Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 413
Filtrar
1.
Ultrasound Obstet Gynecol ; 58(6): 940-942, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34453368

RESUMO

We report on a fetal case of Ebstein's anomaly with severe tricuspid regurgitation, functional pulmonary atresia and progressive circular shunting (CS) across a widely patent ductus arteriosus (DA) and regurgitant pulmonary valve, contributing to significant systemic hypoperfusion. To mitigate the extent of CS and allow the pregnancy to continue, maternal non-steroidal anti-inflammatory drug (NSAID) therapy with indomethacin was started at 33 + 5 weeks to induce DA constriction. Rather than achieving the desired narrowing of the DA, the treatment led to its complete closure and only minimal antegrade flow across the pulmonary valve. While closure of the DA resulted in the anticipated improvement in fetal hemodynamics, at birth, the child was at risk of severe hypoxemia and its consequences due to the lack of adequate pulmonary perfusion. Reduction and eventual discontinuation of the NSAID treatment did not result in DA reopening. Our experience illustrates the risk of unintended irreversible DA closure when NSAIDs are used to treat CS. © 2021 International Society of Ultrasound in Obstetrics and Gynecology.


Assuntos
Anti-Inflamatórios não Esteroides/administração & dosagem , Permeabilidade do Canal Arterial/tratamento farmacológico , Canal Arterial/efeitos dos fármacos , Anomalia de Ebstein/tratamento farmacológico , Indometacina/administração & dosagem , Administração Oral , Administração Retal , Permeabilidade do Canal Arterial/embriologia , Anomalia de Ebstein/embriologia , Anomalia de Ebstein/patologia , Feminino , Humanos , Troca Materno-Fetal , Ilustração Médica , Gravidez , Atresia Pulmonar/tratamento farmacológico , Atresia Pulmonar/embriologia , Insuficiência da Valva Pulmonar/tratamento farmacológico , Insuficiência da Valva Pulmonar/embriologia , Insuficiência da Valva Tricúspide/tratamento farmacológico , Insuficiência da Valva Tricúspide/embriologia
3.
Ultrasound Obstet Gynecol ; 58(3): 420-427, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-33502049

RESUMO

OBJECTIVES: To test the hypotheses that estimated mean pulmonary arterial pressure (MPAP) decreases and pulmonary vascular maturation, assessed by the ratio of pulmonary arterial flow acceleration time to ejection time (AT/ET ratio), increases after reversal of fetal ductus arteriosus constriction by reducing maternal intake of the causal agent (prostaglandin inhibitors, such as polyphenol-rich foods or non-steroidal anti-inflammatory drugs), and that these effects are independent of gestational age, which are inferences not yet demonstrated in the clinical setting. METHODS: This was a prospective cohort study comparing Doppler echocardiographic ductal flow dynamics, MPAP and pulmonary arterial flow AT/ET ratio in third-trimester fetuses (≥ 28 weeks' gestation) with ductus arteriosus constriction, at the time of diagnosis and after 2 weeks of reduced maternal intake of prostaglandin inhibitors either by suspending the use of pharmacological agents with potential for prostaglandin inhibition or by restricting the consumption of polyphenol-rich foods. MPAP was estimated using the Dabestani equation (MPAP = 90 - (0.62 × AT)), and pulmonary vascular maturity was assessed using the AT/ET ratio, according to reported validation studies. Student's t-test was used for comparison of variables at diagnosis with those after reversal of ductal constriction. Change in MPAP and pulmonary AT/ET ratio between the two assessments was compared with the expected change in the same gestational period in normal fetuses based on reference curves of MPAP and pulmonary AT/ET ratio constructed in normal fetuses from healthy pregnant women at 19-37 weeks' gestation, encompassing the same gestational age range as the study group (28-37 weeks). RESULTS: Seventy pregnancies with fetal ductus arteriosus constriction were included in the study. After 2 weeks of reduced maternal intake of prostaglandin inhibitors, normalization of mean systolic (change from 1.86 ± 0.34 m/s at diagnosis to 1.38 ± 0.41 m/s; P < 0.001) and diastolic (change from 0.41 ± 0.11 m/s to 0.21 ± 0.065 m/s; P < 0.001) ductal velocities and of mean pulsatility index (change from 1.99 ± 0.20 to 2.55 ± 0.42; P < 0.001) was demonstrated. MPAP decreased between the assessments (change from 66.7 ± 6.90 mmHg at diagnosis to 54.5 ± 6.70 mmHg after 2 weeks; P < 0.001) and mean pulmonary AT/ET ratio increased (change from 0.20 ± 0.06 to 0.33 ± 0.07; P < 0.001). Change in MPAP between diagnosis and after 2 weeks of reduced maternal intake of prostaglandin inhibitors was -12.2 ± 0.30 mmHg, which was 5.3-times higher than that in 305 normal fetuses over 2 weeks during the same gestational period (-2.3 ± 0.19 mmHg) (P < 0.001), and change in pulmonary AT/ET ratio between the two assessments was 0.13 ± 0.08, which was 8.7-times higher than that in normal fetuses in the same gestational period (0.015 ± 0.08) (P < 0.001). CONCLUSIONS: Resolution of fetal ductal constriction is followed by a fall in MPAP and by an increase in pulmonary vascular maturity, to a significantly greater degree than is observed in normal fetuses in the same gestational-age period. © 2021 International Society of Ultrasound in Obstetrics and Gynecology.


Assuntos
Canal Arterial/patologia , Feto/irrigação sanguínea , Hipertensão Pulmonar/embriologia , Cuidado Pré-Natal/métodos , Adulto , Pressão Arterial , Velocidade do Fluxo Sanguíneo , Constrição Patológica/induzido quimicamente , Constrição Patológica/embriologia , Canal Arterial/efeitos dos fármacos , Canal Arterial/embriologia , Ecocardiografia Doppler , Feminino , Desenvolvimento Fetal/efeitos dos fármacos , Feto/embriologia , Idade Gestacional , Humanos , Hipertensão Pulmonar/etiologia , Polifenóis/efeitos adversos , Gravidez , Estudos Prospectivos , Antagonistas de Prostaglandina/efeitos adversos , Artéria Pulmonar/embriologia , Artéria Pulmonar/crescimento & desenvolvimento , Artéria Pulmonar/fisiopatologia , Fluxo Pulsátil , Volume Sistólico , Ultrassonografia Pré-Natal
4.
Eur J Clin Pharmacol ; 77(7): 1019-1028, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33410971

RESUMO

PURPOSE: Acetaminophen (APAP) is available over-the-counter and widely regarded as safe for use in pregnancy. APAP has been used to close a persistently patent ductus arteriosus. Fetal constriction/closure of the ductus arteriosus (FCCDA), of public health interest given the drug's widespread use during pregnancy, is being monitored globally, including by the European Medicines Agency Pharmacovigilance Risk Assessment Committee. Our objective was to share a comprehensive signal evaluation of FCCDA with in utero APAP exposure to determine if the totality of evidence is sufficiently more consistent with one of the following two possibilities: (1) APAP never contributes to FCCDA (null hypothesis or HO) versus (2) APAP may in some cases be at least a contributory cause of in utero DA narrowing (alternative hypothesis or HA) to justify risk communication. METHODS: To assess the relative support for HO versus HA, we synthesize and interpret within an Austin Bradford Hill criteria framework a comprehensive, cross-disciplinary set of published information and de novo analysis, including toxicology, epidemiology, clinical pharmacology, and clinical and quantitative pharmacovigilance analysis of spontaneous reports. RESULTS: While residual uncertainty remains, the totality of information is more compatible with HA than H0, to the extent that it is reasonably possible that APAP may sometimes be at least a contributory cause of FCCDA. CONCLUSION: It is reasonably possible that APAP may sometimes be at least a contributory cause of FCCDA, and this should therefore be communicated to stakeholders. TRIAL REGISTRATION: CLINICALTRIALS. GOV REGISTRATION: NOT APPLICABLE.


Assuntos
Acetaminofen/efeitos adversos , Analgésicos não Narcóticos/efeitos adversos , Canal Arterial/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Constrição , Feminino , Humanos , Gravidez
5.
Cells ; 9(12)2020 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-33297453

RESUMO

ß3-adrenoreceptor (ß3-AR), a G-protein coupled receptor, has peculiar regulatory properties in response to oxygen and widespread localization. ß3-AR is expressed in the most frequent neoplasms, also occurring in pregnant women, and its blockade reduces tumor growth, indicating ß3-AR-blockers as a promising alternative to antineoplastic drugs during pregnancy. However, ß3-AR involvement in prenatal morphogenesis and the consequences of its blockade for the fetus remain unknown. In this study, after the demonstrated expression of ß3-AR in endothelial and smooth muscle cells of ductus arteriosus (DA), C57BL/6 pregnant mice were acutely treated at 18.5 of gestational day (GD) with indomethacin or with the selective ß3-AR antagonist SR59230A, or chronically exposed to SR59230A from 15.5 to 18.5 GD. Six hours after the last treatment, fetuses were collected. Furthermore, newborn mice were treated straight after birth with BRL37344, a ß3-AR agonist, and sacrificed after 7 h. SR59230A, at the doses demonstrated effective in reducing cancer progression (10 and 20 mg/kg) in acute and chronic mode, did not induce fetal DA constriction and did not impair the DA ability to close after birth, whereas at the highest dose (40 mg/kg), it was shown to cause DA constriction and preterm-delivery. BRL37344 administered immediately after birth did not alter the physiological DA closure.


Assuntos
Antagonistas de Receptores Adrenérgicos beta 3/farmacologia , Permeabilidade do Canal Arterial/metabolismo , Canal Arterial/metabolismo , Receptores Adrenérgicos beta 3/metabolismo , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Progressão da Doença , Canal Arterial/efeitos dos fármacos , Permeabilidade do Canal Arterial/tratamento farmacológico , Etanolaminas/farmacologia , Feminino , Indometacina/farmacologia , Masculino , Exposição Materna , Camundongos , Camundongos Endogâmicos C57BL , Gravidez , Prenhez , Propanolaminas/farmacologia , Receptores Acoplados a Proteínas G/metabolismo , Fatores de Tempo
6.
Early Hum Dev ; 145: 105042, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32442828

RESUMO

BACKGROUND: We previously reported in a randomised trial that early intravenous paracetamol accelerated contraction of ductus arteriosus in very preterm infants (<32 gestation weeks). AIMS: To monitor sequentially paracetamol effects on the blood pressure and brain tissue oxygenation in the infants participating the trial. METHODS: In a double-blind trial, intravenous paracetamol or placebo was infused to 48 very premature infants starting within 24 h of birth for four days. Besides the ductus arteriosus, we systematically measured blood pressure, peripheral (spO2) and regional cerebral oxygen saturation (rcSO2), and cerebral fractional tissue oxygen extraction (cFTOE) during the study period. RESULTS: Compared to the placebo, the paracetamol loading dose transiently decreased the arterial blood pressure. During treatment, the paracetamol-treated infants had higher spO2 (p = .042) and rcSO2 (p = .036) values than the placebo group infants. Additionally, the cFTOE values were lower in the paracetamol group during the study without statistical significance. All infants with closed ductus had higher tissue oxygenation and a lower cFTOE than infants with open ductus. CONCLUSIONS: Paracetamol caused modest haemodynamic effects and increased cerebral oxygenation. They were mostly due to early contraction of ductus. Additional direct drug-effects in brain are not ruled-out.


Assuntos
Acetaminofen/efeitos adversos , Analgésicos não Narcóticos/efeitos adversos , Pressão Sanguínea/efeitos dos fármacos , Recém-Nascido Prematuro , Consumo de Oxigênio/efeitos dos fármacos , Acetaminofen/administração & dosagem , Acetaminofen/farmacologia , Analgésicos não Narcóticos/administração & dosagem , Analgésicos não Narcóticos/farmacologia , Encéfalo/efeitos dos fármacos , Circulação Cerebrovascular , Canal Arterial/efeitos dos fármacos , Feminino , Humanos , Recém-Nascido , Injeções Intravenosas , Masculino , Oxigênio/sangue
7.
Ann Thorac Surg ; 110(6): 2088-2095, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32246933

RESUMO

BACKGROUND: To improve survival of patients with hypoplastic left heart syndrome, combination therapy with bilateral pulmonary artery banding and prostaglandin E1 (PGE1)-mediated ductal patency was developed as an alternative for high-risk neonates in Japan. However, the effect of long-term PGE1 administration on ductus arteriosus remains unclear. Synchrotron radiation-based X-ray phase-contrast tomography (XPCT) enables clear visualization of soft tissues at an approximate spatial resolution of 12.5 µm. We aimed to investigate morphologic changes in ductus arteriosus after long-term PGE1 infusion using XPCT. METHODS: Seventeen ductus arteriosus tissue samples from patients with hypoplastic left heart syndrome were obtained during the Norwood procedure. The median duration of lipo-prostaglandin E1 (lipo-PGE1) administration was 48 days (range, 3 to 123). Structural analysis of ductus arteriosus was performed and compared with conventional histologic analysis. RESULTS: The XPCT was successfully applied to quantitative measurements of ductal media. Significant correlation was found between the duration of lipo-PGE1 infusion and mass density of ductal media (R = 0.723, P = .001). The duration of lipo-PGE1 administration was positively correlated with elastic fiber staining (R = 0.799, P < .001) and negatively correlated with smooth muscle formation (R = -0.83, P < .001). No significant increase in intimal cushion formation was found after long-term lipo-PGE1 administration. Expression of ductus arteriosus dominant PGE2-receptor EP4 almost disappeared in specimens when lipo-PGE1 was administered over 3 days. CONCLUSIONS: Disorganized elastogenesis and little intimal cushion formation after long-term lipo-PGE1 administration suggest that ductus arteriosus remodeled to the elastic artery phenotype. Because EP4 was downregulated and ductus arteriosus exhibited elastic characteristics, the dosage of lipo-PGE1 might be decreased after a definite administration period.


Assuntos
Alprostadil/administração & dosagem , Canal Arterial/efeitos dos fármacos , Síndrome do Coração Esquerdo Hipoplásico/terapia , Vasodilatadores/administração & dosagem , Estudos de Coortes , Esquema de Medicação , Canal Arterial/diagnóstico por imagem , Elasticidade , Feminino , Humanos , Síndrome do Coração Esquerdo Hipoplásico/diagnóstico por imagem , Recém-Nascido , Masculino , Tomografia Computadorizada por Raios X
8.
J Physiol Pharmacol ; 71(5)2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33475092

RESUMO

Prenatal treatment with magnesium sulfate (MgSO4) has neuroprotective effects in very preterm infants but its use has been associated with an increased rate of patent ductus arteriosus (DA). MgSO4 is a vasodilator and thus may exert a direct relaxant effect in the DA. We aimed to investigate the vasoactive effects of MgSO4 in the DA using the chicken embryo as experimental model. DA rings from 15-d (E15), 17-d (E17) and 19-d (E19) chicken embryos (total incubation: 21-d) were mounted in a wire myograph for isometric tension recordings. Exposure of DA rings to 21% O2 induced a tonic contraction which was relaxed by MgSO4 (2.4 - 7.2 mmol L-1) in a concentration-dependent manner (mean maximal relaxation E19: 51.4%, SE 6.3; EC50: 3.5 mmol L-1, SE 0.7). The relaxation evoked by MgSO4 was not significantly different between E15, E17 and E19 DA and was not affected by removal of the endothelium or by the presence of the nitric oxide synthase inhibitor L-NAME, the soluble guanylate cyclase inhibitor ODQ, or the cyclooxygenase inhibitor indomethacin. In contrast, when the DA rings were incubated in Ca2+-free solution, or in the presence of inhibitors of L-type Ca2+ channels (nifedipine), or large-conductance Ca2+-activated K+ (BKCa) channels (iberiotoxin), MgSO4-induced relaxation was impaired. Preincubation of the DA rings with MgSO4 concentrations ranging from 0 to 6.0 mmol L-1 did not significantly affect O2-induced contraction that was only impaired by a concentration of 7.2 mmol L-1. In conclusion, MgSO4 induced endothelium-independent relaxation of chicken DA and this relaxation appeared to be mediated through stimulation of BKCa channels and blockade of transmembrane flux of extracellular Ca2+. However, O2-induced DA contraction was only impaired by suprapharmacological concentrations of MgSO4 (> 6.0 mmol L-1). Therefore, our data suggest that the higher incidence of patent DA in preterm infants exposed to MgSO4 is unlikely to be due to a direct pharmacological effect of the drug on the DA.


Assuntos
Canal Arterial/efeitos dos fármacos , Sulfato de Magnésio/farmacologia , Animais , Cálcio/metabolismo , Embrião de Galinha , Dinoprostona/fisiologia , Canal Arterial/fisiologia , Permeabilidade do Canal Arterial/induzido quimicamente , Óxido Nítrico/fisiologia , Vasodilatação/efeitos dos fármacos
9.
J Matern Fetal Neonatal Med ; 33(4): 645-650, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29986620

RESUMO

Introduction: Premature birth is a leading cause of neonatal morbidity and mortality. Since gestational age at birth is the most important predictive factor of adverse neonatal outcomes, strategies to postpone premature labor are of major importance. Studies on tocolytic drugs show that COX inhibitors such as indomethacin are superior to others in terms of efficiency in delaying birth, but results concerning neonatal outcomes associated with prenatal exposure to these drugs show controversial results. Indomethacin is also used in the postnatal age for pharmacologic treatment of patent ductus arteriosus (PDA), but no data concerning the effects of antenatal exposure on postnatal ductal patency are available.Methods: In this study, we focused primarily on the association between antenatal indomethacin (AI) and postnatal patency of ductus arteriosus while our secondary aim was to highlight any possible influence of AI exposure on adverse neonatal outcomes. We performed a retrospective analysis of 241 medical records of newborns born before 33 weeks' gestation and exposed to antenatal tocolysis. Obstetrical data and neonatal outcomes of newborns exposed to AI were compared to those of neonates exposed to other tocolytic drugs. Early ductal closure (EDC) was defined when functional echocardiography performed within 24 hours of life showed a closed duct. Occurrence of intraventricular hemorrhage (IVH), periventricular leukomalacia (PVL), respiratory distress syndrome (RDS), chronic lung disease (CLD), necrotizing enterocolitis (NEC), sepsis, and PDA were compared between the groups and the diagnosis of at least one of III-IV grade IVH, PVL, CLD, sepsis, surgical NEC, or death was defined as a severe outcome.Results: The univariate analysis showed that infants in the AI group were at a higher risk of IVH, CLD, RDS, sepsis, and PDA. The incidence of severe outcomes also appeared to be higher in this group, while no effect of AI on PDA was observed. Since we noticed that infants exposed to AI had a lower gestational age and worse clinical conditions at birth when compared to the controls, we considered this as a confounding factor. To overcome this bias, we performed a multivariate analysis that evidenced no significant role of AI on the occurrence of severe outcomes. On the other hand, a possible association was confirmed for all degrees of IVH (OR: 3.16, 95% CI : [1.41; 7.05]) and sepsis (OR: 2.81, 95% CI: [1.24; 6,28]).Conclusions: The unexpected result shown by the multivariate analysis was the association between AI exposure and EDC (OR: 2.52, 95% CI: [1.02; 6.21]). This result, which has never been evidenced in previous studies, has great clinical importance. It is well known that PDA is more frequent at lower gestational ages, thus reducing the incidence of PDA could lead to an improvement of overall outcomes in extremely preterm newborns.


Assuntos
Canal Arterial/efeitos dos fármacos , Indometacina/efeitos adversos , Tocolíticos/efeitos adversos , Feminino , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Gravidez , Estudos Retrospectivos
10.
Pediatr Res ; 87(6): 991-997, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31816622

RESUMO

BACKGROUND: Indomethacin treatment for patent ductus arteriosus (PDA) is associated with acute kidney injury (AKI). Fenoldopam, a dopamine (DA) DA1-like receptor agonist dilates the renal vasculature and may preserve renal function during indomethacin treatment. However, limited information exists on DA receptor-mediated signaling in the ductus and fenoldopam may prevent ductus closure given its vasodilatory nature. METHODS: DA receptor expression in CD-1 mouse vessels was analyzed by qPCR and immunohistochemistry. Concentration-response curves were established using pressure myography. Pretreatment with SCH23390 (DA1-like receptor antagonist), phentolamine (α -adrenergic receptor antagonist) or indomethacin addressed mechanisms for DA-induced changes. Fenoldopam's effects on postnatal ductus closure were evaluated in vivo. RESULTS: DA1 receptors were expressed equally in ductus and aorta. High-dose DA induced modest vasoconstriction under newborn O2 conditions. Phentolamine inhibited DA-induced constriction, while SCH23390 augmented constriction, consistent with a vasodilatory role for DA1 receptors. Despite this, fenoldopam had little effect on ductus tone nor indomethacin- or O2-induced constriction and did not impair postnatal closure in vivo. CONCLUSION(S): DA receptors are present in the ductus but have limited physiologic effects. DA-induced ductus vasoconstriction is mediated via α-adrenergic pathways. The absence of DA1-mediated impairment of ductus closure supports the study of potential role for fenoldopam during PDA treatment.


Assuntos
Agonistas de Dopamina/farmacologia , Dopamina/metabolismo , Permeabilidade do Canal Arterial/tratamento farmacológico , Canal Arterial/efeitos dos fármacos , Fenoldopam/farmacologia , Receptores de Dopamina D1/agonistas , Vasoconstrição/efeitos dos fármacos , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia , Animais , Canal Arterial/metabolismo , Canal Arterial/fisiopatologia , Permeabilidade do Canal Arterial/metabolismo , Permeabilidade do Canal Arterial/fisiopatologia , Feminino , Indometacina/toxicidade , Camundongos , Oxigênio/toxicidade , Gravidez , Receptores de Dopamina D1/metabolismo , Transdução de Sinais
11.
J Pharmacol Exp Ther ; 370(3): 350-359, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31201216

RESUMO

Glucose-stimulated insulin secretion from pancreatic ß-cells is controlled by ATP-regulated potassium (KATP) channels composed of Kir6.2 and sulfonylurea receptor 1 (SUR1) subunits. The KATP channel-opener diazoxide is FDA-approved for treating hyperinsulinism and hypoglycemia but suffers from off-target effects on vascular KATP channels and other ion channels. The development of more specific openers would provide critically needed tool compounds for probing the therapeutic potential of Kir6.2/SUR1 activation. Here, we characterize a novel scaffold activator of Kir6.2/SUR1 that our group recently discovered in a high-throughput screen. Optimization efforts with medicinal chemistry identified key structural elements that are essential for VU0071063-dependent opening of Kir6.2/SUR1. VU0071063 has no effects on heterologously expressed Kir6.1/SUR2B channels or ductus arteriole tone, indicating it does not open vascular KATP channels. VU0071063 induces hyperpolarization of ß-cell membrane potential and inhibits insulin secretion more potently than diazoxide. VU0071063 exhibits metabolic and pharmacokinetic properties that are favorable for an in vivo probe and is brain penetrant. Administration of VU0071063 inhibits glucose-stimulated insulin secretion and glucose-lowering in mice. Taken together, these studies indicate that VU0071063 is a more potent and specific opener of Kir6.2/SUR1 than diazoxide and should be useful as an in vitro and in vivo tool compound for investigating the therapeutic potential of Kir6.2/SUR1 expressed in the pancreas and brain.


Assuntos
Ativação do Canal Iônico/efeitos dos fármacos , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Receptores de Sulfonilureias/metabolismo , Xantinas/farmacologia , Xantinas/farmacocinética , Animais , Canal Arterial/efeitos dos fármacos , Canal Arterial/fisiologia , Glucose/farmacologia , Células HEK293 , Humanos , Secreção de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Camundongos , Relação Estrutura-Atividade , Vasodilatação/efeitos dos fármacos , Xantinas/química
12.
Pediatr Res ; 86(1): 28-38, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30965358

RESUMO

The ductus arteriosus (DA) is probably the most intriguing vessel in postnatal hemodynamic transition. DA patency in utero is an active state, in which prostaglandin E2 (PGE2) and nitric monoxide (NO), play an important role. Since the DA gets programmed for postnatal closure as gestation advances, in preterm infants the DA frequently remains patent (PDA). PGE2 exposure programs functional postnatal closure by inducing gene expression of ion channels and phosphodiesterases and anatomical closure by inducing intimal thickening. Postnatally, oxygen inhibits potassium and activates calcium channels, which ultimately leads to a rise in intracellular calcium concentration consequently inducing phosphorylation of the myosin light chain and thereby vasoconstriction of the DA. Since ion channel expression is lower in preterm infants, oxygen induced functional vasoconstriction is attenuated in comparison with full term newborns. Furthermore, the preterm DA is more sensitive to both PGE2 and NO compared to the term DA pushing the balance toward less constriction. In this review we explain the physiology of DA patency in utero and subsequent postnatal functional closure. We will focus on the pathobiology of PDA in preterm infants and the (un)intended effect of antenatal exposure to medication on both fetal and neonatal DA vascular tone.


Assuntos
Dinoprostona/metabolismo , Permeabilidade do Canal Arterial/fisiopatologia , Hemodinâmica , Doenças do Prematuro/fisiopatologia , Oxigênio/metabolismo , Animais , Animais Recém-Nascidos , Canal Arterial/efeitos dos fármacos , Permeabilidade do Canal Arterial/tratamento farmacológico , Permeabilidade do Canal Arterial/cirurgia , Homeostase , Humanos , Incidência , Recém-Nascido , Recém-Nascido Prematuro , Cadeias Leves de Miosina/metabolismo , Fosforilação , Prostaglandinas/metabolismo , Vasoconstrição , Vasodilatação
13.
Am J Physiol Regul Integr Comp Physiol ; 316(6): R716-R724, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30840485

RESUMO

The glucocorticosteroid betamethasone is routinely administered via maternal intramuscular injection to enhance fetal lung maturation before anticipated preterm birth. Although antenatal betamethasone increases fetal pulmonary arterial (PA) blood flow, whether this agent alters the contribution of 1) right ventricular (RV) output or 2) left-to-right shunting across the ductus arteriosus to rises in PA blood flow after preterm birth is unknown. To address this question, anesthetized control (n = 7) and betamethasone-treated (n = 7) preterm fetal lambs (gestation 127 ± 1 days, means ± SD) were instrumented with aortic, pulmonary, and left atrial catheters as well as ductus arteriosus and left PA flow probes to calculate RV output, with hemodynamics measured for 30 min after cord clamping and mechanical ventilation. Mean PA blood flow was higher in betamethasone-treated than in control lambs over the initial 10 min after birth (P < 0.05). This higher PA flow was accompanied by 1) a greater pulmonary vascular conductance (P ≤ 0.025), 2) a larger proportion of RV output passing to lungs (P ≤ 0.01), despite a fall in this output, and 3) earlier reversal and a greater magnitude (P ≤ 0.025) of net ductal shunting, due to the combination of higher left-to-right (P ≤ 0.025) and lesser right-to-left phasic shunting (P ≤ 0.025). These results suggest that antenatal betamethasone augments the initial rise in PA blood flow after birth in preterm lambs, with this augmented rise supported by the combination of 1) a greater redistribution of RV output toward the lungs and 2) a faster and larger reversal in net ductal shunting underpinned not only by greater left-to-right, but also by lesser right-to-left phasic shunting.


Assuntos
Betametasona/administração & dosagem , Canal Arterial/efeitos dos fármacos , Glucocorticoides/administração & dosagem , Pulmão/irrigação sanguínea , Nascimento Prematuro , Circulação Pulmonar/efeitos dos fármacos , Volume Sistólico/efeitos dos fármacos , Função Ventricular Direita/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Velocidade do Fluxo Sanguíneo , Modelos Animais de Doenças , Esquema de Medicação , Canal Arterial/fisiopatologia , Feminino , Idade Gestacional , Gravidez , Carneiro Doméstico , Fatores de Tempo
14.
J Perinatol ; 39(5): 599-607, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30850756

RESUMO

OBJECTIVE: To evaluate the effectiveness of drugs used to constrict patent ductus arteriosus (PDA) in newborns < 28 weeks. METHODS: We performed a secondary analysis of the multi-center PDA-TOLERATE trial (NCT01958320). Infants with moderate-to-large PDAs were randomized 1:1 at 8.1 ± 2.1 days to either Drug treatment (n = 104) or Conservative management (n = 98). Drug treatments were assigned by center rather than within center (acetaminophen: 5 centers, 27 infants; ibuprofen: 7 centers, 38 infants; indomethacin: 7 centers, 39 infants). RESULTS: Indomethacin produced the greatest constriction (compared with spontaneous constriction during Conservative management): RR (95% CI) = 3.21 (2.05-5.01)), followed by ibuprofen = 2.03 (1.05-3.91), and acetaminophen = 1.33 (0.55-3.24). The initial rate of acetaminophen-induced constriction was 27%. Infants with persistent moderate-to-large PDA after acetaminophen were treated with indomethacin. The final rate of constriction after acetaminophen ± indomethacin was 60% (similar to the rate in infants receiving indomethacin-alone (62%)). CONCLUSION: Indomethacin was more effective than acetaminophen in producing ductus constriction.


Assuntos
Acetaminofen/uso terapêutico , Permeabilidade do Canal Arterial/tratamento farmacológico , Ibuprofeno/uso terapêutico , Indometacina/uso terapêutico , Vasoconstrição/efeitos dos fármacos , Administração Intravenosa , Administração Oral , Tratamento Conservador , Canal Arterial/efeitos dos fármacos , Feminino , Humanos , Lactente Extremamente Prematuro , Recém-Nascido , Masculino , São Francisco , Resultado do Tratamento
15.
Circ J ; 83(3): 654-661, 2019 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-30726804

RESUMO

BACKGROUND: Antenatal betamethasone (BMZ) is a standard therapy for reducing respiratory distress syndrome in preterm infants. Recently, some reports have indicated that BMZ promotes ductus arteriosus (DA) closure. DA closure requires morphological remodeling; that is, intimal thickening (IT) formation; however, the role of BMZ in IT formation has not yet been reported. Methods and Results: First, DNA microarray analysis using smooth muscle cells (SMCs) of rat preterm DA on gestational day 20 (pDASMCs) stimulated with BMZ was performed. Among 58,717 probe sets, ADP-ribosyltransferase 3 (Art3) was markedly increased by BMZ stimulation. Quantitative reverse transcription polymerase chain reaction (RT-PCR) confirmed the BMZ-induced increase of Art3 in pDASMCs, but not in aortic SMCs. Immunocytochemistry showed that BMZ stimulation increased lamellipodia formation. BMZ significantly increased total paxillin protein expression and the ratio of phosphorylated to total paxillin. A scratch assay demonstrated that BMZ stimulation promoted pDASMC migration, which was attenuated byArt3-targeted siRNAs transfection. pDASMC proliferation was not promoted by BMZ, which was analyzed by a 5'-bromo-2'-deoxyuridine (BrdU) assay. Whether BMZ increased IT formation in vivo was examined. BMZ or saline was administered intravenously to maternal rats on gestational days 18 and 19, and DA tissues were obtained on gestational day 20. The ratio of IT to tunica media was significantly higher in the BMZ-treated group. CONCLUSIONS: These data suggest that antenatal BMZ administration promotes DA IT through Art3-mediated DASMC migration.


Assuntos
Betametasona/farmacologia , Canal Arterial/efeitos dos fármacos , Túnica Íntima/efeitos dos fármacos , ADP Ribose Transferases/efeitos dos fármacos , Animais , Movimento Celular/efeitos dos fármacos , Canal Arterial/patologia , Feminino , Miócitos de Músculo Liso/metabolismo , Gravidez , Ratos
16.
Br J Clin Pharmacol ; 85(1): 245-251, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30300944

RESUMO

Recent case reports describe an association between maternal paracetamol intake and fetal ductus arteriosus constriction or closure. To put these cases into perspective and explore causality, a structured literature search was conducted. The World Health Organization Uppsala Monitoring Centre (WHO-UMC) causality tool was applied to the cases retrieved. The search resulted in 12 papers with 25 case descriptions, of which one case was classified as unlikely, nine as possible, 11 as probable and four as certain. Consequently, we concluded that a causal relationship between maternal paracetamol intake and fetal ductus arteriosus constriction or closure is likely. These findings suggest that pharmacovigilance studies on paracetamol safety during pregnancy are warranted to quantify the event and put the current findings into clinical perspective. Although analgesia during pregnancy and during the peripartum period is of obvious relevance, alternative analgesics such as opioids or other nonsteroidal anti-inflammatory drugs also have side effects.


Assuntos
Acetaminofen/efeitos adversos , Analgésicos não Narcóticos/efeitos adversos , Canal Arterial/patologia , Doenças do Recém-Nascido/epidemiologia , Exposição Materna/efeitos adversos , Constrição Patológica/induzido quimicamente , Constrição Patológica/epidemiologia , Canal Arterial/efeitos dos fármacos , Feminino , Humanos , Incidência , Recém-Nascido , Doenças do Recém-Nascido/induzido quimicamente , Exposição Materna/estatística & dados numéricos , Gravidez
17.
Fetal Diagn Ther ; 45(5): 339-344, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30157479

RESUMO

INTRODUCTION: The use of perioperative tocolytic agents in fetal surgery is imperative to prevent preterm labor. Indomethacin, a well-known tocolytic agent, can cause ductus arteriosus (DA) constriction. We sought to determine whether a relationship exists between preoperative indomethacin dosing and fetal DA constriction. MATERIALS AND METHODS: This is an IRB-approved, single-center retrospective observational case series of 42 pregnant mothers who underwent open fetal myelomeningocele repair. Preoperatively, mothers received either 1 (QD) or 2 (BID) indomethacin doses. Maternal anesthetic drug exposures and fetal cardiac dysfunction measures were collected from surgical and anesthesia records and intraoperative fetal echocardiography. Pulsatility Index was used to calculate DA constriction severity. Comparative testing between groups was performed using t- and chi-square testing. RESULTS: DA constriction was observed in all fetuses receiving BID indomethacin and in 71.4% of those receiving QD dosing (p = 0.0002). Severe DA constriction was observed only in the BID group (35.7%). QD indomethacin group received more intraoperative magnesium sulfate (p < 0.0001). Minimal fetal cardiac dysfunction (9.5%) and bradycardia (9.5%) were observed in all groups independent of indomethacin dosing. CONCLUSIONS: DA constriction was the most frequent and severe in the BID indomethacin group. QD indomethacin and greater magnesium sulfate dosing was associated with reduced DA constriction.


Assuntos
Anti-Inflamatórios não Esteroides/administração & dosagem , Canal Arterial/cirurgia , Terapias Fetais/métodos , Indometacina/administração & dosagem , Meningomielocele/cirurgia , Tocolíticos/administração & dosagem , Constrição , Relação Dose-Resposta a Droga , Canal Arterial/diagnóstico por imagem , Canal Arterial/efeitos dos fármacos , Feminino , Humanos , Meningomielocele/diagnóstico por imagem , Meningomielocele/tratamento farmacológico , Gravidez , Estudos Retrospectivos , Ultrassonografia Pré-Natal/métodos
18.
Clin Sci (Lond) ; 132(18): 2045-2058, 2018 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-30219798

RESUMO

The physiologic process of postnatal ductus arteriosus (DA) closure consists of vasoconstriction followed by vascular remodeling. We have recently reported that B-type natriuretic peptide (BNP), a potent vasodilator, also has anti-remodeling effects in pulmonary vasculature. However, its effects on DA have not been elucidated. We investigated whether BNP can prevent DA closure, and if so, the underlying mechanisms. Using in vivo studies, we examined effects of BNP (10 mg/kg, ip at birth) on DA closure in neonatal rats within 4 h after birth. We found that in control rats, the DA spontaneously closed at 4 h with a decreased DA diameter, enhanced intimal thickening, and luminal occlusion. BNP prevented DA closure at 4 h with a preserved DA diameter, attenuated intimal thickening, and preserved luminal patency. Ex vivo, BNP attenuated oxygen-induced vasoconstriction of isolated DA rings of newborn rats. These vasodilating effects were blunted by Rp-8-Br-PET-cGMPS, a cGMP inhibitor. In vitro, BNP inhibited angiotensin II (Ang II)-induced proliferation and migration of DA smooth muscle cells (DASMCs). BNP inhibited Ang II-induced mitochondrial reactive oxygen species (ROS) production and calcium overload in DASMCs. Finally, BNP inhibited Ang II-induced ERK1/2 activation. These in vitro effects were antagonized by Rp-8-Br-PET-cGMPS. In conclusion, BNP prevents postnatal DA closure by both vasodilation and anti-remodeling through the cGMP pathway. The mechanisms underlying anti-remodeling effects include anti-poliferation and anti-migration, with attenuation of mitochondrial ROS production and intracellular calcium and ERK1/2 signaling. Therefore, the BNP/cGMP pathway can be a promising therapeutic target for clinical management of DA patency.


Assuntos
Canal Arterial/efeitos dos fármacos , Peptídeo Natriurético Encefálico/farmacologia , Remodelação Vascular/efeitos dos fármacos , Vasodilatação/efeitos dos fármacos , Angiotensina II/farmacologia , Animais , Animais Recém-Nascidos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacologia , Canal Arterial/citologia , Canal Arterial/fisiologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/fisiologia , Ratos Wistar , Tionucleotídeos/farmacologia , Fatores de Tempo , Remodelação Vascular/fisiologia , Vasodilatação/fisiologia
19.
J Neonatal Perinatal Med ; 11(3): 273-279, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30149471

RESUMO

OBJECTIVE: Patent ductus arteriosus is a common problem frequently encountered in preterm infants. We aimed to study the risk factors associated with reopening of patent ductus arteriosus and their short term outcomes in preterm infants. METHODS: A total of 162 preterm infants born between November 2013 and December 2015 with gestaional age less than 32 weeks and treated for hemodynamically significant patent ductus arteriosus are included in our study. RESULTS: 113(69.8%) showed permanent closure and 49(30.2%) infants revealed symptoms of reopening after effective closure of patent ductus arteriosus. Low birth weight and small gestational age were more common in reopening group. Multivariete analysis showed that sepsis and multiple courses of drug treatment were independent factors affecting reopening of hemodynamically significant patent ductus arteriosus (OR: 3.01, 95% CI 1.48-6.13, p = 0.002) and (OR: 2.67, 95% CI 1.23-5.82, p = 0.013) respectively. Reopened group had a remarkable higher rate of developing necrotising nnterocolitis, bronchopulmonary dysplasia and retinopathy of prematurity than the closed group. (16.3% vs 4.4%, p = 0.01, 55.1% vs 28.3%, p = 0.001 and 55.1% vs 23.0%, p = 0.0001 respectively). CONCLUSION: Late neonatal sepsis and the need of multiple drug courses to close patent ductus arteriosus are risk factors affecting the reopening of patent ductus arteriosus in preterm infants.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Permeabilidade do Canal Arterial/tratamento farmacológico , Canal Arterial/efeitos dos fármacos , Hemodinâmica/efeitos dos fármacos , Ibuprofeno/uso terapêutico , Doenças do Prematuro/tratamento farmacológico , Relação Dose-Resposta a Droga , Canal Arterial/fisiopatologia , Permeabilidade do Canal Arterial/fisiopatologia , Feminino , Idade Gestacional , Humanos , Lactente , Recém-Nascido , Recém-Nascido Prematuro , Doenças do Prematuro/fisiopatologia , Masculino , Estudos Retrospectivos , Resultado do Tratamento
20.
Pediatr Res ; 84(3): 458-465, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29976969

RESUMO

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.


Assuntos
Betametasona/uso terapêutico , Permeabilidade do Canal Arterial/tratamento farmacológico , Canal Arterial/efeitos dos fármacos , Animais , Ecocardiografia , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Recém-Nascido Prematuro , Exposição Materna , Camundongos , Oxigênio/metabolismo , Papio , Reação em Cadeia da Polimerase , Prostaglandinas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...