Your browser doesn't support javascript.
loading
A yeast-based genomic strategy highlights the cell protein networks altered by FTase inhibitor peptidomimetics.
Porcu, Giampiero; Wilson, Cathal; Di Giandomenico, Daniele; Ragnini-Wilson, Antonella.
Afiliação
  • Porcu G; Department of Biology, University of Rome Tor Vergata, Italy.
Mol Cancer ; 9: 197, 2010 Jul 23.
Article em En | MEDLINE | ID: mdl-20653956
ABSTRACT

BACKGROUND:

Farnesyltransferase inhibitors (FTIs) are anticancer agents developed to inhibit Ras oncoprotein activities. FTIs of different chemical structure act via a conserved mechanism in eukaryotic cells. They have low toxicity and are active on a wide range of tumors in cellular and animal models, independently of the Ras activation state. Their ultimate mechanism of action, however, remains undetermined. FTase has hundred of substrates in human cells, many of which play a pivotal role in either tumorigenesis or in pro-survival pathways. This lack of knowledge probably accounts for the failure of FTIs at clinical stage III for most of the malignancies treated, with the notable exception of haematological malignancies. Understanding which cellular pathways are the ultimate targets of FTIs in different tumor types and the basis of FTI resistance is required to improve the efficacy of FTIs in cancer treatment.

RESULTS:

Here we used a yeast-based cellular assay to define the transcriptional changes consequent to FTI peptidomimetic administration in conditions that do not substantially change Ras membrane/cytosol distribution. Yeast and cancer cell lines were used to validate the results of the network analysis. The transcriptome of yeast cells treated with FTase inhibitor I was compared with that of untreated cells and with an isogenic strain genetically inhibited for FTase activity (Deltaram1). Cells treated with GGTI-298 were analyzed in a parallel study to validate the specificity of the FTI response. Network analysis, based on gene ontology criteria, identified a cell cycle gene cluster up-regulated by FTI treatment that has the Aurora A kinase IPL1 and the checkpoint protein MAD2 as hubs. Moreover, TORC1-S6K-downstream effectors were found to be down-regulated in yeast and mammalian FTI-treated cells. Notably only FTIs, but not genetic inhibition of FTase, elicited up-regulation of ABC/transporters.

CONCLUSIONS:

This work provides a view of how FTIs globally affect cell activity. It suggests that the chromosome segregation machinery and Aurora A association with the kinetochore as well as TORC1-S6K downstream effectors are among the ultimate targets affected by the transcriptional deregulation caused by FTI peptidomimetics. Moreover, it stresses the importance of monitoring the MDR response in patients treated with FTIs.
Assuntos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Genoma Fúngico / Mimetismo Molecular / Inibidores Enzimáticos / Farnesiltranstransferase Tipo de estudo: Prognostic_studies Limite: Humans Idioma: En Revista: Mol Cancer Assunto da revista: NEOPLASIAS Ano de publicação: 2010 Tipo de documento: Article País de afiliação: Itália

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Genoma Fúngico / Mimetismo Molecular / Inibidores Enzimáticos / Farnesiltranstransferase Tipo de estudo: Prognostic_studies Limite: Humans Idioma: En Revista: Mol Cancer Assunto da revista: NEOPLASIAS Ano de publicação: 2010 Tipo de documento: Article País de afiliação: Itália