4.8 Article

ANGPTL2-containing small extracellular vesicles from vascular endothelial cells accelerate leukemia progression

Journal

JOURNAL OF CLINICAL INVESTIGATION
Volume 131, Issue 1, Pages -

Publisher

AMER SOC CLINICAL INVESTIGATION INC
DOI: 10.1172/JCI138986

Keywords

-

Funding

  1. Ministry of Science and Technology of China [2020YFE0203000, 2019YFA0801800, 2016YFA0100600, 2017YFA0103400, 2018YFA0107000]
  2. National Natural Science Foundation of China [81730006, 81825001, 81922002, 81421002, 81721004, 81890990, 81861148029, 81870086, 81670106]
  3. Chinese Academy of Medical Sciences (CAMS) Initiative for Innovative Medicine [2017-I2M-3-009, 2016-I2M-1-017]
  4. Natural Science Foundation of Shanghai [17ZR1415500]
  5. Shanghai Science and Technology Commission [19XD1422100]
  6. CAMS Fundamental Research Funds for Central Research Institutes [2019RC310003]
  7. SKLEH-Pilot Research Grant [Z20-04]
  8. Distinguished Young Scholars of Tianjin [19JCJQJC63400]

Ask authors/readers for more resources

Reducing SEV production from endothelial cells significantly delays leukemia progression, without affecting normal hematopoiesis. These findings reveal a role of niche-specific SEVs in cancer development and suggest that targeting ANGPTL2-SEVs released from endothelial cells may be a potential strategy to interfere with certain types of AML.
Small extracellular vesicles (SEVs) are functional messengers of certain cellular niches that permit noncontact cell communications. Whether niche-specific SEVs fulfill this role in cancer is unclear. Here, we used 7 cell type-specific mouse Cre lines to conditionally knock out Vps33b in Cdh5(+) or Tie2(+) endothelial cells (ECs), Lepr(+) BM perivascular cells, Osx(+) osteoprogenitor cells, Pf4(+) megakaryocytes, and Tcf21(+ )spleen stromal cells. We then examined the effects of reduced SEV secretion on progression of MLL-AF9-induced acute myeloid leukemia (AML), as well as normal hematopoiesis. Blocking SEV secretion from ECs, but not perivascular cells, megakaryocytes, or spleen stromal cells, markedly delayed the leukemia progression. Notably, reducing SEV production from ECs had no effect on normal hematopoiesis. Protein analysis showed that EC-derived SEVs contained a high level of ANGPTL2, which accelerated leukemia progression via binding to the LILRB2 receptor. Moreover, ANGPTL2-SEVs released from ECs were governed by VPS33B. Importantly, ANGPTL2-SEVs were also required for primary human AML cell maintenance. These findings demonstrate a role of niche-specific SEVs in cancer development and suggest targeting of ANGPTL2-SEVs from ECs as a potential strategy to interfere with certain types of AML.

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