Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Int J Mol Sci ; 24(10)2023 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-37240239

RESUMO

Silver nanoparticles are widely used in various industrial and biomedical applications; however, little is known about their potential cardiotoxicity after pulmonary exposure, particularly in hypertensive subjects. We assessed the cardiotoxicity of polyethylene glycol (PEG)-coated AgNPs in hypertensive (HT) mice. Saline (control) or PEG-AgNPs (0.5 mg/kg) were intratracheally (i.t.) instilled four times (on days 7, 14, 21, and 28 post-angiotensin II or vehicle [saline] infusion). On day 29, various cardiovascular parameters were evaluated. Systolic blood pressure and heart rate were higher in PEG-AgNPs-treated HT mice than in saline-treated HT or PEG-AgNPs-treated normotensive mice. The heart histology of PEG-AgNPs-treated HT mice had comparatively larger cardiomyocyte damage with fibrosis and inflammatory cells when compared with saline-treated HT mice. Similarly, the relative heart weight and the activities of lactate dehydrogenase and creatine kinase-MB and the concentration of brain natriuretic peptide concentration were significantly augmented in heart homogenates of HT mice treated with PEG-AgNPs compared with HT mice treated with saline or normotensive animals exposed to PEG-AgNPs. Similarly, the concentrations of endothelin-1, P-selectin, vascular cell adhesion molecule-1, and intercellular adhesion molecule-1 in heart homogenates were significantly higher than in the other two groups when HT mice were exposed to PEG-AgNPs. Markers of inflammation and oxidative and nitrosative stress were significantly elevated in heart homogenates of HT mice given PEG-AgNPs compared with HT mice treated with saline or normotensive animals exposed to PEG-AgNPs. The hearts of HT mice exposed to PEG-AgNPs had significantly increased DNA damage than those of HT mice treated with saline or normotensive mice treated with AgNPs. In conclusion, the cardiac injury caused by PEG-AgNPs was aggravated in hypertensive mice. The cardiotoxicity of PEG-AgNPs in HT mice highlights the importance of an in-depth assessment of their toxicity before using them in clinical settings, particularly in patients with pre-existing cardiovascular diseases.


Assuntos
Hipertensão , Nanopartículas Metálicas , Animais , Camundongos , Pressão Sanguínea , Prata/farmacologia , Nanopartículas Metálicas/toxicidade , Cardiotoxicidade , Polietilenoglicóis , Hipertensão/induzido quimicamente
2.
Toxicol Appl Pharmacol ; 367: 36-50, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30639276

RESUMO

Pulmonary exposure to silver nanoparticles (AgNPs) revealed the potential of nanoparticles to cause pulmonary toxicity, cross the alveolar-capillary barrier, and distribute to remote organs. However, the mechanism underlying the effects of AgNPs on the cardiovascular system remains unclear. Hence, we investigated the cardiovascular mechanisms of pulmonary exposure to AgNPs (10 nm) with varying coatings [polyvinylpyrrolidone (PVP) and citrate (CT)], concentrations (0.05, 0.5 and 5 mg/kg body weight), and time points (1 and 7 days) in BALB/C mice. Silver ions (Ag+) were used as ionic control. Exposure to AgNPs induced lung inflammation. In heart, tumor necrosis factor α, interleukin 6, total antioxidants, reduced glutathione and 8-isoprostane significantly increased for both AgNPs. Moreover, AgNPs caused oxidative DNA damage and apoptosis in the heart. The plasma concentration of fibrinogen, plasminogen activation inhibitor-1 and brain natriuretic peptide were significantly increased for both coating AgNPs. Likewise, the prothrombin time and activated partial thromboplastin time were significantly decreased. Additionally, the PVP- and CT- AgNPs induced a significant dose-dependent increase in thrombotic occlusion time in cerebral microvessels at both time points. In vitro study on mice whole blood exhibited significant platelet aggregation for both particle types. Compared with AgNPs, Ag+ increased thrombogenicity and markers of oxidative stress, but did not induce either DNA damage or apoptosis in the heart. In conclusion, pulmonary exposure to AgNPs caused cardiac oxidative stress, DNA damage and apoptosis, alteration of coagulation markers and thrombosis. Our findings provide a novel mechanistic insight into the cardiovascular pathophysiological effects of lung exposure to AgNPs.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Ácido Cítrico/toxicidade , Cardiopatias/induzido quimicamente , Trombose Intracraniana/induzido quimicamente , Nanopartículas Metálicas/toxicidade , Miócitos Cardíacos/efeitos dos fármacos , Povidona/toxicidade , Prata/toxicidade , Animais , Apoptose/efeitos dos fármacos , Cardiotoxicidade , Dano ao DNA , Relação Dose-Resposta a Droga , Feminino , Cardiopatias/metabolismo , Cardiopatias/patologia , Mediadores da Inflamação/metabolismo , Exposição por Inalação , Trombose Intracraniana/sangue , Masculino , Camundongos Endogâmicos BALB C , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Estresse Oxidativo/efeitos dos fármacos , Agregação Plaquetária/efeitos dos fármacos , Propriedades de Superfície , Fatores de Tempo
3.
Cell Physiol Biochem ; 49(4): 1577-1588, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30223265

RESUMO

BACKGROUND/AIMS: Silver nanoparticles (AgNPs) are increasingly used as antimicrobial agents and drug carriers in various biomedical fields. AgNPs can encounter erythrocytes either directly following intravenous injection, or indirectly via translocation from the site of administration. However, information regarding the pathophysiological effects and possible mechanism of action of AgNPs on the erythrocytes are still inadequately studied. Thus, the aim of our study was to investigate the mechanism underlying the effect of coating and concentration of AgNPs on mouse erythrocytes in vitro. METHODS: We studied the interaction of polyvinylpyrrolidone (PVP) and citrate (CT) coated AgNPs (10 nm) at various concentrations (2.5, 10, 40 µg/ml) with mouse erythrocytes in vitro using various techniques including transmission electron microscopy (TEM), hemolysis, and colorimetric measurement of markers of oxidative stress comprising malondialdehyde (MDA), reduced glutathione (GSH), and catalase (CAT). Intracellular calcium (Ca2+) was determined using Fura 2AM fluorescence. Annexin V was quantified using ELISA and the caspase 3 was determined both flurometrically and by western blot technique. RESULTS: Following incubation of the erythrocytes with AgNPs, both PVP- and CT- AgNPs induced significant and dose - dependent increase in hemolysis. TEM revealed that both PVP- and CT- AgNPs were taken up by erythrocytes. The erythrocyte susceptibility to lipid peroxidation measured by MDA was significantly increased in both PVP-and CT- AgNPs. The concentration of GSH and CAT activity were significantly decreased by both types of AgNPs. Additionally, PVP- and CT- AgNPs significantly increased intracellular Ca2+ in a dose -dependent manner. Likewise, the concentration of the cellular protein annexin V was significantly and dose - dependently enhanced by both types of AgNPs. Furthermore, PVP- and CT- AgNPs induced significant increase in calpain activity in incubated erythrocytes. CONCLUSION: We conclude that both PVP- and CT- AgNPs causes hemolysis, and are taken up by erythrocytes. Moreover, we demonstrated that AgNPs induces oxidative stress and eryptosis. These findings provide evidence for the potential pathophysiological effect of PVP-and CT- AgNPs on erythrocyte physiology.


Assuntos
Ácido Cítrico/química , Eriptose/efeitos dos fármacos , Nanopartículas Metálicas/química , Estresse Oxidativo/efeitos dos fármacos , Povidona/química , Prata/química , Animais , Cálcio/metabolismo , Caspase 3/metabolismo , Catalase/metabolismo , Eritrócitos/citologia , Eritrócitos/efeitos dos fármacos , Eritrócitos/metabolismo , Glutationa/metabolismo , Hemólise/efeitos dos fármacos , Peroxidação de Lipídeos/efeitos dos fármacos , Malondialdeído/metabolismo , Nanopartículas Metálicas/toxicidade , Camundongos Endogâmicos BALB C , Microscopia Eletrônica de Transmissão , Tamanho da Partícula , Ratos
4.
Oxid Med Cell Longev ; 2022: 2079630, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35111278

RESUMO

With advent of nanotechnology, silver nanoparticles, AgNPs owing majorly to their antibacterial properties, are used widely in food industry and biomedical applications implying human exposure by various routes including inhalation. Several reports have suggested AgNPs induced pathophysiological effects in a cardiovascular system. However, cardiovascular diseases such as hypertension may interfere with AgNPs-induced response, yet majority of them are understudied. The aim of this work was to evaluate the thrombotic complications in response to polyethylene glycol- (PEG-) coated AgNPs using an experimental hypertensive (HT) mouse model. Saline (control) or PEG-AgNPs (0.5 mg/kg) were intratracheally (i.t.) instilled four times, i.e., on days 7, 14, 21, and 28 post-angiotensin II-induced HT, or vehicle (saline) infusion. On day 29, various parameters were assessed including thrombosis in pial arterioles and venules, platelet aggregation in whole blood in vitro, plasma markers of coagulation, and fibrinolysis and systemic oxidative stress. Pulmonary exposure to PEG-AgNPs in HT mice induced an aggravation of in vivo thrombosis in pial arterioles and venules compared to normotensive (NT) mice exposed to PEG-AgNPs or HT mice given saline. The prothrombin time, activated partial thromboplastin time, and platelet aggregation in vitro were exacerbated after exposure to PEG-AgNPs in HT mice compared with either NT mice exposed to nanoparticles or HT mice exposed to saline. Elevated concentrations of fibrinogen, plasminogen activator inhibitor-1, and von Willebrand factor were seen after the exposure to PEG-AgNPs in HT mice compared with either PEG-AgNPs exposed NT mice or HT mice given with saline. Likewise, the plasma levels of superoxide dismutase and nitric oxide were augmented by PEG-AgNPs in HT mice compared with either NT mice exposed to nanoparticles or HT mice exposed to saline. Collectively, these results demonstrate that PEG-AgNPs can potentially exacerbate the in vivo and in vitro procoagulatory and oxidative stress effect in HT mice and suggest that population with hypertension are at higher risk of the toxicity of PEG-AgNPs.


Assuntos
Hipertensão/patologia , Nanopartículas Metálicas/toxicidade , Agregação Plaquetária/efeitos dos fármacos , Prata/química , Angiotensina II/efeitos adversos , Angiotensina II/farmacologia , Animais , Modelos Animais de Doenças , Feminino , Fibrinogênio/metabolismo , Hipertensão/etiologia , Masculino , Nanopartículas Metálicas/química , Camundongos , Camundongos Endogâmicos BALB C , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética , Tempo de Tromboplastina Parcial , Polietilenoglicóis/química , Tempo de Protrombina , Fator de von Willebrand/metabolismo
5.
Toxicology ; 222(3): 195-201, 2006 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-16584826

RESUMO

Particulate air pollution is associated with respiratory and cardiovascular morbidity and mortality. However, important uncertainties remain in the quantification of extrapulmonary translocation of ultrafine particles into blood circulation. Therefore, the widely used radioiodinated technique was applied to radiolabel polystyrene particles with an average diameter of 56.4 and 202 nm, respectively. The extrapulmonary distribution of these particles (3.7 x 10(5) Bq/rat) was quantified at 0.5, 2, 24 and 120 h after intratracheal instillation in rats. Moreover, we have taken into account the possible involvement of pulmonary inflammation in this process. Rats which received a single intratracheal instillation of free 125I or a single intravenous injection of labeled ultrafine particles served as control. The results indicated that the pulmonary deposition of radioactivity was almost unchanged for both sizes. Only small amounts of radioactivity (1.64-2.49%) were recovered in blood shortly after administration of both types of particle, in healthy rats. However, the extent of particle translocation into the blood of the ultrafine size following the pretreatment with lipopolysaccharides was significantly higher (from 1.96 +/- 0.67 to 4.73 +/- 0.31%) compared to larger particles (from 2.19 +/- 0.77 to 2.21 +/- 0.64%). In conclusion, our findings suggest that only a small fraction of intratracheal-instilled ultrafine particles can pass rapidly into systemic circulation, but this translocation is markedly increased following LPS pretreatment. Thus, pulmonary inflammation seems to play a major role in enhancing the extrapulmonary translocation of particles.


Assuntos
Lipopolissacarídeos/toxicidade , Pulmão/efeitos dos fármacos , Poliestirenos/farmacocinética , Administração por Inalação , Animais , Fluoracetatos , Injeções Intravenosas , Radioisótopos do Iodo , Pulmão/metabolismo , Pulmão/patologia , Masculino , Tamanho da Partícula , Pneumonia/metabolismo , Poliestirenos/administração & dosagem , Poliestirenos/sangue , Compostos Radiofarmacêuticos , Ratos , Ratos Sprague-Dawley , Tálio , Distribuição Tecidual , Ácido Trifluoracético
6.
Circulation ; 110(12): 1670-7, 2004 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-15364808

RESUMO

BACKGROUND: Particulate air pollution is associated with cardiovascular diseases and myocardial infarction (MI). METHODS AND RESULTS: We investigated the relationship between airway inflammation and thrombosis 24 hours after intratracheal (IT) instillation of diesel exhaust particles (DEP; 50 microg/hamster). Mild thrombosis was induced in the femoral vein by endothelial injury, and the consequences of airway inflammation on thrombogenicity were studied via online video microscopy. Lung inflammation and histamine analysis in bronchoalveolar lavage (BAL) and plasma were performed after pretreatment with dexamethasone (DEX) or sodium cromoglycate (SC). DEP induced airway inflammation and histamine release in BAL and in plasma, and increased thrombosis, without elevating plasma von Willebrand factor (vWF) levels. The IT instillation of 400-nm positively charged polystyrene particles (500 microg/hamster), serving as particles that do not penetrate into the circulation, equally produced airway inflammation, histamine release, and enhanced thrombosis. Histamine in plasma resulted from basophil activation. Intraperitoneal (IP) pretreatment with DEX (5 mg/kg) abolished the DEP-induced histamine increase in BAL and plasma and abrogated airway inflammation and thrombogenicity. The IT pretreatment with DEX (0.5 mg/kg) showed a partial but parallel inhibition of all of these parameters. Pretreatment with SC (40 mg/kg, IP) strongly inhibited airway inflammation, thrombogenicity, and histamine release. CONCLUSIONS: Our results are compatible with the triggering of mast cell degranulation and histamine release by DEP. Histamine plays an initial central role in airway inflammation, further release of histamine by circulating basophils, and peripheral thrombotic events. Antiinflammatory pretreatment can abrogate the peripheral thrombogenicity by preventing histamine release from mast cells.


Assuntos
Poluentes Atmosféricos/toxicidade , Anti-Inflamatórios/uso terapêutico , Cromolina Sódica/uso terapêutico , Dexametasona/uso terapêutico , Mastócitos/efeitos dos fármacos , Pneumonia/prevenção & controle , Trombose/prevenção & controle , Vasculite/prevenção & controle , Emissões de Veículos/toxicidade , Animais , Anti-Inflamatórios/farmacologia , Líquido da Lavagem Broncoalveolar/química , Cricetinae , Cromolina Sódica/farmacologia , Grânulos Citoplasmáticos/metabolismo , Dexametasona/farmacologia , Relação Dose-Resposta a Droga , Feminino , Histamina/análise , Liberação de Histamina/efeitos dos fármacos , Instilação de Medicamentos , Pulmão/patologia , Masculino , Mastócitos/metabolismo , Mastócitos/fisiologia , Mesocricetus , Microscopia de Vídeo , Especificidade de Órgãos , Tamanho da Partícula , Agregação Plaquetária/efeitos dos fármacos , Pneumonia/induzido quimicamente , Poliestirenos/toxicidade , Trombose/induzido quimicamente , Traqueia , Vasculite/induzido quimicamente , Fator de von Willebrand/análise
7.
Toxicology ; 208(1): 105-13, 2005 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-15664437

RESUMO

Particulate air pollution is associated with respiratory and cardiovascular morbidity and mortality. It has been suggested that ultrafine particles are able to translocate from the airways into the bloodstream in vivo. We have investigated this in an isolated perfused and ventilated rabbit lung preparation lacking pulmonary lymphatic flow. Fluorescent polystyrene particles of different diameters (24, 110 or 190 nm) and surface chemistry (carboxylate or amine modified) were injected either intratracheally (i.t.) or intravascularly (i.v.) and, after a period of 2 h, their presence in the perfusion liquid or in the bronchoalveolar lavage (BAL) fluid, was assessed by spectrofluorimetry. Vascular pressures and lung weights were monitored. Following the i.t. administration, no particle translocation was observed from the alveoli into the vascular compartment. Similarly, no particle translocation was found after i.v. administration of particles. However, when microvascular permeability was pharmacologically increased by administering histamine (10(-4) M) in the vascular compartment, inducing a positive driving force provided by fluid filtration, a fluorescent signal in BAL was recorded (2.5 +/- 1% of the dose of particles administered), suggesting a translocation of particles through the alveolo-capillary barrier. We conclude that ultrafine polystyrene particles cannot significantly diffuse from lung into the vascular compartment in our model, but they are able to translocate in the opposite direction when the microvascular permeability is increased by histamine. The relevance of these ex vivo findings for the in vivo translocation of inhaled ultrafine particles remains to be established.


Assuntos
Poluentes Atmosféricos/farmacocinética , Barreira Alveolocapilar/metabolismo , Permeabilidade Capilar/efeitos dos fármacos , Poliestirenos/farmacocinética , Poluentes Atmosféricos/química , Animais , Pressão Sanguínea/fisiologia , Líquido da Lavagem Broncoalveolar/química , Corantes Fluorescentes , Técnicas In Vitro , Pulmão/metabolismo , Pulmão/ultraestrutura , Modelos Biológicos , Tamanho da Partícula , Poliestirenos/química , Coelhos , Espectrometria de Fluorescência
8.
Toxicol Lett ; 149(1-3): 243-53, 2004 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15093270

RESUMO

Particulate air pollution is associated with cardiovascular morbidity and mortality. Fine particles with a diameter <2.5 microm (PM2.5) have an important role in triggering biological responses. These particles, and particularly the ultrafine fraction (<100 nm) penetrate deeply into the respiratory tract. Recently, we have demonstrated that ultrafine particles are able to translocate from the lung into the systemic circulation in hamsters and humans. In urban areas, diesel engines are considered to be the major source of PM2.5. We therefore evaluated the acute effect (1 h) of diesel exhaust particles (DEP) in a hamster model of peripheral vascular thrombosis induced by free-radical mediated endothelial injury, using intravenous Rose Bengal and local illumination. Intratracheal doses of 5-500 microg of DEP per animal induced inflammation with elevation of neutrophils, total proteins and histamine in bronchoalveolar lavage. DEP enhanced experimental arterial and venous platelet rich-thrombus formation in vivo. Blood samples taken from hamsters 30 and 60 min after instillation of DEP caused platelet activation, when analyzed in the Platelet Function Analyser (PFA-100). The direct addition of DEP to untreated hamster blood also caused platelet aggregation. These effects persisted up to 24 h after instillation. Our results provide plausible mechanistic explanations for the epidemiologically established link between air pollution and acute cardiovascular effects.


Assuntos
Poluentes Atmosféricos/toxicidade , Doenças Cardiovasculares/etiologia , Trombose/induzido quimicamente , Adulto , Poluentes Atmosféricos/farmacocinética , Animais , Líquido da Lavagem Broncoalveolar/citologia , Doenças Cardiovasculares/patologia , Doenças Cardiovasculares/fisiopatologia , Cricetinae , Humanos , Exposição por Inalação , Masculino , Pessoa de Meia-Idade , Neutrófilos/efeitos dos fármacos , Tamanho da Partícula , Testes de Função Plaquetária , Pneumonia/induzido quimicamente , Pneumonia/patologia , Poliestirenos , Compostos Radiofarmacêuticos , Pertecnetato Tc 99m de Sódio , Trombose/sangue , Emissões de Veículos/toxicidade
9.
Toxicol Appl Pharmacol ; 186(1): 38-45, 2003 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-12583991

RESUMO

Particulate air pollution is associated with cardiorespiratory effects and ultrafine particles (UFPs, diameter < 100 nm) are believed to play an important role. We studied the acute (1 h) effect of intratracheally instilled unmodified (60 nm), negatively charged carboxylate-modified (60 nm), or positively charged amine-modified (60 or 400 nm) polystyrene particles on bronchoalveolar lavage (BAL) indices and on peripheral thrombosis in hamster. The latter was assessed by measuring the extent of photochemically induced thrombosis in a femoral vein via transillumination. Unmodified and negative UFPs did not modify thrombosis and BAL indices. Positive UFPs increased thrombosis at 500 microg per animal (+ 341 +/- 96%) and at 50 microg per animal (+ 533 +/- 122%), but not at 5 microg per animal. Neutrophils, lactate dehydrogenase, and histamine were increased in BAL at all these doses but protein concentration was increased only at 500 microg per animal. Positive 400-nm particles (500 microg per animal) did not affect thrombosis, although they led to a neutrophil influx and an increase in BAL proteins and histamine. Using the Platelet Function Analyser (PFA-100), the platelets of hamsters were activated by the in vitro addition of positive UFPs and 400-nm particles to blood. We conclude that intratracheally administered positive ultrafine and 400-nm particles induce pulmonary inflammation within 1 h. Positive UFPs, but not the 400-nm particles enhance thrombosis. Hence, particle-induced lung inflammation and thrombogenesis can be partially uncoupled.


Assuntos
Poluentes Atmosféricos/toxicidade , Pneumonia/induzido quimicamente , Poliestirenos/toxicidade , Trombose/induzido quimicamente , Animais , Líquido da Lavagem Broncoalveolar/química , Cricetinae , Feminino , Intubação Intratraqueal , Masculino , Tamanho da Partícula , Pneumonia/patologia , Poliestirenos/administração & dosagem , Trombose/patologia
10.
Am J Respir Crit Care Med ; 166(7): 998-1004, 2002 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-12359661

RESUMO

Particulate air pollution is associated with cardiovascular morbidity and mortality. To investigate this association, we studied the effect of ultrafine (60 nm) polystyrene particles on thrombus formation in a hamster model after intravenous and intratracheal administration of unmodified, carboxylate-polystyrene, or amine-polystyrene particles. Unmodified particles had no effect on thrombosis up to 5 mg/kg. Carboxylate-polystyrene particles significantly inhibited thrombus formation at 500 and 100 microg/kg body weight but not at 50 microg/kg body weight. In contrast, amine-polystyrene particles significantly enhanced thrombosis at 500 and 50 microg/kg body weight but not at 5 microg/kg body weight. Similarly, the intratracheal instillation of 5,000 microg of amine-polystyrene particles significantly increased thrombus formation. The unmodified particles and carboxylate-polystyrene particles had no effect. During platelet aggregation in human platelet-rich plasma, induced with 1.25 microM ADP, unmodified particles had no effect up to 100 microg/ml, and carboxylate-polystyrene particles weakly enhanced platelet aggregation at 25 to 100 microg/ml. However, amine-polystyrene particles (50 and 100 microg/ml) induced platelet aggregation themselves and strongly increased the ADP-induced aggregation. We conclude that the presence of (ultrafine) particles in the circulation may affect hemostasis. The observed in vivo prothrombotic tendency results, at least in part, from platelet activation by positively charged amine-polystyrene particles.


Assuntos
Coloides/administração & dosagem , Cardiopatias/patologia , Silicones/administração & dosagem , Trombose/patologia , Difosfato de Adenosina/administração & dosagem , Animais , Coagulação Sanguínea/efeitos dos fármacos , Cricetinae , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Veia Femoral/patologia , Injeções Intravenosas , Microscopia Confocal , Microscopia Eletrônica , Tamanho da Partícula , Material Particulado , Agregação Plaquetária/efeitos dos fármacos , Poliestirenos/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA