Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Osteoarthritis Cartilage ; 29(9): 1335-1345, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34144150

RESUMEN

OBJECTIVE: Cartilage loss observed in osteoarthritis (OA) is prevented when osteoclasts in the subchondral bone are inhibited in mice. Here, we investigated the role of the osteoclast secretome and of the lipid mediator sphingosine 1-phosphate (S1P) in chondrocyte metabolism and OA. MATERIALS AND METHODS: We used SphK1LysMCre and wild type mice to assess the effect of murine osteoclast secretome in chondrocyte metabolism. Gene and protein expressions of matrix metalloproteinase (Mmp) were quantified in chondrocytes and explants by RT-qPCR and Western blots. SphK1LysMCre mice or wild type mice treated with S1P2 receptor inhibitor JTE013 or anti-S1P neutralizing antibody sphingomab are analyzed by OA score and immunohistochemistry. RESULTS: The osteoclast secretome increased the expression of Mmp3 and Mmp13 in murine chondrocytes and cartilage explants and activated the JNK signaling pathway, which led to matrix degradation. JTE013 reversed the osteoclast-mediated chondrocyte catabolism and protected mice against OA, suggesting that osteoclastic S1P contributes to cartilage damage in OA via S1P/S1P2 signaling. The activity of sphingosine kinase 1 (SphK1) increased with osteoclast differentiation, and its expression was enhanced in subchondral bone of mice with OA. The expression of Mmp3 and Mmp13 in chondrocytes was low upon stimulation with the secretome of Sphk1-lacking osteoclasts. Cartilage damage was significantly reduced in SphK1LysMCre mice, but not the synovial inflammation. Finally, intra-articular administration of sphingomab inhibited the cartilage damage and synovial inflammation. CONCLUSIONS: Lack of S1P in myeloid cells and local S1P neutralization alleviates from osteoarthritis in mice. These data identify S1P as a therapeutic target in OA.


Asunto(s)
Condrocitos/metabolismo , Lisofosfolípidos/antagonistas & inhibidores , Osteoartritis/metabolismo , Osteoartritis/prevención & control , Osteoclastos/metabolismo , Secretoma/metabolismo , Esfingosina/análogos & derivados , Animales , Masculino , Ratones , Esfingosina/antagonistas & inhibidores
3.
Oncogenesis ; 5: e209, 2016 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-26974204

RESUMEN

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.

4.
Prog Urol ; 26(3): 159-67, 2016 Mar.
Artículo en Francés | MEDLINE | ID: mdl-26896427

RESUMEN

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.


Asunto(s)
Neoplasias de la Próstata Resistentes a la Castración/patología , Receptores Androgénicos/biosíntesis , Hipoxia Tumoral , Microambiente Tumoral , Humanos , Masculino , Esferoides Celulares , Células Tumorales Cultivadas
5.
Leukemia ; 22(5): 971-9, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18401414

RESUMEN

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.


Asunto(s)
Apoptosis , Proteínas de Fusión bcr-abl/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Fosfotransferasas (Aceptor de Grupo Alcohol)/fisiología , Piperazinas/farmacología , Pirimidinas/farmacología , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis , Benzamidas , Benzoquinonas/farmacología , Resistencia a Antineoplásicos , Humanos , Mesilato de Imatinib , Leucemia Mielógena Crónica BCR-ABL Positiva/enzimología , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Transducción de Señal
6.
Cell Death Differ ; 14(2): 197-208, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16645635

RESUMEN

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.


Asunto(s)
Apoptosis , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Receptores de Somatostatina/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Somatostatina/metabolismo , Animales , Apoptosis/efectos de los fármacos , Caspasas/metabolismo , Activación Enzimática/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Ratones , Modelos Biológicos , Imitación Molecular/efectos de los fármacos , Células 3T3 NIH , Fosforilación/efectos de los fármacos , Proteína Fosfatasa 1 , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Transducción de Señal/efectos de los fármacos , Transfección , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/farmacología
7.
Leukemia ; 20(1): 95-102, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16281067

RESUMEN

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.


Asunto(s)
Resistencia a Múltiples Medicamentos , Leucemia Mieloide/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Enfermedad Aguda , Apoptosis/efectos de los fármacos , Benzoquinonas/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ceramidas/biosíntesis , Doxorrubicina/farmacología , Resistencia a Antineoplásicos , Etopósido/farmacología , Células HL-60 , Humanos , Leucemia Mieloide/tratamiento farmacológico , Mitocondrias/efectos de los fármacos , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/farmacología , Interferencia de ARN/fisiología , Receptores de Lisoesfingolípidos/metabolismo
8.
Circ Res ; 89(11): 957-68, 2001 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-11717151

RESUMEN

Sphingolipids have emerged as a new class of lipid mediators. In response to various extracellular stimuli, sphingolipid turnover can be stimulated in vascular cells and cardiac myocytes. Subsequent generation of sphingolipid molecules such as ceramide, sphingosine, and sphingosine-1-phosphate, is followed by regulation of ion fluxes and activation of various signaling pathways leading to smooth muscle cell proliferation, endothelial cell differentiation or apoptotic cell death, cell contraction, retraction, or migration. The importance of sphingolipids in cardiovascular signaling is illustrated by recent observations implicating them in physiological processes such as vasculogenesis as well as in frequent pathological conditions, including atherosclerosis and its complications.


Asunto(s)
Cardiopatías/etiología , Miocardio/metabolismo , Esfingolípidos/fisiología , Animales , Apoptosis , División Celular , Enfermedad de la Arteria Coronaria/etiología , Humanos , Transporte Iónico , Contracción Miocárdica , Daño por Reperfusión Miocárdica/etiología , Miocardio/citología , Neovascularización Fisiológica , Traumatismos por Radiación/etiología , Transducción de Señal , Esfingolípidos/química
9.
Blood ; 98(9): 2828-36, 2001 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-11675357

RESUMEN

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.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteínas Portadoras/antagonistas & inhibidores , Grupo Citocromo c/antagonistas & inhibidores , Leucemia/patología , Lisofosfolípidos , Mitocondrias/metabolismo , Proteínas Mitocondriales/antagonistas & inhibidores , Esfingosina/análogos & derivados , Esfingosina/farmacología , Proteínas Reguladoras de la Apoptosis , Proteínas Portadoras/efectos de los fármacos , Proteínas Portadoras/metabolismo , Ceramidas/farmacología , Medio de Cultivo Libre de Suero/farmacología , Grupo Citocromo c/efectos de los fármacos , Grupo Citocromo c/metabolismo , Células HL-60 , Humanos , Péptidos y Proteínas de Señalización Intracelular , Células Jurkat , Proteínas Mitocondriales/efectos de los fármacos , Proteínas Mitocondriales/metabolismo , Transporte de Proteínas/efectos de los fármacos , Receptores del Factor de Necrosis Tumoral/fisiología , Acetato de Tetradecanoilforbol/farmacología , Células U937
10.
J Clin Invest ; 108(1): 143-51, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11435466

RESUMEN

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.


Asunto(s)
Apoptosis/fisiología , Proteínas/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Clorometilcetonas de Aminoácidos/farmacología , Animales , Antígenos CD/efectos de los fármacos , Apoptosis/efectos de los fármacos , Caspasas/metabolismo , Línea Celular Transformada , Células Cultivadas/efectos de los fármacos , Ceramidas/biosíntesis , Ceramidas/farmacología , Inhibidores de Cisteína Proteinasa/farmacología , Daunorrubicina/farmacología , Activación Enzimática/efectos de los fármacos , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Genes Dominantes , Humanos , Hidrólisis , Molécula 1 de Adhesión Intercelular/biosíntesis , Molécula 1 de Adhesión Intercelular/genética , Péptidos y Proteínas de Señalización Intracelular , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Ratones Noqueados , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Estructura Terciaria de Proteína , Proteínas/genética , Receptores del Factor de Necrosis Tumoral/efectos de los fármacos , Receptores Tipo I de Factores de Necrosis Tumoral , Proteínas Recombinantes de Fusión/fisiología , Sistemas de Mensajero Secundario , Esfingomielina Fosfodiesterasa/metabolismo , Esfingomielinas/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Factor de Necrosis Tumoral alfa/toxicidad , Células U937
11.
Cell Death Differ ; 8(2): 162-71, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11313718

RESUMEN

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.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Caspasas/metabolismo , Grupo Citocromo c/metabolismo , Doxorrubicina/farmacología , Esfingosina/biosíntesis , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/metabolismo , Antineoplásicos/metabolismo , Apoptosis/fisiología , Neoplasias de la Mama/metabolismo , Caspasa 7 , Doxorrubicina/metabolismo , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Femenino , Humanos , Mitocondrias/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína bcl-X
12.
J Neurochem ; 76(5): 1573-84, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11238741

RESUMEN

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.


Asunto(s)
Apoptosis/fisiología , Caspasas/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proteínas Serina-Treonina Quinasas , Esfingosina/análogos & derivados , Neoplasias de las Glándulas Suprarrenales , Animales , Apoptosis/efectos de los fármacos , Caspasa 2 , Caspasa 3 , Caspasa 7 , Medio de Cultivo Libre de Suero , Proteínas Quinasas JNK Activadas por Mitógenos , Cinética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Factor de Crecimiento Nervioso/farmacología , Células PC12 , Toxina del Pertussis , Feocromocitoma , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Ratas , Proteínas Recombinantes/metabolismo , Esfingosina/farmacología , Transfección , Factores de Virulencia de Bordetella/farmacología
13.
FASEB J ; 15(2): 297-9, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11156942

RESUMEN

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.


Asunto(s)
Apoptosis/fisiología , Fumonisinas , Lisosomas/enzimología , Esfingomielina Fosfodiesterasa/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Ácidos Carboxílicos/farmacología , Caspasa 3 , Caspasas/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Ceramidas/metabolismo , Daunorrubicina/toxicidad , Doxorrubicina/toxicidad , Inhibidores Enzimáticos/farmacología , Humanos , Linfocitos/enzimología , Linfocitos/patología , Linfocitos/fisiología , Enfermedades de Niemann-Pick , Péptido Hidrolasas/metabolismo , Valores de Referencia , Receptor fas/fisiología
14.
Cancer Res ; 60(16): 4468-74, 2000 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-10969794

RESUMEN

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.


Asunto(s)
Apoptosis/efectos de la radiación , Neoplasias de la Próstata/patología , Tolerancia a Radiación/fisiología , Esfingosina/análogos & derivados , Esfingosina/fisiología , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Caspasas/metabolismo , Ceramidas/metabolismo , Ceramidas/fisiología , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Humanos , Masculino , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/fisiología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/radioterapia , Tolerancia a Radiación/efectos de los fármacos , Fármacos Sensibilizantes a Radiaciones/farmacología , Esfingomielina Fosfodiesterasa/farmacología , Esfingosina/metabolismo , Esfingosina/farmacología , Células Tumorales Cultivadas/efectos de la radiación
15.
J Biol Chem ; 275(21): 15691-700, 2000 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-10747891

RESUMEN

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.


Asunto(s)
Apoptosis , Fumonisinas , Mitocondrias/metabolismo , Esfingosina/metabolismo , Receptor fas/metabolismo , Clorometilcetonas de Aminoácidos/farmacología , Anticuerpos Monoclonales/farmacología , Proteína Proapoptótica que Interacciona Mediante Dominios BH3 , Ácidos Carboxílicos/farmacología , Proteínas Portadoras/metabolismo , Caspasas/metabolismo , Ceramidas/metabolismo , Inhibidores de Cisteína Proteinasa/farmacología , Grupo Citocromo c/metabolismo , Activación Enzimática , Regulación de la Expresión Génica , Humanos , Células Jurkat , Oligopéptidos/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Esfingosina/análogos & derivados , Esfingosina/farmacología , Transfección , Proteína bcl-X , Receptor fas/inmunología
16.
Blood ; 94(10): 3583-92, 1999 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-10552970

RESUMEN

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.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis , Grupo Citocromo c/metabolismo , Éteres Fosfolípidos/farmacología , Receptor fas/metabolismo , Transporte Biológico , Caspasas/metabolismo , Grupo Citocromo c/fisiología , Citosol/metabolismo , Relación Dosis-Respuesta a Droga , Portadores de Fármacos , Sistemas de Liberación de Medicamentos , Activación Enzimática , Humanos , Células Jurkat , Laminas , Liposomas , Mitocondrias/metabolismo , Proteínas Nucleares/metabolismo , Péptido Hidrolasas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Transducción de Señal , Factores de Tiempo , Células U937 , Proteína bcl-X
17.
J Cell Biol ; 147(3): 545-58, 1999 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-10545499

RESUMEN

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.


Asunto(s)
División Celular , Supervivencia Celular , Lisofosfolípidos , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Receptores Acoplados a Proteínas G , Sistemas de Mensajero Secundario , Esfingosina/análogos & derivados , Animales , Apoptosis/efectos de los fármacos , Caspasa 3 , Inhibidores de Caspasas , Caspasas/metabolismo , División Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Citosol/metabolismo , ADN/biosíntesis , Activación Enzimática/efectos de los fármacos , Humanos , Ratones , Péptido Hidrolasas/metabolismo , Toxina del Pertussis , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Factor de Crecimiento Derivado de Plaquetas/farmacología , Receptores de Superficie Celular/antagonistas & inhibidores , Receptores de Superficie Celular/metabolismo , Receptores Lisofosfolípidos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Sistemas de Mensajero Secundario/efectos de los fármacos , Esfingosina/antagonistas & inhibidores , Esfingosina/genética , Esfingosina/metabolismo , Esfingosina/farmacología , Transfección , Factores de Virulencia de Bordetella/farmacología , Receptor fas/metabolismo
18.
FEBS Lett ; 454(1-2): 137-41, 1999 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-10413111

RESUMEN

Incorporation of ET-18-OCH3 into well-characterized liposomes known as ELL-12 has eliminated its gastrointestinal and hemolytic toxicity without loss of growth inhibiting activity. ET-18-OCH3, but not ELL-12, blunted the increase in membrane protein kinase C (PKC) activity induced by 12-O-tetradecanoylphorbol 13-myristate (TPA) and markedly reduced levels of PKC alpha in NIH 3T3 fibroblasts. Furthermore, prolonged treatment with ELL-12 neither inhibited TPA-induced translocations of PKC alpha and PKC delta to the particulate fraction nor caused down-regulation, and did not affect the cellular distribution of TPA-insensitive PKC zeta. In Jurkat T cells, where ELL-12 markedly induced apoptosis that was blocked by an inhibitor of caspase-3-like activities, it had no effect on PKC activity or translocation induced by TPA. Thus, it seems unlikely that PKC is involved in the therapeutic effects of ELL-12.


Asunto(s)
Liposomas/metabolismo , Éteres Fosfolípidos/farmacología , Proteína Quinasa C/metabolismo , Células 3T3 , Animales , Apoptosis/efectos de los fármacos , División Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Células Jurkat , Ratones
19.
Biochemistry ; 37(37): 12892-8, 1998 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-9737868

RESUMEN

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.


Asunto(s)
Ceramidas/metabolismo , Inhibidores Enzimáticos/farmacología , Lisofosfolípidos , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Esfingosina/análogos & derivados , Células 3T3 , Animales , Activación Enzimática/efectos de los fármacos , Humanos , Ratones , Células PC12 , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Ratas , Esfingosina/metabolismo , Esfingosina/farmacología , Células Tumorales Cultivadas
20.
J Cell Biol ; 142(1): 229-40, 1998 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-9660876

RESUMEN

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.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Proteínas Inmediatas-Precoces/metabolismo , Lisofosfolípidos , Receptores de Superficie Celular/metabolismo , Receptores Acoplados a Proteínas G , Esfingosina/análogos & derivados , Células 3T3 , Animales , División Celular , Línea Celular , Supervivencia Celular , AMP Cíclico/metabolismo , Células HL-60 , Humanos , Proteínas Inmediatas-Precoces/genética , Ratones , Células PC12 , Ratas , Receptores Lisofosfolípidos , Transducción de Señal , Esfingosina/metabolismo , Esfingosina/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA