Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neurotoxicology ; 102: 1-11, 2024 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-38461971

RESUMO

Although overexposure to manganese (Mn) is known to cause neurotoxic damage, effective exposure markers for assessing Mn loading in Mn-exposed workers are lacking. Here, we construct a Mn-exposed rat model to perform correlation analysis between Mn-induced neurological damage and Mn levels in various biological samples. We combine this analysis with epidemiological investigation to assess whether Mn concentrations in red blood cells (MnRBCs) and urine (MnU) can be used as valid exposure markers. The results show that Mn exposure resulted in neurotoxic damage in rats and that MnRBCs correlated well with neurological damage, showing potential as a novel Mn exposure biomarker. These findings provide a basis for health monitoring of Mn-exposed workers and the development of more appropriate biological exposure limits.

2.
Ecotoxicol Environ Saf ; 272: 116029, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38290312

RESUMO

Manganese is essential trace elements, to participate in the body a variety of biochemical reactions, has important physiological functions, such as stimulate the immune cell proliferation, strengthen the cellular immunity, etc. However, excessive manganese exposure can cause damage to multiple systems of the body.The immune system is extremely vulnerable to external toxicants, however manganese research on the immune system are inadequate and biomarkers are lacking. Therefore, here we applied a manganese-exposed rat model to make preliminary observations on the immunotoxic effects of manganese. We found that manganese exposure inhibited humoral immune function in rats by decreasing peripheral blood IgG (ImmunoglobulinG, IgG), IgM (ImmunoglobulinM, IgM) and complement C3 levels; It also regulates rat cellular immune activity by influencing peripheral blood, spleen, and thymus T cell numbers and immune organ ICs (Immune Checkpoints, ICs) and cytokine expression. Furthermore, it was revealed that the impact of manganese exposure on the immune function of rats exhibited a correlation with both the dosage and duration of exposure. Notably, prolonged exposure to high doses of manganese had the most pronounced influence on rat immune function, primarily manifesting as immunosuppression.The above findings suggest that manganese exposure leads to impaired immune function and related changes in immune indicators, or may provide clues for the discovery of its biomarkers.


Assuntos
Manganês , Linfócitos T , Ratos , Animais , Manganês/toxicidade , Imunoglobulina M , Imunoglobulina G , Biomarcadores
3.
Front Public Health ; 11: 1289838, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38026392

RESUMO

Mn (Manganese, Mn) is an essential trace element involved in various biological processes such as the regulation of immune, nervous and digestive system functions. However, excessive Mn exposure can lead to immune damage. Occupational workers in cement and ferroalloy manufacturing and other related industries are exposed to low levels of Mn for a long time. Mn exposure is one of the important occupational hazards, but the research on the effect of Mn on the immune system of the occupational population is not complete, and there is no reliable biomarker. Therefore, this study aimed to evaluate the immunotoxicity of Mn from the soluble immune checkpoint TIM-3 (T-cell immunoglobulin and mucin containing protein 3, TIM-3) and complement C3. A total of 144 Mn-exposed workers were recruited from a bus manufacturing company and a railroad company in Henan Province. An inductively coupled plasma mass spectrometer was used to detect the concentration of RBC Mn (Red blood cell Mn, RBC Mn), and ELISA kits were used to detect serum complement C3 and TIM-3. Finally, the subjects were statistically analyzed by dividing them into low and high Mn groups based on the median RBC Mn concentration. We found that Mn exposure resulted in elevated serum TIM-3 expression and decreased complement C3 expression in workers; that serum TIM-3 and complement C3 expression showed a dose-response relationship with RBC Mn; and that the mediating effect of complement C3 between RBC Mn and TIM-3 was found to be significant. The above findings indicate that this study has a preliminary understanding of the effect of Mn exposure on the immune system of the occupational population exposed to Mn, and complement C3 and TIM-3 may be biomarkers of Mn exposure, which may provide clues for the prevention and control of Mn occupational hazards.


Assuntos
Complemento C3 , Receptor Celular 2 do Vírus da Hepatite A , Humanos , Manganês/toxicidade , Biomarcadores
4.
Environ Toxicol Pharmacol ; 100: 104150, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37207490

RESUMO

This study evaluated the effects of BTEX exposure on oxidative stress; it analyzed the correlation between oxidative stress and peripheral blood counts and estimated the benchmark dose (BMD) of BTEX compounds. This study recruited 247 exposed workers and 256 controls; physical examination data were collected and serum oxidative stress levels were measured. Relationships between BTEX exposure and biomarkers were analyzed using Mann-Whitney U, generalized linear model, and chi-square trend tests. Environmental Protection Agency Benchmark Dose Software was used to calculate the BMD and lower confidence limit of the BMD (BMDL) for BTEX exposure. The total antioxidant capacity (T-AOC) correlated positively with peripheral blood counts, and negatively with the cumulative exposure dose. On using T-AOC as the outcome variable, the estimated BMD and BMDL for BTEX exposure were 3.57 mg/m3 and 2.20 mg/m3, respectively. Based on T-AOC, the calculated occupational exposure limit of BTEX was 0.055 mg/m3.


Assuntos
Benzeno , Exposição Ocupacional , Humanos , Benzeno/toxicidade , Benzeno/análise , Benchmarking , População do Leste Asiático , Exposição Ocupacional/efeitos adversos , Exposição Ocupacional/análise , Estresse Oxidativo , Antioxidantes , Derivados de Benzeno
5.
Shock ; 59(6): 955-965, 2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-37119808

RESUMO

ABSTRACT: Blast lung injuries (BLIs) are frequent because of industrial accidents and terrorist groups. Bone marrow mesenchymal stem cells (BMSCs) and exosomes derived from BMSCs (BMSCs-Exo) have become a hot topic in modern biology because of their significance in damage healing, immune regulation, and gene therapy. The aim of this study is to investigate the effect of BMSCs and BMSCs-Exo on BLI in rats caused by gas explosion. Here, BMSCs and BMSCs-Exo were transplanted into BLI rats via tail vein and then evaluated pathological alterations, oxidative stress, apoptosis, autophagy, and pyroptosis in the lung tissue. Through histopathology and changes in malondialdehyde (MDA) and superoxide dismutase (SOD) contents, we discovered that oxidative stress and inflammatory infiltration in the lungs were significantly reduced by BMSCs and BMSCs-Exo. After treatment with BMSCs and BMSCs-Exo, apoptosis-related proteins, such as cleaved caspase-3 and Bax, were significantly decreased, and the ratio of Bcl-2/Bax was significantly increased; the level of pyroptosis-associated proteins, including NLRP3, GSDMD-N, cleaved caspase-1, IL-1ß, and IL-18, were decreased; autophagy-related proteins, beclin-1 and LC3, were downregulated while P62 was upregulated; the number of autophagosomes was decreased. In summary, BMSCs and BMSCs-Exo attenuate BLI caused by gas explosion, which may be associated with apoptosis, aberrant autophagy, and pyroptosis.


Assuntos
Exossomos , Lesão Pulmonar , Células-Tronco Mesenquimais , Humanos , Lesão Pulmonar/metabolismo , Proteína X Associada a bcl-2/metabolismo , Exossomos/metabolismo , Células-Tronco Mesenquimais/metabolismo , Apoptose
6.
Exp Cell Res ; 424(2): 113522, 2023 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-36796747

RESUMO

High mobility group protein B1 (HMGB1), a highly conserved non-histone nuclear protein, is highly expressed in fibrotic diseases; however, the role of HMGB1 in pulmonary fibrosis has not been fully elucidated. In this study, an epithelial-mesenchymal transition (EMT) model was constructed using transforming growth factor-ß1 (TGF-ß1) to stimulate BEAS-2B cells in vitro, and HMGB1 was knocked down or overexpressed to observe its effects on cell proliferation, migration and EMT. Meanwhile, string system, immunoprecipitation and immunofluorescence analyses were applied to identify and examine the relationship between HMGB1 and its potential interacting protein Brahma-related gene 1 (BRG1), and to explore the mechanism of interaction between HMGB1 and BRG1 in EMT. The results indicate that exogenous increase in HMGB1 promotes cell proliferation and migration and facilitates EMT by enhancing the PI3K/Akt/mTOR signaling pathway, whereas silencing HMGB1 has the opposite effect. Mechanistically, HMGB1 exerts these functions by interacting with BRG1, which may enhance BRG1 function and activate the PI3K/Akt/mTOR signaling pathway, thereby promoting EMT. These results suggest that HMGB1 is important for EMT and is a potential therapeutic target for the treatment of pulmonary fibrosis.


Assuntos
Proteína HMGB1 , Fibrose Pulmonar , Humanos , Transição Epitelial-Mesenquimal , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fibrose Pulmonar/metabolismo , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Fator de Crescimento Transformador beta1/metabolismo
7.
Biomed Pharmacother ; 155: 113638, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36099794

RESUMO

Silicosis is an incurable lung disease that can progress even when exposure to silica dust has ended. Lipid metabolism plays an important role in the occurrence and development of silicosis. However, the mechanistic details have not been fully elucidated. This was investigated in the current study by high-performance liquid chromatography-mass spectrometry-based lipidomic analysis of lung tissue in a mouse model of silicosis. Lipid profiles and key metabolic enzymes were compared between silica and control groups. The lipidomic analysis revealed differentially-expressed lipids in the lungs of silicosis mice compared with controls. Among the identified lipid metabolism-related enzymes, the expression of lysophosphatidylcholine acyltransferase 1 (LPCAT1) was significantly down-regulated at the transcript and protein levels. LPCAT1 overexpression in vivo using adeno-associated virus altered the balance between phosphatidylcholine and lysophosphatidylcholine and inhibited the development of silicosis in mice. These results indicate that LPCAT1 dysregulation leads to abnormal lipid metabolism and silicosis, and is a potential therapeutic target for the treatment of silica-induced pulmonary fibrosis.


Assuntos
Fibrose Pulmonar , Silicose , Animais , Camundongos , 1-Acilglicerofosfocolina O-Aciltransferase/genética , 1-Acilglicerofosfocolina O-Aciltransferase/metabolismo , Aciltransferases/metabolismo , Poeira , Metabolismo dos Lipídeos , Lisofosfatidilcolinas , Fosfatidilcolinas/uso terapêutico , Fibrose Pulmonar/induzido quimicamente , Dióxido de Silício/toxicidade , Silicose/tratamento farmacológico
8.
Artigo em Inglês | MEDLINE | ID: mdl-35886594

RESUMO

PURPOSE: High mobility group protein 1 (HMGB1) is a highly conserved DNA-binding nuclear protein that participates in the occurrence and development of silicosis. HMGB1 binds to its specific receptor and activates phosphatidylinositol 3-kinase (PI3K)/protein kinase B, (PKB; Akt)/mammalian target of rapamycin (mTOR) pathway. Brahma-related genes 1 (BRG1; SMARCA4) is the core subunit of SWI/SNF. HMGB1 activates the Akt pathway through BRG1 to promote the proliferation of prostate cancer. Glycyrrhizic acid is a new pharmacological inhibitor of HMGB1, which may inhibit the occurrence and development of silicosis. We speculate that glycyrrhizic acid inhibits the interaction between HMGB1 and BRG1 through the PI3K/Akt/mTOR pathway to affect the progression of silicosis. METHODS: We carried out an in vitro study and stimulated A549 with TGF-ß1 to establish an epithelial-mesenchymal transition (EMT) model, knocked down the HMGB1 and BRG1 genes in cells, observed the expression of EMT markers, and detected the interaction between HMGB1 and BRG1 by co-immunoprecipitation. In vivo, we injected glycyrrhizic acid into the mouse silicosis model to inhibit the expression of HMGB1. RESULTS: Both HMGB1 and BRG1 were highly expressed in the process of EMT. After knocking down HMGB1 and BRG1, the process of EMT was inhibited through the PI3K/Akt/mTOR pathway, and their expressions were influenced by each other. HMGB1 and BRG1 interact with each other in A549 cells. HMGB1 and BRG1 are also highly expressed in the mouse silicosis model, and glycyrrhizic acid can inhibit the expression of HMGB1/BRG1 through the PI3K/Akt/mTOR pathway. CONCLUSION: Glycyrrhizic acid can inhibit the interaction between HMGB1 and BRG1 through the PI3K/Akt/mTOR pathway to affect the progression of silicosis.


Assuntos
Proteína HMGB1 , Fibrose Pulmonar , Silicose , Animais , Proliferação de Células , Ácido Glicirrízico/farmacologia , Ácido Glicirrízico/uso terapêutico , Proteína HMGB1/genética , Masculino , Mamíferos/metabolismo , Camundongos , Proteínas Nucleares/genética , Fosfatidilinositol 3-Quinase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Silicose/tratamento farmacológico , Serina-Treonina Quinases TOR/metabolismo
9.
Artigo em Inglês | MEDLINE | ID: mdl-35742351

RESUMO

The purpose of this study was to investigate the expression of immune checkpoint cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and T cell immunoglobulin and mucin domain 3 (TIM-3) in the peripheral blood T lymphocytes of medical radiologists. The study incorporated 100 male medical radiologists and 107 male healthy controls. The expressions of CTLA-4 and TIM-3 among CD4+ and CD8+ lymphocytes were detected by flow cytometry. The expression levels of CTLA-4 and TIM-3 in the CD4+T cells of radiation workers were lower than those of healthy controls (p < 0.05). Correlation analysis showed that the CD8+CTLA-4 expression level was significantly positively correlated with individual cumulative dose (rs = 0.260, p = 0.001, <0.05), while the expression level of CD8+TIM-3 was negatively correlated (rs = −0.180, p = 0.027, <0.05). Low-dose radiation exposure affects the expression of CTLA-4 and TIM-3 in human peripheral blood T lymphocytes. Future studies need to focus on exploring the mechanisms by which CTLA-4 and TIM-3 expression changes in response to low-dose radiation exposure.


Assuntos
Linfócitos T CD8-Positivos , Receptor Celular 2 do Vírus da Hepatite A , Linfócitos T CD4-Positivos/metabolismo , Antígeno CTLA-4 , Receptor Celular 2 do Vírus da Hepatite A/metabolismo , Humanos , Masculino , Radiologistas
10.
Artigo em Inglês | MEDLINE | ID: mdl-35682354

RESUMO

BACKGROUND: Excessive accumulation of extracellular matrix is a key feature of pulmonary fibrosis (PF), and myofibroblasts are the main producers of extracellular matrix. Fibroblasts are the major source of myofibroblasts, but the mechanisms of transdifferentiation are unclear. METHODS: In vitro, transforming growth factor-ß1 was used to induce NIH-3T3 cell transdifferentiation. DMOG was used to increase hypoxia-inducible factor-1α subunit (HIF-1α) expression. KC7F2 and siRNA decreased HIF-1α expression. In vivo, silica particles were used to induce PF in C57BL/6N mice, and KC7F2 was used to reduce HIF-1α expression in C57BL/6N mice. Western blot was used to detect the expression of collagen type 1 alpha 1(COL1A1), α-smooth muscle actin (α-SMA), SMAD family member (SAMD) 3, Phospho-SMAD3 (PSMAD3), and HIF-1α. PCR was used to detect the expression of COL1A1, α-SMA, and HIF-1α. Immunohistochemistry was used to detect the expression of COL1A1 and HIF-1α. RESULTS: In vitro, compared to the control group, COL1A1, α-SMA, PSMAD3, and HIF-1α expression were elevated in the DMOG group, and COL1A1, α-SMA, PSMAD3, and HIF-1α expression were decreased in the KC7F2 group and siRNA group. Compared to the DMOG group, COL1A1, α-SMA, and PSMAD3 expression were decreased in the DMOG + SIS3 group. In vivo, compared to the saline group, COL1A1, α-SMA, PSMAD3, and HIF-1α expression were increased in the pulmonary tissue of C57BL/6N mice in the silica group. Compared to the silica group, COL1A1, α-SMA, PSMAD3, and HIF-1α expression and the degree of PF were decreased in the silica + KC7F2 group. CONCLUSION: Inhibition of HIF-1α reduced α-SMA, decreased COL1A1 expression, and attenuated the degree of PF in C57BL/6N mice. Therefore, HIF-1α may be a new target for the treatment of silica-induced PF.


Assuntos
Fibrose Pulmonar , Animais , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Camundongos , Camundongos Endogâmicos C57BL , Fibrose Pulmonar/induzido quimicamente , RNA Interferente Pequeno/genética , Dióxido de Silício/toxicidade
11.
Biomed Pharmacother ; 148: 112768, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35247717

RESUMO

Pulmonary fibrosis induced by silica particles is defined as silicosis, which is an incurable disease. The pathogenesis of silicosis is not completely clear, but it's certain that immune system dysfunction is closely related to it. Immune checkpoint inhibitors (ICIs) are emerging immunotherapeutic agents that mainly target adaptive immune cells, and there is abundant evidence that ICIs are of great value in cancer treatment. However, whether these attractive agents can be implemented in silicosis treatment is unclear. In this study, we explored the efficacy of small molecule inhibitors targeted PD-1/PD-L1 and CTLA-4 on silica-induced pulmonary fibrosis in mice. ICIs were injected intraperitoneally into mice that received silica instillation twice a week. The mice were sacrificed 7 and 28 days after the injection. The lungs, spleen, hilar lymph nodes, thymus, and peripheral blood of mice were collected and subjected to histological examination, flow cytometry analysis, and mRNA and protein quantification. Our results demonstrated that silica exposure caused damage to multiple immune organs in mice, leading to an imbalance in systemic immune homeostasis. Specifically, proportions and subtypes of T and B cells were significantly altered, and the expressions of PD-1, PD-L1 and CTLA-4 were abnormal on these cells. Both PD-1/PD-L1 and CTLA-4 inhibitor administration modulated silica-induced immune system disruption, however, only PD-1/PD-L1 signaling inhibition showed significant amelioration of silicosis. Our findings confirmed for the first time the potential value of ICIs for the treatment of silica-induced pulmonary fibrosis, and this may provide new ideas for the treatment of other fibrosis-related diseases.


Assuntos
Inibidores de Checkpoint Imunológico/farmacologia , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/patologia , Dióxido de Silício/efeitos adversos , Animais , Subpopulações de Linfócitos B/efeitos dos fármacos , Antígeno B7-H1/efeitos dos fármacos , Antígeno CTLA-4/efeitos dos fármacos , Homeostase/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Insuficiência de Múltiplos Órgãos/induzido quimicamente , Insuficiência de Múltiplos Órgãos/patologia , Receptor de Morte Celular Programada 1/efeitos dos fármacos , RNA Mensageiro , Subpopulações de Linfócitos T/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...