10113452131822Toxicol Mech MethodsToxicol. Mech. MethodsToxicology mechanisms and methods1537-65161537-652423194015459347710.3109/15376516.2012.753967HHSPA701765ArticlePulmonary toxicity and fibrogenic response of carbon nanotubesMankeAmruta1WangLiying2RojanasakulYon1Department of Pharmaceutical Sciences, West Virginia University, Morgantown, WV, USAPhysiology and Pathology Research Branch, National Institute for Occupational Safety and Health, Morgantown, WV, USAAddress for correspondence: Yon Rojanasakul, PhD, Department of Basic Pharmaceutical Sciences, West Virginia University, Morgantown, WV 26506, USA. Tel: (304)293-1476. Fax: (304)293-2576. yrojan@hsc.wvu.edu289201516120133201305102015233196206

Carbon nanotubes (CNTs) have been a subject of intensive research for a wide range of applications. However, because of their extremely small size and light weight, CNTs are readily inhaled into human lungs resulting in increased rates of pulmonary disorders, most notably fibrosis. Several studies have demonstrated the fibrogenic effects of CNTs given their ability to translocate into the surrounding areas in the lung causing granulomatous lesions and interstitial and sub-pleural fibrosis. However, the mechanisms underlying the disease process remain obscure due to the lack of understanding of the cellular interactions and molecular targets involved. Interestingly, certain physicochemical properties of CNTs have been shown to affect their respiratory toxicity, thereby becoming significant determinants of fibrogenesis. CNT-induced fibrosis involves a multitude of cell types and is characterized by the early onset of inflammation, oxidative stress and accumulation of extracellular matrix. Increased reactive oxygen species activate various cytokine/growth factor signaling cascades resulting in increased expression of inflammatory and fibrotic genes. Profibrotic growth factors and cytokines contribute directly to fibroblast proliferation and collagen production. Given the role of multiple players during the pathogenesis of CNT-induced fibrosis, the objective of this review is to summarize the key findings and discuss major cellular and molecular events governing pulmonary fibrosis. We also discuss the physicochemical properties of CNTs and their effects on pulmonary toxicities as well as various biological factors contributing to the development of fibrosis.

Angiogenesisepithelial mesenchymal transitioninflammationlung fibrosisoxidative stress
Introduction

In recent years, a variety of nanomaterials have revolutionized the industrial field with their rapidly emerging applications in the areas of biotechnology, electronics, medicinal drug delivery, cosmetics, material science and aerospace engineering. Among the pool of recently developed nanomaterials, carbon nanotubes (CNTs) have generated great interest commercially with their unique physicochemical properties such as high tensile strength and conductivity (Donaldson et al., 2010; Maynard et al., 2004). With abundant novel applications, the CNT market has been projected to expand substantially within the next decade. However, such massive production is fraught with concerns for environmental and occupational exposure. According to a National Science Foundation (NSF) report, about 6 million workers will be involved with the nanotechnology industry by 2020 including 2 million within the United States, thus indicating a possible prevalence (Patel, 2011). Human exposures to manufactured nanomaterials are most likely to be observed in workers than the general population (Bergamaschi, 2009). A study reported exposure to polyacrylate paint containing nanoparticle within a group of female workers. Affected workers clinically presented with pleural effusions, progressive pulmonary fibrosis, pleural damage and death (Song et al., 2009). Moreover, a risk-assessment study on titanium dioxide nanomaterial observed occupational exposure within factory workers beyond the acceptable limits during the packaging process (DuPont™, 2008). However, nanomaterial exposure and dosimetry data are insufficient for humans owing to the difficulty in detection and accurate measurement tools for this unique and rapidly growing nanomaterial industry. The extraordinary properties of CNTs need to compete with reports of CNT-associated toxicities, thus indicating careful monitoring of human health and safety during their use. Depending upon the type of exposure, CNTs may penetrate the body through various routes such as the lungs and gut. CNTs are high aspect ratio nanomaterials (HARNs) having at least one of their dimensions of the order of 100 nm or less in size, as per the British Standards Institution (BSI) (2007). CNT structure facilitates their entry, deposition and residence in the lungs and pleura, resulting in incomplete phagocytosis and clearance from the lungs (Stella, 2011). Owing to their bio-persistent and non-biodegradable nature, and particularly the resemblance to needle-like asbestos fibers, CNTs are believed to induce biologically harmful effects (Lam et al., 2004). CNTs are similar to asbestos in their fibrous morphology, biopersistence, surface reactivity and the ability to translocate within the alveolar regions and the deeper pleura of the lung. Upon pulmonary exposure, CNTs generate an acute inflammatory response, activate several cell signaling pathways, induce genotoxicity, mesothelioma, diffuse interstitial fibrosis and granulomas similar to that observed in asbestos-exposed animals and humans. However, they differ in their chemical composition, surface charge and the ability to (i) enter mesothelial cells and (ii) induce direct fibrogenic effects (Jaurand et al., 2009; Nagai et al., 2011; Wang et al., 2010b).

Lung toxicity appears to be the major consequence of CNT exposure, ultimately contributing toward granuloma formation, epithelial hypertrophy and early onset of fibrosis (Lam et al., 2004; Muller et al., 2005; Shvedova et al., 2005; Warheit et al., 2004). Accumulating evidence in the literature demonstrates the fibrogenic potential of CNTs. Toxicity reports have indicated the ability of CNTs to translocate into the surrounding areas of the lung causing systemic toxicity, granulomatous lesions, interstitial and sub-pleural fibrosis (Jia et al., 2005; Park et al., 2011; Shvedova et al., 2008). However, the interactions of CNTs with the host at the molecular and cellular levels remain largely unknown. Identification of molecular targets and intracellular signaling is essential to the development of specific biomarkers for risk assessment and early detection of CNT-induced pathogenesis. The pathologic effects of CNTs are likely to be influenced by their physicochemical properties, thus we will first describe the physical and chemical properties of CNTs and their associated cellular toxicities. We will then focus on acute and chronic responses to CNT exposure involving oxidative stress, inflammation, genotoxicity and fibrosis. Finally, cellular mediators and mechanisms of fibrosis as well as biological factors influencing the fibrogenic process will be discussed.

Types and properties of CNTs

As per the British Standards Institute Report (2007), CNTs are HARNs having at least one of their dimensions, i.e. the diameter less than 100 nm whereas the lengths vary from a wide range of micrometers. CNTs are engineered nanomaterials made of graphene sheets that have been rolled into seamless cylindrical structure. They are manufactured mainly via arc discharge, chemical vapor deposition and laser ablation. All the three methods basically involve thermal elimination of carbon atoms from carbon sources including graphite, or gaseous carbon-bearing compounds such as CO, methane, ethylene or other hydrocarbons (Lam et al., 2006). Post-synthesis, CNTs are purified to eliminate residual organics such as soot or amorphous carbon and metals. They are classified into two main types known as single-walled carbon nanotubes (SWCNTs) and multi-walled carbon nanotubes (MWCNTs), both demonstrating outstanding chemical and thermal stability. MWCNTs comprise of several single-walled tubes layered onto each other. SWCNTs can be viewed as a single thick-graphite layer rolled into a cylindrical tube (Sinha & Yeow, 2005).

The nanoscale and large surface area of CNTs allow them to interact efficiently with cells, albeit in an undefined manner. Whether CNTs are inherently toxic or is it a wide array of external factors such as length, surface modification, degree of dispersion and the presence of metal impurities playing a role in CNT-induced toxicity is still a subject of intense investigations. Current literature reveals that CNTs based on their type, fiber length, dispersion status and functionality exert considerable variations in toxicities.

Physicochemical properties of CNTs affecting toxicities

Physicochemical factors alter the cytotoxicity of CNTs with respect to their cellular uptake, internalization, phagocytosis and clearance from the body (Borm et al., 2006; Oberdörster et al., 2005). For instance, even though the functionalization of CNTs may aid their solubility and dispersion, it has been speculated to facilitate their uptake and internalization into the systemic circulation. More detailed discussions of the effects of physicochemical properties of CNTs on their biological activities are provided below and are summarized in Tables 1 and 2.

Particle dispersion

The biopersistence of CNTs is critical in determining lung toxicity endpoints (Murray et al., 2012; Oberdörster et al., 2005). Factors such as dispersion and aggregation can determine the persistence of CNTs within the lung. Previous studies have shown that the distribution of CNTs within the lung can influence the degree of fibrosis (Muller et al., 2005). Poorly dispersed SWCNTs were found to be restricted to the proximal alveolar regions resulting in granulomatous lesions, whereas well-dispersed CNTs deposited deeper into the alveolar interstitial and pleural areas of the lung causing parenchymal granulomas and interstitial fibrosis (Mercer et al., 2008; Murray et al., 2012). A study comparing inhalation and pharyngeal aspiration of MWCNTs reported a significant increase in fibrosis after pharyngeal aspiration. MWCNT inhalation resulted in collagen deposition in peribronchial and interstitial areas (Mercer et al., 2011).

CNTs can induce direct fibrotic effects without any signs of inflammatory response depending upon their entry/deposition into the deeper lung tissue. For instance, the entry of SWCNTs into the alveolar interstitial space evades their macrophage engulfment (Shvedova et al., 2005). The ability of dispersed SWCNTs to enter the alveolar interstitium and stimulate resident fibroblasts has been suggested to play a key role in SWCNT-induced interstitial fibrosis (Mercer et al., 2008). Subsequent studies identified the mechanism for the direct fibrogenic effects of dispersed SWCNTs. Their findings demonstrated increased cell proliferation, collagen production and upregulation of matrix metalloproteinase (MMP)-2 and MMP-9 in the region surrounding interstitialized CNTs after 2 d of exposure without any signs of persistent inflammation (Wang et al., 2010a, 2011b).

Surface chemistry and functionalization

Surface reactivity of CNTs is a key property enhancing their applicability. During their interaction with biological tissues, surface chemistry plays a key role in determining the toxic responses. Surface charge present on acid-functionalized CNTs elicits inflammatory response. Functionalization of CNTs enhances the degree of oxidative stress and inflammation, thereby governing cytotoxicity in multiple cell lines including lung epithelial cells, lung tumor cells, bone marrow cells and lymphocytes (Bottini et al., 2006; Patlolla et al., 2010b; Saxena et al., 2007; Warheit et al., 2004). In contrast, some studies showed that functionalization of CNTs inversely affected their toxicity (Jain et al., 2011; Sayes et al., 2006; Zhang et al., 2007). It is likely that the effect of CNT functionalization on toxicity is functional group-specific and dependent on the toxicological outcomes such as cell death, DNA damage, fibrosis, which are poorly understood at present.

Particle dimension

Particle length can govern the penetration of CNTs in the interstitium or pleura of the lung. A number of studies have illustrated the effects of CNT length on inflammation and granuloma formation (Poland et al., 2008), cytotoxicity (Liu et al., 2012), oxidative stress (Kagan et al., 2010) and epithelial barrier function (Rotoli et al., 2009). Furthermore, the length can influence the retention and clearance of CNTs from the lung, thereby contributing to the asbestos-like responses in the pleural cavity of the lung leading to acute inflammation and progressive fibrosis (Murphy et al., 2011). The thickness of CNTs contributes to their inflammogenicity and carcinogenicity in mesothelial cells with thinner MWCNTs causing a more pronounced effect (Nagai et al., 2011).

Metal contaminants

The presence of transition metals has been shown to modify the respiratory toxicity of CNTs. Metal contaminants introduced into CNTs during their synthesis affect CNT-induced oxidative stress, inflammation and loss of cell viability (Warheit et al., 2004). The effect of metal contaminants on CNT-induced lung fibrosis has not been thoroughly demonstrated, although it is likely to play a key role. Studies suggest that Ni, Co and Fe are toxic and fibrogenic upon their interaction with CNTs (Lam et al., 2004; Pacurari et al., 2008; Pulskamp et al., 2007; Shvedova et al., 2003, 2005, 2008).

In addition to the physicochemical properties of CNTs described above, dose-metric parameters such as surface area and mass dose have been shown to be important in the toxic risk assessment of CNTs (Shvedova et al., 2012). There is a need for careful monitoring of CNT properties while documenting their toxicity. Hence, it is necessary to accurately assess the potential hazards of CNTs attributable to their properties in order to fully realize their novel applications.

Biodistribution of CNTs

Very few studies have addressed the systemic distribution of CNTs after being deposited in the lungs. Upon clearance from the spleen and liver, SWCNTs are eliminated from the blood circulation via renal excretion (Singh et al., 2006). Intratracheal instillation of MWCNTs results in their uptake via the lymphatic and phagocytic systems similar to asbestos fibers. However, MWCNTs are retained in the lungs 6 months post-exposure indicating their persistent nature (Elgrabli et al., 2008). Extra-pulmonary toxicities of MWCNTs have been reported following their translocation to the liver and kidney (Reddy et al., 2010), although the underlying mechanism is unclear. Alterations in membrane permeability due to pulmonary inflammation can lead to particle distribution extending beyond the lung (Zhu et al., 2009). MWCNT exposure increases the permeability of microvascular endothelium via increased cell migration, reactive oxygen species (ROS) response and actin remodeling (Pacurari et al., 2012). Further investigations of the biodistribution and pharmacokinetics of CNTs are essential in understanding their toxicities.

Biological activities CNTsOxidative stress

One of the most frequently reported toxicity endpoints is the formation of ROS, which can be either protective or harmful during biological interactions. Oxidative stress is an imbalance between the production of ROS and their elimination by the host’s defense systems. Oxidative stress amounts to DNA damage, lipid peroxidation and activation of signaling networks associated with loss of cell growth, fibrosis and carcinogenesis (Knaapen et al., 2004; Valko et al., 2006). Following the exposure to CNTs, ROS are induced intrinsically within the cell, extrinsically or indirectly via the effect of internalized CNTs on mitochondrial respiration. The critical factors driving CNT-induced ROS generation include active redox cycling on the surface of nanoparticles (NPs), oxidative functional groups on the NPs and NP-cell interactions, especially in the lungs where there is a rich pool of ROS producers like neutrophils and macrophages (Knaapen et al., 2004). CNT-induced oxidative stress is mainly followed by inflammation, cell injury, apoptosis and activation of cellular signaling pathways such as mitogen-activated protein kinase (MAPK) and nuclear factor (NF)-κB, which are implicated in the pathogenesis of lung fibrosis (Bonner, 2002, 2007). Several studies have demonstrated the role of CNTs in oxidative stress, one of the mechanisms being mitochondrial damage (He et al., 2011). MWCNT exposure induces ROS production in a variety of cell lines as well as in vivo (Clichici et al., 2012; He et al., 2011; Mitchell et al., 2007; Reddy et al., 2010). Likewise, studies have demonstrated SWCNT-induced ROS generation in multiple cell lines (Azad et al., 2012; Manna et al., 2005; Shvedova et al., 2012) and activation of intracellular signaling pathways including MAPK, Akt, AP-1 and NF-κB in mesothelial cells in a dose-dependent manner (Pacurari et al., 2008). These findings indicate that CNT-induced oxidative stress may serve as an important intermediate endpoint while assessing pulmonary toxicity of CNTs.

Inflammation

Inflammation is commonly observed upon inhalation of CNTs. Characterization of the inflammatory process upon CNT exposure is necessary since inflammation is associated with other pathologic disorders such as fibrosis and cancer. Given the interplay between the inflammatory response and ROS generation, both effects are closely linked and one leads to the other (Donaldson & Poland, 2012). Particle deposition in the lung causes recruitment of inflammatory cells that generate ROS, clastogenic factors and cytokines, either harming or stimulating resident lung cells (Bonner, 2007). Oxidative stress produced from CNT exposure activates pro-inflammatory transcription factors such as NF-κB, AP-1 and MAPK (Pacurari et al., 2008). The inflammatory cytokines tumor necrosis factor (TNF)-α and interleukin (IL)-1β are well-known mediators of fibrotic lung diseases by activating the pro-fibrotic transforming growth factor (TGF)-β and platelet-derived growth factor (PDGF), respectively. Such stimulation leads to differentiation of fibroblasts to myofibroblasts and their production of extracellular matrix (ECM) proteins (Bonner, 2007; Mangum et al., 2006; Sime et al., 1998). Several reports indicate that MWCNTs and SWCNTs induce a panel of pro-fibrotic inflammatory cytokines and chemokines in human lung cells including TNF-α and IL-8 (Brown et al., 2007), IL-1β, IL-6, IL-10 and monocyte chemoattractant protein (MCP)-1 (He et al., 2011), NLRP3 inflammasome (Pälomaki et al., 2011), IL-13/33 (Beamer et al., 2012) and inflammatory enzymes such as cyclooxygenase (COX)-2 and inducible nitric oxide synthase (iNOS) (Lee et al., 2012). Intratracheal and pharyngeal aspiration of CNTs induce robust inflammatory effects in vivo leading to the onset of fibrogenesis (Shvedova et al., 2008). Additionally, ineffective internalization of nano-sized CNTs by macrophages amounts to transient inflammation, suggesting that the rate of CNT elimination may be important in assessing the severity of the associated inflammatory responses (Dong et al., 2012; Kagan et al., 2010).

Genotoxicity

Given the association of inflammatory responses and lung fibrosis as well as the striking similarities between CNTs and asbestos, it is essential to assess the genotoxic potential of CNT exposure (Hubbard et al., 2000). Genotoxicity may be elicited either by the direct interaction of fibrous particles with the genetic material or by secondary damage from particle-induced ROS generation. CNT-induced sustained inflammation and oxidative stress can result in DNA damage and abnormal cell growth, possibly leading to carcinogenesis and fibrogenesis (Jaurand, 1997; Jaurand et al., 2009). A number of studies have provided evidence for the genotoxic effects of CNTs (Kisin et al., 2007, 2011; Patlolla et al., 2010a, b). Oxidative stress-dependent DNA breakage and DNA repair, and the activation of signaling pathways including poly ADP-ribose polymerase (PARP), AP-1, NF-κB, p38 and Akt were reported in human mesothelial cells exposed to SWCNTs (Pacurari et al., 2008). K-ras mutations were observed in mouse lungs upon SWCNT exposure (Shvedova et al., 2008). Chronic SWCNT exposure induced a genotypic change characterized by deregulated p53 amounting to apoptosis-resistant malignant phenotype (Wang et al., 2011a). MWCNTs cause mutations because of their clastogenic (DNA break) and aneuploidic (chromosomal loss) effects (Muller et al., 2008). Mutational changes such as micronuclei formation, DNA damage (Zhu et al., 2007) and p53-independent expression of p21 and cell cycle arrest (Zhang & Yan, 2012) upon MWCNT exposure have been reported in a variety of cell lines.

Permeability barrier function

Alterations in respiratory barrier function is of particular importance to CNT-induced toxicity since respiratory epithelial cells present a protective barrier against inhaled particles and constitute a major determinant of the interaction of the particles with other body compartments. Epithelial cells are responsible for the formation and maintenance of tight junction barrier, only permitting polarized secretory functions and preventing access to xenobiotics and pathogens (Schneeberger & Lynch, 2004). Consequently, respiratory barrier function plays a pivotal role in CNT-related inhalation hazards. MWCNTs have been shown to alter the paracellular permeability of airway epithelial cells by interfering with the formation of tight junctions. The permeability-altering effect of CNTs was shown to be dependent on fiber length and functionalization (Rotoli et al., 2008, 2009). However, the underlying mechanisms of alteration and its effect on CNT-induced pathogenesis have not been clearly elucidated.

CNT-induced pulmonary fibrosis

A variety of environmental and occupational agents including fibrous particles, metals, drugs and microbes are able to induce pulmonary fibrosis. During the development of fibrosis, several common cellular events occur including epithelial cell injury, infiltration of inflammatory cells, proliferation and transformation of fibroblasts into myofibroblasts, and synthesis and deposition of ECM (He et al., 2011). Unique to CNTs is their non-biodegradable and biopersistent nature which likely prolongs the fibrotic process. Multiple factors determine the severity and duration of CNT-induced fibrosis, which are discussed below and are summarized in Figure 1.

Role of ROS

ROS is widely known to be involved in epithelial cell injury and fibrogenesis (Inghilleri et al., 2006). CNT exposure results in ROS-dependent activation of several transcription factors and signaling pathways including NF-κB, signal transducer and activator of transcription (STAT)-1, MAPK and receptor tyrosine kinases (RTK), which are involved in the regulation of inflammation and fibrosis (Bonner, 2007). For instance, MWCNTs induce fibroblast to myofibroblast differentiation via ROS-dependent NF-κB activation (He et al., 2011), whereas SWCNTs induce collagen production and angiogenesis via ROS-dependent p38-MAPK activation (Azad et al., 2012). MAPK activation is also responsible for the enhancing effect of ROS on Transforming growth factor-beta (TGF-β)-mediated fibrogenic response in human lung cells (Thannickal et al., 2004). Furthermore, ROS has been shown to play a critical role in TGF-β-mediated fibroblast to myofibroblast differentiation with the differentiated cells serving as an additional source of ROS production (Thannickal & Fanburg, 1995). CNT-induced ROS generation is likely to be a result of frustrated phagocytosis which refers to the failure of the macrophage to entirely engulf the long fibers (<15 mm), thus resulting in an inflammatory condition (Donaldson et al., 2010).

Additionally, specific properties of CNTs such as metal contaminants including iron, cobalt, tungsten and vanadium as well as reactive groups on the CNT surface that have been attributed to pulmonary fibrotic response may induce oxidative stress (Antonini, 2003; Bonner et al., 2000; Shvedova et al., 2003; Warheit et al., 2004). Some of the common metal contaminants associated with pulmonary fibrosis include iron, cobalt, tungsten and vanadium.

Role of inflammation

Numerous inflammatory and pro-fibrotic mediators such as TNF-α, IL-1β and TGF-β have been implicated in the pathogenesis of fibrosis. Infiltration of immune cells such as eosinophils, neutrophils and macrophages results in tissue injury and loss of epithelial integrity, thus promoting tissue repair and fibrosis (Wallace et al., 2007). As mentioned earlier, CNT exposure initiates an inflammatory cascade of cytokines in association with oxidative stress. ROS-dependent activation of RTK, MAPK, Akt and NF-κB results in the expression of genes involved in inflammation and fibrosis. Upregulation of TNF-α stimulates TGF-β production, collagen deposition, ECM remodeling and angiogenesis during fibrosis (Sime et al., 1998). TGF-β further enhances fibrogenesis by activating connective tissue growth factor (CTGF). Similarly, IL-1β triggers the secretion of PDGF on lung fibroblasts, enhancing their proliferation (Lindroos et al., 1997). There are reports documenting inflammation-induced fibrosis following CNT exposure. For examples, MWCNTs induce TNF-α resulting in fibrosis in vivo (Muller et al., 2005). SWCNTs upon pharyngeal aspiration induce robust inflammation and an early onset of fibrogenic response in vivo characterized by the secretion of TNF-α, IL-1β and TGF-β as well as several biomarkers of oxidative stress (Shvedova et al., 2005). The time course of TNF-α and IL-1β corresponds to neutrophil influx, whereas that for TGF-β is dependent on macrophage recruitment. The extent of fibrosis is dose and time dependent (Shvedova et al., 2005). Inflammation-driven fibrosis causes granuloma formation associated with epithelial hypertrophy, alveolar thickening and interstitial fibrosis (Muller et al., 2005; Shvedova et al., 2008). Intratracheal instillation of SWCNTs results in early onset of lung fibrosis driven by the secretion of a panel of inflammatory cytokines (Park et al., 2011). In general, inflammation-mediated fibrogenesis can cause deterioration of pulmonary functions.

