Supplementation with Akkermansia muciniphila in overweight and obese human volunteers: a proof-of-concept exploratory study

Clara Depommier, Amandine Everard, Céline Druart, Hubert Plovier, Matthias Van Hul, Sara Vieira-Silva, Gwen Falony, Jeroen Raes, Dominique Maiter, Nathalie M Delzenne, Marie de Barsy, Audrey Loumaye, Michel P Hermans, Jean-Paul Thissen, Willem M de Vos, Patrice D Cani, Clara Depommier, Amandine Everard, Céline Druart, Hubert Plovier, Matthias Van Hul, Sara Vieira-Silva, Gwen Falony, Jeroen Raes, Dominique Maiter, Nathalie M Delzenne, Marie de Barsy, Audrey Loumaye, Michel P Hermans, Jean-Paul Thissen, Willem M de Vos, Patrice D Cani

Abstract

Metabolic syndrome is characterized by a constellation of comorbidities that predispose individuals to an increased risk of developing cardiovascular pathologies as well as type 2 diabetes mellitus1. The gut microbiota is a new key contributor involved in the onset of obesity-related disorders2. In humans, studies have provided evidence for a negative correlation between Akkermansia muciniphila abundance and overweight, obesity, untreated type 2 diabetes mellitus or hypertension3-8. Since the administration of A. muciniphila has never been investigated in humans, we conducted a randomized, double-blind, placebo-controlled pilot study in overweight/obese insulin-resistant volunteers; 40 were enrolled and 32 completed the trial. The primary end points were safety, tolerability and metabolic parameters (that is, insulin resistance, circulating lipids, visceral adiposity and body mass). Secondary outcomes were gut barrier function (that is, plasma lipopolysaccharides) and gut microbiota composition. In this single-center study, we demonstrated that daily oral supplementation of 1010 A. muciniphila bacteria either live or pasteurized for three months was safe and well tolerated. Compared to placebo, pasteurized A. muciniphila improved insulin sensitivity (+28.62 ± 7.02%, P = 0.002), and reduced insulinemia (-34.08 ± 7.12%, P = 0.006) and plasma total cholesterol (-8.68 ± 2.38%, P = 0.02). Pasteurized A. muciniphila supplementation slightly decreased body weight (-2.27 ± 0.92 kg, P = 0.091) compared to the placebo group, and fat mass (-1.37 ± 0.82 kg, P = 0.092) and hip circumference (-2.63 ± 1.14 cm, P = 0.091) compared to baseline. After three months of supplementation, A. muciniphila reduced the levels of the relevant blood markers for liver dysfunction and inflammation while the overall gut microbiome structure was unaffected. In conclusion, this proof-of-concept study (clinical trial no. NCT02637115 ) shows that the intervention was safe and well tolerated and that supplementation with A. muciniphila improves several metabolic parameters.

Conflict of interest statement

Competing financial interests

A.E., C.D., H.P., P.D.C. and W.M.d.V. are inventors of patent applications (PCT/EP2013/073972; PCT/EP2016/071327, PCT/EP2016/060033 filed in European Patent Office (EP), Australia (AU), Brazil (BR), Canada (CA), China (CN), Eurasian Patent Organization (EA), Israel (IL), India (IN), Hong Kong (HK), Japan (JP), South Korea (KR), Mexico (MX), New Zealand (NZ), and the United States (US)) dealing with the use of A.muciniphila and its components in the context of obesity and related disorders. P.D.C. and W.M.d.V. are co-founders of A-Mansia Biotech SA.

Figures

Extended data figure 1. Flow chart of…
Extended data figure 1. Flow chart of the interventional study.
Diagram of the participant selection procedure, with the following information: number of individuals enrolled at each step of the study progress; number of individuals included in the final analysis; details of events that led to a reduction in the group size.
Extended data figure 2. Changes in inflammatory…
Extended data figure 2. Changes in inflammatory parameters and GLP-1.
(a) Soluble CD40 Ligand (sCD40L), (b) Growth-related oncogene (GRO/CXCL1), (c) Monocyte Chemoattractant Protein-1 (MCP-1) and (d) Glucagon-like peptide-1 (GLP-1). Differential values (MD and MD from placebo) are expressed as mean ± SEM, either as raw data or as percentages. Bars represent mean change from baseline value per group, with their standard errors. Mann-Whitney tests or unpaired T-tests were performed to compare differential values of both treated groups versus the Placebo group (inter-group changes), according to the distribution. The respective P value are indicated in the table. Lines represent raw values before and after 3 months of supplementation. Distribution of values within each group for each timing is illustrated by a box and whisker. In the box plots, the line in the middle of the box is plotted at the median, the inferior and superior limit of the box correspond to the 25th and the 75th percentiles respectively. Wilcoxon matched-pairs signed ranks tests or Paired T-tests were performed to verify changes from baseline (intra-group changes), according to the distribution and, when drawn, the capped line above the concerned group shows the corresponding P-value. Changes between 0 and 3 months across the 3 groups were analyzed with Kruskal-Wallis or One-way ANOVA according to the distribution and group-wise comparisons were performed using Bonferroni and Tuckey’s adjustment for multiple testing, respectively. MD, Mean Difference. Placebo, n = 11; Pasteurized, n =12; Alive, n=9 for all parameters except for GRO: Placebo, n = 7; Pasteurized, n =10; Alive, n=8. All tests were performed in two-tailed.
Extended data figure 3. Changes in faecal…
Extended data figure 3. Changes in faecal microbiome.
(a)Akkermansia muciniphila abundance in feces evaluated by Q-PCR. Differential values (MD and MD from placebo) are expressed as mean ± SEM as raw. Bars represent mean change from baseline value per group, with their standard errors. Mann-Whitney tests were performed to compare differential values of both treated groups versus the Placebo group (inter-group changes), according to the distribution. The respective P value are indicated in the table. Lines represent raw values before and after 3 months of supplementation. Distribution of values within each group for each timing is illustrated by a box and whisker. In the box plots, the line in the middle of the box is plotted at the median, the inferior and superior limit of the box correspond to the 25th and the 75th percentiles respectively. Wilcoxon matched-pairs signed ranks tests were performed to verify changes from baseline (intra-group changes), according to the distribution. When the difference is significative, a capped line is marked above the concerned group with the corresponding P value. Kruskal-Wallis analysis were used to compare changes between 0 and 3 months across the 3 groups, according to the distribution. MD, Mean Difference. Placebo, n = 11; Pasteurized, n =12; Alive, n=9. All tests were performed in two-tailed. *P < 0.05. (b) Visualization of the participants’ faecal microbiota composition at baseline and endpoint of the intervention. Faecal microbiota dissimilarity between samples is represented by principal coordinates analysis (genus-level Aitchison distance), with 6 sample groups - corresponding to the 3 different treatment arms at baseline or at endpoint represented by confidence ellipses (80% confidence interval). Intervention effects are symbolized by colored arrows, with direction and length corresponding to the shift in group centroid coordinates from baseline to endpoint for each treatment arm (re-scaled x4 and re-centered at baseline global centroid). Placebo, n = 11; Pasteurized, n =12; Alive, n=9.
Figure 1. Changes in parameters related to…
Figure 1. Changes in parameters related to glucose metabolism and WBC.
(a) Insulinemia, (b) Glycemia, (c) Insulin Resistance score, (d) Insulin Sensitivity, (e) DDPIV activity (f) and With blood cell count. Differential values (MD and MD from placebo) are expressed as mean + SEM, either as raw data or as percentages. Bars represent mean change from baseline value per group, with their standard errors. Mann-Whitney tests or unpaired T-tests were performed to compare differential values of both treated groups versus the Placebo group (inter-group changes), according to the distribution. The respective P value are indicated in the table and when the test is significative, bars are marked by * symbol. Lines represent raw values before and after 3 months of supplementation. Distribution of values within each group for each timing is illustrated by a box and whisker. In the box plots, the line in the middle of the box is plotted at the median, the inferior and superior limit of the box correspond to the 25th and the 75th percentiles respectively. The whiskers correspond to the minimum and maximum value. Wilcoxon matched-pairs signed ranks tests or Paired T-tests were performed to verify changes from baseline (intra-group changes), according to the distribution. When the difference is significative, a capped line is marked above the concerned group with the corresponding P value. Changes between 0 and 3 months across the 3 groups were analyzed with Kruskal-Wallis or One-way ANOVA according to the distribution and group-wise comparisons were performed using Bonferroni and Tuckey’s adjustment for multiple testing, respectively. When the difference is significative, a line is marked above the concerned groups with the corresponding P value. DPPIV, dipeptidyl peptidase-IV ; MD, Mean Difference; WBC, white blood cells. Placebo, n = 11; Pasteurized, n =12; Alive, n=9 for all parameters except for WBC : Placebo, n = 11; Pasteurized, n =11; Alive, n=8. All tests were performed in two-tailed. *P < 0.05.
Figure 2. Changes in parameters related to…
Figure 2. Changes in parameters related to lipid metabolism.
(a) Total cholesterol, (b) LDL-cholesterol and (c) Triglycerides. Differential values (MD and MD from placebo) are expressed as mean ± SEM, either as raw data or as percentages. Bars represent mean change from baseline value per group, with their standard errors. Mann-Whitney tests or unpaired T-tests were performed to compare differential values of both treated groups versus the Placebo group (inter-group changes), according to the distribution. The respective P value are indicated in the table and when the test is significative, bars are marked by * symbol. Lines represent raw values before and 3 months after receiving treatment. Distribution of values within each group for each timing is illustrated by a box and whisker. In the box plots, the line in the middle of the box is plotted at the median, the inferior and superior limit of the box correspond to the 25th and the 75th percentiles respectively. The whiskers correspond to the minimum and maximum value. Wilcoxon matched-pairs signed ranks tests or Paired T-tests were performed to verify changes from baseline (intra-group changes), according to the distribution,when drawn, the capped line above the concerned group shows the corresponding P-value. When the difference is significative, a capped line is marked above the concerned group. Changes between 0 and 3 months across the 3 groups were analyzed with Kruskal-Wallis or One-way ANOVA according to the distribution and group-wise comparisons were performed using Bonferroni and Tuckey’s adjustment for multiple testing, respectively. When the difference is significative, a line is marked above the concerned groups with the corresponding P value. MD, Mean Difference. Placebo, n = 11; Pasteurized, n =12; Alive, n=9 for all parameters. All tests were performed in two-tailed. *P < 0.05.
Figure 3. Changes in hepatic and general…
Figure 3. Changes in hepatic and general enzymes.
(a) Alanine aminotransferase activity (ALT), (b) Aspartate aminotransferase activity (AST), (c) ɣ-Glutamyltransferase (GGT), (d) Lipopolysaccharides (LPS), (e) Lactate Dehydrogenase (LDH) and (f) Creatine Kinase (CK). Differential values (MD and MD from placebo) are expressed as mean ± SEM, either as raw data or as percentages. Bars represent mean change from baseline value per group, with their standard errors. Mann-Whitney tests were performed to compare differential values of both treated groups versus the Placebo group (inter-group changes), according to the distribution. The respective P value are indicated in the table and when the test is significative, bars are marked by * symbol. Lines represent raw values before and after 3 months of supplementation. Distribution of values within each group for each timing is illustrated by a box and whisker. In the box plots, the line in the middle of the box is plotted at the median, the inferior and superior limit of the box correspond to the 25th and the 75th percentiles respectively. The whiskers correspond to the minimum and maximum value. Wilcoxon matched-pairs signed ranks tests were performed to verify changes from baseline (intra-group changes), according to the distribution. When the difference is significative, a capped line is marked above the concerned group with the corresponding P value. Kruskal-Wallis analysis were used to compare changes between 0 and 3 months across the 3 groups according to the distribution. All group-wise comparisons were performed using Bonferroni’s adjustment for multiple testing. When the difference is significative, a line is marked above the concerned groups with the corresponding P value. MD, Mean Difference. Placebo, n = 11; Pasteurized, n =12; Alive, n=9 for all parameters except for Creatine Kinase : Placebo, n = 11; Pasteurized, n =11; Alive, n=8. All tests were performed in two-tailed. *P < 0.05.
Figure 4. Changes in anthropometric parameters.
Figure 4. Changes in anthropometric parameters.
(a) Body weight, (b) Fat mass, (c) Hip circumference and (d) Waist circumference. Differential values (MD and MD from placebo) are expressed as mean ± SEM, either as raw data or as percentages. Bars represent mean change from baseline value per group, with their standard errors. Mann-Whitney tests were performed to compare differential values of both treated groups versus the Placebo group (inter-group changes), according to the distribution. The respective P value are indicated in the table. Lines represent raw values before and after 3 months of supplementation. Distribution of values within each group for each timing is illustrated by a box and whisker. In the box plots, the line in the middle of the box is plotted at the median, the inferior and superior limit of the box correspond to the 25th and the 75th percentiles respectively. The whiskers correspond to the minimum and maximum value. Wilcoxon matched-pairs signed ranks tests were performed to verify changes from baseline (intra-group changes), according to the distribution and, when drawn, the capped line above the concerned group shows the corresponding P-value. Kruskal-Wallis analysis was used to compare changes between 0 and 3 months across the 3 groups according to the distribution. All group-wise comparisons were performed using Bonferroni’s adjustment for multiple testing. MD, Mean Difference. Placebo, n = 11; Pasteurized, n =12; Alive, n=9 for all parameters except for Hip and Waist: Placebo, n = 10; Pasteurized, n =12; Alive, n=9. All tests were performed in two-tailed. MD, Mean Difference. All tests were performed in two-tailed.

References

    1. O'Neill S, O'Driscoll L. Metabolic syndrome: a closer look at the growing epidemic and its associated pathologies. Obesity reviews : an official journal of the International Association for the Study of Obesity. 2015;16:1–12.
    1. Cani PD, et al. Microbial regulation of organismal energy homeostasis. Nature Metabolism. 2019;1:34–46.
    1. Brahe LK, et al. Specific gut microbiota features and metabolic markers in postmenopausal women with obesity. Nutrition & diabetes. 2015;5:e159.
    1. Zhang X, et al. Human gut microbiota changes reveal the progression of glucose intolerance. PLoS One. 2013;8:e71108.
    1. Yassour M, et al. Sub-clinical detection of gut microbial biomarkers of obesity and type 2 diabetes. Genome Med. 2016;8:17.
    1. Li J, et al. Gut microbiota dysbiosis contributes to the development of hypertension. Microbiome. 2017;5:14.
    1. Liu R, et al. Gut microbiome and serum metabolome alterations in obesity and after weight-loss intervention. Nat Med. 2017;23:859–868.
    1. Dao MC, et al. Akkermansia muciniphila and improved metabolic health during a dietary intervention in obesity: relationship with gut microbiome richness and ecology. Gut. 2016;65:426–436.
    1. Cani PD, de Vos WM. Next-Generation Beneficial Microbes: The Case of Akkermansia muciniphila. Frontiers in microbiology. 2017;8:1765.
    1. Everard A, et al. Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proceedings of the National academy of sciences of the United States of America. 2013;110:9066–9071.
    1. Plovier H, et al. A purified membrane protein from Akkermansia muciniphila or the pasteurized bacterium improves metabolism in obese and diabetic mice. Nat Med. 2017;23:107–113.
    1. Everard A, et al. Intestinal epithelial N-acylphosphatidylethanolamine phospholipase D links dietary fat to metabolic adaptations in obesity and steatosis. Nature communications. 2019;10:457.
    1. Atkin SL, et al. Effect of dipeptidyl peptidase-4 inhibitors on circulating tumor necrosis factor-alpha concentrations: A systematic review and meta-analysis of controlled trials. J Diabetes Complications. 2017;31:1458–1464.
    1. Akoumianakis I, Antoniades C. Dipeptidyl peptidase IV inhibitors as novel regulators of vascular disease. Vascul Pharmacol. 2017;96-98:1–4.
    1. Olivares M, et al. The Potential Role of the Dipeptidyl Peptidase-4-Like Activity From the Gut Microbiota on the Host Health. Frontiers in microbiology. 2018;9:1900.
    1. Veronelli A, et al. White blood cells in obesity and diabetes: effects of weight loss and normalization of glucose metabolism. Diabetes Care. 2004;27:2501–2502.
    1. Ohshita K, et al. Elevated white blood cell count in subjects with impaired glucose tolerance. Diabetes Care. 2004;27:491–496.
    1. Gu Y, et al. White blood cells count as an indicator to identify whether obesity leads to increased risk of type 2 diabetes. Diabetes research and clinical practice. 2018;141:140–147.
    1. Zhang H, et al. White blood cell subtypes and risk of type 2 diabetes. J Diabetes Complications. 2017;31:31–37.
    1. Twig G, et al. White blood cells count and incidence of type 2 diabetes in young men. Diabetes Care. 2013;36:276–282.
    1. Shen J, et al. Low-Density Lipoprotein Receptor Signaling Mediates the Triglyceride-Lowering Action of Akkermansia muciniphila in Genetic-Induced Hyperlipidemia. Arteriosclerosis, thrombosis, and vascular biology. 2016;36:1448–1456.
    1. Li J, Lin S, Vanhoutte PM, Woo CW, Xu A. Akkermansia Muciniphila Protects Against Atherosclerosis by Preventing Metabolic Endotoxemia-Induced Inflammation in Apoe-/- Mice. Circulation. 2016;133:2434–2446.
    1. Ras RT, et al. Consumption of plant sterol-enriched foods and effects on plasma plant sterol concentrations--a meta-analysis of randomized controlled studies. Atherosclerosis. 2013;230:336–346.
    1. Wannamethee SG, Shaper AG, Lennon L, Whincup PH. Hepatic enzymes, the metabolic syndrome, and the risk of type 2 diabetes in older men. Diabetes Care. 2005;28:2913–2918.
    1. Rantala AO, et al. Gamma-glutamyl transpeptidase and the metabolic syndrome. J Intern Med. 2000;248:230–238.
    1. Lim JS, Lee DH, Park JY, Jin SH, Jacobs DR., Jr A strong interaction between serum gamma-glutamyltransferase and obesity on the risk of prevalent type 2 diabetes: results from the Third National Health and Nutrition Examination Survey. Clin Chem. 2007;53:1092–1098.
    1. Fraser A, et al. Alanine aminotransferase, gamma-glutamyltransferase, and incident diabetes: the British Women's Heart and Health Study and meta-analysis. Diabetes Care. 2009;32:741–750.
    1. Hanninen A, et al. Akkermansia muciniphila induces gut microbiota remodelling and controls islet autoimmunity in NOD mice. Gut. 2018;67:1445–1453.
    1. Grander C, et al. Recovery of ethanol-induced Akkermansia muciniphila depletion ameliorates alcoholic liver disease. Gut. 2017
    1. Wu W, et al. Protective Effect of Akkermansia muciniphila against Immune-Mediated Liver Injury in a Mouse Model. Frontiers in microbiology. 2017;8:1804.
    1. Harte AL, et al. Elevated endotoxin levels in non-alcoholic fatty liver disease. J Inflamm (Lond) 2010;7:15.
    1. Cani PD, et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes. 2007;56:1761–1772.
    1. Miele L, et al. Increased intestinal permeability and tight junction alterations in nonalcoholic fatty liver disease. Hepatology. 2009;49:1877–1887.
    1. Lassenius MI, et al. Bacterial endotoxin activity in human serum is associated with dyslipidemia, insulin resistance, obesity, and chronic inflammation. Diabetes Care. 2011;34:1809–1815.
    1. Allin KH, et al. Aberrant intestinal microbiota in individuals with prediabetes. Diabetologia. 2018;61:810–820.
    1. Wu H, et al. Metformin alters the gut microbiome of individuals with treatment-naive type 2 diabetes, contributing to the therapeutic effects of the drug. Nat Med. 2017;23:850–858.
    1. Dao MC, et al. Akkermansia muciniphila and improved metabolic health during a dietary intervention in obesity: relationship with gut microbiome richness and ecology. Gut. 2016;65:426–436.
    1. Scherbaum WA, et al. Efficacy and tolerability of vildagliptin in drug-naive patients with type 2 diabetes and mild hyperglycaemia*. Diabetes, obesity & metabolism. 2008;10:675–682.
    1. Rosenstock J, Rigby SP, Ford DM, Tao B, Chou HS. The glucose and lipid effects of colesevelam as monotherapy in drug-naive type 2 diabetes. Hormone and metabolic research = Hormon- und Stoffwechselforschung = Hormones et metabolisme. 2014;46:348–353.
    1. Kim SG, et al. Efficacy and safety of lobeglitazone monotherapy in patients with type 2 diabetes mellitus over 24-weeks: a multicenter, randomized, double-blind, parallel-group, placebo controlled trial. PLoS One. 2014;9:e92843.
    1. Werner M, Tonjes A, Stumvoll M, Thiery J, Kratzsch J. Assay-dependent variability of serum insulin levels during oral glucose tolerance test: influence on reference intervals for insulin and on cut-off values for insulin sensitivity indices. Clin Chem Lab Med. 2008;46:240–246.
    1. Borza D, et al. Influence of assay-dependent variability of serum insulin levels on insulin sensitivity indices. Clin Chem Lab Med. 2008;46:1655–1656.
    1. Wallace TM, Levy JC, Matthews DR. Use and abuse of HOMA modeling. Diabetes Care. 2004;27:1487–1495.
    1. Dennis JM, et al. Precision Medicine in Type 2 Diabetes: Clinical Markers of Insulin Resistance Are Associated With Altered Short- and Long-term Glycemic Response to DPP-4 Inhibitor Therapy. Diabetes Care. 2018;41:705–712.
    1. Vandeputte D, et al. Quantitative microbiome profiling links gut community variation to microbial load. Nature. 2017;551:507–511.
    1. Callahan BJ, et al. DADA2: High-resolution sample inference from Illumina amplicon data. Nature methods. 2016;13:581–583.
    1. Wang Q, Garrity GM, Tiedje JM, Cole JR. Naive Bayesian classifier for rapid assignment of rRNA sequences into the new bacterial taxonomy. Appl Environ Microbiol. 2007;73:5261–5267.
    1. Oksanen JB, et al. vegan: Community Ecology Package. R package version 2.5-3. 2018
    1. Gloor GB, Reid G. Compositional analysis: a valid approach to analyze microbiome high-throughput sequencing data. Can J Microbiol. 2016;62:692–703.

Source: PubMed

3
Suscribir