Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Paediatr Respir Rev ; 23: 72-77, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27856214

RESUMO

Intrauterine inflammation, or chorioamnionitis, is a major contributor to preterm birth. Prematurity per se is associated with considerable morbidity and mortality resulting from lung immaturity but exposure to chorioamnionitis reduces the risk of neonatal respiratory distress syndrome (RDS) in preterm infants. Animal experiments have identified that an increase in pulmonary surfactant production by the preterm lungs likely underlies this decreased risk of RDS in infants exposed to chorioamnionitis. Further animal experimentation has shown that infectious or inflammatory agents in amniotic fluid exert their effects on lung development by direct effects within the developing respiratory tract, and probably not by systemic pathways. Differences in the effects of intrauterine inflammation and glucocorticoids demonstrate that canonical glucocorticoid-mediated lung maturation is not responsible for inflammation-induced changes in lung development. Animal experimentation is identifying alternative lung maturational pathways, and transgenic animals and cell culture techniques will allow identification of novel mechanisms of lung maturation that may lead to new treatments for the prevention of RDS.


Assuntos
Corioamnionite/metabolismo , Pulmão , Pneumonia , Síndrome do Desconforto Respiratório do Recém-Nascido , Animais , Descoberta de Drogas , Feminino , Idade Gestacional , Glucocorticoides/metabolismo , Humanos , Pulmão/crescimento & desenvolvimento , Pulmão/metabolismo , Pulmão/fisiopatologia , Pneumonia/complicações , Pneumonia/metabolismo , Pneumonia/fisiopatologia , Gravidez , Nascimento Prematuro/etiologia , Nascimento Prematuro/metabolismo , Nascimento Prematuro/fisiopatologia , Prostaglandinas/metabolismo , Síndrome do Desconforto Respiratório do Recém-Nascido/etiologia , Síndrome do Desconforto Respiratório do Recém-Nascido/metabolismo , Síndrome do Desconforto Respiratório do Recém-Nascido/fisiopatologia , Síndrome do Desconforto Respiratório do Recém-Nascido/prevenção & controle
2.
J Physiol ; 591(8): 2127-37, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23420658

RESUMO

Intrauterine inflammation is associated with preterm birth and poor long-term cardiopulmonary outcomes. We aimed to determine the effect of intrauterine inflammation on the cardiopulmonary and cerebral haemodynamic transition at birth, and the response to subsequent haemodynamic challenge. Fetal instrumentation was performed at ∼112 days gestation (term is 147 days) for measurement of cardiopulmonary and cerebral haemodynamics. At 118 days, inflammation was induced by intra-amniotic administration of lipopolysaccharide (LPS; n = 7); controls (n = 5) received intra-amniotic saline. At 125 days lambs were delivered and mechanically ventilated. Arterial blood gases, pulmonary and systemic arterial blood pressures and flows were measured during the perinatal period. At 10 min a haemodynamic challenge was administered by increasing positive end-expiratory pressure. During the first 10 min after birth, LPS-exposed lambs had higher pulmonary vascular resistance and lower pulmonary blood flow and left ventricular output than controls. Carotid arterial blood flow was higher in LPS-exposed lambs than controls between 3 and 7 min after delivery, and cerebral oxygen delivery was higher at 5 min. During the haemodynamic challenge, pulmonary blood flow and left ventricular output were reduced in controls but not in LPS-exposed lambs; a transient reduction in brachiocephalic arterial pressure occurred in LPS-exposed lambs but not in controls. Intrauterine inflammation altered the cardiopulmonary and cerebral haemodynamic transition at birth and reduced the cardiopulmonary response to a haemodynamic challenge after birth. The transient reduction in brachiocephalic arterial pressure suggests intrauterine inflammation may alter cerebrovascular control following an increase in positive end-expiratory pressure.


Assuntos
Circulação Cerebrovascular , Inflamação/fisiopatologia , Circulação Pulmonar , Útero/fisiopatologia , Lesão Pulmonar Aguda/genética , Lesão Pulmonar Aguda/fisiopatologia , Animais , Artérias Carótidas/fisiopatologia , Feminino , Feto , Regulação da Expressão Gênica , Idade Gestacional , Hemodinâmica , Inflamação/genética , Lipopolissacarídeos , Masculino , Respiração com Pressão Positiva , Gravidez , Ovinos
3.
Dev Neurosci ; 35(2-3): 272-82, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23571644

RESUMO

Intrauterine infection, such as occurs in chorioamnionitis, is a principal cause of preterm birth and is a strong risk factor for neurological morbidity and cerebral palsy. This study aims to examine whether human amnion epithelial cells (hAECs) can be used as a potential therapeutic agent to reduce brain injury induced by intra-amniotic administration of lipopolysaccharide (LPS) in preterm fetal sheep. Pregnant ewes underwent surgery at approximately 110 days of gestation (term is approx. 147 days) for implantation of catheters into the amniotic cavity, fetal trachea, carotid artery and jugular vein. LPS was administered at 117 days; hAECs were labeled with carboxyfluorescein succinimidyl ester and administered at 0, 6 and 12 h, relative to LPS administration, into the fetal jugular vein, trachea or both. Control fetuses received an equivalent volume of saline. Brains were collected 7 days later for histological assessment of brain injury. Microglia (Iba-1-positive cells) were present in the brain of all fetuses and were significantly increased in the cortex, subcortical and periventricular white matter in fetuses that received LPS, indicative of inflammation. Inflammation was reduced in fetuses that received hAECs. In LPS fetuses, the number of TUNEL-positive cells was significantly elevated in the cortex, periventricular white matter, subcortical white matter and hippocampus compared with controls, and reduced in fetuses that received hAECs in the cortex and periventricular white matter. Within the fetal brains studied there was a significant positive correlation between the number of Iba-1-immunoreactive cells and the number of TUNEL-positive cells (R(2) = 0.19, p < 0.001). The administration of hAECs protects the developing brain when administered concurrently with the initiation of intrauterine inflammation.


Assuntos
Âmnio/citologia , Lesões Encefálicas/etiologia , Lesões Encefálicas/prevenção & controle , Corioamnionite/patologia , Células Epiteliais/transplante , Animais , Lesões Encefálicas/patologia , Corioamnionite/imunologia , Corioamnionite/metabolismo , Citocinas/análise , Citocinas/biossíntese , Modelos Animais de Doenças , Feminino , Feto , Humanos , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Gravidez , Carneiro Doméstico
4.
Am J Physiol Lung Cell Mol Physiol ; 302(7): L664-78, 2012 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-22287609

RESUMO

Intra-amniotic (IA) lipopolysaccharide (LPS) induces intrauterine and fetal lung inflammation and increases lung surfactant and compliance in preterm sheep; however, the mechanisms are unknown. Prostaglandins (PGs) are inflammatory mediators, and PGE(2) has established roles in fetal lung surfactant production. The aim of our first study was to determine PGE(2) concentrations in response to IA LPS and pulmonary gene expression for PG synthetic [prostaglandin H synthase-2 (PGHS-2) and PGE synthase (PGES)] and PG-metabolizing [prostaglandin dehydrogenase (PGDH)] enzymes and PGE(2) receptors. Our second study aimed to block LPS-induced increases in PGE(2) with a PGHS-2 inhibitor (nimesulide) and determine lung inflammation and surfactant protein mRNA expression. Pregnant ewes received an IA saline or LPS injection at 118 days of gestation. In study 1, fetal plasma and amniotic fluid were sampled before and at 2, 4, 6, 12, and 24 h after injection and then daily, and fetuses were delivered 2 or 7 days later. Amniotic fluid PGE(2) concentrations increased (P < 0.05) 12 h and 3-6 days after LPS. Fetal lung PGHS-2 mRNA and PGES mRNA increased 2 (P = 0.0084) and 7 (P = 0.014) days after LPS, respectively. In study 2, maternal intravenous nimesulide or vehicle infusion began immediately before LPS or saline injection and continued until delivery 2 days later. Nimesulide inhibited LPS-induced increases in PGE(2) and decreased fetal lung IL-1ß and IL-8 mRNA (P ≤ 0.002) without altering lung inflammatory cell infiltration. Nimesulide decreased surfactant protein (SP)-A (P = 0.05), -B (P = 0.05), and -D (P = 0.0015) but increased SP-C mRNA (P = 0.023). Thus PGHS-2 mediates, at least in part, fetal pulmonary responses to inflammation.


Assuntos
Corioamnionite/metabolismo , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Pulmão/embriologia , Pulmão/metabolismo , Pneumonia/embriologia , Útero/imunologia , Líquido Amniótico/química , Animais , Corioamnionite/imunologia , Inibidores de Ciclo-Oxigenase 2/farmacologia , Dinoprostona/sangue , Feminino , Interleucina-1beta/biossíntese , Interleucina-1beta/genética , Interleucina-8/biossíntese , Interleucina-8/genética , Lipopolissacarídeos/imunologia , Pulmão/patologia , Pulmão/fisiopatologia , Complacência Pulmonar , Pneumonia/imunologia , Pneumonia/metabolismo , Pneumonia/fisiopatologia , Gravidez , Proteínas Associadas a Surfactantes Pulmonares/biossíntese , Surfactantes Pulmonares/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Distribuição Aleatória , Ovinos , Sulfonamidas/farmacologia
5.
Clin Exp Pharmacol Physiol ; 39(9): 824-30, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22816773

RESUMO

1. Intrauterine infection or inflammation is common in cases of preterm birth. Preterm infants are at risk of acute respiratory distress as a result of lung immaturity; evidence of exposure to infection and/or inflammation before birth is associated with a reduced risk of neonatal respiratory distress syndrome (RDS). Experimentally induced intrauterine inflammation or infection in sheep causes a precocious increase in pulmonary surfactant in the preterm lungs that improves preterm lung function, consistent with the reduced risk of RDS in human infants exposed to infection and/or inflammation before birth. 2. The effects of intrauterine inflammation on fetal lung development appear to result from direct action of proinflammatory stimuli within the lungs rather than by systemic signals, such as the classical glucocorticoid-mediated lung maturation pathway. However, paracrine and/or autocrine production and/or metabolism of glucocorticoids in fetal lung tissue may occur as a result of inflammation-induced changes in the expression of 11ß-hydroxysteroid dehydrogenase (types 1 and 2). 3. Likely candidates that mediate inflammation-induced surfactant production by the preterm lung include prostaglandin E2 and/or other arachidonic acid metabolites. Intrauterine inflammation induces the expression of enzymes responsible for prostaglandin production in fetal lung tissue. Inhibition of prostaglandin production prevents, at least in part, the effects of inflammation on fetal lungs. 4. Our experiments are identifying mechanisms of surfactant production by the preterm lungs that may be exploited as novel therapies for preventing respiratory distress in preterm infants. Elucidation of the effects of inflammation on the fetal lungs and other organs will allow more refined approaches to the care of preterm infants exposed to inflammation in utero.


Assuntos
Corioamnionite/fisiopatologia , Endometrite/fisiopatologia , Pulmão/embriologia , Organogênese , Complicações Infecciosas na Gravidez/fisiopatologia , Nascimento Prematuro/etiologia , Corticosteroides/uso terapêutico , Animais , Displasia Broncopulmonar/etiologia , Displasia Broncopulmonar/prevenção & controle , Corioamnionite/imunologia , Corioamnionite/microbiologia , Endometrite/imunologia , Endometrite/microbiologia , Feminino , Humanos , Hidrocortisona/metabolismo , Recém-Nascido , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pulmão/fisiopatologia , Organogênese/efeitos dos fármacos , Gravidez , Complicações Infecciosas na Gravidez/imunologia , Complicações Infecciosas na Gravidez/microbiologia , Nascimento Prematuro/imunologia , Nascimento Prematuro/metabolismo , Nascimento Prematuro/fisiopatologia , Prostaglandina-Endoperóxido Sintases/metabolismo , Prostaglandinas/metabolismo , Surfactantes Pulmonares/metabolismo , Síndrome do Desconforto Respiratório do Recém-Nascido/etiologia , Síndrome do Desconforto Respiratório do Recém-Nascido/prevenção & controle
6.
Am J Obstet Gynecol ; 205(2): 156.e26-33, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21640967

RESUMO

OBJECTIVE: The purpose of this study was to determine whether human amnion epithelial cells (hAECs) can modulate the pulmonary developmental consequences of intrauterine inflammation in fetal sheep that are exposed to intraamniotic lipopolysaccharide (LPS) injection. STUDY DESIGN: At 117 days' gestation, fetal sheep (n=16) received intraamniotic LPS (20 mg). hAECs were delivered at 0, 6, and 12 hours into the fetal jugular vein (n=4), trachea (n=4), or both (n=4). Controls (n=6) received equivalent administration of saline solution. Lungs were collected at 124 days. RESULTS: Intraamniotic LPS caused pulmonary inflammation and altered lung structure and function. hAECs attenuated changes in lung function and structure that had been induced by LPS: lung volume, 40 cm H2O (P<.05, intravenous+intratracheal hAECs vs LPS), tissue-to-airspace ratio (P<.05, intravenous+intratracheal hAECs vs LPS), and septal crest density (P<.001, all hAEC groups vs LPS). Leukocyte infiltration of the lungs was not reduced by hAECs; however, inflammatory cytokines were reduced (tumor necrosis factor-α, P<.01, vs LPS; interleukin-1b, P<.01, vs LPS; interleukin-6, P<.01 vs LPS). Surfactant protein A and C messenger RNA was increased by LPS, although this was not statistically significant (P>.05 vs control); there were significant increases in all hAEC-treated animals (surfactant protein-A, P<.05 vs LPS; surfactant protein-C, P<.01 vs LPS). CONCLUSION: Human amnion epithelial cells attenuate the fetal pulmonary inflammatory response to experimental intrauterine inflammation and reduce, but (as administered in our study) do not prevent, consequent alterations in lung development.


Assuntos
Células Epiteliais/transplante , Lesão Pulmonar/terapia , Pneumonia/terapia , Prenhez , Âmnio , Animais , Células Cultivadas , Modelos Animais de Doenças , Feminino , Doenças Fetais/terapia , Humanos , Injeções , Lipopolissacarídeos/farmacologia , Lesão Pulmonar/induzido quimicamente , Lesão Pulmonar/patologia , Pneumonia/induzido quimicamente , Pneumonia/patologia , Gravidez , Distribuição Aleatória , Valores de Referência , Sensibilidade e Especificidade , Ovinos , Carneiro Doméstico , Transplante de Células-Tronco/métodos
7.
Reprod Sci ; 19(7): 740-7, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22421448

RESUMO

Intrauterine inflammation is a common antecedent of preterm birth and can alter the development of the fetal thymus, the site of development, and maturation of T lymphocytes. The effects of intrauterine inflammation on specific thymic T lymphocyte populations are largely unknown. We hypothesized that intrauterine inflammation would alter fetal thymic T cell populations. Immunohistochemistry was used to quantitate the relative proportions of thymic cortical and medullary cell populations in fetal sheep 7 days after intra-amniotic lipopolysaccharide (LPS) injection. The proportions of CD8⁺and MHC II⁺ cells in the fetal thymus were reduced in response to LPS. The ratio of CD4:CD8 cells was increased by LPS exposure. No changes were observed in the percentage of CD4⁺, γδ(WC1)⁺, CD45R⁺B cells, or CD44⁺ cells. These studies indicate that intrauterine inflammation impacts thymic composition of CD8 T cells and the development and/or activation of CD4 T cells in the fetal thymus.


Assuntos
Corioamnionite/imunologia , Modelos Animais de Doenças , Sistema Imunitário/embriologia , Timo/embriologia , Animais , Corioamnionite/metabolismo , Corioamnionite/patologia , Corioamnionite/fisiopatologia , Feminino , Sistema Imunitário/imunologia , Sistema Imunitário/metabolismo , Sistema Imunitário/patologia , Lipopolissacarídeos , Gravidez , Nascimento Prematuro/etiologia , Carneiro Doméstico , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/patologia , Timo/imunologia , Timo/metabolismo , Timo/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA