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










Base de dados
Intervalo de ano de publicação
1.
Pediatr Res ; 90(1): 131-139, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33753894

RESUMO

BACKGROUND: Extremely low gestational age newborns (ELGANs) are at risk of neurodevelopmental impairments that may originate in early NICU care. We hypothesized that early oxygen saturations (SpO2), arterial pO2 levels, and supplemental oxygen (FiO2) would associate with later neuroanatomic changes. METHODS: SpO2, arterial blood gases, and FiO2 from 73 ELGANs (GA 26.4 ± 1.2; BW 867 ± 179 g) during the first 3 postnatal days were correlated with later white matter injury (WM, MRI, n = 69), secondary cortical somatosensory processing in magnetoencephalography (MEG-SII, n = 39), Hempel neurological examination (n = 66), and developmental quotients of Griffiths Mental Developmental Scales (GMDS, n = 58). RESULTS: The ELGANs with later WM abnormalities exhibited lower SpO2 and pO2 levels, and higher FiO2 need during the first 3 days than those with normal WM. They also had higher pCO2 values. The infants with abnormal MEG-SII showed opposite findings, i.e., displayed higher SpO2 and pO2 levels and lower FiO2 need, than those with better outcomes. Severe WM changes and abnormal MEG-SII were correlated with adverse neurodevelopment. CONCLUSIONS: Low oxygen levels and high FiO2 need during the NICU care associate with WM abnormalities, whereas higher oxygen levels correlate with abnormal MEG-SII. The results may indicate certain brain structures being more vulnerable to hypoxia and others to hyperoxia, thus emphasizing the role of strict saturation targets. IMPACT: This study indicates that both abnormally low and high oxygen levels during early NICU care are harmful for later neurodevelopmental outcomes in preterm neonates. Specific brain structures seem to be vulnerable to low and others to high oxygen levels. The findings may have clinical implications as oxygen is one of the most common therapies given in NICUs. The results emphasize the role of strict saturation targets during the early postnatal period in preterm infants.


Assuntos
Lesões Encefálicas/etiologia , Hipóxia/complicações , Lactente Extremamente Prematuro , Lesões Encefálicas/diagnóstico por imagem , Feminino , Idade Gestacional , Humanos , Recém-Nascido , Unidades de Terapia Intensiva Neonatal , Magnetoencefalografia , Masculino , Oximetria/métodos , Oxigênio/sangue , Oxigenoterapia
2.
Pediatr Res ; 89(5): 1253-1260, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-32663837

RESUMO

BACKGROUND: Antenatal glucocorticoids (GCs) reduce respiratory distress syndrome (RDS) in preterm infants and are associated with reduced lung liquid content. Our aim was to assess whether airway gene expression of mediators of pulmonary epithelial sodium and liquid absorption, and further, respiratory morbidity, associate with cord blood GC concentrations. METHODS: The study included 64 infants delivered <32 weeks gestation. Cortisol and betamethasone in umbilical cord blood were quantified with liquid chromatography-tandem mass spectrometry. The total GC concentration was calculated. Gene expression of the epithelial sodium channel (ENaC), Na,K-ATPase, and serum- and GC-inducible kinase 1 at <2 h and at 1 day postnatally in nasal epithelial cell samples was quantified with reverse transcription-polymerase chain reaction. The mean oxygen supplementation during the first 72 h was calculated. RESULTS: Concentrations of cord blood betamethasone and total GC were significantly lower in infants with RDS and correlated with mean oxygen supplementation. Expression of αENaC and α1- and ß1Na,K-ATPase at <2 h correlated with betamethasone and total GC concentrations. Expression of Na,K-ATPase was lower in infants with RDS. CONCLUSION: Enhancement of lung liquid absorption via increased expression of sodium transporters may contribute to the beneficial pulmonary effects of antenatal GCs. IMPACT: RDS is related to lower umbilical cord blood GC concentrations and lower airway expression of sodium transporters. In addition to the timing of antenatal GC treatment, resulting concentrations may be of importance in preventing RDS. Induction of sodium transport may be a factor contributing to the pulmonary response to antenatal GCs.


Assuntos
Betametasona/química , Glucocorticoides/metabolismo , Síndrome do Desconforto Respiratório do Recém-Nascido/fisiopatologia , Sódio/química , Transporte Biológico , Estudos Transversais , Canais Epiteliais de Sódio/genética , Feminino , Sangue Fetal/metabolismo , Perfilação da Expressão Gênica , Humanos , Lactente , Recém-Nascido , Recém-Nascido Prematuro , Masculino , Estudos Prospectivos , ATPase Trocadora de Sódio-Potássio/metabolismo
3.
Arterioscler Thromb Vasc Biol ; 28(2): 309-14, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18079408

RESUMO

OBJECTIVE: Activated mast cells (MCs) induce endothelial cell (EC) apoptosis in vitro and are present at sites of plaque erosions in vivo. To further elucidate the role of MCs in endothelial apoptosis and consequently in plaque erosion, we have studied the molecular mechanisms involved in MC-induced EC apoptosis. METHODS AND RESULTS: Primary cultures of rat cardiac microvascular ECs (RCMECs) and human coronary artery ECs (HCAECs) were treated either with rat MC releasate (ie, mediators released on MC activation), rat chymase and tumor necrosis factor-alpha (TNF-alpha), or with human chymase and TNF-alpha, respectively. MC releasate induced RCMEC apoptosis by inactivating the focal adhesion kinase (FAK) and Akt-dependent survival signaling pathway, and apoptosis was partially inhibited by chymase and TNF-alpha inhibitors. Chymase avidly degraded both vitronectin (VN) and fibronectin (FN) produced by the cultured RCMECs. In addition, MC releasate inhibited the activation of NF-kappaB (p65) and activated caspase-8 and -9. Moreover, in HCAECs, human chymase and TNF-alpha induced additive levels of apoptosis. CONCLUSIONS: Activated MCs induce EC apoptosis by multiple mechanisms: chymase inactivates the FAK-mediated cell survival signaling, and TNF-alpha triggers apoptosis. Thus, by inducing EC apoptosis, MCs may contribute to plaque erosion and complications of atherosclerosis.


Assuntos
Apoptose/fisiologia , Quimases/metabolismo , Células Endoteliais/fisiologia , Mastócitos/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Células Cultivadas , Vasos Coronários/citologia , Proteína-Tirosina Quinases de Adesão Focal/fisiologia , Humanos , Masculino , Proteínas Proto-Oncogênicas c-akt/fisiologia , Ratos , Ratos Wistar , Transdução de Sinais
4.
Coron Artery Dis ; 18(8): 663-7, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18004118

RESUMO

INTRODUCTION: Chemically modified tetracyclines (CMTs) are a group of nonantimicrobial derivatives of tetracycline, which exert antiproliferative and anticollagenolytic properties. The molecular mechanisms, however, are poorly understood. MATERIALS AND METHODS: The effect of CMT-3 on cultured, subconfluent rat aortic smooth muscle cells (SMCs) was analyzed by [(3)H]-thymidine incorporation, counting cell numbers, and flow cytometry analysis. RESULTS: CMT-3 inhibited the incorporation of [(3)H]-thymidine and reduced the cell number dose-dependently, with approximately 60% inhibition at the maximal CMT-3 concentration used (20 mumol/l). CMT-3 decreased the SMC proportion in S-phase and gradually increased the proportion at G2/M. Initially, the proportion of cells in G1-phase increased and then gradually decreased back to baseline as the CMT-3 concentration increased. CMT-3 treatment of confluent SMCs for 24 h did not induce apoptosis. CONCLUSIONS: CMT-3 inhibited SMC proliferation by inducing cell cycle arrest at the G2/M restriction point. Nonetheless, CMT-3 did not induce SMC apoptosis.


Assuntos
Proliferação de Células/efeitos dos fármacos , Músculo Liso Vascular/efeitos dos fármacos , Tetraciclina/farmacologia , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Replicação do DNA , Citometria de Fluxo , Masculino , Músculo Liso Vascular/citologia , Ratos , Ratos Wistar
5.
Exp Cell Res ; 312(8): 1289-98, 2006 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-16460729

RESUMO

Chymase released from activated mast cells induces apoptosis of vascular smooth muscle cells (SMCs) in vitro by degrading the pericellular matrix component fibronectin, so causing disruption of focal adhesion complexes and Akt dephosphorylation, which are necessary for cell adhesion and survival. However, the molecular mechanisms of chymase-mediated apoptosis downstream of Akt have remained elusive. Here, we show by means of RT-PCR, Western blotting, EMSA, immunocytochemistry and confocal microscopy, that chymase induces SMC apoptosis by disrupting NF-kappaB-mediated survival signaling. Following chymase treatment, the translocation of active NF-kappaB/p65 to the nucleus was partly abolished and the amount of nuclear p65 was reduced. Pretreatment of SMCs with chymase also inhibited LPS- and IL-1beta-induced nuclear translocation of p65. The chymase-induced degradation of p65 was mediated by active caspases. Loss of NF-kappaB-mediated transactivation resulted in downregulation of bcl-2 mRNA and protein expression, leading to mitochondrial swelling and release of cytochrome c. The apoptotic process involved activation of both caspase 9 and caspase 8. The results reveal that, by disrupting the NF-kappaB-mediated survival-signaling pathway, activated chymase-secreting mast cells can mediate apoptosis of cultured arterial SMCs. Since activated mast cells colocalize with apoptotic SMCs in vulnerable areas of human atherosclerotic plaques, they may participate in the weakening and rupture of atherosclerotic plaques.


Assuntos
Apoptose/fisiologia , Mastócitos/enzimologia , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , NF-kappa B/metabolismo , Serina Endopeptidases/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Animais , Apoptose/efeitos dos fármacos , Aterosclerose/enzimologia , Aterosclerose/fisiopatologia , Caspases/efeitos dos fármacos , Caspases/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Quimases , Citocromos c/metabolismo , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Mediadores da Inflamação/farmacologia , Masculino , Mastócitos/metabolismo , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , NF-kappa B/efeitos dos fármacos , NF-kappa B/genética , Proteínas Proto-Oncogênicas c-bcl-2/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Serina Endopeptidases/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Fator de Transcrição RelA/efeitos dos fármacos , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo , Ativação Transcricional/efeitos dos fármacos , Ativação Transcricional/fisiologia
6.
Arterioscler Thromb Vasc Biol ; 24(8): 1350-8, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15191939

RESUMO

The 2 major general concepts about the cell biology of atherogenesis, growth of smooth muscle cells, and lipid accumulation in macrophages, ie, foam cell formation, have not been able to satisfactorily explain the genesis of acute coronary syndromes. Rather, the basic pathology behind the acute atherothrombotic events relates to erosion and rupture of unstable coronary plaques. At the cellular level, we now understand that a switch from cellular growth to cellular death, notably apoptosis, could be involved in turning at least some types of atherosclerotic plaques unstable. Because intimal cells require a proper matrix environment for normal function and survival, the vulnerability of an atherosclerotic plaque may critically depend on the integrity of the pericellular matrix of the plaque cells. In vitro studies have revealed that plaque-infiltrating inflammatory cells, such as macrophages, T-lymphocytes, and mast cells, by secreting a variety of proteases capable of degrading pericellular matrix components, induce death of endothelial cells and smooth muscle cells, and so provide a mechanistic explanation for inflammation-dependent plaque erosion and rupture. Thus, a novel link between inflammation and acute coronary syndromes is emerging. For a more explicit understanding of the role of proteases released by inflammatory cells in the conversion of a clinically silent plaque into a dangerous and potentially killing plaque, animal models of plaque erosion and rupture need to be established.


Assuntos
Arteriosclerose/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Peptídeo Hidrolases/fisiologia , Animais , Apoptose , Arteriosclerose/etiologia , Arteriosclerose/patologia , Vasos Sanguíneos/lesões , Adesão Celular , Fibrose , Humanos , Hipercolesterolemia/complicações , Células Matadoras Naturais/fisiologia , Macrófagos/fisiologia , Mastócitos/fisiologia , Modelos Cardiovasculares , Neutrófilos/fisiologia , Fator de Crescimento Derivado de Plaquetas/fisiologia , Coelhos , Ruptura Espontânea , Transdução de Sinais , Linfócitos T Citotóxicos/fisiologia
7.
Biochem Pharmacol ; 66(8): 1493-8, 2003 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-14555226

RESUMO

The fibrous cap of a lipid-containing atherosclerotic plaque consists of collagen produced by arterial smooth muscle cells (SMCs) of synthetic phenotype. A thick cap protects the lipid-rich core, whereas a thin cap predisposes it to rupture, with ensuing acute clinical complications, such as myocardial infarction. Among the pathological mechanisms leading to plaque weakening and rupture, one possibility is loss of the matrix-synthesizing SMCs. Indeed, caps of ruptured coronary plaques contain a reduced number of SMCs. In contrast, in such lesions, the number of activated inflammatory cells, such as mast cells, is increased, suggesting that they may regulate the SMC number. We have shown that heparin proteoglycans secreted by activated mast cells can efficiently inhibit proliferation of SMCs in vitro and reduce their ability to produce collagen. Chymase, a neutral serine protease secreted by activated mast cells, can also inhibit SMC-mediated collagen synthesis by a transforming growth factor-beta-dependent and -independent mechanism, and moreover, cause degradation of the collagen matrix by activating latent interstitial collagenase (MMP-1). Furthermore, chymase can induce SMC apoptosis by degrading the extracellular matrix component fibronectin necessary for SMC adhesion, with subsequent disruption of focal adhesions and loss of outside-in survival signaling. Thus, activated mast cells may participate in the weakening and rupture of atherosclerotic plaques by secreting mediators, such as heparin proteoglycans and chymase, which affect the growth, function and death of arterial SMCs.


Assuntos
Apoptose , Arteriosclerose/patologia , Mastócitos/patologia , Músculo Liso Vascular/patologia , Ruptura/etiologia , Animais , Arteriosclerose/complicações , Divisão Celular , Matriz Extracelular , Humanos , Inflamação/etiologia
8.
Arterioscler Thromb Vasc Biol ; 23(2): 238-43, 2003 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-12588765

RESUMO

OBJECTIVE: Chymase released from activated mast cells has been shown to induce apoptosis of vascular smooth muscle cells (SMCs) in vitro. The proteolytic activity of chymase is essential for the proapoptotic effect, but the mechanism of chymase-induced apoptosis has remained unknown. METHODS AND RESULTS: Here we show by means of FACS analysis, immunohistochemistry, and Western blotting that mast cell-derived chymase induces SMC apoptosis by a mechanism involving degradation of an extracellular matrix component, fibronectin (FN), with subsequent disruption of focal adhesions. The FN degradation products induced SMC apoptosis of similar magnitude and with similar changes in outside-in signaling, as did chymase. Sodium orthovanadate, an inhibitor of tyrosine phosphatases, inhibited the chymase-induced SMC apoptosis. Focal adhesion kinase (FAK), one of the key mediators of integrin-extracellular matrix interactions and cell survival, was rapidly degraded in the presence of chymase or FN degradation products. Loss of phosphorylated FAK (p-FAK) resulted in a rapid dephosphorylation of the p-FAK-dependent downstream mediator Akt. CONCLUSIONS: The results suggest that chymase-secreting mast cells can mediate apoptosis of neighboring SMCs through a mechanism involving degradation of pericellular FN and disruption of the p-FAK-dependent cell-survival signaling cascade.


Assuntos
Apoptose/fisiologia , Fibronectinas/metabolismo , Adesões Focais/metabolismo , Mastócitos/enzimologia , Mastócitos/fisiologia , Músculo Liso Vascular/citologia , Músculo Liso Vascular/fisiologia , Serina Endopeptidases/fisiologia , Animais , Aorta/citologia , Sobrevivência Celular/fisiologia , Células Cultivadas , Quimases , Masculino , Ratos , Ratos Wistar , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...