Interaction and Metabolic Pathways: Elucidating the Role of Gut Microbiota in Gestational Diabetes Mellitus Pathogenesis
Abstract
:1. Introduction
2. Gut Microbiota’s Role in Gestational Diabetes Mellitus
2.1. Early Pregnancy: Gut Microbiota, Host Overweight, and GDM Predection
2.2. Second Trimester of Pregnancy: Gut Microbiota, Host GDM, and Metabolomis Pathogensis
2.3. Third Trimester of Pregnancy: Gut Microbiota Dysbiosis and Host Postpartum Diabetes Risk
2.4. Neonatal Impact: Maternal GDM and Neonatal Microbiota
3. Microbial Metabolites in GDM
3.1. Role of Different SCFAs
3.2. Role of Bile Acids
3.3. Trimethylamine N-Oxide, TMAO
4. Microbes and Microbial Metabolites in the Prevention and Treatment of GDM
4.1. Probiotic Supplements
4.2. Prebiotic Supplementation
5. Conclusions and Future Perspectives
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Buchanan, T.A.; Xiang, A.H. Gestational diabetes mellitus. J. Clin. Investig. 2005, 115, 485–491. [Google Scholar] [CrossRef] [PubMed]
- McIntyre, H.D.; Catalano, P.; Zhang, C.; Desoye, G.; Mathiesen, E.R.; Damm, P. Gestational diabetes mellitus. Nat. Rev. Dis. Primers 2019, 5, 47. [Google Scholar] [CrossRef]
- Lowe, W.L.; Scholtens, D.M.; Lowe, L.P.; Kuang, A.; Nodzenski, M.; Talbot, O.; Catalano, P.M.; Linder, B.; Brickman, W.J.; Clayton, P.; et al. Association of Gestational Diabetes With Maternal Disorders of Glucose Metabolism and Childhood Adiposity. JAMA 2018, 320, 1005–1016. [Google Scholar] [CrossRef] [PubMed]
- Ong, K.L.; Stafford, L.K.; McLaughlin, S.A.; Boyko, E.J.; Vollset, S.E.; Smith, A.E.; Dalton, B.E.; Duprey, J.; Cruz, J.A.; Hagins, H.; et al. Global, regional, and national burden of diabetes from 1990 to 2021, with projections of prevalence to 2050: A systematic analysis for the Global Burden of Disease Study 2021. Lancet 2023, 402, 203–234. [Google Scholar] [CrossRef] [PubMed]
- Billionnet, C.c.; Mitanchez, D.; Weill, A.; Nizard, J.; Alla, F.ß.; Hartemann, A.s.; Jacqueminet, S. Gestational diabetes and adverse perinatal outcomes from 716,152 births in France in 2012. Diabetologia 2017, 60, 636–644. [Google Scholar] [CrossRef] [PubMed]
- Song, C.; Lyu, Y.; Li, C.; Liu, P.; Li, J.; Ma, R.C.; Yang, X. Long-term risk of diabetes in women at varying durations after gestational diabetes: A systematic review and meta-analysis with more than 2 million women. Obes. Rev. 2018, 19, 421–429. [Google Scholar] [CrossRef] [PubMed]
- Sanna, S.; van Zuydam, N.R.; Mahajan, A.; Kurilshikov, A.; Vich Vila, A.; Võsa, U.; Mujagic, Z.; Masclee, A.A.M.; Jonkers, D.M.A.E.; Oosting, M.; et al. Causal Relationships among the Gut Microbiome, Short-Chain Fatty Acids and Metabolic Diseases. Nat. Genet. 2019, 51, 600–605. [Google Scholar] [CrossRef] [PubMed]
- Pedersen, H.K.; Gudmundsdottir, V.; Nielsen, H.B.r.; Hyotylainen, T.; Nielsen, T.; Jensen, B.A.H.; Forslund, K.; Hildebrand, F.; Prifti, E.; Falony, G.; et al. Human gut microbes impact host serum metabolome and insulin sensitivity. Nature 2016, 535, 376–381. [Google Scholar] [CrossRef]
- Perry, R.J.; Peng, L.; Barry, N.A.; Cline, G.W.; Zhang, D.; Cardone, R.L.; Petersen, K.F.; Kibbey, R.G.; Goodman, A.L.; Shulman, G.I. Acetate Mediates a Microbiome-Brain-β-Cell Axis to Promote Metabolic Syndrome. Nature 2016, 534, 213–217. [Google Scholar] [CrossRef]
- Jia, W.; Xie, G.; Jia, W. Bile Acid–Microbiota Crosstalk in Gastrointestinal Inflammation and Carcinogenesis. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 111–128. [Google Scholar] [CrossRef]
- Levy, M.; Thaiss, C.A.; Zeevi, D.; Dohnalová, L.; Zilberman-Schapira, G.; Mahdi, J.A.; David, E.; Savidor, A.; Korem, T.; Herzig, Y.; et al. Microbiota-Modulated Metabolites Shape the Intestinal Microenvironment by Regulating NLRP6 Inflammasome Signaling. Cell 2015, 163, 1428–1443. [Google Scholar] [CrossRef]
- Pinto, Y.; Frishman, S.; Turjeman, S.; Eshel, A.; Nuriel-Ohayon, M.; Shrossel, O.; Ziv, O.; Walters, W.; Parsonnet, J.; Ley, C.; et al. Gestational diabetes is driven by microbiota-induced inflammation months before diagnosis. Gut 2023, 72, 918–928. [Google Scholar] [CrossRef] [PubMed]
- Lloyd-Price, J.; Arze, C.; Ananthakrishnan, A.N.; Schirmer, M.; Avila-Pacheco, J.; Poon, T.W.; Andrews, E.; Ajami, N.J.; Bonham, K.S.; Brislawn, C.J.; et al. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature 2019, 569, 655–662. [Google Scholar] [CrossRef] [PubMed]
- Nagata, N.; Tohya, M.; Fukuda, S.; Suda, W.; Nishijima, S.; Takeuchi, F.; Ohsugi, M.; Tsujimoto, T.; Nakamura, T.; Shimomura, A.; et al. Effects of bowel preparation on the human gut microbiome and metabolome. Sci. Rep. 2019, 9, 4042. [Google Scholar] [CrossRef] [PubMed]
- Canfora, E.E.; van der Beek, C.M.; Jocken, J.W.E.; Goossens, G.H.; Holst, J.J.; Olde Damink, S.W.M.; Lenaerts, K.; Dejong, C.H.C.; Blaak, E.E. Colonic infusions of short-chain fatty acid mixtures promote energy metabolism in overweight/obese men: A randomized crossover trial. Sci. Rep. 2017, 7, 2360. [Google Scholar] [CrossRef] [PubMed]
- Baldini, F.; Heinken, A.; Heirendt, L.; Magnusdottir, S.; Fleming, R.M.T.; Thiele, I. The Microbiome Modeling Toolbox: From microbial interactions to personalized microbial communities. Bioinformatics 2019, 35, 2332–2334. [Google Scholar] [CrossRef]
- Nicholson, J.K.; Holmes, E.; Kinross, J.; Burcelin, R.; Gibson, G.; Jia, W.; Pettersson, S. Host-Gut Microbiota Metabolic Interactions. Science 2012, 336, 1262–1267. [Google Scholar] [CrossRef] [PubMed]
- Staley, C.; Weingarden, A.R.; Khoruts, A.; Sadowsky, M.J. Interaction of gut microbiota with bile acid metabolism and its influence on disease states. Appl. Microbiol. Biotechnol. 2017, 101, 47–64. [Google Scholar] [CrossRef]
- Racicot, K.; Kwon, J.-Y.; Aldo, P.; Silasi, M.; Mor, G. Understanding the complexity of the immune system during pregnancy. Am. J. Reprod. Immunol. 2014, 72, 107–116. [Google Scholar] [CrossRef]
- Thaweethai, T.; Soetan, Z.; James, K.; Florez, J.C.; Powe, C.E. Distinct Insulin Physiology Trajectories in Euglycemic Pregnancy and Gestational Diabetes Mellitus. Diabetes Care 2023, 46, 2137–2146. [Google Scholar] [CrossRef]
- Fugmann, M.; Breier, M.; Rottenkolber, M.; Banning, F.; Ferrari, U.; Sacco, V.; Grallert, H.; Parhofer, K.G.; Seissler, J.; Clavel, T.; et al. The stool microbiota of insulin resistant women with recent gestational diabetes, a high risk group for type 2 diabetes. Sci. Rep. 2015, 5, 13212. [Google Scholar] [CrossRef] [PubMed]
- Koren, O.; Goodrich, J.K.; Cullender, T.C.; Spor, A.; Laitinen, K.; Bäckhed, H.K.; Gonzalez, A.; Werner, J.J.; Angenent, L.T.; Knight, R.; et al. Host Remodeling of the Gut Microbiome and Metabolic Changes during Pregnancy. Cell 2012, 150, 470–480. [Google Scholar] [CrossRef] [PubMed]
- Pavlo, P.; Kamyshna, I.; Kamyshnyi, A. Effects of metformin on the gut microbiota: A systematic review. Mol. Metab. 2023, 77, 101805. [Google Scholar] [CrossRef]
- Molina-Vega, M.; Picón-César, M.J.; Gutiérrez-Repiso, C.; Fernández-Valero, A.; Lima-Rubio, F.; González-Romero, S.; Moreno-Indias, I.; Tinahones, F.J. Metformin Action over Gut Microbiota Is Related to Weight and Glycemic Control in Gestational Diabetes Mellitus: A Randomized Trial. Biomed. Pharmacother. 2022, 145, 112465. [Google Scholar] [CrossRef] [PubMed]
- Shin, N.-R.; Lee, J.-C.; Lee, H.-Y.; Kim, M.-S.; Whon, T.W.; Lee, M.-S.; Bae, J.-W. An increase in the Akkermansia spp. population induced by metformin treatment improves glucose homeostasis in diet-induced obese mice. Gut 2014, 63, 727–735. [Google Scholar] [CrossRef] [PubMed]
- Tocci, V.; Mirabelli, M.; Salatino, A.; Sicilia, L.; Giuliano, S.; Brunetti, F.S.; Chiefari, E.; De Sarro, G.; Foti, D.P.; Brunetti, A. Metformin in Gestational Diabetes Mellitus: To Use or Not to Use, That Is the Question. Pharmaceuticals 2023, 16, 1318. [Google Scholar] [CrossRef] [PubMed]
- Lebeer, S.; Vanderleyden, J.; De Keersmaecker, S.C.J. Host interactions of probiotic bacterial surface molecules: Comparison with commensals and pathogens. Nat. Rev. Microbiol. 2010, 8, 171–184. [Google Scholar] [CrossRef]
- O’Callaghan, A.; van Sinderen, D. Bifidobacteria and Their Role as Members of the Human Gut Microbiota. Front. Microbiol. 2016, 7, 925. [Google Scholar] [CrossRef]
- Levy, M.; Kolodziejczyk, A.A.; Thaiss, C.A.; Elinav, E. Dysbiosis and the immune system. Nat. Rev. Immunol. 2017, 17, 219–232. [Google Scholar] [CrossRef]
- Sassone-Corsi, M.; Nuccio, S.P.; Liu, H.; Hernandez, D.; Vu, C.T.; Takahashi, A.A.; Edwards, R.A.; Raffatellu, M. Microcins mediate competition among Enterobacteriaceae in the inflamed gut. Nature 2016, 540, 280–283. [Google Scholar] [CrossRef]
- Gomez-Arango, L.F.; Barrett, H.L.; McIntyre, H.D.; Callaway, L.K.; Morrison, M.; Dekker Nitert, M.; Group, S.T. Connections Between the Gut Microbiome and Metabolic Hormones in Early Pregnancy in Overweight and Obese Women. Diabetes 2016, 65, 2214–2223. [Google Scholar] [CrossRef] [PubMed]
- Hu, P.; Chen, X.; Chu, X.; Fan, M.; Ye, Y.; Wang, Y.; Han, M.; Yang, X.; Yuan, J.; Zha, L.; et al. Association of Gut Microbiota during Early Pregnancy with Risk of Incident Gestational Diabetes Mellitus. J. Clin. Endocrinol. Metab. 2021, 106, e4128–e4141. [Google Scholar] [CrossRef] [PubMed]
- Ma, S.; You, Y.; Huang, L.; Long, S.; Zhang, J.; Guo, C.; Zhang, N.; Wu, X.; Xiao, Y.; Tan, H. Alterations in Gut Microbiota of Gestational Diabetes Patients During the First Trimester of Pregnancy. Front. Cell Infect. Microbiol. 2020, 10, 58. [Google Scholar] [CrossRef] [PubMed]
- Mokkala, K.; Houttu, N.; Vahlberg, T.; Munukka, E.; Rönnemaa, T.; Laitinen, K. Gut Microbiota Aberrations Precede Diagnosis of Gestational Diabetes Mellitus. Acta Diabetol. 2017, 54, 1147–1149. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Liang, Y.-Y.; Liu, L.-Y.; Jia, Y.; Li, Y.; Cai, J.-N.; Shu, Y.; Tan, J.-Y.; Chen, P.-Y.; Li, H.-W.; Cai, H.-H.; et al. Correlation between gut microbiota and glucagon-like peptide-1 in patients with gestational diabetes mellitus. World J. Diabetes 2022, 13, 861–876. [Google Scholar] [CrossRef] [PubMed]
- Wei, J.; Qing, Y.; Zhou, H.; Liu, J.; Qi, C.; Gao, J. 16S rRNA gene amplicon sequencing of gut microbiota in gestational diabetes mellitus and their correlation with disease risk factors. J. Endocrinol. Investig. 2022, 45, 279–289. [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, 1840765. [Google Scholar] [CrossRef]
- Ye, G.; Zhang, L.; Wang, M.; Chen, Y.; Gu, S.; Wang, K.; Leng, J.; Gu, Y.; Xie, X. The Gut Microbiota in Women Suffering from Gestational Diabetes Mellitus with the Failure of Glycemic Control by Lifestyle Modification. J. Diabetes Res. 2019, 2019, 6081248. [Google Scholar] [CrossRef]
- Su, Y.; Wang, H.K.; Gan, X.P.; Chen, L.; Cao, Y.N.; Cheng, D.C.; Zhang, D.Y.; Liu, W.Y.; Li, F.F.; Xu, X.M. Alterations of gut microbiota in gestational diabetes patients during the second trimester of pregnancy in the Shanghai Han population. J. Transl. Med. 2021, 19, 366. [Google Scholar] [CrossRef]
- Smits, L.P.; Kootte, R.S.; Levin, E.; Prodan, A.; Fuentes, S.; Zoetendal, E.G.; Wang, Z.; Levison, B.S.; Cleophas, M.C.P.; Kemper, E.M.; et al. Effect of Vegan Fecal Microbiota Transplantation on Carnitine- and Choline-Derived Trimethylamine-N-Oxide Production and Vascular Inflammation in Patients With Metabolic Syndrome. J. Am. Heart Assoc. 2018, 7, e008342. [Google Scholar] [CrossRef] [PubMed]
- Quigley, E.M. Gut bacteria in health and disease. Gastroenterol. Hepatol. 2013, 9, 560–569. [Google Scholar]
- Qin, J.; Li, Y.; Cai, Z.; Li, S.; Zhu, J.; Zhang, F.; Liang, S.; Zhang, W.; Guan, Y.; Shen, D.; et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature 2012, 490, 55–60. [Google Scholar] [CrossRef]
- Org, E.; Blum, Y.; Kasela, S.; Mehrabian, M.; Kuusisto, J.; Kangas, A.J.; Soininen, P.; Wang, Z.; Ala-Korpela, M.; Hazen, S.L.; et al. Relationships between gut microbiota, plasma metabolites, and metabolic syndrome traits in the METSIM cohort. Genome Biol. 2017, 18, 70. [Google Scholar] [CrossRef]
- Ferretti, P.; Pasolli, E.; Tett, A.; Asnicar, F.; Gorfer, V.; Fedi, S.; Armanini, F.; Truong, D.T.; Manara, S.; Zolfo, M.; et al. Mother-to-Infant Microbial Transmission from Different Body Sites Shapes the Developing Infant Gut Microbiome. Cell Host Microbe 2018, 24, 133–145.e135. [Google Scholar] [CrossRef] [PubMed]
- Ferrocino, I.; Ponzo, V.; Gambino, R.; Zarovska, A.; Leone, F.; Monzeglio, C.; Goitre, I.; Rosato, R.; Romano, A.; Grassi, G.; et al. Changes in the gut microbiota composition during pregnancy in patients with gestational diabetes mellitus (GDM). Sci. Rep. 2018, 8, 12216. [Google Scholar] [CrossRef]
- Crusell, M.K.W.; 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, 89. [Google Scholar] [CrossRef]
- Allin, K.H.; Tremaroli, V.; Caesar, R.; Jensen, B.A.H.; Damgaard, M.T.F.; Bahl, M.I.; Licht, T.R.; Hansen, T.H.; Nielsen, T.; Dantoft, T.M.; et al. Aberrant intestinal microbiota in individuals with prediabetes. Diabetologia 2018, 61, 810–820. [Google Scholar] [CrossRef]
- Chen, T.; Qin, Y.; Chen, M.; Zhang, Y.; Wang, X.; Dong, T.; Chen, G.; Sun, X.; Lu, T.; White, R.A., 3rd; et al. Gestational diabetes mellitus is associated with the neonatal gut microbiota and metabolome. BMC Med. 2021, 19, 120. [Google Scholar] [CrossRef]
- Zhu, Q.; Yang, X.; Zhang, Y.; Shan, C.; Shi, Z. Role of the Gut Microbiota in the Increased Infant Body Mass Index Induced by Gestational Diabetes Mellitus. mSystems 2022, 7, e0046522. [Google Scholar] [CrossRef]
- Crusell, M.K.W.; 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. Comparative Studies of the Gut Microbiota in the Offspring of Mothers With and Without Gestational Diabetes. Front. Cell. Infect. Microbiol. 2020, 10, 536282. [Google Scholar] [CrossRef] [PubMed]
- Neri, C.; Serafino, E.; Morlando, M.; Familiari, A. Microbiome and Gestational Diabetes: Interactions with Pregnancy Outcome and Long-Term Infant Health. J. Diabetes Res. 2021, 2021, 9994734. [Google Scholar] [CrossRef] [PubMed]
- Ponzo, V.; Ferrocino, I.; Zarovska, A.; Amenta, M.B.; Leone, F.; Monzeglio, C.; Rosato, R.; Pellegrini, M.; Gambino, R.; Cassader, M.; et al. The microbiota composition of the offspring of patients with gestational diabetes mellitus (GDM). PLoS ONE 2019, 14, e0226545. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Zheng, J.; Shi, W.; Du, N.; Xu, X.; Zhang, Y.; Ji, P.; Zhang, F.; Jia, Z.; Wang, Y.; et al. Dysbiosis of maternal and neonatal microbiota associated with gestational diabetes mellitus. Gut 2018, 67, 1614–1625. [Google Scholar] [CrossRef] [PubMed]
- Hu, J.; Nomura, Y.; Bashir, A.; Fernandez-Hernandez, H.; Itzkowitz, S.; Pei, Z.; Stone, J.; Loudon, H.; Peter, I. Diversified microbiota of meconium is affected by maternal diabetes status. PLoS ONE 2013, 8, e78257. [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, 3044534. [Google Scholar] [CrossRef]
- Smilowitz, J.T.; O’Sullivan, A.; Barile, D.; German, J.B.; Lönnerdal, B.; Slupsky, C.M. The human milk metabolome reveals diverse oligosaccharide profiles. J. Nutr. 2013, 143, 1709–1718. [Google Scholar] [CrossRef]
- Sundekilde, U.K.; Downey, E.; O’Mahony, J.A.; O’Shea, C.A.; Ryan, C.A.; Kelly, A.L.; Bertram, H.C. The Effect of Gestational and Lactational Age on the Human Milk Metabolome. Nutrients 2016, 8, 304. [Google Scholar] [CrossRef]
- Yefet, E.; Bar, L.; Izhaki, I.; Iskander, R.; Massalha, M.; Younis, J.S.; Nachum, Z. Effects of Probiotics on Glycemic Control and Metabolic Parameters in Gestational Diabetes Mellitus: Systematic Review and Meta-Analysis. Nutrients 2023, 15, 1633. [Google Scholar] [CrossRef]
- Pammi, M.; Cope, J.; Tarr, P.I.; Warner, B.B.; Morrow, A.L.; Mai, V.; Gregory, K.E.; Kroll, J.S.; McMurtry, V.; Ferris, M.J.; et al. Intestinal dysbiosis in preterm infants preceding necrotizing enterocolitis: A systematic review and meta-analysis. Microbiome 2017, 5, 31. [Google Scholar] [CrossRef]
- Sordillo, J.E.; Zhou, Y.; McGeachie, M.J.; Ziniti, J.; Lange, N.; Laranjo, N.; Savage, J.R.; Carey, V.; O’Connor, G.; Sandel, M.; et al. Factors influencing the infant gut microbiome at age 3–6 months: Findings from the ethnically diverse Vitamin D Antenatal Asthma Reduction Trial (VDAART). J. Allergy Clin. Immunol. 2017, 139, 482–491.e414. [Google Scholar] [CrossRef]
- Sun, Z.; Pan, X.F.; Li, X.; Jiang, L.; Hu, P.; Wang, Y.; Ye, Y.; Wu, P.; Zhao, B.; Xu, J.; et al. The Gut Microbiome Dynamically Associates with Host Glucose Metabolism throughout Pregnancy: Longitudinal Findings from a Matched Case-Control Study of Gestational Diabetes Mellitus. Adv. Sci. 2023, 10, e2205289. [Google Scholar] [CrossRef]
- Ríos-Covián, D.; Ruas-Madiedo, P.; Margolles, A.; Gueimonde, M.; de Los Reyes-Gavilán, C.G.; Salazar, N. Intestinal Short Chain Fatty Acids and their Link with Diet and Human Health. Front. Microbiol. 2016, 7, 185. [Google Scholar] [CrossRef] [PubMed]
- Agus, A.; Planchais, J.; Sokol, H. Gut Microbiota Regulation of Tryptophan Metabolism in Health and Disease. Cell Host Microbe 2018, 23, 716–724. [Google Scholar] [CrossRef] [PubMed]
- Lin, H.V.; Frassetto, A.; Kowalik, E.J., Jr.; Nawrocki, A.R.; Lu, M.M.; Kosinski, J.R.; Hubert, J.A.; Szeto, D.; Yao, X.; Forrest, G.; et al. Butyrate and propionate protect against diet-induced obesity and regulate gut hormones via free fatty acid receptor 3-independent mechanisms. PLoS ONE 2012, 7, e35240. [Google Scholar] [CrossRef] [PubMed]
- Joyce, S.A.; MacSharry, J.; Casey, P.G.; Kinsella, M.; Murphy, E.F.; Shanahan, F.; Hill, C.; Gahan, C.G. Regulation of host weight gain and lipid metabolism by bacterial bile acid modification in the gut. Proc. Natl. Acad. Sci. USA 2014, 111, 7421–7426. [Google Scholar] [CrossRef] [PubMed]
- LeBlanc, J.G.; Milani, C.; de Giori, G.S.; Sesma, F.; van Sinderen, D.; Ventura, M. Bacteria as vitamin suppliers to their host: A gut microbiota perspective. Curr. Opin. Biotechnol. 2013, 24, 160–168. [Google Scholar] [CrossRef] [PubMed]
- He, J.; Zhang, P.; Shen, L.; Niu, L.; Tan, Y.; Chen, L.; Zhao, Y.; Bai, L.; Hao, X.; Li, X.; et al. Short-Chain Fatty Acids and Their Association with Signalling Pathways in Inflammation, Glucose and Lipid Metabolism. Int. J. Mol. Sci. 2020, 21, 6356. [Google Scholar] [CrossRef]
- Walter, J.; Martínez, I.; Rose, D.J. Holobiont nutrition: Considering the role of the gastrointestinal microbiota in the health benefits of whole grains. Gut Microbes 2013, 4, 340–346. [Google Scholar] [CrossRef]
- Louis, P.; Flint, H.J. Formation of propionate and butyrate by the human colonic microbiota. Environ. Microbiol. 2017, 19, 29–41. [Google Scholar] [CrossRef]
- Morrison, D.J.; Preston, T. Formation of short chain fatty acids by the gut microbiota and their impact on human metabolism. Gut Microbes 2016, 7, 189–200. [Google Scholar] [CrossRef] [PubMed]
- Tang, C.; Ahmed, K.; Gille, A.; Lu, S.; Gröne, H.J.; Tunaru, S.; Offermanns, S. Loss of FFA2 and FFA3 increases insulin secretion and improves glucose tolerance in type 2 diabetes. Nat. Med. 2015, 21, 173–177. [Google Scholar] [CrossRef] [PubMed]
- Kimura, I.; Ozawa, K.; Inoue, D.; Imamura, T.; Kimura, K.; Maeda, T.; Terasawa, K.; Kashihara, D.; Hirano, K.; Tani, T.; et al. The gut microbiota suppresses insulin-mediated fat accumulation via the short-chain fatty acid receptor GPR43. Nat. Commun. 2013, 4, 1829. [Google Scholar] [CrossRef]
- Roy, R.; Nguyen-Ngo, C.; Lappas, M. Short-chain fatty acids as novel therapeutics for gestational diabetes. J. Mol. Endocrinol. 2020, 65, 21–34. [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] [PubMed]
- Ziętek, M.; Celewicz, Z.; Szczuko, M. Short-Chain Fatty Acids, Maternal Microbiota and Metabolism in Pregnancy. Nutrients 2021, 13, 1244. [Google Scholar] [CrossRef] [PubMed]
- Layden, B.T.; Angueira, A.R.; Brodsky, M.; Durai, V.; Lowe, W.L., Jr. Short chain fatty acids and their receptors: New metabolic targets. Transl. Res. 2013, 161, 131–140. [Google Scholar] [CrossRef]
- Kebede, M.A.; Alquier, T.; Latour, M.G.; Poitout, V. Lipid receptors and islet function: Therapeutic implications? Diabetes Obes. Metab. 2009, 11 (Suppl. S4), 10–20. [Google Scholar] [CrossRef]
- Wang, S.; Liu, Y.; Qin, S.; Yang, H. Composition of Maternal Circulating Short-Chain Fatty Acids in Gestational Diabetes Mellitus and Their Associations with Placental Metabolism. Nutrients 2022, 14, 3727. [Google Scholar] [CrossRef]
- Kong, M.; Lu, Z.; Zhong, C.; Gao, Q.; Zhou, X.; Chen, R.; Xiong, G.; Hao, L.; Yang, X.; Yang, N. A higher level of total bile acid in early mid-pregnancy is associated with an increased risk of gestational diabetes mellitus: A prospective cohort study in Wuhan, China. J. Endocrinol. Investig. 2020, 43, 1097–1103. [Google Scholar] [CrossRef]
- Li, J.; Huo, X.; Cao, Y.F.; Li, S.N.; Du, Z.; Shao, P.; Leng, J.; Zhang, C.; Sun, X.Y.; Ma, R.C.W.; et al. Bile acid metabolites in early pregnancy and risk of gestational diabetes in Chinese women: A nested case-control study. EBioMedicine 2018, 35, 317–324. [Google Scholar] [CrossRef] [PubMed]
- Chiang, J.Y. Bile acid metabolism and signaling. Compr. Physiol. 2013, 3, 1191–1212. [Google Scholar] [CrossRef] [PubMed]
- Lefebvre, P.; Cariou, B.; Lien, F.; Kuipers, F.; Staels, B. Role of bile acids and bile acid receptors in metabolic regulation. Physiol. Rev. 2009, 89, 147–191. [Google Scholar] [CrossRef] [PubMed]
- Ridlon, J.M.; Harris, S.C.; Bhowmik, S.; Kang, D.J.; Hylemon, P.B. Consequences of bile salt biotransformations by intestinal bacteria. Gut Microbes 2016, 7, 22–39. [Google Scholar] [CrossRef] [PubMed]
- Wahlström, A.; Sayin, S.I.; Marschall, H.U.; Bäckhed, F. Intestinal Crosstalk between Bile Acids and Microbiota and Its Impact on Host Metabolism. Cell Metab. 2016, 24, 41–50. [Google Scholar] [CrossRef]
- Joyce, S.A.; Shanahan, F.; Hill, C.; Gahan, C.G. Bacterial bile salt hydrolase in host metabolism: Potential for influencing gastrointestinal microbe-host crosstalk. Gut Microbes 2014, 5, 669–674. [Google Scholar] [CrossRef] [PubMed]
- Kumar, M.; Babaei, P.; Ji, B.; Nielsen, J. Human gut microbiota and healthy aging: Recent developments and future prospective. Nutr. Healthy Aging 2016, 4, 3–16. [Google Scholar] [CrossRef] [PubMed]
- Zheng, J.; Wittouck, S.; Salvetti, E.; Franz, C.; Harris, H.M.B.; Mattarelli, P.; O’Toole, P.W.; Pot, B.; Vandamme, P.; Walter, J.; et al. A taxonomic note on the genus Lactobacillus: Description of 23 novel genera, emended description of the genus Lactobacillus Beijerinck 1901, and union of Lactobacillaceae and Leuconostocaceae. Int. J. Syst. Evol. Microbiol. 2020, 70, 2782–2858. [Google Scholar] [CrossRef]
- Kageyama, A.; Benno, Y. Catenibacterium mitsuokai gen. nov., sp. nov., a gram-positive anaerobic bacterium isolated from human faeces. Int. J. Syst. Evol. Microbiol. 2000; 50, Pt 4, 1595–1599. [Google Scholar] [CrossRef]
- Yoshimoto, S.; Loo, T.M.; Atarashi, K.; Kanda, H.; Sato, S.; Oyadomari, S.; Iwakura, Y.; Oshima, K.; Morita, H.; Hattori, M.; et al. Obesity-induced gut microbial metabolite promotes liver cancer through senescence secretome. Nature 2013, 499, 97–101. [Google Scholar] [CrossRef]
- Randrianarisoa, E.; Lehn-Stefan, A.; Wang, X.; Hoene, M.; Peter, A.; Heinzmann, S.S.; Zhao, X.; Königsrainer, I.; Königsrainer, A.; Balletshofer, B.; et al. Relationship of Serum Trimethylamine N-Oxide (TMAO) Levels with early Atherosclerosis in Humans. Sci. Rep. 2016, 6, 26745. [Google Scholar] [CrossRef]
- Dambrova, M.; Latkovskis, G.; Kuka, J.; Strele, I.; Konrade, I.; Grinberga, S.; Hartmane, D.; Pugovics, O.; Erglis, A.; Liepinsh, E. Diabetes is Associated with Higher Trimethylamine N-oxide Plasma Levels. Exp. Clin. Endocrinol. Diabetes 2016, 124, 251–256. [Google Scholar] [CrossRef]
- Cho, C.E.; Taesuwan, S.; Malysheva, O.V.; Bender, E.; Tulchinsky, N.F.; Yan, J.; Sutter, J.L.; Caudill, M.A. Trimethylamine-N-oxide (TMAO) response to animal source foods varies among healthy young men and is influenced by their gut microbiota composition: A randomized controlled trial. Mol. Nutr. Food Res. 2017, 61, 1600324. [Google Scholar] [CrossRef]
- Heianza, Y.; Ma, W.; Manson, J.E.; Rexrode, K.M.; Qi, L. Gut Microbiota Metabolites and Risk of Major Adverse Cardiovascular Disease Events and Death: A Systematic Review and Meta-Analysis of Prospective Studies. J. Am. Heart Assoc. 2017, 6, e004947. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Zeisel, S.H.; Wishnok, J.S.; Blusztajn, J.K. Formation of methylamines from ingested choline and lecithin. J. Pharmacol. Exp. Ther. 1983, 225, 320–324. [Google Scholar] [PubMed]
- Zhang, A.Q.; Mitchell, S.C.; Smith, R.L. Dietary precursors of trimethylamine in man: A pilot study. Food Chem. Toxicol. 1999, 37, 515–520. [Google Scholar] [CrossRef]
- Sun, X.; Jiao, X.; Ma, Y.; Liu, Y.; Zhang, L.; He, Y.; Chen, Y. Trimethylamine N-oxide induces inflammation and endothelial dysfunction in human umbilical vein endothelial cells via activating ROS-TXNIP-NLRP3 inflammasome. Biochem. Biophys. Res. Commun. 2016, 481, 63–70. [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]
- Lappas, M.; Permezel, M.; Rice, G.E. Release of proinflammatory cytokines and 8-isoprostane from placenta, adipose tissue, and skeletal muscle from normal pregnant women and women with gestational diabetes mellitus. J. Clin. Endocrinol. Metab. 2004, 89, 5627–5633. [Google Scholar] [CrossRef]
- Overton, H.A.; Babbs, A.J.; Doel, S.M.; Fyfe, M.C.; Gardner, L.S.; Griffin, G.; Jackson, H.C.; Procter, M.J.; Rasamison, C.M.; Tang-Christensen, M.; et al. Deorphanization of a G protein-coupled receptor for oleoylethanolamide and its use in the discovery of small-molecule hypophagic agents. Cell Metab. 2006, 3, 167–175. [Google Scholar] [CrossRef]
- Lan, H.; Vassileva, G.; Corona, A.; Liu, L.; Baker, H.; Golovko, A.; Abbondanzo, S.J.; Hu, W.; Yang, S.; Ning, Y.; et al. GPR119 is required for physiological regulation of glucagon-like peptide-1 secretion but not for metabolic homeostasis. J. Endocrinol. 2009, 201, 219–230. [Google Scholar] [CrossRef] [PubMed]
- Wang, L.; Christophersen, C.T.; Sorich, M.J.; Gerber, J.P.; Angley, M.T.; Conlon, M.A. Increased abundance of Sutterella spp. and Ruminococcus torques in feces of children with autism spectrum disorder. Mol. Autism 2013, 4, 42. [Google Scholar] [CrossRef] [PubMed]
- Fanning, S.; Hall, L.J.; Cronin, M.; Zomer, A.; MacSharry, J.; Goulding, D.; Motherway, M.O.; Shanahan, F.; Nally, K.; Dougan, G.; et al. Bifidobacterial surface-exopolysaccharide facilitates commensal-host interaction through immune modulation and pathogen protection. Proc. Natl. Acad. Sci. USA 2012, 109, 2108–2113. [Google Scholar] [CrossRef]
- Zietek, T.; Rath, E.; Haller, D.; Daniel, H. Intestinal organoids for assessing nutrient transport, sensing and incretin secretion. Sci. Rep. 2015, 5, 16831. [Google Scholar] [CrossRef] [PubMed]
- Sridharan, G.V.; Choi, K.; Klemashevich, C.; Wu, C.; Prabakaran, D.; Pan, L.B.; Steinmeyer, S.; Mueller, C.; Yousofshahi, M.; Alaniz, R.C.; et al. Prediction and quantification of bioactive microbiota metabolites in the mouse gut. Nat. Commun. 2014, 5, 5492. [Google Scholar] [CrossRef]
- Janeiro, M.H.; Ramírez, M.J.; Milagro, F.I.; Martínez, J.A.; Solas, M. Implication of Trimethylamine N-Oxide (TMAO) in Disease: Potential Biomarker or New Therapeutic Target. Nutrients 2018, 10, 1398. [Google Scholar] [CrossRef] [PubMed]
- Zhu, Y.; Barupal, D.K.; Ngo, A.L.; Quesenberry, C.P.; Feng, J.; Fiehn, O.; Ferrara, A. Predictive Metabolomic Markers in Early to Mid-pregnancy for Gestational Diabetes Mellitus: A Prospective Test and Validation Study. Diabetes 2022, 71, 1807–1817. [Google Scholar] [CrossRef]
- Dodd, D.; Spitzer, M.H.; Van Treuren, W.; Merrill, B.D.; Hryckowian, A.J.; Higginbottom, S.K.; Le, A.; Cowan, T.M.; Nolan, G.P.; Fischbach, M.A.; et al. A gut bacterial pathway metabolizes aromatic amino acids into nine circulating metabolites. Nature 2017, 551, 648–652. [Google Scholar] [CrossRef]
- Krishnan, S.; Ding, Y.; Saedi, N.; Choi, M.; Sridharan, G.V.; Sherr, D.H.; Yarmush, M.L.; Alaniz, R.C.; Jayaraman, A.; Lee, K. Gut Microbiota-Derived Tryptophan Metabolites Modulate Inflammatory Response in Hepatocytes and Macrophages. Cell Rep. 2018, 23, 1099–1111. [Google Scholar] [CrossRef]
- Halkjaer, S.I.; Nilas, L.; Carlsen, E.M.; Cortes, D.; Halldórsson, T.I.; Olsen, S.F.; Pedersen, A.E.; Krogfelt, K.A.; Petersen, A.M. Effects of probiotics (Vivomixx®) in obese pregnant women and their newborn: Study protocol for a randomized controlled trial. Trials 2016, 17, 491. [Google Scholar] [CrossRef]
- Lindsay, K.L.; Kennelly, M.; Culliton, M.; Smith, T.; Maguire, O.C.; Shanahan, F.; Brennan, L.; McAuliffe, F.M. Probiotics in obese pregnancy do not reduce maternal fasting glucose: A double-blind, placebo-controlled, randomized trial (Probiotics in Pregnancy Study). Am. J. Clin. Nutr. 2014, 99, 1432–1439. [Google Scholar] [CrossRef] [PubMed]
- Luoto, R.; Laitinen, K.; Nermes, M.; Isolauri, E. Impact of maternal probiotic-supplemented dietary counselling on pregnancy outcome and prenatal and postnatal growth: A double-blind, placebo-controlled study. Br. J. Nutr. 2010, 103, 1792–1799. [Google Scholar] [CrossRef] [PubMed]
- Wickens, K.L.; Barthow, C.A.; Murphy, R.; Abels, P.R.; Maude, R.M.; Stone, P.R.; Mitchell, E.A.; Stanley, T.V.; Purdie, G.L.; Kang, J.M.; et al. Early pregnancy probiotic supplementation with Lactobacillus rhamnosus HN001 may reduce the prevalence of gestational diabetes mellitus: A randomised controlled trial. Br. J. Nutr. 2017, 117, 804–813. [Google Scholar] [CrossRef] [PubMed]
- Geurts, L.; Neyrinck, A.M.; Delzenne, N.M.; Knauf, C.; Cani, P.D. Gut microbiota controls adipose tissue expansion, gut barrier and glucose metabolism: Novel insights into molecular targets and interventions using prebiotics. Benef. Microbes 2014, 5, 3–17. [Google Scholar] [CrossRef] [PubMed]
- Taylor, B.L.; Woodfall, G.E.; Sheedy, K.E.; O’Riley, M.L.; Rainbow, K.A.; Bramwell, E.L.; Kellow, N.J. Effect of Probiotics on Metabolic Outcomes in Pregnant Women with Gestational Diabetes: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Nutrients 2017, 9, 461. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Lindsay, K.L.; Brennan, L.; Kennelly, M.A.; Maguire, O.C.; Smith, T.; Curran, S.; Coffey, M.; Foley, M.E.; Hatunic, M.; Shanahan, F.; et al. Impact of probiotics in women with gestational diabetes mellitus on metabolic health: A randomized controlled trial. Am. J. Obstet. Gynecol. 2015, 212, 496.e1–e11. [Google Scholar] [CrossRef]
- Callaway, L.K.; McIntyre, H.D.; Barrett, H.L.; Foxcroft, K.; Tremellen, A.; Lingwood, B.E.; Tobin, J.M.; Wilkinson, S.; Kothari, A.; Morrison, M.; et al. Probiotics for the Prevention of Gestational Diabetes Mellitus in Overweight and Obese Women: Findings From the SPRING Double-Blind Randomized Controlled Trial. Diabetes Care 2019, 42, 364–371. [Google Scholar] [CrossRef]
- Hasain, Z.; Raja Ali, R.A.; Ahmad, H.F.; Abdul Rauf, U.F.; Oon, S.F.; Mokhtar, N.M. The Roles of Probiotics in the Gut Microbiota Composition and Metabolic Outcomes in Asymptomatic Post-Gestational Diabetes Women: A Randomized Controlled Trial. Nutrients 2022, 14, 3878. [Google Scholar] [CrossRef]
- Nabhani, Z.; Hezaveh, S.J.G.; Razmpoosh, E.; Asghari-Jafarabadi, M.; Gargari, B.P. The effects of synbiotic supplementation on insulin resistance/sensitivity, lipid profile and total antioxidant capacity in women with gestational diabetes mellitus: A randomized double blind placebo controlled clinical trial. Diabetes Res. Clin. Pract. 2018, 138, 149–157. [Google Scholar] [CrossRef]
- Gibson, G.R.; Probert, H.M.; Loo, J.V.; Rastall, R.A.; Roberfroid, M.B. Dietary modulation of the human colonic microbiota: Updating the concept of prebiotics. Nutr. Res. Rev. 2004, 17, 259–275. [Google Scholar] [CrossRef]
- Roberfroid, M. Prebiotics: The concept revisited. J. Nutr. 2007, 137, 830s–837s. [Google Scholar] [CrossRef] [PubMed]
- Slavin, J. Fiber and prebiotics: Mechanisms and health benefits. Nutrients 2013, 5, 1417–1435. [Google Scholar] [CrossRef] [PubMed]
- Salminen, S.; Collado, M.C.; Endo, A.; Hill, C.; Lebeer, S.; Quigley, E.M.M.; Sanders, M.E.; Shamir, R.; Swann, J.R.; Szajewska, H.; et al. The International Scientific Association of Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of postbiotics. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 649–667. [Google Scholar] [CrossRef] [PubMed]
- Kellow, N.J.; Coughlan, M.T.; Reid, C.M. Metabolic benefits of dietary prebiotics in human subjects: A systematic review of randomised controlled trials. Br. J. Nutr. 2014, 111, 1147–1161. [Google Scholar] [CrossRef]
- Roberfroid, M.; Gibson, G.R.; Hoyles, L.; McCartney, A.L.; Rastall, R.; Rowland, I.; Wolvers, D.; Watzl, B.; Szajewska, H.; Stahl, B.; et al. Prebiotic effects: Metabolic and health benefits. Br. J. Nutr. 2010, 104 (Suppl. S2), S1–S63. [Google Scholar] [CrossRef]
- Canfora, E.E.; Jocken, J.W.; Blaak, E.E. Short-chain fatty acids in control of body weight and insulin sensitivity. Nat. Rev. Endocrinol. 2015, 11, 577–591. [Google Scholar] [CrossRef]
- Everard, A.; Lazarevic, V.; Derrien, M.; Girard, M.; Muccioli, G.G.; Neyrinck, A.M.; Possemiers, S.; Van Holle, A.; François, P.; de Vos, W.M.; et al. Responses of gut microbiota and glucose and lipid metabolism to prebiotics in genetic obese and diet-induced leptin-resistant mice. Diabetes 2011, 60, 2775–2786. [Google Scholar] [CrossRef]
- Kakiyama, G.; Pandak, W.M.; Gillevet, P.M.; Hylemon, P.B.; Heuman, D.M.; Daita, K.; Takei, H.; Muto, A.; Nittono, H.; Ridlon, J.M.; et al. Modulation of the fecal bile acid profile by gut microbiota in cirrhosis. J. Hepatol. 2013, 58, 949–955. [Google Scholar] [CrossRef]
- Brighton, C.A.; Rievaj, J.; Kuhre, R.E.; Glass, L.L.; Schoonjans, K.; Holst, J.J.; Gribble, F.M.; Reimann, F. Bile Acids Trigger GLP-1 Release Predominantly by Accessing Basolaterally Located G Protein-Coupled Bile Acid Receptors. Endocrinology 2015, 156, 3961–3970. [Google Scholar] [CrossRef]
- Kumar, D.P.; Rajagopal, S.; Mahavadi, S.; Mirshahi, F.; Grider, J.R.; Murthy, K.S.; Sanyal, A.J. Activation of transmembrane bile acid receptor TGR5 stimulates insulin secretion in pancreatic β cells. Biochem. Biophys. Res. Commun. 2012, 427, 600–605. [Google Scholar] [CrossRef]
- Everard, A.; Belzer, C.; Geurts, L.; Ouwerkerk, J.P.; Druart, C.; Bindels, L.B.; Guiot, Y.; Derrien, M.; Muccioli, G.G.; Delzenne, N.M.; et al. Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc. Natl. Acad. Sci. USA 2013, 110, 9066–9071. [Google Scholar] [CrossRef]
- Monte, M.J.; Marin, J.J.; Antelo, A.; Vazquez-Tato, J. Bile acids: Chemistry, physiology, and pathophysiology. World J. Gastroenterol. 2009, 15, 804–816. [Google Scholar] [CrossRef] [PubMed]
- Koeth, R.A.; Wang, Z.; Levison, B.S.; Buffa, J.A.; Org, E.; Sheehy, B.T.; Britt, E.B.; Fu, X.; Wu, Y.; Li, L.; et al. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat. Med. 2013, 19, 576–585. [Google Scholar] [CrossRef]
- Wang, Z.; Klipfell, E.; Bennett, B.J.; Koeth, R.; Levison, B.S.; Dugar, B.; Feldstein, A.E.; Britt, E.B.; Fu, X.; Chung, Y.M.; et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature 2011, 472, 57–63. [Google Scholar] [CrossRef] [PubMed]
- Le Gall, G.; Noor, S.O.; Ridgway, K.; Scovell, L.; Jamieson, C.; Johnson, I.T.; Colquhoun, I.J.; Kemsley, E.K.; Narbad, A. Metabolomics of fecal extracts detects altered metabolic activity of gut microbiota in ulcerative colitis and irritable bowel syndrome. J. Proteome Res. 2011, 10, 4208–4218. [Google Scholar] [CrossRef] [PubMed]
- Bennett, B.J.; de Aguiar Vallim, T.Q.; Wang, Z.; Shih, D.M.; Meng, Y.; Gregory, J.; Allayee, H.; Lee, R.; Graham, M.; Crooke, R.; et al. Trimethylamine-N-oxide, a metabolite associated with atherosclerosis, exhibits complex genetic and dietary regulation. Cell Metab. 2013, 17, 49–60. [Google Scholar] [CrossRef] [PubMed]
- Laitinen, K.; Poussa, T.; Isolauri, E. Probiotics and dietary counselling contribute to glucose regulation during and after pregnancy: A randomised controlled trial. Br. J. Nutr. 2009, 101, 1679–1687. [Google Scholar] [CrossRef] [PubMed]
Title | Study Type | Timing of Analysis | Country and/or Ethnicity | Sample Size | Main Results | |||
---|---|---|---|---|---|---|---|---|
GDM | non = GDM | Microbial Abundance and Correlations | Inferred Functional | Gut Microbiota Profile in GDM Patients in Comparison to Controls (Phylum/Order/Family/Genus) | ||||
Mokkala et al. [34] | Prospective cohort study | 12.9 weeks of gestation | Finland | 15 | 60 | Ruminococcaceae abundance is positively correlated with fasting glucose. | Ruminococcaceae may impede glucose homeostasis by affecting inflammation, insulin signaling, SCFA production, and energy harvest. | Genus: Increased abundance of an unidentified genus of Ruminococcaceae family in GDM patients. |
Ma et al. [33] | Nested case–control study | 10–15 weeks of gestation | Chinese Han ethnicity | 98 | 98 | Eisenbergiella and Tyzzerella are positively correlated with fasting glucose. Parabacteroides, Parasutterella, and Ruminococcaceae were negatively correlated with fasting glucose. | Predicted enrichment of sphingolipid metabolism, starch/sucrose metabolism pathways in GDM. Reduction in lysine biosynthesis and nitrogen metabolism pathways in GDM. | Genus: Higher Eisenbergiella, Tyzzerella 4, Lachnospiraceae NK4A136 in GDM group. Higher Parabacteroides, Megasphaera, Ruminococcaceae, Eubacterium in controls. |
Gomez et al. [31] | Cross-sectional study | mean 16 weeks of gestation | Australia, mostly Caucasian | 18 | 70 (29 overweight, 41 obese)—18 developed GDM later | The genus Collinsella (phylum Actinobacteria) was positively correlated with insulin, C-peptide, HOMA-IR, triglycerides, and VLDL cholesterol. The genus Coprococcus (family Lachnospiraceae) was positively correlated with GIP. Bacteroidaceae positively and Prevotellaceae negatively correlated with ghrelin. | Maternal metabolic hormones. | Increased Actinobacteria. |
Su et al. [40] | Cross-sectional study | 24–28 weeks of gestation | Shanghai, China; Han ethnicity | 21 | 32 | Bacteroidetes positively correlated with 1hPG, FINS, 1hPIN, and HOMA-IR. Proteobacteria, Actinobacteria, and Verrucomicrobia negatively correlated with 1hPG. Ruminococcaceae UCG014 negatively correlated with glycemic traits. Incertae Sedis positively correlated with FPG and 1hPG. Akkermansia negatively correlated with 1hPG. | GDM patients had higher microbial gene functions related to amino sugar and nucleotide sugar metabolism. Bacteroides genus positively correlated with amino sugar/nucleotide sugar metabolism. Controls had higher gene functions related to two-component system, ABC transporters, and transporters | Phyla: Bacteroidetes higher, and Proteobacteria, Actinobacteria, Verrucomicrobia lower in GDM. Genera: Bacteroides Parabacteroides higher, Akkermansia Rhodococcus lower in GDM. |
Wang et al. [54] | Case–control study | 24–28 weeks of gestation | China | 59 | 48 | Lachnospiraceae OTUs positively correlated with glucose levels. Enterobacteriaceae OTUs negatively correlated with glucose levels. | Disturbances in fecal and urinary metabolites related to amino acid and carbohydrate metabolism. | At the family level, there was increased Lachnospiraceae and decreased Enterobacteriaceae and Ruminococcaceae in GDM. |
Wei et al. [37] | Case–control study | 24–28 weeks of gestation | China | 15 | 18 | S. infantis positively correlated with glucose levels. | - | GDM patients had increased Ruminococcus bromii, Clostridium colinum, and Streptococcus infantis at genus level compared to controls. |
Chen et al. [49] | Cross-sectional study | 24–28 weeks of gestation | China | 30 | 28 | Prevotella and Romboutsia genera negatively correlated with 2 h glucose in GDM Aureimonas, Kosakonia species positively correlated and Peptostreptococcus negatively correlated with fasting glucose in GDM. | Depletion of beneficial acetate and lactate-producing bacteria like Bifidobacterium, and butyrate-producing bacteria like Eubacterium suggests beneficial SCFA production may be reduced in GDM. | At the genus level, 54 differentially abundant taxa identified, with 42 genera depleted in GDM (e.g., Prevotella, Romboutsia). GDM had lower levels of beneficial bacteria like Bifidobacterium, Eubacterium, and Prevotella species. GDM was enriched in Blautia hydrogenotrophica and some Corynebacterium, Lactobacillus species. |
Liang et al. [36] | Case–control study | Second trimester | China | 35 | 25 | Ruminococcaceae_UCG-002, Ruminococcaceae_UCG-005, Clostridium_sensu_stricto_1, and Streptococcus were more abundant in controls compared to GDM patients. Bacteroides and Lachnoclostridium were more abundant in GDM patients compared to controls. Paraprevotella, Roseburia, Faecalibacterium, and Ruminococcaceae_UCG-002 were negatively correlated with glucose. Ruminococcaceae_UCG-002 was negatively correlated with HbA1c. Bacteroides was positively correlated with glucose. Sutterella, Oscillibacter, and Bifidobacterium were positively correlated with GLP-1 levels. | Paraprevotella, Roseburia, and Faecalibacterium negatively correlated with glucose. Bacteroides positively correlated with glucose. Sutterella, Oscillibacter, Bifidobacterium was positively associated with GLP-1. | At the phylum level, Firmicutes, Bacteroidetes, Proteobacteria, and Actinobacteria were the main phyla in both groups. Firmicutes and Bacteroidetes decreased and increased in GDM, respectively. At the family level, Ruminococcaceae, Lachnospiraceae, and Christensenellaceae were more abundant in NGT. Bacteroidaceae increased in GDM. At the genus level, Ruminococcaceae_UCG-002, Ruminococcaceae_UCG-005, and Clostridium_sensu_stricto_1 were lower in GDM, while Bacteroides and Lachnoclostridium were higher in GDM. |
Sun et al. [62] | Nested case–control study | 24–28 weeks of gestation | China | 120 | 120 | Fecal short-chain fatty acids like propionate and butyrate increased more from first to second trimester in controls versus GDM patient. | Microbial pathways related to dietary fiber fermentation (e.g., mannan degradation) were lower in abundance in GDM patients. | At the phylum level, the Firmicutes-to-Bacteroidetes (F/B) ratio decreased with advancing gestation in controls but not in GDM patients. Depleted in GDM: Ruminococcus bromii, Alistipes putredinis, Bacteroides ovatus, Bifidobacterium dentium. Enriched in GDM: Escherichia coli, Fusobacterium mortiferum, Bacteroides massiliensis, Eubacterium ramulus, Anaerostipes hadrus. |
Ferrocinoo et al. [46] | Prospective cohort study | 24–28 weeks of gestation, 38 weeks of gestation | Italy | 41 | 0 | Predicted metagenomic analysis suggested enrichment of pathways involved in carbohydrate metabolism and LPS biosynthesis in third trimester. | Faecalibacterium was significantly associated with fasting glucose; Collinsella (directly) and Blautia (inversely) were associated with insulin and with homeostasis model assessment of insulin resistance, while Sutterella was associated with C-reactive protein levels. Consistent with this latter association, the predicted metagenomes showed a correlation between those taxa and inferred KEGG genes associated with lipopolysaccharide biosynthesis | At the phylum level, Firmicutes increased and Bacteroidetes and Actinobacteria decreased from 2nd to third trimester. At the genus level, Blautia, Butyricicoccus, Clostridium, Coprococcus, Dorea, Faecalibacterium, L-Ruminococcus, and Lachnospiraceae increased, while Bacteroides, Collinsella and Rikenellaceae decreased. |
Kuang et al. [35] | Cross-sectional study | 21–29 weeks of gestation | china | 43 | 81 | Gut microbiota abundance correlated with glucose levels: Positively correlated genera: Parabacteroides, Megamonas, Klebsiella, etc. Negatively correlated genera: Alistipes, Bifidobacterium, Eubacterium, etc. | Inferred functional characters in GDM: Increased membrane transport, energy metabolism pathways, LPS and PTS systems; Decreased amino acid metabolism pathways. | Order: Decreased Clostridiales in GDM Family: Decreased Coriobacteriaceae in GDM Genus: Increased Parabacteroides, Megamonas, and Phascolarctobacterium in GDM; Decreased Ruminiclostridium, Roseburia, Eggerthella, Fusobacterium, Haemophilus, Mitsukella, and Aggregatibacter in GDM |
Crusell et al. [47] | Cross-sectional study | Analysis in third trimester and 8 months postpartum | Danish white pregnant women | In third trimester 50, postpartum 43 | In third trimester157, postpartum79 | Collinsella positively correlated with fasting glucose (adjusted for BMI) Butyricicoccus negatively correlated with insulin sensitivity (adjusted for BMI). Prevotella and Faecalitalea positively correlated with 2 h glucose (adjusted for BMI). | Two species of Blautia were associated with lower levels of plasma hsCRP, pointing to the occurrence of various subspecies of Blautia with opposite functionality related to host metabolism. | Phylum Actinobacteria higher in GDM. Genera Collinsella, Rothia, Desulfovibrio higher in GDM. Genera Faecalibacterium, Bacteroides, Isobaculum lower in GDM. 17 species-level OTUs differentially abundant between GDM and controls. |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Mao, L.; Gao, B.; Chang, H.; Shen, H. Interaction and Metabolic Pathways: Elucidating the Role of Gut Microbiota in Gestational Diabetes Mellitus Pathogenesis. Metabolites 2024, 14, 43. https://doi.org/10.3390/metabo14010043
Mao L, Gao B, Chang H, Shen H. Interaction and Metabolic Pathways: Elucidating the Role of Gut Microbiota in Gestational Diabetes Mellitus Pathogenesis. Metabolites. 2024; 14(1):43. https://doi.org/10.3390/metabo14010043
Chicago/Turabian StyleMao, Lindong, Biling Gao, Hao Chang, and Heqing Shen. 2024. "Interaction and Metabolic Pathways: Elucidating the Role of Gut Microbiota in Gestational Diabetes Mellitus Pathogenesis" Metabolites 14, no. 1: 43. https://doi.org/10.3390/metabo14010043
APA StyleMao, L., Gao, B., Chang, H., & Shen, H. (2024). Interaction and Metabolic Pathways: Elucidating the Role of Gut Microbiota in Gestational Diabetes Mellitus Pathogenesis. Metabolites, 14(1), 43. https://doi.org/10.3390/metabo14010043