Increased fatty acid oxidation promotes mitochondrial ROS (mtROS) production and Ca 2+ entry via TRPV4 in microvascular endothelial cells in PAH

2020 ◽  
Vol 34 (S1) ◽  
pp. 1-1 ◽  
Author(s):  
Karthik Suresh ◽  
Laura Servinsky ◽  
Nicolas Philip ◽  
Haiyang Jiang ◽  
John Huetsch ◽  
...  
2019 ◽  
Vol 316 (5) ◽  
pp. L740-L750 ◽  
Author(s):  
Hira Raheel ◽  
Siavash Ghaffari ◽  
Negar Khosraviani ◽  
Victoria Mintsopoulos ◽  
Derek Auyeung ◽  
...  

In healthy blood vessels, albumin crosses the endothelium to leave the circulation by transcytosis. However, little is known about the regulation of albumin transcytosis or how it differs in different tissues; its physiological purpose is also unclear. Using total internal reflection fluorescence microscopy, we quantified transcytosis of albumin across primary human microvascular endothelial cells from both lung and skin. We then validated our in vitro findings using a tissue-specific knockout mouse model. We observed that albumin transcytosis was saturable in the skin but not the lung microvascular endothelial cells, implicating a receptor-mediated process. We identified the scavenger receptor CD36 as being both necessary and sufficient for albumin transcytosis across dermal microvascular endothelium, in contrast to the lung where macropinocytosis dominated. Mutations in the apical helical bundle of CD36 prevented albumin internalization by cells. Mice deficient in CD36 specifically in endothelial cells exhibited lower basal permeability to albumin and less basal tissue edema in the skin but not in the lung. Finally, these mice also exhibited a smaller subcutaneous fat layer despite having identical total body weights and circulating fatty acid levels as wild-type animals. In conclusion, CD36 mediates albumin transcytosis in the skin but not the lung. Albumin transcytosis may serve to regulate fatty acid delivery from the circulation to tissues.


2019 ◽  
Vol 33 (S1) ◽  
Author(s):  
Karthik Suresh ◽  
Laura Servinsky ◽  
Nicolas Philip ◽  
Zack Hill ◽  
Haiyang Jiang ◽  
...  

Author(s):  
Laurel Yong-Hwa Lee ◽  
William M. Oldham ◽  
Huamei He ◽  
Ruisheng Wang ◽  
Ryan Mulhern ◽  
...  

Background: Endothelial cells depend on glycolysis for much of their energy production. Impaired endothelial glycolysis has been associated with various vascular pathobiologies, including impaired angiogenesis and atherogenesis. Interferon-gamma (IFN- γ )-producing CD4 + and CD8 + T-lymphocytes have been identified as the predominant pathologic cell subsets in human atherosclerotic plaques. While the immunological consequences of these cells have been extensively evaluated, their IFN- γ -mediated metabolic effects on endothelial cells remain unknown. The purpose of this study was to determine the metabolic consequences of the T-lymphocyte cytokine, IFN- γ , on human coronary artery endothelial cells (HCAEC). Methods: The metabolic effects of IFN- γ on primary HCAEC were assessed by unbiased transcriptomic and metabolomic analyses combined with real-time extracellular flux analyses and molecular mechanistic studies. Cellular phenotypic correlations were made by measuring altered endothelial intracellular cyclic guanosine monophosphate (cGMP) content, wound healing capacity, and adhesion molecule expression. Results: IFN- γ exposure inhibited basal glycolysis of quiescent primary HCAEC by 20% through the global transcriptional suppression of glycolytic enzymes resulting from decreased basal hypoxia inducible factor 1α (HIF1α) nuclear availability in normoxia. The decrease in HIF1α activity was a consequence of IFN- γ -induced tryptophan catabolism resulting in ARNT (aryl hydrocarbon receptor nuclear translocator)/HIF1β sequestration by the kynurenine-activated aryl hydrocarbon receptor (AHR). Additionally, IFN- γ resulted in a 23% depletion of intracellular NAD + in HCAEC. This altered glucose metabolism was met with concomitant activation of fatty acid oxidation, which augmented its contribution to intracellular ATP balance by over 20%. These metabolic derangements were associated with adverse endothelial phenotypic changes, including decreased basal intracellular cGMP, impaired endothelial migration, and a switch to a pro-inflammatory state. Conclusions: IFN- γ impairs endothelial glucose metabolism via altered tryptophan catabolism destabilizing HIF1, depletes NAD + , and results in a metabolic shift toward increased fatty acid oxidation. This work suggests a novel mechanistic basis for pathologic T-lymphocyte-endothelial interactions in atherosclerosis mediated by IFN- γ , linking endothelial glucose, tryptophan, and fatty acid metabolism with NAD(H) and ATP generation, and their adverse endothelial functional consequences.


Sign in / Sign up

Export Citation Format

Share Document