Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Death Differ ; 8(4): 403-10, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11550092

RESUMO

APRIL (a proliferation-inducing ligand) is a newly identified member of the tumor necrosis factor (TNF) family. Tumor growth-promoting as well as apoptosis-inducing effects of APRIL have been described. Here, we report that five of 12 human malignant glioma cell lines express APRIL. APRIL gene transfer experiments revealed that malignant glioma cells are refractory to growth-promoting activity of APRIL in vitro and in vivo. Interestingly, ectopic expression of APRIL confers minor protection from apoptotic cell death induced by the death ligands, CD95 ligand (CD95L) and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)/Apo2 ligand (Apo2L). This antiapoptotic activity is specific for death ligand/receptor-mediated apoptosis since APRIL does not protect glioma cells from the cytotoxicity of the drugs, teniposide, vincristine, lomustine or cisplatin. Ectopic expression of APRIL is associated with the upregulation of X-linked inhibitor of apoptosis protein (XIAP), providing a possible explanation for the antiapoptotic activity observed here. In contrast, APRIL does not regulate the expression levels of the antiapoptotic proteins FLICE-inhibitory protein (FLIP), Bcl-2 or Bcl-X(L). These findings suggest that APRIL is involved in the regulation of death ligand-induced apoptotic signaling in malignant glioma cells.


Assuntos
Apoptose , Glioma/patologia , Glicoproteínas de Membrana/farmacologia , Proteínas de Membrana/fisiologia , Proteínas , Fator de Necrose Tumoral alfa/farmacologia , Fator de Necrose Tumoral alfa/fisiologia , Receptor fas/farmacologia , Animais , Proteínas Reguladoras de Apoptose , Divisão Celular , Glioma/genética , Glioma/metabolismo , Humanos , Proteínas de Membrana/genética , Camundongos , Camundongos Nus , Biossíntese de Proteínas , RNA Neoplásico/biossíntese , Ligante Indutor de Apoptose Relacionado a TNF , Transfecção , Células Tumorais Cultivadas , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral , Fator de Necrose Tumoral alfa/genética , Regulação para Cima , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X
2.
Cell Death Differ ; 8(6): 595-602, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11536010

RESUMO

Most human malignant glioma cell lines are susceptible to CD95 ligand (CD95L)-induced apoptosis. Here, we report that glioma cells are also susceptible to the cytotoxic effects of exogenous C2-ceramide. This form of cell death exhibits some morphological features of apoptosis as assessed by electron microscopy, but is unaffected by the broad spectrum caspase inhibitor, zVAD-fmk. Further, CD95L-induced apoptosis is synergistically enhanced by coexposure of the glioma cells to CD95L and C2-ceramide. CD95L-induced caspase 3-like activity, cytochrome c release and cleavage of caspases 3, 8, 9 and poly(ADP-ribose)polymerase (PARP) increase substantially after cotreatment with CD95L and C2-ceramide compared with CD95L treatment alone. None of these events occur in response to cytotoxic concentrations of C2-ceramide alone. C2-ceramide does not alter CD95 expression. Gene transfer-mediated enhancement of CD95 expression results not only in increased susceptibility to CD95L, but also in increased sensitivity to C2-ceramide. We conclude that (i) synergistic induction of apoptosis by C2-ceramide and CD95L depend on a cross-talk between the two signal transduction pathways and that (ii) C2-ceramide, independently of its sensitizing effects on CD95-dependent caspase activation, is also capable of triggering an apoptotic signaling cascade that is unaffected by zVAD-fmk-mediated caspase inhibition, but promoted by high levels of CD95 expression.


Assuntos
Apoptose/efeitos dos fármacos , Caspases/metabolismo , Neuroglia/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Esfingosina/farmacologia , Receptor fas/metabolismo , Clorometilcetonas de Aminoácidos/farmacologia , Inibidores de Caspase , Grupo dos Citocromos c/metabolismo , Sinergismo Farmacológico , Ativação Enzimática/efeitos dos fármacos , Proteína Ligante Fas , Humanos , Immunoblotting , Glicoproteínas de Membrana/metabolismo , Microscopia Eletrônica , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Neuroglia/patologia , Neuroglia/ultraestrutura , Poli(ADP-Ribose) Polimerases/metabolismo , Esfingosina/análogos & derivados , Células Tumorais Cultivadas
3.
Oncogene ; 20(35): 4757-67, 2001 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-11521188

RESUMO

Human glioma cell lines differ in their requirement for the inhibition of protein synthesis to activate the CD95-dependent killing pathway. CD95 ligand (CD95L) induced mitochondrial cytochrome c release and processing of caspases 3, 7, 8 and 9 in LN-18 cells in the absence of an inhibitor of protein synthesis, cycloheximide (CHX). These biochemical changes were observed in LN-229 cells only in the presence of CHX. The viral caspase inhibitor, cytokine response modifier (crm)-A, inhibited mitochondrial cytochrome c release, caspase processing and cell death under all conditions. Ectopic expression of BCL-X(L) prevented processing of caspase 8 in LN-18 cells but not in LN-229 cells. Thus, caspase 8 activation is amplified through the release of cytochrome c in LN-18 cells but occurs mainly at the receptor in LN-229 cells. In contrast to BCL-2, BCL-X(L), X-linked inhibitor-of-apoptosis protein (XIAP) and FLICE-inhibitory protein (FLIP), the levels of the cyclin-dependent kinase (CDK) inhibitor, p21Waf/Cip1, rapidly decreased in response to CHX. P21 antisense oligonucleotides promoted caspase activation and mitochondrial cytochrome c release and induced strong sensitization to CD95-mediated apoptosis. These data place potentiating effects of CHX (i) to the activation of caspase 8 at the receptor in LN-229 cells as well as (ii) to a down-stream target at least in LN-18 cells, but probably both cell lines, that may be identical with p21Waf/Cip1.


Assuntos
Apoptose , Ciclinas/fisiologia , Cicloeximida/farmacologia , Glioma/patologia , Peptídeos e Proteínas de Sinalização Intracelular , Inibidores da Síntese de Proteínas/farmacologia , Receptor fas/fisiologia , Animais , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3 , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD , Proteínas de Transporte/metabolismo , Proteínas de Transporte/fisiologia , Inibidor de Quinase Dependente de Ciclina p21 , Grupo dos Citocromos c/metabolismo , Camundongos , Proteínas Proto-Oncogênicas c-bcl-2/fisiologia , Células Tumorais Cultivadas , Proteína bcl-X
4.
Oncogene ; 20(31): 4128-37, 2001 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-11464279

RESUMO

Death ligands such as CD95 ligand (CD95L) or tumor necrosis factor-related apoptosis-inducing ligand/Apo2 ligand (TRAIL/Apo2L) induce apoptosis in radiochemotherapy-resistant human malignant glioma cell lines. The death-signaling TRAIL receptors 2 (TRAIL-R2/death receptor (DR) 5) and TRAIL-R1/DR4 were expressed more abundantly than the non-death-inducing (decoy) receptors TRAIL-R3/DcR1 and TRAIL-R4/DcR2 in 12 human glioma cell lines. Four of the 12 cell lines were TRAIL/Apo2L-sensitive in the absence of a protein synthesis inhibitor, cycloheximide (CHX). Three of the 12 cell lines were still TRAIL/Apo2L-resistant in the presence of CHX. TRAIL-R2 expression predicted sensitivity to apoptosis. Coexposure to TRAIL/Apo2L and cytotoxic drugs such as topotecan, lomustine (1-(2-chloroethyl)-3-cyclohexyl-1-nitrosourea, CCNU) or temozolomide resulted in synergistic killing. Synergistic killing was more often observed in cell lines retaining wild-type p53 activity (U87MG, LN-229) than in p53 mutant cell lines (LN-18, T98G, U373MG). Drug exposure resulted in enhanced TRAIL-R2 expression, but decreased TRAIL-R4 expression in U87MG cells. Ectopic expression of dominant-negative p53(V135A) abrogated the drug-induced changes in TRAIL-R2 and TRAIL-R4 expression, but had no effect on synergy. Thus, neither wild-type p53 function nor changes in TRAIL receptor expression were required for synergy. In contrast, synergy resulted possibly from drug-induced cytochrome c release from mitochondria, serving as an amplifier of the TRAIL/Apo2L-mediated cascade of caspase activation. These data provide novel insights into the role of the TRAIL/Apo2L system in malignant gliomas and illustrate that TRAIL/Apo2L-based immunochemotherapy may be an effective therapeutic strategy for these lethal neoplasms.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Apoptose/fisiologia , Neoplasias Encefálicas/patologia , Grupo dos Citocromos c/metabolismo , Glioma/patologia , Lomustina/farmacologia , Glicoproteínas de Membrana/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Proteínas Reguladoras de Apoptose , Western Blotting , Neoplasias Encefálicas/enzimologia , Neoplasias Encefálicas/metabolismo , Caspase 8 , Caspase 9 , Caspases/metabolismo , Citometria de Fluxo , Glioma/enzimologia , Glioma/metabolismo , Humanos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF , Células Tumorais Cultivadas
5.
J Immunol ; 166(12): 7238-43, 2001 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-11390472

RESUMO

TGF-beta is a putative mediator of immunosuppression associated with malignant glioma and other types of cancer. Subtilisin-like proprotein convertases such as furin are thought to mediate TGF-beta processing. Here we report that human malignant glioma cell lines express furin mRNA and protein, exhibit furin-like protease (FLP) activity, and release active furin into the cell culture supernatant. FLP activity is not modulated by exogenous TGF-beta or neutralizing TGF-beta Abs. Exposure of LN-18 and T98G glioma cell lines to the furin inhibitor, decanoyl-Arg-Val-Lys-Arg-chloromethylketone, inhibits processing of the TGF-beta1 and TGF-beta2 precursor molecules and, consequently, the release of mature bioactive TGF-beta molecules. Ectopic expression of PDX, a synthetic antitrypsin analog with antifurin activity, in the glioma cells inhibits FLP activity, TGF-beta processing, and TGF-beta release. Thus, subtilisin-like proprotein convertases may represent a novel target for the immunotherapy of malignant glioma and other cancers or pathological conditions characterized by enhanced TGF-beta bioactivity.


Assuntos
Citoplasma/enzimologia , Glioblastoma/enzimologia , Imunossupressores/metabolismo , Precursores de Proteínas/metabolismo , Processamento de Proteína Pós-Traducional/imunologia , Subtilisinas/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Linhagem Celular Transformada , Citoplasma/fisiologia , Ativação Enzimática/genética , Ativação Enzimática/imunologia , Furina , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Imunossupressores/antagonistas & inibidores , Precursores de Proteínas/antagonistas & inibidores , Precursores de Proteínas/biossíntese , Subtilisinas/antagonistas & inibidores , Subtilisinas/biossíntese , Transfecção , Fator de Crescimento Transformador beta/antagonistas & inibidores , Fator de Crescimento Transformador beta/biossíntese , Fator de Crescimento Transformador beta2 , Células Tumorais Cultivadas , alfa 1-Antitripsina/genética , alfa 1-Antitripsina/fisiologia
6.
J Neurochem ; 75(6): 2288-97, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11080180

RESUMO

The proteasome is a multiprotein complex that is involved in the intracellular protein degradation in eukaryotes. Here, we show that human malignant glioma cells are susceptible to apoptotic cell death induced by the proteasome inhibitors, MG132 and lactacystin. The execution of the apoptotic death program involves the processing of caspases 2, 3, 7, 8, and 9. Apoptosis is inhibited by ectopic expression of X-linked inhibitor of apoptosis (XIAP) and by coexposure to the broad-spectrum caspase inhibitor, benzoyl-VAD-fluoromethyl ketone (zVAD-fmk), but not by the preferential caspase 8 inhibitor, crm-A. It is interesting that specific morphological alterations induced by proteasome inhibition, such as dilated rough endoplasmic reticulum and the formation of cytoplasmic vacuoles and dense mitochondrial deposits, are unaffected by zVAD-fmk. Apoptosis is also inhibited by ectopic expression of Bcl-2 or by an inhibitor of protein synthesis, cycloheximide. Further, cytochrome c release and disruption of mitochondrial membrane potential are prominent features of apoptosis triggered by proteasome inhibition. Bcl-2 is a stronger inhibitor of cytochrome c release than zVAD-fmk. XIAP and crm-A fail to modulate cytochrome c release. These data place cytochrome c release downstream of Bcl-2 activity but upstream of XIAP- and crm-A-sensitive caspases. The partial inhibition of cytochrome c release by zVAD-fmk indicates a positive feedback loop that may involve cytochrome c release and zVAD-fmk-sensitive caspases. Finally, death ligand/receptor interactions, including the CD95/CD95 ligand system, do not mediate apoptosis induced by proteasome inhibition in human malignant glioma cells.


Assuntos
Acetilcisteína/análogos & derivados , Antineoplásicos/farmacologia , Apoptose , Caspases/metabolismo , Grupo dos Citocromos c/metabolismo , Glioma/metabolismo , Complexos Multienzimáticos/antagonistas & inibidores , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Acetilcisteína/farmacologia , Animais , Antibacterianos/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Cicloeximida/farmacologia , Cisteína Endopeptidases , Inibidores de Cisteína Proteinase/farmacologia , Relação Dose-Resposta a Droga , Proteína Ligante Fas , Glioma/patologia , Humanos , Lactamas , Leupeptinas/farmacologia , Glicoproteínas de Membrana/metabolismo , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/enzimologia , Complexo de Endopeptidases do Proteassoma , Inibidores da Síntese de Proteínas/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Proteínas Proto-Oncogênicas c-bcl-2/genética , Transgenes , Células Tumorais Cultivadas , Receptor fas/metabolismo
7.
J Neuroimmunol ; 110(1-2): 214-22, 2000 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-11024552

RESUMO

The efficacy of glucocorticoids in the treatment of multiple sclerosis may involve the induction of T cell apoptosis. Here, we report that glucocorticoids have two different effects on the vulnerability of human antigen-specific T cells: (i) steroids induce T cell apoptosis in a CD95-independent, but caspase-dependent manner; (ii) steroids protect T cells from CD95-mediated apoptosis which, however, is also caspase-dependent. An increase in BCL-2 expression is observed upon incubation with steroids. Thus, inhibition of CD95-mediated T cell apoptosis may be an undesirable side-effect resulting in survival of activated T cells and the maintenance of pathogenic immune responses might explain the lack of long-term glucocorticoid therapy.


Assuntos
Apoptose/efeitos dos fármacos , Apoptose/imunologia , Autoantígenos/imunologia , Linfócitos T CD4-Positivos/citologia , Glucocorticoides/farmacologia , Metilprednisolona/farmacologia , Linfócitos T CD4-Positivos/enzimologia , Linfócitos T CD4-Positivos/imunologia , Caspase 3 , Caspases/metabolismo , Cumarínicos/farmacologia , Fragmentação do DNA/imunologia , Proteína Ligante Fas , Corantes Fluorescentes/farmacologia , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/imunologia , Humanos , Glicoproteínas de Membrana/imunologia , Esclerose Múltipla/imunologia , Oligopeptídeos/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/imunologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Mensageiro/análise , Receptor fas/genética , Receptor fas/imunologia
8.
Oncogene ; 19(19): 2338-45, 2000 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-10822385

RESUMO

CD95L-induced apoptosis involves caspase activation and is facilitated when RNA and protein synthesis are inhibited. Here, we report that hyperthermia sensitizes malignant glioma cells to CD95L- and APO2L-induced apoptosis in the absence, but not in the presence, of inhibitors of RNA and protein synthesis. Hyperthermia does not alter CD95 expression at the cell surface and does not modulate the morphology of CD95-mediated cell death on electron microscopy. Bcl-2 gene transfer inhibits apoptosis and abrogates the sensitization mediated by hyperthermia. Hyperthermia does not overcome resistance to apoptosis conferred by the viral caspase inhibitor, crm-A, indicating the absolute requirement for the activation of crm-A-sensitive caspases, probably caspase 8, for apoptosis. CD95L-evoked DEVD-amc-cleaving caspase activity is enhanced by hyperthermia, suggesting that hyperthermia operates upstream of caspase processing to promote apoptosis. There is no uniformly enhanced processing of three caspase 3 substrates, poly-ADP ribose polymerase (PARP), protein kinase C (PKC) delta and DNA fragmentation factor (DFF) 45. Yet, hyperthermia promotes CD95L-evoked DNA fragmentation. Interestingly, hyperthermia enhances the CD95L-evoked release of cytochrome c in the absence, but not in the presence, of CHX. In contrast, the reduction of the mitochondrial membrane potential is enhanced by hyperthermia both in the absence and presence of CHX, and enhanced cytochrome c release is not associated with significantly enhanced caspase 9 processing. The potentiation of cytochrome c release at hyperthermic conditions in the absence of CHX is abrogated by Bcl-2. Thus, either hyperthermia or inhibition of protein synthesis by CHX potentiate cytotoxic cytokine-induced apoptosis. These pathways show no synergy, but rather redundance, indicating that CHX may function to promote apoptosis in response to cytotoxic cytokines by inhibiting the synthesis of specific proteins whose synthesis, function or degradation is temperature-sensitive.


Assuntos
Apoptose/fisiologia , Grupo dos Citocromos c/metabolismo , Glioma/metabolismo , Hipertermia Induzida , Glicoproteínas de Membrana/metabolismo , Proteínas Virais , Proteínas Reguladoras de Apoptose , Caspase 8 , Caspase 9 , Inibidores de Caspase , Caspases/metabolismo , Cumarínicos/metabolismo , Cicloeximida/farmacologia , Proteína Ligante Fas , Glioma/patologia , Glioma/terapia , Humanos , Isoenzimas/efeitos dos fármacos , Isoenzimas/metabolismo , Glicoproteínas de Membrana/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/efeitos dos fármacos , Oligopeptídeos/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Proteína Quinase C/efeitos dos fármacos , Proteína Quinase C/metabolismo , Proteína Quinase C-delta , Inibidores da Síntese de Proteínas/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Serpinas/metabolismo , Transdução de Sinais , Ligante Indutor de Apoptose Relacionado a TNF , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/metabolismo
9.
Cell Physiol Biochem ; 9(3): 117-25, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10494025

RESUMO

The proteasome is a multiprotein complex involved in the degradation of ubiquitinated proteins. Three proteasome inhibitors, calpain inhibitor I, lactacystin and MG132, induced apoptosis in several human malignant glioma cell lines. Although proteasome inhibitors induced p53 accumulation in a cell line retaining wild-type p53 activity, p53 activity was dispensable for apoptosis since transdominant-negative p53 abrogated p53-dependent p21 induction but did not modulate apoptosis. Further, p21 was induced by higher concentrations of proteasome inhibitors in a p53-independent manner both in p53 wild-type and in p53 mutant cell lines. Although there was a strong G2/M arrest in response to proteasome inhibition in glioma cells, this G2/M arrest was also observed in p21(-/-) colon carcinoma cells, suggesting that p21 is dispensable for the G2/M arrest associated with proteasome inhibition. Interestingly, the p21(-/-) cells were more resistant to protease inhibitors than parental p21(+/+) cells. In summary, our data indicate that proteasome inhibition induces a p21-independent G2/M arrest and p53-independent apoptosis in human malignant glioma cells.


Assuntos
Acetilcisteína/análogos & derivados , Apoptose/efeitos dos fármacos , Cisteína Endopeptidases/metabolismo , Inibidores de Cisteína Proteinase/farmacologia , Glicoproteínas/farmacologia , Leupeptinas/farmacologia , Complexos Multienzimáticos/metabolismo , Acetilcisteína/farmacologia , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Neoplasias do Colo , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/metabolismo , Inibidores Enzimáticos/metabolismo , Glioma , Humanos , Cinética , Complexo de Endopeptidases do Proteassoma , Inibidores de Serina Proteinase/farmacologia , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
10.
Oncogene ; 18(36): 5044-53, 1999 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-10490841

RESUMO

Death ligand/receptor interactions and caspase activation mediate drug-induced apoptosis in certain cancer cells. The molecular mechanisms responsible for the chemoresistance of human malignant gliomas are largely unknown. Here, we report that malignant glioma cells co-express CD95 and CD95L without undergoing suicidal or fratricidal apoptosis. Glioma cells do not commit CD95/CD95L-dependent suicide or fratricide even when RNA and protein synthesis are inhibited. This is because ectopic expression of the viral caspase inhibitor, crm-A, or exposure to a neutralizing CD95L antibody, block apoptosis induced by exogenous CD95L but not cell death induced by cytotoxic concentrations of inhibitors of RNA and protein synthesis. Although some cytotoxic drugs enhance the expression of CD95 or CD95L, crm-A fails to block drug-induced cytotoxic and clonogenic cell death, suggesting that the drug-induced changes in CD95 and CD95L expression are epiphenomenal. There is also no difference in drug-induced apoptosis between crm-A-transfected and control cells as assessed by electron microscopy, in situ DNA end labeling and DNA fragmentation. Further, glioma cells selected for resistance to CD95L do not acquire cross-resistance to chemotherapy. However, the broad spectrum caspase inhibitor, ZVAD-fmk, inhibits drug-induced cytotoxic cell death, suggesting a role of crm-A-insensitive caspases in drug-induced apoptosis of glioma cells. Thus, drug resistance of malignant glioma cells may involve deficiencies in two interrelated pathways that mediate death in order tumor cell types: (i) death ligand/receptor signalling; and (ii) caspase activation.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Caspases/metabolismo , Glioma/patologia , Receptores do Fator de Necrose Tumoral/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Ativação Enzimática , Glioma/enzimologia , Glioma/ultraestrutura , Humanos , Microscopia Eletrônica
11.
Cancer Lett ; 142(1): 11-6, 1999 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-10424775

RESUMO

The role of non-caspase protease activation in drug-induced cell death of glioma cells was examined. Neither calpain inhibitors I or II, phenylmethylsulfonyl fluoride (PMSF), Nalpha -p-tosyl-L-lysine chloromethyl ketone (TLCK), N-tosyl-L-phenylalanine chloromethyl ketone (TPCK), E64, leupeptin nor pepstatin inhibit the cytotoxicity of vincristine, cisplatin, doxorubicin, cytarabine, camptothecin, BCNU or VM26 in two malignant glioma cell lines, T98G and LN-229. However, DNA fragmentation induced by VM26 is inhibited by calpain inhibitor I, PMSF, TLCK and TPCK, and that induced by camptothecin by calpain inhibitors I and II and TPCK. Moreover, protease inhibitors fail to abrogate CD95 ligand-induced apoptosis even though DNA fragmentation is attenuated by calpain inhibitor II and TPCK. Thus, non-caspase protease activation is not required for drug-induced apoptosis of glioma cells. Protease inhibitor-mediated inhibition of DNA fragmentation operates downstream of the commitment point for cell death.


Assuntos
Antineoplásicos/farmacologia , Fragmentação do DNA/efeitos dos fármacos , Glioma/tratamento farmacológico , Inibidores de Proteases/farmacologia , Antineoplásicos/uso terapêutico , Morte Celular/efeitos dos fármacos , Antagonismo de Drogas , Glioma/genética , Glioma/patologia , Humanos , Inibidores de Proteases/uso terapêutico , Células Tumorais Cultivadas
12.
Cell Physiol Biochem ; 9(1): 29-37, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10352342

RESUMO

The temperature-sensitive murine p53val135 mutant was introduced into 3 human malignant glioma cell lines to examine the effects of the p53 status on BCL-2 family protein expression, CD95 expression, and sensitivity to CD95 ligand (CD95L)-induced apoptosis. p53val135 behaves as a dominant negative mutant at 38.5 degrees C but assumes p53 wild-type properties. In order to dissect (i) specific effects of wild-type versus mutant p53, and (ii) transdominant-negative versus gain-of-function effects of mutant p53, we included glioma cell lines with functional wild-type (LN-229), mutant (LN-18) or deleted (LN-308) p53 genes. Wild-type, but not mutant, p53val135 promoted G2/M arrest and accumulation of BAK protein in all cell lines. The levels of other BCL-2 family members including BAX, BCL-2, BCL-X or MCL-1 were not consistently modulated by mutant or wild-type p53val135. Wild-type, but not mutant, p53val135 enhanced CD95 expression in all cell lines. CD95L-evoked caspase 3 activity was unaffected by wild-type p53 in all cell lines. Unexpectedly, mutant p53val135 differentially modulated caspase 3 activity in a gain-of-function fashion in that caspase 3 activity induced by CD95L was enhanced in LN-229 and LN-308 cells but reduced in LN-18 cells. Yet, mutant p53val135 enhanced the sensitivity to CD95L in LN-18 cells, had no effect in LN-229 cells, and decreased the sensitivity of LN-308 cells. Corresponding to the unaltered CD95L-evoked caspase 3 activity, wild-type p53val135 had no major effect on CD95L-induced apoptosis, except for a moderate sensitization of LN-229 cells but only when protein synthesis was inhibited. Thus, wild-type p53 induces BAK and CD95 expression in human glioma cells without enhancing their susceptibility to CD95-mediated apoptosis, and mutant p53 modulates CD95L-evoked apoptotic signalling in a gain-of-function fashion up-stream and down-stream of caspase 3 activation.


Assuntos
Apoptose/fisiologia , Regulação Neoplásica da Expressão Gênica , Genes p53 , Glicoproteínas de Membrana/fisiologia , Proteínas de Membrana/genética , Proteína Supressora de Tumor p53/metabolismo , Receptor fas/genética , Animais , Neoplasias Encefálicas , Caspase 3 , Caspases/metabolismo , Sobrevivência Celular , Proteína Ligante Fas , Glioma , Humanos , Células K562 , Camundongos , Proteínas Recombinantes/metabolismo , Transfecção , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética , Proteína Killer-Antagonista Homóloga a bcl-2
13.
Cell Death Differ ; 6(4): 370-6, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10381630

RESUMO

The inhibitor-of-apoptosis (IAP) proteins are a novel family of antiapoptotic proteins that are thought to inhibit cell death via direct inhibition of caspases. Here, we report that human malignant glioma cell lines express XIAP, HIAP-1 and HIAP-2 mRNA and proteins. NAIP was not expressed. IAP proteins were not cleaved during CD95 ligand (CD95L)-induced apoptosis, and loss of IAP protein expression was not responsible for the potentiation of CD95L-induced apoptosis when protein synthesis was inhibited. LN-18 cells are highly sensitive to CD95-mediated apoptosis, whereas LN-229 cells require co-exposure to CD95L and a protein synthesis inhibitor, CHX, to acquire sensitivity to apoptosis. Adenoviral XIAP gene transfer blocked caspase 8 and 3 processing in both cell lines in the absence of CHX. Apoptosis was blocked in the absence and in the presence of CHX. However, XIAP failed to block caspase 8 processing in LN-229 cells in the presence of CHX. There was considerable overlap of the effects of XIAP on caspase processing with those of BCL-2 and the viral caspase inhibitor crm-A. These data define complex regulatory mechanisms for CD95-mediated apoptosis in glioma cells and indicate that there may be a distinct pathway of death receptor-mediated apoptosis that is readily activated when protein synthesis is inhibited. The constitutive expression of natural caspase inhibitors may play a role in the resistance of these cells to apoptotic stimuli that directly target caspases, including radiochemotherapy and immune-mediated tumor cell lysis.


Assuntos
Apoptose/genética , Neoplasias Encefálicas , Regulação Neoplásica da Expressão Gênica , Glioma , Adenoviridae/genética , Elementos Antissenso (Genética) , Caspases/metabolismo , Técnicas de Transferência de Genes , Humanos , Proteínas de Neoplasias/genética , Proteínas/genética , Células Tumorais Cultivadas/enzimologia , Células Tumorais Cultivadas/patologia , Células Tumorais Cultivadas/fisiologia , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X , Receptor fas/genética
14.
J Pharmacol Exp Ther ; 289(3): 1306-12, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10336521

RESUMO

Betulinic acid (BA), a pentacyclic triterpene, is an experimental cytotoxic agent for malignant melanoma. Here, we show that BA triggers apoptosis in five human glioma cell lines. BA-induced apoptosis requires new protein, but not RNA, synthesis, is independent of p53, and results in p21 protein accumulation in the absence of a cell cycle arrest. BA-induced apoptosis involves the activation of caspases that cleave poly(ADP ribose)polymerase. Interactions of death ligand/receptor pairs of the CD95/CD95 ligand family do not mediate BA-induced caspase activation. BA enhances the levels of BAX and BCL-2 proteins but does not alter the levels of BCL-xS or BCL-xL. Ectopic expression of BCL-2 prevents BA-induced caspase activation, DNA fragmentation, and cell death. Furthermore, BA induces the formation of reactive oxygen species that are essential for BA-triggered cell death. The generation of reactive oxygen species is blocked by BCL-2 and requires new protein synthesis but is unaffected by caspase inhibitors, suggesting that BA toxicity sequentially involves new protein synthesis, formation of reactive oxygen species, and activation of crm-A-insensitive caspases.


Assuntos
Antineoplásicos Fitogênicos/toxicidade , Apoptose/efeitos dos fármacos , Caspases/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Espécies Reativas de Oxigênio/fisiologia , Triterpenos/toxicidade , Ciclo Celular/efeitos dos fármacos , Divisão Celular , Sobrevivência Celular , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/genética , Cicloeximida/farmacologia , Inibidores de Cisteína Proteinase/farmacologia , Fragmentação do DNA , Ativação Enzimática , Proteína Ligante Fas , Glioma , Humanos , Cinética , Glicoproteínas de Membrana/farmacologia , Oligopeptídeos/farmacologia , Triterpenos Pentacíclicos , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Células Tumorais Cultivadas , Proteína X Associada a bcl-2 , Proteína bcl-X , Receptor fas/efeitos dos fármacos , Receptor fas/fisiologia , Ácido Betulínico
15.
J Neuroimmunol ; 87(1-2): 121-9, 1998 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-9670853

RESUMO

CD95 ligand (CD95L)-induced apoptosis is a novel immunotherapeutic approach to malignant glioma. Here, we report that interferon-alpha (IFN-alpha) sensitizes LN-229 and T98G human malignant glioma cells to CD95L-induced apoptosis. In contrast to the effects of IFN-gamma and TNF-alpha which sensitize glioma cells to CD95 antibody-induced apoptosis in part by enhancing CD95 expression, IFN-alpha has no effect on CD95 expression at the cell surface of LN-229 and T98G cells. To confirm that changes in CD95 expression are not required for the effects of IFN-alpha, we show that IFN-alpha enhances CD95L-induced apoptosis even in CD95-transfected LN-308 glioma cells. These LN-308 cells have little endogenous CD95 expression but express high levels of CD95 from a stably integrated CD95 expression plasmid. The sensitizing effects of IFN-alpha appear to be independent of cell cycle effects of IFN-alpha and are unaffected by ectopic expression of the bcl-2 proto-oncogene. IFN-alpha enhances CD95L-induced activation of caspase-3, a critical mediator of CD95L-induced cell death. IFN-alpha also increases the cytotoxic effects of BCNU, teniposide and cytarabine in both cell lines, and of vincristine in LN-229 cells. Doxorubicin and 5-fluorouracil toxicity are unaffected by IFN-alpha. IFN-alpha may be a useful adjunct to novel strategies of immunochemotherapy for malignant gliomas that target CD95-mediated apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Caspases , Glioma/fisiopatologia , Interferon-alfa/farmacologia , Receptor fas/farmacologia , Antineoplásicos Alquilantes/farmacologia , Carmustina/farmacologia , Caspase 3 , Membrana Celular/metabolismo , Cisteína Endopeptidases/metabolismo , Sinergismo Farmacológico , Expressão Gênica/fisiologia , Genes bcl-2/genética , Glioma/metabolismo , Glioma/patologia , Humanos , Proto-Oncogene Mas , Células Tumorais Cultivadas , Receptor fas/metabolismo
16.
Br J Cancer ; 77(3): 404-11, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9472635

RESUMO

The anti-tumour alkaloid taxol shows strong cytotoxic and antiproliferative activity in two human malignant glioma cell lines, T98G and LN-229. CD95 (Fas/APO-1) ligand is a novel cytotoxic cytokine of the tumour necrosis factor (TNF) family that exerts prominent antiglioma activity. At clinically relevant taxol concentrations of 5-100 nM, taxol and CD95 ligand showed significant synergistic cytotoxicity and growth inhibition. High concentrations of taxol induced G/M cell cycle arrest in both cell lines. The synergy of taxol and CD95 ligand was independent of cell cycle effects of taxol as synergy was achieved at much lower taxol concentrations than G2/M arrest and as cell cycle effects of taxol were unaffected by co-exposure to CD95 ligand. Similarly, high concentrations of taxol were required to induce p53 activity in the p53 wild-type cell line LN-229. This effect was not modulated by CD95 ligand, suggesting that synergy is also independent of p53 activation. However, taxol induced a mobility shift of the bcl-2 protein on immunoblot analysis, indicative of bcl-2 phosphorylation. Bcl-2 phosphorylation on serine was confirmed by immunoprecipitation and phosphoserine immunoblot analysis. Considering (1) that phosphorylation of bcl-2 interferes with its heterodimerization with bax and (2) the inhibition of CD95-mediated apoptosis by bcl-2, we propose that taxol sensitizes malignant glioma cells to CD95 ligand by increasing the functional bax/bcl-2 rheostat in favour of bax and thus cell death.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Glioma/tratamento farmacológico , Glicoproteínas de Membrana/farmacologia , Paclitaxel/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteína Supressora de Tumor p53/fisiologia , Sinergismo Farmacológico , Proteína Ligante Fas , Fase G2/efeitos dos fármacos , Glioma/patologia , Humanos , Imunoterapia , Mitose/efeitos dos fármacos , Fosforilação , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/farmacologia , Receptor fas/análise
17.
Cell Death Differ ; 5(10): 894-900, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10203695

RESUMO

Susceptibility to CD95 (Fas/APO-1)-mediated apoptosis in human glioma cells depends on CD95 expression and unknown factors that regulate signal transduction. Thus, LN-18 cells are highly sensitive to CD95 ligand (CD95L) whereas LN-229 cells require coexposure to inhibitors of RNA or protein synthesis for induction of apoptosis. Here, we report that caspase 8 and 3 activation, poly(ADP-ribose)polymerase cleavage and apoptosis are inhibited by the lipoxygenase inhibitor, nordihydroguaretic acid (NDGA), or ectopic expression of crm-A or bcl-2. CD95L-induced glioma cell apoptosis does not involve ceramide generation. Apoptosis induced by exogenous ceramide resembles CD95-mediated apoptosis in that bcl-2 is protective but differs in that NDGA and crm-A have no effect and in that cycloheximide (CHX) inhibits rather than potentiates ceramide-induced cell death. We conclude that caspase 8 and caspase 3 activation, but not ceramide generation, are required for CD95 ligand-induced apoptosis of glioma cells and that bcl-2, crm-A and NDGA all act upstream of caspases to inhibit apoptosis.


Assuntos
Apoptose , Caspases/metabolismo , Glioma/enzimologia , Glioma/patologia , Masoprocol/farmacologia , Glicoproteínas de Membrana/fisiologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Serpinas/metabolismo , Proteínas Virais , Animais , Apoptose/efeitos dos fármacos , Caspase 3 , Caspase 8 , Caspase 9 , Sobrevivência Celular , Ceramidas/metabolismo , Cicloeximida/farmacologia , Inibidores de Cisteína Proteinase/farmacologia , Ativação Enzimática , Proteína Ligante Fas , Humanos , Camundongos , Serpinas/genética , Transdução de Sinais , Transfecção , Células Tumorais Cultivadas , Receptor fas/fisiologia
18.
FEBS Lett ; 409(1): 17-23, 1997 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-9199495

RESUMO

CD95 ligand is a cytotoxic cytokine that induces apoptosis. Here we report that CD95-mediated apoptosis of human malignant glioma cells is associated with arachidonic acid (AA) release. Inhibitors of phospholipase A2, phospholipase C or diacylglycerol lipase have minor effects on AA release and fail to modulate apoptosis. Formation of two AA metabolites generated during CD95-dependent apoptosis is attenuated by the lipoxygenase inhibitor, nordihydroguaretic acid (NDGA). NDGA also blocks CD95 ligand-induced apoptosis. This effect is independent of antioxidant properties of NDGA. Lipoxygenase may thus play a critical role in CD95 ligand-induced apoptosis of human malignant glioma cells.


Assuntos
Apoptose/efeitos dos fármacos , Apoptose/imunologia , Glioma/terapia , Inibidores de Lipoxigenase/farmacologia , Glicoproteínas de Membrana/fisiologia , Receptor fas/metabolismo , Animais , Ácido Araquidônico/metabolismo , Proteína Ligante Fas , Glioma/imunologia , Glioma/metabolismo , Humanos , Ligantes , Masoprocol/farmacologia , Camundongos , Fosfolipases/antagonistas & inibidores , Células Tumorais Cultivadas
19.
J Mol Cell Cardiol ; 26(7): 797-808, 1994 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-7966348

RESUMO

Calcium-free incubation followed by exposure to calcium damages naturally occurring cardiac muscle preparations irreversibly. Whether the observed calcium overload during calcium repletion is a primary cause for, or a secondary consequence of, sarcolemmal disruption and cell injury is controversial. We used cultured embryonic chicken heart muscle cells to correlate ionic, metabolic, and ultrastructural changes during calcium depletion (0Ca, 1 mM EGTA) and repletion. After 10 min of calcium depletion, intracellular Na increased four-fold above control levels, intracellular K decreased by 26%, total cell Ca decreased by 81%, and cytosolic ionized Ca concentration decreased by 87%. Within 10 min of subsequent calcium repletion, total cell Ca transiently increased to four-fold above control, cytosolic ionized Ca concentration transiently increased to twice control, and both Na and K returned toward control levels; by 3 h of calcium repletion, physiological cation (Na, K, Ca) contents were restored and adenine nucleotide contents were normal. Long-term (i.e. 120 min) calcium depletion did not significantly reduce cell ATP levels, but increased adenine nucleotide turnover as indicated by adenosine and lactate release; after 60 min of subsequent calcium repletion, ionic and metabolic parameters were returned to control levels. During calcium depletion (both short- and long-term) and subsequent repletion, no ultrastructural changes occurred. When Mg was also removed during calcium depletion, the ionic changes during depletion and subsequent repletion were enhanced. When 10 microM CCCP was present during calcium depletion and repletion, cytosolic ionized Ca concentration increased to six-fold above control with no increase in total cell Ca content, suggesting that the increased Ca is buffered, in part, by mitochondria. These results indicate that an increase in Ca per se, occurring when high energy phosphate levels and/or cellular Ca buffering capacity are maintained, does not seem to be associated with irreversible cell injury.


Assuntos
Cálcio/metabolismo , Miocárdio/metabolismo , Animais , Soluções Tampão , Células Cultivadas , Embrião de Galinha , Íons , Modelos Biológicos , Miocárdio/citologia
20.
Eur J Pharmacol ; 208(3): 261-4, 1991 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-1663050

RESUMO

In cultured rat heart muscle cells, reversible long-term ATP depletion induces a decrease in beta-adrenoceptor density and a fall in isoproterenol- as well as forskolin-stimulated cAMP formation. However, isoproterenol-stimulated adenylyl cyclase activity in membrane preparations is not reduced after ATP depletion. These results suggest that the decreased responsiveness to catecholamines during myocardial ischemia cannot be explained by alterations of the beta-adrenoceptor-adenylyl cyclase system alone.


Assuntos
Trifosfato de Adenosina/metabolismo , Adenilil Ciclases/metabolismo , Miocárdio/metabolismo , Receptores Adrenérgicos beta/metabolismo , Animais , Catecolaminas/metabolismo , Células Cultivadas , Doença das Coronárias/enzimologia , Doença das Coronárias/metabolismo , Desoxiglucose/farmacologia , Miocárdio/citologia , Miocárdio/ultraestrutura , Ratos , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...