Antioxidants, Gut Microbiota, and Cardiovascular Programming: Unraveling a Triad of Early-Life Interactions
Abstract
1. Introduction
2. Materials and Methods
3. Distinct Roles of Gut Microbiota in Overt CVD and Cardiovascular Programming
3.1. The Role of Gut Microbiota in CVD
3.1.1. Gut Microbiota and Hypertension
3.1.2. TMAO and CVD
3.1.3. Impact of Short-Chain Fatty Acids on CVD
3.1.4. Inflammation and Immune Dysregulation
3.1.5. Bile Acid Metabolism
3.2. Impact of Early-Life Gut Microbiota on Cardiovascular Programming
3.2.1. Early-Life Gut Microbiota
3.2.2. Human and Animal Evidence of Cardiovascular Programming
3.2.3. Mechanisms Underlying Cardiovascular Programming
3.2.4. Oxidative Stress and Its Link to Gut Microbiota in Cardiovascular Programming
4. Antioxidants as a Reprogramming Strategy
4.1. Vitamins and Micronutrients
4.2. Amino Acids
4.3. Melatonin
4.4. Polyphenols
4.5. SCFAs
4.6. N-Acetylcysteine
4.7. Synthetic Antioxidants
4.8. Others
5. Conclusions and Future Perspective
Author Contributions
Funding
Data Availability Statement
Acknowledgments
Conflicts of Interest
Abbreviations
ROS | Reactive Oxygen Species |
BP | Blood Pressure |
CVD | Cardiovascular Disease |
CKD | Chronic Kidney Disease |
NO | Nitric Oxide |
H2S | Hydrogen Sulfide |
BCAAs | Branched-Chain Amino Acids |
AhR | Aryl Hydrocarbon Receptor |
DOHaD | Developmental Origins of Health and Disease |
TMA | Trimethylamine |
TMAO | Trimethylamine N-oxide |
SCFAs | Short-Chain Fatty Acids |
NAC | N-acetylcysteine |
SHR | Spontaneously Hypertensive Rat |
DMF | Dimethyl Fumarate |
ADMA | Asymmetric Dimethylarginine |
EGCG | Epigallocatechin Gallate |
RAAS | Renin–Angiotensin–Aldosterone System |
LPS | Lipopolysaccharide |
NRF2 | Nuclear factor erythroid 2-related factor 2 |
FXR | Farnesoid X Receptor |
TGR5 | G protein–coupled Bile Acid Receptor 1 |
GPR41 | G-protein-coupled Receptor 41 |
GPR43 | G-protein-coupled Receptor 43 |
GPR109A | G-protein-coupled Receptor 109A |
References
- Arnett, D.K.; Blumenthal, R.S.; Albert, M.A.; Buroker, A.B.; Goldberger, Z.D.; Hahn, E.J.; Himmelfarb, C.D.; Khera, A.; Lloyd-Jones, D.; McEvoy, J.W.; et al. 2019 ACC/AHA Guideline on the Primary Prevention of Cardiovascular Disease: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation 2019, 140, e596–e646. [Google Scholar] [CrossRef]
- Thomas, H.; Diamond, J.; Vieco, A.; Chaudhuri, S.; Shinnar, E.; Cromer, S.; Perel, P.; Mensah, G.A.; Narula, J.; Johnson, C.O.; et al. Global Atlas of Cardiovascular Disease 2000-2016: The Path to Prevention and Control. Glob. Heart 2018, 13, 143–163. [Google Scholar] [CrossRef]
- Huang, M.; Li, J.; Zhao, X.; Fu, R.; Li, X.; Jiang, W. Global and regional prevalence and cardiovascular risk of primary aldosteronism: A systematic review and meta-analysis. Curr. Probl. Cardiol. 2024, 49, 102791. [Google Scholar] [CrossRef]
- Barker, D.J.; Eriksson, J.G.; Forsén, T.; Osmond, C. Fetal origins of adult disease: Strength of effects and biological basis. Int. J. Epidemiol. 2002, 31, 1239. [Google Scholar] [CrossRef]
- Gluckman, P.D.; Hanson, M.A.; Buklijas, T. A Conceptual Framework for the Developmental Origins of Health and Disease. J. Dev. Orig. Health Dis. 2010, 1, 6–18. [Google Scholar] [CrossRef]
- Santos, K.L. The programming of cardiovascular disease. J. Dev. Orig. Health Dis. 2015, 6, 366–376. [Google Scholar]
- Loche, E.; Ozanne, S.E. Early nutrition, epigenetics, and cardiovascular disease. Curr. Opin. Lipidol. 2016, 27, 449–458. [Google Scholar] [CrossRef]
- LaKind, J.S.; Lehmann, G.M.; Davis, M.H.; Hines, E.P.; Marchitti, S.A.; Alcala, C.; Lorber, M. Infant Dietary Exposures to Environmental Chemicals and Infant/Child Health: A Critical Assessment of the Literature. Environ. Health Perspect. 2018, 126, 96002. [Google Scholar] [CrossRef]
- Cottrell, E.C.; Seckl, J.R. Prenatal stress, glucocorticoids and the programming of adult disease. Front. Behav. Neurosci. 2009, 3, 19. [Google Scholar] [CrossRef]
- Hsu, C.N.; Hou, C.Y.; Hsu, W.H.; Tain, Y.L. Cardiovascular Diseases of Developmental Origins: Preventive Aspects of Gut Microbiota-Targeted Therapy. Nutrients 2021, 13, 2290. [Google Scholar] [CrossRef]
- Bourque, S.L.; Davidge, S.T. Developmental programming of cardiovascular function: A translational perspective. Clin. Sci. 2020, 134, 3023–3046. [Google Scholar] [CrossRef]
- Rodríguez-Rodríguez, P.; Ramiro-Cortijo, D.; Reyes-Hernández, C.G.; López de Pablo, A.L.; González, M.C.; Arribas, S.M. Implication of Oxidative Stress in Fetal Programming of Cardiovascular Disease. Front. Physiol. 2018, 9, 602. [Google Scholar] [CrossRef] [PubMed]
- Zhao, C.; He, L.; Li, L.; Deng, F.; Zhang, M.; Wang, C.; Qiu, J.; Gao, Q. Prenatal glucocorticoids exposure and adverse cardiovascular effects in offspring. Front. Endocrinol. 2024, 15, 1430334. [Google Scholar] [CrossRef]
- Rogers, L.K.; Velten, M. Maternal inflammation, growth retardation, and preterm birth: Insights into adult cardiovascular disease. Life Sci. 2011, 89, 417–421. [Google Scholar] [CrossRef]
- Benincasa, G.; Napoli, C.; DeMeo, D.L. Transgenerational Epigenetic Inheritance of Cardiovascular Diseases: A Network Medicine Perspective. Matern. Child Health J. 2024, 28, 617–630. [Google Scholar] [CrossRef]
- Tang, W.H.W.; Bäckhed, F.; Landmesser, U.; Hazen, S.L. Intestinal Microbiota in Cardiovascular Health and Disease: JACC State-of-the-Art Review. J. Am. Coll. Cardiol. 2019, 73, 2089–2105. [Google Scholar] [CrossRef]
- Witkowski, M.; Weeks, T.L.; Hazen, S.L. Gut Microbiota and Cardiovascular Disease. Circ. Res. 2020, 127, 553–570. [Google Scholar] [CrossRef]
- Scarmozzino, F.; Poli, A.; Visiolim, F. Microbiota and cardiovascular disease risk: A scoping review. Pharmacol. Res. 2020, 159, 104952. [Google Scholar] [CrossRef]
- Hu, T.; Wu, Q.; Yao, Q.; Jiang, K.; Yu, J.; Tang, Q. Short-chain fatty acid metabolism and multiple effects on cardiovascular diseases. Ageing Res. Rev. 2022, 81, 101706. [Google Scholar] [CrossRef]
- Thomas, M.S.; Fernandez, M.L. Trimethylamine N-Oxide (TMAO), Diet and Cardiovascular Disease. Curr. Atheroscler. Rep. 2021, 23, 12. [Google Scholar] [CrossRef]
- Matamoros, S.; Gras-Leguen, C.; Le Vacon, F.; Potel, G.; De La Cochetiere, M.F. Development of intestinal microbiota in infants and its impact on health. Trends Microbiol. 2013, 21, 167–173. [Google Scholar] [CrossRef]
- Sarkar, A.; Yoo, J.Y.; Valeria Ozorio Dutra, S.; Morgan, K.H.; Groer, M. The Association between Early-Life Gut Microbiota and Long-Term Health and Diseases. J. Clin. Med. 2021, 10, 459. [Google Scholar] [CrossRef]
- Młynarska, E.; Hajdys, J.; Czarnik, W.; Fularski, P.; Leszto, K.; Majchrowicz, G.; Lisińska, W.; Rysz, J.; Franczyk, B. The Role of Antioxidants in the Therapy of Cardiovascular Diseases-A Literature Review. Nutrients 2024, 16, 2587. [Google Scholar] [CrossRef]
- Manson, J.E.; Gaziano, J.M.; Jonas, M.A.; Hennekens, C.H. Antioxidants and cardiovascular disease: A review. J. Am. Coll. Nutr. 1993, 12, 426–432. [Google Scholar] [CrossRef]
- Tain, Y.L.; Joles, J.A. Reprogramming: A Preventive Strategy in Hypertension Focusing on the Kidney. Int. J. Mol. Sci. 2015, 17, 23. [Google Scholar] [CrossRef] [PubMed]
- Paauw, N.D.; van Rijn, B.B.; Lely, A.T.; Joles, J.A. Pregnancy as a critical window for blood pressure regulation in mother and child: Programming and reprogramming. Acta Physiol. 2017, 219, 241–259. [Google Scholar] [CrossRef]
- Aljunaidy, M.M.; Morton, J.S.; Cooke, C.M.; Davidge, S.T. Prenatal hypoxia and placental oxidative stress: Linkages to developmental origins of cardiovascular disease. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2017, 313, R395–R399. [Google Scholar] [CrossRef]
- Diniz, M.S.; Magalhães, C.C.; Tocantins, C.; Grilo, L.F.; Teixeira, J.; Pereira, S.P. Nurturing through Nutrition: Exploring the Role of Antioxidants in Maternal Diet during Pregnancy to Mitigate Developmental Programming of Chronic Diseases. Nutrients 2023, 15, 4623. [Google Scholar] [CrossRef]
- Rock, C.R.; Miller, S.L.; Allison, B.J. The Use of Antioxidants for Cardiovascular Protection in Fetal Growth Restriction: A Systematic Review. Antioxidants 2024, 13, 1400. [Google Scholar] [CrossRef]
- Li, X.Y.; Meng, L.; Shen, L.; Ji, H.F. Regulation of gut microbiota by vitamin C, vitamin E and β-carotene. Food Res. Int. 2023, 169, 112749. [Google Scholar] [CrossRef]
- Wan, M.L.Y.; Co, V.A.; El-Nezami, H. Dietary polyphenol impact on gut health and microbiota. Crit. Rev. Food Sci. Nutr. 2021, 61, 690–711. [Google Scholar] [CrossRef]
- Żółkiewicz, J.; Marzec, A.; Ruszczyński, M.; Feleszko, W. Postbiotics-A Step Beyond Pre- and Probiotics. Nutrients 2020, 12, 2189. [Google Scholar] [CrossRef]
- Wang, Y.; Wu, Y.; Wang, Y.; Xu, H.; Mei, X.; Yu, D.; Wang, Y.; Li, W. Antioxidant Properties of Probiotic Bacteria. Nutrients 2017, 9, 521. [Google Scholar] [CrossRef]
- Verhaar, B.J.H.; Prodan, A.; Nieuwdorp, M.; Muller, M. Gut Microbiota in Hypertension and Atherosclerosis: A Review. Nutrients 2020, 12, 2982. [Google Scholar] [CrossRef]
- Xu, W.; Chen, D.; Zhou, H.L. S-Nitrosylation: Mechanistic Links between Nitric Oxide Signaling and Atherosclerosis. Curr. Atheroscler. Rep. 2025, 27, 78. [Google Scholar] [CrossRef]
- Zheng, S.; Liu, Z.; Liu, H.; Lim, J.Y.; Li, D.W.H.; Zhang, S.; Luo, F.; Wang, X.; Sun, C.; Tang, R.; et al. Research development on gut microbiota and vulnerable atherosclerotic plaque. Heliyon 2024, 10, e25186. [Google Scholar] [CrossRef]
- Ni, M.; Wang, Y.; Zhang, M.; Zhang, P.F.; Ding, S.F.; Liu, C.X.; Liu, X.L.; Zhao, Y.X.; Zhang, Y. Atherosclerotic plaque disruption induced by stress and lipopolysaccharide in apolipoprotein E knockout mice. Am. J. Physiol. Heart Circ. Physiol. 2009, 296, H1598–H1606. [Google Scholar] [CrossRef] [PubMed]
- Khodor, S.A.; Reichert, B.; Shatat, I.F. The microbiome and blood pressure: Can microbes regulate our blood pressure? Front. Pediatr. 2017, 5, 138. [Google Scholar] [CrossRef]
- Yang, T.; Richards, E.M.; Pepine, C.J.; Raizada, M.K. The gut microbiota and the brain-gut-kidney axis in hypertension and chronic kidney disease. Nat. Rev. Nephrol. 2018, 14, 442–456. [Google Scholar] [CrossRef]
- Cai, M.; Lin, L.; Jiang, F.; Peng, Y.; Li, S.; Chen, L.; Lin, Y. Gut microbiota changes in patients with hypertension: A systematic review and meta-analysis. J. Clin. Hypertens. 2023, 25, 1053–1068. [Google Scholar] [CrossRef]
- Santisteban, M.M.; Qi, Y.; Zubcevic, J.; Kim, S.; Yang, T.; Shenoy, V.; Cole-Jeffrey, C.T.; Lobaton, G.O.; Stewart, D.C.; Rubiano, A.; et al. Hypertension-Linked Pathophysiological Alterations in the Gut. Circ. Res. 2017, 120, 312–323. [Google Scholar] [CrossRef]
- Li, J.; Zhao, F.; Wang, Y.; Chen, J.; Tao, J.; Tian, G.; Wu, S.; Liu, W.; Cui, Q.; Geng, B.; et al. Gut microbiota dysbiosis contributes to the development of hypertension. Microbiome 2017, 5, 14. [Google Scholar] [CrossRef]
- Merra, G.; Noce, A.; Marrone, G.; Cintoni, M.; Tarsitano, M.G.; Capacci, A.; De Lorenzo, A. Influence of Mediterranean Diet on Human Gut Microbiota. Nutrients 2020, 13, 7. [Google Scholar] [CrossRef]
- Thushara, R.M.; Gangadaran, S.; Solati, Z.; Moghadasian, M.H. Cardiovascular benefits of probiotics: A review of experimental and clinical studies. Food Funct. 2016, 7, 632–642. [Google Scholar] [CrossRef] [PubMed]
- Pandey, K.R.; Naik, S.R.; Vakil, B.V. Probiotics, prebiotics and synbiotics-A review. J. Food Sci. Technol. 2015, 52, 7577–7587. [Google Scholar] [CrossRef]
- Leshem, A.; Horesh, N.; Elinav, E. Fecal microbial transplantation and its potential application in cardiometabolic syndrome. Front. Immunol. 2019, 10, 1341. [Google Scholar] [CrossRef]
- Velasquez, M.T.; Ramezani, A.; Manal, A.; Raj, D.S. Trimethylamine N-Oxide: The good, the bad and the unknown. Toxins 2016, 8, 326. [Google Scholar] [CrossRef]
- Schiattarella, G.G.; Sannino, A.; Toscano, E.; Giugliano, G.; Gargiulo, G.; Franzone, A.; Trimarco, B.; Esposito, G.; Perrino, C. Gut microbe-generated metabolite trimethylamine-N-oxide as cardiovascular risk biomarker: A systematic review and dose-response meta-analysis. Eur. Heart J. 2017, 38, 2948–2956. [Google Scholar] [CrossRef]
- Seldin, M.M.; Meng, Y.; Qi, H.; Zhu, W.; Wang, Z.; Hazen, S.L.; Lusis, A.J.; Shih, D.M. Trimethylamine N-Oxide promotes vascular inflammation through signaling of mitogen-activated protein kinase and nuclear factor-κB. J. Am. Heart Assoc. 2016, 5, e002767. [Google Scholar] [CrossRef]
- Yang, Y.; Karampoor, S.; Mirzaei, R.; Borozdkin, L.; Zhu, P. The interplay between microbial metabolites and macrophages in cardiovascular diseases: A comprehensive review. Int. Immunopharmacol. 2023, 121, 110546. [Google Scholar] [CrossRef]
- Querio, G.; Antoniotti, S.; Geddo, F.; Levi, R.; Gallo, M.P. Modulation of Endothelial Function by TMAO, a Gut Microbiota-Derived Metabolite. Int. J. Mol. Sci. 2023, 24, 5806. [Google Scholar] [CrossRef] [PubMed]
- Aldana-Hernández, P.; Leonard, K.A.; Zhao, Y.Y.; Curtis, J.M.; Field, C.J.; Jacobs, R.L. Dietary Choline or Trimethylamine N-oxide Supplementation Does Not Influence Atherosclerosis Development in Ldlr-/- and Apoe-/- Male Mice. J. Nutr. 2020, 150, 249–255. [Google Scholar] [CrossRef]
- Chen, H.; Li, J.; Li, N.; Liu, H.; Tang, J. Increased circulating trimethylamine N-oxide plays a contributory role in the development of endothelial dysfunction and hypertension in the RUPP rat model of preeclampsia. Hypertens. Pregnancy 2019, 38, 96–104. [Google Scholar] [CrossRef]
- Hamad, A.; Ozkan, M.H.; Uma, S. Trimethylamine-N-oxide (TMAO) Selectively Disrupts Endothelium-Dependent Hyperpolarization-Type Relaxations in a Time-Dependent Manner in Rat Superior Mesenteric Artery. Biol. Pharm. Bull. 2021, 44, 1220–1229. [Google Scholar] [CrossRef]
- Li, T.; Gua, C.; Wu, B.; Chen, Y. Increased circulating trimethylamine N-oxide contributes to endothelial dysfunction in a rat model of chronic kidney disease. Biochem. Biophys. Res. Commun. 2018, 495, 2071–2077. [Google Scholar] [CrossRef]
- Koh, A.; De Vadder, F.; Kovatcheva-Datchary, P.; Bäckhed, F. From Dietary Fiber to Host Physiology: Short-Chain Fatty Acids as Key Bacterial Metabolites. Cell 2016, 165, 1332–1345. [Google Scholar] [CrossRef]
- Fusco, W.; Lorenzo, M.B.; Cintoni, M.; Porcari, S.; Rinninella, E.; Kaitsas, F.; Lener, E.; Mele, M.C.; Gasbarrini, A.; Collado, M.C.; et al. Short-Chain Fatty-Acid-Producing Bacteria: Key Components of the Human Gut Microbiota. Nutrients 2023, 15, 2211. [Google Scholar] [CrossRef]
- Pluznick, J.L. Microbial short-chain fatty acids and blood pressure regulation. Curr. Hypertens. Rep. 2017, 19, 25. [Google Scholar] [CrossRef]
- Ratajczak, W.; Rył, A.; Mizerski, A.; Walczakiewicz, K.; Sipak, O.; Laszczyńska, M. Immunomodulatory potential of gut microbiome-derived short-chain fatty acids (SCFAs). Acta Biochim. Pol. 2019, 66, 1–12. [Google Scholar] [CrossRef]
- Hays, K.E.; Pfaffinger, J.M.; Ryznar, R. The interplay between gut microbiota, short-chain fatty acids, and implications for host health and disease. Gut Microbes 2024, 16, 2393270. [Google Scholar] [CrossRef]
- Madella, A.M.; Van Bergenhenegouwen, J.; Garssen, J.; Masereeuw, R.; Overbeek, S.A. Microbial-Derived Tryptophan Catabolites, Kidney Disease and Gut Inflammation. Toxins 2022, 14, 645. [Google Scholar] [CrossRef]
- Velasquez, M.T.; Centron, P.; Barrows, I.; Dwivedi, R.; Raj, D.S. Gut Microbiota and cardiovascular uremic toxicities. Toxins 2018, 10, 287. [Google Scholar] [CrossRef]
- Sallée, M.; Dou, L.; Cerini, C.; Poitevin, S.; Brunet, P.; Burtey, S. The aryl hydrocarbon receptor-activating effect of uremic toxins from tryptophan metabolism: A new concept to understand cardiovascular complications of chronic kidney disease. Toxins 2014, 6, 934–949. [Google Scholar] [CrossRef]
- Hung, S.C.; Kuo, K.L.; Wu, C.C.; Tarng, D.C. Indoxyl sulfate: A novel cardiovascular risk factor in chronic kidney disease. J. Am. Heart Assoc. 2017, 6, e005022. [Google Scholar] [CrossRef] [PubMed]
- Hubbard, T.D.; Murray, I.A.; Perdew, G.H. Indole and tryptophan metabolism: Endogenous and dietary routes to ah receptor activation. Drug Metab. Dispos. 2015, 43, 1522–1535. [Google Scholar] [CrossRef]
- Wilck, N.; Matus, M.G.; Kearney, S.M.; Olesen, S.W.; Forslund, K.; Bartolomaeus, H.; Haase, S.; Mähler, A.; Balogh, A.; Markó, L.; et al. Salt-responsive gut commensal modulates TH17 axis and disease. Nature 2017, 551, 585–589. [Google Scholar] [CrossRef]
- Ren, J.; Crowley, S.D. Role of T-cell activation in salt-sensitive hypertension. Am. J. Physiol. Heart Circ. Physiol. 2019, 316, H1345–H1353. [Google Scholar] [CrossRef]
- Gryp, T.; Vanholder, R.; Vaneechoutte, M.; Glorieux, G. p-Cresyl Sulfate. Toxins 2017, 9, 52. [Google Scholar] [CrossRef]
- Zhang, Z.; Lv, T.; Wang, X.; Wu, M.; Zhang, R.; Yang, X.; Fu, Y.; Liu, Z. Role of the microbiota-gut-heart axis between bile acids and cardiovascular disease. Biomed. Pharmacother. 2024, 174, 116567. [Google Scholar] [CrossRef]
- Hanafi, N.I.; Mohamed, A.S.; Sheikh Abdul Kadir, S.H.; Othman, M.H.D. Overview of bile acids signaling and perspective on the signal of ursodeoxycholic acid, the most hydrophilic bile acid, in the heart. Biomolecules 2018, 8, 159. [Google Scholar] [CrossRef]
- Fiorucci, S.; Distrutti, E. Linking liver metabolic and vascular disease via bile acid signaling. Trends Mol. Med. 2022, 28, 51–66. [Google Scholar] [CrossRef]
- Miyazaki-Anzai, S.; Masuda, M.; Levi, N.; Keenan, A.L.; Miyazaki, M. Dual activation of the bile acid nuclear receptor FXR and G-protein-coupled receptor TGR5 protects mice against atherosclerosis. PLoS ONE 2014, 9, e108270. [Google Scholar] [CrossRef]
- Crispi, F.; Sepúlveda-Martínez, Á.; Crovetto, F.; Gómez, O.; Bijnens, B.; Gratacós, E. Main Patterns of Fetal Cardiac Remodeling. Fetal Diagn. Ther. 2020, 47, 337–344. [Google Scholar] [CrossRef]
- Arrieta, M.C.; Stiemsma, L.T.; Amenyogbe, N.; Brown, E.M.; Finlay, B. The intestinal microbiome in early life: Health and disease. Front. Immunol. 2014, 5, 427. [Google Scholar] [CrossRef]
- Nagareddy, P.; Smyth, S.S. Inflammation and thrombosis in cardiovascular disease. Curr. Opin. Hematol. 2013, 20, 457–463. [Google Scholar] [CrossRef] [PubMed]
- Alexander, B.T.; South, A.M.; August, P.; Bertagnolli, M.; Ferranti, E.P.; Grobe, J.L.; Jones, E.J.; Loria, A.S.; Safdar, B.; Sequeira-Lopez, M.L.S.; et al. Appraising the Preclinical Evidence of the Role of the Renin-Angiotensin-Aldosterone System in Antenatal Programming of Maternal and Offspring Cardiovascular Health Across the Life Course: Moving the Field Forward: A Scientific Statement From the American Heart Association. Hypertension 2023, 80, e75–e89. [Google Scholar] [PubMed]
- Milani, C.; Duranti, S.; Bottacini, F.; Casey, E.; Turroni, F.; Mahony, J.; Belzer, C.; Delgado Palacio, S.; Arboleya Montes, S.; Mancabelli, L.; et al. The First Microbial Colonizers of the Human Gut: Composition, Activities, and Health Implications of the Infant Gut Microbiota. Microbiol. Mol. Biol. Rev. 2017, 81, e00036-17. [Google Scholar] [CrossRef] [PubMed]
- Xiao, L.; Zhao, F. Microbial transmission, colonisation and succession: From pregnancy to infancy. Gut 2023, 72, 772–786. [Google Scholar] [CrossRef]
- Zhou, L.; Xiao, X. The role of gut microbiota in the effects of maternal obesity during pregnancy on offspring metabolism. Biosci. Rep. 2018, 38, BSR20171234. [Google Scholar] [CrossRef]
- Dabelea, D. The predisposition to obesity and diabetes in offspring of diabetic mothers. Diabetes Care 2007, 30 (Suppl. S2), S169–S174. [Google Scholar] [CrossRef]
- Groer, M.; Luciano, A.A.; Dishaw, L.J.; Ashmeade, T.L.; Miller, E.M.; Gilbert, J.A. Development of the preterm infant gut microbiome: A research priority. Microbiome 2014, 2, 38. [Google Scholar] [CrossRef]
- Unger, S.; Stintzi, A.; Shah, P.; Mack, D.; O’Connor, D.L. Gut microbiota of the very- low-birth-weight infant. Pediatr. Res. 2015, 77, 205–213. [Google Scholar] [CrossRef]
- Mischke, M.; Plösch, T. More than just a gut instinct–the potential interplay between a baby’s nutrition, its gut microbiome, and the epigenome. Am. J. Physiol. Integr. Comp. Physiol. 2013, 304, R1065–R1069. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Huang, Y.; Lu, W.; Zeng, M.; Hu, X.; Su, Z.; Liu, Y.; Liu, Z.; Yuan, J.; Li, L.; Zhang, X.; et al. Mapping the early life gut microbiome in neonates with critical congenital heart disease: Multiomics insights and implications for host metabolic and immunological health. Microbiome 2022, 10, 245. [Google Scholar] [CrossRef]
- Tsiaoussis, J.; Antoniou, M.N.; Koliarakis, I.; Mesnage, R.; Vardavas, C.I.; Izotov, B.N.; Psaroulaki, A.; Tsatsakis, A. Effects of single and combined toxic exposures on the gut microbiome: Current knowledge and future directions. Toxicol. Lett. 2019, 312, 72–97. [Google Scholar] [CrossRef]
- Singh, R.D.; Koshta, K.; Tiwari, R.; Khan, H.; Sharma, V.; Srivastava, V. Developmental Exposure to Endocrine Disrupting Chemicals and Its Impact on Cardio-Metabolic-Renal Health. Front. Toxicol. 2021, 3, 663372. [Google Scholar] [CrossRef]
- Roseboom, T.; de Rooij, S.; Painter, R. The Dutch famine and its long-term consequences for adult health. Early Hum. Dev. 2006, 82, 485–491. [Google Scholar] [CrossRef]
- Santos, M.S.; Joles, J.A. Early determinants of cardiovascular disease. Best Pract. Res. Clin. Endocrinol. Metab. 2012, 26, 581–597. [Google Scholar]
- Hult, M.; Tornhammar, P.; Ueda, P.; Chima, C.; Bonamy, A.-K.E.; Ozumba, B.; Norman, M. Hypertension, diabetes and overweight: Looming legacies of the biafran famine. PLoS ONE 2010, 5, e13582. [Google Scholar] [CrossRef]
- Dalziel, S.R.; Walker, N.K.; Parag, V.; Mantell, C.; Rea, H.H.; Rodgers, A.; Harding, J.E. Cardiovascular risk factors after antenatal exposure to betamethasone: 30-year follow-up of a randomised controlled trial. Lancet 2005, 365, 1856–1862. [Google Scholar] [CrossRef]
- Antonucci, R.; Zaffanello, M.; Puxeddu, E.; Porcella, A.; Cuzzolin, L.; Pilloni, M.D.; Fanos, V. Use of non-steroidal anti-inflammatory drugs in pregnancy: Impact on the fetus and newborn. Curr. Drug Metab. 2012, 13, 474–490. [Google Scholar] [CrossRef] [PubMed]
- Barker, D.J.; Winter, P.D.; Osmond, C.; Margetts, B.; Simmonds, S.J. Weight in infancy and death from ischaemic heart disease. Lancet 1989, 2, 577–580. [Google Scholar] [CrossRef]
- Halvorsen, C.P.; Andolf, E.; Hu, J.; Pilo, C.; Winbladh, B.; Norman, M. Discordant twin growth in utero and differences in blood pressure and endothelial function at 8 years of age. J. Intern. Med. 2006, 259, 155–163. [Google Scholar] [CrossRef] [PubMed]
- Guimarães, K.S.L.; Braga, V.A.; Noronha, S.I.S.R.; Costa, W.K.A.D.; Makki, K.; Cruz, J.C.; Brandão, L.R.; Chianca Junior, D.A.; Meugnier, E.; Leulier, F.; et al. Lactiplantibacillus plantarum WJL administration during pregnancy and lactation improves lipid profile, insulin sensitivity and gut microbiota diversity in dyslipidemic dams and protects male offspring against cardiovascular dysfunction in later life. Food Funct. 2020, 11, 8939–8950. [Google Scholar] [CrossRef]
- Hsu, C.N.; Chang-Chien, G.P.; Lin, S.; Hou, C.Y.; Tain, Y.L. Targeting on gut microbial metabolite trimethylamine-N-Oxide and short-chain fatty acid to prevent maternal high-fructose-diet-induced developmental programming of hypertension in adult male offspring. Mol. Nutr. Food Res. 2019, 63, e1900073. [Google Scholar] [CrossRef]
- Hsu, C.N.; Hou, C.Y.; Chang-Chien, G.P.; Lin, S.; Yang, H.W.; Tain, Y.L. Perinatal resveratrol therapy prevents hypertension programmed by maternal chronic kidney disease in adult male offspring: Implications of the gut microbiome and their metabolites. Biomedicines 2020, 8, 567. [Google Scholar] [CrossRef]
- Hsu, C.N.; Chan, J.Y.H.; Wu, K.L.H.; Yu, H.R.; Lee, W.C.; Hou, C.Y.; Tain, Y.L. Altered gut microbiota and its metabolites in hypertension of developmental origins: Exploring differences between fructose and antibiotics exposure. Int. J. Mol. Sci. 2021, 22, 2674. [Google Scholar] [CrossRef]
- Hsu, C.N.; Hou, C.Y.; Chang-Chien, G.P.; Lin, S.; Chan, J.Y.H.; Lee, C.T.; Tain, Y.L. Maternal resveratrol therapy protected adult rat offspring against hypertension programmed by combined exposures to asymmetric dimethylarginine and trimethylamine-Noxide. J. Nutr. Biochem. 2021, 93, 108630. [Google Scholar] [CrossRef]
- Li, H.B.; Yang, T.; Richards, E.M.; Pepine, C.J.; Raizada, M.K. Maternal Treatment with captopril persistently alters gut-brain communication and attenuates hypertension of male offspring. Hypertension 2020, 75, 1315–1324. [Google Scholar] [CrossRef]
- Friedman, J.E.; Dobrinskikh, E.; Alfonso-Garcia, A.; Fast, A.; Janssen, R.C.; Soderborg, T.K.; Anderson, A.L.; Reisz, J.A.; D’Alessandro, A.; Frank, D.N.; et al. Pyrroloquinoline quinone prevents developmental programming of microbial dysbiosis and macrophage polarization to attenuate liver fibrosis in offspring of obese mice. Hepatol. Commun. 2018, 2, 313–328. [Google Scholar] [CrossRef]
- Sherman, S.B.; Sarsour, N.; Salehi, M.; Schroering, A.; Mell, B.; Joe, B.; Hill, J.W. Prenatal androgen exposure causes hypertension and gut microbiota dysbiosis. Gut Microbes 2018, 9, 400–421. [Google Scholar] [CrossRef]
- de Oliveira, Y.; Cavalcante, R.G.S.; Cavalcanti Neto, M.P.; Magnani, M.; Braga, V.A.; de Souza, E.L.; de Brito Alves, J.L. Oral administration of Lactobacillus fermentum post-weaning improves the lipid profile and autonomic dysfunction in rat offspring exposed to maternal dyslipidemia. Food Funct. 2020, 11, 5581–5594. [Google Scholar] [CrossRef]
- Marzullo, P.; Di Renzo, L.; Pugliese, G.; De Siena, M.; Barrea, L.; Muscogiuri, G.; Colao, A.; Savastano, S. Obesity Programs of nutrition, Education, Research and Assessment (OPERA) Group. From obesity through gut microbiota to cardiovascular diseases: A dangerous journey. Int. J. Obes. Suppl. 2020, 10, 35–49. [Google Scholar] [CrossRef] [PubMed]
- Blackmore, H.L.; Ozanne, S.E. Programming of cardiovascular disease across the life-course. J. Mol. Cell. Cardiol. 2015, 83, 122–130. [Google Scholar] [CrossRef] [PubMed]
- Tain, Y.L.; Hsu, C.N. Interplay between maternal nutrition and epigenetic programming on offspring hypertension. J. Nutr. Biochem. 2024, 127, 109604. [Google Scholar] [CrossRef]
- Pluznick, J.L.; Protzko, R.J.; Gevorgyan, H.; Peterlin, Z.; Sipos, A.; Han, J.; Brunet, I.; Wan, L.X.; Rey, F.; Wang, T.; et al. Olfactory receptor responding to gut microbiota-derived signals plays a role in renin secretion and blood pressure regulation. Proc. Natl. Acad. Sci. USA 2013, 110, 4410–4415. [Google Scholar] [CrossRef]
- Hsu, C.N.; Lin, I.C.; Yu, H.R.; Huang, L.T.; Tiao, M.M.; Tain, Y.L. Maternal tryptophan supplementation protects adult rat offspring against hypertension programmed by maternal chronic kidney disease: Implication of tryptophan-metabolizing microbiome and aryl hydrocarbon receptor. Int. J. Mol. Sci. 2020, 21, 4552. [Google Scholar] [CrossRef]
- Oliveira Andrade, J.M.; de Farias Lelis, D.; Mafra, V.; Cota, J. The angiotensin converting enzyme 2 (ACE2), gut microbiota, and cardiovascular health. Protein Pept. Lett. 2017, 24, 827–832. [Google Scholar] [CrossRef]
- Dennery, P.A. Oxidative stress in development: Nature or nurture? Free Radic. Biol. Med. 2010, 49, 1147–1151. [Google Scholar]
- Paixão, A.D.; Alexander, B.T. How the kidney is impacted by the perinatal maternal environment to develop hypertension. Biol. Reprod. 2013, 89, 144. [Google Scholar] [CrossRef]
- Liu, Y.; Qi, L.; Wu, J.; Xu, T.; Yang, C.; Chen, X.; Lv, J.; Xu, Z. Prenatal high-salt diet impaired vasodilatation with reprogrammed renin-angiotensin system in offspring rats. J. Hypertens. 2018, 36, 2369–2379. [Google Scholar] [CrossRef]
- Svitok, P.; Okuliarova, M.; Varga, I.; Zeman, M. Renal impairment induced by prenatal exposure to angiotensin II in male rat offspring. Exp. Biol. Med. 2019, 244, 923–931. [Google Scholar] [CrossRef]
- Tain, Y.L.; Hsieh, C.S.; Lin, I.C.; Chen, C.C.; Sheen, J.M.; Huang, L.T. Effects of maternal L-citrulline supplementation on renal function and blood pressure in offspring exposed to maternal caloric restriction: The impact of nitric oxide pathway. Nitric Oxide 2010, 23, 34–41. [Google Scholar] [CrossRef] [PubMed]
- Do Nascimento, L.C.P.; Neto, J.P.R.C.; de Andrade Braga, V.; Lagranha, C.J.; de Brito Alves, J.L. Maternal exposure to high-fat and high-cholesterol diet induces arterial hypertension and oxidative stress along the gut-kidney axis in rat offspring. Life Sci. 2020, 261, 118367. [Google Scholar] [CrossRef] [PubMed]
- Chen, L.; Zadi, Z.H.; Zhang, J.; Scharf, S.M.; Pae, E.K. Intermittent hypoxia in utero damages postnatal growth and cardiovascular function in rats. J. Appl. Physiol. 2018, 124, 821–830. [Google Scholar] [CrossRef]
- Hsu, C.N.; Yang, H.W.; Hou, C.Y.; Chang-Chien, G.P.; Lin, S.; Tain, Y.L. Maternal Adenine-Induced Chronic Kidney Disease Programs Hypertension in Adult Male Rat Offspring: Implications of Nitric Oxide and Gut Microbiome Derived Metabolites. Int. J. Mol. Sci. 2020, 21, 7237. [Google Scholar] [CrossRef]
- Piecha, G.; Koleganova, N.; Ritz, E.; Müller, A.; Fedorova, O.V.; Bagrov, A.Y.; Lutz, D.; Schirmacher, P.; Gross-Weissmann, M.L. High salt intake causes adverse fetal programming–vascular effects beyond blood pressure. Nephrol. Dial. Transplant. 2012, 27, 3464–3476. [Google Scholar] [CrossRef]
- Campbell, E.L.; Colgan, S.P. Control and dysregulation of redox signalling in the gastrointestinal tract. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 106–120. [Google Scholar] [CrossRef]
- Halliwell, B. Understanding mechanisms of antioxidant action in health and disease. Nat. Rev. Mol. Cell Biol. 2024, 25, 13–33. [Google Scholar] [CrossRef]
- Buonocore, G.; Groenendaal, F. Anti-oxidant strategies. Semin. Fetal Neonatal Med. 2007, 12, 287–295. [Google Scholar] [CrossRef]
- Barteková, M.; Adameová, A.; Görbe, A.; Ferenczyová, K.; Pecháňová, O.; Lazou, A.; Dhalla, N.S.; Ferdinandy, P.; Giricz, Z. Natural and synthetic antioxidants targeting cardiac oxidative stress and redox signaling in cardiometabolic diseases. Free Radic. Biol. Med. 2021, 169, 446–477. [Google Scholar] [CrossRef] [PubMed]
- National Academy of Sciences, Institute of Medicine. Dietary Reference Intakes for Vitamin C, Vitamin E, Selenium, and Carotenoids: A Report of the Panel on Dietary Antioxidants and Related Compounds, Subcommittees on Upper Reference Levels of Nutrients and on Interpretation and Use of Dietary Reference Intakes, and the Standing Committee on Scientific Evaluation of Dietary Reference Intakes, Food and Nutrition Board; Institute of Medicine, 17 National Academy Press: Washington, DC, USA, 2000. [Google Scholar]
- Tain, Y.L.; Hsu, C.N. Metabolic Syndrome Programming and Reprogramming: Mechanistic Aspects of Oxidative Stress. Antioxidants 2022, 11, 2108. [Google Scholar] [CrossRef] [PubMed]
- Tain, Y.L.; Hsu, C.N. Amino Acids during Pregnancy and Offspring Cardiovascular-Kidney-Metabolic Health. Nutrients 2024, 16, 1263. [Google Scholar] [CrossRef]
- Tain, Y.L.; Hsu, C.N. Melatonin Use during Pregnancy and Lactation Complicated by Oxidative Stress: Focus on Offspring’s Cardiovascular-Kidney-Metabolic Health in Animal Models. Antioxidants 2024, 13, 226. [Google Scholar] [CrossRef]
- Zhang, F.F.; Barr, S.I.; McNulty, H.; Li, D.; Blumberg, J.B. Health effects of vitamin and mineral supplements. BMJ 2020, 369, 2511. [Google Scholar] [CrossRef]
- Said, H.M.; Nexo, E. Gastrointestinal Handling of Water-Soluble Vitamins. Compr. Physiol. 2018, 8, 1291–1311. [Google Scholar] [CrossRef]
- Niki, E. Role of vitamin E as a lipid-soluble peroxyl radical scavenger: In vitro and in vivo evidence. Free Radic. Biol. Med. 2014, 66, 3–12. [Google Scholar] [CrossRef]
- Wang, J.; Yin, N.; Deng, Y.; Wei, Y.; Huang, Y.; Pu, X.; Li, L.; Zheng, Y.; Guo, J.; Yu, J.; et al. Ascorbic Acid Protects against Hypertension through Downregulation of ACE1 Gene Expression Mediated by Histone Deacetylation in Prenatal InflammationInduced Offspring. Sci. Rep. 2016, 6, 39469. [Google Scholar] [CrossRef]
- Farias, J.S.; Santos, K.M.; Lima, N.K.S.; Cabral, E.V.; Aires, R.S.; Veras, A.C.; Paixão, A.D.; Vieira, L.D. Maternal endotoxemia induces renal collagen deposition in adult offspring: Role of NADPH oxidase/TGF-β1/MMP-2signalingpathway. Arch. Biochem. Biophys. 2020, 684, 108306. [Google Scholar] [CrossRef]
- Franco Mdo, C.; Ponzio, B.F.; Gomes, G.N.; Gil, F.Z.; Tostes, R.; Carvalho, M.H.; Fortes, Z.B. Micronutrient prenatal supplementation prevents the development of hypertension and vascular endothelial damage induced by intrauterine malnutrition. Life Sci. 2009, 85, 327–333. [Google Scholar] [CrossRef]
- Palinski, W.; D’Armiento, F.P.; Witztum, J.L.; de Nigris, F.; Casanada, F.; Condorelli, M.; Silvestre, M.; Napoli, C. Maternal hypercholesterolemia and treatment during pregnancy influence the long-term progression of atherosclerosis in offspring of rabbits. Circ. Res. 2001, 89, 991–996. [Google Scholar] [CrossRef]
- Torrens, C.; Brawley, L.; Anthony, F.W.; Dance, C.S.; Dunn, R.; Jackson, A.A.; Poston, L.; Hanson, M.A. Folate supplementation during pregnancy improves offspring cardiovascular dysfunction induced by protein restriction. Hypertension 2006, 47, 982–987. [Google Scholar] [CrossRef]
- Korsmo, H.W.; Edwards, K.; Dave, B.; Jack-Roberts, C.; Yu, H.; Saxena, A.; Salvador, M.; Dembitzer, M.; Phagoora, J.; Jiang, X. Prenatal Choline Supplementation during High-Fat Feeding Improves Long-Term Blood Glucose Control in Male Mouse Offspring. Nutrients 2020, 12, 144. [Google Scholar] [CrossRef]
- Korsmo, H.W.; Kadam, I.; Reaz, A.; Bretter, R.; Saxena, A.; Johnson, C.H.; Caviglia, J.M.; Jiang, X. Prenatal Choline Supplement in a Maternal Obesity Model Modulates Offspring Hepatic Lipidomes. Nutrients 2023, 15, 965. [Google Scholar] [CrossRef]
- Wang, J.; Wu, Z.; Li, D.; Li, N.; Dindot, S.V.; Satterfield, M.C.; Bazer, F.W.; Wu, G. Nutrition, epigenetics, and metabolic syndrome. Antioxid. Redox Signal. 2012, 17, 282–301. [Google Scholar] [CrossRef]
- Li, K.; Wahlqvist, M.L.; Li, D. Nutrition, One-Carbon Metabolism and Neural Tube Defects: A Review. Nutrients 2016, 8, 741. [Google Scholar] [CrossRef]
- Terstappen, F.; Tol, A.J.C.; Gremmels, H.; Wever, K.E.; Paauw, N.D.; Joles, J.A.; Beek, E.M.V.; Lely, A.T. Prenatal Amino Acid Supplementation to Improve Fetal Growth: A Systematic Review and Meta-Analysis. Nutrients 2020, 12, 2535. [Google Scholar] [CrossRef]
- Ali, S.S.; Ahsan, H.; Zia, M.K.; Siddiqui, T.; Khan, F.H. Understanding oxidants and antioxidants: Classical team with new players. J. Food Biochem. 2020, 44, e13145. [Google Scholar] [CrossRef]
- Luiking, Y.C.; Ten Have, G.A.M.; Wolfe, R.R.; Deutz, N.E.P. Arginine de novo and nitric oxide production in disease states. Am. J. Physiol. Endocrinol. Metab. 2012, 303, E1177–E1189. [Google Scholar] [CrossRef]
- Schwedhelm, E.; Maas, R.; Freese, R.; Jung, D.; Lukacs, Z.; Jambrecina, A.; Spickler, W.; Schulze, F.; Boger, R.H. Pharmacokinetic and pharmacodynamic properties of oral L-citrulline and L-arginine: Impact on nitric oxide metabolism. Br. J. Clin. Pharmacol. 2008, 65, 51–59. [Google Scholar] [CrossRef]
- Luo, K.; Chen, P.; Li, S.; Li, W.; He, M.; Wang, T.; Chen, J. Effect of L-arginine supplementation on the hepatic phosphatidylinositol 3-kinase signaling pathway and gluconeogenic enzymes in early intrauterine growth-restricted rats. Exp. Ther. Med. 2017, 14, 2355–2360. [Google Scholar] [CrossRef]
- Tain, Y.L.; Sheen, J.M.; Chen, C.C.; Yu, H.R.; Tiao, M.M.; Kuo, H.C.; Huang, L.T. Maternal citrulline supplementation prevents prenatal dexamethasone-induced programmed hypertension. Free Radic. Res. 2014, 48, 580–586. [Google Scholar] [CrossRef]
- Tain, Y.L.; Lee, W.C.; Hsu, C.N.; Lee, W.C.; Huang, L.T.; Lee, C.T.; Lin, C.Y. Asymmetric dimethylarginine is associated with developmental programming of adult kidney disease and hypertension in offspring of streptozotocin-treated mothers. PLoS ONE 2013, 8, e55420. [Google Scholar] [CrossRef]
- Tain, Y.L.; Hou, C.Y.; Chang-Chien, G.P.; Lin, S.; Hsu, C.N. Perinatal Use of Citrulline Rescues Hypertension in Adult Male Offspring Born to Pregnant Uremic Rats. Int. J. Mol. Sci. 2024, 25, 1612. [Google Scholar] [CrossRef]
- Boucknooghe, T.; Remacle, C.; Reusens, B. Is taurine a functional nutrient? Curr. Opin. Clin. Nutr. Metab. Care 2006, 9, 728–733. [Google Scholar] [CrossRef]
- Thaeomor, A.; Teangphuck, P.; Chaisakul, J.; Seanthaweesuk, S.; Somparn, N.; Roysommuti, S. Perinatal Taurine Supplementation Prevents Metabolic and Cardiovascular Effects of Maternal Diabetes in Adult Rat Offspring. Adv. Exp. Med. Biol. 2017, 975, 295–305. [Google Scholar]
- Thaeomor, A.; Tangnoi, C.; Seanthaweesuk, S.; Somparn, N.; Roysommuti, S. Perinatal Taurine Supplementation Prevents the Adverse Effects of Maternal Dyslipidemia on Growth and Cardiovascular Control in Adult Rat Offspring. Adv. Exp. Med. Biol. 2019, 1155, 415–427. [Google Scholar]
- Mensegue, M.F.; Burgueño, A.L.; Tellechea, M.L. Perinatal taurine exerts a hypotensive effect in male spontaneously hypertensive rats and down-regulates endothelial oxide nitric synthase in the aortic arch. Clin. Exp. Pharmacol. Physiol. 2020, 47, 780–789. [Google Scholar] [CrossRef]
- Tain, Y.L.; Hou, C.Y.; Chang-Chien, G.P.; Lin, S.; Hsu, C.N. Protective Role of Taurine on Rat Offspring Hypertension in the Setting of Maternal Chronic Kidney Disease. Antioxidants 2023, 12, 2059. [Google Scholar] [CrossRef]
- Hsu, C.N.; Hou, C.Y.; Chang-Chien, G.P.; Lin, S.; Tain, Y.L. Dietary Supplementation with Cysteine during Pregnancy Rescues Maternal Chronic Kidney Disease-Induced Hypertension in Male Rat Offspring: The Impact of Hydrogen Sulfide and Microbiota-Derived Tryptophan Metabolites. Antioxidants 2022, 11, 483. [Google Scholar] [CrossRef] [PubMed]
- Jackson, A.A.; Dunn, R.L.; Marchand, M.C.; Langley-Evans, S.C. Increased systolic blood pressure in rats induced by a maternal low-protein diet is reversed by dietary supplementation with glycine. Clin. Sci. 2002, 103, 633–639. [Google Scholar] [CrossRef]
- Fujii, T.; Yura, S.; Tatsumi, K.; Kondoh, E.; Mogami, H.; Fujita, K.; Kakui, K.; Aoe, S.; Itoh, H.; Sagawa, N.; et al. Branched-chain amino acid supplemented diet during maternal food restriction prevents developmental hypertension in adult rat offspring. J. Dev. Orig. Health Dis. 2011, 2, 176–183. [Google Scholar] [CrossRef]
- Fortunato, M.C.; Neves, J.; Pais, M.L.; Fonseca, C.; Silva, D.; Martins, J.; Castelo-Branco, M.; Fortuna, A.; Gonçalves, J. Maternal Tryptophan Supplementation Alters Offspring Gut-Brain Axis and Behavior in a Sex-Specific Manner. J. Neurochem. 2025, 169, e70161. [Google Scholar] [CrossRef]
- Tamura, H.; Nakamura, Y.; Terron, M.P.; Flores, L.J.; Manchester, L.C.; Tan, D.X.; Sugino, N.; Reiter, R.J. Melatonin and pregnancy in the human. Reprod. Toxicol. 2008, 25, 291–303. [Google Scholar] [CrossRef]
- Reiter, R.J.; Mayo, J.C.; Tan, D.X.; Sainz, R.M.; Alatorre-Jimenez, M.; Qin, L. Melatonin as an antioxidant: Under promises but over delivers. J. Pineal Res. 2016, 61, 253–278. [Google Scholar] [CrossRef]
- Reiter, R.J.; Tan, D.X.; Terron, M.P.; Flores, L.J.; Czarnocki, Z. Melatonin and its metabolites: New findings regarding their production and their radical scavenging actions. Acta Biochim. Pol. 2007, 54, 1–9. [Google Scholar] [CrossRef]
- Bonmatí-Carrión, M.Á.; Rol, M.A. Melatonin as a Mediator of the Gut Microbiota-Host Interaction: Implications for Health and Disease. Antioxidants 2023, 13, 34. [Google Scholar] [CrossRef]
- Tain, Y.L.; Huang, L.T.; Hsu, C.N. Developmental Programming of Adult Disease: Reprogramming by Melatonin? Int. J. Mol. Sci. 2017, 18, 426. [Google Scholar] [CrossRef]
- Tain, Y.L.; Lin, Y.J.; Chan, J.Y.H.; Lee, C.T.; Hsu, C.N. Maternal melatonin or agomelatine therapy prevents programmed hypertension in male offspring of mother exposed to continuous light. Biol. Reprod. 2017, 97, 636–643. [Google Scholar] [CrossRef]
- Lee, S.K.; Sirajudeen, K.N.; Sundaram, A.; Zakaria, R.; Singh, H.J. Effects of antenatal, postpartum and post-weaning melatonin supplementation on blood pressure and renal antioxidant enzyme activities in spontaneously hypertensive rats. J. Physiol. Biochem. 2011, 67, 249–257. [Google Scholar] [CrossRef]
- Kim, C.Y.; Lee, B.N.; Kim, J.S. Effects of maternal-melatonin treatment on open-field behaviors and hypertensive phenotype in spontaneously hypertensive rats’ (SHR) offspring. Exp. Anim. 2002, 51, 69–74. [Google Scholar] [CrossRef]
- Hansell, J.A.; Richter, H.G.; Camm, E.J.; Herrera, E.A.; Blanco, C.E.; Villamor, E.; Patey, O.V.; Lock, M.C.; Trafford, A.W.; Galli, G.L.J.; et al. Maternal melatonin: Effective intervention against developmental programming of cardiovascular dysfunction in adult offspring of complicated pregnancy. J. Pineal Res. 2022, 72, e12766. [Google Scholar] [CrossRef]
- Goszcz, K.; Deakin, S.J.; Duthie, G.G.; Stewart, D.; Leslie, S.J.; Megson, I.L. Antioxidants in Cardiovascular Therapy: Panacea or False Hope? Front. Cardiovasc. Med. 2015, 2, 29. [Google Scholar] [CrossRef] [PubMed]
- Petrosillo, G.; Moro, N.; Ruggiero, F.M.; Paradies, G. Melatonin Inhibits Cardiolipin Peroxidation in Mitochondria and Prevents the Mitochondrial Permeability Transition and Cytochrome c Release. Free Radic. Biol. Med. 2009, 47, 969–974. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.; Li, L.N.; Guo, S.; Zhao, X.Y.; Liu, Y.Z.; Liang, C.; Tu, S.; Wang, D.; Li, L.; Dong, J.Z.; et al. Melatonin Improves Cardiac Function in a Mouse Model of Heart Failure with Preserved Ejection Fraction. Redox Biol. 2018, 18, 211–221. [Google Scholar] [CrossRef]
- Veneroso, C.; Tuñón, M.J.; González-Gallego, J.; Collado, P.S. Melatonin Reduces Cardiac Inflammatory Injury Induced by Acute Exercise. J. Pineal Res. 2009, 47, 184–191. [Google Scholar] [CrossRef]
- Li, F.; Lai, J.; Ma, F.; Cai, Y.; Li, S.; Feng, Z.; Lu, Z.; Liu, X.; Ke, Q.; Hao, H.; et al. Maternal melatonin supplementation shapes gut microbiota and protects against inflammation in early life. Int. Immunopharmacol. 2023, 120, 110359. [Google Scholar] [CrossRef]
- Hsu, C.N.; Yang, H.W.; Hou, C.Y.; Chang-Chien, G.P.; Lin, S.; Tain, Y.L. Melatonin Prevents Chronic Kidney Disease-Induced Hypertension in Young Rat Treated with Adenine: Implications of Gut Microbiota-Derived Metabolites. Antioxidants 2021, 10, 1211. [Google Scholar] [CrossRef]
- Vine, T.; Brown, G.M.; Frey, B.N. Melatonin use during pregnancy and lactation: A scoping review of human studies. Braz. J. Psychiatry 2022, 44, 342–348. [Google Scholar] [CrossRef]
- Boutin, J.A.; Kennaway, D.J.; Jockers, R. Melatonin: Facts, Extrapolations and Clinical Trials. Biomolecules 2023, 13, 943. [Google Scholar] [CrossRef]
- Chen, Y.C.; Tain, Y.L.; Sheen, J.M.; Huang, L.T. Melatonin utility in neonates and children. J. Formos. Med. Assoc. 2012, 111, 57–66. [Google Scholar] [CrossRef]
- Zeb, A. Concept, mechanism, and applications of phenolic antioxidants in foods. J. Food Biochem. 2020, 44, e13394. [Google Scholar] [CrossRef]
- Iqbal, I.; Wilairatana, P.; Saqib, F.; Nasir, B.; Wahid, M.; Latif, M.F.; Iqbal, A.; Naz, R.; Mubarak, M.S. Plant Polyphenols and Their Potential Benefits on Cardiovascular Health: A Review. Molecules 2023, 28, 6403. [Google Scholar] [CrossRef]
- Hussain, T.; Tan, B.; Yin, Y.; Blachier, F.; Tossou, M.C.; Rahu, N. Oxidative Stress and Inflammation: What Polyphenols Can Do for Us? Oxidative Med. Cell. Longev. 2016, 2016, 7432797. [Google Scholar] [CrossRef] [PubMed]
- Urquiaga, I.; Leighton, F. Plant polyphenol antioxidants and oxidative stress. Biol. Res. 2000, 33, 55–64. [Google Scholar] [CrossRef] [PubMed]
- Singh, A.P.; Singh, R.; Verma, S.S.; Rai, V.; Kaschula, C.H.; Maiti, P.; Gupta, S.C. Health benefits of resveratrol: Evidence from clinical studies. Med. Res. Rev. 2019, 39, 1851–1891. [Google Scholar] [CrossRef]
- Cheng, H.; Zhang, D.; Wu, J.; Liu, J.; Zhou, Y.; Tan, Y.; Feng, W.; Peng, C. Interactions between gut microbiota and polyphenols: A mechanistic and metabolomic review. Phytomedicine 2023, 119, 154979. [Google Scholar] [CrossRef]
- Wang, Z.M.; Zhou, B.; Wang, Y.S.; Gong, Q.Y.; Wang, Q.M.; Yan, J.J.; Gao, W.; Wang, L.S. Black and green tea consumption and the risk of coronary artery disease: A meta-analysis. Am. J. Clin. Nutr. 2011, 93, 506–515. [Google Scholar] [CrossRef]
- Hartley, L.; Flowers, N.; Holmes, J.; Clarke, A.; Stranges, S.; Hooper, L.; Rees, K. Green and black tea for the primary prevention of cardiovascular disease. Cochrane Database Syst. Rev. 2013, 2013, CD009934. [Google Scholar] [CrossRef]
- Chiva-Blanch, G.; Badimon, L. Effects of Polyphenol Intake on Metabolic Syndrome: Current Evidences from Human Trials. Oxidative Med. Cell. Longev. 2017, 2017, 5812401. [Google Scholar] [CrossRef] [PubMed]
- Ellwood, L.; Torun, G.; Bahar, Z.; Fernandez, R. Effects of flavonoid-rich fruits on hypertension in adults: A systematic review. JBI Database Syst. Rev. Implement. Rep. 2019, 17, 2075–2105. [Google Scholar] [CrossRef] [PubMed]
- Durazzo, A.; Lucarini, M.; Souto, E.B.; Cicala, C.; Caiazzo, E.; Izzo, A.A.; Novellino, E.; Santini, A. Polyphenols: A concise overview on the chemistry, occurrence, and human health. Phytother. Res. 2019, 33, 2221–2243. [Google Scholar] [CrossRef] [PubMed]
- Liang, C.; Oest, M.E.; Prater, M.R. Intrauterine exposure to high saturated fat diet elevates risk of adult-onset chronic diseases in C57BL/6 mice. Birth Defects Res. B Dev. Reprod. Toxicol. 2009, 86, 377–384. [Google Scholar] [CrossRef]
- Lamothe, J.; Khurana, S.; Tharmalingam, S.; Williamson, C.; Byrne, C.J.; Lees, S.J.; Khaper, N.; Kumar, A.; Tai, T.C. Oxidative Stress Mediates the Fetal Programming of Hypertension by Glucocorticoids. Antioxidants 2021, 10, 531. [Google Scholar] [CrossRef]
- Kursvietiene, L.; Staneviciene, I.; Mongirdiene, A.; Bernatoniene, J. Multiplicity of effects and health benefits of resveratrol. Medicina 2016, 52, 148–155. [Google Scholar] [CrossRef]
- Zhang, Y.; Cheng, J.; Jian, W. Unlocking Resveratrol’s Potential: Targeting Ferroptosis in Atherosclerosis Through MAPK1. Food Sci. Nutr. 2025, 13, e70466. [Google Scholar] [CrossRef]
- Ros, P.; Díaz, F.; Freire-Regatillo, A.; Argente-Arizón, P.; Barrios, V.; Argente, J.; Chowen, J.A. Resveratrol Intake during Pregnancy and Lactation Modulates the Early Metabolic Effects of Maternal Nutrition Differently in Male and Female Offspring. Endocrinology 2018, 159, 810–825. [Google Scholar] [CrossRef]
- Zou, T.; Chen, D.; Yang, Q.; Wang, B.; Zhu, M.J.; Nathanielsz, P.W.; Du, M. Resveratrol supplementation of high-fat diet-fed pregnant mice promotes brown and beige adipocyte development and prevents obesity in male offspring. J. Physiol. 2017, 595, 1547–1562. [Google Scholar] [CrossRef]
- Liu, T.Y.; Yu, H.R.; Tsai, C.C.; Huang, L.T.; Chen, C.C.; Sheen, J.M.; Tiao, M.M.; Tain, Y.L.; Lin, I.C.; Lai, Y.J.; et al. Resveratrol intake during pregnancy and lactation re-programs adiposity and ameliorates leptin resistance in male progeny induced by maternal high-fat/high sucrose plus postnatal high-fat/high sucrose diets via fat metabolism regulation. Lipids Health Dis. 2020, 19, 174. [Google Scholar] [CrossRef]
- Tain, Y.L.; Lee, W.C.; Wu, K.L.H.; Leu, S.; Chan, J.Y.H. Resveratrol Prevents the Development of Hypertension Programmed by Maternal Plus Post-Weaning High-Fructose Consumption through Modulation of Oxidative Stress, Nutrient-Sensing Signals, and Gut Microbiota. Mol. Nutr. Food Res. 2018, 30, e1800066. [Google Scholar] [CrossRef]
- Chen, H.E.; Lin, Y.J.; Lin, I.C.; Yu, H.R.; Sheen, J.M.; Tsai, C.C.; Huang, L.T.; Tain, Y.L. Resveratrol prevents combined prenatal NG-nitro-L-arginine-methyl ester (L-NAME) treatment plus postnatal high-fat diet induced programmed hypertension in adult rat offspring: Interplay between nutrient-sensing signals, oxidative stress and gut microbiota. J. Nutr. Biochem. 2019, 70, 28–37. [Google Scholar] [CrossRef]
- Scalbert, A.; Williamson, G. Dietary intake and bioavailability of polyphenols. J. Nutr. 2000, 130, 2073S–2085S. [Google Scholar] [CrossRef]
- Ferrer, M.; Buey, B.; Grasa, L.; Mesonero, J.E.; Latorre, E. Protective role of short-chain fatty acids on intestinal oxidative stress induced by TNF-α. Cell Stress Chaperones 2024, 29, 769–776. [Google Scholar] [CrossRef]
- Qin, X.; Zhang, M.; Chen, S.; Tang, Y.; Cui, J.; Ding, G. Short-chain fatty acids in fetal development and metabolism. Trends Mol. Med. 2024, 31, 625–639. [Google Scholar] [CrossRef] [PubMed]
- Hsu, C.N.; Yu, H.R.; Chan, J.Y.H.; Lee, W.C.; Lin, I.C.; Wu, K.L.H.; Hou, C.Y.; Chang-Chien, G.P.; Lin, S.; Tain, Y.L. Maternal Acetate Supplementation Reverses Blood Pressure Increase in Male Offspring Induced by Exposure to Minocycline during Pregnancy and Lactation. Int. J. Mol. Sci. 2022, 23, 7924. [Google Scholar] [CrossRef] [PubMed]
- Hsu, C.N.; Yu, H.R.; Lin, I.C.; Tiao, M.M.; Huang, L.T.; Hou, C.Y.; Chang-Chien, G.P.; Lin, S.; Tain, Y.L. Sodium butyrate modulates blood pressure and gut microbiota in maternal tryptophan-free diet-induced hypertension rat offspring. J. Nutr. Biochem. 2022, 108, 109090. [Google Scholar] [CrossRef]
- Tain, Y.L.; Hou, C.Y.; Chang-Chien, G.P.; Lin, S.; Tzeng, H.T.; Lee, W.C.; Wu, K.L.H.; Yu, H.R.; Chan, J.Y.H.; Hsu, C.N. Reprogramming Effects of Postbiotic Butyrate and Propionate on Maternal High-Fructose Diet-Induced Offspring Hypertension. Nutrients 2023, 15, 1682. [Google Scholar] [CrossRef]
- Šalamon, Š.; Kramar, B.; Marolt, T.P.; Poljšak, B.; Milisav, I. Medical and Dietary Uses of N-Acetylcysteine. Antioxidants 2019, 8, 111. [Google Scholar] [CrossRef]
- Van Goor, H.; van den Born, J.C.; Hillebrands, J.L.; Joles, J.A. Hydrogen sulfide in hypertension. Curr. Opin. Nephrol. Hypertens. 2016, 25, 107–113. [Google Scholar] [CrossRef]
- Hsu, C.N.; Hou, C.Y.; Chang-Chien, G.P.; Lin, S.; Tain, Y.L. Maternal N-Acetylcysteine Therapy Prevents Hypertension in Spontaneously Hypertensive Rat Offspring: Implications of Hydrogen Sulfide-Generating Pathway and Gut Microbiota. Antioxidants 2020, 9, 856. [Google Scholar] [CrossRef]
- Tain, Y.L.; Lee, C.T.; Chan, J.Y.; Hsu, C.N. Maternal melatonin or N-acetylcysteine therapy regulates hydrogen sulfide-generating pathway and renal transcriptome to prevent prenatal NG-Nitro-L-arginine-methyl ester (L-NAME)-induced fetal programming of hypertension in adult male offspring. Am. J. Obstet. Gynecol. 2016, 215, 636.e1–636.e72. [Google Scholar] [CrossRef] [PubMed]
- Tain, Y.L.; Hsu, C.N.; Lee, C.T.; Lin, Y.J.; Tsai, C.C. N-Acetylcysteine prevents programmed hypertension in male rat offspring born to suramin-treated mothers. Biol. Reprod. 2016, 95, 8. [Google Scholar] [CrossRef] [PubMed]
- Tai, I.H.; Sheen, J.M.; Lin, Y.J.; Yu, H.R.; Tiao, M.M.; Chen, C.C.; Huang, L.T.; Tain, Y.L. Maternal N-acetylcysteine therapy regulates hydrogen sulfide-generating pathway and prevents programmed hypertension in male offspring exposed to prenatal dexamethasone and postnatal high-fat diet. Nitric Oxide 2016, 53, 6–12. [Google Scholar] [CrossRef]
- Di Masi, A.; Ascenzi, P. H2S: A “double face” molecule in health and disease. Biofactors 2013, 39, 186–196. [Google Scholar] [CrossRef]
- Huang, S.H.; Leonard, S.; Shi, X.; Goins, M.R.; Vallyathan, V. Antioxidant activity of lazaroid (U-75412E) and its protective effects against crystalline silica-induced cytotoxicity. Free Radic. Biol. Med. 1998, 24, 529–536. [Google Scholar] [CrossRef]
- Dodson, M.; Castro-Portuguez, R.; Zhang, D.D. NRF2 plays a critical role in mitigating lipid peroxidation and ferroptosis. Redox Biol. 2019, 23, 101107. [Google Scholar] [CrossRef]
- Mills, E.L.; Ryan, D.G.; Prag, H.A.; Dikovskaya, D.; Menon, D.; Zaslona, Z.; Jedrychowski, M.P.; Costa, A.S.H.; Higgins, M.; Hams, E.; et al. Itaconate is an anti-inflammatory metabolite that activates Nrf2 via alkylation of KEAP1. Nature 2018, 556, 113–117. [Google Scholar] [CrossRef]
- Hsu, C.N.; Lin, Y.J.; Yu, H.R.; Lin, I.C.; Sheen, J.M.; Huang, L.T.; Tain, Y.L. Protection of Male Rat Offspring against Hypertension Programmed by Prenatal Dexamethasone Administration and Postnatal High-Fat Diet with the Nrf2 Activator Dimethyl Fumarate during Pregnancy. Int. J. Mol. Sci. 2019, 20, 3957. [Google Scholar] [CrossRef]
- Wilcox, C.S. Effects of tempol and redox-cycling nitroxides in models of oxidative stress. Pharmacol. Ther. 2010, 126, 119–145. [Google Scholar] [CrossRef]
- Pang, M.; Wang, S.; Shi, T.; Chen, J. Overview of MitoQ on prevention and management of cardiometabolic diseases: A scoping review. Front. Cardiovasc. Med. 2025, 12, 1506460. [Google Scholar] [CrossRef]
- Zhou, R.; Ma, P.; Xiong, A.; Xu, Y.; Wang, Y.; Xu, Q. Protective effects of low dose rosuvastatin on isoproterenol-induced chronic heart failure in rats by regulation of DDAH-ADMA-NO pathway. Cardiovasc. Ther. 2017, 35, e12241. [Google Scholar] [CrossRef] [PubMed]
- Bal, F.; Bekpinar, S.; Unlucerci, Y.; Kusku-Kiraz, Z.; Önder, S.; Uysal, M.; Gurdol, F. Antidiabetic drug metformin is effective on the metabolism of asymmetric dimethylarginine in experimental liver injury. Diabetes Res. Clin. Pract. 2014, 106, 295–302. [Google Scholar] [CrossRef]
- He, H.; Li, X.; Wang, H.; Zhang, W.; Jiang, H.; Wang, S.; Yuan, L.; Liu, Y.; Liu, X. Effects of salvianolic acid A on plasma and tissue dimethylarginine levels in a rat model of myocardial infarction. J. Cardiovasc. Pharmacol. 2013, 61, 482–488. [Google Scholar] [CrossRef] [PubMed]
- Zhang, W.; Zhang, J.; Liu, Y.K.; Liu, J.; Wang, X.; Xu, Q.; Wang, Y.; Xu, X.; Dai, G. Cardioprotective effects of oxymatrine on isoproterenol-induced heart failure via regulation of DDAH/ADMA metabolism pathway in rats. Eur. J. Pharmacol. 2014, 745, 29–35. [Google Scholar] [CrossRef]
- Onozato, M.L.; Tojo, A.; Leiper, J.; Fujita, T.; Palm, F.; Wilcox, C.S. Expression of NG,NG-dimethylarginine dimethylaminohydrolase and protein arginine N-methyltransferase isoforms in diabetic rat kidney: Effects of angiotensin II receptor blockers. Diabetes 2008, 57, 172–180. [Google Scholar] [CrossRef]
- Ojima, A.; Ishibashi, Y.; Matsui, T.; Maeda, S.; Nishino, Y.; Takeuchi, M.; Fukami, K.; Yamagishi, S. Glucagon-like peptide-1 receptor agonist inhibits asymmetric dimethylarginine generation in the kidney of streptozotocin-induced diabetic rats by blocking advanced glycation end product-induced protein arginine methyltranferase-1 expression. Am. J. Pathol. 2013, 182, 132–141. [Google Scholar] [CrossRef]
- Tain, Y.L.; Wu, K.L.H.; Lee, W.C.; Leu, S.; Chan, J.Y.H. Prenatal Metformin Therapy Attenuates Hypertension of Developmental Origin in Male Adult Offspring Exposed to Maternal High-Fructose and Post-Weaning High-Fat Diets. Int. J. Mol. Sci. 2018, 19, 1066. [Google Scholar] [CrossRef]
- Petakh, P.; Kamyshna, I.; Kamyshnyi, A. Effects of metformin on the gut microbiota: A systematic review. Mol. Metab. 2023, 77, 101805. [Google Scholar] [CrossRef]
- Kleniewska, P.; Pawliczak, R. The Link Between Dysbiosis, Inflammation, Oxidative Stress, and Asthma-The Role of Probiotics, Prebiotics, and Antioxidants. Nutrients 2024, 17, 16. [Google Scholar] [CrossRef]
- Mishra, V.; Shah, C.; Mokashe, N.; Chavan, R.; Yadav, H.; Prajapati, J. Probiotics as potential antioxidants: A systematic review. J. Agric. Food Chem. 2015, 63, 3615–3626. [Google Scholar] [CrossRef]
- Yuksel, S.; Yigit, A.A.; Cinar, M.; Atmaca, N.; Onaran, Y. Oxidant and Antioxidant Status of Human Breast Milk during Lactation Period. Dairy Sci. Technol. 2015, 95, 295–302. [Google Scholar] [CrossRef]
- Hinde, K.; Lewis, Z.T. MICROBIOTA. Mother’s littlest helpers. Science 2015, 348, 1427–1428. [Google Scholar] [CrossRef]
- Lyons, K.E.; Ryan, C.A.; Dempsey, E.M.; Ross, R.P.; Stanton, C. Breast Milk, a Source of Beneficial Microbes and Associated Benefits for Infant Health. Nutrients 2020, 12, 1039. [Google Scholar] [CrossRef]
- Andreas, N.J.; Kampmann, B.; Mehring Le-Doare, K. Human breast milk: A review on its composition and bioactivity. Early Hum. Dev. 2015, 91, 629–635. [Google Scholar] [CrossRef]
- Binns, C.; Lee, M.; Low, W.Y. The Long-Term Public Health Benefits of Breastfeeding. Asia Pac. J. Public Health 2016, 28, 7–14. [Google Scholar]
- Singhal, A.; Cole, T.J.; Lucas, A. Early Nutrition in Preterm Infants and Later Blood Pressure: Two Cohorts after Randomised Trials. Lancet 2001, 357, 413–419. [Google Scholar] [CrossRef]
- l-Khuffash, A.; Jain, A.; Lewandowski, A.J.; Levy, P.T. Preventing disease in the 21st century: Early breast milk exposure and later cardiovascular health in premature infants. Pediatr. Res. 2020, 87, 385–390. [Google Scholar] [CrossRef]
- Section on Breastfeeding. Breastfeeding and the use of human milk. Pediatrics 2012, 129, e827–e841. [Google Scholar] [CrossRef]
- Larson, K.; Russ, S.A.; Kahn, R.S.; Flores, G.; Goodman, E.; Cheng, T.L.; Halfon, N. Health Disparities: A Life Course Health Development Perspective and Future Research Directions. In Handbook of Life Course Health Development [Internet]; Halfon, N., Forrest, C.B., Lerner, R.M., Faustman, E.M., Eds.; Springer: Cham, Switzerland, 2018. [Google Scholar]
- Halliwell, B. The antioxidant paradox. Lancet 2000, 355, 1179–1180. [Google Scholar] [CrossRef]
- Hussain, T.; Murtaza, G.; Metwally, E.; Kalhoro, D.H.; Kalhoro, M.S.; Rahu, B.A.; Sahito, R.G.A.; Yin, Y.; Yang, H.; Chughtai, M.I.; et al. The Role of Oxidative Stress and Antioxidant Balance in Pregnancy. Mediat. Inflamm. 2021, 2021, 9962860. [Google Scholar] [CrossRef]
- Villanueva, C.; Kross, R.D. Antioxidant-induced stress. Int. J. Mol. Sci. 2012, 13, 2091–2109. [Google Scholar] [CrossRef]
Category | Examples | Main Mechanisms | Microbiota Modulation | Key Evidence | Translational Considerations |
---|---|---|---|---|---|
Vitamins and Micronutrients | Vitamin C, Vitamin E, Selenium, Folic Acid, Choline, Betaine | ROS scavenging, lipid peroxidation inhibition, methyl donor for epigenetic regulation | Limited direct reprogramming studies; known to influence microbiota composition | Prevented hypertension and CVD in offspring under maternal inflammation, nutrient restriction, hypercholesterolemia | Widely available; human data limited for developmental microbiota–vascular effects |
Amino Acids | Arginine, Citrulline, Taurine, Cysteine, Glycine, BCAAs, Tryptophan | NO production, H2S production, antioxidant defense enhancement | Alters hypertension-associated microbes; restores beneficial genera (Bifidobacterium, Oscillibacter); reduces harmful bacteria (Akkermansia, Alistipes) | Prevented hypertension in maternal CKD, diabetes, dexamethasone exposure; modulated AhR signaling | Requires understanding of dose/timing; maternal–fetal amino acid metabolism critical |
Melatonin | Endogenous hormone | ROS scavenging, ↑ antioxidant enzymes, ↑ NO, Nrf2 activation, ↓ inflammation | Restores microbial diversity; ↑ Firmicutes, Roseburia; ↓ TMAO dysregulation | Prevented hypertension/CVD in multiple DOHaD models; improved gut and vascular health | Safe in some neonatal uses; pregnancy safety uncertain; further long-term data needed |
Polyphenols | Quercetin, EGCG, Resveratrol | ROS scavenging, NO bioavailability ↑, metal chelation, endogenous antioxidant activation | ↑ Bifidobacterium, Lactobacillus, diversity; normalize Firmicutes/Bacteroidetes ratio | Resveratrol protected against hypertension in CKD, high-fructose, metabolic overload models | Poor bioavailability; interindividual absorption variability; targeted delivery strategies needed |
SCFAs | Acetate, Butyrate, Propionate | Antioxidant effects, SCFA receptor activation, RAS balance restoration, TMAO metabolism modulation | ↑ Roseburia, Bifidobacterium; enhance microbial diversity | Prevented hypertension in high-fructose, minocycline, tryptophan-deficient models | Potential as microbiota–metabolite therapy; diet-dependent production |
N-acetylcysteine (NAC) | NAC (from Allium plants) | Glutathione precursor, H2S synthesis, ROS reduction, ↑ NO bioavailability | ↑ Actinobacteria, Bifidobacterium, sulfur-oxidizing bacteria; ↑ fecal thiosulfate | Prevented hypertension in maternal suramin, SHR, CKD models | Dual antioxidant–microbiota actions; promising for oxidative-stress-driven hypertension |
Synthetic Antioxidants | Lazaroids, DMF (Nrf2 activator), Tempol, MitoQ | Lipid peroxidation inhibition, antioxidant gene activation, ADMA metabolism modulation | Limited data; some agents (metformin) modulate microbiota | DMF prevented hypertension in dexamethasone + high-fat model; metformin reduced ADMA and hypertension | Many agents already clinically available; need microbiota mechanism studies |
Others | Probiotics, Prebiotics, Postbiotics, Breast Milk | Antioxidant and anti-inflammatory effects, microbiome shaping | Direct microbiota delivery and modulation | Breastfeeding linked to lower long-term CVD risk; probiotics/prebiotics show benefit in animal models | Ethical barriers in pregnancy trials; breast milk offers natural antioxidant + probiotic synergy |
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. |
© 2025 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
Hsu, C.-N.; Lin, Y.-J.; Hou, C.-Y.; Chen, Y.-W.; Chang-Chien, G.-P.; Lin, S.-F.; Tain, Y.-L. Antioxidants, Gut Microbiota, and Cardiovascular Programming: Unraveling a Triad of Early-Life Interactions. Antioxidants 2025, 14, 1049. https://doi.org/10.3390/antiox14091049
Hsu C-N, Lin Y-J, Hou C-Y, Chen Y-W, Chang-Chien G-P, Lin S-F, Tain Y-L. Antioxidants, Gut Microbiota, and Cardiovascular Programming: Unraveling a Triad of Early-Life Interactions. Antioxidants. 2025; 14(9):1049. https://doi.org/10.3390/antiox14091049
Chicago/Turabian StyleHsu, Chien-Ning, Ying-Jui Lin, Chih-Yao Hou, Yu-Wei Chen, Guo-Ping Chang-Chien, Shu-Fen Lin, and You-Lin Tain. 2025. "Antioxidants, Gut Microbiota, and Cardiovascular Programming: Unraveling a Triad of Early-Life Interactions" Antioxidants 14, no. 9: 1049. https://doi.org/10.3390/antiox14091049
APA StyleHsu, C.-N., Lin, Y.-J., Hou, C.-Y., Chen, Y.-W., Chang-Chien, G.-P., Lin, S.-F., & Tain, Y.-L. (2025). Antioxidants, Gut Microbiota, and Cardiovascular Programming: Unraveling a Triad of Early-Life Interactions. Antioxidants, 14(9), 1049. https://doi.org/10.3390/antiox14091049