1
|
Cavalcanti BC, Soares BM, Barreto FS, Magalhães HIF, Ferreira JRDO, Almeida ATAD, Araújo Beserra Filho JI, Silva J, Dos Santos HS, Marinho ES, Furtado CLM, Moraes Filho MOD, Pessoa C, Ferreira PMP. Hellebrigenin triggers death of promyelocytic leukemia cells by non-genotoxic ways. Toxicon 2024; 238:107591. [PMID: 38160738 DOI: 10.1016/j.toxicon.2023.107591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 12/12/2023] [Accepted: 12/28/2023] [Indexed: 01/03/2024]
Abstract
Bufadienolides are digitalis-like aglycones mainly found in skin secretions of toads. Among their biological properties, the mechanisms of antiproliferative action on tumor cells remain unclear for many compounds, including against leukemia cells. Herein, it was evaluated the mechanisms involved in the antiproliferative and genotoxic actions of hellebrigenin on tumor cell lines and in silico capacity to inhibit the human topoisomerase IIa enzyme. Firstly, its cytotoxic action was investigated by colorimetric assays in human tumor and peripheral blood mononuclear cells (PBMC). Next, biochemical and morphological studies were detailed by light microscopy (trypan blue dye exclusion), immunocytochemistry (BrdU uptake), flow cytometry and DNA/chromosomal damages (Cometa and aberrations). Finally, computational modelling was used to search for topoisomerase inhibition. Hellebrigenin reduced proliferation, BrdU incorporation, viability, and membrane integrity of HL-60 leukemia cells. Additionally, it increased G2/M arrest, internucleosomal DNA fragmentation, mitochondrial depolarization, and phosphatidylserine externalization in a concentration-dependent manner. In contrast to doxorubicin, hellebrigenin did not cause DNA strand breaks in HL-60 cell line and lymphocytes, and it interacts with ATPase domain residues of human topoisomerase IIa, generating a complex of hydrophobic and van der Waals interactions and hydrogen bonds. So, hellebrigenin presented potent anti-leukemic activity at concentrations as low as 0.06 μM, a value comparable to the clinical anticancer agent doxorubicin, and caused biochemical changes suggestive of apoptosis without genotoxic/clastogenic-related action, but it probably triggers catalytic inhibition of topoisomerase II. These findings also emphasize toad steroid toxins as promising lead antineoplasic compounds with relatively low cytotoxic action on human normal cells.
Collapse
Affiliation(s)
- Bruno Coêlho Cavalcanti
- Laboratory of Experimental Oncology (LOE), Drug Research and Development Center, Federal University of Ceará, Fortaleza, Brazil
| | - Bruno Marques Soares
- Laboratory of Experimental Oncology (LOE), Drug Research and Development Center, Federal University of Ceará, Fortaleza, Brazil
| | - Francisco Stefânio Barreto
- Laboratory of Experimental Oncology (LOE), Drug Research and Development Center, Federal University of Ceará, Fortaleza, Brazil
| | | | | | - Ana Tárcila Alves de Almeida
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Federal University of Piauí, Teresina, Brazil
| | - José Ivo Araújo Beserra Filho
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Federal University of Piauí, Teresina, Brazil
| | - Jacilene Silva
- Department of Biological Chemistry, Regional University of Cariri, Crato, Brazil
| | | | - Emmanuel Silva Marinho
- Group of Theoretical Chemistry and Electrochemistry, State University of Ceará, Limoeiro do Norte, Brazil
| | - Cristiana Libardi Miranda Furtado
- Laboratory of Experimental Oncology (LOE), Drug Research and Development Center, Federal University of Ceará, Fortaleza, Brazil; Experimental Biology Center, University of Fortaleza, Fortaleza, Brazil
| | - Manoel Odorico de Moraes Filho
- Laboratory of Experimental Oncology (LOE), Drug Research and Development Center, Federal University of Ceará, Fortaleza, Brazil
| | - Cláudia Pessoa
- Laboratory of Experimental Oncology (LOE), Drug Research and Development Center, Federal University of Ceará, Fortaleza, Brazil.
| | - Paulo Michel Pinheiro Ferreira
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Federal University of Piauí, Teresina, Brazil.
| |
Collapse
|
2
|
Nowak P, Bil-Lula I, Śliwińska-Mossoń M. A Cross-Talk about Radioresistance in Lung Cancer-How to Improve Radiosensitivity According to Chinese Medicine and Medicaments That Commonly Occur in Pharmacies. Int J Mol Sci 2023; 24:11206. [PMID: 37446385 DOI: 10.3390/ijms241311206] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/21/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
Lung cancer is one of the most common cancers in the population and is characterized by non-specific symptoms that delay the diagnosis and reduce the effectiveness of oncological treatment. Due to the difficult placement of the tumor, one of the main methods of lung cancer treatment is radiotherapy, which damages the DNA of cancer cells, inducing their apoptosis. However, resistance to ionizing radiation may develop during radiotherapy cycles, leading to an increase in the number of DNA points of control that protect cells from apoptosis. Cancer stem cells are essential for radioresistance, and due to their ability to undergo epithelial-mesenchymal transition, they modify the phenotype, bypassing the genotoxic effect of radiotherapy. It is therefore necessary to search for new methods that could improve the cytotoxic effect of cells through new mechanisms of action. Chinese medicine, with several thousand years of tradition, offers a wide range of possibilities in the search for compounds that could be used in conventional medicine. This review introduces the potential candidates that may present a radiosensitizing effect on lung cancer cells, breaking their radioresistance. Additionally, it includes candidates taken from conventional medicine-drugs commonly available in pharmacies, which may also be significant candidates.
Collapse
Affiliation(s)
- Paulina Nowak
- Scientific Club of Specialized Biological Analyzes, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Iwona Bil-Lula
- Department of Medical Laboratory Diagnostics, Division of Clinical Chemistry and Laboratory Hematology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Mariola Śliwińska-Mossoń
- Department of Medical Laboratory Diagnostics, Division of Clinical Chemistry and Laboratory Hematology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| |
Collapse
|
3
|
Ye Q, Zhou X, Han F, Zheng C. Toad venom-derived bufadienolides and their therapeutic application in prostate cancers: Current status and future directions. Front Chem 2023; 11:1137547. [PMID: 37007051 PMCID: PMC10060886 DOI: 10.3389/fchem.2023.1137547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/07/2023] [Indexed: 03/18/2023] Open
Abstract
Cancer is the second leading cause of death worldwide. Specially, the high incidence rate and prevalence of drug resistance have rendered prostate cancer (PCa) a great threat to men’s health. Novel modalities with different structures or mechanisms are in urgent need to overcome these two challenges. Traditional Chinese medicine toad venom-derived agents (TVAs) have shown to possess versatile bioactivities in treating certain diseases including PCa. In this work, we attempted to have an overview of bufadienolides, the major bioactive components in TVAs, in the treatment of PCa in the past decade, including their derivatives developed by medicinal chemists to antagonize certain drawbacks of bufadienolides such as innate toxic effect to normal cells. Generally, bufadienolides can effectively induce apoptosis and suppress PCa cells in-vitro and in-vivo, majorly mediated by regulating certain microRNAs/long non-coding RNAs, or by modulating key pro-survival and pro-metastasis players in PCa. Importantly, critical obstacles and challenges using TVAs will be discussed and possible solutions and future perspectives will also be presented in this review. Further in-depth studies are clearly needed to decipher the mechanisms, e.g., targets and pathways, toxic effects and fully reveal their application. The information collected in this work may help evoke more effects in developing bufadienolides as therapeutic agents in PCa.
Collapse
Affiliation(s)
- Qingmei Ye
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education, Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, Hainan, China
- Hainan General Hospital & Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Xin Zhou
- The Fifth People’s Hospital of Hainan Province & Affiliated Dermatology Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Fangxuan Han
- Hainan General Hospital & Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Caijuan Zheng
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education, Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, Hainan, China
- *Correspondence: Caijuan Zheng,
| |
Collapse
|
4
|
Gu R, Zhang Q. Effects of low-dose bufalin combined with hydroxycamptothecin on human castration-resistant prostate cancer xenografts in nude mice. Exp Ther Med 2021; 22:1015. [PMID: 34373701 PMCID: PMC8343571 DOI: 10.3892/etm.2021.10447] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 12/11/2019] [Indexed: 12/19/2022] Open
Abstract
Prostate cancer is the most prevalent tumor found in men worldwide. Despite the efficiency of primary endocrine prostate cancer therapies, more efficient drugs are needed to tackle the most advanced and resistant forms of this condition. The present study investigated the antitumor effects of low-dose bufalin combined with hydroxycamptothecin on castration-resistant prostate cancer (CRPC) in mice, as well as the possible mechanisms of apoptosis induction. CRPC xenograft tumors were generated in mice and, subsequently, mice received appropriate doses of bufalin, hydroxycamptothecin or a combination of the two drugs. Tumors from each treatment group were removed, and the tumor volume, weight and inhibition rate of each group was determined. Hematoxylin and eosin staining was performed for pathological analysis and TUNEL staining was used to assess the level of apoptosis in the xenografts. Immunohistochemistry was used for the analysis of proliferating cell nuclear antigen expression and the expression of Bax, Bcl-XL, p53, programmed cell death 4 (PDCD4), phosphorylated (p)-AKT and glycogen synthase kinase (GSK)-3β was determined by western blotting. Treatment with bufalin significantly (P<0.05) reduced tumor volumes compared with the negative control group, reducing tumor volumes to lower levels when combined with hydroxycampothecin. The combination of bufalin (0.6 or 0.8 mg/kg) and hydroxycampothecin significantly (P<0.05) induced higher levels of cell apoptosis compared with the administration of bufalin or hydroxycampothecin alone. The combination of bufalin and hydroxycampothecin also increased the expression of apoptosis-related proteins Bax, p53, PDCD4 and GSK-3β, and decreased the expression of Bcl-XL and p-AKT compared with a single drug treatment. The present study suggested that the combination of bufalin and hydroxycampothecin improved the inhibitory effects of both drugs on CRPC tumors in vivo, potentially via the regulation of the PI3K/AKT/GSK-3β and p53-dependent apoptosis signaling pathways.
Collapse
Affiliation(s)
- Renze Gu
- Department of Urology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Qingchuan Zhang
- Department of Urology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| |
Collapse
|
5
|
Fujii E, Inada Y, Kakoki M, Nishimura N, Endo S, Fujiwara S, Wada N, Kawano Y, Okuno Y, Sugimoto T, Hata H. Bufalin induces DNA damage response under hypoxic condition in myeloma cells. Oncol Lett 2018; 15:6443-6449. [PMID: 29616114 PMCID: PMC5876453 DOI: 10.3892/ol.2018.8091] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 02/13/2018] [Indexed: 12/13/2022] Open
Abstract
Hypoxia serves a crucial role in the development of drug resistance in various cancer cells. Therefore, many attempts targeting hypoxia are underway to overcome the drug resistance mediated by hypoxia. This strategy is useful for multiple myeloma (MM) cells, as MM cells reside within the bone marrow, where oxygen concentrations are relatively low. A natural compound library was screened to identify compounds exerting cytotoxicity in MM cells under hypoxic conditions. Bufalin exhibited marked cytotoxicity to MM cells under normoxic and hypoxic conditions. No significant toxicity was observed in lymphocytes obtained from healthy donors. Under normoxic conditions, bufalin induced a DNA double strand break (DSB) response, ROS induction and apoptosis within 24 with a rapid response compared with melphalan. Interestingly, the bufalin-induced DSB response was not impaired by low oxygen concentrations while the DSB response by melphalan was reduced. Furthermore, treatment with bufalin abolished HIF-1α expression under hypoxia, suggesting that bufalin exerts cytotoxicity under hypoxia by regulating HIF-1α. These results indicate that bufalin induces apoptosis in MM cells through DSB under hypoxic conditions by inhibiting HIF-1α, suggesting that bufalin could be useful for eradication of drug-resistant MM cells in the hypoxic microenvironment.
Collapse
Affiliation(s)
- Eri Fujii
- Graduate School of Health Sciences, Course of Medical Laboratory Sciences, Kumamoto University, Kumamoto 862-0976, Japan.,Department of Clinical Laboratory, Osaka University Hospital, Suita, Osaka 565-0871, Japan
| | - Yuki Inada
- Graduate School of Health Sciences, Course of Medical Laboratory Sciences, Kumamoto University, Kumamoto 862-0976, Japan
| | - Misaki Kakoki
- Graduate School of Health Sciences, Course of Medical Laboratory Sciences, Kumamoto University, Kumamoto 862-0976, Japan
| | - Nao Nishimura
- Department of Hematology, Faculty of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Shinya Endo
- Department of Hematology, Faculty of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Shiho Fujiwara
- Department of Hematology, Faculty of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Naoko Wada
- Department of Hematology, Faculty of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Yawara Kawano
- Department of Hematology, Faculty of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Yutaka Okuno
- Department of Hematology, Faculty of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Toshiya Sugimoto
- Graduate School of Health Sciences, Course of Medical Laboratory Sciences, Kumamoto University, Kumamoto 862-0976, Japan
| | - Hiroyuki Hata
- Department of Hematology, Faculty of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan.,Division of Informative Clinical Sciences, Faculty of Medical Sciences, Kumamoto University, Kumamoto 862-0976, Japan
| |
Collapse
|
6
|
Zhang X, Huang Q, Wang X, Xu Y, Xu R, Han M, Huang B, Chen A, Qiu C, Sun T, Wang F, Li X, Wang J, Zhao P, Wang X. Bufalin enhances radiosensitivity of glioblastoma by suppressing mitochondrial function and DNA damage repair. Biomed Pharmacother 2017; 94:627-635. [DOI: 10.1016/j.biopha.2017.07.136] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Revised: 07/21/2017] [Accepted: 07/25/2017] [Indexed: 10/19/2022] Open
|
7
|
Lanatoside C suppressed colorectal cancer cell growth by inducing mitochondrial dysfunction and increased radiation sensitivity by impairing DNA damage repair. Oncotarget 2017; 7:6074-87. [PMID: 26756216 PMCID: PMC4868741 DOI: 10.18632/oncotarget.6832] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 12/07/2015] [Indexed: 12/19/2022] Open
Abstract
Cardiac glycosides are clinically used for cardiac arrhythmias. In this study, we investigated the mechanism responsible for anti-cancer and radiosensitizing effects of lanatoside C in colorectal cancer cells. Lanatoside C-treated cells showed classic patterns of autophagy, which may have been caused by lanatoside C-induced mitochondrial aggregation or degeneration. This mitochondrial dysfunction was due to disruption of K+ homeostasis, possibly through inhibition of Na+/K+-ATPase activity. In addition, lanatoside C sensitized HCT116 cells (but not HT-29 cells) to radiation in vitro. γ-H2AX, a representative marker of DNA damage, were sustained longer after combination of irradiation with lanatoside C, suggesting lanatoside C impaired DNA damage repair processes. Recruitment of 53BP1 to damaged DNA, a critical initiation step for DNA damage repair signaling, was significantly suppressed in lanatoside C-treated HCT116 cells. This may have been due to defects in the RNF8- and RNF168-dependent degradation of KDM4A/JMJD2A that increases 53BP1 recruitment to DNA damage sites. Although lanatoside C alone reduced tumor growth in the mouse xenograft tumor model, combination of lanatoside C and radiation inhibited tumor growth more than single treatments. Thus, lanatoside C could be a potential molecule for anti-cancer drugs and radiosensitizing agents.
Collapse
|
8
|
Bufalin Inhibits Proliferation and Induces Apoptosis in Osteosarcoma Cells by Downregulating MicroRNA-221. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:7319464. [PMID: 28074104 PMCID: PMC5198087 DOI: 10.1155/2016/7319464] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 10/20/2016] [Accepted: 11/02/2016] [Indexed: 12/13/2022]
Abstract
Bufalin, a major component of the Chinese medicine ChanSu, which is prepared from the skin and parotid venom glands of toads, has shown cytotoxicity in several malignant tumors. Here, we reported that bufalin inhibited proliferation and induced mitochondria-dependent apoptosis in U-2OS and Saos-2 osteosarcoma cells with intracellular reactive oxygen species (ROS) production. By microRNA (miR) array analysis and quantitative reverse transcription polymerase chain reaction, we found that miR-221 was downregulated after treatment with bufalin. In accordance with TargetScan prediction and luciferase reporter assay, Bcl2 binding component 3 (BBC3) was the direct target of miR-221. Furthermore, upregulating miR-221 by its MIMIC and suppressing BBC3 by small interfering RNA (siRNA) reversed the effects of bufalin on osteosarcoma cells. Collectively, our data indicate that bufalin inhibits cell proliferation and induces mitochondria-dependent apoptosis in osteosarcoma cells through downregulating miR-221 and triggering BBC3 expression.
Collapse
|
9
|
Inhibition of topoisomerase IIα sensitizes FaDu cells to ionizing radiation by diminishing DNA repair. Tumour Biol 2015; 36:8985-92. [PMID: 26081617 DOI: 10.1007/s13277-015-3587-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 05/19/2015] [Indexed: 11/27/2022] Open
Abstract
Despite the high efficiency of ionizing radiation (IR) to inactivate malignant tumours in general, an appreciable number of individual patients cannot be cured by standard IR. Head and neck tumours are not likely to be cured even by high-dose radiotherapy or chemotherapy. Accordingly, combined therapy is one of the most applicable strategies. Topoisomerase IIα is a ubiquitous enzyme that removes knots and tangles from the genetic material by generating and subsequently resealing of transient double-strand breaks. Due to its unique mechanism of action, topoisomerase IIα is the target of many chemotherapeutic agents such as etoposide. The aim of the present study is to examine the effect of inhibiting topoisomerase IIα by etoposide on the response of squamous cell carcinoma to IR. Results of the present study demonstrated a radiosensitizing effect for the topoisomerase IIα inhibitor etoposide on exponentially growing squamous cell carcinoma (FaDu) cell line especially at low radiation doses. This effect was found to be due to inhibition, by etoposide, of the repair of radiation-induced DNA damage. Cell cycle studies showed that the concentration of etoposide that sensitized the cells to radiation had no effect on the distribution of cells at different phases of the cell cycle. Synchronization of FaDu cells in different cell cycle phases revealed that proliferating G1 and G2 cells are responsible for sensitization of cells at low doses of ionizing radiation. It might, therefore, be concluded that topoisomerase II enzyme may be involved in the repair of radiation-induced DNA damage and consequently its inhibition constitute a strategy for sensitizing tumour cells to ionizing radiation.
Collapse
|
10
|
Wu SH, Wu TY, Hsiao YT, Lin JH, Hsu SC, Hsia TC, Yang ST, Hsu WH, Chung JG. Bufalin induces cell death in human lung cancer cells through disruption of DNA damage response pathways. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2014; 42:729-42. [PMID: 24871662 DOI: 10.1142/s0192415x14500475] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Bufalin is a key component of a Chinese medicine (Chan Su) and has been proved effective in killing various cancer cells. Its role in inducing DNA damage and the inhibition of the DNA damage response (DDR) has been reported, but none have studied such action in lung cancer in detail. In this study, we demonstrated bufalin-induced DNA damage and condensation in NCI-H460 cells through a comet assay and DAPI staining, respectively. Western blotting indicated that bufalin suppressed the protein levels associated with DNA damage and repair, such as a DNA dependent serine/threonine protein kinase (DNA-PK), DNA repair proteins breast cancer 1, early onset (BRCA1), 14-3-3 σ (an important checkpoint keeper of DDR), mediator of DNA damage checkpoint 1 (MDC1), O6-methylguanine-DNA methyltransferase (MGMT) and p53 (tumor suppressor protein). Bufalin could activate phosphorylated p53 in NCI-H460 cells. DNA damage in NCI-H460 cells after treatment with bufalin up-regulated its ATM and ATR genes, which encode proteins functioning as sensors in DDR, and also up-regulated the gene expression (mRNA) of BRCA1 and DNA-PK. But bufalin suppressed the gene expression (mRNA) of p53 and 14-3-3 σ, however, bufalin did not significantly affect the mRNA of MGMT. In conclusion, bufalin induced DNA damage in NCI-H460 cells and also inhibited its DNA repair and checkpoint function.
Collapse
Affiliation(s)
- Shin-Hwar Wu
- Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan, ROC , Division of Critical Care Medicine, Department of Medicine, Changhua Christian Hospital, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Huang H, Cao Y, Wei W, Liu W, Lu SY, Chen YB, Wang Y, Yan H, Wu YL. Targeting poly (ADP-ribose) polymerase partially contributes to bufalin-induced cell death in multiple myeloma cells. PLoS One 2013; 8:e66130. [PMID: 23762475 PMCID: PMC3676346 DOI: 10.1371/journal.pone.0066130] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 05/02/2013] [Indexed: 11/18/2022] Open
Abstract
Despite recent pharmaceutical advancements in therapeutic drugs, multiple myeloma (MM) remains an incurable disease. Recently, ploy(ADP-ribose) polymerase 1 (PARP1) has been shown as a potentially promising target for MM therapy. A previous report suggested bufalin, a component of traditional Chinese medicine ("Chan Su"), might target PARP1. However, this hypothesis has not been verified. We here showed that bufalin could inhibit PARP1 activity in vitro and reduce DNA-damage-induced poly(ADP-ribosyl)ation in MM cells. Molecular docking analysis revealed that the active site of bufalin interaction is within the catalytic domain of PAPR1. Thus, PARP1 is a putative target of bufalin. Furthermore, we showed, for the first time that the proliferation of MM cell lines (NCI-H929, U266, RPMI8226 and MM.1S) and primary CD138(+) MM cells could be inhibited by bufalin, mainly via apoptosis and G2-M phase cell cycle arrest. MM cell apoptosis was confirmed by apoptotic cell morphology, Annexin-V positive cells, and the caspase3 activation. We further evaluated the role of PARP1 in bufalin-induced apoptosis, discovering that PARP1 overexpression partially suppressed bufalin-induced cell death. Moreover, bufalin can act as chemosensitizer to enhance the cell growth-inhibitory effects of topotecan, camptothecin, etoposide and vorinostat in MM cells. Collectively, our data suggest that bufalin is a novel PARP1 inhibitor and a potentially promising therapeutic agent against MM alone or in combination with other drugs.
Collapse
Affiliation(s)
- He Huang
- Department of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yang Cao
- Department of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Wei
- Department of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Liu
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Universities E-Institute for Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shao-Yong Lu
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Universities E-Institute for Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu-Bao Chen
- Department of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Wang
- Department of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hua Yan
- Department of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- * E-mail: (HY); (YLW)
| | - Ying-Li Wu
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Universities E-Institute for Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- * E-mail: (HY); (YLW)
| |
Collapse
|
12
|
Cosimi S, Orta L, Mateos S, Cortés F. The mycotoxin ochratoxin A inhibits DNA topoisomerase II and induces polyploidy in cultured CHO cells. Toxicol In Vitro 2009; 23:1110-5. [PMID: 19490938 DOI: 10.1016/j.tiv.2009.05.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2009] [Revised: 05/05/2009] [Accepted: 05/26/2009] [Indexed: 11/25/2022]
Abstract
Ochratoxin A (OTA), a known nephrotoxin and carcinogenic mycotoxin, was investigated to examine its effectiveness to induce cytotoxicity and DNA damage (Comet assay), as well as its possible inhibition of topoisomerase II (topo II) catalytic activity in cultured Chinese hamster ovary (CHO) cells. The analysis of OTA-induced DNA strand breaks as well as the flow cytometric assessment of polyploidy has provided evidence that is consistent with the idea of a mixed mode of action of the mycotoxin: in addition to its genotoxic activity, OTA may also interfere with chromosome distribution during cell division.
Collapse
Affiliation(s)
- Simona Cosimi
- Department of Cell Biology, Faculty of Biology, University of Seville, Av. Reina Mercedes 6, E-41012 Seville, Spain
| | | | | | | |
Collapse
|
13
|
Abstract
Amphibians have been found to be a source of agents with anticancer properties. Bufalin, for example, is an anticancer agent that may induce apoptosis by its interaction with other genes and cellular components. Certain peptides with anticancer activities have been found in amphibian skin; they include magainins, aureins, citropin 1.1 and gaegurins. These peptides may exert a cytotoxic effect on human cancer cells through various mechanisms. Onconase, amphinase, cSBL (sialic acid-binding lectin purified from Rana catesbeiana eggs) and jSBL (sialic acid-binding lectin purified from Rana japonica eggs), which belong to the RNase A family, were purified from the oocyte cells and eggs of three amphibians, and they induce cytotoxicity by degrading cellular RNA. This paper discusses the medical and pharmaceutical significance of products derived from amphibians.
Collapse
|
14
|
Yu CH, Kan SF, Pu HF, Jea Chien E, Wang PS. Apoptotic signaling in bufalin- and cinobufagin-treated androgen-dependent and -independent human prostate cancer cells. Cancer Sci 2008; 99:2467-76. [PMID: 19037992 PMCID: PMC11159935 DOI: 10.1111/j.1349-7006.2008.00966.x] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2008] [Revised: 08/03/2008] [Accepted: 08/09/2008] [Indexed: 01/08/2023] Open
Abstract
Prostate cancer has its highest incidence in the USA and is becoming a major concern in Asian countries. Bufadienolides are extracts of toxic glands from toads and are used as anticancer agents, mainly on leukemia cells. In the present study, the antiproliferative and apoptotic mechanisms of bufalin and cinobufagin on prostate cancer cells were investigated. Proliferation of LNCaP, DU145, and PC3 cells was measured by 3-(4,5-dimethylthiazol-2-yle)-2,5-diphenyltetrazolium bromide assay and the doubling time (tD) was calculated. Bufalin and cinobufagin caused changes in the tD of three prostate cancer cell lines, which were more significant than that of human mesangial cells. In addition, bufadienolides induced prostate cancer cell apoptosis more significantly than that in breast epithelial cell lines. After treatment, the caspase-3 activity and protein expression of caspase-3, -8, and -9 were elevated. The expression of other apoptotic modulators, including mitochondrial Bax and cytosolic cytochrome c, were also increased. However, expression of p53 was only enhanced in LNCaP cells. Downregulation of p53 by antisense TP53 restored the cell viability suppressed by bufalienolides. Furthermore, the increased expression of Fas was more significant in DU145 and PC3 cells with mutant p53 than in LNCaP cells. Transfection of Fas small interfering RNA restored cell viability in the bufadienolide-treated cells. These results suggest that bufalin and cinobufagin suppress cell proliferation and cause apoptosis in prostate cancer cells via a sequence of apoptotic modulators, including Bax, cytochrome c, and caspases. The upstream mediators might be p53 and Fas in androgen-dependent LNCaP cells and Fas in androgen-independent DU145 and PC3 cells.
Collapse
Affiliation(s)
- Ching-Han Yu
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan
| | | | | | | | | |
Collapse
|
15
|
Mateos S, Domínguez I, Cantero G, Pastor N, Campanella C, Cortés F. The high rate of endoreduplication in the repair deficient CHO mutant EM9 parallels a reduced level of methylated deoxycytidine in DNA. Mutat Res 2008; 644:24-30. [PMID: 18640132 DOI: 10.1016/j.mrfmmm.2008.06.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2007] [Revised: 05/30/2008] [Accepted: 06/20/2008] [Indexed: 11/26/2022]
Abstract
It has been recently proposed that hypomethylation of DNA induced by 5-azacytidine (5-azaC) leads to reduced chromatid decatenation that ends up in endoreduplication, most likely due to a failure in topo II function [S. Mateos, I. Domínguez, N. Pastor, G. Cantero, F. Cortés, The DNA demethylating 5-azaC induces endoreduplication in cultured Chinese hamster cells, Mutat. Res. 578 (2005) 33-42]. The Chinese hamster mutant cell line EM9 has a high spontaneous frequency of endoreduplication as compared to its parental line AA8. In order to see if this is related to the degree of DNA methylation, we have investigated the basal levels of both endpoints in AA8 and EM9, as well as the effect of extensive 5-azaC-induced demethylation on the production of endoreduplication. Based on the correlation between the levels of DNA methylation and indices of endoreduplication we propose that genomic DNA hypomethylation in EM9 cell line is probably an important factor that bears significance in relation to the high basal level of endoreduplication observed in this cell line.
Collapse
Affiliation(s)
- Santiago Mateos
- Department of Cell Biology, Faculty of Biology, University of Seville, Avda Reina Mercedes 6, 41012 Seville, Spain.
| | | | | | | | | | | |
Collapse
|
16
|
Dodson AW, Taylor TJ, Knipe DM, Coen DM. Inhibitors of the sodium potassium ATPase that impair herpes simplex virus replication identified via a chemical screening approach. Virology 2007; 366:340-8. [PMID: 17544048 PMCID: PMC2099250 DOI: 10.1016/j.virol.2007.05.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2007] [Revised: 03/27/2007] [Accepted: 05/01/2007] [Indexed: 01/07/2023]
Abstract
Small molecules can provide valuable tools to investigate virus biology. We developed a chemical screening approach to identify small molecule inhibitors of poorly understood, pre-early gene expression steps in herpes simplex virus infection, using green fluorescent protein fused to an early protein. Our assay identified ouabain, a cardiac glycoside. Ouabain reversibly decreased viral yield by 100-fold without affecting cellular metabolic activity in an overnight assay. The antiviral potencies of other cardiac glycosides correlated with their potencies against the known target of these compounds, the cellular sodium potassium ATPase. Ouabain had a reduced effect if added 8 h post-infection. It did not inhibit viral attachment or entry, but did reduce the expression of viral immediate-early and early genes by at least 5-fold. Collectively, these results implicate a cellular target that was hitherto not considered important for a stage of HSV replication prior to viral gene expression.
Collapse
Affiliation(s)
- Allen W. Dodson
- Department of Biological Chemistry and Molecular Pharmacology
| | - Travis J Taylor
- Department of Microbiology and Molecular Genetics
- Committee on Virology, Harvard Medical School Boston, MA 02115, USA
| | - David M. Knipe
- Department of Microbiology and Molecular Genetics
- Committee on Virology, Harvard Medical School Boston, MA 02115, USA
| | - Donald M. Coen
- Department of Biological Chemistry and Molecular Pharmacology
- Committee on Virology, Harvard Medical School Boston, MA 02115, USA
- To Whom Correspondence should be addressed: Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, 250 Longwood Avenue, Boston, MA, 02215. Phone: +1 617 432-1691. Fax: +1 617 432-3833,
| |
Collapse
|
17
|
Cortés F, Pastor N, Mateos S, Domínguez I. Topoisomerase inhibitors as therapeutic weapons. Expert Opin Ther Pat 2007. [DOI: 10.1517/13543776.17.5.521] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
18
|
Mateos S, Hajji N, Pastor N, Cortés F. Modulation of radiation response by inhibiting topoisomerase II catalytic activity. Mutat Res 2006; 599:105-15. [PMID: 16574164 DOI: 10.1016/j.mrfmmm.2006.02.002] [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] [Received: 07/28/2005] [Revised: 02/09/2006] [Accepted: 02/14/2006] [Indexed: 05/08/2023]
Abstract
Due to the essential role played by DNA topoisomerases (topos) in cell survival, the use of topoisomerase inhibitors as chemotherapeutic drugs in combination with radiation has become a common strategy for the treatment of cancer. Catalytic inhibitors of these enzymes would be promising to improve the effectiveness of radiation and therefore, it appears reasonable to incorporate them in combined modality trials. In this work, we have investigated the capacity of both ICRF-193 and Aclarubicin (ACLA), two catalytic inhibitors of topoisomerase II (Topo II), to modulate radiation response in Chinese hamster V79 cell line and its radiosensitive mutant irs2. We also have explored potential mechanisms underlying these interactions. Experiments were performed in the presence and absence of either ICRF-193 or ACLA, and topo II activity was measured using an assay based upon decatenation of kinetoplast DNA (kDNA). For the combined experiments cells were incubated for 3 h in the presence of various inhibitor concentrations and irradiated 30 min prior to the end of treatments and cell survival was determined by clonogenic assay. DNA-damaging activity was measured by single-cell gel electrophoresis. Our results demonstrate that combinations of catalytic inhibitors of topo II and radiation produce an increase in cell killing induced by ionising radiation. The mechanism of radiation enhancement may involve a direct or indirect participation of topo II in the repair of radiation-induced DNA damage.
Collapse
Affiliation(s)
- Santiago Mateos
- Department of Cell Biology, Cell Culture and Radiobiology Research Group, University of Seville, Avda. Reina Mercedes n degree 6, 41012 Seville, Spain.
| | | | | | | |
Collapse
|
19
|
Hofland KF, Thougaard AV, Dejligbjerg M, Jensen LH, Kristjansen PEG, Rengtved P, Sehested M, Jensen PB. Combining Etoposide and Dexrazoxane Synergizes with Radiotherapy and Improves Survival in Mice with Central Nervous System Tumors. Clin Cancer Res 2005; 11:6722-9. [PMID: 16166453 DOI: 10.1158/1078-0432.ccr-05-0698] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The treatment of patients with brain metastases is presently ineffective, but cerebral chemoradiotherapy using radiosensitizing agents seems promising. Etoposide targets topoisomerase II, resulting in lethal DNA breaks; such lesions may increase the effect of irradiation, which also depends on DNA damage. Coadministration of the topoisomerase II catalytic inhibitor dexrazoxane in mice allows for more than 3-fold higher dosing of etoposide. We hypothesized that dexrazoxane combined with escalated etoposide doses might improve the efficacy of cerebral radiotherapy. EXPERIMENTAL DESIGN Mice with cerebrally inoculated Ehrlich ascites tumor (EHR2) cells were treated with combinations of etoposide + dexrazoxane + cerebral radiotherapy. Similar chemotherapy and radiation combinations were investigated by clonogenic assays using EHR2 cells, and by DNA double-strand break assay through quantification of phosphorylated histone H2AX (gammaH2AX). RESULTS Escalated etoposide dosing (90 mg/kg) combined with dexrazoxane (125 mg/kg) and cerebral radiotherapy (10 Gy x 1) increased the median survival by 60% (P = 0.001) without increased toxicity, suggesting that escalated etoposide levels may indeed represent a new strategy for improving radiotherapy. Interestingly, 125 mg/kg dexrazoxane combined with normal etoposide doses (34 mg/kg) also increased survival from radiotherapy, but only by 27% (P = 0.002). This indicates a direct dexrazoxane modulation of the combined effects of etoposide and radiation in brain tumors. Further, in vitro, concurrent dexrazoxane, etoposide, and irradiation significantly increased DNA double-strand breaks. CONCLUSION Combining etoposide (high or normal doses) and dexrazoxane synergizes with cerebral radiotherapy and significantly improves survival in mice with central nervous system tumors. This regimen may thus improve radiation therapy of central nervous system tumors.
Collapse
MESH Headings
- Animals
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Blood-Brain Barrier/drug effects
- Blood-Brain Barrier/pathology
- Blood-Brain Barrier/radiation effects
- Central Nervous System Neoplasms/drug therapy
- Central Nervous System Neoplasms/pathology
- Central Nervous System Neoplasms/radiotherapy
- Combined Modality Therapy
- DNA Damage
- DNA, Neoplasm/drug effects
- DNA, Neoplasm/genetics
- DNA, Neoplasm/radiation effects
- Dose-Response Relationship, Drug
- Dose-Response Relationship, Radiation
- Etoposide/administration & dosage
- Female
- Mice
- Mice, Inbred Strains
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/pathology
- Neoplasms, Experimental/radiotherapy
- Razoxane/administration & dosage
- Survival Analysis
- Time Factors
- Treatment Outcome
- Tumor Cells, Cultured
Collapse
|