Oridonin inhibits tumor growth and metastasis through anti-angiogenesis by blocking the Notch signaling

PLoS One. 2014 Dec 8;9(12):e113830. doi: 10.1371/journal.pone.0113830. eCollection 2014.

Abstract

While significant progress has been made in understanding the anti-inflammatory and anti-proliferative effects of the natural diterpenoid component Oridonin on tumor cells, little is known about its effect on tumor angiogenesis or metastasis and on the underlying molecular mechanisms. In this study, Oridonin significantly suppressed human umbilical vascular endothelial cells (HUVECs) proliferation, migration, and apillary-like structure formation in vitro. Using aortic ring assay and mouse corneal angiogenesis model, we found that Oridonin inhibited angiogenesis ex vivo and in vivo. In our animal experiments, Oridonin impeded tumor growth and metastasis. Immunohistochemistry analysis further revealed that the expression of CD31 and vWF protein in xenografts was remarkably decreased by the Oridonin. Furthermore, Oridonin reinforced endothelial cell-cell junction and impaired breast cancer cell transendothelial migration. Mechanistically, Oridonin not only down-regulated Jagged2 expression and Notch1 activity but also decreased the expression of their target genes. In conclusion, our results demonstrated an original role of Oridonin in inhibiting tumor angiogenesis and propose a mechanism. This study also provides new evidence supporting the central role of Notch in tumor angiogenesis and suggests that Oridonin could be a potential drug candidate for angiogenesis related diseases.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Angiogenesis Inhibitors / pharmacology*
  • Animals
  • Antineoplastic Agents / pharmacology*
  • Calcium-Binding Proteins / metabolism
  • Cell Communication / drug effects
  • Cell Line, Tumor
  • Cell Movement / drug effects
  • Cell Proliferation / drug effects
  • Disease Models, Animal
  • Diterpenes, Kaurane / pharmacology*
  • Human Umbilical Vein Endothelial Cells / drug effects
  • Human Umbilical Vein Endothelial Cells / metabolism
  • Humans
  • Intercellular Signaling Peptides and Proteins / metabolism
  • Male
  • Membrane Proteins / metabolism
  • Mice
  • Neoplasm Metastasis
  • Neoplasms / drug therapy
  • Neoplasms / metabolism*
  • Neoplasms / pathology*
  • Neovascularization, Pathologic / drug therapy
  • Neovascularization, Pathologic / metabolism*
  • Rats
  • Receptors, Notch / metabolism*
  • Serrate-Jagged Proteins
  • Signal Transduction / drug effects*
  • Tumor Burden / drug effects
  • Vascular Endothelial Growth Factor A / pharmacology
  • Xenograft Model Antitumor Assays

Substances

  • Angiogenesis Inhibitors
  • Antineoplastic Agents
  • Calcium-Binding Proteins
  • Diterpenes, Kaurane
  • Intercellular Signaling Peptides and Proteins
  • Membrane Proteins
  • Receptors, Notch
  • Serrate-Jagged Proteins
  • Vascular Endothelial Growth Factor A
  • oridonin

Grants and funding

This study was partially supported by Major State Basic Research Development Program of China (2015CB910400). National Natural Science Foundation of China (81272463, 81472788, 81330049). Program for Changjiang Scholars and Innovative Research Team in University (IRT1128). Innovation Program of Shanghai Municipal Education Commission (13zz034). The Science and Technology Commission of Shanghai Municipality (11DZ2260300). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.