Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Am J Respir Crit Care Med ; 205(10): 1186-1201, 2022 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-35286238

RESUMO

Rationale: Bronchopulmonary dysplasia, a chronic respiratory condition originating from preterm birth, is associated with abnormal neurodevelopment. Currently, there is an absence of effective therapies for bronchopulmonary dysplasia and its associated brain injury. In preclinical trials, mesenchymal stromal cell therapies demonstrate promise as a therapeutic alternative for bronchopulmonary dysplasia. Objectives: To investigate whether a multifactorial neonatal mouse model of lung injury perturbs neural progenitor cell function and to assess the ability of human umbilical cord-derived mesenchymal stromal cell extracellular vesicles to mitigate pulmonary and neurologic injury. Methods: Mice at Postnatal Day 7 or 8 were injected intraperitoneally with LPS and ventilated with 40% oxygen at Postnatal Day 9 or 10 for 8 hours. Treated animals received umbilical cord-mesenchymal stromal cell-derived extracellular vesicles intratracheally preceding ventilation. Lung morphology, vascularity, and inflammation were quantified. Neural progenitor cells were isolated from the subventricular zone and hippocampus and assessed for self-renewal, in vitro differentiation ability, and transcriptional profiles. Measurements and Main Results: The multifactorial lung injury model produced alveolar and vascular rarefaction mimicking bronchopulmonary dysplasia. Neural progenitor cells from lung injury mice showed reduced neurosphere and oligodendrocyte formation, as well as inflammatory transcriptional signatures. Mice treated with mesenchymal stromal cell extracellular vesicles showed significant improvement in lung architecture, vessel formation, and inflammatory modulation. In addition, we observed significantly increased in vitro neurosphere formation and altered neural progenitor cell transcriptional signatures. Conclusions: Our multifactorial lung injury model impairs neural progenitor cell function. Observed pulmonary and neurologic alterations are mitigated by intratracheal treatment with mesenchymal stromal cell-derived extracellular vesicles.


Assuntos
Displasia Broncopulmonar , Vesículas Extracelulares , Lesão Pulmonar , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Nascimento Prematuro , Animais , Displasia Broncopulmonar/terapia , Feminino , Humanos , Recém-Nascido , Pulmão , Lesão Pulmonar/terapia , Camundongos , Gravidez
2.
Cytotherapy ; 20(1): 108-125, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29056548

RESUMO

BACKGROUND AIMS: Bronchopulmonary dysplasia (BPD), a chronic lung disease characterized by disrupted lung growth, is the most common complication in extreme premature infants. BPD leads to persistent pulmonary disease later in life. Alveolar epithelial type 2 cells (AEC2s), a subset of which represent distal lung progenitor cells (LPCs), promote normal lung growth and repair. AEC2 depletion may contribute to persistent lung injury in BPD. We hypothesized that induced pluripotent stem cell (iPSC)-derived AECs prevent lung damage in experimental oxygen-induced BPD. METHODS: Mouse AECs (mAECs), miPSCs/mouse embryonic stem sells, human umbilical cord mesenchymal stromal cells (hUCMSCs), human (h)iPSCs, hiPSC-derived LPCs and hiPSC-derived AECs were delivered intratracheally to hyperoxia-exposed newborn mice. Cells were pre-labeled with a red fluorescent dye for in vivo tracking. RESULTS: Airway delivery of primary mAECs and undifferentiated murine pluripotent cells prevented hyperoxia-induced impairment in lung function and alveolar growth in neonatal mice. Similar to hUCMSC therapy, undifferentiated hiPSCs also preserved lung function and alveolar growth in hyperoxia-exposed neonatal NOD/SCID mice. Long-term assessment of hiPSC administration revealed local teratoma formation and cellular infiltration in various organs. To develop a clinically relevant cell therapy, we used a highly efficient method to differentiate hiPSCs into a homogenous population of AEC2s. Airway delivery of hiPSC-derived AEC2s and hiPSC-derived LPCs, improved lung function and structure and resulted in long-term engraftment without evidence of tumor formation. CONCLUSIONS: hiPSC-derived AEC2 therapy appears effective and safe in this model and warrants further exploration as a therapeutic option for BPD and other lung diseases characterized by AEC injury.


Assuntos
Células Epiteliais Alveolares/citologia , Hiperóxia/complicações , Células-Tronco Pluripotentes Induzidas/citologia , Lesão Pulmonar/etiologia , Lesão Pulmonar/terapia , Animais , Animais Recém-Nascidos , Diferenciação Celular , Modelos Animais de Doenças , Humanos , Células-Tronco Pluripotentes Induzidas/ultraestrutura , Lesão Pulmonar/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Oxigênio , Teratoma/patologia
3.
Pediatr Res ; 79(5): 748-53, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26761124

RESUMO

BACKGROUND: Little is known about the effects of propofol on oxidative stress and its effect on key structures of the contractile apparatus as the myosin light chain 2 (MLC2) and the p38MAPK survival pathway in the preterm heart. We hypothesized that propofol administration could attenuate the hypoxic myocardial injury after birth asphyxia. METHODS: Pregnant ewes were randomized to receive either propofol or isoflurane anesthesia. A total of 44 late-preterm lambs were subjected to in utero umbilical cord occlusion (UCO), resulting in asphyxia and cardiac arrest, or sham treatment. After emergency cesarean delivery, each fetus was resuscitated, mechanically ventilated, and supported under anesthesia for 8 h using the same anesthetic as the one received by its mother. RESULTS: At 8 h after UCO, occurrence of reactive oxygen species and activation of inducible nitric oxide synthase in the heart were lower in association with propofol anesthesia than with isoflurane. This was accompanied by less degradation of MLC2 but higher p38MAPK level and in echocardiography with a trend toward a higher median left ventricular fractional shortening. CONCLUSION: The use of propofol resulted in less oxidative stress and was associated with less cytoskeletal damage of the contractile apparatus than the use of isoflurane anesthesia.


Assuntos
Asfixia/fisiopatologia , Parada Cardíaca/fisiopatologia , Estresse Oxidativo/efeitos dos fármacos , Propofol/administração & dosagem , Animais , Animais Recém-Nascidos , Asfixia Neonatal/fisiopatologia , Miosinas Cardíacas/metabolismo , Ecocardiografia , Feminino , Feto/efeitos dos fármacos , Coração/fisiopatologia , Isoflurano/administração & dosagem , Peroxidação de Lipídeos , Cadeias Leves de Miosina/metabolismo , Gravidez , Prenhez , Distribuição Aleatória , Carneiro Doméstico , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
4.
Am J Respir Cell Mol Biol ; 50(1): 96-105, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23962064

RESUMO

Understanding how alveoli and the underlying capillary network develop and how these mechanisms are disrupted in disease states is critical for developing effective therapies for lung regeneration. Recent evidence suggests that lung angiogenesis promotes lung development and repair. Vascular endothelial growth factor (VEGF) preserves lung angiogenesis and alveolarization in experimental O2-induced arrested alveolar growth in newborn rats, but combined VEGF+angiopoietin 1 treatment is necessary to correct VEGF-induced vessel leakiness. Hypoxia-inducible factors (HIFs) are transcription factors that activate multiple O2-sensitive genes, including those encoding for angiogenic growth factors, but their role during postnatal lung growth is incompletely understood. By inducing the expression of a range of angiogenic factors in a coordinated fashion, HIF may orchestrate efficient and safe angiogenesis superior to VEGF. We hypothesized that HIF inhibition impairs alveolarization and that HIF activation regenerates irreversible O2-induced arrested alveolar growth. HIF inhibition by intratracheal dominant-negative adenovirus (dnHIF-1α)-mediated gene transfer or chetomin decreased lung HIF-1α, HIF-2α, and VEGF expression and led to air space enlargement and arrested lung vascular growth. In experimental O2-induced arrested alveolar growth in newborn rats, the characteristic features of air space enlargement and loss of lung capillaries were associated with decreased lung HIF-1α and HIF-2α expression. Intratracheal administration of Ad.HIF-1α restored HIF-1α, endothelial nitric oxide synthase, VEGF, VEGFR2, and Tie2 expression and preserved and rescued alveolar growth and lung capillary formation in this model. HIFs promote normal alveolar development and may be useful targets for alveolar regeneration.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Hipóxia/metabolismo , Alvéolos Pulmonares/fisiopatologia , Regeneração/fisiologia , Animais , Animais Recém-Nascidos , Neovascularização Patológica/metabolismo , Neovascularização Patológica/fisiopatologia , Óxido Nítrico Sintase Tipo III/metabolismo , Oxigênio/metabolismo , Alvéolos Pulmonares/metabolismo , Ratos , Receptor TIE-2/metabolismo , Fatores de Transcrição/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
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.
Birth Defects Res A Clin Mol Teratol ; 100(3): 217-26, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24619857

RESUMO

Bronchopulmonary dysplasia (BPD) is the most common adverse outcome in extreme preterm neonates (born before 28 weeks gestation). BPD is characterized by interrupted lung growth and may predispose to early-onset emphysema and poor lung function in later life. At present, there is no treatment for BPD. Recent advances in stem/progenitor cell biology have enabled the exploration of endogenous lung progenitor populations in health and disease. In parallel, exogenous stem/progenitor cell administration has shown promise in protecting the lung from injury in the experimental setting. This review will provide an outline of the progenitor populations that have currently been identified in all tissue compartments of the distal lung and how they may be affected in BPD. A thorough understanding of the lung's endogenous progenitor populations during normal development, injury and repair may one day allow us to harness their regenerative capacity.


Assuntos
Displasia Broncopulmonar , Lactente Extremamente Prematuro , Pulmão , Células-Tronco , Displasia Broncopulmonar/metabolismo , Displasia Broncopulmonar/patologia , Displasia Broncopulmonar/fisiopatologia , Humanos , Recém-Nascido , Pulmão/metabolismo , Pulmão/patologia , Pulmão/fisiopatologia
8.
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
9.
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
10.
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
11.
Pediatr Res ; 73(4 Pt 1): 427-34, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23329199

RESUMO

BACKGROUND: Cardiac dysfunction is reported to occur after severe perinatal asphyxia. We hypothesized that anesthesia of the mother with propofol during emergency cesarean section (c-section) would result in less cardiac injury (troponin T) in preterm fetuses exposed to global severe asphyxia in utero than anesthesia with isoflurane. We tested whether propofol decreases the activity of proapoptotic caspase-3 by activating the antiapoptotic AKT kinase family and the signal transducer and activator of transcription-3 (STAT-3). METHODS: Pregnant ewes were randomized to receive either propofol or isoflurane anesthesia. A total of 44 late-preterm lambs were subjected to in utero umbilical cord occlusion (UCO), resulting in asphyxia and cardiac arrest, or sham treatment. After emergency c-section, each fetus was resuscitated, mechanically ventilated, and supported under anesthesia for 8 h using the same anesthetic as the one received by its mother. RESULTS: At 8 h after UCO, the fetuses whose mothers had received propofol anesthesia had lower plasma troponin T levels, and showed a trend toward a higher median left ventricular ejection fraction (LVEF) of 84% as compared with 74% for those whose mothers had received isoflurane. Postasphyxia activation of caspase-3 was lower in association with propofol anesthesia than with isoflurane. Postasphyxia levels of STAT-3 and the AKT kinase family rose 655% and 500%, respectively with the use of propofol anesthesia for the mother. CONCLUSION: The use of propofol for maternal anesthesia results in less cardiac injury in late-preterm lambs subjected to asphyxia than the use of isoflurane anesthesia. The underlying mechanism may be activation of the antiapoptotic STAT-3 and AKT pathways.


Assuntos
Anestésicos Intravenosos/administração & dosagem , Asfixia Neonatal/terapia , Cesárea , Parada Cardíaca/terapia , Miocárdio/patologia , Nascimento Prematuro , Propofol/administração & dosagem , Disfunção Ventricular Esquerda/prevenção & controle , Anestésicos Inalatórios/administração & dosagem , Animais , Animais Recém-Nascidos , Asfixia Neonatal/sangue , Asfixia Neonatal/fisiopatologia , Biomarcadores/sangue , Caspase 3/metabolismo , Modelos Animais de Doenças , Ativação Enzimática , Feminino , Idade Gestacional , Parada Cardíaca/sangue , Parada Cardíaca/fisiopatologia , Humanos , Recém-Nascido , Isoflurano/administração & dosagem , Ligadura , Miocárdio/metabolismo , Fosforilação , Gravidez , Proteínas Proto-Oncogênicas c-akt/metabolismo , Respiração Artificial , Ressuscitação , Fator de Transcrição STAT3/metabolismo , Ovinos , Volume Sistólico/efeitos dos fármacos , Fatores de Tempo , Troponina T/sangue , Ultrassonografia , Cordão Umbilical/cirurgia , Disfunção Ventricular Esquerda/sangue , Disfunção Ventricular Esquerda/diagnóstico por imagem , Disfunção Ventricular Esquerda/fisiopatologia , Função Ventricular Esquerda/efeitos dos fármacos
12.
J Immunol ; 187(5): 2688-95, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21784974

RESUMO

The chorioamnionitis associated with preterm delivery is often polymicrobial with ureaplasma being the most common isolate. To evaluate interactions between the different proinflammatory mediators, we hypothesized that ureaplasma exposure would increase fetal responsiveness to LPS. Fetal sheep were given intra-amniotic (IA) injections of media (control) or Ureaplasma parvum serovar 3 either 7 or 70 d before preterm delivery. Another group received an IA injection of Escherichia coli LPS 2 d prior to delivery. To test for interactions, IA U. parvum-exposed animals were challenged with IA LPS and delivered 2 d later. All animals were delivered at 124 ± 1-d gestation (term = 150 d). Compared with the 2-d LPS exposure group, the U. parvum 70 d + LPS group had 1) decreased lung pro- and anti-inflammatory cytokine expression and 2) fewer CD3(+) T lymphocytes, CCL2(+), myeloperoxidase(+), and PU.1(+) cells in the lung. Interestingly, exposure to U. parvum for 7 d did not change responses to a subsequent IA LPS challenge, and exposure to IA U. parvum alone induced mild lung inflammation. Exposure to U. parvum increased pulmonary TGF-ß1 expression but did not change mRNA expression of either the receptor TLR4 or some of the downstream mediators in the lung. Monocytes from fetal blood and lung isolated from U. parvum 70 d + LPS but not U. parvum 7 d + LPS animals had decreased in vitro responsiveness to LPS. These results are consistent with the novel finding of downregulation of LPS responses by chronic but not acute fetal exposures to U. parvum. The findings increase our understanding of how chorioamnionitis-exposed preterm infants may respond to lung injury and postnatal nosocomial infections.


Assuntos
Imunidade Inata , Pneumonia/imunologia , Complicações Infecciosas na Gravidez/imunologia , Efeitos Tardios da Exposição Pré-Natal/imunologia , Infecções por Ureaplasma/imunologia , Animais , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/citologia , Corioamnionite/imunologia , Infecção Hospitalar/imunologia , Citocinas/análise , Citocinas/biossíntese , Citocinas/imunologia , Feminino , Feto , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/toxicidade , Gravidez , Carneiro Doméstico , Ureaplasma , Infecções por Ureaplasma/complicações
13.
Lung ; 191(1): 77-86, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23117276

RESUMO

BACKGROUND: Recruitment manoeuvres are widely used in clinical practice to open the lung and prevent lung injury by derecruitment, although the evidence is still discussed. In this study two different recruitment manoeuvres were compared to no recruitment manoeuvres (control) in ventilated sheep with acute respiratory distress syndrome (ARDS), induced by lung lavage. METHODS: We performed a prospective, randomised study in 26 ventilated sheep with ARDS, to evaluate the effect of two different recruitment manoeuvres on gas exchange, blood pressure and lung injury. The two different recruitment manoeuvres, the high pressure recruitment manoeuvre (HPRM), with high peak pressure, and the smooth and moderate recruitment manoeuvre (SMRM), with lower peak pressure, were compared to controls (no recruitment) after disconnection. Oxygenation index and ventilation efficacy index were calculated to evaluate gas exchange. Lung injury was assessed by inflammatory response in broncho-alveolar lavage fluid (BALF) and blood and histology of the lung. RESULTS: Oxygenation index improved significantly after both recruitment manoeuvres compared with controls, but no significant difference was found between the recruitment manoeuvres. Blood pressure decreased after HPRM but not after SMRM. HPRM induced a higher number of total cells and more neutrophils in the BALF. In the histology of the lung, mean alveolar size was increased in the dorsocranial region of the lung of SMRM compared to controls. CONCLUSION: Recruitment manoeuvres improved oxygenation, but SMRM was superior, with respect to hemodynamics and pulmonary inflammation, in ventilated sheep suffering from ARDS induced by lung lavage.


Assuntos
Lavagem Broncoalveolar/efeitos adversos , Respiração Artificial/métodos , Síndrome do Desconforto Respiratório/etiologia , Síndrome do Desconforto Respiratório/fisiopatologia , Animais , Pressão Sanguínea/fisiologia , Modelos Animais de Doenças , Feminino , Lesão Pulmonar/patologia , Lesão Pulmonar/fisiopatologia , Respiração com Pressão Positiva , Estudos Prospectivos , Troca Gasosa Pulmonar/fisiologia , Ovinos
14.
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
15.
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
16.
Artif Organs ; 36(6): 512-6, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22309513

RESUMO

The concept of an artificial placenta has been pursued in experimental research since the early 1960s. The principle has yet to be successfully implemented in neonatal care despite the constant evolution in extracorporeal life support technology and advancements in neonatal intensive care in general. For more than three decades, the physical dimensions of the required equipment necessitated pump-driven circuits; however, recent advances in oxygenator technology have allowed exploration of the simpler and physiologically preferable concept of pumpless arteriovenous oxygenation. We expect that further miniaturization of the extracorporeal circuit will allow the implementation of the concept into clinical application as an assist device. To this end, NeonatOx (Fig. 1), a custom-made miniaturized oxygenator with a filling volume of 20 mL, designed by our own group, has been successfully implemented with a preterm lamb model of less than 2000 g body weight as an assist device. We provide an overview of milestones in the history of extracorporeal membrane oxygenation of the preterm newborn juxtaposed against current and future technological advancements. Key limitations, which need to be addressed in order to make mechanical gas exchange a clinical treatment option of prematurity-related lung failure, are also identified.


Assuntos
Órgãos Artificiais/história , Oxigenação por Membrana Extracorpórea/história , Recém-Nascido Prematuro/fisiologia , Placenta/fisiologia , Animais , Cateterismo/história , Oxigenação por Membrana Extracorpórea/instrumentação , Oxigenação por Membrana Extracorpórea/métodos , Feminino , História do Século XX , História do Século XXI , Humanos , Recém-Nascido , Gravidez
17.
Am J Respir Cell Mol Biol ; 45(5): 969-76, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21562314

RESUMO

Bronchopulmonary dysplasia (BPD), associated with chorioamnionitis, results from the simultaneous effects of disrupted lung development, lung injury, and repair superimposed on the developing lung. Caveolins (Cavs) are implicated as major modulators of lung injury and remodeling by multiple signaling pathways, although Cavs have been minimally studied in the injured developing lung. We hypothesized that chorioamnionitis-associated antenatal lung inflammation would decrease the expression of Cav-1 in preterm fetal lungs. We tested whether changes occurred in the transcription factors Smad2/3, Smad1/5, Stat3, and Stat1, and we also studied the activation of acid-sphingomyelinase (a-SMase) with the generation of ceramide, along with changes in the expression of heme oxygenase-1 (HO-1) as indicators of possible Cav-1-mediated effects. Fetal sheep were exposed to 10 mg of intra-amniotic endotoxin or saline for 2, 7, or 2 + 7 days before preterm delivery at 124 days of gestation. The expression of Cav-1 and HO-1 and the phosphorylation of Smad and Stat were evaluated by real-time PCR, Western blotting, and/or immunohistochemistry. The activity of a-SMase and the concentrations of ceramide were measured. Intra-amniotic endotoxin decreased Cav-1 mRNA and protein expression in the lungs, with a maximum reduction of Cav-1 mRNA to 50% ± 7% of the control value (P < 0.05), and of Cav-1 protein expression to 20% ± 5% of the control value (P < 0.05). Decreased concentrations of Cav-1 were associated with the elevated phosphorylation of Smad2/3, Stat3, and Stat1, but not of Smad1/5. The expression of HO-1, a-SMase activity, and ceramide increased. Antenatal inflammation decreased the expression of Cav-1 in the preterm fetal lung. The decreased expression of Cav-1 was associated with the activation of the Smad2/3, Stat, and a-SMase/ceramide pathways, and with the increased expression of HO-1. The decreased concentrations of Cav-1 and changes in other signaling pathways may contribute to BPD.


Assuntos
Caveolina 1/metabolismo , Corioamnionite/metabolismo , Pneumonia/metabolismo , Nascimento Prematuro , Transdução de Sinais , Animais , Animais Recém-Nascidos , Ceramidas/biossíntese , Endotoxinas/farmacologia , Feminino , Feto/metabolismo , Heme Oxigenase-1/biossíntese , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Fosforilação , Gravidez , Ovinos , Esfingomielina Fosfodiesterase/metabolismo , Fatores de Transcrição/metabolismo
18.
Am J Physiol Lung Cell Mol Physiol ; 301(3): L285-95, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21665964

RESUMO

Clinical and epidemiological studies implicate IL-1 as an important mediator of perinatal inflammation. We tested the hypothesis that intra-amniotic IL-1α would induce pulmonary and systemic fetal inflammatory responses. Sheep with singleton fetuses were given an intra-amniotic injection of recombinant sheep IL-1α (100 µg) and were delivered 1, 3, or 7 days later, at 124 ± 1 days gestation (n=5-8/group). A separate group of sheep were given two intra-amniotic IL-1α injections (100 µg dose each): 7 days and again 1 day prior to delivery. IL-1α induced a robust increase in monocytes, neutrophils, lymphocytes, and IL-8 protein in bronchoalveolar lavage fluid. H(2)O(2) secretion was increased in inflammatory cells isolated from lungs of IL-1α-exposed lambs upon LPS challenge in vitro compared with control monocytes. T lymphocytes were recruited to the lung. IL-1ß, cyclooxygenase-1, and cyclooxygenase-2 mRNA expression increased in the lung 1 day after intra-amniotic IL-1α exposure. Lung volumes increased 7 days after intra-amniotic IL-1α exposure, with minimal anatomic changes in air space morphology. The weight of the posterior mediastinal lymph node draining the lung and the gastrointestinal tract doubled, inducible nitric oxide synthase (NOSII)-positive cells increased, and Foxp3-positive T-regulatory lymphocytes decreased in the lymph node after IL-1α exposure. In the blood, neutrophil counts and plasma haptoglobin increased after IL-1α exposure. Compared with a single exposure, exposure to intra-amniotic IL-1α 7 days and again 1 day before delivery had a variable effect (increases in some inflammatory markers, but not pulmonary cytokines). IL-1α is a potent mediator of the fetal inflammatory response syndrome.


Assuntos
Doenças Fetais/imunologia , Feto/imunologia , Inflamação/imunologia , Interleucina-1alfa/farmacologia , Âmnio , Animais , Líquido da Lavagem Broncoalveolar/imunologia , Feminino , Feto/efeitos dos fármacos , Fatores de Transcrição Forkhead/biossíntese , Pulmão , Linfonodos/metabolismo , Mediastinite/patologia , Gravidez , Carneiro Doméstico
19.
Artif Organs ; 35(11): 997-1001, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21995519

RESUMO

Gas exchange in premature neonates is regularly impaired by structural and functional immaturity of the lung. Mechanical ventilation, which is vitally important to sustain oxygenation and CO(2) elimination, causes, at the same time, mechanical and inflammatory destruction of lung tissue. To date, extracorporeal oxygenation is not a treatment option, one reason among others being the size of available oxygenators and cannulas. We hypothesized that a substantial improvement in gas exchange can be achieved by maintenance of the fetal cardiopulmonary bypass and interposition of a suitable passively driven (arteriovenous) membrane oxygenator. In close cooperation between engineers and neonatologists, we developed a miniaturized oxygenator and adapted cannulas to be used as a pumpless extracorporeal lung support that is connected to the circulation via cannulation of the umbilical cord vessels. First in vitro and in vivo studies show promising results. We regard this as one step on the way to clinical application of the artificial placenta.


Assuntos
Cateterismo/instrumentação , Oxigenação por Membrana Extracorpórea/instrumentação , Pulmão/fisiologia , Oxigenadores , Cordão Umbilical/irrigação sanguínea , Animais , Monóxido de Carbono/metabolismo , Desenho de Equipamento , Feminino , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Masculino , Oxigênio/metabolismo , Distribuição Aleatória , Ovinos
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA