4.6 Article

Gastric Cancer-Derived Extracellular Vesicles (EVs) Promote Angiogenesis via Angiopoietin-2

Journal

CANCERS
Volume 14, Issue 12, Pages -

Publisher

MDPI
DOI: 10.3390/cancers14122953

Keywords

gastric cancer (GC); endothelial cells (ECs); angiogenesis; angiopoetin-2 (ANG2); extracellular vesicles (EVs)

Categories

Funding

  1. Cancer Biology Research Center (CBRC), Tel Aviv University

Ask authors/readers for more resources

Angiogenesis is a crucial process for the growth and metastasis of gastric cancer. Angiopoietin-2 is an important driver of tumor angiogenesis and has shown potential as a target for antiangiogenic therapy. Extracellular vesicles play a significant role in tumor progression, including angiogenesis. This study investigated the interaction between gastric cancer and endothelial cells mediated by vesicles, focusing on angiopoietin-2. The results demonstrated the expression of angiopoietin-2 in primary gastric cancer tissues and omental metastasis tissues. Gastric cancer vesicles were found to induce proliferation, migration, invasion, and tube formation of endothelial cells. The vesicles also contained high levels of proangiogenic proteins, including angiopoietin-2. Knockdown of angiopoietin-2 showed that it mediates the proangiogenic effects of gastric cancer vesicles. These findings reveal a novel mechanism by which gastric cancer cells induce angiogenesis and suggest its potential as a target for cancer therapy.
Simple Summary Angiogenesis is the formation of new blood vessels, which is essential for gastric cancer growth and metastasis. Angiopoietin-2 is a key driver of tumor angiogenesis and has recently emerged as a promising target for antiangiogenic therapy. Extracellular vesicles play an important role in tumor progression including angiogenesis. We explored the crosstalk between gastric cancer and endothelial cells mediated by vesicles, with a specific focus on angiopoietin-2. We show that primary gastric cancer and omental metastasis tissues express angiopoietin-2. We isolated gastric cancer vesicles and demonstrated that they induce the proliferation, migration, invasion, and tube formation of endothelial cells. Characterization of the angiogenic profile of these vesicles revealed high levels of proangiogenic proteins including angiopoietin-2. Using angiopoietin-2 knockdown, we demonstrate that angiopoietin-2 mediates the proangiogenic effects of the gastric cancer vesicles. Our findings suggest a new mechanism via which gastric cancer cells induce angiogenesis. Such a mechanism may be used as a target for cancer therapy. Angiogenesis is an important control point of gastric cancer (GC) progression and metastasis. Angiopoietin-2 (ANG2) is a key driver of tumor angiogenesis and metastasis, and it has been identified in primary GC tissues. Extracellular vesicles (EVs) play an important role in mediating intercellular communication through the transfer of proteins between cells. However, the expression of ANG2 in GC-EVs has never been reported. Here, we characterized the EV-mediated crosstalk between GC and endothelial cells (ECs), with particular focus on the role of ANG2. We first demonstrate that ANG2 is expressed in GC primary and metastatic tissues. We then isolated EVs from two different GC cell lines and showed that these EVs enhance EC proliferation, migration, invasion, and tube formation in vitro and in vivo. Using an angiogenesis protein array, we showed that GC-EVs contain high levels of proangiogenic proteins, including ANG2. Lastly, using Lenti viral ANG2-shRNA, we demonstrated that the proangiogenic effects of the GC-EVs were mediated by ANG2 through the activation of the PI3K/Akt signal transduction pathway. Our data suggest a new mechanism via which GC cells induce angiogenesis. This knowledge may be utilized to develop new therapies in gastric cancer.

Authors

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

Reviews

Primary Rating

4.6
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available