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.
Physiol Rep ; 4(12)2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27354543

RESUMEN

Platelet-activating factor (PAF) acting via its receptor (PAFR) is implicated in the pathogenesis of persistent pulmonary hypertension of the newborn (PPHN). Effects of long-term oxygen therapy on newborn lung are not well understood; therefore, we studied the effect of oxygen tension on ovine newborn pulmonary artery smooth muscle cells (NBPASMC). Our global hypothesis is that PPHN results from failure of newborn lamb pulmonary system to downregulate PAFR activity or to upregulate vasodilatory cyclic nucleotides (Cnucs) activity. NBPASMC from newborns 6-12 days old were studied in vitro at three different oxygen tensions (pO2, [Torr]: hypoxia, <40; normoxia, 80-100; and hyperoxia, >100 Torr often clinically imposed upon newborns with PPHN) PAFR- and Cnucs mediated effects were determined. PAFR and PKA Cα mRNA expression as well as prostacyclin, thromboxane, cAMP production, and DNA synthesis was studied to assess PAFR-mediated hypertrophy and/or hyperplasia. Hypoxia and hyperoxia increased specific PAFR binding. PAF treatment during hyperoxia increased PAFR gene, but decreased PKA-Cα gene expression. Hypoxia and hyperoxia increased NBPASMC proliferation via PAFR signaling. Baseline prostacyclin level was ninefold greater than in fetal PASMC, whereas baseline thromboxane was sevenfold less suggesting greater postnatal cyclooxygenase activity in NBPASMC PAF decreased, while forskolin and 8-Br-cAMP increased cAMP production. Decrease of PAFR effects by Cnucs indicates that normal newborn PA physiology favors vasodilator pathways to minimize PAF-induced hypertrophy or hyperplasia. We speculate that failure of newborn lung to anchor downregulation of vasoconstrictors with upregulation of vasodilators leads to PPHN.


Asunto(s)
Hiperoxia/metabolismo , Hipoxia/metabolismo , Miocitos del Músculo Liso/metabolismo , Terapia por Inhalación de Oxígeno/efectos adversos , Síndrome de Circulación Fetal Persistente/metabolismo , Glicoproteínas de Membrana Plaquetaria/metabolismo , Arteria Pulmonar/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , AMP Cíclico/metabolismo , Síndrome de Circulación Fetal Persistente/fisiopatología , Síndrome de Circulación Fetal Persistente/terapia , Unión Proteica , Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo , Arteria Pulmonar/citología , Ovinos , Transducción de Señal , Vasoconstricción
2.
Mol Genet Metab Rep ; 4: 11-8, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26966681

RESUMEN

Platelet activating factor (PAF) modulates ovine fetal pulmonary hemodynamic. PAF acts through its receptors (PAFR) in pulmonary vascular smooth muscle cells (PVSMC) to phosphorylate and induce nuclear translocation of NF-kB p65 leading to PVSMC proliferation. However, the interaction of NF-kB p65 and PAF in the nuclear domain to effect PVSMC cell growth is not clearly defined. We used siRNA-dependent translation initiation arrest to study a mechanism by which NF-kB p65 regulates PAF stimulation of PVSMC proliferation. Our hypotheses are: (a) PAF induces NF-kB p65 DNA binding and (b) NF-kB p65 siRNA attenuates PAF stimulation of PVSMC proliferation. For DNA binding, cells were fed 10 nM PAF with and without PAFR antagonists WEB 2170, CV 3988 or BN 52021 and incubated for 12 h. DNA binding was measured by specific ELISA. For NF-kB p65 siRNA effect, starved cells transfected with the siRNA were incubated for 24 h with and without 10 nM PAF. Cell proliferation was measured by DNA synthesis while expression of NF-kB p65 and PAFR protein was measured by Western blotting. In both studies, the effect of 10% FBS alone was used as the positive control. In general, PAF stimulated DNA binding which was inhibited by PAFR antagonists. siRNAs to NF-kB p65 and PAFR significantly attenuated cell proliferation compared to 10% FBS and PAF effect. Inclusion of PAF in siRNA-treated cells did not reverse inhibitory effect of NF-kB p65 siRNA on DNA synthesis. PAFR expression was inhibited in siRNA-treated cells. These data show that PAF-stimulation of PVSMC proliferation occurs via a PAFR-NF-kB p65 linked pathway.

3.
Pulm Circ ; 4(3): 482-95, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25621162

RESUMEN

Initiation, progression, and resolution of vaso-occlusive pain episodes in sickle cell disease (SCD) have been recognized as reperfusion injury, which provokes an inflammatory response in the pulmonary circulation. Some 5-lipoxygenase (5-lox) metabolites are potent vasoconstrictors in the pulmonary circulation. We studied stimulation of production of the inflammatory eicosanoids leukotrienes (LTs) and prostaglandin E2 (PGE2) by isolated rat lungs perfused with sickle (HbSS) erythrocytes. Our hypothesis is that HbSS erythrocytes produce more LTs than normal (HbAA) erythrocytes, which can induce vaso-occlusive episodes in SCD patients. Lung perfusates were collected at specific time points and purified by high-pressure liquid chromatography, and LTC4 and PGE2 contents were measured by enzyme-linked immunosorbent assay (ELISA). Rat lung explants were also cultured with purified HbAA and HbSS peptides, and 5-lox, cyclooxygenase 1/2, and platelet-activating factor receptor (PAFR) proteins were measured by Western blotting, while prostacyclin and LTs produced by cultured lung explants were measured by ELISA. Lung weight gain and blood gas data were not different among the groups. HbSS-perfused lungs produced more LTC4 and PGE2 than HbAA-perfused lungs: 10.40 ± 0.62 versus 0.92 ± 0.2 ng/g dry lung weight (mean ± SEM; P = 0.0001) for LTC4. Inclusion of autologous platelets (platelet-rich plasma) elevated LTC4 production to 12.6 ± 0.96 and 7 ± 0.60 ng/g dry lung weight in HbSS and HbAA perfusates, respectively. HbSS lungs also expressed more 5-lox and PAFR. The data suggest that HbSS erythrocytes and activated platelets in patient's pulmonary microcirculation will enhance the synthesis and release of the proinflammatory mediators LTC4 and PGE2, both of which may contribute to onset of the acute chest syndrome in SCD.

4.
Int Arch Allergy Immunol ; 158(1): 18-26, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22212397

RESUMEN

BACKGROUND: Racemic albuterol is a 50:50 mixture of the (R)- and (S)-enantiomers of albuterol. Its clinical efficacy resides in the (R)-enantiomer (levalbuterol). Studies have shown that (S)-albuterol induces human bronchial smooth muscle cell (HBSMC) proliferation via a pathway linked to platelet-activating factor (PAF), but the underlying mechanism by which (S)-albuterol augments PAF effects is not clear. In this study, we compared effect of levalbuterol and (S)-albuterol on PAF receptor (PAFr)-mediated signaling and PAF metabolism by HBSMCs after incubation with the albuterol isomers. METHODS: PAF binding and inositol phosphate (IP(3)) release were studied on adherent cultured cells. PAFr protein expression was measured by Western blotting, PAF synthesis and catabolism were measured in membrane and cytosolic proteins of cells incubated with albuterol isomers. RESULTS: Compared to control conditions, (S)-albuterol increased PAF binding by 70% after 30 min of preincubation and by 150% after 24 h of preincubation. Levalbuterol had no effect on PAF binding under both conditions. (S)-albuterol also augmented PAF stimulation of IP(3) release, while levalbuterol and the racemic mixture had no effect. WEB 2170, a PAFr antagonist, inhibited the ability of (S)-albuterol to increase PAF binding or stimulate IP(3) release. (S)-albuterol stimulated PAFr protein expression. With PAF metabolism, (S)-albuterol treatment augmented PAF synthesis, but significantly inhibited PAF catabolism. CONCLUSIONS: Our data suggest that one mechanism by which (S)-albuterol stimulates HBSMC proliferation involves upregulation of PAFr-mediated effects including increased PAF synthesis and decreased PAF catabolism.


Asunto(s)
Albuterol/farmacología , Bronquios/efectos de los fármacos , Broncodilatadores/farmacología , Miocitos del Músculo Liso/efectos de los fármacos , Factor de Activación Plaquetaria/biosíntesis , Glicoproteínas de Membrana Plaquetaria/fisiología , Receptores Acoplados a Proteínas G/fisiología , Azepinas/farmacología , Bronquios/metabolismo , Línea Celular , Proliferación Celular/efectos de los fármacos , Humanos , Inositol 1,4,5-Trifosfato/metabolismo , Miocitos del Músculo Liso/metabolismo , Glicoproteínas de Membrana Plaquetaria/antagonistas & inhibidores , Glicoproteínas de Membrana Plaquetaria/biosíntesis , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/biosíntesis , Transducción de Señal/efectos de los fármacos , Triazoles/farmacología
5.
Mol Genet Metab ; 101(4): 400-8, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20813571

RESUMEN

Hypoxia augments PAF receptor (PAFr) binding and PAFr protein expression in venous SMC (SMC-PV). We compared effect of acute and prolonged hypoxia (pO(2)<40 torr) on PAFr-mediated responses in arterial SMC (SMC-PA) and SMC-PV. Cells were studied for 30 min (acute) or for 48 h (prolonged) hypoxia and compared to normoxic (pO(2) ~100 torr) conditions. PAF binding was quantified in fmol/10(6) cells (mean ± SEM). PAF binding in normoxia were SMC-PA, 5.2 ± 0.2 and in SMC-PV, 19.3 ± 1.1; values in acute hypoxia were SMC-PA, 7.7 ± 0.4 and in SMC-PV, 27.8 ± 1.7. Prolonged hypoxia produced 6-fold increase in binding in SMC-PA, but only 2-fold increase in SMC-PV, but binding in SMC-PV was still higher. Acute hypoxia augmented inositol phosphate release by 50% and 40% in SMC-PA and SMC-PV, respectively. During normoxia, PAFr mRNA expression by both cell types was similar, but expression in hypoxia by SMC-PA was greater. In SMC-PA, hypoxia and PAF augmented intracellular calcium flux. Re-exposure of cells to 30 min normoxia after 48 h hypoxia decreased binding by 45-60%, suggesting immediate down-regulation of hypoxia-induced PAFr-mediated effects. We speculate that re-oxygenation immediately reverses hypoxia effect probably due to oxygen tension-dependent reversibility of PAFr activation and suggest that exposure of the neonate to prolonged state of hypoxia will vilify oxygen exchange capacity of the neonatal lungs.


Asunto(s)
Hipoxia/metabolismo , Músculo Liso Vascular/metabolismo , Glicoproteínas de Membrana Plaquetaria/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Calcio/metabolismo , Células Cultivadas , Regulación hacia Abajo , Femenino , Feto , Hipoxia/genética , Fosfatos de Inositol/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Oxígeno/metabolismo , Oxígeno/farmacología , Factor de Activación Plaquetaria/farmacología , Glicoproteínas de Membrana Plaquetaria/biosíntesis , Glicoproteínas de Membrana Plaquetaria/genética , Embarazo , Arteria Pulmonar/citología , Arteria Pulmonar/metabolismo , Venas Pulmonares/citología , Venas Pulmonares/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores Acoplados a Proteínas G/biosíntesis , Receptores Acoplados a Proteínas G/genética , Ovinos
6.
High Alt Med Biol ; 10(4): 365-72, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-20043379

RESUMEN

High altitude long-term hypoxia (LTH) in the fetus may result in pulmonary vascular smooth muscle cell (PVSMC) proliferation and pulmonary vascular remodeling. Our objective was to determine if epidermal growth factor receptor (EGFR) is involved in hypoxia induced PVSMC proliferation or in pulmonary vascular remodeling in ovine fetuses exposed to high altitude LTH. Fetuses of pregnant ewes that were held at 3820-m altitude from *30 to 140 days (LTH) gestation and sea level control pregnant ewes were delivered near term. Morphometric analyses and immunohistochemistry were done on fetal lung sections. Pulmonary arteries of LTH fetuses exhibited medial wall thickening and distal muscularization. Western blot analyses done on protein isolated from pulmonary arteries demonstrated an upregulation of EGFR. This upregulation was attributed in part to PVSMC in the medial wall by immunohistochemistry.Proliferation of fetal ovine PVSMC after 24 h of hypoxia (2% O2) was attenuated by inhibition of EGFR with 250 nmol tyrphostin 4-(3-chloroanilino)-6,7-dimethoxyquinazoline (AG1478), a specific EGFR protein tyrosine kinase inhibitor, when measured by [3H]-thymidine incorporation. Our data indicate that EGFR plays a role in fetal ovine pulmonary vascular remodeling following long-term fetal hypoxia and that inhibition of EGFR signaling may ameliorate hypoxia-induced pulmonary vascular remodeling.


Asunto(s)
Altitud , Receptores ErbB/metabolismo , Hipoxia Fetal/fisiopatología , Músculo Liso Vascular/fisiopatología , Arteria Pulmonar/fisiopatología , Actinas/metabolismo , Animales , Proliferación Celular , Células Cultivadas , Receptores ErbB/antagonistas & inhibidores , Femenino , Hipoxia Fetal/etiología , Hipoxia Fetal/metabolismo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Embarazo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Arteria Pulmonar/metabolismo , Arteria Pulmonar/patología , Quinazolinas , Oveja Doméstica , Tirfostinos/farmacología
7.
Int Arch Allergy Immunol ; 146(4): 321-33, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18362475

RESUMEN

BACKGROUND: Racemic albuterol is a 50:50 mixture of the R-isomer, levalbuterol, and the S-isomer, S-albuterol. S-Albuterol increases airway hyperresponsiveness to spasmogens, exacerbates asthmatic conditions and stimulates cell growth, whereas levalbuterol attenuates cell growth in culture. The mechanisms of S-albuterol-induced cell proliferation are not well understood. We studied the role of albuterol isomers and intracellular cell cycle regulators on proliferation of human bronchial smooth muscle cells. METHODS: Serum-starved cells (72 h) were fed test agents for 24 h and cell proliferation was measured. The expression of nuclear factor-kappaB inhibitory protein IkappaBalpha, nuclear factor-kappaB, cyclin-dependent kinases 2 and 4, interleukin (IL)-6, and retinoblastoma and platelet-activating factor (PAF) receptor protein were measured by Western blotting. RESULTS: S-Albuterol, PAF and platelet-derived growth factor stimulated cell proliferation, but levalbuterol and the racemic mixture inhibited cell proliferation compared with the effect of 5% fetal bovine serum alone. The proliferative effect of platelet-derived growth factor on S-albuterol was not additive, suggesting that the 2 mediators act by different mechanisms. S-Albuterol induced greater expression of all the measured proteins than either levalbuterol, the racemic mixture or 5% fetal bovine serum. S-Albuterol stimulated IL-6 secretion and abolished the ability of levalbuterol to inhibit IL-6 secretion. CONCLUSION: Our data show that S-albuterol stimulates cell proliferation by activating expression and phosphorylation of several intracellular mitogenic proteins and may exacerbate asthma by stimulating the release of IL-6. Induction of PAF receptor protein expression by S-albuterol strongly suggests that S-albuterol may exert its adverse effects by binding to a G protein-coupled receptor such as the PAF receptor.


Asunto(s)
Albuterol/farmacología , Asma/tratamiento farmacológico , Broncodilatadores/farmacología , Miocitos del Músculo Liso/efectos de los fármacos , Albuterol/efectos adversos , Asma/metabolismo , Asma/patología , Western Blotting , Broncodilatadores/efectos adversos , Proliferación Celular/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina/biosíntesis , Quinasa 4 Dependiente de la Ciclina/biosíntesis , Sinergismo Farmacológico , Humanos , Quinasa I-kappa B/biosíntesis , Interleucina-6/biosíntesis , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , FN-kappa B/biosíntesis , Fosforilación/efectos de los fármacos , Factor de Activación Plaquetaria/farmacología , Glicoproteínas de Membrana Plaquetaria/biosíntesis , Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Factor de Crecimiento Derivado de Plaquetas/farmacología , Receptores Acoplados a Proteínas G/biosíntesis , Proteína de Retinoblastoma/biosíntesis
8.
Clin Exp Pharmacol Physiol ; 35(2): 148-58, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17892503

RESUMEN

1. Endothelial nitric oxide synthase (NOS3) is important for vascular homeostasis. The role of protein kinase G (PKG) in regulation of NOS3 activity was studied in primary cultures of newborn lamb lung microvascular endothelial cells (LMVEC). 2. We determined the presence of PKG in fetal and neonatal LMVEC as well as subcellular localization of PKG isoforms in the neonatal cells by fluorescence immunohistochemistry. We used diaminofluorescein (DAF) fluorophore to measure nitric oxide (NO) production from neonatal LMVEC. We confirmed that NO measured was from constitutive NOS3 by inhibiting it with NOS inhibitors. 3. To identify a role for PKG in basal NO production, we measured NO release from LMVEC cells using 4-amino-5-methylamino-2',7'-difluorofluorescein (DAF-FM; 0.5-0.8 micromol/L) with and without prior stimulation with the PKG activator 8-bromo-cGMP (8-Br-cGMP; 0.3 and 3 micromol/L) or prior PKG inhibition with beta-phenyl-1,N2-etheno-8-bromoguanosine-3',5'-cyclic monophosphorothionate (BPC; 0.3 and 3 micromol/L). With the same drugs, we determined the role of PKG on cellular expression of NOS3 and serine 116 phosphorylated NOS (pSer116-NOS) by qualitative and quantitative immunofluorescence assays, as well as western blotting. 4. Because PKG 1 beta was distributed throughout the cytosol in a punctate expression, we used 2 mmol/L cyclodextrin, a cholesterol extractor, to determine a role for lipid vesicles in PKG regulation of NO production. 5. Protein kinase G 1 beta gave a punctate appearance, indicating its presence in intracellular vesicles. Nitric oxide production decreased by approximately 20% with 300 nmol/L and 3 micromol/L 8-Br cGMP (P < 0.05) and increased by 20.8 +/- 3.7% with 3 micromol/L BPC (P < 0.001), indicating that both stimulated and basal PKG activity has inhibitory effects on basal NOS3 function. Nitric oxide synthase immunofluorescence and immunoblot expression were decreased and pSer116-NOS immunofluorescence was increased by 800 nmol/L 8-Br-cGMP and 170 micromol/L (Z)-1-[2-(2-aminoethyl)-N-(2-ammonio-ethyl)amino]diazen-1-ium-1, 2-diolate (DETANONOate). The effect of cyclodextrin indicated that cholesterol extraction interfered with PKG inhibition of NOS. Further examination of pSer116-NOS by immunohistochemistry showed it abundant in the endoplasmic reticulum and colocalized with PKG 1 beta, especially in nuclear vesicles. 6. We conclude that endothelial PKG is involved in endogenous regulation of basal NOS3 activity with the involvement of lipid structures, the endoplasmic reticulum and the nucleus. Protein kinase G 1 beta is colocalized with pSer116-NOS, indicating that PKG action may involve serine 116 phosphorylation on NOS.


Asunto(s)
Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Células Endoteliales/enzimología , Pulmón/irrigación sanguínea , Lípidos de la Membrana/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Óxido Nítrico/metabolismo , Orgánulos/enzimología , Animales , Animales Recién Nacidos , Western Blotting , Núcleo Celular/metabolismo , Células Cultivadas , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacología , Ciclodextrinas/farmacología , Citosol/enzimología , Relación Dosis-Respuesta a Droga , Retículo Endoplásmico/enzimología , Células Endoteliales/efectos de los fármacos , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Fluoresceínas , Colorantes Fluorescentes , Pulmón/embriología , Microcirculación/enzimología , Microscopía Fluorescente/métodos , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Compuestos Nitrosos/farmacología , Orgánulos/efectos de los fármacos , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Subunidades de Proteína/metabolismo , Transporte de Proteínas , Serina/metabolismo , Ovinos , Factores de Tiempo
9.
Am J Physiol Lung Cell Mol Physiol ; 293(6): L1475-82, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17951313

RESUMEN

Platelet-activating factor (PAF) is implicated in pathogenesis of chronic hypoxia-induced pulmonary hypertension in some animal models and in neonates. Effects of chronic hypoxia on PAF receptor (PAF-R) system in fetal pulmonary vasculature are unknown. We investigated the effect of chronic high altitude hypoxia (HAH) in fetal lambs [pregnant ewes were kept at 3,801 m (12,470 ft) altitude from approximately 35 to 145 days gestation] on PAF-R-mediated effects in the pulmonary vasculature. Age-matched controls were kept at sea level. Intrapulmonary arteries were isolated, and smooth muscle cells (SMC-PA) were cultured from HAH and control fetuses. To determine presence of pulmonary vascular remodeling, lung tissue sections were subjected to morphometric analysis. Percentage medial wall thickness was significantly increased (P < 0.05) in arteries at all levels in the HAH lambs. PAF-R protein expression studied by immunocytochemistry and Western blot analysis on lung tissue SMC-PA demonstrated greater PAF-R expression in HAH lambs. PAF-R binding (femtomoles per 10(6) cells) in HAH SMC-PA was 90.3 +/- 4.08 and 66% greater than 54.3 +/- 4.9 in control SMC-PA. Pulmonary arteries from HAH fetuses synthesized >3-fold PAF than vessels from controls. Compared with controls SMC-PA of HAH lambs demonstrated 139% and 40% greater proliferation in 10% FBS alone and with 10 nM PAF, respectively. Our data demonstrate that exposure of ovine fetuses to HAH will result in significant upregulation of PAF synthesis, PAF-R expression, and PAF-R-mediated effects in pulmonary arteries. These findings suggest that increased PAF-R protein expression and increased PAF binding contribute to pulmonary vascular remodeling in these animals and may predispose them to persistent pulmonary hypertension after birth.


Asunto(s)
Mal de Altura/fisiopatología , Feto/fisiopatología , Hipoxia/fisiopatología , Factor de Activación Plaquetaria/metabolismo , Arteria Pulmonar/fisiopatología , Animales , Peso al Nacer/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Enfermedad Crónica , Femenino , Feto/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/patología , Factor de Activación Plaquetaria/biosíntesis , Factor de Activación Plaquetaria/farmacología , Glicoproteínas de Membrana Plaquetaria/metabolismo , Embarazo , Arteria Pulmonar/patología , Receptores Acoplados a Proteínas G/metabolismo , Oveja Doméstica
10.
Am J Physiol Heart Circ Physiol ; 292(6): H2773-81, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17322418

RESUMEN

We have previously reported that platelet-activating factor (PAF) is present in very high levels in the ovine fetal lung and circulation and that PAF serves as an important physiological vasoconstrictor of the pulmonary circulation in utero. However, it is not known whether PAF stimulates pulmonary vascular smooth muscle cell (SMC) proliferation. In this study, we used ovine fetal pulmonary venous SMCs as our model system to study the effects and mechanisms of action of PAF on SMC proliferation. We found that PAF induced SMC proliferation in a dose-dependent manner. PAF also stimulated activation of both ERK and p38 but not c-Jun NH(2) terminal kinase (JNK) mitogen-activated protein (MAP) kinase pathways. PAF (10 nM) induced phosphorylation of epidermal growth factor receptor (EGFR). Specific inhibition of EGFR by AG-1478 and by the expression of a dominant-negative EGFR mutant in SMCs attenuated PAF-stimulated cell proliferation. Inhibition of heparin-binding EGF-like growth factor (HB-EGF) release by CRM-197 and inhibition of matrix metalloproteinases (MMP) by GM-6001 abolished PAF-induced MAP kinase activation and cell proliferation. Increased alkaline phosphatase (AP) activity after PAF treatment in AP-HB-EGF fusion construct-transfected SMCs indicated that PAF induced the release of HB-EGF within 1 min. Gelatin zymography data showed that PAF stimulated MMP-2 activity and MMP-9 activity within 1 min. These results suggest that PAF promotes pulmonary vascular SMC proliferation via transactivation of EGFR through MMP activation and HB-EGF, resulting in p38 and ERK activation and that EGFR transactivation is essential for the mitogenic effect of PAF in pulmonary venous SMC.


Asunto(s)
Proliferación Celular , Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Factor de Activación Plaquetaria/metabolismo , Transducción de Señal , Animales , Proteínas Bacterianas/farmacología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Dipéptidos/farmacología , Relación Dosis-Respuesta a Droga , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Factor de Crecimiento Epidérmico/genética , Receptores ErbB/efectos de los fármacos , Receptores ErbB/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Factor de Crecimiento Similar a EGF de Unión a Heparina , Péptidos y Proteínas de Señalización Intercelular , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/embriología , Mutación , Miocitos del Músculo Liso/efectos de los fármacos , Fosforilación , Factor de Activación Plaquetaria/farmacología , Venas Pulmonares/citología , Venas Pulmonares/embriología , Venas Pulmonares/metabolismo , Quinazolinas , Ovinos , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Transfección , Tirfostinos/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
11.
J Pharmacol Exp Ther ; 320(2): 728-37, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17085546

RESUMEN

At birth, release of endogenous vasodilators such as nitric oxide and prostacyclin facilitate pulmonary vasodilation via the cyclic nucleotides, cGMP and cAMP. Interaction of cyclic nucleotides and platelet-activating factor (PAF)-mediated responses in pulmonary vascular smooth muscle is not known. We studied the effects of cGMP and cAMP on PAF-mediated responses in ovine fetal intrapulmonary venous smooth muscle cells. Studies were done in hypoxia or normoxia with buffer with 8-Br-cGMP (BGMP) and 8-Br-cAMP (BAMP), as well as cGMP-dependent protein kinase (PKG) and cAMP-dependent protein kinase (PKA) inhibitors. All groups were treated with 1 nM PAF and incubated for 30 min for the binding assay or 20 min for measurement of inositol 1,4,5-phosphate (IP(3)) production. BGMP and BAMP decreased PAF binding in normoxia by 63 and 14%, respectively. Incubations with the PKG inhibitor Rp-8-(4-chlorophenylthio)-guanosine-3',5'-cyclic monophosphorothioate sodium and the PKA inhibitor Rp-adenosine-3',5'-cyclic monophosphorothioate abrogated the inhibitory effects of BGMP and BAMP. PAF-stimulated IP(3) production was 8565 +/- 314 dpm/10(6) cells in hypoxia and 5418 +/- 118 dpm/10(6) cells in normoxia, a 40% decrease. BGMP attenuated PAF-stimulated IP(3) production by 67 and 37% in hypoxia and normoxia, respectively; the value for BAMP was 44% under both conditions. Pretreatment with PKG or PKA inhibitor abrogated BGMP and BAMP inhibition of IP(3) release. PAF receptor (PAFr) protein expression decreased in normoxia, but pretreatment with 10 nM PAF up-regulated PAFr expression. Pretreatment with PAF decreased expression and activities of PKG or PKA proteins in normoxia and hypoxia. Our data demonstrate the existence of cGMP/cAMP-PAF cross-talk in pulmonary vascular smooth muscle cells, which may be one mechanism by which PAFr-mediated vasoconstriction is down-regulated at birth.


Asunto(s)
8-Bromo Monofosfato de Adenosina Cíclica/farmacología , GMP Cíclico/análogos & derivados , Pulmón/irrigación sanguínea , Músculo Liso Vascular/efectos de los fármacos , Factor de Activación Plaquetaria/farmacología , Animales , AMP Cíclico/fisiología , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , GMP Cíclico/farmacología , GMP Cíclico/fisiología , Proteínas Quinasas Dependientes de GMP Cíclico/fisiología , Femenino , Feto , Inositol 1,4,5-Trifosfato/metabolismo , Músculo Liso Vascular/fisiología , Glicoproteínas de Membrana Plaquetaria/fisiología , Embarazo , Receptores Acoplados a Proteínas G/fisiología , Ovinos
12.
Am J Physiol Lung Cell Mol Physiol ; 291(5): L1079-93, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16997880

RESUMEN

We determined the effect of oxygen [approximately 100 Torr (normoxia) and approximately 30-40 Torr (hypoxia)] on functions of endothelial nitric oxide (NO) synthase (NOS-3) and its negative regulator caveolin-1 in ovine fetal and neonatal lung microvascular endothelial cells (MVECs). Fetal NOS-3 activity, measured as NO production with 0.5-0.9 microM 4-amino-5-methylamino-2,7-difluorofluorescein, was decreased in hypoxia by 14.4% (P < 0.01), inhibitable by the NOS inhibitor N-nitro-L-arginine, and dependent on extracellular arginine. Caveolar function, assessed as FITC-BSA (160 microg/ml) endocytosis, was decreased in hypoxia by 13.5% in fetal and 22.8% in neonatal MVECs (P < 0.01). NOS-3 and caveolin-1 were physically associated, as demonstrated by coimmunoprecipitation and colocalization, and functionally associated, as shown by cross-activation of endocytosis, by their specific antibodies and activation of NOS by albumin. Caveolin peptide, containing the sequence for the PKC phosphorylation site of caveolin, and caveolin antiserum against the site increased NO production and endocytosis by 12.3% (P < 0.05) and 16% (P < 0.05), respectively, in normoxia and increased endocytosis by 25% (P < 0.001) in hypoxia. PMA decreased NO production in normoxia and hypoxia by 19.32% (P < 0.001) and 11.8% (P < 0.001) and decreased endocytosis in normoxia by 20.35% (P < 0.001). PKC kinase activity was oxygen sensitive, and threonine phosphorylation was enhanced in hypoxia. Pertussis toxin increased caveolar and NOS functions. These data support our hypothesis that increased Po(2) at birth promotes dissociation of caveolin-1 and NOS-3, with an increase in their activities, and that PKC and an oxygen-sensitive cell surface G protein-coupled receptor regulate caveolin-1 and NOS-3 interactions in fetal and neonatal lung MVECs.


Asunto(s)
Caveolina 1/metabolismo , Células Endoteliales/enzimología , Hipoxia/metabolismo , Pulmón/irrigación sanguínea , Óxido Nítrico Sintasa de Tipo III/metabolismo , Oxígeno/farmacología , Animales , Especificidad de Anticuerpos , Caveolina 1/inmunología , Células Cultivadas , Regulación hacia Abajo/fisiología , Endocitosis/fisiología , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Hipoxia/tratamiento farmacológico , Pulmón/embriología , Pulmón/crecimiento & desarrollo , Microcirculación/efectos de los fármacos , Microcirculación/enzimología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/inmunología , Toxina del Pertussis/farmacología , Fosforilación , Embarazo , Proteína Quinasa C/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Serina/metabolismo , Ovinos , Treonina/metabolismo
13.
Life Sci ; 79(23): 2157-64, 2006 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-16890244

RESUMEN

The purpose of this study was to examine the levels of norepinephrine (NE) turnover in skin tissues and to determine the effect of nitric oxide (NO) on NE production in acupuncture points (acupoints) and meridians. The rats were pretreated with alpha-methyl-tyrosine methyl ester and intravenously infused with L-(2,3,5,6-(3)H)-tyrosine. Blood was withdrawn and skin tissues were excised from the low skin resistance points, non-acupoint, and non-meridian areas located on leg, arm, or trunk. The results showed that the skin NE concentration and (3)H-NE release in acupoints were significantly higher than those in non-acupoints and non-meridian controls. (3)H-NE releases in the acupoints were increased by intravenous infusion of 2-N,N-diethylamino-diazenolate-2-oxide, an NO donor, but lowered by N(G)-Propyl-L-arginine, an inhibitor of neuronal NO synthesis. NE turnover rates in the acupoints were lower in the NO donor treated group while the inhibitor of NO synthesis reversed the trend. In contrast, NE turnover rates were not altered by NO donor and inhibitor of NO synthesis in non-acupoint and non-meridian control tissues. This is the first evidence that NE turnover was consistently decreased in acupoints and enhanced NE synthesis/release in acupoints were facilitated by presence of an NO donor and inhibited by an inhibitor of NO synthesis. The data suggest that skin NE synthesis/release in acupoints/meridians is increased in skin acupoints, which is modulated by L-arginine-derived NO synthesis in sympathetic nervous system.


Asunto(s)
Puntos de Acupuntura , Óxido Nítrico/metabolismo , Norepinefrina/biosíntesis , Piel/metabolismo , Animales , Arginina/análogos & derivados , Arginina/farmacología , Dietilaminas/farmacología , Masculino , Donantes de Óxido Nítrico/farmacología , Ratas , Ratas Sprague-Dawley , Piel/inervación , Sistema Nervioso Simpático/metabolismo
14.
Int Arch Allergy Immunol ; 139(3): 225-36, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16446544

RESUMEN

BACKGROUND: Racemic albuterol is a mixture of (R)- and (S)-enantiomers of albuterol. Its pharmacological activity and clinical efficacy reside in the (R)-enantiomer (levalbuterol), but the (S)-enantiomer exacerbates airway reactivity in nonclinical models. The role of albuterols in airway smooth muscle cell (SMC) proliferation is not well understood. METHODS: The effect of levalbuterol on human bronchial SMC growth was compared with the effects of racemic albuterol and (S)-albuterol. Cells were fed albuterols and 3H-thymidine in 5% FBS and incubated for 24 h. The effect of (S)-albuterol on levalbuterol actions was also studied and so were the effects of cAMP/PKA, PI-3 kinase, NK-kappaB, and retinoblastoma (Rb) proteins on albuterols and human bronchial SMC proliferation. RESULTS: Levalbuterol inhibited cell proliferation at low concentrations. The growth-inhibitory effect of levalbuterol occurs via activation of the cAMP/PKA pathway. Addition of (S)-albuterol to levalbuterol decreased the growth-inhibitory effect of levalbuterol, and (S)-albuterol attenuated levalbuterol-induced cAMP release by 65%. Levalbuterol inhibited NF-kappaB and Rb protein expressions. ICI-118551 abrogated the inhibitory properties of levalbuterol. The PAF receptor antagonist CV-3988 inhibited (S)-albuterol-induced cell growth, with no effect on levalbuterol. CONCLUSIONS: Levalbuterol inhibits cell growth by activating the cAMP/PKA pathway and inhibiting PI-3 kinase, NF-kappaB and Rb protein expression, and (S)-albuterol induces cell growth by activating PAF-receptor-mediated cell signaling.


Asunto(s)
Agonistas Adrenérgicos beta/farmacología , Albuterol/farmacología , Asma/tratamiento farmacológico , Broncodilatadores/farmacología , Músculo Liso/efectos de los fármacos , Antagonistas Adrenérgicos beta/farmacología , Albuterol/antagonistas & inhibidores , Asma/patología , Broncodilatadores/antagonistas & inhibidores , Línea Celular , Proliferación Celular/efectos de los fármacos , Cromonas/farmacología , Colforsina/farmacología , AMP Cíclico/inmunología , Inhibidores Enzimáticos/farmacología , Humanos , Isoproterenol/farmacología , Morfolinas/farmacología , Músculo Liso/citología , Músculo Liso/inmunología , FN-kappa B/inmunología , Fosfatidilinositol 3-Quinasas/inmunología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Éteres Fosfolípidos/farmacología , Inhibidores de Agregación Plaquetaria/farmacología , Propanolaminas/farmacología , Proteína de Retinoblastoma/inmunología
15.
Am J Physiol Lung Cell Mol Physiol ; 288(5): L879-86, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15618453

RESUMEN

Circulating levels of platelet-activating factor (PAF) are high in the fetus, and PAF is active in maintaining high PVR in fetal hypoxia (Ibe BO, Hibler S, Raj J. J Appl Physiol 85: 1079-1085, 1998). PAF synthesis by fetal pulmonary vascular smooth muscle cells (PVSMC) is high in hypoxia, but how oxygen tension affects PAF receptor (PAF-r) binding in PVSMC is not known. We studied the effect of oxygen tension on PAF-r binding and signaling in fetal PVSMC. PAF binding was saturable. PAF-r density (B(max): fmol/10(6) cells; means +/- SE, n = 6), 25.2 +/- 0.77 during hypoxia (Po(2) <40 Torr), was higher than 13.9 +/- 0.44 during normoxia (Po(2) approximately 100 Torr). K(d) was twofold lower in hypoxia than normoxia. PAF-r protein expression, 35-40% greater in hypoxia, was inhibited by cycloheximide, a protein synthesis inhibitor, suggesting translational regulation. IP(3) release, an index of PAF-r-mediated cell signaling, was greater in hypoxia (EC(50): hypoxia, 2.94 +/- 0.61; normoxia, 5.85 +/- 0.51 nM). Exogenous PAF induced 50-90% greater intracellular calcium flux in cells during hypoxia, indicating hypoxia augments PAF-r-mediated cell signaling. PAF-r phosphorylation, with or without 5 nM PAF, was 40% greater in hypoxia. These data show 1) hypoxia upregulates PAF-r binding, PAF-r phosphorylation, and PAF-r-mediated intracellular signaling, evidenced by augmented IP(3) production and intracellular Ca(2+) flux; and 2) hypoxia-induced PAF-r phosphorylation results in activation of PAF-r-mediated signal transduction. The data suggest the fetal hypoxic environment facilitates PAF-r binding and signaling, thereby promoting PAF-mediated pulmonary vasoconstriction and maintenance of high PVR in utero.


Asunto(s)
Hipoxia Fetal/metabolismo , Hipoxia/metabolismo , Músculo Liso Vascular/metabolismo , Oxígeno/metabolismo , Glicoproteínas de Membrana Plaquetaria/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Calcio/metabolismo , Femenino , Hipoxia Fetal/fisiopatología , Hipoxia/fisiopatología , Fosfatos de Inositol/metabolismo , Pulmón/irrigación sanguínea , Pulmón/embriología , Fosforilación , Embarazo , Ovinos , Transducción de Señal/fisiología
16.
Mol Genet Metab ; 77(3): 237-48, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12409272

RESUMEN

We have demonstrated that platelet activating factor (PAF) plays an important physiological role in the maintenance of high pulmonary vascular tone in fetal lambs, a role attributable to increased PAF receptor binding (J. Appl. Physiol. 85 (1998) 1079; Am J. Physiol. 278 (2000) H1168). In this study, we examined the possibility that increased PAF synthesis via de novo and remodeling pathways as well as decreased PAF catabolism in hypoxic state of fetal lungs may account for the PAF action in vivo. We investigated effect of oxygen tension on PAF synthesis by ovine fetal intrapulmonary venous (PV) and arterial (PA) smooth muscle cells pulsed with [3H]choline (de novo), or [3H]acetate (remodeling), while PAF catabolism was studied by assay of acetylhydrolase (PAF-Ah) activity. Hypoxia stimulated PAF synthesis by choline incorporation (pmol/10(6)cells) in both PVSMC (1.14+/-0.13 vs 0.53+/-0.05) and PASMC (0.39+/-0.12 vs 0.22+/-0.04). Hypoxia stimulated PAF synthesis via remodeling pathway only in PVSMC (408+/-32 vs 225+/-17) which was 5-fold greater than in PASMC (77+/-15 vs 105+/-24), however, with A23187 in remodeling pathway, PAF synthesis increased 5-fold compared to baseline conditions and then synthesis in hypoxia was greater than in normoxia in both cell types. Phospholipase A2 protein expression was significantly higher in hypoxia in both cells and was approximately 2-fold higher in PVSMC. PAF-Ah activity (nmol lyso-PAF/min/mg protein) was greater in hypoxia vs normoxia in PVSMC (0.81+/-0.24 vs 0.44+/-0.088), but in PASMC activity was less in hypoxia vs normoxia (1.68+/-0.24 vs 3.93+/-0.44). Compared to PVSMC PAF-Ah activity in PASMC was 4-fold higher in hypoxia. Our data demonstrate that (1) PAF synthesis in intrapulmonary SMC of fetal lambs occurs by both de novo studied by choline incorporation and remodeling pathways, the latter being predominant. (2) There is heterogeneity in PAF synthetic and catabolic activities in lung vasculature of fetal lambs. We conclude that increased PAF synthesis in veins by the two synthetic pathways coupled with decreased catabolism will result in a higher venous PAF levels in the hypoxic environment of fetal lungs. We speculate that in vivo, a high PAF level in veins will make more PAF available for binding to its receptors so as to sustain the desired high venous tone in the fetal pulmonary circulation.


Asunto(s)
Músculo Liso Vascular/metabolismo , Oxígeno/metabolismo , Fosfolipasas A/genética , Factor de Activación Plaquetaria/metabolismo , 1-Alquil-2-acetilglicerofosfocolina Esterasa , Acetil-CoA C-Acetiltransferasa/metabolismo , Animales , Colina/metabolismo , Diacilglicerol Colinafosfotransferasa/metabolismo , Femenino , Pulmón/irrigación sanguínea , Pulmón/metabolismo , Fosfolipasas A/metabolismo , Fosfolipasas A2 , Factor de Activación Plaquetaria/biosíntesis , Ovinos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...