Role of pre-existing inflammation

Little has been reported about the risk and possibility of a fibrogenic response following CNT exposure in conditions with pre-existing inflammation. Studies have reported how bacterial-derived products modify CNT-related toxicities. In mice with prior bacterial infection, pharyngeal aspiration of SWCNTs promotes inflammatory response, collagen synthesis, reduces phagocytosis of bacteria by macrophages and bacterial clearance from the lungs, thereby increasing host susceptibility to lung fibrosis (Shvedova et al., 2008). Preexposure with lipopolysaccharide, an endotoxin, exaggerates the fibrotic effects of CNTs by (i) intensifying acute inflammatory response (Inoue et al., 2009), (ii) increasing PDGF and its receptor resulting in fibroblast chemotaxis and proliferation (Cesta et al., 2010) and by (iii) elevating the expression of TNF-α and IL-1β resulting in collagen synthesis and ECM deposition (Shvedova et al., 2008). Together, these results support the role of pre-existing inflammation in promoting CNT-induced lung fibrosis.

Role of angiogenesis

Angiogenesis is essential in the formation of new blood vessels, wound healing and tissue repair, thus important in fibrogenesis. Vascular endothelial growth factor (VEGF) regulates the angiogenic response by controlling the migration, proliferation and vasculature of endothelial cells. Initial studies demonstrated neovascularization leading to anastomoses between the systemic and pulmonary microvasculature of patients with pulmonary fibrosis (Turner-Warwick, 1963). Subsequent studies showed structural and functional changes in the vasculature involving the formation of new vessels, remodeling of alveolar capillaries, and imbalance between the angiogenic and angiostatic chemokines (Keane, 2004). These alterations signify the role of altered vascular homeostasis in the pathogenesis of lung fibrosis (Peão et al., 1994; Turner-Warwick, 1963). Interestingly, aberrant expression of VEGF has been reported during idiopathic myelofibrosis and diffuse lung fibrosis (Chaudhary et al., 2007). Besides being a pro-fibrogenic cytokine, TGF-β has the ability to promote angiogenesis. VEGF and TGF-β interact closely with respect to angiogenic and fibrogenic effects (Ferrari et al., 2009). A recent in vitro study showed that SWCNT-induced fibroblast proliferation and collagen production is mediated by VEGF and TGF-β1 (Azad et al., 2012). This study also suggested a positive feedback loop between TGF-β and VEGF during angiogenesis, which promotes the fibrotic process after SWCNT treatment. The observed fibrogenic and angiogenic effects were found to be mediated by ROS-dependent p38 MAPK activation. Inhibitors of ROS, p38 MAPK, VEGF and TGF-β strongly inhibited the pathologic effects of SWCNT. Further in vivo studies are necessary to substantiate the role of these cellular mediators in CNT-induced angiogenesis and fibrosis.

Role of epithelial mesenchymal transition

Epithelial mesenchymal transition (EMT), a process characterized by the transition of fully differentiated epithelial cells to a mesenchymal phenotype, has been suggested to play a key role in fibrosis by serving as a cellular source of resident fibroblasts/myofibroblasts. However, whether or not EMT is a major source of these interstitial lung cells during fibrosis in vivo is still unclear. EMT is brought about by various external stimuli including growth factors, cytokines and hormones (Willis & Borok, 2007). EMT promotes fibrogenesis by causing a loss of epithelial barrier that acts as a defense during advanced stage fibrosis (Chapman, 2011) and by generating mesenchymal cells that expand the pool of fibroblasts/myofibroblasts. In transgenic mice in which the fate of alveolar epithelial cells (AECs) can be tracked, it was found that vimentin-positive mesenchymal cells in injured lungs were mostly of alveolar epithelial origin, suggesting epithelial cells as the main source of mesenchymal expansion (Kim et al., 2006). Epithelial cells from patients with idiopathic pulmonary fibrosis (IPF) also co-express epithelial and mesenchymal markers, supporting the involvement of EMT in fibrosis (Kim et al., 2006; Willis et al., 2005). Furthermore, AECs undergo EMT via stimulation with TGF-β during fibrosis and are an important source of myofibroblasts (Radisky et al., 2007). In contrast to the above findings, a lineage-tracing study in a murine bleomycin-induced lung fibrosis model showed that EMT was not the origin of myofibroblasts as evidenced by the lack of AECs expressing the mesenchymal markers (Rock et al., 2011). However, a recent study showed that such EMT required proper ECM microenvironment, notably type I collagen (DeMaio et al., 2012). Thus, several factors may influence EMT and its fibrotic outcome, including experimental conditions, type of stimuli and the difference between the murine models and human disease.

With regards to CNT-induced EMT, a recent in vivo study demonstrated that SWCNT exposure induced EMT of lung epithelial cells which contributed significantly to fibroblast expansion (Chang et al., 2012). SWCNT exposure also resulted in an activation of TGF-β/p-Smad2 and β-catenin in epithelial cells, which was suggested to play a key role in SWCNT-induced EMT. Given the paucity of data on CNT-induced EMT, future studies are warranted to elucidate the role of EMT and related signaling pathways in CNT-induced lung fibrosis.

Profibrogenic mediators

A variety of fibrogenic cytokines and growth factors are involved in the regulation of pulmonary fibrosis. Key cytokines and growth factors implicated in the pathogenesis of CNT-induced fibrosis are highlighted below.

Transforming growth factor-beta

In the lung, TGF-β is produced mainly from alveolar macrophages and epithelial cells in response to various fibrogenic stimuli (Agostini & Gurrieri, 2006). Both SWCNTs and MWCNTs have been shown to induce TGF-β secretion in vitro (Azad et al., 2012; Mishra et al., 2012; Wang et al., 2010b) and in vivo (Shvedova et al., 2005; Wang et al., 2011b). In addition to its well-established role in EMT and angiogenesis, TGF-β can directly stimulate fibroblasts to produce collagen. Clinical evidence demonstrated that TGF-β is elevated in the lung of IPF patients and rodent models. TGF-β is capable of inducing cytoskeletal rearrangements and differentiation of fibroblasts to myofibroblasts, resulting in increased ECM production and deposition during fibrosis (Leask & Abraham, 2004). Additionally, a positive feedback loop exists between TGF-β and ROS generation. TGF-β stimulates ROS by reducing antioxidant levels in many lung cells, whereas ROS can activate TGF-β to mediate its fibrotic effects. Oxidative stress can thus significantly initiate a cascade of fibrogenic responses via TGF-β signaling (Thannickal & Fanburg, 1995).

Platelet-derived growth factor

PDGF is a key mediator of fibroblast proliferation and chemotaxis (Lindroos et al., 1997). During NP-induced fibrosis, PDGF isoforms induce fibroblast to myofibroblast differentiation and proliferation, thus increasing the immature collagenous tissue in the lung (Bonner, 2002; Mangum et al., 2006). PDGF has been previously implicated in asbestosis and coal dust-induced pneumoconiosis (Vanhee et al., 1994). Exposure of macrophages to MWCNTs induced PDGF secretion, mitochondrial damage and oxidative stress, which promote fibrogenesis (He et al., 2011). PDGF was also shown to be upregulated during SWCNT-induced interstitial fibrosis (Mangum et al., 2006). These studies indicate that coordinated activation of PDGF can drive CNT-induced fibrosis.

Osteopontin

Osteopontin (OPN), a chemotactic cytokine secreted mainly by macrophages, exhibits leukocyte chemotaxis during tissue repair. It induces proliferation and migration of epithelial cells and fibroblasts, and is known to play a key role in ECM remodeling during lung fibrosis (Pardo et al., 2005). Its pro-fibrotic role is implicated by the induction of MMP-7 during SWCNT-induced lung inflammation (Mangum et al., 2006). However, direct evidence for the role of OPN in CNT-induced fibrosis is still lacking.

Matrix metalloproteinases

The MMP family of enzymes plays an important role during wound healing and ECM repair. They are present in moderate levels during normal conditions; however, upon cellular injury and inflammation, their levels rise which helps to recruit immune cells to the site of injury. These fibrotic mediators along with TGF-β can potentially drive NP-induced fibrogenic response (Chakrabarti & Patel, 2005). Recent reports demonstrated the role of MMPs in CNT-induced fibrogenesis. For instance, CNTs induced MMP-9 and in conjunction with TGF-β stimulated fibroblast proliferation, collagen synthesis and secretion of fibroblast growth factor (FGF)-2 which is a potent angiogenic factor and key stimulant for fibroblast proliferation and collagen production (Khalil et al., 2005; Mishra et al., 2012; Wang et al., 2010b). These studies also indicated the early onset of fibrosis brought about by CNTs without persistent inflammation. Thus, activating fibroblast proliferation and collagen synthesis appears sufficient for the atypical rapid interstitial fibrosis caused by CNTs.

Conclusions

This review encompasses the acute and chronic pulmonary responses to CNT exposure and their linkage. The ability of CNTs to cause acute toxicities and chronic fibrotic effects depends on several physicochemical factors such as particle dimension, dispersion status, functionalization and the presence of transition metals. Understanding these factors will enable the design of safer CNT products and their utilization. The cytotoxic and fibrogenic effects of CNTs appear to be associated with their ability to induce oxidative stress and inflammatory and fibrogenic cytokines. There is a close connection between oxidative stress and inflammatory response, as well as cross-talk between inflammation and fibrosis as indicated by the multifunctional roles of the induced cytokines, e.g. TGF-β, PDGF and MMPs. Interestingly, however, there have been reports showing CNT-induced fibrosis with minimum inflammation and oxidative injury, suggesting alternative pathways and mechanisms of fibrosis. Apart from the molecular and cellular changes, other biological factors such as angiogenesis and EMT can also influence fibrosis. EMT may contribute to the increased fibroblasts/myofibroblasts during CNT-induced fibrosis through TGF-β, Smad and β-catenin signaling. Similarly, angiogenesis may promote fibrosis through VEGF-mediated fibroblast proliferation and collagen synthesis. Together, these findings provide a mechanistic framework for the induction of fibrosis by CNTs which could facilitate the development of potential biomarkers and drug targets for diagnosis and treatment of the disease.

This work is supported by the National Institutes of Health grant R01-HL076340 and by the National Science Foundation grant EPS-1003907.

Declaration of interest

The authors report no declarations of interest.

Research findings and conclusions are those of the authors and do not necessarily represent the views of the National Institute for Occupational Safety and Health.

AbbreviationsCNT

Carbon nanotube

NP

Nanoparticle

SWCNT

Single-walled carbon nanotube

MWCNT

Multi-walled carbon nanotube

MMP

Matrix metalloproteinase

ROS

Reactive oxygen species

MAPK

Mitogen-activated protein kinase

NF-κB

Nuclear factor kappa-light-chain-enhancer of activated B cells

AP-1

Activator protein

ECM

Extracellular matrix

TNF-α

Tumor necrosis factor-alpha

IL-1β

Interleukin-1beta

PDGF

Platelet-derived growth factor

TGF-β

Transforming growth factor-beta

COX-2

Cyclooxygenase-2

iNOS

Inducible nitric oxide synthase

MCP-1

Monocyte chemoattractant protein

STAT-1

Signal transducer and activator of transcription

RTK

Receptor tyrosine kinase

CTGF

Connective tissue growth factor

VEGF

Vascular endothelial growth factor

EMT

Epithelial mesenchymal transition

AECs

Alveolar epithelial cells

IPF

Idiopathic pulmonary fibrosis

FGF

Fibroblast growth factor

OPN

Osteopontin

AgostiniCGurrieriC2006Chemokine/cytokine cocktail in idiopathic pulmonary fibrosisProc Am Thorac Soc33576316738201AntoniniJM2003Health effects of weldingCrit Rev Toxicol336110312585507AzadNIyerAKWangL2012Reactive oxygen species-mediated p38 MAPK regulates carbon nanotube-induced fibrogenic and angiogenic responsesNanotoxicology10.3109/17435390.2011.647929BeamerCAGirtsmanTASeaverBP2012IL-33 mediates multi-walled carbon nanotube (MWCNT)-induced airway hyper-reactivity via the mobilization of innate helper cells in the lungNanotoxicology10.3109/17435390.201.702230BergamaschiE2009Occupational exposure to nanomaterials: present knowledge and future developmentNanotoxicology3194201BonnerJC2002The epidermal growth factor receptor at the crossroads of airway remodelingAm J Physiol Lung Cell Mol Physiol283L5283012169571BonnerJC2007Lung fibrotic responses to particle exposureToxicol Pathol351485317325983BonnerJCRiceABMoomawCRMorganDL2000Airway fibrosis in rats induced by vanadium pentoxideAm J Lung Cell Mol Physiol278L20916BormPJRobbinsDHauboldS2006The potential risks of nanomaterials: a review carried out for ECETOCPart Fibre Toxicol31116907977BottiniMBrucknerSNikaK2006Multi-walled carbon nanotubes induce T lymphocyte apoptosisToxicol Lett160121616125885British Standards Institution (BSI) 2007Nanotechnologies Part 2: guide to safe handling and disposal of manufactured nanomaterialsBSI PD669922007BrownDKinlochIBangertU2007An in vitro study of the potential of carbon nanotubes and nanofibres to induce inflammatory mediators and frustrated phagocytosisCarbon45174356CestaMFRyman-RasmussenJWallaceDG2010Bacterial lipopolysaccharide enhances PDGF signaling and pulmonary fibrosis in rats exposed to carbon nanotubesAm J Respir Cell Mol Biol431425119738159ChakrabartiSPatelKD2005Matrix metalloproteinase-2 (MMP-2) and MMP-9 in pulmonary pathologyExp Lung Res3159962116019990ChangCCTsaiMLHuangHC2012Epithelial-mesenchymal transition contributes to SWCNT-induced pulmonary fibrosisNanotoxicology66001021711127ChapmanHA2011Epithelial-mesenchymal interactions in pulmonary fibrosisAnn Rev Physiol734133521054168ChaudharyNIRothGJHilbergF2007Inhibition of PDGF, VEGF and FGF signalling attenuates fibrosisEur Respir J299768517301095ClichiciSBirisARTabaranFFilipA2012Transient oxidative stress and inflammation after intraperitoneal administration of multi-walled carbon nanotubes functionalized with single strand DNA in ratsToxicol Appl Pharmacol2592819222280989DeMaioLBuckleySTKrishnaveniMS2012Ligand-independent transforming growth factor-β type I receptor signalling mediates type I collagen-induced epithelial-mesenchymal transitionJ Pathol2266334421984393DonaldsonKMurphyFADuffinRPolandCA2010Asbestos, carbon nanotubes and the pleural mesothelium: a review of the hypothesis regarding the role of long fibre retention in the parietal pleura, inflammation and mesotheliomaPart Fibre Toxicol7520307263DonaldsonKPolandCA2012Inhaled nanoparticles and lung cancer – what we can learn from conventional particle toxicologySwiss Med Wkly142w1354722714122DongPXWanBWangZX2012Exposure of single-walled carbon nanotubes impairs the functions of primarily cultured murine peritoneal macrophagesNanotoxicology10.3109/17435390.2012.694487DuPont™2008Nano risk framework output worksheet for: DuPont™ Crystar® 6920 PET poly-(ethylene terephthalate) resin with sepiolite clay, Pangel S-9 as an encapsulated nanodispersed fillerAvailable from: http://www.epa.gov/opptintr/nano/dupont2.pdf [last accessed 2 Jan 2013]ElgrabliDFlorianiMAbella-GallartS2008Biodistribution and clearance of instilled carbon nanotubes in rat lungPart Fibre Toxicol52019068117FenoglioIAldieriEGazzanoE2012Thickness of multiwalled carbon nanotubes affects their lung toxicityChem Res Toxicol25748222128750FerrariGCookBDTerushkinV2009Transforming growth factor-beta 1 (TGF-beta1) induces angiogenesis through vascular endothelial growth factor (VEGF)-mediated apoptosisJ Cell Physiol2194495819180561HeXYoungSSchwegler-BerryD2011Multiwalled carbon nanotubes induce a fibrogenic response by stimulating reactive oxygen species production, activating NF-κB signaling, and promoting fibroblast-to-myofibroblast transformationChemical Res Toxicol24223748HubbardRVennALewisSBrittonJ2000Lung cancer and cryptogenic fibrosing alveolitis. A population-based cohort studyAm J Respir Crit Care Med1615810619790InghilleriSMorbiniPOggionniT2006In situ assessment of oxidant and nitrogenic stress in bleomycin pulmonary fibrosisHistochem Cell Biol125661916307278InoueKKoikeEYanagisawaR2009Effects of multi-walled carbon nanotubes on a murine allergic airway inflammation modelToxicol Appl Pharmacol2373061619371758JainSThakareVSDasM2011Toxicity of multiwalled carbon nanotubes with end defects critically depends on their functionalization densityChem Res Toxicol2420283921978239JaurandMC1997Mechanisms of fiber-induced genotoxicityEnviron Health Perspect1051073849400703JaurandMFRenierADaubriacJ2009Mesothelioma: do asbestos and carbon nanotubes pose the same health risk?Particle Fibre Toxicol616JiaGWangHYanL2005Cytotoxicity of carbon nanomaterials: single-wall nanotube, multi-wall nanotube, and fullereneEnviron Sci Technol3913788315787380KaganVEKonduruNVFengW2010Carbon nanotubes degraded by neutrophil myeloperoxidase induce less pulmonary inflammationNature Nanotechnol5354920364135KaganVETyurinaYYTyurinVA2006Direct and indirect effects of single walled carbon nanotubes on RAW 264.7 macro phages: role of ironToxicol Lett1658810016527436KangSHerzbergMRodriguesDFElimelechM2008Antibacterial effects of carbon nanotubes: size does matter!Langmuir2464091318512881KeaneMP2004Angiogenesis and pulmonary fibrosis: feast or famine?Am J Respir Crit Care Med170207915280169KhalilNXuYDO’ConnorRDuronioV2005Proliferation of pulmonary interstitial fibroblasts is mediated by transforming growth factor-beta1-induced release of extracellular fibroblast growth factor-2 and phosphorylation of p38 MAPK and JNKJ Biol Chem28043000916246848KimKKKuglerMCWoltersPJ2006Alveolar epithelial cell mesenchymal transition develops in vivo during pulmonary fibrosis and is regulated by the extracellular matrixProc Natl Acad Sci USA10313180516924102KisinERMurrayARKeaneMJ2007Single-walled carbon nanotubes: geno- and cytotoxic effects in lung fibroblast V79 cellsJ Toxicol Environ Health Part A702071918049996KisinERMurrayARSargentL2011Genotoxicity of carbon nanofibers: are they potentially more or less dangerous than carbon nanotubes or asbestos?Toxicol Appl Pharmacol25211021310169KnaapenAMBormPJAlbrechtCSchinsRP2004Inhaled particles and lung cancer. Part A: mechanismsInt J Cancer10979980915027112LamCJamesJTMcCluskeyRHunterRL2004Pulmonary toxicity of single-wall carbon nanotubes in mice 7 and 90 days after intratracheal instillationToxicol Sci771263414514958LamCWJamesJTMcCluskeyR2006A review of carbon nanotube toxicity and assessment of potential occupational and environmental health risksCrit Rev Toxicol3618921716686422LeaskAAbrahamDJ2004TGF-beta signaling and the fibrotic responseFASEB J: Off Publ Fed Am Soc Exp Biol1881627LeeJKSayersBCChunKS2012Multi-walled carbon nanotubes induce COX-2 and iNOS expression via MAP Kinase-dependent and -independent mechanisms in mouse RAW264.7 macrophagesParticle Fibre Toxicol914LindroosPMCoinPGBadgettA1997Alveolar macrophages stimulated with titanium dioxide, chrysotile asbestos, and residual oil fly ash upregulate the PDGF receptor-alpha on lung fibroblasts through an IL-1beta-dependent mechanismAm J Respir Cell Mol Biol16283929070613LiuJLegrosSMaG2012Influence of surface functionalization and particle size on the aggregation kinetics of engineered nanoparticlesChemosphere879182422349061MagrezAKasasSSalicioV2006Cellular toxicity of carbon-based nanomaterialsNano Lett61121516771565MangumJBTurpinEAAntao-MenezesA2006Single-walled carbon nanotube (SWCNT)-induced interstitial fibrosis in the lungs of rats is associated with increased levels of PDGF mRNA and the formation of unique intercellular carbon structures that bridge alveolar macrophages in situParticle Fibre Toxicol315MannaSKSarkarSBarrJ2005Single-walled carbon nanotube induces oxidative stress and activates nuclear transcription factor-kappaB in human keratinocytesNano Lett516768416159204MaynardADBaronPAFoleyM2004Exposure to carbon nanotube material: aerosol release during the handling of unrefined single-walled carbon nanotube materialJ Toxicol Environ Health Part A678710714668113MercerRRHubbsAFScabilloniJF2011Pulmonary fibrotic response to aspiration of multi-walled carbon nanotubesParticle