Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Pediatr Res ; 79(4): 637-46, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26705118

RESUMO

BACKGROUND: Accumulating evidence suggests that c-kit-positive cells are present in the remodeled pulmonary vasculature bed of patients with pulmonary hypertension (PH). Whether stem cell factor (SCF)/c-kit-regulated pathways potentiate pulmonary vascular remodeling is unknown. Here, we tested the hypothesis that attenuated c-kit signaling would decrease chronic hypoxia-induced pulmonary vascular remodeling by decreasing pulmonary vascular cell mitogenesis. METHODS: Neonatal FVB/NJ mice treated with nonimmune IgG (placebo), or c-kit neutralizing antibody (ACK2) as well as c-kit mutant mice (WBB6F1-Kit(W-v/+)) and their congenic controls, were exposed to normoxia (FiO2 = 0.21) or hypoxia (FiO2 = 0.12) for 2 wk. Following this exposure, right ventricular systolic pressure (RVSP), right ventricular hypertrophy (RVH), pulmonary vascular cell proliferation, and remodeling were evaluated. RESULTS: As compared to chronically hypoxic controls, c-kit mutant mice had decreased RVSP, RVH, pulmonary vascular remodeling, and proliferation. Consistent with these findings, administration of ACK2 to neonatal mice with chronic hypoxia-induced PH decreased RVSP, RVH, pulmonary vascular cell proliferation, and remodeling. This attenuation in PH was accompanied by decreased extracellular signal-regulated protein kinase (ERK) 1/2 activation. CONCLUSION: SCF/c-kit signaling may potentiate chronic hypoxia-induced vascular remodeling by modulating ERK activation. Inhibition of c-kit activity may be a potential strategy to alleviate PH.


Assuntos
Modelos Animais de Doenças , Hipóxia/fisiopatologia , Pulmão/irrigação sanguínea , Transdução de Sinais , Fator de Células-Tronco/metabolismo , Remodelação Vascular , Animais , Animais Recém-Nascidos , Anticorpos Neutralizantes/imunologia , Doença Crônica , Hipóxia/metabolismo , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Mutantes , Fator de Células-Tronco/imunologia
2.
Am J Respir Cell Mol Biol ; 53(6): 793-801, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25909334

RESUMO

Lung inflammation plays a key role in the pathogenesis of bronchopulmonary dysplasia (BPD), a chronic lung disease of premature infants. The challenge in BPD management is the lack of effective and safe antiinflammatory agents. Leukadherin-1 (LA1) is a novel agonist of the leukocyte surface integrin CD11b/CD18 that enhances leukocyte adhesion to ligands and vascular endothelium and thus reduces leukocyte transendothelial migration and influx to the injury sites. Its functional significance in preventing hyperoxia-induced neonatal lung injury is unknown. We tested the hypothesis that administration of LA1 is beneficial in preventing hyperoxia-induced neonatal lung injury, an experimental model of BPD. Newborn rats were exposed to normoxia (21% O2) or hyperoxia (85% O2) and received twice-daily intraperitoneal injection of LA1 or placebo for 14 days. Hyperoxia exposure in the presence of the placebo resulted in a drastic increase in the influx of neutrophils and macrophages into the alveolar airspaces. This increased leukocyte influx was accompanied by decreased alveolarization and angiogenesis and increased pulmonary vascular remodeling and pulmonary hypertension (PH), the pathological hallmarks of BPD. However, administration of LA1 decreased macrophage infiltration in the lungs during hyperoxia. Furthermore, treatment with LA1 improved alveolarization and angiogenesis and decreased pulmonary vascular remodeling and PH. These data indicate that leukocyte recruitment plays an important role in the experimental model of BPD induced by hyperoxia. Targeting leukocyte trafficking using LA1, an integrin agonist, is beneficial in preventing lung inflammation and protecting alveolar and vascular structures during hyperoxia. Thus, targeting integrin-mediated leukocyte recruitment and inflammation may provide a novel strategy in preventing and treating BPD in preterm infants.


Assuntos
Anti-Inflamatórios/farmacologia , Benzoatos/farmacologia , Displasia Broncopulmonar/prevenção & controle , Hiperóxia/tratamento farmacológico , Tioidantoínas/farmacologia , Animais , Animais Recém-Nascidos , Anti-Inflamatórios/uso terapêutico , Benzoatos/uso terapêutico , Displasia Broncopulmonar/etiologia , Avaliação Pré-Clínica de Medicamentos , Hiperóxia/complicações , Hipertensão Pulmonar/tratamento farmacológico , Pulmão/irrigação sanguínea , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Infiltração de Neutrófilos , Ratos Sprague-Dawley , Tioidantoínas/uso terapêutico , Resultado do Tratamento , Remodelação Vascular
3.
Neonatology ; 107(4): 304-11, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25825119

RESUMO

BACKGROUND: Lung inflammation is a key factor in the pathogenesis of bronchopulmonary dysplasia (BPD). Stromal-derived factor-1 (SDF-1) and its receptor chemokine receptor 4 (CXCR4) modulate the inflammatory response. It is not known if antagonism of CXCR4 alleviates lung inflammation in neonatal hyperoxia-induced lung injury. OBJECTIVE: We aimed to determine whether CXCR4 antagonism would attenuate lung injury in rodents with experimental BPD by decreasing pulmonary inflammation. METHODS: Newborn rats exposed to normoxia (room air, RA) or hyperoxia (FiO2 = 0.9) from postnatal day 2 (P2) to P16 were randomized to receive the CXCR4 antagonist, AMD3100 or placebo (PL) from P5 to P15. Lung alveolarization, angiogenesis and inflammation were evaluated at P16. RESULTS: Compared to the RA pups, hyperoxic PL pups had a decrease in alveolarization, reduced lung vascular density and increased lung inflammation. In contrast, AMD3100-treated hyperoxic pups had improved alveolarization and increased angiogenesis. This improvement in lung structure was accompanied by a decrease in the macrophage and neutrophil counts in the bronchoalveolar lavage fluid and reduced lung myeloperoxidase activity. CONCLUSION: CXCR4 antagonism decreases lung inflammation and improves alveolar and vascular structure in neonatal rats with experimental BPD. These findings suggest a novel therapeutic strategy to alleviate lung injury in preterm infants with BPD.


