4.3 Article

C3G promotes a selective release of angiogenic factors from activated mouse platelets to regulate angiogenesis and tumor metastasis

Journal

ONCOTARGET
Volume 8, Issue 67, Pages 110994-111011

Publisher

IMPACT JOURNALS LLC
DOI: 10.18632/oncotarget.22339

Keywords

C3G; platelet secretome; angiogenesis; Vamp-7; metastasis

Funding

  1. Spanish Ministry of Economy and Competitiveness [SAF2013-48210-C2-1-R, SAF2016-76588-C2-2-R, SAF2013-48210-C2-2-R, SAF2016-76588-C2-1-R]
  2. Council of Education of Junta de Castilla y Leon, Spain [SA157A12-1, SA017U16]
  3. Council of Health of Junta de Castilla y Leon, Spain [GRS 991/A/14]
  4. European FEDER Program
  5. Dr. Moraza Foundation PhD fellowship
  6. Salamanca University
  7. Complutense University

Ask authors/readers for more resources

Previous observations indicated that C3G (RAPGEF1) promotes a-granule release, evidenced by the increase in P-selectin exposure on the platelet surface following its activation. The goal of the present study is to further characterize the potential function of C3G as a modulator of the platelet releasate and its implication in the regulation of angiogenesis. Proteomic analysis revealed a decreased secretion of anti-angiogenic factors from activated transgenic C3G and C3G.Cat platelets. Accordingly, the secretome from both transgenic platelets had an overall pro-angiogenic effect as evidenced by an in vitro capillary-tube formation assay with HUVECs (human umbilical vein endothelial cells) and by two in vivo models of heterotopic tumor growth. In addition, transgenic C3G expression in platelets greatly increased mouse melanoma cells metastasis. Moreover, immunofluorescence microscopy showed that the pro-angiogenic factors VEGF and bFGF were partially retained into a-granules in thrombin-and ADP-activated mouse platelets from both, C3G and C3G.Cat transgenic mice. The observed interaction between C3G and Vesicle-associated membrane protein (Vamp)-7 could explain these results. Concomitantly, increased platelet spreading in both transgenic platelets upon thrombin activation supports this novel function of C3G in a-granule exocytosis. Collectively, our data point out to the co-existence of Rap1GEF-dependent and independent mechanisms mediating C3G effects on platelet secretion, which regulates pathological angiogenesis in tumors and other contexts. The results herein support an important role for platelet C3G in angiogenesis and metastasis.

Authors

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

Reviews

Primary Rating

4.3
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available