Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Biol Pharm Bull ; 47(1): 60-71, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-37926527

ABSTRACT

Residual cancer cells after radiation therapy may acquire malignant phenotypes such as enhanced motility and migration ability, and therefore it is important to identify targets for preventing radiation-induced malignancy in order to increase the effectiveness of radiotherapy. G-Protein-coupled receptors (GPCRs) such as adenosine A2B receptor and cannabinoid receptors (CB1, CB2, and GPR55) may be involved, as they are known to have roles in proliferation, invasion, migration and tumor growth. In this study, we investigated the involvement of A2B and cannabinoid receptors in γ-radiation-induced enhancement of cell migration and actin remodeling, as well as the involvement of cannabinoid receptors in cell migration enhancement via activation of A2B receptor in human lung cancer A549 cells. Antagonists or knockdown of A2B, CB1, CB2, or GPR55 receptor suppressed γ-radiation-induced cell migration and actin remodeling. Furthermore, BAY60-6583 (an A2B receptor-specific agonist) enhanced cell migration and actin remodeling in A549 cells, and this enhancement was suppressed by antagonists or knockdown of CB2 or GPR55, though not CB1 receptor. Our results indicate that A2B receptors and cannabinoid CB1, CB2, and GPR55 receptors all contribute to γ-radiation-induced acquisition of malignant phenotypes, and in particular that interactions of A2B receptor and cannabinoid CB2 and GPR55 receptors play a role in promoting cell migration and actin remodeling. A2B receptor-cannabinoid receptor pathways may be promising targets for blocking the appearance of malignant phenotypes during radiotherapy of lung cancer.


Subject(s)
Cannabinoids , Lung Neoplasms , Humans , A549 Cells , Actins , Cannabinoids/pharmacology , Cannabinoids/metabolism , Lung Neoplasms/radiotherapy , Receptor, Adenosine A2B , Receptors, Cannabinoid
2.
Biol Pharm Bull ; 40(6): 878-887, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28344198

ABSTRACT

Melanoma is highly malignant, and generally exhibits radioresistance, responding poorly to radiation therapy. We previously reported that activation of P2X7, P2Y6, and P2Y12 receptors is involved in the DNA damage response after γ-irradiation of human lung adenocarcinoma A549 cells. However, it is not clear whether these receptors are also involved in the case of melanoma cells, although P2X7 receptor is highly expressed in various cancers, including melanoma. Here, we show that P2X7 receptor antagonist enhances radiation-induced cytotoxicity in B16 melanoma cells in vitro and in vivo. We confirmed that these cells express P2X7 receptor mRNA and exhibit P2X7 receptor-mediated activities, such as ATP-induced pore formation and cytotoxicity. We further examined the radiosensitizing effect of P2X7 receptor antagonist Brilliant Blue G (BBG) in vitro by colony formation assay of B16 cells. γ-Irradiation dose-dependently reduced cell survival, and pretreatment with BBG enhanced the radiation-induced cytotoxicity. BBG pretreatment also decreased the number of DNA repair foci in nuclei, supporting involvement of P2X7 receptor in the DNA damage response. Finally, we investigated the radiosensitizing effect of BBG on B16 melanoma cells inoculated into the hind footpad of C57BL/6 mice. Neither 1 Gy γ-irradiation alone nor BBG alone suppressed the increase of tumor volume, but the combination of irradiation and BBG significantly suppressed tumor growth. Our results suggest that P2X7 receptor antagonist BBG has a radiosensitizing effect in melanoma in vitro and in vivo. BBG, which is used as a food coloring agent, appears to be a promising candidate as a radiosensitizer.


Subject(s)
Gamma Rays/therapeutic use , Melanoma, Experimental/therapy , Purinergic P2X Receptor Antagonists/therapeutic use , Radiation-Sensitizing Agents/therapeutic use , Receptors, Purinergic P2X7/metabolism , Rosaniline Dyes/therapeutic use , Animals , Cell Line, Tumor , DNA Damage , DNA Repair , Humans , Male , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Purinergic P2X Receptor Antagonists/pharmacology , Radiation-Sensitizing Agents/pharmacology , Receptors, Purinergic P2X7/genetics , Rosaniline Dyes/pharmacology , Tumor Burden/drug effects
3.
Biol Pharm Bull ; 39(7): 1224-30, 2016 Jul 01.
Article in English | MEDLINE | ID: mdl-27150432

ABSTRACT

Radiosensitizers are used in cancer therapy to increase the γ-irradiation susceptibility of cancer cells, including radioresistant hypoxic cancer cells within solid tumors, so that radiotherapy can be applied at doses sufficiently low to minimize damage to adjacent normal tissues. Radiation-induced DNA damage is repaired by multiple repair systems, and therefore these systems are potential targets for radiosensitizers. We recently reported that the transient receptor potential vanilloid type 1 (TRPV1) channel is involved in early responses to DNA damage after γ-irradiation of human lung adenocarcinoma A549 cells. Therefore, we hypothesized that TRPV1 channel inhibitors would have a radiosensitizing effect by blocking repair of radiation-induced cell damage. Here, we show that pretreatment of A549 cells with the TRPV1 channel inhibitors capsazepine, AMG9810, SB366791 and BCTC suppressed the γ-ray-induced activation of early DNA damage responses, i.e., activation of the protein kinase ataxia-telangiectasia mutated (ATM) and accumulation of p53-binding protein 1 (53BP1). Further, the decrease of survival fraction at one week after γ-irradiation (2.0 Gy) was enhanced by pretreatment of cells with these inhibitors. On the other hand, inhibitor pretreatment did not affect cell viability, the number of apoptotic or necrotic cells, or DNA synthesis at 24 h after irradiation. These results suggest that inhibition of DNA repair by TRPV1 channel inhibitors in irradiated A549 cells caused gradual loss of proliferative ability, rather than acute facilitation of apoptosis or necrosis. TRPV1 channel inhibitors could be novel candidates for radiosensitizers to improve the efficacy of radiation therapy, either alone or in combination with other types of radiosensitizers.


Subject(s)
Gamma Rays , Radiation-Sensitizing Agents/pharmacology , TRPV Cation Channels/antagonists & inhibitors , Acrylamides/pharmacology , Anilides/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Capsaicin/analogs & derivatives , Capsaicin/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Cinnamates/pharmacology , DNA Damage/drug effects , Humans , Mice , Necrosis/chemically induced , Neoplasms/metabolism , Pyrazines/pharmacology , Pyridines/pharmacology , TRPV Cation Channels/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...