ZHX2 promotes HIF1α oncogenic signaling in triple-negative breast cancer

  1. Wentong Fang
  2. Chengheng Liao  Is a corresponding author
  3. Rachel Shi
  4. Jeremy M Simon
  5. Travis S Ptacek
  6. Giada Zurlo
  7. Youqiong Ye
  8. Leng Han
  9. Cheng Fan
  10. Lei Bao
  11. Christopher Llynard Ortiz
  12. Hong-Rui Lin
  13. Ujjawal Manocha
  14. Weibo Luo
  15. Yan Peng
  16. William Y Kim
  17. Lee-Wei Yang
  18. Qing Zhang  Is a corresponding author
  1. The First Affiliated Hospital of Nanjing Medical University, China
  2. University of Texas Southwestern Medical Center, United States
  3. University of North Carolina School of Medicine, United States
  4. The University of Alabama at Birmingham, United States
  5. Shanghai Jiao Tong University School of Medicine, China
  6. The University of Texas Health Science Center at Houston McGovern Medical School, United States
  7. Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Taiwan
  8. The University of Texas Southwestern Medical Center, United States
  9. Department of Pathology, University of Texas Southwestern Medical Center, United States
  10. National Tsing Hua University, Taiwan

Abstract

Triple-negative breast cancer (TNBC) is an aggressive and highly lethal disease, which warrants the critical need to identify new therapeutic targets. We show that Zinc Fingers and Homeoboxes 2 (ZHX2) is amplified or overexpressed in TNBC cell lines and patients. Functionally, depletion of ZHX2 inhibited TNBC cell growth and invasion in vitro, orthotopic tumor growth and spontaneous lung metastasis in vivo. Mechanistically, ZHX2 bound with hypoxia inducible factor (HIF) family members and positively regulated HIF1a activity in TNBC. Integrated ChIP-Seq and gene expression profiling demonstrated that ZHX2 co-occupied with HIF1a on transcriptionally active promoters marked by H3K4me3 and H3K27ac, thereby promoting gene expression. Among the identified ZHX2 and HIF1a co-regulated genes, overexpression of AP2B1, COX20, KDM3A, or PTGES3L could partially rescue TNBC cell growth defect by ZHX2 depletion, suggested that these downstream targets contribute to the oncogenic role of ZHX2 in an accumulative fashion. Furthermore, multiple residues (R491, R581 and R674) on ZHX2 are important in regulating its phenotype, which correspond with their roles on controlling ZHX2 transcriptional activity in TNBC cells. These studies establish that ZHX2 activates oncogenic HIF1a signaling, therefore serving as a potential therapeutic target for TNBC.

Data availability

•Sequencing data have been deposited in GEO under accession codes GSE175487

The following data sets were generated

Article and author information

Author details

  1. Wentong Fang

    The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0003-0047-1198
  2. Chengheng Liao

    Department of Pathology, University of Texas Southwestern Medical Center, Dallas, United States
    For correspondence
    chengheng.liao@utsouthwestern.edu
    Competing interests
    The authors declare that no competing interests exist.
  3. Rachel Shi

    Department of Pathology, University of Texas Southwestern Medical Center, Dallas, United States
    Competing interests
    The authors declare that no competing interests exist.
  4. Jeremy M Simon

    Neuroscience Center; Carolina Institute for Developmental Disabilities; Department of Genetics, University of North Carolina School of Medicine, Chapel Hill, United States
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0003-3906-1663
  5. Travis S Ptacek

    Department of Microbiology, The University of Alabama at Birmingham, Birmingham, United States
    Competing interests
    The authors declare that no competing interests exist.
  6. Giada Zurlo

    Department of Pathology, University of Texas Southwestern Medical Center, Dallas, United States
    Competing interests
    The authors declare that no competing interests exist.
  7. Youqiong Ye

    Shanghai Institute of Immunology, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
    Competing interests
    The authors declare that no competing interests exist.
  8. Leng Han

    The University of Texas Health Science Center at Houston McGovern Medical School, Houston, United States
    Competing interests
    The authors declare that no competing interests exist.
  9. Cheng Fan

    Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel hill, United States
    Competing interests
    The authors declare that no competing interests exist.
  10. Lei Bao

    Department of Pathology, University of Texas Southwestern Medical Center, Dallas, United States
    Competing interests
    The authors declare that no competing interests exist.
  11. Christopher Llynard Ortiz

    Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-3114-7369
  12. Hong-Rui Lin

    Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
    Competing interests
    The authors declare that no competing interests exist.
  13. Ujjawal Manocha

    Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel hill, United States
    Competing interests
    The authors declare that no competing interests exist.
  14. Weibo Luo

    Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, United States
    Competing interests
    The authors declare that no competing interests exist.
  15. Yan Peng

    Department of Pathology, University of Texas Southwestern Medical Center, Dallas, United States
    Competing interests
    The authors declare that no competing interests exist.
  16. William Y Kim

    Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, United States
    Competing interests
    The authors declare that no competing interests exist.
  17. Lee-Wei Yang

    Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-3971-6386
  18. Qing Zhang

    Department of Pathology, University of Texas Southwestern Medical Center, Dallas, United States
    For correspondence
    qing.zhang@utsouthwestern.edu
    Competing interests
    The authors declare that no competing interests exist.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-6595-8995

Funding

National Cancer Institute (R01CA211732)

  • Qing Zhang

National Cancer Institute (R01CA256833)

  • Qing Zhang

Cancer Prevention and Research Institute of Texas (RR190058)

  • Qing Zhang

American Cancer Society (RSG-18-059-01-TBE)

  • Qing Zhang

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

Ethics

Animal experimentation: All animal experiments were in compliance with National Institutes of Health guidelines and were approved by the University of Texas, Southwestern Medical Center Institutional Animal Care and Use Committee.

Copyright

© 2021, Fang 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

  • 1,547
    views
  • 258
    downloads
  • 22
    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. Wentong Fang
  2. Chengheng Liao
  3. Rachel Shi
  4. Jeremy M Simon
  5. Travis S Ptacek
  6. Giada Zurlo
  7. Youqiong Ye
  8. Leng Han
  9. Cheng Fan
  10. Lei Bao
  11. Christopher Llynard Ortiz
  12. Hong-Rui Lin
  13. Ujjawal Manocha
  14. Weibo Luo
  15. Yan Peng
  16. William Y Kim
  17. Lee-Wei Yang
  18. Qing Zhang
(2021)
ZHX2 promotes HIF1α oncogenic signaling in triple-negative breast cancer
eLife 10:e70412.
https://doi.org/10.7554/eLife.70412

Share this article

https://doi.org/10.7554/eLife.70412

Further reading

    1. Cancer Biology
    2. Cell Biology
    Francesca Butera, Julia E Sero ... Chris Bakal
    Research Article

    The canonical NF-κB transcription factor RELA is a master regulator of immune and stress responses and is upregulated in PDAC tumours. In this study, we characterised previously unexplored endogenous RELA-GFP dynamics in PDAC cell lines through live single cell imaging. Our observations revealed that TNFα stimulation induces rapid, sustained, and non-oscillatory nuclear translocation of RELA. Through Bayesian analysis of single cell datasets with variation in nuclear RELA, we predicted that RELA heterogeneity in PDAC cell lines is dependent on F-actin dynamics. RNA-seq analysis identified distinct clusters of RELA-regulated gene expression in PDAC cells, including TNFα-induced RELA upregulation of the actin regulators NUAK2 and ARHGAP31. Further, siRNA-mediated depletion of ARHGAP31 and NUAK2 altered TNFα-stimulated nuclear RELA dynamics in PDAC cells, establishing a novel negative feedback loop that regulates RELA activation by TNFα. Additionally, we characterised the NF-κB pathway in PDAC cells, identifying how NF-κB/IκB proteins genetically and physically interact with RELA in the absence or presence of TNFα. Taken together, we provide computational and experimental support for interdependence between the F-actin network and the NF-κB pathway with RELA translocation dynamics in PDAC.

    1. Cancer Biology
    Danielle Algranati, Roni Oren ... Efrat Shema
    Research Article

    Diffuse midline gliomas (DMGs) are aggressive and fatal pediatric tumors of the central nervous system that are highly resistant to treatments. Lysine to methionine substitution of residue 27 on histone H3 (H3-K27M) is a driver mutation in DMGs, reshaping the epigenetic landscape of these cells to promote tumorigenesis. H3-K27M gliomas are characterized by deregulation of histone acetylation and methylation pathways, as well as the oncogenic MYC pathway. In search of effective treatment, we examined the therapeutic potential of dual targeting of histone deacetylases (HDACs) and MYC in these tumors. Treatment of H3-K27M patient-derived cells with Sulfopin, an inhibitor shown to block MYC-driven tumors in vivo, in combination with the HDAC inhibitor Vorinostat, resulted in substantial decrease in cell viability. Moreover, transcriptome and epigenome profiling revealed synergistic effect of this drug combination in downregulation of prominent oncogenic pathways such as mTOR. Finally, in vivo studies of patient-derived orthotopic xenograft models showed significant tumor growth reduction in mice treated with the drug combination. These results highlight the combined treatment with PIN1 and HDAC inhibitors as a promising therapeutic approach for these aggressive tumors.