Hepatic lipid overload triggers biliary epithelial cell activation via E2Fs

  1. Ece Yildiz
  2. Gaby El Alam
  3. Alessia Perino
  4. Antoine Jalil
  5. Pierre-Damien Denechaud
  6. Katharina Huber
  7. Lluis Fajas
  8. Johan Auwerx
  9. Giovanni Sorrentino
  10. Kristina Schoonjans  Is a corresponding author
  1. École Polytechnique Fédérale de Lausanne, Switzerland
  2. Inserm UMR1048, France
  3. University of Lausanne, Switzerland
  4. University of Trieste, Italy

Abstract

During severe or chronic hepatic injury, biliary epithelial cells (BECs) undergo rapid activation into proliferating progenitors, a crucial step required to establish a regenerative process known as ductular reaction (DR). While DR is a hallmark of chronic liver diseases, including advanced stages of non-alcoholic fatty liver disease (NAFLD), the early events underlying BEC activation are largely unknown. Here, we demonstrate that BECs readily accumulate lipids during high-fat diet feeding in mice and upon fatty acid treatment in BEC-derived organoids. Lipid overload induces metabolic rewiring to support the conversion of adult cholangiocytes into reactive BECs. Mechanistically, we found that lipid overload activates the E2F transcription factors in BECs, which drives cell cycle progression while promoting glycolytic metabolism. These findings demonstrate that fat overload is sufficient to reprogram BECs into progenitor cells in the early stages of NAFLD and provide new insights into the mechanistic basis of this process, revealing unexpected connections between lipid metabolism, stemness, and regeneration.

Data availability

Computational analysis was performed using established packages mentioned in previous sections, and no new code was generated. RNA-Seq data have been deposited in GEO under accession code GSE217739. Two publicly available RNA-Seq datasets of mouse BECs with accession numbers GSE123133 (Aloia et al., 2019) and GSE125688 (Pepe-Mooney et al., 2019) were downloaded from the GEO and used for GSEA and over-representation enrichment analysis as mentioned previously. Source code is available at https://github.com/auwerxlab/Yildiz_eLife_2023.

The following data sets were generated
The following previously published data sets were used

Article and author information

Author details

  1. Ece Yildiz

    Laboratory of Metabolic Signaling, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
    Competing interests
    The authors declare that no competing interests exist.
  2. Gaby El Alam

    Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
    Competing interests
    The authors declare that no competing interests exist.
  3. Alessia Perino

    Laboratory of Metabolic Signaling, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
    Competing interests
    The authors declare that no competing interests exist.
  4. Antoine Jalil

    Laboratory of Metabolic Signaling, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
    Competing interests
    The authors declare that no competing interests exist.
  5. Pierre-Damien Denechaud

    Institute of Metabolic and Cardiovascular Diseases, Inserm UMR1048, Toulouse, France
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0003-3502-4814
  6. Katharina Huber

    Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
    Competing interests
    The authors declare that no competing interests exist.
  7. Lluis Fajas

    Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-1283-9503
  8. Johan Auwerx

    Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
    Competing interests
    The authors declare that no competing interests exist.
  9. Giovanni Sorrentino

    Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
    Competing interests
    The authors declare that no competing interests exist.
  10. Kristina Schoonjans

    Laboratory of Metabolic Signaling, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
    For correspondence
    kristina.schoonjans@epfl.ch
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0003-1247-4265

Funding

École Polytechnique Fédérale de Lausanne

  • Kristina Schoonjans

Kristian Gerhard Jebsen Foundation

  • Kristina Schoonjans

Swiss National Science Foundation (310030_189178)

  • Kristina Schoonjans

Swiss National Science Foundation (31003A_179435)

  • Johan Auwerx

AIRC Start-Up 2020 (24322)

  • Giovanni Sorrentino

The authors declare no competing interests.

Reviewing Editor

  1. Peter Tontonoz, University of California, Los Angeles, United States

Ethics

Animal experimentation: All the animal experiments were authorized by the Veterinary Office of the Canton of Vaud, Switzerland, under license authorizations VD3721 and VD2627.b.

Version history

  1. Received: July 16, 2022
  2. Preprint posted: July 28, 2022 (view preprint)
  3. Accepted: March 3, 2023
  4. Accepted Manuscript published: March 6, 2023 (version 1)
  5. Version of Record published: March 21, 2023 (version 2)

Copyright

© 2023, Yildiz et al.

This article is distributed under the terms of the Creative Commons Attribution License permitting unrestricted use and redistribution provided that the original author and source are credited.

Metrics

  • 4,152
    views
  • 238
    downloads
  • 0
    citations

Views, downloads and citations are aggregated across all versions of this paper published by eLife.

Download links

A two-part list of links to download the article, or parts of the article, in various formats.

Downloads (link to download the article as PDF)

Open citations (links to open the citations from this article in various online reference manager services)

Cite this article (links to download the citations from this article in formats compatible with various reference manager tools)

  1. Ece Yildiz
  2. Gaby El Alam
  3. Alessia Perino
  4. Antoine Jalil
  5. Pierre-Damien Denechaud
  6. Katharina Huber
  7. Lluis Fajas
  8. Johan Auwerx
  9. Giovanni Sorrentino
  10. Kristina Schoonjans
(2023)
Hepatic lipid overload triggers biliary epithelial cell activation via E2Fs
eLife 12:e81926.
https://doi.org/10.7554/eLife.81926

Share this article

https://doi.org/10.7554/eLife.81926

Further reading

    1. Cell Biology
    2. Stem Cells and Regenerative Medicine
    Rajdeep Banerjee, Thomas J Meyer ... David D Roberts
    Research Article

    Extramedullary erythropoiesis is not expected in healthy adult mice, but erythropoietic gene expression was elevated in lineage-depleted spleen cells from Cd47−/− mice. Expression of several genes associated with early stages of erythropoiesis was elevated in mice lacking CD47 or its signaling ligand thrombospondin-1, consistent with previous evidence that this signaling pathway inhibits expression of multipotent stem cell transcription factors in spleen. In contrast, cells expressing markers of committed erythroid progenitors were more abundant in Cd47−/− spleens but significantly depleted in Thbs1−/− spleens. Single-cell transcriptome and flow cytometry analyses indicated that loss of CD47 is associated with accumulation and increased proliferation in spleen of Ter119CD34+ progenitors and Ter119+CD34 committed erythroid progenitors with elevated mRNA expression of Kit, Ermap, and Tfrc. Induction of committed erythroid precursors is consistent with the known function of CD47 to limit the phagocytic removal of aged erythrocytes. Conversely, loss of thrombospondin-1 delays the turnover of aged red blood cells, which may account for the suppression of committed erythroid precursors in Thbs1−/− spleens relative to basal levels in wild-type mice. In addition to defining a role for CD47 to limit extramedullary erythropoiesis, these studies reveal a thrombospondin-1-dependent basal level of extramedullary erythropoiesis in adult mouse spleen.

    1. Developmental Biology
    2. Stem Cells and Regenerative Medicine
    Mayank Verma, Yoko Asakura ... Atsushi Asakura
    Research Article Updated

    Endothelial and skeletal muscle lineages arise from common embryonic progenitors. Despite their shared developmental origin, adult endothelial cells (ECs) and muscle stem cells (MuSCs; satellite cells) have been thought to possess distinct gene signatures and signaling pathways. Here, we shift this paradigm by uncovering how adult MuSC behavior is affected by the expression of a subset of EC transcripts. We used several computational analyses including single-cell RNA-seq (scRNA-seq) to show that MuSCs express low levels of canonical EC markers in mice. We demonstrate that MuSC survival is regulated by one such prototypic endothelial signaling pathway (VEGFA-FLT1). Using pharmacological and genetic gain- and loss-of-function studies, we identify the FLT1-AKT1 axis as the key effector underlying VEGFA-mediated regulation of MuSC survival. All together, our data support that the VEGFA-FLT1-AKT1 pathway promotes MuSC survival during muscle regeneration, and highlights how the minor expression of select transcripts is sufficient for affecting cell behavior.