Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 189
Filter
1.
J Pharmacol Exp Ther ; 389(1): 51-60, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38296645

ABSTRACT

Glioblastoma (GBM) is the most frequently diagnosed primary central nervous system tumor in adults. Despite the standard of care therapy, which includes surgical resection, temozolomide chemotherapy, radiation and the newly added tumor-treating fields, median survival remains only ∼20 months. Unfortunately, GBM has a ∼100% recurrence rate, but after recurrence there are no Food and Drug Administration-approved therapies to limit tumor growth and enhance patient survival, as these tumors are resistant to temozolomide (TMZ). Recently, our laboratory reported that lucanthone slows GBM by inhibiting autophagic flux through lysosome targeting and decreases the number of Olig2+ glioma stem-like cells (GSC) in vitro and in vivo. We now additionally report that lucanthone efficiently abates stemness in patient-derived GSC and reduces tumor microtube formation in GSC, an emerging hallmark of treatment resistance in GBM. In glioma tumors derived from cells with acquired resistance to TMZ, lucanthone retains the ability to perturb tumor growth, inhibits autophagy by targeting lysosomes, and reduces Olig2 positivity. We also find that lucanthone may act as an inhibitor of palmitoyl protein thioesterase 1. Our results suggest that lucanthone may function as a potential treatment option for GBM tumors that are not amenable to TMZ treatment. SIGNIFICANCE STATEMENT: We report that the antischistosome agent lucanthone impedes tumor growth in a preclinical model of temozolomide-resistant glioblastoma and reduces the numbers of stem-like glioma cells. In addition, it acts as an autophagy inhibitor, and its mechanism of action may be via inhibition of palmitoyl protein thioesterase 1. As there are no defined therapies approved for recurrent, TMZ-resistant tumor, lucanthone could emerge as a treatment for glioblastoma tumors that may not be amenable to TMZ both in the newly diagnosed and recurrent settings.


Subject(s)
Brain Neoplasms , Glioblastoma , Glioma , Lucanthone , Humans , Temozolomide/pharmacology , Temozolomide/therapeutic use , Glioblastoma/drug therapy , Glioblastoma/metabolism , Lucanthone/pharmacology , Lucanthone/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/pathology , Glioma/drug therapy , Glioma/pathology , Xenograft Model Antitumor Assays , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , Membrane Proteins , Thiolester Hydrolases
2.
Int J Mol Sci ; 23(1)2021 Dec 21.
Article in English | MEDLINE | ID: mdl-35008442

ABSTRACT

A lucanthone, one of the family of thioxanthenones, has been reported for its inhibitory effects of apurinic endonuclease-1 and autophagy. In this study, we investigated whether lucanthone could enhance tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis in various cancer cells. Combined treatment with lucanthone and TRAIL significantly induced apoptosis in human renal carcinoma (Caki and ACHN), prostate carcinoma (PC3), and lung carcinoma (A549) cells. However, combined treatment did not induce apoptosis in normal mouse kidney cells (TCMK-1) and normal human skin fibroblast (HSF). Lucanthone downregulated protein expression of deubiquitinase DUB3, and a decreased expression level of DUB3 markedly led to enhance TRAIL-induced apoptosis. Ectopic expression of DUB3 inhibited combined treatment with lucanthone and TRAIL-induced apoptosis. Moreover, lucanthone increased expression level of DR5 mRNA via downregulation of miR-216a-5p. Transfection of miR-216a-5p mimics suppressed the lucanthone-induced DR5 upregulation. Taken together, these results provide the first evidence that lucanthone enhances TRAIL-induced apoptosis through DR5 upregulation by downregulation of miR-216a-5p and DUB3-dependent Mcl-1 downregulation in human renal carcinoma cells.


Subject(s)
Endopeptidases/metabolism , Lucanthone/pharmacology , MicroRNAs/genetics , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Neoplasms/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/pharmacology , A549 Cells , Animals , Autophagy/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Down-Regulation , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , PC-3 Cells , Up-Regulation
3.
SLAS Discov ; 26(3): 345-351, 2021 03.
Article in English | MEDLINE | ID: mdl-33267713

ABSTRACT

A novel bioinformatic approach for drug repurposing against emerging viral epidemics like Covid-19 is described. It exploits the COMPARE algorithm, a public program from the National Cancer Institute (NCI) to sort drugs according to their patterns of growth inhibitory profiles from a diverse panel of human cancer cell lines. The data repository of the NCI includes the growth inhibitory patterns of more than 55,000 molecules. When candidate drug molecules with ostensible anti-SARS-CoV-2 activities were used as seeds (e.g., hydroxychloroquine, ritonavir, and dexamethasone) in COMPARE, the analysis uncovered several molecules with fingerprints similar to the seeded drugs. Interestingly, despite the fact that the uncovered drugs were from various pharmacological classes (antiarrhythmic, nucleosides, antipsychotic, alkaloids, antibiotics, and vitamins), they were all reportedly known from published literature to exert antiviral activities via different modes, confirming that COMPARE analysis is efficient for predicting antiviral activities of drugs from various pharmacological classes. Noticeably, several of the uncovered drugs can be readily tested, like didanosine, methotrexate, vitamin A, nicotinamide, valproic acid, uridine, and flucloxacillin. Unlike pure in silico methods, this approach is biologically more relevant and able to pharmacologically correlate compounds regardless of their chemical structures. This is an untapped resource, reliable and readily exploitable for drug repurposing against current and future viral outbreaks.


Subject(s)
Antiviral Agents/pharmacology , Computational Biology/methods , Drug Repositioning/methods , Algorithms , COVID-19 , Cell Line , Data Mining/methods , Databases, Pharmaceutical , Dexamethasone/chemistry , Dexamethasone/pharmacology , Drug Discovery/methods , Humans , Lucanthone/pharmacology , SARS-CoV-2/drug effects
4.
J Chem Inf Model ; 56(9): 1872-83, 2016 09 26.
Article in English | MEDLINE | ID: mdl-27564845

ABSTRACT

Although Camptothecin and its analogs as Topoisomerase I poisons can effectively treat cancers, serious drug resistance has been identified for this class of drugs. Recent computational studies have indicated that the mutations near the active binding site of the drug can significantly weaken the drug binding and cause drug resistance. However, only Topotecan and three mutations have been previously analyzed. Here we present a comprehensive binding study of 10 Topoisomerase I mutants (N722S, N722A, D533G, D533N, G503S, G717V, T729A, F361S, G363C, and R364H) and 8 poisons including 7 Camptothecin analogs as well as a new generation Topoisomerase I drug, Lucanthone. Utilizing Glide docking followed by MMGBSA calculations, we determined the binding energy for each complex. We examine the relative binding energy changes with reference to the wild type, which are linked to the degree of drug resistance. On this set of mutant complexes, Topotecan and Camptothecin showed much smaller binding energies than a set of new Camptothecin derivatives (Lurtotecan, SN38, Gimatecan, Exatecan, and Belotecan) currently under clinical trials. We observed that Lucanthone exhibited comparable results to Topotecan and Camptothecin, indicating that it may serve as a promising candidate for future studies as a Topoisomerase I poison. Our docked results on Topotecan were also validated by a set of molecular dynamics simulations. In addition to a good agreement on the MMGBSA binding energy change, our simulation data also shows there is larger conformation fluctuation upon the mutations. These results may be utilized to further advancements of Topoisomerase I drugs that are resistant to mutations.


Subject(s)
Antineoplastic Agents/pharmacology , Camptothecin/pharmacology , DNA Topoisomerases, Type I/genetics , Drug Resistance, Neoplasm/genetics , Lucanthone/pharmacology , Models, Molecular , Mutation , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Camptothecin/chemistry , Camptothecin/metabolism , DNA Topoisomerases, Type I/chemistry , DNA Topoisomerases, Type I/metabolism , Humans , Lucanthone/chemistry , Lucanthone/metabolism , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Conformation , Sequence Homology, Amino Acid , Thermodynamics
5.
Nanomedicine ; 11(1): 109-18, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25131339

ABSTRACT

We report use of PEG-DSPE coated oxidized graphene nanoribbons (O-GNR-PEG-DSPE) as agent for delivery of anti-tumor drug Lucanthone (Luc) into Glioblastoma Multiformae (GBM) cells targeting base excision repair enzyme APE-1 (Apurinic endonuclease-1). Lucanthone, an endonuclease inhibitor of APE-1, was loaded onto O-GNR-PEG-DSPEs using a simple non-covalent method. We found its uptake by GBM cell line U251 exceeding 67% and 60% in APE-1-overexpressing U251, post 24h. However, their uptake was ~38% and 29% by MCF-7 and rat glial progenitor cells (CG-4), respectively. TEM analysis of U251 showed large aggregates of O-GNR-PEG-DSPE in vesicles. Luc-O-GNR-PEG-DSPE was significantly toxic to U251 but showed little/no toxicity when exposed to MCF-7/CG-4 cells. This differential uptake effect can be exploited to use O-GNR-PEG-DSPEs as a vehicle for Luc delivery to GBM, while reducing nonspecific cytotoxicity to the surrounding healthy tissue. Cell death in U251 was necrotic, probably due to oxidative degradation of APE-1.


Subject(s)
Brain Neoplasms/drug therapy , Drug Delivery Systems , Glioblastoma/drug therapy , Graphite/chemistry , Lucanthone/chemistry , Animals , Cell Line, Tumor , Coculture Techniques , DNA-(Apurinic or Apyrimidinic Site) Lyase/chemistry , Flow Cytometry , Humans , Hydrogen-Ion Concentration , Mice , Microscopy, Electron, Transmission , Nanotubes, Carbon/chemistry , Neoplasm Transplantation , Neuroglia/metabolism , Oxygen/chemistry , Rats , Stem Cells/cytology
6.
Xenobiotica ; 44(1): 89-93, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23777287

ABSTRACT

1. Once in a while, during drug metabolism studies, an unusual or unexpected pathway is unearthed. 2. Such quirky finds open a refreshing hiatus, providing a departure from the, perhaps now mundane, textbook routes. 3. This brief missive draws attention to an interesting anecdote that may be unknown to some and concerns a substituted thioxanthenone drug.


Subject(s)
Lucanthone/chemistry , Lucanthone/metabolism , Lucanthone/pharmacokinetics , Metabolic Networks and Pathways/physiology , Toluidines/chemistry , Humans , Lucanthone/urine , Molecular Structure , Toluidines/urine
7.
Angew Chem Int Ed Engl ; 52(31): 7936-56, 2013 Jul 29.
Article in English | MEDLINE | ID: mdl-23813602

ABSTRACT

After malaria, schistosomiasis (or bilharzia) is the second most prevalent disease in Africa, and is occurring in over 70 countries in tropical and subtropical regions. It is estimated that 600 million people are at risk of infection, 200 million people are infected, and at least 200,000 deaths per year are associated with the disease. All schistosome species are transmitted through contact with fresh water that is infested with free-swimming forms of the parasite, which is known as cercariae and produced by snails. When located in the blood vessels of the host, larval and adult schistosomes digest red cells to acquire amino acids for growth and development. Vaccine candidates have been unsuccessful up to now. Against such devastating parasitic disease, the antischistosomal arsenal is currently limited to a single drug, praziquantel, which has been used for more than 35 years. Because the question of the reduction of the activity of praziquantel was raised recently, it is thus urgent to create new and safe antischistosomal drugs that should be combined with praziquantel to develop efficient bitherapies.


Subject(s)
Imidazoles/therapeutic use , Niacin/analogs & derivatives , Praziquantel/therapeutic use , Schistosomiasis/drug therapy , Animals , Drug Resistance , Hemeproteins/metabolism , Hemoglobins/metabolism , Humans , Lucanthone/analogs & derivatives , Lucanthone/chemistry , Lucanthone/therapeutic use , Niacin/therapeutic use , Oxadiazoles/chemistry , Oxadiazoles/therapeutic use , Praziquantel/pharmacology , Schistosoma/drug effects , Schistosoma/growth & development , Schistosoma/metabolism , Schistosomiasis/parasitology
8.
PLoS One ; 6(9): e23679, 2011.
Article in English | MEDLINE | ID: mdl-21935361

ABSTRACT

Lucanthone and hycanthone are thioxanthenone DNA intercalators used in the 1980s as antitumor agents. Lucanthone is in Phase I clinical trial, whereas hycanthone was pulled out of Phase II trials. Their potential mechanism of action includes DNA intercalation, inhibition of nucleic acid biosyntheses, and inhibition of enzymes like topoisomerases and the dual function base excision repair enzyme apurinic endonuclease 1 (APE1). Lucanthone inhibits the endonuclease activity of APE1, without affecting its redox activity. Our goal was to decipher the precise mechanism of APE1 inhibition as a prerequisite towards development of improved therapeutics that can counteract higher APE1 activity often seen in tumors. The IC(50) values for inhibition of APE1 incision of depurinated plasmid DNA by lucanthone and hycanthone were 5 µM and 80 nM, respectively. The K(D) values (affinity constants) for APE1, as determined by BIACORE binding studies, were 89 nM for lucanthone/10 nM for hycanthone. APE1 structures reveal a hydrophobic pocket where hydrophobic small molecules like thioxanthenones can bind, and our modeling studies confirmed such docking. Circular dichroism spectra uncovered change in the helical structure of APE1 in the presence of lucanthone/hycanthone, and notably, this effect was decreased (Phe266Ala or Phe266Cys or Trp280Leu) or abolished (Phe266Ala/Trp280Ala) when hydrophobic site mutants were employed. Reduced inhibition by lucanthone of the diminished endonuclease activity of hydrophobic mutant proteins (as compared to wild type APE1) supports that binding of lucanthone to the hydrophobic pocket dictates APE1 inhibition. The DNA binding capacity of APE1 was marginally inhibited by lucanthone, and not at all by hycanthone, supporting our hypothesis that thioxanthenones inhibit APE1, predominantly, by direct interaction. Finally, lucanthone-induced degradation was drastically reduced in the presence of short and long lived free radical scavengers, e.g., TRIS and DMSO, suggesting that the mechanism of APE1 breakdown may involve free radical-induced peptide bond cleavage.


