Please use this identifier to cite or link to this item: http://hdl.handle.net/2445/175351
Title: Stimulation of soluble guanylate cyclase exerts antiinflammatory actions in the liver through a VASP/NF-κB/NLRP3 inflammasome circuit
Author: Flores Costa, Roger
Duran Güell, Marta
Casulleras, Mireia
López Vicario, Cristina
Alcaraz-Quiles, José
Díaz Lorca, Maria Alba
Lozano Salvatella, Juan José
Titos Rodríguez, Esther
Hall, Katherine
Sarno, Renee
Masferrer, Jaime L.
Clària i Enrich, Joan
Keywords: Hepatitis
Malalties del fetge
Inflamació
Hepatitis
Liver diseases
Inflammation
Issue Date: 26-Oct-2020
Publisher: National Academy of Sciences
Abstract: Soluble guanylate cyclase (sGC) catalyzes the conversion of guanosine triphosphate into cyclic guanosine-3',5'-monophosphate, a key second messenger in cell signaling and tissue homeostasis. It was recently demonstrated that sGC stimulation is associated with a marked antiinflammatory effect in the liver of mice with experimental nonalcoholic steatohepatitis (NASH). Here, we investigated the mechanisms underlying the antiinflammatory effect of the sGC stimulator praliciguat (PRL) in the liver. Therapeutic administration of PRL exerted antiinflammatory and antifibrotic actions in mice with choline-deficient l-amino acid-defined high-fat diet-induced NASH. The PRL antiinflammatory effect was associated with lower F4/80- and CX3CR1-positive macrophage infiltration into the liver in parallel with lower Ly6CHigh- and higher Ly6CLow-expressing monocytes in peripheral circulation. The PRL antiinflammatory effect was also associated with suppression of hepatic levels of interleukin (IL)-1β, NLPR3 (NACHT, LRR, and PYD domain-containing protein 3), ASC (apoptosis-associated speck-like protein containing a caspase-recruitment domain), and active cleaved-caspase-1, which are components of the NLRP3 inflammasome. In Kupffer cells challenged with the classical inflammasome model of lipopolysaccharide plus adenosine triphosphate, PRL inhibited the priming (expression of Il1b and Nlrp3) and blocked the release of mature IL-1β. Mechanistically, PRL induced the protein kinase G (PKG)-mediated phosphorylation of the VASP (vasodilator-stimulated phosphoprotein) Ser239 residue which, in turn, reduced nuclear factor-κB (NF-κB) activity and Il1b and Nlrp3 gene transcription. PRL also reduced active cleaved-caspase-1 levels independent of pannexin-1 activity. These data indicate that sGC stimulation with PRL exerts antiinflammatory actions in the liver through mechanisms related to a PKG/VASP/NF-κB/NLRP3 inflammasome circuit.
Note: Versió postprint del document publicat a: https://doi.org/10.1073/pnas.2000466117
It is part of: Proceedings of the National Academy of Sciences of the United States of America - PNAS, 2020, vol. 117, num. 45, p. 28263-28274
URI: http://hdl.handle.net/2445/175351
Related resource: https://doi.org/10.1073/pnas.2000466117
ISSN: 0027-8424
Appears in Collections:Articles publicats en revistes (IDIBAPS: Institut d'investigacions Biomèdiques August Pi i Sunyer)
Articles publicats en revistes (Biomedicina)

Files in This Item:
File Description SizeFormat 
709164.pdf2.6 MBAdobe PDFView/Open


Items in DSpace are protected by copyright, with all rights reserved, unless otherwise indicated.