Your browser doesn't support javascript.
loading
The splicing factor RBM17 drives leukemic stem cell maintenance by evading nonsense-mediated decay of pro-leukemic factors.
Liu, Lina; Vujovic, Ana; Deshpande, Nandan P; Sathe, Shashank; Anande, Govardhan; Chen, He Tian Tony; Xu, Joshua; Minden, Mark D; Yeo, Gene W; Unnikrishnan, Ashwin; Hope, Kristin J; Lu, Yu.
Afiliação
  • Liu L; Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada.
  • Vujovic A; Department of Biochemistry and Biomedical Sciences, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada.
  • Deshpande NP; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
  • Sathe S; Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
  • Anande G; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
  • Chen HTT; Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
  • Xu J; Adult Cancer Program, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW, Australia.
  • Minden MD; Prince of Wales Clinical School, University of New South Wales, Sydney, NSW, Australia.
  • Yeo GW; Department of Cellular and Molecular Medicine, Stem Cell Program and Institute for Genomic Medicine, University of California at San Diego, San Diego, CA, USA.
  • Unnikrishnan A; Adult Cancer Program, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW, Australia.
  • Hope KJ; Prince of Wales Clinical School, University of New South Wales, Sydney, NSW, Australia.
  • Lu Y; Department of Biochemistry and Biomedical Sciences, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada.
Nat Commun ; 13(1): 3833, 2022 07 04.
Article em En | MEDLINE | ID: mdl-35781533
ABSTRACT
Chemo-resistance in acute myeloid leukemia (AML) patients is driven by leukemic stem cells (LSCs) resulting in high rates of relapse and low overall survival. Here, we demonstrate that upregulation of the splicing factor, RBM17 preferentially marks and sustains LSCs and directly correlates with shorten patient survival. RBM17 knockdown in primary AML cells leads to myeloid differentiation and impaired colony formation and in vivo engraftment. Integrative multi-omics analyses show that RBM17 repression leads to inclusion of poison exons and production of nonsense-mediated decay (NMD)-sensitive transcripts for pro-leukemic factors and the translation initiation factor, EIF4A2. We show that EIF4A2 is enriched in LSCs and its inhibition impairs primary AML progenitor activity. Proteomic analysis of EIF4A2-depleted AML cells shows recapitulation of the RBM17 knockdown biological effects, including pronounced suppression of proteins involved in ribosome biogenesis. Overall, these results provide a rationale to target RBM17 and/or its downstream NMD-sensitive splicing substrates for AML treatment.
Assuntos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Células-Tronco Neoplásicas / Leucemia Mieloide Aguda / Fatores de Processamento de RNA Limite: Humans Idioma: En Ano de publicação: 2022 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Células-Tronco Neoplásicas / Leucemia Mieloide Aguda / Fatores de Processamento de RNA Limite: Humans Idioma: En Ano de publicação: 2022 Tipo de documento: Article