Your browser doesn't support javascript.
loading
Krüppel-like factor 4 (KLF4) induces mitochondrial fusion and increases spare respiratory capacity of human glioblastoma cells.
Wang, Shuyan; Shi, Xiaohai; Wei, Shuang; Ma, Ding; Oyinlade, Olutobi; Lv, Sheng-Qing; Ying, Mingyao; Zhang, Yu Alex; Claypool, Steven Michael; Watkins, Paul; Xia, Shuli.
Afiliação
  • Wang S; From the Cell Therapy Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.
  • Shi X; the Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland 21205.
  • Wei S; the Departments of Neurology and.
  • Ma D; the Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland 21205.
  • Oyinlade O; the Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland 21205.
  • Lv SQ; the Departments of Neurology and.
  • Ying M; the Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan 430030, China, and.
  • Zhang YA; the Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland 21205.
  • Claypool SM; the Departments of Neurology and.
  • Watkins P; the Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland 21205.
  • Xia S; the Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China.
J Biol Chem ; 293(17): 6544-6555, 2018 04 27.
Article em En | MEDLINE | ID: mdl-29507094
ABSTRACT
Krüppel-like factor 4 (KLF4) is a zinc finger transcription factor critical for the regulation of many cellular functions in both normal and neoplastic cells. Here, using human glioblastoma cells, we investigated KLF4's effects on cancer cell metabolism. We found that forced KLF4 expression promotes mitochondrial fusion and induces dramatic changes in mitochondrial morphology. To determine the impact of these changes on the cellular functions following, we analyzed how KLF4 alters glioblastoma cell metabolism, including glucose uptake, glycolysis, pentose phosphate pathway, and oxidative phosphorylation. We did not identify significant differences in baseline cellular metabolism between control and KLF4-expressing cells. However, when mitochondrial function was impaired, KLF4 significantly increased spare respiratory capacity and levels of reactive oxygen species in the cells. To identify the biological effects of these changes, we analyzed proliferation and survival of control and KLF4-expressing cells under stress conditions, including serum and nutrition deprivation. We found that following serum starvation, KLF4 altered cell cycle progression by arresting the cells at the G2/M phase and that KLF4 protected cells from nutrition deprivation-induced death. Finally, we demonstrated that methylation-dependent KLF4-binding activity mediates mitochondrial fusion. Specifically, the downstream targets of KLF4-mCpG binding, guanine nucleotide exchange factors, serve as the effector of KLF4-induced mitochondrial fusion, cell cycle arrest, and cell protection. Our experimental system provides a robust model for studying the interactions between mitochondrial morphology and function, mitochondrial dynamics and metabolism, and mitochondrial fusion and cell death during tumor initiation and progression.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Consumo de Oxigênio / Divisão Celular / Fase G2 / Glioblastoma / Fatores de Transcrição Kruppel-Like / Dinâmica Mitocondrial / Proteínas de Neoplasias Tipo de estudo: Prognostic_studies Limite: Humans Idioma: En Ano de publicação: 2018 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Consumo de Oxigênio / Divisão Celular / Fase G2 / Glioblastoma / Fatores de Transcrição Kruppel-Like / Dinâmica Mitocondrial / Proteínas de Neoplasias Tipo de estudo: Prognostic_studies Limite: Humans Idioma: En Ano de publicação: 2018 Tipo de documento: Article