Your browser doesn't support javascript.
loading
The eukaryotic translation initiation factor eIF4E reprograms alternative splicing.
Ghram, Mehdi; Morris, Gavin; Culjkovic-Kraljacic, Biljana; Mars, Jean-Clement; Gendron, Patrick; Skrabanek, Lucy; Revuelta, Maria Victoria; Cerchietti, Leandro; Guzman, Monica L; Borden, Katherine L B.
Afiliação
  • Ghram M; Department of Pathology and Cell Biology, Institute for Research in Immunology and Cancer, University of Montreal, Montreal, QC, Canada.
  • Morris G; Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, QC, Canada.
  • Culjkovic-Kraljacic B; Department of Pathology and Cell Biology, Institute for Research in Immunology and Cancer, University of Montreal, Montreal, QC, Canada.
  • Mars JC; Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, QC, Canada.
  • Gendron P; Department of Pathology and Cell Biology, Institute for Research in Immunology and Cancer, University of Montreal, Montreal, QC, Canada.
  • Skrabanek L; Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, QC, Canada.
  • Revuelta MV; Department of Pathology and Cell Biology, Institute for Research in Immunology and Cancer, University of Montreal, Montreal, QC, Canada.
  • Cerchietti L; Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, QC, Canada.
  • Guzman ML; Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, QC, Canada.
  • Borden KLB; Department of Physiology and Biophysics, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA.
EMBO J ; 42(7): e110496, 2023 04 03.
Article em En | MEDLINE | ID: mdl-36843541
ABSTRACT
Aberrant splicing is typically attributed to splice-factor (SF) mutation and contributes to malignancies including acute myeloid leukemia (AML). Here, we discovered a mutation-independent means to extensively reprogram alternative splicing (AS). We showed that the dysregulated expression of eukaryotic translation initiation factor eIF4E elevated selective splice-factor production, thereby impacting multiple spliceosome complexes, including factors mutated in AML such as SF3B1 and U2AF1. These changes generated a splicing landscape that predominantly supported altered splice-site selection for ~800 transcripts in cell lines and ~4,600 transcripts in specimens from high-eIF4E AML patients otherwise harboring no known SF mutations. Nuclear RNA immunoprecipitations, export assays, polysome analyses, and mutational studies together revealed that eIF4E primarily increased SF production via its nuclear RNA export activity. By contrast, eIF4E dysregulation did not induce known SF mutations or alter spliceosome number. eIF4E interacted with the spliceosome and some pre-mRNAs, suggesting its direct involvement in specific splicing events. eIF4E induced simultaneous effects on numerous SF proteins, resulting in a much larger range of splicing alterations than in the case of mutation or dysregulation of individual SFs and providing a novel paradigm for splicing control and dysregulation.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Leucemia Mieloide Aguda / Processamento Alternativo Limite: Humans Idioma: En Revista: EMBO J Ano de publicação: 2023 Tipo de documento: Article País de afiliação: Canadá

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Leucemia Mieloide Aguda / Processamento Alternativo Limite: Humans Idioma: En Revista: EMBO J Ano de publicação: 2023 Tipo de documento: Article País de afiliação: Canadá