Randomized Controlled-Feeding Study of Dietary Emulsifier Carboxymethylcellulose Reveals Detrimental Impacts on the Gut Microbiota and Metabolome

Benoit Chassaing, Charlene Compher, Brittaney Bonhomme, Qing Liu, Yuan Tian, William Walters, Lisa Nessel, Clara Delaroque, Fuhua Hao, Victoria Gershuni, Lillian Chau, Josephine Ni, Meenakshi Bewtra, Lindsey Albenberg, Alexis Bretin, Liam McKeever, Ruth E Ley, Andrew D Patterson, Gary D Wu, Andrew T Gewirtz, James D Lewis, Benoit Chassaing, Charlene Compher, Brittaney Bonhomme, Qing Liu, Yuan Tian, William Walters, Lisa Nessel, Clara Delaroque, Fuhua Hao, Victoria Gershuni, Lillian Chau, Josephine Ni, Meenakshi Bewtra, Lindsey Albenberg, Alexis Bretin, Liam McKeever, Ruth E Ley, Andrew D Patterson, Gary D Wu, Andrew T Gewirtz, James D Lewis

Abstract

Background & aims: Epidemiologic and murine studies suggest that dietary emulsifiers promote development of diseases associated with microbiota dysbiosis. Although the detrimental impact of these compounds on the intestinal microbiota and intestinal health have been demonstrated in animal and in vitro models, impact of these food additives in healthy humans remains poorly characterized.

Methods: To examine this notion in humans, we performed a double-blind controlled-feeding study of the ubiquitous synthetic emulsifier carboxymethylcellulose (CMC) in which healthy adults consumed only emulsifier-free diets (n = 9) or an identical diet enriched with 15 g per day of CMC (n = 7) for 11 days.

Results: Relative to control subjects, CMC consumption modestly increased postprandial abdominal discomfort and perturbed gut microbiota composition in a way that reduced its diversity. Moreover, CMC-fed subjects exhibited changes in the fecal metabolome, particularly reductions in short-chain fatty acids and free amino acids. Furthermore, we identified 2 subjects consuming CMC who exhibited increased microbiota encroachment into the normally sterile inner mucus layer, a central feature of gut inflammation, as well as stark alterations in microbiota composition.

Conclusions: These results support the notion that the broad use of CMC in processed foods may be contributing to increased prevalence of an array of chronic inflammatory diseases by altering the gut microbiome and metabolome (ClinicalTrials.gov, number NCT03440229).

Keywords: Emulsifier; Metabolism; Metabolome; Microbiota.

Conflict of interest statement

Conflict of interest

Dr Lewis consulted or served on an advisory board for Eli Lilly and Company, Samsung Bioepis, UCB, Bristol-Myers Squibb, Nestle Health Science, Merck, Celgene, Janssen Pharmaceuticals, Bridge Biotherapeutics, Entasis Therapeutics, AbbVie, Pfizer, Gilead, Arena Pharmaceuticals, Protagonist Therapeutics, Amgen, and Scipher Medicine. He has had research funding from Nestle Health Science, Takeda, Janssen Pharmaceuticals, and AbbVie. The remaining authors disclose no conflicts.

Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.

Figures

Figure 1.
Figure 1.
Effect of CMC consumption on metabolic parameters. (A) Schematic representation of the inpatient study that enabled daily monitoring and specimen collections before, during, and after CMC consumption, or lack thereof, and presenting timing of oral glucose tolerance tests, intestinal biopsies, and feces collection. (B) Biomorphometric characterization of the study’s participants at the beginning of the study. (C) Effect of dietary emulsifier CMC consumption on various metabolic parameters, measured both pre- and postintervention. (DF) Effect of dietary emulsifier CMC consumption on weight (D), PROMISE gas/bloating (E), and belly pain (F) scores, measured both pre- and postintervention. OGTT, oral glucose tolerance test. Significance was determined using Mann-Whitney test; *P < 0.05 compared with control group.
Figure 2.
Figure 2.
Effect of CMC consumption on microbiota composition. (A) PCoA of the unweighted UniFrac distance matrix of the study participants’ microbiota assessed by 16S rRNA gene sequencing. All time points are included in the representation, and samples are colored by participants. (B) PCoA of the Bray-Curtis distance matrix at days 0, 9, and 14 of study participants’ microbiota composition after normalization of every SV based on day 4 value, with samples colored by group. (C) Changes in the microbial community structure over time, as measured by Bray-Curtis distance from day 4 to subsequent days, for each group. (D) Fecal bacterial load assessed by 16S quantitative polymerase chain reaction (qPCR). (E) Changes in Evenness and Shannon alpha diversity measures for CMC intervention versus control groups, at days 0, 9, and 14. Significance was determined using 2-way ANOVA corrected for multiple comparisons with a Bonferroni posttest (E), multiple t tests (E) or Permanova analysis (A, B).
Figure 3.
Figure 3.
Effect of CMC consumption on fecal metagenome. (A) PCoA of the Bray-Curtis dissimilarity of study’s participants metagenome (uniref90 categories) assessed by shotgun sequencing. Days 4 and 14 are included in the representation, and samples are colored by participants. (B, C) PCoA of the Bray-Curtis distance matrix at day 4 (B) and 14 (C) of study’s participants metagenome assessed by shotgun sequencing, with samples colored by group.
Figure 4.
Figure 4.
Effect of CMC consumption on the fecal metabolome. (A) Changes of the fecal level of bioactive lipopolysaccharide from day 0 to subsequent days measured with HEK-TLR4 reporter cells. (B) Changes of the fecal level of bioactive flagellin from day 0 to subsequent days measured with HEK-TLR5 reporter cells. (C) Changes of the fecal level of the inflammatory marker Lipocalin-2 from day 0 to subsequent days. (D) PCoA of the Euclidean distance at days 0, 9, and 14 of study participants’ fecal metabolome, with samples colored by group. (E) Heatmap presenting participants fecal metabolome over the course of the study. (F) Changes of the fecal level of CMC from day 0 to subsequent days in both control and CMC-treated groups. (G) Changes of the fecal level of CMC from day 0 to subsequent days in control group. Significance was determined using 2-way ANOVA corrected for multiple comparisons with a Bonferroni posttest or repeated t tests corrected with the false discovery rate approach for (G); *P < .05 compared with control group for (F), *P < .05 compared with day 0 for (G).
Figure 5.
Figure 5.
Intersubject variability in the response to CMC consumption. (A) Effect of dietary emulsifier CMC consumption on microbiota localization (distance of the closest bacteria from the surface of the epithelium), measured both pre- and postintervention. (B) Changes of the Bray-Curtis distance matrix, for each study’s participant from the CMC-treated group, from day 4 to subsequent days. (C) Changes of the Bray-Curtis distance matrix, for the CMC-Insensitive and the CMC-Sensitive groups, from day 4 to subsequent days. (D) Changes of the fecal level of bioactive LPS from day 0 to subsequent days measured with HEK-TLR4 reporter cells. (E) Changes of the fecal level of bioactive flagellin from day 0 to subsequent days measured with HEK-TLR5 reporter cells. (F) Changes of the fecal level of the inflammatory marker Lipocalin-2 from day 0 to subsequent days. (G) Effect of dietary emulsifier CMC consumption on fecal metagenome measured through Bray-Curtis distance. (H) Biomorphometric characterization of study’s participants at the beginning of the study and according to CMC sensitivity status. Significance was determined using 1-way ANOVA corrected for multiple comparisons with a Bonferroni posttest (A and G) or 2-way ANOVA corrected for multiple comparisons with a Bonferroni posttest (C, D, E, and F). BMI, body mass index; DBP, diastolic blood pressure; IEC, intestinal epithelial cell; LPS, lipopolysaccharide; NS, not statistically significant; SBP, systolic blood pressure.

References

    1. Marion-Letellier R, Amamou A, Savoye G, et al. Inflammatory bowel diseases and food additives: to add fuel on the flames. Nutrients 2019;11:1111.
    1. Martinez Steele E, Juul F, Neri D, et al. Dietary share of ultra-processed foods and metabolic syndrome in the US adult population. Prev Med 2019;125:40–48.
    1. Caruso R, Lo BC, Nunez G. Host-microbiota interactions in inflammatory bowel disease. Nat Rev Immunol 2020;20:411–426.
    1. Neurath MF. Host-microbiota interactions in inflammatory bowel disease. Nat Rev Gastroenterol Hepatol 2020;17:76–77.
    1. Dabke K, Hendrick G, Devkota S. The gut microbiome and metabolic syndrome. J Clin Invest 2019;129:4050–4057.
    1. Schultz J. Carboxymethylcellulose as a colloid laxative. Am J Dig Dis 1949;16:319–322.
    1. Smits CH, Veldman A, Verkade HJ, et al. The inhibitory effect of carboxymethylcellulose with high viscosity on lipid absorption in broiler chickens coincides with reduced bile salt concentration and raised microbial numbers in the small intestine. Poult Sci 1998;77:1534–1539.
    1. Naimi S, Viennois E, Gewirtz AT, et al. Direct impact of commonly used dietary emulsifiers on human gut microbiota. Microbiome 2021;9:66.
    1. Viennois E, Bretin A, Dube PE, et al. Dietary emulsifiers directly impact adherent-invasive E. coli gene expression to drive chronic intestinal inflammation. Cell Rep 2020;33:108229.
    1. Viennois E, Merlin D, Gewirtz AT, et al. Dietary emulsifier-induced low-grade inflammation promotes colon carcinogenesis. Cancer Res 2017;77:27–40.
    1. Chassaing B, Van de Wiele T, De Bodt J, et al. Dietary emulsifiers directly alter human microbiota composition and gene expression ex vivo potentiating intestinal inflammation. Gut 2017;66:1414–1427.
    1. Chassaing B, Koren O, Goodrich JK, et al. Dietary emulsifiers impact the mouse gut microbiota promoting colitis and metabolic syndrome. Nature 2015;519:92–96.
    1. Chassaing B, Raja SM, Lewis JD, et al. Colonic microbiota encroachment correlates with dysglycemia in humans. Cell Mol Gastroenterol Hepatol 2017;4:205–221.
    1. Krebs-Smith SM, Pannucci TE, Subar AF, et al. Update of the Healthy Eating Index: HEI-2015. J Acad Nutr Diet 2018;118:1591–1602.
    1. Panizza CE, Shvetsov YB, Harmon BE, et al. Testing the predictive validity of the Healthy Eating Index-2015 in the multiethnic cohort: is the score associated with a reduced risk of all-cause and cause-specific mortality? Nutrients 2018;10:452.
    1. Petimar J, Smith-Warner SA, Fung TT, et al. Recommendation-based dietary indexes and risk of colorectal cancer in the Nurses’ Health Study and Health Professionals Follow-up Study. Am J Clin Nutr 2018;108:1092–1103.
    1. Doucet E, Laviolette M, Imbeault P, et al. Total peptide YY is a correlate of postprandial energy expenditure but not of appetite or energy intake in healthy women. Metabolism 2008;57:1458–1464.
    1. Doucet E, Pomerleau M, Harper ME. Fasting and postprandial total ghrelin remain unchanged after short-term energy restriction. J Clin Endocrinol Metab 2004;89:1727–1732.
    1. Chassaing B, Srinivasan G, Delgado MA, et al. Fecal lipocalin 2, a sensitive and broadly dynamic non-invasive biomarker for intestinal inflammation. PLoS One 2012;7:e44328.
    1. Jiang L, Huang J, Wang Y, et al. Eliminating the dication-induced intersample chemical-shift variations for NMR-based biofluid metabonomic analysis. Analyst 2012;137:4209–4219.
    1. Tian Y, Zhang L, Wang Y, et al. Age-related topographical metabolic signatures for the rat gastrointestinal contents. J Proteome Res 2012;11:1397–1411.
    1. Armenta JM, Cortes DF, Pisciotta JM, et al. Sensitive and rapid method for amino acid quantitation in malaria biological samples using AccQ.Tag ultra performance liquid chromatography-electrospray ionization-MS/MS with multiple reaction monitoring. Anal Chem 2010;82:548–558.
    1. Cole CR, Frem JC, Schmotzer B, et al. The rate of bloodstream infection is high in infants with short bowel syndrome: relationship with small bowel bacterial over-growth, enteral feeding, and inflammatory and immune responses. J Pediatr 2010;156:941–947.e1.
    1. Fedirko V, Tran HQ, Gewirtz AT, et al. Exposure to bacterial products lipopolysaccharide and flagellin and hepatocellular carcinoma: a nested case-control study. BMC Med 2017;15:72.
    1. Wu GD, Chen J, Hoffmann C, et al. Linking long-term dietary patterns with gut microbial enterotypes. Science 2011;334:105–108.
    1. Sokol H, Pigneur B, Watterlot L, et al. Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc Natl Acad Sci U S A 2008;105:16731–16736.
    1. Quevrain E, Maubert MA, Michon C, et al. Identification of an anti-inflammatory protein from Faecalibacterium prausnitzii, a commensal bacterium deficient in Crohn’s disease. Gut 2016;65:415–425.
    1. Lenoir M, Martin R, Torres-Maravilla E, et al. Butyrate mediates anti-inflammatory effects of Faecalibacterium prausnitzii in intestinal epithelial cells through Dact3. Gut Microbes 2020;12:1–16.
    1. Larsen OFA, Claassen E. The mechanistic link between health and gut microbiota diversity. Sci Rep 2018;8:2183.
    1. Human Microbiome Project C Structure, function and diversity of the healthy human microbiome. Nature 2012;486:207–214.
    1. Bar A, Van Ommen B, Timonen M. Metabolic disposition in rats of regular and enzymatically depolymerized sodium carboxymethylcellulose. Food Chem Toxicol 1995;33:901–907.
    1. Johansson ME, Gustafsson JK, Holmen-Larsson J, et al. Bacteria penetrate the normally impenetrable inner colon mucus layer in both murine colitis models and patients with ulcerative colitis. Gut 2014;63:281–291.
    1. Chazelas E, Druesne-Pecollo N, Esseddik Y, et al. Exposure to food additive mixtures in 106,000 French adults from the NutriNet-Sante cohort. Sci Rep 2021;11:19680.
    1. Suez J, Korem T, Zeevi D, et al. Artificial sweeteners induce glucose intolerance by altering the gut microbiota. Nature 2014;514:181–186.

Source: PubMed

Подписаться