Modulation of gut microbiota dysbioses in type 2 diabetic patients by macrobiotic Ma-Pi 2 diet

Marco Candela, Elena Biagi, Matteo Soverini, Clarissa Consolandi, Sara Quercia, Marco Severgnini, Clelia Peano, Silvia Turroni, Simone Rampelli, Paolo Pozzilli, Mario Pianesi, Francesco Fallucca, Patrizia Brigidi, Marco Candela, Elena Biagi, Matteo Soverini, Clarissa Consolandi, Sara Quercia, Marco Severgnini, Clelia Peano, Silvia Turroni, Simone Rampelli, Paolo Pozzilli, Mario Pianesi, Francesco Fallucca, Patrizia Brigidi

Abstract

The gut microbiota exerts a role in type 2 diabetes (T2D), and deviations from a mutualistic ecosystem layout are considered a key environmental factor contributing to the disease. Thus, the possibility of improving metabolic control in T2D by correcting gut microbiome dysbioses through diet has been evaluated. Here, we explore the potential of two different energy-restricted dietary approaches - the fibre-rich macrobiotic Ma-Pi 2 diet or a control diet recommended by Italian professional societies for T2D treatment - to correct gut microbiota dysbioses in T2D patients. In a previous 21-d open-label MADIAB trial, fifty-six overweight T2D patients were randomised to the Ma-Pi 2 or the control diet. For the present study, stools were collected before and after intervention from a subset of forty MADIAB participants, allowing us to characterise the gut microbiota by 16S rRNA sequencing and imputed metagenomics. To highlight microbiota dysbioses in T2D, the gut microbiota of thirteen normal-weight healthy controls were characterised. According to our findings, both diets were effective in modulating gut microbiome dysbioses in T2D, resulting in an increase of the ecosystem diversity and supporting the recovery of a balanced community of health-promoting SCFA producers, such as Faecalibacterium, Roseburia, Lachnospira, Bacteroides and Akkermansia. The Ma-Pi 2 diet, but not the control diet, was also effective in counteracting the increase of possible pro-inflammatory groups, such as Collinsella and Streptococcus, in the gut ecosystem, showing the potential to reverse pro-inflammatory dysbioses in T2D, and possibly explaining the greater efficacy in improving the metabolic control.

Keywords: CTR control diet; Dysbiosis; FBG fasting blood glucose; Fibre-rich diets; GM gut microbiota; Gut microbiota; KO KEGG Orthology; Macrobiotic diets; PCoA principal coordinates analysis; T2D type 2 diabetes; Type 2 diabetes.

Figures

Fig. 1
Fig. 1
Comparison of the gut microbiota compositional structure between overweight type 2 diabetes (T2D) patients at baseline and healthy controls. (a) Principal coordinates analysis (PCoA) based on weighted UniFrac distances shows separation between forty overweight T2D patients at T0 and thirteen normal-weight healthy controls. , Healthy controls; , T2D patients. P<0·001; permutation test with pseudoF ratios. (b) Superimposition of microbial genera on the PCoA plot in order to identify the genera involved in this separation. Sphere width is proportional to the mean relative abundance of the genus across all samples. The two components explain 37·8 and 23·9 % of the variance, respectively. , , Centroids for each group with indication of standard errors on each coordinate axis; uncl, unclassified.
Fig. 2
Fig. 2
Variation of the weighted UniFrac PC2 coordinates between the study groups. , Ma-Pi 2 diet group at T0; , control (CTR) diet group at T0; , healthy controls; , Ma-Pi 2 diet group at T1; , CTR diet group at T1. For each group, average (±sem, error bar) PC2 coordinates are shown. The significance of the differences between the PC2 coordinates of the groups is indicated as follows: *P<0·001 (Ma-Pi 2 diet group at T0 v. T1), ** P=0·01 (CTR diet group at T0 v. T1), ***P=0·01 (Ma-Pi 2 diet group at T0 v. healthy controls), **** P=0·03 (CTR diet group at T0 v.healthy controls); Wilcoxon’s signed rank-sum test.
Fig. 3
Fig. 3
Comparison of the gut microbiota compositional structure of type 2 diabetes (T2D) patients before and after the nutritional interventions. (a) Principal coordinates analysis (PCoA) based on weighted UniFrac distances for T2D subjects following the Ma-Pi 2 diet (n 21) shows separation between T0 () and T1 (). The two components explain 35·0 and 24·8 % of the variance, respectively. P=0·01; permutation test with pseudo F ratios. (b) PCoA based on weighted UniFrac distances for T2D subjects following the control (CTR) diet (n 19). , T0; , T1. The two components explain 32·0 and 20·2 % of the variance, respectively.P=0·04; permutation test with pseudo F ratios. Lines connect T0 and T1 samples from the same patient. , responding bacterial genera and biochemical parameters, respectively; , direction of significant correlations; , , , , centroids for each time point.
Fig. 4
Fig. 4
Impact of dietary interventions on the taxonomic structure of the core microbiota in type 2 diabetes (T2D) patients. Heat maps were calculated for both Ma-Pi 2 and control (CTR) diet, based on the log-ratio of the median genus abundance in T2D patients (T0 and T1 samples) and healthy controls. The colour code and segment line reveal the deviation in terms of fold change from the median profile of healthy subjects (). T2D patients following the Ma-Pi 2 diet (n 21); T2D patients following the CTR diet (n 19); healthy controls (n 13).
Fig. 5
Fig. 5
Impact of dietary components on the gut microbiota taxonomic structure. Cladogram obtained with the GraPhlAn tool, showing the family-level gut microbiota profile of T1 samples from both intervention groups with a phylum-based colour code (, Firmicutes; , Bacteroidetes; , Proteobacteria; , Actinobacteria; , Verrucomicrobia; , Synergistetes). Families with relative abundance of at least 0·5 % in at least two samples are plotted. Larger circles identify bacterial families having a positive correlation with at least one dietary component; the names of these families are reported. Filled circles identify bacterial families that showed significantly higher abundance in T1 samples of the Ma-Pi 2 diet group () or the CTR diet group (). Bacterial family-food component correlations are indicated by filled boxes in the external rings of the plot, referring to the list of dietary components. T2D patients following the Ma-Pi 2 diet (n 21); T2D patients following the CTR diet (n19).
Fig. 6
Fig. 6
Functional dysbioses of the gut microbiome in type 2 diabetes (T2D) patients. Metabolic pathways were superimposed on the principal component analysis plot based on Euclidean distances, and the pathways responsible for the separation are shown. , Healthy controls (n13); , T2D patients (n 40). Sphere width is proportional to the mean relative abundance of the function across all samples. The two components explain 14·0 and 9·2 % of the variance, respectively. P=0·03; permutation test with pseudoF ratios. , , Centroids for each group with indication of standard errors on each coordinate axis.
Fig. 7
Fig. 7
Impact of Ma-Pi 2 dietary intervention on the functional configuration of the gut microbiome in type 2 diabetes (T2D) patients. Metabolic pathways were superimposed on the principal component analysis plot based on Euclidean distances in T2D patients before (T0, ) and after (T1, ) the Ma-Pi 2 diet (n 21). The two components explain 22·3 and 14·9 % of the variance, respectively. P=0·007; permutation test with pseudo F ratios. , , Centroids for each group with indication of standard errors on each coordinate axis.

References

    1. Xu Y (2013) Prevalence and control of diabetes in Chinese adults. JAMA 310, 948.
    1. Zhang P, Zhang X, Brown J, et al. (2010) Global healthcare expenditure on diabetes for 2010 and 2030. Diabetes Res Clin Pract 87, 293–301.
    1. Zhang X, Shen D, Fang Z, et al. (2013) Human gut microbiota changes reveal the progression of glucose intolerance. PLOS ONE 8, e71108.
    1. Xu Z, Malmer D, Langille MGI, et al. (2014) Which is more important for classifying microbial communities: who’s there or what they can do? ISME J 8, 2357–2359.
    1. Qin J, Li R, Raes J, et al. (2010) A human gut microbial gene catalogue established by metagenomic sequencing. Nature 464, 59–65.
    1. Karlsson FH, Tremaroli V, Nookaew I, et al. (2013) Gut metagenome in European women with normal, impaired and diabetic glucose control. Nature 498, 99–103.
    1. Larsen N, Vogensen FK, van den Berg FWJ, et al. (2010) Gut microbiota in human adults with type 2 diabetes differs from non-diabetic adults. PLoS ONE 5, e9085.
    1. Tremaroli V & Bäckhed F (2012) Functional interactions between the gut microbiota and host metabolism. Nature 489, 242–249.
    1. De Vadder F, Kovatcheva-Datchary P, Goncalves D, et al. (2014) Microbiota-generated metabolites promote metabolic benefits via gut-brain neural circuits. Cell 156, 84–96.
    1. Backhed F, Ding H, Wang T, et al. (2004) The gut microbiota as an environmental factor that regulates fat storage. Proc Natl Acad Sci U S A 101, 15718–15723.
    1. Tilg H & Moschen AR (2014) Microbiota and diabetes: an evolving relationship. Gut 63, 1513–1521.
    1. Khan MT, Nieuwdorp M & Bäckhed F (2014) Microbial modulation of insulin sensitivity. Cell Metab 20, 753–760.
    1. Amar J, Chabo C, Waget A, et al. (2011) Intestinal mucosal adherence and translocation of commensal bacteria at the early onset of type 2 diabetes: molecular mechanisms and probiotic treatment. EMBO Mol Med 3, 559–572.
    1. Burcelin R (2012) Regulation of metabolism: a cross talk between gut microbiota and its human host. Physiology (Bethesda) 27, 300–307.
    1. Cani PD, Amar J, Iglesias MA, et al. (2007) Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 56, 1761–1772.
    1. Everard A & Cani PD (2013) Diabetes, obesity and gut microbiota. Best Pract Res Clin Gastroenterol 27, 73–83.
    1. Karlsson F, Tremaroli V, Nielsen J, et al. (2013) Assessing the human gut microbiota in metabolic diseases. Diabetes 62, 3341–3349.
    1. Turnbaugh PJ, Hamady M, Yatsunenko T, et al. (2009) A core gut microbiome in obese and lean twins. Nature 457, 480–484.
    1. David LA, Maurice CF, Carmody RN, et al. (2013) Diet rapidly and reproducibly alters the human gut microbiome. Nature 505, 559–563.
    1. He C, Shan Y & Song W (2015) Targeting gut microbiota as a possible therapy for diabetes. Nutr Res 35, 361–367.
    1. Soare A, Khazrai YM, Del Toro R, et al. (2014) The effect of the macrobiotic Ma-Pi 2 diet vs. the recommended diet in the management of type 2 diabetes: the randomized controlled MADIAB trial. Nutr Metab 11, 39.
    1. Craig WJ & Mangels AR, & American Dietetic Association (2009) Position of the American Dietetic Association: vegetarian diets. J Am Diet Assoc 9, 1266–1282.
    1. Centanni M, Turroni S, Consolandi C, et al. (2013) The enterocyte-associated intestinal microbiota of breast-fed infants and adults responds differently to a TNF-α-mediated pro-inflammatory stimulus. PLOS ONE 8, e81762.
    1. Klindworth A, Pruesse E, Schweer T, et al. (2013) Evaluation of general 16S ribosomal RNA gene PCR primers for classical and next-generation sequencing-based diversity studies. Nucleic Acids Res 41, e1.
    1. Masella AP, Bartram AK, Truszkowski JM, et al. (2012) PANDAseq: paired-end assembler for illumina sequences. BMC Bioinformatics 3, 31.
    1. Caporaso JG, Kuczynski J, Stombaugh J, et al. (2010) QIIME allows analysis of high-throughput community sequencing data. Nat Methods 7, 335–336.
    1. Edgar RC (2010) Search and clustering orders of magnitude faster than BLAST. Bioinformatics 26, 2460–2461.
    1. Langille MGI, Zaneveld J, Caporaso JG, et al. (2013) Predictive functional profiling of microbial communities using 16S rRNA marker gene sequences. Nat Biotechnol 31, 814–821.
    1. Kanehisa M, Goto S, Sato Y, et al. (2012) KEGG for integration and interpretation of large-scale molecular data sets. Nucleic Acids Res 40, D109–D114.
    1. Asnicar F, Weingart G, Tickle TL, et al. (2015) Compact graphical representation of phylogenetic data and metadata with GraPhlAn. PeerJ 3, e1029.
    1. Lozupone CA, Stombaugh J, Gonzalez A, et al. (2013) Meta-analyses of studies of the human microbiota. Genome Res 23, 1704–1714.
    1. Marchesi JR, Dutilh BE, Hall N, et al. (2011) Towards the human colorectal cancer microbiome. PLoS ONE 6, e20447.
    1. Morgan XC, Tickle TL, Sokol H, et al. (2012) Dysfunction of the intestinal microbiome in inflammatory bowel disease and treatment. Genome Biol 13, R79.
    1. Thams P & Capito K (1999) L-arginine stimulation of glucose-induced insulin secretion through membrane depolarization and independent of nitric oxide. Eur J Endocrinol 140, 87–93.
    1. Holmes E, Loo RL, Stamler J, et al. (2008) Human metabolic phenotype diversity and its association with diet and blood pressure. Nature 453, 396–400.
    1. Nicholson JK, Holmes E, Kinross J, et al. (2012) Host-gut microbiota metabolic interactions. Science 336, 1262–1267.
    1. Sorrell TC, Muller M & Sztelma K (1992) Bacterial metabolism of human polymorphonuclear leukocyte-derived arachidonic acid. Infect Immun 60, 1779–1785.
    1. Ridley CP, Lee HY & Khosla C (2008) Evolution of polyketide synthases in bacteria. Proc Natl Acad Sci U S A 105, 4595–4600.
    1. Faber F & Bäumler AJ (2014) The impact of intestinal inflammation on the nutritional environment of the gut microbiota. Immunol Lett 162, 48–53.
    1. Wieland Brown LC, Penaranda C, Kashyap PC, et al. (2013) Production of α-galactosylceramide by a prominent member of the human gut microbiota. PLoS Biol 11, e1001610.
    1. Biagi E, Nylund L, Candela M, et al. (2010) Through ageing, and beyond: gut microbiota and inflammatory status in seniors and centenarians. PLoS ONE 5, e10667.
    1. Tong M, Li X, Wegener Parfrey L, et al. (2013) A modular organization of the human intestinal mucosal microbiota and its association with inflammatory bowel disease. PLOS ONE 8, e80702.
    1. Joossens M, Huys G, Cnockaert M, et al. (2011) Dysbiosis of the faecal microbiota in patients with Crohn’s disease and their unaffected relatives. Gut 5, 631–637.
    1. Karlsson FH, Fåk F, Nookaew I, et al. (2012) Symptomatic atherosclerosis is associated with an altered gut metagenome. Nat Commun 3, 1245.
    1. Jiang W, Wu N, Wang X, et al. (2015) Dysbiosis gut microbiota associated with inflammation and impaired mucosal immune function in intestine of humans with non-alcoholic fatty liver disease. Sci Rep 5, 8096.
    1. Kamada N, Seo SU, Chen GY, et al. (2013) Role of the gut microbiota in immunity and inflammatory disease. Nat Rev Immunol 13, 321–335.
    1. Qin J, Li Y, Cai Z, et al. (2012) A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature 490, 55–60.
    1. Flint HJ, Duncan SH, Scott KP, et al. (2015) Links between diet, gut microbiota composition and gut metabolism. Proc Nutr Soc 74, 13–22.
    1. Russell WR, Hoyles L, Flint HJ, et al. (2013) Colonic bacterial metabolites and human health. Curr Opin Microbiol 16, 246–254.
    1. Trompette A, Gollwitzer ES, Yadava K, et al. (2014) Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nat Med 20, 159–166.
    1. Segain JP, Raingeard de la Blétière D, Bourreille A, et al. (2000) Butyrate inhibits inflammatory responses through NFkappaB inhibition: implications for Crohn’s disease. Gut 47, 397–403.
    1. Johnson AMF & Olefsky JM (2013) The origins and drivers of insulin resistance. Cell 152, 673–684.
    1. Sonnenburg ED & Sonnenburg JL (2014) Starving our microbial self: the deleterious consequences of a diet deficient in microbiota-accessible carbohydrates. Cell Metab 20, 779–786.
    1. Everard A, Belzer C, Geurts L, et al. (2013) Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc Natl Acad Sci U S A 110, 9066–9071.
    1. Xu J, Lian F, Zhao L, et al. (2015) Structural modulation of gut microbiota during alleviation of type 2 diabetes with a Chinese herbal formula. ISME J 9, 552–562.
    1. Brahe LK, Le Chatelier E, Prifti E, et al. (2015) Specific gut microbiota features and metabolic markers in postmenopausal women with obesity. Nutr Diabetes 5, e159.
    1. Donath MY & Shoelson SE (2011) Type 2 diabetes as an inflammatory disease. Nat Rev Immunol 11, 98–107.
    1. Schnorr SL, Candela M, Rampelli S, et al. (2014) Gut microbiome of the Hadza hunter-gatherers. Nat Commun 5, 3654.

Source: PubMed

3
Sottoscrivi