FBXL2 targets IP3R3 for ubiquitin-mediated degradation to limit Ca2+ flux to mitochondria and, consequently, apoptosis. Efficient replication of hepatitis C virus (HCV) requires geranylgeranylation of FBXL2. Here, we show that the viral protein NS5A forms a trimeric complex with IP3R3 and FBXL2, unmasking IP3R3’s degron in the absence of inositol 1,4,5-trisphosphate (IP3) stimulation. FBXL2 knockdown or expression of a stable IP3R3 mutant causes persistent Ca2+ flux and sensitizes cells to apoptosis, resulting in the inhibition of viral replication. Importantly, the effect of FBXL2 silencing is rescued by depleting IP3R3, but not p85β another established FBXL2 substrate, indicating that the anti-HCV effect of FBXL2 knockdown is largely due to IP3R3 stabilization. Finally, disruption of the FBXL2-NS5A-IP3R3 complex using somatic cell genetics or pharmacologic inhibition results in IP3R3 stabilization and suppression of HCV replication. This study reveals an IP3-independent molecular mechanism through which HCV promotes IP3R3 degradation, thereby inhibiting virus-induced apoptosis and establishing chronic infection. Kuchay et al. show that the HCV NS5A protein forms a complex with IP3R3 and FBXL2 and promotes constitutive FBXL2-mediated degradation of IP3R3. IP3R3 degradation inhibits calcium flux, mitochondrial calcium overload, and apoptosis. Thus, NS5A contributes to a cellular environment that is permissive for chronic HCV infection.

NS5A Promotes Constitutive Degradation of IP3R3 to Counteract Apoptosis Induced by Hepatitis C Virus

Giorgi, Carlotta;Pinton, Paolo;
2018

Abstract

FBXL2 targets IP3R3 for ubiquitin-mediated degradation to limit Ca2+ flux to mitochondria and, consequently, apoptosis. Efficient replication of hepatitis C virus (HCV) requires geranylgeranylation of FBXL2. Here, we show that the viral protein NS5A forms a trimeric complex with IP3R3 and FBXL2, unmasking IP3R3’s degron in the absence of inositol 1,4,5-trisphosphate (IP3) stimulation. FBXL2 knockdown or expression of a stable IP3R3 mutant causes persistent Ca2+ flux and sensitizes cells to apoptosis, resulting in the inhibition of viral replication. Importantly, the effect of FBXL2 silencing is rescued by depleting IP3R3, but not p85β another established FBXL2 substrate, indicating that the anti-HCV effect of FBXL2 knockdown is largely due to IP3R3 stabilization. Finally, disruption of the FBXL2-NS5A-IP3R3 complex using somatic cell genetics or pharmacologic inhibition results in IP3R3 stabilization and suppression of HCV replication. This study reveals an IP3-independent molecular mechanism through which HCV promotes IP3R3 degradation, thereby inhibiting virus-induced apoptosis and establishing chronic infection. Kuchay et al. show that the HCV NS5A protein forms a complex with IP3R3 and FBXL2 and promotes constitutive FBXL2-mediated degradation of IP3R3. IP3R3 degradation inhibits calcium flux, mitochondrial calcium overload, and apoptosis. Thus, NS5A contributes to a cellular environment that is permissive for chronic HCV infection.
2018
Kuchay, Shafi; Saeed, Mohsan; Giorgi, Carlotta; Li, Jie; Hoffmann, Hans-Heinrich; Pinton, Paolo; Rice, Charles M.; Pagano, Michele
File in questo prodotto:
File Dimensione Formato  
269.pdf

accesso aperto

Descrizione: Full text editoriale
Tipologia: Full text (versione editoriale)
Licenza: Creative commons
Dimensione 1.68 MB
Formato Adobe PDF
1.68 MB Adobe PDF Visualizza/Apri

I documenti in SFERA sono protetti da copyright e tutti i diritti sono riservati, salvo diversa indicazione.

Utilizza questo identificativo per citare o creare un link a questo documento: https://hdl.handle.net/11392/2400158
Citazioni
  • ???jsp.display-item.citation.pmc??? 13
  • Scopus 18
  • ???jsp.display-item.citation.isi??? 19
social impact