Physicochemical characteristics of nanomaterials that affect pulmonary inflammation

The increasing manufacture and use of products based on nanotechnology raises concerns for both workers and consumers. Various studies report induction of pulmonary inflammation after inhalation exposure to nanoparticles, which can vary in aspects such as size, shape, charge, crystallinity, chemical composition, and dissolution rate. Each of these aspects can affect their toxicity, although it is largely unknown to what extent. The aim of the current review is to analyse published data on inhalation of nanoparticles to identify and evaluate the contribution of their physicochemical characteristics to the onset and development of pulmonary inflammation. Many physicochemical characteristics of nanoparticles affect their lung deposition, clearance, and pulmonary response that, in combination, ultimately determine whether pulmonary inflammation will occur and to what extent. Lung deposition is mainly determined by the physical properties of the aerosol (size, density, shape, hygroscopicity) in relation to airflow and the anatomy of the respiratory system, whereas clearance and translocation of nanoparticles are mainly determined by their geometry and surface characteristics. Besides size and chemical composition, other physicochemical characteristics influence the induction of pulmonary inflammation after inhalation. As some nanoparticles dissolve, they can release toxic ions that can damage the lung tissue, making dissolution rate an important characteristic that affects lung inflammation. Fibre-shaped materials are more toxic to the lungs compared to spherical shaped nanoparticles of the same chemical composition. In general, cationic nanoparticles are more cytotoxic than neutral or anionic nanoparticles. Finally, surface reactivity correlates well with observed pulmonary inflammation. With all these characteristics affecting different stages of the events leading to pulmonary inflammation, no unifying dose metric could be identified to describe pulmonary inflammation for all nanomaterials, although surface reactivity might be a useful measure. To determine the extent to which the various characteristics influence the induction of pulmonary inflammation, the effect of these characteristics on lung deposition, clearance, and pulmonary response should be systematically evaluated. The results can then be used to facilitate risk assessment by categorizing nanoparticles according to their characteristics.

[1]  Jürgen Seitz,et al.  Size dependence of the translocation of inhaled iridium and carbon nanoparticle aggregates from the lung of rats to the blood and secondary target organs , 2009, Inhalation toxicology.

[2]  B. van Ravenzwaay,et al.  Comparative inhalation toxicity of multi-wall carbon nanotubes, graphene, graphite nanoplatelets and low surface carbon black , 2013, Particle and Fibre Toxicology.

[3]  Mark R Wiesner,et al.  Comparison of the abilities of ambient and manufactured nanoparticles to induce cellular toxicity according to an oxidative stress paradigm. , 2006, Nano letters.

[4]  Sir,et al.  Nanomaterials in consumer products : Update of products on the European market in 2010 , 2011 .

[5]  S. Becker,et al.  Murine Pulmonary Inflammatory Responses Following Instillation of Size-Fractionated Ambient Particulate Matter , 2003, Journal of toxicology and environmental health. Part A.

[6]  J. Arts,et al.  Five-day inhalation toxicity study of three types of synthetic amorphous silicas in Wistar rats and post-exposure evaluations for up to 3 months. , 2007, Food and chemical toxicology : an international journal published for the British Industrial Biological Research Association.

[7]  Mark Bradley,et al.  Differential pro-inflammatory effects of metal oxide nanoparticles and their soluble ions in vitro and in vivo; zinc and copper nanoparticles, but not their ions, recruit eosinophils to the lungs , 2012, Nanotoxicology.

[8]  W. MacNee,et al.  Progressive severe lung injury by zinc oxide nanoparticles; the role of Zn2+ dissolution inside lysosomes , 2011, Particle and Fibre Toxicology.

[9]  P. Morrow,et al.  Dust overloading of the lungs: update and appraisal. , 1992, Toxicology and applied pharmacology.

[10]  Vincent Castranova,et al.  Distribution and persistence of pleural penetrations by multi-walled carbon nanotubes , 2010, Particle and Fibre Toxicology.

[11]  T. Xia,et al.  Toxic Potential of Materials at the Nanolevel , 2006, Science.

[12]  Craig A. Poland,et al.  Asbestos, carbon nanotubes and the pleural mesothelium: a review of the hypothesis regarding the role of long fibre retention in the parietal pleura, inflammation and mesothelioma , 2010, Particle and Fibre Toxicology.

[13]  W. D. de Jong,et al.  The effect of particle size on the cytotoxicity, inflammation, developmental toxicity and genotoxicity of silver nanoparticles. , 2011, Biomaterials.

[14]  Wolfgang G Kreyling,et al.  The effect of primary particle size on biodistribution of inhaled gold nano-agglomerates. , 2013, Biomaterials.

[15]  Peter Gehr,et al.  A three-dimensional cellular model of the human respiratory tract to study the interaction with particles. , 2005, American journal of respiratory cell and molecular biology.

[16]  Tsun-Jen Cheng,et al.  Pulmonary toxicity of inhaled nanoscale and fine zinc oxide particles: Mass and surface area as an exposure metric , 2011, Inhalation toxicology.

[17]  J. Vincent,et al.  Kinetics of deposition and clearance of inhaled mineral dusts during chronic exposure. , 1985, British journal of industrial medicine.

[18]  Wei Yang,et al.  Inhaled nanoparticles--a current review. , 2008, International journal of pharmaceutics.

[19]  Z. Reich,et al.  Cargo surface hydrophobicity is sufficient to overcome the nuclear pore complex selectivity barrier , 2009, The EMBO journal.

[20]  E. Teller,et al.  ADSORPTION OF GASES IN MULTIMOLECULAR LAYERS , 1938 .

[21]  Nianqiang Wu,et al.  Acute pulmonary dose–responses to inhaled multi-walled carbon nanotubes , 2012, Nanotoxicology.

[22]  Jürgen Pauluhn,et al.  Subchronic 13-week inhalation exposure of rats to multiwalled carbon nanotubes: toxic effects are determined by density of agglomerate structures, not fibrillar structures. , 2010, Toxicological sciences : an official journal of the Society of Toxicology.

[23]  T. Xia,et al.  Understanding biophysicochemical interactions at the nano-bio interface. , 2009, Nature materials.

[24]  Manuela Semmler-Behnke,et al.  The role of macrophages in the clearance of inhaled ultrafine titanium dioxide particles. , 2008, American journal of respiratory cell and molecular biology.

[25]  Flemming R. Cassee,et al.  Multiple Path Particle Dosimetry model (MPPD v1.0): A model for human and rat airway particle dosimetry , 2002 .

[26]  Vicki Stone,et al.  Carbon black nanoparticles induce type II epithelial cells to release chemotaxins for alveolar macrophages , 2005, Particle and Fibre Toxicology.

[27]  Oleg G. Poluektov,et al.  Improving Optical and Charge Separation Properties of Nanocrystalline TiO2 by Surface Modification with Vitamin C , 1999 .

[28]  K. Wittmaack In Search of the Most Relevant Parameter for Quantifying Lung Inflammatory Response to Nanoparticle Exposure: Particle Number, Surface Area, or What? , 2006, Environmental health perspectives.

[29]  Lijuan Wang,et al.  Acute toxicity of ferric oxide and zinc oxide nanoparticles in rats. , 2010, Journal of nanoscience and nanotechnology.

[30]  Tian Xia,et al.  The role of oxidative stress in ambient particulate matter-induced lung diseases and its implications in the toxicity of engineered nanoparticles. , 2008, Free radical biology & medicine.

[31]  Matthias Ochs,et al.  Interactions of nanoparticles with pulmonary structures and cellular responses. , 2008, American journal of physiology. Lung cellular and molecular physiology.

[32]  Kenneth A. Dawson,et al.  Nanoparticle size and surface properties determine the protein corona with possible implications for biological impacts , 2008, Proceedings of the National Academy of Sciences.

[33]  Marianne Geiser,et al.  Deposition and biokinetics of inhaled nanoparticles , 2010, Particle and Fibre Toxicology.

[34]  N. Herlin‐Boime,et al.  In vitro investigation of oxide nanoparticle and carbon nanotube toxicity and intracellular accumulation in A549 human pneumocytes. , 2008, Toxicology.

[35]  Suresh K Varghese,et al.  Particle deposition in human respiratory system: Deposition of concentrated hygroscopic aerosols , 2009, Inhalation toxicology.

[36]  B. Lehnert,et al.  Correlation between particle size, in vivo particle persistence, and lung injury. , 1994, Environmental health perspectives.

[37]  D. Ingber,et al.  Reconstituting Organ-Level Lung Functions on a Chip , 2010, Science.

[38]  N. Jacobsen,et al.  Role of oxidative damage in toxicity of particulates , 2010, Free radical research.

[39]  J. Nakanishi,et al.  Comparative pulmonary toxicity study of nano-TiO(2) particles of different sizes and agglomerations in rats: different short- and long-term post-instillation results. , 2009, Toxicology.

[40]  P. Baron,et al.  Unusual inflammatory and fibrogenic pulmonary responses to single-walled carbon nanotubes in mice. , 2005, American journal of physiology. Lung cellular and molecular physiology.

[41]  R Tardif,et al.  Effects of inhaled nano-TiO2 aerosols showing two distinct agglomeration states on rat lungs. , 2012, Toxicology letters.

[42]  K. Rabe,et al.  Human neutrophil defensins induce lung epithelial cell proliferation in vitro , 2002, Journal of leukocyte biology.

[43]  K. Donaldson,et al.  Inhaled nanoparticles and lung cancer - what we can learn from conventional particle toxicology. , 2012, Swiss medical weekly.

[44]  B. Brunekreef,et al.  IMMUNE BIOMARKERS IN RELATION TO EXPOSURE TO PARTICULATE MATTER: A Cross-Sectional Survey in 17 Cities of Central Europe , 2000, Inhalation toxicology.

[45]  D. Grainger,et al.  Nanoparticles in the Lung , 2010 .

[46]  D. Thwaites CHAPTER 12 , 1999 .

[47]  Agnes B Kane,et al.  Biological interactions of graphene-family nanomaterials: an interdisciplinary review. , 2012, Chemical research in toxicology.

[48]  A. Proykova,et al.  Scientific Basis for the Definition of the Term “nanomaterial” , 2010 .

[49]  Andrea Steinbrück,et al.  Comprehensive analysis of the effects of CdSe quantum dot size, surface charge, and functionalization on primary human lung cells. , 2012, ACS nano.

[50]  Inge Mangelsdorf,et al.  Change in agglomeration status and toxicokinetic fate of various nanoparticles in vivo following lung exposure in rats , 2012, Inhalation toxicology.

[51]  Robert Landsiedel,et al.  Comparing fate and effects of three particles of different surface properties: nano-TiO(2), pigmentary TiO(2) and quartz. , 2009, Toxicology letters.

[52]  J. Martens,et al.  Oxidative stress and proinflammatory effects of carbon black and titanium dioxide nanoparticles: role of particle surface area and internalized amount. , 2009, Toxicology.

[53]  Wolfgang G Kreyling,et al.  Nanoparticles in the lung , 2010, Nature Biotechnology.

[54]  J. Finkelstein,et al.  Acute pulmonary effects of ultrafine particles in rats and mice. , 2000, Research report.

[55]  Ken Donaldson,et al.  Education: Ultrafine Particles , 2001 .

[56]  J. Tschopp,et al.  Nanoparticles activate the NLR pyrin domain containing 3 (Nlrp3) inflammasome and cause pulmonary inflammation through release of IL-1α and IL-1β , 2010, Proceedings of the National Academy of Sciences.

[57]  Arno C Gutleb,et al.  An improved 3D tetraculture system mimicking the cellular organisation at the alveolar barrier to study the potential toxic effects of particles on the lung , 2013, Particle and Fibre Toxicology.

[58]  Myrtill Simkó,et al.  Risks from accidental exposures to engineered nanoparticles and neurological health effects: A critical review , 2010, Particle and Fibre Toxicology.

[59]  J. Heyder,et al.  Instillation of Six Different Ultrafine Carbon Particles Indicates a Surface Area Threshold Dose for Acute Lung Inflammation in Mice , 2005, Environmental health perspectives.

[60]  W. J. Bair,et al.  The ICRP Human Respiratory Tract Model for Radiological Protection , 1995 .

[61]  Yuh-Jeen Huang,et al.  Effects of various physicochemical characteristics on the toxicities of ZnO and TiO nanoparticles toward human lung epithelial cells. , 2011, The Science of the total environment.

[62]  Christian Mühlfeld,et al.  Translocation and cellular entering mechanisms of nanoparticles in the respiratory tract. , 2008, Swiss medical weekly.

[63]  Monty Liong,et al.  Cationic polystyrene nanosphere toxicity depends on cell-specific endocytic and mitochondrial injury pathways. , 2008, ACS nano.

[64]  Stephen S. Olin,et al.  THE RELEVANCE OF THE RAT LUNG RESPONSE TO PARTICLE OVERLOAD FOR HUMAN RISK ASSESSMENT: A Workshop Consensus Report , 2000, Inhalation toxicology.

[65]  V. Grassian,et al.  Inhalation Exposure Study of Titanium Dioxide Nanoparticles with a Primary Particle Size of 2 to 5 nm , 2006, Environmental health perspectives.

[66]  Robert A Hoke,et al.  Development of a base set of toxicity tests using ultrafine TiO2 particles as a component of nanoparticle risk management. , 2007, Toxicology letters.

[67]  M. L. Laucks,et al.  Aerosol Technology Properties, Behavior, and Measurement of Airborne Particles , 2000 .

[68]  K. Donaldson,et al.  Type 1 pulmonary epithelial cells: a new compartment involved in the slow phase of particle clearance from alveoli , 2012, Nanotoxicology.

[69]  Feng Yang,et al.  Differential mouse pulmonary dose and time course responses to titanium dioxide nanospheres and nanobelts. , 2013, Toxicological sciences : an official journal of the Society of Toxicology.

[70]  Vladimir Murashov,et al.  Adverse Effects of Engineered Nanomaterials Exposure, Toxicology, and Impact on Human Health , 2013 .

[71]  I. Yu,et al.  Subchronic inhalation toxicity of silver nanoparticles. , 2009, Toxicological sciences : an official journal of the Society of Toxicology.

[72]  W. MacNee,et al.  The pro-inflammatory effects of low-toxicity low-solubility particles, nanoparticles and fine particles, on epithelial cells in vitro: the role of surface area , 2007, Occupational and Environmental Medicine.

[73]  Flemming R Cassee,et al.  Impact of agglomeration state of nano- and submicron sized gold particles on pulmonary inflammation , 2010, Particle and Fibre Toxicology.

[74]  Lang Tran,et al.  The biologically effective dose in inhalation nanotoxicology. , 2013, Accounts of chemical research.

[75]  W G Kreyling,et al.  Intracellular particle dissolution in alveolar macrophages. , 1992, Environmental health perspectives.

[76]  Markus Schulz,et al.  Genotoxicity investigations on nanomaterials: methods, preparation and characterization of test material, potential artifacts and limitations--many questions, some answers. , 2009, Mutation research.

[77]  W. MacNee,et al.  Ultrafine particles , 2001, Occupational and environmental medicine.

[78]  Charalambos Kaittanis,et al.  Surface-charge-dependent cell localization and cytotoxicity of cerium oxide nanoparticles. , 2010, ACS nano.

[79]  Michael J Oldham,et al.  New developments in aerosol dosimetry , 2010, Inhalation toxicology.

[80]  G Oberdörster,et al.  Dosimetric principles for extrapolating results of rat inhalation studies to humans, using an inhaled Ni compound as an example. , 1989, Health physics.

[81]  A. Karim,et al.  Toxicological Evaluation of Lung Responses After Intratracheal Exposure to Non-Dispersed Titanium Dioxide Nanorods , 2011, Journal of toxicology and environmental health. Part A.

[82]  Hak Soo Choi,et al.  Rapid translocation of nanoparticles from the lung airspaces to the body , 2010, Nature Biotechnology.

[83]  Craig A. Poland,et al.  Carbon nanotubes introduced into the abdominal cavity of mice show asbestos-like pathogenicity in a pilot study. , 2008, Nature nanotechnology.

[84]  V L Roggli,et al.  Persistence of long, thin chrysotile asbestos fibers in the lungs of rats. , 1994, Environmental health perspectives.

[85]  G. Oberdörster,et al.  Nanotoxicology: An Emerging Discipline Evolving from Studies of Ultrafine Particles , 2005, Environmental health perspectives.

[86]  Ken Donaldson,et al.  Use of silver nanowires to determine thresholds for fibre length-dependent pulmonary inflammation and inhibition of macrophage migration in vitro , 2012, Particle and Fibre Toxicology.

[87]  R. J. Talbot,et al.  Significance of fibre length in the clearance of asbestos fibres from the lung. , 1978, British journal of industrial medicine.

[88]  Xinli Huang,et al.  Neutrophils in acute lung injury. , 2012, Frontiers in bioscience.

[89]  Amanda Hayes,et al.  Nanoparticles: a review of particle toxicology following inhalation exposure , 2012, Inhalation toxicology.

[90]  Benoit Nemery,et al.  Cytokine production by co-cultures exposed to monodisperse amorphous silica nanoparticles: the role of size and surface area. , 2012, Toxicology letters.

[91]  Flemming R. Cassee,et al.  Particle size-dependent total mass deposition in lungs determines inhalation toxicity of cadmium chloride aerosols in rats. Application of a multiple path dosimetry model , 2002, Archives of Toxicology.

[92]  Jürgen Pauluhn,et al.  Pulmonary toxicity and fate of agglomerated 10 and 40 nm aluminum oxyhydroxides following 4-week inhalation exposure of rats: toxic effects are determined by agglomerated, not primary particle size. , 2009, Toxicological sciences : an official journal of the Society of Toxicology.

[93]  Jürgen Pauluhn,et al.  Retrospective analysis of 4-week inhalation studies in rats with focus on fate and pulmonary toxicity of two nanosized aluminum oxyhydroxides (boehmite) and pigment-grade iron oxide (magnetite): the key metric of dose is particle mass and not particle surface area. , 2009, Toxicology.

[94]  A. Gutleb,et al.  Potential of coculture in vitro models to study inflammatory and sensitizing effects of particles on the lung. , 2011, Toxicology in vitro : an international journal published in association with BIBRA.

[95]  Wim H. De Jong,et al.  Interactions with the Human Body , 2012 .

[96]  A. Ledbetter,et al.  Systemic translocation of particulate matter-associated metals following a single intratracheal instillation in rats. , 2007, Toxicological sciences : an official journal of the Society of Toxicology.

[97]  D. Chandler Interfaces and the driving force of hydrophobic assembly , 2005, Nature.

[98]  V J Feron,et al.  Subchronic inhalation toxicity of amorphous silicas and quartz dust in rats. , 1991, Food and chemical toxicology : an international journal published for the British Industrial Biological Research Association.

[99]  Wolfgang Koch,et al.  Chronic Inhalation Exposure of Wistar Rats and two Different Strains of Mice to Diesel Engine Exhaust, Carbon Black, and Titanium Dioxide , 1995 .

[100]  Wolfgang Koch, Werner Stöber A SIMPLE PULMONARY RETENTION MODEL ACCOUNTING FOR DISSOLUTION AND MACROPHAGE-MEDIATED REMOVAL OF DEPOSITED POLYDISPERSE PARTICLES , 2001, Inhalation toxicology.

[101]  P. Morrow,et al.  Possible mechanisms to explain dust overloading of the lungs. , 1988, Fundamental and applied toxicology : official journal of the Society of Toxicology.

[102]  Marianne Geiser,et al.  Update on macrophage clearance of inhaled micro- and nanoparticles. , 2010, Journal of aerosol medicine and pulmonary drug delivery.

[103]  Jing Wang,et al.  Nano-bio effects: interaction of nanomaterials with cells. , 2013, Nanoscale.

[104]  W. Kreyling Lung deposition and biokinetics of inhaled nanoparticles , 2013 .

[105]  Ken Donaldson,et al.  Graphene-based nanoplatelets: a new risk to the respiratory system as a consequence of their unusual aerodynamic properties. , 2012, ACS nano.

[106]  Craig A. Poland,et al.  Zeta potential and solubility to toxic ions as mechanisms of lung inflammation caused by metal/metal oxide nanoparticles. , 2012, Toxicological sciences : an official journal of the Society of Toxicology.

[107]  David M. Brown,et al.  Proinflammogenic Effects of Low-Toxicity and Metal Nanoparticles In Vivo and In Vitro: Highlighting the Role of Particle Surface Area and Surface Reactivity , 2007, Inhalation toxicology.

[108]  David B Warheit,et al.  Pulmonary bioassay studies with nanoscale and fine-quartz particles in rats: toxicity is not dependent upon particle size but on surface characteristics. , 2007, Toxicological sciences : an official journal of the Society of Toxicology.

[109]  R. Williams,et al.  Influence of particle size on regional lung deposition--what evidence is there? , 2011, International journal of pharmaceutics.

[110]  L. Nicod Pulmonary Defence Mechanisms , 1999, Respiration.

[111]  Nianqiang Wu,et al.  Mouse pulmonary dose- and time course-responses induced by exposure to multi-walled carbon nanotubes. , 2010, Toxicology.

[112]  G. Oster,et al.  Quartz hemolysis as related to its surface functionalities. , 1981, Environmental research.

[113]  V. Castranova Overview of Current Toxicological Knowledge of Engineered Nanoparticles , 2011, Journal of occupational and environmental medicine.

[114]  Antonio Nunes,et al.  Length-dependent retention of carbon nanotubes in the pleural space of mice initiates sustained inflammation and progressive fibrosis on the parietal pleura. , 2011, The American journal of pathology.

[115]  Roel P F Schins,et al.  Inhaled particles and lung cancer. Part A: Mechanisms , 2004, International journal of cancer.

[116]  Nancy A. Monteiro-Riviere,et al.  Cellular uptake mechanisms and toxicity of quantum dots in dendritic cells. , 2011, Nanomedicine.

[117]  Ken Donaldson,et al.  Possible genotoxic mechanisms of nanoparticles: Criteria for improved test strategies , 2010, Nanotoxicology.

[118]  Vicki H. Grassian,et al.  Silver nanoparticles in simulated biological media: a study of aggregation, sedimentation, and dissolution , 2011 .

[119]  Sir,et al.  Nanomaterial in consumer products : Detection, characterisation and interpretation , 2011 .

[120]  E. Niki,et al.  Comparison of acute oxidative stress on rat lung induced by nano and fine-scale, soluble and insoluble metal oxide particles: NiO and TiO2 , 2012, Inhalation toxicology.

[121]  Rogene F. Henderson,et al.  Concepts In Inhalation Toxicology , 1995 .

[122]  Alexandra Kroll,et al.  Testing Metal‐Oxide Nanomaterials for Human Safety , 2010, Advanced materials.

[123]  T. Webb,et al.  Pulmonary toxicity study in rats with three forms of ultrafine-TiO2 particles: differential responses related to surface properties. , 2007, Toxicology.

[124]  David B Warheit,et al.  Changing the dose metric for inhalation toxicity studies: Short-term study in rats with engineered aerosolized amorphous silica nanoparticles , 2010, Inhalation toxicology.

[125]  Flemming R Cassee,et al.  Comparative hazard identification of nano- and micro-sized cerium oxide particles based on 28-day inhalation studies in rats , 2014, Nanotoxicology.

[126]  Agnes G Oomen,et al.  Tissue distribution of inhaled micro- and nano-sized cerium oxide particles in rats: results from a 28-day exposure study. , 2012, Toxicological sciences : an official journal of the Society of Toxicology.

[127]  Adriele Prina-Mello,et al.  The threshold length for fiber-induced acute pleural inflammation: shedding light on the early events in asbestos-induced mesothelioma. , 2012, Toxicological sciences : an official journal of the Society of Toxicology.

[128]  R. Müller,et al.  Influence of surface charge density on protein adsorption on polymeric nanoparticles: analysis by two-dimensional electrophoresis. , 2002, European journal of pharmaceutics and biopharmaceutics : official journal of Arbeitsgemeinschaft fur Pharmazeutische Verfahrenstechnik e.V.

[129]  Thomas Kuhlbusch,et al.  Particle and Fibre Toxicology BioMed Central Review The potential risks of nanomaterials: a review carried out for ECETOC , 2006 .

[130]  Maria Hammer,et al.  Time-response relationship of nano and micro particle induced lung inflammation. Quartz as reference compound , 2010, Human & experimental toxicology.

[131]  K. Donaldson,et al.  Differences in the extent of inflammation caused by intratracheal exposure to three ultrafine metals: role of free radicals. , 1998, Journal of toxicology and environmental health. Part A.

[132]  Kirsten Peters,et al.  Lung epithelial cell lines in coculture with human pulmonary microvascular endothelial cells: development of an alveolo-capillary barrier in vitro , 2004, Laboratory Investigation.

[133]  Albert Duschl,et al.  Shape matters: effects of silver nanospheres and wires on human alveolar epithelial cells , 2011, Particle and Fibre Toxicology.

[134]  J. West,et al.  Correlating nanoscale titania structure with toxicity: a cytotoxicity and inflammatory response study with human dermal fibroblasts and human lung epithelial cells. , 2006, Toxicological sciences : an official journal of the Society of Toxicology.

[135]  A. Peters,et al.  Particulate Matter Air Pollution and Cardiovascular Disease: An Update to the Scientific Statement From the American Heart Association , 2010, Circulation.

[136]  Vicki Stone,et al.  Nano-silver – feasibility and challenges for human health risk assessment based on open literature , 2010, Nanotoxicology.

[137]  J. Harington,et al.  Enhancement of haemolytic activity of asbestos by heat-labile factors in fresh serum. , 1971, La Medicina del lavoro.

[138]  W. Kreyling,et al.  TRANSLOCATION OF ULTRAFINE INSOLUBLE IRIDIUM PARTICLES FROM LUNG EPITHELIUM TO EXTRAPULMONARY ORGANS IS SIZE DEPENDENT BUT VERY LOW , 2002, Journal of toxicology and environmental health. Part A.

[139]  Meng Wang,et al.  Comparative study of pulmonary responses to nano- and submicron-sized ferric oxide in rats. , 2008, Toxicology.

[140]  Stella M. Marinakos,et al.  Size-controlled dissolution of organic-coated silver nanoparticles. , 2012, Environmental science & technology.

[141]  B. van Ravenzwaay,et al.  Inhalation toxicity of multiwall carbon nanotubes in rats exposed for 3 months. , 2009, Toxicological sciences : an official journal of the Society of Toxicology.

[142]  Icrp Human Respiratory Tract Model for Radiological Protection , 1994 .

[143]  C. Stanier,et al.  An Algorithm for Combining Electrical Mobility and Aerodynamic Size Distributions Data when Measuring Ambient Aerosol Special Issue of Aerosol Science and Technology on Findings from the Fine Particulate Matter Supersites Program , 2004 .

[144]  G. Daston,et al.  Toxicology of nanoparticles. , 2012, Advanced drug delivery reviews.

[145]  A. Maynard,et al.  Airborne Nanostructured Particles and Occupational Health , 2005 .

[146]  Nicklas Raun Jacobsen,et al.  Biodistribution of gold nanoparticles in mouse lung following intratracheal instillation , 2009, Chemistry Central journal.

[147]  M. Bawendi,et al.  Renal clearance of quantum dots , 2007, Nature Biotechnology.

[148]  J. Jung,et al.  Twenty-Eight-Day Inhalation Toxicity Study of Silver Nanoparticles in Sprague-Dawley Rats , 2007, Inhalation toxicology.

[149]  Jongheop Yi,et al.  Oxidative stress and apoptosis induced by titanium dioxide nanoparticles in cultured BEAS-2B cells. , 2008, Toxicology letters.

[150]  Jürgen Pauluhn,et al.  Poorly soluble particulates: searching for a unifying denominator of nanoparticles and fine particles for DNEL estimation. , 2011, Toxicology.

[151]  K. Rock,et al.  Silica crystals and aluminum salts activate the NALP3 inflammasome through phagosomal destabilization , 2008, Nature Immunology.

[152]  V. Vallyathan,et al.  Contrasting respirable quartz and kaolin retention of lecithin surfactant and expression of membranolytic activity following phospholipase A2 digestion. , 1992, Journal of toxicology and environmental health.