학술논문

Elevated MARCKS phosphorylation contributes to unresponsiveness of breast cancer to paclitaxel treatment
Document Type
article
Source
Oncotarget. 6(17)
Subject
Biomedical and Clinical Sciences
Oncology and Carcinogenesis
Breast Cancer
Cancer
2.1 Biological and endogenous factors
Development of treatments and therapeutic interventions
5.1 Pharmaceuticals
Aetiology
Animals
Antineoplastic Agents
Phytogenic
Breast Neoplasms
Cell Line
Tumor
Female
Humans
Intracellular Signaling Peptides and Proteins
MCF-7 Cells
Membrane Proteins
Mice
Mice
Nude
Myristoylated Alanine-Rich C Kinase Substrate
Neovascularization
Pathologic
Paclitaxel
Peptide Fragments
Phosphorylation
RNA Interference
RNA
Small Interfering
Tubulin Modulators
phospho-MARCKS
MANS peptide
paclitaxel
mitotic inhibitor
breast cancer
Oncology and carcinogenesis
Language
Abstract
Accumulating evidence has suggested that myristoylated alanine-rich C-kinase substrate (MARCKS) is critical for regulating multiple pathophysiological processes. However, the molecular mechanism underlying increased phosphorylation of MARCKS at Ser159/163 (phospho-MARCKS) and its functional consequence in neoplastic disease remain to be established. Herein, we investigated how phospho-MARCKS is regulated in breast carcinoma, and its role in the context of chemotherapy. In a screen of patients with breast tumors, we find that the abundance of phospho-MARCKS, not MARCKS protein per se, increased in breast cancers and positively correlated with tumor grade and metastatic status. Among chemotherapeutic agents, mitotic inhibitors, including paclitaxel, vincristine or eribulin, notably promoted phospho-MARCKS accumulation in multiple breast cancer cells. We further show that phospho-MARCKS acted upstream of Src activation upon paclitaxel exposure. Reduction of phospho-MARCKS by knockdown of MARCKS or pharmacological agents increased paclitaxel sensitivity. Particularly, a known phospho-MARCKS inhibitor, MANS peptide, was demonstrated to increase paclitaxel efficacy and attenuate angiogenesis/metastasis of xenografted breast cancer cells by decreasing abundance of phospho-MARCKS and messages of inflammatory mediators. Our data suggest that unresponsiveness of breast cancer to paclitaxel treatment is, at least in part, mediated by phospho-MARCKS and also provide an alternative therapeutic strategy against breast cancer by improving taxanes sensitivity.