Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Hazard Mater ; 430: 128485, 2022 05 15.
Article in English | MEDLINE | ID: mdl-35739668

ABSTRACT

As a newly emerging hazardous material, airborne nanoplastics are easily inhaled and accumulated in human and animal alveoli. We previously found that polystyrene nanoplastics (PS-NPs) induced apoptosis and inflammation of human alveolar epithelial A549 cells, implying they increase the risk of pulmonary fibrosis. In this study, we investigated whether PS-NPs induce epithelial-to-mesenchymal transition (EMT), the prelude to lung fibrosis, in A549 cells. A549 cells treated with PS-NPs of different sizes and surface charges exhibited increased migration and EMT markers accompanied with up-regulation of reactive oxygen species (ROS) and NADPH oxidase 4 (NOX4), an ROS generator located in the mitochondria and endoplasmic reticulum (ER). Moreover, PS-NPs caused mitochondrial dysfunction as demonstrated by membrane potential changes and impaired cellular energy metabolism. PS-NPs also activated ER stress as indicated by the up-regulated ER stress markers. As expected, smaller PS-NPs with a positive surface charge had stronger effects. Furthermore, the effects of PS-NPs on A549 cells were reversed by NOX4 gene knock-down, which verified the involvement of NOX4. Our results suggest that PS-NPs induce EMT in A549 cells through multiple mechanisms, and NOX4 is a key mediator in this process. Our findings contribute to understanding the toxicological mechanisms of nanoplastics on the respiratory system.


Subject(s)
Microplastics , Pulmonary Fibrosis , A549 Cells , Animals , Epithelial-Mesenchymal Transition , Humans , Polystyrenes , Reactive Oxygen Species/metabolism
2.
J Hazard Mater ; 424(Pt B): 127508, 2022 02 15.
Article in English | MEDLINE | ID: mdl-34688005

ABSTRACT

Nanoplastics can be ingested by organisms and penetrate biological barriers to affect multiple physiological functions. However, few studies have focused on the effects of nanoplastics on the mammalian immune system. We evaluated the effects and underlying mechanism of nanoplastics of varying particle sizes and surface charges on murine splenic lymphocytes. We found that nanoplastics penetrated into splenic lymphocytes and that nanoplastics of a diameter of 50 nm were absorbed more efficiently by the cells. The nanoplastics decreased cell viability, induce cell apoptosis, up-regulated apoptosis-related protein expression, elicited the production of reactive oxygen species, altered mitochondrial membrane potential, and impaired mitochondrial function. Positively charged nanoplastics exerted the strongest toxicity. Negatively charged and uncharged nanoplastics caused oxidative stress and mitochondrial structural damage in lymphocytes, while positively charged nanoplastics induced endogenous apoptosis directly. Moreover, nanoplastics inhibited the expression of activated T cell markers on the T cell surface, while inhibiting the differentiation of CD8+ T cells and the expression of helper T cell cytokines. In terms of the mechanism, a series of key signaling molecules in the pathways of T cell activation and function were markedly down-regulated after exposure to nanoplastics.


Subject(s)
Microplastics , Polystyrenes , Animals , CD8-Positive T-Lymphocytes , Mice , Particle Size , Reactive Oxygen Species
3.
Elife ; 102021 11 16.
Article in English | MEDLINE | ID: mdl-34783655

ABSTRACT

Influenza pandemics pose public health threats annually for lacking vaccine that provides cross-protection against novel and emerging influenza viruses. Combining conserved antigens that induce cross-protective antibody responses with epitopes that activate cross-protective T cell responses might be an attractive strategy for developing a universal vaccine. In this study, we constructed a recombinant protein named NMHC that consists of influenza viral conserved epitopes and a superantigen fragment. NMHC promoted the maturation of bone marrow-derived dendritic cells and induced CD4+ T cells to differentiate into Th1, Th2, and Th17 subtypes. Mice vaccinated with NMHC produced high levels of immunoglobulins that cross-bound to HA fragments from six influenza virus subtypes with high antibody titers. Anti-NMHC serum showed potent hemagglutinin inhibition effects to highly divergent group 1 (H1 subtype) and group 2 (H3 subtype) influenza virus strains. Furthermore, purified anti-NMHC antibodies bound to multiple HAs with high affinities. NMHC vaccination effectively protected mice from infection and lung damage when exposed to two subtypes of H1N1 influenza virus. Moreover, NMHC vaccination elicited CD4+ and CD8+ T cell responses that cleared the virus from infected tissues and prevented virus spread. In conclusion, this study provides proof of concept that NMHC vaccination triggers B and T cell immune responses against multiple influenza virus infections. Therefore, NMHC might be a candidate universal broad-spectrum vaccine for the prevention and treatment of multiple influenza viruses.


Subject(s)
Influenza A virus/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Recombinant Proteins/immunology , Animals , B-Lymphocytes/immunology , Cross Protection , Epitopes/immunology , Female , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Immunity, Cellular , Influenza A Virus, H1N1 Subtype/immunology , Influenza A virus/genetics , Mice, Inbred BALB C , Superantigens/immunology , T-Lymphocytes/immunology
4.
Biomed Pharmacother ; 143: 112204, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34560552

ABSTRACT

As a member of superantigens, Staphylococcal Enterotoxin C2 (SEC2) can potently activate T cells expressing specific Vß repertoires and has been applied in clinic for tumor immunotherapy in China for more than 20 years. However, excessive activation of T cells by over-stimulation with superantigen are always followed by eliciting regulatory T cells (Tregs) induction and functional immunosuppression, which brings uncertainties to SEC2 application in tumor immunotherapy. In this study, we found that SEC2 could induce CD4+CD25+Foxp3+ Tregs from the murine splenocytes in dose and time related manners. The induced Tregs with high expression of GITR and CTLA-4 and low expression of CD127 were TCR Vß8.2-specific and have character of IL-10 production in a SEC2 dose-depended manner. Importantly, SEC2-induced CD4+ Tregs showed the potent capacity of suppressing proliferation of intact murine splenocytes response to SEC2. Furthermore, by using specific inhibitors or neutralizing antibody, we proved that the signaling pathways of TCR-NFAT/AP-1, IL-2-STAT5, and TGF-ß-Smad3 play crucial roles in Tregs induction by SEC2. These findings will help us better understand the balance of immune stimulation and immunosuppression mediated by SEC2 and provide valuable guidance for SEC2 application in antitumor immunology.


Subject(s)
Enterotoxins/pharmacology , Signal Transduction/drug effects , T-Lymphocytes, Regulatory/drug effects , Animals , Cells, Cultured , Female , Immunophenotyping , Interleukin-10/metabolism , Interleukin-2/metabolism , Mice, Inbred BALB C , NFATC Transcription Factors/metabolism , Phenotype , Receptors, Antigen, T-Cell, alpha-beta/metabolism , STAT5 Transcription Factor/metabolism , Smad3 Protein/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Transcription Factor AP-1/metabolism , Transforming Growth Factor beta/metabolism
5.
Int J Pharm ; 586: 119498, 2020 Aug 30.
Article in English | MEDLINE | ID: mdl-32505575

ABSTRACT

Solid tumors are intrinsically resistant to immunotherapy because of the major challenges including the immunosuppression and poor penetration of drugs and lymphocytes into solid tumors due to the complicated tumor microenvironment (TME). Our previous study has created a novel superantigen mutant ST-4 to efficiently active the T lymphocytes and alleviate immune suppression. In the present study, to accumulate ST-4 into the TME, we constructed a recombinant protein, ST-4-iRGD, by fusing ST-4 to a tumor-homing peptide, iRGD. We hypothesized that ST-4-iRGD could internalize into the TME through iRGD-mediated tumor targeting and tumor tissue penetrating to activate the regional immunoreaction. The results of in vitro studies showed that ST-4-iRGD achieved improved tumor targeting and cytotoxicity in mouse B16F10 melanoma cells. The iRGD-mediated tumor tissue penetration was further confirmed by imaging and immunofluorescence studies in vivo, wherein higher distribution of ST-4-iRGD was observed in the mouse 4T1 breast tumor model. Moreover, ST-4-iRGD exhibited enhanced anti-solid tumor characteristics and induced improved lymphocyte infiltration in the B16F10 and 4T1 models. In conclusion, using iRGD to facilitate better dissemination of the therapeutic agent ST-4 throughout a solid tumor mass is feasible, and ST-4-iRGD may be a potential candidate for efficient cancer immunotherapy in the future.


Subject(s)
Breast Neoplasms/therapy , Immunotherapy/methods , Melanoma, Experimental/therapy , Oligopeptides/administration & dosage , Superantigens/administration & dosage , Animals , Breast Neoplasms/immunology , Cell Line, Tumor , Female , Melanoma, Experimental/immunology , Mice , Mice, Inbred BALB C , Mutation , Superantigens/genetics , Superantigens/metabolism , T-Lymphocytes/immunology , Tumor Microenvironment , Xenograft Model Antitumor Assays
6.
Sci Total Environ ; 694: 133794, 2019 Dec 01.
Article in English | MEDLINE | ID: mdl-31756791

ABSTRACT

As a kind of newly emerging pollutant, nanoplastics are easily to be ingested by organisms, and cause severe damage to biological functions because of their small size, high specific surface area, and strong biological penetration. Recently, there are increasing reports of numerous airborne microplastics, including polystyrene (PS), being detected in atmospheric samples, which implies a potential risk to the human respiratory system. In this work, we evaluated the effects of polystyrene nanoparticles of two different sizes (PS-NP25: 25 nm diameter and PS-NP70: 70 nm diameter) on the human alveolar epithelial A549 cell line including internalization, cell viability, cell cycle, apoptosis, and associated gene transcription and protein expression. Results showed that PS-NP25 was internalized more rapidly and efficiently into the cytoplasm of A549 than PS-NP70. PS-NPs significantly affected the cell viability, caused cell cycle S phrase arrest, activated inflammatory gene transcription, and changed the expression of proteins associated with cell cycle and pro-apoptosis. PS-NPs induced significant up-regulation of pro-inflammatory cytokines such as IL-8, NF-κB, and TNF-α, as well as pro-apoptotic proteins such as DR5, caspase-3, caspase-8, caspase-9, and cytochrome c, which revealed that PS-NPs triggered a TNF-α-associated apoptosis pathway. This study suggests that exposure duration, diameter, and concentration are the key factors for evaluating the toxicological effects of PS-NPs on alveolar epithelial cells. More attention must be focused on the risk of nanoplastic-related air pollution and the environmental toxicological effects of nanoplastics on humans and other terrestrial mammals.


Subject(s)
Environmental Pollutants/toxicity , Nanoparticles/toxicity , Plastics/toxicity , Cell Line , Epithelial Cells , Humans , Lung , Toxicity Tests
SELECTION OF CITATIONS
SEARCH DETAIL
...