Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cancer Res ; 62(15): 4439-44, 2002 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-12154052

RESUMO

The cytotoxic effect of the chemotherapeutic drug etoposide (VP-16) is thought to result from its ability to induce DNA damage and thereby to trigger apoptosis. Internucleosomal DNA fragmentation occurs late during apoptosis in many cell types. However, whereas human osteosarcoma cells undergo internucleosomal DNA fragmentation during staurosporine-induced apoptosis, they fail to do so in response to VP-16. Recently, we showed that these cells also do not express the poly(ADP-ribosyl)ation-regulated Ca(2+)- and Mg(2+)-dependent endonuclease DNAS1L3. The possibility that this deficiency underlies the failure of these cells to undergo internucleosomal DNA fragmentation in response to VP-16 was investigated. The proteolytic processing and consequent activation of procaspase-3, cleavage of the inhibitory subunit of DNA fragmentation factor, and the degradation of DNA into 50-kb fragments occurred similarly in osteosarcoma cells exposed to either staurosporine or VP-16. However, the additional processing of the 50-kb DNA fragments to oligonucleosomal fragments was not apparent in the VP-16-treated cells. Ectopic expression of DNAS1L3 conferred on osteosarcoma cells the ability to undergo VP-16-induced internucleosomal DNA fragmentation. Furthermore, expression of DNAS1L3 markedly potentiated the cytotoxic effect of VP-16 in these cells. Both DNAS1L3-mediated and staurosporine-induced internucleosomal DNA fragmentation were Ca(2+) dependent, but only the DNAS1L3-mediated DNA cleavage was blocked by expression of a caspase-3-resistant mutant of poly(ADP-ribose) polymerase-1. The present work results suggest a direct relation between the activity of a chemotherapeutic drug (VP-16) and a specific endonuclease (DNAS1L3). They also indicate that internucleosomal DNA fragmentation plays an active role in apoptosis and that the failure of cancer cells to undergo such DNA degradation may contribute to the development of resistance to chemotherapeutic drugs.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Neoplasias Ósseas/enzimologia , Fragmentação do DNA/efeitos dos fármacos , Endodesoxirribonucleases/fisiologia , Etoposídeo/farmacologia , Osteossarcoma/enzimologia , Poli(ADP-Ribose) Polimerases/fisiologia , Antineoplásicos Fitogênicos/toxicidade , Proteínas Reguladoras de Apoptose , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/patologia , Cálcio/metabolismo , Cálcio/fisiologia , Caspase 3 , Caspases/metabolismo , Fragmentação do DNA/fisiologia , Endodesoxirribonucleases/biossíntese , Endodesoxirribonucleases/genética , Ativação Enzimática , Etoposídeo/toxicidade , Humanos , Nucleossomos/efeitos dos fármacos , Nucleossomos/metabolismo , Osteossarcoma/tratamento farmacológico , Osteossarcoma/patologia , Poli(ADP-Ribose) Polimerases/metabolismo , Proteínas/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Estaurosporina/farmacologia , Transfecção , Células Tumorais Cultivadas
2.
Biochem Biophys Res Commun ; 341(2): 653-62, 2006 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-16427601

RESUMO

A functional relationship between the apoptotic endonuclease DNAS1L3 and the chemotherapeutic drug VP-16 was established. The lymphoma cell line, Daudi, exhibited a significant resistance to VP-16 treatment in comparison to the lymphoma/leukemia cell line, U-937. While U-937 cells degraded their DNA into internucleosomal fragments, Daudi cells failed to undergo such fragmentation in response to the drug. Activation of both caspase-3 and DNA fragmentation factor was not sufficient to trigger internucleosomal DNA fragmentation in Daudi cells. No correlation was found between expression levels of topoisomerase-II, Pgp, Bcl-2, Bax, or Bad and decreased sensitivity of Daudi cells to VP-16. Daudi cells failed to express DNAS1L3 and ectopic expression of this protein significantly sensitized the cells to VP-16. An enhancement of caspase-3 activity and collapse of mitochondrial membrane potential underlie DNAS1L3-mediated sensitization of Daudi cells to VP-16, which may be a direct result of DNAS1L3-mediated increase in PARP-1-activating DNA breaks after VP-16 treatment. Our results suggest that DNAS1L3 plays an active role in lymphoma cell sensitization to VP-16 and that its deficiency may constitute a novel mechanism of drug resistance in these cells.


Assuntos
Antineoplásicos/farmacologia , Apoptose , Endodesoxirribonucleases/biossíntese , Etoposídeo/farmacologia , Linfoma/tratamento farmacológico , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Antineoplásicos Fitogênicos/farmacologia , Proteínas Reguladoras de Apoptose , Cálcio/metabolismo , Caspase 3 , Caspases/metabolismo , Linhagem Celular Tumoral , DNA/metabolismo , Dano ao DNA , Fragmentação do DNA , DNA Topoisomerases Tipo II/metabolismo , Desoxirribonucleases/química , Desoxirribonucleases/metabolismo , Resistencia a Medicamentos Antineoplásicos , Ativação Enzimática , Deleção de Genes , Regulação Neoplásica da Expressão Gênica , Humanos , Immunoblotting , Marcação In Situ das Extremidades Cortadas , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Magnésio/metabolismo , Potenciais da Membrana , Nucleossomos/metabolismo , Proteínas de Ligação a Poli-ADP-Ribose , Proteínas/química , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sensibilidade e Especificidade , Transfecção , Células U937 , Proteína X Associada a bcl-2/metabolismo , Proteína de Morte Celular Associada a bcl/metabolismo
3.
J Cell Physiol ; 207(2): 407-12, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16419035

RESUMO

Platelet-derived growth factor-B (PDGF-B) has been implicated in the pathogenesis of proliferative retinopathies and other scarring disorders in the eye. In this study, we sought to test the therapeutic potential of an aptamer that selectively binds PDGF-B, ARC126, and its PEGylated derivative, ARC127. Both ARC126 and ARC127 blocked PDGF-B-induced proliferation of cultured fibroblasts with an IC50 of 4 nM. Pharmacokinetic studies in rabbits showed similar peak vitreous concentrations of approximately 110 microM after intravitreous injection of 1 mg of either ARC126 or ARC127, but the terminal half-life was longer for ARC127 (98 versus 43 h). Efficacy was tested in rho/PDGF-B transgenic mice that express PDGF-B in photoreceptors and develop severe proliferative retinopathy resulting in retinal detachment. Compared to eyes injected with 20 microg of scrambled aptamer in which five of six developed detachments (three total and two partial), eyes injected with ARC126 (no detachment in five of six and one partial detachment), or ARC127 (no detachment in six of six) had significantly fewer retinal detachments. They also showed a significant reduction in epiretinal membrane formation. These data demonstrate that a single intravitreous injection of an aptamer that specifically binds PDGF-B is able to significantly reduce epiretinal membrane formation and retinal detachment in rho/PDGF-B mice. These striking effects in an aggressive model of proliferative retinopathy suggest that ARC126 and ARC127 should be considered for treatment of diseases in which PDGF-B has been implicated, including ischemic retinopathies such as proliferative diabetic retinopathy, proliferative vitreoretinopathy (PVR), and choroidal neovascularization.


Assuntos
Aptâmeros de Nucleotídeos/farmacologia , Proteínas Proto-Oncogênicas c-sis/genética , Doenças Retinianas/tratamento farmacológico , Células 3T3 , Animais , Aptâmeros de Nucleotídeos/farmacocinética , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Membrana Epirretiniana/tratamento farmacológico , Olho/efeitos dos fármacos , Olho/metabolismo , Olho/patologia , Injeções , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-sis/antagonistas & inibidores , Coelhos , Retina/efeitos dos fármacos , Retina/patologia , Descolamento Retiniano/tratamento farmacológico , Rodopsina/genética
4.
Pharmacol Toxicol ; 90(1): 38-50, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12005112

RESUMO

Acetaminophen is a widely used analgesic and antipyretic drug that exhibits toxicity at high doses to the liver and kidneys. This toxicity has been attributed to cytochrome P-450-generated metabolites which covalently modify target proteins. Recently, acetaminophen, in its unmetabolized form, has been shown to affect a variety of cells and tissues, for instance, testicular and lymphoid tissues and lymphocyte cell lines. The effects on cell viability of acetaminophen at a concentration comparable to that achieved in plasma during acetaminophen toxicity have now been examined with a hepatoma cell line SK-Hep1, primary human peripheral blood lymphocytes and human Jurkat T cells. Acetaminophen reduced cell viability in a time-dependent manner. Staining of cells with annexin-V also revealed that acetaminophen induced, after 8 hr of treatment, a loss of the asymmetry of membrane phospholipids, which is an early event associated with apoptosis. Acetaminophen triggered the release of cytochrome c from mitochondria into the cytosol, activation of caspase-3, 8, and 9, cleavage of poly(ADP-ribose) polymerase, and degradation of lamin B1 and DNA. Whereas cleavage of DNA into internucleosomal fragments was apparent in acetaminophen treated SK-Hep1 and primary lymphocytes, DNA was only degraded to 50-kb fragments in treated Jurkat cells. Overexpression of the antiapoptotic protein Bcl-XL prevented these various apoptotic events induced by acetaminophen in Jurkat cells. Caspase-8 activation was a postmictochondrial event and occurred in a Fas-independent manner. These results demonstrate that acetaminophen induces caspases-dependent apoptosis with mitochondria as a primary target. These results also reiterate the potential role of apoptosis in acetaminophen hepatic and extrahepatic toxicity.


Assuntos
Acetaminofen/farmacologia , Analgésicos não Narcóticos/farmacologia , Apoptose/efeitos dos fármacos , Caspases/metabolismo , Linfócitos/efeitos dos fármacos , Caspases/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Fragmentação do DNA , Ativação Enzimática/efeitos dos fármacos , Humanos , Células Tumorais Cultivadas/efeitos dos fármacos
5.
Biochem Biophys Res Commun ; 290(2): 796-801, 2002 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-11785971

RESUMO

DNA fragmentation factor (DFF) comprises DFF45 and DFF40 subunits, the former of which acts as an inhibitor of the latter (the catalytic subunit) and whose cleavage by caspase-3 results in DFF activation. Disruption of the DFF45 gene blocks the generation of 50-kb DNA fragments and confers resistance to apoptosis. We recently suggested that the early fragmentation of DNA by DFF and the consequent activation of poly(ADP-ribose) polymerase-1 (PARP-1), mitochondrial dysfunction, and activation of caspase-3 contribute to an amplification loop in the apoptotic process. To verify the existence of such a loop, we have now examined the effects of restoring DFF expression in DFF45-deficient fibroblasts. Co-transfection of mouse DFF45(-/-) fibroblasts with plasmids encoding human DFF40 and DFF45 reversed the apoptosis resistance normally observed in these cells. The DFF45(-/-) cells regained the ability to fragment their DNA into 50-kb pieces in response to TNF, which resulted in a marked activation of PARP-1 and a concomitant depletion of intracellular NAD. DFF expression also resulted in an increase both in cytochrome c release into the cytosol and in caspase-3 activation triggered by TNF. These results support the importance of DFF, PARP-1, mitochondria, and caspase-3 in an amplification phase of TNF-induced apoptosis.


Assuntos
Apoptose/fisiologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Proteínas/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Trifosfato de Adenosina/metabolismo , Animais , Proteínas Reguladoras de Apoptose , Caspase 3 , Caspases/metabolismo , Células Cultivadas , Grupo dos Citocromos c/metabolismo , Fragmentação do DNA/efeitos dos fármacos , Fragmentação do DNA/genética , Desoxirribonucleases/genética , Desoxirribonucleases/metabolismo , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Fibroblastos/citologia , Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Camundongos Mutantes , NAD/metabolismo , Proteínas de Ligação a Poli-ADP-Ribose , Subunidades Proteicas , Proteínas/genética , Transfecção
6.
Am J Respir Cell Mol Biol ; 28(3): 322-9, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12594058

RESUMO

Airway inflammation is a central feature of asthma and chronic obstructive pulmonary disease. Reactive oxygen species (ROS) contribute to inflammation by damaging DNA, which, in turn, results in the activation of poly(ADP-ribose) polymerase-1 (PARP-1) and depletion of its substrate, nicotinamide adenine dinucleotide. Here we show that prevention of PARP-1 activation protects against both ROS-induced airway epithelial cell injury in vitro and airway inflammation in vivo. H(2)O(2) induced the generation of ROS, PARP-1 activation and concomitant nicotinamide adenine dinucleotide depletion, and release of lactate dehydrogenase in A549 human airway epithelial cells. These effects were blocked by the PARP-1 inhibitor 3-aminobenzamide (3-AB). Furthermore, 3-AB inhibited both activation of the proinflammatory transcription factor nuclear factor-kappaB and expression of the interleukin-8 gene induced by H(2)O(2) in these cells. In a murine model of allergen-induced asthma, 3-AB prevented airway inflammation elicited by ovalbumin. Moreover, PARP-1 knockout mice were resistant to such ovalbumin-induced inflammation. These protective effects were associated with an inhibition of expression of the inducible nitric oxide synthase. These results implicate PARP-1 activation in airway inflammation, and suggest this enzyme as a potential target for the development of new therapeutic strategies in the treatment of asthma as well as other respiratory disorders such as chronic obstructive pulmonary disease.


Assuntos
Asma/prevenção & controle , Pulmão/patologia , Poli(ADP-Ribose) Polimerases/fisiologia , Alérgenos/administração & dosagem , Alérgenos/imunologia , Animais , Benzamidas/farmacologia , Linhagem Celular , Inibidores Enzimáticos/farmacologia , Expressão Gênica/efeitos dos fármacos , Humanos , Peróxido de Hidrogênio/farmacologia , Inflamação/induzido quimicamente , Inflamação/prevenção & controle , Interleucina-8/metabolismo , L-Lactato Desidrogenase/análise , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/metabolismo , Óxido Nítrico Sintase/metabolismo , Ovalbumina/administração & dosagem , Ovalbumina/imunologia , Oxidantes/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases , Poli(ADP-Ribose) Polimerases/genética , Ativação Transcricional/efeitos dos fármacos
7.
J Biol Chem ; 277(1): 372-8, 2002 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-11694507

RESUMO

Several endonucleases are implicated in the internucleosomal DNA fragmentation associated with apoptosis. The human Ca2+- and Mg2+-dependent endonuclease DNAS1L3 is inhibited by poly(ADP-ribosyl)ation in vitro, and its activation during apoptosis shows a time course similar to that of the cleavage of poly(ADP-ribose) polymerase-1 (PARP-1). The role of the cleavage and consequent inactivation of PARP-1 by caspase-3 in the activation of DNAS1L3 has now been investigated further both in vitro and in vivo. In an in vitro system based on purified recombinant proteins and NAD, caspase-3 prevented the inhibition of DNAS1L3 endonuclease activity by wild-type PARP-1 but not that induced by a caspase-3-resistant PARP-1 mutant. The induction by etoposide of apoptosis in human osteosarcoma cells (which were shown not to express endogenous DNAS1L3) was accompanied by internucleosomal DNA fragmentation only after transfection of the cells with a plasmid encoding DNAS1L3. This DNA fragmentation in etoposide-treated cells was blocked by 1,2-bis(2-aminophenoxy)-ethane-N,N,N',N'-tetraacetic acid, an inhibitor of intracellular Ca2+ release. Expression of the endonuclease subunit of DNA fragmentation factor (DFF40) and cleavage of its inhibitor, DFF45, were not sufficient to cause internucleosomal DNA fragmentation in osteosarcoma cells during etoposide-induced apoptosis. Coexpression of caspase-3-resistant PARP-1 mutant with DNAS1L3 in osteosarcoma cells blocked etoposide-induced internucleosomal DNA fragmentation and resulted in persistent poly(ADP-ribosyl)ation of DNAS1L3; it did not, however, prevent the activation of caspase-3 and the consequent cleavage of endogenous PARP-1. These results indicate that PARP-1 cleavage during apoptosis is not simply required to prevent excessive depletion of NAD and ATP but is also necessary to release DNAS1L3 from poly(ADP-ribosyl)ation-mediated inhibition.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Endodesoxirribonucleases/metabolismo , Etoposídeo/farmacologia , Poli Adenosina Difosfato Ribose/metabolismo , Poli(ADP-Ribose) Polimerases/fisiologia , Caspase 3 , Caspases/fisiologia , Fragmentação do DNA , Ativação Enzimática , Osteossarcoma/metabolismo , Osteossarcoma/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA