Developmental Programming of the Fetal Immune System by Maternal Western-Style Diet: Mechanisms and Implications for Disease Pathways in the Offspring
Abstract
:1. Introduction
2. Immune Cell Development: Hematopoiesis
3. Impact of Maternal Obesity/WSD on Maternal and Placental Immune Function
3.1. Maternal Obesity Changes Placental Immunity
3.2. Cord Blood Changes in Offspring from Obese Pregnancies
4. Impact of Maternal Obesity on the Fetal Liver and Offspring Immunity
5. Microglia Activation by a Maternal WSD
6. Maternal Obesity Influences the Fetal Microbiome and Immune Development
7. Metabolites Regulate Fetal Metabolism and Immune Development
Metabolite | Mechanism | Effect | References |
---|---|---|---|
Acetate | FFAR2 and acetyl-CoA carboxylase | Promoted lipid metabolism and reduced appetite | [191,192] |
Propionate | FFAR2 and FFAR3 | Promoted glucose metabolism | [191] |
Butyrate | FFAR3 | Anti-inflammatory effects and promoted lipid metabolism | [191] |
Indole-lactic Acid | AHR and HCA3 | Improved gut health and immune responses | [193] |
Phenyllactic Acid | Mineral utilization | Increased lymphoid cell count without infection | [194] |
4-Hydroxyphenyllactic Acid | DCAR-1 | Triggered increased innate immune responses | [195] |
Tryptamine | AHR | Suppressed inflammation | [185] |
Indole-3-acetate | AHR | Suppressed inflammation | [185] |
Indole-3-ethanol | MyoIIA, ezrin | Maintained gut permeability | [186] |
Indole-3-pyruvate | MyoIIA, ezrin | Maintained gut permeability | [186] |
Indole-3-aldehye | MyoIIA, ezrin | Maintained gut permeability | [186] |
Indole-3-carbinol | UCPs, PPAR, sirtuin-1, leptin, aP2 | Decreased adipogenesis, thermogenesis, and inflammation | [187] |
8. Role of Epigenetic Programming in Immune Cells from Offspring Exposed to WSD
9. Conclusions and Future Directions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Finucane, M.M.; Stevens, G.A.; Cowan, M.J.; Danaei, G.; Lin, J.K.; Paciorek, C.J.; Singh, G.M.; Gutierrez, H.R.; Lu, Y.; Bahalim, A.N.; et al. National, regional, and global trends in body-mass index since 1980: Systematic analysis of health examination surveys and epidemiological studies with 960 country-years and 9.1 million participants. Lancet 2011, 377, 557–567. [Google Scholar] [CrossRef]
- Kelly, T.; Yang, W.; Chen, C.S.; Reynolds, K.; He, J. Global burden of obesity in 2005 and projections to 2030. Int. J. Obes. 2008, 32, 1431–1437. [Google Scholar] [CrossRef] [PubMed]
- Brines, J.; Rigourd, V.; Billeaud, C. The First 1000 Days of Infant. Healthcare 2022, 10, 106. [Google Scholar] [CrossRef]
- Likhar, A.; Patil, M.S. Importance of Maternal Nutrition in the First 1000 Days of Life and Its Effects on Child Development: A Narrative Review. Cureus 2022, 14, e30083. [Google Scholar] [CrossRef] [PubMed]
- Simione, M.; Moreno-Galarraga, L.; Perkins, M.; Price, S.N.; Luo, M.; Kotelchuck, M.; Blake-Lamb, T.L.; Taveras, E.M. Effects of the First 1000 Days Program, a systems-change intervention, on obesity risk factors during pregnancy. BMC Pregnancy Childbirth 2021, 21, 729. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Sureshchandra, S.; Doratt, B.M.; Mendza, N.; Varlamov, O.; Rincon, M.; Marshall, N.E.; Messaoudi, I. Maternal obesity blunts antimicrobial responses in fetal monocytes. eLife 2023, 12, e81320. [Google Scholar] [CrossRef] [PubMed]
- Kobayashi, M.; Yoshimoto, M. Multiple waves of fetal-derived immune cells constitute adult immune system. Immunol. Rev. 2023, 315, 11–30. [Google Scholar] [CrossRef] [PubMed]
- Mass, E.; Gentek, R. Fetal-Derived Immune Cells at the Roots of Lifelong Pathophysiology. Front. Cell Dev. Biol. 2021, 9, 648313. [Google Scholar] [CrossRef]
- Donald, K.; Finlay, B.B. Early-life interactions between the microbiota and immune system: Impact on immune system development and atopic disease. Nat. Rev. Immunol. 2023, 23, 735–748. [Google Scholar] [CrossRef]
- Palis, J.; Yoder, M.C. Yolk-sac hematopoiesis: The first blood cells of mouse and man. Exp. Hematol. 2001, 29, 927–936. [Google Scholar] [CrossRef] [PubMed]
- Brittain, T. Molecular aspects of embryonic hemoglobin function. Mol. Asp. Med. 2002, 23, 293–342. [Google Scholar] [CrossRef] [PubMed]
- Chia, S.L.; Kapoor, S.; Carvalho, C.; Bajenoff, M.; Gentek, R. Mast cell ontogeny: From fetal development to life-long health and disease. Immunol. Rev. 2023, 315, 31–53. [Google Scholar] [CrossRef] [PubMed]
- Ginhoux, F.; Greter, M.; Leboeuf, M.; Nandi, S.; See, P.; Gokhan, S.; Mehler, M.F.; Conway, S.J.; Ng, L.G.; Stanley, E.R.; et al. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science 2010, 330, 841–845. [Google Scholar] [CrossRef] [PubMed]
- Tober, J.; Koniski, A.; McGrath, K.E.; Vemishetti, R.; Emerson, R.; de Mesy-Bentley, K.K.; Waugh, R.; Palis, J. The megakaryocyte lineage originates from hemangioblast precursors and is an integral component both of primitive and of definitive hematopoiesis. Blood 2007, 109, 1433–1441. [Google Scholar] [CrossRef] [PubMed]
- McGrath, K.E.; Frame, J.M.; Fegan, K.H.; Bowen, J.R.; Conway, S.J.; Catherman, S.C.; Kingsley, P.D.; Koniski, A.D.; Palis, J. Distinct Sources of Hematopoietic Progenitors Emerge before HSCs and Provide Functional Blood Cells in the Mammalian Embryo. Cell Rep. 2015, 11, 1892–1904. [Google Scholar] [CrossRef] [PubMed]
- Gomez Perdiguero, E.; Klapproth, K.; Schulz, C.; Busch, K.; Azzoni, E.; Crozet, L.; Garner, H.; Trouillet, C.; de Bruijn, M.F.; Geissmann, F.; et al. Tissue-resident macrophages originate from yolk-sac-derived erythro-myeloid progenitors. Nature 2015, 518, 547–551. [Google Scholar] [CrossRef] [PubMed]
- Hoeffel, G.; Chen, J.; Lavin, Y.; Low, D.; Almeida, F.F.; See, P.; Beaudin, A.E.; Lum, J.; Low, I.; Forsberg, E.C.; et al. C-Myb+ erythro-myeloid progenitor-derived fetal monocytes give rise to adult tissue-resident macrophages. Immunity 2015, 42, 665–678. [Google Scholar] [CrossRef] [PubMed]
- Li, Z.; Liu, S.; Xu, J.; Zhang, X.; Han, D.; Liu, J.; Xia, M.; Yi, L.; Shen, Q.; Xu, S.; et al. Adult Connective Tissue-Resident Mast Cells Originate from Late Erythro-Myeloid Progenitors. Immunity 2018, 49, 640–653.e5. [Google Scholar] [CrossRef]
- Calvanese, V.; Mikkola, H.K.A. The genesis of human hematopoietic stem cells. Blood 2023, 142, 519–532. [Google Scholar] [CrossRef]
- Hall, T.; Sriram, P.; McKinney-Freeman, S. Murine Fetal Bone Marrow HSPCs Undergo a Dramatic Shift in Frequency at Birth. Blood 2019, 134, 2471. [Google Scholar] [CrossRef]
- Zhang, Y.; Liu, F. The evolving views of hematopoiesis: From embryo to adulthood and from in vivo to in vitro. J. Genet. Genom. 2023, 51, 3–15. [Google Scholar] [CrossRef] [PubMed]
- Sun, S.; Wijanarko, K.; Liani, O.; Strumila, K.; Ng, E.S.; Elefanty, A.G.; Stanley, E.G. Lymphoid cell development from fetal hematopoietic progenitors and human pluripotent stem cells. Immunol. Rev. 2023, 315, 154–170. [Google Scholar] [CrossRef] [PubMed]
- O’Byrne, S.; Elliott, N.; Rice, S.; Buck, G.; Fordham, N.; Garnett, C.; Godfrey, L.; Crump, N.T.; Wright, G.; Inglott, S.; et al. Discovery of a CD10-negative B-progenitor in human fetal life identifies unique ontogeny-related developmental programs. Blood 2019, 134, 1059–1071. [Google Scholar] [CrossRef] [PubMed]
- Yoshimoto, M.; Porayette, P.; Glosson, N.L.; Conway, S.J.; Carlesso, N.; Cardoso, A.A.; Kaplan, M.H.; Yoder, M.C. Autonomous murine T-cell progenitor production in the extra-embryonic yolk sac before HSC emergence. Blood 2012, 119, 5706–5714. [Google Scholar] [CrossRef] [PubMed]
- Luis, T.C.; Luc, S.; Mizukami, T.; Boukarabila, H.; Thongjuea, S.; Woll, P.S.; Azzoni, E.; Giustacchini, A.; Lutteropp, M.; Bouriez-Jones, T.; et al. Initial seeding of the embryonic thymus by immune-restricted lympho-myeloid progenitors. Nat. Immunol. 2016, 17, 1424–1435. [Google Scholar] [CrossRef] [PubMed]
- Krangel, M.S. Mechanics of T cell receptor gene rearrangement. Curr. Opin. Immunol. 2009, 21, 133–139. [Google Scholar] [CrossRef] [PubMed]
- Jackson, T.R.; Ling, R.E.; Roy, A. The Origin of B-cells: Human Fetal B Cell Development and Implications for the Pathogenesis of Childhood Acute Lymphoblastic Leukemia. Front. Immunol. 2021, 12, 637975. [Google Scholar] [CrossRef] [PubMed]
- Pabst, O.; Nowosad, C.R. B cells and the intestinal microbiome in time, space and place. Semin. Immunol. 2023, 69, 101806. [Google Scholar] [CrossRef]
- Dorshkind, K.; Crooks, G. Layered immune system development in mice and humans. Immunol. Rev. 2023, 315, 5–10. [Google Scholar] [CrossRef]
- Racicot, K.; Kwon, J.Y.; Aldo, P.; Silasi, M.; Mor, G. Understanding the complexity of the immune system during pregnancy. Am. J. Reprod. Immunol. 2014, 72, 107–116. [Google Scholar] [CrossRef] [PubMed]
- Wanaditya, G.K.; Putra, I.W.A.; Aryana, M.B.D.; Mulyana, R.S. Obesity in Pregnant Women and Its Impact on Maternal and Neonatal Morbidity. Eur. J. Med. Health Sci. 2023, 5, 17–21. [Google Scholar] [CrossRef]
- Bravo-Flores, E.; Mancilla-Herrera, I.; Espino, Y.S.S.; Ortiz-Ramirez, M.; Flores-Rueda, V.; Ibarguengoitia-Ochoa, F.; Ibanez, C.A.; Zambrano, E.; Solis-Paredes, M.; Perichart-Perera, O.; et al. Macrophage Populations in Visceral Adipose Tissue from Pregnant Women: Potential Role of Obesity in Maternal Inflammation. Int. J. Mol. Sci. 2018, 19, 1074. [Google Scholar] [CrossRef] [PubMed]
- Pendeloski, K.P.T.; Ono, E.; Torloni, M.R.; Mattar, R.; Daher, S. Maternal obesity and inflammatory mediators: A controversial association. Am. J. Reprod. Immunol. 2017, 77, e12674. [Google Scholar] [CrossRef] [PubMed]
- Tinius, R.A.; Blankenship, M.M.; Furgal, K.E.; Cade, W.T.; Pearson, K.J.; Rowland, N.S.; Pearson, R.C.; Hoover, D.L.; Maples, J.M. Metabolic flexibility is impaired in women who are pregnant and overweight/obese and related to insulin resistance and inflammation. Metabolism 2020, 104, 154142. [Google Scholar] [CrossRef] [PubMed]
- Pantham, P.; Aye, I.L.; Powell, T.L. Inflammation in maternal obesity and gestational diabetes mellitus. Placenta 2015, 36, 709–715. [Google Scholar] [CrossRef] [PubMed]
- Sureshchandra, S.; Marshall, N.E.; Wilson, R.M.; Barr, T.; Rais, M.; Purnell, J.Q.; Thornburg, K.L.; Messaoudi, I. Inflammatory Determinants of Pregravid Obesity in Placenta and Peripheral Blood. Front. Physiol. 2018, 9, 1089. [Google Scholar] [CrossRef] [PubMed]
- Sen, S.; Iyer, C.; Klebenov, D.; Histed, A.; Aviles, J.A.; Meydani, S.N. Obesity impairs cell-mediated immunity during the second trimester of pregnancy. Am. J. Obstet. Gynecol. 2013, 208, 139.e1–139.e8. [Google Scholar] [CrossRef] [PubMed]
- Moffett-King, A. Natural killer cells and pregnancy. Nat. Rev. Immunol. 2002, 2, 656–663. [Google Scholar] [CrossRef]
- Nagamatsu, T.; Schust, D.J. The contribution of macrophages to normal and pathological pregnancies. Am. J. Reprod. Immunol. 2010, 63, 460–471. [Google Scholar] [CrossRef]
- Suryawanshi, H.; Morozov, P.; Straus, A.; Sahasrabudhe, N.; Max, K.E.A.; Garzia, A.; Kustagi, M.; Tuschl, T.; Williams, Z. A single-cell survey of the human first-trimester placenta and decidua. Sci. Adv. 2018, 4, eaau4788. [Google Scholar] [CrossRef] [PubMed]
- Burke, S.D.; Barrette, V.F.; Gravel, J.; Carter, A.L.; Hatta, K.; Zhang, J.; Chen, Z.; Leno-Duran, E.; Bianco, J.; Leonard, S.; et al. Uterine NK cells, spiral artery modification and the regulation of blood pressure during mouse pregnancy. Am. J. Reprod. Immunol. 2010, 63, 472–481. [Google Scholar] [CrossRef] [PubMed]
- Yao, Y.; Xu, X.H.; Jin, L. Macrophage Polarization in Physiological and Pathological Pregnancy. Front. Immunol. 2019, 10, 792. [Google Scholar] [CrossRef] [PubMed]
- Paul, N.; Sultana, Z.; Fisher, J.J.; Maiti, K.; Smith, R. Extracellular vesicles- crucial players in human pregnancy. Placenta 2023, 140, 30–38. [Google Scholar] [CrossRef] [PubMed]
- Zhang, T.N.; Wang, W.; Huang, X.M.; Gao, S.Y. Non-Coding RNAs and Extracellular Vehicles: Their Role in the Pathogenesis of Gestational Diabetes Mellitus. Front. Endocrinol. 2021, 12, 664287. [Google Scholar] [CrossRef]
- Mir, I.N.; Chalak, L.F.; Liao, J.; Johnson-Welch, S.; Brown, L.S.; Longoria, C.; Savani, R.C.; Rosenfeld, C.R. Fetal-placental crosstalk occurs through fetal cytokine synthesis and placental clearance. Placenta 2018, 69, 1–8. [Google Scholar] [CrossRef] [PubMed]
- Sacks, G.P.; Studena, K.; Sargent, K.; Redman, C.W. Normal pregnancy and preeclampsia both produce inflammatory changes in peripheral blood leukocytes akin to those of sepsis. Am. J. Obs. Gynecol. 1998, 179, 80–86. [Google Scholar] [CrossRef] [PubMed]
- Batorsky, R.; Ceasrine, A.M.; Shook, L.L.; Kislal, S.; Bordt, E.A.; Devlin, B.A.; Perlis, R.H.; Slonim, D.K.; Bilbo, S.D.; Edlow, A.G. Hofbauer cells and fetal brain microglia share transcriptional profiles and responses to maternal diet-induced obesity. bioRxiv 2023. [Google Scholar] [CrossRef]
- Castellana, B.; Perdu, S.; Kim, Y.; Chan, K.; Atif, J.; Marziali, M.; Beristain, A.G. Maternal obesity alters uterine NK activity through a functional KIR2DL1/S1 imbalance. Immunol. Cell Biol. 2018, 96, 805–819. [Google Scholar] [CrossRef]
- Fukui, A.; Funamizu, A.; Yokota, M.; Yamada, K.; Nakamua, R.; Fukuhara, R.; Kimura, H.; Mizunuma, H. Uterine and circulating natural killer cells and their roles in women with recurrent pregnancy loss, implantation failure and preeclampsia. J. Reprod. Immunol. 2011, 90, 105–110. [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]
- Challier, J.C.; Basu, S.; Bintein, T.; Minium, J.; Hotmire, K.; Catalano, P.M.; Hauguel-de Mouzon, S. Obesity in pregnancy stimulates macrophage accumulation and inflammation in the placenta. Placenta 2008, 29, 274–281. [Google Scholar] [CrossRef]
- Wilson, R.M.; Marshall, N.E.; Jeske, D.R.; Purnell, J.Q.; Thornburg, K.; Messaoudi, I. Maternal obesity alters immune cell frequencies and responses in umbilical cord blood samples. Pediatr. Allergy Immunol. 2015, 26, 344–351. [Google Scholar] [CrossRef]
- Basu, S.; Haghiac, M.; Surace, P.; Challier, J.C.; Guerre-Millo, M.; Singh, K.; Waters, T.; Minium, J.; Presley, L.; Catalano, P.M.; et al. Pregravid obesity associates with increased maternal endotoxemia and metabolic inflammation. Obesity 2011, 19, 476–482. [Google Scholar] [CrossRef] [PubMed]
- Esteve-Sole, A.; Luo, Y.; Vlagea, A.; Deya-Martinez, A.; Yague, J.; Plaza-Martin, A.M.; Juan, M.; Alsina, L. B Regulatory Cells: Players in Pregnancy and Early Life. Int. J. Mol. Sci. 2018, 19, 2099. [Google Scholar] [CrossRef] [PubMed]
- Rosser, E.C.; Mauri, C. Regulatory B cells: Origin, phenotype, and function. Immunity 2015, 42, 607–612. [Google Scholar] [CrossRef]
- Flores-Borja, F.; Bosma, A.; Ng, D.; Reddy, V.; Ehrenstein, M.R.; Isenberg, D.A.; Mauri, C. CD19+CD24hiCD38hi B cells maintain regulatory T cells while limiting TH1 and TH17 differentiation. Sci. Transl. Med. 2013, 5, 173ra123. [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] [PubMed]
- Sureshchandra, S.; Mendoza, N.; Jankeel, A.; Wilson, R.M.; Marshall, N.E.; Messaoudi, I. Phenotypic and Epigenetic Adaptations of Cord Blood CD4+ T Cells to Maternal Obesity. Front. Immunol. 2021, 12, 617592. [Google Scholar] [CrossRef]
- Dosch, N.C.; Guslits, E.F.; Weber, M.B.; Murray, S.E.; Ha, B.; Coe, C.L.; Auger, A.P.; Kling, P.J. Maternal Obesity Affects Inflammatory and Iron Indices in Umbilical Cord Blood. J. Pediatr. 2016, 172, 20–28. [Google Scholar] [CrossRef]
- McCloskey, K.; Ponsonby, A.L.; Collier, F.; Allen, K.; Tang, M.L.K.; Carlin, J.B.; Saffery, R.; Skilton, M.R.; Cheung, M.; Ranganathan, S.; et al. The association between higher maternal pre-pregnancy body mass index and increased birth weight, adiposity and inflammation in the newborn. Pediatr. Obes. 2018, 13, 46–53. [Google Scholar] [CrossRef] [PubMed]
- Sureshchandra, S.; Marshall, N.E.; Messaoudi, I. Impact of pregravid obesity on maternal and fetal immunity: Fertile grounds for reprogramming. J. Leukoc. Biol. 2019, 106, 1035–1050. [Google Scholar] [CrossRef] [PubMed]
- Chan, W.K.; Chuah, K.H.; Rajaram, R.B.; Lim, L.L.; Ratnasingam, J.; Vethakkan, S.R. Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD): A State-of-the-Art Review. J. Obes. Metab. Syndr. 2023, 32, 197–213. [Google Scholar] [CrossRef]
- 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] [PubMed]
- Gallardo, J.M.; Gomez-Lopez, J.; Medina-Bravo, P.; Juarez-Sanchez, F.; Contreras-Ramos, A.; Galicia-Esquivel, M.; Sanchez-Urbina, R.; Klunder-Klunder, M. Maternal obesity increases oxidative stress in the newborn. Obesity 2015, 23, 1650–1654. [Google Scholar] [CrossRef] [PubMed]
- Brumbaugh, D.E.; Friedman, J.E. Developmental origins of nonalcoholic fatty liver disease. Pediatr. Res. 2014, 75, 140–147. [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] [PubMed]
- Mouralidarane, A.; Soeda, J.; Visconti-Pugmire, C.; Samuelsson, A.M.; Pombo, J.; Maragkoudaki, X.; Butt, A.; Saraswati, R.; Novelli, M.; Fusai, G.; et al. Maternal obesity programs offspring nonalcoholic fatty liver disease by innate immune dysfunction in mice. Hepatology 2013, 58, 128–138. [Google Scholar] [CrossRef] [PubMed]
- Cohen, C.C.; Francis, E.C.; Perng, W.; Sauder, K.A.; Scherzinger, A.; Sundaram, S.S.; Shankar, K.; Dabelea, D. Exposure to maternal fuels during pregnancy and offspring hepatic fat in early childhood: The healthy start study. Pediatr. Obes. 2022, 17, e12902. [Google Scholar] [CrossRef]
- O’Rourke, R.W. Metabolic thrift and the genetic basis of human obesity. Ann. Surg. 2014, 259, 642–648. [Google Scholar] [CrossRef]
- Wells, J.C. Thrift: A guide to thrifty genes, thrifty phenotypes and thrifty norms. Int. J. Obes. 2009, 33, 1331–1338. [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]
- Tacke, F. Targeting hepatic macrophages to treat liver diseases. J. Hepatol. 2017, 66, 1300–1312. [Google Scholar] [CrossRef] [PubMed]
- Heymann, F.; Hammerich, L.; Storch, D.; Bartneck, M.; Huss, S.; Russeler, V.; Gassler, N.; Lira, S.A.; Luedde, T.; Trautwein, C.; et al. Hepatic macrophage migration and differentiation critical for liver fibrosis is mediated by the chemokine receptor C-C motif chemokine receptor 8 in mice. Hepatology 2012, 55, 898–909. [Google Scholar] [CrossRef] [PubMed]
- Friedman, S.L.; Ratziu, V.; Harrison, S.A.; Abdelmalek, M.F.; Aithal, G.P.; Caballeria, J.; Francque, S.; Farrell, G.; Kowdley, K.V.; Craxi, A.; et al. A randomized, placebo-controlled trial of cenicriviroc for treatment of nonalcoholic steatohepatitis with fibrosis. Hepatology 2018, 67, 1754–1767. [Google Scholar] [CrossRef] [PubMed]
- Pitzalis, C.; Jones, G.W.; Bombardieri, M.; Jones, S.A. Ectopic lymphoid-like structures in infection, cancer and autoimmunity. Nat. Rev. Immunol. 2014, 14, 447–462. [Google Scholar] [CrossRef] [PubMed]
- Sutti, S.; Jindal, A.; Locatelli, I.; Vacchiano, M.; Gigliotti, L.; Bozzola, C.; Albano, E. Adaptive immune responses triggered by oxidative stress contribute to hepatic inflammation in NASH. Hepatology 2014, 59, 886–897. [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-γ 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]
- Ferreyra Solari, N.E.; Inzaugarat, M.E.; Baz, P.; De Matteo, E.; Lezama, C.; Galoppo, M.; Galoppo, C.; Chernavsky, A.C. The role of innate cells is coupled to a Th1-polarized immune response in pediatric nonalcoholic steatohepatitis. J. Clin. Immunol. 2012, 32, 611–621. [Google Scholar] [CrossRef]
- 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]
- 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]
- Iwasaki, A.; Medzhitov, R. Control of adaptive immunity by the innate immune system. Nat. Immunol. 2015, 16, 343–353. [Google Scholar] [CrossRef] [PubMed]
- Netea, M.G.; Joosten, L.A.; Latz, E.; Mills, K.H.; Natoli, G.; Stunnenberg, H.G.; O’Neill, L.A.; Xavier, R.J. Trained immunity: A program of innate immune memory in health and disease. Science 2016, 352, aaf1098. [Google Scholar] [CrossRef] [PubMed]
- Kucuksezer, U.C.; Ozdemir, C.; Akdis, M.; Akdis, C.A. Influence of Innate Immunity on Immune Tolerance. Acta Med. Acad. 2020, 49, 164–180. [Google Scholar] [CrossRef] [PubMed]
- Bauer, M.; Weis, S.; Netea, M.G.; Wetzker, R. Remembering Pathogen Dose: Long-Term Adaptation in Innate Immunity. Trends Immunol. 2018, 39, 438–445. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Ghazarian, M.; Revelo, X.S.; Nohr, M.K.; Luck, H.; Zeng, K.; Lei, H.; Tsai, S.; Schroer, S.A.; Park, Y.J.; Chng, M.H.Y.; et al. Type I Interferon Responses Drive Intrahepatic T cells to Promote Metabolic Syndrome. Sci. Immunol. 2017, 2, eaai7616. [Google Scholar] [CrossRef] [PubMed]
- Grohmann, M.; Wiede, F.; Dodd, G.T.; Gurzov, E.N.; Ooi, G.J.; Butt, T.; Rasmiena, A.A.; Kaur, S.; Gulati, T.; Goh, P.K.; et al. Obesity Drives STAT-1-Dependent NASH and STAT-3-Dependent HCC. Cell 2018, 175, 1289–1306.e20. [Google Scholar] [CrossRef] [PubMed]
- Thapa, M.; Chinnadurai, R.; Velazquez, V.M.; Tedesco, D.; Elrod, E.; Han, J.H.; Sharma, P.; Ibegbu, C.; Gewirtz, A.; Anania, F.; et al. Liver fibrosis occurs through dysregulation of MyD88-dependent innate B-cell activity. Hepatology 2015, 61, 2067–2079. [Google Scholar] [CrossRef]
- Odaka, Y.; Nakano, M.; Tanaka, T.; Kaburagi, T.; Yoshino, H.; Sato-Mito, N.; Sato, K. The influence of a high-fat dietary environment in the fetal period on postnatal metabolic and immune function. Obesity 2010, 18, 1688–1694. [Google Scholar] [CrossRef]
- Tsiantoulas, D.; Sage, A.P.; Mallat, Z.; Binder, C.J. Targeting B cells in atherosclerosis: Closing the gap from bench to bedside. Arter. Thromb. Vasc. Biol. 2015, 35, 296–302. [Google Scholar] [CrossRef] [PubMed]
- Magalhaes, M.S.; Potter, H.G.; Ahlback, A.; Gentek, R. Developmental programming of macrophages by early life adversity. Int. Rev. Cell Mol. Biol. 2022, 368, 213–259. [Google Scholar] [CrossRef] [PubMed]
- Saijo, K.; Glass, C.K. Microglial cell origin and phenotypes in health and disease. Nat. Rev. Immunol. 2011, 11, 775–787. [Google Scholar] [CrossRef] [PubMed]
- Rivera, H.M.; Christiansen, K.J.; Sullivan, E.L. The role of maternal obesity in the risk of neuropsychiatric disorders. Front. Neurosci. 2015, 9, 194. [Google Scholar] [CrossRef] [PubMed]
- Mattei, D.; Ivanov, A.; Ferrai, C.; Jordan, P.; Guneykaya, D.; Buonfiglioli, A.; Schaafsma, W.; Przanowski, P.; Deuther-Conrad, W.; Brust, P.; et al. Maternal immune activation results in complex microglial transcriptome signature in the adult offspring that is reversed by minocycline treatment. Transl. Psychiatry 2017, 7, e1120. [Google Scholar] [CrossRef] [PubMed]
- Solmi, M.; Veronese, N.; Thapa, N.; Facchini, S.; Stubbs, B.; Fornaro, M.; Carvalho, A.F.; Correll, C.U. Systematic review and meta-analysis of the efficacy and safety of minocycline in schizophrenia. CNS Spectr. 2017, 22, 415–426. [Google Scholar] [CrossRef] [PubMed]
- Dunn, G.A.; Mitchell, A.J.; Selby, M.; Fair, D.A.; Gustafsson, H.C.; Sullivan, E.L. Maternal diet and obesity shape offspring central and peripheral inflammatory outcomes in juvenile non-human primates. Brain Behav. Immun. 2022, 102, 224–236. [Google Scholar] [CrossRef] [PubMed]
- Mitchell, A.J.; Khambadkone, S.G.; Dunn, G.; Bagley, J.; Tamashiro, K.L.K.; Fair, D.; Gustafsson, H.; Sullivan, E.L. Maternal Western-style diet reduces social engagement and increases idiosyncratic behavior in Japanese macaque offspring. Brain Behav. Immun. 2022, 105, 109–121. [Google Scholar] [CrossRef] [PubMed]
- Papadakis, S.; Thompson, J.R.; Feczko, E.; Miranda-Dominguez, O.; Dunn, G.A.; Selby, M.; Mitchell, A.J.; Sullivan, E.L.; Fair, D.A. Perinatal Western-style diet exposure associated with decreased microglial counts throughout the arcuate nucleus of the hypothalamus in Japanese macaques. J. Neurophysiol. 2024, 131, 241–260. [Google Scholar] [CrossRef]
- Subhramanyam, C.S.; Wang, C.; Hu, Q.; Dheen, S.T. Microglia-mediated neuroinflammation in neurodegenerative diseases. Semin. Cell Dev. Biol. 2019, 94, 112–120. [Google Scholar] [CrossRef]
- Guo, D.H.; Yamamoto, M.; Hernandez, C.M.; Khodadadi, H.; Baban, B.; Stranahan, A.M. Visceral adipose NLRP3 impairs cognition in obesity via IL-1R1 on CX3CR1+ cells. J. Clin. Investig. 2020, 130, 1961–1976. [Google Scholar] [CrossRef] [PubMed]
- Smith, S.E.; Li, J.; Garbett, K.; Mirnics, K.; Patterson, P.H. Maternal immune activation alters fetal brain development through interleukin-6. J. Neurosci. 2007, 27, 10695–10702. [Google Scholar] [CrossRef] [PubMed]
- Sasaki, T.; Tome, S.; Takei, Y. Intraventricular IL-17A administration activates microglia and alters their localization in the mouse embryo cerebral cortex. Mol. Brain 2020, 13, 93. [Google Scholar] [CrossRef] [PubMed]
- Aron-Wisnewsky, J.; Vigliotti, C.; Witjes, J.; Le, P.; Holleboom, A.G.; Verheij, J.; Nieuwdorp, M.; Clement, K. Gut microbiota and human NAFLD: Disentangling microbial signatures from metabolic disorders. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 279–297. [Google Scholar] [CrossRef] [PubMed]
- Conway, J.; Duggal, N.A. Ageing of the gut microbiome: Potential influences on immune senescence and inflammageing. Ageing Res. Rev. 2021, 68, 101323. [Google Scholar] [CrossRef] [PubMed]
- Nichols, R.G.; Davenport, E.R. The relationship between the gut microbiome and host gene expression: A review. Hum. Genet. 2021, 140, 747–760. [Google Scholar] [CrossRef] [PubMed]
- Cox, T.O.; Lundgren, P.; Nath, K.; Thaiss, C.A. Metabolic control by the microbiome. Genome Med. 2022, 14, 80. [Google Scholar] [CrossRef]
- Human Microbiome Project, C. Structure, function and diversity of the healthy human microbiome. Nature 2012, 486, 207–214. [Google Scholar] [CrossRef]
- Turnbaugh, P.J.; Ley, R.E.; Hamady, M.; Fraser-Liggett, C.M.; Knight, R.; Gordon, J.I. The human microbiome project. Nature 2007, 449, 804–810. [Google Scholar] [CrossRef]
- Ley, R.E.; Backhed, F.; Turnbaugh, P.; Lozupone, C.A.; Knight, R.D.; Gordon, J.I. Obesity alters gut microbial ecology. Proc. Natl. Acad. Sci. USA 2005, 102, 11070–11075. [Google Scholar] [CrossRef]
- Turnbaugh, P.J.; Ley, R.E.; Mahowald, M.A.; Magrini, V.; Mardis, E.R.; Gordon, J.I. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 2006, 444, 1027–1031. [Google Scholar] [CrossRef] [PubMed]
- David, L.A.; Maurice, C.F.; Carmody, R.N.; Gootenberg, D.B.; Button, J.E.; Wolfe, B.E.; Ling, A.V.; Devlin, A.S.; Varma, Y.; Fischbach, M.A.; et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014, 505, 559–563. [Google Scholar] [CrossRef] [PubMed]
- Wilson, A.S.; Koller, K.R.; Ramaboli, M.C.; Nesengani, L.T.; Ocvirk, S.; Chen, C.; Flanagan, C.A.; Sapp, F.R.; Merritt, Z.T.; Bhatti, F.; et al. Diet and the Human Gut Microbiome: An International Review. Dig. Dis. Sci. 2020, 65, 723–740. [Google Scholar] [CrossRef] [PubMed]
- Singh, R.K.; Chang, H.W.; Yan, D.; Lee, K.M.; Ucmak, D.; Wong, K.; Abrouk, M.; Farahnik, B.; Nakamura, M.; Zhu, T.H.; et al. Influence of diet on the gut microbiome and implications for human health. J. Transl. Med. 2017, 15, 73. [Google Scholar] [CrossRef] [PubMed]
- Koren, O.; Konnikova, L.; Brodin, P.; Mysorekar, I.U.; Collado, M.C. The maternal gut microbiome in pregnancy: Implications for the developing immune system. Nat. Rev. Gastroenterol. Hepatol. 2024, 21, 35–45. [Google Scholar] [CrossRef] [PubMed]
- Jennison, E.; Byrne, C.D. The role of the gut microbiome and diet in the pathogenesis of non-alcoholic fatty liver disease. Clin. Mol. Hepatol. 2021, 27, 22–43. [Google Scholar] [CrossRef]
- Kuang, Y.S.; Lu, J.H.; Li, S.H.; Li, J.H.; Yuan, M.Y.; He, J.R.; Chen, N.N.; Xiao, W.Q.; Shen, S.Y.; Qiu, L.; et al. Connections between the human gut microbiome and gestational diabetes mellitus. Gigascience 2017, 6, gix058. [Google Scholar] [CrossRef]
- Sugino, K.Y.; Hernandez, T.L.; Barbour, L.A.; Kofonow, J.M.; Frank, D.N.; Friedman, J.E. A maternal higher-complex carbohydrate diet increases bifidobacteria and alters early life acquisition of the infant microbiome in women with gestational diabetes mellitus. Front. Endocrinol. 2022, 13, 921464. [Google Scholar] [CrossRef]
- Miko, E.; Csaszar, A.; Bodis, J.; Kovacs, K. The Maternal-Fetal Gut Microbiota Axis: Physiological Changes, Dietary Influence, and Modulation Possibilities. Life 2022, 12, 424. [Google Scholar] [CrossRef]
- Arboleya, S.; Suarez, M.; Fernandez, N.; Mantecon, L.; Solis, G.; Gueimonde, M.; de Los Reyes-Gavilan, C.G. C-section and the Neonatal Gut Microbiome Acquisition: Consequences for Future Health. Ann. Nutr. Metab. 2018, 73 (Suppl. S3), 17–23. [Google Scholar] [CrossRef]
- Rios-Covian, D.; Langella, P.; Martin, R. From Short- to Long-Term Effects of C-Section Delivery on Microbiome Establishment and Host Health. Microorganisms 2021, 9, 2122. [Google Scholar] [CrossRef] [PubMed]
- DeVries, A.; McCauley, K.; Fadrosh, D.; Fujimura, K.E.; Stern, D.A.; Lynch, S.V.; Vercelli, D. Maternal prenatal immunity, neonatal trained immunity, and early airway microbiota shape childhood asthma development. Allergy 2022, 77, 3617–3628. [Google Scholar] [CrossRef] [PubMed]
- Thorburn, A.N.; McKenzie, C.I.; Shen, S.; Stanley, D.; Macia, L.; Mason, L.J.; Roberts, L.K.; Wong, C.H.; Shim, R.; Robert, R.; et al. Evidence that asthma is a developmental origin disease influenced by maternal diet and bacterial metabolites. Nat. Commun. 2015, 6, 7320. [Google Scholar] [CrossRef] [PubMed]
- Kimura, I.; Miyamoto, J.; Ohue-Kitano, R.; Watanabe, K.; Yamada, T.; Onuki, M.; Aoki, R.; Isobe, Y.; Kashihara, D.; Inoue, D.; et al. Maternal gut microbiota in pregnancy influences offspring metabolic phenotype in mice. Science 2020, 367, eaaw8429. [Google Scholar] [CrossRef] [PubMed]
- van Dijk, S.J.; Zhou, J.; Peters, T.J.; Buckley, M.; Sutcliffe, B.; Oytam, Y.; Gibson, R.A.; McPhee, A.; Yelland, L.N.; Makrides, M.; et al. Effect of prenatal DHA supplementation on the infant epigenome: Results from a randomized controlled trial. Clin. Epigenet. 2016, 8, 114. [Google Scholar] [CrossRef] [PubMed]
- Yassour, M.; Vatanen, T.; Siljander, H.; Hamalainen, A.M.; Harkonen, T.; Ryhanen, S.J.; Franzosa, E.A.; Vlamakis, H.; Huttenhower, C.; Gevers, D.; et al. Natural history of the infant gut microbiome and impact of antibiotic treatment on bacterial strain diversity and stability. Sci. Transl. Med. 2016, 8, 343ra381. [Google Scholar] [CrossRef] [PubMed]
- Wang, S.; Ryan, C.A.; Boyaval, P.; Dempsey, E.M.; Ross, R.P.; Stanton, C. Maternal Vertical Transmission Affecting Early-Life Microbiota Development. Trends Microbiol. 2020, 28, 28–45. [Google Scholar] [CrossRef] [PubMed]
- Li, W.; Tapiainen, T.; Brinkac, L.; Lorenzi, H.A.; Moncera, K.; Tejesvi, M.V.; Salo, J.; Nelson, K.E. Vertical Transmission of Gut Microbiome and Antimicrobial Resistance Genes in Infants Exposed to Antibiotics at Birth. J. Infect. Dis. 2021, 224, 1236–1246. [Google Scholar] [CrossRef] [PubMed]
- Guarner, F.; Bourdet-Sicard, R.; Brandtzaeg, P.; Gill, H.S.; McGuirk, P.; van Eden, W.; Versalovic, J.; Weinstock, J.V.; Rook, G.A. Mechanisms of disease: The hygiene hypothesis revisited. Nat. Clin. Pract. Gastroenterol. Hepatol. 2006, 3, 275–284. [Google Scholar] [CrossRef]
- 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]
- Hong, J.Y.; Medzhitov, R. On developmental programming of the immune system. Trends Immunol. 2023, 44, 877–889. [Google Scholar] [CrossRef] [PubMed]
- Lin, C.; Lin, Y.; Zhang, H.; Wang, G.; Zhao, J.; Zhang, H.; Chen, W. Intestinal ‘Infant-Type’ Bifidobacteria Mediate Immune System Development in the First 1000 Days of Life. Nutrients 2022, 14, 1498. [Google Scholar] [CrossRef] [PubMed]
- Turroni, F.; Peano, C.; Pass, D.A.; Foroni, E.; Severgnini, M.; Claesson, M.J.; Kerr, C.; Hourihane, J.; Murray, D.; Fuligni, F.; et al. Diversity of bifidobacteria within the infant gut microbiota. PLoS ONE 2012, 7, e36957. [Google Scholar] [CrossRef] [PubMed]
- Turroni, F.; van Sinderen, D.; Ventura, M. Genomics and ecological overview of the genus Bifidobacterium. Int. J. Food Microbiol. 2011, 149, 37–44. [Google Scholar] [CrossRef] [PubMed]
- Asakuma, S.; Hatakeyama, E.; Urashima, T.; Yoshida, E.; Katayama, T.; Yamamoto, K.; Kumagai, H.; Ashida, H.; Hirose, J.; Kitaoka, M. Physiology of consumption of human milk oligosaccharides by infant gut-associated bifidobacteria. J. Biol. Chem. 2011, 286, 34583–34592. [Google Scholar] [CrossRef] [PubMed]
- Russell, J.T.; Roesch, L.F.W.; Ordberg, M.; Ilonen, J.; Atkinson, M.A.; Schatz, D.A.; Triplett, E.W.; Ludvigsson, J. Genetic risk for autoimmunity is associated with distinct changes in the human gut microbiome. Nat. Commun. 2019, 10, 3621. [Google Scholar] [CrossRef] [PubMed]
- Vatanen, T.; Kostic, A.D.; d’Hennezel, E.; Siljander, H.; Franzosa, E.A.; Yassour, M.; Kolde, R.; Vlamakis, H.; Arthur, T.D.; Hamalainen, A.M.; et al. Variation in Microbiome LPS Immunogenicity Contributes to Autoimmunity in Humans. Cell 2016, 165, 842–853. [Google Scholar] [CrossRef] [PubMed]
- Ley, R.E.; Turnbaugh, P.J.; Klein, S.; Gordon, J.I. Microbial ecology: Human gut microbes associated with obesity. Nature 2006, 444, 1022–1023. [Google Scholar] [CrossRef]
- Weaver, L.K.; Minichino, D.; Biswas, C.; Chu, N.; Lee, J.J.; Bittinger, K.; Albeituni, S.; Nichols, K.E.; Behrens, E.M. Microbiota-dependent signals are required to sustain TLR-mediated immune responses. JCI Insight 2019, 4, e124370. [Google Scholar] [CrossRef]
- Soderborg, T.K.; Carpenter, C.M.; Janssen, R.C.; Weir, T.L.; Robertson, C.E.; Ir, D.; Young, B.E.; Krebs, N.F.; Hernandez, T.L.; Barbour, L.A.; et al. Gestational Diabetes Is Uniquely Associated With Altered Early Seeding of the Infant Gut Microbiota. Front. Endocrinol. 2020, 11, 603021. [Google Scholar] [CrossRef]
- Chen, T.; Qin, Y.; Chen, M.; Zhang, Y.; Wang, X.; Dong, T.; Chen, G.; Sun, X.; Lu, T.; White, R.A., III; et al. Gestational diabetes mellitus is associated with the neonatal gut microbiota and metabolome. BMC Med. 2021, 19, 120. [Google Scholar] [CrossRef] [PubMed]
- Pinto, Y.; Frishman, S.; Turjeman, S.; Eshel, A.; Nuriel-Ohayon, M.; Shrossel, O.; Ziv, O.; Walters, W.; Parsonnet, J.; Ley, C.; et al. Gestational diabetes is driven by microbiota-induced inflammation months before diagnosis. Gut 2023, 72, 918–928. [Google Scholar] [CrossRef] [PubMed]
- Jones, J.M.; Reinke, S.N.; Mousavi-Derazmahalleh, M.; Garssen, J.; Jenmalm, M.C.; Srinivasjois, R.; Silva, D.; Keelan, J.; Prescott, S.L.; Palmer, D.J.; et al. Maternal prebiotic supplementation during pregnancy and lactation modifies the microbiome and short chain fatty acid profile of both mother and infant. Clin. Nutr. 2024, 43, 969–980. [Google Scholar] [CrossRef] [PubMed]
- Reynolds, H.M.; Bettini, M.L. Early-life microbiota-immune homeostasis. Front. Immunol. 2023, 14, 1266876. [Google Scholar] [CrossRef] [PubMed]
- Barman, P.K.; Goodridge, H.S. Microbial Sensing by Hematopoietic Stem and Progenitor Cells. Stem Cells 2022, 40, 14–21. [Google Scholar] [CrossRef] [PubMed]
- Paucar Iza, Y.A.; Brown, C.C. Early life imprinting of intestinal immune tolerance and tissue homeostasis. Immunol. Rev. 2024, 323, 303–315. [Google Scholar] [CrossRef] [PubMed]
- Kohler, A.; Delbauve, S.; Smout, J.; Torres, D.; Flamand, V. Very early-life exposure to microbiota-induced TNF drives the maturation of neonatal pre-cDC1. Gut 2021, 70, 511–521. [Google Scholar] [CrossRef]
- Constantinides, M.G.; Belkaid, Y. Early-life imprinting of unconventional T cells and tissue homeostasis. Science 2021, 374, eabf0095. [Google Scholar] [CrossRef] [PubMed]
- Sefik, E.; Geva-Zatorsky, N.; Oh, S.; Konnikova, L.; Zemmour, D.; McGuire, A.M.; Burzyn, D.; Ortiz-Lopez, A.; Lobera, M.; Yang, J.; et al. MUCOSAL IMMUNOLOGY. Individual intestinal symbionts induce a distinct population of RORγ+ regulatory T cells. Science 2015, 349, 993–997. [Google Scholar] [CrossRef]
- Eshleman, E.M.; Shao, T.Y.; Woo, V.; Rice, T.; Engleman, L.; Didriksen, B.J.; Whitt, J.; Haslam, D.B.; Way, S.S.; Alenghat, T. Intestinal epithelial HDAC3 and MHC class II coordinate microbiota-specific immunity. J. Clin. Investig. 2023, 133, 1–13. [Google Scholar] [CrossRef]
- Al Nabhani, Z.; Dulauroy, S.; Marques, R.; Cousu, C.; Al Bounny, S.; Dejardin, F.; Sparwasser, T.; Berard, M.; Cerf-Bensussan, N.; Eberl, G. A Weaning Reaction to Microbiota Is Required for Resistance to Immunopathologies in the Adult. Immunity 2019, 50, 1276–1288.e5. [Google Scholar] [CrossRef] [PubMed]
- Mishra, A.; Lai, G.C.; Yao, L.J.; Aung, T.T.; Shental, N.; Rotter-Maskowitz, A.; Shepherdson, E.; Singh, G.S.N.; Pai, R.; Shanti, A.; et al. Microbial exposure during early human development primes fetal immune cells. Cell 2021, 184, 3394–3409.e20. [Google Scholar] [CrossRef] [PubMed]
- Hu, M.; Eviston, D.; Hsu, P.; Marino, E.; Chidgey, A.; Santner-Nanan, B.; Wong, K.; Richards, J.L.; Yap, Y.A.; Collier, F.; et al. Decreased maternal serum acetate and impaired fetal thymic and regulatory T cell development in preeclampsia. Nat. Commun. 2019, 10, 3031. [Google Scholar] [CrossRef] [PubMed]
- Sanidad, K.Z.; Rager, S.L.; Carrow, H.C.; Ananthanarayanan, A.; Callaghan, R.; Hart, L.R.; Li, T.; Ravisankar, P.; Brown, J.A.; Amir, M.; et al. Gut bacteria-derived serotonin promotes immune tolerance in early life. Sci. Immunol. 2024, 9, eadj4775. [Google Scholar] [CrossRef] [PubMed]
- Khosravi, A.; Yanez, A.; Price, J.G.; Chow, A.; Merad, M.; Goodridge, H.S.; Mazmanian, S.K. Gut microbiota promote hematopoiesis to control bacterial infection. Cell Host Microbe 2014, 15, 374–381. [Google Scholar] [CrossRef] [PubMed]
- Josefsdottir, K.S.; Baldridge, M.T.; Kadmon, C.S.; King, K.Y. Antibiotics impair murine hematopoiesis by depleting the intestinal microbiota. Blood 2017, 129, 729–739. [Google Scholar] [CrossRef] [PubMed]
- Staffas, A.; Burgos da Silva, M.; Slingerland, A.E.; Lazrak, A.; Bare, C.J.; Holman, C.D.; Docampo, M.D.; Shono, Y.; Durham, B.; Pickard, A.J.; et al. Nutritional Support from the Intestinal Microbiota Improves Hematopoietic Reconstitution after Bone Marrow Transplantation in Mice. Cell Host Microbe 2018, 23, 447–457.e4. [Google Scholar] [CrossRef]
- Trompette, A.; Gollwitzer, E.S.; Yadava, K.; Sichelstiel, A.K.; Sprenger, N.; Ngom-Bru, C.; Blanchard, C.; Junt, T.; Nicod, L.P.; Harris, N.L.; et al. Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nat. Med. 2014, 20, 159–166. [Google Scholar] [CrossRef]
- Clarke, T.B.; Davis, K.M.; Lysenko, E.S.; Zhou, A.Y.; Yu, Y.; Weiser, J.N. Recognition of peptidoglycan from the microbiota by Nod1 enhances systemic innate immunity. Nat. Med. 2010, 16, 228–231. [Google Scholar] [CrossRef]
- Yanez, A.; Coetzee, S.G.; Olsson, A.; Muench, D.E.; Berman, B.P.; Hazelett, D.J.; Salomonis, N.; Grimes, H.L.; Goodridge, H.S. Granulocyte-Monocyte Progenitors and Monocyte-Dendritic Cell Progenitors Independently Produce Functionally Distinct Monocytes. Immunity 2017, 47, 890–902.e4. [Google Scholar] [CrossRef]
- Menezes, S.; Melandri, D.; Anselmi, G.; Perchet, T.; Loschko, J.; Dubrot, J.; Patel, R.; Gautier, E.L.; Hugues, S.; Longhi, M.P.; et al. The Heterogeneity of Ly6Chi Monocytes Controls Their Differentiation into iNOS+ Macrophages or Monocyte-Derived Dendritic Cells. Immunity 2016, 45, 1205–1218. [Google Scholar] [CrossRef] [PubMed]
- Villani, A.C.; Satija, R.; Reynolds, G.; Sarkizova, S.; Shekhar, K.; Fletcher, J.; Griesbeck, M.; Butler, A.; Zheng, S.; Lazo, S.; et al. Single-cell RNA-seq reveals new types of human blood dendritic cells, monocytes, and progenitors. Science 2017, 356, eaah4573. [Google Scholar] [CrossRef] [PubMed]
- Jonscher, K.R.; Abrams, J.; Friedman, J.E. Maternal Diet Alters Trained Immunity in the Pathogenesis of Pediatric NAFLD. J. Cell. Immunol. 2020, 2, 315–325. [Google Scholar] [CrossRef] [PubMed]
- Luo, Y.; Chen, G.L.; Hannemann, N.; Ipseiz, N.; Kronke, G.; Bauerle, T.; Munos, L.; Wirtz, S.; Schett, G.; Bozec, A. Microbiota from Obese Mice Regulate Hematopoietic Stem Cell Differentiation by Altering the Bone Niche. Cell Metab. 2015, 22, 886–894. [Google Scholar] [CrossRef]
- Bermick, J.; Schaller, M. Chorioamnionitis exposure dampens the preterm monocyte response to subsequent challenges. Immunol. Cell Biol. 2018, 96, 789–791. [Google Scholar] [CrossRef] [PubMed]
- de Jong, E.; Hancock, D.G.; Wells, C.; Richmond, P.; Simmer, K.; Burgner, D.; Strunk, T.; Currie, A.J. Exposure to chorioamnionitis alters the monocyte transcriptional response to the neonatal pathogen Staphylococcus epidermidis. Immunol. Cell Biol. 2018, 96, 792–804. [Google Scholar] [CrossRef]
- Rios-Covian, D.; Ruas-Madiedo, P.; Margolles, A.; Gueimonde, M.; de Los Reyes-Gavilan, C.G.; Salazar, N. Intestinal Short Chain Fatty Acids and their Link with Diet and Human Health. Front. Microbiol. 2016, 7, 185. [Google Scholar] [CrossRef] [PubMed]
- Koh, A.; De Vadder, F.; Kovatcheva-Datchary, P.; Backhed, F. From Dietary Fiber to Host Physiology: Short-Chain Fatty Acids as Key Bacterial Metabolites. Cell 2016, 165, 1332–1345. [Google Scholar] [CrossRef] [PubMed]
- Hsu, C.N.; Tain, Y.L. Developmental Programming and Reprogramming of Hypertension and Kidney Disease: Impact of Tryptophan Metabolism. Int. J. Mol. Sci. 2020, 21, 8705. [Google Scholar] [CrossRef]
- Patil, N.Y.; Friedman, J.E.; Joshi, A.D. Role of Hepatic Aryl Hydrocarbon Receptor in Non-Alcoholic Fatty Liver Disease. Receptors 2023, 2, 1–15. [Google Scholar] [CrossRef]
- Cummings, J.H.; Pomare, E.W.; Branch, W.J.; Naylor, C.P.; Macfarlane, G.T. Short chain fatty acids in human large intestine, portal, hepatic and venous blood. Gut 1987, 28, 1221–1227. [Google Scholar] [CrossRef] [PubMed]
- Johnstone, R.W. Histone-deacetylase inhibitors: Novel drugs for the treatment of cancer. Nat. Rev. Drug Discov. 2002, 1, 287–299. [Google Scholar] [CrossRef] [PubMed]
- Chang, P.V.; Hao, L.; Offermanns, S.; Medzhitov, R. The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition. Proc. Natl. Acad. Sci. USA 2014, 111, 2247–2252. [Google Scholar] [CrossRef]
- Flint, H.J.; Scott, K.P.; Louis, P.; Duncan, S.H. The role of the gut microbiota in nutrition and health. Nat. Rev. Gastroenterol. Hepatol. 2012, 9, 577–589. [Google Scholar] [CrossRef] [PubMed]
- Singh, N.; Thangaraju, M.; Prasad, P.D.; Martin, P.M.; Lambert, N.A.; Boettger, T.; Offermanns, S.; Ganapathy, V. Blockade of dendritic cell development by bacterial fermentation products butyrate and propionate through a transporter (Slc5a8)-dependent inhibition of histone deacetylases. J. Biol. Chem. 2010, 285, 27601–27608. [Google Scholar] [CrossRef]
- Beisner, J.; Filipe Rosa, L.; Kaden-Volynets, V.; Stolzer, I.; Gunther, C.; Bischoff, S.C. Prebiotic Inulin and Sodium Butyrate Attenuate Obesity-Induced Intestinal Barrier Dysfunction by Induction of Antimicrobial Peptides. Front. Immunol. 2021, 12, 678360. [Google Scholar] [CrossRef]
- Fang, W.; Xue, H.; Chen, X.; Chen, K.; Ling, W. Supplementation with Sodium Butyrate Modulates the Composition of the Gut Microbiota and Ameliorates High-Fat Diet-Induced Obesity in Mice. J. Nutr. 2019, 149, 747–754. [Google Scholar] [CrossRef] [PubMed]
- Zhou, D.; Pan, Q.; Xin, F.Z.; Zhang, R.N.; He, C.X.; Chen, G.Y.; Liu, C.; Chen, Y.W.; Fan, J.G. Sodium butyrate attenuates high-fat diet-induced steatohepatitis in mice by improving gut microbiota and gastrointestinal barrier. World J. Gastroenterol. 2017, 23, 60–75. [Google Scholar] [CrossRef] [PubMed]
- Kimura, I.; Ozawa, K.; Inoue, D.; Imamura, T.; Kimura, K.; Maeda, T.; Terasawa, K.; Kashihara, D.; Hirano, K.; Tani, T.; et al. The gut microbiota suppresses insulin-mediated fat accumulation via the short-chain fatty acid receptor GPR43. Nat. Commun. 2013, 4, 1829. [Google Scholar] [CrossRef]
- Le Poul, E.; Loison, C.; Struyf, S.; Springael, J.Y.; Lannoy, V.; Decobecq, M.E.; Brezillon, S.; Dupriez, V.; Vassart, G.; Van Damme, J.; et al. Functional characterization of human receptors for short chain fatty acids and their role in polymorphonuclear cell activation. J. Biol. Chem. 2003, 278, 25481–25489. [Google Scholar] [CrossRef]
- Chen, D.; Wang, Y.Y.; Li, S.P.; Zhao, H.M.; Jiang, F.J.; Wu, Y.X.; Tong, Y.; Pang, Q.F. Maternal propionate supplementation ameliorates glucose and lipid metabolic disturbance in hypoxia-induced fetal growth restriction. Food Funct. 2022, 13, 10724–10736. [Google Scholar] [CrossRef] [PubMed]
- Badawy, A.A. Tryptophan metabolism, disposition and utilization in pregnancy. Biosci. Rep. 2015, 35, e00261. [Google Scholar] [CrossRef] [PubMed]
- Cussotto, S.; Delgado, I.; Anesi, A.; Dexpert, S.; Aubert, A.; Beau, C.; Forestier, D.; Ledaguenel, P.; Magne, E.; Mattivi, F.; et al. Tryptophan Metabolic Pathways Are Altered in Obesity and Are Associated With Systemic Inflammation. Front. Immunol. 2020, 11, 557. [Google Scholar] [CrossRef] [PubMed]
- Lischka, J.; Schanzer, A.; Baumgartner, M.; de Gier, C.; Greber-Platzer, S.; Zeyda, M. Tryptophan Metabolism Is Associated with BMI and Adipose Tissue Mass and Linked to Metabolic Disease in Pediatric Obesity. Nutrients 2022, 14, 286. [Google Scholar] [CrossRef] [PubMed]
- Krishnan, S.; Ding, Y.; Saedi, N.; Choi, M.; Sridharan, G.V.; Sherr, D.H.; Yarmush, M.L.; Alaniz, R.C.; Jayaraman, A.; Lee, K. Gut Microbiota-Derived Tryptophan Metabolites Modulate Inflammatory Response in Hepatocytes and Macrophages. Cell Rep. 2018, 23, 1099–1111. [Google Scholar] [CrossRef] [PubMed]
- Scott, S.A.; Fu, J.; Chang, P.V. Microbial tryptophan metabolites regulate gut barrier function via the aryl hydrocarbon receptor. Proc. Natl. Acad. Sci. USA 2020, 117, 19376–19387. [Google Scholar] [CrossRef] [PubMed]
- Choi, Y.; Kim, Y.; Park, S.; Lee, K.W.; Park, T. Indole-3-carbinol prevents diet-induced obesity through modulation of multiple genes related to adipogenesis, thermogenesis or inflammation in the visceral adipose tissue of mice. J. Nutr. Biochem. 2012, 23, 1732–1739. [Google Scholar] [CrossRef]
- Shinde, R.; McGaha, T.L. The Aryl Hydrocarbon Receptor: Connecting Immunity to the Microenvironment. Trends Immunol. 2018, 39, 1005–1020. [Google Scholar] [CrossRef] [PubMed]
- Bock, K.W. Aryl hydrocarbon receptor (AHR), integrating energy metabolism and microbial or obesity-mediated inflammation. Biochem. Pharmacol. 2021, 184, 114346. [Google Scholar] [CrossRef]
- Ghiboub, M.; Verburgt, C.M.; Sovran, B.; Benninga, M.A.; de Jonge, W.J.; Van Limbergen, J.E. Nutritional Therapy to Modulate Tryptophan Metabolism and Aryl Hydrocarbon-Receptor Signaling Activation in Human Diseases. Nutrients 2020, 12, 2846. [Google Scholar] [CrossRef]
- den Besten, G.; van Eunen, K.; Groen, A.K.; Venema, K.; Reijngoud, D.J.; Bakker, B.M. The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J. Lipid Res. 2013, 54, 2325–2340. [Google Scholar] [CrossRef] [PubMed]
- Frost, G.; Sleeth, M.L.; Sahuri-Arisoylu, M.; Lizarbe, B.; Cerdan, S.; Brody, L.; Anastasovska, J.; Ghourab, S.; Hankir, M.; Zhang, S.; et al. The short-chain fatty acid acetate reduces appetite via a central homeostatic mechanism. Nat. Commun. 2014, 5, 3611. [Google Scholar] [CrossRef]
- Laursen, M.F.; Sakanaka, M.; von Burg, N.; Morbe, U.; Andersen, D.; Moll, J.M.; Pekmez, C.T.; Rivollier, A.; Michaelsen, K.F.; Molgaard, C.; et al. Bifidobacterium species associated with breastfeeding produce aromatic lactic acids in the infant gut. Nat. Microbiol. 2021, 6, 1367–1382. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.P.; Yoo, J.S.; Lee, J.H.; Jang, H.D.; Kim, H.J.; Shin, S.O.; Seong, S.I.; Kim, I.H. Effects of phenyllactic acid on growth performance, nutrient digestibility, microbial shedding, and blood profile in pigs. J. Anim. Sci. 2009, 87, 3235–3243. [Google Scholar] [CrossRef]
- Zugasti, O.; Bose, N.; Squiban, B.; Belougne, J.; Kurz, C.L.; Schroeder, F.C.; Pujol, N.; Ewbank, J.J. Activation of a G protein-coupled receptor by its endogenous ligand triggers the innate immune response of Caenorhabditis elegans. Nat. Immunol. 2014, 15, 833–838. [Google Scholar] [CrossRef]
- Waldrop, S.W.; Niemiec, S.; Wood, C.; Gyllenhammer, L.E.; Jansson, T.; Friedman, J.E.; Tryggestad, J.B.; Borengasser, S.J.; Davidson, E.J.; Yang, I.V.; et al. Cord blood DNA methylation of immune and lipid metabolism genes is associated with maternal triglycerides and child adiposity. Obesity 2024, 32, 187–199. [Google Scholar] [CrossRef] [PubMed]
- van der Heijden, C.; Noz, M.P.; Joosten, L.A.B.; Netea, M.G.; Riksen, N.P.; Keating, S.T. Epigenetics and Trained Immunity. Antioxid. Redox Signal 2018, 29, 1023–1040. [Google Scholar] [CrossRef] [PubMed]
- Fanucchi, S.; Dominguez-Andres, J.; Joosten, L.A.B.; Netea, M.G.; Mhlanga, M.M. The Intersection of Epigenetics and Metabolism in Trained Immunity. Immunity 2021, 54, 32–43. [Google Scholar] [CrossRef] [PubMed]
- Suter, M.; Bocock, P.; Showalter, L.; Hu, M.; Shope, C.; McKnight, R.; Grove, K.; Lane, R.; Aagaard-Tillery, K. Epigenomics: Maternal high-fat diet exposure in utero disrupts peripheral circadian gene expression in nonhuman primates. FASEB J. 2011, 25, 714–726. [Google Scholar] [CrossRef]
- Suter, M.A.; Takahashi, D.; Grove, K.L.; Aagaard, K.M. Postweaning exposure to a high-fat diet is associated with alterations to the hepatic histone code in Japanese macaques. Pediatr. Res. 2013, 74, 252–258. [Google Scholar] [CrossRef]
- Harmancioglu, B.; Kabaran, S. Maternal high fat diets: Impacts on offspring obesity and epigenetic hypothalamic programming. Front. Genet. 2023, 14, 1158089. [Google Scholar] [CrossRef]
- Borengasser, S.J.; Zhong, Y.; Kang, P.; Lindsey, F.; Ronis, M.J.; Badger, T.M.; Gomez-Acevedo, H.; Shankar, K. Maternal obesity enhances white adipose tissue differentiation and alters genome-scale DNA methylation in male rat offspring. Endocrinology 2013, 154, 4113–4125. [Google Scholar] [CrossRef] [PubMed]
- Reynolds, C.M.; Segovia, S.A.; Vickers, M.H. Experimental Models of Maternal Obesity and Neuroendocrine Programming of Metabolic Disorders in Offspring. Front. Endocrinol. 2017, 8, 245. [Google Scholar] [CrossRef] [PubMed]
- Ross, M.G.; Desai, M. Developmental programming of appetite/satiety. Ann. Nutr. Metab. 2014, 64 (Suppl. S1), 36–44. [Google Scholar] [CrossRef] [PubMed]
- Chen, H.; Simar, D.; Morris, M.J. Hypothalamic neuroendocrine circuitry is programmed by maternal obesity: Interaction with postnatal nutritional environment. PLoS ONE 2009, 4, e6259. [Google Scholar] [CrossRef]
- Vickers, M.H.; Breier, B.H.; Cutfield, W.S.; Hofman, P.L.; Gluckman, P.D. Fetal origins of hyperphagia, obesity, and hypertension and postnatal amplification by hypercaloric nutrition. Am. J. Physiol. Endocrinol. Metab. 2000, 279, E83–E87. [Google Scholar] [CrossRef]
- Desai, M.; Han, G.; Ross, M.G. Programmed hyperphagia in offspring of obese dams: Altered expression of hypothalamic nutrient sensors, neurogenic factors and epigenetic modulators. Appetite 2016, 99, 193–199. [Google Scholar] [CrossRef]
- Sardinha, F.L.; Telles, M.M.; Albuquerque, K.T.; Oyama, L.M.; Guimaraes, P.A.; Santos, O.F.; Ribeiro, E.B. Gender difference in the effect of intrauterine malnutrition on the central anorexigenic action of insulin in adult rats. Nutrition 2006, 22, 1152–1161. [Google Scholar] [CrossRef]
- Galjaard, S.; Devlieger, R.; Van Assche, F.A. Fetal growth and developmental programming. J. Perinat. Med. 2013, 41, 101–105. [Google Scholar] [CrossRef]
- Gali Ramamoorthy, T.; Allen, T.J.; Davies, A.; Harno, E.; Sefton, C.; Murgatroyd, C.; White, A. Maternal overnutrition programs epigenetic changes in the regulatory regions of hypothalamic Pomc in the offspring of rats. Int. J. Obes. 2018, 42, 1431–1444. [Google Scholar] [CrossRef]
- Plagemann, A.; Harder, T.; Brunn, M.; Harder, A.; Roepke, K.; Wittrock-Staar, M.; Ziska, T.; Schellong, K.; Rodekamp, E.; Melchior, K.; et al. Hypothalamic proopiomelanocortin promoter methylation becomes altered by early overfeeding: An epigenetic model of obesity and the metabolic syndrome. J. Physiol. 2009, 587, 4963–4976. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Funato, H.; Oda, S.; Yokofujita, J.; Igarashi, H.; Kuroda, M. Fasting and high-fat diet alter histone deacetylase expression in the medial hypothalamus. PLoS ONE 2011, 6, e18950. [Google Scholar] [CrossRef] [PubMed]
- Dalvi, P.S.; Chalmers, J.A.; Luo, V.; Han, D.Y.; Wellhauser, L.; Liu, Y.; Tran, D.Q.; Castel, J.; Luquet, S.; Wheeler, M.B.; et al. High fat induces acute and chronic inflammation in the hypothalamus: Effect of high-fat diet, palmitate and TNF-α on appetite-regulating NPY neurons. Int. J. Obes. 2017, 41, 149–158. [Google Scholar] [CrossRef] [PubMed]
- Grayson, B.E.; Levasseur, P.R.; Williams, S.M.; Smith, M.S.; Marks, D.L.; Grove, K.L. Changes in melanocortin expression and inflammatory pathways in fetal offspring of nonhuman primates fed a high-fat diet. Endocrinology 2010, 151, 1622–1632. [Google Scholar] [CrossRef] [PubMed]
- Cheng, C.; Chen, W.; Jin, H.; Chen, X. A Review of Single-Cell RNA-Seq Annotation, Integration, and Cell-Cell Communication. Cells 2023, 12, 1970. [Google Scholar] [CrossRef] [PubMed]
- Chen, B.; de Launoit, E.; Renier, N.; Schneeberger, M. Maternal nutritional programming shapes the cerebral landscape. Trends Endocrinol. Metab. 2023, 35, 367–370. [Google Scholar] [CrossRef] [PubMed]
- Wu, R.; Prachyathipsakul, T.; Zhuang, J.; Liu, H.; Han, Y.; Liu, B.; Gong, S.; Qiu, J.; Wong, S.; Ribbe, A.; et al. Conferring liver selectivity to a thyromimetic using a novel nanoparticle increases therapeutic efficacy in a diet-induced obesity animal model. PNAS Nexus 2023, 2, pgad252. [Google Scholar] [CrossRef]
- Ayyangar, U.; Karkhanis, A.; Tay, H.; Afandi, A.F.B.; Bhattacharjee, O.; Ks, L.; Lee, S.H.; Chan, J.; Raghavan, S. Metabolic rewiring of macrophages by epidermal-derived lactate promotes sterile inflammation in the murine skin. EMBO J. 2024, 43, 1113–1134. [Google Scholar] [CrossRef]
Subjects (Year) | Physiological Outcomes | Programming Effects | Reference |
---|---|---|---|
222 human infants and NOD mice (2016) |
|
| [137] |
Germ-free mice colonized with microbes of infants from obese or NW mothers (2018) |
|
| [130] |
17,055 neonates from the general population (2019) |
|
| [136] |
Antibiotic-treated and germ-free mice (2019) |
|
| [139] |
46 full-term neonates from obese mother with and without GDM (2020) |
|
| [140] |
418 mothers and their neonates (2021) |
|
| [141] |
394 pregnant women in the first trimester and germ-free mice (2023) |
|
| [142] |
65 mother-infant pairs, half receiving a prebiotic (2024) |
|
| [143] |
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
Nelson, B.N.; Friedman, J.E. Developmental Programming of the Fetal Immune System by Maternal Western-Style Diet: Mechanisms and Implications for Disease Pathways in the Offspring. Int. J. Mol. Sci. 2024, 25, 5951. https://doi.org/10.3390/ijms25115951
Nelson BN, Friedman JE. Developmental Programming of the Fetal Immune System by Maternal Western-Style Diet: Mechanisms and Implications for Disease Pathways in the Offspring. International Journal of Molecular Sciences. 2024; 25(11):5951. https://doi.org/10.3390/ijms25115951
Chicago/Turabian StyleNelson, Benjamin N., and Jacob E. Friedman. 2024. "Developmental Programming of the Fetal Immune System by Maternal Western-Style Diet: Mechanisms and Implications for Disease Pathways in the Offspring" International Journal of Molecular Sciences 25, no. 11: 5951. https://doi.org/10.3390/ijms25115951
APA StyleNelson, B. N., & Friedman, J. E. (2024). Developmental Programming of the Fetal Immune System by Maternal Western-Style Diet: Mechanisms and Implications for Disease Pathways in the Offspring. International Journal of Molecular Sciences, 25(11), 5951. https://doi.org/10.3390/ijms25115951