Your browser doesn't support javascript.
loading
A heterozygous p.S143P mutation in LMNA associates with proteasome dysfunction and enhanced autophagy-mediated degradation of mutant lamins A and C.
West, Gun; Turunen, Minttu; Aalto, Anna; Virtanen, Laura; Li, Song-Ping; Heliö, Tiina; Meinander, Annika; Taimen, Pekka.
Afiliación
  • West G; Institute of Biomedicine and FICAN West Cancer Centre, University of Turku, Turku, Finland.
  • Turunen M; InFLAMES Research Flagship Center, University of Turku and Åbo Akademi University, Turku, Finland.
  • Aalto A; Institute of Biomedicine and FICAN West Cancer Centre, University of Turku, Turku, Finland.
  • Virtanen L; InFLAMES Research Flagship Center, University of Turku and Åbo Akademi University, Turku, Finland.
  • Li SP; InFLAMES Research Flagship Center, University of Turku and Åbo Akademi University, Turku, Finland.
  • Heliö T; Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland.
  • Meinander A; Institute of Biomedicine and FICAN West Cancer Centre, University of Turku, Turku, Finland.
  • Taimen P; InFLAMES Research Flagship Center, University of Turku and Åbo Akademi University, Turku, Finland.
Front Cell Dev Biol ; 10: 932983, 2022.
Article en En | MEDLINE | ID: mdl-36111332
ABSTRACT
Lamins A and C are nuclear intermediate filament proteins that form a proteinaceous meshwork called lamina beneath the inner nuclear membrane. Mutations in the LMNA gene encoding lamins A and C cause a heterogenous group of inherited degenerative diseases known as laminopathies. Previous studies have revealed altered cell signaling pathways in lamin-mutant patient cells, but little is known about the fate of mutant lamins A and C within the cells. Here, we analyzed the turnover of lamins A and C in cells derived from a dilated cardiomyopathy patient with a heterozygous p.S143P mutation in LMNA. We found that transcriptional activation and mRNA levels of LMNA are increased in the primary patient fibroblasts, but the protein levels of lamins A and C remain equal in control and patient cells because of a meticulous interplay between autophagy and the ubiquitin-proteasome system (UPS). Both endogenous and ectopic expression of p.S143P lamins A and C cause significantly reduced activity of UPS and an accumulation of K48-ubiquitin chains in the nucleus. Furthermore, K48-ubiquitinated lamins A and C are degraded by compensatory enhanced autophagy, as shown by increased autophagosome formation and binding of lamins A and C to microtubule-associated protein 1A/1B-light chain 3. Finally, chaperone 4-PBA augmented protein degradation by restoring UPS activity as well as autophagy in the patient cells. In summary, our results suggest that the p.S143P-mutant lamins A and C have overloading and deleterious effects on protein degradation machinery and pharmacological interventions with compounds enhancing protein degradation may be beneficial for cell homeostasis.
Palabras clave

Texto completo: 1 Banco de datos: MEDLINE Tipo de estudio: Risk_factors_studies Idioma: En Revista: Front Cell Dev Biol Año: 2022 Tipo del documento: Article País de afiliación: Finlandia

Texto completo: 1 Banco de datos: MEDLINE Tipo de estudio: Risk_factors_studies Idioma: En Revista: Front Cell Dev Biol Año: 2022 Tipo del documento: Article País de afiliación: Finlandia