Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Pharmacol Res ; 167: 105528, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33667685

RESUMO

Geranylgeranyl diphosphate synthase (GGDPS), an enzyme in the isoprenoid biosynthetic pathway (IBP), produces the isoprenoid (geranylgeranyl pyrophosphate, GGPP) used in protein geranylgeranylation reactions. Our prior studies utilizing triazole bisphosphonate-based GGDPS inhibitors (GGSIs) have revealed that these agents represent a novel strategy by which to induce cancer cell death, including multiple myeloma and pancreatic cancer. Statins inhibit the rate-limiting enzyme in the IBP and potentiate the effects of GGSIs in vitro. The in vivo effects of combination therapy with statins and GGSIs have not been determined. Here we evaluated the effects of combining VSW1198, a novel GGSI, with a statin (lovastatin or pravastatin) in CD-1 mice. Twice-weekly dosing with VSW1198 at the previously established maximally tolerated dose in combination with a statin led to hepatotoxicity, while once-weekly VSW1198-based combinations were feasible. No abnormalities in kidney, spleen, brain or skeletal muscle were observed with combination therapy. Combination therapy disrupted protein geranylgeranylation in vivo. Evaluation of hepatic isoprenoid levels revealed decreased GGPP levels in the single drug groups and undetectable GGPP levels in the combination groups. Additional studies with combinations using 50% dose-reductions of either VSW1198 or lovastatin revealed minimal hepatotoxicity with expected on-target effects of diminished GGPP levels and disruption of protein geranylgeranylation. Combination statin/GGSI therapy significantly slowed tumor growth in a myeloma xenograft model. Collectively, these studies are the first to demonstrate that combination IBP inhibitor therapy alters isoprenoid levels and disrupts protein geranylgeranylation in vivo as well as slows tumor growth in a myeloma xenograft model, thus providing the framework for future clinical exploration.


Assuntos
Vias Biossintéticas/efeitos dos fármacos , Diterpenos/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Inibidores de Hidroximetilglutaril-CoA Redutases/administração & dosagem , Prenilação de Proteína/efeitos dos fármacos , Terpenos/metabolismo , Triazóis/administração & dosagem , Animais , Vias Biossintéticas/fisiologia , Linhagem Celular Tumoral , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/patologia , Diterpenos/toxicidade , Avaliação Pré-Clínica de Medicamentos/métodos , Quimioterapia Combinada , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/toxicidade , Farnesiltranstransferase/antagonistas & inibidores , Farnesiltranstransferase/metabolismo , Feminino , Inibidores de Hidroximetilglutaril-CoA Redutases/toxicidade , Lovastatina/administração & dosagem , Lovastatina/toxicidade , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Pravastatina/administração & dosagem , Pravastatina/toxicidade , Prenilação de Proteína/fisiologia , Terpenos/antagonistas & inibidores , Triazóis/toxicidade , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
2.
J Biochem Mol Toxicol ; 27(6): 287-94, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23761184

RESUMO

Statins are potent drugs, used as lipid-lowering agents in cardiovascular diseases. Hepatotoxicity is one of the serious adverse effects of statins, and the exact mechanism of hepatotoxicity is not yet clear. In this study, the cytotoxic effects of the most commonly used statins, that is, atorvastatin, lovastatin, and simvastatin toward isolated rat hepatocytes, were evaluated. Markers, such as cell death, reactive oxygen species (ROS) formation, lipid peroxidation, mitochondrial membrane potential, and the amount of reduced and oxidized glutathione in the statin-treated hepatocytes, were investigated. It was found that the statins caused cytotoxicity toward rat hepatocytes dose dependently. An elevation in ROS formation, accompanied by a significant amount of lipid peroxidation and mitochondrial depolarization, was observed. Cellular glutathione reservoirs were decreased, and a significant amount of oxidized glutathione was formed. This study suggests that the adverse effect of statins toward hepatocytes is mediated through oxidative stress and the hepatocytes mitochondria play an important role in the statin-induced toxicity.


Assuntos
Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Inibidores de Hidroximetilglutaril-CoA Redutases/toxicidade , Animais , Atorvastatina , Morte Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Glutationa/metabolismo , Dissulfeto de Glutationa/metabolismo , Hepatócitos/patologia , Ácidos Heptanoicos/toxicidade , Peroxidação de Lipídeos/efeitos dos fármacos , Lovastatina/toxicidade , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Pirróis/toxicidade , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Sinvastatina/toxicidade
3.
Eur Rev Med Pharmacol Sci ; 15(9): 985-91, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22013720

RESUMO

OBJECTIVE: Evidences from cell culture experiments suggest a link between cholesterol and nervous system disease. Statins may have neurotoxic or neuroprotective effects, but these effects remain controversial. Therefore, the present study was aimed to investigate the possible toxicity of statins on a neurite outgrowth in mouse neuroblastoma NB2a cell line. MATERIALS AND METHODS: We have utilized d-cAMP-induced terminally differentiated NB2a cells in culture as an experimental model to study the effects of statins. The cell survival and proliferation were studied by MTT. Measurement of neurite outgrowth was done by neurotoxicity screening test. NB2a cell differentiation was achieved by serum free medium plus 0.5 mM dibutyryl cAMP. Cells were incubated for 24 hours at 37 degrees C. After this period, lovastatin, mevastatin and atorvastatin were added to wells at different concentrations (1, 3, 10, 100 microM). Approximately 100 cells were chosen for each sample and examined randomly 24 hours later, from 10 different fields. Total length of neurite was photographed microscopically and measured by image analyze software. Changes in neurite lengths were expressed as % inhibition compared to that of the control group. RESULTS: Results showed that three statins at high concentrations induced neurite inhibition, inhibited proliferation and reduced the viability of differentiated neuroblastoma NB2a cells. CONCLUSIONS: Our results suggest that statins could act as a neurotoxic agent at high doses depending upon their concentrations. These results require further investigation at ultra structural and molecular levels to understand long term side effects for clinical safety of statins.


Assuntos
Inibidores de Hidroximetilglutaril-CoA Redutases/toxicidade , Neuritos/efeitos dos fármacos , Neuroblastoma/patologia , Animais , Atorvastatina , Diferenciação Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ácidos Heptanoicos/toxicidade , Lovastatina/análogos & derivados , Lovastatina/toxicidade , Camundongos , Neuritos/patologia , Pirróis/toxicidade
4.
Cancer Lett ; 288(1): 57-67, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19632769

RESUMO

The aim of this study was to investigate the potential cytostatic/cytotoxic effects of HMG-CoA reductase inhibitors and two orphan drugs registered for the treatment of advanced renal cell carcinoma, i.e. sorafenib and sunitinib against several different tumor cell lines. Cytostatic/cytotoxic effects were measured using crystal violet or MTT reduction assays. Cell cycle regulation was investigated using flow cytometry and Western blotting. The combination of lovastatin and sorafenib (but not sunitinib) produced synergistic cytostatic/cytotoxic effects against all tested tumor cell lines. In this study, an impairment of the protein prenylation, especially geranylgeranylation, resulted predominantly in the potentiation of the cytostatic/cytotoxic activity of sorafenib, in cell cycle arrest in G1 phase, and, in poor induction of apoptosis. Moreover, due to the fact that it has been well documented that sorafenib compromises the heart function, we studied the interaction of lovastatin and sorafenib using rat cardiomyoblast line H9c2. The combination showed strong synergistic cardiotoxic effects. Statins and tyrosine kinase inhibitors were used at doses that are achievable clinically, which makes the combination promising for future studies, especially in urooncology, bearing in mind possible cardiotoxic effects.


Assuntos
Benzenossulfonatos/farmacologia , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Indóis/farmacologia , Lovastatina/farmacologia , Neoplasias/patologia , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Pirróis/farmacologia , Animais , Apoptose/efeitos dos fármacos , Benzenossulfonatos/toxicidade , Western Blotting , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Citometria de Fluxo , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/toxicidade , Indóis/toxicidade , Lovastatina/toxicidade , Camundongos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Niacinamida/análogos & derivados , Compostos de Fenilureia , Inibidores de Proteínas Quinases/toxicidade , Prenilação de Proteína , Piridinas/toxicidade , Pirróis/toxicidade , Ratos , Sorafenibe , Sunitinibe
5.
Br J Pharmacol ; 158(7): 1777-86, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19888963

RESUMO

BACKGROUND AND PURPOSE: Statins, inhibitors of hydroxymethylglutaryl-CoA reductase, reduce the intracellular synthesis of cholesterol and prevent the onset of atherosclerosis. They also decrease the synthesis of isoprenoid molecules, such as the side chain of ubiquinone and geranylgeranyl pyrophosphate. As a consequence, statins impair mitochondrial metabolism and the activation of small monomeric GTPases (such as Rho and Ras), causing toxic effects. To date, a successful strategy to prevent statin toxicity is lacking. EXPERIMENTAL APPROACH: In human monocytic THP-1 cells, we measured the synthesis of cholesterol and isoprenoids, mitochondrial electron flow, the activity of RhoA and Rac, cell death and proliferation. KEY RESULTS: Mevastatin reduced the synthesis of cholesterol, geranylgeranyl pyrophosphate and ubiquinone, mitochondrial electron transport, activity of RhoA and Rac, and cell proliferation, accompanied by increased cell death. Geranylgeraniol, a cell-permeable analogue of geranylgeranyl pyrophosphate, reversed all these effects of mevastatin, without affecting its ability to reduce cholesterol synthesis. Notably, geranylgeraniol was more effective than the addition of exogenous ubiquinone, which rescued mitochondrial respiratory activity and reversed mevastatin cytotoxicity, but did not alter the decrease in cell proliferation. The same results were obtained in human liver HepG2 cells. CONCLUSIONS AND IMPLICATIONS: Geranylgeraniol had a broader protective effect against the cytotoxicity of statins than exogenous ubiquinone. Therefore, geranylgeraniol may be a more useful and practical means of limiting the toxicities of statins, without reducing their efficacy as cholesterol lowering agents.


Assuntos
Colesterol/biossíntese , Diterpenos/farmacologia , Inibidores de Hidroximetilglutaril-CoA Redutases/toxicidade , Lovastatina/análogos & derivados , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Transporte de Elétrons/efeitos dos fármacos , Células Hep G2 , Humanos , Lovastatina/toxicidade , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Terpenos/metabolismo , Ubiquinona/farmacologia
6.
Drug Chem Toxicol ; 32(1): 17-20, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19514934

RESUMO

The 3-hydroxy-3-methylglutaryl (HMG)-coenzyme A reductase inhibitor, lovastatin (lova), has been reported to both sensitize to, and protect against, the toxic effects of the antitumor anthracycline doxorubicin (dox) in cellular and in vivo systems. The mechanism by which these effects occur has not yet been determined. In the present study, lova is shown to enhance the genotoxicity of dox in the V79 cell in vitro micronucleus assay and to do so, most likely, via noncovalent interaction with DNA adjacent to sites of dox binding. These studies confirm and extend the experimental evidence strongly suggesting the importance of noncovalent drug/DNA interactions in cellular responses to genotoxic stimuli.


Assuntos
Antibióticos Antineoplásicos/toxicidade , Doxorrubicina/toxicidade , Lovastatina/toxicidade , Mutagênicos/toxicidade , Animais , Linhagem Celular , Cricetinae , Cricetulus , DNA/efeitos dos fármacos , DNA/metabolismo , Sinergismo Farmacológico , Inibidores de Hidroximetilglutaril-CoA Redutases/toxicidade , Testes para Micronúcleos
7.
Toxicol Appl Pharmacol ; 193(3): 346-55, 2003 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-14678744

RESUMO

The objective of this study was to evaluate the cardiac toxicity of the HMG-CoA reductase inhibitors by testing the hypothesis that lovastatin induces apoptotic and/or oncotic cell death in the myocyte element of the heart and further that cell death is mediated through interruption of the mevalonate pathway and that apoptosis is induced through activation of caspase-2 and caspase-3. Cardiomyocytes were cultured from embryonic chick heart. Lovastatin-induced apoptosis in these cells was demonstrated by three independent techniques, namely (1) FACS analysis of low DNA content by propidium iodide (PI); (2) microscopic assessment for cellular changes of apoptosis; and (3) FACS analysis of cells stained with PI and fluorescein diacetate. Lovastatin produced a concentration-dependent increase in apoptotic cell death and 100 microM lovastatin showed over a 4-fold increase in apoptosis compared to control. Lovastatin also induced oncotic cell death, as there was a 2.5-fold increase in the amount of oncotic cell death compared to control. Lovastatin-induced apoptosis operated, in part, through the mevalonate pathway. The caspase-2 inhibitor z-VDVAD-fmk and the caspase-3 inhibitor Ac-DEVD-CHO reduced the extent of lovastatin-induced cardiac apoptosis. In contrast, lovastatin-induced oncosis was not only insensitive to these caspase-2 or -3 inhibitors but occurred through a mevalonate-independent mechanism of action. In summary, lovastatin-induced cardiotoxicity is complex and represents the sum of two distinct modes of cell death operating in part through the mevalonate pathway with the apoptotic component subject to modification by inhibitors of the initiator caspase, caspase-2, as well as the effector caspase, caspase-3.


Assuntos
Apoptose/efeitos dos fármacos , Inibidores de Caspase , Inibidores de Hidroximetilglutaril-CoA Redutases/toxicidade , Lovastatina/toxicidade , Miócitos Cardíacos/efeitos dos fármacos , Animais , Caspase 2 , Caspase 3 , Células Cultivadas , Embrião de Galinha , Inibidores de Cisteína Proteinase/farmacologia , DNA/metabolismo , Citometria de Fluxo , Técnicas In Vitro , Microscopia de Fluorescência , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/metabolismo , Propídio
8.
Leuk Res ; 25(8): 651-60, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11397469

RESUMO

Cytosine arabinoside (ara-C) is widely used for the treatment of leukemias and displays significant toxicities. Lovastatin, an HMG-CoA reductase inhibitor, is extensively used to treat hypercholesterolemia. To determine whether lovastatin could augment ara-C's activity we have examined their effects in the human erythroleukemia K562 cell line and the ara-C resistant ARAC8D cell line. A synergistic interaction between the two drugs was found. We have demonstrated that the interaction does not occur at the level of RAS but may involve lovastatin's effect of downregulating MAPK activity and preventing ara-C-induced MAPK activation. These studies represent the first description of a potentially beneficial interaction between lovastatin and ara-C that could be applied to the treatment of human leukemia.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Citarabina/farmacologia , Células K562/efeitos dos fármacos , Lovastatina/farmacologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/toxicidade , Apoptose/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Citarabina/administração & dosagem , Citarabina/toxicidade , Sinergismo Farmacológico , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/administração & dosagem , Inibidores de Hidroximetilglutaril-CoA Redutases/toxicidade , Células K562/patologia , Lovastatina/administração & dosagem , Lovastatina/toxicidade , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas ras/fisiologia
9.
Int J Biochem Cell Biol ; 32(9): 957-65, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11084375

RESUMO

Lovastatin and simvastatin are HMG-CoA reductase inhibitors widely used as antihyperlipidemic drugs, which also display antiproliferative properties. In the present paper, we provide evidence that both lovastatin and simvastatin are modulators of the purified bovine pituitary 20 S proteasome, since they mildly stimulate the chymotrypsin-like activity and inhibit the peptidylglutamylpeptide hydrolyzing activity without interfering with the trypsin-like activity. However, those effects are only observed when the closed ring forms of the drugs are used, while the opened ring form of lovastatin acts as a mild inhibitor of the chymotrypsin like activity. The closed ring form of lovastatin is much more potent as a cytotoxic agent on the Colon-26 (C-26) colon carcinoma cell line than the opened ring form, which is only mildly cytostatic. Moreover, neither the cytotoxic effects nor the effects on 20 S proteasome activities are prevented by mevalonate, which by itself inhibits the trypsin-like activity of the proteasome. Neither the opened ring nor the closed ring form of lovastatin induces an accumulation of ubiquitin-protein conjugates, which is observed after treatment with lactacystin, a selective proteasome inhibitor. In contrast with the opened ring form of lovastatin, the closed ring form induces the disappearance of detectable p27(kip1) from C-26 cells. Altogether, our results indicate that the closed ring form of lovastatin induces cytotoxic effects independent of its HMG-CoA inhibiting activity, however, those effects are mediated by a complex modulation of proteasome activity rather than by inhibition of the 20 S proteasome.


Assuntos
Cisteína Endopeptidases/metabolismo , Lovastatina/farmacologia , Complexos Multienzimáticos/metabolismo , Sinvastatina/farmacologia , Animais , Antineoplásicos/toxicidade , Western Blotting , Inibidores do Crescimento/toxicidade , Lovastatina/toxicidade , Camundongos , Complexo de Endopeptidases do Proteassoma , Sinvastatina/toxicidade , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/enzimologia
10.
J Neurooncol ; 42(1): 1-11, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10360474

RESUMO

Medulloblastoma is a malignant paediatric central nervous system tumor with a poor prognosis, stimulating the evaluation of improved treatment strategies. Lovastatin, a competitive inhibitor of 3-hydroxy-3-methylglutaryl coenzyme A reductase, is currently used to treat patients with hypercholesterolemia. This compound also inhibits the production of non-steroidal mevalonate derivatives that are implicated in the control of cellular proliferation, and can induce cell-cycle arrest in vitro. We recently showed that lovastatin inhibited growth and promoted apoptosis of neuroblastoma, the peripheral nervous system 'cousin' of medulloblastoma. Therefore the potential of lovastatin as a possible anticancer drug against medulloblastoma was evaluated in vitro. Four medulloblastoma cell lines, Daoy, UW228, D341 Med and D283 Med, were treated with 1-40 microM of lovastatin in vitro. Analysis of cell morphologic changes, cell viability, DNA fragmentation and flow cytometry in all four cell lines showed growth inhibition and induction of apoptosis with lovastatin treatment. As little as 10 microM of lovastatin was sufficient to cause a marked reduction in cell numbers, and more than 20 microM of lovastatin induced >90% cells to undergo apoptosis, after intervals ranging between 36 and 96 h, depending on the cell line. Lovastatin induced apoptosis in these cell lines was concomitant with cell cycle arrest in G1. The attached cell lines UW228 and Daoy were more sensitive to lovastatin than D283 Med and D341 Med. Daoy cells which survived several cycles of lovastatin treatment could still be induced to undergo apoptosis after longer treatment times. The efficient induction of apoptosis by lovastatin favours this drug as a potential new avenue of therapeutic intervention for medulloablastoma.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias Cerebelares/patologia , Lovastatina/toxicidade , Meduloblastoma/patologia , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Criança , Citometria de Fluxo , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/toxicidade , Células Tumorais Cultivadas
11.
Int J Cancer ; 81(4): 560-7, 1999 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-10225445

RESUMO

Lovastatin, a drug commonly used in the treatment of hypercholesterolemia, has previously been reported to exert potentiated antitumor activity when combined with either tumor necrosis factor-alpha (TNF-alpha), cisplatin or doxorubicin in a melanoma model in mice. Since lovastatin interferes with the function of ras oncogene-encoded (Ras) proteins, we have investigated the antitumor activity of lovastatin and TNF-alpha using a Ha-ras-transformed murine tumor model. In in vitro studies, lovastatin inhibited the growth of cells transformed with Ha-ras oncogene (Ras-3T3 and HBL100-ras cells) more effectively than control NIH-3T3 and HBL100-neo cells. In in vivo experiments, the Ras-3T3 tumor demonstrated significantly increased sensitivity to combined treatment with both lovastatin (50 mg/kg) and TNF-alpha (1 microg/day) compared with either agent alone. Combined treatment with both agents also resulted in greater inhibition of blood-vessel formation. Ras-3T3 tumor cells produced increased amounts of vascular endothelial growth factor (VEGF) and lovastatin effectively suppressed VEGF production by these cells. Our results suggest that lovastatin increases antitumor activity of TNF-alpha against tumor cells transformed with v-Ha-ras oncogene via inhibition of tumor-induced blood-vessel formation.


Assuntos
Antineoplásicos/uso terapêutico , Transformação Celular Neoplásica , Genes ras , Lovastatina/toxicidade , Lovastatina/uso terapêutico , Neoplasias Experimentais/tratamento farmacológico , Neovascularização Patológica/prevenção & controle , Fator de Necrose Tumoral alfa/toxicidade , Fator de Necrose Tumoral alfa/uso terapêutico , Células 3T3 , Animais , Antineoplásicos/toxicidade , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Fatores de Crescimento Endotelial/biossíntese , Fatores de Crescimento Endotelial/genética , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Linfocinas/biossíntese , Linfocinas/genética , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias Experimentais/irrigação sanguínea , Neoplasias Experimentais/genética , Neovascularização Patológica/patologia , Transfecção , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
12.
Antibiot Khimioter ; 44(2): 13-8, 1999.
Artigo em Russo | MEDLINE | ID: mdl-10202552

RESUMO

It was shown in vitro that high concentrations of lovastatin, a competitive inhibitor of hydroxymethyl glutaryl coenzyme A (HMG-CoA) reductase inhibited human malignant cells MOLT-4. The activity of lovastatin in doses of 50-250 microM was dose-dependent. Addition of mevalonate in a concentration of 3 mM to the growth medium completely prevented the cytotoxic effect of 100 microM of lovastatin. At the same time, exogenous mevalonate did not decrease the cytotoxicity of the anthracycline antibiotic carminomycin. Moreover, in a high concentration (7 mM) mevalonate slowly but significantly inhibited the growth of the malignant target cells and the effect was added to the cytotoxic effect of carminomycin low concentrations (0.08 to 0.175 microgram/ml). The results and the literature data suggested that combination of mevalonate, HMG-CoA reductase inhibitors and anthracyclines could be useful in tumor chemotherapy. The suggestion needs further investigation.


Assuntos
Antibióticos Antineoplásicos/toxicidade , Antineoplásicos/toxicidade , Carrubicina/toxicidade , Lovastatina/toxicidade , Ácido Mevalônico/toxicidade , Linfócitos T/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Depressão Química , Relação Dose-Resposta a Droga , Interações Medicamentosas , Humanos , Células Tumorais Cultivadas/efeitos dos fármacos
13.
Biochem Mol Biol Int ; 38(2): 345-55, 1996 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-8850530

RESUMO

Selenocysteine tRNA[Ser]Sec isoacceptors contain the modified nucleotide i6A immediately 3' to the anticodon. Because synthesis of i6A is expected to be inhibited by lovastatin, the status of tRNA[Ser]Sec isoacceptors was examined in human breast carcinoma cells. As part of the initial characterization of these cells, it was determined that an adriamycin resistant derivative of the MCF-7 cell line exhibited a dramatic increase in the sensitivity to the killing effects of lovastatin relative to the parental MCF-7 cells. When MCF-7Adr cells were incubated with high levels of lovastatin, there was a dramatic perturbation in the distribution of isoacceptors within the selenocysteine tRNA population. Lovastatin may therefore be a useful reagent for both the study of differential killing of drug-resistant tumor cells and selenoprotein biosynthesis.


Assuntos
Neoplasias da Mama/química , Carcinoma/química , Lovastatina/toxicidade , RNA de Transferência Aminoácido-Específico/análise , Antibióticos Antineoplásicos/farmacologia , Doxorrubicina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Glutationa Peroxidase/metabolismo , Humanos , Selênio/metabolismo , Células Tumorais Cultivadas
14.
Jpn J Cancer Res ; 85(6): 633-8, 1994 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8063617

RESUMO

Simvastatin (SV), a 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor, inhibits the synthesis of mevalonic acid. The dose-dependent (0.1-100 micrograms/ml) cytotoxicity of SV towards human (MIAPaCa-2, Panc-1, HPC-1, HPC-3, HPC-4, PK-1, PK-9) and hamster (T2) pancreatic carcinoma cell lines was determined by MTT assay. At up to 20 micrograms/ml of SV, the effect was reversible and was restored by 60 micrograms/ml mevalonic acid. Point mutation of Ki-ras at codon 12 in each cell line was detected by means of the modified polymerase chain reaction. The concentration of SV necessary to achieve 50% cytotoxicity was about 10 micrograms/ml, and at this concentration of SV, DNA synthesis assayed in terms of [3H]thymidine uptake, isoprenylation of p21ras examined by Western blotting and cell progression from G1 to S phase of the cell cycle analyzed by flow cytometry were all inhibited. Isoprenylation inhibitors of p21ras, such as SV, are expected to be useful for the treatment of pancreatic cancer.


Assuntos
Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/genética , Genes ras , Lovastatina/análogos & derivados , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Mutação Puntual , Adenocarcinoma/metabolismo , Animais , Sequência de Bases , Ciclo Celular/fisiologia , Códon , Cricetinae , DNA de Neoplasias/biossíntese , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases , Lovastatina/toxicidade , Dados de Sequência Molecular , Neoplasias Pancreáticas/metabolismo , Prenilação de Proteína/efeitos dos fármacos , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Sensibilidade e Especificidade , Sinvastatina , Células Tumorais Cultivadas/efeitos dos fármacos
15.
Mol Aspects Med ; 15 Suppl: s195-206, 1994.
Artigo em Inglês | MEDLINE | ID: mdl-7752831

RESUMO

Alcohol metabolism may result in oxidant stress and free radical injury through a variety of mechanisms. Lovastatin may also produce oxidant stress by reducing levels of an endogenous antioxidant, coenzyme Q (CoQ). The separate and combined effects of ethanol, 20 EN% in a total liquid diet, and lovastatin, 67 mg/kg diet, on alpha-tocopherol, retinol palmitate, CoQ9 and thiobarbituric acid reactive (TBAR) material in liver from rats were determined. The effect of exogenous CoQ10 on these treatment groups was also determined. Food consumption, weight gain, liver lipid and TBAR material were similar between treatment groups. Compared to control animals, ethanol reduced retinol palmitate significantly, from 143 to 90 micrograms/g wet weight. Lovastatin had no effect on retinal palmitate nor did it act additively with ethanol. Ethanol decreased liver alpha-tocopherol from 28 to 12 micrograms/g wet weight and lovastatin diminished it to 12 micrograms; no additive effect was evident. Ethanol had no effect, but lovastatin decreased CoQ9 from 83 to 55 micrograms/g wet weight. Supplementation with CoQ10 did not modulate the effect of ethanol on retinal palmitate, but it did reverse the effect of lovastatin on CoQ9. Supplementary CoQ10 did not alter control levels of alpha-tocopherol, but it appeared to reverse most of the decrease in alpha-tocopherol attributable to ethanol or lovastatin separately. It only partially reversed the effect of ethanol and lovastatin combined on alpha-tocopherol, however. As expected, lovastatin had no effect on CoQ10 levels in supplemented animals. Ethanol, either separately or in combination with lovastatin, diminished liver stores of CoQ10 by almost 40%. We conclude that 20 EN% ethanol given in a liquid diet for 5 weeks is sufficient to lower retinol palmitate and that lovastatin reduces CoQ9. Both diminish alpha-tocopherol, an effect largely overcome by CoQ10 supplementation with either drug alone, but not with the combination. Since many individuals chronically consume the levels of ethanol represented by this experiment, and since a certain number of those also take lovastatin, further research into the possible clinical significance of these observations is warranted.


Assuntos
Antioxidantes/análise , Etanol/farmacologia , Fígado/efeitos dos fármacos , Lovastatina/farmacologia , Substâncias Reativas com Ácido Tiobarbitúrico/análise , Ubiquinona/análogos & derivados , Animais , Peso Corporal/efeitos dos fármacos , Coenzimas , Dieta , Diterpenos , Avaliação Pré-Clínica de Medicamentos , Etanol/toxicidade , Fígado/química , Hepatopatias Alcoólicas/prevenção & controle , Lovastatina/toxicidade , Tamanho do Órgão/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Ésteres de Retinil , Ubiquinona/análise , Ubiquinona/farmacologia , Vitamina A/análogos & derivados , Vitamina A/análise , Vitamina E/análise
16.
Am J Cardiol ; 62(15): 16J-27J, 1988 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-3055920

RESUMO

Administration of lovastatin to animals at high dosage levels produces a broad spectrum of toxicity. This toxicity is expected based on the critical nature of the target enzyme (HMG CoA reductase) and the magnitude of the dosage levels used. The information reviewed in this paper demonstrates that these adverse findings in animals do not predict significant risk in humans. The reason for this derives from the fact that all the available evidence suggests that the adverse effects observed are produced by an exaggeration of the desired biochemical effect of the drug at high dosage levels. The presence of clear and high no-effect doses for these toxic effects along with the fact that most of the changes observed are clearly mechanism-based (directly attributable to inhibition of mevalonate synthesis) indicate that it is unlikely that similar changes will be observed at the therapeutic dosage levels in humans. This hypothesis is supported by the extensive human safety experience described by Tobert in the following report.


Assuntos
Inibidores de Hidroximetilglutaril-CoA Redutases , Lovastatina/toxicidade , Animais , Catarata/induzido quimicamente , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Fígado/efeitos dos fármacos , Lovastatina/administração & dosagem , Neoplasias Experimentais/induzido quimicamente , Fatores de Risco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA