Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 104
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Mol Med ; 29(1): 70, 2023 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-37226090

RESUMO

BACKGROUND: Bronchopulmonary dysplasia (BPD) is the most common and serious chronic lung disease in preterm infants with pathological characteristics of arrested lung development. DNA double-strand breaks (DSBs) are a serious manifestation of oxidative stress damage, but little is known about the role of DSBs in BPD. The current study set out to detect DSB accumulation and cell cycle arrest in BPD and study the expression of genes related to DNA damage and repair in BPD through DNA damage signaling pathway-based PCR array to determine a suitable target to improve arrested lung development associated with BPD. METHODS: DSB accumulation and cell cycle arrest were detected in a BPD animal model and primary cells, then a DNA damage signaling pathway-based PCR array was used to identify the target of DSB repair in BPD. RESULTS: DSB accumulation and cell cycle arrest were shown in BPD animal model, primary type II alveolar epithelial cells (AECII) and cultured cells after exposure to hyperoxia. Of the 84 genes in the DNA damage-signaling pathway PCR array, eight genes were overexpressed and 11 genes were repressed. Rad1, an important protein for DSB repair, was repressed in the model group. Real-time PCR and western blots were used to verify the microarray results. Next, we confirmed that silencing Rad1 expression aggravated the accumulation of DSBs and cell cycle arrest in AECII cells, whereas its overexpression alleviated DSB accumulation and cell cycle arrest. CONCLUSIONS: The accumulation of DSBs in AECII might be an important cause of alveolar growth arrest associated with BPD. Rad1 could be an effective target for intervention to improve this arrest in lung development associated with BPD.


Assuntos
Displasia Broncopulmonar , Exonucleases , Parada Cardíaca , Animais , Ratos , Células Epiteliais Alveolares , Displasia Broncopulmonar/genética , Pontos de Checagem do Ciclo Celular/genética , DNA , Quebras de DNA de Cadeia Dupla
2.
Pediatr Res ; 92(6): 1543-1554, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35220399

RESUMO

BACKGROUND: Reducing neuroinflammatory damage is an effective strategy for treating white-matter damage (WMD) in premature infants. Caffeine can ameliorate hypoxia-ischemia-induced brain WMD; however, its neuroprotective effect and mechanism against hypoxic-ischemic WMD remain unclear. METHODS: We used 3-day-old Sprague-Dawley rats to establish a model of cerebral hypoxia-ischemia-induced brain WMD after unilateral common carotid artery ligation and hypoxia exposure (8% O2 + 92% N2) for 2.5 h. Mechanism experiments were conducted to detect M1/M2 polarization and activation of microglia and NLRP3 inflammasome. RESULTS: Caffeine inhibited NLRP3 inflammasome activation, reduced microglial Iba-1 activation, inhibited microglia M1 polarization, and promoted microglia M2 polarization by downregulating CD86 and iNOS protein expression, inhibiting the transcription of the proinflammatory TNF-α and IL-1ß, upregulating CD206 and Arg-1 expression, and promoting the transcription of the anti-inflammatory factors IL-10 and TGF-ß. Importantly, we found that these caffeine-mediated effects could be reversed after inhibiting A2aR activity. CONCLUSIONS: Caffeine improved long-term cognitive function in neonatal rats with hypoxic-ischemic WMD via A2aR-mediated inhibition of NLRP3 inflammasome activation, reduction of microglial activation, regulation of the phenotypic polarization of microglia and the release of inflammatory factors, and improvement of myelination development. IMPACT: The direct protective effect of caffeine on hypoxic-ischemic white-matter damage (WMD) and its mechanism remains unclear. This study elucidated this mechanism using neonatal rats as an animal model of hypoxia-ischemia-induced cerebral WMD. The findings demonstrated caffeine as a promising therapeutic tool against immature WMD to protect neonatal cognitive function. We found that caffeine pretreatment reduced WMD in immature brains via regulation of microglial activation and polarization by adenosine A2a receptor, thereby, providing a scientific basis for future clinical application of caffeine.


Assuntos
Hipóxia-Isquemia Encefálica , Inflamassomos , Ratos , Animais , Animais Recém-Nascidos , Ratos Sprague-Dawley , Inflamassomos/metabolismo , Microglia/metabolismo , Cafeína/farmacologia , Cafeína/uso terapêutico , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Hipóxia-Isquemia Encefálica/metabolismo , Isquemia
3.
Pediatr Res ; 83(5): 1024-1030, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29281615

RESUMO

BackgroundThe toll-like receptor 4 (TLR4) has been reported to play an important role in necrotizing enterocolitis (NEC). As an established regulator of TLR4, vitamin D has been demonstrated to be intestinal-protective. This study aims at finding out whether the vitamin D/vitamin D receptor (VDR) pathway ameliorates NEC by regulating TLR4.MethodsSerum 25-hydrovitamin D (25(OH)D) was tested and compared in 15 preterm infants with NEC, 12 preterm infants without known complications and 20 healthy term infants. Neonatal Wistar rats were grouped and NEC was induced through formula feeding and cold/asphyxia stress. Vitamin D and the vehicle were administered to compare the microscopic structure, apoptotic protein expression, intestinal barrier function, inflammatory response, and TLR4 expression.ResultsPreterm infants with NEC had significantly lower 25(OH)D levels than those without NEC and healthy subjects. VDR expression was suppressed, whereas TLR4 expression was elevated in the NEC intestine. Vitamin D may increase the survival rate, alleviate structure damage, and preserve intestinal barrier function. These were achieved partly through restoration of VDR and suppression of TLR4.ConclusionNEC infants have lower levels of vitamin D. The vitamin D/VDR pathway protects against intestinal injury of NEC partly through suppressing the expression of TLR4.


