Abstract

Cancer cell-stromal cell crosstalk is orchestrated by a plethora of ligand-receptor interactions generating a tumor microenvironment (TME) which favors tumor growth. The high pro-angiogenic nature of the TME perpetuates the chaotic network of structurally immature, low pericyte-covered vessels characteristic of the tumor vasculature. We previously demonstrated that chloroquine (CQ) -a lysosomotropic agent used as first-generation autophagy blocker in clinical trials- induced tumor vessel normalization and reduced tumor hypoxia. CQ improved both vessel structure and maturation, whereas the conditional knockout of the crucial autophagy gene Atg5 in endothelial cells (ECs) did not, thus highlighting a potential differential role for EC-associated autophagy and the lysosomes in pathological tumor angiogenesis. However, how CQ or ATG5-deficiency in ECs affect angiogenic signals regulating EC-pericyte interface and therefore vessel maturation, remains unknown. Here, we show that in ECs CQ constrained VEGF-A-mediated VEGF receptor (VEGFR)2 phosphorylation, a driver of angiogenic signaling. In the presence of CQ we observed increased expression of the decoy receptor VEGFR1 and of a lower molecular weight form of VEGFR2, suggesting receptor cleavage. Consequently, VEGF-A-driven EC spheroid sprouting was reduced by CQ treatment. Furthermore, CQ significantly affected the transcription and secretion of platelet-derived growth factor (PDGF)-AB/BB (upregulated) and Endothelin-1 (EDN1, downregulated), both modulators of perivascular cell (PC) behavior. In contrast, silencing of ATG5 in ECs had no effect on VEGFR2 to VEGFR1 ratio nor on PDGFB and EDN1 expression. Accordingly, mice harboring B16F10 melanoma tumors treated with CQ, displayed both an increased number of αSMA+ PCs covering tumor vessels and co-expressed PDGF receptor-β, enabling PDGF ligand dependent recruitment. Moreover, upon CQ treatment the tumoral expression of angiopoietin-1 (Angpt1), which retains mural cells, and induces vessel stabilization by binding to the EC-localized cognate receptor (TIE2), was increased thus supporting the vessel normalization function of CQ. These features associated with improved tumor vasculature were not phenocopied by the specific deletion of Atg5 in ECs. In conclusion, this study further unravels endothelial cell autonomous and non-autonomous mechanisms by which CQ “normalizes” the intercellular communication in the tumor vasculature independent of autophagy.

Highlights

  • Physiological angiogenesis is a multistep process that involves, migration and proliferation of endothelial cells (ECs), remodeling of the extracellular matrix and functional maturation of the newly assembled vessels

  • We first evaluated the effects of CQ treatment or ATG5 silencing on the VEGFR1, and VEGFR2 expression in human umbilical vein ECs (HUVECs)

  • CQ treatment of HUVECs induced a significant increase in the VEGFR1 gene expression -without altering the expression of VEGFR2- already after 24 h and led to a significant decrease in the VEGFR2/VEGFR1 mRNA expression ratio (Supplementary Figures 1A,B)

Read more

Summary

Introduction

Physiological angiogenesis is a multistep process that involves, migration and proliferation of endothelial cells (ECs), remodeling of the extracellular matrix and functional maturation of the newly assembled vessels. The latter process features the recruitment of perivascular cells (PCs), principally classified as pericytes or vascular smooth muscle cells (vSMCs), which envelop the endothelial wall to ameliorate vessel stability [1]. Tumor vessels are characterized by chaotic branching, ill-coverage of vesselstabilizing PCs, and high level of leakiness [2] This aberrant vascular phenotype supports crucial tumor microenvironment (TME) conditions including hypoxia, acidity, and high interstitial pressure, which promote tumor progression by e.g., dampening antitumor immunity, selecting for the most aggressive cancer cell subclones, and reducing the efficacy of therapies [3]. An interesting shift in concept assumes that rather than pruning the vasculature, Abbreviations: ANGPT, angiopoietin; αSMA, alpha smooth muscle actin; CQ, chloroquine; EC, endothelial cell; EDN1, endothelin-1; HB-EGF, heparin-binding epidermal growth factor-like growth factor; HUVEC, human umbilical cord endothelial cell; NICD, notch intracellular domain; PC, perivascular cell; PDGF, platelet-derived growth factor; PDGFR, PDGF receptor; PECAM1, platelet endothelial cell adhesion molecule-1; TME, tumor microenvironment; TGFβ1, transforming growth factor beta 1; VEGF, vascular endothelial growth factor; VEGFR, VEGF receptor; vSMC, vascular smooth muscle cell

Objectives
Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call