Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 361
Filter
Add more filters

Publication year range
1.
Arterioscler Thromb Vasc Biol ; 38(10): 2382-2395, 2018 10.
Article in English | MEDLINE | ID: mdl-30354214

ABSTRACT

Objective- Vascular calcification is a major risk factor for rupture of atherosclerotic plaques. High expression of BMP2 (bone morphogenetic protein 2) in lesions suggests its importance in vascular calcification during atherosclerosis. Teniposide is a Topo II (DNA topoisomerase II) inhibitor and is used for cancer treatment. Previously, we reported that teniposide activated macrophage ABCA1 (ATP-binding cassette transporter A1) expression and free cholesterol efflux indicating Topo II inhibitors may demonstrate antiatherogenic properties. Herein, we investigated the effects of teniposide on the development of atherosclerosis and vascular calcification in apoE-/- (apoE deficient) mice. Approach and Results- apoE-/- mice were fed high-fat diet containing teniposide for 16 weeks, or prefed high-fat diet for 12 weeks followed by high-fat diet containing teniposide for 4 weeks. Atherosclerosis and vascular calcification were determined. Human aortic smooth muscle cells were used to determine the mechanisms for teniposide-inhibited vascular calcification. Teniposide reduced atherosclerotic lesions. It also substantially reduced vascular calcification without affecting bone structure. Mechanistically, teniposide reduced vascular calcification by inactivating BMP2/(pi-Smad1/5/8 [mothers against decapentaplegic homolog 1, 5, and 8])/RUNX2 (runt-related transcription factor 2) axis in a p53-dependent manner. Furthermore, activated miR-203-3p by teniposide functioned as a link between activated p53 expression and inhibited BMP2 expression in inhibition of calcification. Conclusions- Our study demonstrates that teniposide reduces vascular calcification by regulating p53-(miR-203-3p)-BMP2 signaling pathway, which contributes to the antiatherogenic properties of Topo II inhibitors.


Subject(s)
Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Teniposide/pharmacology , Topoisomerase II Inhibitors/pharmacology , Vascular Calcification/prevention & control , 3' Untranslated Regions , Alkaline Phosphatase/metabolism , Animals , Aorta/drug effects , Aorta/enzymology , Aorta/pathology , Aortic Diseases/enzymology , Aortic Diseases/genetics , Aortic Diseases/pathology , Atherosclerosis/enzymology , Atherosclerosis/genetics , Atherosclerosis/pathology , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/metabolism , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Disease Models, Animal , Female , Humans , Mice, Knockout, ApoE , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/pathology , Signal Transduction/drug effects , Smad Proteins/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Vascular Calcification/enzymology , Vascular Calcification/genetics , Vascular Calcification/pathology
2.
Biochem Biophys Res Commun ; 506(4): 1040-1046, 2018 12 02.
Article in English | MEDLINE | ID: mdl-30409428

ABSTRACT

The switch of vascular smooth muscle cells (SMCs) from the contractile phenotype to proliferative one can make contributions to atherosclerosis and neointima formation. MiR-21 can prevent the rupture of advanced lesion plaques. We previously reported the protection of DNA topoisomerase II (Topo II) inhibitors against atherosclerosis and vascular calcification. However, it remains unknown if Topo II inhibitors can change SMC phenotypes. Herein, we show that teniposide protected SMC phenotype switching during atherosclerosis by enhancing expression of smooth muscle α-actin (SMA) while reducing osteopontin (OPN) expression in aortic lesion plaques. In vitro, teniposide induced expression of smooth muscle protein 22-α and calponin 1, but inhibited expression of OPN and epiregulin in human aortic SMCs (HASMCs). Moreover, teniposide attenuated platelet derived growth factor-BB-induced HASMC proliferation and migration. Mechanistically, the effect of teniposide on SMC phenotypes was completed, at least in part, by activating miR-21 expression. In addition, teniposide ameliorated ligation-induced carotid artery remodeling in C57BL/6J mice by regulating SMA and OPN expression. Taken together, our study demonstrates that teniposide regulates SMC phenotype switching by upregulating expression of contractile genes in a miR-21-dependent manner, and this function is an important anti-atherogenic mechanism of teniposide.


Subject(s)
MicroRNAs/metabolism , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Teniposide/pharmacology , Animals , Cell Proliferation/drug effects , Female , Humans , Male , Mice, Inbred C57BL , MicroRNAs/genetics , Muscle Contraction/drug effects , Myocytes, Smooth Muscle/drug effects , Phenotype , Proto-Oncogene Proteins c-sis/pharmacology , Vascular Remodeling/drug effects
3.
Inflammopharmacology ; 26(2): 395-402, 2018 Apr.
Article in English | MEDLINE | ID: mdl-28825192

ABSTRACT

Bone pain associated with advanced tumor metastasis is the most severe threat to life quality of patients. Highly efficient and low-toxic therapeutics is of urgent need for this complication. Bone tumor metastasis was established by direct bone inoculation of Walker 256 mammary gland carcinoma cells. Bone nociception was measured by mechanical allodynia, thermal hyperalgesia and spontaneous flinches. P2X7R level was determined by immunoblotting. The inward current was recorded by a patch clamp. The related cytokines were determined by ELISA. Our results showed that teniposide (TN) treatment significantly ameliorated bone nociception associated with tumor inoculation to a comparable extent with P2X7-specific inhibitor, BBG, in rat model. The efficient blockade of inward current generation and pro-inflammatory cytokines secretion were observed upon administration with TN. Our data highlighted the therapeutic potency of TN in this complication associated with tumor metastasis and warrants further clinical investigations.


Subject(s)
Bone Neoplasms/complications , Nociception/drug effects , Pain/drug therapy , Receptors, Purinergic P2X7/metabolism , Teniposide/pharmacology , Animals , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Carcinoma/pathology , Cell Line, Tumor , Cytokines/metabolism , Disease Models, Animal , Female , Hyperalgesia/drug therapy , Hyperalgesia/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Mammary Glands, Animal/pathology , Pain/etiology , Pain/metabolism , Pain Measurement/methods , Pain Threshold/drug effects , Rats , Rats, Wistar
4.
J Biol Chem ; 290(23): 14418-29, 2015 Jun 05.
Article in English | MEDLINE | ID: mdl-25914138

ABSTRACT

Cholesteryl ester transfer protein (CETP) transfers cholesteryl esters from high density lipoprotein to triglyceride-rich lipoproteins. CETP expression can be transcriptionally activated by liver X receptor (LXR). Etoposide and teniposide are DNA topoisomerase II (Topo II) inhibitors. Etoposide has been reported to inhibit atherosclerosis in rabbits with un-fully elucidated mechanisms. In this study we determined if Topo II activity can influence cholesterol metabolism by regulating hepatic CETP expression. Inhibition of Topo II by etoposide, teniposide, or Topo II siRNA increased CETP expression in human hepatic cell line, HepG2 cells, which was associated with increased CETP secretion and mRNA expression. Meanwhile, inhibition of LXR expression by LXR siRNA attenuated induction of CETP expression by etoposide and teniposide. Etoposide and teniposide induced LXRα expression and LXRα/ß nuclear translocation while inhibiting expression of receptor interacting protein 140 (RIP140), an LXR co-repressor. In vivo, administration of teniposide moderately reduced serum lipid profiles, induced CETP expression in the liver, and activated reverse cholesterol transport in CETP transgenic mice. Our study demonstrates a novel function of Topo II inhibitors in cholesterol metabolism by activating hepatic CETP expression and reverse cholesterol transport.


Subject(s)
Cholesterol Ester Transfer Proteins/genetics , Cholesterol/metabolism , DNA Topoisomerases, Type II/metabolism , Etoposide/pharmacology , Gene Expression Regulation/drug effects , Teniposide/pharmacology , Topoisomerase II Inhibitors/pharmacology , Animals , Cholesterol Ester Transfer Proteins/metabolism , Hep G2 Cells , Humans , Liver/drug effects , Liver/metabolism , Liver X Receptors , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Orphan Nuclear Receptors/genetics , Orphan Nuclear Receptors/metabolism , Protein Transport/drug effects , Transcriptional Activation/drug effects
5.
Biochim Biophys Acta ; 1831(6): 1134-45, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23466610

ABSTRACT

ATP-binding cassette transporter A1 (ABCA1) facilitates cholesterol efflux and thereby inhibits lipid-laden macrophage/foam cell formation and atherosclerosis. ABCA1 expression is transcriptionally regulated by activation of liver X receptor (LXR). Both etoposide and teniposide are DNA topoisomerase II (Topo II) inhibitors and are chemotherapeutic medications used in the treatment of various cancers. Interestingly, etoposide inhibits atherosclerosis in rabbits by unclear mechanisms. Herein, we report the effects of etoposide and teniposide on macrophage ABCA1 expression and cholesterol efflux. Both etoposide and teniposide increased macrophage free cholesterol efflux. This increase was associated with increased ABCA1 mRNA and protein expression. Etoposide and teniposide also increased ABCA1 promoter activity in an LXR-dependent manner and formation of the LXRE-LXR/RXR complex indicating that transcriptional induction had occurred. Expression of ABCG1 and fatty acid synthase (FAS), another two LXR-targeted genes, was also induced by etoposide and teniposide. In vivo, administration of mice with either etoposide or teniposide induced macrophage ABCA1 expression and enhanced reverse cholesterol transport from macrophages to feces. Taken together, our study indicates that etoposide and teniposide increase macrophage ABCA1 expression and cholesterol efflux that may be attributed to the anti-atherogenic properties of etoposide. Our study also describes a new function for Topo II inhibitors in addition to their role in anti-tumorigenesis.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Cholesterol/metabolism , Foam Cells/drug effects , Gene Expression Regulation/drug effects , Macrophages/drug effects , Orphan Nuclear Receptors/metabolism , Topoisomerase II Inhibitors/pharmacology , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/metabolism , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Cells, Cultured , Electrophoretic Mobility Shift Assay , Etoposide/pharmacology , Fluorescent Antibody Technique , Foam Cells/cytology , Foam Cells/metabolism , Liver X Receptors , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Orphan Nuclear Receptors/antagonists & inhibitors , Orphan Nuclear Receptors/genetics , Promoter Regions, Genetic/genetics , RNA, Small Interfering/genetics , Retinoid X Receptors/genetics , Retinoid X Receptors/metabolism , Teniposide/pharmacology
6.
BMC Cancer ; 14: 611, 2014 Aug 25.
Article in English | MEDLINE | ID: mdl-25151861

ABSTRACT

BACKGROUND: Although the incidence of glioma is relatively low, it is the most malignant tumor of the central nervous system. The prognosis of high-grade glioma patient is very poor due to the difficulties in complete resection and resistance to radio-/chemotherapy. Therefore, it is worth investigating the molecular mechanisms involved in glioma drug resistance. MicroRNAs have been found to play important roles in tumor progression and drug resistance. Our previous work showed that miR-181b is involved in the regulation of temozolomide resistance. In the current study, we investigated whether miR-181b also plays a role in antagonizing the effect of teniposide. METHODS: MiR-181b expression was measured in 90 glioma patient tissues and its relationship to prognosis of these patients was analyzed. Cell sensitivity to teniposide was tested in 48 primary cultured glioma samples. Then miR-181b stably overexpressed U87 cells were generated. The candidate genes of miR-181b from our previous study were reanalyzed, and the interaction between miR-181b and target gene MDM2 was confirmed by dual luciferase assay. Cell sensitivity to teniposide was detected on miR-181b over expressed and MDM2 down regulated cells. RESULTS: Our data confirmed the low expression levels of miR-181b in high-grade glioma tissues, which is related to teniposide resistance in primary cultured glioma cells. Overexpression of miR-181b increased glioma cell sensitivity to teniposide. Through target gene prediction, we found that MDM2 is a candidate target of miR-181b. MDM2 knockdown mimicked the sensitization effect of miR-181b. Further study revealed that miR-181b binds to the 3'-UTR region of MDM2 leading to the decrease in MDM2 levels and subsequent increase in teniposide sensitivity. Partial restoration of MDM2 attenuated the sensitivity enhancement by miR-181b. CONCLUSIONS: MiR-181b is an important positive regulator on glioma cell sensitivity to teniposide. It confers glioma cell sensitivity to teniposide through binding to the 3'-UTR region of MDM2 leading to its reduced expression. Our findings not only reveal the novel mechanism involved in teniposide resistance, but also shed light on the optimization of glioma treatment in the future.


Subject(s)
Biomarkers, Tumor/metabolism , Glioma/drug therapy , MicroRNAs/metabolism , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Teniposide/pharmacology , Biomarkers, Tumor/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic/drug effects , Glioma/genetics , Glioma/pathology , Humans , MicroRNAs/genetics , Prognosis , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Cells, Cultured
7.
Cell Death Dis ; 15(5): 322, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38719798

ABSTRACT

Metastatic dissemination from the primary tumor is a complex process that requires crosstalk between tumor cells and the surrounding milieu and involves the interplay between numerous cellular-signaling programs. Epithelial-mesenchymal transition (EMT) remains at the forefront of orchestrating a shift in numerous cellular programs, such as stemness, drug resistance, and apoptosis that allow for successful metastasis. Till date, there is limited success in therapeutically targeting EMT. Utilizing a high throughput screen of FDA-approved compounds, we uncovered a novel role of the topoisomerase inhibitor, Teniposide, in reversing EMT. Here, we demonstrate Teniposide as a potent modulator of the EMT program, specifically through an IRF7-NMI mediated response. Furthermore, Teniposide significantly reduces the expression of the key EMT transcriptional regulator, Zinc Finger E-Box Binding Homeobox 2 (ZEB2). ZEB2 downregulation by Teniposide inhibited RNA polymerase I (Pol I) activity and rRNA biogenesis. Importantly, Teniposide treatment markedly reduced pulmonary colonization of breast cancer cells. We have uncovered a novel role of Teniposide, which when used at a very low concentration, mitigates mesenchymal-like invasive phenotype. Overall, its ability to target EMT and rRNA biogenesis makes Teniposide a viable candidate to be repurposed as a therapeutic option to restrict breast cancer metastases.


Subject(s)
Breast Neoplasms , Down-Regulation , Epithelial-Mesenchymal Transition , RNA Polymerase I , Teniposide , Zinc Finger E-box Binding Homeobox 2 , Epithelial-Mesenchymal Transition/drug effects , Humans , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/genetics , Female , Zinc Finger E-box Binding Homeobox 2/metabolism , Zinc Finger E-box Binding Homeobox 2/genetics , Cell Line, Tumor , Down-Regulation/drug effects , RNA Polymerase I/metabolism , Teniposide/pharmacology , Animals , Mice , Gene Expression Regulation, Neoplastic/drug effects
8.
Mutagenesis ; 27(1): 31-9, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21821840

ABSTRACT

The ability of topoisomerase II inhibitor, teniposide, to induce aneuploidy and meiotic delay in somatic and germinal cells of male mice was investigated by fluorescence in situ hybridisation (FISH) assay using labelled DNA probes and 5-bromo-2'-deoxyuridine (BrdU) incorporation assay, respectively. Colchicine and mitomycin C were used as a positive control aneugen and clastogen, respectively, and these compounds produced the expected responses. Using FISH assay with centromeric and telomeric DNA probes for erythrocyte, micronuclei (MN) showed that teniposide is not only clastogenic but also aneugenic in somatic cells in vivo. The assay also showed that chromosomes can be enclosed in the MN before and after centromere separation. By using the BrdU incorporation assay, it could be shown that the meiotic delay caused by teniposide in germ cells was ∼48 h. Disomic and diploid sperms were shown in epididymal sperm hybridised with DNA probes specific for chromosomes 8, X and Y after teniposide treatment. The prevalence of autodiploid (XX88 and YY88) sperm and disomic XX8 or YY8 sperm indicates that the second meiotic division was more sensitive to teniposide than the first meiotic division. The results also suggest that earlier prophase stages contribute relatively less to teniposide-induced aneuploidy. Both the clastogenic and the aneugenic potential of teniposide can give rise to the development of secondary tumours and abnormal reproductive outcomes in cured cancer patients and medical personnel exposing to drug regimens that include teniposide. Thus, genetic counselling of these patients should take place before the start of chemotherapy and should take the present results into consideration.


Subject(s)
Aneugens/pharmacology , Cytogenetic Analysis , Spermatozoa/drug effects , Teniposide/pharmacology , Animals , Bromodeoxyuridine/metabolism , Colchicine/toxicity , DNA Probes/genetics , Erythrocytes/drug effects , In Situ Hybridization, Fluorescence/methods , Male , Mice , Micronucleus Tests , Mitomycin/toxicity , Mitosis/drug effects
9.
J Exp Med ; 175(1): 23-7, 1992 Jan 01.
Article in English | MEDLINE | ID: mdl-1309853

ABSTRACT

Cytotoxic T lymphocytes (CTL) kill their target cells via a contact-dependent mechanism that results in the perturbation of the target cell's plasma membrane and the fragmentation of the target cell's DNA into nucleosomal particles. The membrane disruption is presumed to be due to the action of perforin, while the DNA fragmentation is thought to be by the activation of an endogenous nuclease(s). DNA topoisomerases I and II are nuclear enzymes with inherent endonuclease activities. We have investigated their role in the CTL-induced DNA fragmentation process. We report that in CTL killing assays, the treatment of target cells with topoisomerase I and II inhibitors blocks the CTL-induced DNA fragmentation process, but not the lysis of the target cell.


Subject(s)
Amsacrine/pharmacology , Camptothecin/pharmacology , DNA Damage , DNA/genetics , T-Lymphocytes, Cytotoxic/immunology , Teniposide/pharmacology , Topoisomerase I Inhibitors , Topoisomerase II Inhibitors , Animals , Cells, Cultured , Cytotoxicity, Immunologic , DNA/drug effects , DNA/isolation & purification , Female , Killer Cells, Natural/immunology , Lymphocyte Depletion , Lymphocytic choriomeningitis virus/immunology , Male , Mice , Mice, Inbred Strains , T-Lymphocytes, Cytotoxic/drug effects
10.
Cell Mol Neurobiol ; 30(6): 955-60, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20526804

ABSTRACT

Brain tumor stem-like cells (BTSLCs) have been implied to play an important role in genesis and development of glioma. However, their characteristics on proliferation and drug-resistance are uncertain thoroughly. In this experiment, some of the biological characteristics about BTSLCs were explored. Twenty cases of different grades of human glioma tissues were obtained from clinic. The primary glioma cells were collected and CD133(+) cells from them were purified by magnetic cell sorting assay. The BTSLCs were identified by testing the expression of CD133, Nestin, NSE, and GFAP, along with the culture process. WST-8 assay kit was used to evaluate the proliferating situation of CD133(+) cells in the different grade gliomas, and to compare the drug-resistance between the CD133(+) and CD133( - ) cells in the medium containing different concentrations of teniposide (VM-26). The results showed that the CD133(+) cells could regenerate by self-renewal, then generate and different into NSE(+) and GFAP(+) cells, respectively. CD133(+) cells in the high grade of gliomas showed the faster generation than the ones in the low grade. The number of survived CD133(+) cells in the medium containing VM-26 was much more than the CD133(-) ones in it. Therefore, it was implied that the CD133(+) BTSLCs existed in the glioma tissues possessed the more tolerant ability to the VM-26, and could proliferate much more easily in the high-grade glioma.


Subject(s)
Brain Neoplasms/pathology , Drug Resistance, Neoplasm , Glioma/pathology , Neoplastic Stem Cells/pathology , AC133 Antigen , Antigens, CD/metabolism , Brain Neoplasms/metabolism , Cell Death/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Flow Cytometry , Glioma/metabolism , Glycoproteins/metabolism , Humans , Neoplastic Stem Cells/metabolism , Peptides/metabolism , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Teniposide/pharmacology , Tumor Cells, Cultured
11.
Nucleic Acids Res ; 36(19): 6165-74, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18824478

ABSTRACT

Although centromere function has been conserved through evolution, apparently no interspecies consensus DNA sequence exists. Instead, centromere DNA may be interconnected through the formation of certain DNA structures creating topological binding sites for centromeric proteins. DNA topoisomerase II is a protein, which is located at centromeres, and enzymatic topoisomerase II activity correlates with centromere activity in human cells. It is therefore possible that topoisomerase II recognizes and interacts with the alpha satellite DNA of human centromeres through an interaction with potential DNA structures formed solely at active centromeres. In the present study, human topoisomerase IIalpha-mediated cleavage at centromeric DNA sequences was examined in vitro. The investigation has revealed that the enzyme recognizes and cleaves a specific hairpin structure formed by alpha satellite DNA. The topoisomerase introduces a single-stranded break at the hairpin loop in a reaction, where DNA ligation is partly uncoupled from the cleavage reaction. A mutational analysis has revealed, which features of the hairpin are required for topoisomerease IIalpha-mediated cleavage. Based on this a model is discussed, where topoisomerase II interacts with two hairpins as a mediator of centromere cohesion.


Subject(s)
Antigens, Neoplasm/metabolism , Centromere/chemistry , DNA Topoisomerases, Type II/metabolism , DNA, Satellite/chemistry , DNA-Binding Proteins/metabolism , Base Sequence , DNA, Satellite/metabolism , DNA-Binding Proteins/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Humans , Models, Biological , Molecular Sequence Data , Nucleic Acid Conformation , Teniposide/pharmacology , Topoisomerase II Inhibitors
12.
J Med Chem ; 63(6): 2877-2893, 2020 03 26.
Article in English | MEDLINE | ID: mdl-32084316

ABSTRACT

As an FDA-approved drug, teniposide, was utilized in cancer treatment but was accompanied by a strong side effect in long-term clinical trials. This work discovered potential candidate drugs with low toxicity by modifying the molecule structure of teniposide through a structure-guided drug design approach. The IC50 value of novel 4,6-O-thenylidene-ß-d-glucopyranoside-(2″-acetamido, 3″-acetyl-di-S-5-fluorobenzothizole/5-fluorobenzoxazole)-4'-demethylepipodophyllotoxin (compounds 15 and 16) was 120.4-125.1 µM, which was significantly improved by around 10 times more than teniposide (11.5-22.3 µM) against healthy human cells (i.e., HL-7702, H8, MRC-5, and HMEC). In vivo studies demonstrated compounds 15 and 16 significantly suppressed the tumor growth in the HepG2 cell xenograft model without exhibiting obvious toxicity (LD50 values of 208.45 and 167.52 mg/kg), which was lower than that of teniposide (LD50 = 46.12 mg/kg). Compounds 15 and 16 caused mild γH2AX phosphorylation for low DNA toxicity and less inhibition of PI3K/Akt. Compounds 15 and 16 might be potential antitumor drugs with low toxicity.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Podophyllotoxin/analogs & derivatives , Teniposide/analogs & derivatives , Teniposide/pharmacology , Animals , Antineoplastic Agents/toxicity , Cell Line , DNA Damage/drug effects , Hep G2 Cells , Humans , Male , Mice, Inbred C57BL , Molecular Docking Simulation , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors/chemistry , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Phosphoinositide-3 Kinase Inhibitors/toxicity , Podophyllotoxin/chemistry , Podophyllotoxin/pharmacology , Podophyllotoxin/toxicity , Teniposide/toxicity
13.
Curr Med Chem ; 16(3): 327-49, 2009.
Article in English | MEDLINE | ID: mdl-19149581

ABSTRACT

Podophyllotoxin is an important and much sought after antimitotic natural lead compound, since it paved the way for three hemisynthetic derivatives of podophyllotoxin, e.g., etoposide, teniposide and etopophos, which are widely used as anticancer drugs and show good clinical effects against several types of neoplasms. Although the publication of the recent reviews by Gordaliza in 2004 and You in 2005, which covered the literatures concerning podophyllotoxin until the early part of 2003, there have been significant number of works carried out on podophyllotoxin recently. Therefore, this review presents up-to-date coverage of podophyllotoxin in regard to hemisynthesis, biosynthesis, biological activities, mode of action and structure-activity relationship.


Subject(s)
Antineoplastic Agents, Phytogenic , Podophyllotoxin , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line, Tumor , Etoposide/analogs & derivatives , Etoposide/chemistry , Etoposide/pharmacology , Humans , Inhibitory Concentration 50 , Organophosphorus Compounds/chemistry , Organophosphorus Compounds/pharmacology , Podophyllotoxin/analogs & derivatives , Podophyllotoxin/chemistry , Podophyllotoxin/pharmacology , Structure-Activity Relationship , Teniposide/chemistry , Teniposide/pharmacology
14.
J Cell Biol ; 136(1): 29-43, 1997 Jan 13.
Article in English | MEDLINE | ID: mdl-9008701

ABSTRACT

Here we report that DNA decatenation is not a physical requirement for the formation of mammalian chromosomes containing a two-armed chromosome scaffold. 2-aminopurine override of G2 arrest imposed by VM-26 or ICRF-193, which inhibit topoisomerase II (topo II)-dependent DNA decatenation, results in the activation of p34cdc2 kinase and entry into mitosis. After override of a VM-26-dependent checkpoint, morphologically normal compact chromosomes form with paired axial cores containing topo II and ScII. Despite its capacity to form chromosomes of normal appearance, the chromatin remains covalently complexed with topo II at continuous levels during G2 arrest with VM-26. Override of an ICRF-193 block, which inhibits topo II-dependent decatenation at an earlier step than VM-26, also generates chromosomes with two distinct, but elongated, parallel arms containing topo II and ScII. These data demonstrate that DNA decatenation is required to pass a G2 checkpoint, but not to restructure chromatin for chromosome formation. We propose that the chromosome core structure is templated during interphase, before DNA decatenation, and that condensation of the two-armed chromosome scaffold can therefore occur independently of the formation of two intact and separate DNA helices.


Subject(s)
Avian Proteins , Chromosomes/metabolism , DNA/metabolism , G2 Phase/physiology , 2-Aminopurine/pharmacology , Animals , Antimetabolites/pharmacology , CHO Cells , Cell Cycle Proteins , Cell Line , Chromatin/metabolism , Cricetinae , DNA Topoisomerases, Type II/physiology , DNA-Binding Proteins/metabolism , Diketopiperazines , Enzyme Inhibitors/pharmacology , Kidney , Mitosis , Models, Genetic , Nuclear Proteins/metabolism , Piperazines/pharmacology , Teniposide/pharmacology , Topoisomerase II Inhibitors
15.
J Cell Biol ; 160(5): 645-55, 2003 Mar 03.
Article in English | MEDLINE | ID: mdl-12604590

ABSTRACT

The condensin complex and topoisomerase II (topo II) have different biochemical activities in vitro, and both are required for mitotic chromosome condensation. We have used Xenopus egg extracts to investigate the functional interplay between condensin and topo II in chromosome condensation. When unreplicated chromatin is directly converted into chromosomes with single chromatids, the two proteins must function together, although they are independently targeted to chromosomes. In contrast, the requirement for topo II is temporarily separable from that of condensin when chromosome assembly is induced after DNA replication. This experimental setting allows us to find that, in the absence of condensin, topo II becomes enriched in an axial structure within uncondensed chromatin. Subsequent addition of condensin converts this structure into mitotic chromosomes in an ATP hydrolysis-dependent manner. Strikingly, preventing DNA replication by the addition of geminin or aphidicolin disturbs the formation of topo II-containing axes and alters the binding property of topo II with chromatin. Our results suggest that topo II plays an important role in an early stage of chromosome condensation, and that this function of topo II is tightly coupled with prior DNA replication.


Subject(s)
Adenosine Triphosphatases/metabolism , Cell Nucleus/enzymology , Chromosome Structures/metabolism , DNA Replication/genetics , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/metabolism , Eukaryotic Cells/enzymology , Mitosis/genetics , Adenosine Triphosphatases/genetics , Animals , Cell Nucleus/ultrastructure , Chromatin/genetics , Chromatin/metabolism , Chromosome Structures/genetics , DNA Topoisomerases, Type II/genetics , DNA-Binding Proteins/genetics , Eukaryotic Cells/cytology , Female , Male , Multiprotein Complexes , Nucleic Acid Synthesis Inhibitors/pharmacology , Oocytes , Spermatozoa , Teniposide/pharmacology , Xenopus laevis
16.
J Cell Biol ; 111(5 Pt 1): 1753-62, 1990 Nov.
Article in English | MEDLINE | ID: mdl-2172257

ABSTRACT

We have examined the effects of topoisomerase inhibitors on the phosphorylation of histones in chromatin during the G2 and the M phases of the cell cycle. Throughout the G2 phase of BHK cells, addition of the topoisomerase II inhibitor VM-26 prevented histone H1 phosphorylation, accompanied by the inhibition of intracellular histone H1 kinase activity. However, VM-26 had no inhibitory effect on the activity of the kinase in vitro, suggesting an indirect influence on histone H1 kinase activity. Entry into mitosis was also prevented, as monitored by the absence of nuclear lamina depolymerization, chromosome condensation, and histone H3 phosphorylation. In contrast, the topoisomerase I inhibitor, camptothecin, inhibited histone H1 phosphorylation and entry into mitosis only when applied at early G2. In cells that were arrested in mitosis, VM-26 induced dephosphorylation of histones H1 and H3, DNA breaks, and partial chromosome decondensation. These changes in chromatin parameters probably reverse the process of chromosome condensation, unfolding condensed regions to permit the repair of strand breaks in the DNA that were induced by VM-26. The involvement of growth-associated histone H1 kinase in these processes raises the possibility that the cell detects breaks in the DNA through their effects on the state of DNA supercoiling in constrained domains or loops. It would appear that histone H1 kinase and topoisomerase II work coordinately in both chromosome condensation and decondensation, and that this process participates in the VM-26-induced G2 arrest of the cell.


Subject(s)
Chromosomes/metabolism , DNA Topoisomerases, Type II/physiology , Histones/metabolism , Protamine Kinase/metabolism , Teniposide/pharmacology , Animals , Aphidicolin , Cell Cycle/drug effects , Cells, Cultured , Chromosomes/drug effects , DNA Damage , Demecolcine/pharmacology , Diterpenes/pharmacology , G1 Phase , G2 Phase/physiology , Metaphase/physiology , Mitosis/physiology , Nuclear Envelope/metabolism , Phosphorylation , Protamine Kinase/drug effects , Topoisomerase I Inhibitors , Topoisomerase II Inhibitors
17.
J Cell Biol ; 120(3): 613-24, 1993 Feb.
Article in English | MEDLINE | ID: mdl-8381119

ABSTRACT

A normal consequence of mitosis in eukaryotes is the repression of transcription. Using Xenopus egg extracts shifted to a mitotic state by the addition of purified cyclin, we have for the first time been able to reproduce a mitotic repression of transcription in vitro. Active RNA polymerase III transcription is observed in interphase extracts, but strongly repressed in extracts converted to mitosis. With the topoisomerase II inhibitor VM-26, we demonstrate that this mitotic repression of RNA polymerase III transcription does not require normal chromatin condensation. Similarly; in vitro mitotic repression of transcription does not require the presence of nucleosome structure or involve a general repressive chromatin-binding protein, as inhibition of chromatin formation with saturating amounts of non-specific DNA has no effect on repression. Instead, the mitotic repression of transcription appears to be due to phosphorylation of a component of the transcription machinery by a mitotic protein kinase, either cdc2 kinase and/or a kinase activated by it. Mitotic repression of RNA polymerase III transcription is observed both in complete mitotic cytosol and when a kinase-enriched mitotic fraction is added to a highly simplified 5S RNA transcription reaction. We present evidence that, upon depletion of cdc2 kinase, a secondary protein kinase activity remains and can mediate this in vitro mitotic repression of transcription.


Subject(s)
Mitosis/physiology , Oocytes/physiology , Transcription, Genetic , Animals , CDC2 Protein Kinase/isolation & purification , CDC2 Protein Kinase/metabolism , Cell-Free System , Cyclins/pharmacology , Female , Interphase/physiology , Mitosis/drug effects , Models, Biological , Oocytes/cytology , Protamine Kinase/metabolism , Protein Kinases/isolation & purification , Protein Kinases/metabolism , RNA Polymerase III/metabolism , Templates, Genetic , Teniposide/pharmacology , Topoisomerase II Inhibitors , Transcription Factors/isolation & purification , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Xenopus laevis
18.
J Clin Invest ; 129(11): 4850-4862, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31408442

ABSTRACT

Checkpoint blockade antibodies have been approved as immunotherapy for multiple types of cancer, but the response rate and efficacy are still limited. There are few immunogenic cell death (ICD)-inducing drugs available that can kill cancer cells, enhance tumor immunogenicity, increase the in vivo immune infiltration, and thereby boosting a tumor response to immunotherapy. So far, the ICD markers have been identified as the few immuno-stimulating characteristics of dead cells, but whether the presence of such ICD markers on tumor cells translates into enhanced antitumor immunity in vivo is still investigational. To identify anticancer drugs that could induce tumor cell death and boost T cell response, we performed drug screenings based on both an ICD reporter assay and T cell activation assay. We identified that teniposide, a DNA topoisomerase II inhibitor, could induce high mobility group box 1 (HMGB1) release and type I interferon signaling in tumor cells, and teniposide-treated tumor cells could activate antitumor T cell response both in vitro and in vivo. Mechanistically, teniposide induced tumor cell DNA damage and innate immune signaling including NF-κB activation and STING-dependent type I interferon signaling, both of which contribute to the activation of dendritic cells and subsequent T cells. Furthermore, teniposide potentiated the antitumor efficacy of anti-PD1 on multiple types of mouse tumor models. Our findings showed that teniposide could trigger tumor immunogenicity, and enabled a potential chemo-immunotherapeutic approach to potentiate the therapeutic efficacy of anti-PD1 immunotherapy.


Subject(s)
Immunity, Cellular/drug effects , Membrane Proteins/immunology , Neoplasm Proteins/immunology , Neoplasms, Experimental/drug therapy , Nucleotidyltransferases/immunology , Signal Transduction/drug effects , Teniposide/pharmacology , Topoisomerase II Inhibitors/pharmacology , Animals , Cell Line, Tumor , Female , HEK293 Cells , Humans , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Neoplasm Proteins/genetics , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Nucleotidyltransferases/genetics , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Xenograft Model Antitumor Assays
19.
Sci Rep ; 8(1): 13159, 2018 09 03.
Article in English | MEDLINE | ID: mdl-30177851

ABSTRACT

The transcription factor MYB plays key roles in hematopoietic cells and has been implicated the development of leukemia. MYB has therefore emerged as an attractive target for drug development. Recent work has suggested that targeting MYB by small-molecule inhibitors is feasible and that inhibition of MYB has potential as a therapeutic approach against acute myeloid leukemia. To facilitate the identification of small-molecule MYB inhibitors we have re-designed and improved a previously established cell-based screening assay and have employed it to screen a natural product library for potential inhibitors. Our work shows that teniposide and etoposide, chemotherapeutic agents causing DNA-damage by inhibiting topoisomerase II, potently inhibit MYB activity and induce degradation of MYB in AML cell lines. MYB inhibition is suppressed by caffeine, suggesting that MYB is inhibited indirectly via DNA-damage signalling. Importantly, ectopic expression of an activated version of MYB in pro-myelocytic NB4 cells diminished the anti-proliferative effects of teniposide, suggesting that podophyllotoxins disrupt the proliferation of leukemia cells not simply by inducing general DNA-damage but that their anti-proliferative effects are boosted by inhibition of MYB. Teniposide and etoposide therefore act like double-edged swords that might be particularly effective to inhibit tumor cells with deregulated MYB.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Etoposide/pharmacology , Gene Expression Regulation, Leukemic , Podophyllotoxin/pharmacology , Proto-Oncogene Proteins c-myb/antagonists & inhibitors , Teniposide/pharmacology , Cell Line, Tumor , DNA Damage , Dose-Response Relationship, Drug , High-Throughput Screening Assays , Humans , Leukocytes/drug effects , Leukocytes/metabolism , Leukocytes/pathology , Proto-Oncogene Proteins c-myb/genetics , Proto-Oncogene Proteins c-myb/metabolism , Signal Transduction , Small Molecule Libraries/pharmacology
20.
Mol Cell Biol ; 6(4): 985-92, 1986 Apr.
Article in English | MEDLINE | ID: mdl-3023886

ABSTRACT

Similar to its inhibitory effect on mammalian DNA topoisomerase II, the cytotoxic drug VM26 (teniposide) also interferes with the breakage-reunion reaction of Drosophila melanogaster DNA topoisomerase II. VM26 induces topoisomerase II-mediated DNA breakage in vitro and in cultured D. melanogaster cells presumably by stabilizing an enzyme-DNA cleavable complex. The drug-induced DNA breaks on D. melanogaster hsp70 genes were mapped in cultured cells using the indirect end-labeling procedure. Multiple and specific cleavage sites occurred at both the 3' and 5' ends of the hsp70 genes. A number of these cellular topoisomerase II cleavage sites mapped close to the DNase I-hypersensitive regions of the hsp70 genes. The intensities of several topoisomerase II cleavage sites changed significantly on heat shock induction. Treatment of cultured D. melanogaster cells with VM26 at 25 degrees C resulted in the stimulation of transcription of the hsp70 genes. These results suggest that inhibition of DNA topoisomerase II may lead to heat shock transcription.


Subject(s)
Chromatin/metabolism , DNA Topoisomerases, Type II/metabolism , Drosophila melanogaster/genetics , Heat-Shock Proteins/genetics , Animals , DNA Damage , DNA Restriction Enzymes , Drosophila melanogaster/enzymology , Nucleic Acid Hybridization , Teniposide/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL