scholarly journals P-Glycoprotein Transport of Neurotoxic Pesticides

2015 ◽  
Vol 355 (1) ◽  
pp. 99-107 ◽  
Author(s):  
Sarah E. Lacher ◽  
Kasse Skagen ◽  
Joachim Veit ◽  
Rachel Dalton ◽  
Erica L. Woodahl
2013 ◽  
Vol 102 (10) ◽  
pp. 3830-3837 ◽  
Author(s):  
Analia Novak ◽  
Griselda Delli Carpini ◽  
María Laura Ruiz ◽  
Marcelo G. Luquita ◽  
Modesto C. Rubio ◽  
...  

2011 ◽  
Vol 338 (1) ◽  
pp. 372-380 ◽  
Author(s):  
Mark Jean Gnoth ◽  
Ulf Buetehorn ◽  
Uwe Muenster ◽  
Thomas Schwarz ◽  
Steffen Sandmann

2012 ◽  
Vol 33 (3) ◽  
pp. 381-388 ◽  
Author(s):  
Tara A Cartwright ◽  
Christopher R Campos ◽  
Ronald E Cannon ◽  
David S Miller

At the blood–brain and blood–spinal cord barriers, P-glycoprotein, an ATP-driven drug efflux pump, is a major obstacle to central nervous system (CNS) pharmacotherapy. Recently, we showed that signaling through tumor necrosis factor-α (TNF-α), sphingolipids, and sphingosine-1-phosphate receptor 1 (S1PR1) rapidly and reversibly reduced basal P-glycoprotein transport activity in the rat blood–brain barrier. The present study extends those findings to the mouse blood–brain and blood–spinal cord barriers and, importantly, identifies multidrug resistance-associated protein 1 (Mrp1, Abcc1) as the transporter that mediates S1P efflux from brain and spinal cord endothelial cells. In brain and spinal cord capillaries isolated from wild-type mice, TNF-α, sphingosine, S1P, the S1PR agonist fingolimod (FTY720), and its active, phosphorylated metabolite, FTY720P, reduced P-glycoprotein transport activity; these effects were abolished by a specific S1PR1 antagonist. In brain and spinal cord capillaries isolated from Mrp1-null mice, neither TNF-α nor sphingosine nor FTY720 reduced P-glycoprotein transport activity. However, S1P and FTY720P had the same S1PR1-dependent effects on transport activity as in capillaries from wild-type mice. Thus, deletion of Mrp1 alone terminated endogenous signaling to S1PR1. These results identify Mrp1 as the transporter essential for S1P efflux from the endothelial cells and thus for inside-out S1P signaling to P-glycoprotein at the blood–brain and blood–spinal cord barriers.


2008 ◽  
Vol 356 (1-2) ◽  
pp. 351-353 ◽  
Author(s):  
Jiangeng Huang ◽  
Luqin Si ◽  
Lingli Jiang ◽  
Zhaoze Fan ◽  
Jun Qiu ◽  
...  

2013 ◽  
Vol 67 (4) ◽  
pp. 390-394 ◽  
Author(s):  
S-Y Wang ◽  
K-M Duan ◽  
Y Li ◽  
Y Mei ◽  
H Sheng ◽  
...  

2012 ◽  
Vol 26 (S1) ◽  
Author(s):  
David S. Miller ◽  
Christopher R. Campos ◽  
Brian T. Hawkins ◽  
Ronald Cannon

Biochemistry ◽  
1995 ◽  
Vol 34 (38) ◽  
pp. 12210-12220 ◽  
Author(s):  
David Piwnica-Worms ◽  
Vallabhaneni V. Rao ◽  
James F. Kronauge ◽  
James M. Croop

2011 ◽  
Vol 31 (6) ◽  
pp. 1371-1375 ◽  
Author(s):  
Xueqian Wang ◽  
Brian T Hawkins ◽  
David S Miller

Upregulation of blood-brain barrier (BBB) P-glycoprotein expression causes central nervous system (CNS) pharmacoresistance. However, activation of BBB protein kinase C-β1 (PKC-β1) rapidly reduces basal P-glycoprotein transport activity. We tested whether PKC-β1 activation would reverse CNS drug resistance caused by dioxin acting through aryl hydrocarbon receptor. A selective PKC-β1 agonist abolished the increase in P-glycoprotein activity induced by dioxin in isolated rat brain capillaries and reversed the effect of dioxin on brain uptake of verapamil in dioxin-dosed rats. Thus, targeting BBB PKC-β1 may be an effective strategy to improve drug delivery to the brain, even in drug-resistant individuals.


Sign in / Sign up

Export Citation Format

Share Document