ahctf1 and kras mutations combine to amplify oncogenic stress and restrict liver overgrowth in a zebrafish model of hepatocellular carcinoma

  1. Kimberly J Morgan
  2. Karen Doggett
  3. Fansuo Geng
  4. Stephen Mieruszynski
  5. Lachlan Whitehead
  6. Kelly A Smith
  7. Benjamin M Hogan
  8. Cas Simons
  9. Gregory J Baillie
  10. Ramyar Molania
  11. Anthony T Papenfuss
  12. Thomas E Hall
  13. Elke A Ober
  14. Didier YR Stainier
  15. Zhiyuan Gong
  16. Joan Kathleen Heath  Is a corresponding author
  1. Walter and Eliza Hall Institute of Medical Research, Australia
  2. University of Technology Sydney, Australia
  3. University of Melbourne, Australia
  4. Peter MacCallum Cancer Centre, Australia
  5. Murdoch Children's Research Institute, Australia
  6. University of Queensland, Australia
  7. University of Copenhagen, Denmark
  8. Max Planck Institute for Heart and Lung Research, Germany
  9. National University of Singapore, Singapore

Abstract

The nucleoporin (NUP) ELYS, encoded by AHCTF1, is a large multifunctional protein with essential roles in nuclear pore assembly and mitosis. Using both larval and adult zebrafish models of hepatocellular carcinoma (HCC), in which the expression of an inducible mutant kras transgene (krasG12V) drives hepatocyte-specific hyperplasia and liver enlargement, we show that reducing ahctf1 gene dosage by 50% markedly decreases liver volume, while non-hyperplastic tissues are unaffected. We demonstrate that in the context of cancer, ahctf1 heterozygosity impairs nuclear pore formation, mitotic spindle assembly and chromosome segregation, leading to DNA damage and activation of a Tp53-dependent transcriptional program that induces cell death and cell cycle arrest. Heterozygous expression of both ahctf1 and ranbp2 (encoding a second nucleoporin), or treatment of heterozygous ahctf1 larvae with the nucleocytoplasmic transport inhibitor, Selinexor, completely blocks krasG12V-driven hepatocyte hyperplasia. Gene expression analysis of patient samples in the Liver hepatocellular carcinoma (LIHC) dataset in The Cancer Genome Atlas shows that high expression of one or more of the transcripts encoding the ten components of the NUP107-160 sub-complex, which includes AHCTF1, is positively correlated with worse overall survival. These results provide a strong and feasible rationale for the development of novel cancer therapeutics that target ELYS function and suggest potential avenues for effective combinatorial treatments.

Data availability

A new RNA sequencing dataset has been deposited in GEO under accession ID GSE220282. Existing datasets analysed during the current study are available in the cBioPortal Cancer Genomics database (http://www.cbioportal.org). All data generated/analysed during this study are included in the Figures and figure supplements and Source Data files are provided for Figures 1-7.

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

Article and author information

Author details

  1. Kimberly J Morgan

    Epigenetic and Development Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
    Competing interests
    No competing interests declared.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0001-7871-2583
  2. Karen Doggett

    Epigenetic and Development Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
    Competing interests
    No competing interests declared.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-8676-8461
  3. Fansuo Geng

    University of Technology Sydney, Sydney, Australia
    Competing interests
    No competing interests declared.
  4. Stephen Mieruszynski

    Epigenetics and Development Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
    Competing interests
    No competing interests declared.
  5. Lachlan Whitehead

    Centre for Dynamic Imaging, Advanced Technology and Biology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
    Competing interests
    No competing interests declared.
  6. Kelly A Smith

    Department of Physiology, University of Melbourne, Parkville, Australia
    Competing interests
    No competing interests declared.
  7. Benjamin M Hogan

    Program in Organogenesis and Cancer, Peter MacCallum Cancer Centre, Melbourne, Australia
    Competing interests
    No competing interests declared.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-0651-7065
  8. Cas Simons

    Murdoch Children's Research Institute, Parkville, Australia
    Competing interests
    No competing interests declared.
  9. Gregory J Baillie

    Institute for Molecular Biosciences, University of Queensland, Brisbane, Australia
    Competing interests
    No competing interests declared.
  10. Ramyar Molania

    Bioinformatics Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
    Competing interests
    No competing interests declared.
  11. Anthony T Papenfuss

    Bioinformatics Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
    Competing interests
    No competing interests declared.
  12. Thomas E Hall

    Institute for Molecular Biosciences, University of Queensland, Brisbane, Australia
    Competing interests
    No competing interests declared.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-7718-7614
  13. Elke A Ober

    Danish Stem Cell Center, University of Copenhagen, Copenhagen, Denmark
    Competing interests
    Elke A Ober, Reviewing editor, eLife.
  14. Didier YR Stainier

    Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
    Competing interests
    Didier YR Stainier, Senior editor, eLife.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-0382-0026
  15. Zhiyuan Gong

    Department of Biological Science, National University of Singapore, Singapore, Singapore
    Competing interests
    No competing interests declared.
  16. Joan Kathleen Heath

    Epigenetics and Development Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
    For correspondence
    joan.heath@wehi.edu.au
    Competing interests
    No competing interests declared.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0001-6955-232X

Funding

National Health and Medical Research Council (Project GNT 1024878)

  • Joan Kathleen Heath

Australian Government (Graduate Student Training Program)

  • Kimberly J Morgan

Ludwig Institute for Cancer Research (Ludwig Member Support Package to Joan Heath)

  • Joan Kathleen Heath

The funders had no role in study design, data collection and interpretation, or the decision to submit the work for publication.

Reviewing Editor

  1. Hao Zhu, University of Texas Southwestern Medical Center, United States

Ethics

Animal experimentation: All husbandry and experimental procedures performed on zebrafish followed standard operating procedures and were conducted with the approval of the Animal Ethics Committees of the Walter and Eliza Hall Institute and The University of Melbourne, Parkville, Victoria, Australia. WEHI-AEC approved project 2019.014, project title: Zebrafish disease models and mechanisms.

Version history

  1. Preprint posted: August 27, 2021 (view preprint)
  2. Received: August 27, 2021
  3. Accepted: January 16, 2023
  4. Accepted Manuscript published: January 17, 2023 (version 1)
  5. Version of Record published: February 3, 2023 (version 2)

Copyright

© 2023, Morgan 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

  • 1,642
    views
  • 166
    downloads
  • 2
    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. Kimberly J Morgan
  2. Karen Doggett
  3. Fansuo Geng
  4. Stephen Mieruszynski
  5. Lachlan Whitehead
  6. Kelly A Smith
  7. Benjamin M Hogan
  8. Cas Simons
  9. Gregory J Baillie
  10. Ramyar Molania
  11. Anthony T Papenfuss
  12. Thomas E Hall
  13. Elke A Ober
  14. Didier YR Stainier
  15. Zhiyuan Gong
  16. Joan Kathleen Heath
(2023)
ahctf1 and kras mutations combine to amplify oncogenic stress and restrict liver overgrowth in a zebrafish model of hepatocellular carcinoma
eLife 12:e73407.
https://doi.org/10.7554/eLife.73407

Share this article

https://doi.org/10.7554/eLife.73407

Further reading

    1. Cancer Biology
    2. Immunology and Inflammation
    Nicholas J Mullen, Surendra K Shukla ... Pankaj K Singh
    Research Article

    Pyrimidine nucleotide biosynthesis is a druggable metabolic dependency of cancer cells, and chemotherapy agents targeting pyrimidine metabolism are the backbone of treatment for many cancers. Dihydroorotate dehydrogenase (DHODH) is an essential enzyme in the de novo pyrimidine biosynthesis pathway that can be targeted by clinically approved inhibitors. However, despite robust preclinical anticancer efficacy, DHODH inhibitors have shown limited single-agent activity in phase 1 and 2 clinical trials. Therefore, novel combination therapy strategies are necessary to realize the potential of these drugs. To search for therapeutic vulnerabilities induced by DHODH inhibition, we examined gene expression changes in cancer cells treated with the potent and selective DHODH inhibitor brequinar (BQ). This revealed that BQ treatment causes upregulation of antigen presentation pathway genes and cell surface MHC class I expression. Mechanistic studies showed that this effect is (1) strictly dependent on pyrimidine nucleotide depletion, (2) independent of canonical antigen presentation pathway transcriptional regulators, and (3) mediated by RNA polymerase II elongation control by positive transcription elongation factor B (P-TEFb). Furthermore, BQ showed impressive single-agent efficacy in the immunocompetent B16F10 melanoma model, and combination treatment with BQ and dual immune checkpoint blockade (anti-CTLA-4 plus anti-PD-1) significantly prolonged mouse survival compared to either therapy alone. Our results have important implications for the clinical development of DHODH inhibitors and provide a rationale for combination therapy with BQ and immune checkpoint blockade.

    1. Cancer Biology
    2. Cell Biology
    Savvas Nikolaou, Amelie Juin ... Laura M Machesky
    Research Article Updated

    Pancreatic ductal adenocarcinoma carries a dismal prognosis, with high rates of metastasis and few treatment options. Hyperactivation of KRAS in almost all tumours drives RAC1 activation, conferring enhanced migratory and proliferative capacity as well as macropinocytosis. Macropinocytosis is well understood as a nutrient scavenging mechanism, but little is known about its functions in trafficking of signalling receptors. We find that CYRI-B is highly expressed in pancreatic tumours in a mouse model of KRAS and p53-driven pancreatic cancer. Deletion of Cyrib (the gene encoding CYRI-B protein) accelerates tumourigenesis, leading to enhanced ERK and JNK-induced proliferation in precancerous lesions, indicating a potential role as a buffer of RAC1 hyperactivation in early stages. However, as disease progresses, loss of CYRI-B inhibits metastasis. CYRI-B depleted tumour cells show reduced chemotactic responses to lysophosphatidic acid, a major driver of tumour spread, due to impaired macropinocytic uptake of the lysophosphatidic acid receptor 1. Overall, we implicate CYRI-B as a mediator of growth and signalling in pancreatic cancer, providing new insights into pathways controlling metastasis.