Your browser doesn't support javascript.
loading
Identification of a novel quinoline-based DNA demethylating compound highly potent in cancer cells.
Zwergel, Clemens; Schnekenburger, Michael; Sarno, Federica; Battistelli, Cecilia; Manara, Maria Cristina; Stazi, Giulia; Mazzone, Roberta; Fioravanti, Rossella; Gros, Christina; Ausseil, Frédéric; Florean, Cristina; Nebbioso, Angela; Strippoli, Raffaele; Ushijima, Toshikazu; Scotlandi, Katia; Tripodi, Marco; Arimondo, Paola B; Altucci, Lucia; Diederich, Marc; Mai, Antonello; Valente, Sergio.
Afiliación
  • Zwergel C; Department of Chemistry and Technologies of Drugs, Sapienza University of Rome, P.le A. Moro 5, 00185, Rome, Italy.
  • Schnekenburger M; Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, 9 rue Edward Steichen, L-2540, Luxembourg City, Luxembourg.
  • Sarno F; Department of Medicine of Precision, University of Studi della Campania Luigi Vanvitelli, Vico L. De Crecchio 7, 80138, Naples, Italy.
  • Battistelli C; Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy.
  • Manara MC; Laboratory of Experimental Oncology, IRCCS - Istituto Ortopedico Rizzoli, via di Barbiano, 1/10, Bologna, 40136, Italy.
  • Stazi G; Department of Chemistry and Technologies of Drugs, Sapienza University of Rome, P.le A. Moro 5, 00185, Rome, Italy.
  • Mazzone R; Department of Chemistry and Technologies of Drugs, Sapienza University of Rome, P.le A. Moro 5, 00185, Rome, Italy.
  • Fioravanti R; Department of Chemistry and Technologies of Drugs, Sapienza University of Rome, P.le A. Moro 5, 00185, Rome, Italy.
  • Gros C; Center for High-Throughput Chemical Biology, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1S6, Canada.
  • Ausseil F; Pierre Fabre Laboratories, 3 Avenue Hubert Curien, Toulouse, 31100, France.
  • Florean C; Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, 9 rue Edward Steichen, L-2540, Luxembourg City, Luxembourg.
  • Nebbioso A; Department of Medicine of Precision, University of Studi della Campania Luigi Vanvitelli, Vico L. De Crecchio 7, 80138, Naples, Italy.
  • Strippoli R; Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy.
  • Ushijima T; National Institute for Infectious Diseases L. Spallanzani, IRCCS, Via Portuense, 292, Rome, 00149, Italy.
  • Scotlandi K; Division of Epigenomics, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.
  • Tripodi M; Laboratory of Experimental Oncology, IRCCS - Istituto Ortopedico Rizzoli, via di Barbiano, 1/10, Bologna, 40136, Italy.
  • Arimondo PB; Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy.
  • Altucci L; National Institute for Infectious Diseases L. Spallanzani, IRCCS, Via Portuense, 292, Rome, 00149, Italy.
  • Diederich M; Pasteur Institute, Cenci-Bolognetti Foundation, Sapienza University of Rome, P.le A. Moro 5, 00185, Rome, Italy.
  • Mai A; Epigenetic Chemical Biology, Institut Pasteur, CNRS UMR3523, 28 rue du Docteur Roux, Paris, 75724, France.
  • Valente S; Department of Medicine of Precision, University of Studi della Campania Luigi Vanvitelli, Vico L. De Crecchio 7, 80138, Naples, Italy.
Clin Epigenetics ; 11(1): 68, 2019 05 06.
Article en En | MEDLINE | ID: mdl-31060628
ABSTRACT

BACKGROUND:

DNA methyltransferases (DNMTs) are epigenetic enzymes involved in embryonic development, cell differentiation, epithelial to mesenchymal transition, and control of gene expression, whose overexpression or enhanced catalytic activity has been widely reported in cancer initiation and progression. To date, two DNMT inhibitors (DNMTi), 5-azacytidine (5-AZA) and 5-aza-2'-deoxycytidine (DAC), are approved for the treatment of myelodysplastic syndromes and acute myeloid leukemia. Nevertheless, they are chemically instable and quite toxic for healthy cells; thus, the discovery of novel DNMTi is urgent.

RESULTS:

Here, we report the identification of a new quinoline-based molecule, MC3353, as a non-nucleoside inhibitor and downregulator of DNMT. This compound was able, in promoter demethylating assays, to induce enhanced green fluorescence protein (EGFP) gene expression in HCT116 cells and transcription in a cytomegalovirus (CMV) promoter-driven luciferase reporter system in KG-1 cells. Moreover, MC3353 displayed a strong antiproliferative activity when tested on HCT116 colon cancer cells after 48 h of treatment at 0.5 µM. At higher doses, this compound provided a cytotoxic effect in double DNMT knockout HCT116 cells. MC3353 was also screened on a different panel of cancer cells (KG-1 and U-937 acute myeloid leukemia, RAJI Burkitt's lymphoma, PC-3 prostate cancer, and MDA-MB-231 breast cancer), where it arrested cell proliferation and reduced viability after 48 h of treatment with IC50 values ranging from 0.3 to 0.9 µM. Compared to healthy cell models, MC3353 induced apoptosis (e.g., U-937 and KG-1 cells) or necrosis (e.g., RAJI cells) at lower concentrations. Importantly, together with the main DNMT3A enzyme inhibition, MC3353 was also able to downregulate the DNMT3A protein level in selected HCT116 and PC-3 cell lines. Additionally, this compound provided impairment of the epithelial-to-mesenchymal transition (EMT) by inducing E-cadherin while reducing matrix metalloproteinase (MMP2) mRNA and protein levels in PC-3 and HCT116 cells. Last, tested on a panel of primary osteosarcoma cell lines, MC3353 markedly inhibited cell growth with low single-digit micromolar IC50 ranging from 1.1 to 2.4 µM. Interestingly, in Saos-2 osteosarcoma cells, MC3353 induced both expression of genes and mineralized the matrix as evidence of osteosarcoma to osteoblast differentiation.

CONCLUSIONS:

The present work describes MC3353 as a novel DNMTi displaying a stronger in cell demethylating ability than both 5-AZA and DAC, providing re-activation of the silenced ubiquitin C-terminal hydrolase L1 (UCHL1) gene. MC3353 displayed dose- and time-dependent antiproliferative activity in several cancer cell types, inducing cell death and affecting EMT through E-cadherin and MMP2 modulation. In addition, this compound proved efficacy even in primary osteosarcoma cell models, through the modulation of genes involved in osteoblast differentiation.
Asunto(s)
Palabras clave

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: ADN-Citosina Metilasas / Inhibidores Enzimáticos / Aminoquinolinas / Neoplasias Tipo de estudio: Diagnostic_studies Límite: Humans Idioma: En Revista: Clin Epigenetics Año: 2019 Tipo del documento: Article País de afiliación: Italia

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: ADN-Citosina Metilasas / Inhibidores Enzimáticos / Aminoquinolinas / Neoplasias Tipo de estudio: Diagnostic_studies Límite: Humans Idioma: En Revista: Clin Epigenetics Año: 2019 Tipo del documento: Article País de afiliación: Italia