Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Respir Res ; 16: 4, 2015 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-25591994

RESUMEN

BACKGROUND: Earlier studies have reported that transforming growth factor beta 1(TGFß1) is a critical mediator of hyperoxia-induced acute lung injury (HALI) in developing lungs, leading to impaired alveolarization and a pulmonary phenotype of bronchopulmonary dysplasia (BPD). However, the mechanisms responsible for the TGFß1-induced inflammatory signals that lead to cell death and abnormal alveolarization are poorly understood. We hypothesized that TGFß1 signaling via TGFßR2 is necessary for the pathogenesis of the BPD pulmonary phenotype resulting from HALI. METHODS: We utilized lung epithelial cell-specific TGFß1 overexpressing transgenic and TGFßR2 null mutant mice to evaluate the effects on neonatal mortality as well as pulmonary inflammation and apoptosis in developing lungs. Lung morphometry was performed to determine the impaired alveolarization and multicolor flow cytometry studies were performed to detect inflammatory macrophages and monocytes in lungs. Apoptotic cell death was measured with TUNEL assay, immunohistochemistry and western blotting and protein expression of angiogenic mediators were also analyzed. RESULTS: Our data reveals that increased TGFß1 expression in newborn mice lungs leads to increased mortality, macrophage and immature monocyte infiltration, apoptotic cell death specifically in Type II alveolar epithelial cells (AECs), impaired alveolarization, and dysregulated angiogenic molecular markers. CONCLUSIONS: Our study has demonstrated the potential role of inhibition of TGFß1 signaling via TGFßR2 for improved survival, reduced inflammation and apoptosis that may provide insights for the development of potential therapeutic strategies targeted against HALI and BPD.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Apoptosis , Pulmón/metabolismo , Neumonía/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Factor de Crecimiento Transformador beta1/biosíntesis , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/patología , Lesión Pulmonar Aguda/fisiopatología , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/patología , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Genotipo , Humanos , Hiperoxia/complicaciones , Pulmón/patología , Pulmón/fisiopatología , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Fenotipo , Neumonía/genética , Neumonía/patología , Neumonía/fisiopatología , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/deficiencia , Receptores de Factores de Crecimiento Transformadores beta/genética , Transducción de Señal , Factores de Tiempo , Factor de Crecimiento Transformador beta1/genética , Regulación hacia Arriba
2.
Am J Respir Cell Mol Biol ; 48(6): 749-57, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23470621

RESUMEN

We noted a marked increase in cyclooxygenase-2 (Cox2) and the activation of the endoplasmic reticulum (ER) stress pathway in newborn murine lung on exposure to hyperoxia and IFN-γ. We sought to evaluate Cox2-mediated ER stress pathway activation in hyperoxia-induced and IFN-γ-mediated injury in developing lungs. We applied in vivo genetic gain-of-function and genetic/chemical inhibition, as well as in vitro loss-of-function genetic strategies. Hyperoxia-induced and IFN-γ-mediated impaired alveolarization was rescued by Cox2 inhibition, using celecoxib. The use of small interfering RNA against the ER stress pathway mediator, the C/EBP homologous protein (CHOP; also known as growth arrest and DNA damage-inducible gene 153/GADD153), alleviated cell death in alveolar epithelial cells as well as in hyperoxia-induced and IFN-γ-mediated murine models of bronchopulmonary dysplasia (BPD). In addition, CHOP siRNA also restored alveolarization in the in vivo models. Furthermore, as evidence of clinical relevance, we show increased concentrations of Cox2 and ER stress pathway mediators in human lungs with BPD. Cox2, via CHOP, may significantly contribute to the final common pathway of hyperoxia-induced and IFN-γ-mediated injury in developing lungs and human BPD.


Asunto(s)
Ciclooxigenasa 2/metabolismo , Estrés del Retículo Endoplásmico , Hiperoxia/patología , Interferón gamma/metabolismo , Animales , Animales Recién Nacidos , Displasia Broncopulmonar/metabolismo , Displasia Broncopulmonar/patología , Celecoxib , Muerte Celular , Ciclooxigenasa 2/genética , Inhibidores de la Ciclooxigenasa 2/farmacología , Humanos , Inmunohistoquímica , Recién Nacido , Interferón gamma/genética , Pulmón/efectos de los fármacos , Pulmón/embriología , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Pirazoles/farmacología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Sulfonamidas/farmacología , Factor de Transcripción CHOP/genética , Factor de Transcripción CHOP/metabolismo
3.
Respir Res ; 14: 27, 2013 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-23448134

RESUMEN

BACKGROUND: The role and mechanism of action of MIF in bronchopulmonary dysplasia (BPD) are not known. We hypothesized that increased MIF signaling would ameliorate the pulmonary phenotype of BPD in the mouse lung. METHODS: We studied newborn wild type (WT), MIF knockout (MIFKO), and lung MIF transgenic (MIFTG) mice in room air and a BPD model, and examined the effects of administering a small molecule MIF agonist and antagonist. Lung morphometry was performed and mRNA and protein expression of vascular mediators were analyzed. RESULTS: The pulmonary phenotype of MIFKO and MIFTG mice lungs in room air (RA) and BPD model were comparable to the WT-BPD mice at postnatal (PN) day 14. Vascular endothelial growth factor (VEGF)-A, -R1 and Angiopoietin (Ang)1 mRNA were decreased, and Ang2 increased in the WT-BPD, MIFKO-RA, MIFKO-BPD, MIFTG-RA and MIFTG-BPD mice lungs, compared to appropriate controls. The protein expression of Ang1 in the MIFKO-RA was similar to WT-RA, but decreased in MIFTG-RA, and decreased in all the BPD groups. Ang2 was increased in MIFKO-RA, MIFTG-RA and in all 3 BPD groups. Tie2 was increased in WT-BPD compared to WT-RA, but decreased in MIFKO- and MIFTG- RA and BPD groups. VEGFR1 was uniformly decreased in MIFKO-RA, MIFTG-RA and in all 3 BPD groups. VEGF-A had a similar expression across all RA and BPD groups. There was partial recovery of the pulmonary phenotype in the WT-BPD model treated with the MIF agonist, and in the MIFTG mice treated with the MIF antagonist. CONCLUSIONS: These data point to the careful regulatory balance exerted by MIF in the developing lung and response to hyperoxia and support the potential therapeutic value of small molecule MIF modulation in BPD.


Asunto(s)
Displasia Broncopulmonar/etiología , Displasia Broncopulmonar/metabolismo , Factores Quimiotácticos/metabolismo , Hiperoxia/complicaciones , Hiperoxia/metabolismo , Pulmón/metabolismo , Macrófagos/inmunología , Animales , Animales Recién Nacidos , Humanos , Recién Nacido , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Regulación hacia Arriba
4.
Mediators Inflamm ; 2013: 457189, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24347826

RESUMEN

RATIONALE: Hyperoxia exposure to developing lungs-critical in the pathogenesis of bronchopulmonary dysplasia-may augment lung inflammation by inhibiting anti-inflammatory mediators in alveolar macrophages. OBJECTIVE: We sought to determine the O2-induced effects on the polarization of macrophages and the role of anti-inflammatory BRP-39 in macrophage phenotype and neonatal lung injury. METHODS: We used RAW264.7, peritoneal, and bone marrow derived macrophages for polarization (M1/M2) studies. For in vivo studies, wild-type (WT) and BRP-39(-/-) mice received continuous exposure to 21% O2 (control mice) or 100% O2 from postnatal (PN) 1 to PN7 days, along with intranasal lipopolysaccharide (LPS) administered on alternate days (PN2, -4, and -6). Lung histology, bronchoalveolar lavage (BAL) cell counts, BAL protein, and cytokines measurements were performed. MEASUREMENTS AND MAIN RESULTS: Hyperoxia differentially contributed to macrophage polarization by enhancing LPS induced M1 and inhibiting interleukin-4 induced M2 phenotype. BRP-39 absence led to further enhancement of the hyperoxia and LPS induced M1 phenotype. In addition, BRP-39(-/-) mice were significantly more sensitive to LPS plus hyperoxia induced lung injury and mortality compared to WT mice. CONCLUSIONS: These findings collectively indicate that BRP-39 is involved in repressing the M1 proinflammatory phenotype in hyperoxia, thereby deactivating inflammatory responses in macrophages and preventing neonatal lung injury.


Asunto(s)
Glicoproteínas/fisiología , Hiperoxia/complicaciones , Inflamación/complicaciones , Lesión Pulmonar/etiología , Animales , Animales Recién Nacidos , Polaridad Celular , Proteína 1 Similar a Quitinasa-3 , Lipopolisacáridos/toxicidad , Macrófagos , Ratones , Ratones Endogámicos C57BL , Fenotipo
5.
Biochemistry ; 51(28): 5642-54, 2012 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-22686371

RESUMEN

MIP-2/CXCL2 is a murine chemokine related to human chemokines that possesses the Glu-Leu-Arg (ELR) activation motif and activates CXCR2 for neutrophil chemotaxis. We determined the structure of MIP-2 to 1.9 Å resolution and created a model with its murine receptor CXCR2 based on the coordinates of human CXCR4. Chemokine-induced migration of cells through specific G-protein coupled receptors is regulated by glycosaminoglycans (GAGs) that oligomerize chemokines. MIP-2 GAG-binding residues were identified that interact with heparin disaccharide I-S by NMR spectroscopy. A model GAG/MIP-2/CXCR2 complex that supports a 2:2 complex between chemokine and receptor was created. Mutants of these disaccharide-binding residues were made and tested for heparin binding, in vitro neutrophil chemotaxis, and in vivo neutrophil recruitment to the mouse peritoneum and lung. The mutants have a 10-fold decrease in neutrophil chemotaxis in vitro. There is no difference in neutrophil recruitment between wild-type MIP-2 and mutants in the peritoneum, but all activity of the mutants is lost in the lung, supporting the concept that GAG regulation of chemokines is tissue-dependent.


Asunto(s)
Quimiocina CXCL2/química , Glicosaminoglicanos/química , Receptores de Interleucina-8B/química , Alanina/genética , Animales , Líquido del Lavado Bronquioalveolar/citología , Células Cultivadas , Quimiocina CXCL2/genética , Quimiocina CXCL2/metabolismo , Quimiotaxis de Leucocito , Cristalografía por Rayos X , Disacáridos/química , Femenino , Glicosaminoglicanos/metabolismo , Heparina/análogos & derivados , Heparina/química , Humanos , Pulmón/citología , Pulmón/inmunología , Ratones , Ratones Endogámicos BALB C , Modelos Moleculares , Mutación , Neutrófilos/inmunología , Neutrófilos/fisiología , Resonancia Magnética Nuclear Biomolecular , Cavidad Peritoneal/citología , Unión Proteica , Conformación Proteica , Multimerización de Proteína , Receptores de Interleucina-8B/metabolismo
6.
Front Physiol ; 11: 266, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32327998

RESUMEN

BACKGROUND: Invasive mechanical ventilation (IMV) has become one of the mainstays of therapy in NICUs worldwide, as a result of which premature babies with extremely low birth weight have been able to survive. Although lifesaving, IMV can result in lung inflammation and injury. Surfactant therapy is considered a standard of care in preterm infants with immature lungs. Recently, small molecule inhibitors like siRNAs and miRNAs have been used for therapeutic purposes. Ddit3 (CHOP), Ang2 and miR34a are known to be upregulated in experimental lung injury. We wanted to test whether inhibitors for these molecules (CHOP siRNA, Ang2 siRNA, and miR34a antagomir) if used alone or with a combination with surfactant (Curosurf®) would help in reducing ventilation and hyperoxia-induced injury in an experimental lung injury model. METHODS: Preterm rabbits born by cesarean section were intratracheally instilled with the three small molecule inhibitors with or without Curosurf® prior to IMV and hyperoxia exposure. Prior to testing the inhibitors in rabbits, these small molecule inhibitors were transfected in mouse lung epithelial cells (MLE12 and AECII) and delivered to neonatal mouse pups intranasally as a proof of concept that surfactant (Curosurf®) could be used as an effective vehicle for administration of such drugs. Survival, pulmonary function tests, histopathology, immunostaining, quantitative PCR and western blotting were done to see the adjuvant effect of surfactant with these three small molecule inhibitors. RESULTS: Our data shows that Curosurf® can facilitate transfection of small molecules in MLE12 cells with the same and/or increased efficiency as Lipofectamine. Surfactant given alone or as an adjuvant with small molecule inhibitors increases survival, decreases IMV and hyperoxia-induced inflammation, improves pulmonary function and lung injury scores in preterm rabbit kits. CONCLUSION: Our study shows that Curosurf® can be used successfully as an adjuvant therapy with small molecule inhibitors for CHOP/Ang2/miR34a. In this study, of the three inhibitors used, miR34a inhibitor seemed to be the most promising compound to combat IMV and hyperoxia-induced lung injury in preterm rabbits.

7.
PLoS One ; 11(1): e0147588, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26799210

RESUMEN

BACKGROUND: The role of vascular endothelial growth factor (VEGF)-induced 3 different nitric oxide synthase (NOS) isoforms in lung development and injury in the newborn (NB) lung are not known. We hypothesized that VEGF-induced specific NOS pathways are critical regulators of lung development and injury. METHODOLOGY: We studied NB wild type (WT), lung epithelial cell-targeted VEGF165 doxycycline-inducible overexpressing transgenic (VEGFTG), VEGFTG treated with a NOS1 inhibitor (L-NIO), VEGFTG x NOS2-/- and VEGFTG x NOS3+/- mice in room air (RA) for 7 postnatal (PN) days. Lung morphometry (chord length), vascular markers (Ang1, Ang2, Notch2, vWF, CD31 and VE-cadherin), cell proliferation (Ki67), vascular permeability, injury and oxidative stress markers (hemosiderin, nitrotyrosine and 8-OHdG) were evaluated. RESULTS: VEGF overexpression in RA led to increased chord length and vascular markers at PN7, which were significantly decreased to control values in VEGFTG x NOS2-/- and VEGFTG x NOS3+/- lungs. However, we found no noticeable effect on chord length and vascular markers in the VEGFTG / NOS1 inhibited group. In the NB VEGFTG mouse model, we found VEGF-induced vascular permeability in the NB murine lung was partially dependent on NOS2 and NOS3-signaling pathways. In addition, the inhibition of NOS2 and NOS3 resulted in a significant decrease in VEGF-induced hemosiderin, nitrotyrosine- and 8-OHdG positive cells at PN7. NOS1 inhibition had no significant effect. CONCLUSION: Our data showed that the complete absence of NOS2 and partial deficiency of NOS3 confers protection against VEGF-induced pathologic lung vascular and alveolar developmental changes, as well as injury markers. Inhibition of NOS1 does not have any modulating role on VEGF-induced changes in the NB lung. Overall, our data suggests that there is a significant differential regulation in the NOS-mediated effects of VEGF overexpression in the developing mouse lung.


Asunto(s)
Vasos Sanguíneos/crecimiento & desarrollo , Lesión Pulmonar/etiología , Pulmón/crecimiento & desarrollo , Óxido Nítrico Sintasa/metabolismo , Factor A de Crecimiento Endotelial Vascular/fisiología , Animales , Animales Recién Nacidos/crecimiento & desarrollo , Vasos Sanguíneos/metabolismo , Femenino , Pulmón/metabolismo , Lesión Pulmonar/metabolismo , Masculino , Ratones , Ratones Noqueados , Óxido Nítrico Sintasa/fisiología , Óxido Nítrico Sintasa de Tipo I/metabolismo , Óxido Nítrico Sintasa de Tipo I/fisiología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo II/fisiología , Isoformas de Proteínas/metabolismo
8.
Ann Thorac Surg ; 98(5): 1699-704, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25258157

RESUMEN

BACKGROUND: Cardiopulmonary bypass subjects patients' blood to hemodilution and nonphysiologic conditions, resulting in a systemic inflammatory response. Modified ultrafiltration (MUF) counteracts hemodilution and has also been postulated to improve outcomes by proinflammatory cytokine removal. The objective of this study was to investigate whether the benefits of MUF include the removal of proinflammatory mediators, such as angiopoietin-2 (angpt-2). We hypothesize that some of the clinical benefits of MUF are related to the preferential removal of angpt-2. METHODS: We performed a prospective cohort study in children 18 years old or younger undergoing cardiopulmonary bypass. Serum samples were obtained from each patient preoperatively, after cardiopulmonary bypass, and on intensive care unit admission. A fluid sample from the MUF effluent was also analyzed. Angpt-1, angpt-2, interleukin-8, and interleukin-10 levels were determined by enzyme-linked immunosorbent assay. RESULTS: Thirty-one patients were enrolled. Angpt-1 levels significantly decreased across all time points (p<0.01). Angpt-2 concentrations were significantly elevated at intensive care unit admission when compared with both preoperative and post-cardiopulmonary bypass levels (p<0.01). The angpt-2:1 ratio significantly increased after cardiopulmonary bypass to intensive care unit admission (p<0.01). There was no significant difference between the angpt-2 or angpt-1 percentage of extraction within MUF effluent. Interleukin-8 and interleukin-10 significantly increased from preoperative to intensive care unit admission (both p<0.01). CONCLUSIONS: The results of this study demonstrate that MUF removes both proinflammatory and antiinflammatory mediators equally. This study suggests that the clinical benefits of MUF cannot be attributed to the removal of larger quantities of proinflammatory mediators such as angpt-2 and interleukin-8.


Asunto(s)
Angiopoyetinas/sangre , Procedimientos Quirúrgicos Cardíacos , Cardiopatías Congénitas/sangre , Hemofiltración/métodos , Adolescente , Puente Cardiopulmonar , Niño , Preescolar , Femenino , Cardiopatías Congénitas/terapia , Humanos , Lactante , Inflamación/sangre , Masculino , Estudios Prospectivos , Procedimientos Quirúrgicos Torácicos , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda