Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
PLoS One ; 8(5): e63076, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23658799

RESUMEN

PURPOSE: Breast cancers that over-express a lipoxygenase or cyclooxygenase are associated with poor survival possibly because they overproduce metabolites that alter the cancer's malignant behaviors. However, these metabolites and behaviors have not been identified. We here identify which metabolites among those that stimulate breast cancer cell proliferation in vitro are associated with rapidly proliferating breast cancer. EXPERIMENTAL DESIGN: We used selective ion monitoring-mass spectrometry to quantify in the cancer and normal breast tissue of 27 patients metabolites that stimulate (15-, 12-, 5-hydroxy-, and 5-oxo-eicosatetraenoate, 13-hydroxy-octadecaenoate [HODE]) or inhibit (prostaglandin [PG]E2 and D2) breast cancer cell proliferation. We then related their levels to each cancer's proliferation rate as defined by its Mib1 score. RESULTS: 13-HODE was the only metabolite strongly, significantly, and positively associated with Mib1 scores. It was similarly associated with aggressive grade and a key component of grade, mitosis, and also trended to be associated with lymph node metastasis. PGE2 and PGD2 trended to be negatively associated with these markers. No other metabolite in cancer and no metabolite in normal tissue had this profile of associations. CONCLUSIONS: Our data fit a model wherein the overproduction of 13-HODE by 15-lipoxygenase-1 shortens breast cancer survival by stimulating its cells to proliferate and possibly metastasize; no other oxygenase-metabolite pathway, including cyclooxygenase-PGE2/D2 pathways, uses this specific mechanism to shorten survival.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Ácidos Grasos/metabolismo , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Proliferación Celular , Humanos , Persona de Mediana Edad , Clasificación del Tumor , Metástasis de la Neoplasia
2.
Carcinogenesis ; 34(1): 176-82, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23066085

RESUMEN

Fatty acid metabolism impacts multiple intracellular signaling pathways in many cell types, but its role in prostate cancer cells is still unclear. Our previous studies have shown that the n-3 polyunsaturated fatty acid docosahexaenoic acid (DHA) induces apoptosis in human prostate cancer cells by a syndecan-1 (SDC-1)-dependent mechanism. Here, we examined the contribution of lipoxygenase (LOX)- and cyclooxygenase (COX)-mediated DHA metabolism to this effect. Pan-LOX inhibitor (nordihydroguaiaretic acid), 15-LOX inhibitor (luteolin) or 15/12-LOX inhibitor (baicalein) blocked the induced effect of DHA on SDC-1 expression and apoptosis in human prostate cancer cells, whereas 5-LOX inhibitor, AA861, was ineffective. Human prostate cancer cells lines (PC3, LNCaP and DU145 cells) expressed two 15-LOX isoforms, 15-LOX-1 and 15-LOX-2, with higher 15-LOX-1 and lower 15-LOX-2 expressions compared with human epithelial prostate cells. Knockdown of 15-LOX-1 blocked the effect of DHA on SDC-1 expression and caspase-3 activity, whereas silencing 15-LOX-2, 5-LOX, COX-1, COX-2 or 12-LOX had no effect. Moreover, the ability of DHA to inhibit the activity of the PDK/Akt (T308) signaling pathway was abrogated by silencing 15-LOX-1. These findings demonstrate that 15-LOX-1-mediated metabolism of DHA is required for it to upregulate SDC-1 and trigger the signaling pathway that elicits apoptosis in prostate cancer cells.


Asunto(s)
Apoptosis , Araquidonato 15-Lipooxigenasa/metabolismo , Ácidos Docosahexaenoicos/metabolismo , Neoplasias de la Próstata/patología , Transducción de Señal , Sindecano-1/metabolismo , Humanos , Masculino , Neoplasias de la Próstata/metabolismo
3.
PLoS One ; 7(9): e45480, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23029040

RESUMEN

A 15-LOX, it is proposed, suppresses the growth of prostate cancer in part by converting arachidonic, eicosatrienoic, and/or eicosapentaenoic acids to n-6 hydroxy metabolites. These metabolites inhibit the proliferation of PC3, LNCaP, and DU145 prostate cancer cells but only at ≥1-10 µM. We show here that the 15-LOX metabolites of docosahexaenoic acid (DHA), 17-hydroperoxy-, 17-hydroxy-, 10,17-dihydroxy-, and 7,17-dihydroxy-DHA inhibit the proliferation of these cells at ≥0.001, 0.01, 1, and 1 µM, respectively. By comparison, the corresponding 15-hydroperoxy, 15-hydroxy, 8,15-dihydroxy, and 5,15-dihydroxy metabolites of arachidonic acid as well as DHA itself require ≥10-100 µM to do this. Like DHA, the DHA metabolites a) induce PC3 cells to activate a peroxisome proliferator-activated receptor-γ (PPARγ) reporter, express syndecan-1, and become apoptotic and b) are blocked from slowing cell proliferation by pharmacological inhibition or knockdown of PPARγ or syndecan-1. The DHA metabolites thus slow prostate cancer cell proliferation by engaging the PPARγ/syndecan-1 pathway of apoptosis and thereby may contribute to the prostate cancer-suppressing effects of not only 15-LOX but also dietary DHA.


Asunto(s)
Araquidonato 15-Lipooxigenasa/metabolismo , Ácidos Docosahexaenoicos/farmacología , Neoplasias de la Próstata/metabolismo , Apoptosis/efectos de los fármacos , Caspasas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Humanos , Masculino , PPAR gamma/metabolismo , Transducción de Señal/efectos de los fármacos , Sindecano-1/metabolismo
4.
Neoplasia ; 12(10): 826-36, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20927321

RESUMEN

Evidence indicates that diets enriched in n-3 polyunsaturated fatty acids (n-3 PUFAs) reduce the risk of prostate cancer, but biochemical mechanisms are unclear. Syndecan-1 (SDC-1), a transmembrane heparan sulfate proteoglycan, supports the integrity of the epithelial compartment. In tumor cells of epithelial lineage, SDC-1 is generally downregulated. This may result in perturbation of homeostasis and lead to progression of malignancy. Our studies have shown that the n-3 PUFA species, docosahexaenoic acid (DHA), increases SDC-1 expression in prostate tissues of Pten knockout (Pten(P-/-)) mice/cells and human prostate cancer cells. We have now determined that DHA-mediated up-regulation of SDC-1 induces apoptosis. Bovine serum albumin-bound DHA and exogenous human recombinant SDC-1 ecotodomain were delivered to PC3 and LNCaP cells in the presence or absence of SDC-1 small interfering (si)RNA. In the presence of control siRNA, both DHA and SDC-1 ectodomain induced apoptosis, whereas SDC-1 silencing blocked DHA-induced but not SDC-1 ectodomain-induced apoptosis. Downstream effectors of SDC-1 signaling linked to n-3 PUFA-induced apoptosis involved the 3'-phosphoinositide-dependent kinase 1 (PDK1)/Akt/Bad integrating network. A diet enriched in n-3 PUFA decreased phosphorylation of PDK1, Akt (T308), and Bad in prostates of Pten(P-/-) mice. Similar results were observed in human prostate cancer cells in response to DHA and SDC-1 ectodomain. The effect of DHA on PDK1/Akt/Bad signaling was abrogated by SDC-1 siRNA. These findings define a mechanism by which SDC-1-dependent suppression of phosphorylation of PDK1/Akt/Bad mediates n-3 PUFA-induced apoptosis in prostate cancer.


Asunto(s)
Apoptosis/efectos de los fármacos , Ácidos Docosahexaenoicos/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sindecano-1/metabolismo , Proteína Letal Asociada a bcl/metabolismo , Proteínas Quinasas Dependientes de 3-Fosfoinosítido , Animales , Western Blotting , Bovinos , Humanos , Técnicas para Inmunoenzimas , Masculino , Ratones , Ratones Noqueados , Neoplasias Hormono-Dependientes/tratamiento farmacológico , Neoplasias Hormono-Dependientes/metabolismo , Neoplasias Hormono-Dependientes/patología , Fosfohidrolasa PTEN/fisiología , Fosforilación/efectos de los fármacos , Neoplasias de la Próstata/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Albúmina Sérica Bovina/metabolismo , Transducción de Señal/efectos de los fármacos , Tasa de Supervivencia , Sindecano-1/antagonistas & inhibidores , Sindecano-1/genética , Células Tumorales Cultivadas , Proteína Letal Asociada a bcl/genética
5.
Neoplasia ; 11(10): 1042-53, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19794963

RESUMEN

Epidermal growth factor receptor (EGFR) and androgen receptor (AR) pathways play pivotal roles in prostate cancer progression. Therefore, agents with dual-targeting ability may have important therapeutic potential. Decorin, a proteoglycan present in the tumor microenvironment, is known to regulate matrix assembly, growth factor binding, and receptor tyrosine kinase activity. Here, we show that in prostate-specific Pten(P-/-) mice, a genetically defined, immune-competent mouse model of prostate cancer, systemic delivery of decorin inhibits tumor progression by targeting cell proliferation and survival pathways. Moreover, in human prostate cancer cells, we show that decorin specifically inhibits EGFR and AR phosphorylation and cross talk between these pathways. This prevents AR nuclear translocation and inhibits the production of prostate specific antigen. Further, the phosphatidylinositol-3 kinase (PI3K)/Akt cell survival pathway is suppressed leading to tumor cell apoptosis. Those findings highlight the effectiveness of decorin in the presence of a powerful genetic cancer risk and implicate decorin as a potential new agent for prostate cancer therapy by targeting EGFR/AR-PI3K-Akt pathways.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Factor de Crecimiento Epidérmico/metabolismo , Proteínas de la Matriz Extracelular/farmacología , Neoplasias de la Próstata/patología , Proteoglicanos/farmacología , Receptores Androgénicos/metabolismo , Animales , Western Blotting , Línea Celular Tumoral , Núcleo Celular/metabolismo , Decorina , Receptores ErbB/genética , Receptores ErbB/metabolismo , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Transporte de Proteínas/efectos de los fármacos , Proteoglicanos/genética , Proteoglicanos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Receptor Cross-Talk/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
6.
PPAR Res ; 2008: 358052, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18769551

RESUMEN

Omega-3 (or n-3) polyunsaturated fatty acids (PUFAs) and their metabolites are natural ligands for peroxisome proliferator receptor activator (PPAR)gamma and, due to the effects of PPARgamma on cell proliferation, survival, and differentiation, are potential anticancer agents. Dietary intake of omega-3 PUFAs has been associated with a reduced risk of certain cancers in human populations and in animal models. In vitro studies have shown that omega-3 PUFAs inhibit cell proliferation and induce apoptosis in cancer cells through various pathways but one of which involves PPARgamma activation. The differential activation of PPARgamma and PPARgamma-regulated genes by specific dietary fatty acids may be central to their distinct roles in cancer. This review summarizes studies relating PUFAs to PPARgamma and cancer and offers a new paradigm relating an n-3 PUFA through PPARgamma to the expression of the cell surface proteoglycan, syndecan-1, and to the death of cancer cells.

7.
J Biol Chem ; 283(26): 18441-9, 2008 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-18450755

RESUMEN

Syndecan 1 is the major proteoglycan produced by epithelial cells. It is strategically localized at the plasma membrane to participate in growth factor signaling and cell-cell and cell-matrix interactions. Its expression may modulate the properties of epithelial lineage tumor cells in which it is generally down-regulated compared with nontumor progenitors. The present study examined the regulation of syndecan 1 in prostate epithelial cells by n-3 polyunsaturated fatty acids. In prostate tissue of mice, syndecan 1 immunostaining was demonstrated in epithelial cells throughout each gland. In animals fed an n-3 polyunsaturated fatty acid-enriched diet, syndecan 1 mRNA was increased in all prostate glands. In the human prostate cancer cell line, PC-3, delivery of exogenous n-3 (but not n-6) fatty acids resulted in up-regulation of syndecan 1 expression. This effect was mimicked by a peroxisome proliferator-activated receptor (PPAR) gamma agonist, troglitazone, and inhibited in the presence of a PPARgamma antagonist and in cells transfected with dominant negative PPARgamma cDNA. Using a luciferase gene driven either by a PPAR response element or by a DR-1 site present in the syndecan 1 promoter, reporter activation was increased by n-3 low density lipoprotein, docosahexaenoic acid, and troglitazone, whereas activity of a luciferase gene placed downstream of a mutant DR-1 site was unresponsive. These findings indicate that syndecan 1 is up-regulated by n-3 fatty acids by a transcriptional pathway involving PPARgamma. This mechanism may contribute to the chemopreventive properties of n-3 fatty acids in prostate cancer.


Asunto(s)
Ácidos Grasos Omega-3/metabolismo , Regulación Neoplásica de la Expresión Génica , Regulación de la Expresión Génica , Próstata/metabolismo , Sindecano-1/biosíntesis , Animales , Línea Celular Tumoral , Cromanos/farmacología , Humanos , Masculino , Ratones , PPAR gamma/agonistas , PPAR gamma/antagonistas & inhibidores , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Sindecano-1/genética , Tiazolidinedionas/farmacología , Distribución Tisular , Troglitazona
8.
Cancer Res ; 68(8): 2912-9, 2008 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-18413760

RESUMEN

Diets enriched in n-3 polyunsaturated fatty acids (n-3 PUFA) may protect against breast cancer but biochemical mechanisms are unclear. Our studies showed that the n-3 fatty acid docosahexaenoic acid (DHA) up-regulated syndecan-1 (SDC-1) in human breast cancer cells, and we tested the hypothesis that DHA-mediated up-regulation of SDC-1 induces apoptosis. DHA was delivered to MCF-7 cells by n-3 PUFA-enriched low-density lipoproteins (LDL) or by albumin in the presence or absence of SDC-1 small interfering RNA. The n-3 PUFA induced apoptosis, which was blocked by SDC-1 silencing. We also confirmed that SDC-1 up-regulation and apoptosis promotion by n-3 PUFA was mediated by peroxisome proliferator-activated receptor gamma (PPAR gamma). Using a luciferase gene driven by either a PPAR response element or a DR-1 site present in the SDC-1 promoter, reporter activities were enhanced by n-3 LDL, DHA, and PPAR gamma agonist, whereas activity of a luciferase gene placed downstream of a mutant DR-1 site was unresponsive. Cotransfection with dominant-negative PPAR gamma DNA eliminated the increase in luciferase activity. These data provide strong evidence that SDC-1 is a molecular target of n-3 PUFA in human breast cancer cells through activation of PPAR gamma and that n-3 PUFA-induced apoptosis is mediated by SDC-1. This provides a novel mechanism for the chemopreventive effects of n-3 PUFA in breast cancer.


Asunto(s)
Apoptosis/efectos de los fármacos , Ácidos Grasos Omega-3/farmacología , PPAR gamma/fisiología , Sindecano-1/genética , Animales , Antineoplásicos/farmacología , Neoplasias de la Mama , Línea Celular Tumoral , Chlorocebus aethiops , Femenino , Aceites de Pescado , Humanos , Reacción en Cadena de la Polimerasa , ARN Neoplásico/genética , ARN Neoplásico/aislamiento & purificación
9.
Biochim Biophys Acta ; 1783(8): 1544-50, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18406359

RESUMEN

Cytosolic phospholipase A2 (cPLA2)alpha responds to the rise in cytosolic Ca2+ ([Ca2+]i) attending cell stimulation by moving to intracellular membranes, releasing arachidonic acid (AA) from these membranes, and thereby initiating the synthesis of various lipid mediators. Under some conditions, however, cPLA2alpha translocation occurs without any corresponding changes in [Ca2+]i. The signal for such responses has not been identified. Using confocal microscopy to track fluorescent proteins fused to cPLA2alpha or cPLA2alpha's C2 domain, we find that AA mimics Ca2+ ionophores in stimulating cPLA(2)alpha translocations to the perinuclear ER and to a novel site, the lipid body. Unlike the ionophores, AA acted independently of [Ca2+](i) rises and did not translocate the proteins to the Golgi. AA's action did not involve its metabolism to eicosanoids or acylation into cellular lipids. Receptor agonists also stimulated translocations targeting lipid bodies. We propose that AA is a signal for Ca2+-independent cPLA2alpha translocation and that lipid bodies are common targets of cPLA2alpha and contributors to stimulus-induced lipid mediator synthesis.


Asunto(s)
Colorantes Fluorescentes/análisis , Fosfolipasas A2 Grupo IV/metabolismo , Proteínas Luminiscentes/análisis , Calcio/metabolismo , Línea Celular , Aparato de Golgi/enzimología , Fosfolipasas A2 Grupo IV/genética , Humanos , Lípidos/análisis , Proteínas Luminiscentes/genética , Microscopía Confocal , Orgánulos/química , Orgánulos/enzimología , Transporte de Proteínas
11.
Biochim Biophys Acta ; 1736(3): 228-36, 2005 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-16154383

RESUMEN

MDA-MB-231, MCF7, and SKOV3 cancer cells, but not HEK-293 cells, expressed mRNA for the leukocyte G protein-coupled 5-oxo-eicosatetraenoate (ETE) OXE receptor. 5-Oxo-ETE, 5-oxo-15-OH-ETE, and 5-HETE stimulated the cancer cell lines but not HEK-293 cells to mount pertussis toxin-sensitive proliferation responses. Their potencies in eliciting this response were similar to their known potencies in activating leukocytes and OXE receptor-transfected cells. However, high concentrations of 5-oxo-ETE and 5-oxo-15-OH-ETE, but not 5-HETE, arrested growth and caused apoptosis in all four cell lines; these responses were pertussis toxin-resistant. The same high concentrations of the oxo-ETEs but again not 5-HETE also activated peroxisome proliferator-activated receptor (PPAR)-gamma. Pharmacological studies indicated that this activation did not mediate their effects on proliferation. These results are the first to implicate the OXE receptor in malignant cell growth and to show that 5-oxo-ETEs activate cell death programs as well as PPARgamma independently of this receptor.


Asunto(s)
Ácidos Araquidónicos/farmacología , Proliferación Celular/efectos de los fármacos , Receptores Eicosanoides/fisiología , Anilidas/farmacología , Apoptosis/efectos de los fármacos , Ácidos Araquidónicos/metabolismo , Sitios de Unión/genética , Caspasa 3 , Caspasas/metabolismo , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Expresión Génica/genética , Humanos , Ácidos Hidroxieicosatetraenoicos/metabolismo , Ácidos Hidroxieicosatetraenoicos/farmacología , Mitosis/efectos de los fármacos , PPAR gamma/antagonistas & inhibidores , PPAR gamma/genética , PPAR gamma/metabolismo , Receptores Activados del Proliferador del Peroxisoma/genética , Toxina del Pertussis/farmacología , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/metabolismo , Prostaglandina D2/análogos & derivados , Prostaglandina D2/metabolismo , Prostaglandina D2/farmacología , Unión Proteica , Receptores Eicosanoides/genética , Transfección
12.
Biochim Biophys Acta ; 1733(2-3): 120-9, 2005 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15863359

RESUMEN

Platelet-activating factor (1-O-alkyl-2-acetyl-sn-glycero-3-phosphocholine; PAF) is a potent inflammatory mediator produced by cells in response to physical or chemical stress. The mechanisms linking cell injury to PAF synthesis are unknown. We used liquid chromatography-tandem mass spectrometry to investigate stress-induced PAF synthesis in human neutrophils. PAF synthesis induced by extracellular pH 5.4 correlated with the activation of a stress-activated kinase, p38 mitogen-activated protein kinase (MAPK), and was blocked by the p38 MAPK inhibitor SB 203580. A key enzyme of PAF synthesis, acetyl-CoA:lysoPAF acetyltransferase, which we have previously shown is a target of p38 MAPK, was also activated in an SB 203580-sensitive fashion. Another MAPK pathway, extracellular signal-regulated kinase-1/2 (ERK-1/2), was also activated. Surprisingly, the pharmacological blockade of the ERK-1/2 pathway with PD 98059 did not block, but rather enhanced, PAF accumulation. Two unexpected actions of PD 98059 may underlie this phenomenon: an augmentation of stress-induced p38 MAPK phosphorylation and an inhibition of PAF catabolism. The latter effect did not appear to be due to a direct inhibition of PAF acetylhydrolase. Finally, similar results were obtained using another form of cellular stress, hypertonic sodium chloride. These data are consistent with a model in which stress-induced PAF accumulation is regulated positively by p38 MAPK and negatively by ERK-1/2. Such a model contrasts with the PAF accumulation induced by other forms of stimulation, which we and others have found is up-regulated by both p38 MAPK and ERK-1/2.


Asunto(s)
Neutrófilos/metabolismo , Factor de Activación Plaquetaria/metabolismo , Acetiltransferasas/metabolismo , Cromatografía Liquida , Dimetilsulfóxido , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Humanos , Concentración de Iones de Hidrógeno , Imidazoles/farmacología , Espectrometría de Masas , Presión Osmótica , Estrés Oxidativo , Fosforilación , Factor de Activación Plaquetaria/biosíntesis , Piridinas/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
13.
Clin Cancer Res ; 10(24): 8275-83, 2004 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-15623603

RESUMEN

PURPOSE: Omega-3 (n-3) fatty acids (FA) have been proposed to confer tumor-inhibitory properties. In vivo, dietary FA are delivered to tumor cells by two main routes: low-density lipoproteins (LDL) and albumin complexes. High FA concentration in LDL and up-regulation of LDL receptors in tumor cells suggest that the LDL receptor pathway may be the major route for FA delivery. We compared effects of n-3FA delivered to human cancer cells by LDL and albumin. EXPERIMENTAL DESIGN: LDL was isolated from plasma of African Green monkeys fed diets enriched in fish oil (n-3 FA) or linoleic acid (n-6FA) and used to deliver FA to MCF-7 and PC3 cancer cells. Cell proliferation, apoptosis, and changes in global gene expression were monitored. RESULTS: Both LDL and albumin were effective in delivering FA to tumor cells and modifying the composition of cell phospholipids. The molar ratio of 20:4 (n-6) to 20:5 (n-3) in phosphatidylcholine and phosphatidylethanolamine was profoundly decreased. Although cell phospholipids were similarly modified by LDL and albumin-delivered FA, effects on cell proliferation and on transcription were markedly different. LDL-delivered n-3 FA were more effective at inhibiting cell proliferation and inducing apoptosis. Expression microarray profiling showed that a significantly higher number of genes were regulated by LDL-delivered than albumin-delivered n-3 FA with little overlap between the two sets of genes. CONCLUSIONS: These results show the importance of the LDL receptor pathway in activating molecular mechanisms responsible for the tumor inhibitory properties of n-3FA.


Asunto(s)
Albúminas/administración & dosificación , Neoplasias de la Mama/patología , Sistemas de Liberación de Medicamentos , Ácidos Grasos Omega-3/administración & dosificación , Lipoproteínas LDL/administración & dosificación , Neoplasias de la Próstata/patología , Animales , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/metabolismo , Proliferación Celular/efectos de los fármacos , Chlorocebus aethiops , Colesterol/metabolismo , Femenino , Aceites de Pescado/administración & dosificación , Perfilación de la Expresión Génica , Humanos , Ácido Linoleico/administración & dosificación , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosfatidilcolinas/metabolismo , Fosfatidiletanolaminas/metabolismo , Neoplasias de la Próstata/metabolismo , Triglicéridos/metabolismo , Células Tumorales Cultivadas
14.
Br J Pharmacol ; 139(2): 388-98, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12770944

RESUMEN

1 Since most inflammatory mediators that stimulate granulocyte responsiveness also delay apoptosis, it is often assumed that activation and longevity are causally related. Using isolated human peripheral blood neutrophils and eosinophils, we examined this association by exploiting the proinflammatory lipid mediators, the leukotrienes (LTs), and investigated granulocyte function and apoptosis. 2 LTB(4) induced elevation of intracellular free Ca(2+) concentration ([Ca(2+)](i)), cell polarisation and retardation of neutrophil apoptosis, although the antiapoptotic effect occurred only at concentrations > or =300 nM. LTB(4)-induced activation was attenuated by CP-105,696, a BLT1-specific antagonist suggesting classical LTB(4) receptor BLT1 involvement. 3 Despite demonstrating the presence of the neutrophil intracellular LTB(4) receptor peroxisome-proliferator activator receptor-alpha (PPARalpha) in neutrophils, the selective PPARalpha agonist WY-14,643 did not mimic LTB(4)-induced prosurvival effects. 4 LTB(4)-induced survival, however, also appeared to be mediated by BLT1 since CP-105,696 inhibited the LTB(4)-mediated antiapoptotic effect. Furthermore, based on studies with CP-105,696 and 5-lipoxygenase inhibitors, lipopolysaccharide (LPS)-, granulocyte-macrophage colony-stimulating factor (GM-CSF)-, dexamethasone- and dibutyryl-cAMP (db-cAMP)-induced delay of neutrophil apoptosis did not involve autocrine production of LTB(4). 5 Although LTB(4) and LTD(4) induced human eosinophil [Ca(2+)](i) elevation and polarization, these LTs did not influence eosinophil apoptosis. Furthermore, LTB(4)- and LTD(4)-induced eosinophil activation was attenuated by CP-105,696 and the Cys-LT(1) receptor antagonist montelukast, respectively, highlighting specific receptor dependency. 6 Thus, mediator-triggered granulocyte activation and antiapoptotic pathways are distinct events that can be differentially regulated.


Asunto(s)
Apoptosis/efectos de los fármacos , Eosinófilos/citología , Leucotrieno B4/fisiología , Leucotrieno D4/fisiología , Neutrófilos/citología , Acetatos/farmacología , Adyuvantes Inmunológicos/farmacología , Apoptosis/fisiología , Benzopiranos/farmacología , Calcio/metabolismo , Ácidos Carboxílicos/farmacología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Ciclopropanos , Eosinófilos/metabolismo , Eosinófilos/fisiología , Humanos , Antagonistas de Leucotrieno/farmacología , Leucotrieno B4/farmacología , Leucotrieno D4/farmacología , Neutrófilos/metabolismo , Neutrófilos/fisiología , Quinolinas/farmacología , Sulfuros
15.
Cancer Res ; 62(23): 6817-9, 2002 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-12460891

RESUMEN

5(S)-Hydroxy-6,8,11,14-E,Z,Z,Z-eicosatetraenoate (5-HETE) causes PC3 cells to grow by an unknown mechanism. We find that it also induces the cells to activate extracellular signal-regulated kinases and Akt. Pertussis toxin inhibits both responses. 5-HETE, 5-oxo-6,8,11,14-E,Z,Z,Z-eicosatetraenoate, and 5-oxo-15-hydroxy-eicosatetraenoate are known to stimulate leukocytes by a receptor coupled to pertussis toxin-sensitive G proteins. Their respective relative potencies in leukocytes are 1, 10, and 3. In PC3 cells, however, these values are 10, 1, and 0. PC3 cells, we propose, express a non-leukocyte-type, G protein-coupled, 5-HETE receptor. This novel receptor and the extracellular signal-regulated kinase and Akt pathways it recruits may contribute to the progression of prostate adenocarcinoma.


Asunto(s)
Ácidos Hidroxieicosatetraenoicos/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Neoplasias de la Próstata/patología , Proteínas Serina-Treonina Quinasas , Receptores Eicosanoides/fisiología , Benzoquinonas/farmacología , Proteínas de Unión al GTP/fisiología , Humanos , Ácidos Hidroxieicosatetraenoicos/antagonistas & inhibidores , Ácidos Hidroxieicosatetraenoicos/fisiología , Indoles/farmacología , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Toxina del Pertussis/farmacología , Fosforilación/efectos de los fármacos , Neoplasias de la Próstata/enzimología , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Receptores Eicosanoides/metabolismo , Estimulación Química , Células Tumorales Cultivadas
16.
Biochim Biophys Acta ; 1592(2): 175-84, 2002 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-12379481

RESUMEN

Human neutrophils (PMN) are potentially a major source of platelet-activating factor (PAF) produced during inflammatory responses. The stimulated synthesis of PAF in PMN is carried out by a phospholipid remodeling pathway involving three enzymes: acetyl-CoA:lyso-PAF acetyltransferase (acetyltransferase), type IV phospholipase A(2) (cPLA(2)) and CoA-independent transacylase (CoA-IT). However, the coordinated actions and the regulatory mechanisms of these enzymes in PAF synthesis are poorly defined. A23187 has been widely used to activate the remodeling pathway, but it has not been shown how closely its actions mimic those of physiological stimuli. Here we address this important problem and compare responses of the three remodeling enzymes and PAF synthesis by intact cells. In both A23187- and N-formyl-methionyl-leucyl-phenylalanine (fMLP)-stimulated PMN, acetyltransferase activation is blocked by SB 203580, a p38 MAP kinase inhibitor, but not by PD 98059, which blocks activation of the ERKs. In contrast, either agent attenuated cPLA(2) activation. Correlating with these results, SB 203580 decreased stimulated PAF formation by 60%, whereas PD 98059 had little effect. However, the combination of both inhibitors decreased PAF formation to control levels. Although a role for CoA-IT in PAF synthesis is recognized, we did not detect activation of the enzyme in stimulated PMN. CoA-IT thus appears to exhibit full activity in resting as well as stimulated cells. We conclude that the calcium ionophore A23187 and the receptor agonist fMLP both act through common pathways to stimulate PAF synthesis, with p38 MAP kinase regulating acetyltransferase and supplementing ERK activation of cPLA(2).


Asunto(s)
Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neutrófilos/metabolismo , Factor de Activación Plaquetaria/biosíntesis , Acetiltransferasas/antagonistas & inhibidores , Acetiltransferasas/metabolismo , Aciltransferasas/antagonistas & inhibidores , Aciltransferasas/metabolismo , Calcimicina/farmacología , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , N-Formilmetionina Leucil-Fenilalanina/farmacología , Fosfolipasas A/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...