Fibre Toxicol821MercerRRScabilloniJWangL2008Alteration of deposition pattern and pulmonary response as a result of improved dispersion of aspirated single-walled carbon nanotubes in a mouse modelAm J Physiol Lung Cell Mol Physiol294L879718024722MishraARojanasakulYChenB2012Assessment of pulmonary fibrogenic potential of multiwalled carbon nanotubes in human lung cellsJ NanomaterMitchellLAGaoJWalRV2007Pulmonary and systemic immune response to inhaled multiwalled carbon nanotubesToxicol Sci: Off J Soc Toxicol10020314MullerJDecordierIHoetPH2008Clastogenic and aneugenic effects of multi-wall carbon nanotubes in epithelial cellsCarcinogenesis294273318174261MullerJHuauxFMoreauN2005Respiratory toxicity of multi-wall carbon nanotubesToxicol Appl Pharmacol2072213116129115MurphyFAPolandCADuffinR2011Length-dependent retention of carbon nanotubes in the pleural space of mice initiates sustained inflammation and progressive fibrosis on the parietal pleuraAm J Pathol178258760021641383MurrayARKisinERTkachAV2012Factoring-in agglomeration of carbon nanotubes and nanofibers for better prediction of their toxicity versus asbestosParticle Fibre Toxicol910NagaiHOkazakiYChewSH2011Diameter and rigidity of multiwalled carbon nanotubes are critical factors in mesothelial injury and carcinogenesisProc Natl Acad Sci USA108E1330822084097OberdörsterGMaynardADonaldsonK2005Principles for characterizing the potential human health effects from exposure to nanomaterials: elements of a screening strategyParticle Fibre Toxicol28PacurariMQianYFuW2012Cell permeability, migration, and reactive oxygen species induced by multiwalled carbon nanotubes in human microvascular endothelial cellsJ Toxicol Environ Health Part A751294722251262PacurariMYinXJZhaoJ2008Raw single-wall carbon nanotubes induce oxidative stress and activate MAPKs, AP-1, NF-kappaB, and Akt in normal and malignant human mesothelial cellsEnviron Health Perspect11612111718795165PalomäkiJVälimäkiESundJ2011Long, needle-like carbon nanotubes and asbestos activate the NLRP3 inflammasome through a similar mechanismACS Nano568617021800904PardoAGibsonKCisnerosJ2005Up-regulation and profibrotic role of osteopontin in human idiopathic pulmonary fibrosisPlos Med2e25116128620ParkERohJKimS2011A single intratracheal instillation of single-walled carbon nanotubes induced early lung fibrosis and subchronic tissue damage in miceArchiv Toxicol85112131PatelV2011Global carbon nanotubes market outlook: industry beckonsNanotech Insights2315PatlollaAKHussainSMSchlagerJJ2010aComparative study of the clastogenicity of functionalized and nonfunctionalized multi-walled carbon nanotubes in bone marrow cells of Swiss-Webster miceEnviron Toxicol256082120549644PatlollaAKnightenBTchounwouP2010bMulti-walled carbon nanotubes induce cytotoxicity, genotoxicity and apoptosis in normal human dermal fibroblast cellsEthn Dis20S1-657220521388PeãoMNAguasAPde SáCMGrandeNR1994Neoformation of blood vessels in association with rat lung fibrosis induced by bleomycinAnat Record23857677509580PolandCADuffinRKinlochI2008Carbon nanotubes introduced into the abdominal cavity of mice show asbestos-like pathogenicity in a pilot studyNature Nanotechnol3423818654567PulskampKDiabatéSKrugHF2007Carbon nanotubes show no sign of acute toxicity but induce intracellular reactive oxygen species in dependence on contaminantsToxicol Lett168587417141434RadiskyDCKennyPABissellMJ2007Fibrosis and cancer: do myofibroblasts come also from epithelial cells via EMT?J Cell Biochem101830917211838ReddyARKrishnaDRReddyYNHimabinduV2010Translocation and extra pulmonary toxicities of multi wall carbon nanotubes in ratsToxicol Mech Meth2026772RockJRBarkauskasCECronceMJ2011Multiple stromal populations contribute to pulmonary fibrosis without evidence for epithelial to mesenchymal transitionProc Natl Acad Sci USA108208678RotoliBMBussolatiOBarilliA2009Airway barrier dysfunction induced by exposure to carbon nanotubes in vitro: which role for fiber length?Human Exp Toxicol283618RotoliBMBussolatiOBianchiMG2008Non-functionalized multi-walled carbon nanotubes alter the paracellular permeability of human airway epithelial cellsToxicol Lett1789510218403140SatoYYokoyamaAShibataK2005Influence of length on cytotoxicity of multi-walled carbon nanotubes against human acute monocytic leukemia cell line THP-1 in vitro and subcutaneous tissue of rats in vivoMol BioSyst11768216880981SaxenaRWilliamsWMcGeeJ2007Enhanced in vitro and in vivo toxicity of polydispersed acid-functionalized single-wall carbon nanotubesNanotoxicology1291300SayesCMLiangFHudsonJL2006Functionalization density dependence of single-walled carbon nanotubes cytotoxicity in vitroToxicol Lett1611354216229976SchneebergerEELynchRD2004The tight junction: a multifunctional complexAm J Physiol Cell Physiol286C12132815151915ShvedovaAACastranovaVKisinER2003Exposure to carbon nanotube material: assessment of nanotube cytotoxicity using human keratinocyte cellsJ Toxicol Environ Health Part A6619092614514433ShvedovaAAKapralovAAFengWH2012Impaired clearance and enhanced pulmonary inflammatory/fibrotic response to carbon nanotubes in myeloperoxidase-deficient micePlos One7e3092322479306ShvedovaAAKisinERMercerR2005Unusual inflammatory and fibrogenic pulmonary responses to single-walled carbon nanotubes in miceAm J Physiol Lung Cell Mol Physiol289L69870815951334ShvedovaAAKisinEMurrayAR2008Inhalation vs. aspiration of single-walled carbon nanotubes in C57BL/6 mice: inflammation, fibrosis, oxidative stress, and mutagenesisAm J Physiol Lung Cell Mol Physiol295L5526518658273SimePJMarrRAGauldieD1998Transfer of tumor necrosis factor-alpha to rat lung induces severe pulmonary inflammation and patchy interstitial fibrogenesis with induction of transforming growth factor-beta1 and myofibroblastsAm J Pathol153825329736031SinghRPantarottoDLacerdaL2006Tissue biodistribution and blood clearance rates of intravenously administered carbon nanotube radiotracersProc Natl Acad Sci USA10333576216492781SinhaNYeowJT2005Carbon nanotubes for biomedical applicationsIEEE Trans Nanobioscience41809516117026SongYLiXDuX2009Exposure to nanoparticles is related to pleural effusion, pulmonary fibrosis and granulomaEur Respir J345596719696157StellaGM2011Carbon nanotubes and pleural damage: perspectives of nanosafety in the light of asbestos experienceBiointerphases6P11721721837ThannickalVJFanburgBL1995Activation of an H2O-generating NADH oxidase in human lung fibroblasts by transforming growth factor beta 1J Biol Chem2703033488530457ThannickalVJToewsGBWhiteES2004Mechanisms of pulmonary fibrosisAnn Rev Med5539541714746528TianFCuiDSchwarzH2006Cytotoxicity of single-wall carbon nanotubes on human fibroblastsToxicol In Vitro2012021216697548Turner-WarwickM1963Precapillary Systemic-pulmonary anastomosesThorax182253714064617ValkoMRhodesCJMoncolJ2006Free radicals, metals and antioxidants in oxidative stress-induced cancerChem Biol Interact16014016430879VanheeDGossetPWallaertB1994Mechanisms of fibrosis in coal workers’ pneumoconiosis. Increased production of platelet-derived growth factor, insulin-like growth factor type I, and transforming growth factor beta and relationship to disease severityAm J Respir Crit Care Med1501049557921435WallaceWAHFitchPMSimpsonAJHowieSEM2007Inflammation-associated remodelling and fibrosis in the lung – a process and an end pointInt J Exp Pathol881031017408453WangLCastranovaVMishraA2010aDispersion of single-walled carbon nanotubes by a natural lung surfactant for pulmonary in vitro and in vivo toxicity studiesParticle Fibre Toxicol731WangLLuanpitpongSCastranovaV2011aCarbon nanotubes induce malignant transformation and tumorigenesis of human lung epithelial cellsNano Lett11279680321657258WangRMikoryakCLiS2011cCytotoxicity screening of single-walled carbon nanotubes: detection and removal of cytotoxic contaminants from carboxylated carbon nanotubesMol Pharm813516121688794WangLRojanasakulYCastranovaV2010bDirect fibrogenic effects of dispersed single-walled carbon nanotubes on human lung fibroblastsJ Toxicol Environ Health Part A734102220155582WangXXiaTNtimSA2011bDispersal state of multiwalled carbon nanotubes elicits profibrogenic cellular responses that correlate with fibrogenesis biomarkers and fibrosis in the murine lungACS Nano597728722047207WarheitDBLaurenceBRReedKL2004Comparative pulmonary toxicity assessment of single-wall carbon nanotubes in ratsToxicol Sci: Off J Soc Toxicol7711725WillisBCBorokZ2007TGF-beta-induced EMT: mechanisms and implications for fibrotic lung diseaseAm J Physiol Lung Cell Mol Physiol293L5253417631612WillisBCLieblerJMLuby-PhelpsK2005Induction of epithelial-mesenchymal transition in alveolar epithelial cells by transforming growth factor-[beta]1: potential role in idiopathic pulmonary fibrosisAm J Pathol16613213215855634ZhangYYanB2012Cell cycle regulation by carboxylated multiwalled carbon nanotubes through p53-independent induction of p21 under the control of the BMP signaling pathwayChem Res Toxicol2512122122428663ZhangLWZengLBarronARMonteiro-RiviereNA2007Biological interactions of functionalized single-wall carbon nanotubes in human epidermal keratinocytesInt J Toxicol261031317454250ZhuLChangDWDaiLHongY2007DNA damage induced by multiwalled carbon nanotubes in mouse embryonic stem cellsNano Lett73592718044946ZhuMTFengWYWangY2009Particokinetics and extrapulmonary translocation of intratracheally instilled ferric oxide nanoparticles in rats and the potential health risk assessmentToxicol Sci: Off J Soc Toxicol10734251

Mechanisms of lung fibrosis: irritants such as nanoparticles induce epithelial injury resulting in infiltration of immune cells such as neutrophils, eosinophils and alveolar macrophages at the site of tissue injury. Activated neutrophils can exaggerate the ROS response. Moreover, ROS generation upon particle–cell interactions activates cytokine growth receptor cascade. ROS-dependent activation of RTKs, MAPK, Akt and NF-κB results in expression of genes related to inflammation and fibrosis. ROS can also activate TGF-β to mediate the fibrogenic effects. Recruitment of leukocytes induces key pro-fibrotic cytokines including TNF-α, IL-1β and IL-13, which can further damage the epithelial cells. TNF-α and IL-1β stimulation upregulates TGF-β and PDGF, respectively, which in turn increase collagen production via fibroblast and myofibroblast proliferation. Alternatively, fibroblasts can directly induce fibrosis via proliferation and differentiation into myofibroblasts. In addition, epithelial cells undergoing EMT expand the pool of fibroblasts and myofibroblasts thereby driving fibrogenesis.

Effects of physicochemical properties of CNTs on their biological activities based on size and surface area.

Type of CNTSystemEffectStudy
Purified MWCNT, short (220 nm) and long (825 nm)Human acute monocytic leukemia THP-1 cell lineLonger tubes induce increased inflammationSato et al. (2005)
SWCNT, long (0.5–100 mm) and short (0.5–2 mm) MWCNT, long (5–9 mm) and short (0.5–2 mm)Human epithelial Calu-3Longer MWCNTs and SWCNTs cause significant disruption of barrier functionRotoli et al. (2009)
MWCNT, long (13 μm) and (56 μm), tangled (1–5 mm) and (5–20 mm)In vivoLength-dependent inflammation and granuloma formationPoland et al. (2008)
MWCNT, short (1–10 mm), long tangled (10–50 mm), long needle-like (450 mm), asbestos (4.6 mm) and carbon blackPrimary human macrophagesEnhanced activation of NRLP3 inflammasome and secretion of IL-1β, IL-1α by longer MWCNTsPalomaki et al. (2011)
MWCNT, long (3–14 mm) and short (1.5 mm)Mammalian immune and epithelial cancer cells RAW264.7 macro-phages and MCF-7Increased cytotoxicity with longer MWCNTsLiu et al. (2012)
MWCNT, long, short, tangled, nickel nanowires, long and shortIn vivoLength-dependent retention of CNTs into lung pleura resulting in sustained inflammation and progressive fibrosisMurphy et al. (2011)
MWCNT, dispersed thin (50 nm), aggregative (2–20 nm), thick (150nm)Human peritoneal mesothelial cellsThinner MWCNTs induced more potent cytotoxic response in terms of inflammagenocity and carcinogenicityNagai et al. (2011)
Purified MWCNT, thick (70 nm) and thin (9.4 nm)Murine alveolar macrophages and in vivo in ratsThinner MWCNTs more toxic than the thicker ones both in vitro and in vivoFenoglio et al. (2012)
SWCNT and MWCNTBacteria (E. coli K12) cellsSWCNTs are much more toxic to bacteria than MWCNTs based on diameter differencesKang et al. (2008)
SWCNT (138 m2/g), carbon nanofibers, CNF (21 m2/g), asbestos (8 m2/g)In vivo C57BL/6 miceSWCNTs with higher surface area induced increased oxidative stress, inflammation, pulmonary damage and fibrosis than CNF and asbestosMurray et al. (2012)
SWCNT, MWCNT, active carbon, carbon black and carbon graphiteHuman fibroblast cellsSWCNTs with smaller surface area more toxic than their larger counterpartsTian et al. (2006)
MWCNT, CNF carbon nanoparticlesHuman lung tumor cellsSize and aspect ratio dependent cytotoxicity of MWCNTMagrez et al. (2006)

Effects of physicochemical properties of CNTs on their biological activities based on (A) functionalization; (B) presence of metal impurities; (C) dispersion status.

Panel A
SWCNT, control and acid functionalized (AF-SWCNT)LA4 mouse lung epithelial cells and in vivo in CD1 miceAF-SWCNTs more cytotoxic than SWCNTs in vitro; exerted stronger inflammatory response in vivo than control SWCNTsSaxena et al. (2007)
MWCNT, functionalized and non-functionalizedIn vivo bone marrow cells of Swiss-Webster miceFunctionalized MWCNTs induced greater clastogenic/genotoxic effects compared to the non-functionalized counterpartsPatlolla et al. (2010a)
SWCNT, purified, raw and carboxylatedNRK cell lineCarboxylated SWCNTs more cytotoxic as compared to purified and raw samplesWang et al. (2011c)
SWCNT, SWCNT-phenyl-SO3H, SWCNT-phenyl-SO3Na and SWCNT-phenyl-(COOH)2Human dermal fibroblastsCytotoxicity dependent on the degree of sidewall functionalizationSayes et al. (2006)
MWCNT, pristine and carboxylatedIn vivo miceThe degree of functionalization was inversely proportional to hepatic toxicityJain et al. (2011)
MWCNT, CNF, carbon nanoparticlesHuman lung tumor cellsFunctionalized carbon nanoparticles most toxic as compared to MWCNTs and CNFsMagrez et al. (2006)
SWCNT, purified and 6-amino-hexanoic acid-derivatized (AHA-SWCNT)Human epidermal keratinocytesFunctionalization induced mild cytotoxic responses and maintained cell viabilityZhang et al. (2007)
Panel B
30 wt% iron-rich SWCNTHuman keratinocytesLoss of cell viability, oxidative stress due to the catalytic activity of iron content associated within SWCNTsShvedova et al. (2003)
26 wt% iron-rich SWCNTMurine RAW 264.7 macrophagesLoss of intracellular thiols (GSH) and lipid hydroperoxides accumulation within the macrophagesKagan et al. (2006)
Panel C
SWCNT, poor and well dispersedIn vivo C57BL/6 micePoorly dispersed SWCNTs – proximal alveolar regions resulting in granulomatous lesions; well-dispersed CNTs-alveolar interstitial and pleural areas causing parenchymal granulomas and interstitial fibrosisMercer et al. (2008)
SWCNT, Survanta dispersed (SD-SWCNT) and non-dispersed (ND-SWCNT)Human lung epithelial BEAS-2B cellsSD-SWCNT induced more potent fibrogenic response both in vitro and in vivo as compared to ND-SWCNTWang et al. (2011a)