Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
2.
Eur J Clin Nutr ; 69(10): 1105-8, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25920422

ABSTRACT

BACKGROUND/OBJECTIVES: An elevated body mass index (BMI) in childhood is a significant risk factor for cardiovascular disease, and it may pose an additional risk to children and adults with palliated univentricular congenital heart disease. However, little is known about longitudinal development of obesity in this population. The objective of this study is to determine the prevalence of overweight (OW) and obese (OB) habitus at the time of Fontan palliative surgery, to track changes in BMI after surgery, and ultimately to determine whether factors such as gender, ethnicity, preoperative heart defect and ventricular dominance are associated with later development of OW or OB. SUBJECTS/METHODS: A retrospective chart review of 84 patients undergoing Fontan palliation was performed. Demographic data including gender, ethnicity, preoperative heart defect and ventricular dominance were recorded. Height, weight and BMI were obtained at the time of Fontan and on a yearly basis post surgery. RESULTS: At the time of Fontan palliation, 10.7% of patients were OB or OW. During the five years following palliation, the percentage of OB or OW patients trended upward, from 20.3% the year following surgery to 30% at 5 years post Fontan. Repeated measures generalized estimating equation showed a significant association between Hispanic ethnicity and increased BMI Z-scores for the 5 years after Fontan palliation (P<0.001); there was no association between BMI Z-scores and patient sex, lesion or ventricular dominance. CONCLUSIONS: During the first 5 years after Fontan palliation, there is a trend toward increasing percentages of OB and OW patients. In addition, there is a significant association between Hispanic ethnicity and being OW or OB before and after surgery. Further study is needed to determine whether OW/OB status is associated with worse health outcomes in this patient population.


Subject(s)
Body Mass Index , Heart Defects, Congenital/surgery , Obesity/etiology , Palliative Care , Postoperative Complications , Child , Female , Hispanic or Latino , Humans , Male , Obesity/epidemiology , Overweight/epidemiology , Overweight/etiology
3.
Am J Physiol Lung Cell Mol Physiol ; 281(5): L1051-7, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11597895

ABSTRACT

Utilizing aortopulmonary vascular graft placement, we established a lamb model of pulmonary hypertension that mimics congenital heart disease with increased pulmonary blood flow. We previously demonstrated that endothelial nitric oxide synthase (eNOS) is increased in lambs at age 4 wk. However, these lambs display a selective impairment of endothelium-dependent pulmonary vasodilation that is suggestive of a derangement downstream of NO release. Thus our objective was to characterize potential alterations in the expression and activity of soluble guanylate cyclase (sGC) and phosphodiesterase type 5 (PDE5) induced by increased pulmonary blood flow and pulmonary hypertension. Late-gestational fetal lambs (n = 10) underwent in utero placement of an aortopulmonary vascular graft (shunt). Western blotting analysis on lung tissue from 4-wk-old shunted lambs and age-matched controls showed that protein for both subunits of sGC was increased in shunted lamb lungs compared with age-matched controls. Similarly, cGMP levels were increased in shunted lamb lungs compared with age-matched controls. However, PDE5 expression and activity were also increased in shunted lambs. Thus although cGMP generation was increased, concomitant upregulation of PDE5 expression and activity may have (at least partially) limited and accounted for the impairment of endothelium-dependent pulmonary vasodilation in shunted lambs.


Subject(s)
3',5'-Cyclic-GMP Phosphodiesterases/metabolism , Hypertension, Pulmonary/physiopathology , Pulmonary Circulation/physiology , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 5 , Female , Guanylate Cyclase , Hypertension, Pulmonary/metabolism , Immunoblotting , Immunohistochemistry , Lung/cytology , Lung/enzymology , Lung/metabolism , Nitric Oxide/metabolism , Pregnancy , Sheep , Soluble Guanylyl Cyclase
4.
Am J Physiol Lung Cell Mol Physiol ; 281(5): L1264-70, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11597919

ABSTRACT

Infants with increased pulmonary blood flow secondary to congenital heart disease suffer from tachypnea, dyspnea, and recurrent pulmonary infections. We have recently established a model of pulmonary hypertension secondary to increased pulmonary blood flow in lambs after in utero placement of an aortopulmonary vascular graft. The purpose of the present study was to utilize our animal model to determine the effects on the expression of surfactant proteins A (SP-A), B (SP-B), and C (SP-C). At age 4 wk, SP-A mRNA content in lambs decreased to 61.4 +/- 8% of age-matched control value (n = 5; P < 0.05). In addition, SP-A protein content was decreased to 50 +/- 12% of control value (n = 6; P < 0.0001). Although we did not observe statistically significant changes in SP-B mRNA content, SP-B protein was decreased to 74 +/- 25% of control value (n = 4; P < 0.02). There was no difference in SP-C mRNA. These data show that in a model of congenital heart disease with pulmonary hypertension secondary to increased pulmonary blood flow, there is a decrease in SP-A gene expression as well as a decrease in SP-A and SP-B protein contents.


Subject(s)
Hypertension, Pulmonary/physiopathology , Lung/metabolism , Proteolipids/metabolism , Pulmonary Circulation , Pulmonary Surfactants/metabolism , Animals , Animals, Newborn , Child , Disease Models, Animal , Female , Heart Defects, Congenital/physiopathology , Humans , Pregnancy , Proteolipids/genetics , Pulmonary Surfactant-Associated Protein A , Pulmonary Surfactant-Associated Proteins , Pulmonary Surfactants/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sheep , Tissue Extracts/chemistry
5.
Circ Res ; 89(4): 357-64, 2001 Aug 17.
Article in English | MEDLINE | ID: mdl-11509453

ABSTRACT

Our previous studies have demonstrated that inhaled nitric oxide (NO) decreases nitric oxide synthase (NOS) activity in vivo and that this inhibition is associated with rebound pulmonary hypertension upon acute withdrawal of inhaled NO. We have also demonstrated that inhaled NO elevates plasma endothelin-1 (ET-1) levels and that pretreatment with PD156707, an ETA receptor antagonist, blocks the rebound hypertension. The objectives of this study were to further elucidate the role of ET-1 in the rebound pulmonary hypertension upon acute withdrawal of inhaled NO. Inhaled NO (40 ppm) delivered to thirteen 4-week-old lambs decreased NOS activity by 36.2% in control lambs (P<0.05), whereas NOS activity was preserved in PD156707-treated lambs. When primary cultures of pulmonary artery smooth muscle cells were exposed to ET-1, superoxide production increased by 33% (P<0.05). This increase was blocked by a preincubation with PD156707. Furthermore, cotreatment of cells with ET-1 and NO increased peroxynitrite levels by 26% (P<0.05), whereas preincubation of purified human endothelial nitric oxide synthase (eNOS) protein with peroxynitrite generated a nitrated enzyme with 50% activity relative to control (P<0.05). Western blot analysis of peripheral lung extracts obtained after 24 hours of inhaled NO revealed a 90% reduction in 3-nitrotyrosine residues (P<0.05) in PD156707-treated lambs. The nitration of eNOS was also reduced by 40% in PD156707-treated lambs (P<0.05). These data suggest that the reduction of NOS activity associated with inhaled NO therapy may involve ETA receptor-mediated superoxide production. ETA receptor antagonists may prevent rebound pulmonary hypertension by protecting endogenous eNOS activity during inhaled NO therapy.


Subject(s)
Endothelin-1/metabolism , Hypertension, Pulmonary/metabolism , Nitrates/metabolism , Nitric Oxide/adverse effects , Superoxides/metabolism , Tyrosine/analogs & derivatives , Administration, Inhalation , Animals , Blotting, Western , Cells, Cultured , Dioxoles/pharmacology , Disease Models, Animal , Endothelin Receptor Antagonists , Endothelin-1/pharmacology , Enzyme Activation/drug effects , Humans , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/prevention & control , Lung/chemistry , Lung/metabolism , Microscopy, Fluorescence , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Nitrates/chemistry , Nitric Oxide/administration & dosage , Nitric Oxide/pharmacology , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase/chemistry , Nitric Oxide Synthase/isolation & purification , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type III , Pulmonary Artery/cytology , Pulmonary Artery/drug effects , Pulmonary Artery/metabolism , Receptor, Endothelin A , Secondary Prevention , Sheep , Tyrosine/analysis , Tyrosine/biosynthesis
6.
Am J Physiol Heart Circ Physiol ; 280(2): H777-85, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11158977

ABSTRACT

Clinically significant increases in pulmonary vascular resistance have been noted on acute withdrawal of inhaled nitric oxide (NO). Endothelin (ET)-1 is a vasoactive peptide produced by the vascular endothelium that may participate in the pathophysiology of pulmonary hypertension. The objectives of this study were to determine the effects of inhaled NO on endogenous ET-1 production in vivo in the intact lamb and to determine the potential role of ET-1 in the rebound pulmonary hypertension associated with the withdrawal of inhaled NO. Seven 1-mo-old vehicle-treated control lambs and six PD-156707 (an ET(A) receptor antagonist)-treated lambs were mechanically ventilated. Inhaled NO (40 parts per million) was administered for 24 h and then acutely withdrawn. After 24 h of inhaled NO, plasma ET-1 levels increased by 119.5 +/- 42.2% (P < 0.05). Western blot analysis revealed that protein levels of preproET-1, endothelin-converting enzyme-1alpha, and ET(A) and ET(B) receptors were unchanged. On acute withdrawal of NO, pulmonary vascular resistance (PVR) increased by 77.8% (P < 0.05) in control lambs but was unchanged (-5.5%) in PD-156707-treated lambs. Inhaled NO increased plasma ET-1 concentrations but not gene expression in the intact lamb, and ET(A) receptor blockade prevented the increase in PVR after NO withdrawal. These data suggest a role for ET-1 in the rebound pulmonary hypertension noted on acute withdrawal of inhaled NO.


Subject(s)
Endothelin-1/physiology , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/physiopathology , Nitric Oxide/pharmacology , Administration, Inhalation , Animals , Animals, Newborn , Blotting, Western , Dioxoles/pharmacology , Endothelin Receptor Antagonists , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Hypertension, Pulmonary/metabolism , Lung/blood supply , Lung/chemistry , Lung/enzymology , Metalloendopeptidases/analysis , Pulmonary Circulation/drug effects , Pulmonary Circulation/physiology , Receptor, Endothelin A , Receptor, Endothelin B , Receptors, Endothelin/analysis , Receptors, Endothelin/physiology , Respiration, Artificial , Sheep , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/physiopathology , Vascular Resistance/drug effects , Vascular Resistance/physiology
7.
Lancet ; 357(9250): 118-20, 2001 Jan 13.
Article in English | MEDLINE | ID: mdl-11197402

ABSTRACT

To investigate the oxygenation and haemodynamic dose response to inhaled nitric oxide in neonates with persistent pulmonary hypertension (PPHN), we gave seven neonates nitric oxide and measured directly pulmonary arterial pressure. Inhaled nitric oxide produced peak improvement in oxygenation at 5 parts per million (ppm) whereas peak improvement in the pulmonary-to-systemic arterial pressure ratio did not occur until a nitric oxide dose of 20 ppm, which suggests that an Initial dose of 20 ppm is optimum for the treatment of PPHN.


Subject(s)
Hypertension, Pulmonary/drug therapy , Nitric Oxide/administration & dosage , Vasodilator Agents/administration & dosage , Administration, Inhalation , Humans , Infant, Newborn
8.
Am J Physiol Heart Circ Physiol ; 280(1): H311-7, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11123246

ABSTRACT

Congenital heart disease associated with increased pulmonary blood flow produces pulmonary hypertension. To characterize vascular alterations in the nitric oxide (NO)-cGMP cascade induced by increased pulmonary blood flow and pulmonary hypertension, 10 fetal lambs underwent in utero placement of an aortopulmonary vascular graft (shunt). When the lambs were 4-6 wk of age, we assessed responses of pulmonary arteries (PAs) and pulmonary veins (PVs) isolated from lungs of control and shunted lambs. PVs from control and shunted lambs relaxed similarly to exogenous NO (S-nitrosyl-acetyl-penicillamine), to NO produced endogenously (zaprinast and A-23187), and to cGMP (atrial natriuretic peptide). In contrast, relaxations to A-23187 and zaprinast were blunted in PAs isolated from shunted lambs relative to controls. Inhibitors of NO synthase (NOS) and soluble guanylate cyclase constricted control but not shunt PAs, indicating reduced basal NOS activity in shunt PAs. Pretreatment of shunt PAs with the substrates L-arginine and sepiapterin, a precursor for tetrahydrobiopterin synthesis, did not augment A-23187 relaxations. However, pretreatment with superoxide dismutase and catalase significantly enhanced A-23187 relaxations in shunt PAs. We conclude that increased pulmonary blood flow induces an impairment of endothelium-dependent relaxation that is selective to PAs. The impaired relaxation may be mediated in part by excess superoxide production.


Subject(s)
Endothelium, Vascular/physiopathology , Hypertension, Pulmonary/physiopathology , Muscle, Smooth, Vascular/physiopathology , Pulmonary Circulation , 3',5'-Cyclic-GMP Phosphodiesterases/antagonists & inhibitors , Animals , Atrial Natriuretic Factor/pharmacology , Calcimycin/pharmacology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Fetus , Guanylate Cyclase/antagonists & inhibitors , Hypertension, Pulmonary/congenital , In Vitro Techniques , Muscle Relaxation , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type III , Phosphodiesterase Inhibitors/pharmacology , Pulmonary Artery/physiopathology , Pulmonary Veins/physiopathology , Purinones/pharmacology , Sheep
9.
Circulation ; 102(19 Suppl 3): III172-8, 2000 Nov 07.
Article in English | MEDLINE | ID: mdl-11082382

ABSTRACT

BACKGROUND: After cardiopulmonary bypass (CPB), altered vascular reactivity is a major source of complications, particularly for children with increased pulmonary blood flow. Although changes in agonist-induced NO activity are well described after CPB, potential changes in basal NO production and their role in post-CPB pulmonary hypertension remain unclear. By using aortopulmonary vascular graft placement in the fetal lamb (shunt lambs), we established a unique model of pulmonary hypertension that mimics congenital heart disease with increased pulmonary blood flow. The objective of the present study was to investigate potential alterations in endogenous NO production after CPB in lambs with normal and increased pulmonary blood flow. METHODS AND RESULTS: Vascular pressures and blood flows were monitored in 1-month-old lambs (n=7) with increased pulmonary blood flow and 6 age-matched control lambs. After shunt closure, hypothermic CPB (25 degrees C) was performed for 2 hours. The hemodynamic variables were monitored for 4 hours after CPB. Before, during, and after CPB, peripheral lung biopsies were performed to determine tissue NO, nitrite, nitrate, and cGMP concentrations; total NO synthase (NOS) activity; and endothelial NOS protein levels. Hypothermic CPB increased both mean pulmonary arterial pressure and left pulmonary vascular resistance (P:<0.05). The increase in pulmonary arterial pressure induced in shunt lambs was greater than that induced in control lambs (P:<0.05). Four hours after CPB, tissue concentrations of NO, nitrite, nitrate, and cGMP were decreased to approximately 70% of pre-CPB levels in both control and shunt lambs (P:<0.05). Total NOS activity and endothelial NOS protein levels were unchanged. CONCLUSIONS: Modest decreases in basal NO production, the inability to increase NO production, or both may play a role in the altered pulmonary vascular reactivity after CPB. The decrease in NO is independent of gene expression. However, other mechanisms for this decrease, such as substrate or cofactor availability, warrant further study.


Subject(s)
Cardiopulmonary Bypass/adverse effects , Hypertension, Pulmonary/etiology , Lung/blood supply , Lung/metabolism , Nitric Oxide/metabolism , Pulmonary Circulation , Animals , Blood Flow Velocity , Blood Pressure , Blotting, Western , Cyclic GMP/metabolism , Disease Models, Animal , Nitrates/metabolism , Nitric Oxide/analysis , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type III , Pulmonary Artery/diagnostic imaging , Sheep , Ultrasonography
10.
Crit Care Med ; 28(8): 2997-3001, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10966285

ABSTRACT

OBJECTIVES: To characterize transthoracic intracardiac catheter uses and associated morbidities in pediatric patients recovering from congenital heart defect surgery and to identify potential risk factors associated with their use. DESIGN: Prospective data collection and review. SETTING: An 18-bed pediatric intensive care unit (PICU) in a tertiary care university hospital. PATIENTS: All pediatric patients between October 1, 1996, and September 30, 1997, who were recovering from congenital heart defect surgery and had transthoracic intracardiac catheters in place. MEASUREMENTS AND MAIN RESULTS: Catheter use, associated morbidity, necessary interventions, and risk factors for complications of catheter use were identified. During this period, 523 catheters (276 right atrial, 155 left atrial, 68 common atrial, and 24 right ventricular or pulmonary artery catheters) in 351 PICU patients were studied. Mean age was 23.1+/-45.1 months (median, 4.98 months); 138 patients (39.3%) were <3 months old. The rate of occurrence of bleeding with catheter removal (mediastinal output in the hour after removal that was more than twice the previous average hourly output) was 36.7%, and bleeding occurred more frequently with left atrial catheters (47%; odds ratio, 2.0; p < .05). However, interventions after catheter removal were required for only 8.3% (42/504) of catheters removed, and hemodynamic compromise occurred with the removal of only 2.6% (13/504) of catheters. Interventions included fluid resuscitation (35 cases), pleural drainage (three cases), catheter wiring for retention (one case), chest tube suctioning (two cases), and surgical removal (one case). No associated deaths occurred. In a multivariate logistic regression analysis, age <3 months (odds ratio, 4.74), catheter location (left atrial: odds ratio, 4.97; pulmonary artery: odds ratio, 12.48), and platelet count of <50,000 (odds ratio, 8.59) were identified as risk factors associated with a need for intervention after catheter removal (p < .05). Other complications included blood cultures positive for organisms (1.5%), thrombus (0.6%), and catheter nonfunction (10.9%). Prematurity was a risk factor for thrombus and nonfunction. CONCLUSIONS: Use of transthoracic intracardiac catheters in pediatric patients is safe. Young infants and pediatric patients with thrombocytopenia or with catheters in the left atrial or pulmonary artery position have a greater need for interventions after catheter removal, warranting added precautions.


Subject(s)
Cardiac Catheterization/adverse effects , Catheters, Indwelling/adverse effects , Heart Defects, Congenital/surgery , Postoperative Care , Postoperative Complications/etiology , Adolescent , Child , Child, Preschool , Hemodynamics , Humans , Infant , Infant, Newborn , Postoperative Complications/epidemiology , Postoperative Complications/physiopathology , Prospective Studies , Risk Factors , Treatment Outcome
11.
Crit Care Med ; 28(4): 1180-4, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10809302

ABSTRACT

OBJECTIVES: To investigate the efficacy, safety, and patterns of management of open sternotomy and delayed sternal closure in infants who were left with an open sternum after cardiac surgery and to assess these patterns for possible correlation with outcome. DESIGN: Retrospective chart review with statistical analysis. SETTING: Pediatric cardiac surgery service at a regional referral center based in an urban university teaching hospital. PATIENTS: All 128 patients <1 yr of age who were left with an open sternum after cardiac surgery with cardiopulmonary bypass during the 4-yr period from July, 1992 to June, 1996. INTERVENTIONS: Procedures for managing open sternotomy and delayed sternal closure were analyzed retrospectively. No interventions were undertaken for the study. MEASUREMENTS AND RESULTS: Of the 128 patients, 14 (11%) died before sternal closure; delayed sternal closure was performed in the remaining 114. Of these 114, 13 died in the early postoperative period. During sternal closure, significant increases were noted in pulmonary arterial (from 21.1+/-7.6 mm Hg to 26.1+/-6.5 mm Hg; p = .006), left atrial (from 8.4+/-3.4 mm Hg to 11.5+/-3.7 mm Hg; p < .001), and right atrial pressures (from 7.3+/-2.5 mm Hg to 9.8+/-2.5 mm Hg; p < .001). In addition, mean airway pressure (from 7.4+/-2.0 mm Hg to 8.6+/-2.4 mm Hg; p < .001) and peak inspiratory pressure (from 29.3+/-5.4 mm Hg to 31.3+/-5.6 mm Hg; p = .004) increased. Sternal wound infection occurred in one patient. CONCLUSIONS: Delayed sternal closure is an effective approach to the management of neonates and infants at risk for hemodynamic, respiratory, or hemostatic instability early after cardiac surgery. Significant changes in hemodynamics and respiratory variables occur during sternal closure, often requiring adjustment of inotropic and ventilatory management. (Crit Care Med 2000; 28: 1180-1184)


Subject(s)
Cardiac Surgical Procedures , Postoperative Care/methods , Sternum/surgery , Cardiac Surgical Procedures/methods , Cardiac Surgical Procedures/mortality , Cardiac Surgical Procedures/statistics & numerical data , Cardiopulmonary Bypass , Combined Modality Therapy , Hospital Mortality , Humans , Infant , Infant, Newborn , Postoperative Care/statistics & numerical data , Respiration, Artificial , Retrospective Studies , Time Factors , Treatment Outcome
12.
J Am Coll Cardiol ; 35(2): 463-7, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10676695

ABSTRACT

OBJECTIVE: To determine the effect of pulmonary blood flow (Qp) on nitric oxide (NO) production in patients with increased Qp due to an atrial septal defect (ASD). BACKGROUND: Alterations in pulmonary vascular NO production have been implicated in the development of pulmonary hypertension secondary to increased Qp. In vitro, acute changes in flow or shear stress alter NO production. However, the effect of Qp on lung NO production in vivo is unclear. METHODS: Nineteen patients (2.4-61 years of age, median 17) with secundum ASD undergoing device closure were studied. Before, and 30 min after ASD closure, exhaled NO and plasma nitrate concentration were measured by chemiluminescence (NOA 280, Sievers, Boulder, Colorado). RESULTS: Before ASD closure, all patients had increased Qp (Qp: systemic blood flow [Qs] of 2.0 +/- 0.7) and normal mean pulmonary arterial pressure (13.4 +/- 3.1 mm Hg). Atrial septal defect device closure decreased Qp from 6.0 +/- 2.5 to 3.6 +/- 1.3 L/min/m2 (p < 0.05). Mean pulmonary arterial pressure was unchanged. Associated with the decrease in Qp, both exhaled NO (-22.1%, p < 0.05) and plasma nitrate concentrations (-17.9%, p < 0.05) decreased. CONCLUSIONS: These data represent the first demonstration that acute changes in Qp alter pulmonary NO production in vivo in humans. Exhaled NO determinations may provide a noninvasive assessment of pulmonary vascular NO production in patients with congenital heart disease. Potential correlations between exhaled NO, pulmonary vascular reactivity and pulmonary hypertension warrant further study.


Subject(s)
Cardiac Surgical Procedures/instrumentation , Heart Septal Defects, Atrial/surgery , Nitric Oxide/biosynthesis , Pulmonary Circulation/physiology , Adolescent , Adult , Biomarkers/blood , Blood Flow Velocity , Cardiac Catheterization , Child , Child, Preschool , Female , Heart Septal Defects, Atrial/blood , Heart Septal Defects, Atrial/physiopathology , Humans , Male , Middle Aged , Nitric Oxide/blood , Prognosis , Pulmonary Artery/physiopathology , Pulmonary Wedge Pressure
13.
Pediatr Res ; 47(1): 97-106, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10625089

ABSTRACT

Plasma concentrations of endothelin-1 (ET-1) are increased in children with congenital heart disease associated with increased pulmonary blood flow. However, the role of ET-1 in the pathophysiology of pulmonary hypertension remains unclear. Preproendothelin-1 gene expression is increased in adults with advanced pulmonary hypertension. To characterize potential early molecular alterations in the ET-1 cascade induced by increased pulmonary blood flow and pulmonary hypertension, fetal lambs underwent in utero placement of an aortopulmonary vascular graft (shunt). RNase protection assays and Western blot analysis were performed on lung tissue prepared from 4-wk-old shunt lambs and age-matched controls. Endothelin-converting enzyme-1 [the enzyme responsible for the production of active ET-1 from big ET-1, mRNA (411%, p<0.05)] and protein (170%, p<0.05) were increased in lung tissue prepared from shunt lambs, compared with age-matched controls. Endothelin type A receptor (the receptor that mediates vasoconstriction), mRNA (246%, p<0.05), and protein (176%, p<0.05) also were increased in lung tissue prepared from shunt lambs compared with age-matched controls. Conversely, endothelin type B receptor (the receptor that mediates vasodilation), mRNA (46%, p<0.05), and protein (65%, p<0.05) were decreased in shunt lambs. Both the mRNA and protein levels for preproendothelin-were unchanged. Thus we conclude that increased pulmonary blood flow and pulmonary hypertension induce early alterations in the ET-1 cascade that result in increased ET-1 production, increased ET-1-mediated vasoconstriction, and decreased vasodilation. These early alterations in gene expression may contribute to the development of pulmonary hypertension and its associated enhanced pulmonary vascular reactivity.


Subject(s)
Endothelin-1/blood , Hypertension, Pulmonary/metabolism , Lung/blood supply , Animals , Base Sequence , DNA Primers , Endothelin-1/biosynthesis , Endothelin-1/genetics , Female , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Endothelin/genetics , Regional Blood Flow , Sheep
14.
Am J Physiol ; 277(5): H1849-56, 1999 11.
Article in English | MEDLINE | ID: mdl-10564139

ABSTRACT

Life-threatening increases in pulmonary vascular resistance have been noted on acute withdrawal of inhaled nitric oxide (NO), although the mechanisms remain unknown. In vitro data suggest that exogenous NO exposure inhibits endothelial NO synthase (NOS) activity. Thus the objectives of this study were to determine the effects of inhaled NO therapy and its acute withdrawal on endogenous NOS activity and gene expression in vivo in the intact lamb. Six 1-mo-old lambs were mechanically ventilated and instrumented to measure vascular pressures and left pulmonary blood flow. Inhaled NO (40 ppm) acutely decreased left pulmonary vascular resistance by 27. 5 +/- 4.7% (P < 0.05). This was associated with a 207% increase in plasma cGMP concentrations (P < 0.05). After 6 h of inhaled NO, NOS activity was reduced to 44.3 +/- 5.9% of pre-NO values (P < 0.05). After acute withdrawal of NO, pulmonary vascular resistance increased by 52.1 +/- 11.6% (P < 0.05) and cGMP concentrations decreased. Both returned to pre-NO values within 60 min. One hour after NO withdrawal, NOS activity increased by 48.4 +/- 19.1% to 70% of pre-NO values (P < 0.05). Western blot analysis revealed that endothelial NOS protein levels remained unchanged throughout the study period. These data suggest a role for decreased endogenous NOS activity in the rebound pulmonary hypertension noted after acute withdrawal of inhaled NO.


Subject(s)
Enzyme Inhibitors/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide/pharmacology , Administration, Inhalation , Animals , Cyclic GMP/blood , Endothelium, Vascular/enzymology , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/adverse effects , Gene Expression/drug effects , Hypertension, Pulmonary/chemically induced , Nitric Oxide/administration & dosage , Nitric Oxide/adverse effects , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Pulmonary Circulation/drug effects , Sheep , Substance Withdrawal Syndrome , Time Factors , Vascular Resistance/drug effects
15.
Artif Organs ; 23(11): 970-4, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10564299

ABSTRACT

Congenital heart disease with increased pulmonary blood flow commonly leads to the development of pulmonary hypertension and increased vascular reactivity. These serious sequelae are associated with the following two major categories of congenital heart defects: those resulting in increased pulmonary blood flow and increased pulmonary arterial pressure and those resulting in increased pulmonary venous pressure. Recent evidence that the pulmonary vascular endothelium is an important determinant of vascular tone has led to the hypothesis that endothelial injury, secondary to congenital heart disease with increased pulmonary blood flow, disrupts these regulatory mechanisms and thereby plays a role in the development of pulmonary hypertension and its associated increased vascular reactivity. In many animal models, endothelial dysfunction is a precursor for smooth muscle dysfunction, and there is an apparent progression from endothelial dysfunction to smooth muscle dysfunction as vascular changes progress. We established a chronic model of pulmonary hypertension with increased pulmonary blood flow in young lambs by placing a systemic-to-pulmonary shunt in utero. In this model, we found significant physiologic and molecular alternations of both the nitric oxide (NO) and endothelin signaling pathways, two important mechanisms by which the endothelium regulates pulmonary vascular tone. These alterations occur extremely early and precede severe anatomic changes. Early endothelial damage may contribute to the development of pulmonary hypertension and its associated enhanced pulmonary vascular reactivity.


Subject(s)
Heart Defects, Congenital/physiopathology , Hypertension, Pulmonary/physiopathology , Animals , Blood Pressure/physiology , Cyclic GMP/physiology , Disease Models, Animal , Endothelin-1/physiology , Endothelium, Vascular/physiopathology , Heart Defects, Congenital/complications , Hypertension, Pulmonary/etiology , Muscle, Smooth, Vascular/physiopathology , Nitric Oxide/physiology , Pulmonary Circulation/physiology , Sheep , Signal Transduction/physiology , Vasodilator Agents/pharmacology , Vasomotor System/physiopathology , Venous Pressure/physiology
16.
Pediatr Pulmonol ; 28(1): 3-11, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10406044

ABSTRACT

Inhaled nitric oxide (NO) is currently used as an adjuvant therapy for a variety of pulmonary hypertensive disorders. In both animal and human studies, inhaled NO induces selective, dose-dependent pulmonary vasodilation. However, its potential interactions with other simultaneously used pulmonary vasodilator therapies have not been studied. Therefore, the objective of this study was to determine the potential dose-response interactions of inhaled NO, oxygen, and alkalosis therapies. Fourteen newborn lambs (age 1-6 days) were instrumented to measure vascular pressures and left pulmonary artery blood flow. After recovery, the lambs were sedated and mechanically ventilated. During steady-state pulmonary hypertension induced by U46619 (a thromboxane A2 mimic), the lambs were exposed to the following conditions: Protocol A, inhaled NO (0, 5, 40, and 80 ppm) and inspired oxygen concentrations (FiO2) of 0.21, 0.50, and 1.00; and Protocol B, inhaled NO (0, 5, 40, and 80 ppm) and arterial pH levels of 7.30, 7.40, 7.50, and 7.60. Each condition (in randomly chosen order) was maintained for 10 min, and all variables were allowed to return to baseline between conditions. Inhaled NO, oxygen, and alkalosis produced dose-dependent decreases in mean pulmonary arterial pressures (P < 0.05). Systemic arterial pressure remained unchanged. At 5 ppm of inhaled NO, alkalosis and oxygen induced further dose-dependent decreases in mean pulmonary arterial pressures (P < 0.05). At inhaled NO doses > 5 ppm, alkalosis induced further dose-independent decreases in mean pulmonary arterial pressure, while oxygen did not. We conclude that in this animal model, oxygen, alkalosis, and inhaled NO induced selective, dose-dependent pulmonary vasodilation. However, when combined, a systemic arterial pH > 7.40 augmented inhaled NO-induced pulmonary vasodilation, while an FiO2 > 0.5 did not. Therefore, weaning high FiO2 during inhaled NO therapy should be considered, since it may not diminish the pulmonary vasodilating effects. Further studies are warranted to guide the clinical weaning strategies of these pulmonary vasodilator therapies.


Subject(s)
Alkalosis/etiology , Hypertension, Pulmonary/drug therapy , Nitric Oxide/administration & dosage , Oxygen/administration & dosage , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid , Administration, Inhalation , Alkalosis/physiopathology , Analysis of Variance , Animals , Animals, Newborn , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Hydrogen-Ion Concentration/drug effects , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/therapy , Linear Models , Respiratory Function Tests , Sheep , Treatment Outcome , Vasodilation/drug effects , Vasodilation/physiology
17.
Pediatr Pulmonol ; 27(3): 147-56, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10213252

ABSTRACT

Acute lung injury produces pulmonary hypertension, altered vascular reactivity, and endothelial injury. To determine whether acute lung injury impairs the endothelium-dependent regulation of pulmonary vascular tone, 16 lambs were studied during U46619-induced pulmonary hypertension without acute lung injury, or air embolization-induced pulmonary hypertension with acute lung injury. The hemodynamic responses to endothelium-dependent (acetylcholine, ATP, ET-1, and 4 Ala ET-1 [an ETb receptor agonist]) and endothelium-independent (nitroprusside and isoproterenol) vasodilators were compared. During U46619-induced pulmonary hypertension, all vasodilators decreased pulmonary arterial pressure and vascular resistance (P < 0.05). During air embolization-induced pulmonary hypertension, the pulmonary vasodilating effects of acetylcholine, ATP, and 4 Ala ET-1 were attenuated (P < 0.05); the pulmonary vasodilating effects of nitroprusside and isoproterenol were unchanged; and the pulmonary vasodilating effects of ET-1 were reversed, producing pulmonary vasoconstriction (P < 0.05). During air embolization, the pulmonary vasoconstricting effects of ET-1 were blocked by BQ 123, an ETa receptor antagonist. The systemic effects of the vasoactive drugs were similar during both conditions. We conclude that pulmonary hypertension with acute lung injury induced by air embolization results in endothelial dysfunction; there is selective impairment of endothelium-dependent pulmonary vasodilation and an altered response to ET-1 from pulmonary vasodilation to vasoconstriction. This altered response to ET-1 is associated with decreased ETb receptor-mediated vasodilation and increased ETa receptor-mediated vasoconstriction. Endothelial injury and dysfunction account, in part, for the altered regulation of pulmonary vascular tone during pulmonary hypertension with acute lung injury.


Subject(s)
Endothelium, Vascular/physiopathology , Hypertension, Pulmonary/physiopathology , Lung Injury , Lung/physiopathology , Pulmonary Circulation/physiology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid , Acute Disease , Animals , Disease Models, Animal , Embolism, Air , Endothelin-1/metabolism , Hemodynamics/drug effects , Hemodynamics/physiology , Hypertension, Pulmonary/chemically induced , Nitric Oxide/metabolism , Pulmonary Circulation/drug effects , Random Allocation , Reference Values , Respiration, Artificial , Sheep , Vascular Resistance , Vasoconstrictor Agents , Vasodilator Agents/pharmacology
18.
J Thorac Cardiovasc Surg ; 117(2): 314-23, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9918974

ABSTRACT

BACKGROUND: Children with increased pulmonary blood flow may experience morbidity as the result of increased pulmonary vascular resistance after operations in which cardiopulmonary bypass is used. Plasma levels of endothelin-1, a potent vasoactive substance implicated in pulmonary hypertension, are increased after cardiopulmonary bypass. OBJECTIVES: In a lamb model of increased pulmonary blood flow after in utero placement of an aortopulmonary shunt, we characterized the changes in pulmonary vascular resistance induced by hypothermic cardiopulmonary bypass and investigated the role of endothelin-1 and endothelin-A receptor activation in postbypass pulmonary hypertension. METHODS: In eleven 1-month-old lambs, the shunt was closed, and vascular pressures and blood flows were monitored. An infusion of a selective endothelin-A receptor blocker (PD 156707; 1.0 mg/kg/h) or drug vehicle (saline solution) was then begun 30 minutes before cardiopulmonary bypass and continued for 4 hours after bypass. The hemodynamic variables were monitored, and plasma endothelin-1 concentrations were determined before, during, and for 6 hours after cardiopulmonary bypass. RESULTS: After 90 minutes of hypothermic cardiopulmonary bypass, both pulmonary arterial pressure and pulmonary vascular resistance increased significantly in saline-treated lambs during the 6-hour study period (P <.05). In lambs pretreated with PD 156707, pulmonary arterial pressure and pulmonary vascular resistance decreased (P <. 05). After bypass, plasma endothelin-1 concentrations increased in all lambs; there was a positive correlation between postbypass pulmonary vascular resistance and plasma endothelin-1 concentrations (P <.05). CONCLUSIONS: This study suggests that endothelin-A receptor-induced pulmonary vasoconstriction mediates, in part, the rise in pulmonary vascular resistance after cardiopulmonary bypass. Endothelin-A receptor antagonists may decrease morbidity in children at risk for postbypass pulmonary hypertension. This potential therapy warrants further investigation.


Subject(s)
Cardiopulmonary Bypass , Endothelin Receptor Antagonists , Pulmonary Artery/physiology , Pulmonary Circulation/physiology , Vascular Resistance/physiology , Analysis of Variance , Animals , Cardiopulmonary Bypass/instrumentation , Cardiopulmonary Bypass/methods , Dioxoles/pharmacology , Disease Models, Animal , Endothelin-1/blood , Endothelin-1/drug effects , Female , Fetus , Hemodynamics/drug effects , Hemodynamics/physiology , Hypertension, Pulmonary/blood , Hypertension, Pulmonary/physiopathology , Hypertension, Pulmonary/surgery , Linear Models , Pregnancy , Pulmonary Artery/drug effects , Pulmonary Circulation/drug effects , Receptors, Endothelin/drug effects , Receptors, Endothelin/physiology , Sheep , Time Factors , Vascular Resistance/drug effects
19.
J Thorac Cardiovasc Surg ; 117(2): 343-51, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9918976

ABSTRACT

BACKGROUND: Fetal cardiac bypass causes placental dysfunction, characterized by increased placental vascular resistance, decreased placental blood flow, hypoxia, and acidosis. Vasoactive factors produced by the vascular endothelium, such as nitric oxide and endothelin 1, are important regulators of placental vascular tone and may contribute to this placental dysfunction. METHODS: To investigate the role of the vascular endothelium in placental dysfunction related to fetal cardiac bypass, we studied 3 groups of fetal sheep. In the first group (n = 7) we determined placental hemodynamic responses before and after bypass to an endothelium-dependent vasodilator (acetylcholine), an endothelium-independent vasodilator (nitroprusside), and endothelin 1. In the second group (n = 8) a nonspecific endothelin receptor blocker (PD 145065) was administered and placental hemodynamic values were measured before and after bypass. In the third group (n = 5) endothelin 1 levels were measured before and after bypass. RESULTS: Before fetal cardiac bypass exogenous endothelin 1 decreased placental blood flow by 9% and increased placental resistance by 9%. After bypass endothelin 1 decreased placental flow by 47% and increased resistance by 106%. There was also a significant attenuation of the placental vascular relaxation response to acetylcholine after bypass, whereas the response to nitroprusside was not significantly altered. In fetuses that received the PD 145065, placental vascular resistance increased significantly less than in control fetuses (28% versus 62%). Similarly, placental blood flow decreased significantly more (from 6. 3 +/- 3.1 to 28.3 +/- 10.4 pg/mL; P =.01) in control fetuses than in fetuses receiving PD 145065 (33% versus 20%). Umbilical venous endothelin 1 levels increased significantly in fetuses exposed to fetal bypass but did not change in control fetuses. CONCLUSIONS: The basal endothelial regulatory mechanisms of placental vascular tone were deranged after fetal cardiac bypass. Endothelin receptor blockade, which substantially reduced postbypass placental dysfunction, and other interventions aimed at preserving endothelial function may be effective means of optimizing fetal outcome after cardiac bypass.


Subject(s)
Cardiac Surgical Procedures , Endothelium, Vascular/physiopathology , Fetal Heart/surgery , Placenta Diseases/physiopathology , Placenta/physiopathology , Postoperative Complications/physiopathology , Acetylcholine/pharmacology , Analysis of Variance , Animals , Cardiac Surgical Procedures/methods , Endothelin Receptor Antagonists , Endothelin-1/pharmacology , Endothelium, Vascular/drug effects , Female , Hemodynamics , Nitroprusside/pharmacology , Oligopeptides/pharmacology , Placenta/drug effects , Pregnancy , Sheep , Vasodilator Agents/pharmacology
20.
J Perinatol ; 19(6 Pt 1): 441-6, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10685275

ABSTRACT

OBJECTIVE: Alveolar capillary dysplasia is a rare cause of persistent pulmonary hypertension of the newborn. Infants with this condition die despite maximal medical intervention including inhaled nitric oxide therapy and extracorporeal membrane oxygenation. To date, diagnosis of this lethal condition was made by open lung biopsy or during postmortem examination. We examined the possibility that distinct cardiac catheterization findings could be used in the diagnosis of this lethal disorder. STUDY DESIGN: We present three infants with fatal persistent pulmonary hypertension of the newborn refractory to extracorporeal membrane oxygenation and inhaled nitric oxide therapy, two with postmortem autopsy confirmation of alveolar capillary dysplasia. Each infant underwent cardiac catheterization to complete the diagnostic evaluations. RESULTS: Significant right ventricular hypertension and normal pulmonary venous return were demonstrated, but a markedly diminished or absent capillary blush phase was noted in each infant. This finding is distinct from the normal capillary blush seen in infants with persistent pulmonary hypertension of the newborn of other etiologies. CONCLUSION: Cardiac catheterization may provide a useful alternative to tissue examination in the diagnosis of alveolar capillary dysplasia.


Subject(s)
Cardiac Catheterization , Pulmonary Alveoli/blood supply , Angiography , Capillaries/abnormalities , Extracorporeal Membrane Oxygenation , Fatal Outcome , Female , Humans , Infant, Newborn , Lung/pathology , Male , Nitric Oxide/therapeutic use , Persistent Fetal Circulation Syndrome/diagnosis , Persistent Fetal Circulation Syndrome/etiology , Persistent Fetal Circulation Syndrome/physiopathology , Persistent Fetal Circulation Syndrome/therapy , Pulmonary Artery/diagnostic imaging , Pulmonary Veins/physiopathology , Ventricular Function, Right
SELECTION OF CITATIONS
SEARCH DETAIL
...