Short-Chain Fatty-Acid-Producing Bacteria: Key Components of the Human Gut Microbiota
Abstract
:1. Introduction on Human Gut Microbiota
2. Structure and Functions of SCFAs in Physiological Conditions
3. SCFAs-Producing Bacteria of the Human Gut Microbiota
4. SCFAs Production as a Marker of Healthy Gut Ecosystem
5. Mechanistic Involvement of SCFAs in the Development of Human Diseases
5.1. SCFAs and IBD
5.2. SCFAs and Colorectal Cancer (CRC)
5.3. SCFAs and Cardiovascular Diseases (CVDs)
5.4. SCFAs and Metabolic Diseases
6. Therapeutic use of SCFAs in Clinical Practice
7. How to Foster the Production of SCFAs in the Human Gastrointestinal Tract
7.1. Diet
7.2. Prebiotics
7.3. Probiotics
7.4. Fecal Microbiota Transplantation
8. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Lozupone, C.A.; Stombaugh, J.I.; Gordon, J.I.; Jansson, J.K.; Knight, R. Diversity, stability and resilience of the human gut microbiota. Nature 2012, 489, 220–230. [Google Scholar] [CrossRef]
- Ianiro, G.; Bruno, G.; Lopetuso, L.; Beghella, F.B.; Laterza, L.; D’Aversa, F.; Gigante, G.; Cammarota, G.; Gasbarrini, A. Role of yeasts in healthy and impaired gut microbiota: The gut mycome. Curr. Pharm. Des. 2014, 20, 4565–4569. [Google Scholar] [CrossRef]
- Ianiro, G.; Iorio, A.; Porcari, S.; Masucci, L.; Sanguinetti, M.; Perno, C.F.; Gasbarrini, A.; Putignani, L.; Cammarota, G. How the gut parasitome affects human health. Ther. Adv. Gastroenterol. 2022, 15, 17562848221091524. [Google Scholar] [CrossRef]
- Siena, M.D.; Laterza, L.; Matteo, M.V.; Mignini, I.; Schepis, T.; Rizzatti, G.; Ianiro, G.; Rinninella, E.; Cintoni, M.; Gasbarrini, A. Gut and Reproductive Tract Microbiota Adaptation during Pregnancy: New Insights for Pregnancy-Related Complications and Therapy. Microorganisms 2021, 9, 473. [Google Scholar] [CrossRef]
- Arumugam, M.; Raes, J.; Pelletier, E.; Le Paslier, D.; Yamada, T.; Mende, D.R.; Fernandes, G.R.; Tap, J.; Bruls, T.; Batto, J.-M.; et al. Enterotypes of the human gut microbiome. Nature 2011, 473, 174–180. [Google Scholar] [CrossRef]
- Rath, S.; Rud, T.; Karch, A.; Pieper, D.H.; Vital, M. Pathogenic functions of host microbiota. Microbiome 2018, 6, 174. [Google Scholar] [CrossRef] [PubMed]
- Hornef, M. Pathogens, Commensal Symbionts, and Pathobionts: Discovery and Functional Effects on the Host. ILAR J. 2015, 56, 159–162. [Google Scholar] [CrossRef]
- Ianiro, G.; Molina-Infante, J.; Gasbarrini, A. Gastric Microbiota. Helicobacter 2015, 20 (Suppl. S1), 68–71. [Google Scholar] [CrossRef] [PubMed]
- Lopetuso, L.R.; Severgnini, M.; Pecere, S.; Ponziani, F.R.; Boskoski, I.; Larghi, A.; Quaranta, G.; Masucci, L.; Ianiro, G.; Camboni, T.; et al. Esophageal microbiome signature in patients with Barrett’s esophagus and esophageal adenocarcinoma. PLoS ONE 2020, 15, e0231789. [Google Scholar] [CrossRef]
- Viggiano, D.; Ianiro, G.; Vanella, G.; Bibbò, S.; Bruno, G.; Simeone, G.; Mele, G. Gut barrier in health and disease: Focus on childhood. Eur. Rev. Med. Pharmacol. Sci. 2015, 19, 1077–1085. [Google Scholar] [PubMed]
- Cardinale, V.; Capurso, G.; Ianiro, G.; Gasbarrini, A.; Arcidiacono, P.G.; Alvaro, D. Intestinal permeability changes with bacterial translocation as key events modulating systemic host immune response to SARS-CoV-2: A working hypothesis. Dig. Liver Dis. Off. J. Ital. Soc. Gastroenterol. Ital. Assoc. Study Liver 2020, 52, 1383–1389. [Google Scholar] [CrossRef] [PubMed]
- Cianci, R.; Franza, L.; Schinzari, G.; Rossi, E.; Ianiro, G.; Tortora, G.; Gasbarrini, A.; Gambassi, G.; Cammarota, G. The Interplay between Immunity and Microbiota at Intestinal Immunological Niche: The Case of Cancer. Int. J. Mol. Sci. 2019, 20, 501. [Google Scholar] [CrossRef] [PubMed]
- Bibbò, S.; Lopetuso, L.R.; Ianiro, G.; Di Rienzo, T.; Gasbarrini, A.; Cammarota, G. Role of microbiota and innate immunity in recurrent Clostridium difficile infection. J. Immunol. Res. 2014, 2014, 462740. [Google Scholar] [CrossRef] [PubMed]
- D’Aversa, F.; Tortora, A.; Ianiro, G.; Ponziani, F.R.; Annicchiarico, B.E.; Gasbarrini, A. Gut microbiota and metabolic syndrome. Intern. Emerg. Med. 2013, 8 (Suppl. S1), S11–S15. [Google Scholar] [CrossRef]
- Ianiro, G.; Mangiola, F.; Di Rienzo, T.A.; Bibbò, S.; Franceschi, F.; Greco, A.V.; Gasbarrini, A. Levothyroxine absorption in health and disease, and new therapeutic perspectives. Eur. Rev. Med. Pharmacol. Sci. 2014, 18, 451–456. [Google Scholar]
- Valdes, A.M.; Walter, J.; Segal, E.; Spector, T.D. Role of the gut microbiota in nutrition and health. BMJ 2018, 361, k2179. [Google Scholar] [CrossRef]
- Lin, D.; Medeiros, D.M. The microbiome as a major function of the gastrointestinal tract and its implication in micronutrient metabolism and chronic diseases. Nutr. Res. 2023, 112, 30–45. [Google Scholar] [CrossRef]
- Settanni, C.R.; Ianiro, G.; Bibbò, S.; Cammarota, G.; Gasbarrini, A. Gut microbiota alteration and modulation in psychiatric disorders: Current evidence on fecal microbiota transplantation. Prog. Neuropsychopharmacol. Biol. Psychiatry 2021, 109, 110258. [Google Scholar] [CrossRef]
- Valles-Colomer, M.; Menni, C.; Berry, S.E.; Valdes, A.M.; Spector, T.D.; Segata, N. Cardiometabolic health, diet and the gut microbiome: A meta-omics perspective. Nat. Med. 2023, 29, 551–561. [Google Scholar]
- Rinninella, E.; Tohumcu, E.; Raoul, P.; Fiorani, M.; Cintoni, M.; Mele, M.C.; Cammarota, G.; Gasbarrini, A.; Ianiro, G. The role of diet in shaping human gut microbiota. Best Pract. Res. Clin. Gastroenterol. 2023, 62–63, 101828. [Google Scholar] [CrossRef]
- Dumitrascu, D.L.; Bakulin, I.; Berzigotti, A.; Cravo, M.; Gombošová, L.; Lukas, M.; Pietrzak, A.; Remes-Troche, J.M.; Romero-Gómez, M.; Balmori, M.A.; et al. Update on the Role of Rifaximin in Digestive Diseases. J. Gastrointest. Liver Dis. 2023, 32, 92–109. [Google Scholar] [CrossRef]
- Nambiar, R.B.; Perumal, A.B.; Shittu, T.; Sadiku, E.R.; Sellamuthu, P.S. Editorial: Probiotics, prebiotics, synbiotics, postbiotics, & paraprobiotics—New perspective for functional foods and nutraceuticals. Front. Nutr. 2023, 10, 1164676. [Google Scholar] [PubMed]
- Mosca, A.; Abreu, Y.; Abreu, A.T.; Gwee, K.A.; Ianiro, G.; Tack, J.; Nguyen, T.V.H.; Hill, C. The clinical evidence for postbiotics as microbial therapeutics. Gut Microbes 2022, 14, 2117508. [Google Scholar] [CrossRef] [PubMed]
- Baunwall, S.M.D.; Terveer, E.M.; Dahlerup, J.F.; Erikstrup, C.; Arkkila, P.; Vehreschild, M.J.; Ianiro, G.; Gasbarrini, A.; Sokol, H.; Kump, P.K.; et al. The use of Faecal Microbiota Transplantation (FMT) in Europe: A Europe-wide survey. Lancet Reg. Health Eur. 2021, 9, 100181. [Google Scholar] [CrossRef]
- Mortensen, P.B.; Clausen, M.R. Short-chain fatty acids in the human colon: Relation to gastrointestinal health and disease. Scand. J. Gastroenterol. Suppl. 1996, 216, 132–148. [Google Scholar] [CrossRef]
- Topping, D.L.; Clifton, P.M. Short-chain fatty acids and human colonic function: Roles of resistant starch and nonstarch polysaccharides. Physiol. Rev. 2001, 81, 1031–1064. [Google Scholar] [CrossRef] [PubMed]
- Topping, D.L. Short-chain fatty acids produced by intestinal bacteria. Asia Pac. J. Clin. Nutr. 1996, 5, 15–19. [Google Scholar]
- Kimura, I.; Ichimura, A.; Ohue-Kitano, R.; Igarashi, M. Free Fatty Acid Receptors in Health and Disease. Physiol. Rev. 2020, 100, 171–210. [Google Scholar] [CrossRef]
- Bolognini, D.; Tobin, A.B.; Milligan, G.; Moss, C.E. The Pharmacology and Function of Receptors for Short-Chain Fatty Acids. Mol. Pharmacol. 2016, 89, 388–398. [Google Scholar] [CrossRef]
- Kimura, I.; Inoue, D.; Maeda, T.; Hara, T.; Ichimura, A.; Miyauchi, S.; Kobayashi, M.; Hirasawa, A.; Tsujimoto, G. Short-chain fatty acids and ketones directly regulate sympathetic nervous system via G protein-coupled receptor 41 (GPR41). Proc. Natl. Acad. Sci. USA 2011, 108, 8030–8035. [Google Scholar] [CrossRef]
- Priyadarshini, M.; Layden, B.T. FFAR3 modulates insulin secretion and global gene expression in mouse islets. Islets 2015, 7, e1045182. [Google Scholar] [CrossRef] [PubMed]
- Priyadarshini, M.; Villa, S.R.; Fuller, M.; Wicksteed, B.; Mackay, C.R.; Alquier, T.; Poitout, V.; Mancebo, H.; Mirmira, R.G.; Gilchrist, A.; et al. An Acetate-Specific GPCR, FFAR2, Regulates Insulin Secretion. Mol. Endocrinol. Baltim. Md. 2015, 29, 1055–1066. [Google Scholar] [CrossRef]
- Tolhurst, G.; Heffron, H.; Lam, Y.S.; Parker, H.E.; Habib, A.M.; Diakogiannaki, E.; Cameron, J.; Grosse, J.; Reimann, F.; Gribble, F.M. Short-chain fatty acids stimulate glucagon-like peptide-1 secretion via the G-protein-coupled receptor FFAR2. Diabetes 2012, 61, 364–371. [Google Scholar] [CrossRef] [PubMed]
- Akiba, Y.; Inoue, T.; Kaji, I.; Higashiyama, M.; Narimatsu, K.; Iwamoto, K.; Watanabe, M.; Guth, P.H.; Engel, E.; Kuwahara, A.; et al. Short-chain Fatty Acid Sensing in Rat Duodenum. J. Physiol. 2015, 593, 585–599. [Google Scholar] [CrossRef]
- Sivaprakasam, S.; Bhutia, Y.D.; Yang, S.; Ganapathy, V. Short-Chain Fatty Acid Transporters: Role in Colonic Homeostasis. Compr. Physiol. 2017, 8, 299–314. [Google Scholar] [PubMed]
- Chen, G.; Ran, X.; Li, B.; Li, Y.; He, D.; Huang, B.; Fu, S.; Liu, J.; Wang, W. Sodium Butyrate Inhibits Inflammation and Maintains Epithelium Barrier Integrity in a TNBS-induced Inflammatory Bowel Disease Mice Model. EBioMedicine 2018, 30, 317–325. [Google Scholar] [CrossRef] [PubMed]
- Liang, L.; Liu, L.; Zhou, W.; Yang, C.; Mai, G.; Li, H.; Chen, Y. Gut microbiota-derived butyrate regulates gut mucus barrier repair by activating the macrophage/WNT/ERK signaling pathway. Clin. Sci. 2022, 136, 291–307. [Google Scholar] [CrossRef]
- Liu, P.; Wang, Y.; Yang, G.; Zhang, Q.; Meng, L.; Xin, Y.; Jiang, X. The role of short-chain fatty acids in intestinal barrier function, inflammation, oxidative stress, and colonic carcinogenesis. Pharmacol. Res. 2021, 165, 105420. [Google Scholar] [CrossRef]
- De Vadder, F.; Kovatcheva-Datchary, P.; Goncalves, D.; Vinera, J.; Zitoun, C.; Duchampt, A.; Bäckhed, F.; Mithieux, G. Microbiota-Generated Metabolites Promote Metabolic Benefits via Gut-Brain Neural Circuits. Cell 2014, 156, 84–96. [Google Scholar] [CrossRef]
- Mirzaei, R.; Bouzari, B.; Hosseini-Fard, S.R.; Mazaheri, M.; Ahmadyousefi, Y.; Abdi, M.; Jalalifar, S.; Karimitabar, Z.; Teimoori, A.; Keyvani, H.; et al. Role of microbiota-derived short-chain fatty acids in nervous system disorders. Biomed. Pharmacother. Biomed. Pharmacother. 2021, 139, 111661. [Google Scholar] [CrossRef]
- Yang, L.L.; Millischer, V.; Rodin, S.; MacFabe, D.F.; Villaescusa, J.C.; Lavebratt, C. Enteric short-chain fatty acids promote proliferation of human neural progenitor cells. J. Neurochem. 2020, 154, 635–646. [Google Scholar] [CrossRef] [PubMed]
- Nankova, B.B.; Agarwal, R.; MacFabe, D.F.; La Gamma, E.F. Enteric bacterial metabolites propionic and butyric acid modulate gene expression, including CREB-dependent catecholaminergic neurotransmission, in PC12 cells—Possible relevance to autism spectrum disorders. PLoS ONE 2014, 9, e103740. [Google Scholar] [CrossRef] [PubMed]
- Hoyles, L.; Snelling, T.; Umlai, U.-K.; Nicholson, J.K.; Carding, S.R.; Glen, R.C.; McArthur, S. Microbiome–host systems interactions: Protective effects of propionate upon the blood–brain barrier. Microbiome 2018, 6, 55. [Google Scholar] [CrossRef] [PubMed]
- Dalile, B.; Van Oudenhove, L.; Vervliet, B.; Verbeke, K. The role of short-chain fatty acids in microbiota-gut-brain communication. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 461–478. [Google Scholar] [CrossRef] [PubMed]
- Frost, G.; Sleeth, M.L.; Sahuri-Arisoylu, M.; Lizarbe, B.; Cerdan, S.; Brody, L.; Anastasovska, J.; Ghourab, S.; Hankir, M.; Zhang, S.; et al. The short-chain fatty acid acetate reduces appetite via a central homeostatic mechanism. Nat. Commun. 2014, 5, 3611. [Google Scholar] [CrossRef]
- González Hernández, M.A.; Canfora, E.E.; Jocken, J.W.E.; Blaak, E.E. The Short-Chain Fatty Acid Acetate in Body Weight Control and Insulin Sensitivity. Nutrients 2019, 11, 1943. [Google Scholar] [CrossRef]
- Yoshida, H.; Ishii, M.; Akagawa, M. Propionate suppresses hepatic gluconeogenesis via GPR43/AMPK signaling pathway. Arch. Biochem. Biophys. 2019, 672, 108057. [Google Scholar] [CrossRef]
- den Besten, G.; Bleeker, A.; Gerding, A.; van Eunen, K.; Havinga, R.; van Dijk, T.H.; Oosterveer, M.H.; Jonker, J.W.; Groen, A.K.; Reijngoud, D.-J.; et al. Short-Chain Fatty Acids Protect Against High-Fat Diet-Induced Obesity via a PPARγ-Dependent Switch From Lipogenesis to Fat Oxidation. Diabetes 2015, 64, 2398–2408. [Google Scholar] [CrossRef]
- Weitkunat, K.; Schumann, S.; Nickel, D.; Kappo, K.A.; Petzke, K.J.; Kipp, A.P.; Blaut, M.; Klaus, S. Importance of propionate for the repression of hepatic lipogenesis and improvement of insulin sensitivity in high-fat diet-induced obesity. Mol. Nutr. Food Res. 2016, 60, 2611–2621. [Google Scholar] [CrossRef]
- Xiong, Y.; Miyamoto, N.; Shibata, K.; Valasek, M.A.; Motoike, T.; Kedzierski, R.M.; Yanagisawa, M. Short-chain fatty acids stimulate leptin production in adipocytes through the G protein-coupled receptor GPR41. Proc. Natl. Acad. Sci. USA 2004, 101, 1045–1050. [Google Scholar] [CrossRef]
- Soliman, M.M.; Ahmed, M.M.; Salah-Eldin, A.-E.; Abdel-Aal, A.A.-A. Butyrate regulates leptin expression through different signaling pathways in adipocytes. J. Vet. Sci. 2011, 12, 319–323. [Google Scholar] [CrossRef] [PubMed]
- Deng, M.; Qu, F.; Chen, L.; Liu, C.; Zhang, M.; Ren, F.; Guo, H.; Zhang, H.; Ge, S.; Wu, C.; et al. SCFAs alleviated steatosis and inflammation in mice with NASH induced by MCD. J. Endocrinol. 2020, 245, 425–437. [Google Scholar] [CrossRef] [PubMed]
- Shimizu, H.; Masujima, Y.; Ushiroda, C.; Mizushima, R.; Taira, S.; Ohue-Kitano, R.; Kimura, I. Dietary short-chain fatty acid intake improves the hepatic metabolic condition via FFAR3. Sci. Rep. 2019, 9, 16574. [Google Scholar] [CrossRef]
- Kondo, T.; Kishi, M.; Fushimi, T.; Ugajin, S.; Kaga, T. Vinegar intake reduces body weight, body fat mass, and serum triglyceride levels in obese Japanese subjects. Biosci. Biotechnol. Biochem. 2009, 73, 1837–1843. [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]
- Hu, T.; Wu, Q.; Yao, Q.; Jiang, K.; Yu, J.; Tang, Q. Short-chain fatty acid metabolism and multiple effects on cardiovascular diseases. Ageing Res. Rev. 2022, 81, 101706. [Google Scholar] [CrossRef]
- Mortensen, F.V.; Jørgensen, B.; Christiansen, H.M.; Sloth-Nielsen, J.; Wolff, B.; Hessov, I. Short-chain fatty acid enemas stimulate plasminogen activator inhibitor-1 after abdominal aortic graft surgery: A double-blinded, placebo-controlled study. Thromb. Res. 2000, 98, 361–366. [Google Scholar] [CrossRef]
- Maslowski, K.M.; Vieira, A.T.; Ng, A.; Kranich, J.; Sierro, F.; Yu, D.; Schilter, H.C.; Rolph, M.S.; Mackay, F.; Artis, D.; et al. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature 2009, 461, 1282–1286. [Google Scholar] [CrossRef]
- Zapolska-Downar, D.; Naruszewicz, M. Propionate reduces the cytokine-induced VCAM-1 and ICAM-1 expression by inhibiting nuclear factor-kappa B (NF-kappaB) activation. J. Physiol. Pharmacol. Off. J. Pol. Physiol. Soc. 2009, 60, 123–131. [Google Scholar]
- Cox, M.A.; Jackson, J.; Stanton, M.; Rojas-Triana, A.; Bober, L.; Laverty, M.; Yang, X.; Zhu, F.; Liu, J.; Wang, S.; et al. Short-chain fatty acids act as antiinflammatory mediators by regulating prostaglandin E(2) and cytokines. World J. Gastroenterol. 2009, 15, 5549–5557. [Google Scholar] [CrossRef]
- Lucas, J.L.; Mirshahpanah, P.; Haas-Stapleton, E.; Asadullah, K.; Zollner, T.M.; Numerof, R.P. Induction of Foxp3+ regulatory T cells with histone deacetylase inhibitors. Cell. Immunol. 2009, 257, 97–104. [Google Scholar] [CrossRef] [PubMed]
- n-Butyrate Anergized Effector CD4+ T Cells Independent of Regulatory T cell Generation or Activity—Fontenelle—2012—Scandinavian Journal of Immunology—Wiley Online Library. Available online: https://onlinelibrary.wiley.com/doi/10.1111/j.1365-3083.2012.02740.x (accessed on 9 April 2023).
- Silva, Y.P.; Bernardi, A.; Frozza, R.L. The Role of Short-Chain Fatty Acids From Gut Microbiota in Gut-Brain Communication. Front. Endocrinol. 2020, 11, 25. [Google Scholar] [CrossRef] [PubMed]
- Selma-Royo, M.; Tarrazó, M.; García-Mantrana, I.; Gómez-Gallego, C.; Salminen, S.; Collado, M.C. Shaping Microbiota During the First 1000 Days of Life. Adv. Exp. Med. Biol. 2019, 1125, 3–24. [Google Scholar] [PubMed]
- Garcia-Mantrana, I.; Selma-Royo, M.; Alcantara, C.; Collado, M.C. Shifts on Gut Microbiota Associated to Mediterranean Diet Adherence and Specific Dietary Intakes on General Adult Population. Front. Microbiol. 2018, 9, 890. [Google Scholar] [CrossRef] [PubMed]
- Wong, J.M.W.; de Souza, R.; Kendall, C.W.C.; Emam, A.; Jenkins, D.J.A. Colonic health: Fermentation and short chain fatty acids. J. Clin. Gastroenterol. 2006, 40, 235–243. [Google Scholar] [CrossRef] [PubMed]
- Odamaki, T.; Kato, K.; Sugahara, H.; Hashikura, N.; Takahashi, S.; Xiao, J.-Z.; Abe, F.; Osawa, R. Age-related changes in gut microbiota composition from newborn to centenarian: A cross-sectional study. BMC Microbiol. 2016, 16, 90. [Google Scholar] [CrossRef]
- Tsukuda, N.; Yahagi, K.; Hara, T.; Watanabe, Y.; Matsumoto, H.; Mori, H.; Higashi, K.; Tsuji, H.; Matsumoto, S.; Kurokawa, K.; et al. Key bacterial taxa and metabolic pathways affecting gut short-chain fatty acid profiles in early life. ISME J. 2021, 15, 2574–2590. [Google Scholar] [CrossRef]
- Biddle, A.; Stewart, L.; Blanchard, J.; Leschine, S. Untangling the Genetic Basis of Fibrolytic Specialization by Lachnospiraceae and Ruminococcaceae in Diverse Gut Communities. Diversity 2013, 5, 627–640. [Google Scholar] [CrossRef]
- Herrmann, E.; Young, W.; Reichert-Grimm, V.; Weis, S.; Riedel, C.U.; Rosendale, D.; Stoklosinski, H.; Hunt, M.; Egert, M. In Vivo Assessment of Resistant Starch Degradation by the Caecal Microbiota of Mice Using RNA-Based Stable Isotope Probing-A Proof-of-Principle Study. Nutrients 2018, 10, 179. [Google Scholar] [CrossRef]
- Ghosh, T.S.; Shanahan, F.; O’Toole, P.W. The gut microbiome as a modulator of healthy ageing. Nat. Rev. Gastroenterol. Hepatol. 2022, 19, 565–584. [Google Scholar] [CrossRef]
- Rios-Covian, D.; Gueimonde, M.; Duncan, S.H.; Flint, H.J.; de los Reyes-Gavilan, C.G. Enhanced butyrate formation by cross-feeding between Faecalibacterium prausnitzii and Bifidobacterium adolescentis. FEMS Microbiol. Lett. 2015, 362, fnv176. [Google Scholar] [CrossRef]
- Rodríguez-Carrio, J.; Salazar, N.; Margolles, A.; González, S.; Gueimonde, M.; de Los Reyes-Gavilán, C.G.; Suárez, A. Free Fatty Acids Profiles Are Related to Gut Microbiota Signatures and Short-Chain Fatty Acids. Front. Immunol. 2017, 8, 823. [Google Scholar] [CrossRef]
- Matsuki, T.; Yahagi, K.; Mori, H.; Matsumoto, H.; Hara, T.; Tajima, S.; Ogawa, E.; Kodama, H.; Yamamoto, K.; Yamada, T.; et al. A key genetic factor for fucosyllactose utilization affects infant gut microbiota development. Nat. Commun. 2016, 7, 11939. [Google Scholar] [CrossRef]
- Sela, D.A.; Mills, D.A. Nursing our microbiota: Molecular linkages between bifidobacteria and milk oligosaccharides. Trends Microbiol. 2010, 18, 298–307. [Google Scholar] [CrossRef]
- Hugenholtz, F.; Mullaney, J.A.; Kleerebezem, M.; Smidt, H.; Rosendale, D.I. Modulation of the microbial fermentation in the gut by fermentable carbohydrates. Bioact. Carbohydr. Diet. Fibre 2013, 2, 133–142. [Google Scholar] [CrossRef]
- Walsh, C.; Lane, J.A.; van Sinderen, D.; Hickey, R.M. Human milk oligosaccharide-sharing by a consortium of infant derived Bifidobacterium species. Sci. Rep. 2022, 12, 4143. [Google Scholar] [CrossRef]
- Henrick, B.M.; Rodriguez, L.; Lakshmikanth, T.; Pou, C.; Henckel, E.; Arzoomand, A.; Olin, A.; Wang, J.; Mikes, J.; Tan, Z.; et al. Bifidobacteria-mediated immune system imprinting early in life. Cell 2021, 184, 3884–3898.e11. [Google Scholar] [CrossRef] [PubMed]
- Saturio, S.; Nogacka, A.M.; Alvarado-Jasso, G.M.; Salazar, N.; de Los Reyes-Gavilán, C.G.; Gueimonde, M.; Arboleya, S. Role of Bifidobacteria on Infant Health. Microorganisms 2021, 9, 2415. [Google Scholar] [CrossRef] [PubMed]
- Fukuda, S.; Toh, H.; Taylor, T.D.; Ohno, H.; Hattori, M. Acetate-producing bifidobacteria protect the host from enteropathogenic infection via carbohydrate transporters. Gut Microbes 2012, 3, 449–454. [Google Scholar] [CrossRef] [PubMed]
- Deleu, S.; Machiels, K.; Raes, J.; Verbeke, K.; Vermeire, S. Short chain fatty acids and its producing organisms: An overlooked therapy for IBD? EBioMedicine 2021, 66, 103293. [Google Scholar] [CrossRef] [PubMed]
- Ragsdale, S.W.; Pierce, E. Acetogenesis and the Wood-Ljungdahl pathway of CO(2) fixation. Biochim. Biophys. Acta 2008, 1784, 1873–1898. [Google Scholar] [CrossRef] [PubMed]
- Schug, Z.T.; Vande Voorde, J.; Gottlieb, E. The metabolic fate of acetate in cancer. Nat. Rev. Cancer 2016, 16, 708–717. [Google Scholar] [CrossRef] [PubMed]
- Conlon, M.A.; Bird, A.R. The impact of diet and lifestyle on gut microbiota and human health. Nutrients 2014, 7, 17–44. [Google Scholar] [CrossRef] [PubMed]
- Reichardt, N.; Duncan, S.H.; Young, P.; Belenguer, A.; McWilliam Leitch, C.; Scott, K.P.; Flint, H.J.; Louis, P. Phylogenetic distribution of three pathways for propionate production within the human gut microbiota. ISME J. 2014, 8, 1323–1335. [Google Scholar] [CrossRef]
- De Vadder, F.; Kovatcheva-Datchary, P.; Zitoun, C.; Duchampt, A.; Bäckhed, F.; Mithieux, G. Microbiota-Produced Succinate Improves Glucose Homeostasis via Intestinal Gluconeogenesis. Cell Metab. 2016, 24, 151–157. [Google Scholar] [CrossRef]
- Salonen, A.; Lahti, L.; Salojärvi, J.; Holtrop, G.; Korpela, K.; Duncan, S.H.; Date, P.; Farquharson, F.; Johnstone, A.M.; Lobley, G.E.; et al. Impact of diet and individual variation on intestinal microbiota composition and fermentation products in obese men. ISME J. 2014, 8, 2218–2230. [Google Scholar] [CrossRef]
- Watanabe, Y.; Nagai, F.; Morotomi, M. Characterization of Phascolarctobacterium succinatutens sp. nov., an asaccharolytic, succinate-utilizing bacterium isolated from human feces. Appl. Environ. Microbiol. 2012, 78, 511–518. [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]
- Scott, K.P.; Martin, J.C.; Campbell, G.; Mayer, C.-D.; Flint, H.J. Whole-genome transcription profiling reveals genes up-regulated by growth on fucose in the human gut bacterium “Roseburia inulinivorans”. J. Bacteriol. 2006, 188, 4340–4349. [Google Scholar] [CrossRef]
- Litvak, Y.; Byndloss, M.X.; Bäumler, A.J. Colonocyte metabolism shapes the gut microbiota. Science 2018, 362, eaat9076. [Google Scholar] [CrossRef]
- Manson, J.M.; Rauch, M.; Gilmore, M.S. The commensal microbiology of the gastrointestinal tract. Adv. Exp. Med. Biol. 2008, 635, 15–28. [Google Scholar] [PubMed]
- Rivera-Chávez, F.; Zhang, L.F.; Faber, F.; Lopez, C.A.; Byndloss, M.X.; Olsan, E.E.; Xu, G.; Velazquez, E.M.; Lebrilla, C.B.; Winter, S.E.; et al. Depletion of Butyrate-Producing Clostridia from the Gut Microbiota Drives an Aerobic Luminal Expansion of Salmonella. Cell Host Microbe 2016, 19, 443–454. [Google Scholar] [CrossRef]
- Parada Venegas, D.; De la Fuente, M.K.; Landskron, G.; González, M.J.; Quera, R.; Dijkstra, G.; Harmsen, H.J.M.; Faber, K.N.; Hermoso, M.A. Short Chain Fatty Acids (SCFAs)-Mediated Gut Epithelial and Immune Regulation and Its Relevance for Inflammatory Bowel Diseases. Front. Immunol. 2019, 10, 277. [Google Scholar] [CrossRef]
- Singh, V.; Lee, G.; Son, H.; Koh, H.; Kim, E.S.; Unno, T.; Shin, J.-H. Butyrate producers, “The Sentinel of Gut”: Their intestinal significance with and beyond butyrate, and prospective use as microbial therapeutics. Front. Microbiol. 2022, 13, 1103836. [Google Scholar] [CrossRef]
- Vacca, M.; Celano, G.; Calabrese, F.M.; Portincasa, P.; Gobbetti, M.; De Angelis, M. The Controversial Role of Human Gut Lachnospiraceae. Microorganisms 2020, 8, 573. [Google Scholar] [CrossRef] [PubMed]
- Maturana, J.L.; Cárdenas, J.P. Insights on the Evolutionary Genomics of the Blautia Genus: Potential New Species and Genetic Content Among Lineages. Front. Microbiol. 2021, 12, 660920. [Google Scholar] [CrossRef] [PubMed]
- Nie, K.; Ma, K.; Luo, W.; Shen, Z.; Yang, Z.; Xiao, M.; Tong, T.; Yang, Y.; Wang, X. Roseburia intestinalis: A Beneficial Gut Organism From the Discoveries in Genus and Species. Front. Cell. Infect. Microbiol. 2021, 11, 757718. [Google Scholar] [CrossRef] [PubMed]
- Louis, P.; Young, P.; Holtrop, G.; Flint, H.J. Diversity of human colonic butyrate-producing bacteria revealed by analysis of the butyryl-CoA:acetate CoA-transferase gene. Environ. Microbiol. 2010, 12, 304–314. [Google Scholar] [CrossRef]
- Chassard, C.; Bernalier-Donadille, A. H2 and acetate transfers during xylan fermentation between a butyrate-producing xylanolytic species and hydrogenotrophic microorganisms from the human gut. FEMS Microbiol. Lett. 2006, 254, 116–122. [Google Scholar] [CrossRef]
- Schwab, C.; Ruscheweyh, H.-J.; Bunesova, V.; Pham, V.T.; Beerenwinkel, N.; Lacroix, C. Trophic Interactions of Infant Bifidobacteria and Eubacterium hallii during L-Fucose and Fucosyllactose Degradation. Front. Microbiol. 2017, 8, 95. [Google Scholar] [CrossRef]
- Martín, R.; Bermúdez-Humarán, L.G.; Langella, P. Searching for the Bacterial Effector: The Example of the Multi-Skilled Commensal Bacterium Faecalibacterium prausnitzii. Front. Microbiol. 2018, 9, 346. [Google Scholar] [CrossRef] [PubMed]
- Miquel, S.; Martín, R.; Rossi, O.; Bermúdez-Humarán, L.G.; Chatel, J.M.; Sokol, H.; Thomas, M.; Wells, J.M.; Langella, P. Faecalibacterium prausnitzii and human intestinal health. Curr. Opin. Microbiol. 2013, 16, 255–261. [Google Scholar] [CrossRef] [PubMed]
- Machiels, K.; Joossens, M.; Sabino, J.; De Preter, V.; Arijs, I.; Eeckhaut, V.; Ballet, V.; Claes, K.; Van Immerseel, F.; Verbeke, K.; et al. A decrease of the butyrate-producing species Roseburia hominis and Faecalibacterium prausnitzii defines dysbiosis in patients with ulcerative colitis. Gut 2014, 63, 1275–1283. [Google Scholar] [CrossRef] [PubMed]
- Franzosa, E.A.; Sirota-Madi, A.; Avila-Pacheco, J.; Fornelos, N.; Haiser, H.J.; Reinker, S.; Vatanen, T.; Hall, A.B.; Mallick, H.; McIver, L.J.; et al. Gut microbiome structure and metabolic activity in inflammatory bowel disease. Nat. Microbiol. 2019, 4, 293–305. [Google Scholar] [CrossRef] [PubMed]
- Anand, S.; Kaur, H.; Mande, S.S. Comparative In silico Analysis of Butyrate Production Pathways in Gut Commensals and Pathogens. Front. Microbiol. 2016, 7, 1945. [Google Scholar] [CrossRef]
- Vital, M.; Howe, A.C.; Tiedje, J.M. Revealing the bacterial butyrate synthesis pathways by analyzing (meta)genomic data. mBio 2014, 5, e00889. [Google Scholar] [CrossRef]
- Rasmussen, H.S.; Holtug, K.; Mortensen, P.B. Degradation of amino acids to short-chain fatty acids in humans. An in vitro study. Scand. J. Gastroenterol. 1988, 23, 178–182. [Google Scholar] [CrossRef]
- Macfarlane, G.T.; Gibson, G.R.; Beatty, E.; Cummings, J.H. Estimation of short-chain fatty acid production from protein by human intestinal bacteria based on branched-chain fatty acid measurements. FEMS Microbiol. Lett. 1992, 101, 81–88. [Google Scholar] [CrossRef]
- Özcan, E.; Sela, D.A. Inefficient Metabolism of the Human Milk Oligosaccharides Lacto-N-tetraose and Lacto-N-neotetraose Shifts Bifidobacterium longum subsp. infantis Physiology. Front. Nutr. 2018, 5, 46. [Google Scholar] [CrossRef]
- Engels, C.; Ruscheweyh, H.-J.; Beerenwinkel, N.; Lacroix, C.; Schwab, C. The Common Gut Microbe Eubacterium hallii also Contributes to Intestinal Propionate Formation. Front. Microbiol. 2016, 7, 713. [Google Scholar] [CrossRef]
- Jost, T.; Lacroix, C.; Braegger, C.P.; Chassard, C. New insights in gut microbiota establishment in healthy breast fed neonates. PLoS ONE 2012, 7, e44595. [Google Scholar] [CrossRef] [PubMed]
- Pham, V.T.; Lacroix, C.; Braegger, C.P.; Chassard, C. Early colonization of functional groups of microbes in the infant gut. Environ. Microbiol. 2016, 18, 2246–2258. [Google Scholar] [CrossRef] [PubMed]
- Donohoe, D.R.; Garge, N.; Zhang, X.; Sun, W.; O’Connell, T.M.; Bunger, M.K.; Bultman, S.J. The microbiome and butyrate regulate energy metabolism and autophagy in the mammalian colon. Cell Metab. 2011, 13, 517–526. [Google Scholar] [CrossRef] [PubMed]
- Paone, P.; Cani, P.D. Mucus barrier, mucins and gut microbiota: The expected slimy partners? Gut 2020, 69, 2232–2243. [Google Scholar] [CrossRef]
- Geerlings, S.Y.; Kostopoulos, I.; de Vos, W.M.; Belzer, C. Akkermansia muciniphila in the Human Gastrointestinal Tract: When, Where, and How? Microorganisms 2018, 6, 75. [Google Scholar] [CrossRef] [PubMed]
- Belzer, C.; Chia, L.W.; Aalvink, S.; Chamlagain, B.; Piironen, V.; Knol, J.; de Vos, W.M. Microbial Metabolic Networks at the Mucus Layer Lead to Diet-Independent Butyrate and Vitamin B12 Production by Intestinal Symbionts. mBio 2017, 8, e00770-e17. [Google Scholar] [CrossRef]
- Gaudier, E.; Rival, M.; Buisine, M.-P.; Robineau, I.; Hoebler, C. Butyrate enemas upregulate Muc genes expression but decrease adherent mucus thickness in mice colon. Physiol. Res. 2009, 58, 111–119. [Google Scholar] [CrossRef]
- Blaak, E.E.; Canfora, E.E.; Theis, S.; Frost, G.; Groen, A.K.; Mithieux, G.; Nauta, A.; Scott, K.; Stahl, B.; van Harsselaar, J.; et al. Short chain fatty acids in human gut and metabolic health. Benef. Microbes 2020, 11, 411–455. [Google Scholar] [CrossRef]
- Yao, Y.; Kim, G.; Shafer, S.; Chen, Z.; Kubo, S.; Ji, Y.; Luo, J.; Yang, W.; Perner, S.P.; Kanellopoulou, C.; et al. Mucus sialylation determines intestinal host-commensal homeostasis. Cell 2022, 185, 1172–1188.e28. [Google Scholar] [CrossRef]
- Jung, T.-H.; Park, J.H.; Jeon, W.-M.; Han, K.-S. Butyrate modulates bacterial adherence on LS174T human colorectal cells by stimulating mucin secretion and MAPK signaling pathway. Nutr. Res. Pract. 2015, 9, 343–349. [Google Scholar] [CrossRef]
- Johansson, M.E.V.; Sjövall, H.; Hansson, G.C. The gastrointestinal mucus system in health and disease. Nat. Rev. Gastroenterol. Hepatol. 2013, 10, 352–361. [Google Scholar] [CrossRef] [PubMed]
- Fang, J.; Wang, H.; Zhou, Y.; Zhang, H.; Zhou, H.; Zhang, X. Slimy partners: The mucus barrier and gut microbiome in ulcerative colitis. Exp. Mol. Med. 2021, 53, 772–787. [Google Scholar] [CrossRef] [PubMed]
- Tang, Y.; Chen, Y.; Jiang, H.; Robbins, G.T.; Nie, D. G-protein-coupled receptor for short-chain fatty acids suppresses colon cancer. Int. J. Cancer 2011, 128, 847–856. [Google Scholar] [CrossRef] [PubMed]
- Yang, T.; Santisteban, M.M.; Rodriguez, V.; Li, E.; Ahmari, N.; Carvajal, J.M.; Zadeh, M.; Gong, M.; Qi, Y.; Zubcevic, J.; et al. Gut dysbiosis is linked to hypertension. Hypertension 2015, 65, 1331–1340. [Google Scholar] [CrossRef]
- Ley, R.E.; Bäckhed, F.; Turnbaugh, P.; Lozupone, C.A.; Knight, R.D.; Gordon, J.I. Obesity alters gut microbial ecology. Proc. Natl. Acad. Sci. USA 2005, 102, 11070–11075. [Google Scholar] [CrossRef]
- Medzhitov, R. Inflammation 2010: New adventures of an old flame. Cell 2010, 140, 771–776. [Google Scholar] [CrossRef] [PubMed]
- Chen, L.; Deng, H.; Cui, H.; Fang, J.; Zuo, Z.; Deng, J.; Li, Y.; Wang, X.; Zhao, L. Inflammatory responses and inflammation-associated diseases in organs. Oncotarget 2017, 9, 7204–7218. [Google Scholar] [CrossRef]
- Kuriakose, T.; Kanneganti, T.-D. Pyroptosis in Antiviral Immunity. Curr. Top. Microbiol. Immunol. 2019, 13, 1–19. [Google Scholar] [CrossRef]
- Zhang, G.; Wang, J.; Zhao, Z.; Xin, T.; Fan, X.; Shen, Q.; Raheem, A.; Lee, C.R.; Jiang, H. Regulated necrosis, a proinflammatory cell death, potentially counteracts pathogenic infections. Cell Death Dis. 2022, 13, 637. [Google Scholar] [CrossRef]
- Patankar, J.V.; Becker, C. Cell death in the gut epithelium and implications for chronic inflammation. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 543–556. [Google Scholar] [CrossRef] [PubMed]
- Pascal, V.; Pozuelo, M.; Borruel, N.; Casellas, F.; Campos, D.; Santiago, A.; Martinez, X.; Varela, E.; Sarrabayrouse, G.; Machiels, K.; et al. A microbial signature for Crohn’s disease. Gut 2017, 66, 813–822. [Google Scholar] [CrossRef]
- De Preter, V.; Arijs, I.; Windey, K.; Vanhove, W.; Vermeire, S.; Schuit, F.; Rutgeerts, P.; Verbeke, K. Impaired butyrate oxidation in ulcerative colitis is due to decreased butyrate uptake and a defect in the oxidation pathway. Inflamm. Bowel Dis. 2012, 18, 1127–1136. [Google Scholar] [CrossRef]
- Singh, N.; Gurav, A.; Sivaprakasam, S.; Brady, E.; Padia, R.; Shi, H.; Thangaraju, M.; Prasad, P.D.; Manicassamy, S.; Munn, D.H.; et al. Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis. Immunity 2014, 40, 128–139. [Google Scholar] [CrossRef]
- Arpaia, N.; Campbell, C.; Fan, X.; Dikiy, S.; van der Veeken, J.; deRoos, P.; Liu, H.; Cross, J.R.; Pfeffer, K.; Coffer, P.J.; et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 2013, 504, 451–455. [Google Scholar] [CrossRef]
- Smith, P.M.; Howitt, M.R.; Panikov, N.; Michaud, M.; Gallini, C.A.; Bohlooly-Y, M.; Glickman, J.N.; Garrett, W.S. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 2013, 341, 569–573. [Google Scholar] [CrossRef]
- Lee, S.U.; In, H.J.; Kwon, M.S.; Park, B.; Jo, M.; Kim, M.-O.; Cho, S.; Lee, S.; Lee, H.-J.; Kwak, Y.S.; et al. β-Arrestin 2 mediates G protein-coupled receptor 43 signals to nuclear factor-κB. Biol. Pharm. Bull. 2013, 36, 1754–1759. [Google Scholar] [CrossRef] [PubMed]
- Chen, W.; Liu, F.; Ling, Z.; Tong, X.; Xiang, C. Human intestinal lumen and mucosa-associated microbiota in patients with colorectal cancer. PLoS ONE 2012, 7, e39743. [Google Scholar] [CrossRef]
- Chang, P.V.; Hao, L.; Offermanns, S.; Medzhitov, R. The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition. Proc. Natl. Acad. Sci. USA 2014, 111, 2247–2252. [Google Scholar] [CrossRef] [PubMed]
- Macia, L.; Tan, J.; Vieira, A.T.; Leach, K.; Stanley, D.; Luong, S.; Maruya, M.; Ian McKenzie, C.; Hijikata, A.; Wong, C.; et al. Metabolite-sensing receptors GPR43 and GPR109A facilitate dietary fibre-induced gut homeostasis through regulation of the inflammasome. Nat. Commun. 2015, 6, 6734. [Google Scholar] [CrossRef]
- Xiu, W.; Chen, Q.; Wang, Z.; Wang, J.; Zhou, Z. Microbiota-derived short chain fatty acid promotion of Amphiregulin expression by dendritic cells is regulated by GPR43 and Blimp-1. Biochem. Biophys. Res. Commun. 2020, 533, 282–288. [Google Scholar] [CrossRef] [PubMed]
- Yang, W.; Yu, T.; Huang, X.; Bilotta, A.J.; Xu, L.; Lu, Y.; Sun, J.; Pan, F.; Zhou, J.; Zhang, W.; et al. Intestinal microbiota-derived short-chain fatty acids regulation of immune cell IL-22 production and gut immunity. Nat. Commun. 2020, 11, 4457. [Google Scholar] [CrossRef]
- Wong, S.H.; Yu, J. Gut microbiota in colorectal cancer: Mechanisms of action and clinical applications. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 690–704. [Google Scholar] [CrossRef] [PubMed]
- Castellarin, M.; Warren, R.L.; Freeman, J.D.; Dreolini, L.; Krzywinski, M.; Strauss, J.; Barnes, R.; Watson, P.; Allen-Vercoe, E.; Moore, R.A.; et al. Fusobacterium nucleatum infection is prevalent in human colorectal carcinoma. Genome Res. 2012, 22, 299–306. [Google Scholar] [CrossRef] [PubMed]
- Wang, T.; Cai, G.; Qiu, Y.; Fei, N.; Zhang, M.; Pang, X.; Jia, W.; Cai, S.; Zhao, L. Structural segregation of gut microbiota between colorectal cancer patients and healthy volunteers. ISME J. 2012, 6, 320–329. [Google Scholar] [CrossRef] [PubMed]
- Meira, L.B.; Bugni, J.M.; Green, S.L.; Lee, C.-W.; Pang, B.; Borenshtein, D.; Rickman, B.H.; Rogers, A.B.; Moroski-Erkul, C.A.; McFaline, J.L.; et al. DNA damage induced by chronic inflammation contributes to colon carcinogenesis in mice. J. Clin. Investig. 2008, 118, 2516–2525. [Google Scholar] [CrossRef]
- Matthews, G.M.; Howarth, G.S.; Butler, R.N. Short-chain fatty acids induce apoptosis in colon cancer cells associated with changes to intracellular redox state and glucose metabolism. Chemotherapy 2012, 58, 102–109. [Google Scholar] [CrossRef]
- Davie, J.R. Inhibition of histone deacetylase activity by butyrate. J. Nutr. 2003, 133, 2485S–2493S. [Google Scholar] [CrossRef]
- Yan, Q.; Gu, Y.; Li, X.; Yang, W.; Jia, L.; Chen, C.; Han, X.; Huang, Y.; Zhao, L.; Li, P.; et al. Alterations of the Gut Microbiome in Hypertension. Front. Cell. Infect. Microbiol. 2017, 7, 381. [Google Scholar] [CrossRef]
- Yang, T.; Magee, K.L.; Colon-Perez, L.M.; Larkin, R.; Liao, Y.-S.; Balazic, E.; Cowart, J.R.; Arocha, R.; Redler, T.; Febo, M.; et al. Impaired butyrate absorption in the proximal colon, low serum butyrate and diminished central effects of butyrate on blood pressure in spontaneously hypertensive rats. Acta Physiol. 2019, 226, e13256. [Google Scholar] [CrossRef]
- Pluznick, J.L.; Protzko, R.J.; Gevorgyan, H.; Peterlin, Z.; Sipos, A.; Han, J.; Brunet, I.; Wan, L.-X.; Rey, F.; Wang, T.; et al. Olfactory receptor responding to gut microbiota-derived signals plays a role in renin secretion and blood pressure regulation. Proc. Natl. Acad. Sci. USA 2013, 110, 4410–4415. [Google Scholar] [CrossRef] [PubMed]
- Natarajan, N.; Hori, D.; Flavahan, S.; Steppan, J.; Flavahan, N.A.; Berkowitz, D.E.; Pluznick, J.L. Microbial short chain fatty acid metabolites lower blood pressure via endothelial G protein-coupled receptor 41. Physiol. Genom. 2016, 48, 826–834. [Google Scholar] [CrossRef] [PubMed]
- Lal, S.; Kirkup, A.J.; Brunsden, A.M.; Thompson, D.G.; Grundy, D. Vagal afferent responses to fatty acids of different chain length in the rat. Am. J. Physiol. Gastrointest. Liver Physiol. 2001, 281, G907–G915. [Google Scholar] [CrossRef] [PubMed]
- Jie, Z.; Xia, H.; Zhong, S.-L.; Feng, Q.; Li, S.; Liang, S.; Zhong, H.; Liu, Z.; Gao, Y.; Zhao, H.; et al. The gut microbiome in atherosclerotic cardiovascular disease. Nat. Commun. 2017, 8, 845. [Google Scholar] [CrossRef]
- Aguilar, E.C.; Leonel, A.J.; Teixeira, L.G.; Silva, A.R.; Silva, J.F.; Pelaez, J.M.N.; Capettini, L.S.A.; Lemos, V.S.; Santos, R.A.S.; Alvarez-Leite, J.I. Butyrate impairs atherogenesis by reducing plaque inflammation and vulnerability and decreasing NFκB activation. Nutr. Metab. Cardiovasc. Dis. 2014, 24, 606–613. [Google Scholar] [CrossRef] [PubMed]
- Kim, K.N.; Yao, Y.; Ju, S.Y. Short Chain Fatty Acids and Fecal Microbiota Abundance in Humans with Obesity: A Systematic Review and Meta-Analysis. Nutrients 2019, 11, 2512. [Google Scholar] [CrossRef]
- Müller, M.; Hernández, M.A.G.; Goossens, G.H.; Reijnders, D.; Holst, J.J.; Jocken, J.W.E.; van Eijk, H.; Canfora, E.E.; Blaak, E.E. Circulating but not faecal short-chain fatty acids are related to insulin sensitivity, lipolysis and GLP-1 concentrations in humans. Sci. Rep. 2019, 9, 12515. [Google Scholar] [CrossRef]
- 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]
- Samuel, B.S.; Shaito, A.; Motoike, T.; Rey, F.E.; Backhed, F.; Manchester, J.K.; Hammer, R.E.; Williams, S.C.; Crowley, J.; Yanagisawa, M.; et al. Effects of the gut microbiota on host adiposity are modulated by the short-chain fatty-acid binding G protein-coupled receptor, Gpr41. Proc. Natl. Acad. Sci. USA 2008, 105, 16767–16772. [Google Scholar] [CrossRef]
- Chambers, E.S.; Viardot, A.; Psichas, A.; Morrison, D.J.; Murphy, K.G.; Zac-Varghese, S.E.K.; MacDougall, K.; Preston, T.; Tedford, C.; Finlayson, G.S.; et al. Effects of targeted delivery of propionate to the human colon on appetite regulation, body weight maintenance and adiposity in overweight adults. Gut 2015, 64, 1744–1754. [Google Scholar] [CrossRef]
- Larraufie, P.; Martin-Gallausiaux, C.; Lapaque, N.; Dore, J.; Gribble, F.M.; Reimann, F.; Blottiere, H.M. SCFAs strongly stimulate PYY production in human enteroendocrine cells. Sci. Rep. 2018, 8, 74. [Google Scholar] [CrossRef]
- Pingitore, A.; Chambers, E.S.; Hill, T.; Maldonado, I.R.; Liu, B.; Bewick, G.; Morrison, D.J.; Preston, T.; Wallis, G.A.; Tedford, C.; et al. The diet-derived short chain fatty acid propionate improves beta-cell function in humans and stimulates insulin secretion from human islets in vitro. Diabetes Obes. Metab. 2017, 19, 257–265. [Google Scholar] [CrossRef]
- Li, G.; Yao, W.; Jiang, H. Short-chain fatty acids enhance adipocyte differentiation in the stromal vascular fraction of porcine adipose tissue. J. Nutr. 2014, 144, 1887–1895. [Google Scholar] [CrossRef] [PubMed]
- Hong, Y.-H.; Nishimura, Y.; Hishikawa, D.; Tsuzuki, H.; Miyahara, H.; Gotoh, C.; Choi, K.-C.; Feng, D.D.; Chen, C.; Lee, H.-G.; et al. Acetate and propionate short chain fatty acids stimulate adipogenesis via GPCR43. Endocrinology 2005, 146, 5092–5099. [Google Scholar] [CrossRef] [PubMed]
- Jocken, J.W.E.; González Hernández, M.A.; Hoebers, N.T.H.; van der Beek, C.M.; Essers, Y.P.G.; Blaak, E.E.; Canfora, E.E. Short-Chain Fatty Acids Differentially Affect Intracellular Lipolysis in a Human White Adipocyte Model. Front. Endocrinol. 2017, 8, 372. [Google Scholar] [CrossRef] [PubMed]
- Ge, H.; Li, X.; Weiszmann, J.; Wang, P.; Baribault, H.; Chen, J.-L.; Tian, H.; Li, Y. Activation of G protein-coupled receptor 43 in adipocytes leads to inhibition of lipolysis and suppression of plasma free fatty acids. Endocrinology 2008, 149, 4519–4526. [Google Scholar] [CrossRef]
- Al-Lahham, S.; Roelofsen, H.; Rezaee, F.; Weening, D.; Hoek, A.; Vonk, R.; Venema, K. Propionic acid affects immune status and metabolism in adipose tissue from overweight subjects. Eur. J. Clin. Investig. 2012, 42, 357–364. [Google Scholar] [CrossRef]
- Zhao, Y.; Liu, J.; Hao, W.; Zhu, H.; Liang, N.; He, Z.; Ma, K.Y.; Chen, Z.-Y. Structure-Specific Effects of Short-Chain Fatty Acids on Plasma Cholesterol Concentration in Male Syrian Hamsters. J. Agric. Food Chem. 2017, 65, 10984–10992. [Google Scholar] [CrossRef]
- Butzner, J.D.; Parmar, R.; Bell, C.J.; Dalal, V. Butyrate enema therapy stimulates mucosal repair in experimental colitis in the rat. Gut 1996, 38, 568–573. [Google Scholar] [CrossRef]
- Breuer, R.I.; Soergel, K.H.; Lashner, B.A.; Christ, M.L.; Hanauer, S.B.; Vanagunas, A.; Harig, J.M.; Keshavarzian, A.; Robinson, M.; Sellin, J.H.; et al. Short chain fatty acid rectal irrigation for left-sided ulcerative colitis: A randomised, placebo controlled trial. Gut 1997, 40, 485–491. [Google Scholar] [CrossRef]
- Jamka, M.; Kokot, M.; Kaczmarek, N.; Bermagambetova, S.; Nowak, J.K.; Walkowiak, J. The Effect of Sodium Butyrate Enemas Compared with Placebo on Disease Activity, Endoscopic Scores, and Histological and Inflammatory Parameters in Inflammatory Bowel Diseases: A Systematic Review of Randomised Controlled Trials. Complement. Med. Res. 2021, 28, 344–356. [Google Scholar] [CrossRef]
- Vernero, M.; De Blasio, F.; Ribaldone, D.G.; Bugianesi, E.; Pellicano, R.; Saracco, G.M.; Astegiano, M.; Caviglia, G.P. The Usefulness of Microencapsulated Sodium Butyrate Add-On Therapy in Maintaining Remission in Patients with Ulcerative Colitis: A Prospective Observational Study. J. Clin. Med. 2020, 9, 3941. [Google Scholar] [CrossRef] [PubMed]
- Pietrzak, A.; Banasiuk, M.; Szczepanik, M.; Borys-Iwanicka, A.; Pytrus, T.; Walkowiak, J.; Banaszkiewicz, A. Sodium Butyrate Effectiveness in Children and Adolescents with Newly Diagnosed Inflammatory Bowel Diseases-Randomized Placebo-Controlled Multicenter Trial. Nutrients 2022, 14, 3283. [Google Scholar] [CrossRef] [PubMed]
- Denton, A.S.; Andreyev, H.J.N.; Forbes, A.; Maher, E.J. Systematic review for non-surgical interventions for the management of late radiation proctitis. Br. J. Cancer 2002, 87, 134–143. [Google Scholar] [CrossRef] [PubMed]
- Pinto, A.; Fidalgo, P.; Cravo, M.; Midões, J.; Chaves, P.; Rosa, J.; dos Anjos Brito, M.; Leitão, C.N. Short chain fatty acids are effective in short-term treatment of chronic radiation proctitis: Randomized, double-blind, controlled trial. Dis. Colon Rectum 1999, 42, 788–795, discussion 795–796. [Google Scholar] [CrossRef] [PubMed]
- Long, X.; Li, M.; Li, L.-X.; Sun, Y.-Y.; Zhang, W.-X.; Zhao, D.-Y.; Li, Y.-Q. Butyrate promotes visceral hypersensitivity in an IBS-like model via enteric glial cell-derived nerve growth factor. Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc. 2018, 30, e13227. [Google Scholar] [CrossRef]
- Shaidullov, I.F.; Sorokina, D.M.; Sitdikov, F.G.; Hermann, A.; Abdulkhakov, S.R.; Sitdikova, G.F. Short chain fatty acids and colon motility in a mouse model of irritable bowel syndrome. BMC Gastroenterol. 2021, 21, 37. [Google Scholar] [CrossRef]
- Nozu, T.; Miyagishi, S.; Nozu, R.; Takakusaki, K.; Okumura, T. Butyrate inhibits visceral allodynia and colonic hyperpermeability in rat models of irritable bowel syndrome. Sci. Rep. 2019, 9, 19603. [Google Scholar] [CrossRef]
- Pozuelo, M.; Panda, S.; Santiago, A.; Mendez, S.; Accarino, A.; Santos, J.; Guarner, F.; Azpiroz, F.; Manichanh, C. Reduction of butyrate- and methane-producing microorganisms in patients with Irritable Bowel Syndrome. Sci. Rep. 2015, 5, 12693. [Google Scholar] [CrossRef]
- Lewandowski, K.; Kaniewska, M.; Karłowicz, K.; Rosołowski, M.; Rydzewska, G. The effectiveness of microencapsulated sodium butyrate at reducing symptoms in patients with irritable bowel syndrome. Przeglad Gastroenterol. 2022, 17, 28–34. [Google Scholar] [CrossRef]
- Krokowicz, L.; Stojcev, Z.; Kaczmarek, B.F.; Kociemba, W.; Kaczmarek, E.; Walkowiak, J.; Krokowicz, P.; Drews, M.; Banasiewicz, T. Microencapsulated sodium butyrate administered to patients with diverticulosis decreases incidence of diverticulitis—A prospective randomized study. Int. J. Colorectal Dis. 2014, 29, 387–393. [Google Scholar] [CrossRef]
- Zhao, Z.-H.; Wang, Z.-X.; Zhou, D.; Han, Y.; Ma, F.; Hu, Z.; Xin, F.-Z.; Liu, X.-L.; Ren, T.-Y.; Zhang, F.; et al. Sodium Butyrate Supplementation Inhibits Hepatic Steatosis by Stimulating Liver Kinase B1 and Insulin-Induced Gene. Cell. Mol. Gastroenterol. Hepatol. 2021, 12, 857–871. [Google Scholar] [CrossRef] [PubMed]
- Yu, Z.; Han, J.; Chen, H.; Wang, Y.; Zhou, L.; Wang, M.; Zhang, R.; Jin, X.; Zhang, G.; Wang, C.; et al. Oral Supplementation With Butyrate Improves Myocardial Ischemia/Reperfusion Injury via a Gut-Brain Neural Circuit. Front. Cardiovasc. Med. 2021, 8, 718674. [Google Scholar] [CrossRef]
- Nutting, C.W.; Islam, S.; Daugirdas, J.T. Vasorelaxant effects of short chain fatty acid salts in rat caudal artery. Am. J. Physiol.-Heart Circ. Physiol. 1991, 261, H561–H567. [Google Scholar] [CrossRef] [PubMed]
- Mortensen, F.V.; Nielsen, H.; Mulvany, M.J.; Hessov, I. Short chain fatty acids dilate isolated human colonic resistance arteries. Gut 1990, 31, 1391–1394. [Google Scholar] [CrossRef]
- Wang, L.; Zhu, Q.; Lu, A.; Liu, X.; Zhang, L.; Xu, C.; Liu, X.; Li, H.; Yang, T. Sodium butyrate suppresses angiotensin II-induced hypertension by inhibition of renal (pro)renin receptor and intrarenal renin–angiotensin system. J. Hypertens. 2017, 35, 1899. [Google Scholar] [CrossRef] [PubMed]
- Bridgeman, S.C.; Northrop, W.; Melton, P.E.; Ellison, G.C.; Newsholme, P.; Mamotte, C.D.S. Butyrate generated by gut microbiota and its therapeutic role in metabolic syndrome. Pharmacol. Res. 2020, 160, 105174. [Google Scholar] [CrossRef]
- Roshanravan, N.; Mahdavi, R.; Alizadeh, E.; Jafarabadi, M.A.; Hedayati, M.; Ghavami, A.; Alipour, S.; Alamdari, N.M.; Barati, M.; Ostadrahimi, A. Effect of Butyrate and Inulin Supplementation on Glycemic Status, Lipid Profile and Glucagon-Like Peptide 1 Level in Patients with Type 2 Diabetes: A Randomized Double-Blind, Placebo-Controlled Trial. Horm. Metab. Res. 2017, 49, 886–891. [Google Scholar] [CrossRef]
- Bouter, K.E.; Bakker, G.J.; Levin, E.; Hartstra, A.V.; Kootte, R.S.; Udayappan, S.D.; Katiraei, S.; Bahler, L.; Gilijamse, P.W.; Tremaroli, V.; et al. Differential metabolic effects of oral butyrate treatment in lean versus metabolic syndrome subjects. Clin. Transl. Gastroenterol. 2018, 9, e155. [Google Scholar] [CrossRef]
- Facchin, S.; Vitulo, N.; Calgaro, M.; Buda, A.; Romualdi, C.; Pohl, D.; Perini, B.; Lorenzon, G.; Marinelli, C.; D’Incà, R.; et al. Microbiota changes induced by microencapsulated sodium butyrate in patients with inflammatory bowel disease. Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc. 2020, 32, e13914. [Google Scholar] [CrossRef]
- Rinninella, E.; Raoul, P.; Cintoni, M.; Franceschi, F.; Miggiano, G.A.D.; Gasbarrini, A.; Mele, M.C. What is the Healthy Gut Microbiota Composition? A Changing Ecosystem across Age, Environment, Diet, and Diseases. Microorganisms 2019, 7, 14. [Google Scholar] [CrossRef]
- Cronin, P.; Joyce, S.A.; O’Toole, P.W.; O’Connor, E.M. Dietary Fibre Modulates the Gut Microbiota. Nutrients 2021, 13, 1655. [Google Scholar] [CrossRef]
- So, D.; Whelan, K.; Rossi, M.; Morrison, M.; Holtmann, G.; Kelly, J.T.; Shanahan, E.R.; Staudacher, H.M.; Campbell, K.L. Dietary fiber intervention on gut microbiota composition in healthy adults: A systematic review and meta-analysis. Am. J. Clin. Nutr. 2018, 107, 965–983. [Google Scholar] [CrossRef] [PubMed]
- Wagenaar, C.A.; van de Put, M.; Bisschops, M.; Walrabenstein, W.; de Jonge, C.S.; Herrema, H.; van Schaardenburg, D. The Effect of Dietary Interventions on Chronic Inflammatory Diseases in Relation to the Microbiome: A Systematic Review. Nutrients 2021, 13, 3208. [Google Scholar] [CrossRef] [PubMed]
- Ojo, O.; Feng, Q.-Q.; Ojo, O.O.; Wang, X.-H. The Role of Dietary Fibre in Modulating Gut Microbiota Dysbiosis in Patients with Type 2 Diabetes: A Systematic Review and Meta-Analysis of Randomised Controlled Trials. Nutrients 2020, 12, 3239. [Google Scholar] [CrossRef]
- Rew, L.; Harris, M.D.; Goldie, J. The ketogenic diet: Its impact on human gut microbiota and potential consequent health outcomes: A systematic literature review. Gastroenterol. Hepatol. Bed Bench 2022, 15, 326–342. [Google Scholar]
- Gibson, G.R.; Hutkins, R.; Sanders, M.E.; Prescott, S.L.; Reimer, R.A.; Salminen, S.J.; Scott, K.; Stanton, C.; Swanson, K.S.; Cani, P.D.; et al. Expert consensus document: The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 491–502. [Google Scholar] [CrossRef]
- François, I.E.J.A.; Lescroart, O.; Veraverbeke, W.S.; Marzorati, M.; Possemiers, S.; Evenepoel, P.; Hamer, H.; Houben, E.; Windey, K.; Welling, G.W.; et al. Effects of a wheat bran extract containing arabinoxylan oligosaccharides on gastrointestinal health parameters in healthy adult human volunteers: A double-blind, randomised, placebo-controlled, cross-over trial. Br. J. Nutr. 2012, 108, 2229–2242. [Google Scholar] [CrossRef] [PubMed]
- Damen, B.; Cloetens, L.; Broekaert, W.F.; François, I.; Lescroart, O.; Trogh, I.; Arnaut, F.; Welling, G.W.; Wijffels, J.; Delcour, J.A.; et al. Consumption of breads containing in situ-produced arabinoxylan oligosaccharides alters gastrointestinal effects in healthy volunteers. J. Nutr. 2012, 142, 470–477. [Google Scholar] [CrossRef] [PubMed]
- Walton, G.E.; Lu, C.; Trogh, I.; Arnaut, F.; Gibson, G.R. A randomised, double-blind, placebo controlled cross-over study to determine the gastrointestinal effects of consumption of arabinoxylan-oligosaccharides enriched bread in healthy volunteers. Nutr. J. 2012, 11, 36. [Google Scholar] [CrossRef] [PubMed]
- Rauch, C.E.; Mika, A.S.; McCubbin, A.J.; Huschtscha, Z.; Costa, R.J.S. Effect of prebiotics, probiotics, and synbiotics on gastrointestinal outcomes in healthy adults and active adults at rest and in response to exercise-A systematic literature review. Front. Nutr. 2022, 9, 1003620. [Google Scholar] [CrossRef]
- Kim, Y.A.; Keogh, J.B.; Clifton, P.M. Probiotics, prebiotics, synbiotics and insulin sensitivity. Nutr. Res. Rev. 2018, 31, 35–51. [Google Scholar] [CrossRef] [PubMed]
- Schneider, S.M.; Girard-Pipau, F.; Anty, R.; van der Linde, E.G.M.; Philipsen-Geerling, B.J.; Knol, J.; Filippi, J.; Arab, K.; Hébuterne, X. Effects of total enteral nutrition supplemented with a multi-fibre mix on faecal short-chain fatty acids and microbiota. Clin. Nutr. Edinb. Scotl. 2006, 25, 82–90. [Google Scholar] [CrossRef] [PubMed]
- Kamarul Zaman, M.; Chin, K.-F.; Rai, V.; Majid, H.A. Fiber and prebiotic supplementation in enteral nutrition: A systematic review and meta-analysis. World J. Gastroenterol. 2015, 21, 5372–5381. [Google Scholar] [CrossRef] [PubMed]
- Markowiak-Kopeć, P.; Śliżewska, K. The Effect of Probiotics on the Production of Short-Chain Fatty Acids by Human Intestinal Microbiome. Nutrients 2020, 12, 1107. [Google Scholar] [CrossRef]
- Wang, L.; Zhang, J.; Guo, Z.; Kwok, L.; Ma, C.; Zhang, W.; Lv, Q.; Huang, W.; Zhang, H. Effect of oral consumption of probiotic Lactobacillus planatarum P-8 on fecal microbiota, SIgA, SCFAs, and TBAs of adults of different ages. Nutrition 2014, 30, 776–783.e1. [Google Scholar] [CrossRef]
- Hor, Y.-Y.; Lew, L.-C.; Jaafar, M.H.; Lau, A.S.-Y.; Ong, J.-S.; Kato, T.; Nakanishi, Y.; Azzam, G.; Azlan, A.; Ohno, H.; et al. Lactobacillus sp. improved microbiota and metabolite profiles of aging rats. Pharmacol. Res. 2019, 146, 104312. [Google Scholar] [CrossRef] [PubMed]
- Pérez Burillo, S.; Pastoriza de la Cueva, S.; Gironés, A.; Avellaneda, A.; Francino, M.P.; Rufián Henares, J.Á. Potential probiotic salami with dietary fiber modulates metabolism and gut microbiota in a human intervention study. J. Funct. Foods 2020, 66, 103790. [Google Scholar] [CrossRef]
- Hou, H.; Chen, D.; Zhang, K.; Zhang, W.; Liu, T.; Wang, S.; Dai, X.; Wang, B.; Zhong, W.; Cao, H. Gut microbiota-derived short-chain fatty acids and colorectal cancer: Ready for clinical translation? Cancer Lett. 2022, 526, 225–235. [Google Scholar] [CrossRef]
- Williams, L.M.; Stoodley, I.L.; Berthon, B.S.; Wood, L.G. The Effects of Prebiotics, Synbiotics, and Short-Chain Fatty Acids on Respiratory Tract Infections and Immune Function: A Systematic Review and Meta-Analysis. Adv. Nutr. 2022, 13, 167–192. [Google Scholar] [CrossRef]
- Keller, J.J.; Ooijevaar, R.E.; Hvas, C.L.; Terveer, E.M.; Lieberknecht, S.C.; Högenauer, C.; Arkkila, P.; Sokol, H.; Gridnyev, O.; Mégraud, F.; et al. A standardised model for stool banking for faecal microbiota transplantation: A consensus report from a multidisciplinary UEG working group. United Eur. Gastroenterol. J. 2021, 9, 229–247. [Google Scholar] [CrossRef]
- Cammarota, G.; Ianiro, G.; Magalini, S.; Gasbarrini, A.; Gui, D. Decrease in Surgery for Clostridium difficile Infection After Starting a Program to Transplant Fecal Microbiota. Ann. Intern. Med. 2015, 163, 487–488. [Google Scholar] [CrossRef] [PubMed]
- Ianiro, G.; Punčochář, M.; Karcher, N.; Porcari, S.; Armanini, F.; Asnicar, F.; Beghini, F.; Blanco-Míguez, A.; Cumbo, F.; Manghi, P.; et al. Variability of strain engraftment and predictability of microbiome composition after fecal microbiota transplantation across different diseases. Nat. Med. 2022, 28, 1913–1923. [Google Scholar] [CrossRef] [PubMed]
- Ianiro, G.; Rossi, E.; Thomas, A.M.; Schinzari, G.; Masucci, L.; Quaranta, G.; Settanni, C.R.; Lopetuso, L.R.; Armanini, F.; Blanco-Miguez, A. Faecal microbiota transplantation for the treatment of diarrhoea induced by tyrosine-kinase inhibitors in patients with metastatic renal cell carcinoma. Nat. Commun. 2020, 11, 4333. [Google Scholar] [CrossRef] [PubMed]
- Chen, R.; Xu, Y.; Wu, P.; Zhou, H.; Lasanajak, Y.; Fang, Y.; Tang, L.; Ye, L.; Li, X.; Cai, Z.; et al. Transplantation of fecal microbiota rich in short chain fatty acids and butyric acid treat cerebral ischemic stroke by regulating gut microbiota. Pharmacol. Res. 2019, 148, 104403. [Google Scholar] [CrossRef]
- El-Salhy, M.; Valeur, J.; Hausken, T.; Gunnar Hatlebakk, J. Changes in fecal short-chain fatty acids following fecal microbiota transplantation in patients with irritable bowel syndrome. Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc. 2021, 33, e13983. [Google Scholar] [CrossRef]
- Xu, Z.; Mak, J.W.Y.; Lin, Y.; Yang, K.; Liu, Q.; Zhang, F.; Lau, L.; Tang, W.; Ching, J.Y.; Tun, H.M.; et al. Mixed-donor faecal microbiota transplantation was associated with increased butyrate-producing bacteria for obesity. Gut 2023. [Google Scholar] [CrossRef]
- Bibbò, S.; Settanni, C.R.; Porcari, S.; Bocchino, E.; Ianiro, G.; Cammarota, G.; Gasbarrini, A. Fecal Microbiota Transplantation: Screening and Selection to Choose the Optimal Donor. J. Clin. Med. 2020, 9, 1757. [Google Scholar] [CrossRef]
Disease | SCFA | Model | Function | Ref. |
---|---|---|---|---|
Inflammatory bowel disease | Acetate | Gpr43−/−, Gpr109a−/−, Nlrp3−/− and Nlrp6−/− mice | Induces NLRP3 inflammosome activation to maintain tissue homeostasis | [133] |
Butyrate | Niacr1+/− Apc min/+ and Niacr1−/− Apc min/+ mice | Increases colonic DCs and macrophages’ production of IL-10, inducing Treg generation | [127] | |
Foxp3 ΔCNS1, Foxp3 GFP, Foxp3 Thy1.1 and Gpr109a−/− mice | Promotes Treg differentiation through enhancing Foxp3 activity | [128] | ||
GPR109a−/− and WT mice | Inhibits AKT and NF-κB p65 signaling pathways in macrophages | [131] | ||
BMDM cells, C57BL/6 and CX3CR1-GFP/+ mice | Reduces NO, IL-6 and IL-12p40 secretion by macrophages | [132] | ||
GPR43−/−, Prdm1−/− and WT mice | Increases AREG expression levels in DCs to promote tissue repair | [134] | ||
Cdx2-IEC monolayer | Induces production of claudin-1 to enhance barrier functions | [138] | ||
Propionate | Gpr43−/− and Gpr43+/+ mice | Promotes Treg differentiation through GPR-43 | [129] | |
All SCFAs | HeLa and HEK293 cell lines | Inhibit NF-κB activity through GPR43—βarrestin interactions | [130] | |
Isolated human neutrophils, monocytes and PBMC | Promotes anti-inflammatory effects via the regulation of PGE2, cytokine and chemokine release | [58] | ||
CD4+ T cells and ILCs | Induces production of IL-22 to promote barrier functions | [135] | ||
Colorectal cancer | Butyrate | Caco-2 cell line | Enhances cancer cells’ apoptosis by alterations in the redox state and D-glucose metabolism | [140] |
MCF-7 (T5) and MDA MB 231 cell lines | Arrests cancer cells’ proliferation through upregulation of p21 | [141] | ||
Propionate | Caco-2, HCT116, HCT8, HT-29, SW620, SW480, CBS, FET and MOSER cell lines | Arrests cancer cells’ proliferation through p21 upregulation and decrease in cyclin D3, CDK-1 and CDK-2 | [142] | |
Hypertension | Acetate and propionate | Olfr78−/− and Gpr41−/− mice | Increase blood pressure through Olfr-78 | [144] |
Gpr41−/− and WT mice | Reduces blood pressure by binding GRP-41 | [145] | ||
Butyrate | Vagotomized Sheffield strain male Wistar rats | Reduces blood pressure through the regulation of afferent vagal terminals | [146] | |
Atherosclerosis | Butyrate | ApoE −/− mice | Reduces CCL-2, VCAM-1, and MMP-2 production to stabilize atherosclerotic plaques | [148] |
Obesity | Acetate | C57BL/6 male mice | Decreases appetite through central hypothalamic mechanisms | [43] |
Propionate | Isolated human colonic cells | Reduces food intake through the secretion of PPY and GLP-1 via GPR-41 | [153] | |
Propionate and butyrate | NCI-h716 and HuTu-80 cells | Reduce food intake through the secretion of PPY via inhibition of HDACs | [154] | |
Metabolic syndrome | Acetate | Isolated adipocytes from GPR43 knockout mice | Decreases lipid plasma levels through inhibition of lipolysis via GPR-43 | [159] |
Propionate | Human subjects and in vitro isolated human islets | Enhances glucose-stimulated insulin release and increases β-cell mass | [155] | |
Human adipose tissue culture | Decreases lipid plasma levels by stimulating lipogenesis | [160] | ||
Propionate and butyrate | Stromal vascular fraction of the porcine subcutaneous fat | Stimulates adipocyte differentiation | [156] | |
All SCFAs | PPARγ f/f and PPARγ lox/lox mice | Regulate gluconeogenesis and lipogenesis through PPARγ downregulation | [46] | |
Male Golden hamsters | Decrease cholesterol plasma levels by enhancing its hepatic uptake | [161] |
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. |
© 2023 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
Fusco, W.; Lorenzo, M.B.; Cintoni, M.; Porcari, S.; Rinninella, E.; Kaitsas, F.; Lener, E.; Mele, M.C.; Gasbarrini, A.; Collado, M.C.; et al. Short-Chain Fatty-Acid-Producing Bacteria: Key Components of the Human Gut Microbiota. Nutrients 2023, 15, 2211. https://doi.org/10.3390/nu15092211
Fusco W, Lorenzo MB, Cintoni M, Porcari S, Rinninella E, Kaitsas F, Lener E, Mele MC, Gasbarrini A, Collado MC, et al. Short-Chain Fatty-Acid-Producing Bacteria: Key Components of the Human Gut Microbiota. Nutrients. 2023; 15(9):2211. https://doi.org/10.3390/nu15092211
Chicago/Turabian StyleFusco, William, Manuel Bernabeu Lorenzo, Marco Cintoni, Serena Porcari, Emanuele Rinninella, Francesco Kaitsas, Elena Lener, Maria Cristina Mele, Antonio Gasbarrini, Maria Carmen Collado, and et al. 2023. "Short-Chain Fatty-Acid-Producing Bacteria: Key Components of the Human Gut Microbiota" Nutrients 15, no. 9: 2211. https://doi.org/10.3390/nu15092211
APA StyleFusco, W., Lorenzo, M. B., Cintoni, M., Porcari, S., Rinninella, E., Kaitsas, F., Lener, E., Mele, M. C., Gasbarrini, A., Collado, M. C., Cammarota, G., & Ianiro, G. (2023). Short-Chain Fatty-Acid-Producing Bacteria: Key Components of the Human Gut Microbiota. Nutrients, 15(9), 2211. https://doi.org/10.3390/nu15092211