BMC Biology (Jan 2018)

A role for endothelial nitric oxide synthase in intestinal stem cell proliferation and mesenchymal colorectal cancer

  • Jon Peñarando,
  • Laura M. López-Sánchez,
  • Rafael Mena,
  • Silvia Guil-Luna,
  • Francisco Conde,
  • Vanessa Hernández,
  • Marta Toledano,
  • Victoria Gudiño,
  • Michela Raponi,
  • Caroline Billard,
  • Carlos Villar,
  • César Díaz,
  • José Gómez-Barbadillo,
  • Juan De la Haba-Rodríguez,
  • Kevin Myant,
  • Enrique Aranda,
  • Antonio Rodríguez-Ariza

DOI
https://doi.org/10.1186/s12915-017-0472-5
Journal volume & issue
Vol. 16, no. 1
pp. 1 – 14

Abstract

Read online

Abstract Background Nitric oxide (NO) has been highlighted as an important agent in cancer-related events. Although the inducible nitric oxide synthase (iNOS) isoform has received most attention, recent studies in the literature indicate that the endothelial isoenzyme (eNOS) can also modulate different tumor processes including resistance, angiogenesis, invasion, and metastasis. However, the role of eNOS in cancer stem cell (CSC) biology and mesenchymal tumors is unknown. Results Here, we show that eNOS was significantly upregulated in VilCre ERT2 Apc fl/+ and VilCre ERT2 Apc fl/fl mouse intestinal tissue, with intense immunostaining in hyperproliferative crypts. Similarly, the more invasive VilCre ERT2 Apc fl/+ Pten fl/+ mouse model showed an overexpression of eNOS in intestinal tumors whereas this isoform was not expressed in normal tissue. However, none of the three models showed iNOS expression. Notably, when 40 human colorectal tumors were classified into different clinically relevant molecular subtypes, high eNOS expression was found in the poor relapse-free and overall survival mesenchymal subtype, whereas iNOS was absent. Furthermore, Apc fl/fl organoids overexpressed eNOS compared with wild-type organoids and NO depletion with the scavenger carboxy-PTIO (c-PTIO) decreased the proliferation and the expression of stem-cell markers, such as Lgr5, Troy, Vav3, and Slc14a1, in these intestinal organoids. Moreover, specific NO depletion also decreased the expression of CSC-related proteins in human colorectal cancer cells such as β-catenin and Bmi1, impairing the CSC phenotype. To rule out the contribution of iNOS in this effect, we established an iNOS-knockdown colorectal cancer cell line. NO-depleted cells showed a decreased capacity to form tumors and c-PTIO treatment in vivo showed an antitumoral effect in a xenograft mouse model. Conclusion Our data support that eNOS upregulation occurs after Apc loss, emerging as an unexpected potential new target in poor-prognosis mesenchymal colorectal tumors, where NO scavenging could represent an interesting therapeutic alternative to targeting the CSC subpopulation.

Keywords