Pub Date : 2022-11-01DOI: 10.1080/17435390.2023.2177205
Min-Sung Kang, Gwang-Hee Lee, Mi-Jin Yang, Myeong-Chang Sung, Hyoung-Yun Han, Byoung-Seok Lee, Bosung Baek, Dong-Wan Kim, Eun-Jung Park
Pulmonary effects of inhaled microfibers are an emerging public health concern. In this study, we investigated toxicity following pulmonary exposure to synthetic polyethylene oxide fibroin (PEONF) and silk fibroin (SFNF) nanofibers and the cellular responses. When instilled intratracheally weekly for four weeks, body weight gain was significantly reduced in female mice exposed to the higher dose of SFNF when compared with the control group. The total number of cells in the lungs was more significant in all treated groups than in the control, whereas the relative portion of neutrophils and eosinophils increased significantly only in female mice exposed to SFNF. Both types of nanofibers induced notable pathological changes and increased pulmonary expression of MCP-1α, CXCL1, and TGF-β. More importantly, blood calcium, creatinine kinase, sodium, and chloride concentration were affected significantly, showing sex- and material-dependent differences. The relative portion of eosinophils increased only in SFNF-treated mice. In addition, both types of nanofibers induced necrotic and late apoptotic cell death in alveolar macrophages after 24 h of exposure, with accompanying oxidative stress, increased NO production, cell membrane rupture, intracellular organelle damage, and intracellular calcium accumulation. Additionally, multinucleated giant cells were formed in cells exposed to PEONF or SFNF. Taken together, the findings indicate that inhaled PEONF and SFNF may cause systemic adverse health effects with lung tissue damage, showing differences by sex- and material. Furthermore, PEONF- and SFNF-induced inflammatory response may be partly due to the low clearance of dead (or damaged) pulmonary cells and the excellent durability of PEONF and SFNF.
{"title":"Comparison of toxicity and cellular responses following pulmonary exposure to different types of nanofibers.","authors":"Min-Sung Kang, Gwang-Hee Lee, Mi-Jin Yang, Myeong-Chang Sung, Hyoung-Yun Han, Byoung-Seok Lee, Bosung Baek, Dong-Wan Kim, Eun-Jung Park","doi":"10.1080/17435390.2023.2177205","DOIUrl":"https://doi.org/10.1080/17435390.2023.2177205","url":null,"abstract":"<p><p>Pulmonary effects of inhaled microfibers are an emerging public health concern. In this study, we investigated toxicity following pulmonary exposure to synthetic polyethylene oxide fibroin (PEONF) and silk fibroin (SFNF) nanofibers and the cellular responses. When instilled intratracheally weekly for four weeks, body weight gain was significantly reduced in female mice exposed to the higher dose of SFNF when compared with the control group. The total number of cells in the lungs was more significant in all treated groups than in the control, whereas the relative portion of neutrophils and eosinophils increased significantly only in female mice exposed to SFNF. Both types of nanofibers induced notable pathological changes and increased pulmonary expression of MCP-1α, CXCL1, and TGF-β. More importantly, blood calcium, creatinine kinase, sodium, and chloride concentration were affected significantly, showing sex- and material-dependent differences. The relative portion of eosinophils increased only in SFNF-treated mice. In addition, both types of nanofibers induced necrotic and late apoptotic cell death in alveolar macrophages after 24 h of exposure, with accompanying oxidative stress, increased NO production, cell membrane rupture, intracellular organelle damage, and intracellular calcium accumulation. Additionally, multinucleated giant cells were formed in cells exposed to PEONF or SFNF. Taken together, the findings indicate that inhaled PEONF and SFNF may cause systemic adverse health effects with lung tissue damage, showing differences by sex- and material. Furthermore, PEONF- and SFNF-induced inflammatory response may be partly due to the low clearance of dead (or damaged) pulmonary cells and the excellent durability of PEONF and SFNF.</p>","PeriodicalId":18899,"journal":{"name":"Nanotoxicology","volume":"16 9-10","pages":"935-954"},"PeriodicalIF":5.0,"publicationDate":"2022-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9465389","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-11-01DOI: 10.1080/17435390.2022.2159891
Małgorzata Kubczak, Sylwia Michlewska, Michael Karimov, Alexander Ewe, Achim Aigner, Maria Bryszewska, Maksim Ionov
Polyethylenimines (PEIs) have been previously introduced for siRNA delivery. In particular, in the case of higher molecular weight PEIs, this is associated with toxicity, while low molecular weight PEIs are often insufficient for siRNA complexation. The tyrosine-modification of PEIs has been shown to enhance PEI efficacy and biocompatibility. This paper evaluates a set of tyrosine-modified low molecular weight linear or branched polyethylenimines as efficient carriers of siRNA. Complexation efficacies and biophysical complex properties were analyzed by zeta potential, dynamic light scattering and circular dichroism measurements as well as gel electrophoresis. Biological knockdown was studied in 2 D cell culture and 3 D ex vivo tissue slice air-liquid interface culture. The results demonstrate that siRNAs were able to form stable complexes with all tested polymers. Complexation was able to protect siRNA from degradation by RNase and to mediate target gene knockdown, as determined on the mRNA level and in PC3-Luc3/EGFP and HCT116-Luc3/EGFP expressing reporter cells on the protein level, using flow cytometry and confocal microscopy. The direct comparison of the studied polymers revealed differences in biological efficacies. Moreover, the tyrosine-modified PEIs showed high biocompatibility, as determined by LDH release and mitochondria integrity (J-aggregate assay) as well as caspase 3/7 (apoptosis) and H2O2 levels (ROS). In 3 D tissue slices, complexes based on LP10Y proved to be most efficient, by combining tissue penetration with efficient gene expression knockdown.
{"title":"Comparison of tyrosine-modified low molecular weight branched and linear polyethylenimines for siRNA delivery.","authors":"Małgorzata Kubczak, Sylwia Michlewska, Michael Karimov, Alexander Ewe, Achim Aigner, Maria Bryszewska, Maksim Ionov","doi":"10.1080/17435390.2022.2159891","DOIUrl":"https://doi.org/10.1080/17435390.2022.2159891","url":null,"abstract":"<p><p>Polyethylenimines (PEIs) have been previously introduced for siRNA delivery. In particular, in the case of higher molecular weight PEIs, this is associated with toxicity, while low molecular weight PEIs are often insufficient for siRNA complexation. The tyrosine-modification of PEIs has been shown to enhance PEI efficacy and biocompatibility. This paper evaluates a set of tyrosine-modified low molecular weight linear or branched polyethylenimines as efficient carriers of siRNA. Complexation efficacies and biophysical complex properties were analyzed by zeta potential, dynamic light scattering and circular dichroism measurements as well as gel electrophoresis. Biological knockdown was studied in 2 D cell culture and 3 D <i>ex vivo</i> tissue slice air-liquid interface culture. The results demonstrate that siRNAs were able to form stable complexes with all tested polymers. Complexation was able to protect siRNA from degradation by RNase and to mediate target gene knockdown, as determined on the mRNA level and in PC3-Luc3/EGFP and HCT116-Luc3/EGFP expressing reporter cells on the protein level, using flow cytometry and confocal microscopy. The direct comparison of the studied polymers revealed differences in biological efficacies. Moreover, the tyrosine-modified PEIs showed high biocompatibility, as determined by LDH release and mitochondria integrity (J-aggregate assay) as well as caspase 3/7 (apoptosis) and H<sub>2</sub>O<sub>2</sub> levels (ROS). In 3 D tissue slices, complexes based on LP10Y proved to be most efficient, by combining tissue penetration with efficient gene expression knockdown.</p>","PeriodicalId":18899,"journal":{"name":"Nanotoxicology","volume":"16 9-10","pages":"867-882"},"PeriodicalIF":5.0,"publicationDate":"2022-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9097650","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-08-03DOI: 10.1080/17435390.2022.2105172
Lidia Strużyńska, Beata Dąbrowska-Bouta, Grzegorz Sulkowski
The increasing production and use of silver nanoparticles (AgNPs) as an antimicrobial agent in an array of medical and commercial products, including those designed for infants and children, poses a substantial risk of exposure during the developmental period. This review summarizes current knowledge on developmental neurotoxicity of AgNPs in both pre- and post-natal stages with a focus on the biological specificity of immature organisms that predisposes them to neurotoxic insults as well as the molecular mechanisms underlying AgNP-induced neurotoxicity. The current review revealed that AgNPs increase the permeability of the blood-brain barrier (BBB) and selectively damage neurons in the brain of immature rats exposed pre and postnatally. Among the AgNP-induced molecular mechanisms underlying toxic insult is cellular stress, which can consequently lead to cell death. Glutamatergic neurons and NMDAR-mediated neurotransmission also appear to be a target for AgNPs during the postnatal period of exposure. Collected data indicate also that our current knowledge of the impact of AgNPs on the developing nervous system remains insufficient and further studies are required during different stages of development with investigation of environmentally-relevant doses of exposure.
{"title":"Developmental neurotoxicity of silver nanoparticles: the current state of knowledge and future directions.","authors":"Lidia Strużyńska, Beata Dąbrowska-Bouta, Grzegorz Sulkowski","doi":"10.1080/17435390.2022.2105172","DOIUrl":"10.1080/17435390.2022.2105172","url":null,"abstract":"<p><p>The increasing production and use of silver nanoparticles (AgNPs) as an antimicrobial agent in an array of medical and commercial products, including those designed for infants and children, poses a substantial risk of exposure during the developmental period. This review summarizes current knowledge on developmental neurotoxicity of AgNPs in both pre- and post-natal stages with a focus on the biological specificity of immature organisms that predisposes them to neurotoxic insults as well as the molecular mechanisms underlying AgNP-induced neurotoxicity. The current review revealed that AgNPs increase the permeability of the blood-brain barrier (BBB) and selectively damage neurons in the brain of immature rats exposed pre and postnatally. Among the AgNP-induced molecular mechanisms underlying toxic insult is cellular stress, which can consequently lead to cell death. Glutamatergic neurons and NMDAR-mediated neurotransmission also appear to be a target for AgNPs during the postnatal period of exposure. Collected data indicate also that our current knowledge of the impact of AgNPs on the developing nervous system remains insufficient and further studies are required during different stages of development with investigation of environmentally-relevant doses of exposure.</p>","PeriodicalId":18899,"journal":{"name":"Nanotoxicology","volume":" ","pages":"1-26"},"PeriodicalIF":5.0,"publicationDate":"2022-08-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"40666203","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-08-01DOI: 10.1080/17435390.2022.2149359
Delia Cavallo, Cinzia Lucia Ursini, Anna Maria Fresegna, Aureliano Ciervo, Fabio Boccuni, Riccardo Ferrante, Francesca Tombolini, Raffaele Maiello, Pieranna Chiarella, Giuliana Buresti, Valentina Del Frate, Diana Poli, Roberta Andreoli, Luisana Di Cristo, Stefania Sabella, Sergio Iavicoli
During nanomaterial (NM) production, workers could be exposed, particularly by inhalation, to NMs and other chemicals used in the synthesis process, so it is important to have suitable biomarkers to monitor potential toxic effects. Aim of this study was to evaluate the effectiveness of the introduction of exposure mitigation measures on workers unintentionally exposed to graphene co-pollutants during production process monitoring the presumable reduction of workplace NM contamination and of early genotoxic and oxidative effects previously found on these workers. We used Buccal Micronucleus Cytome (BMCyt) assay and Fpg-comet test, resulted the most sensitive biomarkers on our first biomonitoring work, to measure the genotoxic effects. We also detected urinary oxidized nucleic acid bases 8-oxoGua, 8-oxoGuo and 8-oxodGuo to evaluate oxidative damage. The genotoxic and oxidative effects were assessed on the same graphene workers (N = 6) previously studied, comparing the results with those found in the first biomonitoring and with the control group (N = 11). This was achieved 6 months after the installation of a special filter hood (where to perform the phases at higher risk of NM emission) and the improvement of environmental and personal protective equipment. Particle number concentration decreased after the mitigation measures. We observed reduction of Micronucleus (MN) frequency and oxidative DNA damage and increase of 8-oxodGuo excretion compared to the first biomonitoring. These results, although limited by the small subject number, showed the efficacy of adopted exposure mitigation measures and the suitability of used sensitive and noninvasive biomarkers to bio-monitor over time workers involved in graphene production process.
{"title":"A follow-up study on workers involved in the graphene production process after the introduction of exposure mitigation measures: evaluation of genotoxic and oxidative effects.","authors":"Delia Cavallo, Cinzia Lucia Ursini, Anna Maria Fresegna, Aureliano Ciervo, Fabio Boccuni, Riccardo Ferrante, Francesca Tombolini, Raffaele Maiello, Pieranna Chiarella, Giuliana Buresti, Valentina Del Frate, Diana Poli, Roberta Andreoli, Luisana Di Cristo, Stefania Sabella, Sergio Iavicoli","doi":"10.1080/17435390.2022.2149359","DOIUrl":"https://doi.org/10.1080/17435390.2022.2149359","url":null,"abstract":"<p><p>During nanomaterial (NM) production, workers could be exposed, particularly by inhalation, to NMs and other chemicals used in the synthesis process, so it is important to have suitable biomarkers to monitor potential toxic effects. Aim of this study was to evaluate the effectiveness of the introduction of exposure mitigation measures on workers unintentionally exposed to graphene co-pollutants during production process monitoring the presumable reduction of workplace NM contamination and of early genotoxic and oxidative effects previously found on these workers. We used Buccal Micronucleus Cytome (BMCyt) assay and Fpg-comet test, resulted the most sensitive biomarkers on our first biomonitoring work, to measure the genotoxic effects. We also detected urinary oxidized nucleic acid bases 8-oxoGua, 8-oxoGuo and 8-oxodGuo to evaluate oxidative damage. The genotoxic and oxidative effects were assessed on the same graphene workers (<i>N</i> = 6) previously studied, comparing the results with those found in the first biomonitoring and with the control group (<i>N</i> = 11). This was achieved 6 months after the installation of a special filter hood (where to perform the phases at higher risk of NM emission) and the improvement of environmental and personal protective equipment. Particle number concentration decreased after the mitigation measures. We observed reduction of Micronucleus (MN) frequency and oxidative DNA damage and increase of 8-oxodGuo excretion compared to the first biomonitoring. These results, although limited by the small subject number, showed the efficacy of adopted exposure mitigation measures and the suitability of used sensitive and noninvasive biomarkers to bio-monitor over time workers involved in graphene production process.</p>","PeriodicalId":18899,"journal":{"name":"Nanotoxicology","volume":"16 6-8","pages":"776-790"},"PeriodicalIF":5.0,"publicationDate":"2022-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10766666","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
We and others have previously demonstrated that exposure to nickel nanoparticles (Nano-Ni) caused fibrogenic and carcinogenic effects; however, the underlying mechanisms are still not fully understood. This study aimed to investigate the effects of Nano-Ni on epithelial-mesenchymal transition (EMT) in human bronchial epithelial cells (BEAS-2B) and its underlying mechanisms since EMT is involved in both cancer pathogenesis and tissue fibrosis. Our results showed that exposure to Nano-Ni, compared to the control Nano-TiO2, caused a remarkable decrease in the expression of E-cadherin and an increase in the expression of vimentin and α-SMA, indicating an inducible role of Nano-Ni in EMT development in human bronchial epithelial cells. HIF-1α nuclear accumulation, HDAC3 upregulation, and decreased histone acetylation were also observed in the cells exposed to Nano-Ni, but not in those exposed to Nano-TiO2. Pretreatment of the cells with a specific HIF-1α inhibitor, CAY10585, or HIF-1α-specific siRNA transfection prior to Nano-Ni exposure resulted in the restoration of E-cadherin and abolished Nano-Ni-induced upregulation of vimentin and α-SMA, suggesting a crucial role of HIF-1α in Nano-Ni-induced EMT development. CAY10585 pretreatment also attenuated the HDAC3 upregulation and increased histone acetylation. Inhibition of HDAC3 with specific siRNA significantly restrained Nano-Ni-induced reduction in histone acetylation and restored EMT-related protein expression to near control levels. In summary, our findings suggest that exposure to Nano-Ni promotes the development of EMT in human bronchial epithelial cells by decreasing histone acetylation through HIF-1α-mediated HDAC3 upregulation. Our findings may provide information for further understanding of the molecular mechanisms of Nano-Ni-induced fibrosis and carcinogenesis.
{"title":"Nickel nanoparticles induce epithelial-mesenchymal transition in human bronchial epithelial cells via the HIF-1α/HDAC3 pathway.","authors":"Jiali Yuan, Yiqun Mo, Yuanbao Zhang, Yue Zhang, Qunwei Zhang","doi":"10.1080/17435390.2022.2142169","DOIUrl":"10.1080/17435390.2022.2142169","url":null,"abstract":"<p><p>We and others have previously demonstrated that exposure to nickel nanoparticles (Nano-Ni) caused fibrogenic and carcinogenic effects; however, the underlying mechanisms are still not fully understood. This study aimed to investigate the effects of Nano-Ni on epithelial-mesenchymal transition (EMT) in human bronchial epithelial cells (BEAS-2B) and its underlying mechanisms since EMT is involved in both cancer pathogenesis and tissue fibrosis. Our results showed that exposure to Nano-Ni, compared to the control Nano-TiO<sub>2</sub>, caused a remarkable decrease in the expression of E-cadherin and an increase in the expression of vimentin and α-SMA, indicating an inducible role of Nano-Ni in EMT development in human bronchial epithelial cells. HIF-1α nuclear accumulation, HDAC3 upregulation, and decreased histone acetylation were also observed in the cells exposed to Nano-Ni, but not in those exposed to Nano-TiO<sub>2</sub>. Pretreatment of the cells with a specific HIF-1α inhibitor, CAY10585, or HIF-1α-specific siRNA transfection prior to Nano-Ni exposure resulted in the restoration of E-cadherin and abolished Nano-Ni-induced upregulation of vimentin and α-SMA, suggesting a crucial role of HIF-1α in Nano-Ni-induced EMT development. CAY10585 pretreatment also attenuated the HDAC3 upregulation and increased histone acetylation. Inhibition of HDAC3 with specific siRNA significantly restrained Nano-Ni-induced reduction in histone acetylation and restored EMT-related protein expression to near control levels. In summary, our findings suggest that exposure to Nano-Ni promotes the development of EMT in human bronchial epithelial cells by decreasing histone acetylation through HIF-1α-mediated HDAC3 upregulation. Our findings may provide information for further understanding of the molecular mechanisms of Nano-Ni-induced fibrosis and carcinogenesis.</p>","PeriodicalId":18899,"journal":{"name":"Nanotoxicology","volume":"16 6-8","pages":"695-712"},"PeriodicalIF":5.0,"publicationDate":"2022-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9892310/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10749962","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-08-01Epub Date: 2022-11-28DOI: 10.1080/17435390.2022.2144774
Yan Li, Yawen Zhu, Bosen Zhao, Qing Yao, Hailin Xu, Songqing Lv, Ji Wang, Zhiwei Sun, Yanbo Li, Caixia Guo
The adverse effects of amorphous silica nanoparticles (SiNPs) exposure on the respiratory system were increasingly recognized, however, its potential pathogenesis still remains not fully elucidated. So, this study aimed to explore its effects on pulmonary injury, and to investigate related mechanisms. Histological investigations illustrated SiNPs triggered the lung injury, mainly manifested as alveolar structure destruction, collagen deposition, and mitochondrial ultrastructural injury. In particular, SiNPs greatly enhanced pulmonary ROS and TUNEL positive rate in lungs, both of which were positively correlated with lung impairments. Further, the underlying mechanisms were investigated in cultured human bronchial epithelial cells (16HBE). Consistent with the in vivo findings, SiNPs caused the impairments on mitochondrial structure, as well as the activation of ROS generation and oxidative injury. Upon SiNPs stimuli, mitochondrial respiration was greatly inhibited, while Ca2+ overload in cytosol and mitochondria owing to ER calcium release was noticed, resulting in mitochondrial-dependent epithelial apoptosis. More importantly, mitochondrial dynamics was imbalanced toward a fission type, as evidenced by upregulated DRP1 and its phosphorylation at Ser616 (DRP1s616), while downregulated DRP1s637, and also MFN1, MFN2. Mechanistic investigations revealed that the activation of ROS/Ca2+ signaling promoted DRP1-mediated mitochondrial fission by SiNPs, forming a vicious cycle, and ultimately contributing to apoptosis in 16HBE. In summary, our results disclosed SiNPs caused pulmonary injury through the induction of epithelial apoptosis via a ROS/Ca2+/DRP1-mediated mitochondrial fission axis.
{"title":"Amorphous silica nanoparticles caused lung injury through the induction of epithelial apoptosis via ROS/Ca<sup>2+</sup>/DRP1-mediated mitochondrial fission signaling.","authors":"Yan Li, Yawen Zhu, Bosen Zhao, Qing Yao, Hailin Xu, Songqing Lv, Ji Wang, Zhiwei Sun, Yanbo Li, Caixia Guo","doi":"10.1080/17435390.2022.2144774","DOIUrl":"10.1080/17435390.2022.2144774","url":null,"abstract":"<p><p>The adverse effects of amorphous silica nanoparticles (SiNPs) exposure on the respiratory system were increasingly recognized, however, its potential pathogenesis still remains not fully elucidated. So, this study aimed to explore its effects on pulmonary injury, and to investigate related mechanisms. Histological investigations illustrated SiNPs triggered the lung injury, mainly manifested as alveolar structure destruction, collagen deposition, and mitochondrial ultrastructural injury. In particular, SiNPs greatly enhanced pulmonary ROS and TUNEL positive rate in lungs, both of which were positively correlated with lung impairments. Further, the underlying mechanisms were investigated in cultured human bronchial epithelial cells (16HBE). Consistent with the <i>in vivo</i> findings, SiNPs caused the impairments on mitochondrial structure, as well as the activation of ROS generation and oxidative injury. Upon SiNPs stimuli, mitochondrial respiration was greatly inhibited, while Ca<sup>2+</sup> overload in cytosol and mitochondria owing to ER calcium release was noticed, resulting in mitochondrial-dependent epithelial apoptosis. More importantly, mitochondrial dynamics was imbalanced toward a fission type, as evidenced by upregulated DRP1 and its phosphorylation at Ser616 (DRP1<sup>s616</sup>), while downregulated DRP1<sup>s637</sup>, and also MFN1, MFN2. Mechanistic investigations revealed that the activation of ROS/Ca<sup>2+</sup> signaling promoted DRP1-mediated mitochondrial fission by SiNPs, forming a vicious cycle, and ultimately contributing to apoptosis in 16HBE. In summary, our results disclosed SiNPs caused pulmonary injury through the induction of epithelial apoptosis <i>via</i> a ROS/Ca<sup>2+</sup>/DRP1-mediated mitochondrial fission axis.</p>","PeriodicalId":18899,"journal":{"name":"Nanotoxicology","volume":"16 6-8","pages":"713-732"},"PeriodicalIF":5.0,"publicationDate":"2022-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9309815","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-08-01DOI: 10.1080/17435390.2022.2152751
Anne Thoustrup Saber, Niels Hadrup, Andrew Williams, Alicja Mortensen, Jozef Szarek, Zdenka Kyjovska, Alexander Kurz, Nicklas Raun Jacobsen, Håkan Wallin, Sabina Halappanavar, Ulla Vogel
The inclusion of nanoparticles can increase the quality of certain products. One application is the inclusion of Zinc oxide (ZnO) nanoparticles in a glass coating matrix to produce a UV-absorbing coating for glass sheets. Yet, the question is whether the inclusion of ZnO in the matrix induces toxicity at low exposure levels. To test this, mice were given single intratracheal instillation of 1) ZnO powder (ZnO), 2) ZnO in a glass matrix coating in its liquid phase (ZnO-Matrix), and 3) the matrix with no ZnO (Matrix). Doses of ZnO were 0.23, 0.67, and 2 µg ZnO/mouse. ZnO Matrix doses had equal amounts of ZnO, while Matrix was adjusted to have an equal volume of matrix as ZnO Matrix. Post-exposure periods were 1, 3, or 28 d. Endpoints were pulmonary inflammation as bronchoalveolar lavage (BAL) fluid cellularity, genotoxicity in lung and liver, measured by comet assay, histopathology of lung and liver, and global gene expression in lung using microarrays. Neutrophil numbers were increased to a similar extent with ZnO and ZnO-Matrix at 1 and 3 d. Only weak genotoxicity without dose-response effects was observed in the lung. Lung histology showed an earlier onset of inflammation in material-exposed groups as compared to controls. Microarray analysis showed a stronger response in terms of the number of differentially regulated genes in ZnO-Matrix exposed mice as compared to Matrix only. Activated canonical pathways included inflammatory and cardiovascular ones. In conclusion, the pulmonary toxicity of ZnO was not changed by formulation in a liquid matrix for glass coating.
{"title":"Unchanged pulmonary toxicity of ZnO nanoparticles formulated in a liquid matrix for glass coating.","authors":"Anne Thoustrup Saber, Niels Hadrup, Andrew Williams, Alicja Mortensen, Jozef Szarek, Zdenka Kyjovska, Alexander Kurz, Nicklas Raun Jacobsen, Håkan Wallin, Sabina Halappanavar, Ulla Vogel","doi":"10.1080/17435390.2022.2152751","DOIUrl":"https://doi.org/10.1080/17435390.2022.2152751","url":null,"abstract":"<p><p>The inclusion of nanoparticles can increase the quality of certain products. One application is the inclusion of Zinc oxide (ZnO) nanoparticles in a glass coating matrix to produce a UV-absorbing coating for glass sheets. Yet, the question is whether the inclusion of ZnO in the matrix induces toxicity at low exposure levels. To test this, mice were given single intratracheal instillation of 1) ZnO powder (ZnO), 2) ZnO in a glass matrix coating in its liquid phase (ZnO-Matrix), and 3) the matrix with no ZnO (Matrix). Doses of ZnO were 0.23, 0.67, and 2 µg ZnO/mouse. ZnO Matrix doses had equal amounts of ZnO, while Matrix was adjusted to have an equal volume of matrix as ZnO Matrix. Post-exposure periods were 1, 3, or 28 d. Endpoints were pulmonary inflammation as bronchoalveolar lavage (BAL) fluid cellularity, genotoxicity in lung and liver, measured by comet assay, histopathology of lung and liver, and global gene expression in lung using microarrays. Neutrophil numbers were increased to a similar extent with ZnO and ZnO-Matrix at 1 and 3 d. Only weak genotoxicity without dose-response effects was observed in the lung. Lung histology showed an earlier onset of inflammation in material-exposed groups as compared to controls. Microarray analysis showed a stronger response in terms of the number of differentially regulated genes in ZnO-Matrix exposed mice as compared to Matrix only. Activated canonical pathways included inflammatory and cardiovascular ones. In conclusion, the pulmonary toxicity of ZnO was not changed by formulation in a liquid matrix for glass coating.</p>","PeriodicalId":18899,"journal":{"name":"Nanotoxicology","volume":"16 6-8","pages":"812-827"},"PeriodicalIF":5.0,"publicationDate":"2022-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10750484","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-08-01DOI: 10.1080/17435390.2022.2144775
Nikita Dinger, Valeria Panzetta, Carmela Russo, Paolo Antonio Netti, Mariano Sirignano
Carbon nanomaterials are an inventive class of materials with wide applications in state-of-the-art bioimaging and therapeutics. They allow a broad range of tunable and integrated advantages of structural flexibility, chemical and thermal stability, upright electrical conductivity, and the option of scale-up and mass production. In the context of nanomedicine, carbon nanomaterials have been used extensively to mitigate the serious side effects of conventional chemotherapy and also to enable early cancer diagnostics, given their wide range of tunable properties. A class of carbon nanomaterials, called carbon dots (CDs) are small carbon-based nanoparticles and have been a valued discovery due to their photoluminescence, low photobleaching, and high surface area to mass ratio. The process of producing these CDs had so far been a high energy demanding process involving wet chemistry for purification. A one-step tunable production of luminescent CDs from fuel rich combustion reactors was recently presented by our group. In this paper, we explore the effects of these yellow luminescent combustion-generated CDs in MCF7 adenocarcinoma and MCF10a normal breast epithelial cells. We observed that these CDs, also at nontoxic doses, can affect basic cellular functions, such as cell cycle and proliferation; induce substantial changes on the physical parameters of the plasma membrane; and change the overall appearance of a cell in terms of morphology.
{"title":"<i>In vitro</i> effects of combustion generated carbon dots on cellular parameters in healthy and cancerous breast cells.","authors":"Nikita Dinger, Valeria Panzetta, Carmela Russo, Paolo Antonio Netti, Mariano Sirignano","doi":"10.1080/17435390.2022.2144775","DOIUrl":"https://doi.org/10.1080/17435390.2022.2144775","url":null,"abstract":"<p><p>Carbon nanomaterials are an inventive class of materials with wide applications in state-of-the-art bioimaging and therapeutics. They allow a broad range of tunable and integrated advantages of structural flexibility, chemical and thermal stability, upright electrical conductivity, and the option of scale-up and mass production. In the context of nanomedicine, carbon nanomaterials have been used extensively to mitigate the serious side effects of conventional chemotherapy and also to enable early cancer diagnostics, given their wide range of tunable properties. A class of carbon nanomaterials, called carbon dots (CDs) are small carbon-based nanoparticles and have been a valued discovery due to their photoluminescence, low photobleaching, and high surface area to mass ratio. The process of producing these CDs had so far been a high energy demanding process involving wet chemistry for purification. A one-step tunable production of luminescent CDs from fuel rich combustion reactors was recently presented by our group. In this paper, we explore the effects of these yellow luminescent combustion-generated CDs in MCF7 adenocarcinoma and MCF10a normal breast epithelial cells. We observed that these CDs, also at nontoxic doses, can affect basic cellular functions, such as cell cycle and proliferation; induce substantial changes on the physical parameters of the plasma membrane; and change the overall appearance of a cell in terms of morphology.</p>","PeriodicalId":18899,"journal":{"name":"Nanotoxicology","volume":"16 6-8","pages":"733-756"},"PeriodicalIF":5.0,"publicationDate":"2022-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10757312","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-08-01DOI: 10.1080/17435390.2022.2149360
Kinga Malinowska, Bożena Bukowska, Ireneusz Piwoński, Marek Foksiński, Aneta Kisielewska, Ewelina Zarakowska, Daniel Gackowski, Paulina Sicińska
Plastic nanoparticles are widely spread in the biosphere, but health risk associated with their effect on the human organism has not yet been assessed. The purpose of this study was to determine the genotoxic potential of non-functionalized polystyrene nanoparticles (PS-NPs) of different diameters of 29, 44, and 72 nm in human peripheral blood mononuclear cells (PBMCs) (in vitro). To select non-cytotoxic concentrations of tested PS-NPs, we analyzed metabolic activity of PBMCs incubated with these particles in concentrations ranging from 0.001 to 1000 µg/mL. Then, PS-NPs were used in concentrations from 0.0001 to 100 μg/mL and incubated with tested cells for 24 h. Physico-chemical properties of PS-NPs in media and suspension were analyzed using dynamic light scattering (DLS), atomic force microscopy (AFM), scanning electron microscopy (SEM) and zeta potential. For the first time, we investigated the mechanism of genotoxic action of PS-NPs based on detection of single/double DNA strand-breaks and 8-oxo-2'-deoxyguanosine (8-oxodG) formation, as well as determination of oxidative modification of purines and pyrimidines and repair efficiency of DNA damage. Obtained results have shown that PS-NPs caused a decrease in PBMCs metabolic activity, increased single/double-strand break formation, oxidized purines and pyrimidines and increased 8oxodG levels. The resulting damage was completely repaired in the case of the largest PS-NPs. It was also found that extent of genotoxic changes in PBMCs depended on the size of tested particles and their ζ-potential value.
{"title":"Polystyrene nanoparticles: the mechanism of their genotoxicity in human peripheral blood mononuclear cells.","authors":"Kinga Malinowska, Bożena Bukowska, Ireneusz Piwoński, Marek Foksiński, Aneta Kisielewska, Ewelina Zarakowska, Daniel Gackowski, Paulina Sicińska","doi":"10.1080/17435390.2022.2149360","DOIUrl":"https://doi.org/10.1080/17435390.2022.2149360","url":null,"abstract":"<p><p>Plastic nanoparticles are widely spread in the biosphere, but health risk associated with their effect on the human organism has not yet been assessed. The purpose of this study was to determine the genotoxic potential of non-functionalized polystyrene nanoparticles (PS-NPs) of different diameters of 29, 44, and 72 nm in human peripheral blood mononuclear cells (PBMCs) (<i>in vitro</i>). To select non-cytotoxic concentrations of tested PS-NPs, we analyzed metabolic activity of PBMCs incubated with these particles in concentrations ranging from 0.001 to 1000 µg/mL. Then, PS-NPs were used in concentrations from 0.0001 to 100 μg/mL and incubated with tested cells for 24 h. Physico-chemical properties of PS-NPs in media and suspension were analyzed using dynamic light scattering (DLS), atomic force microscopy (AFM), scanning electron microscopy (SEM) and zeta potential. For the first time, we investigated the mechanism of genotoxic action of PS-NPs based on detection of single/double DNA strand-breaks and 8-oxo-2'-deoxyguanosine (8-oxodG) formation, as well as determination of oxidative modification of purines and pyrimidines and repair efficiency of DNA damage. Obtained results have shown that PS-NPs caused a decrease in PBMCs metabolic activity, increased single/double-strand break formation, oxidized purines and pyrimidines and increased 8oxodG levels. The resulting damage was completely repaired in the case of the largest PS-NPs. It was also found that extent of genotoxic changes in PBMCs depended on the size of tested particles and their ζ-potential value.</p>","PeriodicalId":18899,"journal":{"name":"Nanotoxicology","volume":"16 6-8","pages":"791-811"},"PeriodicalIF":5.0,"publicationDate":"2022-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10766665","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2022-08-01DOI: 10.1080/17435390.2022.2147460
Tao Wu, Shengmin Xu, Biao Chen, Lingzhi Bao, Jie Ma, Wei Han, An Xu, Kwan Ngok Yu, Lijun Wu, Shaopeng Chen
Ambient PM2.5 is one of the environmental risk factors and was correlated with senescence-related diseases based on the epidemiologic investigation. However, little is known about senescence induced by PM2.5 as well as the underlying mechanisms. In this study, we demonstrated that PM2.5 exposure aggravated cellular senescence in vivo and in vitro, and disrupted micronuclei (MN) played a vital role in this process. Our results suggested that the nuclear envelope (NE) of PM2.5-induced MN was ruptured. Subsequently, cGAS was found to localize to approximately 80% of the disrupted MN but few for intact MN. Upon examination of cGAMP and SA-β-Gal, the cGAS-STING pathway was found activated and related to cellular senescence induced by PM2.5. Taken together, we reported a novel finding that PM2.5 exposure causes cellular senescence via DNA damage, MN formation, and cGAS activation. These results revealed the potential toxicity of PM2.5 and its related mechanisms in cellular senescence.
{"title":"Ambient PM2.5 exposure causes cellular senescence via DNA damage, micronuclei formation, and cGAS activation.","authors":"Tao Wu, Shengmin Xu, Biao Chen, Lingzhi Bao, Jie Ma, Wei Han, An Xu, Kwan Ngok Yu, Lijun Wu, Shaopeng Chen","doi":"10.1080/17435390.2022.2147460","DOIUrl":"https://doi.org/10.1080/17435390.2022.2147460","url":null,"abstract":"<p><p>Ambient PM2.5 is one of the environmental risk factors and was correlated with senescence-related diseases based on the epidemiologic investigation. However, little is known about senescence induced by PM2.5 as well as the underlying mechanisms. In this study, we demonstrated that PM2.5 exposure aggravated cellular senescence <i>in vivo</i> and <i>in vitro</i>, and disrupted micronuclei (MN) played a vital role in this process. Our results suggested that the nuclear envelope (NE) of PM2.5-induced MN was ruptured. Subsequently, cGAS was found to localize to approximately 80% of the disrupted MN but few for intact MN. Upon examination of cGAMP and SA-β-Gal, the cGAS-STING pathway was found activated and related to cellular senescence induced by PM2.5. Taken together, we reported a novel finding that PM2.5 exposure causes cellular senescence <i>via</i> DNA damage, MN formation, and cGAS activation. These results revealed the potential toxicity of PM2.5 and its related mechanisms in cellular senescence.</p>","PeriodicalId":18899,"journal":{"name":"Nanotoxicology","volume":"16 6-8","pages":"757-775"},"PeriodicalIF":5.0,"publicationDate":"2022-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10757313","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}