Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Am J Physiol Lung Cell Mol Physiol ; 305(5): L341-51, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23812633

RESUMO

Stimulation of MAS oncogene receptor (MAS) or angiotensin (Ang) receptor type 2 (AT2) may be novel therapeutic options for neonatal chronic lung disease (CLD) by counterbalancing the adverse effects of the potent vasoconstrictor angiotensin II, consisting of arterial hypertension (PAH)-induced right ventricular hypertrophy (RVH) and pulmonary inflammation. We determined the cardiopulmonary effects in neonatal rats with CLD of daily treatment during continuous exposure to 100% oxygen for 10 days with specific ligands for MAS [cyclic Ang-(1-7); 10-50 µg·kg(-1)·day(-1)] and AT2 [dKcAng-(1-7); 5-20 µg·kg(-1)·day(-1)]. Parameters investigated included lung and heart histopathology, fibrin deposition, vascular leakage, and differential mRNA expression in the lungs of key genes involved in the renin-angiotensin system, inflammation, coagulation, and alveolar development. We investigated the role of nitric oxide synthase inhibition with N(ω)-nitro-l-arginine methyl ester (25 mg·kg(-1)·day(-1)) during AT2 agonist treatment. Prophylactic treatment with agonists for MAS or AT2 for 10 days diminished cardiopulmonary injury by reducing alveolar septum thickness and medial wall thickness of small arterioles and preventing RVH. Both agonists attenuated the pulmonary influx of inflammatory cells, including macrophages (via AT2) and neutrophils (via MAS) but did not reduce alveolar enlargement and vascular alveolar leakage. The AT2 agonist attenuated hyperoxia-induced fibrin deposition. In conclusion, stimulation of MAS or AT2 attenuates cardiopulmonary injury by reducing pulmonary inflammation and preventing PAH-induced RVH but does not affect alveolar and vascular development in neonatal rats with experimental CLD. The beneficial effects of AT2 activation on experimental CLD were mediated via a NOS-independent mechanism.


Assuntos
Angiotensina I/farmacologia , Hiperóxia/complicações , Hipertensão Pulmonar/prevenção & controle , Hipertrofia Ventricular Direita/prevenção & controle , Lesão Pulmonar/etiologia , Fragmentos de Peptídeos/farmacologia , Pneumonia/prevenção & controle , Proteínas Proto-Oncogênicas/agonistas , Receptor Tipo 2 de Angiotensina/agonistas , Receptores Acoplados a Proteínas G/agonistas , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Western Blotting , Lavagem Broncoalveolar , Proliferação de Células/efeitos dos fármacos , Hiperóxia/patologia , Hipertensão Pulmonar/etiologia , Hipertensão Pulmonar/metabolismo , Hipertrofia Ventricular Direita/etiologia , Hipertrofia Ventricular Direita/metabolismo , Lesão Pulmonar/patologia , Masculino , Oxigênio/metabolismo , Pneumonia/etiologia , Pneumonia/metabolismo , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas/metabolismo , RNA Mensageiro/genética , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real , Receptor Tipo 2 de Angiotensina/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Sistema Renina-Angiotensina/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
2.
Am J Physiol Lung Cell Mol Physiol ; 304(4): L264-75, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23292811

RESUMO

Ambrisentan, an endothelin receptor type A antagonist, may be a novel therapeutic agent in neonatal chronic lung disease (CLD) by blocking the adverse effects of the vasoconstrictor endothelin-1, especially pulmonary arterial hypertension (PAH)-induced right ventricular hypertrophy (RVH). We determined the cardiopulmonary effects of ambrisentan treatment (1-20 mg·kg(-1)·day(-1)) in neonatal rats with CLD in 2 models: early treatment during continuous exposure to hyperoxia for 10 days and late treatment starting on day 6 in rat pups exposed postnatally to hyperoxia for 9 days, followed by a 9-day recovery period in room air. Parameters investigated included survival, lung and heart histopathology, right ventricular function, fibrin deposition, and differential mRNA expression in the lungs. In the early treatment model, we investigated the role of nitric oxide synthase (NOS) inhibition with N(ω)-nitro-L-arginine methyl ester (L-NAME; 25 mg·kg(-1)·day(-1)) during ambrisentan treatment. In the early treatment model, ambrisentan improved survival with reduced lung fibrin and collagen III deposition, arterial medial wall thickness, and RVH. These changes were not affected by L-NAME administration. Ambrisentan did not reduce the influx of macrophages and neutrophils or prevent reduced irregular elastin expression. In the late treatment model, ambrisentan diminished PAH, RVH, and right ventricular peak pressure, demonstrating that RVH is reversible in the neonatal period. Alveolarization and vascularization were not affected by ambrisentan. In conclusion, ambrisentan prolongs survival and reduces lung injury, PAH, and RVH via a NOS-independent mechanism but does not affect inflammation and alveolar and vascular development in neonatal rats with CLD.


Assuntos
Hiperóxia/fisiopatologia , Hipertensão Pulmonar/tratamento farmacológico , Fenilpropionatos/uso terapêutico , Alvéolos Pulmonares/efeitos dos fármacos , Piridazinas/uso terapêutico , Animais , Animais Recém-Nascidos/crescimento & desenvolvimento , Hipertensão Pulmonar Primária Familiar , Coração/efeitos dos fármacos , Coração/crescimento & desenvolvimento , Hipertrofia Ventricular Direita/prevenção & controle , Pulmão/efeitos dos fármacos , Pulmão/crescimento & desenvolvimento , NG-Nitroarginina Metil Éster/farmacologia , Fenilpropionatos/administração & dosagem , Alvéolos Pulmonares/crescimento & desenvolvimento , Piridazinas/administração & dosagem , Ratos , Fatores de Tempo
3.
Am J Physiol Lung Cell Mol Physiol ; 302(1): L56-67, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21949154

RESUMO

Phosphodiesterase (PDE) 4 inhibitors are potent anti-inflammatory drugs with antihypertensive properties, and their therapeutic role in bronchopulmonary dysplasia (BPD) is still controversial. We studied the role of PDE4 inhibition with piclamilast on normal lung development and its therapeutic value on pulmonary hypertension (PH) and right ventricular hypertrophy (RVH) in neonatal rats with hyperoxia-induced lung injury, a valuable model for premature infants with severe BPD. The cardiopulmonary effects of piclamilast treatment (5 mg·kg(-1)·day(-1)) were investigated in two models of experimental BPD: 1) daily treatment during continuous exposure to hyperoxia for 10 days; and 2) late treatment and injury-recovery in which pups were exposed to hyperoxia or room air for 9 days, followed by 9 or 42 days of recovery in room air combined with treatment started on day 6 of oxygen exposure until day 18. Prophylactic piclamilast treatment reduced pulmonary fibrin deposition, septum thickness, arteriolar wall thickness, arteriolar vascular smooth muscle cell proliferation and RVH, and prolonged survival. In the late treatment and injury-recovery model, hyperoxia caused persistent aberrant alveolar and vascular development, PH, and RVH. Treatment with piclamilast in both models reduced arteriolar wall thickness, attenuated RVH, and improved right ventricular function in the injury recovery model, but did not restore alveolarization or angiogenesis. Treatment with piclamilast did not show adverse cardiopulmonary effects in room air controls in both models. In conclusion, PDE4 inhibition attenuated and partially reversed PH and RVH, but did not advance alveolar development in neonatal rats with hyperoxic lung injury or affect normal lung and heart development.


Assuntos
Benzamidas , Displasia Broncopulmonar , Traumatismos Cardíacos/tratamento farmacológico , Hiperóxia/tratamento farmacológico , Inibidores da Fosfodiesterase 4 , Piridinas , Animais , Animais Recém-Nascidos , Benzamidas/farmacologia , Benzamidas/uso terapêutico , Displasia Broncopulmonar/tratamento farmacológico , Displasia Broncopulmonar/patologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/metabolismo , Modelos Animais de Doenças , Coração/efeitos dos fármacos , Coração/fisiopatologia , Traumatismos Cardíacos/fisiopatologia , Humanos , Hiperóxia/fisiopatologia , Hipertensão Pulmonar/tratamento farmacológico , Hipertensão Pulmonar/fisiopatologia , Hipertrofia Ventricular Direita/tratamento farmacológico , Hipertrofia Ventricular Direita/fisiopatologia , Recém-Nascido , Pulmão/efeitos dos fármacos , Pulmão/fisiopatologia , Estresse Oxidativo/efeitos dos fármacos , Oxigênio/farmacologia , Inibidores da Fosfodiesterase 4/farmacologia , Inibidores da Fosfodiesterase 4/uso terapêutico , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/crescimento & desenvolvimento , Piridinas/farmacologia , Piridinas/uso terapêutico , Ratos
4.
Am J Physiol Heart Circ Physiol ; 297(5): H1606-16, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19783775

RESUMO

Pulmonary arterial hypertension (PAH) is a chronic lung disease that leads to right ventricular (RV) hypertrophy (RVH), remodeling, and failure. We tested treatment with bone marrow-derived mesenchymal stem cells (MSCs) obtained from donor rats with monocrotaline (MCT)-induced PAH to recipient rats with MCT-induced PAH on pulmonary artery pressure, lung pathology, and RV function. This model was chosen to mimic autologous MSC therapy. On day 1, PAH was induced by MCT (60 mg/kg) in 20 female Wistar rats. On day 14, rats were treated with 10(6) MSCs intravenously (MCT + MSC) or with saline (MCT60). MSCs were obtained from donor rats with PAH at 28 days after MCT. A control group received saline on days 1 and 14. On day 28, the RV function of recipient rats was assessed, followed by isolation of the lungs and heart. RVH was quantified by the weight ratio of the RV/(left ventricle + interventricular septum). MCT induced an increase of RV peak pressure (from 27 + or - 5 to 42 +/- 17 mmHg) and RVH (from 0.25 + or - 0.04 to 0.47 + or - 0.12), depressed the RV ejection fraction (from 56 + or - 11 to 43 + or - 6%), and increased lung weight (from 0.96 + or - 0.15 to 1.66 + or - 0.32 g), including thickening of the arteriolar walls and alveolar septa. MSC treatment attenuated PAH (31 + or - 4 mmHg) and RVH (0.32 + or - 0.07), normalized the RV ejection fraction (52 + or - 5%), reduced lung weight (1.16 + or - 0.24 g), and inhibited the thickening of the arterioles and alveolar septa. We conclude that the application of MSCs from donor rats with PAH reduces RV pressure overload, RV dysfunction, and lung pathology in recipient rats with PAH. These results suggest that autologous MSC therapy may alleviate cardiac and pulmonary symptoms in PAH patients.


Assuntos
Hipertensão Pulmonar/cirurgia , Hipertrofia Ventricular Direita/prevenção & controle , Pulmão/patologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Miocárdio/patologia , Disfunção Ventricular Direita/prevenção & controle , Função Ventricular Direita , Animais , Arteríolas/patologia , Débito Cardíaco , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Feminino , Frequência Cardíaca , Hipertensão Pulmonar/induzido quimicamente , Hipertensão Pulmonar/patologia , Hipertensão Pulmonar/fisiopatologia , Hipertrofia Ventricular Direita/induzido quimicamente , Hipertrofia Ventricular Direita/patologia , Hipertrofia Ventricular Direita/fisiopatologia , Mediadores da Inflamação/metabolismo , Pulmão/irrigação sanguínea , Pulmão/metabolismo , Pulmão/fisiopatologia , Células-Tronco Mesenquimais/metabolismo , Monocrotalina , Contração Miocárdica , Miocárdio/metabolismo , Alvéolos Pulmonares/patologia , Artéria Pulmonar/patologia , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Recuperação de Função Fisiológica , Volume Sistólico , Fatores de Tempo , Transplante Homólogo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Disfunção Ventricular Direita/induzido quimicamente , Disfunção Ventricular Direita/patologia , Disfunção Ventricular Direita/fisiopatologia , Pressão Ventricular , Remodelação Ventricular
5.
Respir Res ; 10: 30, 2009 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-19402887

RESUMO

BACKGROUND: Phosphodiesterase-5 inhibition with sildenafil has been used to treat severe pulmonary hypertension and bronchopulmonary dysplasia (BPD), a chronic lung disease in very preterm infants who were mechanically ventilated for respiratory distress syndrome. METHODS: Sildenafil treatment was investigated in 2 models of experimental BPD: a lethal neonatal model, in which rat pups were continuously exposed to hyperoxia and treated daily with sildenafil (50-150 mg/kg body weight/day; injected subcutaneously) and a neonatal lung injury-recovery model in which rat pups were exposed to hyperoxia for 9 days, followed by 9 days of recovery in room air and started sildenafil treatment on day 6 of hyperoxia exposure. Parameters investigated include survival, histopathology, fibrin deposition, alveolar vascular leakage, right ventricular hypertrophy, and differential mRNA expression in lung and heart tissue. RESULTS: Prophylactic treatment with an optimal dose of sildenafil (2 x 50 mg/kg/day) significantly increased lung cGMP levels, prolonged median survival, reduced fibrin deposition, total protein content in bronchoalveolar lavage fluid, inflammation and septum thickness. Treatment with sildenafil partially corrected the differential mRNA expression of amphiregulin, plasminogen activator inhibitor-1, fibroblast growth factor receptor-4 and vascular endothelial growth factor receptor-2 in the lung and of brain and c-type natriuretic peptides and the natriuretic peptide receptors NPR-A, -B, and -C in the right ventricle. In the lethal and injury-recovery model we demonstrated improved alveolarization and angiogenesis by attenuating mean linear intercept and arteriolar wall thickness and increasing pulmonary blood vessel density, and right ventricular hypertrophy (RVH). CONCLUSION: Sildenafil treatment, started simultaneously with exposure to hyperoxia after birth, prolongs survival, increases pulmonary cGMP levels, reduces the pulmonary inflammatory response, fibrin deposition and RVH, and stimulates alveolarization. Initiation of sildenafil treatment after hyperoxic lung injury and continued during room air recovery improves alveolarization and restores pulmonary angiogenesis and RVH in experimental BPD.


Assuntos
Modelos Animais de Doenças , Fibrina/metabolismo , Hipertrofia Ventricular Direita/tratamento farmacológico , Hipertrofia Ventricular Direita/mortalidade , Piperazinas/administração & dosagem , Pneumonia/tratamento farmacológico , Pneumonia/mortalidade , Sulfonas/administração & dosagem , Animais , Animais Recém-Nascidos , Humanos , Hiperóxia/tratamento farmacológico , Hiperóxia/metabolismo , Hiperóxia/mortalidade , Hipertrofia Ventricular Direita/metabolismo , Lesão Pulmonar/tratamento farmacológico , Lesão Pulmonar/metabolismo , Lesão Pulmonar/mortalidade , Inibidores de Fosfodiesterase/administração & dosagem , Pneumonia/metabolismo , Purinas/administração & dosagem , Ratos , Ratos Wistar , Citrato de Sildenafila , Análise de Sobrevida , Taxa de Sobrevida , Resultado do Tratamento
6.
Br J Pharmacol ; 156(2): 388-96, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19154431

RESUMO

BACKGROUND AND PURPOSE: A combination of early neurodevelopmental insult(s) and young-adult stress exposure may be involved in the development of schizophrenia. We studied prepulse inhibition (PPI) regulation in rats after an early stress, maternal deprivation, combined with a later stress, simulated by chronic corticosterone treatment, and also determined whether changes in brain dopamine receptor density were involved. EXPERIMENTAL APPROACH: Rats were subjected to either 24 h maternal deprivation on postnatal day 9, corticosterone treatment from 8 to 10 weeks of age, or both. At 12 weeks of age, the rats were injected with 0.1, 0.3 or 1.0 mg.kg(-1) of apomorphine or 0.5 or 2.5 mg.kg(-1) of amphetamine and PPI was determined using automated startle boxes. Dopamine D(1) and D(2) receptor levels were assessed in the nucleus accumbens and caudate nucleus using receptor autoradiography. KEY RESULTS: Young-adult treatment with corticosterone resulted in attenuated disruption of PPI by apomorphine and amphetamine. In some rats, maternal deprivation resulted in reduced baseline PPI which added to the effect of corticosterone treatment. There was no down-regulation of dopamine D(1) or D(2) receptors. CONCLUSIONS AND IMPLICATIONS: These results confirm and extend our finding of an inhibitory interaction of developmental stress on dopaminergic regulation of PPI. No corresponding changes in dopamine receptor density were observed in brain regions with a major involvement in PPI regulation, suggesting long-lasting desensitization of dopamine receptor signalling or indirect changes in PPI regulation.


Assuntos
Núcleo Caudado/metabolismo , Corticosterona/farmacologia , Núcleo Accumbens/metabolismo , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , Reflexo de Sobressalto , Estresse Psicológico/metabolismo , Anfetamina/farmacologia , Animais , Animais Recém-Nascidos , Apomorfina/farmacologia , Autorradiografia , Núcleo Caudado/efeitos dos fármacos , Privação Materna , Núcleo Accumbens/efeitos dos fármacos , Ratos , Ratos Wistar , Receptores de Dopamina D1/agonistas , Receptores de Dopamina D2/agonistas , Reflexo de Sobressalto/efeitos dos fármacos , Estresse Psicológico/psicologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA