1
|
Yu Y, Shang Y, Shi S, He Y, Shi W, Wang M, Wang Q, Xu D, Shi C, Chen H. Combination of arsenic trioxide and apatinib synergistically inhibits small cell lung cancer by down-regulating VEGFR2/mTOR and Akt/c-Myc signaling pathway via GRB10. Hereditas 2024; 161:29. [PMID: 39223679 PMCID: PMC11367874 DOI: 10.1186/s41065-024-00330-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 08/10/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Small cell lung carcinoma (SCLC) is characterized by -poor prognosis, -high predilection for -metastasis, -proliferation, and -absence of newer therapeutic options. Elucidation of newer pathways characterizing the disease may allow for development of targeted therapies and consequently favorable outcomes. METHODS The current study explored the combinatorial action of arsenic trioxide (ATO) and apatinib (APA) in vitro and in vivo. In vitro models were tested using -H446 and -H196 SCLC cell lines. The ability of drugs to reduce -metastasis, -cell proliferation, and -migration were assessed. Using bioinformatic analysis, differentially expressed genes were determined. Gene regulation was assessed using gene knock down models and confirmed using Western blots. The in vivo models were used to confirm the resolution of pathognomic features in the presence of the drugs. Growth factor receptor bound protein (GRB) 10 expression levels of human small cell lung cancer tissues and adjacent tissues were detected by IHC. RESULTS In combination, ATO and APA were found to significantly reduce -cell proliferation, -migration, and -metastasis in both the cell lines. Cell proliferation was found to be inhibited by activation of Caspase-3, -7 pathway. In the presence of drugs, it was found that expression of GRB10 was stabilized. The silencing of GRB10 was found to negatively regulate the VEGFR2/Akt/mTOR and Akt/GSK-3β/c-Myc signaling pathway. Concurrently, absence of metastasis and reduction of tumor volume were confirmed in vivo. The immunohistochemical results confirmed that the expression level of GRB10 in adjacent tissues was significantly higher than that in human small cell lung cancer tissues. CONCLUSIONS Synergistically, ATO and APA have a more significant impact on inhibiting cell proliferation than each drug independently. ATO and APA may be mediating its action through the stabilization of GRB10 thus acting as a tumor suppressor. We thus, preliminarily report the impact of GRB10 stability as a target for SCLC treatment.
Collapse
Affiliation(s)
- Yao Yu
- Harbin Medical University, Harbin, Heilongjiang Province, China
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yu Shang
- Department of Respiration, The First Hospital of Harbin, Harbin, Heilongjiang Province, China
| | - Si Shi
- Harbin Medical University, Harbin, Heilongjiang Province, China
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yaowu He
- Harbin Medical University, Harbin, Heilongjiang Province, China
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Wenchao Shi
- Harbin Medical University, Harbin, Heilongjiang Province, China
- Department of Pulmonary and Critical Care Medicine, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Menghan Wang
- Harbin Medical University, Harbin, Heilongjiang Province, China
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Qi Wang
- Harbin Medical University, Harbin, Heilongjiang Province, China
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Dandan Xu
- Harbin Medical University, Harbin, Heilongjiang Province, China
- Department of Geriatric Respiratory Medicine, Heilongjiang Provincial Hospital, Harbin, China
| | - Ce Shi
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University (HMU), Harbin, Heilongjiang Province, China.
| | - Hong Chen
- Harbin Medical University, Harbin, Heilongjiang Province, China.
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China.
| |
Collapse
|
2
|
Liu F, Deng Y, Wang A, Yang T, Ke H, Tang Y, Wu H, Chen H. Harness arsenic in medicine: current status of arsenicals and recent advances in drug delivery. Expert Opin Drug Deliv 2024; 21:867-880. [PMID: 38913024 DOI: 10.1080/17425247.2024.2372363] [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: 01/21/2024] [Accepted: 06/21/2024] [Indexed: 06/25/2024]
Abstract
INTRODUCTION Arsenicals have a special place in the history of human health, acting both as poison and medicine. Having been used to treat a variety of diseases in the past, the success of arsenic trioxide (ATO) in treating acute promyelocytic leukemia (APL) in the last century marked its use as a drug in modern medicine. To expand their role against cancer, there have been clinical uses of arsenicals worldwide and progress in the development of drug delivery for various malignancies, especially solid tumors. AREAS COVERED In this review, conducted on Google Scholar [1977-2024], we start with various forms of arsenicals, highlighting the well-known ATO. The mechanism of action of arsenicals in cancer therapy is then overviewed. A summary of the research progress in developing new delivery approaches (e.g. polymers, inorganic frameworks, and biomacromolecules) in recent years is provided, addressing the challenges and opportunities in treating various malignant tumors. EXPERT OPINION Reducing toxicity and enhancing therapeutic efficacy are guidelines for designing and developing new arsenicals and drug delivery systems. They have shown potential in the fight against cancer and emerging pathogens. New technologies and strategies can help us harness the potency of arsenicals and make better products.
Collapse
Affiliation(s)
- Fan Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, and College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Yibin Deng
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, and College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Anru Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, and College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Tao Yang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, and College of Pharmaceutical Sciences, Soochow University, Suzhou, China
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Hengte Ke
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, and College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Yongan Tang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, and College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Hong Wu
- Department of Pharmaceutical Analysis, School of Pharmacy, Air Force Medical University, Xi'an, China
| | - Huabing Chen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, and College of Pharmaceutical Sciences, Soochow University, Suzhou, China
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute for Interdisciplinary Drug Research and Translational Sciences, Soochow University, Suzhou, China
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou, China
| |
Collapse
|
3
|
Dubey S, Mishra N, Goswami N, Siddiqui MQ, Varma AK. Multimodal approach to characterize the tetrameric form of human PML-RBCC domain and ATO-mediated conformational changes. Int J Biol Macromol 2022; 223:468-478. [PMID: 36356867 DOI: 10.1016/j.ijbiomac.2022.11.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/17/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022]
Abstract
RING-B box-coiled coil (RBCC) domain of promyelocytic leukemia (PML) comprises a zinc finger motif that is targeted by arsenic trioxide (ATO) to treat acute promyelocytic leukemia (APL) pathogenesis. Preliminary evidence suggests that the PML-RBCC has different functional characteristics, but no structural details have been reported despite its importance in differential expression and cell-cycle regulation. Therefore, the recombinant h-PML-RBCC protein was purified to its homogeneity, and characterized for oligomeric behaviour which indicated that RBCC domain exists as a tetramer in solution. Furthermore, nano-DSF and circular-dichroism demonstrated that the tetrameric form preserves its native conformation along with thermal stability (Tm = 83.2 °C). In-silico-based PML-RBCC structure was used to perform the molecular dynamics simulation for 300 ns in the presence of zinc atoms, which demonstrated the differential dynamic of PML-RBCC tetrameric chains. MMPBSA analysis also indicated the role of hydrophobic interactions that favours stable tetrameric structure of PML-RBCC. ATO-induced secondary and tertiary structure changes were observed in PML-RBCC using circular dichroism and fluorescence spectroscopy. Dynamic light scattering and transmission electron microscopy revealed ATO-induced higher-order oligomerization and aggregation of PML-RBCC. The unique oligomeric nature of the h-PML-RBCC protein and its interactions with ATO will help to understand the mechanism of APL pathogenesis and drug resistance.
Collapse
Affiliation(s)
- Suchita Dubey
- Advanced Centre for Treatment, Research and Education in Cancer, Kharghar, Navi Mumbai, Maharashtra 410210, India; Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai 400094, India
| | - Neha Mishra
- Advanced Centre for Treatment, Research and Education in Cancer, Kharghar, Navi Mumbai, Maharashtra 410210, India; Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai 400094, India
| | - Nabajyoti Goswami
- Advanced Centre for Treatment, Research and Education in Cancer, Kharghar, Navi Mumbai, Maharashtra 410210, India
| | - M Quadir Siddiqui
- Advanced Centre for Treatment, Research and Education in Cancer, Kharghar, Navi Mumbai, Maharashtra 410210, India
| | - Ashok K Varma
- Advanced Centre for Treatment, Research and Education in Cancer, Kharghar, Navi Mumbai, Maharashtra 410210, India; Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai 400094, India.
| |
Collapse
|
4
|
Burska AN, Ilyassova B, Dildabek A, Khamijan M, Begimbetova D, Molnár F, Sarbassov DD. Enhancing an Oxidative "Trojan Horse" Action of Vitamin C with Arsenic Trioxide for Effective Suppression of KRAS-Mutant Cancers: A Promising Path at the Bedside. Cells 2022; 11:3454. [PMID: 36359850 PMCID: PMC9657932 DOI: 10.3390/cells11213454] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/19/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022] Open
Abstract
The turn-on mutations of the KRAS gene, coding a small GTPase coupling growth factor signaling, are contributing to nearly 25% of all human cancers, leading to highly malignant tumors with poor outcomes. Targeting of oncogenic KRAS remains a most challenging task in oncology. Recently, the specific G12C mutant KRAS inhibitors have been developed but with a limited clinical outcome because they acquire drug resistance. Alternatively, exploiting a metabolic breach of KRAS-mutant cancer cells related to a glucose-dependent sensitivity to oxidative stress is becoming a promising indirect cancer targeting approach. Here, we discuss the use of a vitamin C (VC) acting in high dose as an oxidative "Trojan horse" agent for KRAS-mutant cancer cells that can be potentiated with another oxidizing drug arsenic trioxide (ATO) to obtain a potent and selective cytotoxic impact. Moreover, we outline the advantages of VC's non-natural enantiomer, D-VC, because of its distinctive pharmacokinetics and lower toxicity. Thus, the D-VC and ATO combination shows a promising path to treat KRAS-mutant cancers in clinical settings.
Collapse
Affiliation(s)
- Agata N. Burska
- Department of Biology, Nazarbayev University, Astana 010000, Kazakhstan
| | | | - Aruzhan Dildabek
- Department of Biology, Nazarbayev University, Astana 010000, Kazakhstan
| | - Medina Khamijan
- Department of Biology, Nazarbayev University, Astana 010000, Kazakhstan
| | - Dinara Begimbetova
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan
| | - Ferdinand Molnár
- Department of Biology, Nazarbayev University, Astana 010000, Kazakhstan
| | - Dos D. Sarbassov
- Department of Biology, Nazarbayev University, Astana 010000, Kazakhstan
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan
| |
Collapse
|
5
|
Lospinoso Severini L, Ghirga F, Bufalieri F, Quaglio D, Infante P, Di Marcotullio L. The SHH/GLI signaling pathway: a therapeutic target for medulloblastoma. Expert Opin Ther Targets 2020; 24:1159-1181. [PMID: 32990091 DOI: 10.1080/14728222.2020.1823967] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Medulloblastoma (MB) is a heterogeneous tumor of the cerebellum that is divided into four main subgroups with distinct molecular and clinical features. Sonic Hedgehog MB (SHH-MB) is the most genetically understood and occurs predominantly in childhood. Current therapies consist of aggressive and non-targeted multimodal approaches that are often ineffective and cause long-term complications. These problems intensify the need to develop molecularly targeted therapies to improve outcome and reduce treatment-related morbidities. In this scenario, Hedgehog (HH) signaling, a developmental pathway whose deregulation is involved in the pathogenesis of several malignancies, has emerged as an attractive druggable pathway for SHH-MB therapy. AREAS COVERED This review provides an overview of the advancements in the HH antagonist research field. We place an emphasis on Smoothened (SMO) and glioma-associated oncogene homolog (GLI) inhibitors and immunotherapy approaches that are validated in preclinical SHH-MB models and that have therapeutic potential for MB patients. Literature from Pubmed and data reported on ClinicalTrial.gov up to August 2020 were considered. EXPERT OPINION Extensive-omics analysis has enhanced our knowledge and has transformed the way that MB is studied and managed. The clinical use of SMO antagonists has yet to be determined, however, future GLI inhibitors and multitargeting approaches are promising.
Collapse
Affiliation(s)
| | - Francesca Ghirga
- Center for Life NanoScience@Sapienza, Istituto Italiano di Tecnologia , 00161, Rome, Italy
| | - Francesca Bufalieri
- Department of Molecular Medicine, University of Rome La Sapienza , 00161, Rome, Italy
| | - Deborah Quaglio
- Department of Chemistry and Technology of Drugs, University of Rome La Sapienza, 00185 , Rome, Italy
| | - Paola Infante
- Center for Life NanoScience@Sapienza, Istituto Italiano di Tecnologia , 00161, Rome, Italy
| | - Lucia Di Marcotullio
- Department of Molecular Medicine, University of Rome La Sapienza , 00161, Rome, Italy.,Istituto Pasteur-Fondazione Cenci Bolognetti, University of Rome La Sapienza , 00161, Rome, Italy
| |
Collapse
|
6
|
An K, Xue MJ, Zhong JY, Yu SN, Lan TS, Qi ZQ, Xia JJ. Arsenic trioxide ameliorates experimental autoimmune encephalomyelitis in C57BL/6 mice by inducing CD4 + T cell apoptosis. J Neuroinflammation 2020; 17:147. [PMID: 32375831 PMCID: PMC7201567 DOI: 10.1186/s12974-020-01829-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 04/27/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is an immune-mediated disease of the central nervous system characterized by severe white matter demyelination. Because of its complex pathogenesis, there is no definite cure for MS. Experimental autoimmune encephalomyelitis (EAE) is an ideal animal model for the study of MS. Arsenic trioxide (ATO) is an ancient Chinese medicine used for its therapeutic properties with several autoimmune diseases. It is also used to inhibit acute immune rejection due to its anti-inflammatory and immunosuppressive properties. However, it is unclear whether ATO has a therapeutic effect on EAE, and the underlying mechanisms have not yet been clearly elucidated. In this study, we attempted to assess whether ATO could be used to ameliorate EAE in mice. METHODS ATO (0.5 mg/kg/day) was administered intraperitoneally to EAE mice 10 days post-immunization for 8 days. On day 22 post-immunization, the spinal cord, spleen, and blood were collected to analyze demyelination, inflammation, microglia activation, and the proportion of CD4+ T cells. In vitro, for mechanistic studies, CD4+ T cells were sorted from the spleen of naïve C57BL/6 mice and treated with ATO and then used for an apoptosis assay, JC-1 staining, imaging under a transmission electron microscope, and western blotting. RESULTS ATO delayed the onset of EAE and alleviated the severity of EAE in mice. Treatment with ATO also attenuated demyelination, alleviated inflammation, reduced microglia activation, and decreased the expression levels of IL-2, IFN-γ, IL-1β, IL-6, and TNF-α in EAE mice. Moreover, the number and proportion of CD4+ T cells in the spinal cord, spleen, and peripheral blood were reduced in ATO-treated EAE mice. Finally, ATO induced CD4+ T cell apoptosis via the mitochondrial pathway both in vitro and in vivo. Additionally, the administration of ATO had no adverse effect on the heart, liver, or kidney function, nor did it induce apoptosis in the spinal cord. CONCLUSIONS Overall, our findings indicated that ATO plays a protective role in the initiation and progression of EAE and has the potential to be a novel drug in the treatment of MS.
Collapse
Affiliation(s)
- Ke An
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Meng-Jiao Xue
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Jia-Ying Zhong
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Sheng-Nan Yu
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, China
- Department of Obstetrics and Gynecology, Zhongshan Hospital, Xiamen University, Xiamen, Fujian, China
| | - Tian-Shu Lan
- Key Laboratory of Functional and Clinical Translational Medicine, Fujian Province University, Xiamen Medical College, Xiamen, Fujian, China
| | - Zhong-Quan Qi
- School of Medicine, Guangxi University, Nanning, Guangxi, China.
| | - Jun-Jie Xia
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
7
|
Luo H, Wang J, Li M, Luo J, Ni P, Zhao K, Wu FX, Pan Y. Computational Drug Repositioning with Random Walk on a Heterogeneous Network. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2019; 16:1890-1900. [PMID: 29994051 DOI: 10.1109/tcbb.2018.2832078] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Drug repositioning is an efficient and promising strategy to identify new indications for existing drugs, which can improve the productivity of traditional drug discovery and development. Rapid advances in high-throughput technologies have generated various types of biomedical data over the past decades, which lay the foundations for furthering the development of computational drug repositioning approaches. Although many researches have tried to improve the repositioning accuracy by integrating information from multiple sources and different levels, it is still appealing to further investigate how to efficiently exploit valuable data for drug repositioning. In this study, we propose an efficient approach, Random Walk on a Heterogeneous Network for Drug Repositioning (RWHNDR), to prioritize candidate drugs for diseases. First, an integrated heterogeneous network is constructed by combining multiple sources including drugs, drug targets, diseases and disease genes data. Then, a random walk model is developed to capture the global information of the heterogeneous network. RWHNDR takes advantage of drug targets and disease genes data more comprehensively for drug repositioning. The experiment results show that our approach can achieve better performance, compared with other state-of-the-art approaches which prioritized candidate drugs based on multi-source data.
Collapse
|
8
|
Carpenter RL, Ray H. Safety and Tolerability of Sonic Hedgehog Pathway Inhibitors in Cancer. Drug Saf 2019; 42:263-279. [PMID: 30649745 DOI: 10.1007/s40264-018-0777-5] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The hedgehog pathway, for which sonic hedgehog (Shh) is the most prominent ligand, is highly conserved and is tightly associated with embryonic development in a number of species. This pathway is also tightly associated with the development of several types of cancer, including basal cell carcinoma (BCC) and acute promyelocytic leukemia, among many others. Inactivating mutations in Patched-1 (PTCH1), leading to ligand-independent pathway activation, are frequent in several cancer types, but most prominent in BCC. This has led to the development of several compounds targeting this pathway as a cancer therapeutic. These compounds target the inducers of this pathway in Smoothened (SMO) and the GLI transcription factors, although targeting SMO has had the most success. Despite the many attempts at targeting this pathway, only three US FDA-approved drugs for cancers affect the Shh pathway. Two of these compounds, vismodegib and sonidegib, target SMO to suppress signaling from either PTCH1 or SMO mutations that lead to upregulation of the pathway. The other approved compound is arsenic trioxide, which can suppress this pathway at the level of the GLI proteins, although current evidence suggests it also has other targets. This review focuses on the safety and tolerability of these clinically approved drugs targeting the Shh pathway, along with a discussion on other Shh pathway inhibitors being developed.
Collapse
Affiliation(s)
- Richard L Carpenter
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, 1001 E. 3rd St, Bloomington, IN, 47405, USA. .,Medical Sciences, Indiana University School of Medicine, 1001 E. 3rd St, Bloomington, IN, 47405, USA. .,Simon Cancer Center, Indiana University School of Medicine, 535 Barnhill Dr., Indianapolis, IN, 46202, USA.
| | - Haimanti Ray
- Medical Sciences, Indiana University School of Medicine, 1001 E. 3rd St, Bloomington, IN, 47405, USA
| |
Collapse
|
9
|
Gao J, Wang G, Wu J, Zuo Y, Zhang J, Jin X. Skp2 Expression Is Inhibited by Arsenic Trioxide through the Upregulation of miRNA-330-5p in Pancreatic Cancer Cells. Mol Ther Oncolytics 2019; 12:214-223. [PMID: 30847385 PMCID: PMC6389777 DOI: 10.1016/j.omto.2019.01.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 01/27/2019] [Indexed: 12/26/2022] Open
Abstract
Arsenic trioxide (ATO) has been found to exert its anti-cancer activity in various human malignancies. In our previous report, we have shown that ATO inhibited cell growth and invasion via downregulation of Skp2 in pancreatic cancer (PC) cells. It has been extensively demonstrated that microRNAs (miRNAs) play a pivotal role in tumorigenesis. ATO might induce PC cell apoptosis and regulate Skp2 downregulation through the regulation of miRNAs. One study has demonstrated that miR-330-5p exerts a tumor-suppressive function in PC cell lines. Here, we investigated the role of miRNA-330-5p in ATO-mediated anti-tumor activity and explored whether ATO could regulate miR-330-5p in PC cells. We found that ATO treatment upregulated the expression of miR-330-5p. Moreover, miR-330-5p inhibitor rescued the ATO-mediated tumor-suppressive function. The combination of miR-330-5p mimic with ATO reduced cell growth, motility, and invasion, and enhanced apoptosis to a greater degree in PC cells. This study suggests that the combination of miR-330-5p mimic with ATO may be a potential therapeutic strategy for the treatment of PC.
Collapse
Affiliation(s)
- Jiankun Gao
- Department of Basic Medical Science, Sichuan College of Traditional Chinese Medicine, Mianyang, Sichuan 621000, China
- Corresponding author: Jiankun Gao, Department of Basic Medical Science, Sichuan College of Traditional Chinese Medicine, Mianyang, 621000 Sichuan, China.
| | - Gu Wang
- Department of Basic Medical Science, Sichuan College of Traditional Chinese Medicine, Mianyang, Sichuan 621000, China
| | - Jingrong Wu
- Department of Basic Medical Science, Sichuan College of Traditional Chinese Medicine, Mianyang, Sichuan 621000, China
| | - Yu Zuo
- Department of Basic Medical Science, Sichuan College of Traditional Chinese Medicine, Mianyang, Sichuan 621000, China
| | - Jing Zhang
- Department of Basic Medical Science, Sichuan College of Traditional Chinese Medicine, Mianyang, Sichuan 621000, China
| | - Xintian Jin
- Department of Thoracic Oncosurgery, Jilin Province Cancer Hospital, Changchun, Jilin 130012, China
- Corresponding author: Xintian Jin, Department of Thoracic Oncosurgery, Jilin Province Cancer Hospital, Changchun, Jilin 130012, China.
| |
Collapse
|
10
|
Lakshmaiah KC, Chaudhuri T, Babu GK, Lokanatha D, Jacob LA, Suresh Babu MC, Rudresha AH, Lokesh KN, Rajeev LK. Safety and antitumor activity of arsenic trioxide plus infusional 5-fluorouracil, leucovorin, and irinotecan as second-line chemotherapy for refractory metastatic colorectal cancer: A pilot study from South India. Indian J Cancer 2018; 54:631-633. [PMID: 30082548 DOI: 10.4103/ijc.ijc_374_17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
BACKGROUND After failing oxaliplatin-based first-line chemotherapy (CT), approximately 4%-21% of patients with metastatic colorectal cancer (mCRC) respond to irinotecan-based second-line treatment. Earlier studies have demonstrated that arsenic trioxide (ATO) can significantly resensitize resistant colon cancer to 5-fluorouracil (5-FU) by downregulating thymidylate synthase (TS). We hypothesized that a combination of ATO with infusional 5-FU, leucovorin, and irinotecan (FOLFIRI) regimen in mCRC patients refractory to first-line FOLFOX/CAPOX could further improve the outcome of second-line CT. MATERIALS AND METHODS Patients were administered ATO 0.15 mg/kg/day on days 1-2 along with FOLFIRI regimen at standard doses every 2 weeks, until disease progression, unacceptable toxicity, or patients' refusal. Responses to CT were reported according to RECIST 1.1. Adverse events were classified based on CTCAE version 4.0. RESULTS Between September 2016 and July 2017, 17 patients with refractory mCRC were treated with this investigational combination. The median age was 49 years; 13 males and 4 females; ECOG PS 0-1/2, 14/3. The most common site of metastases was liver (n = 11) followed by peritoneum (n = 7) and number of involved metastatic sites 1-2/≥3, 9/8. After 6 cycles of CT, overall response rate and disease control rate were 17.6% and 82.4%, respectively (complete remission = 0, partial remission = 3 patients, stable disease = 11 patients). Median progression-free survival was 5.3 months (95% confidence interval [CI]: 4.3-7.0) and median overall survival was 9 months (95% CI: 7.4-10.5) from the initiation of ATO plus FOLFIRI. The toxicities were as follows: Grade 1/2 toxicity: fatigue (7 patients), constipation (2), and nausea and vomiting (2); Grade 3 toxicity: fatigue (3), neutropenia (2), febrile neutropenia (1), diarrhea (2), and QTc prolongation (1). No patient experienced Grade 4 toxicities. CONCLUSIONS The addition of ATO to FOLFIRI regimen as second-line CT in patients with refractory mCRC offered an encouraging antitumor effect at the cost of manageable toxicity.
Collapse
Affiliation(s)
- K C Lakshmaiah
- Department of Medical Oncology, Kidwai Memorial Institute of Oncology, Bengaluru, Karnataka, India
| | - Tamojit Chaudhuri
- Department of Medical Oncology, Kidwai Memorial Institute of Oncology, Bengaluru, Karnataka, India
| | - Govind K Babu
- Department of Medical Oncology, Kidwai Memorial Institute of Oncology, Bengaluru, Karnataka, India
| | - Dasappa Lokanatha
- Department of Medical Oncology, Kidwai Memorial Institute of Oncology, Bengaluru, Karnataka, India
| | - Linu Abraham Jacob
- Department of Medical Oncology, Kidwai Memorial Institute of Oncology, Bengaluru, Karnataka, India
| | - M C Suresh Babu
- Department of Medical Oncology, Kidwai Memorial Institute of Oncology, Bengaluru, Karnataka, India
| | - A H Rudresha
- Department of Medical Oncology, Kidwai Memorial Institute of Oncology, Bengaluru, Karnataka, India
| | - K N Lokesh
- Department of Medical Oncology, Kidwai Memorial Institute of Oncology, Bengaluru, Karnataka, India
| | - L K Rajeev
- Department of Medical Oncology, Kidwai Memorial Institute of Oncology, Bengaluru, Karnataka, India
| |
Collapse
|
11
|
Huang P, Zhang YH, Zheng XW, Liu YJ, Zhang H, Fang L, Zhang YW, Yang C, Islam K, Wang C, Naranmandura H. Phenylarsine oxide (PAO) induces apoptosis in HepG2 cells via ROS-mediated mitochondria and ER-stress dependent signaling pathways. Metallomics 2017; 9:1756-1764. [PMID: 28831476 DOI: 10.1039/c7mt00179g] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Arsenic trioxide (As2O3) is an old drug that has recently been reintroduced as a therapeutic agent for acute promyelocytic leukemia (APL). Although As2O3 is also applied to treat other types of cancer in vitro and in vivo, it has been reported that single agent As2O3 has poor efficacy against non-hematologic malignant cancers in clinical trials. Recently, a few reports have indicated that organic arsenic compounds can be a possible alternative for the treatment of As2O3-resistant cancers. In this study, we aimed to investigate whether the organic arsenic compound phenylarsine oxide (PAO) has potent cytotoxic effects against human hepatocellular carcinoma (HCC) HepG2 cells. Our results showed that PAO not only had a potent inhibitory effect on the proliferation of HepG2 cells but also activated apoptosis-related proteins (e.g., caspase-3 and -9 and poly-ADP ribose polymerase) in a dose- and time-dependent manner. Furthermore, intracellular ROS were specifically accumulated in the mitochondria and endoplasmic reticulum (ER) after exposure to PAO, implying that they are the target organelles for PAO-induced cytotoxicity. Additionally, when the cells were pretreated with antioxidant N-acetylcysteine (NAC), apoptosis and ER-stress were attenuated significantly, suggesting that induction of apoptosis and cell death probably occurs through the ROS-mediated mitochondria and ER-stress dependent signaling pathways.
Collapse
Affiliation(s)
- Ping Huang
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Yu Hua Zhang
- Zhejiang Provincial People's Hospital, Hangzhou 310014, China
| | - Xiao Wei Zheng
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Yu Jia Liu
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Hong Zhang
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Luo Fang
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Yi Wen Zhang
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Chang Yang
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou, 310058, China and Department of Toxicology, School of Medicine and Public Health, Zhejiang University, Hangzhou, 310058, China.
| | - Khairul Islam
- Department of Toxicology, School of Medicine and Public Health, Zhejiang University, Hangzhou, 310058, China.
| | - Chao Wang
- Department of Toxicology, School of Medicine and Public Health, Zhejiang University, Hangzhou, 310058, China.
| | - Hua Naranmandura
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou, 310058, China and Department of Toxicology, School of Medicine and Public Health, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
12
|
Moghaddaskho F, Eyvani H, Ghadami M, Tavakkoly-Bazzaz J, Alimoghaddam K, Ghavamzadeh A, Ghaffari SH. Demethylation and alterations in the expression level of the cell cycle-related genes as possible mechanisms in arsenic trioxide-induced cell cycle arrest in human breast cancer cells. Tumour Biol 2017; 39:1010428317692255. [PMID: 28218039 DOI: 10.1177/1010428317692255] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Arsenic trioxide (As2O3) has been used clinically as an anti-tumor agent. Its mechanisms are mostly considered to be the induction of apoptosis and cell cycle arrest. However, the detailed molecular mechanisms of its anti-cancer action through cell cycle arrest are poorly known. Furthermore, As2O3 has been shown to be a potential DNA methylation inhibitor, inducing DNA hypomethylation. We hypothesize that As2O3 may affect the expression of cell cycle regulatory genes by interfering with DNA methylation patterns. To explore this, we examined promoter methylation status of 24 cell cycle genes in breast cancer cell lines and in a normal breast tissue sample by methylation-specific polymerase chain reaction and/or restriction enzyme-based methods. Gene expression level and cell cycle distribution were quantified by real-time polymerase chain reaction and flow cytometric analyses, respectively. Our methylation analysis indicates that only promoters of RBL1 (p107), RASSF1A, and cyclin D2 were aberrantly methylated in studied breast cancer cell lines. As2O3 induced CpG island demethylation in promoter regions of these genes and restores their expression correlated with DNA methyltransferase inhibition. As2O3 also induced alterations in messenger RNA expression of several cell cycle-related genes independent of demethylation. Flow cytometric analysis revealed that the cell cycle arrest induced by As2O3 varied depending on cell lines, MCF-7 at G1 phase and both MDA-MB-231 and MDA-MB-468 cells at G2/M phase. These changes at transcriptional level of the cell cycle genes by the molecular mechanisms dependent and independent of demethylation are likely to represent the mechanisms of cell cycle redistribution in breast cancer cells, in response to As2O3 treatment.
Collapse
Affiliation(s)
- Farima Moghaddaskho
- 1 Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.,2 Medical Genetics Department, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Haniyeh Eyvani
- 1 Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.,2 Medical Genetics Department, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Ghadami
- 2 Medical Genetics Department, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Javad Tavakkoly-Bazzaz
- 2 Medical Genetics Department, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kamran Alimoghaddam
- 1 Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Ardeshir Ghavamzadeh
- 1 Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed H Ghaffari
- 1 Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Arsenic Trioxide Induces T Cell Apoptosis and Prolongs Islet Allograft Survival in Mice. Transplantation 2015; 99:1796-806. [PMID: 25919768 DOI: 10.1097/tp.0000000000000735] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND T cell-mediated immune rejection is a key barrier to islet transplantation. Preliminary studies have shown that arsenic trioxide (As2O3) can inhibit T cell responses and prolong heart allograft survival. Here, we sought to investigate the possibility of using As2O3 to prolong islet allograft survival in an acute rejection model of Balb/c to C57B/6 mice. METHODS Recipient mice were treated with As2O3 and/or rapamycin after islet allograft transplantation. At day 10 after transplantation, the graft, spleen, lymph nodes, and blood of the recipient mice were recovered for analysis. In vitro, to further examine the mechanism underlying As2O3 protection of islet allografts against T cell-mediated rejection, mixed lymphocyte reaction and apoptosis analyses of T cells were performed. The phosphorylation levels of IκBα and p38 were also evaluated to confirm the proliferation and apoptosis of As2O3-treated T cells. RESULTS We found that As2O3 prolonged islet allograft survival by reducing inflammatory reactions, influencing cytokine synthesis and secretion and T-cell subset proportions, and inhibiting T-cell responses. Furthermore, As2O3 and rapamycin showed a synergistic effect in suppressing islet allotransplant rejection. CONCLUSIONS Arsenic trioxide may prevent allograft rejection by inhibiting T-cell proliferation and inducing T-cell apoptosis.
Collapse
|
14
|
Liu SH, Yang RS, Yen YP, Chiu CY, Tsai KS, Lan KC. Low-Concentration Arsenic Trioxide Inhibits Skeletal Myoblast Cell Proliferation via a Reactive Oxygen Species-Independent Pathway. PLoS One 2015; 10:e0137907. [PMID: 26359868 PMCID: PMC4567280 DOI: 10.1371/journal.pone.0137907] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 08/22/2015] [Indexed: 12/25/2022] Open
Abstract
Myoblast proliferation and differentiation are essential for skeletal muscle regeneration. Myoblast proliferation is a critical step in the growth and maintenance of skeletal muscle. The precise action of inorganic arsenic on myoblast growth has not been investigated. Here, we investigated the in vitro effect of inorganic arsenic trioxide (As2O3) on the growth of C2C12 myoblasts. As2O3 decreased myoblast growth at submicromolar concentrations (0.25–1 μM) after 72 h of treatment. Submicromolar concentrations of As2O3 did not induce the myoblast apoptosis. Low-concentration As2O3 (0.5 and 1 μM) significantly suppressed the myoblast cell proliferative activity, which was accompanied by a small proportion of bromodeoxyuridine (BrdU) incorporation and decreased proliferating cell nuclear antigen (PCNA) protein expression. As2O3 (0.5 and 1 μM) increased the intracellular arsenic content but did not affect the reactive oxygen species (ROS) levels in the myoblasts. Cell cycle analysis indicated that low-concentrations of As2O3 inhibited cell proliferation via cell cycle arrest in the G1 and G2/M phases. As2O3 also decreased the protein expressions of cyclin D1, cyclin E, cyclin B1, cyclin-dependent kinase (CDK) 2, and CDK4, but did not affect the protein expressions of p21 and p27. Furthermore, As2O3 inhibited the phosphorylation of Akt. Insulin-like growth factor-1 significantly reversed the inhibitory effect of As2O3 on Akt phosphorylation and cell proliferation in the myoblasts. These results suggest that submicromolar concentrations of As2O3 alter cell cycle progression and reduce myoblast proliferation, at least in part, through a ROS-independent Akt inhibition pathway.
Collapse
Affiliation(s)
- Shing Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Rong-Sen Yang
- Departments of Orthopaedic, College of Medicine and Hospital, National Taiwan University, Taipei, Taiwan
| | - Yuan-Peng Yen
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chen-Yuan Chiu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Keh-Sung Tsai
- Departments of Laboratory Medicine, College of Medicine and Hospital, National Taiwan University, Taipei, Taiwan
| | - Kuo-Cheng Lan
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
15
|
Gao JK, Wang LX, Long B, Ye XT, Su JN, Yin XY, Zhou XX, Wang ZW. Arsenic Trioxide Inhibits Cell Growth and Invasion via Down- Regulation of Skp2 in Pancreatic Cancer Cells. Asian Pac J Cancer Prev 2015; 16:3805-10. [PMID: 25987041 DOI: 10.7314/apjcp.2015.16.9.3805] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Arsenic trioxide (ATO) has been found to exert anti-cancer activity in various human malignancies. However, the molecular mechanisms by which ATO inhibits tumorigenesis are not fully elucidated. In the current study, we explored the molecular basis of ATO-mediated tumor growth inhibition in pancreatic cancer cells. We used multiple approaches such as MTT assay, wound healing assay, Transwell invasion assay, annexin V-FITC, cell cycle analysis, RT-PCR and Western blotting to achieve our goal. We found that ATO treatment effectively caused cell growth inhibition, suppressed clonogenic potential and induced G2-M cell cycle arrest and apoptosis in pancreatic cancer cells. Moreover, we observed a significant down-regulation of Skp2 after treatment with ATO. Furthermore, we revealed that ATO regulated Skp2 downstream genes such as FOXO1 and p53. These findings demonstrate that inhibition of Skp2 could be a novel strategy for the treatment of pancreatic cancer by ATO.
Collapse
Affiliation(s)
- Jian-Kun Gao
- Department of Basic Medical Sciences, Sichuan College of Traditional Chinese Medicine, Mianyang, Sichuan, China E-mail : ,
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Smith AH, Marshall G, Yuan Y, Steinmaus C, Liaw J, Smith MT, Wood L, Heirich M, Fritzemeier RM, Pegram MD, Ferreccio C. Rapid reduction in breast cancer mortality with inorganic arsenic in drinking water. EBioMedicine 2014; 1:58-63. [PMID: 25580451 PMCID: PMC4286879 DOI: 10.1016/j.ebiom.2014.10.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Arsenic trioxide is effective in treating promyelocytic leukemia, and laboratory studies demonstrate that arsenic trioxide causes apoptosis of human breast cancer cells. Region II in northern Chile experienced very high concentrations of inorganic arsenic in drinking water, especially in the main city Antofagasta from 1958 until an arsenic removal plant was installed in 1970. METHODS We investigated breast cancer mortality from 1950 to 2010 among women in Region II compared to Region V, which had low arsenic water concentrations. We conducted studies on human breast cancer cell lines and compared arsenic exposure in Antofagasta with concentrations inducing apoptosis in laboratory studies. FINDINGS Before 1958, breast cancer mortality rates were similar, but in 1958-1970 the rates in Region II were half those in Region V (rate ratio RR = 0·51, 95% CI 0·40-0·66; p<0·0001). Women under the age of 60 experienced a 70% reduction in breast cancer mortality during 1965-1970 (RR=0·30, 0·17-0·54; p<0·0001). Breast cancer cell culture studies showed apoptosis at arsenic concentrations close to those estimated to have occurred in people in Region II. INTERPRETATION We found biologically plausible major reductions in breast cancer mortality during high exposure to inorganic arsenic in drinking water which could not be attributed to bias or confounding. We recommend clinical trial assessment of inorganic arsenic in the treatment of advanced breast cancer.
Collapse
Affiliation(s)
- Allan H Smith
- Arsenic Research Group, School of Public Health, University of California, Berkeley, Berkeley, CA (Prof AH Smith MD, Y Yuan MPH, Assoc Prof C Steinmaus MD, J Liaw MPH); Departamento de Estadística, Facultad de Matemáticas, Pontificia Universidad Catòlica de Chile, Santiago, Chile (Prof G Marshall PhD); Environmental Health Sciences, School of Public Health, University of California, Berkeley, Berkeley, CA (Prof MT Smith PhD); Breast Cancer Oncology Program, Stanford Cancer Institute, Stanford, CA (L Wood, M Heirich, RM Fritzemeier MS, Prof MD Pegram MD); Departamento de Salud Pública, Escuela de Medicina, Pontificia Universidad Catòlica de Chile, and Advanced Center for Chronic Diseases ACCDIS, Santiago, Chile (Prof C Ferreccio MD)
| | - Guillermo Marshall
- Arsenic Research Group, School of Public Health, University of California, Berkeley, Berkeley, CA (Prof AH Smith MD, Y Yuan MPH, Assoc Prof C Steinmaus MD, J Liaw MPH); Departamento de Estadística, Facultad de Matemáticas, Pontificia Universidad Catòlica de Chile, Santiago, Chile (Prof G Marshall PhD); Environmental Health Sciences, School of Public Health, University of California, Berkeley, Berkeley, CA (Prof MT Smith PhD); Breast Cancer Oncology Program, Stanford Cancer Institute, Stanford, CA (L Wood, M Heirich, RM Fritzemeier MS, Prof MD Pegram MD); Departamento de Salud Pública, Escuela de Medicina, Pontificia Universidad Catòlica de Chile, and Advanced Center for Chronic Diseases ACCDIS, Santiago, Chile (Prof C Ferreccio MD)
| | - Yan Yuan
- Arsenic Research Group, School of Public Health, University of California, Berkeley, Berkeley, CA (Prof AH Smith MD, Y Yuan MPH, Assoc Prof C Steinmaus MD, J Liaw MPH); Departamento de Estadística, Facultad de Matemáticas, Pontificia Universidad Catòlica de Chile, Santiago, Chile (Prof G Marshall PhD); Environmental Health Sciences, School of Public Health, University of California, Berkeley, Berkeley, CA (Prof MT Smith PhD); Breast Cancer Oncology Program, Stanford Cancer Institute, Stanford, CA (L Wood, M Heirich, RM Fritzemeier MS, Prof MD Pegram MD); Departamento de Salud Pública, Escuela de Medicina, Pontificia Universidad Catòlica de Chile, and Advanced Center for Chronic Diseases ACCDIS, Santiago, Chile (Prof C Ferreccio MD)
| | - Craig Steinmaus
- Arsenic Research Group, School of Public Health, University of California, Berkeley, Berkeley, CA (Prof AH Smith MD, Y Yuan MPH, Assoc Prof C Steinmaus MD, J Liaw MPH); Departamento de Estadística, Facultad de Matemáticas, Pontificia Universidad Catòlica de Chile, Santiago, Chile (Prof G Marshall PhD); Environmental Health Sciences, School of Public Health, University of California, Berkeley, Berkeley, CA (Prof MT Smith PhD); Breast Cancer Oncology Program, Stanford Cancer Institute, Stanford, CA (L Wood, M Heirich, RM Fritzemeier MS, Prof MD Pegram MD); Departamento de Salud Pública, Escuela de Medicina, Pontificia Universidad Catòlica de Chile, and Advanced Center for Chronic Diseases ACCDIS, Santiago, Chile (Prof C Ferreccio MD)
| | - Jane Liaw
- Arsenic Research Group, School of Public Health, University of California, Berkeley, Berkeley, CA (Prof AH Smith MD, Y Yuan MPH, Assoc Prof C Steinmaus MD, J Liaw MPH); Departamento de Estadística, Facultad de Matemáticas, Pontificia Universidad Catòlica de Chile, Santiago, Chile (Prof G Marshall PhD); Environmental Health Sciences, School of Public Health, University of California, Berkeley, Berkeley, CA (Prof MT Smith PhD); Breast Cancer Oncology Program, Stanford Cancer Institute, Stanford, CA (L Wood, M Heirich, RM Fritzemeier MS, Prof MD Pegram MD); Departamento de Salud Pública, Escuela de Medicina, Pontificia Universidad Catòlica de Chile, and Advanced Center for Chronic Diseases ACCDIS, Santiago, Chile (Prof C Ferreccio MD)
| | - Martyn T Smith
- Arsenic Research Group, School of Public Health, University of California, Berkeley, Berkeley, CA (Prof AH Smith MD, Y Yuan MPH, Assoc Prof C Steinmaus MD, J Liaw MPH); Departamento de Estadística, Facultad de Matemáticas, Pontificia Universidad Catòlica de Chile, Santiago, Chile (Prof G Marshall PhD); Environmental Health Sciences, School of Public Health, University of California, Berkeley, Berkeley, CA (Prof MT Smith PhD); Breast Cancer Oncology Program, Stanford Cancer Institute, Stanford, CA (L Wood, M Heirich, RM Fritzemeier MS, Prof MD Pegram MD); Departamento de Salud Pública, Escuela de Medicina, Pontificia Universidad Catòlica de Chile, and Advanced Center for Chronic Diseases ACCDIS, Santiago, Chile (Prof C Ferreccio MD)
| | - Lily Wood
- Arsenic Research Group, School of Public Health, University of California, Berkeley, Berkeley, CA (Prof AH Smith MD, Y Yuan MPH, Assoc Prof C Steinmaus MD, J Liaw MPH); Departamento de Estadística, Facultad de Matemáticas, Pontificia Universidad Catòlica de Chile, Santiago, Chile (Prof G Marshall PhD); Environmental Health Sciences, School of Public Health, University of California, Berkeley, Berkeley, CA (Prof MT Smith PhD); Breast Cancer Oncology Program, Stanford Cancer Institute, Stanford, CA (L Wood, M Heirich, RM Fritzemeier MS, Prof MD Pegram MD); Departamento de Salud Pública, Escuela de Medicina, Pontificia Universidad Catòlica de Chile, and Advanced Center for Chronic Diseases ACCDIS, Santiago, Chile (Prof C Ferreccio MD)
| | - Marissa Heirich
- Arsenic Research Group, School of Public Health, University of California, Berkeley, Berkeley, CA (Prof AH Smith MD, Y Yuan MPH, Assoc Prof C Steinmaus MD, J Liaw MPH); Departamento de Estadística, Facultad de Matemáticas, Pontificia Universidad Catòlica de Chile, Santiago, Chile (Prof G Marshall PhD); Environmental Health Sciences, School of Public Health, University of California, Berkeley, Berkeley, CA (Prof MT Smith PhD); Breast Cancer Oncology Program, Stanford Cancer Institute, Stanford, CA (L Wood, M Heirich, RM Fritzemeier MS, Prof MD Pegram MD); Departamento de Salud Pública, Escuela de Medicina, Pontificia Universidad Catòlica de Chile, and Advanced Center for Chronic Diseases ACCDIS, Santiago, Chile (Prof C Ferreccio MD)
| | - Rebecca M Fritzemeier
- Arsenic Research Group, School of Public Health, University of California, Berkeley, Berkeley, CA (Prof AH Smith MD, Y Yuan MPH, Assoc Prof C Steinmaus MD, J Liaw MPH); Departamento de Estadística, Facultad de Matemáticas, Pontificia Universidad Catòlica de Chile, Santiago, Chile (Prof G Marshall PhD); Environmental Health Sciences, School of Public Health, University of California, Berkeley, Berkeley, CA (Prof MT Smith PhD); Breast Cancer Oncology Program, Stanford Cancer Institute, Stanford, CA (L Wood, M Heirich, RM Fritzemeier MS, Prof MD Pegram MD); Departamento de Salud Pública, Escuela de Medicina, Pontificia Universidad Catòlica de Chile, and Advanced Center for Chronic Diseases ACCDIS, Santiago, Chile (Prof C Ferreccio MD)
| | - Mark D Pegram
- Arsenic Research Group, School of Public Health, University of California, Berkeley, Berkeley, CA (Prof AH Smith MD, Y Yuan MPH, Assoc Prof C Steinmaus MD, J Liaw MPH); Departamento de Estadística, Facultad de Matemáticas, Pontificia Universidad Catòlica de Chile, Santiago, Chile (Prof G Marshall PhD); Environmental Health Sciences, School of Public Health, University of California, Berkeley, Berkeley, CA (Prof MT Smith PhD); Breast Cancer Oncology Program, Stanford Cancer Institute, Stanford, CA (L Wood, M Heirich, RM Fritzemeier MS, Prof MD Pegram MD); Departamento de Salud Pública, Escuela de Medicina, Pontificia Universidad Catòlica de Chile, and Advanced Center for Chronic Diseases ACCDIS, Santiago, Chile (Prof C Ferreccio MD)
| | - Catterina Ferreccio
- Arsenic Research Group, School of Public Health, University of California, Berkeley, Berkeley, CA (Prof AH Smith MD, Y Yuan MPH, Assoc Prof C Steinmaus MD, J Liaw MPH); Departamento de Estadística, Facultad de Matemáticas, Pontificia Universidad Catòlica de Chile, Santiago, Chile (Prof G Marshall PhD); Environmental Health Sciences, School of Public Health, University of California, Berkeley, Berkeley, CA (Prof MT Smith PhD); Breast Cancer Oncology Program, Stanford Cancer Institute, Stanford, CA (L Wood, M Heirich, RM Fritzemeier MS, Prof MD Pegram MD); Departamento de Salud Pública, Escuela de Medicina, Pontificia Universidad Catòlica de Chile, and Advanced Center for Chronic Diseases ACCDIS, Santiago, Chile (Prof C Ferreccio MD)
| |
Collapse
|
17
|
Zhang Q, Vakili MR, Li XF, Lavasanifar A, Le XC. Polymeric micelles for GSH-triggered delivery of arsenic species to cancer cells. Biomaterials 2014; 35:7088-100. [DOI: 10.1016/j.biomaterials.2014.04.072] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 04/17/2014] [Indexed: 11/25/2022]
|
18
|
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: 2.8] [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
|
19
|
Arsenic trioxide induces cervical cancer apoptosis, but specifically targets human papillomavirus-infected cell populations. Anticancer Drugs 2013; 23:280-7. [PMID: 22245994 DOI: 10.1097/cad.0b013e32834f1fd3] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES Human papillomavirus (HPV) is directly associated with the occurrence and development of cervical cancer. Targeting HPV infection has become the priority in treatment and prevention. Some treatment strategies have shown a limited therapeutic potential in suppressing and reversing the oncogenic effects of HPVs, but are compromised by the toxicity, immune suppression and the expense. Arsenic trioxide (As2O3) has shown therapeutic efficacy in the treatment of haematological and solid cancer and has been demonstrated to effectively induce apoptosis of HPV-infected cervical cancer cells in vitro. Here, we examined the effects and possible molecular pathway of As2O3-induced apoptosis in HPV-infected and noninfected cervical cancer cells. METHODS As2O3 was added to HPV-infected cell lines HeLa and CaSki and the HPV-negative cell line C33A at concentrations from 1 to 10 µmol/l. Cell proliferation rates were evaluated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays after exposure. Expressions of tumour suppressor gene p53, activated caspase-3 and cell cycle distribution were evaluated in relation to HPV-E6 protein expression by confocal microscopy immunofluorescent staining and flow cytometry. RESULTS As2O3 reduced cell populations by 16% in C33A but by 48-60% in HPV-infected cell lines CaSki and HeLa. The expression of HPV-E6 proteins was drastically down-regulated in a dose-dependent manner, whereas p53 and activated caspase-3 expressions increased in the HPV-infected cell lines. Flow cytometry demonstrated cell cycle arrest in S-G2/M phases, and increasing apoptotic bodies were seen in HPV-infected lines only. CONCLUSION As2O3, at low concentrations, inhibited HPV-E6 protein expression, leading to up-regulated p53 levels, induced S to G2/M arrest and apoptosis.
Collapse
|
20
|
Suppression of pancreatic tumor growth by targeted arsenic delivery with anti-CD44v6 single chain antibody conjugated nanoparticles. Biomaterials 2013; 34:6175-84. [DOI: 10.1016/j.biomaterials.2013.04.056] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Accepted: 04/27/2013] [Indexed: 01/09/2023]
|
21
|
Wu CC, D'Argenio D, Asgharzadeh S, Triche T. TARGETgene: a tool for identification of potential therapeutic targets in cancer. PLoS One 2012; 7:e43305. [PMID: 22952662 PMCID: PMC3432038 DOI: 10.1371/journal.pone.0043305] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Accepted: 07/18/2012] [Indexed: 11/19/2022] Open
Abstract
The vast array of in silico resources and data of high throughput profiling currently available in life sciences research offer the possibility of aiding cancer gene and drug discovery process. Here we propose to take advantage of these resources to develop a tool, TARGETgene, for efficiently identifying mutation drivers, possible therapeutic targets, and drug candidates in cancer. The simple graphical user interface enables rapid, intuitive mapping and analysis at the systems level. Users can find, select, and explore identified target genes and compounds of interest (e.g., novel cancer genes and their enriched biological processes), and validate predictions using user-defined benchmark genes (e.g., target genes detected in RNAi screens) and curated cancer genes via TARGETgene. The high-level capabilities of TARGETgene are also demonstrated through two applications in this paper. The predictions in these two applications were then satisfactorily validated by several ways, including known cancer genes, results of RNAi screens, gene function annotations, and target genes of drugs that have been used or in clinical trial in cancer treatments. TARGETgene is freely available from the Biomedical Simulations Resource web site (http://bmsr.usc.edu/Software/TARGET/TARGET.html).
Collapse
Affiliation(s)
- Chia-Chin Wu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America.
| | | | | | | |
Collapse
|
22
|
Moskalev AA, Smit-McBride Z, Shaposhnikov MV, Plyusnina EN, Zhavoronkov A, Budovsky A, Tacutu R, Fraifeld VE. Gadd45 proteins: relevance to aging, longevity and age-related pathologies. Ageing Res Rev 2012; 11:51-66. [PMID: 21986581 PMCID: PMC3765067 DOI: 10.1016/j.arr.2011.09.003] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Revised: 09/25/2011] [Accepted: 09/27/2011] [Indexed: 12/12/2022]
Abstract
The Gadd45 proteins have been intensively studied, in view of their important role in key cellular processes. Indeed, the Gadd45 proteins stand at the crossroad of the cell fates by controlling the balance between cell (DNA) repair, eliminating (apoptosis) or preventing the expansion of potentially dangerous cells (cell cycle arrest, cellular senescence), and maintaining the stem cell pool. However, the biogerontological aspects have not thus far received sufficient attention. Here we analyzed the pathways and modes of action by which Gadd45 members are involved in aging, longevity and age-related diseases. Because of their pleiotropic action, a decreased inducibility of Gadd45 members may have far-reaching consequences including genome instability, accumulation of DNA damage, and disorders in cellular homeostasis - all of which may eventually contribute to the aging process and age-related disorders (promotion of tumorigenesis, immune disorders, insulin resistance and reduced responsiveness to stress). Most recently, the dGadd45 gene has been identified as a longevity regulator in Drosophila. Although further wide-scale research is warranted, it is becoming increasingly clear that Gadd45s are highly relevant to aging, age-related diseases (ARDs) and to the control of life span, suggesting them as potential therapeutic targets in ARDs and pro-longevity interventions.
Collapse
Affiliation(s)
- Alexey A Moskalev
- Group of Molecular Radiobiology and Gerontology, Institute of Biology, Komi Science Center of Russian Academy of Sciences.
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Research Spotlight: Iron chelation: deciphering novel molecular targets for cancer therapy. The tip of the iceberg of a web of iron-regulated molecules. Future Med Chem 2011; 3:1983-6. [DOI: 10.4155/fmc.11.154] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The response of cells to cellular iron depletion is complex with multiple molecules and signaling pathways being involved. Indeed, this is far broader than just the effect on the classical target, ribonucleotide reductase. It is likely that a network of interactions exists between the molecular players and that the relationships currently known only represent the ‘tip of an iceberg’ in terms of understanding the response of cells to iron deprivation. This article describes some of the research being undertaken in this area by the Richardson group at the University of Sydney, Australia.
Collapse
|
24
|
Emdad L, Qadeer ZA, Bederson LB, Kothari HP, Uzzaman M, Germano IM. Is there a common upstream link for autophagic and apoptotic cell death in human high-grade gliomas? Neuro Oncol 2011; 13:725-35. [PMID: 21727211 DOI: 10.1093/neuonc/nor053] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The prognosis of patients with human high-grade gliomas (HGGs) remains dismal despite major advances in their management, due mainly to the high resistance of these infiltrative tumor cells to programmed cell death (PCD). Most therapeutic strategies for HGGs are aimed to maximize PCD type I, apoptosis or type II, autophagy. These are predominantly distinctive processes, but many studies suggest a cross-talk between the two. A better understanding of the link between PCD types I and II might allow development of more effective therapies for HGGs. In this study, we examined whether there is a common upstream signaling event responsible for both apoptotic and autophagic PCD using 3 chemotherapeutic agents in human HGG cells. Our study shows that each agent caused a significant decrease in cell viability in each of the HGG cell lines tested. The increase rate of apoptosis and autophagy varied among cell lines and chemotherapeutic agents used. Increased expression of cytidine-cytidine-adenosine-adenosine-thymidine (C)/enhancer binding protein (EBP) homologous transcription factor C/EBP homologous protein (CHOP)/growth arrest and DNA damage-inducible gene 153 (GADD153) was documented after use of either pro-autophagic or pro-apoptotic agents. The involvement of CHOP/GADD153 in both type I and type II PCD was confirmed by overexpression and gene-silencing studies. Gene silencing by small-interfering RNA-mediated CHOP/GADD153 resulted in increased cell viability, decreased upregulation of microtubule-associated protein light-chain 3' type II (LC3II) and cleaved caspase-3, and inhibition of apoptosis and autophagy. Exogenous expression of CHOP/GADD153 triggered apoptosis and autophagy in the absence of other stimuli. The clinical significance of these findings was supported by the evidence that celecoxib, a nonsteroidal anti-inflammatory drug known to induce GADD153-mediated apoptosis, strongly increases both type I and type II PCD in HGG cells when combined with another inducer of GADD153. These data suggest that CHOP/GADD153 should be investigated as a novel targetable signaling step to improve therapies for HGGs.
Collapse
Affiliation(s)
- Luni Emdad
- Department of Neurosurgery, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | | | | | | | |
Collapse
|
25
|
Li G, He S, Chang L, Lu H, Zhang H, Zhang H, Chiu J. GADD45α and annexin A1 are involved in the apoptosis of HL-60 induced by resveratrol. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2011; 18:704-9. [PMID: 21277758 DOI: 10.1016/j.phymed.2010.11.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2010] [Revised: 10/15/2010] [Accepted: 11/29/2010] [Indexed: 02/05/2023]
Abstract
Resveratrol (3,4',5-trihydroxy-trans-stilbene), one of secondary metabolites of low molecular weight present in plant, has various important biological effects. It can induce apoptosis in human leukemia cell types in vitro, although the mechanism is not fully understood. In the present study, we demonstrated reduced viability and DNA synthesis, as well as increased proportion of the subdiploid cell population, in HL-60 cells as determined by cell cycle analysis with resveratrol. Resveratrol treatment resulted in a gradual time-dependent decrease in the expression of anti-apoptotic Bcl-2 and increase in that of Bax, annexin A1, growth arrest- and DNA damage-induced gene 45α (GADD45α), and cleaved caspase-3. In addition, resveratrol markedly increased caspase-3 activity in cells. Our results suggest that resveratrol could inhibit the proliferation and induce apoptosis of HL-60 cells through a GADD45α and annexin A1/caspase-3 pathway.
Collapse
Affiliation(s)
- Guanwu Li
- Open Laboratory for Tumor Molecular Biology/Department of Biochemistry, Shantou University Medical College, Shantou, China
| | | | | | | | | | | | | |
Collapse
|
26
|
Podolsky L, Oh M, Subbarayan PR, Francheschi D, Livingstone A, Ardalan B. 5-Fluorouracil/Leucovorin and arsenic trioxide for patients with refractory/relapsed colorectal carcinoma: a clinical experience. Acta Oncol 2011; 50:602-5. [PMID: 20950119 DOI: 10.3109/0284186x.2010.524934] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
27
|
Wang Y, Zhang Y, Yang L, Cai B, Li J, Zhou Y, Yin L, Yang L, Yang BF, Lu YJ. Arsenic trioxide induces the apoptosis of human breast cancer MCF-7 cells through activation of caspase-3 and inhibition of HERG channels. Exp Ther Med 2011; 2:481-486. [PMID: 22977528 DOI: 10.3892/etm.2011.224] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2010] [Accepted: 02/28/2011] [Indexed: 01/27/2023] Open
Abstract
Arsenic trioxide (As(2)O(3)) has been widely used to treat patients with acute promyelocytic leukemia and has also been shown to exhibit therapeutic effects on various types of solid tumors, including gastric cancer and lung carcinoma. Breast cancer is a type of solid tumor whose incidence has been increasing for many years. The present study was designed to investigate the effects of As(2)O(3) on the human breast cancer cell line MCF-7, and to explore its potential mechanisms. The MTT assay demonstrated that As(2)O(3) decreased the cellular viability of MCF-7 cells in a concentration-dependent manner. Morphological observation, the TUNEL assay and flow cytometric analysis revealed that apoptosis was involved in the process. An assay for caspase-3 activity suggested that the apoptosis was mediated through caspase-3 activation. Further investigation indicated that protein levels of the human ether-a-go-go-related gene (HERG) were markedly downregulated by As(2)O(3). Taken together, the results indicate that arsenic trioxide induces the apoptosis of human breast cancer MCF-7 cells at least in part through the activation of caspase-3 and the decrease in HERG expression.
Collapse
Affiliation(s)
- Ying Wang
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine and Pharmaceutics, and
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Gupta S, Sathishkumar S, Ahmed MM. Influence of cell cycle checkpoints and p53 function on the toxicity of temozolomide in human pancreatic cancer cells. Pancreatology 2010; 10:565-79. [PMID: 20980775 PMCID: PMC2992636 DOI: 10.1159/000317254] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2009] [Accepted: 06/06/2010] [Indexed: 12/11/2022]
Abstract
BACKGROUND Though an increased efficacy of carmustine and temozolomide (TMZ) has been demonstrated by inactivation of O(6)-methylguanine-DNA methyltransferase (MGMT) with O(6)-benzyl-guanine (BG) in human pancreatic tumors refractive to alkylating agents, the regulatory mechanisms have not been explored. METHODS The effects of TMZ and BG on apoptosis, cell growth, the mitotic index, cell cycle distribution, and protein expression were studied by TUNEL, cell counting, flow cytometry, and Western blot analysis, respectively. RESULTS The wt-p53 human pancreatic tumor cell line Capan-2 and p53-efficient mouse embryonic fibroblasts (MEFs) were more responsive to treatment with TMZ + BG than mutant p53 Capan-1 and p53-null MEFs. S phase delay with a subsequent G2/M arrest was observed in Capans in response to BG + TMZ. The G1-to-S transition delay in Capan-2 was associated with p53-dependent apoptosis and was distinctly different from the presumed mismatch repair (MMR) killing operative during the G2/M arrest. The effect of p53 on BG + TMZ toxicity was supported by a marked change in apoptosis when p53 function was restored/inactivated. There was an early induction of MMR proteins in p53-efficient lines. CONCLUSION p53 provokes a classic proapoptotic response by delaying G1-to-S progression, but it may also facilitate cell killing by enhancing MMR-related cell cycle arrest and cell death.
Collapse
Affiliation(s)
- Seema Gupta
- Department of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, Fla., USA,Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Fla., USA
| | | | - Mansoor M. Ahmed
- Department of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, Fla., USA,Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Fla., USA,*Mansoor M. Ahmed, PhD, Department of Radiation Oncology, 1475 NW 12th Ave (D-31) Miami, FL 33136 (USA), Tel. +1 305 243 5454, Fax +1 305 243 1854, E-Mail
| |
Collapse
|
29
|
Walker AM, Stevens JJ, Ndebele K, Tchounwou PB. Arsenic trioxide modulates DNA synthesis and apoptosis in lung carcinoma cells. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2010; 7:1996-2007. [PMID: 20632473 PMCID: PMC2864039 DOI: 10.3390/ijerph7051996] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Arsenic trioxide, the trade name Trisenox, is a drug used to treat acute promyleocytic leukemia (APL). Studies have demonstrated that arsenic trioxide slows cancer cells growth. Although arsenic influences numerous signal-transduction pathways, cell-cycle progression, and/or apoptosis, its apoptotic mechanisms are complex and not entirely delineated. The primary objective of this research was to evaluate the effects of arsenic trioxide on DNA synthesis and to determine whether arsenic-induced apoptosis is mediated via caspase activation, p38 mitogen-activated protein kinase (MAPK), and cell cycle arrest. To achieve this goal, lung cancer cells (A549) were exposed to various concentrations (0, 2, 4, 6, 8, and 10 microg/mL) of arsenic trioxide for 48 h. The effect of arsenic trioxide on DNA synthesis was determined by the [3H]thymidine incorporation assay. Apoptosis was determined by the caspase-3 fluorescein isothiocyanate (FITC) assay, p38 MAP kinase activity was determined by an immunoblot assay, and cell-cycle analysis was evaluated by the propidium iodide assay. The [3H]thymidine-incorporation assay revealed a dose-related cytotoxic response at high levels of exposure. Furthermore, arsenic trioxide modulated caspase 3 activity and induced p38 MAP kinase activation in A549 cells. However, cell-cycle studies showed no statistically significant differences in DNA content at subG1 check point between control and arsenic trioxide treated cells.
Collapse
Affiliation(s)
- Alice M. Walker
- Molecular Toxicology Research Laboratory, NIH RCMI-Center for Environmental Health, College of Science, Engineering and Technology, Jackson State University, 1400 JR Lynch Street, Box 18540, Jackson, MS 39217, USA; E-Mail:
- Molecular and Cellular Biology Research Laboratory, NIH RCMI-Center for Environmental Health, College of Science, Engineering and Technology, Jackson State University, 1400 JR Lynch Street, Box 18540, Jackson, MS 39217, USA; E-Mail:
| | - Jacqueline J. Stevens
- Molecular and Cellular Biology Research Laboratory, NIH RCMI-Center for Environmental Health, College of Science, Engineering and Technology, Jackson State University, 1400 JR Lynch Street, Box 18540, Jackson, MS 39217, USA; E-Mail:
| | - Kenneth Ndebele
- Laboratory of Cancer Immunology: Target Identification and Validation, College of Science, Engineering and Technology, Jackson State University, 1400 JR Lynch Street, Box 18540, Jackson, MS 39217, USA; E-Mail:
| | - Paul B. Tchounwou
- Molecular Toxicology Research Laboratory, NIH RCMI-Center for Environmental Health, College of Science, Engineering and Technology, Jackson State University, 1400 JR Lynch Street, Box 18540, Jackson, MS 39217, USA; E-Mail:
- Author to whom correspondence should be addressed; E-Mail:
; Tel.: +1-601-979-0777; Fax: +1-601-979-0570
| |
Collapse
|
30
|
p53 Activates Either Survival or Apoptotic Signaling Responses in Lupulone-Treated Human Colon Adenocarcinoma Cells and Derived Metastatic Cells. Transl Oncol 2010; 3:286-92. [PMID: 20885891 DOI: 10.1593/tlo.10124] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2010] [Revised: 05/24/2010] [Accepted: 06/02/2010] [Indexed: 12/31/2022] Open
Abstract
The SW480 cell line is derived from a human colon adenocarcinoma, and SW620 cells are derived from a lymph node metastasis of the same patient. We have previously shown that lupulone induces apoptosis in SW480 cells, through a cross talk between the TRAIL-death receptor pathway and the mitochondrial apoptotic pathway. In SW620 cells, lupulone induced apoptosis only through TRAIL-death receptor activation. Both cell lines exhibit the same p53 mutations. Because p53 plays a central role in the response to cellular stresses by upregulating the transcription of several genes controlling apoptosis, we aimed to study the involvement of p53 on lupulone-triggered apoptosis. Our data show that in SW620 cells, lupulone upregulated p53 gene expression and caused a cloistering of p53 in the nucleus, allowing p53 to play a proapoptotic role by activating the TRAIL-death receptor pathway. In contrast, in lupulone-treated SW480 cells, p53 was translocated to the cytoplasm where it initiated a survival response associated with the up-regulation of antiapoptotic Bcl-2 and Mcl-1 proteins in an attempt to preserve mitochondrial integrity. These prosurvival effects of p53 in lupulone-treated SW480 cells were reversed by pifithrin-α, an inhibitor of p53 function, which caused a blocking of p53 in the nucleus leading to the down-regulation of Bcl-2 and Mcl-1, the up-regulation of proapoptotic Bax protein and TRAIL-death receptors leading to enhanced cell death. Our data support different functions of the same mutated p53 in colon adenocarcinoma and derived metastatic cells in response to the chemopreventive agent lupulone.
Collapse
|
31
|
Up-regulation of GADD45alpha expression by NSAIDs leads to apoptotic and necrotic colon cancer cell deaths. Apoptosis 2010; 14:1341-51. [PMID: 19757064 DOI: 10.1007/s10495-009-0399-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Growth arrest and DNA damage inducible 45 alpha (GADD45alpha) is a central player in mediating apoptosis induced by a variety of stress stimuli and genotoxic agents. Regular usage of nonselective nonsteroidal anti-inflammatory drugs (NSAIDs) such as indomethacin and sulindac is associated with reduced risk for various cancers, including colon cancer. The role of GADD45alpha in NSAID-induced colon cancer cell cytotoxicity is unknown. In this study, we report that indomethacin and sulindac sulfide treatments up-regulate GADD45alpha mRNA expression and protein levels in colon cancer HT-29, RKO and Caco-2 cells. This up-regulation of GADD45alpha is accompanied by necrotic cell death and apoptosis. Anti-sense suppression of GADD45alpha expression inhibited indomethacin and sulindac sulfide-induced necrotic cell death and apoptosis. These findings confirm a role for GADD45alpha in NSAID-induced cytotoxicity, a mechanism for the anti-neoplastic effect of NSAIDs in colon tumorigenesis and cancer growth.
Collapse
|
32
|
Qu GP, Xiu QY, Li B, Liu YA, Zhang LZ. Arsenic trioxide inhibits the growth of human lung cancer cell lines via cell cycle arrest and induction of apoptosis at both normoxia and hypoxia. Toxicol Ind Health 2009; 25:505-15. [PMID: 19825857 DOI: 10.1177/0748233709345936] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Arsenic trioxide (As(2)O(3)) has been established to be an effective agent for treating acute promyleocytic leukemia. Laboratory data suggest that As(2)O(3) induces apoptosis of several solid tumor cells including lung cancer cells. Regions of tissue hypoxia often arise in aggressive solid tumors, and hypoxic tumors exhibit augmented invasiveness and metastatic ability in several malignancies. Furthermore, hypoxia may impair the treatment efficiency; therefore, we studied the cytotoxic effect of As(2)O(3) on human lung adenocarcinoma cell lines A549 and A549/R (resistant to vincristine, adriamycin and mitomycin etc.) grown under normoxic and hypoxic (1% oxygen) conditions. At both normoxia and hypoxia, 5, 10 and 15 microM As(2)O(3) induced evident growth inhibition and apoptosis in A549 cells as well as A549/R cells after 48 hours of exposure. In contrast, the conventional chemotherapeutic drug vincristine showed lowered efficiency in hypoxic A549 cells. As(2)O(3) induced G(2)/M cell cycle arrest in both normoxic and hypoxic A549 cells. As(2)O(3) significantly decreased the messenger RNA (mRNA) levels of Cyclin B(1) and survivin and the protein levels of Cyclin B(1), phospho-CDC(2) (Thr 161) and survivin in both normoxic and hypoxic A549 cells. Together, our findings indicated that As(2)O(3) significantly inhibited the proliferation of lung cancer cells via G(2)/M cell cycle arrest and induction of apoptosis at both normoxia and hypoxia, and the induction of apoptosis was associated with down regulation of survivin.
Collapse
Affiliation(s)
- Ge-ping Qu
- Department of Respiratory Diseases, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | | | | | | | | |
Collapse
|
33
|
Abstract
Arsenic is a metalloid that generates various biological effects on cells and tissues. Depending on the specific tissue exposed and the time and degree of exposure, diverse responses can be observed. In humans, prolonged and/or high dose exposure to arsenic can have a variety of outcomes, including the development of malignancies, severe gastrointestinal toxicities, diabetes, cardiac arrhythmias, and death. On the other hand, one arsenic derivative, arsenic trioxide (As(2)O(3)), has important antitumor properties. This agent is a potent inducer of antileukemic responses, and it is now approved by the Food and Drug Administration for the treatment of acute promyelocytic leukemia in humans. The promise and therapeutic potential of arsenic and its various derivatives have been exploited for hundreds of years. Remarkably, research focused on the potential use of arsenic compounds in the treatment of human diseases remains highly promising, and it is an area of active investigation. An emerging approach of interest and therapeutic potential involves efforts to target and block cellular pathways activated in a negative feedback manner during treatment of cells with As(2)O(3). Such an approach may ultimately provide the means to selectively enhance the suppressive effects of this agent on malignant cells and render normally resistant tumors sensitive to its antineoplastic properties.
Collapse
Affiliation(s)
- Leonidas C Platanias
- Robert H. Lurie Comprehensive Cancer Center and Division of Hematology-Oncology, Northwestern University Medical School, Chicago, Illinois 60611, USA.
| |
Collapse
|
34
|
Payne CM, Bernstein C, Dvorak K, Bernstein H. Hydrophobic bile acids, genomic instability, Darwinian selection, and colon carcinogenesis. Clin Exp Gastroenterol 2008; 1:19-47. [PMID: 21677822 PMCID: PMC3108627 DOI: 10.2147/ceg.s4343] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Sporadic colon cancer is caused predominantly by dietary factors. We have selected bile acids as a focus of this review since high levels of hydrophobic bile acids accompany a Western-style diet, and play a key role in colon carcinogenesis. We describe how bile acid-induced stresses cause cell death in susceptible cells, contribute to genomic instability in surviving cells, impose Darwinian selection on survivors and enhance initiation and progression to colon cancer. The most likely major mechanisms by which hydrophobic bile acids induce stresses on cells (DNA damage, endoplasmic reticulum stress, mitochondrial damage) are described. Persistent exposure of colon epithelial cells to hydrophobic bile acids can result in the activation of pro-survival stress-response pathways, and the modulation of numerous genes/proteins associated with chromosome maintenance and mitosis. The multiple mechanisms by which hydrophobic bile acids contribute to genomic instability are discussed, and include oxidative DNA damage, p53 and other mutations, micronuclei formation and aneuploidy. Since bile acids and oxidative stress decrease DNA repair proteins, an increase in DNA damage and increased genomic instability through this mechanism is also described. This review provides a mechanistic explanation for the important link between a Western-style diet and associated increased levels of colon cancer.
Collapse
Affiliation(s)
- Claire M Payne
- Department of Cell Biology and Anatomy, College of Medicine, University of Arizona, Tucson, Arizona, USA
| | | | | | | |
Collapse
|
35
|
Bae-Jump VL, Chunxiao Zhou, Boggess JF, Gehrig PA. Arsenic Trioxide (As2O3) Inhibits Expression of Estrogen Receptor—alpha Through Regulation of the Mitogen-activated Protein Kinase (MAPK) Pathway in Endometrial Cancer Cells. Reprod Sci 2008; 15:1011-7. [DOI: 10.1177/1933719108324134] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Victoria L. Bae-Jump
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina, Chapel Hill, North Carolina,
| | - Chunxiao Zhou
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina, Chapel Hill, North Carolina
| | - John F. Boggess
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina, Chapel Hill, North Carolina
| | - Paola A. Gehrig
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
36
|
Sodium arsenite induces ROS generation, DNA oxidative damage, HO-1 and c-Myc proteins, NF-kappaB activation and cell proliferation in human breast cancer MCF-7 cells. Mutat Res 2008; 674:109-15. [PMID: 18996220 DOI: 10.1016/j.mrgentox.2008.09.021] [Citation(s) in RCA: 146] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2008] [Accepted: 09/29/2008] [Indexed: 12/11/2022]
Abstract
Epidemiological evidence has associated exposure to arsenic (As) in drinking water with an increased incidence of human cancers in the skin, bladder, liver, kidney and lung. Sodium arsenite mimics the effects of estradiol and induces cell proliferation in the estrogen responsive breast cancer cell line MCF-7. Therefore, our aim was to further explore the ability of sodium arsenite to induce MCF-7 epithelial breast cell proliferation and some of its underlying mechanisms by studying ROS production, c-Myc and HO-1 protein levels, 8-OHdG formation and NF-kappaB activation. Low arsenite concentrations (0.5-5 microM) induced ROS production and ROS-related depolarization of the mitochondrial membrane suggesting that mitochondria played an important role in the oxidative effects of As. ROS-mediated DNA damage as measured by the presence of 8-OHdG DNA-adducts in their nuclei, IkappaB phosphorylation, NF-kappaB activation and increases in c-Myc and HO-1 protein levels were also observed, suggesting that these factors play a relevant role in the arsenite induced MCF-7 cell recruitment into the S-phase of the cell cycle and cell proliferation observed. In conclusion, arsenite activates several pathways involved in MCF-7 cell proliferation suggesting that arsenite exposure may pose a risk for breast cancer in human exposed populations notwithstanding that most studies to date have not yet implicated this metalloid as a cofactor in the etiology of this disease.
Collapse
|
37
|
Seo JH, Moon HS, Kim IY, Guo DD, Lee HG, Choi YJ, Cho CS. PEGylated conjugated linoleic acid stimulation of apoptosis via a p53-mediated signaling pathway in MCF-7 breast cancer cells. Eur J Pharm Biopharm 2008; 70:621-6. [DOI: 10.1016/j.ejpb.2008.05.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2007] [Accepted: 05/09/2008] [Indexed: 12/26/2022]
|
38
|
Wang X, Wang G, Dong D, Fu S, Yang B. Inhibition on LS-174T cell growth and activity of telomerase in vitro and in vivo by arsenic trioxide. ACTA ACUST UNITED AC 2008; 60:481-8. [DOI: 10.1016/j.etp.2008.04.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2007] [Accepted: 04/21/2008] [Indexed: 01/02/2023]
|
39
|
Zhang X, Yang F, Shim JY, Kirk KL, Anderson DE, Chen X. Identification of arsenic-binding proteins in human breast cancer cells. Cancer Lett 2007; 255:95-106. [PMID: 17499915 PMCID: PMC2853370 DOI: 10.1016/j.canlet.2007.03.025] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2007] [Revised: 03/29/2007] [Accepted: 03/29/2007] [Indexed: 01/22/2023]
Abstract
As a cancer chemotherapeutic drug, arsenic acts on numerous intracellular signal transduction pathways in cancer cells. However, its mechanism of actions is still not fully understood. Previous studies suggest that arsenic reacts with closely spaced cysteine (Cys) residues of proteins with high Cys content and accessible sulfhydryl (SH) groups. In this study, human breast cancer cell line MCF-7 was examined as a cellular model to explore arsenic-binding proteins and the mechanism of binding. An arsenic-biotin conjugate was synthesized by coupling the pentafluorophenol ester of biotin with p-aminophenylarsenoxide. Arsenic-binding proteins were eluted with streptavidin resin from arsenic-biotin treated MCF-7 cells, separated by polyacrylamide gel electrophoresis, and identified by matrix assisted laser desorption ionization mass spectrometry (MALDI-MS). Arsenic-binding properties of two of these proteins, beta-tubulin and pyruvate kinase M2 (PKM2), were studied further in vitro and the biological consequences of this binding was evaluated. Binding assay with Western blotting confirmed binding of beta-tubulin and PKM2 by arsenic in a concentration-dependent manner. Arsenic binding inhibited tubulin polymerization, but surprisingly had no effect on PKM2 activity. Molecular modeling showed that binding of Cys(12) alone or vicinal Cys residues (Cys(12) and Cys(213)) of beta-tubulin by arsenic blocked the active site for access of GTP, which is necessary for tubulin polymerization. On the contrary, all Cys residues of PKM2 were far away from the active site of the enzyme. In summary, this study confirmed beta-tubulin and PKM2 as arsenic-binding proteins in MCF-7 cells. Functional consequence of such binding may depend on whether arsenic binding causes conformational changes or blocks active sites of target proteins.
Collapse
Affiliation(s)
- Xinyan Zhang
- Cancer Research Program, Julius L Chambers Biomedical/Biotechnology Research Institute, North Carolina Central University, Durham, NC 27707, USA
| | | | | | | | | | | |
Collapse
|
40
|
Dilda PJ, Hogg PJ. Arsenical-based cancer drugs. Cancer Treat Rev 2007; 33:542-64. [PMID: 17624680 DOI: 10.1016/j.ctrv.2007.05.001] [Citation(s) in RCA: 281] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2007] [Revised: 05/21/2007] [Accepted: 05/23/2007] [Indexed: 01/04/2023]
Abstract
Arsenic is a semi-metal or metalloid with two biologically important oxidation states, As(III) and As(V). As(III), in particular, reacts with closely spaced protein thiols, forming stable cyclic dithioarsinite complexes in which both sulfur atoms are bound to arsenic. It is this reaction that is mostly responsible for arsenics cytotoxicity. Arsenic compounds have been used as medicinal agents for many centuries for the treatment of diseases such as psoriasis, syphilis, and rheumatosis. From the 1700's until the introduction of and use of modern chemotherapy and radiation therapy in the mid 1900's, arsenic was a mainstay in the treatment of leukemia. Concerns about the toxicity of arsenical compounds led eventually to their abandonment for the treatment of cancer. The discovery in the 1980's that arsenic trioxide induces complete remission in a high percentage of patients with acute promyelocytic leukemia has awakened interest in this metalloid for the treatment of human disease. In particular, a new class or organoarsenicals are being trialed for the treatment of hematological malignancies and solid tumors. In this review, we discuss the arsenical-based compounds used in the past and present for the treatment of various forms of cancer. Mechanisms of action and selectivity and acute and chronic toxicities are discussed along with the prospects of this class of molecule.
Collapse
Affiliation(s)
- Pierre J Dilda
- UNSW Cancer Research Centre, University of New South Wales and Department of Haematology, Prince of Wales Hospital, Sydney 2052, Australia
| | | |
Collapse
|
41
|
Jiang HY, Jiang L, Wek RC. The eukaryotic initiation factor-2 kinase pathway facilitates differential GADD45a expression in response to environmental stress. J Biol Chem 2006; 282:3755-65. [PMID: 17170114 DOI: 10.1074/jbc.m606461200] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Phosphorylation of eukaryotic initiation factor-2 (eIF2) regulates general and gene-specific translation in response to diverse environmental stresses. Central to gene expression induced by eIF2 phosphorylation is the preferential translation of ATF4, a basic zipper transcription activator. Phosphorylation of eIF2 and its attendant induction of ATF4 can lead to different patterns of gene expression depending on the environmental stress. This is of fundamental importance because eIF2 kinases can induce the expression of genes involved in survival as well as in apoptosis. In this report, we explore the molecular basis for why there can be differential expression of GADD45a, a stress-responsive protein that regulates genome stability, apoptosis, and immune responses. We find that whereas ATF4 is required for GADD45a transcription during many different environmental stresses, GADD45a protein accumulates only during a limited number of stress arrangements. The basis for this difference between measurable GADD45a mRNA and protein lies in the observation that GADD45a protein is labile. Those stress agents that enhance ATF4-directed GADD45a transcription and impede the turnover of GADD45a protein by blocking ubiquitin/proteasome-mediated degradation elevate GADD45a protein levels. By comparison, those stress arrangements that trigger ATF4 levels and GADD45a transcription, but do not perturb the proteasome pathway, only elevate GADD45a mRNA levels. This study highlights the molecular mechanisms by which environmental stresses can differentially control central regulatory proteins targeted by the eIF2 kinase pathway.
Collapse
Affiliation(s)
- Hao-Yuan Jiang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | | | | |
Collapse
|
42
|
Liu B, Pan S, Dong X, Qiao H, Jiang H, Krissansen GW, Sun X. Opposing effects of arsenic trioxide on hepatocellular carcinomas in mice. Cancer Sci 2006; 97:675-81. [PMID: 16827809 PMCID: PMC11159334 DOI: 10.1111/j.1349-7006.2006.00230.x] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Arsenic trioxide (As2O3) is a potent antitumor agent used to treat acute promyelocytic leukemia (APL) and, more recently, solid tumors. However, the dose of As2O3 required to suppress human xenographs in mice is markedly higher than that used to treat APL in humans. Paradoxically, low doses of As2O3 stimulate angiogenesis, which might be expected to promote tumor growth. Clearly, appropriate dosages of As2O3 are required to treat human patients to avoid toxicity and undesirable side effects. In the present study, we investigated As2O3 with respect to its toxicity and effects on tumor growth, angiogenesis and cell apoptosis using H22 hepatocellular carcinoma (HCC) cells in a mouse model of HCC. As2O3 inhibited tumor growth and angiogenesis, and enhanced tumor cell apoptosis at doses greater than 1 mg/kg, but mice lost weight and failed to thrive at doses of 4 mg/kg and greater. In contrast, low doses (<1 mg/kg) of As2O3 promoted tumor growth, upregulated the expression of vascular endothelial growth factor and tumor angiogenesis, and had no effect on tumor cell apoptosis. In vitro studies demonstrated that As2O3 inhibited the proliferation of H22 tumor cells and bovine aortic endothelial cells, and induced their apoptosis in a dose- and time-dependent fashion, suggesting that the mechanism of As2O3-mediated inhibition of tumor growth is due to direct effects of the drug on both tumor cells and endothelia. In summary, different doses of As2O3 have opposing effects on tumor growth and angiogenesis. The results demonstrate that As2O3 has a narrow window of therapeutic opportunity with respect to dosage, and that low doses of the drug as used in metronomic therapy should be used with extreme caution.
Collapse
Affiliation(s)
- Bing Liu
- The Hepatosplenic Surgery Center of Heilongjiang Province/Department of General Surgery, The First Clinical College of Harbin Medical University, Harbin 150001, China
| | | | | | | | | | | | | |
Collapse
|
43
|
Tsou TC, Tsai FY, Yeh SC, Chang LW. ATM/ATR-related checkpoint signals mediate arsenite-induced G2/M arrest in primary aortic endothelial cells. Arch Toxicol 2006; 80:804-10. [PMID: 16645841 DOI: 10.1007/s00204-006-0110-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2006] [Accepted: 04/12/2006] [Indexed: 01/08/2023]
Abstract
Epidemiological studies have demonstrated a high association of inorganic arsenic exposure with vascular disease. Our recent in vitro studies have linked this vascular damage to vascular endothelial dysfunction induced by arsenic exposure. However, cell-cycle arrest induced by arsenic and its involvement in vascular dysfunction remain to be clarified. In this study, we employed primary porcine aortic endothelial cells to investigate regulatory mechanisms of G2/M phase arrest induced by arsenite. Our study revealed that lower concentrations of arsenite (1 and 3 microM) increased cell proliferation, whereas higher concentrations of arsenite (10, 20, and 30 microM) inhibited cell proliferation together with correlated increases in G2/M phase arrest. We found that this arsenite-induced G2/M phase arrest was accompanied by accumulation and/or phosphorylation of checkpoint-related molecules, including p53, Cdc25B, Cdc25C, and securin. Inhibition of activations of these checkpoint-related molecules by caffeine significantly attenuated the 30-microM arsenite-induced G2/M phase arrest by 93%. Our data suggest that the DNA damage responsive kinases ATM (ataxia-telangiectasia mutated) and ATR (ATM and Rad3-related) play critical roles in arsenite-induced G2/M phase arrest in aortic endothelial cells possibly via regulation of checkpoint-related signaling molecules including p53, Cdc25B, Cdc25C, and securin.
Collapse
Affiliation(s)
- Tsui-Chun Tsou
- Division of Environmental Health and Occupational Medicine, National Health Research Institutes, 35 Keyan Road, Zhunan Town, Miaoli County 35053, Taiwan.
| | | | | | | |
Collapse
|
44
|
Liu ZM, Huang HS. As2O3-induced c-Src/EGFR/ERK signaling is via Sp1 binding sites to stimulate p21WAF1/CIP1 expression in human epidermoid carcinoma A431 cells. Cell Signal 2006; 18:244-55. [PMID: 15961274 DOI: 10.1016/j.cellsig.2005.04.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2005] [Revised: 04/19/2005] [Accepted: 04/19/2005] [Indexed: 11/30/2022]
Abstract
Arsenic has been effectively used to treat acute promyelocytic leukemia, and can induce cell cycle arrest or apoptosis in human solid tumors. Previously, we have demonstrated that As2O3 can induce p21WAF1/CIP1 (p21) expression in A431 cells and then due to cellular cytotoxicity. Presently, we have clarified these signaling events and compared them with EGF. Using reporter assay, RT-PCR and Western blotting, we show that c-Src activation might be a prerequisite for As2O3-induced EGFR/Ras/Raf/ERK signaling. Furthermore, with the aids of 5'-deletion and site-directed mutagenesis, we demonstrate that Sp1 binding sites, ranging from -64 to -84 bp, are essential for As2O3- or EGF-regulated p21 expression. Finally, our experiments utilizing cycloheximide prompt the suggestion that the stability of mRNA or protein also contributes to As2O3- or EGF-induced p21 expression. Taken together, we conclude that the Sp1 binding sites are required for As2O3-induced p21 gene transcription through c-Src/EGFR/Ras/Raf/ERK pathway. Furthermore, post-transcriptional or post-translational stabilization mechanism is also essential for As2O3-induced p21 expression. EGF-induced p21 expression may involve similar mechanisms as those that operate in the As2O3-mediated reactions in A431 cells.
Collapse
Affiliation(s)
- Zi-Miao Liu
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | | |
Collapse
|
45
|
Binet F, Cavalli H, Moisan E, Girard D. Arsenic trioxide (AT) is a novel human neutrophil pro-apoptotic agent: effects of catalase on AT-induced apoptosis, degradation of cytoskeletal proteins and de novo protein synthesis. Br J Haematol 2006; 132:349-58. [PMID: 16409300 DOI: 10.1111/j.1365-2141.2005.05866.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The anti-cancer drug arsenic trioxide (AT) induces apoptosis in a variety of transformed or proliferating cells. However, little is known regarding its ability to induce apoptosis in terminally differentiated cells, such as neutrophils. Because neutropenia has been reported in some cancer patients after AT treatment, we hypothesised that AT could induce neutrophil apoptosis, an issue that has never been investigated. Herein, we found that AT-induced neutrophil apoptosis and gelsolin degradation via caspases. AT did not increase neutrophil superoxide production and did not induce mitochondrial generation of reactive oxygen species. AT-induced apoptosis in PLB-985 and X-linked chronic granulomatous disease (CGD) cells (PLB-985 cells deficient in gp91(phox) mimicking CGD) at the same potency. Addition of catalase, an inhibitor of H2O2, reversed AT-induced apoptosis and degradation of the cytoskeletal proteins gelsolin, alpha-tubulin and lamin B1. Unexpectedly, AT-induced de novo protein synthesis, which was reversed by catalase. Cycloheximide partially reversed AT-induced apoptosis. We conclude that AT induces neutrophil apoptosis by a caspase-dependent mechanism and via de novo protein synthesis. H2O2 is of major importance in AT-induced neutrophil apoptosis but its production does not originate from nicotinamide adenine dinucleotide phosphate dehydrogenase activation and mitochondria. Cytoskeletal structures other than microtubules can now be considered as novel targets of AT.
Collapse
Affiliation(s)
- François Binet
- INRS-Institut Armand-Frappier, Université du Québec, Pointe-Claire, PQ, Canada
| | | | | | | |
Collapse
|
46
|
Ryu KH, Woo SY, Lee MY, Jung YJ, Yoo ES, Seoh JY, Kie JH, Shin HY, Ahn HS. Morphological and biochemical changes induced by arsenic trioxide in neuroblastoma cell lines. Pediatr Hematol Oncol 2005; 22:609-21. [PMID: 16166054 DOI: 10.1080/08880010500198897] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Arsenic trioxide has recently been shown to inhibit growth and induce apoptosis in a variety of hematologic malignancies, but very little is known about its effects on solid tumors and especially on neuroblastoma cells that have self-differentiating characteristics. To demonstrate the growth inhibition induced in neuroblastoma cells (the SH-SY5Y and SK-N-AS cell line) and acute promyelocytic leukemia cells (HL-60) by arsenic trioxide (As2O3), the viable cell numbers were counted after trypan blue staining. Apoptosis was assessed by the cell morphology, by flow cytometry with annexin-V staining, and by Western blot analysis for the apoptosis-related proteins (bcl-2 and PARP). To decide the dose for the clinical application of As2O3, normal peripheral blood lymphocytes were also examined. The growth and survival of the SH-SY5Y and SK-N-AS cells were markedly inhibited by As2O3 treatment at a 3 microM concentration before the changes of the normal lymphocytes were observed. The apoptotic cells showed a shrunken cell nucleus, and an increase in the number and balloon-like swelling of the mitochondria at 72 h after the As2O3 was added. Apoptosis of the annexin-V-positive cell proportion in the neuroblastoma cell lines was increased with increasing the exposure time and the concentration of As2O3, just like the HL-60 cells. Bcl-2 downregulation and PARP degradation were also noted all the cell lines, but these changes were not statistically significant among the 3 cell lines. Taken together, these results indicate that As2O3 is an excellent candidate as a therapeutic agent for the treatment of neuroblastoma.
Collapse
Affiliation(s)
- Kyung-Ha Ryu
- Department of Pediatrics and Ewha Medical Research Institute, Ewha Woman's University College of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Liu JB, Qin SK, Li J. Effect of arsenic trioxide on pancreatic carcinoma cell line PC-3 in vitro. Shijie Huaren Xiaohua Zazhi 2005; 13:1713-1716. [DOI: 10.11569/wcjd.v13.i14.1713] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To explore the biological effect of arsenic trioxide (As2O3) on human pancreatic carcinoma cell line PC-3 and its mechanism.
METHODS: MTT assay was used to observe the inhibitory actions of As2O3 on PC-3 cells at various concentrations. The apoptotic rate and growth cycle of the cells were detected by flow cytometry. The changes of the cells ultrastructures were observed under electron microscope. The expression of apoptosis-related gene protein (Fas, Fas-L, Bcl-2, Bax) was detected by immunohistochemical staining.
RESULTS: As2O3 inhibited the proliferation of PC-3 cells in a concentration- and time-dependent manner. Marked apoptosis peak was observed and the cells were mainly blocked in S phase (14.86-63.66%). PC-3 cells showed obvious feature of apoptosis under electron microscope, such as intact cell membrane, pyknosis of chromatin, nuclear fragmentation and apoptotic body formation. Bcl-2 protein was weakly expressed (+++) in controls, but strongly expressed (+) in As2O3-treated cells. Fas-L protein were weakly (+) and negatively (-) expressed in controls respectively, but strongly expressed (+++) in As2O3-treated cells.
CONCLUSION: As2O3 can inhibit the proliferation and induce the apoptosis of human pancreatic carcinoma cell PC-3 and the mechanism is probably related to its effect on the regulation of Fas, Fas-L and Bcl-2 expression.
Collapse
|