Dietary Impacts on Gestational Diabetes: Connection between Gut Microbiome and Epigenetic Mechanisms
Abstract
:1. Introduction
2. Gestational Diabetes Mellitus (GDM)
2.1. The Influence of GDM on Maternal Health and Pregnancy Outcomes
2.2. The influence of GDM on Fetal Development and Offspring Metabolic Outcomes
3. Epigenetic Reprogramming during Early Development
3.1. Epigenetic Regulation in GDM
3.1.1. DNA Methylation
3.1.2. Histone Modifications
3.1.3. Non-Coding RNAs (ncRNAs)
3.2. Epigenetics Bioactive Diets
3.3. Effects of Maternal Epigenetics Diets in GDM
4. Gut Microbiome, Metabolome and Diet
4.1. Establishment of Fetal Gut Microbiome
4.2. Gut Microbiota-Produced Metabolites through Maternal Diets
4.3. The Roles of Gut Microbiome and Metabolome in Development of GDM
5. Interplay between Maternal Diets, Gut Microbiome, Metabolome, and Epigenome on GDM Pathogenesis
5.1. Maternal Diets Alter Gut Microbiome and Metabolome
5.2. Crosstalk between Maternal Diets, Gut Microbiome, Metabolome and Epigenome in the Pathogenesis of Metabolic Disorders
6. Recent Clinical Trials through Maternal Diet in the Prevention of GDM
7. Conclusions
Author Contributions
Funding
Data Availability Statement
Conflicts of Interest
References
- Plows, J.F.; Stanley, J.L.; Baker, P.N.; Reynolds, C.M.; Vickers, M.H. The Pathophysiology of Gestational Diabetes Mellitus. Int. J. Mol. Sci. 2018, 19, 3342. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, H.; Li, N.; Chivese, T.; Werfalli, M.; Sun, H.; Yuen, L.; Hoegfeldt, C.A.; Powe, C.E.; Immanuel, J.; Karuranga, S.; et al. IDF Diabetes Atlas: Estimation of Global and Regional Gestational Diabetes Mellitus Prevalence for 2021 by International Association of Diabetes in Pregnancy Study Group’s Criteria. Diabetes Res. Clin. Pr. 2022, 183, 109050. [Google Scholar] [CrossRef] [PubMed]
- Yamada, H.; Hirayama Kato, E.; Tsuruga, R.; Ebina, Y.; Kobashi, G.; Sagawa, T.; Makita, Z.; Koike, T.; Fujimoto, S. Insulin Response Patterns Contribute to Different Perinatal Risks in Gestational Diabetes. Gynecol. Obstet. Investig. 2001, 51, 103–109. [Google Scholar] [CrossRef] [PubMed]
- Caughey, A.B.; Turrentine, M. ACOG Practice Bulletin No. 190 Summary: Gestational Diabetes Mellitus. Obstet. Gynecol. 2018, 131, 406–408. [Google Scholar]
- Alejandro, E.; Mamerto, T.; Chung, G.; Villavieja, A.; Gaus, N.; Morgan, E.; Pineda-Cortel, M. Gestational Diabetes Mellitus: A Harbinger of the Vicious Cycle of Diabetes. Int. J. Mol. Sci. 2020, 21, 5003. [Google Scholar] [CrossRef]
- Aplin, J.D.; Myers, J.E.; Timms, K.; Westwood, M. Tracking placental development in health and disease. Nat. Rev. Endocrinol. 2020, 16, 479–494. [Google Scholar] [CrossRef]
- Gómez-Roig, M.D.; Pascal, R.; Cahuana, M.J.; García-Algar, O.; Sebastiani, G.; Andreu-Fernández, V.; Martínez, L.; Rodríguez, G.; Iglesia, I.; Ortiz-Arrabal, O.; et al. Environmental Exposure during Preg-nancy: Influence on Prenatal Development and Early Life: A Comprehensive Review. Fetal Diagn Ther. 2021, 48, 245–257. [Google Scholar] [CrossRef]
- Chu, A.H.Y.; Godfrey, K.M. Gestational Diabetes Mellitus and Developmental Programming. Ann. Nutr. Metab. 2020, 76, 4–15. [Google Scholar] [CrossRef]
- Li, Y. Epigenetic Mechanisms Link Maternal Diets and Gut Microbiome to Obesity in the Offspring. Front. Genet. 2018, 9. [Google Scholar] [CrossRef]
- Kapur, K.; Kapur, A.; Hod, M. Nutrition Management of Gestational Diabetes Mellitus. Ann. Nutr. Metab. 2020, 76, 1–13. [Google Scholar] [CrossRef]
- Li, Y.; Saldanha, S.N.; Tollefsbol, T.O. Impact of Epigenetic Dietary Compounds on Transgenerational Prevention of Human Diseases. AAPS J. 2014, 16, 27–36. [Google Scholar] [CrossRef] [PubMed]
- Tiffon, C. The Impact of Nutrition and Environmental Epigenetics on Human Health and Disease. Int. J. Mol. Sci. 2018, 19, 3425. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, S.; Chen, M.; Li, Y.; Tollefsbol, T.O. Prenatal epigenetics diets play protective roles against environmental pollution. Clin. Epigenetics 2019, 11, 1–31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, M.; Li, S.; Arora, I.; Yi, N.; Sharma, M.; Li, Z.; Tollefsbol, T.O.; Li, Y. Maternal soybean diet on prevention of obesity-related breast cancer through early-life gut microbiome and epigenetic regulation. J. Nutr. Biochem. 2022, 110, 109119. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; Buckhaults, P.; Li, S.; Tollefsbol, T. Temporal Efficacy of a Sulforaphane-Based Broccoli Sprout Diet in Prevention of Breast Cancer through Modulation of Epigenetic Mechanisms. Cancer Prev. Res. 2018, 11, 451–464. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, M.; Li, S.; Srinivasasainagendra, V.; Sharma, M.; Li, Z.; Tiwari, H.; Tollefsbol, T.O.; Li, Y. Maternal soybean genistein on pre-vention of later-life breast cancer through inherited epigenetic regulations. Carcinogenesis. 2022, 43, 190–202. [Google Scholar] [CrossRef]
- Zhu, W.; Shen, Y.; Liu, J.; Fei, X.; Zhang, Z.; Li, M.; Chen, X.; Xu, J.; Zhu, Q.; Zhou, W.; et al. Epigenetic alternations of microRNAs and DNA methylation contribute to gestational diabetes mellitus. J. Cell. Mol. Med. 2020, 24, 13899–13912. [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]
- Ponzo, V.; Fedele, D.; Goitre, I.; Leone, F.; Lezo, A.; Monzeglio, C.; Finocchiaro, C.; Ghigo, E.; Bo, S. Diet-Gut Microbiota Interactions and Gestational Diabetes Mellitus (GDM). Nutrients 2019, 11, 330. [Google Scholar] [CrossRef] [Green Version]
- Ziętek, M.; Celewicz, Z.; Szczuko, M. Short-chain fatty acids, maternal microbiota, and metabolism in pregnancy. Nutrients 2021, 13, 1244. [Google Scholar] [CrossRef]
- Li, X.; Yu, D.; Wang, Y.; Yuan, H.; Ning, X.; Rui, B.; Lei, Z.; Yuan, J.; Yan, J.; Li, M. The Intestinal Dysbiosis of Mothers with Gestational Diabetes Mellitus (GDM) and Its Impact on the Gut Microbiota of Their Newborns. Can. J. Infect. Dis. Med. Microbiol. 2021, 2021, 1–12. [Google Scholar] [CrossRef] [PubMed]
- Verduci, E.; Giannì, M.; Vizzari, G.; Vizzuso, S.; Cerasani, J.; Mosca, F.; Zuccotti, G. The Triad Mother-Breast Milk-Infant as Predictor of Future Health: A Narrative Review. Nutrients 2021, 13, 486. [Google Scholar] [CrossRef] [PubMed]
- Khodayarpardo, P.; Pascual, L.M.; Collado, M.C.; Martinez-Costa, C. Impact of lactation stage, gestational age and mode of delivery on breast milk microbiota. J. Perinatol. 2014, 34, 599–605. [Google Scholar] [CrossRef] [PubMed]
- Nyangahu, D.D.; Lennard, K.S.; Brown, B.; Darby, M.; Wendoh, J.M.; Havyarimana, E.; Smith, P.; Butcher, J.; Stintzi, A.; Mulder, N.J.; et al. Disruption of maternal gut microbiota during gestation alters offspring microbiota and immunity. Microbiome 2018, 6, 1–10. [Google Scholar] [CrossRef] [Green Version]
- Li, P.; Zhong, C.; Li, S.; Sun, T.; Huang, H.; Chen, X.; Zhu, Y.; Hu, X.; Peng, X.; Zhang, X.; et al. Plasma concentration of trimethylamine-N-oxide and risk of gestational diabetes mellitus. Am. J. Clin. Nutr. 2018, 108, 603–610. [Google Scholar] [CrossRef] [Green Version]
- Latino, C.; Gianatti, E.J.; Mehta, S.; Lo, J.; Devine, A.; Christophersen, C. Does a high dietary intake of resistant starch affect glycaemic control and alter the gut microbiome in women with gestational diabetes? A randomised control trial protocol. BMC Pregnancy Childbirth 2022, 22, 1–13. [Google Scholar] [CrossRef]
- Hardy, T.M.; Tollefsbol, T.O. Epigenetic diet: Impact on the epigenome and cancer. Epigenomics 2011, 3, 503–518. [Google Scholar] [CrossRef] [Green Version]
- Belda, E.; Voland, L.; Tremaroli, V.; Falony, G.; Adriouch, S.; Assmann, K.E.; Prifti, E.; Aron-Wisnewsky, J.; Debédat, J.; Le Roy, T. Impairment of gut microbial biotin metabolism and host biotin status in severe obesity: Effect of biotin and prebiotic sup-plementation on improved metabolism. Gut 2022, 71, 2463–2480. [Google Scholar] [CrossRef]
- Krautkramer, K.A.; Kreznar, J.H.; Romano, K.A.; Vivas, E.I.; Barrett-Wilt, G.A.; Rabaglia, M.E.; Keller, M.P.; Attie, A.D.; Rey, F.E.; Denu, J.M. Diet-Microbiota Interactions Mediate Global Epigenetic Programming in Multiple Host Tissues. Mol. Cell 2016, 64, 982–992. [Google Scholar] [CrossRef] [Green Version]
- Brown, J.; Alwan, N.A.; West, J.; Brown, S.; McKinlay, C.J.; Farrar, D.; Crowther, C.A. Lifestyle interventions for the treatment of women with gestational diabetes. Cochrane Database Syst. Rev. 2017, 5, CD011970. [Google Scholar] [CrossRef] [Green Version]
- Mustad, V.A.; Huynh, D.T.; López-Pedrosa, J.M.; Campoy, C.; Rueda, R. The Role of Dietary Carbohydrates in Gestational Diabetes. Nutrients 2020, 12, 385. [Google Scholar] [CrossRef] [PubMed]
- Nguyen-Ngo, C.; Jayabalan, N.; Salomon, C.; Lappas, M. Molecular pathways disrupted by gestational diabetes mellitus. J. Mol. Endocrinol. 2019, 63, R51–R72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Djelmiš, J.; Desoye, G.; Ivaniševic, M. (Eds.) Diabetology of Pregnancy. In Frontiers in Diabetes; Karger: Basel, Switzerland, 2005; Volume 17, pp. 310–319. [Google Scholar]
- Franzago, M.; Fraticelli, F.; Stuppia, L.; Vitacolonna, E. Nutrigenetics, epigenetics and gestational diabetes: Consequences in mother and child. Epigenetics 2019, 14, 215–235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Catalano, P.M.; Ehrenberg, H.M. The short and long term implications of maternal obesity on the mother and her off-spring. British J. Obstet. Gynecology. 2006, 113, 1126–1133. [Google Scholar] [CrossRef] [PubMed]
- Choudhury, A.A.; Devi Rajeswari, V. Gestational diabetes mellitus—A metabolic and reproductive disorder. Biomed Pharmacother. 2021, 143, 112183. [Google Scholar] [CrossRef] [PubMed]
- Catalano, P.M.; Nizielski, S.E.; Shao, J.; Preston, L.; Qiao, L.; Friedman, J.E. Downregulated IRS-1 and PPARγ in obese women with gestational diabetes: Relationship to free fatty acids during pregnancy. Am. J. Physiol. Endocr. Metab. 2002, 282, E522–E533. [Google Scholar] [CrossRef] [Green Version]
- Stettler, C.; Christ, E.; Diem, P. (Eds.) Novelties in Diabetes. Endocr. Dev. 2016, 31, 163–178. [Google Scholar]
- Ehrenberg, H.M.; Mercer, B.M.; Catalano, P.M. The influence of obesity and diabetes on the prevalence of macrosomia. Am. J. Obstet. Gynecol. 2004, 191, 964–968. [Google Scholar] [CrossRef]
- Kc, K.; Shakya, S.; Zhang, H. Gestational Diabetes Mellitus and Macrosomia: A Literature Review. Ann. Nutr. Metab. 2015, 66, 14–20. [Google Scholar] [CrossRef]
- Wendland, E.M.; Torloni, M.R.; Falavigna, M.; Trujillo, J.; Dode, M.A.; Campos, M.A.; Duncan, B.B.; Schmidt, M.I. Gestational diabetes and pregnancy outcomes–a systematic review of the World Health Organization (WHO) and the International Association of Diabetes in Pregnancy Study Groups (IADPSG) di-agnostic criteria. BMC Pregnancy Childbirth 2012, 12, 23. [Google Scholar] [CrossRef] [Green Version]
- Su, W.-J.; Chen, Y.-L.; Huang, P.-Y.; Shi, X.-L.; Yan, F.-F.; Chen, Z.; Yan, B.; Song, H.-Q.; Lin, M.-Z.; Li, X.-J. Effects of Prepregnancy Body Mass Index, Weight Gain, and Gestational Diabetes Mellitus on Pregnancy Outcomes: A Population-Based Study in Xiamen, China, 2011. Ann. Nutr. Metab. 2019, 75, 31–38. [Google Scholar] [CrossRef] [PubMed]
- Nguyen, M.T.; Ouzounian, J.G. Evaluation and Management of Fetal Macrosomia. Obstet. Gynecol. Clin. North Am. 2021, 48, 387–399. [Google Scholar] [CrossRef] [PubMed]
- El Hajj, N.; Schneider, E.; Lehnen, H.; Haaf, T. Epigenetics and life-long consequences of an adverse nutritional and diabetic in-trauterine environment. Reproduction 2014, 148, R111–R120. [Google Scholar] [CrossRef]
- Sweeting, A.N.; Wong, J.; Appelblom, H.; Ross, G.P.; Kouru, H.; Williams, P.F.; Sairanen, M.; Hyett, J.A. A Novel Early Pregnancy Risk Prediction Model for Gestational Diabetes Mellitus. Fetal Diagn. Ther. 2019, 45, 76–84. [Google Scholar] [CrossRef]
- Pan, X.-F.; Huang, Y.; Li, X.; Wang, Y.; Ye, Y.; Chen, H.; Marklund, M.; Wen, Y.; Liu, Y.; Zeng, H.; et al. Circulating fatty acids and risk of gestational diabetes mellitus: Prospective analyses in China. Eur. J. Endocrinol. 2021, 185, 87–97. [Google Scholar] [CrossRef] [PubMed]
- Pippen, J.; Stetson, B.; Doherty, L.; Varner, M.W.; Casey, B.M.; Reddy, U.M.; Wapner, R.J.; Rouse, D.J.; Tita, A.T.N.; Thorp, J.M., Jr.; et al. Eunice Kennedy Shriver National Institute of child health human development maternal-fetal medicine units network. Neonatal birthweight, infant feeding, and childhood metabolic markers. Am. J. Perinatol. 2022, 39, 584–591. [Google Scholar] [PubMed]
- Mustaniemi, S.; Vääräsmäki, M.; Eriksson, J.G.; Gissler, M.; Laivuori, H.; Ijäs, H.; Bloigu, A.; Kajantie, E.; Morin-Papunen, L. Polycystic ovary syndrome and risk factors for gestational diabetes. Endocr. Connect. 2018, 7, 859–869. [Google Scholar] [CrossRef]
- Ruchat, S.-M.; Hivert, M.-F.; Bouchard, L. Epigenetic programming of obesity and diabetes by in utero exposure to gestational diabetes mellitus. Nutr. Rev. 2013, 71, S88–S94. [Google Scholar] [CrossRef]
- Silva-Zolezzi, I.; Samuel, T.M.; Spieldenner, J. Maternal nutrition: Opportunities in the prevention of gestational diabetes. Nutr. Rev. 2017, 75, 32–50. [Google Scholar] [CrossRef]
- Sweet, C.B.; Grayson, S.; Polak, M. Management Strategies for Neonatal Hypoglycemia. J. Pediatr. Pharmacol. Ther. 2013, 18, 199–208. [Google Scholar] [CrossRef] [Green Version]
- Brumbaugh, D.E.; Tearse, P.; Cree-Green, M.; Fenton, L.Z.; Brown, M.; Scherzinger, A.; Reynolds, R.; Alston, M.; Hoffman, C.; Pan, Z.; et al. Intrahepatic Fat Is Increased in the Neonatal Offspring of Obese Women with Gestational Diabetes. J. Pediatr. 2013, 162, 930–936. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lorenzo-Almorós, A.; Hang, T.; Peiró, C.; Soriano-Guillén, L.; Egido, J.; Tuñón, J.; Lorenzo, Ó. Predictive and diagnostic biomarkers for gestational diabetes and its associated metabolic and cardiovascular diseases. Cardiovasc. Diabetol. 2019, 18, 140. [Google Scholar] [CrossRef] [PubMed]
- Agarwal, P.; Morriseau, T.S.; Kereliuk, S.M.; Doucette, C.A.; Wicklow, B.A.; Dolinsky, V.W. Maternal obesity, diabetes during preg-nancy and epigenetic mechanisms that influence the developmental origins of cardiometabolic disease in the offspring. Crit. Rev. Clin. Lab. Sci. 2018, 55, 71–101. [Google Scholar] [CrossRef] [PubMed]
- Law, P.-P.; Holland, M.L. DNA methylation at the crossroads of gene and environment interactions. Essays Biochem. 2019, 63, 717–726. [Google Scholar] [CrossRef]
- Sun, C.; Fan, J.-G.; Qiao, L. Potential Epigenetic Mechanism in Non-Alcoholic Fatty Liver Disease. Int. J. Mol. Sci. 2015, 16, 5161–5179. [Google Scholar] [CrossRef] [Green Version]
- Deshpande, S.S.; Balasinor, N.H. Placental Defects: An Epigenetic Perspective. Reprod. Sci. 2018, 25, 1143–1160. [Google Scholar] [CrossRef]
- Nelissen, E.C.; van Montfoort, A.P.; Dumoulin, J.C.; Evers, J.L. Epigenetics and the placenta. Hum. Reprod. Updat. 2010, 17, 397–417. [Google Scholar] [CrossRef] [Green Version]
- Caniçais, C.; Vasconcelos, S.; Ramalho, C.; Marques, C.J.; Dória, S. Deregulation of imprinted genes expression and epigenetic regulators in placental tissue from intrauterine growth restriction. J. Assist. Reprod. Genet. 2021, 38, 791–801. [Google Scholar] [CrossRef]
- Meng, H.; Cao, Y.; Qin, J.; Song, X.; Zhang, Q.; Shi, Y.; Cao, L. DNA Methylation, Its Mediators and Genome Integrity. Int. J. Biol. Sci. 2015, 11, 604–617. [Google Scholar] [CrossRef] [Green Version]
- Moore, L.D.; Le, T.; Fan, G. DNA methylation and its basic function. Neuropsychopharmacology 2013, 38, 23–38. [Google Scholar] [CrossRef] [Green Version]
- Li, S.; Tollefsbol, T.O. DNA methylation methods: Global DNA methylation and methylomic analyses. Methods 2021, 187, 28–43. [Google Scholar] [CrossRef] [PubMed]
- Howe, C.G.; Cox, B.; Fore, R.; Jungius, J.; Kvist, T.; Lent, S.; Miles, H.E.; Salas, L.A.; Rifas-Shiman, S.; Starling, A.P.; et al. Maternal Gestational Diabetes Mellitus and Newborn DNA Methylation: Findings from the Pregnancy and Childhood Epigenetics Consortium. Diabetes Care 2020, 43, 98–105. [Google Scholar] [CrossRef] [PubMed]
- Nomura, Y.; Lambertini, L.; Rialdi, A.; Lee, M.; Ba, E.Y.M.; Grabie, M.; Bs, I.M.; Huynh, N.; Finik, J.; Davey, M.; et al. Global Methylation in the Placenta and Umbilical Cord Blood from Pregnancies With Maternal Gestational Diabetes, Preeclampsia, and Obesity. Reprod. Sci. 2014, 21, 131–137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ruchat, S.-M.; Houde, A.-A.; Voisin, G.; St-Pierre, J.; Perron, P.; Baillargeon, J.-P.; Gaudet, D.; Hivert, M.-F.; Brisson, D.; Bouchard, L. Gestational diabetes mellitus epigenetically affects genes predominantly involved in metabolic diseases. Epigenetics 2013, 8, 935–943. [Google Scholar] [CrossRef] [Green Version]
- Wang, L.; Fan, H.; Zhou, L.; Wu, Y.; Lu, H.; Luo, J. Altered expression of PGC-1 α and PDX1 and their methylation status are associated with fetal glucose metabolism in gestational diabetes mellitus. Biochem. Biophys. Res. Commun. 2018, 501, 300–306. [Google Scholar] [CrossRef]
- Barlow, D.P.; Bartolomei, M.S. Genomic imprinting in mammals. Cold Spring Harb Perspect Biol. 2014, 6, a018382. [Google Scholar] [CrossRef] [Green Version]
- Inbar-Feigenberg, M.; Choufani, S.; Butcher, D.T.; Roifman, M.; Weksberg, R. Basic concepts of epigenetics. Fertil. Steril. 2013, 99, 607–615. [Google Scholar] [CrossRef]
- Petry, C.; Mooslehner, K.; Prentice, P.; Hayes, M.; Nodzenski, M.; Scholtens, D.; Hughes, I.; Acerini, C.; Ong, K.; Lowe, W.; et al. Associations between a fetal imprinted gene allele score and late pregnancy maternal glucose concentrations. Diabetes Metab. 2017, 43, 323–331. [Google Scholar] [CrossRef]
- Zhang, Y.; Sun, Z.; Jia, J.; Du, T.; Zhang, N.; Tang, Y.; Fang, Y.; Fang, D. Overview of Histone Modification. Adv. Exp. Med. Biol. 2021, 1283, 1–16. [Google Scholar] [CrossRef]
- Zhou, B.-R.; Bai, Y. Chromatin structures condensed by linker histones. Essays Biochem. 2019, 63, 75–87. [Google Scholar] [CrossRef]
- Tolsma, T.O.; Hansen, J.C. Post-translational modifications and chromatin dynamics. Essays Biochem. 2019, 63, 89–96. [Google Scholar] [CrossRef] [PubMed]
- Ramazi, S.; Allahverdi, A.; Zahiri, J. Evaluation of post-translational modifications in histone proteins: A review on histone modification defects in developmental and neurological disorders. J. Biosci. 2020, 45, 33184251. [Google Scholar] [CrossRef]
- Li, Y.; Seto, E. HDACs and HDAC Inhibitors in Cancer Development and Therapy. Cold Spring Harb. Perspect. Med. 2016, 6, a026831. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schuettengruber, B.; Bourbon, H.-M.; Di Croce, L.; Cavalli, G. Genome Regulation by Polycomb and Trithorax: 70 Years and Counting. Cell 2017, 171, 34–57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hepp, P.; Hutter, S.; Knabl, J.; Hofmann, S.; Kuhn, C.; Mahner, S.; Jeschke, U. Histone H3 Lysine 9 Acetylation is Downregulated in GDM Placentas and Calcitriol Supplementation Enhanced This Effect. Int. J. Mol. Sci. 2018, 19, 4061. [Google Scholar] [CrossRef] [Green Version]
- Li, Y.; Li, G.; Guo, X.; Yao, H.; Wang, G.; Li, C. Non-coding RNA in bladder cancer. Cancer Lett. 2020, 485, 38–44. [Google Scholar] [CrossRef]
- Panni, S.; Lovering, R.C.; Porras, P.; Orchard, S. Non-coding RNA regulatory networks. Biochim. Biophys. Acta Gene Regul. Mech. 2020, 1863, 194417. [Google Scholar] [CrossRef]
- Li, Y. Modern epigenetics methods in biological research. Methods 2021, 187, 104–113. [Google Scholar] [CrossRef]
- Moen, G.H.; Sommer, C.; Prasad, R.B.; Sletner, L.; Groop, L.; Qvigstad, E.; Birkeland, K.I. Mechanisms in Endocrinology: Ep-igenetic modifications and gestational diabetes: A systematic review of published literature. Eur. J. Endocrinol. 2017, 176, R247–R267. [Google Scholar] [CrossRef] [Green Version]
- Ozata, D.M.; Gainetdinov, I.; Zoch, A.; O’Carroll, D.; Zamore, P.D. PIWI-interacting RNAs: Small RNAs with big functions. Nat. Rev. Genet. 2019, 20, 89–108. [Google Scholar] [CrossRef] [Green Version]
- Sliwinska, A.; Kasinska, M.A.; Drzewoski, J. MicroRNAs and metabolic disorders - where are we heading? Arch. Med. Sci. 2017, 13, 885–896. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, Z.-N.; Jiang, Y.; Liu, X.-Q.; Yang, M.-M.; Chen, C.; Zhao, B.-H.; Huang, H.-F.; Luo, Q. MiRNAs in Gestational Diabetes Mellitus: Potential Mechanisms and Clinical Applications. J. Diabetes Res. 2021, 2021, 1–14. [Google Scholar] [CrossRef] [PubMed]
- Dias, S.; Pheiffer, C.; Abrahams, Y.; Rheeder, P.; Adam, S. Molecular Biomarkers for Gestational Diabetes Mellitus. Int. J. Mol. Sci. 2018, 19, 2926. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, W.; Cao, D.; Wang, Y.; Ren, W. LncRNA MEG8 is upregulated in gestational diabetes mellitus (GDM) and predicted kidney injury. J. Diabetes its Complicat. 2021, 35, 107749. [Google Scholar] [CrossRef] [PubMed]
- Li, J.; Du, B.; Geng, X.; Zhou, L. lncRNA SNHG17 is Downregulated in Gestational Diabetes Mellitus (GDM) and Has Predictive Values. Diabetes Metab. Syndr. Obes. Targets Ther. 2021, ume 14, 831–838. [Google Scholar] [CrossRef]
- Mentch, S.J.; Locasale, J.W. One-carbon metabolism and epigenetics: Understanding the specificity. Ann. N. Y. Acad. Sci. 2016, 1363, 91–98. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zeisel, S.H. Choline: Critical Role During Fetal Development and Dietary Requirements in Adults. Annu. Rev. Nutr. 2006, 26, 229–250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bernhard, W.; Poets, C.F.; Franz, A.R. Choline and choline-related nutrients in regular and preterm infant growth. Eur. J. Nutr. 2019, 58, 931–945. [Google Scholar] [CrossRef] [PubMed]
- Santangelo, C.; Zicari, A.; Mandosi, E.; Scazzocchio, B.; Mari, E.; Morano, S.; Masella, R. Could gestational diabetes mellitus be managed through dietary bioactive compounds? Current knowledge and future perspectives. Br. J. Nutr. 2016, 115, 1129–1144. [Google Scholar] [CrossRef]
- Khan, M.A.; Hussain, A.; Sundaram, M.K.; Alalami, U.; Gunasekera, D.; Ramesh, L.; Hamza, A.; Quraishi, U. (−)-Epigallocatechin-3-gallate reverses the expression of various tumor-suppressor genes by inhibiting DNA methyltransferases and histone deacetylases in human cervical cancer cells. Oncol. Rep. 2015, 33, 1976–1984. [Google Scholar] [CrossRef] [Green Version]
- Rothenberg, D.O.; Zhou, C.; Zhang, L. A Review on the Weight-Loss Effects of Oxidized Tea Polyphenols. Molecules 2018, 23, 1176. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Heber, D.; Zhang, Y.; Yang, J.; Ma, J.E.; Henning, S.M.; Li, Z. Green Tea, Black Tea, and Oolong Tea Polyphenols Reduce Visceral Fat and Inflammation in Mice Fed High-Fat, High-Sucrose Obesogenic Diets. J. Nutr. 2014, 144, 1385–1393. [Google Scholar] [CrossRef] [PubMed]
- Sharma, M.; Arora, I.; Chen, M.; Wu, H.; Crowley, M.R.; Tollefsbol, T.O.; Li, Y. Therapeutic Effects of Dietary Soybean Genistein on Triple-Negative Breast Cancer via Regulation of Epigenetic Mechanisms. Nutrients 2021, 13, 3944. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; Liu, L.; Andrews, L.G.; Tollefsbol, T.O. Genistein depletes telomerase activity through cross-talk between genetic and epi-genetic mechanisms. Int. J. Cancer 2009, 125, 286–296. [Google Scholar] [CrossRef] [Green Version]
- Li, Y.; Liu, L.; Tollefsbol, T.O. Glucose restriction can extend normal cell lifespan and impair precancerous cell growth through epigenetic control of hTERT and p16 expression. FASEB J. 2010, 24, 1442–1453. [Google Scholar] [CrossRef] [Green Version]
- Li, Y.; Meeran, S.M.; Patel, S.N.; Chen, H.; Hardy, T.M.; Tollefsbol, T.O. Epigenetic reactivation of estrogen receptor-α (ERα) by genistein enhances hormonal therapy sensitivity in ERα-negative breast cancer. Mol. Cancer 2013, 12, 9. [Google Scholar] [CrossRef] [Green Version]
- Choi, J.S.; Koh, I.-U.; Song, J. Genistein reduced insulin resistance index through modulating lipid metabolism in ovariectomized rats. Nutr. Res. 2012, 32, 844–855. [Google Scholar] [CrossRef]
- Behloul, N.; Wu, G. Genistein: A promising therapeutic agent for obesity and diabetes treatment. Eur. J. Pharmacol. 2013, 698, 31–38. [Google Scholar] [CrossRef]
- Gilbert, E.R.; Liu, D. Anti-diabetic functions of soy isoflavone genistein: Mechanisms underlying its effects on pancreatic β-cell function. Food Funct. 2013, 4, 200–212. [Google Scholar] [CrossRef] [Green Version]
- Sakamoto, Y.; Naka, A.; Ohara, N.; Kondo, K.; Iida, K. Daidzein regulates proinflammatory adipokines thereby improving obe-sity-related inflammation through PPARγ. Mol. Nutr. Food Res. 2014, 58, 718–726. [Google Scholar] [CrossRef] [PubMed]
- Vanduchova, A.; Anzenbacher, P.; Anzenbacherova, E. Isothiocyanate from Broccoli, Sulforaphane, and Its Properties. J. Med. Food. 2019, 22, 121–126. [Google Scholar] [CrossRef] [PubMed]
- Gao, L.; Cheng, D.; Yang, J.; Wu, R.; Li, W.; Kong, A.-N. Sulforaphane epigenetically demethylates the CpG sites of the miR-9-3 promoter and reactivates miR-9-3 expression in human lung cancer A549 cells. J. Nutr. Biochem. 2018, 56, 109–115. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.; Fu, X.; Chen, Z.; Luo, T.; Zhu, C.; Ji, Y.; Bian, Z. The Protective Effects of Sulforaphane on High-Fat Diet-Induced Obesity in Mice Through Browning of White Fat. Front. Pharmacol. 2021, 12. [Google Scholar] [CrossRef] [PubMed]
- Li, Z.; Li, Y. Bioactive Dietary Compounds and Epigenetics in Women’s Reproductive Cancers. Compr. Pharmacol. 2021, 595–610. [Google Scholar] [CrossRef]
- Sharma, M.; Tollefsbol, T.O. Combinatorial epigenetic mechanisms of sulforaphane, genistein and sodium butyrate in breast cancer inhibition. Exp. Cell Res. 2022, 416. [Google Scholar] [CrossRef]
- Galiniak, S.; Aebisher, D.; Bartusik-Aebisher, D. Health benefits of resveratrol administration. Acta Biochim. Pol. 2019, 66, 13–21. [Google Scholar] [CrossRef] [Green Version]
- Sedlak, L.; Wojnar, W.; Zych, M.; Wyględowska-Promieńska, D.; Mrukwa-Kominek, E.; Kaczmarczyk-Sedlak, I. Effect of Resveratrol, a Dietary-Derived Polyphenol, on the Oxidative Stress and Polyol Pathway in the Lens of Rats with Streptozotocin-Induced Diabetes. Nutrients 2018, 10, 1423. [Google Scholar] [CrossRef] [Green Version]
- Udenigwe, C.; Ramprasath, V.R.; Aluko, R.E.; Jones, P.J.H. Potential of resveratrol in anticancer and anti-inflammatory therapy. Nutr. Rev. 2008, 66, 445–454. [Google Scholar] [CrossRef]
- Kim, C.W.; Hwang, K.A.; Choi, K.C. Anti-metastatic potential of resveratrol and its metabolites by the inhibition of epitheli-al-mesenchymal transition, migration, and invasion of malignant cancer cells. Phytomedicine 2016, 23, 1787–1796. [Google Scholar] [CrossRef]
- Divella, R.; Daniele, A.; Savino, E.; Paradiso, A. Anticancer Effects of Nutraceuticals in the Mediterranean Diet: An Epigenetic Diet Model. Cancer Genom. Proteom. 2020, 17, 335–350. [Google Scholar] [CrossRef]
- Shimron-Nachmias, L.; Frishman, S.; Hod, M. Dietary management of diabetic pregnancy. Harefuah 2006, 145, 768–780. [Google Scholar] [PubMed]
- Chen, X.; Zhang, Y.; Chen, H.; Jiang, Y.; Wang, Y.; Wang, D.; Li, M.; Dou, Y.; Sun, X.; Huang, G.; et al. Association of Maternal Folate and Vitamin B12 in Early Pregnancy with Gestational Diabetes Mellitus: A Prospective Cohort Study. Diabetes Care 2021, 44, 217–223. [Google Scholar] [CrossRef] [PubMed]
- Setola, E.; Monti, L.; Galluccio, E.; Palloshi, A.; Fragasso, G.; Paroni, R.; Magni, F.; Sandoli, E.; Lucotti, P.; Costa, S.; et al. Insulin resistance and endothelial function are improved after folate and vitamin B12 therapy in patients with metabolic syndrome: Relationship between homocysteine levels and hyperinsulinemia. Eur. J. Endocrinol. 2004, 151, 483–489. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nam, J.; Greenwald, E.; Jack-Roberts, C.; Ajeeb, T.T.; Malysheva, O.V.; Caudill, M.A.; Axen, K.; Saxena, A.; Semernina, E.; Nanobashvili, K.; et al. Choline prevents fetal overgrowth and normalizes placental fatty acid and glucose metabolism in a mouse model of maternal obesity. J. Nutr. Biochem. 2017, 49, 80–88. [Google Scholar] [CrossRef] [PubMed]
- Esmaeelpanah, E.; Razavi, B.M.; Hosseinzadeh, H. Green tea and metabolic syndrome: A 10-year research update review. Iran J. Basic Med. Sci. 2021, 24, 1159–1172. [Google Scholar] [CrossRef]
- Zhang, H.; Su, S.; Yu, X.; Li, Y. Dietary epigallocatechin 3-gallate supplement improves maternal and neonatal treatment outcome of gestational diabetes mellitus: A double-blind randomised controlled trial. J. Hum. Nutr. Diet. 2017, 30, 753–758. [Google Scholar] [CrossRef]
- Yuan, L.-J.; Qin, Y.; Wang, L.; Zeng, Y.; Chang, H.; Wang, J.; Wang, B.; Wan, J.; Chen, S.-H.; Zhang, Q.-Y.; et al. Capsaicin-containing chili improved postprandial hyperglycemia, hyperinsulinemia, and fasting lipid disorders in women with gestational diabetes mellitus and lowered the incidence of large-for-gestational-age newborns. Clin. Nutr. 2016, 35, 388–393. [Google Scholar] [CrossRef]
- Jamilian, M.; Asemi, Z. The Effect of Soy Intake on Metabolic Profiles of Women with Gestational Diabetes Mellitus. J. Clin. Endocrinol. Metab. 2015, 100, 4654–4661. [Google Scholar] [CrossRef]
- Lu, X.; Wu, F.; Jiang, M.; Sun, X.; Tian, G. Curcumin ameliorates gestational diabetes in mice partly through activating AMPK. Pharm. Biol. 2019, 57, 250–254. [Google Scholar] [CrossRef] [Green Version]
- Hajimoosayi, F.; Jahanian Sadatmahalleh, S.; Kazemnejad, A.; Pirjani, R. Effect of ginger on the blood glucose level of women with gestational diabetes mellitus (GDM) with impaired glucose tolerance test (GTT): A randomized double-blind place-bo-controlled trial. BMC Complement Med. Ther. 2020, 20, 116. [Google Scholar] [CrossRef]
- Zhang, G.; Wang, X.; Ren, B.; Zhao, Q.; Zhang, F. The Effect of Resveratrol on Blood Glucose and Blood Lipids in Rats with Gesta-tional Diabetes Mellitus. Evid. Based Complement Alternat. Med. 2021, 2021, 2956795. [Google Scholar] [CrossRef] [PubMed]
- Mahabady, M.K.; Shamsi, M.M.; Ranjbar, R.; Tabandeh, M.R.; Khazaeel, K. Quercetin improved histological structure and upregulated adiponectin and adiponectin receptors in the placenta of rats with gestational diabetes mellitus. Placenta 2021, 106, 49–57. [Google Scholar] [CrossRef] [PubMed]
- Faroughi, F.; Charandabi, S.M.; Javadzadeh, Y.; Mirghafourvand, M. Effects of Garlic Pill on Blood Glucose Level in Borderline Gestational Diabetes Mellitus: A Triple Blind, Randomized Clinical Trial. Iran. Red Crescent Med. J. 2018, 20. in press. [Google Scholar] [CrossRef]
- 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] [Green Version]
- Lamichhane, S.; Sen, P.; Dickens, A.M.; Orešič, M.; Bertram, H.C. Gut metabolome meets microbiome: A methodological perspective to understand the relationship between host and microbe. Methods 2018, 149, 3–12. [Google Scholar] [CrossRef]
- Wilmanski, T.; Rappaport, N.; Earls, J.C.; Magis, A.T.; Manor, O.; Lovejoy, J.; Omenn, G.S.; Hood, L.; Gibbons, S.M.; Price, N.D. Blood metabolome predicts gut microbiome α-diversity in humans. Nat. Biotechnol. 2019, 37, 1217–1228. [Google Scholar] [CrossRef]
- Kato, T.; Yamazaki, K.; Nakajima, M.; Date, Y.; Kikuchi, J.; Hase, K.; Ohno, H.; Yamazaki, K. Oral Administration of Porphyromonas gingivalis Alters the Gut Microbiome and Serum Metabolome. Msphere 2018, 3, e00460-18. [Google Scholar] [CrossRef] [Green Version]
- 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] [Green Version]
- Bittinger, K.; Zhao, C.; Li, Y.; Ford, E.; Friedman, E.S.; Ni, J.; Kulkarni, C.V.; Cai, J.; Tian, Y.; Liu, Q. Bacterial colonization reprograms the neonatal gut metabolome. Nat. Microbiol. 2020, 5, 838–847. [Google Scholar] [CrossRef]
- Friesen, R.W.; Novak, E.M.; Hasman, D.; Innis, S.M. Relationship of Dimethylglycine, Choline, and Betaine with Oxoproline in Plasma of Pregnant Women and Their Newborn Infants. J. Nutr. 2007, 137, 2641–2646. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gong, X.; Du, Y.; Li, X.; Yang, J.; Zhang, X.; Wei, Y.; Zhao, Y. Maternal Plasma Betaine in Middle Pregnancy Was Associated with Decreased Risk of GDM in Twin Pregnancy: A Cohort Study. Diabetes Metab. Syndr. Obesity Targets Ther. 2021, ume 14, 2495–2504. [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. [Google Scholar] [CrossRef]
- Mayo, B.; Vázquez, L.; Flórez, A.B. Equol: A Bacterial Metabolite from The Daidzein Isoflavone and Its Presumed Beneficial Health Effects. Nutrients 2019, 11, 2231. [Google Scholar] [CrossRef] [Green Version]
- Qiao, Y.; Sun, J.; Xia, S.; Tang, X.; Shi, Y.; Le, G. Effects of resveratrol on gut microbiota and fat storage in a mouse model with high-fat-induced obesity. Food Funct. 2014, 5, 1241–1249. [Google Scholar] [CrossRef]
- Chaplin, A.; Carpéné, C.; Mercader, J. Resveratrol, Metabolic Syndrome, and Gut Microbiota. Nutrients 2018, 10, 1651. [Google Scholar] [CrossRef] [Green Version]
- Hasain, Z.; Mokhtar, N.M.; Kamaruddin, N.A.; Ismail, N.A.M.; Razalli, N.H.; Gnanou, J.V.; Ali, R.A.R. Gut Microbiota and Gestational Diabetes Mellitus: A Review of Host-Gut Microbiota Interactions and Their Therapeutic Potential. Front. Cell. Infect. Microbiol. 2020, 10, 188. [Google Scholar] [CrossRef]
- Agus, A.; Clément, K.; Sokol, H. Gut microbiota-derived metabolites as central regulators in metabolic disorders. Gut 2021, 70, 1174–1182. [Google Scholar] [CrossRef]
- Gao, Y.; Chen, H.; Li, J.; Ren, S.; Yang, Z.; Zhou, Y.; Xuan, R. Alterations of gut microbiota-derived metabolites in gestational diabetes mellitus and clinical significance. J. Clin. Lab. Anal. 2022, 36. [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, 1–12. [Google Scholar] [CrossRef] [Green Version]
- Crusell, M.K.; Hansen, T.H.; Nielsen, T.; Allin, K.H.; Rühlemann, M.C.; Damm, P.; Vestergaard, H.; Rørbye, C.; Jørgensen, N.R.; Christiansen, O.B.; et al. Gestational diabetes is associated with change in the gut microbiota composition in third trimester of pregnancy and postpartum. Microbiome 2018, 6, 1–9. [Google Scholar] [CrossRef]
- Wang, X.; Liu, H.; Li, Y.; Huang, S.; Zhang, L.; Cao, C.; Baker, P.N.; Tong, C.; Zheng, P.; Qi, H. Altered gut bacterial and metabolic signatures and their interaction in gestational diabetes mellitus. Gut Microbes 2020, 12, 1–13. [Google Scholar] [CrossRef] [PubMed]
- Bentley-Lewis, R.; Huynh, J.; Xiong, G.; Lee, H.; Wenger, J.; Clish, C.; Nathan, D.; Thadhani, R.; Gerszten, R. Metabolomic profiling in the prediction of gestational diabetes mellitus. Diabetologia 2015, 58, 1329–1332. [Google Scholar] [CrossRef] [PubMed]
- Hou, W.; Meng, X.; Zhao, A.; Zhao, W.; Pan, J.; Tang, J.; Huang, Y.; Li, H.; Jia, W.; Liu, F.; et al. Development of Multimarker Diagnostic Models from Metabolomics Analysis for Gestational Diabetes Mellitus (GDM). Mol. Cell. Proteom. 2018, 17, 431–441. [Google Scholar] [CrossRef] [Green Version]
- Su, M.; Nie, Y.; Shao, R.; Duan, S.; Jiang, Y.; Wang, M.; Xing, Z.; Sun, Q.; Liu, X.; Xu, W. Diversified gut microbiota in newborns of mothers with gestational diabetes mellitus. PLOS ONE 2018, 13, e0205695. [Google Scholar] [CrossRef]
- Chen, T.; Qin, Y.; Chen, M.; Zhang, Y.; Wang, X.; Dong, T.; Chen, G.; Sun, X.; Lu, T.; Xia, Y. Gestational diabetes mellitus is associated with the neonatal gut microbiota and metabolome. BMC Med. 2021, 19, 120. [Google Scholar] [CrossRef] [PubMed]
- Sierra, A.C.; Ramos-Lopez, O.; Riezu-Boj, J.I.; Milagro, F.I.; Martinez, J.A. Diet, Gut Microbiota, and Obesity: Links with Host Genetics and Epigenetics and Potential Applications. Adv. Nutr. 2019, 10, S17–S30. [Google Scholar] [CrossRef] [Green Version]
- Sharma, M.; Li, Y.; Stoll, M.L.; Tollefsbol, T.O. The Epigenetic Connection Between the Gut Microbiome in Obesity and Diabetes. Front. Genet. 2020, 10, 1329. [Google Scholar] [CrossRef] [Green Version]
- Myatt, L.; Maloyan, A. Obesity and Placental Function. Semin. Reprod. Med. 2016, 34, 42–49. [Google Scholar] [CrossRef]
- Gibson, G.R.; Roberfroid, M.B. Dietary Modulation of the Human Colonic Microbiota: Introducing the Concept of Prebiotics. J. Nutr. 1995, 125, 1401–1412. [Google Scholar] [CrossRef]
- Paul, H.A.; Bomhof, M.R.; Vogel, H.J.; Reimer, R.A. Diet-induced changes in maternal gut microbiota and metabolomic profiles influence programming of offspring obesity risk in rats. Sci. Rep. 2016, 6, 20683. [Google Scholar] [CrossRef] [Green Version]
- Zhang, C.; He, X.; Sheng, Y.; Yang, C.; Xu, J.; Zheng, S.; Liu, J.; Xu, W.; Luo, Y.; Huang, K. Allicin-induced host-gut microbe interactions improves energy homeostasis. FASEB J. 2020, 34, 10682–10698. [Google Scholar] [CrossRef] [PubMed]
- Sharma, M.; Arora, I.; Stoll, M.L.; Li, Y.; Morrow, C.D.; Barnes, S.; Berryhill, T.F.; Li, S.; Tollefsbol, T.O. Nutritional combinatorial impact on the gut microbiota and plasma short-chain fatty acids levels in the prevention of mammary cancer in Her2/neu estrogen receptor-negative transgenic mice. PLoS ONE 2020, 15, e0234893. [Google Scholar] [CrossRef] [PubMed]
- Hsu, C.N.; Hou, C.Y.; Lee, C.T.; Chan, J.Y.; Tain, Y.L. The interplay between maternal and post-weaning high-fat diet and gut microbiota in the developmental programming of hypertension. Nutrients 2019, 11, 1982. [Google Scholar] [CrossRef] [Green Version]
- Li, S.; You, J.; Wang, Z.; Liu, Y.; Wang, B.; Du, M.; Zou, T. Curcumin alleviates high-fat diet-induced hepatic steatosis and obesity in association with modulation of gut microbiota in mice. Food Res. Int. 2021, 143, 110270. [Google Scholar] [CrossRef]
- He, C.; Huang, L.; Lei, P.; Liu, X.; Li, B.; Shan, Y. Sulforaphane Normalizes Intestinal Flora and Enhances Gut Barrier in Mice with BBN-Induced Bladder Cancer. Mol. Nutr. Food Res. 2018, 62, e1800427. [Google Scholar] [CrossRef]
- Okubo, T.; Ishihara, N.; Oura, A.; Serit, M.; Kim, M.; Yamamoto, T.; Mitsuoka, T. In Vivo Effects of Tea Polyphenol Intake on Human Intestinal Microflora and Metabolism. Biosci. Biotechnol. Biochem. 1992, 56, 588–591. [Google Scholar] [CrossRef]
- Lee, H.C.; Jenner, A.M.; Low, C.S.; Lee, Y.K. Effect of tea phenolics and their aromatic fecal bacterial metabolites on intestinal mi-crobiota. Res. Microbiol. 2006, 157, 876–884. [Google Scholar] [CrossRef]
- Hu, W.; Huang, L.; Zhou, Z.; Yin, L.; Tang, J. Diallyl Disulfide (DADS) Ameliorates Intestinal Candida albicans Infection by Modu-lating the Gut microbiota and Metabolites and Providing Intestinal Protection in Mice. Front. Cell Infect. Microbiol. 2022, 11, 743454. [Google Scholar] [CrossRef]
- Huang, H.; Krishnan, H.B.; Pham, Q.; Yu, L.L.; Wang, T.T.Y. Soy and Gut Microbiota: Interaction and Implication for Human Health. J. Agric. Food Chem. 2016, 64, 8695–8709. [Google Scholar] [CrossRef]
- Tamura, K.; Sasaki, H.; Shiga, K.; Miyakawa, H.; Shibata, S. The Timing Effects of Soy Protein Intake on Mice Gut Microbiota. Nutrients 2019, 12, 87. [Google Scholar] [CrossRef] [Green Version]
- Gill, I.R.; Uno, J.K. The impact of dietary soy on Gut microbiome. FASEB 2016, 30 (suppl. 1). [Google Scholar]
- Zhu, Y.; Shi, X.; Lin, X.; Ye, K.; Xu, X.; Li, C.; Zhou, G. Beef, Chicken, and Soy Proteins in Diets Induce Different Gut Microbiota and Metabolites in Rats. Front. Microbiol. 2017, 8, 1395. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dioletis, E.; Paiva, R.S.; Kaffe, E.; Secor, E.R.; Weiss, T.R.; Fields, M.R.; Ouyang, X.; Ali, A. The fermented soy beverage Q-CAN® plus induces beneficial changes in the oral and intestinal microbiome. BMC Nutr. 2021, 7, 1–15. [Google Scholar] [CrossRef] [PubMed]
- Etxeberria, U.; Arias, N.; Boqué, N.; Macarulla, M.; Portillo, M.; Martínez, J.; Milagro, F. Reshaping faecal gut microbiota composition by the intake of trans-resveratrol and quercetin in high-fat sucrose diet-fed rats. J. Nutr. Biochem. 2015, 26, 651–660. [Google Scholar] [CrossRef]
- Zhang, B.; Xu, Y.; Lv, H.; Pang, W.; Wang, J.; Ma, H.; Wang, S. Intestinal pharmacokinetics of resveratrol and regulatory effects of resveratrol metabolites on gut barrier and gut microbiota. Food Chem. 2021, 357, 129532. [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 pro-grammed by maternal chronic kidney disease in adult male offspring: Implications of the gut microbiome and their me-tabolites. Biomedicines 2020, 8, 567. [Google Scholar] [CrossRef]
- Shi, T.; Bian, X.; Yao, Z.; Wang, Y.; Gao, W.; Guo, C. Quercetin improves gut dysbiosis in antibiotic-treated mice. Food Funct. 2020, 11, 8003–8013. [Google Scholar] [CrossRef]
- Porras, D.; Nistal, E.; Martínez-Flórez, S.; Pisonero-Vaquero, S.; Olcoz, J.L.; Jover, R.; González-Gallego, J.; García-Mediavilla, M.V.; Sánchez-Campos, S. Protective effect of quercetin on high-fat diet-induced non-alcoholic fatty liver disease in mice is mediated by modulating intestinal microbiota imbalance and related gut-liver axis activation. Free. Radic. Biol. Med. 2017, 102, 188–202. [Google Scholar] [CrossRef]
- Shinohara, K.; Ohashi, Y.; Kawasumi, K.; Terada, A.; Fujisawa, T. Effect of apple intake on fecal microbiota and metabolites in humans. Anaerobe 2010, 16, 510–515. [Google Scholar] [CrossRef]
- Hu, H.; Zhang, S.; Liu, F.; Zhang, P.; Muhammad, Z.; Pan, S. Role of the Gut Microbiota and Their Metabolites in Modulating the Cholesterol-Lowering Effects of Citrus Pectin Oligosaccharides in C57BL/6 Mice. J. Agric. Food Chem. 2019, 67, 11922–11930. [Google Scholar] [CrossRef]
- Li, W.; Zhang, K.; Yang, H. Pectin Alleviates High Fat (Lard) Diet-Induced Nonalcoholic Fatty Liver Disease in Mice: Possible Role of Short-Chain Fatty Acids and Gut Microbiota Regulated by Pectin. J. Agric. Food Chem. 2018, 66, 8015–8025. [Google Scholar] [CrossRef] [PubMed]
- Marounek, M.; Volek, Z.; Synytsya, A.; Čopíková, J. Effect of pectin and amidated pectin on cholesterol homeostasis and cecal metabolism in rats fed a high-cholesterol diet. Physiol. Res. 2007, 433–442. [Google Scholar] [CrossRef] [PubMed]
- Rodriguez-Melendez, R.; Zempleni, J. Regulation of gene expression by biotin (review). J. Nutr. Biochem. 2003, 14, 680–690. [Google Scholar] [CrossRef]
- Dakshinamurti, K. Biotin--a regulator of gene expression. J. Nutr. Biochem. 2005, 16, 419–423. [Google Scholar] [CrossRef] [PubMed]
- Berni Canani, R.; Di Costanzo, M.; Leone, L. The epigenetic effects of butyrate: Potential therapeutic implications for clinical practice. Clin. Epigenetics 2012, 4, 4. [Google Scholar] [CrossRef] [PubMed]
- Bishop, K.S.; Xu, H.; Marlow, G. Epigenetic Regulation of Gene Expression Induced by Butyrate in Colorectal Cancer: Involvement of MicroRNA. Genet. Epigenetics 2017, 9. [Google Scholar] [CrossRef]
- Romano, K.A.; Martinez-Del Campo, A.; Kasahara, K.; Chittim, C.L.; Vivas, E.I.; Amador-Noguez, D.; Balskus, E.P.; Rey, F.E. Metabolic, Epigenetic, and Transgenerational Effects of Gut Bacterial Choline Consumption. Cell Host Microbe 2017, 22, 279–290. [Google Scholar] [CrossRef] [Green Version]
- Romano, K.A.; Rey, F.E. Is maternal microbial metabolism an early-life determinant of health? Lab. Anim. 2018, 47, 239–243. [Google Scholar] [CrossRef]
- Ortega Ávila, J.G.; Echeverri, I.; de Plata, C.A.; Castillo, A. Impact of oxidative stress during pregnancy on fetal epigenetic patterns and early origin of vascular diseases. Nutr. Rev. 2015, 73, 12–21. [Google Scholar] [CrossRef] [PubMed]
- Amirani, E.; Asemi, Z.; Taghizadeh, M. The effects of selenium plus probiotics supplementation on glycemic status and serum lipoproteins in patients with gestational diabetes mellitus: A randomized, double-blind, placebo-controlled trial. Clin. Nutr. ESPEN 2022, 48, 56–62. [Google Scholar] [CrossRef]
- Karamali, M.; Dadkhah, F.; Sadrkhanlou, M.; Jamilian, M.; Ahmadi, S.; Tajabadi-Ebrahimi, M.; Jafari, P.; Asemi, Z. Effects of probiotic supplementation on glycaemic control and lipid profiles in gestational diabetes: A randomized, double-blind, placebo-controlled trial. Diabetes Metab. 2016, 42, 234–241. [Google Scholar] [CrossRef] [PubMed]
- Assaf-Balut, C.; de la Torre, N.G.; Fuentes, M.; Durán, A.; Bordiú, E.; del Valle, L.; Valerio, J.; Jiménez, I.; Herraiz, M.A.; Izquierdo, N.; et al. A High Adherence to Six Food Targets of the Mediterranean Diet in the Late First Trimester is Associated with a Reduction in the Risk of Materno-Foetal Outcomes: The St. Carlos Gestational Diabetes Mellitus Prevention Study. Nutrients 2018, 11, 66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Assaf-Balut, C.; Garcia De La Torre, N.; Durán, A.; Fuentes, M.; Bordiú, E.; Del Valle, L.; Familiar, C.; Ortolá, A.; Jiménez, I.; Herraiz, M.A.; et al. A Mediterranean diet with additional extra virgin olive oil and pistachios reduces the incidence of gestational diabetes mellitus (GDM): A randomized controlled trial: The St. Carlos GDM prevention study. PLoS ONE 2017, 12, e0185873. [Google Scholar] [CrossRef] [PubMed]
- Basu, A.; Feng, D.; Planinic, P.; Ebersole, J.L.; Lyons, T.J.; Alexander, J.M. Dietary Blueberry and Soluble Fiber Supplementation Reduces Risk of Gestational Diabetes in Women with Obesity in a Randomized Controlled Trial. J. Nutr. 2021, 151, 1128–1138. [Google Scholar] [CrossRef] [PubMed]
- Asemi, Z.; Samimi, M.; Tabassi, Z.; Esmaillzadeh, A. The effect of DASH diet on pregnancy outcomes in gestational diabetes: A randomized controlled clinical trial. Eur. J. Clin. Nutr. 2014, 68, 490–495. [Google Scholar] [CrossRef] [Green Version]
- Babadi, M.; Khorshidi, A.; Aghadavood, E.; Samimi, M.; Kavossian, E.; Bahmani, F.; Mafi, A.; Shafabakhsh, R.; Satari, M.; Asemi, Z. The Effects of Probiotic Supplementation on Genetic and Metabolic Profiles in Patients with Gestational Diabetes Mellitus: A Randomized, Double-Blind, Placebo-Controlled Trial. Probiotics Antimicrob. Proteins 2019, 11, 1227–1235. [Google Scholar] [CrossRef]
- Santamaria, A.; Alibrandi, A.; Di Benedetto, A.; Pintaudi, B.; Corrado, F.; Facchinetti, F.; D’Anna, R. Clinical and metabolic outcomes in pregnant women at risk for gestational diabetes mellitus supplemented with myo-inositol: A secondary analysis from 3 RCTs. Am. J. Obstet. Gynecol. 2018, 219, 300.e1. [Google Scholar] [CrossRef]
- U.S. Department of Agriculture; U.S. Department of Health and Human Services. Dietary Guidelines for Americans, 2020–2025, 9th Edition. December 2020; pp. 107–120. Available online: DietaryGuidelines.gov (accessed on 28 November 2022).
Diets | Microbial Profiles | Metabolite Profiles |
---|---|---|
High fat diet | ↓ Bacteroides, Prevotella, Lactobacillus [19,154], and Bifidobacterium [19] | ↓ SCFAs (acetic, propionic acids, butyric, and isobutyric) [154,155] |
↑ Clostridium [151] and Firmicutes/Bacteroidetes ratio [154,155] | ↑ 3-hydroxybutyrate, acetone and acetoacetate, and 2-oxoisocaproate [151] | |
↑ isoleucine, leucine, and valine [151] and ↓ alanine, proline [151] | ||
↑ o-phosphocholine, cytidine [151] | ||
Curcumin, e.g., turmeric | ↑ Akkermansia, Bacteroides, Parabacteroides, Alistipes, and Alloprevotella [155] | ↑ acetate, propionic acid, butyric, and isobutyric [155] |
↓ Firmicutes/Bacteroidetes ratio, Desulfovibrio, and Ruminococcaceae [155] | ||
Sulforaphane, e.g., kale | ↑ Bacteroides fragilis, Clostridium cluster 1 [156], Bacteroidetes, and Lactobacillus [153] | ↑ butyric acid [156] |
↓ Actinobacteria, Proteobacteria, and Lactococcus [153] | ||
Green tea polyphenol | ↑ Bifidobacterium [157] and Bacteroidetes [153] | ↑ acetate and propionate [157] |
↓ Clostridium perfringens, C. difficile, [157,158], and Proteobacteria [153] | ↑ isobutyrate, valerate, and hexanoate [153] | |
Prebiotics, e.g., banana | ↑ Bifidobacterium and Bacteroides/Prevotella spp. [151] | ↑ myo-inositol [151] |
↓ Methanobrevibacter spp., Roseburia spp., Clostridium coccoides, and C. leptum [151] | ↑ arginine, ornithine, citrulline, and proline [151] | |
Diallyl disulfide, e.g., garlic | ↓ Parabacteroides, Faecalibacterium, Escherichia-Shigella, and Streptococcus [159] | ↑ 4-(2-aminophenyl)-2,4-dioxobutanoic acid, and kynurenic acid [159] |
↑ Ruminiclostridium, Oscillibacter, Ruminococcaceae, and Prevotellaceae [159] | ↑ 5-hydroxyindoleacetic acid, quinoline-4,8-diol, 4-(2-amino-3-hydroxyphenyl)-2,4-dioxobutanoic acid, 3-methyldioxyindole, 2-formaminobenzoylacetate, lipoxin A4, and cholic acids [159] | |
↑ Bifidobacterium, Bacteroidetes, and Lactobacillus [152] | ||
↑ Bacteroidetes/Firmicutes ratio [152] | ||
↑ SCAFs (acetate, valine) [152] | ||
Soy | ↑ Bacteroidetes, proteobacteria, Enterococcus, Lactobacilli, and Bifidobacterium [160,161,162] | ↑ SCAFs (butyric acid and lactic acid) [160,161,163] |
↓ Clostridia, Enterobacteria, Firmicutes [160,163] | ||
↑ Blautia and Veillonellaceae [164] | ||
Resveratrol, e.g., grapes | ↑ Lactobacillus, Bacteroidetes/Firmicutes ratio, and Bifidobacterium [135,165] | 3, 4′-dihydroxy-trans-stilbene and 3,4′-dihydroxy benzyl (lunularin) [135] |
↓ Enterococcus faecalis [135], Clostridia spp., Parabacteroides distasonis, and Gracilibacter thermotolerans [165] | glucuronide, sulfate [136,166] | |
↓ TMAO [167] | ||
Quercetin, e.g., citrus fruits | ↓ Firmicutes/Bacteroidetes ratio, Eubacterium cylindroides [165], Escherichia-shigella [168], Desulfovibrio, and Helicobacter [169] | ↑ butyrate [168,169], acetate, and propionate [169] |
Pectin, e.g., apples | ↑ Lactobacillus, Bifidobacterium [170,171,172] and Bacteroidetes [171] | ↑ acetate [171,173], propionate, and butyrate [171,172,173] |
↓ C. perfringens, Enterobacteriaceae, Pseudomonas [170], and Firmicutes [171] |
Clinical Trial | Dose/Supplement | Health Outcome on GDM | Reference |
---|---|---|---|
Effect of the Mediterranean diet supplemented with extra virgin oil and pistachios on GDM | Daily consumption of ≥40 mL of extra virgin oil and 25–30 g of pistachios in addition to basic Med Diet recommendations for 16 weeks | ↓ maternal fasting blood glucose, insulin resistance, and GWG ↓ neonatal LGA | [184] |
Dietary blueberry and fiber supplements for GDM women | 280 g whole blueberries and 12 g soluble fiber daily for 18 weeks | ↓ maternal weight gain and blood glucose | [185] |
Effect of DASH diet on GDM | Daily intake of a diet rich in fruits, vegetables, whole grains, low-fat dairy products, and a diet low in saturated fats, cholesterol, and refined grains and sweets for 4 weeks | Improved pregnancy and fetal health outcomes Reduced the need for insulin therapy ↓ fetal macrosomia | [186] |
Probiotic supplements | Daily intake of probiotic capsules (2 × 109 CFU/g each of a Lactobacillus spp. and b Bifidobacterium spp.) | ↑ PPAR-γ, HDL and antioxidant capacity ↓ TNF-α, fasting glucose, insulin resistance, TG, and VLDL | [187] |
Myo-inositol supplements | 4 g/day throughout pregnancy | Reduced the risk of preterm birth, macrosomia, and maternal weight gain | [188] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 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
Bankole, T.; Winn, H.; Li, Y. Dietary Impacts on Gestational Diabetes: Connection between Gut Microbiome and Epigenetic Mechanisms. Nutrients 2022, 14, 5269. https://doi.org/10.3390/nu14245269
Bankole T, Winn H, Li Y. Dietary Impacts on Gestational Diabetes: Connection between Gut Microbiome and Epigenetic Mechanisms. Nutrients. 2022; 14(24):5269. https://doi.org/10.3390/nu14245269
Chicago/Turabian StyleBankole, Taiwo, Hung Winn, and Yuanyuan Li. 2022. "Dietary Impacts on Gestational Diabetes: Connection between Gut Microbiome and Epigenetic Mechanisms" Nutrients 14, no. 24: 5269. https://doi.org/10.3390/nu14245269
APA StyleBankole, T., Winn, H., & Li, Y. (2022). Dietary Impacts on Gestational Diabetes: Connection between Gut Microbiome and Epigenetic Mechanisms. Nutrients, 14(24), 5269. https://doi.org/10.3390/nu14245269