PLoS ONE 2015-01-01

High-Throughput Assay Development for Cystine-Glutamate Antiporter (xc-) Highlights Faster Cystine Uptake than Glutamate Release in Glioma Cells.

Ajit G Thomas, Rita Sattler, Karen Tendyke, Kara A Loiacono, Hans Hansen, Vishal Sahni, Yutaka Hashizume, Camilo Rojas, Barbara S Slusher

Index: PLoS ONE 10 , e0127785, (2015)

Full Text: HTML

Abstract

The cystine-glutamate antiporter (system xc-) is a Na+-independent amino acid transporter that exchanges extracellular cystine for intracellular glutamate. It is thought to play a critical role in cellular redox processes through regulation of intracellular glutathione synthesis via cystine uptake. In gliomas, system xc- expression is universally up-regulated while that of glutamate transporters down-regulated, leading to a progressive accumulation of extracellular glutamate and excitotoxic cell death of the surrounding non-tumorous tissue. Additionally, up-regulation of system xc- in activated microglia has been implicated in the pathogenesis of several neurodegenerative disorders mediated by excess glutamate. Consequently, system xc- is a new drug target for brain cancer and neuroinflammatory diseases associated with excess extracellular glutamate. Unfortunately no potent and selective small molecule system xc- inhibitors exist and to our knowledge, no high throughput screening (HTS) assay has been developed to identify new scaffolds for inhibitor design. To develop such an assay, various neuronal and non-neuronal human cells were evaluated as sources of system xc-. Human glioma cells were chosen based on their high system xc- activity. Using these cells, [14C]-cystine uptake and cystine-induced glutamate release assays were characterized and optimized with respect to cystine and protein concentrations and time of incubation. A pilot screen of the LOPAC/NINDS libraries using glutamate release demonstrated that the logistics of the assay were in place but unfortunately, did not yield meaningful pharmacophores. A larger, HTS campaign using the 384-well cystine-induced glutamate release as primary assay and the 96-well 14C-cystine uptake as confirmatory assay is currently underway. Unexpectedly, we observed that the rate of cystine uptake was significantly faster than the rate of glutamate release in human glioma cells. This was in contrast to the same rates of cystine uptake and glutamate release previously reported in normal human fibroblast cells.


Related Compounds

Related Articles:

Aptamer-based polyvalent ligands for regulated cell attachment on the hydrogel surface.

2015-04-13

[Biomacromolecules 16(4) , 1382-9, (2015)]

Osmoregulatory bicarbonate secretion exploits H(+)-sensitive haemoglobins to autoregulate intestinal O2 delivery in euryhaline teleosts.

2014-10-01

[J. Comp. Physiol. B, Biochem. Syst. Environ. Physiol. 184(7) , 865-76, (2014)]

Polymerization of affinity ligands on a surface for enhanced ligand display and cell binding.

2014-12-08

[Biomacromolecules 15(12) , 4561-9, (2014)]

Continuous syntheses of Pd@Pt and Cu@Ag core-shell nanoparticles using microwave-assisted core particle formation coupled with galvanic metal displacement.

2014-08-07

[Nanoscale 6(15) , 8720-5, (2014)]

Effect of (2)H and (18)O water isotopes in kinesin-1 gliding assay.

2014-01-01

[PeerJ 2 , e284, (2014)]

More Articles...