Food Groups' Effects on Gut Microbiota and T2DM: History
Please note this is an old version of this entry, which may differ significantly from the current revision.

Nutrition is one of the most influential environmental factors in both taxonomical shifts in gut microbiota as well as in the development of type 2 diabetes mellitus (T2DM). Considering the effects of macro- and micronutrients on gut microbiota and T2DM, food groups and dietary patterns are major determinants of the gut microbiota–metabolic disorder axis. 

  • gut bacteria
  • macronutrients
  • micronutrients
  • food groups
  • insulin resistance

1. Cereals and Cereal Products

Cereals are composed of whole grains, wheat, oats, rye, and barley, which have been shown to have beneficial effects on metabolic health and gut microbiota [1][2]. Over the years, an increasing number of studies have elucidated the effects of cereal products on T2DM, largely showing that the components within cereals decrease the risk of developing the disease and related sequelae [3][4][5][6]. Dietary recommendations for cereals in metabolic disease include increasing the intake of cereals with whole grains and limiting the intake of refined grains or cereals with processed sugars and artificial sweeteners [7]. Both whole wheat intake and the intake of barley, oat, and rye have been associated with improved blood glucose levels [8][9] and increased insulin sensitivity [10]. It should be noted that the beneficial effects of cereals appear when the intake is high, at least 4 g of β-glucans daily [9][11]. For example, a supplement of up to 50 g of whole grains per day was associated with a 25% decrease in the risk of T2DM [12]. Similarly, the consumption of two servings of whole grains per day was associated with a 21% decrease in the risk of T2DM [13], while a refined grain intake of 200–400 g per day was associated with a 6–14% increase in the risk of T2DM [12]. The composition of the whole grains such as magnesium, phytochemicals, isoflavins, and lignins was also associated with beneficial effects in T2DM [14]. Adding cereal fiber to meals reduced post-prandial insulin release, indicating the important roles of fiber in improving insulin sensitivity [6]. Taken together, these data provide strong evidence for cereal products in reducing the risk of T2DM development.
Further, cereal-based dietary approaches are shown to affect multiple metabolic parameters in individuals already diagnosed with T2DM, some of which may be related to changes in gut microbiota. For example, after a 3-month adherence to high dietary fiber-based cereals, there were favorable trends in lipids, HgbA1, body mass index (BMI), adipose distribution, and fasting C-peptide levels [3]. Similarly, high fiber rye, a component of healthy cereal, is shown to improve similar parameters when compared to refined wheats [4]. In addition to improving metabolic parameters, the high fiber rye diet produced important changes in gut microbiota, including elevated SCFA-producing Agathobacter and decreased Ruminococcus torques, with associated increases in plasma butyrate concentrations [4]. When compared to refined grains, the whole grain has immunomodulatory effects that were associated with microbial composition alterations [15]. For example, the introduction of wheat grains after a 2-week Western-style diet improved SCFAs, increased SCFA-producing Lachnospira, and reduced the pro-inflammatory Enterobacteriaceae family which correlated with positive changes in effector memory T-cell activity and acute innate immune response [15]. Other immunomodulatory effects of cereals have also been described in the literature, with the reduced activity of pro-inflammatory cytokines, TNF-α and IL-6, being observed after consumption [16][17]. In rodent studies, the effects of wheat also improved GPR41/43 receptor expression and enhanced GLP-1 secretion with concomitant increases in SCFA-producing bacteria, providing further insights into the multitude of effects that cereals have on metabolic disease [18].
In addition to the changes described above, it seems that a general increase in Bifidobacterium and Lactobacillus spp. is common after cereal consumption, an effect consistent with other recent studies [5]. Previous studies have also shown that diets rich in whole wheat compared to refined wheats exhibit an abundance of Bifidobacterium and relative decreases in Bacteroides after a 12-week intervention [19]. The type of cereal consumed is also important in determining microbiota shifts. For example, an increase in the abundance of Bifidobacterium and Lactobacillus was seen in the gut microbiota of people who consumed whole grain cereals for breakfast, compared to the microbiota of people who consumed cereals based on wheat bran [20]. A corn-based cereal diet increased the abundance of fecal Bifidobacteirum after a 3-week intervention, as compared to a refined-corn-based cereal [21]. These changes in the composition of the gut microbiota could be observed even at a low intake of whole corn (29.6% of the recommended total of 48 g [21]). However, the opposite is also true, with the sugar additives and processing seen in refined cereals having been shown to have negative effects on both gut microbial composition and related metabolites [22]. Therefore, it is evident that eating cereals with naturally occurring fibers can be beneficial in preventing or treating metabolic derangements in T2DM, while avoiding refined cereals and cereals with additives is also important.

2. Fruits and Vegetables

In general, some of the healthiest foods are considered vegetables and fruits, due to their content of dietary fiber, vitamins, minerals, and flavonoids [23]. Multiple studies have demonstrated the inverse associations between the consumption of green leafy vegetables and the risk of developing T2DM [24][25][26], the consumption of fruits and T2DM [27], as well as the intake of mixed fruits and vegetables and T2DM [28]. Specifically, an intake of 0.2 servings per day of green leafy vegetables reduced the risk for type 2 diabetes by 13% [25], with similar findings in another meta-analysis showing a risk reduction of 14% [26]. Changes in microbial shifts after the consumption of fruits and vegetables have also been described with study findings showing a decreased abundance of the Lachnospiraceae family, including Ruminococcus, and increased concentrations of Faecalibacterium and Lactobacillus [29]. Further metagenomic sequencing studies combining two large human cohorts have shown changes that include an increased abundance of Faecalibacterium prausnitzii, Akkermansia muciniphila, Ruminococcaceae, Clostridiales, and Acidaminococcus and a decrease in the abundance of Fusobacterium [30].
Vegetables and fruits are sources of antioxidants that have been associated with augmenting glucose metabolism, by improving oxidative stress [31], particularly given their high content of flavonoids and polyphenols [32]. Interestingly, flavonoids are shown to modulate gut microbiota-related metabolic processes, particularly through the suppression of lipogenesis and the upregulation of lipolysis, via the FXR pathway in bile acid metabolism [33]. These effects of flavonoids were corelated with increased Akkermansia and reductions in Lachnoclostridium, Desulfovibrio, Colidextribacter, and Blautia, all of which are strongly associated with metabolic parameters [33]. Further, flavonoid-based dietary interventions alleviated inflammation as measured through LPS/TLR-4, TNF-α, IL-6, and IL-10, while also improving insulin resistance, HgbA1c, and oral glucose tolerance [34]. Interestingly, GLP-1 release was also enhanced following flavonoid introduction. The beneficial effects of fruit and vegetable flavonoids also are shown by the improvement of intestinal barrier integrity, as well as promoting islet cell proliferation and the suppression of islet cell apoptosis [35]. Flavonoids also modulate glucose metabolism by the upregulation of the IRS/AKT signaling pathway to increase GLUT4 translocation and the synthesis of glycogen, while concomitantly improving the Firmicutes-to-Bacteroidetes ratio [36]. As such, flavonoids, a major component of fruits and vegetables, exert a multitude of metabolic benefits at the intersection between gut microbiota and glucose homeostasis.
In addition to flavonoids, fruits and vegetables are comprised of other beneficial bioactive phytochemical-based nutrients, including vitamin C and carotenoids, which contribute to insulin sensitivity [37][38]. Also, green leafy vegetables contain magnesium which is inversely associated with an increased risk for type 2 diabetes [39]. The association between fruit and vegetable intake and a reduced risk of type 2 diabetes may be due to their dietary fiber content [40] and the subsequent effects of weight loss in overweight individuals [41]. Fruit and vegetable juices, depending on their content, may have differing outcomes on both gut microbiota and T2DM [42][43][44][45]. Fruit juices that are altered by added sugar or artificial sweeteners pose harmful risks to the gut metabolic profile [42]. For example, the artificial sweetening of fruit beverages results in modest changes in gut microbiota, particularly in the ratio of Firmicutes to Bacteroidetes [42]. However, the introduction of natural fruit or vegetable extracts or juice generally has favorable effects [43][44][45]. In a prediabetic rodent model, blueberry juice improved the microbiota composition as well as metabolic parameters including insulin signaling, inflammation, ketogenesis, and fatty acid oxidation [44]. Similarly, pomegranate juice can reduce the post-prandial glycemic response after eating a high-carbohydrate meal, primarily breads [46]. Overall, fruits and vegetables are an important food group in maintaining a healthy microbiota profile because diets high in fruits, vegetables, legumes, and whole grains are accompanied by optimal body weight, reduced inflammation, and lower insulin resistance.

3. Milk and Dairy Products

Dairy products are rich in protein, B vitamins, and minerals, such as calcium, magnesium, potassium, phosphorus, and zinc, all of which have important effects on gut microbiota composition [47]. Dairy proteins, especially whey proteins, are associated with improved insulin sensitivity and a reduced risk of type 2 diabetes [48]. Interestingly, high quantities of dairy consumption (two servings per day) in adolescence were associated with a 38% decreased risk of developing T2DM in middle-aged women [49]. Further, an inverse correlation was observed between the intake of skimmed or semi-skimmed dairy products and the risk of type 2 diabetes [50]. This decreased risk was seen with 200 g of skimmed dairy product intake, with an improvement in risk up to 6% with every additional 200 g, up to a daily total of 600 g [12]. Another study has shown that one serving of dairy per day has beneficial effects on T2DM risk reduction of 9% in men and 4% in women [51][52]. Dairy consumption produces specific compositional changes in gut microbiota. For example, the introduction of dairy products or intake of yogurt for three weeks led to decreased Bacteroides fragilis [47] and an abundant growth of Lactobacillus and Bifidobacterium [53]. Similarly, the consumption of kefir, a yogurt-based drink, over the next 4 weeks increased the abundance of Lactobacillus [54][55], with associated elevated levels in fecal SCFAs [55]. Interestingly, in studies on murine models, yogurt-derived Lactobacillus plantarum has been shown to ameliorate the reduction in pancreatic β-cell mass with notable improvements in insulin resistance [56]. Taken together, these studies show that dairy consumption prompts significant changes in the composition of gut microbiota that are beneficial to the host in mitigating the deleterious effects of T2DM.

4. Meat and Meat Products

The recommendations for patients with type 2 diabetes regarding the intake of meat and meat products are similar to the recommendations for healthy individuals, i.e., one portion/day or the equivalent of 100–150 g of lean meat per day [57]. Lean meat and meat products are sources of protein with high biological value, but they are also important sources of iron and vitamin B12 [58]. However, red meats are shown to exert negative effects in both contributing to T2DM development and worsening the condition [57]. Several positive associations have been reported between the intake of processed red meats and increased blood glucose concentrations, insulin levels, and risk for obesity [59][60]. Moreover, the risk for type 2 diabetes was associated with the intake of red meat up to 100 g per day [12] but also with the intake of up to 50 g per day of processed meat products [12][61]. These effects have been attributed to the content of heterocyclic amines and nitrates affecting glucose metabolism [62][63]. These metabolites contribute to insulin resistance through adverse effects on pancreatic β-cell function and insulin-like growth factor (IGF-1) [64]. Further, these inorganic nitrates, present in processed meats, promote DNA damage through conversion to cytotoxic agents such as peroxy-nitrite as well as reactive oxygen species, which increase pro-inflammatory cytokine production and hamper glucose homeostasis [65]. Red meats also enhance the presence of dietary advanced glycosylated end products (dAGEs), the result of the Maillard reaction that occurs between amino acids and reducing sugars [66]. These dAGE products are shown to increase insulin resistance, while a restricted intake of dietary glycoxidation products improved insulin sensitivity in diabetic mice [67]. Additionally, it has been shown that hyperglycemia further enhances the glycation process, thus worsening the complications of uncontrolled diabetes. Therefore, red meats are a source of inorganic nitrates and substrates for the generation of dAGEs, which may contribute to the development of insulin resistance and complicate pre-existing diabetes.
Red meat may also be detrimental to gut microbial composition. It has been shown that red meat decreases Lactobacillus, Paralactobacillus, and Prevotella, while also decreasing SCFAs in animal models [68]. Further, the administration of beef, a red meat derivate, in mouse and rat study models led to an increase in the amount of Clostridium and Blautia and a decrease in the amount of Bifidobacterium and Akkermansia [69]. The addition of butyrate containing starch was shown to reverse the negative effects of red meat diet adherence through increased abundances of Clostridium coccoides, Clostridium leptum, Lactobacillus spp., Parabacteroides distasonis, and Ruminococcus bromii, but it also showed a decrease in the amount of Ruminococcus torques, Ruminococcus gnavus, and Escherichia coli [70]. However, the effects on gut microbial composition are dependent on the type of meat and proteins they contain [71]. For example, a study evaluating the gut microbiota of individuals consuming chicken meat is characterized by the highest proportion of Prevotella 9 (22.45%), followed by Dialister, Faecalibacterium, Megamonas, Prevotella, Roseburia, Alloprevotella, Ruminococcaceae, Eubacterium, and Succinivibrio, while the gut microbiota of individuals consuming pork is characterized by the highest proportion of Bacteroides (17.3%), followed by Faecalibacterium, Roseburia, Dialister, Ruminococcus, Blautia, Megamonas, Agathobacter, Subdoligranulum, and Eubacterium [72]. On the other hand, pork intake decreased the amount of Blautia, Bifidobacterium, and Alistipes and increased the amount of Akkermansia muciniphila and Ruminococcaceae [73]. Collectively, the intake of pork meat induced low-grade inflammation and induced oxidative stress and the upregulation of lipid metabolism genes such as PPAR-α and PPAR-γ [73]. Further, an increase in the abundance of Lactobacillus and a decrease in SCFA levels and SCFA-producing bacterial species such as Fusobacterium, Bacteroides, and Prevotella have been reported in laboratory mice fed beef, pork, or fish proteins, compared to mice that were given protein from sources other than meat, such as soy or casein [74]. Similarly, laboratory rats fed chicken meat had the highest abundance of Lactobacillus, compared to laboratory rats fed soy, which had the highest abundance of Ruminococcus and the lowest abundance of Lactobacillus [75]. The administration of beef in mouse and rat study models led to an increase in the amount of Clostridium and Blautia and a decrease in the amount of Bifidobacterium and Akkermansia [69]. Collectively, these changes indicate that meats derived from chicken have more favorable effects on gut microbiota and insulin resistance as compared to pork and red meats.

5. Nuts, Oils, and Oilseeds

Tree nuts have been shown to exert favorable effects on gut microbiota and metabolic parameters [76][77]. For example, replacing starchy foods with peanuts or almonds in patients with type 2 diabetes led to improvements in blood glucose, HgbA1c, and inflammatory markers [76]. In addition, the daily intake of raw or roasted almonds for 4 weeks promoted Bifidobacterium spp. and Lactobacillus spp. and inhibited the growth of Enterococcus spp. Interestingly, the administration of raw almonds had a greater Bifidobacteria-promoting effect than roasted almonds, with both roasted and raw almonds having a potential prebiotic effect, including regulating gut bacteria and improving metabolic activities [77]. Similarly, nut intake promotes an increase in the abundance of Faecalibacterium, Clostridium, Dialister, and Roseburia and a decrease in the abundance of Ruminococcus, Dorea, Oscillopira, and Bifidobacterium [78]. Pistachio consumption led to an increase in the abundance of potentially beneficial, butyrate-producing bacteria [79], while eating whole, roasted, or chopped almonds is associated with an increase in the abundance of Lachnospira and Roseburia [80]. These alterations in gut microbiota were associated with a concomitant decrease in pro-inflammatory secondary bile acid production and LDL cholesterol, two interrelated parameters in the development of hyperglycemia and insulin resistance [78]. Also, a diet enriched with 20% peanut protein was effective in increasing the amount of Bifidobacterium and reducing the amount of Enterobacteria and Clostridium perfringens in rats [81].
Oilseeds are important sources of polyunsaturated and monounsaturated fatty acids [82][83] and their consumption has been associated with a decreased risk for type 2 diabetes [84]. For example, dietary flaxseed oil, given its rich composition of omega-3, was associated with decreased Firmicutes and pro-inflammatory markers such as IL-1β, TNF-α, and IL-6 and increased Bacteroidetes and Alistipes that negatively correlate with LPS production [85]. Further, the direct markers of hyperglycemia showed significant improvement, particularly in fasting blood glucose and glycated hemoglobin. Interestingly, superoxide dismutase (SOD) activity was increased as well, with previous studies showing that SOD activity can improve diabetes-induced mitochondrial electron transport dysfunction and diabetes complications such as retinopathy [86]. Meta-analyses of human studies confirm these beneficial anti-inflammatory effects of oilseeds, with decreased CRP and IL-6 activity leading to improved endothelial function and metabolic activity [87]. Oilseeds cause significant changes in the gut composition profile, such as increased Lactobacillus spp. and SCFAs, with reduced production of harmful metabolites such as TMAO [88]. As such, oilseeds and nuts serve as healthy food sources that are intricate components of the Mediterranean diet and modulate important metabolic processes associated with T2DM. A summary of food groups and the mechanisms by which they impact T2DM is presented in Table 1.
Table 1. Food groups and resulting effects on T2DM.
Food Group Study Period Outcome Measured Results/Implications Subject Type Reference
Cereals and Cereal Products Meta-Analysis Diabetes risk Two servings of whole grains decreased the risk of developing T2DM by 21%
Refined grain intake increased the risk of developing T2DM by 6–14%
Humans [12]
3 months Metabolic Parameters Improvements in lipid quality, HgbA1c, BMI, adipose distribution, and fasting C-peptide levels Humans [3]
6 weeks Gut Microbiota and Inflammatory Markers Whole grains improved effector memory T-cell activity and acute innate immune response
Increased quantity of SCFAs and SCFA-producing genera including Lachnospira
Decreased relative abundances of pro-inflammatory bacterial family Enterobacteriaceae
Humans [15]
9 weeks Gut Microbiota and Gut Hormones Increased SCFA-producing species
Increased GLP-1 secretion
Mice [18]
12 weeks Gut Microbiota Increased Bifidobacterium and decreased Bacteroides Humans [19]
Fruits and Vegetables Meta-Analysis Diabetes risk Intake of 0.2 servings per day of green leafy vegetables reduced the risk for type 2 diabetes by 13% Humans [25]
  Gut Microbiota and Metabolic Parameters Increased Akkermansia
Reduced Lachnoclostridium, Desulfovibrio, Colidextribacter, and Blautia
Upregulation of lipolysis through the FXR, bile acid metabolism pathway
Mice [33]
  Inflammatory Markers and Metabolic Parameters Reduced LPS/TLR-4 activity, TNF-α, and IL-6
Improved IL-10
Improved insulin resistance and HgbA1c
Increased GLP-1 secretion
Mice [34]
  Glucose Metabolism and Gut Microbiota Upregulation of the IRS/AKT signaling pathway to increase GLUT4 translocation and synthesis of glycogen
Improved Firmicutes-to-Bacteroidetes ratio
Mice [36]
Milk and Dairy Products   Diabetes risk One serving of dairy per day has beneficial effects on T2DM risk reduction of 9% in men and 4% in women Humans [51][52]
3 weeks Gut Microbiota Increased Bifidobacterium and Lactobacillus spp. Increased serum IgA
Decreased Bacteroides fragilis
Humans [47][53]
  Gut Microbiota and Pancreatic Function Lactobacillus isolated from yogurt increased SCFA levels and SCFA receptors, GPR41/43
Increased SCFA-producing genera
Inhibited reduction of β-cell mass
Mice [56]
Meat and Meat Products Meta-Analysis Diabetes Risk Risk for T2DM is increased with intake of 100 g of red meat per day
Risk for T2DM is increased with intake of 50 g of processed meat per day
Humans [12][61]
  Gut Microbiota Red meat decreases Lactobacillus, Paralactobacillus, and Prevotella, while also decreasing SCFAs Dogs [68]
1–4 weeks Gut Microbiota Increased Clostridium and Blautia
Decreased Bifidobacterium and Akkermansia
Mice [69]
3 months Gut Microbiota, Inflammatory and Metabolic parameters Pork meat decreased Blautia, Bifidobacterium, and Alistipes
Induced low-grade inflammation
Induced oxidative stress
Upregulated lipid metabolism genes including PPAR-α and PPAR-γ
Mice [73]
Nuts, Oils and Oilseeds 3 months Parameters of T2DM Peanuts or almonds in patients with T2DM improved blood glucose, HgbA1c, and inflammatory markers like IL-6 expression Humans [76]
6 weeks Gut Microbiota and Metabolic Parameters Nut intake increased the abundance of Faecalibacterium, Clostridium, Dialister, and Roseburia and decreased the abundance of Ruminococcus, Dorea, Oscillopira, and Bifidobacterium
Decreased pro-inflammatory bile acid production and LDL cholesterol
Humans [78]
5 weeks Gut Microbiota and Inflammatory Parameters Dietary flaxseed oil decreased severity of T2DM, improved the Firmicutes-to-Bacteroidetes ratio, while increasing Alistipes
Reduction in IL-1β, TNF-α, IL-6, and LPS production
Rats [85]
Abbreviations: T2DM, type 2 diabetes mellitus, HgbA1c, Hemoglobin A1c; BMI, body mass index; SCFA, short-chain fatty acid; GLP-1, glucagon-like peptide 1; FXR, Farsenoid X Receptor; LPS, lipopolysaccharides; TLR-4, Toll-like receptor 4; TNF-α, Tumor Necrosis Factor alpha; IL, interleukin; GLUT4, Glucose Transporter 4; IRS, insulin receptor substrate; IgA, Immunoglobulin A; PPAR, peroxisome proliferator-activated receptor; LDL, low density lipoprotein.

This entry is adapted from the peer-reviewed paper 10.3390/nu16020269

References

  1. Kulathunga, J.; Simsek, S. A Review: Cereals on Modulating the Microbiota/Metabolome for Metabolic Health. Curr. Nutr. Rep. 2022, 11, 371–385.
  2. van Trijp, M.P.H.; Schutte, S.; Esser, D.; Wopereis, S.; Hoevenaars, F.P.M.; Hooiveld, G.; Afman, L.A. Minor Changes in the Composition and Function of the Gut Microbiota During a 12-Week Whole Grain Wheat or Refined Wheat Intervention Correlate with Liver Fat in Overweight and Obese Adults. J. Nutr. 2021, 151, 491–502.
  3. Li, X.; Shi, Y.; Wei, D.; Ni, W.; Zhu, N.; Yan, X. Impact of a high dietary fiber cereal meal intervention on body weight, adipose distribution, and cardiovascular risk among individuals with type 2 diabetes. Front. Endocrinol. 2023, 14, 1283626.
  4. Iversen, K.N.; Dicksved, J.; Zoki, C.; Fristedt, R.; Pelve, E.A.; Langton, M.; Landberg, R. The Effects of High Fiber Rye, Compared to Refined Wheat, on Gut Microbiota Composition, Plasma Short Chain Fatty Acids, and Implications for Weight Loss and Metabolic Risk Factors (the RyeWeight Study). Nutrients 2022, 14, 1669.
  5. Fava, F.; Ulaszewska, M.M.; Scholz, M.; Stanstrup, J.; Nissen, L.; Mattivi, F.; Vermeiren, J.; Bosscher, D.; Pedrolli, C.; Tuohy, K.M. Impact of wheat aleurone on biomarkers of cardiovascular disease, gut microbiota and metabolites in adults with high body mass index: A double-blind, placebo-controlled, randomized clinical trial. Eur. J. Nutr. 2022, 61, 2651–2671.
  6. Gaesser, G.A.; Rodriguez, J.; Patrie, J.T.; Whisner, C.M.; Angadi, S.S. Effects of Glycemic Index and Cereal Fiber on Postprandial Endothelial Function, Glycemia, and Insulinemia in Healthy Adults. Nutrients 2019, 11, 2387.
  7. Williams, P.G. The benefits of breakfast cereal consumption: A systematic review of the evidence base. Adv. Nutr. 2014, 5, 636s–673s.
  8. Harris Jackson, K.; West, S.G.; Vanden Heuvel, J.P.; Jonnalagadda, S.S.; Ross, A.B.; Hill, A.M.; Grieger, J.A.; Lemieux, S.K.; Kris-Etherton, P.M. Effects of whole and refined grains in a weight-loss diet on markers of metabolic syndrome in individuals with increased waist circumference: A randomized controlled-feeding trial. Am. J. Clin. Nutr. 2014, 100, 577–586.
  9. Tosh, S.M. Review of human studies investigating the post-prandial blood-glucose lowering ability of oat and barley food products. Eur. J. Clin. Nutr. 2013, 67, 310–317.
  10. Pereira, M.A.; Jacobs, D.R., Jr.; Pins, J.J.; Raatz, S.K.; Gross, M.D.; Slavin, J.L.; Seaquist, E.R. Effect of whole grains on insulin sensitivity in overweight hyperinsulinemic adults. Am. J. Clin. Nutr. 2002, 75, 848–855.
  11. Nilsson, A.C.; Ostman, E.M.; Granfeldt, Y.; Björck, I.M. Effect of cereal test breakfasts differing in glycemic index and content of indigestible carbohydrates on daylong glucose tolerance in healthy subjects. Am. J. Clin. Nutr. 2008, 87, 645–654.
  12. Schwingshackl, L.; Hoffmann, G.; Lampousi, A.M.; Knüppel, S.; Iqbal, K.; Schwedhelm, C.; Bechthold, A.; Schlesinger, S.; Boeing, H. Food groups and risk of type 2 diabetes mellitus: A systematic review and meta-analysis of prospective studies. Eur. J. Epidemiol. 2017, 32, 363–375.
  13. Seo, Y.S.; Lee, H.B.; Kim, Y.; Park, H.Y. Dietary Carbohydrate Constituents Related to Gut Dysbiosis and Health. Microorganisms 2020, 8, 427.
  14. Davison, K.M.; Temple, N.J. Cereal fiber, fruit fiber, and type 2 diabetes: Explaining the paradox. J. Diabetes Complicat. 2018, 32, 240–245.
  15. Vanegas, S.M.; Meydani, M.; Barnett, J.B.; Goldin, L.; Kane, A.; Rasmussen, H.; Brown, C.; Vangay, P.; Knights, D.; Jonnalagadda, S.; et al. Substituting whole grains for refined grains in a 6-wk randomized trial has a modest effect on gut microbiota and immune and inflammatory markers of healthy adults. Am. J. Clin. Nutr. 2017, 105, 635–650.
  16. Li, C.; Wang, X.; Sun, S.; Liu, S.; Huan, Y.; Li, R.; Liu, Q.; Cao, H.; Zhou, T.; Lei, L.; et al. Effects of a ready-to-eat cereal formula powder on glucose metabolism, inflammation, and gut microbiota in diabetic db/db mice. Food Sci. Nutr. 2020, 8, 4523–4533.
  17. Roager, H.M.; Vogt, J.K.; Kristensen, M.; Hansen, L.B.S.; Ibrügger, S.; Mærkedahl, R.B.; Bahl, M.I.; Lind, M.V.; Nielsen, R.L.; Frøkiær, H.; et al. Whole grain-rich diet reduces body weight and systemic low-grade inflammation without inducing major changes of the gut microbiome: A randomised cross-over trial. Gut 2019, 68, 83–93.
  18. Liu, J.; Zhang, D.; Yang, Z.; Hao, Y.; Wang, Z.; Wang, J.; Wang, Z. Wheat Alkylresorcinols Modulate Glucose Homeostasis through Improving GLP-1 Secretion in High-Fat-Diet-Induced Obese Mice. J. Agric. Food Chem. 2023, 71, 16125–16136.
  19. Christensen, E.G.; Licht, T.R.; Kristensen, M.; Bahl, M.I. Bifidogenic effect of whole-grain wheat during a 12-week energy-restricted dietary intervention in postmenopausal women. Eur. J. Clin. Nutr. 2013, 67, 1316–1321.
  20. Costabile, A.; Klinder, A.; Fava, F.; Napolitano, A.; Fogliano, V.; Leonard, C.; Gibson, G.R.; Tuohy, K.M. Whole-grain wheat breakfast cereal has a prebiotic effect on the human gut microbiota: A double-blind, placebo-controlled, crossover study. Br. J. Nutr. 2008, 99, 110–120.
  21. Carvalho-Wells, A.L.; Helmolz, K.; Nodet, C.; Molzer, C.; Leonard, C.; McKevith, L.; Thielecke, F.; Jackson, K.G.; Tuohy, K.M. Determination of the in vivo prebiotic potential of a maize-based whole grain breakfast cereal: A human feeding study. Br. J. Nutr. 2010, 104, 1353–1356.
  22. Yu, D.; Zhu, L.; Gao, M.; Yin, Z.; Zhang, Z.; Zhu, L.; Zhan, X. A Comparative Study of the Effects of Whole Cereals and Refined Cereals on Intestinal Microbiota. Foods 2023, 12, 2847.
  23. Basiak-Rasała, A.; Różańska, D.; Zatońska, K. Food groups in dietary prevention of type 2 diabetes. Rocz. Państw. Zakł. Hig. 2019, 70, 347–357.
  24. Cooper, A.J.; Forouhi, N.G.; Ye, Z.; Buijsse, L.; Arriola, L.; Balkau, L.; Barricarte, A.; Beulens, J.W.; Boeing, H.; Büchner, F.L.; et al. Fruit and vegetable intake and type 2 diabetes: EPIC-InterAct prospective study and meta-analysis. Eur. J. Clin. Nutr. 2012, 66, 1082–1092.
  25. Li, M.; Fan, Y.; Zhang, X.; Hou, W.; Tang, Z. Fruit and vegetable intake and risk of type 2 diabetes mellitus: Meta-analysis of prospective cohort studies. BMJ Open 2014, 4, e005497.
  26. Carter, P.; Gray, L.J.; Troughton, J.; Khunti, K.; Davies, M.J. Fruit and vegetable intake and incidence of type 2 diabetes mellitus: Systematic review and meta-analysis. BMJ 2010, 341, c4229.
  27. Mursu, J.; Virtanen, J.K.; Tuomainen, T.P.; Nurmi, T.; Voutilainen, S. Intake of fruit, berries, and vegetables and risk of type 2 diabetes in Finnish men: The Kuopio Ischaemic Heart Disease Risk Factor Study. Am. J. Clin. Nutr. 2014, 99, 328–333.
  28. Wang, P.Y.; Fang, J.C.; Gao, Z.H.; Zhang, C.; Xie, S.Y. Higher intake of fruits, vegetables or their fiber reduces the risk of type 2 diabetes: A meta-analysis. J. Diabetes Investig. 2016, 7, 56–69.
  29. Lakshmanan, A.P.; Mingione, A.; Pivari, F.; Dogliotti, E.; Brasacchio, C.; Murugesan, S.; Cusi, D.; Lazzaroni, M.; Soldati, L.; Terranegra, A. Modulation of gut microbiota: The effects of a fruits and vegetables supplement. Front. Nutr. 2022, 9, 930883.
  30. Jiang, Z.; Sun, T.Y.; He, Y.; Gou, W.; Zuo, L.S.; Fu, Y.; Miao, Z.; Shuai, M.; Xu, F.; Xiao, C.; et al. Dietary fruit and vegetable intake, gut microbiota, and type 2 diabetes: Results from two large human cohort studies. BMC Med. 2020, 18, 371.
  31. Sotoudeh, G.; Abshirini, M.; Bagheri, F.; Siassi, F.; Koohdani, F.; Aslany, Z. Higher dietary total antioxidant capacity is inversely related to prediabetes: A case-control study. Nutrition 2018, 46, 20–25.
  32. Rienks, J.; Barbaresko, J.; Oluwagbemigun, K.; Schmid, M.; Nöthlings, U. Polyphenol exposure and risk of type 2 diabetes: Dose-response meta-analyses and systematic review of prospective cohort studies. Am. J. Clin. Nutr. 2018, 108, 49–61.
  33. Duan, R.; Guan, X.; Huang, K.; Zhang, Y.; Li, S.; Xia, J.; Shen, M. Flavonoids from Whole-Grain Oat Alleviated High-Fat Diet-Induced Hyperlipidemia via Regulating Bile Acid Metabolism and Gut Microbiota in Mice. J. Agric. Food Chem. 2021, 69, 7629–7640.
  34. Yang, T.; Zhou, W.; Xu, W.; Ran, L.; Yan, Y.; Lu, L.; Mi, J.; Zeng, X.; Cao, Y. Modulation of gut microbiota and hypoglycemic/hypolipidemic activity of flavonoids from the fruits of Lycium barbarum on high-fat diet/streptozotocin-induced type 2 diabetic mice. Food Funct. 2022, 13, 11169–11184.
  35. Su, M.; Tang, T.; Tang, W.; Long, Y.; Wang, L.; Liu, M. Astragalus improves intestinal barrier function and immunity by acting on intestinal microbiota to treat T2DM: A research review. Front. Immunol. 2023, 14, 1243834.
  36. Gong, X.; Xiong, L.; Bi, C.; Zhang, B. Diosmetin ameliorate type 2 diabetic mellitus by up-regulating Corynebacterium glutamicum to regulate IRS/PI3K/AKT-mediated glucose metabolism disorder in KK-Ay mice. Phytomedicine 2021, 87, 153582.
  37. Tarwadi, K.; Agte, V. Potential of commonly consumed green leafy vegetables for their antioxidant capacity and its linkage with the micronutrient profile. Int. J. Food Sci. Nutr. 2003, 54, 417–425.
  38. Harding, A.H.; Wareham, N.J.; Bingham, S.A.; Khaw, K.; Luben, R.; Welch, A.; Forouhi, N.G. Plasma vitamin C level, fruit and vegetable consumption, and the risk of new-onset type 2 diabetes mellitus: The European prospective investigation of cancer--Norfolk prospective study. Arch. Intern. Med. 2008, 168, 1493–1499.
  39. Larsson, S.C.; Wolk, A. Magnesium intake and risk of type 2 diabetes: A meta-analysis. J. Intern. Med. 2007, 262, 208–214.
  40. Liese, A.D.; Roach, A.K.; Sparks, K.C.; Marquart, L.; D’Agostino, R.B., Jr.; Mayer-Davis, E.J. Whole-grain intake and insulin sensitivity: The Insulin Resistance Atherosclerosis Study. Am. J. Clin. Nutr. 2003, 78, 965–971.
  41. Schwingshackl, L.; Hoffmann, G.; Kalle-Uhlmann, T.; Arregui, M.; Buijsse, L.; Boeing, H. Fruit and Vegetable Consumption and Changes in Anthropometric Variables in Adult Populations: A Systematic Review and Meta-Analysis of Prospective Cohort Studies. PLoS ONE 2015, 10, e0140846.
  42. Ramne, S.; Brunkwall, L.; Ericson, U.; Gray, N.; Kuhnle, G.G.C.; Nilsson, P.M.; Orho-Melander, M.; Sonestedt, E. Gut microbiota composition in relation to intake of added sugar, sugar-sweetened beverages and artificially sweetened beverages in the Malmö Offspring Study. Eur. J. Nutr. 2021, 60, 2087–2097.
  43. Hu, R.; Zeng, F.; Wu, L.; Wan, X.; Chen, Y.; Zhang, J.; Liu, B. Fermented carrot juice attenuates type 2 diabetes by mediating gut microbiota in rats. Food Funct. 2019, 10, 2935–2946.
  44. Nunes, S.; Viana, S.D.; Preguiça, I.; Alves, A.; Fernandes, R.; Teodoro, J.S.; Matos, P.; Figueirinha, A.; Salgueiro, L.; André, A.; et al. Blueberry Counteracts Prediabetes in a Hypercaloric Diet-Induced Rat Model and Rescues Hepatic Mitochondrial Bioenergetics. Nutrients 2021, 13, 4192.
  45. Fidélix, M.; Milenkovic, D.; Sivieri, K.; Cesar, T. Microbiota modulation and effects on metabolic biomarkers by orange juice: A controlled clinical trial. Food Funct. 2020, 11, 1599–1610.
  46. Kerimi, A.; Nyambe-Silavwe, H.; Gauer, J.S.; Tomás-Barberán, F.A.; Williamson, G. Pomegranate juice, but not an extract, confers a lower glycemic response on a high-glycemic index food: Randomized, crossover, controlled trials in healthy subjects. Am. J. Clin. Nutr. 2017, 106, 1384–1393.
  47. Aslam, H.; Marx, W.; Rocks, T.; Loughman, A.; Chandrasekaran, V.; Ruusunen, A.; Dawson, S.L.; West, M.; Mullarkey, E.; Pasco, J.A.; et al. The effects of dairy and dairy derivatives on the gut microbiota: A systematic literature review. Gut Microbes 2020, 12, 1799533.
  48. Pal, S.; Ellis, V.; Dhaliwal, S. Effects of whey protein isolate on body composition, lipids, insulin and glucose in overweight and obese individuals. Br. J. Nutr. 2010, 104, 716–723.
  49. Malik, V.S.; Sun, Q.; van Dam, R.M.; Rimm, E.B.; Willett, W.C.; Rosner, L.; Hu, F.B. Adolescent dairy product consumption and risk of type 2 diabetes in middle-aged women. Am. J. Clin. Nutr. 2011, 94, 854–861.
  50. Tong, X.; Dong, J.Y.; Wu, Z.W.; Li, W.; Qin, L.Q. Dairy consumption and risk of type 2 diabetes mellitus: A meta-analysis of cohort studies. Eur. J. Clin. Nutr. 2011, 65, 1027–1031.
  51. Choi, H.K.; Willett, W.C.; Stampfer, M.J.; Rimm, E.; Hu, F.B. Dairy consumption and risk of type 2 diabetes mellitus in men: A prospective study. Arch. Intern. Med. 2005, 165, 997–1003.
  52. Liu, S.; Choi, H.K.; Ford, E.; Song, Y.; Klevak, A.; Buring, J.E.; Manson, J.E. A prospective study of dairy intake and the risk of type 2 diabetes in women. Diabetes Care 2006, 29, 1579–1584.
  53. Link-Amster, H.; Rochat, F.; Saudan, K.Y.; Mignot, O.; Aeschlimann, J.M. Modulation of a specific humoral immune response and changes in intestinal flora mediated through fermented milk intake. FEMS Immunol. Med. Microbiol. 1994, 10, 55–63.
  54. Yılmaz, İ.; Dolar, M.E.; Özpınar, H. Effect of administering kefir on the changes in fecal microbiota and symptoms of inflammatory bowel disease: A randomized controlled trial. Turk. J. Gastroenterol. 2019, 30, 242–253.
  55. Butler, M.I.; Bastiaanssen, T.F.S.; Long-Smith, C.; Berding, K.; Morkl, S.; Cusack, A.M.; Strain, C.; Busca, K.; Porteous-Allen, P.; Claesson, M.J.; et al. Recipe for a Healthy Gut: Intake of Unpasteurised Milk Is Associated with Increased Lactobacillus Abundance in the Human Gut Microbiome. Nutrients 2020, 12, 1468.
  56. Wang, G.; Song, J.; Huang, Y.; Li, X.; Wang, H.; Zhang, Y.; Suo, H. Lactobacillus plantarum SHY130 isolated from yak yogurt attenuates hyperglycemia in C57BL/6J mice by regulating the enteroinsular axis. Food Funct. 2022, 13, 675–687.
  57. Gray, A.; Threlkeld, R.J. Nutritional Recommendations for Individuals with Diabetes. In Endotext; Feingold, K.R., Anawalt, L., Blackman, M.R., Boyce, A., Chrousos, G., Corpas, E., Eds.; MDText.com, Inc.: South Dartmouth, MA, USA, 2000.
  58. Gille, D.; Schmid, A. Vitamin B12 in meat and dairy products. Nutr. Rev. 2015, 73, 106–115.
  59. Fretts, A.M.; Follis, J.L.; Nettleton, J.A.; Lemaitre, R.N.; Ngwa, J.S.; Wojczynski, M.K.; Kalafati, I.P.; Varga, T.V.; Frazier-Wood, A.C.; Houston, D.K.; et al. Consumption of meat is associated with higher fasting glucose and insulin concentrations regardless of glucose and insulin genetic risk scores: A meta-analysis of 50,345 Caucasians. Am. J. Clin. Nutr. 2015, 102, 1266–1278.
  60. Rouhani, M.H.; Salehi-Abargouei, A.; Surkan, P.J.; Azadbakht, L. Is there a relationship between red or processed meat intake and obesity? A systematic review and meta-analysis of observational studies. Obes. Rev. 2014, 15, 740–748.
  61. Pan, A.; Sun, Q.; Bernstein, A.M.; Schulze, M.B.; Manson, J.E.; Willett, W.C.; Hu, F.B. Red meat consumption and risk of type 2 diabetes: 3 cohorts of US adults and an updated meta-analysis. Am. J. Clin. Nutr. 2011, 94, 1088–1096.
  62. de la Monte, S.M.; Tong, M.; Lawton, M.; Longato, L. Nitrosamine exposure exacerbates high fat diet-mediated type 2 diabetes mellitus, non-alcoholic steatohepatitis, and neurodegeneration with cognitive impairment. Mol. Neurodegener. 2009, 4, 54.
  63. Micha, R.; Michas, G.; Mozaffarian, D. Unprocessed red and processed meats and risk of coronary artery disease and type 2 diabetes—An updated review of the evidence. Curr. Atheroscler. Rep. 2012, 14, 515–524.
  64. Tong, M.; Neusner, A.; Longato, L.; Lawton, M.; Wands, J.R.; de la Monte, S.M. Nitrosamine exposure causes insulin resistance diseases: Relevance to type 2 diabetes mellitus, non-alcoholic steatohepatitis, and Alzheimer’s disease. J. Alzheimers Dis. 2009, 17, 827–844.
  65. Wolk, A. Potential health hazards of eating red meat. J. Intern. Med. 2017, 281, 106–122.
  66. White, D.L.; Collinson, A. Red meat, dietary heme iron, and risk of type 2 diabetes: The involvement of advanced lipoxidation endproducts. Adv. Nutr. 2013, 4, 403–411.
  67. Hofmann, S.M.; Dong, H.J.; Li, Z.; Cai, W.; Altomonte, J.; Thung, S.N.; Zeng, F.; Fisher, E.A.; Vlassara, H. Improved insulin sensitivity is associated with restricted intake of dietary glycoxidation products in the db/db mouse. Diabetes 2002, 51, 2082–2089.
  68. Sandri, M.; Dal Monego, S.; Conte, G.; Sgorlon, S.; Stefanon, B. Raw meat based diet influences faecal microbiome and end products of fermentation in healthy dogs. BMC Vet. Res. 2017, 13, 65.
  69. Albracht-Schulte, K.; Islam, T.; Johnson, P.; Moustaid-Moussa, N. Systematic Review of Beef Protein Effects on Gut Microbiota: Implications for Health. Adv. Nutr. 2021, 12, 102–114.
  70. Le Leu, R.K.; Winter, J.M.; Christophersen, C.T.; Young, G.P.; Humphreys, K.J.; Hu, Y.; Gratz, S.W.; Miller, R.B.; Topping, D.L.; Bird, A.R.; et al. Butyrylated starch intake can prevent red meat-induced O6-methyl-2-deoxyguanosine adducts in human rectal tissue: A randomised clinical trial. Br. J. Nutr. 2015, 114, 220–230.
  71. Li, J.; Li, Y.; Feng, S.; He, K.; Guo, L.; Chen, W.; Wang, M.; Zhong, L.; Wu, C.; Peng, X.; et al. Differential Effects of Dietary White Meat and Red Meat on NAFLD Progression by Modulating Gut Microbiota and Metabolites in Rats. Oxid. Med. Cell Longev. 2022, 2022, 6908934.
  72. Shi, J.; Zhao, D.; Zhao, F.; Wang, C.; Zamaratskaia, G.; Li, C. Chicken-eaters and pork-eaters have different gut microbiota and tryptophan metabolites. Sci. Rep. 2021, 11, 11934.
  73. Zhang, M.; Zou, X.; Zhao, D.; Zhao, F.; Li, C. Pork Meat Proteins Alter Gut Microbiota and Lipid Metabolism Genes in the Colon of Adaptive Immune-Deficient Mice. Mol. Nutr. Food Res. 2020, 64, e1901105.
  74. Zhu, Y.; Lin, X.; Li, H.; Li, Y.; Shi, X.; Zhao, F.; Xu, X.; Li, C.; Zhou, G. Intake of Meat Proteins Substantially Increased the Relative Abundance of Genus Lactobacillus in Rat Feces. PLoS ONE 2016, 11, e0152678.
  75. Zhu, Y.; Shi, X.; Lin, X.; Ye, K.; Xu, X.; Li, C.; Zhou, G. Beef, Chicken, and Soy Proteins in Diets Induce Different Gut Microbiota and Metabolites in Rats. Front. Microbiol. 2017, 8, 1395.
  76. Hou, Y.Y.; Ojo, O.; Wang, L.L.; Wang, Q.; Jiang, Q.; Shao, X.Y.; Wang, X.H. A Randomized Controlled Trial to Compare the Effect of Peanuts and Almonds on the Cardio-Metabolic and Inflammatory Parameters in Patients with Type 2 Diabetes Mellitus. Nutrients 2018, 10, 1565.
  77. Liu, Z.; Wang, W.; Huang, G.; Zhang, W.; Ni, L. In vitro and in vivo evaluation of the prebiotic effect of raw and roasted almonds (Prunus amygdalus). J. Sci. Food Agric. 2016, 96, 1836–1843.
  78. Holscher, H.D.; Guetterman, H.M.; Swanson, K.S.; An, R.; Matthan, N.R.; Lichtenstein, A.H.; Novotny, J.A.; Baer, D.J. Walnut Consumption Alters the Gastrointestinal Microbiota, Microbially Derived Secondary Bile Acids, and Health Markers in Healthy Adults: A Randomized Controlled Trial. J. Nutr. 2018, 148, 861–867.
  79. Ukhanova, M.; Wang, X.; Baer, D.J.; Novotny, J.A.; Fredborg, M.; Mai, V. Effects of almond and pistachio consumption on gut microbiota composition in a randomised cross-over human feeding study. Br. J. Nutr. 2014, 111, 2146–2152.
  80. Holscher, H.D.; Taylor, A.M.; Swanson, K.S.; Novotny, J.A.; Baer, D.J. Almond Consumption and Processing Affects the Composition of the Gastrointestinal Microbiota of Healthy Adult Men and Women: A Randomized Controlled Trial. Nutrients 2018, 10, 126.
  81. Peng, M.; Bitsko, E.; Biswas, D. Functional properties of peanut fractions on the growth of probiotics and foodborne bacterial pathogens. J. Food Sci. 2015, 80, M635–M641.
  82. Jiang, R.; Manson, J.E.; Stampfer, M.J.; Liu, S.; Willett, W.C.; Hu, F.B. Nut and peanut butter consumption and risk of type 2 diabetes in women. JAMA 2002, 288, 2554–2560.
  83. Pan, X.; Siloto, R.M.; Wickramarathna, A.D.; Mietkiewska, E.; Weselake, R.J. Identification of a pair of phospholipid:diacylglycerol acyltransferases from developing flax (Linum usitatissimum L.) seed catalyzing the selective production of trilinolenin. J. Biol. Chem. 2013, 288, 24173–24188.
  84. Viguiliouk, E.; Kendall, C.W.; Blanco Mejia, S.; Cozma, A.I.; Ha, V.; Mirrahimi, A.; Jayalath, V.H.; Augustin, L.S.; Chiavaroli, L.; Leiter, L.A.; et al. Effect of tree nuts on glycemic control in diabetes: A systematic review and meta-analysis of randomized controlled dietary trials. PLoS ONE 2014, 9, e103376.
  85. Zhu, L.; Sha, L.; Li, K.; Wang, Z.; Wang, T.; Li, Y.; Liu, P.; Dong, X.; Dong, Y.; Zhang, X.; et al. Dietary flaxseed oil rich in omega-3 suppresses severity of type 2 diabetes mellitus via anti-inflammation and modulating gut microbiota in rats. Lipids Health Dis. 2020, 19, 20.
  86. Lee, J.Y.; Kim, M.; Oh, S.B.; Kim, H.Y.; Kim, C.; Kim, T.Y.; Park, Y.H. Superoxide dismutase 3 prevents early stage diabetic retinopathy in streptozotocin-induced diabetic rat model. PLoS ONE 2022, 17, e0262396.
  87. Schwingshackl, L.; Christoph, M.; Hoffmann, G. Effects of Olive Oil on Markers of Inflammation and Endothelial Function-A Systematic Review and Meta-Analysis. Nutrients 2015, 7, 7651–7675.
  88. Sánchez-Quintero, M.J.; Delgado, J.; Medina-Vera, D.; Becerra-Muñoz, V.M.; Queipo-Ortuño, M.I.; Estévez, M.; Plaza-Andrades, I.; Rodríguez-Capitán, J.; Sánchez, P.L.; Crespo-Leiro, M.G.; et al. Beneficial Effects of Essential Oils from the Mediterranean Diet on Gut Microbiota and Their Metabolites in Ischemic Heart Disease and Type-2 Diabetes Mellitus. Nutrients 2022, 14, 4650.
More
This entry is offline, you can click here to edit this entry!
Video Production Service