Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2383 2023-05-15 07:37:10 |
2 format Meta information modification 2383 2023-05-17 03:33:40 | |
3 format -3 word(s) 2380 2023-05-22 04:04:42 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Mazhar, M.; Zhu, Y.; Qin, L. Dietary Fibers and Intestinal Microbiota Affects T2D. Encyclopedia. Available online: https://encyclopedia.pub/entry/44281 (accessed on 20 April 2024).
Mazhar M, Zhu Y, Qin L. Dietary Fibers and Intestinal Microbiota Affects T2D. Encyclopedia. Available at: https://encyclopedia.pub/entry/44281. Accessed April 20, 2024.
Mazhar, Muhammad, Yong Zhu, Likang Qin. "Dietary Fibers and Intestinal Microbiota Affects T2D" Encyclopedia, https://encyclopedia.pub/entry/44281 (accessed April 20, 2024).
Mazhar, M., Zhu, Y., & Qin, L. (2023, May 15). Dietary Fibers and Intestinal Microbiota Affects T2D. In Encyclopedia. https://encyclopedia.pub/entry/44281
Mazhar, Muhammad, et al. "Dietary Fibers and Intestinal Microbiota Affects T2D." Encyclopedia. Web. 15 May, 2023.
Dietary Fibers and Intestinal Microbiota Affects T2D
Edit

Foods contain dietary fibers which can be classified into soluble and insoluble forms. The nutritional composition of fast foods is considered unhealthy because it negatively affects the production of short-chain fatty acids (SCFAs). Dietary fiber is resistant to digestive enzymes in the gut, which modulates the anaerobic intestinal microbiota (AIM) and fabricates SCFAs. Acetate, butyrate, and propionate are dominant in the gut and are generated via Wood–Ljungdahl and acrylate pathways. In pancreatic dysfunction, the release of insulin/glucagon is impaired, leading to hyperglycemia. SCFAs enhance insulin sensitivity or secretion, beta-cell function, leptin release, mitochondrial function, and intestinal gluconeogenesis in human organs, which positively affects type 2 diabetes (T2D). Research models have shown that SCFAs either enhance the release of peptide YY (PYY) and glucagon-like peptide-1 (GLP-1) from L-cells (entero-endocrine), or promotes the release of leptin hormone in adipose tissues through G-protein receptors GPR-41 and GPR-43. Dietary fiber is a component that influences the production of SCFAs by AIM, which may have beneficial effects on T2D.

dietary fibers intestinal microbiota short-chain fatty acids fermentation

1. Dietary Fiber, Inflammatory Markers, and Type 2 Diabetes (T2D)

T2D is among the major diseases associated with a low level of inflammatory processes, characterized by amendments in the secretion of cytokines [1]. The amount of inflammatory markers (IL-6, TNF-α, and LPS) in T2D is increased, which is associated with dysfunction in insulin resistance and β-cell activity, and the amount of LPS in diabetic patients is twice as high as that in healthy individuals [2]. A high-fat diet is associated with metabolic endotoxemia caused by serum LPS, resulting in obesity and insulin resistance [3], and high serum LPS enhances TNF-α and inhibits insulin signals [4]. An excess amount of ANK-α indirectly inhibits insulin signaling by serine-307 phosphorylation in the substrate of the insulin receptor [5]. According to scientific reports, the composition of the diet can positively affect the inflammatory process; Lactobacillus spp. and Bifidobacterium spp., which are stimulated by dietary fiber, show anti-inflammatory properties [6]. Dietary fiber at 40 g/day can reduce the level of TNF-α [7].

2. Effects of Fructose on SCFAs and T2D

Sugar is an important source of energy in the daily diet, and there is increasing evidence that high sugar intake causes a number of major diet-related health problems, such as T2D and obesity [8][9]. Dietary factors influence blood glucose homeostasis in T2D; blood glucose levels rise when fructose is converted into glucose in the liver. This conversion takes time, so a small portion of fructose is converted into glucose, resulting in a lower increase in blood glucose levels [10]; therefore, the glycemic index of fructose is only 23 [11]. In addition to contributing to blood glucose homeostasis, fructose has also been shown to improve glycemic control at moderate levels [12][13]. The health effects of fructose are closely related to the consumption amount. Ultimately, it was determined that a high-fructose diet and a certain gut microbiota profile may be associated with the inflammation of the liver, pancreas, and colon. With low or inadequate fructose intake, no adverse effects were found on body weight, fasting blood glucose, histology, gut microbiota, or colonic SCFA levels [14][15][16]. Some evidence showed that fructose causes insulin resistance in the liver, which can negatively impact blood glucose homeostasis [17].

3. Effects of Lipids on SCFAs and T2D

A lipid molecule is mostly made up of repeating units named fatty acids. There are two types of fatty acids, saturated and unsaturated. Humans get most of their energy from fatty acids, which are the main components of triacylglycerols found in oils and fats [18][19]. Long-term consumption of a high-fat diet affects gut microbiota composition in animal models as well as in humans, which directly impacts SCFA production and host health [20]. High-fat diets containing medium-chain fatty acids, monounsaturated fatty acids, and polyunsaturated fatty acids, low-fat diets containing long-chain fatty acids, and diets with high Bacteroidetes or Firmicutes ratios were associated with increased SCFA production [21].

