Microbiota Short-Chain Fatty Acids Modulate Antioxidant Defences: Comparison
Please note this is a comparison between Version 4 by Wendy Huang and Version 3 by Wendy Huang.

Food nutrients play a key role in human metabolism and health via the modulation of multiple mechanisms, including energy metabolism, intestinal homeostasis, antioxidant homeostasis, and immune responses. The intestine is an essential organ involved in human nutrition, the metabolic activity of gut microbes is essential for maintaining host health, and alterations in its composition induce metabolic shifts that may have adverse effects. The consensus on microbiota-mediated healthy effects on the host is based on the microbe-induced biotransformation of food components into bioactive metabolites. Bioactive molecules exhibit, in combination with food components, the ability to modulate the metabolic pathways of the host or to modify the composition and metabolism of the microbiota. Studies indicated the efficacy of the carbohydrates accessible to the microbiota (MACs), polyphenols, and polyunsaturated fatty acids (PUFAs) in increasing the microbial population with the ability to yield biologically active metabolites (e.g., polyphenol metabolites, short-chain fatty acids (SCFAs)) capable of modulating redox homeostasis of the host.

  • MACs
  • polyphenols
  • PUFAs
  • gut microbiota
  • active metabolites

1. Introduction

The beneficial effects associated with the diversity of the microbial population arise from the metabolic activities of specific microbial populations. Under eubiotic conditions, the commensal relationship between the microbiota and the host mainly consists of the capacity of bacteria to generate bioactive metabolites, starting from the ingested food, which exhibits the ability to modulate different metabolic pathways of the host [1]. For example, the production of carboxylic acids with aliphatic tails with fewer than six carbon atoms such as acetate (C2), propionate (C3), and butyrate (C4), resulting from the anaerobic fermentation of dietary plant polysaccharides, is the most relevant metabolic activity of enteric microbiota. These molecules are collectively referred to as Short-chain Fatty Acids (SCFAs) [2].

The growth of anaerobic SCFA-producing bacteria is favored by the low oxygen concentrations in the intestine where the two most abundant populations, namely, Bacteroidetes and Firmicutes, mainly produce acetate/propionate and butyrate, respectively [3]. Interestingly, due to butyrate generation during acetate metabolism, their coexistence can be consequential to mutual metabolic gain, thus resulting from the utilization of acetate produced by Bacteroidetes and Firmicutes to produce butyrate and propionate [4]. This example strongly supports the concept that the production of SCFAs is finely tuned by the balance of the bacterial species present in the gut.

The homeostatic condition of the intestinal microbiota can be restored by the level of SCFAs, and many studies in vivo describe the link between gut dysbiosis and the production of SCFAs (Table 1).
Table 1. Studies reporting the link between gut dysbiosis/production of SCFAs in several human diseases. An increase or decrease in the levels considered is indicated by upward (↑) or downward arrow (↓), respectively.
Disease Model Microbiota Alteration

Production of SCFAs
Ref.
Diabetes   Randomized clinical trial

High-fiber diet
Type 2 diabetes

↓ SCFAs

High fiber intake

↑ SCFAs

↑ SCFA-producing bacteria
[5]
Meta analysis

Dietary fiber
↑ Butyrate, propionate

Bifidobacterium
[6]
Inflammatory Bowel Disease (IBD)   313 patients ↓ Acetate-to-butyrate converter

Firmicutes (Roseburia)

↓ Propionate

↑ Pathogens (Enterobacteriaceae, Proteobacteria)
[7]
  127 patients

87 healthy controls
↓ Butyrate-producing bacteria

(Firmicutes)

↓ SCFAs (acetate, propionate, butyrate)
[8]
  10 inactive Crohn patients

10 healthy controls
↓ SCFA-producing bacteria

Roseburia inulinivorans,

Ruminococcus torques,

Clostridium lavalense,

Bacteroides uniformis

Faecalibacterium prausnitzii
[9]
Nonalcoholic Fatty Liver

Disease
  14 nonalcoholic fatty liver,

18 nonalcoholic steatohepatitis

27 healthy controls
↑ SCFA levels

↑ SCFA-producing bacteria

(Fusobacteriaceae, Prevotellaceae)
[10]
  25 nonalcoholic fatty liver

25 nonalcoholic steatohepatitis

25 healthy donors
Ruminococcaceae

Clostridiales

Bacteroidetes

Firmicutes
[11]
  30 patients F0/1 fibrosis stage

27 patients F ≥ 2 fibrosis
Bacteroidetes (F ≥ 2)

Ruminococcus (F ≥ 2)

Prevotella
[12]
Neurodegeneration Parkinson’s Disease Nonparametric meta-analysis Akkermansia

↓ Fecal SCFAs (acetate, propionate, butyrate)
[13]
96 patients

85 controls
↓ Fecal SCFAs

↑ Plasma SCFAs

↑ Pro-inflammatory bacteria
[14]
95 patients

33 controls
↓ Fecal SCFAs

(propionic acetic, butyric)

↑ Plasma SCFA

(propionic acetic)
[15]
Alzheimer’s Disease 25 patients Firmicutes, Bifidobacterium

Bacteroidetes
[16]
33 dementia

22 mild cognitive impairment

120 subjective cognitive decline
↓ SCFA-producing bacteria (Ruminococcus, Eubacterium)

↑ AD biomarkers (Amyloid-β1-42 and p-tau concentrations)
[17]
Mouse model

Sodium butyrate supplementation
↓ Amyloid-β1-42 protein (40%) [18]

References

  1. Descamps, H.C.; Herrmann, B.; Wiredu, D.; Thaiss, C.A. The path toward using microbial metabolites as therapies. EBioMedicine 2019, 44, 747–754.
  2. 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.
  3. Louis, P.; Flint, H.J. Formation of propionate and butyrate by the human colonic microbiota. Environ. Microbiol. 2017, 19, 29–41.
  4. Mahowald, M.; Rey, F.; Seedorf, H.; Turnbaugh, P.; Fulton, R.; Wollam, A.; Shah, N.; Wang, C.; Magrini, V.; Wilson, R.; et al. Characterizing a model human gut microbiota composed of members of its two dominant bacterial phyla. Proc. Natl. Acad. Sci. USA 2009, 106, 5859–5864.
  5. Zhao, L.; Zhang, F.; Ding, X.;Wu, G.; Lam, Y.Y.;Wang, X.; Fu, H.; Xue, X.; Lu, C.; Ma, J.; et al. Gut bacteria selectively promoted by dietary fibers alleviate type 2 diabetes. Science 2018, 359, 1151–1156.
  6. 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. [CrossRef] Antioxidants 2023, 12, 1073 17 of 22
  7. Imhann, F.; Vich Vila, A.; Bonder, M.J.; Fu, J.; Gevers, D.; Visschedijk, M.C.; Spekhorst, L.M.; Alberts, R.; Franke, L.; van Dullemen, H.M.; et al. Interplay of host genetics and gut microbiota underlying the onset and clinical presentation of inflammatory bowel disease. Gut 2018, 67, 108–119.
  8. 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.
  9. Takahashi, K.; Nishida, A.; Fujimoto, T.; Fujii, M.; Shioya, M.; Innaeda, H.; Lnatorni, O.; Bamba, S.; Andoh, A.; Sugimoto, M. Reduced Abundance of Butyrate-Producing Bacteria Species in the Fecal Microbial Community in Crohn’s Disease. Digestion 2016, 93, 59–65.
  10. Rau, M.; Rehman, A.; Dittrich, M.; Groen, A.K.; Hermanns, H.M.; Seyfried, F.; Beyersdorf, N.; Dandekar, T.; Rosenstiel, P.; Geier, A. Fecal SCFAs and SCFA-producing bacteria in gut microbiome of human NAFLD as a putative link to systemic T-cell activation and advanced disease. United Eur. Gastroenterol. J. 2018, 6, 1496–1507.
  11. Tsai, M.C.; Liu, Y.Y.; Lin, C.C.; Wang, C.C.; Wu, Y.J.; Yong, C.C.; Chen, K.D.; Chuah, S.K.; Yao, C.C.; Huang, P.Y.; et al. Gut Microbiota Dysbiosis in Patients with Biopsy-Proven Nonalcoholic Fatty Liver Disease: A Cross-Sectional Study in Taiwan. Nutrients 2020, 12, 820.
  12. Boursier, J.; Mueller, O.; Barret, M.; Machado, M.; Fizanne, L.; Araujo-Perez, F.; Guy, C.D.; Seed, P.C.; Rawls, J.F.; David, L.A.; et al. The severity of nonalcoholic fatty liver disease is associated with gut dysbiosis and shift in the metabolic function of the gut microbiota. Hepatology 2016, 63, 764–775.
  13. Hirayama, M.; Ohno, K. Parkinson’s Disease and Gut Microbiota. Ann. Nutr. Metab. 2021, 77 (Suppl. 2), 28–35.
  14. Chen, S.J.; Chen, C.C.; Liao, H.Y.; Lin, Y.T.; Wu, Y.W.; Liou, J.M.; Wu, M.S.; Kuo, C.H.; Lin, C.H. Association of Fecal and Plasma Levels of Short-Chain Fatty Acids with Gut Microbiota and Clinical Severity in Patients with Parkinson Disease. Neurology 2022,98, e848–e858.
  15. Yang, X.; Ai, P.; He, X.; Mo, C.; Zhang, Y.; Xu, S.; Lai, Y.; Qian, Y.; Xiao, Q. Parkinson’s Disease Is Associated with ImpairedGut-Blood Barrier for Short-Chain Fatty Acids. Mov. Disord. 2022, 37, 1634–1643.
  16. Vogt, N.M.; Kerby, R.L.; Dill-McFarland, K.A.; Harding, S.J.; Merluzzi, A.P.; Johnson, S.C.; Carlsson, C.M.; Asthana, S.; Zetterberg, H.; Blennow, K.; et al. Gut microbiome alterations in Alzheimer’s disease. Sci. Rep. 2017, 7, 13537.
  17. Verhaar, B.J.H.; Hendriksen, H.M.A.; de Leeuw, F.A.; Doorduijn, A.S.; van Leeuwenstijn, M.; Teunissen, C.E.; Barkhof, F.; Scheltens, P.; Kraaij, R.; van Duijn, C.M.; et al. Gut Microbiota Composition Is Related to AD Pathology. Front. Immunol. 2021, 12, 794519.
  18. Fernando, W.M.A.D.; Martins, I.J.; Morici, M.; Bharadwaj, P.; Rainey-Smith, S.R.; Lim, W.L.F.; Martins, R.N. Sodium ButyrateReduces Brain Amyloid-Levels and Improves Cognitive Memory Performance in an Alzheimer’s Disease Transgenic Mouse Model at an Early Disease Stage. J. Alzheimer’s Dis. 2020, 74, 91–99.
  19. Barcenilla, A.; Pryde, S.; Martin, J.; Duncan, S.; Stewart, C.; Henderson, C.; Flint, H. Phylogenetic relationships of butyrate producing bacteria from the human gut. Appl. Environ. Microbiol. 2000, 66, 1654–1661.
  20. Ohata, A.; Usami, M.; Miyoshi, M. Short-chain fatty acids alter tight junction permeability in intestinal monolayer cells via lipoxygenase activation. Nutrition 2005, 21, 838–847.
  21. 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.
  22. Colgan, S.P.; Taylor, C.T. Hypoxia: An alarm signal during intestinal inflammation. Nat. Rev. Gastroenterol. Hepatol. 2010, 7, 281–287.
  23. Bohnhoff, M.; Miller, C.P.; Martin, W.R. Resistance of the mouse’s intestinal tract to experimental salmonella infection. II. factors responsible for its loss following streptomycin treatment. J. Exp. Med. 1964, 120, 817–828.
  24. Rolfe, R.D. Role of volatile fatty acids in colonization resistance to Clostridium difficile. Infect. Immun. 1984, 45, 185–191
  25. Cummings, J.H.; Pomare, E.W.; Branch,W.J.; Naylor, C.P.; Macfarlane, G.T. Short chain fatty acids in human large intestine, portal, hepatic and venous blood. Gut 1987, 28, 1221–1227.
  26. Lin, H.V.; Frassetto, A.; Kowalik, E.J.; 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.
  27. Bergman, E.N. Energy contributions of volatile fatty acids from the gastrointestinal tract in various species. Physiol. Rev. 1990, 70, 567–590.
  28. Macfarlane, G.T.; Macfarlane, S. Fermentation in the human large intestine: Its physiologic consequences and the potential contribution of prebiotics. J. Clin. Gastroenterol. 2011, 45, S120–S127.
  29. 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.
  30. 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
  31. Hamer, H.M.; Jonkers, D.; Venema, K.; Vanhoutvin, S.; Troost, F.J.; Brummer, R.J. Review article: The role of butyrate on colonic function. Aliment. Pharmacol. Ther. 2008, 27, 104–119.
  32. Meesters, R.; van Eijk, H.; ten Have, G.; de Graaf, A.; Venema, K.; van Rossum, B.; Deutz, N. Application of liquid chromatography mass spectrometry to measure the concentrations and study the synthesis of short chain fatty acids following stable isotope infusions. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 2007, 854, 57–62.
  33. Moreau, N.; Goupry, S.; Antignac, J.; Monteau, F.; Le Bizec, B.; Champ, M.; Martin, L.; Dumon, H. Simultaneous measurement of plasma concentrations and C-13-enrichment of short-chain fatty acids, lactic acid and ketone bodies by gas chromatography coupled to mass spectrometry. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 2003, 784, 395–403.
  34. 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.
  35. Yajima, T. Contractile effect of short-chain fatty acids on the isolated colon of the rat. J. Physiol. 1985, 368, 667–678.
  36. Plaisancié, P.; Dumoulin, V.; Chayvialle, J.A.; Cuber, J.C. Luminal peptide YY-releasing factors in the isolated vascularly perfused rat colon. J. Endocrinol. 1996, 151, 421–429. [CrossRef]
  37. Fåk, F.; Jakobsdottir, G.; Kulcinskaja, E.; Marungruang, N.; Matziouridou, C.; Nilsson, U.; Stålbrand, H.; Nyman, M. The physico-chemical properties of dietary fibre determine metabolic responses, short-chain Fatty Acid profiles and gut microbiota composition in rats fed low- and high-fat diets. PLoS ONE 2015, 10, e0127252.
  38. Kilner, J.; Corfe, B.; McAuley, M.; Wilkinson, S. A deterministic oscillatory model of microtubule growth and shrinkage for differential actions of short chain fatty acids. Mol. Biosyst. 2016, 12, 93–101.
  39. Vincent, A.D.; Wang, X.Y.; Parsons, S.P.; Khan, W.I.; Huizinga, J.D. Abnormal absorptive colonic motor activity in germ-free mice is rectified by butyrate, an effect possibly mediated by mucosal serotonin. Am. J. Physiol. Gastrointest. Liver Physiol. 2018, 315, G896–G907.
  40. Gao, Z.; Yin, J.; Zhang, J.; Ward, R.E.; Martin, R.J.; Lefevre, M.; Cefalu, W.T.; Ye, J. Butyrate improves insulin sensitivity and increases energy expenditure in mice. Diabetes 2009, 58, 1509–1517.
  41. Fushimi, T.; Suruga, K.; Oshima, Y.; Fukiharu, M.; Tsukamoto, Y.; Goda, T. Dietary acetic acid reduces serum cholesterol and triacylglycerols in rats fed a cholesterol-rich diet. Br. J. Nutr. 2006, 95, 916–924.
  42. De Vadder, F.; Kovatcheva-Datchary, P.; Goncalves, D.; Vinera, J.; Zitoun, C.; Duchampt, A.; Backhed, F.; Mithieux, G. Microbiota-Generated Metabolites Promote Metabolic Benefits via Gut-Brain Neural Circuits. Cell 2014, 156, 84–96.
  43. Ukai, M.; Tomura, A.; Ito, M. Cholesterol-synthesis in germfree and conventional rats. J. Nutr. 1976, 106, 1175–1183.
  44. Wang, G.; Yu, Y.;Wang, Y.;Wang, J.; Guan, R.; Sun, Y.; Shi, F.; Gao, J.; Fu, X. Role of SCFAs in gut microbiome and glycolysis for colorectal cancer therapy. J. Cell. Physiol. 2019, 234, 17023–17049.
  45. Reigstad, C.S.; Salmonson, C.E.; Rainey, J.F.; Szurszewski, J.H.; Linden, D.R.; Sonnenburg, J.L.; Farrugia, G.; Kashyap, P.C. Gut microbes promote colonic serotonin production through an effect of short-chain fatty acids on enterochromaffin cells. FASEB J.2015, 29, 1395–1403.
  46. Vijay, N.; Morris, M.E. Role of monocarboxylate transporters in drug delivery to the brain. Curr. Pharm. Des. 2014, 20, 1487–1498.
  47. Daly, K.; Shirazi-Beechey, S. Microarray analysis of butyrate regulated genes in colonic epithelial cells. DNA Cell Biol. 2006, 25,49–62.
  48. Campos-Perez, W.; Martinez-Lopez, E. Effects of short chain fatty acids on metabolic and inflammatory processes in human health. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2021, 1866, 158900.
  49. González-Bosch, C.; Boorman, E.; Zunszain, P.A.; Mann, G.E. Short-chain fatty acids as modulators of redox signaling in health and disease. Redox Biol. 2021, 47, 102165.
  50. Visekruna, A.; Luu, M. The Role of Short-Chain Fatty Acids and Bile Acids in Intestinal and Liver Function, Inflammation, and Carcinogenesis. Front. Cell Dev. Biol. 2021, 9, 703218.
  51. Brown, A.J.; Goldsworthy, S.M.; Barnes, A.A.; Eilert, M.M.; Tcheang, L.; Daniels, D.; Muir, A.I.; Wigglesworth, M.J.; Kinghorn, I.; Fraser, N.J.; et al. The Orphan G protein-coupled receptors GPR41 and GPR43 are activated by propionate and other short chain carboxylic acids. J. Biol. Chem. 2003, 278, 11312–11319.
  52. Karaki, S.; Mitsui, R.; Hayashi, H.; Kato, I.; Sugiya, H.; Iwanaga, T.; Furness, J.B.; Kuwahara, A. Short-chain fatty acid receptor, GPR43, is expressed by enteroendocrine cells and mucosal mast cells in rat intestine. Cell Tissue Res. 2006, 324, 353–360.
  53. Ahmed, K.; Tunaru, S.; Offermanns, S. GPR109A, GPR109B and GPR81, a family of hydroxy-carboxylic acid receptors. Trends Pharmacol. Sci. 2009, 30, 557–562.
  54. Dorsam, R.T.; Gutkind, J.S. G-protein-coupled receptors and cancer. Nat. Rev. Cancer 2007, 7, 79–94.
  55. Schotterl, S.; Brennenstuhl, H.; Naumann, U. Modulation of immune responses by histone deacetylase inhibitors. Crit. Rev. Oncog. 2015, 20, 139–154.
  56. Weiss, U.; Möller, M.; Husseini, S.A.; Manderscheid, C.; Häusler, J.; Geisslinger, G.; Niederberger, E. Inhibition of HDAC Enzymes Contributes to Differential Expression of Pro-Inflammatory Proteins in the TLR-4 Signaling Cascade. Int. J. Mol. Sci. 2020, 21, 8943.
  57. 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.
  58. Sun, M.;Wu,W.; Chen, L.; Yang,W.; Huang, X.; Ma, C.; Chen, F.; Xiao, Y.; Zhao, Y.; Yao, S.; et al. Microbiota-derived short-chain fatty acids promote Th1 cell IL-10 production to maintain intestinal homeostasis. Nat. Commun. 2018, 9, 3555.
  59. Salazar, N.; Arboleya, S.; Fernández-Navarro, T.; de Los Reyes-Gavilán, C.G.; Gonzalez, S.; Gueimonde, M. Age-Associated Changes in Gut Microbiota and Dietary Components Related with the Immune System in Adulthood and Old Age: A Cross-Sectional Study. Nutrients 2019, 11, 1765.
  60. Matsumaru, D.; Motohashi, H. The KEAP1-NRF2 System in Healthy Aging and Longevity. Antioxidants 2021, 10, 1929.
  61. Li, H.; Shang, Z.; Liu, X.; Qiao, Y.;Wang, K.; Qiao, J. Alleviates Enterotoxigenic. Front. Immunol. 2021, 12, 771826.
  62. Dong,W.; Jia, Y.; Liu, X.; Zhang, H.; Li, T.; Huang,W.; Chen, X.;Wang, F.; Sun,W.;Wu, H. Sodium butyrate activates NRF2 to ameliorate diabetic nephropathy possibly via inhibition of HDAC. J. Endocrinol. 2017, 232, 71–83.
  63. Wu, J.; Jiang, Z.; Zhang, H.; Liang, W.; Huang, W.; Li, Y.; Wang, Z.; Wang, J.; Jia, Y.; Liu, B.; et al. Sodium butyrate attenuates diabetes-induced aortic endothelial dysfunction via P300-mediated transcriptional activation of Nrf2. Free Radic. Biol. Med. 2018,124, 454–465.
More
Video Production Service