Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Am J Physiol Lung Cell Mol Physiol ; 325(3): L360-L367, 2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-37431589

RESUMO

Bronchopulmonary dysplasia (BPD) is the most common chronic lung disease in preterm infants, and pulmonary hypertension (PH) develops in 25%-40% of patients with BPD, increasing morbidity and mortality. BPD-PH is characterized by vasoconstriction and vascular remodeling. Nitric oxide (NO) is a pulmonary vasodilator and apoptotic mediator made in the pulmonary endothelium by NO synthase (eNOS). Asymmetric dimethylarginine (ADMA) is an endogenous eNOS inhibitor, primarily metabolized by dimethylarginine dimethylaminohydrolase-1 (DDAH1). Our hypothesis is that DDAH1 knockdown in human pulmonary microvascular endothelial cells (hPMVEC) will result in lower NO production, decreased apoptosis, and greater proliferation of human pulmonary arterial smooth muscle cells (hPASMC), whereas DDAH1 overexpression will have the opposite effect. hPMVECs were transfected with small interfering RNA targeting DDAH1 (siDDAH1)/scramble or adenoviral vector containing DDAH1 (AdDDAH1)/AdGFP for 24 h and co-cultured for 24 h with hPASMC. Analyses included Western blot for cleaved and total caspase-3, caspase-8, caspase-9, ß-actin; trypan blue exclusion for viable cell numbers; terminal deoxynucleotide transferase dUTP nick end labeling (TUNEL); and BrdU incorporation. Small interfering RNA targeting DDAH1 (siDDAH1) transfected into hPMVEC resulted in lower media nitrites, cleaved caspase-3 and caspase-8 protein expression, and TUNEL staining; and greater viable cell numbers and BrdU incorporation in co-cultured hPASMC. Adenoviral-mediated transfection of the DDAH1 gene (AdDDAH1) into hPMVEC resulted in greater cleaved caspase-3 and caspase-8 protein expression and lower viable cell numbers in co-cultured hPASMC. Partial recovery of hPASMC viable cell numbers after AdDDAH1-hPMVEC transfection was observed when media were treated with hemoglobin to sequester NO. In conclusion, hPMVEC-DDAH1-mediated NO production positively regulates hPASMC apoptosis, which may prevent/attenuate aberrant pulmonary vascular proliferation/remodeling in BPD-PH.NEW & NOTEWORTHY BPD-PH is characterized by vascular remodeling. NO is an apoptotic mediator made in the pulmonary endothelium by eNOS. ADMA is an endogenous eNOS inhibitor metabolized by DDAH1. EC-DDAH1 overexpression resulted in greater cleaved caspase-3 and caspase-8 protein expression and lower viable cell numbers in co-cultured SMC. After NO sequestration, SMC viable cell numbers partially recovered despite EC-DDAH1 overexpression. EC-DDAH1-mediated NO production positively regulates SMC apoptosis, which may prevent/attenuate aberrant pulmonary vascular proliferation/remodeling in BPD-PH.


Assuntos
Displasia Broncopulmonar , Hipertensão Pulmonar , Lactente , Humanos , Recém-Nascido , Óxido Nítrico/metabolismo , Caspase 3/metabolismo , Caspase 8/metabolismo , Células Endoteliais/metabolismo , Técnicas de Cocultura , Remodelação Vascular , Bromodesoxiuridina , Recém-Nascido Prematuro , Hipertensão Pulmonar/metabolismo , Arginina/metabolismo , RNA Interferente Pequeno , Apoptose , Miócitos de Músculo Liso/metabolismo
2.
J Neonatal Perinatal Med ; 15(1): 113-121, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34151866

RESUMO

BACKGROUND: Bronchopulmonary dysplasia (BPD) is chronic lung disease of prematurity and pulmonary hypertension (PH) is a major contributor to morbidity and mortality in BPD patients. Nitric oxide (NO) is a vasodilator and apoptotic mediator made by NO synthase (NOS). NOS is inhibited by asymmetric dimethylarginine (ADMA), and dimethylarginine dimethylaminohydrolase (DDAH) hydrolyzes ADMA. Previously, in a BPD patient cohort, we identified single nucleotide polymorphism (SNP) DDAH1 rs480414 (G > A) that was protective against developing PH. This study aims to determine functional consequences of the DDAH1 SNP in lymphoblastoid cell lines (LCLs) derived from neonatal cord blood. We tested the hypothesis that DDAH1 SNP (AA) results in DDAH1 gain of function, leading to greater NO-mediated apoptosis compared to DDAH1 wild-type (GG) in LCLs. METHODS: LCLs were analyzed by Western blot (DDAH1, cleaved and total caspase-3 and -8, and ß-actin), and RT-PCR (DDAH1, iNOS). Cell media assayed for nitrites with chemiluminescence NO analyzer, and conversion of ADMA to L-citrulline was measured by spectrophotometry. RESULTS: LCLs with DDAH1 SNP had similar levels of DDAH1 protein and mRNA expression, as well as DDAH activity, compared to DDAH1 WT LCLs. There were also no changes in cleaved caspase-3 and -8 protein levels. LCLs with DDAH1 SNP had similar iNOS mRNA expression. Nitrite levels in media were lower for DDAH1 SNP LCLs compared to DDAH1 WT LCLs (p < 0.05). CONCLUSION: Contrary to our hypothesis, we found that NO production was lower in DDAH1 SNP LCLs, indicative of a loss of function phenotype.


Assuntos
Displasia Broncopulmonar , Hipertensão Pulmonar , Displasia Broncopulmonar/genética , Linhagem Celular , Sangue Fetal , Humanos , Hipertensão Pulmonar/genética , Hipertensão Pulmonar/prevenção & controle , Recém-Nascido , Óxido Nítrico , Polimorfismo de Nucleotídeo Único
3.
Pediatr Res ; 92(3): 631-636, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-34795389

RESUMO

Pulmonary artery acceleration time (PAT) and PAT: ejection time (PATET) ratio are echocardiographic measurements of pulmonary arterial hypertension (PAH). These noninvasive quantitative measurements are ideal to follow longitudinally through the clinical course of PAH, especially as it relates to the need for and/or response to treatment. This review article focuses on the current literature of PATET measurement for infants and children as it relates to the shortening of the PATET ratio in PAH. At the same time, further development of PATET as an outcome measure for PAH in preclinical models, particularly mice, such that the field can move forward to human clinical studies that are both safe and effective. Here, we present what is known about PATET in infants and children and discuss what is known in preclinical models with particular emphasis on neonatal mouse models. In both animal models and human disease, PATET allows for longitudinal measurements in the same individual, leading to more precise determinations of disease/model progression and/or response to therapy. IMPACT: PATET ratio is a quantitative measurement by a noninvasive technique, Doppler echocardiography, providing clinicians a more precise/accurate, safe, and longitudinal assessment of pediatric PAH. We present a brief history/state of the art of PATET ratio to predict PAH in adults, children, infants, and fetuses, as well as in small animal models of PAH. In a preliminary study, PATET shortened by 18% during acute hypoxic exposure compared to pre-hypoxia. Studies are needed to establish PATET, especially in mouse models of disease, such as bronchopulmonary, as a routine measure of PAH.


Assuntos
Hipertensão Pulmonar , Hipertensão Arterial Pulmonar , Adulto , Animais , Criança , Ecocardiografia , Ecocardiografia Doppler/métodos , Humanos , Hipertensão Pulmonar/diagnóstico por imagem , Lactente , Camundongos , Artéria Pulmonar/diagnóstico por imagem
4.
Physiol Rep ; 7(12): e14150, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31209995

RESUMO

Nitric Oxide (NO) is an endogenous pulmonary vasodilator produced by endothelial NO synthase (eNOS). Asymmetric dimethyl L-arginine (ADMA) is an endogenous inhibitor of eNOS activity. In endothelial cells, ADMA is hydrolyzed to L-citrulline primarily by dimethylarginine dimethyl-aminohydrolase-1 (DDAH1). We tested the hypothesis that DDAH1 expression is essential for maintaining NO production in human fetal pulmonary microvascular endothelial cells (hfPMVEC), such that knockdown of DDAH1 expression will lead to decreased NO production resulting in less caspase-3 activation and less tube formation. We found that hfPMVEC transfected with DDAH1 siRNA had lower NO production than control, with no difference in eNOS protein levels between groups. hfPMVEC transfected with DDAH1 siRNA had lower protein levels of cleaved caspase-3 and -8 than control. Both DDAH1 siRNA- and ADMA-treated hfPMVEC had greater numbers of viable cells than controls. Angiogenesis was assessed using tube formation assays in matrigel, and tube formation was lower after either DDAH1 siRNA transfection or ADMA treatment than controls. Addition of an NO donor restored cleaved caspase-3 and -8 protein levels after DDAH1 siRNA transfection in hfPMVEC to essentially the levels seen in scramble control. Addition of a putative caspase-3 inhibitor to DDAH1 siRNA transfected and NO-donor treated cells led to greater numbers of viable cells and far less angiogenesis than in any other group studied. We conclude that in hfPMVEC, DDAH1 is central to the regulation of NO-mediated caspase-3 activation and the resultant apoptosis and angiogenesis. Our findings suggest that DDAH1 may be a potential therapeutic target in pulmonary hypertensive disorders.


Assuntos
Amidoidrolases/fisiologia , Apoptose/fisiologia , Pulmão/irrigação sanguínea , Neovascularização Fisiológica/fisiologia , Amidoidrolases/genética , Caspase 3/metabolismo , Caspase 8/metabolismo , Células Endoteliais/citologia , Ativação Enzimática/fisiologia , Técnicas de Silenciamento de Genes , Humanos , Pulmão/embriologia , Microvasos/citologia , Óxido Nítrico/biossíntese , Doadores de Óxido Nítrico/farmacologia , RNA Interferente Pequeno/genética
5.
Clin Exp Pharmacol Physiol ; 45(6): 556-562, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29266319

RESUMO

The L-arginine/NO pathway is an important regulator of pulmonary hypertension, the leading cause of mortality in patients with the chronic lung disease of prematurity, bronchopulmonary dysplasia. L-arginine can be metabolized by NO synthase (NOS) to form L-citrulline and NO, a potent vasodilator. Alternatively, L-arginine can be metabolized by arginase to form urea and L-ornithine, a precursor to collagen and proline formation important in vascular remodelling. In the current study, we hypothesized that C3H/HeN mice exposed to prolonged hyperoxia would have increased arginase expression and pulmonary vascular wall cell proliferation. C3H/HeN mice were exposed to 14 days of 85% O2 or room air and lung homogenates analyzed by western blot for protein levels of arginase I, arginase II, endothelial NOS (eNOS), ornithine decarboxylase (ODC), ornithine aminotransferase (OAT), and α-smooth muscle actin (α-SMA). Hyperoxia did not change arginase I or eNOS protein levels. However, arginase II protein levels were 15-fold greater after hyperoxia exposure than in lungs exposed to room air. Greater protein levels of ODC and OAT were found in lungs following hyperoxic exposure than in room air animals. α-SMA protein levels were found to be 7-fold greater in the hyperoxia exposed lungs than in room air lungs. In the hyperoxia exposed lungs there was evidence of greater pulmonary vascular wall cell proliferation by α-SMA immunohistochemistry than in room air lungs. Taken together, these data are consistent with a more proliferative vascular phenotype, and may explain the propensity of patients with bronchopulmonary dysplasia to develop pulmonary hypertension.


Assuntos
Actinas/metabolismo , Arginase/biossíntese , Displasia Broncopulmonar/complicações , Displasia Broncopulmonar/metabolismo , Hiperóxia/complicações , Animais , Displasia Broncopulmonar/patologia , Proliferação de Células , Modelos Animais de Doenças , Indução Enzimática , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Ornitina Descarboxilase/metabolismo , Ornitina-Oxo-Ácido Transaminase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...