28.01.2015 Views

A single intratracheal instillation of single-walled carbon nanotubes ...

A single intratracheal instillation of single-walled carbon nanotubes ...

A single intratracheal instillation of single-walled carbon nanotubes ...

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

Arch Toxicol (2011) 85:1121–1131<br />

DOI 10.1007/s00204-011-0655-8<br />

ORGAN TOXICITY AND MECHANISMS<br />

A <strong>single</strong> <strong>intratracheal</strong> <strong>instillation</strong> <strong>of</strong> <strong>single</strong>-<strong>walled</strong> <strong>carbon</strong><br />

<strong>nanotubes</strong> induced early lung fibrosis and subchronic tissue<br />

damage in mice<br />

Eun-Jung Park • Jinkyu Roh • Soo-Nam Kim •<br />

Min-sung Kang • Young-Ah Han • Younghun Kim •<br />

Jin Tae Hong • Kyunghee Choi<br />

Received: 13 October 2010 / Accepted: 13 January 2011 / Published online: 7 April 2011<br />

Ó Springer-Verlag 2011<br />

Abstract Large amounts <strong>of</strong> nanomaterials may reach<br />

both the natural and occupational environments. This represents<br />

a potential health hazard. People have forecasted<br />

that CNTs may lead to the toxicity such as mesothelioma<br />

and fibrosis like asbestos. To identify dominant immune<br />

responses induced by SWCNTs, we investigated the composition<br />

<strong>of</strong> bronchioalveolar lavage (BAL) cells, the<br />

secretion <strong>of</strong> cytokine and collagen, histopathology, protein<br />

expression, and cell phenotypes over time after a <strong>single</strong><br />

administration <strong>of</strong> <strong>single</strong>-<strong>walled</strong> <strong>carbon</strong> <strong>nanotubes</strong><br />

(SWCNT). In our results, the number <strong>of</strong> total cells and<br />

macrophages remained at the up-regulated level until Day<br />

28, neutrophils rapidly increased at Day 1, and lymphocytes<br />

increased from Day 7. In the BAL fluid, proinflammatory<br />

cytokines rapidly increased at Day 1 and<br />

remained at an up-regulated level throughout the<br />

Electronic supplementary material The online version <strong>of</strong> this<br />

article (doi:10.1007/s00204-011-0655-8) contains supplementary<br />

material, which is available to authorized users.<br />

E.-J. Park (&) K. Choi<br />

Environmental Health Risk Research Department,<br />

National Institute <strong>of</strong> Environmental Research, Kyungseo-dong,<br />

Seo-gu, Incheon 404-708, Korea<br />

e-mail: pej303@korea.kr<br />

J. Roh Y. Kim<br />

Department <strong>of</strong> Chemical Engineering, Kwangwoon University,<br />

447-1, Wolgye-dong, Nowon-gu, Seoul 139-701, Korea<br />

S.-N. Kim M. Kang Y.-A. Han<br />

Inhalation Toxicology Center, KIT Jeongeup Campus,<br />

1051, Shinjeong-dong, Jeongeup, Korea<br />

J. T. Hong<br />

College <strong>of</strong> Pharmacy and Medical Research Center,<br />

Chungbuk National University, 410, Seongbong-ro,<br />

Heungdeok-gu, Cheongju, Chungbuk 361-763, Korea<br />

experimental period. IL-12 and IL-10 rapidly increased at<br />

Day 1 after administration and remained at a similar level<br />

until Day 28. IFN-c and IL-4 reached the maximum at Day<br />

1, and IL-5, TGF-b, and collagen reached the maximum at<br />

Day 7. IL-13 and IL-17 increased in a time-dependent<br />

manner. The distribution <strong>of</strong> B cells and cytotoxic T cells<br />

markedly increased at Days 7 and 14, and fibrotic lesions<br />

were histopathologically observed at Days 7 and 14. The<br />

expressions <strong>of</strong> caspase-3, p53, COL1A1, COX-2, iNOS,<br />

MMP-9, and MMP-2 were also markedly increased at Days<br />

7 and 14. In addition, the expression <strong>of</strong> mesothelin, iNOS,<br />

MMP-9, and p53 was up-regulated until Day 28. Based on<br />

these findings, we suggest that a <strong>single</strong> <strong>intratracheal</strong><br />

<strong>instillation</strong> <strong>of</strong> SWCNTs may induce early lung fibrosis and<br />

subchronic tissue damage.<br />

Keywords SWCNT Lung fibrosis Mesothelin <br />

Cytokine Inflammation<br />

Introduction<br />

The use <strong>of</strong> manufactured nanomaterials is rapidly<br />

increasing due to nanotechnology advances in different<br />

industries and consumer products. Many feel that these<br />

nanotechnologies have resulted in colossal benefits in<br />

improving the quality <strong>of</strong> life. However, the large amounts<br />

<strong>of</strong> nanomaterials may consequently reach the natural, as<br />

well as occupational, environment and thus represent a<br />

potential health hazard (Shvedova and Kagan 2010; Lam<br />

et al. 2006; Maynard et al. 2004; Warheit et al. 2004). Song<br />

et al. reported symptoms, including non-specific pulmonary<br />

inflammation, pulmonary fibrosis, and foreign-body granulomas<br />

<strong>of</strong> the pleura, in workers exposed to nanoparticles<br />

for 5–13 months (Song et al. 2009).<br />

123


1122 Arch Toxicol (2011) 85:1121–1131<br />

Some nanomaterials <strong>of</strong> importance are <strong>carbon</strong> <strong>nanotubes</strong><br />

(CNTs). CNTs have a unique length-to-diameter ratio,<br />

which is significantly larger than that <strong>of</strong> any other material.<br />

CNTs have been categorized as <strong>single</strong>-<strong>walled</strong> <strong>carbon</strong><br />

<strong>nanotubes</strong> (SWCNTs) and multi-<strong>walled</strong> <strong>carbon</strong> <strong>nanotubes</strong><br />

(MWCNTs), according to the number <strong>of</strong> layers, and both<br />

types <strong>of</strong> CNTs are listed as high-priority groups in the<br />

Organization for Economic Co-operation and Development<br />

(OECD) Steering Group for Test Guidelines.<br />

Many researchers have forecasted that CNTs may lead<br />

to mesothelioma and fibrosis, similar to asbestos, due to<br />

their unique morphology (Pacurari et al. 2008; Takagi et al.<br />

2008; Sakamoto et al. 2009, Donaldson et al. 2010).<br />

Mesothelioma is a cancer that develops from the protective<br />

lining that covers many <strong>of</strong> the body’s internal organs, the<br />

mesothelium, and it is most commonly generated in the<br />

pleura, peritoneum, heart, pericardium, and tunica vaginalis<br />

(Chang and Pastan 1996). Fibrosis is generated in<br />

various organs, such as the lung, liver, heart, and intestine,<br />

as a kind <strong>of</strong> autoimmune disease, and it forms excess<br />

fibrous connective tissue in an organ or tissue as a reparative<br />

or reactive process.<br />

The outbreak <strong>of</strong> environment-related disease is caused<br />

by the destruction <strong>of</strong> immune balance by the xenobiotics<br />

inflowing into the body. In previous reports, some<br />

researchers have suggested that SWCNT induces oxidative<br />

stress and fibrotic inflammatory responses (Folkmann et al.<br />

2009; Pacurari et al. 2008; Shvedova et al. 2008a, b; Chou<br />

et al. 2008; Sharma et al. 2007; Manna et al. 2005). For<br />

example, nicotinamide adenine dinucleotide phosphate<br />

(NADPH) oxidase-deficient C57BL/6 mice showed an<br />

increase in the accumulation <strong>of</strong> neutrophils and a decrease<br />

<strong>of</strong> fibrosis in the lung (Shvedova et al. 2008a, b), and the<br />

pharyngeal aspiration <strong>of</strong> SWCNT elicited a robust but<br />

acute inflammation with an early onset <strong>of</strong> progressive<br />

fibrosis and granulomas in C57BL/6 mice (Shvedova et al.<br />

2005). Furthermore, Chou et al. studied innate and adaptive<br />

immune responses related to chronic pulmonary inflammation<br />

and granuloma formation caused by SWCNT (Chou<br />

et al. 2008). They suggested that the uptake <strong>of</strong> SWCNT<br />

(500 lg) into the macrophages activates various transcription<br />

factors, such as NF-kB and AP-1, and this leads to<br />

oxidative stress, the release <strong>of</strong> pro-inflammatory cytokines,<br />

the recruitment <strong>of</strong> leukocytes, the induction <strong>of</strong> protective<br />

and anti-apoptotic gene expression, and the activation <strong>of</strong> T<br />

cells.<br />

The treatment <strong>of</strong> SWCNT together with OVA strongly<br />

increased serum levels <strong>of</strong> OVA-specific IgE, the number <strong>of</strong><br />

eosinophils in bronchial alveolar lavage fluid (BALF), and<br />

the secretion <strong>of</strong> Th2-associated cytokines in the mediastinal<br />

lymph node (Nygaard et al. 2009). However, there is<br />

still not enough data on the immunotoxicity <strong>of</strong> SWCNTs.<br />

In this study, we investigated the change <strong>of</strong> cytokines and<br />

cells (NK, NKT, B, and T) to identify whether SWCNTs<br />

induce Th1 or Th2 response. Furthermore, we explored<br />

proteins related to lung tissue damage.<br />

Materials and methods<br />

Animals<br />

Male, 6-week-old (40–42 days old, 26 ± 1 g), ICR mice<br />

were purchased from the Orient Bio INC. (Seongnam,<br />

Gyunggi-do, Korea) and were acclimated the animal to<br />

room conditions prior to the initiation <strong>of</strong> the study. The<br />

environmental conditions were a temperature <strong>of</strong> 23 ± 1°C,<br />

relative humidity <strong>of</strong> 55 ± 5%, and a 12-h light/dark cycle.<br />

All the animals used in this study were cared for in<br />

accordance with the principles outlined in the ‘‘Guide for<br />

the Care and Use <strong>of</strong> Laboratory Animals’’ issued by the<br />

Animal Care and Use Committee <strong>of</strong> the National Veterinary<br />

Research and Quarantine Service (NVRQS). To<br />

observe time-dependent change, 16 mice were treated per<br />

time point.<br />

Preparation <strong>of</strong> test material<br />

SWCNTs (approximately 10% weight metal contents,<br />

1.2 nm in diameter, 2–10 lm in length; ASP-100F, Hanhwa<br />

Nanotech, Korea) were dispersed into DI water by<br />

sonication. Because SWCNTs are very hydrophobic and<br />

immiscible to water, 40 mg sodium dodecyl sulfate (SDS,<br />

Sigma–Aldrich) was added to 200 ml <strong>of</strong> the SWCNTtreated<br />

water as a stabilizer. Then, the solution was treated<br />

with sonification (ULH-700S, ULSSO HI-Tech, Korea) for<br />

30 min, 4 kHz (20% PWR), and was then diluted with 29<br />

PBS. As shown in Fig. 1, SWCNT was relatively well<br />

dispersed in the PBS buffer (A, JEM1010, JEOL, Japan),<br />

showed the peak <strong>of</strong> graphite band in Raman spectroscopy<br />

(B, T64000, HORIABA Jobin–Yvon, France), and the<br />

average length was 0.76 lm (C, ELS-8000, Otuska Electronics,<br />

Japan) (Yang et al. 2005; Yu et al. 2010; Wei et al.<br />

2005).<br />

Intratracheal <strong>instillation</strong> and sample preparation<br />

Intratracheal <strong>instillation</strong> and sample preparation was conducted<br />

by a special technician from the Korea Institute <strong>of</strong><br />

Toxicology, one <strong>of</strong> the GLP institutes in Korea. SWCNTs<br />

were delivered using a 24-gauge catheter at a 100 lg/kg<br />

dose by <strong>intratracheal</strong> administration under light tiletamine<br />

123


Arch Toxicol (2011) 85:1121–1131 1123<br />

Fig. 1 Physicochemical properties <strong>of</strong> SWCNTs suspended in PBS a TEM image, b Raman spectroscopy, and c Size distribution<br />

anesthesia. The animals were killed at Days 1, 7, 14, and 28<br />

after treatment. The control group (4 mice per time point)<br />

was treated with a vehicle control solution that was manufactured<br />

by the same method.<br />

At the selected time intervals after administration,<br />

about 1.2 ml <strong>of</strong> blood was collected per mouse from the<br />

saphenous vein. The samples harvested from the 16 mice<br />

per time point were pooled to four test samples for<br />

analysis <strong>of</strong> the BAL cell count, cell phenotype, cytokines,<br />

and collagen (n = 4). Whole blood was centrifuged at<br />

3,000 rpm for 10 min to make serum, and 400–450 ll <strong>of</strong><br />

serum was obtained from each mouse. BAL fluid was<br />

obtained by cannulating the trachea and lavaging the<br />

lungs with 1 ml <strong>of</strong> cold sterile (Ca2 plus Mg2)-free<br />

PBS (0.15 M, pH 7.2). Approximately 500–600 ll <strong>of</strong><br />

BAL fluid was harvested per mouse, and this was centrifuged<br />

at 3,000 rpm for 10 min.<br />

BAL fluid analysis<br />

Total cells in the BAL fluid were quantified by hemocytometric<br />

counting. Cell differentials were performed on<br />

cytocentrifuged preparations fixed in methanol and stained<br />

with Diff-Quick (Thermo Shandon, PA, USA). Distributions<br />

<strong>of</strong> the alveolar macrophages, neutrophils, and lymphocytes<br />

were assessed by their characteristic cell shapes.<br />

Measurement <strong>of</strong> cytokines<br />

The concentrations <strong>of</strong> each cytokine in the supernatant <strong>of</strong><br />

the BAL fluid and serum were determined using commercially<br />

available ELISA kits (eBioscience, San Diego,<br />

CA, USA). First, each well <strong>of</strong> the microplates was coated<br />

with 100 ll <strong>of</strong> capture antibody and incubated overnight at<br />

4°C. After washing and blocking with assay diluent and<br />

BAL fluid, serum or standard antibody was added to the<br />

individual wells. The plates were then maintained at room<br />

temperature for 2 h. Next, the plates were washed, and<br />

biotin-conjugated detecting antibody was added to each<br />

well. Then, the plates were incubated at room temperature<br />

for 1 h. After incubation, the plates were washed again and<br />

further incubated with avidin–HRP for 30 min before<br />

detection using TMB solution. Finally, the reactions were<br />

stopped by adding 1 M H 3 PO 4 , and the absorbance at<br />

450 nm was measured with an ELISA reader (Molecular<br />

Devices, Sunnyvale, CA, USA). The amount <strong>of</strong> cytokine<br />

123


1124 Arch Toxicol (2011) 85:1121–1131<br />

was calculated from the linear portion <strong>of</strong> the generated<br />

standard curve (Park et al. 2009a, b).<br />

Immunophenotyping<br />

All monoclonal antibodies were purchased from eBioscience<br />

(San Diego, CA, USA). T cells (CD3, 1:50), B cells<br />

(CD19, 1:50), NK cells (DX5, 1:100), CD4 T cells<br />

(CD4 , 1:160), and CD8 T cells (CD8 , 1:50) were<br />

identified using directly conjugated anti-mouse antibodies.<br />

First, the blood samples were blocked with Fc-block (eBioscience,<br />

San Diego, CA, USA) to reduce non-specific antibody<br />

binding. The cells were then incubated in the dark with<br />

10 ll <strong>of</strong> the appropriate fluorochrome-conjugated antibody<br />

for 20 min at 4°C. The cells were then washed with fluorescence-activated<br />

cell sorter (FACS) buffer. The blood was<br />

lysed for 5 min with FACS lysis buffer (BD Bioscience,<br />

Franklin Lakes, NJ, USA) at room temperature and then rewashed<br />

with FACS buffer. Finally, each sample was fixed<br />

with 1% paraformaldehyde until further analysis.<br />

Flow cytometry analysis was performed using the<br />

FACSCalibur system (BD Biosciences, Franklin Lakes, NJ,<br />

USA). Control samples were matched for each fluorochrome.<br />

Data were analyzed using CellQuest s<strong>of</strong>tware<br />

(Becton–Dickinson, Franklin Lakes, NJ, USA) (Silva et al.<br />

2005; Vendrame et al. 2006).<br />

Measurement <strong>of</strong> collagen<br />

The collagen concentration was measured using a mouse<br />

hydroxyproline ELISA kit (USCNLIFE SCIENCE and<br />

TECHNOLOGY CO., LTD., Wuhan, China). First, 100 ll<br />

<strong>of</strong> standard, blank, or sample was added per well and the<br />

plates were incubated for 2 h at 37°C. Then, the liquid was<br />

removed from each well and detection reagent A working<br />

solution was added to each well. The plates were then<br />

incubated at 37°C for 1 h. After washing, detection reagent<br />

B working solution was added to each well and incubated<br />

at 37°C for 1 h. After re-washing, substrate solution was<br />

added to each well and incubated at 37°C for 15 min<br />

protecting from light. Finally, the reaction was terminated<br />

by the addition <strong>of</strong> stop solution, and the absorbance was<br />

measured at 450 nm using a microplate reader (Molecular<br />

Devices, Sunnyvale, CA, USA). The amounts <strong>of</strong> secreted<br />

collagen were calculated from the linear portion <strong>of</strong> the<br />

prepared standard curve.<br />

Protein expression in tissue<br />

Lung tissue was homogenized with a protein extraction<br />

solution (PRO-PREP TM , Cat. No. 17081, iNtRON<br />

biotechnology, Kyunggi, Korea), and lysates were centrifuged<br />

at 13,000 rpm for 10 min. The protein concentration<br />

was measured by the Bradford method (Bio-Rad<br />

Protein Assay, Bio-Rad Laboratories Inc., Hercules, CA),<br />

and equal amounts <strong>of</strong> proteins (40 lg) were separated on<br />

a SDS/1%-polyacrylamide gel and then transferred to a<br />

nitrocellulose membrane (Hybond ECL, Amersham<br />

Pharmacia Biotech Inc., Piscataway, NJ, USA). Blots<br />

were blocked for 2 h at room temperature with 5% (w/v)<br />

non-fat dried milk in Tris-buffered saline (10 mM Tris,<br />

pH 8.0, and 150 mM NaCl) solution containing 0.05%<br />

Tween-20. The membranes were immunoblotted with<br />

primary specific antibodies: rabbit polyclonal for rabbit<br />

polyclonal <strong>of</strong> caspase-3 and ADP ribose polymerase<br />

(PARP) (1:1,000 dilution; Cell Signaling Technology, Inc.<br />

Beverly, MA, USA), COX-2 (1:500 dilution; Cayman<br />

Chemical, MI, USA), and MMP-2 (1:500 dilution; Santa<br />

Cruz Biotechnology Inc. CA, USA); rabbit monoclonal<br />

for b-actin (1:2,000 dilution; Cell Signaling Technology,<br />

Inc. Beverly, MA, USA); mouse monoclonal for mesothelin,<br />

p53, and MMP-9 (1:1,000 dilution; Santa Cruz<br />

Biotechnology Inc. CA, USA), and iNOS (1:1,000 dilution;<br />

BD Biosciences, CA, USA); and goat polyclonal for<br />

COL1A1 (1:1,000 dilution; Santa Cruz Biotechnology<br />

Inc. CA, USA). The blots were then incubated with the<br />

corresponding conjugated anti-mouse, anti-rabbit, or antigoat<br />

immunoglobulin G-horseradish peroxidase (1:2,000<br />

dilution; Santa Cruz Biotechnology Inc.). Immunoreactive<br />

proteins were detected with the ECL western blotting<br />

detection system.<br />

Histopathology<br />

Histopathological analysis was performed at the Korea<br />

Institute <strong>of</strong> Toxicology (Yuseong-gu, Daejeon, Korea) on<br />

tissue harvested from six mice per group. The lung, brain,<br />

and thymus from mice in the control group and the treated<br />

group were fixed with 10% neutral buffered formalin and<br />

processed using routine histological techniques. After<br />

paraffin embedding, 3 lm sections were cut and stained<br />

with hematoxylin and eosin (H&E) for histopathological<br />

evaluation.<br />

Statistical analysis<br />

The results obtained from the chemically treated groups<br />

were compared to those <strong>of</strong> the control group. The values<br />

were compared using Dunnett’s t test after one-way<br />

ANOVA, and levels <strong>of</strong> significance were represented<br />

compared to the control group.<br />

123


Arch Toxicol (2011) 85:1121–1131 1125<br />

Results<br />

Cell distribution in BAL fluid<br />

The number <strong>of</strong> total cells recovered in the BAL fluid <strong>of</strong><br />

mice treated with SWCNTs did not significantly increase at<br />

Day 1 after administration but did significantly increase by<br />

Day 7 (Fig. 2a) compared with the control. When the<br />

composition <strong>of</strong> cells in the BAL fluid was analyzed, the<br />

percentage composition <strong>of</strong> neutrophils, which is characteristic<br />

<strong>of</strong> the initial stage <strong>of</strong> inflammation, showed maximum<br />

(8.41 ± 0.01%) increase at Day 1. The distribution<br />

ratio <strong>of</strong> macrophages increased again by Day 7. The distribution<br />

ratio <strong>of</strong> lymphocytes increased to 0.7 ± 0.0%,<br />

1.8 ± 0.0%, 3.4 ± 0.0, and 2.1 ± 0.0% at Days 1, 7, 14,<br />

and 28, respectively, compared to 0.4 ± 0.0% <strong>of</strong> the control<br />

(Fig. 2b).<br />

Cytokines in BAL<br />

To identify inflammatory responses to the administration<br />

<strong>of</strong> SWCNT, we measured the concentrations <strong>of</strong> proinflammatory<br />

cytokines (IL-1, TNF-a, and IL-6), Th0<br />

cytokine (IL-2), Th1-type cytokines (IL-12 and IFN-c),<br />

and Th2-type cytokines (IL-4, IL-5, IL-10, IL-13, and<br />

IL-17) in the BAL fluid over time (Table 1). Proinflammatory<br />

cytokines, such as IL-1b, TNF-a, and IL-6,<br />

rapidly increased at Day 1 after <strong>instillation</strong> and were upregulated<br />

during the experimental period. IL-2 reached<br />

the maximum at Day 7. IL-12 and IL-10 rapidly increased<br />

at Day 1 and remained at a similar level until Day 28.<br />

IFN-c and IL-4 reached the maximum at Day 1, and IL-5<br />

reached the maximum at Day 7. IL-13 and IL-17<br />

increased in a time-dependent manner.<br />

Cytokines in the blood<br />

As shown in Table 2, the level <strong>of</strong> IL-6 reached the maximum<br />

at Day 7, and that <strong>of</strong> IL-12 reached the maximum at<br />

Day 14 after treatment. IL-17 was up-regulated during the<br />

experimental period. However, neither IL-1, TNF-a, IL-2,<br />

IFN-c, IL-4, IL-5, IL-10, nor IL-13 was detected at any <strong>of</strong><br />

the designed time points.<br />

Secretion <strong>of</strong> TGF-b and collagen<br />

In the BAL fluid, the concentration <strong>of</strong> TGF-b was<br />

103.3 ± 6.6 pg/ml, 146.2 ± 9.2 pg/ml, 106.9 ± 1.1 pg/ml,<br />

and 117.6 ± 2.0 pg/ml at Days 1, 7, 14, and 28,<br />

respectively, after treatment. The concentration for the<br />

control was 7.2 ± 0.1 pg/ml (Fig. 3). In blood, the<br />

concentration <strong>of</strong> TGF-b reached the maximum at Day 7<br />

(45.1 ± 9.5 pg/ml).<br />

Fig. 2 Changes <strong>of</strong> cell distribution in BAL fluid after treatment<br />

SWCNTs (n = 4). Mice were treated with 100 lg/kg <strong>of</strong> SWCNTs<br />

and then killed on the designated Day (1, 7, 14, or 28). Total cells in<br />

the BAL fluid were quantified by hemocytometric counting a, and the<br />

distributions <strong>of</strong> alveolar macrophages, neutrophils, and lymphocytes<br />

were identified by their characteristic cell shapes b. Level <strong>of</strong> control<br />

group is mean ± SD <strong>of</strong> the values measured at each time point.<br />

*P \ 0.05; **P \ 0.01<br />

The concentration <strong>of</strong> collagen following administration<br />

<strong>of</strong> 100 lg/kg <strong>of</strong> SWCNT was also measured. As shown in<br />

Fig. 4, the concentration <strong>of</strong> collagen in the BAL fluid and<br />

in blood reached the maximum <strong>of</strong> 16.5 ± 0.4 ng/ml and<br />

928.7 ± 150.5 ng/ml at Days 14 and 7, respectively. The<br />

concentration <strong>of</strong> collagen in the control group was<br />

2.81 ± 0.00 ng/ml and 659.59 ± 79.47 ng/ml in BAL and<br />

blood, respectively.<br />

123


1126 Arch Toxicol (2011) 85:1121–1131<br />

Table 1 Changes in cytokine levels in BAL fluid after a <strong>single</strong> treatment <strong>of</strong> SWCNTs (n = 4)<br />

Control Day 1 Day 7 Day 14 Day 28<br />

IL-1 b 19.9 ± 12.7 64.1 ± 9.5* 73.4 ± 19.3* 82.9 ± 29.4* 89.0 ± 41.0<br />

TNF-a 16.4 ± 13.5 43.6 ± 12.8* 44.3 ± 4.4* 41.6 ± 8.8* 41.6 ± 6.8*<br />

IL-6 22.0 ± 5.8 115.6 ± 16.8** 137.2 ± 11.9** 151.3 ± 45.2** 129.2 ± 24.7**<br />

IL-2 5.1 ± 5.9 66.9 ± 23.0** 151.5 ± 27.0** 77.8 ± 3.5** 81.5 ± 22.9**<br />

IL-12 14.1 ± 2.9 49.1 ± 14.7* 45.7 ± 6.0** 45.5 ± 8.1* 52.6 ± 12.3*<br />

IFN-c 7.0 ± 2.3 32.2 ± 6.1** 16.9 ± 3.6* 26.1 ± 7.5* 23.5 ± 3.6*<br />

IL-4 20.2 ± 1.9 59.8 ± 7.2* 47.6 ± 1.9** 50.7 ± 6.9* 48.6 ± 4.3*<br />

IL-5 15.4 ± 1.4 35.8 ± 8.4* 45.8 ± 5.8** 36.4 ± 8.0* 41.5 ± 10.7*<br />

IL-10 64.6 ± 2.7 100.0 ± 17.0* 97.0 ± 13.8* 93.7 ± 11.1* 120.1 ± 45.9<br />

IL-13 13.1 ± 3.8 44.3 ± 5.3* 43.1 ± 4.1* 50.83 ± 4.4** 56.9 ± 4.6**<br />

IL-17 6.1 ± 3.9 36.2 ± 7.4* 38.4 ± 8.0* 45.2 ± 23.5 48.2 ± 30.1<br />

BAL fluid was harvested and pooled (400 ll per mouse) at Days 1, 7, 14, and 28 after treatment<br />

Cytokine concentrations were determined using ELISA kits. Level <strong>of</strong> control group was calculated as mean ± SD <strong>of</strong> the values obtained from<br />

4 mice per time point. * P \ 0.05; ** P \ 0.01<br />

Table 2 Changes in cytokine levels in the blood after a <strong>single</strong> treatment <strong>of</strong> SWCNTs (n = 4)<br />

Control Day 1 Day 7 Day 14 Day 28<br />

IL-1 b ND ND ND ND 1.2 ± 0.0<br />

TNF-a ND ND ND ND ND<br />

IL-6 ND 4.5 ± 0.1** 26.6 ± 2.6** 12.6 ± 0.8** ND<br />

IL-2 ND ND ND ND ND<br />

IL-12 38.1 ± 2.3 58.5 ± 3.8* 46.5 ± 0.9* 76.1 ± 22.5 52.4 ± 16.9<br />

IFN-c ND ND ND ND ND<br />

IL-4 ND ND ND ND ND<br />

IL-5 ND ND ND ND 4.15 ± 0.56<br />

IL-10 ND ND ND ND ND<br />

IL-13 1.5 ± 0.0 2.7 ± 0.0 3.3 ± 0.0 1.9 ± 0.0 2.4 ± 0.0<br />

IL-17 52.1 ± 1.5 85.3 ± 31.6 62.3 ± 8.3 98.5 ± 1.9* 62.4 ± 12.5<br />

Serum was harvested and pooled (400 ll per mouse) at Days 1, 7, 14, and 28 after treatment<br />

Cytokine concentrations were determined using ELISA kits. Level <strong>of</strong> control group was calculated as mean ± SD <strong>of</strong> the values obtained from<br />

4 mice per time point. * P \ 0.05; ** P \ 0.01<br />

Lymphocyte phenotype<br />

A <strong>single</strong> <strong>instillation</strong> <strong>of</strong> 100 lg/kg SWCNTs significantly<br />

increased the proportion <strong>of</strong> T cells in blood at Day 1.<br />

However, the proportion <strong>of</strong> B cells in blood lymphocytes<br />

had rapidly increased by 85.80 ± 5.20%, 70.33 ± 2.68%,<br />

and 62.80 ± 5.81% at Days 7, 14, and 28, respectively,<br />

compared to 59.03 ± 0.95% <strong>of</strong> the control (Table 3;<br />

Fig. 5a, b). The ratio <strong>of</strong> CD4/CD8 T cells also exhibited<br />

a significant change compared to the control group<br />

(Table 3; Fig. 5c, d). The ratio <strong>of</strong> CD4/CD8 T cells in<br />

the blood lymphocytes <strong>of</strong> the SWCNT-treated group was<br />

4.05 ± 0.11, 3.31 ± 0.09, 3.75 ± 0.03, and 4.98 ± 0.19 at<br />

Days 1, 7, 14, and 28, respectively, while that <strong>of</strong> the control<br />

group was 4.44 ± 0.20.<br />

Histopathology <strong>of</strong> lung tissue<br />

We observed histopathological changes in the lung, brain,<br />

and thymus. Figure 6 and Table 4 show the histopathological<br />

changes in the lung tissue following treatment with<br />

SWCNTs. SWCNTs induced fibrotic pathology by Day 7,<br />

but the severity <strong>of</strong> the symptoms decreased with time. At<br />

Day 1, phagocytosis by macrophages was observed, and<br />

cell infiltration and fibrosis were observed in one <strong>of</strong> the<br />

samples tested. By Day 7, the number <strong>of</strong> individuals<br />

showing cell infiltration and fibrosis had increased. No<br />

pathological symptom, including phagocytosis, cell infiltration,<br />

or fibrosis, was observed at Day 28. No pathological<br />

changes were observed in the brain and thymus (data<br />

not shown).<br />

123


Arch Toxicol (2011) 85:1121–1131 1127<br />

Protein expression by SWCNT<br />

As shown in Fig. 7, the expression <strong>of</strong> the apoptosis-related<br />

proteins p53 and caspase-3 and the inflammatory proteins<br />

COX-2 and iNOS showed maximum induction at Day 7<br />

and Day 14, respectively. The expression <strong>of</strong> the tissue<br />

damage-related proteins COL1A1, MMP-2, MMP-9, and<br />

mesothelin also markedly increased until Day 28 after<br />

treatment.<br />

Discussion<br />

Fig. 3 Levels <strong>of</strong> TGF-b in BAL fluid and in blood after a <strong>single</strong><br />

<strong>instillation</strong> <strong>of</strong> SWCNTs (n = 4). BAL fluid and serum were harvested<br />

and pooled at Days 1, 7, 14, and 28 after treatment with 100 lg/kg <strong>of</strong><br />

SWCNTs. The TGF-b concentration in each sample was determined<br />

using commercially available kits. Level <strong>of</strong> control group is<br />

mean ± SD <strong>of</strong> the values measured at each time point. *P \ 0.05;<br />

**P \ 0.01<br />

Fig. 4 Levels <strong>of</strong> collagen in BAL fluid and in blood after a <strong>single</strong><br />

treatment with SWCNTs (n = 4). BAL fluid and serum were<br />

harvested and pooled at Days 1, 7, 14, and 28 after treatment with<br />

100 lg/kg <strong>of</strong> SWCNTs. The collagen concentration in each sample<br />

was determined using commercially available kits. Level <strong>of</strong> control<br />

group is mean ± SD <strong>of</strong> the values measured at each time point.<br />

*P \ 0.05<br />

Human beings are constantly exposed to environmental<br />

pollutants since birth. The development <strong>of</strong> new technology<br />

and materials has improved the quality <strong>of</strong> life for humans,<br />

but it also entails the possibility <strong>of</strong> exposure to new<br />

materials. One <strong>of</strong> the recent hot issues is nanoparticles,<br />

including CNTs. Extensive toxicological studies with in<br />

vitro and in vivo models are required for the safe application<br />

<strong>of</strong> nanoparticles. In our study, 2 <strong>of</strong> the 64 mice died<br />

at Day 3 instilled with SWCNTs, one death due to pigmented<br />

histiocytes and the other due to a hemorrhage in the<br />

lung.<br />

Although the raw materials are same, nanoparticles can<br />

have different physicochemical properties according to the<br />

manufacturing process. The toxicity <strong>of</strong> nanoparticles<br />

depends on various parameters, such as the purity <strong>of</strong> the<br />

samples, structure, size distribution, surface area, surface<br />

chemistry, surface charge, and agglomeration state (Kagan<br />

et al. 2006; Murray et al. 2009; Herzog et al. 2009). In one<br />

study, exposure <strong>of</strong> JB6P cells (murine epidermal cells)<br />

to unpurified SWCNT (30% iron) resulted in the production<br />

<strong>of</strong> ESR detectable hydroxyl radicals and caused a<br />

significant dose-dependent activation <strong>of</strong> the transcription<br />

factor, AP-1. However, no significant changes in AP-1<br />

activation were detected when partially purified SWCNT<br />

(0.23% iron) was introduced to the cells (Murray et al.<br />

2009). In another study, SWCNT dispersed in dipalmitoylphosphatidylcholine,<br />

the main component <strong>of</strong> lung lining<br />

fluid, suppressed the production <strong>of</strong> inflammatory<br />

mediators, such as IL-8, IL-6, TNF-a, and MCP-1, in<br />

human lung epithelial cells (Herzog et al. 2009). In this<br />

study, we used SWCNT dispersed by ultra-sonication and<br />

used SDS as a surfactant.<br />

Nanoparticles entering the body trigger the innate<br />

immune system. This is usually resolved by macrophages<br />

phagocyting the nanoparticles. Some researchers have<br />

suggested that SWCNTs impair the phagocytic function <strong>of</strong><br />

macrophages in both a cytotoxic and a non-cytotoxic state<br />

(Witasp et al. 2009; Nygaard et al. 2009; Shvedova et al.<br />

2008a, b). For example, SWCNTs with low metal impurity<br />

content did not exert direct cytotoxic effects on human<br />

123


1128 Arch Toxicol (2011) 85:1121–1131<br />

Table 3 Lymphocyte phenotypes in blood after treatment with SWCNTs (n = 4)<br />

Control Day 1 Day 7 Day 14 Day 28<br />

NK cell 0.13 ± 0.06 0.63 ± 0.15 0.50 ± 0.10 0.63 ± 0.19 0.37 ± 006<br />

NKT cell 0.03 ± 0.06 0.13 ± 0.06 0.10 ± 0.00 0.18 ± 0.05 0.27 ± 0.06<br />

B cell 59.03 ± 0.95 51.63 ± 5.58 85.80 ± 5.20* 70.33 ± 2.68* 62.80 ± 5.81<br />

T cell 40.77 ± 0.95 47.63 ± 5.71 13.60 ± 5.16** 28.88 ± 2.81* 36.57 ± 5.80<br />

CD4/CD8 4.44 ± 0.20 4.05 ± 0.11 3.31 ± 0.09* 3.75 ± 0.03* 4.98 ± 0.19<br />

Level <strong>of</strong> control group is mean ± SD <strong>of</strong> the values measured at each time point. * P \ 0.05; ** P \ 0.01<br />

Fig. 5 Lymphocyte phenotypes<br />

in blood after treatment with<br />

SWCNTs. The experiment was<br />

repeated 4 times using samples<br />

pooled by n = 4 and<br />

representative data are shown.<br />

Control samples were matched<br />

for each fluorochrome:<br />

a lymphocyte <strong>of</strong> control,<br />

b lymphocyte at Day 7,<br />

c T subtype <strong>of</strong> control, and<br />

d T subtype at Day 7. a and<br />

b consist <strong>of</strong> Q1-NK, Q2-NKT,<br />

Q3-B, and Q4-T cells<br />

monocyte-derived macrophages (HMDM). However,<br />

SWCNT suppressed the chemotaxis <strong>of</strong> primary human<br />

monocytes. Moreover, macrophage engulfment <strong>of</strong> apoptotic<br />

target cells was significantly impaired following the preincubation<br />

<strong>of</strong> HMDM with SWCNT at non-cytotoxic<br />

concentrations (Witasp et al. 2009).<br />

Other researchers reported the inflow <strong>of</strong> immune cells<br />

into target sites by the phagocytes <strong>of</strong> SWCNTs (Shvedova<br />

et al. 2005; Inoue et al. 2008). The pharyngeal aspiration <strong>of</strong><br />

SWCNTs (about 500 lg/kg) induced an early neutrophil<br />

accumulation in BAL fluid at Day 1, followed by lymphocytes<br />

at Day 3 and macrophages at Day 7 (Shvedova<br />

et al. 2005). The concentration <strong>of</strong> MIP-1a and MCP-1 in<br />

BAL fluid significantly increased by about 32.2-fold and<br />

10.7-fold, respectively, at 24 h after <strong>instillation</strong> with<br />

4 mg/kg SWCNT compared to the vehicle control. The<br />

number <strong>of</strong> total cells and neutrophils in BAL fluid also<br />

significantly increased by about 1.7-fold and 22.4-fold,<br />

respectively, compared to the vehicle control (Inoue et al.<br />

2008). In the present study, the number <strong>of</strong> total cells in the<br />

BAL fluid increased 1.7-fold, 3.9-fold, 4.8-fold, and 3.2-fold<br />

at Days 1, 7, 14, and 28, respectively, after <strong>instillation</strong> with<br />

100 lg/kg SWCNTs in relation to the control group. The<br />

inflow <strong>of</strong> neutrophils was markedly increased at Day 1, and<br />

the proportion <strong>of</strong> lymphocytes to the number <strong>of</strong> total cells<br />

increased by 5.1-fold, 9.5-fold, and 5.7-fold at Days 7, 14,<br />

and 28 after treatment, respectively, relative to the control.<br />

Pro-inflammatory cytokines, such as IL-1b, TNF-a, and<br />

IL-6, play an important role in the initiation <strong>of</strong> the immune<br />

system when it comes to removing xenobiotics. In prior<br />

reports, the secretion <strong>of</strong> TNF-a and IL-1b was elevated by<br />

16-fold and tenfold, respectively, at Day 1 by the pharyngeal<br />

aspiration <strong>of</strong> SWCNT (about 500 lg/kg) (Shvedova<br />

et al. 2005). The concentration <strong>of</strong> IL-1b in BAL fluid<br />

123


Arch Toxicol (2011) 85:1121–1131 1129<br />

Fig. 6 Histopathological changes in lung tissue after a <strong>single</strong> <strong>intratracheal</strong> treatment <strong>of</strong> SWCNTs. Lung sections were stained with hematoxylin<br />

and eosin stains (9200): a solvent control, b Day 1, c Day 7, and d Day 28. Tissue <strong>of</strong> control group was randomly chosen from each time point<br />

Table 4 Histopathological changes in lung tissue after a <strong>single</strong> <strong>intratracheal</strong> treatment <strong>of</strong> SWCNTs (n = 6)<br />

Control Day 1 Day 7 Day 14 Day 28<br />

F I F I F I F I F I<br />

Pigmented macrophage 4/6 ± or 4/6 ± or 5/6 ± or 2/6 ±<br />

Inflammatory cell 1/6 ± 3/6 ± or 3/6 ± or <br />

Fibrosis, interstitial 1/6 ± 3/6 ± 1/6 ±<br />

Micr<strong>of</strong>ibrillar material 1/6 ±<br />

Epithelial degeneration, bronchiole 1/6 ± 1/6 ±<br />

Tissue <strong>of</strong> control group was randomly chosen from each time point. F Frequency <strong>of</strong> the events, I Intensity <strong>of</strong> the events, ± minimal; mild<br />

significantly increased to about 6.8-fold at 24 h after<br />

treatment with 4 mg/kg <strong>of</strong> SWCNT compared to the<br />

vehicle control, but its level in the blood stream did not<br />

increase (Inoue et al. 2008). In this study, the concentration<br />

<strong>of</strong> IL-1b, TNF-a, and IL-6 rapidly increased by 3.2-fold,<br />

2.7-fold, and 5.2-fold relative to the control at Day 1 after<br />

<strong>instillation</strong> and remained at similar levels during the<br />

experimental period.<br />

In addition, macrophages secrete IL-2 to stimulate naïve<br />

T cells to trigger the adaptive immune system. Following<br />

this, activated T cells are differentiated into Th1 cells and<br />

Th2 cells. These differentiated cells secrete IFN-c and<br />

IL-10 to stimulate themselves or to inhibit the activation <strong>of</strong><br />

another, respectively. Th1 and Th2 cells have very different<br />

functions. Th2 cells are the most effective activators <strong>of</strong><br />

B cells, especially in primary responses, whereas Th1 cells<br />

123


1130 Arch Toxicol (2011) 85:1121–1131<br />

Con. 1 7 14 28<br />

β- Actin<br />

COX-2<br />

iNOS<br />

Caspase-3<br />

p53<br />

MMP-9<br />

MMP-2<br />

COL1A1<br />

Mesothelin<br />

Fig. 7 Changes in protein expression following treatment with<br />

SWCNTs. Results were confirmed by several separate experiments,<br />

and representative images are shown. Tissue <strong>of</strong> control group was<br />

pooled from all mice in the control group<br />

are crucial for activating macrophages. However, under<br />

many circumstances in vivo, there is a mixed Th1 and Th2<br />

response, and the overall effect is determined by the<br />

responses that are dominant. In this study, Th0-type cytokine<br />

(IL-2), Th1-type cytokines (IL-12, IFN-c), and Th2-<br />

type cytokines (IL-4, IL-5, IL-10, IL-13) were markedly<br />

increased at Day 1 in the BAL fluid and steadily remained<br />

at the up-regulated level during the experimental period.<br />

We hypothesize that the sustained increase <strong>of</strong> pro-inflammatory<br />

and Th1-type cytokine may be the effect <strong>of</strong> the<br />

sustained inflow <strong>of</strong> macrophages into damaged sites.<br />

Inoue et al. suggested that any histopathological opinion<br />

on the attack <strong>of</strong> fibrotic lesions was not observed after the<br />

administration <strong>of</strong> 4 mg/kg <strong>of</strong> SWCNTs. However, Shvedova<br />

et al. (2005) suggested that the fibrotic response<br />

proceeded with time, as the alveolar wall was significantly<br />

thicker at 60 days than it was at 28 days postexposure to<br />

roughly 2 mg/kg <strong>of</strong> SWCNTs. Moreover, the deposition <strong>of</strong><br />

collagen and elastin was observed in granulomatous<br />

regions, as well as in the areas distant from granulomas. In<br />

our study, the fibrotic lesions histopathologically peaked on<br />

Day 7, and the severity <strong>of</strong> the symptoms decreased with<br />

time. In addition, cell composition in lymphocytes shifted<br />

from T cell dominant at Days 1 and 28 to B cell dominant<br />

at Days 7 and 14. The population <strong>of</strong> B cells in lymphocytes<br />

increased to 1.45-fold and 1.19-fold at Day 7 and Day 14,<br />

respectively, compared to that <strong>of</strong> the control group. The<br />

ratio <strong>of</strong> cytotoxic T cells relative to helper T cells also<br />

significantly increased until Day 14 after treatment. Cytotoxic<br />

T cells are capable <strong>of</strong> inducing death in somatic<br />

tumor cells, infected cells, damaged cells, and dysfunctional<br />

cells through the release <strong>of</strong> cytotoxins, such as perforins,<br />

granzymes, and granulysin. The theory that early<br />

outbreak <strong>of</strong> fibrosis is caused by exposure to SWCNTs is<br />

also supported by the expression <strong>of</strong> inflammatory response-<br />

(iNOS and COX-2), apoptosis- (p53 and caspase-3), and<br />

matrix damage- (MMP-9, MMP-2, and COL1A1) related<br />

protein, which peaked at Day 7 and Day 14.<br />

Some researchers also have suggested that the retention<br />

<strong>of</strong> serve is the initiator <strong>of</strong> mesothelial injury and inflammation<br />

that over time leads to pleural pathology, including<br />

mesothelioma (Pacurari et al. 2008; Takagi et al. 2008;<br />

Sakamoto et al. 2009; Donaldson et al. 2010). TGF-b<br />

exhibits three major activities: inhibition <strong>of</strong> cell proliferation,<br />

immunosuppression, and enhancement <strong>of</strong> the formation<br />

<strong>of</strong> the extracellular matrix (Lawrence 1996). Collagen<br />

is the main protein <strong>of</strong> connective tissue and is related to the<br />

repair <strong>of</strong> injured matrices (Kershenobich Stalnikowitz and<br />

Weissbrod 2003; Karsenty and Park 1995; Cutroneo 2007).<br />

It is known that the expression <strong>of</strong> mesothelin in normal<br />

tissue is limited to mesothelial cells, but is over-expressed<br />

in several human tumors, including mesothelioma and<br />

ovarian, lung, and pancreatic adenocarcinomas (Hassan<br />

and Ho 2008). In our study, the expression <strong>of</strong> mesothelin<br />

increased in a time-dependent manner. Furthermore, at Day<br />

28, although the fibrotic lesions were almost completely<br />

resolved, the secretion <strong>of</strong> TGF-b and collagen and the<br />

expression <strong>of</strong> p53 related to the repair <strong>of</strong> damaged DNA<br />

were still being up-regulated.<br />

Based on these findings, we suggest that a <strong>single</strong> <strong>intratracheal</strong><br />

<strong>instillation</strong> <strong>of</strong> SWCNTs may induce early lung<br />

fibrosis and subchronic tissue damage. Thus, sufficient<br />

verification <strong>of</strong> the toxicity <strong>of</strong> SWCNT is necessary before<br />

the expansion <strong>of</strong> its market size to avoid unforeseen<br />

consequences.<br />

Acknowledgments This work was supported by Ministry <strong>of</strong> Environment<br />

as the Eco-technopia 21 project and National Institute <strong>of</strong><br />

Environmental Research.<br />

References<br />

Chang K, Pastan I (1996) Molecular cloning <strong>of</strong> mesothelin, a<br />

differentiation antigen present on mesothelium, mesotheliomas,<br />

and ovarian cancers. Proc Natl Acad Sci USA 93(1):136–143<br />

123


Arch Toxicol (2011) 85:1121–1131 1131<br />

Chou CC, Hsiao HY, Hong QS, Chen CH, Peng YW, Chen HW,<br />

Yang PC (2008) Single-<strong>walled</strong> <strong>carbon</strong> <strong>nanotubes</strong> can induce<br />

pulmonary injury in mouse model. Nano Lett 8(2):437–445<br />

Cutroneo KR (2007) TGF-beta-induced fibrosis and SMAD signaling:<br />

oligo decoys as natural therapeutics for inhibition <strong>of</strong> tissue<br />

fibrosis and scarring. Wound Repair Regen 15(Suppl 1):S54–S60<br />

Donaldson K, Murphy FA, Duffin R, Poland CA (2010) Asbestos,<br />

<strong>carbon</strong> <strong>nanotubes</strong> and the pleural mesothelium: a review <strong>of</strong> the<br />

hypothesis regarding the role <strong>of</strong> long fibre retention in the<br />

parietal pleura, inflammation and mesothelioma. Part Fibre<br />

Toxicol 7:5<br />

Folkmann JK, Risom L, Jacobsen NR, Wallin H, L<strong>of</strong>t S, Møller P<br />

(2009) Oxidatively damaged DNA in rats exposed by oral<br />

gavage to C60 fullerenes and <strong>single</strong>-<strong>walled</strong> <strong>carbon</strong> <strong>nanotubes</strong>.<br />

Environ Health Perspect 117(5):703–708<br />

Hassan R, Ho M (2008) Mesothelin targeted cancer immunotherapy.<br />

Eur J Cancer 44(1):46–53<br />

Herzog E, Byrne HJ, Casey A, Davoren M, Lenz AG, Maier KL,<br />

Duschl A, Oostingh GJ (2009) SWCNT suppress inflammatory<br />

mediator responses in human lung epithelium in vitro. Toxicol<br />

Appl Pharmacol 234(3):378–390<br />

Inoue K, Takano H, Koike E, Yanagisawa R, Sakurai M, Tasaka S,<br />

Ishizaka A, Shimada A (2008) Effects <strong>of</strong> pulmonary exposure to<br />

<strong>carbon</strong> <strong>nanotubes</strong> on lung and systemic inflammation with<br />

coagulatory disturbance induced by lipopolysaccharide in mice.<br />

Exp Biol Med (Maywood) 233(12):1583–1590<br />

Kagan VE, Tyurina YY, Tyurin VA, Konduru NV, Potapovich AI,<br />

Osipov AN, Kisin ER, Schwegler-Berry D, Mercer R, Castranova<br />

V, Shvedova AA (2006) Direct and indirect effects <strong>of</strong><br />

<strong>single</strong> <strong>walled</strong> <strong>carbon</strong> <strong>nanotubes</strong> on RAW 264.7 macrophages:<br />

role <strong>of</strong> iron. Toxicol Lett 165(1):88–100<br />

Karsenty G, Park RW (1995) Regulation <strong>of</strong> type I collagen genes<br />

expression. Int Rev Immunol 12(2–4):177–185<br />

Kershenobich Stalnikowitz D, Weissbrod AB (2003) Liver fibrosis<br />

and inflammation. A review. Ann Hepatol 2(4):159–163<br />

Lam CW, James JT, McCluskey R, Arepalli S, Hunter RL (2006) A<br />

review <strong>of</strong> <strong>carbon</strong> nanotube toxicity and assessment <strong>of</strong> potential<br />

occupational and environmental health risks. Crit Rev Toxicol<br />

36(3):189–217<br />

Lawrence DA (1996) Transforming growth factor-beta: a general<br />

review. Eur Cytokine Netw 7(3):363–374<br />

Manna SK, Sarkar S, Barr J, Wise K, Barrera EV, Jejelowo O, Rice-<br />

Fichtg AC, Ramesh GT (2005) Single-<strong>walled</strong> <strong>carbon</strong> nanotube<br />

induces oxidative stress and activates nuclear transcription factorkappaB<br />

in human keratinocytes. Nano Lett 5(9):1676–1684<br />

Maynard AD, Baron PA, Foley M, Shvedova AA, Kisin ER,<br />

Castranova V (2004) Exposure to <strong>carbon</strong> nanotube material:<br />

aerosol release during the handling <strong>of</strong> unrefined <strong>single</strong>-<strong>walled</strong><br />

<strong>carbon</strong> nanotube material. J Toxicol Environ Health A<br />

67(1):87–107<br />

Murray AR, Kisin E, Leonard SS, Young SH, Kommineni C, Kagan<br />

VE, Castranova V, Shvedova AA (2009) Oxidative stress and<br />

inflammatory response in dermal toxicity <strong>of</strong> <strong>single</strong>-<strong>walled</strong><br />

<strong>carbon</strong> <strong>nanotubes</strong>. Toxicology 257(3):161–171<br />

Nygaard UC, Hansen JS, Samuelsen M, Alberg T, Marioara CD,<br />

Løvik M (2009) Single-<strong>walled</strong> and multi-<strong>walled</strong> <strong>carbon</strong> <strong>nanotubes</strong><br />

promote allergic immune responses in mice. Toxicol Sci<br />

109(1):113–123<br />

Pacurari M, Yin XJ, Zhao J, Ding M, Leonard SS, Schwegler-Berry<br />

D, Ducatman BS, Sbarra D, Hoover MD, Castranova V,<br />

Vallyathan V (2008) Raw <strong>single</strong>-wall <strong>carbon</strong> <strong>nanotubes</strong> induce<br />

oxidative stress and activate MAPKs, AP-1, NF-kappaB, and<br />

Akt in normal and malignant human mesothelial cells. Environ<br />

Health Perspect 116(9):1211–1217<br />

Park EJ, Cho WS, Jeong J, Yi J, Choi K, Park K (2009a) Proinflammatory<br />

and potential allergic responses resulting from B<br />

cell activation in mice treated with multi-<strong>walled</strong> <strong>carbon</strong> <strong>nanotubes</strong><br />

by <strong>intratracheal</strong> <strong>instillation</strong>. Toxicology 259:113–121<br />

Park EJ, Yoon J, Choi K, Yi J, Park K (2009b) Induction <strong>of</strong> chronic<br />

inflammation in mice treated with titanium dioxide nanoparticles<br />

by <strong>intratracheal</strong> <strong>instillation</strong>. Toxicology 260:37–46<br />

Sakamoto Y, Nakae D, Fukumori N, Tayama K, Maekawa A, Imai K,<br />

Hirose A, Nishimura T, Ohashi N, Ogata A (2009) Induction <strong>of</strong><br />

mesothelioma by a <strong>single</strong> intrascrotal administration <strong>of</strong> multiwall<br />

<strong>carbon</strong> nanotube in intact male Fischer 344 rats. J Toxicol<br />

Sci 34(1):65–76<br />

Sharma CS, Sarkar S, Periyakaruppan A, Barr J, Wise K, Thomas R,<br />

Wilson BL, Ramesh GT (2007) Single-<strong>walled</strong> <strong>carbon</strong> <strong>nanotubes</strong><br />

induces oxidative stress in rat lung epithelial cells. J Nanosci<br />

Nanotechnol. 7(7):2466–2472<br />

Shvedova AA, Kagan VE (2010) The role <strong>of</strong> nanotoxicology in<br />

realizing the ‘helping without harm’ paradigm <strong>of</strong> nanomedicine:<br />

lessons from studies <strong>of</strong> pulmonary effects <strong>of</strong> <strong>single</strong>-<strong>walled</strong><br />

<strong>carbon</strong> <strong>nanotubes</strong>. J Intern Med 267(1):106–118<br />

Shvedova AA, Kisin ER, Mercer R, Murray AR, Johnson VJ,<br />

Potapovich AI, Tyurina YY, Gorelik O, Arepalli S, Schwegler-<br />

Berry D, Hubbs AF, Antonini J, Evans DE, Ku BK, Ramsey D,<br />

Maynard A, Kagan VE, Castranova V, Baron P (2005) Unusual<br />

inflammatory and fibrogenic pulmonary responses to <strong>single</strong><strong>walled</strong><br />

<strong>carbon</strong> <strong>nanotubes</strong> in mice. Am J Physiol Lung Cell Mol<br />

Physiol 289(5):L698–L708<br />

Shvedova AA, Fabisiak JP, Kisin ER, Murray AR, Roberts JR,<br />

Tyurina YY, Antonini JM, Feng WH, Kommineni C, Reynolds J,<br />

Barchowsky A, Castranova V, Kagan VE (2008a) Sequential<br />

exposure to <strong>carbon</strong> <strong>nanotubes</strong> and bacteria enhances pulmonary<br />

inflammation and infectivity. Am J Respir Cell Mol Biol<br />

38(5):579–590<br />

Shvedova AA, Kisin ER, Murray AR, Kommineni C, Castranova V,<br />

Fadeel B, Kagan VE (2008b) Increased accumulation <strong>of</strong><br />

neutrophils and decreased fibrosis in the lung <strong>of</strong> NADPH<br />

oxidase-deficient C57BL/6 mice exposed to <strong>carbon</strong> <strong>nanotubes</strong>.<br />

Toxicol Appl Pharmacol 231(2):235–240<br />

Silva IA, Graber J, Nyland JF, Silbergeld EK (2005) In vitro HgCl 2<br />

exposure <strong>of</strong> immune cells at different stages <strong>of</strong> maturation:<br />

effects on phenotype and function. Environ Res 98:341–348<br />

Song Y, Li X, Du X (2009) Exposure to nanoparticles is related to<br />

pleural effusion, pulmonary fibrosis and granuloma. Eur Respir J<br />

34(3):559–567<br />

Takagi A, Hirose A, Nishimura T, Fukumori N, Ogata A, Ohashi N,<br />

Kitajima S, Kanno J (2008) Induction <strong>of</strong> mesothelioma in p53 ±<br />

mouse by intraperitoneal application <strong>of</strong> multi-wall <strong>carbon</strong><br />

nanotube. J Toxicol Sci 33(1):105–116<br />

Vendrame M, Gemma C, Pennypacker KR, Bickford PC, Davis SC,<br />

Sanberg PR, Willing AE (2006) Cord blood rescues strokeinduced<br />

changes in splenocyte phenotype and function. Exp<br />

Neurol 199:191–200<br />

Warheit DB, Laurence BR, Reed KL, Roach DH, Reynolds GA,<br />

Webb TR (2004) Comparative pulmonary toxicity assessment<br />

<strong>of</strong> <strong>single</strong>-wall <strong>carbon</strong> <strong>nanotubes</strong> in rats. Toxicol Sci<br />

77(1):117–125<br />

Wei G, Zhou H, Liu Z, Song Y, Wang L, Sun L, Li Z (2005) One-step<br />

synthesis <strong>of</strong> silver nanoparticles, nanorods, and nanowires on the<br />

surface <strong>of</strong> DNA network. J Phys Chem B 109:8738–8743<br />

Witasp E, Shvedova AA, Kagan VE, Fadeel B (2009) Single-<strong>walled</strong><br />

<strong>carbon</strong> <strong>nanotubes</strong> impair human macrophage engulfment <strong>of</strong><br />

apoptotic cell corpses. Inhal Toxicol 21(Suppl 1):131–136<br />

Yang CM, Park JS, An KH, Lim SC, Seo K, Kim B, Park KA, Han S,<br />

Park CY, Lee YH (2005) Selective removal <strong>of</strong> metallic <strong>single</strong><strong>walled</strong><br />

<strong>carbon</strong> <strong>nanotubes</strong> with small diameters by using nitric<br />

and sulfuric acids. J Phys Chem B 109(41):19242–19248<br />

Yu X, Zhang J, Choi W, Choi JY, Kim JM, Gan L, Liu Z (2010) Nano<br />

Lett 10(9):3343–3349<br />

123

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!