Your browser doesn't support javascript.
loading
Oncogenic Cells of Renal Embryonic Lineage Sensitive to the Small-Molecule Inhibitor QC6352 Display Depletion of KDM4 Levels and Disruption of Ribosome Biogenesis.
Pichavaram, Prahalathan; Jablonowski, Carolyn M; Fang, Jie; Fleming, Andrew M; Gil, Hyea Jin; Boghossian, Andrew S; Rees, Matthew G; Ronan, Melissa M; Roth, Jennifer A; Morton, Christopher L; Zambetti, Gerard P; Davidoff, Andrew M; Yang, Jun; Murphy, Andrew J.
Afiliação
  • Pichavaram P; Department of Surgery, St. Jude Children's Research Hospital, Memphis, Tennessee.
  • Jablonowski CM; Department of Surgery, St. Jude Children's Research Hospital, Memphis, Tennessee.
  • Fang J; Department of Surgery, St. Jude Children's Research Hospital, Memphis, Tennessee.
  • Fleming AM; Department of Surgery, St. Jude Children's Research Hospital, Memphis, Tennessee.
  • Gil HJ; Department of Surgery, University of Tennessee Health Science Center, Memphis, Tennessee.
  • Boghossian AS; Department of Surgery, St. Jude Children's Research Hospital, Memphis, Tennessee.
  • Rees MG; Broad Institute of Harvard and MIT, Cambridge, Massachusetts.
  • Ronan MM; Broad Institute of Harvard and MIT, Cambridge, Massachusetts.
  • Roth JA; Broad Institute of Harvard and MIT, Cambridge, Massachusetts.
  • Morton CL; Broad Institute of Harvard and MIT, Cambridge, Massachusetts.
  • Zambetti GP; Department of Surgery, St. Jude Children's Research Hospital, Memphis, Tennessee.
  • Davidoff AM; Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee.
  • Yang J; Department of Surgery, St. Jude Children's Research Hospital, Memphis, Tennessee.
  • Murphy AJ; Department of Surgery, University of Tennessee Health Science Center, Memphis, Tennessee.
Mol Cancer Ther ; 23(4): 478-491, 2024 Apr 02.
Article em En | MEDLINE | ID: mdl-37988559
ABSTRACT
The histone lysine demethylases KDM4A-C are involved in physiologic processes including stem cell identity and self-renewal during development, DNA damage repair, and cell-cycle progression. KDM4A-C are overexpressed and associated with malignant cell behavior in multiple human cancers and are therefore potential therapeutic targets. Given the role of KDM4A-C in development and cancer, we aimed to test the potent, selective KDM4A-C inhibitor QC6352 on oncogenic cells of renal embryonic lineage. The anaplastic Wilms tumor cell line WiT49 and the tumor-forming human embryonic kidney cell line HEK293 demonstrated low nanomolar QC6352 sensitivity. The cytostatic response to QC6352 in WiT49 and HEK293 cells was marked by induction of DNA damage, a DNA repair-associated protein checkpoint response, S-phase cell-cycle arrest, profound reduction of ribosomal protein gene and rRNA transcription, and blockade of newly synthesized proteins. QC6352 caused reduction of KDM4A-C levels by a proteasome-associated mechanism. The cellular phenotype caused by QC6352 treatment of reduced migration, proliferation, tumor spheroid growth, DNA damage, and S-phase cell-cycle arrest was most closely mirrored by knockdown of KDM4A as determined by siRNA knockdown of KDM4A-C. QC6352 sensitivity correlated with high basal levels of ribosomal gene transcription in more than 900 human cancer cell lines. Targeting KDM4A may be of future therapeutic interest in oncogenic cells of embryonic renal lineage or cells with high basal expression of ribosomal protein genes.
Assuntos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Proteínas Ribossômicas / Histona Desmetilases com o Domínio Jumonji / Compostos Heterocíclicos de 4 ou mais Anéis Idioma: En Ano de publicação: 2024 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Proteínas Ribossômicas / Histona Desmetilases com o Domínio Jumonji / Compostos Heterocíclicos de 4 ou mais Anéis Idioma: En Ano de publicação: 2024 Tipo de documento: Article