Your browser doesn't support javascript.
loading
Tumor suppressor KIF1Bß regulates mitochondrial apoptosis in collaboration with YME1L1.
Ando, Koji; Yokochi, Tomoki; Mukai, Akira; Wei, Gao; Li, Yuanyuan; Kramer, Sonja; Ozaki, Toshinori; Maehara, Yoshihiko; Nakagawara, Akira.
  • Ando K; Division of Biochemistry and Innovative Cancer Therapeutics, Chiba Cancer Center Research Institute, Chiba, Japan.
  • Yokochi T; Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
  • Mukai A; Division of Biochemistry and Innovative Cancer Therapeutics, Chiba Cancer Center Research Institute, Chiba, Japan.
  • Wei G; Division of Biochemistry and Innovative Cancer Therapeutics, Chiba Cancer Center Research Institute, Chiba, Japan.
  • Li Y; Division of Biochemistry and Innovative Cancer Therapeutics, Chiba Cancer Center Research Institute, Chiba, Japan.
  • Kramer S; Division of Biochemistry and Innovative Cancer Therapeutics, Chiba Cancer Center Research Institute, Chiba, Japan.
  • Ozaki T; Division of Molecular Medicine, Life Science Research Institute, Saga Medical Center Koseikan, Saga, Japan.
  • Maehara Y; Division of Biochemistry and Innovative Cancer Therapeutics, Chiba Cancer Center Research Institute, Chiba, Japan.
  • Nakagawara A; Division of Anti-Tumor Research, Chiba Cancer Center Research Institute, Chiba, Japan.
Mol Carcinog ; 58(7): 1134-1144, 2019 07.
Article en En | MEDLINE | ID: mdl-30859632
ABSTRACT
KIF1Bß, a member of the kinesin superfamily of motor proteins, is a haploinsufficient tumor suppressor mapped to chromosome 1p36.2, which is frequently deleted in neural crest-derived tumors, including neuroblastoma and pheochromocytoma. While KIF1Bß acts downstream of the nerve growth factor (NGF) pathway to induce apoptosis, further molecular functions of this gene product have largely been unexplored. In this study, we report that KIF1Bß destabilizes the morphological structure of mitochondria, which is critical for cell survival and apoptosis. We identified YME1L1, a mitochondrial metalloprotease responsible for the cleavage of the mitochondrial GTPase OPA1, as a physical interacting partner of KIF1Bß. KIF1Bß interacted with YME1L1 through its death-inducing region, as initiated the protease activity of YME1L1 to cleave the long forms of OPA1, resulting in mitochondrial fragmentation. Overexpression of YME1L1 promoted apoptosis, while knockdown of YME1L1 promoted cell growth. High YME1L1 expression was significantly associated with a better prognosis in neuroblastoma. Furthermore, in NGF-deprived PC12 cells, KIF1Bß and YME1L1 were upregulated, accompanied by mitochondrial fragmentation and apoptotic cell death. Small interfering RNA-mediated knockdown of either protein alone, however, remarkably inhibited the NGF depletion-induced apoptosis. Our findings indicate that tumor suppressor KIF1Bß plays an important role in intrinsic mitochondria-mediated apoptosis through the regulation of structural and functional dynamics of mitochondria in collaboration with YME1L1. Dysfunction of the KIF1Bß/YME1L1/OPA1 mechanism may be involved in malignant biological features of neural crest-derived tumors as well as the initiation and progression of neurodegenerative diseases.
Asunto(s)
Palabras clave

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Metaloendopeptidasas / Cinesinas / Apoptosis / Proteínas Mitocondriales / ATPasas Asociadas con Actividades Celulares Diversas / Mitocondrias / Neuroblastoma Tipo de estudio: Prognostic_studies Límite: Humans Idioma: En Año: 2019 Tipo del documento: Article

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Metaloendopeptidasas / Cinesinas / Apoptosis / Proteínas Mitocondriales / ATPasas Asociadas con Actividades Celulares Diversas / Mitocondrias / Neuroblastoma Tipo de estudio: Prognostic_studies Límite: Humans Idioma: En Año: 2019 Tipo del documento: Article