Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
J Perinatol ; 38(6): 636-638, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29467523

RESUMEN

TYPE OF INVESTIGATION: Prognosis; exploratory secondary analysis of an interventional randomized controlled trial. QUESTION: In extremely preterm infant (<28 weeks), is early low-dose hydrocortisone compared to placebo associated with neurodevelopmental impairment at 2 years of age? METHODS: Patients: Surviving infants enrolled in the PREMILOC trial conducted in France between 2008 and 2014. INTERVENTION: Double-blind, multicenter, randomized, placebo-controlled trial of infants born between 24 0/7 weeks and 27 6/7 weeks of gestation and before 24 h of postnatal age, assigned to receive either placebo or low-dose hydrocortisone (0.5 mg/kg twice per day for 7 days, followed by 0.5 mg/kg per day for 3 days). MAIN RESULTS: For the pre-specified exploratory outcome, the distribution of patients without neurodevelopmental impairment (73% in the hydrocortisone group vs. 70% in the placebo group), with mild neurodevelopmental impairment (20% in the hydrocortisone group vs. 18% in the placebo group), or with moderate to severe neurodevelopmental impairment (7% in the hydrocortisone group vs. 11% in the placebo group) was not found to be statistically significantly different between the two groups (p = 0.33). Qualitative assessment of patients using standardized neurological examination also was not statistically significantly different between groups (p = 0.87). STUDY CONCLUSION: In this follow-up study of premature infants who were randomly assigned at birth to receive low-dose hydrocortisone or placebo for 10 days, hydrocortisone treatment was not associated with any adverse effects on neurodevelopmental outcome at 22 months of corrected age.


Asunto(s)
Displasia Broncopulmonar/tratamiento farmacológico , Dexametasona/uso terapéutico , Hidrocortisona/uso terapéutico , Mortalidad Infantil/tendencias , Recien Nacido Extremadamente Prematuro , Trastornos del Neurodesarrollo/tratamiento farmacológico , Displasia Broncopulmonar/mortalidad , Displasia Broncopulmonar/prevención & control , Cuidados Críticos/métodos , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Esquema de Medicación , Femenino , Francia , Humanos , Lactante , Recién Nacido , Unidades de Cuidado Intensivo Neonatal , Modelos Logísticos , Masculino , Trastornos del Neurodesarrollo/diagnóstico , Examen Neurológico/métodos , Embarazo
2.
PLoS One ; 12(8): e0180957, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28792962

RESUMEN

Bronchopulmonary dysplasia (BPD), a common complication of preterm birth, is associated with pulmonary hypertension (PH) in 25% of infants with moderate to severe BPD. Neonatal mice exposed to hyperoxia for 14d develop lung disease similar to BPD, with evidence of associated PH. The cyclic guanosine monophosphate (cGMP) signaling pathway has not been well studied in BPD-associated PH. In addition, there is little data about the natural history of hyperoxia-induced PH in mice or the utility of phosphodiesterase-5 (PDE5) inhibition in established disease. C57BL/6 mice were placed in room air or 75% O2 within 24h of birth for 14d, followed by recovery in room air for an additional 7 days (21d). Additional pups were treated with either vehicle or sildenafil for 7d during room air recovery. Mean alveolar area, pulmonary artery (PA) medial wall thickness (MWT), RVH, and vessel density were evaluated at 21d. PA protein from 21d animals was analyzed for soluble guanylate cyclase (sGC) activity, PDE5 activity, and cGMP levels. Neonatal hyperoxia exposure results in persistent alveolar simplification, RVH, decreased vessel density, increased MWT, and disrupted cGMP signaling despite a period of room air recovery. Delayed treatment with sildenafil during room air recovery is associated with improved RVH and decreased PA PDE5 activity, but does not have significant effects on alveolar simplification, PA remodeling, or vessel density. These data are consistent with clinical studies suggesting inconsistent effects of sildenafil treatment in infants with BPD-associated PH.


Asunto(s)
Displasia Broncopulmonar/patología , Hiperoxia/patología , Hipertensión Pulmonar/patología , Oxígeno/metabolismo , Inhibidores de Fosfodiesterasa 5/farmacología , Citrato de Sildenafil/farmacología , Animales , Animales Recién Nacidos , GMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/metabolismo , Modelos Animales de Enfermedad , Guanilato Ciclasa/metabolismo , Hipertrofia Ventricular Derecha/patología , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Alveolos Pulmonares/fisiología , Arteria Pulmonar/fisiología , Transducción de Señal , Remodelación Vascular
3.
PLoS One ; 12(7): e0180384, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28671996

RESUMEN

To investigate the impact of photoreceptor oxidative stress on photoreceptor degeneration in mice carrying the rd8 mutation (C57BL/6N). We compared the hyperoxia-induced proliferative retinopathy (HIPR) model in two mouse strains (C57BL/6J and C57BL/6N). Pups were exposed to 75% oxygen, starting at birth and continuing for 14 days (P14). Mice were euthanized at P14, or allowed to recover in room air for one day (P15), seven days (P21), or 14 days (P28). We quantified retinal thickness and the length of residual photoreceptors not affected by rosette formation. In addition we explored differences in retinal immunostaining for NADPH oxidase 4 (NOX4), Rac1, vascular endothelium, and activated Mϋller cells. We analyzed photoreceptor oxidative stress using DCF staining in cross sections and quantified NOX4 protein levels using western blotting. C57BL/6N mice in HIPR showed increased oxidative stress, NOX4, and Rac1 in the photoreceptors at P14 and P15 compared to C57BL/6J. In addition, we observed significant progression of photoreceptor degeneration, with significantly accelerated rosette formation in C57BL/6N under HIPR, compared to their room air counterparts. Furthermore, C57BL/6N under HIPR had significantly thinner central retinas than C57BL/6J in HIPR. We did not find a difference in vascular disruption or Mϋller cell activation comparing the two strains in hyperoxia. In HIPR, the C57BL/6N strain carrying the rd8 mutation showed significantly accelerated photoreceptor degeneration, mediated via exacerbated photoreceptor oxidative stress, which we believe relates to Rac1-NOX dysregulation in the setting of Crb1 loss-of-function.


Asunto(s)
Hiperoxia/metabolismo , Estrés Oxidativo , Células Fotorreceptoras/metabolismo , Enfermedades de la Retina/metabolismo , Animales , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Mutación , Neovascularización Patológica , Especies Reactivas de Oxígeno/metabolismo
4.
Am J Pathol ; 187(2): 401-417, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27939131

RESUMEN

Necrotizing enterocolitis (NEC) is a devastating intestinal disease that has been associated with Cronobacter sakazakii and typically affects premature infants. Although NEC has been actively investigated, little is known about the mechanisms underlying the pathophysiology of epithelial injury and intestinal barrier damage. Cyclic adenosine monophosphate (cAMP) and protein kinase A (PKA) are important mediators and regulators of apoptosis. To test the hypothesis that C. sakazakii increases cAMP and PKA activation in experimental NEC resulting in increased epithelial apoptosis, we investigated the effects of C. sakazakii on cAMP and PKA in vitro and in vivo. Specifically, rat intestinal epithelial cells and a human intestinal epithelial cell line were infected with C. sakazakii, and cAMP levels and phosphorylation of PKA were measured. An increase in cAMP was demonstrated after infection, as well as an increase in phosphorylated PKA. Similarly, increased intestinal cAMP and PKA phosphorylation were demonstrated in a rat pup model of NEC. These increases were correlated with increased intestinal epithelial apoptosis. The additional of a PKA inhibitor (KT5720) significantly ameliorated these effects and decreased the severity of experimental NEC. Findings were compared with results from human tissue samples. Collectively, these observations indicate that cAMP and PKA phosphorylation are associated with increased apoptosis in NEC and that inhibition of PKA activation protects against apoptosis and experimental NEC.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Enterocolitis Necrotizante/metabolismo , Animales , Apoptosis/fisiología , Western Blotting , Cronobacter sakazakii , Modelos Animales de Enfermedad , Enterocolitis Necrotizante/patología , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Reacción en Cadena de la Polimerasa , Ratas , Ratas Sprague-Dawley
5.
PLoS One ; 11(11): e0166886, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27861592

RESUMEN

Bronchopulmonary dysplasia (BPD) is a major cause of neonatal morbidity in premature infants, occurring as a result of arrested lung development combined with multiple postnatal insults. Infants with BPD exposed to supplemental oxygen are at risk of retinopathy of prematurity as well. Thus, we studied the effects of hyperoxia on the retinal vasculature in a murine model of BPD. The retinal phenotype of this model, which we termed hyperoxia-induced proliferative retinopathy (HIPR), shows severe disruption of retinal vasculature and loss of vascular patterning, disorganized intra-retinal angiogenesis, inflammation and retinal detachment. Neonatal mice were subjected to 75% oxygen exposure from postnatal day (P)0 to P14 to model BPD, then allowed to recover in room air for 1 (P15), 7 (P21), or 14 days (P28). We quantified retinal thickness, protein levels of HIF-1α, NOX2, and VEGF, and examined the cellular locations of these proteins by immunohistochemistry. We examined the retinal blood vessel integrity and inflammatory markers, including macrophages (F4/80) and lymphocytes (CD45R). Compared to controls, normal retinal vascular development was severely disrupted and replaced by a disorganized sheet of intra-retinal angiogenesis in the HIPR mice. At all time-points, HIPR showed persistent hyaloidal vasculature and a significantly thinner central retina compared to controls. HIF-1α protein levels were increased at P15, while VEGF levels continued to increase until P21. Intra-retinal fibrinogen was observed at P21 followed by sub-retinal deposition in at P28. Inflammatory lymphocytes and macrophages were observed at P21 and P28, respectively. This model presents a severe phenotype of disrupted retinal vascular development, intra-retinal angiogenesis inflammation and retinal detachment.


Asunto(s)
Hiperoxia/metabolismo , Vitreorretinopatía Proliferativa/etiología , Vitreorretinopatía Proliferativa/metabolismo , Animales , Colágeno/metabolismo , Modelos Animales de Enfermedad , Expresión Génica , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inmunohistoquímica , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , NADPH Oxidasa 2 , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Oxígeno/metabolismo , Retina/metabolismo , Retina/patología , Células Ganglionares de la Retina/metabolismo , Neovascularización Retiniana/metabolismo , Neovascularización Retiniana/patología , Vasos Retinianos/metabolismo , Vasos Retinianos/patología , Retinitis/genética , Retinitis/metabolismo , Retinitis/patología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Vitreorretinopatía Proliferativa/patología
6.
Pediatr Res ; 80(5): 635-640, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27434118

RESUMEN

Understanding mechanisms of childhood disease and development of rational therapeutics are fundamental to progress in pediatric intensive care specialties. However, Division Chiefs and Department Chairs face unique challenges when building effective laboratory-based research programs in Neonatal and Pediatric Intensive Care, owing to high clinical demands necessary to maintain competence as well as financial pressures arising from fund flow models and the current extramural funding climate. Given these factors, the role of institutional support that could facilitate successful transition of promising junior faculty to independent research careers is ever more important. Would standardized guidelines of such support provide greater consistency among institutions? We addressed preliminary questions during a national focus group, a workshop and a survey of junior and senior academicians to solicit recommendations for optimal levels of protected time and resources when starting an independent laboratory. The consensus was that junior faculty should be assigned no more than 8 wk clinical service and should obtain start-up funds of $500K-1M exclusive of a 5-y committed salary support. Senior respondents placed a higher premium on protected time than junior faculty.


Asunto(s)
Cuidados Críticos , Neonatología , Pediatría , Médicos , Investigación Biomédica Traslacional , Centros Médicos Académicos/organización & administración , Selección de Profesión , Cuidados Críticos/organización & administración , Grupos Focales , Guías como Asunto , Hospitales Pediátricos/organización & administración , Humanos , Satisfacción en el Trabajo , Cuerpo Médico de Hospitales , Mentores , Neonatología/organización & administración , Pediatría/organización & administración , Desarrollo de Programa , Encuestas y Cuestionarios , Investigación Biomédica Traslacional/organización & administración , Recursos Humanos
7.
Pediatr Res ; 79(5): 759-65, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26756781

RESUMEN

BACKGROUND: Exposure of neonatal mice to hyperoxia results in pulmonary vascular remodeling and aberrant phosphodiesterase type 5 (PDE5) signaling. Although glucocorticoids are frequently utilized in the NICU, little is known about their effects on the developing pulmonary vasculature and on PDE5. We sought to determine the effects of hydrocortisone (HC) on pulmonary vascular development and on PDE5 in a neonatal mouse model of hyperoxic lung injury. METHODS: C57BL/6 mice were placed in 21% O2 or 75% O2 within 24 h of birth and received HC (1, 5, or 10 mg/kg subcutaneously every other day) or vehicle. At 14 d, right ventricular hypertrophy (RVH), medial wall thickness (MWT), lung morphometry, and pulmonary artery (PA) PDE5 activity were assessed. PDE5 activity was measured in isolated pulmonary artery smooth muscle cells exposed to 21 or 95% O2 ± 100 nmol/l HC for 24 h. RESULTS: Hyperoxia resulted in alveolar simplification, RVH, increased MWT, and increased PA PDE5 activity. HC decreased hyperoxia-induced RVH and attenuated MWT. HC had dose-dependent effects on alveolar simplification. HC decreased hyperoxia-induced PDE5 activity both in vivo and in vitro. CONCLUSIONS: HC decreases hyperoxia-induced pulmonary vascular remodeling and attenuates PDE5 activity. These findings suggest that HC may protect against hyperoxic injury in the developing pulmonary vasculature.


Asunto(s)
Glucocorticoides/farmacología , Hidrocortisona/farmacología , Hiperoxia/patología , Lesión Pulmonar/patología , Pulmón/crecimiento & desarrollo , Animales , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/metabolismo , Relación Dosis-Respuesta a Droga , Elastina/metabolismo , Humanos , Hiperoxia/metabolismo , Hipertensión Pulmonar/fisiopatología , Hipertrofia Ventricular Derecha/fisiopatología , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Alveolos Pulmonares/metabolismo , Arteria Pulmonar/patología , Transducción de Señal
8.
JPEN J Parenter Enteral Nutr ; 40(6): 890-5, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-25700180

RESUMEN

Peripherally inserted central catheters (PICCs) are widely used in the pediatric population, and their use continues to grow in popularity. These catheters provide a reliable source of venous access to neonatal patients but can also be the cause of life-threatening complications. There are several well-documented complications such as infections, catheter thrombosis, vascular extravasations, and fractured catheters. However, the complication of vascular erosion into the pleural space using both small and silicone-based catheters is rarely described. After obtaining institutional review board approval, we identified 4 cases to review of PICCs complicated by vascular erosions in the past 2 years. Herein, we also review the current literature of PICC complications. Getting the catheter tip as close to the atrial-caval junction as possible and confirmation of this placement are of the utmost importance. The thick wall of the vena cava near the atrium seems to be less likely to perforate; in addition, this position provides increased volume and turbulence to help dilute the hyperosmolar fluid, which seems to also be a factor in this complication. A daily screening chest x-ray in patients with upper extremity PICCs and ongoing parenteral nutrition (PN) are not necessary at this time given the overall low rate of vascular erosion and concerns regarding excessive radiation exposure in pediatric populations. However, a low threshold for chest x-ray imaging in patients with even mild respiratory symptoms in the setting of upper extremity PN is recommended.


Asunto(s)
Vasos Sanguíneos/lesiones , Cateterismo Venoso Central/métodos , Cateterismo Periférico/efectos adversos , Enfermedades del Prematuro , Recien Nacido Prematuro , Enfermedades Vasculares/etiología , Infecciones Relacionadas con Catéteres , Catéteres Venosos Centrales/efectos adversos , Enfermedades en Gemelos , Femenino , Humanos , Recién Nacido , Enfermedades del Prematuro/etiología , Enfermedades del Prematuro/terapia , Masculino , Nutrición Parenteral , Cavidad Pleural , Derrame Pleural/diagnóstico por imagen , Radiografía , Venas Cavas
9.
Am J Physiol Lung Cell Mol Physiol ; 309(4): L369-77, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26092998

RESUMEN

Animal models demonstrate that exposure to supraphysiological oxygen during the neonatal period compromises both lung and pulmonary vascular development, resulting in a phenotype comparable to bronchopulmonary dysplasia (BPD). Our prior work in murine models identified postnatal maturation of antioxidant enzyme capacities as well as developmental regulation of mitochondrial oxidative stress in hyperoxia. We hypothesize that consequences of hyperoxia may also be developmentally regulated and mitochondrial reactive oxygen species (ROS) dependent. To determine whether age of exposure impacts the effect of hyperoxia, neonatal mice were placed in 75% oxygen for 72 h at either postnatal day 0 (early postnatal) or day 4 (late postnatal). Mice exposed to early, but not late, postnatal hyperoxia demonstrated decreased alveolarization and septation, increased muscularization of resistance pulmonary arteries, and right ventricular hypertrophy (RVH) compared with normoxic controls. Treatment with a mitochondria-specific antioxidant, (2-(2,2,6,6-tetramethylpiperidin-1-oxyl-4-ylamino)-2-oxoethyl)triphenylphosphonium chloride (mitoTEMPO), during early postnatal hyperoxia protected against compromised alveolarization and RVH. In addition, early, but not late, postnatal hyperoxia resulted in induction of NOX1 expression that was mitochondrial ROS dependent. Because early, but not late, exposure resulted in compromised lung and cardiovascular development, we conclude that the consequences of hyperoxia are developmentally regulated and decrease with age. Attenuated disease in mitoTEMPO-treated mice implicates mitochondrial ROS in the pathophysiology of neonatal hyperoxic lung injury, with potential for amplification of ROS signaling through NOX1 induction. Furthermore, it suggests a potential role for targeted antioxidant therapy in the prevention or treatment of BPD.


Asunto(s)
Displasia Broncopulmonar/enzimología , Hiperoxia/enzimología , Animales , Inducción Enzimática , Hipertrofia Ventricular Derecha/enzimología , Hipertrofia Ventricular Derecha/etiología , Pulmón/enzimología , Pulmón/crecimiento & desarrollo , Pulmón/patología , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , NADH NADPH Oxidorreductasas/genética , NADH NADPH Oxidorreductasas/metabolismo , NADPH Oxidasa 1 , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/metabolismo
10.
Am J Physiol Heart Circ Physiol ; 308(12): H1575-82, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-25862831

RESUMEN

Pulmonary hypertension (PH) and right ventricular hypertrophy (RVH) affect 25-35% of premature infants with significant bronchopulmonary dysplasia (BPD), increasing morbidity and mortality. We sought to determine the role of phosphodiesterase 5 (PDE5) in the right ventricle (RV) and left ventricle (LV) in a hyperoxia-induced neonatal mouse model of PH and RVH. After birth, C57BL/6 mice were placed in room air (RA) or 75% O2 (CH) for 14 days to induce PH and RVH. Mice were euthanized at 14 days or recovered in RA for 14 days or 42 days prior to euthanasia at 28 or 56 days of age. Some pups received sildenafil or vehicle (3 mg·kg(-1)·dose(-1) sc) every other day from P0. RVH was assessed by Fulton's index [RV wt/(LV + septum) wt]. PDE5 protein expression was analyzed via Western blot, PDE5 activity was measured by commercially available assay, and cGMP was measured by enzyme-linked immunoassay. Hyperoxia induced RVH in mice after 14 days, and RVH did not resolve until 56 days of age. Hyperoxia increased PDE5 expression and activity in RV, but not LV + S, after 14 days. PDE5 expression normalized by 28 days of age, but PDE5 activity did not normalize until 56 days of age. Sildenafil given during hyperoxia prevented RVH, decreased RV PDE5 activity, and increased RV cGMP levels. Mice with cardiac-specific overexpression of PDE5 had increased RVH in RA. These findings suggest normal RV PDE5 function is disrupted by hyperoxia, and elevated PDE5 contributes to RVH and remodeling. Therefore, in addition to impacting the pulmonary vasculature, sildenafil also targets PDE5 in the neonatal mouse RV and decreases RVH.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/metabolismo , Ventrículos Cardíacos/metabolismo , Hiperoxia/complicaciones , Hipertensión Pulmonar/etiología , Hipertrofia Ventricular Derecha/etiología , Sistemas de Mensajero Secundario , Función Ventricular Derecha , Remodelación Ventricular , Animales , Animales Recién Nacidos , Antihipertensivos/farmacología , AMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/genética , Modelos Animales de Enfermedad , Regulación hacia Abajo , Ventrículos Cardíacos/fisiopatología , Hiperoxia/tratamiento farmacológico , Hiperoxia/metabolismo , Hiperoxia/fisiopatología , Hipertensión Pulmonar/tratamiento farmacológico , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/fisiopatología , Hipertrofia Ventricular Derecha/metabolismo , Hipertrofia Ventricular Derecha/fisiopatología , Hipertrofia Ventricular Derecha/prevención & control , Ratones Endogámicos C57BL , Ratones Transgénicos , Inhibidores de Fosfodiesterasa 5/farmacología , Piperazinas/farmacología , Purinas/farmacología , Sistemas de Mensajero Secundario/efectos de los fármacos , Citrato de Sildenafil , Sulfonamidas/farmacología , Factores de Tiempo , Función Ventricular Derecha/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos
11.
Int J Mol Sci ; 16(3): 6373-90, 2015 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-25809610

RESUMEN

Pulmonary hypertension (PH) complicates bronchopulmonary dysplasia (BPD) in 25% of infants. Superoxide dismutase 2 (SOD2) is an endogenous mitochondrial antioxidant, and overexpression protects against acute lung injury in adult mice. Little is known about SOD2 in neonatal lung disease and PH. C57Bl/6 mice and isogenic SOD2+/+ and SOD2-/+ mice were placed in room air (control) or 75% O2 (chronic hyperoxia, CH) for 14 days. Right ventricular hypertrophy (RVH) was assessed by Fulton's index. Medial wall thickness (MWT) and alveolar area were assessed on formalin fixed lung sections. Pulmonary artery smooth muscle cells (PASMC) were placed in 21% or 95% O2 for 24 h. Lung and PASMC protein were analyzed for SOD2 expression and activity. Oxidative stress was measured with a mitochondrially-targeted sensor, mitoRoGFP. CH lungs have increased SOD2 expression, but unchanged activity. SOD2-/+ PASMC have decreased expression and activity at baseline, but increased SOD2 expression in hyperoxia. Hyperoxia increased mitochondrial ROS in SOD2+/+ and SOD2-/+ PASMC. SOD2+/+ and SOD2-/+ CH pups induced SOD2 expression, but not activity, and developed equivalent increases in RVH, MWT, and alveolar area. Since SOD2-/+ mice develop equivalent disease, this suggests other antioxidant systems may compensate for partial SOD2 expression and activity in the neonatal period during hyperoxia-induced oxidative stress.


Asunto(s)
Miocitos del Músculo Liso/metabolismo , Arteria Pulmonar/citología , Arteria Pulmonar/metabolismo , Superóxido Dismutasa/metabolismo , Animales , Hipoxia de la Célula/fisiología , Modelos Animales de Enfermedad , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/fisiopatología , Hipertrofia Ventricular Derecha/metabolismo , Ratones , Ratones Endogámicos C57BL , Oxidación-Reducción
13.
J Clin Endocrinol Metab ; 100(1): E30-40, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25343232

RESUMEN

CONTEXT: Fetal growth restriction with abnormal umbilical artery Doppler velocimetry (FGRadv), reflective of elevated fetoplacental vascular resistance, is associated with increased risks of fetal morbidity and mortality even in comparison to those of growth-restricted fetuses with normal placental blood flow. One major cause of this abnormally elevated fetoplacental vascular resistance is the aberrantly formed, thin, elongated villous vessels that are seen in FGRadv placentas. OBJECTIVE: The purpose of this study was to determine the role of fetoplacental endothelial cells (ECs) in angiogenesis in normal pregnancies and in those complicated by FGRadv. DESIGN AND PARTICIPANTS: Human placental specimens were obtained from FGRadv and gestational age-matched, appropriately grown control pregnancies for EC isolation/culture and for immunohistochemical studies. Additional mechanistic studies were performed on ECs isolated from subjects with term, uncomplicated pregnancies. MAIN OUTCOME MEASURES: We evaluated tube formation and differential angiogenic gene expression in FGRadv and control ECs, and we used ECs from uncomplicated pregnancies to further elucidate the molecular mechanisms by which angiogenesis is impaired in FGRadv pregnancies. RESULTS: Tube formation assays showed that FGRadv ECs demonstrate fewer branch points and total length compared with those from gestational age-matched controls, and this defect was not rescued by exposure to hypoxia. FGRadv ECs also demonstrated lower aryl hydrocarbon receptor nuclear translocator (ARNT) expression. ARNT knockdown resulted in suppression of key angiogenic genes including vascular endothelial growth factor A expression and led to deficient tube formation. CONCLUSIONS: ARNT expression in the placental vasculature mediates key angiogenic expression and fetoplacental EC angiogenesis, and low ARNT expression in FGRadv ECs appears to be a key factor in deficient angiogenesis. This, in turn, results in malformed thin villous vessels that structurally contribute to the abnormally elevated fetoplacental vascular resistance that is associated with high morbidity and mortality in fetal growth restriction.


Asunto(s)
Translocador Nuclear del Receptor de Aril Hidrocarburo/metabolismo , Células Endoteliales/metabolismo , Retardo del Crecimiento Fetal/metabolismo , Neovascularización Fisiológica/fisiología , Placenta/metabolismo , Arterias Umbilicales/diagnóstico por imagen , Femenino , Retardo del Crecimiento Fetal/diagnóstico por imagen , Humanos , Placenta/diagnóstico por imagen , Embarazo , Ultrasonografía , Factor A de Crecimiento Endotelial Vascular/metabolismo , Resistencia Vascular/fisiología
14.
Am J Physiol Lung Cell Mol Physiol ; 307(11): L868-76, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25326578

RESUMEN

Excess superoxide has been implicated in pulmonary hypertension (PH). We previously found lung overexpression of the antioxidant extracellular superoxide dismutase (EC-SOD) attenuates PH and pulmonary artery (PA) remodeling. Although comprising a small fraction of total SOD activity in most tissues, EC-SOD is abundant in arteries. We hypothesize that the selective loss of vascular EC-SOD promotes hypoxia-induced PH through redox-sensitive signaling pathways. EC-SOD(loxp/loxp) × Tg(cre/SMMHC) mice (SMC EC-SOD KO) received tamoxifen to conditionally deplete smooth muscle cell (SMC)-derived EC-SOD. Mice were exposed to hypobaric hypoxia for 35 days, and PH was assessed by right ventricular systolic pressure measurements and right ventricle hypertrophy. Vascular remodeling was evaluated by morphometric analysis and two-photon microscopy for collagen. We examined cGMP content and soluble guanylate cyclase expression and activity in lung, lung phosphodiesterase 5 (PDE5) expression and activity, and expression of endothelial nitric oxide synthase and GTP cyclohydrolase-1 (GTPCH-1), the rate-limiting enzyme in tetrahydrobiopterin synthesis. Knockout of SMC EC-SOD selectively decreased PA EC-SOD without altering total lung EC-SOD. PH and vascular remodeling induced by chronic hypoxia was augmented in SMC EC-SOD KO. Depletion of SMC EC-SOD did not impact content or activity of lung soluble guanylate cyclase or PDE5, yet it blunted the hypoxia-induced increase in cGMP. Although total eNOS was not altered, active eNOS and GTPCH-1 decreased with hypoxia only in SMC EC-SOD KO. We conclude that the localized loss of PA EC-SOD augments chronic hypoxic PH. In addition to oxidative inactivation of NO, deletion of EC-SOD seems to reduce eNOS activity, further compromising pulmonary vascular function.


Asunto(s)
Hipertensión Pulmonar/terapia , Hipoxia/terapia , Superóxido Dismutasa/genética , Animales , Presión Sanguínea , GMP Cíclico/biosíntesis , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/biosíntesis , Antagonistas de Estrógenos/farmacología , GTP Ciclohidrolasa/biosíntesis , Guanilato Ciclasa/biosíntesis , Hipertrofia Ventricular Derecha/fisiopatología , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo III/biosíntesis , Arteria Pulmonar/patología , Receptores Citoplasmáticos y Nucleares/biosíntesis , Transducción de Señal , Guanilil Ciclasa Soluble , Tamoxifeno/farmacología
15.
Antioxid Redox Signal ; 21(13): 1819-22, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25111369

RESUMEN

Respiratory diseases are increasingly recognized as having their origins during perinatal and early postnatal lung development, a time of significant adaptation to large changes in redox conditions as well as to mechanical forces. This Forum of the journal presents a Forum highlighting studies of the interplay between reactive oxygen/nitrogen species and the systems that have evolved to degrade them or exploit them, as well as the cellular repair processes which respond to early life redox stress in the lung. This group of authors suggests new understandings of these events that may point the way to improved therapeutic approaches.


Asunto(s)
Pulmón/metabolismo , Pulmón/patología , Animales , Animales Recién Nacidos , Humanos , Recién Nacido , Oxidación-Reducción , Estrés Oxidativo/fisiología , Especies de Nitrógeno Reactivo/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Enfermedades Respiratorias/metabolismo , Enfermedades Respiratorias/patología
16.
Pulm Circ ; 4(1): 71-81, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25006423

RESUMEN

Phosphodiesterase-5 (PDE5) is the primary phosphodiesterase in the pulmonary vasculature. It degrades cyclic guanosine monophosphate (cGMP) and inhibits cGMP-mediated vasorelaxation. We previously reported that hydrocortisone treatment decreased hyperoxia-induced PDE5 activity and markers of oxidative stress in lambs with persistent pulmonary hypertension of the newborn (PPHN) ventilated with 100% O2. The objective of our study was to determine the molecular mechanism by which hydrocortisone downregulates PDE5 and oxidative stress in fetal pulmonary artery smooth muscle cells (FPASMCs) from PPHN lambs. PPHN FPASMC were incubated for 24 hours in either 21% or 95% O2. Some cells were treated with 100 nM hydrocortisone and/or ±1 µM helenalin, an inhibitor of nuclear factor κ B (NFκB), a redox-sensitive transcription factor. Exposure to hyperoxia led to increased PDE5 activity, oxidative stress, and NFκB activity. Pretreatment of PPHN FPASMC with hydrocortisone normalized PDE5 activity, decreased cytosolic oxidative stress, increased expression of extracellular superoxide dismutase and NFκB inhibitory protein, and decreased NFκB activity. Similarly, treatment with NFκB inhibitor, helenalin, decreased PDE5 activity. These data suggest that hyperoxia activates NFκB, which in turn induces PDE5 activity in PPHN FPASMC, whereas treatment with hydrocortisone attenuates these changes by blocking reactive oxygen species-induced NFκB activity.

17.
Invest Ophthalmol Vis Sci ; 55(3): 1493-501, 2014 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-24519428

RESUMEN

PURPOSE: We sought to determine the effect of sildenafil on retinal vascular changes in a mouse model of oxygen-induced retinopathy (OIR). METHODS: Vascular defects in OIR mice were quantified by measuring vaso-obliteration at postnatal days 12 and 17 (P12 and P17) and neovascularization at P17 to compare sildenafil-treated to dextrose-treated OIR mice. Retinal HIF1α protein expression was quantified by Western blotting and normalized to that of ß-actin. Right ventricular hypertrophy was measured by Fulton's index as a surrogate for hyperoxia-induced pulmonary hypertension. RESULTS: At P12, OIR mice treated with sildenafil demonstrated a 24% reduction in vaso-obliteration (P < 0.05), whereas at P17, treated animals showed a 50% reduction in neovascularization (P < 0.05) compared to dextrose-treated controls. Sildenafil-treated OIR mice had stabilization of retinal HIF1α at P12, immediately after hyperoxia. At P17, sildenafil-treated OIR mice had decreased HIF1α relative to untreated mice. OIR mice developed right ventricle hypertrophy that was significant compared to that in room air controls, which was abrogated by sildenafil. CONCLUSIONS: Sildenafil treatment significantly decreased retinal vaso-obliteration and neovascularization in a mouse OIR model. These effects are likely due to sildenafil-induced HIF1α stabilization during hyperoxia exposure. Furthermore, we confirm disease overlap by showing that OIR mice also develop hyperoxia-induced right ventricular hypertrophy, which is prevented by sildenafil. This study is a first step toward delineating a potential therapeutic role for sildenafil in OIR and further suggests that there may be common pathophysiologic mechanisms underlying hyperoxia-induced retinal and pulmonary vascular disease.


Asunto(s)
Piperazinas/farmacología , Neovascularización Retiniana/tratamiento farmacológico , Vasos Retinianos/fisiopatología , Retinopatía de la Prematuridad/complicaciones , Sulfonas/farmacología , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Purinas/farmacología , Neovascularización Retiniana/etiología , Neovascularización Retiniana/patología , Vasos Retinianos/efectos de los fármacos , Vasos Retinianos/patología , Retinopatía de la Prematuridad/tratamiento farmacológico , Retinopatía de la Prematuridad/patología , Citrato de Sildenafil , Vasoconstricción/efectos de los fármacos , Vasodilatadores/farmacología
18.
Early Hum Dev ; 90(4): 195-200, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24480606

RESUMEN

BACKGROUND: Preterm infants are at risk for postnatal growth failure (PGF). Identification of biomarkers that are associated with neonatal growth may help reduce PGF and associated long-term morbidity. OBJECTIVE: To investigate the associations between cord blood vascular endothelial growth factor (VEGF) and its soluble receptor (sFlt-1) with birth weight (BW) and postnatal growth in premature infants. STUDY DESIGN AND METHODS: From an ongoing birth cohort, 123 premature infants from 23 to 36 weeks gestational age (GA) were studied. Cord blood plasma VEGF and sFlt-1 were measured via enzyme-linked immunoassay. Growth parameters and nutritional information were evaluated. Multivariate logistic regression models were constructed to evaluate the associations of VEGF and sFlt-1 on PGF, defined as weight <10th percentile at 36 weeks corrected age or discharge. RESULTS: VEGF was positively correlated, and sFlt-1 was negatively correlated with BW and BW-for-GA percentiles. Higher cord blood VEGF levels were associated with reduced risk of PGF (OR=0.7; 95% CI=0.5-0.9), while higher sFlt-1 levels appeared to increase the risk of PGF (OR=1.6; 95% CI=1.1-2.4). The above biomarker associations were attenuated after adjustment for maternal preeclampsia, fetal growth restriction and related neonatal characteristics, and when taking into account placental vascular pathologies. Longitudinal growth patterns by mean weight and length percentiles were consistently lower among infants with low VEGF/sFlt-1 ratios. CONCLUSIONS: Our data support that intrauterine regulation of angiogenesis is an important mechanism of fetal and postnatal growth. Cord blood VEGF and sFlt-1 are useful in elucidating how intrauterine processes may have long-standing effects on developing premature infants.


Asunto(s)
Peso al Nacer , Trastornos del Crecimiento/diagnóstico , Recien Nacido Prematuro/crecimiento & desarrollo , Factor A de Crecimiento Endotelial Vascular/sangre , Receptor 1 de Factores de Crecimiento Endotelial Vascular/sangre , Adulto , Biomarcadores/sangre , Endotelio Vascular/crecimiento & desarrollo , Femenino , Sangre Fetal/metabolismo , Trastornos del Crecimiento/sangre , Humanos , Recién Nacido , Recien Nacido Prematuro/sangre , Masculino , Neovascularización Fisiológica
19.
Am J Perinatol ; 31(3): 223-30, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23690052

RESUMEN

OBJECTIVES: To characterize postnatal growth failure (PGF), defined as weight < 10th percentile for postmenstrual age (PMA) in preterm (≤ 27 weeks' gestation) infants with severe bronchopulmonary dysplasia (sBPD) at specified time points during hospitalization, and to compare these in subgroups of infants who died/underwent tracheostomy and others. STUDY DESIGN: Retrospective review of data from the multicenter Children's Hospital Neonatal Database (CHND). RESULTS: Our cohort (n = 375) had a mean ± standard deviation gestation of 25 ± 1.2 weeks and birth weight of 744 ± 196 g. At birth, 20% of infants were small for gestational age (SGA); age at referral to the CHND neonatal intensive care unit (NICU) was 46 ± 50 days. PGF rates at admission and at 36, 40, 44, and 48 weeks' PMA were 33, 53, 67, 66, and 79% of infants, respectively. Tube feedings were administered to > 70% and parenteral nutrition to a third of infants between 36 and 44 weeks' PMA. At discharge, 34% of infants required tube feedings and 50% had PGF. A significantly greater (38 versus 17%) proportion of infants who died/underwent tracheostomy (n = 69) were SGA, compared with those who did not (n = 306; p < 0.01). CONCLUSIONS: Infants with sBPD commonly had progressive PGF during their NICU hospitalization. Fetal growth restriction may be a marker of adverse outcomes in this population.


Asunto(s)
Displasia Broncopulmonar/fisiopatología , Trastornos del Crecimiento/etiología , Aumento de Peso , Displasia Broncopulmonar/complicaciones , Displasia Broncopulmonar/terapia , Femenino , Humanos , Recién Nacido , Recien Nacido Prematuro , Masculino , Estudios Retrospectivos , Traqueostomía
20.
Am J Respir Cell Mol Biol ; 50(2): 369-78, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24032519

RESUMEN

Pulmonary hypertension (PH) occurs in 25 to 35% of premature infants with significant bronchopulmonary dysplasia (BPD). Neonatal mice exposed to 14 days of hyperoxia develop BPD-like lung injury and PH. To determinne the impact of hyperoxia on pulmonary artery (PA) cyclic guanosine monophosphate (cGMP) signaling in a murine model of lung injury and PH, neonatal C57BL/6 mice were placed in room air, 75% O2 for 14 days (chronic hyperoxia [CH]) or 75% O2 for 24 hours, followed by 13 days of room air (acute hyperoxia with recovery [AHR]) with or without sildenafil. At 14 days, mean alveolar area, PA medial wall thickness (MWT), right ventricular hypertrophy (RVH), and vessel density were assessed. PA protein was analyzed for cGMP, soluble guanylate cyclase, and PDE5 activity. CH and AHR mice had RVH, but only CH mice had increased alveolar area and MWT and decreased vessel density. In CH and AHR PAs, soluble guanylate cyclase activity was decreased, and PDE5 activity was increased. In CH mice, sildenafil attenuated MWT and RVH but did not improve mean alveolar area or vessel density. In CH and AHR PAs, sildenafil decreased PDE5 activity and increased cGMP. Our results indicate that prolonged hyperoxia leads to lung injury, PH, RVH, and disrupted PA cGMP signaling. Furthermore, 24 hours of hyperoxia causes RVH and disrupted PA cGMP signaling that persists for 13 days. Sildenafil reduced RVH and restored vascular cGMP signaling but did not attenuate lung injury. Thus, hyperoxia can rapidly disrupt PA cGMP signaling in vivo with sustained effects, and concurrent sildenafil therapy can be protective.


Asunto(s)
Guanosina Monofosfato/metabolismo , Hiperoxia/metabolismo , Hipertensión Pulmonar/tratamiento farmacológico , Piperazinas/farmacología , Arteria Pulmonar/metabolismo , Transducción de Señal , Sulfonas/farmacología , Animales , GMP Cíclico/metabolismo , Hiperoxia/complicaciones , Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/metabolismo , Pulmón/metabolismo , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/metabolismo , Ratones , Ratones Endogámicos C57BL , Arteria Pulmonar/patología , Purinas/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Citrato de Sildenafil
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...