Your browser doesn't support javascript.
loading
ZRF1 controls the retinoic acid pathway and regulates leukemogenic potential in acute myeloid leukemia.
Demajo, S; Uribesalgo, I; Gutiérrez, A; Ballaré, C; Capdevila, S; Roth, M; Zuber, J; Martín-Caballero, J; Di Croce, L.
Affiliation
  • Demajo S; 1] Centre de Regulació Genòmica (CRG), Barcelona, Spain [2] UPF, Barcelona, Spain.
  • Uribesalgo I; 1] Centre de Regulació Genòmica (CRG), Barcelona, Spain [2] UPF, Barcelona, Spain.
  • Gutiérrez A; 1] Centre de Regulació Genòmica (CRG), Barcelona, Spain [2] UPF, Barcelona, Spain.
  • Ballaré C; 1] Centre de Regulació Genòmica (CRG), Barcelona, Spain [2] UPF, Barcelona, Spain.
  • Capdevila S; Unidad de Animal de Laboratorio, PRBB, Barcelona, Spain.
  • Roth M; Institute of Molecular Pathology (IMP), Vienna, Austria.
  • Zuber J; Institute of Molecular Pathology (IMP), Vienna, Austria.
  • Martín-Caballero J; Unidad de Animal de Laboratorio, PRBB, Barcelona, Spain.
  • Di Croce L; 1] Centre de Regulació Genòmica (CRG), Barcelona, Spain [2] UPF, Barcelona, Spain [3] Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
Oncogene ; 33(48): 5501-10, 2014 Nov 27.
Article in En | MEDLINE | ID: mdl-24292673
ABSTRACT
Acute myeloid leukemia (AML) is frequently linked to epigenetic abnormalities and deregulation of gene transcription, which lead to aberrant cell proliferation and accumulation of undifferentiated precursors. ZRF1, a recently characterized epigenetic factor involved in transcriptional regulation, is highly overexpressed in human AML, but it is not known whether it plays a role in leukemia progression. Here, we demonstrate that ZRF1 depletion decreases cell proliferation, induces apoptosis and enhances cell differentiation in human AML cells. Treatment with retinoic acid (RA), a differentiating agent currently used to treat certain AMLs, leads to a functional switch of ZRF1 from a negative regulator to an activator of differentiation. At the molecular level, ZRF1 controls the RA-regulated gene network through its interaction with the RA receptor α (RARα) and its binding to RA target genes. Our genome-wide expression study reveals that ZRF1 regulates the transcription of nearly half of RA target genes. Consistent with our in vitro observations that ZRF1 regulates proliferation, apoptosis, and differentiation, ZRF1 depletion strongly inhibits leukemia progression in a xenograft mouse model. Finally, ZRF1 knockdown cooperates with RA treatment in leukemia suppression in vivo. Taken together, our data reveal that ZRF1 is a key transcriptional regulator in leukemia progression and suggest that ZRF1 inhibition could be a novel strategy to be explored for AML treatment.
Subject(s)

Full text: 1 Collection: 01-internacional Health context: 1_ASSA2030 / 6_ODS3_enfermedades_notrasmisibles Database: MEDLINE Main subject: Tretinoin / Leukemia, Myeloid, Acute / Signal Transduction / Gene Expression Regulation, Neoplastic / Oncogene Proteins / DNA-Binding Proteins Type of study: Prognostic_studies Limits: Animals / Humans Language: En Journal: Oncogene Year: 2014 Document type: Article

Full text: 1 Collection: 01-internacional Health context: 1_ASSA2030 / 6_ODS3_enfermedades_notrasmisibles Database: MEDLINE Main subject: Tretinoin / Leukemia, Myeloid, Acute / Signal Transduction / Gene Expression Regulation, Neoplastic / Oncogene Proteins / DNA-Binding Proteins Type of study: Prognostic_studies Limits: Animals / Humans Language: En Journal: Oncogene Year: 2014 Document type: Article