Your browser doesn't support javascript.
loading
Loss of DIP2C in RKO cells stimulates changes in DNA methylation and epithelial-mesenchymal transition.
Larsson, Chatarina; Ali, Muhammad Akhtar; Pandzic, Tatjana; Lindroth, Anders M; He, Liqun; Sjöblom, Tobias.
Afiliação
  • Larsson C; Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, Dag Hammarskjölds väg 20, SE-751 85, Uppsala, Sweden.
  • Ali MA; Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, Dag Hammarskjölds väg 20, SE-751 85, Uppsala, Sweden.
  • Pandzic T; Current address: Department of Medical Biochemistry and Microbiology, Uppsala University, BMC, Husargatan 3, SE-751 23, Uppsala, Sweden.
  • Lindroth AM; Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, Dag Hammarskjölds väg 20, SE-751 85, Uppsala, Sweden.
  • He L; Department of System Cancer Science, Graduate School of Cancer Science and Policy, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, 10408, Goyang-si, Republic of Korea.
  • Sjöblom T; Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, Dag Hammarskjölds väg 20, SE-751 85, Uppsala, Sweden.
BMC Cancer ; 17(1): 487, 2017 Jul 17.
Article em En | MEDLINE | ID: mdl-28716088
ABSTRACT

BACKGROUND:

The disco-interacting protein 2 homolog C (DIP2C) gene is an uncharacterized gene found mutated in a subset of breast and lung cancers. To understand the role of DIP2C in tumour development we studied the gene in human cancer cells.

METHODS:

We engineered human DIP2C knockout cells by genome editing in cancer cells. The growth properties of the engineered cells were characterised and transcriptome and methylation analyses were carried out to identify pathways deregulated by inactivation of DIP2C. Effects on cell death pathways and epithelial-mesenchymal transition traits were studied based on the results from expression profiling.

RESULTS:

Knockout of DIP2C in RKO cells resulted in cell enlargement and growth retardation. Expression profiling revealed 780 genes for which the expression level was affected by the loss of DIP2C, including the tumour-suppressor encoding CDKN2A gene, the epithelial-mesenchymal transition (EMT) regulator-encoding ZEB1, and CD44 and CD24 that encode breast cancer stem cell markers. Analysis of DNA methylation showed more than 30,000 sites affected by differential methylation, the majority of which were hypomethylated following loss of DIP2C. Changes in DNA methylation at promoter regions were strongly correlated to changes in gene expression, and genes involved with EMT and cell death were enriched among the differentially regulated genes. The DIP2C knockout cells had higher wound closing capacity and showed an increase in the proportion of cells positive for cellular senescence markers.

CONCLUSIONS:

Loss of DIP2C triggers substantial DNA methylation and gene expression changes, cellular senescence and epithelial-mesenchymal transition in cancer cells.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Proteínas Nucleares / Proteínas de Transporte / Neoplasias do Colo / Metilação de DNA / Transição Epitelial-Mesenquimal Idioma: En Ano de publicação: 2017 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Proteínas Nucleares / Proteínas de Transporte / Neoplasias do Colo / Metilação de DNA / Transição Epitelial-Mesenquimal Idioma: En Ano de publicação: 2017 Tipo de documento: Article