Human WDR5 promotes breast cancer growth and metastasis via KMT2-independent translation regulation

  1. Wesley L Cai
  2. Jocelyn Fang-Yi Chen
  3. Huacui Chen
  4. Emily Wingrove
  5. Sarah J Kurley
  6. Lok Hei Chan
  7. Meiling Zhang
  8. Anna Arnal-Estape
  9. Minghui Zhao
  10. Amer Balabaki
  11. Wenxue Li
  12. Xufen Yu
  13. Ethan D Krop
  14. Yali Dou
  15. Yansheng Liu
  16. Jian Jin
  17. Thomas F Westbrook
  18. Don X Nguyen  Is a corresponding author
  19. Qin Yan  Is a corresponding author
  1. University of Pittsburgh Medical Center, United States
  2. Yale University, United States
  3. Baylor College of Medicine, United States
  4. Icahn School of Medicine at Mount Sinai, United States
  5. University of Southern California, United States

Abstract

Metastatic breast cancer remains a major cause of cancer related deaths in women and there are few effective therapies against this advanced disease. Emerging evidence suggests that key steps of tumor progression and metastasis are controlled by reversible epigenetic mechanisms. Using an in vivo genetic screen, we identified WDR5 as an actionable epigenetic regulator that is required for metastatic progression in models of triple-negative breast cancer. We found that knockdown of WDR5 in breast cancer cells independently impaired their tumorigenic as well as metastatic capabilities. Mechanistically, WDR5 promotes cell growth by increasing ribosomal gene expression and translation efficiency in a KMT2-independent manner. Consistently, pharmacological inhibition or degradation of WDR5 impedes cellular translation rate and the clonogenic ability of breast cancer cells. Furthermore, combination of WDR5-targeting with mTOR inhibitors leads to potent suppression of translation and proliferation of breast cancer cells. These results reveal novel therapeutic strategies to treat metastatic breast cancer.

Data availability

RNA-seq data have been deposited into the National Center for Biotechnology Information (NCBI) Gene Expression Omnibus database under GSE196666. Reviewer token: qhqpeackxnebvqn.

The following data sets were generated

Article and author information

Author details

  1. Wesley L Cai

    Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, United States
    Competing interests
    No competing interests declared.
  2. Jocelyn Fang-Yi Chen

    Department of Pathology, Yale University, New Haven, United States
    Competing interests
    No competing interests declared.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0001-7281-8686
  3. Huacui Chen

    Department of Pathology, Yale University, New Haven, United States
    Competing interests
    No competing interests declared.
  4. Emily Wingrove

    Department of Pathology, Yale University, New Haven, United States
    Competing interests
    No competing interests declared.
  5. Sarah J Kurley

    Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, United States
    Competing interests
    No competing interests declared.
  6. Lok Hei Chan

    Department of Pathology, Yale University, New Haven, United States
    Competing interests
    No competing interests declared.
  7. Meiling Zhang

    Department of Pathology, Yale University, New Haven, United States
    Competing interests
    No competing interests declared.
  8. Anna Arnal-Estape

    Department of Pathology, Yale University, New Haven, United States
    Competing interests
    No competing interests declared.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0003-0490-7040
  9. Minghui Zhao

    Department of Pathology, Yale University, New Haven, United States
    Competing interests
    No competing interests declared.
  10. Amer Balabaki

    Department of Pathology, Yale University, New Haven, United States
    Competing interests
    No competing interests declared.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-1509-886X
  11. Wenxue Li

    Department of Pharmacology, Yale University, West Haven, United States
    Competing interests
    No competing interests declared.
  12. Xufen Yu

    Mount Sinai Center for Therapeutics Discovery, Icahn School of Medicine at Mount Sinai, New York, United States
    Competing interests
    No competing interests declared.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0001-7794-7890
  13. Ethan D Krop

    Department of Pathology, Yale University, New Haven, United States
    Competing interests
    No competing interests declared.
  14. Yali Dou

    Department of Medicine, University of Southern California, Los Angeles, United States
    Competing interests
    No competing interests declared.
  15. Yansheng Liu

    Department of Pharmacology, Yale University, West Haven, United States
    Competing interests
    No competing interests declared.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-2626-3912
  16. Jian Jin

    Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, United States
    Competing interests
    Jian Jin, . The Jin laboratory received research funds un-related to this study from Celgene Corporation, Levo Therapeutics, Inc., Cullgen, Inc. and Cullinan Oncology, Inc. J.J. is a cofounder, scientific advisory board member and equity shareholder in Cullgen, Inc. and a consultant for Cullgen, Inc., EpiCypher, Inc. and Accent Therapeutics, Inc..
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-2387-3862
  17. Thomas F Westbrook

    Department of Molecular and Human Genetics, Baylor College of Medicine, Boston, United States
    Competing interests
    No competing interests declared.
  18. Don X Nguyen

    Department of Pathology, Yale University, New Haven, United States
    For correspondence
    don.nguyen@yale.edu
    Competing interests
    Don X Nguyen, has received research funding un-related to this study from AstraZeneca Inc..
  19. Qin Yan

    Department of Pathology, Yale University, New Haven, United States
    For correspondence
    qin.yan@yale.edu
    Competing interests
    No competing interests declared.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0003-4077-453X

Funding

National Science Foundation (Graduate Research FellowshipDGE-1122492)

  • Wesley L Cai

National Cancer Institute (F31CA243295)

  • Jocelyn Fang-Yi Chen

Congressionally Directed Medical Research Programs (W81XWH-15-1-0117 and W81XWH-21-1-0411)

  • Qin Yan

National Cancer Institute (R01CA237586)

  • Qin Yan

National Cancer Institute (R01CA166376)

  • Don X Nguyen

National Cancer Institute (P30CA016359)

  • Qin Yan

Yale Cancer Center (Class of '61 Cancer Research Award)

  • Qin Yan

Yale Cancer Center (Class of '61 Cancer Research Award)

  • Don X Nguyen

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 procedures have been approved by the Institutional Animal Care and Use Committee of Yale University under animal protocol 2021-11286.

Version history

  1. Received: February 25, 2022
  2. Preprint posted: March 30, 2022 (view preprint)
  3. Accepted: August 24, 2022
  4. Accepted Manuscript published: August 31, 2022 (version 1)
  5. Version of Record published: October 20, 2022 (version 2)

Copyright

© 2022, Cai 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

  • 2,226
    views
  • 519
    downloads
  • 13
    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. Wesley L Cai
  2. Jocelyn Fang-Yi Chen
  3. Huacui Chen
  4. Emily Wingrove
  5. Sarah J Kurley
  6. Lok Hei Chan
  7. Meiling Zhang
  8. Anna Arnal-Estape
  9. Minghui Zhao
  10. Amer Balabaki
  11. Wenxue Li
  12. Xufen Yu
  13. Ethan D Krop
  14. Yali Dou
  15. Yansheng Liu
  16. Jian Jin
  17. Thomas F Westbrook
  18. Don X Nguyen
  19. Qin Yan
(2022)
Human WDR5 promotes breast cancer growth and metastasis via KMT2-independent translation regulation
eLife 11:e78163.
https://doi.org/10.7554/eLife.78163

Share this article

https://doi.org/10.7554/eLife.78163

Further reading

    1. Cancer Biology
    Fang Huang, Zhenwei Dai ... Yang Wang
    Research Article

    Aberrant alternative splicing is well-known to be closely associated with tumorigenesis of various cancers. However, the intricate mechanisms underlying breast cancer metastasis driven by deregulated splicing events remain largely unexplored. Here, we unveiled that RBM7 is decreased in lymph node and distant organ metastases of breast cancer as compared to primary lesions and low expression of RBM7 is correlated with the reduced disease-free survival of breast cancer patients. Breast cancer cells with RBM7 depletion exhibited an increased potential for lung metastasis compared to scramble control cells. The absence of RBM7 stimulated breast cancer cell migration, invasion, and angiogenesis. Mechanistically, RBM7 controlled the splicing switch of MFGE8, favoring the production of the predominant isoform of MFGE8, MFGE8-L. This resulted in the attenuation of STAT1 phosphorylation and alterations in cell adhesion molecules. MFGE8-L exerted an inhibitory effect on the migratory and invasive capability of breast cancer cells, while the truncated isoform MFGE8-S, which lack the second F5/8 type C domain had the opposite effect. In addition, RBM7 negatively regulates the NF-κB cascade and an NF-κB inhibitor could obstruct the increase in HUVEC tube formation caused by RBM7 silencing. Clinically, we noticed a positive correlation between RBM7 expression and MFGE8 exon7 inclusion in breast cancer tissues, providing new mechanistic insights for molecular-targeted therapy in combating breast cancer.

    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.