English
 
Help Privacy Policy Disclaimer
  Advanced SearchBrowse

Item

ITEM ACTIONSEXPORT

Released

Journal Article

TRAF2 Is a Novel Ubiquitin E3 Ligase for the Na,K-ATPase beta-Subunit That Drives Alveolar Epithelial Dysfunction in Hypercapnia

MPS-Authors
/persons/resource/persons248863

Herold,  Susanne
Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Max Planck Society;

/persons/resource/persons224311

Morty,  Rory E.
Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Max Planck Society;

/persons/resource/persons224334

Seeger,  Werner
Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Max Planck Society;

/persons/resource/persons248912

Vadasz,  Istvan
Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Max Planck Society;

External Resource
No external resources are shared
Fulltext (restricted access)
There are currently no full texts shared for your IP range.
Fulltext (public)
There are no public fulltexts stored in PuRe
Supplementary Material (public)
There is no public supplementary material available
Citation

Gabrielli, N. M., Mazzocchi, L. C., Kryvenko, V., Tello, K., Herold, S., Morty, R. E., et al. (2021). TRAF2 Is a Novel Ubiquitin E3 Ligase for the Na,K-ATPase beta-Subunit That Drives Alveolar Epithelial Dysfunction in Hypercapnia. FRONTIERS IN CELL AND DEVELOPMENTAL BIOLOGY, 9: 689983. doi:10.3389/fcell.2021.689983.


Cite as: https://hdl.handle.net/21.11116/0000-0008-F5E0-6
Abstract
Several acute and chronic lung diseases are associated with alveolar hypoventilation leading to accumulation of CO2 (hypercapnia). The beta-subunit of the Na,K-ATPase plays a pivotal role in maintaining epithelial integrity by functioning as a cell adhesion molecule and regulating cell surface stability of the catalytic alpha-subunit of the transporter, thereby, maintaining optimal alveolar fluid balance. Here, we identified the E3 ubiquitin ligase for the Na, K-ATPase beta-subunit, which promoted polyubiquitination, subsequent endocytosis and proteasomal degradation of the protein upon exposure of alveolar epithelial cells to elevated CO2 levels, thus impairing alveolar integrity. Ubiquitination of the Na, K-ATPase beta-subunit required lysine 5 and 7 and mutating these residues (but not other lysines) prevented trafficking of Na,K-ATPase from the plasma membrane and stabilized the protein upon hypercapnia. Furthermore, ubiquitination of the Na, K-ATPase beta-subunit was dependent on prior phosphorylation at serine 11 by protein kinase C (PKC)-zeta. Using a protein microarray, we identified the tumor necrosis factor receptor-associated factor 2 (TRAF2) as the E3 ligase driving ubiquitination of the Na, K-ATPase beta-subunit upon hypercapnia. Of note, prevention of Na, K-ATPase beta-subunit ubiquitination was necessary and sufficient to restore the formation of cell-cell junctions under hypercapnic conditions. These results suggest that a hypercapnic environment in the lung may lead to persistent epithelial dysfunction in affected patients. As such, the identification of the E3 ligase for the Na, K-ATPase may provide a novel therapeutic target, to be employed in patients with acute or chronic hypercapnic respiratory failure, aiming to restore alveolar epithelial integrity.