Your browser doesn't support javascript.
loading
A mutation in the PRKAR1B gene drives pathological mechanisms of neurodegeneration across species.
Benjamin-Zukerman, Tal; Shimon, Gilat; Gaine, Marie E; Dakwar, Anwar; Peled, Netta; Aboraya, Mohammad; Masri-Ismail, Ashar; Safadi-Safa, Rania; Solomon, Meir; Lev-Ram, Varda; Rissman, Robert A; Mayrhofer, Johanna E; Raffeiner, Andrea; Mol, Merel O; Argue, Benney M R; McCool, Shaylah; Doan, Binh; van Swieten, John; Stefan, Eduard; Abel, Ted; Ilouz, Ronit.
Afiliação
  • Benjamin-Zukerman T; The Azrieli Faculty of Medicine, Bar Ilan University, Safed 1311502, Israel.
  • Shimon G; The Azrieli Faculty of Medicine, Bar Ilan University, Safed 1311502, Israel.
  • Gaine ME; Department of Pharmaceutical Sciences and Experimental Therapeutics, University of Iowa, Iowa City, IA 52242, USA.
  • Dakwar A; Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52242, USA.
  • Peled N; The Azrieli Faculty of Medicine, Bar Ilan University, Safed 1311502, Israel.
  • Aboraya M; The Azrieli Faculty of Medicine, Bar Ilan University, Safed 1311502, Israel.
  • Masri-Ismail A; The Azrieli Faculty of Medicine, Bar Ilan University, Safed 1311502, Israel.
  • Safadi-Safa R; The Azrieli Faculty of Medicine, Bar Ilan University, Safed 1311502, Israel.
  • Solomon M; The Azrieli Faculty of Medicine, Bar Ilan University, Safed 1311502, Israel.
  • Lev-Ram V; The Azrieli Faculty of Medicine, Bar Ilan University, Safed 1311502, Israel.
  • Rissman RA; Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA.
  • Mayrhofer JE; Department of Physiology and Neurosciences, Alzheimer's Therapeutic Research Institute, Keck School of Medicine of University of Southern California, San Diego CA 92121, USA.
  • Raffeiner A; Institute of Molecular Biology, Institute of Biochemistry and Center for Molecular Biosciences, University of Innsbruck, Innsbruck, 6020 Tyrol, Austria.
  • Mol MO; Tyrolean Cancer Research Institute (TKFI), Innsbruck, 6020 Tyrol, Austria.
  • Argue BMR; Institute of Molecular Biology, Institute of Biochemistry and Center for Molecular Biosciences, University of Innsbruck, Innsbruck, 6020 Tyrol, Austria.
  • McCool S; Tyrolean Cancer Research Institute (TKFI), Innsbruck, 6020 Tyrol, Austria.
  • Doan B; Department of Neurology and Alzheimer Center Erasmus MC, Erasmus University Medical Center, 3015 CE Rotterdam, The Netherlands.
  • van Swieten J; Department of Pharmaceutical Sciences and Experimental Therapeutics, University of Iowa, Iowa City, IA 52242, USA.
  • Stefan E; Department of Pharmaceutical Sciences and Experimental Therapeutics, University of Iowa, Iowa City, IA 52242, USA.
  • Abel T; Department of Pharmaceutical Sciences and Experimental Therapeutics, University of Iowa, Iowa City, IA 52242, USA.
  • Ilouz R; Department of Neurology and Alzheimer Center Erasmus MC, Erasmus University Medical Center, 3015 CE Rotterdam, The Netherlands.
Brain ; 2024 May 14.
Article em En | MEDLINE | ID: mdl-38743596
ABSTRACT
Protein Kinase A (PKA) neuronal function is controlled by the interaction of a regulatory (R) subunit dimer to two catalytic (C) subunits. Recently, the L50R variant in the gene encoding the RIß subunit was identified in individuals with a novel neurodegenerative disease. However, the mechanisms driving the disease phenotype remained unknown. In this study, we generated a mouse model carrying the RIß-L50R mutation to replicate the human disease phenotype and study its progression with age. We examined postmortem brains of affected individuals as well as live cell cultures. Employing biochemical assays, immunohistochemistry, and behavioral assessments, we investigated the impact of the mutation on PKA complex assembly, protein aggregation and neuronal degeneration. We reveal that RIß is an aggregation-prone protein that progressively accumulates in wildtype and Alzheimer's mouse models with age, while aggregation is accelerated in the RIß-L50R mouse model. We define RIß-L50R as a causal mutation driving an age-dependent behavioral and disease phenotype in human and mouse models. Mechanistically, this mutation disrupts RIß dimerization, leading to aggregation of its monomers. Intriguingly, interaction with the C-subunit protects the RIß-L50R from self-aggregating, in a dose-dependent manner. Furthermore, cAMP signaling induces RIß-L50R aggregation. The pathophysiological mechanism elucidated here for a newly recognized neurodegenerative disease, in which protein aggregation is the result of disrupted homodimerization, sheds light on a remarkably under-appreciated but potentially common mechanism across several neurodegenerative diseases.
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Idioma: En Ano de publicação: 2024 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Idioma: En Ano de publicação: 2024 Tipo de documento: Article