4.8 Article

Antiinflammatory adaptation to hypoxia through adenosine-mediated cullin-1 deneddylation

Journal

JOURNAL OF CLINICAL INVESTIGATION
Volume 117, Issue 3, Pages 703-711

Publisher

AMER SOC CLINICAL INVESTIGATION INC
DOI: 10.1172/JCI30049

Keywords

-

Funding

  1. NHLBI NIH HHS [R01 HL060569, HL60569] Funding Source: Medline
  2. NIDCR NIH HHS [P01 DE013499, DE13499] Funding Source: Medline
  3. NIDDK NIH HHS [R01 DK050189, DK50189, R37 DK050189, R29 DK050189] Funding Source: Medline
  4. NATIONAL HEART, LUNG, AND BLOOD INSTITUTE [R01HL060569] Funding Source: NIH RePORTER
  5. NATIONAL INSTITUTE OF DENTAL &CRANIOFACIAL RESEARCH [P01DE013499] Funding Source: NIH RePORTER
  6. NATIONAL INSTITUTE OF DIABETES AND DIGESTIVE AND KIDNEY DISEASES [R29DK050189, R37DK050189, R01DK050189] Funding Source: NIH RePORTER

Ask authors/readers for more resources

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 and inflammatory properties of HPC. cDNA microarray analysis of lung tissue from mice subjected to hypoxia or HPC identified a cluster of NF-kappa B-regulated genes whose expression is attenuated by HPC. Studies using an NF-kappa B luciferase reporter assay confirmed a significant suppression of NF-kappa B 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-kappa B 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-KB. 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-KB activity through deneddylation of Cul-1. These results define a molecular regulatory pathway by which Ado provides potent and inflammatory properties.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available