1
|
Ma Y, Hossen MM, Huang JJ, Yin Z, Du J, Ye Z, Zeng M, Huang Z. Growth arrest and DNA damage-inducible 45: a new player on inflammatory diseases. Front Immunol 2025; 16:1513069. [PMID: 40083548 PMCID: PMC11903704 DOI: 10.3389/fimmu.2025.1513069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 02/07/2025] [Indexed: 03/16/2025] Open
Abstract
Growth arrest and DNA damage-inducible 45 (GADD45) proteins are critical stress sensors rapidly induced in response to genotoxic/physiological stress and regulate many cellular functions. Even though the primary function of the proteins is to block the cell cycle, inhibit cell proliferation, promote cell apoptosis, and repair DNA damage to cope with the damage caused by internal and external stress on the body, evidence has shown that GADD45 also has the function to modulate innate and adaptive immunity and plays a broader role in inflammatory and autoimmune diseases. In this review, we focus on the immunomodulatory role of GADD45 in inflammatory and autoimmune diseases. First, we describe the regulatory factors that affect the expression of GADD45. Then, we introduce its immunoregulatory roles on immune cells and the critical signaling pathways mediated by GADD45. Finally, we discuss its immunomodulatory effects in various inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- Yanmei Ma
- Rheumatology Research Institute, Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
- Department of Immunology, Biological Therapy Institute, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Health Science Center, Shenzhen University, Shenzhen, China
- Joint Research Laboratory for Rheumatology of Shenzhen University Health Science Center and Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
| | - Md Munnaf Hossen
- Rheumatology Research Institute, Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
- Department of Immunology, Biological Therapy Institute, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Health Science Center, Shenzhen University, Shenzhen, China
- Joint Research Laboratory for Rheumatology of Shenzhen University Health Science Center and Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
| | - Jennifer Jin Huang
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, United States
| | - Zhihua Yin
- Rheumatology Research Institute, Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
- Joint Research Laboratory for Rheumatology of Shenzhen University Health Science Center and Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
| | - Jing Du
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - Zhizhong Ye
- Rheumatology Research Institute, Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
- Joint Research Laboratory for Rheumatology of Shenzhen University Health Science Center and Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
| | - Miaoyu Zeng
- Rheumatology Research Institute, Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
- Joint Research Laboratory for Rheumatology of Shenzhen University Health Science Center and Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
| | - Zhong Huang
- Department of Immunology, Biological Therapy Institute, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Health Science Center, Shenzhen University, Shenzhen, China
- Joint Research Laboratory for Rheumatology of Shenzhen University Health Science Center and Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
| |
Collapse
|
2
|
Yuan J, Mo Y, Zhang Y, Zhang Y, Zhang Q. HMGB1 derived from lung epithelial cells after cobalt nanoparticle exposure promotes the activation of lung fibroblasts. Nanotoxicology 2024; 18:565-581. [PMID: 39295432 PMCID: PMC11581909 DOI: 10.1080/17435390.2024.2404074] [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: 05/10/2024] [Revised: 08/11/2024] [Accepted: 09/07/2024] [Indexed: 09/21/2024]
Abstract
We have previously demonstrated that exposure to cobalt nanoparticles (Nano-Co) caused extensive interstitial fibrosis and inflammatory cell infiltration in mouse lungs. However, the underlying mechanisms of Nano-Co-induced pulmonary fibrosis remain unclear. In this study, we investigated the role of high-mobility group box 1 (HMGB1) in the epithelial cell-fibroblast crosstalk in Nano-Co-induced pulmonary fibrosis. Our results showed that Nano-Co exposure caused remarkable production and release of HMGB1, as well as nuclear accumulation of HIF-1α in human bronchial epithelial cells (BEAS-2B) in a dose- and a time-dependent manner. Pretreatment with CAY10585, an inhibitor against HIF-1α, significantly blocked the overexpression of HMGB1 in cell lysate and the release of HMGB1 in the supernatant of BEAS-2B cells induced by Nano-Co exposure, indicating that Nano-Co exposure induces HIF-1α-dependent HMGB1 overexpression and release. In addition, treatment of lung fibroblasts (MRC-5) with conditioned media from Nano-Co-exposed BEAS-2B cells caused increased RAGE expression, MAPK signaling activation, and enhanced expression of fibrosis-associated proteins, such as fibronectin, collagen 1, and α-SMA. However, conditioned media from Nano-Co-exposed BEAS-2B cells with HMGB1 knockdown had no effects on the activation of MRC-5 fibroblasts. Finally, inhibition of ERK1/2, p38, and JNK all abolished MRC-5 activation induced by conditioned media from Nano-Co-exposed BEAS-2B cells, suggesting that MAPK signaling might be a key downstream signal of HMGB1/RAGE to promote MRC-5 fibroblast activation. These findings have important implications for understanding the pro-fibrotic potential of Nano-Co.
Collapse
Affiliation(s)
- Jiali Yuan
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA
| | - Yiqun Mo
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA
| | - Yue Zhang
- Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yuanbao Zhang
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA
| | - Qunwei Zhang
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA
| |
Collapse
|
3
|
Kanika, Khan R. Functionalized nanomaterials targeting NLRP3 inflammasome driven immunomodulation: Friend or Foe. NANOSCALE 2023; 15:15906-15928. [PMID: 37750698 DOI: 10.1039/d3nr03857b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/27/2023]
Abstract
The advancement in drug delivery systems in recent times has significantly enhanced therapeutic effects by enabling site-specific targeting through nanocarriers. These nanocarriers serve as invaluable tools for pharmacotherapeutic advancements against various disorders that enhance the effectiveness of encapsulated drugs by reducing their toxicity and increasing the efficacy of less potent drugs, thereby improving the therapeutic index. Inflammasomes, protein complexes located in the activated immune cell cytoplasm, regulate the activation of caspases involved in inflammation. However, aberrant activation of inflammasomes can result in uncontrolled tissue responses, contributing to the development of various diseases. Therefore, achieving a precise balance between inflammasome inhibition and activation is crucial for effectively treating inflammatory disorders through targeted functionalized nanocarriers. Despite the wealth of available data on the relevance of functionalized nanocarriers in inflammatory disorders, the nanotechnological potential to modulate inflammasomes has not been adequately explored. In this comprehensive review, we highlight the latest research on the modulation of the inflammasome cascade, both upregulating and downregulating its function, using nanocarriers in the context of inflammatory disorders. The utilization of nanocarriers as a therapeutic strategy holds immense potential for researchers aiming to effectively target and modulate inflammasomes in the treatment of inflammatory disorders, thus improving disease severity outcomes.
Collapse
Affiliation(s)
- Kanika
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, 5 Sahibzada Ajit Singh Nagar, Punjab, Pin - 140306, India.
| | - Rehan Khan
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, 5 Sahibzada Ajit Singh Nagar, Punjab, Pin - 140306, India.
| |
Collapse
|
4
|
Yuan J, Mo Y, Zhang Y, Zhang Y, Zhang Q. Nickel nanoparticles induce autophagy and apoptosis via HIF-1α/mTOR signaling in human bronchial epithelial cells. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 329:121670. [PMID: 37080518 PMCID: PMC10231338 DOI: 10.1016/j.envpol.2023.121670] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/31/2023] [Accepted: 04/17/2023] [Indexed: 05/03/2023]
Abstract
With the rapid development of nanotechnology, the potential adverse health effects of nanoparticles have been caught more attention and become global concerns. However, the underlying mechanisms in metal nanoparticle-induced toxic effects are still largely obscure. In this study, we investigated whether exposure to nickel nanoparticles (Nano-Ni) and titanium dioxide nanoparticles (Nano-TiO2) would alter autophagy and apoptosis levels in normal human bronchial epithelial BEAS-2B cells and the underlying mechanisms involved in this process. Our results showed that the expressions of autophagy- and apoptosis-associated proteins were dysregulated in cells exposed to Nano-Ni. However, exposure to the same doses of Nano-TiO2 had no significant effects on these proteins. In addition, exposure to Nano-Ni, but not Nano-TiO2, led to nuclear accumulation of HIF-1α and decreased phosphorylation of mTOR in BEAS-2B cells. Inhibition of HIF-1α by CAY10585 abolished Nano-Ni-induced decreased phosphorylation of mTOR, while activation of mTOR by MHY1485 did not affect Nano-Ni-induced nuclear accumulation of HIF-1α. Furthermore, both HIF-1α inhibition and mTOR activation abolished Nano-Ni-induced autophagy but enhanced Nano-Ni-induced apoptosis. Blockage of autophagic flux by Bafilomycin A1 exacerbated Nano-Ni-induced apoptosis, while activation of autophagy by Rapamycin effectively rescued Nano-Ni-induced apoptosis. In conclusion, our results demonstrated that Nano-Ni exposure caused increased levels of autophagy and apoptosis via the HIF-1α/mTOR signaling axis. Nano-Ni-induced autophagy has a protective role against Nano-Ni-induced apoptosis. These findings provide us with further insight into Nano-Ni-induced toxicity.
Collapse
Affiliation(s)
- Jiali Yuan
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, 40202, USA
| | - Yiqun Mo
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, 40202, USA
| | - Yue Zhang
- Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Yuanbao Zhang
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, 40202, USA
| | - Qunwei Zhang
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, 40202, USA.
| |
Collapse
|
5
|
G S, S VP, E P, G A. Comparative synthesis and characterization of nanocomposites using chemical and green approaches including a comparison study on in vivo and in vitro biological properties. NANOSCALE ADVANCES 2023; 5:767-785. [PMID: 36756509 PMCID: PMC9890937 DOI: 10.1039/d2na00677d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 12/09/2022] [Indexed: 06/18/2023]
Abstract
In this study, the anti-diabetic, anti-inflammatory, anti-cytotoxic, and antibacterial effects of various substances were studied in vitro. Malachite green's photocatalytic effects were used to determine the optimised sample while it was exposed to visible light. The intended nanocomposites were created without any contaminants, according to XRD data. The overall characterisation results of the green synthesis of CS/SiO2/TiO2/CeO2/Fe3O4 nanocomposites (CSTCF(G)) were superior to those of the chemical synthesis of CS/SiO2/TiO2/CeO2/Fe3O4 nanocomposites (CSTCF(C)). At the five doses examined, the green synthesis of CS/SiO2/TiO2/CeO2/Fe3O4 nanocomposites (CSTCF(G)) and chemical synthesis of CS/SiO2/TiO2/CeO2/Fe3O4 nanocomposites (CSTCF(C)) resulted in higher α-glucosidase inhibition percentages in the antidiabetic assay. HaCaT cells and MCF-7 cells were less harmful when treated with chemically synthesized CS/SiO2/TiO2/CeO2/Fe3O4 nanocomposites (CSTCF(C)), and green synthesized CS/SiO2/TiO2/CeO2/Fe3O4 nanocomposites (CSTCF(G)). From the results of the cytotoxicity tests against MCF-7 cells and HaCaT cells using the nanocomposites, the IC50 values of Salacia reticulata, green synthesized CS/SiO2/TiO2/CeO2/Fe3O4 nanocomposites (CSTCF(G)), and chemically synthesized CS/SiO2/TiO2/CeO2/Fe3O4 nanocomposites (CSTCF(C)) were calculated. This research work shows that the green synthesized CS/SiO2/TiO2/CeO2/Fe3O4 nanocomposites (CSTCF(G)) have strong anti-inflammatory, antibacterial and anti-diabetic properties, as well as considerable suppression of high activation in in vivo zebrafish embryo toxicity. The novelty of this study focused on the revelation that green synthesized nanocomposites are more affordable, environmentally friendly and biocompatible than chemically synthesized ones.
Collapse
Affiliation(s)
- Sabeena G
- Sri Paramakalyani Centre of Excellence in Environmental Sciences, Manonmaniam Sundaranar University Alwarkurichi - 627412 India
| | - Vainath Praveen S
- Sri Paramakalyani Centre of Excellence in Environmental Sciences, Manonmaniam Sundaranar University Alwarkurichi - 627412 India
| | - Pushpalakshmi E
- Sri Paramakalyani Centre of Excellence in Environmental Sciences, Manonmaniam Sundaranar University Alwarkurichi - 627412 India
| | - Annadurai G
- Sri Paramakalyani Centre of Excellence in Environmental Sciences, Manonmaniam Sundaranar University Alwarkurichi - 627412 India
| |
Collapse
|
6
|
A weight of evidence review of the genotoxicity of titanium dioxide (TiO2). Regul Toxicol Pharmacol 2022; 136:105263. [DOI: 10.1016/j.yrtph.2022.105263] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/26/2022] [Accepted: 09/10/2022] [Indexed: 11/06/2022]
|
7
|
Yuan J, Mo L, Mo Y, Zhang Y, Zhang Y, Zhang Q. A protective role of autophagy in fine airborne particulate matter-induced apoptosis in LN-229 cells. Toxicology 2022; 477:153271. [PMID: 35872226 PMCID: PMC10825875 DOI: 10.1016/j.tox.2022.153271] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/15/2022] [Accepted: 07/20/2022] [Indexed: 01/16/2023]
Abstract
Air pollution is a public health threat and global epidemiological studies have shown that ambient air pollutants are closely related to various poor health conditions, including neurodegenerative diseases. Here, we evaluated the toxic effects and the underlying mechanisms of fine airborne particulate matter (PM2.5) on human glioblastoma LN-229 cells. Our results showed that exposure of LN-229 cells to PM2.5 (≥ 200 μg/mL) significantly reduced cell viability. PM2.5 exposure increased autophagy, apoptosis, and ROS production in the cells. Pre-treatment with a ROS scavenger, catalase, or depletion of mtDNA (ρ0 cells) abolished PM2.5-induced autophagy and apoptosis. PM2.5 exposure also activated MAPK signals in cells, which were blocked by catalase pre-treatment or mtDNA depletion. Furthermore, inhibition of JNK, but not ERK1/2 or p38, attenuated PM2.5-induced autophagy and apoptosis in cells. Finally, suppression of autophagy with Bafilomycin A1 or Beclin 1 siRNA exacerbated PM2.5-induced apoptosis, indicating a protective role of autophagy against PM2.5-induced apoptosis. Our results demonstrated that exposure of LN-229 cells to PM2.5 caused autophagy and apoptosis through PM2.5-induced ROS generation, mainly by mitochondria, and JNK activation. Autophagy may have a transient protective response in PM2.5-induced apoptosis. These findings have important implications for understanding the potential neurotoxicity of PM2.5.
Collapse
Affiliation(s)
- Jiali Yuan
- Department of Epidemiology & Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA
| | - Luke Mo
- duPont Manual High School, Louisville, KY, USA
| | - Yiqun Mo
- Department of Epidemiology & Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA
| | - Yuanbao Zhang
- Department of Epidemiology & Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA
| | - Yue Zhang
- Department of Epidemiology & Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA
| | - Qunwei Zhang
- Department of Epidemiology & Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
8
|
Mo Y, Zhang Y, Zhang Y, Yuan J, Mo L, Zhang Q. Nickel nanoparticle-induced cell transformation: involvement of DNA damage and DNA repair defect through HIF-1α/miR-210/Rad52 pathway. J Nanobiotechnology 2021; 19:370. [PMID: 34789290 PMCID: PMC8600818 DOI: 10.1186/s12951-021-01117-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 11/02/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Nickel nanoparticles (Nano-Ni) are increasingly used in industry and biomedicine with the development of nanotechnology. However, the genotoxic and carcinogenic effects of Nano-Ni and the underlying mechanisms are still unclear. METHODS At first, dose-response (0, 10, 20, and 30 μg/mL) and time-response (0, 3, 6, 12, and 24 h) studies were performed in immortalized normal human bronchial epithelial cells BEAS-2B to observe the effects of Nano-Ni on DNA damage response (DDR)-associated proteins and the HIF-1α/miR-210/Rad52 pathway by real-time PCR or Western blot. Then, a Hsp90 inhibitor (1 µM of 17-AAG, an indirect HIF-1α inhibitor), HIF-1α knock-out (KO) cells, and a miR-210 inhibitor (20 nM) were used to determine whether Nano-Ni-induced Rad52 down-regulation was through HIF-1α nuclear accumulation and miR-210 up-regulation. In the long-term experiments, cells were treated with 0.25 and 0.5 µg/mL of Nano-Ni for 21 cycles (~ 150 days), and the level of anchorage-independent growth was determined by plating the cells in soft agar. Transduction of lentiviral particles containing human Rad52 ORF into BEAS-2B cells was used to observe the role of Rad52 in Nano-Ni-induced cell transformation. Nano-Ni-induced DNA damage and dysregulation of HIF-1α/miR-210/Rad52 pathway were also investigated in vivo by intratracheal instillation of 50 µg per mouse of Nano-Ni. gpt delta transgenic mice were used to analyze mutant frequency and mutation spectrum in mouse lungs after Nano-Ni exposure. RESULTS Nano-Ni exposure caused DNA damage at both in vitro and in vivo settings, which was reflected by increased phosphorylation of DDR-associated proteins such as ATM at Ser1981, p53 at Ser15, and H2AX. Nano-Ni exposure also induced HIF-1α nuclear accumulation, miR-210 up-regulation, and down-regulation of homologous recombination repair (HRR) gene Rad52. Inhibition of or knocking-out HIF-1α or miR-210 ameliorated Nano-Ni-induced Rad52 down-regulation. Long-term low-dose Nano-Ni exposure led to cell malignant transformation, and augmentation of Rad52 expression significantly reduced Nano-Ni-induced cell transformation. In addition, increased immunostaining of cell proliferation markers, Ki-67 and PCNA, was observed in bronchiolar epithelial cells and hyperplastic pneumocytes in mouse lungs at day 7 and day 42 after Nano-Ni exposure. Finally, using gpt delta transgenic mice revealed that Nano-Ni exposure did not cause increased gpt mutant frequency and certain DNA mutations, such as base substitution and small base insertions/deletions, are not the main types of Nano-Ni-induced DNA damage. CONCLUSIONS This study unraveled the mechanisms underlying Nano-Ni-induced cell malignant transformation; the combined effects of Nano-Ni-induced DNA damage and DNA repair defects through HIF-1α/miR-210/Rad52 pathway likely contribute to Nano-Ni-induced genomic instability and ultimately cell transformation. Our findings will provide information to further elucidate the molecular mechanisms of Nano-Ni-induced genotoxicity and carcinogenicity.
Collapse
Affiliation(s)
- Yiqun Mo
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, 485 E. Gray Street, Louisville, KY 40202 USA
| | - Yue Zhang
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, 485 E. Gray Street, Louisville, KY 40202 USA
| | - Yuanbao Zhang
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, 485 E. Gray Street, Louisville, KY 40202 USA
| | - Jiali Yuan
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, 485 E. Gray Street, Louisville, KY 40202 USA
| | - Luke Mo
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, 485 E. Gray Street, Louisville, KY 40202 USA
| | - Qunwei Zhang
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, 485 E. Gray Street, Louisville, KY 40202 USA
| |
Collapse
|
9
|
EFSA Panel on Food Additives and Flavourings (FAF), Younes M, Aquilina G, Castle L, Engel K, Fowler P, Frutos Fernandez MJ, Fürst P, Gundert‐Remy U, Gürtler R, Husøy T, Manco M, Mennes W, Moldeus P, Passamonti S, Shah R, Waalkens‐Berendsen I, Wölfle D, Corsini E, Cubadda F, De Groot D, FitzGerald R, Gunnare S, Gutleb AC, Mast J, Mortensen A, Oomen A, Piersma A, Plichta V, Ulbrich B, Van Loveren H, Benford D, Bignami M, Bolognesi C, Crebelli R, Dusinska M, Marcon F, Nielsen E, Schlatter J, Vleminckx C, Barmaz S, Carfí M, Civitella C, Giarola A, Rincon AM, Serafimova R, Smeraldi C, Tarazona J, Tard A, Wright M. Safety assessment of titanium dioxide (E171) as a food additive. EFSA J 2021; 19:e06585. [PMID: 33976718 PMCID: PMC8101360 DOI: 10.2903/j.efsa.2021.6585] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The present opinion deals with an updated safety assessment of the food additive titanium dioxide (E 171) based on new relevant scientific evidence considered by the Panel to be reliable, including data obtained with TiO2 nanoparticles (NPs) and data from an extended one-generation reproductive toxicity (EOGRT) study. Less than 50% of constituent particles by number in E 171 have a minimum external dimension < 100 nm. In addition, the Panel noted that constituent particles < 30 nm amounted to less than 1% of particles by number. The Panel therefore considered that studies with TiO2 NPs < 30 nm were of limited relevance to the safety assessment of E 171. The Panel concluded that although gastrointestinal absorption of TiO2 particles is low, they may accumulate in the body. Studies on general and organ toxicity did not indicate adverse effects with either E 171 up to a dose of 1,000 mg/kg body weight (bw) per day or with TiO2 NPs (> 30 nm) up to the highest dose tested of 100 mg/kg bw per day. No effects on reproductive and developmental toxicity were observed up to a dose of 1,000 mg E 171/kg bw per day, the highest dose tested in the EOGRT study. However, observations of potential immunotoxicity and inflammation with E 171 and potential neurotoxicity with TiO2 NPs, together with the potential induction of aberrant crypt foci with E 171, may indicate adverse effects. With respect to genotoxicity, the Panel concluded that TiO2 particles have the potential to induce DNA strand breaks and chromosomal damage, but not gene mutations. No clear correlation was observed between the physico-chemical properties of TiO2 particles and the outcome of either in vitro or in vivo genotoxicity assays. A concern for genotoxicity of TiO2 particles that may be present in E 171 could therefore not be ruled out. Several modes of action for the genotoxicity may operate in parallel and the relative contributions of different molecular mechanisms elicited by TiO2 particles are not known. There was uncertainty as to whether a threshold mode of action could be assumed. In addition, a cut-off value for TiO2 particle size with respect to genotoxicity could not be identified. No appropriately designed study was available to investigate the potential carcinogenic effects of TiO2 NPs. Based on all the evidence available, a concern for genotoxicity could not be ruled out, and given the many uncertainties, the Panel concluded that E 171 can no longer be considered as safe when used as a food additive.
Collapse
|
10
|
Liu L, Kong L. Research progress on the carcinogenicity of metal nanomaterials. J Appl Toxicol 2021; 41:1334-1344. [PMID: 33527484 DOI: 10.1002/jat.4145] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 01/16/2021] [Accepted: 01/22/2021] [Indexed: 12/21/2022]
Abstract
With the rapid development of nanotechnology, new nanomaterials with enormous potentials continue to emerge, especially metal nanomaterials. Metal nanomaterials possess the characteristics of metals and nanomaterials, so they are widely used in many fields. But at the same time, whether the use or release of metal nan4omaterials into the environment is toxic to human beings and animals has now attained widespread attention at home and abroad. Currently, it is an indisputable fact that cancer ranks among the top causes of death among residents worldwide. The properties of causing DNA damage and mutations possessed by these metal nanomaterials make them unpredictable influences in the body, subsequently leading to genotoxicity and carcinogenicity. Due to the increasing evidence of their roles in carcinogenicity, this article reviews the toxicological and carcinogenic effects of metal nanomaterials, including nano-metal elements (nickel nanoparticles, silver nanoparticles, and cobalt nanoparticles) and nano-metal oxides (titanium dioxide nanoparticles, silica nanoparticles, zinc oxide nanoparticles, and alumina nanoparticles). This article provides a reference for the researchers and policymakers to use metal nanomaterials rationally in modern industries and biomedicine.
Collapse
Affiliation(s)
- Lin Liu
- Key Laboratory of Environment Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Lu Kong
- Key Laboratory of Environment Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| |
Collapse
|
11
|
Mo Y, Zhang Y, Mo L, Wan R, Jiang M, Zhang Q. The role of miR-21 in nickel nanoparticle-induced MMP-2 and MMP-9 production in mouse primary monocytes: In vitro and in vivo studies. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 267:115597. [PMID: 33254626 PMCID: PMC7708676 DOI: 10.1016/j.envpol.2020.115597] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/12/2020] [Accepted: 09/02/2020] [Indexed: 05/02/2023]
Abstract
Exposure to metal nanoparticles causes both pulmonary and systemic effects. Nanoparticles can enter the circulation and act directly or indirectly on blood cells, such as monocytes. Monocytes/macrophages are among the first cells to home to inflammatory sites and play a key role in the immune response. Here we investigated the effects of nickel nanoparticles (Nano-Ni), partially [O]-passivated Nano-Ni (Nano-Ni-P), and carbon-coated Nano-Ni (Nano-Ni-C) on MMP-2 and MMP-9 production in mouse primary monocytes both in vitro and in vivo and explored the potential mechanisms involved. The dose- and time-response studies showed that exposure of primary monocytes from wild-type (WT) mice to 30 μg/mL of Nano-Ni for 24 h caused significant MMP-2 and MMP-9 production; therefore, these dose and time point were chosen for the following in vitro studies. Nano-Ni and Nano-Ni-P caused miR-21 upregulation, as well as MMP-2, MMP-9, TIMP-1 and TIMP-2 upregulation in monocytes from WT, but not miR-21 knock-out (KO), mice, indicating the important role of miR-21 in Nano-Ni-induced MMPs and TIMPs upregulation. However, Nano-Ni-C did not cause these effects, suggesting surface modification of Nano-Ni, such as carbon coating, alleviates Nano-Ni-induced miR-21 and MMPs upregulation. These results were further confirmed by in vivo studies by intratracheal instillation of nickel nanoparticles into WT and miR-21 KO mice. Finally, our results demonstrated that exposure of primary monocytes from WT mice to Nano-Ni and Nano-Ni-P caused downregulation of RECK, a direct miR-21 target, suggesting the involvement of miR-21/RECK pathway in Nano-Ni-induced MMP-2 and MMP-9 production.
Collapse
Affiliation(s)
- Yiqun Mo
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA
| | - Yue Zhang
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA
| | - Luke Mo
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA
| | - Rong Wan
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA
| | - Mizu Jiang
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA
| | - Qunwei Zhang
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
12
|
Mo Y, Zhang Y, Wan R, Jiang M, Xu Y, Zhang Q. miR-21 mediates nickel nanoparticle-induced pulmonary injury and fibrosis. Nanotoxicology 2020; 14:1175-1197. [PMID: 32924694 PMCID: PMC7984410 DOI: 10.1080/17435390.2020.1808727] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 07/18/2020] [Accepted: 08/06/2020] [Indexed: 12/26/2022]
Abstract
We and other groups have demonstrated that exposure to nickel nanoparticles (Nano-Ni) results in severe and persistent lung inflammation and fibrosis, but the underlying mechanisms remain unclear. Here, we propose that miR-21 may play an important role in Nano-Ni-induced lung inflammation, injury, and fibrosis. Our dose- and time-response studies demonstrated that exposure of C57BL/6J (WT) mice to Nano-Ni resulted in upregulation of miR-21, proinflammatory cytokines, and profibrotic mediators. Histologically, exposure to Nano-Ni caused severe pulmonary inflammation and fibrosis. Based on the dose- and time-response studies, we chose a dose of 50 µg of Nano-Ni per mouse to compare the effects of Nano-Ni on WT with those on miR-21 KO mouse lungs. At day 3 post-exposure, Nano-Ni caused severe acute lung inflammation and injury that were reflected by increased neutrophil count, CXCL1/KC level, LDH activity, total protein concentration, MMP-2/9 protein levels and activities, and proinflammatory cytokines in the BALF or lung tissues from WT mice, which were confirmed histologically. Although Nano-Ni had similar effects on miR-21 KO mice, the above-mentioned levels were significantly lower than those in WT mice. Histologically, lungs from WT mice exposed to Nano-Ni had infiltration of a large number of polymorphonuclear (PMN) cells and macrophages in the alveolar space and interstitial tissues. However, exposure of miR-21 KO mice to Nano-Ni only caused mild acute lung inflammation and injury. At day 42 post-exposure, Nano-Ni caused extensive pulmonary fibrosis and chronic inflammation in the WT mouse lungs. However, exposure of miR-21 KO mice to Nano-Ni only caused mild lung fibrosis and chronic lung inflammation. Our results also showed that exposure to Nano-Ni caused upregulation of TGF-β1, phospho-Smad2, COL1A1, and COL3A1 in both WT and miR-21 KO mouse lungs. However, levels were significantly lower in miR-21 KO mice than in WT mice, except TGF-β1, which was similar in both kinds of mice. Decreased expression of Smad7 was observed in WT mouse lungs, but not in miR-21 KO mice. Our results demonstrated that knocking out miR-21 ameliorated Nano-Ni-induced pulmonary inflammation, injury, and fibrosis, suggesting the important role of miR-21 in Nano-Ni-induced pulmonary toxicity.
Collapse
Affiliation(s)
- Yiqun Mo
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA
| | - Yue Zhang
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA
| | - Rong Wan
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA
| | - Mizu Jiang
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA
| | - Youqiong Xu
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA
| | - Qunwei Zhang
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA
| |
Collapse
|
13
|
Feng S, Zhang Z, Mo Y, Tong R, Zhong Z, Chen Z, He D, Wan R, Gao M, Mo Y, Zhang Q, Huang Y. Activation of NLRP3 inflammasome in hepatocytes after exposure to cobalt nanoparticles: The role of oxidative stress. Toxicol In Vitro 2020; 69:104967. [PMID: 32805375 DOI: 10.1016/j.tiv.2020.104967] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/21/2020] [Accepted: 08/09/2020] [Indexed: 12/21/2022]
Abstract
With the increased use of nanomaterials and increased exposure of humans to various nanomaterials, the potential health effects of nanomaterials cannot be ignored. The hepatotoxicity of cobalt nanoparticles (Nano-Co) is largely unknown and the underlying mechanisms remain obscure. The purpose of this study was to exam the hepatotoxicity induced by Nano-Co and its potential mechanisms. Our results showed that exposure of human fetal hepatocytes L02 to Nano-Co caused a dose- and a time-dependent cytotoxicity. Besides the generation of reactive oxygen species (ROS) and mitochondrial reactive oxygen species (mtROS), exposure to Nano-Co also caused activation of NOD-like receptor protein 3 (NLRP3) inflammasome in hepatocytes. After silencing NLRP3, one component of NLRP3 inflammasome, expression by siRNA strategy, we found that upregulation of NLRP3-related proteins was abolished in hepatocytes exposed to Nano-Co. Using antioxidants to scavenge ROS and mtROS, we demonstrated that Nano-Co-induced mtROS generation was related to Nano-Co-induced NLRP3 inflammasome activation. Our findings demonstrated that Nano-Co exposure may promote intracellular oxidative stress damage, and mtROS may mediate the activation of NLRP3 inflammasome in hepatocytes exposed to Nano-Co, suggesting an important role of ROS/NLRP3 pathway in Nano-Co-induced hepatotoxicity. These results provide scientific insights into the hepatotoxicity of Nano-Co and a basis for the prevention and treatment of Nano-Co-induced cytotoxicity.
Collapse
Affiliation(s)
- Sisi Feng
- Department of Pathology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, PR China
| | - Zhenyu Zhang
- Department of Emergency, Xiang'An Hospital of Xiamen University, Xiamen, Fujian, PR China
| | - Yiqing Mo
- Community Health Care Center, Changqing Chaoming Street, Xiacheng District, Hangzhou, Zhejiang, PR China
| | - Ruirui Tong
- Department of Pathology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, PR China
| | - Zexiang Zhong
- Department of Spine Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, PR China
| | - Zhong Chen
- Department of Pathology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, PR China
| | - Dan He
- Department of Pathology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, PR China
| | - Rong Wan
- Department of Pathology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, PR China; Institute of Oncology, Fujian Medical University, Fuzhou, Fujian, PR China; Diagnostic Pathology Center, Fujian Medical University, Fuzhou, Fujian, PR China
| | - Meiqin Gao
- Department of Pathology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, PR China; Institute of Oncology, Fujian Medical University, Fuzhou, Fujian, PR China; Diagnostic Pathology Center, Fujian Medical University, Fuzhou, Fujian, PR China
| | - Yiqun Mo
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA
| | - Qunwei Zhang
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA
| | - Yang Huang
- Department of Pathology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, PR China; Institute of Oncology, Fujian Medical University, Fuzhou, Fujian, PR China; Diagnostic Pathology Center, Fujian Medical University, Fuzhou, Fujian, PR China.
| |
Collapse
|
14
|
Laumonier T, Ruffieux E, Paccaud J, Kindler V, Hannouche D. In vitro evaluation of human myoblast function after exposure to cobalt and chromium ions. J Orthop Res 2020; 38:1398-1406. [PMID: 31883135 DOI: 10.1002/jor.24579] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 12/23/2019] [Indexed: 02/04/2023]
Abstract
The replacement of a native hip joint by a metal-on-metal prosthesis may induce deleterious inflammatory side effects that are associated with the release of wear particles and metal ions. These events are referred to the adverse reaction to metal debris (ARMD) and the adverse local tissue reaction (ALTR). While wear particles seem involved in ARMD, the role of metal ions in ALTR and their impact on myoblasts, located in the prosthesis vicinity, has not been fully identified. To clarify this issue we investigated, using an in vitro culture system, the effect of cobalt and/or chromium ions (Co2+ and/or Cr3+ ) on human myoblast proliferation, cellular differentiation, and inflammatory marker expression. Freshly isolated human myoblasts were cultured in media supplemented with graded concentrations of Co2+ and/or Cr3+ . Co2+ induced a concentration-dependent decrease of both myoblast viability and myogenic differentiation while Cr3+ did not. Co2+ or Co2+ /Cr3+ also induced the upregulation of ICAM-1, whereas HLA-DR expression was unaffected. Moreover, allogenic monocytes induced the synergistic increase of Co2+ -induced ICAM-1 expression. We also found that Co2+ stabilized HIF-1α and increased TLR4, tumor necrosis factor-alpha (TNF-α), and interleukin 1β (IL-1β) expression in a dose and time-dependent manner in human myoblasts. This study showed that Co2+ , but not Cr3+ , was toxic toward myoblasts and induced, in the surviving cells, expression of inflammatory markers such as ICAM-1, TLR4, TNF-α, and IL-1β. This suggests that Co2+ , most efficiently in the presence of monocytes, may be a key inducer of ALTR, which may, if severe and long-lasting, eventually result in prosthesis loosening.
Collapse
Affiliation(s)
- Thomas Laumonier
- Department of Orthopedic Surgery, Faculty of Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Etienne Ruffieux
- Department of Orthopedic Surgery, Faculty of Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Joris Paccaud
- Department of Orthopedic Surgery, Faculty of Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Vincent Kindler
- Department of Orthopedic Surgery, Faculty of Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Didier Hannouche
- Department of Orthopedic Surgery, Faculty of Medicine, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
15
|
Danilevich VN, Kozlov SA, Shevchuk TV, Oleinikov VA, Sizova SV, Khodarovich YM, Mulyukin AL. Ribonucleic acid (RNA) condensation by thermal cycling with metal cations: yield of nanoparticles and their applicability for transfection. J Biomol Struct Dyn 2019; 38:3959-3971. [PMID: 31543001 DOI: 10.1080/07391102.2019.1671228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
To the present, different efficient but expensive, multistage, and time-consuming technologies have been developed to deliver ribonucleic acids (RNA) into eukaryotic cells. Here, we report a simple and feasible solution to design RNA nanocarriers based on nucleic acid condensation by bi- and trivalent metal ions during thermal cycling. Efficient RNA conversion to nanoparticles with small size (10-50 nm) suitable for transfection was achieved using cations Ni2+, Co2+ or Cu2+ alone or in combination with Ca2+ at the specially selected concentrations (2.0 mM-3.5 mM), low ionic strength, and narrow pH range (8.0-8.5). Other ions - Mn2+, Zn2+, Tb3+, or Gd3+ - caused RNA-cleaving effect that was abolished in the presence of Ni2+, Co2+, Zn2+, or Cu2+. Naked RNA-metal ion nanoparticles were extremely unstable in phosphate buffer and sensitive to serum ribonucleases (RNases), and this problem was solved by treatment with polyarginines-16 and 8. Polyarginine-stabilized nanoparticles, containing malachite green (MG) aptamer RNA and metal cations, crossed the cell membrane, dissociated in the cytoplasm, and preserved the functionality of transported RNA, as judged from efficient transfection of human embryonic kidney 293 cells. The technology, involving RNA condensation by metal cations, can be used as a cheap alternative to produce nanoscale carriers to deliver various RNAs into cells in vitro and in vivo.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Vasily N Danilevich
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Science, Moscow, Russia
| | - Sergey A Kozlov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Science, Moscow, Russia
| | - Taras V Shevchuk
- Branch of the M. M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Pushchino, Moscow, Russia
| | - Vladimir A Oleinikov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Science, Moscow, Russia
| | - Svetlana V Sizova
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Science, Moscow, Russia
| | - Yuriy M Khodarovich
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Science, Moscow, Russia
| | - Andrey L Mulyukin
- Winogradsky Institute of Microbiology, Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
16
|
Kanwal Z, Raza MA, Riaz S, Manzoor S, Tayyeb A, Sajid I, Naseem S. Synthesis and characterization of silver nanoparticle-decorated cobalt nanocomposites (Co@AgNPs) and their density-dependent antibacterial activity. ROYAL SOCIETY OPEN SCIENCE 2019; 6:182135. [PMID: 31218038 PMCID: PMC6549958 DOI: 10.1098/rsos.182135] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 03/26/2019] [Indexed: 05/10/2023]
Abstract
Magnetic cores loaded with metallic nanoparticles can be promising nano-carriers for successful drug delivery at infectious sites. We report fabrication, characteristic analysis and in vitro antibacterial performance of nanocomposites comprising cobalt cores (Co-cores) functionalized with a varied concentration of silver nanoparticles (AgNPs). A two-step polyol process synchronized with the transmetalation reduction method was used. Co-cores were synthesized with cobalt acetate, and decoration of AgNPs was carried out with silver acetate. The density of AgNPs was varied by changing the amount of silver content as 0.01, 0.1 and 0.2 g in the synthesis solution. Both AgNPs and Co-cores were spherical having a size range of 30-80 nm and 200 nm to more than 1 µm, respectively, as determined by scanning electron microscopy. The metallic nature and face-centred cubic crystalline phase of prepared nanocomposites were confirmed by X-ray diffraction. Biocompatibility analysis confirmed high cell viability of MCF7 at low concentrations of tested particles. The antibacterial performance of nanocomposites (Co@AgNPs) against Escherichia coli and Bacillus subtilis was found to be AgNPs density-dependent, and nanocomposites with the highest AgNPs density exhibited the maximum bactericidal efficacy. We therefore propose that Co@AgNPs as effective drug containers for various biomedical applications.
Collapse
Affiliation(s)
- Zakia Kanwal
- Department of Zoology, Lahore College for Women University, Jail Road, Lahore 54000, Pakistan
| | - Muhammad Akram Raza
- Centre of Excellence in Solid State Physics, University of the Punjab, Quid-e-Azam Campus, Lahore 54590, Pakistan
| | - Saira Riaz
- Centre of Excellence in Solid State Physics, University of the Punjab, Quid-e-Azam Campus, Lahore 54590, Pakistan
| | - Saher Manzoor
- Centre of Excellence in Solid State Physics, University of the Punjab, Quid-e-Azam Campus, Lahore 54590, Pakistan
| | - Asima Tayyeb
- School of Biological Sciences, University of the Punjab, Quid-e-Azam Campus, Lahore 54590, Pakistan
| | - Imran Sajid
- Department of Microbiology and Molecular Genetics, University of the Punjab, Quid-e-Azam Campus, Lahore 54590, Pakistan
| | - Shahzad Naseem
- Centre of Excellence in Solid State Physics, University of the Punjab, Quid-e-Azam Campus, Lahore 54590, Pakistan
| |
Collapse
|
17
|
Ertuğrul H, Yalçın B, Güneş M, Kaya B. Ameliorative effects of melatonin against nano and ionic cobalt induced genotoxicity in two in vivo Drosophila assays. Drug Chem Toxicol 2019; 43:279-286. [DOI: 10.1080/01480545.2019.1585444] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Havva Ertuğrul
- Faculty of Sciences, Department of Biology, Akdeniz University, Antalya, Turkey
| | - Burçin Yalçın
- Faculty of Sciences, Department of Biology, Akdeniz University, Antalya, Turkey
| | - Merve Güneş
- Faculty of Sciences, Department of Biology, Akdeniz University, Antalya, Turkey
| | - Bülent Kaya
- Faculty of Sciences, Department of Biology, Akdeniz University, Antalya, Turkey
| |
Collapse
|
18
|
Lison D, van den Brule S, Van Maele-Fabry G. Cobalt and its compounds: update on genotoxic and carcinogenic activities. Crit Rev Toxicol 2018; 48:522-539. [PMID: 30203727 DOI: 10.1080/10408444.2018.1491023] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
This article summarizes recent experimental and epidemiological data on the genotoxic and carcinogenic activities of cobalt compounds. Emphasis is on the respiratory system, but endogenous exposure from Co-containing alloys used in endoprostheses, and limited data on nanomaterials and oral exposures are also considered. Two groups of cobalt compounds are differentiated on the basis of their mechanisms of toxicity: (1) those essentially involving the solubilization of Co(II) ions, and (2) metallic materials for which both surface corrosion and release of Co(II) ions act in concert. For both groups, identified genotoxic and carcinogenic mechanisms are non-stochastic and thus expected to exhibit a threshold. Cobalt compounds should, therefore, be considered as genotoxic carcinogens with a practical threshold. Accumulating evidence indicates that chronic inhalation of cobalt compounds can induce respiratory tumors locally. No evidence of systemic carcinogenicity upon inhalation, oral or endogenous exposure is available. The scarce data available for Co-based nanosized materials does not allow deriving a specific mode of action or assessment for these species.
Collapse
Affiliation(s)
- D Lison
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Catholic University of Louvain, Brussels, Belgium
| | - S van den Brule
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Catholic University of Louvain, Brussels, Belgium
| | - G Van Maele-Fabry
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Catholic University of Louvain, Brussels, Belgium
| |
Collapse
|
19
|
Vieira LFDA, Lins MP, Viana IMMN, dos Santos JE, Smaniotto S, Reis MDDS. Metallic nanoparticles reduce the migration of human fibroblasts in vitro. NANOSCALE RESEARCH LETTERS 2017; 12:200. [PMID: 28314368 PMCID: PMC5355407 DOI: 10.1186/s11671-017-1982-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Accepted: 03/06/2017] [Indexed: 05/04/2023]
Abstract
Nanoparticles have extremely wide applications in the medical and biological fields. They are being used in biosensors, local drug delivery, diagnostics, and medical therapy. However, the potential effects of nanoparticles on target cell and tissue function, apart from cytotoxicity, are not completely understood. Thus, the aim of this study was to investigate the in vitro effects of silver nanoparticles (AgNPs) and gold nanoparticles (AuNPs) on human fibroblasts with respect to their interaction with the extracellular matrix and in cell migration. Immunofluorescence analysis revealed that treatment with AgNPs or AuNPs decreased collagen and laminin production at all the concentrations tested (0.1, 1, and 10 μg/mL). Furthermore, cytofluorometric analysis showed that treatment with AgNPs reduced the percentage of cells expressing the collagen receptor very late antigen 2, α2β1 integrin (VLA-2) and the laminin receptor very late antigen 6, α6β1 integrin (VLA-6). In contrast, AuNP treatment increased and decreased the percentages of VLA-2-positive and VLA-6-positive cells, respectively, as compared to the findings for the controls. Analysis of cytoskeletal reorganization showed that treatment with both types of nanoparticles increased the formation of stress fibres and number of cell protrusions and impaired cell polarity. Fibroblasts exposed to different concentrations of AuNPs and AgNPs showed reduced migration through transwell chambers in the functional chemotaxis assay. These results demonstrated that metal nanoparticles may influence fibroblast function by negatively modulating the deposition of extracellular matrix molecules (ECM) and altering the expression of ECM receptors, cytoskeletal reorganization, and cell migration.
Collapse
Affiliation(s)
- Larissa Fernanda de Araújo Vieira
- Laboratory of Cell Biology, Institute of Biological Sciences and Health, Federal University of Alagoas, CEP 57072-970 Maceió, Alagoas Brazil
| | - Marvin Paulo Lins
- Laboratory of Cell Biology, Institute of Biological Sciences and Health, Federal University of Alagoas, CEP 57072-970 Maceió, Alagoas Brazil
| | - Iana Mayane Mendes Nicácio Viana
- Laboratory of Cell Biology, Institute of Biological Sciences and Health, Federal University of Alagoas, CEP 57072-970 Maceió, Alagoas Brazil
| | - Jeniffer Estevão dos Santos
- Laboratory of Cell Biology, Institute of Biological Sciences and Health, Federal University of Alagoas, CEP 57072-970 Maceió, Alagoas Brazil
| | - Salete Smaniotto
- Laboratory of Cell Biology, Institute of Biological Sciences and Health, Federal University of Alagoas, CEP 57072-970 Maceió, Alagoas Brazil
| | - Maria Danielma dos Santos Reis
- Laboratory of Cell Biology, Institute of Biological Sciences and Health, Federal University of Alagoas, CEP 57072-970 Maceió, Alagoas Brazil
| |
Collapse
|
20
|
Wan R, Mo Y, Zhang Z, Jiang M, Tang S, Zhang Q. Cobalt nanoparticles induce lung injury, DNA damage and mutations in mice. Part Fibre Toxicol 2017; 14:38. [PMID: 28923112 PMCID: PMC5604172 DOI: 10.1186/s12989-017-0219-z] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 09/11/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND We and other groups have demonstrated that exposure to cobalt nanoparticles (Nano-Co) caused oxidative stress and inflammation, which have been shown to be strongly associated with genotoxic and carcinogenic effects. However, few studies have reported Nano-Co-induced genotoxic effects in vivo. Here, we propose that Nano-Co may have high genotoxic effects due to their small size and high surface area, which have high capacity for causing oxidative stress and inflammation. METHODS gpt delta transgenic mice were used as our in vivo study model. They were intratracheally instilled with 50 μg per mouse of Nano-Co. At day 1, 3, 7 and 28 after exposure, bronchoalveolar lavage (BAL) was performed and the number of neutrophils, CXCL1/KC level, LDH activity and concentration of total protein in the BAL fluid (BALF) were determined. Mouse lung tissues were collected for H&E staining, and Ki-67, PCNA and γ-H2AX immunohistochemical staining. 8-OHdG level in the genomic DNA of mouse lungs was determined by an OxiSelect™ Oxidative DNA Damage ELISA Kit, and mutant frequency and mutation spectrum in the gpt gene were also determined in mouse lungs at four months after Nano-Co exposure by 6-TG selection, colony PCR, and DNA sequencing. RESULTS Exposure of mice to Nano-Co (50 μg per mouse) resulted in extensive acute lung inflammation and lung injury which were reflected by increased number of neutrophils, CXCL1/KC level, LDH activity and concentration of total protein in the BALF, and infiltration of large amount of neutrophils and macrophages in the alveolar space and interstitial tissues. Increased immunostaining of cell proliferation markers, Ki-67 and PCNA, and the DNA damage marker, γ-H2AX, was also observed in bronchiolar epithelial cells and hyperplastic type II pneumocytes in mouse lungs at day 7 after Nano-Co exposure. At four months after exposure, extensive interstitial fibrosis and proliferation of interstitial cells with inflammatory cells infiltrating the alveolar septa were observed. Moreover, Nano-Co caused increased level of 8-OHdG in genomic DNA of mouse lung tissues. Nano-Co also induced a much higher mutant frequency as compared to controls, and the most common mutation was G:C to T:A transversion, which may be explained by Nano-Co-induced increased formation of 8-OHdG. CONCLUSION Our study demonstrated that exposure to Nano-Co caused oxidative stress, lung inflammation and injury, and cell proliferation, which further resulted in DNA damage and DNA mutation. These findings have important implications for understanding the potential health effects of nanoparticle exposure.
Collapse
Affiliation(s)
- Rong Wan
- Department of Pathology, Fujian Medical University, Fuzhou, People’s Republic of China
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, 485 E. Gray Street, Louisville, KY 40202 USA
| | - Yiqun Mo
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, 485 E. Gray Street, Louisville, KY 40202 USA
| | - Zhenyu Zhang
- Seven-year Program of Clinical Medicine, Fujian Medical University, Fuzhou, People’s Republic of China
| | - Mizu Jiang
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, 485 E. Gray Street, Louisville, KY 40202 USA
- Department of Gastroenterology, Children’s Hospital, Zhejiang University, Hangzhou, People’s Republic of China
| | - Shichuan Tang
- Beijing Municipal Institute of Labor Protection, Beijing, People’s Republic of China
| | - Qunwei Zhang
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, 485 E. Gray Street, Louisville, KY 40202 USA
- Beijing Municipal Institute of Labor Protection, Beijing, People’s Republic of China
- Department of Preventive Medicine, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, 350122 People’s Republic of China
| |
Collapse
|
21
|
Che B, Luo Q, Zhai B, Fan G, Liu Z, Cheng K, Xin L. Cytotoxicity and genotoxicity of nanosilver in stable GADD45α promoter-driven luciferase reporter HepG2 and A549 cells. ENVIRONMENTAL TOXICOLOGY 2017; 32:2203-2211. [PMID: 28568508 DOI: 10.1002/tox.22433] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 04/25/2017] [Accepted: 04/25/2017] [Indexed: 06/07/2023]
Abstract
OBJECTIVES The intense commercial application of silver nanoparticles (AgNPs) has been raising concerns about their potential adverse health effects to human. This study aimed to explore the potency of AgNPs to induce GADD45α gene, an important stress sensor, and its relationships with the cytotoxicity and genotoxicity elicited by AgNPs. METHODS Two established HepG2 and A549 cell lines containing the GADD45α promoter-driven luciferase reporter were treated with increasing concentrations of AgNPs for 48 hours. After the treatment, transcriptional activation of GADD45α indicated by luciferase activity, cell viability, cell cycle arrest, and levels of genotoxicity were determined. The uptake and intracellular localization of AgNPs, cellular Ag doses as well as Ag+ release were also detected. RESULTS AgNPs could activate GADD45α gene at the transcriptional level as demonstrated by the dose-dependent increases in luciferase activity in both the reporter cells. The relative luciferase activity was greater than 12× the control level in HepG2-luciferase cells at the highest concentration tested where the cell viability decreased to 17.0% of the control. These results was generally in accordance with the positive responses in cytotoxicity, cell cycle arrest of Sub G1 and G2/M phase, Olive tail moment, micronuclei frequency, and the cellular Ag content. CONCLUSIONS The cytotoxicity and genotoxicity of AgNPs seems to occur mainly via particles uptake and the subsequent liberation of ions inside the cells. And furthermore, the GADD45α promoter-driven luciferase reporter cells, especially the HepG2-luciferase cells, could provide a new and valuable tool for predicting nanomaterials genotoxicity in humans.
Collapse
Affiliation(s)
- Bizhong Che
- School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou, Jiangsu, 215123, China
| | - Qiulin Luo
- School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou, Jiangsu, 215123, China
| | - Bingzhong Zhai
- School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou, Jiangsu, 215123, China
| | - Guoqiang Fan
- Suzhou Industrial Park Centers for Disease Control and Prevention, 58 Suqian Road, Suzhou, Jiangsu, China
| | - Zhiyong Liu
- School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou, 215123, China
| | - Kaiming Cheng
- Suzhou Industrial Park Centers for Disease Control and Prevention, 58 Suqian Road, Suzhou, Jiangsu, China
| | - Lili Xin
- School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou, Jiangsu, 215123, China
| |
Collapse
|
22
|
Nyga A, Hart A, Tetley TD. Importance of the HIF pathway in cobalt nanoparticle-induced cytotoxicity and inflammation in human macrophages. Nanotoxicology 2015; 9:905-17. [PMID: 25676618 DOI: 10.3109/17435390.2014.991430] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Recent, unexpected high failure rates of metal-on-metal hip implants have reintroduced the issue of cobalt toxicity. An adverse reaction to cobalt ions and cobalt-induced lung injury occurs during environmental exposure and is now strictly controlled. Currently adverse reaction occurs to cobalt nanoparticles during wear and tear of metal-on-metal hip implants of which the underlying mechanism is not fully understood. The putative role of the hypoxia-inducible factor (HIF) pathway in the mechanism of cobalt nanoparticle (Co-NPs) toxicity was examined using the U937 cell line, human alveolar macrophages and monocyte-derived macrophages. Co-NPs (5-20 μg/ml)-induced cytotoxicity (viability ranged from 75% to <20% of control, respectively) and reactive oxygen species (ROS), whereas a comparable concentration of cobalt ions (Co(II); up to 350 μM) did not. Co-NPs induced HIF-1α stabilization. Addition of ascorbic acid (100 µM) and glutathione (1 mM) both prevented the increased ROS. However, only treatment with ascorbic acid reduced HIF-1α levels and prevented cell death, indicating that a ROS-independent pathway is involved in Co-NPs-induced cytotoxicity. Replenishing intracellular ascorbate, which is crucial in preventing HIF pathway activation, modified Co-induced HIF target gene expression and the inflammatory response, by decreasing interleukin-1 beta (IL-1β) mRNA and protein expression. Addition of glutathione had no effect on Co-NPs-induced HIF target gene expression or inflammatory response. Thus, Co-NPs induce the HIF pathway by depleting intracellular ascorbate, leading to HIF stabilization and pathway activation. This suggests a strong, ROS-independent role for HIF activation in Co-NPs-induced cytotoxicity and a possible role for HIF in metal-on-metal hip implant pathology.
Collapse
Affiliation(s)
- Agata Nyga
- a National Heart & Lung Institute, Imperial College London , London , UK and
| | - Alister Hart
- b Institute of Orthopaedics & Musculoskeletal Science, Royal National Orthopaedic Hospital, University College London , London , UK
| | - Teresa D Tetley
- a National Heart & Lung Institute, Imperial College London , London , UK and
| |
Collapse
|