Assuntos
Displasia Broncopulmonar/tratamento farmacológico , Compostos Heterocíclicos/uso terapêutico , Hiperóxia/complicações , Hipertensão Pulmonar/tratamento farmacológico , Alvéolos Pulmonares/patologia , Receptores CXCR4/antagonistas & inibidores , Animais , Animais Recém-Nascidos , Benzilaminas , Ciclamos , Modelos Animais de Doenças , Inflamação , Ratos , Ratos Sprague-Dawley
4.
Cell Transplant ; 24(1): 85-95, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-23759597

RESUMO

Recent studies suggest that bone marrow (BM)-derived stem cells have therapeutic efficacy in neonatal hyperoxia-induced lung injury (HILI). c-kit, a tyrosine kinase receptor that regulates angiogenesis, is expressed on several populations of BM-derived cells. Preterm infants exposed to hyperoxia have decreased lung angiogenesis. Here we tested the hypothesis that administration of BM-derived c-kit(+) cells would improve angiogenesis in neonatal rats with HILI. To determine whether intratracheal (IT) administration of BM-derived c-kit(+) cells attenuates neonatal HILI, rat pups exposed to either normobaric normoxia (21% O2) or hyperoxia (90% O2) from postnatal day (P) 2 to P15 were randomly assigned to receive either IT BM-derived green fluorescent protein (GFP)(+) c-kit(-) cells (PL) or BM-derived GFP(+) c-kit(+) cells on P8. The effect of cell therapy on lung angiogenesis, alveolarization, pulmonary hypertension, vascular remodeling, cell proliferation, and apoptosis was determined at P15. Cell engraftment was determined by GFP immunostaining. Compared to PL, the IT administration of BM-derived c-kit(+) cells to neonatal rodents with HILI improved alveolarization as evidenced by increased lung septation and decreased mean linear intercept. This was accompanied by an increase in lung vascular density, a decrease in lung apoptosis, and an increase in the secretion of proangiogenic factors. There was no difference in pulmonary vascular remodeling or the degree of pulmonary hypertension. Confocal microscopy demonstrated that 1% of total lung cells were GFP(+) cells. IT administration of BM-derived c-kit(+) cells improves lung alveolarization and angiogenesis in neonatal HILI, and this may be secondary to an improvement in the lung angiogenic milieu.


Assuntos
Células da Medula Óssea/metabolismo , Transplante de Medula Óssea , Hiperóxia/terapia , Lesão Pulmonar/terapia , Alvéolos Pulmonares , Células-Tronco/metabolismo , Aloenxertos , Animais , Células da Medula Óssea/patologia , Humanos , Hiperóxia/complicações , Hiperóxia/metabolismo , Hiperóxia/patologia , Lesão Pulmonar/etiologia , Lesão Pulmonar/metabolismo , Lesão Pulmonar/patologia , Masculino , Neovascularização Fisiológica , Proteínas Proto-Oncogênicas c-kit , Alvéolos Pulmonares/irrigação sanguínea , Alvéolos Pulmonares/crescimento & desenvolvimento , Alvéolos Pulmonares/patologia , Ratos , Ratos Transgênicos , Células-Tronco/patologia
5.
Am J Respir Cell Mol Biol ; 51(1): 104-13, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24484510

RESUMO

Bronchopulmonary dysplasia (BPD) is the most common and serious chronic lung disease of preterm infants. The development of pulmonary hypertension (PH) significantly increases the mortality and morbidity of this disease. ß-Catenin signaling plays an important role in tissue development and remodeling. Aberrant ß-catenin signaling is associated with clinical and experiment models of BPD. To test the hypothesis that inhibition of ß-catenin signaling is beneficial in promoting alveolar and vascular development and preventing PH in experimental BPD, we examined the effects of ICG001, a newly developed pharmacological inhibitor of ß-catenin, in preventing hyperoxia-induced BPD in neonatal rats. Newborn rat pups were randomized at postnatal day (P)2 to room air (RA) + DMSO (placebo), RA + ICG001, 90% FiO2 (O2) + DMSO, or O2 + ICG001. ICG001 (10 mg/kg) or DMSO was given by daily intraperitoneal injection for 14 days during continuous exposure to RA or hyperoxia. Primary human pulmonary arterial smooth muscle cells (PASMCs) were cultured in RA or hyperoxia (95% O2) in the presence of DMSO or ICG001 for 24 to 72 hours. Treatment with ICG001 significantly increased alveolarization and reduced pulmonary vascular remodeling and PH during hyperoxia. Furthermore, administering ICG001 decreased PASMC proliferation and expression of extracellular matrix remodeling molecules in vitro under hyperoxia. Finally, these structural, cellular, and molecular effects of ICG001 were associated with down-regulation of multiple ß-catenin target genes. These data indicate that ß-catenin signaling mediates hyperoxia-induced alveolar impairment and PH in neonatal animals. Targeting ß-catenin may provide a novel strategy to alleviate BPD in preterm infants.


Assuntos
Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Displasia Broncopulmonar/prevenção & controle , Modelos Animais de Doenças , Hiperóxia/prevenção & controle , Hipertensão Pulmonar/prevenção & controle , Alvéolos Pulmonares/efeitos dos fármacos , Pirimidinonas/farmacologia , beta Catenina/antagonistas & inibidores , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Western Blotting , Displasia Broncopulmonar/metabolismo , Displasia Broncopulmonar/patologia , Proliferação de Células/efeitos dos fármacos , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Imunofluorescência , Humanos , Hiperóxia/metabolismo , Hiperóxia/patologia , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/patologia , Técnicas Imunoenzimáticas , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/patologia , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , beta Catenina/genética , beta Catenina/metabolismo
6.
Pediatr Res ; 74(6): 682-8, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24153399

RESUMO

BACKGROUND: Stem cell factor (SCF) and its receptor, c-kit, are modulators of angiogenesis. Neonatal hyperoxia-induced lung injury (HILI) is characterized by disordered angiogenesis. The objective of this study was to determine whether exogenous SCF improves recovery from neonatal HILI by improving angiogenesis. METHODS: Newborn rats assigned to normoxia (RA: 20.9% O2) or hyperoxia (90% O2) from postnatal day (P) 2 to 15, received daily injections of SCF 100 µg/kg or placebo (PL) from P15 to P21. Lung morphometry was performed at P28. Capillary tube formation in SCF-treated hyperoxia-exposed pulmonary microvascular endothelial cells (HPMECs) was determined by Matrigel assay. RESULTS: As compared with RA, hyperoxic-PL pups had decrease in alveolarization and in lung vascular density, and this was associated with increased right ventricular systolic pressure (RVSP), right ventricular hypertrophy, and vascular remodeling. In contrast, SCF-treated hyperoxic pups had increased angiogenesis, improved alveolarization, and attenuation of pulmonary hypertension as evidenced by decreased RVSP, right ventricular hypertrophy, and vascular remodeling. Moreover, in an in vitro model, SCF increased capillary tube formation in hyperoxia-exposed HPMECs. CONCLUSION: Exogenous SCF restores alveolar and vascular structure in neonatal rats with HILI by promoting neoangiogenesis. These findings suggest a new strategy to treat lung diseases characterized by dysangiogenesis.


Assuntos
Hiperóxia/tratamento farmacológico , Lesão Pulmonar/tratamento farmacológico , Fator de Células-Tronco/uso terapêutico , Animais , Animais Recém-Nascidos , Hiperóxia/fisiopatologia , Lesão Pulmonar/fisiopatologia , Neovascularização Fisiológica/efeitos dos fármacos , Placebos , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/crescimento & desenvolvimento , Ratos , Ratos Sprague-Dawley , Fator de Células-Tronco/farmacologia
7.
Am J Respir Cell Mol Biol ; 48(5): 578-88, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23328640

RESUMO

The pathological hallmarks of bronchopulmonary dysplasia (BPD), a chronic lung disease of premature infants, include inflammation, arrested alveolarization, and dysregulated angiogenesis. Severe BPD is often complicated by pulmonary hypertension (PH) that significantly increases morbidity and mortality. Glycogen synthase kinase (GSK)-3ß plays a pivotal role in embryonic development, cell proliferation and survival, and inflammation by modulating multiple signaling pathways, particularly the nuclear transcription factor, NF-κB, and Wnt/ß-catenin pathways. Aberrant GSK-3ß signaling is linked to BPD. We tested the hypothesis that inhibition of GSK-3ß is beneficial in preventing hyperoxia-induced neonatal lung injury, an experimental model of BPD. Newborn rats were exposed to normoxia or hyperoxia (90% oxygen), and received daily intraperitoneal injections of placebo (DMSO) or SB216763, a specific pharmacological inhibitor of GSK-3ß, for 14 days. Hyperoxia exposure in the presence of the placebo increased GSK-3ß phosphorylation, which was correlated with increased inflammation, decreased alveolarization and angiogenesis, and increased pulmonary vascular remodeling and PH. However, treatment with SB216763 decreased phosphorylation of NF-κB p65, expression of monocyte chemotactic protein-1, and lung inflammation during hyperoxia. Furthermore, treatment with the GSK-3ß inhibitor also improved alveolarization and angiogenesis, and decreased pulmonary vascular remodeling and PH. These data indicate that GSK-3ß signaling plays an important role in the pathogenesis of hyperoxia-induced neonatal lung injury, and that inhibition of GSK-3ß is beneficial in preventing inflammation and protecting alveolar and vascular structures during hyperoxia. Thus, targeting GSK-3ß signaling may offer a novel strategy to prevent and treat preterm infants with BPD.


Assuntos
Displasia Broncopulmonar/tratamento farmacológico , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Hiperóxia/tratamento farmacológico , Indóis/administração & dosagem , Maleimidas/administração & dosagem , Inibidores de Proteínas Quinases/administração & dosagem , Remodelação das Vias Aéreas/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Displasia Broncopulmonar/enzimologia , Displasia Broncopulmonar/etiologia , Avaliação Pré-Clínica de Medicamentos , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , Hiperóxia/complicações , Hiperóxia/enzimologia , Hipertensão Pulmonar/enzimologia , Hipertensão Pulmonar/etiologia , Hipertensão Pulmonar/prevenção & controle , Recém-Nascido , Injeções Intraperitoneais , Pulmão/irrigação sanguínea , Pulmão/efeitos dos fármacos , Pulmão/patologia , Fosforilação , Pneumonia/tratamento farmacológico , Pneumonia/enzimologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Fator de Transcrição RelA/metabolismo
8.
Pediatr Res ; 73(1): 46-53, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23138401

RESUMO

BACKGROUND: Mesenchymal stem cell (MSC) therapy may prevent neonatal hyperoxia-induced lung injury (HILI). There are, however, no clear data on the therapeutic efficacy of MSC therapy in established HILI, the duration of the reparative effects, and the exact mechanisms of repair. The main objective of this study was to evaluate whether the long-term reparative effects of a single intratracheal (IT) dose of MSCs or MSC-conditioned medium (CM) are comparable in established HILI. METHODS: Newborn rats exposed to normoxia or hyperoxia from postnatal day (P)2)-P16 were randomized to receive IT MSCs, IT CM, or IT placebo (PL) on P9. Alveolarization and angiogenesis were evaluated at P16, P30, and P100. RESULTS: At all time periods, there were marked improvements in alveolar and vascular development in hyperoxic pups treated with MSCs or CM as compared with PL. This was associated with decreased expression of inflammatory mediators and an upregulation of angiogenic factors. Of note, at P100, the improvements were more substantial with MSCs as compared with CM. CONCLUSION: These data suggest that acute effects of MSC therapy in HILI are mainly paracrine mediated; however, optimum long-term improvement following HILI requires treatment with the MSCs themselves or potentially repetitive administration of CM.


Assuntos
Meios de Cultivo Condicionados/farmacologia , Hiperóxia/complicações , Lesão Pulmonar/tratamento farmacológico , Lesão Pulmonar/terapia , Transplante de Células-Tronco Mesenquimais/métodos , Análise de Variância , Animais , Animais Recém-Nascidos , Feminino , Hipertrofia Ventricular Direita/patologia , Lesão Pulmonar/etiologia , Masculino , Neovascularização Fisiológica/efeitos dos fármacos , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/crescimento & desenvolvimento , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real
9.
Pediatr Res ; 71(6): 682-8, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22337226

RESUMO

INTRODUCTION: Chemokines may directly participate in the pathogenesis of neonatal chronic hypoxia-induced pulmonary hypertension (PH). Although stromal-derived factor-1 (SDF-1) has been shown to be involved in PH, the role of its most recently discovered receptor, chemokine receptor type 7 (CXCR7), remains unclear. We sought to determine whether antagonism of the CXCR7 receptor would decrease pulmonary vascular remodeling in newborn mice exposed to chronic hypoxia by decreasing pulmonary vascular cell proliferation. METHODS: Neonatal mice were exposed to hypoxia (fractional inspired oxygen concentration = 0.12) or room air (RA) for 2 wk. After 1 wk of exposure, mice received daily injections of placebo or a CXCR7 antagonist (CCX771) from postnatal day 7 (P7) to P14. Right ventricular systolic pressure (RVSP), the ratio of the weight of the right ventricle to left ventricle + septum (RV/LV + S), and pulmonary vascular cell proliferation and remodeling were determined at P14. RESULTS: As compared with mice exposed to RA, hypoxia placebo mice had a significant increase in the lung protein expression of CXCR7. Although hypoxic placebo-treated mice had a significant increase in RVSP, RV/LV+S, and pulmonary vascular cell proliferation and remodeling, the administration of CCX771 markedly decreased these changes. DISCUSSION: These results indicate that antagonism of CXCR7 may be a potent strategy to decrease PH and vascular remodeling.


Assuntos
Hipertensão Pulmonar/etiologia , Hipertensão Pulmonar/prevenção & controle , Hipóxia/complicações , Receptores CXCR/antagonistas & inibidores , Animais , Animais Recém-Nascidos , Artérias/efeitos dos fármacos , Artérias/patologia , Adesão Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Feminino , Hipertensão Pulmonar/patologia , Pulmão/irrigação sanguínea , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos , Modelos Animais , Gravidez , Receptores CXCR/efeitos dos fármacos , Receptores CXCR/metabolismo
10.
Am J Respir Cell Mol Biol ; 45(6): 1169-77, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21659659

RESUMO

Despite recent advances in neonatal intensive care and surfactant therapy, bronchopulmonary dysplasia (BPD) continues to be one of the most common long-term pulmonary complications associated with preterm birth. Clinical efforts to prevent and treat BPD have been largely unsuccessful due to its multifactorial nature and poorly understood disease process. Connective tissue growth factor (CTGF) is a matricellular protein that plays an important role in tissue development and remodeling. Previous studies have demonstrated that hyperoxia exposure up-regulates CTGF expression in neonatal rat lungs. Whether CTGF overexpression plays a role in the pathogenesis of BPD, and whether CTGF antagonism has a therapeutic potential for BPD, are unknown. In the present study, we examined CTGF expression in lung autopsy specimens from patients with BPD and control subjects with no BPD. We assessed the effect of a CTGF-neutralizing monoclonal antibody (CTGF Ab) on preventing hyperoxia-induced lung injury in neonatal rats. Our study demonstrates that CTGF expression is increased in BPD lungs. In newborn rats, exposure to 90% oxygen for 14 days resulted in activation of ß-catenin signaling, decreased alveolarization and vascular development, and physiological and histological evidence of pulmonary hypertension (PH). However, treatment with CTGF Ab prevented ß-catenin signaling activation, improved alveolarization and vascular development, and attenuated PH during hyperoxia. These data indicate that CTGF-ß-catenin signaling plays a critical role in the pathogenesis of experimental BPD. CTGF antagonism may offer a novel therapeutic strategy to alleviate BPD and PH in neonates.


Assuntos
Anticorpos Monoclonais Murinos/farmacologia , Anticorpos Neutralizantes/farmacologia , Displasia Broncopulmonar/tratamento farmacológico , Fator de Crescimento do Tecido Conjuntivo/antagonistas & inibidores , Hiperóxia/tratamento farmacológico , Animais , Animais Recém-Nascidos , Displasia Broncopulmonar/metabolismo , Displasia Broncopulmonar/patologia , Fator de Crescimento do Tecido Conjuntivo/biossíntese , Humanos , Hiperóxia/metabolismo , Hiperóxia/patologia , Hipertensão Pulmonar/tratamento farmacológico , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/patologia , Recém-Nascido , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , beta Catenina/metabolismo
11.
Am J Physiol Lung Cell Mol Physiol ; 300(3): L330-40, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21239535

RESUMO

The pathological hallmarks of bronchopulmonary dysplasia (BPD), one of the most common long-term pulmonary complications associated with preterm birth, include arrested alveolarization, abnormal vascular growth, and variable interstitial fibrosis. Severe BPD is often complicated by pulmonary hypertension characterized by excessive pulmonary vascular remodeling and right ventricular hypertrophy that significantly contributes to the mortality and morbidity of these infants. Connective tissue growth factor (CTGF) is a multifunctional protein that coordinates complex biological processes during tissue development and remodeling. We have previously shown that conditional overexpression of CTGF in airway epithelium under the control of the Clara cell secretory protein promoter results in BPD-like architecture in neonatal mice. In this study, we have generated a doxycycline-inducible double transgenic mouse model with overexpression of CTGF in alveolar type II epithelial (AT II) cells under the control of the surfactant protein C promoter. Overexpression of CTGF in neonatal mice caused dramatic macrophage and neutrophil infiltration in alveolar air spaces and perivascular regions. Overexpression of CTGF also significantly decreased alveolarization and vascular development. Furthermore, overexpression of CTGF induced pulmonary vascular remodeling and pulmonary hypertension. Most importantly, we have also demonstrated that these pathological changes are associated with activation of integrin-linked kinase (ILK)/glucose synthesis kinase-3ß (GSK-3ß)/ß-catenin signaling. These data indicate that overexpression of CTGF in AT II cells results in lung pathology similar to those observed in infants with severe BPD and that ILK/GSK-3ß/ß-catenin signaling may play an important role in the pathogenesis of severe BPD.


Assuntos
Fator de Crescimento do Tecido Conjuntivo/metabolismo , Hipertensão Pulmonar/complicações , Lesão Pulmonar/complicações , Lesão Pulmonar/etiologia , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/patologia , Animais , Animais Recém-Nascidos , Núcleo Celular/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Hipertensão Pulmonar/patologia , Pulmão/irrigação sanguínea , Pulmão/enzimologia , Pulmão/patologia , Camundongos , Modelos Biológicos , Fosforilação , Pneumonia/complicações , Pneumonia/metabolismo , Pneumonia/patologia , beta Catenina/metabolismo
12.
Neonatology ; 99(1): 57-64, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-20639678

RESUMO

BACKGROUND: A single dose of NONOate attenuates pulmonary hypertension (PH) induced by group B Streptococcus (GBS) infusion and this is accompanied by a decrease in systemic vascular resistance (SVR). OBJECTIVE: The objective of the study was to determine whether two doses of the NONOate sustain the attenuation in GBS-induced PH without further systemic compromise. METHODS: 15 anesthetized newborn piglets were randomized to receive placebo (n = 8) or two doses of nebulized DPTA/NO (n = 7) at 15 and 75 min after GBS-induced PH. Pulmonary artery (Ppa) and systemic (Psa) pressures, cardiac output (CO) and arterial blood gases were obtained at baseline and every 15 min until 180 min during GBS infusion. RESULTS: Ppa and pulmonary vascular resistance (PVR) decreased significantly after the first dose of nebulized DPTA/NO and this effect was maintained after the second dose. Psa and SVR decreased after the first dose of DPTA/NO to values close to baseline and no further changes in systemic circulation were observed with repeated treatment. PVR/SVR increased with GBS infusion, but decreased after the first dose of DPTA/NO and remained significantly lower for 180 min. CO was significantly higher in the DPTA/NO group. Changes in Ppa, PVR, Psa, SVR, and CO with GBS infusion were not modified by placebo infusion. PaCO(2), base deficit, and pH did not differ between groups. PaO(2) was significantly lower in the DPTA/NO group after the second dose. CONCLUSION: These data demonstrated that GBS-induced PH is attenuated with two doses of DPTA/NO without significant systemic effect. The vasodilatory effect is more pronounced in the pulmonary than in the systemic vasculature, as suggested by lower PVR/SVR in the DPTA/NO group. We speculate that NONOates may have a clinical application in the management of PH in neonates.


Assuntos
Alcenos/administração & dosagem , Hipertensão Pulmonar/tratamento farmacológico , Doadores de Óxido Nítrico/administração & dosagem , Óxido Nítrico/administração & dosagem , Infecções Estreptocócicas/tratamento farmacológico , Streptococcus agalactiae , Administração por Inalação , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Esquema de Medicação , Hipertensão Pulmonar/etiologia , Nebulizadores e Vaporizadores , Infecções Estreptocócicas/complicações , Suínos , Resultado do Tratamento
13.
Exp Lung Res ; 36(2): 111-9, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20205596

RESUMO

Current data suggest that Toll-like receptor 4 (TLR4), a key molecule in the innate immune response, may also be activated following tissue injury. Activation of this receptor is known to induce the production of several proinflammatory cytokines. Given that pulmonary inflammation has been shown to be a key contributor to chronic hypoxia-induced pulmonary vascular remodeling, the authors hypothesized that TLR4-deficient mice would be less susceptible to pulmonary hypertension (PH) as compared to mice with intact TLR4. TLR4-deficient and TLR4-intact strains of inbred mice were exposed to 4, 8, and 16 weeks of hypoxia (0.10 FiO(2)) or normoxia (0.21 FiO(2)) in a normobaric chamber. After chronic hypoxic exposure, TLR4-intact mice developed significant PH evidenced by increased right ventricular systolic pressure, right ventricular hypertrophy, and pulmonary artery medial thickening. In contrast, TLR4-deficient mice had no significant change in any of these parameters and this was associated with decreased pulmonary vascular inflammatory response as compared to the TLR4-intact mice. These results suggest that TLR4 deficiency may decrease the susceptibility to developing PH by attenuating the pulmonary vascular inflammatory response to chronic hypoxia.


Assuntos
Hipertensão Pulmonar/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Hipóxia/complicações , Metaloproteinase 9 da Matriz/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Pressão Sanguínea , Hipertensão Pulmonar/etiologia , Hipertensão Pulmonar/fisiopatologia , Hipertrofia Ventricular Direita , Hipóxia/fisiopatologia , Inflamação/metabolismo , Pulmão/irrigação sanguínea , Pulmão/fisiopatologia , Camundongos , Camundongos Endogâmicos C3H , Receptor 4 Toll-Like/genética
14.
Circ Res ; 104(11): 1293-301, 2009 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-19423843

RESUMO

Exposure of the neonatal lung to chronic hypoxia produces significant pulmonary vascular remodeling, right ventricular hypertrophy, and decreased lung alveolarization. Given recent data suggesting that stem cells could contribute to pulmonary vascular remodeling and right ventricular hypertrophy, we tested the hypothesis that blockade of SDF-1 (stromal cell-derived factor 1), a key stem cell mobilizer or its receptor, CXCR4 (CXC chemokine receptor 4), would attenuate and reverse hypoxia-induced cardiopulmonary remodeling in newborn mice. Neonatal mice exposed to normoxia or hypoxia were randomly assigned to receive daily intraperitoneal injections of normal saline, AMD3100, or anti-SDF-1 antibody from postnatal day 1 to 7 (preventive strategy) or postnatal day 7 to 14 (therapeutic strategy). As compared to normal saline, inhibition of the SDF-1/CXCR4 axis significantly improved lung alveolarization and decreased pulmonary hypertension, right ventricular hypertrophy, vascular remodeling, vascular cell proliferation, and lung or right ventricular stem cell expressions to near baseline values. We therefore conclude that the SDF-1/CXCR4 axis both prevents and reverses hypoxia-induced cardiopulmonary remodeling in neonatal mice, by decreasing progenitor cell recruitment to the pulmonary vasculature, as well as by decreasing pulmonary vascular cell proliferation. These data offer novel insights into the role of the SDF-1/CXCR4 axis in the pathogenesis of neonatal hypoxia-induced cardiopulmonary remodeling and have important therapeutic implications.


Assuntos
Quimiocina CXCL12/antagonistas & inibidores , Compostos Heterocíclicos/uso terapêutico , Hipertensão Pulmonar/prevenção & controle , Hipóxia/complicações , Receptores CXCR4/antagonistas & inibidores , Animais , Animais Recém-Nascidos , Benzilaminas , Células da Medula Óssea/fisiologia , Ciclamos , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/fisiopatologia , Humanos , Hipertensão Pulmonar/tratamento farmacológico , Hipertensão Pulmonar/fisiopatologia , Hipóxia/fisiopatologia , Recém-Nascido , Recém-Nascido Prematuro , Camundongos , Circulação Pulmonar/fisiologia , Sístole/efeitos dos fármacos , Sístole/fisiologia , Remodelação Ventricular/efeitos dos fármacos , Remodelação Ventricular/fisiologia
15.
Neonatology ; 95(2): 132-40, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-18776727

RESUMO

BACKGROUND: Acute episodes of hypoxemia in ventilated preterm infants are triggered by changes in ventilation, lung volume (LV) and respiratory system compliance (C(RS)) that are not prevented by conventional synchronized intermittent mandatory ventilation (SIMV). OBJECTIVE: To assess in a rabbit model of episodic hypoxemia the individual and combined efficacy of targeted tidal volume (V(T)) and minute ventilation (V'(E)) by automatic adjustment of peak inspiratory pressure (PIP) and ventilator rate, respectively. METHODS: Six young New Zealand white rabbits were ventilated with SIMV, targeted V(T), targeted V'(E), and combined targeted V'(E) + V(T) in random sequence. Hypoxemia episodes were induced by apnea alone or by apnea combined with a reduction in LV and C(RS). Apnea was induced by a bolus of propofol. The reduction in LV and C(RS) was induced by chest compression with a cuff. PaO(2) and PaCO(2) were measured continuously by an indwelling arterial electrode. RESULTS: During SIMV, apnea caused a decrease in ventilation and PaO(2). This was attenuated during targeted V'(E) and targeted V'(E) + V(T). Apnea plus a reduction in LV and C(RS) caused a greater decrease in ventilation and PaO(2) during SIMV. These changes were attenuated during targeted V(T) and targeted V'(E). The attenuation was more pronounced during targeted V'(E) + V(T). CONCLUSION: In this animal model, targeted V'(E) was effective in reducing hypoxemia caused by apnea. When apnea was accompanied by a reduction in LV and C(RS), the combined adjustment of PIP and ventilator rate was more effective than each individually. This combined strategy may be effective in ameliorating acute episodes of hypoxemia in preterm infants but this remains to be proven.


Assuntos
Hipóxia/fisiopatologia , Hipóxia/terapia , Ventilação com Pressão Positiva Intermitente/métodos , Ventilação Pulmonar/fisiologia , Mecânica Respiratória/fisiologia , Animais , Apneia/induzido quimicamente , Apneia/complicações , Apneia/fisiopatologia , Calibragem , Modelos Animais de Doenças , Hipóxia/etiologia , Pulmão/fisiopatologia , Complacência Pulmonar/fisiologia , Pressão/efeitos adversos , Propofol/farmacologia , Coelhos , Volume de Ventilação Pulmonar/fisiologia
16.
Neonatology ; 93(4): 263-8, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18043007

RESUMO

BACKGROUND: Angiotensin-converting enzyme activity is increased in newborn infants with respiratory distress syndrome and in animals with alveolar hypoxia. OBJECTIVE: To test whether angiotensin II (Ang II) mediates the pulmonary vasoconstriction induced by acute hypoxia in newborn piglets. METHODS: Eight unanesthetized chronically instrumented newborn piglets (mean +/- SEM; age 6.6 +/- 0.6 days; weight 2,181 +/- 174 g) were randomly assigned to receive a saline solution or the Ang II type 1 receptor (AT(1)) antagonist, losartan, in a crossover study design, with an interval of at least 48 h between the first and second study. Pulmonary artery (Ppa), wedge, systemic arterial (Psa) and right atrial pressures, cardiac output (CO), pulmonary (PVR) and systemic (SVR) vascular resistances, and arterial blood gases were obtained in room air, before and during the saline or losartan infusion (6 mg/kg followed by 3 mg/kg/h), and during 6 h of hypoxia (FiO(2) = 0.11) and saline or losartan infusion. Data were analyzed by repeated measures analysis of variance. RESULTS: The pulmonary vasoconstriction induced by acute hypoxia was significantly attenuated during losartan infusion, while Psa, SVR, CO, pH, PaCO(2), PaO(2) and base excess did not differ between groups. During room air, Ppa, PVR, Psa, SVR and CO values were not modified by saline or losartan infusion. CONCLUSION: These data suggest that the pulmonary vasoconstriction induced by acute hypoxia in newborn piglets is partially mediated by Ang II, acting via AT(1).


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Antagonistas de Receptores de Angiotensina , Hipóxia/fisiopatologia , Losartan/farmacologia , Artéria Pulmonar/fisiopatologia , Receptor Tipo 1 de Angiotensina/fisiologia , Animais , Animais Recém-Nascidos , Débito Cardíaco , Constrição Patológica , Modelos Animais de Doenças , Hemodinâmica/efeitos dos fármacos , Suínos
17.
Pediatr Res ; 63(1): 46-50, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18043517

RESUMO

The ventilatory response to hypoxia is influenced by the balance between inhibitory (GABA, glycine, and taurine) and excitatory (glutamate and aspartate) brainstem amino acid (AA) neurotransmitters. To assess the effects of AA in the nucleus tractus solitarius (NTS) on the ventilatory response to hypoxia at 1 and 2 wk of age, inhibitory and excitatory AA were sampled by microdialysis in unanesthetized and chronically instrumented piglets. Microdialysis samples from the NTS area were collected at 5-min intervals and minute ventilation (VE), arterial blood pressure (ABP), and arterial blood gases (ABG) were measured while the animals were in quiet sleep. A biphasic ventilatory response to hypoxia was observed in wk 1 and 2, but the decrease in VE at 10 and 15 min was more marked in wk 1. This was associated with an increase in inhibitory AA during hypoxia in wk 1. Excitatory AA levels were elevated during hypoxia in wk 1 and 2. Changes in ABP, pH, and ABG during hypoxia were not different between weeks. These data suggest that the larger depression in the ventilatory response to hypoxia observed in younger piglets is mediated by predominance of the inhibitory AA neurotransmitters, GABA, glycine, and taurine, in the NTS.


Assuntos
Aminoácidos/metabolismo , Hipóxia/metabolismo , Neurotransmissores/metabolismo , Ventilação Pulmonar , Centro Respiratório/metabolismo , Núcleo Solitário/metabolismo , Equilíbrio Ácido-Base , Animais , Animais Recém-Nascidos , Ácido Aspártico/metabolismo , Pressão Sanguínea , Dióxido de Carbono/sangue , Modelos Animais de Doenças , Ácido Glutâmico/metabolismo , Glicina/metabolismo , Frequência Cardíaca , Hipóxia/fisiopatologia , Microdiálise , Oxigênio/sangue , Centro Respiratório/fisiopatologia , Mecânica Respiratória , Núcleo Solitário/fisiopatologia , Suínos , Taurina/metabolismo , Volume de Ventilação Pulmonar , Fatores de Tempo , Ácido gama-Aminobutírico/metabolismo
18.
Pulm Pharmacol Ther ; 21(1): 54-60, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-17251044

RESUMO

BACKGROUND: Volume-induced lung injury is associated with lung inflammation. Pentoxifylline inhibits cytokine release and modulates neutrophil function. OBJECTIVE: To evaluate the efficacy of pentoxifylline in the attenuation of lung inflammation induced by high tidal volume ventilation. DESIGN: Adult rats were randomly assigned to receive saline as placebo or pentoxifylline (100mg/kg over 30 min, followed by 50mg/kg/h) before and during 4h of high tidal volume ventilation (20 ml/kg). Bronchoalveolar fluid inflammatory mediators were measured at baseline and after 4h of ventilation. Lung tissue myeloperoxidase activity and wet/dry lung weight were assessed upon completion of the study. RESULTS: Bronchoalveolar tumor necrosis factor-alpha (pentoxifylline vs. placebo; 192+/-61 vs. 543+/-99 pg/ml; p<0.007) and thromboxane B(2) (262+/-26 vs. 418+/-49 pg/ml; p<0.02) concentrations, lung myeloperoxidase activity (0.5+/-0.1 vs. 1.2+/-0.2U/mg; p<0.003) and wet/dry weight (6.1+/-0.2 vs. 7.1+/-0.3; p<0.01) were all significantly lower in the pentoxifylline-treated group. CONCLUSION: Pentoxifylline was effective in reducing inflammatory lung injury associated with high tidal volume ventilation.


Assuntos
Mediadores da Inflamação/metabolismo , Pulmão/efeitos dos fármacos , Pentoxifilina/farmacologia , Inibidores de Fosfodiesterase/farmacologia , Pneumonia/imunologia , Respiração Artificial/efeitos adversos , Animais , Líquido da Lavagem Broncoalveolar/citologia , Feminino , Pulmão/imunologia , Peroxidase/metabolismo , Pneumonia/etiologia , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Tromboxano B2/metabolismo , Volume de Ventilação Pulmonar
19.
Pediatr Res ; 57(3): 378-83, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15585676

RESUMO

NONOates are chemical compounds that are stable as solids but generate nitric oxide (NO) in aqueous solutions. When nebulized or instilled intratracheally, NONOates can attenuate pulmonary hypertension in adult animals with lung injury. To assess the effect of a nebulized NONOate, DPTA/NO, on group B Streptococcus (GBS)-induced pulmonary hypertension in newborn piglets, we studied 20 anesthetized and mechanically ventilated piglets (4-10 d). They were randomly assigned to receive nebulized placebo solution or DPTA/NO (100 mg) 15 min after sustained pulmonary hypertension. Pulmonary artery and wedge, systemic, and right atrial pressures; cardiac output; and arterial blood gases were obtained at baseline and every 15 min during 120 min of continuous GBS infusion (6 x 10(8) CFU/min). Methemoglobin levels were measured at baseline and 60 min. A significant decrease in pulmonary artery pressure, pulmonary vascular resistance (PVR), systemic arterial pressure, and systemic vascular resistance (SVR) was observed after DPTA/NO nebulization (p <0.001). Whereas the increase in PVR/SVR observed after GBS infusion was sustained for 120 min in the placebo group, this ratio decreased after DPTA/NO nebulization and remained significantly lower throughout the study period (p <0.01). Cardiac output, arterial blood gases, and methemoglobin values did not differ between groups. These data demonstrate that the pulmonary hypertension induced by GBS infusion is markedly attenuated by DPTA/NO nebulization. The lower PVR/SVR observed in the treated group indicates that the vasodilatory effect of NONOate is more pronounced in the pulmonary than systemic vasculature. Therefore, NONOates may have clinical application in the management of pulmonary hypertension secondary to sepsis in neonates.


Assuntos
Alcenos/administração & dosagem , Alcenos/uso terapêutico , Hipertensão Pulmonar/tratamento farmacológico , Óxido Nítrico/administração & dosagem , Óxido Nítrico/uso terapêutico , Streptococcus agalactiae , Adulto , Alcenos/farmacologia , Animais , Animais Recém-Nascidos , Hemodinâmica/efeitos dos fármacos , Humanos , Hipertensão Pulmonar/etiologia , Recém-Nascido , Óxido Nítrico/farmacologia , Distribuição Aleatória , Infecções Estreptocócicas/complicações , Infecções Estreptocócicas/tratamento farmacológico , Suínos
20.
Biol Neonate ; 85(3): 195-202, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-14707432

RESUMO

NONOates, novel NO donors, are complexes of NO with nucleophiles which spontaneously and nonenzymatically release NO in aqueous solution. This study sought to determine the cardiopulmonary effects of the nebulized NONOate dipropylenetriamine (DPTA)/NO in newborn piglets with acute hypoxia-induced pulmonary hypertension. Twenty sedated and mechanically ventilated piglets (4-10 days old) exposed to hypoxia (Fi(O2) = 0.14) were randomly assigned to receive nebulized saline as placebo (PL) or DPTA/NO (75 mg) after 30 min of hypoxia. Pulmonary artery (P(pa)) and wedge pressures, systemic (P(sa)) and right atrial pressures, cardiac output (CO) and arterial blood gas were measured at baseline and every 15 min for 2 h. Methemoglobin levels were measured at baseline and 1 h after drug nebulization. Data (means +/- SD) were analyzed by repeated-measures analysis of variance. Acute hypoxia resulted in an increase in P(pa) and pulmonary vascular resistance (PVR), which was significantly attenuated by DPTA/NO nebulization as compared to the PL group (p < 0.0001). Changes in P(sa), CO, systemic vascular resistance (SVR), arterial blood gas and methemoglobin levels were not different between groups. In contrast to the increase in PVR/SVR observed during hypoxia in the PL group, there was a significant decrease in this ratio after NONOate administration (p < 0.0001). These data show that acute hypoxic pulmonary hypertension in newborn piglets is markedly attenuated by NONOate nebulization. This response is predominantly in the pulmonary vasculature as the PVR/SVR was significantly lower in the treated group. We speculate that NONOates may have clinical application in the treatment of persistent pulmonary hypertension of the newborn.


Assuntos
Alcenos/administração & dosagem , Hipertensão Pulmonar/tratamento farmacológico , Hipóxia/fisiopatologia , Doadores de Óxido Nítrico/administração & dosagem , Doença Aguda , Animais , Animais Recém-Nascidos , Gasometria , Pressão Sanguínea/efeitos dos fármacos , Modelos Animais de Doenças , Hipertensão Pulmonar/sangue , Hipertensão Pulmonar/fisiopatologia , Hipóxia/sangue , Nebulizadores e Vaporizadores , Distribuição Aleatória , Testes de Função Respiratória , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...