Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Fetal Pediatr Pathol ; 42(5): 766-774, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37341579

RESUMO

Introduction: Oxidative stress and inflammation have proven to be key factors contributing to the occurrence of BPD. Erythromycin has been shown to be effective in treating the redox imbalance seen in many non-bacterial infectious chronic inflammatory diseases. Methods: Ninety-six premature rats were randomly divided into air + saline chloride group, air + erythromycin group, hyperoxia + saline chloride group and hyperoxia + erythromycin group. Lung tissue specimens were collected from 8 premature rats in each group on days 1, 7 and 14, respectively. Results: Pulmonary pathological changes in premature rats after hyperoxia exposure were similar to those of BPD. Hyperoxia exposure induced high expression of GSH, TNF-α, and IL-1ß. Erythromycin intervention caused a further increase in GSH expression and a decrease in TNF-α and IL-1ß expression. Conclusion: GSH, TNF-α and IL-1ß are all involved in the development of BPD. Erythromycin may alleviate BPD by enhancing the expression of GSH and inhibiting the release of inflammatory mediators.


Assuntos
Displasia Broncopulmonar , Hiperóxia , Lesão Pulmonar , Animais , Ratos , Lesão Pulmonar/tratamento farmacológico , Lesão Pulmonar/etiologia , Lesão Pulmonar/metabolismo , Hiperóxia/complicações , Hiperóxia/metabolismo , Hiperóxia/patologia , Citocinas/metabolismo , Citocinas/farmacologia , Eritromicina/farmacologia , Eritromicina/metabolismo , Animais Recém-Nascidos , Fator de Necrose Tumoral alfa/metabolismo , Cloretos/metabolismo , Cloretos/farmacologia , Pulmão , Inflamação/patologia , Displasia Broncopulmonar/complicações , Displasia Broncopulmonar/metabolismo , Modelos Animais de Doenças
2.
BMC Cardiovasc Disord ; 21(1): 514, 2021 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-34689755

RESUMO

BACKGROUND: Pulmonary hypertension (PH) is a common complication of bronchopulmonary dysplasia (BPD) in very-low-birth-weight infants (VLBWIs). Although recent studies have increased awareness that PH contributes significantly to the high morbidity and mortality of BPD, the risk factors and clinical characteristics for PH in VLBWIs are little known. OBJECTIVES: To investigate the risk factors and clinical characteristics for BPD-associated pulmonary hypertension (BPD-PH) in VLBWIs. METHODS: A retrospective case-control observational study of VLBWIs with BPD admitted to a neonatal intensive care unit (NICU) over 4 years. According to echocardiograms confirming elevated pulmonary artery pressure after 28 days after birth, we divided BPD infants into PH group (n = 18) and non-PH group (n = 65). We compared pre- and postnatal characteristics between VLBWIs with or without PH. Multivariable logistic regression analysis was conducted with backward selection. RESULTS: A total of 83 infants with BPD were divided into PH group (n = 18) or non-PH group (n = 65). The average birth weight of the infants with BPD was 1078.1 g. Compared with those infants of the non-PH group, the birth weight of BPD-PH infants was significantly lower (968.1 ± 187.7 vs. 1108.5 ± 185.8, P = 0.006). Infants in the PH group had a higher incidence of patent ductus arteriosus (PDA) and underwent longer durations of oxygen therapy and mechanical ventilation compared to those in the non-PH group. In all subjects, birth weight (OR 0.995; 95% CI 0.991-0.999; P = 0.025) and PDA (OR 13.355; 95% CI 2.950-60.469; P = 0.001) were found to be specific risk factors for BPD-PH in this cohort. CONCLUSIONS: The study shows PDA and birth weight are specific risk factors for BPD-PH in VLBWIs.


Assuntos
Displasia Broncopulmonar/complicações , Permeabilidade do Canal Arterial/complicações , Hipertensão Pulmonar/etiologia , Recém-Nascido de muito Baixo Peso , Peso ao Nascer , Estudos de Casos e Controles , Feminino , Humanos , Recém-Nascido , Masculino , Oxigenoterapia , Respiração Artificial , Estudos Retrospectivos , Fatores de Risco
3.
Transl Pediatr ; 10(2): 265-275, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33708512

RESUMO

BACKGROUND: Bronchopulmonary dysplasia (BPD) is a common respiratory disease in premature infants and is characterized by alveolar and pulmonary vascular dysplasia. Long-term oxygen exposure can cause BPD in preterm infants. Numerous studies have shown that long non-coding ribonucleic acid (lncRNA) is involved in the process of biological metabolism; however, its role in the development of BPD is unclear. Apoptosis-induced factor (AIF) is a key component involved in apoptosis. The Kelch-like ECH-associated protein 1/nuclear factor erythroid-2-related factor 2 (Keap1/Nrf2) signaling pathway is a body-derived antioxidant signaling pathway. METHODS: In this study, the relative expression of metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), AIF, Keap1, and Nrf2 was detected by real-time polymerase chain reaction (PCR). Also, the apoptosis of A549 cells was detected by flow cytometry. RESULTS: The results showed that, compared to the control group, the expression of MALAT1 increased significantly, and AIF decreased substantially in BPD premature infants. In the A549 hyperoxic lung injury model, compared with the air group, the expression of MALAT1 in the hyperoxia group decreased markedly, while the expression of Keap1 and Nrf2 increased considerably. Furthermore, compared with the control plasmid transfection air group (NC group), the expression of Keap1 and Nrf2 increased significantly in the small interfering RNA (siRNA) group. CONCLUSIONS: These results indicate that MALAT1 can play a protective role in BPD via the reduction of apoptosis and anti-oxidation, offering clinicians a new way to prevent and treat BPD.

4.
Mol Med Rep ; 23(3)2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33495821

RESUMO

Bronchopulmonary dysplasia (BPD) is one of the main causes of chronic lung disease in premature infants. Acute lung injury following exposure to hyperoxia contributes to the development of BPD in preterm infants. The nuclear factor­erythroid 2­related factor 2 (Nrf2) signaling pathway is an endogenous antioxidant defense mechanism that is involved in the pathogenesis of numerous hyperoxia­induced diseases. In the present study, the expression of Nrf2, Kelch­like ECH­associated protein 1 (Keap1) and NAD(P)H quinone oxidoreductase 1 enzyme (NQO1) was detected in A549 cells exposed to hyperoxia and transfection with small interfering RNA (siRNA) using reverse transcription­quantitative polymerase chain reaction and western blotting, and cellular apoptosis was detected using flow cytometry. The results demonstrated that apoptosis increased significantly following exposure of the cells to hyperoxia, and Nrf2, Keap1 and NQO1 expression levels were significantly upregulated under hyperoxic conditions. Furthermore, following transfection with Nrf2 siRNA, the expression levels of these genes were significantly downregulated and apoptosis was significantly increased compared with the respective values in untransfected cells. These findings suggest that the Nrf2­Keap1­antioxidant response element­NQO1 signaling pathway may play a protective role in hyperoxia­induced lung injury via the inhibition of apoptosis.


Assuntos
Elementos de Resposta Antioxidante , Apoptose , Hiperóxia/metabolismo , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Pulmão/metabolismo , NAD(P)H Desidrogenase (Quinona)/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Transdução de Sinais , Células A549 , Humanos , Hiperóxia/genética , Proteína 1 Associada a ECH Semelhante a Kelch/genética , Pulmão/patologia , NAD(P)H Desidrogenase (Quinona)/genética , Fator 2 Relacionado a NF-E2/genética
5.
Front Pediatr ; 8: 271, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32537448

RESUMO

Bronchopulmonary dysplasia (BPD) is one of the common chronic lung diseases (CLD) of premature infants, which causes unpredictable consequences to the family and society. Therefore, the pathogenesis and prevention methods of BPD are the focus of current research, and the establishment of an effective and appropriate animal model of BPD in premature infants is the key to the research. In this study, premature rats were exposed to hyperoxia environment. Compared with the air group, the body weight and alveolar radiation count of the hyperoxia group decreased significantly, but there was no significant difference in body length. HE staining was used to observe the pathological changes of BPD in the lung tissue. The above results proved that under the hyperoxia condition, the BPD animal model of premature infants was successfully established, which provided a new choice for the future research of BPD.

6.
Mol Cell Probes ; 51: 101529, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32036037

RESUMO

Bronchopulmonary dysplasia (BPD) is a chronic lung disease with long-term sequelae including neurodevelopmental delay. Although the precise mechanism of BPD is not well defined, oxidative stress is thought to be involved in the pathogenesis process of BPD. Nrf2 (Nuclear factor erythroid 2-related factor 2)-Keap1 (Kelch-like ECH associated protein 1)-ARE (Antioxidant Reaction Elements) signaling pathway is one of the main protective mechanisms of BPD, which can induce cytoprotective gene expression, such as heme oxygenase-1 (HO-1), nicotinamide quinone oxidoreductase 1 (NQO1) and so on. We exposed premature rats to hyperoxia and identified lung developmental retardation in preterm rats, with similar pathological changes as BPD. The expression of Nrf2 and HO-1 in premature rats was significantly higher after hyperoxia exposure. To explore the changes of Nrf2 and HO-1 in premature rats and enhance their beneficial functions may provide new treatment strategies for infants at risk of BPD.


Assuntos
Displasia Broncopulmonar/metabolismo , Heme Oxigenase-1/metabolismo , Pulmão/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Animais , Elementos de Resposta Antioxidante/genética , Displasia Broncopulmonar/complicações , Displasia Broncopulmonar/genética , Displasia Broncopulmonar/patologia , Feminino , Heme Oxigenase-1/genética , Humanos , Hiperóxia/complicações , Recém-Nascido , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Pulmão/enzimologia , Pulmão/patologia , Masculino , NAD(P)H Desidrogenase (Quinona)/metabolismo , Fator 2 Relacionado a NF-E2/genética , Ratos , Ratos Sprague-Dawley , Fatores de Risco , Transdução de Sinais/genética
7.
J Cell Mol Med ; 24(1): 965-972, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31713992

RESUMO

Bronchopulmonary dysplasia (BPD) is a chronic lung disease that affects the quality of life of infants. At present, premature exposure to hyperoxia for extended periods of time is believed to affect the development of lung tissue and vascularity, resulting in BPD. The oxidative stress caused by hyperoxia exposure is an important risk factor for BPD in premature infants. Nuclear factor E2-related factor 2 (Nrf2) is an important regulator of antioxidant mechanisms. As a microRNA, microRNA-125b (miR-125b) plays an important role in cell proliferation, differentiation and apoptosis. Although the Nrf2/ARE pathway has been extensively studied, little is known about the regulatory role of microRNAs in Nrf2 expression. In this study, the expression levels of Nrf2 and miR-125b in the lung tissues of premature Sprague Dawley (SD) rats and A549 cells exposed to hyperoxia were detected by quantitative real-time polymerase chain reaction (qRT-PCR), and the apoptosis of A549 cells was detected by flow cytometry. The results showed that Nrf2 and miRNA-125b in the lung tissues of premature rats increased significantly upon exposure to hyperoxia and played a protective role. Nrf2 was suppressed by small interfering RNA (siRNA) in A549 cells, miR-125b was similarly inhibited, and apoptosis was significantly increased. These results suggest that miR-125b helps protect against BPD as a downstream target of Nrf2.


Assuntos
Apoptose , Hiperóxia/fisiopatologia , Neoplasias Pulmonares/patologia , Pulmão/patologia , MicroRNAs/genética , Fator 2 Relacionado a NF-E2/antagonistas & inibidores , Estresse Oxidativo , Células A549 , Animais , Animais Recém-Nascidos , Diferenciação Celular , Proliferação de Células , Feminino , Humanos , Pulmão/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Masculino , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , RNA Interferente Pequeno/genética , Ratos , Ratos Sprague-Dawley
8.
Zhongguo Dang Dai Er Ke Za Zhi ; 21(6): 594-600, 2019 Jun.
Artigo em Chinês | MEDLINE | ID: mdl-31208516

RESUMO

OBJECTIVE: To study the effect of hyperoxic exposure on the dynamic expression of heme oxygenase-1 (HO-1) and glutamate-L-cysteine ligase catalytic subunit (GCLC) in the lung tissue of preterm neonatal rats. METHODS: Cesarean section was performed for rats on day 21 of gestation to obtain 80 preterm rats, which were randomly divided into air group and hyperoxia group after one day of feeding. The rats in the air group were housed in room air under atmospheric pressure, and those in the hyperoxia group were placed in an atmospheric oxygen tank (oxygen concentration 85%-95%) in the same room. Eight rats each were selected from each group on days 1, 4, 7, 10, and 14, and lung tissue samples were collected. Hematoxylin and eosin staining was used to observe the pathological changes of lung tissue at different time points after air or hyperoxic exposure. Western blot and RT-qPCR were used to measure the protein and mRNA expression of HO-1 and GCLC in the lung tissue of preterm rats at different time points after air or hyperoxic exposure. RESULTS: Compared with the air group, the hyperoxia group had a significant reduction in the body weight (P<0.05). Compared with the air group, the hyperoxia group had structural disorder, widening of alveolar septa, a reduction in the number of alveoli, and simplification of the alveoli on the pathological section of lung tissue. Compared with the air group, the hyperoxia group had significantly lower relative mRNA expression of HO-1 in the lung tissue on day 7 and significantly higher expression on days 10 and 14 (P<0.05). Compared with the air group, the hyperoxia group had significantly lower mRNA expression of GCLC in the lung tissue on days 1, 4, and 7 and significantly higher expression on day 10 (P<0.05). Compared with the air group, the hyperoxia group had significantly higher protein expression of HO-1 in the lung tissue on all days, and the protein expression of GCLC had same results as HO-1, except on day 1 (P<0.05). CONCLUSIONS: Hyperoxia exposure may lead to growth retardation and lung developmental retardation in preterm rats. Changes in the protein and mRNA expression of HO-1 and GCLC in the lung tissue of preterm rats may be associated with the pathogenesis of hyperoxia-induced lung injury in preterm rats.


Assuntos
Hiperóxia , Animais , Animais Recém-Nascidos , Domínio Catalítico , Cesárea , Cisteína , Feminino , Glutamatos , Heme Oxigenase-1 , Humanos , Recém-Nascido , Pulmão , Gravidez , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA