Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Br J Clin Pharmacol ; 83(11): 2503-2516, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28635050

RESUMEN

AIM: We encountered a case of fetal toxicity due to ductus arteriosus (DA) constriction in a 36-week pregnant woman who had applied multiple ketoprofen patches. The aim of the present study was to present the case and develop a model to predict quantitatively the fetal toxicity risk of transdermal administration of ketoprofen. METHODS: Human placenta perfusion studies were conducted to estimate transplacental pharmacokinetic (PK) parameters. Using a developed model and these parameters, human fetal plasma concentration profiles of ketoprofen administered to mothers were simulated. Using pregnant rats, DA constriction and fetal plasma drug concentration after ketoprofen administration were measured, fitted to an Emax model, and extrapolated to humans. RESULTS: Transplacental transfer value at the steady state of ketoprofen was 4.82%, which was approximately half that of antipyrine (passive marker). The model and PK parameters predicted almost equivalent mother and fetus drug concentrations at steady state after transdermal ketoprofen administration in humans. Maximum DA constriction and maximum plasma concentration of ketoprofen after administration to rat dams were observed at different times: 4 h and 1 h, respectively. The model accurately described the delay in DA constriction with respect to the fetal ketoprofen concentration profile. The model with effect compartment and the obtained parameters predicted that use of multiple ketoprofen patches could potentially cause severe DA constriction in the human fetus, and that fetal toxicity might persist after ketoprofen discontinuation by the mother, as observed in our case. CONCLUSION: The present approach successfully described the sustained fetal toxicity after discontinuing the transdermal administration of ketoprofen.


Asunto(s)
Antiinflamatorios no Esteroideos/efectos adversos , Conducto Arterial/efectos de los fármacos , Cetoprofeno/efectos adversos , Dolor de la Región Lumbar/tratamiento farmacológico , Modelos Biológicos , Complicaciones del Embarazo/tratamiento farmacológico , Adulto , Animales , Antiinflamatorios no Esteroideos/farmacocinética , Antipirina/farmacocinética , Biomarcadores Farmacológicos/metabolismo , Constricción Patológica/inducido químicamente , Conducto Arterial/patología , Femenino , Humanos , Cetoprofeno/farmacocinética , Intercambio Materno-Fetal/efectos de los fármacos , Modelos Animales , Perfusión/métodos , Placenta/metabolismo , Embarazo , Tercer Trimestre del Embarazo/efectos de los fármacos , Ratas , Ratas Wistar , Medición de Riesgo/métodos , Parche Transdérmico/efectos adversos
2.
Pediatr Int ; 59(12): 1246-1251, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28901668

RESUMEN

BACKGROUND: Diazoxide, an ATP-sensitive potassium channel opener, is the main therapeutic agent for treating hyperinsulinemic hypoglycemia. The aim of this study was to determine the in vivo ductus arteriosus (DA)-dilating effects of diazoxide in fetal and neonatal rats. METHODS: Near-term rat pups delivered via cesarean section were housed at 33°C. After rapid whole-body freezing, the ductus arteriosus (DA) diameter was measured using a microscope and a micrometer. Full-term pregnant rats (gestational day 21) were injected i.p. with diazoxide (10 and 100 mg/kg) 4 h before delivery, and the neonatal DA diameter was measured at 0, 30, or 60 min after birth. The newborn rats were also injected i.p. with diazoxide (10 and 100 mg/kg) at birth or 60 min after birth. DA was measured at 0, 30, or 60 min after injection. In the fetal investigation, the effect of diazoxide was studied via simultaneous application of indomethacin (10 mg/kg) and L-nitroarginine methyl ester (L-NAME) on gestational days 21 and 19. RESULTS: The control rats had rapid postnatal DA constriction (diameter, 0.80 and 0.08 mm at 0 and 60 min after birth, respectively). Diazoxide had a dose-dependent inhibitory effect on postnatal DA constriction. Prenatal diazoxide (10 mg/kg) inhibited postnatal DA closure (0.20 mm at 60 min after birth). The diazoxide injection (10 mg) at birth inhibited postnatal DA closure (0.14 mm at 60 min after birth). Diazoxide injection in 60-min-old rats dilated the constricted DA at 60 min (0.10 mm vs. 0.02 mm in the controls). In the fetal investigation, diazoxide inhibited the fetal DA constrictive effect of indomethacin and L-NAME. CONCLUSION: Diazoxide attenuates postnatal DA constriction and dilates a closing DA in fetal and neonatal rats.


Asunto(s)
Diazóxido/farmacología , Conducto Arterial/efectos de los fármacos , Vasodilatadores/farmacología , Animales , Animales Recién Nacidos , Diazóxido/administración & dosificación , Dilatación Patológica/inducido químicamente , Conducto Arterial/fisiopatología , Femenino , Canales KATP/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Ratas , Ratas Wistar , Vasodilatadores/administración & dosificación
3.
Nat Genet ; 39(8): 1013-7, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17603482

RESUMEN

Noonan syndrome is characterized by short stature, facial dysmorphia and a wide spectrum of congenital heart defects. Mutations of PTPN11, KRAS and SOS1 in the RAS-MAPK pathway cause approximately 60% of cases of Noonan syndrome. However, the gene(s) responsible for the remainder are unknown. We have identified five different mutations in RAF1 in ten individuals with Noonan syndrome; those with any of four mutations causing changes in the CR2 domain of RAF1 had hypertrophic cardiomyopathy (HCM), whereas affected individuals with mutations leading to changes in the CR3 domain did not. Cells transfected with constructs containing Noonan syndrome-associated RAF1 mutations showed increased in vitro kinase and ERK activation, and zebrafish embryos with morpholino knockdown of raf1 demonstrated the need for raf1 for the development of normal myocardial structure and function. Thus, our findings implicate RAF1 gain-of-function mutations as a causative agent of a human developmental disorder, representing a new genetic mechanism for the activation of the MAPK pathway.


Asunto(s)
Mutación Missense , Síndrome de Noonan/genética , Proteínas Proto-Oncogénicas c-raf/genética , Animales , Línea Celular , Línea Celular Transformada , Femenino , Corazón/embriología , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Miocardio/metabolismo , Estructura Terciaria de Proteína , Proteína Tirosina Fosfatasa no Receptora Tipo 11 , Proteínas Tirosina Fosfatasas/genética , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Proto-Oncogénicas c-raf/química , Proteínas Proto-Oncogénicas c-raf/metabolismo , Pez Cebra , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
4.
Pediatr Res ; 67(2): 173-6, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19809378

RESUMEN

Furosemide increases prostaglandin production and may be associated with patent ductus arteriosus (PDA). We aimed to clarify the in vivo ductus-dilating effects of furosemide in neonatal rats. Near-term rat pups delivered by a cesarean section were housed at 33 degrees C. After a rapid whole-body freezing, the DA diameter was measured using a microscope and a micrometer. Pregnant rats (gestational day 21) were s.c. injected with furosemide 4 h before delivery, and the neonatal DA was examined 0, 15, 30, 60, and 120 min after birth. Furosemide was also s.c. injected into 60-min-old rats and the DA diameter was examined 30, 60, and 120 min later. The control rats showed a rapid postnatal DA constriction (diameter: 0.80 and 0.08 mm at 0 and 60 min after birth, respectively). Prenatally administered furosemide delayed postnatal DA closure (0.36 mm at 60 min after birth). Furosemide injection in 60-min-old rats dilated the constricted DA at 60 min (0.25 versus 0.02 mm in the controls). Indomethacin inhibited furosemide-induced DA dilatation. Furosemide delays DA closure and dilates the constricted DA in neonatal rats. If furosemide has similar effects in human preterm neonates, caution may be warranted in its use in the treatment of infants with PDA.


Asunto(s)
Conducto Arterioso Permeable/patología , Conducto Arterial/patología , Animales , Animales Recién Nacidos , Cesárea , Inhibidores de la Ciclooxigenasa/farmacología , Dilatación Patológica , Modelos Animales de Enfermedad , Conducto Arterial/efectos de los fármacos , Conducto Arterial/metabolismo , Conducto Arterioso Permeable/inducido químicamente , Conducto Arterioso Permeable/metabolismo , Conducto Arterioso Permeable/prevención & control , Femenino , Furosemida/administración & dosificación , Edad Gestacional , Indometacina/farmacología , Inyecciones Subcutáneas , Embarazo , Prostaglandinas E/metabolismo , Ratas , Ratas Wistar , Factores de Tiempo
5.
Am J Med Genet A ; 149A(10): 2216-9, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19725129

RESUMEN

Here we report on a patient with multiple lentigines, hypertelorism, short stature, arachnodactyly, scoliosis, dissecting aneurysm, hypertrophic cardiomyopathy and developmental delay, and a family history of Marfan syndrome. The patient is affected with both Marfan and LEOPARD syndromes. Mutational screening of the FBN1 gene showed a c.1464T>A (p.C488X) mutation and screening of the PTPN11 gene showed a c.836A>G (p.Y279C) mutation. We conclude that each mutation contributed independently to individual features in the ocular and cardiovascular systems, although short stature was more significantly influenced by the p.Y279C change in PTPN11 rather than the mutation in FBN1. To our knowledge, this is the first report of mutations in both FBN1 and PTPN11 with combined phenotypes of Marfan and LEOPARD syndromes.


Asunto(s)
Síndrome LEOPARD/complicaciones , Síndrome LEOPARD/genética , Síndrome de Marfan/complicaciones , Síndrome de Marfan/genética , Adulto , Análisis Mutacional de ADN , Resultado Fatal , Fibrilina-1 , Fibrilinas , Genotipo , Humanos , Masculino , Proteínas de Microfilamentos/genética , Linaje , Fenotipo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética
6.
Prostaglandins Other Lipid Mediat ; 79(1-2): 34-42, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16516808

RESUMEN

We studied the transplacental ductal constrictive effects of a selective cyclooxygenase (COX)-1 inhibitor (SC560), six selective COX-2 inhibitors including rofecoxib, and a non-selective COX inhibitor (indomethacin). Each drug was administered to the pregnant rats, and fetal ductus arteriosus (DA) was studied with a whole-body freezing method. The inner diameter ratio of the DA to the main pulmonary artery (DA/PA) was 1.02+/-0.03 (mean+/-S.E.M.) in controls. Every drug constricted the DA dose-dependently. In preterm rats on the 19th day of gestation, 10mg/kg of SC560, rofecoxib and indomethacin caused ductal constriction, with DA/PA reduced to 0.76+/-0.02, 0.80+/-0.03 and 0.75+/-0.02, respectively. In near-term on the 21st day, 10mg/kg of them caused ductal constriction, with DA/PA to 0.74+/-0.04, 0.26+/-0.02 and 0.33+/-0.05. In conclusion, both COX-1 and COX-2 selective inhibitors constrict fetal DA. They are not better alternatives for the fetus than non-selective COX inhibitors for tocolysis.


Asunto(s)
Inhibidores de la Ciclooxigenasa/efectos adversos , Conducto Arterioso Permeable/fisiopatología , Enfermedades Fetales/fisiopatología , Tocólisis/efectos adversos , Vasoconstricción/efectos de los fármacos , Animales , Ciclooxigenasa 1/metabolismo , Ciclooxigenasa 2/metabolismo , Inhibidores de la Ciclooxigenasa/administración & dosificación , Relación Dosis-Respuesta a Droga , Conducto Arterioso Permeable/inducido químicamente , Conducto Arterioso Permeable/enzimología , Femenino , Enfermedades Fetales/inducido químicamente , Enfermedades Fetales/enzimología , Feto/anomalías , Feto/enzimología , Feto/patología , Feto/fisiopatología , Embarazo , Ratas , Ratas Wistar
7.
Paediatr Drugs ; 8(1): 55-69, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16494512

RESUMEN

This article reviews reports of ACE inhibitor use in pediatric heart failure and summarizes the present implications for clinical practice. Captopril, enalapril, and cilazapril are orally active ACE inhibitors, and widely used in pediatric cardiology, although more than ten other ACE inhibitors have been applied clinically in adults. Effects of ACE inhibitors on the renin-angiotensin-aldosterone system in pediatric patients are similar to those in adults. ACE inhibitors lower aortic pressure and systemic vascular resistance, do not affect pulmonary vascular resistance significantly, and lower left atrial and right atrial pressures in pediatric patients with heart failure. In infants with a large ventricular septal defect and pulmonary hypertension, ACE inhibitors decrease left-to-right shunt in those infants with elevated systemic vascular resistance. ACE inhibitors induce a small increase in left ventricular ejection fraction, left ventricular fractional shortening, and systemic blood flow in children with left ventricular dysfunction, mitral regurgitation, and aortic regurgitation. These beneficial effects usually persist long term without the development of tolerance. Therapeutic trials of ACE inhibitors have been reported in children with heart failure and divergent hemodynamics, including myocardial dysfunction, left-to-right shunt, such as large ventricular septal defect and pulmonary hypertension, aortic or mitral regurgitation, and Fontan circulation. Hypotension and renal failure usually occur within 5 days after starting ACE inhibition or increasing the dose and, in most cases, recovery is seen after reduction or cessation of the drug. With all ACE inhibitors, smaller doses are administered initially to prevent excessive hypotension, and doses are increased gradually to the target dose. Captopril is administered orally, usually every 8 hours. Daily doses range from 0.3 to 1.5 mg/kg in children. Enalapril is administered orally, once or twice a day, and daily doses range from 0.1 to 0.5 mg/kg. Enalaprilat is administered intravenously, one to three times a day, in doses ranging from 0.01 to 0.05 mg/kg/dose. For the treatment of chronic heart failure in children, ACE inhibitors are essential along with other medications including diuretics, digoxin, and beta-blockers (beta-adrenoceptor antagonists).


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Gasto Cardíaco Bajo/tratamiento farmacológico , Adolescente , Inhibidores de la Enzima Convertidora de Angiotensina/efectos adversos , Inhibidores de la Enzima Convertidora de Angiotensina/farmacocinética , Gasto Cardíaco Bajo/fisiopatología , Niño , Preescolar , Enfermedad Crónica , Ensayos Clínicos como Asunto , Hemodinámica/efectos de los fármacos , Humanos , Lactante , Sistema Renina-Angiotensina/efectos de los fármacos
8.
Lancet ; 362(9393): 1366-73, 2003 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-14585638

RESUMEN

BACKGROUND: Del22q11.2 syndrome is the most frequent known chromosomal microdeletion syndrome, with an incidence of 1 in 4000-5000 livebirths. It is characterised by a 3-Mb deletion on chromosome 22q11.2, cardiac abnormalities, T-cell deficits, cleft palate facial anomalies, and hypocalcaemia. At least 30 genes have been mapped to the deleted region. However, the association of these genes with the cause of this syndrome is not clearly understood. METHODS: To test for the chromosomal deletion at 22q11.2, we did fluorescence in-situ hybridisation analysis with ten probes on 22q11.2 in 235 unrelated patients with clinically diagnosed del22q11.2 syndrome. To investigate mutations in the coding sequence of TBX1, we also did genetic analysis in 13 patients from ten families who have the 22q11.2 syndrome phenotype but no detectable deletion of 22q11.2. FINDINGS: 96% (225 of 235) of patients had a defined 1.5-3-Mb deletion at 22q11.2. We identified three mutations of TBX1 in two unrelated patients without the 22q11.2 deletion-one with sporadic conotruncal anomaly face syndrome/velocardiofacial syndrome and one with sporadic DiGeorge's syndrome-and in three patients from a family with conotruncal anomaly face syndrome/velocardiofacial syndrome. We did not record these three mutations in 555 healthy controls (1110 chromosomes; p<0.0001). INTERPRETATION: Our results suggest that the TBX1 mutation is responsible for five major phenotypes in del22q11.2 syndrome. Therefore, we conclude that TBX1 is a major genetic determinant of the del22q11.2 syndrome.


Asunto(s)
Cromosomas Humanos Par 22/genética , Síndrome de DiGeorge/genética , Eliminación de Gen , Proteínas de Dominio T Box/genética , Anomalías Múltiples/genética , Femenino , Cardiopatías Congénitas/genética , Humanos , Hibridación Fluorescente in Situ , Masculino , Mutación , Fenotipo
9.
Prostaglandins Other Lipid Mediat ; 78(1-4): 117-28, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16303610

RESUMEN

Prostaglandin E1 is used to reopen the constricted ductus arteriosus in neonates with ductus-dependent circulation. To clarify possible prostanoid receptor agonists that can reopen the neonatal ductus with fewer side effects, we studied in vivo reopening of the neonatal ductus arteriosus by AE1-329, a prostanoid EP4-receptor agonist, in the rat. Neonatal rats were incubated at 33 degrees C. The inner diameter of the ductus was measured with a microscope and a micrometer following rapid whole-body freezing. Intraesophageal pressure was measured with a Millar micro-tip transducer. The ductus arteriosus constricted quickly after birth, and the inner diameter was 0.80 and 0.08 mm at 0 and 60 min after birth. PGE1 and AE1-329, injected subcutaneously at 60 min after birth, dilated the ductus dose-dependently. Thirty minutes after injection of 10 ng/g of PGE1 and AE1-329, the ductus diameter was 0.52 and 0.65 mm, respectively. The ductus-dilating effect of PGE1 was maximal at 15-30 min, and disappeared at 2 h. The ductus-dilating effect of AE1-329 was prolonged, the ductus was widely open at 6 h, and closed at 12 h after injection of 10 ng/g AE1-329. AE1-259-01 (EP2 agonist) (100 ng/g) did not dilate the neonatal ductus. Respiration was depressed by PGE1, but not by AE1-329. These results indicate the major role of EP4 in the neonatal ductus and that AE1-329, an EP4 agonist, can be used to dilate the neonatal constricted ductus without the side effects shown by EP3, including apnea.


Asunto(s)
Alprostadil/farmacología , Animales Recién Nacidos , Conducto Arterial/efectos de los fármacos , Receptores de Prostaglandina E/agonistas , Animales , Femenino , Embarazo , Ratas , Ratas Wistar , Subtipo EP4 de Receptores de Prostaglandina E
10.
Am J Cardiol ; 91(6): 693-8, 2003 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-12633800

RESUMEN

Little is known about the growth potential of pulmonary stenotic lesions after balloon angioplasty (BA) in patients after the arterial switch operation. The aim of this study was to evaluate the growth potential of pulmonary stenotic lesions after BA and assess the midterm results of BA for pulmonary artery stenosis after the arterial switch operation. Thirty-seven patients who had undergone 52 procedures had repeat catheterization at a median of 43 years (range 1.2 to 9.3 ys) after BA. To adjust growth-related changes in the size of the pulmonary artery, the stenotic diameter was expressed as a percentage of normal (%N). An immediate increase of 63 +/- 45% in the stenotic diameter and a reduction of 51 +/- 33% in the pressure gradient occurred across the stenotic lesions after BA. The right ventricular-aortic systolic pressure ratio decreased from 0.67 +/- 0.24 to 0.51 +/- 0.12 after BA (p <0.0001). Compared with immediate data after BA, there was no significant change in the growth-adjusted diameter of the stenotic lesions (68 +/- 26 %N after BA vs 65 +/- 25 %N at follow-up, p = 0.08), and the pressure gradient (16 +/- 13 mm Hg after BA vs 20 +/- 21 mm Hg at follow-up, p = 0.10). The ventricular-aortic systolic pressure ratio also did not change (0.51 +/- 0.12 after BA vs 0.50 +/- 0.21 at follow-up, p = 0.57). Restenosis occurred in 3 of 26 vessels (12%) after successful BA in which the diameter increased >50% after BA. Our data suggest that pulmonary stenotic lesions after BA develop with age in growing children after the arterial switch operation, and the efficacy of the BA may be long lasting.


Asunto(s)
Angioplastia de Balón/efectos adversos , Complicaciones Posoperatorias , Arteria Pulmonar/crecimiento & desarrollo , Arteria Pulmonar/cirugía , Estenosis de la Válvula Pulmonar/etiología , Estenosis de la Válvula Pulmonar/cirugía , Adolescente , Adulto , Niño , Preescolar , Femenino , Estudios de Seguimiento , Humanos , Lactante , Masculino , Persona de Mediana Edad , Evaluación de Resultado en la Atención de Salud , Arteria Pulmonar/diagnóstico por imagen , Estenosis de la Válvula Pulmonar/diagnóstico por imagen , Radiografía , Recurrencia , Reoperación , Factores de Tiempo , Insuficiencia del Tratamiento
11.
Am J Cardiol ; 105(11): 1617-24, 2010 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-20494672

RESUMEN

Cardiovascular anomalies are present in 80% of neonates with 22q11.2 deletion syndrome. Three genes in chromosome 22q11.2 (TBX1, CRKL, and ERK2) have been identified whose haploinsufficiency causes dysfunction of the neural crest cell and anterior heart field and anomalies of 22q11.2 deletion syndrome. The most common diseases are conotruncal anomalies, which include tetralogy of Fallot (TF), TF with pulmonary atresia, truncus arteriosus, and interrupted aortic arch. A high prevalence of the deletion is noted in patients with TF with absent pulmonary valve, TF associated with pulmonary atresia and major aortopulmonary collateral arteries, truncus arteriosus, and type B interruption of aortic arch. Right aortic arch, aberrant subclavian artery, cervical origin of the subclavian artery, crossing pulmonary arteries, and major aortopulmonary collateral arteries are frequently associated with cardiovascular anomalies associated with 22q11.2 deletion syndrome. Virtually every type of congenital heart defect has been described early in the context of a 22q11.2 deletion. In conclusion, conotruncal anomaly associated with aortic arch and ductus arteriosus anomalies should increase the suspicion of 22q11.2 deletion.


Asunto(s)
Síndrome de DiGeorge/genética , Cardiopatías Congénitas/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Biomarcadores/metabolismo , Humanos , Proteínas Nucleares/genética , Prevalencia , Proteína Proto-Oncogénica c-ets-2/genética , Proteínas de Dominio T Box/genética
12.
Neonatology ; 96(2): 125-31, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19279396

RESUMEN

BACKGROUND: Magnesium sulfate (MgSO4) is used therapeutically for eclampsia and tocolysis. Some reports have suggested a relationship between therapeutic MgSO4 and patent ductus arteriosus (DA) in preterm infants. OBJECTIVES: To clarify patent DA induction by MgSO4 in preterm infants, we studied the increase in serum Mg concentrations and fetal dilatation and postnatal delayed closure of the ductus, using transplacental MgSO4 in rats. METHODS: Fetal and neonatal ductus diameters were measured with a microscope and a micrometer after rapid whole-body freezing. In the postnatal study, 21-day pregnant dams were administered a subcutaneous injection of MgSO4 1-3 h before delivery, and the ductus was studied 0, 15, 30, 60 and 120 min after birth. In the fetal study, MgSO4 (1 g/kg) and indomethacin (10 mg/kg) were simultaneously administered to 21-day dams and the fetal ductus was studied 1, 2 and 4 h later. Serum Mg concentration was measured in the dams and newborns. RESULTS: Neonatal Mg concentrations increased from 3.8 to 4.7 and 5.8 mg/dl at 1 and 3 h after maternal administration of MgSO4. Following MgSO4 administration 3 h before birth, closure of the neonatal DA was delayed. The ductus diameter was 0.88 mm (0.80 mm in control) at 0 min, and 0.26 mm (0.08 mm in the control) at 60 min after birth. In the fetal study, MgSO4 initially reversed and later attenuated the ductus-constricting effect of indomethacin. CONCLUSIONS: Hypermagnesemia induced by transplacental MgSO4 attenuates the fetal ductus-constricting effects of indomethacin, and delays postnatal ductal closure in rats.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Conducto Arterioso Permeable/tratamiento farmacológico , Conducto Arterial/efectos de los fármacos , Indometacina/farmacología , Sulfato de Magnesio/farmacología , Intercambio Materno-Fetal , Tocolíticos/farmacología , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Antagonismo de Drogas , Conducto Arterial/patología , Conducto Arterial/fisiopatología , Conducto Arterioso Permeable/patología , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/embriología , Femenino , Sulfato de Magnesio/farmacocinética , Masculino , Contracción Muscular/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Embarazo , Ratas , Ratas Wistar , Tocolíticos/farmacocinética
13.
Neonatology ; 95(4): 324-31, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19077393

RESUMEN

BACKGROUND: Fasudil hydrochloride, a Rho kinase inhibitor, was reported to dilate the constricted ductus arteriosus in vitro, and was suggested as a neonatal bridge to heart surgery. OBJECTIVES: To clarify the in vivo effectiveness of fasudil to dilate the neonatal ductus arteriosus, and its usefulness as a bridge to heart surgery. METHODS: We studied the dose and timing of administration of fasudil in the neonatal rat. Postnatal ductal closure was studied on the 21st gestational day following cesarean section and incubation for 30-480 min in room air at 33 degrees C, with a rapid whole-body freezing method. In control rats, the ductus closed rapidly after birth, and the ductal diameter was 0.80 and 0.12 mm at 0 and 30 min after birth. Fasudil was injected peritoneally into the neonate, and the ductus was studied 30 min after injection and 60 min after birth. RESULTS: Fasudil, 10 mg/kg, injected within 5 min after birth, dilated the ductus completely to 0.8 mm, and the dose used clinically, 1 mg/kg, dilated the ductus to 0.4 mm. The ductus-dilating effect of fasudil decreased rapidly as the neonatal ductus constricted. Fasudil, 100 mg/kg, dilated the ductus completely, but induced severe respiratory depression and frequent death in 1-hour-old rats. CONCLUSIONS: Fasudil dilates the neonatal ductus arteriosus dose-dependently in the rat. Fasudil in large doses dilates the neonatal ductus completely, but is associated with fatal side effects, including respiratory depression.


Asunto(s)
1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , Animales Recién Nacidos/fisiología , Conducto Arterial/efectos de los fármacos , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología , Quinasas Asociadas a rho/antagonistas & inhibidores , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/efectos adversos , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Animales , Procedimientos Quirúrgicos Cardiovasculares/métodos , Relación Dosis-Respuesta a Droga , Conducto Arterial/fisiología , Femenino , Masculino , Modelos Animales , Embarazo , Ratas , Ratas Wistar , Insuficiencia Respiratoria/inducido químicamente , Vasodilatación/fisiología , Vasodilatadores/efectos adversos
14.
Neonatology ; 96(2): 69-79, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19225238

RESUMEN

BACKGROUND: Indomethacin is used to close the patent ductus arteriosus in premature infants and for tocolysis of preterm labor. Clinically and experimentally, early in utero exposure to indomethacin induces the paradoxical delay of postnatal closure of the ductus arteriosus. OBJECTIVES: To clarify the pharmacological nature of the delay of closure of the ductus arteriosus in the rat. METHODS: We studied early in utero exposure to indomethacin (dose and timing) in addition to other drugs, inducing a delay in postnatal ductal closure. Pregnant rats at near term were studied by cesarean section on gestational day 21 (D21), incubated in room air at 33 degrees C, followed by rapid whole-body freezing. RESULTS: The delay in closure of the ductus arteriosus was dose dependent. A large dose of indomethacin (10 mg/kg) 1 or 2 days before birth induced a delay of 3-4 times. A timing study revealed maximum delay with administration of indomethacin 2 days before birth and minimum delay with administration 5 days before. Aspirin, ibuprofen, the selective COX1 inhibitor SC 560, the selective COX2 inhibitor rofecoxib and a prostaglandin EP(4) receptor blocker, ONO-208, all delayed neonatal ductal closure following maternal administration on D19 and D20. CONCLUSIONS: The delay by indomethacin was dose dependent. The maximum delay was induced by 2 doses of 10 mg/kg indomethacin on D19 and D20. The delay was induced by a decreased stimulus to the prostaglandin EP(4) receptor system in the last 2 days in utero. The delay was temporary with recovery 3 days or more after exposure.


Asunto(s)
Inhibidores de la Ciclooxigenasa/farmacología , Conducto Arterioso Permeable/inducido químicamente , Conducto Arterial/efectos de los fármacos , Indometacina/farmacología , Animales , Animales Recién Nacidos , Antiinflamatorios no Esteroideos/administración & dosificación , Antiinflamatorios no Esteroideos/farmacología , Aspirina/administración & dosificación , Aspirina/farmacología , Inhibidores de la Ciclooxigenasa/administración & dosificación , Inhibidores de la Ciclooxigenasa/sangre , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Conducto Arterial/anomalías , Conducto Arterioso Permeable/embriología , Conducto Arterioso Permeable/patología , Femenino , Feto/anomalías , Feto/efectos de los fármacos , Edad Gestacional , Ibuprofeno/administración & dosificación , Ibuprofeno/farmacología , Indometacina/administración & dosificación , Indometacina/sangre , Masculino , Exposición Materna , Embarazo , Pirazoles/administración & dosificación , Pirazoles/farmacología , Ratas , Ratas Wistar
15.
Neonatology ; 92(2): 139-44, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17409775

RESUMEN

BACKGROUND: Alpha-human atrial natriuretic peptide (hANP) reportedly increases in premature infants with patent ductus arteriosus (PDA). OBJECTIVES: To clarify a possible hANP effect to reopen the postnatal ductus, we studied in vivo reopening of the postnatal DA by a recombinant hANP, carperitide, in rats. METHODS: Near-term rat pups were incubated at 33 degrees C following caesarean section. The inner diameter of the ductus was measured with a microscope and a micrometer following rapid whole-body freezing. The DA constricted quickly after birth, and the inner diameter was 0.80 and 0.08 mm at 0 min (fetal state) and 60 min after birth. hANP concentration in the pup blood and the ductus-dilating effect of hANP were studied by subcutaneous injection of hANP at 60 min after birth, and by measurement 7, 15, 30 and 60 min later. RESULTS: The peak hANP concentration was 790 pg/ml at 7 min with 1 mg/kg, which is similar to the level seen in preterm infants with symptomatic PDA. hANP dilated the postnatal ductus dose dependently and maximally at 7 min after injection. hANP dilated the postnatal constricted ductus completely to 0.79 mm in diameter with a large dose (10 mg/kg) and to 0.55 mm with 1 mg/kg. CONCLUSIONS: hANP reopens the constricted postnatal DA dose dependently in rats. The increased hANP, accompanying premature PDA, may delay closure of the DA.


Asunto(s)
Factor Natriurético Atrial/farmacología , Dilatación , Conducto Arterioso Permeable , Conducto Arterial/efectos de los fármacos , Animales , Animales Recién Nacidos , Factor Natriurético Atrial/sangre , Criopreservación , Relación Dosis-Respuesta a Droga , Conducto Arterial/embriología , Conducto Arterial/patología , Conducto Arterioso Permeable/patología , Conducto Arterioso Permeable/fisiopatología , Secciones por Congelación , Inyecciones Subcutáneas , Modelos Animales , Ratas , Ratas Wistar , Proteínas Recombinantes
16.
Biol Neonate ; 89(1): 42-9, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16155385

RESUMEN

BACKGROUND: The closing mechanisms of the ductus venosus (DV) have not yet been revealed. OBJECTIVES: The aims of this study were to document the perinatal closing process of the DV, to study the suppression of prostaglandins by indomethacin, and to determine the effects of umbilical blood flow to the fetal DV. METHODS: The proximal and distal DV diameters were studied in near-term fetal and neonatal rats with the rapid whole-body freezing method. RESULTS: The DV diameter changed sensitively at birth, and decreased by 10% immediately after the cessation of the umbilical circulation. Umbilical hemorrhage caused an additional decrease in the DV diameter compared with neonate without the hemorrhage. The neonates showed a DV diameter decreased by 20% at 30 min and 30% at 60 min after birth. The fetal DV was tubular, and the neonatal DV was horn-shaped with a smaller inlet than outlet. A small dose (0.1 mg/kg) of indomethacin administered to the pregnant rats induced a reduction in prostaglandins and decreased the fetal DV diameter to 80% of the control. Indomethacin at a large dose (10 mg/kg), administered to the dams, induced a reduction in prostaglandins, severe constriction of the ductus arteriosus, and decreased blood flow through the descending aorta umbilicus, and caused a further reduction in the DV diameter to 70-80% of the control. A large dose of nifedipine (10 mg/kg), which causes cardiac suppression and heart failure in the fetus, was administered to near-term rats to study the effect of decreased fetal cardiac output and blood flow passing through the DV. Nifedipine induced a 20% decrease in the DV diameter for 2-8 h. In all 1-hour-old neonates with or without pretreatment, the inlet diameter of the DV was reduced more than the outlet diameter, and the DV showed a horn-shaped morphology. CONCLUSION: In conclusion, perinatal cessation of the umbilical circulation and umbilical hemorrhage are associated with an immediate decrease in DV diameter. The DV diameter is also reduced in other conditions associated with decreased umbilical blood flow, such as induced by nifedipine which leads to heart failure and constricting of the ductus arteriosus induced by indomethacin. The constricting effect of a small dose of indomethacin suggests that prostaglandins dilate the DV physiologically.


Asunto(s)
Feto/irrigación sanguínea , Hemorragia/patología , Prostaglandinas/sangre , Cordón Umbilical/fisiopatología , Venas/embriología , Animales , Velocidad del Flujo Sanguíneo , Femenino , Embarazo , Ratas , Ratas Wistar , Venas/patología
17.
Biol Neonate ; 89(4): 251-6, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16319452

RESUMEN

BACKGROUND: Clinically, it appears that phosphodiesterase 3 (PDE 3) inhibitors, which are used for acute cardiac failure in premature infants, dilate the ductus arteriosus (DA). OBJECTIVES: To clarify the ductus-dilating effects of PDE 3 inhibitors in near-term rat pups and their differential effects in near-term and preterm fetal rats, in in vivo studies. METHODS: The in vivo ductal diameter of rat pups and fetuses was measured using a rapid whole-body freezing method, by cutting on a freezing microtome and measuring with a microscope and micrometer. Eight to twenty pups and fetuses were studied in each group. Milrinone and amrinone (specific inhibitors of PDE 3) were injected into 1-hour-old pups and the DA was studied 0.5 and 1 h later. The differential effects of these PDE 3 inhibitors on the near-term and preterm ductus were studied by injecting indomethacin (10 mg/kg) and PDE 3 inhibitors into 21D (21st day of pregnancy: term-21.5 days) and 19D dams and studying the fetal ductus 4 and 8 h later. RESULTS: Milrinone and amrinone dilated the postnatal ductus dose-dependently. Large doses of these drugs dilated it completely, and clinically equivalent doses dilated it minimally. Milrinone and amrinone prevented constriction of the fetal ductus by indomethacin. Their ductus-dilating effects were more potent in the preterm than in the near-term fetuses, and clinically equivalent doses of these PDE 3 inhibitors dilated preterm ductus completely. CONCLUSION: In rats, PDE 3 inhibitors reopen the constricted postnatal DA slightly. PDE 3 inhibitors dilate the fetal DA constricted with indomethacin effectively and more sensitively in preterm than in near-term fetuses.


Asunto(s)
3',5'-AMP Cíclico Fosfodiesterasas/antagonistas & inhibidores , Conducto Arterial/efectos de los fármacos , Conducto Arterial/enzimología , Inhibidores de Proteínas Quinasas/farmacología , 3',5'-AMP Cíclico Fosfodiesterasas/metabolismo , Animales , Animales Recién Nacidos , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3 , Conducto Arterial/embriología , Conducto Arterial/crecimiento & desarrollo , Conducto Arterioso Permeable/tratamiento farmacológico , Femenino , Indometacina/farmacología , Milrinona/farmacología , Embarazo , Inhibidores de Proteínas Quinasas/uso terapéutico , Ratas , Ratas Wistar
18.
Circ J ; 69(3): 354-8, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15731544

RESUMEN

BACKGROUND: To find a better treatment for patent ductus arteriosus (PDA) in premature infants, the present study investigated the synergism of clinical doses of dexamethasone, indomethacin, and rofecoxib, a selective cyclooxygenase-2 (COX-2) inhibitor, on the fetal ductus arteriosus (DA) in rats. METHODS AND RESULTS: Dexamethasone (0.3 mg/kg), indomethacin (0.3 mg/kg), and rofecoxib (0.3 mg/kg), alone or in combination, were administered to preterm (d19) and near-term (d21) fetal rats. The ratio of the inner diameter of the DA to that of the main pulmonary artery (PA) (DA/PA) was studied at 2, 4 and 8 h after drug administration, using a rapid whole-body freezing method. In near-term rats, the combined administration of dexamethasone and indomethacin caused severe constriction, with a DA/PA ratio of 0.52+/-0.08 at 8 h, whereas the DA/PA ratios were 0.83+/-0.03 and 0.90+/-0.02 with dexamethasone and indomethacin, respectively. Combined administration of dexamethasone and rofecoxib also caused severe constriction, with a DA/PA ratio of 0.64+/-0.07 at 8 h, compared with the DA/PA ratio of 0.92+/-0.03 with rofecoxib alone. CONCLUSIONS: Combined therapy may be an option in the medical management of PDA in premature infants before considering surgical treatment.


Asunto(s)
Antiinflamatorios/administración & dosificación , Conducto Arterial/efectos de los fármacos , Feto/efectos de los fármacos , Vasoconstricción/efectos de los fármacos , Animales , Dexametasona/administración & dosificación , Evaluación Preclínica de Medicamentos , Quimioterapia Combinada , Conducto Arterial/fisiología , Conducto Arterioso Permeable/tratamiento farmacológico , Feto/irrigación sanguínea , Indometacina/administración & dosificación , Lactonas/administración & dosificación , Modelos Animales , Ratas , Ratas Wistar , Sulfonas/administración & dosificación
19.
Pediatr Res ; 58(1): 42-5, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15774847

RESUMEN

A recent in vitro study showed that sildenafil, a type 5 phosphodiesterase inhibitor, dilated the constricted ductus arteriosus of neonatal rabbits. We studied the in vivo ductus-dilating effects of sildenafil in fetal and neonatal rats. Ductus diameters were measured with whole-body freezing and cutting on a freezing microtome. Indomethacin (10 mg/kg) constricted the fetal ductus severely at 4 and 8 h after orogastric administration to the dams. Sildenafil, administered orogastrically and simultaneously with indomethacin, dilated the near-term fetal [21 fetal days (FD)] ductus constricted by indomethacin completely with 1 mg/kg at 8 h after administration. The preterm fetal ductus was more sensitive to sildenafil at 19FD. The ductus constricted rapidly after birth, and the ductal diameter was only 10% of the fetal diameter at 1 h after birth. The ductus-dilating effect of sildenafil was studied by i.p. injection at 1 h after birth, and the ductus diameter was studied 30 and 60 min later. Sildenafil dilated the neonatal constricted ductus moderately with a massive dose (100 mg/kg) and only minimally with 1 mg/kg. In conclusion, sildenafil, a type 5 phosphodiesterase inhibitor, dilated the constricted fetal ductus completely at 8 h with 1 mg/kg in the near-term fetus and completely with a smaller dose (0.1 mg/kg) in the preterm fetus. However, sildenafil dilated the neonatal constricted ductus only moderately with large doses and minimally with 1 mg/kg. Probably, sildenafil is useful clinically for treating idiopathic and secondary fetal ductal constriction and not useful for dilation of the neonatal constricted ductus.


Asunto(s)
Conducto Arterial/efectos de los fármacos , Conducto Arterial/patología , Inhibidores de Fosfodiesterasa/farmacología , Hidrolasas Diéster Fosfóricas/metabolismo , 3',5'-GMP Cíclico Fosfodiesterasas , Animales , Antiinflamatorios no Esteroideos/farmacología , Peso Corporal , GMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5 , Congelación , Indometacina/farmacología , Piperazinas/farmacología , Prostaglandinas E/metabolismo , Purinas , Ratas , Ratas Wistar , Citrato de Sildenafil , Sulfonas , Factores de Tiempo
20.
Pediatr Res ; 58(5): 971-5, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16257930

RESUMEN

Indomethacin is used to constrict the patent ductus arteriosus in premature infants. To clarify possible prostanoid receptor antagonists that can constrict the ductus, we studied in vivo constriction of the fetal and neonatal ductus arteriosus by AE3-208, a prostanoid EP4-receptor antagonist, in rats. Following quick cesarean section of near-term pregnant rats (21 d), neonates were incubated in room air at 33 degrees C. The inner diameter of the ductus was measured with a microscope and a micrometer following rapid whole-body freezing of the fetus and neonate, and sectioning of the thorax in the frontal plane on a freezing microtome. In the control, the ductus arteriosus constricted quickly after birth, and the inner diameter was 0.80 mm in the fetus and 0.06 mm at 90 min after birth. AE3-208, administered orogastrically to the dam, constricted the fetal ductus dose dependently. Maximal ductal constriction was observed 4 h after administration, and the ductal diameters were 0.06 mm and 0.26 mm after administration of 10 mg/kg and 10 ng/kg of AE3-208, respectively. In neonatal rats, AE3-208 injected subcutaneously at 30 min after birth, inhibited dilatation of the ductus by PGE1 dose dependently. PGE1 (10 microg/kg) was injected subcutaneously to the 1-h-old neonatal rat, and the ductal diameters were 0.53 mm and 0.19 mm without and with pretreatment of AE3-208 (10 microg/kg), respectively. These results indicate the major role of EP4 in the fetal and neonatal ductus and show that an EP4 antagonist can be used to constrict the patent ductus of premature infants.


Asunto(s)
Conducto Arterial/efectos de los fármacos , Feto/efectos de los fármacos , Naftalenos/farmacología , Fenilbutiratos/farmacología , Receptores de Prostaglandina E/antagonistas & inhibidores , Vasoconstricción/efectos de los fármacos , Animales , Animales Recién Nacidos , Conducto Arterial/fisiopatología , Feto/fisiopatología , Ratas , Subtipo EP4 de Receptores de Prostaglandina E
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA