Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Respir Res ; 24(1): 39, 2023 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-36732726

RESUMO

BACKGROUND: Ibuprofen is a nonsteroidal anti-inflammatory drug that is commonly used to stimulate closure of a patent ductus arteriosus (PDA) in very premature infants and may lead to aberrant neonatal lung development and bronchopulmonary dysplasia (BPD). METHODS: We investigated the effect of ibuprofen on angiogenesis in human umbilical cord vein endothelial cells (HUVECs) and the therapeutic potential of daily treatment with 50 mg/kg of ibuprofen injected subcutaneously in neonatal Wistar rat pups with severe hyperoxia-induced experimental BPD. Parameters investigated included growth, survival, lung histopathology and mRNA expression. RESULTS: Ibuprofen inhibited angiogenesis in HUVECs, as shown by reduced tube formation, migration and cell proliferation via inhibition of the cell cycle S-phase and promotion of apoptosis. Treatment of newborn rat pups with ibuprofen reduced pulmonary vessel density in the developing lung, but also attenuated experimental BPD by reducing lung inflammation, alveolar enlargement, alveolar septum thickness and small arteriolar wall thickening. CONCLUSIONS: In conclusion, ibuprofen has dual effects on lung development: adverse effects on angiogenesis and beneficial effects on alveolarization and inflammation. Therefore, extrapolation of the beneficial effects of ibuprofen to premature infants with BPD should be done with extreme caution.


Assuntos
Displasia Broncopulmonar , Hiperóxia , Recém-Nascido , Animais , Ratos , Humanos , Ibuprofeno/farmacologia , Ibuprofeno/uso terapêutico , Células Endoteliais/metabolismo , Animais Recém-Nascidos , Ratos Wistar , Pulmão , Displasia Broncopulmonar/tratamento farmacológico , Displasia Broncopulmonar/prevenção & controle , Displasia Broncopulmonar/patologia , Hiperóxia/metabolismo
2.
Blood ; 133(19): 2090-2099, 2019 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-30898865

RESUMO

Tissue factor, coagulation factor XII, platelets, and neutrophils are implicated as important players in the pathophysiology of (experimental) venous thrombosis (VT). Their role became evident in mouse models in which surgical handlings were required to provoke VT. Combined inhibition of the natural anticoagulants antithrombin (Serpinc1) and protein C (Proc) using small interfering RNA without additional triggers also results in a venous thrombotic phenotype in mice, most notably with vessel occlusion in large veins of the head. VT is fatal but is fully rescued by thrombin inhibition. In the present study, we used this VT mouse model to investigate the involvement of tissue factor, coagulation factor XII, platelets, and neutrophils. Antibody-mediated inhibition of tissue factor reduced the clinical features of VT, the coagulopathy in the head, and fibrin deposition in the liver. In contrast, genetic deficiency in, and small interfering RNA-mediated depletion of, coagulation factor XII did not alter VT onset, severity, or thrombus morphology. Antibody-mediated depletion of platelets fully abrogated coagulopathy in the head and liver fibrin deposition. Although neutrophils were abundant in thrombotic lesions, depletion of circulating Ly6G-positive neutrophils did not affect onset, severity, thrombus morphology, or liver fibrin deposition. In conclusion, VT after inhibition of antithrombin and protein C is dependent on the presence of tissue factor and platelets but not on coagulation factor XII and circulating neutrophils. This study shows that distinct procoagulant pathways operate in mouse VT, dependent on the triggering stimulus.


Assuntos
Plaquetas/metabolismo , Fator XII/metabolismo , Neutrófilos/metabolismo , Tromboplastina/metabolismo , Trombose Venosa/sangue , Animais , Antitrombina III/antagonistas & inibidores , Modelos Animais de Doenças , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Proteína C/antagonistas & inibidores
3.
Blood ; 127(21): 2630-7, 2016 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-26932804

RESUMO

Recently, platelets, neutrophils, and factor XII (FXII) have been implicated as important players in the pathophysiology of venous thrombosis. Their role became evident in mouse models in which surgical handling was used to provoke thrombosis. Inhibiting anticoagulation in mice by using small interfering RNA (siRNA) targeting Serpinc1 and Proc also results in a thrombotic phenotype, which is spontaneous (no additional triggers) and reproducibly results in clots in the large veins of the head and fibrin deposition in the liver. This thrombotic phenotype is fatal but can be fully rescued by thrombin inhibition. The mouse model was used in this study to investigate the role of platelets, neutrophils, and FXII. After administration of siRNAs targeting Serpinc1 and Proc, antibody-mediated depletion of platelets fully abrogated the clinical features as well as microscopic aspects in the head. This was corroborated by strongly reduced fibrin deposition in the liver. Whereas neutrophils were abundant in siRNA-triggered thrombotic lesions, antibody-mediated depletion of circulating Ly6G-positive neutrophils did not affect onset, severity, or thrombus morphology. In addition, absence of circulating neutrophils did not affect quantitative liver fibrin deposition. Remarkably, siRNA-mediated depletion of plasma FXII accelerated the onset of the clinical phenotype; mice were affected with more severe thrombotic lesions. To summarize, in this study, onset and severity of the thrombotic phenotype are dependent on the presence of platelets but not circulating neutrophils. Unexpectedly, FXII has a protective effect. This study challenges the proposed roles of neutrophils and FXII in venous thrombosis pathophysiology.


Assuntos
Plaquetas/metabolismo , Fator XII/metabolismo , Neutrófilos/metabolismo , Trombose Venosa/metabolismo , Animais , Antígenos Ly/metabolismo , Antitrombina III/antagonistas & inibidores , Antitrombina III/metabolismo , Plaquetas/patologia , Feminino , Fibrina/metabolismo , Fígado/metabolismo , Fígado/patologia , Camundongos , Neutrófilos/patologia , RNA Interferente Pequeno/farmacologia , Trombose Venosa/patologia
4.
Crit Care Med ; 44(12): e1226-e1235, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27513359

RESUMO

OBJECTIVES: A growing body of evidence suggests that age affects the main pathophysiologic mechanisms of the acute respiratory distress syndrome. This may imply the need for developing age-tailored therapies for acute respiratory distress syndrome. However, underlying molecular mechanisms governing age-related susceptibility first need to be unraveled. In a rat model of acute lung injury, we investigated whether age affects the balance between the two key enzymes of the pulmonary renin-angiotensin system, angiotensin-converting enzyme, and angiotensin-converting enzyme 2. We hypothesized that aging shifts the balance toward the lung injury-promoting angiotensin-converting enzyme, which may form an explanation for the differences in severity of lung injury between different age groups. DESIGN: Prospective, randomized controlled animal study. SETTING: University medical research laboratory. SUBJECTS: Infant (15 ± 2 d), juvenile (37 ± 2 d), adult (4 ± 0.2 mo), and elderly (19.5 ± 0.5 mo) male RCCHan Wistar rats. INTERVENTIONS: Lung injury was induced by intratracheal administration of lipopolysaccharide (5 mg/kg) and 4 hours of mechanical ventilation (15 mL/kg). MEASUREMENTS AND MAIN RESULTS: In lipopolysaccharide-exposed and mechanical ventilated rats, angiotensin-converting enzyme activity in bronchoalveolar lavage fluid increased 3.2-fold in elderly when compared with infants. No changes in bronchoalveolar lavage fluid angiotensin-converting enzyme 2 activity were found. In addition, membrane-bound angiotensin-converting enzyme activity decreased. Together with the presence of angiotensin-converting enzyme-sheddase ADAM9 (a disintegrin and metalloproteinase domain-containing protein 9) and an age-dependent increase in tumor necrosis factor-α, an activator of ADAM9, these results indicate increased shedding of angiotensin-converting enzyme in the alveolar compartment, thereby shifting the balance toward the injurious pathway. This imbalance was associated with an increased inflammatory mediator response and more lung injury (wet-to-dry ratio and histology) in elderly rats. CONCLUSIONS: Increasing age is associated with an imbalance of the pulmonary renin-angiotensin system, which correlates with aggravated inflammation and more lung injury. These changes might form the ground for new therapeutic strategies in terms of dosing and effectiveness of renin-angiotensin system-modulating agents for treatment of acute respiratory distress syndrome.


Assuntos
Lesão Pulmonar Aguda/fisiopatologia , Sistema Renina-Angiotensina/fisiologia , Lesão Pulmonar Aguda/etiologia , Fatores Etários , Animais , Pulmão/fisiopatologia , Masculino , Ratos , Ratos Wistar , Síndrome do Desconforto Respiratório/etiologia , Síndrome do Desconforto Respiratório/fisiopatologia
5.
Am J Physiol Lung Cell Mol Physiol ; 309(10): L1037-40, 2015 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-26432873

RESUMO

Supplemental oxygen after premature birth results in aberrant airway, alveolar, and pulmonary vascular development with an increased risk for bronchopulmonary dysplasia, and development of wheeze and asthma, pulmonary hypertension, and chronic obstructive pulmonary disease in survivors. Although stimulation of the nitric oxide (NO)-soluble guanylate cyclase (sGC)-cGMP signal transduction pathway has significant beneficial effects on disease development in animal models, so far this could not be translated to the clinic. Oxidative stress reduces the NO-sGC-cGMP pathway by oxidizing heme-bound sGC, resulting in inactivation or degradation of sGC. Reduced sGC activity and/or expression is associated with pathology due to premature birth, oxidative stress-induced lung injury, including impaired alveolar maturation, smooth muscle cell (SMC) proliferation and contraction, impaired airway relaxation and vasodilation, inflammation, pulmonary hypertension, right ventricular hypertrophy, and an aggravated response toward hyperoxia-induced neonatal lung injury. Recently, Britt et al. (10) demonstrated that histamine-induced Ca(2+) responses were significantly elevated in hyperoxia-exposed fetal human airway SMCs compared with normoxic controls and that this hyperoxia-induced increase in the response was strongly reduced by NO-independent stimulation and activation of sGC. These recent studies highlight the therapeutic potential of sGC modulators in the treatment of preterm infants for respiratory distress with supplemental oxygen. Such treatment is aimed at improving aberrant alveolar and vascular development of the neonatal lung and preventing the development of wheezing and asthma in survivors of premature birth. In addition, these studies highlight the suitability of fetal human airway SMCs as a translational model for pathological airway changes in the neonate.


Assuntos
Ativadores de Enzimas/farmacologia , Guanilato Ciclase/fisiologia , Receptores Citoplasmáticos e Nucleares/fisiologia , Síndrome do Desconforto Respiratório do Recém-Nascido/tratamento farmacológico , Animais , Asma/etiologia , Ativadores de Enzimas/uso terapêutico , Humanos , Terapia de Alvo Molecular , Oxigenoterapia/efeitos adversos , Síndrome do Desconforto Respiratório do Recém-Nascido/enzimologia , Transdução de Sinais , Guanilil Ciclase Solúvel
6.
Am J Physiol Lung Cell Mol Physiol ; 309(3): L262-70, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-26047641

RESUMO

Because therapeutic options are lacking for bronchopulmonary dysplasia (BPD), there is an urgent medical need to discover novel targets/drugs to treat this neonatal chronic lung disease. Metformin, a drug commonly used to lower blood glucose in type 2 diabetes patients, may be a novel therapeutic option for BPD by reducing pulmonary inflammation and fibrosis and improving vascularization. We investigated the therapeutic potential of daily treatment with 25 and 100 mg/kg metformin, injected subcutaneously in neonatal Wistar rats with severe experimental BPD, induced by continuous exposure to 100% oxygen for 10 days. Parameters investigated included survival, lung and heart histopathology, pulmonary fibrin and collagen deposition, vascular leakage, right ventricular hypertrophy, and differential mRNA expression in the lungs of key genes involved in BPD pathogenesis, including inflammation, coagulation, and alveolar development. After daily metformin treatment rat pups with experimental BPD had reduced mortality, alveolar septum thickness, lung inflammation, and fibrosis, demonstrated by a reduced influx of macrophages and neutrophils and hyperoxia-induced collagen III and fibrin deposition (25 mg/kg), as well as improved vascularization (100 mg/kg) compared with control treatment. However, metformin did not ameliorate alveolar enlargement, small arteriole wall thickening, vascular alveolar leakage, and right ventricular hypertrophy. In conclusion metformin prolongs survival and attenuates pulmonary injury by reducing pulmonary inflammation, coagulation, and fibrosis but does not affect alveolar development or prevent pulmonary arterial hypertension and right ventricular hypertrophy in neonatal rats with severe hyperoxia-induced experimental BPD.


Assuntos
Anti-Inflamatórios/farmacologia , Displasia Broncopulmonar/tratamento farmacológico , Metformina/farmacologia , Animais , Animais Recém-Nascidos , Anti-Inflamatórios/uso terapêutico , Displasia Broncopulmonar/complicações , Displasia Broncopulmonar/imunologia , Permeabilidade Capilar/efeitos dos fármacos , Colágeno/metabolismo , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Elastina/metabolismo , Fibrina/metabolismo , Expressão Gênica/efeitos dos fármacos , Hipertrofia Ventricular Direita/etiologia , Hipertrofia Ventricular Direita/prevenção & controle , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Metformina/uso terapêutico , Ratos Wistar
7.
Blood ; 121(21): 4413-6, 2013 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-23550037

RESUMO

Mice deficient in the anticoagulants antithrombin (Serpinc1) or protein C (Proc) display premature death due to thrombosis-related coagulopathy, thereby precluding their use in gene function studies and thrombosis models. We used RNA interference to silence Serpinc1 and/or Proc in normal adult mice. The severe coagulopathy that followed combined "knockdown" of these genes is reported. Two days after siRNA injection, thrombi (occlusive) were observed in vessels (large and medium-sized) in multiple tissues, and hemorrhages were prominent in the ocular, mandibular, and maxillary areas. Tissue fibrin deposition and reduction of plasma fibrinogen accompanied this phenotype. The coagulopathy was prevented by dabigatran etexilate treatment. Silencing of Serpinc1 alone yielded a comparable but milder phenotype with later onset. The phenotype was absent when Proc was targeted alone. We conclude that RNA interference of Serpinc1 and/or Proc allows for evaluation of the function of these genes in vivo and provides a novel, controlled mouse model for spontaneous venous thrombosis.


Assuntos
Antitrombina III/genética , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Proteína C/genética , Trombose Venosa/genética , Trombose Venosa/fisiopatologia , Doença Aguda , Animais , Antitrombina III/fisiologia , Transtornos da Coagulação Sanguínea/genética , Transtornos da Coagulação Sanguínea/fisiopatologia , Feminino , Inativação Gênica , Fígado/fisiologia , Camundongos , Fenótipo , Proteína C/fisiologia , RNA Interferente Pequeno/genética , Índice de Gravidade de Doença
8.
Respir Res ; 16: 131, 2015 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-26498483

RESUMO

BACKGROUND: Dendritic cells (DCs) as professional antigen presenting cells (APCs) play a critical role in the regulation of host immune responses. DCs evolve from immature, antigen-capturing cells, to mature antigen-presenting cells. The relative contribution of DCs to cigarette smoke-induced inflammation is not well documented. In the current study, we investigated a modulatory effect of cigarette smoke extract (CSE) on differentiation, maturation and function of DCs. METHODS: Primary murine DCs were grown from bone marrow cells with GM-CSF. Development of DC was analyzed by expression of CD11c, MHCII, CD86, CD40 and CD83 using flow cytometry. Murine DC's and human L428 cells were co-cultured with CSE for various periods of time. Functional activity was analyzed by measuring FITC-dextran uptake, cytokine production and the ability to stimulate T cell activation in a mixed lymphocyte reaction. RESULTS: Our results show that short-term CSE stimulation (~24 h) influence the maturation status of newly differentiated and immature DCs towards more mature cells as revealed by upregulation of MHCII, CD83, CD86, CD40, reduction in antigen up-take capacity and enhanced secretion of pro-inflammatory (IL-12, IL-6 and TNF-α) cytokines. Interestingly, long-term CSE exposure, time- and concentration-dependently, suppressed the development of functional DCs. This suppression was demonstrated by a decline in CD11c/MHCII, CD83, CD86 and CD40 expression, the production of cytokines and ability to stimulate T lymphocytes. Moreover, CSE significantly suppressed the endocytosis function of mouse DCs which was not due to diminished DC viability. Similar to mouse DCs, long-term co-culturing of the human L428 DC cell line with CSE time-dependently suppressed the expression of CD54. CONCLUSIONS: The present study provides evidence that CSE modulates DC-mediated immune responses via affecting both the function and maturation of DCs. The suppressive effects of cigarette smoke on DC function might lead to impaired immune responses to various infections.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Células Dendríticas/efeitos dos fármacos , Ativação Linfocitária/efeitos dos fármacos , Fumaça/efeitos adversos , Fumar/efeitos adversos , Biomarcadores/metabolismo , Linhagem Celular Tumoral , Técnicas de Cocultura , Citocinas/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Relação Dose-Resposta a Droga , Humanos , Fenótipo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Fatores de Tempo
9.
Am J Physiol Lung Cell Mol Physiol ; 307(3): L261-72, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24951776

RESUMO

Intervening in angiotensin (Ang)-II type 2 receptor (AT2) signaling may have therapeutic potential for bronchopulmonary dysplasia (BPD) by attenuating lung inflammation and preventing arterial hypertension (PAH)-induced right ventricular hypertrophy (RVH). We first investigated the role of AT2 inhibition with PD123319 (0.5 and 2 mg·kg(-1)·day(-1)) on the beneficial effect of AT2 agonist LP2-3 (5 µg/kg twice a day) on RVH in newborn rats with hyperoxia-induced BPD. Next we determined the cardiopulmonary effects of PD123319 (0.1 mg·kg(-1)·day(-1)) in two 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 lung and heart histopathology, fibrin deposition, vascular leakage, and differential mRNA expression. Ten days of coadministration of LP2-3 and PD123319 abolished the beneficial effects of LP2-3 on RVH in experimental BPD. In the early treatment model PD123319 attenuated cardiopulmonary injury by reducing alveolar septal thickness, pulmonary influx of inflammatory cells, including macrophages and neutrophils, medial wall thickness of small arterioles, and extravascular collagen III deposition, and by preventing RVH. In the late treatment model PD123319 diminished PAH and RVH, demonstrating that PAH is reversible in the neonatal period. At high concentrations PD123319 blocks the beneficial effects of the AT2-agonist LP2-3 on RVH. At low concentrations PD123319 attenuates cardiopulmonary injury by reducing pulmonary inflammation and fibrosis and preventing PAH-induced RVH but does not affect alveolar and vascular development in newborn rats with experimental BPD.


Assuntos
Traumatismos Cardíacos/tratamento farmacológico , Hiperóxia/patologia , Imidazóis/farmacologia , Lesão Pulmonar/tratamento farmacológico , Piridinas/farmacologia , Receptor Tipo 2 de Angiotensina/metabolismo , Bloqueadores do Receptor Tipo 2 de Angiotensina II/farmacologia , Animais , Animais Recém-Nascidos , Displasia Broncopulmonar/tratamento farmacológico , Displasia Broncopulmonar/metabolismo , Displasia Broncopulmonar/patologia , Fibrina/metabolismo , Traumatismos Cardíacos/metabolismo , Traumatismos Cardíacos/patologia , Hiperóxia/metabolismo , Hipertrofia Ventricular Direita/tratamento farmacológico , Hipertrofia Ventricular Direita/metabolismo , Hipertrofia Ventricular Direita/patologia , Ligantes , Lesão Pulmonar/metabolismo , Lesão Pulmonar/patologia , Pneumonia/tratamento farmacológico , Pneumonia/metabolismo , Pneumonia/patologia , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/patologia , Ratos , Ratos Wistar
10.
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
11.
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
12.
Eur J Pharmacol ; 961: 176189, 2023 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-37951489

RESUMO

The renin-angiotensin system is one of the most complex regulatory systems that controls multiple organ functions. One of its key components, angiotensin II (Ang II), stimulates two G-protein coupled class A receptors: the Ang II type 1 (AT1) receptor and the Ang II type 2 (AT2) receptor. While stimulation of the AT1 receptor causes G-protein-dependent signaling and arrestin recruitment, the AT2 receptor seems to have a constitutively active-like conformation and appears to act via G-protein-dependent and -independent pathways. Overstimulation of the AT1 receptor may lead to unwanted effects like inflammation and fibrosis. In contrast, stimulation of the AT2 receptor leads to opposite effects thus restoring the balance. However, the role of the AT2 receptor has become controversial due to beneficial effects of putative AT2 receptor antagonists. The two first synthetic AT2 receptor-selective ligands, peptide CGP42112 and small molecule PD123319, were initially both considered antagonists. CGP42112 was subsequently considered a partial agonist and it was recently demonstrated to be a full agonist. Based on the search-term PD123319 in Pubmed, 1652 studies have investigated putative AT2 receptor antagonist PD123319. Here, we put forward literature that shows beneficial effects of PD123319 alone, even at doses too low for antagonist efficacy. These beneficial effects appear compatible with agonist-like activity via the AT2 receptor. Taken together, a more consistent image of a therapeutic role of stimulated AT2 receptor emerges which may clarify current controversies.


Assuntos
Receptor Tipo 1 de Angiotensina , Receptor Tipo 2 de Angiotensina , Receptor Tipo 2 de Angiotensina/metabolismo , Receptor Tipo 1 de Angiotensina/metabolismo , Sistema Renina-Angiotensina , Transdução de Sinais , Angiotensina II/farmacologia , Proteínas de Ligação ao GTP/metabolismo , Receptores de Angiotensina/metabolismo , Antagonistas de Receptores de Angiotensina
13.
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
14.
Front Pediatr ; 10: 943054, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36016884

RESUMO

Previous studies suggest that platelets are involved in fetal and adult lung development, but their role in postnatal lung development especially after premature birth is elusive. There is an urgent need to scrutinize this topic because the incidence of bronchopulmonary dysplasia (BPD), a chronic lung disease after premature birth, remains high. We have previously shown impaired platelet biogenesis in infants and rats with BPD. In this study, we investigated the role of anti-CD41 antibody-induced platelet depletion during normal postnatal lung development and thrombopoietin (TPO)-induced platelet biogenesis in mice with experimental BPD. We demonstrate that platelet deficient mice develop a BPD-like phenotype, characterized by enlarged alveoli and vascular remodeling of the small pulmonary arteries, resulting in pulmonary arterial hypertension (PAH)-induced right ventricular hypertrophy (RVH). Vascular remodeling was potentially caused by endothelial dysfunction demonstrated by elevated von Willebrand factor (vWF) concentration in plasma and reduced vWF staining in lung tissue with platelet depletion. Furthermore, TPO-induced platelet biogenesis in mice with experimental BPD improved alveolar simplification and ameliorated vascular remodeling. These findings demonstrate that platelets are indispensable for normal postnatal lung development and attenuation of BPD, probably by maintaining endothelial function.

15.
Am J Respir Crit Care Med ; 182(10): 1239-50, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-20622042

RESUMO

RATIONALE: Apelin, a potent vasodilator and angiogenic factor, may be a novel therapeutic agent in neonatal chronic lung disease, including bronchopulmonary dysplasia. OBJECTIVES: To determine the beneficial effect of apelin in neonatal rats with hyperoxia-induced lung injury, a model for premature infants with bronchopulmonary dysplasia. METHODS: The cardiopulmonary effects of apelin treatment (62 µg/kg/d) were studied in neonatal rats by exposure to 100% oxygen, using two treatment strategies: early concurrent treatment during continuous exposure to hyperoxia for 10 days and late treatment and recovery in which treatment was started on Day 6 after hyperoxic injury for 9 days and continued during the 9-day recovery period. We investigated in both models the role of the nitric oxide-cyclic guanosine monophosphate (cGMP) pathway in apelin treatment by specific inhibition of the nitric oxide synthase activity with N(ω)-nitro-L-arginine methyl ester (L-NAME, 25 mg/kg/d). MEASUREMENTS AND MAIN RESULTS: Parameters investigated include survival, lung and heart histopathology, pulmonary fibrin deposition and inflammation, alveolar vascular leakage, lung cGMP levels, right ventricular hypertrophy, and differential mRNA expression in lung and heart tissue. Prophylactic treatment with apelin improved alveolarization and angiogenesis, increased lung cGMP levels, and reduced pulmonary fibrin deposition, inflammation, septum thickness, arteriolar wall thickness, and right ventricular hypertrophy. These beneficial effects were completely absent in the presence of L-NAME. In the injury-recovery model apelin also improved alveolarization and angiogenesis, reduced arteriolar wall thickness, and attenuated right ventricular hypertrophy. CONCLUSIONS: Apelin reduces pulmonary inflammation, fibrin deposition, and right ventricular hypertrophy, and partially restores alveolarization in rat pups with neonatal hyperoxic lung injury via a nitric oxide synthase-dependent mechanism.


Assuntos
Proteínas de Transporte/uso terapêutico , Traumatismos Cardíacos/tratamento farmacológico , Hiperóxia/tratamento farmacológico , Lesão Pulmonar/tratamento farmacológico , Animais , Animais Recém-Nascidos , Apelina , Receptores de Apelina , Proteínas de Transporte/biossíntese , Proteínas de Transporte/fisiologia , GMP Cíclico/análise , Traumatismos Cardíacos/fisiopatologia , Hiperóxia/fisiopatologia , Hibridização In Situ , Inflamação/tratamento farmacológico , Inflamação/patologia , Inflamação/fisiopatologia , Peptídeos e Proteínas de Sinalização Intercelular , Pulmão/química , Pulmão/crescimento & desenvolvimento , Pulmão/patologia , Pulmão/fisiopatologia , Miocárdio/metabolismo , Miocárdio/patologia , NG-Nitroarginina Metil Éster/farmacologia , Ratos , Ratos Wistar , Receptores Acoplados a Proteínas G/biossíntese , Receptores Acoplados a Proteínas G/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
Front Pediatr ; 9: 625031, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34026682

RESUMO

Bronchopulmonary dysplasia (BPD) is a neonatal chronic lung disease characterized by an arrest in alveolar and vascular development. BPD is secondary to lung immaturity, ventilator-induced lung injury, and exposure to hyperoxia in extremely premature infants, leading to a lifelong impairment of lung function. Recent studies indicate that the lung plays an important role in platelet biogenesis. However, the dynamic change of platelet production during lung development and BPD pathogenesis remains to be elucidated. We investigated the dynamic change of platelet parameters in extremely premature infants during BPD development, and in newborn rats during their normal development from birth to adulthood. We further studied the effect of hyperoxia exposure on platelet production and concomitant pulmonary maldevelopment in an experimental BPD rat model induced by prolonged exposure to hyperoxia. We detected a physiological increase in platelet count from birth to 36 weeks postmenstrual age in extremely premature infants, but platelet counts in extremely premature infants who developed BPD were persistently lower than gestational age-matched controls. In line with clinical findings, exposure to hyperoxia significantly decreased the platelet count in neonatal rats. Lung morphometry analysis demonstrated that platelet counts stabilized with the completion of lung alveolarization in rats. Our findings indicate a close association between platelet biogenesis and alveolarization in the developing lung. This phenomenon might explain the reduced platelet count in extremely premature infants with BPD.

17.
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
18.
Nephrol Dial Transplant ; 24(3): 801-8, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18842674

RESUMO

BACKGROUND: Acute pyelonephritis is one of the most common bacterial infections. Tissue-type plasminogen activator (tPA) is a potent fibrinolytic agent, but can play a role in inflammatory processes as well. METHODS: We induced pyelonephritis in tPA(-/-) and C57BL/6 wild-type (WT) mice by intravesical inoculation with 10(10) CFU uropathogenic Escherichia coli 1677. The mice were killed after 24 and 48 h, after which bacterial outgrowth and cytokine levels in kidney homogenates were determined. Influx of neutrophils was quantified by myeloperoxidase-ELISA. Neutrophil phagocytosis and oxidative burst were measured. RESULTS: The tPA(-/-) kidneys contained significantly higher numbers of E. coli CFU, accompanied by higher levels of interleukin-1beta (IL-1beta) and tumour necrosis factor-alpha (TNF-alpha). The number of infiltrating neutrophils was similar in tPA(-/-) and WT mice at both time points, suggesting that tPA(-/-) neutrophils have a lower ability to eliminate E. coli. Phagocytosis of E. coli organisms was not diminished in tPA(-/-) neutrophils. Interestingly, tPA(-/-) neutrophils showed a significantly lower ability to generate an oxidative burst reaction upon stimulation with E. coli than WT neutrophils. Incubation with recombinant tPA reversed this effect completely. CONCLUSIONS: These results show that deletion of the tPA-gene in mice leads to lower bactericidal potential of tPA(-/-) neutrophils, which results in significantly more bacterial outgrowth during experimental pyelonephritis.


Assuntos
Infecções por Escherichia coli/genética , Infecções por Escherichia coli/metabolismo , Pielonefrite/etiologia , Pielonefrite/metabolismo , Ativador de Plasminogênio Tecidual/metabolismo , Animais , Modelos Animais de Doenças , Infecções por Escherichia coli/patologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/fisiologia , Fagocitose , Pielonefrite/patologia , RNA Mensageiro/metabolismo , Explosão Respiratória/fisiologia , Ativador de Plasminogênio Tecidual/genética
19.
Eur J Pharmacol ; 831: 52-59, 2018 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-29750914

RESUMO

In the gastro-intestinal tract, short chain fatty acids (SCFAs) have protective effects on epithelial cells. However, their effects on inflammatory cytokine production by endothelial and immune cells and the recruitment of immune cells and their trans-migration across the endothelial layer remain controversial. Both cell types are associated with the initiation and development of inflammatory diseases, such as atherosclerosis and sepsis. SCFAs modulate immune and inflammatory responses via activation of free fatty acid (FFA) receptors type 2 and 3 (FFA2 and FFA3 receptors), G protein-coupled receptor 109A (GPR109A) and inhibition of histone deacetylases (HDACs). This review will focus on the effects of SCFAs on lipopolysaccharide (LPS)- or tumor necrosis factor-alpha (TNFα)-induced inflammatory response on endothelial and immune cells function, and an overview is presented on the underlying mechanisms of the effects of SCFAs on both immune and endothelial cells, including HDACs, FFA2 and FFA3 receptors and GPR109A regulation of nuclear factor-kappa B (NF-κB) activation and mitogen-activated protein kinase (MAPK) signaling pathways.


Assuntos
Anti-Inflamatórios/farmacologia , Células Endoteliais/efeitos dos fármacos , Ácidos Graxos Voláteis/farmacologia , Sistema Imunitário/efeitos dos fármacos , Inflamação/tratamento farmacológico , Animais , Anti-Inflamatórios/toxicidade , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Ácidos Graxos Voláteis/toxicidade , Inibidores de Histona Desacetilases/farmacologia , Humanos , Sistema Imunitário/imunologia , Sistema Imunitário/metabolismo , Inflamação/induzido quimicamente , Inflamação/imunologia , Inflamação/metabolismo , Mediadores da Inflamação/metabolismo , Receptores de Superfície Celular/agonistas , Receptores de Superfície Celular/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo , Receptores Nicotínicos/metabolismo , Transdução de Sinais/efeitos dos fármacos
20.
Front Physiol ; 8: 155, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28382003

RESUMO

Aim: Survivors of neonatal chronic lung disease or bronchopulmonary dysplasia (BPD) suffer from compromised lung function and are at high risk for developing lung injury by multiple insults later in life. Because neonatal lysophosphatidic acid receptor-1 (LPAR1)-deficient rats are protected against hyperoxia-induced lung injury, we hypothesize that LPAR1-deficiency may protect adult survivors of BPD from a second hit response against lipopolysaccharides (LPS)-induced lung injury. Methods: Directly after birth, Wistar control and LPAR1-deficient rat pups were exposed to hyperoxia (90%) for 8 days followed by recovery in room air. After 7 weeks, male rats received either LPS (2 mg kg-1) or 0.9% NaCl by intraperitoneal injection. Alveolar development and lung inflammation were investigated by morphometric analysis, IL-6 production, and mRNA expression of cytokines, chemokines, coagulation factors, and an indicator of oxidative stress. Results: LPAR1-deficient and control rats developed hyperoxia-induced neonatal emphysema, which persisted into adulthood, as demonstrated by alveolar enlargement and decreased vessel density. LPAR1-deficiency protected against LPS-induced lung injury. Adult controls with BPD exhibited an exacerbated response toward LPS with an increased expression of pro-inflammatory mRNAs, whereas LPAR1-deficient rats with BPD were less sensitive to this "second hit" with a decreased pulmonary influx of macrophages and neutrophils, interleukin-6 (IL-6) production, and mRNA expression of IL-6, monocyte chemoattractant protein-1, cytokine-induced neutrophil chemoattractant 1, plasminogen activator inhibitor-1, and tissue factor. Conclusion: LPAR1-deficient rats have increased hyperoxia-induced BPD survival rates and, despite the presence of neonatal emphysema, are less sensitive to an aggravated "second hit" than Wistar controls with BPD. Intervening in LPA-LPAR1-dependent signaling may not only have therapeutic potential for neonatal chronic lung disease, but may also protect adult survivors of BPD from sequelae later in life.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA