Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
1.
Pediatr Res ; 84(3): 458-465, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29976969

RESUMEN

BACKGROUND: Although studies involving preterm infants ≤34 weeks gestation report a decreased incidence of patent ductus arteriosus after antenatal betamethasone, studies involving younger gestation infants report conflicting results. METHODS: We used preterm baboons, mice, and humans (≤276/7 weeks gestation) to examine betamethasone's effects on ductus gene expression and constriction both in vitro and in vivo. RESULTS: In mice, betamethasone increased the sensitivity of the premature ductus to the contractile effects of oxygen without altering the effects of other contractile or vasodilatory stimuli. Betamethasone's effects on oxygen sensitivity could be eliminated by inhibiting endogenous prostaglandin/nitric oxide signaling. In mice and baboons, betamethasone increased the expression of several developmentally regulated genes that mediate oxygen-induced constriction (K+ channels) and inhibit vasodilator signaling (phosphodiesterases). In human infants, betamethasone increased the rate of ductus constriction at all gestational ages. However, in infants born ≤256/7 weeks gestation, betamethasone's contractile effects were only apparent when prostaglandin signaling was inhibited, whereas at 26-27 weeks gestation, betamethasone's contractile effects were apparent even in the absence of prostaglandin inhibitors. CONCLUSIONS: We speculate that betamethasone's contractile effects may be mediated through genes that are developmentally regulated. This could explain why betamethasone's effects vary according to the infant's developmental age at birth.


Asunto(s)
Betametasona/uso terapéutico , Conducto Arterioso Permeable/tratamiento farmacológico , Conducto Arterial/efectos de los fármacos , Animales , Ecocardiografía , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Recien Nacido Prematuro , Exposición Materna , Ratones , Oxígeno/metabolismo , Papio , Reacción en Cadena de la Polimerasa , Prostaglandinas/metabolismo
2.
Am J Physiol Heart Circ Physiol ; 307(5): H732-40, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24993047

RESUMEN

Sepsis is strongly associated with patency of the ductus arteriosus (PDA) in critically ill newborns. Inflammation and the aminoglycoside antibiotics used to treat neonatal sepsis cause smooth muscle relaxation, but their contribution to PDA is unknown. We examined whether: 1) lipopolysaccharide (LPS) or inflammatory cytokines cause relaxation of the ex vivo mouse DA; 2) the aminoglycosides gentamicin, tobramycin, or amikacin causes DA relaxation; and 3) newborn infants treated with aminoglycosides have an increased risk of symptomatic PDA (sPDA). Changes in fetal mouse DA tone were measured by pressure myography in response to LPS, TNF-α, IFN-γ, macrophage-inflammatory protein 2, IL-15, IL-13, CXC chemokine ligand 12, or three aminoglycosides. A clinical database of inborn patients of all gestations was analyzed for association between sPDA and aminoglycoside treatment. Contrary to expectation, neither LPS nor any of the inflammatory mediators caused DA relaxation. However, each of the aminoglycosides caused concentration-dependent vasodilation in term and preterm mouse DAs. Pretreatment with indomethacin and N-(G)-nitro-L-arginine methyl ester did not prevent gentamicin-induced DA relaxation. Gentamicin-exposed DAs developed less oxygen-induced constriction than unexposed DAs. Among 488,349 infants who met the study criteria, 40,472 (8.3%) had sPDA. Confounder-adjusted odds of sPDA were higher in gentamicin-exposed infants, <25 wk and >32 wk. Together, these findings suggest that factors other than inflammation contribute to PDA. Aminoglycoside-induced vasorelaxation and inhibition of oxygen-induced DA constriction support the paradox that antibiotic treatment of sepsis may contribute to DA relaxation. This association was also found in newborn infants, suggesting that antibiotic selection may be an important consideration in efforts to reduce sepsis-associated PDA.


Asunto(s)
Conducto Arterioso Permeable/fisiopatología , Conducto Arterial/efectos de los fármacos , Gentamicinas/farmacología , Sepsis/complicaciones , Vasodilatación , Animales , Quimiocina CXCL12/farmacología , Conducto Arterial/fisiopatología , Conducto Arterioso Permeable/etiología , Humanos , Técnicas In Vitro , Indometacina/farmacología , Recién Nacido , Interferón gamma/farmacología , Interleucinas/farmacología , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , NG-Nitroarginina Metil Éster/farmacología , Factor de Necrosis Tumoral alfa/farmacología
3.
J Mol Cell Cardiol ; 59: 86-94, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23454087

RESUMEN

Persistent patency of the ductus arteriosus (PDA) is a common problem in preterm infants. The antacid cimetidine is a potent antagonist of the H2 histamine receptor but it also inhibits certain cytochrome P450 enzymes (CYPs), which may affect DA patency. We examined whether cimetidine contributes to PDA and is mediated by CYP inhibition rather than H2 blockade. Analysis of a clinical trial to prevent lung injury in premature infants revealed a significant association between cimetidine treatment and PDA. Cimetidine and ranitidine, both CYP inhibitors as well as H2 blockers, caused relaxation of the term and preterm mouse DA. CYP enzymes that are inhibited by cimetidine were expressed in DA subendothelial smooth muscle. The selective CYP3A inhibitor ketoconazole induced greater DA relaxation than cimetidine, whereas famotidine and other H2 antagonists with less CYP inhibitory effects caused less dilation. Histamine receptors were developmentally regulated and localized in DA smooth muscle. However, cimetidine caused DA relaxation in histamine-deficient mice, consistent with CYP inhibition, not H2 antagonism, as the mechanism for PDA. Oxygen-induced DA constriction was inhibited by both cimetidine and famotidine. These studies show that antacids and other compounds with CYP inhibitory properties pose a significant and previously unrecognized risk for PDA in critically ill newborn infants.


Asunto(s)
Cimetidina/efectos adversos , Sistema Enzimático del Citocromo P-450/metabolismo , Conducto Arterioso Permeable/inducido químicamente , Conducto Arterioso Permeable/metabolismo , Antagonistas de los Receptores H2 de la Histamina/efectos adversos , Humanos , Inmunohistoquímica , Recién Nacido , Cetoconazol/efectos adversos , Reacción en Cadena de la Polimerasa , Ensayos Clínicos Controlados Aleatorios como Asunto , Ranitidina/efectos adversos , Receptores Histamínicos/metabolismo , Estudios Retrospectivos
4.
Pediatr Res ; 72(2): 122-8, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22565502

RESUMEN

BACKGROUND: Increased oxygen tension at birth regulates physiologic events that are essential to postnatal survival, but the accompanying oxidative stress may also generate isoprostanes. We hypothesized that isoprostanes regulate ductus arteriosus (DA) function during postnatal vascular transition. METHODS: Isoprostanes were measured by gas chromatography-mass spectrometry. DA tone was assessed by pressure myography. Gene expression was measured by quantitative PCR. RESULTS: Oxygen exposure was associated with increased 8-iso-prostaglandin (PG)F2α in newborn mouse lungs. Both 8-iso-PGE2 and 8-iso-PGF2α induced concentration-dependent constriction of the isolated term DA, which was reversed by the thromboxane A2 (TxA2) receptor antagonist SQ29548. SQ29548 pretreatment unmasked an isoprostane-induced DA dilation mediated by the EP4 PG receptor. Exposure of the preterm DA to 8-iso-PGE2 caused unexpected DA relaxation that was reversed by EP4 antagonism. In contrast, exposure to 8-iso-PGF2α caused preterm DA constriction via TxA2 receptor activation. Further investigation revealed the predominance of the TxA2 receptor at term, whereas the EP4 receptor was expressed and functionally active from mid-gestation onward. CONCLUSION: This study identifies a novel physiological role for isoprostanes during postnatal vascular transition and provide evidence that oxidative stress may act on membrane lipids to produce vasoactive mediators that stimulate physiological DA closure at birth or induce pathological patency of the preterm DA.


Asunto(s)
Conducto Arterioso Permeable/metabolismo , Conducto Arterial/crecimiento & desarrollo , Isoprostanos/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Vasodilatación/efectos de los fármacos , Análisis de Varianza , Animales , Animales Recién Nacidos , Compuestos Bicíclicos Heterocíclicos con Puentes , Dinoprost/análogos & derivados , Dinoprost/metabolismo , Dinoprostona/análogos & derivados , Dinoprostona/metabolismo , Conducto Arterial/efectos de los fármacos , Conducto Arterioso Permeable/fisiopatología , Ácidos Grasos Insaturados , Femenino , Cromatografía de Gases y Espectrometría de Masas , Perfilación de la Expresión Génica , Hidrazinas/farmacología , Isoprostanos/farmacología , Ratones , Miografía , Estrés Oxidativo/fisiología , Oxígeno/análisis , Embarazo , Nacimiento Prematuro/metabolismo , Receptores de Tromboxano A2 y Prostaglandina H2/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA