Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Int J Mol Sci ; 25(18)2024 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-39337630

RESUMO

Bronchopulmonary dysplasia (BPD) is the most common lung complication of prematurity. Despite extensive research, our understanding of its pathophysiology remains limited, as reflected by the stable prevalence of BPD. Prematurity is the primary risk factor for BPD, with oxidative stress (OS) and inflammation playing significant roles and being closely linked to premature birth. Understanding the interplay and temporal relationship between OS and inflammation is crucial for developing new treatments for BPD. Animal studies suggest that OS and inflammation can exacerbate each other. Clinical trials focusing solely on antioxidants or anti-inflammatory therapies have been unsuccessful. In contrast, vitamin A and caffeine, with antioxidant and anti-inflammatory properties, have shown some efficacy, reducing BPD by about 10%. However, more than one-third of very preterm infants still suffer from BPD. New therapeutic agents are needed. A novel tripeptide, N-acetyl-lysyltyrosylcysteine amide (KYC), is a reversible myeloperoxidase inhibitor and a systems pharmacology agent. It reduces BPD severity by inhibiting MPO, enhancing antioxidative proteins, and alleviating endoplasmic reticulum stress and cellular senescence in a hyperoxia rat model. KYC represents a promising new approach to BPD treatment.


Assuntos
Displasia Broncopulmonar , Inflamação , Estresse Oxidativo , Displasia Broncopulmonar/metabolismo , Displasia Broncopulmonar/patologia , Displasia Broncopulmonar/tratamento farmacológico , Estresse Oxidativo/efeitos dos fármacos , Humanos , Animais , Inflamação/metabolismo , Inflamação/tratamento farmacológico , Antioxidantes/farmacologia , Antioxidantes/uso terapêutico , Recém-Nascido , Recém-Nascido Prematuro , Ratos , Anti-Inflamatórios/uso terapêutico , Anti-Inflamatórios/farmacologia
2.
Am J Physiol Lung Cell Mol Physiol ; 312(5): L586-L598, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28213471

RESUMO

Rodent pups exposed to hyperoxia develop lung changes similar to bronchopulmonary dysplasia (BPD) in extremely premature infants. Oxidative stress from hyperoxia can injure developing lungs through endoplasmic reticulum (ER) stress. Early caffeine treatment decreases the rate of BPD, but the mechanisms remain unclear. We hypothesized that caffeine attenuates hyperoxia-induced lung injury through its chemical chaperone property. Sprague-Dawley rat pups were raised either in 90 (hyperoxia) or 21% (normoxia) oxygen from postnatal day 1 (P1) to postnatal day 10 (P10) and then recovered in 21% oxygen until P21. Caffeine (20 mg/kg) or normal saline (control) was administered intraperitoneally daily starting from P2. Lungs were inflation-fixed for histology or snap-frozen for immunoblots. Blood caffeine levels were measured in treated pups at euthanasia and were found to be 18.4 ± 4.9 µg/ml. Hyperoxia impaired alveolar formation and increased ER stress markers and downstream effectors; caffeine treatment attenuated these changes at P10. Caffeine also attenuated the hyperoxia-induced activation of cyclooxygenase-2 and markers of apoptosis. In conclusion, hyperoxia-induced alveolar growth impairment is mediated, in part, by ER stress. Early caffeine treatment protects developing lungs from hyperoxia-induced injury by attenuating ER stress.


Assuntos
Cafeína/farmacologia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Hiperóxia/complicações , Hiperóxia/patologia , Lesão Pulmonar/etiologia , Lesão Pulmonar/patologia , Animais , Apoptose/efeitos dos fármacos , Cafeína/sangue , Ciclo-Oxigenase 2/metabolismo , Metabolismo Energético/efeitos dos fármacos , Feminino , Proteínas de Choque Térmico/metabolismo , Hiperóxia/enzimologia , Pulmão/irrigação sanguínea , Pulmão/efeitos dos fármacos , Pulmão/patologia , Lesão Pulmonar/enzimologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Modelos Biológicos , Neovascularização Fisiológica/efeitos dos fármacos , Biogênese de Organelas , Estresse Oxidativo/efeitos dos fármacos , Peroxidase/metabolismo , Pneumonia/complicações , Pneumonia/patologia , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/patologia , Ratos Sprague-Dawley , Resposta a Proteínas não Dobradas/efeitos dos fármacos
3.
Am J Physiol Cell Physiol ; 310(1): C80-8, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26491046

RESUMO

Nitration of both protein-bound and free tyrosine by reactive nitrogen species results in the formation of nitrotyrosine (NT). We previously reported that free NT impairs microtubule polymerization and uncouples endothelial nitric oxide synthase (eNOS) function in pulmonary artery endothelial cells (PAEC). Because microtubules modulate mitochondrial function, we hypothesized that increased NT levels during inflammation and oxidative stress will lead to mitochondrial dysfunction in PAEC. PAEC isolated from fetal lambs were exposed to varying concentrations of free NT. At low concentrations (1-10 µM), NT increased nitration of mitochondrial electron transport chain (ETC) protein subunit complexes I-V and state III oxygen consumption. Higher concentrations of NT (50 µM) caused decreased microtubule acetylation, impaired eNOS interactions with mitochondria, and decreased ETC protein levels. We also observed increases in heat shock protein-90 nitration, mitochondrial superoxide formation, and fragmentation of mitochondria in PAEC. Our data suggest that free NT accumulation may impair microtubule polymerization and exacerbate reactive oxygen species-induced cell damage by causing mitochondrial dysfunction.


Assuntos
Células Endoteliais/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Artéria Pulmonar/efeitos dos fármacos , Tirosina/análogos & derivados , Acetilação , Animais , Células Cultivadas , DNA Mitocondrial/biossíntese , Relação Dose-Resposta a Droga , Complexo de Proteínas da Cadeia de Transporte de Elétrons/metabolismo , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Idade Gestacional , Proteínas de Choque Térmico HSP90/metabolismo , Microtúbulos/efeitos dos fármacos , Microtúbulos/metabolismo , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Óxido Nítrico Sintase Tipo III/metabolismo , Consumo de Oxigênio/efeitos dos fármacos , Ligação Proteica , Artéria Pulmonar/metabolismo , Artéria Pulmonar/patologia , Ovinos , Superóxidos/metabolismo , Tirosina/toxicidade
4.
J Formos Med Assoc ; 114(4): 359-62, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25839770

RESUMO

BACKGROUND/PURPOSE: Insulin-like growth factors (IGFs) and their binding proteins (IGFBPs) are known to modulate fetal growth but their role in intrauterine growth of monochorionic twins (MCT) has not been studied. METHODS: Cord venous blood was collected directly after birth. IGF-1 and IGFBP-3 in the cord venous blood were quantified by radioimmunoassay. Birth weights (BWs) were obtained electronically. Placentas were examined for chorionicity. RESULTS: Cord blood was collected in 37 pairs of MCT (15 pairs were males). BWs ranged from 564 to 3240 g, and gestational ages (GAs) were between 24 weeks and 39 weeks. There was a correlation between BW and cord venous blood IGFBP-3 concentration (r = 0.28, p = 0.015), but not between BW and cord venous blood IGF-1 level. There was no difference in IGF-1 between the heavier twins (30.8 ± 61.8 ng/mL) and lighter twins (33.2 ± 63.7 ng/mL), but a trend (p = 0.096) of higher IGFBP-3 level was demonstrated in heavier twins (3.14 ± 1.23 µg/mL) than in lighter twins (2.71 ± 1.19 µg/mL). The IGFBP-3 levels were higher (p = 0.042) in female twins (3.20 ± 1.33 µg/mL) than in male twins (2.64 ± 1.04 µg/mL). The IGF-1 level of the heavier twins correlated significantly to their lighter co-twin (r = 0.73, p < 0.001). CONCLUSION: Our data showed that cord venous blood IGF-1 level might be controlled mainly by genetic factors. IGFBP-3 might play an important role in fetal growth.


Assuntos
Sangue Fetal/química , Retardo do Crescimento Fetal/metabolismo , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/sangue , Fator de Crescimento Insulin-Like I/química , Peso ao Nascer , Feminino , Idade Gestacional , Humanos , Recém-Nascido , Masculino , Taiwan , Gêmeos
5.
J Formos Med Assoc ; 114(2): 174-9, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25678180

RESUMO

BACKGROUND/PURPOSE: Neutropenia is a risk factor for nosocomial infections (NI) in very-low-birth-weight (VLBW) infants. Although recombinant human granulocyte colony stimulating factor (rhG-CSF) increases the neutrophil counts in neutropenic VLBW infants, its long-term efficacy for early neutropenia (EN) remains unknown. METHODS: In this case-controlled study, charts of VLBW recipients of rhG-CSF for EN (total neutrophil count <1.5 × 10(9)/L during first 7 days) were reviewed and compared to gestational age, total neutrophil count, and birth weight matched infants unexposed to rhG-CSF. RESULTS: Twenty-seven infants were identified in each group. Mortality and morbidity did not differ between the two groups. Rate of NI (16/27 vs. 4/27, p = 0.002, odds ratio = 8.36) as well as the total number of episodes of NI (22 vs. 4, p = 0.007) were higher in rhG-CSF (+) group than in the rhG-CSF (-) group. CONCLUSION: Our experience does not show benefit in empirical use of rhG-CSF in preventing NI in VLBW infants with EN.


Assuntos
Infecção Hospitalar/prevenção & controle , Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Recém-Nascido Prematuro , Recém-Nascido de muito Baixo Peso , Neutropenia/tratamento farmacológico , Estudos de Casos e Controles , Feminino , Florida , Idade Gestacional , Humanos , Lactente , Recém-Nascido , Unidades de Terapia Intensiva Neonatal , Contagem de Leucócitos , Modelos Logísticos , Masculino , Análise Multivariada , Neutropenia/complicações , Neutropenia/mortalidade , Proteínas Recombinantes/uso terapêutico
6.
Antioxidants (Basel) ; 13(8)2024 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-39199135

RESUMO

Bronchopulmonary dysplasia (BPD) is a lung complication of premature births. The leading causes of BPD are oxidative stress (OS) from oxygen treatment, infection or inflammation, and mechanical ventilation. OS activates alveolar myeloid cells with subsequent myeloperoxidase (MPO)-mediated OS. Premature human neonates lack sufficient antioxidative capacity and are susceptible to OS. Unopposed OS elicits inflammation, endoplasmic reticulum (ER) stress, and cellular senescence, culminating in a BPD phenotype. Poor nutrition, patent ductus arteriosus, and infection further aggravate OS. BPD survivors frequently suffer from reactive airway disease, neurodevelopmental deficits, and inadequate exercise performance and are prone to developing early-onset chronic obstructive pulmonary disease. Rats and mice are commonly used to study BPD, as they are born at the saccular stage, comparable to human neonates at 22-36 weeks of gestation. The alveolar stage in rats and mice starts at the postnatal age of 5 days. Because of their well-established antioxidative capacities, a higher oxygen concentration (hyperoxia, HOX) is required to elicit OS lung damage in rats and mice. Neutrophil infiltration and ER stress occur shortly after HOX, while cellular senescence is seen later. Studies have shown that MPO plays a critical role in the process. A novel tripeptide, N-acetyl-lysyltyrosylcysteine amide (KYC), a reversible MPO inhibitor, attenuates BPD effectively. In contrast, the irreversible MPO inhibitor-AZD4831-failed to provide similar efficacy. Interestingly, KYC cannot offer its effectiveness without the existence of MPO. We review the mechanisms by which this anti-MPO agent attenuates BPD.

7.
J Formos Med Assoc ; 112(4): 177-84, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23537863

RESUMO

Persistent pulmonary hypertension of the newborn (PPHN) is a severe pulmonary disorder which occurs at a rate of one in every 500 live births. About 10-50% of the victims will die of the problem and 7-20% of the survivors develop long-term impairments such as hearing deficit, chronic lung disease, and intracranial bleed. Most adult survivors show evidence of augmented pulmonary vasoreactivity, suggesting a phenotypical change. Several animal models have been used to study the pathophysiology and help to develop new therapeutic modality for PPHN. The etiology of PPHN can be classified into three groups: (1) abnormally constricted pulmonary vasculature as a result of parenchymal diseases; (2) hypoplastic pulmonary vasculature; and (3) normal parenchyma with remodeled pulmonary vasculature. Impaired vasorelaxation of pulmonary artery and reduced blood vessel density in lungs are two characteristic findings in PPHN. Medical treatment includes sedation, oxygen, mechanical ventilation, vasorelaxants (inhaled nitric oxide, inhaled or intravenous prostacyclin, intravenous prostaglandin E1, magnesium sulfate), and inotropic agents. Phosphodiesterase inhibitors have recently been studied as another therapeutic agent for PPHN. Endothelin-1 (ET-1) inhibitors have been studied in animals and a case of premature infant with PPHN successfully treated with an ET-I inhibitor has been reported in the literature. Surfactants have been reported as an adjunct treatment for PPHN as a complication of meconium aspiration syndrome. Even with the introduction of several new therapeutic modalities there has been no significant change in survival rate. Extracorporeal membrane oxygenator is used when medical treatment fails and the patient is considered to have a recoverable cause of PPHN.


Assuntos
Síndrome da Persistência do Padrão de Circulação Fetal/etiologia , Animais , Modelos Animais de Doenças , Oxigenação por Membrana Extracorpórea , Retardo do Crescimento Fetal/fisiopatologia , Humanos , Óxido Nítrico Sintase Tipo III/fisiologia , Síndrome da Persistência do Padrão de Circulação Fetal/diagnóstico , Síndrome da Persistência do Padrão de Circulação Fetal/terapia , Respiração Artificial , Fatores de Risco
8.
Pediatr Res ; 69(2): 112-7, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21057377

RESUMO

Infection is known to impair the growth of developing lungs. It is known that plasma free nitrotyrosine (NT) levels can reach 150 µM during sepsis. Free NT incorporates into microtubules and impairs cell function. We hypothesize that free NT perturbs the angiogenic activity of pulmonary artery endothelial cells (PAEC) in developing lungs. PAEC from fetal lamb lungs were incubated with NT (1-100 µM). We examined the effects of NT on tube formation, cell proliferation, apoptosis, and α-tubulin assembly in PAEC. We assessed superoxide anion (O2) and NO levels in PAEC during NT exposure. Effects of NT on endothelial NO synthase (eNOS) were examined with respect to eNOS-dimer formation and the association of eNOS chaperone, heat-shock-protein-90 (hsp90). NT decreased tube formation and increased apoptosis in PAEC. NT also decreased NO levels, increased NOS-dependent O2 generation, and promoted α-tubulin depolymerization. Although NT increased eNOS homodimer formation, it decreased the hsp90 association with eNOS. Our data suggest that increased NT formation during sepsis may uncouple eNOS activity and increase oxidative stress. Because NO plays an important role in angiogenesis and vasodilation, these observations suggest a mechanism for the impaired vasodilation and angiogenesis during sepsis in the developing lung.


Assuntos
Células Endoteliais/enzimologia , Pulmão/irrigação sanguínea , Pulmão/enzimologia , Neovascularização Fisiológica , Óxido Nítrico Sintase Tipo III/metabolismo , Artéria Pulmonar/enzimologia , Sepse/enzimologia , Tirosina/análogos & derivados , Animais , Apoptose , Proliferação de Células , Células Cultivadas , Cesárea , Células Endoteliais/patologia , Idade Gestacional , Proteínas de Choque Térmico HSP90/metabolismo , Pulmão/crescimento & desenvolvimento , Microtúbulos/metabolismo , Óxido Nítrico/metabolismo , Nascimento Prematuro , Multimerização Proteica , Artéria Pulmonar/crescimento & desenvolvimento , Artéria Pulmonar/patologia , Sepse/patologia , Sepse/fisiopatologia , Ovinos , Superóxido Dismutase/metabolismo , Superóxidos/metabolismo , Tubulina (Proteína)/metabolismo , Tirosina/metabolismo
9.
Free Radic Res ; 41(2): 127-34, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17364938

RESUMO

Reactive oxygen and nitrogen species have been implicated in ischemia-reperfusion (I/R) injury. Metalloporphyrins (MP) are stable catalytic antioxidants that can scavenge superoxide, hydrogen peroxide, peroxynitrite and lipid peroxyl radicals. Studies were conducted with three manganese-porphyrin (MnP) complexes with varying superoxide dimutase (SOD) and catalase catalytic activity to determine if the MnP attenuates I/R injury in isolated perfused mouse livers. The release of the hepatocellular enzymes alanine aminotransferase (ALT), aspartate aminotransferase (AST) and lactate dehydrogenase (LDH) was maximal at 1 min reperfusion, decreased rapidly and increased gradually by 90 min. Manganese tetrakis-(N-ethyl-2 pyridyl) porphyrin (MnTE-2-PyP) decreased ALT, AST, LDH at 1-90 min reperfusion, while manganese tetrakis-(N-methyl-2 pyridyl) porphyrin (MnTM-2-PyP) and manganese tetrakis-(ethoxycarbonyl) porphyrin (MnTECP) decreased ALT and LDH from 5 to 90 min reperfusion. The release of thiobarbituric acid-reacting substances (TBARS) was diminished by MnTE-2-PyP and MnTM-2-PyP at 90 min. The extent of protein nitration (nitrotyrosine, NT) was decreased in all three MnPs treated livers. These results demonstrate that MnP complexes can attenuate hepatic I/R injury and may have therapeutic implications in disease states involving oxidants.


Assuntos
Antioxidantes/uso terapêutico , Isquemia/tratamento farmacológico , Hepatopatias/prevenção & controle , Fígado/efeitos dos fármacos , Metaloporfirinas/uso terapêutico , Traumatismo por Reperfusão/prevenção & controle , Alanina Transaminase/metabolismo , Animais , Antioxidantes/farmacologia , Aspartato Aminotransferases/metabolismo , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , L-Lactato Desidrogenase/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Fígado/irrigação sanguínea , Masculino , Metaloporfirinas/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Nitrosação/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Substâncias Reativas com Ácido Tiobarbitúrico/análise , Tirosina/análogos & derivados , Tirosina/análise
10.
Biol Neonate ; 90(4): 218-25, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16636534

RESUMO

BACKGROUND: Ventilation with higher PaCO(2) goals may reduce lung injury and bronchopulmonary dysplasia (BPD). The effect may be enhanced by using a higher PaCO(2) goal than in previous trials. OBJECTIVE: To determine the clinical benefits and safety of higher PaCO(2) goals for ventilated preterm infants. STUDY DESIGN: Preterm infants with a gestational age between 23 and 28 completed weeks receiving mechanical ventilation within 6 h of birth were randomized to be managed with either a PaCO(2) target between 55 and 65 mm Hg (7.3- 8.7 kPa, minimal ventilation) or 35 and 45 mm Hg (4.7- 6.0 kPa, routine ventilation) for the first 7 days of life. The primary outcome measure was BPD, defined as need for mechanical ventilation or supplemental oxygen at 36 weeks postmenstrual age, or death. The neurodevelopmental status was assessed at 18-22 months corrected age. RESULTS: The trial was stopped early after enrolling 31% of the projected sample size. Enrolled infants had a median birth weight of 640 g. BPD or death occurred in 21/33 (64%) infants after minimal ventilation and 19/32 (59%) infants after routine ventilation. Minimal ventilation was associated with trends towards higher mortality and higher incidence of neurodevelopmental impairment, and a significantly increased combined outcome of mental impairment or death (p < 0.05). CONCLUSION: Minimal ventilation as performed in this study did not improve clinical outcome, and may have been associated with a worse neurodevelopmental outcome.


Assuntos
Recém-Nascido Prematuro , Respiração Artificial/métodos , Síndrome do Desconforto Respiratório do Recém-Nascido/terapia , Algoritmos , Encéfalo/crescimento & desenvolvimento , Displasia Broncopulmonar/mortalidade , Displasia Broncopulmonar/prevenção & controle , Feminino , Idade Gestacional , Humanos , Recém-Nascido , Recém-Nascido de muito Baixo Peso , Masculino , Síndrome do Desconforto Respiratório do Recém-Nascido/mortalidade , Resultado do Tratamento
11.
J Pediatr ; 142(6): 656-61, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12838194

RESUMO

OBJECTIVE: Vitamin A supplementation reduces bronchopulmonary dysplasia (BPD)/death in extremely low birth weight neonates. It was hypothesized that compared with the standard regimen of 5000 IU 3 times per week for 4 weeks, (1) a higher dose (10,000 IU 3 x per week) would increase serum retinol and retinol binding protein (RBP) and lower relative dose responses (RDR), and (2) once-per-week dosing (15,000 IU once per week) would lead to equivalent levels, RBP, and RDR. STUDY DESIGN: Extremely low birth weight neonates (n = 120) receiving O(2)/mechanical ventilation at 24 hours were randomly assigned to (1) standard, (2) higher dose, or (3) once-per-week regimens. Measures of vitamin A deficiency were serum retinol <20 microg/dL, RBP <2.5 mg/dL, and/or RDR >10% on day 28. BPD was defined as O(2)/mechanical ventilation at 36 weeks' postmenstrual age. RESULTS: Groups were similar at enrollment (median gestational age, 25 weeks; birth weight, 689 g). Possible toxicity was seen in <5%. The higher dose regimen did not increase retinol or RBP, decrease RDR, or improve outcomes. Infants in the once-per-week regimen had lower retinol levels and higher RDR without an effect on outcomes. CONCLUSIONS: Compared with the standard regimen, once-per-week dosing worsened, and higher doses did not reduce, vitamin A deficiency. Therefore, the standard regimen is recommended.


Assuntos
Recém-Nascido de muito Baixo Peso , Vitamina A/administração & dosagem , Displasia Broncopulmonar/prevenção & controle , Feminino , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Doenças do Prematuro/prevenção & controle , Masculino , Proteínas de Ligação ao Retinol/análise , Vitamina A/sangue
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa