Share this post on:

Dark. Cells were then harvested, washed, and resuspended in PBS and analyzed immediately using flow cytometry with the Ex488 nm/ Em525 nm.Mechanisms of Temporin-1CEa Induced 101043-37-2 site CytotoxicityFigure 6. Transmembrane potential of breast cancer cells after peptides treatment. The transmembrane potential depolarization of cancer cells (A: MDA-MB-231 and B: MCF-7) were determined using the anionic dye, DiBAC4(3). doi:10.1371/journal.pone.0060462.gDetermination of Reactive Oxygen Species (ROS) ProductionThe intracellular accumulation of ROS was determined using a sensitive free-radical indicator, 29,79-dichlorofluorescin-diacetate (DCFH-DA). The nonfluorescent DCFH can be oxidized by ROS to form green fluorescent molecule, 29,79-dichlorofluorescein (DCF). Temporin-1CEa-treated MCF-7 or MDA-MB-231 cells were incubated with 25 mM DCFH-DA in darkness for 30 min. After incubation, cells were collected, washed with PBS, resuspended in PBS and then subjected to flow cytometry for analysis.DiscussionIn the present and our previous work, we have shown that temporin-1CEa, an AMP from the skin secretions of the Chinese brown frog (Rana chensinensis), is a potent antitumor agent. AMPs usually cause cancer cells to undergo rapid cell death through a direct cell membrane-disrupting effect, but some AMPs can trigger regulated cell death through intracellular signaling mechanisms. For example, previous research reports have indicated that some AMPs may promote K562 cancer cells death through intracellular calcium mechanisms, participation of free Cucurbitacin I site radicals and caspase-3 signaling pathway [16]. Previous research has also demonstrated that LfcinB disrupts cancer cell membranes, causing the loss of membrane integrity due to the formation oftransmembrane pores that allow an uptake of the peptide into the cytoplasmic compartment of the cancer cell. The internalized LfcinB further colocalizes with the negatively charged mitochondria [25,26] and results in cell death via an apoptotic process that involves the sequential generation of reactive oxygen species, the loss of Dwm, and the activation of the caspase cascade [15]. Furthermore, certain AMPs have been shown to be potent inhibitors of tumor angiogenesis, which is associated with tumor progression [27,28]. Although temporin-1CEa treatment triggered a rapid cell death and resulted in striking morphological changes in MDA-MB-231 and MCF-7 cells, the results of the present study revealed an interesting concentration-related mechanism 15755315 involved in temporin-1CEa-induced rapid cytotoxicity. The plasma membrane disruption during temporin-1CEa exposure is a progressive doseresponse process with gradually increased permeability. After being exposed to peptides of lower concentrations, the plasma membrane became permeable to only small molecules, including PI (668 Da), EthD-1 (857 Da) or calcium. However, after being exposed to peptides of higher concentrations, the membrane was disrupted to be permeable for FITC-labeled temporin-1CEa (about 2310 Da) and LDH (140 kDa). When cancer cells were treated with 20 mM FITC-labeled temporin-1CEa, the non-detectable intracellular green fluorescence suggested that temporin-1CEa at this concentration was prevented from influx into cancer cells. The membrane-boundMechanisms of Temporin-1CEa Induced CytotoxicityFigure 7. Elevation of intracellular calcium concentration in MDA-MB-231 or MCF-7 cells following temporin-1CEa treatment in either a calcium-containing solution (A-B) or calcium-fre.Dark. Cells were then harvested, washed, and resuspended in PBS and analyzed immediately using flow cytometry with the Ex488 nm/ Em525 nm.Mechanisms of Temporin-1CEa Induced CytotoxicityFigure 6. Transmembrane potential of breast cancer cells after peptides treatment. The transmembrane potential depolarization of cancer cells (A: MDA-MB-231 and B: MCF-7) were determined using the anionic dye, DiBAC4(3). doi:10.1371/journal.pone.0060462.gDetermination of Reactive Oxygen Species (ROS) ProductionThe intracellular accumulation of ROS was determined using a sensitive free-radical indicator, 29,79-dichlorofluorescin-diacetate (DCFH-DA). The nonfluorescent DCFH can be oxidized by ROS to form green fluorescent molecule, 29,79-dichlorofluorescein (DCF). Temporin-1CEa-treated MCF-7 or MDA-MB-231 cells were incubated with 25 mM DCFH-DA in darkness for 30 min. After incubation, cells were collected, washed with PBS, resuspended in PBS and then subjected to flow cytometry for analysis.DiscussionIn the present and our previous work, we have shown that temporin-1CEa, an AMP from the skin secretions of the Chinese brown frog (Rana chensinensis), is a potent antitumor agent. AMPs usually cause cancer cells to undergo rapid cell death through a direct cell membrane-disrupting effect, but some AMPs can trigger regulated cell death through intracellular signaling mechanisms. For example, previous research reports have indicated that some AMPs may promote K562 cancer cells death through intracellular calcium mechanisms, participation of free radicals and caspase-3 signaling pathway [16]. Previous research has also demonstrated that LfcinB disrupts cancer cell membranes, causing the loss of membrane integrity due to the formation oftransmembrane pores that allow an uptake of the peptide into the cytoplasmic compartment of the cancer cell. The internalized LfcinB further colocalizes with the negatively charged mitochondria [25,26] and results in cell death via an apoptotic process that involves the sequential generation of reactive oxygen species, the loss of Dwm, and the activation of the caspase cascade [15]. Furthermore, certain AMPs have been shown to be potent inhibitors of tumor angiogenesis, which is associated with tumor progression [27,28]. Although temporin-1CEa treatment triggered a rapid cell death and resulted in striking morphological changes in MDA-MB-231 and MCF-7 cells, the results of the present study revealed an interesting concentration-related mechanism 15755315 involved in temporin-1CEa-induced rapid cytotoxicity. The plasma membrane disruption during temporin-1CEa exposure is a progressive doseresponse process with gradually increased permeability. After being exposed to peptides of lower concentrations, the plasma membrane became permeable to only small molecules, including PI (668 Da), EthD-1 (857 Da) or calcium. However, after being exposed to peptides of higher concentrations, the membrane was disrupted to be permeable for FITC-labeled temporin-1CEa (about 2310 Da) and LDH (140 kDa). When cancer cells were treated with 20 mM FITC-labeled temporin-1CEa, the non-detectable intracellular green fluorescence suggested that temporin-1CEa at this concentration was prevented from influx into cancer cells. The membrane-boundMechanisms of Temporin-1CEa Induced CytotoxicityFigure 7. Elevation of intracellular calcium concentration in MDA-MB-231 or MCF-7 cells following temporin-1CEa treatment in either a calcium-containing solution (A-B) or calcium-fre.

Share this post on:

Author: Gardos- Channel