4. Short-Chain Fatty Acids (SCFAs)

SCFAs are organic acids produced in the human gut, where the AIM resides [22]. Quantitatively, these fatty acids are measured in millimoles, and they are predominately represented by acetate, butyrate, and propionate [23]. These three SCFAs are discussed here. The dietary carbon flow is based on SCFAs [24], and their production is fairly well understood and characterized [25][26]. The ratio and concentration of SCFAs depend on the microbial composition and the substrate (dietary fiber) provided to the GM [27]. The molar ratio of acetate, propionate, and butyrate is 3:1:1. SCFAs constitute 90–95% of the colon, whereas formic acid is present in a smaller proportion [23]. As a result of antibiotic treatment depleting the microbiota, mice were found to produce lower amounts of SCFAs, compared to mice that did not receive antibiotics [28]. A diet rich in prebiotics may be particularly effective at increasing SCFA production in diabetes [29]. According to previous studies, individuals with T2D have lower proportions of microbiota species producing butyrate [29][30].

4.1. The Contribution of Gut Microbiota Producing SCFAs

Dietary fibers are resistant to gut digestive enzymes, which contribute to the production of SCFAs during colonic fermentation [31]. Acetate, propionate, and butyrate are dominant SCFAs in the gut [6]. These fatty acids are composed of 1–6 carbon atoms and are naturally saturated [32]. Present-day research has shown a significant role for AIM, and the metabolites produced during dietary fiber fermentation positively contribute to T2D [33]. Gut intestinal microbiota, including Clostridiales spp. SS3/4, Roseburia inulinivorans, Roseburia intestinalis, Faecalibacterium prausnitzii, and Eubacterium rectale, produce butyrate, which has a protective role in T2D, even though these species are decreased in diabetes [34]. In addition, oral administration of Clostridium butyricum in obese diabetic rats was found to modulate gut microbiota to produce butyrate, leading to reduced proportions of Bacteroides and Firmicutes spp. [35].
In diet-induced diabetes, chitosan and antibiotics targeting Gram-negative intestinal microbes may be considered antidiabetic agents [36][37]. Cross-feeding GM metabolizes lactate into acetate, propionate, and butyrate in the gut fermentation process, in which propionate and butyrate are produced in limited quantities owing to selected GM, while acetate is a regular product in the gut [24]. Propionate is produced during the fermentation of propiogenic substrate (fucose/rhamnose) by Akkermansia municiphilla, whereas butyrate is produced through RS fermentation by Eubacterium hallii, Eubacterium rectale, Faecalibacterium prausnitzii, and Ruminococcus bromii in the gut; moreover, butyrogenic bacteria ferment pyruvate, lactate, and acetate into butyrate [23]. Acetate, propionate, and butyrate are the energy sources for the human body. Butyrate is directly utilized in the liver, heart, brain, and colon; propionate is used for gluconeogenesis in the liver, and acetate is used as fuel in peripheral tissues [38].
The responsiveness of free fatty acid receptors (FFAR-2 and -3) is proportional to the length of the carbon chain. For example, acetate and propionate are more responsive to FFAR-2, whereas butyrate and propionate are more responsive to FFAR-3 [39]. Medium (FFAR-1) and long-chain (FFAR-4) fatty acids were found to positively respond to inflammation and insulin secretion [40]. FFAR-1 enhances specific pancreatic β-cell activity, while in T2D, this activity is downregulated, resulting in FFAR-1 inhibition and insulin resistance [41]. FFAR-4 boosts these fatty acids (unsaturated) to stimulate glucagon-like peptide-1 (GLP-1), secreting insulin from β-cells [42]. Propionate and butyrate may positively regulate obesity and T2D when administered orally [43][44][45].

4.2. Production of SCFAs via Anaerobic Bacterial Pathways and the Role of Akkermansia Muciniphila in T2D

The non-digestible carbohydrates are hydrolyzed by the AIM into monosaccharides and oligosaccharides during anaerobic fermentation in the colon [46]. For the metabolization of monosaccharides into phosphoenolpyruvate (PEP), the Embden–Meyerhof–Parnas pathway (sugars containing 6-c) and the pentose phosphate pathway (sugars containing 5-c) are utilized [25]. Eventually, organic acids/alcohols are formed during PEP fermentation. Nicotinamide adenine dinucleotide (NAD) + hydrogen (H) (NADH) is produced during the reaction of an acidic protein, glyceraldehyde-3-phosphate dehydrogenase (GAPDH). First is the traditional fermentation pathway, in which lactate/ethanol is produced from the reduction of pyruvate. Second, pyruvate is reduced to acetyl-CoA (ACA) and NADH to NAD+ [47]. The second pathway produces excess amounts of H2 molecules by using two major routes, pyruvate (exergonic) and NADH (endergonic) via ferredoxin oxidoreductase and hydrogenase, respectively. Despite depleting/consuming H2 molecules, the AIM is a primary participant in the fermentation process when H2 pressure in the large intestine (lumen) is low [48]. Third, the fundamental electron transport chain (ETC) proceeds with anaerobes, starting with PEP carboxylation and the reduction of oxaloacetate into fumarate [49]. The electrons are accepted by fumarate from NADH; NADH dehydrogenase and fumarate reductase constituted an ordinary electron transfer chain (OETC) [49][50]. NADH-dehydrogenase contributes to the transport of protons across the cell membrane, resulting in the chemiosmotic synthesis of ATP. Succinate (produced by fumarate reductase) is transformed into methylmalonate once the preferential load of CO2 is reduced. PEP can also be recycled from oxaloacetate through the carboxylation process.
SCFAs are the end product of the fermentation pathways. Pyruvate is transformed into ACA, releasing H2 and CO2 molecules. Hydrolysis of ACA leads to the formation of acetate, or it can also be produced by the Wood–Ljungdahl pathway utilizing CO2, wherein CO2 is reduced to CO coupled with CoASH and a methyl group and converted to ACA [51][52]. Propionate is formed either by utilizing PEP via OETC or by reducing lactate to propionate via the acrylate pathway [25]. These pathways accommodate supplementary NADH associated with lactate fermentation. The condensation of ACA (2 molecules) results in the formation of butyrate, which is subsequently reduced to butyryl CoA. ACA is produced from lactate, and then lactate is utilized by gut bacteria to produce butyrate [53]. Two pathways are involved in the formation of butyrate: the traditional pathway uses phosphotransbutyrylase and butyrate kinase to convert butyryl CoA into butyrate, accompanying ATP formation, and in the alternative pathway, butyryl CoA is converted to butyrate via butyryl-CoA: acetate CoA transferase [54][55]. The exogenic utilization of acetate to form butyrate and ACA involves cross-feeding among acetate and butyrate-producing bacteria [56][57]; the human GM prefers the alternative over the traditional pathway [54].
The symbiotic association between GM and the human body is significant in SCFA production [58]. The primary metabolites (H2 molecules) produced to get acetate must be utilized by secondary fermenters to reduce the burden of these molecules and accelerate the oxidation of NADH via primary fermenters [59]. The human body provides the CO2 molecules required in the OETC, and an average of 0.7 kg/day of CO2 is produced by the human organism [60]. By exchanging SCFA anions, some of that production is secreted into the gut (lumen) as HCO3, which is likely a significant pH-regulating mechanism, since protons in the gut (lumen) generated during the formation of SCFAs are neutralized by bicarbonate to produce CO2 [59]. Subsequently, much is known about the biochemistry of SCFA production from carbohydrates via the AIM. However, further study is still needed to determine whether SCFAs, as the significant output of indigestible carbohydrates via the AIM, have beneficial effects in T2D.
Akkermansia muciniphila is the only representative Gram-negative Verrucomicrobia inhabiting human intestinal mucosa [61]. In the studies by Derrien, gene sequence analysis revealed that multiple genes are associated with mucin encoding, and a single chromosome containing 2176 genes with 55.8% GC content was found in the MucT type strain of A. muciniphila (ATCC BAA-835 1/4 CIP107961T) [62][63]. This immobile, oval-shaped microorganism is purely anaerobic and contains chemical organotrophic material that can endure low levels of oxygen. The enzymes produced by A. muciniphila were responsible for the breakdown of mucin, and the mucin in the mucosal layer of the epithelium was used as a source of carbon and nitrogen. In order to release the sulfate, A. muciniphila splits these compounds into acetic and propionic compounds [64][65]. According to an analysis of its 16SrRNA signature, A. muciniphila makes up 3 to 5% of the gut microbiome even in healthy adults, but the amount depends on several factors. Age has been closely associated with stability in humans. This species begins to colonize at a young age and ranges from 5.0 to 8.8 log cells/g in a year, which is comparable to the adult stage, although it decreases with age [66][67]. The combined effects of an excess amount of A. muciniphila supplementation can positively affect metabolic disorders including T2D, and early vancomycin therapy may help control the progression of autoimmune diabetes by early colonization of the intestinal tract with A. muciniphila [68][69].

4.3. Effects of SCFAs on T2D

SCFAs are metabolites of gut microbe fermentation that result from indigestible dietary fiber and may have a beneficial role in T2D [70]. Compared to normal animals, diabetic rodents that consumed a high-fat diet with streptozotocin showed lower levels of acetate, propionate, and butyrate [71][72]. It was found that T2D patients had lower fecal butyrate and propionate concentrations, as well as acetate concentrations, than healthy subjects [73]. Improved insulin secretion/sensitivity, reduced fat accumulation, intestinal gluconeogenesis (IGN) triggering, and inflammation are the mechanisms by which SCFAs can positively affect T2D [22][74]. A study using homeostatic model assessment of insulin resistance (HOMA-IR) observed an adverse correlation between blood insulin levels and total SCFAs, including acetate and propionate [75]. In vitro and in vivo studies showed that propionate can enhance the release of glucose-stimulated insulin, sustain β-cell mass by decreasing trans-differentiation in α-cells, obstruct apoptosis, and assist in proliferation [22][76]. Moreover, it was shown in mouse models that butyrate improved insulin sensitivity [44][77]. These mechanisms support energy consumption and boost mitochondrial functions [44].
Propionate- or butyrate-induced IGN affects glucose homeostasis, the cAMP-dependent pathway, and the gut–brain neural circuit [78]. Acetate enhances the suppression of lipogenesis in the liver and decreases lipid aggregation in adipose tissues, while glucose transporter-4 genes and myoglobin are enhanced in the abdominal muscles of diabetic rats [79]. The peroxisome proliferator-activated receptor-α (PPAR-α) gene was upregulated in the presence of acetate, which may suppress body fat aggregation [80][81][82]. Furthermore, SCFA supplementation reduces hepatic steatosis and body weight [83]. In vitro and in vivo models showed that SCFAs either enhance the release of peptide YY (PYY) and GLP-1 from L-cells (entero-endocrine), or promote the release of leptin hormone satiation in adipose tissues through G-protein receptors (GPR-41 and/or GPR-43) [84][85][86][87]. SCFAs promote lipid oxidation and energy consumption and were found to increase fasting fat oxidation and PYY concentration during colonic infusion in obese subjects [88]. Butyrate may weaken inflammation generated by the interaction of macrophages and adipocytes by decreasing lipolysis and obstructing inflammatory signals [89]. These fatty acids showed beneficial effects on T2D by reducing the production of TNK-α, IL-6, and monocyte chemoattractant protein-1 (MCP-1); nuclear factor kappa-B (NF-κB) activity was also constrained. Propionate had a positive influence on T2D, participating in the downregulation of inflammatory chemokines and cytokines, such as CC chemokine ligand-5 (CCL-5) and TNF-α [90].

References

  1. Esser, N.; Legrand-Poels, S.; Piette, J.; Scheen, A.J.; Paquot, N. Inflammation as a link between obesity, metabolic syndrome and type 2 diabetes. Diabetes Res. Clin. Pract. 2014, 105, 141–150.
  2. Creely, S.J.; McTernan, P.G.; Kusminski, C.M.; Fisher, F.M.; Da Silva, N.; Khanolkar, M.; Evans, M.; Harte, A.; Kumar, S. Lipopolysaccharide activates an innate immune system response in human adipose tissue in obesity and type 2 diabetes. Am. J. Physiol.-Endocrinol. Metab. 2007, 292, E740–E747.
  3. Pedro, M.N.; Magro, D.O.; da Silva, E.U.P.P.; Guadagnini, D.; Santos, A.; de Jesus Pedro, R.; Saad, M.J.A. Plasma levels of lipopolysaccharide correlate with insulin resistance in HIV patients. Diabetol. Metab. Syndr. 2018, 10, 5.
  4. Peraldi, P.; Hotamisligil, G.S.; Buurman, W.A.; White, M.F.; Spiegelman, B.M. Tumor necrosis factor (TNF)-α inhibits insulin signaling through stimulation of the p55 TNF receptor and activation of sphingomyelinase. J. Biol. Chem. 1996, 271, 13018–13022.
  5. Copps, K.D.; Hancer, N.J.; Opare-Ado, L.; Qiu, W.; Walsh, C.; White, M.F. Irs1 serine 307 promotes insulin sensitivity in mice. Cell Metab. 2010, 11, 84–92.
  6. 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.
  7. Bodinham, C.; Smith, L.; Thomas, E.L.; Bell, J.D.; Swann, J.R.; Costabile, A.; Russell-Jones, D.; Umpleby, A.M.; Robertson, M.D. Efficacy of increased resistant starch consumption in human type 2 diabetes. Endocr. Connect. 2014, 3, 75–84.
  8. Laughlin, M.R. Normal roles for dietary fructose in carbohydrate metabolism. Nutrients 2014, 6, 3117–3129.
  9. Taskinen, M.-R.; Packard, C.J.; Borén, J. Dietary fructose and the metabolic syndrome. Nutrients 2019, 11, 1987.
  10. Teff, K.L.; Elliott, S.S.; Tschöp, M.; Kieffer, T.J.; Rader, D.; Heiman, M.; Townsend, R.R.; Keim, N.L.; D’alessio, D.; Havel, P.J. Dietary fructose reduces circulating insulin and leptin, attenuates postprandial suppression of ghrelin, and increases triglycerides in women. J. Clin. Endocrinol. Metab. 2004, 89, 2963–2972.
  11. Segal, M.S.; Gollub, E.; Johnson, R.J. Is the fructose index more relevant with regards to cardiovascular disease than the glycemic index? Eur. J. Nutr. 2007, 46, 406–417.
  12. Sievenpiper, J.L.; de Souza, R.J.; Cozma, A.I.; Chiavaroli, L.; Ha, V.; Mirrahimi, A. Fructose vs. glucose and metabolism: Do the metabolic differences matter? Curr. Opin. Lipidol. 2014, 25, 8–19.
  13. Cozma, A.I.; Sievenpiper, J.L.; De Souza, R.J.; Chiavaroli, L.; Ha, V.; Wang, D.D.; Mirrahimi, A.; Yu, M.E.; Carleton, A.J.; Di Buono, M. Effect of fructose on glycemic control in diabetes: A systematic review and meta-analysis of controlled feeding trials. Diabetes Care 2012, 35, 1611–1620.
  14. Kolderup, A.; Svihus, B. Fructose metabolism and relation to atherosclerosis, type 2 diabetes, and obesity. J. Nutr. Metab. 2015, 2015, 823081.
  15. de Farias Lelis, D.; Andrade, J.M.O.; Almenara, C.C.P.; Broseguini-Filho, G.B.; Mill, J.G.; Baldo, M.P. High fructose intake and the route towards cardiometabolic diseases. Life Sci. 2020, 259, 118235.
  16. Hannou, S.A.; Haslam, D.E.; McKeown, N.M.; Herman, M.A. Fructose metabolism and metabolic disease. J. Clin. Investig. 2018, 128, 545–555.
  17. Havel, P.J. Dietary fructose: Implications for dysregulation of energy homeostasis and lipid/carbohydrate metabolism. Nutr. Rev. 2005, 63, 133–157.
  18. Hawkesworth, S.; Dangour, A.D.; Johnston, D.; Lock, K.; Poole, N.; Rushton, J.; Uauy, R.; Waage, J. Feeding the world healthily: The challenge of measuring the effects of agriculture on health. Philos. Trans. R. Soc. B Biol. Sci. 2010, 365, 3083–3097.
  19. Kearney, J. Food consumption trends and drivers. Philos. Trans. R. Soc. B Biol. Sci. 2010, 365, 2793–2807.
  20. Machate, D.J.; Figueiredo, P.S.; Marcelino, G.; Guimarães, R.d.C.A.; Hiane, P.A.; Bogo, D.; Pinheiro, V.A.Z.; Oliveira, L.C.S.d.; Pott, A. Fatty acid diets: Regulation of gut microbiota composition and obesity and its related metabolic dysbiosis. Int. J. Mol. Sci. 2020, 21, 4093.
  21. Portincasa, P.; Bonfrate, L.; Vacca, M.; De Angelis, M.; Farella, I.; Lanza, E.; Khalil, M.; Wang, D.Q.-H.; Sperandio, M.; Di Ciaula, A. Gut microbiota and short chain fatty acids: Implications in glucose homeostasis. Int. J. Mol. Sci. 2022, 23, 1105.
  22. Mandaliya, D.K.; Seshadri, S. Short Chain Fatty Acids, pancreatic dysfunction and type 2 diabetes. Pancreatology 2019, 19, 617–622.
  23. 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.
  24. 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.
  25. Miller, T.L.; Wolin, M.J. Pathways of acetate, propionate, and butyrate formation by the human fecal microbial flora. Appl. Environ. Microbiol. 1996, 62, 1589–1592.
  26. Flint, H.J.; Duncan, S.H.; Scott, K.P.; Louis, P. Links between diet, gut microbiota composition and gut metabolism. Proc. Nutr. Soc. 2015, 74, 13–22.
  27. Havenaar, R. Intestinal health functions of colonic microbial metabolites: A review. Benef. Microbes 2011, 2, 103–114.
  28. Robertson, M.D. Prebiotics and type 2 diabetes: Targeting the gut microbiota for improved glycaemic control? Pract. Diabetes 2020, 37, 133–137.
  29. Karlsson, F.H.; Tremaroli, V.; Nookaew, I.; Bergström, G.; Behre, C.J.; Fagerberg, B.; Nielsen, J.; Bäckhed, F. Gut metagenome in European women with normal, impaired and diabetic glucose control. Nature 2013, 498, 99–103.
  30. Li, Q.; Chang, Y.; Zhang, K.; Chen, H.; Tao, S.; Zhang, Z. Implication of the gut microbiome composition of type 2 diabetic patients from northern China. Scientific reports 2020, 10, 5450.
  31. Roy, C.C.; Kien, C.L.; Bouthillier, L.; Levy, E. Short-chain fatty acids: Ready for prime time? Nutr. Clin. Pract. 2006, 21, 351–366.
  32. 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.
  33. Brunkwall, L.; Orho-Melander, M. The gut microbiome as a target for prevention and treatment of hyperglycaemia in type 2 diabetes: From current human evidence to future possibilities. Diabetologia 2017, 60, 943–951.
  34. Qin, J.; Li, Y.; Cai, Z.; Li, S.; Zhu, J.; Zhang, F.; Liang, S.; Zhang, W.; Guan, Y.; Shen, D. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature 2012, 490, 55–60.
  35. Jia, L.; Li, D.; Feng, N.; Shamoon, M.; Sun, Z.; Ding, L.; Zhang, H.; Chen, W.; Sun, J.; Chen, Y.Q. Anti-diabetic effects of Clostridium butyricum CGMCC0313. 1 through promoting the growth of gut butyrate-producing bacteria in type 2 diabetic mice. Sci. Rep. 2017, 7, 7046.
  36. Patel, S.; Mandaliya, D.; Prajapati, B.; Kumar, S.; Seshadri, S. Cefdinir Microsphere modulated microflora and liver immunological response to diet induced diabetes in mice. Endocr. Metab. Immune Disord.-Drug Targets (Former. Curr. Drug Targets-Immune Endocr. Metab. Disord.) 2019, 19, 349–357.
  37. Jena, P.K.; Singh, S.; Prajapati, B.; Nareshkumar, G.; Mehta, T.; Seshadri, S. Impact of targeted specific antibiotic delivery for gut microbiota modulation on high-fructose-fed rats. Appl. Biochem. Biotechnol. 2014, 172, 3810–3826.
  38. Fleming, S.; Fitch, M.; DeVries, S.; Liu, M.; Kight, C. Nutrient utilization by cells isolated from rat jejunum, cecum and colon. J. Nutr. 1991, 121, 869–878.
  39. 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. 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.
  40. Itoh, Y.; Kawamata, Y.; Harada, M.; Kobayashi, M.; Fujii, R.; Fukusumi, S.; Ogi, K.; Hosoya, M.; Tanaka, Y.; Uejima, H. Free fatty acids regulate insulin secretion from pancreatic β cells through GPR40. Nature 2003, 422, 173–176.
  41. Kristinsson, H.; Bergsten, P.; Sargsyan, E. Free fatty acid receptor 1 (FFAR1/GPR40) signaling affects insulin secretion by enhancing mitochondrial respiration during palmitate exposure. Biochim. Biophys. Acta (BBA)-Mol. Cell Res. 2015, 1853, 3248–3257.
  42. Hirasawa, A.; Tsumaya, K.; Awaji, T.; Katsuma, S.; Adachi, T.; Yamada, M.; Sugimoto, Y.; Miyazaki, S.; Tsujimoto, G. Free fatty acids regulate gut incretin glucagon-like peptide-1 secretion through GPR120. Nat. Med. 2005, 11, 90–94.
  43. Lin, H.V.; Frassetto, A.; Kowalik Jr, E.J.; Nawrocki, A.R.; Lu, M.M.; Kosinski, J.R.; Hubert, J.A.; Szeto, D.; Yao, X.; Forrest, G. 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.
  44. 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.
  45. Guo, Y.; Xiao, Z.; Wang, Y.; Yao, W.; Liao, S.; Yu, B.; Zhang, J.; Zhang, Y.; Zheng, B.; Ren, B. Sodium butyrate ameliorates streptozotocin-induced type 1 diabetes in mice by inhibiting the HMGB1 expression. Front. Endocrinol. 2018, 9, 630.
  46. Tang, R.; Li, L. Modulation of short-chain fatty acids as potential therapy method for type 2 diabetes mellitus. Can. J. Infect. Dis. Med. Microbiol. 2021, 2021, 6632266.
  47. Fischbach, M.A.; Sonnenburg, J.L. Eating for two: How metabolism establishes interspecies interactions in the gut. Cell Host Microbe 2011, 10, 336–347.
  48. Macfarlane, S.; Macfarlane, G.T. Regulation of short-chain fatty acid production. Proc. Nutr. Soc. 2003, 62, 67–72.
  49. Macy, J.M.; Ljungdahl, L.G.; Gottschalk, G. Pathway of succinate and propionate formation in Bacteroides fragilis. J. Bacteriol. 1978, 134, 84–91.
  50. Macy, J.M.; Probst, I. The biology of gastrointestinal bacteroides. Annu. Rev. Microbiol. 1979, 33, 561–594.
  51. Pryde, S.E.; Duncan, S.H.; Hold, G.L.; Stewart, C.S.; Flint, H.J. The microbiology of butyrate formation in the human colon. FEMS Microbiol. Lett. 2002, 217, 133–139.
  52. Ragsdale, S.W.; Pierce, E. Acetogenesis and the Wood–Ljungdahl pathway of CO2 fixation. Biochim. Biophys. Acta (BBA)-Proteins Proteom. 2008, 1784, 1873–1898.
  53. Duncan, S.H.; Louis, P.; Flint, H.J. Lactate-utilizing bacteria, isolated from human feces, that produce butyrate as a major fermentation product. Appl. Environ. Microbiol. 2004, 70, 5810–5817.
  54. Louis, P.; Flint, H.J. Diversity, metabolism and microbial ecology of butyrate-producing bacteria from the human large intestine. FEMS Microbiol. Lett. 2009, 294, 1–8.
  55. Duncan, S.H.; Barcenilla, A.; Stewart, C.S.; Pryde, S.E.; Flint, H.J. Acetate utilization and butyryl coenzyme A (CoA): Acetate-CoA transferase in butyrate-producing bacteria from the human large intestine. Appl. Environ. Microbiol. 2002, 68, 5186–5190.
  56. Venema, K. Role of gut microbiota in the control of energy and carbohydrate metabolism. Curr. Opin. Clin. Nutr. Metab. Care 2010, 13, 432–438.
  57. Duncan, S.H.; Holtrop, G.; Lobley, G.E.; Calder, A.G.; Stewart, C.S.; Flint, H.J. Contribution of acetate to butyrate formation by human faecal bacteria. Br. J. Nutr. 2004, 91, 915–923.
  58. Malard, F.; Dore, J.; Gaugler, B.; Mohty, M. Introduction to host microbiome symbiosis in health and disease. Mucosal Immunol. 2021, 14, 547–554.
  59. Den Besten, G.; Van Eunen, K.; Groen, A.K.; Venema, K.; Reijngoud, D.-J.; Bakker, B.M. The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J. Lipid Res. 2013, 54, 2325–2340.
  60. El-Khoury, A.E.; Sánchez, M.; Fukagawa, N.K.; Gleason, R.E.; Young, V.R. Similar 24-h pattern and rate of carbon dioxide production, by indirect calorimetry vs. stable isotope dilution, in healthy adults under standardized metabolic conditions. J. Nutr. 1994, 124, 1615–1627.
  61. Xu, Y.; Wang, N.; Tan, H.-Y.; Li, S.; Zhang, C.; Feng, Y. Function of Akkermansia muciniphila in obesity: Interactions with lipid metabolism, immune response and gut systems. Front. Microbiol. 2020, 11, 219.
  62. Derrien, M.; Vaughan, E.E.; Plugge, C.M.; de Vos, W.M. Akkermansia muciniphila gen. nov., sp. nov., a human intestinal mucin-degrading bacterium. Int. J. Syst. Evol. Microbiol. 2004, 54, 1469–1476.
  63. Zhai, Q.; Feng, S.; Arjan, N.; Chen, W. A next generation probiotic, Akkermansia muciniphila. Crit. Rev. Food Sci. Nutr. 2019, 59, 3227–3236.
  64. Huang, K.; Wang, M.M.; Kulinich, A.; Yao, H.L.; Ma, H.Y.; Martínez, J.E.; Duan, X.C.; Chen, H.; Cai, Z.P.; Flitsch, S.L. Biochemical characterisation of the neuraminidase pool of the human gut symbiont Akkermansia muciniphila. Carbohydr. Res. 2015, 415, 60–65.
  65. Ottman, N.; Huuskonen, L.; Reunanen, J.; Boeren, S.; Klievink, J.; Smidt, H.; Belzer, C.; De Vos, W.M. Characterization of outer membrane proteome of Akkermansia muciniphila reveals sets of novel proteins exposed to the human intestine. Front. Microbiol. 2016, 7, 1157.
  66. Collado, M.C.; Derrien, M.; Isolauri, E.; de Vos, W.M.; Salminen, S. Intestinal integrity and Akkermansia muciniphila, a mucin-degrading member of the intestinal microbiota present in infants, adults, and the elderly. Appl. Environ. Microbiol. 2007, 73, 7767–7770.
  67. Wang, L.; Christophersen, C.T.; Sorich, M.J.; Gerber, J.P.; Angley, M.T.; Conlon, M.A. Low relative abundances of the mucolytic bacterium Akkermansia muciniphila and Bifidobacterium spp. in feces of children with autism. Appl. Environ. Microbiol. 2011, 77, 6718–6721.
  68. Hansen, C.; Krych, L.; Nielsen, D.; Vogensen, F.; Hansen, L.; Sørensen, S.; Buschard, K.; Hansen, A. Early life treatment with vancomycin propagates Akkermansia muciniphila and reduces diabetes incidence in the NOD mouse. Diabetologia 2012, 55, 2285–2294.
  69. Corb Aron, R.; Abid, A.; Vesa, C.; Nechifor, A.; Behl, T.; Ghitea, T.; Munteanu, M.; Fratila, O.; Andronie-Cioara, F.; Toma, M. Recognizing the Benefits of Pre-/Probiotics in Metabolic Syndrome and Type 2 Diabetes Mellitus Considering the Influence of Akkermansia muciniphila as a Key Gut Bacterium. Microorganisms 2021, 9, 618.
  70. y Abreu, A.T.A.; Milke-García, M.; Argüello-Arévalo, G.; Calderón-de la Barca, A.; Carmona-Sánchez, R.; Consuelo-Sánchez, A.; Coss-Adame, E.; García-Cedillo, M.; Hernández-Rosiles, V.; Icaza-Chávez, M. Dietary fiber and the microbiota: A narrative review by a group of experts from the Asociación Mexicana de Gastroenterología. Rev. Gastroenterol. Méx. (Engl. Ed.) 2021, 86, 287–304.
  71. Li, L.; Pan, M.; Pan, S.; Li, W.; Zhong, Y.; Hu, J.; Nie, S. Effects of insoluble and soluble fibers isolated from barley on blood glucose, serum lipids, liver function and caecal short-chain fatty acids in type 2 diabetic and normal rats. Food Chem. Toxicol. 2020, 135, 110937.
  72. Cao, Y.; Yao, G.; Sheng, Y.; Yang, L.; Wang, Z.; Yang, Z.; Zhuang, P.; Zhang, Y. JinQi Jiangtang tablet regulates gut microbiota and improve insulin sensitivity in type 2 diabetes mice. Journal of diabetes research 2019, 2019, 1872134.
  73. Adachi, K.; Sugiyama, T.; Yamaguchi, Y.; Tamura, Y.; Izawa, S.; Hijikata, Y.; Ebi, M.; Funaki, Y.; Ogasawara, N.; Goto, C. Gut microbiota disorders cause type 2 diabetes mellitus and homeostatic disturbances in gut-related metabolism in Japanese subjects. J. Clin. Biochem. Nutr. 2019, 64, 231–238.
  74. 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.
  75. Yamaguchi, Y.; Adachi, K.; Sugiyama, T.; Shimozato, A.; Ebi, M.; Ogasawara, N.; Funaki, Y.; Goto, C.; Sasaki, M.; Kasugai, K. Association of intestinal microbiota with metabolic markers and dietary habits in patients with type 2 diabetes. Digestion 2016, 94, 66–72.
  76. Pingitore, A.; Chambers, E.S.; Hill, T.; Maldonado, I.R.; Liu, B.; Bewick, G.; Morrison, D.J.; Preston, T.; Wallis, G.A.; Tedford, C. 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.
  77. Sanna, S.; van Zuydam, N.R.; Mahajan, A.; Kurilshikov, A.; Vich Vila, A.; Võsa, U.; Mujagic, Z.; Masclee, A.A.; Jonkers, D.M.; Oosting, M. Causal relationships among the gut microbiome, short-chain fatty acids and metabolic diseases. Nat. Genet. 2019, 51, 600–605.
  78. 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.
  79. Saboo, B.; Misra, A.; Kalra, S.; Mohan, V.; Aravind, S.; Joshi, S.; Chowdhury, S.; Sahay, R.; Kesavadev, J.; John, M. Role and importance of high fiber in diabetes management in India. Diabetes Metab. Syndr. Clin. Res. Rev. 2022, 16, 102480.
  80. Kondo, T.; Kishi, M.; Fushimi, T.; Kaga, T. Acetic acid upregulates the expression of genes for fatty acid oxidation enzymes in liver to suppress body fat accumulation. J. Agric. Food Chem. 2009, 57, 5982–5986.
  81. Yamashita, H.; Maruta, H.; Jozuka, M.; Kimura, R.; Iwabuchi, H.; Yamato, M.; Saito, T.; Fujisawa, K.; Takahashi, Y.; Kimoto, M. Effects of acetate on lipid metabolism in muscles and adipose tissues of type 2 diabetic Otsuka Long-Evans Tokushima Fatty (OLETF) rats. Biosci. Biotechnol. Biochem. 2009, 73, 570–576.
  82. Yamashita, H.; Fujisawa, K.; Ito, E.; Idei, S.; Kawaguchi, N.; Kimoto, M.; Hiemori, M.; Tsuji, H. Improvement of obesity and glucose tolerance by acetate in Type 2 diabetic Otsuka Long-Evans Tokushima Fatty (OLETF) rats. Biosci. Biotechnol. Biochem. 2007, 71, 1236–1243.
  83. 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. 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.
  84. 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.
  85. Larraufie, P.; Martin-Gallausiaux, C.; Lapaque, N.; Dore, J.; Gribble, F.; Reimann, F.; Blottiere, H. SCFAs strongly stimulate PYY production in human enteroendocrine cells. Sci. Rep. 2018, 8, 74.
  86. 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.
  87. Al-Lahham, S.a.H.; Roelofsen, H.; Priebe, M.; Weening, D.; Dijkstra, M.; Hoek, A.; Rezaee, F.; Venema, K.; Vonk, R.J. Regulation of adipokine production in human adipose tissue by propionic acid. Eur. J. Clin. Investig. 2010, 40, 401–407.
  88. Canfora, E.E.; van der Beek, C.M.; Jocken, J.W.; Goossens, G.H.; Holst, J.J.; Olde Damink, S.W.; Lenaerts, K.; Dejong, C.H.; 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.
  89. Ohira, H.; Fujioka, Y.; Katagiri, C.; Mamoto, R.; Aoyama-Ishikawa, M.; Amako, K.; Izumi, Y.; Nishiumi, S.; Yoshida, M.; Usami, M. Butyrate attenuates inflammation and lipolysis generated by the interaction of adipocytes and macrophages. J. Atheroscler. Thromb. 2013, 20, 425–442.
  90. Al-Lahham, S.A.; 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.
More
Information
Subjects: Microbiology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 337
Entry Collection: Gastrointestinal Disease
Revisions: 3 times (View History)
Update Date: 22 May 2023
1000/1000