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.
J Am Heart Assoc ; 3(1): e000471, 2014 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-24419735

RESUMO

BACKGROUND: The core region of a myocardial infarction is notoriously unsupportive of cardiomyocyte survival. However, there has been less investigation of the potentially beneficial spontaneous recruitment of endogenous bone marrow progenitor cells (BMPCs) within infarcted areas. In the current study we examined the role of tissue oxygenation and derived toxic species in the control of BMPC engraftment during postinfarction heart remodeling. METHODS AND RESULTS: For assessment of cellular origin, local oxygenation, redox status, and fate of cells in the infarcted region, myocardial infarction in mice with or without LacZ(+) bone marrow transplantation was induced by coronary ligation. Sham-operated mice served as controls. After 1 week, LacZ(+) BMPC-derived cells were found inhomogeneously distributed into the infarct zone, with a lower density at its core. Electron paramagnetic resonance (EPR) oximetry showed that pO2 in the infarct recovered starting on day 2 post-myocardial infarction, concomitant with wall thinning and erythrocytes percolating through muscle microruptures. Paralleling this reoxygenation, increased generation of reactive oxygen/nitrogen species was detected at the infarct core. This process delineated a zone of diminished BMPC engraftment, and at 1 week infiltrating cells displayed immunoreactive 3-nitrotyrosine and apoptosis. In vivo treatment with a superoxide dismutase mimetic significantly reduced reactive oxygen species formation and amplified BMPC accumulation. This treatment also salvaged wall thickness by 43% and left ventricular ejection fraction by 27%, with significantly increased animal survival. CONCLUSIONS: BMPC engraftment in the infarct inversely mirrored the distribution of reactive oxygen/nitrogen species. Antioxidant treatment resulted in increased numbers of engrafted BMPCs, provided functional protection to the heart, and decreased the incidence of myocardial rupture and death.


Assuntos
Células da Medula Óssea/metabolismo , Infarto do Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Espécies Reativas de Nitrogênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Células-Tronco/metabolismo , Remodelação Ventricular , Animais , Antioxidantes/farmacologia , Apoptose , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/patologia , Transplante de Medula Óssea , Diferenciação Celular , Movimento Celular , Modelos Animais de Doenças , Óperon Lac , Masculino , Camundongos , Camundongos Transgênicos , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Transplante de Células-Tronco , Células-Tronco/efeitos dos fármacos , Células-Tronco/patologia , Fatores de Tempo , Tirosina/análogos & derivados , Tirosina/metabolismo , Remodelação Ventricular/efeitos dos fármacos
2.
Biochem Biophys Res Commun ; 409(3): 412-7, 2011 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-21575599

RESUMO

Systemic cell-free hemoglobin (Hb) released via hemolysis disrupts vascular homeostasis, in part, through the scavenging of nitric oxide (NO). Sodium nitrite (NaNO(2)) therapy can attenuate the hypertensive effects of Hb. However, the chemical reactivity of NaNO(2) with Hb may enhance heme- or iron-mediated toxicities. Here, we investigate the effect of NaNO(2) on the central nervous system (CNS) in guinea pigs exposed to systemic cell-free Hb. Intravascular infusion of NaNO(2), at doses sufficient to alleviate Hb-mediated blood pressure changes, reduced the expression of occludin, but not zona occludens-1 (ZO-1) or claudin-5, in cerebral tight junctions 4h after Hb infusion. This was accompanied by increased perivascular heme oxygenase-1 expression, neuronal iron deposition, increased astrocyte and microglial activation, and reduced expression of neuron-specific nuclear protein (NeuN). These CNS changes were not observed in animals treated with Hb or NaNO(2) alone. Taken together, these findings suggest that the use of nitrite salts to treat systemic Hb exposure may promote acute CNS toxicity.


Assuntos
Doenças do Sistema Nervoso Central/induzido quimicamente , Sistema Nervoso Central/efeitos dos fármacos , Hemoglobinas/metabolismo , Hemólise , Nitrito de Sódio/toxicidade , Animais , Sistema Nervoso Central/metabolismo , Doenças do Sistema Nervoso Central/metabolismo , Claudinas/metabolismo , Cobaias , Humanos , Masculino , Proteínas de Membrana/metabolismo , Ocludina , Oxirredução , Fosfoproteínas/metabolismo , Proteína da Zônula de Oclusão-1
3.
Am J Pathol ; 178(3): 1316-28, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21356382

RESUMO

Systemic exposure to cell-free hemoglobin (Hb) or its breakdown products after hemolysis or with the use of Hb-based oxygen therapeutics may alter the function and integrity of the blood-brain barrier. Using a guinea pig exchange transfusion model, we investigated the effect of a polymerized cell-free Hb (HbG) on the expression of endothelial tight junction proteins (zonula occludens 1, claudin-5, and occludin), astrocyte activation, IgG extravasation, heme oxygenase (HO), iron deposition, oxidative end products (4-hydroxynonenal adducts and 8-hydroxydeoxyguanosine), and apoptosis (cleaved caspase 3). Reduced zonula occludens 1 expression was observed after HbG transfusion as evidenced by Western blot and confocal microscopy. Claudin-5 distribution was altered in small- to medium-sized vessels. However, total expression of claudin-5 and occludin remained unchanged except for a notable increase in occludin 72 hours after HbG transfusion. HbG-transfused animals also showed increased astrocytic glial fibrillary acidic protein expression and IgG extravasation after 72 hours. Increased HO activity and HO-1 expression with prominent enhancement of HO-1 immunoreactivity in CD163-expressing perivascular cells and infiltrating monocytes/macrophages were also observed. Consistent with oxidative stress, HbG increased iron deposition, 4-hydroxynonenal and 8-hydroxydeoxyguanosine immunoreactivity, and cleaved caspase-3 expression. Systemic exposure to an extracellular Hb triggers blood-brain barrier disruption and oxidative stress, which may have important implications for the use of Hb-based therapeutics and may provide indirect insight on the central nervous system vasculopathies associated with excessive hemolysis.


Assuntos
Barreira Hematoencefálica/patologia , Hemoglobinas/administração & dosagem , Hemoglobinas/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Astrócitos/efeitos dos fármacos , Astrócitos/patologia , Transfusão de Sangue , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/enzimologia , Caspase 3/metabolismo , Sistema Livre de Células/efeitos dos fármacos , Indução Enzimática/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/metabolismo , Cobaias , Heme/metabolismo , Heme Oxigenase (Desciclizante)/biossíntese , Imunoglobulina G/metabolismo , Ferro/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/metabolismo
4.
Antioxid Redox Signal ; 12(2): 199-208, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19659432

RESUMO

Abstract Heme catabolism and iron sequestration systems play an important role in regulating the response to extracellular hemoglobin (Hb). We previously reported that extracellular Hb oxidizes more readily in the circulation of guinea pigs, a nonascorbate (AA)-producing species with similar plasma and tissue antioxidant status to humans, compared to rats, an AA-producing species. To determine whether these two species exhibit differences in heme catabolism and iron sequestration at the level of the kidney, we examined heme oxygenase (HO), H- and L-ferritin expression, nonheme iron deposition, and renal AA content following transfusion with polymerized bovine hemoglobin (HbG). Both species showed similar rates of hemoglobinuria but urinary HbG was significantly more oxidized in guinea pigs. HbG enhanced HO activity in both species but appeared greater and more sustained in guinea pigs. Conversely, rats showed a greater and more rapid induction of H- and L-ferritin as well as greater iron accumulation and AA content. Furthermore, ferrous and ferric iron deposits were detected in rats while only ferric iron was observed in guinea pigs. These findings suggest significant differences in the renal handling of HbG which may be important for understanding how endogenous antioxidant defenses may modulate the renal response to extracellular Hb.


Assuntos
Ácido Ascórbico/metabolismo , Ferritinas/metabolismo , Heme Oxigenase (Desciclizante)/metabolismo , Hemoglobinas/metabolismo , Rim/enzimologia , Rim/metabolismo , Animais , Transfusão de Componentes Sanguíneos , Western Blotting , Bovinos , Creatinina/sangue , Cobaias , Heme Oxigenase-1/metabolismo , Hemoglobinas/administração & dosagem , Hemoglobinúria/sangue , Ferro/metabolismo , Masculino , Polímeros/metabolismo , Ratos , Ratos Sprague-Dawley , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
5.
J Clin Invest ; 119(8): 2271-80, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19620788

RESUMO

Release of hemoglobin (Hb) into the circulation is a central pathophysiologic event that contributes to morbidity and mortality in chronic hemolytic anemias and severe malaria. These toxicities arise from Hb-mediated vasoactivity, possibly due to NO scavenging and localized tissue oxidative processes. Currently, there is no established treatment that targets circulating extracellular Hb. Here, we assessed the role of haptoglobin (Hp), the primary scavenger of Hb in the circulation, in limiting the toxicity of cell-free Hb infusion. Using a canine model, we found that glucocorticoid stimulation of endogenous Hp synthesis prevented Hb-induced hemodynamic responses. Furthermore, guinea pigs administered exogenous Hp displayed decreased Hb-induced hypertension and oxidative toxicity to extravascular environments, such as the proximal tubules of the kidney. The ability of Hp to both attenuate hypertensive responses during Hb exposure and prevent peroxidative toxicity in extravascular compartments was dependent on Hb-Hp complex formation, which likely acts through sequestration of Hb rather than modulation of its NO- and O2-binding characteristics. Our data therefore suggest that therapies involving supplementation of endogenous Hb scavengers may be able to treat complications of acute and chronic hemolysis, as well as counter the adverse effects associated with Hb-based oxygen therapeutics.


Assuntos
Haptoglobinas/farmacologia , Hemoglobinas/metabolismo , Hipertensão/prevenção & controle , Animais , Cães , Feminino , Glucocorticoides/farmacologia , Cobaias , Haptoglobinas/metabolismo , Hemoglobinas/toxicidade , Ferro/metabolismo , Masculino , Óxido Nítrico/metabolismo , Estresse Oxidativo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA