Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Int J Mol Sci ; 22(16)2021 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-34445253

RESUMO

Bronchopulmonary dysplasia (BPD) is the most common complication of prematurity and a key contributor to the large health care burden associated with prematurity, longer hospital stays, higher hospital costs, and frequent re-hospitalizations of affected patients through the first year of life and increased resource utilization throughout childhood. This disease is associated with abnormal pulmonary function that may lead to BPD-associated pulmonary hypertension (PH), a major contributor to neonatal mortality and morbidity. In the absence of any definitive treatment options, this life-threatening disease is associated with high resource utilization during and after neonatal intensive care unit (NICU) stay. The goal of this study was to test the safety and efficacy of a small molecule derivative of chitin, AVR-48, as prophylactic therapy for preventing experimental BPD in a mouse model. Two doses of AVR-48 were delivered either intranasally (0.11 mg/kg), intraperitoneally (10 mg/kg), or intravenously (IV) (10 mg/kg) to newborn mouse pups on postnatal day (P)2 and P4. The outcomes were assessed by measuring total inflammatory cells in the broncho-alveolar lavage fluid (BALF), chord length, septal thickness, and radial alveolar counts of the alveoli, Fulton's Index (for PH), cell proliferation and cell death by immunostaining, and markers of inflammation by Western blotting and ELISA. The bioavailability and safety of the drug were assessed by pharmacokinetic and toxicity studies in both neonatal mice and rat pups (P3-P5). Following AVR-48 treatment, alveolar simplification was improved, as evident from chord length, septal thickness, and radial alveolar counts; total inflammatory cells were decreased in the BALF; Fulton's Index was decreased and lung inflammation and cell death were decreased, while angiogenesis and cell proliferation were increased. AVR-48 was found to be safe and the no-observed-adverse-effect level (NOAEL) in rat pups was determined to be 100 mg/kg when delivered via IV dosing with a 20-fold safety margin. With no reported toxicity and with a shorter half-life, AVR-48 is able to reverse the worsening cardiopulmonary phenotype of experimental BPD and BPD-PH, compared to controls, thus positioning it as a future drug candidate.


Assuntos
Displasia Broncopulmonar , Quitina , Hipertensão Pulmonar , Neovascularização Fisiológica/efeitos dos fármacos , Alvéolos Pulmonares , Animais , Animais Recém-Nascidos , Displasia Broncopulmonar/tratamento farmacológico , Displasia Broncopulmonar/metabolismo , Displasia Broncopulmonar/patologia , Quitina/química , Quitina/farmacologia , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Hipertensão Pulmonar/tratamento farmacológico , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/patologia , Camundongos , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/patologia , Ratos
2.
Zhongguo Dang Dai Er Ke Za Zhi ; 20(5): 410-415, 2018 May.
Artigo em Chinês | MEDLINE | ID: mdl-29764580

RESUMO

OBJECTIVE: To study the effect of rhubarb on neonatal rats with bronchopulmonary dysplasia (BPD) induced by hyperoxia. METHODS: A total of 64 rats (postnatal day 4) were randomly divided into four groups: air control, rhubarb control, hyperoxia model, and hyperoxia+rhubarb (n=16 each). The rats in the hyperoxia model and hyperoxia+rhubarb groups were exposed to hyperoxia (60% O2) to establish a BPD model. The rats in the rhubarb control and hyperoxia+rhubarb groups were given rhubarb extract suspension (600 mg/kg) by gavage daily. The pathological changes of lung tissue were evaluated by hematoxylin-eosin staining on postnatal days 14 and 21. The content of malondialdehyde (MDA) and the activity of superoxide dismutase (SOD) were measured by spectrophotometry. The mRNA and protein expression levels of tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6) were determined by RT-PCR and Western blot respectively. RESULTS: The hyperoxia model group showed reduced alveolar number, increased alveolar volume, and simplified alveolar structure, which worsened over the time of exposure to hyperoxia. These pathological changes were significantly reduced in the hyperoxia+rhubarb group. On postnatal days 14 and 21, compared with the air control and rhubarb control groups, the hyperoxia model group had significantly reduced radical alveolar count (RAC), significantly reduced activity of SOD in the lung tissue, and significantly increased content of MDA and mRNA and protein expression levels of TNF-α and IL-6 (P<0.05). Compared with the hyperoxia model group, the hyperoxia+rhubarb group had significantly increased RAC, significantly increased activity of SOD in the lung tissue, and significantly reduced content of MDA and mRNA and protein expression levels of TNF-α and IL-6 (P<0.05). CONCLUSIONS: Rhubarb may play a protective role in rats with BPD induced by hyperoxia through inhibiting inflammatory response and oxidative stress.


Assuntos
Displasia Broncopulmonar/prevenção & controle , Hiperóxia/complicações , Extratos Vegetais/uso terapêutico , Rheum , Animais , Animais Recém-Nascidos , Displasia Broncopulmonar/metabolismo , Displasia Broncopulmonar/patologia , Modelos Animais de Doenças , Pulmão/metabolismo , Pulmão/patologia , Ratos , Ratos Sprague-Dawley , Superóxido Dismutase/metabolismo , Fator de Necrose Tumoral alfa/genética
3.
Biochem Biophys Res Commun ; 495(2): 1972-1979, 2018 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-29242152

RESUMO

The incidence and mortality rates of bronchopulmonary dysplasia (BPD) remain very high. Therefore, novel therapies are imminently needed to improve the outcome of this disease. Human umbilical cord-derived mesenchymal stem cells (UC-MSCs) show promising therapeutic effects on oxygen-induced model of BPD. In our experiment, UC-MSCs were intratracheally delivered into the newborn rats exposed to hyperoxia, a well-established BPD model. This study demonstrated that UC-MSCs reduce elastin expression stimulated by 90% O2 in human lung fibroblasts-a (HLF-a), and inhibit HLF-a transdifferentiation into myofibroblasts. In addition, the therapeutic effects of UC-MSCs in neonatal rats with BPD, UC-MSCs could inhibit lung elastase activity and reduce aberrant elastin expression and deposition in the lung of BPD rats. Overall, this study suggested that UC-MSCs could ameliorate aberrant elastin expression in the lung of hyperoxia-induced BPD model which may be associated with suppressing increased TGFß1 activation.


Assuntos
Displasia Broncopulmonar/imunologia , Displasia Broncopulmonar/patologia , Transplante de Células-Tronco de Sangue do Cordão Umbilical/métodos , Elastina/metabolismo , Pulmão/imunologia , Pulmão/patologia , Transplante de Células-Tronco Mesenquimais/métodos , Animais , Animais Recém-Nascidos , Células Cultivadas , Humanos , Oxigenoterapia Hiperbárica , Hiperóxia/metabolismo , Hiperóxia/patologia , Hiperóxia/prevenção & controle , Lesão Pulmonar/imunologia , Lesão Pulmonar/patologia , Lesão Pulmonar/prevenção & controle , Ratos , Ratos Sprague-Dawley , Resultado do Tratamento
4.
J Reprod Immunol ; 124: 21-29, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-29035757

RESUMO

Bronchopulmonary dysplasia (BPD) and BPD-associated pulmonary hypertension (BPD-PH) are chronic inflammatory cardiopulmonary diseases with devastating short- and long-term consequences for infants born prematurely. The immature lungs of preterm infants are ill-prepared to achieve sufficient gas exchange, thus usually necessitating immediate commencement of respiratory support and oxygen supplementation. These therapies are life-saving, but they exacerbate the tissue damage that is inevitably inflicted on a preterm lung forced to perform gas exchange. Together, air-breathing and necessary therapeutic interventions disrupt normal lung development by aggravating pulmonary inflammation and vascular remodelling, thus frequently precipitating BPD and PH via an incompletely understood pathogenic cascade. BPD and BPD-PH share common risk factors, such as low gestational age at birth, fetal growth restriction and perinatal maternal inflammation; however, these risk factors are not unique to BPD or BPD-PH. Occurring in 17-24% of BPD patients, BPD-PH substantially worsens the morbidity and mortality attributable to BPD alone, thus darkening their outlook; for example, BPD-PH entails a mortality of up to 50%. The absence of a safe and effective therapy for BPD and BPD-PH renders neonatal cardiopulmonary disease an area of urgent unmet medical need. Besides the need to develop new therapeutic strategies, a major challenge for clinicians is the lack of a reliable method for identifying babies at risk of developing BPD and BPD-PH. In addition to discussing current knowledge on pathophysiology, diagnosis and treatment of BPD-PH, we highlight emerging biomarkers that could enable clinicians to predict disease-risk and also optimise treatment of BPD-PH in our tiniest patients.


Assuntos
Displasia Broncopulmonar/epidemiologia , Displasia Broncopulmonar/patologia , Hipertensão Pulmonar/epidemiologia , Doenças do Recém-Nascido/epidemiologia , Recém-Nascido Prematuro/fisiologia , Nascimento Prematuro/epidemiologia , Animais , Desenvolvimento Fetal , Humanos , Oxigenoterapia Hiperbárica , Hipertensão Pulmonar/patologia , Lactente , Recém-Nascido , Doenças do Recém-Nascido/patologia , Inflamação , Nascimento Prematuro/patologia , Respiração , Remodelação Vascular
5.
Am J Physiol Lung Cell Mol Physiol ; 308(10): L983-L1001, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25770179

RESUMO

This paper is focused on unique insights provided by the preterm lamb physiological model of bronchopulmonary dysplasia (BPD). Connections are also made to insights provided by the former preterm baboon model of BPD, as well as to rodent models of lung injury to the immature, postnatal lung. The preterm lamb and baboon models recapitulate the clinical setting of preterm birth and respiratory failure that require prolonged ventilation support for days or weeks with oxygen-rich gas. An advantage of the preterm lamb model is the large size of preterm lambs, which facilitates physiological studies for days or weeks during the evolution of neonatal chronic lung disease (CLD). To this advantage is linked an integrated array of morphological, biochemical, and molecular analyses that are identifying the role of individual genes in the pathogenesis of neonatal CLD. Results indicate that the mode of ventilation, invasive mechanical ventilation vs. less invasive high-frequency nasal ventilation, is related to outcomes. Our approach also includes pharmacological interventions that test causality of specific molecular players, such as vitamin A supplementation in the pathogenesis of neonatal CLD. The new insights that are being gained from our preterm lamb model may have important translational implications about the pathogenesis and treatment of BPD in preterm human infants.


Assuntos
Modelos Animais de Doenças , Recém-Nascido Prematuro , Respiração Artificial , Animais , Animais Recém-Nascidos , Displasia Broncopulmonar/patologia , Displasia Broncopulmonar/fisiopatologia , Displasia Broncopulmonar/terapia , Doença Crônica , Humanos , Recém-Nascido , Pulmão , Ovinos
6.
Birth Defects Res A Clin Mol Teratol ; 100(3): 134-44, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24578124

RESUMO

Since Northway's original description of BPD almost 45 years ago, the clinical presentation of BPD has evolved into a disease process, which mostly involves extremely premature infants. This new form of BPD is the result of multiple antenatal and postnatal factors that can cause injury to the developing lung leading to altered alveolar and vascular development. Over the years, there has been considerable increase in knowledge of factors that contribute to the development of BPD. This has led to different strategies for prevention as well as management of BPD. Some of these strategies have been successful and have withstood the test of clinical trials, such as vitamin A supplementation, post-natal steroids, caffeine, and volume targeted ventilation. The evidence for other interventions has been weak or negative. With better understanding of the complex and multifactorial pathogenesis of BPD, it is quite clear that any single therapy is very unlikely to eliminate this problem unless it reduces prematurity. Further development in prevention and treatment of BPD will likely need a multi-pronged strategy with novel therapeutic agents acting at various stages of the disease process.


Assuntos
Displasia Broncopulmonar , Recém-Nascido Prematuro , Displasia Broncopulmonar/etiologia , Displasia Broncopulmonar/patologia , Displasia Broncopulmonar/terapia , Cafeína/uso terapêutico , Estimulantes do Sistema Nervoso Central/uso terapêutico , Ensaios Clínicos como Assunto , Humanos , Recém-Nascido , Alvéolos Pulmonares/patologia , Respiração Artificial , Esteroides/uso terapêutico , Vitamina A/uso terapêutico , Vitaminas/uso terapêutico
7.
Antimicrob Agents Chemother ; 57(5): 2127-33, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23439637

RESUMO

Ureaplasma respiratory tract colonization is associated with bronchopulmonary dysplasia (BPD) in preterm infants. Previously, we demonstrated that a single intravenous (i.v.) dose of azithromycin (10 mg/kg of body weight) is safe but inadequate to eradicate Ureaplasma spp. in preterm infants. We performed a nonrandomized, single-arm open-label study of the pharmacokinetics (PK) and safety of intravenous 20-mg/kg single-dose azithromycin in 13 mechanically ventilated neonates with a gestational age between 24 weeks 0 days and 28 weeks 6 days. Pharmacokinetic data from 25 neonates (12 dosed with 10 mg/kg i.v. and 13 dosed with 20 mg/kg i.v.) were analyzed using a population modeling approach. Using a two-compartment model with allometric scaling of parameters on body weight (WT), the population PK parameter estimates were as follows: clearance, 0.21 liter/h × WT(kg)(0.75) [WT(kg)(0.75) indicates that clearance was allometrically scaled on body weight (in kilograms) with a fixed exponent of 0.75]; intercompartmental clearance, 2.1 liters/h × WT(kg)(0.75); central volume of distribution (V), 1.97 liters × WT (kg); and peripheral V, 17.9 liters × WT (kg). There was no evidence of departure from dose proportionality in azithromycin exposure over the tested dose range. The calculated area under the concentration-time curve over 24 h in the steady state divided by the MIC90 (AUC24/MIC90) for the single dose of azithromycin (20 mg/kg) was 7.5 h. Simulations suggest that 20 mg/kg for 3 days will maintain azithromycin concentrations of >MIC50 of 1 µg/ml for this group of Ureaplasma isolates for ≥ 96 h after the first dose. Azithromycin was well tolerated with no drug-related adverse events. One of seven (14%) Ureaplasma-positive subjects and three of six (50%) Ureaplasma-negative subjects developed physiologic BPD. Ureaplasma was eradicated in all treated Ureaplasma-positive subjects. Simulations suggest that a multiple-dose regimen may be efficacious for microbial clearance, but the effect on BPD remains to be determined.


Assuntos
Antibacterianos/uso terapêutico , Azitromicina/uso terapêutico , Displasia Broncopulmonar/tratamento farmacológico , Recém-Nascido Prematuro , Modelos Estatísticos , Sistema Respiratório/efeitos dos fármacos , Ureaplasma/efeitos dos fármacos , Antibacterianos/farmacocinética , Área Sob a Curva , Azitromicina/farmacocinética , Peso Corporal , Displasia Broncopulmonar/microbiologia , Displasia Broncopulmonar/patologia , Relação Dose-Resposta a Droga , Esquema de Medicação , Feminino , Idade Gestacional , Humanos , Recém-Nascido , Injeções Intravenosas , Masculino , Testes de Sensibilidade Microbiana , Sistema Respiratório/microbiologia , Sistema Respiratório/patologia , Resultado do Tratamento , Ureaplasma/crescimento & desenvolvimento
8.
Nutrition ; 28(11-12): 1186-91, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23044165

RESUMO

OBJECTIVES: Supplementation studies of glutamine, arginine, and docosahexaenoic acid (DHA) have established the safety of each of these nutrients in neonates. However, the potential for a more stable and soluble dipeptide, arginyl-glutamine (Arg-Gln) or DHA, a long-chain ω-3 fatty acid with anti-inflammatory properties, to exert benefits on hyperoxia-induced lung injury has not to our knowledge been investigated. The aim of this study was to investigate whether Arg-Gln dipeptide or DHA could attenuate markers of injury and inflammation in neonatal mouse lungs exposed to hyperoxia. METHODS: Seven-day-old mouse pups were placed with their dams in 75% oxygen for 5 d. After 5 d of hyperoxic exposure (postnatal days 7-12), pups were removed from hyperoxia and allowed to recover in atmospheric conditions for 5 d (postnatal days 12-17). Mouse pups received Arg-Gln (5 g · kg⁻¹ · d⁻¹) or DHA (5 g · kg⁻¹ · d⁻¹) or saline orally from postnatal days 12 through 17. Histologic changes, myeloperoxidase, lactate dehydrogenase, inflammatory cytokines, and nuclear factor-κB inhibitor levels were checked in each group. RESULTS: The Arg-Gln and DHA prevented the development of key markers of injury, including histologic changes, myeloperoxidase, lactate dehydrogenase, and inflammatory cytokines interleukin-6 and C-X-C motif ligand 1 (CXCL1)/keratinocyte-derived chemokine (KC). The highly beneficial effects of Arg-Gln on the reversal of oxygen-induced lung damage was associated with restoration of levels of nuclear factor-κB inhibitor. CONCLUSION: The Arg-Gln and DHA, with protective effects on hyperoxic lung injury in neonatal mice, are promising nutritional adjuncts that may prevent lung damage owing to oxygen toxicity in infants.


Assuntos
Displasia Broncopulmonar/prevenção & controle , Suplementos Nutricionais , Dipeptídeos/uso terapêutico , Modelos Animais de Doenças , Ácidos Docosa-Hexaenoicos/uso terapêutico , Hiperóxia/fisiopatologia , Pulmão/fisiopatologia , Animais , Animais Recém-Nascidos , Câmaras de Exposição Atmosférica , Biomarcadores/sangue , Biomarcadores/metabolismo , Displasia Broncopulmonar/etiologia , Displasia Broncopulmonar/metabolismo , Displasia Broncopulmonar/patologia , Citocinas/sangue , Citocinas/metabolismo , Feminino , Humanos , Recém-Nascido , Mediadores da Inflamação/sangue , Mediadores da Inflamação/metabolismo , Lactação , Pulmão/metabolismo , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
9.
Pediatr Res ; 68(6): 519-25, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20805789

RESUMO

Bronchopulmonary dysplasia (BPD) is characterized by arrested alveolar development and complicated by pulmonary hypertension (PH). NO promotes alveolar growth. Inhaled NO (iNO) ameliorates the BPD phenotype in experimental models and in some premature infants. Arginosuccinate synthetase (ASS) and arginosuccinate lyase (ASL) convert L-citrulline to L-arginine; L-citrulline is regenerated during NO synthesis from L-arginine. Plasma levels of these NO precursors are low in PH. We hypothesized that L-citrulline prevents experimental O2-induced BPD in newborn rats. Rat pups were assigned from birth through postnatal day (P) 14 to room air (RA), RA + L-citrulline, 95% hyperoxia (BPD model), and 95%O2 + L-citrulline. Rat pups exposed to hyperoxia had fewer and enlarged air spaces and decreased capillary density, mimicking human BPD. This was associated with decreased plasma L-arginine and L-citrulline concentrations on P7. L-citrulline treatment significantly increased plasma L-arginine and L-citrulline concentrations and increased ASL protein expression in hyperoxia. L-citrulline preserved alveolar and vascular growth in O2-exposed pups and decreased pulmonary arterial medial wall thickness (MWT) and right ventricular hypertrophy (RVH). Increased lung arginase (ARG) activity in O2-exposed pups was reversed by L-citrulline treatment. L-citrulline supplementation prevents hyperoxia-induced lung injury and PH in newborn rats. L-citrulline may represent a novel therapeutic alternative to iNO for prevention of BPD.


Assuntos
Animais Recém-Nascidos , Displasia Broncopulmonar/prevenção & controle , Displasia Broncopulmonar/fisiopatologia , Citrulina/uso terapêutico , Hipertensão Pulmonar/prevenção & controle , Alvéolos Pulmonares/crescimento & desenvolvimento , Animais , Arginina/sangue , Displasia Broncopulmonar/sangue , Displasia Broncopulmonar/patologia , Citrulina/sangue , Citrulina/farmacologia , Modelos Animais de Doenças , Humanos , Hipertensão Pulmonar/fisiopatologia , Recém-Nascido , Pulmão/patologia , Pulmão/fisiopatologia , Lesão Pulmonar , Óxido Nítrico/sangue , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/fisiopatologia , Ratos
10.
Biol Neonate ; 89(4): 303-12, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16770070

RESUMO

BACKGROUND: Recent improvements in the survival of extremely preterm infants have been accompanied by evolution in the pathogenesis and histopathology of bronchopulmonary dysplasia (BPD). Although oxygen and barotrauma-induced injury remain important contributing factors, pulmonary developmental arrest appears to play an equally important causal role in prolonged respiratory illness, especially among the most immature surviving preterm newborns. To date, clinical trials have failed to demonstrate a substantial benefit of a single treatment or preventive strategy for BPD. OBJECTIVES: To evaluate the current evidence in favor of treatments that might prevent BPD. METHODS: Review of clinical studies of preventive treatment strategies for BPD. RESULTS: High frequency oscillatory ventilation, permissive hypercapnea, and inhaled nitric oxide might offer benefit to infants at risk of BPD. These and other potential preventive therapies for BPD, such as superoxide dismutase, inositol, and alpha(1)-proteinase inhibitor, deserve further study. CONCLUSIONS: Although some current treatments offer promise, no preventive therapy for BPD has proven safe and effective, except for intramuscular vitamin A. Additional studies of respiratory technologies, management strategies, and protective molecules are needed.


Assuntos
Displasia Broncopulmonar/prevenção & controle , Displasia Broncopulmonar/terapia , Animais , Antioxidantes/metabolismo , Displasia Broncopulmonar/metabolismo , Displasia Broncopulmonar/patologia , Pressão Positiva Contínua nas Vias Aéreas , Glucocorticoides/uso terapêutico , Humanos , Fenômenos Fisiológicos da Nutrição do Lactente , Recém-Nascido , Inibidores de Proteases/uso terapêutico
11.
Pediatr Res ; 43(6): 719-26, 1998 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9621979

RESUMO

The antioxidant vitamins ascorbic acid (AA) and alpha-tocopherol (alpha-TP) effectively inhibit oxygen free radical-induced lipid peroxidation. Using a premature baboon model of hyperoxia-induced bronchopulmonary dysplasia (BPD), we measured concentrations of AA, alpha-TP, and conjugated dienes (CD, marker of lipid peroxidation) in four animals (hyperoxic antioxidant group) receiving high dose antioxidant vitamin supplementation (AA, 100 mg x kg x(-1) x d(-1); alpha-TP; 20 mg x kg x(-1) x d(-1)) and one animal receiving standard dose antioxidant vitamin supplementation (AA, 10 mg x kg x(-1) x d(-1); alpha-TP, 1 mg x kg x(-1) x d(-1)). Respiratory and histopathologic data were compared with data from 10 historical animals exposed to hyperoxia (hyperoxic control group) and 11 historical animals treated as required with oxygen (normoxic control group) who had received standard dose antioxidant vitamin supplementation. Compared with standard dose antioxidant vitamin supplementation, high dose antioxidant vitamin supplementation effectively raised AA concentrations in plasma (37 +/- 22 micromol/L and 395 +/- 216 micromol/L, respectively) and tracheal aspirates (62 +/- 35 micromol/L and 286 +/- 205 micromol/L, respectively), and alpha-TP concentrations in plasma (10.1 +/- 2.5 micromol/L and 24.6 +/- 17.5 micromol/L, respectively). However, there was no apparent effect on tracheal aspirate CD concentrations (482 +/- 333 micromol/L and 1050 +/- 1111 micromol/L, respectively), and respiratory parameters in the hyperoxic antioxidant group were comparable to those of the hyperoxic control group but significantly worse than in the normoxic control group. Finally, no protective effect of high dose antioxidant vitamin supplementation was noted at the histopathologic level.


Assuntos
Animais Recém-Nascidos/fisiologia , Antioxidantes/uso terapêutico , Displasia Broncopulmonar/prevenção & controle , Pulmão/patologia , Vitamina E/uso terapêutico , Animais , Displasia Broncopulmonar/etiologia , Displasia Broncopulmonar/patologia , Modelos Animais de Doenças , Idade Gestacional , Humanos , Hiperóxia , Recém-Nascido , Pulmão/efeitos dos fármacos , Papio , Projetos Piloto , Análise de Regressão
12.
Lab Invest ; 59(1): 25-35, 1988 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-3260640

RESUMO

Twelve pairs of fetal lambs were used to test the hypothesis that the necrotizing tracheobronchitis followed by squamous metaplasia seen in premature infants who develop chronic bronchopulmonary dysplasia might be related to low retinol stores and might, therefore, be reversed by retinol supplementation. Epidermal growth factor (EGF) was used to model the growth factor stimulus initiated by chronic wounding of the airways, and retinol was used as a differentiator of proliferating cells stimulated by EGF. Saline-treated animals were used as controls, as were fetal lambs receiving retinol alone or EGF alone. The effects of EGF on tracheal and bronchial epithelium consisted of proliferation of basal and intermediate cells, necrosis and slough of lining ciliated and mucous-producing cells, followed by squamous metaplasia. In fetal lambs given retinol, plasma, liver and lung retinol levels rose and mucous producing cells were increased in number. In the presence of EGF plus retinol, differentiation of mucous-producing cells was accelerated. We believe that this fetal lamb model with low initial levels of retinol in plasma, liver and lung, treated with EGF may mimic human premature infants with chronic bronchopulmonary dysplasia, and that the addition of retinol in amounts sufficient to raise their tissue levels produces a more normal surface epithelium in conducting airways.


Assuntos
Displasia Broncopulmonar/etiologia , Fator de Crescimento Epidérmico/farmacologia , Traqueia/efeitos dos fármacos , Deficiência de Vitamina A/complicações , Vitamina A/farmacologia , Análise de Variância , Animais , Animais Recém-Nascidos , Displasia Broncopulmonar/tratamento farmacológico , Displasia Broncopulmonar/patologia , Diferenciação Celular , Divisão Celular , Modelos Animais de Doenças , Fator de Crescimento Epidérmico/administração & dosagem , Fator de Crescimento Epidérmico/uso terapêutico , Epitélio/efeitos dos fármacos , Epitélio/patologia , Epitélio/ultraestrutura , Feminino , Feto , Humanos , Recém-Nascido , Metaplasia , Microscopia Eletrônica , Gravidez , Ovinos , Traqueia/patologia , Traqueia/ultraestrutura , Vitamina A/administração & dosagem , Vitamina A/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA