Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Redox Biol ; 72: 103158, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38631121

RESUMO

Exposure to PM2.5 is correlated with cardiac remodeling, of which cardiac hypertrophy is one of the main clinical manifestations. Ferroptosis plays an important role in cardiac hypertrophy. However, the potential mechanism of PM2.5-induced cardiac hypertrophy through ferroptosis remains unclear. This study aimed to explore the molecular mechanism of cardiac hypertrophy caused by PM2.5 and the intervention role of MitoQ involved in this process. The results showed that PM2.5 could induce cardiac hypertrophy and dysfunction in mice. Meanwhile, the characteristics of ferroptosis were observed, such as iron homeostasis imbalance, lipid peroxidation, mitochondrial damage and abnormal expression of key molecules. MitoQ treatment could effectively mitigate these alternations. After treating human cardiomyocyte AC16 with PM2.5, ferroptosis activator (Erastin) and inhibitor (Fer-1), it was found that PM2.5 could promote ferritinophagy and lead to lipid peroxidation, mitochondrial dysfunction as well as the accumulation of intracellular and mitochondrial labile iron. Subsequently, mitophagy was activated and provided an additional source of labile iron, enhancing the sensitivity of AC16 cells to ferroptosis. Furthermore, Fer-1 alleviated PM2.5-induced cytotoxicity and iron overload in the cytoplasm and mitochondria of AC16 cells. It was worth noting that during the process of PM2.5 caused ferroptosis, abnormal iron metabolism mediated the activation of ferritinophagy and mitophagy in a temporal order. In addition, NCOA4 knockdown reversed the iron homeostasis imbalance and lipid peroxidation caused by PM2.5, thereby alleviating ferroptosis. In summary, our study found that iron homeostasis imbalance-mediated the crosstalk of ferritinophagy and mitophagy played an important role in PM2.5-induced ferroptosis and cardiac hypertrophy.


Assuntos
Autofagia , Cardiomegalia , Ferroptose , Homeostase , Ferro , Miócitos Cardíacos , Material Particulado , Cardiomegalia/metabolismo , Cardiomegalia/etiologia , Cardiomegalia/patologia , Animais , Camundongos , Ferro/metabolismo , Autofagia/efeitos dos fármacos , Humanos , Material Particulado/efeitos adversos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Miócitos Cardíacos/efeitos dos fármacos , Coativadores de Receptor Nuclear/metabolismo , Coativadores de Receptor Nuclear/genética , Peroxidação de Lipídeos/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/efeitos dos fármacos , Linhagem Celular
2.
Sci Total Environ ; 903: 166010, 2023 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-37541522

RESUMO

BACKGROUND: This review aimed to establish a holistic perspective of long-term PM exposure and cardiometabolic diseases, identify long-term PM-related cardiovascular and metabolic risk factors, and provide practical significance to preventative measures. METHOD: A combination of computer and manual retrieval was used to search for keywords in PubMed (2903 records), Embase (2791 records), Web of Science (5488 records) and Cochrane Library (163 records). Finally, a total of 82 articles were considered in this meta-analysis. Stata 13.0 was accustomed to inspecting the studies' heterogeneity and calculating the combined effect value (RR) by selecting the matching models. The subgroup analysis, sensitivity analysis and publication bias tests were also performed. RESULTS: Meta-analysis figured an association between PM and cardiometabolic diseases. PM2.5 (per 10 µg/m3 increase) boosted the risk of hypertension (RR = 1.14, 95 % CI: 1.09-1.19), coronary heart disease (CHD) (RR = 1.21, 95 % CI: 1.08-1.35), diabetes (RR = 1.16, 95 % CI: 1.11-1.21) and stroke (including ischemic stroke and hemorrhagic stroke). PM10 (per 10 µg/m3 increase) elevated the incidence of hypertension (RR = 1.11, 95 % CI: 1.07-1.16) and diabetes (RR = 1.26, 95 % CI: 1.08-1.47). PM1 (per 10 µg/m3 increase) exposure increased the risk of total dyslipidemia, yielding the RR of 1.10 (95 % CI: 1.01-1.18). Furthermore, the elderly, overweight and higher background pollutant level were potentially susceptible to related diseases. CONCLUSION: There was a virtual connection between long-term exposure to PM and cardiometabolic diseases. PM2.5 or PM10 (per 10 µg/m3) increased the risk of hypertension, CHD, diabetes, stroke and dyslipidemia, causing cardiovascular "multimorbidity" in high-risk populations.

3.
Chemosphere ; 330: 138745, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37088202

RESUMO

Pulmonary fibrosis is a severe pulmonary disease, and may related to PM2.5 exposure. Our study aims to explore the pathogenesis of PM2.5-induced pulmonary fibrosis, and MitoQ protective effect in this process. Our results find that inflammatory cells aggregation and pulmonary fibrosis in mice lung after PM2.5 exposure. Moreover, Collagen I/III overproduction, EMT and TGF-ß1/Smad2 pathway activation in mice lung and BEAS-2B after PM2.5 exposure. Fortunately, these changes were partially ameliorated after MitoQ treatment. Meanwhile, severe oxidative stress, mitochondrial homeostasis imbalance, overproduction of 8-oxoG (7,8-dihydro-8-oxoguanine), as well as the inhibition of SIRT3/OGG1 pathway have founded in mice lung or BEAS-2B after PM2.5 exposure, which were alleviated by MitoQ treatment. Collectively, our study found that oxidative stress, especially mitochondrial oxidative stress participates in the PM2.5-induced pulmonary fibrosis, and MitoQ intervention had a protective effect on this progress. Moreover, mitochondrial DNA homeostasis might participate in the pulmonary fibrosis caused by PM2.5 exposure. Our study provides a novel pathogenesis of PM2.5-caused pulmonary fibrosis and a possible targeted therapy for the pulmonary diseases triggered by PM2.5.


Assuntos
Fibrose Pulmonar , Animais , Camundongos , Fibrose Pulmonar/induzido quimicamente , DNA Mitocondrial/metabolismo , Mitocôndrias/metabolismo , Estresse Oxidativo , Material Particulado/efeitos adversos , Homeostase
4.
J Adv Res ; 48: 227-257, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-35998874

RESUMO

BACKGROUND: Nowadays, cigarette smoking remains the leading cause of chronic disease and premature death, especially cardiovascular disease. As an emerging tobacco product, e-cigarettes have been advocated as alternatives to canonical cigarettes, and thus may be an aid to promote smoking cessation. However, recent studies indicated that e-cigarettes should not be completely harmless to the cardiovascular system. AIM OF REVIEW: This review aimed to build up an integral perspective of cigarettes and e-cigarettes-related cardiovascular toxicity. KEY SCIENTIFIC CONCEPTS OF REVIEW: This review adopted the adverse outcome pathway (AOP) framework as a pivotal tool and aimed to elucidate the association between the molecular initiating events (MIEs) induced by cigarette and e-cigarette exposure to the cardiovascular adverse outcome. Since the excessive generation of reactive oxygen species (ROS) has been widely approved to play a critical role in cigarette smoke-related CVD and may also be involved in e-cigarette-induced toxic effects, the ROS overproduction and subsequent oxidative stress are regarded as essential parts of this framework. As far as we know, this should be the first AOP framework focusing on cigarette and e-cigarette-related cardiovascular toxicity, and we hope our work to be a guide in exploring the biomarkers and novel therapies for cardiovascular injury.


Assuntos
Rotas de Resultados Adversos , Doenças Cardiovasculares , Sistema Cardiovascular , Sistemas Eletrônicos de Liberação de Nicotina , Produtos do Tabaco , Espécies Reativas de Oxigênio , Nicotiana
5.
Environ Pollut ; 315: 120437, 2022 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-36272612

RESUMO

Fine particulate matter (PM2.5) exposure has been proved to increase the cardiovascular disease risk. However, there is a lack of comprehensive knowledge on whether a high-fat diet (HFD) affects PM2.5-induced cardiovascular disease. The purpose of this study was to investigate the impairment of lipid metabolism and vascular function by PM2.5 and HFD exposure in ApoE-/- mice. Oil red O staining indicated that co-treatment of PM2.5 and HFD resulted in markedly lipid deposition in the mice aorta. Blood biochemical analysis demonstrated that co-exposure of PM2.5 and HFD could cause dyslipidemia in vivo. Vascular Doppler ultrasound and histopathological analysis found that the functional and structural alterations with fibrosis and calcified remodeling of the vessels were detected after PM2.5 and HFD exposure. For in-depth study, the genome-wide transcriptional analysis performed in macrophages was further revealed that the endoplasmic reticulum stress, immune system process, regulation of cell proliferation etc. were response to PM2.5 exposure; while Lipid and atherosclerosis signaling pathways had a critical role in PM2.5-induced vascular injury. Results from validation experiments manifested that the release of supernatant in PM2.5- or ox-LDL-treated macrophages could decrease the cell viability and increase the lipid ROS in vascular smooth muscle cells (VSMCs). Moreover, the up-regulations of CCL2, IL-6 and IL-1ß in aortic arch of mice were observed after co-exposure with PM2.5 and HFD. Our data hinted that PM2.5 could affect the lipid metabolism reprogramming and induce vascular remodeling, accompanied with synergistic effects of HFD.


Assuntos
Doenças Cardiovasculares , Dieta Hiperlipídica , Camundongos , Animais , Dieta Hiperlipídica/efeitos adversos , Metabolismo dos Lipídeos , Remodelação Vascular , Material Particulado/toxicidade , Camundongos Endogâmicos C57BL
6.
Nanotoxicology ; 16(3): 290-309, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35653618

RESUMO

Epidemiological studies demonstrate that fine particulate matter (PM2.5) promotes the development of atherosclerosis. However, the mechanism insight of PM2.5-induced atherosclerosis is still lacking. The aim of this study was to explore the biological effects of hypoxia-inducible factor 1α (HIF-1α) on PM2.5-triggered atherosclerosis. The vascular stiffness, carotid intima-media thickness (CIMT), lipid and atherosclerotic lesion were increased when von Hippel-Lindau (VHL)-null mice were exposed to PM2.5. Yet, knockout of HIF-1α markedly decreased the PM2.5-triggered atherosclerotic lesion. We firstly performed microarray analysis in PM2.5-treated bone morrow-derived macrophages (BMDMs), which showed that PM2.5 significantly changed the genes expression patterns and affected biological processes such as phagocytosis, apoptotic cell clearance, cellular response to hypoxia, apoptotic process and inflammatory response. Moreover, the data showed knockout of HIF-1α remarkably relieved PM2.5-induced defective efferocytosis. Mechanistically, PM2.5 inhibited the level of genes and proteins of efferocytosis receptor c-Mer tyrosine kinase (MerTK), especially in VHL-null BMDMs. In addition, PM2.5 increased the genes and proteins of a disintegrin and metallopeptidase domain 17 (ADAM17), which caused the MerTK cleavage to form soluble MerTK (sMer) in plasma and cellular supernatant. The sMer was significantly up-regulated in plasma of VHL-null PM2.5-exposed mice. Moreover, PM2.5 could induce defective efferocytosis and activate inflammatory response through MerTK/IFNAR1/STAT1 signaling pathway in macrophages. Our results demonstrate that PM2.5 could induce defective efferocytosis and inflammation by activating HIF-1α in macrophages, ultimately resulting in accelerating atherosclerotic lesion formation and development. Our data suggest HIF-1α in macrophages might be a potential target for PM2.5-related atherosclerosis.


Assuntos
Aterosclerose , Espessura Intima-Media Carotídea , Animais , Aterosclerose/induzido quimicamente , Aterosclerose/metabolismo , Hipóxia/metabolismo , Hipóxia/patologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Macrófagos , Camundongos , Material Particulado/toxicidade , Fagocitose , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/metabolismo , Receptores Proteína Tirosina Quinases/farmacologia , c-Mer Tirosina Quinase/metabolismo
7.
Sci Total Environ ; 839: 156392, 2022 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-35660447

RESUMO

INTRODUCTION: A growing number of epidemiological evidence reveals that electronic cigarettes (E-cigs) were associated with pneumonia, hypertension and atherosclerosis, but the toxicological evaluation and mechanism of E-cigs were largely unknown. OBJECTIVE: Our study was aimed to explore the adverse effects on organs and metabolomics changes in C57BL/6J mice after acute exposure to E-cigs. METHODS AND RESULTS: Hematoxylin and eosin (H&E) staining found pathological changes in tissues after acute exposure to E-cigs, such as inflammatory cell infiltration, nuclear pyknosis, and intercellular interstitial enlargement. E-cigs could increase apoptosis-positive cells in a time-dependent way using Terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL) assay. Oxidative damage indicators of reactive oxygen species (ROS), malondialdehyde (MDA) and 4-hydroxynonena (4-HNE) were also elevated after E-cigs exposure. There was an increasing trend of total glycerol and cholesterol in serum, while the glucose and liver enzymes including alanine aminotransferase (ALT), aspartate transaminase (AST), gamma-glutamyltranspeptidase (γ-GT) had no significant change compared to that of control. Further, Q Exactive high field (HF) mass spectrometer was used to conduct metabolomics, which revealed that differential metabolites including l-carnitine, Capryloyl glycine, etc. Trend analysis showed the type of compounds that change over time. Pathway enrichment analysis indicated that E-cigs affected 24 metabolic pathways, which were mainly regulated amino acid metabolism, further affected the tricarboxylic acid (TCA) cycle. Additionally, metabolites-diseases network analysis found that the type 2 diabetes mellitus, propionic acidemia, defect in long-chain fatty acids transport and lung cancer may be related to E-cigs exposure. CONCLUSIONS: Our findings provided important clues for metabolites biomarkers of E-cigs acute exposure and are beneficial for disease prevention.


Assuntos
Diabetes Mellitus Tipo 2 , Sistemas Eletrônicos de Liberação de Nicotina , Acidemia Propiônica , Animais , Metabolômica , Camundongos , Camundongos Endogâmicos C57BL
8.
Free Radic Biol Med ; 181: 166-179, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35149217

RESUMO

It is reported that oxidative stress homeostasis was involved in PM2.5-induced foam cell formation and progression of atherosclerosis, but the exact molecular mechanism is still unclear. Melatonin is an effective antioxidant that could reverse the cardiopulmonary injury. The main purpose of this study is to investigate the latent mechanism of PM2.5-triggered atherosclerosis development and the protective role of melatonin administration. Vascular Doppler ultrasound showed that PM2.5 exposure reduced aortic elasticity in ApoE-/- mice. Meanwhile, blood biochemical and pathological analysis demonstrated that PM2.5 exposure caused dyslipidemia, elicited oxidative damage of aorta and was accompanied by an increase in atherosclerotic plaque area; while the melatonin administration could effectively alleviate PM2.5-induced macrophage M1 polarization and atherosclerosis in mice. Further investigation verified that NADPH oxidase 2 (NOX2) and mitochondria are two prominent sources of PM2.5-induced ROS production in vascular macrophages. Whereas, the combined use of two ROS-specific inhibitors and adopted with melatonin markedly rescued PM2.5-triggered macrophage M1 polarization and foam cell formation by inhibiting NOX2-mediated crosstalk of Keap1/Nrf2/NF-κB and TLR4/TRAF6/NF-κB signaling pathways. Our results demonstrated that NOX2-mediated oxidative stress homeostasis is critical for PM2.5-induced atherosclerosis and melatonin might be a potential treatment for air pollution-related cardiovascular diseases.


Assuntos
Aterosclerose , Melatonina , Animais , Aterosclerose/metabolismo , Homeostase , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Macrófagos/metabolismo , Melatonina/metabolismo , Melatonina/farmacologia , Camundongos , NADPH Oxidase 2/genética , NADPH Oxidase 2/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo , Material Particulado/metabolismo , Material Particulado/toxicidade
9.
J Hazard Mater ; 430: 128368, 2022 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-35149491

RESUMO

Fine particulate matter (PM2.5) exposure is a major threat to public health, and is listed as one of the leading factors associated with global premature mortality. Among the adverse health effects on multiple organs or tissues, the influence of PM2.5 exposure on cardiovascular system has drawn more and more attention. Although numerous studies have investigated the mechanisms responsible for the cardiovascular toxicity of PM2.5, the various mechanisms have not been integrated due to the variety of the study models, different levels of toxicity assessment endpoints, etc. Adverse Outcome Pathway (AOP) framework is a useful tool to achieve this goal so as to facilitate comprehensive understanding of toxicity assessment of PM2.5 on cardiovascular system. This review aims to illustrate the causal mechanistic relationships of PM2.5-triggered cardiovascular toxicity from different levels (from molecular/cellular/organ to individual/population) by using AOP framework. Based on the AOP Wiki and published literature, we propose an AOP framework focusing on the cardiovascular toxicity induced by PM2.5 exposure. The molecular initiating event (MIE) is identified as reactive oxygen species generation, followed by the key events (KEs) of oxidative damage and mitochondria dysfunction, which induces vascular endothelial dysfunction via vascular endothelial cell autophagy dysfunction, vascular fibrosis via vascular smooth muscle cell activation, cardiac dysregulation via myocardial apoptosis, and cardiac fibrosis via fibroblast proliferation and myofibroblast differentiation, respectively; all of the above cardiovascular injuries ultimately elevate cardiovascular morbidity and mortality in the general population. As far as we know, this is the first work on PM2.5-related cardiovascular AOP construction. In the future, more work needs to be done to explore new markers in the safety assessment of cardiovascular toxicity induced by PM2.5.


Assuntos
Rotas de Resultados Adversos , Poluentes Atmosféricos , Doenças Cardiovasculares , Poluentes Atmosféricos/metabolismo , Poluentes Atmosféricos/toxicidade , Doenças Cardiovasculares/induzido quimicamente , Doenças Cardiovasculares/epidemiologia , Humanos , Miocárdio/metabolismo , Estresse Oxidativo , Material Particulado/metabolismo , Material Particulado/toxicidade
10.
Genes Environ ; 43(1): 47, 2021 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-34654488

RESUMO

Cardiovascular disease (CVD) has become the leading cause of death worldwide, which seriously threatens human life and health. Epidemiological studies have confirmed the occurrence and development of CVD are closely related to air pollution. In particular, fine particulate matter (PM2.5) is recognized as an important environmental factor contributing to increased morbidity, mortality and hospitalization rates among adults and children. However, the underlying mechanism by which PM2.5 promotes CVD development remains unclear. With the development of epigenetics, recent studies have shown that PM2.5 exposure may induce or aggravate CVD through epigenetic changes. In order to better understand the potential mechanisms, this paper reviews the epigenetic changes of CVD caused by PM2.5. We summarized the epigenetic mechanisms of PM2.5 causing cardiovascular pathological damage and functional changes, mainly involving DNA methylation, non-coding RNA, histone modification and chromosome remodeling. It will provide important clues for exploring the biological mechanisms affecting cardiovascular health.

11.
Chemosphere ; 282: 131124, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34374342

RESUMO

Environmental exposure to nanoplastics is inevitable as the application of nanoplastics in our daily life is more and more extensively. So, the adverse effects of nanoplastics on human health are also gaining greater concerns. However, the subsequent toxicological response to nanoplastics, especially on cardiovascular damage was still largely unknown. In this regard, the evaluation of cardiovascular effects of nanoplastics was performed in zebrafish embryos. The results indicated that the no observed adverse effect level (NOAEL) of nanoplastics is 50 µg/mL. The pericardial toxicity and hemodynamic changes were assessed by Albino (melanin allele) mutant zebrafish line. Severe pericardial edema was observed in zebrafish embryos after exposure to nanoplastics. At the concentration higher than NOAEL, nanoplastics significantly decreased the cardiac output (CO) and blood flow velocity. The fluorescence images manifested that the nanoplastics could inhibit the subintestinal angiogenesis of transgenic zebrafish embryos line Tg (fli-1: EGFP), which might disturb the cardiovascular formation and development. The resulting vascular endothelial dysfunction and hypercoagulable state of circulating blood further accelerated thrombosis. Reactive oxidative stress (ROS) and systemic inflammation were also found in Wild AB and Tg (mpo: GFP) zebrafish embryos, respectively. We also found many neutrophils recruiting in the tail vein where the zebrafish embryo thrombosis occurred. Our data suggested that nanoplastics could trigger the cardiovascular toxicity in zebrafish embryos, which could provide an essential clue for the safety assessment of nanoplastics.


Assuntos
Nanopartículas , Peixe-Zebra , Animais , Embrião não Mamífero , Humanos , Microplásticos , Pericárdio , Polietileno
12.
NanoImpact ; 24: 100353, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-35559812

RESUMO

Currently, the potential applications of polydopamine (PDA) nanoparticles in the biomedical field are being extensively studied, such as cell internalization, biocompatible surface modification, biological imaging, nano-drug delivery, cancer diagnosis, and treatment. However, the subsequent toxicological response to PDA nanoparticles, especially on nervous system damage was still largely unknown. In this regard, the evaluation of the neurotoxicity of PDA nanoparticles was performed in the developing zebrafish larvae. Results of the transmission electron microscope (TEM), diameter analysis, 1H NMR, and thermogravimetric analysis (TGA) indicated that PDA nanoparticles had high stability without any depolymerization; the maximum non-lethal dose (MNLD) and LD10 of PDA nanoparticles for zebrafish were determined to be 0.5 mg/mL and 4 mg/mL. Pericardial edema and uninflated swim bladders were observed in zebrafish larvae after exposure to PDA nanoparticles. At a concentration higher than MNLD, the fluorescence images manifested that the PDA nanoparticles could inhibit the axonal growth of peripheral motor neurons in zebrafish, which might affect the movement distances and speed, disturb the movement trace, finally resulting in impaired motor function. However, in further investigating the mechanism of PDA nanoparticles-induced neurotoxicity in zebrafish larvae, we did not find apoptosis of central neurocytes. Our data suggested that PDA nanoparticles might trigger neurotoxicity in zebrafish, which could provide an essential clue for the safety assessment of PDA nanoparticles.


Assuntos
Nanopartículas , Peixe-Zebra , Animais , Indóis/toxicidade , Larva , Nanopartículas/toxicidade , Polímeros/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA