KRIT1 is a scaffolding protein that regulates multiple molecular mechanisms, including cell-cell and cell-matrix adhesion and redox homeostasis and signaling. However, rather little is known about how KRIT1 is itself regulated. KRIT1 is found in both the cytoplasm and the nucleus, yet the upstream signaling proteins and mechanisms that regulate KRIT1 nucleocytoplasmic shuttling are not well understood. Here, we identify a key role for protein kinase C (PKC). In particular, we found that PKC activation promotes the redox-dependent cytoplasmic localization of KRIT1, whereas inhibition of PKC or treatment with the antioxidant N-acetylcysteine leads to KRIT1 nuclear accumulation. Moreover, we demonstrated that the N-terminal region of KRIT1 is crucial for the ability of PKC to regulate KRIT1 nucleocytoplasmic shuttling, and may be a target for PKC-dependent regulatory phosphorylation events. Finally, we found that silencing of PKCα, but not PKCδ, inhibits phorbol-12-myristate-13-acetate (PMA)-induced cytoplasmic enrichment of KRIT1, suggesting a major role for PKCα in regulating KRIT1 nucleocytoplasmic shuttling. Overall, our findings identify PKCα as a novel regulator of KRIT1 subcellular compartmentalization, thus shedding new light on the physiopathological functions of this protein.

Protein kinase Cα regulates the nucleocytoplasmic shuttling of KRIT1

Perrelli, Andrea
Co-first
;
Retta, Saverio Francesco
Last
2021-01-01

Abstract

KRIT1 is a scaffolding protein that regulates multiple molecular mechanisms, including cell-cell and cell-matrix adhesion and redox homeostasis and signaling. However, rather little is known about how KRIT1 is itself regulated. KRIT1 is found in both the cytoplasm and the nucleus, yet the upstream signaling proteins and mechanisms that regulate KRIT1 nucleocytoplasmic shuttling are not well understood. Here, we identify a key role for protein kinase C (PKC). In particular, we found that PKC activation promotes the redox-dependent cytoplasmic localization of KRIT1, whereas inhibition of PKC or treatment with the antioxidant N-acetylcysteine leads to KRIT1 nuclear accumulation. Moreover, we demonstrated that the N-terminal region of KRIT1 is crucial for the ability of PKC to regulate KRIT1 nucleocytoplasmic shuttling, and may be a target for PKC-dependent regulatory phosphorylation events. Finally, we found that silencing of PKCα, but not PKCδ, inhibits phorbol-12-myristate-13-acetate (PMA)-induced cytoplasmic enrichment of KRIT1, suggesting a major role for PKCα in regulating KRIT1 nucleocytoplasmic shuttling. Overall, our findings identify PKCα as a novel regulator of KRIT1 subcellular compartmentalization, thus shedding new light on the physiopathological functions of this protein.
2021
134
3
jcs.250217
-
https://pubmed.ncbi.nlm.nih.gov/33443102/
https://jcs.biologists.org/content/134/3/jcs250217.long
https://jcs.biologists.org/content/joces/134/3/jcs250217.full.pdf?with-ds=yes
CCM1/KRIT1; Cerebral Cavernous Malformation (CCM); Nucleocytoplasmic shuttling; PKC signaling; PKCα; PKCδ; Phorbol esters; Phosphoproteomics; Redox signaling
De Luca, Elisa; Perrelli, Andrea; Swamy, Harsha; Nitti, Mariapaola; Passalacqua, Mario; Furfaro, Anna Lisa; Salzano, Anna Maria; Scaloni, Andrea; Glad...espandi
File in questo prodotto:
File Dimensione Formato  
2021 JCS - Protein kinase Cα regulates the nucleocytoplasmic shuttling of KRIT1 (+SI).pdf

Accesso aperto

Descrizione: Articolo principale
Tipo di file: PDF EDITORIALE
Dimensione 8.84 MB
Formato Adobe PDF
8.84 MB Adobe PDF Visualizza/Apri

I documenti in IRIS 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/2318/1770702
Citazioni
  • ???jsp.display-item.citation.pmc??? 8
  • Scopus 8
  • ???jsp.display-item.citation.isi??? 9
social impact