Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Neuroinflammation ; 21(1): 16, 2024 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-38200558

RESUMEN

BACKGROUND: Preterm birth is often associated with chorioamnionitis and leads to increased risk of neurodevelopmental disorders, such as autism. Preterm birth can lead to cerebellar underdevelopment, but the mechanisms of disrupted cerebellar development in preterm infants are not well understood. The cerebellum is consistently affected in people with autism spectrum disorders, showing reduction of Purkinje cells, decreased cerebellar grey matter, and altered connectivity. METHODS: Preterm rhesus macaque fetuses were exposed to intra-amniotic LPS (1 mg, E. coli O55:B5) at 127 days (80%) gestation and delivered by c-section 5 days after injections. Maternal and fetal plasma were sampled for cytokine measurements. Chorio-decidua was analyzed for immune cell populations by flow cytometry. Fetal cerebellum was sampled for histology and molecular analysis by single-nuclei RNA-sequencing (snRNA-seq) on a 10× chromium platform. snRNA-seq data were analyzed for differences in cell populations, cell-type specific gene expression, and inferred cellular communications. RESULTS: We leveraged snRNA-seq of the cerebellum in a clinically relevant rhesus macaque model of chorioamnionitis and preterm birth, to show that chorioamnionitis leads to Purkinje cell loss and disrupted maturation of granule cells and oligodendrocytes in the fetal cerebellum at late gestation. Purkinje cell loss is accompanied by decreased sonic hedgehog signaling from Purkinje cells to granule cells, which show an accelerated maturation, and to oligodendrocytes, which show accelerated maturation from pre-oligodendrocytes into myelinating oligodendrocytes. CONCLUSION: These findings suggest a role of chorioamnionitis on disrupted cerebellar maturation associated with preterm birth and on the pathogenesis of neurodevelopmental disorders among preterm infants.


Asunto(s)
Corioamnionitis , Nacimiento Prematuro , Recién Nacido , Femenino , Lactante , Animales , Humanos , Embarazo , Proteínas Hedgehog , Macaca mulatta , Escherichia coli , Recien Nacido Prematuro , Cerebelo , ARN Nuclear Pequeño
2.
Nat Commun ; 14(1): 8452, 2023 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-38114516

RESUMEN

Lung epithelial regeneration after acute injury requires coordination cellular coordination to pattern the morphologically complex alveolar gas exchange surface. During adult lung regeneration, Wnt-responsive alveolar epithelial progenitor (AEP) cells, a subset of alveolar type 2 (AT2) cells, proliferate and transition to alveolar type 1 (AT1) cells. Here, we report a refined primary murine alveolar organoid, which recapitulates critical aspects of in vivo regeneration. Paired scRNAseq and scATACseq followed by transcriptional regulatory network (TRN) analysis identified two AT1 transition states driven by distinct regulatory networks controlled in part by differential activity of Nkx2-1. Genetic ablation of Nkx2-1 in AEP-derived organoids was sufficient to cause transition to a proliferative stressed Krt8+ state, and AEP-specific deletion of Nkx2-1 in adult mice led to rapid loss of progenitor state and uncontrolled growth of Krt8+ cells. Together, these data implicate dynamic epigenetic maintenance via Nkx2-1 as central to the control of facultative progenitor activity in AEPs.


Asunto(s)
Epigenómica , Pulmón , Animales , Ratones , Diferenciación Celular , Células Epiteliales , Homeostasis , Células Madre
3.
Am J Respir Cell Mol Biol ; 68(4): 430-443, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36542853

RESUMEN

Mutations in the FOXF1 (forkhead box F1) gene, encoding the mesenchymal FOX (forkhead box) transcription factor, are linked to alveolar capillary dysplasia with misalignment of pulmonary veins (ACDMPV), a severe congenital disorder associated with the loss of alveolar capillaries and lung hypoplasia. Although proangiogenic functions of FOXF1 have been extensively studied, the role of FOXF1 in mesenchymal-epithelial signaling during lung development remains uncharacterized. Herein, we used murine lung organoids to demonstrate that the S52F FOXF1 mutation (found in patients with ACDMPV) stimulates canonical WNT/ß-catenin signaling in type 2 alveolar epithelial cells (AEC2s), leading to increased proliferation of AEC2s and decreased differentiation of AEC2s into type 1 alveolar epithelial cells (AEC1s). Alveolar organoids containing Foxf1WT/S52F lung fibroblasts and wild-type epithelial cells grew faster on Matrigel and exhibited AEC2 hyperplasia. AEC2 hyperplasia and loss of AEC1s were found in the lungs of Foxf1WT/S52F embryos, a mouse model of ACDMPV. Activation of canonical WNT/ß-catenin signaling in AEC2s of lung organoids and Foxf1WT/S52F mice was associated with decreased expression of noncanonical WNT5A (Wnt family member 5A) ligand in lung fibroblasts. Mechanistically, FOXF1 directly activates the Wnt5a gene transcription through an evolutionarily conserved +6320/+6326 region located in the first intron of the Wnt5a gene. Site-directed mutagenesis of the +6320/+6326 region prevented the transcriptional activation of the Wnt5a enhancer by FOXF1. Treatment with exogenous WNT5A ligand inhibited the effects of the S52F FOXF1 mutation on canonical WNT/ß-catenin signaling in alveolar organoids, preventing aberrant AEC2 expansion and restoring differentiation of AEC1s. Activation of either FOXF1 or WNT5A may provide an attractive strategy to improve lung function in patients with ACDMPV.


Asunto(s)
Factores de Transcripción Forkhead , Síndrome de Circulación Fetal Persistente , Proteína Wnt-5a , Animales , Humanos , Ratones , beta Catenina/genética , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Hiperplasia , Ligandos , Morfogénesis , Activación Transcripcional , Proteína Wnt-5a/genética , Proteína Wnt-5a/metabolismo , Vía de Señalización Wnt
4.
JCI Insight ; 7(18)2022 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-35980752

RESUMEN

Accurate estimate of fetal maturity could provide individualized guidance for delivery of complicated pregnancies. However, current methods are invasive, have low accuracy, and are limited to fetal lung maturation. To identify diagnostic gestational biomarkers, we performed transcriptomic profiling of lung and brain, as well as cell-free RNA from amniotic fluid of preterm and term rhesus macaque fetuses. These data identify potentially new and prior-associated gestational age differences in distinct lung and neuronal cell populations when compared with existing single-cell and bulk RNA-Seq data. Comparative analyses found hundreds of genes coincidently induced in lung and amniotic fluid, along with dozens in brain and amniotic fluid. These data enable creation of computational models that accurately predict lung compliance from amniotic fluid and lung transcriptome of preterm fetuses treated with antenatal corticosteroids. Importantly, antenatal steroids induced off-target gene expression changes in the brain, impinging upon synaptic transmission and neuronal and glial maturation, as this could have long-term consequences on brain development. Cell-free RNA in amniotic fluid may provide a substrate of global fetal maturation markers for personalized management of at-risk pregnancies.


Asunto(s)
Líquido Amniótico , Ácidos Nucleicos Libres de Células , Líquido Amniótico/metabolismo , Animales , Ácidos Nucleicos Libres de Células/metabolismo , Femenino , Desarrollo Fetal , Macaca mulatta , Embarazo , Transcriptoma
5.
Sci Transl Med ; 14(638): eabl8574, 2022 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-35353543

RESUMEN

Perinatal inflammatory stress is associated with early life morbidity and lifelong consequences for pulmonary health. Chorioamnionitis, an inflammatory condition affecting the placenta and fluid surrounding the developing fetus, affects 25 to 40% of preterm births. Severe chorioamnionitis with preterm birth is associated with significantly increased risk of pulmonary disease and secondary infections in childhood, suggesting that fetal inflammation may markedly alter the development of the lung. Here, we used intra-amniotic lipopolysaccharide (LPS) challenge to induce experimental chorioamnionitis in a prenatal rhesus macaque (Macaca mulatta) model that mirrors structural and temporal aspects of human lung development. Inflammatory injury directly disrupted the developing gas exchange surface of the primate lung, with extensive damage to alveolar structure, particularly the close association and coordinated differentiation of alveolar type 1 pneumocytes and specialized alveolar capillary endothelium. Single-cell RNA sequencing analysis defined a multicellular alveolar signaling niche driving alveologenesis that was extensively disrupted by perinatal inflammation, leading to a loss of gas exchange surface and alveolar simplification, with notable resemblance to chronic lung disease in newborns. Blockade of the inflammatory cytokines interleukin-1ß and tumor necrosis factor-α ameliorated LPS-induced inflammatory lung injury by blunting stromal responses to inflammation and modulating innate immune activation in myeloid cells, restoring structural integrity and key signaling networks in the developing alveolus. These data provide new insight into the pathophysiology of developmental lung injury and suggest that modulating inflammation is a promising therapeutic approach to prevent fetal consequences of chorioamnionitis.


Asunto(s)
Corioamnionitis , Nacimiento Prematuro , Animales , Corioamnionitis/inducido químicamente , Corioamnionitis/patología , Femenino , Pulmón/patología , Macaca mulatta , Embarazo , Nacimiento Prematuro/prevención & control , Intercambio Gaseoso Pulmonar
6.
JCI Insight ; 5(24)2020 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-33328385

RESUMEN

Respiratory complicˆations are the major cause of morbidity and mortality among preterm infants, which is partially prevented by the administration of antenatal corticosteroids (ACS). Most very preterm infants are exposed to chorioamnionitis, but short- and long-term effects of ACS treatment in this setting are not well defined. In low-resource settings, ACS increased neonatal mortality by perhaps increasing infection. We report that treatment with low-dose ACS in the setting of inflammation induced by intraamniotic lipopolysaccharide (LPS) in rhesus macaques improves lung compliance and increases surfactant production relative to either exposure alone. RNA sequencing shows that these changes are mediated by suppression of proliferation and induction of mesenchymal cellular death via TP53. The combined exposure results in a mature-like transcriptomic profile with inhibition of extracellular matrix development by suppression of collagen genes COL1A1, COL1A2, and COL3A1 and regulators of lung development FGF9 and FGF10. ACS and inflammation also suppressed signature genes associated with proliferative mesenchymal progenitors similar to the term gestation lung. Treatment with ACS in the setting of inflammation may result in early respiratory advantage to preterm infants, but this advantage may come at a risk of abnormal extracellular matrix development, which may be associated with increased risk of chronic lung disease.


Asunto(s)
Corticoesteroides/farmacología , Pulmón/efectos de los fármacos , Nacimiento Prematuro/tratamiento farmacológico , Corticoesteroides/efectos adversos , Animales , Animales Recién Nacidos , Corioamnionitis/tratamiento farmacológico , Corioamnionitis/genética , Dexametasona/farmacología , Modelos Animales de Enfermedad , Femenino , Glucocorticoides/farmacología , Inflamación/tratamiento farmacológico , Inflamación/genética , Macaca mulatta , Masculino , Embarazo , Surfactantes Pulmonares/farmacología
7.
PLoS One ; 14(9): e0222817, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31536601

RESUMEN

Antenatal corticosteroids (ACS) are standard of care for women at risk of preterm delivery, although choice of drug, dose or route have not been systematically evaluated. Further, ACS are infrequently used in low resource environments where most of the mortality from prematurity occurs. We report proof of principle experiments to test betamethasone-phosphate (Beta-P) or dexamethasone-phosphate (Dex-P) given orally in comparison to the clinical treatment with the intramuscular combination drug beta-phosphate plus beta-acetate in a Rhesus Macaque model. First, we performed pharmacokinetic studies in non-pregnant monkeys to compare blood levels of the steroids using oral dosing with Beta-P, Dex-P and an effective maternal intramuscular dose of the beta-acetate component of the clinical treatment. We then evaluated maternal and fetal blood steroid levels with limited fetal sampling under ultrasound guidance in pregnant macaques. We found that oral Beta is more slowly cleared from plasma than oral Dex. The blood levels of both drugs were lower in maternal plasma of pregnant than in non-pregnant macaques. Using the pharmacokinetic data, we treated groups of 6-8 pregnant monkeys with oral Beta-P, oral Dex-P, or the maternal intramuscular clinical treatment and saline controls and measured pressure-volume curves to assess corticosteroid effects on lung maturation at 5d. Oral Beta-P improved the pressure-volume curves similarly to the clinical treatment. Oral Dex-P gave more variable and nonsignificant responses. We then compared gene expression in the fetal lung, liver and hippocampus between oral Beta-P and the clinical treatment by RNA-sequencing. The transcriptomes were largely similar with small gene expression differences in the lung and liver, and no differences in the hippocampus between the groups. As proof of principle, ACS therapy can be effective using inexpensive and widely available oral drugs. Clinical dosing strategies must carefully consider the pharmacokinetics of oral Beta-P or Dex-P to minimize fetal exposure while achieving the desired treatment responses.


Asunto(s)
Corticoesteroides/administración & dosificación , Betametasona/análogos & derivados , Dexametasona/análogos & derivados , Modelos Animales , Atención Prenatal/métodos , Administración Oral , Corticoesteroides/sangre , Corticoesteroides/farmacocinética , Animales , Betametasona/administración & dosificación , Betametasona/sangre , Betametasona/farmacocinética , Dexametasona/administración & dosificación , Dexametasona/sangre , Dexametasona/farmacocinética , Femenino , Madurez de los Órganos Fetales/efectos de los fármacos , Madurez de los Órganos Fetales/genética , Perfilación de la Expresión Génica/métodos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/embriología , Humanos , Inyecciones Intramusculares , Hígado/efectos de los fármacos , Hígado/embriología , Hígado/metabolismo , Pulmón/efectos de los fármacos , Pulmón/embriología , Pulmón/metabolismo , Macaca mulatta , Embarazo , Nacimiento Prematuro/genética , Nacimiento Prematuro/metabolismo
8.
Pediatr Res ; 86(5): 589-594, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31365919

RESUMEN

BACKGROUND: The use of antenatal corticosteroids (ACS) in low-resource environments is sporadic. Further, drug choice, dose, and route of ACS are not optimized. We report the pharmacokinetics and pharmacodynamics of oral dosing of ACS using a preterm sheep model. METHODS: We measured pharmacokinetics of oral betamethasone-phosphate (Beta-P) and dexamethasone-phosphate (Dex-P) using catheterized pregnant sheep. We compared fetal lung maturation responses of oral Beta-P and Dex-P to the standard treatment with 2 doses of the i.m. mixture of Beta-P and betamethasone-acetate at 2, 5, and 7 days after initiation of ACS. RESULTS: Oral Dex-P had lower bioavailability than Beta-P, giving a lower maximum maternal and fetal concentration. A single oral dose of 0.33 mg/kg of Beta-P was equivalent to the standard clinical treatment assessed at 2 days; 2 doses of 0.16 mg/kg of oral Beta-P were equivalent to the standard clinical treatment at 7 days as assessed by lung mechanics and gas exchange after preterm delivery and ventilation. In contrast, oral Dex-P was ineffective because of its decreased bioavailability. CONCLUSION: Using a sheep model, we demonstrate the use of pharmacokinetics to develop oral dosing strategies for ACS. Oral dosing is feasible and may facilitate access to ACS in low-resource environments.


Asunto(s)
Betametasona/análogos & derivados , Dexametasona/análogos & derivados , Glucocorticoides/administración & dosificación , Ovinos/embriología , Administración Oral , Animales , Betametasona/administración & dosificación , Betametasona/farmacocinética , Disponibilidad Biológica , Dexametasona/administración & dosificación , Dexametasona/farmacocinética , Femenino , Glucocorticoides/farmacocinética , Pulmón/crecimiento & desarrollo , Embarazo
9.
Sci Rep ; 9(1): 9039, 2019 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-31227752

RESUMEN

Antenatal corticosteroids (ANS) are the major intervention to decrease respiratory distress syndrome and mortality from premature birth and are standard of care. The use of ANS is expanding to include new indications and gestational ages, although the recommended dosing was never optimized. The most widely used treatment is two intramuscular doses of a 1:1 mixture of betamethasone-phosphate (Beta-P) and betamethasone-acetate (Beta-Ac) - the clinical drug. We tested in a primate model the efficacy of the slow release Beta-Ac alone for enhancing fetal lung maturation and to reduce fetal corticosteroid exposure and potential toxic effects. Pregnant rhesus macaques at 127 days of gestation (80% of term) were treated with either the clinical drug (0.25 mg/kg) or Beta-Ac (0.125 mg/kg). Beta-Ac alone increased lung compliance and surfactant concentration in the fetal lung equivalently to the clinical drug. By transcriptome analyses the early suppression of genes associated with immune responses and developmental pathways were less affected by Beta-Ac than the clinical drug. Promoter and regulatory analysis prediction identified differentially expressed genes targeted by the glucocorticoid receptor in the lung. At 5 days the clinical drug suppressed genes associated with neuronal development and differentiation in the fetal hippocampus compared to control, while low dose Beta-Ac alone did not. A low dose ANS treatment with Beta-Ac should be assessed for efficacy in human trials.


Asunto(s)
Corticoesteroides/administración & dosificación , Regulación del Desarrollo de la Expresión Génica , Pulmón/efectos de los fármacos , Animales , Femenino , Madurez de los Órganos Fetales/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/embriología , Pulmón/embriología , Macaca mulatta , Masculino , Embarazo , Regiones Promotoras Genéticas , Transcriptoma
10.
Am J Obstet Gynecol ; 218(1): 132.e1-132.e9, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29138038

RESUMEN

BACKGROUND: Antenatal steroids are standard of care for women who are at risk of preterm delivery; however, antenatal steroid dosing and formulation have not been evaluated adequately. The standard clinical 2-dose treatment with betamethasone-acetate+betamethasone-phosphate is more effective than 2 doses of betamethasone-phosphate for the induction of lung maturation in preterm fetal sheep. We hypothesized that the slowly released betamethasone-acetate component induces similar lung maturation to betamethasone-phosphate+betamethasone-acetate with decreased dose and fetal exposure. OBJECTIVE: The purpose of this study was to investigate pharmacokinetics and fetal lung maturation of antenatal betamethasone-acetate in preterm fetal sheep. STUDY DESIGN: Groups of 10 singleton-pregnant ewes received 1 or 2 intramuscular doses 24 hours apart of 0.25 mg/kg/dose of betamethasone-phosphate+betamethasone-acetate (the standard of care dose) or 1 intramuscular dose of 0.5 mg/kg, 0.25 mg/kg, or 0.125 mg/kg of betamethasone-acetate. Fetuses were delivered 48 hours after the first injection at 122 days of gestation (80% of term) and ventilated for 30 minutes, with ventilator settings, compliance, vital signs, and blood gas measurements recorded every 10 minutes. After ventilation, we measured static lung pressure-volume curves and sampled the lungs for messenger RNA measurements. Other groups of pregnant ewes and fetuses were catheterized and treated with intramuscular injections of betamethasone-phosphate 0.125 mg/kg, betamethasone-acetate 0.125 mg/kg, or betamethasone-acetate 0.5 mg/kg. Maternal and fetal betamethasone concentrations in plasma were measured for 24 hours. RESULTS: All betamethasone-treated groups had increased messenger RNA expression of surfactant proteins A, B, and C, ATP-binding cassette subfamily A member 3, and aquaporin-5 compared with control animals. Treatment with 1 dose of intramuscular betamethasone-acetate 0.125mg/kg improved dynamic and static lung compliance, gas exchange, and ventilation efficiency similarly to the standard treatment of 2 doses of 0.25 m/kg of betamethasone-acetate+betamethasone-phosphate. Betamethasone-acetate 0.125 mg/kg resulted in lower maternal and fetal peak plasma concentrations and decreased fetal exposure to betamethasone compared with betamethasone-phosphate 0.125 mg/kg. CONCLUSION: A single dose of betamethasone-acetate results in similar fetal lung maturation as the 2-dose clinical formulation of betamethasone-phosphate+betamethasone-acetate with decreased fetal exposure to betamethasone. A lower dose of betamethasone-acetate may be an effective alternative to induce fetal lung maturation with less risk to the fetus.


Asunto(s)
Betametasona/administración & dosificación , Madurez de los Órganos Fetales/efectos de los fármacos , Glucocorticoides/administración & dosificación , Pulmón/efectos de los fármacos , Subfamilia A de Transportadores de Casete de Unión al ATP/genética , Subfamilia A de Transportadores de Casete de Unión al ATP/metabolismo , Animales , Acuaporina 5/genética , Acuaporina 5/metabolismo , Betametasona/análogos & derivados , Betametasona/farmacocinética , Relación Dosis-Respuesta a Droga , Femenino , Glucocorticoides/farmacocinética , Modelos Animales , Embarazo , Proteínas Asociadas a Surfactante Pulmonar/genética , Proteínas Asociadas a Surfactante Pulmonar/metabolismo , ARN Mensajero/metabolismo , Ovinos
11.
PLoS One ; 12(9): e0184938, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28957335

RESUMEN

BACKGROUND: Intrauterine infection is a primary cause of preterm birth and fetal injury. The pro-inflammatory role of the fetal skin in the setting of intrauterine infection remains poorly characterized. Whether or not inflammation of the fetal skin occurs in primates remains unstudied. Accordingly, we hypothesized that: i) the fetal primate skin would mount a pro-inflammatory response to preterm birth associated pro-inflammatory agents (lipopolysaccharides from Escherichia coli, live Ureaplasma parvum, interleukin-1ß) and; ii) that inhibiting interleukin-1 signaling would decrease the skin inflammatory response. METHODS: Rhesus macaques with singleton pregnancies received intraamniotic injections of either sterile saline (control) or one of three pro-inflammatory agonists: E. coli lipopolysaccharides, interluekin-1ß or live U. parvum under ultrasound guidance. A fourth group of animals received both E. coli lipopolysaccharide and interleukin-1 signaling inhibitor interleukin-1 receptor antagonist (Anakinra) prior to delivery. Animals were surgically delivered at approximately 130 days' gestational age. RESULTS: Intraamniotic lipopolysaccharide caused an inflammatory skin response characterized by increases in interluekin-1ß,-6 and -8 mRNA at 16 hours. There was a modest inflammatory response to U. parvum, but interleukin-1ß alone caused no inflammatory response in the fetal skin. Intraamniotic Anakinra treatment of lipopolysaccharide-exposed animals significantly reduced skin inflammation. CONCLUSIONS: Intraamniotic lipopolysaccharide and U. parvum were associated with modest increases in the expression of inflammatory mediators in primate fetal skin. Although administration of Interleukin-1ß alone did not elicit an inflammatory response, lipopolysaccharide-driven skin inflammation was decreased following intraamniotic Anakinra therapy. These findings provide support for the role of the fetal skin in the development of the fetal inflammatory response.


Asunto(s)
Corioamnionitis/patología , Feto/patología , Inflamación/patología , Piel/patología , Animales , Corioamnionitis/genética , Modelos Animales de Enfermedad , Femenino , Inflamación/complicaciones , Inflamación/genética , Mediadores de Inflamación/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , Queratinas/metabolismo , Lipopolisacáridos , Macaca mulatta , Masculino , Reacción en Cadena de la Polimerasa , Embarazo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ureaplasma/fisiología
12.
Pediatr Res ; 81(3): 496-503, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27861467

RESUMEN

BACKGROUND: Dexamethasone-phosphate (Dex-PO4) and the combination betamethasone-phosphate (Beta-PO4) + betamethasone-acetate (Beta-Ac) are the most used antenatal corticosteroids to promote fetal lung maturation. We compared fetal lung maturation induced by Beta-Ac+Beta-PO4, Dex-PO4, or Beta-PO4 alone. METHODS: Pregnant ewes received two intramuscular doses 24 h apart of 0.25 mg/kg/dose of Beta-Ac+Beta-PO4, Dex-PO4 or Beta-PO4; or 2 doses of 0.125 mg/kg/dose of Beta-PO4 at 6, 12, or 24 h intervals. Fetuses were delivered 48 h after the first dose and ventilated for 30 min. We assessed ventilatory variables, vital signs, and blood gas. After ventilation pressure-volume curves were measured and lungs were sampled for analysis. RESULTS: All treatments improved lung compliance and ventilation efficiency. Only Beta-Ac + Beta-PO4 required lower positive inspiratory pressure compared with control. Beta-Ac + Beta-PO4 and Beta-PO4 alone, but not Dex-PO4, increased the mRNA of surfactant proteins compared with control. Low-dose Beta-PO4 did not increase mRNA of surfactant proteins. There were no differences among Beta-PO4 treatment intervals. CONCLUSION: Beta-Ac + Beta-PO4 given as two doses 24 h apart was more effective in promoting fetal lung maturation than Dex-PO4 or Beta-PO4 alone, consistent with a prolonged exposure provided by the Beta-Ac + Beta-PO4. These results support the clinical use of combined Beta-Ac + Beta-PO4 preparations over phosphate corticosteroids alone for fetal lung maturation.


Asunto(s)
Betametasona/administración & dosificación , Dexametasona/administración & dosificación , Pulmón/efectos de los fármacos , Pulmón/embriología , Corticoesteroides/administración & dosificación , Corticoesteroides/uso terapéutico , Animales , Betametasona/análogos & derivados , Betametasona/uso terapéutico , Presión Sanguínea , Dexametasona/uso terapéutico , Femenino , Madurez de los Órganos Fetales , Edad Gestacional , Glucocorticoides/administración & dosificación , Glucocorticoides/uso terapéutico , Inyecciones Intramusculares , Pulmón/fisiología , Masculino , Embarazo , Ovinos , Oveja Doméstica , Factores de Tiempo
13.
J Neuroinflammation ; 13(1): 238, 2016 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-27596440

RESUMEN

BACKGROUND: Chorioamnionitis is associated with an increased risk of brain injury in preterm neonates. Inflammatory changes in brain could underlie this injury. Here, we evaluated whether neuroinflammation is induced by chorioamnionitis in a clinically relevant model. METHODS: Rhesus macaque fetuses were exposed to either intra-amniotic (IA) saline, or IA lipopolysaccharide (LPS) (1 mg) 16 or 48 h prior to delivery at 130 days (85 % of gestation) (n = 4-5 animals/group). We measured cytokines in the cerebrospinal fluid (CSF), froze samples from the left brain for molecular analysis, and immersion fixed the right brain hemisphere for immunohistology. We analyzed the messenger RNA (mRNA) levels of the pro-inflammatory cytokines IL-1ß, CCL2, TNF-α, IL-6, IL-8, IL-10, and COX-2 in the periventricular white matter (PVWM), cortex, thalamus, hippocampus, and cerebellum by RT-qPCR. Brain injury was assessed by immunohistology for myelin basic protein (MBP), IBA1 (microglial marker), GFAP (astrocyte marker), OLIG2 (oligodendrocyte marker), NeuN (neuronal marker), CD3 (T cells), and CD14 (monocytes). Microglial proliferation was assessed by co-immunostaining for IBA1 and Ki67. Data were analyzed by ANOVA with Tukey's post-test. RESULTS: IA LPS increased mRNA expression of pro-inflammatory cytokines in the PVWM, thalamus, and cerebellum, increased IL-6 concentration in the CSF, and increased apoptosis in the periventricular area after 16 h. Microglial proliferation in the white matter was increased 48 h after IA LPS. CONCLUSIONS: LPS-induced chorioamnionitis caused neuroinflammation, microglial proliferation, and periventricular apoptosis in a clinically relevant model of chorioamnionitis in fetal rhesus macaques. These findings identify specific responses in the fetal brain and support the hypothesis that neuroinflammatory changes may mediate the adverse neurodevelopmental outcomes associated with chorioamnionitis.


Asunto(s)
Corioamnionitis/inducido químicamente , Citocinas/metabolismo , Lipopolisacáridos/toxicidad , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Animales , Apoptosis/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/patología , Proteínas de Unión al Calcio , Corioamnionitis/patología , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Citocinas/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Antígeno Ki-67/metabolismo , Macaca mulatta , Proteínas de Microfilamentos , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Proteína Básica de Mielina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Embarazo , Prostaglandina-E Sintasas/metabolismo , ARN Mensajero/metabolismo , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Factores de Tiempo
14.
Am J Obstet Gynecol ; 214(5): 627.e1-627.e16, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26965447

RESUMEN

BACKGROUND: Preterm birth (PTB) is a leading cause of neonatal morbidity and mortality and is not uncommonly associated with chorioamnionitis. We recently have demonstrated that the placenta harbors a unique microbiome with similar flora to the oral community. We also have shown an association of these placental microbiota with PTB, history of antenatal infection, and excess maternal weight gain. On the basis of these previous observations, we hypothesized that the placental membranes would retain a microbiome community that would vary in association with preterm birth and chorioamnionitis. OBJECTIVE: In the current study, we aimed to examine the differences in the placental membrane microbiome in association with PTB in both the presence and absence of chorioamnionitis and/or funisitis using state-of-the-science whole-genome shotgun metagenomics. STUDY DESIGN: This was a cross-sectional analysis with 6 nested spontaneous birth cohorts (n = 9-15 subjects/cohort): Term gestations without chorioamnionitis, term with chorioamnionitis, preterm without chorioamnionitis, preterm with mild chorioamnionitis, preterm with severe chorioamnionitis, and preterm with chorioamnionitis and funisitis. Histologic analysis was performed with Redline's criteria, and inflammatory cytokines were analyzed in the cord blood. DNA from placental membranes was extracted from sterile swabs collected at delivery, and whole-genome shotgun sequencing was performed on the Illumina HiSeq platform. Filtered microbial DNA sequences were annotated and analyzed with MG-RAST (ie, Metagenomic Rapid Annotations using Subsystems Technology) and R. RESULTS: Subjects were assigned to cohorts on the basis of gestational age at delivery and independent scoring of histologic chorioamnionitis. We found that preterm subjects with severe chorioamnionitis and funisitis had increases in cord blood inflammatory cytokines. Of interest, although the placental membrane microbiome was altered in association with severity of histologic chorioamnionitis (permutational multivariate analysis of variance P = .005), there was no observable impact with either betamethasone or antibiotic treatment. In preterm subjects with chorioamnionitis, we found a high abundance of both urogenital and oral commensal bacteria. These alterations in the microbiome were accompanied by significant variation (P < .05) in microbial metabolic pathways important in the glucose-fed pentose phosphate pathway (term subjects), or glycerophopholipid metabolism, and the biosynthesis of the siderophore group nonribosomal peptides (preterm subjects). CONCLUSION: Consistent with ours and others previous findings, women who experienced spontaneous PTB harbor placental microbiota that further differed by severity of chorioamnionitis. Integrative metagenomic analysis revealed significant variation in distinct bacterial metabolic pathways, which we speculate may contribute to risk of preterm birth with and without severe chorioamnionitis.


Asunto(s)
Corioamnionitis/microbiología , Microbiota , Placenta/microbiología , Nacimiento Prematuro , Butiratos/metabolismo , Estudios Transversales , ADN Bacteriano/genética , Femenino , Glicerofosfolípidos/metabolismo , Humanos , Metagenómica , Vía de Pentosa Fosfato , Embarazo , Riboflavina/metabolismo , Análisis de Secuencia de ADN , Índice de Severidad de la Enfermedad , Nacimiento a Término
15.
Reprod Sci ; 23(1): 69-80, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26156854

RESUMEN

UNLABELLED: To understand the changes in the structural integrity of fetal membranes during intrauterine inflammation, we evaluated the time course of expression and localization of damage-associated molecular patterns (DAMPs) and injury/remodeling in collagen and vascular smooth muscle. Time-mated ewes received intra-amniotic (IA) saline or IA lipopolysaccharide (LPS) for 5 hours to 15 days prior to a preterm delivery at 125 ± 2 days (n = 5-7 animals/group). The DAMP high mobility group box 1 (HMGB1) protein assessed by Western blot was induced within 24 hours after IA LPS in the fetal membranes, and HMGB1 expression was localized to amnion epithelium, chorion vascular endothelium, and infiltrating inflammatory cells by immunohistology. Markers of vascular injury, intercellular adhesion molecule 1, and tissue plasminogen activator messenger RNA (mRNA) expression increased 5 to 12 hours after IA LPS in the chorioamnion indicating vascular injury. Chorion vascular remodeling with increased chorion arteriolar smooth muscle actin expression by morphometric analyses of immunohistology was noted 15 days after IA LPS. Collagen expression was nonhomogeneous by histochemical staining, and there was a trend toward decreased mRNA expression of collagen subunit COL5A1 after IA LPS. CONCLUSIONS: Intrauterine inflammation induced early increases in HMGB1 in the chorioamnion with a concomitant vascular injury followed by chorion arteriolar hypertrophy. There was nonhomogeneous collagen expression in the chorioamnion. These results have implications for understanding the pathogenesis of IA inflammation-induced preterm rupture of membranes.


Asunto(s)
Alarminas/metabolismo , Colágeno/metabolismo , Membranas Extraembrionarias/metabolismo , Inflamación/metabolismo , Amnios/metabolismo , Amnios/patología , Líquido Amniótico/metabolismo , Animales , Membranas Extraembrionarias/patología , Femenino , Proteína HMGB1/metabolismo , Inflamación/inducido químicamente , Inflamación/patología , Molécula 1 de Adhesión Intercelular/metabolismo , Lipopolisacáridos , Embarazo , Ovinos , Activador de Tejido Plasminógeno/metabolismo
16.
PLoS One ; 10(10): e0140701, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26473607

RESUMEN

INTRODUCTION: Ex-vivo uterine environment (EVE) therapy uses an artificial placenta to provide gas exchange and nutrient delivery to a fetus submerged in an amniotic fluid bath. Development of EVE may allow us to treat very premature neonates without mechanical ventilation. Meanwhile, elevations in fetal inflammation are associated with adverse neonatal outcomes. In the present study, we analysed fetal survival, inflammation and pulmonary maturation in preterm lambs maintained on EVE therapy using a parallelised umbilical circuit system with a low priming volume. METHODS: Ewes underwent surgical delivery at 115 days of gestation (term is 150 days), and fetuses were transferred to EVE therapy (EVE group; n = 5). Physiological parameters were continuously monitored; fetal blood samples were intermittently obtained to assess wellbeing and targeted to reference range values for 2 days. Age-matched animals (Control group; n = 6) were surgically delivered at 117 days of gestation. Fetal blood and tissue samples were analysed and compared between the two groups. RESULTS: Fetal survival time in the EVE group was 27.0 ± 15.5 (group mean ± SD) hours. Only one fetus completed the pre-determined study period with optimal physiological parameters, while the other 4 animals demonstrated physiological deterioration or death prior to the pre-determined study end point. Significant elevations (p<0.05) in: i) inflammatory proteins in fetal plasma; ii) selected cytokine/chemokine mRNA expression levels in fetal tissues; and iii) histological inflammatory score in fetal lung, were observed in the EVE group compared to the Control group. There was no significant difference (p>0.05) in surfactant protein mRNA expression level between the two groups. CONCLUSION: In this study, we achieved limited fetal survival using EVE therapy. Despite this, EVE therapy only induced a modest fetal inflammatory response and did not promote lung maturation. These data provide additional insight into markers of treatment efficacy for the assessment of future studies.


Asunto(s)
Órganos Artificiales , Enfermedades Fetales/terapia , Placenta , Nacimiento Prematuro/terapia , Animales , Modelos Animales de Enfermedad , Femenino , Enfermedades Fetales/patología , Inflamación/patología , Inflamación/terapia , Embarazo , Nacimiento Prematuro/patología , Ovinos
17.
Pediatr Res ; 77(6): 740-8, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25760552

RESUMEN

BACKGROUND: Intrauterine Candida albicans infection causes severe fetal inflammatory responses and fetal injury in an ovine model. We hypothesized that intra-amniotic antifungal therapy with fluconazole would decrease the adverse fetal effects of intra-amniotic C. albicans in sheep. METHODS: Sheep received an intra-amniotic injection of 10(7) colony-forming units C. albicans. After 2 d, animals were then randomized to: (i) intra-amniotic and fetal intraperitoneal saline with delivery after 24 h (3 d C. albicans group); (ii) intra-amniotic and fetal intraperitoneal injections of fluconazole with delivery after either 24 h (3 d C. albicans plus 1 d fluconazole group) or 72 h (5 d C. albicans plus 3 d fluconazole group). Controls received intra-amniotic injections of saline followed by intra-amniotic and fetal intraperitoneal fluconazole injections. RESULTS: Intra-amniotic C. albicans caused severe fetal inflammatory responses characterized by decreases in lymphocytes and platelets, an increase in posterior mediastinal lymph node weight and proinflammatory mRNA responses in the fetal lung, liver, and spleen. Fluconazole treatment temporarily decreased the pulmonary and chorioamnion inflammatory responses. CONCLUSION: The severe fetal inflammatory responses caused by intra-amniotic C. albicans infection were transiently decreased with fluconazole. A timely fetal delivery of antimicrobial agents may prevent fetal injury associated with intrauterine infection.


Asunto(s)
Antifúngicos/uso terapéutico , Candidiasis/tratamiento farmacológico , Enfermedades Fetales/tratamiento farmacológico , Enfermedades Fetales/microbiología , Fluconazol/uso terapéutico , Inflamación/tratamiento farmacológico , Análisis de Varianza , Animales , Candidiasis/patología , Femenino , Técnicas Histológicas , Inflamación/patología , Embarazo , Ovinos
18.
PLoS One ; 10(3): e0119281, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25793992

RESUMEN

BACKGROUND: A fetal inflammatory response (FIR) in sheep can be induced by intraamniotic or selective exposure of the fetal lung or gut to lipopolysaccharide (LPS). The oral, nasal, and pharyngeal cavities (ONP) contain lymphoid tissue and epithelium that are in contact with the amniotic fluid. The ability of the ONP epithelium and lymphoid tissue to initiate a FIR is unknown. OBJECTIVE: To determine if FIR occurs after selective ONP exposure to LPS in fetal sheep. METHODS: Using fetal recovery surgery, we isolated ONP from the fetal lung, GI tract, and amniotic fluid by tracheal and esophageal ligation and with an occlusive glove fitted over the snout. LPS (5 mg) or saline was infused with 24 h Alzet pumps secured in the oral cavity (n = 7-8/group). Animals were delivered 1 or 6 days after initiation of the LPS or saline infusions. RESULTS: The ONP exposure to LPS had time-dependent systemic inflammatory effects with changes in WBC in cord blood, an increase in posterior mediastinal lymph node weight at 6 days, and pro-inflammatory mRNA responses in the fetal plasma, lung, and liver. Compared to controls, the expression of surfactant protein A mRNA increased 1 and 6 days after ONP exposure to LPS. CONCLUSION: ONP exposure to LPS alone can induce a mild FIR with time-dependent inflammatory responses in remote fetal tissues not directly exposed to LPS.


Asunto(s)
Corioamnionitis/etiología , Lipopolisacáridos/efectos adversos , Boca/inmunología , Nariz/inmunología , Faringe/inmunología , Animales , Biomarcadores , Citocinas/sangre , Citocinas/metabolismo , Femenino , Mediadores de Inflamación/sangre , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/administración & dosificación , Hígado/inmunología , Hígado/metabolismo , Hígado/patología , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/patología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/patología , Embarazo , Ovinos
19.
Pediatr Res ; 76(5): 441-7, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25105257

RESUMEN

BACKGROUND: Damage-associated molecular patterns (DAMPs) and antimicrobial peptides (AMPs) are components of pulmonary innate immunity and tissue repair. We hypothesized that DAMPs and AMPs would increase in response to fetal pulmonary inflammation caused by chorioamnionitis in a time-dependent manner. METHODS: Fetal sheep were exposed to intra-amniotic saline or lipopolysaccharide (LPS) (10 mg) between 5 h and 15 d prior to preterm delivery at 125 ± 2 d. Lung tissue mRNAs for proinflammatory cytokines; AMPs: myeloid AMP-29 (MAP29), dodecapeptide, sheep ß-defensin-1 (SBD1), and sheep ß-defensin-2 (SBD2); and DAMPs: interleukin (IL)-1α, lactoferrin, heat-shock protein-70 (HSP70), high-mobility group box protein-B1 (HMGB1), and receptor for advanced glycation endproducts (RAGE) were measured by reverse-transcriptase quantitative polymerase chain reaction. Immunohistochemistry of DAMPs and in situ hybridization of AMPs was performed. RESULTS: IL-1α, IL-1ß, IL-6, IL-8, IL-10, MCP-1, and tumor necrosis factor (TNF)-α mRNA increased after LPS exposure. MAP29, dodecapeptide, SBD1, and SBD2 mRNA were suppressed at 24 h. MAP29 and dodecapeptide mRNA then increased at 8 d. Lactoferrin increased at 24 h. There were no changes for HMGB1, HSP70, or RAGE. MAP29 and dodecapeptide localized to alveolar cells, increased 8 d after exposure to LPS. CONCLUSION: AMPs are initially suppressed in the fetal lung by LPS-induced chorioamnionitis. The late induction of MAP29 and dodecapeptide may be related to lung repair.


Asunto(s)
Líquido Amniótico/metabolismo , Péptidos Catiónicos Antimicrobianos/metabolismo , Corioamnionitis/metabolismo , Lipopolisacáridos , Pulmón/metabolismo , Animales , Péptidos Catiónicos Antimicrobianos/genética , Corioamnionitis/inducido químicamente , Corioamnionitis/genética , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Edad Gestacional , Mediadores de Inflamación/metabolismo , Pulmón/crecimiento & desarrollo , Embarazo , Nacimiento Prematuro , ARN Mensajero/metabolismo , Ovinos , Factores de Tiempo
20.
Pediatr Res ; 75(6): 716-22, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24632681

RESUMEN

BACKGROUND: Preventing preterm birth and subsequent adverse neonatal sequelae is among the greatest clinical challenges of our time. Recent studies suggest a role for Candida spp. in preterm birth and fetal injury, as a result of their colonization of either the vagina and/or the amniotic cavity. We hypothesized that intraamniotic Candida albicans would cause a vigorous, acute fetal inflammatory response. METHODS: Sheep carrying singleton pregnancies received single intraamniotic injections of either saline (control) or 10(7) colony-forming units C. albicans 1 or 2 d prior to surgical delivery and euthanasia at 124 ± 2 d gestation. RESULTS: Colonization of the amniotic cavity by C. albicans resulted in a modest inflammatory response at 1 d and florid inflammation at 2 d, characterized by fetal thrombocytopenia, lymphopenia, and significant increases of inflammatory cytokines/chemokines in the fetal membranes skin, lung, and the amniotic fluid. CONCLUSION: Acute colonization of the amniotic cavity by C. albicans causes severe intrauterine inflammation and fetal injury. C. albicans is a potent fetal pathogen that can contribute to adverse pregnancy outcomes.


Asunto(s)
Candida albicans , Candidiasis/veterinaria , Feto/microbiología , Inflamación/fisiopatología , Enfermedades de las Ovejas/microbiología , Enfermedades de las Ovejas/fisiopatología , Útero/fisiopatología , Análisis de Varianza , Animales , Candidiasis/fisiopatología , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Feto/metabolismo , Hidrocortisona/sangre , Pulmón/patología , Embarazo , ARN Mensajero/metabolismo , Ovinos , Piel/patología , Útero/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...