Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
1.
Arch Pediatr ; 24(4): 360-362, 2017 Apr.
Artículo en Francés | MEDLINE | ID: mdl-28242153

RESUMEN

Papillon-Lefèvre syndrome (PLS) is a rare primary immunodeficiency, which combines severe periodontal disease with edentulism and palmoplantar keratosis (PPK). PLS is inherited as an autosomal recessive trait and is due to mutations in the cathepsin C gene. The biological properties of the neutrophils (PN) are altered, leading to a gingival dysbiosis and bacterial overgrowth, with intense inflammation of the periodontium. We report the observation of a 4-year-old girl who presented to the clinic with gingivitis, partial edentulism, and PPK, whose diagnosis, raised after a long delay, was suggested by null cathepsin C activity and confirmed by the presence of heterozygous mutations in exon 4: c.628C>T, pArg210* and in exon 7: c.1286G>A, p.Trp429*. A multidisciplinary approach transformed the functional and esthetic prognosis and psychological behavior of this child. This classical observation describes this poorly known phenotype.


Asunto(s)
Enfermedad de Papillon-Lefevre/diagnóstico , Catepsina C/genética , Preescolar , Terapia Combinada , Análisis Mutacional de ADN , Diagnóstico Tardío , Exones , Femenino , Tamización de Portadores Genéticos , Humanos , Comunicación Interdisciplinaria , Colaboración Intersectorial , Enfermedad de Papillon-Lefevre/genética , Enfermedad de Papillon-Lefevre/terapia , Grupo de Atención al Paciente , Fenotipo , Pronóstico
3.
Oncogene ; 34(8): 996-1005, 2015 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-24632610

RESUMEN

Epithelial-to-mesenchymal transition (EMT) promotes cell motility, which is important for the metastasis of malignant cells, and blocks CD95-mediated apoptotic signaling triggered by immune cells and chemotherapeutic regimens. CD95L, the cognate ligand of CD95, can be cleaved by metalloproteases and released as a soluble molecule (cl-CD95L). Unlike transmembrane CD95L, cl-CD95L does not induce apoptosis but triggers cell motility. Electron paramagnetic resonance was used to show that EMT and cl-CD95L treatment both led to augmentation of plasma membrane fluidity that was instrumental in inducing cell migration. Compaction of the plasma membrane is modulated, among other factors, by the ratio of certain lipids such as sphingolipids in the membrane. An integrative analysis of gene expression in NCI tumor cell lines revealed that expression of ceramide synthase-6 (CerS6) decreased during EMT. Furthermore, pharmacological and genetic approaches established that modulation of CerS6 expression/activity in cancer cells altered the level of C16-ceramide, which in turn influenced plasma membrane fluidity and cell motility. Therefore, this study identifies CerS6 as a novel EMT-regulated gene that has a pivotal role in the regulation of cell migration.


Asunto(s)
Membrana Celular/fisiología , Movimiento Celular/genética , Transición Epitelial-Mesenquimal/genética , Fluidez de la Membrana/genética , Proteínas de la Membrana/genética , Neoplasias/patología , Esfingosina N-Aciltransferasa/genética , Células Cultivadas , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Células HL-60 , Humanos , Células Jurkat , Células K562
4.
Rev Neurol (Paris) ; 170(6-7): 445-53, 2014.
Artículo en Francés | MEDLINE | ID: mdl-24746394

RESUMEN

INTRODUCTION: Cerebrotendinous xanthomatosis, a metabolic leukodystrophy with an autosomal recessive inheritance, is secondary to deficiency of sterol 27-hydroxylase, an enzyme involved in cholesterol catabolism. Classical symptoms include clinical or infraclinical xanthomas affecting the skin and tendons, early cataracts, neurological signs and diarrhea. Brain imaging reveals involvement of the dentate nuclei and periventricular white matter hyperintensities. The diagnosis is based on an increased cholestanol level in serum, confirmed by the presence of a mutation in the CYP27A1 gene. Treatment is based on chenodeoxycholic acid. METHOD: We report a retrospective multicentric study of 15 cases of cerebrotendinous xanthomatosis diagnosed in French adults. Clinical, molecular and MRI findings were recorded in all patients. RESULTS: The average age at diagnosis was 39years (range 27-65). Disease onset occurred in childhood in 73% of patients and in adulthood in 27%. All patients with a pediatric onset were diagnosed during adulthood (age range 28-65years). Clinical symptoms variably associated cerebellar syndrome, pyramidal syndrome, cognitive decline, epilepsy, neuropathy (sought in 10 of our patients, present in forms in 8), psychiatric disorders, cataract and xanthomas. One patient had an atypical presentation: monoparesis associated with xanthomas. Brain MRI was abnormal in all: findings consisted in T2-weighted hyperintensity of the dentate nuclei (47%), periventricular leuoencephalopathy (73%) which preferentially involved the posterior cerebral part (60%), leucoencephalopathy with a vascular pattern (7%), hyperintensity of the cortico-spinal tracts (53%), globi pallidi, corpus callosum and cerebral atrophy (33%). Serum cholestanol was elevated in 93% of patients. The most frequent mutation was 1183C>T (n=5/15). Under treatment with chenodeoxycholic acid, eight patients improved initially, followed by stabilization in five of them, and worsening in the others. Four patients died. CONCLUSION: Patients with the xanthoma-neurological disorder association should be tested for cerebrotendinous xanthomatosis. The disease often begins in childhood with a diagnostic delay but also in adulthood. Involvement of the dentate nuclei is specific but not sensitive and the supratentorial leucoencephalopathy is not specific but with an antero-posterior gradient. A vascular distribution and involvement of the corpus callosum are possible. Serum cholestanol assay is very reliable: an elevated level provides the diagnosis, which must nevertheless be confirmed by molecular biology.


Asunto(s)
Xantomatosis Cerebrotendinosa , Adulto , Edad de Inicio , Anciano , Sustitución de Aminoácidos , Encéfalo/patología , Ácido Quenodesoxicólico/uso terapéutico , Colestanotriol 26-Monooxigenasa/deficiencia , Colestanotriol 26-Monooxigenasa/genética , Femenino , Genes Recesivos , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Mutación Missense , Estudios Retrospectivos , Evaluación de Síntomas , Xantomatosis Cerebrotendinosa/tratamiento farmacológico , Xantomatosis Cerebrotendinosa/epidemiología , Xantomatosis Cerebrotendinosa/patología
5.
Oncogene ; 33(26): 3364-73, 2014 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-23893239

RESUMEN

Despite progress in the understanding of the biology and genetics of melanoma, no effective treatment against this cancer is available. The adjacent microenvironment has an important role in melanoma progression. Defining the molecular signals that control the bidirectional dialog between malignant cells and the surrounding stroma is crucial for efficient targeted therapy. Our study aimed at defining the role of sphingosine-1-phosphate (S1P) in melanoma-stroma interactions. Transcriptomic analysis of human melanoma cell lines showed increased expression of sphingosine kinase-1 (SPHK1), the enzyme that produces S1P, as compared with normal melanocytes. Such an increase was also observed by immunohistochemistry in melanoma specimens as compared with nevi, and occurred downstream of ERK activation because of BRAF or NRAS mutations. Importantly, migration of melanoma cells was not affected by changes in SPHK1 activity in tumor cells, but was stimulated by comparable modifications of S1P-metabolizing enzymes in cocultured dermal fibroblasts. Reciprocally, incubation of fibroblasts with the conditioned medium from SPHK1-expressing melanoma cells resulted in their differentiation to myofibroblasts, increased production of matrix metalloproteinases and enhanced SPHK1 expression and activity. In vivo tumorigenesis experiments showed that the lack of S1P in the microenvironment prevented the development of orthotopically injected melanoma cells. Finally, local tumor growth and dissemination were enhanced more efficiently by coinjection of wild-type skin fibroblasts than by fibroblasts from Sphk1(-/-) mice. This report is the first to document that SPHK1/S1P modulates the communication between melanoma cells and dermal fibroblasts. Altogether, our findings highlight SPHK1 as a potential therapeutic target in melanoma progression.


Asunto(s)
Fibroblastos/patología , Melanoma/patología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Neoplasias Cutáneas/patología , Aldehído-Liasas/genética , Animales , Comunicación Celular/genética , Diferenciación Celular/genética , Movimiento Celular , Transformación Celular Neoplásica/genética , Quinasas MAP Reguladas por Señal Extracelular , Femenino , Fibroblastos/enzimología , GTP Fosfohidrolasas/genética , Perfilación de la Expresión Génica , Células HEK293 , Humanos , Lisofosfolípidos/metabolismo , Metaloproteinasa 2 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/biosíntesis , Melanoma/enzimología , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miofibroblastos/enzimología , Miofibroblastos/patología , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Proteínas Proto-Oncogénicas B-raf/genética , Interferencia de ARN , ARN Mensajero/biosíntesis , ARN Interferente Pequeño , Neoplasias Cutáneas/enzimología , Esfingosina/análogos & derivados , Esfingosina/metabolismo
6.
Leukemia ; 27(11): 2129-38, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23568147

RESUMEN

Previous reports demonstrate that metformin, an anti-diabetic drug, can decrease the risk of cancer and inhibit cancer cell growth. However, its mechanism in cancer cells is still unknown. Metformin significantly blocks cell cycle and inhibits cell proliferation and colony formation of leukemic cells. However, the apoptotic response to metformin varies. Furthermore, daily treatment with metformin induces apoptosis and reduces tumor growth in vivo. While metformin induces early and transient activation of AMPK, inhibition of AMPKα1/2 does not abrogate anti-proliferative or pro-apoptotic effects of metformin. Metformin decreases electron transport chain complex I activity, oxygen consumption and mitochondrial ATP synthesis, while stimulating glycolysis for ATP and lactate production, pentose phosphate pathway for purine biosynthesis, fatty acid metabolism, as well as anaplerotic and mitochondrial gene expression. Importantly, leukemic cells with high basal AKT phosphorylation, glucose consumption or glycolysis exhibit a markedly reduced induction of the Pasteur effect in response to metformin and are resistant to metformin-induced apoptosis. Accordingly, glucose starvation or treatment with deoxyglucose or an AKT inhibitor induces sensitivity to metformin. Overall, metformin elicits reprogramming of intermediary metabolism leading to inhibition of cell proliferation in all leukemic cells and apoptosis only in leukemic cells responding to metformin with AKT phosphorylation and a strong Pasteur effect.


Asunto(s)
Apoptosis/efectos de los fármacos , Hipoglucemiantes/farmacología , Leucemia/tratamiento farmacológico , Leucemia/patología , Metformina/farmacología , Mitocondrias/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Western Blotting , Proliferación Celular/efectos de los fármacos , Cromatografía Liquida , Glucosa/metabolismo , Glucólisis/efectos de los fármacos , Humanos , Técnicas para Inmunoenzimas , Ácido Láctico/metabolismo , Leucemia/metabolismo , Ratones , Ratones Desnudos , Mitocondrias/efectos de los fármacos , Consumo de Oxígeno/efectos de los fármacos , Fosforilación/efectos de los fármacos , ARN Interferente Pequeño/genética , Espectrometría de Masa por Ionización de Electrospray , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Cell Death Differ ; 17(4): 642-54, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19779494

RESUMEN

Ceramide can be converted into sphingomyelin by sphingomyelin synthases (SMS) 1 and 2. In this study, we show that in human leukemia Jurkat cells, which express mainly SMS1, Fas ligand (FasL) treatment inhibited SMS activity in a dose- and time-dependent manner before nuclear fragmentation. The SMS inhibition elicited by FasL (1) was abrogated by benzyloxycarbonyl valyl-alanyl-aspartyl-(O-methyl)-fluoromethylketone (zVAD-fmk), a broad-spectrum caspase inhibitor; (2) did not occur in caspase-8-deficient cells and (3) was not affected in caspase-9-deficient cells. Western blot experiments showed SMS1 cleavage in a caspase-dependent manner upon FasL treatment. In a cell-free system, caspase-2, -7, -8 and -9, but not caspase-3 and -10, cleaved SMS1. In HeLa cells, SMS1 was Golgi localized and relocated throughout the cytoplasm in cells exhibiting an early apoptotic phenotype on FasL treatment. zVAD-fmk prevented FasL-induced SMS1 relocation. Thus, FasL-mediated SMS1 inhibition and relocation depend on caspase activation and likely represent proximal events in Fas signaling. FasL-induced ceramide production and cell death were enhanced in cells stably expressing an siRNA against SMS1. Conversely, in cells stably overexpressing SMS1, FasL neither increased ceramide generation nor efficiently induced cell death. Altogether, our data show that SMS1 is a novel caspase target that is functionally involved in the regulation of FasL-induced apoptosis.


Asunto(s)
Apoptosis/fisiología , Caspasas/metabolismo , Proteína Ligando Fas/metabolismo , Leucemia/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Esfingomielinas/metabolismo , Transferasas (Grupos de Otros Fosfatos Sustitutos)/metabolismo , Clorometilcetonas de Aminoácidos/farmacología , Ceramidas/metabolismo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Proteína Ligando Fas/farmacología , Aparato de Golgi/enzimología , Células HeLa , Humanos , Células Jurkat , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética , Interferencia de ARN/fisiología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal/fisiología , Transferasas (Grupos de Otros Fosfatos Sustitutos)/antagonistas & inhibidores , Transferasas (Grupos de Otros Fosfatos Sustitutos)/genética
8.
Leukemia ; 20(3): 392-9, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16397504

RESUMEN

Ceramide metabolism has emerged as a potential target for anticancer therapy. Here, the potential usefulness of two novel synthetic ceramide analogs as anti-leukemic drugs was investigated. Compounds AD2646 and AD2687 were able to dose-and time-dependently decrease the viability of Jurkat leukemic cells. This was accompanied by an accumulation of endogenous ceramide owing to perturbed ceramide metabolism. Cytotoxicity involved caspase activation but also necrotic-like features, as evidenced by phosphatidylserine externalization, membrane permeability, hypodiploidy, caspase processing and only partial protection from cell death by a pan-caspase inhibitor. Ceramide analogs also induced cell death in Jurkat mutants that are deficient in cell death signaling proteins, including FADD, caspase-8 and 10, and RIP. While overexpression of Bcl-xL did not suppress ceramide accumulation, it conferred robust protection from caspase activation and cell death. Altogether, these novel ceramide analogs are able to kill leukemic cells through distinct pathways implicating caspase activation and mitochondrial events, and represent a new group of bioactive molecules with potential applications in anticancer therapy.


Asunto(s)
Caspasas/metabolismo , Muerte Celular/fisiología , Ceramidas/farmacología , Leucemia de Células T/patología , Citometría de Flujo , Humanos , Células Jurkat , Leucemia de Células T/enzimología , Proteína bcl-X/fisiología
9.
Gene Ther ; 13(5): 440-8, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16251995

RESUMEN

Posterior capsule opacification (PCO) is a common complication of cataract surgery. Using adenovirus(Ad)-mediated gene transfer, we overexpressed the proapoptotic molecules p53, procaspase 3, Bax, and TRAIL to induce therapeutic programmed cell death of residual lens cells to prevent PCO. Overexpressed TRAIL did not induce apoptosis in cultured rabbit lens cells or in human lens cells. Overexpressed p53 induced apoptosis of lens cells in vitro and ex vivo, but was unable to prevent PCO in vivo. Overexpressed procaspase 3 was associated with engagement of many components of the apoptotic pathway, including cleavage of intracellular caspase targets such as PARP and inter-nucleosome DNA fragmentation. Even when only slightly overexpressed, Bax caused apoptosis of transduced rabbit and human lens cells by engaging the mitochondrial pathway, including catalytic activation of the caspases. A single in vivo injection of Ad vectors expressing either Bax or procaspase 3 into the capsular bag at the end of phacoemulsification prevented PCO in rabbits. These experiments show that Ad-mediated Bax or procaspase 3 overexpression is capable of inducing therapeutic programmed cell death in vitro and in vivo in residual lens cells and preventing PCO in a rabbit model of PCO. Manipulation of proapoptotic molecule expression could be a novel gene therapy approach for prevention of PCO.


Asunto(s)
Terapia Genética/métodos , Cápsula del Cristalino/patología , Facoemulsificación , Complicaciones Posoperatorias/terapia , Adenoviridae/genética , Animales , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/genética , Caspasa 3 , Caspasas/genética , Línea Celular Transformada , Proliferación Celular , Células Epiteliales/patología , Epitelio Corneal/patología , Regulación de la Expresión Génica , Genes p53 , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Humanos , Glicoproteínas de Membrana/genética , Complicaciones Posoperatorias/patología , Conejos , Ligando Inductor de Apoptosis Relacionado con TNF , Transducción Genética/métodos , Factor de Necrosis Tumoral alfa/genética , Proteína X Asociada a bcl-2/genética
10.
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
11.
Int J Cancer ; 94(2): 157-65, 2001 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-11668492

RESUMEN

Multidrug-resistant (MDR) cancer cells have been shown to have an accumulation of glucosylceramide (GlcCer). In this study, we aim at localizing, at subcellular level, where these lipids accumulate. Neutral lipids and phospholipid containing organelles have been identified using confocal fluorescence microscopy and microspectrofluorometry by monitoring the emission of the fluorescent probe Nile-red. Data from confocal fluorescence microscopy analysis shows accumulation of neutral lipids in cytoplasmic droplets of MDR human carcinoma MCF7R cells. Microspectrofluorometric measurements show an increase of the gold-yellow emission intensity in MCF7R cells, corresponding to neutral lipids. Similar observations were made in human MDR vincristine-HL60 and doxorubicin-KB selected cells. Total cellular glucosylceramide (GlcCer) measurements using [(3)H]-palmitic acid and thin layer chromatography show a significant increase of GlcCer in MCF7R cells. Moreover, MCF7R cells treated with fluorescent GlcCer-bodipy exhibit an accumulation of this lipid in cytoplasmic droplets. Treatment of MCF7R cells with 1-phenyl-2-palmitoylamino-3-morpholino-1-propanolol (PPMP), a potent inhibitor of GlcCer synthase, attenuates the Nile-red fluorescence emission emanating from these structures and reverses MDR. Moreover, Golgi compartments stained with fluorescent PPMP-bodipy, show an increase in the Golgi compartments density. Treatment of MCF7R cells with cyclosporine A (CSA), tamoxifen (TMX) and 3'-azido-3'deoxythymidine (AZT) leads to the same effect observed in the presence of PPMP. Treatment of MCF7 and MCF7R with the beta-glucosidase inhibitor conduritol beta-epoxide (CBE) significantly increases resistance to daunorubicin only in MCF7R cells. These data demonstrate also that: (i) CSA, an inhibitor of MDR, has an additional target in addition to P-glycoprotein; and (ii) TMX (used in breast cancer treatment and prevention) and AZT (used in the treatment of HIV) could have side effects by disturbing lipid metabolism and inhibiting many cellular functions required in normal cells.


Asunto(s)
Citoplasma/metabolismo , Glucosilceramidas/metabolismo , Neoplasias/metabolismo , Núcleo Celular/metabolismo , Daunorrubicina/farmacocinética , Daunorrubicina/farmacología , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Glucosilceramidas/análisis , Humanos , Inositol/análogos & derivados , Inositol/farmacología , Microscopía Confocal , Neoplasias/tratamiento farmacológico , Células Tumorales Cultivadas
12.
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
13.
Mol Pharmacol ; 60(3): 488-96, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11502879

RESUMEN

Reduced glutathione and N-acetylcysteine can inhibit both apoptosis and necrosis of several cell types, suggesting a critical role for reactive oxygen species (ROS) in cell death. However, how the cellular defense against oxidative stress is connected with other cell death mediators remains unclear. We selectively investigated the interaction of seleno-glutathione peroxidase-1 (GPx-1), the major enzyme responsible for peroxide detoxification in mammalian cells, with the cytotoxic response of T47D human breast cancer cells to doxorubicin, an anticancer drug known to promote production of ROS and apoptotic mediator ceramide. The sensitivity to doxorubicin-mediated cell death was compared in T47D/H3 containing low levels of endogenous GPx and T47D/GPx2 transfectant cells, which overexpress GPx-1. We show that T47D/GPx2 cells were significantly more resistant than T47D/H3 cells to doxorubicin (1 microM). The glutathione precursor, N-acetylcysteine also partially protected T47D/H3 cells from the lethal effect of doxorubicin, whereas L-buthionine-(S,R)-sulfoximine, an inhibitor of glutathione biosynthesis, sensitized both GPx-1--deficient and -proficient cells. Interestingly, in addition to a decrease in ROS production, the activation of neutral sphingomyelinase, sphingomyelin hydrolysis, and ceramide generation in response to doxorubicin was impaired in T47D/GPx2 cells compared with control cells. In contrast, GPx overexpression did not protect breast cancer cells from cell death induced by exogenous cell-permeant ceramide. Moreover, the basal activity of neutral sphingomyelinase was considerably lower in T47D/GPx2. Taken together, these results indicate that GPx-1 can regulate doxorubicin-induced cell death signaling at least in part by interfering with the activation of the sphingomyelin-ceramide pathway.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis , Ceramidas/metabolismo , Doxorrubicina/farmacología , Glutatión Peroxidasa/metabolismo , Acetilcisteína/farmacología , Antimetabolitos Antineoplásicos/farmacología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Butionina Sulfoximina/farmacología , Interacciones Farmacológicas , Depuradores de Radicales Libres/farmacología , Humanos , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Transfección , Células Tumorales Cultivadas , Glutatión Peroxidasa GPX1
14.
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
15.
Mol Pharmacol ; 59(5): 955-9, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11306675

RESUMEN

Cannabinoids exert most of their effects through the CB(1) receptor. This G protein-coupled receptor signals inhibition of adenylyl cyclase, modulation of ion channels, and stimulation of mitogen- and stress-activated protein kinases. In this article, we report that Delta(9)-tetrahydrocannabinol (THC), the major active component of marijuana, induces sphingomyelin hydrolysis in primary astrocytes but not in other cells expressing the CB(1) receptor, such as primary neurons, U373 MG astrocytoma cells, and Chinese hamster ovary cells transfected with the CB(1) receptor cDNA. THC-evoked sphingomyelin breakdown in astrocytes was also exerted by the endogenous cannabinoid anandamide and the synthetic cannabinoid HU-210 and was prevented by the selective CB(1) antagonist SR141716. By contrast, the effect of THC was not blocked by pertussis toxin, pointing to a lack of involvement of G(i/o) proteins. A role for the adaptor protein FAN in CB(1) receptor-coupled sphingomyelin breakdown is supported by two observations: 1) coimmunoprecipitation experiments show that the binding of FAN to the CB(1) receptor is enhanced by THC and prevented by SR141716; 2) cells expressing a dominant-negative form of FAN are refractory to THC-induced sphingomyelin breakdown. This is the first report showing that a G-protein-coupled receptor induces sphingomyelin hydrolysis through FAN and that the CB(1) cannabinoid receptor may signal independently of G(i/o) proteins.


Asunto(s)
Astrocitos/metabolismo , Proteínas/metabolismo , Receptores de Droga/metabolismo , Esfingomielinas/metabolismo , Toxina de Adenilato Ciclasa , Animales , Astrocitos/efectos de los fármacos , Cannabinoides/metabolismo , Células Cultivadas , Hidrólisis , Péptidos y Proteínas de Señalización Intracelular , Toxina del Pertussis , Ratas , Receptores de Cannabinoides , Receptores de Droga/efectos de los fármacos , Factores de Virulencia de Bordetella/farmacología
16.
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
17.
Biochem J ; 355(Pt 3): 859-68, 2001 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-11311151

RESUMEN

Sphingomyelinases hydrolyse sphingomyelin to ceramide, a process involved in signal-transduction routes leading to apoptosis and various other cellular responses. In the present study, we investigated the sphingomyelinase content of caveolae, invaginated plasma-membrane microdomains that contain a variety of signalling molecules. These structures are highly enriched in sphingomyelin as well as in ceramide, which suggests that metabolism of these lipids might, to some extent, occur locally. By cell fractionation, we demonstrate that, in addition to a previously reported minute amount of acidic sphingomyelinase activity, a substantial amount of neutral sphingomyelinase activity resides in caveolae of human skin fibroblasts. This caveolar neutral sphingomyelinase activity was also detected in Niemann-Pick disease type A fibroblasts, which are completely devoid of functional acidic sphingomyelinase. Neutral (but not acidic) sphingomyelinase activity was specifically inhibited by a peptide that corresponds to the scaffolding domain of caveolin, which suggests a direct molecular interaction between the two proteins. In addition, this finding implies a cytosolic orientation of the caveolar neutral sphingomyelinase. Interestingly, stimulation of fibroblasts with tumour necrosis factor alpha (TNFalpha) resulted in a partial shift of its p55 receptor to caveolin-enriched membrane fractions and the appearance of caveolin-sensitive neutral sphingomyelinase activity in the non-caveolar fractions. These results suggest that (part of) the presently identified caveolar neutral sphingomyelinase activity is involved in TNFalpha signalling.


Asunto(s)
Caveolinas/farmacología , Transducción de Señal/fisiología , Esfingomielina Fosfodiesterasa/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Caveolas/metabolismo , Caveolinas/química , Células Cultivadas , Colesterol/deficiencia , Colesterol/metabolismo , Activación Enzimática/efectos de los fármacos , Humanos , Enfermedades de Niemann-Pick/patología , Estructura Terciaria de Proteína , Esfingolípidos/metabolismo , Esfingomielina Fosfodiesterasa/antagonistas & inhibidores
18.
Proc Natl Acad Sci U S A ; 98(6): 3428-33, 2001 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-11248095

RESUMEN

Fabry disease is a lipid storage disorder resulting from mutations in the gene encoding the enzyme alpha-galactosidase A (alpha-gal A; EC ). We previously have demonstrated long-term alpha-gal A enzyme correction and lipid reduction mediated by therapeutic ex vivo transduction and transplantation of hematopoietic cells in a mouse model of Fabry disease. We now report marked improvement in the efficiency of this gene-therapy approach. For this study we used a novel bicistronic retroviral vector that engineers expression of both the therapeutic alpha-gal A gene and the human IL-2Ralpha chain (huCD25) gene as a selectable marker. Coexpression of huCD25 allowed selective immunoenrichment (preselection) of a variety of transduced human and murine cells, resulting in enhanced intracellular and secreted alpha-gal A enzyme activities. Of particular significance for clinical applicability, mobilized CD34(+) peripheral blood hematopoietic stem/progenitor cells from Fabry patients have low-background huCD25 expression and could be enriched effectively after ex vivo transduction, resulting in increased alpha-gal A activity. We evaluated effects of preselection in the mouse model of Fabry disease. Preselection of transduced Fabry mouse bone marrow cells elevated the level of multilineage gene-corrected hematopoietic cells in the circulation of transplanted animals and improved in vivo enzymatic activity levels in plasma and organs for more than 6 months after both primary and secondary transplantation. These studies demonstrate the potential of using a huCD25-based preselection strategy to enhance the clinical utility of ex vivo hematopoietic stem/progenitor cell gene therapy of Fabry disease and other disorders.


Asunto(s)
Enfermedad de Fabry/terapia , Terapia Genética/métodos , Receptores de Interleucina-2/genética , alfa-Galactosidasa/genética , Células 3T3 , Animales , Trasplante de Médula Ósea , Modelos Animales de Enfermedad , Enfermedad de Fabry/metabolismo , Enfermedad de Fabry/patología , Expresión Génica , Vectores Genéticos , Células HeLa , Humanos , Ratones , Ratones Endogámicos C57BL , Receptores de Interleucina-2/metabolismo , Retroviridae/genética , alfa-Galactosidasa/metabolismo
19.
Biochem Biophys Res Commun ; 281(2): 289-94, 2001 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-11181043

RESUMEN

Exposure of human keratinocytes to UVA radiation induced an increase in ceramide (CER) intracellular content, with a dose-dependent effect within the range of 4-9 J/cm(2). The production of CER reached a maximum 2 h after UVA irradiation. The increase of CER was proportional to the intracellular content of reactive oxygen species, was prevented by the antioxidant vitamin E, and enhanced by the prooxidant buthionine-sulfoximine, suggesting the involvement of an oxidative stress. UVA decreased both neutral and acid sphingomyelinase activities measured in vitro. A direct cleavage of sphingomyelin to CER by UVA, recently described, was not observed under our experimental conditions. We also show that, downstream of CER, UVA activated the Ser/Thr kinases ERK, JNK, and p38. Since ceramide has been shown to play a role in stress kinase activation, our results provide a possible mechanism for UVA-induced activation of stress kinases via ceramide formation. However, the actual mechanisms whereby CER is produced in cultured cells under UVA exposure remain to be specified.


Asunto(s)
Ceramidas/efectos de la radiación , Proteínas Quinasas JNK Activadas por Mitógenos , Rayos Ultravioleta , Línea Celular , Ceramidas/metabolismo , Relación Dosis-Respuesta en la Radiación , Activación Enzimática/efectos de la radiación , Humanos , MAP Quinasa Quinasa 4 , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Esfingomielina Fosfodiesterasa/metabolismo , Esfingomielinas/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA