Your browser doesn't support javascript.
loading
Systems level profiling of chemotherapy-induced stress resolution in cancer cells reveals druggable trade-offs.
Saavedra-García, Paula; Roman-Trufero, Monica; Al-Sadah, Hibah A; Blighe, Kevin; López-Jiménez, Elena; Christoforou, Marilena; Penfold, Lucy; Capece, Daria; Xiong, Xiaobei; Miao, Yirun; Parzych, Katarzyna; Caputo, Valentina S; Siskos, Alexandros P; Encheva, Vesela; Liu, Zijing; Thiel, Denise; Kaiser, Martin F; Piazza, Paolo; Chaidos, Aristeidis; Karadimitris, Anastasios; Franzoso, Guido; Snijders, Ambrosius P; Keun, Hector C; Oyarzún, Diego A; Barahona, Mauricio; Auner, Holger W.
  • Saavedra-García P; Cancer Cell Protein Metabolism, Department of Immunology and Inflammation, Imperial College London, London W12 0NN, United Kingdom.
  • Roman-Trufero M; The Hugh and Josseline Langmuir Centre for Myeloma Research, Imperial College London, London W12 0NN, United Kingdom.
  • Al-Sadah HA; Cancer Cell Protein Metabolism, Department of Immunology and Inflammation, Imperial College London, London W12 0NN, United Kingdom.
  • Blighe K; The Hugh and Josseline Langmuir Centre for Myeloma Research, Imperial College London, London W12 0NN, United Kingdom.
  • López-Jiménez E; Cancer Cell Protein Metabolism, Department of Immunology and Inflammation, Imperial College London, London W12 0NN, United Kingdom.
  • Christoforou M; The Hugh and Josseline Langmuir Centre for Myeloma Research, Imperial College London, London W12 0NN, United Kingdom.
  • Penfold L; Clinical Bioinformatics Research, London W1B 3HH, United Kingdom.
  • Capece D; Cancer Cell Protein Metabolism, Department of Immunology and Inflammation, Imperial College London, London W12 0NN, United Kingdom.
  • Xiong X; The Hugh and Josseline Langmuir Centre for Myeloma Research, Imperial College London, London W12 0NN, United Kingdom.
  • Miao Y; Cancer Cell Protein Metabolism, Department of Immunology and Inflammation, Imperial College London, London W12 0NN, United Kingdom.
  • Parzych K; The Hugh and Josseline Langmuir Centre for Myeloma Research, Imperial College London, London W12 0NN, United Kingdom.
  • Caputo VS; Cancer Cell Protein Metabolism, Department of Immunology and Inflammation, Imperial College London, London W12 0NN, United Kingdom.
  • Siskos AP; Cellular Stress, MRC London Institute of Medical Sciences, London W12 0NN, United Kingdom.
  • Encheva V; Centre for Molecular Immunology and Inflammation, Department of Immunology and Inflammation, Imperial College London, London W12 0NN, United Kingdom.
  • Liu Z; Cancer Cell Protein Metabolism, Department of Immunology and Inflammation, Imperial College London, London W12 0NN, United Kingdom.
  • Thiel D; The Hugh and Josseline Langmuir Centre for Myeloma Research, Imperial College London, London W12 0NN, United Kingdom.
  • Kaiser MF; Cancer Cell Protein Metabolism, Department of Immunology and Inflammation, Imperial College London, London W12 0NN, United Kingdom.
  • Piazza P; The Hugh and Josseline Langmuir Centre for Myeloma Research, Imperial College London, London W12 0NN, United Kingdom.
  • Chaidos A; Cancer Cell Protein Metabolism, Department of Immunology and Inflammation, Imperial College London, London W12 0NN, United Kingdom.
  • Karadimitris A; The Hugh and Josseline Langmuir Centre for Myeloma Research, Imperial College London, London W12 0NN, United Kingdom.
  • Franzoso G; Department of Surgery and Cancer, Imperial College London, London W12 0NN, United Kingdom.
  • Snijders AP; Proteomics Platform, The Francis Crick Institute, London NW1 1AT, United Kingdom.
  • Keun HC; Department of Mathematics, Imperial College London, London SW7 2AZ, United Kingdom.
  • Oyarzún DA; Department of Brain Sciences, Imperial College London, London W12 0NN, United Kingdom.
  • Barahona M; UK Dementia Research Institute at Imperial College, London W12 0NN, United Kingdom.
  • Auner HW; Department of Mathematics, Imperial College London, London SW7 2AZ, United Kingdom.
Proc Natl Acad Sci U S A ; 118(17)2021 04 27.
Article en En | MEDLINE | ID: mdl-33883278
ABSTRACT
Cancer cells can survive chemotherapy-induced stress, but how they recover from it is not known. Using a temporal multiomics approach, we delineate the global mechanisms of proteotoxic stress resolution in multiple myeloma cells recovering from proteasome inhibition. Our observations define layered and protracted programs for stress resolution that encompass extensive changes across the transcriptome, proteome, and metabolome. Cellular recovery from proteasome inhibition involved protracted and dynamic changes of glucose and lipid metabolism and suppression of mitochondrial function. We demonstrate that recovering cells are more vulnerable to specific insults than acutely stressed cells and identify the general control nonderepressable 2 (GCN2)-driven cellular response to amino acid scarcity as a key recovery-associated vulnerability. Using a transcriptome analysis pipeline, we further show that GCN2 is also a stress-independent bona fide target in transcriptional signature-defined subsets of solid cancers that share molecular characteristics. Thus, identifying cellular trade-offs tied to the resolution of chemotherapy-induced stress in tumor cells may reveal new therapeutic targets and routes for cancer therapy optimization.
Asunto(s)
Palabras clave

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Estrés Fisiológico / Neoplasias Tipo de estudio: Prognostic_studies Límite: Humans Idioma: En Año: 2021 Tipo del documento: Article

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Estrés Fisiológico / Neoplasias Tipo de estudio: Prognostic_studies Límite: Humans Idioma: En Año: 2021 Tipo del documento: Article