 1388. https://doi.org/10.3390/nu16091388