Subject(s)
DNA-(Apurinic or Apyrimidinic Site) Lyase/antagonists & inhibitors , Hycanthone/chemistry , Lucanthone/chemistry , Cell Line, Tumor , Circular Dichroism , DNA/chemistry , Glioblastoma/metabolism , Humans , Hydrogen Bonding , Indoles/pharmacology , Inhibitory Concentration 50 , Mutation , Oxidation-Reduction , Plasmids/metabolism , Protein Binding , Recombinant Proteins , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
9.
J Biol Chem ; 286(8): 6602-13, 2011 Feb 25.
Article in English | MEDLINE | ID: mdl-21148553

ABSTRACT

Cellular stress induced by nutrient deprivation, hypoxia, and exposure to many chemotherapeutic agents activates an evolutionarily conserved cell survival pathway termed autophagy. This pathway enables cancer cells to undergo self-digestion to generate ATP and other essential biosynthetic molecules to temporarily avoid cell death. Therefore, disruption of autophagy may sensitize cancer cells to cell death and augment chemotherapy-induced apoptosis. Chloroquine and its analog hydroxychloroquine are the only clinically relevant autophagy inhibitors. Because both of these agents induce ocular toxicity, novel inhibitors of autophagy with a better therapeutic index are needed. Here we demonstrate that the small molecule lucanthone inhibits autophagy, induces lysosomal membrane permeabilization, and possesses significantly more potent activity in breast cancer models compared with chloroquine. Exposure to lucanthone resulted in processing and recruitment of microtubule-associated protein 1 light chain 3 (LC3) to autophagosomes, but impaired autophagic degradation as revealed by transmission electron microscopy and the accumulation of p62/SQSTM1. Microarray analysis, qRT-PCR, and immunoblotting determined that lucanthone stimulated a large induction in cathepsin D, which correlated with cell death. Accordingly, knockdown of cathepsin D reduced lucanthone-mediated apoptosis. Subsequent studies using p53(+/+) and p53(-/-) HCT116 cells established that lucanthone induced cathepsin D expression and reduced cancer cell viability independently of p53 status. In addition, lucanthone enhanced the anticancer activity of the histone deacetylase inhibitor vorinostat. Collectively, our results demonstrate that lucanthone is a novel autophagic inhibitor that induces apoptosis via cathepsin D accumulation and enhances vorinostat-mediated cell death in breast cancer models.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Breast Neoplasms/drug therapy , Cathepsin D/metabolism , Lucanthone/pharmacology , Schistosomicides/pharmacology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Antineoplastic Agents/agonists , Antineoplastic Agents/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cathepsin D/genetics , Cell Line, Tumor , Drug Synergism , Gene Expression Profiling , Humans , Hydroxamic Acids/agonists , Hydroxamic Acids/pharmacology , Intracellular Membranes/metabolism , Lucanthone/agonists , Lysosomes/genetics , Lysosomes/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Permeability/drug effects , Phagosomes/genetics , Phagosomes/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Schistosomicides/agonists , Sequestosome-1 Protein , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Vorinostat
10.
J Radiat Res ; 51(4): 393-404, 2010.
Article in English | MEDLINE | ID: mdl-20679741

ABSTRACT

Since radiation therapy remains a primary treatment modality for gliomas, the radioresistance of glioma cells and targets to modify their radiation tolerance are of significant interest. Human apurinic endonuclease 1 (Ape1, Ref-1, APEX, HAP1, AP endo) is a multifunctional protein involved in base excision repair of DNA and a redox-dependent transcriptional co-activator. This study investigated whether there is a direct relationship between Ape1 and radioresistance in glioma cells, employing the human U87 and U251 cell lines. U87 is intrinsically more radioresistant than U251, which is partly attributable to more cycling U251 cells found in G2/M, the most radiosensitive cell stage, while more U87 cells are found in S and G1, the more radioresistant cell stages. But observed radioresistance is also related to Ape1 activity. U87 has higher levels of Ape1 than does U251, as assessed by Western blot and enzyme activity assays (approximately 1.5-2 fold higher in cycling cells, and approximately 10 fold higher at G2/M). A direct relationship was seen in cells transfected with CMV-Ape1 constructs; there was a dose-dependent relationship between increasing Ape1 overexpression and increasing radioresistance. Conversely, knock down by siRNA or by pharmacological down regulation of Ape1 resulted in decreased radioresistance. The inhibitors lucanthone and CRT004876 were employed, the former a thioxanthene previously under clinical evaluation as a radiosensitizer for brain tumors and the latter a more specific Ape1 inhibitor. These data suggest that Ape1 may be a useful target for modifying radiation tolerance.


Subject(s)
DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Glioma/metabolism , Glioma/radiotherapy , Base Sequence , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/radiotherapy , Cell Cycle , Cell Line, Tumor , DNA Damage , DNA Repair , DNA-(Apurinic or Apyrimidinic Site) Lyase/antagonists & inhibitors , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Gene Expression , Glioma/genetics , Glioma/pathology , Humans , Indoles/pharmacology , Lucanthone/pharmacology , RNA, Small Interfering/genetics , Radiation Tolerance/drug effects , Radiation Tolerance/genetics , Radiation Tolerance/physiology
11.
Mol Aspects Med ; 28(3-4): 375-95, 2007.
Article in English | MEDLINE | ID: mdl-17560642

ABSTRACT

With our growing understanding of the pathways involved in cell proliferation and signaling, targeted therapies, in the treatment of cancer are entering the clinical arena. New and emerging targets are proteins involved in DNA repair pathways. Inhibition of various proteins in the DNA repair pathways sensitizes cancer cells to DNA damaging agents such as chemotherapy and/or radiation. We study the apurinic endonuclease 1/redox factor-1 (Ape1/Ref-1) and believe that its crucial function in DNA repair and reduction-oxidation or redox signaling make it an excellent target for sensitizing tumor cells to chemotherapy. Ape1/Ref-1 is an essential enzyme in the base excision repair (BER) pathway which is responsible for the repair of DNA caused by oxidative and alkylation damage. As importantly, Ape1/Ref-1 also functions as a redox factor maintaining transcription factors in an active reduced state. Ape1/Ref-1 stimulates the DNA binding activity of numerous transcription factors that are involved in cancer promotion and progression such as AP-1 (Fos/Jun), NFkappaB, HIF-1alpha, CREB, p53 and others. We will discuss what is known regarding the pharmacological targeting of the DNA repair activity, as well as the redox activity of Ape1/Ref-1, and explore the budding clinical utility of inhibition of either of these functions in cancer treatment. A brief discussion of the effect of polymorphisms in its DNA sequence is included because of Ape1/Ref-1's importance to maintenance and integrity of the genome. Experimental modification of Ape1/Ref-1 activity changes the response of cells and of organisms to DNA damaging agents, suggesting that Ape1/Ref-1 may also be a productive target of chemoprevention. In this review, we will provide an overview of Ape1/Ref-1's activities and explore the potential of this protein as a target in cancer treatment as well as its role in chemoprevention.


Subject(s)
DNA Repair , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Neoplasms , Benzoquinones/metabolism , Carboxylic Acids/chemistry , Carboxylic Acids/metabolism , DNA Damage , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Humans , Hydroxylamines/metabolism , Isoflavones/metabolism , Lucanthone/metabolism , Neoplasms/metabolism , Neoplasms/prevention & control , Neoplasms/therapy , Oxidation-Reduction , Oxidative Stress , Propionates/metabolism , Resveratrol , Stilbenes/metabolism
12.
Cell Stress Chaperones ; 10(1): 37-45, 2005.
Article in English | MEDLINE | ID: mdl-15832946

ABSTRACT

Pretreatment of human leukemia THP-1 cells with heat shock protein Hsp70 (Hsp70) protected them from the cell-lethal effects of the topoisomerase II inhibitor, lucanthone and from ionizing radiation. Cell viability was scored in clonogenic assays of single cells grown in liquid medium containing 0.5% methyl cellulose. Colonies were observed and rapidly scored after staining with the tetrazolium salt, 3-(4,5-dimethylthiazol-2-yl)-2,5 diphenyl tetrazolium bromide. The frequency of abasic sites in the deoxyribonucleic acid (DNA) of THP-1 cells was reduced when these cells were treated with Hsp70. Hsp70 is presumed to have protected the cells by promoting repair of cell DNA, in agreement with previous studies that showed that Hsp70 enhanced base excision repair by purified enzymes. The shoulders of radiation dose-response curves were enhanced by pretreatment of cells with Hsp70 and, importantly, were reduced when cells were transfected with ribonucleic acid designed to silence Hsp70. Hsp70 influenced repair of sublethal damage after radiation.


Subject(s)
Colony-Forming Units Assay , Enzyme Inhibitors/pharmacology , Gamma Rays/adverse effects , HSP70 Heat-Shock Proteins/metabolism , Lucanthone/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Cytoprotection , DNA Damage/drug effects , DNA Damage/radiation effects , DNA Repair , Dose-Response Relationship, Radiation , Humans , Leukemia, Monocytic, Acute/pathology , RNA, Small Interfering/metabolism
13.
Anticancer Res ; 24(4): 2127-34, 2004.
Article in English | MEDLINE | ID: mdl-15330152

ABSTRACT

Cells repair DNA damage via four main mechanisms, however, damage induced by alkylators and oxidative damage is predominantly repaired by the DNA base excision repair (BER) pathway. The AP endonuclease, APE1, is one of the main enzymes in the BER pathway. It is abundant in human cells and accounts for nearly all of the abasic site cleavage activity observed in cellular extracts. APE1 expression is elevated in a variety of cancers and a high APE1 expression has been associated with poor outcome to chemoradiotherapy. The small molecule lucanthone has been shown to enhance the killing ability of ionizing radiation in cells and preliminary evidence suggests that lucanthone may inhibit AP endonuclease. Given the role APE1 plays in repairing oxidative and ionizing radiation DNA damage, the reports of lucanthone as an ionizing radiation enhancer and the potential use of lucanthone as an AP endonuclease inhibitor, we examined whether lucanthone could inhibit APE1 endonuclease activity. We report that lucanthone inhibits the repair activity of APE1, but not its redox function or exonuclease activity on mismatched nucleotides. Lucanthone also appears to inhibit exonuclease III family members (APE1 and ExoIII), but not endonuclease IV AP endonucleases, nor bifunctional glycosylase/lyases such as endonuclease VIII or formamidopyrimidine-DNA glycosylase (Fpg). Furthermore, the addition of lucanthone inhibits APE1 repair activity from cellular extracts and enhances the cell killing effect of the laboratory alkylating agent methyl methanesulfonate (MMS) and the clinically relevant agent temozolomide (TMZ). Given these initial findings, it would be of interest to further develop lucanthone as an APE1 inhibitor through the use of structure-function studies as a means of enhancing the sensitization of tumors to chemotherapeutic agents.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , DNA Repair/drug effects , DNA-(Apurinic or Apyrimidinic Site) Lyase/antagonists & inhibitors , Lucanthone/pharmacology , Antineoplastic Agents, Alkylating/administration & dosage , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Cell Line, Tumor , DNA Repair/physiology , DNA, Neoplasm/drug effects , DNA, Neoplasm/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Drug Synergism , Endonucleases/antagonists & inhibitors , Endonucleases/metabolism , Humans , Lucanthone/administration & dosage
14.
J Magn Reson ; 164(1): 128-35, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12932464

ABSTRACT

Three- and four-frequency nuclear magnetic-resonance probes have been designed for the study of small amounts of protein. Both "HX" (1H, X, and 2H channels) and "triple-resonance" (1H, 15N, 13C, and 2H) probes were implemented using a single transmit/receive coil and multiple-frequency impedance matching circuits. The coil used was a six-turn solenoid with an observe volume of 15 microl. A variable pitch design was used to improve the B1 homogeneity of the coil. Two-dimensional HSQC spectra of approximately 1mM single labeled 15N- and double labeled 15N/13C-proteins were acquired in experimental times of approximately 2h. Triple-resonance capability of the small-volume triple-resonance probe was demonstrated by acquiring three-dimensional HNCO spectra from the same protein samples. In addition to enabling very small quantities of protein to be used, the extremely short pulse widths (1H = 4, 15N = 4, and 13C = 2 micros) of this particular design result in low power decoupling and wide-bandwidth coverage, an important factor for the ever-higher operating frequencies used for protein NMR studies.


Subject(s)
Lucanthone/analogs & derivatives , Magnetics/instrumentation , Microchemistry/instrumentation , Nuclear Magnetic Resonance, Biomolecular/instrumentation , Proteins/chemistry , Transducers , Carbon Isotopes , Equipment Design , Lucanthone/chemistry , Microchemistry/methods , Nitrogen Isotopes , Nuclear Magnetic Resonance, Biomolecular/methods , Protons , Quality Control , Reproducibility of Results , Sensitivity and Specificity , Ubiquitin/chemistry
15.
Cancer Invest ; 20(7-8): 983-91, 2002.
Article in English | MEDLINE | ID: mdl-12449731

ABSTRACT

Abasic sites in HeLa cell DNA were increased in frequency by exposing the cells to lucanthone. Cell growth in the presence of lucanthone caused progressive accumulation of abasic sites and loss of cellular DNA. After 2 hr in 8 microM lucanthone, the abundance of abasic sites was 2.4 fold greater than the background of 9.9 +/- 2.0 SE abasic sites/10(6) nucleotides; 80 microM lucanthone in the growth medium increased the level 12.6 +/- 2.5 SE fold and decreased the DNA content in HeLa cells to one-half of the value obtained in untreated cells. The frequency of abasic sites in cellular DNA was determined by the aldehyde reactive probe method, with reference to abasic sites created in plasmid pBR322. The ability of lucanthone to inhibit the normal repair of abasic sites might reflect inhibition of apurinic/apyrimidinic endonuclease (HAP1) by the drug, thereby preventing an early step in the base excision repair pathway. Unrepaired abasic sites prevalent after ionizing radiation are cytotoxic lesions that promote DNA strand breaks. These results suggest a rationale for the joint lethal effects of lucanthone and ionizing radiation in cells and the accelerated tumor regression observed in cancer patients who received the combined therapy.


Subject(s)
Carbon-Oxygen Lyases/metabolism , DNA Damage/drug effects , DNA, Neoplasm/drug effects , HeLa Cells/drug effects , Lucanthone/pharmacology , Binding Sites , Carbon-Oxygen Lyases/genetics , DNA Repair/drug effects , DNA-(Apurinic or Apyrimidinic Site) Lyase , HeLa Cells/enzymology , Humans , Plasmids/drug effects
16.
Biochem Pharmacol ; 58(8): 1307-12, 1999 Oct 15.
Article in English | MEDLINE | ID: mdl-10487533

ABSTRACT

Lucanthone is an antitumour drug used as an adjuvant in radiation therapy. The drug intercalates into DNA and inhibits topoisomerase II. An indazole analogue of lucanthone (IA-5) was examined for its ability to modulate topoisomerase II-DNA cleavable complex formation in vitro. The drug contains a methylbenzothiopyranoindazole chromophore instead of the methyl-thioxanthenone nucleus of lucanthone. Using a radiolabelled linear plasmid DNA as a substrate, both lucanthone and the indazole analogue were shown to promote the cleavage of DNA by human topoisomerase II. Sequencing experiments with different restriction fragments indicated that the indazole drug promoted DNA cleavage primarily at sites having a C on the 3' side of the cleaved bond (-1 position). By contrast, in the same sequencing methodology lucanthone exerted a much weaker effect on topoisomerase II. The sequence selectivity of IA-5 is reminiscent of that of the anticancer drug mitoxantrone and its anthrapyrazole analogue losoxantrone, which is structurally close to IA-5. Binding to DNA and topoisomerase II inhibition are two distinct processes contributing separately to the cytotoxic activity of the indazole drug.


Subject(s)
DNA Topoisomerases, Type II/metabolism , DNA/drug effects , Indazoles/pharmacology , Lucanthone/pharmacology , DNA/metabolism , DNA Topoisomerases, Type II/drug effects , Humans , Indazoles/chemistry , Lucanthone/analogs & derivatives , Molecular Structure
17.
Int J Radiat Oncol Biol Phys ; 43(1): 89-93, 1999 Jan 01.
Article in English | MEDLINE | ID: mdl-9989518

ABSTRACT

PURPOSE: To determine if lucanthone crossed the blood-brain barrier in experimental animals; and to determine accelerated tumor regression of human brain metastases treated jointly with lucanthone and whole brain radiation. METHODS AND MATERIALS: The organ distribution of 3H lucanthone in mice and 125I lucanthone in rats was determined to learn if lucanthone crossed the blood-brain barrier. Size determinations were made of patients' brain metastases from magnetic resonance images or by computed tomography before and after treatment with 30 Gy whole brain radiation alone or with lucanthone. RESULTS: The time course of lucanthone's distribution in brain was identical to that in muscle and heart after intraperitoneal or intravenous administration in experimental animals. Lucanthone, therefore, readily crossed the blood-brain barrier in experimental animals. CONCLUSION: Compared with radiation alone, the tumor regression in patients with brain metastases treated with lucanthone and radiation was accelerated, approaching significance using a permutation test at p = 0.0536.


Subject(s)
Blood-Brain Barrier , Brain Neoplasms/radiotherapy , Brain Neoplasms/secondary , Enzyme Inhibitors/therapeutic use , Lucanthone/therapeutic use , Topoisomerase II Inhibitors , Animals , Brain Neoplasms/blood , Enzyme Inhibitors/blood , Enzyme Inhibitors/pharmacokinetics , Female , Humans , Lucanthone/blood , Lucanthone/pharmacokinetics , Male , Mice , Mice, Inbred C3H , Rats , Rats, Sprague-Dawley
18.
Int J Radiat Oncol Biol Phys ; 37(5): 1133-7, 1997 Mar 15.
Article in English | MEDLINE | ID: mdl-9169823

ABSTRACT

PURPOSE: To determine whether lucanthone can inhibit human topoisomerases in vitro. METHODS AND MATERIALS: Lucanthone was incubated with human topoisomerases II and I together with their plasmid substrates, to determine if lucanthone interfered with the catalytic activities of topoisomerases and if it enhanced the formation of DNA strand breaks, as determined by agarose gel electrophoresis of the resultant plasmid forms. RESULTS: Incubation of the enzymes with lucanthone inhibited the catalytic activity of topoisomerases II and I. With topoisomerase II, it increased the abundance of DNA double strand breaks (cleavable complexes). CONCLUSION: Lucanthone, like actinomycin D, inhibited topoisomerases II and I. It may act to enhance the yield of DNA double strand breaks in cells through a mechanism of topoisomerase II inhibition.


Subject(s)
DNA Damage , DNA, Superhelical/drug effects , Lucanthone/pharmacology , Topoisomerase I Inhibitors , Topoisomerase II Inhibitors , Antineoplastic Agents, Phytogenic/pharmacology , Etoposide/pharmacology , Humans
19.
Biochemistry ; 32(23): 5985-93, 1993 Jun 15.
Article in English | MEDLINE | ID: mdl-8389585

ABSTRACT

DNAase I footprinting has been used to probe the DNA sequence selectivity of the antitumor intercalating agents lucanthone (1), hycanthone (2), 6-chlorolucanthone (7), and four indazole analogs (IA-3-IA-6). The latter have a benzothiopyranoindazole chromophore substituted with a diethylaminoethyl side chain identical to that attached to the thioxanthenone chromophore of compounds 1, 2, and 7. IA-3 and IA-5 are lucanthone analogs bearing a methyl group at position 4, whereas IA-4 and IA-6 are hycanthone analogs bearing a hydroxymethyl group. IA-3 and IA-4 have an additional chloro group at position 6. Studies employing the 160-bp tyrT DNA fragment as substrate to assay inhibition of DNAase I-mediated cleavage show that both lucanthone and hycanthone bind preferentially to AT sites. They discriminate against GC-rich sequences as well as short runs of a single base, which are often cut more readily in the presence of the drugs compared to the control. The indazole analogs exhibit more pronounced selectivity of binding to AT sequences and promote enhanced DNAase I cleavage both at GC-rich sequences and at homooligomeric runs of adenines or thymines. The results of further DNAase I cleavage inhibition assays, performed with three more restriction fragments having different base pair arrangements, are fully consistent with those obtained with the tyrT fragment. They reveal that the preferred binding sequences for lucanthone, hycanthone, and the indazole analogs are predominantly composed of alternating A and T residues.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
DNA/metabolism , Hycanthone/metabolism , Indazoles/metabolism , Intercalating Agents/metabolism , Lucanthone/metabolism , Base Sequence , DNA/chemistry , DNA Restriction Enzymes , Deoxyribonuclease I , Molecular Sequence Data , Osmium Tetroxide , Substrate Specificity
20.
Article in English | MEDLINE | ID: mdl-1360350

ABSTRACT

1. Carbohydrates were extracted from total tissue extracts of Biomphalaria alexandrina snails and were analyzed to their monosaccharides using GLC. 2. The snails were chemically treated with thioxanthone derivatives (compounds I, II, III) and the change in the monosaccharide constituents of their carbohydrates was investigated. 3. The isolated monosaccharides from native and chemically pretreated snails were injected into mice and their protective effects were examined after infection of mice with cercariae of Schistosoma mansoni. 4. The results showed that the main monosaccharides in carbohydrates of snails were galactose, glucose, fucose and mannose and that chemical treatment caused a drop in the galactose content. 5. Moreover, monosaccharide fractions from snails treated with compound III were the most effective in inducing protection against Schistosoma infection in mice.


Subject(s)
Biomphalaria/chemistry , Biomphalaria/drug effects , Hycanthone/pharmacology , Lucanthone/pharmacology , Monosaccharides/immunology , Schistosomiasis mansoni/prevention & control , Thioxanthenes/pharmacology , Animals , Biomphalaria/immunology , Carbohydrates/chemistry , Carbohydrates/isolation & purification , Intestines/parasitology , Lethal Dose 50 , Liver/parasitology , Mice , Monosaccharides/isolation & purification , Parasite Egg Count , Schistosomiasis mansoni/immunology , Vaccines/immunology , Xanthones
SELECTION OF CITATIONS
SEARCH DETAIL
...