• Title/Summary/Keyword: Arsenic trioxide

Search Result 39, Processing Time 0.035 seconds

Effects of Arsenic Trioxide on Cell Cycle Related Proteins (Cyclin D1, p21, p27) Expression During Urethane-induced Lung Carcinogenesis in Mice (Urethane으로 유발된 생쥐 폐샘암종 발생과정에서 세포주기 관련인자(Cyclin D1, p21, and p27)에 대한 비소의 효과)

  • Yim Sung-Hyuk;Jeong Ji-Hoon;Gyeon Jong-Man;Park Eon-Sub
    • YAKHAK HOEJI
    • /
    • v.50 no.2
    • /
    • pp.84-92
    • /
    • 2006
  • The present study investigated an effect of arsenic trioxide on the urethane-induced lung carcinogenesis in mice. To understand its carcinogenesis, we examined proliferating cell nuclear antigen (PCNA), apoptotic index as well as cell cycle-related proteins (cyclin D1, p21, and p27). Urethane was injected intraperitoneally in ICR mice, and then they were sacrificed at 5, 15, or 25 weeks following treatment of arsenic trioxide. Arsenic trioxide was given with tap water at a concentration of 1 mg/l (low-dose) and 5mg/1 (high-dose) for 25 weeks. During the carcinogenesis, sequential histological changes from hyperplasia to adenomas, and ultimately to overt carcinomas were noted. The development of hyperplasias, adenomas, and carcinomas in the lung were slightly increased by the treatment of low-dose arsenic trioxide. However, there is no correlation between dose and tumor multiplicity. The administration of low-dose arsenic trioxide, significantly increased the tumor size. The proliferative index observed on 5 weeks after significantly increased. Cyclin D1 and p21 protein, cell cycle related proteins, were more significantly increased in hyperplasia and adenoma in low dose arsenic treated group than urethane alone group. The p27 protein expression did not show any significantly changes with arsenic treated or untreated group. Low dose exposure to arsenic trioxide resulted in increased expression of cyclin D1 and p21 protein. The present results indicate that low-dose treatment of arsenic trioxide, but not high dose of it, partly modulate the cellular proliferation, cyclin D1, and p21 protein expression, and that this effect may contribute to accelerated development of lung adenocarcinomas in urethane-induced mice.

OSTEOMYELITIS ON MAXILLA CAUSED BY ARSENIC TRIOXIDE (비소에 의해 유도된 상악골 골수염의 증례보고)

  • Choi, Bo-Young;Yoo, Dae-Hyun;Choi, Mun-Ki;Choi, Jung-Goo;Lee, Young-Jin;Jo, Byung-Ho
    • Maxillofacial Plastic and Reconstructive Surgery
    • /
    • v.30 no.6
    • /
    • pp.593-598
    • /
    • 2008
  • Arsenic trioxide is one of the 'tooth pulp devitalizing agents' used through the dental history when anaesthesia was not available. But owing to its capacity to kill cells in surrounding tissues, the use of arsenic trioxide in vital pulpotomy has been reduced. Arsenic trioxide can cause necrosis of gingiva, bone which can cause osteomyelitis of the jaws. But some dentists still continue to use arsenic trioxide in their endodontic practices. The purpose of this article is to present arsenic trioxide induced osteomyelitis on maxilla and treatment process.

The anti-tumor effect of combined treatment with arsenic trioxide and interferone-α on transplanted murine Lewis lung carcinoma

  • Seo, Deug-Log;Yang, Je-Hoon;Won, Chung-Kil;Kim, Myeong-Ok;Lee, Jong-Hwan;Kwark, Soo-Dong;Koh, Phil-Ok
    • Korean Journal of Veterinary Research
    • /
    • v.45 no.1
    • /
    • pp.17-23
    • /
    • 2005
  • In the present study, we expected the anti-tumor effect by combined treatment of arsenic trioxide and interferon (IFN)-${\alpha}$ on murine Lewis lung carcinoma (LL2) cells through in vivo study. As a experimental model, LL2 cells ($1{\times}10^{6}$/mouse) were injected subcutaneously into the back region of mice. When the tumor volume reached $100mm^3$, mice were treated with 1 mg/kg arsenic trioxide, 50000 IU IFN-${\alpha}$, or arsenic trioxide and IFN-${\alpha}$. The development of tumor cells was significantly inhibited by combined treatment with arsenic trioxide and IFN-${\alpha}$. In arsenic trioxide and IFN-${\alpha}$ treated group, apoptotic index was reached a peak valve at 48 hr after the treatment and it was restored to approximately the control level at 8 days. Also, positive signals of Bax and Bad were increased at 48 to 96 hr and decreased at 8 day. Whereas, positive cells of Bcl-2 were steadily decreased at 12 to 48 hr and restored to the background level at 8 days. Our data showed that immunoreactivity of Bcl-2 was decreased at 12 to 48 hr, while positive signals of Bax and Bad were increased in accordance with apoptotic index at these times. In conclusion, our results suggest that the combined treatment with arsenic trioxide and IFN-${\alpha}$ significantly inhibited the growth of LL2 tumor cells and induced apoptosis through the up and down-regulation of Bcl-2 gene family.

Different Responses to Arsenic Trioxide between NB4 and UF-1, Acute Promyelocytic Leukemia Cell Lines (급성 전골수성 백혈병 세포주간의 삼산화비소에 대한 반응)

  • Kim, Hye-Ran;Choi, Yoon-Jeong;Ryu, Seong-Yeoll;Lee, Young-Seok;Lee, Sang-Hwa
    • Journal of Life Science
    • /
    • v.16 no.5
    • /
    • pp.759-766
    • /
    • 2006
  • Acute promyelocytic leukemia (APL) is a myeloid leukemia caused by over-expression of fusion protein, PML/RAR$({\alpha})$, which was the result of chromosomal translocation and induces the blockage of differentiation of affected promyelocytes. Pharmacological dose of retinoic acid induces the activation of and subsequent degradation of PML/RAR$({\alpha})$ fusion protein, and then APL cells undergo through the normal differentiation pathway. Arsenic trioxide has proved effective in causing remission of acute promyelocytic leukemia by inducing apoptosis of this tumor cells, whereas the heterogeneity of cellular susceptibility to this cytotoxic agent limited its usage on more types of tumors in clinic. This work showed that arsenic trioxide could induce apoptosis of a panel of acute promyelocytic leukemic cell lines, all-trans-retinoic acid (ATRA) sensitive NB4 cells and ATRA resistant UF-1 cell. They were investigated with regard to the correlation between the inherent or intrinsic cellular level of GSH and the apoptotic susceptibility of the cells to arsenic trioxide. We manifested, in two cell types, the inherently existed difference in intracellular GSH level reactive to the arsenic trioxide, and a positive correlation between the GSH level and their apoptotic sensitivity to arsenic trioxide. And it showed that arsenic trioxide could differentiate promyelocytic cancer cells to the cells possessed of dendritic cell surface markers. Unravelling the cause of the different susceptibility between leukemic cells and proving that promyelocyte could be differentiated to dendritic cells by arsenic trioxide will help not only to understand the mechanism underlying the complete remission of acute promyelocytic leukemia induced by arsenic trioxide, but also to expand its clinical usage.

Inducing Apoptosis of NCI-H157 Human Lung Carcinoma Cells via Activation of Caspase Cascade by Combination Treatment with Arsenic Trioxide and Sulindac (NCI-H157 폐암 세포주에서 Caspase Cascade 활성을 통한 Arsenic Trioxide와 Sulindac 병합요법의 세포고사효과)

  • Kim, Hak Ryul;Yang, Sei Hoon;Jeong, Eun Taik
    • Tuberculosis and Respiratory Diseases
    • /
    • v.56 no.4
    • /
    • pp.381-392
    • /
    • 2004
  • Arsenic trioxide($As_2O_3$) was introduced into the treatment of refractory or relapsed acute promyelocytic Ieukemia. Some investigators have reported that arsenic trioxide had induced apoptosis in a variety of solid human tumor cell lines, including non-small cell lung cancer. Non-steroidal anti-inflammatory drugs(NSAIDs) are powerful chemopreventive agents for gastrointestinal cancers and the growth of established tumors are reduced by inducing apoptosis. It's also reported that NSAIDs enhanced tumor response to chemotherapeutic drugs or radiation. In this study, we aimed to determine whether combination of arsenic trioxide with sulindac augmented its apoptotic potential in NCI-H157 human lung cancer cells. The human lung cancer cell line NCI-H157 was treated with arsenic trioxide and sulindac. Cell viability was measured by the MTT assay. Apoptosis was measured by nuclear staining and flow cytometric analysis. The catalytic activity of the caspase families were measured by the fluorogenic cleavage of biosubstrates. The western blotting were also performed to define the mechanical basis of apoptosis. Combination treatment of arsenic trioxide and sulindac decreased the viability of NCI-H157 human lung cancer cells in a dose-dependent manner. The catalytic activity of caspase-3, 8 and 9 proteases were increased after combination treatment. Consistently PARP was cleaved from 116kDa to 85kDa fragments, and the expression of ICAD was decreased by time-dependent manner. Also combination treatment increased the expression of Fas and Fas/L. Combination therapy of arsenic trioxide with sulindac augments cell death and induces apoptosis via the activation of caspase cascade in NCI-H157 human lung carcinoma cells.

Combined Treatment with Coptidis Rhizoma Extract and Arsenic Trioxide Enhanced Apoptosis through Diverse Pathways in H157 Cells

  • Youn, Myung-Ja;Kim, Yun-Ha;Kim, Hyung-Jin;Song, Je-Ho;Jeon, Ho-Sung;Yu, Dong-Hee;Sul, Jeong-Dug;So, Hong-Seob;Park, Rae-Kil
    • Journal of Physiology & Pathology in Korean Medicine
    • /
    • v.23 no.6
    • /
    • pp.1449-1459
    • /
    • 2009
  • Coptidis rhizoma (huanglian) is an herb that is widely used in traditional Chinese medicine that has recently been shown to possess anticancer activity. However, the molecular mechanism underlying the anticancer effects of this herb is poorly understood. In this study, we investigated the anticancer activity of a combination of CR extract and arsenic trioxide, as well as the apoptotic pathway associated with its mechanism of action in human lung cancer H157 cells. Combined treatment of H157 cells with CR extract and arsenic trioxide resulted in significant apoptotic death. In addition, combined treatment with CR extract and arsenic trioxide acted in concert to induce a loss of mitochondrial membrane potential (${\Delta}{\Psi}$), the release of cytochrome c from mitochondria, and an increase in the expression of pro-apoptotic p53 and Bax protein, which resulted in activation of caspases and apoptosis. CR extract combined with arsenic trioxide also increased the lipid peroxidation, mRNA expression of DR4 and DR5 and caspase-8 activity. These data indicate that combined treatment with CR extract and arsenic trioxide enhanced apoptotic cell death in H157 cells through diverse pathways, including mitochondrial dysfunction and death receptors, particularly DR4 and DR5. Thus, this treatment may be an effective from of chemotherapy.

The Effect of Taxol and Arsenic Trioxide in HT-29 Spheroid Cells

  • Lee In-Soo;Choi Hyun-Il;Han Hye-Eun;Lee Hye-Young;Kim Tae-Ue
    • Biomedical Science Letters
    • /
    • v.12 no.3
    • /
    • pp.153-160
    • /
    • 2006
  • Human colon cancer is the second most fatal disease among a variety of cancers to cause cancer death in U.S.A. and its incidence rate is currently increased in Korea. Recently, many studies have been being progressed on the efficacy of diverse combination treatments. But results of these studies in vitro were not similar those in vivo. This study compared the anticancer reactions between each use of arsenic trioxide and taxol against human colon cancer HT-29 cell line and combined use of two drugs. And these results compared with the results of HT-29 spheroid cells having similar characteristics to the solid tumor in vivo. The spheroid of HT-29 cells was formed by using a multicellular spheroid system and the result was observed through electron microscopy. In vitro cytotoxicity of each use of arsenic trioxide and taxol was evaluated in HT-29 monolayer cells. The $IC_{50}$ value for arsenic trioxide was to be $33{\mu}M$ and taxol was to be 18nM. The result treated with the combination of taxol and arsenic trioxide decreased the cytotoxicity on the HT-29 monolayer cells. The spheroid cells represented higher resistance against drugs than the monolayer cells. I demonstrated DNA fragmentation after incubation with concentrations more than $10{\mu}M$ arsenic trioxide and 100nM taxol for 48h, on the monolayer cells. But the results of HT-29 cell line treated with the combination of taxol and arsenic trioxide was the same as the outcome of control samples that were not treated with any drug. And I don't demonstrated DNA fragmentation on the spheroid cells. These results suggest that apoptosis was not induced in the use of the combination can be thought as that arsenic trioxide might work as an antagonist to inhibit a taxol mechanism to induce apoptosis. And the spheroid cells represented higher resistance against drugs than the monolayer cells.

  • PDF

Arsenic Trioxide Promotes Paclitaxel Cytotoxicity in Resistant Breast Cancer Cells

  • Bakhshaiesh, Tayebeh Oghabi;Armat, Marzie;Shanehbandi, Dariush;Sharifi, Simin;Baradaran, Behzad;Hejazi, Mohammad Saeed;Samadi, Nasser
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.16 no.13
    • /
    • pp.5191-5197
    • /
    • 2015
  • A partial response or resistance to chemotherapeutic agents is considered as a main obstacle in treatment of patients with cancer, including breast cancer. Refining taxane-based treatment procedures using adjuvant or combination treatment is a novel strategy to increase the efficiency of chemotherapy. PPM1D is a molecule activated by reactive oxygen species. whose expression is reported to modulate the recruitment of DNA repair molecules. In this study we examined the impact of arsenic trioxide on efficacy of paclitaxel-induced apoptosis in paclitaxel-resistant MCF-7 cells. We also investigated the expression of PPM1D and TP53 genes in response to this combination treatment. Resistant cells were developed from the parent MCF-7 cell line by applying increasing concentrations of paclitaxel. MTT assays were applied to determine the rate of cell survival. DAPI staining using fluorescent microscopy was employed to study apoptotic bodies. Real-time RT-PCR analysis was also applied to determine PPM1D mRNA levels. Our results revealed that combination of arsenic trioxide and paclitaxel elevates the efficacy of the latter in induction of apoptosis in MCF-7/PAC resistant cells. Applying arsenic trioxide also caused significant decreases in PPM1D mRNA levels (p<0.05). Our findings suggest that arsenic trioxide increases paclitaxel-induced apoptosis by down regulation of PPM1D expression. PPM1D dependent signaling can be considered as a novel target to improve the efficacy of chemotherapeutic agents in resistant breast cancer cells.

Suppression of Arsenic Trioxide-induced Apoptosis in HeLa Cells by N-Acetylcysteine

  • Han, Yong Hwan;Kim, Sung Zoo;Kim, Suhn Hee;Park, Woo Hyun
    • Molecules and Cells
    • /
    • v.26 no.1
    • /
    • pp.18-25
    • /
    • 2008
  • Arsenic trioxide (ATO) can affect many biological functions such as apoptosis and differentiation in various cells. We investigated the involvement of ROS and GSH in ATO-induced HeLa cell death using ROS scavengers, especially N-acetylcysteine (NAC). ATO increased intracellular ${O_2}^{{\cdot}-}$ levels and reduced intracellular GSH content. The ROS scavengers, Tempol, Tiron and Trimetazidine, did not significantly reduce levels of ROS or GSH depletion in ATO-treated HeLa cells. Nor did they reduce the apoptosis induced by ATO. In contrast, treatment with NAC reduced ROS levels and GSH depletion in the ATO-treated HeLa cells and prevented ATO-induced apoptosis. Treatment with exogenous SOD and catalase reduced the depletion of GSH content in ATO-treated cells. Catalase strongly protected the cells from ATO-induced apoptosis. In addition, treatment with SOD, catalase and NAC slightly inhibited the G1 phase accumulation induced by ATO. In conclusion, NAC protects HeLa cells from apoptosis induced by ATO by up-regulating intracellular GSH content and partially reducing the production of ${O_2}^{{\cdot}-}$.

Oxidative Stress by Arsenic Trioxide in Cultured Rat Cardiomyocytes, $H_9C_2$ Cells (배양 심근세포에서 저농도 삼산화비소에 의한 산화적 스트레스 발생)

  • Park Eun-Jung;Park Kwang-Sik
    • Environmental Analysis Health and Toxicology
    • /
    • v.21 no.1 s.52
    • /
    • pp.71-79
    • /
    • 2006
  • Epidemiologic studies have showed a close correlation between arsenic exposure and heart disease such as, cardiovascular problem, ischemic heart disease, infarction, atherosclerosis and hypertension in human. It may increase the mortality of high risk group with heart disease. Regarding the mechanism studies of heart failure, blood vessel, vascular smooth muscle cells and endothelial cells have long been focused as the primary targets in arsenic exposure but there are only a few studies on the cardiomyocytes. In this study, the generation of oxidative stress by low dose of arsenic trioxide was investigated in rat cardiomyocytes. By direct measurement of reactive oxygen species and fluorescent microscopic observation using fluorescent dye 2',7'-dichlorofluorescin diacetate, reactive oxygen species were found to be generated without cell death, where cells are treated with 0.1 ppm arsenic for 24 hours. With the induction of reactive oxygen species, GSH level was decreased by the same treatment. However, DNA damage did not seem to be serious by DAPI staining, while high dose of arsenic (2 ppm for 24 hrs) caused fragmentation of DNA. To identify the molecular biomarkers of low-dose arsenic exposure, gene expression was also investigated with whole genome microarray. As results, 9,022 genes were up-regulated including heme oxygenase-l and glutathione S-transrerase, which are well-known biomarkers of oxidative stress. 9,404 genes were down-regulated including endothelial type gp 91-phox gene by the treatment of 0.1 ppm arsenic for 24 hours. This means that biological responses of cardiomyocytes may be altered by ROS induced by low level arsenic without cell death, and this alteration may be detected clearly by molecular biomarkers such as heme oxygenase-1.