Your browser doesn't support javascript.
loading
KCa3.1 channel inhibition sensitizes malignant gliomas to temozolomide treatment.
D'Alessandro, Giuseppina; Grimaldi, Alfonso; Chece, Giuseppina; Porzia, Alessandra; Esposito, Vincenzo; Santoro, Antonio; Salvati, Maurizio; Mainiero, Fabrizio; Ragozzino, Davide; Di Angelantonio, Silvia; Wulff, Heike; Catalano, Myriam; Limatola, Cristina.
Afiliação
  • D'Alessandro G; Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy.
  • Grimaldi A; IRCCS Neuromed, Pozzilli, Italy.
  • Chece G; Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy.
  • Porzia A; Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy.
  • Esposito V; Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy.
  • Santoro A; IRCCS Neuromed, Pozzilli, Italy.
  • Salvati M; Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy.
  • Mainiero F; Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy.
  • Ragozzino D; Department of Science and Medical Surgical Biotechnology, Sapienza University of Rome, Rome, Italy.
  • Di Angelantonio S; IRCCS Neuromed, Pozzilli, Italy.
  • Wulff H; Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy.
  • Catalano M; Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy.
  • Limatola C; IRCCS Neuromed, Pozzilli, Italy.
Oncotarget ; 7(21): 30781-96, 2016 May 24.
Article em En | MEDLINE | ID: mdl-27096953
ABSTRACT
Malignant gliomas are among the most frequent and aggressive cerebral tumors, characterized by high proliferative and invasive indexes. Standard therapy for patients, after surgery and radiotherapy, consists of temozolomide (TMZ), a methylating agent that blocks tumor cell proliferation. Currently, there are no therapies aimed at reducing tumor cell invasion. Ion channels are candidate molecular targets involved in glioma cell migration and infiltration into the brain parenchyma. In this paper we demonstrate that i) blockade of the calcium-activated potassium channel KCa3.1 with TRAM-34 has co-adjuvant effects with TMZ, reducing GL261 glioma cell migration, invasion and colony forming activity, increasing apoptosis, and forcing cells to pass the G2/M cell cycle phase, likely through cdc2 de-phosphorylation; ii) KCa3.1 silencing potentiates the inhibitory effect of TMZ on glioma cell viability; iii) the combination of TMZ/TRAM-34 attenuates the toxic effects of glioma conditioned medium on neuronal cultures, through a microglia dependent mechanism since the effect is abolished by clodronate-induced microglia killing; iv) TMZ/TRAM-34 co-treatment increases the number of apoptotic tumor cells, and the mean survival time in a syngeneic mouse glioma model (C57BL6 mice implanted with GL261 cells); v) TMZ/TRAM-34 co-treatment reduces cell viability of GBM cells and cancer stem cells (CSC) freshly isolated from patients.Taken together, these data suggest a new therapeutic approach for malignant glioma, targeting both glioma cell proliferating and migration, and demonstrate that TMZ/TRAM-34 co-treatment affects both glioma cells and infiltrating microglia, resulting in an overall reduction of tumor cell progression.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Pirazóis / Neoplasias Encefálicas / Antineoplásicos Alquilantes / Dacarbazina / Canais de Potássio Ativados por Cálcio de Condutância Intermediária / Glioma Idioma: En Revista: Oncotarget Ano de publicação: 2016 Tipo de documento: Article País de afiliação: Itália

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Pirazóis / Neoplasias Encefálicas / Antineoplásicos Alquilantes / Dacarbazina / Canais de Potássio Ativados por Cálcio de Condutância Intermediária / Glioma Idioma: En Revista: Oncotarget Ano de publicação: 2016 Tipo de documento: Article País de afiliação: Itália