Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Ecotoxicol Environ Saf ; 270: 115889, 2024 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-38150751

RESUMEN

Silica nanoparticles (SiNPs) have been widely used in electronics, chemistry, and biomedicine. Human exposure to SiNPs and possible health effects have attracted much attention. The potential cardiovascular toxicity of SiNPs and their related mechanisms are still unclear. Therefore, in this study, we investigated the toxic effects of SiNPs on human umbilical vein endothelial cells (HUVECs). We found that SiNPs could induce HUVECs ferroptosis. The results showed that the level of intracellular divalent iron and lipid peroxidation increased, and mitochondrial cristae decreased. In addition, the pretreatment of the iron chelator deferoxamine mesylate (DFO) could alleviate the ferroptosis of cells. Interestingly, pretreatment of 3-methyladenine (3-MA), an autophagy/PI3K inhibitor could partially inhibit autophagy and reduce ferroptosis, which indicated that autophagy played an important role in cell ferroptosis. Additionally, after knocking down nuclear receptor coactivator 4 (NCOA4), Ferritin Heavy Chain 1 (FTH1) expression was up-regulated, and the levels of divalent iron and lipid peroxidation decreased, which suggested that NCOA4 mediated the ferroptosis of HUVECs induced by SiNPs. In conclusion, this study shows that SiNPs can induce cardiovascular toxicity in which there is ferroptosis. NCOA4-mediated ferritinophagy and resultant ferroptosis by SiNPs may play an important role. This study provides a new theoretical strategy for the treatment and prevention of cardiovascular diseases in the future.


Asunto(s)
Ferroptosis , Nanopartículas , Humanos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Dióxido de Silicio/toxicidad , Fosfatidilinositol 3-Quinasas/metabolismo , Hierro/metabolismo , Factores de Transcripción/metabolismo , Nanopartículas/toxicidad , Autofagia , Coactivadores de Receptor Nuclear/genética , Coactivadores de Receptor Nuclear/metabolismo
2.
Int J Mol Med ; 52(1)2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37264973

RESUMEN

Nanoplastics (NPs) are a newly discovered type of environmental pollutant. The potential for neurotoxicity caused by NPs and their mechanisms are unclear. The present study aimed to determine the molecular mechanism underlying neurotoxicity induced by NPs. Microglia (BV2) cells were used for in vitro studies, and it was found that NPs invaded cells, activated inflammasomes, induced the release of significant quantities of inflammatory factors by detection of inflammatory response­associated proteins through Western blot and ELISA. By detection of FITC, SOD, GSH, cellular iron level, and ferroptosis­related proteins, it was found that NPs compromised the anti­oxidative mechanisms of cells, increased intracellular lipid peroxidation and Fe2+ concentration and triggered inflammatory reactions and ferroptosis. Pretreatment with reactive oxygen species (ROS) inhibitor N­acetylcysteine (NAC) alleviated induction of inflammatory reactions and ferroptosis of cells. In addition, inhibiting expression of c­Jun N­terminal kinase (JNK) increased expression of heme oxygenase (HO­1), resulting in decreased ferroptosis, indicating that the JNK/HO­1 signaling pathway was involved in NP­induced effects on ferroptosis in BV2 cells. In conclusion, NPs could induce inflammatory responses and ferroptosis in BV2 cells. JNK/HO­1 mediated ferroptosis may serve an important role in the toxicity of microglia induced by NPs. This study provided novel evidence for the toxic effects of NPs and highlighted a theoretical mechanistic basis for safe prevention and treatment of plastic pollution­induced neurotoxicity.


Asunto(s)
Ferroptosis , Humanos , Microplásticos/metabolismo , Microplásticos/farmacología , Microglía/metabolismo , Sistema de Señalización de MAP Quinasas , Especies Reactivas de Oxígeno/metabolismo , Inflamación/metabolismo , Ferritinas/metabolismo , Ferritinas/farmacología , Oxidorreductasas/metabolismo , Oxidorreductasas/farmacología
3.
Iran J Public Health ; 52(1): 1-9, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36824257

RESUMEN

Background: Cell transplantation is a promising therapeutic strategy for pulmonary fibrosis. In order to clarify the alveolar type II epithelial cell potential utility in the treatment of lung disease, we conducted a meta-analysis, to evaluate alveolar type II epithelial cells in animal models of lung injury and pulmonary fibrosis. Methods: This review followed the recommendations from the PRISMA statements, Comprehensive retrieval method was used to search Embase, PubMed, Cochrane, Chinese Knowledge Infrastructure, VIP and Wanfang databases. A total of 7 studies and 286 model rats were included. Two researchers independently screened the identified studies, based on inclusion and exclusion criteria. All analyses were conducted using Review Manager V.5.3 software. The combined standard mean difference (SMD) and 95% confidence interval (CI) of data from the included studies were calculated using fixed or random-effects models. Results: The analysis of three outcome indexes showed that the heterogeneity of the oxygen saturation group was high (I2=85%), the lung weight group (I2=64%) was close to moderate heterogeneity, and the lung hydroxyproline content group (I2=0) was not heterogeneous. Conclusion: Meta-analysis showed that transplantation of alveolar type II epithelial cells has beneficial effects, and no obvious adverse reactions. Alveolar type II epithelial cell transplantation can significantly reduce the intervention group and lung hydroxyproline content weight, improve the blood oxygen saturation, lung histo-pathology showed significant improvement in pulmonary fibrosis.

4.
Int J Nanomedicine ; 17: 5247-5264, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36388872

RESUMEN

Introduction: Silica nanoparticles (SiNPs) have been widely used in food, cosmetics, medicine and other fields; however, there have been growing concerns regarding their potential adverse effects on health. A large number of studies have confirmed that SiNPs with small particle diameters can pass through the blood brain barrier, causing irreversible damage to the nervous system. This study aims to further explore the molecular mechanism of neurotoxicity of SiNPs and provide a toxicological basis for the medical application of SiNPs. Methods: We conducted an in vitro study using neuroimmune cells (mouse microglial cells, BV2) of the central nervous system to study inflammation and ferroptosis after exposure to SiNPs. We detected cell viability, morphology and ultrastructure, antioxidant function, inflammation, and ferroptosis-related proteins to explore the role of pyroptosis and ferroptosis in the damage of BV2 cells induced by SiNPs. We further explored the relationship between the inflammatory response and ferroptosis induced by SiNPs by silencing the NOD-like receptor thermal protein domain associated protein 3 (NLRP3) gene and inhibiting ferroptosis. Results: The results showed that SiNPs could invade the cytoplasm, change the ultrastructure, activate NLRP3 inflammasomes, release a large number of inflammatory factors, and trigger inflammatory reaction. We also found that SiNPs could disrupt cellular antioxidant function, increase intracellular ferrous ion level and induce ferroptosis. In addition, both inflammation and ferroptosis are alleviated in NLRP3 gene-silenced cells. Conclusion: SiNPs could induce BV2 cytotoxicity through inflammatory response and ferroptosis, which may be mediated by the activation of the NLRP3 inflammasomes.


Asunto(s)
Nanopartículas , Dióxido de Silicio , Animales , Ratones , Dióxido de Silicio/toxicidad , Dióxido de Silicio/química , Inflamasomas/metabolismo , Microglía/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Antioxidantes/metabolismo , Nanopartículas/toxicidad , Nanopartículas/química , Inflamación/inducido químicamente , Inflamación/metabolismo
5.
J Clin Sleep Med ; 18(10): 2397-2404, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-35808945

RESUMEN

STUDY OBJECTIVES: To investigate the prevalence of positional obstructive sleep apnea (P-OSA) and rapid eye movement-related OSA (REM-OSA) in children with OSA and identify related factors. METHODS: This was a cross-sectional study among children aged 2-12 years diagnosed with OSA using overnight polysomnography (PSG) between August 1, 2020, and July 31, 2021. Demographics, anthropometrics, PSG, and OSA-18 questionnaire data were recorded. RESULTS: Data from a total of 474 children were available for analysis. Children had a median age of 4.8 (4.1, 6.4) years, 66.7% were male, and 23.2% were obese. The prevalence of P-OSA was 38.2% and that of REM-OSA was 43.0%. P-OSA was correlated with age and obstructive apnea-hypopnea index (OAHI; odds ratio [OR] = 1.172, 0.947; P = .005, < 0.001, respectively), but not sex, obesity, and adenoid and tonsil size (OR = 1.265, 0.785, 0.826, 0.989; P = .258, 0.327, 0.153, 0.905, respectively). REM-OSA was correlated with age, adenoid size, tonsil size, and OAHI (OR = 0.876, 1.320, 1.387, 1.021; P = .024, 0.040, 0.001, 0.042) but not with sex and obesity (OR = 0.910, 1.281; P = .643, 0.315). CONCLUSIONS: The prevalence of P-OSA was 38.2% and that of REM-OSA was 43.0% in children with OSA. Age was correlated with both the prevalence of P-OSA and REM-OSA, with an increasing and decreasing prevalence as children grew older, respectively. The severity of OSA was significantly associated with the prevalence of both P-OSA and REM-OSA. Adenoid and tonsil size were correlated with the prevalence of REM-OSA but not P-OSA. Obesity and sex were not associated with the prevalence of P-OSA or REM-OSA. CITATION: Wu Y, Zheng L, Cui G, Xu Z, Ni X. Subtypes of obstructive sleep apnea in children and related factors. J Clin Sleep Med. 2022;18(10):2397-2404.


Asunto(s)
Apnea Obstructiva del Sueño , Niño , Estudios Transversales , Femenino , Humanos , Masculino , Obesidad/complicaciones , Polisomnografía , Apnea Obstructiva del Sueño/complicaciones , Sueño REM
6.
Chemosphere ; 291(Pt 2): 132944, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34793849

RESUMEN

Microplastics (MPs) are new environmental pollutants and have received widespread attention in recent years, but the toxicity of the MPs remains to be fully elucidated. To explore the effect of MPs on hepatotoxicity in mice and unravel the mechanism of pyroptosis and ferroptosis in the process of liver injury, we treated mice with 5.0 µm polypropylene microplastics (MPs) at 0.1, 0.5 and 1 mg/mL for 4 weeks. Results revealed that MPs could damage liver structure and function with broken and reduced mitochondrial cristae, as well as increased levels of aspartate minotransferase (AST), alanine aminotransferase (ALT), AST/ALT, alkaline phosphatase (ALP) and lactate dehydrogenase (LDH). Treatment with MPs resulted in pyroptosis as evidenced by increasing expressions of interleukin IL-1ß, IL-18. Additionally, MPs were shown to induce the NOD-like receptor protein 3 (NLRP3) inflammasomes and apoptosis associated speck-like protein (ASC) containing a caspase recruitment domain activation in liver tissue, enabling activation of Caspase-1-dependent signaling pathway induced by inflammatory stimuli resulting from oxidative stress. In addition, the increase of malondialdehyde (MDA) and decrease of glutathione (GSH) and superoxide dismutase (SOD) in the liver indicated that MPs could induce oxidative damage. Moreover, MPs induced lipid peroxidation in the liver of mice could activate the expression of ferroptosis related proteins, including iron metabolism, such as transferrin receptor (TFRC) was active but ferritin heavy chain 1 (FTH1) was inhibited; amino acid metabolism, such as XCT system and glutathione peroxidase 4 (GPX4) were inhibited; lipid metabolism, such as acyl-CoA synthetase long-chain family member 4 (ACSL4) was inhibited. Collectively, these findings evidenced that pyroptosis and ferroptosis occurred in MPs-induced liver injury accompanied by intense oxidative stress and inflammation.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas , Ferroptosis , Animales , Ratones , Microplásticos , Estrés Oxidativo , Plásticos , Poliestirenos , Piroptosis
7.
Environ Toxicol ; 36(7): 1389-1401, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33764603

RESUMEN

Silica nanoparticles (SiNPs) as one of the most productive nano-powder, has been extensively applied in various fields. There has been increasing concern about the adverse effects of SiNPs on the health of ecological organisms and human. The potential cardiovascular toxicity of SiNPs and involved mechanisms remain elusive. Hence, in this study, we investigated the cardiovascular toxicity of SiNPs (60 nm) and explored the underlying mechanisms using H9c2 cardiomyocytes. Results showed that SiNPs induced oxidative stress and activated the Nrf2/HO-1 antioxidant pathway. Autophagy was also activated by SiNPs. Interestingly, N-acetyl-L-cysteine (NAC)attenuated autophagy after inhibiting reactive oxygen species (ROS). Meanwhile, down-regulation of Nrf2 enhanced autophagy. In summary, these data indicated that SiNPs induce autophagy in H9c2 cardiomyocytes through oxidative stress, and the Nrf2/HO-1 pathway has a negative regulatory effect on autophagy. This study provides new evidence for the cardiovascular toxicity of SiNPs and provides a reference for the safe use of nanomaterials in the future.


Asunto(s)
Nanopartículas , Dióxido de Silicio , Autofagia , Humanos , Factor 2 Relacionado con NF-E2/genética , Nanopartículas/toxicidad , Estrés Oxidativo , Especies Reactivas de Oxígeno , Transducción de Señal , Dióxido de Silicio/toxicidad
8.
Toxicol Mech Methods ; 30(9): 646-655, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32746757

RESUMEN

The use of silica nanoparticles (SiNPs) is increasing in popularity; however, the emissions released during manufacturing, use and during the disposal stages potentially harm the environment. SiNPs can enter the body and cause cardiac toxicity indirectly or directly. However, toxicological data on SiNPs in cardiac cells in vitro, and the detailed molecular mechanisms by which damage is caused remain unclear. In the present study, oxidative stress-mediated apoptosis and cytotoxicity induced by SiNPs in H9c2 cells were examined. H9c2 cells were used to explore the mechanisms of toxicity by treating cells with 0, 25, 50, 100, and 200 µg/ml SiNPs, with and without 3 mM of the reactive oxygen species (ROS) scavenger, N-acetyl-l-cysteine (NAC), for 24 h. The results showed that SiNPs decreased cell viability and proliferation by increasing the release of lactate dehydrogenase (LDH) and inducing apoptosis in H9c2 cells. ROS levels were significantly increased in a dose-dependent manner. Additionally, the levels of superoxide dismutase (SOD), glutathione (GSH), and GSH-peroxidase (Px) were significantly decreased following exposure to SiNPs. Treatment with NAC attenuated LDH release; the levels of ROS, SOD, GSH, and GSH-Px production were increased, and SiNPs-induced mitochondrial pathway-dependent apoptosis was reduced. These results demonstrate that apoptosis and cytotoxicity induced by SiNPs in H9c2 cells are a result of ROS-mediated oxidative stress. These data suggest that exposure to SiNPs is a potential risk factor for cardiovascular disease.


Asunto(s)
Apoptosis/efectos de los fármacos , Cardiopatías/inducido químicamente , Mitocondrias Cardíacas/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Nanopartículas/toxicidad , Estrés Oxidativo/efectos de los fármacos , Dióxido de Silicio/toxicidad , Animales , Cardiotoxicidad , Línea Celular , Relación Dosis-Respuesta a Droga , Cardiopatías/metabolismo , Cardiopatías/patología , Mitocondrias Cardíacas/metabolismo , Mitocondrias Cardíacas/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Ratas , Especies Reactivas de Oxígeno/metabolismo , Medición de Riesgo , Transducción de Señal , Factores de Tiempo
9.
Int J Mol Med ; 43(3): 1229-1240, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30628656

RESUMEN

Diseases of the cardiac system caused by silicon dioxide exposure have captured wide public attention. Upon entering the blood circulation, ultrafine particles have the potential to influence cardiomyocytes, leading to myocardial ischemia or even cardiac failure, and the molecular mechanisms remain to be completely elucidated. In this study, the toxicity of ultrafine particles on cardiomyocytes from rats exposed to silica nanoparticles was observed. Rats were randomly divided into a normal saline control group and three exposure groups (2, 5 and 10 mg/kg·body weight) that were intratracheally treated with 60­nm silica nanoparticles. Alterations in body weight, routine blood factors and myocardial enzymes, histopathological and microstructural alterations, apoptosis and the expression of apoptosis­associated proteins were assessed at the end of the exposure period. The silicon levels in the heart and serum, and myocardial enzymes in exposed rats were significantly increased in a dose­dependent manner. In addition, exposure to the silica nanoparticles caused notable histological and ultrastructural alterations in the hearts of these animals. Furthermore, a significant apoptotic effect was observed in the exposure groups. The present data suggest that silica nanoparticles may enter the circulatory system through the lungs, and are distributed to the heart causing cardiovascular injury. Silica nanoparticle­induced apoptosis via the mitochondrial pathway may serve an important role in observed cardiac damage.


Asunto(s)
Apoptosis , Mitocondrias/metabolismo , Miocitos Cardíacos/metabolismo , Nanopartículas , Dióxido de Silicio , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Biomarcadores , Expresión Génica , Inmunohistoquímica , Masculino , Miocitos Cardíacos/ultraestructura , Nanopartículas/química , Nanopartículas/ultraestructura , Ratas , Dióxido de Silicio/química
10.
Stem Cell Res Ther ; 9(1): 110, 2018 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-29673394

RESUMEN

BACKGROUND: Silicosis has been topping the list of high-incidence occupational diseases in developing countries and cannot be completely cured. Recent advances in stem cell research have made possible the treatment of various diseases including lung fibrosis. The application of stem cell therapy in occupational diseases, in particular the use of adipose-derived mesenchymal stem cells (AD-MSCs) in treatment of silicosis, has not yet been reported. The aim of the study is to explore the intervening effect of silica-induced lung fibrosis in rats. METHODS: In this study, we investigated the anti-pulmonary fibrosis effects of the transplantation of AD-MSCs in rats in which lung fibrosis was induced by oral tracheal intubation with silica suspension. Twenty rats were divided into four groups: control group (n = 5), exposure group (n = 5), vehicle group (n = 5) and treatment group (n = 5). AD-MSCs were given to rats after exposure to silica for 24 h. Twenty-eight days after AD-MSC transplantation, we examined the organ coefficient, inflammatory cytokines, apoptosis, pathological and fibrotic changes in lung tissue. RESULTS: Results showed that exposure to silica for 28 days induced an increase of the lung coefficient with significant pulmonary fibrosis. Treatment with AD-MSC transplantation led to a remissive effect on pulmonary fibrosis. We found that after AD-MSC transplantation the inflammatory response decreased and Caspase-3 protein expression significantly decreased with a significant increase of the Bcl-2/Bax ratio. CONCLUSIONS: Anti-inflammatory and anti-apoptosis of AD-MSCs may play important roles in their anti-pulmonary fibrosis effect. Our data suggest that transplantation of AD-MSCs holds promise for potential interference in the formation of silicosis through regulating inflammatory and apoptotic processes.


Asunto(s)
Antiinflamatorios/uso terapéutico , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Madre Mesenquimatosas/metabolismo , Fibrosis Pulmonar/terapia , Silicosis/terapia , Animales , Antiinflamatorios/farmacología , Apoptosis , Modelos Animales de Enfermedad , Masculino , Fibrosis Pulmonar/patología , Ratas , Ratas Sprague-Dawley , Silicosis/complicaciones
11.
Int J Nanomedicine ; 12: 2179-2188, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28356735

RESUMEN

Gap junction intercellular communication (GJIC) between cardiomyocytes is essential for synchronous heart contraction and relies on connexin-containing channels. Connexin 43 (Cx43) is a major component involved in GJIC in heart tissue, and its abnormal expression is closely associated with various cardiac diseases. Silica nanoparticles (SNPs) are known to induce cardiovascular toxicity. However, the mechanisms through which GJIC plays a role in cardiomyocytes apoptosis induced by SNPs remain unknown. The aim of the present study is to determine whether SNPs-decreased GJIC promotes apoptosis in rat cardiomyocytes cell line (H9c2 cells) via the mitochondrial pathway using CCK-8 Kit, scrape-loading dye transfer technique, Annexin V/PI double-staining assays, and Western blot analysis. The results showed that SNPs elicited cytotoxicity in H9c2 cells in a time- and concentration-dependent manner. SNPs also reduced GJIC in H9c2 cells in a concentration-dependent manner through downregulation of Cx43 and upregulation of P-Cx43. Inhibition of gap junctions by gap junction blocker carbenoxolone disodium resulted in decreased survival and increased apoptosis, whereas enhancement of the gap junctions by retinoic acid led to enhanced survival but decreased apoptosis. Furthermore, SNPs-induced apoptosis through the disrupted functional gap junction was correlated with abnormal expressions of the proteins involved in the mitochondrial pathway-related apoptosis such as Bcl-2/Bax, cytochrome C, Caspase-9, and Caspase-3. Taken together, our results provide the first evidence that SNPs-decreased GJIC promotes apoptosis in cardiomyocytes via the mitochondrial pathway. In addition, downregulation of GJIC by SNPs in cardiomyocytes is mediated through downregulation of Cx43 and upregulation of P-Cx43. These results suggest that in rat cardiomyocytes cell line, GJIC plays a protective role in SNPs-induced apoptosis and that GJIC may be one of the targets for SNPs-induced biological effects.


Asunto(s)
Apoptosis/efectos de los fármacos , Comunicación Celular , Uniones Comunicantes/metabolismo , Mitocondrias/metabolismo , Miocitos Cardíacos/citología , Nanopartículas/química , Transducción de Señal/efectos de los fármacos , Dióxido de Silicio/farmacología , Animales , Comunicación Celular/efectos de los fármacos , Línea Celular , Uniones Comunicantes/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Nanopartículas/ultraestructura , Ratas , Factores de Tiempo , Tretinoina/farmacología
12.
Stem Cells Int ; 2015: 516215, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26294918

RESUMEN

The mechanisms behind the repairing effects of the cotransplantation of olfactory ensheathing cells (OECs) with bone marrow mesenchymal stromal cells (BMSCs) have not been fully understood. Therefore, we investigated the effects of the cotransplantation of OECs with BMSCs on antiapoptotic effects in adult rats for which the models of SCI are induced. We examined the changes in body weight, histopathological changes, apoptosis, and the expressions of apoptosis-related proteins after 14 days and 28 days after transplantation. We also assessed animal locomotion using BBB test. We found that treatment with OECs and BMSCs had a remissive effect on behavioral outcome and histopathological changes induced SCI. Furthermore, we observed the significant antiapoptotic effect on cotransplant treated group. In addition, cotransplantation of OECs with BMSCs was found to have more significant repairing effect than that of OECs or BMSCs alone. Furthermore, the recovery of hind limb could be related to antiapoptotic effect of OECs and BMSCs through downregulating the apoptotic pathways. Finally, our data suggested the cotransplantation of OECs with BMSCs holds promise for a potential cure after SCI through the ability to incorporate into the spinal cord.

13.
Cardiovasc Toxicol ; 13(3): 194-207, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23322373

RESUMEN

The purpose of this work was to investigate the cardiovascular toxicity of different sizes and different dosages of silica nanoparticles in Wistar rats. The three silica nanoparticles (30, 60, and 90 nm) and one fine silica particles (600 nm) at three doses of 2, 5, and 10 (mg/Kg bw) were used in the present experiment. After intratracheal instillation for a total of 16 times, concentration of Si in hearts and serum was measured by inductively coupled plasma optical emission spectrometer. The hematology parameters were analyzed by an automated hematology analyzer, and the inflammatory reaction, oxidative stress, endothelial dysfunction, and the myocardial enzymes in serum were measured by kits. Our results showed intratracheal-instilled silica nanoparticles could pass through the alveolar-capillary barrier into systemic circulation. Concentration of Si in the heart and serum depended on the particles size and dosage. The levels of reactive oxygen species (ROS) at 5, 10 mg/Kg bw of the three silica nanoparticles were higher than the fine silica particles. Blood levels of inflammation-related high-sensitivity C-reactive protein and cytokines such as interleukin-1beta (IL-1ß), interleukin-6 (IL-6), and tumor necrosis factor-alpha were increased after exposure to three silica nanoparticles at 10 mg/Kg bw. Moreover, the levels of IL-1ß and IL-6 at 10 mg/Kg bw of silica nanoparticles (30 nm) were higher than the fine silica particles. Significant decrease in superoxide dismutase, glutathione peroxidase and significant increase in malondialdehyde were observed at 10 mg/Kg bw of the three silica nanoparticles. A significant decrease in nitric oxide (NO) production was induced which coincided with the reduction of nitric oxide synthase (NOS) activity and the excessive generation of ROS in rats. The levels of intercellular adhesion molecule-l and vascular cell adhesion molecule-l elevated significantly after exposure to three silica nanoparticles at 10 mg/Kg bw, which are considered as early steps of endothelial dysfunction. We conclude that cardiovascular toxicity of silica nanoparticles could be related to the particles size and dosage. Oxidative stress could be involved in inflammatory reaction and endothelial dysfunction, all of which could aggravate cardiovascular toxicology. In addition, endothelial NO/NOS system disorder caused by nanoparticles could be one of the mechanisms for endothelial dysfunction.


Asunto(s)
Enfermedades Cardiovasculares/inducido químicamente , Nanopartículas/toxicidad , Dióxido de Silicio/toxicidad , Animales , Biomarcadores/sangre , Recuento de Células Sanguíneas , Coagulación Sanguínea/efectos de los fármacos , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/patología , Relación Dosis-Respuesta a Droga , Endotelio Vascular/efectos de los fármacos , Indicadores y Reactivos , Mediadores de Inflamación/metabolismo , Intubación Intratraqueal , Masculino , Microscopía Electrónica de Transmisión , Miocardio/enzimología , Miocardio/metabolismo , Nanopartículas/administración & dosificación , Nanopartículas/metabolismo , Estrés Oxidativo/efectos de los fármacos , Tamaño de la Partícula , Ratas , Ratas Wistar , Dióxido de Silicio/administración & dosificación , Dióxido de Silicio/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...