Your browser doesn't support javascript.
loading
Inhibition of Histone Demethylases LSD1 and UTX Regulates ERα Signaling in Breast Cancer.
Benedetti, Rosaria; Dell'Aversana, Carmela; De Marchi, Tommaso; Rotili, Dante; Liu, Ning Qing; Novakovic, Boris; Boccella, Serena; Di Maro, Salvatore; Cosconati, Sandro; Baldi, Alfonso; Niméus, Emma; Schultz, Johan; Höglund, Urban; Maione, Sabatino; Papulino, Chiara; Chianese, Ugo; Iovino, Francesco; Federico, Antonio; Mai, Antonello; Stunnenberg, Hendrik G; Nebbioso, Angela; Altucci, Lucia.
Afiliação
  • Benedetti R; Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy.
  • Dell'Aversana C; Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy.
  • De Marchi T; Institute Experimental Endocrinology and Oncology "Gaetano Salvatore" (IEOS)-National Research Council (CNR) Via Sergio Pansini, 5-80131 Napoli, Italy.
  • Rotili D; Department of Oncology and Pathology, Lund University, SE-221 00 Lund, Sweden.
  • Liu NQ; Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Rome, Italy.
  • Novakovic B; Department of Molecular Biology, Radboud University, 6500 HB Nijmegen, The Netherlands.
  • Boccella S; Murdoch Children's Research Institute and Department of Paediatrics, University of Melbourne, Melbourne, Parkville Victoria 3052, Australia.
  • Di Maro S; Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", University of Campania "Luigi Vanvitelli", 80138 Naples, Italy.
  • Cosconati S; Dipartimento di Scienze e Tecnologie Ambientali Biologiche e Farmaceutiche, University of Campania 'Luigi Vanvitelli', 81100 Caserta, Italy.
  • Baldi A; Dipartimento di Scienze e Tecnologie Ambientali Biologiche e Farmaceutiche, University of Campania 'Luigi Vanvitelli', 81100 Caserta, Italy.
  • Niméus E; Dipartimento di Scienze e Tecnologie Ambientali Biologiche e Farmaceutiche, University of Campania 'Luigi Vanvitelli', 81100 Caserta, Italy.
  • Schultz J; Department of Oncology and Pathology, Lund University, SE-221 00 Lund, Sweden.
  • Höglund U; Department of Surgery, Skånes University Hospital, 222 29 Lund, Sweden.
  • Maione S; Kancera AB, Banvaktsvagen 22, SE-17148 Solna, Sweden.
  • Papulino C; Adlego Biomedical AB, P.O. Box 42, SE-751 03 Uppsala, Sweden.
  • Chianese U; Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", University of Campania "Luigi Vanvitelli", 80138 Naples, Italy.
  • Iovino F; Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy.
  • Federico A; Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy.
  • Mai A; Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", Via L. De Crecchio 7, 80138 Naples, Italy.
  • Stunnenberg HG; Faculty of Medicine and Health Technology, Tampere University, 33100 Tampere, Finland.
  • Nebbioso A; Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Rome, Italy.
  • Altucci L; Department of Molecular Biology, Radboud University, 6500 HB Nijmegen, The Netherlands.
Cancers (Basel) ; 11(12)2019 Dec 16.
Article em En | MEDLINE | ID: mdl-31888209
ABSTRACT
In breast cancer, Lysine-specific demethylase-1 (LSD1) and other lysine demethylases (KDMs), such as Lysine-specific demethylase 6A also known as Ubiquitously transcribed tetratricopeptide repeat, X chromosome (UTX), are co-expressed and co-localize with estrogen receptors (ERs), suggesting the potential use of hybrid (epi)molecules to target histone methylation and therefore regulate/redirect hormone receptor signaling. Here, we report on the biological activity of a dual-KDM inhibitor (MC3324), obtained by coupling the chemical properties of tranylcypromine, a known LSD1 inhibitor, with the 2OG competitive moiety developed for JmjC inhibition. MC3324 displays unique features not exhibited by the single moieties and well-characterized mono-pharmacological inhibitors. Inhibiting LSD1 and UTX, MC3324 induces significant growth arrest and apoptosis in hormone-responsive breast cancer model accompanied by a robust increase in H3K4me2 and H3K27me3. MC3324 down-regulates ERα in breast cancer at both transcriptional and non-transcriptional levels, mimicking the action of a selective endocrine receptor disruptor. MC3324 alters the histone methylation of ERα-regulated promoters, thereby affecting the transcription of genes involved in cell surveillance, hormone response, and death. MC3324 reduces cell proliferation in ex vivo breast cancers, as well as in breast models with acquired resistance to endocrine therapies. Similarly, MC3324 displays tumor-selective potential in vivo, in both xenograft mice and chicken embryo models, with no toxicity and good oral efficacy. This epigenetic multi-target approach is effective and may overcome potential mechanism(s) of resistance in breast cancer.
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Idioma: En Ano de publicação: 2019 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Idioma: En Ano de publicação: 2019 Tipo de documento: Article