Somatic Gain of KRAS Function in the Endothelium Is Sufficient to Cause Vascular Malformations That Require MEK but Not PI3K Signaling

Circ Res. 2020 Aug 28;127(6):727-743. doi: 10.1161/CIRCRESAHA.119.316500. Epub 2020 Jun 17.

Abstract

Rationale: We previously identified somatic activating mutations in the KRAS (Kirsten rat sarcoma viral oncogene homologue) gene in the endothelium of the majority of human sporadic brain arteriovenous malformations; a disorder characterized by direct connections between arteries and veins. However, whether this genetic abnormality alone is sufficient for lesion formation, as well as how active KRAS signaling contributes to arteriovenous malformations, remains unknown.

Objective: To establish the first in vivo models of somatic KRAS gain of function in the endothelium in both mice and zebrafish to directly observe the phenotypic consequences of constitutive KRAS activity at a cellular level in vivo, and to test potential therapeutic interventions for arteriovenous malformations.

Methods and results: Using both postnatal and adult mice, as well as embryonic zebrafish, we demonstrate that endothelial-specific gain of function mutations in Kras (G12D or G12V) are sufficient to induce brain arteriovenous malformations. Active KRAS signaling leads to altered endothelial cell morphogenesis and increased cell size, ectopic sprouting, expanded vessel lumen diameter, and direct connections between arteries and veins. Furthermore, we show that these lesions are not associated with altered endothelial growth dynamics or a lack of proper arteriovenous identity but instead seem to feature exuberant angiogenic signaling. Finally, we demonstrate that KRAS-dependent arteriovenous malformations in zebrafish are refractory to inhibition of the downstream effector PI3K but instead require active MEK (mitogen-activated protein kinase kinase 1) signaling.

Conclusions: We demonstrate that active KRAS expression in the endothelium is sufficient for brain arteriovenous malformations, even in the setting of uninjured adult vasculature. Furthermore, the finding that KRAS-dependent lesions are reversible in zebrafish suggests that MEK inhibition may represent a promising therapeutic treatment for arteriovenous malformation patients. Graphical Abstract: A graphical abstract is available for this article.

Keywords: brain; cell size; endothelium, vascular; models, animal; vascular disease.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, Non-U.S. Gov't
  • Research Support, U.S. Gov't, Non-P.H.S.
  • Video-Audio Media

MeSH terms

  • Animals
  • Cells, Cultured
  • Disease Models, Animal
  • Endothelial Cells / enzymology*
  • Endothelial Cells / pathology
  • Female
  • Gain of Function Mutation*
  • Genetic Predisposition to Disease
  • Human Umbilical Vein Endothelial Cells / enzymology
  • Human Umbilical Vein Endothelial Cells / pathology
  • Humans
  • Intracranial Arteriovenous Malformations / enzymology
  • Intracranial Arteriovenous Malformations / genetics*
  • Intracranial Arteriovenous Malformations / pathology
  • Intracranial Hemorrhages / enzymology
  • Intracranial Hemorrhages / genetics
  • Intracranial Hemorrhages / pathology
  • MAP Kinase Kinase 1 / antagonists & inhibitors
  • MAP Kinase Kinase 1 / metabolism*
  • Male
  • Mice, Transgenic
  • Permeability
  • Phenotype
  • Phosphatidylinositol 3-Kinase / metabolism
  • Phosphoinositide-3 Kinase Inhibitors / pharmacology
  • Proto-Oncogene Proteins p21(ras) / genetics*
  • Signal Transduction
  • Zebrafish / embryology
  • Zebrafish / genetics
  • Zebrafish Proteins

Substances

  • KRAS protein, human
  • Phosphoinositide-3 Kinase Inhibitors
  • Zebrafish Proteins
  • Phosphatidylinositol 3-Kinase
  • MAP Kinase Kinase 1
  • MAP2K1 protein, human
  • MAP2K1 protein, zebrafish
  • Map2k1 protein, mouse
  • Hras protein, mouse
  • Kras protein, zebrafish
  • Proto-Oncogene Proteins p21(ras)