Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Oncogenesis ; 5: e209, 2016 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-26974204

RESUMO

The sphingosine kinase-1/sphingosine 1-phosphate (SphK1/S1P) signaling pathway has been reported to modulate the expression of the canonical transcription factor hypoxia-inducible HIF-1α in multiple cell lineages. HIF-2α is also frequently overexpressed in solid tumors but its role has been mostly studied in clear cell renal cell carcinoma (ccRCC), the most common form of kidney cancer, where HIF-2α has been established as a driver of a more aggressive disease. In this study, the role of SphK1/S1P signaling with regard to HIF-2α was investigated in various cancer cell models including ccRCC cells. Under hypoxic conditions or in ccRCC lacking a functional von Hippel-Lindau (VHL) gene and expressing high levels of HIF-2α, SphK1 activity controls HIF-2α expression and transcriptional activity through a phospholipase D (PLD)-driven mechanism. SphK1 silencing promotes a VHL-independent HIF-2α loss of expression and activity and reduces cell proliferation in ccRCC. Importantly, downregulation of SphK1 is associated with impaired Akt and mTOR signaling in ccRCC. Taking advantage of a monoclonal antibody neutralizing extracellular S1P, we show that inhibition of S1P extracellular signaling blocks HIF-2α accumulation in ccRCC cell lines, an effect mimicked when the S1P transporter Spns2 or the S1P receptor 1 (S1P1) is silenced. Here, we report the first evidence that the SphK1/S1P signaling pathway regulates the transcription factor hypoxia-inducible HIF-2α in diverse cancer cell lineages notably ccRCC, where HIF-2α has been established as a driver of a more aggressive disease. These findings demonstrate that SphK1/S1P signaling may act as a canonical regulator of HIF-2α expression in ccRCC, giving support to its inhibition as a therapeutic strategy that could contribute to reduce HIF-2 activity in ccRCC.

2.
Prog Urol ; 26(3): 159-67, 2016 Mar.
Artigo em Francês | MEDLINE | ID: mdl-26896427

RESUMO

PURPOSE: Tumor hypoxia and its biological consequence lead to microenvironment adaptation of tumor initiation, promotion and progression. The aim of the study was to observe the influence of hypoxia on the expression of the androgen receptor (AR), using an original model of multicellular spheroids obtained from castration-resistant prostate tumor cells. MATERIAL: Two human castration-resistant prostate cancer cell lines have been used to generate multicellular tumor spheroids (MTS). The conditions and duration of incubation modulated the final size of the MTS and the intrinsic hypoxia gradient. The expression of AR was studied by immunohistochemistry (IHC) and secretion of PSA measured in the culture medium. RESULTS: The IHC expression of AR was characterized by a decreasing gradient from the periphery to the center of MTS (less intense in central hypoxic zone), corresponding to a nuclear translocation of activated AR. This result was proportionally correlated with the duration of hypoxic incubation period. Hypoxia caused significant increase in AR expression at 6h of oxygen deprivation. This activation of AR was correlated with transcriptional activity increase of target genes, including increased secretion of PSA. CONCLUSION: This demonstration of activation, increased expression and increased transcriptional activity of AR by hypoxia is the first to have been made with an original model of hypoxia, closer to reality than previous models, i.e. close to tissue hypoxia observed in primary prostate cancer. LEVEL OF EVIDENCE: 4.


Assuntos
Neoplasias de Próstata Resistentes à Castração/patologia , Receptores Androgênicos/biossíntese , Hipóxia Tumoral , Microambiente Tumoral , Humanos , Masculino , Esferoides Celulares , Células Tumorais Cultivadas
3.
Leukemia ; 22(5): 971-9, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18401414

RESUMO

We examined the involvement of sphingosine kinase-1 (SphK1), which governs the ceramide/sphingosine-1-phosphate balance, in susceptibility to imatinib of either sensitive or resistant chronic myeloid leukemia cells. Imatinib-sensitive LAMA84-s displayed marked SphK1 inhibition coupled with increased content of ceramide and decreased pro-survival sphingosine-1-phosphate. Conversely, no changes in the sphingolipid metabolism were observed in LAMA84-r treated with imatinib. Overcoming imatinib resistance in LAMA84-r with farnesyltransferase or MEK/ERK inhibitors as well as with cytosine arabinoside led to SphK1 inhibition. Overexpression of SphK1 in LAMA84-s cells impaired apoptosis and inhibited the effects of imatinib on caspase-3 activation, cytochrome c and Smac release from mitochondria through modulation of Bim, Bcl-xL and Mcl-1 expression. Pharmacological inhibition of SphK1 with F-12509a or its silencing by siRNA induced apoptosis of both imatinib-sensitive and -resistant cells, suggesting that SphK1 inhibition was critical for apoptosis signaling. We also show that imatinib-sensitive and -resistant primary cells from chronic myeloid leukemia patients can be successfully killed in vitro by the F-12509a inhibitor. These results uncover the involvement of SphK1 in regulating imatinib-induced apoptosis and establish that SphK1 is a downstream effector of the Bcr-Abl/Ras/ERK pathway inhibited by imatinib but upstream regulator of Bcl-2 family members.


Assuntos
Apoptose , Proteínas de Fusão bcr-abl/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Fosfotransferases (Aceptor do Grupo Álcool)/fisiologia , Piperazinas/farmacologia , Pirimidinas/farmacologia , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose , Benzamidas , Benzoquinonas/farmacologia , Resistencia a Medicamentos Antineoplásicos , Humanos , Mesilato de Imatinib , Leucemia Mielogênica Crônica BCR-ABL Positiva/enzimologia , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Transdução de Sinais
4.
Cell Death Differ ; 14(2): 197-208, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16645635

RESUMO

Somatostatin is a multifunctional hormone that modulates cell proliferation, differentiation and apoptosis. Mechanisms for somatostatin-induced apoptosis are at present mostly unsolved. Therefore, we investigated whether somatostatin receptor subtype 2 (sst2) induces apoptosis in the nontransformed murine fibroblastic NIH3T3 cells. Somatostatin receptor subtype 2 expression induced an executioner caspase-mediated apoptosis through a tyrosine phosphatase SHP-1 (Src homology domain phosphatase-1)-dependent stimulation of nuclear factor kappa B (NF-kappaB) activity and subsequent inhibition of the mitogen-activated protein kinase JNK. Tumor necrosis factor alpha (TNFalpha) stimulated both NF-kappaB and c-Jun NH2-terminal kinase (JNK) activities, which had opposite action on cell survival. Importantly, sst2 sensitized NIH3T3 cells to TNFalpha-induced apoptosis by (1) upregulating TNFalpha receptor protein expression, and sensitizing to TNFalpha-induced caspase-8 activation; (2) enhancing TNFalpha-mediated activation of NF-kappaB, resulting in JNK inhibition and subsequent executioner caspase activation and cell death. We have here unraveled a novel signaling mechanism for a G protein-coupled receptor, which directly triggers apoptosis and crosstalks with a death receptor to enhance death ligand-induced apoptosis.


Assuntos
Apoptose , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Receptores de Somatostatina/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Somatostatina/metabolismo , Animais , Apoptose/efeitos dos fármacos , Caspases/metabolismo , Ativação Enzimática/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Camundongos , Modelos Biológicos , Mimetismo Molecular/efeitos dos fármacos , Células NIH 3T3 , Fosforilação/efeitos dos fármacos , Proteína Fosfatase 1 , Proteína Tirosina Fosfatase não Receptora Tipo 6/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Transdução de Sinais/efeitos dos fármacos , Transfecção , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/farmacologia
5.
Leukemia ; 20(1): 95-102, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16281067

RESUMO

We examined the involvement of sphingosine kinase-1, a critical regulator of the sphingolipid balance, in susceptibility to antineoplastic agents of either sensitive or multidrug-resistant acute myeloid leukemia cells. Contrary to parental HL-60 cells, doxorubicin and etoposide failed to trigger apoptosis in chemoresistant HL-60/Doxo and HL-60NP16 cells overexpressing MRP1 and MDR1, respectively. Chemosensitive HL-60 cells displayed sphingosine kinase-1 inhibition coupled with ceramide generation. In contrast, chemoresistant HL-60/ Doxo and HL-60/VP16 had sustained sphingosine kinase-1 activity and did not produce ceramide during treatment. Enforced expression of sphingosine kinase-1 in chemosensitive HL-60 cells resulted in marked inhibition of apoptosis that was mediated by blockade of mitochondrial cytochrome c efflux hence suggesting a control of apoptosis at the pre-mitochondrial level. Incubation with cell-permeable ceramide of chemoresistant cells led to a sphingosine kinase-1 inhibition and apoptosis both prevented by sphingosine kinase-1 over-expression. Furthermore, F-12509a, a new sphingosine kinase inhibitor, led to ceramide accumulation, decrease in sphingosine 1-phosphate content and caused apoptosis equally in chemosensitive and chemoresistant cell lines that is inhibited by adding sphingosine 1-phosphate or overexpressing sphingosine kinase-1. F-12509a induced classical apoptosis hallmarks namely nuclear fragmentation, caspase-3 cleavage as well as downregulation of antiapoptotic XIAP, and release of cytochrome c and SMAC/Diablo.


Assuntos
Resistência a Múltiplos Medicamentos , Leucemia Mieloide/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Doença Aguda , Apoptose/efeitos dos fármacos , Benzoquinonas/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Ceramidas/biossíntese , Doxorrubicina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Etoposídeo/farmacologia , Células HL-60 , Humanos , Leucemia Mieloide/tratamento farmacológico , Mitocôndrias/efeitos dos fármacos , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/farmacologia , Interferência de RNA/fisiologia , Receptores de Lisoesfingolipídeo/metabolismo
6.
Blood ; 98(9): 2828-36, 2001 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-11675357

RESUMO

Sphingosine 1-phosphate (S-1P) has been implicated as a second messenger preventing apoptosis by counteracting activation of executioner caspases. Here it is reported that S-1P prevents apoptosis and executioner caspase-3 activation by inhibiting the translocation of cytochrome c and Smac/DIABLO from mitochondria to the cytosol induced by anti-Fas, tumor necrosis factor-alpha (TNF-alpha), serum deprivation, and cell-permeable ceramides in the human acute leukemia Jurkat, U937, and HL-60 cell lines. Furthermore, the tumor promoter 12-O-tetradecanoyl-phorbol-13-acetate, which stimulates sphingosine kinase, the enzyme responsible for S-1P production, also inhibits cytochrome c and Smac/DIABLO release. In contrast, dimethylsphingosine (DMS), a specific inhibitor of sphingosine kinase, sensitizes cells to cytochrome c and Smac/DIABLO release triggered by anti-Fas, TNF-alpha, serum deprivation, or ceramide. DMS-induced mitochondrial apoptogenic factor leakage can likewise be overcome by S-1P cotreatment. Hence, S-1P, likely generated through a protein kinase C- mediated activation of sphingosine kinase, inhibits the apoptotic cascade upstream of the release of the mitochondrial apoptogenic factors, cytochrome c, and Smac/DIABLO in human acute leukemia cells.


Assuntos
Apoptose/efeitos dos fármacos , Proteínas de Transporte/antagonistas & inibidores , Grupo dos Citocromos c/antagonistas & inibidores , Leucemia/patologia , Lisofosfolipídeos , Mitocôndrias/metabolismo , Proteínas Mitocondriais/antagonistas & inibidores , Esfingosina/análogos & derivados , Esfingosina/farmacologia , Proteínas Reguladoras de Apoptose , Proteínas de Transporte/efeitos dos fármacos , Proteínas de Transporte/metabolismo , Ceramidas/farmacologia , Meios de Cultura Livres de Soro/farmacologia , Grupo dos Citocromos c/efeitos dos fármacos , Grupo dos Citocromos c/metabolismo , Células HL-60 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Células Jurkat , Proteínas Mitocondriais/efeitos dos fármacos , Proteínas Mitocondriais/metabolismo , Transporte Proteico/efeitos dos fármacos , Receptores do Fator de Necrose Tumoral/fisiologia , Acetato de Tetradecanoilforbol/farmacologia , Células U937
7.
J Clin Invest ; 108(1): 143-51, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11435466

RESUMO

TNF-alpha is a pleiotropic cytokine activating several signaling pathways initiated at distinct intracellular domains of the TNF receptors. Although the C-terminal region is believed to be responsible for apoptosis induction, the functions of more membrane-proximal domains, including the domain that couples to neutral sphingomyelinase activation, are not yet fully elucidated. The roles of this region and of the associated adapter protein FAN (factor associated with neutral SMase activation) in the cytotoxic response to TNF have been investigated. We have now shown that stable expression in human fibroblasts of a dominant negative form of FAN abrogates TNF-induced ceramide generation from sphingomyelin hydrolysis and reduces caspase processing, thus markedly inhibiting TNF-triggered apoptosis. However, the cytotoxic responses to daunorubicin and exogenous ceramide remain unaltered, as do the TNF-induced p42/p44 MAPK activation and CD54 expression. Fibroblasts from FAN-knockout mice also proved to be resistant to TNF toxicity. These findings highlight the previously unrecognized role of the adapter protein FAN in signaling cell death induction by TNF.


Assuntos
Apoptose/fisiologia , Proteínas/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Clorometilcetonas de Aminoácidos/farmacologia , Animais , Antígenos CD/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Caspases/metabolismo , Linhagem Celular Transformada , Células Cultivadas/efeitos dos fármacos , Ceramidas/biossíntese , Ceramidas/farmacologia , Inibidores de Cisteína Proteinase/farmacologia , Daunorrubicina/farmacologia , Ativação Enzimática/efeitos dos fármacos , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Genes Dominantes , Humanos , Hidrólise , Molécula 1 de Adesão Intercelular/biossíntese , Molécula 1 de Adesão Intercelular/genética , Peptídeos e Proteínas de Sinalização Intracelular , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Camundongos Knockout , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Estrutura Terciária de Proteína , Proteínas/genética , Receptores do Fator de Necrose Tumoral/efeitos dos fármacos , Receptores Tipo I de Fatores de Necrose Tumoral , Proteínas Recombinantes de Fusão/fisiologia , Sistemas do Segundo Mensageiro , Esfingomielina Fosfodiesterase/metabolismo , Esfingomielinas/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Fator de Necrose Tumoral alfa/toxicidade , Células U937
8.
Cell Death Differ ; 8(2): 162-71, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11313718

RESUMO

Treatment of human breast carcinoma MCF7 cells with doxorubicin, one of the most active antineoplastic agents used in clinical oncology, induces apoptosis and leads to increases in sphingosine levels. The transient generation of this sphingolipid mediator preceded cytochrome c release from the mitochondria and activation of the executioner caspase-7 in MCF7 cells which do not express caspase-3. Bcl-x(L) overexpression did not affect sphingosine generation whereas it reduced apoptosis triggered by doxorubicin and completely blocked apoptosis triggered by sphingosine. Exogenous sphingosine-induced apoptosis was also accompanied by cytochrome c release and activation of caspase-7 in a Bcl-x(L)-sensitive manner. Furthermore, neither doxorubicin nor sphingosine treatment affected expression of Fas ligand or induced activation of the apical caspase-8, indicating a Fas/Fas ligand-independent mechanism. Our results suggest that a further metabolite of ceramide, sphingosine, may also be involved in mitochondria-mediated apoptotic signaling induced by doxorubicin in human breast cancer cells.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Caspases/metabolismo , Grupo dos Citocromos c/metabolismo , Doxorrubicina/farmacologia , Esfingosina/biossíntese , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/metabolismo , Antineoplásicos/metabolismo , Apoptose/fisiologia , Neoplasias da Mama/metabolismo , Caspase 7 , Doxorrubicina/metabolismo , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Feminino , Humanos , Mitocôndrias/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteína bcl-X
9.
J Neurochem ; 76(5): 1573-84, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11238741

RESUMO

Sphingosine-1-phosphate (SPP), a bioactive sphingolipid metabolite, suppresses apoptosis of many types of cells, including rat pheochromocytoma PC12 cells. Elucidating the molecular mechanism of action of SPP is complicated by many factors, including uptake and metabolism, as well as activation of specific G-protein-coupled SPP receptors, known as the endothelial differentiation gene-1 (EDG-1) family. In this study, we overexpressed type 1 sphingosine kinase (SPHK1), the enzyme that converts sphingosine to SPP, in order to examine more directly the role of intracellularly generated SPP in neuronal survival. Enforced expression of SPHK1 in PC12 cells resulted in significant increases in kinase activity, with corresponding increases in intracellular SPP levels and concomitant decreases in both sphingosine and ceramide, and marked suppression of apoptosis induced by trophic factor withdrawal or by C(2)-ceramide. NGF, which protects PC12 cells from serum withdrawal-induced apoptosis, also stimulated SPHK1 activity. Surprisingly, overexpression of SPHK1 had no effect on activation of two known NGF-stimulated survival pathways, extracellular signal regulated kinase ERK 1/2 and Akt. However, trophic withdrawal-induced activation of the stress activated protein kinase, c-Jun amino terminal kinase (SAPK/JNK), and activation of the executionary caspases 2, 3 and 7, were markedly suppressed. Moreover, this abrogation of caspase activation, which was prevented by the SPHK inhibitor N,N-dimethylsphingosine, was not affected by pertussis toxin treatment, indicating that the cytoprotective effect was likely not mediated by binding of SPP to cell surface G(i)-coupled SPP receptors. In agreement, there was no detectable release of SPP into the culture medium, even after substantially increasing cellular SPP levels by NGF or sphingosine treatment. In contrast to PC12 cells, C6 astroglioma cells secreted SPP, suggesting that SPP might be one of a multitude of known neurotrophic factors produced and secreted by glial cells. Collectively, our results indicate that SPHK/SPP may play an important role in neuronal survival by regulating activation of SAPKs and caspases.


Assuntos
Apoptose/fisiologia , Caspases/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Proteínas Serina-Treonina Quinases , Esfingosina/análogos & derivados , Neoplasias das Glândulas Suprarrenais , Animais , Apoptose/efeitos dos fármacos , Caspase 2 , Caspase 3 , Caspase 7 , Meios de Cultura Livres de Soro , Proteínas Quinases JNK Ativadas por Mitógeno , Cinética , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fator de Crescimento Neural/farmacologia , Células PC12 , Toxina Pertussis , Feocromocitoma , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Ratos , Proteínas Recombinantes/metabolismo , Esfingosina/farmacologia , Transfecção , Fatores de Virulência de Bordetella/farmacologia
10.
FASEB J ; 15(2): 297-9, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11156942

RESUMO

Stress-induced activation of an acidic sphingomyelinase leading to generation of ceramide, an important lipid mediator, has been associated with apoptosis; however, the implication of this hydrolase has been questioned. The present study aimed at re-evaluating the role of this lysosomal enzyme in apoptosis initiated by different apoptotic inducers. The sensitivity of a series of acid sphingomyelinase-deficient cell lines derived from Niemann-Pick disease patients to stress-induced apoptosis was investigated. We have now shown that stress stimuli, such as anthracyclines, ionizing radiation, and Fas ligation trigger similar apoptotic hallmarks in normal and acid sphingomyelinase-deficient cell lines. Retrovirus-mediated gene correction of enzyme deficiency in Niemann-Pick cells does not modify response to apoptosis. Ceramide production is comparable in normal and Niemann-Pick cells, and increased activity of neutral sphingomyelinase is observed. Thus, our findings cast serious doubts that lysosomal sphingomyelinase activation is responsible for stress-induced apoptosis of cultured cells.


Assuntos
Apoptose/fisiologia , Fumonisinas , Lisossomos/enzimologia , Esfingomielina Fosfodiesterase/metabolismo , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Ácidos Carboxílicos/farmacologia , Caspase 3 , Caspases/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Ceramidas/metabolismo , Daunorrubicina/toxicidade , Doxorrubicina/toxicidade , Inibidores Enzimáticos/farmacologia , Humanos , Linfócitos/enzimologia , Linfócitos/patologia , Linfócitos/fisiologia , Doenças de Niemann-Pick , Peptídeo Hidrolases/metabolismo , Valores de Referência , Receptor fas/fisiologia
11.
Cancer Res ; 60(16): 4468-74, 2000 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-10969794

RESUMO

Ceramide has been implicated as an important component of radiation-induced apoptosis of human prostate cancer cells. We examined the role of the sphingolipid metabolites--ceramide, sphingosine, and sphingosine-1-phosphate--in susceptibility to radiation-induced apoptosis in prostate cancer cell lines with different sensitivities to gamma-irradiation. Exposure of radiation-sensitive TSU-Pr1 cells to 8-Gy irradiation led to a sustained increase in ceramide, beginning after 12 h of treatment and increasing to 2.5- to 3-fold within 48 h. Moreover, irradiation of TSU-Pr1 cells also produced a marked and rapid 50% decrease in the activity of sphingosine kinase, the enzyme that phosphorylates sphingosine to form sphingosine-1-phosphate. In contrast, the radiation-insensitive cell line, LNCaP, had sustained sphingosine kinase activity and did not produce elevated ceramide levels on 8-Gy irradiation. Although LNCaP cells are highly resistant to gamma-irradiation-induced apoptosis, they are sensitive to the death-inducing effects of tumor necrosis factor alpha, which also increases ceramide levels in these cells (K. Kimura et al., Cancer Res., 59: 1606-1614, 1999). Moreover, we found that although irradiation alone did not increase sphingosine levels in LNCaP cells, tumor necrosis factor alpha plus irradiation induced significantly higher sphingosine levels and markedly reduced intracellular levels of sphingosine-1-phosphate. The elevation of sphingosine levels either by exogenous sphingosine or by treatment with the sphingosine kinase inhibitor N,N-dimethylsphingosine induced apoptosis and also sensitized LNCaP cells to gamma-irradiation-induced apoptosis. Our data suggest that the relative levels of sphingolipid metabolites may play a role in determining the radiosensitivity of prostate cancer cells, and that the enhancement of ceramide and sphingosine generation could be of therapeutic value.


Assuntos
Apoptose/efeitos da radiação , Neoplasias da Próstata/patologia , Tolerância a Radiação/fisiologia , Esfingosina/análogos & derivados , Esfingosina/fisiologia , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Caspases/metabolismo , Ceramidas/metabolismo , Ceramidas/fisiologia , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Humanos , Masculino , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/fisiologia , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/radioterapia , Tolerância a Radiação/efeitos dos fármacos , Radiossensibilizantes/farmacologia , Esfingomielina Fosfodiesterase/farmacologia , Esfingosina/metabolismo , Esfingosina/farmacologia , Células Tumorais Cultivadas/efeitos da radiação
12.
J Biol Chem ; 275(21): 15691-700, 2000 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-10747891

RESUMO

Exposure to anti-Fas antibody in Jurkat cells (type II cells), which are characterized by a weak caspase-8 activation at the death-inducing signaling complex (DISC), induced a biphasic increase in ceramide levels. The early generation of ceramide preceded transient activation of acidic ceramidase and subsequent production of sphingosine, followed by cytochrome c release, activation of caspases-2, -3, -6, -7, -8, and -9, Bid cleavage, and a later sustained ceramide accumulation. The caspase inhibitor benzyloxycarbonyl-Val-Ala-Asp-fluoromethyl ketone inhibited early increases of ceramide and sphingosine, whereas overexpression of Bcl-x(L) had no effect, and both prevented the later sustained ceramide accumulation. Exogenous sphingosine, as well as cell-permeable C(2)-ceramide, induced cytochrome c release from mitochondria in a caspase-independent fashion leading to activation of caspase-9 and executioner caspases and, surprisingly, activation of the initiator caspase-8 and processing of its substrate Bid. These effects were also completely abolished by Bcl-x(L) overexpression. Our results suggest that sphingosine might also be involved in the mitochondria-mediated pathway of Fas-induced cell death in type II cells.


Assuntos
Apoptose , Fumonisinas , Mitocôndrias/metabolismo , Esfingosina/metabolismo , Receptor fas/metabolismo , Clorometilcetonas de Aminoácidos/farmacologia , Anticorpos Monoclonais/farmacologia , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3 , Ácidos Carboxílicos/farmacologia , Proteínas de Transporte/metabolismo , Caspases/metabolismo , Ceramidas/metabolismo , Inibidores de Cisteína Proteinase/farmacologia , Grupo dos Citocromos c/metabolismo , Ativação Enzimática , Regulação da Expressão Gênica , Humanos , Células Jurkat , Oligopeptídeos/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Esfingosina/análogos & derivados , Esfingosina/farmacologia , Transfecção , Proteína bcl-X , Receptor fas/imunologia
13.
Blood ; 94(10): 3583-92, 1999 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-10552970

RESUMO

ELL-12, a liposome formulation of the ether-lipid 1-O-octadecyl-2-O-methyl-sn-glycero-3-phosphocholine (ET-18-OCH(3)), is a nonmyelosuppressive antiproliferative agent that is more effective and less toxic than the ether lipid itself in tumor model systems. We found that ELL-12 induced apoptosis in Jurkat, H9, and U-937 cells that was preceded by activation of executioner caspases. In addition, ELL-12 triggered release of cytochrome c from mitochondria to the cytoplasm before caspase-9 activation. Apoptosis, activation of caspases, and cytochrome c release were blocked by Bcl-x(L) overexpression in Jurkat T cells, suggesting a critical role for mitochondria in ELL-12-triggered cell death. Furthermore, ELL-12 had no effect on expression of CD95 ligand, and inhibition of the Fas signaling pathway with antagonistic anti-CD95 antibody did not affect apoptosis induced by ELL-12. Hence, ELL-12 could be a promising adjunct for the treatment of tumors in addition to myelosuppressive chemotherapeutic drugs and/or those that use the CD95-ligand/receptor system to trigger apoptosis.


Assuntos
Antineoplásicos/farmacologia , Apoptose , Grupo dos Citocromos c/metabolismo , Éteres Fosfolipídicos/farmacologia , Receptor fas/metabolismo , Transporte Biológico , Caspases/metabolismo , Grupo dos Citocromos c/fisiologia , Citosol/metabolismo , Relação Dose-Resposta a Droga , Portadores de Fármacos , Sistemas de Liberação de Medicamentos , Ativação Enzimática , Humanos , Células Jurkat , Laminas , Lipossomos , Mitocôndrias/metabolismo , Proteínas Nucleares/metabolismo , Peptídeo Hidrolases/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Transdução de Sinais , Fatores de Tempo , Células U937 , Proteína bcl-X
14.
J Cell Biol ; 147(3): 545-58, 1999 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-10545499

RESUMO

Sphingosine-1-phosphate (SPP) is a bioactive lipid that has recently been identified as the ligand for the EDG family of G protein-coupled cell surface receptors. However, the mitogenic and survival effects of exogenous SPP may not correlate with binding to cell-surface receptors (Van Brocklyn, J.R., M.J. Lee, R. Menzeleev, A. Olivera, L. Edsall, O. Cuvillier, D.M. Thomas, P.J.P. Coopman, S. Thangada, T. Hla, and S. Spiegel. 1998. J. Cell Biol. 142:229-240). The recent cloning of sphingosine kinase, a unique lipid kinase responsible for the formation of SPP, has provided a new tool to investigate the role of intracellular SPP. Expression of sphingosine kinase markedly increased SPP levels in NIH 3T3 fibroblasts and HEK293 cells, but no detectable secretion of SPP into the medium was observed. The increased sphingosine kinase activity in NIH 3T3 fibroblasts was sufficient to promote growth in low- serum media, expedite the G(1)/S transition, and increase DNA synthesis and the proportion of cells in the S phase of the cell cycle with a concomitant increase in cell numbers. Transient or stable overexpression of sphingosine kinase in NIH 3T3 fibroblasts or HEK293 cells protected against apoptosis induced by serum deprivation or ceramide elevation. N,N-Dimethylsphingosine, a competitive inhibitor of sphingosine kinase, blocked the effects of sphingosine kinase overexpression on cell proliferation and suppression of apoptosis. In contrast, pertussis toxin did not abrogate these biological responses. In Jurkat T cells, overexpression of sphingosine kinase also suppressed serum deprivation- and ceramide-induced apoptosis and, to a lesser extent, Fas-induced apoptosis, which correlated with inhibition of DEVDase activity, as well as inhibition of the executionary caspase-3. Taken together with ample evidence showing that growth and survival factors activate sphingosine kinase, our results indicate that SPP functions as a second messenger important for growth and survival of cells. Hence, SPP belongs to a novel class of lipid mediators that can function inside and outside cells.


Assuntos
Divisão Celular , Sobrevivência Celular , Lisofosfolipídeos , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Receptores Acoplados a Proteínas G , Sistemas do Segundo Mensageiro , Esfingosina/análogos & derivados , Animais , Apoptose/efeitos dos fármacos , Caspase 3 , Inibidores de Caspase , Caspases/metabolismo , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Citosol/metabolismo , DNA/biossíntese , Ativação Enzimática/efeitos dos fármacos , Humanos , Camundongos , Peptídeo Hidrolases/metabolismo , Toxina Pertussis , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fator de Crescimento Derivado de Plaquetas/farmacologia , Receptores de Superfície Celular/antagonistas & inibidores , Receptores de Superfície Celular/metabolismo , Receptores de Lisofosfolipídeos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Sistemas do Segundo Mensageiro/efeitos dos fármacos , Esfingosina/antagonistas & inibidores , Esfingosina/genética , Esfingosina/metabolismo , Esfingosina/farmacologia , Transfecção , Fatores de Virulência de Bordetella/farmacologia , Receptor fas/metabolismo
15.
Biochemistry ; 37(37): 12892-8, 1998 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-9737868

RESUMO

Sphingosine 1-phosphate (SPP), a lipid second messenger formed by the action of sphingosine kinase, has been implicated in regulating diverse biological processes, including growth, survival, and differentiation. N,N-Dimethylsphingosine (DMS) inhibits sphingosine kinase and has been used to investigate the biological roles of SPP; however, little is known of the mechanism of inhibition of sphingosine kinase by DMS. In addition, DMS has been shown to inhibit protein kinase C in vitro. Here we report that DMS is a competitive inhibitor of sphingosine kinase from U937 monoblastic leukemia cells, Swiss 3T3 fibroblasts, and PC12 pheochromocytoma cells. DMS decreases basal levels of SPP and prevents increases in SPP in response to physiological stimuli known to activate sphingosine kinase. DMS also effectively increases cellular levels of ceramide in a variety of cell types, and resetting of the ceramide/SPP rheostat may account for the pro-apoptotic effects of DMS. Moreover, DMS, at concentrations which effectively inhibit sphingosine kinase, has no effect on protein kinase C activity or its membrane translocation. Thus, DMS acts as a specific competitive inhibitor of sphingosine kinase in diverse cell types and is a useful tool to elucidate the role of SPP as an intracellular second messenger.


Assuntos
Ceramidas/metabolismo , Inibidores Enzimáticos/farmacologia , Lisofosfolipídeos , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Proteína Quinase C/metabolismo , Esfingosina/análogos & derivados , Células 3T3 , Animais , Ativação Enzimática/efeitos dos fármacos , Humanos , Camundongos , Células PC12 , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Ratos , Esfingosina/metabolismo , Esfingosina/farmacologia , Células Tumorais Cultivadas
16.
J Cell Biol ; 142(1): 229-40, 1998 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-9660876

RESUMO

Sphingosine-1-phosphate (SPP), a bioactive lipid, acts both intracellularly and extracellularly to cause pleiotropic biological responses. Recently, we identified SPP as a ligand for the G protein-coupled receptor Edg-1 (Lee, M.-J., J.R. Van Brocklyn, S. Thangada, C.H. Liu, A.R. Hand, R. Menzeleev, S. Spiegel, and T. Hla. 1998. Science. 279:1552-1555). Edg-1 binds SPP with remarkable specificity as only sphinganine-1-phosphate displaced radiolabeled SPP, while other sphingolipids did not. Binding of SPP to Edg-1 resulted in inhibition of forskolin-stimulated cAMP accumulation, in a pertussis toxin-sensitive manner. In contrast, two well-characterized biological responses of SPP, mitogenesis and prevention of apoptosis, were clearly unrelated to binding to Edg-1 and correlated with intracellular uptake. SPP also stimulated signal transduction pathways, including calcium mobilization, activation of phospholipase D, and tyrosine phosphorylation of p125(FAK), independently of edg-1 expression. Moreover, DNA synthesis in Swiss 3T3 fibroblasts was significantly and specifically increased by microinjection of SPP. Finally, SPP suppresses apoptosis of HL-60 and pheochromocytoma PC12 cells, which do not have specific SPP binding or expression of Edg-1 mRNA. Conversely, sphinganine-1-phosphate, which binds to and signals via Edg-1, does not have any significant cytoprotective effect. Thus, SPP is a prototype for a novel class of lipid mediators that act both extracellularly as ligands for cell surface receptors and intracellularly as second messengers.


Assuntos
Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Proteínas Imediatamente Precoces/metabolismo , Lisofosfolipídeos , Receptores de Superfície Celular/metabolismo , Receptores Acoplados a Proteínas G , Esfingosina/análogos & derivados , Células 3T3 , Animais , Divisão Celular , Linhagem Celular , Sobrevivência Celular , AMP Cíclico/metabolismo , Células HL-60 , Humanos , Proteínas Imediatamente Precoces/genética , Camundongos , Células PC12 , Ratos , Receptores de Lisofosfolipídeos , Transdução de Sinais , Esfingosina/metabolismo , Esfingosina/fisiologia
17.
Ann N Y Acad Sci ; 845: 11-8, 1998 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-9668339

RESUMO

Recent evidence suggests that branching pathways of sphingolipid metabolism may mediate either apoptotic or mitogenic responses depending on the cell type and the nature of the stimulus. While ceramide has been shown to be an important regulatory component of apoptosis induced by tumor necrosis factor alpha and Fas ligand, sphingosine-1-phosphate (SPP), a further metabolite of ceramide, has been implicated as a second messenger in cellular proliferation and survival induced by platelet-derived growth factor, nerve growth factor, and serum. SPP protects cells from apoptosis resulting from elevations of ceramide. Inflammatory cytokines stimulate sphingomyelinase, but not ceramidase, leading to accumulation of ceramide, whereas growth signals also leading to accumulation of ceramide, whereas growth signals also stimulate ceramidase and sphingosine kinase leading to increased SPP levels. We propose that the dynamic balance between levels of sphingolipid metabolites, ceramide, and SPP, and consequent regulation of different family members of mitogen-activated protein kinases (JNK versus ERK), is an important factor that determines whether a cell survives or dies.


Assuntos
Apoptose/fisiologia , Divisão Celular/fisiologia , Lisofosfolipídeos , Receptores Acoplados a Proteínas G , Transdução de Sinais , Esfingosina/análogos & derivados , Animais , Apoptose/efeitos dos fármacos , Ceramidas/farmacologia , Ceramidas/fisiologia , Humanos , Fatores de Crescimento Neural/farmacologia , Receptores de Superfície Celular/fisiologia , Receptores de Lisofosfolipídeos , Esfingosina/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Fator de Necrose Tumoral alfa/fisiologia , Receptor fas/fisiologia
18.
Cancer Res ; 58(9): 1817-24, 1998 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-9581819

RESUMO

Sphingolipid breakdown products [ceramide, sphingosine, and sphingosine-1-phosphate (SPP)] are emerging as a new class of bioactive molecules. In agreement with previous studies, treatment of human promyelocytic leukemia HL-60 cells with 1-alpha,25-dihydroxyvitamin D3 [1,25-(OH)2D3] induced a transient increase of ceramide levels within 2 h, which then returned to basal levels within 8 h. In contrast, sphingosine kinase activity increased more slowly and reached maximal levels only after 20 h of exposure, leading to a concomitant increase in SPP level. Unlike treatments with cell-permeable ceramide analogues or sphingomyelinase, which induce apoptosis, 1,25-(OH)2D3 did not induce apoptosis, despite the early formation of ceramide. Moreover, prolonged treatment of HL-60 cells with 1,25-(OH)2D3 suppressed ceramide-induced apoptosis. There was a correlation between the time course and dose response of the activation of sphingosine kinase by 1,25-(OH)2D3 and the protection against apoptosis. In contrast, treatment with all-trans-retinoic acid neither stimulated sphingosine kinase activity nor protected cells from ceramide-induced apoptosis. Treatment with SPP protected HL-60 cells from ceramide-induced apoptosis, and N,N-dimethylsphingosine (DMS), a competitive inhibitor of sphingosine kinase, prevented the survival effect of 1,25-(OH)2D3. The effect of DMS was counteracted by SPP, suggesting that SPP is a critical component of the cytoprotective effect of 1,25-(OH)2D3. Chelerythrine chloride, an inhibitor of protein kinase C, markedly reduced sphingosine kinase activity and the apoptosis-sparing effect of 1,25-(OH)2D3, and conversely, the tumor promoter 12-O-tetradecanoylphorhol-13-acetate not only suppressed ceramide-induced apoptosis but also stimulated sphingosine kinase activity. Moreover, the protective effect of 12-O-tetradecanoylphorbol-13-acetate was blocked by DMS. Collectively, our observations indicate that the cytoprotective effect of 1,25-(OH)2D3 is mediated by SPP, which is formed as a consequence of activation of sphingosine kinase.


Assuntos
Apoptose/efeitos dos fármacos , Calcitriol/farmacologia , Células HL-60/efeitos dos fármacos , Lisofosfolipídeos , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Alcaloides , Benzofenantridinas , Carcinógenos/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Ceramidas/metabolismo , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Células HL-60/enzimologia , Células HL-60/patologia , Humanos , Fenantridinas/farmacologia , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Esfingosina/farmacologia , Acetato de Tetradecanoilforbol/farmacologia , Fatores de Tempo
19.
Biochemistry (Mosc) ; 63(1): 69-73, 1998 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9526097

RESUMO

Recent evidence suggests that branching pathways of sphingolipid metabolism may mediate either apoptotic or mitogenic responses depending on the cell type and the nature of the stimulus. While ceramide has been shown to be an important regulatory component of apoptosis induced by tumor necrosis factor alpha and the Fas ligand, sphingosine-1-phosphate (SPP), a further metabolite of ceramide, has been implicated as a second messenger in cellular proliferation and survival induced by platelet-derived growth factor, neuronal growth factor, and serum. SPP protects cells from apoptosis resulting from elevations of ceramide. Inflammatory cytokines stimulate sphingomyelinase, but not ceramidase, leading to accumulation of ceramide, whereas growth signals also stimulate ceramidase and sphingosine kinase leading to increased SPP levels. We propose that the dynamic balance between levels of sphingolipid metabolites, ceramide, and SPP and consequent regulation of different members of the mitogen-activated protein kinases (JNK versus ERK) family is an important factor that determines whether a cell survives or dies.


Assuntos
Lisofosfolipídeos , Esfingosina/análogos & derivados , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Humanos , Fatores de Crescimento Neural/farmacologia , Sistemas do Segundo Mensageiro , Transdução de Sinais , Esfingosina/farmacologia , Esfingosina/fisiologia
20.
J Biol Chem ; 273(5): 2910-6, 1998 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-9446602

RESUMO

Ceramide, a sphingolipid generated by the hydrolysis of membrane-associated sphingomyelin, appears to play a role as a gauge of apoptosis. A further metabolite of ceramide, sphingosine 1-phosphate (SPP), prevents ceramide-mediated apoptosis, and it has been suggested that the balance between intracellular ceramide and SPP levels may determine the cell fate (Cuvillier, O., Pirianov, G, Kleuser, B., Vanek, P. G., Coso, O. A., Gutkind, J. S., and Spiegel, S. (1996) Nature 381, 800-803). Here, we investigated the role of SPP and the protein kinase C activator, phorbol ester 12-O-tetradecanoylphorbol-13-acetate (TPA), in the caspase cascade leading to the proteolysis of poly(ADP-ribose) polymerase (PARP) and lamins. In Jurkat T cells, Fas ligation or addition of exogenous C2-ceramide induced activations of caspase-3/CPP32 and caspase-7/Mch3 followed by PARP cleavage, effects that can be blocked either by SPP or TPA. Furthermore, both SPP and TPA inhibit the activation of caspase-6/Mch2 and subsequent lamin B cleavage. Ceramide, in contrast to Fas ligation, did not induce activation of caspase-8/FLICE and neither SPP nor TPA were able to prevent this activation. Thus, SPP, likely generated via protein kinase C-mediated activation of sphingosine kinase, suppresses the apoptotic pathway downstream of FLICE but upstream of the executioner caspases, caspase-3, -6, and -7.


Assuntos
Apoptose/efeitos dos fármacos , Caspases , Cisteína Endopeptidases/efeitos dos fármacos , Lisofosfolipídeos , Esfingosina/análogos & derivados , Linfócitos T/efeitos dos fármacos , Caspase 3 , Caspase 6 , Caspase 7 , Cisteína Endopeptidases/metabolismo , Ativação Enzimática/efeitos dos fármacos , Humanos , Células Jurkat , Lamina Tipo B , Laminas , Modelos Biológicos , Proteínas Nucleares/metabolismo , Poli Adenosina Difosfato Ribose/metabolismo , Transdução de Sinais , Esfingosina/metabolismo , Esfingosina/farmacologia , Acetato de Tetradecanoilforbol/farmacologia , Receptor fas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA