Your browser doesn't support javascript.
loading
Identification and characterization of a BRAF fusion oncoprotein with retained autoinhibitory domains.
Weinberg, Florian; Griffin, Ricarda; Fröhlich, Martina; Heining, Christoph; Braun, Sandra; Spohr, Corinna; Iconomou, Mary; Hollek, Viola; Röring, Michael; Horak, Peter; Kreutzfeldt, Simon; Warsow, Gregor; Hutter, Barbara; Uhrig, Sebastian; Neumann, Olaf; Reuss, David; Heiland, Dieter Henrik; von Kalle, Christof; Weichert, Wilko; Stenzinger, Albrecht; Brors, Benedikt; Glimm, Hanno; Fröhling, Stefan; Brummer, Tilman.
Afiliação
  • Weinberg F; Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
  • Griffin R; Centre for Biological Signalling Studies BIOSS, University of Freiburg, Freiburg, Germany.
  • Fröhlich M; Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
  • Heining C; Division of Applied Bioinformatics, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany.
  • Braun S; Department of Translational Medical Oncology, NCT Dresden, Dresden, and DKFZ, Heidelberg, Germany.
  • Spohr C; University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany.
  • Iconomou M; German Cancer Consortium (DKTK), Dresden, Germany.
  • Hollek V; Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
  • Röring M; Centre for Biological Signalling Studies BIOSS, University of Freiburg, Freiburg, Germany.
  • Horak P; Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
  • Kreutzfeldt S; Faculty of Biology, University of Freiburg, Freiburg, Germany.
  • Warsow G; Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany.
  • Hutter B; Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
  • Uhrig S; Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
  • Neumann O; Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
  • Reuss D; Department of Translational Medical Oncology, NCT Heidelberg and DKFZ, Heidelberg, Germany.
  • Heiland DH; DKTK, Heidelberg, Germany.
  • von Kalle C; Department of Translational Medical Oncology, NCT Heidelberg and DKFZ, Heidelberg, Germany.
  • Weichert W; DKTK, Heidelberg, Germany.
  • Stenzinger A; Omics IT and Data Management Core Facility, DKFZ, Heidelberg, Germany.
  • Brors B; Division of Theoretical Bioinformatics, DKFZ, Heidelberg, Germany.
  • Glimm H; Division of Applied Bioinformatics, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany.
  • Fröhling S; Division of Applied Bioinformatics, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany.
  • Brummer T; Faculty of Biosciences, Heidelberg University, Heidelberg, Germany.
Oncogene ; 39(4): 814-832, 2020 01.
Article em En | MEDLINE | ID: mdl-31558800
ABSTRACT
Fusion proteins involving the BRAF serine/threonine kinase occur in many cancers. The oncogenic potential of BRAF fusions has been attributed to the loss of critical N-terminal domains that mediate BRAF autoinhibition. We used whole-exome and RNA sequencing in a patient with glioblastoma multiforme to identify a rearrangement between TTYH3, encoding a membrane-resident, calcium-activated chloride channel, and BRAF intron 1, resulting in a TTYH3-BRAF fusion protein that retained all features essential for BRAF autoinhibition. Accordingly, the BRAF moiety of the fusion protein alone, which represents full-length BRAF without the amino acids encoded by exon 1 (BRAFΔE1), did not induce MEK/ERK phosphorylation or transformation. Likewise, neither the TTYH3 moiety of the fusion protein nor full-length TTYH3 provoked ERK pathway activity or transformation. In contrast, TTYH3-BRAF displayed increased MEK phosphorylation potential and transforming activity, which were caused by TTYH3-mediated tethering of near-full-length BRAF to the (endo)membrane system. Consistent with this mechanism, a synthetic approach, in which BRAFΔE1 was tethered to the membrane by fusing it to the cytoplasmic tail of CD8 also induced transformation. Furthermore, we demonstrate that TTYH3-BRAF signals largely independent of a functional RAS binding domain, but requires an intact BRAF dimer interface and activation loop phosphorylation sites. Cells expressing TTYH3-BRAF exhibited increased MEK/ERK signaling, which was blocked by clinically achievable concentrations of sorafenib, trametinib, and the paradox breaker PLX8394. These data provide the first example of a fully autoinhibited BRAF protein whose oncogenic potential is dictated by a distinct fusion partner and not by a structural change in BRAF itself.
Assuntos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Piridonas / Pirimidinonas / Sulfonamidas / Proteínas de Fusão Oncogênica / Glioblastoma / Proteínas Proto-Oncogênicas B-raf / Compostos Heterocíclicos com 2 Anéis Tipo de estudo: Diagnostic_studies Limite: Aged / Female / Humans Idioma: En Revista: Oncogene Ano de publicação: 2020 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Piridonas / Pirimidinonas / Sulfonamidas / Proteínas de Fusão Oncogênica / Glioblastoma / Proteínas Proto-Oncogênicas B-raf / Compostos Heterocíclicos com 2 Anéis Tipo de estudo: Diagnostic_studies Limite: Aged / Female / Humans Idioma: En Revista: Oncogene Ano de publicação: 2020 Tipo de documento: Article