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

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Pediatr Res ; 78(6): 657-63, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26322409

RESUMO

BACKGROUND: Hyperoxia and hypoxia influence morbidity and mortality of preterm infants. Automated closed-loop control of the fraction of inspired oxygen (FiO(2)) has been shown to facilitate oxygen supplementation in the neonatal intensive care unit (NICU), but has not yet been tested during preterm resuscitation. We hypothesized that fully automated FiO(2) control based on predefined oxygen saturation (SpO(2)) targets was applicable in both preterm resuscitation and ventilation. METHODS: Twenty-two preterm lambs were operatively delivered and intubated in a modified EXIT procedure. They were randomized to receive standardized resuscitation with either automated or manual FiO(2) control, targeting SpO(2) according to the Dawson curve in the first 10 min and SpO(2) 90-95% hereafter. Automated FiO(2) control also was applied during surfactant replacement therapy and subsequent ventilation. RESULTS: Time within target range did not differ significantly between manual and automated FiO(2) control during resuscitation, however automated FiO(2) control significantly avoided hyperoxia. Automated FiO(2) control was feasible during surfactant replacement and kept SpO(2) within target range significantly better than manual control during subsequent ventilation. CONCLUSION: In our model, fully automated FiO(2) control was feasible in rapidly changing physiologic conditions during postnatal resuscitation and prevented hyperoxia. We conclude that closed loop FiO(2) control is a promising tool for the delivery room.


Assuntos
Hiperóxia/prevenção & controle , Pulmão/fisiopatologia , Oxigenoterapia/métodos , Nascimento Prematuro , Respiração Artificial/métodos , Síndrome do Desconforto Respiratório do Recém-Nascido/terapia , Ressuscitação/métodos , Animais , Animais Recém-Nascidos , Automação , Produtos Biológicos/farmacologia , Modelos Animais de Doenças , Estudos de Viabilidade , Feminino , Idade Gestacional , Hiperóxia/etiologia , Hiperóxia/fisiopatologia , Intubação Intratraqueal , Pulmão/efeitos dos fármacos , Pulmão/embriologia , Masculino , Oxigenoterapia/efeitos adversos , Fosfolipídeos/farmacologia , Gravidez , Surfactantes Pulmonares/farmacologia , Respiração Artificial/efeitos adversos , Síndrome do Desconforto Respiratório do Recém-Nascido/fisiopatologia , Ressuscitação/efeitos adversos , Ovinos , Fatores de Tempo
2.
Pediatr Res ; 78(6): 664-9, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26322413

RESUMO

BACKGROUND: Surfactant replacement therapy is the gold standard treatment of neonatal respiratory distress (RDS). Nebulization is a noninvasive mode of surfactant administration. We administered Poractant alfa (Curosurf) via a vibrating perforated membrane nebulizer (eFlow Neonatal Nebulizer) to spontaneously breathing preterm lambs during binasal continuous positive pressure ventilation (CPAP). METHODS: Sixteen preterm lambs were operatively delivered at a gestational age of 133 ± 1 d (term ~150 d), and connected to CPAP applied via customized nasal prongs. Nebulization was performed (i) with saline or (ii) with surfactant for 3 h in humidified or (iii) nonhumidified air, and with surfactant (iv) for 60 min or (v) for 30 min. We measured arterial oxygenation, lung gas volumes and surfactant pool size and deposition. RESULTS: Nebulization of surfactant in humidified air for 3 h improved oxygenation and lung function, and surfactant was preferentially distributed to the lower lung lobes. Shorter nebulization times and 3 h nebulization in dry air did not show these effects. Nebulized surfactant reached all lung lobes, however the increase of surfactant pool size missed statistical significance. CONCLUSION: Positive effects of surfactant nebulization to spontaneously breathing preterm lambs depend on treatment duration, surfactant dose, air humidity, and surfactant distribution within the lung.


Assuntos
Produtos Biológicos/administração & dosagem , Pressão Positiva Contínua nas Vias Aéreas , Pulmão/efeitos dos fármacos , Membranas Artificiais , Nebulizadores e Vaporizadores , Fosfolipídeos/administração & dosagem , Nascimento Prematuro , Surfactantes Pulmonares/administração & dosagem , Respiração/efeitos dos fármacos , Síndrome do Desconforto Respiratório do Recém-Nascido/terapia , Administração por Inalação , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Desenho de Equipamento , Idade Gestacional , Pulmão/fisiopatologia , Síndrome do Desconforto Respiratório do Recém-Nascido/fisiopatologia , Ovinos , Fatores de Tempo , Vibração
3.
Pediatr Res ; 77(1-1): 29-35, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25285474

RESUMO

BACKGROUND: Intrauterine inflammation activates the fetal immune system and can result in organ injury and postnatal complications in preterm infants. As the spleen is an important site for peripheral immune activation, we asked how the fetal spleen would respond to intrauterine inflammation over time. We hypothesized that intraamniotic lipopolysaccharide (IA LPS) exposure induces acute and persistent changes in the splenic cytokine profile and T-cell composition that may contribute to the sustained fetal inflammatory response after chorioamnionitis. METHODS: Fetal sheep were exposed to IA LPS 5, 12, and 24 h and 2, 4, 8, or 15 d before delivery at 125 d of gestational age (term = 150 d). Splenic cytokine mRNA levels and cleaved caspase-3, CD3, and Foxp3 expression were evaluated. RESULTS: IA LPS increased interleukin (IL)1, IL4, IL5, and IL10 mRNA by twofold 24 h after injection. Interferon gamma increased by fivefold, whereas IL23 decreased 15 d post-LPS exposure. Cleaved caspase-3-positive cells increased 2 and 8 d after LPS exposure. CD3 immunoreactivity increased within 5 h with increased Foxp3-positive cells at 12 h. CONCLUSION: Intrauterine inflammation induced a rapid and sustained splenic immune response with persistent changes in the cytokine profile. This altered immune status may drive sustained inflammation and injury in other fetal organs.


Assuntos
Líquido Amniótico/metabolismo , Lipopolissacarídeos/química , Baço/imunologia , Líquido Amniótico/efeitos dos fármacos , Animais , Apoptose , Complexo CD3/metabolismo , Caspase 3/metabolismo , Corioamnionite/fisiopatologia , Citocinas/metabolismo , Feminino , Feto/metabolismo , Idade Gestacional , Sistema Imunitário , Inflamação , Interleucina-23/metabolismo , Modelos Animais , Gravidez , Prenhez , RNA Mensageiro/metabolismo , Ovinos , Baço/metabolismo
4.
Pediatr Res ; 76(2): 166-70, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24796373

RESUMO

BACKGROUND: A new technique was proposed to administer surfactant to spontaneous breathing preterm infants by placing a thin catheter through the vocal cords. This technique was not studied with respect to oxygenation, gas exchange, surfactant distribution, and lung mechanics. We tested the technique of less-invasive surfactant administration (LISA) in a spontaneous breathing preterm lamb model. METHODS: Preterm lambs (n = 12) of 133-134 d gestational age were randomized to the following three groups: (i) continuous positive airway pressure (CPAP) only, (ii) CPAP + LISA, and (iii) intubation and mechanical ventilation with surfactant administration. Surfactant was labeled with samarium oxide. During the next 180 min, blood gas analyses were performed. Postmortem, lungs were removed and surfactant distribution was assessed, and pressure-volume curves were performed. RESULTS: Pao2 in the LISA-treated lambs was significantly higher than in the lambs that exclusively received CPAP. Moreover, Pao2 values were similar between the LISA-treated and the intubated lambs. Overall, surfactant deposition was less in the LISA lambs, with significantly less surfactant distributed to the right upper lobe. Lung compliance was better in the intubated lambs compared with the LISA-treated lambs, although this did not reach significance. CONCLUSION: LISA improved oxygenation, similar to conventional surfactant application techniques, despite lower surfactant deposition and lung compliance.


Assuntos
Cateterismo Periférico/métodos , Complacência Pulmonar/efeitos dos fármacos , Procedimentos Cirúrgicos Minimamente Invasivos/métodos , Oxigênio/sangue , Surfactantes Pulmonares/administração & dosagem , Surfactantes Pulmonares/farmacologia , Análise de Variância , Animais , Animais Recém-Nascidos , Pressão Positiva Contínua nas Vias Aéreas/métodos , Óxidos , Pressão Parcial , Surfactantes Pulmonares/metabolismo , Samário , Ovinos , Prega Vocal/cirurgia
5.
Pediatr Res ; 75(4): 500-6, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24441106

RESUMO

BACKGROUND: Intra-amniotic lipopolysaccharide (LPS) exposure may affect neonatal outcome by altering fetal lung and immune system development. We hypothesized that intra-amniotic LPS exposure would cause persistent fetal pulmonary responses as the lungs develop in utero. METHODS: Fetal lambs were exposed to intra-amniotic LPS at 118 or at 118 and 123 d of gestational age (GA) with delivery at 125, 133, or 140 d (term = 147 d). Immune responses, PU.1 expression, Toll-like receptor (TLR)-1,2,4,6 mRNA levels, mast cell levels, and pulmonary elastin deposition were evaluated. RESULTS: After a single dose of LPS, pulmonary inflammatory responses were observed with increases of (i) PU.1 and TLR1 at 125 d GA and (ii) monocytes, lymphocytes, TLR2, and TLR6 at 133 d GA. Repetitive LPS exposure resulted in (i) increases of neutrophils, monocytes, PU.1, and TLR1 at 125 d GA; (ii) increases of neutrophils, PU.1, and TLR2 at 133 d GA; and (iii) decreases of mast cells, elastin foci, TLR4, and TLR6 at early gestation. At 140 d GA, only PU.1 was increased after repetitive LPS exposure. CONCLUSION: The preterm fetal lung can respond to a single exposure or repeated exposures from intra-amniotic LPS in multiple ways, but the absence of inflammatory and structural changes in LPS-exposed fetuses delivered near term suggest that the fetus can resolve an inflammatory stimulus in utero with time.


Assuntos
Lipopolissacarídeos/farmacologia , Pulmão/embriologia , Prenhez , Ovinos/embriologia , Animais , Peso Corporal , Feminino , Pulmão/efeitos dos fármacos , Tamanho do Órgão , Gravidez
6.
Pediatr Res ; 75(2): 281-7, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24232635

RESUMO

BACKGROUND: Antenatal inflammation and maternal corticosteroids induce fetal lung maturation but interfere with late lung development. Canonical Wingless-Int (Wnt) signaling directs lung development and repair. We showed that intra-amniotic (IA) lipopolysaccharide (LPS) exposure disrupted developmental signaling pathways in the preterm lamb lungs. Therefore, we hypothesized that pulmonary Wnt signaling was altered by exposure to IA LPS and/or antenatal corticosteroids. METHODS: Ovine fetuses were exposed to IA LPS, maternal intramuscular betamethasone, a control saline injection, or a combination thereof at 107 and/or 114 d gestational age (term = 150 d gestational age) before delivery at 121 d gestational age. RESULTS: IA LPS exposure decreased the lung expression of lymphoid enhancer-binding factor 1 (LEF1), a major Wnt pathway effector. WNT1, WNT4, and downstream messenger ß-catenin decreased after LPS exposure. WNT7b mRNA increased fourfold 14 d post-LPS exposure. Betamethasone treatment 7 d before LPS exposure prevented the reduction in LEF1 expression, whereas betamethasone administration after LPS normalized the LPS-induced increase in Wnt7b mRNA. CONCLUSION: IA LPS exposure decreased canonical Wnt signaling in the developing lung. Antenatal corticosteroids before or after IA inflammation had different effects on pulmonary Wnt signaling. This study provides new insights into possible mechanisms by which prenatal inflammation affects lung development and how corticosteroid can be beneficial in this setting.


Assuntos
Betametasona/administração & dosagem , Lipopolissacarídeos/administração & dosagem , Pulmão/patologia , Via de Sinalização Wnt , Animais , Betametasona/química , Feminino , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Inflamação , Lipopolissacarídeos/química , Pulmão/metabolismo , Fator 1 de Ligação ao Facilitador Linfoide/metabolismo , Exposição Materna , Fosforilação , Gravidez , Prenhez , Ovinos , Carneiro Doméstico , Fatores de Tempo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
7.
Am J Physiol Lung Cell Mol Physiol ; 304(6): L438-44, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23333802

RESUMO

Inflammation and antenatal glucocorticoids, the latter given to mothers at risk for preterm birth, affect lung development and may contribute to the development of bronchopulmonary dysplasia (BPD). The effects of the combined exposures on inflammation and antenatal glucocorticoids on transforming growth factor (TGF)-ß signaling are unknown. TGF-ß and its downstream mediators are implicated in the etiology of BPD. Therefore, we asked whether glucocorticoids altered intra-amniotic lipopolysaccharide (LPS) effects on TGF-ß expression, its signaling molecule phosphorylated sma and mothers against decapentaplegic homolog 2 (pSmad2), and the downstream mediators connective tissue growth factor (CTGF) and caveolin-1 (Cav-1). Ovine singleton fetuses were randomized to receive either an intra-amniotic injection of LPS and/or maternal betamethasone (BTM) intramuscularly 7 and/or 14 days before delivery at 120 days gestational age (GA; term = 150 days GA). Saline was used for controls. Protein levels of TGF-ß1 and -ß2 were measured by ELISA. Smad2 phosphorylation was assessed by immunohistochemistry and Western blot. CTGF and Cav-1 mRNA and protein levels were determined by RT-PCR and Western blot. Free TGF-ß1 and -ß2 and total TGF-ß1 levels were unchanged after LPS and/or BTM exposure, although total TGF-ß2 increased in animals exposed to BTM 7 days before LPS. pSmad2 immunostaining increased 7 days after LPS exposure although pSmad2 protein expression did not increase. Similarly, CTGF mRNA and protein levels increased 7 days after LPS exposure as Cav-1 mRNA and protein levels decreased. BTM exposure before LPS prevented CTGF induction and Cav-1 downregulation. This study demonstrated that the intrauterine inflammation-induced TGF-ß signaling can be inhibited by antenatal glucocorticoids in fetal lungs.


Assuntos
Betametasona/administração & dosagem , Caveolina 1/metabolismo , Glucocorticoides/administração & dosagem , Pulmão/metabolismo , Fator de Crescimento Transformador beta/fisiologia , Animais , Caveolina 1/genética , Fator de Crescimento do Tecido Conjuntivo/genética , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Feminino , Feto/efeitos dos fármacos , Feto/imunologia , Feto/metabolismo , Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Injeções Intramusculares , Lipopolissacarídeos/farmacologia , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Troca Materno-Fetal , Fosforilação , Gravidez , Processamento de Proteína Pós-Traducional , Ovinos , Proteína Smad2/metabolismo , Fator de Crescimento Transformador beta/metabolismo
8.
J Neuroinflammation ; 10: 13, 2013 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-23347579

RESUMO

BACKGROUND: Hypoxic-ischemic encephalopathy (HIE) is one of the most important causes of brain injury in preterm infants. Preterm HIE is predominantly caused by global hypoxia-ischemia (HI). In contrast, focal ischemia is most common in the adult brain and known to result in cerebral inflammation and activation of the peripheral immune system. These inflammatory responses are considered to play an important role in the adverse outcomes following brain ischemia. In this study, we hypothesize that cerebral and peripheral immune activation is also involved in preterm brain injury after global HI. METHODS: Preterm instrumented fetal sheep were exposed to 25 minutes of umbilical cord occlusion (UCO) (n = 8) at 0.7 gestation. Sham-treated animals (n = 8) were used as a control group. Brain sections were stained for ionized calcium binding adaptor molecule 1 (IBA-1) to investigate microglial proliferation and activation. The peripheral immune system was studied by assessment of circulating white blood cell counts, cellular changes of the spleen and influx of peripheral immune cells (MPO-positive neutrophils) into the brain. Pre-oligodendrocytes (preOLs) and myelin basic protein (MBP) were detected to determine white matter injury. Electro-encephalography (EEG) was recorded to assess functional impairment by interburst interval (IBI) length analysis. RESULTS: Global HI resulted in profound activation and proliferation of microglia in the hippocampus, periventricular and subcortical white matter. In addition, non-preferential mobilization of white blood cells into the circulation was observed within 1 day after global HI and a significant influx of neutrophils into the brain was detected 7 days after the global HI insult. Furthermore, global HI resulted in marked involution of the spleen, which could not be explained by increased splenic apoptosis. In concordance with cerebral inflammation, global HI induced severe brain atrophy, region-specific preOL vulnerability, hypomyelination and persistent suppressed brain function. CONCLUSIONS: Our data provided evidence that global HI in preterm ovine fetuses resulted in profound cerebral inflammation and mobilization of the peripheral innate immune system. These inflammatory responses were paralleled by marked injury and functional loss of the preterm brain. Further understanding of the interplay between preterm brain inflammation and activation of the peripheral immune system following global HI will contribute to the development of future therapeutic interventions in preterm HIE.


Assuntos
Encéfalo/imunologia , Encéfalo/patologia , Movimento Celular/imunologia , Hipóxia-Isquemia Encefálica/imunologia , Hipóxia-Isquemia Encefálica/patologia , Animais , Animais Recém-Nascidos , Feminino , Feto/imunologia , Feto/patologia , Imunidade Inata , Microglia/imunologia , Microglia/patologia , Gravidez , Ovinos
9.
Am J Obstet Gynecol ; 208(6): 429-37, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23313727

RESUMO

In recent years, translational research with various animal models has been helpful to answer basic questions about the effect of antenatal inflammation on maturation and development of the fetal lung and immune system. The fetal lung and immune systems are very plastic and their development can be conditioned and influenced by both endogenous and/or exogenous factors. Antenatal inflammation can induce pulmonary inflammation, leading to lung injury and remodeling in the fetal lung. Exposure to antenatal inflammation can induce interleukin-1α production, which enhances surfactant protein and lipid synthesis thereby promoting lung maturation. Interleukin-1α is therefore a candidate for the link between lung inflammation and lung maturation, preventing respiratory distress syndrome in preterm infants. Antenatal inflammation can, however, cause structural changes in the fetal lung and affect the expression of growth factors, such as transforming growth factor-beta, connective tissue growth factor, fibroblast growth factor-10, or bone morphogenetic protein-4, which are essential for branching morphogenesis. These alterations cause alveolar and microvascular simplification resembling the histology of bronchopulmonary dysplasia. Antenatal inflammation may also affect neonatal outcome by modulating the responsiveness of the immune system. Lipopolysaccharide-tolerance (endotoxin hyporesponsiveness/immunoparalysis), induced by exposure to inflammation in utero, may prevent fetal lung damage, but increases susceptibility to postnatal infections. Moreover, prenatal exposure to inflammation appears to be a predisposition for the development of adverse neonatal outcomes, like bronchopulmonary dysplasia, if the preterm infant is exposed to a second postnatal hit, such as mechanical ventilation oxygen exposure, infections, or steroids.


Assuntos
Corioamnionite/fisiopatologia , Sistema Imunitário/fisiologia , Inflamação/fisiopatologia , Pulmão/fisiologia , Período Periparto , Animais , Feminino , Humanos , Tolerância Imunológica/fisiologia , Gravidez
10.
Am J Physiol Lung Cell Mol Physiol ; 303(9): L778-87, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-22962010

RESUMO

Chorioamnionitis and antenatal corticosteroids mature the fetal lung functionally but disrupt late-gestation lung development. Because Sonic Hedgehog (Shh) signaling is a major pathway directing lung development, we hypothesized that chorioamnionitis and antenatal corticosteroids modulated Shh signaling, resulting in an altered fetal lung structure. Time-mated ewes with singleton ovine fetuses received an intra-amniotic injection of lipopolysaccharide (LPS) and/or maternal intramuscular betamethasone 7 and/or 14 days before delivery at 120 days gestational age (GA) (term = 150 days GA). Intra-amniotic LPS exposure decreased Shh mRNA levels and Gli1 protein expression, which was counteracted by both betamethasone pre- or posttreatment. mRNA and protein levels of fibroblast growth factor 10 and bone morphogenetic protein 4, which are important mediators of lung development, increased 2-fold and 3.5-fold, respectively, 14 days after LPS exposure. Both 7-day and 14-day exposure to LPS changed the mRNA levels of elastin (ELN) and collagen type I alpha 1 (Col1A1) and 2 (Col1A2), which resulted in fewer elastin foci and increased collagen type I deposition in the alveolar septa. Corticosteroid posttreatment prevented the decrease in ELN mRNA and increased elastin foci and decreased collagen type I deposition in the fetal lung. In conclusion, fetal lung exposure to LPS was accompanied by changes in key modulators of lung development resulting in abnormal lung structure. Betamethasone treatment partially prevented the changes in developmental processes and lung structure. This study provides new insights into clinically relevant prenatal exposures and fetal lung development.


Assuntos
Betametasona/farmacologia , Corioamnionite/metabolismo , Feto/metabolismo , Glucocorticoides/farmacologia , Proteínas Hedgehog/metabolismo , Lipopolissacarídeos/farmacologia , Alvéolos Pulmonares/metabolismo , Animais , Betametasona/uso terapêutico , Proteína Morfogenética Óssea 4/genética , Proteína Morfogenética Óssea 4/metabolismo , Proliferação de Células , Corioamnionite/tratamento farmacológico , Corioamnionite/imunologia , Colágeno Tipo I/metabolismo , Elastina/metabolismo , Feminino , Feto/efeitos dos fármacos , Feto/embriologia , Feto/patologia , Fator 10 de Crescimento de Fibroblastos/genética , Fator 10 de Crescimento de Fibroblastos/metabolismo , Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Glucocorticoides/uso terapêutico , Proteínas de Choque Térmico HSP70/metabolismo , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Gravidez , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/embriologia , Alvéolos Pulmonares/patologia , Ovinos , Transdução de Sinais , Transativadores/genética , Transativadores/metabolismo , Proteína GLI1 em Dedos de Zinco
11.
Am J Physiol Lung Cell Mol Physiol ; 302(4): L380-9, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22160306

RESUMO

The proinflammatory stimulus of chorioamnionitis is commonly associated with preterm delivery. Women at risk of preterm delivery receive antenatal glucocorticoids to functionally mature the fetal lung. However, the effects of the combined exposures of chorioamnionitis and antenatal glucocorticoids on the fetus are poorly understood. Time-mated ewes with singleton fetuses received an intra-amniotic injection of lipopolysaccharide (LPS) either preceding or following maternal intramuscular betamethasone 7 or 14 days before delivery, and the fetuses were delivered at 120 days gestational age (GA) (term = 150 days GA). Gestation matched controls received intra-amniotic and maternal intramuscular saline. Compared with saline controls, intra-amniotic LPS increased inflammatory cells in the bronchoalveolar lavage and myeloperoxidase, Toll-like receptor 2 and 4 mRNA, PU.1, CD3, and Foxp3-positive cells in the fetal lung. LPS-induced lung maturation measured as increased airway surfactant and improved lung gas volumes. Intra-amniotic LPS-induced inflammation persisted until 14 days after exposure. Betamethasone treatment alone induced modest lung maturation but, when administered before intra-amniotic LPS, suppressed lung inflammation. Interestingly, betamethasone treatment after LPS did not counteract inflammation but enhanced lung maturation. We conclude that the order of exposures of intra-amniotic LPS or maternal betamethasone had large effects on fetal lung inflammation and maturation.


Assuntos
Betametasona/uso terapêutico , Corioamnionite/tratamento farmacológico , Desenvolvimento Fetal/efeitos dos fármacos , Glucocorticoides/uso terapêutico , Pulmão/embriologia , Âmnio , Animais , Líquido da Lavagem Broncoalveolar/citologia , Corioamnionite/etiologia , Corioamnionite/imunologia , Citocinas/genética , Citocinas/metabolismo , Feminino , Maturidade dos Órgãos Fetais/efeitos dos fármacos , Maturidade dos Órgãos Fetais/imunologia , Expressão Gênica , Inflamação/tratamento farmacológico , Inflamação/imunologia , Lipopolissacarídeos/farmacologia , Pulmão/efeitos dos fármacos , Pulmão/enzimologia , Pulmão/imunologia , Masculino , Acetato de Medroxiprogesterona/uso terapêutico , Peroxidase/metabolismo , Fosfatidilcolinas/metabolismo , Gravidez , Nascimento Prematuro/imunologia , Nascimento Prematuro/prevenção & controle , Proteína C Associada a Surfactante Pulmonar/genética , Proteína C Associada a Surfactante Pulmonar/metabolismo , Proteína D Associada a Surfactante Pulmonar/genética , Proteína D Associada a Surfactante Pulmonar/metabolismo , Distribuição Aleatória , Ovinos , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo
12.
Am J Physiol Lung Cell Mol Physiol ; 299(6): L852-60, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20935228

RESUMO

Bronchopulmonary dysplasia is associated with chorioamnionitis and fetal lung inflammation. Ureaplasma species are the bacteria most frequently isolated from chorioamnionitis. Very chronic ureaplasma colonization of amniotic fluid causes low-grade lung inflammation and functional lung maturation in fetal sheep. Less is known about shorter exposures of the fetal lung. Therefore, we hypothesized that ureaplasmas would cause an acute inflammatory response that would alter lung development. Singleton ovine fetuses received intra-amniotic Ureaplasma parvum serovar 3 or control media at 110, 117, or 121 days and were delivered at 124 days gestational age (term = 150 days). Inflammation was assessed by 1) cell counts in bronchoalveolar lavage fluid (BALF), and 2) cytokine mRNA measurements, immunohistochemistry, and flow cytometry for inflammatory cells and elastin and α-smooth muscle actin (α-SMA) staining in lung tissue. Neutrophils were increased in BALF 3 days after exposure to ureaplasmas (P = 0.01). Myeloperoxidase-positive cells increased after 3 days (P = 0.03), and major histocompatibility complex (MHC) class II-positive cells increased after 14 days of ureaplasma exposure (P = 0.001). PU.1 (macrophage marker)- or CD3 (T lymphocyte marker)-positive cells were not induced by ureaplasmas. CD3-positive cells in the posterior mediastinal lymph node increased in ureaplasma-exposed animals at 3, 7, and 14 days (P = 0.002). Focal elastin depositions decreased in alveolar septa at 14 days (P = 0.002), whereas α-SMA increased in arteries and bronchioli. U. parvum induced a mild acute inflammatory response and changed elastin and α-SMA deposition in the lung, which may affect lung structure and subsequent development.


Assuntos
Feto , Pneumonia , Ovinos , Infecções por Ureaplasma/imunologia , Ureaplasma/patogenicidade , Animais , Líquido da Lavagem Broncoalveolar/citologia , Citocinas/genética , Citocinas/metabolismo , Feminino , Feto/imunologia , Feto/microbiologia , Feto/fisiopatologia , Idade Gestacional , Humanos , Masculino , Pneumonia/imunologia , Pneumonia/microbiologia , Pneumonia/patologia , Gravidez , Infecções por Ureaplasma/metabolismo , Infecções por Ureaplasma/patologia
13.
Front Pediatr ; 5: 186, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28913327

RESUMO

BACKGROUND: Poractant alfa (Curosurf®) and Bovactant (Alveofact®) are two animal-derived pulmonary surfactants preparations approved for the treatment of neonatal respiratory distress syndrome (nRDS). They differ in their source, composition, pharmaceutical form, and clinical dose. How much these differences affect the acute pulmonary response to treatment is unknown. OBJECTIVES: Comparing these two surfactant preparations in two different animal models of respiratory distress focusing on the short-term response to treatment. METHODS: Poractant alfa and Bovactant were administered in a 50-200 mg/kg dose range to surfactant-depleted adult rabbits with acute respiratory distress syndrome induced by lavage and to preterm lambs (127-129 days gestational age) with nRDS induced by developmental immaturity. The acute impact of surfactant therapy on gas exchange and pulmonary mechanics was assessed for 1 h in surfactant-depleted rabbits and for 3 h in preterm lambs. RESULTS: Overall, treatment with Bovactant 50 mg/kg or Poractant alfa 50 mg/kg did not achieve full recovery of the rabbits' respiratory conditions, as indicated by significantly lower arterial oxygenation and carbon dioxide values. By contrast, the two approved doses for clinical use of Poractant alfa (100 and 200 mg/kg) achieved a rapid and sustained recovery in both animal models. The comparison of the ventilation indices of the licensed doses of Bovactant (50 mg/kg) and Poractant alfa (100 mg/kg) showed a superior performance of the latter preparation in both animal models. At equal phospholipid doses, Poractant alfa was superior to Bovactant in terms of arterial oxygenation in both animal models. In preterm lambs, surfactant replacement therapy with Poractant alfa at either 100 or 200 mg/kg was associated with significantly higher lung gas volumes compared to Bovactant treatment with 100 mg/kg. CONCLUSION: At the licensed doses, the acute pulmonary response to Poractant alfa was significantly better than the one observed after Bovactant treatment, either at 50 or at 100 mg/kg dose, in two animal models of pulmonary failure.

14.
Mol Cell Pediatr ; 2(1): 4, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26542294

RESUMO

BACKGROUND: Term and near-term infants are at high risk of developing brain injury and life-long disability if they have suffered from severe perinatal asphyxia. We hypothesized that propofol administration to the maternal-fetal unit can diminish cerebral injury in term and near-term infant fetuses in states of progressive severe asphyxia. METHODS: Forty-four late preterm lambs underwent total umbilical cord occlusion (UCO) or sham treatment in utero. UCO resulted in global asphyxia and cardiac arrest. After emergency cesarean section under either maternal propofol or isoflurane anesthesia, the fetuses were resuscitated and subsequently anesthetized the same way as their mothers. RESULTS: Asphyctic lambs receiving isoflurane showed a significant increase of total and low-frequency spectral power in bursts indicating seizure activity and more burst-suppression with a marked increase of interburst interval length during UCO. Asphyctic lambs receiving propofol showed less EEG changes. Propofol increased levels of anti-apoptotic B-cell lymphoma-extra large (Bcl-xL) and phosphorylated STAT-3 and reduced the release of cytochrome c from the mitochondria and the protein levels of activated cysteinyl aspartate-specific protease (caspase)-3, -9, and N-methyl-d-aspartate (NMDA) receptor. CONCLUSIONS: Improvement of fetal EEG during and after severe asphyxia could be achieved by propofol treatment of the ovine maternal-fetal unit. The underlying mechanism is probably the reduction of glutamate-induced cytotoxicity by down-regulation of NMDA receptors and an inhibition of the mitochondrial apoptotic pathway.

15.
CNS Neurol Disord Drug Targets ; 14(1): 77-84, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25613498

RESUMO

OBJECTIVE: To better understand the inflammatory response in the central nervous system (CNS) after lipopolysaccharide (LPS)-induced chorioamnionitis. STUDY DESIGN: Fetal sheep were exposed to intra-amniotic LPS 2 or 14 days before preterm delivery at 125 days of gestation. mRNA levels of cytokines, TLRs and anti-oxidants were determined in different CNS regions. RESULTS: Interleukin 1ß levels increased in hippocampus, cortex and cerebellum 2 days after LPS exposure, while Interleukin 8 levels increased in the periventricular white matter as well. Levels returned back to control levels after 14 days. Tumor necrosis factor-α levels increased in hippocampus and cortex after 2 days. Toll like receptor 4 levels was upregulated in all grey matter regions 2 and 14 days after exposure. Glutathione s-transferase mRNA levels were lower after 2 and 14 days in all grey matter regions. CONCLUSION: Intra-amniotic LPS exposure causes acute and region-specific changes in inflammatory markers in the fetal brain, with grey matter being more affected than white matter. CONDENSATION: Intra-amniotic LPS exposure causes acute and region-specific changes in cytokines, TLR and anti-oxidants levels, with grey matter being more affected than white matter.


Assuntos
Sistema Nervoso Central/patologia , Corioamnionite/induzido quimicamente , Corioamnionite/fisiopatologia , Inflamação/etiologia , Inflamação/patologia , Lipopolissacarídeos/toxicidade , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Catalase/genética , Catalase/metabolismo , Sistema Nervoso Central/embriologia , Sistema Nervoso Central/metabolismo , Corioamnionite/veterinária , Citocinas/genética , Citocinas/metabolismo , Feminino , Feto , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Inflamação/veterinária , Gravidez , RNA Mensageiro/metabolismo , Ovinos , Fatores de Tempo , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo
16.
Reprod Sci ; 20(8): 946-56, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23314960

RESUMO

RATIONALE: Chorioamnionitis induces preterm delivery and acute involution of the fetal thymus which is associated with postnatal inflammatory disorders. We studied the immune response, cell composition, and architecture of the fetal thymus following intraamniotic lipopolysaccharide (LPS) exposure. METHODS: Time-mated ewes received an intraamniotic injection of LPS 5, 12, or 24 hours or 2, 4, 8, or 15 days before delivery at 125 days gestational age (term = 150 days). RESULTS: The LPS exposure resulted in decreased blood lymphocytes within 5 hours and decreased thymic corticomedullary ratio within 24 hours. Thymic interleukin 6 (IL6) and IL17 messenger RNA (mRNA) increased 5-fold 24 hours post-LPS exposure. Increased toll-like receptor 4 (TLR4) mRNA and nuclear factor κB positive cells at 24 hours after LPS delivery demonstrated acute thymic activation. Both TLR4 and IL1 mRNA increased by 5-fold and the number of Foxp3-positive cells (Foxp3+ cells) decreased 15 days after exposure. CONCLUSION: Intraamniotic LPS exposure caused a proinflammatory response, involution, and a persistent depletion of thymic Foxp3+ cells indicating disturbance of the fetal immune homeostasis.


Assuntos
Corioamnionite/imunologia , Lipopolissacarídeos/administração & dosagem , Timo/imunologia , Líquido Amniótico , Animais , Apoptose , Proliferação de Células , Corioamnionite/induzido quimicamente , Corioamnionite/patologia , Modelos Animais de Doenças , Feminino , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Mediadores da Inflamação/metabolismo , Injeções , Interleucina-17/genética , Interleucina-17/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , NF-kappa B/metabolismo , Gravidez , RNA Mensageiro/metabolismo , Ovinos , Timo/embriologia , Timo/patologia , Fatores de Tempo , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo
17.
Neonatology ; 104(1): 49-55, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23711546

RESUMO

BACKGROUND: Bronchopulmonary dysplasia (BPD) is one of the most common complications after preterm birth and is associated with intrauterine exposure to bacteria. Transforming growth factor-ß (TGFß) is implicated in the development of BPD. OBJECTIVES: We hypothesized that different and/or multiple bacterial signals could elicit divergent TGFß signaling responses in the developing lung. METHODS: Time-mated pregnant Merino ewes received an intra-amniotic injection of lipopolysaccharide (LPS) and/or Ureaplasma parvum serovar 3 (UP) at 117 days' and/or 121/122 days' gestational age (GA). Controls received an equivalent injection of saline and or media. Lambs were euthanized at 124 days' GA (term = 150 days' GA). TGFß1, TGFß2, TGFß3, TGFß receptor (R)1 and TGFßR2 protein levels, Smad2 phosphorylation and elastin deposition were evaluated in lung tissue. RESULTS: Total TGFß1 and TGFß2 decreased by 24 and 51% after combined UP+LPS exposure, whereas total TGFß1 increased by 31% after 7 days' LPS exposure but not after double exposures. Alveolar expression of TGFßR2 decreased 75% after UP, but remained unaltered after double exposures. Decreased focal elastin deposition after single LPS exposure was prevented by double exposures. CONCLUSIONS: TGFß signaling components and elastin responded differently to intrauterine LPS and UP exposure. Multiple bacterial exposures attenuated TGFß signaling and normalized elastin deposition.


Assuntos
Inflamação/fisiopatologia , Pulmão/embriologia , Complicações na Gravidez/fisiopatologia , Ovinos/embriologia , Transdução de Sinais , Fator de Crescimento Transformador beta/fisiologia , Âmnio/efeitos dos fármacos , Animais , Corioamnionite , Modelos Animais de Doenças , Elastina/análise , Feminino , Idade Gestacional , Lipopolissacarídeos/administração & dosagem , Pulmão/química , Fosforilação , Gravidez , Receptores de Fatores de Crescimento Transformadores beta/análise , Proteína Smad2/metabolismo , Fator de Crescimento Transformador beta1/análise , Fator de Crescimento Transformador beta2/análise , Fator de Crescimento Transformador beta3/análise , Ureaplasma
18.
PLoS One ; 8(8): e73031, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23991170

RESUMO

Hypoxic-ischemic encephalopathy (HIE) in preterm infants is a severe disease for which no curative treatment is available. Cerebral inflammation and invasion of activated peripheral immune cells have been shown to play a pivotal role in the etiology of white matter injury, which is the clinical hallmark of HIE in preterm infants. The objective of this study was to assess the neuroprotective and anti-inflammatory effects of intravenously delivered mesenchymal stem cells (MSC) in an ovine model of HIE. In this translational animal model, global hypoxia-ischemia (HI) was induced in instrumented preterm sheep by transient umbilical cord occlusion, which closely mimics the clinical insult. Intravenous administration of 2 x 10(6) MSC/kg reduced microglial proliferation, diminished loss of oligodendrocytes and reduced demyelination, as determined by histology and Diffusion Tensor Imaging (DTI), in the preterm brain after global HI. These anti-inflammatory and neuroprotective effects of MSC were paralleled by reduced electrographic seizure activity in the ischemic preterm brain. Furthermore, we showed that MSC induced persistent peripheral T-cell tolerance in vivo and reduced invasion of T-cells into the preterm brain following global HI. These findings show in a preclinical animal model that intravenously administered MSC reduced cerebral inflammation, protected against white matter injury and established functional improvement in the preterm brain following global HI. Moreover, we provide evidence that induction of T-cell tolerance by MSC might play an important role in the neuroprotective effects of MSC in HIE. This is the first study to describe a marked neuroprotective effect of MSC in a translational animal model of HIE.


Assuntos
Encéfalo/embriologia , Hipóxia-Isquemia Encefálica/imunologia , Tolerância Imunológica , Células-Tronco Mesenquimais/imunologia , Linfócitos T/imunologia , Animais , Sequência de Bases , Primers do DNA , Modelos Animais de Doenças , Imageamento por Ressonância Magnética , Reação em Cadeia da Polimerase , Convulsões/prevenção & controle , Ovinos
19.
Exp Neurol ; 250: 293-303, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24120465

RESUMO

Hypoxic-ischemic encephalopathy (HIE) is common in preterm infants, but currently no curative therapy is available. Cell-based therapy has a great potential in the treatment of hypoxic-ischemic preterm brain injury. Granulocyte-colony stimulating factor (G-CSF) is known to mobilize endogenous hematopoietic stem cells (HSC) and promotes proliferation of endogenous neural stem cells. On these grounds, we hypothesized that systemic G-CSF would be neuroprotective in a large translational animal model of hypoxic-ischemic injury in the preterm brain. Global hypoxia-ischemia (HI) was induced by transient umbilical cord occlusion in instrumented preterm sheep. G-CSF treatment (100µg/kg intravenously, during five consecutive days) was started one day before the global HI insult to ascertain mobilization of endogenous stem cells within the acute phase after global HI. Mobilization of HSC and neutrophils was studied by flow cytometry. Brain sections were stained for microglia (IBA-1), myelin basic protein (MBP) and myeloperoxidase (MPO) to study microglial proliferation, white matter injury and neutrophil invasion respectively. Electrographic seizure activity was analyzed using amplitude-integrated electroencephalogram (aEEG). G-CSF effectively mobilized CD34-positive HSC in the preterm sheep. In addition, G-CSF caused marked mobilization of neutrophils, but did not influence enhanced invasion of neutrophils into the preterm brain after global HI. Microglial proliferation and hypomyelination following global HI were reduced as a result of G-CSF treatment. G-CSF did not cause a reduction of the electrographic seizure activity after global HI. In conclusion, G-CSF induced mobilization of endogenous stem cells which was associated with modulation of the cerebral inflammatory response and reduced white matter injury in an ovine model of preterm brain injury after global HI. G-CSF treatment did not improve neuronal function as shown by seizure analysis. Our study shows that G-CSF treatment has neuroprotective potential following hypoxic-ischemic injury in the preterm brain.


Assuntos
Encefalite/patologia , Hipóxia Fetal/complicações , Fator Estimulador de Colônias de Granulócitos/farmacologia , Hipóxia-Isquemia Encefálica/complicações , Fármacos Neuroprotetores/farmacologia , Animais , Modelos Animais de Doenças , Eletrocardiografia , Eletroencefalografia , Encefalite/etiologia , Hipóxia Fetal/patologia , Feto , Citometria de Fluxo , Mobilização de Células-Tronco Hematopoéticas , Hipóxia-Isquemia Encefálica/patologia , Imuno-Histoquímica , Fibras Nervosas Mielinizadas/efeitos dos fármacos , Convulsões/etiologia , Ovinos
20.
PLoS One ; 8(12): e81644, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24358119

RESUMO

RATIONALE: Chorioamnionitis and antenatal glucocorticoids are common exposures for preterm infants and can affect the fetal brain, contributing to cognitive and motor deficits in preterm infants. The effects of antenatal glucocorticoids on the brain in the setting of chorioamnionitis are unknown. We hypothesized that antenatal glucocorticoids would modulate inflammation in the brain and prevent hippocampal and white matter injury after intra-amniotic lipopolysaccharide (LPS) exposure. METHODS: Time-mated ewes received saline (control), an intra-amniotic injection of 10 mg LPS at 106d GA or 113d GA, maternal intra-muscular betamethasone (0.5 mg/kg maternal weight) alone at 113d GA, betamethasone at 106d GA before LPS or betamethasone at 113d GA after LPS. Animals were delivered at 120d GA (term=150d). Brain structure volumes were measured on T2-weighted MRI images. The subcortical white matter (SCWM), periventricular white matter (PVWM) and hippocampus were analyzed for microglia, astrocytes, apoptosis, proliferation, myelin and pre-synaptic vesicles. RESULTS: LPS and/or betamethasone exposure at different time-points during gestation did not alter brain structure volumes on MRI. Betamethasone alone did not alter any of the measurements. Intra-amniotic LPS at 106d or 113d GA induced inflammation as indicated by increased microglial and astrocyte recruitment which was paralleled by increased apoptosis and hypomyelination in the SCWM and decreased synaptophysin density in the hippocampus. Betamethasone before the LPS exposure at 113d GA prevented microglial activation and the decrease in synaptophysin. Betamethasone after LPS exposure increased microglial infiltration and apoptosis. CONCLUSION: Intra-uterine LPS exposure for 7d or 14d before delivery induced inflammation and injury in the fetal white matter and hippocampus. Antenatal glucocorticoids aggravated the inflammatory changes in the brain caused by pre-existing intra-amniotic inflammation. Antenatal glucocorticoids prior to LPS reduced the effects of intra-uterine inflammation on the brain. The timing of glucocorticoid administration in the setting of chorioamnionitis can alter outcomes for the fetal brain.


Assuntos
Líquido Amniótico/efeitos dos fármacos , Betametasona/farmacologia , Encefalite/imunologia , Feto/efeitos dos fármacos , Glucocorticoides/farmacologia , Lipopolissacarídeos/farmacologia , Líquido Amniótico/imunologia , Animais , Feminino , Gravidez , Ovinos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA