Title

Rational Development of a Cytotoxic Peptide To Trigger Cell Death

Authors

Authors

R. J. Boohaker; G. Zhang; M. W. Lee; K. N. Nemec; S. Santra; J. M. Perez;A. R. Khaled

Comments

Authors: contact us about adding a copy of your work at STARS@ucf.edu

Abbreviated Journal Title

Mol. Pharm.

Keywords

necrosis; nanoparticles; metabolism; mitochondria; lipid membranes; CHRONIC LYMPHOCYTIC-LEUKEMIA; ANTIMICROBIAL PEPTIDES; PROAPOPTOTIC; PEPTIDE; ANTICANCER ACTIVITY; APOPTOTIC PROTEINS; CANCER-CELLS; SOLID; TUMORS; IN-VIVO; BAX; NANOPARTICLES; Medicine, Research & Experimental; Pharmacology & Pharmacy

Abstract

Defects in the apoptotic machinery can contribute to tumor formation and resistance to treatment, creating a need to identify new agents that kill cancer cells by alternative mechanisms. To this end, we examined the cytotoxic properties of a novel peptide, CT20p, derived from the C-terminal, alpha-9 helix of Box, an amphipathic domain with putative membrane binding properties. Like many antimicrobial peptides, CT20p contains clusters of hydrophobic and cationic residues that could enable the peptide to associate with lipid membranes. CT20p caused the release of calcein from mitochondrial-like lipid vesicles without disrupting vesicle integrity and, when expressed as a fusion protein in cells, localized to mitochondria. The amphipathic nature of CT20p allowed it to be encapsulated in polymeric nanoparticles (NPs) that have the capacity to harbor targeting molecules, dyes or drugs. The resulting CT20p-NPs proved an effective killer, in vitro, of colon and breast cancer cells, and in vivo, using a murine breast cancer tumor model. By introducing CT20p to Bax deficient cells, we demonstrated that the peptide's lethal activity was independent of endogenous Bax. CT20p also caused an increase in the mitochondria] membrane potential that was followed by plasma membrane rupture and cell death, without the characteristic membrane asymmetry associated with apoptosis. We determined that cell death triggered by the CT20p-NPs was minimally dependent on effector caspases and resistant to Bcl-2 overexpression, suggesting that it acts independently of the intrinsic apoptotic death pathway. Furthermore, use of CT20p with the apoptosis-inducing drug, cisplatin, resulted in additive toxicity. These results reveal the novel features of CT20p that allow nanoparticle-mediated delivery to tumors and the potential application in combination therapies to activate multiple death pathways in cancer cells.

Journal Title

Molecular Pharmaceutics

Volume

9

Issue/Number

7

Publication Date

1-1-2012

Document Type

Article

Language

English

First Page

2080

Last Page

2093

WOS Identifier

WOS:000305917600022

ISSN

1543-8384

Share

COinS