Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 537
Filtrar
2.
Biomolecules ; 12(3)2022 03 11.
Artigo em Inglês | MEDLINE | ID: mdl-35327629

RESUMO

Within this review, sex-specific differences in alveolar epithelial functions are discussed with special focus on preterm infants and the respiratory disorders associated with premature birth. First, a short overview about fetal lung development, the challenges the lung faces during perinatal lung transition to air breathing and respiratory distress in preterm infants is given. Next, clinical observations concerning sex-specific differences in pulmonary morbidity of human preterm infants are noted. The second part discusses potential sex-specific causes of pulmonary complications, including pulmonary steroid receptors and local lung steroid metabolism. With regard to pulmonary steroid metabolism, it is important to highlight which steroidogenic enzymes are expressed at which stage during fetal lung development. Thereafter, we review the knowledge concerning sex-specific aspects of lung growth and maturation. Special focus is given to alveolar epithelial Na+ transport as a driver of perinatal lung transition and the sex differences that were noted in this process.


Assuntos
Síndrome do Desconforto Respiratório do Recém-Nascido , Feminino , Feto/metabolismo , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Pulmão/metabolismo , Masculino , Gravidez , Síndrome do Desconforto Respiratório do Recém-Nascido/etiologia , Síndrome do Desconforto Respiratório do Recém-Nascido/metabolismo , Caracteres Sexuais
3.
Pediatr Pulmonol ; 57(5): 1325-1330, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35170262

RESUMO

ABCA3 is a phospholipid transporter protein required for surfactant assembly in lamellar bodies of alveolar type II cells. Biallelic pathogenic ABCA3 variants cause severe neonatal respiratory distress syndrome or childhood interstitial lung disease. However, ABCA3 genotype alone does not explain the diversity in disease presentation, severity, and progression. Additionally, monoallelic ABCA3 variants have been reported in infants and children with ABCA3-deficient phenotypes. The effects of most ABCA3 variants identified in patients have not been characterized at the RNA level. ABCA3 allele-specific expression occurs in some cell types due to epigenetic regulation. We obtained lung tissue at transplant or autopsy from 16 infants and children with ABCA3 deficiency due to compound heterozygous ABCA3 variants for biologic characterization of the predicted effects of ABCA3 variants at the RNA level and determination of ABCA3 allele expression. We extracted DNA and RNA from frozen lung tissue and reverse-transcribed cDNA from mRNA. We performed Sanger sequencing to assess allele-specific expression by comparing the heights of variant nucleotide peaks in amplicons from genomic DNA and cDNA. We found similar genomic and cDNA variant nucleotide peak heights and no evidence of allele-specific expression among explant or autopsy samples with biallelic missense ABCA3 variants (n = 6). We observed allele-specific expression of missense alleles in trans with frameshift (n = 4) or nonsense (n = 1) variants, attributable to nonsense-mediated decay. The missense variant c.53 A > G;p.Gln18Arg, located near an exon-intron junction, encoded abnormal splicing with skipping of exon 4. Biologic characterization of ABCA3 variants can inform discovery of variant-specific disease mechanisms.


Assuntos
Epigênese Genética , Síndrome do Desconforto Respiratório do Recém-Nascido , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Criança , DNA/metabolismo , DNA Complementar/metabolismo , Humanos , Recém-Nascido , Pulmão/patologia , Mutação , Nucleotídeos/metabolismo , RNA/metabolismo , Síndrome do Desconforto Respiratório do Recém-Nascido/metabolismo
4.
Bioengineered ; 13(1): 508-520, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34969358

RESUMO

Neonatal acute respiratory distress syndrome (ARDS) has high morbidity and mortality rates worldwide, but there is a lack of pharmacologic treatment and clinical targeted therapies. In this study, we aimed to explore the effects of Lipocalin-2 (LCN2) on ferroptosis-mediated inflammation and oxidative stress in neonatal ARDS and the potential mechanism. In this study, we established an in vivo ARDS mouse model and an in vitro ARDS cell model by LPS (Lipopolysaccharide) stimulation. Lung tissue injury was evaluated by wet/dry ratios and histopathological examination. LCN2 expression was detected by qRT-PCR and Western blot. Inflammatory factors, oxidative stress and apoptosis were also detected. Ferroptosis was identified by detection of Fe2+ level and ferroptosis-associated protein expressions. Mitogen-activated protein kinases (MAPK)/extracellular signal-regulated kinase (ERK) pathway signaling was examined by Western blot analysis. The data revealed that LCN2 expression was significantly upregulated in neonatal mice with ARDS. Interference with LCN2 protected LPS-induced lung in neonatal mouse by reducing the radio of wet/dry and alleviating pathological damages. In addition, LCN2 silencing repressed LPS-induced inflammation, oxidative stress in vivo and in vitro, as well as apoptosis. Meanwhile, decreased level of Fe2+ and transferrin while increased levels of ferritin heavy chain 1 (FTH1) and glutathione peroxidase 4 (GPX4) were observed. The expression MAPK/ERK pathway was inhibited by depletion of LCN2. The present results suggest that LCN2 knockdown protected LPS-induced ARDS model via inhibition of ferroptosis-related inflammation and oxidative stress by inhibiting the MAPK/ERK pathway, thereby presenting a novel target for the treatment of ARDS.


Assuntos
Ferroptose , Lipocalina-2/genética , Lipopolissacarídeos/efeitos adversos , RNA Interferente Pequeno/administração & dosagem , Síndrome do Desconforto Respiratório do Recém-Nascido/tratamento farmacológico , Síndrome do Desconforto Respiratório do Recém-Nascido/genética , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Ferroptose/efeitos dos fármacos , Inativação Gênica , Sistema de Sinalização das MAP Quinases , Camundongos , Estresse Oxidativo/efeitos dos fármacos , RNA Interferente Pequeno/farmacologia , Distribuição Aleatória , Síndrome do Desconforto Respiratório do Recém-Nascido/induzido quimicamente , Síndrome do Desconforto Respiratório do Recém-Nascido/metabolismo , Transdução de Sinais , Regulação para Cima
5.
Int J Mol Sci ; 22(19)2021 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-34638622

RESUMO

ABCA3 is a crucial protein of pulmonary surfactant biosynthesis, associated with recessive pulmonary disorders such as neonatal respiratory distress and interstitial lung disease. Mutations are mostly private, and accurate interpretation of variants is mandatory for genetic counseling and patient care. We used 3D structure information to complete the set of available bioinformatics tools dedicated to medical decision. Using the experimental structure of human ABCA4, we modeled at atomic resolution the human ABCA3 3D structure including transmembrane domains (TMDs), nucleotide-binding domains (NBDs), and regulatory domains (RDs) in an ATP-bound conformation. We focused and mapped known pathogenic missense variants on this model. We pinpointed amino-acids within the NBDs, the RDs and within the interfaces between the NBDs and TMDs intracellular helices (IHs), which are predicted to play key roles in the structure and/or the function of the ABCA3 transporter. This theoretical study also highlighted the possible impact of ABCA3 variants in the cytosolic part of the protein, such as the well-known p.Glu292Val and p.Arg288Lys variants.


Assuntos
Transportadores de Cassetes de Ligação de ATP/química , Transportadores de Cassetes de Ligação de ATP/genética , Variação Genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Trifosfato de Adenosina/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Biologia Computacional , Humanos , Doenças Pulmonares Intersticiais/genética , Doenças Pulmonares Intersticiais/metabolismo , Modelos Moleculares , Mutação , Mutação de Sentido Incorreto , Conformação Proteica , Domínios Proteicos , Surfactantes Pulmonares/metabolismo , Síndrome do Desconforto Respiratório do Recém-Nascido/genética , Síndrome do Desconforto Respiratório do Recém-Nascido/metabolismo , Homologia de Sequência de Aminoácidos
6.
Am J Physiol Lung Cell Mol Physiol ; 321(6): L1036-L1043, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34585605

RESUMO

Invasive mechanical ventilation and oxygen toxicity are postnatal contributors to chronic lung disease of prematurity, also known as bronchopulmonary dysplasia (BPD). Cyfra 21-1 is a soluble fragment of cytokeratin 19, which belongs to the cytoskeleton stabilizing epithelial intermediate filaments. As a biomarker of structural integrity, Cyfra 21-1 might be associated with airway injury and lung hypoplasia in neonates. Serum Cyfra 21-1 concentrations for 80 preterm and 80 healthy term newborns were measured within 48 h after birth. Preterm infants with the combined endpoint BPD/mortality had significantly higher Cyfra 21-1 levels compared with those without fulfilling BPD/mortality criteria (P = 0.01). Also, severe RDS (>grade III) was associated with higher Cyfra levels (P = 0.01). Total duration of oxygen therapy was more than five times longer in neonates with high Cyfra 21-1 levels (P = 0.01). Infants with higher Cyfra 21-1 values were more likely to receive mechanical ventilation (50% vs. 17.5%). However, the duration of mechanical ventilation was similar between groups. The median Cyfra value was 1.93 ng/mL (IQR: 1.68-2.53 ng/mL) in healthy term neonates and 8.5 ng/mL (IQR: 3.6-16.0 ng/mL) in preterm infants. Using ROC analysis, we calculated a Cyfra cutoff > 8.5 ng/mL to predict BPD/death with an AUC of 0.795 (P = 0.004), a sensitivity of 88.9%, and a specificity of 55%. Mortality was predicted with a cutoff > 17.4 ng/mL (AUC: 0.94; P = 0.001), a sensitivity of 100%, and a specificity of 84%. These findings suggest that Cyfra 21-1 concentration might be useful to predict poor outcome in premature infants.


Assuntos
Biomarcadores/metabolismo , Displasia Broncopulmonar/mortalidade , Recém-Nascido Prematuro/crescimento & desenvolvimento , Queratina-19/metabolismo , Respiração Artificial/mortalidade , Síndrome do Desconforto Respiratório do Recém-Nascido/mortalidade , Displasia Broncopulmonar/metabolismo , Displasia Broncopulmonar/patologia , Displasia Broncopulmonar/terapia , Estudos de Casos e Controles , Feminino , Humanos , Recém-Nascido , Masculino , Prognóstico , Síndrome do Desconforto Respiratório do Recém-Nascido/metabolismo , Síndrome do Desconforto Respiratório do Recém-Nascido/patologia , Síndrome do Desconforto Respiratório do Recém-Nascido/terapia , Taxa de Sobrevida
7.
Comput Math Methods Med ; 2021: 7710129, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34471421

RESUMO

OBJECTIVE: We aimed to explore the level of PS, cell viability, inflammatory factors, and apoptosis in neonatal respiratory distress syndrome (ARDS). Besides, we explored the potential relationship between ACE2, SIRT1/eNOS pathway, and hypoxia-induced AT II cell damage. METHODS: The hUC-MSC-derived AT II cells were verified by IF and ICC, whereas qRT-PCR was used for PS and AT II cell marker (CK-8 and KGF). The AT II cell damage model was established by hypoxia exposure. The enhanced expression of ACE2 was tested after transfection with pcDNA3.1-ACE2 by western blot. The effects of hypoxia and ACE2 on AT II cells were evaluated by MTT, western blot, ELISA, and flow cytometry. The involvement of the SIRT1/eNOS pathway in AT II cell's protective functions against NRDS was verified with the addition of SIRT1 inhibitor EX527. RESULTS: Based on the successful differentiation of AT II cells from hUC-MSCs and the buildup of AT II cell damage model, the overexpressed ACE2 impeded the hypoxia-induced cellular damage of AT II cells. It also counteracted the inhibitory effects of hypoxia on the secretion of PS. Overexpression of ACE2 rescued the cell viability and suppressed the secretion of inflammatory cytokines and the apoptosis of AT II cells triggered by hypoxia. And ACE2 activated the SIRT1/eNOS pathway to play its cell-protective and anti-inflammatory roles. CONCLUSION: Our findings provided information that ACE2 prevented AT II cells from inflammatory damage through activating the SIRT1/eNOS pathway, which suggested that ACE2 might become a novel protective agent applied in the protection and treatment of NRDS.


Assuntos
Enzima de Conversão de Angiotensina 2/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Alvéolos Pulmonares/lesões , Alvéolos Pulmonares/metabolismo , Surfactantes Pulmonares/metabolismo , Sirtuína 1/metabolismo , Enzima de Conversão de Angiotensina 2/genética , Apoptose , Carbazóis/farmacologia , Diferenciação Celular , Hipóxia Celular , Sobrevivência Celular , Células Cultivadas , Biologia Computacional , Feminino , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Redes e Vias Metabólicas/efeitos dos fármacos , Alvéolos Pulmonares/efeitos dos fármacos , Síndrome do Desconforto Respiratório do Recém-Nascido/etiologia , Síndrome do Desconforto Respiratório do Recém-Nascido/metabolismo , Síndrome do Desconforto Respiratório do Recém-Nascido/prevenção & controle , Sirtuína 1/antagonistas & inibidores , Regulação para Cima
8.
Am J Physiol Lung Cell Mol Physiol ; 321(2): L291-L307, 2021 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-34132118

RESUMO

ATP-binding cassette class A3 (ABCA3) is a lipid transporter that plays a critical role in pulmonary surfactant function. The substitution of valine for glutamic acid at codon 292 (E292V) produces a hypomorphic variant that accounts for a significant portion of ABCA3 mutations associated with lung disorders spanning from neonatal respiratory distress syndrome and childhood interstitial lung disease to diffuse parenchymal lung disease (DPLD) in adults including pulmonary fibrosis. The mechanisms by which this and similar ABCA3 mutations disrupt alveolar type 2 (AT2) cell homeostasis and cause DPLD are largely unclear. The present study, informed by a patient homozygous for the E292V variant, used an in vitro and a preclinical murine model to evaluate the mechanisms by which E292V expression promotes aberrant lung injury and parenchymal remodeling. Cell lines stably expressing enhanced green fluorescent protein (EGFP)-tagged ABCA3 isoforms show a functional deficiency of the ABCA3E292V variant as a lipid transporter. AT2 cells isolated from mice constitutively homozygous for ABCA3E292V demonstrate the presence of small electron-dense lamellar bodies, time-dependent alterations in macroautophagy, and induction of apoptosis. These changes in AT2 cell homeostasis are accompanied by a spontaneous lung phenotype consisting of both age-dependent inflammation and fibrillary collagen deposition in alveolar septa. Older ABCA3E292V mice exhibit increased vulnerability to exogenous lung injury by bleomycin. Collectively, these findings support the hypothesis that the ABCA3E292V variant is a susceptibility factor for lung injury through effects on surfactant deficiency and impaired AT2 cell autophagy.


Assuntos
Transportadores de Cassetes de Ligação de ATP , Células Epiteliais Alveolares , Autofagia , Regulação da Expressão Gênica , Lesão Pulmonar , Mutação de Sentido Incorreto , Transportadores de Cassetes de Ligação de ATP/biossíntese , Transportadores de Cassetes de Ligação de ATP/genética , Células Epiteliais Alveolares/metabolismo , Células Epiteliais Alveolares/patologia , Substituição de Aminoácidos , Animais , Lesão Pulmonar/genética , Lesão Pulmonar/metabolismo , Lesão Pulmonar/patologia , Camundongos , Camundongos Mutantes , Fibrose Pulmonar/genética , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/patologia , Síndrome do Desconforto Respiratório do Recém-Nascido/metabolismo , Síndrome do Desconforto Respiratório do Recém-Nascido/patologia
9.
Pediatr Res ; 90(5): 1039-1043, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-33531681

RESUMO

BACKGROUND: Chorioamnionitis is associated with preterm delivery and morbidities; its role in lung disease is controversial. The aim of this study is to assess the effect of chorioamnionitis on metabolite and lipid profiles of epithelial lining fluid in preterm newborns with respiratory distress syndrome (RDS). METHODS: The study involved 30 newborns with RDS, born from mothers with or without histological chorioamnionitis (HCA): HCA+, N = 10; HCA-, N = 20. Patients had a gestational age ≤30 weeks; the groups were matched for age and birth weights. Tracheal aspirates were collected within 24 h after birth and analyzed using liquid chromatography/mass spectrometry-based untargeted lipidomics. RESULTS: According to Mann-Whitney U tests, 570 metabolite features had statistically significantly higher or lower concentrations (p < 0.05) in tracheal aspirates of HCA+ compared to HCA-, and 241 metabolite features were putatively annotated and classified. The most relevant changes involved higher levels of glycerophospholipids (fold change 2.42-17.69) and sphingolipids, with lower concentration of all annotated sphingomyelins in HCA+ (fold change 0.01-0.50). CONCLUSIONS: Untargeted lipidomics of tracheal aspirates suggested the production of lipid mediators in the context of an ongoing inflammatory status in HCA+ babies. However, the effect of chorioamnionitis on epithelial lining fluid composition deserves further investigations on a larger group of infants. IMPACT: Our lipidomics investigation on tracheal aspirates of preterm newborns at birth suggested that exposure to maternal histological chorioamnionitis may cause changes in epithelial lining fluid composition. This is the first description of epithelial lining fluid lipidomic profiles in preterm infants with and without exposition to chorioamnionitis. These results could provide novel link between placental membrane inflammation and newborns' respiratory outcome.


Assuntos
Corioamnionite/metabolismo , Lipidômica , Surfactantes Pulmonares/metabolismo , Síndrome do Desconforto Respiratório do Recém-Nascido/metabolismo , Feminino , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Masculino , Gravidez , Síndrome do Desconforto Respiratório do Recém-Nascido/complicações
10.
Biomed Res Int ; 2021: 4051504, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33490270

RESUMO

Neonatal respiratory distress syndrome (NRDS) is a common disease that occurs in premature infants. However, the mechanisms underlying the disease remain unclear. microRNAs (miRNAs) have been indicated to play a crucial role in the development of NRDS. In this study, we aimed to explore the regulatory mechanisms of miR-296-5p in NRDS. The expression levels of miR-296-5p in preterm infants with NRDS were determined using quantitative reverse-transcription polymerase chain reaction (RT-qPCR). A549 cells were transfected with lentiviral vectors encoding miR-296-5p, and the transfection efficiency was determined using RT-qPCR. Flow cytometry and CCK8 assay were performed to measure apoptosis and proliferation of A549 cells, respectively. The protein levels of pulmonary surfactant SP-A (SFTPA1), SP-B, Wnt7b, and ß-catenin were measured using western blotting. We demonstrated an upregulation of miR-296-5p in NRDS. The miR-296-5p was successfully overexpressed in A549 cells via lentivirus transfection, and the upregulation of miR-296-5p inhibited cell proliferation and secretion of SP-A and SP-B and also induced downregulation of the Wnt7b/ß-catenin in vitro. Therefore, miR-296-5p inhibits cell proliferation and secretion of pulmonary surfactants in A549 cells via downregulation of Wnt7b/ß-catenin signaling.


Assuntos
MicroRNAs , Surfactantes Pulmonares/metabolismo , Síndrome do Desconforto Respiratório do Recém-Nascido , Via de Sinalização Wnt/genética , Células A549 , Apoptose/genética , Sobrevivência Celular/genética , Regulação para Baixo/genética , Humanos , Recém-Nascido , MicroRNAs/genética , MicroRNAs/metabolismo , Síndrome do Desconforto Respiratório do Recém-Nascido/genética , Síndrome do Desconforto Respiratório do Recém-Nascido/metabolismo
11.
Sci Rep ; 11(1): 22, 2021 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-33420141

RESUMO

During postnatal adaptation pulmonary surfactant may be inactivated by lipopolysaccharide (LPS). We evaluated the effect of surfactant therapy in combination with antibiotic polymyxin B (PxB) in double-hit model of neonatal lung injury. Surfactant (poractant alfa, Curosurf) was exposed to smooth (S) LPS without/with PxB and tested in captive bubble surfactometer. Preterm rabbits received intratracheally saline (control) or S-LPS and were ventilated with 100% oxygen. After 30 min, LPS-treated animals received no treatment, or surfactant (200 mg/kg) without/with 3% PxB; controls received the same dose of surfactant. Animals were ventilated for further 2 h. In vitro, addition of 5% S-LPS to surfactant increased minimum surface tension (γmin) and addition of 1-3% PxB to surfactant/S-LPS mixture restored γmin to low values. Animals only given S-LPS had lower lung compliance and lung gas volume (LGV) compared to surfactant groups. Treatment with surfactant/PxB, but not with surfactant only, restored LGV. Addition of PxB to the surfactant increased the alveolar expansion. S-LPS interferes with surface activity of the pulmonary surfactant and PxB improves the resistance of surfactant to LPS-induced inactivation. In our neonatal model of respiratory distress syndrome surfactant gives positive response even in simultaneous exposure to S-LPS, when enriched with PxB.


Assuntos
Lesão Pulmonar/tratamento farmacológico , Lesão Pulmonar/metabolismo , Polimixina B/farmacologia , Surfactantes Pulmonares/metabolismo , Animais , Animais Recém-Nascidos , Produtos Biológicos/administração & dosagem , Produtos Biológicos/antagonistas & inibidores , Modelos Animais de Doenças , Feminino , Humanos , Técnicas In Vitro , Recém-Nascido , Lipopolissacarídeos/toxicidade , Pulmão/efeitos dos fármacos , Pulmão/patologia , Pulmão/fisiopatologia , Complacência Pulmonar/efeitos dos fármacos , Lesão Pulmonar/induzido quimicamente , Masculino , Fosfolipídeos/administração & dosagem , Fosfolipídeos/antagonistas & inibidores , Polimixina B/administração & dosagem , Surfactantes Pulmonares/administração & dosagem , Surfactantes Pulmonares/agonistas , Síndrome do Desconforto Respiratório do Recém-Nascido/tratamento farmacológico , Síndrome do Desconforto Respiratório do Recém-Nascido/metabolismo
12.
Am J Physiol Lung Cell Mol Physiol ; 320(2): L193-L204, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33112186

RESUMO

Premature infants, especially those with bronchopulmonary dysplasia (BPD), develop recurrent severe respiratory viral illnesses. We have shown that hyperoxic exposure of immature mice, a model of BPD, increases lung IL-12-producing Clec9a+ CD103+ dendritic cells (DCs), pro-inflammatory responses, and airway hyperreactivity following rhinovirus (RV) infection. However, the requirement for CD103+ DCs and Clec9a, a DAMP receptor that binds necrotic cell cytoskeletal filamentous actin (F-actin), for RV-induced inflammatory responses has not been demonstrated. To test this, 2-day-old C57BL/6J, CD103+ DC-deficient Batf3-/- or Clec9agfp-/- mice were exposed to normoxia or hyperoxia for 14 days. Also, selected mice were treated with neutralizing antibody against CD103. Immediately after hyperoxia, the mice were inoculated with RV intranasally. We found that compared with wild-type mice, hyperoxia-exposed Batf3-/- mice showed reduced levels of IL-12p40, IFN-γ, and TNF-α, fewer IFN-γ-producing CD4+ T cells, and decreased airway responsiveness following RV infection. Similar effects were observed in anti-CD103-treated and Clec9agfp-/- mice. Furthermore, hyperoxia increased airway dead cell number and extracellular F-actin levels. Finally, studies in preterm infants with respiratory distress syndrome showed that tracheal aspirate CLEC9A expression positively correlated with IL12B expression, consistent with the notion that CLEC9A+ cells are responsible for IL-12 production in humans as well as mice. We conclude that CD103+ DCs and Clec9a are required for hyperoxia-induced pro-inflammatory responses to RV infection. In premature infants, Clec9a-mediated activation of CD103+ DCs may promote pro-inflammatory responses to viral infection, thereby driving respiratory morbidity.


Assuntos
Antígenos CD/metabolismo , Células Dendríticas/imunologia , Hiperóxia/fisiopatologia , Cadeias alfa de Integrinas/metabolismo , Lectinas Tipo C/fisiologia , Pulmão/imunologia , Pneumonia/imunologia , Receptores Imunológicos/fisiologia , Síndrome do Desconforto Respiratório do Recém-Nascido/imunologia , Animais , Animais Recém-Nascidos , Antígenos CD/genética , Fatores de Transcrição de Zíper de Leucina Básica/fisiologia , Feminino , Humanos , Recém-Nascido , Recém-Nascido Prematuro/imunologia , Cadeias alfa de Integrinas/genética , Pulmão/metabolismo , Pulmão/patologia , Pulmão/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções por Picornaviridae/complicações , Infecções por Picornaviridae/virologia , Pneumonia/virologia , Proteínas Repressoras/fisiologia , Síndrome do Desconforto Respiratório do Recém-Nascido/metabolismo , Síndrome do Desconforto Respiratório do Recém-Nascido/patologia , Rhinovirus/isolamento & purificação
13.
Peptides ; 135: 170398, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33022295

RESUMO

Previous studies have shown that, oxytocin has anticonvulsant and neuroprotective effects. One of the most important complications of Hypercapnic-hypoxia is drug resistance epilepsy. Effects of chronic intraperitoneal oxytocin treatment on gliosis, neuroinflammation and seizure activity was investigated in a model in which rats were exposed to hypoxia on postnatal day 1 and later challenged to the seizure-inducing pentylenetetrazol Forty pups were included in the study on their first day of birth. 16 pups were exposed to 100% CO2 for 5 minutes and other 16 pups for 10 minutes. The remaining 8 pups comprised the control group. Groups were classified according to oxytocin administration within the first 4 weeks. Pentylenetetrazol was administered 6 months after the oxytocin treatment. The Racine's Convulsion Scale and onset times of first myoclonic jerk (FMJ) were evaluated. To determine the mechanisms by which oxytocin exerted its effects on hypercapnic-anoxia exposed rats, we performed CA1 total neuron count & CA1 GFAP immunostaining, and measured brain levels of TNF-α and GAD-67. The Racine scale and TNF-α values were significantly lower in both groups that received oxytocin, while time-to-FMJ and GAD-67 level were significantly higher. The histopathological evaluations showed that oxytocin had significant ameliorative effects (especially regarding gliosis) on the hippocampus of hypoxic rats. Regarding the results of present study, it can be speculated that after acute hypercapnic-anoxia exposure, chronic Oxytocin treatment has long lasting therapeutic potential on rats, possibly by reducing the gliosis with its anti-inflammatory feature and by activating the GABA pathway.


Assuntos
Gliose/tratamento farmacológico , Ocitocina/farmacologia , Síndrome do Desconforto Respiratório do Recém-Nascido/tratamento farmacológico , Insuficiência Respiratória/tratamento farmacológico , Animais , Animais Recém-Nascidos , Anti-Inflamatórios , Encéfalo/efeitos dos fármacos , Modelos Animais de Doenças , Neurônios GABAérgicos/efeitos dos fármacos , Gliose/metabolismo , Gliose/patologia , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Hipóxia/tratamento farmacológico , Hipóxia/patologia , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Inflamação/patologia , Fármacos Neuroprotetores/farmacologia , Ratos , Síndrome do Desconforto Respiratório do Recém-Nascido/metabolismo , Síndrome do Desconforto Respiratório do Recém-Nascido/patologia , Insuficiência Respiratória/metabolismo , Insuficiência Respiratória/patologia
14.
Pediatr Pulmonol ; 55(11): 2964-2969, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32770804

RESUMO

BACKGROUND: Hypoxia and asphyxia are known to induce surfactant inactivation in newborns. Deleted in Malignant Brain Tumors 1 (DMBT1) is an innate immunity protein with functions in epithelial differentiation and angiogenesis. It was detected in hyaline membranes of infants with respiratory distress syndrome. Human recombinant DMBT1 is able to increase the surface tension of exogenous surfactant preparations in a dose-dependent manner. METHODS: Immunohistochemistry was performed on lung sections of infants who died due to pre-, peri- or postnatal hypoxia. The lung epithelial cell line A549 was stably transfected with a DMBT1 (DMBT1+ cells) expression plasmid or with an empty plasmid (DMBT1- cells). The cells were cultured in normoxic or hypoxic conditions, and then DMBT1 as well as HIF-1α RNA expression were analyzed by using real-time-polymerase chain reaction. Human recombinant DMBT1 was added to the modified porcine natural surfactant Curosurf to examine the effect of DMBT1 on surfactant ultrastructure with electron microscopy. RESULTS: DMBT1 expression was upregulated in human lung tissue after fetal/peri-/postnatal hypoxia. In addition, in vitro experiments showed increased DMBT1 RNA expression in A549 cells after hypoxia. HIF-1α was upregulated in both DMBT1+ and DMBT1- cells in response to hypoxia. The addition of human recombinant DMBT1 to Curosurf caused an impaired surfactant ultrastructure. CONCLUSIONS: DMBT1 is upregulated in response to hypoxia and there seems to be a link between hypoxia and surfactant inactivation.


Assuntos
Produtos Biológicos/administração & dosagem , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Ligação a DNA/metabolismo , Células Epiteliais/metabolismo , Hipóxia/metabolismo , Pulmão/metabolismo , Fosfolipídeos/administração & dosagem , Surfactantes Pulmonares/administração & dosagem , Síndrome do Desconforto Respiratório do Recém-Nascido/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteínas de Ligação ao Cálcio/genética , Linhagem Celular , Proteínas de Ligação a DNA/genética , Humanos , Hipóxia/genética , Recém-Nascido , Pulmão/citologia , Síndrome do Desconforto Respiratório do Recém-Nascido/genética , Proteínas Supressoras de Tumor/genética , Regulação para Cima
15.
Pediatr Res ; 87(5): 940-945, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31715622

RESUMO

BACKGROUND: In preterm infants on moderately high ventilator support, the addition of budesonide to surfactant lowered bronchopulmonary dysplasia (BPD) rates by 20% without increased morbidity or mortality. The aim of this cohort comparison was to determine the safety and efficacy of the combination in infants with milder respiratory distress syndrome (RDS). METHODS: In August 2016 we began administering budesonide (0.25 mg/kg) mixed with surfactant (Survanta 4 mL/kg) to all infants ≤ 1250 g who failed CPAP and required intubation. Infants were compared to a historical cohort (2013-2016) who received surfactant alone. RESULTS: BPD or death did not change between the historical surfactant cohort (71%, n = 294) and the budesonide cohort (69%, n = 173). Budesonide was associated with a decrease in the need for continued mechanical ventilation, severe BPD type II or death (19-12%), grade III BPD or death (31-21%), and the median gestational age at discharge was 1 week earlier. Histologic chorioamnionitis was associated with decreased budesonide effects. Secondary morbidities (NEC, IVH, ROP, Sepsis) were similar. CONCLUSION: Overall BPD rates remained unchanged with the addition of budesonide. Budesonide was associated with decreased severity of BPD, decreased mechanical ventilation use, earlier discharge, and similar short-term outcomes.


Assuntos
Displasia Broncopulmonar/tratamento farmacológico , Budesonida/administração & dosagem , Síndrome do Desconforto Respiratório do Recém-Nascido/tratamento farmacológico , Síndrome do Desconforto Respiratório do Recém-Nascido/metabolismo , Tensoativos/administração & dosagem , Corioamnionite , Feminino , Humanos , Recém-Nascido , Masculino , Alta do Paciente , Segurança do Paciente , Gravidez , Surfactantes Pulmonares/uso terapêutico , Respiração Artificial , Resultado do Tratamento
16.
Int J Mol Sci ; 21(1)2019 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-31861781

RESUMO

Preterm infants frequently suffer from respiratory distress syndrome (RDS), possibly due to lower expression of epithelial Na+ channels (ENaC). RDS incidence is sex-specific, affecting males almost twice as often. Despite the use of antenatal glucocorticoids (GCs), the sex difference persists. It is still controversial whether both sexes benefit equally from GCs. We previously showed that Na+ transport is higher in female compared with male fetal distal lung epithelial (FDLE) cells. Since GCs increase Na+ transport, we hypothesized that their stimulating effect might be sex-specific. We analyzed FDLE cells with Ussing chambers and RT-qPCR in the presence or absence of fetal serum. In serum-free medium, GCs increased the ENaC activity and mRNA expression, independent of sex. In contrast, GCs did not increase the Na+ transport in serum-supplemented media and abolished the otherwise observed sex difference. Inhibition of the GC receptor in the presence of serum did not equalize Na+ transport between male and female cells. The GC-induced surfactant protein mRNA expression was concentration and sex-specific. In conclusion, female and male FDLE cells exhibit no sex difference in response to GCs with regard to Na+ transport, and GR activity does not contribute to the higher Na+ transport in females.


Assuntos
Células Epiteliais Alveolares/efeitos dos fármacos , Canais Epiteliais de Sódio/metabolismo , Glucocorticoides/farmacologia , Células Epiteliais Alveolares/metabolismo , Animais , Animais Recém-Nascidos , Transporte Biológico/efeitos dos fármacos , Células Cultivadas , Feminino , Humanos , Masculino , Ratos , Síndrome do Desconforto Respiratório do Recém-Nascido/tratamento farmacológico , Síndrome do Desconforto Respiratório do Recém-Nascido/metabolismo , Caracteres Sexuais , Sódio/metabolismo
17.
Eur Rev Med Pharmacol Sci ; 23(21): 9548-9556, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31773706

RESUMO

OBJECTIVE: The aim of this study was to investigate the effect of micro-ribonucleic acid (miR)-200a on respiratory distress syndrome (RDS) in newborn rabbits by regulating the Wnt/ß-catenin signaling pathway. MATERIALS AND METHODS: In this work, newborn rabbits aged three days were selected from our laboratory as research objects. The messenger RNA (mRNA) and protein expression levels of miR-200a, ß-catenin and interleukin-10 (IL-10) in blood samples of healthy newborn rabbits and newborn rabbits with RDS were determined by fluorescence quantitative Polymerase Chain Reaction (PCR) and Western blotting, respectively. Lentivirus-packaged plasmids containing miR-200a were then injected into newborn rabbits suffering from RDS. After 2 d, the mRNA and protein expression levels of miR-200a, ß-catenin and IL-10 in blood samples of newborn rabbits in different treatment groups were measured. Meanwhile, lung sections were collected from newborn rabbits in different treatment groups. After that, the sections were observed via hematoxylin and eosin (H&E) staining. At the same time, lung coefficient of newborn rabbits in different treatment groups was also measured. RESULTS: Compared with healthy newborn rabbits, the mRNA and protein expression levels of miR-200a and IL-10 in the blood of newborn rabbits with RDS decreased significantly (p<0.05), while ß-catenin increased markedly (p<0.05). The mRNA and protein expression levels of ß-catenin and IL-10 in newborn RDS rabbits with miR-200a over-expression and knockout were detected as well. The results revealed that lowly expressed miR-200a could remarkably promote the expression level of ß-catenin, whereas inhibiting the expression of IL-10. However, highly expressed miR-200a could significantly inhibit the expression level of ß-catenin and promote the expression level of IL-10. H&E staining results manifested that miR-200a knockout markedly promoted the increase of pulmonary alveoli with increased lung coefficients. However, the up-regulation of miR-200a could reduce lung coefficients and remarkably improve RDS. CONCLUSIONS: MiR-200a regulates RDS in newborn rabbits by regulating the Wnt/ß-catenin signaling pathway.


Assuntos
MicroRNAs/metabolismo , Síndrome do Desconforto Respiratório do Recém-Nascido/metabolismo , beta Catenina/metabolismo , Animais , Animais Recém-Nascidos , Linhagem Celular , Humanos , Injeções Intravenosas , MicroRNAs/administração & dosagem , MicroRNAs/genética , Coelhos , Síndrome do Desconforto Respiratório do Recém-Nascido/patologia , Via de Sinalização Wnt/genética
18.
Eur Rev Med Pharmacol Sci ; 23(17): 7550-7556, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31539145

RESUMO

OBJECTIVE: Acute respiratory distress syndrome (ARDS) threatens humans' health worldwide, causing huge labor and economic cost investment. This study aims to explore whether mesenchymal stem cells (MSCs) affect RDS in newborn swines via the Janus kinase-signal transducers and activators of transcription (JAK-STAT) signaling pathway by the establishment of the model of the disease. MATERIALS AND METHODS: The phosphorylation of the JAK-STAT signal transduction proteins was first detected via Western blotting to verify the regulatory effect of MSCs on RDS in newborn swines through the JAK-STAT signaling pathway. Then, the Reverse Transcription-Polymerase Chain Reaction (RT-PCR) was utilized to analyze the influences of the injection of MSCs into the blood of newborn model RDS swines on inflammatory factors in vivo. To further demonstrate the signal transduction function put forwarded, the RT-PCR and enzyme-linked immunosorbent assay (ELISA) were adopted to analyze the influences of the JAK-STAT signaling pathway inhibitor on the expression of the signature proteins of RDS in newborn swines and the changes in the inflammatory factors. RESULTS: MSCs induced the phosphorylation of JAK and STAT, and they activated the JAK-STAT signal transduction of RDS in newborn swines. Compared with those in normal saline group, the interleukin (IL)-2, IL-6, IL-8, and tumor necrosis factor-α (TNF-α) expression levels in MSC group were increased, namely, MSCs substantially promoted their expression levels (p<0.05), but those of IL-10 and IL-13 were significantly decreased (p<0.05). CONCLUSIONS: The inhibitor of the JAK-STAT signaling pathway can suppress the therapeutic effect of MSCs on RDS in newborn swines.


Assuntos
Janus Quinases/metabolismo , Síndrome do Desconforto Respiratório do Recém-Nascido/patologia , Fator de Transcrição STAT3/metabolismo , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Interleucina-10/sangue , Interleucina-6/sangue , Janus Quinases/antagonistas & inibidores , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Fosforilação/efeitos dos fármacos , Piridinas/farmacologia , Síndrome do Desconforto Respiratório do Recém-Nascido/metabolismo , Síndrome do Desconforto Respiratório do Recém-Nascido/terapia , Fator de Transcrição STAT3/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Suínos , Fator de Necrose Tumoral alfa/sangue , Tirfostinas/farmacologia , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
19.
Eur Rev Med Pharmacol Sci ; 23(16): 7042-7048, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31486505

RESUMO

OBJECTIVE: Acute respiratory distress syndrome (ARDS) is greatly threatening human health with high morbidity and mortality. The pathogenesis of ARDS is closely related to the inflammatory response in patients. The micro-ribonucleic acid (miR)-155/nuclear factor-κB (NF-κB) signaling pathway is crucial in regulating the expression of inflammation-related genes. Therefore, the influences of miR-155/NF-κB signaling pathway on inflammatory factors in ARDS in neonatal pigs were explored in this study. MATERIALS AND METHODS: The model of ARDS in neonatal pigs was established first. The expression levels of miR-155, NF-κB-related proteins, and inflammatory factors in model group and control group were detected, and their differences were compared. Moreover, after treatment with the miR-155/NF-κB signaling pathway inhibitor, the changes of the inflammatory factors expression in ARDS neonatal pigs were observed at different time points. RESULTS: In the model group, the levels of miR-155 and NF-κB-related proteins were significantly increased, and the levels of inflammatory factors, including interleukin-1ß (IL-1ß), tumor necrosis factor-α (TNF-α), and IL-6, were also increased synchronously. However, the levels of IL-4 and IL-10 declined significantly. In addition, it was proved that after treatment with the inhibitor in model group the mRNA expressions of miR-155/NF-κB signaling pathway-related proteins were significantly inhibited, and the levels of IL-1ß, TNF-α, and IL-6 were also significantly inhibited (p<0.05). The levels of IL-4 and IL-10 remarkably rose after treatment with the inhibitor for 24 h (p<0.05). CONCLUSIONS: The miR-155/NF-κB signaling pathway influenced the changes of inflammatory factors in ARDS in neonatal pigs, which might be a potential target for eliminating the inflammatory response after ARDS in neonatal pigs.


Assuntos
Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , MicroRNAs/metabolismo , NF-kappa B/metabolismo , Síndrome do Desconforto Respiratório do Recém-Nascido/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Modelos Animais de Doenças , Interleucina-1beta/genética , Interleucina-6/genética , MicroRNAs/genética , NF-kappa B/genética , Transdução de Sinais , Suínos , Fator de Necrose Tumoral alfa/genética
20.
Mol Cell Neurosci ; 100: 103400, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31472222

RESUMO

Several studies have investigated the general role of chloride-based neurotransmission (GABAA and glycinergic signaling) in respiratory rhythmogenesis and pattern formation. In several brain regions, developmental alterations in these signaling pathways have been shown to be mediated by changes in cation-chloride cotransporter (CC) expression. For instance, CC expression changes during the course of neonatal development in medullary respiratory nuclei and other brain/spinal cord regions in a manner which decreases the cellular import, and increases the export, of chloride ions, shifting reversal potentials for chloride to progressively more negative values with maturation. In slice preparations of the same, this is related to an excitatory-to-inhibitory shift of GABAA- and glycinergic signaling. In medullary slices, GABAA-/glycinergic signaling in the early neonatal period is excitatory, becoming inhibitory over time. Additionally, blockade of the Na+/K+/2Cl- cotransporter, which imports these ions via secondary active transport, converts excitatory response to inhibitory ones. These effects have not yet been demonstrated at the individual respiratory-related neuron level to occur in intact (in vivo or in situ) animal preparations, which in contrast to slices, possess normal network connectivity and natural sources of tonic drive. Developmental changes in respiratory rhythm generating and pattern forming pontomedullary respiratory circuitry may contribute to critical periods, during which there exist increased risk for perinatal respiratory disturbances of central, obstructive, or hypoxia/hypercapnia-induced origin, including the sudden infant death syndrome. Thus, better characterizing the neurochemical maturation of the central respiratory network will enhance our understanding of these conditions, which will facilitate development of targeted therapies for respiratory disturbances in neonates and infants.


Assuntos
Relógios Biológicos , Potenciais Pós-Sinápticos Inibidores , Respiração , Síndrome do Desconforto Respiratório do Recém-Nascido/fisiopatologia , Animais , Glicina/metabolismo , Humanos , Síndrome do Desconforto Respiratório do Recém-Nascido/metabolismo , Ácido gama-Aminobutírico/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA