Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
1.
Curr Opin Cell Biol ; 8(2): 159-67, 1996 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-8791422

RESUMEN

This review emphasizes the generation of glycerolipid and sphingolipid second messengers, and their molecular targets. The role of the phosphatidylinositol transfer protein and phospholipase D in signal transmission, and the structures of the 1, 2-diacylglycerol and calcium-binding sites of protein kinase C are discussed. Further, ceramide signaling through protein kinases and the role of cross-talk in the signaling of apoptosis and inflammation are addressed.


Asunto(s)
Lípidos/fisiología , Sistemas de Mensajero Secundario/fisiología , Transducción de Señal , Animales , Humanos , Transducción de Señal/fisiología , Esfingolípidos/fisiología
2.
Nat Med ; 9(3): 322-30, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12563314

RESUMEN

Pseudomonas aeruginosa infection is a serious complication in patients with cystic fibrosis and in immunocompromised individuals. Here we show that P. aeruginosa infection triggers activation of the acid sphingomyelinase and the release of ceramide in sphingolipid-rich rafts. Ceramide reorganizes these rafts into larger signaling platforms that are required to internalize P. aeruginosa, induce apoptosis and regulate the cytokine response in infected cells. Failure to generate ceramide-enriched membrane platforms in infected cells results in an unabated inflammatory response, massive release of interleukin (IL)-1 and septic death of mice. Our findings show that ceramide-enriched membrane platforms are central to the host defense against this potentially lethal pathogen.


Asunto(s)
Ceramidas/metabolismo , Microdominios de Membrana/inmunología , Microdominios de Membrana/metabolismo , Infecciones por Pseudomonas/metabolismo , Pseudomonas aeruginosa/fisiología , Esfingomielina Fosfodiesterasa/metabolismo , beta-Ciclodextrinas , Animales , Apoptosis/fisiología , Trasplante de Médula Ósea , Células Cultivadas , Ciclodextrinas/farmacología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Activación Enzimática , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/fisiología , Filipina/farmacología , Colorantes Fluorescentes/metabolismo , Humanos , Ionóforos/farmacología , Microdominios de Membrana/química , Ratones , Nistatina/farmacología , Infecciones por Pseudomonas/inmunología , Transducción de Señal/fisiología , Esfingomielina Fosfodiesterasa/genética , Receptor fas/metabolismo
3.
Nat Med ; 6(10): 1109-14, 2000 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11017141

RESUMEN

The time at which ovarian failure (menopause) occurs in females is determined by the size of the oocyte reserve provided at birth, as well as by the rate at which this endowment is depleted throughout post-natal life. Here we show that disruption of the gene for acid sphingomyelinase in female mice suppressed the normal apoptotic deletion of fetal oocytes, leading to neonatal ovarian hyperplasia. Ex vivo, oocytes lacking the gene for acid sphingomyelinase or wild-type oocytes treated with sphingosine-1-phosphate resisted developmental apoptosis and apoptosis induced by anti-cancer therapy, confirming cell autonomy of the death defect. Moreover, radiation-induced oocyte loss in adult wild-type female mice, the event that drives premature ovarian failure and infertility in female cancer patients, was completely prevented by in vivo therapy with sphingosine-1-phosphate. Thus, the sphingomyelin pathway regulates developmental death of oocytes, and sphingosine-1-phosphate provides a new approach to preserve ovarian function in vivo.


Asunto(s)
Apoptosis/efectos de los fármacos , Oocitos/citología , Oocitos/efectos de los fármacos , Esfingomielina Fosfodiesterasa/genética , Esfingosina/análogos & derivados , Animales , Apoptosis/genética , Apoptosis/efectos de la radiación , Supervivencia Celular/genética , Relación Dosis-Respuesta a Droga , Doxorrubicina/farmacología , Femenino , Lisofosfolípidos/farmacología , Masculino , Ratones , Ratones Mutantes , Oocitos/efectos de la radiación , Esfingomielina Fosfodiesterasa/metabolismo , Esfingomielinas/metabolismo , Esfingosina/farmacología
4.
J Exp Med ; 180(2): 525-35, 1994 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-8046331

RESUMEN

Recent investigations provided evidence that the sphingomyelin signal transduction pathway mediates apoptosis for tumor necrosis factor alpha (TNF-alpha) in several hematopoietic and nonhematopoietic cells. In this pathway, TNF-receptor interaction initiates sphingomyelin hydrolysis to ceramide by a sphingomyelinase. Ceramide acts as a second messenger stimulating a ceramide-activated serine/threonine protein kinase. The present studies show that ionizing radiation, like TNF, induces rapid sphingomyelin hydrolysis to ceramide and apoptosis in bovine aortic endothelial cells. Elevation of ceramide with exogenous ceramide analogues was sufficient for induction of apoptosis. Protein kinase C activation blocked both radiation-induced sphingomyelin hydrolysis and apoptosis, and apoptosis was restored by ceramide analogues added exogenously. Ionizing radiation acted directly on membrane preparations devoid of nuclei, stimulating sphingomyelin hydrolysis enzymatically through a neutral sphingomyelinase. These studies provide the first conclusive evidence that apoptotic signaling can be generated by interaction of ionizing radiation with cellular membranes and suggest an alternative to the hypothesis that direct DNA damage mediates radiation-induced cell kill.


Asunto(s)
Apoptosis/efectos de la radiación , Membrana Celular/efectos de la radiación , Ceramidas/biosíntesis , Radiación Ionizante , Esfingomielinas/metabolismo , Animales , Bovinos , Membrana Celular/metabolismo , Células Cultivadas , Daño del ADN , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/ultraestructura , Humanos , Sistemas de Mensajero Secundario , Transducción de Señal , Esfingomielina Fosfodiesterasa/metabolismo
5.
J Exp Med ; 186(11): 1831-41, 1997 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-9382882

RESUMEN

The endotoxic shock syndrome is characterized by systemic inflammation, multiple organ damage, circulatory collapse and death. Systemic release of tumor necrosis factor (TNF)-alpha and other cytokines purportedly mediates this process. However, the primary tissue target remains unidentified. The present studies provide evidence that endotoxic shock results from disseminated endothelial apoptosis. Injection of lipopolysaccharide (LPS), and its putative effector TNF-alpha, into C57BL/6 mice induced apoptosis in endothelium of intestine, lung, fat and thymus after 6 h, preceding nonendothelial tissue damage. LPS or TNF-alpha injection was followed within 1 h by tissue generation of the pro-apoptotic lipid ceramide. TNF-binding protein, which protects against LPS-induced death, blocked LPS-induced ceramide generation and endothelial apoptosis, suggesting systemic TNF is required for both responses. Acid sphingomyelinase knockout mice displayed a normal increase in serum TNF-alpha in response to LPS, yet were protected against endothelial apoptosis and animal death, defining a role for ceramide in mediating the endotoxic response. Furthermore, intravenous injection of basic fibroblast growth factor, which acts as an intravascular survival factor for endothelial cells, blocked LPS-induced ceramide elevation, endothelial apoptosis and animal death, but did not affect LPS-induced elevation of serum TNF-alpha. These investigations demonstrate that LPS induces a disseminated form of endothelial apoptosis, mediated sequentially by TNF and ceramide generation, and suggest that this cascade is mandatory for evolution of the endotoxic syndrome.


Asunto(s)
Apoptosis/efectos de los fármacos , Ceramidas/biosíntesis , Endotelio Vascular/efectos de los fármacos , Lipopolisacáridos/farmacología , Receptores del Factor de Necrosis Tumoral , Choque Séptico/patología , Factor de Necrosis Tumoral alfa/farmacología , Tejido Adiposo/irrigación sanguínea , Animales , Capilares/efectos de los fármacos , Capilares/patología , Proteínas Portadoras/farmacología , Endotelio Vascular/patología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Mucosa Intestinal/irrigación sanguínea , Lipopolisacáridos/antagonistas & inhibidores , Lipopolisacáridos/toxicidad , Pulmón/irrigación sanguínea , Ratones , Ratones Endogámicos C57BL , Receptores Tipo I de Factores de Necrosis Tumoral , Choque Séptico/inducido químicamente , Transducción de Señal , Organismos Libres de Patógenos Específicos , Esfingomielina Fosfodiesterasa/farmacología , Esfingomielinas/metabolismo , Timo/irrigación sanguínea , Receptores Señuelo del Factor de Necrosis Tumoral
6.
Trends Cell Biol ; 2(8): 232-6, 1992 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-14731480

RESUMEN

Sphingomyelin used to be considered only as a structural molecule of the outer leaflet of the plasma membrane of mammalian cells. However, in the last five years it has become clear that sphingomyelin is rapidly degraded and resynthesized in a pathway that could function in signal transduction. Indeed, it is now known that this pathway plays a role in signal transduction for at least one cytokine, tumour necrosis factor alpha. It seems possible that other cell surface receptors will also utilize this pathway for signalling.

7.
Science ; 255(5052): 1715-8, 1992 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-1313189

RESUMEN

The mechanism of tumor necrosis factor (TNF)-alpha signaling is unknown. TNF-alpha signaling may involve sphingomyelin hydrolysis to ceramide by a sphingomyelinase and stimulation of a ceramide-activated protein kinase. In a cell-free system, TNF-alpha induced a rapid reduction in membrane sphingomyelin content and a quantitative elevation in ceramide concentrations. Ceramide-activated protein kinase activity also increased. Kinase activation was mimicked by addition of sphingomyelinase but not by phospholipases A2, C, or D. Reconstitution of this cascade in a cell-free system demonstrates tight coupling to the receptor, suggesting this is a signal transduction pathway for TNF-alpha.


Asunto(s)
Ceramidas/fisiología , Proteínas Quinasas/metabolismo , Receptores de Superficie Celular/fisiología , Transducción de Señal/efectos de los fármacos , Esfingomielina Fosfodiesterasa/fisiología , Esfingomielinas/fisiología , Factor de Necrosis Tumoral alfa/farmacología , Sistema Libre de Células , Activación Enzimática , Receptores ErbB/metabolismo , Humanos , Técnicas In Vitro , Fosforilación , Receptores del Factor de Necrosis Tumoral , Sistemas de Mensajero Secundario , Células Tumorales Cultivadas
8.
Science ; 259(5094): 519-22, 1993 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-8424175

RESUMEN

The mechanism of interleukin-1 (IL-1) signaling is unknown. Tumor necrosis factor-alpha uses a signal transduction pathway that involves sphingomyelin hydrolysis to ceramide and stimulation of a ceramide-activated protein kinase. In intact EL4 thymoma cells, IL-1 beta similarly stimulated a rapid decrease of sphingomyelin and an elevation of ceramide, and enhanced ceramide-activated protein kinase activity. This cascade was also activated by IL-1 beta in a cell-free system, demonstrating tight coupling to the receptor. Exogenous sphingomyelinase, but not phospholipases A2, C, or D, in combination with phorbol ester replaced IL-1 beta to stimulate IL-2 secretion. Thus, IL-1 beta signals through the sphingomyelin pathway.


Asunto(s)
Ceramidas/metabolismo , Interleucina-1/farmacología , Transducción de Señal/efectos de los fármacos , Esfingomielinas/metabolismo , Secuencia de Aminoácidos , Animales , Sistema Libre de Células , Relación Dosis-Respuesta a Droga , Interleucina-2/biosíntesis , Cinética , Ratones , Datos de Secuencia Molecular , Proteínas Quinasas/metabolismo , Esfingomielina Fosfodiesterasa/farmacología , Especificidad por Sustrato , Timoma , Neoplasias del Timo , Células Tumorales Cultivadas , Fosfolipasas de Tipo C/farmacología
9.
Science ; 293(5528): 293-7, 2001 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-11452123

RESUMEN

Gastrointestinal (GI) tract damage by chemotherapy or radiation limits their efficacy in cancer treatment. Radiation has been postulated to target epithelial stem cells within the crypts of Lieberkühn to initiate the lethal GI syndrome. Here, we show in mouse models that microvascular endothelial apoptosis is the primary lesion leading to stem cell dysfunction. Radiation-induced crypt damage, organ failure, and death from the GI syndrome were prevented when endothelial apoptosis was inhibited pharmacologically by intravenous basic fibroblast growth factor (bFGF) or genetically by deletion of the acid sphingomyelinase gene. Endothelial, but not crypt, cells express FGF receptor transcripts, suggesting that the endothelial lesion occurs before crypt stem cell damage in the evolution of the GI syndrome. This study provides a basis for new approaches to prevent radiation damage to the bowel.


Asunto(s)
Apoptosis , Endotelio Vascular/efectos de la radiación , Mucosa Intestinal/efectos de la radiación , Intestinos/efectos de la radiación , Animales , Anexina A5/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Médula Ósea/efectos de la radiación , Trasplante de Médula Ósea , Capilares , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/patología , Factores de Crecimiento de Fibroblastos/farmacología , Humanos , Etiquetado Corte-Fin in Situ , Mucosa Intestinal/irrigación sanguínea , Mucosa Intestinal/citología , Mucosa Intestinal/patología , Intestinos/irrigación sanguínea , Intestinos/patología , Ratones , Ratones Endogámicos C57BL , Neoplasias/patología , Neoplasias/radioterapia , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Esfingomielina Fosfodiesterasa/deficiencia , Esfingomielina Fosfodiesterasa/genética , Esfingomielina Fosfodiesterasa/metabolismo , Células Madre/efectos de la radiación , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/metabolismo , Irradiación Corporal Total
10.
Sci Immunol ; 4(42)2019 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-31811055

RESUMEN

Despite the importance of intestinal stem cells (ISCs) for epithelial maintenance, there is limited understanding of how immune-mediated damage affects ISCs and their niche. We found that stem cell compartment injury is a shared feature of both alloreactive and autoreactive intestinal immunopathology, reducing ISCs and impairing their recovery in T cell-mediated injury models. Although imaging revealed few T cells near the stem cell compartment in healthy mice, donor T cells infiltrating the intestinal mucosa after allogeneic bone marrow transplantation (BMT) primarily localized to the crypt region lamina propria. Further modeling with ex vivo epithelial cultures indicated ISC depletion and impaired human as well as murine organoid survival upon coculture with activated T cells, and screening of effector pathways identified interferon-γ (IFNγ) as a principal mediator of ISC compartment damage. IFNγ induced JAK1- and STAT1-dependent toxicity, initiating a proapoptotic gene expression program and stem cell death. BMT with IFNγ-deficient donor T cells, with recipients lacking the IFNγ receptor (IFNγR) specifically in the intestinal epithelium, and with pharmacologic inhibition of JAK signaling all resulted in protection of the stem cell compartment. In addition, epithelial cultures with Paneth cell-deficient organoids, IFNγR-deficient Paneth cells, IFNγR-deficient ISCs, and purified stem cell colonies all indicated direct targeting of the ISCs that was not dependent on injury to the Paneth cell niche. Dysregulated T cell activation and IFNγ production are thus potent mediators of ISC injury, and blockade of JAK/STAT signaling within target tissue stem cells can prevent this T cell-mediated pathology.


Asunto(s)
Interferón gamma/inmunología , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Células Madre/inmunología , Linfocitos T/inmunología , Animales , Muerte Celular , Mucosa Intestinal/patología , Ratones
11.
Prog Lipid Res ; 30(1): 1-38, 1991.
Artículo en Inglés | MEDLINE | ID: mdl-1771169

RESUMEN

This comprehensive review was necessitated by recent observations suggesting that sphingomyelin and derivatives may serve second messenger functions. It has attempted to remain true to the theme of cellular signalling. Hence, it has focussed on the lipids involved primarily with respect to their metabolism and properties in mammalian systems. The enzymology involved has been emphasized. An attempt was made to define directions in which signals may be flowing. However, the evidence presented to date is insufficient to conclusively designate the mechanisms of stimulated lipid metabolism. Hence, the proposed pathways must be viewed as preliminary. Further, the biologic functions of these lipids is for the most part uncertain. Thus, it is difficult to presently integrate this sphingomyelin pathway into the greater realm of cell biology. Nevertheless, the present evidence appears to suggest that a sphingomyelin pathway is likely to possess important bioregulatory functions. Hopefully, interest in this novel pathway will grow and allow a more complete understanding of the roles of these sphingolipids in physiology and pathology.


Asunto(s)
Transducción de Señal/fisiología , Esfingomielinas/metabolismo , Animales , Membrana Celular/metabolismo , Ceramidas/metabolismo , Humanos , Mamíferos/metabolismo , Esfingomielinas/biosíntesis , Esfingomielinas/química
12.
Cancer Res ; 60(2): 321-7, 2000 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-10667583

RESUMEN

Injury to the central nervous system (CNS) by ionizing radiation may be a consequence of damage to the vascular endothelium. Recent studies showed that radiation-induced apoptosis of endothelial cells in vitro and in the lung in vivo is mediated by the lipid second messenger ceramide via activation of acid sphingomyelinase (ASM). This apoptotic response to radiation can be inhibited by basic fibroblast growth factor or by genetic mutation of ASM. In the CNS, single-dose radiation has been shown to result in a 15% loss of endothelial cells within 24 h, but whether or not this loss is associated with apoptosis remains unknown. In the present studies, dose- and time-dependent induction of apoptosis was observed in the C57BL/6 mouse CNS. Apoptosis was quantified by terminal deoxynucleotidyl transferase-mediated nick end labeling, and specific endothelial apoptosis was determined by histochemical double labeling with terminal deoxynucleotidyl transferase-mediated nick end labeling and Lycopersicon esculentum lectin. Beginning at 4 h after single-dose radiation, apoptosis was ongoing for 24 h and peaked at 12 h at an incidence of 0.7-1.4% of the total cells in spinal cord sections. Up to 20% of the apoptotic cells were endothelial. This effect was also seen in multiple regions of the brain (medulla, pons, and hippocampus). A significant reduction of radiation-induced apoptosis was observed after i.v. basic fibroblast growth factor treatment (0.45-4.5 microg/mouse). Identical results were noted in C3H/HeJ mice. Furthermore, irradiated ASM knockout mice displayed as much as a 70% reduction in endothelial apoptosis. This study demonstrates that ionizing radiation induces early endothelial cell apoptosis throughout the CNS. These data are consistent with recent evidence linking radiation-induced stress with ceramide and suggest approaches to modify the apoptotic response in control of radiation toxicity in the CNS.


Asunto(s)
Apoptosis/efectos de la radiación , Sistema Nervioso Central/irrigación sanguínea , Circulación Cerebrovascular/efectos de la radiación , Endotelio Vascular/efectos de la radiación , Factor 2 de Crecimiento de Fibroblastos/farmacología , Esfingomielina Fosfodiesterasa/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Encéfalo/citología , Encéfalo/fisiología , Encéfalo/efectos de la radiación , Circulación Cerebrovascular/efectos de los fármacos , Circulación Cerebrovascular/fisiología , Radioisótopos de Cesio , Relación Dosis-Respuesta a Droga , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Etiquetado Corte-Fin in Situ , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , Microglía/efectos de los fármacos , Microglía/fisiología , Microglía/efectos de la radiación , Esfingomielina Fosfodiesterasa/deficiencia , Esfingomielina Fosfodiesterasa/genética , Médula Espinal/efectos de los fármacos , Médula Espinal/fisiología , Médula Espinal/efectos de la radiación , Irradiación Corporal Total
13.
Cancer Res ; 58(10): 2260-4, 1998 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-9605775

RESUMEN

Protein kinase C (PKC) activation is often antiapoptotic, although in a few cell types PKC initiates apoptosis by an unknown mechanism. Recent investigations showed that activation of PKC alpha by 12-O-tetradecanoylphorbol 13-acetate (TPA) induced apoptosis in LNCaP prostate cancer cells. The present studies examine the mechanism of this effect and show that de novo ceramide generation through the enzyme ceramide synthase is required. TPA induced rapid ceramide generation, which was detectable by 1 h and increased linearly for 12 h. TPA-induced apoptosis was measurable by 12 h and was progressive for 48 h. Investigations into the mechanism of TPA-induced ceramide generation revealed that acid and neutral sphingomyelinase activities were not enhanced. However, TPA induced an increase in ceramide synthase activity that persisted for at least 16 h. Treatment with fumonisin B1, a specific natural inhibitor of ceramide synthase, abrogated both ceramide production and TPA-induced apoptosis. Ceramide analogues bypassed fumonisin B1 inhibition to initiate apoptosis directly. Thus, ceramide appears to be a necessary signal for TPA-induced apoptosis in LNCaP cells. This represents the first description of a pathway by which PKC may signal apoptosis.


Asunto(s)
Amidohidrolasas/metabolismo , Apoptosis/efectos de los fármacos , Carcinógenos/farmacología , Ceramidas/biosíntesis , Fumonisinas , Proteína Quinasa C/efectos de los fármacos , Acetato de Tetradecanoilforbol/farmacología , Amidohidrolasas/efectos de los fármacos , Ácidos Carboxílicos/farmacología , Carcinógenos Ambientales/farmacología , Ceramidasas , Interacciones Farmacológicas , Humanos , Masculino , Proteína Quinasa C/metabolismo , Células Tumorales Cultivadas/efectos de los fármacos
14.
Cancer Res ; 59(20): 5194-201, 1999 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-10537297

RESUMEN

Cell lines derived from human prostate cancer are regarded as relatively resistant to both radiation-induced clonogenic death and apoptosis. Here we attempted to modulate the response of LNCaP prostate cancer cells to radiation therapy (XRT) by pretreatment with 12-O-tetradecanoylphorbol acetate (TPA), a known apoptogenic agent in LNCaP cells. Using plateau-phase cultures, we investigated the response of these cells to XRT, TPA, and a combination of XRT and TPA. LNCaP irradiation did not result in ceramide generation or apoptosis. However, pretreatment with TPA enabled XRT to generate ceramide via activation of the enzyme ceramide synthase and signal apoptosis. Apoptosis was abrogated by the competitive inhibitor of ceramide synthase, fumonisin B1. Furthermore, when transplanted orthotopically into the prostate of nude mice, LNCaP cells produced tumors that recapitulated the responses of LNCaP cells in vitro. XRT or TPA failed to signal apoptosis in LNCaP tumors, whereas a combination of the two resulted in substantial (20-25%) apoptosis within 24 h. There was an additional benefit associated with this regimen because TPA pretreatment protected the adjacent rectum from radiation-induced apoptosis. This represents the first description of signaling-based therapy designed to overcome one form of radiation resistance expressed preferentially in LNCaP human prostate cancer cells.


Asunto(s)
Apoptosis , Oxidorreductasas/fisiología , Neoplasias de la Próstata/radioterapia , Fármacos Sensibilizantes a Radiaciones/farmacología , Acetato de Tetradecanoilforbol/farmacología , Animales , Apoptosis/efectos de los fármacos , Ceramidas/biosíntesis , Activación Enzimática/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Desnudos , Oxidorreductasas/efectos de los fármacos , Oxidorreductasas/efectos de la radiación , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/patología , Tolerancia a Radiación , Células Tumorales Cultivadas
15.
Oncogene ; 17(25): 3277-85, 1998 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-9916990

RESUMEN

Mammalian systems respond to environmental stress by either adapting or undergoing programmed cell death. While there is general agreement that the caspase family of proteases serve as the effectors of the apoptotic death response, the signaling apparatus involved in the decision to activate the caspase system is less clear. In the past few years, the sphingomyelin and c-Jun Kinase (JNK)/Stress-activated Protein Kinase (SAPK) pathways have been linked to the death response in many cellular systems. These signaling systems are found throughout the animal kingdom, and ceramide signaling is conserved through yeast. Since yeast do not undergo apoptosis, the sphingomyelin pathway appears evolutionarily older than the caspase-mediated death programs. While recent reviews by several groups have broadly surveyed ceramide signaling in apoptosis, this paper examines the role of sphingomyelinases and the JNK/SAPK pathway in coordinate signaling of apoptosis.


Asunto(s)
Apoptosis , Proteínas Quinasas Dependientes de Calcio-Calmodulina/fisiología , Ceramidas/fisiología , Proteínas Quinasas Activadas por Mitógenos , Transducción de Señal , Estrés Fisiológico/patología , Animales , Caspasas/metabolismo , Ceramidas/biosíntesis , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos , Oxidorreductasas/metabolismo , Esfingomielina Fosfodiesterasa/metabolismo , Estrés Fisiológico/metabolismo
16.
Biochim Biophys Acta ; 1179(2): 203-7, 1993 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-8218362

RESUMEN

We previously reported that protein kinase C (PKC) stimulation through phorbol ester (TPA) treatment enhances the effects of all-trans retinoic acid (RA) on immunophenotypic differentiation and RA nuclear receptor (RAR) activation in the multipotential human teratocarcinoma (TC) cell line NTera-2/clone D1 (abbreviated NT2/D1). This study extends prior work in NT2/D1 cells by demonstrating that PKC pathway activation is an early effect of RA treatment which regulates RAR transcriptional activity. RA activated the PKC pathway prior to induction of RAR-beta expression at 6 h, which is an established early marker of RAR activation in NT2/D1 cells. RA caused a transient 1.3-fold increase in intracellular diacylglycerol (DG) at 2 min and a translocation of the gamma isozyme of PKC (PKC-gamma) within 5 min. Transient co-transfection studies provided evidence that PKC pathway activation plays a role in the regulation of RAR-beta expression. In these studies a constitutively active PKC-gamma augmented the RA-mediated transactivation of a luciferase reporter containing the native RAR-beta promoter which has a retinoic-acid-response element (RARE). These findings reveal that PKC pathway activation is an early step in RA-mediated human TC differentiation and that PKC-gamma can potentiate the effects of RA on RAR transcriptional activation.


Asunto(s)
Proteína Quinasa C/metabolismo , Teratocarcinoma/patología , Tretinoina/farmacología , Diferenciación Celular , Membrana Celular/metabolismo , Citosol/metabolismo , Diglicéridos/metabolismo , Activación Enzimática , Humanos , Células Tumorales Cultivadas/efectos de los fármacos
17.
Cell Death Differ ; 3(2): 171-6, 1996 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17180079

RESUMEN

In this article, we review the role of sphingomyelinases and ceramide in the Fas-mediated apoptosis signal transduction cascade. Several stimuli, including ligation of Fas, have been shown to enhance either neutral and/or acidic sphingomyelinase activity and increase ceramide content in intact cells or cell membrane preparations. Ceramide seems to have different functions, including induction of apoptosis, growth arrest, and/or differentiation, depending on cell type or location of sphingomyelin hydrolysis within the cell. Several putative targets for ceramide activity, including a kinase and a phosphatase, have also been identified. While ceramide and acidic sphingomyelinase activity appear to be involved in apoptotic signalling for Fas and other members of the tumour necrosis factor receptor family, it is clear that other signals and mechanisms are necessary for Fas-mediated apoptosis.

18.
Cell Death Differ ; 2(4): 253-7, 1995 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17180030

RESUMEN

Ceramide, a long chain sphingolipid that is generated intracellularly upon hydrolysis of membrane-associated sphingomyelin, has recently been implicated as a second messenger-like molecule that is produced distal to ligation of the tumour necrosis factor receptor type 1 (TNFR1), as well as the related Fas (CD95/Apo-1) molecule. It is well established that ligation of TNFR1 or Fas leads to apoptosis in most cases. Furthermore, it has also recently been demonstrated that exposure to cell-permeable synthetic ceramides can result in apoptosis in many cases. These and other observations have led to the hypothesis that accumulation of intracellular ceramide may be a common element of several pathways that result in apoptosis. Here we show that exposure to synthetic ceramides triggers apoptosis in the human T lymphoblastoid cell lines, CEM and Jurkat, and that overexpression of the apoptosis-repressor protein, Bcl-2, renders these cells resistant to the apoptosis-inducing effects of ceramide, as well as to several other stimuli. Since exposure to ceramides can result in either cell proliferation, differentiation, cycle arrest, or death, the level of Bcl-2 expression in a cell may be an important factor in determining the outcome of signals that result in intracellular generation of this sphingolipid.

20.
Leukemia ; 16(9): 1596-602, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12200669

RESUMEN

The bioactive sphingolipid metabolite sphingosine-1-phosphate (S1P), formed by activation of sphingosine kinase in response to diverse stimuli, is an important lipid mediator that has novel dual actions - both inside and outside of cells. S1P is the ligand for a family of five G protein-coupled receptors. Activation of these GPCRs by S1P or dihydro-S1P regulates diverse processes, including cell migration, angiogenesis, vascular maturation, heart development, and neurite retraction. There is also abundant evidence that S1P can function as a second messenger important for regulation of calcium homeostasis, cell growth, and suppression of apoptosis. In many cases, the intracellular level of S1P and ceramide, another important sphingolipid metabolite associated with cell death and cell growth arrest, coordinately determine cell fate. Changes in S1P and ceramide have been implicated in a number of pathological conditions in which apoptosis plays an important role. Importantly, radiation-induced oocyte loss in adult female mice, the event that drives premature ovarian failure and infertility in female cancer patients, was completely prevented by in vivo therapy with S1P. Understanding the biosynthesis, metabolism and functions of S1P can uncover new targets for the pharmaceutical and therapeutic applications of S1P.


Asunto(s)
Lisofosfolípidos , Neoplasias/tratamiento farmacológico , Esfingosina/análogos & derivados , Esfingosina/uso terapéutico , Animales , Fenómenos Fisiológicos Celulares , Femenino , Proteínas de Unión al GTP/metabolismo , Humanos , Ratones , Transducción de Señal , Esfingosina/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA