Your browser doesn't support javascript.
loading
Comparative analysis of SEC61A1 mutant R236C in two patient-derived cellular platforms.
Weiand, Matthias; Sandfort, Vanessa; Nadzemova, Oksana; Schierwagen, Robert; Trebicka, Jonel; Schlevogt, Bernhard; Kabar, Iyad; Schmidt, Hartmut; Zibert, Andree.
Afiliación
  • Weiand M; Medizinische Klinik B, Universitätsklinikum Münster, Münster, Germany.
  • Sandfort V; Medizinische Klinik B, Universitätsklinikum Münster, Münster, Germany.
  • Nadzemova O; Medizinische Klinik B, Universitätsklinikum Münster, Münster, Germany.
  • Schierwagen R; Medizinische Klinik B, Universitätsklinikum Münster, Münster, Germany.
  • Trebicka J; Medizinische Klinik B, Universitätsklinikum Münster, Münster, Germany.
  • Schlevogt B; Department of Gastroenterology, Medical Center Osnabrück, Osnabrück, Germany.
  • Kabar I; Medizinische Klinik B, Universitätsklinikum Münster, Münster, Germany.
  • Schmidt H; Klinik für Gastroenterologie und Hepatologie, Uniklinik Essen, Essen, Germany.
  • Zibert A; Medizinische Klinik B (Gastroenterologie, Hepatologie, Endokrinologie, Klinische Infektiologie), Universitätsklinikum Münster, Albert-Schweitzer-Campus 1, Gebäude A14, 48149, Münster, Germany. andree.zibert@ukmuenster.de.
Sci Rep ; 14(1): 9506, 2024 04 25.
Article en En | MEDLINE | ID: mdl-38664472
ABSTRACT
SEC61A1 encodes a central protein of the mammalian translocon and dysfunction results in severe disease. Recently, mutation R236C was identified in patients having autosomal dominant polycystic liver disease (ADPLD). The molecular phenotype of R236C was assessed in two cellular platforms. Cells were immortalized by retroviral transduction of an oncogene (UCi) or reprogrammed to induced pluripotent stem cells (iPSC) that were differentiated to cholangiocyte progenitor-like cells (CPLC). UCi and CPLC were subjected to analyses of molecular pathways that were associated with development of disease. UCi displayed markers of epithelial cells, while CPLCs expressed typical markers of both cholangiocytes and hepatocytes. Cells encoding R236C showed a stable, continuous proliferation in both platforms, however growth rates were reduced as compared to wildtype control. Autophagy, cAMP synthesis, and secretion of important marker proteins were reduced in R236C-expressing cells. In addition, R236C induced increased calcium leakiness from the ER to the cytoplasm. Upon oxidative stress, R236C led to a high induction of apoptosis and necrosis. Although the grade of aberrant cellular functions differed between the two platforms, the molecular phenotype of R236C was shared suggesting that the mutation, regardless of the cell type, has a dominant impact on disease-associated pathways.
Asunto(s)

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Células Madre Pluripotentes Inducidas / Canales de Translocación SEC Límite: Humans Idioma: En Revista: Sci Rep Año: 2024 Tipo del documento: Article País de afiliación: Alemania

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Células Madre Pluripotentes Inducidas / Canales de Translocación SEC Límite: Humans Idioma: En Revista: Sci Rep Año: 2024 Tipo del documento: Article País de afiliación: Alemania