Your browser doesn't support javascript.
loading
UCH-L1 bypasses mTOR to promote protein biosynthesis and is required for MYC-driven lymphomagenesis in mice.
Hussain, Sajjad; Bedekovics, Tibor; Liu, Qiuying; Hu, Wenqian; Jeon, Haeseung; Johnson, Sarah H; Vasmatzis, George; May, Danielle G; Roux, Kyle J; Galardy, Paul J.
Afiliação
  • Hussain S; Department of Pediatric and Adolescent Medicine.
  • Bedekovics T; Department of Pediatric and Adolescent Medicine.
  • Liu Q; Department of Biochemistry and Molecular Biology.
  • Hu W; Department of Biochemistry and Molecular Biology.
  • Jeon H; Department of Pediatric and Adolescent Medicine.
  • Johnson SH; Center for Individualized Medicine-Biomarker Discovery, and.
  • Vasmatzis G; Center for Individualized Medicine-Biomarker Discovery, and.
  • May DG; Department of Molecular Medicine, Mayo Clinic, Rochester, MN.
  • Roux KJ; Enabling Technology Group, Sanford Research, Sioux Falls, SD.
  • Galardy PJ; Enabling Technology Group, Sanford Research, Sioux Falls, SD.
Blood ; 132(24): 2564-2574, 2018 12 13.
Article em En | MEDLINE | ID: mdl-30257881
ABSTRACT
The mechanistic target of rapamycin (mTOR) is a central regulator of cellular proliferation and metabolism. Depending on its binding partners, mTOR is at the core of 2 complexes that either promote protein biosynthesis (mTOR complex 1; mTORC1) or provide survival and proliferation signals (mTORC2). Protein biosynthesis downstream of mTORC1 plays an important role in MYC-driven oncogenesis with translation inhibitors garnering increasing therapeutic attention. The germinal center B-cell oncogene UCHL1 encodes a deubiquitinating enzyme that regulates the balance between mTOR complexes by disrupting mTORC1 and promoting mTORC2 assembly. While supporting mTORC2-dependent growth and survival signals may contribute to its role in cancer, the suppression of mTORC1 activity is enigmatic, as its phosphorylation of its substrate 4EBP1 promotes protein biosynthesis. To address this, we used proximity-based proteomics to identify molecular complexes with which UCH-L1 associates in malignant B cells. We identified a novel association of UCH-L1 with the translation initiation complex eIF4F, the target of 4EBP1. UCH-L1 associates with and promotes the assembly of eIF4F and stimulates protein synthesis through a mechanism that requires its catalytic activity. Because of the importance of mTOR in MYC-driven oncogenesis, we used novel mutant Uchl1 transgenic mice and found that catalytic activity is required for its acceleration of lymphoma in the Eµ-myc model. Further, we demonstrate that mice lacking UCH-L1 are resistant to MYC-induced lymphomas. We conclude that UCH-L1 bypasses the need for mTORC1-dependent protein synthesis by directly promoting translation initiation, and that this mechanism may be essential for MYC in B-cell malignancy.
Assuntos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Transformação Celular Neoplásica / Linfoma de Células B / Ubiquitina Tiolesterase / Serina-Treonina Quinases TOR / Proteínas de Neoplasias Idioma: En Ano de publicação: 2018 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Transformação Celular Neoplásica / Linfoma de Células B / Ubiquitina Tiolesterase / Serina-Treonina Quinases TOR / Proteínas de Neoplasias Idioma: En Ano de publicação: 2018 Tipo de documento: Article