Autophagy inhibitor 3-methyladenine protects against endothelial cell barrier dysfunction in acute lung injury

SA Slavin, A Leonard, V Grose… - American Journal of …, 2018 - journals.physiology.org
SA Slavin, A Leonard, V Grose, F Fazal, A Rahman
American Journal of Physiology-Lung Cellular and Molecular …, 2018journals.physiology.org
Autophagy is an evolutionarily conserved cellular process that facilitates the continuous
recycling of intracellular components (organelles and proteins) and provides an alternative
source of energy when nutrients are scarce. Recent studies have implicated autophagy in
many disorders, including pulmonary diseases. However, the role of autophagy in
endothelial cell (EC) barrier dysfunction and its relevance in the context of acute lung injury
(ALI) remain uncertain. Here, we provide evidence that autophagy is a critical component of …
Autophagy is an evolutionarily conserved cellular process that facilitates the continuous recycling of intracellular components (organelles and proteins) and provides an alternative source of energy when nutrients are scarce. Recent studies have implicated autophagy in many disorders, including pulmonary diseases. However, the role of autophagy in endothelial cell (EC) barrier dysfunction and its relevance in the context of acute lung injury (ALI) remain uncertain. Here, we provide evidence that autophagy is a critical component of EC barrier disruption in ALI. Using an aerosolized bacterial lipopolysaccharide (LPS) inhalation mouse model of ALI, we found that administration of the autophagy inhibitor 3-methyladenine (3-MA), either prophylactically or therapeutically, markedly reduced lung vascular leakage and tissue edema. 3-MA was also effective in reducing the levels of proinflammatory mediators and lung neutrophil sequestration induced by LPS. To test the possibility that autophagy in EC could contribute to lung vascular injury, we addressed its role in the mechanism of EC barrier disruption. Knockdown of ATG5, an essential regulator of autophagy, attenuated thrombin-induced EC barrier disruption, confirming the involvement of autophagy in the response. Similarly, exposure of cells to 3-MA, either before or after thrombin, protected against EC barrier dysfunction by inhibiting the cleavage and loss of vascular endothelial cadherin at adherens junctions, as well as formation of actin stress fibers. 3-MA also reversed LPS-induced EC barrier disruption. Together, these data imply a role of autophagy in lung vascular injury and reveal the protective and therapeutic utility of 3-MA against ALI.
American Physiological Society