1
|
Lotfi N, Yousefi Z, Golabi M, Khalilian P, Ghezelbash B, Montazeri M, Shams MH, Baghbadorani PZ, Eskandari N. The potential anti-cancer effects of quercetin on blood, prostate and lung cancers: An update. Front Immunol 2023; 14:1077531. [PMID: 36926328 PMCID: PMC10011078 DOI: 10.3389/fimmu.2023.1077531] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 02/07/2023] [Indexed: 03/08/2023] Open
Abstract
Cancer is caused by abnormal proliferation of cells and aberrant recognition of the immune system. According to recent studies, natural products are most likely to be effective at preventing cancer without causing any noticeable complications. Among the bioactive flavonoids found in fruits and vegetables, quercetin is known for its anti-inflammatory, antioxidant, and anticancer properties. This review aims to highlight the potential therapeutic effects of quercetin on some different types of cancers including blood, lung and prostate cancers.
Collapse
Affiliation(s)
- Noushin Lotfi
- Department of Medical Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zahra Yousefi
- School of Allied Medical Sciences, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Marjan Golabi
- Department of Medical Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Parvin Khalilian
- Department of Medical Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Behrooz Ghezelbash
- Department of Medical Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mina Montazeri
- Department of Medical Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Hossein Shams
- Department of Medical Immunology, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | | | - Nahid Eskandari
- Department of Medical Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
2
|
Identification of a novel catalytic inhibitor of topoisomerase II alpha that engages distinct mechanisms in p53 wt or p53 -/- cells to trigger G2/M arrest and senescence. Cancer Lett 2022; 526:284-303. [PMID: 34843865 DOI: 10.1016/j.canlet.2021.11.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 11/10/2021] [Accepted: 11/19/2021] [Indexed: 12/18/2022]
Abstract
We report a novel topoisomerase IIα inhibitor, mercaptopyridine oxide (MPO), which induces G2/M arrest and senescence with distinctly different cell cycle regulators (p21 or p14ARF) in HCT116p 53WT and HCT116 p53-/- cells, respectively. MPO treatment induced defective topoisomerase IIα-mediated decatenation process and inhibition of the enzyme's catalytic activity that stalled entry into mitosis. Topoisomerase IIα inhibition was associated with ROS-mediated activation of ATM-Chk2 kinase axis in HCT116 p53WT cells, but not in HCT116 p53-/- cells displaying early Chk1 activation. Results suggest that E2F1 stabilization might link MPO-induced p53 phospho-activation in HCT116 p53WT cells or p14ARF induction in HCT116 p53-/- cells. Also, interaction between topoisomerase IIα and Chk1 was induced in both cell lines, which could be important for decatenation checkpoint activation, even upon p53 ablation. Notably, TCGA dataset analyses revealed topoisomerase IIα upregulation across a wide array of cancers, which was associated with lower overall survival. Corroborating that increased topoisomerase IIα expression might offer susceptibility to the novel inhibitor, MPO (5 μM) induced strong inhibition in colony forming ability of pancreatic and hepatocellular cancer cell lines. These data highlight a novel topoisomerase IIα inhibitor and provide proof-of-concept for its therapeutic potential against cancers even with loss-of-function of p53.
Collapse
|
3
|
Sun D, Li WY, Chen SH, Zhi ZF, Lin HS, Fan JT, Fan YJ. shRNA-Mediated Suppression of γ-Synuclein Leading to Downregulation of p38/ERK/JNK Phosphorylation and Cell Cycle Arrest in Endometrial Cancer Cells. Mol Biol 2021. [DOI: 10.1134/s0026893320060114] [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]
|
4
|
Hong FU, Castro M, Linse K. Tumor-specific lytic path “hyperploid progression mediated death”: Resolving side effects through targeting retinoblastoma or p53 mutant. World J Clin Oncol 2020; 11:854-867. [PMID: 33312882 PMCID: PMC7701912 DOI: 10.5306/wjco.v11.i11.854] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/08/2020] [Accepted: 10/20/2020] [Indexed: 02/06/2023] Open
Abstract
A major advance was made to reduce the side effects of cancer therapy via the elucidation of the tumor-specific lytic path “hyperploid progression-mediated death” targeting retinoblastoma (Rb) or p53-mutants defective in G1 DNA damage checkpoint. The genetic basis of human cancers was uncovered through the cloning of the tumor suppressor Rb gene. It encodes a nuclear DNA-binding protein whose self-interaction is regulated by cyclin-dependent kinases. A 3D-structure of Rb dimer is shown, confirming its multimeric status. Rb assumes a central role in cell cycle regulation and the “Rb pathway” is universally inactivated in human cancers. Hyperploidy refers to a state in which cells contain one or more extra chromosomes. Hyperploid progression occurs due to continued cell-cycling without cytokinesis in G1 checkpoint-defective cancer cells. The evidence for the triggering of hyperploid progression-mediated death in RB-mutant human retinoblastoma cells is shown. Hence, the very genetic mutation that predisposes to cancer can be exploited to induce lethality. The discovery helped to establish the principle of targeted cytotoxic cancer therapy at the mechanistic level. By triggering the lytic path, targeted therapy with tumor specificity at the genetic level can be developed. It sets the stage for systematically eliminating side effects for cytotoxic cancer therapy.
Collapse
Affiliation(s)
- Frank-Un Hong
- Department of Research and Development, Bio-Synthesis, Lewisville, TX 75057, United States
| | - Miguel Castro
- Department of Research and Development, Bio-Synthesis, Lewisville, TX 75057, United States
| | - Klaus Linse
- Department of Research and Development, Bio-Synthesis, Lewisville, TX 75057, United States
| |
Collapse
|
5
|
Zhu L, Wu Q, Quan B, Yang J, Yang J, Hou W, Cheng L. Autophagy inhibition by reversine and its suppressive effects on human hepatocellular carcinoma cells. Biochem Biophys Res Commun 2020; 528:105-111. [PMID: 32456791 DOI: 10.1016/j.bbrc.2020.05.054] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 05/08/2020] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Therapy for human hepatocellular carcinoma (HCC) remains a great challenge for physicians and patients worldwide. The anti-tumor effects of reversine have attracted much more concerns. MATERIALS AND METHODS This study evaluated the growth regulatory effects of reversine on HCC cells lines. Meanwhile, the underlying mechanism including autophagy modulation was also identified. RESULTS reversine markedly inhibited the proliferation of both HCC cells and induced cell apoptosis and multinuclear in a dose-dependent manner. In addition, the decreased ratio of LC3II/LC3I as well as elevated p62 expression were observed under reversine treatment, indicating the autophagy inhibition by reversine in HepG2 cell line. Moreover, modulation of autophagy with rapamycin and chloroquine significantly attenuated and enhanced the cytostatic effects of reversine, respectively. CONCLUSIONS reversine could reduce the cell viability of HCC cells via inducing cell apoptosis and polyploidy. In addition, cell autophagy was involved and might play a protective role in HCC cells, the joint use of autophagy inhibitor enhanced reversine-mediating antitumor effects. Our data offered novel ideas for comprehensive therapeutic regimes on human hepatocellular carcinoma.
Collapse
Affiliation(s)
- Lingling Zhu
- Department of Infectious Disease, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Qiongle Wu
- Department of Infectious Disease, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Bin Quan
- Department of Infectious Disease, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Jianghua Yang
- Department of Infectious Disease, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Jinsun Yang
- Department of Infectious Disease, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Weishun Hou
- Department of Infectious Disease, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Li Cheng
- Department of Urology, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui Province, China.
| |
Collapse
|
6
|
Zhang X, Huang J, Yu C, Xiang L, Li L, Shi D, Lin F. Quercetin Enhanced Paclitaxel Therapeutic Effects Towards PC-3 Prostate Cancer Through ER Stress Induction and ROS Production. Onco Targets Ther 2020; 13:513-523. [PMID: 32021294 PMCID: PMC6970612 DOI: 10.2147/ott.s228453] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 12/16/2019] [Indexed: 12/24/2022] Open
Abstract
Introduction Prostate cancer is one of the most common cancers threatening public health worldwide. Although chemotherapy plays an important role in treating prostate cancer, it leads to many adverse effects and is prone to drug resistance. Quercetin, a natural product, is used in traditional Chinese medicine because of its strong antitumor activity and few side effects. Methods In this study, we combined quercetin and paclitaxel to kill prostate cancer cells in vivo and in vitro, and we investigated the relevant mechanism of this combination treatment. After the cancer cells were treated with quercetin or/and paclitaxel, cell growth inhibition, apoptosis, the cell cycle, reactive oxygen species (ROS) generation, and several endoplasmic reticulum (ER) stress signaling pathway related gene expressions were evaluated. Results The combined treatment with quercetin and paclitaxel significantly inhibited cell proliferation, increased apoptosis, arrested the cell cycle at the G2/M phase, inhibited cell migration, dramatically induced ER stress to occur, and increased ROS generation. In a PC-3 cancer-bearing murine model, this combination treatment exerted the most beneficial therapeutic effects, and quercetin increased the cancer cell-killing effects of paclitaxel, with nearly no side effects compared with the single paclitaxel treatment group. Conclusion Combination treatment possessed enhanced anti-cancer effects, and these results will provide a basis for treating prostate cancer using a combination of quercetin and paclitaxel.
Collapse
Affiliation(s)
- Xiangyu Zhang
- Department of Pathology, Jining First People's Hospital, Jining Medical University, Jining 272000, People's Republic of China
| | - Jingwen Huang
- Anhui Province Key Laboratory of Translational Cancer Research Affiliated to Bengbu Medical University, Bengbu 233004, People's Republic of China
| | - Chao Yu
- Department of Clinical Medicine, Jining Medical University, Jining 272000, People's Republic of China
| | - Longquan Xiang
- Department of Pathology, Jining First People's Hospital, Jining Medical University, Jining 272000, People's Republic of China
| | - Liang Li
- Department of Pathology, Jining First People's Hospital, Jining Medical University, Jining 272000, People's Republic of China
| | - Dongmei Shi
- Department of Dermatology, Jining First People's Hospital, Jining Medical University, Jining 272000, People's Republic of China
| | - Fanzhong Lin
- Department of Pathology, Jining First People's Hospital, Jining Medical University, Jining 272000, People's Republic of China
| |
Collapse
|
7
|
Cheng L, Wang H, Guo K, Wang Z, Zhang Z, Shen C, Chen L, Lin J. Reversine, a substituted purine, exerts an inhibitive effect on human renal carcinoma cells via induction of cell apoptosis and polyploidy. Onco Targets Ther 2018. [PMID: 29520153 PMCID: PMC5833753 DOI: 10.2147/ott.s158198] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Background Human renal cell carcinoma (RCC) is the most common type of kidney cancer that arises from the renal epithelium. Up to 33.3% of RCC patients treated with local tumor resections will subsequently develop recurrence or metastases. Thus, optimized therapeutic regimes are urgently needed to improve the prognosis of RCC. Reversine was recently reported to exert critical roles in cancer therapy. Materials and methods This study evaluated the anti-tumor effects of reversine on cell viability, colony formation, apoptosis, and cell cycle in 786-O and ACHN cell lines. Results It was demonstrated that reversine significantly inhibited the proliferation of both cell lines in time- and dose-dependent manners. Polyploidy formation was observed under high-concentration reversine treatment. In addition, reversine induced cell death via caspase-dependent apoptotic pathways, which could be partially inhibited by Z-VAD-FMK, a pan-caspase inhibitor. Conclusion Reversine could effectively suppress the proliferation of human RCC cells, and may serve as a novel therapeutic regimen for RCC in clinical practice.
Collapse
Affiliation(s)
- Li Cheng
- Department of Urology, Peking University First Hospital, Beijing, China.,Institute of Urology, Peking University, Beijing, China
| | - Hao Wang
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
| | - Kecun Guo
- Department of Urology, The Second People's Hospital of Liaocheng, Shandong, China
| | - Zicheng Wang
- Department of Urology, Peking University First Hospital, Beijing, China.,Institute of Urology, Peking University, Beijing, China
| | - Zhongyuan Zhang
- Department of Urology, Peking University First Hospital, Beijing, China.,Institute of Urology, Peking University, Beijing, China.,National Urological Cancer Center, Beijing, China
| | - Cheng Shen
- Department of Urology, Peking University First Hospital, Beijing, China.,Institute of Urology, Peking University, Beijing, China.,National Urological Cancer Center, Beijing, China
| | - Liang Chen
- Medical Center of Reproductive and Genetics, Peking University First Hospital, Beijing, China
| | - Jian Lin
- Department of Urology, Peking University First Hospital, Beijing, China.,Institute of Urology, Peking University, Beijing, China.,National Urological Cancer Center, Beijing, China
| |
Collapse
|
8
|
Wen L, Xiao B, Shi Y, Han F. PERK signalling pathway mediates single prolonged stress-induced dysfunction of medial prefrontal cortex neurons. Apoptosis 2018; 22:753-768. [PMID: 28391375 DOI: 10.1007/s10495-017-1371-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Post-traumatic stress disorder (PTSD) is characterized with abnormal learning and memory. Impairments in learning and memory are closely associated with apoptosis in the medial prefrontal cortex (mPFC). We previously examined the endoplasmic reticulum (ER) stress was involved in the apoptosis in the mPFC of PTSD. The PERK pathway plays the important role in the ER stress-induced apoptosis. The aim of the present study was to explore the role of PERK pathway in neuronal apoptosis in the mPFC of rat models of PTSD. We used the single prolonged stress (SPS) to mimic PTSD in rats and studied the effects of the PERK pathway in mPFC. Learning and memory behavior were examined by Morris water maze and novel object recognition tests. Apoptosis in mPFC was detected by TUNEL staining. Our results showed decreased learning memory and increased apoptosis of mPFC neurons in rats exposed to SPS. SPS exposure upregulate mRNA expressions of PERK, p-PERK, eIF2α, p-eIF2α, nuclear ATF4 and C/EBP-homologous protein (CHOP) in mPFC neurons, but the protein levels of these molecules showed difference in magnitude and time course. GSK2606414 (an antagonist of PERK) treatment successfully reversed the above changes. These results suggested that the PERK pathway mediated SPS-induced neural apoptosis in the mPFC. These findings will be helpful in understanding mPFC-related pathogenesis of PTSD.
Collapse
Affiliation(s)
- Lili Wen
- PTSD Lab, Department of Histo-Embryology, Basic Medical Sciences College, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China
| | - Bing Xiao
- PTSD Lab, Department of Histo-Embryology, Basic Medical Sciences College, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China
| | - Yuxiu Shi
- PTSD Lab, Department of Histo-Embryology, Basic Medical Sciences College, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China
| | - Fang Han
- PTSD Lab, Department of Histo-Embryology, Basic Medical Sciences College, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China.
| |
Collapse
|
9
|
Krushkal J, Zhao Y, Hose C, Monks A, Doroshow JH, Simon R. Concerted changes in transcriptional regulation of genes involved in DNA methylation, demethylation, and folate-mediated one-carbon metabolism pathways in the NCI-60 cancer cell line panel in response to cancer drug treatment. Clin Epigenetics 2016; 8:73. [PMID: 27347216 PMCID: PMC4919895 DOI: 10.1186/s13148-016-0240-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 06/15/2016] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Aberrant patterns of DNA methylation are abundant in cancer, and epigenetic pathways are increasingly being targeted in cancer drug treatment. Genetic components of the folate-mediated one-carbon metabolism pathway can affect DNA methylation and other vital cell functions, including DNA synthesis, amino acid biosynthesis, and cell growth. RESULTS We used a bioinformatics tool, the Transcriptional Pharmacology Workbench, to analyze temporal changes in gene expression among epigenetic regulators of DNA methylation and demethylation, and one-carbon metabolism genes in response to cancer drug treatment. We analyzed gene expression information from the NCI-60 cancer cell line panel after treatment with five antitumor agents, 5-azacytidine, doxorubicin, vorinostat, paclitaxel, and cisplatin. Each antitumor agent elicited concerted changes in gene expression of multiple pathway components across the cell lines. Expression changes of FOLR2, SMUG1, GART, GADD45A, MBD1, MTR, MTHFD1, and CTH were significantly correlated with chemosensitivity to some of the agents. Among many genes with concerted expression response to individual antitumor agents were genes encoding DNA methyltransferases DNMT1, DNMT3A, and DNMT3B, epigenetic and DNA repair factors MGMT, GADD45A, and MBD1, and one-carbon metabolism pathway members MTHFD1, TYMS, DHFR, MTR, MAT2A, SLC19A1, ATIC, and GART. CONCLUSIONS These transcriptional changes are likely to influence vital cellular functions of DNA methylation and demethylation, cellular growth, DNA biosynthesis, and DNA repair, and some of them may contribute to cytotoxic and apoptotic action of the drugs. This concerted molecular response was observed in a time-dependent manner, which may provide future guidelines for temporal selection of genetic drug targets for combination drug therapy treatment regimens.
Collapse
Affiliation(s)
- Julia Krushkal
- />Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, 9609 Medical Center Dr., Rockville, MD 20850 USA
| | - Yingdong Zhao
- />Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, 9609 Medical Center Dr., Rockville, MD 20850 USA
| | - Curtis Hose
- />Molecular Pharmacology Group, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702 USA
| | - Anne Monks
- />Molecular Pharmacology Group, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702 USA
| | - James H. Doroshow
- />Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD 20892 USA
| | - Richard Simon
- />Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, 9609 Medical Center Dr., Rockville, MD 20850 USA
| |
Collapse
|
10
|
Yamada T, Das Gupta TK, Beattie CW. p28-Mediated Activation of p53 in G2–M Phase of the Cell Cycle Enhances the Efficacy of DNA Damaging and Antimitotic Chemotherapy. Cancer Res 2016; 76:2354-65. [DOI: 10.1158/0008-5472.can-15-2355] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 01/15/2016] [Indexed: 11/16/2022]
|
11
|
Castilla C, Flores ML, Medina R, Pérez-Valderrama B, Romero F, Tortolero M, Japón MA, Sáez C. Prostate cancer cell response to paclitaxel is affected by abnormally expressed securin PTTG1. Mol Cancer Ther 2014; 13:2372-83. [PMID: 25122070 DOI: 10.1158/1535-7163.mct-13-0405] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PTTG1 protein, the human securin, has a central role in sister chromatid separation during mitosis, and its altered expression has been reported in many tumor types. Paclitaxel is a widely used chemotherapeutic drug, whose mechanism of action is related to its ability to arrest cells in mitosis and the subsequent induction of the intrinsic apoptotic pathway. By using two prostate cancer cell lines with different responses to paclitaxel treatment, we have identified two situations in which PTTG1 influences cell fate differentially. In slippage-prone PC3 cells, both PTTG1 downregulation and overexpression induce an increase in mitotic cells that is associated with diminished apoptosis after paclitaxel treatment. In LNCaP cells, however, PTTG1 downregulation prevents mitotic entry and, subsequently, inhibits mitosis-associated, paclitaxel-induced apoptosis. In contrast, PTTG1 overexpression induces an increase in mitotic cells and apoptosis after paclitaxel treatment. We have also identified a role for Mcl-1 protein in preventing apoptosis during mitosis in PC3 cells, as simultaneous PTTG1 and Mcl-1 silencing enhances mitosis-associated apoptosis after paclitaxel treatment. The finding that a more efficient mitotic arrest alone in PC3 cells is not enough to increase apoptosis was also confirmed with the observation that a selected paclitaxel-resistant PC3 cell line showed an apoptosis-resistant phenotype associated with increased mitosis upon paclitaxel treatment. These findings could contribute to identify putative responsive and nonresponsive cells and help us to approach incomplete responses to paclitaxel in the clinical setting.
Collapse
Affiliation(s)
- Carolina Castilla
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain
| | - M Luz Flores
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain
| | - Rafael Medina
- Department of Urology, Hospital Universitario Virgen del Rocío, Seville, Spain
| | | | - Francisco Romero
- Department of Microbiology, Faculty of Biology, Universidad de Sevilla, Seville, Spain
| | - María Tortolero
- Department of Microbiology, Faculty of Biology, Universidad de Sevilla, Seville, Spain
| | - Miguel A Japón
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain. Department of Pathology, Hospital Universitario Virgen del Rocío, Seville, Spain.
| | - Carmen Sáez
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain. Department of Pathology, Hospital Universitario Virgen del Rocío, Seville, Spain.
| |
Collapse
|
12
|
Riva G, Baronchelli S, Paoletta L, Butta V, Biunno I, Lavitrano M, Dalprà L, Bentivegna A. In vitro anticancer drug test: A new method emerges from the model of glioma stem cells. Toxicol Rep 2014; 1:188-199. [PMID: 28962238 PMCID: PMC5598297 DOI: 10.1016/j.toxrep.2014.05.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 05/08/2014] [Accepted: 05/12/2014] [Indexed: 11/03/2022] Open
Abstract
Glioblastoma multiforme (GBM) is a grade IV astrocytoma and the most common malignant brain tumor. Current therapies provide a median survival of 12–15 months after diagnosis, due to the high recurrence rate. The failure of current therapies may be due to the presence, within the tumor, of cells characterized by enhanced self-renewal capacity, multilineage differentiation potential and elevated invasive behavior, called glioma stem cells (GSCs). To evaluate the pharmacological efficacy of selected drugs on six GSC lines, we set up a multiple drug responsivity assay based on the combined evaluation of cytomorphological and functional parameters, including the analysis of polymorphic nuclei, mitotic index and cell viability. In order to understand the real pharmacological efficacy of the tested drugs, we assigned a specific drug responsivity score to each GSC line, integrating the data produced by multiple assays. In this work we explored the antineoplastic effects of paclitaxel (PTX), an inhibitor of microtubule depolymerization, utilized as standard treatment in several cancers, and of valproic acid (VPA), an inhibitor of histone deacetylases (HDACs) with multiple anticancer properties. We classified the six GSC lines as responsive or resistant to these drugs, on the basis of their responsivity scores. This method can also be useful to identify the best way to combine two or more drugs. In particular, we utilized the pro-differentiating effect of VPA to improve the PTX effectiveness and we observed a significant reduction of cell viability compared to single treatments.
Collapse
Affiliation(s)
- Gabriele Riva
- Department of Surgery and Translational Medicine, University of Milano-Bicocca, via Cadore 48, 20900 Monza, Italy
| | - Simona Baronchelli
- Department of Surgery and Translational Medicine, University of Milano-Bicocca, via Cadore 48, 20900 Monza, Italy.,Institute for Genetic and Biomedical Research - National Research Council (IRGB-CNR), via Fantoli 16/15, 20138 Milan, Italy
| | - Laura Paoletta
- Department of Surgery and Translational Medicine, University of Milano-Bicocca, via Cadore 48, 20900 Monza, Italy
| | - Valentina Butta
- Department of Surgery and Translational Medicine, University of Milano-Bicocca, via Cadore 48, 20900 Monza, Italy
| | - Ida Biunno
- Institute for Genetic and Biomedical Research - National Research Council (IRGB-CNR), via Fantoli 16/15, 20138 Milan, Italy.,IRCCS MultiMedica, Science and Technology Pole, via Fantoli 16/15, 20138 Milan, Italy
| | - Marialuisa Lavitrano
- Department of Surgery and Translational Medicine, University of Milano-Bicocca, via Cadore 48, 20900 Monza, Italy
| | - Leda Dalprà
- Department of Surgery and Translational Medicine, University of Milano-Bicocca, via Cadore 48, 20900 Monza, Italy.,Medical Genetics Laboratory, S. Gerardo Hospital, via Pergolesi 33, 20900 Monza, Italy
| | - Angela Bentivegna
- Department of Surgery and Translational Medicine, University of Milano-Bicocca, via Cadore 48, 20900 Monza, Italy
| |
Collapse
|
13
|
Synuclein γ Compromises Spindle Assembly Checkpoint and Renders Resistance to Antimicrotubule Drugs. Mol Cancer Ther 2014; 13:699-713. [DOI: 10.1158/1535-7163.mct-13-0671] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
14
|
Cui SY, Wang R, Chen LB. MicroRNAs: key players of taxane resistance and their therapeutic potential in human cancers. J Cell Mol Med 2013; 17:1207-17. [PMID: 24106980 PMCID: PMC4159023 DOI: 10.1111/jcmm.12131] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2013] [Accepted: 08/15/2013] [Indexed: 01/01/2023] Open
Abstract
The successful long-term use of taxane for cancer therapy is often prevented by the development of drug resistance in clinic. Thus, exploring the mechanisms involved is a first step towards rational strategies to overcome taxane resistance. Taxane resistance-related microRNA (miRNAs) are under investigation and miRNAs could induce the taxane resistance of tumour cells by regulating cell cycle distribution, survival and/or apoptosis pathways, drug transports, epithelial–mesenchymal transition and cancer stem cell. This article summarizes current research involving miRNAs as regulators of key target genes for tanxanxe chemoresistance and discusses the complex regulatory networks of miRNAs. Also, the authors will envisage future developments towards the potential use of targeting miRNAs as a novel strategy for improving response of tumour patients to taxane. miRNAs play critical roles in taxane chemoresistance and the miRNA-based therapies will be helpful for overcoming drug resistance and developing more effective personalized anti-cancer treatment strategies. Further research studies should be performed to promote therapeutic–clinical use of taxane resistance-related miRNAs in cancer patients, especially in those patients with taxane-resistant cancers.
Collapse
Affiliation(s)
- Shi-Yun Cui
- Department of Medical Oncology, School of Medicine, Jinling Hospital, Nanjing University, Nanjing, Jiangsu, China
| | | | | |
Collapse
|
15
|
Kunos CA, Stefan T, Jacobberger JW. Cabazitaxel-induced stabilization of microtubules enhances radiosensitivity in ovarian cancer cells. Front Oncol 2013; 3:226. [PMID: 24066277 PMCID: PMC3776429 DOI: 10.3389/fonc.2013.00226] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Accepted: 08/16/2013] [Indexed: 12/21/2022] Open
Abstract
Background: Up to 40% of women with ovarian cancer have short disease-free intervals due to molecular mechanisms of chemotherapy resistance. New therapeutic strategies are sought. Ovarian cancers are sensitive to radiochemotherapy. The taxane cabazitaxel (XRP6258, Jevtana) promotes tubulin assembly and stabilizes microtubules against depolymerization in cells, acting similarly in mechanism to paclitaxel. Here, sequences of cabazitaxel-radiation co-administration are tested for drug-alone cytotoxicity and optimal radiosensitization. Materials and Methods: SKOV3, OVCAR3, and TOV-112D ovarian cancer cells were administered cabazitaxel 24 h before (first), 18 h before (second), together (third), or 24 h after (fourth) a single radiation dose, and then, investigated by clonogenic assay and flow cytometric assays. Radiation dose-cell survival data were fitted by two-stage multivariate analyses of variance. High-content flow cytometry partitioned cabazitaxel effects into G2-phase versus M-phase events by DNA content, cyclin A2, and phospho-S10-histone H3 (PHH3). Paclitaxel served as a comparator. Findings: Cabazitaxel cytotoxicity and radiosensitization were dose dependent. Cabazitaxel added 24 h before radiation was the most lethal schedule. DNA content measurements by flow cytometry showed that cabazitaxel-treated cells accumulated in the radiosensitive G2/M 4C DNA complement compartment. Cytometry also showed that surviving cabazitaxel-induced cell cycle arrested cells resolve the arrest by entering 4C or by 8C DNA complement cell cycles. Interpretation: The radiosensitizing effect of cabazitaxel was schedule dependent, due to cell cycle redistribution, and best when cabazitaxel was given 24 h before radiation. Clinical trials of administering both cabazitaxel and radiation should be explored in women with chemoresistant ovarian cancer.
Collapse
Affiliation(s)
- Charles A Kunos
- Department of Radiation Oncology, Case Western Reserve School of Medicine, Cleveland , OH , USA
| | | | | |
Collapse
|
16
|
Li Y, Han F, Shi Y. Increased neuronal apoptosis in medial prefrontal cortex is accompanied with changes of Bcl-2 and Bax in a rat model of post-traumatic stress disorder. J Mol Neurosci 2013; 51:127-37. [PMID: 23381833 DOI: 10.1007/s12031-013-9965-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 01/14/2013] [Indexed: 10/27/2022]
Abstract
Post-traumatic stress disorder (PTSD) is an anxiety disorder caused by traumatic experience, which affects a patient's quality of life and social stability. The objective of this study was to determine the apoptosis-related genes B-cell lymphoma 2 (Bcl-2) and BCL2-associated X (Bax) expressions and medial prefrontal cortex (mPFC) neuronal apoptosis after PTSD in rat model and therefore to provide experimental evidence to reveal PTSD pathogenesis. The single-prolonged stress (SPS) method was used to set up the rat PTSD models. Chemiluminescence was used to determine serum corticosterone levels. Neuronal apoptosis was detected using transmission electron microscopy, Hoechst staining, and terminal deoxynucleotidyl transferase dUTP nick end labeling staining. Immunohistochemistry, immunofluorescence, RT-PCR, and Western blot were used to detect the expressions of Bcl-2 and Bax protein in mPFC. Our results showed an increased mPFC neuronal apoptosis after SPS stimulation. The number of apoptotic cells peaked on day 7. The expressions of Bcl-2 and Bax peaked on days 4 and 7. The Bcl-2/Bax ratio elevated on days 1 and 4 but decreased markedly on day 7. These results indicated that SPS stimulation increased the number of apoptotic neurons, up-regulated the expressions of Bcl-2 and Bax, and altered the Bcl-2/Bax ratio in the mPFC of PTSD rats.
Collapse
Affiliation(s)
- Yana Li
- Department of Histology and Embryology, Institute of Pathology and Pathophysiology, Basic Medical Sciences College, China Medical University, No. 92 Beima Road, Heping District, Shenyang, 110001, Liaoning Province, China
| | | | | |
Collapse
|
17
|
Liu Y, Bowen NJ, Matyunina L, McDonald J, Prausnitz MR. Gene transfection enhanced by ultrasound exposure combined with drug treatment guided by gene chip analysis. Int J Hyperthermia 2012; 28:349-61. [PMID: 22621736 DOI: 10.3109/02656736.2012.669513] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
PURPOSE Heterogeneous bioeffects have been reported in previous studies of ultrasound-mediated gene delivery. The goal of this study is to identify the differences between cells that take up plasmid DNA (pDNA) after sonication but are not transfected and cells that similarly take up pDNA but are transfected. We used these findings to select drugs that regulate intracellular processes expected to enhance gene transfection in combination with US. MATERIALS AND METHODS Gene expression among DU145 human prostate cancer cells after ultrasound-mediated transfection was analyzed using Affymetrix GeneChip Human Genome U133 Plus 2.0 Arrays. Drug treatments suggested by the microarray analysis were combined with US exposure to regulate the corresponding intracellular processes. Cell viability and transfection efficiency were determined by flow cytometry to analyze the effects of US combined with drug treatment. RESULTS Genes such as GADD45α (growth arrest and DNA-damage inducible, alpha) and Topoisomerase IIα were found to be associated with successful transfection. Drugs that regulate GADD45α and Topoisomerase IIα (e.g., ethyl methanesulfomate, amsacrine and chloroquine) were shown to increase ultrasound-mediated transfection efficiency by up to 2 fold. CONCLUSIONS Among cells with pDNA uptake after sonication, we found that genes are differentially expressed among transfected cells versus non-transfected cells. Regulation of the expression level of GADD45α and TOP2α and other intracellular processes can yield higher efficiency of ultrasound-mediated gene transfection. This suggests that a strategy to increase gene transfection efficiency involving the combination of sonication and regulation of intracellular processes using drugs could further enhance US-mediated gene transfection.
Collapse
Affiliation(s)
- Ying Liu
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332-0100, USA
| | | | | | | | | |
Collapse
|
18
|
Llovera L, Mansilla S, Portugal J. Apoptotic-like death occurs through a caspase-independent route in colon carcinoma cells undergoing mitotic catastrophe. Cancer Lett 2012; 326:114-21. [PMID: 22885806 DOI: 10.1016/j.canlet.2012.08.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Revised: 06/29/2012] [Accepted: 08/01/2012] [Indexed: 01/11/2023]
Abstract
We have examined the relationship between chemotherapy-induced mitotic catastrophe and cell death by apoptosis in both wild-type and p53(-/-) HCT116 human colon carcinoma cells treated with nanomolar concentrations of paclitaxel (PTX), a drug that acts on tubulin altering the normal development of mitosis. After treatment, HCT116 cells entered mitosis regardless of the presence of functional p53, which resulted in changes in the distribution of cells in the different phases of the cell cycle, and in cell death. In the presence of PTX, the percentage of polyploid cells observed was higher in p53-deficient cells, indicating that mitotic slippage was favored compared to wild-type cells, with the presence of large multinucleate cells. PTX caused mitotic catastrophe and about 50-60% cells that were entering an aberrant mitosis died through an apoptotic-like pathway characterized by the presence of phosphatidylserine in the outer cell membrane, which occurred in the absence of significant activation of caspases. Lack of p53 facilitated endoreduplication and polyploidy in PTX-treated cells, but cells were still killed with similar efficacy through the same apoptotic-like mechanism in the absence of caspase activity.
Collapse
Affiliation(s)
- Laia Llovera
- Instituto de Biologia Molecular de Barcelona, CSIC, Parc Cientific de Barcelona, Barcelona, Spain
| | | | | |
Collapse
|
19
|
Babbio F, Castiglioni I, Cassina C, Gariboldi MB, Pistore C, Magnani E, Badaracco G, Monti E, Bonapace IM. Knock-down of methyl CpG-binding protein 2 (MeCP2) causes alterations in cell proliferation and nuclear lamins expression in mammalian cells. BMC Cell Biol 2012; 13:19. [PMID: 22783988 PMCID: PMC3477090 DOI: 10.1186/1471-2121-13-19] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Accepted: 07/03/2012] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND MeCP2 (CpG-binding protein 2) is a nuclear multifunctional protein involved in several cellular processes, like large-scale chromatin reorganization and architecture, and transcriptional regulation. In recent years, a non-neuronal role for MeCP2 has emerged in cell growth and proliferation. Mutations in the MeCP2 gene have been reported to determine growth disadvantages in cultured lymphocyte cells, and its functional ablation suppresses cell growth in glial cells and proliferation in mesenchymal stem cells and prostate cancer cells. MeCP2 interacts with lamin B receptor (LBR) and with Heterochromatin Protein 1 (HP1) at the nuclear envelope (NE), suggesting that it could be part of complexes involved in attracting heterochromatin at the nuclear periphery and in mediating gene silencing. The nuclear lamins, major components of the lamina, have a role in maintaining NE integrity, in orchestrating mitosis, in DNA replication and transcription, in regulation of mitosis and apoptosis and in providing anchoring sites for chromatin domains.In this work, we inferred that MeCP2 might have a role in nuclear envelope stability, thereby affecting the proliferation pattern of highly proliferating systems. RESULTS By performing knock-down (KD) of MeCP2 in normal murine (NIH-3 T3) and in human prostate transformed cells (PC-3 and LNCaP), we observed a strong proliferation decrease and a defect in the cell cycle progression, with accumulation of cells in S/G2M, without triggering a strong apoptotic and senescent phenotype. In these cells, KD of MeCP2 evidenced a considerable decrease of the levels of lamin A, lamin C, lamin B1 and LBR proteins. Moreover, by confocal analysis we confirmed the reduction of lamin A levels, but we also observed an alteration in the shape of the nuclear lamina and an irregular nuclear rim. CONCLUSIONS Our results that indicate reduced levels of NE components, are consistent with a hypothesis that the deficiency of MeCP2 might cause the lack of a key "bridge" function that links the peripheral heterochromatin to the NE, thereby causing an incorrect assembly of the NE itself, together with a decreased cell proliferation and viability.
Collapse
Affiliation(s)
- Federica Babbio
- Department of Theoretical and Applied Sciences, Insubria University, via A. da Giussano 10, Busto Arsizio, 21052, Italy
| | - Ilaria Castiglioni
- Department of Theoretical and Applied Sciences, Insubria University, via A. da Giussano 10, Busto Arsizio, 21052, Italy
| | - Chiara Cassina
- Department of Theoretical and Applied Sciences, Insubria University, via A. da Giussano 10, Busto Arsizio, 21052, Italy
| | - Marzia Bruna Gariboldi
- Department of Theoretical and Applied Sciences, Insubria University, via A. da Giussano 10, Busto Arsizio, 21052, Italy
| | - Christian Pistore
- Department of Theoretical and Applied Sciences, Insubria University, via A. da Giussano 10, Busto Arsizio, 21052, Italy
| | - Elena Magnani
- Department of Theoretical and Applied Sciences, Insubria University, via A. da Giussano 10, Busto Arsizio, 21052, Italy
| | - Gianfranco Badaracco
- Department of Biotechnologies and Life Sciences, Insubria University, via H. J. Dunant 3, Varese 21100, Italy
| | - Elena Monti
- Department of Theoretical and Applied Sciences, Insubria University, via A. da Giussano 10, Busto Arsizio, 21052, Italy
| | - Ian Marc Bonapace
- Department of Theoretical and Applied Sciences, Insubria University, via A. da Giussano 10, Busto Arsizio, 21052, Italy
| |
Collapse
|
20
|
Reddy MVR, Akula B, Cosenza SC, Lee CM, Mallireddigari MR, Pallela VR, Subbaiah DRCV, Udofa A, Reddy EP. (Z)-1-aryl-3-arylamino-2-propen-1-ones, highly active stimulators of tubulin polymerization: synthesis, structure-activity relationship (SAR), tubulin polymerization, and cell growth inhibition studies. J Med Chem 2012; 55:5174-87. [PMID: 22587519 DOI: 10.1021/jm300176j] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Tubulin, the major structural component of microtubules, is a target for the development of anticancer agents. A series of (Z)-1-aryl-3-arylamino-2-propen-1-one (10) were synthesized and evaluated for antiproliferative activity in cell-based assay. The most active compound (Z)-1-(2-bromo-3,4,5-trimethoxyphenyl)-3-(3-hydroxy-4-methoxyphenylamino)prop-2-en-1-one (10ae) was tested in 20 tumor cell lines including multidrug resistant phenotype and was found to induce apoptosis in all these cell lines with similar GI(50) values. Flow cytometry studies showed that 10ae arrested the cells in G2/M phase of cell cycle. In addition to G2/M block, these compounds caused microtubule stabilization like paclitaxel and induced apoptosis via activation of the caspase family. The observations made in this investigation demonstrate that (Z)-1-Aryl-3-arylamino-2-propen-1-one (10) represents a new class of microtubule-stabilizing agents.
Collapse
Affiliation(s)
- M V Ramana Reddy
- Department of Oncological Sciences, Mount Sinai School of Medicine, Icahn Medical Institute, 1425 Madison Avenue, New York, New York 10029-6514, United States.
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Scharadin TM, Jiang H, Martin S, Eckert RL. TIG3 interaction at the centrosome alters microtubule distribution and centrosome function. J Cell Sci 2012; 125:2604-14. [PMID: 22427689 DOI: 10.1242/jcs.096495] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
TIG3 is an important pro-differentiation regulator that is expressed in the suprabasal epidermis. We have shown that TIG3 activates selective keratinocyte differentiation-associated processes leading to cornified envelope formation. However, TIG3 also suppresses cell proliferation by an unknown mechanism. Our present studies suggest that cessation of growth is mediated through the impact of TIG3 on the centrosome and microtubules. The centrosome regulates microtubule function in interphase cells and microtubule spindle formation in mitotic cells. We show that TIG3 colocalizes with γ-tubulin and pericentrin at the centrosome. Localization of TIG3 at the centrosome alters microtubule nucleation and reduces anterograde microtubule growth, increases acetylation and detyrosination of α-tubulin, increases insoluble tubulin and drives the formation of a peripheral microtubule ring adjacent to the plasma membrane. In addition, TIG3 suppresses centrosome separation, but not duplication, and reduces cell proliferation. We propose that TIG3 regulates the formation of the peripheral microtubule ring observed in keratinocytes of differentiated epidermis and also has a role in the cessation of proliferation in these cells.
Collapse
Affiliation(s)
- Tiffany M Scharadin
- Department of Biochemistry, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | | | | | | |
Collapse
|
22
|
Synergistic antitumor effects of novel HDAC inhibitors and paclitaxel in vitro and in vivo. PLoS One 2011; 6:e29085. [PMID: 22194993 PMCID: PMC3241602 DOI: 10.1371/journal.pone.0029085] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Accepted: 11/21/2011] [Indexed: 12/01/2022] Open
Abstract
Preclinical studies support the therapeutic potential of histone deacetylases inhibitors (HDACi) in combination with taxanes. The efficacy of combination has been mainly ascribed to a cooperative effect on microtubule stabilization following tubulin acetylation. In the present study we investigated the effect of paclitaxel in combination with two novel HDACi, ST2782 or ST3595, able to induce p53 and tubulin hyperacetylation. A synergistic effect of the paclitaxel/ST2782 (or ST3595) combination was found in wild-type p53 ovarian carcinoma cells, but not in a p53 mutant subline, in spite of a marked tubulin acetylation. Such a synergistic interaction was confirmed in additional human solid tumor cell lines harboring wild-type p53 but not in those expressing mutant or null p53. In addition, a synergistic cytotoxic effect was found when ST2782 was combined with the depolymerising agent vinorelbine. In contrast to SAHA, which was substantially less effective in sensitizing cells to paclitaxel-induced apoptosis, ST2782 prevented up-regulation of p21WAF1/Cip1 by paclitaxel, which has a protective role in response to taxanes, and caused p53 down-regulation, acetylation and mitochondrial localization of acetylated p53. The synergistic antitumor effects of the paclitaxel/ST3595 combination were confirmed in two tumor xenograft models. Our results support the relevance of p53 modulation as a major determinant of the synergistic interaction observed between paclitaxel and novel HDACi and emphasize the therapeutic interest of this combination.
Collapse
|
23
|
Lee Y, Kim H, Lee J, Kim K. Anticancer activity of S-allylmercapto-L-cysteine on implanted tumor of human gastric cancer cell. Biol Pharm Bull 2011; 34:677-81. [PMID: 21532156 DOI: 10.1248/bpb.34.677] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Allylmercapto glutathione S-conjugate, S-allylmercapto-L-cysteine (SAMC), which is biotransformed from allyl sulfides and from naturally occurring water-soluble garlic derivatives, has been known to inhibit tumorigenesis. We found that SAMC was able to induce apoptosis in gastric cancer cells in vitro. We report that SAMC inhibited tumor growth rate by 31.36% and 37.78% at doses of 100 and 300 mg/kg, respectively. Apoptosis in the implanted tumor cells was manifested by apoptotic characteristics, including morphological changes of chromatin crescent, cell shrinkage and membrane blebbing. The apoptosis index of 100 mg/kg and 300 mg/kg of SAMC was 20.74 ± 2.50% and 30.61 ± 2.42%, respectively, by terminal deoxy-nucleotidyl transferase-mediated deoxyuridine triphosphate (dUTP) nick-end labeling (TUNEL) staining. The positive rate of B-cell lymphoma 2 (bcl-2) protein expression of control, 100 mg/kg SAMC and 300 mg/kg SAMC was 15.20 ± 1.67%, 10.94 ± 1.57%, and 8.24 ± 1.07%, respectively, by immunohistochemical staining. The positive rate of bax protein expression of control, 100 mg/kg SAMC and 300 mg/kg SAMC was 15.30 ± 1.90%, 23.18 ± 1.81%, and 25.26 ± 3.03%, respectively. We also observed decreases in bcl-2 mRNA and increases in bax mRNA by SAMC in a dose-dependent manner by reverse transcription-polymerase chain reaction (RT-PCR). These results suggest that SAMC may regulate bcl-2 and bax to induce apoptosis in transplanted tumor cells.
Collapse
Affiliation(s)
- Yongkyu Lee
- Department of Food and Biotechnology, Dongseo University, Korea.
| | | | | | | |
Collapse
|
24
|
Cucurbitacin IIa: a novel class of anti-cancer drug inducing non-reversible actin aggregation and inhibiting survivin independent of JAK2/STAT3 phosphorylation. Br J Cancer 2011; 104:781-9. [PMID: 21304528 PMCID: PMC3048206 DOI: 10.1038/bjc.2011.10] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Background: Cucurbitacin (Cuc) and triterpene-derived natural products exhibit anti-cancer potential in addition to their conspicuous anti-bacterial and anti-inflammatory activity. Recently, inhibition of Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) signaling was shown to underlie the effects of Cuc family on inducing cell death in cancer. Method: We purified Cuc IIa, the active component from the medicinal plant Hemsleya amalils Diels, which shows different structural modifications from other Cuc derivatives. We investigated the mechanisms of its inhibitory effects on cancer cells in vitro and tumour growth in vivo. Results: Cuc IIa induced the irreversible clustering of filamentous actin and arrested cell cycle by the increases in G2/M populations. Cuc IIa resulted in the reduced phospho-Histone H3 and markedly increased cleavage of poly-(ADP-ribose) polymerase or PARP, immediate upstream of DNA breakdown as the result of caspase activation, consistent with mitotic blockage-induced cell death. However, unlike other Cuc members, Cuc IIa did not suppress JAK2/STAT3 phosphorylation or alter phosphorylation of mitogen-activated protein kinases. Instead, the expression of the cell cycle-regulated Inhibitor of Apoptosis Protein (IAP) survivin was reduced. Introducing oncoprotein δ-catenin, which increased survivin expression and suppressed small GTPase RhoA, reduced efficacy of Cuc IIa to induce cell death. Supporting the effects of Cuc IIa on actin cytoskeletal signaling, RhoA phosphorylation was reduced suggesting its increased activity. Conclusion: Cuc IIa is a novel class of anti-cancer drug in suppression of cancer cell expansion by disrupting the actin cytoskeleton and directing the cell to undergo PARP-mediated apoptosis through the inhibition of survivin downstream of JAK2/STAT3.
Collapse
|
25
|
Chiou CT, Chen GS, Chen ML, Li H, Shi L, Huang XH, Dai WM, Chern JW. Synthesis of anti-microtubule N-(2-Arylindol-7-yl)benzenesulfonamide derivatives and their antitumor mechanisms. ChemMedChem 2010; 5:1489-97. [PMID: 20672297 DOI: 10.1002/cmdc.201000228] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2010] [Indexed: 01/26/2023]
Affiliation(s)
- Chun-Tang Chiou
- School of Pharmacy, National Taiwan University, Taipei 10051, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Docetaxel-Resistant Prostate Cancer Cells Remain Sensitive to S-Trityl-l-Cysteine–Mediated Eg5 Inhibition. Mol Cancer Ther 2010; 9:1730-9. [DOI: 10.1158/1535-7163.mct-09-1103] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
27
|
Ting C, Lee Y, Wong C, Wong A, Lung H, Lung M, Lo K, Wong R, Mak N. 2-Methoxyestradiol induces endoreduplication through the induction of mitochondrial oxidative stress and the activation of MAPK signaling pathways. Biochem Pharmacol 2010; 79:825-41. [DOI: 10.1016/j.bcp.2009.10.018] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2009] [Revised: 10/20/2009] [Accepted: 10/21/2009] [Indexed: 01/13/2023]
|
28
|
Creighton CJ, Fountain MD, Yu Z, Nagaraja AK, Zhu H, Khan M, Olokpa E, Zariff A, Gunaratne PH, Matzuk MM, Anderson ML. Molecular profiling uncovers a p53-associated role for microRNA-31 in inhibiting the proliferation of serous ovarian carcinomas and other cancers. Cancer Res 2010; 70:1906-15. [PMID: 20179198 DOI: 10.1158/0008-5472.can-09-3875] [Citation(s) in RCA: 207] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
MicroRNAs (miRNA) regulate complex patterns of gene expression, and the relevance of altered miRNA expression to ovarian cancer remains to be elucidated. By comprehensively profiling expression of miRNAs and mRNAs in serous ovarian tumors and cell lines and normal ovarian surface epithelium, we identified hundreds of potential miRNA-mRNA targeting associations underlying cancer. Functional overexpression of miR-31, the most underexpressed miRNA in serous ovarian cancer, repressed predicted miR-31 gene targets including the cell cycle regulator E2F2. MIR31 and CDKN2A, which encode p14(ARF) and p16(INK4A), are located at 9p21.3, a genomic region commonly deleted in ovarian and other cancers. p14(ARF) promotes p53 activity, and E2F2 overexpression in p53 wild-type cells normally leads via p14(ARF) to an induction of p53-dependent apoptosis. In a number of serous cancer cell lines with a dysfunctional p53 pathway (i.e., OVCAR8, OVCA433, and SKOV3), miR-31 overexpression inhibited proliferation and induced apoptosis; however, in other lines (i.e., HEY and OVSAYO) with functional p53, miR-31 had no effect. Additionally, the osteosarcoma cell line U2OS and the prostate cancer cell line PC3 (p14(ARF)-deficient and p53-deficient, respectively) were also sensitive to miR-31. Furthermore, miR-31 overexpression induced a global gene expression pattern in OVCAR8 associated with better prognosis in tumors from patients with advanced stage serous ovarian cancer, potentially affecting many genes underlying disease progression. Our findings reveal that loss of miR-31 is associated with defects in the p53 pathway and functions in serous ovarian cancer and other cancers, suggesting that patients with cancers deficient in p53 activity might benefit from therapeutic delivery of miR-31.
Collapse
Affiliation(s)
- Chad J Creighton
- Department of Medicine, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Ding J, Han F, Shi Y. Single-prolonged stress induces apoptosis in the amygdala in a rat model of post-traumatic stress disorder. J Psychiatr Res 2010; 44:48-55. [PMID: 19586638 DOI: 10.1016/j.jpsychires.2009.06.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2008] [Revised: 05/12/2009] [Accepted: 06/02/2009] [Indexed: 12/22/2022]
Abstract
OBJECTIVE To detect the apoptosis-related Bax and Bcl-2 gene expression and apoptotic cell death in the amygdala region in the single-prolonged stress (SPS) rats. METHODS A total of 100 male Wistar rats were randomly divided into a normal control group and SPS groups of 1d, 4d, 7d, and 14d. The expression of Bax and Bcl-2 was detected using immunohistochemistry and Western Blotting; TUNEL-staining and double-labeled flow cytometry (FCM) were employed for the detection and quantification of the apoptotic cells in the amygdala; morphological change of the subcellular structure in amygdala was observed by using the transmission electron microscopy (TEM). RESULTS The ratio of Bax/Bcl-2 peaked at SPS 4d and then gradually decreased. The apoptosis peaked at SPS 4d. The TUNEL-positive cells were found in each SPS group and the TUNEL-positive cells rate peaked at SPS 4d. The morphological change of amygdala cells in each SPS group bears typical apoptotic characteristics. CONCLUSIONS In the SPS rat brain, we found apoptotic process in the amygdala region which may relate to the pathogenesis of amygdala abnormal function in PTSD.
Collapse
Affiliation(s)
- Jinlan Ding
- Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China
| | | | | |
Collapse
|
30
|
Holtan SG, Creedon DJ, Haluska P, Markovic SN. Cancer and Pregnancy: Parallels in Growth, Invasion, and Immune Modulation and Implications for Cancer Therapeutic Agents. Mayo Clin Proc 2009. [DOI: 10.4065/84.11.985] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
31
|
Holtan SG, Creedon DJ, Haluska P, Markovic SN. Cancer and pregnancy: parallels in growth, invasion, and immune modulation and implications for cancer therapeutic agents. Mayo Clin Proc 2009; 84:985-1000. [PMID: 19880689 PMCID: PMC2770910 DOI: 10.1016/s0025-6196(11)60669-1] [Citation(s) in RCA: 221] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Many proliferative, invasive, and immune tolerance mechanisms that support normal human pregnancy are also exploited by malignancies to establish a nutrient supply and evade or edit the host immune response. In addition to the shared capacity for invading through normal tissues, both cancer cells and cells of the developing placenta create a microenvironment supportive of both immunologic privilege and angiogenesis. Systemic alterations in immunity are also detectable, particularly with respect to a helper T cell type 2 polarization evident in advanced cancers and midtrimester pregnancy. This review summarizes the similarities between growth and immune privilege in cancer and pregnancy and identifies areas for further investigation. Our PubMed search strategy included combinations of terms such as immune tolerance, pregnancy, cancer, cytokines, angiogenesis, and invasion. We did not place any restrictions on publication dates. The knowledge gained from analyzing similarities and differences between the physiologic state of pregnancy and the pathologic state of cancer could lead to identification of new potential targets for cancer therapeutic agents.
Collapse
Affiliation(s)
| | | | | | - Svetomir N. Markovic
- From the Division of Hematology (S.G.H., S.N.M.), Department of Oncology (S.G.H., P.H., S.N.M.), and Department of Obstetrics and Gynecology (D.J.C.), Mayo Clinic, Rochester, MN
| |
Collapse
|
32
|
Palayoor ST, Mitchell JB, Cerna D, Degraff W, John-Aryankalayil M, Coleman CN. PX-478, an inhibitor of hypoxia-inducible factor-1alpha, enhances radiosensitivity of prostate carcinoma cells. Int J Cancer 2008; 123:2430-7. [PMID: 18729192 DOI: 10.1002/ijc.23807] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Overexpression of hypoxia-inducible factor-1alpha (HIF-1alpha) in human tumors is associated with poor prognosis and poor outcome to radiation therapy. Inhibition of HIF-1alpha is considered as a promising approach in cancer therapy. The purpose of this study was to test the efficacy of a novel HIF-1alpha inhibitor PX-478 as a radiosensitizer under normoxic and hypoxic conditions in vitro. PC3 and DU 145 prostate carcinoma cells were treated with PX-478 for 20 hr, and HIF-1alpha protein level and clonogenic cell survival were determined under normoxia and hypoxia. Effects of PX-478 on cell cycle distribution and phosphorylation of H2AX histone were evaluated. PX-478 decreased HIF-1alpha protein in PC3 and DU 145 cells. PX-478 produced cytotoxicity in both cell lines with enhanced toxicity under hypoxia for DU-145. PX-478 (20 mumol/L) enhanced the radiosensitivity of PC3 cells irradiated under normoxic and hypoxic condition with enhancement factor (EF) 1.4 and 1.56, respectively. The drug was less effective in inhibiting HIF-1alpha and enhancing radiosensitivity of DU 145 cells compared to PC3 cells with EF 1.13 (normoxia) and 1.25 (hypoxia) at 50 mumol/L concentration. PX-478 induced S/G2M arrest in PC3 but not in DU 145 cells. Treatment of PC3 and DU 145 cells with the drug resulted in phosphorylation of H2AX histone and prolongation of gammaH2AX expression in the irradiated cells. PX-478 is now undergoing Phase I clinical trials as an oral agent. Although the precise mechanism of enhancement of radiosensitivity remains to be identified, this study suggests a potential role for PX-478 as a clinical radiation enhancer.
Collapse
Affiliation(s)
- Sanjeewani T Palayoor
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | | | | | | | | | | |
Collapse
|
33
|
Singh VK, Jia Z. Targeting synuclein-gamma to counteract drug resistance in cancer. Expert Opin Ther Targets 2008; 12:59-68. [PMID: 18076370 DOI: 10.1517/14728222.12.1.59] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND Expression of synuclein-gamma (SNCG) protein is elevated in the advanced stages of many types of cancers, including ovarian, lung, liver, esophagus, colon, prostate and, in particular, breast. In breast carcinoma, SNCG is causatively linked to stimulated proliferation, metastasis and drug resistance. OBJECTIVE To establish SNCG as a potential therapeutic target and to discuss clinical use of SNCG inhibiting peptide. METHODS This review focuses on the plausible mechanisms of SNCG activity, SNCG mediated drug resistance and its inhibition. RESULTS/CONCLUSION Evidence based research shows that the aberrant expression of SNCG has a strong correlation with breast cancer progression and poor clinical outcome. A peptide based inhibitor counters activity of SNCG, which may be developed as an adjuvant therapy.
Collapse
Affiliation(s)
- Vinay K Singh
- CIHR Post-doctoral Fellow in Transdisciplinary Cancer Research, Queen's University, Department of Biochemistry, Kingston, Ontario, K7L 3N6, Canada
| | | |
Collapse
|
34
|
Zhou HB, Chen JM, Cai JT, Du Q, Wu CN. Anticancer activity of genistein on implanted tumor of human SG7901 cells in nude mice. World J Gastroenterol 2008; 14:627-31. [PMID: 18203299 PMCID: PMC2681158 DOI: 10.3748/wjg.14.627] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
AIM: To investigate genistein-induced apoptosis of implanted tumors of SG7901 cells in nude mice, and the relationship between this apoptosis and expression of Bcl-2 and Bax.
METHODS: Establishing a transplanted tumor model by injecting human SG7901 cells into subcutaneous tissue of nude mice. Genistein (0.5, 1 and 1.5 mg/kg) was directly injected adjacent to the tumor, six times at 2-d intervals. Then, changes in tumor volume were measured continuously and tumor inhibition rate of each group was calculated. We observed the morphological alterations by transmission electron microscopy (TEM), measured the apoptotic rate by the TUNEL staining method, and detected the expression of apoptosis-regulated gene Bcl-2 and bax by immunohistochemical staining and RT-PCR.
RESULTS: Genistein 0.5, 1 and 1.5 mg/kg significantly inhibited carcinoma growth when it was injected near the tumor by 10.8%, 29.9% and 39.6%, respectively. Genistein induced implanted tumor cells to undergo apoptosis, with apoptotic characteristics seen by TEM. The apoptosis index was increased progressively with increasing genistein dose (28.9% ± 1.2%, 33.8% ± 1.6% and 37.7% ± 1.2%). The positive rate of Bcl-2 protein was decreased progressively (11.9% ± 0.9%, 5.9% ± 0.7% and 4.2% ± 0.6%), and the positive rate of bax protein was increased progressively (0.9% ± 1.7%, 24.9% ± 0.8% and 29.6% ± 1.7%) by immunohistochemical staining, with increasing dose of genistein. The density of Bcl-2 mRNA decreased progressively and the density of bax mRNA increased progressively with elongation of time by RT-PCR.
CONCLUSION: Genistein was able to induce apoptosis of transplanted tumor cells. This apoptosis may be mediated by down-regulation of the apoptosis-regulated gene Bcl-2 and up-regulation of apoptosis-regulated gene bax.
Collapse
|
35
|
Zhang AL, Russell PJ, Knittel T, Milross C. Paclitaxel enhanced radiation sensitization for the suppression of human prostate cancer tumor growth via a p53 independent pathway. Prostate 2007; 67:1630-40. [PMID: 17823933 DOI: 10.1002/pros.20638] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND This study investigated the influence of p53 status on treatment using combined paclitaxel and irradiation for human prostate cancer (PC) in vitro and in vivo. METHODS Enhancement of the radiation response by paclitaxel was determined by MTT and clonogenic assays in four sublines of the human PC cell line, LNCaP, stably transfected to express different p53 mutations found in PC patients. Suppression of xenograft growth by combined paclitaxel and radiation was assessed in NOD.SCID mice in vivo. Expression of p53 and downstream functional proteins, p21 and Bax, was assessed by Western blotting. RESULTS Paclitaxel (8-10 nM) suppressed cell proliferation by 50% by inducing G2M mitotic arrest in LNCaP cell lines transfected to overexpress wild-type or mutant p53. Exposure to 20 nM paclitaxel before radiation therapy enhanced cytotoxicity in clonogenic assays. The dose and duration of paclitaxel exposure were important in inducing both G2M arrest and cell growth suppression and were critical factors in paclitaxel/irradiation combination therapy. Western blotting indicated that combination therapy increased p21 protein expression to varying degrees in all cell lines. In vivo studies indicated that paclitaxel pre-treatment followed by irradiation significantly suppressed tumor growth compared with either treatment alone. CONCLUSIONS Pre-treatment with paclitaxel enhances radiation efficacy on cell killing and suppression of growth of human PC cell lines in vitro and in vivo via p53 independent pathways. Paclitaxel has potential for use as a radiosensitizer in the treatment of patients with PC with either wild-type or mutant p53 genetic status.
Collapse
Affiliation(s)
- An Ling Zhang
- Oncology Research Centre, Prince of Wales Hospital, Randwick, NSW, Australia
| | | | | | | |
Collapse
|
36
|
Copeland RL, Das JR, Kanaan YM, Taylor RE, Tizabi Y. Antiapoptotic effects of nicotine in its protection against salsolinol-induced cytotoxicity. Neurotox Res 2007; 12:61-9. [PMID: 17513200 DOI: 10.1007/bf03033901] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Salsolinol (1-methyl-6,7-dihydroxy-1,2,3,4-tetrahydroisoquinoline), a metabolite of dopamine, may act as an endogenous neurotoxin and contribute to the etiology of Parkinson's disease (PD). The inverse relationship between smoking and PD prompted our previous investigation and the report of protective effects of nicotine against salsolinol-induced toxicity in cultured SH-SY5Y cells (Copeland et al., Neurotox. Res. 8:289, 2005). These cells, derived from human neuroblastoma cells, express dopaminergic activity and are used as a model of nigral dopaminergic cells, the major site of pathology in PD. The purpose of the current study was to investigate whether apoptotic or antiapoptotic mechanisms were responsible for the observed effects of salsolinol and nicotine, respectively. Moreover, it was of interest to determine whether the actions of nicotine are mediated through nicotinic receptors. SH-SY5Y cells were exposed to 0.4 or 0.7 mM salsolinol with and without pretreatment in combination of 0.1 mM nicotine and 0.1 mM mecamylamine and were exposed for 24 and 48 h. Various parameters including cell cycle perturbations (reflected in propidium iodide DNA staining); cell cycle regulator retinoblastoma protein (reflected in the Western blot), apoptosis (reflected in annexin V/propidium iodide staining followed by flow cytometry) were analyzed. Salsolinol caused an arrest of the cells in G1-phase of cell cycle and an increase in apoptotic indices, whereas pretreatment with nicotine attenuated or completely blocked the effects of salsolinol. Nicotine effects in turn, were totally blocked by mecamylamine (0.1 mM). The results suggest that apoptosis is a major mechanism for salsolinol-induced toxicity and that antiapoptotic effects of nicotine, mediated by nicotinic receptors, may play a primary role in its neuroprotective effects. Hence, nicotinic agonists in combination with other antiapoptotic agents may be of substantial benefit in at least a subpopulation of Parkinson patients.
Collapse
Affiliation(s)
- Robert L Copeland
- Department of Pharmacology, College of Medicine, Howard University, Washington, DC 20059, USA
| | | | | | | | | |
Collapse
|
37
|
Chen KC, Peng CC, Peng RY, Su CH, Chiang HS, Yan JH, Hsieh-Li HM. Unique formosan mushroom Antrodia camphorata differentially inhibits androgen-responsive LNCaP and -independent PC-3 prostate cancer cells. Nutr Cancer 2007; 57:111-21. [PMID: 17516868 DOI: 10.1080/01635580701268360] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Antrodia camphorata (AC), a precious and unique folkloric medicinal mushroom enriched in polyphenolics, isoflavonoids, triterpenoids, and polysaccharides, has been diversely used in Formosa (Taiwan) since the 18th century. In this study, prostate cancer (PCa) cell lines PC-3 (androgen independent) and LNCaP (androgen responsive) were treated with AC crude extract (ACCE) at 50-200 microg/mL, respectively, for 48 h. At the minimum effective dose 150 microg/mL, LNCaP showed a G1/S phase arrest with significant apoptosis. Such dose-dependent behavior of LNCaP cells in response to ACCE was confirmed to proceed as Akt-->p53-->p21-->CDK4/cyclin D1-->G1/S-phase arrest-->apoptosis, which involved inhibiting cyclin D1 activity and preventing pRb phosphorylation. In contrast, being without p53, PC-3 cells showed a G2/M-phase arrest mediated through pathway p21-->cyclin B1/Cdc2-->G2/M-phase arrest, however, with limited degree of apoptosis, implicating that ACCE is able to differentially inhibit the growth of different PCa cells by modulating different cell cycle signaling pathways. We conclude that this unique Formosan mushroom, A. camphorata, due to its nontoxicity, might be used as a good adjuvant anticancer therapy for prostate cancers despite its androgen-responsive behaviors, which has long been a serious drawback often encountered clinically in hormonal refractory cases treated by antihormonal therapies and chemotherapeutics.
Collapse
Affiliation(s)
- Kuan-Chou Chen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | | | | | | | | | | | | |
Collapse
|
38
|
Sharma A, Comstock CE, Knudsen ES, Cao KH, Hess-Wilson JK, Morey LM, Barrera J, Knudsen KE. Retinoblastoma tumor suppressor status is a critical determinant of therapeutic response in prostate cancer cells. Cancer Res 2007; 67:6192-203. [PMID: 17616676 PMCID: PMC4133940 DOI: 10.1158/0008-5472.can-06-4424] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The retinoblastoma tumor suppressor protein (RB), a critical mediator of cell cycle progression, is functionally inactivated in the majority of human cancers, including prostatic adenocarcinoma. The importance of RB tumor suppressor function in this disease is evident because 25% to 50% of prostatic adenocarcinomas harbor aberrations in RB pathway. However, no previous studies challenged the consequence of RB inactivation on tumor cell proliferation or therapeutic response. Here, we show that RB depletion facilitates deregulation of specific E2F target genes, but does not confer a significant proliferative advantage in the presence of androgen. However, RB-deficient cells failed to elicit a cytostatic response (compared with RB proficient isogenic controls) when challenged with androgen ablation, AR antagonist, or combined androgen blockade. These data indicate that RB deficiency can facilitate bypass of first-line hormonal therapies used to treat prostate cancer. Given the established effect of RB on DNA damage checkpoints, these studies were then extended to determine the impact of RB depletion on the response to cytotoxic agents used to treat advanced disease. In this context, RB-deficient prostate cancer cells showed enhanced susceptibility to cell death induced by only a selected subset of cytotoxic agents (antimicrotubule agents and a topoisomerase inhibitor). Combined, these data indicate that RB depletion dramatically alters the cellular response to therapeutic intervention in prostate cancer cells and suggest that RB status could potentially be developed as a marker for effectively directing therapy.
Collapse
Affiliation(s)
- Ankur Sharma
- Departments of Cell and Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Clay E.S. Comstock
- Departments of Cell and Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Erik S. Knudsen
- Departments of Cell and Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Center for Environmental Genetics, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Barrett Cancer Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Khanh H. Cao
- Department of Biomedical Engineering, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Janet K. Hess-Wilson
- Departments of Cell and Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Lisa M. Morey
- Departments of Cell and Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Jason Barrera
- Departments of Psychiatry, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Karen E. Knudsen
- Departments of Cell and Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Center for Environmental Genetics, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Barrett Cancer Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
39
|
Supino R, Favini E, Cuccuru G, Zunino F, Scovassi AI. Effect of paclitaxel on intracellular localization of c-Myc and P-c-Myc in prostate carcinoma cell lines. Ann N Y Acad Sci 2007; 1095:175-81. [PMID: 17404030 DOI: 10.1196/annals.1397.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The proto-oncogene c-myc is involved in multiple cell pathways with opposite effects on cell outcome of death or proliferation. It has been proposed that these different roles depend on the sequestration of c-Myc protein in cellular compartments and/or its phosphorylation. We speculated that subcellular localization of c-Myc protein and of its phosphorylated form (P-c-Myc) could have a role in the different response to paclitaxel (PTX) in two prostate carcinoma cell lines, PC3 and DU145, which undergo either multinucleation or c-myc-dependent apoptosis, respectively. c-myc is amplified only in PC3, but a similar extent of c-Myc phosphorylation was observed in both cell lines after PTX treatment. We found that PTX-induced upregulation of c-myc in DU145 cells, not occurring in PC3 cells, cannot be ascribed to a different protein localization, and that a comparable c-Myc and P-c-Myc nuclear translocation occurs in both cell lines after drug treatment. Thus, subcellular localization of c-Myc and P-c-Myc is not crucial in determining the mode of cell death in these prostate carcinoma cell lines.
Collapse
Affiliation(s)
- Rosanna Supino
- Istituto Nazionale Tumori, Via Venezian 1, 20133 Milano, Italy.
| | | | | | | | | |
Collapse
|
40
|
Gandellini P, Folini M, Bandiera R, De Cesare M, Binda M, Veronese S, Daidone MG, Zunino F, Zaffaroni N. Down-regulation of human telomerase reverse transcriptase through specific activation of RNAi pathway quickly results in cancer cell growth impairment. Biochem Pharmacol 2007; 73:1703-14. [PMID: 17321502 DOI: 10.1016/j.bcp.2007.01.035] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2006] [Revised: 01/23/2007] [Accepted: 01/24/2007] [Indexed: 01/09/2023]
Abstract
Targeting of human telomerase reverse transcriptase (hTERT) by different small interfering RNAs (siRNAs) resulted in a variable degree of telomerase activity inhibition in PC-3 and DU145 prostate cancer cells. In addition, transfection with siRNA5 and siRNA41, which caused high levels ( approximately 80 and approximately 55%, respectively) of enzyme activity inhibition in both cell lines, led to a marked reduction of hTERT mRNA and protein expression and a significant inhibition of cell proliferation within a few days, without concomitant telomere shortening or telomeric 3' overhang impairment. Such an antiproliferative effect was not ascribable to the activation of non-specific responses, since siRNA5 and siRNA41 did not induce the expression of 2'-5' oligoadenylate synthetase-1 and were able to cause a significant growth impairment also in HCT 116 colon cancer cells, which have a defective interferon pathway. Cell growth inhibition was indeed associated with hTERT down-regulation, as it was almost completely rescued in siRNA-treated HCT 116 cells co-transfected with an hTERT-expressing vector. Moreover, siRNA5 and siRNA41 failed to affect the proliferation of hTERT-negative U2-OS osteosarcoma cells. Interestingly, transfection with siRNA5 significantly reduced the tumorigenic and growth potential of PC-3 cells when xenotransplanted into nude mice. Such data suggest siRNA-mediated hTERT down-regulation as an efficient strategy to impair prostate cancer cell growth.
Collapse
Affiliation(s)
- Paolo Gandellini
- Department of Experimental Oncology and Laboratories, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Perego P, Ciusani E, Gatti L, Carenini N, Corna E, Zunino F. Sensitization to gimatecan-induced apoptosis by tumor necrosis factor-related apoptosis inducing ligand in prostate carcinoma cells. Biochem Pharmacol 2006; 71:791-8. [PMID: 16438941 DOI: 10.1016/j.bcp.2005.12.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2005] [Revised: 12/19/2005] [Accepted: 12/19/2005] [Indexed: 11/27/2022]
Abstract
Since the intrinsic resistance of prostate carcinoma likely reflects a low susceptibility to drug-induced apoptosis, in this study we explored the possibility of sensitizing prostate carcinoma cells to apoptosis by combination of TRAIL with camptothecins. Indeed, these agents are known to activate different pathways of apoptosis. Topotecan- and gimatecan induced moderate up-regulation of TRAIL-R1 and -R2 which resulted in a different cell response to the combination in androgen-independent cells (DU-145 and PC-3). In DU-145 cells apoptosis was increased by lower TRAIL concentrations and was earlier than in PC-3 cells, as shown using Annexin V-binding assay. The relative resistance of PC-3 cells to drug-induced apoptosis was associated with constitutive Akt activation, higher levels of cFLIP-L and Bcl-2, and lower levels of Bax. The different expression/activation of apoptosis-related factors appears to influence the sensitization of prostate carcinoma cells by TRAIL. Potentiation of camptothecin-induced apoptosis by TRAIL appears dependent on cooperation between extrinsic and intrinsic pathways, as documented by loss of the sensitization to apoptosis following reduction of caspase 8 after small interfering RNA transfection. The efficacy of the approach may be critically dependent on the intrinsic susceptibility to apoptosis of different tumors. These observations support that the activation of multiple signals could enhance apoptotic response and suggest the therapeutic interest of the TRAIL/camptothecin combination.
Collapse
Affiliation(s)
- Paola Perego
- Preclinical Chemotherapy and Pharmacology Unit, Department of Experimental Oncology and Laboratories, Istituto Nazionale per lo Studio e la Cura dei Tumori, Via Venezian 1, 20133 Milan, Italy.
| | | | | | | | | | | |
Collapse
|
42
|
Plasencia C, Dayam R, Wang Q, Pinski J, Burke TR, Quinn DI, Neamati N. Discovery and preclinical evaluation of a novel class of small-molecule compounds in hormone-dependent and -independent cancer cell lines. Mol Cancer Ther 2005; 4:1105-13. [PMID: 16020668 DOI: 10.1158/1535-7163.mct-04-0288] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We discovered a series of salicylhydrazide class of compounds with remarkable anticancer activity against a panel of hormone receptor-positive and -negative cell lines. In the present study, we evaluated the in vitro activity of SC21 and SC23 against a range of human tumor cell types and the in vivo efficacy of compound SC21 in a PC3 human prostate cancer xenograft model in mice. We also determined the effects of SC21 on cell cycle regulation and apoptosis. Our in vitro results show that salicylhydrazides are highly potent compounds effective in both hormone receptor-positive and -negative cancer cells. SC21 induced apoptosis and blocked the cell cycle in G(0)/G(1) or S phase, depending on the cell lines used and irrespective of p53, p21, pRb, and p16 status. SC21 effectively reduced the tumor growth in mice without apparent toxicity. Although the mechanism of action of SC21 is not completely elucidated, the effect on cell cycle, the induction of apoptosis and the activity against a panel of tumor cell lines of different origins prompted us to carry out an in-depth preclinical evaluation of SC21.
Collapse
Affiliation(s)
- Carmen Plasencia
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, 90089, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Simoens C, Vermorken JB, Korst AEC, Pauwels B, De Pooter CMJ, Pattyn GGO, Lambrechts HAJ, Breillout F, Lardon F. Cell cycle effects of vinflunine, the most recent promising Vinca alkaloid, and its interaction with radiation, in vitro. Cancer Chemother Pharmacol 2005; 58:210-8. [PMID: 16317558 DOI: 10.1007/s00280-005-0147-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2005] [Accepted: 10/12/2005] [Indexed: 01/07/2023]
Abstract
PURPOSE Vinflunine (VFL) is a novel third generation Vinca alkaloid with superior antitumour activity in preclinical models and an anticipated more favourable toxicity profile compared to the other Vinca alkaloids. METHOD We investigate the radiosensitising properties of VFL and its cell cycle effects in four human tumour cell lines (ECV304, MCF-7, H292, and CAL-27). The sulforhodamine B test was used to determine cell survival, and cell cycle analysis was performed by flow cytometry. Radiosensitisation (RS) was represented by dose enhancement factors (DEFs). RESULTS Twenty-four hours treatment with VFL before radiation caused dose-dependent RS in all cell lines. This was most pronounced in ECV304 cells with RS already at VFL concentrations that reduced cell survival by 10% (IC10). DEFs ranged from 1.57 to 2.29 in the different cell lines. A concentration-dependent G2/M block was observed (starting at 4 h of incubation). After maximal G2/M blockade cells started cycling again, mainly by mitosis, while a small portion of cells started a polyploid cell cycle. Also drug removal immediately caused recycling of cells and induction of a polyploid cell population. The polyploid cell population was most impressively noticeable after prolonged incubation with VFL (48 h), in particular in CAL-27 and ECV304. This was never observed in a tested normal fibroblast cell line (Fi 360). The fate of these cells is of particular interest, but yet uncertain. CONCLUSION VFL has radiosensitising potential. The exact role of the cell cycle effects of VFL in its radiosensitising mechanism is still not fully elucidated and requires further study.
Collapse
Affiliation(s)
- Cindy Simoens
- Laboratory of Cancer Research and Clinical Oncology, Department of Medical Oncology, University of Antwerp, 2610 Wilrijk, Belgium.
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Ramanathan B, Jan KY, Chen CH, Hour TC, Yu HJ, Pu YS. Resistance to paclitaxel is proportional to cellular total antioxidant capacity. Cancer Res 2005; 65:8455-60. [PMID: 16166325 DOI: 10.1158/0008-5472.can-05-1162] [Citation(s) in RCA: 214] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Paclitaxel, one of the most commonly prescribed chemotherapeutic agents, is active against a wide spectrum of human cancer. The mechanism of its cytotoxicity, however, remains controversial. Our results indicate that paclitaxel treatment increases levels of superoxide, hydrogen peroxide, nitric oxide (NO), oxidative DNA adducts, G2-M arrest, and cells with fragmented nuclei. Antioxidants pyruvate and selenium, the NO synthase inhibitor N(omega)-nitro-L-arginine methyl ester, and the NO scavenger manganese (III) 2-(4-carboxyphenyl)-4,4,5,5-tetramethyl-imidazoline-1-oxyl-3-oxide all decreased paclitaxel-mediated DNA damage and sub-G1 cells. In contrast, the glutamylcysteine synthase inhibitor buthionine sulfoximine (BSO) and the superoxide dismutase (SOD) inhibitor 2-methoxyestradiol (2-ME) increased the sub-G1 fraction in paclitaxel-treated cells. These results suggest that reactive oxygen and nitrogen species are involved in paclitaxel cytotoxicity. This notion is further supported with the observation that concentrations of paclitaxel required to inhibit cell growth by 50% correlate with total antioxidant capacity. Moreover, agents such as arsenic trioxide (As2O3), BSO, 2-ME, PD98059, U0126 [mitogen-activated protein/extracellular signal-regulated kinase inhibitors], and LY294002 (phosphatidylinositol 3-kinase/Akt inhibitor), all of which decrease clonogenic survival, also decrease the total antioxidant capacity of paclitaxel-treated cells, regardless whether they are paclitaxel sensitive or paclitaxel resistant. These results suggest that paclitaxel chemosensitivity may be predicted by taking total antioxidant capacity measurements from clinical tumor samples. This, in turn, may then improve treatment outcomes by selecting out potentially responsive patients.
Collapse
|
45
|
Toiyama Y, Tanaka K, Konishi N, Mohri Y, Tonouchi H, Miki C, Kusunoki M. Administration sequence-dependent antitumor effects of paclitaxel and 5-fluorouracil in the human gastric cancer cell line MKN45. Cancer Chemother Pharmacol 2005; 57:368-75. [PMID: 16075279 DOI: 10.1007/s00280-005-0057-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2005] [Accepted: 05/24/2005] [Indexed: 10/25/2022]
Abstract
BACKGROUND The clinical outcome of gastric cancer patients has been improved by combination of 5-fluorouracil (5-FU) and paclitaxel (PXL). However, the optimal schedule of this combination has not been determined. METHODS The efficacies of sequential administrations of 5-FU and PXL on the gastric cancer cell line MKN45 were investigated using a WST-8 colorimetric assay. The cell cycle distribution of each drug was evaluated by flow-cytometry. Furthermore, the mechanism of antitumor activity enhancement by the administration sequence was investigated by western blotting. RESULTS MKN45 cell growth was significantly inhibited by each drug in a dose- and time-dependent manner. The cytotoxicities of PXL followed by 5-FU were significantly greater than those of 5-FU followed by PXL. The flow-cytometric analysis revealed that PXL exposure caused viable cell accumulation in G2/M phase in a dose-dependent manner. Western blotting showed that PXL exposure followed by 5-FU up-regulated Chk1 and Wee1 protein expressions until PXL removal and 5-FU exposure, when these expressions gradually decreased to their basal levels. 14-3-3sigma protein expression was significantly up-regulated upon PXL treatment followed by 5-FU. Interestingly, Mad2 protein expression with PXL treatment followed by 5-FU gradually increased after the PXL removal and 5-FU exposure. CONCLUSIONS PXL followed by 5-FU administration may be the optimal sequence for treatment of gastric cancer. The enhanced viable cell accumulation after PXL pretreatment may be related to G2 arrest. After PXL removal and 5-FU exposure, the cells progressing to M phase may undergo cell death by mitotic catastrophe due to DNA damage caused by 5-FU exposure.
Collapse
Affiliation(s)
- Yuji Toiyama
- The Second Department of Surgery, Mie University School of Medicine, Tsu, Mie, Japan
| | | | | | | | | | | | | |
Collapse
|
46
|
Zhou HB, Yan Y, Cai JT, Du Q, Chen JM. Genistein induces apoptosis in human primary gastric carcinoma cells. Shijie Huaren Xiaohua Zazhi 2005; 13:504-507. [DOI: 10.11569/wcjd.v13.i4.504] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the apoptosis in primary gastric cancer cells induced by genistein, and its relationship with bcl-2 and bax.
METHODS: MTT assay was used to determine cell growth inhibition. Transmission electron microscope and TUNEL staining were used to quantitatively and qualitatively detect apoptosis. Immunohistochemistry and RT-PCR were used to detect the expression of apoptosis-regulated genes bcl-2 and bax.
RESULTS: Genistein inhibited the growth of primary gastric cancer cells in a dose- and time-dependent manner. Genistein induced primary gastric cancer cells to undergo apoptosis with typically apoptotic characteristics. TUNEL assay showed that the percentage of apoptotic cells was increased gradually along with the time of genistein treatment(1.25±0.30%, 4.97±0.80%, 18.44±1.92%, 35.18±0.35%, and 43.93±1.11% at 0, 24, 48, 72 and 96 h after treatment, respectively, P<0.05). The percentage of bcl-2 protein positive cells was significantly reduced (36.34±0.72%, 21.62±0.08%, 10.60±0.49%,7.21±0.45%,and 4.540.36% at 0, 24, 48, 72 and 96 h after treatment, respectively, P<0.01), whereas the percentage of bax protein positive cells was markedly increased (10.73±0.57%, 20.630.86%, 34.3±0.81%, 45.96±0.42%, and 58.61±1.46%, at 0, 24, 48, 72 and 96 h after treatment, respectively, P<0.01). After exposed to 20 mmol/L genistein for 24,48,72 and 96 h, bcl-2 mRNA was decreased, while bax mRNA was increased progressively with elongation of genistein treatment time.
CONCLUSION: Genistein can induce apoptosis in primary gastric cancer, which may be mediated by down-regulating the apoptosis-regulated gene bcl-2 and up-regulating the apoptosis-regulated gene bax.
Collapse
|
47
|
Zhou HB, Chen JJ, Wang WX, Cai JT, Du Q. Anticancer activity of resveratrol on implanted human primary gastric carcinoma cells in nude mice. World J Gastroenterol 2005; 11:280-4. [PMID: 15633232 PMCID: PMC4205418 DOI: 10.3748/wjg.v11.i2.280] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the apoptosis of implanted primary gastric cancer cells in nude mice induced by resveratrol and the relation between this apoptosis and expression of bcl-2 and bax.
METHODS: A transplanted tumor model was established by injecting human primary gastric cancer cells into subcutaneous tissue of nude mice. Resveratrol (500 mg/kg, 1000 mg/kg and 1500 mg/kg) was directly injected beside tumor body 6 times at an interval of 2 d. Then changes of tumor volume were measured continuously and tumor inhibition rate of each group was calculated. We observed the morphologic alterations by electron microscope, measured the apoptotic rate by TUNEL staining method, detected the expression of apoptosis-regulated genes bcl-2 and bax by immunohistoch-emical staining and PT-PCR.
RESULTS: Resveratrol could significantly inhibit carcinoma growth when it was injected near the carcinoma. An inhibitory effect was observed in all therapeutic groups and the inhibition rate of resveratrol at the dose of 500 mg/kg, 1000 mg/kg and 1500 mg/kg was 10.58%, 29.68% and 39.14%, respectively. Resveratrol induced implanted tumor cells to undergo apoptosis with apoptotic characteristics, including morphological changes of chromatin condensation, chromatin crescent formation, nucleus fragmentation. The inhibition rate of 0.2 mL of normal saline solution, 1 500 mg/kg DMSO, 500 mg/kg resveratrol, 1000 mg/kg resveratrol, and 1500 mg/kg resveratrol was 13.68±0.37%, 13.8±0.43%, 48.7±1.07%, 56.44±1.39% and 67±0.96%, respectively. The positive rate of bcl-2 protein of each group was 29.48±0.51%, 27.56±1.40%, 11.86±0.97%, 5.7±0.84% and 3.92±0.85%, respectively by immunohistochemical staining. The positive rate of bax protein of each group was 19.34±0.35%, 20.88±0.91%, 40.02±1.20%, 45.72±0.88% and 52.3±1.54%, respectively by immunohistochemical staining. The density of bcl-2 mRNA in 0.2 mL normal saline solution, 1500 mg/kg DMSO, 500 mg/kg resveratrol, 1000 mg/kg resveratrol, and 1500 mg/kg resveratrol decreased progressively and the density of bax mRNA in 0.2 mL normal saline solution, 1500 mg/kg DMSO, 500 mg/kg resveratrol, 1000 mg/kg resveratrol, and 1500 mg/kg increased progressively with elongation of time by RT-PCR.
CONCLUSION: Resveratrol is able to induce apoptosis of transplanted tumor cells. This apoptosis may be mediated by down-regulating apoptosis-regulated gene bcl-2 and up-regulating the expression of apoptosis-regulated gene bax.
Collapse
Affiliation(s)
- Hai-Bo Zhou
- Department of Gastroenterology, Second Hospital of Zhejiang University, Hangzhou 310009, Zhejiang Province, China.
| | | | | | | | | |
Collapse
|
48
|
Ioffe ML, White E, Nelson DA, Dvorzhinski D, DiPaola RS. Epothilone induced cytotoxicity is dependent on p53 status in prostate cells. Prostate 2004; 61:243-7. [PMID: 15368466 DOI: 10.1002/pros.20108] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
BACKGROUND Epothilones are a promising class of drugs in clinical trials of prostate cancer that target the microtubules, similar to taxanes, and induce apoptosis in taxane resistant tumors. The tumor suppressor p53 is one important molecular mechanism of chemotherapy resistance that in some studies predicted tumor sensitivity to paclitaxel. We hypothesized that epothilone induced cytotoxicity would be influenced by the status of p53 in prostate cells. METHODS LNCaP, DU145, and a transformed rat prostate (RP) epithelial cell line with a temperature sensitive mutant p53 (val 135) were studied for the effect of epothilone on cell viability, cell cycle, and cell cycle checkpoint proteins. RESULTS Epothilone had greater cytotoxicity in p53 mutant cancer cells compared to wild type cells. We confirmed our findings by creating a transformed RP epithelial cell line with a temperature sensitive mutant p53 (val 135). Using a tetrazolium (MTT) assay we found that epothilone (100 nM) decreased cell viability in RP cells by 90% with mutant p53 compared to 45% with wild type p53 (P < 0.01). Epothilone induced G2/M arrest in 50% of cells with mutant p53 compared to 25% with wild type p53 (P < 0.01). To begin to understand mechanism of epothilone induced G2/M arrest, we assessed cell cycle checkpoint proteins. We found that the effect to enhance G2/M cell cycle arrest was associated with dephosphorylation of cdc2 in both p53 wild type and p53 mutant RP cells. CONCLUSIONS These results demonstrate that epothilone is more active against transformed prostate epithelial cells with mutant compared to wild type p53. Epothilone is capable of dephosphorylation of cdc2 in both p53 wild type and mutant cells, which is associated with G2/M cell cycle arrest. These data provide a basis for further study of p53, and the phosphorylation status of cdc-2, as markers for epothilone sensitivity in clinical studies.
Collapse
Affiliation(s)
- Margarita L Ioffe
- The Cancer Institute of New Jersey, Robert Wood Johnson Medical School/UMDNJ, New Brunswick, New Jersey 08901, USA
| | | | | | | | | |
Collapse
|
49
|
Cassinelli G, Supino R, Zuco V, Lanzi C, Scovassi AI, Semple SC, Zunino F. Role of c-myc protein in hormone refractory prostate carcinoma: cellular response to paclitaxel. Biochem Pharmacol 2004; 68:923-31. [PMID: 15294455 DOI: 10.1016/j.bcp.2004.06.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2004] [Accepted: 06/07/2004] [Indexed: 11/26/2022]
Abstract
Amplification of the c-MYC proto-oncogene is a frequent alteration in hormone refractory prostate carcinomas (HRPC). In an attempt to investigate the role of c-myc in the cellular response to paclitaxel (PTX), we used two HRPC cell lines, DU145 and PC3, characterised by different levels of the protein and by different behaviour in response to taxane. In both cell lines, PTX-induced cell death was a caspase-mediated apoptosis. In DU145 cells, PTX induced an early apoptotic response associated with upregulation of c-myc restricted to the G2/M cell population. This event appeared delayed in the presence of c-myc antisense (AS-c-myc), suggesting an upstream regulation of the protein expression. In addition, the antisense approach provided evidence of an involvement of c-myc in the apoptotic response to the taxane. In contrast, in PC3 cells, the overexpressed c-myc was not modulated by drug-treatment and the addition of AS-c-myc did not affect the cell growth inhibition of PTX. In both cell lines, PTX-induced c-myc phosphorylation was concomitant with the mitotic arrest and not related to the modulation of the activation state of AKT and MAPK kinases. Our data indicate that the cellular response to PTX of HRPC cells can involve c-myc and suggest that its pro-apoptotic role is affected by the genetic background, thus supporting a complex and differentiated HRPC cell response to taxanes.
Collapse
|
50
|
Zhou HB, Chen JJ, Wang WX, Cai JT, Du Q. Apoptosis of human primary gastric carcinoma cells induced by genistein. World J Gastroenterol 2004; 10:1822-5. [PMID: 15188515 PMCID: PMC4572278 DOI: 10.3748/wjg.v10.i12.1822] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
AIM: To investigate the apoptosis in primary gastric cancer cells induced by genistein, and the relationship between this apoptosis and expression of bcl-2 and bax.
METHODS: MTT assay was used to determine the cell growth inhibitory rate in vitro. Transmission electron microscope and TUNEL staining were used to quantitatively and qualitatively detect the apoptosis of primary gastric cancer cells before and after genistein treatment. Immunohistochemical staining and RT-PCR were used to detect the expression of apoptosis-associated genes bcl-2 and bax.
RESULTS: Genistein inhibited the growth of primary gastric cancer cells in dose-and time-dependent manner. Genistein induced primary gastric cancer cells to undergo apoptosis with typically apoptotic characteristics. TUNEL assay showed that after the treatment of primary gastric cancer cells with genistein for 24 to 96 h, the apoptotic rates of primary gastric cancer cells increased time-dependently. Immunohistochemical staining showed that after the treatment of primary gastric cancer cells with genistein for 24 to 96 h, the positivity rates of Bcl-2 proteins were apparently reduced with time and the positivity rates of Bax proteins were apparently increased with time. After exposed to genistein at 20 μmol/L for 24, 48, 72 and 96 respectively, the density of bcl-2 mRNA decreased progressively and the density of bax mRNA increased progressively with elongation of time.
CONCLUSION: Genistein is able to induce the apoptosis in primary gastric cancer cells. This apoptosis may be mediated by down-regulating the apoptosis- associated bcl-2 gene and up-regulating the expression of apoptosis-associated bax gene.
Collapse
Affiliation(s)
- Hai-Bo Zhou
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University, Hangzhou 310009, Zhejiang Province, China.
| | | | | | | | | |
Collapse
|