MEIS-mediated suppression of human prostate cancer growth and metastasis through HOXB13-dependent regulation of proteoglycans

  1. Calvin VanOpstall
  2. Srikanth Perike
  3. Hannah Brechka
  4. Marc Gillard
  5. Sophia Lamperis
  6. Baizhen Zhu
  7. Ryan Brown
  8. Raj Bhanvadia
  9. Donald J Vander Griend  Is a corresponding author
  1. The University of Chicago, United States
  2. The University of Illinois at Chicago, United States

Abstract

The molecular roles of HOX transcriptional activity in human prostate epithelial cells remain unclear, impeding the implementation of new treatment strategies for cancer prevention and therapy. MEIS proteins are transcription factors that bind and direct HOX protein activity. MEIS proteins are putative tumor suppressors that are frequently silenced in aggressive forms of prostate cancer. Here we show that MEIS1 expression is sufficient to decrease proliferation and metastasis of prostate cancer cells in vitro and in vivo murine xenograft models. HOXB13 deletion demonstrates that the tumor-suppressive activity of MEIS1 is dependent on HOXB13. Integration of ChIP-seq and RNA-seq data revealed direct and HOXB13-dependent regulation of proteoglycans including decorin (DCN) as a mechanism of MEIS1-driven tumor suppression. These results define and underscore the importance of MEIS1-HOXB13 transcriptional regulation in suppressing prostate cancer progression and provide a mechanistic framework for the investigation of HOXB13 mutants and oncogenic cofactors when MEIS1/2 are silenced.

Data availability

RNA-seq and ChIP-seq raw and analyzed data have been deposited at the Gene Expression Omnibus and Sequence Read Archive under the accession number GSE132717.

The following previously published data sets were used

Article and author information

Author details

  1. Calvin VanOpstall

    Ben May Institute for Cancer Research, The University of Chicago, Chicago, United States
    Competing interests
    The authors declare that no competing interests exist.
  2. Srikanth Perike

    Pathology, The University of Illinois at Chicago, Chicago, United States
    Competing interests
    The authors declare that no competing interests exist.
  3. Hannah Brechka

    Surgery, The University of Chicago, Chicago, United States
    Competing interests
    The authors declare that no competing interests exist.
  4. Marc Gillard

    Surgery, The University of Chicago, Chicago, United States
    Competing interests
    The authors declare that no competing interests exist.
  5. Sophia Lamperis

    Pathology, The University of Illinois at Chicago, Chicago, United States
    Competing interests
    The authors declare that no competing interests exist.
  6. Baizhen Zhu

    Surgery, The University of Chicago, Chicago, United States
    Competing interests
    The authors declare that no competing interests exist.
  7. Ryan Brown

    Pathology, The University of Illinois at Chicago, Chicago, United States
    Competing interests
    The authors declare that no competing interests exist.
  8. Raj Bhanvadia

    Pritzker School of Medicine, The University of Chicago, Chicago, United States
    Competing interests
    The authors declare that no competing interests exist.
  9. Donald J Vander Griend

    Pathology, The University of Illinois at Chicago, Chicago, United States
    For correspondence
    dvanderg@uic.edu
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0003-4421-5698

Funding

U.S. Department of Defense (PC130587)

  • Donald J Vander Griend

U.S. Department of Defense (PC180414)

  • Donald J Vander Griend

National Institutes of Health (P50 CA180995)

  • Donald J Vander Griend

National Institutes of Health (T32 CA009594)

  • Calvin VanOpstall
  • Hannah Brechka
  • Donald J Vander Griend

National Institutes of Health (F31CA232651)

  • Calvin VanOpstall

National Institutes of Health (P30CA014599)

  • Donald J Vander Griend

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

Reviewing Editor

  1. Wilbert Zwart, Netherlands Cancer Institute, Netherlands

Ethics

Animal experimentation: All animal studies were carried out in strict accordance with the recommendations in the Guide for the Care and Use of Laboratory Animals of the National Institutes of Health. The protocol was approved by the University of Chicago Institutional Animal Care and Use Committee (IACUC) (protocol #72231) as well as by the University of Illinois at Chicago IACUC (protocol #18-100).

Version history

  1. Received: November 14, 2019
  2. Accepted: June 17, 2020
  3. Accepted Manuscript published: June 18, 2020 (version 1)
  4. Version of Record published: July 20, 2020 (version 2)

Copyright

© 2020, VanOpstall 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

  • 25,641
    views
  • 298
    downloads
  • 29
    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. Calvin VanOpstall
  2. Srikanth Perike
  3. Hannah Brechka
  4. Marc Gillard
  5. Sophia Lamperis
  6. Baizhen Zhu
  7. Ryan Brown
  8. Raj Bhanvadia
  9. Donald J Vander Griend
(2020)
MEIS-mediated suppression of human prostate cancer growth and metastasis through HOXB13-dependent regulation of proteoglycans
eLife 9:e53600.
https://doi.org/10.7554/eLife.53600

Share this article

https://doi.org/10.7554/eLife.53600

Further reading

    1. Cancer Biology
    2. Cell Biology
    Stefanie Schmieder
    Insight

    Mutations in the gene for β-catenin cause liver cancer cells to release fewer exosomes, which reduces the number of immune cells infiltrating the tumor.

    1. Cancer Biology
    2. Cell Biology
    Dongyue Jiao, Huiru Sun ... Kun Gao
    Research Article

    Enhanced protein synthesis is a crucial molecular mechanism that allows cancer cells to survive, proliferate, metastasize, and develop resistance to anti-cancer treatments, and often arises as a consequence of increased signaling flux channeled to mRNA-bearing eukaryotic initiation factor 4F (eIF4F). However, the post-translational regulation of eIF4A1, an ATP-dependent RNA helicase and subunit of the eIF4F complex, is still poorly understood. Here, we demonstrate that IBTK, a substrate-binding adaptor of the Cullin 3-RING ubiquitin ligase (CRL3) complex, interacts with eIF4A1. The non-degradative ubiquitination of eIF4A1 catalyzed by the CRL3IBTK complex promotes cap-dependent translational initiation, nascent protein synthesis, oncogene expression, and cervical tumor cell growth both in vivo and in vitro. Moreover, we show that mTORC1 and S6K1, two key regulators of protein synthesis, directly phosphorylate IBTK to augment eIF4A1 ubiquitination and sustained oncogenic translation. This link between the CRL3IBTK complex and the mTORC1/S6K1 signaling pathway, which is frequently dysregulated in cancer, represents a promising target for anti-cancer therapies.