Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Microbiome-derived ethanol in nonalcoholic fatty liver disease

Abstract

To test the hypothesis that the gut microbiota of individuals with nonalcoholic fatty liver disease (NAFLD) produce enough ethanol to be a driving force in the development and progression of this complex disease, we performed one prospective clinical study and one intervention study. Ethanol was measured while fasting and 120 min after a mixed meal test (MMT) in 146 individuals. In a subset of 37 individuals and in an external validation cohort, ethanol was measured in portal vein blood. In an intervention study, ten individuals with NAFLD and ten overweight but otherwise healthy controls were infused with a selective alcohol dehydrogenase (ADH) inhibitor before an MMT. When compared to fasted peripheral blood, median portal vein ethanol concentrations were 187 (interquartile range (IQR), 17–516) times higher and increased with disease progression from 2.1 mM in individuals without steatosis to 8.0 mM in NAFL 21.0 mM in nonalcoholic steatohepatitis. Inhibition of ADH induced a 15-fold (IQR,1.6- to 20-fold) increase in peripheral blood ethanol concentrations in individuals with NAFLD, although this effect was abolished after antibiotic treatment. Specifically, Lactobacillaceae correlated with postprandial peripheral ethanol concentrations (Spearman’s rho, 0.42; P < 10−5) in the prospective study. Our data show that the first-pass effect obscures the levels of endogenous ethanol production, suggesting that microbial ethanol could be considered in the pathogenesis of this highly prevalent liver disease.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Ethanol concentrations in different blood compartments and studies.
Fig. 2: Gut microbiome analyses of the studies.

Similar content being viewed by others

Data availability

Fecal metagenomics and liver transcriptomics data have been deposited in the European Nucleotide Archive (ENA; PRJEB47902) and European Genome-Phenome Archive (EGAS00001005704), respectively. A comprehensive data analysis report can be found at https://amcmc.github.io/BARIA_ETHANASH/.Source data which are provided with this paper.

References

  1. Zhu, L. et al. Characterization of gut microbiomes in nonalcoholic steatohepatitis (NASH) patients: a connection between endogenous alcohol and NASH. Hepatology 57, 601–609 (2013).

    Article  CAS  Google Scholar 

  2. Yuan, J. et al. Fatty liver disease caused by high-alcohol-producing Klebsiella pneumoniae. Cell Metab. 30, 675–688 (2019).

    Article  CAS  Google Scholar 

  3. Younossi, Z. et al. Global perspectives on non-alcoholic fatty liver disease and non-alcoholic steatohepatitis. Hepatology https://doi.org/10.1002/hep.30251 (2018).

    Article  PubMed  Google Scholar 

  4. Farrell, G. C. & Larter, C. Z. Nonalcoholic fatty liver disease: from steatosis to cirrhosis. Hepatology 43, S99–S112 (2006).

    Article  CAS  Google Scholar 

  5. Sym, W. K., Teaberry, V., Choi, S. S. & Diehl, A. M. Similarities and differences in the pathogenesis of alcoholic and nonalcoholic steatohepatitis. Semin Liver Dis. 29, 200–210 (2009).

    Article  Google Scholar 

  6. Diehl, A. M. & Day, C. Cause, pathogenesis, and treatment of nonalcoholic steatohepatitis. N. Engl. J. Med. 377, 2063–2072 (2017).

    Article  CAS  Google Scholar 

  7. Thursz, M. et al. EASL clinical practice guidelines: management of alcohol-related liver disease. J. Hepatol. 69, 154–181 (2018).

    Article  Google Scholar 

  8. Engstler, A. J. et al. Insulin resistance alters hepatic ethanol metabolism: studies in mice and children with non-alcoholic fatty liver disease. Gut 65, 1–8 (2015).

    Google Scholar 

  9. Cope, K., Risby, T. & Diehl, A. M. Increased gastrointestinal ethanol production in obese mice: implications for fatty liver disease pathogenesis. Gastroenterology 119, 1340–1347 (2000).

    Article  CAS  Google Scholar 

  10. de Medeiros, I. C. & de Lima, J. G. Is nonalcoholic fatty liver disease an endogenous alcoholic fatty liver disease?—A mechanistic hypothesis. Med. Hypotheses 85, 148–152 (2015).

    Article  Google Scholar 

  11. Simic, M., Ajdukovic, N., Veselinovic, I., Mitrovic, M. & Djurendic-Brenesel, M. Endogenous ethanol production in patients with diabetes mellitus as a medicolegal problem. Forensic Sci. Int. 216, 97–100 (2012).

    Article  CAS  Google Scholar 

  12. Jeon, S. & Carr, R. Alcohol effects on hepatic lipid metabolism. J. Lipid Res. 61, 470–479 (2020).

    Article  CAS  Google Scholar 

  13. Cederbaum, A. I. Alcohol metabolism. Clin. Liver Dis. 16, 667 (2012).

    Article  Google Scholar 

  14. Wilson, D. F. & Matschinsky, F. M. Ethanol metabolism: the good, the bad, and the ugly. Med. Hypotheses 140, 109638 (2020).

    Article  CAS  Google Scholar 

  15. van Olden, C. et al. A systems biology approach to understand gut microbiota and host metabolism in morbid obesity: design of the BARIA Longitudinal Cohort Study. Journal of Internal Medicine joim.13157 https://doi.org/10.1111/joim.13157 (2020).

  16. Nascimbeni, F. et al. Clinical validation of the FLIP algorithm and the SAF score in patients with non-alcoholic fatty liver disease. J. Hepatol. https://doi.org/10.1016/j.jhep.2019.12.008 (2020).

    Article  PubMed  Google Scholar 

  17. Mardinoglu, A. et al. An integrated understanding of the rapid metabolic benefits of a carbohydrate-restricted diet on hepatic steatosis in humans. Cell Metab. 27, 1547–1558 (2018).

    Article  Google Scholar 

  18. Francque, S. M. et al. A randomized, controlled trial of the Pan-PPAR agonist lanifibranor in NASH. N. Engl. J. Med. 385, 1547–1558 (2021).

    Article  CAS  Google Scholar 

  19. Elshaghabee, F. et al. Ethanol production by selected intestinal microorganisms and lactic acid bacteria growing under different nutritional conditions. Front. Microbiol. 7, 47 (2016).

    Article  Google Scholar 

  20. Pyo, J. H. et al. Proton pump inhibitors use and the risk of fatty liver disease: a nationwide cohort study. J. Gastroenterol. Hepatol. 36, 1235–1243 (2021).

    Article  CAS  Google Scholar 

  21. Krautkramer, K. A., Fan, J. & Bäckhed, F. Gut microbial metabolites as multi-kingdom intermediates. Nat. Rev. Microbiol. 19, 77–94 (2020).

    Article  Google Scholar 

  22. Fell, J. C. & Scherer, M. Estimation of the potential effectiveness of lowering the blood alcohol concentration (BAC) limit for driving from 0.08 to 0.05 grams per deciliter in the United States. Alcohol. Clin. Exp. Res. 41, 2128–2139 (2017).

    Article  Google Scholar 

  23. Nosova, T. et al. Aldehyde dehydrogenase activity and acetate production by aerobic bacteria representing the normal flora of human large intestine. Alcohol Alcohol. 31, 555–564 (1996).

    Article  CAS  Google Scholar 

  24. Frantz, J. C. & McCallum, R. E. Growth yields and fermentation balance of Bacteroides fragilis cultured in glucose-enriched medium. J. Bacteriol. 137, 1263–1270 (1979).

    Article  CAS  Google Scholar 

  25. Amaretti, A. et al. Kinetics and metabolism of Bifidobacterium adolescentis MB 239 growing on glucose, galactose, lactose, and galactooligosaccharides. Appl. Environ. Microbiol. 73, 3637–3644 (2007).

    Article  CAS  Google Scholar 

  26. Weimer, P. J. & Zeikus, J. G. Fermentation of cellulose and cellobiose by Clostridium thermocellum in the absence and presence of Methanobacterium thermoautotrophicum. Appl. Environ. Microbiol. 33, 289–297 (1977).

    Article  CAS  Google Scholar 

  27. del Chierico, F. et al. Gut microbiota profiling of pediatric nonalcoholic fatty liver disease and obese patients unveiled by an integrated meta-omics-based approach. Hepatology 65, 451–464 (2017).

    Article  CAS  Google Scholar 

  28. Deschasaux, M. et al. Depicting the composition of gut microbiota in a population with varied ethnic origins but shared geography. Nat. Med. 24, 1526–1531 (2018).

    Article  CAS  Google Scholar 

  29. He, Y. et al. Regional variation limits applications of healthy gut microbiome reference ranges and disease models. Nat. Med. 24, 1532–1535 (2018).

    Article  CAS  Google Scholar 

  30. Fuchs, H. F. et al. Benefits of bariatric surgery do not reach obese men. J. Laparoendosc. Adv. Surg. Tech. A 25, 196–201 (2015).

    Article  Google Scholar 

  31. Seitz, H. K. et al. Alcoholic liver disease. Nat. Rev. Dis. Prim. 4, 16 (2018).

    Article  Google Scholar 

  32. Meijnikman, A. S. et al. Distinct differences in gut microbial composition and functional potential from lean to morbidly obese subjects. J. Intern. Med. https://doi.org/10.1111/joim.13137 (2020).

  33. Kleiner, D. E. et al. Design and validation of a histological scoring system for nonalcoholic fatty liver disease. Hepatology https://doi.org/10.1002/hep.20701 (2005).

  34. Bedosa, P. & FLIP Pathology Consortium Utility and appropriateness of the fatty liver inhibition of progression (FLIP) algorithm and steatosis, activity, and fibrosis (SAF) score in the evaluation of biopsies of nonalcoholic fatty liver disease.Hepatology 60, 565–575 (2014).

    Article  Google Scholar 

  35. Costea, P. I. et al. Towards standards for human fecal sample processing in metagenomic studies. Nat. Biotechnol. https://doi.org/10.1038/nbt.3960 (2017).

    Article  PubMed  Google Scholar 

  36. Karlsson, F. H., Nookaew, I. & Nielsen, J. Metagenomic data utilization and analysis (MEDUSA) and construction of a global gut microbial gene catalogue. PLoS Comput. Biol. 10, e1003706 (2014).

    Article  Google Scholar 

  37. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods https://doi.org/10.1038/nmeth.1923 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Chamberlain, S. A. & Szöcs, E. taxize: taxonomic search and retrieval in R. F1000Res 2, 191 (2013).

  39. Soverini, M. et al. HumanMycobiomeScan: a new bioinformatics tool for the characterization of the fungal fraction in metagenomic samples. BMC Genomics 20, 496 (2019).

  40. Edgar, R. C. Search and clustering orders of magnitude faster than BLAST. Bioinformatics 26, 2460–2461 (2010).

    Article  CAS  Google Scholar 

  41. Edgar, R. C. UNOISE2: improved error-correction for Illumina 16S and ITS amplicon sequencing. Preprint at bioRxiv 081257 https://doi.org/10.1101/081257 (2016).

  42. Wang, Q., Garrity, G. M., Tiedje, J. M. & Cole, J. R. Naive Bayesian classifier for rapid assignment of rRNA sequences into the new bacterial taxonomy. Appl. Environ. Microbiol. 73, 5261–5267 (2007).

    Article  CAS  Google Scholar 

  43. Quast, C. et al. The SILVA ribosomal RNA gene database project: improved data processing and web-based tools. Nucleic Acids Res. 41, D590–D596 (2013).

    Article  CAS  Google Scholar 

  44. Bolger, A. M., Lohse, M. & Usadel, B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014).

    Article  CAS  Google Scholar 

  45. Bray, N. L., Pimentel, H., Melsted, P. & Pachter, L. Near-optimal probabilistic RNA-seq quantification. Nat. Biotechnol. 34, 525–527 (2016).

    Article  CAS  Google Scholar 

  46. R Core Team. R: A Language and Environment for Statistical Computing. (R Foundation for Statistical Computing, 2013).

  47. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  Google Scholar 

  48. Oksanen, J. et al. Package ‘vegan’. Community Ecology Package. (2019).

  49. Benjamini, Y. & Hochberg, Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. Roy. Stat. Soc. B 57, 289–300 (1995).

    Google Scholar 

Download references

Acknowledgements

M.N. is supported by a ZONMW VICI grant 2020 (number 09150182010020). H.H. is supported by a Senior Fellowship of the Dutch Diabetes Research Foundation (2019.82.004). A.S.M. is supported by a Lilly/EFSD grant for Young Investigators. O.A. and V.T. are appointed on a NNF GUTMMM grant 2016 NNF15OC0016798 (to M.N., T.W.S. and F.B.). The study reported here was additionally supported by Le Ducq consortium grant 17CVD01 to F.B. and M.N. M.N, H.H. and A.K.G. are supported by a Dutch Heart Foundation CVON IN CONTROL-2 consortium grant. S.F. has a senior clinical research mandate from the Fund for Scientific Research (FWO) Flanders (1802154N). A.G.H. is supported by the Amsterdam UMC Fellowship, two TKI-PPP grants from Health~Holland and the Gilead Research Scholar grant.

Author information

Authors and Affiliations

Authors

Contributions

A.S., M.N., A.K., FB. and V.G. conceived and designed the studies. A.S.M., H.H., M.D., O.A., S.B., M.L.D.B. and J.V. managed the clinical studies and collection of stool samples and clinical data. M.K. was responsible for the study medication. A.S.M., O.A., A.G.H. and M.T. were responsible for the inclusion of participants. E.D., J.W., A.V., C.D.B., L.V. and S.F. delivered the samples of the validation cohort. M.D., V.T., F.B., T.S. and J.N. oversaw the processing of the data. M.D., A.K. and A.S.M. analyzed the data. A.S.M., M.D., H.H., U.B., A.K. and M.N. wrote the manuscript, with input from all the authors.

Corresponding author

Correspondence to Max Nieuwdorp.

Ethics declarations

Competing interests

M.N. is on the scientific board of Caelus Pharmaceuticals, Netherlands. F.B. is on the scientific board of Metabogen AB, Sweden. However, none of these are directly relevant for the current paper. S.F. has acted as advisor and/or lecturer for Roche, Gilead, Abbvie, Bayer, BMS, MSD, Janssen, Actelion, Astellas, Genfit, Inventiva, Intercept, Genentech, Galmed, Promethera, Coherus and NGM Bio. However, none of these are directly relevant for the current paper. A.G.H. has consulted for Novo Nordisk, Gilead, Amgen, Echosens and Julius Clinical. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Medicine thanks Jasmohan Bajaj and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: Jennifer Sargent, in collaboration with the Nature Medicine team.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Portal ethanol concentrations linked to NAFLD disease markers in two independent cohorts.

Portal ethanol concentrations linked to histological scores for NAFLD (a–d). Portal vein ethanol in individuals with NAFL and NASH compared to those without steatosis (e). Ethanol concentrations between different SAF-Activity scores (f) and Nonalcoholic Fatty Liver Disease Activity Scores (g). Box plots feature the median (center line), upper and lower quartiles (box limits), 1.5× the interquartile range (whiskers), points outside of boxplot range are outliers. Significant differences were determined by two-tailed Mann–Whitney test (****P < 0.0001;***P < 0.001; **P < 0.01; *P < 0.05).

Source data

Extended Data Fig. 2 Peripheral ethanol concentrations linked to NAFLD disease markers in fasted and post prandial state.

Peripheral ethanol concentrations in relation with histological scores for NAFLD (a–d). Ethanol concentrations in individuals with NAFL and NASH compared to those without steatosis (e). Peripheral ethanol concentrations between different SAF-Activity scores (f) and Nonalcoholic Fatty Liver Disease Activity Scores (g). Box plots feature the median (center line), upper and lower quartiles (box limits), 1.5× the interquartile range (whiskers), points outside of boxplot range are outliers. Significant differences were determined by two-tailed Mann–Whitney test (****P < 0.0001;***P < 0.001; **P < 0.01; *P < 0.05).

Source data

Extended Data Fig. 3 Correlations between portal, fasted and post-prandial peripheral ethanol levels and insulin levels.

Spearman’s Rho correlations are given, and lines represent an overall linear fit, with grey areas the standard error with a 0.95 confidence interval.

Source data

Extended Data Fig. 4 Correlations between portal and peripheral ethanol levels.

Correlations between portal and fasting peripheral ethanol (a) and post prandial peripheral ethanol (b) concentrations. Correlation strength was tested with Spearman’s rank correlation. Colored lines represent a linear fit for each of the NAFLD classes while black represents the overall fit.

Source data

Extended Data Fig. 5 MA plots of liver gene expression in association with NAFLD and ethanol concentrations.

MA plots for fasted peripheral (a), post prandial peripheral (b) and portal (c) ethanol concentrations, between No Steatosis and NAFL (d) and NASH (e). Significant differentially abundant genes are highlighted in blue. NADH2A and CYB2E1 specifically are highlighted in red.

Source data

Extended Data Fig. 6 Small intestinal Streptococcus abundance associates with NAFLD and ethanol concentrations.

Relative abundance is increased in NAFL and NASH (a) and correlates with fasted (b) and post-prandial (c) peripheral and portal (d) ethanol concentrations. Difference in abundance was tested with two-tailed Mann–Whitney test Line represents a linear fit with grey areas the standard error with a 0.95 confidence interval. Spearman’s Rho correlation coefficients are shown. Box plots feature the median (center line), upper and lower quartiles (box limits), 1.5× the interquartile range (whiskers), points outside of boxplot range are outliers.

Source data

Extended Data Fig. 7 Prospective cohort microbiome alpha diversity analysis.

Shannon’s diversity Index with NAFLD classes (a) shown with violin plots, portal ethanol concentrations (b), fasted peripheral ethanol concentrations (c), post prandial ethanol concentrations (d). Differences in alpha diversity between NAFLD classes were determined by Kruskal-Wallis test. Correlations were tested using spearman’s rank correlation.

Source data

Extended Data Fig. 8 Prospective cohort microbiome beta diversity analysis.

NAFLD classes (a), portal ethanol concentrations (b), fasted peripheral ethanol concentrations (c), post prandial ethanol concentrations (d). Bray-Curtis was used as a distance metrics and dissimilarity was tested using permanova using 1000 permutations.

Source data

Extended Data Fig. 9 Prospective cohort microbiome associations.

S. cerevisiae abundance associations with NAFLD classes (a), portal ethanol concentrations (b), fasted peripheral ethanol concentrations (c), post prandial ethanol concentrations (d). Significant differences in abundance were determined by two-tailed Mann–Whitney test (*P < 0.05). Correlations were tested using spearman’s rank correlation.

Source data

Extended Data Fig. 10 Microbiome characteristics of the intervention study.

Alpha- diversity in NASH (a), PCoA of Bray-Curtis dissimilarity between NAFLD class (b), PCoA of Bray Curtis dissimilarity with rate of ethanol accumulation after 4-methylpyrazole infusion (c), Volcano plot with differential taxa abundance analysis (d). Alpha diversity metrics was compared using a two-tailed Mann–Whitney test. Box plots feature the median (center line), upper and lower quartiles (box limits), 1.5× the interquartile range (whiskers), points outside of boxplot range are outliers. Beta diversity was tested using permanova using 1000 permutations. Differential taxa abundance was tested using deSeq2 and observed p values were Benjamini & Hochberg corrected.

Source data

Supplementary information

Reporting Summary

Supplementary Tables

Supplementary Table 1. Patient characteristics table portal subcohort. Supplementary Table 2. Patient characteristics table portal validation cohort. Supplementary Table 3. Differential expression and gene set enrichment analysis results. Supplementary Table 4. Correlation analysis between ethanol concentrations and fecal microbiome. Supplementary Table 5. Correlation analysis between KEGG orthologous functions and ethanol levels. Supplementary Table 6 DESeq2 analyses of gut microbiome in the intervention study.

Source data

Source Data Fig. 1

Source data for Fig. 1

Source Data Fig. 2

Source data for Fig. 2

Source Data Extended Data Fig. 1

Source data for Extended Data Fig. 1

Source Data Extended Data Fig. 2

Source data for Extended Data Fig. 2

Source Data Extended Data Fig. 3

Source data for Extended Fig. 3

Source Data Extended Data Fig. 4

Source data for Extended Data Fig. 4

Source Data Extended Data Fig. 5

Source data for Extended Data Fig. 5

Source Data Extended Data Fig. 6

Source data for Extended Data Fig. 6

Source Data Extended Data Fig. 7

Source data for Extended Data Fig. 7

Source Data Extended Data Fig. 8

Source data for Extended Data Fig. 8

Source Data Extended Data Fig. 9

Source data for Extended Data Fig. 9

Source Data Extended Data Fig. 10

Source data for Extended Data Fig. 10

Rights and permissions

Springer Nature or its licensor holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Meijnikman, A.S., Davids, M., Herrema, H. et al. Microbiome-derived ethanol in nonalcoholic fatty liver disease. Nat Med 28, 2100–2106 (2022). https://doi.org/10.1038/s41591-022-02016-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-022-02016-6

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing