Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 137
Filter
1.
Anal Chem ; 96(22): 9270-9277, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38770656

ABSTRACT

Developing a specific, sensitive, rapid, and on-site method for detecting pathogenic bacteria in food samples is critical to ensuring public safety. This article demonstrates a CRISPR/Cas13a system and a chemiluminescence resonance energy transfer (CRET) (CRISPR/Cas 13a-assisted CRET)-based strategy for sensitive and on-site detection of pathogenic bacteria in real samples. Once the hybrid double strand of aptamerS. aureus-cRNA recognizes the target model bacteria of Staphylococcus aureus (S. aureus), the released cRNA would bind with CRISPR/Cas 13a to form a complex of cRNA-CRISPR/Cas 13a, which could cleave the RNA molecule in the detecting probe of horseradish peroxidase (HRP) modified-gold nanoparticles (AuNPs) linked by RNA (AuNPs-RNA-HRP), resulting in an enhanced chemiluminescence signal due to the CRET "OFF" phenomenon after introducing the chemiluminescence substrate of luminol. The CRISPR/Cas 13a-assisted CRET strategy successfully detected S. aureus in drinking water and milk with detection limits of 20 and 30 cfu/mL, respectively, within the recovery of 90.07-105.50%. Furthermore, after integrating with an immunochromatographic test strip (ICTS), the CRISPR/Cas 13a-assisted CRET strategy achieved the on-site detection of as low as 102 cfu/mL of S. aureus in drinking water and milk via a smartphone, which is about 10 times lower than that in the previously reported AuNPs-based colorimetric ICTS, demonstrating a convenient and sensitive detection method for S. aureus in real samples.


Subject(s)
CRISPR-Cas Systems , Gold , Milk , Staphylococcus aureus , Staphylococcus aureus/isolation & purification , Staphylococcus aureus/genetics , CRISPR-Cas Systems/genetics , Gold/chemistry , Milk/microbiology , Animals , Luminescent Measurements , Drinking Water/microbiology , Metal Nanoparticles/chemistry , Biosensing Techniques/methods , Limit of Detection , Luminescence
2.
Chem Res Toxicol ; 37(2): 429-438, 2024 02 19.
Article in English | MEDLINE | ID: mdl-38193392

ABSTRACT

The increasing nanoparticle (NP) applications in the biomedical field have become an emerging concern regarding human health. NP exposure may play a role in the accelerating Alzheimer's disease (AD) progression; however, the etiology of this disorder is complex and remains largely unclear. Here, we identified that intravenous injection of silica NPs (SiNPs) caused the blood-brain barrier breakdown via downregulating tight junction-related gene expressions. Meanwhile, SiNPs upregulate the transport receptor for advanced glycation end products (RAGE) that govern the ß-amyloid (Aß) influx to the brain; however, low-density lipoprotein receptor-related protein 1 (LRP1) that controls the efflux of Aß from the brain was not affected. Consequently, an increase in Aß burden in the brain of SiNP-challenged APP/PS1 mice was found. Intriguingly, plasma apolipoprotein E (ApoE) adsorbed on the surface of SiNPs partially relieves this effect. Using ApoE knockout (ApoE-/-) mice, we confirmed that SiNPs covered with serum without ApoE showed further elevated AD symptoms. Together, this study offered a compilation of data to support the potential risk factors of NP exposure and AD pathology.


Subject(s)
Alzheimer Disease , Nanoparticles , Peptide Fragments , Humans , Mice , Animals , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Presenilin-1/genetics , Amyloid beta-Peptides/metabolism , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Silicon Dioxide , Mice, Transgenic , Disease Models, Animal
3.
Environ Sci Technol ; 58(4): 1832-1841, 2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38230996

ABSTRACT

The massive production of plastics causes the ubiquitous existence of microplastics (MPs) in the biota, therefore, posing exposure risks and potential health concerns to human beings. However, the exact mechanisms of MPs-induced toxicities and abnormalities are largely unknown. In this study, we developed a mouse model of gavage polystyrene microplastics (PS MPs) for 30 days. We found that PS MPs can damage the intestinal barrier, accumulate in the liver tissue, and cause injury. The liver and intestine are both highly associated with bile acid (BA) metabolism. Indeed, we found that PS MPs dysregulate BA synthesis and efflux-related gene expression in the liver, causing cholestasis. Tandemly, PS MPs alter the ratio of primary to secondary BA in the feces by affecting the composition of the intestinal flora. At last, PS MPs alter mice's fecal BA profile, which affects normal BA metabolism. Taken together, the present study provides robust data on the mechanism of toxicity of MPs causing the disturbance of BA metabolism via a 4-step gut-liver loop.


Subject(s)
Cholestasis , Plastics , Humans , Animals , Mice , Microplastics , Liver , Polystyrenes , Bile Acids and Salts
4.
Proc Natl Acad Sci U S A ; 118(44)2021 11 02.
Article in English | MEDLINE | ID: mdl-34716267

ABSTRACT

The rapid development of nanotechnology has greatly benefited modern science and engineering and also led to an increased environmental exposure to nanoparticles (NPs). While recent research has established a correlation between the exposure of NPs and cardiovascular diseases, the intrinsic mechanisms of such a connection remain unclear. Inhaled NPs can penetrate the air-blood barrier from the lung to systemic circulation, thereby intruding the cardiovascular system and generating cardiotoxic effects. In this study, on-site cardiovascular damage was observed in mice upon respiratory exposure of silica nanoparticles (SiNPs), and the corresponding mechanism was investigated by focusing on the interaction of SiNPs and their encountered biomacromolecules en route. SiNPs were found to collect a significant amount of apolipoprotein A-I (Apo A-I) from the blood, in particular when the SiNPs were preadsorbed with pulmonary surfactants. While the adsorbed Apo A-I ameliorated the cytotoxic and proinflammatory effects of SiNPs, the protein was eliminated from the blood upon clearance of the NPs. However, supplementation of Apo A-I mimic peptide mitigated the atherosclerotic lesion induced by SiNPs. In addition, we found a further declined plasma Apo A-I level in clinical silicosis patients than coronary heart disease patients, suggesting clearance of SiNPs sequestered Apo A-I to compromise the coronal protein's regular biological functions. Together, this study has provided evidence that the protein corona of SiNPs acquired in the blood depletes Apo A-I, a biomarker for prediction of cardiovascular diseases, which gives rise to unexpected toxic effects of the nanoparticles.


Subject(s)
Apolipoprotein A-I/deficiency , Cardiovascular Diseases/etiology , Nanoparticles/adverse effects , Adsorption/drug effects , Animals , Apolipoprotein A-I/blood , Cardiovascular System , Lung , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nanoparticles/chemistry , Nanotechnology , Oxidative Stress/drug effects , Particle Size , Signal Transduction/drug effects , Silicon Dioxide/adverse effects , Silicon Dioxide/chemistry
5.
Small ; 19(25): e2208249, 2023 06.
Article in English | MEDLINE | ID: mdl-36929641

ABSTRACT

Confirming bacterial infection at an early stage and distinguishing between sterile inflammation and bacterial infection is still highly needed for efficient treatment. Here, in situ highly sensitive magnetic resonance imaging (MRI) bacterial infection in vivo based on a peptide-modified magnetic resonance tuning (MRET) probe (MPD-1) that responds to matrix metallopeptidase 2 (MMP-2) highly expressed in bacteria-infected microenvironments is achieved. MPD-1 is an assembly of magnetic nanoparticle (MNP) bearing with gadolinium ion (Gd3+ ) modified MMP-2-cleavable self-assembled peptide (P1 ) and bacteria-targeting peptide (P), and it shows T2 -weighted signal due to the assemble of MNP and MRET ON phenomenon between MNP assembly and Gd3+ . Once MPD-1 accumulates at the bacterially infected site, P1 included in MPD-1 is cleaved explicitly by MMP-2, which triggers the T2 contrast agent of MPD-1 to disassemble into the monomer of MNP, leading the recovery of T1 -weighted signal. Simultaneously, Gd3+ detaches from MNP, further enhancing the T1 -weighted signal due to MRET OFF. The sensitive MRI of Staphylococcus aureus (low to 104 CFU) at the myositis site and accurate differentiation between sterile inflammation and bacterial infection based on the proposed MPD-1 probe suggests that this novel probe would be a promising candidate for efficiently detecting bacterial infection in vivo.


Subject(s)
Bacterial Infections , Infectious Disease Medicine , Magnetic Resonance Imaging , Bacterial Infections/diagnosis , Magnetic Resonance Imaging/instrumentation , Infectious Disease Medicine/instrumentation , Infectious Disease Medicine/methods , Matrix Metalloproteinase 2/analysis , Matrix Metalloproteinase 2/metabolism , Metal Nanoparticles/chemistry , Gadolinium/chemistry , Peptides/chemistry , Molecular Probes/chemistry , Molecular Probes/metabolism , Molecular Probes/standards , Animals , Mice , RAW 264.7 Cells , Staphylococcus aureus/isolation & purification , Sensitivity and Specificity , Staphylococcal Infections/diagnosis
6.
Langmuir ; 39(5): 1997-2007, 2023 02 07.
Article in English | MEDLINE | ID: mdl-36706054

ABSTRACT

Humans may intendedly or unintendedly be exposed to nanomaterials through food, water, and air. Upon exposure, nanomaterials can pierce the bloodstream and translocate to secondary organs, including the brain, which warrants increased concern for the potential health impacts of nanomaterials. Due to their large surface area and interaction energy, nanomaterials can adsorb surrounding proteins. The misfolding and self-aggregation of amyloid-ß (Aß) have been considered significant factors in the pathogenesis of Alzheimer's disease. We thus hypothesize that brain-targeted nanomaterials may modulate Aß aggregation and cause related neurotoxicity. Here, we showed that TiO2 nanoparticles (NPs) and their aminated analogue (TiO2-NH2 NPs) adsorb the Aß42 peptide and accelerate its early oligomerization. Molecular dynamics simulation indicated that the adsorption onto TiO2 NPs and TiO2-NH2 NPs surfaces can stabilize the ß-sheet-rich conformations formed by the Aß42 peptide. The binding sites between TiO2-NH2 NPs and the Aß42 oligomer surface were mainly concentrated in the hydrophobic core region, and the ß-sheet conformation spontaneously formed by Aß42 oligomers can be better stabilized through a hydrogen bond, electrostatic attraction, and hydrophobic interaction. This study will further help in the understanding of nanomaterial-related neurotoxicities and the regulation of their applications.


Subject(s)
Alzheimer Disease , Nanoparticles , Humans , Peptide Fragments/chemistry , Amyloid beta-Peptides/chemistry , Molecular Dynamics Simulation
7.
Chem Res Toxicol ; 35(3): 378-382, 2022 03 21.
Article in English | MEDLINE | ID: mdl-35060696

ABSTRACT

With growing applications of plastic products, there is a growing threat to human health. Therefore, it is important to understand their toxicological behaviors. The release of neutrophil extracellular traps (NETs) was clarified as a new immune defense mechanism against intruders. Here, we discovered that polystyrene nanoplastics (PS NPs) induce NET formation, with involvement of reactive oxygen species, peptidyl arginine deiminase 4 (PAD4), and neutrophil elastase. Moreover, overexpression of PAD4 induced by PS NPs further mediates histone citrullination and chromatin depolymerization. These results provide important information and promising strategies for safety and immunotoxicity assessment of NPs.


Subject(s)
Extracellular Traps , Animals , Mice , Microplastics , Neutrophils , Polystyrenes , Protein-Arginine Deiminase Type 4
8.
Chem Res Toxicol ; 35(4): 597-605, 2022 04 18.
Article in English | MEDLINE | ID: mdl-35168318

ABSTRACT

Polychlorinated biphenyls (PCBs) are a group of persistent organic environmental pollutants with various toxic effects. Our previous research found that a highly reactive quinone metabolite of PCBs, namely, PCB29-pQ, causes excessive reactive oxygen species (ROS) production and different toxic actions. Neutrophil extracellular traps (NETs), the product of NETosis, are one of the newly discovered programmed cell deaths. Recent studies have suggested the association of NET formation with excess ROS. The objective of this study was to investigate the influence of PCB29-pQ exposure on NETs and its possible molecular mechanisms. Using scanning electron microscopy, immunofluorescence microscopy, and the quantitative analysis of extracellular DNA, we found that PCB29-pQ exposure induces the formation of NETs in mouse bone marrow. Mechanistically, our results suggested that PCB29-pQ induces histone citrullination and chromatin decondensation, which are necessary processes for NET formation. Moreover, PCB29-pQ exposure increases ROS and autophagy levels, while ROS and autophagy inhibitors significantly reverse NET formation. These results indicated that PCB29-pQ-induced NET formation was mediated by the intracellular ROS level and autophagy signaling. In general, our research uncovered a toxicity mechanism of PCB29-pQ, which suggested the necessity of evaluating its immunotoxicity during the risk assessment of PCB exposure.


Subject(s)
Extracellular Traps , Polychlorinated Biphenyls , Animals , Benzoquinones , Bone Marrow/metabolism , Extracellular Traps/metabolism , Mice , Neutrophils/metabolism , Polychlorinated Biphenyls/toxicity , Quinones , Reactive Oxygen Species/metabolism
9.
Environ Sci Technol ; 56(12): 8319-8325, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35576522

ABSTRACT

Nanoparticles (NPs) can make their way to the brain and cause in situ damage, which is a concern for nanomaterial application and airborne particulate matter exposure. Our recent study indicated that respiratory exposure to silica nanoparticles (SiO2 NPs) caused unexpected cardiovascular toxic effects. However, the toxicities of SiO2 NPs in other organs have warranted further investigation. To confirm the accumulation of SiO2 NPs in the brain, we introduced SiO2 NPs with different diameters into mice via intranasal instillation (INI) and intravenous injection (IVI) in parallel. We found that SiO2 NPs may target the brain through both olfactory and systemic routes, but the size of SiO2 NPs and delivery routes both significantly affected their brain accumulation. Surprisingly, while equivalent SiO2 NPs were found in the brain regions, brain lesions were distinctly much higher in INI than in the IVI group. Mechanistically, we showed that SiO2 NPs introduced via INI induced brain apoptosis and autophagy, while the SiO2 NPs introduced via IVI only induced autophagy in the brain.


Subject(s)
Nanoparticles , Silicon Dioxide , Animals , Apoptosis , Brain , Mice , Nanoparticles/toxicity , Particulate Matter , Silicon Dioxide/toxicity
10.
Small ; 17(44): e2103627, 2021 11.
Article in English | MEDLINE | ID: mdl-34554653

ABSTRACT

Despite the significant advances of imaging techniques nowadays, accurate diagnosis of bacterial infections and real-time monitoring the efficacy of antibiotic therapy in vivo still remain huge challenges. Herein, a self-assembling peptide (FFYEGK) and vancomycin (Van) antibiotic molecule co-modified gadolinium (Gd) MRI nanoaggregate probe (GFV) for detecting Staphylococcus aureus (S. aureus) infection in vivo and monitoring the treatment of S. aureus-infected myositis by using daptomycin (Dap) antibiotic as model are designed and fabricated. The as-prepared GFV probe bears Van molecules, making itself good bacteria-specific targeting, and the peptide in the probe can enhance the longitudinal relaxivity rate (r1 ) after self-assembly due to the π-π stacking. The study showed that, based on the GFV probe, bacterial infections and sterile inflammation can be discriminated, and as few as 105 cfu S. aureus can be detected in vivo with high specificity and accurately. Moreover, the T1 signal of GFV probe at the S. aureus-infected site in mice correlates with the increasing time of Dap treating, indicating the possibility of monitoring the efficacy of antibacterial agents for infected mice based on the as proposed GFV probe. This study shows the potential of GFV probe for diagnosis, evaluation, and prognosis of infectious diseases in clinics.


Subject(s)
Anti-Infective Agents , Staphylococcal Infections , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Bacteria , Magnetic Resonance Imaging , Mice , Microbial Sensitivity Tests , Staphylococcal Infections/drug therapy , Staphylococcus aureus
11.
Chem Res Toxicol ; 34(9): 2125-2134, 2021 09 20.
Article in English | MEDLINE | ID: mdl-34428026

ABSTRACT

Polybrominated diphenyl ethers (PBDEs) are used worldwide in brominated flame retardants. Although due to the forbiddance of their application, PBDEs continuously exist in the environment due to their persistence. Therefore, it is important to expand the understanding of their potential toxicities and human risks. The underlying cardiovascular toxicological mechanisms of PBDEs are still largely unknown. Our previous studies indicated that PBDE quinone-type metabolite (PBDEQ) exposure causes reactive oxygen species (ROS)-driven cytotoxicity and various types of programmed cell death. Here, we first reported PBDEQ exposure induces atherosclerosis progression in bone marrow-derived macrophages (BMDMs) isolated from wild-type C57BL/6 or CD36-/- mice and J774A.1 macrophage models. First, we found that PBDEQ exposure induced lipid accumulation in oxidized low-density lipid (Ox-LDL)-treated J774A.1 macrophages. Consistently, in J774A.1 macrophages, PBDEQ exposure resulted in NLR family pyrin domain containing 3 (NLRP3) inflammasome activation and pyroptosis. CD36, a scavenger receptor responsible for the mediation of Ox-LDL uptake, was upregulated upon PBDEQ treatment. On the contrary, genetic knockout of CD36 or CD36 silencing by small interfering RNA efficiently attenuates PBDEQ-promoted lipid accumulation in BMDMs and J774A.1 macrophages. These findings highlight the effect of CD36 on the cardiovascular toxicity of PBDEs, which provides a better understanding of the pro-atherosclerosis effect of PBDEs.


Subject(s)
Atherosclerosis/etiology , Benzoquinones/toxicity , Halogenated Diphenyl Ethers/toxicity , Inflammasomes/drug effects , Lipid Metabolism/drug effects , Pyroptosis/drug effects , Animals , CD36 Antigens/metabolism , Cell Line , Foam Cells/drug effects , Male , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
12.
Anal Chem ; 92(16): 11462-11468, 2020 08 18.
Article in English | MEDLINE | ID: mdl-32693581

ABSTRACT

The intracellular invasion and survival of a pathogen like Staphylococcus aureus (S. aureus) within host cells enable them to resist antibiotic treatment and colonize long-term in the host, which leads to a series of clinical issues. Rapid and specific detection of intracellular bacteria is important in diagnosis of infection and guiding antibiotic administration. Herein, this work reports a simple one-step fluorescence resonance energy transfer (FRET) platform-based strategy to achieve specific and rapid detection of S. aureus in specimens of phagocytic cells. The aptamer modified quantum dots (Aptamer-QDs) and antibiotic molecule of Teicoplanin functionalized-gold nanoparticles (Teico-AuNPs) dual-recognition units to S. aureus are employed as energy donor and acceptor, respectively. Based on the "off" to "on" signal readout mode, when in the presence of target S. aureus, the donor and acceptor are close to each other and bring high FRET efficiency, which is suitable for analysis of intracellular S. aureus. After it was incubated with the sample for 2 h, the as-prepared FRET sensor showed selectivity to the target S. aureus, and the changed fluorescence signal shows an obvious variation with increasing concentration of S. aureus in pure buffer. When the FRET strategy was further applied to assay intracellular S. aureus, there was an obvious fluorescence signal change obtained both by spectrum analysis and visual fluorescence microscope observation when the average number of S. aureus in one host cell (NS. aureus/cell) was as low as 1, which can be attributed to the high fluorescence quenching efficiency of about 41.3%. It could be envisioned that this FRET nanoprobe with high fluorescence quenching efficiency may provide a simple approach for the facile, selective, and rapid diagnosis of an intracellular bacterial infection.


Subject(s)
Aptamers, Nucleotide/chemistry , Bacterial Load/methods , Fluorescence Resonance Energy Transfer/methods , Fluorescent Dyes/chemistry , Quantum Dots/chemistry , Staphylococcus aureus/isolation & purification , Animals , Gold/chemistry , Limit of Detection , Macrophages/microbiology , Metal Nanoparticles/chemistry , Mice , RAW 264.7 Cells , Staphylococcus aureus/chemistry , Teicoplanin/chemistry
13.
Anal Chem ; 92(22): 14990-14998, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33104346

ABSTRACT

Gold nanoparticle (AuNP)-based sensors have been extensively applied for sensing or imaging. It is known that a protein shell named protein corona (PC) formed around the nanomaterials could not only block the desired function of nanomaterials but also affect their behavior, which is a hot and important issue needing consideration. Therefore, we hypothesize that the formation of PC around AuNPs could inevitably affect the AuNP-based target assay. In this work, the effects of PC on the detection results in sensors based on AuNPs were studied. Three types of noncovalent molecule-AuNP sensors including AuNP-dichlorofluorescein, AuNP-aptamer, and AuNP-antibody-DNA were constructed, and several typical proteins (bovine serum albumin, fibrinogen, hemoglobin, and ß-lactoglobulin), milk, and fetal bovine serum were selected as models for the formation of PCs. This study shows that the PC could cause the loss of detection signals (up to 80%) and result in positive deviation of the measuring value compared with the true value. Moreover, the loss of detection signals could also increase the limits of detection (almost 10 times), decreasing the sensitivity of the three types of sensors, as proposed in this work compared to that without PC. Moreover, the polyethylene glycol backfilling strategy could not resolve the negative effects of PC on noncovalent molecule-AuNP sensors. The impacts of PC on detection results from noncovalent molecule-AuNP sensors would cause misdiagnosis or wasted production, which needs careful reconsideration of the AuNP-based detection in application fields like clinic diagnosis, food safety control, and so forth.


Subject(s)
Biosensing Techniques/methods , Gold/chemistry , Metal Nanoparticles/chemistry , Protein Corona/chemistry , Animals , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/metabolism , DNA/chemistry , Polyethylene Glycols , Protein Corona/metabolism
14.
Toxicol Appl Pharmacol ; 407: 115241, 2020 11 15.
Article in English | MEDLINE | ID: mdl-32937103

ABSTRACT

Neurological diseases were often characterized by progressive neuronal death, and emerging evidences suggested that ferroptosis may be an active driver of multiple neurodegenerative diseases. However, the mechanisms underlying ferroptosis in neuron cells are unclear. Here, we demonstrated that ferroptotic stimuli caused injury in neuron-like PC12 cells by modulating the expression of proteins involved in iron metabolism and lipid peroxidation at multiple levels, such as altering iron import/export, activating ferritinophagy, and decreasing glutathione (GSH) level. Nuclear factor erythroid 2-related factor 2 (Nrf2) regulates multiple genes involved in ferroptosis, however, its exact role remain elusive. Our mechanistic inquiry revealed that Nrf2 expression enhanced iron storage capacity by increasing ferritin heavy chain 1 (FTH1) expression in PC12 cells. Moreover, Nrf2 alleviated the decrease in GSH level by promoting the expression of genes related to GSH synthesis, including solute carrier family 7 member 11 (SLC7A11) and cysteine ligase (GCL). The contribution of Nrf2 on ferroptosis resistance was further verified by increasing cell tolerance to oxidative stress. Furthermore, Nfe2l2 (Nrf2) knockdown sensitized cells to ferroptotic cell death. Taken together, our findings suggested that iron accumulation caused by altering iron metabolism and the decrease of GSH content are key factors in determining ferroptosis in PC12 cells, and Nrf2 inhibits ferroptosis by combating iron-induced oxidative stress. Our present study provided new clues for the intervention and prevention against ferroptosis-associated neurological diseases.


Subject(s)
Ferroptosis/drug effects , Glutathione/biosynthesis , Iron Overload/drug therapy , NF-E2-Related Factor 2/biosynthesis , Neurons/drug effects , Amino Acid Transport Systems, Acidic/biosynthesis , Amino Acid Transport Systems, Acidic/genetics , Animals , Gene Knockdown Techniques , Glutathione/deficiency , Iron/metabolism , Lipid Peroxidation/drug effects , NF-E2-Related Factor 2/drug effects , PC12 Cells , RNA, Small Interfering , Rats
15.
Chem Res Toxicol ; 33(8): 2022-2042, 2020 08 17.
Article in English | MEDLINE | ID: mdl-32677429

ABSTRACT

Polychlorinated biphenyls (PCBs) are notorious persistent organic pollutants that were banned in the last century. However, PCBs still remain ubiquitous in the ecosystem due to their persistence and bioaccumulative potency against environmental and biological degradation. Albeit there is no longer the permission of commercial production of PCBs, they were continuously released into global biota via illegal disposal of e-waste or as byproducts of industrial supplies. The role of oxidative stress is often implicated in PCBs' toxicology. PCBs, especially coplanar ones, have a high affinity toward aryl hydrocarbon receptors and inducing CYP1A1, which is considered as a source of oxidative stress. Although commercial PCBs and coplanar individual PCBs, for example, PCB 77 and 126, induced oxidative stresses have been extensively investigated, PCB metabolite-induced oxidative stress has received less attention. PCBs can undergo phase I metabolism which metabolizes the parent PCBs into hydroquinone/semiquinone/quinone metabolites as a futile redox cycle, producing downstream reactive oxygen species (ROS) as byproducts. PCBs can also undergo phase II metabolism yielding methylsulfonyl metabolites that deplete glutathione and such. PCB metabolites induce oxidative stress generally via direct production of ROS or indirect scavenge antioxidant and inhibit antioxidant enzymes, disturbing cellular redox balance. This review aims to provide a critical summary of PCBs metabolism, PCBs parents, and daughter metabolite-induced oxidative stress. We especially focus on the connection between parent PCBs and downstream metabolites, to encourage research associated with PCB metabolite-induced oxidative stress.


Subject(s)
Polychlorinated Biphenyls/metabolism , Oxidative Stress/drug effects , Polychlorinated Biphenyls/pharmacology
16.
Chem Res Toxicol ; 33(6): 1497-1507, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32434321

ABSTRACT

Polychlorinated biphenyls (PCBs) are persistent organic environmental pollutants. According to previous epidemiological reports, PCBs exposure is highly related to atherosclerosis. However, studies of PCBs metabolites and atherosclerosis and corresponding mechanism studies are scarce. In this study, we evaluated the effect of 2,3,5-trichloro-6-phenyl-[1,4]-benzoquinone (PCB29-pQ), a presumptive PCB metabolite, on atherosclerosis. Aortic plaques were increased in PCB29-pQ-treated ApoE-/- mice [intraperitoneally (i.p.) injection of 5 mg/kg body weight of PCB29-pQ once a week for 12 continuous weeks, high-fat feeding]. We observed lipids accumulation and the release of interleukin-1 beta (IL-1ß), tumor necrosis factor alpha (TNF-α), and interleukin-6 (IL-6) in ApoE-/- mice. In addition, we found that PCB29-pQ promoted the levels of total cholesterol, free cholesterol, triglyceride, and cholesteryl ester. Mechanism investigation indicated that PCB29-pQ induces the activation of three branches of endoplasmic reticulum (ER) stress response, that is, phosphorylated protein kinase R-like ER kinase (p-PERK), eukaryotic translation initiation factor 2α (eIF2α) and transcription factor 6 (ATF6), which is responsible for downstream necrosis. More importantly, we found the silence of CD36 is able to reverse PCB29-pQ-induced adverse effects completely. Overall, PCB29-pQ exposure resulted in lipid accumulation, ER stress response, apoptosis, and pro-inflammatory cytokines release via CD36, ultimately leading to atherosclerosis.


Subject(s)
Atherosclerosis/chemically induced , Benzoquinones/toxicity , CD36 Antigens/metabolism , Endoplasmic Reticulum Stress/drug effects , Environmental Pollutants/toxicity , Lipid Metabolism/drug effects , Polychlorinated Biphenyls/toxicity , Animals , Apoptosis/drug effects , Atherosclerosis/metabolism , CD36 Antigens/genetics , Cholesterol/metabolism , Cytokines/metabolism , Diet, High-Fat , Male , Mice , Mice, Knockout, ApoE , Necrosis/chemically induced , RAW 264.7 Cells
17.
Environ Sci Technol ; 54(18): 11485-11493, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32786567

ABSTRACT

The adverse biological and ecological consequences of plastic debris have become a serious problem worldwide. Evidences have uncovered the accumulation of nanoplastics (NPs) in organisms. In a complex biological environment, proteins are prone to adsorbed onto the NPs' surface and form a protein corona layer, which mediates the interaction of NPs with cells. Here, we discovered the interaction of polystyrene (PS) NPs with protein fetal bovine serum (FBS) and altered cytotoxic effects. Mechanistically, prefabricated FBS protein corona mediated the relief of autophagic flux blockage, autophagosomes accumulation, and lysosomal damage in RAW264.7 cells caused by PS NPs. Using an individual fluorescent protein bovine serum albumin (BSA) as a corona surrogate, we demonstrated that coronal BSA remains, at least partially, on the surface of PS NPs during the initial stage of internalization and protects cell membrane from PS NPs-induced damage. However, along with the degradation of corona in lysosomes, reappearance of cytotoxicity was observed. Herein, we provided a proof of principle of the manipulation of corona on NPs' toxicity and we expect the result will promote the further safety assessment of NPs.


Subject(s)
Nanoparticles , Protein Corona , Animals , Cattle , Lysosomes , Microplastics , Polystyrenes
18.
Part Fibre Toxicol ; 17(1): 46, 2020 09 18.
Article in English | MEDLINE | ID: mdl-32948194

ABSTRACT

BACKGROUND: With the development of zinc oxide nanoparticles (ZnO NPs) in the field of nanotechnology, their toxicological effects are attracting increasing attention, and the mechanisms for ZnO NPs neurotoxicity remain obscure. In an attempt to address concerns regarding neurotoxicity of ZnO NPs, we explored the relationship between free zinc ions, reactive oxygen species (ROS) and neurotoxic mechanisms in ZnO NPs-exposed PC12 cells. RESULT: This study demonstrated the requirement of free zinc ions shed by ZnO NPs to over generation of intracellular ROS. Next, we identified autophagic cell death was the major mode of cell death induced by ZnO NPs, and autophagosome accumulation resulted from not only induction of autophagy, but also blockade of autophagy flux. We concluded that autophagic cell death, resulting from zinc ions-ROS-c-Jun N-terminal kinase (JNK)-autophagy positive feedback loop and blockade of autophagosomal-lysosomal fusion, played a major role in the neurotoxicity of ZnO NPs. CONCLUSION: Our study contributes to a better understanding of the neurotoxicity of ZnO NPs and might be useful for designing and developing new biosafety nanoparticles in the future.


Subject(s)
Metal Nanoparticles/toxicity , Zinc Oxide/toxicity , Autophagic Cell Death , Autophagosomes , Autophagy , Cell Death , Nanoparticles , Reactive Oxygen Species , Toxicity Tests
19.
Chem Res Toxicol ; 32(12): 2422-2432, 2019 12 16.
Article in English | MEDLINE | ID: mdl-31680514

ABSTRACT

Polychlorinated biphenyls (PCBs) are organic environmental pollutants that are accused of various toxic effects. PCB exposure is widely believed to be associated with atherosclerosis, but the underlying mechanisms are unclear. Although PCBs are easily metabolized, there is rarely information on the effects of their metabolites on atherosclerosis. Currently, we evaluate the effect of 2,3,5-trichloro-6-phenyl-[1,4]-benzoquinone (PCB29-pQ) on the critical phase of atherosclerosis development, that is, the formation of macrophage-derived foam cells. We exposed Ox-LDL-induced RAW264.7 cells to 2.5 µM and 5 µM PCB29-pQ. Varieties of evidence have demonstrated that PCB29-pQ promotes foam cell formation and develops proinflammatory cascade and cell necroptosis. In detail, we observed that PCB29-pQ increased levels of total cholesterol (TC), free cholesterol (FC), triglyceride (TG), and cholesteryl ester (CE) by increasing the cholesterol influx and reducing the cholesterol efflux. Moreover, we found that PCB29-pQ induced inflammatory cytokines, such as tumor necrosis factor (TNF-α), interleukin 6 (IL-6), and IL-1ß, released by activating the mitogen-activated protein kinase (MAPK)-nuclear factor kappa B (NF-κB) inflammatory pathway. In addition, we demonstrated that PCB29-pQ induced cell necroptosis via receptor interacting protein kinases 1 and 3 (RIPK1/3) and a mixed-lineage kinase domain-like (MLKL) pathway. Finally, the overproduction of reactive oxygen species (ROS) by PCB29-pQ played significant roles in these processes, which could be reversed with an antioxidant. Overall, our results indicated that PCB29-pQ promoted the macrophage formation of foam cells, inflammation, and cell necroptosis.


Subject(s)
Benzoquinones/toxicity , Cell Differentiation/drug effects , Environmental Pollutants/toxicity , Foam Cells/metabolism , Macrophages/drug effects , Polychlorinated Biphenyls/toxicity , Animals , Atherosclerosis/etiology , Cytokines/metabolism , Mice , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B p50 Subunit/metabolism , Necroptosis/drug effects , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
20.
Chem Res Toxicol ; 32(12): 2509-2516, 2019 12 16.
Article in English | MEDLINE | ID: mdl-31687807

ABSTRACT

Polybrominated diphenyl ethers (PBDEs) have been detected ubiquitously in biological and environmental samples. Growing epidemiological data suggested the obvious correlation of PBDEs exposure with adverse health outcomes toward human beings, but exact molecular mechanism(s) are limited. Especially, the toxicological information regarding PBDEs metabolites is missing. Thereafter, this study intends to explore unidentified cell death modalities caused by PBDEs reactive quinone-type metabolite, PBDEQ. We found that PBDEQ induces autophagy in an ROS-dependent manner. Interestingly, the results indicated that PBDEQ degraded ferritin and activated a selective autophagy (termed as ferritinophagy) by using NCOA4 as its cargo receptor. These processes may further promote the release of iron and ROS. These results suggested the incidence of ferritinophagy induced by PBDEQ, which may contribute to PBDE exposure-caused diseases and dysfunctions.


Subject(s)
Autophagy/drug effects , Ferritins/metabolism , Halogenated Diphenyl Ethers/toxicity , Nuclear Receptor Coactivators/metabolism , Quinones/toxicity , Animals , Ferroptosis/drug effects , Iron/metabolism , PC12 Cells , Rats , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL