Akkermansia muciniphila inversely correlates with the onset of inflammation, altered adipose tissue metabolism and metabolic disorders during obesity in mice

Marc Schneeberger, Amandine Everard, Alicia G Gómez-Valadés, Sébastien Matamoros, Sara Ramírez, Nathalie M Delzenne, Ramon Gomis, Marc Claret, Patrice D Cani, Marc Schneeberger, Amandine Everard, Alicia G Gómez-Valadés, Sébastien Matamoros, Sara Ramírez, Nathalie M Delzenne, Ramon Gomis, Marc Claret, Patrice D Cani

Abstract

Recent evidence indicates that the gut microbiota plays a key role in the pathophysiology of obesity. Indeed, diet-induced obesity (DIO) has been associated to substantial changes in gut microbiota composition in rodent models. In the context of obesity, enhanced adiposity is accompanied by low-grade inflammation of this tissue but the exact link with gut microbial community remains unknown. In this report, we studied the consequences of high-fat diet (HFD) administration on metabolic parameters and gut microbiota composition over different periods of time. We found that Akkermansia muciniphila abundance was strongly and negatively affected by age and HFD feeding and to a lower extend Bilophila wadsworthia was the only taxa following an opposite trend. Different approaches, including multifactorial analysis, showed that these changes in Akkermansia muciniphila were robustly correlated with the expression of lipid metabolism and inflammation markers in adipose tissue, as well as several circulating parameters (i.e., glucose, insulin, triglycerides, leptin) from DIO mice. Thus, our data shows the existence of a link between gut Akkermansia muciniphila abundance and adipose tissue homeostasis on the onset of obesity, thus reinforcing the beneficial role of this bacterium on metabolism.

Figures

Figure 1. Kinetic evolution of body weight…
Figure 1. Kinetic evolution of body weight and adiposity following chronic HFD administration.
(A) Body weight gain (g) and (B) adiposity (% of eWAT on total body weight) measured after 3 weeks (3w); 6 weeks (6w); 12 weeks (12w) and 16 weeks (16w) of a high-fat diet (HFD) or a control diet (CT) (n = 6/group). Data are presented as the mean ± SEM. Data are significantly different (P < 0.05) according to the unpaired two-tailed Student t-test. *indicates a significant difference versus CT (P < 0.05).
Figure 2. Time series evolution of markers…
Figure 2. Time series evolution of markers of inflammation and macrophage infiltration in white adipose tissue following HFD administration.
mRNA expression of (A) Tnf (encoding TNF-α), (B) Ccl2 (encoding MCP-1), (C) Itgax encoding CD11c), (D) Emr1 (encoding F4/80), (E) Lbp (encoding LBP), (F) Il6 (encoding Il-6) and (G) Il1b (encoding IL-1β) measured in the adipose tissue after 3 weeks (3w); 6 weeks (6w); 12 weeks (12w) and 16 weeks (16w) of a high-fat diet (HFD) or a control diet (CT) (n = 6/group). Data are presented as the mean ± SEM. Data are significantly different (P < 0.05) according to the unpaired two-tailed Student t-test. *indicates a significant difference versus CT (P < 0.05).
Figure 3. Time series evolution of markers…
Figure 3. Time series evolution of markers of fatty acid oxidation and browning in white adipose tissue following HFD treatment.
mRNA expression of (A) Pppargc1a (encoding PGC1-α), (B) Pppargc1b (encoding PGC1-β), (C) Cpt1a (encoding CPT-1a), (D) Acacb (encoding ACC2), (E) Cidea (encoding CIDEA), (F) Elovl3 (encoding ELOVL3), (G) Prdm16 (encoding PRDM16) and (H) Acox1 (encoding ACOX1) measured in the adipose tissue after 3 weeks (3w); 6 weeks (6w); 12 weeks (12w) and 16 weeks (16w) of a high-fat diet (HFD) or a control diet (CT) (n = 6/group). Data are presented as the mean ± SEM. Data are significantly different (P < 0.05) according to the unpaired two-tailed Student t-test. *indicates a significant difference versus CT (P < 0.05).
Figure 4. Time series evolution of markers…
Figure 4. Time series evolution of markers of lipogenesis and adipogenesis in white adipose tissue following HFD treatment.
mRNA expression of (A) Dgat2 (encoding DGAT2), (B) Acaca (encoding ACC1), (C) Fasn (encoding FAS), (D) Pparg (encoding PPAR-γ) and (E) Cebpa (encoding CEBP-α) measured in the adipose tissue after 3 weeks (3w); 6 weeks (6w); 12 weeks (12w) and 16 weeks (16w) of a high-fat diet (HFD) or a control diet (CT) (n = 6/group). Data are presented as the mean ± SEM. Data are significantly different (P < 0.05) according to the unpaired two-tailed Student t-test. *indicates a significant difference versus CT (P < 0.05).
Figure 5. Time series evolution of orexigenic…
Figure 5. Time series evolution of orexigenic and anorexigenic markers in the hypothalamus and circulating levels of metabolic parameters following HFD treatment.
mRNA expression of (A) AgRP (encoding AGRP), (B) Npy (encoding NPY), (C) Pomc (encoding POMC) measured in the hypothalamus,. Circulating levels of (D) glucose, (E) triglycerides, (F) leptin and (G) insulin measured in the serum of mice after 3 weeks (3w); 6 weeks (6w); 12 weeks (12w) and 16 weeks (16w) of a high-fat diet (HFD) or a control diet (CT) (n = 6/group). Data are presented as the mean ± SEM. Data are significantly different (P < 0.05) according to the unpaired two-tailed Student t-test. *indicates a significant difference versus CT (P < 0.05).
Figure 6. Time series evolution of specific…
Figure 6. Time series evolution of specific gut bacteria following HFD treatment.
Quantification of (A) Akkermansia muciniphila, (B) Bifidobacterium spp., (C) Roseburia spp. (D) Lactobacillus spp., (E) Bacteroides/Prevotella spp, (F) Bilophila wadsworthia and (G) total gut bacteria abundance after 3 weeks (3w); 6 weeks (6w); 12 weeks (12w) and 16 weeks (16w) of a high-fat diet (HFD) or a control diet (CT) and expressed as Log cells per g of caecal content (n = 6/group). Data are presented as the mean ± SEM. Data are significantly different (P < 0.05) according to the unpaired two-tailed Student t-test. *indicates a significant difference versus CT (P < 0.05).
Figure 7. Individuals factor map.
Figure 7. Individuals factor map.
The individual factor map presents the repartition of the samples (dots) in the multifactorial analysis’ plane. Time and diet (squares) are presented as illustrative (inactive) qualitative factors. Samples are colored according to treatment’s duration. Circles regroup all samples from a specific diet for a single time point, except week 3 where all samples are grouped together.
Figure 8. Hierarchical clustering.
Figure 8. Hierarchical clustering.
Hierarchical clustering of the samples on the principal components of the multifactorial analysis. This graph illustrates the similarities and the clustering between different samples. The smaller the distance linking samples is (resulting from the addition of horizontal distances in dark line), the more similar these samples are for the parameters measured.
Figure 9. Correlation circle and heat map…
Figure 9. Correlation circle and heat map correlation showing associations between bacterial taxa and adipose tissue metabolic parameters.
Multifactorial analysis (A) correlation map and (B) heat map of the Spearman r correlations between the bacterial genera and the metabolic parameters measured in the adipose tissue of mice after 3 weeks (3w); 6 weeks (6w); 12 weeks (12w) and 16 weeks (16w) of a high-fat diet (HFD) or a CT diet (CT) (n = 6/group). Data are presented as the mean ± SEM. Squared cells depict significance following the Spearman correlation and FDR correction for multiple comparisons *P < 0.05.

References

    1. Everard A. et al. Microbiome of prebiotic-treated mice reveals novel targets involved in host response during obesity. The ISME journal 8, 2116–2130, doi: 10.1038/ismej.2014.45 (2014).
    1. Everard A., Matamoros S., Geurts L., Delzenne N. M. & Cani P. D. Saccharomyces boulardii Administration Changes Gut Microbiota and Reduces Hepatic Steatosis, Low-Grade Inflammation, and Fat Mass in Obese and Type 2 Diabetic db/db Mice. mBio 5, doi: 10.1128/mBio.01011-14 (2014).
    1. Everard A. et al. Intestinal epithelial MyD88 is a sensor switching host metabolism towards obesity according to nutritional status. Nature communications 5, 5648, doi: 10.1038/ncomms6648 (2014).
    1. Cotillard A. et al. Dietary intervention impact on gut microbial gene richness. Nature 500, 585–588, doi: 10.1038/nature12480 (2013).
    1. Le Chatelier E. et al. Richness of human gut microbiome correlates with metabolic markers. Nature 500, 541–546, doi: 10.1038/nature12506 (2013).
    1. Tremaroli V. & Backhed F. Functional interactions between the gut microbiota and host metabolism. Nature 489, 242–249, doi: 10.1038/nature11552 (2012).
    1. David L. A. et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 505, 559–563, doi: 10.1038/nature12820 (2014).
    1. Cani P. D. et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 56, 1761–1772 (2007).
    1. Cani P. D. et al. Selective increases of bifidobacteria in gut microflora improve high-fat-diet-induced diabetes in mice through a mechanism associated with endotoxaemia. Diabetologia 50, 2374–2383 (2007).
    1. Cani P. D. et al. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes 57, 1470–1481 (2008).
    1. Turnbaugh P. J., Backhed F., Fulton L. & Gordon J. I. Diet-induced obesity is linked to marked but reversible alterations in the mouse distal gut microbiome. Cell Host.Microbe 3, 213–223 (2008).
    1. Hildebrandt M. A. et al. High-fat diet determines the composition of the murine gut microbiome independently of obesity. Gastroenterology 137, 1716–1724 (2009).
    1. Serino M. et al. Metabolic adaptation to a high-fat diet is associated with a change in the gut microbiota. Gut 61, 543–553, doi: 10.1136/gutjnl-2011-301012 (2012).
    1. Everard A. et al. Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc Natl Acad Sci USA 110, 9066–9071, doi: 10.1073/pnas.1219451110 (2013).
    1. Wang J. H., Bose S., Kim H. G., Han K. S. & Kim H. Fermented Rhizoma Atractylodis Macrocephalae alleviates high fat diet-induced obesity in association with regulation of intestinal permeability and microbiota in rats. Scientific reports 5, 8391, doi: 10.1038/srep08391 (2015).
    1. Dewulf E. M. et al. Inulin-type fructans with prebiotic properties counteract GPR43 overexpression and PPARgamma-related adipogenesis in the white adipose tissue of high-fat diet-fed mice. J. Nutr. Biochem. 22, 712–722 (2011).
    1. Neyrinck A. M. et al. Curcuma longa Extract Associated with White Pepper Lessens High Fat Diet-Induced Inflammation in Subcutaneous Adipose Tissue. PLoS One 8, 0081252 (2013).
    1. Neyrinck A. M. et al. Polyphenol-rich extract of pomegranate peel alleviates tissue inflammation and hypercholesterolaemia in high-fat diet-induced obese mice: potential implication of the gut microbiota. The British journal of nutrition 109, 802–809, doi: 10.1017/S0007114512002206 (2013).
    1. Devkota S. et al. Dietary-fat-induced taurocholic acid promotes pathobiont expansion and colitis in Il10−/− mice. Nature 487, 104–108, doi: 10.1038/nature11225 (2012).
    1. Qiu Y. et al. Eosinophils and type 2 cytokine signaling in macrophages orchestrate development of functional beige fat. Cell 157, 1292–1308, doi: 10.1016/j.cell.2014.03.066 (2014).
    1. Rao R. R. et al. Meteorin-like Is a Hormone that Regulates Immune-Adipose Interactions to Increase Beige Fat Thermogenesis. Cell 157, 1279–1291, doi: 10.1016/j.cell.2014.03.065 (2014).
    1. Muccioli G. G. et al. The endocannabinoid system links gut microbiota to adipogenesis. Mol. Syst. Biol. 6, 392 (2010).
    1. Geurts L. et al. Adipose tissue NAPE-PLD controls fat mass development by altering the browning process and gut microbiota. Nature communications 6, 6495, doi: 10.1038/ncomms7495 (2015).
    1. Schele E. et al. The gut microbiota reduces leptin sensitivity and the expression of the obesity-suppressing neuropeptides proglucagon (Gcg) and brain-derived neurotrophic factor (Bdnf) in the central nervous system. Endocrinology 154, 3643–3651, doi: 10.1210/en.2012-2151 (2013).
    1. Wueest S., Yang X., Liu J., Schoenle E. J. & Konrad D. Inverse regulation of basal lipolysis in perigonadal and mesenteric fat depots in mice. American journal of physiology. Endocrinology and metabolism 302, E153–160, doi: 10.1152/ajpendo.00338.2011 (2012).
    1. Stanton M. C. et al. Inflammatory Signals shift from adipose to liver during high fat feeding and influence the development of steatohepatitis in mice. J Inflamm (Lond) 8, 8, doi: 10.1186/1476-9255-8-8 (2011).
    1. Anhe F. F. et al. A polyphenol-rich cranberry extract protects from diet-induced obesity, insulin resistance and intestinal inflammation in association with increased Akkermansia spp. population in the gut microbiota of mice. Gut 64, 872–883, doi: 10.1136/gutjnl-2014-307142 (2015).
    1. Shin N. R. et al. An increase in the Akkermansia spp. population induced by metformin treatment improves glucose homeostasis in diet-induced obese mice. Gut 63, 727–735, doi: 10.1136/gutjnl-2012-303839 (2014).
    1. Kim H. et al. Modulation of the intestinal microbiota is associated with lower plasma cholesterol and weight gain in hamsters fed chardonnay grape seed flour. J Agric Food Chem 63, 1460–1467, doi: 10.1021/jf5026373 (2015).
    1. Dewulf E. M. et al. Insight into the prebiotic concept: lessons from an exploratory, double blind intervention study with inulin-type fructans in obese women. Gut, doi: 10.1136/gutjnl-2012-303304 (2012).
    1. Cani P. D. & Van Hul M. Novel opportunities for next-generation probiotics targeting metabolic syndrome. Curr Opin Biotechnol 32, 21–27, doi: 10.1016/j.copbio.2014.10.006 (2015).
    1. Parks B. W. et al. Genetic control of obesity and gut microbiota composition in response to high-fat, high-sucrose diet in mice. Cell Metab 17, 141–152, doi: 10.1016/j.cmet.2012.12.007 (2013).
    1. Carmody R. N. et al. Diet dominates host genotype in shaping the murine gut microbiota. Cell host & microbe 17, 72- 84, doi: 10.1016/j.chom.2014.11.010 (2015).
    1. Hamilton M. K., Boudry G., Lemay D. G. & Raybould H. E. Changes in intestinal barrier function and gut microbiota in high-fat diet-fed rats are dynamic and region dependent. American journal of physiology. Gastrointestinal and liver physiology 308, G840–851, doi: 10.1152/ajpgi.00029.2015 (2015).
    1. Caesar R., Tremaroli V., Kovatcheva-Datchary P., Cani P. D. & Backhed F. Crosstalk between Gut Microbiota and Dietary Lipids Aggravates WAT Inflammation through TLR Signaling. Cell Metab, doi: 10.1016/j.cmet.2015.07.026 (2015).
    1. Dao M. C. et al. Akkermansia muciniphila and improved metabolic health during a dietary intervention in obesity: relationship with gut microbiome richness and ecology. Gut, doi: 10.1136/gutjnl-2014-308778 (2015).
    1. Everard A. et al. Responses of gut microbiota and glucose and lipid metabolism to prebiotics in genetic obese and diet-induced leptin-resistant mice. Diabetes 60, 2775–2786 (2011).
    1. 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 73, 7767–7770, doi: 10.1128/AEM.01477-07 (2007).
    1. Sonoyama K. et al. Response of gut microbiota to fasting and hibernation in Syrian hamsters. Appl Environ Microbiol 75, 6451–6456, doi: 10.1128/AEM.00692-09 (2009).
    1. Remely M. et al. Increased gut microbiota diversity and abundance of Faecalibacterium prausnitzii and Akkermansia after fasting: a pilot study. Wiener klinische Wochenschrift, doi: 10.1007/s00508-015-0755-1 (2015).
    1. Briggs D. I., Enriori P. J., Lemus M. B., Cowley M. A. & Andrews Z. B. Diet-induced obesity causes ghrelin resistance in arcuate NPY/AgRP neurons. Endocrinology 151, 4745–4755, doi: 10.1210/en.2010-0556 (2010).
    1. Claret M. et al. AMPK is essential for energy homeostasis regulation and glucose sensing by POMC and AgRP neurons. J.Clin.Invest 117, 2325–2336 (2007).
    1. Dewulf E. M. et al. Insight into the prebiotic concept: lessons from an exploratory, double blind intervention study with inulin-type fructans in obese women. Gut 62, 1112–1121, doi: 10.1136/gutjnl-2012-303304 (2013).
    1. Benjamini Y. & Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. Journal of the Royal Statistical Society. Series B (Methodological) 57, 289–300 (1995).

Source: PubMed

3
Suscribir