Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Pediatr Res ; 94(5): 1684-1695, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37349511

RESUMO

BACKGROUND: Prenatal or postnatal lung inflammation and oxidative stress disrupt alveolo-vascular development leading to bronchopulmonary dysplasia (BPD) with and without pulmonary hypertension. L-citrulline (L-CIT), a nonessential amino acid, alleviates inflammatory and hyperoxic lung injury in preclinical models of BPD. L-CIT modulates signaling pathways mediating inflammation, oxidative stress, and mitochondrial biogenesis-processes operative in the development of BPD. We hypothesize that L-CIT will attenuate lipopolysaccharide (LPS)-induced inflammation and oxidative stress in our rat model of neonatal lung injury. METHODS: Newborn rats during the saccular stage of lung development were used to investigate the effect of L-CIT on LPS-induced lung histopathology and pathways involved in inflammatory, antioxidative processes, and mitochondrial biogenesis in lungs in vivo, and in primary culture of pulmonary artery smooth muscle cells, in vitro. RESULTS: L-CIT protected the newborn rat lung from LPS-induced: lung histopathology, ROS production, NFκB nuclear translocation, and upregulation of gene and protein expression of inflammatory cytokines (IL-1ß, IL-8, MCP-1α, and TNF-α). L-CIT maintained mitochondrial morphology, increased protein levels of PGC-1α, NRF1, and TFAM (transcription factors involved in mitochondrial biogenesis), and induced SIRT1, SIRT3, and superoxide dismutases protein expression. CONCLUSION: L-CIT may be efficacious in decreasing early lung inflammation and oxidative stress mitigating progression to BPD. IMPACT: The nonessential amino acid L-citrulline (L-CIT) mitigated lipopolysaccharide (LPS)-induced lung injury in the early stage of lung development in the newborn rat. This is the first study describing the effect of L-CIT on the signaling pathways operative in bronchopulmonary dysplasia (BPD) in a preclinical inflammatory model of newborn lung injury. If our findings translate to premature infants, L-CIT could decrease inflammation, oxidative stress and preserve mitochondrial health in the lung of premature infants at risk for BPD.


Assuntos
Displasia Broncopulmonar , Hiperóxia , Lesão Pulmonar , Pneumonia , Humanos , Recém-Nascido , Feminino , Gravidez , Animais , Ratos , Animais Recém-Nascidos , Displasia Broncopulmonar/metabolismo , Lipopolissacarídeos/farmacologia , Citrulina/farmacologia , Citrulina/metabolismo , Pulmão , Pneumonia/metabolismo , Inflamação/metabolismo , Modelos Animais de Doenças
2.
Physiol Rep ; 8(17): e14553, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32889775

RESUMO

Preterm infants are at high risk for developing bronchopulmonary dysplasia and pulmonary hypertension from inflammatory lung injury. In adult models, adiponectin (APN)-an adipocyte-derived hormone-protects the lung from inflammatory injury and pulmonary vascular remodeling. Cord blood APN levels in premature infants born < 26 weeks gestation are 5% of the level in infants born at term. We previously reported the expression profile of APN and its receptors in neonatal rat lung homogenates during the first 3 weeks of postnatal development. Here, we characterize the expression profile of APN and its receptors in specific lung cells and the effects of exogenous recombinant APN (rAPN) on lipopolysaccharide-(LPS)-induced cytokine and chemokine production in total lung homogenates and specific lung cells. In vitro, rAPN added to primary cultures of pulmonary artery smooth muscle cells attenuated the expression of LPS-induced pro-inflammatory cytokines while increasing the expression of anti-inflammatory cytokines. In vivo, intraperitoneal rAPN (2 mg/kg), given 4 hr prior to intrapharyngeal administration of LPS (5 mg/kg) to newborn rats at postnatal day 4, significantly reduced gene and protein expression of the pro-inflammatory cytokine IL-1ß and reduced protein expression of the chemokines monocyte chemoattractant protein (MCP-1) and macrophage inflammatory protein-1 alpha (MIP-1α) in the lung. LPS-induced histopathological changes in the lung were also decreased. Moreover, rAPN given 20 hr after intrapharyngeal LPS had a similar effect on lung inflammation. These findings suggest a role for APN in protecting the lung from inflammation during early stages of lung development.


Assuntos
Adiponectina/uso terapêutico , Anti-Inflamatórios/uso terapêutico , Displasia Broncopulmonar/tratamento farmacológico , Pneumonia/tratamento farmacológico , Adiponectina/farmacologia , Animais , Animais Recém-Nascidos , Anti-Inflamatórios/farmacologia , Displasia Broncopulmonar/etiologia , Células Cultivadas , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Lipopolissacarídeos/toxicidade , Pulmão/efeitos dos fármacos , Pulmão/crescimento & desenvolvimento , Pulmão/metabolismo , Pneumonia/etiologia , Ratos , Ratos Sprague-Dawley , Receptores de Adiponectina/genética , Receptores de Adiponectina/metabolismo , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico
3.
Cell Rep ; 27(10): 3006-3018.e4, 2019 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-31167144

RESUMO

Gut mesenchyme provides key stem cell niche signals such as Wnt ligands, but how these signals are regulated is unclear. Because Hedgehog (Hh) signaling is critical for gut mesenchymal development and tumorigenesis, we investigated Hh-mediated mechanisms by analyzing mice deleted for key negative regulators of Hh signaling, Sufu and/or Spop, in the gut mesenchyme, and demonstrated their dosage-dependent roles. Although these mutants exhibit abnormal mesenchymal cell growth and functionally defective muscle layers, villification is completed with proper mesenchymal clustering, implying a permissive role for Hh signaling. These mesenchymal defects are partially rescued by Gli2 reduction. Consistent with increased epithelial proliferation caused by abnormal Hh activation in development, Sufu reduction promotes intestinal tumorigenesis, whereas Gli2 heterozygosity suppresses it. Our analyses of chromatin and GLI2 binding genomic regions reveal its transcriptional regulation of stem cell niche signals through enhancers, providing mechanistic insight into the intestinal stem cell niche in development and tumorigenesis.


Assuntos
Transformação Celular Neoplásica , Intestino Delgado/metabolismo , Proteínas Repressoras/metabolismo , Complexos Ubiquitina-Proteína Ligase/metabolismo , Proteína Gli2 com Dedos de Zinco/metabolismo , Actinas/metabolismo , Animais , Proliferação de Células , Embrião de Mamíferos/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Proteínas Hedgehog/metabolismo , Intestino Delgado/crescimento & desenvolvimento , Intestino Delgado/patologia , Camundongos , Camundongos Knockout , Proteínas dos Microfilamentos/metabolismo , Contração Muscular , Proteínas Musculares/metabolismo , Músculos/metabolismo , Músculos/fisiologia , Proteínas Repressoras/deficiência , Proteínas Repressoras/genética , Transdução de Sinais , Nicho de Células-Tronco , Fator de Crescimento Transformador beta/metabolismo , Complexos Ubiquitina-Proteína Ligase/deficiência , Complexos Ubiquitina-Proteína Ligase/genética , Proteínas Wnt/metabolismo , Proteína Gli2 com Dedos de Zinco/genética
4.
Adv Exp Med Biol ; 1071: 151-157, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30357746

RESUMO

Premature infants have chronic intermittent hypoxia (CIH) that increases morbidity, and the youngest and the smallest premature infants are at the greatest risk. The combination of lung injury from inflammation/oxidative stress causing low functional residual capacity combined with frequent short apneas leads to CIH. Adiponectin (APN) is an adipose-derived adipokine that protects the lung from inflammation and oxidative stress. Premature and small for gestational age (SGA) infants have minimal body fat and low levels of circulating APN. To begin to understand the potential role of APN in lung protection during lung development, we characterized the developmental profile of APN and APN receptors (AdipoR1 and AdipoR2) protein and mRNA expression in the newborn rat lung at fetal day (FD) 19, and postnatal days (PD) 1, 4, 7, 10, 14, 21, and 28. Protein levels in lung homogenates were measured by western blot analyses; relative mRNA expression was detected by quantitative PCR (qPCR); and serum high molecular weight (HMW) APN was measured using enzyme-linked immunosorbent assay (ELISA). Results: APN protein and mRNA levels were lowest at FD19 and PD1, increased 2.2-fold at PD4, decreased at PD10, and then increased again at PD21. AdipoR1 protein and mRNA levels peaked at PD1, followed by a threefold drop by PD4, and remained low until PD21. AdipoR2 protein and mRNA levels also peaked at PD1, but remained high at PD4, followed by a 1.7-fold drop by PD10 that remained low by PD21. Serum APN levels detected by ELISA did not differ from PD4 to PD28. To date, this is the first report characterizing APN and APN receptor protein and mRNA expression in the rat lung during development. The developmental stage of the newborn rat lung models that of the premature human infant; both are in the saccular stage of lung development. In the newborn rat lung, alveolarization begins at PD4, peaks at PD10, and ends at PD21. Importantly, we found that AdipoR1 receptor protein and mRNA expression is lowest during lung alveolarization (PD4 to PD21). Thus, we speculate that low levels of AdipoR1 during lung alveolarization contributes to the increased susceptibility to developing acute lung edema and chronic lung injury such as bronchopulmonary dysplasia (BPD) in premature human infants.


Assuntos
Hipóxia/fisiopatologia , Lesão Pulmonar/fisiopatologia , Receptores de Adiponectina/metabolismo , Adiponectina/metabolismo , Animais , Animais Recém-Nascidos , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Ratos
5.
Exp Eye Res ; 169: 54-59, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29421328

RESUMO

Retinopathy of prematurity (ROP) is one of the leading causes of blindness in preterm Infants. Anti-vascular endothelial growth factor (VEGF) is emerging as a promising treatment, but there is insufficient evidence on their safety. We investigate the effect of systemic anti-VEGF in rat pups with equivalent maturity to a 32 week neonate. A single dose of either anti-VEGF antibody (n = 7) or saline (control group; n = 6) was administered to newborn rats intra-peritoneally on the first day of life. 14 days' post treatment, the serum concentration of anti-VEGF was measured and the brain, lung, heart, kidney and liver were harvested and weighed. The heart was processed to measure the Fulton index (a surrogate for pulmonary hypertension). All other organs were processed for mRNA expression of VEGF and VEGF-receptors (R1&R2). No group differences in body and organ weights were noted. The anti-VEGF was still detected in serum 14 days post Injection and resulted in increase in lung (p < 0.002) and kidney (p < 0.01) VEGF mRNA expressions and the lung (p < 0.02) VEGF-R1 and kidney (P < 0.001) VEGF-R2 mRNA expressions. The treated pups exhibited increased total heart weight (p < 0.01) and Fulton Index (p < 0.05). No changes were seen in the liver and brain. Anti-VEGF antibody did not affect mortality, total body and organ weights, but was associated with pulmonary hypertension. Expression of lung and kidney VEGF and its receptors was increased, whilst the brain and liver did not show changes. Dosing experiments can now be targeted to assess safety threshold and at anti-VEGF dose used in human ROP treatment.


Assuntos
Inibidores da Angiogênese/administração & dosagem , Animais Recém-Nascidos/crescimento & desenvolvimento , Encéfalo/crescimento & desenvolvimento , Rim/crescimento & desenvolvimento , Fígado/crescimento & desenvolvimento , Pulmão/crescimento & desenvolvimento , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Peso Corporal , Encéfalo/metabolismo , Injeções Intraperitoneais , Rim/metabolismo , Fígado/metabolismo , Pulmão/metabolismo , Tamanho do Órgão , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Fator A de Crescimento do Endotélio Vascular/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
7.
J Pediatr Surg ; 52(5): 744-748, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28168986

RESUMO

PURPOSE: The aim of this study is to assess gastric emptying in experimental necrotizing enterocolitis (NEC) and its diagnostic significance using non-invasive ultrasound imaging. METHODS: Fourteen neonatal mice (C57BL/6) were randomized into two groups: NEC [n=10] and control [n=4]. NEC was induced by gavage feeding of hyperosmolar formula, hypoxia, and lipopolysaccharide between postnatal day 5 (P5) and 9 (P9). Stomach volume was measured using a 40-MHz ultrasound transducer on P5 and P9. Gastric residual volume was calculated in control mice from two measurements at 4h interval and in NEC mice from two measurements immediately after gavage feeding and 4h post-fasting. The distal ileum was harvested for histology and quantitative PCR analysis on P9. RESULTS: On P9, NEC mice had a greater gastric residual volume compared to control (p=0.002) indicating delay in gastric emptying. Positive correlations were found between gastric residual volume and both IL-6 mRNA expression level and histological bowel damage (p=0.035, p=0.012, respectively). CONCLUSIONS: During experimental NEC there is a delay in stomach emptying which is related to the severity of the disease. Ultrasound assessment of gastric emptying is a new non-invasive imaging modality that could be used to predict the severity of NEC.


Assuntos
Enterocolite Necrosante/fisiopatologia , Esvaziamento Gástrico , Íleo/patologia , Índice de Gravidade de Doença , Animais , Animais Recém-Nascidos , Biomarcadores/metabolismo , Enterocolite Necrosante/diagnóstico por imagem , Enterocolite Necrosante/metabolismo , Enterocolite Necrosante/patologia , Íleo/metabolismo , Interleucina-6/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Distribuição Aleatória , Ultrassonografia
8.
Eur J Pediatr ; 175(10): 1343-51, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27595847

RESUMO

UNLABELLED: The objective of this study was to analyze the effect of I/E ratio on carbon dioxide (CO2) elimination during high-frequency oscillatory ventilation (HFOV) combined with volume guarantee (VG). Five 2-day-old piglets were studied before and after a bronchoalveolar lavage (BAL). The effect of an I/E ratio of 1:1 and 1:2 with (VG-ON) and without VG (VG-OFF) on PaCO2, as well as delta and mean airway pressures at the airway opening (∆Phf-ao, mPaw-ao) and at the tracheal level (∆Phf-t, mPaw-t) were evaluated at frequencies of 5, 8, 11, and 14 Hz. With the VG-ON, PaCO2 was significant lower with the I/E ratio of 1:2 at 5 Hz compared with the 1:1. mPaw-t was higher than mPaw-ao, with 1:1 I/E ratio, and on VG-ON, this difference was statistically significant. CONCLUSION: "In this animal study and with this ventilator, the I/E ratio of 1:1 compared to 1:2 in HFOV and VG-ON did not produce a higher CO2 lavage as when HFOV was used without the VG modality. Even more, a lower PaCO2 was found when using the lower frequency and 1:2 ratio compared to 1:1. So in contrast to non-VG HFOV mode, using a fixed tidal volume, no significant changes on CO2 elimination are observed during HFOV when the I/E ratios of 1:1 and 1:2 are compared at different frequencies." WHAT IS KNOWN: •The tidal volume on HFOV is determinant in CO 2 removal, and this is generated by delta pressure and the length of the inspiratory time. What is New: •HFOV combined with VG, an I/E ratio of 1:2 is more effective to remove CO 2 , and this is not related to the tidal volume.


Assuntos
Gasometria , Dióxido de Carbono/sangue , Modelos Animais de Doenças , Ventilação de Alta Frequência/métodos , Lesão Pulmonar/prevenção & controle , Síndrome do Desconforto Respiratório do Recém-Nascido/prevenção & controle , Análise de Variância , Animais , Animais Recém-Nascidos , Expiração , Inalação , Troca Gasosa Pulmonar , Distribuição Aleatória , Suínos , Volume de Ventilação Pulmonar
9.
Am J Physiol Lung Cell Mol Physiol ; 308(6): L503-10, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25595650

RESUMO

Arginase is an enzyme that limits substrate L-arginine bioavailability for the production of nitric oxide by the nitric oxide synthases and produces L-ornithine, which is a precursor for collagen formation and tissue remodeling. We studied the pulmonary vascular effects of arginase inhibition in an established model of repeated systemic bleomycin sulfate administration in neonatal rats that results in pulmonary hypertension and lung injury mimicking the characteristics typical of bronchopulmonary dysplasia. We report that arginase expression is increased in the lungs of bleomycin-exposed neonatal rats and that treatment with the arginase inhibitor amino-2-borono-6-hexanoic acid prevented the bleomycin-induced development of pulmonary hypertension and deposition of collagen. Arginase inhibition resulted in increased L-arginine and L-arginine bioavailability and increased pulmonary nitric oxide production. Arginase inhibition also normalized the expression of inducible nitric oxide synthase, and reduced bleomycin-induced nitrative stress while having no effect on bleomycin-induced inflammation. Our data suggest that arginase is a promising target for therapeutic interventions in neonates aimed at preventing lung vascular remodeling and pulmonary hypertension.


Assuntos
Aminocaproatos/farmacologia , Antibióticos Antineoplásicos/efeitos adversos , Arginase/antagonistas & inibidores , Bleomicina/efeitos adversos , Compostos de Boro/farmacologia , Colágeno/metabolismo , Hipertensão Pulmonar , Pulmão/enzimologia , Remodelação Vascular/efeitos dos fármacos , Animais , Antibióticos Antineoplásicos/farmacologia , Arginase/metabolismo , Arginina/metabolismo , Bleomicina/farmacologia , Displasia Broncopulmonar/induzido quimicamente , Displasia Broncopulmonar/enzimologia , Displasia Broncopulmonar/patologia , Displasia Broncopulmonar/prevenção & controle , Modelos Animais de Doenças , Hipertensão Pulmonar/induzido quimicamente , Hipertensão Pulmonar/enzimologia , Hipertensão Pulmonar/patologia , Hipertensão Pulmonar/prevenção & controle , Pulmão/patologia , Lesão Pulmonar/induzido quimicamente , Lesão Pulmonar/enzimologia , Lesão Pulmonar/patologia , Lesão Pulmonar/prevenção & controle , Óxido Nítrico/metabolismo , Ratos , Ratos Sprague-Dawley
10.
Am J Perinatol ; 30(2): 171-6, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24915560

RESUMO

OBJECTIVE: The aim of the article is to review the effectiveness of neonatal noninvasive high-frequency ventilation (NIHFV) in preventing endotracheal mechanical ventilation. STUDY DESIGN: Retrospective case series including all 79 instances of NIHFV use at four participating centers between July 2010 and September 2012. RESULTS: In 73% of cases, NIHFV was used as rescue after another noninvasive mode, and prophylactically (postextubation) in the remainder. In 58% of cases, infants transitioned to another noninvasive mode, without requiring intubation. There were significant reductions in the mean (SD) number of apneas, bradycardias, or desaturations (over 6 hours) (3.2 [0.4] vs. 1.2 [0.3]; p < 0.001), FiO2 (48 [3] vs. 40 [2]%; p < 0.001) and CO2 levels (74 [6] vs. 62 [4] mm Hg; p = 0.025] with NIHFV. No NIHFV-related complications were noted. CONCLUSIONS: NIHFV is a promising NIV mode that may help prevent or delay intubation and deserves further clinical research.


Assuntos
Displasia Broncopulmonar/prevenção & controle , Ventilação de Alta Frequência/métodos , Ventilação não Invasiva/métodos , Insuficiência Respiratória/terapia , Estudos de Coortes , Feminino , Humanos , Lactente Extremamente Prematuro , Recém-Nascido , Recém-Nascido Prematuro , Unidades de Terapia Intensiva Neonatal , Masculino , Estudos Retrospectivos , Lesão Pulmonar Induzida por Ventilação Mecânica/prevenção & controle
11.
Am J Physiol Lung Cell Mol Physiol ; 306(2): L207-15, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24242011

RESUMO

Inhaled nitric oxide (NO) and other cGMP- or cAMP-dependent pulmonary vasodilators are often used in combination for the treatment of the persistent pulmonary hypertension of the newborn syndrome. There is in vitro evidence to indicate that NO downregulate the pulmonary vascular response to cGMP-dependent agonists raising concern as to whether a synergistic effect is observed when employing a combined strategy in newborns. Hypothesizing that a synergistic effect is absent, we evaluated newborn and juvenile rat pulmonary arteries to determine the individual and combined vasodilatory effect of cGMP- and cAMP-dependent agonists. In precontracted near-resistance pulmonary arteries, the addition of sildenafil reduced vasorelaxation response to NO donor S-nitroso-N-acetyl penicillamine (SNAP). A similar decrease in SNAP-induced vasodilation was observed in arteries pretreated with BAY 41-2272 (10(-9) M), a soluble guanylate cyclase stimulator cGMP, and its downstream protein kinase activator. cGMP also reduced the vasorelaxant response to the cAMP-dependent forskolin. Inhibition of endogenous vascular NO generation enhanced SNAP-induced relaxation. The present data suggest that the mechanism involved in the cGMP desensitization to other relaxant agonists involves downregulation of the small heat shock protein HSP20 and is evident in rat pulmonary and systemic vascular smooth muscle cells. In newborn rats with chronic hypoxia-induced pulmonary hypertension, the combination of sildenafil and inhaled NO resulted in a lesser reduction in pulmonary vascular resistance compared with their individual effect. These data suggest that clinical exposure to one cGMP-dependent pulmonary vasodilator may affect the response to other cGMP- or cAMP-mediated agonists.


Assuntos
GMP Cíclico/metabolismo , Hipóxia/metabolismo , Músculo Liso Vascular/fisiologia , Circulação Pulmonar/efeitos dos fármacos , Circulação Pulmonar/fisiologia , Vasodilatadores/farmacologia , Animais , Animais Recém-Nascidos , Doença Crônica , Feminino , Proteínas de Choque Térmico HSP20/metabolismo , Proteínas de Choque Térmico HSP27/metabolismo , Hipóxia/fisiopatologia , Masculino , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Óxido Nítrico/metabolismo , Doadores de Óxido Nítrico/farmacologia , Fosforilação/fisiologia , Piperazinas/farmacologia , Cultura Primária de Células , Inibidores de Proteínas Quinases/farmacologia , Purinas/farmacologia , Ratos , Ratos Sprague-Dawley , Citrato de Sildenafila , Sulfonas/farmacologia , Vasodilatação/efeitos dos fármacos , Vasodilatação/fisiologia
12.
Redox Biol ; 1: 297-303, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24024164

RESUMO

BACKGROUND: Fetal-to-neonatal transition is associated with oxidative stress. In preterm infants, immaturity of the antioxidant system favours supplemental oxygen-derived morbidity and mortality. OBJECTIVES: To assess if prolonging in utero-like oxygenation during the fetal-to-neonatal transition limits oxidative stress in the lung and brain, improving postnatal adaptation of mice pups. MATERIAL AND METHODS: Inspiratory oxygen fraction (FiO2) in pregnant mice was reduced from 21% (room air) to 14% (hypoxia) 8-12 h prior to delivery and reset to 21% 6-8 h after birth. The control group was kept at 21% during the procedure. Reduced (GSH) and oxidized (GSSG) glutathione and its precursors [γ-glutamyl cysteine (γ-GC) and L-cysteine (CySH)] content and expression of several redox-sensitive genes were evaluated in newborn lung and brain tissue 1 (P1) and 7 (P7) days after birth. RESULTS: As compared with control animals, the GSH/GSSG ratio was increased in the hypoxic group at P1 and P7 in the lung, and at P7 in the brain. In the hypoxic group a significant increase in the mRNA levels of NAD(P)H:quinone oxidoreductase 1 (noq1), Sulfiredoxin 1 (srnx1) and Glutathione Peroxidase 1 (gpx) was found in lung tissue at P1, as well as a significant increase in gpx in brain tissue at P7. CONCLUSIONS: Delaying the increase in tissue oxygenation to occur after birth reduces short-and-long-term oxidative stress in the lung. Similar yet more subtle effects were found in the brain. Apparently, the fetal-to-neonatal transition under hypoxic conditions appears to have protective qualities.


Assuntos
Encéfalo/metabolismo , Pulmão/metabolismo , NAD(P)H Desidrogenase (Quinona)/genética , Animais , Animais Recém-Nascidos , Hipóxia Celular , Feminino , Regulação da Expressão Gênica , Glutationa/metabolismo , Glutationa Peroxidase/genética , Camundongos , Camundongos Endogâmicos C57BL , Estresse Oxidativo , Oxirredutases atuantes sobre Doadores de Grupo Enxofre/genética , Gravidez , Glutationa Peroxidase GPX1
13.
J. pediatr. (Rio J.) ; 89(3): 226-242, maio-jun. 2013. ilus, tab
Artigo em Português | LILACS | ID: lil-679302

RESUMO

OBJETIVOS: Embora reconhecida há décadas, ainda pouco se sabe a respeito da etiologia, fisiopatologia e prevenção da hipertensão pulmonar persistente neonatal (HPPN), e seu tratamento continua a ser um grande desafio para os neonatologistas. Nesta revisão, vamos abordar as características clínicas e os mecanismos fisiopatológicos da síndrome, assim como seu tratamento geral e específico. FONTES DE DADOS: Fizemos uma revisão nas bases de dados PubMed, Cochrane Library e MRei Consult , procurando por artigos relacionados à síndrome e publicados entre 1995 e 2011. SÍNTESE DE DADOS: São discutidos os fatores de risco e os mecanismos fisiopatológicos da síndrome. O quadro clínico depende dos diferentes fatores envolvidos, que provavelmente estão relacionados com a etiologia e o mecanismo fisiopatológico. Além das medidas utilizadas para permitir a queda da resistência vascular pulmonar após o nascimento, alguns casos necessitam de vasodilatadores pulmonares. Embora o óxido nítrico tenha se provado efetivo, recentemente, outros vasodilatadores têm sido usados, mas ainda faltam evidências clínicas para comprovar seus benefícios no tratamento da HPPN. CONCLUSÕES: Apesar dos recentes avanços tecnológicos e dos novos conhecimentos fisiopatológicos, a mortalidade associada à HPPN ainda é de 10%. São necessárias mais pesquisas clínicas e resultados experimentais baseados em evidências para prevenir, tratar e reduzir a morbimortalidade associada a esta síndrome neonatal.


OBJECTIVES: Although recognized for decades, little is known about the etiology, physiopathology, and prevention of persistent pulmonary hypertension of the newborn (PPHN). and its treatment remains a major challenge for neonatologists. In this review, the clinical features and physiopathology of the syndrome will be addressed, as well as its general and specific treatments. DATA SOURCE: A review was carried out in PubMed, Cochrane Library, and MRei consult databases, searching for articles related to the syndrome and published between 1995 and 2011. DATA SYNTHESIS: Risk factors and the physiopathological mechanisms of the syndrome are discussed. The clinical picture depends on the different factors involved, which are probably related to the etiology and physiopathological mechanisms. In addition to the measures used to allow the decrease in pulmonary vascular resistance after birth, some cases will require pulmonary vasodilators. Although nitric oxide has proved effective, other vasodilators have been recently used, but clinical evidence is still lacking to demonstrate their benefits in the treatment of PPHN. CONCLUSIONS: Despite recent technological advances and new physiopathological knowledge, mortality associated with PPHN remains at 10%. More clinical research and evidence-based experimental results are needed to prevent, treat, and reduce the morbidity/mortality associated with this neonatal syndrome.


Assuntos
Humanos , Recém-Nascido , Hipertensão Pulmonar , Administração por Inalação , Hipertensão Pulmonar/diagnóstico , Hipertensão Pulmonar/etiologia , Hipertensão Pulmonar/fisiopatologia , Hipertensão Pulmonar/terapia , Óxido Nítrico/uso terapêutico , Vasodilatadores/uso terapêutico
14.
PLoS One ; 7(10): e46618, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23071598

RESUMO

Increased arginase activity contributes to airway nitric oxide (NO) deficiency in cystic fibrosis (CF). Whether down-stream products of arginase activity contribute to CF lung disease is currently unknown. The objective of this study was to test whether L-ornithine derived polyamines are present in CF airways and contribute to airway pathophysiology. Polyamine concentrations were measured in sputum of patients with CF and in healthy controls, using liquid chromatography-tandem mass spectrometry. The effect of spermine on airway smooth muscle mechanical properties was assessed in bronchial segments of murine airways, using a wire myograph. Sputum polyamine concentrations in stable CF patients were similar to healthy controls for putrescine and spermidine but significantly higher for spermine. Pulmonary exacerbations were associated with an increase in sputum and spermine levels. Treatment for pulmonary exacerbations resulted in decreases in arginase activity, L-ornithine and spermine concentrations in sputum. The changes in sputum spermine with treatment correlated significantly with changes in L-ornithine but not with sputum inflammatory markers. Incubation of mouse bronchi with spermine resulted in an increase in acetylcholine-induced force and significantly reduced nitric oxide-induced bronchial relaxation. The polyamine spermine is increased in CF airways. Spermine contributes to airways obstruction by reducing the NO-mediated smooth muscle relaxation.


Assuntos
Poliaminas Biogênicas/metabolismo , Brônquios/metabolismo , Fibrose Cística/metabolismo , Ornitina/metabolismo , Escarro/metabolismo , Acetilcolina/farmacologia , Acetilcolina/fisiologia , Adolescente , Animais , Arginase/metabolismo , Poliaminas Biogênicas/fisiologia , Brônquios/enzimologia , Brônquios/fisiopatologia , Estudos de Casos e Controles , Criança , Feminino , Humanos , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Contração Muscular/efeitos dos fármacos , Relaxamento Muscular , Músculo Liso/fisiopatologia , Óxido Nítrico/farmacologia , Óxido Nítrico/fisiologia , Adulto Jovem
15.
Pediatrics ; 127(6): e1615-20, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21536610

RESUMO

High-flow hepatic vascular anomalies with arteriovenous shunting commonly manifest during the neonatal period with signs and symptoms of congestive heart failure, but to our knowledge, they have never been described in patients with hereditary hemorrhagic telangiectasia (HHT). We report here our experience with 3 patients with hepatic arteriovenous malformations (AVMs) who presented with symptoms of high-output congestive heart failure during the neonatal period and were subsequently diagnosed with HHT. Imaging showed large hypervascular lesions and multiple hepatic arteriovenous shunts that differentiated these lesions from liver hemangiomas. Transcatheter embolization was performed in all cases. One infant died of sepsis shortly after embolization; follow-up at the age of 2.5 years of the surviving infants revealed involution of the vascular lesions and no evidence of symptom recurrence. We conclude that severe symptoms related to hepatic AVMs in HHT can occur in the neonatal period and that HHT should therefore be included in the differential diagnosis of symptomatic neonatal hepatic vascular malformations. Imaging plays a key role in differentiating hepatic AVMs from hemangiomas, because the latter require additional pharmacologic treatments. Early transcatheter embolization seems to be effective, but long-term outcomes still need to be assessed.


Assuntos
Hepatopatias/etiologia , Fígado/irrigação sanguínea , Telangiectasia Hemorrágica Hereditária/complicações , Malformações Vasculares/complicações , Angiografia , Diagnóstico Diferencial , Embolização Terapêutica/métodos , Feminino , Seguimentos , Humanos , Recém-Nascido , Fígado/diagnóstico por imagem , Hepatopatias/diagnóstico , Hepatopatias/terapia , Masculino , Ultrassonografia , Malformações Vasculares/diagnóstico , Malformações Vasculares/terapia
16.
Am J Respir Crit Care Med ; 183(10): 1363-8, 2011 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-21278301

RESUMO

RATIONALE: Airway nitric oxide is reduced in cystic fibrosis airways. Asymmetric dimethylarginine is an endogenous nitric oxide synthase inhibitor that may contribute to nitric oxide deficiency in cystic fibrosis. OBJECTIVES: To test the hypothesis that asymmetric dimethylarginine is increased in cystic fibrosis and contributes to nitric oxide deficiency and airway obstruction. METHODS: The concentrations of asymmetric dimethylarginine, symmetric dimethylarginine, and l-arginine were measured in sputum of clinically stable patients with cystic fibrosis, in patients with cystic fibrosis before and after treatment for a pulmonary exacerbation, and in healthy control subjects, using liquid chromatography-tandem mass spectrometry. MEASUREMENTS AND MAIN RESULTS: Asymmetric dimethylarginine was increased in cystic fibrosis compared with control sputum, and the l-arginine/asymmetric dimethylarginine ratio was decreased. Symmetric dimethylarginine exceeded asymmetric dimethylarginine concentrations in control sputum, but this ratio was reversed in cystic fibrosis. Treatment for pulmonary exacerbation resulted in a decrease in sputum asymmetric dimethylarginine and an improved l-arginine/asymmetric dimethylarginine ratio. The treatment-related decrease in asymmetric dimethylarginine correlated significantly with an increase in sputum nitric oxide metabolites and improvement in pulmonary function. The activity of the asymmetric dimethylarginine-metabolizing enzyme, dimethylarginine dimethylaminohydrolase, was higher in cystic fibrosis sputum before rather than after treatment, suggesting that the accumulation of asymmetric dimethylarginine is caused by increased production, not decreased degradation, of asymmetric dimethylarginine. CONCLUSIONS: Asymmetric dimethylarginine is increased in cystic fibrosis airways and may contribute to airway obstruction in patients with cystic fibrosis by reducing nitric oxide formation.


Assuntos
Arginina/análogos & derivados , Fibrose Cística/metabolismo , Óxido Nítrico/metabolismo , Sistema Respiratório/metabolismo , Adolescente , Obstrução das Vias Respiratórias/complicações , Arginina/metabolismo , Criança , Cromatografia Líquida , Fibrose Cística/complicações , Feminino , Humanos , Pulmão/metabolismo , Masculino , Escarro/metabolismo , Espectrometria de Massas em Tandem
17.
Rev. bras. crescimento desenvolv. hum ; 20(3): 762-768, 2010. graf, tab
Artigo em Inglês | LILACS | ID: lil-603663

RESUMO

OBJETIVO: o acúmulo de líquido peritoneal é um achado comum em muitas crianças com distúrbios abdominais e sua geração é secundária ao aumento da permeabilidade vascular. A contribuição da circulação arterial versus venosa para a formação de edema e acúmulo de líquido peritoneal é mal compreendida. Estudos realizados in vivo mais de duas décadas atrás, sugeriram que a vênulas são mais importantes que os vasos arteriais no processo de formação de edema. O objetivo deste trabalho é analisar os efeitos das mudanças na pressão intravascular e a inflamação mediada pela bradicinina na permeabilidade venosa e vascular mesentérica de ratos. MÉTODO: artérias mesentéricas (MA) e veias (MV) foram montadas em cânulas de vidro, por via intravenosa preenchida com dextran fluorescentes e incremental pressurizado acima de seus valores fisiológicos in vivo. Permeabilidade vascular para dextrana foi determinada a 100, 200 e 300 por cento de pressões fisiológicas. RESULTADOS: a permeabilidade vascular estava presente em todas as medidas para ambos os vasos e sua magnitude diretamente proporcional à pressão intravascular. A bradicinina (10-5 M) aumentou significativamente a permeabilidade da MV, mas não no MA. CONCLUSÃO: o acúmulo de líquido abdominal relacionado à doença inflamatória intestinal é mais provável de ser secundária a venosa, em oposição à fuga vascular arterial


Assuntos
Ratos , Bradicinina/efeitos adversos , Permeabilidade Capilar , Sistema Cardiovascular , Endotélio Vascular , Edema/fisiopatologia , Artérias Mesentéricas , Veias Mesentéricas , Músculo Liso Vascular , Peritônio/fisiopatologia
18.
Am J Physiol Lung Cell Mol Physiol ; 297(5): L920-30, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19749000

RESUMO

Permissive hypercapnia, achieved using low tidal volume ventilation, has been an effective protective strategy in patients with acute respiratory distress syndrome. To date, no such protective effect has been demonstrated for the chronic neonatal lung injury, bronchopulmonary dysplasia. The objective of our study was to determine whether evolving chronic neonatal lung injury, using a rat model, is resistant to the beneficial effects of hypercapnia or simply requires a less conservative approach to hypercapnia than that applied clinically to date. Neonatal rats inhaled air or 60% O2 for 14 days with or without 5.5% CO2. Lung parenchymal neutrophil and macrophage numbers were significantly increased by hyperoxia alone, which was associated with interstitial thickening and reduced secondary crest formation. The phagocyte influx, interstitial thickening, and impaired alveolar formation were significantly attenuated by concurrent hypercapnia. Hyperoxic pups that received 5.5% CO2 had a significant increase in alveolar number relative to air-exposed pups. Increased tyrosine nitration, a footprint for peroxynitrite-mediated reactions, arteriolar medial wall thickening, and both reduced small peripheral pulmonary vessel number and VEGF and angiopoietin-1 (Ang-1) expression, which were observed with hyperoxia, was attenuated by concurrent hypercapnia. We conclude that evolving chronic neonatal lung injury in a rat model is responsive to the beneficial effects of hypercapnia. Inhaled 5.5% CO2 provided a significant degree of protection against parenchymal and vascular injury in an animal model of chronic neonatal lung injury likely due, at least in part, to its inhibition of a phagocyte influx.


Assuntos
Vasos Sanguíneos/fisiologia , Hipercapnia/complicações , Hipercapnia/fisiopatologia , Lesão Pulmonar/fisiopatologia , Lesão Pulmonar/terapia , Actinas/metabolismo , Ar , Animais , Animais Recém-Nascidos , Gasometria , Vasos Sanguíneos/efeitos dos fármacos , Dióxido de Carbono/farmacologia , Elastina/metabolismo , Imunofluorescência , Frequência Cardíaca/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Pulmão/patologia , Lesão Pulmonar/complicações , Neutrófilos/citologia , Neutrófilos/efeitos dos fármacos , Neutrófilos/enzimologia , Tamanho do Órgão/efeitos dos fármacos , Peroxidase/metabolismo , Ratos , Frações Subcelulares/efeitos dos fármacos , Frações Subcelulares/metabolismo , Tirosina/análogos & derivados , Tirosina/metabolismo , Fator de von Willebrand/metabolismo
19.
Am J Physiol Lung Cell Mol Physiol ; 296(1): L115-20, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18952757

RESUMO

The factors controlling the pulmonary vascular resistance under physiological conditions are poorly understood. We have previously reported on an apparent cross talk between the airway and adjacent pulmonary arterial bed where a factor likely derived from the bronchial epithelial cells reduced the magnitude of agonist-stimulated force in the vascular smooth muscle. The main purpose of this investigation was to evaluate whether bronchial epithelial cells release a pulmonary arterial smooth muscle relaxant factor. Conditioned media from SPOC-1 or BEAS-2B, a rat- and a human-derived bronchial epithelial cell line, respectively, were utilized. This media significantly relaxed precontracted adult but not fetal pulmonary arterial muscle in an oxygen tension-dependent manner. This response was mediated via soluble guanylate cyclase, involving AKT/PI3-kinase and neuronal nitric oxide synthase. Airway epithelial cell-conditioned media increased AKT phosphorylation in pulmonary smooth muscle cells (SMC) and reduced intracellular calcium change following ATP stimulation to a significantly greater extent than observed for bronchial SMC. The present data strongly support the evidence for bronchial epithelial cells releasing a stable and soluble factor capable of inducing pulmonary arterial SMC relaxation. We speculate that under physiological conditions, the maintenance of a low pulmonary vascular resistance, postnatally, is in part modulated by the airway epithelium.


Assuntos
Hipertensão Pulmonar/fisiopatologia , Músculo Liso Vascular/fisiologia , Artéria Pulmonar/fisiologia , Mucosa Respiratória/fisiologia , Vasoconstrição/fisiologia , Animais , Brônquios/fisiologia , Cálcio/metabolismo , Meios de Cultivo Condicionados/farmacologia , Feminino , Guanilato Ciclase/metabolismo , Hipóxia/fisiopatologia , Músculo Liso Vascular/citologia , Óxido Nítrico/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Gravidez , Proteínas Proto-Oncogênicas c-akt/metabolismo , Artéria Pulmonar/citologia , Ratos , Receptores Citoplasmáticos e Nucleares/metabolismo , Mucosa Respiratória/citologia , Guanilil Ciclase Solúvel , Vasodilatação/fisiologia
20.
Pediatr Res ; 64(3): 251-5, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18458659

RESUMO

Sildenafil is a pulmonary vasodilator shown to be effective in neonates, but conflicting data exist regarding its effect on arterial oxygenation. To address this issue, we tested the sildenafil effect on the piglet's hypoxic pulmonary vasoconstriction (HPV) response. A segmental lung atelectasis was created by obstructing the corresponding bronchus. Total pulmonary and specific flows to the atelectatic and contra-lateral lobes were measured by magnetic resonance (MR) before and 30-min post sildenafil (0.2 and 1 mg/kg i.v.) or saline administration. Flow was reduced (p < 0.01) in the atelectatic and increased in the contra-lateral lobe indicating an effective HPV response. Sildenafil at both doses significantly (p < 0.01) increased flow solely to the atelectatic lobe. At a dose of 1 mg/kg, sildenafil induced a decrease in Pao2 from 285 +/- 37 to 161 +/- 22 mm Hg (p < 0.01). We conclude that the HPV response in the newborn is capable of almost completely reducing blood flow to nonventilated lung units and is reversed following sildenafil i.v. administration in a dose-dependent manner. In the presence of lung parenchymal disease, the use of i.v. sildenafil as a pulmonary vasodilator may worsen arterial oxygenation by reversing the HPV response in nonventilated lung units.


Assuntos
Hipóxia/fisiopatologia , Pulmão/irrigação sanguínea , Piperazinas/farmacologia , Atelectasia Pulmonar/fisiopatologia , Sulfonas/farmacologia , Vasoconstrição/efeitos dos fármacos , Vasodilatadores/farmacologia , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Infusões Intravenosas , Imageamento por Ressonância Magnética , Piperazinas/administração & dosagem , Circulação Pulmonar/efeitos dos fármacos , Circulação Pulmonar/fisiologia , Ventilação Pulmonar/fisiologia , Purinas/administração & dosagem , Purinas/farmacologia , Fluxo Sanguíneo Regional/efeitos dos fármacos , Fluxo Sanguíneo Regional/fisiologia , Citrato de Sildenafila , Sulfonas/administração & dosagem , Suínos , Vasoconstrição/fisiologia , Vasodilatadores/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA