1
|
Zhao M, Chen Q, Chen X, Gong S, Wang M, Zhao S, Wang S, Du W, Xu Y, Peng L, Yao Y. Tripartite motif-containing 32 regulated by miR-6236-p5 inhibited silica-induced apoptosis of alveolar macrophages. Toxicology 2024; 511:154042. [PMID: 39742911 DOI: 10.1016/j.tox.2024.154042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/19/2024] [Accepted: 12/29/2024] [Indexed: 01/04/2025]
Abstract
Apoptosis of alveolar macrophages (AMs) induced by silica is one of the crucial driving factors of silicosis inflammation and fibrosis. However, the mechanism of silica-induced AMs apoptosis remains unclear. In this study, transcriptome sequencing identified 11 differentially expressed (DE)-mRNAs enriched in the regulation of apoptotic signaling pathways in AMs treated with 250μg/mL silica for 24h, of which tripartite motif-containing 32 (Trim32) was the most significant and down-regulated. The decreased Trim32 promoted AMs apoptosis, while Trim32 overexpression inhibited the apoptosis of AMs induced by silica at 250μg/mL for 24h. MiR-6236-p5 was then identified by MiRNA sequencing as the most significant DE-miRNA potentially regulating Trim32 expression, and the interaction between miR-6236-p5 and Trim32 3'-UTR was confirmed by dual luciferase reporter gene assay. Treated with 100nM miR-6236-p5 inhibitor increased the expression of Trim32 and inhibited the apoptosis of AMs induced by silica at 250μg/mL for 24h, while miR-6236-p5 mimic promoted the apoptosis of silica-induced AMs. In conclusion, this study identified Trim32 regulated by miR-6236-p5 played an important role in silica-induced AMs apoptosis based on RNA sequencing, which provided a novel clue for exploring the mechanism of silica-induced AMs apoptosis.
Collapse
Affiliation(s)
- Manyu Zhao
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China; Laboratory of Precision Therapeutics, Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qing Chen
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China
| | - Xuxi Chen
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China
| | - Shuyu Gong
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China
| | - Mengzhu Wang
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China
| | - Shanshan Zhao
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China
| | - Sihan Wang
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China
| | - Wen Du
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China; West China Occupational Pneumoconiosis Cohort Study (WCOPCS) working group, Research Center for Prevention and Therapy of Occupational Disease, West China-PUMC C.C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China
| | - Yunyi Xu
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China
| | - Lijun Peng
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China; West China Occupational Pneumoconiosis Cohort Study (WCOPCS) working group, Research Center for Prevention and Therapy of Occupational Disease, West China-PUMC C.C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China.
| | - Yuqin Yao
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China; West China Occupational Pneumoconiosis Cohort Study (WCOPCS) working group, Research Center for Prevention and Therapy of Occupational Disease, West China-PUMC C.C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
2
|
Jin X, Chen Y, Xu B, Tian H. Exercise-Mediated Protection against Air Pollution-Induced Immune Damage: Mechanisms, Challenges, and Future Directions. BIOLOGY 2024; 13:247. [PMID: 38666859 PMCID: PMC11047937 DOI: 10.3390/biology13040247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/29/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024]
Abstract
Air pollution, a serious risk factor for human health, can lead to immune damage and various diseases. Long-term exposure to air pollutants can trigger oxidative stress and inflammatory responses (the main sources of immune impairment) in the body. Exercise has been shown to modulate anti-inflammatory and antioxidant statuses, enhance immune cell activity, as well as protect against immune damage caused by air pollution. However, the underlying mechanisms involved in the protective effects of exercise on pollutant-induced damage and the safe threshold for exercise in polluted environments remain elusive. In contrast to the extensive research on the pathogenesis of air pollution and the preventive role of exercise in enhancing fitness, investigations into exercise resistance to injury caused by air pollution are still in their infancy. In this review, we analyze evidence from humans, animals, and cell experiments on the combined effects of exercise and air pollution on immune health outcomes, with an emphasis on oxidative stress, inflammatory responses, and immune cells. We also propose possible mechanisms and directions for future research on exercise resistance to pollutant-induced damage in the body. Furthermore, we suggest strengthening epidemiological studies at different population levels and investigations on immune cells to guide how to determine the safety thresholds for exercise in polluted environments.
Collapse
Affiliation(s)
| | | | - Bingxiang Xu
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (X.J.); (Y.C.)
| | - Haili Tian
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (X.J.); (Y.C.)
| |
Collapse
|
3
|
Zhao M, Wang M, Chen X, Gao Y, Chen Q, Wang L, Bao Q, Sun D, Du W, Xu Y, Xie L, Jiang X, Zhang L, Peng L, Zhang B, Yao Y. Targeting progranulin alleviated silica particles-induced pulmonary inflammation and fibrosis via decreasing Il-6 and Tgf-β1/Smad. JOURNAL OF HAZARDOUS MATERIALS 2024; 465:133199. [PMID: 38103296 DOI: 10.1016/j.jhazmat.2023.133199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 10/12/2023] [Accepted: 12/05/2023] [Indexed: 12/19/2023]
Abstract
Long term exposure to silica particles leads to various diseases, among which silicosis is of great concern. Silicosis is an interstitial lung disease caused by inhalation of silica particles in production environments. However, the mechanisms underlying silicosis remains unclear. Our previous studies revealed that progranulin (Pgrn) promoted the expression of pro-inflammatory factors in alveolar macrophages treated with silica particles and the secretion of extracellular matrix of pulmonary fibroblasts. Nevertheless, the role of Pgrn in silica particles-induced silicosis in vivo was unknown. This study found that silica particles increased Pgrn expression in silicosis patients. Pgrn deficiency reduced lung inflammation and fibrosis in silica particles-induced silicosis mouse models. Subsequently, based on transcriptional sequencing and interleukin (Il) -6 knockout mouse models, results demonstrated that Pgrn deficiency might decrease silicosis inflammation by reducing the production of Il-6, thereby modulating pulmonary fibrosis in the early stage of silicosis mouse models. Furthermore, another mechanism through which Pgrn deficiency reduced fibrosis in silicosis mouse models was the regulation of the transforming growth factor (Tgf) -β1/Smad signaling pathway. Conclusively, Pgrn contributed to silicosis inflammation and fibrosis induced by silica particles, indicating that Pgrn could be a promising therapeutic target.
Collapse
Affiliation(s)
- Manyu Zhao
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Mengzhu Wang
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Xuxi Chen
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Ying Gao
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Qing Chen
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Liqun Wang
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Qixue Bao
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Donglei Sun
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Wen Du
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China; West China Occupational Pneumoconiosis Cohort Study (WCOPCS) working group, Research Center For Prevention and Therapy of Occupational Disease, West China-PUMC C.C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Yunyi Xu
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Linshen Xie
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China; West China Occupational Pneumoconiosis Cohort Study (WCOPCS) working group, Research Center For Prevention and Therapy of Occupational Disease, West China-PUMC C.C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Xia Jiang
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China; West China Occupational Pneumoconiosis Cohort Study (WCOPCS) working group, Research Center For Prevention and Therapy of Occupational Disease, West China-PUMC C.C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Ling Zhang
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China; West China Occupational Pneumoconiosis Cohort Study (WCOPCS) working group, Research Center For Prevention and Therapy of Occupational Disease, West China-PUMC C.C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Lijun Peng
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China; West China Occupational Pneumoconiosis Cohort Study (WCOPCS) working group, Research Center For Prevention and Therapy of Occupational Disease, West China-PUMC C.C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Ben Zhang
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China; West China Occupational Pneumoconiosis Cohort Study (WCOPCS) working group, Research Center For Prevention and Therapy of Occupational Disease, West China-PUMC C.C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China; Departments of Cardiology, Neurology, and Oncology, Hainan General Hospital and Hainan Affiliated Hospital, Hainan Medical University, Haikou 570311, China.
| | - Yuqin Yao
- Molecular Toxicology Laboratory of Sichuan Provincial Education office, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China; West China Occupational Pneumoconiosis Cohort Study (WCOPCS) working group, Research Center For Prevention and Therapy of Occupational Disease, West China-PUMC C.C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
4
|
Bredeck G, Dobner J, Stahlmecke B, Fomba KW, Herrmann H, Rossi A, Schins RPF. Saharan dust induces NLRP3-dependent inflammatory cytokines in an alveolar air-liquid interface co-culture model. Part Fibre Toxicol 2023; 20:39. [PMID: 37864207 PMCID: PMC10588053 DOI: 10.1186/s12989-023-00550-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 10/09/2023] [Indexed: 10/22/2023] Open
Abstract
BACKGROUND Epidemiological studies have related desert dust events to increased respiratory morbidity and mortality. Although the Sahara is the largest source of desert dust, Saharan dust (SD) has been barely examined in toxicological studies. Here, we aimed to assess the NLRP3 inflammasome-caspase-1-pathway-dependent pro-inflammatory potency of SD in comparison to crystalline silica (DQ12 quartz) in an advanced air-liquid interface (ALI) co-culture model. Therefore, we exposed ALI co-cultures of alveolar epithelial A549 cells and macrophage-like differentiated THP-1 cells to 10, 21, and 31 µg/cm² SD and DQ12 for 24 h using a Vitrocell Cloud system. Additionally, we exposed ALI co-cultures containing caspase (CASP)1-/- and NLRP3-/- THP-1 cells to SD. RESULTS Characterization of nebulized DQ12 and SD revealed that over 90% of agglomerates of both dusts were smaller than 2.5 μm. Characterization of the ALI co-culture model revealed that it produced surfactant protein C and that THP-1 cells remained viable at the ALI. Moreover, wild type, CASP1-/-, and NLRP3-/- THP-1 cells had comparable levels of the surface receptors cluster of differentiation 14 (CD14), toll-like receptor 2 (TLR2), and TLR4. Exposing ALI co-cultures to non-cytotoxic doses of DQ12 and SD did not induce oxidative stress marker gene expression. SD but not DQ12 upregulated gene expressions of interleukin 1 Beta (IL1B), IL6, and IL8 as well as releases of IL-1β, IL-6, IL-8, and tumor necrosis factor α (TNFα). Exposing wild type, CASP1-/-, and NLRP3-/- co-cultures to SD induced IL1B gene expression in all co-cultures whereas IL-1β release was only induced in wild type co-cultures. In CASP1-/- and NLRP3-/- co-cultures, IL-6, IL-8, and TNFα releases were also reduced. CONCLUSIONS Since surfactants can decrease the toxicity of poorly soluble particles, the higher potency of SD than DQ12 in this surfactant-producing ALI model emphasizes the importance of readily soluble SD components such as microbial compounds. The higher potency of SD than DQ12 also renders SD a potential alternative particulate positive control for studies addressing acute inflammatory effects. The high pro-inflammatory potency depending on NLRP3, CASP-1, and IL-1β suggests that SD causes acute lung injury which may explain desert dust event-related increased respiratory morbidity and mortality.
Collapse
Affiliation(s)
- Gerrit Bredeck
- IUF - Leibniz Research Institute for Environmental Medicine, 40225, Düsseldorf, Germany.
| | - Jochen Dobner
- IUF - Leibniz Research Institute for Environmental Medicine, 40225, Düsseldorf, Germany
| | - Burkhard Stahlmecke
- Institut für Umwelt & Energie, Technik & Analytik e. V. (IUTA), 47229, Duisburg, Germany
| | - Khanneh Wadinga Fomba
- Atmospheric Chemistry Department (ACD), Leibniz Institute for Tropospheric Research (TROPOS), 04318, Leipzig, Germany
| | - Hartmut Herrmann
- Atmospheric Chemistry Department (ACD), Leibniz Institute for Tropospheric Research (TROPOS), 04318, Leipzig, Germany
| | - Andrea Rossi
- IUF - Leibniz Research Institute for Environmental Medicine, 40225, Düsseldorf, Germany
| | - Roel P F Schins
- IUF - Leibniz Research Institute for Environmental Medicine, 40225, Düsseldorf, Germany
| |
Collapse
|
5
|
Plasma Cytokine Profiling Reveals Differences between Silicotic Patients with Simple Silicosis and Those with Progressive Massive Fibrosis Caused by Engineered Stone. Int J Mol Sci 2023; 24:ijms24021541. [PMID: 36675056 PMCID: PMC9860830 DOI: 10.3390/ijms24021541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/05/2023] [Accepted: 01/11/2023] [Indexed: 01/14/2023] Open
Abstract
Engineered stone silicosis has become an occupational epidemic disease that progresses rapidly to progressive massive fibrosis with respiratory failure and death, and there is no effective treatment. Silica deposition in the lung triggers a series of inflammatory reactions with the participation of multiple cytokines and cellular mediators whose role in the development and progression of the disease is largely unknown. We hypothesized that differences in plasma cytokine levels exist between patients diagnosed with simple silicosis (SS) and patients diagnosed with progressive massive fibrosis (PMF). Plasma samples from 91 ES silicosis patients, diagnosed and classified by chest radiography and/or high-resolution computed tomography with SS (n = 53) and PMF (n = 38), were assayed by multiplex assays for levels of 34 cytokines. Additionally, a healthy volunteer control group (n = 22) was included. Plasma levels of a high number of cytokines were significantly higher in subjects with silicosis than in healthy control subjects. Moreover, the levels of IL-1RA, IL-8, IL-10, IL-16, IL-18, TNF-α, MIP-1α, G-CSF and VEGF were significantly elevated in PMF compared to SS patients. This study shows that plasma cytokine levels differ between healthy people and silicosis patients, and some of them are also significantly elevated in patients with PMF compared with patients with SS, which could indicate their involvement in the severity of the disease, be considered as biomarkers and could be explored as future therapeutic targets for the disease.
Collapse
|
6
|
Pulmonary Toxicity of Silica Linked to Its Micro- or Nanometric Particle Size and Crystal Structure: A Review. NANOMATERIALS 2022; 12:nano12142392. [PMID: 35889616 PMCID: PMC9318389 DOI: 10.3390/nano12142392] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/10/2022] [Accepted: 07/11/2022] [Indexed: 02/06/2023]
Abstract
Silicon dioxide (SiO2) is a mineral compound present in the Earth’s crust in two mineral forms: crystalline and amorphous. Based on epidemiological and/or biological evidence, the pulmonary effects of crystalline silica are considered well understood, with the development of silicosis, emphysema, chronic bronchitis, or chronic obstructive pulmonary disease. The structure and capacity to trigger oxidative stress are recognized as relevant determinants in crystalline silica’s toxicity. In contrast, natural amorphous silica was long considered nontoxic, and was often used as a negative control in experimental studies. However, as manufactured amorphous silica nanoparticles (or nanosilica or SiNP) are becoming widely used in industrial applications, these paradigms must now be reconsidered at the nanoscale (<100 nm). Indeed, recent experimental studies appear to point towards significant toxicity of manufactured amorphous silica nanoparticles similar to that of micrometric crystalline silica. In this article, we present an extensive review of the nontumoral pulmonary effects of silica based on in vitro and in vivo experimental studies. The findings of this review are presented both for micro- and nanoscale particles, but also based on the crystalline structure of the silica particles.
Collapse
|
7
|
Comparing α-Quartz-Induced Cytotoxicity and Interleukin-8 Release in Pulmonary Mono- and Co-Cultures Exposed under Submerged and Air-Liquid Interface Conditions. Int J Mol Sci 2022; 23:ijms23126412. [PMID: 35742856 PMCID: PMC9224477 DOI: 10.3390/ijms23126412] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/01/2022] [Accepted: 06/03/2022] [Indexed: 02/06/2023] Open
Abstract
The occupational exposure to particles such as crystalline quartz and its impact on the respiratory tract have been studied extensively in recent years. For hazard assessment, the development of physiologically more relevant in-vitro models, i.e., air-liquid interface (ALI) cell cultures, has greatly progressed. Within this study, pulmonary culture models employing A549 and differentiated THP-1 cells as mono-and co-cultures were investigated. The different cultures were exposed to α-quartz particles (Min-U-Sil5) with doses ranging from 15 to 66 µg/cm2 under submerged and ALI conditions and cytotoxicity as well as cytokine release were analyzed. No cytotoxicity was observed after ALI exposure. Contrarily, Min-U-Sil5 was cytotoxic at the highest dose in both submerged mono- and co-cultures. A concentration-dependent release of interleukin-8 was shown for both exposure types, which was overall stronger in co-cultures. Our findings showed considerable differences in the toxicological responses between ALI and submerged exposure and between mono- and co-cultures. A substantial influence of the presence or absence of serum in cell culture media was noted as well. Within this study, the submerged culture was revealed to be more sensitive. This shows the importance of considering different culture and exposure models and highlights the relevance of communication between different cell types for toxicological investigations.
Collapse
|
8
|
Skuland T, Låg M, Gutleb AC, Brinchmann BC, Serchi T, Øvrevik J, Holme JA, Refsnes M. Pro-inflammatory effects of crystalline- and nano-sized non-crystalline silica particles in a 3D alveolar model. Part Fibre Toxicol 2020; 17:13. [PMID: 32316988 PMCID: PMC7175518 DOI: 10.1186/s12989-020-00345-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 04/07/2020] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Silica nanoparticles (SiNPs) are among the most widely manufactured and used nanoparticles. Concerns about potential health effects of SiNPs have therefore risen. Using a 3D tri-culture model of the alveolar lung barrier we examined effects of exposure to SiNPs (Si10) and crystalline silica (quartz; Min-U-Sil) in the apical compartment consisting of human alveolar epithelial A549 cells and THP-1-derived macrophages, as well as in the basolateral compartment with Ea.hy926 endothelial cells. Inflammation-related responses were measured by ELISA and gene expression. RESULTS Exposure to both Si10 and Min-U-Sil induced gene expression and release of CXCL8, interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), interleukin-1α (IL-1α) and interleukin-1β (IL-1β) in a concentration-dependent manner. Cytokine/chemokine expression and protein levels were highest in the apical compartment. Si10 and Min-U-Sil also induced expression of adhesion molecules ICAM-1 and E-selectin in the apical compartment. In the basolateral endothelial compartment we observed marked, but postponed effects on expression of all these genes, but only at the highest particle concentrations. Geneexpressions of heme oxygenase-1 (HO-1) and the metalloproteases (MMP-1 and MMP-9) were less affected. The IL-1 receptor antagonist (IL-1RA), markedly reduced effects of Si10 and Min-U-Sil exposures on gene expression of cytokines and adhesion molecules, as well as cytokine-release in both compartments. CONCLUSIONS Si10 and Min-U-Sil induced gene expression and release of pro-inflammatory cytokines/adhesion molecules at both the epithelial/macrophage and endothelial side of a 3D tri-culture. Responses in the basolateral endothelial cells were only induced at high concentrations, and seemed to be mediated by IL-1α/β released from the apical epithelial cells and macrophages.
Collapse
Affiliation(s)
- Tonje Skuland
- Section of Air Pollution and Noise, Department of Environment and Health, Norwegian Institute of Public Health, PO Box 4404 Nydalen, N-0403, Oslo, Norway.
| | - Marit Låg
- Section of Air Pollution and Noise, Department of Environment and Health, Norwegian Institute of Public Health, PO Box 4404 Nydalen, N-0403, Oslo, Norway
| | - Arno C Gutleb
- Environmental Research and Innovation (ERIN), Luxembourg Institute of Science and Technology (LIST), Belvaux, Grand Duchy of Luxembourg, Luxembourg
| | - Bendik C Brinchmann
- Section of Air Pollution and Noise, Department of Environment and Health, Norwegian Institute of Public Health, PO Box 4404 Nydalen, N-0403, Oslo, Norway
- Department of Occupational Medicine and Epidemiology, National Institute of Occupational Health, Oslo, Norway
| | - Tommaso Serchi
- Environmental Research and Innovation (ERIN), Luxembourg Institute of Science and Technology (LIST), Belvaux, Grand Duchy of Luxembourg, Luxembourg
| | - Johan Øvrevik
- Section of Air Pollution and Noise, Department of Environment and Health, Norwegian Institute of Public Health, PO Box 4404 Nydalen, N-0403, Oslo, Norway
- Department of Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Jørn A Holme
- Section of Air Pollution and Noise, Department of Environment and Health, Norwegian Institute of Public Health, PO Box 4404 Nydalen, N-0403, Oslo, Norway
| | - Magne Refsnes
- Section of Air Pollution and Noise, Department of Environment and Health, Norwegian Institute of Public Health, PO Box 4404 Nydalen, N-0403, Oslo, Norway
| |
Collapse
|
9
|
Ambient fine particulate matter induce toxicity in lung epithelial-endothelial co-culture models. Toxicol Lett 2019; 301:133-145. [DOI: 10.1016/j.toxlet.2018.11.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 11/13/2018] [Accepted: 11/22/2018] [Indexed: 01/09/2023]
|
10
|
Oh H, Kim CY, Ryu B, Kim U, Kim J, Lee JM, Lee BH, Moon J, Jung CR, Park JH. Respiratory Toxicity of Polyhexamethylene Guanidine Phosphate Exposure in Zebrafish. Zebrafish 2018; 15:460-472. [PMID: 30133415 DOI: 10.1089/zeb.2018.1571] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Humidifier disinfectants containing polyhexamethylene guanidine phosphate (PHMG-P) can induce pulmonary toxicity and has caused human casualties in South Korea since 2006. Thereby, the safety evaluation of household chemicals such as PHMG-P has garnered increased importance. However, many limitations, such as the lack of specialized facilities and animal welfare concerns associated with the use of murine models, persist. Zebrafish gills have high functional and structural similarity to mammalian lungs. Moreover, zebrafish are sensitive to toxic substances, resulting in changes in behavioral or ventilatory patterns. Based on these facts, in this study, we aimed to evaluate the pulmonary toxicity of PHMG-P in zebrafish. Zebrafish exposed to PHMG-P showed an increase in mRNA levels of inflammatory factors persisting for 28 days along with histopathologic changes in the gills. An exposure time-dependent alteration in infiltration of inflammatory cells and destruction of gill lamellae was observed. In addition, an increase in mRNA levels of fibrosis factors was observed in gills exposed to PHMG-P for 28 days, as assessed by collagen staining with Masson's trichrome. These results supported the cellular level results. Taken together, our results reveal pulmonary toxic effects of PHMG-P and suggest useful markers for evaluating pulmonary toxicity.
Collapse
Affiliation(s)
- Hanseul Oh
- 1 Department of Laboratory Animal Medicine, College of Veterinary Medicine, Seoul National University , Seoul, Republic of Korea
| | - C-Yoon Kim
- 2 Department of Medicine, School of Medicine, Konkuk University , Seoul, Republic of Korea
| | - Bokyeong Ryu
- 1 Department of Laboratory Animal Medicine, College of Veterinary Medicine, Seoul National University , Seoul, Republic of Korea
| | - Ukjin Kim
- 1 Department of Laboratory Animal Medicine, College of Veterinary Medicine, Seoul National University , Seoul, Republic of Korea
| | - Jin Kim
- 1 Department of Laboratory Animal Medicine, College of Veterinary Medicine, Seoul National University , Seoul, Republic of Korea
| | - Ji-Min Lee
- 1 Department of Laboratory Animal Medicine, College of Veterinary Medicine, Seoul National University , Seoul, Republic of Korea
| | - Byoung-Hee Lee
- 3 National Institute of Biological Resources , Incheon, Republic of Korea
| | - Jisook Moon
- 4 Department of Biotechnology, College of Life Science, CHA University , Seoul, Republic of Korea
| | - Cho-Rok Jung
- 5 Gene Therapy Research Unit, KRIBB , Daejeon, Republic of Korea
| | - Jae-Hak Park
- 1 Department of Laboratory Animal Medicine, College of Veterinary Medicine, Seoul National University , Seoul, Republic of Korea
| |
Collapse
|
11
|
Li J, Yao W, Zhang L, Bao L, Chen H, Wang D, Yue Z, Li Y, Zhang M, Hao C. Genome-wide DNA methylation analysis in lung fibroblasts co-cultured with silica-exposed alveolar macrophages. Respir Res 2017; 18:91. [PMID: 28499430 PMCID: PMC5429546 DOI: 10.1186/s12931-017-0576-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 05/08/2017] [Indexed: 01/14/2023] Open
Abstract
Background Exposure to crystalline silica is considered to increase the risk of lung fibrosis. The primary effector cell, the myofibroblast, plays an important role in the deposition of extracellular matrix (ECM). DNA methylation change is considered to have a potential effect on myofibroblast differentiation. Therefore, the present study was designed to investigate the genome-wide DNA methylation profiles of lung fibroblasts co-cultured with alveolar macrophages exposed to crystalline silica in vitro. Methods AM/fibroblast co-culture system was established. CCK8 was used to assess the toxicity of AMs. mRNA and protein expression of collagen I, α-SMA, MAPK9 and TGF-β1 of fibroblasts after AMs exposed to 100 μg /ml SiO2 for 0–, 24–, or 48 h were determined by means of quantitative real-time PCR, immunoblotting and immunohistochemistry. Genomic DNA of fibroblasts was isolated using MeDIP-Seq to sequence. R software, GO, KEGG and Cytoscape were used to analyze the data. Results SiO2 exposure increased the expression of collagen I and α-SMA in fibroblasts in co-culture system. Analysis of fibroblast methylome identified extensive methylation changes involved in several signaling pathways, such as the MAPK signaling pathway and metabolic pathways. Several candidates, including Tgfb1 and Mapk9, are hubs who can connect the gene clusters. MAPK9 mRNA expression was significantly higher in fibroblast exposed to SiO2 in co-culture system for 48 h. MAPK9 protein expression was increased at both 24-h and 48-h treatment groups. TGF-β1 mRNA expression of fibroblast has a time-dependent manner, but we didn’t observe the TGF-β1 protein expression. Conclusion Tgfb1 and Mapk9 are helpful to explore the mechanism of myofibroblast differentiation. The genome-wide DNA methylation profiles of fibroblasts in this experimental silicosis model will be useful for future studies on epigenetic gene regulation during myofibroblast differentiation. Electronic supplementary material The online version of this article (doi:10.1186/s12931-017-0576-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Juan Li
- College of Public Health, Zhengzhou University, No.100, Kexue Road, Zhengzhou city, Henan province, China
| | - Wu Yao
- College of Public Health, Zhengzhou University, No.100, Kexue Road, Zhengzhou city, Henan province, China
| | - Lin Zhang
- College of Public Health, Zhengzhou University, No.100, Kexue Road, Zhengzhou city, Henan province, China
| | - Lei Bao
- College of Public Health, Zhengzhou University, No.100, Kexue Road, Zhengzhou city, Henan province, China
| | - Huiting Chen
- College of Public Health, Zhengzhou University, No.100, Kexue Road, Zhengzhou city, Henan province, China
| | - Di Wang
- College of Public Health, Zhengzhou University, No.100, Kexue Road, Zhengzhou city, Henan province, China
| | - Zhongzheng Yue
- College of Public Health, Zhengzhou University, No.100, Kexue Road, Zhengzhou city, Henan province, China
| | - Yiping Li
- College of Public Health, Zhengzhou University, No.100, Kexue Road, Zhengzhou city, Henan province, China
| | - Miao Zhang
- College of Public Health, Zhengzhou University, No.100, Kexue Road, Zhengzhou city, Henan province, China
| | - Changfu Hao
- College of Public Health, Zhengzhou University, No.100, Kexue Road, Zhengzhou city, Henan province, China.
| |
Collapse
|
12
|
Herseth JI, Volden V, Bolling AK. Particulate matter-mediated release of long pentraxin 3 (PTX3) and vascular endothelial growth factor (VEGF) in vitro: Limited importance of endotoxin and organic content. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2017; 80:105-119. [PMID: 28071984 DOI: 10.1080/15287394.2016.1257399] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 11/02/2016] [Indexed: 06/06/2023]
Abstract
Exposure to particulate matter (PM) is associated with adverse health effects, but it is still relatively unknown which role PM sources and physicochemical properties play in the observed effects. It was postulated that PM in vitro induces release of long pentraxin 3 (PTX3) and vascular endothelial growth factor (VEGF) and that endotoxin and organic compounds present in the PM regulate this release. A contact coculture of THP-1 human leukemia monocytes and A549 human adenocarcinoma alveolar pneumocytes was exposed to PM from Traffic, Wood, Diesel, and Quartz (10-40 µg/cm2) for 12-64 h to determine release of PTX3 and VEGF. The role of endotoxin and the organic fraction in the mediator release was assessed using polymyxin B sulfate and organic extracts, respectively. Finally, antagonists were used to investigate whether the early proinflammatory cytokines interleukin (IL)-1 and tumor necrosis factor (TNF)-α affected the PTX3 and VEGF release. All PM samples induced a time-dependent release of both PTX3 and VEGF. Traffic mediated the greatest release of PTX3, whereas Wood and Diesel were more potent inducers of VEGF. The endotoxin content did not markedly affect release of either mediator, while the organic fraction exerted no significant effect on VEGF release and limited influence on PTX3 release. In addition, the IL-1 and TNF-α agonists affected PTX3 release more strongly than VEGF release. In conclusion, the current data show a limited impact of endotoxin and organic compounds on PTX3 and VEGF release. Further, the observed differences in response patterns may point toward differential regulation of PM-mediated release of PTX3 and VEGF.
Collapse
Affiliation(s)
- J I Herseth
- a Faculty of Health Sciences , Oslo and Akershus University College of Applied Sciences , Oslo , Norway
| | - V Volden
- a Faculty of Health Sciences , Oslo and Akershus University College of Applied Sciences , Oslo , Norway
| | - A K Bolling
- b Department of Air Pollution and Noise , Norwegian Institute of Public Health , Oslo , Norway
| |
Collapse
|
13
|
Liu T, Dai W, Li C, Liu F, Chen Y, Weng D, Chen J. Baicalin Alleviates Silica-Induced Lung Inflammation and Fibrosis by Inhibiting the Th17 Response in C57BL/6 Mice. JOURNAL OF NATURAL PRODUCTS 2015; 78:3049-3057. [PMID: 26605988 DOI: 10.1021/acs.jnatprod.5b00868] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Silicosis is an inflammatory and fibrotic lung disease caused by inhalation of silica. Th17 cells play a key role in causing silica-induced lung inflammation and fibrosis. Baicalin, a compound isolated from the Chinese herb Huangqin, could suppress the differentiation of Th17 cells and alleviate inflammation. However, there are very few reports of the immunoregulatory mechanisms of baicalin in experimental silica-induced lung inflammation and fibrosis. In our study, mice were exposed to silica by intratracheal instillation, and in this way we established an experimental silicosis model. To elucidate the effects and mechanisms of baicalin in silica-induced inflammation and fibrosis, we used baicalin to treat the developed mouse model of silicosis. Treatment with baicalin attenuated the accumulation of inflammatory cells and led to milder pathological inflammatory and fibrotic changes in lung tissues. Baicalin affected the immunological balance between Th17 and Treg responses. Therefore, baicalin caused a decrease in Th17 cells by stimulating Treg cells and by inhibiting IL-6 and IL-23. We further demonstrated that silica-induced Th1 and Th2 immune responses were both inhibited by increased Treg activation, which was promoted by baicalin. Our findings confirmed the potential functions of baicalin in inhibiting the Th17 response and reducing silica-induced inflammation and fibrosis.
Collapse
Affiliation(s)
- Tao Liu
- Department of Occupational and Environmental Health, School of Public Health, and ‡Department of Natural Products Chemistry, School of Pharmacy, China Medical University , Shenyang 110122, People's Republic of China
| | - Wujing Dai
- Department of Occupational and Environmental Health, School of Public Health, and ‡Department of Natural Products Chemistry, School of Pharmacy, China Medical University , Shenyang 110122, People's Republic of China
| | - Chao Li
- Department of Occupational and Environmental Health, School of Public Health, and ‡Department of Natural Products Chemistry, School of Pharmacy, China Medical University , Shenyang 110122, People's Republic of China
| | - Fangwei Liu
- Department of Occupational and Environmental Health, School of Public Health, and ‡Department of Natural Products Chemistry, School of Pharmacy, China Medical University , Shenyang 110122, People's Republic of China
| | - Ying Chen
- Department of Occupational and Environmental Health, School of Public Health, and ‡Department of Natural Products Chemistry, School of Pharmacy, China Medical University , Shenyang 110122, People's Republic of China
| | - Dong Weng
- Department of Occupational and Environmental Health, School of Public Health, and ‡Department of Natural Products Chemistry, School of Pharmacy, China Medical University , Shenyang 110122, People's Republic of China
| | - Jie Chen
- Department of Occupational and Environmental Health, School of Public Health, and ‡Department of Natural Products Chemistry, School of Pharmacy, China Medical University , Shenyang 110122, People's Republic of China
| |
Collapse
|
14
|
Immune Homeostasis in Epithelial Cells: Evidence and Role of Inflammasome Signaling Reviewed. J Immunol Res 2015; 2015:828264. [PMID: 26355424 PMCID: PMC4556877 DOI: 10.1155/2015/828264] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 07/07/2015] [Indexed: 12/12/2022] Open
Abstract
The epithelium regulates the interaction between the noxious xenogenous, as well as the microbial environment and the immune system, not only by providing a barrier but also by expressing a number of immunoregulatory membrane receptors, and intracellular danger sensors and their downstream effectors. Amongst these are a number of inflammasome sensor subtypes, which have been initially characterized in myeloid cells and described to be activated upon assembly into multiprotein complexes by microbial and environmental triggers. This review compiles a vast amount of literature that supports a pivotal role for inflammasomes in the various epithelial barriers of the human body as essential factors maintaining immune signaling and homeostasis.
Collapse
|
15
|
Willoughby JA. Predicting Respiratory Toxicity Using a Human 3D Airway (EpiAirway™) Model Combined with Multiple Parametric Analysis. ACTA ACUST UNITED AC 2015. [DOI: 10.1089/aivt.2014.0003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
16
|
Perkins TN, Peeters PM, Shukla A, Arijs I, Dragon J, Wouters EFM, Reynaert NL, Mossman BT. Indications for distinct pathogenic mechanisms of asbestos and silica through gene expression profiling of the response of lung epithelial cells. Hum Mol Genet 2014; 24:1374-89. [PMID: 25351596 DOI: 10.1093/hmg/ddu551] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Occupational and environmental exposures to airborne asbestos and silica are associated with the development of lung fibrosis in the forms of asbestosis and silicosis, respectively. However, both diseases display distinct pathologic presentations, likely associated with differences in gene expression induced by different mineral structures, composition and bio-persistent properties. We hypothesized that effects of mineral exposure in the airway epithelium may dictate deviating molecular events that may explain the different pathologies of asbestosis versus silicosis. Using robust gene expression-profiling in conjunction with in-depth pathway analysis, we assessed early (24 h) alterations in gene expression associated with crocidolite asbestos or cristobalite silica exposures in primary human bronchial epithelial cells (NHBEs). Observations were confirmed in an immortalized line (BEAS-2B) by QRT-PCR and protein assays. Utilization of overall gene expression, unsupervised hierarchical cluster analysis and integrated pathway analysis revealed gene alterations that were common to both minerals or unique to either mineral. Our findings reveal that both minerals had potent effects on genes governing cell adhesion/migration, inflammation, and cellular stress, key features of fibrosis. Asbestos exposure was most specifically associated with aberrant cell proliferation and carcinogenesis, whereas silica exposure was highly associated with additional inflammatory responses, as well as pattern recognition, and fibrogenesis. These findings illustrate the use of gene-profiling as a means to determine early molecular events that may dictate pathological processes induced by exogenous cellular insults. In addition, it is a useful approach for predicting the pathogenicity of potentially harmful materials.
Collapse
Affiliation(s)
- Timothy N Perkins
- Department of Pathology, University of Vermont College of Medicine, Burlington, VT, USA, Department of Respiratory Medicine, Maastricht University Medical Centre, Maastricht University, Maastricht, The Netherlands,
| | - Paul M Peeters
- Department of Pathology, University of Vermont College of Medicine, Burlington, VT, USA, Department of Respiratory Medicine, Maastricht University Medical Centre, Maastricht University, Maastricht, The Netherlands,
| | - Arti Shukla
- Department of Pathology, University of Vermont College of Medicine, Burlington, VT, USA
| | - Ingrid Arijs
- Department of Gastroenterology, Translational Research Center for Gastrointestinal Disorders (TARGID), and Gene Expression Unit, Department of Molecular Cell Biology, KU Leuven, Leuven, Belgium
| | - Julie Dragon
- Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, VT, USA
| | - Emiel F M Wouters
- Department of Respiratory Medicine, Maastricht University Medical Centre, Maastricht University, Maastricht, The Netherlands
| | - Niki L Reynaert
- Department of Respiratory Medicine, Maastricht University Medical Centre, Maastricht University, Maastricht, The Netherlands
| | - Brooke T Mossman
- Department of Pathology, University of Vermont College of Medicine, Burlington, VT, USA
| |
Collapse
|
17
|
Song L, Weng D, Dai W, Tang W, Chen S, Li C, Chen Y, Liu F, Chen J. Th17 can regulate silica-induced lung inflammation through an IL-1β-dependent mechanism. J Cell Mol Med 2014; 18:1773-84. [PMID: 25091058 PMCID: PMC4196653 DOI: 10.1111/jcmm.12341] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 05/14/2014] [Indexed: 12/16/2022] Open
Abstract
Silicosis is an occupational lung disease caused by the inhalation of silica dust and characterized by lung inflammation and fibrosis. Interleukin (IL)-1β is induced by silica and functions as the key pro-inflammatory cytokine in this process. The Th17 response, which is induced by IL-1β, has been reported very important in chronic human lung inflammatory diseases. To elucidate the underlying mechanisms of IL-1β and IL-17 in silicosis, we used anakinra and an anti-IL-17 monoclonal antibody (mAb) to block the receptor of IL-1β (IL-RI) and IL-17, respectively, in a mouse model of silicosis. We observed increased IL-1β expression and an enhanced Th17 response after silica instillation. Treatment with an IL-1 type I receptor (IL-1RI) antagonist anakinra substantially decreased silica-induced lung inflammation and the Th17 response. Lung inflammation and the accumulation of inflammatory cells were attenuated in the IL-17-neutralized silicosis group. IL-17 may promote lung inflammation by modulating the differentiation of Th1 and regulatory T cells (Tregs) and by regulating the production of IL-22 and IL-1β during the lung inflammation of silicosis. Silica may induce IL-1β production from alveolar macrophages and promote inflammation by initiating a Th17 response via an IL-1β/IL-1RI-dependent mechanism. The Th17 response could induce lung inflammation during the pathogenesis of silicosis by regulating the homoeostasis of the Th immune responses and affecting the production of IL-22 and IL-1β. This study describes a potentially important inflammatory mechanism of silicosis that may bring about novel therapies for this inflammatory and fibrotic disease.
Collapse
Affiliation(s)
- Laiyu Song
- Division of Pneumoconiosis, School of Public Health, China Medical UniversityShenyang, China
- Department of Clinical Immunology, Dalian Medical UniversityDalian, China
| | - Dong Weng
- Department of Respiratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of MedicineShanghai, China
| | - Wujing Dai
- Division of Pneumoconiosis, School of Public Health, China Medical UniversityShenyang, China
| | - Wen Tang
- Division of Pneumoconiosis, School of Public Health, China Medical UniversityShenyang, China
| | - Shi Chen
- Division of Pneumoconiosis, School of Public Health, China Medical UniversityShenyang, China
| | - Chao Li
- Division of Pneumoconiosis, School of Public Health, China Medical UniversityShenyang, China
| | - Ying Chen
- Division of Pneumoconiosis, School of Public Health, China Medical UniversityShenyang, China
| | - Fangwei Liu
- Division of Pneumoconiosis, School of Public Health, China Medical UniversityShenyang, China
| | - Jie Chen
- Division of Pneumoconiosis, School of Public Health, China Medical UniversityShenyang, China
| |
Collapse
|
18
|
Mossman BT, Glenn RE. Bioreactivity of the crystalline silica polymorphs, quartz and cristobalite, and implications for occupational exposure limits (OELs). Crit Rev Toxicol 2013; 43:632-60. [PMID: 23863112 DOI: 10.3109/10408444.2013.818617] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Silica or silicon dioxides (SiO₂) are naturally occurring substances that comprise the vast majority of the earth's crust. Because of their prevalence and commercial applications, they have been widely studied for their potential to induce pulmonary fibrosis and other disorders. Historically, the focus in the workplace has been on the development of inflammation and fibrotic lung disease, the basis for promulgating workplace standards to protect workers. Crystalline silica (CS) polymorphs, predominantly quartz and cristobalite, are used in industry but are different in their mineralogy, chemistry, surface features, size dimensions and association with other elements naturally and during industrial applications. Epidemiologic, clinical and experimental studies in the literature historically have predominantly focused on quartz polymorphs. Thus, in this review, we summarize past scientific evaluations and recent peer-reviewed literature with an emphasis on cristobalite, in an attempt to determine whether quartz and cristobalite polymorphs differ in their health effects, toxicity and other properties that may dictate the need for various standards of protection in the workplace. In addition to current epidemiological and clinical reports, we review in vivo studies in rodents as well as cell culture studies that shed light on mechanisms intrinsic to the toxicity, altered cell responses and protective or defense mechanisms in response to these minerals. The medical and scientific literature indicates that the mechanisms of injury and potential causation of inflammation and fibrotic lung disease are similar for quartz and cristobalite. Our analysis of these data suggests similar occupational exposure limits (OELs) for these minerals in the workplace.
Collapse
Affiliation(s)
- Brooke T Mossman
- Department of Pathology, University of Vermont College of Medicine, Burlington, VT 05405-0068, USA.
| | | |
Collapse
|
19
|
Peeters PM, Perkins TN, Wouters EFM, Mossman BT, Reynaert NL. Silica induces NLRP3 inflammasome activation in human lung epithelial cells. Part Fibre Toxicol 2013; 10:3. [PMID: 23402370 PMCID: PMC3607900 DOI: 10.1186/1743-8977-10-3] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Accepted: 01/29/2013] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND In myeloid cells the inflammasome plays a crucial role in innate immune defenses against pathogen- and danger-associated patterns such as crystalline silica. Respirable mineral particles impinge upon the lung epithelium causing irreversible damage, sustained inflammation and silicosis. In this study we investigated lung epithelial cells as a target for silica-induced inflammasome activation. METHODS A human bronchial epithelial cell line (BEAS-2B) and primary normal human bronchial epithelial cells (NHBE) were exposed to toxic but nonlethal doses of crystalline silica over time to perform functional characterization of NLRP3, caspase-1, IL-1β, bFGF and HMGB1. Quantitative RT-PCR, caspase-1 enzyme activity assay, Western blot techniques, cytokine-specific ELISA and fibroblast (MRC-5 cells) proliferation assays were performed. RESULTS We were able to show transcriptional and translational upregulation of the components of the NLRP3 intracellular platform, as well as activation of caspase-1. NLRP3 activation led to maturation of pro-IL-1β to secreted IL-1β, and a significant increase in the unconventional release of the alarmins bFGF and HMGB1. Moreover, release of bFGF and HMGB1 was shown to be dependent on particle uptake. Small interfering RNA experiments using siNLRP3 revealed the pivotal role of the inflammasome in diminished release of pro-inflammatory cytokines, danger molecules and growth factors, and fibroblast proliferation. CONCLUSION Our novel data indicate the presence and functional activation of the NLRP3 inflammasome by crystalline silica in human lung epithelial cells, which prolongs an inflammatory signal and affects fibroblast proliferation, mediating a cadre of lung diseases.
Collapse
Affiliation(s)
- Paul M Peeters
- Department of Respiratory Medicine, Maastricht University Medical Centre+ (MUMC+), Maastricht University, Maastricht, The Netherlands
| | | | | | | | | |
Collapse
|
20
|
Ramos-Godínez MDP, González-Gómez BE, Montiel-Dávalos A, López-Marure R, Alfaro-Moreno E. TiO2 nanoparticles induce endothelial cell activation in a pneumocyte-endothelial co-culture model. Toxicol In Vitro 2012; 27:774-81. [PMID: 23261642 DOI: 10.1016/j.tiv.2012.12.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 10/24/2012] [Accepted: 12/12/2012] [Indexed: 01/08/2023]
Abstract
The effects of particulate matter (PM) on endothelial cells have been evaluated in vitro by exposing isolated endothelial cells to different types of PM. Although some of the findings from these experiments have been corroborated by in vivo studies, an in vitro model that assesses the interaction among different cell types is necessary to achieve more realistic assays. We developed an in vitro model that mimics the alveolar-capillary interface, and we challenged the model using TiO nanoparticles (TiO-NPs). Human umbilical endothelial cells (HUVECs) were cultured on the basolateral side of a membrane and pneumocytes (A549) on the apical side. Confluent co-cultures were exposed on the apical side to 10 μg/cm of TiO-NPs or 10 ng/mL of TNFα for 24 h. Unexposed cultures were used as negative controls. We evaluated monocyte adhesion to HUVECs, adhesion molecule expression, nitric oxide concentration and proinflammatory cytokine release. The TiO-NPs added to the pneumocytes induced a 3- to 4-fold increase in monocyte adhesion to the HUVECs and significant increases in the expression of adhesion molecules (4-fold for P-selectin at 8 h, and about 8- and 10-fold for E-selectin, ICAM-1, VCAM-1 and PECAM-1 at 24 h). Nitric oxide production also increased significantly (2-fold). These results indicate that exposing pneumocytes to TiO-NPs causes endothelial cell activation.
Collapse
Affiliation(s)
- María Del Pilar Ramos-Godínez
- Environmental Toxicology Laboratory, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Mexico
| | | | | | | | | |
Collapse
|
21
|
The α(1)AT and TIMP-1 Gene Polymorphism in the Development of Asthma. Comp Funct Genomics 2012; 2012:968267. [PMID: 23226977 PMCID: PMC3512250 DOI: 10.1155/2012/968267] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Accepted: 10/13/2012] [Indexed: 01/23/2023] Open
Abstract
Asthma has been an inflammatory disorder accompanied by tissue remodeling and protease-antiprotease imbalance in lungs. The SNPs of alpha-1 antitrypsin (α(1)AT) and tissue inhibitor of metalloproteinase-1 (TIMP-1) genes were studied for their association with asthma. Genotyping of α(1)AT and TIMP-1 genes was performed in 202 asthmatics and 204 controls. Serum levels of α(1)AT, TIMP-1 and cytokines were estimated to find if the interplay between genotypes and cellular biomarkers determines the pathogenesis of asthma. The analysis of results showed significantly low level of α(1)AT in the serum of asthmatics as compared to controls (P = 0.001), whereas cytokines were elevated in patients. No significant difference was observed in the concentration of TIMP-1 in patients and controls. Genotyping led to the identification of 3 SNPs (Val213Ala, Glu363Lys, and Glu376Asp) in α(1)AT gene. The novel SNP Glu363Lys of α(1)AT was found to be associated with asthma (P = 0.001). The analysis of TIMP-1 gene showed the occurrence of seven SNPs, including a novel intronic SNP at base G3774A. The allele frequency of G3774A and Phe124Phe was significantly higher in asthmatics as compared to controls. Thus, the SNP Glu363Lys of α(1)AT and G3774A and Phe124Phe of TIMP-1 could be important genetic markers for use in better management of the disease.
Collapse
|
22
|
Guo L, Zhu N, Guo Z, Li GK, Chen C, Sang N, Yao QC. Particulate matter (PM10) exposure induces endothelial dysfunction and inflammation in rat brain. JOURNAL OF HAZARDOUS MATERIALS 2012; 213-214:28-37. [PMID: 22365138 DOI: 10.1016/j.jhazmat.2012.01.034] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Revised: 12/05/2011] [Accepted: 01/11/2012] [Indexed: 05/04/2023]
Abstract
Epidemiological studies suggest that particulate matter (PM(10)) inhalation was associated with adverse effects on brain-related health, however, existing experimental data lacked relevant evidences. In this study, we treated Wistar rats with PM(10) at different concentrations (0.3, 1, 3 and 10 mg/kg body weight (bw)), and investigated endothelial dysfunction and inflammatory responses in the brain. The results indicate that mild pathological abnormal occurred after 15-day exposure (five times with 3 days each), followed by the changes of endothelial mediators (ET-1 and eNOS) and inflammatory markers (IL-1β, TNF-α, COX-2, iNOS and ICAM-1). Also, the sample up-regulated bax/bcl-2 ratio and p53 expression, and induced neuronal apoptosis. It implicates that PM(10) exerted injuries to mammals' brain, and the mechanisms might be involved in endothelial dysfunction and inflammatory responses.
Collapse
Affiliation(s)
- Lin Guo
- College of Environment and Resource, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | | | | | | | | | | | | |
Collapse
|
23
|
Perkins TN, Shukla A, Peeters PM, Steinbacher JL, Landry CC, Lathrop SA, Steele C, Reynaert NL, Wouters EFM, Mossman BT. Differences in gene expression and cytokine production by crystalline vs. amorphous silica in human lung epithelial cells. Part Fibre Toxicol 2012; 9:6. [PMID: 22300531 PMCID: PMC3337246 DOI: 10.1186/1743-8977-9-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Accepted: 02/02/2012] [Indexed: 12/21/2022] Open
Abstract
Background Exposure to respirable crystalline silica particles, as opposed to amorphous silica, is associated with lung inflammation, pulmonary fibrosis (silicosis), and potentially with lung cancer. We used Affymetrix/GeneSifter microarray analysis to determine whether gene expression profiles differed in a human bronchial epithelial cell line (BEAS 2B) exposed to cristobalite vs. amorphous silica particles at non-toxic and equal surface areas (75 and 150 × 106μm2/cm2). Bio-Plex analysis was also used to determine profiles of secreted cytokines and chemokines in response to both particles. Finally, primary human bronchial epithelial cells (NHBE) were used to comparatively assess silica particle-induced alterations in gene expression. Results Microarray analysis at 24 hours in BEAS 2B revealed 333 and 631 significant alterations in gene expression induced by cristobalite at low (75) and high (150 × 106μm2/cm2) amounts, respectively (p < 0.05/cut off ≥ 2.0-fold change). Exposure to amorphous silica micro-particles at high amounts (150 × 106μm2/cm2) induced 108 significant gene changes. Bio-Plex analysis of 27 human cytokines and chemokines revealed 9 secreted mediators (p < 0.05) induced by crystalline silica, but none were induced by amorphous silica. QRT-PCR revealed that cristobalite selectively up-regulated stress-related genes and cytokines (FOS, ATF3, IL6 and IL8) early and over time (2, 4, 8, and 24 h). Patterns of gene expression in NHBE cells were similar overall to BEAS 2B cells. At 75 × 106μm2/cm2, there were 339 significant alterations in gene expression induced by cristobalite and 42 by amorphous silica. Comparison of genes in response to cristobalite (75 × 106μm2/cm2) revealed 60 common, significant gene alterations in NHBE and BEAS 2B cells. Conclusions Cristobalite silica, as compared to synthetic amorphous silica particles at equal surface area concentrations, had comparable effects on the viability of human bronchial epithelial cells. However, effects on gene expression, as well as secretion of cytokines and chemokines, drastically differed, as the crystalline silica induced more intense responses. Our studies indicate that toxicological testing of particulates by surveying viability and/or metabolic activity is insufficient to predict their pathogenicity. Moreover, they show that acute responses of the lung epithelium, including up-regulation of genes linked to inflammation, oxidative stress, and proliferation, as well as secretion of inflammatory and proliferative mediators, can be indicative of pathologic potential using either immortalized lines (BEAS 2B) or primary cells (NHBE). Assessment of the degree and magnitude of these responses in vitro are suggested as predictive in determining the pathogenicity of potentially harmful particulates.
Collapse
Affiliation(s)
- Timothy N Perkins
- Department of Pathology, University of Vermont College of Medicine, 89 Beaumont Avenue, Burlington, VT 05405, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Klein SG, Hennen J, Serchi T, Blömeke B, Gutleb AC. Potential of coculture in vitro models to study inflammatory and sensitizing effects of particles on the lung. Toxicol In Vitro 2011; 25:1516-34. [PMID: 21963807 DOI: 10.1016/j.tiv.2011.09.006] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2011] [Revised: 07/18/2011] [Accepted: 09/06/2011] [Indexed: 12/30/2022]
Abstract
Exposure to particulate matter (PM) like nanoparticles (NPs) has increased in the last century due to increased combustion processes, road traffic, etc. In addition, the progress in chemical and cosmetic industry led to many new compounds, e.g. fragrances, which humans are exposed to every day. Many chemicals are known to act as contact and some as respiratory sensitizers, causing allergic reactions. Exposure to small particles of less than 100 nm in diameter is linked with an increased risk of respiratory diseases, such as asthma or rhinitis. To date already more than 1000 customer products contain eNPs without knowing much about the health effects. In comparison to chemicals, the mechanisms by which PM and eNPs can cause sensitization are still not fully understood. Validated and regulatory accepted in vitro models to assess this hazard in its full range are still missing. While a huge number of animal studies contributed to our knowledge about sensitization processes, knowledge on involved cellular mechanisms is still limited. In this review relevant in vitro models to study and elucidate these mechanisms in more detail are presented and their potential to serve as part of a tiered testing strategy is discussed.
Collapse
Affiliation(s)
- Sebastian G Klein
- Department Environment and Agro-biotechnologies (EVA), Centre de Recherche Public, Gabriel Lippmann, 41 rue du Brill, L-4422 Belvaux, Luxembourg
| | | | | | | | | |
Collapse
|
25
|
Li H, Han M, Guo L, Li G, Sang N. Oxidative stress, endothelial dysfunction and inflammatory response in rat heart to NO₂ inhalation exposure. CHEMOSPHERE 2011; 82:1589-1596. [PMID: 21168897 DOI: 10.1016/j.chemosphere.2010.11.055] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2010] [Revised: 11/06/2010] [Accepted: 11/21/2010] [Indexed: 05/30/2023]
Abstract
Epidemiological studies suggest that NO₂ inhalation is associated with adverse effects on heart-related health, however, existing experimental data lack relevant evidences. In this study, a role for oxidative stress, endothelial dysfunction and inflammatory responses in the heart of rats treated with different concentrations of NO₂ (0, 5, 10 and 20 mg m⁻³) was investigated. Mild heart pathology occurred after 7-d exposure (6 h d⁻¹). Marked oxidative stress were induced as evaluated by reduction/induction of antioxidants (Cu/Zn-SOD, Mn-SOD and GPx) activity and increasing formation of MDA and PCO. Also, mRNA and protein biomarkers of vasoconstriction (ET-1, eNOS) and inflammation (TNF-α, IL-1β and ICAM-1) were up-regulated, and p53 mRNA expression, bax/bcl-2 ratio and the mean number of TUNEL-positive myocytes were increased as well. All the results implicate that NO₂ exerted injuries to mammals' heart, and the damage mechanisms were possibly associated with oxidative stress, endothelial dysfunction and inflammation.
Collapse
Affiliation(s)
- Hongyan Li
- College of Environment and Resource, Center of Environmental Science and Engineering, Shanxi University, Taiyuan, Shanxi, PR China
| | | | | | | | | |
Collapse
|
26
|
Yun Y, Hou L, Sang N. SO(2) inhalation modulates the expression of pro-inflammatory and pro-apoptotic genes in rat heart and lung. JOURNAL OF HAZARDOUS MATERIALS 2011; 185:482-488. [PMID: 20951496 DOI: 10.1016/j.jhazmat.2010.09.057] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2010] [Revised: 09/16/2010] [Accepted: 09/16/2010] [Indexed: 05/30/2023]
Abstract
SO(2) is a common air pollutant, and human exposure to SO(2) has become increasingly widespread due to the combustion of fossil fuels. The epidemiological studies have linked SO(2) exposure not only with many respiratory responses, but also with cardiovascular diseases. Also, its possible toxicity has been implicated by determining oxidative stress, DNA damage and membrane channel alteration in rat heart and lung. However, its detailed mechanisms remain unclear. In the present study, rats were treated with 7, 14 and 28 mg/m(3) SO(2) for 6h/day for 7 days, and the mRNA levels of TNF-α, IL-1β, iNOS, ICAM-1, Bax and Bcl-2 and subsequent insults were determined in the heart and lung. The results indicate that SO(2) inhalation markedly elevated TNF-α and IL-1β mRNA levels and secretions, enhanced iNOS and ICAM-1 mRNA levels and the ratio of Bax/Bcl-2 in a concentration-dependent manner, and induced occurrence of apoptosis. This suggests that SO(2) inhalation induced an inflammatory response and subsequent insults via modulating pro-inflammatory and pro-apoptotic genes in the heart and lung, which contributed to the increased risk of respiratory and cardiovascular diseases.
Collapse
Affiliation(s)
- Yang Yun
- College of Environment and Resource, Center of Environmental Science and Engineering, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | | | | |
Collapse
|
27
|
Napierska D, Thomassen LCJ, Lison D, Martens JA, Hoet PH. The nanosilica hazard: another variable entity. Part Fibre Toxicol 2010; 7:39. [PMID: 21126379 PMCID: PMC3014868 DOI: 10.1186/1743-8977-7-39] [Citation(s) in RCA: 477] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2010] [Accepted: 12/03/2010] [Indexed: 11/10/2022] Open
Abstract
Silica nanoparticles (SNPs) are produced on an industrial scale and are an addition to a growing number of commercial products. SNPs also have great potential for a variety of diagnostic and therapeutic applications in medicine. Contrary to the well-studied crystalline micron-sized silica, relatively little information exists on the toxicity of its amorphous and nano-size forms. Because nanoparticles possess novel properties, kinetics and unusual bioactivity, their potential biological effects may differ greatly from those of micron-size bulk materials. In this review, we summarize the physico-chemical properties of the different nano-sized silica materials that can affect their interaction with biological systems, with a specific emphasis on inhalation exposure. We discuss recent in vitro and in vivo investigations into the toxicity of nanosilica, both crystalline and amorphous. Most of the in vitro studies of SNPs report results of cellular uptake, size- and dose-dependent cytotoxicity, increased reactive oxygen species levels and pro-inflammatory stimulation. Evidence from a limited number of in vivo studies demonstrates largely reversible lung inflammation, granuloma formation and focal emphysema, with no progressive lung fibrosis. Clearly, more research with standardized materials is needed to enable comparison of experimental data for the different forms of nanosilicas and to establish which physico-chemical properties are responsible for the observed toxicity of SNPs.
Collapse
Affiliation(s)
- Dorota Napierska
- Unit of Lung Toxicology, Katholieke Universiteit Leuven, Herestraat 49, 3000 Leuven, Belgium
| | | | | | | | | |
Collapse
|
28
|
Ramery E, Fievez L, Fraipont A, Bureau F, Lekeux P. Characterization of pentraxin 3 in the horse and its expression in airways. Vet Res 2009; 41:18. [PMID: 19863902 PMCID: PMC2789332 DOI: 10.1051/vetres/2009066] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2009] [Accepted: 10/28/2009] [Indexed: 01/05/2023] Open
Abstract
The long pentraxin 3 (PTX3) plays an important role in host defence and its over-expression may contribute to airway injury. The aim of the present study was therefore to characterize in more detail PTX3 and its expression in the horses’ airway. Six healthy horses and six horses affected by recurrent airway obstruction (R.A.O.) were submitted to a dusty environment challenge. PTX3 DNA and cDNA were cloned and sequenced. PTX3 expression was evaluated by RT-qPCR, Western blotting and immuno-histochemistry in bronchoalveolar lavage fluid (BALF) cells, BALF supernatant and bronchial epithelial cells. An alternative splicing of the second exon of PTX3 occurred, resulting in two forms of the protein: “spliced” (32 kDa) and “full length” (42 kDa). PTX3 was detected in BALF macrophages, neutrophils and bronchial epithelial cells. It was over-expressed in the BALF supernatant from R.A.O.-affected horses in crisis. However, dust was unable to induce PTX3 in BALF cells ex vivo, indicating that dust is an indirect inducer of PTX3. Dust exposure in-vivo induced PTX3 in BALF macrophages but there was no significant difference between healthy and R.A.O.-affected horses. Conversely, PTX3 was over-expressed in the bronchial epithelial cells from R.A.O.-affected horses in crisis. These data indicate a differential regulatory mechanism in inflammatory and bronchial epithelial cells and offer therapeutically interesting perspectives.
Collapse
Affiliation(s)
- Eve Ramery
- Department for Functional Sciences, Faculty of Veterinary Medicine, University of Liege, Bvd de Colonster, 20, B-4000 Liege, Belgium.
| | | | | | | | | |
Collapse
|
29
|
Park MVDZ, Lankveld DPK, van Loveren H, de Jong WH. The status of in vitro toxicity studies in the risk assessment of nanomaterials. Nanomedicine (Lond) 2009; 4:669-85. [DOI: 10.2217/nnm.09.40] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Nanotechnology applications already on the market or in development promise great benefits for humans as well as the environment. Simultaneously, the pressure to advance the development of fast methods for evaluating the potential risks of increased human exposure to nanomaterials is augmented. One way forward would be to enhance the role of in vitro toxicity studies in risk assessment procedures of nanomaterials. However, to maximize the use of in vitro assays for this purpose, their values and limitations need to be revealed. Even in risk assessment frameworks for regular chemicals, in vitro studies play a minor role. A comparative analysis of published in vitro data with nanomaterials demonstrates that there are a number of issues that need resolving before in vitro studies can play a role in the risk assessment of nanomaterials.
Collapse
Affiliation(s)
- Margriet VDZ Park
- Laboratory for Health Protection Research, National Institute for Public Health & the Environment (RIVM), Bilthoven, The Netherlands
- Department of Health Risk Analysis & Toxicology, Maastricht University, Maastricht, The Netherlands
| | - Daniëlle PK Lankveld
- Laboratory for Health Protection Research, National Institute for Public Health & the Environment (RIVM), Bilthoven, The Netherlands
| | - Henk van Loveren
- Laboratory for Health Protection Research, National Institute for Public Health & the Environment (RIVM), Bilthoven, The Netherlands
- Department of Health Risk Analysis & Toxicology, Maastricht University, Maastricht, The Netherlands
| | - Wim H de Jong
- Laboratory for Health Protection Research, National Institute for Public Health & the Environment (RIVM), Bilthoven, The Netherlands
| |
Collapse
|