1
|
Natural gum as bio-reductant to green synthesize silver nanoparticles: assessing the apoptotic efficacy on MCF-7 and SH-SY5Y cell lines and their antimicrobial potential. Polym Bull (Berl) 2021. [DOI: 10.1007/s00289-020-03238-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
2
|
Wahiduzzaman M, Ota A, Hosokawa Y. Novel Mechanistic Insights into the Anti-cancer Mode of Arsenic Trioxide. Curr Cancer Drug Targets 2021; 20:115-129. [PMID: 31736446 DOI: 10.2174/1568009619666191021122006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 08/23/2019] [Accepted: 09/19/2019] [Indexed: 12/19/2022]
Abstract
Arsenic, a naturally-occurring toxic element, and a traditionally-used drug, has received a great deal of attention worldwide due to its curative anti-cancer properties in patients with acute promyelocytic leukemia. Among the arsenicals, arsenic trioxide has been most widely used as an anti-cancer drug. Recent advances in cancer therapeutics have led to a paradigm shift away from traditional cytotoxic drugs towards the targeting of proteins closely associated with driving the cancer phenotype. Due to the diverse anti-cancer effects of ATO on different types of malignancies, numerous studies have made efforts to uncover the mechanisms of ATO-induced tumor suppression. From in vitro cellular models to studies in clinical settings, ATO has been extensively studied. The outcomes of these studies have opened doors to establishing improved molecular-targeted therapies for cancer treatment. The efficacy of ATO has been augmented by combination with other drugs. In this review, we discuss recent arsenic-based cancer therapies and summarize the novel underlying molecular mechanisms of the anti-cancer effects of ATO.
Collapse
Affiliation(s)
- Md Wahiduzzaman
- Department of Biochemistry, School of Medicine, Aichi Medical University, Nagakute, Aichi 480-1195, Japan
| | - Akinobu Ota
- Department of Biochemistry, School of Medicine, Aichi Medical University, Nagakute, Aichi 480-1195, Japan
| | - Yoshitaka Hosokawa
- Department of Biochemistry, School of Medicine, Aichi Medical University, Nagakute, Aichi 480-1195, Japan
| |
Collapse
|
3
|
Reactive oxygen species (ROS): Critical roles in breast tumor microenvironment. Crit Rev Oncol Hematol 2021; 160:103285. [DOI: 10.1016/j.critrevonc.2021.103285] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 01/18/2021] [Accepted: 02/27/2021] [Indexed: 02/06/2023] Open
|
4
|
Karmous I, Pandey A, Haj KB, Chaoui A. Efficiency of the Green Synthesized Nanoparticles as New Tools in Cancer Therapy: Insights on Plant-Based Bioengineered Nanoparticles, Biophysical Properties, and Anticancer Roles. Biol Trace Elem Res 2020; 196:330-342. [PMID: 31512171 DOI: 10.1007/s12011-019-01895-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Accepted: 09/05/2019] [Indexed: 12/14/2022]
Abstract
The aim of this work is to review the current knowledge on the efficiency of plant-based synthesized nanoparticles in medical field, particularly in the prevention, diagnosis, and therapy of cancer. For this, we examine the advantages of nanotechnological tools. Besides, a particular attention was given to understand the mechanism by which plant-based bioengineered nanoparticles can interact with components of cancerous cells. Green biosynthesized nanoparticles seem to be novel tool for prognostic biomarkers for cancer diagnosis and drug delivery in tumor cells. They can act either by leading to the damage of tumor cells, or by the protection of healthy cells, via mechanisms involving the specific properties of nanoparticles themselves and the antioxidative and antitumor properties found in plants. However, special attention should be given to the choice of plant species, extracts, and the toxic dose of some phytocompounds during the biosynthesis process. An increase in metal or trace element release from metal and metal oxide biosynthesized nanoparticles can lead to greater oxidative stress, which is associated with higher risk of cancer. Hence, plant-based nanosystems should be more developed to increase their specific targeting of the cancerous cells, in order to preserve the healthy ones.
Collapse
Affiliation(s)
- Inès Karmous
- Applied Institute of Biology of Medenine, University of Gabes, Gabes, Tunisia.
- Plant Toxicology and Molecular Biology of Microorganism, Faculty of Sciences of Bizerta, 7021, Zarzouna, Tunisia.
| | - Ashish Pandey
- SUNUM, Sabanci University, Orhanli, Tuzla, 34956, Istanbul, Turkey
| | - Khemais Ben Haj
- Applied Institute of Biology of Medenine, University of Gabes, Gabes, Tunisia
| | - Abdelilah Chaoui
- Plant Toxicology and Molecular Biology of Microorganism, Faculty of Sciences of Bizerta, 7021, Zarzouna, Tunisia
| |
Collapse
|
5
|
A candidate for lung cancer treatment: arsenic trioxide. Clin Transl Oncol 2019; 21:1115-1126. [PMID: 30756240 DOI: 10.1007/s12094-019-02054-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 01/29/2019] [Indexed: 12/14/2022]
Abstract
Arsenic trioxide (ATO), a highly effective drug in treating acute promyelocytic leukemia with low toxicity, demonstrates a significant effect on lung cancer. The anti-cancer mechanisms of ATO include inhibition of cancer stem-like cells, induction of apoptosis, anti-angiogenesis, sensitization of chemotherapy and radiotherapy, anti-cancer effects of hypoxia, and immunoregulation properties. In addition, some studies have reported that different lung cancers respond differently to ATO. It was concluded on numerous studies that the rational combination of administration and encapsulation of ATO have promising potentials in increasing drug efficacy and decreasing adverse drug effects. We reviewed the efficacy of ATO in the treatment of lung cancer in recent years to provide some views for further study.
Collapse
|
6
|
Hassanzade A, Mandegary A, Sharif E, Rasooli R, Mohammadnejad R, Masoumi-Ardekani Y. Cyclooxygenase inhibitors combined with deuterium-enriched water augment cytotoxicity in A549 lung cancer cell line via activation of apoptosis and MAPK pathways. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2018; 21:508-516. [PMID: 29922432 PMCID: PMC6000214 DOI: 10.22038/ijbms.2018.25366.6269] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Objectives Combination chemotherapy is a rational strategy to increase patient response and tolerability and to decrease adverse effects and drug resistance. Recently, the use of non-steroidal anti-inflammatory drugs (NSAIDs) has been reported to be associated with reduction in occurrence of a variety of cancers including lung cancer. On the other hand, growing evidences suggest that deuterium-enriched water (DEW, D2O) and deuterium-depleted water (DDW) play a role both in treatment and prevention of cancers. In the present study, we examined the effects of DEW and DDW in combination with two NSAIDs, celecoxib and indomethacin, on A549 human non-small lung cancer cell to identify novel treatment options. Materials and Methods The cytotoxicity of celecoxib or indomethacin, alone and in combination with DDW and DEW was determined. The COX-2, MAPK pathway proteins, the anti-apoptotic Bcl2 and pro-apoptotic Bax proteins and caspase-3 activity were studied for cytotoxic combinations. Results Co-administration of selective and non-selective COX-2 inhibitors with DEW led to a remarkable increase in cytotoxicity and apoptosis of A549 cells. These events were associated with activation of p38 and JNK MAPKs and decreasing pro-survival proteins Bcl-2, COX-2 and ERK1/2. Furthermore, the combination therapy activated caspase-3, and the apoptosis mediator, and disabled poly ADP-ribose polymerase (PARP), the key DNA repair enzyme, by cleaving it. Conclusion The combination of DEW with NSAIDs might be effective against lung cancer cells by influence on principal cell signalling pathways, and this has a potential to become a candidate for chemotherapy.
Collapse
Affiliation(s)
- Abdolreza Hassanzade
- Pharmaceutics Research Centre, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.,Department of Medicinal Chemistry, School of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Ali Mandegary
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.,Department of Pharmacology & Toxicology, School of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Elham Sharif
- Pharmaceutics Research Centre, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.,Department of Pharmacology & Toxicology, School of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Rokhsana Rasooli
- Pharmaceutics Research Centre, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.,Department of Pharmacology & Toxicology, School of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Reza Mohammadnejad
- Pharmaceutics Research Centre, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.,Department of Pharmacology & Toxicology, School of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Yaser Masoumi-Ardekani
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
7
|
Bendale Y, Bendale V, Natu R, Paul S. Biosynthesized Platinum Nanoparticles Inhibit the Proliferation of Human Lung-Cancer Cells in vitro and Delay the Growth of a Human Lung-Tumor Xenograft in vivo: -In vitro and in vivo Anticancer Activity of bio-Pt NPs. J Pharmacopuncture 2016; 19:114-21. [PMID: 27386144 PMCID: PMC4931296 DOI: 10.3831/kpi.2016.19.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Objectives: Lung cancer remains a deadly disease with unsatisfactory overall survival. Cisplatin, a standard platinum (Pt)-based chemotherapeutic agent, has the potential to inhibit the growth of lung cancer. Its use, however, is occasionally limited by severe organ toxicity. However, until now, no systematic study has been conducted to verify its efficacy with proper experimental support in vivo. Therefore, we examined whether biosynthesized Pt nanoparticles (NPs) inhibited human lung cancer in vitro and in vivo to validate their use in alternative and complementary medicine. Methods: We evaluated the in vitro and the in vivo anticancer efficiencies of biosynthesized Pt NPs in a subcutaneous xenograft model with A549 cells. Severe combined immune deficient mice (SCID) were divided into four groups: group 1 being the vehicle control group and groups 2, 3 and 4 being the experimental groups. Once the tumor volume had reached 70 ─ 75 mm3, the progression profile of the tumor growth kinetics and the body weights of the mice were measured every week for 6 weeks after oral administration of Pt NPs. Doses of Pt NPs of 500, 1,000 and 2,000 mg/kg of body weight were administered to the experimental groups and a dose of honey was administered to the vehicle control group. The efficacy was quantified by using the delay in tumor growth following the administration of Pt NPs of A549 human-lung-cancer xenografts growing in SCID mice. Results: The in vitro cytotoxicity evaluation indicated that Pt NPs, in a dose-dependent manner, inhibited the growth of A549 cells, and the in vivo evaluation showed that Pt NPs at the mid and high doses effectively inhibited and delayed the growth of lung cancer in SCID mice. Conclusion: These findings confirm the antitumor properties of biosynthesized Pt NPs and suggest that they may be a cost-effective alternative for the treatment of patients with lung cancer.
Collapse
Affiliation(s)
- Yogesh Bendale
- Research and Development Section, Rasayani Biologics Private Limited, Pune, India
| | - Vineeta Bendale
- Research and Development Section, Rasayani Biologics Private Limited, Pune, India
| | - Rammesh Natu
- Research and Development Section, Rasayani Biologics Private Limited, Pune, India
| | - Saili Paul
- Research and Development Section, Rasayani Biologics Private Limited, Pune, India
| |
Collapse
|
8
|
He Y, Du Z, Ma S, Liu Y, Li D, Huang H, Jiang S, Cheng S, Wu W, Zhang K, Zheng X. Effects of green-synthesized silver nanoparticles on lung cancer cells in vitro and grown as xenograft tumors in vivo. Int J Nanomedicine 2016; 11:1879-87. [PMID: 27217750 PMCID: PMC4862350 DOI: 10.2147/ijn.s103695] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Silver nanoparticles (AgNPs) have now been recognized as promising therapeutic molecules and are extending their use in cancer diagnosis and therapy. This study demonstrates for the first time the antitumor activity of green-synthesized AgNPs against lung cancer in vitro and in vivo. Cytotoxicity effect was explored on human lung cancer H1299 cells in vitro by MTT and trypan blue assays. Apoptosis was measured by morphological assessment, and nuclear factor-κB (NF-κB) transcriptional activity was determined by a luciferase reporter gene assay. The expressions of phosphorylated stat3, bcl-2, survivin, and caspase-3 were examined by Western blot analysis. AgNPs showed dose-dependent cytotoxicity and stimulation of apoptosis in H1299 cells. The effects on H1299 cells correlated well with the inhibition of NF-κB activity, a decrease in bcl-2, and an increase in caspase-3 and survivin expression. AgNPs significantly suppressed the H1299 tumor growth in a xenograft severe combined immunodeficient (SCID) mouse model. The results demonstrate the anticancer activities of AgNPs, suggesting that they may act as potential beneficial molecules in lung cancer chemoprevention and chemotherapy, especially for early-stage intervention.
Collapse
Affiliation(s)
- Yan He
- Allan H Conney Laboratory for Anticancer Drug Research, School of Chemical Engineering and Light Industry, Guandong University of Technology, Guangzhou, People's Republic of China
| | - Zhiyun Du
- Allan H Conney Laboratory for Anticancer Drug Research, School of Chemical Engineering and Light Industry, Guandong University of Technology, Guangzhou, People's Republic of China
| | - Shijing Ma
- Allan H Conney Laboratory for Anticancer Drug Research, School of Chemical Engineering and Light Industry, Guandong University of Technology, Guangzhou, People's Republic of China
| | - Yue Liu
- Susan Lehman Cullman Laboratory for Cancer Research, Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Dongli Li
- Allan H Conney Laboratory for Anticancer Drug Research, School of Chemical Engineering and Light Industry, Guandong University of Technology, Guangzhou, People's Republic of China
| | - Huarong Huang
- Allan H Conney Laboratory for Anticancer Drug Research, School of Chemical Engineering and Light Industry, Guandong University of Technology, Guangzhou, People's Republic of China
| | - Sen Jiang
- Allan H Conney Laboratory for Anticancer Drug Research, School of Chemical Engineering and Light Industry, Guandong University of Technology, Guangzhou, People's Republic of China
| | - Shupeng Cheng
- Allan H Conney Laboratory for Anticancer Drug Research, School of Chemical Engineering and Light Industry, Guandong University of Technology, Guangzhou, People's Republic of China
| | - Wenjing Wu
- Allan H Conney Laboratory for Anticancer Drug Research, School of Chemical Engineering and Light Industry, Guandong University of Technology, Guangzhou, People's Republic of China
| | - Kun Zhang
- Allan H Conney Laboratory for Anticancer Drug Research, School of Chemical Engineering and Light Industry, Guandong University of Technology, Guangzhou, People's Republic of China
| | - Xi Zheng
- Allan H Conney Laboratory for Anticancer Drug Research, School of Chemical Engineering and Light Industry, Guandong University of Technology, Guangzhou, People's Republic of China; Susan Lehman Cullman Laboratory for Cancer Research, Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| |
Collapse
|
9
|
Li H, Tang Z, Zhu H, Ge H, Cui S, Jiang W. Proteomic study of benign and malignant pleural effusion. J Cancer Res Clin Oncol 2016; 142:1191-200. [PMID: 26945985 DOI: 10.1007/s00432-016-2130-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2015] [Accepted: 02/08/2016] [Indexed: 12/18/2022]
Abstract
BACKGROUND Lung adenocarcinoma can easily cause malignant pleural effusion which was difficult to discriminate from benign pleural effusion. Now there was no biomarker with high sensitivity and specificity for the malignant pleural effusion. PURPOSE This study used proteomics technology to acquire and analyze the protein profiles of the benign and malignant pleural effusion, to seek useful protein biomarkers with diagnostic value and to establish the diagnostic model. METHODS We chose the weak cationic-exchanger magnetic bead (WCX-MB) to purify peptides in the pleural effusion, used matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF-MS) to obtain peptide expression profiles from the benign and malignant pleural effusion samples, established and validated the diagnostic model through a genetic algorithm (GA) and finally identified the most promising protein biomarker. RESULTS A GA diagnostic model was established with spectra of 3930.9 and 2942.8 m/z in the training set including 25 malignant pleural effusion and 26 benign pleural effusion samples, yielding both 100 % sensitivity and 100 % specificity. The accuracy of diagnostic prediction was validated in the independent testing set with 58 malignant pleural effusion and 34 benign pleural effusion samples. Blind evaluation was as follows: the sensitivity was 89.6 %, specificity 88.2 %, PPV 92.8 %, NPV 83.3 % and accuracy 89.1 % in the independent testing set. The most promising peptide biomarker was identified successfully: Isoform 1 of caspase recruitment domain-containing protein 9 (CARD9), with 3930.9 m/z, was decreased in the malignant pleural effusion. CONCLUSIONS This model is suitable to discriminate benign and malignant pleural effusion and CARD9 can be used as a new peptide biomarker.
Collapse
Affiliation(s)
- Hongqing Li
- Department of Respiratory Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, 200040, China
| | - Zhonghao Tang
- Department of Respiratory Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, 200040, China
| | - Huili Zhu
- Department of Respiratory Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, 200040, China.
| | - Haiyan Ge
- Department of Respiratory Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, 200040, China
| | - Shilei Cui
- Department of Respiratory Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, 200040, China
| | - Weiping Jiang
- Department of Respiratory Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai, 200040, China
| |
Collapse
|
10
|
Yun SM, Woo SH, Oh ST, Hong SE, Choe TB, Ye SK, Kim EK, Seong MK, Kim HA, Noh WC, Lee JK, Jin HO, Lee YH, Park IC. Melatonin enhances arsenic trioxide-induced cell death via sustained upregulation of Redd1 expression in breast cancer cells. Mol Cell Endocrinol 2016; 422:64-73. [PMID: 26607805 DOI: 10.1016/j.mce.2015.11.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 10/24/2015] [Accepted: 11/12/2015] [Indexed: 10/22/2022]
Abstract
Melatonin is implicated in various physiological functions, including anticancer activity. However, the mechanism(s) of its anticancer activity is not well understood. In the present study, we investigated the combined effects of melatonin and arsenic trioxide (ATO) on cell death in human breast cancer cells. Melatonin enhanced the ATO-induced apoptotic cell death via changes in the protein levels of Survivin, Bcl-2, and Bax, thus affecting cytochrome c release from the mitochondria to the cytosol. Interestingly, we found that the cell death induced by co-treatment with melatonin and ATO was mediated by sustained upregulation of Redd1, which was associated with increased production of reactive oxygen species (ROS). Combined treatment with melatonin and ATO induced the phosphorylation of JNK and p38 MAP kinase downstream from Redd1 expression. Rapamycin and S6K1 siRNA enhanced, while activation of mTORC1 by transfection with TSC2 siRNA suppressed the cell death induced by melatonin and ATO treatment. Taken together, our findings suggest that melatonin enhances ATO-induced apoptotic cell death via sustained upregulation of Redd1 expression and inhibition of mTORC1 upstream of the activation of the p38/JNK pathways in human breast cancer cells.
Collapse
Affiliation(s)
- Sun-Mi Yun
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 139-706, Republic of Korea
| | - Sang Hyeok Woo
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 139-706, Republic of Korea; KIRAMS Radiation Biobank, Korea Institute of Radiological & Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 139-709, Republic of Korea
| | - Sang Taek Oh
- Department of Radiation Oncology, College of Medicine, Yonsei University, Seoul 120-752, Republic of Korea
| | - Sung-Eun Hong
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 139-706, Republic of Korea
| | - Tae-Boo Choe
- Department of Microbiological Engineering, Kon-Kuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 143-701, Republic of Korea
| | - Sang-Kyu Ye
- Department of Pharmacology, Seoul National University College of Medicine, 103 Daehangno, Jongno-gu Seoul 110-799, Republic of Korea
| | - Eun-Kyu Kim
- Department of Surgery, Seoul National University Bundang Hospital, 82 Gumi-ro, 173 Beon-gil, Bundang-gu, Seongnam-si, Gyenggi-do 463-707, Republic of Korea
| | - Min Ki Seong
- Department of Surgery, Korea Cancer Center Hospital, Korea Institute of Radiological & Medical Sciences, 215-4 Gongneung-dong, Nowon-gu, Seoul 139-706, Republic of Korea
| | - Hyun-A Kim
- Department of Surgery, Korea Cancer Center Hospital, Korea Institute of Radiological & Medical Sciences, 215-4 Gongneung-dong, Nowon-gu, Seoul 139-706, Republic of Korea
| | - Woo Chul Noh
- Department of Surgery, Korea Cancer Center Hospital, Korea Institute of Radiological & Medical Sciences, 215-4 Gongneung-dong, Nowon-gu, Seoul 139-706, Republic of Korea
| | - Jin Kyung Lee
- KIRAMS Radiation Biobank, Korea Institute of Radiological & Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 139-709, Republic of Korea
| | - Hyeon-Ok Jin
- KIRAMS Radiation Biobank, Korea Institute of Radiological & Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 139-709, Republic of Korea
| | - Yun-Han Lee
- Department of Radiation Oncology, College of Medicine, Yonsei University, Seoul 120-752, Republic of Korea.
| | - In-Chul Park
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 139-706, Republic of Korea.
| |
Collapse
|
11
|
Yang Y, Yang Y, Hou J, Ding Y, Zhang T, Zhang Y, Wang J, Shi C, Fu W, Cai Z. The Hydroxyl at Position C1 of Genipin Is the Active Inhibitory Group that Affects Mitochondrial Uncoupling Protein 2 in Panc-1 Cells. PLoS One 2016; 11:e0147026. [PMID: 26771380 PMCID: PMC4714807 DOI: 10.1371/journal.pone.0147026] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 12/27/2015] [Indexed: 12/14/2022] Open
Abstract
Genipin (GNP) effectively inhibits uncoupling protein 2 (UCP2), which regulates the leakage of protons across the inner mitochondrial membrane. UCP2 inhibition may induce pancreatic adenocarcinoma cell death by increasing reactive oxygen species (ROS) levels. In this study, the hydroxyls at positions C10 (10-OH) and C1 (1-OH) of GNP were hypothesized to be the active groups that cause these inhibitory effects. Four GNP derivatives in which the hydroxyl at position C10 or C1 was replaced with other chemical groups were synthesized and isolated. Differences in the inhibitory effects of GNP and its four derivatives on pancreatic carcinoma cell (Panc-1) proliferation were assessed. The effects of GNP and its derivatives on apoptosis, UCP2 inhibition and ROS production were also studied to explore the relationship between GNP’s activity and its structure. The derivatives with 1-OH substitutions, geniposide (1-GNP1) and 1-ethyl-genipin (1-GNP2) lacked cytotoxic effects, while the other derivatives that retained 1-OH, 10-piv-genipin (10-GNP1) and 10-acetic acid-genipin (10-GNP2) exerted biological effects similar to those of GNP, even in the absence of 10-OH. Thus, 1-OH is the key functional group in the structure of GNP that is responsible for GNP’s apoptotic effects. These cytotoxic effects involve the induction of Panc-1 cell apoptosis through UCP2 inhibition and subsequent ROS production.
Collapse
Affiliation(s)
- Yang Yang
- Laboratory of Immunology and Virology, Experiment Center For Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yifu Yang
- Laboratory of Immunology and Virology, Experiment Center For Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jianwei Hou
- Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yue Ding
- Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- * E-mail: (TZ); (YD)
| | - Tong Zhang
- Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- * E-mail: (TZ); (YD)
| | - Yong Zhang
- Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jianying Wang
- Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chenchen Shi
- Laboratory of Immunology and Virology, Experiment Center For Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenwei Fu
- Innovation of Traditional Chinese Medicine Laboratory, College of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhenzhen Cai
- Laboratory of Immunology and Virology, Experiment Center For Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
12
|
Jiang W, Mitra R, Lin CC, Wang Q, Cheng F, Zhao Z. Systematic dissection of dysregulated transcription factor-miRNA feed-forward loops across tumor types. Brief Bioinform 2015; 17:996-1008. [PMID: 26655252 PMCID: PMC5142013 DOI: 10.1093/bib/bbv107] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 10/23/2015] [Indexed: 02/07/2023] Open
Abstract
Transcription factor and microRNA (miRNA) can mutually regulate each other and jointly regulate their shared target genes to form feed-forward loops (FFLs). While there are many studies of dysregulated FFLs in a specific cancer, a systematic investigation of dysregulated FFLs across multiple tumor types (pan-cancer FFLs) has not been performed yet. In this study, using The Cancer Genome Atlas data, we identified 26 pan-cancer FFLs, which were dysregulated in at least five tumor types. These pan-cancer FFLs could communicate with each other and form functionally consistent subnetworks, such as epithelial to mesenchymal transition-related subnetwork. Many proteins and miRNAs in each subnetwork belong to the same protein and miRNA family, respectively. Importantly, cancer-associated genes and drug targets were enriched in these pan-cancer FFLs, in which the genes and miRNAs also tended to be hubs and bottlenecks. Finally, we identified potential anticancer indications for existing drugs with novel mechanism of action. Collectively, this study highlights the potential of pan-cancer FFLs as a novel paradigm in elucidating pathogenesis of cancer and developing anticancer drugs.
Collapse
Affiliation(s)
- Wei Jiang
- *These authors contributed equally to this work
| | | | | | | | | | | |
Collapse
|
13
|
Lam SK, Li YY, Zheng CY, Leung LL, Ho JCM. E2F1 downregulation by arsenic trioxide in lung adenocarcinoma. Int J Oncol 2014; 45:2033-43. [PMID: 25174355 DOI: 10.3892/ijo.2014.2609] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 07/01/2014] [Indexed: 11/05/2022] Open
Abstract
Lung cancer is one of the most common cancers worldwide. Arsenic trioxide (ATO) has been approved by the US Food and Drug Administration for the treatment of acute promyelocytic leukemia. Nonetheless preliminary data have suggested potential activity of ATO in solid tumors including lung cancer. This study aimed to examine the underlying mechanisms of ATO in the treatment of lung adenocarcinoma. Using a panel of 7 lung adenocarcinoma cell lines, the effects of ATO treatment on cell viability, expression of E2F1 and its downstream targets, phosphatidylserine externalization, mitochondrial membrane depolarization and alteration of apoptotic/anti-apoptotic factors were studied. Tumor growth inhibition in vivo was investigated using a nude mouse xenograft model. ATO decreased cell viability with clinically achievable concentrations (8 µM) in all cell lines investigated. This was accompanied by reduced expression of E2F1, cyclin A2, skp2, c-myc, thymidine kinase and ribonucleotide reductase M1, while p-c-Jun was upregulated. Cell viability was significantly decreased with E2F1 knockdown. Treatment with ATO resulted in phosphatidylserine externalization in H23 cells and mitochondrial membrane depolarization in all cell lines, associated with truncation of Bid, downregulation of Bcl-2, upregulation of Bax and Bak, caspase-9 and -3 activation and PARP cleavage. Using the H358 xenograft model, the tumor growth was suppressed in the ATO treatment group during 8 days of treatment, associated with downregulation of E2F1 and upregulation of truncated Bid and cleaved caspase-3. In conclusion, ATO has potent in vitro and in vivo activity in lung adenocarcinoma, partially mediated through E2F1 downregulation and apoptosis.
Collapse
Affiliation(s)
- Sze-Kwan Lam
- Division of Respiratory Medicine, Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, SAR, P.R. China
| | - Yuan-Yuan Li
- Division of Respiratory Medicine, Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, SAR, P.R. China
| | - Chun-Yan Zheng
- Division of Respiratory Medicine, Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, SAR, P.R. China
| | - Leanne Lee Leung
- Division of Respiratory Medicine, Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, SAR, P.R. China
| | - James Chung-Man Ho
- Division of Respiratory Medicine, Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, SAR, P.R. China
| |
Collapse
|
14
|
Indomethacin-enhanced anticancer effect of arsenic trioxide in A549 cell line: involvement of apoptosis and phospho-ERK and p38 MAPK pathways. BIOMED RESEARCH INTERNATIONAL 2013; 2013:237543. [PMID: 24312908 PMCID: PMC3842073 DOI: 10.1155/2013/237543] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Accepted: 09/21/2013] [Indexed: 02/08/2023]
Abstract
BACKGROUND Focusing on novel drug combinations that target different pathways especially apoptosis and MAPK could be a rationale for combination therapy in successful treatment of lung cancer. Concurrent use of cyclooxygenase (COX) inhibitors with arsenic trioxide (ATO) might be a possible treatment option. METHODS Cytotoxicity of ATO, dexamethasone (Dex), celecoxib (Cel), and Indomethacin (Indo) individually or in combination was determined at 24, 48, and 72 hrs in A549 lung cancer cells. The COX-2 gene and protein expression, MAPK pathway proteins, and caspase-3 activity were studied for the most cytotoxic combinations. RESULTS The IC50s of ATO and Indo were 68.7 μmol/L and 396.5 μmol/L, respectively. Treatment of cells with combinations of clinically relevant concentrations of ATO and Indo resulted in greater growth inhibition and apoptosis induction than did either agent alone. Caspase-3 activity was considerably high in the presence of ATO and Indo but showed no difference in single or combination use. Phosphorylation of p38 and ERK1/2 was remarkable in the concurrent presence of both drugs. CONCLUSIONS Combination therapy with ATO and Indo exerted a very potent in vitro cytotoxic effect against A549 lung cancer cells. Activation of ERK and p38 pathways might be the mechanism of higher cytotoxic effect of ATO-Indo combination.
Collapse
|
15
|
Zeng L, Li J, Wang Y, Qian C, Chen Y, Zhang Q, Wu W, Lin Z, Liang J, Shuai X, Huang K. Combination of siRNA-directed Kras oncogene silencing and arsenic-induced apoptosis using a nanomedicine strategy for the effective treatment of pancreatic cancer. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2013; 10:463-72. [PMID: 24028894 DOI: 10.1016/j.nano.2013.08.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Revised: 08/19/2013] [Accepted: 08/21/2013] [Indexed: 12/18/2022]
Abstract
UNLABELLED The synergetic inhibitory effects on human pancreatic cancer by nanoparticle-mediated siRNA and arsenic therapy were investigated both in vitro and in vivo. Poly(ethylene glycol)-block-poly(L-lysine) were prepared to form siRNA-complexed polyplex and poly(ethylene glycol)-block-poly(DL-lactide) were prepared to form arsenic-encapsulated vesicle, respectively. Down-regulation of the mutant Kras gene by siRNA caused defective abilities of proliferation, clonal formation, migration, and invasion of pancreatic cancer cells, as well as cell cycle arrest at the G0/G1 phase, which substantially enhanced the apoptosis-inducing effect of arsenic administration. Consequently, co-administration of the two nanomedicines encapsulating siRNA or arsenic showed ideal tumor growth inhibition both in vitro and in vivo as a result of synergistic effect of the siRNA-directed Kras oncogene silencing and arsenic-induced cell apoptosis. These results suggest that the combination of mutant Kras gene silencing and arsenic therapy using nanoparticle-mediated delivery strategy is promising for pancreatic cancer treatment. FROM THE CLINICAL EDITOR Treatment of pancreatic cancer remains a major challenge. These authors demonstrate a method that combines a siRNA-based Kras silencing with arsenic delivery to pancreatic cancer cells using nanoparticles, resulting in enhanced apoptosis induction in the treated cells.
Collapse
Affiliation(s)
- Linjuan Zeng
- Department of Gastroenterology, The Second Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Department of Oncology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Jingguo Li
- PCFM Lab of Ministry of Education, School of Chemistry and Chemical Engineering, Sun Yat-Sen University, Guangzhou 510275, China
| | - Yong Wang
- PCFM Lab of Ministry of Education, School of Chemistry and Chemical Engineering, Sun Yat-Sen University, Guangzhou 510275, China
| | - Chenchen Qian
- Department of Gastroenterology, The Second Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yinting Chen
- Department of Gastroenterology, The Second Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qiubo Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wei Wu
- Department of Cardiology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Zhong Lin
- Department of Oncology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Jianzhong Liang
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xintao Shuai
- PCFM Lab of Ministry of Education, School of Chemistry and Chemical Engineering, Sun Yat-Sen University, Guangzhou 510275, China.
| | - Kaihong Huang
- Department of Gastroenterology, The Second Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
16
|
Rathos MJ, Khanwalkar H, Joshi K, Manohar SM, Joshi KS. Potentiation of in vitro and in vivo antitumor efficacy of doxorubicin by cyclin-dependent kinase inhibitor P276-00 in human non-small cell lung cancer cells. BMC Cancer 2013; 13:29. [PMID: 23343191 PMCID: PMC3635914 DOI: 10.1186/1471-2407-13-29] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Accepted: 01/16/2013] [Indexed: 12/17/2022] Open
Abstract
Background In the present study, we show that the combination of doxorubicin with the cyclin-dependent kinase inhibitor P276-00 was synergistic at suboptimal doses in the non-small cell lung carcinoma (NSCLC) cell lines and induces extensive apoptosis than either drug alone in H-460 human NSCLC cells. Methods Synergistic effects of P276-00 and doxorubicin on growth inhibition was studied using the Propidium Iodide (PI) assay. The doses showing the best synergistic effect was determined and these doses were used for further mechanistic studies such as western blotting, cell cycle analysis and RT-PCR. The in vivo efficacy of the combination was evaluated using the H-460 xenograft model. Results The combination of 100 nM doxorubicin followed by 1200 nM P276-00 showed synergistic effect in the p53-positive and p53-mutated cell lines H-460 and H23 respectively as compared to the p53-null cell line H1299. Abrogation of doxorubicin-induced G2/M arrest and induction of apoptosis was observed in the combination treatment. This was associated with induction of tumor suppressor protein p53 and reduction of anti-apoptotic protein Bcl-2. Furthermore, doxorubicin alone greatly induced COX-2, a NF-κB target and Cdk-1, a target of P276-00, which was downregulated by P276-00 in the combination. Doxorubicin when combined with P276-00 in a sequence-specific manner significantly inhibited tumor growth, compared with either doxorubicin or P276-00 alone in H-460 xenograft model. Conclusion These findings suggest that this combination may increase the therapeutic index over doxorubicin alone and reduce systemic toxicity of doxorubicin most likely via an inhibition of doxorubicin-induced chemoresistance involving NF-κB signaling and inhibition of Cdk-1 which is involved in cell cycle progression.
Collapse
Affiliation(s)
- Maggie J Rathos
- Oncology Franchise, Piramal Healthcare Limited, 1-Nirlon Complex, Goregaon, Mumbai 400 063, India
| | | | | | | | | |
Collapse
|
17
|
Ayyanathan K, Kesaraju S, Dawson-Scully K, Weissbach H. Combination of sulindac and dichloroacetate kills cancer cells via oxidative damage. PLoS One 2012; 7:e39949. [PMID: 22866174 PMCID: PMC3398923 DOI: 10.1371/journal.pone.0039949] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Accepted: 06/04/2012] [Indexed: 01/28/2023] Open
Abstract
Sulindac is an FDA-approved non-steroidal anti-inflammatory drug with documented anticancer activities. Our recent studies showed that sulindac selectively enhanced the killing of cancer cells exposed to oxidizing agents via production of reactive oxygen species (ROS) resulting in mitochondrial dysfunction. This effect of sulindac and oxidative stress on cancer cells could be related to the defect in respiration in cancer cells, first described by Warburg 50 years ago, known as the Warburg effect. We postulated that sulindac might enhance the selective killing of cancer cells when combined with any compound that alters mitochondrial respiration. To test this hypothesis we have used dichloroacetate (DCA), which is known to shift pyruvate metabolism away from lactic acid formation to respiration. One might expect that DCA, since it stimulates aerobic metabolism, could stress mitochondrial respiration in cancer cells, which would result in enhanced killing in the presence of sulindac. In this study, we have shown that the combination of sulindac and DCA enhances the selective killing of A549 and SCC25 cancer cells under the conditions used. As predicted, the mechanism of killing involves ROS production, mitochondrial dysfunction, JNK signaling and death by apoptosis. Our results suggest that the sulindac-DCA drug combination may provide an effective cancer therapy.
Collapse
Affiliation(s)
- Kasirajan Ayyanathan
- Center for Molecular Biology and Biotechnology, Charles E. Schmidt College of Science, Florida Atlantic University, Jupiter, Florida, United States of America.
| | | | | | | |
Collapse
|
18
|
Jang M, Kim Y, Won H, Lim S, K R J, Dashdorj A, Min YH, Kim SY, Shokat KM, Ha J, Kim SS. Carbonyl reductase 1 offers a novel therapeutic target to enhance leukemia treatment by arsenic trioxide. Cancer Res 2012; 72:4214-24. [PMID: 22719067 DOI: 10.1158/0008-5472.can-12-1110] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Arsenic trioxide (As2O3) is used, in current practice, as an effective chemotherapeutic agent for acute promyelocytic leukemia (APL). However, the side effects and relatively low efficacy of As2O3 in treating other leukemias have limited its wider use in therapeutic applications. In the present study, we found that the expression of carbonyl reductase 1 (CBR1) affects the resistance to As2O3 in leukemias, including APL; As2O3 upregulated CBR1 expression at the transcriptional level by stimulating the activity of the transcription factor activator protein-1. Moreover, CBR1 overexpression was sufficient to protect cells against As2O3 through modulation of the generation of reactive oxygen species, whereas the attenuation of CBR1 was sufficient to sensitize cells to As2O3. A combination treatment with the specific CBR1 inhibitor hydroxy-PP-Me remarkably increased As2O3-induced apoptotic cell death compared with As2O3 alone, both in vitro and in vivo. These results were confirmed in primary cultured human acute and chronic myeloid leukemia cells, with no significant cell death observed in normal leukocytes. Taken together, our findings indicate that CBR1 contributes to the low efficacy of As2O3 and, therefore, is a rational target for the development of combination chemotherapy with As2O3 in diverse leukemias including APL.
Collapse
Affiliation(s)
- Miran Jang
- Department of Biochemistry and Molecular Biology (BK21 project), Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, School of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Kuo CC, Liu TW, Chen LT, Shiah HS, Wu CM, Cheng YT, Pan WY, Liu JF, Chen KL, Yang YN, Chen SN, Chang JY. Combination of arsenic trioxide and BCNU synergistically triggers redox-mediated autophagic cell death in human solid tumors. Free Radic Biol Med 2011; 51:2195-209. [PMID: 22001324 DOI: 10.1016/j.freeradbiomed.2011.09.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Revised: 09/20/2011] [Accepted: 09/21/2011] [Indexed: 12/14/2022]
Abstract
Arsenic trioxide (As(2)O(3)) is an effective treatment for relapsed or refractory acute promyelocytic leukemia (APL). After the discovery of As(2)O(3) as a promising treatment for APL, several studies investigated the use of As(2)O(3) as a single agent in the treatment of solid tumors; however, its therapeutic efficacy is limited. Thus, the systematic study of the combination of As(2)O(3) with other clinically used chemotherapeutic drugs to improve its therapeutic efficacy in treating human solid tumors is merited. In this study, we demonstrate for the first time, using isobologram analysis, that As(2)O(3) exhibits a synergistic interaction with N,N'-bis(2-chloroethyl)-N-nitrosourea (BCNU). The synergistic augmentation of the cytotoxicity of As(2)O(3) with BCNU is in part through the autophagic cell death machinery in human solid tumor cells. As(2)O(3) and BCNU in combination produce enhanced cytotoxicity via the depletion of reduced glutathione (GSH) and augmentation of reaction oxygen species (ROS) production. Further analysis indicated that the extension of GSH depletion by this combined regimen occurs through the inhibition of the catalytic activity of glutathione reductase. Blocking ROS production with antioxidants or ROS scavengers effectively inhibits cell death and autophagy formation, indicating that redox-mediated autophagic cell death involves the synergism of As(2)O(3) with BCNU. Taken together, this is the first evidence that BCNU could help to extend the therapeutic spectrum of As(2)O(3). These findings will be useful in designing future clinical trials of combination chemotherapy with As(2)O(3) and BCNU, with the potential for broad use against a variety of solid tumors.
Collapse
Affiliation(s)
- Ching-Chuan Kuo
- National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Stępnik M, Ferlińska M, Smok-Pieniążek A, Gradecka-Meesters D, Arkusz J, Stańczyk M. Sulindac and its metabolites: Sulindac sulfide and sulindac sulfone enhance cytotoxic effects of arsenic trioxide on leukemic cell lines. Toxicol In Vitro 2011; 25:1075-84. [DOI: 10.1016/j.tiv.2011.04.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2010] [Revised: 03/10/2011] [Accepted: 04/07/2011] [Indexed: 12/21/2022]
|
21
|
El Fajoui Z, Toscano F, Jacquemin G, Abello J, Scoazec JY, Micheau O, Saurin JC. Oxaliplatin sensitizes human colon cancer cells to TRAIL through JNK-dependent phosphorylation of Bcl-xL. Gastroenterology 2011; 141:663-73. [PMID: 21683075 DOI: 10.1053/j.gastro.2011.04.055] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Revised: 04/03/2011] [Accepted: 04/22/2011] [Indexed: 01/16/2023]
Abstract
BACKGROUND & AIMS Oxaliplatin sensitizes drug-resistant colon cancer cell lines to tumor necrosis factor-related apoptosis inducing ligand (TRAIL), a death receptor ligand that is selective for cancer cells. We investigated the molecular mechanisms by which oxaliplatin sensitizes cancer cells to TRAIL-induced apoptosis. METHODS We incubated the colon cancer cell lines HT29 and V9P, which are resistant to TRAIL, with TRAIL or with oxaliplatin for 2 hours, followed by TRAIL. Annexin V staining was used to measure apoptosis; RNA silencing and immunoblot experiments were used to study the roles of apoptosis-related proteins. Site-directed mutagenesis experiments were used to determine requirements for phosphorylation of Bcl-xL; co-immunoprecipitation experiments were used to analyze the interactions among Bcl-xL, Bax, and Bak, and activation of Bax. RESULTS Oxaliplatin-induced sensitivity to TRAIL required activation of the mitochondrial apoptotic pathway; reduced expression of Bax, Bak, and caspase-9, and stable overexpression of Bcl-xL, reduced TRAIL-induced death of cells incubated with oxaliplatin. Mitochondrial priming was induced in cells that were sensitized by oxaliplatin and required signaling via c-Jun N-terminal kinase and phosphorylation of Bcl-xL. Mimicking constitutive phosphorylation of Bcl-xL by site-directed mutagenesis at serine 62 restored sensitivity of cells to TRAIL. Co-immunoprecipitation experiments showed that oxaliplatin-induced phosphorylation of Bcl-xL disrupted its ability to sequestrate Bax, allowing Bax to interact with Bak to induce TRAIL-mediated apoptosis. CONCLUSIONS Oxaliplatin facilitates TRAIL-induced apoptosis in colon cancer cells by activating c-Jun N-terminal kinase signaling and phosphorylation of Bcl-xL. Oxaliplatin-induced sensitivity to TRAIL might be developed as an approach to cancer therapy.
Collapse
Affiliation(s)
- Zineb El Fajoui
- INSERM U865, University of Lyon, Faculté Laënnec, Lyon, France
| | | | | | | | | | | | | |
Collapse
|
22
|
E3 ubiquitin ligase Hades negatively regulates the exonuclear function of p53. Cell Death Differ 2011; 18:1865-75. [PMID: 21597459 DOI: 10.1038/cdd.2011.57] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Following DNA damage, p53 translocates to the cytoplasm and mitochondria, where it triggers transcription-independent apoptosis by binding to Bcl-2 family proteins. However, little is known about how this exonuclear function of p53 is regulated. Here, we identify and characterize a p53-interacting protein called Hades, an E3 ligase that interacts with p53 in the mitochondria. Hades reduces p53 stability via a mechanism that requires its RING-finger domain with ubiquitin ligase activity. Hades polyubiquitinates p53 in vitro independent of Mdm2 and targets a critical lysine residue in p53 (lysine 24) distinct from those targeted by Mdm2. Hades inhibits a p53-dependent mitochondrial cell death pathway by inhibiting p53 and Bcl-2 interactions. These findings show that Hades-mediated p53 ubiquitination is a novel mechanism for negatively regulating the exonuclear function of p53.
Collapse
|
23
|
Assessment of the involvement of oxidative stress and Mitogen-Activated Protein Kinase signaling pathways in the cytotoxic effects of arsenic trioxide and its combination with sulindac or its metabolites: sulindac sulfide and sulindac sulfone on human leukemic cell lines. Med Oncol 2011; 29:1161-72. [DOI: 10.1007/s12032-011-9920-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Accepted: 03/21/2011] [Indexed: 12/21/2022]
|
24
|
Chen KC, Liu WH, Kao PH, Chang LS. Calcium-stimulated mitogen-activated protein kinase activation elicits Bcl-xL downregulation and Bak upregulation in notexin-treated human neuroblastoma SK-N-SH cells. J Cell Physiol 2009; 222:177-86. [PMID: 19780038 DOI: 10.1002/jcp.21934] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Notechis scutatus scutatus notexin induced apoptotic death of SK-N-SH cells accompanied with downregulation of Bcl-xL, upregulation of Bak, mitochondrial depolarization, and ROS generation. Upon exposure to notexin, Ca(2+)-mediated JNK and p38 MAPK activation were observed in SK-N-SH cells. Production of ROS was a downstream event followed by Ca(2+)-mediated mitochondrial alteration. Notexin-induced cell death, mitochondrial depolarization, and ROS generation were suppressed by SB202190 (p38 MAPK inhibitor) and SP600125 (JNK inhibitor). Moreover, phospho-p38 MAPK and phospho-JNK were proved to be involved in Bcl-xL degradation, and overexpression of Bcl-xL attenuated the cytotoxic effect of notexin. Bak upregulation was elicited by p38 MAPK-mediated ATF-2 activation and JNK-mediated c-Jun activation. Suppression of Bak upregulation by ATF-2 siRNA or c-Jun siRNA attenuated notexin-evoked mitochondrial depolarization and rescued viability of notexin-treated cells. Taken together, our data indicate that notexin-induced apoptotic death of SK-N-SH cells is mediated through mitochondrial alteration triggering by Ca(2+)-evoked p38 MAPK/ATF-2 and JNK/c-Jun signaling pathways.
Collapse
Affiliation(s)
- Ku-Chung Chen
- Institute of Biomedical Sciences, National Sun Yat-Sen University-Kaohsiung Medical University Joint Research Center, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | | | | | | |
Collapse
|
25
|
Yan S, Zhang Q, Zhou B, Xue L, Chen H, Wang Y, Zheng S. Arsenic Trioxide Attenuated the Rejection of Major Histocompatibility Complex Fully-Mismatched Cardiac Allografts in Mice. Transplant Proc 2009; 41:1855-8. [DOI: 10.1016/j.transproceed.2009.02.073] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2008] [Revised: 12/19/2008] [Accepted: 02/09/2009] [Indexed: 11/17/2022]
|
26
|
Doyle D. Notoriety to respectability: a short history of arsenic prior to its present day use in haematology. Br J Haematol 2009; 145:309-17. [DOI: 10.1111/j.1365-2141.2009.07623.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|