Assuntos
Enterocolite Necrosante/terapia , Receptores de Calcitriol/metabolismo , Receptor 4 Toll-Like/metabolismo , Vitamina D/uso terapêutico , Animais , Apoptose , Modelos Animais de Doenças , Enterocolite Necrosante/metabolismo , Feminino , Regulação da Expressão Gênica , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Inflamação , Mucosa Intestinal/metabolismo , Intestinos/fisiopatologia , Masculino , Permeabilidade , Ratos , Ratos Wistar , Transdução de Sinais
4.
Health Econ ; 26(3): 292-304, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-26676999

RESUMO

We provide evidence for the causal impact of social status on longevity by exploiting a natural experiment in which subjects undergo a shift in their social status without considerable economic impact. We gather data on 4190 scientists who were either nominated for or successfully elected to the Chinese Academy of Science or of Engineering. Being elected as an academician in China is a boost in social status (vice-ministerial level) with negligible direct economic impact (US$30 monthly before 2009). After correcting for two sources of bias, (1) some potential academicians decease too young to be elected, leading to selection bias in favor of academicians and (2) the endogenous relationship between health and social status, we find that the enhanced social status of becoming an academician leads to approximately 1.2 years longer life. Copyright © 2015 John Wiley & Sons, Ltd.


Assuntos
Comitês Consultivos/estatística & dados numéricos , Longevidade , Classe Social , Comitês Consultivos/tendências , China , Países em Desenvolvimento , Nível de Saúde , Humanos , Pessoa de Meia-Idade , Política
5.
BMC Public Health ; 17(1): 863, 2017 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-29110657

RESUMO

BACKGROUND: Although social capital as a key determinant of health has been well established in various studies, little is known about how lifestyle factors mediate this relationship. Understanding the cross-relationships between social capital, health, and lifestyle factors is important if health promotion policies are to be effective. The purpose of this study is to explore whether different dimensions of social capital and lifestyle factors are related, and whether lifestyle factors mediate the association between social capital and self-rated health (SRH) and psychological well-being (PWB) in China. METHODS: This study used nationally representative data from the 2014 China Family Panel Studies (n = 28,916). The data reported on three dimensions of individual-level social capital: social trust, social relationship and Chinese Communist Party (CCP) membership. Health was assessed using SRH and PWB. Five lifestyle indicators were recorded: healthy diet, physical activity, smoking, sleeping, and non-overweight status. Logistic regression was used to examine the associations between social capital and lifestyle factors, and whether there was a mediating role of lifestyle. Odds ratios relating health status to social capital were reported before and after adjustment for lifestyle factors. Mediation analysis was then used to calculate the total, direct and indirect effects of social capital on SRH and PWB. RESULTS: The results show that social trust was significantly associated with all five lifestyle factors. Social relationship was significantly associated with four of the five: healthy diet, physical activity, sleeping and non-overweight. CCP membership was only significantly associated with two lifestyle factors: physical activity and non-overweight. Social trust and social relationship were significantly related to both SRH and PWB. CCP membership was only significantly related to SRH. Mediation analysis found modest evidence that lifestyle factors influenced the relationship between all three types of social capital and SRH. In contrast, only social trust and social relationship, but not CCP membership, were mediated by lifestyle factors with respect to PWB. CONCLUSIONS: This study is the first to explore the mediating role of lifestyle factors in the relationship between social capital and health in China. The overall findings suggest that lifestyle factors modestly mediate the association between social capital and health. The degree of mediating effect varies across different dimensions of social capital. Social capital-based health promotion policies would benefit from taking lifestyle factors into account.


Assuntos
Nível de Saúde , Estilo de Vida , Capital Social , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , China , Estudos Transversais , Feminino , Humanos , Modelos Logísticos , Masculino , Pessoa de Meia-Idade , Adulto Jovem
6.
Respir Res ; 17(1): 50, 2016 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-27176222

RESUMO

BACKGROUND: Bronchopulmonary dysplasia (BPD) is a common complication in preterm infants that involves the downregulation of tight junction (TJ) proteins. However, the mechanism underlying downregulation of the expression of TJ proteins during at the early stages of hyperoxia-induced BPD remains to be understood. Here, we aimed to identify the role of caveolin-1 (Cav-1) in hyperoxia-induced pulmonary epithelial barrier breakdown. METHODS: First, we established an in vitro pulmonary epithelial barrier models using primary type II alveolar epithelial cells (AEC-II) from newborn rats. AEC-II was assigned to the hyperoxic (85 % O2/5 % CO2) or normoxic (21 % O2/5 % CO2) groups. Second, AEC-II was transfected with Cav-1-siRNA to downregulate Cav-1 under normoxic exposure. Third, AEC-II was transfected with a cDNA encoding Cav-1 to upregulate Cav-1 expression under hyperoxic exposure. Then, expression levels of Cav-1 and TJ proteins were examined by immunofluorescence staining, reverse transcription-polymerase chain reaction, and Western blotting. The TJ structures visualized using a transmission electron microscope, and transepithelial resistance and apparent permeability coefficient of fluorescein isothiocyanate-dextran, which are indicators of barrier function, were measured. RESULTS: Our data showed that exposure to hyperoxia disrupted the structure and function of the pulmonary epithelial barrier and decreased the ZO-1, occludin, claudin-4, and Cav-1 expression levels. Moreover, Cav-1 knockdown attenuated the expression of the other three genes and disrupted pulmonary epithelial barrier structure and function under normoxic exposure. However, Cav-1 upregulation markedly antagonized the hyperoxia-induced pulmonary epithelial barrier destruction and TJ protein loss. CONCLUSIONS: This is the first study to present evidence illustrating the novel role of Cav-1 downregulation-mediated TJ protein loss in pulmonary epithelial barrier destruction during BPD.


Assuntos
Células Epiteliais Alveolares/metabolismo , Displasia Broncopulmonar/metabolismo , Caveolina 1/metabolismo , Hiperóxia/metabolismo , Oxigênio/metabolismo , Edema Pulmonar/metabolismo , Junções Íntimas/metabolismo , Células Epiteliais Alveolares/ultraestrutura , Animais , Animais Recém-Nascidos , Displasia Broncopulmonar/genética , Displasia Broncopulmonar/patologia , Caveolina 1/genética , Células Cultivadas , Claudina-4/genética , Claudina-4/metabolismo , Impedância Elétrica , Regulação da Expressão Gênica , Hiperóxia/genética , Hiperóxia/patologia , Ocludina/genética , Ocludina/metabolismo , Permeabilidade , Edema Pulmonar/genética , Edema Pulmonar/patologia , Interferência de RNA , Ratos Wistar , Transdução de Sinais , Junções Íntimas/ultraestrutura , Fatores de Tempo , Transfecção , Proteína da Zônula de Oclusão-1/genética , Proteína da Zônula de Oclusão-1/metabolismo
7.
Am J Physiol Lung Cell Mol Physiol ; 308(9): L861-72, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25681436

RESUMO

Supplemental oxygen treatment in preterm infants may cause bronchopulmonary dysplasia (BPD), which is characterized by alveolar simplification and vascular disorganization. Despite type II alveolar epithelial cell (AEC II) damage being reported previously, we found no decrease in the AEC II-specific marker, surfactant protein C (SP-C), in the BPD model in our previous study. We thus speculated that AEC II injury is not a unique mechanism of BPD-related pulmonary epithelial repair dysfunction and that abnormal transdifferentiation can exist. Newborn rats were randomly assigned to model (85% oxygen inhalation) and control groups (room air inhalation). Expressions of AEC I (aquaporin 5, T1α) and AEC II markers (SP-C, SP-B) were detected at three levels: 1) in intact lung tissue, 2) in AEC II isolated from rats in the two groups, and 3) in AEC II isolated from newborn rats, which were further cultured under either hyperoxic or normoxic conditions. In the model group, increased AEC I was observed at both the tissue and cell level, and markedly increased transdifferentiation was observed by immunofluorescent double staining. Transmission electron microscopy revealed morphological changes in alveolar epithelium such as damaged AECs, a fused air-blood barrier structure, and opened tight junctions in the model group. These findings indicate that transdifferentiation of AECs is not suppressed but rather is increased under hyperoxic treatment by compensation; however, such repair during injury cannot offset pulmonary epithelial air exchange and barrier dysfunction caused by structural damage to AECs.


Assuntos
Transdiferenciação Celular/fisiologia , Células Epiteliais/citologia , Oxigênio/farmacologia , Alvéolos Pulmonares/citologia , Mucosa Respiratória/citologia , Animais , Apoptose , Aquaporina 5/biossíntese , Barreira Alveolocapilar , Displasia Broncopulmonar/etiologia , Proliferação de Células , Células Cultivadas , Hiperóxia , Glicoproteínas de Membrana/biossíntese , Oxigênio/administração & dosagem , Peptídeos/metabolismo , Alvéolos Pulmonares/metabolismo , Proteína B Associada a Surfactante Pulmonar/metabolismo , Distribuição Aleatória , Ratos , Ratos Wistar
8.
Respir Res ; 16: 75, 2015 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-26104385

RESUMO

BACKGROUND: Bronchopulmonary dysplasia (BPD) in premature infants is a predominantly secondary occurrence to intrauterine inflammation/infection and postpartum mechanical ventilation; in recent years, an association with epigenetics has also been found. DNA methylation, catalyzed by DNA methyl transferases (DNMTs), and tri-methylation of lysine 27 on histone H3 (H3K27me3), mediated by the methyltransferase, Enhancer of Zeste Homolog 2 (EZH2), are some of the most commonly found modifications in epigenetics. Runt-related transcription factor 3 (RUNX3) is associated with pulmonary epithelial and vascular development and regulates expression at the post-transcriptional level by DNA methylation through DNMT1 or DNMT3b. However, the involvements of these epigenetic factors in the occurrence of BPD are, as yet, unclear. METHODS: Newborn rats were randomly assigned to a model, hyperoxia (85 % O2) or control, normoxia group (21 % O2). Lung tissues and alveolar type 2 (AT2) epithelial cells were collected between 1-14 days. The expression of DNMTs, and EZH2 was detected by immunohistochemistry, Western blot and real-time PCR. The percentage of DNA methylation and H3K27me3 levels in the RUNX3 promoter region was measured by bisulfite sequencing PCR and chromatin immunoprecipitation assay. RUNX3 protein and mRNA expression in AT2 cells was also measured after inhibition using the DNA methylation inhibitor, 5-Aza-2'-deoxycytidine, the H3K27me3 inhibitor, JMJD3, and the EZH2 inhibitor, DZNep. RESULTS: Compared with the control group, RUNX3 protein was downregulated and DNMT3b and EZH2 were highly expressed in lung tissues and AT2 cells of the model group (P < 0.05), while high DNA methylation and H3K27me3 modifications were present in the RUNX3 promoter region, in lung tissues of the model group (P < 0.05). Following hyperoxia in the model group, JMJD3 and DZNep significantly reversed the hyperoxia-induced down-regulation of RUNX3 expression in AT2 cells (P < 0.05), more so than 5-Aza-2'-deoxycytidine (P < 0.05). CONCLUSIONS: 1) DNA methylation and H3K27 trimethylation are present in the BPD model; 2) RUNX3 down-regulation is attributed to both DNMT3b-catalyzed DNA methylation and EZH2-catalyzed histone methylation.


Assuntos
Displasia Broncopulmonar/metabolismo , Subunidade alfa 3 de Fator de Ligação ao Core/biossíntese , Metilação de DNA/fisiologia , Modelos Animais de Doenças , Hiperóxia/metabolismo , Animais , Animais Recém-Nascidos , Displasia Broncopulmonar/patologia , Subunidade alfa 3 de Fator de Ligação ao Core/antagonistas & inibidores , Hiperóxia/patologia , Ratos , Ratos Sprague-Dawley
9.
Mol Cell Biochem ; 408(1-2): 181-9, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26122393

RESUMO

Surfactant protein C (SP-C) deficiency is a risk factor for hyperoxia-induced bronchopulmonary dysplasia in newborn infants. However, the role of SP-C deficiency in the process is unclear. Here, using neonatal rat BPD model and MLE-12, mouse alveolar epithelial type II cell, we examined the changes of SP-C levels during hyperoxia. Immunohistochemistry, immunofluorescence, and ELISA analysis showed SP-C accumulation in alveolar epithelial type II cells. Electron microscopy further demonstrated the accumulation of lamellar bodies and the co-localization of lamellar bodies with autophagosomes in the cytoplasm of alveolar epithelial type II cells. The inhibition of autophagy with 3-Methyladenine and knockdown of Atg7 abolished hyperoxia-induced SP-C accumulation in the cytoplasm. Furthermore, inhibition of JNK signaling with SP600125 suppressed hyperoxia-induced Atg7 expression and SP-C accumulation. These findings suggest that hyperoxia triggers autophagy via JNK signaling-mediated Atg7 expression, which promotes the accumulation of SP-C within alveolar epithelial type II cells. Our data provide a potential approach for hyperoxic lung injury therapy by targeted pharmacological inhibition of autophagic pathway.


Assuntos
Células Epiteliais/citologia , Sistema de Sinalização das MAP Quinases , Alvéolos Pulmonares/metabolismo , Proteína C Associada a Surfactante Pulmonar/metabolismo , Enzimas Ativadoras de Ubiquitina/metabolismo , Animais , Animais Recém-Nascidos , Autofagia , Hipóxia Celular , Linhagem Celular , Camundongos , Alvéolos Pulmonares/citologia , Ratos
10.
Cell Biochem Funct ; 33(1): 44-9, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25515701

RESUMO

Prolonged exposure to hyperoxia leads to acute lung injury. Alveolar type II cells are main target of hyperoxia-induced lung injury. However, the cellular and molecular mechanisms remain unknown. Here, we aimed to investigate the role of placental growth factor (PLGF) in hyperoxia-induced lung injury. Using experimental hyperoxia-induced lung injury model of neonatal rat and mouse lung epithelial type II cells (MLE-12), we examined the levels of PLGF in bronchoalveolar lavage fluid and in the supernatants of MLE-12 cells. Our results revealed that exogenous PLGF induced hyperoxia-induced lung injury. Furthermore, PLGF triggered a shift of vinculin from insoluble to soluble cell fraction, similar to the observation under hyperoxia stimulation. Moreover, we observed significantly reduced phosphorylation of focal adhesion kinase and increased permeability in MLE-12 cells treated with PLGF. These results suggest that PLGF triggers focal adhesion disassembly in alveolar type II cells via inhibiting the activation of focal adhesion kinase. Our findings reveal a novel role of PLGF in hyperoxia-induced lung injury and provide a potential target for the management of hyperoxia-induced acute lung injury.


Assuntos
Lesão Pulmonar Aguda/metabolismo , Modelos Animais de Doenças , Hiperóxia/metabolismo , Pulmão/metabolismo , Proteínas da Gravidez/metabolismo , Animais , Animais Recém-Nascidos , Líquido da Lavagem Broncoalveolar , Células Epiteliais , Quinase 1 de Adesão Focal/metabolismo , Adesões Focais/metabolismo , Pulmão/patologia , Camundongos , Permeabilidade , Fator de Crescimento Placentário , Ratos
11.
Exp Lung Res ; 39(10): 417-26, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24298937

RESUMO

Following preterm birth, levels of Krebs von den Lungen-6/mucin 1 (KL-6/MUC1) in serum correlate closely with the development of advanced bronchopulmonary dysplasia (BPD), but the role of KL-6/MUC1 in the development of BPD is unclear. To explore whether a relationship exists between KL-6/MUC1 and pathological changes in BPD and verify such a clinical finding, we established a newborn rat model of 95% oxygen-induced BPD. The development of pulmonary alveoli was evaluated by determining the radial alveolar count (RAC) and examining the location, distribution, and expression of KL-6/MUC1 in pulmonary tissues using a fluorescent immunoassay, Western blot, and reverse transcription polymerase chain reaction. The synchronic expression levels of KL-6/MUC1 in serum, bronchoalveolar lavage fluid (BALF) and pulmonary tissues were examined using an enzyme-linked immunosorbent assay. The mean RAC in the hyperoxia group was significantly lower than in normoxia controls, whereas the expression levels of KL-6/MUC1 were higher. On days 1, 3, 7, and 14, the mean RACs in hyperoxic rats were 15.00, 12.67, 12.00, and 11.33, respectively. The expression levels of KL-6/MUC1 peaked in the experimental group on day 1, and began to decrease slightly after day 3. The expression levels of KL-6/MUC1 in serum and BALF were associated with KL-6/MUC1 expression in pulmonary tissues. We suggest that increased lung KL-6/MUC1 expression appears to be closely associated with impairment of alveolarization in a newborn rat model of hyperoxia-induced BPD. Changes in lung KL-6/MUC1 expression can be evaluated effectively and less invasively by monitoring KL-6/MUC1 in serum and BALF.


Assuntos
Displasia Broncopulmonar/etiologia , Hiperóxia/complicações , Pulmão/metabolismo , Pulmão/patologia , Mucina-1/metabolismo , Animais , Animais Recém-Nascidos , Líquido da Lavagem Broncoalveolar/química , Displasia Broncopulmonar/metabolismo , Displasia Broncopulmonar/patologia , Modelos Animais de Doenças , Expressão Gênica , Mucina-1/sangue , Mucina-1/genética , Alvéolos Pulmonares/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Distribuição Tecidual
12.
J Immunol Res ; 2023: 5149306, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37636861

RESUMO

White matter damage (WMD) is a primary cause of cerebral palsy and cognitive impairment in preterm infants, and no effective treatments are available. Microglia are a major component of the innate immune system. When activated, they form typical pro-inflammatory (M1) and anti-inflammatory (M2) phenotypes and regulate myelin development and synapse formation. Therefore, they may play a pivotal role in hypoxic-ischemic (HI) WMD. Herein, we investigated neural inflammation and long-term microglia phenotypic polarization in a neonatal rat model of hypoxia-ischemia-induced WMD and elucidated the underlying pathophysiological processes. We exposed 3-day-old (P3) Sprague-Dawley rats to hypoxia (8% oxygen) for 2.5 hr after unilateral common carotid artery ligation. The activation of NLRP3 inflammatory bodies, microglia M1/M2 polarization, myelination, and synaptic development in our model were monitored 7, 14, and 21 days after birth. In addition, the Morris water maze test was performed on postnatal Day 28. We confirmed myelination disturbance in the periventricular white matter, abnormal synaptic development, and behavioral changes in the periventricular area during the development of HI WMD. In addition, we found an association between the occurrence and development of HI WMD and activation of the NLRP3 inflammasome, microglial M1/M2 polarization, and the release of inflammatory factors. NLRP3 inhibition can play an anti-inflammatory role by inhibiting the differentiation of microglia into the M1 phenotype, thereby improving myelination and synapse formation. In conclusion, microglia are key mediators of the inflammatory response and exhibit continuous phenotypic polarization 7-21 days after HI-induced WMD. This finding can potentially lead to a new treatment regimen targeting the phenotypic polarization of microglia early after HI-induced brain injury.


Assuntos
Nascimento Prematuro , Substância Branca , Animais , Feminino , Ratos , Hipóxia , Inflamassomos , Isquemia , Microglia , Doenças Neuroinflamatórias , Proteína 3 que Contém Domínio de Pirina da Família NLR , Ratos Sprague-Dawley , Animais Recém-Nascidos
13.
Respir Res ; 13: 36, 2012 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-22559818

RESUMO

BACKGROUND: Prolonged exposure to hyperoxia in neonates can cause hyperoxic acute lung injury (HALI), which is characterized by increased pulmonary permeability and diffuse infiltration of various inflammatory cells. Disruption of the epithelial barrier may lead to altered pulmonary permeability and maintenance of barrier properties requires intact epithelial tight junctions (TJs). However, in neonatal animals, relatively little is known about how the TJ proteins are expressed in the pulmonary epithelium, including whether expression of TJ proteins is regulated in response to hyperoxia exposure. This study determines whether changes in tight junctions play an important role in disruption of the pulmonary epithelial barrier during hyperoxic acute lung injury. METHODS: Newborn rats, randomly divided into two groups, were exposed to hyperoxia (95% oxygen) or normoxia for 1-7 days, and the severity of lung injury was assessed; location and expression of key tight junction protein occludin and ZO-1 were examined by immunofluorescence staining and immunobloting; messenger RNA in lung tissue was studied by RT-PCR; transmission electron microscopy study was performed for the detection of tight junction morphology. RESULTS: We found that different durations of hyperoxia exposure caused different degrees of lung injury in newborn rats. Treatment with hyperoxia for prolonged duration contributed to more serious lung injury, which was characterized by increased wet-to-dry ratio, extravascular lung water content, and bronchoalveolar lavage fluid (BALF):serum FD4 ratio. Transmission electron microscopy study demonstrated that hyperoxia destroyed the structure of tight junctions and prolonged hyperoxia exposure, enhancing the structure destruction. The results were compatible with pathohistologic findings. We found that hyperoxia markedly disrupted the membrane localization and downregulated the cytoplasm expression of the key tight junction proteins occludin and ZO-1 in the alveolar epithelium by immunofluorescence. The changes of messenger RNA and protein expression of occludin and ZO-1 in lung tissue detected by RT-PCR and immunoblotting were consistent with the degree of lung injury. CONCLUSIONS: These data suggest that the disruption of the pulmonary epithelial barrier induced by hyperoxia is, at least in part, due to massive deterioration in the expression and localization of key TJ proteins.


Assuntos
Hiperóxia/metabolismo , Ocludina/biossíntese , Alvéolos Pulmonares/metabolismo , Mucosa Respiratória/metabolismo , Proteína da Zônula de Oclusão-1/biossíntese , Animais , Animais Recém-Nascidos , Feminino , Hiperóxia/patologia , Gravidez , Alvéolos Pulmonares/patologia , Distribuição Aleatória , Ratos , Ratos Wistar , Mucosa Respiratória/patologia , Junções Íntimas/metabolismo , Junções Íntimas/patologia
14.
Pediatr Int ; 54(6): 735-42, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22591391

RESUMO

BACKGROUND: The aim of the present study was to investigate the expression and activity of epithelial sodium channel (ENaC) in hyperoxia-induced bronchopulmonary dysplasia (BPD) in neonatal rats. METHODS: Neonatal rats were exposed to hyperoxia to establish BPD models (control group was exposed to air), lung water was measured and Western blot was applied to detect the expression of three homologous subunits: α-, ß- and γ-ENaC in the lung tissues. Furthermore, ATII cells were isolated from neonatal rats, and primarily cultured under normoxic or hyperoxic conditions. The ENaC expression was also examined in these cells. In addition, the amiloride-sensitive Na(+) currents induced by hyperoxia were recorded using the whole-cell patch clamp technique. RESULTS: The α-ENaC expression was increased after 5 days of hyperoxia in rat lung tissues, whereas not after 1, 3 and 7 days. ATII cells showed α-ENaC expression was reduced after 1 and 2 days' hyperoxia, but no change after 3 days. In contrast, ß- and γ-ENaC expression was increased after hyperoxia in both in vivo and in vitro experiments. The amiloride-sensitive Na(+) currents in hyperoxia-exposed ATII cells were also increased, which was consistent with the upregulated expression of ß- and γ-ENaC. CONCLUSION: Hyperoxia upregulates the expression of ENaC, especially ß- and γ-ENaC subunits, in both neonatal rat lung tissues and ATII cells. Hyperoxia also enhanced the activity of ENaC in neonatal rat ATII cells. Dysfunctional transport of Na(+) may not be a key factor involving pulmonary edema at the early stage of BPD.


Assuntos
Displasia Broncopulmonar/metabolismo , Canais Epiteliais de Sódio/biossíntese , Hiperóxia/complicações , Prenhez , Alvéolos Pulmonares/metabolismo , Animais , Animais Recém-Nascidos , Transporte Biológico , Western Blotting , Displasia Broncopulmonar/etiologia , Displasia Broncopulmonar/patologia , Células Cultivadas , Modelos Animais de Doenças , Feminino , Hiperóxia/metabolismo , Hiperóxia/patologia , Masculino , Técnicas de Patch-Clamp , Gravidez , Alvéolos Pulmonares/patologia , Ratos , Ratos Wistar
15.
Fetal Pediatr Pathol ; 31(1): 11-20, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22233504

RESUMO

The purpose of this investigation is to research the expression and effect of HoxB5 during pulmonary injury and to investigate the repairing ability of alveolar epithelial cells in such processes. Eighty neonatal rats were randomly divided into two groups: a group of high concentration of oxygen and the control group. The high oxygen group would inhale 85 to 90% oxygen and the control group would inhale air. The lung tissues on the 1(st), 3(rd), 7(th), 14(th), and 21(st) days would be obtained, in which immunohistochemical assay and Reverse Transcription Polymerase Chain Reaction (RT-PCR) would be performed to test the expressions of proteins and mRNAs of surfactant protein C (SPC) and AQP5. For expression of HoxB5 protein and its mRNA, immunohistochemical assay, western blot, in-situ hybridization, and RT-PCR would be run. The expression of SPC in the group of high concentration of oxygen was significantly reduced on day 3. Its expressions on day 14 and day 21 were significantly higher than those of the control group (p < 0.05). The expression of AQP5 in the group of high concentration of oxygen progressively decreased and such difference with the control group was significant (p < 0.05). The four experimental methods all showed the expression of HoxB5 in the group with high concentration of oxygen gradually decreased since day 7 (p < 0.05). High concentration of oxygen is damaging to alveolar epithelial cells. Although the number of type II alveolar epithelial cells (AECII) increases, its ability to differentiate and transform is significantly reduced and the reduced expression level of HoxB5 is possibly the reason for AECII to lose differentiation function to AECI.


Assuntos
Regulação da Expressão Gênica/fisiologia , Proteínas de Homeodomínio/genética , Lesão Pulmonar/genética , Células Epiteliais Alveolares/efeitos dos fármacos , Células Epiteliais Alveolares/metabolismo , Células Epiteliais Alveolares/patologia , Animais , Animais Recém-Nascidos , Aquaporina 5/genética , Aquaporina 5/metabolismo , Western Blotting , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Proteínas de Homeodomínio/metabolismo , Hiperóxia/complicações , Hibridização In Situ , Exposição por Inalação , Lesão Pulmonar/etiologia , Lesão Pulmonar/metabolismo , Masculino , Oxigênio/efeitos adversos , Peptídeos/genética , Peptídeos/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
CNS Neurosci Ther ; 28(7): 1019-1032, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35393758

RESUMO

AIM: White matter damage (WMD) is the main cause of cerebral palsy and cognitive impairment in premature infants. Although caffeine has been shown to possess neuroprotective effects in neonatal rats with hypoxic-ischemic WMD, the mechanisms underlying these protective effects are unclear. Herein, proteins modulated by caffeine in neonatal rats with hypoxic-ischemic WMD were evaluated. METHODS: We identified differential proteins and performed functional enrichment analyses between the Sham, hypoxic-ischemic WMD (HI), and HI+caffeine-treated WMD (Caffeine) groups. Confirmed the changes and effect of proteins in animal models and determined cognitive impairment via water maze experiments. RESULTS: In paraventricular tissue, 47 differential proteins were identified between the Sham, HI, and Caffeine groups. Functional enrichment analyses showed that these proteins were related to myelination and axon formation. In particular, the myelin basic protein (MBP), proteolipid protein, myelin-associated glycoprotein precursor, and sirtiun 2 (SIRT2) levels were reduced in the hypoxic-ischemic WMD group, and this effect could be prevented by caffeine. Caffeine alleviated the hypoxic-ischemic WMD-induced cognitive impairment and improved MBP, synaptophysin, and postsynaptic density protein 95 protein levels after hypoxic-ischemic WMD by preventing the HI-induced downregulation of SIRT2; these effects were subsequently attenuated by the SIRT2 inhibitor AK-7. CONCLUSION: Caffeine may have clinical applications in the management of prophylactic hypoxic-ischemic WMD; its effects may be mediated by proteins related to myelin development and synapse formation through SIRT2.


Assuntos
Hipóxia-Isquemia Encefálica , Substância Branca , Animais , Animais Recém-Nascidos , Cafeína/farmacologia , Cafeína/uso terapêutico , Modelos Animais de Doenças , Humanos , Hipóxia , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Hipóxia-Isquemia Encefálica/metabolismo , Proteômica , Ratos , Ratos Sprague-Dawley , Sirtuína 2/metabolismo , Substância Branca/metabolismo
17.
Front Pediatr ; 10: 946747, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36440350

RESUMO

Background: Bronchopulmonary dysplasia (BPD) is the most common neonatal chronic lung disease. However, its exact molecular pathogenesis is not understood. We aimed to identify relevant gene modules that may play crucial roles in the occurrence and development of BPD by weighted gene co-expression network analysis (WGCNA). Methods: We used RNA-Seq data of BPD and healthy control rats from our previous studies, wherein data from 30 samples was collected at days 1, 3, 7, 10, and 14. Data for preprocessing analysis included 17,613 differentially expressed genes (DEGs) with false discovery rate <0.05. Results: We grouped the highly correlated genes into 13 modules, and constructed a network of mRNA gene associations, including the 150 most associated mRNA genes in each module. Lgals8, Srpra, Prtfdc1, and Thap11 were identified as the key hub genes. Enrichment analyses revealed Golgi vesicle transport, coated vesicle, actin-dependent ATPase activity and endoplasmic reticulum pathways associated with these genes involved in the pathological process of BPD in module. Conclusions: This is a study to analyze data obtained from BPD animal model at different time-points using WGCNA, to elucidate BPD-related susceptibility modules and disease-related genes.

18.
Zhongguo Dang Dai Er Ke Za Zhi ; 13(7): 581-5, 2011 Jul.
Artigo em Zh | MEDLINE | ID: mdl-21752328

RESUMO

OBJECTIVE: To study the expression of extracellular signal regulated protein kinase (ERK) 1/2 in lung tissues of newborn rats with chronic lung disease (CLD) caused by hyperoxia. METHODS: Forty-eight full-term newborn rats were randomly divided into two groups: hyperoxia and control. The two groups were exposed to a hyperoxic gas mixture (0.90 O(2)) for an induction of CLD and room air within 12 hrs after birth, respectively. The levels of ERK1/2 protein and mRNA in lung tissues were measured using immunohistochemistry, Western blot and real-time PCR methods on postnatal days 3, 7 and 14. The severity of pulmonary fibrosis was evaluated. RESULTS: The expression of p-ERK protein in lung tissues in the hyperoxia group was significantly higher than that in the control group on postnatal days 7 and 14 (P<0.01). There were no significant differences in the levels of total ERK1/2 protein and ERK1/2 mRNA. CONCLUSIONS: The activation of phosphorated ERK1/2 may lead to lung fibrosis caused by hyperoxia in newborn rats.


Assuntos
Hiperóxia/complicações , Pneumopatias/metabolismo , Pulmão/enzimologia , Proteína Quinase 1 Ativada por Mitógeno/análise , Proteína Quinase 3 Ativada por Mitógeno/análise , Animais , Animais Recém-Nascidos , Doença Crônica , Feminino , Pulmão/patologia , Pneumopatias/etiologia , Pneumopatias/patologia , Masculino , Fosforilação , Fibrose Pulmonar/etiologia , Ratos , Ratos Wistar
19.
Zhongguo Dang Dai Er Ke Za Zhi ; 13(1): 36-9, 2011 Jan.
Artigo em Zh | MEDLINE | ID: mdl-21251385

RESUMO

OBJECTIVE: To study the effects of transforming growth factor-ß1 (TGF-ß1) on the gene expression of connective tissue growth factor (CTGF) in cultured lung fibroblasts of embryonic rats in vitro. METHODS: Wistar rats of embryonic 19 days were used for primary culture of lung fibroblasts (LFs). The cells in the experimental group were treated by different concentrations (1, 5 or 10 ng/mL) and different durations (12, 24 or 48 hrs) of TGF-ß1 to stimulate the LFs. The cells in the control group were cultured in serum-free medium. RT-PCR method was applied to detect CTGF mRNA expression in LFs. RESULTS: Compared with the control group, the levels of CTGF mRNA in LFs in the experimental group increased significantly (P<0.05). CTGF mRNA expression gradually increased with increasing concentration and duration of TGF-ß1 treatment (P<0.05). CONCLUSIONS: TGF-ß1 can stimulate CTGF gene expression in LFs and increase CTGF gene expression in a dose-and time-dependent manner.


Assuntos
Fator de Crescimento do Tecido Conjuntivo/genética , Fibroblastos/metabolismo , Pulmão/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Animais , Feminino , Expressão Gênica/efeitos dos fármacos , Pulmão/citologia , Fibrose Pulmonar/etiologia , RNA Mensageiro/análise , Ratos , Ratos Wistar
20.
Front Pediatr ; 9: 633532, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33681108

RESUMO

Objective: The clinical symptoms of neonatal Prader-Willi syndrome (PWS) are not typical and are easy to miss. The aim of the study was to investigate the clinical features and genetic characteristics of seven cases of neonatal PWS from northern China, and to improve the understanding of PWS in neonates. Methods: We retrospectively analyzed seven infants diagnosed by methylation specific multiplex ligation probe amplification technology (MS-MLPA) in the Neonatology Unit of Shengjing Hospital of China Medical University from September 2016 to July 2020. Results: All seven cases involved full term or nearly full-term infants born to mothers without a history of abnormal pregnancy or delivery. Difficulty in feeding occurred immediately after birth in infants with decreased hypotonia. Five patients had characteristic craniofacial morphology, such as a prominent forehead, narrow face, almond-shaped eyes, small mouth, and downturned mouth. Further, three of the seven infants had patent ductus arteriosus (PDA). In addition, three neonates had hyperammonemia, hypoglycemia, and idiopathic edema, respectively. PWS could be effectively diagnosed and genotyped by MS-MLPA. Conclusion: Neonates with PWS have hypotonia and feeding difficulty. Characteristic facial features and genital hypoplasia are common in neonatal PWS. Infants with PWS may be predisposed to PDA, hypoglycemia, hyperammonemia, and edema.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA