Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Am J Physiol Lung Cell Mol Physiol ; 318(4): L762-L772, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32073878

RESUMEN

Newborn pigs with chronic hypoxia-induced pulmonary hypertension (PH) have evidence of endothelial nitric oxide synthase (eNOS) uncoupling. In this model, we showed that therapies that promote eNOS coupling, either tetrahydrobiopterin (BH4), a NOS cofactor, or l-citrulline, a NO-l-arginine precursor, inhibit PH. We wanted to determine whether cotreatment with l-citrulline and a BH4 compound, sapropterin dihydrochloride, improves NO signaling and chronic hypoxia-induced PH more markedly than either alone. Normoxic (control) and hypoxic piglets were studied. Some hypoxic piglets received sole treatment with l-citrulline or BH4, or were cotreated with l-citrulline and BH4, from day 3 through day 10 of hypoxia. Catheters were placed for hemodynamic measurements, and pulmonary arteries were dissected to assess eNOS dimer-to-monomer ratios and NO production. In untreated hypoxic piglets, pulmonary vascular resistance (PVR) was higher and NO production and eNOS dimer-to-monomer ratios were lower than in normoxic piglets. Compared with the untreated hypoxic group, PVR was lower in hypoxic piglets cotreated with l-citrulline and BH4 and in those treated with l-citrulline alone but not for those treated solely with BH4. NO production and eNOS dimer-to-monomer ratios were greater for all three treated hypoxic groups compared with the untreated group. Notably, greater improvements in PVR, eNOS dimer-to-monomer ratios, and NO production were found in hypoxic piglets cotreated with l-citrulline and BH4 than in piglets treated with either alone. Cotreatment with l-citrulline and BH4 more effectively improves NO signaling and inhibits chronic hypoxia-induced PH than either treatment alone. Combination therapies may offer enhanced therapeutic capacity for challenging clinical conditions, such as chronic neonatal PH.


Asunto(s)
Biopterinas/análogos & derivados , Citrulina/farmacología , Hipertensión Pulmonar/tratamiento farmacológico , Hipoxia/metabolismo , Óxido Nítrico/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Animales Recién Nacidos , Arginina/metabolismo , Biopterinas/farmacología , Hipertensión Pulmonar/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/metabolismo , Porcinos , Resistencia Vascular/efectos de los fármacos
2.
Am J Physiol Heart Circ Physiol ; 316(4): H911-H919, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30794434

RESUMEN

We have previously shown that Na+-coupled neutral amino acid transporter 1 (SNAT1) modulates nitric oxide (NO) production in pulmonary arterial endothelial cells (PAECs) from newborn piglets. Specifically, the ability to increase NO production in response to the l-arginine-NO precursor l-citrulline is dependent on SNAT1 expression. Elucidating factors that regulate SNAT1 expression in PAECs could provide new insights and therapeutic targets relevant to NO production. Our major goals were to determine if reactive oxygen species (ROS) modulate SNAT1 expression in PAECs from newborn piglets and to evaluate the role of NADPH oxidase 1 (NOX1) and uncoupled endothelial NO synthase, enzymatic sources of ROS, in hypoxia-induced increases in SNAT1 expression. Treatment with either H2O2 or xanthine plus xanthine oxidase increased SNAT1 expression in PAECs from newborn piglets cultured under normoxic conditions. Hypoxia-induced increases in SNAT1 expression were inhibited by treatments with the ROS-removing agents catalase and superoxide dismutase, NOX1 siRNA, and the NO synthase inhibitor NG-nitro-l-arginine methyl ester. Both tetrahydropbiopterin (BH4) and l-citrulline, two therapies that decrease ROS by recoupling endothelial NO synthase, reduced the hypoxia-induced increase in SNAT1 expression. BH4 and l-citrulline treatment improved NO production in hypoxic PAECs despite a reduction in SNAT1 expression. In conclusion, SNAT1 expression is modulated by ROS in PAECs from newborn piglets. However, ROS-mediated decreases in SNAT1 expression per se do not implicate a reduction in NO production. Although SNAT1 may be critical to l-citrulline-induced increases in NO production, therapies designed to alter SNAT1 expression may not lead to a concordant change in NO production. NEW & NOTEWORTHY Na+-coupled neutral amino acid transporter 1 (SNAT1) modulates nitric oxide (NO) production in piglet pulmonary arterial endothelial cells. Factors that regulate SNAT1 expression in pulmonary arterial endothelial cells are unclear. Here, we show that ROS-reducing strategies inhibit hypoxia-induced increases in SNAT1 expression. l-Citrulline and tetrahydropbiopterin decrease SNAT1 expression but increase NO production. Although SNAT1 is modulated by ROS, changes in SNAT1 expression may not cause a concordant change in NO production.


Asunto(s)
Sistema de Transporte de Aminoácidos A/metabolismo , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Sistema de Transporte de Aminoácidos A/genética , Animales , Hipoxia de la Célula , Células Cultivadas , NADPH Oxidasas/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Oxígeno/metabolismo , Arteria Pulmonar/citología , Porcinos , Xantina Oxidasa/metabolismo
3.
Eur Respir J ; 51(6)2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29903860

RESUMEN

Pulmonary arterial hypertension (PAH) is a deadly disease with no cure. Alternate conversion of angiotensin II (AngII) to angiotensin-(1-7) (Ang-(1-7)) by angiotensin-converting enzyme 2 (ACE2) resulting in Mas receptor (Mas1) activation improves rodent models of PAH. Effects of recombinant human (rh) ACE2 in human PAH are unknown. Our objective was to determine the effects of rhACE2 in PAH.We defined the molecular effects of Mas1 activation using porcine pulmonary arteries, measured AngII/Ang-(1-7) levels in human PAH and conducted a phase IIa, open-label pilot study of a single infusion of rhACE2 (GSK2586881, 0.2 or 0.4 mg·kg-1 intravenously).Superoxide dismutase 2 (SOD2) and inflammatory gene expression were identified as markers of Mas1 activation. After confirming reduced plasma ACE2 activity in human PAH, five patients were enrolled in the trial. GSK2586881 was well tolerated with significant improvement in cardiac output and pulmonary vascular resistance. GSK2586881 infusion was associated with reduced plasma markers of inflammation within 2-4 h and increased SOD2 plasma protein at 2 weeks.PAH is characterised by reduced ACE2 activity. Augmentation of ACE2 in a pilot study was well tolerated, associated with improved pulmonary haemodynamics and reduced markers of oxidant and inflammatory mediators. Targeting this pathway may be beneficial in human PAH.


Asunto(s)
Citocinas/metabolismo , Hipertensión Pulmonar/tratamiento farmacológico , Peptidil-Dipeptidasa A/farmacología , Arteria Pulmonar/fisiopatología , Adulto , Anciano , Enzima Convertidora de Angiotensina 2 , Animales , Biomarcadores , Citocinas/efectos de los fármacos , Femenino , Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Proyectos Piloto , Prueba de Estudio Conceptual , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Superóxido Dismutasa/metabolismo , Porcinos , Resistencia Vascular/efectos de los fármacos
4.
Am J Physiol Lung Cell Mol Physiol ; 311(4): L743-L753, 2016 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-27542807

RESUMEN

We previously showed that newborn piglets who develop pulmonary hypertension during exposure to chronic hypoxia have diminished pulmonary vascular nitric oxide (NO) production and evidence of endothelial NO synthase (eNOS) uncoupling (Fike CD, Dikalova A, Kaplowitz MR, Cunningham G, Summar M, Aschner JL. Am J Respir Cell Mol Biol 53: 255-264, 2015). Tetrahydrobiopterin (BH4) is a cofactor that promotes eNOS coupling. Current clinical strategies typically invoke initiating treatment after the diagnosis of pulmonary hypertension, rather than prophylactically. The major purpose of this study was to determine whether starting treatment with an oral BH4 compound, sapropterin dihydrochloride (sapropterin), after the onset of pulmonary hypertension would recouple eNOS in the pulmonary vasculature and ameliorate disease progression in chronically hypoxic piglets. Normoxic (control) and hypoxic piglets were studied. Some hypoxic piglets received oral sapropterin starting on day 3 of hypoxia and continued throughout an additional 7 days of hypoxic exposure. Catheters were placed for hemodynamic measurements, and pulmonary arteries were dissected to assess eNOS dimer-to-monomer ratios (a measure of eNOS coupling), NO production, and superoxide (O2·-) generation. Although higher than in normoxic controls, pulmonary vascular resistance was lower in sapropterin-treated hypoxic piglets than in untreated hypoxic piglets. Consistent with eNOS recoupling, eNOS dimer-to-monomer ratios and NO production were greater and O2·- generation was less in pulmonary arteries from sapropterin-treated than untreated hypoxic animals. When started after disease onset, oral sapropterin treatment inhibits chronic hypoxia-induced pulmonary hypertension at least in part by recoupling eNOS in the pulmonary vasculature of newborn piglets. Rescue treatment with sapropterin may be an effective strategy to inhibit further development of pulmonary hypertension in newborn infants suffering from chronic cardiopulmonary conditions associated with episodes of prolonged hypoxia.


Asunto(s)
Biopterinas/análogos & derivados , Hipertensión Pulmonar/tratamiento farmacológico , Óxido Nítrico Sintasa de Tipo III/metabolismo , Administración Oral , Animales , Presión Arterial , Biopterinas/administración & dosificación , Hipoxia de la Célula , Evaluación Preclínica de Medicamentos , Hipertensión Pulmonar/enzimología , Arteria Pulmonar/enzimología , Sus scrofa
5.
Am J Respir Cell Mol Biol ; 53(2): 255-64, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25536367

RESUMEN

Infants with cardiopulmonary disorders associated with hypoxia develop pulmonary hypertension. We previously showed that initiation of oral L-citrulline before and continued throughout hypoxic exposure improves nitric oxide (NO) production and ameliorates pulmonary hypertension in newborn piglets. Rescue treatments, initiated after the onset of pulmonary hypertension, better approximate clinical strategies. Mechanisms by which L-citrulline improves NO production merit elucidation. The objective of this study was to determine whether starting L-citrulline after the onset of pulmonary hypertension inhibits disease progression and improves NO production by recoupling endothelial NO synthase (eNOS). Hypoxic and normoxic (control) piglets were studied. Some hypoxic piglets received oral L-citrulline starting on Day 3 of hypoxia and continuing throughout the remaining 7 days of hypoxic exposure. Catheters were placed for hemodynamic measurements, and pulmonary arteries were dissected to assess NO production and eNOS dimer-to-monomer ratios (a measure of eNOS coupling). Pulmonary vascular resistance was lower in L-citrulline-treated hypoxic piglets than in untreated hypoxic piglets but was higher than in normoxic controls. NO production and eNOS dimer-to-monomer ratios were greater in pulmonary arteries from L-citrulline-treated than from untreated hypoxic animals but were lower than in normoxic controls. When started after disease onset, oral L-citrulline treatment improves NO production by recoupling eNOS and inhibits the further development of chronic hypoxia-induced pulmonary hypertension in newborn piglets. Oral L-citrulline may be a novel strategy to halt or reverse pulmonary hypertension in infants suffering from cardiopulmonary conditions associated with hypoxia.


Asunto(s)
Citrulina/administración & dosificación , Hipertensión Pulmonar/tratamiento farmacológico , Animales , Animales Recién Nacidos , Arginina/sangre , Hipoxia de la Célula , Evaluación Preclínica de Medicamentos , Hipertensión Pulmonar/metabolismo , Sus scrofa
6.
Pulm Circ ; 13(2): e12221, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37063746

RESUMEN

Impaired nitric oxide (NO) signaling contributes to the development of pulmonary hypertension (PH). The l-arginine precursor, l-citrulline, improves NO signaling and has therapeutic potential in PH. However, there is evidence that l-citrulline might increase arginase activity, which in turn, has been shown to contribute to PH. Our major purpose was to determine if l-citrulline increases arginase activity in hypoxic human pulmonary artery endothelial cells (PAECs). In addition, to avoid potential adverse effects from high dose l-citrulline monotherapy, we evaluated whether the effect on NO signaling is greater using co-treatment with l-citrulline and another agent that improves NO signaling, folic acid, than either alone. Arginase activity was measured in human PAECs cultured under hypoxic conditions in the presence of l-citrulline (0-1 mM). NO production and endothelial nitric oxide synthase (eNOS) coupling, as assessed by eNOS dimer-to-monomer ratios, were measured in PAECs treated with l-citrulline and/or folic acid (0.2 µM). Arginase activity increased in hypoxic PAECs treated with 1 mM but not with either 0.05 or 0.1 mM l-citrulline. Co-treatment with folic acid and 0.1 mM l-citrulline increased NO production and eNOS dimer-to-monomer ratios more than treatment with either alone. The potential to increase arginase activity suggests that there might be plasma l-citrulline concentrations that should not be exceeded when using l-citrulline to treat PH. Rather than progressively increasing the dose of l-citrulline as a monotherapy, co-therapy with l-citrulline and folic acid merits consideration, due to the possibility of achieving efficacy at lower doses and minimizing side effects.

7.
Pediatr Res ; 70(2): 136-41, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21516056

RESUMEN

Reactive oxygen species (ROS) have been implicated in the pathogenesis of pulmonary hypertension. ROS might mediate vascular responses, at least in part, by stimulating prostanoid production. Our goals were to determine whether the effect of ROS on vascular tone is altered in resistance pulmonary arteries (PRAs) of newborn piglets with chronic hypoxia-induced pulmonary hypertension and the role, if any, of prostanoids in ROS-mediated responses. In cannulated, pressurized PRA, ROS generated by xanthine (X) plus xanthine oxidase (XO) had minimal effect on vascular tone in control piglets but caused significant vasoconstriction in hypoxic piglets. Both cyclooxygenase inhibition with indomethacin and thromboxane synthase inhibition with dazoxiben significantly blunted constriction to X+XO in hypoxic PRA. X+XO increased prostacyclin production (70 ± 8%) by a greater degree than thromboxane production (50 ± 6%) in control PRA; this was not the case in hypoxic PRA where the increases in prostacyclin and thromboxane production were not statistically different (78 ± 13% versus 216 ± 93%, respectively). Thromboxane synthase expression was increased in PRA from hypoxic piglets, whereas prostacyclin synthase expression was similar in PRA from hypoxic and control piglets. Under conditions of chronic hypoxia, altered vascular responses to ROS may contribute to pulmonary hypertension by a mechanism that involves the prostanoid vasoconstrictor, thromboxane.


Asunto(s)
Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/metabolismo , Hipoxia/complicaciones , Prostaglandinas/metabolismo , Arteria Pulmonar/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Vasoconstricción/fisiología , Animales , Animales Recién Nacidos , Inhibidores de la Ciclooxigenasa/farmacología , Sistema Enzimático del Citocromo P-450/metabolismo , Ensayo de Inmunoadsorción Enzimática , Hipertensión Pulmonar/fisiopatología , Imidazoles/farmacología , Immunoblotting , Indometacina/farmacología , Oxidorreductasas Intramoleculares/metabolismo , Estadísticas no Paramétricas , Sus scrofa , Tromboxano-A Sintasa/antagonistas & inhibidores , Tromboxano-A Sintasa/metabolismo , Vasoconstricción/efectos de los fármacos
8.
Pulm Circ ; 11(2): 20458940211006289, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33948161

RESUMEN

The L-arginine precursor, L-citrulline, re-couples endothelial nitric oxide synthase, increases nitric oxide production, and ameliorates chronic hypoxia-induced pulmonary hypertension in newborn pigs. L-arginine can induce arginase, which, in turn, may diminish nitric oxide production. Our major purpose was to determine if L-citrulline increases arginase activity in hypoxic piglet pulmonary arterial endothelial cells, and if so, concomitantly impacts the ability to increase endothelial nitric oxide synthase re-coupling and nitric oxide production. Piglet pulmonary arterial endothelial cells were cultured in hypoxic conditions with L-citrulline (0-3 mM) and/or the arginase inhibitor S-(2-boronoethyl)-L-cysteine. We measured arginase activity and nitric oxide production. We assessed endothelial nitric oxide synthase coupling by measuring endothelial nitric oxide synthase dimers and monomers. L-citrulline concentrations ≥0.5 mM increased arginase activity in hypoxic pulmonary arterial endothelial cells. L-citrulline concentrations ≥0.1 mM increased nitric oxide production and concentrations ≥0.5 mM elevated endothelial nitric oxide synthase dimer-to-monomer ratios. Co-treatment with L-citrulline and S-(2-boronoethyl)-L-cysteine elevated endothelial nitric oxide synthase dimer-to-monomer ratios more than sole treatment. Despite inducing arginase, L-citrulline increased nitric oxide production and endothelial nitric oxide synthase coupling in hypoxic piglet pulmonary arterial endothelial cells. However, these dose-dependent findings raise the possibility that there could be L-citrulline concentrations that elevate arginase to levels that negate improvements in endothelial nitric oxide synthase dysfunction. Moreover, our findings suggest that combining an arginase inhibitor with L-citrulline merits evaluation as a treatment for chronic hypoxia-induced pulmonary hypertension.

9.
Physiol Rep ; 9(21): e15096, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34762361

RESUMEN

Concomitant with developing pulmonary hypertension (PH), newborn piglets exposed to chronic hypoxia develop pulmonary vascular NO signaling impairments. PH is reduced and NO signaling is improved in chronically hypoxic piglets treated with the NO-arginine precursor, L-citrulline. Folic acid positively impacts NO signaling. We evaluated whether the effect on NO signaling and PH is greater using co-treatment with folic acid and L-citrulline than either alone. From day 3 to day 10 of hypoxia, piglets were treated solely with folic acid, solely with L-citrulline, or co-treated with both. Catheters were placed to measure in vivo hemodynamics. NO production was measured in vitro in dissected pulmonary arteries. Compared to normoxic piglets, pulmonary vascular resistance (PVR) was elevated and NO production was reduced in untreated hypoxic piglets. Regardless of treatment strategy, PVR was less in all three treated groups of hypoxic piglets when compared to the untreated hypoxic group. In addition, for all three groups of treated hypoxic piglets, NO production was higher than the untreated group. Improvements in PVR and NO production did not differ between piglets co-treated with folic acid and L-citrulline and those treated solely with either. Thus, the impact on NO production and PVR was not augmented by combining folic acid and L-citrulline treatments. Nonetheless, treatment with folic acid, either singly or when combined with L-citrulline, increases NO production and inhibits PH in chronically hypoxic newborn piglets. Folic acid merits consideration as a therapy for PH in human infants with chronic heart and lung conditions that are associated with chronic hypoxia.


Asunto(s)
Citrulina/uso terapéutico , Ácido Fólico/uso terapéutico , Hipertensión Pulmonar/tratamiento farmacológico , Óxido Nítrico/metabolismo , Transducción de Señal , Animales , Citrulina/administración & dosificación , Citrulina/farmacología , Combinación de Medicamentos , Femenino , Ácido Fólico/administración & dosificación , Ácido Fólico/farmacología , Hipertensión Pulmonar/etiología , Hipoxia/complicaciones , Masculino , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/metabolismo , Arteria Pulmonar/fisiopatología , Porcinos , Resistencia Vascular
10.
Am J Physiol Heart Circ Physiol ; 299(4): H1190-204, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20693398

RESUMEN

Aberrant interactions between heat shock protein (Hsp)90 and its client proteins could contribute to pulmonary hypertension. We tested the hypotheses that 1) the interaction between Hsp90 and its known client protein, endothelial nitric oxide synthase (eNOS), is impaired in pulmonary resistance arteries (PRAs) from piglets with pulmonary hypertension caused by exposure to 3 or 10 days of hypoxia and 2) Hsp90 interacts with the prostanoid pathway proteins prostacyclin synthase (PGIS) and/or thromboxane synthase (TXAS). We also determined whether Hsp90 antagonism with geldanamycin alters the agonist-induced synthesis of prostacyclin and thromboxane or alters PRA responses to these prostaglandin metabolites. Compared with normoxic piglets, less eNOS coimmunoprecipitated with Hsp90 in PRAs from hypoxic piglets. Despite reduced Hsp90-eNOS interactions, dilation to ACh was enhanced in geldanamycin-treated PRAs from hypoxic, but not normoxic, piglets. In PRAs from all groups of piglets, PGIS and TXAS coimmunoprecipitated with Hsp90. Geldanamycin reduced the ACh-induced synthesis of prostacyclin and thromboxane and altered responses to the thromboxane mimetic U-46619 in PRAs from all groups. Although geldanamycin enhanced responses to prostacyclin in PRAs from both groups of hypoxic piglets, geldanamycin had no effect on prostacyclin responses in PRAs from either group of normoxic piglets. Our findings indicate that Hsp90 influences both prostanoid and eNOS signaling in the pulmonary circulation of newborn piglets and that the impact of pharmacological inhibition of Hsp90 on these signaling pathways is altered during exposure to chronic hypoxia.


Asunto(s)
Animales Recién Nacidos/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Hipertensión Pulmonar/metabolismo , Hipoxia/metabolismo , Oxidorreductasas Intramoleculares/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Tromboxano-A Sintasa/metabolismo , Animales , Benzoquinonas/farmacología , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Hipertensión Pulmonar/etiología , Hipoxia/complicaciones , Lactamas Macrocíclicas/farmacología , Óxido Nítrico Sintasa/metabolismo , Arteria Pulmonar/fisiopatología , Transducción de Señal , Porcinos , Resistencia Vascular/efectos de los fármacos , Resistencia Vascular/fisiología
11.
Pediatr Pulmonol ; 55(10): 2762-2772, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32662946

RESUMEN

BACKGROUND: Dysregulated nitric oxide (NO) signaling contributes to chronic hypoxia (CH)-induced pulmonary hypertension (PH). NO signaling is improved and pulmonary vascular resistance (PVR) is reduced in CH piglets treated with the l-arginine-NO precursor, l-citrulline. We hypothesized that l-citrulline might cause structural changes in the pulmonary circulation that would contribute to the reduction in PVR and that the l-citrulline-induced structural changes would be accompanied by alterations in vascular endothelial growth factor (VEGF) signaling. METHODS: We evaluated small pulmonary arterial (PA) wall thickness, lung capillary density, and protein abundances of VEGF, VEGFR2, and phospho (p)-VEGFR2 in PA and peripheral lung samples of piglets raised in the lab in CH (10%-12% O2 ) from the day of life (DOL) 2 until DOL 11 to 12 or raised in room air (normoxia) by the vendor and studied on arrival to the lab on DOL 11 to 12. Some CH piglets were treated with oral l-citrulline (1-1.5 g/kg/d) starting on the third day of hypoxia. RESULTS: PA wall thickness was 32% less and lung capillary formation was nearly doubled in l-citrulline treated than untreated CH piglets. Both of these l-citrulline-induced structural changes in the pulmonary circulation were accompanied by altered amounts of VEGF protein but not by altered amounts of either VEGFR2 or p-VEGFR2 proteins. CONCLUSIONS: Alterations in the structure of the pulmonary circulation in CH piglets by l-citrulline are unlikely to be mediated by overall VEGF signaling. Nonetheless, l-citrulline- induced structural changes should reduce PVR and thereby contribute to the amelioration of CH-induced PH.


Asunto(s)
Citrulina/farmacología , Hipoxia/fisiopatología , Circulación Pulmonar/efectos de los fármacos , Animales , Animales Recién Nacidos , Capilares/efectos de los fármacos , Capilares/fisiología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/fisiología , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/fisiología , Porcinos , Factor A de Crecimiento Endotelial Vascular/metabolismo
12.
Am J Physiol Lung Cell Mol Physiol ; 297(3): L506-11, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19617312

RESUMEN

Newborn piglets develop pulmonary hypertension and have diminished pulmonary vascular nitric oxide (NO) production when exposed to chronic hypoxia. NO is produced by endothelial NO synthase (eNOS) in the pulmonary vascular endothelium using l-arginine as a substrate and producing l-citrulline as a byproduct. l-Citrulline is metabolized to l-arginine by two enzymes that are colocated with eNOS in pulmonary vascular endothelial cells. The purpose of this study was to determine whether oral supplementation with l-citrulline during exposure of newborn piglets to 10 days of chronic hypoxia would prevent the development of pulmonary hypertension and increase pulmonary NO production. A total of 17 hypoxic and 17 normoxic control piglets were studied. Six of the 17 hypoxic piglets were supplemented with oral l-citrulline starting on the first day of hypoxia. l-Citrulline supplementation was provided orally twice a day. After 10 days of hypoxia or normoxia, the animals were anesthetized, hemodynamic measurements were performed, and the lungs were perfused in situ. Pulmonary arterial pressure and pulmonary vascular resistance were significantly lower in hypoxic animals treated with l-citrulline compared with untreated hypoxic animals (P < 0.001). In vivo exhaled NO production (P = 0.03) and nitrite/nitrate accumulation in the perfusate of isolated lungs (P = 0.04) were significantly higher in l-citrulline-treated hypoxic animals compared with untreated hypoxic animals. l-Citrulline supplementation ameliorated the development of pulmonary hypertension and increased NO production in piglets exposed to chronic hypoxia. We speculate that l-citrulline may benefit neonates exposed to prolonged periods of hypoxia from cardiac or pulmonary causes.


Asunto(s)
Citrulina/farmacología , Hipertensión Pulmonar/etiología , Hipoxia/complicaciones , Aminoácidos/sangre , Animales , Animales Recién Nacidos , Western Blotting , Enfermedad Crónica , Espiración/efectos de los fármacos , Hemodinámica/efectos de los fármacos , Hipertensión Pulmonar/sangre , Hipertensión Pulmonar/fisiopatología , Hipoxia/sangre , Hipoxia/fisiopatología , Técnicas In Vitro , Pulmón/efectos de los fármacos , Pulmón/enzimología , Pulmón/fisiopatología , Nitratos/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Perfusión , Presión , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/fisiopatología , Sus scrofa
13.
J Appl Physiol (1985) ; 99(2): 670-6, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15802364

RESUMEN

The pulmonary vasoconstrictor, thromboxane, may contribute to the development of pulmonary hypertension. Our objective was to determine whether a combined thromboxane synthase inhibitor-receptor antagonist, terbogrel, prevents pulmonary hypertension and the development of aberrant pulmonary arterial responses in newborn piglets exposed to 3 days of hypoxia. Piglets were maintained in room air (control) or 11% O(2) (hypoxic) for 3 days. Some hypoxic piglets received terbogrel (10 mg/kg po bid). Pulmonary arterial pressure, pulmonary wedge pressure, and cardiac output were measured in anesthetized animals. A cannulated artery technique was used to measure responses to acetylcholine. Pulmonary vascular resistance for terbogrel-treated hypoxic piglets was almost one-half the value of untreated hypoxic piglets but remained greater than values for control piglets. Dilation to acetylcholine in preconstricted pulmonary arteries was greater for terbogrel-treated hypoxic than for untreated hypoxic piglets, but it was less for pulmonary arteries from both groups of hypoxic piglets than for control piglets. Terbogrel may ameliorate pulmonary artery dysfunction and attenuate the development of chronic hypoxia-induced pulmonary hypertension in newborns.


Asunto(s)
Hipertensión Pulmonar/tratamiento farmacológico , Hipertensión Pulmonar/etiología , Hipoxia/complicaciones , Hipoxia/tratamiento farmacológico , Piridinas/administración & dosificación , Tromboxanos/antagonistas & inhibidores , Animales , Enfermedad Crónica , Circulación Pulmonar/efectos de los fármacos , Porcinos
14.
J Appl Physiol (1985) ; 98(3): 1111-8; discussion 1091, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15516370

RESUMEN

Our objective was to determine whether cyclooxygenase (COX)-2-dependent metabolites contribute to the altered pulmonary vascular responses that manifest in piglets with chronic hypoxia-induced pulmonary hypertension. Piglets were raised in either room air (control) or hypoxia for 3 days. The effect of the COX-2 selective inhibitor NS-398 on responses to arachidonic acid or acetylcholine (ACh) was measured in endothelium-intact and denuded pulmonary arteries (100- to 400-microm diameter). Pulmonary arterial production of the stable metabolites of thromboxane and prostacyclin was assessed in the presence and absence of NS-398. Dilation to arachidonic acid was greater for intact control than for intact hypoxic arteries, was unchanged by NS-398 in intact arteries of either group, and was augmented by NS-398 in denuded hypoxic arteries. ACh responses, which were dilation in intact control arteries but constriction in intact and denuded hypoxic arteries, were diminished by NS-398 treatment of all arteries. NS-398 reduced prostacyclin production by control pulmonary arteries and reduced thromboxane production by hypoxic pulmonary arteries. COX-2-dependent contracting factors, such as thromboxane, contribute to aberrant pulmonary arterial responses in piglets exposed to 3 days of hypoxia.


Asunto(s)
Ácido Araquidónico/metabolismo , Hipertensión Pulmonar/enzimología , Hipoxia/enzimología , Prostaglandina-Endoperóxido Sintasas/metabolismo , Arteria Pulmonar/enzimología , Animales , Animales Recién Nacidos , Enfermedad Crónica , Ciclooxigenasa 2 , Hipertensión Pulmonar/etiología , Hipoxia/complicaciones , Transducción de Señal , Porcinos , Distribución Tisular
15.
Pediatr Pulmonol ; 40(1): 72-80, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15880376

RESUMEN

Devising therapies that might prevent the onset or progression of pulmonary hypertension in newborns has received little attention. Our major objective was to determine whether sildenafil, a selective phosphodiesterase inhibitor, prevents the development of an early stage of chronic hypoxia-induced pulmonary hypertension in newborn pigs. Another objective was to determine whether sildenafil causes pulmonary vasodilation without systemic vasodilation in piglets with chronic pulmonary hypertension. Piglets were raised in room air (control, n = 5) or 10-11% O(2) (hypoxic, n = 17) for 3 days. Some piglets (n = 4) received oral sildenafil, 12 mg/kg/day, throughout exposure to hypoxia. All piglets were anesthetized and catheterized, and pulmonary arterial pressure (Ppa), pulmonary wedge pressure (Pw), aortic pressure (Ao), and cardiac output (CO) were measured. Then for some piglets raised in hypoxia for 3 days, a single oral sildenafil dose (3 mg/kg, n = 6) or placebo (n = 5) was given, and hemodynamic measurements were repeated. For piglets raised in hypoxia for 3 days, mean Ppa and calculated PVR were elevated above respective values in control piglets. Mean Ppa and PVR did not differ between piglets that received sildenafil throughout exposure to hypoxia and those that did not. For piglets with chronic hypoxia-induced pulmonary hypertension that received a single oral dose of sildenafil, mean Ppa and PVR decreased, while mean Pw, CO, mean Ao, and systemic vascular resistance remained the same. All hemodynamic measurements were unchanged after placebo. Oral sildenafil did not influence the early stage of chronic hypoxia-induced pulmonary hypertension in newborn piglets. However, a single oral dose of sildenafil caused pulmonary vasodilation, without systemic vasodilation, in piglets with chronic hypoxia-induced pulmonary hypertension, which may have therapeutic implications.


Asunto(s)
Hipertensión Pulmonar/tratamiento farmacológico , Hipoxia/complicaciones , Enfermedades del Recién Nacido/tratamiento farmacológico , Piperazinas/administración & dosificación , Vasodilatadores/administración & dosificación , Administración Oral , Animales , Animales Recién Nacidos , Enfermedad Crónica , GMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Relación Dosis-Respuesta a Droga , Hemodinámica/efectos de los fármacos , Humanos , Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/fisiopatología , Recién Nacido , Enfermedades del Recién Nacido/fisiopatología , Pulmón/irrigación sanguínea , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/metabolismo , Purinas , Valores de Referencia , Citrato de Sildenafil , Sulfonas , Porcinos , Resultado del Tratamiento
16.
J Appl Physiol (1985) ; 92(1): 67-74, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11744644

RESUMEN

Pulmonary hypertension and blunted pulmonary vascular responses to ACh develop when newborn pigs are exposed to chronic hypoxia for 3 days. To determine whether a cyclooxygenase (COX)-dependent contracting factor, such as thromboxane, is involved with altered pulmonary vascular responses to ACh, newborn piglets were raised in 11% O(2) (hypoxic) or room air (control) for 3 days. Small pulmonary arteries (100-400 microm diameter) were cannulated and pressurized, and their responses to ACh were measured before and after either the COX inhibitor indomethacin; a thromboxane synthesis inhibitor, dazoxiben or feregrelate; or the thromboxane-PGH(2)-receptor antagonist SQ-29548. In control arteries, indomethacin reversed ACh responses from dilation to constriction. In contrast, hypoxic arteries constricted to ACh before indomethacin and dilated to ACh after indomethacin. Furthermore, ACh constriction in hypoxic arteries was nearly abolished by either dazoxiben, feregrelate, or SQ-29548. These findings suggest that thromboxane is the COX-dependent contracting factor that underlies the constrictor response to ACh that develops in small pulmonary arteries of piglets exposed to 3 days of hypoxia. The early development of thromboxane-mediated constriction may contribute to the pathogenesis of chronic hypoxia-induced pulmonary hypertension in newborns.


Asunto(s)
Animales Recién Nacidos/fisiología , Hipoxia/fisiopatología , Prostaglandina-Endoperóxido Sintasas/fisiología , Circulación Pulmonar/fisiología , Acetilcolina/metabolismo , Animales , Calcimicina/farmacología , Enfermedad Crónica , Endotelio Vascular/fisiología , Femenino , Hipoxia/enzimología , Masculino , Tono Muscular/fisiología , Músculo Liso Vascular/fisiología , S-Nitroso-N-Acetilpenicilamina/farmacología , Porcinos , Tromboxanos/fisiología
17.
Pulm Circ ; 3(3): 551-63, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24618540

RESUMEN

Abstract Changes in voltage-gated K(+) (Kv) channel function contribute to the pathogenesis of pulmonary hypertension. Yet the mechanisms underlying Kv channel impairments in the pulmonary circulation remain unclear. We tested the hypothesis that reactive oxygen species (ROSs) contribute to the Kv channel dysfunction that develops in resistance-level pulmonary arteries (PRAs) of piglets exposed to chronic in vivo hypoxia. Piglets were raised in either room air (control) or hypoxia for 3 or 10 days. To evaluate Kv channel function, responses to the Kv channel antagonist 4-aminopyridine (4-AP) were measured in cannulated PRAs. To assess the influence of ROSs, PRAs were treated with the ROS-removing agent M40403 (which dismutates superoxide to hydrogen peroxide), plus polyethylene glycol catalase (which converts hydrogen peroxide to water). Responses to 4-AP were diminished in PRAs from both groups of hypoxic piglets. ROS-removing agents had no impact on 4-AP responses in PRAs from piglets exposed to 3 days of hypoxia but significantly increased the response to 4-AP in PRAs from piglets exposed to 10 days of hypoxia. Kv channel function is impaired in PRAs of piglets exposed to 3 or 10 days of in vivo hypoxia. ROSs contribute to Kv channel dysfunction in PRAs from piglets exposed to hypoxia for 10 days but are not involved with the Kv channel dysfunction that develops within 3 days of exposure to hypoxia. Therapies to remove ROSs might improve Kv channel function and thereby ameliorate the progression, but not the onset, of pulmonary hypertension in chronically hypoxic newborn piglets.

18.
Neonatology ; 101(1): 28-39, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-21791937

RESUMEN

BACKGROUND: The use of phosphodiesterase 5 (PDE5) inhibitors to treat newborns with pulmonary hypertension is increasing. The effect of PDE5 inhibitors on the neonatal cerebral circulation remains unknown. The neonatal piglet model of chronic hypoxia-induced pulmonary hypertension allows the study of the effects of PDE5 inhibitors on both the pulmonary and cerebral circulations. OBJECTIVES: To determine whether the PDE5 inhibitor, zaprinast, causes dilation in pulmonary and middle cerebral arteries (MCA) of normoxic newborn piglets and those with chronic hypoxia-induced pulmonary hypertension, and to evaluate whether zaprinast alters responses to increased pressure (autoregulatory ability) of the MCA. METHODS: Two-day-old piglets were raised in normoxia or hypoxia for 3 or 10 days. Pulmonary arteries and MCA were isolated and pressurized, after which changes in diameter to zaprinast were measured. MCA pressure-diameter relationships were determined. RESULTS: Dilation to zaprinast was similar in pulmonary arteries from normoxic and hypoxic piglets. Zaprinast dilated MCA from all groups but the response was diminished in MCA from piglets raised in hypoxia for 10 days. MCA pressure-diameter relationships (autoregulation) did not differ between the groups. CONCLUSIONS: Pulmonary artery dilation to zaprinast supports the use of PDE5 inhibitors to treat pulmonary hypertension in neonates. PDE5 inhibitors function as MCA dilators but do not impair the pressure-diameter behavior of the cerebral circulation of either normoxic newborn piglets or those with chronic hypoxia-induced pulmonary hypertension. These findings suggest that cerebral autoregulation is likely to be intact with acute PDE5 inhibitor treatment in infants with pulmonary hypertension in conditions associated with chronic hypoxia.


Asunto(s)
3',5'-AMP Cíclico Fosfodiesterasas/antagonistas & inhibidores , Hipertensión Pulmonar/tratamiento farmacológico , Hipoxia/complicaciones , Arteria Cerebral Media/efectos de los fármacos , Inhibidores de Fosfodiesterasa/farmacología , Arteria Pulmonar/efectos de los fármacos , Purinonas/farmacología , 3',5'-AMP Cíclico Fosfodiesterasas/metabolismo , Animales , Animales Recién Nacidos , Enfermedad Crónica , Modelos Animales de Enfermedad , Hipertensión Pulmonar/etiología , Arteria Cerebral Media/fisiopatología , Arteria Pulmonar/fisiopatología , Porcinos , Vasodilatación/efectos de los fármacos
19.
Cardiovasc Res ; 95(3): 375-84, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22673370

RESUMEN

AIMS: Pulmonary arterial endothelial cells (PAECs) express the enzymes needed for generation of l-arginine from intracellular l-citrulline but do not express the enzymes needed for de novo l-citrulline synthesis. Hence, l-citrulline levels in PAECs are dependent on l-citrulline transport. Once generated, l-arginine can be converted to l-citrulline and nitric oxide (NO) by the enzyme NO synthase. We sought to determine whether hypoxia, a condition aetiologically linked to pulmonary hypertension, alters the transport of l-citrulline and the expression of the sodium-coupled neutral amino acid transporters (SNATs) in PAECs from newborn piglets. METHODS AND RESULTS: PAECs isolated from newborn piglets were cultured under normoxic and hypoxic conditions and used to measure SNAT1, 2, 3, and 5 protein expression and (14)C-l-citrulline uptake. SNAT1 protein expression was increased, while SNAT2, SNAT3, and SNAT5 expression was unaltered in hypoxic PAECs. (14)C-l-citrulline uptake was increased in hypoxic PAECs. Studies with inhibitors of System A (SNAT1/2) and System N (SNAT3/5) revealed that the increased (14)C-l-citrulline uptake was largely due to System A-mediated transport. Additional studies were performed to evaluate SNAT protein expression and l-citrulline levels in lungs of piglets with chronic hypoxia-induced pulmonary hypertension and comparable age controls. Lungs from piglets raised in chronic hypoxia exhibited greater SNAT1 expression and higher l-citrulline levels than lungs from controls. CONCLUSION: Increased SNAT1 expression and the concomitant enhanced ability to transport l-citrulline in PAECs could represent an important regulatory mechanism to counteract NO signalling impairments known to occur during the development of chronic hypoxia-induced pulmonary hypertension in newborns.


Asunto(s)
Sistema de Transporte de Aminoácidos A/metabolismo , Citrulina/metabolismo , Células Endoteliales/metabolismo , Hipoxia/metabolismo , Arteria Pulmonar/metabolismo , Circulación Pulmonar , Sistema de Transporte de Aminoácidos A/antagonistas & inhibidores , Animales , Animales Recién Nacidos , Transporte Biológico , Células Cultivadas , Enfermedad Crónica , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Hipertensión Pulmonar Primaria Familiar , Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/fisiopatología , Hipoxia/complicaciones , Hipoxia/fisiopatología , Moduladores del Transporte de Membrana/farmacología , Óxido Nítrico/metabolismo , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/fisiopatología , Porcinos , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA