scholarly journals Effects of different fatty acid profiles on markers of endothelial function in cultured human dermal microvascular endothelial cells

2010 ◽  
Vol 69 (OCE6) ◽  
Author(s):  
S. C. Cottin ◽  
R. C. Siow ◽  
T. A. Sanders ◽  
W. L. Hall
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.


Author(s):  
Birger Tielemans ◽  
Rik Gijsbers ◽  
Annelies Michiels ◽  
Allard Wagenaar ◽  
Ricard Farre Marti ◽  
...  

2017 ◽  
Vol 312 (1) ◽  
pp. L143-L153 ◽  
Author(s):  
Karthik Suresh ◽  
Laura Servinsky ◽  
Jose Reyes ◽  
Clark Undem ◽  
Joel Zaldumbide ◽  
...  

Elevated levels of reactive oxygen species and intracellular Ca2+ play a key role in endothelial barrier dysfunction in acute lung injury. We previously showed that H2O2-induced increases in intracellular calcium concentrations ([Ca2+]i) in lung microvascular endothelial cells (LMVECs) involve the membrane Ca2+ channel, transient receptor potential vanilloid-4 (TRPV4) and that inhibiting this channel attenuated H2O2-induced barrier disruption in vitro. We also showed that phosphorylation of TRPV4 by the Src family kinase, Fyn, contributes to H2O2-induced Ca2+ influx in LMVEC. In endothelial cells, Fyn is tethered to the cell membrane by CD36, a fatty acid transporter. In this study, we assessed the effect of genetic loss or pharmacological inhibition of CD36 on Ca2+ responses to H2O2. H2O2-induced Ca2+ influx was attenuated in LMVEC isolated from mice lacking CD36 ( CD36−/−). TRPV4 expression and function was unchanged in LMVEC isolated from wild-type (WT) and CD36−/− mice, as well as mice with deficiency for Fyn ( Fyn−/−). TRPV4 immunoprecipitated with Fyn, but this interaction was decreased in CD36−/− LMVEC. The amount of phosphorylated TRPV4 was decreased in LMVEC from CD36−/− mice compared with WT controls. Loss of CD36 altered subcellular localization of Fyn, while inhibition of CD36 fatty acid transport with succinimidyl oleate did not attenuate H2O2-induced Ca2+ influx. Lastly, we found that CD36−/− mice were protected from ischemia-reperfusion injury in vivo. In conclusion, our data suggest that CD36 plays an important role in H2O2-mediated lung injury and that the mechanism may involve CD36-dependent scaffolding of Fyn to the cell membrane to facilitate TRPV4 phosphorylation.


Sign in / Sign up

Export Citation Format

Share Document