1
|
Kaur J, Sharma A, Passi G, Dey P, Khajuria A, Alajangi HK, Jaiswal PK, Barnwal RP, Singh G. Nanomedicine at the Pulmonary Frontier: Immune-Centric Approaches for Respiratory Disease Treatment. Immunol Invest 2024; 53:295-347. [PMID: 38206610 DOI: 10.1080/08820139.2023.2298398] [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: 01/12/2024]
Abstract
Respiratory diseases (RD) are a group of common ailments with a rapidly increasing global prevalence, posing a significant threat to humanity, especially the elderly population, and imposing a substantial burden on society and the economy. RD represents an unmet medical need that requires the development of viable pharmacotherapies. While various promising strategies have been devised to advance potential treatments for RD, their implementation has been hindered by difficulties in drug delivery, particularly in critically ill patients. Nanotechnology offers innovative solutions for delivering medications to the inflamed organ sites, such as the lungs. Although this approach is enticing, delivering nanomedicine to the lungs presents complex challenges that require sophisticated techniques. In this context, we review the potential of novel nanomedicine-based immunomodulatory strategies that could offer therapeutic benefits in managing this pressing health condition.
Collapse
Affiliation(s)
- Jatinder Kaur
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Akanksha Sharma
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
- Department of Biophysics, Panjab University, Chandigarh, India
| | - Gautam Passi
- Department of Biophysics, Panjab University, Chandigarh, India
| | - Piyush Dey
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
- Department of Biophysics, Panjab University, Chandigarh, India
| | - Akhil Khajuria
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Hema Kumari Alajangi
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
- Department of Biophysics, Panjab University, Chandigarh, India
| | - Pradeep Kumar Jaiswal
- Department of Biochemistry and Biophysics, Texas A & M University, College Station, Texas, USA
| | | | - Gurpal Singh
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| |
Collapse
|
2
|
Zhang Y, Mo Y, Zhang Y, Yuan J, Zhang Q. MMP-3-mediated cleavage of OPN is involved in copper oxide nanoparticle-induced activation of fibroblasts. Part Fibre Toxicol 2023; 20:22. [PMID: 37217992 PMCID: PMC10201731 DOI: 10.1186/s12989-023-00532-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 05/08/2023] [Indexed: 05/24/2023] Open
Abstract
BACKGROUND Copper oxide nanoparticles (Nano-CuO) are one of the most produced and used nanomaterials. Previous studies have shown that exposure to Nano-CuO caused acute lung injury, inflammation, and fibrosis. However, the mechanisms underlying Nano-CuO-induced lung fibrosis are still unclear. Here, we hypothesized that exposure of human lung epithelial cells and macrophages to Nano-CuO would upregulate MMP-3, which cleaved osteopontin (OPN), resulting in fibroblast activation and lung fibrosis. METHODS A triple co-culture model was established to explore the mechanisms underlying Nano-CuO-induced fibroblast activation. Cytotoxicity of Nano-CuO on BEAS-2B, U937* macrophages, and MRC-5 fibroblasts were determined by alamarBlue and MTS assays. The expression or activity of MMP-3, OPN, and fibrosis-associated proteins was determined by Western blot or zymography assay. Migration of MRC-5 fibroblasts was evaluated by wound healing assay. MMP-3 siRNA and an RGD-containing peptide, GRGDSP, were used to explore the role of MMP-3 and cleaved OPN in fibroblast activation. RESULTS Exposure to non-cytotoxic doses of Nano-CuO (0.5 and 1 µg/mL) caused increased expression and activity of MMP-3 in the conditioned media of BEAS-2B and U937* cells, but not MRC-5 fibroblasts. Nano-CuO exposure also caused increased production of cleaved OPN fragments, which was abolished by MMP-3 siRNA transfection. Conditioned media from Nano-CuO-exposed BEAS-2B, U937*, or the co-culture of BEAS-2B and U937* caused activation of unexposed MRC-5 fibroblasts. However, direct exposure of MRC-5 fibroblasts to Nano-CuO did not induce their activation. In a triple co-culture system, exposure of BEAS-2B and U937* cells to Nano-CuO caused activation of unexposed MRC-5 fibroblasts, while transfection of MMP-3 siRNA in BEAS-2B and U937* cells significantly inhibited the activation and migration of MRC-5 fibroblasts. In addition, pretreatment with GRGDSP peptide inhibited Nano-CuO-induced activation and migration of MRC-5 fibroblasts in the triple co-culture system. CONCLUSIONS Our results demonstrated that Nano-CuO exposure caused increased production of MMP-3 from lung epithelial BEAS-2B cells and U937* macrophages, which cleaved OPN, resulting in the activation of lung fibroblasts MRC-5. These results suggest that MMP-3-cleaved OPN may play a key role in Nano-CuO-induced activation of lung fibroblasts. More investigations are needed to confirm whether these effects are due to the nanoparticles themselves and/or Cu ions.
Collapse
Affiliation(s)
- Yuanbao Zhang
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, 485 E. Gray Street, Louisville, KY 40202 USA
| | - Yiqun Mo
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, 485 E. Gray Street, Louisville, KY 40202 USA
| | - Yue Zhang
- Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Jiali Yuan
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, 485 E. Gray Street, Louisville, KY 40202 USA
| | - Qunwei Zhang
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, 485 E. Gray Street, Louisville, KY 40202 USA
| |
Collapse
|
3
|
Zhang G, Luo W, Yang W, Li S, Li D, Zeng Y, Li Y. The importance of the
IL
‐1 family of cytokines in nanoimmunosafety and nanotoxicology. WIRES NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1850. [DOI: 10.1002/wnan.1850] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 08/03/2022] [Accepted: 08/11/2022] [Indexed: 11/24/2022]
Affiliation(s)
- Guofang Zhang
- Laboratory of Immunology and Nanomedicine Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences Shenzhen China
| | - Wenhe Luo
- Laboratory of Immunology and Nanomedicine Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences Shenzhen China
| | - Wenjie Yang
- Laboratory of Immunology and Nanomedicine Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences Shenzhen China
| | - Su Li
- Laboratory of Immunology and Nanomedicine Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences Shenzhen China
| | - Dongjie Li
- Laboratory of Immunology and Nanomedicine Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences Shenzhen China
| | - Yanqiao Zeng
- Laboratory of Immunology and Nanomedicine Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences Shenzhen China
| | - Yang Li
- Laboratory of Immunology and Nanomedicine Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences Shenzhen China
| |
Collapse
|
4
|
Abstract
Background Enthusiasm for the use of metal nanoparticles in human and veterinary medicine is high. Many articles describe the effects of metal nanoparticles on microbes in vitro, and a smaller number of articles describe effects on the immune system, which is the focus of this review. Methods Articles were retrieved by performing literature searches in Medline, of the National Institute of Medicine, as well as via Google Scholar. Results In vitro studies show that metal nanoparticles have antimicrobial effects. Some metal nanoparticles augment innate host immune defenses, such as endogenous antimicrobial peptides, and nitric oxide. Metal nanoparticles may also function as vaccine adjuvants. Metal nanoparticles can migrate to locations distant from the site of administration, however, requiring careful monitoring for toxicity. Conclusions Metal nanoparticles show a great deal of potential as immunomodulators, as well as direct antimicrobial effects. Before metal particles can be adopted as therapies; however, more studies are needed to determine how nanoparticles migrate though the body and on possible adverse effects.
Collapse
Affiliation(s)
- John K Crane
- Division of Infectious Diseases, University at Buffalo , Buffalo, New York, USA
| |
Collapse
|
5
|
Cui L, Wang X, Sun B, Xia T, Hu S. Predictive Metabolomic Signatures for Safety Assessment of Metal Oxide Nanoparticles. ACS NANO 2019; 13:13065-13082. [PMID: 31682760 DOI: 10.1021/acsnano.9b05793] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
The widespread use of metal oxide nanoparticles (MOx NPs) poses a risk of exposure that may lead to adverse health effects on humans. Even though a number of toxicological methodologies are available for assessing nanotoxicity, the effect of MOx NPs on cell metabolism in vitro and in vivo remains largely unknown, especially under the exposure to low-dose or supposedly low-toxicity MOx NPs. In this study, liquid chromatography-mass spectrometry (LC-MS) based metabolomics was used to reveal significantly altered metabolites and metabolic pathways in human bronchial epithelial cells exposed to four different types of MOx NPs (ZnO, SiO2, TiO2, and CeO2) at both high (25 μg/mL) and low (12.5 μg/mL) doses. We demonstrated that high-dose ZnO NPs caused severe cytotoxicity with altered metabolism of amino acids, nucleotides, nucleosides, tricarboxylic acid cycle, lipids, inflammation/redox, and fatty acid oxidation, as well as the elevation of toxic and DNA damage related metabolites. Fewer metabolomic alterations were induced by low-dose ZnO NPs. However, most metabolites significantly altered by high-dose ZnO NPs were also slightly changed by low-dose ZnO NPs. On the other hand, the cells exposed to SiO2, TiO2, and CeO2 NPs at either high or low dose displayed low cytotoxicity with similar metabolomic alterations, although each type of NPs induced distinct changes of certain metabolites. These three NPs significantly affected the metabolic pathways of sphingosine-1-phosphate, fatty acid oxidation, folate cycle, inflammation/redox, and lipid metabolism. In addition, dose-dependent effects were observed for a number of metabolites significantly altered by respective MOx NPs. Representative metabolites of the significantly altered metabolic pathways were successfully validated in vitro using enzymatic assays. More importantly, these representative metabolites were further validated in a mouse model after lung exposure to respective NPs, indicating that in vitro metabolomic findings may be used to effectively predict the toxicological effects in vivo. Despite functional assay results demonstrating that the changes in cellular functions were largely reflected by the metabolomic alterations, LC-MS-based metabolomics was sensitive enough to detect the subtle metabolomic changes when functional cellular assays showed no significant difference. Collectively, our studies have unveiled potential metabolic mechanisms of MOx NP-induced nanotoxicity in lung epithelial cells and demonstrated the sensitivity and feasibility of using metabolomic signatures to understand and predict nanotoxicity in vivo.
Collapse
Affiliation(s)
- Li Cui
- School of Dentistry and Jonsson Comprehensive Cancer Center , University of California , Los Angeles , California 90095 , United States
| | - Xiang Wang
- Division of NanoMedicine, Department of Medicine, California NanoSystems Institute , University of California , Los Angeles , California 90095 , United States
| | - Bingbing Sun
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering , Dalian University of Technology , 2 Linggong Road , 116024 , Dalian , China
| | - Tian Xia
- Division of NanoMedicine, Department of Medicine, California NanoSystems Institute , University of California , Los Angeles , California 90095 , United States
| | - Shen Hu
- School of Dentistry and Jonsson Comprehensive Cancer Center , University of California , Los Angeles , California 90095 , United States
| |
Collapse
|
6
|
Ihrie MD, Taylor-Just AJ, Walker NJ, Stout MD, Gupta A, Richey JS, Hayden BK, Baker GL, Sparrow BR, Duke KS, Bonner JC. Inhalation exposure to multi-walled carbon nanotubes alters the pulmonary allergic response of mice to house dust mite allergen. Inhal Toxicol 2019; 31:192-202. [PMID: 31345048 DOI: 10.1080/08958378.2019.1643955] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background: Increasing evidence from rodent studies indicates that inhaled multi-walled carbon nanotubes (MWCNTs) have harmful effects on the lungs. In this study, we examined the effects of inhalation exposure to MWCNTs on allergen-induced airway inflammation and fibrosis. We hypothesized that inhalation pre-exposure to MWCNTs would render mice susceptible to developing allergic lung disease induced by house dust mite (HDM) allergen. Methods: Male B6C3F1/N mice were exposed by whole-body inhalation for 6 h a day, 5 d a week, for 30 d to air control or 0.06, 0.2, and 0.6 mg/m3 of MWCNTs. The exposure atmospheres were agglomerates (1.4-1.8 µm) composed of MWCNTs (average diameter 16 nm; average length 2.4 µm; 0.52% Ni). Mice then received 25 µg of HDM extract by intranasal instillation 6 times over 3 weeks. Necropsy was performed at 3 and 30 d after the final HDM dose to collect serum, bronchoalveolar lavage fluid (BALF), and lung tissue for histopathology. Results: MWCNT exposure at the highest dose inhibited HDM-induced serum IgE levels, IL-13 protein levels in BALF, and airway mucus production. However, perivascular and peribronchiolar inflammatory lesions were observed in the lungs of mice at 3 d with MWCNT and HDM, but not MWCNT or HDM alone. Moreover, combined HDM and MWCNT exposure increased airway fibrosis in the lungs of mice. Conclusions: Inhalation pre-exposure to MWCNTs inhibited HDM-induced TH2 immune responses, yet this combined exposure resulted in vascular inflammation and airway fibrosis, indicating that MWCNT pre-exposure alters the immune response to allergens.
Collapse
Affiliation(s)
- Mark D Ihrie
- a Department of Biological Sciences, North Carolina State University , Raleigh , NC , USA
| | - Alexia J Taylor-Just
- a Department of Biological Sciences, North Carolina State University , Raleigh , NC , USA
| | - Nigel J Walker
- b National Institute of Environmental Health Sciences , Durham , NC , USA
| | - Matthew D Stout
- b National Institute of Environmental Health Sciences , Durham , NC , USA
| | - Amit Gupta
- c Battelle Biomedical Research Centre , Columbus , OH , USA
| | - Jamie S Richey
- c Battelle Biomedical Research Centre , Columbus , OH , USA
| | - Barry K Hayden
- c Battelle Biomedical Research Centre , Columbus , OH , USA
| | | | | | - Katherine S Duke
- a Department of Biological Sciences, North Carolina State University , Raleigh , NC , USA
| | - James C Bonner
- a Department of Biological Sciences, North Carolina State University , Raleigh , NC , USA
| |
Collapse
|
7
|
Abukabda AB, McBride CR, Batchelor TP, Goldsmith WT, Bowdridge EC, Garner KL, Friend S, Nurkiewicz TR. Group II innate lymphoid cells and microvascular dysfunction from pulmonary titanium dioxide nanoparticle exposure. Part Fibre Toxicol 2018; 15:43. [PMID: 30413212 PMCID: PMC6230229 DOI: 10.1186/s12989-018-0280-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 10/24/2018] [Indexed: 01/16/2023] Open
Abstract
Background The cardiovascular effects of pulmonary exposure to engineered nanomaterials (ENM) are poorly understood, and the reproductive consequences are even less understood. Inflammation remains the most frequently explored mechanism of ENM toxicity. However, the key mediators and steps between lung exposure and uterine health remain to be fully defined. The purpose of this study was to determine the uterine inflammatory and vascular effects of pulmonary exposure to titanium dioxide nanoparticles (nano-TiO2). We hypothesized that pulmonary nano-TiO2 exposure initiates a Th2 inflammatory response mediated by Group II innate lymphoid cells (ILC2), which may be associated with an impairment in uterine microvascular reactivity. Methods Female, virgin, Sprague-Dawley rats (8–12 weeks) were exposed to 100 μg of nano-TiO2 via intratracheal instillation 24 h prior to microvascular assessments. Serial blood samples were obtained at 0, 1, 2 and 4 h post-exposure for multiplex cytokine analysis. ILC2 numbers in the lungs were determined. ILC2s were isolated and phosphorylated nuclear factor kappa-light-chain-enhancer of activated B cells (NF-ĸB) levels were measured. Pressure myography was used to assess vascular reactivity of isolated radial arterioles. Results Pulmonary nano-TiO2 exposure was associated with an increase in IL-1ß, 4, 5 and 13 and TNF- α 4 h post-exposure, indicative of an innate Th2 inflammatory response. ILC2 numbers were significantly increased in lungs from exposed animals (1.66 ± 0.19%) compared to controls (0.19 ± 0.22%). Phosphorylation of the transactivation domain (Ser-468) of NF-κB in isolated ILC2 and IL-33 in lung epithelial cells were significantly increased (126.8 ± 4.3% and 137 ± 11% of controls respectively) by nano-TiO2 exposure. Lastly, radial endothelium-dependent arteriolar reactivity was significantly impaired (27 ± 12%), while endothelium-independent dilation (7 ± 14%) and α-adrenergic sensitivity (8 ± 2%) were not altered compared to control levels. Treatment with an anti- IL-33 antibody (1 mg/kg) 30 min prior to nano-TiO2 exposure resulted in a significant improvement in endothelium-dependent dilation and a decreased level of IL-33 in both plasma and bronchoalveolar lavage fluid. Conclusions These results provide evidence that the uterine microvascular dysfunction that follows pulmonary ENM exposure may be initiated via activation of lung-resident ILC2 and subsequent systemic Th2-dependent inflammation. Electronic supplementary material The online version of this article (10.1186/s12989-018-0280-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alaeddin Bashir Abukabda
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center - West Virginia University, Morgantown, WV, 26505-9229, USA.,Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Carroll Rolland McBride
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center - West Virginia University, Morgantown, WV, 26505-9229, USA.,Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Thomas Paul Batchelor
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center - West Virginia University, Morgantown, WV, 26505-9229, USA.,Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - William Travis Goldsmith
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center - West Virginia University, Morgantown, WV, 26505-9229, USA.,Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Elizabeth Compton Bowdridge
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center - West Virginia University, Morgantown, WV, 26505-9229, USA.,Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Krista Lee Garner
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center - West Virginia University, Morgantown, WV, 26505-9229, USA.,Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Sherri Friend
- National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Timothy Robert Nurkiewicz
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center - West Virginia University, Morgantown, WV, 26505-9229, USA. .,Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA. .,National Institute for Occupational Safety and Health, Morgantown, WV, USA.
| |
Collapse
|
8
|
Dobrovolskaia MA, Shurin MR, Kagan VE, Shvedova AA. Ins and Outs in Environmental and Occupational Safety Studies of Asthma and Engineered Nanomaterials. ACS NANO 2017; 11:7565-7571. [PMID: 28737932 PMCID: PMC6481664 DOI: 10.1021/acsnano.7b04916] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
According to the Centers for Disease Control and Prevention, approximately 25 million Americans suffer from asthma. The disease total annual cost is about $56 billion and includes both the direct and indirect costs of medications, hospital stays, missed work, and decreased productivity. Air pollution with xenobiotics, bacterial agents, and industrial nanomaterials, such as carbon nanotubes, contribute to the exacerbation of this condition and are a point of particular attention in environmental toxicology as well as in occupational health and safety research. Mast cell degranulation and activation of Th2 cells triggered either by allergen-specific immunoglobulin E (IgE) or by alternative mechanisms, such as locally produced neurotransmitters, underlie the pathophysiological process of airway constriction during an asthma attack. Other immune and non-immune cell types, including basophils, eosinophils, Th1, Th17, Th9, macrophages, dendritic cells, and smooth muscle cells, are involved in the inflammatory and allergic responses during asthma, which, under chronic conditions, may progress without mast cells, the key trigger of the acute asthma attack. To decipher complex molecular, cellular, and genetic mechanisms, many researchers have attempted to develop in vitro and in vivo models to study asthma. Herein, we summarize the advantages and disadvantages of various models and their applicability to nanoparticle evaluation in asthma research. We further suggest that a framework for both in vitro and in vivo methods should be used to study the impact of engineered nanomaterials on asthma etiology, pathophysiology, and treatment.
Collapse
Affiliation(s)
- Marina A. Dobrovolskaia
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, NCI at Frederick, Frederick, MD 21702, USA
| | - Michael R. Shurin
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Department of Immunology, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Valerian E. Kagan
- Departments of Environmental and Occupational Health, Pharmacology and Chemical Biology, Chemistry and Radiation Oncology and Center for Free and Antioxidant Health, University of Pittsburgh
| | - Anna A. Shvedova
- Health Effects Laboratory Division, National Institute of Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV 26505, USA
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV 26506, USA
| |
Collapse
|
9
|
Abstract
In recent years there has been considerable effort to understand the interaction of nanomaterials with the skin. In this study we use an in vivo mouse model of allergic contact dermatitis to investigate how nanoparticles (NPs) may alter allergic responses in skin. We investigate a variety of NPs that vary in size, charge and composition. Results show that small (<200 nm) negative and neutral charged NPs exhibit an immunosuppressive effect but that positively charged NPs do not. Confocal imaging suggests positively charged NPs may penetrate skin to a lesser extent and thereby are less able interact with and alter the local immune responses. Interestingly, negatively charged silica (20 nm) NPs suppress allergic response to two chemically distinct sensitizers; 1-fluoro-2, 4-dinitrobenzene and 2-deoxyurushiol. Skin wiping and NP application time studies suggest that the immunomodulatory mechanism is not due solely to the blocking of sensitizer adduct formation in skin. Results suggest that NPs modulate early immune events that impact mast cell degranulation. Our study shows for the first time the potential to modulate the elicitation phase of the allergic response which depends on the NP charge and composition. These finding can be used to inform the design topical therapeutics to mitigate allergic responses in skin.
Collapse
Affiliation(s)
- Samreen Jatana
- Department of Biomedical Engineering, University of Rochester, Rochester, New York, USA
| | - Brian C Palmer
- Department of Toxicology, University of Rochester Medical Center, New York, USA
| | - Sarah J Phelan
- Department of Toxicology, University of Rochester Medical Center, New York, USA
| | - Lisa A DeLouise
- Department of Biomedical Engineering, University of Rochester, Rochester, New York, USA. .,Department of Dermatology, University of Rochester Medical Center, Rochester, New York, USA.
| |
Collapse
|
10
|
Neagu M, Piperigkou Z, Karamanou K, Engin AB, Docea AO, Constantin C, Negrei C, Nikitovic D, Tsatsakis A. Protein bio-corona: critical issue in immune nanotoxicology. Arch Toxicol 2017; 91:1031-1048. [PMID: 27438349 PMCID: PMC5316397 DOI: 10.1007/s00204-016-1797-5] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Accepted: 07/06/2016] [Indexed: 01/04/2023]
Abstract
With the expansion of the nanomedicine field, the knowledge focusing on the behavior of nanoparticles in the biological milieu has rapidly escalated. Upon introduction to a complex biological system, nanomaterials dynamically interact with all the encountered biomolecules and form the protein "bio-corona." The decoration with these surface biomolecules endows nanoparticles with new properties. The present review will address updates of the protein bio-corona characteristics as influenced by nanoparticle's physicochemical properties and by the particularities of the encountered biological milieu. Undeniably, bio-corona generation influences the efficacy of the nanodrug and guides the actions of innate and adaptive immunity. Exploiting the dynamic process of protein bio-corona development in combination with the new engineered horizons of drugs linked to nanoparticles could lead to innovative functional nanotherapies. Therefore, bio-medical nanotechnologies should focus on the interactions of nanoparticles with the immune system for both safety and efficacy reasons.
Collapse
Affiliation(s)
- Monica Neagu
- Immunology Department, "Victor Babes" National Institute of Pathology, Bucharest, Romania
- Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Zoi Piperigkou
- Laboratory of Biochemistry, Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Department of Chemistry, University of Patras, Patras, Greece
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Konstantina Karamanou
- Laboratory of Biochemistry, Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Department of Chemistry, University of Patras, Patras, Greece
- Laboratório de Bioquímica e Biologia Cellular de Glicoconjugados, Programa de Glicobiologia, Instituto de Bioquímica Médica Leopoldo De Meis and Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ayse Basak Engin
- Department of Toxicology, Faculty of Pharmacy, Gazi University, Ankara, Turkey
| | - Anca Oana Docea
- Department of Toxicology, Faculty of Pharmacy University of Medicine and Pharmacy Craiova, Craiova, Romania
| | - Carolina Constantin
- Immunology Department, "Victor Babes" National Institute of Pathology, Bucharest, Romania
| | - Carolina Negrei
- Department of Toxicology, Faculty of Pharmacy, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
| | - Dragana Nikitovic
- Laboratory of Anatomy-Histology-Embryology, Medical School, University of Crete, Heraklion, Greece
| | - Aristidis Tsatsakis
- Department of Toxicology and Forensic Sciences, Medical School, University of Crete, Heraklion, Greece.
| |
Collapse
|
11
|
Engineered Nanomaterials and Occupational Allergy. CURRENT TOPICS IN ENVIRONMENTAL HEALTH AND PREVENTIVE MEDICINE 2017. [DOI: 10.1007/978-981-10-0351-6_3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
12
|
Dandley EC, Taylor AJ, Duke KS, Ihrie MD, Shipkowski KA, Parsons GN, Bonner JC. Atomic layer deposition coating of carbon nanotubes with zinc oxide causes acute phase immune responses in human monocytes in vitro and in mice after pulmonary exposure. Part Fibre Toxicol 2016; 13:29. [PMID: 27278808 PMCID: PMC4899913 DOI: 10.1186/s12989-016-0141-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 06/02/2016] [Indexed: 01/07/2023] Open
Abstract
Background Atomic layer deposition (ALD) is a method for applying conformal nanoscale coatings on three-dimensional structures. We hypothesized that surface functionalization of multi-walled carbon nanotubes (MWCNTs) with polycrystalline ZnO by ALD would alter pro-inflammatory cytokine expression by human monocytes in vitro and modulate the lung and systemic immune response following oropharyngeal aspiration in mice. Methods Pristine (U-MWCNTs) were coated with alternating doses of diethyl zinc and water over increasing ALD cycles (10 to 100 ALD cycles) to yield conformal ZnO-coated MWCNTs (Z-MWCNTs). Human THP-1 monocytic cells were exposed to U-MWCNTs or Z-MWCNTs in vitro and cytokine mRNAs measured by Taqman real-time RT-PCR. Male C57BL6 mice were exposed to U- or Z-MWCNTs by oropharyngeal aspiration (OPA) and lung inflammation evaluated at one day post-exposure by histopathology, cytokine expression and differential counting of cells in bronchoalveolar lavage fluid (BALF) cells. Lung fibrosis was evaluated at 28 days. Cytokine mRNAs (IL-6, IL-1β, CXCL10, TNF-α) in lung, heart, spleen, and liver were quantified at one and 28 days. DNA synthesis in lung tissue was measured by bromodeoxyuridine (BrdU) uptake. Results ALD resulted in a conformal coating of MWCNTs with ZnO that increased proportionally to the number of coating cycles. Z-MWCNTs released Zn+2 ions in media and increased IL-6, IL-1β, CXCL10, and TNF-α mRNAs in THP-1 cells in vitro. Mice exposed to Z-MWCNTs by OPA had exaggerated lung inflammation and a 3-fold increase in monocytes and neutrophils in BALF compared to U-MWCNTs. Z-MWCNTs, but not U-MWCNTs, induced IL-6 and CXCL10 mRNA and protein in the lungs of mice and increased IL-6 mRNA in heart and liver. U-MWCNTs but not Z-MWCNTs stimulated airway epithelial DNA synthesis in vivo. Lung fibrosis at 28 days was not significantly different between mice treated with U-MWCNT or Z-MWCNT. Conclusions Pulmonary exposure to ZnO-coated MWCNTs produces a systemic acute phase response that involves the release of Zn+2, lung epithelial growth arrest, and increased IL-6. ALD functionalization with ZnO generates MWCNTs that possess increased risk for human exposure. Electronic supplementary material The online version of this article (doi:10.1186/s12989-016-0141-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Erinn C Dandley
- Department of Chemical & Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, 27695, USA
| | - Alexia J Taylor
- Toxicology Program, Department of Biological Sciences, North Carolina State University, Campus Box 7633, Raleigh, North Carolina, 27695-7633, USA
| | - Katherine S Duke
- Toxicology Program, Department of Biological Sciences, North Carolina State University, Campus Box 7633, Raleigh, North Carolina, 27695-7633, USA
| | - Mark D Ihrie
- Toxicology Program, Department of Biological Sciences, North Carolina State University, Campus Box 7633, Raleigh, North Carolina, 27695-7633, USA
| | - Kelly A Shipkowski
- Toxicology Program, Department of Biological Sciences, North Carolina State University, Campus Box 7633, Raleigh, North Carolina, 27695-7633, USA
| | - Gregory N Parsons
- Department of Chemical & Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, 27695, USA
| | - James C Bonner
- Toxicology Program, Department of Biological Sciences, North Carolina State University, Campus Box 7633, Raleigh, North Carolina, 27695-7633, USA.
| |
Collapse
|
13
|
Pedata P, Petrarca C, Garzillo EM, Di Gioacchino M. Immunotoxicological impact of occupational and environmental nanoparticles exposure: The influence of physical, chemical, and combined characteristics of the particles. Int J Immunopathol Pharmacol 2015; 29:343-53. [PMID: 26684639 DOI: 10.1177/0394632015608933] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 09/07/2015] [Indexed: 01/12/2023] Open
Abstract
While nanotechnology is growing exponentially, the knowledge of the impact of nanoparticles (NPs) on public health and the environment is limited so far. Current nanomaterial research is focused on the applications of nanotechnology, whereas there is little information on exposure assessment and risk characterization associated with NPs. Therefore, it is essential that the factors influencing NPs associated hazards be studied. This review seeks to survey and evaluate the current literature in order to better understand the impact of both airborne and engineered NPs exposure, the mechanisms at the cellular level, and the factors influencing their immunotoxicity. In fact, NPs do have immunotoxicological significance, as immune cells in the bloodstream and tissues do act to eliminate or interact with NPs.Proper characterization of the NPs as well as understanding the processes occurring on the NPs surface when in contact with biological systems is crucial to predict or exclude toxicological effects.
Collapse
Affiliation(s)
- Paola Pedata
- Department of Experimental Medicine, Section of Hygiene, Occupational Medicine and Forensic Medicine, Second University of Naples, Naples, Italy
| | - Claudia Petrarca
- Immunotoxicology and Allergy Unit, Ce.S.I., G. d'Annunzio University Foundation, Chieti, Italy
| | - Elpidio Maria Garzillo
- Department of Experimental Medicine, Section of Hygiene, Occupational Medicine and Forensic Medicine, Second University of Naples, Naples, Italy
| | - Mario Di Gioacchino
- Department of Experimental Medicine and Ageing Science, G. d'Annunzio University Foundation, Chieti, Italy
| |
Collapse
|
14
|
Liu YX, Karsai A, Anderson DS, Silva RM, Uyeminami DL, Van Winkle LS, Pinkerton KE, Liu GY. Single-Cell Mechanics Provides an Effective Means To Probe in Vivo Interactions between Alveolar Macrophages and Silver Nanoparticles. J Phys Chem B 2015; 119:15118-29. [PMID: 26562364 DOI: 10.1021/acs.jpcb.5b07656] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Single-cell mechanics, derived from atomic force microscopy-based technology, provides a new and effective means to investigate nanomaterial-cell interactions upon in vivo exposure. Lung macrophages represent initial and important responses upon introducing nanoparticles into the respiratory tract, as well as particle clearance with time. Cellular mechanics has previously proven effective to probe in vitro nanomaterial-cell interactions. This study extends technology further to probe the interactions between primary alveolar macrophages (AM) and silver nanoparticles (AgNPs) upon in vivo exposure. Two types of AgNPs, 20 and 110 nm, were instilled to rat lung at 0.5 mg AgNPs/kg body weight, and allowed 24 h interaction. The consequences of these interactions were investigated by harvesting the primary AMs while maintaining their biological status. Cellular mechanics measurements revealed the diverse responses among AM cells, due to variations in AgNP uptake and oxidative dissolving into Ag(+). Three major responses are evident: zero to low uptake that does not alter cellular mechanics, intracellular accumulation of AgNPs trigger cytoskeleton rearrangement resulting in the stiffening of mechanics, and damage of cytoskeleton that softens the mechanical profile. These effects were confirmed using confocal imaging of F-actin and measurements of reactive oxygen species production. More detailed intracellular interactions will also be discussed on the basis of this study in conjunction with prior knowledge of AgNP toxicity.
Collapse
Affiliation(s)
- Ying X Liu
- Department of Chemistry, ‡Center for Health and the Environment, §Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, and ∥Department of Pediatrics, School of Medicine, University of California , Davis, California 95616, United States
| | - Arpad Karsai
- Department of Chemistry, ‡Center for Health and the Environment, §Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, and ∥Department of Pediatrics, School of Medicine, University of California , Davis, California 95616, United States
| | - Donald S Anderson
- Department of Chemistry, ‡Center for Health and the Environment, §Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, and ∥Department of Pediatrics, School of Medicine, University of California , Davis, California 95616, United States
| | - Rona M Silva
- Department of Chemistry, ‡Center for Health and the Environment, §Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, and ∥Department of Pediatrics, School of Medicine, University of California , Davis, California 95616, United States
| | - Dale L Uyeminami
- Department of Chemistry, ‡Center for Health and the Environment, §Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, and ∥Department of Pediatrics, School of Medicine, University of California , Davis, California 95616, United States
| | - Laura S Van Winkle
- Department of Chemistry, ‡Center for Health and the Environment, §Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, and ∥Department of Pediatrics, School of Medicine, University of California , Davis, California 95616, United States
| | - Kent E Pinkerton
- Department of Chemistry, ‡Center for Health and the Environment, §Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, and ∥Department of Pediatrics, School of Medicine, University of California , Davis, California 95616, United States
| | - Gang-yu Liu
- Department of Chemistry, ‡Center for Health and the Environment, §Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, and ∥Department of Pediatrics, School of Medicine, University of California , Davis, California 95616, United States
| |
Collapse
|
15
|
Adamcakova-Dodd A, Monick MM, Powers LS, Gibson-Corley KN, Thorne PS. Effects of prenatal inhalation exposure to copper nanoparticles on murine dams and offspring. Part Fibre Toxicol 2015; 12:30. [PMID: 26437892 PMCID: PMC4594905 DOI: 10.1186/s12989-015-0105-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 09/21/2015] [Indexed: 01/04/2023] Open
Abstract
Background Increasing numbers of individuals may be exposed to nanomaterials during pregnancy. The overarching goal of this investigation was to determine if prenatal inhalation exposure to copper nanoparticles (Cu NPs) has an effect on dams and offspring, including an analysis of inflammatory markers (Th1/Th2 cytokine profiles). Methods Physicochemical characterization of Cu NPs was performed. Pregnant and non-pregnant mice (C57Bl/6 J) were exposed to Cu NPs or laboratory air in the whole-body chamber for 4 hrs/day on gestation days (GD) 3–19 (3.5 mg/m3). Animals were euthanized on GD 19 (0 week) or 7 weeks later. Bronchoalveolar lavage (BAL) fluid was analyzed for total and differential cells. Cytokine/chemokine concentrations were determined in the BAL fluid and the plasma of dams/non-pregnant mice and pups. Cu content was determined in the lungs and the blood of dams/non-pregnant mice and pups, in the placentas as well as in the whole bodies of pups immediately after delivery. Lungs and placentas were evaluated for histopathological changes. Gene expression of the Th1/Th2 profiles were analyzed in spleens of pups. Results The survival rate of 7 week old pups exposed to Cu NPs was significantly lower than control pups (73 vs. 97 %). The average litter size, male/female ratio, body weight and lenght at birth were not different between Cu NP-exposed and control mice. Both pregnant and non-pregnant mice exposed to Cu NPs had significant pulmonary inflammation with increased number of neutrophils in the BAL fluid compared to controls. Perivascular lymphoplasmacytic cuffing was found in the lungs of exposed mice and was more pronounced in the non-pregnant group. Similarly, levels of inflammatory cytokines/chemokines IL-12(p40), G-CSF, GM-CSF, KC, MCP-1, MIP-1α, MIP-1β, RANTES and TNF-α in BAL fluid were significantly higher in non-pregnant than pregnant exposed mice. Histopathology evaluation of placentas did not identify any pathological changes. No translocation of Cu into the placenta or the fetus was found by inductively coupled plasma-mass spectroscopy. Expression of several Th1/Th2 or other immune response genes in pups’ spleens were found to be significantly up- or down-regulated. Conclusions Prenatal exposure to Cu NPs caused a profound pulmonary inflammation in dams and strong immunomodulatory effects in offspring. There was no clear polarization of genes expressed in pups’ spleens towards Th1 or Th2 type of response. Electronic supplementary material The online version of this article (doi:10.1186/s12989-015-0105-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Andrea Adamcakova-Dodd
- Department of Occupational and Environmental Health, University of Iowa, College of Public Health, UI Research Park, IREH 170, Iowa City, IA, 52242, USA.
| | - Martha M Monick
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Linda S Powers
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | | | - Peter S Thorne
- Department of Occupational and Environmental Health, University of Iowa, College of Public Health, UI Research Park, IREH 170, Iowa City, IA, 52242, USA
| |
Collapse
|
16
|
Shipkowski KA, Taylor AJ, Thompson EA, Glista-Baker EE, Sayers BC, Messenger ZJ, Bauer RN, Jaspers I, Bonner JC. An Allergic Lung Microenvironment Suppresses Carbon Nanotube-Induced Inflammasome Activation via STAT6-Dependent Inhibition of Caspase-1. PLoS One 2015; 10:e0128888. [PMID: 26091108 PMCID: PMC4474696 DOI: 10.1371/journal.pone.0128888] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 05/02/2015] [Indexed: 01/12/2023] Open
Abstract
Background Multi-walled carbon nanotubes (MWCNTs) represent a human health risk as mice exposed by inhalation display pulmonary fibrosis. Production of IL-1β via inflammasome activation is a mechanism of MWCNT-induced acute inflammation and has been implicated in chronic fibrogenesis. Mice sensitized to allergens have elevated T-helper 2 (Th2) cytokines, IL-4 and IL-13, and are susceptible to MWCNT-induced airway fibrosis. We postulated that Th2 cytokines would modulate MWCNT-induced inflammasome activation and IL-1β release in vitro and in vivo during allergic inflammation. Methods THP-1 macrophages were primed with LPS, exposed to MWCNTs and/or IL-4 or IL-13 for 24 hours, and analyzed for indicators of inflammasome activation. C57BL6 mice were sensitized to house dust mite (HDM) allergen and MWCNTs were delivered to the lungs by oropharyngeal aspiration. Mice were euthanized 1 or 21 days post-MWCNT exposure and evaluated for lung inflammasome components and allergic inflammatory responses. Results Priming of THP-1 macrophages with LPS increased pro-IL-1β and subsequent exposure to MWCNTs induced IL-1β secretion. IL-4 or IL-13 decreased MWCNT-induced IL-1β secretion by THP-1 cells and reduced pro-caspase-1 but not pro-IL-1β. Treatment of THP-1 cells with STAT6 inhibitors, either Leflunomide or JAK I inhibitor, blocked suppression of caspase activity by IL-4 and IL-13. In vivo, MWCNTs alone caused neutrophilic infiltration into the lungs of mice 1 day post-exposure and increased IL-1β in bronchoalveolar lavage fluid (BALF) and pro-caspase-1 immuno-staining in macrophages and airway epithelium. HDM sensitization alone caused eosinophilic inflammation with increased IL-13. MWCNT exposure after HDM sensitization increased total cell numbers in BALF, but decreased numbers of neutrophils and IL-1β in BALF as well as reduced pro-caspase-1 in lung tissue. Despite reduced IL-1β mice exposed to MWCNTs after HDM developed more severe airway fibrosis by 21 days and had increased pro-fibrogenic cytokine mRNAs. Conclusions These data indicate that Th2 cytokines suppress MWCNT-induced inflammasome activation via STAT6-dependent down-regulation of pro-caspase-1 and suggest that suppression of inflammasome activation and IL-1β by an allergic lung microenvironment is a mechanism through which MWCNTs exacerbate allergen-induced airway fibrosis.
Collapse
Affiliation(s)
- Kelly A. Shipkowski
- Department of Biological Sciences, Environmental and Molecular Toxicology Program, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Alexia J. Taylor
- Department of Biological Sciences, Environmental and Molecular Toxicology Program, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Elizabeth A. Thompson
- Department of Biological Sciences, Environmental and Molecular Toxicology Program, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Ellen E. Glista-Baker
- Department of Biological Sciences, Environmental and Molecular Toxicology Program, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Brian C. Sayers
- Department of Biological Sciences, Environmental and Molecular Toxicology Program, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Zachary J. Messenger
- Department of Biological Sciences, Environmental and Molecular Toxicology Program, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Rebecca N. Bauer
- Center for Environmental Medicine, Asthma, and Lung Biology, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Ilona Jaspers
- Center for Environmental Medicine, Asthma, and Lung Biology, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - James C. Bonner
- Department of Biological Sciences, Environmental and Molecular Toxicology Program, North Carolina State University, Raleigh, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
17
|
Lee DS, Im HJ, Lee YS. Radionanomedicine: Widened perspectives of molecular theragnosis. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2015; 11:795-810. [DOI: 10.1016/j.nano.2014.12.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 12/17/2014] [Accepted: 12/18/2014] [Indexed: 10/24/2022]
|
18
|
Dubey AK, Thrivikraman G, Basu B. Absence of systemic toxicity in mouse model towards BaTiO3 nanoparticulate based eluate treatment. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2015; 26:103. [PMID: 25655497 DOI: 10.1007/s10856-015-5414-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 10/24/2014] [Indexed: 06/04/2023]
Abstract
One of the existing issues in implant failure of orthopedic biomaterials is the toxicity induced by the fine particles released during long term use in vivo, leading to acute inflammatory response. In developing a new class of piezobiocomposite to mimic the integrated electrical and mechanical properties of bone, bone-mimicking physical properties as well as in vitro cytocompatibility properties have been achieved with spark plasma sintered hydroxyapatite (HA)-barium titanate (BaTiO3) composites. However, the presence of BaTiO3 remains a concern towards the potential toxicity effect. To address this issue, present work reports the first result to conclusively confirm the non-toxic effect of HA-BaTiO3 piezobiocomposite nanoparticulates, in vivo. Twenty BALB/c mice were intra-articularly injected at their right knee joints with different concentrations of HA-BaTiO3 composite of up to 25 mg/ml. The histopathological examination confirmed the absence of any trace of injected particles or any sign of inflammatory reaction in the vital organs, such as heart, spleen, kidney and liver at 7 days post-exposure period. Rather, the injected nanoparticulates were found to be agglomerated in the vicinity of the knee joint, surrounded by macrophages. Importantly, the absence of any systemic toxicity response in any of the vital organs in the treated mouse model, other than a mild local response at the site of delivery, was recorded. The serum biochemical analyses using proinflammatory cytokines (TNF-α and IL-1β) also complimented to the non-immunogenic response to injected particulates. Altogether, the absence of any inflammatory/adverse reaction will open up myriad of opportunities for BaTiO3 based piezoelectric implantable devices in biomedical applications.
Collapse
Affiliation(s)
- Ashutosh Kumar Dubey
- Laboratory for Biomaterials, Materials Research Centre, Indian Institute of Science, Bangalore, 560012, India
| | | | | |
Collapse
|
19
|
Glista-Baker EE, Taylor AJ, Sayers BC, Thompson EA, Bonner JC. Nickel nanoparticles cause exaggerated lung and airway remodeling in mice lacking the T-box transcription factor, TBX21 (T-bet). Part Fibre Toxicol 2014; 11:7. [PMID: 24499286 PMCID: PMC3931667 DOI: 10.1186/1743-8977-11-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 02/02/2014] [Indexed: 01/22/2023] Open
Abstract
Background Nickel nanoparticles (NiNPs) are increasingly used in a variety of industrial applications, including the manufacturing of multi-walled carbon nanotubes (MWCNTs). While occupational nickel exposure is a known cause of pulmonary alveolitis, fibrosis, and cancer, the health risks of NiNPs are not well understood, especially in susceptible individuals such as asthmatics. The T-box transcription factor Tbx21 (T-bet) maintains Th1 cell development and loss of T-bet is associated with a shift towards Th2 type allergic airway inflammation that characterizes asthma. The purpose of this study was to determine the role of T-bet in susceptibility to lung remodeling by NiNPs or MWCNTs. Methods Wild-type (WT) and T-bet-/- mice were exposed to NiNPs or MWCNTs (4 mg/kg) by oropharyngeal aspiration (OPA). Necropsy was performed at 1 and 21 days. Bronchoalveolar lavage fluid (BALF) was collected for differential counting of inflammatory cells and for measurement of cytokines by ELISA. The left lung was collected for histopathology. The right lung was analyzed for cytokine or mucin (MUC5AC and MUC5B) mRNAs. Results Morphometry of alcian-blue/periodic acid Schiff (AB/PAS)-stained lung tissue showed that NiNPs significantly increased mucous cell metaplasia in T-bet-/- mice at 21 days (p < 0.001) compared to WT mice, and increased MUC5AC and MUC5B mRNAs (p < 0.05). MWCNTs also increased mucous cell metaplasia in T-bet-/- mice, but to a lesser extent than NiNPs. Chronic alveolitis was also increased by NiNPs, but not MWCNTs, in T-bet-/- mice compared to WT mice at 21 days (P < 0.001). NiNPs also increased IL-13 and eosinophils (p < 0.001) in BALF from T-bet-/- mice after 1 day. Interestingly, the chemokine CCL2 in the BALF of T-bet-/- mice was increased at 1 and 21 days (p < 0.001 and p < 0.05, respectively) by NiNPs, and to a lesser extent by MWCNTs at 1 day. Treatment of T-bet-/- mice with a monoclonal anti-CCL2 antibody enhanced NiNP-induced mucous cell metaplasia and MUC5AC mRNA levels (p < 0.05), yet marginally reduced NiNP-induced alveolitis. Conclusion These findings identify T-bet as a potentially important susceptibility factor for NiNP exposure and to a lesser extent for MWCNT exposure, and suggests that individuals with asthma are at greater risk.
Collapse
Affiliation(s)
| | | | | | | | - James C Bonner
- Environmental & Molecular Toxicology Program, Department of Biological Sciences, North Carolina State University, Campus Box 7633, Raleigh, North Carolina 27695, USA.
| |
Collapse
|