Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Más filtros










Intervalo de año de publicación
1.
Oncogene ; 34(5): 661-9, 2015 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-24488010

RESUMEN

Many of the current antitumor therapeutic strategies are based on the perturbation of the cell cycle, especially during mitosis. Antimitotic drugs trigger mitotic checkpoint activation, mitotic arrest and eventually cell death. However, mitotic slippage represents a major mechanism of resistance to these treatments. In an attempt to circumvent the process of slippage, targeting mitotic exit has been proposed as a better strategy to kill tumor cells. In this study, we show that treatments that induce mitotic checkpoint activation and mitotic arrest downregulate FLICE-like inhibitory protein (FLIP) levels and sensitize several tumor cell lines to TRAIL (tumor necrosis factor-related apoptosis-inducing ligand)-induced apoptosis. Interestingly, we also demonstrate that in absence of mitotic checkpoint activation, mitotic arrest induced either by Cdc20 knockdown or overexpression of nondegradable cyclin B is sufficient to induce both FLIP downregulation and sensitivity to TRAIL. In summary, our data suggest that a combination of antimitotic drugs targeting cyclin B degradation and TRAIL might prevent mitotic slippage and allow tumor cells to reach the threshold for apoptosis induction, thereby facilitating tumor suppression.


Asunto(s)
Antimitóticos/administración & dosificación , Neoplasias de la Mama/genética , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/biosíntesis , Ligando Inductor de Apoptosis Relacionado con TNF/biosíntesis , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Proteínas Cdc20/genética , Línea Celular Tumoral , Ciclina B/antagonistas & inhibidores , Resistencia a Antineoplásicos/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Mitosis/efectos de los fármacos , Ligando Inductor de Apoptosis Relacionado con TNF/genética
2.
Cell Death Differ ; 19(12): 1908-16, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22722337

RESUMEN

Increased activation of the epidermal growth factor receptor (EGFR) is frequently observed in tumors, and inhibition of the signaling pathways originated in the EGFR normally renders tumor cells more sensitive to apoptotic stimuli. However, we show that inhibition of EGFR signaling in non-transformed breast epithelial cells by EGF deprivation or gefitinib, an inhibitor of EGFR tyrosine kinase, causes the upregulation of the long isoform of caspase-8 inhibitor FLICE-inhibitory protein (FLIP(L)) and makes these cells more resistant to the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). We demonstrate that the extracellular signal-regulated kinase (ERK)1/2 pathway plays a pivotal role in the regulation of FLIP(L) levels and sensitivity to TRAIL-induced apoptosis by EGF. Upregulation of FLIP(L) upon EGF deprivation correlates with a decrease in c-Myc levels and c-Myc knockdown by siRNA induces FLIP(L) expression. FLIP(L) upregulation and resistance to TRAIL in EGF-deprived cells are reversed following activation of an estrogen activatable form of c-Myc (c-Myc-ER). Finally, constitutive activation of the ERK1/2 pathway in HER2/ERBB2-transformed cells prevents EGF deprivation-induced FLIP(L) upregulation and TRAIL resistance. Collectively, our results suggest that a regulated ERK1/2 pathway is crucial to control FLIP(L) levels and sensitivity to TRAIL in non-transformed cells, and this mechanism may explain the increased sensitivity of tumor cells to TRAIL, in which the ERK1/2 pathway is frequently deregulated.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Línea Celular Tumoral , Resistencia a Antineoplásicos , Factor de Crecimiento Epidérmico/farmacología , Células Epiteliales/metabolismo , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Femenino , Humanos , Proteínas Proto-Oncogénicas c-myc/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Regulación hacia Arriba/efectos de los fármacos
3.
Clin Transl Oncol ; 13(12): 839-47, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22126726

RESUMEN

In tumour cells, activation of the apoptotic machinery by death receptor ligands of the tumour necrosis factor (TNF) receptor superfamily of cytokines represents a novel therapeutic strategy. However, systemic treatment of tumours with TNF-α and CD95 ligand may produce severe toxic effects. The tumour necrosis-related apoptosisinducing ligand (TRAIL) is a member of the TNF family capable of inducing apoptosis in a wide variety of cancer cells upon binding to pro-apoptotic receptors, while having no effect on the majority of normal human cells tested. Interestingly, preclinical studies in mice and nonhuman primates showed no systemic cytotoxicity upon injection of either recombinant TRAIL or agonistic TRAIL-receptor antibodies. Furthermore, these treatments have been shown to effectively suppress the growth of a range of tumour xenografts. Although unwanted effects of some TRAIL preparations have been reported in normal cells, the use of TRAIL receptor agonists could represent a suitable approach in cancer therapy. Here, we shall review our current understanding of apoptotic and non-apoptotic TRAIL signalling, the therapeutic potential of TRAIL-based approaches in cancer treatment, and the results of phase 1 and 2 clinical trials with recombinant TRAIL or agonistic TRAIL receptor antibodies, either as monotherapy or in combination with other chemotherapeutic agents.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Humanos , Ratones
4.
Clin. transl. oncol. (Print) ; 13(12): 839-847, dic. 2011. ilus, tab
Artículo en Inglés | IBECS | ID: ibc-125991

RESUMEN

In tumour cells, activation of the apoptotic machinery by death receptor ligands of the tumour necrosis factor (TNF) receptor superfamily of cytokines represents a novel therapeutic strategy. However, systemic treatment of tumours with TNF-α and CD95 ligand may produce severe toxic effects. The tumour necrosis-related apoptosisinducing ligand (TRAIL) is a member of the TNF family capable of inducing apoptosis in a wide variety of cancer cells upon binding to pro-apoptotic receptors, while having no effect on the majority of normal human cells tested. Interestingly, preclinical studies in mice and nonhuman primates showed no systemic cytotoxicity upon injection of either recombinant TRAIL or agonistic TRAIL-receptor antibodies. Furthermore, these treatments have been shown to effectively suppress the growth of a range of tumour xenografts. Although unwanted effects of some TRAIL preparations have been reported in normal cells, the use of TRAIL receptor agonists could represent a suitable approach in cancer therapy. Here, we shall review our current understanding of apoptotic and non-apoptotic TRAIL signalling, the therapeutic potential of TRAIL-based approaches in cancer treatment, and the results of phase 1 and 2 clinical trials with recombinant TRAIL or agonistic TRAIL receptor antibodies, either as monotherapy or in combination with other chemotherapeutic agents (AU)


Asunto(s)
Humanos , Animales , Masculino , Femenino , Ratones , Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Transducción de Señal , Oncología Médica/métodos , Oncología Médica/organización & administración , Oncología Médica/tendencias
5.
Cell Death Differ ; 17(5): 883-94, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-19942932

RESUMEN

Breast tumor cells are often resistant to tumor necrosis factor-related apoptosis-inducing ligand (tumour necrosis factor-related apoptosis-inducing ligand (TRAIL)/APO-2 L). Here, we describe the sensitization by microtubule-interfering agents (MIAs) to TRAIL-induced apoptosis in breast tumor cells through a mitotic arrest and c-Jun N-terminal kinase (JNK)-dependent mechanism. MIA treatment resulted in BubR1-dependent mitotic arrest leading to the sustained activation of JNK and the proteasome-mediated downregulation of cellular FLICE-inhibitory protein (cFLIP) and myeloid cell leukemia-1 (Mcl-1) expression. The JNK inhibitor SP600125 abrogated MIA-induced mitotic arrest and downregulation of cFLIP and Mcl-1 and reduced the apoptosis caused by the combination of MIAs and TRAIL. Silencing of cFLIP and Mcl-1 expression by RNA interference resulted in a marked sensitization to TRAIL-induced apoptosis. Furthermore, in FLIP-overexpressing cells, MIA-induced sensitization to TRAIL-activated apoptosis was markedly reduced. In summary, our results show that mitotic arrest imposed by MIAs activates JNK and facilitates TRAIL-induced activation of an apoptotic pathway in breast tumor cells by promoting the proteasome-mediated degradation of cFLIP and Mcl-1.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Mitosis/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/uso terapéutico , Moduladores de Tubulina/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Citometría de Flujo , Humanos , Immunoblotting , Proteínas Quinasas JNK Activadas por Mitógenos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Nocodazol/farmacología , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
6.
Cell Death Differ ; 13(11): 1857-65, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16485030

RESUMEN

Breast cancer cells often show increased activity of the mitogen-activated protein kinase (MAPK) pathway. We report here that this pathway reduces their sensitivity to death ligand tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and present the underlying mechanism. Activation of protein kinase C (PKC) inhibited TRAIL-induced apoptosis in a protein synthesis-independent manner. Deliberate activation of MAPK was also inhibitory. In digitonin-permeabilized cells, PKC activation interfered with the capacity of recombinant truncated (t)Bid to release cytochrome c from mitochondria. MAPK activation did not affect TRAIL or tumor necrosis factor (TNF)alpha-induced Bid cleavage. However, it did inhibit translocation of (t)Bid to mitochondria as determined both by subcellular fractionation analysis and confocal microscopy. Steady state tBid mitochondrial localization was prohibited by activation of the MAPK pathway, also when the Bcl-2 homology domain 3 (BH3) domain of tBid was disrupted. We conclude that the MAPK pathway inhibits TRAIL-induced apoptosis in MCF-7 cells by prohibiting anchoring of tBid to the mitochondrial membrane. This anchoring is independent of its interaction with resident Bcl-2 family members.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/metabolismo , Sistema de Señalización de MAP Quinasas , Mitocondrias/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Neoplasias de la Mama/patología , Caspasas/metabolismo , Activación Enzimática/efectos de los fármacos , Humanos , Membranas Mitocondriales/metabolismo , Permeabilidad/efectos de los fármacos , Unión Proteica , Biosíntesis de Proteínas/efectos de los fármacos , Proteína Quinasa C/metabolismo , Transporte de Proteínas/efectos de los fármacos , Receptores del Factor de Necrosis Tumoral/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Acetato de Tetradecanoilforbol/farmacología , Factor de Necrosis Tumoral alfa/farmacología
7.
Oncogene ; 20(48): 7128-33, 2001 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-11704839

RESUMEN

Bc1-2 protein is a potent anti-apoptotic protein that inhibits a mitochondria-operated pathway of apoptosis in many cells. DNA damaging agents and death receptor ligands can activate this mitochondrial apoptotic mechanism. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has been suggested to escape from the inhibitory action of Bc1-2 protein. We show that in human breast tumor MCF-7 cells, TRAIL induced a mitochondrial pathway of apoptosis that involved cytochrome c release from mitochondria and activation of caspase 9. The DNA damaging drug doxorubicin also activated this mitochondria-regulated mechanism of apoptosis, which was inhibited in Bc1-2-overexpressing cells. We also demonstrate that in MCF-7 cells Bc1-2 might confer resistance to TRAIL-induced apoptosis, depending on the expression levels of the anti-apoptotic protein. These results indicate that enhanced expression of Bc1-2 in tumor cells can render these cells less sensitive not only to chemotherapeutic drugs but also to TRAIL.


Asunto(s)
Adenocarcinoma/patología , Antibióticos Antineoplásicos/farmacología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/patología , Doxorrubicina/farmacología , Resistencia a Antineoplásicos/fisiología , Glicoproteínas de Membrana/farmacología , Proteínas de Neoplasias/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/fisiología , Factor de Necrosis Tumoral alfa/farmacología , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Proteínas Reguladoras de la Apoptosis , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Caspasa 9 , Caspasas/metabolismo , Grupo Citocromo c/metabolismo , Daño del ADN , ADN de Neoplasias/efectos de los fármacos , ADN de Neoplasias/genética , Activación Enzimática/efectos de los fármacos , Femenino , Humanos , Mitocondrias/efectos de los fármacos , Proteínas de Neoplasias/análisis , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF , Tubulina (Proteína)/análisis , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/patología , Proteína X Asociada a bcl-2
8.
Cell Death Differ ; 8(2): 172-81, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11313719

RESUMEN

TRAIL causes apoptosis in numerous types of tumor cells. However, the mechanisms regulating TRAIL-induced apoptosis remain to be elucidated. We have investigated the role of PKC in regulating TRAIL-induced mitochondrial events and apoptosis in the Jurkat T cell line. We found a caspase-dependent decline in mitochondrial membrane potential and translocation of cytochrome c from mitochondria into the cytosol in response to TRAIL. Both these events were prevented by PKC activation. Moreover, PKC activation considerably reduced the activation of caspases, PARP cleavage and apoptosis when induced upon TRAIL treatment. MAPK activation was involved in the mechanism of PKC-mediated inhibition of TRAIL-induced cytochrome c release from mitochondria. Furthermore, inhibition of the MAPK pathway partially reversed the PKC-mediated inhibition of TRAIL-induced apoptosis. Besides, PKC activation may also inhibit the TRAIL-induced apoptosis through a MAPK-independent mechanism. Altogether, these results indicate a negative role of PKC in the regulation of apoptotic signals generated upon TRAIL receptor activation.


Asunto(s)
Apoptosis/fisiología , Caspasas/metabolismo , Leucemia de Células T/metabolismo , Glicoproteínas de Membrana/metabolismo , Mitocondrias/metabolismo , Proteína Quinasa C/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Reguladoras de la Apoptosis , Inhibidores de Caspasas , Línea Celular , Activación Enzimática/fisiología , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Forbol 12,13-Dibutirato/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF
9.
J Biol Chem ; 276(21): 17779-87, 2001 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-11279136

RESUMEN

The role of interferon (IFN)-gamma as a sensitizing agent in apoptosis induced by ligation of death receptors has been evaluated in human myeloid leukemia cells. Incubation of U937 cells with IFN-gamma sensitized these cells to apoptosis induced by tumor necrosis factor-alpha, agonistic CD95 antibody, and tumor necrosis factor-related apoptosis-inducing ligand. Other human myeloid leukemic cells were also sensitized by IFN-gamma to death receptor-mediated apoptosis. Treatment of U937 cells with IFN-gamma up-regulated the expression of caspase-8 and potently synergized with death receptor ligation in the processing of caspase-8 and BID cleavage. Concomitantly, a marked down-regulation of BCL-2 protein was also observed in cells incubated with IFN-gamma. Furthermore, the caspase-dependent generation of a 23-kDa fragment of BCL-2 protein, the release of cytochrome c from mitochondria and the activation of caspase-9 were also enhanced upon death receptor ligation in IFN-gamma-treated cells. Ectopically expressed Bcl-2 protein inhibited IFN-gamma-induced sensitization to apoptosis. In summary, these results indicate that IFN-gamma sensitizes human myeloid leukemic cells to a death receptor-induced, mitochondria-mediated pathway of apoptosis.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Interferón gamma/farmacología , Receptores del Factor de Necrosis Tumoral/fisiología , Caspasa 8 , Caspasa 9 , Caspasas/fisiología , Humanos , Leucemia Mieloide/patología , Proteínas Proto-Oncogénicas c-bcl-2/fisiología , Transducción de Señal/efectos de los fármacos , Células U937
10.
Biochem J ; 353(Pt 1): 101-108, 2001 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-11115403

RESUMEN

Treatment of haematopoietic BA/F3 cells with the thymidylate synthase inhibitor 5-fluoro-2'-deoxyuridine (FUdR) activated apoptosis through a mechanism that required continuous protein synthesis and was inhibited by Bcl-2 over-expression. Analysis of p53 levels in cells treated with FUdR indicated a marked accumulation of this protein. Accumulation of p53 was also observed in cells over-expressing Bcl-2. In BA/F3 cells transfected with a cDNA coding for the human papilloma virus protein E6, p53 accumulation after FUdR treatment was inhibited markedly. However, apoptosis was induced in both control and E6 cells to a similar extent. The role of the CD95/CD95 ligand (CD95L) system in FUdR-induced apoptosis was also assessed. As determined by reverse transcriptase PCR, BA/F3 expressed a low constitutive level of CD95L mRNA, which decreased following FUdR treatment. Moreover, blocking CD95-CD95L interactions with antagonistic CD95 monoclonal antibody did not prevent drug-induced apoptosis. Furthermore, analysis of caspase involvement showed important differences in apoptosis induced by CD95-triggering or FUdR treatment. In summary, these results suggest that apoptosis induced by thymineless stress in haematopoietic BA/F3 cells occurs by a mechanism that does not require accumulation of p53 and which is independent of CD95-CD95L interactions.


Asunto(s)
Apoptosis/efectos de los fármacos , Células Sanguíneas/efectos de los fármacos , Floxuridina/farmacología , Glicoproteínas de Membrana/metabolismo , Timidilato Sintasa/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/metabolismo , Receptor fas/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Células Sanguíneas/citología , Células Sanguíneas/enzimología , Células Sanguíneas/metabolismo , Caspasas/metabolismo , Línea Celular , Cicloheximida/farmacología , Fragmentación del ADN/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Proteína Ligando Fas , Ratones , Poli(ADP-Ribosa) Polimerasas/metabolismo , Biosíntesis de Proteínas , Agregación de Receptores , Timidilato Sintasa/metabolismo
11.
Cancer Res ; 60(20): 5673-80, 2000 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-11059759

RESUMEN

In this report, we have assessed the role of IFN-gamma as a sensitizing agent in apoptosis mediated by activation of death receptor CD95 in breast tumor cells. Treatment of the tumor cell lines MCF-7 and MDA-MB231 with IFN-gamma significantly facilitated apoptosis induced by CD95 receptor ligation at the plasma membrane, independently of p53 status. In contrast, IFN-gamma treatment did not enhance the apoptotic effect of the DNA-damaging drug, doxorubicin. Analysis of apoptosis regulators indicated that caspase-8 mRNA and protein levels were up-regulated in both of the cell lines after treatment with IFN-gamma. Furthermore, IFN-gamma sensitized MCF-7 and MDA-MB231 cells to CD95-mediated activation of caspase-8, induction of cytochrome c release from mitochondria, and processing of caspase-9. Release of cytochrome c, caspases activation, and apoptosis were prevented in MCF-7 cells overexpressing Bcl-2. Altogether these results indicate that IFN-gamma, maybe through the elevation of caspase-8 levels, sensitizes human breast tumor cells to a death receptor-mediated, mitochondria-operated pathway of apoptosis.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/patología , Caspasas/biosíntesis , Interferón gamma/farmacología , Mitocondrias/efectos de los fármacos , Receptor fas/fisiología , Anticuerpos Monoclonales/farmacología , Apoptosis/fisiología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/enzimología , Caspasa 8 , Caspasa 9 , Caspasas/metabolismo , Grupo Citocromo c/metabolismo , Activación Enzimática , Humanos , Mitocondrias/enzimología , Mitocondrias/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Células Tumorales Cultivadas/efectos de los fármacos , Proteína p53 Supresora de Tumor/biosíntesis , Proteína p53 Supresora de Tumor/genética , Regulación hacia Arriba/efectos de los fármacos , Receptor fas/inmunología
12.
J Immunol ; 163(9): 4737-46, 1999 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-10528172

RESUMEN

We have recently reported that activation of protein kinase C (PKC) plays a negative role in CD95-mediated apoptosis in human T cell lines. Here we present data indicating that although the PKC-induced mitogen-activated protein kinase pathway could be partially implicated in the abrogation of CD95-mediated apoptosis by phorbol esters in Jurkat T cells, the major inhibitory effect is exerted through a PKC-dependent, mitogen-activated protein kinase-independent signaling pathway. Furthermore, we demonstrate that activation of PKC diminishes CD95 receptor aggregation elicited by agonistic CD95 Abs. On the other hand, it has been reported that UV radiation-induced apoptosis is mediated at least in part by the induction of CD95 oligomerization at the cell surface. Here we show that activation of PKC also inhibits UVB light-induced CD95 aggregation and apoptosis in Jurkat T cells. These results reveal a novel mechanism by which T cells may restrain their sensitivity to CD95-induced cell death through PKC-mediated regulation of CD95 receptor oligomerization at the cell membrane.


Asunto(s)
Apoptosis/inmunología , Proteína Quinasa C/fisiología , Proteínas Serina-Treonina Quinasas , Linfocitos T/enzimología , Linfocitos T/inmunología , Receptor fas/fisiología , Animales , Anticuerpos Monoclonales/farmacología , Apoptosis/efectos de la radiación , Activación Enzimática/efectos de los fármacos , Activación Enzimática/inmunología , Activación Enzimática/efectos de la radiación , Inhibidores Enzimáticos/farmacología , Proteína Ligando Fas , Flavonoides/farmacología , Humanos , Células Jurkat , Ligandos , MAP Quinasa Quinasa 1 , Glicoproteínas de Membrana/biosíntesis , Glicoproteínas de Membrana/efectos de la radiación , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteína Quinasa C/metabolismo , Ratas , Agregación de Receptores/inmunología , Agregación de Receptores/efectos de la radiación , Linfocitos T/fisiología , Linfocitos T/efectos de la radiación , Rayos Ultravioleta , Receptor fas/inmunología , Receptor fas/metabolismo
13.
Cell Death Differ ; 6(3): 271-80, 1999 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10200578

RESUMEN

Induction of CD95 (Fas/APO-1) and CD95 ligand during chemotherapeutic treatment may contribute to the death by apoptosis of some tumor cells. In this study, we have analyzed the role of the CD95 system in genotoxic drug-induced death of human breast tumor cells. Incubation of the breast tumor cell lines MCF-7 and EVSA-T with doxorubicin or methotrexate caused apoptosis after 48 h of treatment. These drugs induced a marked increase in the level of CD95 mRNA and protein in wild-type p53-expressing MCF-7 cells. On the contrary, the breast cancer cell line EVSA-T that expresses high levels of an inactive form of p53, did not up-regulate CD95 upon drug treatment. Elevation of CD95 expression by DNA-damaging drugs was notably blocked in MCF-7 cells expressing the human papillomavirus type 16 E6 protein (E6 cells) which prevented p53 accumulation upon DNA damage. However, E6 cells were still killed by the drugs. Furthermore, the genotoxic drugs did not induce the expression of CD95 ligand in MCF-7 cells at doses that caused apoptosis in these breast tumor cells. Moreover, drug-induced apoptosis of breast tumor cells was not prevented in the presence of either a CD95 antagonistic antibody or a CD95 ligand blocking antibody. We also observed a strong synergism between lower doses of DNA-damaging drugs and CD95 agonistic antibody in the induction of apoptosis in MCF-7 cells. In summary, our data indicate that drug-induced apoptosis of breast tumor cells occurs by a CD95/CD95L-independent mechanism although by elevating the tumor suppressor proteins p53 and CD95, genotoxic drugs may sensitize breast tumor cells to CD95-mediated apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Mutágenos/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Receptor fas/genética , Receptor fas/metabolismo , Apoptosis/genética , Apoptosis/fisiología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Daño del ADN , Proteína Ligando Fas , Femenino , Humanos , Glicoproteínas de Membrana/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , Temperatura , Células Tumorales Cultivadas , Regulación hacia Arriba/efectos de los fármacos
14.
Cell Death Differ ; 5(3): 214-21, 1998 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10200467

RESUMEN

In order to determine whether disruption of mitochondrial function could trigger apoptosis in murine haematopoietic cells, we used the potassium ionophore valinomycin. Valinomycin induces apoptosis in the murine pre-B cell line BAF3, which cannot be inhibited by interleukin-3 addition or Bcl-2 over-expression. Valinomycin triggers rapid loss of mitochondrial membrane potential. This precedes cytoplasmic acidification, which leads to cysteine-active-site protease activation, DNA fragmentation and cell death. Bongkrekic acid, an inhibitor of the mitochondrial permeability transition, prevents acidification and subsequent induction of apoptosis by valinomycin.


Asunto(s)
Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Ionóforos/farmacología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Valinomicina/farmacología , Animales , Ácido Bongcréquico/farmacología , Línea Celular , Concentración de Iones de Hidrógeno , Líquido Intracelular/efectos de los fármacos , Líquido Intracelular/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Ratones , Permeabilidad
15.
Eur J Immunol ; 27(6): 1442-50, 1997 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-9209497

RESUMEN

Activation of protein kinase C (PKC) has been reported to inhibit Fas (APO-1, CD95)-mediated apoptosis in different cellular systems. Human Jurkat leukemic T cells express the Fas antigen in the cell membrane and undergo apoptosis upon cross-linking by anti-Fas monoclonal antibodies (mAb). Cleavage of the apoptosis-associated protease CPP32 and its substrate poly(ADP-ribose)polymerase are observed after the engagement of Fas antigen with mAb. In this report, we show that all these effects are substantially inhibited by the activation of PKC with a phorbol ester. Bisindolylmaleimide, an inhibitor of PKC, prevents phorbol ester-induced down-regulation of Fas signaling. Inhibition of Fas-mediated cell death by phorbol ester is also observed in other human leukemic T cell lines. Cross-linking of Fas antigen by mAb results in the rapid increase in tyrosine phosphorylation of several protein substrates which is further elevated in the presence of the protein tyrosine phosphatase inhibitor, orthovanadate. Furthermore, orthovanadate markedly enhances the cell death response to Fas mAb in different human leukemic T cell lines and human T cell blasts. These effects of orthovanadate on early tyrosine phosphorylation and cell death are clearly diminished by PKC activation. These results strongly suggest that tyrosine phosphorylation is involved in Fas signaling in apoptosis and that PKC plays a negative role in Fas-mediated apoptosis by counteracting at a very early stage the signals generated following cross-linking of this receptor.


Asunto(s)
Apoptosis/inmunología , Proteína Quinasa C/metabolismo , Transducción de Señal/inmunología , Receptor fas/fisiología , Adyuvantes Inmunológicos/farmacología , Apoptosis/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Activación Enzimática/inmunología , Humanos , Células Jurkat , Forbol 12,13-Dibutirato/farmacología , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/fisiología , Transducción de Señal/efectos de los fármacos , Células Tumorales Cultivadas , Vanadatos/farmacología
16.
J Biol Chem ; 272(16): 10624-30, 1997 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-9099710

RESUMEN

Perturbing deoxyribonucleoside triphosphate (dNTP) metabolism with inhibitors of the de novo synthesis of dNTP causes apoptosis in the interleukin-3 (IL-3)-dependent pre-B cell line BAF3. Under these conditions apoptosis is prevented when deoxyribonucleosides for dNTP synthesis are supplied in the culture medium. On the other hand, removal of IL-3 from cultures of BAF3 cells resulted in down-regulation of thymidine kinase activity, rapid imbalance in dNTP levels, and apoptosis. In this study we show that overexpression of a heterologous thymidine kinase, herpes simplex virus thymidine kinase (TK), in BAF3 cells protects these cells from apoptosis induced by either inhibitors of dNTP synthesis or IL-3 deprivation. This protection against apoptosis is abrogated by 9-(4-hydroxybutyl)-N2-phenylguanine, a specific inhibitor of herpes simplex virus-1 TK. These results suggest that deoxyribonucleoside kinases, particularly TK, may be important in the regulation of apoptosis in hemopoietic cells.


Asunto(s)
Apoptosis , Interleucina-3/farmacología , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Timidina Quinasa/biosíntesis , Animales , Apoptosis/efectos de los fármacos , Linfocitos B , Línea Celular , Supervivencia Celular/efectos de los fármacos , Medios de Cultivo Condicionados , Desoxirribonucleótidos/metabolismo , Desoxirribonucleótidos/farmacología , Floxuridina/farmacología , Herpesvirus Humano 1/enzimología , Hidroxiurea/farmacología , Cinética , Metotrexato/farmacología , Ratones , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/metabolismo , Timidina Quinasa/metabolismo , Transfección
17.
J Cell Sci ; 110 ( Pt 5): 653-61, 1997 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-9092947

RESUMEN

ICE-like protease activation and DNA fragmentation are preceded by a decrease in intracellular pH (pHi) during apoptosis in the IL-3 dependent cell line BAF3. Acidification occurs after 7 hours in cells deprived of IL-3 and after 4 hours when cells are treated with etoposide, close to the time of detection of ICE-like protease activity. Increasing extracellular pH reduces ICE-like protease activation and DNA fragmentation. Bcl-2 over-expression both delays acidification and inhibits ICE-like protease activation. Generation of a rapid intracellular pH decrease, using the ionophore nigericin, induces ICE-like protease activation and apoptosis. ZVAD, a cell permeable inhibitor of ICE-like proteases, does not affect acidification but inhibits apoptosis induced by IL-3 removal or nigericin treatment. These data suggest that intracellular acidification triggers apoptosis by directly or indirectly activating ICE-like proteases.


Asunto(s)
Apoptosis , Cisteína Endopeptidasas/metabolismo , Línea Celular , Fragmentación del ADN , Activación Enzimática , Citometría de Flujo , Humanos , Concentración de Iones de Hidrógeno , Hidrólisis , Interleucina-3/farmacología , Poli(ADP-Ribosa) Polimerasas/metabolismo
18.
Experientia ; 52(10-11): 995-1000, 1996 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-8917730

RESUMEN

Fidelity in DNA synthesis and repair is largely dependent on a balanced supply of deoxynucleotide triphosphate (dNTP) pools. Results from different groups have shown that alterations in dNTP supply result in DNA fragmentation and cell death with characteristics of apoptosis. We have recently shown that in apoptosis driven by deprivation of interleukin-3 (IL-3) in a murine hemopoietic cell line, there is a rapid imbalance in the availability of dNTP that precedes DNA fragmentation. In these cells, dNTP pool balance is closely coupled to the function of the salvage pathway of dNTP synthesis. Apoptosis, induced by treatment of these cells with drugs that inhibit the de novo dNTP synthesis, is prevented when dNTP precursors are supplied through the salvage pathway. IL-3 regulates thymidine kinase activity, suggesting that alterations in dNTP metabolism after IL-3 deprivation could be a relevant event in the commitment of hemopoietic cells to apoptosis.


Asunto(s)
Apoptosis , Desoxirribonucleótidos/fisiología , Animales , Muerte Celular , Células Cultivadas , ADN/metabolismo , Reparación del ADN , Interleucina-3/fisiología , Ratones , Modelos Biológicos , Mutación , Neoplasias Experimentales/metabolismo
19.
J Cell Sci ; 109 ( Pt 9): 2393-9, 1996 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-8886988

RESUMEN

DNA fragmentation in isolated nuclei from the murine IL3-dependent bone marrow cell line BAF3 could be stimulated either by decreasing pH below 6.5 or by adding microM calcium at neutral pH. An endonuclease which could also be stimulated either by a decrease in pH, to 6.5, or by the presence of microM calcium at neutral pH, was purified 10(4)-fold from nuclei of BAF3 cells. Digestion of DNA with the purified enzyme resulted in 5'-terminal hydroxyl and 3'-terminal phosphate ends. These characteristics are distinct from those described for other mammalian endonucleases. The possible role of this enzyme in genome digestion during apoptosis is discussed.


Asunto(s)
Apoptosis/fisiología , Endonucleasas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Calcio/farmacología , Línea Celular , Fragmentación del ADN , Activación Enzimática , Concentración de Iones de Hidrógeno , Ratones
20.
FEBS Lett ; 392(3): 299-303, 1996 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-8774867

RESUMEN

HL-60 acute myeloblastic and U937 monoblastoid leukaemic cell lines both cleave poly(ADP-ribose)polymerase (PARP), at the onset of apoptosis, in response to a wide range of cytotoxic agents. This appears to be a common feature of leukaemic cell apoptosis. However, in the chronic myelogenous leukaemic (CML) derived cell line, K562, no such cleavage was detectable. This correlated with previous findings that this cell line is particularly resistant to apoptosis induced by cytotoxic agents. Proteolytic cleavage of PARP and the subsequent progression to apoptosis was inhibited by two protease inhibitors NEM and IOD. As both PARP cleavage and DNA fragmentation appeared closely linked in these cell lines, anti-oxidants (previously shown to be effective inhibitors of DNA fragmentation and apoptosis) were also demonstrated to prevent PARP cleavage. These results combine to suggest that ROI may mediate PARP cleavage, DNA fragmentation and the eventual apoptosis of these cells following cytotoxic insult.


Asunto(s)
Apoptosis/fisiología , Leucemia/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Antioxidantes/farmacología , Cisteína Endopeptidasas/metabolismo , Inhibidores de Cisteína Proteinasa/farmacología , ADN/metabolismo , Humanos , Leucemia/tratamiento farmacológico , Leucemia/patología , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA