Abstract

Leukemia stem cells (LSCs) are regarded as the origins and key therapeutic targets of leukemia, but limited knowledge is available on the key determinants of LSC 'stemness'. Using single-cell RNA-seq analysis, we identify a master regulator, SPI1, the LSC-specific expression of which determines the molecular signature and activity of LSCs in the murine Pten-null T-ALL model. Although initiated by PTEN-controlled b-catenin activation, Spi1 expression and LSC 'stemness' are maintained by a b-catenin-SPI1-HAVCR2 regulatory circuit independent of the leukemogenic driver mutation. Perturbing any component of this circuit either genetically or pharmacologically can prevent LSC formation or eliminate existing LSCs. LSCs lose their 'stemness' when Spi1 expression is silenced by DNA methylation, but Spi1 expression can be reactivated by 5-AZ treatment. Importantly, similar regulatory mechanisms may be also present in human T-ALLs.

Data availability

All the Bulk RNA-seq, Single cell RNA-seq and BiSulfite-seq data for this study are deposited in NCBI Gene Expression Omnibus under the accession number GSE115356.

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

Article and author information

Author details

  1. Haichuan Zhu

    School of Life Sciences, Peking University, Beijing, China
    Competing interests
    No competing interests declared.
  2. Liuzhen Zhang

    School of Life Sciences, Peking University, Beijing, China
    Competing interests
    No competing interests declared.
  3. Yilin Wu

    School of Life Sciences, Peking University, Beijing, China
    Competing interests
    No competing interests declared.
  4. Bingjie Dong

    School of Life Sciences, Peking University, Beijing, China
    Competing interests
    No competing interests declared.
  5. Weilong Guo

    School of Life Sciences, Peking University, Beijing, China
    Competing interests
    No competing interests declared.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0001-5199-1359
  6. Mei Wang

    School of Life Sciences, Peking University, Beijing, China
    Competing interests
    No competing interests declared.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0003-3292-1413
  7. Lu Yang

    School of Life Sciences, Peking University, Beijing, China
    Competing interests
    No competing interests declared.
  8. Xiaoying Fan

    School of Life Sciences, Peking University, Beijing, China
    Competing interests
    No competing interests declared.
  9. Yuliang Tang

    Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
    Competing interests
    No competing interests declared.
  10. Ningshu Liu

    Drug Discovery Oncology, Bayer Pharmaceuticals, Berlin, Germany
    Competing interests
    Ningshu Liu, is an employee of Bayer AG.
  11. Xiaoguang Lei

    Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
    Competing interests
    No competing interests declared.
  12. Hong Wu

    School of Life Sciences, Peking University, Beijing, China
    For correspondence
    Hongwu@pku.edu.cn
    Competing interests
    No competing interests declared.
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0001-7545-7919

Funding

Peking-tsinghua Center for Life science

  • Hong Wu

Beijing Advanced Innovation Center for Genomics

  • Hong Wu

Bayer Pharma

  • Hong Wu

National Key Research (Grant No. 2017YFA0505200)

  • Xiaoguang Lei

National Science Foundation of China

  • Lu Yang

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

Reviewing Editor

  1. A Thomas Look, Harvard Medical School, United States

Ethics

Animal experimentation: All experimental protocols were approved by the Peking University Animal Care and Use Committee (IACUC).This study were approved by the Peking University Animal Care and Use Committee (LSC-WuH-1).

Version history

  1. Received: May 13, 2018
  2. Accepted: November 9, 2018
  3. Accepted Manuscript published: November 9, 2018 (version 1)
  4. Version of Record published: November 23, 2018 (version 2)

Copyright

© 2018, 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

  • 3,225
    views
  • 583
    downloads
  • 31
    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. Haichuan Zhu
  2. Liuzhen Zhang
  3. Yilin Wu
  4. Bingjie Dong
  5. Weilong Guo
  6. Mei Wang
  7. Lu Yang
  8. Xiaoying Fan
  9. Yuliang Tang
  10. Ningshu Liu
  11. Xiaoguang Lei
  12. Hong Wu
(2018)
T-ALL leukemia stem cell 'stemness' is epigenetically controlled by the master regulator SPI1
eLife 7:e38314.
https://doi.org/10.7554/eLife.38314

Share this article

https://doi.org/10.7554/eLife.38314

Further reading

    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.

    1. Cancer Biology
    Chenxi Gao, Huaibin Ge ... Jing Hu
    Research Article

    BRAFV600E mutation is a driver mutation in the serrated pathway to colorectal cancers. BRAFV600E drives tumorigenesis through constitutive downstream extracellular signal-regulated kinase (ERK) activation, but high-intensity ERK activation can also trigger tumor suppression. Whether and how oncogenic ERK signaling can be intrinsically adjusted to a ‘just-right’ level optimal for tumorigenesis remains undetermined. In this study, we found that FAK (Focal adhesion kinase) expression was reduced in BRAFV600E-mutant adenomas/polyps in mice and patients. In Vil1-Cre;BRAFLSL-V600E/+;Ptk2fl/fl mice, Fak deletion maximized BRAFV600E’s oncogenic activity and increased cecal tumor incidence to 100%. Mechanistically, our results showed that Fak loss, without jeopardizing BRAFV600E-induced ERK pathway transcriptional output, reduced EGFR (epidermal growth factor receptor)-dependent ERK phosphorylation. Reduction in ERK phosphorylation increased the level of Lgr4, promoting intestinal stemness and cecal tumor formation. Our findings show that a ‘just-right’ ERK signaling optimal for BRAFV600E-induced cecal tumor formation can be achieved via Fak loss-mediated downregulation of ERK phosphorylation.