MLL3 regulates the CDKN2A tumor suppressor locus in liver cancer

  1. Changyu Zhu
  2. Yadira M Soto-Feliciano  Is a corresponding author
  3. John P Morris 4th
  4. Chun-Hao Huang
  5. Richard P Koche
  6. Yu-jui Ho
  7. Ana Banito
  8. Chun-Wei (David) Chen
  9. Aditya Shroff
  10. Sha Tian
  11. Geulah Livshits
  12. Chi-Chao Chen
  13. Myles Fennell
  14. Scott A Armstrong
  15. C David Allis
  16. Darjus F Tschaharganeh  Is a corresponding author
  17. Scott W Lowe  Is a corresponding author
  1. Memorial Sloan Kettering Cancer Center, United States
  2. Massachusetts Institute of Technology, United States
  3. The University of North Carolina at Chapel Hill, United States
  4. Dana-Farber Cancer Institute, United States
  5. The Rockefeller University, United States
  6. German Cancer Research Center, Germany

Abstract

Mutations in genes encoding components of chromatin modifying and remodeling complexes are among the most frequently observed somatic events in human cancers. For example, missense and nonsense mutations targeting the mixed lineage leukemia family member 3 (MLL3, encoded by KMT2C) histone methyltransferase occur in a range of solid tumors, and heterozygous deletions encompassing KMT2C occur in a subset of aggressive leukemias. Although MLL3 loss can promote tumorigenesis in mice, the molecular targets and biological processes by which MLL3 suppresses tumorigenesis remain poorly characterized. Here we combined genetic, epigenomic, and animal modeling approaches to demonstrate that one of the mechanisms by which MLL3 links chromatin remodeling to tumor suppression is by co-activating the Cdkn2a tumor suppressor locus. Disruption of Kmt2c cooperates with Myc overexpression in the development of murine hepatocellular carcinoma (HCC), in which MLL3 binding to the Cdkn2a locus is blunted, resulting in reduced H3K4 methylation and low expression levels of the locus-encoded tumor suppressors p16/Ink4a and p19/Arf. Conversely, elevated KMT2C expression increases its binding to the CDKN2A locus and co-activates gene transcription. Endogenous Kmt2c restoration reverses these chromatin and transcriptional effects and triggers Ink4a/Arf-dependent apoptosis. Underscoring the human relevance of this epistasis, we found that genomic alterations in KMT2C and CDKN2A were associated with similar transcriptional profiles in human HCC samples. These results collectively point to a new mechanism for disrupting CDKN2A activity during cancer development and, in doing so, link MLL3 to an established tumor suppressor network.

Data availability

Source files of all original gels and Western Blots were provided for the following figures:Figure 1-figure supplement 2B;Figure 4-figure supplement 1A, C, D, E;Figure 5-figure supplement 2B, F, G.RNA sequencing and ChIP sequencing data files that support the findings of this study have been deposited in the Gene Expression Omnibus under the accession code GSE85055, as well as in the Dryad digital repository (doi:10.5061/dryad.7pvmcvdwm; doi:10.5061/dryad.f1vhhmh0h).All other data supporting the findings of this study will be made available upon reasonable request to the corresponding authors.

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

Article and author information

Author details

  1. Changyu Zhu

    Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, United States
    Competing interests
    No competing interests declared.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0003-3583-3638
  2. Yadira M Soto-Feliciano

    Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, United States
    For correspondence
    ysoto@mit.edu
    Competing interests
    No competing interests declared.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-8523-7917
  3. John P Morris 4th

    Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, United States
    Competing interests
    No competing interests declared.
  4. Chun-Hao Huang

    Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, United States
    Competing interests
    No competing interests declared.
  5. Richard P Koche

    Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, United States
    Competing interests
    No competing interests declared.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-6820-5083
  6. Yu-jui Ho

    Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, United States
    Competing interests
    No competing interests declared.
  7. Ana Banito

    Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, United States
    Competing interests
    No competing interests declared.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0003-2188-0003
  8. Chun-Wei (David) Chen

    Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, United States
    Competing interests
    No competing interests declared.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-8737-6830
  9. Aditya Shroff

    Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, United States
    Competing interests
    No competing interests declared.
  10. Sha Tian

    Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, United States
    Competing interests
    No competing interests declared.
  11. Geulah Livshits

    Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, United States
    Competing interests
    No competing interests declared.
  12. Chi-Chao Chen

    Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, United States
    Competing interests
    No competing interests declared.
  13. Myles Fennell

    Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, United States
    Competing interests
    No competing interests declared.
  14. Scott A Armstrong

    Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, United States
    Competing interests
    No competing interests declared.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-9099-4728
  15. C David Allis

    Laboratory of Chromatin Biology and Epigenetics, The Rockefeller University, New York, United States
    Competing interests
    C David Allis, is a co founder of Chroma Therapeutics and Constellation Pharmaceuticals and a Scientific Advisory Board member of EpiCypher..
  16. Darjus F Tschaharganeh

    German Cancer Research Center, Heidelberg, Germany
    For correspondence
    d.tschaharganeh@dkfz.de
    Competing interests
    No competing interests declared.
  17. Scott W Lowe

    Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, United States
    For correspondence
    lowes@mskcc.org
    Competing interests
    Scott W Lowe, is an advisor for and has equity in the following biotechnology companies: ORIC Pharmaceuticals, Faeth Therapeutics, Blueprint Medicines, Geras Bio, Mirimus Inc., and PMV Pharmaceuticals. S.W.L. also acknowledges receiving funding and research support from Agilent Technologies and Calico, for the purposes of massively parallel oligo synthesis and single-cell analytics, respectively..
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-5284-9650

Funding

National Cancer Institute (P01 CA013106)

  • Scott W Lowe

National Cancer Institute (R01 CA233944)

  • Scott W Lowe

National Institute of General Medical Sciences (1K99GM140265-01)

  • Yadira M Soto-Feliciano

National Cancer Institute (1F32CA257103)

  • Changyu Zhu

American Cancer Society (PF-14-066-01-TBE)

  • John P Morris 4th

Helmholtz foundation (VH-NG-1114)

  • Darjus F Tschaharganeh

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 animal experiments were approved by the MSKCC Institutional Animal Care and Use Committee (protocol 11-06-011). Animals were monitored for signs of ill-health by veterinary staff at the Research Animal Resource Center (RARC) at MSKCC and efforts were made to minimize suffering.

Version history

  1. Received: June 7, 2022
  2. Preprint posted: June 9, 2022 (view preprint)
  3. Accepted: May 31, 2023
  4. Accepted Manuscript published: June 1, 2023 (version 1)
  5. Version of Record published: June 19, 2023 (version 2)

Copyright

© 2023, Zhu 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

  • 1,355
    views
  • 192
    downloads
  • 5
    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. Changyu Zhu
  2. Yadira M Soto-Feliciano
  3. John P Morris 4th
  4. Chun-Hao Huang
  5. Richard P Koche
  6. Yu-jui Ho
  7. Ana Banito
  8. Chun-Wei (David) Chen
  9. Aditya Shroff
  10. Sha Tian
  11. Geulah Livshits
  12. Chi-Chao Chen
  13. Myles Fennell
  14. Scott A Armstrong
  15. C David Allis
  16. Darjus F Tschaharganeh
  17. Scott W Lowe
(2023)
MLL3 regulates the CDKN2A tumor suppressor locus in liver cancer
eLife 12:e80854.
https://doi.org/10.7554/eLife.80854

Share this article

https://doi.org/10.7554/eLife.80854

Further reading

    1. Cancer Biology
    Célia Guérin, David Tulasne
    Review Article

    Tyrosine kinase inhibitors (TKI) directed against MET have been recently approved to treat advanced non-small cell lung cancer (NSCLC) harbouring activating MET mutations. This success is the consequence of a long characterization of MET mutations in cancers, which we propose to outline in this review. MET, a receptor tyrosine kinase (RTK), displays in a broad panel of cancers many deregulations liable to promote tumour progression. The first MET mutation was discovered in 1997, in hereditary papillary renal cancer (HPRC), providing the first direct link between MET mutations and cancer development. As in other RTKs, these mutations are located in the kinase domain, leading in most cases to ligand-independent MET activation. In 2014, novel MET mutations were identified in several advanced cancers, including lung cancers. These mutations alter splice sites of exon 14, causing in-frame exon 14 skipping and deletion of a regulatory domain. Because these mutations are not located in the kinase domain, they are original and their mode of action has yet to be fully elucidated. Less than five years after the discovery of such mutations, the efficacy of a MET TKI was evidenced in NSCLC patients displaying MET exon 14 skipping. Yet its use led to a resistance mechanism involving acquisition of novel and already characterized MET mutations. Furthermore, novel somatic MET mutations are constantly being discovered. The challenge is no longer to identify them but to characterize them in order to predict their transforming activity and their sensitivity or resistance to MET TKIs, in order to adapt treatment.

    1. Cancer Biology
    2. Genetics and Genomics
    Kevin Nuno, Armon Azizi ... Ravindra Majeti
    Research Article

    Relapse of acute myeloid leukemia (AML) is highly aggressive and often treatment refractory. We analyzed previously published AML relapse cohorts and found that 40% of relapses occur without changes in driver mutations, suggesting that non-genetic mechanisms drive relapse in a large proportion of cases. We therefore characterized epigenetic patterns of AML relapse using 26 matched diagnosis-relapse samples with ATAC-seq. This analysis identified a relapse-specific chromatin accessibility signature for mutationally stable AML, suggesting that AML undergoes epigenetic evolution at relapse independent of mutational changes. Analysis of leukemia stem cell (LSC) chromatin changes at relapse indicated that this leukemic compartment underwent significantly less epigenetic evolution than non-LSCs, while epigenetic changes in non-LSCs reflected overall evolution of the bulk leukemia. Finally, we used single-cell ATAC-seq paired with mitochondrial sequencing (mtscATAC) to map clones from diagnosis into relapse along with their epigenetic features. We found that distinct mitochondrially-defined clones exhibit more similar chromatin accessibility at relapse relative to diagnosis, demonstrating convergent epigenetic evolution in relapsed AML. These results demonstrate that epigenetic evolution is a feature of relapsed AML and that convergent epigenetic evolution can occur following treatment with induction chemotherapy.