Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
J Pharmacol Exp Ther ; 371(1): 63-74, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31371480

RESUMEN

Sphingosine kinase [(SK), isoforms SK1 and SK2] catalyzes the formation of the bioactive lipid, sphingosine 1-phosphate (S1P). This can be exported from cells and bind to S1P receptors to modulate vascular function. We investigated the effect of short-term hypoxia on SK1 expression and the response of arteries to S1P. SK1 expression in rat aortic and coronary artery endothelial cells was studied using immunofluorescence and confocal microscopy. Responses of rat aortic rings were studied using wire myography and reversible hypoxia induced by bubbling myography chambers with 95% N2:5% CO2 Inhibitors were added 30 minutes before induction of hypoxia. S1P induced endothelium-dependent vasodilation via activation of S1P3 receptors and generation of nitric oxide. Hypoxia significantly increased relaxation to S1P and this was attenuated by (2R)-1-[[(4-[[3-methyl-5-[(phenylsulfonyl)methyl] phenoxy]methyl]phenyl]methyl]-2-pyrrolidinemethanol [(PF-543), SK1 inhibitor] but not (R)-FTY720 methyl ether [(ROMe), SK2 inhibitor]. Hypoxia also increased vessel contractility to the thromboxane mimetic, 9,11-dideoxy-11α,9α-epoxymethanoprostaglandin F2α, which was further increased by PF-543 and ROMe. Hypoxia upregulated SK1 expression in aortic and coronary artery endothelial cells and this was blocked by PF-543 and 2-(p-hydroxyanilino)-4-(p-chlorophenyl)thiazole [(SKi), SK1/2 inhibitor]. The effects of PF-543 and SKi were associated with increased proteasomal/lysosomal degradation of SK1. A short period of hypoxia increases the expression of SK1, which may generate S1P to oppose vessel contraction. Under hypoxic conditions, upregulation of SK1 is likely to lead to increased export of S1P from the cell and vasodilation via activation of endothelial S1P3 receptors. These data have significance for perfusion of tissue during episodes of ischemia.


Asunto(s)
Hipoxia/metabolismo , Lisofosfolípidos/farmacología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Receptores de Esfingosina-1-Fosfato/metabolismo , Esfingosina/análogos & derivados , Vasodilatación , Animales , Aorta/efectos de los fármacos , Aorta/metabolismo , Aorta/fisiología , Vasos Coronarios/efectos de los fármacos , Vasos Coronarios/metabolismo , Vasos Coronarios/fisiopatología , Hipoxia/fisiopatología , Masculino , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Proteolisis , Ratas , Ratas Sprague-Dawley , Esfingosina/farmacología , Regulación hacia Arriba
2.
Leukemia ; 30(11): 2142-2151, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27461062

RESUMEN

Sphingolipids, such as ceramide, sphingosine and sphingosine 1-phosphate (S1P) are bioactive molecules that have important functions in a variety of cellular processes, which include proliferation, survival, differentiation and cellular responses to stress. Sphingolipids have a major impact on the determination of cell fate by contributing to either cell survival or death. Although ceramide and sphingosine are usually considered to induce cell death, S1P promotes survival of cells. Sphingosine kinases (SPHKs) are the enzymes that catalyze the conversion of sphingosine to S1P. There are two isoforms, SPHK1 and SPHK2, which are encoded by different genes. SPHK1 has recently been implicated in contributing to cell transformation, tumor angiogenesis and metastatic spread, as well as cancer cell multidrug-resistance. More recent findings suggest that SPHK2 also has a role in cancer progression. This review is an overview of our understanding of the role of SPHKs and S1P in hematopoietic malignancies and provides information on the current status of SPHK inhibitors with respect to their therapeutic potential in the treatment of hematological cancers.


Asunto(s)
Neoplasias Hematológicas/tratamiento farmacológico , Neoplasias Hematológicas/enzimología , Terapia Molecular Dirigida/métodos , Progresión de la Enfermedad , Humanos , Lisofosfolípidos/antagonistas & inhibidores , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/uso terapéutico , Esfingosina/análogos & derivados , Esfingosina/antagonistas & inhibidores
3.
Oncogene ; 33(26): 3361-3, 2014 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-23873031

RESUMEN

In this issue of Oncogene, Albinet et al. have demonstrated a critical role of melanoma sphingosine kinase 1, which catalyses formation of sphingosine 1-phosphate (S1P), in promoting the differentiation of fibroblasts into myofibroblasts. The myofibroblast sphingosine kinase 1 then promotes the S1P-dependent dissemination (metastasis) of melanoma cells via a S1P receptor 3-mediated mechanism. These findings are of major significance because they provide a novel mechanism of interaction between melanoma and the microenvironment niche in promoting metastasis. These studies therefore identify S1P derived from myofibroblasts and melanoma cells as a novel target for therapeutic intervention.


Asunto(s)
Fibroblastos/patología , Melanoma/patología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Neoplasias Cutáneas/patología , Animales , Femenino , Humanos
4.
Cell Death Dis ; 4: e927, 2013 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-24263101

RESUMEN

Although the ability of bioactive lipid sphingosine-1-phosphate (S1P) to positively regulate anti-apoptotic/pro-survival responses by binding to S1P1 is well known, the molecular mechanisms remain unclear. Here we demonstrate that expression of S1P1 renders CCL39 lung fibroblasts resistant to apoptosis following growth factor withdrawal. Resistance to apoptosis was associated with attenuated accumulation of pro-apoptotic BH3-only protein Bim. However, although blockade of extracellular signal-regulated kinase (ERK) activation could reverse S1P1-mediated suppression of Bim accumulation, inhibition of caspase-3 cleavage was unaffected. Instead S1P1-mediated inhibition of caspase-3 cleavage was reversed by inhibition of phosphatidylinositol-3-kinase (PI3K) and protein kinase C (PKC), which had no effect on S1P1 regulation of Bim. However, S1P1 suppression of caspase-3 was associated with increased expression of anti-apoptotic protein Mcl-1, the expression of which was also reduced by inhibition of PI3K and PKC. A role for the induction of Mcl-1 in regulating endogenous S1P receptor-dependent pro-survival responses in human umbilical vein endothelial cells was confirmed using S1P receptor agonist FTY720-phosphate (FTY720P). FTY720P induced a transient accumulation of Mcl-1 that was associated with a delayed onset of caspase-3 cleavage following growth factor withdrawal, whereas Mcl-1 knockdown was sufficient to enhance caspase-3 cleavage even in the presence of FTY720P. Consistent with a pro-survival role of S1P1 in disease, analysis of tissue microarrays from ER(+) breast cancer patients revealed a significant correlation between S1P1 expression and tumour cell survival. In these tumours, S1P1 expression and cancer cell survival were correlated with increased activation of ERK, but not the PI3K/PKB pathway. In summary, pro-survival/anti-apoptotic signalling from S1P1 is intimately linked to its ability to promote the accumulation of pro-survival protein Mcl-1 and downregulation of pro-apoptotic BH3-only protein Bim via distinct signalling pathways. However, the functional importance of each pathway is dependent on the specific cellular context.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Supervivencia Celular/fisiología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas de la Membrana/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Proteínas Reguladoras de la Apoptosis/genética , Proteína 11 Similar a Bcl2 , Supervivencia Celular/genética , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/genética , Citometría de Flujo , Humanos , Immunoblotting , Inmunohistoquímica , Proteínas de la Membrana/genética , Microscopía Confocal , Fosfatidilinositol 3-Quinasas/genética , Proteína Quinasa C/genética , Proteínas Proto-Oncogénicas/genética , Receptores de Lisoesfingolípidos/genética
5.
Oncogene ; 32(13): 1660-9, 2013 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-22710717

RESUMEN

Ubiquitination of epidermal growth factor receptor (EGFR) is required for downregulation of the receptor by endocytosis. Impairment of this pathway results in constitutively active EGFR, which is associated with carcinogenesis, particularly in lung cancer. We previously demonstrated that the deubiquitinating enzyme ubiquitin-specific protease 2a (USP2a) has oncogenic properties. Here, we show a new role for USP2a as a regulator of EGFR endocytosis. USP2a localizes to early endosomes and associates with EGFR, stabilizing the receptor, which retains active downstream signaling. HeLa cells transiently expressing catalytically active, but not mutant (MUT), USP2a show increased plasma membrane-localized EGFR, as well as decreased internalized and ubiquitinated EGFR. Conversely, USP2a silencing reverses this phenotype. Importantly, USP2a prevents the degradation of MUT in addition to wild-type EGFR. Finally, we observed that USP2a and EGFR proteins are coordinately overexpressed in non-small cell lung cancers. Taken together, our data indicate that USP2a antagonizes EGFR endocytosis and thus amplifies signaling activity from the receptor. Our findings suggest that regulation of deubiquitination could be exploited therapeutically in cancers overexpressing EGFR.


Asunto(s)
Endocitosis/fisiología , Endopeptidasas/fisiología , Receptores ErbB/metabolismo , Proteolisis , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Regulación hacia Abajo/genética , Endocitosis/genética , Endopeptidasas/genética , Endopeptidasas/metabolismo , Receptores ErbB/genética , Regulación Neoplásica de la Expresión Génica , Células HeLa , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Estabilidad Proteica , Ubiquitina Tiolesterasa , Proteasas Ubiquitina-Específicas , Ubiquitinación/genética
6.
Br J Cancer ; 106(8): 1453-9, 2012 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-22460268

RESUMEN

BACKGROUND: We previously reported that sphingosine 1-phosphate receptor 4 (S1P(4)) is expressed and stimulates the ERK-1/2 pathway via a human epidermal growth factor receptor 2 (HER2)-dependent mechanism in oestrogen receptor-negative (ER(-)) MDA-MB-453 breast cancer cells. METHODS: Clinical relevance of S1P(4) and sphingosine kinase 1 (SK1, which catalyses the formation of S1P) was assessed in a cohort of 140 ER(-) breast tumours by immunohistochemistry (IHC) and the weighted histoscore method. Additional evidence for a functional interaction between S1P(4) and SK1 and between HER2 and SK1 was obtained using MDA-MB-453 cells. RESULTS: High S1P(4) expression is associated with shorter disease-free (P=0.014) and disease-specific survival (P=0.004), and was independent on multivariate analysis. In addition, patients with tumours that contain high and low levels of SK1 and S1P(4), respectively, have a significantly shorter disease-free survival (P=0.043) and disease-specific survival (P=0.033) compared with patients whose tumours contain both low S1P(4) and SK1 levels. In addition, high tumour expression of SK1 was significantly associated with shorter disease-specific survival (P=0.0001) in patients with HER2-positive tumours. Treatment of MDA-MB-453 cells with the SK1 inhibitor, SKi (2-(p-hydroxyanilino)-4-(p-chlorophenyl)thiazole) reduced the basal and S1P/S1P(4)-induced activation of ERK-1/2 and altered HER2 trafficking in these cells. CONCLUSION: These findings highlight an important role for S1P(4) and SK1 in ER(-) breast cancer progression.


Asunto(s)
Neoplasias de la Mama/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/biosíntesis , Receptores de Lisoesfingolípidos/biosíntesis , Neoplasias de la Mama/diagnóstico , Femenino , Humanos , Inmunohistoquímica , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Pronóstico , Receptores de Estrógenos/deficiencia , Resultado del Tratamiento , Células Tumorales Cultivadas
7.
Br J Pharmacol ; 161(1): 176-92, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20718749

RESUMEN

BACKGROUND AND PURPOSE: Anandamide and sphingosine-1-phosphate (S1P) both regulate vascular tone in a variety of vessels. This study aimed to examine the mechanisms involved in the regulation of coronary vascular tone by anandamide and S1P, and to determine whether any functional interaction occurs between these receptor systems. EXPERIMENTAL APPROACH: Mechanisms used by anandamide and S1P to regulate rat coronary artery (CA) reactivity were investigated using wire myography. Interactions between S1P and the cannabinoid (CB)(2) receptor were determined using human embryonic kidney 293 (HEK293) cells that stably over-express recombinant CB(2) receptor. KEY RESULTS: Anandamide and S1P induced relaxation of the rat CA. CB(2) receptor antagonists attenuated anandamide-induced relaxation, while S1P-mediated relaxation was dependent on the vascular endothelium and S1P(3). Anandamide treatment resulted in an increase in the phosphorylation of sphingosine kinase-1 within the CA. Conversely, anandamide-mediated relaxation was attenuated by inhibition of sphingosine kinase. Moreover, S1P(3), specifically within the vascular endothelium, was required for anandamide-mediated vasorelaxation. In addition to this, S1P-mediated relaxation was also reduced by CB(2) receptor antagonists and sphingosine kinase inhibition. Further evidence that S1P functionally interacts with the CB(2) receptor was also observed in HEK293 cells over-expressing the CB(2) receptor. CONCLUSIONS AND IMPLICATIONS: In the vascular endothelium of rat CA, anandamide induces relaxation via a mechanism requiring sphingosine kinase-1 and S1P/S1P(3). In addition, we report that S1P may exert some of its effects via a CB(2) receptor- and sphingosine kinase-dependent mechanism, where subsequently formed S1P may have privileged access to S1P(3) to induce vascular relaxation.


Asunto(s)
Ácidos Araquidónicos/farmacología , Vasos Coronarios/fisiología , Lisofosfolípidos/farmacología , Alcamidas Poliinsaturadas/farmacología , Esfingosina/análogos & derivados , Animales , Ácidos Araquidónicos/administración & dosificación , Bloqueadores de los Canales de Calcio/administración & dosificación , Bloqueadores de los Canales de Calcio/farmacología , Fármacos Cardiovasculares/administración & dosificación , Fármacos Cardiovasculares/farmacología , Línea Celular , Dronabinol/análogos & derivados , Dronabinol/farmacología , Endocannabinoides , Humanos , Indoles/farmacología , Indometacina/administración & dosificación , Indometacina/farmacología , Lisofosfolípidos/administración & dosificación , Masculino , Alcamidas Poliinsaturadas/administración & dosificación , Ratas , Ratas Sprague-Dawley , Receptor Cannabinoide CB2/antagonistas & inhibidores , Esfingosina/administración & dosificación , Esfingosina/farmacología , Vasodilatación
8.
Scand J Med Sci Sports ; 20(5): 725-30, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19765240

RESUMEN

The purpose of this investigation was to determine the association between gender and the prevalence and incidence of patellofemoral pain syndrome (PFPS). One thousand five hundred and twenty-five participants from the United States Naval Academy (USNA) were followed for up to 2.5 years for the development of PFPS. Physicians and certified athletic trainers documented the cases of PFPS. PFPS was defined as retropatellar pain during at least two of the following activities: ascending/descending stairs, hopping/jogging, prolonged sitting, kneeling, and squatting, negative findings on examination of knee ligament, menisci, bursa, and synovial plica, and pain on palpation of either the patellar facets or femoral condyles. Poisson and logistic regressions were performed to determine the association between gender and the incidence and prevalence of PFPS, respectively. The incidence rate for PFPS was 22/1000 person-years. Females were 2.23 times (95% CI: 1.19, 4.20) more likely to develop PFPS compared with males. While not statistically significant, the prevalence of PFPS at study enrollment tended to be higher in females (15%) than in males (12%) (P=0.09). Females at the USNA are significantly more likely to develop PFPS than males. Additionally, at the time of admission to the academy, the prevalence of PFPS was not significantly different between genders.


Asunto(s)
Síndrome de Dolor Patelofemoral/epidemiología , Estudios de Cohortes , Femenino , Humanos , Incidencia , Modelos Logísticos , Masculino , Distribución de Poisson , Prevalencia , Distribución por Sexo , Factores Sexuales
9.
Biochem Soc Trans ; 33(Pt 6): 1370-4, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16246121

RESUMEN

Mammalian LPPs (lipid phosphate phosphatases) are integral membrane proteins that belong to a superfamily of lipid phosphatases/phosphotransferases. They have broad substrate specificity in vitro, dephosphorylating PA (phosphatidic acid), S1P (sphingosine 1-phosphate), LPA (lysophosphatidic acid) etc. Their physiological role may include the attenuation of S1P- and LPA-stimulated signalling by virtue of an ecto-activity (i.e. dephosphorylation of extracellular S1P and LPA), thereby limiting the activation of LPA- and S1P-specific G-protein-coupled receptors at the cell surface. However, our recent work suggests that an intracellular action of LPP2 and LPP3 may account for the reduced agonist-stimulated p42/p44 mitogen-activated protein kinase activation of HEK-293 (human embryonic kidney 293) cells. This may involve a reduction in the basal levels of PA and S1P respectively and the presence of an early apoptotic phenotype under conditions of stress (serum deprivation). Additionally, we describe a model whereby LPP2, but not LPP3, may be functionally linked to the phospholipase D1-derived PA-dependent recruitment of sphingosine kinase 1 to the perinuclear compartment. We also consider the potential regulatory mechanisms for LPPs, which may involve oligomerization. Lastly, we highlight many aspects of the LPP biology that remain to be fully defined.


Asunto(s)
Isoenzimas/metabolismo , Lípidos/química , Fosfatos/metabolismo , Fosfatidato Fosfatasa/metabolismo , Sistemas de Mensajero Secundario/fisiología , Animales , Supervivencia Celular , Humanos , Isoenzimas/genética , Fosfatos/química , Fosfatidato Fosfatasa/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo
10.
Biochem Soc Trans ; 31(Pt 6): 1220-5, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14641030

RESUMEN

The formation of complexes between growth factor receptors and members of a family of G-protein-coupled receptors whose natural ligands are S1P (sphingosine 1-phosphate) and LPA (lysophosphatidic acid) represents a new signalling entity. This receptor complex allows for integrated signalling in response to growth factor and/or S1P/LPA and provides a mechanism for more efficient activation (due to integrated close-proximity signalling from both receptor classes) of the p42/p44 MAPK (mitogen-activated protein kinase) pathway. This article provides information on the molecular events at the interface between receptor tyrosine kinases and S1P/LPA receptors. Examples include the PDGF (platelet-derived growth factor)-induced tyrosine phosphorylation of G(i)alpha, released upon S1P(1) receptor activation, which is required for initiation of the p42/p44 MAPK pathway. Critical to this event is the formation of endocytic vesicles containing functionally active PDGFbeta receptor-S1P(1) receptor complexes, which are internalized and relocated with components of the p42/p44 MAPK pathway. We also report examples of cross-talk signal integration between the Trk A (tropomyosin receptor kinase A) receptor and the LPA(1) receptor in terms of the NGF (nerve growth factor)-dependent regulation of the p42/p44 MAPK pathway. NGF induces recruitment of the LPA(1) receptor to the nucleus (delivery might be Trk A-dependent), whereupon the LPA(1) receptor may govern gene expression via novel nuclear signalling processes.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de Superficie Celular/metabolismo , Transducción de Señal , Línea Celular , Humanos , Factor de Crecimiento Nervioso/metabolismo , Proteínas Proto-Oncogénicas c-sis/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores Lisofosfolípidos
11.
Science ; 294(5544): 1005-6, 2001 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-11691972
12.
Circulation ; 104(13): 1538-43, 2001 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-11571249

RESUMEN

BACKGROUND: Mitogenic stimuli present at the site of coronary arterial balloon injury contribute to the progression and development of a restenotic lesion, many signaling through a common pathway involving the small G protein p21(ras). Our aim was to demonstrate in biochemical studies that farnesyl protein transferase inhibitor III (FPTIII) is an inhibitor of p21(ras) processing and that when it is given locally in vivo at the site of coronary balloon injury in a porcine model, it can inhibit neointima formation. METHODS AND RESULTS: FPTIII (1 to 25 micromol/L) concentration-dependently reduced p21(ras) levels in porcine coronary artery smooth muscle cell membranes. FPTIII also prevented p42/p44 MAPK activation and DNA synthesis in response to platelet-derived growth factor in these cells at a concentration of 25 micromol/L. Application of 25 micromol/L FPTIII locally for 15 minutes to balloon-injured porcine coronary arteries in vivo prevented neointima formation assessed at 4 weeks, reduced proteoglycan deposition, and inhibited adventitial hypertrophy. Coronary arteries from FPTIII-treated pigs had no deterioration in contraction or in endothelium-dependent relaxation. CONCLUSIONS: The study demonstrates in the pig that short-term local delivery of inhibitors of p21(ras)-dependent mitogenic signal transduction prevents restenosis after balloon angioplasty.


Asunto(s)
Transferasas Alquil y Aril/antagonistas & inhibidores , Angioplastia Coronaria con Balón/efectos adversos , Inhibidores Enzimáticos/uso terapéutico , Organofosfonatos/uso terapéutico , Complicaciones Posoperatorias/prevención & control , Animales , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/administración & dosificación , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/fisiología , Organofosfonatos/administración & dosificación , Transducción de Señal/efectos de los fármacos , Porcinos , Túnica Íntima/efectos de los fármacos , Túnica Íntima/patología , Enfermedades Vasculares/etiología , Enfermedades Vasculares/prevención & control
13.
Br J Pharmacol ; 134(1): 6-9, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11522591

RESUMEN

Several different molecular species of phosphatidic acid (PA) bind to a G-protein coupled receptor (GPCR) to induce activation of the p42/p44 mitogen-activated protein kinase (p42/p44 MAPK) pathway in HEK 293 cells. PA is active at low nanomolar concentrations and the response is sensitive to pertussis toxin (which uncouples GPCRs from G(i/o)). The de-acylated product of PA, lysophosphatidic acid (LPA), which binds to members of the endothelial differentiation gene (EDG) family of receptors also stimulated p42/p44 MAPK in a pertussis toxin sensitive manner, but with an approximately 100 - 1000 fold lower potency compared with the different molecular species of PA. RT - PCR using gene-specific primers showed that HEK 293 cells express EDG2 and PSP24, the latter being a lipid binding GPCR out with the EDG cluster. We conclude that PA is a novel high potency GPCR agonist.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Lisofosfolípidos/farmacología , Ácidos Fosfatidicos/farmacología , Receptores de Superficie Celular/agonistas , Línea Celular , Relación Dosis-Respuesta a Droga , Humanos , Riñón/citología , Riñón/embriología , Riñón/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/efectos de los fármacos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , ARN/genética , ARN/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Acetato de Tetradecanoilforbol/farmacología
14.
Mol Pharmacol ; 60(1): 63-70, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11408601

RESUMEN

In this study, we have shown that nerve growth factor (NGF)-dependent activation of the p42/p44 mitogen-activated protein kinase (p42/p44 MAPK) pathway in PC12 cells can be partially blocked by pertussis toxin (which inactivates the G proteins G(i/o)). This suggests that the Trk A receptor may use a G protein-coupled receptor pathway to signal to p42/p44 MAPK. This was supported by data showing that the NGF-dependent activation of p42/p44 MAPK is potentiated in cells transfected with G protein-coupled receptor kinase 2 (GRK2) or beta-arrestin I. Moreover, GRK2 is constitutively bound with the Trk A receptor, whereas NGF stimulates the pertussis toxin-sensitive binding of beta-arrestin I to the TrkA receptor-GRK2 complex. Both GRK2 and beta-arrestin I are involved in clathrin-mediated endocytic signaling to p42/p44 MAPK. Indeed, inhibitors of clathrin-mediated endocytosis (e.g., monodansylcadaverine, concanavalin A, and hyperosmolar sucrose) reduced the NGF-dependent activation of p42/p44 MAPK. Finally, we have found that the G protein-coupled receptor-dependent component regulating p42/p44 MAPK is required for NGF-induced differentiation of PC12 cells. Thus, NGF-dependent inhibition of DNA synthesis was partially blocked by PD098059 (inhibitor of MAPK kinase-1 activation) and pertussis toxin. Our findings are the first to show that the Trk A receptor uses a classic G protein-coupled receptor-signaling pathway to promote differentiation of PC12 cells.


Asunto(s)
Arrestinas/fisiología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Factor de Crecimiento Nervioso/metabolismo , Proteínas Serina-Treonina Quinasas/fisiología , Animales , Diferenciación Celular/fisiología , Clatrina/antagonistas & inhibidores , Clatrina/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , ADN/biosíntesis , ADN/efectos de los fármacos , Endocitosis/efectos de los fármacos , Endocitosis/fisiología , Activación Enzimática , Quinasa 2 del Receptor Acoplado a Proteína-G , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/fisiología , Proteínas de Unión al GTP Heterotriméricas/fisiología , Proteína Quinasa 3 Activada por Mitógenos , Células PC12 , Fosfatidilinositol 3-Quinasas/fisiología , Fosforilación , Ratas , Receptor trkA/metabolismo , Tirosina/metabolismo , Quinasas de Receptores Adrenérgicos beta , beta-Arrestinas
15.
J Biol Chem ; 276(30): 28578-85, 2001 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-11359779

RESUMEN

Here we provide evidence to show that the platelet-derived growth factor beta receptor is tethered to endogenous G-protein-coupled receptor(s) in human embryonic kidney 293 cells. The tethered receptor complex provides a platform on which receptor tyrosine kinase and G-protein-coupled receptor signals can be integrated to produce more efficient stimulation of the p42/p44 mitogen-activated protein kinase pathway. This was based on several lines of evidence. First, we have shown that pertussis toxin (which uncouples G-protein-coupled receptors from inhibitory G-proteins) reduced the platelet-derived growth factor stimulation of p42/p44 mitogen-activated protein kinase. Second, transfection of cells with inhibitory G-protein alpha subunit increased the activation of p42/p44 mitogen-activated protein kinase by platelet-derived growth factor. Third, platelet-derived growth factor stimulated the tyrosine phosphorylation of the inhibitory G-protein alpha subunit, which was blocked by the platelet-derived growth factor kinase inhibitor, tyrphostin AG 1296. We have also shown that the platelet-derived growth factor beta receptor forms a tethered complex with Myc-tagged endothelial differentiation gene 1 (a G-protein-coupled receptor whose agonist is sphingosine 1-phosphate) in cells co-transfected with these receptors. This facilitates platelet-derived growth factor-stimulated tyrosine phosphorylation of the inhibitory G-protein alpha subunit and increases p42/p44 mitogen-activated protein kinase activation. In addition, we found that G-protein-coupled receptor kinase 2 and beta-arrestin I can associate with the platelet-derived growth factor beta receptor. These proteins play an important role in regulating endocytosis of G-protein-coupled receptor signal complexes, which is required for activation of p42/p44 mitogen-activated protein kinase. Thus, platelet-derived growth factor beta receptor signaling may be initiated by G-protein-coupled receptor kinase 2/beta-arrestin I that has been recruited to the platelet-derived growth factor beta receptor by its tethering to a G-protein-coupled receptor(s). These results provide a model that may account for the co-mitogenic effect of certain G-protein-coupled receptor agonists with platelet-derived growth factor on DNA synthesis.


Asunto(s)
Lisofosfolípidos , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/química , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptores de Superficie Celular/química , Línea Celular , Células Cultivadas , Relación Dosis-Respuesta a Droga , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Humanos , Proteína Quinasa 1 Activada por Mitógenos , Toxina del Pertussis , Fosforilación , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Pruebas de Precipitina , Unión Proteica , Proteínas Recombinantes/metabolismo , Transducción de Señal , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Transfección , Tirfostinos/farmacología , Factores de Virulencia de Bordetella/farmacología
16.
J Biol Chem ; 276(16): 13452-60, 2001 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-11278307

RESUMEN

Sphingosine 1-phosphate, lysophosphatidic acid, and phosphatidic acid bind to G-protein-coupled receptors to stimulate intracellular signaling in mammalian cells. Lipid phosphate phosphatases (1, 1a, 2, and 3) are a group of enzymes that catalyze de-phosphorylation of these lipid agonists. It has been proposed that the lipid phosphate phosphatases exhibit ecto activity that may function to limit bioavailability of these lipid agonists at their receptors. In this study, we show that the stimulation of the p42/p44 mitogen-activated protein kinase pathway by sphingosine 1-phosphate, lysophosphatidic acid, and phosphatidic acid, all of which bind to G(i/o)-coupled receptors, is substantially reduced in human embyronic kidney 293 cells transfected with lipid phosphate phosphatases 1, 1a, and 2 but not 3. This was correlated with reduced basal intracellular phosphatidic acid and not ecto lipid phosphate phosphatase activity. These findings were supported by results showing that lipid phosphate phosphatases 1, 1a, and 2 also abrogate the stimulation of p42/p44 mitogen-activated protein kinase by thrombin, a peptide G(i/o)-coupled receptor agonist whose bioavailability at its receptor is not subject to regulation by the phosphatases. Furthermore, the lipid phosphate phosphatases have no effect on the stimulation of p42/p44 mitogen-activated protein kinase by other agents that do not use G-proteins to signal, such as serum factors and phorbol ester. Therefore, these findings show that the lipid phosphate phosphatases 1, 1a, and 2 may function to perturb G-protein-coupled receptor signaling per se rather than limiting bioavailability of lipid agonists at their respective receptors.


Asunto(s)
Sistema de Señalización de MAP Quinasas/fisiología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosfatidato Fosfatasa/metabolismo , Receptores de Superficie Celular/fisiología , Línea Celular , Membrana Celular/enzimología , Proteínas de Unión al GTP/metabolismo , Humanos , Isoenzimas/metabolismo , Riñón , Cinética , Lisofosfolípidos/farmacología , Proteína Quinasa 3 Activada por Mitógenos , Forbol 12,13-Dibutirato/farmacología , Ácidos Fosfatidicos/farmacología , Fosfolipasa D/metabolismo , Receptores de Superficie Celular/efectos de los fármacos , Proteínas Recombinantes/metabolismo , Esfingosina/análogos & derivados , Esfingosina/farmacología , Especificidad por Sustrato , Acetato de Tetradecanoilforbol/farmacología , Transfección , Factores de Virulencia de Bordetella/farmacología
17.
Mol Pharmacol ; 58(2): 413-20, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10908310

RESUMEN

Using cultured airway smooth muscle cells, we showed previously that the platelet-derived growth factor (PDGF) receptor uses the G-protein, G(i), to stimulate Grb-2-associated phosphoinositide 3-kinase (PI3K) activity. We also showed that this was an intermediate step in the activation of p42/p44 mitogen-activated protein kinase (p42/p44 MAPK) by PDGF. We now present two lines of evidence that provide further support for this model. First, we report that PDGF stimulates the G(i)-mediated tyrosine phosphorylation of the Grb-2 adaptor protein, Gab1. This phosphorylation appears to be necessary for association of PI3K1a with the Gab1-Grb-2 complex. Second, PI3K appears to promote the subsequent association of dynamin II (which is involved in clathrin-mediated endocytic processing) with the complex. Furthermore, inhibitors of PI3K and clathrin-mediated endocytosis reduced the PDGF-dependent activation of p42/p44 MAPK, suggesting a role for PI3K in the endocytic signaling process leading to stimulation of p42/p44 MAPK. Together, these results begin to define a common signaling model for certain growth factor receptors (e.g., PDGF, insulin, insulin-like growth factor-1, and fibroblast growth factor) which use G(i) to transmit signals to p42/p44 MAPK.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Músculo Liso/enzimología , Fosfoproteínas/metabolismo , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Animales , Células Cultivadas , Dinaminas , Activación Enzimática , GTP Fosfohidrolasas/fisiología , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Cobayas , Proteínas Sustrato del Receptor de Insulina , Músculo Liso/metabolismo , Fosfatidilinositol 3-Quinasas/fisiología , Fosfoproteínas/química , Fosforilación , Tirosina/metabolismo
18.
Biochem J ; 349(Pt 2): 385-402, 2000 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-10880336

RESUMEN

Sphingosine 1-phosphate is formed in cells in response to diverse stimuli, including growth factors, cytokines, G-protein-coupled receptor agonists, antigen, etc. Its production is catalysed by sphingosine kinase, while degradation is either via cleavage to produce palmitaldehyde and phosphoethanolamine or by dephosphorylation. In this review we discuss the most recent advances in our understanding of the role of the enzymes involved in metabolism of this lysolipid. Sphingosine 1-phosphate can also bind to members of the endothelial differentiation gene (EDG) G-protein-coupled receptor family [namely EDG1, EDG3, EDG5 (also known as H218 or AGR16), EDG6 and EDG8] to elicit biological responses. These receptors are coupled differentially via G(i), G(q), G(12/13) and Rho to multiple effector systems, including adenylate cyclase, phospholipases C and D, extracellular-signal-regulated kinase, c-Jun N-terminal kinase, p38 mitogen-activated protein kinase and non-receptor tyrosine kinases. These signalling pathways are linked to transcription factor activation, cytoskeletal proteins, adhesion molecule expression, caspase activities, etc. Therefore sphingosine 1-phosphate can affect diverse biological responses, including mitogenesis, differentiation, migration and apoptosis, via receptor-dependent mechanisms. Additionally, sphingosine 1-phosphate has been proposed to play an intracellular role, for example in Ca(2+) mobilization, activation of non-receptor tyrosine kinases, inhibition of caspases, etc. We review the evidence for both intracellular and extracellular actions, and extensively discuss future approaches that will ultimately resolve the question of dual action. Certainly, sphingosine 1-phosphate will prove to be unique if it elicits both extra- and intra-cellular actions. Finally, we review the evidence that implicates sphingosine 1-phosphate in pathophysiological disease states, such as cancer, angiogenesis and inflammation. Thus there is a need for the development of new therapeutic compounds, such as receptor antagonists. However, identification of the most suitable targets for drug intervention requires a full understanding of the signalling and action profile of this lysosphingolipid. This article describes where the research field is in relation to achieving this aim.


Asunto(s)
Lisofosfolípidos , Transducción de Señal/fisiología , Esfingosina/fisiología , Animales , Apoptosis/fisiología , Diferenciación Celular/fisiología , División Celular/fisiología , Movimiento Celular/fisiología , Células Cultivadas , Ceramidas/fisiología , Humanos , Esfingolípidos/fisiología , Esfingosina/análogos & derivados
19.
Cell Signal ; 12(11-12): 737-43, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11152959

RESUMEN

Previous studies have demonstrated that a number of biochemical actions of ceramide are mediated through protein kinase signalling pathways, such as p42/p44 mitogen-activated protein kinase (p42/p44 MAPK) and c-Jun N-terminal directed protein kinase (JNK). Ceramide-activated protein kinases, such as the kinase suppressor of Ras (KSR) and protein kinase Czeta (PKCzeta), are involved in the regulation of c-Raf, which promotes sequential activation of MEK-1 and p42/p44 MAPK in mammalian cells. However, in cultured airway smooth muscle (ASM) cells, neither KSR nor PKCzeta are involved in the C2-ceramide (C2-Cer)-dependent activation of this kinase cascade. Instead, we found that C2-Cer utilises a novel pathway involving tyrosine kinases, phosphoinositide 3-kinase (PI3K) and conventional PKC isoform(s). We also found that despite its ability to stimulate p42/p44 MAPK, C2-Cer inhibited platelet-derived growth factor (PDGF)-stimulated DNA synthesis. The possibility that growth arrest could be mediated by JNK was discounted on the basis that PDGF, as well as ceramide, stimulated JNK in these cells. Therefore, growth arrest in response to ceramide is mediated by an alternative mechanism.


Asunto(s)
Ceramidas/farmacología , Lisofosfolípidos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Músculo Liso/efectos de los fármacos , Sistema Respiratorio/enzimología , Esfingosina/análogos & derivados , Animales , Permeabilidad de la Membrana Celular , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Cromonas/farmacología , Activación Enzimática/efectos de los fármacos , Cobayas , Isoenzimas/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos , Proteína Quinasa 3 Activada por Mitógenos , Morfolinas/farmacología , Músculo Liso/citología , Músculo Liso/enzimología , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Factor de Crecimiento Derivado de Plaquetas/farmacología , Pruebas de Precipitina , Proteína Quinasa C/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-raf/metabolismo , Quinazolinas , Sistema Respiratorio/citología , Transducción de Señal/efectos de los fármacos , Esfingosina/farmacología , Tirfostinos/farmacología
20.
Pharmacol Ther ; 88(2): 115-31, 2000 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11150592

RESUMEN

Sphingosine 1-phosphate (S1P) is stored in and released from platelets in response to cell activation. However, recent studies show that it is also released from a number of cell types, where it can function as a paracrine/autocrine signal to regulate cell proliferation, differentiation, survival, and motility. This review discusses the role of S1P in cellular regulation, both at the molecular level and in terms of health and disease. The main biochemical routes for S1P synthesis (sphingosine kinase) and degradation (S1P lyase and S1P phosphatase) are described. The major focus is on the ability of S1P to bind to a novel family of G-protein-coupled receptors (endothelial differentiation gene [EDG]-1, -3, -5, -6, and -8) to elicit signal transduction (via G(q)-, G(i)-, G(12)-, G(13)-, and Rho-dependent routes). Effector pathways regulated by S1P are divergent, such as extracellular signal-regulated kinase, p38 mitogen-activated protein kinase, phospholipases C and D, adenylyl cyclase, and focal adhesion kinase, and occur in multiple cell types, such as immune cells, neurones, smooth muscle, etc. This provides a molecular basis for the ability of S1P to act as a pleiotropic bioactive lipid with an important role in cellular regulation. We also give an account of the expanding role for S1P in health and disease; in particular, with regard to its role in atherosclerosis, angiogenesis, cancer, and inflammation. Finally, we describe future directions for S1P research and novel approaches whereby S1P signalling can be manipulated for therapeutic intervention in disease.


Asunto(s)
Arteriosclerosis/fisiopatología , Diferenciación Celular/fisiología , Reguladores de Proteínas de Unión al GTP/fisiología , Regulación de la Expresión Génica , Lisofosfolípidos , Neovascularización Patológica/fisiopatología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Transducción de Señal , Esfingosina/análogos & derivados , Esfingosina/genética , Esfingosina/farmacología , Aldehído-Liasas/metabolismo , Animales , Endotelio/fisiología , Humanos , Inflamación , Neoplasias/fisiopatología , Esfingosina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA