Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 189
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
J Pharmacol Exp Ther ; 389(1): 51-60, 2024 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-38296645

RESUMO

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.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Glioma , Lucantona , Humanos , Temozolomida/farmacologia , Temozolomida/uso terapêutico , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Lucantona/farmacologia , Lucantona/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Recidiva Local de Neoplasia/tratamento farmacológico , Recidiva Local de Neoplasia/patologia , Glioma/tratamento farmacológico , Glioma/patologia , Ensaios Antitumorais Modelo de Xenoenxerto , Antineoplásicos Alquilantes/farmacologia , Antineoplásicos Alquilantes/uso terapêutico , Proteínas de Membrana , Tioléster Hidrolases
2.
Int J Mol Sci ; 23(1)2021 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-35008442

RESUMO

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.


Assuntos
Endopeptidases/metabolismo , Lucantona/farmacologia , MicroRNAs/genética , Proteína de Sequência 1 de Leucemia de Células Mieloides/genética , Neoplasias/genética , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Células A549 , Animais , Autofagia/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Regulação para Baixo , Sinergismo Farmacológico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Células PC-3 , Regulação para Cima
3.
J Chem Inf Model ; 56(9): 1872-83, 2016 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-27564845

RESUMO

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.


Assuntos
Antineoplásicos/farmacologia , Camptotecina/farmacologia , DNA Topoisomerases Tipo I/genética , Resistencia a Medicamentos Antineoplásicos/genética , Lucantona/farmacologia , Modelos Moleculares , Mutação , Antineoplásicos/química , Antineoplásicos/metabolismo , Camptotecina/química , Camptotecina/metabolismo , DNA Topoisomerases Tipo I/química , DNA Topoisomerases Tipo I/metabolismo , Humanos , Lucantona/química , Lucantona/metabolismo , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Conformação Proteica , Homologia de Sequência de Aminoácidos , Termodinâmica
4.
Nanomedicine ; 11(1): 109-18, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25131339

RESUMO

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.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Sistemas de Liberação de Medicamentos , Glioblastoma/tratamento farmacológico , Grafite/química , Lucantona/química , Animais , Linhagem Celular Tumoral , Técnicas de Cocultura , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/química , Citometria de Fluxo , Humanos , Concentração de Íons de Hidrogênio , Camundongos , Microscopia Eletrônica de Transmissão , Nanotubos de Carbono/química , Transplante de Neoplasias , Neuroglia/metabolismo , Oxigênio/química , Ratos , Células-Tronco/citologia
5.
Xenobiotica ; 44(1): 89-93, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23777287

RESUMO

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.


Assuntos
Lucantona/química , Lucantona/metabolismo , Lucantona/farmacocinética , Redes e Vias Metabólicas/fisiologia , Toluidinas/química , Humanos , Lucantona/urina , Estrutura Molecular , Toluidinas/urina
6.
Angew Chem Int Ed Engl ; 52(31): 7936-56, 2013 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-23813602

RESUMO

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.


Assuntos
Imidazóis/uso terapêutico , Niacina/análogos & derivados , Praziquantel/uso terapêutico , Esquistossomose/tratamento farmacológico , Animais , Resistência a Medicamentos , Hemeproteínas/metabolismo , Hemoglobinas/metabolismo , Humanos , Lucantona/análogos & derivados , Lucantona/química , Lucantona/uso terapêutico , Niacina/uso terapêutico , Oxidiazóis/química , Oxidiazóis/uso terapêutico , Praziquantel/farmacologia , Schistosoma/efeitos dos fármacos , Schistosoma/crescimento & desenvolvimento , Schistosoma/metabolismo , Esquistossomose/parasitologia
7.
J Biol Chem ; 286(8): 6602-13, 2011 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-21148553

RESUMO

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.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Catepsina D/metabolismo , Lucantona/farmacologia , Esquistossomicidas/farmacologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Antineoplásicos/agonistas , Antineoplásicos/farmacologia , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Catepsina D/genética , Linhagem Celular Tumoral , Sinergismo Farmacológico , Perfilação da Expressão Gênica , Humanos , Ácidos Hidroxâmicos/agonistas , Ácidos Hidroxâmicos/farmacologia , Membranas Intracelulares/metabolismo , Lucantona/agonistas , Lisossomos/genética , Lisossomos/metabolismo , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Permeabilidade/efeitos dos fármacos , Fagossomos/genética , Fagossomos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Esquistossomicidas/agonistas , Proteína Sequestossoma-1 , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Vorinostat
8.
SLAS Discov ; 26(3): 345-351, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33267713

RESUMO

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.


Assuntos
Antivirais/farmacologia , Biologia Computacional/métodos , Reposicionamento de Medicamentos/métodos , Algoritmos , COVID-19 , Linhagem Celular , Mineração de Dados/métodos , Bases de Dados de Produtos Farmacêuticos , Dexametasona/química , Dexametasona/farmacologia , Descoberta de Drogas/métodos , Humanos , Lucantona/farmacologia , SARS-CoV-2/efeitos dos fármacos
9.
J Cell Biol ; 65(2): 418-27, 1975 May.
Artigo em Inglês | MEDLINE | ID: mdl-1092701

RESUMO

The round nucleoli of chick embryo fibroblast cells, when exposed to adenosine (2 mM)or to a number of adenosine analogues, lose material and unravel over a period of several hours to become beaded strands, 20 mu M in length, termed nucleolar necklaces (NN). Light microscope observations on this process are described. Biochemical experiments have revealed that most of these analogues interfere with both messenger RNA synthesis and ribosome synthesis, causing extensive degradation of the preribosome species containing 32S RNA although most of the preribosomes containing 18S RNA survive. We suggest that it is the depletion from the nucleolus of the adhesive 32S and 28S RNA preribosomes which allows the remaining nucleolar apparatus to spread apart into the NN configuration. Also required for the maintenance of the NN structure is the synthesis of some ribosomal RNA (rRNA) possibly present as rRNA "feathers" on the DNA. The addition of inhibitors of rRNA synthesis such as actinomycin D to the NN-containing cells causes loss of rRNA. Then a contraction and collapse of the NN structure into small dense spheres is observed.


Assuntos
Adenosina/análogos & derivados , Nucléolo Celular/ultraestrutura , Fibroblastos/ultraestrutura , Adenosina/farmacologia , Amanitinas/farmacologia , Animais , Arginina/metabolismo , Benzimidazóis/farmacologia , Nucléolo Celular/efeitos dos fármacos , Células Cultivadas , Embrião de Galinha , Cicloeximida/farmacologia , Dactinomicina/farmacologia , Desoxiadenosinas/farmacologia , Fibroblastos/efeitos dos fármacos , Fígado , Lucantona/farmacologia , Microscopia de Contraste de Fase , Músculos , Toiocamicina/farmacologia , Tubercidina/farmacologia
10.
Science ; 186(4164): 647-8, 1974 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-4413726

RESUMO

N-Oxidation at the diethylamino group of hycanthone, of lucanthone, and of two chlorobenzothiopyranoindazoles resulted in a marked reduction in mutagenic activity, while antischistosomal activity was retained or even enhanced. Introduction of chlorine into the 8-position of benzothiopyranoindazoles reduced acute toxicity but had no effect on chemnotherapeutic potency. These dissociations of biological activities indicate that safer antischistosomal compounds of this class can be developed.


Assuntos
Etilenodiaminas , Mutagênicos , Esquistossomicidas , Tioxantenos , Animais , Bioensaio , Etilenodiaminas/toxicidade , Humanos , Dose Letal Mediana , Lucantona/farmacologia , Lucantona/toxicidade , Camundongos , Mutação/efeitos dos fármacos , Ratos , Esquistossomicidas/toxicidade , Relação Estrutura-Atividade , Tioxantenos/toxicidade
11.
Science ; 172(3987): 1058-60, 1971 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-5573958

RESUMO

Rapid spot-test screening of antischistosomal agents reveals that hycanthone is a potent frameshift mutagen while the closely related compound, miracil D, is nonmutagenic in Salmonella. Both hycanthone and miracil D are frameshift mutagens for T4 bacteriophage during growth in Escherichia coli.


Assuntos
Anti-Helmínticos/farmacologia , Lucantona/farmacologia , Mutagênicos/farmacologia , Xantenos/farmacologia , Colífagos/efeitos dos fármacos , Histidina/metabolismo , Mutação , Salmonella/efeitos dos fármacos
12.
Mol Aspects Med ; 28(3-4): 375-95, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17560642

RESUMO

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.


Assuntos
Reparo do DNA , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/metabolismo , Neoplasias , Benzoquinonas/metabolismo , Ácidos Carboxílicos/química , Ácidos Carboxílicos/metabolismo , Dano ao DNA , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/genética , Humanos , Hidroxilaminas/metabolismo , Isoflavonas/metabolismo , Lucantona/metabolismo , Neoplasias/metabolismo , Neoplasias/prevenção & controle , Neoplasias/terapia , Oxirredução , Estresse Oxidativo , Propionatos/metabolismo , Resveratrol , Estilbenos/metabolismo
13.
J Natl Cancer Inst ; 60(5): 1023-8, 1978 May.
Artigo em Inglês | MEDLINE | ID: mdl-642024

RESUMO

A sublethal dose of 100 mg lucanthone hydrochioride/kg (Miracil D, Nilodin; NSC-14574) administered ip into Chinese hamsters [median lethal dose for 30-day survival (LD50/30) of 315 mg/kg] reduced the radiation tolerance of the small intestine and had little or no effect on the radiation tolerance of the bone marrow. Lucanthone hydrochloride was administered at various times before and after whole-body 60Co gamma-irradiation. The median lethal dose for 7-day survival (LD50/7), indicative of death from gastrointestinal epithelial denudation, was reduced from 1,235 rads to minimum values of 995 rads or 985 rads by lucanthone hydrochloride inoculation 10 hours before irradiation or 7.5 hours post irradiation, respectively. The LD50/30, indicative of death from bone marrow stem cell depletion, remained unaltered at approximately 990 rads over the entire treatment scheme, which indicated that the radioresponsiveness of bone marrow stem cells was unaffected by lucanthone hydrochloride. The lucanthone hydrochloride effect was reversible in that control values of LD50/7 were attained by 40 hours post inoculation. Serum concentration of lucanthone hydrochloride in the Chinese hamster, determined spectrophotometrically, reached a peak of 8 microgram/ml by 1.5 hours post inoculation and then decreased exponentially with a half-life of approximately 6 hours, so that by 30 hours post inoculation it was unmeasurable.


Assuntos
Medula Óssea/efeitos dos fármacos , Intestino Delgado/efeitos dos fármacos , Lucantona/farmacologia , Radiossensibilizantes , Animais , Antibióticos Antineoplásicos/farmacologia , Medula Óssea/efeitos da radiação , Células Cultivadas , Cricetinae , Cricetulus , Relação Dose-Resposta a Droga , Relação Dose-Resposta à Radiação , Feminino , Raios gama , Intestino Delgado/efeitos da radiação , Dose Letal Mediana , Lucantona/sangue , Lucantona/toxicidade , Masculino , Fatores de Tempo
14.
Cancer Res ; 38(3): 781-6, 1978 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-626982

RESUMO

Hycanthone, the most potent mutagen in a series of nine thiaxanthenones, is a potent inducer of nuclear immunoreactivity to antinucleoside antibodies in HeLa cells. This response indicates exposure of single-stranded DNA regions. All classes of mutagens thus far tested share this property with hycanthone. Immunoreactivity to antinucleoside antibodies was also induced by brief exposure to hycanthone, 3 microgram/ml, in human fibroblasts from three normal subjects and in fibroblasts from seven patients with DNA repair deficiencies. Unlike those of many other mutagens, the metabolic effects and immunoreactivity induction of hycanthone were readily reversible. No evidence for covalent attachment of [3H]hycanthone to HeLa macromolecules could be found. Induction of DNA repair synthesis could not be detected by autoradiography after exposure of cells to hycanthone. Exposure of single-stranded DNA regions appears to be an important feature of the mechanism of action of hycanthone as a mutagen. Both hycanthone and lucanthone intercalate with DNA, but hycanthone was much less active than was lucanthone in reducing the rapid sedimentation of cell lysate DNA in alkaline sucrose gradients. Similarities and differences, therefore, have been found in the way the potent and the weak mutagen affect DNA of HeLa cells. This may provide clues to understanding the mechanism of mutagenesis by thiaxanthenones and other mutagens.


Assuntos
Células HeLa/efeitos dos fármacos , Hicantone/farmacologia , Mutação/efeitos dos fármacos , Tioxantenos/farmacologia , Anticorpos , Reparo do DNA/efeitos dos fármacos , DNA de Cadeia Simples/metabolismo , Células HeLa/imunologia , Células HeLa/metabolismo , Imunidade/efeitos dos fármacos , Lucantona/farmacologia , Nucleosídeos/imunologia
15.
Cancer Res ; 37(7 Pt 1): 2177-81, 1977 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-861944

RESUMO

Exposure of HeLa cells to lucanthone (3 microgram/ml) caused dissociation of a fast-sedimenting duplex DNA complex, as judged by lysis and sedimentation in alkaline sucrose gradients. The effect of lucanthone on the DNA complex resembled that of actinomycin D and ionizing radiation. Protein synthesis inhibitors such as cycloheximide or inhibitors of DNA synthesis such as hydroxyurea did not lead to dissociation of the complex. Lucanthone was more active than were hycanthone and five other closely related thiaxanthenones tested. Lucanthone promoted X-ray-induced denaturation of DNA in intact cells, as judged by their nuclear immunoreactivity to antinucleoside antibodies. Lucanthone did not inhibit repair of X-ray-induced DNA single-strand breaks.


Assuntos
DNA/metabolismo , Lucantona/farmacologia , Conformação de Ácido Nucleico/efeitos dos fármacos , Desnaturação de Ácido Nucleico/efeitos dos fármacos , Centrifugação com Gradiente de Concentração , DNA/biossíntese , DNA/efeitos da radiação , Reparo do DNA/efeitos dos fármacos , DNA de Cadeia Simples/metabolismo , Dactinomicina/farmacologia , Células HeLa/efeitos dos fármacos , Células HeLa/metabolismo , Biossíntese de Proteínas , RNA/biossíntese
16.
Biochim Biophys Acta ; 454(2): 207-11, 1976 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-999904

RESUMO

The crystal structure of the drug Miracil D has been determined. Although the accuracy of the analysis is limited by disorder, it is apparent that the thioxanthone ring system is planar. The proximal nitrogen atom of the side-chain probably forms an intramolecular hydrogen bond with the carbonyl oxygen. The rest of the side-chain has a large degree of conformational mobility.


Assuntos
Lucantona , Fenômenos Químicos , Química
17.
Cell Stress Chaperones ; 10(1): 37-45, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15832946

RESUMO

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.


Assuntos
Ensaio de Unidades Formadoras de Colônias , Inibidores Enzimáticos/farmacologia , Raios gama/efeitos adversos , Proteínas de Choque Térmico HSP70/metabolismo , Lucantona/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Citoproteção , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/efeitos da radiação , Reparo do DNA , Relação Dose-Resposta à Radiação , Humanos , Leucemia Monocítica Aguda/patologia , RNA Interferente Pequeno/metabolismo
18.
Int J Radiat Oncol Biol Phys ; 8(3-4): 667-70, 1982.
Artigo em Inglês | MEDLINE | ID: mdl-7107396

RESUMO

The interaction of lucanthone and cyclophosphamide (CYC) was investigated in the Chinese hamster in terms of the LD50/7 and LD50/30. These values may be indicative of gastrointestinal stem cell depletion and bone marrow stem cell depletion, respectively. When a nonlethal dose of 100 mg/kg lucanthone preceded CYC injection, the LD50/7 for CYC reached its minimum value of 470 mg/kg at a treatment interval of 10 hours. Lucanthone administered simultaneously with CYC had no effect on the control LD50/7 of 750 mg/kg, and by 48 hours after lucanthone administration the LD50/7 had returned to the control value. When CYC administration preceded that of lucanthone, the LD50/7 reached a minimum of value of 610 mg/kg at an interval of 5 hours; however, for the entire sequence it was approximately 640 mg/kg over all intervals up to 48 hours. The LD50/30 for CYC was only slightly reduced by the presence of lucanthone, indicating that bone marrow sensitivity to CYC was only marginally affected by lucanthone. These data indicate that lucanthone may interact with CYC damage in much the same way as it interacts with radiation damage, viz, by reducing cellular capacity to accumulate and repair sublethal damage.


Assuntos
Ciclofosfamida/toxicidade , Lucantona/farmacologia , Animais , Cricetinae , Cricetulus , Interações Medicamentosas , Dose Letal Mediana , Fatores de Tempo
19.
Int J Radiat Oncol Biol Phys ; 43(1): 89-93, 1999 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-9989518

RESUMO

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.


Assuntos
Barreira Hematoencefálica , Neoplasias Encefálicas/radioterapia , Neoplasias Encefálicas/secundário , Inibidores Enzimáticos/uso terapêutico , Lucantona/uso terapêutico , Inibidores da Topoisomerase II , Animais , Neoplasias Encefálicas/sangue , Inibidores Enzimáticos/sangue , Inibidores Enzimáticos/farmacocinética , Feminino , Humanos , Lucantona/sangue , Lucantona/farmacocinética , Masculino , Camundongos , Camundongos Endogâmicos C3H , Ratos , Ratos Sprague-Dawley
20.
Int J Radiat Oncol Biol Phys ; 37(5): 1133-7, 1997 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-9169823

RESUMO

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.


Assuntos
Dano ao DNA , DNA Super-Helicoidal/efeitos dos fármacos , Lucantona/farmacologia , Inibidores da Topoisomerase I , Inibidores da Topoisomerase II , Antineoplásicos Fitogênicos/farmacologia , Etoposídeo/farmacologia , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA