Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 110
Filtrar
1.
Int J Mol Sci ; 22(4)2021 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-33671654

RESUMEN

Sphingosine-1-phosphate (S1P), is a signaling sphingolipid which acts as a bioactive lipid mediator. We assessed whether S1P had multiplex effects in regulating the large-conductance Ca2+-activated K+ channel (BKCa) in catecholamine-secreting chromaffin cells. Using multiple patch-clamp modes, Ca2+ imaging, and computational modeling, we evaluated the effects of S1P on the Ca2+-activated K+ currents (IK(Ca)) in bovine adrenal chromaffin cells and in a pheochromocytoma cell line (PC12). In outside-out patches, the open probability of BKCa channel was reduced with a mean-closed time increment, but without a conductance change in response to a low-concentration S1P (1 µM). The intracellular Ca2+ concentration (Cai) was elevated in response to a high-dose (10 µM) but not low-dose of S1P. The single-channel activity of BKCa was also enhanced by S1P (10 µM) in the cell-attached recording of chromaffin cells. In the whole-cell voltage-clamp, a low-dose S1P (1 µM) suppressed IK(Ca), whereas a high-dose S1P (10 µM) produced a biphasic response in the amplitude of IK(Ca), i.e., an initial decrease followed by a sustained increase. The S1P-induced IK(Ca) enhancement was abolished by BAPTA. Current-clamp studies showed that S1P (1 µM) increased the action potential (AP) firing. Simulation data revealed that the decreased BKCa conductance leads to increased AP firings in a modeling chromaffin cell. Over a similar dosage range, S1P (1 µM) inhibited IK(Ca) and the permissive role of S1P on the BKCa activity was also effectively observed in the PC12 cell system. The S1P-mediated IK(Ca) stimulation may result from the elevated Cai, whereas the inhibition of BKCa activity by S1P appears to be direct. By the differentiated tailoring BKCa channel function, S1P can modulate stimulus-secretion coupling in chromaffin cells.


Asunto(s)
Calcio/metabolismo , Células Cromafines/metabolismo , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/metabolismo , Lisofosfolípidos/metabolismo , Esfingosina/análogos & derivados , Animales , Bovinos , Sistema Libre de Células , Células Cromafines/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Electrofisiología/métodos , Lisofosfolípidos/administración & dosificación , Lisofosfolípidos/farmacología , Células PC12 , Ratas , Esfingosina/administración & dosificación , Esfingosina/metabolismo , Esfingosina/farmacología
2.
Acta Neuropsychiatr ; 33(1): 15-21, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32967746

RESUMEN

OBJECTIVE: G protein-coupled receptor 55 (GPR55) is an orphan G protein-coupled receptor with various physiological functions. Recent evidence suggests that this receptor may be involved in the control of motor functions. Therefore, in the present study, we evaluated the effects of intra-striatal administration of GPR55 selective ligands in a rat model of Parkinson's disease. METHODS: Experimental Parkinson was induced by unilateral intra-striatal administration of 6-hydroxydopamine (6-OHDA, 10 µg/rat). L-α-lysophosphatidylinositol (LPI, 1 and 5 µg/rat), an endogenous GPR55 agonist, and ML193 (1 and 5 µg/rat), a selective GPR55 antagonist, were injected into the striatum of 6-OHDA-lesioned rats. Motor performance and balance skills were evaluated using the accelerating rotating rod and the ledged beam tests. The sensorimotor function of the forelimbs and locomotor activity were assessed by the adhesive removal and open field tests, respectively. RESULTS: 6-OHDA-lesioned rats had impaired behaviours in all tests. Intra-striatal administration of LPI in 6-OHDA-lesioned rats increased time on the rotarod, decreased latency to remove the label, with no significant effect on slip steps, and locomotor activity. Intra-striatal administration of ML193 also increased time on the rotarod, decreased latency to remove the label and slip steps in 6-OHDA-lesioned rats mostly at the dose of 1 µg/rat. CONCLUSIONS: This study suggests that the striatal GPR55 is involved in the control of motor functions. However, considering the similar effects of GPR55 agonist and antagonist, it may be concluded that this receptor has a modulatory role in the control of motor deficits in an experimental model of Parkinson.


Asunto(s)
Cuerpo Estriado/efectos de los fármacos , Enfermedad de Parkinson/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Adrenérgicos/administración & dosificación , Adrenérgicos/farmacología , Animales , Estudios de Casos y Controles , Modelos Animales de Enfermedad , Ligandos , Lisofosfolípidos/administración & dosificación , Lisofosfolípidos/farmacología , Masculino , Actividad Motora/efectos de los fármacos , Oxidopamina/administración & dosificación , Oxidopamina/farmacología , Enfermedad de Parkinson/fisiopatología , Ratas , Ratas Wistar , Receptores de Cannabinoides
3.
Acta Vet Hung ; 67(4): 578-587, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31842605

RESUMEN

Sphingosine-1-phosphate (S1P) has been reported as a matriptase activator. The aim of this study was to reveal if S1P can influence hepcidin production. Furthermore, we investigated how S1P can affect the viability and the redox status of primary hepatocytes. Rat primary hepatocytes were cultivated for 72 h and were treated with 50, 200, 1000 ng/ml S1P. Cell-free supernatants were collected every 24 h. Cell viability was tested by a colorimetric method using tetrazolium compound (MTS). The hepcidin levels in the cell-free supernatants were examined with hepcidin sandwich ELISA to determine the effect of S1P on the hepcidin-modulating ability of matriptase. In order to estimate the extent of S1P-generated oxidative stress, extracellular H2O2 measurements were performed by the use of fluorescent dye. Based on the findings, S1P treatment did not cause cell death for 72 h at concentrations up to 1000 ng/ml. S1P did not influence the extracellular H2O2 production for 72 h. The hepcidin levels were significantly suppressed in hepatocytes exposed to S1P treatment. Further studies would be needed to explore the exact mechanism of action of S1P.


Asunto(s)
Hepatocitos/efectos de los fármacos , Hepcidinas/biosíntesis , Lisofosfolípidos/administración & dosificación , Serina Endopeptidasas/metabolismo , Esfingosina/análogos & derivados , Animales , Peróxido de Hidrógeno/metabolismo , Masculino , Modelos Animales , Ratas , Ratas Sprague-Dawley , Esfingosina/administración & dosificación
4.
J Anim Physiol Anim Nutr (Berl) ; 102(6): 1521-1532, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30255521

RESUMEN

The effects of dietary supplemental lysophospholipids (LPLs) and vitamin C (VC) on performance, activity of antioxidant enzymes, and thyroid hormones of broiler chickens reared under thermoneutral and high ambient temperatures were evaluated. A total of 1,680 broiler chicks (Cobb 500) in finishing rearing period (days 21-38 of age) were allotted to two groups: thermoneutral (TN) and heat stress (HS). In the TN group, 480 chicks were subjected to four treatments with four replicates (n = 30) and maintained in usual ambient temperature (24 ± 1°C). In HS group, the remaining 1,200 chicks were subjected to four treatments with 10 replicates (n = 30) and exposed to high ambient temperature (34 ± 1°C for 8 hr daily). In both groups, four iso-caloric and iso-nitrogenous experimental diets based on a 2 × 2 factorial arrangements including supplemental LPLs (0 or 1,000 mg/kg) and VC (0 or 500 mg/kg) were formulated and used. Supplemental LPLs decreased (p < 0.05) body weight gain and increased FCR in the TN and HS groups. In the TN group, increased (p < 0.05) serum glucose was observed in chickens fed with dietary supplemental VC. In the HS group, decreased (p < 0.05) total protein concentration was detected in birds fed with supplemental LPLs. In both TN and HS groups, decreased (p < 0.05) uric acid concentration was detected in chicks fed with the VC-supplemented diets. A significant (p < 0.05) interaction between LPLs and VC on lactate concentration in the TN group was observed. In the HS group, decreased breast malondialdehyde concentration was detected in birds fed with the VC-supplemented diet. In the TN group, increased serum total antioxidant status was detected in birds fed with the LPLs-supplemented diet. In conclusion, LPLs improved oxidative stability and increased the antioxidant capacity of the serum. In addition, vitamin C modified heat stress and reduced lipid peroxidation.


Asunto(s)
Antioxidantes/metabolismo , Ácido Ascórbico/farmacología , Pollos/sangre , Calor , Peroxidación de Lípido/efectos de los fármacos , Lisofosfolípidos/farmacología , Crianza de Animales Domésticos , Animales , Ácido Ascórbico/administración & dosificación , Pollos/fisiología , Suplementos Dietéticos , Femenino , Lisofosfolípidos/administración & dosificación , Masculino , Malondialdehído/sangre , Malondialdehído/metabolismo , Músculo Esquelético/metabolismo , Distribución Aleatoria , Hormonas Tiroideas/sangre
5.
J Lipid Res ; 58(4): 636-648, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28188148

RESUMEN

The pathophysiology of human keratoconus (KC), a bilateral progressive corneal disease leading to protrusion of the cornea, stromal thinning, and scarring, is not well-understood. In this study, we investigated a novel sphingolipid (SPL) signaling pathway through which KC may be regulated. Using human corneal fibroblasts (HCFs) and human KC cells (HKCs), we examined the SPL pathway modulation. Both cell types were stimulated by the three transforming growth factor (TGF)-ß isoforms: TGF-ß1 (T1), TGF-ß2 (T2), and TGF-ß3 (T3). All samples were analyzed using lipidomics and real-time PCR. Our data showed that HKCs have increased levels of signaling SPLs, ceramide (Cer), and sphingosine 1-phosphate (S1P). Treatment with T1 reversed the increase in Cer in HKCs and treatment with T3 reversed the increase in S1P. S1P3 receptor mRNA levels were also significantly upregulated in HKCs, but were reduced to normal levels following T3 treatment. Furthermore, stimulation with Cer and S1P led to significant upregulation of fibrotic markers in HCFs, but not in HKCs. Additionally, stimulation with a Cer synthesis inhibitor (FTY720) led to significant downregulation of specific fibrotic markers in HKCs (TGF-ß1, collagen type III, and α smooth muscle actin) without an effect on healthy HCFs, suggesting a causative role of Cer and S1P in fibrogenesis. Overall, this study suggests an association of the SPL signaling pathway in KC disease and its relation with the TGF-ß pathway.


Asunto(s)
Ceramidas/genética , Queratocono/genética , Lisofosfolípidos/biosíntesis , Esfingolípidos/genética , Esfingosina/análogos & derivados , Línea Celular , Ceramidas/administración & dosificación , Córnea/metabolismo , Córnea/patología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/fisiología , Clorhidrato de Fingolimod/administración & dosificación , Humanos , Queratocono/patología , Lisofosfolípidos/administración & dosificación , ARN Mensajero/genética , Transducción de Señal , Esfingolípidos/aislamiento & purificación , Esfingolípidos/metabolismo , Esfingosina/administración & dosificación , Esfingosina/biosíntesis , Factor de Crecimiento Transformador beta1/administración & dosificación , Factor de Crecimiento Transformador beta2/administración & dosificación , Factor de Crecimiento Transformador beta3/administración & dosificación
6.
Stem Cells ; 34(3): 551-64, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26800320

RESUMEN

Ovarian cancer shows high mortality due to development of resistance to chemotherapy and relapse. Cancer stem cells (CSCs) have been suggested to be a major contributor in developing drug resistance and relapse in ovarian cancer. In this study, we isolated CSCs through sphere culture of A2780, SKOV3, OVCAR3 epithelial ovarian cancer cells and primary ovarian cancer cells from patients. We identified heat-stable factors secreted from ovarian CSCs stimulated migration and proliferation of CSCs. Mass spectrometry and ELISA analysis revealed that lysophosphatidic acid (LPA) was significantly elevated in CSC culture media compared with non-CSC culture media. Treatment of CSCs with LPA resulted in augmented CSC characteristics such as sphere-forming ability, resistance to anticancer drugs, tumorigenic potential in xenograft transplantation, and high expression of CSC-associated genes, including OCT4, SOX2, and aldehyde dehydrogenase 1. Treatment of CSCs with LPA receptor 1-specific inhibitors or silencing of LPA receptor 1 expression abrogated the LPA-stimulated CSC properties. Autotaxin, an LPA-producing enzyme, is highly secreted from ovarian CSCs, and pharmacological inhibition or knockdown of autotaxin markedly attenuated the LPA-producing, tumorigenic, and drug resistance potentials of CSCs. Clinicopathological analysis showed a significant survival disadvantage of patients with positive staining of autotaxin. In addition, we further identified that AKT1 activity was upregulated in ovarian CSCs through an LPA-dependent mechanism and silencing of AKT1 expression led to suppression of CSC characteristics. These results suggest that autotaxin-LPA-LPA receptor 1-AKT1 signaling axis is critical for maintaining CSC characteristics through an autocrine loop and provide a novel therapeutic target for ovarian CSCs.


Asunto(s)
Lisofosfolípidos/administración & dosificación , Neoplasias Glandulares y Epiteliales/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Neoplasias Ováricas/tratamiento farmacológico , Hidrolasas Diéster Fosfóricas/genética , Receptores del Ácido Lisofosfatídico/genética , Ataxina-1/genética , Comunicación Autocrina/efectos de los fármacos , Carcinoma Epitelial de Ovario , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Medios de Cultivo Condicionados , Resistencia a Antineoplásicos/genética , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Glandulares y Epiteliales/genética , Neoplasias Glandulares y Epiteliales/patología , Células Madre Neoplásicas/patología , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Proteínas Proto-Oncogénicas c-akt/genética , Transducción de Señal/efectos de los fármacos
7.
Exp Mol Pathol ; 103(2): 210-217, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28986246

RESUMEN

Sphingosine 1-phosphate (S1P), via binding to its specific receptors of S1PR1, participates in the regulation of both innate and adaptive immunity. Recent reports have identified S1P as a messenger mediating inflammation. However, roles of S1P in Coxsackievirus B3 (CVB3)-induced myocarditis were largely unknown. Here, we investigated the effect of S1P treatment on CVB3-induced myocarditis in vivo. We found that CVB3 infection downregulated S1PR1 expression in spleen and decreased the proportion of invariant natural killer T cells (iNKT) in CD3 positive T cells both in spleen and in blood from left ventricle, which accompanied by severe inflammation lesions and more virus capsid protein (VP1) expression in heart tissue. In comparison, S1P supply upregulated iNKT in the spleen and in blood from left ventricle, which represented the strengthening of anti-inflammatory effects. Indeed, inflammation infiltration, VP1 expression and apoptosis in the myocardium was all downregulated. These results demonstrated that S1P supplement could alleviate CVB3-induced myocarditis.


Asunto(s)
Infecciones por Coxsackievirus/complicaciones , Enterovirus Humano B/patogenicidad , Lisofosfolípidos/farmacología , Miocarditis/prevención & control , Células T Asesinas Naturales/inmunología , Esfingosina/análogos & derivados , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Infecciones por Coxsackievirus/virología , Modelos Animales de Enfermedad , Lisofosfolípidos/administración & dosificación , Masculino , Ratones , Ratones Endogámicos BALB C , Miocarditis/etiología , Miocarditis/metabolismo , Células T Asesinas Naturales/efectos de los fármacos , Receptores de Lisoesfingolípidos/genética , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/administración & dosificación , Esfingosina/farmacología , Receptores de Esfingosina-1-Fosfato , Bazo/efectos de los fármacos , Bazo/inmunología , Bazo/patología
8.
Cell Mol Biol (Noisy-le-grand) ; 63(4): 16-22, 2017 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-28478798

RESUMEN

Sphingosine-1-phosphate (S1P) is a sphingolipid in plasma that plays a critical role in cardiovascular and immune systems. Endothelial surface glycocalyx (ESG) decorating the inner wall of blood vessels is a regulator of multiple vascular functions. To test the hypothesis that S1P can reduce tumor cell adhesion to microvessel walls by protecting the ESG, we quantified the ESG and MDA-MB-231 tumor cell adhesion in the presence and absence of 1µM S1P, and in the presence of the matrix metalloproteinase (MMP) inhibitor in post-capillary venules of rat mesentery. We also measured the microvessel permeability to albumin as an indicator for the microvessel wall integrity. In the absence of S1P, ESG was ~10% of that in the presence of S1P, whereas adherent tumor cells and the permeability to albumin and were ~3.5-fold (after 30 min adhesion) and ~7.7-fold that in the presence of S1P, respectively. In the presence of the MMP inhibitor, the results are similar to those in the presence of S1P. Our results conform to the hypothesis that protecting ESG by S1P inhibits MDA-MB-231 tumor cell adhesion to the microvessel wall.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Adhesión Celular/efectos de los fármacos , Lisofosfolípidos/administración & dosificación , Microvasos/efectos de los fármacos , Esfingosina/análogos & derivados , Animales , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Adhesión Celular/genética , Línea Celular Tumoral , Células Endoteliales/efectos de los fármacos , Femenino , Glicocálix/metabolismo , Humanos , Microvasos/metabolismo , Ratas , Esfingosina/administración & dosificación , Tensoactivos/metabolismo
9.
J Biol Chem ; 290(10): 6349-60, 2015 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-25564613

RESUMEN

Here we report that VEGF-A and IGF-1 differ in their ability to stabilize newly formed blood vessels and endothelial cell tubes. Although VEGF-A failed to support an enduring vascular response, IGF-1 stabilized neovessels generated from primary endothelial cells derived from various vascular beds and mouse retinal explants. In these experimental systems, destabilization/regression was driven by lysophosphatidic acid (LPA). Because previous studies have established that Erk antagonizes LPA-mediated regression, we considered whether Erk was an essential component of IGF-dependent stabilization. Indeed, IGF-1 lost its ability to stabilize neovessels when the Erk pathway was inhibited pharmacologically. Furthermore, stabilization was associated with prolonged Erk activity. In the presence of IGF-1, Erk activity persisted longer than in the presence of VEGF or LPA alone. These studies reveal that VEGF and IGF-1 can have distinct inputs in the angiogenic process. In contrast to VEGF, IGF-1 stabilizes neovessels, which is dependent on Erk activity and associated with prolonged activation.


Asunto(s)
Vasos Sanguíneos/crecimiento & desarrollo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Vasos Retinianos/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Vasos Sanguíneos/metabolismo , Movimiento Celular/genética , Endotelio Vascular/crecimiento & desarrollo , Endotelio Vascular/metabolismo , Factor I del Crecimiento Similar a la Insulina/genética , Lisofosfolípidos/administración & dosificación , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Neovascularización Fisiológica , Vasos Retinianos/crecimiento & desarrollo , Factor A de Crecimiento Endotelial Vascular/genética
10.
IUBMB Life ; 68(5): 365-75, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26959531

RESUMEN

Sphingosine-1-phosphate (S1P) is emerging to have hypoxic preconditioning potential in various preclinical studies. The study aims to evaluate the preclinical preconditioning efficacy of exogenously administered S1P against acute hypobaric hypoxia (HH)-induced pathological disturbances. Male Sprague Dawley rats (200 ± 20 g) were preconditioned with 1, 10, and 100 µg/kg body weight (b.w.) S1P (i.v.) for three consecutive days. On the third day, S1P preconditioned animals, along with hypoxia control animals, were exposed to HH equivalent to 7,620 m (280 mm Hg) for 6 h. Postexposure status of cardiac energy production, circulatory vasoactive mediators, pulmonary and cerebral oxidative damage, and inflammation were assessed. HH exposure led to cardiac energy deficit indicated by low ATP levels and pronounced AMPK activation levels, raised circulatory levels of brain natriuretic peptide and endothelin-1 with respect to total nitrate (NOx), redox imbalance, inflammation, and alterations in NOx levels in the pulmonary and cerebral tissues. These pathological precursors have been routinely reported to be coincident with high-altitude diseases. Preconditioning with S1P, especially 1 µg/kg b.w. dose, was seen to reverse the manifestation of these pathological disturbances. The protective efficacy could be attributed, at least in part, to enhanced activity of cardioprotective protein kinase C and activation of small GTPase Rac1, which led to further induction of hypoxia-adaptive molecular mediators: hypoxia-inducible factor (HIF)-1α and Hsp70. This is a first such report, to the best of our knowledge, elucidating the mechanism of exogenous S1P-mediated HIF-1α/Hsp70 induction. Conclusively, systemic preconditioning with 1 µg/kg b.w. S1P in rats protects against acute HH-induced pathological disturbances. © 2016 IUBMB Life 68(5):365-375, 2016.


Asunto(s)
Hipoxia/prevención & control , Lisofosfolípidos/administración & dosificación , Esfingosina/análogos & derivados , Animales , Citocinas/sangre , Evaluación Preclínica de Medicamentos , Endotelina-1/sangre , Metabolismo Energético , Proteínas HSP70 de Choque Térmico/metabolismo , Hipoxia/sangre , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Miocardio/metabolismo , Miocardio/patología , Péptido Natriurético Encefálico/sangre , Óxido Nítrico/sangre , Oxidación-Reducción , Estrés Oxidativo , Estabilidad Proteica , Ratas Sprague-Dawley , Esfingosina/administración & dosificación , Proteína de Unión al GTP rac1/metabolismo
11.
Nanomedicine ; 12(7): 1775-1784, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27125435

RESUMEN

Effective treatment of brain disorders requires a focus on improving drug permeability across the blood-brain barrier (BBB). Herein, we examined the pharmacokinetic properties of negatively charged iron oxide nanoparticles (IONPs) and the capability of using lysophosphatidic acid (LPA) to transiently disrupt the tight junctions and allow IONPs to enter the brain. Under normal conditions, IONPs had a plasma half-life of six minutes, with the liver and spleen being the major organs of deposition. Treatment with LPA enhanced accumulation of IONPs in the brain and spleen (approximately 4-fold vs. control). LPA and IONP treated mice revealed no sign of peripheral immune cell infiltration in the brain and no significant activation of microglia or astrocytes. These studies show improved delivery efficiency of IONPs following LPA administration. Our findings suggest transient disruption of the BBB may be a safe and effective method for increasing IONP delivery to the brain.


Asunto(s)
Barrera Hematoencefálica , Lisofosfolípidos/farmacología , Nanopartículas , Animales , Encéfalo , Compuestos Férricos , Lisofosfolípidos/administración & dosificación , Ratones , Bazo , Distribución Tisular
12.
Br Poult Sci ; 57(6): 788-798, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27494540

RESUMEN

Three experiments with a 2 × 2 × 2 factorial arrangement were conducted to evaluate maize-based diets for broilers containing different lipid sources [soybean oil (S) or beef tallow (T)] supplemented with or without lysophospholipids and organic acids on nutrient balance (Experiment I, evaluation period of 10-14 d), on liver concentration of fat-soluble vitamins, on jejunal microbiota (Experiment II, sampling at d 14) and on performance (Experiment III, accumulated periods of 1-14, 1-21 and 1-42 d). A total of 1344 male chicks were used. In each experiment, the birds were allotted in a completely randomised design with 8 replications. The lysophospholipids were mainly composed of lysolecithins and the organic acids blend was constituted by lactic (40%), acetic (7%) and butyric acids (1%). An interaction between lipid sources and lysophospholipids was observed on faecal apparent digestibility of lipid (ADL), which improved with lysophospholipids addition in T diets. Broilers fed on S had higher ADL and faecal apparent digestibility of nitrogen-corrected gross energy (ADGEN). It was not possible to demonstrate a significant treatment effect on the liver concentration of vitamins A and E, even with the differences in fatty acid profile between S and T. Enterobacteria values were below the detection threshold. Lysophospholipid supplementation reduced gram-positive cocci in T-fed birds. S diets promoted lower total anaerobe counts compared with T diets, independent of additives. S diets increased BW gain and feed:gain ratio in all evaluation periods. Lysophospholipids and organic acids improved feed:gain ratio at 1-21 d in T diets. Furthermore, main effects were observed for lysophospholipids and organic acids at 1-42 d, which increased BW gain and improved feed:gain ratio, respectively. No positive interactions between additives were found.


Asunto(s)
Ácido Acético/metabolismo , Fenómenos Fisiológicos Nutricionales de los Animales , Ácido Butírico/metabolismo , Pollos , Dieta/veterinaria , Ácido Láctico/metabolismo , Lisofosfolípidos/metabolismo , Ácido Acético/administración & dosificación , Alimentación Animal/análisis , Animales , Ácido Butírico/administración & dosificación , Pollos/crecimiento & desarrollo , Pollos/microbiología , Suplementos Dietéticos/análisis , Grasas/análisis , Yeyuno/microbiología , Ácido Láctico/administración & dosificación , Hígado/química , Lisofosfolípidos/administración & dosificación , Masculino , Distribución Aleatoria , Aceite de Soja/análisis
13.
J Lipid Res ; 56(9): 1747-61, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26175473

RESUMEN

Ganglioside GM2 is the major lysosomal storage compound of Tay-Sachs disease. It also accumulates in Niemann-Pick disease types A and B with primary storage of SM and with cholesterol in type C. Reconstitution of GM2 catabolism with ß-hexosaminidase A and GM2 activator protein (GM2AP) at uncharged liposomal surfaces carrying GM2 as substrate generated only a physiologically irrelevant catabolic rate, even at pH 4.2. However, incorporation of anionic phospholipids into the GM2 carrying liposomes stimulated GM2 hydrolysis more than 10-fold, while the incorporation of plasma membrane stabilizing lipids (SM and cholesterol) generated a strong inhibition of GM2 hydrolysis, even in the presence of anionic phospholipids. Mobilization of membrane lipids by GM2AP was also inhibited in the presence of cholesterol or SM, as revealed by surface plasmon resonance studies. These lipids also reduced the interliposomal transfer rate of 2-NBD-GM1 by GM2AP, as observed in assays using Förster resonance energy transfer. Our data raise major concerns about the usage of recombinant His-tagged GM2AP compared with untagged protein. The former binds more strongly to anionic GM2-carrying liposomal surfaces, increases GM2 hydrolysis, and accelerates intermembrane transfer of 2-NBD-GM1, but does not mobilize membrane lipids.


Asunto(s)
Proteína Activadora de G (M2)/metabolismo , Gangliósido G(M2)/metabolismo , Liposomas/metabolismo , Lípidos de la Membrana/metabolismo , Ceramidas/metabolismo , Colesterol/genética , Colesterol/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Proteína Activadora de G (M2)/genética , Células HEK293 , Humanos , Hidrólisis/efectos de los fármacos , Lisofosfolípidos/administración & dosificación , Lípidos de la Membrana/genética , Monoglicéridos/administración & dosificación , Enfermedades de Niemann-Pick/genética , Enfermedades de Niemann-Pick/metabolismo , Enfermedades de Niemann-Pick/patología , Esfingomielinas/metabolismo , Resonancia por Plasmón de Superficie , Enfermedad de Tay-Sachs/genética , Enfermedad de Tay-Sachs/metabolismo , Enfermedad de Tay-Sachs/patología , Cadena alfa de beta-Hexosaminidasa/metabolismo
14.
J Cell Physiol ; 230(3): 702-15, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25201048

RESUMEN

Sphingosine-1-phosphate (S1P) has been shown to regulate cyclooxygenase-2 (COX-2)/prostaglandin E2 (PGE2 ) expression and IL-6 secretion in various respiratory diseases. However, the mechanisms underlying S1P-induced COX-2 expression and PGE2 production in human tracheal smooth muscle cells (HTSMCs) remain unclear. Here we demonstrated that S1P markedly induced COX-2 expression. S1P also induced PGE2 and IL-6 secretion which were reduced by the inhibitors of COX-2 (NS-398 and celecoxib). Pretreatment with the inhibitor of S1PR1 (W123), S1PR3 (CAY10444), c-Src (PP1), PYK2 (PF431396), MEK1/2 (U0126), p38 MAPK (SB202190), JNK1/2 (SP600125), or AP-1 (Tanshinone IIA) and transfection with siRNA of S1PR1, S1PR3, c-Src, PYK2, p38, p42, JNK2, c-Jun, or c-Fos reduced S1P-induced COX-2 expression and PGE2 /IL-6 secretion. Moreover, S1P induced c-Src, PYK2, p42/p44 MAPK, JNK1/2, p38 MAPK, and c-Jun phosphorylation. We observed that S1P-induced p42/p44 MAPK and JNK1/2, but not p38 MAPK activation was mediated via a c-Src/PYK2-dependent pathway. S1P also enhanced c-Fos, but not c-Jun mRNA and protein expression and the AP-1 promoter activity. S1P-induced c-Fos mRNA and protein expression, c-Jun phosphorylation, and AP-1 promoter activity was reduced by W123, CAY10444, PP1, PF431396, U0126, SP600125, or SB202190. These results demonstrated that S1P-induced COX-2 expression and PGE2 /IL-6 generation was mediated through S1PR1/3/c-Src/PYK2/p42/p44 MAPK- or JNK1/2- and S1PR1/3/c-Src/p38 MAPK-dependent AP-1 activation.


Asunto(s)
Ciclooxigenasa 2/biosíntesis , Interleucina-6/metabolismo , Lisofosfolípidos/administración & dosificación , Esfingosina/análogos & derivados , Factor de Transcripción AP-1/biosíntesis , Familia-src Quinasas/biosíntesis , Proteína Tirosina Quinasa CSK , Línea Celular , Dinoprostona/biosíntesis , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , FN-kappa B/metabolismo , Transducción de Señal/efectos de los fármacos , Esfingosina/administración & dosificación , Factor de Transcripción AP-1/metabolismo , Activación Transcripcional/efectos de los fármacos
15.
Neurol Sci ; 36(11): 2027-33, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26169757

RESUMEN

Lysophosphatidic acid (LPA) is a bioactive phospholipid that activates at least five known G-protein-coupled receptors (GPCRs): LPA1-LPA5. The nervous system is a major locus for LPA1 expression. LPA has been shown to regulate neuronal proliferation, migration, and differentiation during central nervous system development as well as neuronal survival. Furthermore, deficient LPA signaling has been implicated in several neurological disorders including neuropathic pain and schizophrenia. Parkinson's disease (PD) is a neurodegenerative movement disorder that results from the loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNc). The specific molecular pathways that lead to DA neuron degeneration, however, are poorly understood. The influence of LPA in the differentiation of mesenchymal stem cells (MSCs) into DA neurons in vitro and LPA1 expression in a 6-hydroxydopamine (6-OHDA) lesion model of PD in vivo were examined in the present study. LPA induced neuronal differentiation in 80.2 % of the MSC population. These MSCs developed characteristic neuronal morphology and expressed the neuronal marker, neuron-specific enolase (NSE), while expression of the glial marker, glial fibrillary acidic protein (GFAP), was absent. Moreover, 27.6 % of differentiated MSCs were positive for tyrosine hydroxylase (TH), a marker for DA neurons. In the 6-OHDA PD rat model, LPA1 expression in the substantia nigra was significantly reduced compared to control. These results suggest LPA signaling via activation of LPA1 may be necessary for DA neuron development and survival. Furthermore, reduced LPA/LPA1 signaling may be involved in DA neuron degeneration thus contributing to the pathogenesis of PD.


Asunto(s)
Neuronas Dopaminérgicas/fisiología , Lisofosfolípidos/metabolismo , Neurogénesis/fisiología , Trastornos Parkinsonianos/fisiopatología , Receptores del Ácido Lisofosfatídico/metabolismo , Animales , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Fármacos del Sistema Nervioso Central/administración & dosificación , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/patología , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Lisofosfolípidos/administración & dosificación , Masculino , Células Madre Mesenquimatosas/patología , Células Madre Mesenquimatosas/fisiología , Plexo Mientérico/metabolismo , Neurogénesis/efectos de los fármacos , Oxidopamina , Trastornos Parkinsonianos/patología , Fosfopiruvato Hidratasa/metabolismo , Ratas Sprague-Dawley , Transducción de Señal , Sustancia Negra/patología , Sustancia Negra/fisiopatología , Tirosina 3-Monooxigenasa/metabolismo
16.
An Acad Bras Cienc ; 87(4): 2255-63, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26536854

RESUMEN

The number of sheep flocks in Brazil is increasing. It is known that lambs must be slaughtered when young for producing quality meat. The current study evaluated the inclusion of protected methionine, protected lysine, lysophospholipid and amylolytic enzymes in a diet to lambs and their effects on weight gain and quantitative carcass traits at slaughtering. Eighty non-castrated male crossbred Dorper x Santa Inês lambs, 20.57 ± 4.33 kg live weight, were used. The feedlot lasted 64 days and 60 animals were slaughtered. There were no differences for live weight, daily feed intake, feed conversion and average daily weight gain at the first 28 days of feedlot. From the 28th day lysophospholipid treatment presented the highest live weight. Lysophospholipid and amylolytic enzyme presented the best performance in average daily gain, followed by protected methionine, control and protected lysine. Lysophospholipid treatment presented higher daily feed intake rates than protected lysine and protected methionine. Feed conversion was lower for amylolytic enzyme and higher for control. No changing in carcass traits was reported due to additives. Better performance may be achieved with feedlot lambs fed on diets with the addition of amylolytic enzyme and lysophospholipid at the finishing phase.


Asunto(s)
Alimentación Animal/análisis , Suplementos Dietéticos , Ovinos/crecimiento & desarrollo , Aumento de Peso/fisiología , Amilasas/administración & dosificación , Animales , Lisina/administración & dosificación , Lisofosfolípidos/administración & dosificación , Masculino , Metionina/administración & dosificación
17.
Genet Mol Res ; 14(4): 13843-51, 2015 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-26535699

RESUMEN

In this study, we investigated the effects of sphingosine-1-phosphate (S1P) combined with myoblast transplantation on the treatment of acute myocardial infarction and provided a foundation for its clinical application. A rat model of acute myocardial infarction was established by ligating the anterior descending branch of the coronary artery. Serum-free media, myoblasts, myoblasts with S1P liposomes, or myoblasts with liposomes were then injected into the infarcted area. Apoptosis of the transplanted cells was assessed after 24 and 48 h, and changes in heart function and myocardial infarction area were assessed after 4 weeks. After transplantation of S1P into myoblasts, myocardial function was improved compared to that in the other groups. Specifically, the apoptosis of transplanted cells and the area of myocardial infarction decreased significantly (P < 0.01), while cardiac function significantly improved (P < 0.01). The efficacy of S1P and myoblast transplantation on acute myocardial infarction was significantly better than that in the control group (i.e., injection of myoblasts and liposomes) and the serum-free medium group, demonstrating the feasibility of joint S1P and myoblast transplantation for treating myocardial infarction.


Asunto(s)
Lisofosfolípidos/farmacología , Mioblastos/trasplante , Infarto del Miocardio/terapia , Esfingosina/análogos & derivados , Animales , Apoptosis , Biomarcadores , Modelos Animales de Enfermedad , Inmunohistoquímica , Liposomas , Lisofosfolípidos/administración & dosificación , Mioblastos/metabolismo , Mioblastos Esqueléticos/trasplante , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Ratas , Esfingosina/administración & dosificación , Esfingosina/farmacología , Función Ventricular Izquierda
18.
J Cell Physiol ; 229(7): 927-42, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24318513

RESUMEN

Glaucoma, a prevalent blinding disease is commonly associated with increased intraocular pressure due to impaired aqueous humor (AH) drainage through the trabecular meshwork (TM). Although increased TM tissue contraction and stiffness in association with accumulation of extracellular matrix (ECM) are believed to be partly responsible for increased resistance to AH outflow, the extracellular cues and intracellular mechanisms regulating TM cell contraction and ECM production are not well defined. This study tested the hypothesis that sustained activation of Rho GTPase signaling induced by lysophosphatidic acid (LPA), TGF-ß, and connective tissue growth factor (CTGF) influences TM cell plasticity and fibrogenic activity which may eventually impact resistance to AH outflow. Various experiments performed using human TM cells revealed that constitutively active RhoA (RhoAV14), TGF-ß2, LPA, and CTGF significantly increase the levels and expression of Fibroblast Specific Protein-1 (FSP-1), α-smooth muscle actin (αSMA), collagen-1A1 and secretory total collagen, as determined by q-RT-PCR, immunofluorescence, immunoblot, flow cytometry and the Sircol assay. Significantly, these changes appear to be mediated by Serum Response Factor (SRF), myocardin-related transcription factor (MRTF-A), Slug, and Twist-1, which are transcriptional regulators known to control cell plasticity, myofibroblast generation/activation and fibrogenic activity. Additionally, the Rho kinase inhibitor-Y27632 and anti-fibrotic agent-pirfenidone were both found to suppress the TGF-ß2-induced expression of αSMA, FSP-1, and collagen-1A1. Taken together, these observations demonstrate the significance of RhoA/Rho kinase signaling in regulation of TM cell plasticity, fibrogenic activity, and myofibroblast activation, events with potential implications for the pathobiology of elevated intraocular pressure in glaucoma patients.


Asunto(s)
GTP Fosfohidrolasas/metabolismo , Glaucoma/metabolismo , Malla Trabecular/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Proteínas de Unión al Calcio/metabolismo , Línea Celular , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Citometría de Flujo , GTP Fosfohidrolasas/genética , Regulación del Desarrollo de la Expresión Génica , Glaucoma/genética , Glaucoma/patología , Humanos , Presión Intraocular/genética , Lisofosfolípidos/administración & dosificación , Proteína de Unión al Calcio S100A4 , Transducción de Señal/efectos de los fármacos , Malla Trabecular/citología , Factor de Crecimiento Transformador beta2/metabolismo , Proteína de Unión al GTP rhoA/genética
19.
BMC Cancer ; 14: 432, 2014 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-24928086

RESUMEN

BACKGROUND: Oral squamous cell carcinoma is an aggressive neoplasm with serious morbidity and mortality, which typically spreads through local invasive growth. Lysophosphatidic acid (LPA) is involved in a number of biological processes, and may have a role in cancer cell migration and invasiveness. LPA is present in most tissues and can activate cells through six different LPA receptors (LPAR1-6). Although LPA is predominantly promigratory, some of the receptors may have antimigratory effects in certain cells. The signalling mechanisms of LPA are not fully understood, and in oral carcinoma cells the specific receptors and pathways involved in LPA-stimulated migration are unknown. METHODS: The oral carcinoma cell lines E10, SCC-9, and D2 were investigated. Cell migration was studied in a scratch wound assay, and invasion was demonstrated in organotypic three dimensional co-cultures. Protein and mRNA expression of LPA receptors was studied with Western blotting and qRT-PCR. Activation of signalling proteins was examined with Western blotting and isoelectric focusing, and signalling mechanisms were further explored using pharmacological agents and siRNA directed at specific receptors and pathways. RESULTS: LPA stimulated cell migration in the two oral carcinoma cell lines E10 and SCC-9, but was slightly inhibitory in D2. The receptor expression profile and the effects of specific pharmacological antagonist and agonists indicated that LPA-stimulated cell migration was mediated through LPAR3 in E10 and SCC-9. Furthermore, in both these cell lines, the stimulation by LPA was dependent on PKC activity. However, while LPA induced transactivation of EGFR and the stimulated migration was blocked by EGFR inhibitors in E10 cells, LPA did not induce EGFR transactivation in SCC-9 cells. In D2 cells, LPA induced EGFR transactivation, but this was associated with slowing of a very high inherent migration rate in these cells. CONCLUSION: The results demonstrate LPA-stimulated migration in oral carcinoma cells through LPAR3, mediated further by PKC, which acts either in concert with or independently of EGFR transactivation.


Asunto(s)
Carcinoma de Células Escamosas/genética , Receptores ErbB/genética , Neoplasias de la Boca/genética , Receptores del Ácido Lisofosfatídico/biosíntesis , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Receptores ErbB/biosíntesis , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Lisofosfolípidos/administración & dosificación , Neoplasias de la Boca/patología , Proteína Quinasa C/genética , Receptores del Ácido Lisofosfatídico/genética , Transducción de Señal/efectos de los fármacos
20.
J Biomed Sci ; 21: 55, 2014 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-24898615

RESUMEN

BACKGROUND: Adipose tissue provides a readily available source of autologous stem cells. Adipose-derived stem cells (ASCs) have been proposed as a source for endothelial cell substitutes for lining the luminal surface of tissue engineered bypass grafts. Endothelial nitric oxide synthase (eNOS) is a key protein in endothelial cell function. Currently, endothelial differentiation from ASCs is limited by poor eNOS expression. The goal of this study was to investigate the role of three molecules, sphingosine-1-phosphate (S1P), bradykinin, and prostaglandin-E1 (PGE1) in ASC endothelial differentiation. Endothelial differentiation markers (CD31, vWF and eNOS) were used to evaluate the level of ASCs differentiation capability. RESULTS: ASCs demonstrated differentiation capability toward to adipose, osteocyte and endothelial like cell phenotypes. Bradykinin, S1P and PGE were used to promote differentiation of ASCs to an endothelial phenotype. Real-time PCR showed that all three molecules induced significantly greater expression of endothelial differentiation markers CD31, vWF and eNOS than untreated cells. Among the three molecules, S1P showed the highest up-regulation on endothelial differentiation markers. Immunostaining confirmed presence of more eNOS in cells treated with S1P than the other groups. Cell growth measurements by MTT assay, cell counting and EdU DNA incorporation suggest that S1P promotes cell growth during ASCs endothelial differentiation. The S1P1 receptor was expressed in ASC-differentiated endothelial cells and S1P induced up-regulation of PI3K. CONCLUSIONS: S1P up-regulates endothelial cell markers including eNOS in ASCs differentiated to endothelial like cells. This up-regulation appears to be mediated by the up-regulation of PI3K via S1P1 receptor. ASCs treated with S1P offer promising use as endothelial cell substitutes for tissue engineered vascular grafts and vascular networks.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Células Endoteliales/citología , Lisofosfolípidos/administración & dosificación , Óxido Nítrico Sintasa/metabolismo , Esfingosina/análogos & derivados , Tejido Adiposo , Alprostadil/administración & dosificación , Bradiquinina/administración & dosificación , Células Endoteliales/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Receptores de Lisoesfingolípidos/biosíntesis , Esfingosina/administración & dosificación , Receptores de Esfingosina-1-Fosfato , Células Madre/citología , Células Madre/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA