Your browser doesn't support javascript.
loading
KLC1-ROS1 Fusion Exerts Oncogenic Properties of Glioma Cells via Specific Activation of JAK-STAT Pathway.
Fujii, Takashi; Nakano, Yoshiko; Hagita, Daichi; Onishi, Nobuyuki; Endo, Arumu; Nakagawa, Masaya; Yoshiura, Toru; Otsuka, Yohei; Takeuchi, Satoru; Suzuki, Mario; Shimizu, Yuzaburo; Toyooka, Terushige; Matsushita, Yuko; Hibiya, Yuko; Tomura, Satoshi; Kondo, Akihide; Wada, Kojiro; Ichimura, Koichi; Tomiyama, Arata.
Afiliación
  • Fujii T; Department of Brain Disease Translational Research, Juntendo University Faculty of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan.
  • Nakano Y; Department of Neurosurgery, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Saitama, Japan.
  • Hagita D; Department of Neurosurgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan.
  • Onishi N; Department of Pediatrics, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
  • Endo A; Department of Brain Disease Translational Research, Juntendo University Faculty of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan.
  • Nakagawa M; Department of Neurosurgery, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Saitama, Japan.
  • Yoshiura T; Department of Neurosurgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan.
  • Otsuka Y; Department of Clinical Diagnostic Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan.
  • Takeuchi S; Department of Neurosurgery, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Saitama, Japan.
  • Suzuki M; Department of Neurosurgery, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Saitama, Japan.
  • Shimizu Y; Department of Neurosurgery, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Saitama, Japan.
  • Toyooka T; Department of Neurosurgery, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Saitama, Japan.
  • Matsushita Y; Department of Neurosurgery, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Saitama, Japan.
  • Hibiya Y; Department of Neurosurgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan.
  • Tomura S; Department of Neurosurgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan.
  • Kondo A; Department of Neurosurgery, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Saitama, Japan.
  • Wada K; Department of Brain Disease Translational Research, Juntendo University Faculty of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan.
  • Ichimura K; Department of Neurosurgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan.
  • Tomiyama A; Department of Brain Disease Translational Research, Juntendo University Faculty of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan.
Cancers (Basel) ; 16(1)2023 Dec 19.
Article en En | MEDLINE | ID: mdl-38201436
ABSTRACT
Here, we investigated the detailed molecular oncogenic mechanisms of a novel receptor tyrosine kinase (RTK) fusion, KLC1-ROS1, with an adapter molecule, KLC1, and an RTK, ROS1, discovered in pediatric glioma, and we explored a novel therapeutic target for glioma that possesses oncogenic RTK fusion. When wild-type ROS1 and KLC1-ROS1 fusions were stably expressed in the human glioma cell lines A172 and U343MG, immunoblotting revealed that KLC1-ROS1 fusion specifically activated the JAK2-STAT3 pathway, a major RTK downstream signaling pathway, when compared with wild-type ROS1. Immunoprecipitation of the fractionated cell lysates revealed a more abundant association of the KLC1-ROS1 fusion with JAK2 than that observed for wild-type ROS1 in the cytosolic fraction. A mutagenesis study of the KLC1-ROS1 fusion protein demonstrated the fundamental roles of both the KLC1 and ROS1 domains in the constitutive activation of KLC1-ROS1 fusion. Additionally, in vitro assays demonstrated that KLC1-ROS1 fusion upregulated cell proliferation, invasion, and chemoresistance when compared to wild-type ROS1. Combination treatment with the chemotherapeutic agent temozolomide and an inhibitor of ROS1, JAK2, or a downstream target of STAT3, demonstrated antitumor effects against KLC1-ROS1 fusion-expressing glioma cells. Our results demonstrate that KLC1-ROS1 fusion exerts oncogenic activity through serum-independent constitutive activation, resulting in specific activation of the JAK-STAT pathway. Our data suggested that molecules other than RTKs may serve as novel therapeutic targets for RTK fusion in gliomas.
Palabras clave

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Idioma: En Revista: Cancers (Basel) Año: 2023 Tipo del documento: Article País de afiliación: Japón

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Idioma: En Revista: Cancers (Basel) Año: 2023 Tipo del documento: Article País de afiliación: Japón