151
|
Elkady AI, Hussein RAEH, Abu-Zinadah OA. Differential control of growth, apoptotic activity and gene expression in human colon cancer cells by extracts derived from medicinal herbs, Rhazya stricta and Zingiber officinale and their combination. World J Gastroenterol 2014; 20:15275-88. [PMID: 25386076 PMCID: PMC4223261 DOI: 10.3748/wjg.v20.i41.15275] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 06/29/2014] [Accepted: 07/11/2014] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the effects of extracts from Rhazya stricta (R. stricta) and Zingiber officinale (Z. officinale) on human colorectal cancer cells. METHODS Human colorectal cancer cells (HCT116) were subjected to increasing doses of crude alkaloid extracts from R. stricta (CAERS) and crude flavonoid extracts from Z. officinale (CFEZO). Cells were then harvested after 24, 48 or 72 h and cell viability was examined by trypan blue exclusion dye test; clonogenicity and soft agar colony-forming assays were also carried out. Nuclear stain (Hoechst 33342), acridine orange/ethidium bromide double staining, agarose gel electrophoresis and comet assays were performed to assess pro-apoptotic potentiality of the extracts. Quantitative reverse transcription-polymerase chain reaction (qRT-PCR), using gene-specific primers and Western blot analyses were performed to assess the impact of CAERS and CFEZO on the expression levels of key regulatory proteins in HCT116 cells. RESULTS Treatment with a combination of CAERS and CFEZO synergistically suppressed the proliferation, colony formation and anchorage-independent growth of HCT116 cells. Calculated IC50, after 24, 48 and 72 h, were 70, 90 and 130 μg/mL for CAERS, 65, 85 and 120 μg/mL for CFEZO and 20, 25 and 45 μg/mL for both agents, respectively. CAERS- and CFEZO-treated cells exhibited morphologic and biochemical features of apoptotic cell death. The induction of apoptosis was associated with the release of mitochondrial cytochrome c, an increase in the Bax/Bcl-2 ratio, activation of caspases 3 and 9 and cleavage of poly ADP-ribose polymerase. CAERS and CFEZO treatments downregulated expression levels of anti-apoptotic proteins including Bcl-2, Bcl-X, Mcl-1, survivin and XIAP, and upregulated expression levels of proapoptotic proteins such as Bad and Noxa. CAERS and CFEZO treatments elevated expression levels of the oncosuppressor proteins, p53, p21 and p27, and reduced levels of the oncoproteins, cyclin D1, cyclin/cyclin-dependent kinase-4 and c-Myc. CONCLUSION These data suggest that a combination of CAERS and CFEZO is a promising treatment for the prevention of colon cancer.
Collapse
|
152
|
Liu J, Zhang C, Wang XL, Ly P, Belyi V, Xu-Monette ZY, Young KH, Hu W, Feng Z. E3 ubiquitin ligase TRIM32 negatively regulates tumor suppressor p53 to promote tumorigenesis. Cell Death Differ 2014; 21:1792-1804. [PMID: 25146927 PMCID: PMC4211364 DOI: 10.1038/cdd.2014.121] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Revised: 07/12/2014] [Accepted: 07/14/2014] [Indexed: 01/07/2023] Open
Abstract
Tumor suppressor p53 has a key role in maintaining genomic stability and preventing tumorigenesis through its regulation of cellular stress responses, including apoptosis, cell cycle arrest and senescence. To ensure its proper levels and functions in cells, p53 is tightly regulated mainly through post-translational modifications, such as ubiquitination. Here, we identified E3 ubiquitin ligase TRIM32 as a novel p53 target gene and negative regulator to regulate p53-mediated stress responses. In response to stress, such as DNA damage, p53 binds to the p53 responsive element in the promoter of the TRIM32 gene and transcriptionally induces the expression of TRIM32 in cells. In turn, TRIM32 interacts with p53 and promotes p53 degradation through ubiquitination. Thus, TRIM32 negatively regulates p53-mediated apoptosis, cell cycle arrest and senescence in response to stress. TRIM32 is frequently overexpressed in different types of human tumors. TRIM32 overexpression promotes cell oncogenic transformation and tumorigenesis in mice in a largely p53-dependent manner. Taken together, our results demonstrated that as a novel p53 target and a novel negative regulator for p53, TRIM32 has an important role in regulation of p53 and p53-mediated cellular stress responses. Furthermore, our results also revealed that impairing p53 function is a novel mechanism for TRIM32 in tumorigenesis.
Collapse
Affiliation(s)
- Ju Liu
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, New Brunswick, NJ, USA
| | - C Zhang
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, New Brunswick, NJ, USA
| | - X L Wang
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, New Brunswick, NJ, USA
- Department of Breast Surgery, Qilu Hospital, Shandong University, Ji'nan, China
| | - P Ly
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, New Brunswick, NJ, USA
| | - V Belyi
- Center for Systems Biology, Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, New Brunswick, NJ, USA
| | - Z Y Xu-Monette
- Department of Hematopathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - K H Young
- Department of Hematopathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - W Hu
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, New Brunswick, NJ, USA
| | - Z Feng
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, New Brunswick, NJ, USA
| |
Collapse
|
153
|
Yu H, Yue X, Zhao Y, Li X, Wu L, Zhang C, Liu Z, Lin K, Xu-Monette ZY, Young KH, Liu J, Shen Z, Feng Z, Hu W. LIF negatively regulates tumour-suppressor p53 through Stat3/ID1/MDM2 in colorectal cancers. Nat Commun 2014; 5:5218. [PMID: 25323535 PMCID: PMC4203416 DOI: 10.1038/ncomms6218] [Citation(s) in RCA: 148] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 09/10/2014] [Indexed: 12/21/2022] Open
Abstract
Leukaemia inhibitory factor (LIF) has been recently identified as a p53 target gene, which mediates the role of p53 in maternal implantation under normal physiological conditions. Here we report that LIF is a negative regulator of p53; LIF downregulates p53 protein levels and function in human colorectal cancer (CRC) cells. The downregulation of p53 by LIF is mediated by the activation of Stat3, which transcriptionally induces inhibitor of DNA-binding 1 (ID1). ID1 upregulates MDM2, a key negative regulator of p53, and promotes p53 protein degradation. LIF is overexpressed in a large percentage of CRCs. LIF overexpression promotes cellular resistance towards chemotherapeutic agents in cultured CRC cells and colorectal xenograft tumours in a largely p53-dependent manner. Overexpression of LIF is associated with a poor prognosis in CRC patients. Taken together, LIF is a novel negative regulator of p53, overexpression of LIF is an important mechanism for the attenuation of p53, which promotes chemoresistance in CRCs.
Collapse
Affiliation(s)
- Haiyang Yu
- Rutgers Cancer Institute of New Jersey, Rutgers the State University of New Jersey, New Brunswick, NJ 08903
| | - Xuetian Yue
- Rutgers Cancer Institute of New Jersey, Rutgers the State University of New Jersey, New Brunswick, NJ 08903
| | - Yuhan Zhao
- Rutgers Cancer Institute of New Jersey, Rutgers the State University of New Jersey, New Brunswick, NJ 08903
| | - Xiaoyan Li
- Rutgers Cancer Institute of New Jersey, Rutgers the State University of New Jersey, New Brunswick, NJ 08903
- Department of Breast Surgery, Qilu Hospital, Shandong University, Ji’nan, China
| | - Lihua Wu
- Rutgers Cancer Institute of New Jersey, Rutgers the State University of New Jersey, New Brunswick, NJ 08903
- First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Cen Zhang
- Rutgers Cancer Institute of New Jersey, Rutgers the State University of New Jersey, New Brunswick, NJ 08903
| | - Zhen Liu
- Rutgers Cancer Institute of New Jersey, Rutgers the State University of New Jersey, New Brunswick, NJ 08903
| | - Kevin Lin
- Rutgers Cancer Institute of New Jersey, Rutgers the State University of New Jersey, New Brunswick, NJ 08903
| | - Zijun Y. Xu-Monette
- Department of Hematopathology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030
| | - Ken H. Young
- Department of Hematopathology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030
| | - Juan Liu
- Rutgers Cancer Institute of New Jersey, Rutgers the State University of New Jersey, New Brunswick, NJ 08903
| | - Zhiyuan Shen
- Rutgers Cancer Institute of New Jersey, Rutgers the State University of New Jersey, New Brunswick, NJ 08903
| | - Zhaohui Feng
- Rutgers Cancer Institute of New Jersey, Rutgers the State University of New Jersey, New Brunswick, NJ 08903
| | - Wenwei Hu
- Rutgers Cancer Institute of New Jersey, Rutgers the State University of New Jersey, New Brunswick, NJ 08903
| |
Collapse
|
154
|
Truong WT, Su Y, Gloria D, Braet F, Thordarson P. Dissolution and degradation of Fmoc-diphenylalanine self-assembled gels results in necrosis at high concentrations in vitro. Biomater Sci 2014. [PMID: 26218120 DOI: 10.1039/c4bm00244j] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Herein we report an approach to assess in vitro cellular responses to the dissolution or degradation products from Fmoc-diphenylalanine (Fmoc-FF) self-assembled hydrogels. Three cell lines were used in these studies and two-way ANOVA was used to assess (i) the age of gel dissolution and degradation products and (ii) exposure time on cell fate and state, using viability assays in conjunction with time-lapse fluorescence and high-resolution scanning electron microscopy investigation. The studies show that leaching time but not the exposure time affects the overall cell viability. The cytotoxic effect was only observed once the gel is completely dissolved. Further analysis revealed that the principal mechanism of cell death is necrosis. In addition, the effect of chemotherapeutics (5-fluorouracil and paclitaxel) released from the Fmoc-FF gel (with addition before and after gelation) on colorectal cancer cells were investigated using this methodology, demonstrating enhanced activity of these drugs compared to bulk control. This enhanced activity, however, appears to be a combination of the apoptosis caused by the cancer drugs and necrosis caused by gel dissolution and degradation products. Given that in vivo studies by others on Fmoc-peptides that this material is not harmful to animals, our work highlights that conventional in vitro cellular assays may yield conflicting messages when used for the evaluation of cytotoxicity and drug release from self-assembled gels such as Fmoc-FF and that better in vitro models, (e.g. 3D cell culture systems) need to be developed to evaluate these materials for biomedical applications.
Collapse
Affiliation(s)
- Warren Ty Truong
- School of Chemistry, the Australian Centre for Nanomedicine and the ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of New South Wales, Sydney, NSW 2052, Australia.
| | | | | | | | | |
Collapse
|
155
|
Bachmann P, Bensadoun RJ, Besnard I, Bourdel-Marchasson I, Bouteloup C, Crenn P, Goldwasser F, Guérin O, Latino-Martel P, Meuric J, May-Lévin F, Michallet M, Vasson MP, Hébuterne X. Clinical nutrition guidelines of the French Speaking Society of Clinical Nutrition and Metabolism (SFNEP): Summary of recommendations for adults undergoing non-surgical anticancer treatment. Dig Liver Dis 2014; 46:667-74. [PMID: 24794790 DOI: 10.1016/j.dld.2014.01.160] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 01/28/2014] [Indexed: 12/11/2022]
Abstract
Up to 50% of patients with cancer suffer from weight loss and undernutrition (as called cachexia) even though it is rarely screened or properly handled. Patients' prognosis and quality of life could be greatly improved by simple and inexpensive means encompassing nutritional status assessment and effective nutritional care. These guidelines aim to give health professionals and patients practical and up-to-date advice to manage nutrition in the principal situations encountered during the cancer course according to the type of tumour and treatment (i.e. radio and/or chemotherapy). Specific suggestions are made for palliative and elderly patients because of specific risks of undernutrition and related comorbidities in this subset. Levels of evidence and grades of recommendations are detailed as stated by current literature and consensus opinion of clinical experts in each field.
Collapse
|
156
|
El-Zahany EA, Ali MM, Drweesh SA, El-Seidy AMA, Abdel-Wahab BF, Youssef NS. Synthesis, Characterization, and Antiproliferative Activity of Cu2+, V(IV)O2+, Co2+, Mn2+, and Ni2+ Complexes with 3-(2-(4-Methoxyphenylcarbamothioyl)Hydrazinyl)-3-OXO-N-(Thiazol-2-yl)Propanamide against Human Breast Adenocarcinoma Cells. PHOSPHORUS SULFUR 2014. [DOI: 10.1080/10426507.2013.855764] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Eman A. El-Zahany
- Inorganic Chemistry Department, National Research Centre, P.O. 12622 Dokki, Cairo, Egypt
| | - Mamdouh M. Ali
- Biochemistry Department, National Research Centre, P.O. 12622 Dokki, Cairo, Egypt
| | - Sayed A. Drweesh
- Inorganic Chemistry Department, National Research Centre, P.O. 12622 Dokki, Cairo, Egypt
| | - Ahmed M. A. El-Seidy
- Inorganic Chemistry Department, National Research Centre, P.O. 12622 Dokki, Cairo, Egypt
| | - Bakr F. Abdel-Wahab
- Applied Organic Chemistry Department, National Research Centre, P.O. 12622 Dokki, Cairo, Egypt
| | - Nabil S. Youssef
- Inorganic Chemistry Department, National Research Centre, P.O. 12622 Dokki, Cairo, Egypt
| |
Collapse
|
157
|
Winter E, Dal Pizzol C, Filippin-Monteiro FB, Brondani P, Silva AMPW, Silva AH, Bonacorso HG, Martins MAP, Zanatta N, Creczynski-Pasa TB. Antitumoral activity of a trichloromethyl pyrimidine analogue: molecular cross-talk between intrinsic and extrinsic apoptosis. Chem Res Toxicol 2014; 27:1040-9. [PMID: 24848672 DOI: 10.1021/tx500094x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Acute lymphoblastic leukemia (ALL) is a malignant disorder caused by the proliferation of lymphoid progenitor cells and is the most common cancer in children. Cytotoxic nucleoside analogues are important chemotherapeutic agents, which are used in many cancers, including leukemias. In this study, we investigated the effects of the synthetic nucleoside analogue 1-(5,5,5-trichloro-2-methoxy-4-oxopenten-2-yl)-4-trichloromethyl-pyrimidin-2(1H)-one, named compound 3 or C3, on leukemia cell lines. The compound stimulated cell death by apoptosis, evidenced by DNA fragmentation, phosphatidylserine externalization, and caspase-3 activation. Compound 3 seemed to trigger several cell death pathways. The mitochondrial pathway was evidenced through a disturbance of mitochondrial membrane potential, strong cytochrome c liberation, decrease of antiapoptotic Bcl-2 protein expression, and caspase-9 activation. The C3 also induced caspase-8 and -12 activation, an increase in the intracellular calcium level, and an overproduction of reactive oxygen species. Increased caspase 8 activity suggests that the extrinsic pathway was activated and that the ROS production and enzyme activity alteration (glutathione S-transferase, glutathione peroxidase, catalase, and glutathione reductase) might be related to oxidative stress. Finally, the increase in calcium release, CHOP expression, and caspase-12 activity might characterize endoplasmic reticulum stress. Compound 3 was likewise cytotoxic to leukemic and melanoma human cell lines. Taken together, the results contribute to further understanding the new pyrimidine analogue as a potential chemotherapeutic drug or lead molecule.
Collapse
Affiliation(s)
- Evelyn Winter
- Departamento de Ciências Farmacêuticas, Programa de Pós-graduação em Farmácia, Universidade Federal de Santa Catarina , 88040-900 Florianópolis SC, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
158
|
Chemotherapy-Associated Hepatotoxicity and Hepatic Resection for Metastatic Colorectal Cancer. CURRENT COLORECTAL CANCER REPORTS 2014. [DOI: 10.1007/s11888-014-0218-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
159
|
Chen P, Wang H, Duan Z, Zou JX, Chen H, He W, Wang J. Estrogen-related receptor alpha confers methotrexate resistance via attenuation of reactive oxygen species production and P53 mediated apoptosis in osteosarcoma cells. BIOMED RESEARCH INTERNATIONAL 2014; 2014:616025. [PMID: 24967384 PMCID: PMC4055217 DOI: 10.1155/2014/616025] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 04/25/2014] [Indexed: 01/26/2023]
Abstract
Osteosarcoma (OS) is a malignant tumor mainly occurring in children and adolescents. Methotrexate (MTX), a chemotherapy agent, is widely used in treating OS. However, treatment failures are common due to acquired chemoresistance, for which the underlying molecular mechanisms are still unclear. In this study, we report that overexpression of estrogen-related receptor alpha (ERR α ), an orphan nuclear receptor, promoted cell survival and blocked MTX-induced cell death in U2OS cells. We showed that MTX induced ROS production in MTX-sensitive U2OS cells while ERR α effectively blocked the ROS production and ROS associated cell apoptosis. Our further studies demonstrated that ERR α suppressed ROS induction of tumor suppressor P53 and its target genes NOXA and XAF1 which are mediators of P53-dependent apoptosis. In conclusion, this study demonstrated that ERR α plays an important role in the development of MTX resistance through blocking MTX-induced ROS production and attenuating the activation of p53 mediated apoptosis signaling pathway, and points to ERR α as a novel target for improving osteosarcoma therapy.
Collapse
Affiliation(s)
- Peng Chen
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
- Cancer Center, University of California, Davis, Sacramento, CA 95817, USA
| | - Haibin Wang
- State Key Hip Joints Center, First Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 12 Jichang Road, Guangzhou, Guangdong 510405, China
| | - Zhijian Duan
- Cancer Center, University of California, Davis, Sacramento, CA 95817, USA
| | - June X. Zou
- Cancer Center, University of California, Davis, Sacramento, CA 95817, USA
| | - Hongwu Chen
- Cancer Center, University of California, Davis, Sacramento, CA 95817, USA
| | - Wei He
- State Key Hip Joints Center, First Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 12 Jichang Road, Guangzhou, Guangdong 510405, China
| | - Junjian Wang
- Cancer Center, University of California, Davis, Sacramento, CA 95817, USA
| |
Collapse
|
160
|
Ismail AFM, Ali MM, Ismail LFM. Photodynamic therapy mediated antiproliferative activity of some metal-doped ZnO nanoparticles in human liver adenocarcinoma HepG2 cells under UV irradiation. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2014; 138:99-108. [PMID: 24911277 DOI: 10.1016/j.jphotobiol.2014.04.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2014] [Revised: 03/29/2014] [Accepted: 04/09/2014] [Indexed: 01/10/2023]
Abstract
Photodynamic therapy (PDT) is a promising new modality for the treatment of cancer through generation of reactive oxygen species (ROS). In this work, human liver adenocarcinoma cells HepG2 were treated with zinc oxide nanoparticles (ZnO-NPs), metal-doped-ZnO-NPs: Fe-ZnO-NPs Ag-ZnO-NPs, Pb-ZnO-NPs, and Co-ZnO-NPs, Silica-coated ZnO-NPs, titanium dioxide nanoparticles (TiO2-NPs), titanium dioxide nano-tubes (TiO2-NTs) and ZnO-NPs/TiO2-NTs nanocomposite under UV irradiation. Doxorubicin was used as a standard drug. The results demonstrated that the ZnO-NPs, Fe-ZnO-NPs, Ag-ZnO-NPs, Pb-ZnO-NPs, and Co-ZnO-NPs showed cytotoxicity against HepG2 cells, with the median growth inhibitory concentrations (IC50) 42.60, 37.20, 45.10, 77.20 and 56.50 μg/ml, respectively, as compared to doxorubicin (IC50: 20.10 μg/ml). Treatment of the cancer cells with ZnO-NPs, Fe-ZnO-NPs, Ag-ZnO-NPs, Pb-ZnO-NPs, and Co-ZnO-NPs resulted in a significant increase in the activity of SOD and the levels of H2O2 and NO than those of control, accompanied with a significant decrease in the activity of CAT and GSH-Px. Also, depletion of reduced GSH, total protein and nucleic acids levels was observed. In conclusion, metal-doped ZnO-NPs may induce antiproliferative effect on HepG2 cells under UV-irradiation due to generation of ROS. Therefore, they could be included in modern clinical trials after in vivo more investigations, using photodynamic therapy technique.
Collapse
Affiliation(s)
- Amel F M Ismail
- Drug Radiation Research Department, National Center for Radiation Research and Technology, Atomic Energy Authority, Nasr City, Cairo, Egypt.
| | - Mamdouh M Ali
- Biochemistry Department, Division of Genetic Engineering and Biotechnology, National Research Centre, Dokki, Giza, Egypt
| | - Laila F M Ismail
- Chemistry Department, Al-Azhar University, Faculty of Science, Nasr City, Cairo, Egypt
| |
Collapse
|
161
|
Huillard O, Boissier E, Blanchet B, Thomas-Schoemann A, Cessot A, Boudou-Rouquette P, Durand JP, Coriat R, Giroux J, Alexandre J, Vidal M, Goldwasser F. Drug safety evaluation of sorafenib for treatment of solid tumors: consequences for the risk assessment and management of cancer patients. Expert Opin Drug Saf 2014; 13:663-73. [PMID: 24693873 DOI: 10.1517/14740338.2014.907270] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Sorafenib is a multi-tyrosine kinase inhibitor (TKI). Considerable clinical experience has been accumulated since its first Phase III clinical trial in metastatic renal cancer patients in 2007. The management of its early acute toxicity in fit patients is well known. The management of prolonged treatment becomes the new challenge. AREAS COVERED Using sorafenib as a key word for PubMed search, we review preclinical and clinical data and discuss the pharmacokinetics and pharmacodynamics of sorafenib, its acute and cumulative toxicities and their consequences for patient management. EXPERT OPINION The systematic multi-disciplinary risk assessment of cancer patients prior to TKI initiation reduces the risks of acute and late toxicity, especially drug-drug interactions and arterial risks. Sarcopenia is now identified as a major risk of severe toxicity. The very diverse clinical pictures of cumulative toxicity must be known. The monitoring of sorafenib systemic exposure is helpful especially in elderly patients. Moreover, at disease progression, it allows distinguishing between underexposure to sorafenib and truly acquired resistance to the drug. The optimal use of sorafenib should allow improving the reported results of flat-dose. Finally, most of this knowledge could be used for the development and optimal use of the other TKIs.
Collapse
Affiliation(s)
- Olivier Huillard
- Paris Descartes University, Cochin Hospital, AP-HP, Medical Oncology Department, Angiogenesis Inhibitors Multidisciplinary Study Group (CERIA) , Paris , France +33 1 58 41 17 46 ; +33 1 58 41 17 45 ;
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
162
|
Kitazono M, Shinchi H, Ishigami S, Ueno S, Natsugoe S. Effects of a histone deacetylase inhibitor, sodium butyrate, on 53-kDa protein expression and sensitivity to anticancer drugs of pancreatic cancer cells. Curr Ther Res Clin Exp 2014; 71:162-72. [PMID: 24683262 DOI: 10.1016/j.curtheres.2010.06.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2010] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Several tumor-suppressor genes, such as 53-kDa protein (p53), are inactivated in some pancreatic cancers. The lack of a functional p53 has been proposed to be a component of resistance to chemotherapy, resulting in the inhibition of apoptosis. Therefore, reintroduction of wild-type p53 is a commonly used gene therapy strategy for the treatment of various types of cancer, including pancreatic cancer. OBJECTIVE The aim of this study was to examine the ability of the histone deacetylase inhibitor, sodium butyrate (NaB), to modulate the expression of p53. METHODS Five human pancreatic carcinoma cell lines (SW-1990, BxPC-3, PANC-1, MIA PaCa-2, JHP-1) were utilized. Two of the cell lines (SW-1990 and JHP-1) lacked p53 expression, as determined by Western blot analysis, and were investigated further. Expression of p53 was determined by densitometry of all bands present in the Western blot. Drug sensitivity was measured with a tetrazolium-based assay by exposing the cells to graded concentrations of NaB and/or anticancer drugs (cisplatin, fluorouracil, SN-38, and paclitaxel). Apoptosis was observed using gel electrophoresis. RESULTS In the SW-1990 and JHP-1 cell lines, use of 1 mM NaB was found to induce histone acetylation and p53 expression compared with those not treated with NaB (P = 0.01 and P = 0.018, respectively). Sensitivity to cisplatin (P = 0.021), fluorouracil (P = 0.046), and SN-38 (P = 0.039) was significantly enhanced by NaB treatment compared with nontreatment. However, sensitivity to paclitaxel was not significantly different between untreated and NaB-treated cells. A higher frequency of apoptosis was observed in NaB-treated cells compared with that of control cells. CONCLUSION This in vitro study found that NaB induced p53 expression in 2 pancreatic cancer cell lines (SW-1990 and JHP-1). Moreover, NaB acted on a biochemical modulator for antieuplastic therapy. Future research is necessary to assess the value of these findings.
Collapse
Affiliation(s)
- Masaki Kitazono
- Department of Surgical Oncology and Digestive Surgery, Kagoshima University Graduate School, Kagoshima, Japan
| | - Hiroyuki Shinchi
- Department of Surgical Oncology and Digestive Surgery, Kagoshima University Graduate School, Kagoshima, Japan
| | - Sumiya Ishigami
- Department of Surgical Oncology and Digestive Surgery, Kagoshima University Graduate School, Kagoshima, Japan
| | - Shinichi Ueno
- Department of Surgical Oncology and Digestive Surgery, Kagoshima University Graduate School, Kagoshima, Japan
| | - Shoji Natsugoe
- Department of Surgical Oncology and Digestive Surgery, Kagoshima University Graduate School, Kagoshima, Japan
| |
Collapse
|
163
|
Xu Y, Qi J, Yang X, Wu E, Qian SY. Free radical derivatives formed from cyclooxygenase-catalyzed dihomo-γ-linolenic acid peroxidation can attenuate colon cancer cell growth and enhance 5-fluorouracil's cytotoxicity. Redox Biol 2014; 2:610-8. [PMID: 25114837 PMCID: PMC4124262 DOI: 10.1016/j.redox.2014.01.022] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 01/30/2014] [Indexed: 12/27/2022] Open
Abstract
Dihomo-γ-linolenic acid (DGLA) and its downstream fatty acid arachidonic acid (AA) are both nutritionally important ω-6 polyunsaturated fatty acids (ω-6s). Evidence shows that, via COX-mediated peroxidation, DGLA and its metabolites (1-series prostaglandins) are associated with anti-tumor activity, while AA and its metabolites (2-series prostaglandins) could be tightly implicated in various cancer diseases. However, it still remains a mystery why DGLA and AA possess contrasting bioactivities. Our previous studies showed that DGLA could go through an exclusive C-8 oxygenation pathway during COX-catalyzed lipid peroxidation in addition to a C-15 oxygenation pathway shared by both DGLA and AA, and that the exclusive C-8 oxygenation could lead to the production of distinct DGLA׳s free radical derivatives that may be correlated with DGLA׳s anti-proliferation activity. In the present work, we further investigate the anti-cancer effect of DGLA׳s free radical derivatives and their associated molecular mechanisms. Our study shows that the exclusive DGLA׳s free radical derivatives from C-8 oxygenation lead to cell growth inhibition, cell cycle arrest and apoptosis in the human colon cancer cell line HCA-7 colony 29, probably by up-regulating the cancer suppressor p53 and the cell cycle inhibitor p27. In addition, these exclusive radical derivatives were also able to enhance the efficacy of 5-Fluorouracil (5-FU), a widely used chemo-drug for colon cancer. For the first time, we show how DGLA׳s radical pathway and metabolites are associated with DGLA׳s anti-cancer activities and able to sensitize colon cancer cells to chemo-drugs such as 5-FU. Our findings could be used to guide future development of a combined chemotherapy and dietary care strategy for colon cancer treatment.
Collapse
Key Words
- 5-FU, 5-Fluorouracil
- 5-Fluorouracil
- 8-HOA, 8-hydroxyoctanoic acid
- AA, arachidonic acid
- ACN, acetonitrile
- COX, cyclooxygenase
- COX-catalyzed PUFA peroxidation
- Cell cycle and apoptosis
- Colon cancer cell line HCA-7 colony 29
- D5D, delta-5 desaturase
- DGLA, dihomo-γ-linoleic acid
- DGLA׳s free radical derivatives
- DHA, docosahexaenoic acid
- EIC, extracted ion chromatogram
- EPA, eicosapentaenoic acid
- ESR, electron spin resonance
- GC, gas chromatography
- HEX, 1-hexanol
- HOAc, glacial acetic acid
- HPLC/LC, high performance liquid chromatography
- HTA, heptanoic acid
- LC/MS and ESR spin trapping
- MS, mass spectrometry
- PGs, prostaglandins
- PI, propidium iodide
- POBN, α-[4-pyridyl-1-oxide]-N-tert-butyl nitrone
- PUFA, polyunsaturated fatty acid
- SPE, solid phase extraction
- TBS, Tris buffered saline
- TIC, total ion chromatogram
Collapse
Affiliation(s)
- Yi Xu
- Department of Pharmaceutical Sciences, College of Pharmacy, Nursing and Allied Sciences, North Dakota State University, Fargo, ND 58108, USA
| | - Jin Qi
- Department of Pharmaceutical Sciences, College of Pharmacy, Nursing and Allied Sciences, North Dakota State University, Fargo, ND 58108, USA ; Department of Complex Prescription of TCM, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Xiaoyu Yang
- Department of Pharmaceutical Sciences, College of Pharmacy, Nursing and Allied Sciences, North Dakota State University, Fargo, ND 58108, USA
| | - Erxi Wu
- Department of Pharmaceutical Sciences, College of Pharmacy, Nursing and Allied Sciences, North Dakota State University, Fargo, ND 58108, USA
| | - Steven Y Qian
- Department of Pharmaceutical Sciences, College of Pharmacy, Nursing and Allied Sciences, North Dakota State University, Fargo, ND 58108, USA
| |
Collapse
|
164
|
Gorrini C, Harris IS, Mak TW. Modulation of oxidative stress as an anticancer strategy. Nat Rev Drug Discov 2014; 12:931-47. [PMID: 24287781 DOI: 10.1038/nrd4002] [Citation(s) in RCA: 2576] [Impact Index Per Article: 234.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The regulation of oxidative stress is an important factor in both tumour development and responses to anticancer therapies. Many signalling pathways that are linked to tumorigenesis can also regulate the metabolism of reactive oxygen species (ROS) through direct or indirect mechanisms. High ROS levels are generally detrimental to cells, and the redox status of cancer cells usually differs from that of normal cells. Because of metabolic and signalling aberrations, cancer cells exhibit elevated ROS levels. The observation that this is balanced by an increased antioxidant capacity suggests that high ROS levels may constitute a barrier to tumorigenesis. However, ROS can also promote tumour formation by inducing DNA mutations and pro-oncogenic signalling pathways. These contradictory effects have important implications for potential anticancer strategies that aim to modulate levels of ROS. In this Review, we address the controversial role of ROS in tumour development and in responses to anticancer therapies, and elaborate on the idea that targeting the antioxidant capacity of tumour cells can have a positive therapeutic impact.
Collapse
Affiliation(s)
- Chiara Gorrini
- 1] The Campbell Family Institute for Breast Cancer Research, University Health Network, 620 University Avenue, Toronto, Ontario M5G 2C1, Canada. [2]
| | | | | |
Collapse
|
165
|
Das R, Bhattacharya K, Sarkar S, Samanta SK, Pal BC, Mandal C. Mahanine synergistically enhances cytotoxicity of 5-fluorouracil through ROS-mediated activation of PTEN and p53/p73 in colon carcinoma. Apoptosis 2014; 19:149-64. [PMID: 24052409 DOI: 10.1007/s10495-013-0907-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
5-Fluorouracil (5-FU) alone or in combination with other drugs is the main basis of chemotherapeutic treatment in colorectal cancer although patients with microsatellite instability generally show resistance to 5-FU treatment. The present investigation is focussed on the mechanistic insight of a pure herbal carbazole alkaloid, mahanine, as a single or in combination with 5-FU in colon cancer. We demonstrated that mahanine-induced apoptosis involved reactive oxygen species (ROS)-mediated nuclear accumulation of PTEN and its interaction with p53/p73. Mahanine and 5-FU in combination exerted synergistic inhibitory effect on cell viability. This combination also enhanced ROS production, increased tumour suppressor proteins and suppressed chemo-migration. Taken together, our results revealed that mahanine can be a potential chemotherapeutic agent with efficacy to reduce the concentration of toxic 5-FU in colon cancer.
Collapse
Affiliation(s)
- Ranjita Das
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Kolkata, 700032, India
| | | | | | | | | | | |
Collapse
|
166
|
Romero-Canelón I, Sadler PJ. Next-Generation Metal Anticancer Complexes: Multitargeting via Redox Modulation. Inorg Chem 2013; 52:12276-91. [PMID: 23879584 DOI: 10.1021/ic400835n] [Citation(s) in RCA: 307] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
| | - Peter J. Sadler
- Department of Chemistry, University of Warwick, Coventry CV4 7AL, U.K
- Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong,
People’s Republic of China
| |
Collapse
|
167
|
Courtin A, Richards FM, Bapiro TE, Bramhall JL, Neesse A, Cook N, Krippendorff BF, Tuveson DA, Jodrell DI. Anti-tumour efficacy of capecitabine in a genetically engineered mouse model of pancreatic cancer. PLoS One 2013; 8:e67330. [PMID: 23840665 PMCID: PMC3696095 DOI: 10.1371/journal.pone.0067330] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 05/16/2013] [Indexed: 12/17/2022] Open
Abstract
Capecitabine (CAP) is a 5-FU pro-drug approved for the treatment of several cancers and it is used in combination with gemcitabine (GEM) in the treatment of patients with pancreatic adenocarcinoma (PDAC). However, limited pre-clinical data of the effects of CAP in PDAC are available to support the use of the GEMCAP combination in clinic. Therefore, we investigated the pharmacokinetics and the efficacy of CAP as a single agent first and then in combination with GEM to assess the utility of the GEMCAP therapy in clinic. Using a model of spontaneous PDAC occurring in Kras(G12D); p53(R172H); Pdx1-Cre (KPC) mice and subcutaneous allografts of a KPC PDAC-derived cell line (K8484), we showed that CAP achieved tumour concentrations (∼25 µM) of 5-FU in both models, as a single agent, and induced survival similar to GEM in KPC mice, suggesting similar efficacy. In vitro studies performed in K8484 cells as well as in human pancreatic cell lines showed an additive effect of the GEMCAP combination however, it increased toxicity in vivo and no benefit of a tolerable GEMCAP combination was identified in the allograft model when compared to GEM alone. Our work provides pre-clinical evidence of 5-FU delivery to tumours and anti-tumour efficacy following oral CAP administration that was similar to effects of GEM. Nevertheless, the GEMCAP combination does not improve the therapeutic index compared to GEM alone. These data suggest that CAP could be considered as an alternative to GEM in future, rationally designed, combination treatment strategies for advanced pancreatic cancer.
Collapse
Affiliation(s)
- Aurélie Courtin
- Pharmacology and Drug Development Group, Cancer Research UK Cambridge Research Institute, Cambridge, United Kingdom
- University of Cambridge Department of Oncology, Cambridge, United Kingdom, Cambridge, United Kingdom
| | - Frances M. Richards
- Pharmacology and Drug Development Group, Cancer Research UK Cambridge Research Institute, Cambridge, United Kingdom
- University of Cambridge Department of Oncology, Cambridge, United Kingdom, Cambridge, United Kingdom
| | - Tashinga E. Bapiro
- Pharmacology and Drug Development Group, Cancer Research UK Cambridge Research Institute, Cambridge, United Kingdom
- University of Cambridge Department of Oncology, Cambridge, United Kingdom, Cambridge, United Kingdom
| | - Jo L. Bramhall
- Pharmacology and Drug Development Group, Cancer Research UK Cambridge Research Institute, Cambridge, United Kingdom
- University of Cambridge Department of Oncology, Cambridge, United Kingdom, Cambridge, United Kingdom
| | - Albrecht Neesse
- Tumour Modelling and Experimental Medicine Group, Cancer Research UK Cambridge Research Institute, Cambridge, United Kingdom
- University of Cambridge Department of Oncology, Cambridge, United Kingdom, Cambridge, United Kingdom
| | - Natalie Cook
- Tumour Modelling and Experimental Medicine Group, Cancer Research UK Cambridge Research Institute, Cambridge, United Kingdom
- University of Cambridge Department of Oncology, Cambridge, United Kingdom, Cambridge, United Kingdom
| | - Ben-Fillippo Krippendorff
- Pharmacology and Drug Development Group, Cancer Research UK Cambridge Research Institute, Cambridge, United Kingdom
- University of Cambridge Department of Oncology, Cambridge, United Kingdom, Cambridge, United Kingdom
| | - David A. Tuveson
- Tumour Modelling and Experimental Medicine Group, Cancer Research UK Cambridge Research Institute, Cambridge, United Kingdom
- University of Cambridge Department of Oncology, Cambridge, United Kingdom, Cambridge, United Kingdom
| | - Duncan I. Jodrell
- Pharmacology and Drug Development Group, Cancer Research UK Cambridge Research Institute, Cambridge, United Kingdom
- University of Cambridge Department of Oncology, Cambridge, United Kingdom, Cambridge, United Kingdom
| |
Collapse
|
168
|
Zhou S, Ye W, Duan X, Zhang M, Wang J. The noncytotoxic dose of sorafenib sensitizes Bel-7402/5-FU cells to 5-FU by down-regulating 5-FU-induced Nrf2 expression. Dig Dis Sci 2013; 58:1615-26. [PMID: 23314856 DOI: 10.1007/s10620-012-2537-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2012] [Accepted: 12/20/2012] [Indexed: 01/11/2023]
Abstract
BACKGROUND Acquired resistance to 5-fluorouracil (5-FU) is a serious therapeutic obstacle in advanced hepatocellular carcinoma (HCC) patients. AIM To investigate whether nuclear factor erythroid 2-related factor 2 (Nrf2) was associated with drug resistance in 5-FU resistant Bel-7402 (Bel-7402/5-FU) cells, and if sorafenib, an oral multikinase inhibitor targeting the tumor and vasculature, could reverse drug resistance in Bel-7402/5-FU cells at the noncytotoxic dosage. METHODS We used MTT to detect the resistance reversal activity of sorafenib, compared Nrf2 expression in various conditions by western blot and qRT-PCR, and analyzed subcellular localization of Nrf2 by immunofluorescence. RESULTS The endogenous expression of Nrf2 in Bel-7402/5-FU cells was similar to that observed in Bel-7402 cells. However, Nrf2 expression levels were increased by 5-FU treatment in Bel-7402/5-FU cells higher than that in Bel-7402 cells, which is to highlight the Nrf2 contribution to the enhanced resistance of Bel-7402/5-FU cells to 5-FU. Moreover, intracellular Nrf2 protein level was significantly down-regulated by Nrf2-shRNA in Bel-7402/5-FU cells, resulting in partial reversal of 5-FU resistance. Sorafenib down-regulated the increased expression of Nrf2 induced by 5-FU treatment and partly reversed 5-FU resistance in Bel-7402/5-FU cells. CONCLUSIONS These results suggested that more sensitive cell defense mediated by Nrf2 was associated with drug resistance of Bel-7402/5-FU cells. Sorafenib reversed drug resistance, and its reversal mechanism might be due to the suppression of Nrf2 expression induced by 5-FU, indicating the feasibility of using Nrf2 inhibitors to increase efficacy of chemotherapeutic drugs in HCC patients.
Collapse
MESH Headings
- Antimetabolites, Antineoplastic/pharmacology
- Antimetabolites, Antineoplastic/therapeutic use
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Biomarkers, Tumor/metabolism
- Blotting, Western
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/metabolism
- Cell Line, Tumor
- Dose-Response Relationship, Drug
- Down-Regulation
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/physiology
- Fluorescent Antibody Technique
- Fluorouracil/pharmacology
- Fluorouracil/therapeutic use
- Humans
- Liver Neoplasms/drug therapy
- Liver Neoplasms/metabolism
- Multidrug Resistance-Associated Proteins/metabolism
- NF-E2-Related Factor 2/metabolism
- Niacinamide/analogs & derivatives
- Niacinamide/pharmacology
- Niacinamide/therapeutic use
- Phenylurea Compounds/pharmacology
- Phenylurea Compounds/therapeutic use
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Reverse Transcriptase Polymerase Chain Reaction
- Sorafenib
Collapse
Affiliation(s)
- Suna Zhou
- Department of Thoracic Oncology, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Yanta West Road No. 277, Shaanxi, 710061, China
| | | | | | | | | |
Collapse
|
169
|
Identification of lung-cancer-related genes with the shortest path approach in a protein-protein interaction network. BIOMED RESEARCH INTERNATIONAL 2013; 2013:267375. [PMID: 23762832 PMCID: PMC3674655 DOI: 10.1155/2013/267375] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Revised: 04/19/2013] [Accepted: 04/29/2013] [Indexed: 12/30/2022]
Abstract
Lung cancer is one of the leading causes of cancer mortality worldwide. The main types of lung cancer are small cell lung cancer (SCLC) and nonsmall cell lung cancer (NSCLC). In this work, a computational method was proposed for identifying lung-cancer-related genes with a shortest path approach in a protein-protein interaction (PPI) network. Based on the PPI data from STRING, a weighted PPI network was constructed. 54 NSCLC- and 84 SCLC-related genes were retrieved from associated KEGG pathways. Then the shortest paths between each pair of these 54 NSCLC genes and 84 SCLC genes were obtained with Dijkstra's algorithm. Finally, all the genes on the shortest paths were extracted, and 25 and 38 shortest genes with a permutation P value less than 0.05 for NSCLC and SCLC were selected for further analysis. Some of the shortest path genes have been reported to be related to lung cancer. Intriguingly, the candidate genes we identified from the PPI network contained more cancer genes than those identified from the gene expression profiles. Furthermore, these genes possessed more functional similarity with the known cancer genes than those identified from the gene expression profiles. This study proved the efficiency of the proposed method and showed promising results.
Collapse
|
170
|
Govender R, Phulukdaree A, Gengan RM, Anand K, Chuturgoon AA. Silver nanoparticles of Albizia adianthifolia: the induction of apoptosis in human lung carcinoma cell line. J Nanobiotechnology 2013; 11:5. [PMID: 23418790 PMCID: PMC3606379 DOI: 10.1186/1477-3155-11-5] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 01/30/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Silver nanoparticles (AgNP), the most popular nano-compounds, possess unique properties. Albizia adianthifolia (AA) is a plant of the Fabaceae family that is rich in saponins. The biological properties of a novel AgNP, synthesized from an aqueous leaf extract of AA (AA(AgNP)), were investigated on A549 lung cells. Cell viability was determined by the MTT assay. Cellular oxidative status (lipid peroxidation and glutathione (GSH) levels), ATP concentration, caspase-3/-7, -8 and -9 activities were determined. Apoptosis, mitochondrial (mt) membrane depolarization (flow cytometry) and DNA fragmentation (comet assay) were assessed. The expression of CD95 receptors, p53, bax, PARP-1 and smac/DIABLO was evaluated by flow cytometry and/or western blotting. RESULTS Silver nanoparticles of AA caused a dose-dependent decrease in cell viability with a significant increase in lipid peroxidation (5-fold vs. control; p = 0.0098) and decreased intracellular GSH (p = 0.1184). A significant 2.5-fold decrease in cellular ATP was observed upon AA(AgNP) exposure (p = 0.0040) with a highly significant elevation in mt depolarization (3.3-fold vs. control; p < 0.0001). Apoptosis was also significantly higher (1.5-fold) in AA(AgNP) treated cells (p < 0.0001) with a significant decline in expression of CD95 receptors (p = 0.0416). Silver nanoparticles of AA caused a significant 2.5-fold reduction in caspase-8 activity (p = 0.0024) with contrasting increases in caspase-3/-7 (1.7-fold vs. control; p = 0.0180) and -9 activity (1.4-fold vs. control; p = 0.0117). Western blots showed increased expression of smac/DIABLO (4.1-fold) in treated cells (p = 0.0033). Furthermore, AA(AgNP) significantly increased the expression of p53, bax and PARP-1 (1.2-fold; p = 0.0498, 1.6-fold; p = 0.0083 and 1.1-fold; p = 0.0359 respectively). CONCLUSION Data suggests that AA(AgNP) induces cell death in the A549 lung cells via the mt mediated intrinsic apoptotic program. Further investigation is required to potentiate the use of this novel compound in cancer therapy trials.
Collapse
Affiliation(s)
- Rishalan Govender
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Private Bag 7, Congella, Durban, 4013, South Africa
| | | | | | | | | |
Collapse
|
171
|
Govender R, Phulukdaree A, Gengan RM, Anand K, Chuturgoon AA. Silver nanoparticles of Albizia adianthifolia: the induction of apoptosis in human lung carcinoma cell line. J Nanobiotechnology 2013. [PMID: 23418790 DOI: 10.1186/1477–3155–11–5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Silver nanoparticles (AgNP), the most popular nano-compounds, possess unique properties. Albizia adianthifolia (AA) is a plant of the Fabaceae family that is rich in saponins. The biological properties of a novel AgNP, synthesized from an aqueous leaf extract of AA (AA(AgNP)), were investigated on A549 lung cells. Cell viability was determined by the MTT assay. Cellular oxidative status (lipid peroxidation and glutathione (GSH) levels), ATP concentration, caspase-3/-7, -8 and -9 activities were determined. Apoptosis, mitochondrial (mt) membrane depolarization (flow cytometry) and DNA fragmentation (comet assay) were assessed. The expression of CD95 receptors, p53, bax, PARP-1 and smac/DIABLO was evaluated by flow cytometry and/or western blotting. RESULTS Silver nanoparticles of AA caused a dose-dependent decrease in cell viability with a significant increase in lipid peroxidation (5-fold vs. control; p = 0.0098) and decreased intracellular GSH (p = 0.1184). A significant 2.5-fold decrease in cellular ATP was observed upon AA(AgNP) exposure (p = 0.0040) with a highly significant elevation in mt depolarization (3.3-fold vs. control; p < 0.0001). Apoptosis was also significantly higher (1.5-fold) in AA(AgNP) treated cells (p < 0.0001) with a significant decline in expression of CD95 receptors (p = 0.0416). Silver nanoparticles of AA caused a significant 2.5-fold reduction in caspase-8 activity (p = 0.0024) with contrasting increases in caspase-3/-7 (1.7-fold vs. control; p = 0.0180) and -9 activity (1.4-fold vs. control; p = 0.0117). Western blots showed increased expression of smac/DIABLO (4.1-fold) in treated cells (p = 0.0033). Furthermore, AA(AgNP) significantly increased the expression of p53, bax and PARP-1 (1.2-fold; p = 0.0498, 1.6-fold; p = 0.0083 and 1.1-fold; p = 0.0359 respectively). CONCLUSION Data suggests that AA(AgNP) induces cell death in the A549 lung cells via the mt mediated intrinsic apoptotic program. Further investigation is required to potentiate the use of this novel compound in cancer therapy trials.
Collapse
Affiliation(s)
- Rishalan Govender
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Private Bag 7, Congella, Durban, 4013, South Africa
| | | | | | | | | |
Collapse
|
172
|
Wang H, Ao M, Wu J, Yu L. TNFα and Fas/FasL pathways are involved in 9-Methoxycamptothecin-induced apoptosis in cancer cells with oxidative stress and G2/M cell cycle arrest. Food Chem Toxicol 2013; 55:396-410. [PMID: 23369935 DOI: 10.1016/j.fct.2012.12.059] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Revised: 12/11/2012] [Accepted: 12/12/2012] [Indexed: 11/25/2022]
Abstract
9-Methoxycamptothecin (MCPT) has been recently reported to have a strong anticancer activity. However, its detailed mechanism of action in human cancer cells has not been well clarified. The results showed that MCPT induced cytotoxicity in seven human cancer cell lines in a dose dependent manner after 72h, with A2780 and Hela cell lines more sensitive, so the two cell lines were chosen to do further studies. MCPT induced strong G2/M arrest in both A2780 cells and Hela cells after 24h, following by substantial sub-G1 arrest (indicating apoptosis). The apoptosis was verified by staining with Annexin V-FITC and propidium iodide. ROS generation increased significantly in MCPT-induced apoptosis. Meanwhile, the apoptosis appeared to be dependent on caspase-3, -8 and -9 in A2780 cells, and caspase-3 in Hela cells. In addition, MCPT induced up-regulation expression of most of seventeen genes in both cell lines. Western blot verified that changes of TNFα, Fas, P53 and P27 protein level were consistent with their gene expression changes. Taken together, MCPT plays an important role in tumor growth suppression by inducing apoptosis in both cell lines via extrinsic and intrinsic apoptotic pathways, and has the potential to be developed into an antitumor agent.
Collapse
Affiliation(s)
- Haiyan Wang
- Institute of Resource Biology and Biotechnology, Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | | | | | | |
Collapse
|
173
|
Helal M, Das U, Bandy B, Islam A, Nazarali AJ, Dimmock JR. Mitochondrial dysfunction contributes to the cytotoxicity of some 3,5-bis(benzylidene)-4-piperidone derivatives in colon HCT-116 cells. Bioorg Med Chem Lett 2012; 23:1075-8. [PMID: 23305919 DOI: 10.1016/j.bmcl.2012.12.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Revised: 11/27/2012] [Accepted: 12/10/2012] [Indexed: 10/27/2022]
Abstract
The objectives of this study are to investigate the possible ways by which the curcumin analogs 2a and 2b exert their antiproliferative properties. The analogs 2a and 2b have submicromolar IC(50) values towards human HCT-116 colon cancer cells but are far less toxic to human non-malignant CRL-1790 colon cells. Both compounds affected a number of mitochondrial functions in HCT-116 cells namely increasing the intracellular concentrations of reactive oxygen species, inhibiting oxygen consumption and decreasing the mitochondrial membrane potential. These molecules also produced swelling of isolated rat liver mitochondria, supporting a mitochondrial mechanism of cytotoxicity. Both compounds reacted with glutathione in the presence of glutathione S-transferase π and hence they may be classified as thiol alkylators.
Collapse
Affiliation(s)
- Muath Helal
- Drug Design and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Saskatchewan, Canada S7N 5C9
| | | | | | | | | | | |
Collapse
|
174
|
Latino-Martel P, Bachman P. Nutrition chez le patient adulte atteint de cancer : compléments alimentaires antioxydants pendant et au décours du traitement des cancers. NUTR CLIN METAB 2012. [DOI: 10.1016/j.nupar.2012.10.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
175
|
Agrawal M, Gadgil M. Meta analysis of gene expression changes upon treatment of A549 cells with anti-cancer drugs to identify universal responses. Comput Biol Med 2012; 42:1141-9. [PMID: 23063289 DOI: 10.1016/j.compbiomed.2012.09.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Revised: 08/29/2012] [Accepted: 09/11/2012] [Indexed: 11/26/2022]
Abstract
A meta-analysis of publicly available gene expression changes in A549 cells upon treatment with anti-cancer drugs is reported. To reduce false positives, both fold-change and significance level cutoffs were used. Simulated datasets and permutation analysis were used to guide choice of ratio cutoff. Of the genes identified, FDXR is the only gene differentially expressed in six of the seven drug treatments. Though FDXR has been reported to be differentially expressed upon treatment with 5-fluorouracil and its expression correlated to long term disease survival, to our knowledge this is a first study implicating a wide effect of anti-cancer drug treatment on FDXR expression. The other genes identified which are differentially expressed in four out of the seven drug treatments are CDKN1A and PARVB which are upregulated and MYC, HBP1, LDLR, SIM2, ALX1 and GPHN which are downregulated.
Collapse
Affiliation(s)
- Megha Agrawal
- Chemical Engineering and Process Development Division, CSIR-National Chemical Laboratory, Pune 411008, India
| | | |
Collapse
|
176
|
Tomita K, Teratani T, Suzuki T, Oshikawa T, Yokoyama H, Shimamura K, Nishiyama K, Mataki N, Irie R, Minamino T, Okada Y, Kurihara C, Ebinuma H, Saito H, Shimizu I, Yoshida Y, Hokari R, Sugiyama K, Hatsuse K, Yamamoto J, Kanai T, Miura S, Hibi T. p53/p66Shc-mediated signaling contributes to the progression of non-alcoholic steatohepatitis in humans and mice. J Hepatol 2012; 57:837-43. [PMID: 22641095 DOI: 10.1016/j.jhep.2012.05.013] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Revised: 05/14/2012] [Accepted: 05/21/2012] [Indexed: 12/25/2022]
Abstract
BACKGROUND & AIMS The tumor suppressor p53 is a primary sensor of stressful stimuli, controlling a number of biologic processes. The aim of our study was to examine the roles of p53 in non-alcoholic steatohepatitis (NASH). METHODS Male wild type and p53-deficient mice were fed a methionine- and choline-deficient diet for 8 weeks to induce nutritional steatohepatitis. mRNA expression profiles in normal liver samples and liver samples from patients with non-alcoholic liver disease (NAFLD) were also evaluated. RESULTS Hepatic p53 and p66Shc signaling was enhanced in the mouse NASH model. p53 deficiency suppressed the enhanced p66Shc signaling, decreased hepatic lipid peroxidation and the number of apoptotic hepatocytes, and ameliorated progression of nutritional steatohepatitis. In primary cultured hepatocytes, transforming growth factor (TGF)-β treatment increased p53 and p66Shc signaling, leading to exaggerated reactive oxygen species (ROS) accumulation and apoptosis. Deficient p53 signaling inhibited TGF-β-induced p66Shc signaling, ROS accumulation, and hepatocyte apoptosis. Furthermore, expression levels of p53, p21, and p66Shc were significantly elevated in human NAFLD liver samples, compared with results obtained with normal liver samples. Among NAFLD patients, those with NASH had significantly higher hepatic expression levels of p53, p21, and p66Shc compared with the group with simple steatosis. A significant correlation between expression levels of p53 and p66Shc was observed. CONCLUSIONS p53 in hepatocytes regulates steatohepatitis progression by controlling p66Shc signaling, ROS levels, and apoptosis, all of which may be regulated by TGF-β. Moreover, p53/p66Shc signaling in the liver appears to be a promising target for the treatment of NASH.
Collapse
Affiliation(s)
- Kengo Tomita
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa-shi, Saitama 359-8513, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
177
|
Zhuang J, Ma W, Lago CU, Hwang PM. Metabolic regulation of oxygen and redox homeostasis by p53: lessons from evolutionary biology? Free Radic Biol Med 2012; 53:1279-85. [PMID: 22841759 PMCID: PMC3444283 DOI: 10.1016/j.freeradbiomed.2012.07.026] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Revised: 07/20/2012] [Accepted: 07/20/2012] [Indexed: 12/31/2022]
Abstract
The genetic links between p53 and metabolic processes such as oxidative phosphorylation are being studied with increasing interest given that cellular metabolism seems to play an important role in tumorigenesis. This review focuses on how p53 regulation of various metabolic genes may influence redox homeostasis, as the genome is constantly susceptible to oxidative damage, a consequence of living in an aerobic environment. Because p53-like genetic sequences are also found in life forms that may not necessarily benefit from tumor suppression, an evolutionary introduction is given in an attempt to understand why p53 might regulate a basic cellular activity such as metabolism. The presented epidemiologic and experimental data suggest that one reason may be for the homeostatic regulation of oxygen, the essential substrate for reactive oxygen species generation.
Collapse
Affiliation(s)
- Jie Zhuang
- Center for Molecular Medicine, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|
178
|
Harris DR, Mims A, Bunz F. Genetic disruption of USP9X sensitizes colorectal cancer cells to 5-fluorouracil. Cancer Biol Ther 2012; 13:1319-24. [PMID: 22895071 DOI: 10.4161/cbt.21792] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The X-linked deubiquitinase USP9X affects the stability and activity of numerous regulatory proteins that influence cell survival. Recent studies suggest that decreased USP9X expression can confer a selective advantage onto developing cancer cells and thereby promotes disease progression. To examine the effect of USP9X on the cellular responses to anticancer therapies, we derived cancer cell lines in which the USP9X locus was disrupted by homologous recombination. The resulting USP9X-deficient cancer cells exhibited increased activation of apoptotic pathways and markedly decreased clonogenic survival in response to 5-fluorouracil, a chemotherapeutic drug that is widely used for treatment of gastrointestinal malignancies. These unexpected results suggest that cancers with low USP9X expression might be specifically sensitized to some conventional therapeutic agents.
Collapse
Affiliation(s)
- Dennis R Harris
- Department of Radiation Oncology and Molecular Radiation Sciences, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | |
Collapse
|
179
|
|
180
|
Wei J, Liu H, Liu M, Wu N, Zhao J, Xiao L, Han L, Chu E, Lin X. Oleanolic acid potentiates the antitumor activity of 5-fluorouracil in pancreatic cancer cells. Oncol Rep 2012; 28:1339-45. [PMID: 22825128 DOI: 10.3892/or.2012.1921] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Accepted: 04/26/2012] [Indexed: 11/06/2022] Open
Abstract
The antitumor activity of oleanolic (OA) has attracted attention due to its marked antitumor effects and pharmacological safety. In the present study, the effects of the combination of OA and 5-fluorouracil (5-FU) on Panc-28 human pancreatic cells were studied. The results showed that combined use of OA and 5-FU synergistically potentiated cell death effects on Panc-28 cells, and the pro-apoptotic effects were also increased. Further study revealed that the combined treatment could enhance mitochondrial depolarization, lysosomal membrane permeabilization (LMP) and leakage of cathepin D, while the release of cytochrome C did not display significant changes. The expression of apoptosis related proteins was also affected in cells treated with the combination of OA and 5-FU, including activation of caspases-3 and the expression of Bcl-2/Bax, survivin and NF-κB. Our results provide evidence that combination of OA and 5-FU may serve as a novel strategy for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Jianteng Wei
- Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
181
|
Nakvasina MA, Trubicina MS, Solovieva EV, Artyukhov VG. Ways of apoptosis development in human lymphocytes, induced by UV-irradiation. Biophysics (Nagoya-shi) 2012. [DOI: 10.1134/s0006350912040124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
182
|
Zou H, Volonte D, Galbiati F. Interaction of caveolin-1 with Ku70 inhibits Bax-mediated apoptosis. PLoS One 2012; 7:e39379. [PMID: 22745744 PMCID: PMC3380016 DOI: 10.1371/journal.pone.0039379] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Accepted: 05/24/2012] [Indexed: 01/29/2023] Open
Abstract
Caveolin-1, the structural protein component of caveolae, acts as a scaffolding protein that functionally regulates signaling molecules. We show that knockdown of caveolin-1 protein expression enhances chemotherapeutic drug-induced apoptosis and inhibits long-term survival of colon cancer cells. In vitro studies demonstrate that caveolin-1 is a novel Ku70-binding protein, as shown by the binding of the scaffolding domain of caveolin-1 (amino acids 82-101) to the caveolin-binding domain (CBD) of Ku70 (amino acids 471-478). Cell culture data show that caveolin-1 binds Ku70 after treatment with chemotherapeutic drugs. Mechanistically, we found that binding of caveolin-1 to Ku70 inhibits the chemotherapeutic drug-induced release of Bax from Ku70, activation of Bax, translocation of Bax to mitochondria and apoptosis. Potentiation of apoptosis by knockdown of caveolin-1 protein expression is greatly reduced in the absence of Bax expression. Finally, we found that overexpression of wild type Ku70, but not a mutant form of Ku70 that cannot bind to caveolin-1 (Ku70 Φ→A), limits the chemotherapeutic drug-induced Ku70/Bax dissociation and apoptosis. Thus, caveolin-1 acts as an anti-apoptotic protein in colon cancer cells by binding to Ku70 and inhibiting Bax-dependent cell death.
Collapse
Affiliation(s)
- Huafei Zou
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | | | | |
Collapse
|
183
|
Connexin-43 prevents hematopoietic stem cell senescence through transfer of reactive oxygen species to bone marrow stromal cells. Proc Natl Acad Sci U S A 2012; 109:9071-6. [PMID: 22611193 DOI: 10.1073/pnas.1120358109] [Citation(s) in RCA: 122] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Hematopoietic stem cell (HSC) aging has become a concern in chemotherapy of older patients. Humoral and paracrine signals from the bone marrow (BM) hematopoietic microenvironment (HM) control HSC activity during regenerative hematopoiesis. Connexin-43 (Cx43), a connexin constituent of gap junctions (GJs) is expressed in HSCs, down-regulated during differentiation, and postulated to be a self-renewal gene. Our studies, however, reveal that hematopoietic-specific Cx43 deficiency does not result in significant long-term competitive repopulation deficiency. Instead, hematopoietic Cx43 (H-Cx43) deficiency delays hematopoietic recovery after myeloablation with 5-fluorouracil (5-FU). 5-FU-treated H-Cx43-deficient HSC and progenitors (HSC/P) cells display decreased survival and fail to enter the cell cycle to proliferate. Cell cycle quiescence is associated with down-regulation of cyclin D1, up-regulation of the cyclin-dependent kinase inhibitors, p21(cip1.) and p16(INK4a), and Forkhead transcriptional factor 1 (Foxo1), and activation of p38 mitogen-activated protein kinase (MAPK), indicating that H-Cx43-deficient HSCs are prone to senescence. The mechanism of increased senescence in H-Cx43-deficient HSC/P cells depends on their inability to transfer reactive oxygen species (ROS) to the HM, leading to accumulation of ROS within HSCs. In vivo antioxidant administration prevents the defective hematopoietic regeneration, as well as exogenous expression of Cx43 in HSC/P cells. Furthermore, ROS transfer from HSC/P cells to BM stromal cells is also rescued by reexpression of Cx43 in HSC/P. Finally, the deficiency of Cx43 in the HM phenocopies the hematopoietic defect in vivo. These results indicate that Cx43 exerts a protective role and regulates the HSC/P ROS content through ROS transfer to the HM, resulting in HSC protection during stress hematopoietic regeneration.
Collapse
|
184
|
Kim KK, Lange TS, Singh RK, Brard L, Moore RG. Tetrathiomolybdate sensitizes ovarian cancer cells to anticancer drugs doxorubicin, fenretinide, 5-fluorouracil and mitomycin C. BMC Cancer 2012; 12:147. [PMID: 22502731 PMCID: PMC3353246 DOI: 10.1186/1471-2407-12-147] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Accepted: 04/13/2012] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Our recent study showed that tetrathiomolybdate (TM), a drug to treat copper overload disorders, can sensitize drug-resistant endometrial cancer cells to reactive oxygen species (ROS)-generating anticancer drug doxorubicin. To expand these findings in the present study we explore TM efficacy in combination with a spectrum of ROS-generating anticancer drugs including mitomycin C, fenretinide, 5-fluorouracil and doxorubicin in ovarian cancer cells as a model system. METHODS The effects of TM alone or in combination with doxorubicin, mitomycin C, fenretinide, or 5-fluorouracil were evaluated using a sulforhodamine B assay. Flow cytometry was used to detect the induction of apoptosis and ROS generation. Immunoblot analysis was carried out to investigate changes in signaling pathways. RESULTS TM potentiated doxorubicin-induced cytotoxicity and modulated key regulators of apoptosis (PARP, caspases, JNK and p38 MAPK) in SKOV-3 and A2780 ovarian cancer cell lines. These effects were linked to the increased production of ROS, as shown in SKOV-3 cells. ROS scavenging by ascorbic acid blocked the sensitization of cells by TM. TM also sensitized SKOV-3 to mitomycin C, fenretinide, and 5-fluorouracil. The increased cytotoxicity of these drugs in combination with TM was correlated with the activity of ROS, loss of a pro-survival factor (e.g. XIAP) and the appearance of a pro-apoptotic marker (e.g. PARP cleavage). CONCLUSIONS Our data show that TM increases the efficacy of various anticancer drugs in ovarian cancer cells in a ROS-dependent manner.
Collapse
Affiliation(s)
- Kyu Kwang Kim
- Molecular Therapeutics Laboratory, Program in Women's Oncology, Department of Obstetrics and Gynecology, Women and Infants Hospital, Alpert Medical School, Brown University, Providence, RI 02905, USA.
| | | | | | | | | |
Collapse
|
185
|
Abstract
Chronic alcohol exposure inhibits insulin and insulin-like growth factor signaling in the liver and brain by impairing the signaling cascade at multiple levels. These alterations produced by alcohol cause severe hepatic and central nervous system insulin resistance as the cells fail to adequately transmit signals downstream through Erk/mitogen-activated protein kinase (MAPK), which is needed for DNA synthesis and liver regeneration, and phosphatidylinositol 3-kinase (PI3K), which promotes growth, survival, cell motility, glucose utilization, plasticity, and energy metabolism. The robust inhibition of insulin signaling in liver and brain is augmented by additional factors involving the activation of phosphatases such as phosphatase and tensin homologue (PTEN), which further impairs insulin signaling through PI3K/Akt. Thus, intact insulin signaling is important for neuronal survival. Chronic alcohol consumption produces steatohepatitis, which also promotes hepatic insulin resistance, oxidative stress and injury, with the attendant increased generation of "toxic lipids" such as ceramides that increase insulin resistance. The PI3K/Akt signaling cascade is altered by direct interaction with ceramides as well as through PTEN upregulation as a downstream target gene of enhanced p53 transcriptional activity. Cytotoxic ceramides transferred from the liver to the blood can enter the brain due to their lipid-soluble nature, and thereby exert neurodegenerative effects via a liver-brain axis. We postulate that the neurotoxic and neurodegenerative effects of liver-derived ceramides activate pro-inflammatory cytokines and increase lipid adducts and insulin resistance in the brain to impair cognitive and motor function. These observations are discussed in the context of insulin sensitizers as potential cytoprotective agents against liver and brain injury induced by alcohol.
Collapse
MESH Headings
- Alcohol-Induced Disorders, Nervous System/etiology
- Alcohol-Induced Disorders, Nervous System/metabolism
- Alcohol-Induced Disorders, Nervous System/pathology
- Alcohol-Induced Disorders, Nervous System/physiopathology
- Alcoholism/complications
- Alcoholism/drug therapy
- Alcoholism/metabolism
- Alcoholism/pathology
- Alcoholism/physiopathology
- Animals
- Brain/metabolism
- Brain/pathology
- Brain/physiopathology
- DNA Damage
- Fatty Liver, Alcoholic/etiology
- Fatty Liver, Alcoholic/metabolism
- Humans
- Insulin/metabolism
- Insulin Resistance
- Liver/drug effects
- Liver/metabolism
- Liver/pathology
- Liver/physiopathology
- Liver Diseases, Alcoholic/drug therapy
- Liver Diseases, Alcoholic/etiology
- Liver Diseases, Alcoholic/metabolism
- Liver Diseases, Alcoholic/pathology
- Liver Diseases, Alcoholic/physiopathology
- Liver Regeneration
- PPAR gamma/agonists
- PPAR gamma/metabolism
- PTEN Phosphohydrolase/metabolism
- Receptor, IGF Type 1/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Suzanne de la Monte
- Department of Pathology, Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | | | | |
Collapse
|
186
|
Nagel S, Papadakis M, Pfleger K, Grond-Ginsbach C, Buchan AM, Wagner S. Microarray analysis of the global gene expression profile following hypothermia and transient focal cerebral ischemia. Neuroscience 2012; 208:109-22. [PMID: 22366221 DOI: 10.1016/j.neuroscience.2012.01.048] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Revised: 01/26/2012] [Accepted: 01/30/2012] [Indexed: 01/09/2023]
Abstract
BACKGROUND Hypothermia is one of the most robust experimental neuroprotective interventions against cerebral ischemia. Identification of molecular pathways and gene networks together with single genes or gene families that are significantly associated with neuroprotection might help unravel the mechanisms of therapeutic hypothermia. MATERIAL AND METHODS We performed a microarray analysis of ischemic rat brains that underwent 90 min of middle cerebral artery occlusion (MCAO) and 48 h of reperfusion. Hypothermia was induced for 4 h, starting 1 h after MCAO in male Wistar rats. At 48 h, magnetic resonance imaging (MRI) was performed for infarct volumetry, and functional outcome was determined by a neuroscore. The brain gene expression profile of sham (S), ischemia (I), and ischemia plus hypothermia (HI) treatment were compared by analyzing changes of individual genes, pathways, and networks. Real-time reverse-transcribed polymerase chain reaction (RT-PCR) was performed on selected genes to validate the data. RESULTS Rats treated with HI had significantly reduced infarct volumes and improved neuroscores at 48 h compared with I. Of 4067 genes present on the array chip, HI compared with I upregulated 50 (1.23%) genes and downregulated 103 (3.20%) genes equal or greater than twofold. New genes potentially mediating neuroprotection by hypothermia were HNRNPAB, HIG-1, and JAK3. On the pathway level, HI globally suppressed the ischemia-driven gene response. Twelve gene networks were identified to be significantly altered by HI compared with I. The most significantly altered network contained genes participating in apoptosis suppression. CONCLUSIONS Our data suggest that although hypothermia at the pathway level restored gene expression to sham levels, it selectively regulated the expression of several genes implicated in protein synthesis and folding, calcium homeostasis, cellular and synaptic integrity, inflammation, cell death, and apoptosis.
Collapse
Affiliation(s)
- S Nagel
- Department of Neurology, University of Heidelberg, Heidelberg, Germany.
| | | | | | | | | | | |
Collapse
|
187
|
Abstract
PURPOSE OF REVIEW There is an inverse relationship between cancer incidence and cardiorespiratory fitness in large population studies. Mechanistic insights into these observations may strengthen the rationale for encouraging exercise fitness in the clinics for cancer prevention and may promote the development of new preventive strategies. RECENT FINDINGS Studying the multifaceted activities of p53, a critical tumor suppressor gene, has revealed various cellular pathways necessary for adapting to environmental stresses. Genetic connections are being made between p53 and an increasing number of metabolic activities such as oxidative phosphorylation, glycolysis and fatty acid oxidation. In-vivo mouse models show that p53 plays an important role in determining both basal aerobic exercise capacity and its improvement by training. SUMMARY The genetic pathways by which p53 regulates metabolism and exercise may help explain significant epidemiologic observations connecting cardiorespiratory fitness and cancer. Further understanding of these molecular pathways through human translational studies may promote the development of new cancer preventive strategies.
Collapse
|
188
|
Artyukhov VG, Trubitsina MS, Nakvasina MA, Solov’eva EV, Lidokhova OV. Apoptosis development pathways in human lymphocytes induced by UV light and reactive oxygen species. Biophysics (Nagoya-shi) 2011. [DOI: 10.1134/s0006350911060236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
189
|
Bae YS, Oh H, Rhee SG, Yoo YD. Regulation of reactive oxygen species generation in cell signaling. Mol Cells 2011; 32:491-509. [PMID: 22207195 PMCID: PMC3887685 DOI: 10.1007/s10059-011-0276-3] [Citation(s) in RCA: 467] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Accepted: 12/12/2011] [Indexed: 12/19/2022] Open
Abstract
Reactive oxygen species (ROS) including superoxide anion and hydrogen peroxide (H(2)O(2)) are thought to be byproducts of aerobic respiration with damaging effects on DNA, protein, and lipid. A growing body of evidence indicates, however, that ROS are involved in the maintenance of redox homeostasis and various cellular signaling pathways. ROS are generated from diverse sources including mitochondrial respiratory chain, enzymatic activation of cytochrome p450, and NADPH oxidases further suggesting involvement in a complex array of cellular processes. This review summarizes the production and function of ROS. In particular, how cytosolic and membrane proteins regulate ROS generation for intracellular redox signaling will be detailed.
Collapse
Affiliation(s)
- Yun Soo Bae
- Department of Life Science, Division of Life and Pharmaceutical Sciences, Ewha Womans University, Seoul 120-750, Korea
| | - Hyunjin Oh
- Department of Life Science, Division of Life and Pharmaceutical Sciences, Ewha Womans University, Seoul 120-750, Korea
| | - Sue Goo Rhee
- Department of Life Science, Division of Life and Pharmaceutical Sciences, Ewha Womans University, Seoul 120-750, Korea
| | - Young Do Yoo
- Laboratory of Molecular Cell Biology, Graduate School of Medicine, Korea University College of Medicine, Korea University, Seoul 136-705, Korea
| |
Collapse
|
190
|
Ang FY, Fukuzaki Y, Yamanoha B, Kogure S. Immunocytochemical studies on the effect of 405-nm low-power laser irradiation on human-derived A-172 glioblastoma cells. Lasers Med Sci 2011; 27:935-42. [PMID: 22041845 DOI: 10.1007/s10103-011-1009-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2011] [Accepted: 09/23/2011] [Indexed: 11/28/2022]
Abstract
The application of low-power laser irradiation (LLI) affects the cell cycle and cell proliferation in various kinds of cells. LLI at a wavelength of 808 nm and a power of 30 mW has been found to significantly decrease the proliferation rate of cells of the human-derived glioblastoma cell line A-172. To determine if this effect of LLI is specific to 808-nm LLI, the present study was designed to reveal the effects of 405-nm LLI under the same experimental conditions. A-172 glioblastoma cells were cultured in 96-well plates according to the conventional protocol. Two different schedules of 405-nm LLI (27 mW) were tested: longer periods of 20, 40 and 60 min and shorter periods of 1, 2, 3, 5, 10 and 15 min. Cells on a digital image displayed on a computer monitor were counted and the proliferation ratio was determined using MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide) staining. Annexin-V-FLUOS staining and acridine-orange/ethidium-bromide staining were in an immunocytochemical assay to determine if cells were viable or dead (due to apoptosis or necrosis). Cell counting and MTT staining showed that longer 405-nm LLI significantly suppressed the proliferation of A-172 cells at 48 h after LLI (p < 0.05 or p < 0.01) and that the effect of LLI tended to be dose-dependent with morphological changes including cell death. At 90 min after LLI, shorter 405-nm LLI caused necrotic as well as apoptotic cell death, and these effects depended on irradiation time, power and energy density. Detailed analysis revealed that this lethal effect occurred after LLI and was not sustainable. It is concluded that 405-nm LLI has a lethal effect on human-derived glioblastoma A-172 cells, that is different from the suppressive effect without morphological changes induced by 808-nm LLI.
Collapse
Affiliation(s)
- Foong Yee Ang
- Department of Bioinformatics, Graduate School of Engineering, Soka University, 1-236 Tangi-cho, Hachioji city, Tokyo, 192-8577, Japan
| | | | | | | |
Collapse
|
191
|
Qiu W, Wang X, Leibowitz B, Yang W, Zhang L, Yu J. PUMA-mediated apoptosis drives chemical hepatocarcinogenesis in mice. Hepatology 2011; 54:1249-58. [PMID: 21725994 PMCID: PMC3184207 DOI: 10.1002/hep.24516] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
UNLABELLED Hepatocyte death and proliferation contribute to hepatocellular carcinoma development after carcinogen exposure or chronic liver inflammation. However, the role and the molecular targets of hepatocyte death in relation to compensatory proliferation have not been fully characterized. In this study, we investigated the role of p53 up-regulated modulator of apoptosis (PUMA), a BH3-only protein important for both p53-dependent and -independent apoptosis, in a diethylnitrosamine (DEN)-induced liver carcinogenesis model. PUMA deficiency significantly decreased the multiplicity and size of liver tumors. DEN treatment induced p53-independent PUMA expression, PUMA-dependent hepatocyte death, and compensatory proliferation. Furthermore, inhibition or deletion of c-jun N-terminal kinase 1 (JNK1) abrogated PUMA induction, hepatocyte death, and compensatory proliferation. CONCLUSION These results provide direct evidence that JNK1/PUMA-dependent apoptosis promotes chemical hepatocarcinogenesis through compensatory proliferation, and suggest apoptotic inducers as potential therapeutic targets in liver injury and cancer.
Collapse
Affiliation(s)
- Wei Qiu
- Department of Pathology, University of Pittsburgh School of Medicine, University of Pittsburgh Cancer Institute, 5117 Centre Ave., Pittsburgh, PA 15213, USA
| | - Xinwei Wang
- Department of Pathology, University of Pittsburgh School of Medicine, University of Pittsburgh Cancer Institute, 5117 Centre Ave., Pittsburgh, PA 15213, USA
| | - Brian Leibowitz
- Department of Pathology, University of Pittsburgh School of Medicine, University of Pittsburgh Cancer Institute, 5117 Centre Ave., Pittsburgh, PA 15213, USA
| | - Wancai Yang
- University of Illinois at Chicago, Department of Pathology, 840 S. Wood Street, Chicago, IL 60612
| | - Lin Zhang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, University of Pittsburgh Cancer Institute, 5117 Centre Ave., Pittsburgh, PA 15213, USA
| | - Jian Yu
- Department of Pathology, University of Pittsburgh School of Medicine, University of Pittsburgh Cancer Institute, 5117 Centre Ave., Pittsburgh, PA 15213, USA,Correspondence: Jian Yu, Ph.D., Hillman Cancer Center Research Pavilion, Suite 2.26h, 5117 Centre Ave, Pittsburgh, PA 15213. ; Phone: 412-623-7786; Fax: 412-623-7778
| |
Collapse
|
192
|
Abstract
Control of intracellular redox balance has emerged as a primary function of the p53 network, with crucial implications for tumor suppression, aging, and cell metabolism. Mitochondria are central to redox homeostasis, produce energy, and trigger apoptosis and senescence: not surprisingly, many "old" and "new" functions of p53 appear to be based in mitochondria. Genetic and biomolecular evidence indicates that generation of reactive oxygen species (ROS) in mitochondria can be a deliberate and finely regulated cell response on which signaling by environmental stressors, oncogenes, and nutrients converge. p53 orchestrates mitochondrial redox signaling by the coordinated control of at least two key effectors: the superoxide scavenger MnSOD, and the ROS generator p66shc. This review presents recent evidence and emerging questions regarding the p53-MnSOD-p66shc connection, and discusses how dissection of a circuitry comprising a tumor suppressor, an antioxidant, and a molecule regulating cell survival and mammalian lifespan can provide a framework to address important aspects related to the intricate connection between metabolism, aging, and cancer.
Collapse
Affiliation(s)
- Giovambattista Pani
- Institute of General Pathology, Universitá Cattolica School of Medicine, Largo F. Vito #1, Rome, Italy.
| | | |
Collapse
|
193
|
Hafsi H, Hainaut P. Redox control and interplay between p53 isoforms: roles in the regulation of basal p53 levels, cell fate, and senescence. Antioxid Redox Signal 2011; 15:1655-67. [PMID: 21194382 DOI: 10.1089/ars.2010.3771] [Citation(s) in RCA: 223] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The p53 tumor suppressor protein has achieved stardom in molecular oncology owing to frequent inactivation in a large range of cancers. Known as a factor activated by multiple forms of stress and causing a broad suppressive response to DNA damage, its regulation and functions in basal (non-stress) conditions has received relatively little attention. We summarize recent findings highlighting roles of p53 in physiological processes such as stem cell maintenance, development, aging and senescence, and regulation of basal oxidative cell metabolism. We suggest that these properties are regulated through two integrated biochemical systems: the redox-sensing capacity of the p53 protein (due to its structural features and its regulation by redox factors such as thioredoxin, metallothioneins, or the redox-repair enzyme APE1/ref-1), and the expression of p53 as multiple isoforms with antagonist effects. We propose that interactions between p53 and its isoforms Δ40p53 or Δ133p53 play critical roles in intracellular signaling by reactive oxygen species. We also discuss evidence that p53 controls energy production by repressing glycolysis and enhancing mitochondrial oxidative metabolism. Together, these mechanisms suggest that p53 acts not only as a "guardian of the genome" against DNA damage but also as a finely-tuned regulator of redox-dependent physiological processes.
Collapse
Affiliation(s)
- Hind Hafsi
- International Agency for Research on Cancer, 150 cours Albert Thomas, Lyon, France
| | | |
Collapse
|
194
|
Abstract
p53 regulates the cell cycle and deoxyribonucleic acid (DNA) repair pathways as part of its unequivocally important function to maintain genomic stability. Intriguingly, recent studies show that p53 can also transactivate genes involved in coordinating the two major pathways of energy generation to promote aerobic metabolism, but how this serves to maintain genomic stability is less clear. In an attempt to understand the biology, this review presents human epidemiologic data on the inverse relationship between aerobic capacity and cancer incidence that appears to be mirrored by the impact of p53 on aerobic capacity in mouse models. The review summarizes mechanisms by which p53 regulates mitochondrial respiration and proposes how this might contribute to maintaining genomic stability. Although disparate in nature, the data taken together suggest that the promotion of aerobic metabolism by p53 serves as an important tumor suppressor activity and may provide insights for cancer prevention strategies in the future.
Collapse
Affiliation(s)
- Cory U. Lago
- Center for Molecular Medicine, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Ho Joong Sung
- Center for Molecular Medicine, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
- Department of Biomedical Laboratory Science, College of Health Science, Eulji University, GyeongGi-Do, Korea
| | - Wenzhe Ma
- Center for Molecular Medicine, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Ping-yuan Wang
- Center for Molecular Medicine, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Paul M. Hwang
- Center for Molecular Medicine, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
195
|
Jung YS, Qian Y, Chen X. The p73 tumor suppressor is targeted by Pirh2 RING finger E3 ubiquitin ligase for the proteasome-dependent degradation. J Biol Chem 2011; 286:35388-35395. [PMID: 21852228 DOI: 10.1074/jbc.m111.261537] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The p73 gene, a homologue of the p53 tumor suppressor, is expressed as TA and ΔN isoforms. TAp73 has similar activity as p53 and functions as a tumor suppressor whereas ΔNp73 has both pro- and anti-survival functions. While p73 is rarely mutated in spontaneous tumors, the expression status of p73 is linked to the sensitivity of tumor cells to chemotherapy and prognosis for many types of human cancer. Thus, uncovering its regulators in tumors is of great interest. Here, we found that Pirh2, a RING finger E3 ubiquitin ligase, promotes the proteasome-dependent degradation of p73. Specifically, we showed that knockdown of Pirh2 up-regulates, whereas ectopic expression of Pirh2 down-regulates, expression of endogenous and exogenous p73. In addition, Pirh2 physically associates with and promotes TAp73 polyubiquitination both in vivo and in vitro. Moreover, we found that p73 can be degraded by both 20 S and 26 S proteasomes. Finally, we showed that Pirh2 knockdown leads to growth suppression in a TAp73-dependent manner. Taken together, our findings indicate that Pirh2 promotes the proteasomal turnover of TAp73, and thus targeting Pirh2 to restore TAp73-mediated growth suppression in p53-deficient tumors may be developed as a novel anti-cancer strategy.
Collapse
Affiliation(s)
- Yong-Sam Jung
- Comparative Oncology Laboratory, University of California, Davis, California 95616
| | - Yingjuan Qian
- Comparative Oncology Laboratory, University of California, Davis, California 95616
| | - Xinbin Chen
- Comparative Oncology Laboratory, University of California, Davis, California 95616.
| |
Collapse
|
196
|
Coriat R, Marut W, Leconte M, Ba LB, Vienne A, Chéreau C, Alexandre J, Weill B, Doering M, Jacob C, Nicco C, Batteux F. The organotelluride catalyst LAB027 prevents colon cancer growth in the mice. Cell Death Dis 2011; 2:e191. [PMID: 21833029 PMCID: PMC3181419 DOI: 10.1038/cddis.2011.73] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Organotellurides are newly described redox-catalyst molecules with original pro-oxidative properties. We have investigated the in vitro and in vivo antitumoral effects of the organotelluride catalyst LAB027 in a mouse model of colon cancer and determined its profile of toxicity in vivo. LAB027 induced an overproduction of H(2)O(2) by both human HT29 and murine CT26 colon cancer cell lines in vitro. This oxidative stress was associated with a decrease in proliferation and survival rates of the two cell lines. LAB027 triggered a caspase-independent, ROS-mediated cell death by necrosis associated with mitochondrial damages and autophagy. LAB027 also synergized with the cytotoxic drug oxaliplatin to augment its cytostatic and cytotoxic effects on colon cancer cell lines but not on normal fibroblasts. The opposite effects of LAB027 on tumor and on non-transformed cells were linked to differences in the modulation of reduced glutathione metabolism between the two types of cells. In mice grafted with CT26 tumor cells, LAB027 alone decreased tumor growth compared with untreated mice, and synergized with oxaliplatin to further decrease tumor development compared with mice treated with oxaliplatin alone. LAB027 an organotelluride catalyst compound synergized with oxaliplatin to prevent both in vitro and in vivo colon cancer cell proliferation while decreasing the in vivo toxicity of oxaliplatin. No in vivo adverse effect of LAB027 was observed in this model.
Collapse
Affiliation(s)
- R Coriat
- Université Paris Descartes, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris (AP-HP), Laboratoire d'immunologie, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
197
|
Xu XM, Møller SG. Iron-sulfur clusters: biogenesis, molecular mechanisms, and their functional significance. Antioxid Redox Signal 2011; 15:271-307. [PMID: 20812788 DOI: 10.1089/ars.2010.3259] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Iron-sulfur clusters [Fe-S] are small, ubiquitous inorganic cofactors representing one of the earliest catalysts during biomolecule evolution and are involved in fundamental biological reactions, including regulation of enzyme activity, mitochondrial respiration, ribosome biogenesis, cofactor biogenesis, gene expression regulation, and nucleotide metabolism. Although simple in structure, [Fe-S] biogenesis requires complex protein machineries and pathways for assembly. [Fe-S] are assembled from cysteine-derived sulfur and iron onto scaffold proteins followed by transfer to recipient apoproteins. Several predominant iron-sulfur biogenesis systems have been identified, including nitrogen fixation (NIF), sulfur utilization factor (SUF), iron-sulfur cluster (ISC), and cytosolic iron-sulfur protein assembly (CIA), and many protein components have been identified and characterized. In eukaryotes ISC is mainly localized to mitochondria, cytosolic iron-sulfur protein assembly to the cytosol, whereas plant sulfur utilization factor is localized mainly to plastids. Because of this spatial separation, evidence suggests cross-talk mediated by organelle export machineries and dual targeting mechanisms. Although research efforts in understanding iron-sulfur biogenesis has been centered on bacteria, yeast, and plants, recent efforts have implicated inappropriate [Fe-S] biogenesis to underlie many human diseases. In this review we detail our current understanding of [Fe-S] biogenesis across species boundaries highlighting evolutionary conservation and divergence and assembling our knowledge into a cellular context.
Collapse
Affiliation(s)
- Xiang Ming Xu
- Centre for Organelle Research CORE, University of Stavanger, Norway
| | | |
Collapse
|
198
|
Oxidative damage to guanine nucleosides following combination chemotherapy with 5-fluorouracil and oxaliplatin. Cancer Chemother Pharmacol 2011; 69:301-7. [DOI: 10.1007/s00280-011-1700-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Accepted: 06/16/2011] [Indexed: 11/26/2022]
|
199
|
Zhang T, Tang SS, Jin X, Liu FY, Zhang CM, Zhao WX, Zhang S, Sun CD, Xiao XL. c-Myc influences olaquindox-induced apoptosis in human hepatoma G2 cells. Mol Cell Biochem 2011; 354:253-61. [DOI: 10.1007/s11010-011-0825-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2010] [Accepted: 04/15/2011] [Indexed: 12/27/2022]
|
200
|
Artyukhov VG, Trubitsyna MS, Nakvasina MA, Solov’eva EV. DNA fragmentation of human lymphocytes in dynamics of development of apoptosis induced by action of UV radiation and reactive oxygen species. ACTA ACUST UNITED AC 2011. [DOI: 10.1134/s1990519x11020039] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|