4.8 Article

Microenvironment-induced PTEN loss by exosomal microRNA primes brain metastasis outgrowth

Journal

NATURE
Volume 527, Issue 7576, Pages 100-104

Publisher

NATURE PUBLISHING GROUP
DOI: 10.1038/nature15376

Keywords

-

Funding

  1. DOD Center of Excellence [W81XWH-06-2-0033]
  2. NIH [5R00CA158066-05]
  3. DOD [W81XWH-11-1-0003]
  4. Isaiah Fidler Fellowship in Cancer [PO1-CA099031]
  5. Susan G. Komen Breast Cancer Foundation [KG091020]
  6. METAvivor Research Grant
  7. Breast and Ovarian Cancers Moon Shot program
  8. China Medical University Research Fund
  9. Sowell-Huggins Pre-doctoral Fellowship and Professorship in Cancer Research
  10. [RO1-CA112567-06]
  11. [R01CA184836]

Ask authors/readers for more resources

The development of life-threatening cancer metastases at distant organs requires disseminated tumour cells' adaptation to, and co-evolution with, the drastically different microenvironments of metastatic sites(1). Cancer cells of common origin manifest distinct gene expression patterns after metastasizing to different organs(2). Clearly, the dynamic interaction between metastatic tumour cells and extrinsic signals at individual metastatic organ sites critically effects the subsequent metastatic outgrowth(3,4). Yet, it is unclear when and how disseminated tumour cells acquire the essential traits from the microenvironment of metastatic organs that prime their subsequent outgrowth. Here we show that both human and mouse tumour cells with normal expression of PTEN, an important tumour suppressor, lose PTEN expression after dissemination to the brain, but not to other organs. The PTEN level in PTEN-loss brain metastatic tumour cells is restored after leaving the brain microenvironment. This brain microenvironment-dependent, reversible PTEN messenger RNA and protein downregulation is epigenetically regulated by microRNAs from brain astrocytes. Mechanistically, astrocyte-derived exosomes mediate an intercellular transfer of PTEN-targeting microRNAs to metastatic tumour cells, while astrocyte-specific depletion of PTEN-targeting microRNAs or blockade of astrocyte exosome secretion rescues the PTEN loss and suppresses brain metastasis in vivo. Furthermore, this adaptive PTEN loss in brain metastatic tumour cells leads to an increased secretion of the chemokine CCL2, which recruits IBA1-expressing myeloid cells that reciprocally enhance the outgrowth of brain metastatic tumour cells via enhanced proliferation and reduced apoptosis. Our findings demonstrate a remarkable plasticity of PTEN expression in metastatic tumour cells in response to different organ microenvironments, underpinning an essential role of co-evolution between the metastatic cells and their microenvironment during the adaptive metastatic outgrowth. Our findings signify the dynamic and reciprocal cross-talk between tumour cells and the metastatic niche; importantly, they provide new opportunities for effective anti-metastasis therapies, especially of consequence for brain metastasis patients.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available