[HTML][HTML] Antiinflammatory adaptation to hypoxia through adenosine-mediated cullin-1 deneddylation

J Khoury, JC Ibla, AS Neish… - The Journal of clinical …, 2007 - Am Soc Clin Investig
J Khoury, JC Ibla, AS Neish, SP Colgan
The Journal of clinical investigation, 2007Am Soc Clin Investig
A major adaptive pathway for hypoxia is hypoxic preconditioning (HPC), a form of
endogenous protection that renders cells tolerant to severe challenges of hypoxia. We
sought to define the antiinflammatory properties of HPC. cDNA microarray analysis of lung
tissue from mice subjected to hypoxia or HPC identified a cluster of NF-κB–regulated genes
whose expression is attenuated by HPC. Studies using an NF-κB luciferase reporter assay
confirmed a significant suppression of NF-κB activation during HPC. HPC-elicited activity …
A major adaptive pathway for hypoxia is hypoxic preconditioning (HPC), a form of endogenous protection that renders cells tolerant to severe challenges of hypoxia. We sought to define the antiinflammatory properties of HPC. cDNA microarray analysis of lung tissue from mice subjected to hypoxia or HPC identified a cluster of NF-κB–regulated genes whose expression is attenuated by HPC. Studies using an NF-κB luciferase reporter assay confirmed a significant suppression of NF-κB activation during HPC. HPC-elicited activity was conferrable, as a soluble supernatant from HPC-treated cells, and the active fraction was purified and identified as adenosine (Ado). Guided by recent studies demonstrating bacterial inhibition of NF-κB through cullin-1 (Cul-1) deneddylation, we found a dose-dependent deneddylation of Cul-1 by Ado receptor stimulation predominantly mediated by the Ado A2B receptor subtype. Further, siRNA-mediated repression of CSN5, a subunit of the COP9 signalosome responsible for deneddylation of Cul-1, partially reversed HPC-mediated inhibition of NF-κB. Cul-1 deneddylation was evident in a murine model of HPC and lost in animals lacking extracellular Ado (Cd73–/– mice). Taken together, these results demonstrate that HPC induces extracellular accumulation of Ado and suppresses NF-κB activity through deneddylation of Cul-1. These results define a molecular regulatory pathway by which Ado provides potent antiinflammatory properties.
The Journal of Clinical Investigation