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:

The gut–brain axis underlying hepatic encephalopathy in liver cirrhosis

Abstract

Up to 50–70% of patients with liver cirrhosis develop hepatic encephalopathy (HE), which is closely related to gut microbiota dysbiosis, with an unclear mechanism. Here, by constructing gut–brain modules to assess bacterial neurotoxins from metagenomic datasets, we found that phenylalanine decarboxylase (PDC) genes, mainly from Ruminococcus gnavus, increased approximately tenfold in patients with cirrhosis and higher in patients with HE. Cirrhotic, not healthy, mice colonized with R. gnavus showed brain phenylethylamine (PEA) accumulation, along with memory impairment, symmetrical tremors and cortex-specific neuron loss, typically found in patients with HE. This accumulation of PEA was primarily driven by decreased monoamine oxidase-B activity in both the liver and serum due to cirrhosis. Targeting PDC or PEA reversed the neurological symptoms induced by R. gnavus. Furthermore, fecal microbiota transplantation from patients with HE to germ-free cirrhotic mice replicated these symptoms and further corroborated the efficacy of targeting PDC or PEA. Clinically, high baseline PEA levels were linked to a sevenfold increased risk of HE after intrahepatic portosystemic shunt procedures. Our findings expand the understanding of the gut–liver–brain axis and identify a promising therapeutic and predictive target for HE.

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: The gene abundance of R. gnavus-derived PDC is increased in patients with cirrhosis across countries.
Fig. 2: R. gnavus leads to a pathological accumulation of PEA in the brain when MAO-B activity is reduced owing to cirrhosis.
Fig. 3: R. gnavus induces HE-like syndromes in cirrhotic mice via PDC.
Fig. 4: Targeting PDC alleviates neurotoxicity induced by fecal microbiota from patients with HE.
Fig. 5: Cortex-specific neuronal damage is the hallmark of HE fecal-microbiota-induced neurotoxicity.
Fig. 6: Serum PEA level correlates with fecal PDC-encoding gene abundance and high baseline PEA level links to increased risk of HE after TIPS.

Similar content being viewed by others

Data availability

The metagenomic datasets used in this study have been deposited in the European Nucleotide Archive (ENA) at EMBL-EBI under accession number PRJEB65440. The 16S rRNA sequence datasets in this study have been deposited in the ENA at EMBL-EBI under accession number PRJEB66346. All relevant sample-level patient data used in this study are provided in Supplementary Table 17. The six previously published studies used are available under accession numbers PRJEB6337 (ref. 22) and PRJEB14215 (ref. 23) (Supplementary Table 19), and GSE84044 (ref. 31), GSE48452 (ref. 32), GSE49541 (ref. 32) and GSE41919 (ref. 36). Associated sample-level clinical metadata for external datasets were collected from their relevant publications. Additional public databases used in this study include MetaCyc (https://biocyc.org/download.shtml), UniRef90 database (https://ftp.uniprot.org/pub/databases/uniprot/current_release/), GRCm38, M11 (https://www.gencodegenes.org/mouse/release_M11.html) and NCBI37 human genome (https://www.ncbi.nlm.nih.gov/datasets/genome/GCF_000001405.25/). Source data are provided with this paper.

Code availability

All code is available via the lab’s GitHub page at https://github.com/ZJJY-Bioinformatics/Cirrhosis-AADC.

References

  1. Asrani, S. K., Devarbhavi, H., Eaton, J. & Kamath, P. S. Burden of liver diseases in the world. J. Hepatol. 70, 151–171 (2019).

    PubMed  Google Scholar 

  2. GBD 2017 Cirrhosis Collaborators. The global, regional, and national burden of cirrhosis by cause in 195 countries and territories, 1990–2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet Gastroenterol. Hepatol. 5, 245–266 (2020).

    Google Scholar 

  3. Häussinger, D. et al. Hepatic encephalopathy. Nat. Rev. Dis. Primers 8, 43 (2022).

    PubMed  Google Scholar 

  4. Morais, L. H., Schreiber, H. L. IV. & Mazmanian, S. K. The gut microbiota–brain axis in behaviour and brain disorders. Nat. Rev. Microbiol. 19, 241–255 (2021).

    CAS  PubMed  Google Scholar 

  5. Rai, R., Saraswat, V. A. & Dhiman, R. K. Gut microbiota: its role in hepatic encephalopathy. J. Clin. Exp. Hepatol. 5, S29–S36 (2015).

    PubMed  Google Scholar 

  6. Chen, Z. et al. The role of intestinal bacteria and gut–brain axis in hepatic encephalopathy. Front. Cell. Infect. Microbiol. 10, 595759 (2021).

    PubMed  PubMed Central  Google Scholar 

  7. Mancini, A., Campagna, F., Amodio, P. & Tuohy, K. M. Gut:liver:brain axis: the microbial challenge in the hepatic encephalopathy. Food Funct. 9, 1373–1388 (2018).

    CAS  PubMed  Google Scholar 

  8. Patel, V. C. et al. Rifaximin-α reduces gut-derived inflammation and mucin degradation in cirrhosis and encephalopathy: RIFSYS randomised controlled trial. J. Hepatol. 76, 332–342 (2022).

    CAS  PubMed  Google Scholar 

  9. Bass, N. M. et al. Rifaximin treatment in hepatic encephalopathy. N. Engl. J. Med. 362, 1071–1081 (2010).

    CAS  PubMed  Google Scholar 

  10. Riordan, S. M. & Williams, R. Gut flora and hepatic encephalopathy in patients with cirrhosis. N. Engl. J. Med. 362, 1140–1142 (2010).

    PubMed  Google Scholar 

  11. Kao, D. et al. Fecal microbiota transplantation in the management of hepatic encephalopathy. Hepatology 63, 339–340 (2016).

    PubMed  Google Scholar 

  12. Bajaj, J. S. et al. Fecal microbiota transplant from a rational stool donor improves hepatic encephalopathy: a randomized clinical trial. Hepatology 66, 1727–1738 (2017).

    CAS  PubMed  Google Scholar 

  13. Rose, C. F. et al. Hepatic encephalopathy: novel insights into classification, pathophysiology and therapy. J. Hepatol. 73, 1526–1547 (2020).

    PubMed  Google Scholar 

  14. Görg, B. et al. O-GlcNAcylation-dependent upregulation of HO1 triggers ammonia-induced oxidative stress and senescence in hepatic encephalopathy. J. Hepatol. 71, 930–941 (2019).

    PubMed  Google Scholar 

  15. James, J. H., Ziparo, V., Jeppsson, B. & Fischer, J. E. Hyperammonaemia, plasma amino acid imbalance, and blood–brain amino acid transport: a unified theory of portal-systemic encephalopathy. Lancet 314, 772–775 (1979).

    Google Scholar 

  16. Mora, D. & Arioli, S. Microbial urease in health and disease. PLoS Pathog. 10, e1004472 (2014).

    PubMed  PubMed Central  Google Scholar 

  17. Ge, P. S. & Runyon, B. A. Serum ammonia level for the evaluation of hepatic encephalopathy. JAMA 312, 643–644 (2014).

    CAS  PubMed  Google Scholar 

  18. Ninan, J. & Feldman, L. Ammonia levels and hepatic encephalopathy in patients with known chronic liver disease. J. Hosp. Med. 12, 659–661 (2017).

    PubMed  Google Scholar 

  19. Haj, M. & Rockey, D. C. Ammonia levels do not guide clinical management of patients with hepatic encephalopathy caused by cirrhosis. Am. J. Gastroenterol. 115, 723–728 (2020).

    PubMed  Google Scholar 

  20. Valles-Colomer, M. et al. The neuroactive potential of the human gut microbiota in quality of life and depression. Nat. Microbiol. 4, 623–632 (2019).

    CAS  PubMed  Google Scholar 

  21. Li, X. et al. Prevalence and risk factors for minimal hepatic encephalopathy in cirrhotic patients with different etiologies. Portal Hypertens. Cirrhosis 2, 171–180 (2023).

    CAS  Google Scholar 

  22. Qin, N. et al. Alterations of the human gut microbiome in liver cirrhosis. Nature 513, 59–64 (2014).

    CAS  PubMed  Google Scholar 

  23. Hoyles, L. et al. Molecular phenomics and metagenomics of hepatic steatosis in non-diabetic obese women. Nat. Med. 24, 1070–1080 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Oldendorf, W. H. Brain uptake of radiolabeled amino acids, amines, and hexoses after arterial injection. Am. J. Physiol. 221, 1629–1639 (1971).

    CAS  PubMed  Google Scholar 

  25. Gainetdinov, R. R., Hoener, M. C. & Berry, M. D. Trace amines and their receptors. Pharmacol. Rev. 70, 549–620 (2018).

    CAS  PubMed  Google Scholar 

  26. Williams, B. B. et al. Discovery and characterization of gut microbiota decarboxylases that can produce the neurotransmitter tryptamine. Cell Host Microbe 16, 495–503 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Zhang, Y. et al. Discovery of bioactive microbial gene products in inflammatory bowel disease. Nature 606, 754–760 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Li, J. et al. An integrated catalog of reference genes in the human gut microbiome. Nat. Biotechnol. 32, 834–841 (2014).

    CAS  PubMed  Google Scholar 

  29. Grimsby, J. et al. Increased stress response and beta-phenylethylamine in MAOB-deficient mice. Nat. Genet. 17, 206–210 (1997).

    CAS  PubMed  Google Scholar 

  30. Uhlén, M. et al. Tissue-based map of the human proteome. Science 347, 1260419 (2015).

    PubMed  Google Scholar 

  31. Wang, M. et al. Characterization of gene expression profiles in HBV-related liver fibrosis patients and identification of ITGBL1 as a key regulator of fibrogenesis. Sci. Rep. 7, 43446 (2017).

    PubMed  PubMed Central  Google Scholar 

  32. Ahrens, M. et al. DNA methylation analysis in nonalcoholic fatty liver disease suggests distinct disease-specific and remodeling signatures after bariatric surgery. Cell Metab. 18, 296–302 (2013).

    CAS  PubMed  Google Scholar 

  33. Zemtsova, I. et al. Microglia activation in hepatic encephalopathy in rats and humans. Hepatology 54, 204–215 (2011).

    CAS  PubMed  Google Scholar 

  34. Butterworth, R. F. The liver–brain axis in liver failure: neuroinflammation and encephalopathy. Nat. Rev. Gastroenterol. Hepatol. 10, 522–528 (2013).

    CAS  PubMed  Google Scholar 

  35. Ishihara, A. et al. Probing the role of tryptophan-derived secondary metabolism in defense responses against Bipolaris oryzae infection in rice leaves by a suicide substrate of tryptophan decarboxylase. Phytochemistry 72, 7–13 (2011).

    CAS  PubMed  Google Scholar 

  36. Görg, B., Bidmon, H. J. & Häussinger, D. Gene expression profiling in the cerebral cortex of patients with cirrhosis with and without hepatic encephalopathy. Hepatology 57, 2436–2447 (2013).

    PubMed  Google Scholar 

  37. Alexander, A. S. & Nitz, D. A. Retrosplenial cortex maps the conjunction of internal and external spaces. Nat. Neurosci. 18, 1143–1151 (2015).

    CAS  PubMed  Google Scholar 

  38. Tripathi, D. et al. Transjugular intrahepatic portosystemic stent–shunt in the management of portal hypertension. Gut 69, 1173–1192 (2020).

    PubMed  Google Scholar 

  39. Jiang, H. Y. et al. Altered gut microbiota profile in patients with generalized anxiety disorder. J. Psychiatr. Res. 104, 130–136 (2018).

    PubMed  Google Scholar 

  40. Dong, L. et al. Gut microbial characteristics of adult patients with epilepsy. Front. Neurosci. 16, 803538 (2022).

    PubMed  PubMed Central  Google Scholar 

  41. Han, Y. et al. A multi-omics analysis for the prediction of neurocognitive disorders risk among the elderly in Macao. Clin. Transl. Med. 12, e909 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Fischer, J. E. & Baldessarini, R. J. False neurotransmitters and hepatic failure. Lancet 2, 75–80 (1971).

    CAS  PubMed  Google Scholar 

  43. Jalan, R. & Rose, C. F. Heretical thoughts into hepatic encephalopathy. J. Hepatol. 77, 539–548 (2022).

    PubMed  Google Scholar 

  44. Burkhard, P. R., Delavelle, J., Du Pasquier, R. & Spahr, L. Chronic parkinsonism associated with cirrhosis: a distinct subset of acquired hepatocerebral degeneration. Arch. Neurol. 60, 521–528 (2003).

    PubMed  Google Scholar 

  45. Tryc, A. B. et al. Cirrhosis-related Parkinsonism: prevalence, mechanisms and response to treatments. J. Hepatol. 58, 698–705 (2013).

    CAS  PubMed  Google Scholar 

  46. Klos, K. J. et al. Neurologic spectrum of chronic liver failure and basal ganglia T1 hyperintensity on magnetic resonance imaging: probable manganese neurotoxicity. Arch. Neurol. 62, 1385–1390 (2005).

    PubMed  Google Scholar 

  47. Chen, H. et al. A forward chemical genetic screen reveals gut microbiota metabolites that modulate host physiology. Cell 177, 1217–1231.e18 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Weissenborn, K., Berding, G. & Köstler, H. Altered striatal dopamine D2 receptor density and dopamine transport in a patient with hepatic encephalopathy. Metab. Brain Dis. 15, 173–178 (2000).

    CAS  PubMed  Google Scholar 

  49. Yukawa-Muto, Y. et al. Distinct responsiveness to rifaximin in patients with hepatic encephalopathy depends on functional gut microbial species. Hepatol. Commun. 6, 2090–2104 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Dubinkina, V. B. et al. Metagenomic analysis of taxonomic and functional changes in gut microbiota of patients with alcoholic dependence syndrome. Biomed. Khim. 61, 742–749 (2015).

    CAS  PubMed  Google Scholar 

  51. Jiao, M. et al. Alcohol-related elevation of liver transaminase is associated with gut microbiota in male. Front. Med. 9, 823898 (2022).

    Google Scholar 

  52. Assimakopoulos, S. F. et al. Intestinal mucosal proliferation, apoptosis and oxidative stress in patients with liver cirrhosis. Ann. Hepatol. 12, 301–307 (2013).

    PubMed  Google Scholar 

  53. Sang, C. et al. Aile acid profiles are distinct among patients with different etiologies of chronic liver disease. J. Proteome Res. 20, 2340–2351 (2021).

    CAS  PubMed  Google Scholar 

  54. Muthiah, M. D. et al. Development of alcohol-associated hepatitis is associated with specific changes in gut-modified bile acids. Hepatol. Commun. 6, 1073–1089 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Sorribas, M. et al. FXR modulates the gut–vascular barrier by regulating the entry sites for bacterial translocation in experimental cirrhosis. J. Hepatol. 71, 1126–1140 (2019).

    CAS  PubMed  Google Scholar 

  56. Hall, A. B. et al. A novel Ruminococcus gnavus clade enriched in inflammatory bowel disease patients. Genome Med. 9, 103 (2017).

    PubMed  PubMed Central  Google Scholar 

  57. Chua, H. H. et al. Antagonism between gut Ruminococcus gnavus and Akkermansia muciniphila modulates the progression of chronic hepatitis B. Cell. Mol. Gastroenterol. Hepatol. 17, 361–381 (2024).

    CAS  PubMed  Google Scholar 

  58. Bell, A. et al. Elucidation of a sialic acid metabolism pathway in mucus-foraging Ruminococcus gnavus unravels mechanisms of bacterial adaptation to the gut. Nat. Microbiol. 4, 2393–2404 (2019).

    PubMed  PubMed Central  Google Scholar 

  59. Farooqui, T. & Farooqui, A. A. Trace Amines and Their Relevance to Neurological Disorders: A Commentary 257–268 (Academic Press, 2016).

  60. Xu, Z. et al. Ligand recognition and G-protein coupling of trace amine receptor TAAR1. Nature 624, 672–681 (2023).

    CAS  PubMed  Google Scholar 

  61. Rahimi, R. & Rockey, D. C. Hepatic encephalopathy: pharmacological therapies targeting ammonia. Semin. Liver Dis. 36, 48–55 (2016).

    CAS  PubMed  Google Scholar 

  62. McMillin, M. et al. Gli1 activation and protection against hepatic encephalopathy is suppressed by circulating transforming growth factor β1 in mice. J. Hepatol. 61, 1260–1266 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Rath, S., Heidrich, B., Pieper, D. H. & Vital, M. Uncovering the trimethylamine-producing bacteria of the human gut microbiota. Microbiome 5, 54 (2017).

    PubMed  PubMed Central  Google Scholar 

  64. Allen, W. E., Blosser, T. R., Sullivan, Z. A., Dulac, C. & Zhuang, X. Molecular and spatial signatures of mouse brain aging at single-cell resolution. Cell 186, 194–208 (2023).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thanks D. C. Rockey for the useful discussions. This work was supported by the following fundings: National Key Research and Development Program of China (2022YFA0806400, H.Z.), National Science Fund for Distinguished Young Scholars of China (81925026, H.Z.), National Natural Science Foundation of China (82272387, J.G.; 31900101, J.G.; 82130068, H.Z.; 32170111, X.H.; 82372305, X.H.), Guangzhou Key Research Program on Brain Science (202206060001, H.Z.), Natural Science Foundation of Guangdong Province of China (2022A1515010402, X.H.) and Tianjin Science and Technology Plan Project (19ZXDBSY00030, X. Qi).

Author information

Authors and Affiliations

Authors

Contributions

H.Z., J.G., X.H., X. Qi, J.C., Y.H. and E.E.-O. conceived the study and interpreted the data. J.G., H.Z., X. Qi, J.C., X.H., M.H. and Y.X. wrote and revised the paper. H.Z., J.G., X.H. and X. Qi were involved in the funding acquisition. J.G., W.L., Y.X. and Y. Wang performed and interpreted the metagenomic analysis, GBM construction, 16S rRNA sequencing and transcriptome analysis. X. Qi, J.C., H.X., H.W., M.S., S.L., Y.P., R.S., J.Y., X. Quan, Y. Wei, J.G. and C.H. contributed to participant recruitment, sample collection and biobank management. X.H., M.H., Y.X., Y. Wang, Q.X., S.X., L.W. and Z.H. were involved in the construction of the cirrhotic animal model, R. gnavus colonization, FMT experiment, and PDCI and PEA antibody treatment. X.H., M.H., Y.X., Y. Wang, L.W. and Y. Wan performed and interpreted the behavioral experiment, MAO-B activity detection and immunofluorescence analysis. Y.T., F.L., X.H., M.H. and Y.X. were involved in the genetic manipulation. F.X., P.C., M.H., Y.X., J.G. and L.W. performed the HPLC–MS/MS analysis. T.J., M.H., Y.X. and X.H. performed the screening of the PDCI. E.E.-O. and Y.H. provided critical comments on the paper. All authors discussed and approved the paper.

Corresponding authors

Correspondence to Jinjun Chen, Xiaolong Qi, Jie Gao or Hongwei Zhou.

Ethics declarations

Competing interests

J.G., X.H. and H.Z. are listed as inventors on two patents concerning the use of R. gnavus as a therapeutic or diagnostic target for hepatic encephalopathy.

Peer review

Peer review information

Nature Medicine thanks Patricia Bloom and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editors: Alison Farrell and Joao Monteiro, 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 Systematic screening for possible AADCs.

(a) The detailed process for systematic identification of AADCs. (b) Sequence similarity network (SSN) clusters of 329 sequences from IGC with pyridoxal_deC domains in combination with 107 known decarboxylases. Sequences in clusters 2, 5, 6, 7, and 14 (highlighted in red) were chosen for further analysis. (c) Phylogenetic tree analysis of 97 sequences from clusters 2, 5, 6, 7 and 14 in b. Tips colors indicate enzyme types inferred from known sequences. The text numbers represent the CD-hit cluster number of each sequence, with the 39 sequences picked for validation marked by an asterisk; the inner circle shows the SSN cluster of the corresponding sequence; the middle circle indicates the taxonomy of each sequence at the phylum level; the outermost circle shows the conserved domain organization of each sequence.

Extended Data Fig. 2 Colonization of R. gnavus has no significant effect on normal GF mice.

(a) The change of body weight in mice. n = 8 mice per group. Data are presented as mean ± SD. (b-f) Representative image of H&E staining in colon (b), lung (c), liver (d), spleen (e) and kidney (f); Scale bar, 100 μm. (g) Representative image of H&E staining in six brain regions: cortex, hippocampus, thalamus, striatum, amygdala and hypothalamus. The scale bars in the cortex, thalamus, striatum, amygdala and hypothalamus were 100 μm; in the hippocampus it was 400 μm; in the half brain it was 1000 μm.

Source data

Extended Data Fig. 3 The distribution of astrocytes and microglia in 6 brain regions of R. gnavus-colonized cirrhotic mice.

(a) Left panel: representative images of astrocyte (GFAP, red) in brain coronal slice. Middle panel: each white dot in brain section represents one GFAP-positive astrocyte. Right panel: brain section shown the distribution of GFAP-positive astrocytes in 6 brain regions, cyan: cortex; white: hippocampus; green: thalamus; blue: hypothalamus; red: striatum; yellow: amygdala. Scale bars in whole brain section were 1000 µm, in local region were 100 µm. Bar graph showing the density of astrocytes in 6 brain regions. The six brain regions were illustrated below, cortex (CTX); hippocampus (HIP); thalamus (THA); hypothalamus (HYP); striatum (ST); amygdala (AMY). (b) Left panel: representative images of microglia (Iba-1, green) in brain coronal section. Middle panel: each white dot in brain section represents one Iba-1-positive microglial cell. Right panel: the distribution of Iba-1-positive microglial cells in 6 brain regions, cyan: cortex; white: hippocampus; green: thalamus; blue: hypothalamus; red: striatum; yellow: amygdala. Scale bars in whole brain section were 1000 µm, in local region were 100 µm. Bar graph showing the density of microglia in 6 brain regions. Each dot represents a single individual, n = 3 in Con and n = 4 in Rg group. Data are presented as mean ± SD and analyzed by two-sided Mann-Whitney U test (a, b).

Source data

Extended Data Fig. 4 PDC plays a critical role in mediating neurotoxic effects of R. gnavus in cirrhotic mice.

(a) Experimental scheme. (b) The colonization rate of wild-type R. gnavus (Rg-WT) and a RGna_RS09085 insert mutant of R. gnavus (Rg-Mu). (c) Quantification of PEA in the brain. (d) Left panel: schematic of the open field apparatus and central region. Right panel: heatmap of overall activity comparison between the two groups. (e) The ratio of distance traveled in the center to total distance. (f) Left panel: schematic of the water maze apparatus and platform position, the red dot depicts the location of the mouse at the start of the recording. Right panel: heatmap of overall activity comparison between the two groups. (g) Time spent in the platform quadrant in the probe test. (h) The percentage of mice without coma. (i) Left panel: representative images of immunofluorescent labelling of GFAP (astrocyte, red) in the cerebral cortex of each treatment, scale bars, 20 µm. Representative astrocytes were 3D reconstructed (scale bars, 8 µm) and color-coded to indicate the spectrum of volumes (scale bars, 20 µm). Right panel: astrocyte morphological parameters of relative GFAP fluorescence intensity, volume, and number of branch points. (j) Left panel: representative images of immunofluorescent labelling of Iba-1 (microglia, green) in the cerebral cortex of each treatment, scale bars, 20 µm. Representative microglia were 3D reconstructed and their convex hulls (grey shadow) were illustrated, scale bars, 8 µm. Somas and branches were color-coded to indicate the spectrum of soma volumes and branches length, scale bars, 20 µm. Right panel: microglia morphological parameters of the ratio of soma volume/convex hull volume, total branch length, and number of intersections. Each dot represents a single individual, n = 11 mice per group (a-j). Data are presented as mean ± SD. Statistical analysis: two-sided Student’s t-test (b); one-way ANOVA followed by two-sided Dunnett's post hoc test (c, e, g, i, j); two-sided Log-rank (Mantel-Cox) test (h). Panel a created with BioRender.com.

Source data

Extended Data Fig. 5 High-throughput screening and in vitro evaluating of PDCI.

(a) Schematic diagram illustrating the study design for the high-throughput screening and evaluation of PDCI. (b) The OD600 of R. gnavus (Rg) growth in the PYG broth supplemented with 100 μM of candidate inhibitors. n = 3 biological replicates per group. (c) The relative PEA levels in the culture supernatant of Rg supplemented with 10 μM of candidate inhibitors. n = 3 biological replicates per group. (d) The relative PEA levels in the culture supernatant of Rg supplement with 100 μM of candidate inhibitors. n = 3 biological replicates per group. Data are presented as mean ± SD and analyzed by one-way ANOVA followed by two-sided Dunnett's post hoc test (c, d). Panel a created with BioRender.com.

Source data

Extended Data Fig. 6 PEA is required for the neurotoxic effects of R. gnavus.

(a) Experimental scheme. PEA-mAB: PEA monoclonal antibody. (b) Quantification of PEA in the brain. (c) Left panel: schematic of the open field apparatus and central region. Right panel: heatmap of overall activity comparison between the three groups. (d) The ratio of distance traveled in the center to total distance. (e) Left panel: schematic of the water maze apparatus and platform position, the red dot depicts the location of the mouse at the start of the recording. Right panel: heatmap of overall activity comparison between the three groups. (f) Time spent in the platform quadrant in the probe test. (g) The percentage of mice without coma. (h) Left panel: representative images of immunofluorescent labelling of GFAP (astrocyte, red) in the cerebral cortex of each treatment, scale bars, 20 µm. Representative astrocytes were 3D reconstructed (scale bars, 8 µm) and color-coded to indicate the spectrum of volumes (scale bars, 20 µm). Right panel: astrocyte morphological parameters of relative GFAP fluorescence intensity, volume, and number of branch points. (i) Left panel: representative images of immunofluorescent labelling of Iba-1 (microglia, green) in the cerebral cortex of each treatment, scale bars, 20 µm. Representative microglia were 3D reconstructed and their convex hulls (grey shadow) were illustrated, scale bars, 8 µm. Somas and branches were color-coded to indicate the spectrum of soma volumes and branches length, scale bars, 20 µm. Right panel: microglia morphological parameters of the ratio of soma volume/convex hull volume, total branch length, and number of intersections. Each dot represents a single individual, n = 12 mice per group (b-i). Data represent 2 independent experiments and are presented as mean ± SD. Statistical analysis: one-way ANOVA followed by two-sided Dunnett's post hoc test (b, d, f, h, i); two-sided Log-rank (Mantel-Cox) test (g). Panel a created with BioRender.com.

Source data

Extended Data Fig. 7 PEA triggers a HE-like syndromes in cirrhotic mice.

(a) Experimental scheme. (b) Quantification of PEA in the brain of healthy or cirrhotic mice receiving the indicated dose of PEA (n = 5 in cirrhosis control, non-cirrhosis control and non-cirrhosis + 20 μg/g PEA groups; n = 6 in cirrhosis + 1, 10, 20 μg/g PEA groups). (c) Left panel: schematic of the open field apparatus and central region. Right panel: heatmap of overall activity in the open field apparatus. (d) The ratio of distance traveled in the center to total distance. (e) Left panel: schematic of the water maze apparatus and platform position, the red dot depicts the location of the mouse at the start of the recording. Right panel: heatmap of overall activity. (f) Time spent in the platform quadrant in the probe test. (g) All the mice were intraperitoneally injected with 40% CCl4 to induced severe liver damage, line graph shows the percentage of mice without coma. (h) Upper panel: representative images of immunofluorescent labelling of GFAP (astrocyte, red) in the cerebral cortex of each treatment, scale bars, 20 µm. Representative astrocytes were 3D reconstructed (scale bars, 8 µm) and color-coded to indicate the spectrum of volumes (scale bars, 20 µm). Lower panel: astrocyte morphological parameters of relative GFAP fluorescence intensity, volume, and number of branch points. (i) Upper panel: representative images of immunofluorescent labelling of Iba-1 (microglia, green) in the cerebral cortex of each treatment, scale bars, 20 µm. Representative microglia were 3D reconstructed and their convex hulls (grey shadow) were illustrated, scale bars, 8 µm. Somas and branches were color-coded to indicate the spectrum of soma volumes and branches length, scale bars, 20 µm. Lower panel: microglia morphological parameters of the ratio of soma volume/convex hull volume, total branch length, and number of intersections. Each dot represents a single individual, n = 12 mice per group (c-i). Data represent 2 independent experiments and are presented as mean ± SD. Statistical analysis: two-sided Student’s t-test (d, f, h, i); two-sided Log-rank (Mantel-Cox) test (g). Panel a created with BioRender.com.

Source data

Extended Data Fig. 8 Fecal microbiota from HE patient induces neurobehavioral and neuropathological changes in cirrhotic mice.

(a) Germ-free (GF) mice were colonized with fecal microbiota from 3 healthy control (HC) and 3 HE patients (HE). Each donor was transferred to 3-5 recipient mice. GF mice gavaged with saline were used as pure controls. (b) Relative PDC levels in fecal samples from both donors and recipient mice. (c) Quantification of PEA levels in the brains of recipient mice (n = 8 mice per group). (d) Heatmap of overall activity comparison between three groups in open field test. (e) The ratio of distance traveled in the center to total distance. (f) Heatmap of overall activity comparison between three groups in water maze test. (g) Time spent in the platform quadrant in the probe test. (h) Mediation analysis suggested that HE fecal microbiota induce memory deficits through R. gnavus. (i) Left panel: representative images of immunofluorescent labelling of GFAP (astrocyte, red) in the cerebral cortex of each treatment, scale bars, 20 µm. Representative astrocytes were 3D reconstructed (scale bars, 8 µm) and color-coded to indicate the spectrum of volumes (scale bars, 20 µm). Right panel: astrocyte morphological parameters of relative GFAP fluorescence intensity, volume, and number of branch points. (j) Left panel: representative images of immunofluorescent labelling of Iba-1 (microglia, green) in the cerebral cortex of each treatment, scale bars, 20 µm. Representative microglia were 3D reconstructed and their convex hulls (grey shadow) were illustrated, scale bars, 8 µm. Somas and branches were color-coded to indicate the spectrum of soma volumes and branch length, scale bars, 20 µm. Right panel: microglia morphological parameters of the ratio of soma volume/convex hull volume, total branch length, and number of intersections. Each dot represents a single individual, n = 12 in GF and HC, n = 14 in HE (b, d-j). Data are presented as mean ± SD. Statistical analysis: two-sides Student’s t test (b); Mediation analysis using R package ‘mediation’ (h); one-way ANOVA followed by two-sides Tukey’s post hoc test (c, e, g, i, j). Panel a created with BioRender.com.

Source data

Extended Data Fig. 9 Engraftment fidelity for colonization of mice with human microbiota.

(a-b) α-diversity as measured by the Shannon index (a) or Chao index (b). (c-d) Taxonomic profile in donors and recipients (HC or HE) at the phylum level. (e-f) Taxa engraftment in mice at the genus level from 16S rRNA gene sequencing. The cumulative relative abundance of shared taxa in human donors (e), as well as the fraction of donor-derived taxa (genus level) engrafted in the mice relative to taxa in human donor (f). (g) The PCoA of weighted UniFrac distances. (h-i) GraPhlAn plot of LEfSe linear discriminant analysis of microbiome profiles up to the genus level from 16S rRNA gene sequencing of HC or HE (h, mice; i, human donors). n = 3, 3, 12, 12 for DonorsHC, DonorsHE, MiceHC, MiceHE separately, data are presented as mean ± SD (a-f).

Extended Data Fig. 10 Targeting PEA alleviates neurotoxicity induced by fecal microbiota from HE patients.

(a) Schematic outline of the fecal microbiota transplantation experiments. (b) The relative PDC levels in each treatment. (c) Quantification of PEA in the brain of the mouse. (d) Left panel: heatmap of overall activity comparison between three groups in the open field apparatus, white frame indicated the central region. Right panel: the ratio of distance traveled in the center to total distance. (e) Left panel: heatmap of overall activity comparison between three groups in the water maze apparatus, black dashed circle indicated the platform. Right panel: time spent in the platform quadrant in the probe test. (f) All the mice were intraperitoneally injected with 40% CCl4 to induce severe liver damage, line graph shows the percentage of mice without coma. (g) Left panel: representative images of immunofluorescent labelling of GFAP (astrocyte, red) in cerebral cortex of each treatment, scale bars, 20 µm. Representative astrocytes were 3D reconstructed (scale bars, 8 µm) and color-coded to indicate the spectrum of volumes (scale bars, 20 µm). Right panel: astrocyte morphological parameters of relative GFAP fluorescence intensity, volume, and number of branch points. (h) Left panel: representative images of immunofluorescent labelling of Iba-1 (microglia, green) in the cerebral cortex of each treatment, scale bars, 20 µm. Representative microglia were 3D reconstructed and their convex hulls (grey shadow) were illustrated, scale bars, 8 µm. Somas and branches were color-coded to indicate the spectrum of soma volumes and branches length, scale bars, 20 µm. Right panel: microglia morphological parameters of the ratio of soma volume/convex hull volume, total branch length, and number of intersections. Each dot represents a single individual, n = 11 mice per group (a-h). Data are presented as mean ± SD. Statistical analysis: one-way ANOVA followed by two-sides Dunnett's post hoc test (b, c, d, e, g, h); two-sides Log-rank (Mantel-Cox) test (f). Panel a created with BioRender.com.

Source data

Supplementary information

Reporting Summary

Supplementary Tables

Supplementary Tables 1–20.

Supplementary Video 1

R. gnavus induces symmetrical tremor of the large joint in cirrhotic mice.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 10

Statistical source data.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) 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

He, X., Hu, M., Xu, Y. et al. The gut–brain axis underlying hepatic encephalopathy in liver cirrhosis. Nat Med 31, 627–638 (2025). https://doi.org/10.1038/s41591-024-03405-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue date:

  • DOI: https://doi.org/10.1038/s41591-024-03405-9

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