1
|
Ghorbanlou M, Moradi F, Shabani R, Mehdizadeh M. Upregulation of apoptotic genes and downregulation of target genes of Sonic Hedgehog signaling pathway in DAOY medulloblastoma cell line treated with arsenic trioxide. J Chemother 2024; 36:506-519. [PMID: 38130211 DOI: 10.1080/1120009x.2023.2294574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 12/05/2023] [Accepted: 12/07/2023] [Indexed: 12/23/2023]
Abstract
Sonic hedgehog (SHH) medulloblastoma etiology is associated with the SHH molecular pathway activation at different levels. We investigated the effect of arsenic trioxide as a downstream-level inhibitor of the SHH signaling pathway on morphology, cytotoxicity, migration, and SHH-related and apoptotic gene expression of DAOY cells. Cells were treated at various arsenic trioxide (ATO)concentrations (1, 2, 3, 5, and 10 μM) for different times (24 and 48 hr). Following treatments, the morphology of the cells was investigated at ×20 and ×40 magnification by an inverted microscope. Then, cytotoxicity was investigated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and trypan blue assays. Cell migration was analyzed through the wound-healing assay. Furthermore, the expression of SHH-related (GLI1, GLI2, SMO, and MYCN) and apoptotic genes (BAX, BCL2, and TP53) was assessed by real-time quantitative polymerase chain reaction (qPCR). Finally, GLI1, SMO, and MYCN markers were analyzed through immunocytochemistry. Data were analyzed by SPSS (version 16) and P≤0.05 was considered significant. Morphological changes were seen at 3 and 2 μM in 24 and 48 hr of treatment, respectively. The MTT assay showed a dose-dependent cytotoxicity indicating an IC50 value of 3.39±0.35 and 2.05±0.64 μM in 24 and 48hr treatment, respectively. In addition, the trypan blue assay showed higher IC50 values of 4.29±0.25 and 3.92±0.22 μM in 24 and 48 hr treatment, respectively. The wound-healing assay indicated a dose-dependent reduction of cell migration speed showing a 50% reduction at 2.89±0.26 μM. Significant downregulation of GLI1 and GLI2, as well as the upregulation of BAX, BAX/BCL2 ratio, and TP53 were evident. Significant increases in GLI1 and MYCN markers were also evident in immunocytochemistry. ATO, as a downstream effective inhibitor of the SHH pathway, substantially leads to cell death, cell migration inhibition, apoptosis upregulation, and downregulation of SHH target genes in DAOY medulloblastoma. Since ATO is a toxic chemotherapeutic agent, it must be used at low concentrations (2 μM) in order not to damage healthy cells.
Collapse
Affiliation(s)
- Mehrdad Ghorbanlou
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Moradi
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Reproductive Sciences and Technology Research Center, Department of Anatomy, Iran University of Medical Sciences, Tehran, Iran
| | - Ronak Shabani
- Reproductive Sciences and Technology Research Center, Department of Anatomy, Iran University of Medical Sciences, Tehran, Iran
| | - Mehdi Mehdizadeh
- Reproductive Sciences and Technology Research Center, Department of Anatomy, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Firdaus F, Zafeer MF, Anis E, Ahmad F, Hossain MM, Ali A, Afzal M. Evaluation of phyto-medicinal efficacy of thymoquinone against Arsenic induced mitochondrial dysfunction and cytotoxicity in SH-SY5Y cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 54:224-230. [PMID: 30668372 DOI: 10.1016/j.phymed.2018.09.197] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 09/17/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND It is evaluated that a few million individuals worldwide are experiencing Arsenic (As) harmfulness coming about because of anthropogenic discharges. There is likewise proof to propose that As can affect the peripheral, as well as, the central nervous system (CNS). On the contrary, thymoquinone (TQ), a biologically active ingredient of Nigella sativa has exhibited numerous neuro-pharmacological traits since ancient times. HYPOTHESIS/PURPOSE In the present study, the neuroprotective efficacy of TQ was explored by primarily studying its antioxidant and anti-apoptotic potential against Arsenic trioxide (As2O3) induced toxicity in SH-SY5Y human neuroblastoma cell lines. STUDY DESIGN For experimentation, cells were seeded in 96 well tissue culture plates and kept undisturbed for 24 h to attain proper adhesion. After 75-80% confluence, cells were pretreated with 10 µM and 20 µM thymoquinone (TQ) for 1 h After adding 2 µM As, cells were set aside for incubation for 24 h without changing the medium. METHODS The mitigatory effects of TQ with particular reference to cell viability and cytotoxicity, the generation of reactive oxygen species, DNA damage, and mitochondrial dynamics were studied. RESULTS Pretreatment of SH-SY5Y cells with TQ (10 and 20 μM) for an hour and subsequent exposure to 2 μM As2O3 protected the SH-SY5Y cells against the neuro-damaging effects of the latter. Also, the SH-SY5Y cells were better preserved with increased viability, repaired DNA, less free radical generation and balanced transmembrane potential than those exposed to As2O3 alone. TQ pretreatment also inhibited As2O3-induced exacerbation in protein levels of BAX and PARP-1 and restored the loss of Bcl2 levels. CONCLUSION The findings of this study suggest that TQ may prevent neurotoxicity and As2O3-induced apoptosis and cytotoxicity. It is, therefore, worth studying further for its potential to reduce the risks of arsenic-related neurological implications.
Collapse
Affiliation(s)
- Fakiha Firdaus
- Interdisciplinary Brain Research Centre, Faculty of Medicine, Aligarh Muslim University, Aligarh, Uttar Pradesh, India; Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, Uttar Pradesh, India.
| | - Mohd Faraz Zafeer
- Interdisciplinary Brain Research Centre, Faculty of Medicine, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Ehraz Anis
- Interdisciplinary Brain Research Centre, Faculty of Medicine, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Faraz Ahmad
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - M Mobarak Hossain
- Interdisciplinary Brain Research Centre, Faculty of Medicine, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Asif Ali
- Interdisciplinary Brain Research Centre, Faculty of Medicine, Aligarh Muslim University, Aligarh, Uttar Pradesh, India; Department of Biochemistry, Faculty of Medicine, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Mohammad Afzal
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| |
Collapse
|
3
|
Xiong X, Li Y, Liu L, Qi K, Zhang C, Chen Y, Fang J. Arsenic trioxide induces cell cycle arrest and affects Trk receptor expression in human neuroblastoma SK-N-SH cells. Biol Res 2018; 51:18. [PMID: 29898774 PMCID: PMC5998579 DOI: 10.1186/s40659-018-0167-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 06/06/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Arsenic trioxide (As2O3), a drug that has been used in China for approximately two thousand years, induces cell death in a variety of cancer cell types, including neuroblastoma (NB). The tyrosine kinase receptor (Trk) family comprises three members, namely TrkA, TrkB and TrkC. Various studies have confirmed that TrkA and TrkC expression is associated with a good prognosis in NB, while TrkB overexpression can lead to tumor cell growth and invasive metastasis. Previous studies have shown that As2O3 can inhibit the growth and proliferation of a human NB cell line and can also affect the N-Myc mRNA expression. It remains unclear whether As2O3 regulates Trks for the purposes of treating NB. METHODS The aim of the present study was to investigate the effect of As2O3 on Trk expression in NB cell lines and its potential therapeutic efficacy. SK-N-SH cells were grown with increasing doses of As2O3 at different time points. We cultured SK-N-SH cells, which were treated with increasing doses of As2O3 at different time points. Trk expression in the NB samples was quantified by immunohistochemistry, and the cell cycle was analyzed by flow cytometry. TrkA, TrkB and TrkC mRNA expression was evaluated by real-time PCR analysis. RESULTS Immunohistochemical and real-time PCR analyses indicated that TrkA and TrkC were over-expressed in NB, and specifically during stages 1, 2 and 4S of the disease progression. TrkB expression was increased in stage 3 and 4 NB. As2O3 significantly arrested SK-N-SH cells in the G2/M phase. In addition, TrkA, TrkB and TrkC expression levels were significantly upregulated by higher concentrations of As2O3 treatment, notably in the 48-h treatment period. Our findings suggested that to achieve the maximum effect and appropriate regulation of Trk expression in NB stages 1, 2 and 4S, As2O3 treatment should be at relatively higher concentrations for longer delivery times;however, for NB stages 3 and 4, an appropriate concentration and infusion time for As2O3 must be carefully determined. CONCLUSION The present findings suggested that As2O3 induced Trk expression in SK-N-SH cells to varying degrees and may be a promising adjuvant to current treatments for NB due to its apoptotic effects.
Collapse
Affiliation(s)
- Xilin Xiong
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China
- Pediatric Hematology/Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China
| | - Yang Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China
- Pediatric Hematology/Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China
| | - Ling Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China
- Department of Pediatric Hematology/Oncology, Affiliated Hospital of Guangdong Medical College, Zhanjiang, 524000 Guangdong China
| | - Kai Qi
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China
- Pediatric Hematology/Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China
| | - Chi Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China
- Pediatric Hematology/Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China
| | - Yueqin Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China
- Department of Life Sciences, Sun Yat-Sen University, Guangzhou, 510120 Guangdong China
| | - Jianpei Fang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China
- Pediatric Hematology/Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China
| |
Collapse
|
4
|
Liu L, Li Y, Xiong X, Qi K, Zhang C, Fang J, Guo H. Low dose of arsenic trioxide inhibits multidrug resistant-related P-glycoprotein expression in human neuroblastoma cell line. Int J Oncol 2016; 49:2319-2330. [PMID: 27840903 DOI: 10.3892/ijo.2016.3756] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 08/22/2016] [Indexed: 11/06/2022] Open
Abstract
This study investigated arsenic trioxide (As2O3), cisplatin (DDP) and etoposide (Vp16) on the anticancer effects and P-glycoprotein (P-gp) expression in neuroblastoma (NB) SK-N-SH cells. The potential influence of As2O3, DDP and Vp16 currently included in NB routine treatment protocols on cytotoxicity in SK-N-SH cells was measured by flow cytometry and drug half-maximal inhibitory concentration (IC50) was established. Moreover, chemotherapeutic agent-mediated changes of cellular expression levels of resistant-related P-gp, was monitored using western blotting. The data showed that As2O3, DDP and Vp16 significantly inhibited the growth and survival of the SK-N-SH cells at different concentration. Notably, the levels of apoptosis were upregulated in SK-N-SH cells with an acceleration of the exposure time and the concentration of As2O3, DDP and Vp16. As2O3, DDP and Vp16 were observed with their IC50 values on SK-N-SH cells being 3 µM, 8 and 100 µg/ml, respectively. Flow cytometry analysis showed that As2O3 at low concentrations in SK-N-SH cells led to enhanced accumulation of cell populations in G2/M phase with increasing the exposure time, and increased levels of apoptosis. In contrast, we observed that SK-N-SH cell populations arrested in S phase by DDP and Vp16. In vitro examination revealed that following pretreatment of SK-N-SH cells with As2O3, the expression of P-gp was not increased. The expression of P-gp downregulation were noted following the group treated by As2O3 at 2 and 3 µM. Exposed to As2O3 at 3 µM for 72 h, SK-N-SH cells exhibited lower expression of P-gp than 2 µM As2O3 for 72 h. In contrast, the expression of P-gp was upregulated by DDP and VP16. In summary, SK-N-SH cells were responsive to chemotherapeutic agent-induced apoptosis in a dose-dependent and time-dependent manner. In particular, ours findings showed that low dose of As2O3 markedly reduced the P-gp expression and increased apoptotic cell death in human NB cell line.
Collapse
Affiliation(s)
- Ling Liu
- Department of Pediatric, Affiliated Hospital of Guangdong Medical University, Zhan Jiang, Guangdong 524000, P.R. China
| | - Yang Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Xilin Xiong
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Kai Qi
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Chi Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Jianpei Fang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Haixia Guo
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| |
Collapse
|
5
|
Nakaoka T, Ota A, Ono T, Karnan S, Konishi H, Furuhashi A, Ohmura Y, Yamada Y, Hosokawa Y, Kazaoka Y. Combined arsenic trioxide-cisplatin treatment enhances apoptosis in oral squamous cell carcinoma cells. Cell Oncol (Dordr) 2014; 37:119-29. [PMID: 24599717 DOI: 10.1007/s13402-014-0167-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2014] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) accounts for the majority of oral cancers. Despite recent advances in OSCC diagnostics and therapeutics, the overall survival rate still remains low. Here, we assessed the efficacy of a combinatorial arsenic trioxide (ATO) and cisplatin (CDDP) treatment in human OSCC cells. METHODS The combinatorial effect of ATO/CDDP on the growth and apoptosis of OSCC cell lines HSC-2, HSC-3, and HSC-4 was evaluated using MTT and annexin V assays, respectively. Chou-Talalay analyses were preformed to evaluate the combinatorial effects of ATO/CDDP on the dose-reduction index (DRI). To clarify the mechanism underlying the ATO/CDDP anticancer effect, we also examined the involvement of reactive oxygen species (ROS) in ATO/CDDP-induced apoptosis. RESULTS Combination index (CI) analyses revealed that a synergistic interaction of ATO and CDDP elicits a wide range of effects in HSC-2 cells, with CI values ranging from 0.78 to 0.90, where CI < 1 defines synergism. The CI values in HSC-3 and HSC-4 cells ranged from 0.34 to 0.45 and from 0.60 to 0.92, respectively. In addition, ATO/CDDP yielded favorable DRI values ranging from 1.6-fold to 7.71-fold dose reduction. Compared to mono-therapy, ATO/CDDP combinatorial therapy significantly augmented the loss of mitochondrial potential, caspase-3/7 activity and subsequent apoptosis. These changes were all abrogated by the antioxidant N-acetylcysteine. CONCLUSIONS This study provides the first evidence for a synergistic ATO/CDDP anticancer (apoptotic) activity in OSCC cells with a favorable DRI, thereby highlighting its potential as a combinational therapeutic regime in OSCC.
Collapse
Affiliation(s)
- Toshiki Nakaoka
- Department of Oral and Maxillofacial Surgery, Aichi Medical University Hospital, Nagakute, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
|
7
|
Arsenic trioxide inhibits the growth of human glioma stem cells through activating the JNK pathway. Mol Cell Toxicol 2012. [DOI: 10.1007/s13273-012-0023-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
8
|
Ai Z, Pan H, Suo T, Lv C, Wang Y, Tong S, Liu H. Arsenic oxide targets stem cell marker CD133/prominin-1 in gallbladder carcinoma. Cancer Lett 2011; 310:181-7. [PMID: 21788103 DOI: 10.1016/j.canlet.2011.06.035] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2011] [Accepted: 06/24/2011] [Indexed: 11/29/2022]
Abstract
CD133+ tumor cells are responsible for the initiation, propagation and recurrence of tumors, which raises the question of how to effectively target CD133+ tumor cells. Arsenic trioxide (As2O3) has considerable efficacy in treating solid tumors with induction of apoptosis. Here, we found that purified CD133+ gallbladder carcinoma cells are highly resistant to conventional chemotherapy. However, As2O3 effectively induces CD133+ gallbladder carcinoma cells apoptosis. Treatment with As2O3 reduces CD133 expression at transcriptional levels. Furthermore, the ectopic expression of CD133 attenuated the apoptotic effect of As2O3 on cells through activation of AKT signaling pathways. Collectively, As2O3 effectively targets CD133 in gallbladder carcinoma, providing a new mechanism of As2O3-induced cell apoptosis and a better understanding of drug resistance in gallbladder carcinoma.
Collapse
Affiliation(s)
- Zhilong Ai
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
9
|
Sun H, Zhang S. Arsenic trioxide regulates the apoptosis of glioma cell and glioma stem cell via down-regulation of stem cell marker Sox2. Biochem Biophys Res Commun 2011; 410:692-7. [PMID: 21703238 DOI: 10.1016/j.bbrc.2011.06.060] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2011] [Accepted: 06/08/2011] [Indexed: 12/17/2022]
Abstract
Knowledge of the mechanism by which arsenic trioxide exerts the anti-tumor effects may help in designing a more effective regimen for therapy. Transcription factor Sox2, a key gene implicated in maintaining the "stemness" of embryonic and adult stem cells, plays an important role in the carcinogenesis and maintenance of glioblastoma. Here, we found that the expression of Sox2 at transcriptional level was decreased during As(2)O(3)-induced glioma cell apoptosis. And, the ectopic expression of Sox2 attenuated the apoptotic effect of As(2)O(3) on glioma cell. Furthermore, As(2)O(3) inhibited the self-renewal of glioma stem cells, and efficiently induces the apoptosis of glioma stem cells, at least, partly through down-regulation of Sox2. These data identify a previously unrecognized mechanism of the anti-tumor effects of arsenic trioxide.
Collapse
Affiliation(s)
- Hui Sun
- Department of Neurosurgery, The First Affiliated Hospital, Bengbu Medical College, Bengbu, Anhui 233004, People's Republic of China.
| | | |
Collapse
|
10
|
Kim KJ, Jung JM, Cho JY, Woo SY, Cho KA, Ryu KH, Yoo ES. Antitumor effects of imatinib mesylate and synergistic cytotoxicity with an arsenic compound in neuroblastoma cell lines. Exp Ther Med 2011; 2:557-561. [PMID: 22977540 DOI: 10.3892/etm.2011.220] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2010] [Accepted: 02/24/2011] [Indexed: 11/06/2022] Open
Abstract
Neuroblastoma is a common tumor in childhood and exhibits heterogeneity and malignant progression. MYCN expression and amplification profiles are frequently correlated with the efficacy of therapy. Arsenic trioxide and imatinib mesylate (STI-571) have been suggested as promising therapeutic agents for neuroblastoma, which has been shown to be resistant to conventional therapy. In order to ascertain whether the combination of arsenic trioxide and STI-571 exerts a synergistic cytotoxic effect on neuroblastoma cells in relation to MYCN status, we evaluated cellular proliferation after 72 h of exposure to arsenic trioxide and STI-571 with or without siRNA against MYCN in SH-SY5Y, SK-N-SH and SK-N-BE(2) neuroblastoma cells. Arsenic trioxide and STI-571 demonstrated a synergistic inhibitory effect on cellular proliferation, while MYCN knockdown had an antagonistic effect on this combined treatment. These results indicate that STI-571 treatment may prove effective for MYCN-expressing or MYCN-amplified neuroblastoma. Furthermore, siRNA therapy targeted to MYCN should be avoided in combination with STI-571 treatment in cases of neuroblastoma.
Collapse
|
11
|
Zhang N, Wu ZM, McGowan E, Shi J, Hong ZB, Ding CW, Xia P, Di W. Arsenic trioxide and cisplatin synergism increase cytotoxicity in human ovarian cancer cells: therapeutic potential for ovarian cancer. Cancer Sci 2009; 100:2459-64. [PMID: 19769630 PMCID: PMC11159866 DOI: 10.1111/j.1349-7006.2009.01340.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Drug resistance is a major concern in the successful treatment of ovarian cancer. In the present study we report a combinational drug regime using arsenic trioxide (ATO) and cisplatin (CDDP) to increase therapeutic potentiality in ovarian cancer cells. ATO-mediated growth inhibition and apoptosis in human suspension ovarian cancer COC1 cells were evaluated by MTT assay and annexin V assay using flow cytometry, respectively. cDNA arrays were performed to screen ATO-mediated gene expression. Treatment of COC1 cells with ATO alone resulted in growth inhibition and apoptosis with a dose-and time-dependent fashion; further cDNA arrays showed that 34 genes (23 up-regulated genes and 11 down-regulated genes) may strongly associate with the antiproliferative and pro-apoptotic effects induced by ATO. Furthermore, Chou-Talalay analysis was used to evaluate the combinational effect of ATO and CDDP as well as dose-reduction index (DRI) in a panel of ovarian cancer cells including CDDP-sensitive and -resistant cell lines. The combination index (CI) analysis indicated that the interaction effect of ATO/CDDP exhibited a wide range of synergism in all the adherent ovarian cancer cells (A2780, IGROV-1, SKOV-3, and R182) as well as 0.93 to 0.69 for IC(50) to IC(90) in suspension COC1 cells where CI < 1, =1, and >1, define synergism, additive effect, and antagonism, respectively. More intriguingly, the combination of ATO and CDDP yielded favorable DRIs ranging from 1.23-fold to 13.51-fold dose reduction. These results suggest that ATO and its combination with CDDP present therapeutic potential for ovarian cancer, and deserve further preclinical and clinical studies.
Collapse
Affiliation(s)
- Ning Zhang
- Department of Obstetrics and Gynecology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Fröhlich E, Czarnocka B, Brossart P, Wahl R. Antitumor effects of arsenic trioxide in transformed human thyroid cells. Thyroid 2008; 18:1183-93. [PMID: 19014326 DOI: 10.1089/thy.2008.0114] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
BACKGROUND To improve radioiodine treatment of metastasized differentiated thyroid carcinomas, substances that increase iodide uptake are needed. Many tumors are not responsive to retinoic acid as a differentiating agent. Therefore, identification of other differentiating substances is needed. Arsenic trioxide (ATO) was investigated for its potential to increase iodide uptake. METHODS The action of ATO on proliferation, differentiation, and apoptosis was evaluated in follicular and papillary thyroid carcinoma cell lines. To get insight into the mode of action of ATO, coincubations with inhibitors of the phosphoinositide 3 (PI3) kinase pathway (V-Akt Murine Thymoma Viral Oncogene Homolog 1, Akt inhibitors) were performed; glutathione (GSH) levels were determined, as well as synergistic effects of ATO with inhibitors of GSH metabolism, inductors of oxidative stress. As a potential additional target of the pleiotropic action of ATO, its effect on glucose uptake was investigated. The expression of sodium iodide symporter, pendrin, phospho-Akt, and glucose transporter 1 was studied to reveal a potential effect of ATO on the transcription of specific genes. RESULTS ATO reduced proliferation, increased iodide uptake and apoptosis, and, as an additional new mechanism, decreased glucose uptake in transformed thyrocytes. The pharmacological reduction of the amount of reduced GSH was effective in enhancing the differentiating action of ATO, whereas the combination of ATO with Akt-1 inhibitors reduced cell number but did not increase differentiation. CONCLUSIONS Our study suggests a new therapeutic option for postoperative treatment of radioiodine nonresponsive differentiated thyroid carcinomas.
Collapse
MESH Headings
- Adenocarcinoma, Follicular/drug therapy
- Adenocarcinoma, Follicular/metabolism
- Adenocarcinoma, Follicular/pathology
- Adenocarcinoma, Follicular/radiotherapy
- Antineoplastic Agents/pharmacology
- Apoptosis/drug effects
- Arsenic Trioxide
- Arsenicals/pharmacology
- Biological Transport, Active/drug effects
- Carcinoma, Papillary/drug therapy
- Carcinoma, Papillary/metabolism
- Carcinoma, Papillary/pathology
- Carcinoma, Papillary/radiotherapy
- Cell Differentiation/drug effects
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cell Transformation, Neoplastic/drug effects
- Cell Transformation, Neoplastic/metabolism
- Glucose/metabolism
- Glutathione/metabolism
- Humans
- Iodides/metabolism
- Iodine Radioisotopes/therapeutic use
- Oncogene Protein v-akt/antagonists & inhibitors
- Oxides/pharmacology
- Phosphatidylinositol 3-Kinases/metabolism
- Radiation Tolerance
- Reactive Oxygen Species/metabolism
- Thyroid Neoplasms/drug therapy
- Thyroid Neoplasms/metabolism
- Thyroid Neoplasms/pathology
- Thyroid Neoplasms/radiotherapy
Collapse
Affiliation(s)
- Eleonore Fröhlich
- Department of Endocrinology, Metabolism, Nephrology, and Clinical Chemistry, University of Tuebingen, Tuebingen, Germany
| | | | | | | |
Collapse
|
13
|
Wei Y, Liu D, Ge Y, Zhou F, Xu J, Chen H, Yun X, Gu J, Jiang J. Down-regulation of β1,4GalT V at protein level contributes to arsenic trioxide-induced glioma cell apoptosis. Cancer Lett 2008; 267:96-105. [DOI: 10.1016/j.canlet.2008.03.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2007] [Revised: 03/01/2008] [Accepted: 03/10/2008] [Indexed: 12/21/2022]
|
14
|
Florea AM, Büsselberg D. Arsenic trioxide in environmentally and clinically relevant concentrations interacts with calcium homeostasis and induces cell type specific cell death in tumor and non-tumor cells. Toxicol Lett 2008; 179:34-42. [PMID: 18485628 DOI: 10.1016/j.toxlet.2008.03.019] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2008] [Revised: 03/22/2008] [Accepted: 03/30/2008] [Indexed: 12/24/2022]
Abstract
While arsenic compounds are known as environmental toxicants (especially in drinking water) and as carcinogens, some arsenic compounds, like arsenic trioxide (As2O3), are clinically used in humans to treat some forms of cancer (e.g. leukemia). Although arsenic compounds have been studied intensively, their interactions with living cells are still not fully elucidated. We have previously proposed that modulation of intracellular calcium ([Ca2+]i) homeostasis induced by As2O3 could be an important mechanism to induce cytotoxicity. Here we demonstrate, using human cell models (neuroblastoma (SY-5Y) or embryonic kidney cells (HEK)) and confocal microscopy in combination with the calcium sensitive dye fluo 4-AM, that As2O3 interferes with calcium signaling at low (environmentally and clinically relevant concentrations of 100 pM to 1 microM). Within this concentration range, As2O3 had cell type specific cytotoxic effects, with neuroblastoma cells being more sensitive to As2O3 than HEK 293. In addition, by staining with Hoechst 33347 and counting micronucleated cells as well as apoptotic nuclei, As2O3 was found to increase the rate of apoptosis and DNA damage, which was also cell type specific. These results indicate that the As2O3-induced cell death could be triggered or mediated by [Ca2+]i signals and suggest that low concentrations of As2O3 are able to interfere with specific physiological processes in diverse cell models.
Collapse
Affiliation(s)
- Ana-Maria Florea
- Institut für Physiologie, Universitätsklinikum, Universität Duisburg Essen, Essen, Germany.
| | | |
Collapse
|
15
|
Han YH, Kim SZ, Kim SH, Park WH. Intracellular GSH level is a factor in As4.1 juxtaglomerular cell death by arsenic trioxide. J Cell Biochem 2008; 104:995-1009. [DOI: 10.1002/jcb.21685] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
16
|
Peng Z, Peng L, Fan Y, Zandi E, Shertzer HG, Xia Y. A critical role for IkappaB kinase beta in metallothionein-1 expression and protection against arsenic toxicity. J Biol Chem 2007; 282:21487-96. [PMID: 17526490 DOI: 10.1074/jbc.m702510200] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Arsenic is a widespread environmental toxic agent that has been shown to cause diverse tissue and cell damage and at the same time to be an effective anti-cancer therapeutic agent. The objective of this study is to explore the signaling mechanisms involved in arsenic toxicity. We show that the IkappaB kinase beta (IKKbeta) plays a crucial role in protecting cells from arsenic toxicity. Ikkbeta(-)(/)(-) mouse 3T3 fibroblasts have decreased expression of antioxidant genes, such as metallothionein 1 (Mt1). In contrast to wild type and IKKbeta-reconstituted Ikkbeta(-)(/)(-) cells, IKKbeta-null cells display a marked increase in arsenic-induced reactive oxygen species (ROS) accumulation, which leads to activation of the MKK4-c-Jun NH(2)-terminal kinase (JNK) pathway, c-Jun phosphorylation, and apoptosis. Pretreatment with the antioxidant N-acetylcysteine (NAC) and expression of MT1 in the Ikkbeta(-)(/)(-) cells prevented JNK activation; moreover, NAC pretreatment, MT1 expression, MKK4 ablation, and JNK inhibition all protected cells from death induced by arsenic. Our data show that two signaling pathways appear to be important for modulating arsenic toxicity. First, the IKK-NF-kappaB pathway is crucial for maintaining cellular metallothionein-1 levels to counteract ROS accumulation, and second, when this pathway fails, excessive ROS leads to activation of the MKK4-JNK pathway, resulting in apoptosis.
Collapse
Affiliation(s)
- Zhimin Peng
- Department of Environmental Health, University of Cincinnati Medical Center, Cincinnati, Ohio 45267-0056, USA
| | | | | | | | | | | |
Collapse
|
17
|
Swiderek H, Al-Rubeai M. Functional genome-wide analysis of antibody producing NS0 cell line cultivated at different temperatures. Biotechnol Bioeng 2007; 98:616-30. [PMID: 17421047 DOI: 10.1002/bit.21445] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Lowering culture temperature has been reported as a significant factor in the improvement of mammalian cell productivity. To determine the physiological changes which take place at different temperature cultivations, an NS0 cell line producing human-mouse chimeric antibody was cultured at 22, 34 and 37 degrees C. Various cellular parameters such as viability, productivity, metabolism, apoptosis and cell cycle were studied and notable changes were shown to be accompanied by changes in metabolic rates. Reduction of the temperature to 22 degrees C resulted in a decrease in the growth rate, inhibition of antibody production, arrest of cell cycle in G2 phase and delay in apoptosis. A slight increase in antibody production was observed at 34 degrees C due to the increase of growth rate and prolonged stationary phase. To better understand and explore the mechanisms underpinning these biological alterations and to identify the genes involved in the genetic reprogramming, genome-wide analyses were performed using GeneChip Mouse Genome arrays. The examination of differential gene expression induced by temperature reduction demonstrated a specific pattern of gene expression in NS0 cells in response to temperature stress. The effect of temperature on transcription induced changes within a wide range of genes involved in metabolic and signalling pathways. Most deregulated genes involved in essential metabolic pathways (i.e. glycolysis/gluconeogenesis, pentose phosphate pathway and inositol metabolism) were repressed in cells cultured at 22 degrees C. By combining gene expression and physiological changes at different temperatures it was possible to provide greater understanding of cell response to hypothermic conditions.
Collapse
Affiliation(s)
- Halina Swiderek
- UCD School of Chemical and Bioprocess Engineering, University College Dublin, Belfield, Dublin 4, Ireland
| | | |
Collapse
|
18
|
Olivier S, Robe P, Bours V. Can NF-κB be a target for novel and efficient anti-cancer agents? Biochem Pharmacol 2006; 72:1054-68. [PMID: 16973133 DOI: 10.1016/j.bcp.2006.07.023] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2006] [Revised: 07/20/2006] [Accepted: 07/24/2006] [Indexed: 12/23/2022]
Abstract
Since the discovery of the NF-kappaB transcription factor in 1986 and the cloning of the genes coding for NF-kappaB and IkappaB proteins, many studies demonstrated that this transcription factor can, in most cases, protect transformed cells from apoptosis and therefore participate in the onset or progression of many human cancers. Molecular studies demonstrated that ancient widely used drugs, known for their chemopreventive or therapeutic activities against human cancers, inhibit NF-kappaB, usually among other biological effects. It is therefore considered that the anti-cancer activities of NSAIDs (non-steroidal anti-inflammatory drugs) or glucocorticoids are probably partially related to the inhibition of NF-kappaB and new clinical trials are being initiated with old compounds such as sulfasalazine. In parallel, many companies have developed novel agents acting on the NF-kappaB pathway: some of these agents are supposed to be NF-kappaB specific (i.e. IKK inhibitors) while others have wide-range biological activities (i.e. proteasome inhibitors). Today, the most significant clinical data have been obtained with bortezomib, a proteasome inhibitor, for the treatment of multiple myeloma. This review discusses the preclinical and clinical data obtained with these various drugs and their putative future developments.
Collapse
Affiliation(s)
- Sabine Olivier
- Department of Rheumatology, Centre for Biomedical Integrative Genoproteomics, University of Liège, CHU B35, Sart-Tilman, 4000 Liège, Belgium
| | | | | |
Collapse
|