Review Article | OPEN ACCESS DOI: 10.23937/2572-4061.1510035

A Comprehensive Assessment of Hepatotoxicity Induced by Engineered Nanoparticles- A Review

S V S Rana*

Department of Toxicology, Ch Charan Singh University, Meerut, India

Abstract

Liver is the major detoxifying centre of the body. It removes xenobiotics and their metabolites through metabolism or biliary excretion. Hepatocytes constitute 80% of total liver mass and play a major role in storage, synthesis, metabolism and redistribution of essential molecules. Liver has been known to accumulate > 90% of nanomatrerials translocated from other organs. Bioconcentration of nanoparticles may lead to impairment of structure and function of hepatic cells. Therefore, it is critical to review the information available on NP induced hepatotoxicity, underline the gaps in our knowledge on their toxicity and propose future strategies for nanosafety.

Present review discusses recent researches available on the hepatotoxicity of engineered nanoparticles viz. carbon nanotubes (CNTs), nanoparticles of silver (AgNPs), gold (AuNPs), platinum (PtNPs), zinc oxide (ZnONPs), cadmium sulphide (CdSNPs), titanium dioxide (TiO2NPS), iron oxide (IONPs), copper (CuNPs), cerium oxide (CeO2NPs), silicon (SiO2NPs) and dendrimers in cell as well as animal models.

In vitro and in vivo studies show that these NPs elicit specific effects on serum enzymes, inflammatory cytokines, oxidative stress, gene expression and morphology of hepatocytes. Antioxidants like vitamin C and α lipoic acid can reverse the cell injury in some cases. Further, protective effects of ZnONPs and CeO2NPs against experimental hepato-carcinogenesis have also been highlighted. It is suggested that further efforts are required to address health issues concerned with nanomaterials.

Keywords

Liver, Nanoparticles, Carbon nanotubes, Quantum dots, Dendrimers, Cell death

Abbreviations

NPs: Nanoparticles; RES: Reticuloendothelial system; QDs: Quantum dots; ROS: Reactive oxygen species; LPO: Lipid peroxidation; CNTs: Carbon nanotubes; AgNPs: Silver nanoparticles; AuNPs: Gold nanoparticles; ZnONPs: Zinc oxide nanoparticles; PtNPs: Platinum nanoparticles; CdSNPs: Cadmium sulphide nanoparticles; TiO2NPs: Titanium dioxide nanoparticles; IONPs: Iron oxide nanoparticles; CNPs: Copper nanoparticles; CeO2NPs: Cerium oxide nanoparticles; SiO2NPs: Silicon nanoparticles; CCl4: Carbontetrachloride; APAP: Acetaminophen; PAMAM: Poly amidoamine; ALT: Alanine aminotransferase; AST: Aspartate transaminase; LDH: Lactate dehydrogenase; GPx: Glutathione peroxidise; GSH: Reduced glutathione; GA: Glycyrrhizic acid; SD: Sprague Dawley; BBB: Blood brain barrier

Introduction

It was during the meeting of American Physical Society (1959) when Richard. P. Feynman asked, "What would happen if we could arrange the atoms one by one the way we want them"? This meeting gave birth to the concept of nanotechnology. The word nano is linked to a Greek term "nanos" meaning dwarf. Today, nano is popular label in science, technology and medicine. During 1990-2000, new processes for the synthesis of nanoparticles (NPs) were developed. In subsequent years, health hazards posed by NPs were recognized. NPs have emerged as a new class of environmental pollutants that may affect atmosphere, terrestrial and aquatic ecosystems. Their prevalence in ecosystems can be harmful to human beings, animals, plants and aquatic species [1]. Furthermore, occupational exposure to NPs during their production, commercial and biomedical use may lead different health problems and safety issues [2].

The science that mainly addresses safety issues of NPs is known as nanotoxicology. It refers to the studies on interaction between nanostructures and biological systems. Specific responses are elicited by NPs corresponding to their size, shape, composition, surface chemistry and aggregation. Nanoparticles have been classified into two groups i.e. engineered NPs and incidental NPs [3]. Quantam dots, carbon nanotubes, dendrimers and fullerenes which have a diameter < 100 nm are known as engineered NPs (ENPs). Whereas, accidentally generated diesel particles are called incidental NPs. They can enter human body through different portals i.e. dermal, respiratory, gastro-intestinal, ocular, auditory, intravenous and mucous routes. Their toxicity is determined by barrier function and clearance mechanisms at respective portals of entry. Large surface area to volume ratio facilitates interactions between cell membrane and NPs [4]. Modifications in NPs surface may cause undesirable ionic interactions with biological systems [5].

To-date, limited information is available on the absorption and translocation of NPs. Experimental studies made in rodents have demonstrated that NPs deposited in lungs can translocate to the pulmonary interstitium [6]. Translocation of NPs from lungs to secondary organs i.e. liver, kidney, heart and brain depends on their physical properties [7]. Translocation of inhaled NPs to brain has been associated with neurodegenerative diseases caused by air pollutants [8]. Nevertheless, skin is potent barrier to certain nanomaterials. Similarly, NPs undergo limited gastrointestinal absorption except in environmental or occupational exposures. Smaller, hydrophobic and neutral particles are prone to increased absorption.

Target Toxicity of NPs

Skin, pulmonary and reticuloendothelial system (RES) including liver and spleen have been identified as the main target organs for NPs toxicity. NPs are taken over by RES through opsonisation [9]. Aside RES, kidney may be another organ of NP toxicity. Some fullerenes and dendrimers have been known to distribute in renal tissue [10,11]. There are many examples of particulate induced lysosomal dysfunction. Alterations in lysosomal permeability and subsequent release of lysosomal enzymes contributed to apoptosis induced by silica microparticles in alveolar macrophages [12]. Nanoparticles of neodymium oxide [13]; quantam dots [14] and fullerenes [15] are also known to induce autophagy in vitro.

Absorption of Nanoparticles

Size, shape and surface charge determine the uptake of NPs by cells through size selectivity matching their endocytic pits. Absorption of NP may occur through phagocytosis or pinocytosis. Pinocytosis is further classified as macropinocytosis where particles > 1 μm are absorbed. Other mechanisms are clathrin or caveolae mediated endocytosis or clathrin-caveolae independent endocytosis. Caveolae are made by plasma membrane invaginations of 50-80 nm size containing cholesterol and sphingolipid receptors [16,17]. Endocytosis may occur through lipid rafts that offer suitable platforms to facilitate assembly of receptors, adaptors, regulators and other downstream proteins as a signalling complex [18].

Similarly, clathrin coated pits of 100-200 nm have been shown to be associated with scaffold proteins such as AP-2 and eps-15 [19]. Nevertheless, the specific endocytic mechanism by which cells internalize specific NPs such as quantam dots remains unknown (Figure 1) summarizes some of these mechanisms that may occur in a macrophage or epithelial cell.

Figure 1: Cellular uptake of nanoparticles by a macrophage/epithelial cell. View Figure 1

Interaction of Nps with Biological Molecules

The size and shape of NPs is different from bulk particles. Therefore, their interaction with biomolecules i.e. proteins and lipids has been found to be different. They can bind with proteins and form a protein corona. CdSe/ZnS Qds formed a 3.3 nm thick corona with human serum albumin [20]. Further protein binding depends on the type of cell/system and the medium used. The interaction of NPs with biomolecules undergoes conformational changes [21,22]. These conformational changes determine cellular health as the immune system may treat them as foreign object and may try to eliminate them. Protein-NP interaction may allow their escape from endocytic route and promote cytotoxicity [23]. NPs can enter nucleus in some cases. In in vitro settings, they can cross the placental barrier [24,25]. Their accumulation in subcellular structures i.e. endosomes is higher than the free ions. NPs can directly interact with lipids. They can adhere to membranous lipids causing disturbances in membrane fluidity. It facilitates the penetration of NPs into the cell bypassing the endocytic route [23].

Common Mechanisms of NP Induced Cyto-Toxicity

NPs could cause adverse cellular and molecular effects through several mechanisms. These include generation of reactive oxygen species (ROS). Cytoskeletal effects. Intracellular signalling pathways and genotoxicity. NPs can generate ROS through different mechanisms. It can occur through direct effect as a result of exposure to acidic environment or through leached ions [26,27]. Their effects on mitochondria may cause the generation of ROS [28]. Interaction of NPs with redox active proteins such as NADPH oxidase and cell surface receptors may also lead to lipid peroxidation (LPO) [29]). However, kinetics of ROS induction can differ between various NPs.

Intracellular localization of NPs has been known to disrupt the cytoskeleton network. Effects of QDs on cytoskeleton have scarcely been studied. No differences in pheochromocytoma cells incubated with CdTe QDs for 72 h were reported [30]. However, significant structural changes in actin and tubulin networks of 3T3 fibroblasts after incubation with CdSe/ZnSe QDs were demonstrated [31]. Different surface modifications were also found to cause different degrees of cellular effects [32]. Moreover, gold NPs affected the morphology of several cell types such as A 549 human carcinoma lung cells [33]. Secondary effects caused by cytoskeletal changes are yet to be established.

NPs can interfere with delicate balance of cellular homeostasis and thus alter complex intracellular signalling pathways. Possible effects may be genotoxic caused by high levels of ROS [34]. Protein or gene expression may be altered due to perinuclear localization of NPs that may affect the transcription and translation functions [35]. Leaching of metal ions may also modulate protein or gene expression [36]. NPs may also interfere with cell surface receptors [37]. Specific effects of NPs on liver are discussed in following paragraphs.

Carbon nanotubes (CNTs)

Carbon nanotubes were first discovered by Diego and coworkers [38]. They are allotropic modifications of carbon, represented as a sheet of graphene (single layer of graphite) rolled into a cylinder. They are further classified as single walled or SWCNT or multi walled or MWCNT [39,40]. They are used in numerous technological applications including novel drug delivery systems [41]. They can interact with macromolecules such as proteins and DNA [42]. Several workers have investigated their toxicity in different systems. Multiwall carbon nanotubes induced oxidative stress and cytotoxicity in human embryonic kidney (HEK 293) cells [43]. SWCNTs also increased malondialdehyde and decreased GSH level in mice [44]. Only a few reports are available on the hepatotoxicity of CNTs. However, cytotoxicity of SWCNTs on hepatoma HG2 cells has been reported [45]. A dose-effect relationship on the effects of MWCNTs was reported in the liver of Kunming mice exposed to 10 and 60 mg/kg MWCNT [46]. They reported an increase in total bilirubin and AST in MWCNT treated rats as compared to PBS treated rats. Gene expression studies showed changes in G protein coupled receptors, cholesterol synthesis, CYP450, TNFα and NFkB signalling pathways. An interesting study has shown that rats exposed to SWCNT through intratracheal installation exhibited NMR based metabonomic changes in the blood plasma and liver extracts [47]. It has also been hypothesized that CNTs directly induce ROS generation that caused oxidative stress to various cells through inflammation and apoptosis [48]. Combined hepatotoxic effects of MWCNT and cadmium (Cd) were studied in mice [49]. These researchers showed that MWCNT reduced hepatotoxicity of Cd. These observations were based on their findings on serum transaminases, total bilirubin and blood urea nitrogen (BUN). They speculated the role of metallothionein (MT) in MWCNT toxicity.

Quantum dots (QDs)

The structure of quantum dots has been studied by several workers [50,51]. These are special nanocrystals ranging from 1 to 10 nm in diameter. They exhibit unique electronic, optical, magnetic and catalytic properties. QDs show immunotoxiciyty and can induce oxidative stress and DNA damage [52]. It has also been demonstrated that QDs can cause cell death by lipid peroxidation of human neuroblastoma cells [53]. Hepatotoxicity of QDs in the liver of man and animals has also been studied by a few workers. These results are summarized in the following paragraphs.

Silver nanoparticles (AgNPs)

Silver from ancient times has been used as a therapeutic element. However, silver nanoparticles have been used for their antimicrobial activity. Very few workers have studied the cytotoxicity of AgNPs. The effects of different dose regimen on hepatotoxicity of AgNPs in rat have been studied by a few researchers [54]. They performed liver function tests and recorded the bioaccumulation of AgNPs in liver. Though elevated values for ALP were observed, other parameters showed no significant changes in comparison to controls.

Contrarily, hepatoprotective effects of AgNPs against CCl4 induced hepatotoxicity were observed [55]. Authors used AgNPs synthesized using aqueous leaf extract of a mangrove based plant Rhizophora apiculata. In other reports, edema and necrosis in the liver of male ICR mice after a single intravenous (8 μmol/kg) injection of AgSe (5.1 nm) QDs were observed [56,57]. However, mechanisms of cytotoxicity expressed by AgNPs have not yet been established. Silver ions released from medicaments may enter circulation facilitating translocation and accumulation in soft organs like liver and kidney [58].

Effects of two dose regimen i.e. 20 and 50 ppm (14 days) of silver nanoparticles on liver function of BALB/C mice were also studied [59]. These workers noticed that level of serum transaminases viz: alanine aminotransaminase (ALT) and aspartate transaminase (AST) increased significantly in AgNP treated mice. No significant difference was noted in male and female mice.

A study made on approximately 8.7 nm silver nanoparticles administered in albino rats for 28 days at 1, 2, 4 mg/kg b.w. concluded that AgNps induced hepatotoxicity through oxidative stress mechanisms. Bioaccumulation of AgNPs in liver and chromosomal aberrations in bone marrow was also recorded [60]. Oxidative stress induced by AgNPs was found to be a dose dependent phenomenon. Administration of 10 nm citrate stabilized AgNPs at 0.2 mg/kg b.w. to male Wistar rats for 14 days induced mild oxidative stress in brain but not in the liver [61]. A few researchers have attributed AgNPs toxicity to its preferential accumulation in liver. AgNPs in combination with copper and boron as a composite in the dose range of 1-20 mg/kg on early (4-24 h) acute exposure and late phase (96 h) exposure in normal and NLRP-3 deficient mice were found to cause acute liver injury. Elevated values for ALT, AST and LDH along with necrosis, Kupffer cell hyperplasia and lobular granulomas were observed [62].

Developmental hepatotoxicity of AgNPs has also been investigated by a few researchers. Post-gestational (1-19 days) administration of AgNPs (25 mg/kg) through intra-gastric gavage induced oxidative stress in the liver of rat pups. While glutathione peroxidise (GPx) activity and reduced glutathione (GSH) levels were decreased. Malondialdehyde and caspase-9 levels were significantly increased. Histological studies exhibited fatty degeneration [63].

Although antimicrobial effects of silver nanoparticles are well known, it was found to protect against acetaminophen induced hepatotoxicity. Rats were treated with three different dosages of AgNPs (50, 100, 150 μg/kg p.o.) after APAP treatment (2 g/kg p.o. once only). Serum enzymes values and bilirubin level declined after AgNP treatment. This report further suggests a therapeutic value of AgNPs [64].

Recently, a few authors have studied the molecular toxicity of AgNPs on liver microsomal fraction. NPs (5-80 nm) were administered daily to growing Wistar rat for 92 days. Electrophoretic studies revealed the presence of proteosome activator complex (Psme 1) and heat shock protein (HSPd 1) gene. AgNP treatment caused the disappearance of protein of B-2α tubulin chain (tuba 1b gene) from the microsomal fractions [65]. Molecular aspects of AgNPs toxicity along with ultrastructural changes were reported by a group from Thailand. Microarray study revealed up and down regulation of those genes that were not up or down regulated by Ag ion exposed cells. Hep G2 cells in AgNP treated group showed distorted ultrastructural changes [66].

Gold nanoparticles (AuNPs)

The use of AuNPs in nanomedicine has been suggested by a few scientists. Contrarily, a few authors found them to be toxic. Thus biosafety issues related with AuNPs have raised concerns for human health. A few researchers following standard procedures/protocols have confirmed its hepatotoxic effects. Healthy and damaged liver of mice showed differential effects of AuNPs in mice. Hwang, et al. [67] first induced liver injury in mice by feeding them with methionine choline deficient diet and then subjected them to AuNPs treatment. They recorded higher values for ALT, AST and ROS in the mice. They concluded that AuNPs display toxicity in stressed liver. Another study from Reshi and coauthors contradicted these results [68]. They showed that AuNPs ameliorate APAP induced hepatotoxicity in albino rats. They considered AuNPs as potential hepatoprotective agents. It has been demonstrated that intraperitoneal administration of AuNPs induces liver damage through oxidative stress. These effects are alleviated by melanin (an antioxidant) treatment [69].

Coating AuNPs with suitable carrier molecules was also found to affect their toxicity. Polyethylene glycol (PEG) coated AuNPs were less toxic than uncoated particles. These observations were made based on the results on serum transaminases and histopathological findings [70]. Coating of AuNPs with citrate and chitosan also affected the hepatotoxicity of AuNPs. Chitosan capped NPs were less toxic than citrate capped ones in Swiss mice. This observation was made by after analysing the inflammation related genes using RT-PCR [71].

Platinum nanoparticles (PtNPs)

Platinum nanoparticles (PtNPs) are widely used in cosmetics, industry and diagnostics. On absorption, nanoplatinum can accumulate in soft tissues viz: liver, spleen, kidney, lungs and heart. A study from Poland recently reported that PtNPs can induce DNA damage and apoptosis in liver [72]. Dose dependent hepato and renotoxicity of PtNPs has also been demonstrated. NP of 1 nm diameter when administered with CCl4 or cisplastin induced hepatotoxicity whereas those of 8 nm did not exaberate the toxicity in mice [73]. Toxic effects of subnanosized platinum particles (snPt) in mouse liver were also studied. Increased levels of inflammatory cytokines and histopathological changes in liver of mice were observed after intravenous administration of snPt at 15 mg/kg body weight. However, administration of nanosized platinum particles did not produce these abnormalities [74].

Zinc oxide nanoparticles (ZnONPs)

Nanoparticles of zinc are better known for their hepatoprotective than hepatotoxic effects. Zinc oxide nanoparticles (ZnOPs) are widely used in cosmetics, sunscreens, clothes,medicine and electronic devices. Several studies demonstrate that ZnOPs induce oxidative stress and apoptosis in hepatocytes [75]. Furthermore, a study showed that ZnOPs at 200 mg/kg and 400 mg/kg given through gavage to mice for 90 days induced focal necrosis and increase in AST and ALT values [76]. In addition, mRNA expression level and ER stress related genes (grp78, grp74, pdi-3, xbp-1) were also upregulated. These workers reported upregulation of ER stress associated apoptotic protein levels viz: caspase-3, caspase-9 and caspase-12.

Hepatotoxicity of Mn doped ZnS nanoparticles in mice were also studied by the same group of researchers [77]. They estimated ALT, AST, catalase, glutathione peroxidise, superoxide dismutase and malondialdehyde in the liver of Mn doped ZnSNP treated mice. Mn doped ZnSNP did not cause any obvious damage to liver. Contribution of QDs in hepatocyte pyroptosis and inflammation has also been investigated. QDs expressed cytotoxicity in LO2 cells in a dose dependent manner [78]. QDs activate NLP pyrin domain containing 3 (NLRP3) inflammosome in hepatocytes leading to a novel pro-inflammatory form of cell death called pyroptosis. NLRP3 activation was caused by QDs triggered mtROS production and Ca(2+) mobilization. In another study, ZnONPs at a daily dose of 2 mg/kg for 21 days administered to male Wistar rats induced structural changes in the liver. Sinusoidal dialatation, Kupffer cell hyperplasia, inflammatory cells infiltration, necrosis, hydropic degeneration, apoptosis, karyolysis, glycogen depletion and haemosiderosis were reported [79].

It was postulated that ZnONPs offered protection against dimethylnitrosamine induced hepatotoxicity in rat by inhibiting oxidative stress. Moreover values of proinflammatory cytokines viz: TNF-α and IL-12 were also reduced [75].

A comparison of hepatotoxic effects of zinc nanoparticles and zinc oxide nanoparticles in rat has also been made. Intraperitoneal administration of these particles increased the activity of gamma glutamyl transferase (GGT), lactate dehydrogenase (LDH) and caspase-3. Induction of TNF-α was also registered. They concluded that zinc nanoparticles are less toxic than ZnONPS [80].

Cadmium nanoparticles (CdSNPs)

Hepatotoxicity of cadmium microparticles on liver and other organs of man and experimental animals have been studied in the past by several laboratories. However, toxicity of cadmium nanoparticles is poorly known. It was shown that Cd/Se/Te based quantam dots 705 modulated liver redox balance in mice [81]. A dose dependent increase in metallothionein expression and liver function impairment was noted. Further a corresponding increase in oxidative stress, oxidative DNA damage and inflammation was noticed. QDs activated NLRP3 and pyroptosis. It was attributed to mitochondrial ROS and Ca2+ mobilization [82]. Hepatotoxicity of Cds nanoparticles of different size/length was also studied [78]. It was interesting to note that hepatotoxicity of smaller CdSNP was greater than the larger CdS NPs.

Titanium dioxide nanoparticles (TiO2NPs)

Titanium dioxide nanoparticles (TiO2NPs) are now widely used in food, cosmetics, agriculture, medical devices and building engineering related processes. Their effects on human health have recently been reviewed. Liver disorders caused by TiO2NP were reported to increase the level of serum trans-aminases, LPO and oxidative stress in Wistar rats treated with 300 mg/kg of TiO2NP for 14 days by gavage [83]. These researchers showed that glycyrrhizic acid (GA) protected the rats against hepatic injury caused by TiO2NP. In vitro and in vivo toxicity of TiO2NP enhanced under oxidative stress conditions. A study made on BRL-3A liver cells and in the liver of Sprague-Dawley (SD) rat suggested a synergy between oxidative stress and TiO2NP induced hepato-toxicity [84]. Further cell death ratio was significantly enhanced (up to 2.62 fold) in BRL-3A cells exposed to OS and TiO2NP. A comparative study on the hepatotoxicity of TiO2NPs and sodium oleate coated iron oxide nanoparticles (OC-Fe3O4NPs) was made in Wistar rat [85]. Based on results on redox defences, these researchers concluded that OC-Fe3O4NPs do affect redox enzymes but liver is able to retain its functional integrity. TiO2NPs were also found to affect metabolic function of liver. In a study made in mice treated with TiO2NP (21 nm) for 14 days, ultrastructural changes viz: Mitochondrial oedema and gene expression variations were noticed [86].

A few studies have been made to investigate the reversal of TiO2NP toxicity by antioxidants. Oxidative stress induced by TiO2NPs and TiO2 bulk particles (150 mg/kg) in the liver of Sprague-Dawley rats was reversed by extracts of a common herb cinnamon (Cinnamonium cassia). Morphological as well as haematological improvements were also recorded [87]. In another study, tiron was also found to protect against the toxicity of TiO2NPs. While TiO2NPs upregulated the proapoptotic Bax gene and down regulated the antiapoptotic Bcl-2 gene, tiron upregulated Bcl-2 and decreased Bax expression. These results were supported by observations on serum enzymes and histopathological changes [87]. Moreover, quercetin and idebenone also ameliorated hepatotoxicity caused by TiO2NPs. These antioxidants modulated serum enzymes, VEGF, NO, DNA damage in the liver of TiO2NP treated rat liver [88].

Protective role of vitamin A and vitamin E against liver injury caused by TiO2NPs in male Wistar rat has also been determined. Enzyme biomarkers of liver function, histopathological changes, antioxidant enzymes and inflammatory mediators were increased in TiO2NPs (300 mg/kg) treated rats. Vitamin A and E both inhibited these parameters showing an ameliorative effect [89].

Iron oxide nanoparticles (IONPs)

Iron oxide nanoparticles are used in different disciplines of diagnostic science, biomedical sciences and drug delivery systems. On penetration, IONPs are taken up by cell organelle (endosomes/lysosomes) especially in hepatocytes. They contribute to cellular iron pool and release into cytoplasm after decomposition. Magnetic iron oxide nanoparticles can accumulate in liver, spleen, lungs and brain after inhalation. It shows their ability to cross the BBB [90]. Enzyme biomarkers of liver function viz: AST, ALT, and LDH increased in the liver of rats treated with different concentration of IONPs. In IONPs exposed rats at three concentrations i.e. 500, 1000, 2000 mg/kg, an increase in serum transaminases at all concentrations was recorded [91]. Whereas, exposure of rats to bulk iron oxide exhibited no effect. Another study reported that a dose of 10 mg/kg of dextran coated IONPs do not affect functional integrity of liver in Wistar rat [92]. A few studies have been made to observe the effects of superparamagnetic nanoparticles (USP IO-NPs) on LO2 cells [93]. These particles could induce cytotoxicity and caused leakage of LDH. These particles affected several genes especially those related with calcium homeostasis and inflammation i.e. IL, 1B, IL6 and IL8. USP IONPs can cause ER stress as well. ER stress induced by several nanoparticles has recently been discussed by Rana [94]. NPs have been shown to modulate the cross talk between mitochondria and ER. Human hepatic L02 cells when exposed to USPIO- NPS (2.5, 7.5 and 12.5 μg/mL) for 6 hr did not cause hepatic injury. However, it induced apoptosis at 20 μg/mL after 24 hr. It was postulated that the injury is mediated by Cox-2 genes [95].

Copper nanoparticles (CNPs)

Copper nanoparticles (CNPs) work as anti-biotic, anti-microbial and antifungal agents when added to plastic items and textiles. They are used in conductive inks and pastes as a substitute for expensive metals in electronic displays and transmissive conductive thin film applications. Toxicity of CNPs is not fully understood. However, available data show that CNPs at a high dose (200 mg/kg/d for 5 days) could induce overt hepatotoxicity in rats. An integrated metabolomic study revealed mitochondrial failure, enhanced ketogenesis, fatty acid β oxidation and glycolysis to contribute in its toxicity [96]. High dose(s) of CNPs could elevate serum enzymes, triglytcerides and bilirubin. Histopathological observations exhibited necrosis. Further, gene expression studies indicated that genes related to oxidoreductases, metabolism and signal transduction pathways were involved in the development of hepatotoxicity [97]. CNPs have been found to be toxic in fish also. A fresh water fish, Cyprinus carpio, when exposed to CNPs and CuO at 0.5, 1.0 and 1.5 mg/L showed higher values for malondialdehyde in the liver. It was concluded that LPO contributes in the hepatotoxicity of CNPs [98].

Amelioration of CNPs induced hepatotoxicity by extracts of green tea has also been reported [99]. Theses researchers concluded that green tea offered protection against hepatotoxicity of CNPs (20-30 nm) administered at a dose of 40 mg/kg/b w. Green tea improved the activity of liver enzymes, antioxidant status and suppressed DNA fragmentation and the expression of caspase-3 and Bax proteins. Another agent that offered protection against CNPs induced liver damage was α lipoic acid [100]. Status of LPO, NO, copper and apoptotic genes (c-myc and c-jun) improved in CNPs treated rats when co-adminstered with α lipoic acid. These results suggest that CNPs induce hepatotoxicity through oxidative stress in rat.

Cerium oxide nanoparticles (CeO2NP)

Cerium is the second member and the most reactive element in the lanthanide series. Cerium oxide (CeO2/ceria) is considered the most suitable oxide of cerium. Ceria nanoparticles demonstrate the formation of more oxygen vacancies. The large surface area to volume ratio in its nanoparticle enables CeO2 to react differently resulting in unique properties.

Nanoceria is used in solid oxide fuel cells, catalytic applications and photocatalysis. Recently it has drawn considerable attention as a therapeutic agent in the treatment and prevention of diseases associated with oxidative stress. A few reports are available on its hepato-protective rather than hepato-toxic effects. D-galactosamine and lipo-polysaccharide induced hepatotoxicity in rat was alleviated by CeO2 [101]. These workers showed that CeO2NP decreased translocation of cytoplasmic Nrf-2 with concomitant decrease in gene expression of HO-1. These effects were attributed to its antioxidative properties. Results on antioxidant enzymes and histopathological observations supported this conclusion. Another evidence of antioxidative potential of CeO2 is provided by experiments made on monocrotaline induced hepatotoxicity in mice [102]. It was shown that monocotaline induced decrease in hepatic GSH, GPx, GR and GST is normalized after the treatment of CeO2NPs.

A recent study showed that CeO2NP offered protection against diethylnitrosamine induced hepatotoxicity in mice. Pretreatment of CeO2NPs attenuated the activity of antioxidant enzymes and expression of Bcl2 and Cox2. This report again supports the antioxidative role of CeO2NPs [103].

Silicon nanoparticles

Silica nanoparticles are employed in several commercial, agriculture and medical applications. However, the information on their health effects remains elusive. A few studies are available on their effects on structure and function of liver. Size of silica nanoparticles seems to be a confounding factor in their toxicity. It was reported that silica particles having a diameter of 300 to 1000 nm elicited no adverse effects while SP-70 could induce liver injury at 30 mg/kg b.w. [104]. Repeated administration of SP-70 twice a week for four weeks at 10 mg/kg b .w. caused hepatic fibrosis. Serum enzyme markers were also increased. Hasezaki, et al. also confirmed that SP-70 is potent hepato-toxin [105]. Silica nanoparticles (14 nm) were found to induce apoptosis in human liver (HepG2) cells that was regulated by ROS through p53, bax/bcl-2 and caspase pathways. ROS scavenger, vitamin C modulated apoptotic markers [106].

There are reports suggesting that SiO2NPs (10-80) when administered to rats (nm) disturb tricarboxylic cycle and liver metabolism. It can induce oxidative stress and alter liver cell morphology (150 μg) for 90 days [107]. Another study supported these results through global metabolomic study on SiNPs treated human hepatoma cells (HepG2) and ICR mice liver. This study concluded that glutathione metabolism and oxidative stress are amongst the principal causes of SiNP induced hepatotoxicity [108]. SiNPs influence CYP450 both in rat and human hepatocytes [109]. In addition to apoptosis, SiNPs can induce pyroptosis through NLRP3 inflammasome activation which is caused by mesosporous silica nanoparticles (MSN) induced ROS generation [110]. A recent study suggested that SiONPs (10 nm) make profound changes in morphometry, biochemistry, hematology and genes expression of DMES in rat [111].

Dendrimers

The term dendrimer comes from a Greek word meaning "Dendron". Dendron is translated into a tree. A dendrimer possesses a symmetric structure around a core. A dendrimer molecule has hundreds of possible sites to couple to an active species. It possesses a hydrophobic core and hydrophilic periphery. It exhibits a micelle like behaviour. Dendrimers have been explored for encapsulation/scaffolding of hydrophobic compounds and anticancer drugs.

There are certain anticancer drugs i.e. methotrexate and 6-mercaptopurine that exhibit hepatotoxicity. When these drugs were encapsulated by the dendrimer based melamine and administered in C3H mice at subchronic doses, significant reduction in hepatotoxicity was observed [112]. Subsequently, the same group of researchers showed that melamine dendrimer given at 40 mg/kg to mice resulted into hepatotoxicity as determined through serum enzymes and histopathological changes [113]. Role of their route of administration was also discussed [114]. It was interesting to note that the nanomaterials such as poly-amidoamine (PAMAM) that are widely used in pharmaceutical industry caused hepatotoxicity through growth inhibition, mitochondrial injury and apoptosis in human liver cells. Blockage of autophagy in PAMAM treated mice led to hepatoprotection [115].

Conclusion and Perspectives

In recent times, nanoparticles have emerged as a new class of environmental pollutants. Additionally, occupational exposure to NPs may contribute to some unknown health issues in humans. Their toxicity is determined by barrier functions and clearance mechanisms at respective portals of entry. Experimental studies demonstrate their adverse effects on skin, lungs, liver, kidney, heart and brain of experimental animals. The interaction between NPs and biomolecules causes conformational changes that determine cellular health. The adverse effects are categorized as autophagy, apoptosis, necrosis, pyroptosis, oxidative stress, cytoskeleton changes and altered intracellular signalling pathways. Change in gene and protein expression affects transcription and translation functions.

Present review summarizes the specific hepatotoxic effects of ENPs. To note, CNTs can directly induce oxidative stress, inflammation and apoptosis, whereas, silver NPs could cause edema and necrosis. They exhibit protective effects against liver injury caused by CCl4 and acetaminophen. It is interesting to note that subnanosized platinum nanoparticles cause increased secretion of inflammatory cytokines. Smaller CdSNPs induce greater hepatotoxicity than larger NPs. Earlier studies demonstrated TiO2NPs affect metabolic function of liver. IONPs were found to cause ER stress. Copper nanoparticles can cause mitochondrial failure. However, CeO2NPs offer protection against dimethylnitrosamine toxicity. Available reports indicate that redox imbalance is the principal cause of SiNP induced hepatotoxicity. Dendrimers do affect growth inhibition, mitochondrial function and induce apoptosis.

These observations suggest that there appears an urgent need to develop nanosafety research. In vitro studies on NP toxicity on new models like hepatocyte like cells derived from puripotent stem cells using toxicogenomic tools have opened new avenues in NP research [116]. Further research on molecular toxicology of NPs as well as on their therapeutic values is warranted. Defining the interactive mechanism between NPs and biological molecules will be helpful in designing safer nanomaterials.

Conflict of Interest

The author declares no conflict of interest in the preparation of this article.

Acknowledgement

Financial assistance received from Indian Science Congress Association in the form of Sir Asutosh Mookerjee Fellowship to the author is gratefully acknowledged.

References

  1. Colvin VL (2003) The potential environmental impact of engineered nanomaterials. Nat Biotechnol 21: 1166-1170.
  2. Kraegeloh A (2018) Nanosafety research an ongoing story. Chem Res Toxicol 31: 1105.
  3. Stern ST, McNeil (2008) Nanotechnology safety concerns revisited. Toxicol Sci 101: 4-21.
  4. van Doren EAF, Temmerman PRHD, Francisco AD, Mast J (2011) Dtermination of the volume specific surface area by using transmission electron tomography for characterization and definition of nanomaterials. J Nanobiotechnol 9: 17.
  5. Nakamura H, Watano S (2018) Direct permeation of nanoparticles across cell membrane: A review. Kona Powder Particle J 35: 49-65.
  6. Oberdorster G (2000) Toxicology of ultrafine particles: in vivo studies. Philos Trans R Soc London A 358: 2719-2740.
  7. Kreyling WG, Semmler M, Erbe F, Mayer P, Takenaka S (2000) Translocation of ultrafine insoluble iridium particles from lung epithelium to extrapulmonary organs is size dependent but very low. J Toxicology Environ Health A 65: 1513-1530.
  8. Peters A, Veronesi B, Calderon-Garcishienas L, Gehr P, Chen LC (2006) Translocation and potential neurological effects of fine and ultrafine particles update. Part Fibre Toxicol 3: 13.
  9. Ogawara K, Furumoto K, Takakura Y, Hashida M, Higaki K (2001) Surface hydrophobicity of particles is not necessarily the most important determinant in their in vivo disposition after intravenous administration in rats. J Control Release 77: 191-198.
  10. Lee CC, Mackay JA, Frechet JM, Szoka FC (2005) Designing dendrimers for biological applications. Nat Biotechnol 23: 1517-1526.
  11. Nigavekar SS, Sung LY, Llanes M, El-Jawahri A, Lawrences TS (2004) 3H dendrimer nanoparticle organ/yumor distribution. Pharm Res 21: 476-483.
  12. Thibodeau MS, Giardina C, Knecht DA, Helble J, Hubbard AK (2004) Silica induced apoptosis in mouse alveolar macrophages is initiated by lysosomal enzyme activity. Toxicol Sci 80: 34-48.
  13. Chen Y, Yang, L, Feng C, Wen LP (2005) Nano neodymium oxide induces massive vacuolization and autophagic cell death in non small cell lung cancer NCl-H460 cells. Biochem Biophys Res Commun 337: 52-60.
  14. Selverstor O, Zabimyk O, Zscharmack M, Balavina L, Nowicki M (2006) Quantam dots for human mesenchymal stem cell labelling. A size dependent autophagy activation. Nano Lett 6: 2826-2832.
  15. Yamawaki H, Iwai N (2006) Cytotoxicity of water soluble fullerenes in vascular endothelial cells. Am J Physiol Cell Physiol 290: C1495-C1502.
  16. Lajoie P, Nabi I R (2007) Regulation of raft dependent endocytosis. J Cell Mol Med 11: 644-653.
  17. Pelkmans L, Puntener D, Helenius A (2002) Local actin polymerization and dynamin recruitment in SV-40 induced internalization of caveolae. Science 296: 535-539.
  18. Zhang LW, Monteiro-Riviere (2009) Mechanism of quantum dot nanoparticle cellular uptake. Toxicol Sci 110: 138-155.
  19. Swanson JA, Watts C (1995) Macropinocytosis. Trends Cell Biol 11: 424-428.
  20. Rocker C, Potzt F, Zhang WJ, Parak GU (2009) A quantitative fluorescence study of protein monolayer formation on colloidal nanoparticles. Nat Nanotechnology 4: 577-580.
  21. Lacerda SH, Park JJ, Meuse C, Pristinski D, Becker ML, et al. (2010) Interaction of gold nanoparticles with common human blood proteins. ACS Nano 4: 365-379.
  22. Mahmoudi M, Shokrrgozar MA, Sardari S, Moghadam MK, Vali H, et al. (2011) Irreversible changes in protein confirmation due to interaction with superparamagnetic iron oxide nanoparticles. Nanoscale 3: 1127-1138.
  23. Lin J, Zhang H, Chen Z, Zheng Y (2010) Penetration of lipid membranes by gold nanoparticles: Insights into cellular uptake, cytotoxicity and their relationship. ACS Nano 4: 5421-5429.
  24. Gu YJ, Cheng J, Lin CC, Lam YW, Cheng SH, et al. (2009) Nuclear penetration of surface functionalized gold nanoparticles. Toxicol Appl Pharmacol 237: 196-204.
  25. Chu M, Wu Q, Yang H, Yuan R, Hou S, et al. (2010) Transfer of quantum dots from pregnant mice to pups across the placental barrier. Small 6: 670-678.
  26. Stroh A, Zimmer C, Gutzeit C, Jakstadt M, Marschinke F, et al. (2004) Iron oxide particles from molecular magnetic resonance imaging cause transient oxidative stress in rat macrophages. Free Rad Biol Med 36: 976-984.
  27. Jain TK, Reddy MK, Morales MA, Leslic-Pelecky DL, Labhasetwar V (2008) Biodistribution, clearance and biocompatibility of iron oxide magnetic nanoparticles in rats. Mol Pharm 5: 316-327.
  28. Soto K, Garza KM, Murr LE (2007) Cytotoxic effects of aggregated nanomaterials. Acta Biomater 3: 351-358.
  29. Pisanic TR, Jim S, Shubayev VI (2009) Iron oxide magnetic nanoparticle nanotoxicity: Incidence and mechanism. In: Sahu SC, Casciano DA, Nanotoxicity: From in vivo to in vitro models to Health Risks. John Wiley and Sons Ltd, London, 397-425.
  30. Prasad BR, Nikolskaya N, Connolly D, Smith TJ, Byrne SJ, et al. (2010) Long term exposure to CdTe quantam dots on PCl2 cellular activity and the determination of optimum non toxic concentrations for biological use. J Nanobiotechnol 8: 7.
  31. Mahto SK, Park C, Yoon TH, Rhee SW (2010) Assessment of cytocompatibility of surface modified Cd/Se quantum dots for BALB/3T3 fibroblast cells. Toxicology In Vitro 24: 1070-1077.
  32. Gupta AK, Gupta M (2005) Cytotoxicity suppression and cellular uptake enhancement of surface modified magnetic nanoparticles. Biomaterials 26: 1565-1573.
  33. Patra HK, Banerjee S, Chaudhuri U, Lahiri P, Dasgupta AK (2007) Cell selective response to gold nanoparticles. Nanomed 3: 111-119.
  34. Singh N, Manshian B, Jenkins GJ, Griffiths SM, Williams PM, et al. (2009) Nanogenotoxicology: The DNA damaging potential of engineered nanomaterials. Biomaterials 30: 3891-3914.
  35. Pisanic TR 2nd, Blackwell JD, Shubayev VI, Finones RR, Jin S (2007) Nanotoxicity of iron oxide nanoparticle internalization in growing neurones. Biomaterials 28: 2572-2581.
  36. Soenen SJ, Himmeireich U, Nuytten N, Pisanic TR 2nd, Ferrari A, et al. (2010) Intracellular nanoparticle coating stability determines nanoparticle diagnostics efficacy and cell functionality. Small 6: 2136-2145.
  37. Miller IS, Lynch I, Dowling D, Dawson KA, Gallagher WM (2010) Surface induced signalling events control actin rearrangement and motility. J Biomed Mater Res A 93: 293-504.
  38. Iijima S (1991) Helical microtubules of graphite carbon. Nature 354: 56-58.
  39. Diego AR, Carl AB, John CM (2006) Carbon nanotubes in biomedical applications. Nanotechnol Law Bus 3: 263-265.
  40. Grobert N (2007) Carbon nanotubes becoming clean. Materials Today 10: 28-35.
  41. Wu W, Wieckowski S, Pastorin G, Benincasa M, Klumpp C, et al. (2005) Targeted delivery of amphoterricin B to cells by using functionalized carbon nanotubes. Angew Chem Int Engl 44: 6358-6362.
  42. Foldvari M, Bagonluri M (2008) Carbon nanotubes as functional excipients for nanomedicines: II. Drug delivery and biocompatibility issues. Nanomedicine 4: 183-200.
  43. Reddy ARN, Reddy YN, Krishna DR, Himabindu V (2010) Multi walled carbon nanotubes induce oxidative stress and cytotoxicity in human embryonic kidney (HEK 239) cells. Toxicology 272: 11-16.
  44. Yeng ST, Wang X, Jia G, Gu Y, Wang T, et al. (2008) Long term accumulation and low toxicity of single walled carbon nanotubes in intravenously exposed mice. Toxicol Lett 181: 182-189.
  45. Yuan J, Gav H, Sui J, Chen WN, Ching CB (2011) Cytotoxicity of single walled carbon nanotubes on human hepatoma Hep G2 cells.An iTRAq Coupled 2D LC-MS/MS proteome analysis. Toxicol In Vitro 25: 1820-1827.
  46. Ji Z, Zhang D, Li L, Shen X, Deng X, et al. (2009) The hepatotoxicity of multiwalled carbon nanotubes in mice. Nanotechnol 20: 445101.
  47. Bencheng Lin, Huashan Zhang, Zhiqing Lin, Yanjun Fang, Lei Tian, et al. (2013) Studies on single walled carbon nanotubes -induced hepatotoxicity by NMR based metabonomics of rat blood plasma extracts. Nanoscale Res Lett 8: 236.
  48. Muthu M, Abdulla A, Pandey BL (2013) Major toxicities of carbon nanotubes induced by reactive oxygen species- should we worry about the effects on the lungs,liver and normal cells. Nanomedicine 8: 863-866.
  49. Qi Wei, Bi Juanjuan, Tian Longlong, Li Zhan, Liu Peng (2014) The effect of multiwalled carbon nanotubes on hepatotoxicity of cadmium in accumulated cadmium metallothionein in mice. Biomed Res Int 2014: 463161.
  50. Aitken RJ, Credy KS, Tran CL (2004) Nanoparticles: An Occupational Hygiene Review. Health Safety Executive Research Report 274: HSE Books, London.
  51. Pelley JL, Daar AS, Saner MA (2009) State of academic knowledge on toxicity and biological effects of quantam dots. Toxicol Sci 112: 276-296.
  52. Gagne F, Auclair J, Turcotte P, Fourmier M, Gagnon C (2008) Ecotoxicity of CdTe quantam dots to fresh water mussels: Impacts on immune system, oxidative stress and genotoxicity. Aquatic Toxicol 86: 333-340.
  53. Choi AO, Cho SJ, Desbarats J, Lovric J, Maysinger D (2007) Quantum dot induced cell death involves Fas upregulation and lipid peroxidation in human neuroblastoma cells. J Nanobiotechnology 127: 143-153.
  54. Abd-Elhakeem Mohamed A, Badawy Ingy, El-Feky Said, Abdel Ghaffar Faten R, Abd-Elghafar Omar S, et al. (2015) Hepatotoxicity and nephrotoxicity of repeated oral administration of silver nanoparticles in rats. Nanomedicine and Nanobiology 2: 54-58.
  55. Hongru Zhang, Joe Antony Jacob, Ziyu Jiang, Senlei Xu, Ke Sun, et al. (2019) Hepatoprotective effect of silver nanoparticles synthesized using aqueous lead extract of Rhizophora apiculata. Int J Nanomedicine 14: 3517-3524.
  56. Tang H, Yang ST, Yang YF, Ke DM, Liu JH, et al. (2016)Blood clearance, distribution, transformation, excretion and toxicity of near infrared quantum dots Ag2Se in mice. ACS Appl Mater Interfaces 8: 17859-17869.
  57. Tang H, Yang ST, Ke DM, Yang YF, Liu JH, et al. (2017) Biological behaviour and chemical fates of Ag2Se quantum dots in vivo: The effect of surface chemistry. Toxicol Res (Camb) 6: 693-704.
  58. Tang J, Xi T (2008) Status of biological evaluation on silver nanoparticles. Sheng Wu Yi Xue Gong Cheng Xue Za Zhi 25: 958-961.
  59. Heydrnejad MS, Samani RJ, Aghaeivanda S (2015) Toxic effects of silver nanoparticles on liver and some haematological parameters in male and female mice (Mus musculus). Biol Trace Elem Res 165: 153-158.
  60. El Mahdy MM, Eldin TA, Aly HS, Mohammed FF, Shaalan MI (2015) Evaluation of hepatotoxic and genotoxic potential of silver nanoparticles in albino rats. Exp Toxicol Pathol 67: 21-29.
  61. Skalska J, Dabrowska-Bouta B, Struzynska L (2016) Oxidative stress in rat brain but not in liver following oral administration on a low dose of nanoparticulate silver. Food Chem Toxicol 97: 307-315.
  62. Ramadi KB, Mohamed YA, Al-Sbiei A, Almarzooqi S, Bashir G, et al. (2016) Acute systemic exposure to silver based nanoparticles induces hepatotoxicity and NLRP-3 dependent inflammation. Nanotoxicology 10: 1061-1074.
  63. Fatemi M, Moshtaghian J, Ghaedi K, Jafari Dinani N, Naderi G (2017) Effects of silver nanoparticle on the developing liver of rat pups after maternal exposure. Iran J Pharm Res 16: 685-693.
  64. Reshi MS, Uthra C, Yadav D, Sharma S, Asha S, et al. (2017) Silver nanoparticles protect acetaminophen induced acute hepatotoxicity. Regul Toxicol Pharmacol 90: 36-41.
  65. Shipelin VA, Kudan PV, Zgoda VG, Gmoshinskii IV, Khotimchenko SA (2018) Effect of silver nanoparticles on protein composition of rat liver microsomal fraction. Bull Exp Biol Med 166: 80-85.
  66. Kanidta Sooklert, Asarn Wongjarupong, Sarocha Cherdchom, Nicha Wongjarupong, Depicha Jindati, et al. (2019) Molecular and morphological evidence of hepatotoxicity after silver nanoparticle exposure: A systematic review, in Silico and ultrastructure investigations. Toxicol Res 35: 257-270.
  67. Hwang JH, Kim SJ, Kim YH, Nok JR, Gang GT, et al. (2012) Susceptibility to gold nanoparticle induced hepatotoxicity is enhanced in a mouse model of non-alcoholic steatohepatitis. Toxicology 294: 27-35.
  68. Reshi MS, Shukla S (2017) Acetaminophen induced hepatotoxicity: Preventive effects of gold nanoparticles. Toxicology Open Acceess 3: 66.
  69. Mohamed Anwar K Abdelhalim, Sherif A Abdelmottaleb Moussa, Huda Ay Qaid, Mohammed Suliman Al-Ayed (2018) Effect of melamin on gold nanoparticle - induced hepatotoxicity and lipid peroxidation in rats. Int J Nanomedicine 13: 5207-5213.
  70. Patlolla AK, Kumari SA, Tchounwoa PB (2019) A comparison of poly-ethylene glycol coated and uncoated gold nanoparticle mediated hepatotoxicity and oxidative stress in Sprague Dawlwy rats. Int J Nanomedicine 14: 639-647.
  71. Zaki El, El-Seedy AS, Keladu IP, Sharafeldin NA, Abdel Mouaty HM, et al. (2019) Impact of citrate and chitasan capped gold nanoparticles on the liver of Swiss albino mice. Histological and cyto-genotoxic study. Cell Mol Biol (Noisy-le-grand) 65: 9-23.
  72. Czubacka E, Czerczak S (2019) Are platinum nanoparticles safe to human health. Med Pr 70: 487-495.
  73. Isoda K, Daibo T, Yushina K, Yushioka Y, Tsutsumi Y, et al. (2017) Hepatotoxicity, nephrotoxicity and drug/chemical interaction toxicity of platinum nanoparticles in mice. Pharmazie 72: 10-16.
  74. Y Yamagishi, A Watari, Y Hayata, X Li, M Kondoh, et al. (2013) Hepatotoxicity of sub-nanosized platinum particles in mice. Pharmazie 68: 178-182.
  75. Rani V, Verma Y, Rana K, Rana SVS (2018) Zinc oxide nanoparticles inhibit dimethylnitrosamine induced liver injury in rat. Chem Biol Interact 295: 84-92.
  76. Yang X, Shao H, Liu W, Gu W, Shu X, et al. (2015) Endoplasmic stress and oxidative stress are involved in zinc oxide nanoparticle induced hepatotoxicity. Toxicol Lett 234: 40-49.
  77. Yang Y, Lv SV, Yu B, Xu S, Shen J, et al. (2015) Hepatotoxicity assessment of Mn doped ZnS quantam dots after repeated administration in mice. Int J Nanomedicine 10: 5787-5796.
  78. Lu Y, Xu S, Chen H, He M, Deng Y, et al. (2016) Cd/Se/ZnS quantam dots induce hepatocyte pyroptosis and liver inflammation via NLRP3 inflammasome activation. Biomaterials 90: 27-39.
  79. Almansour MI, Alferah MA, Shraideh ZA, Zarar BM (2017) Zinc oxide nanoparticles hepatotoxicity: histological and histochemical study. Environ Toxicol Pharmacol 51: 124-130.
  80. Sizova E, Mireshmikov S, Nechitailo X (2019) Assessment of structural organization of liver and biochemical parameters of blood serum after introduction of zinc nanoparticles and its oxides. Environ Sci Pollut Res Int 26: 17110-17120.
  81. Lin CH, Yang MH, Chang LW, Yang CS, Chang H, et al. (2011) Cd/Se/Te based quantam dot 705 modulated redox homeostasis with hepatotoxicity in mice. Nanotoxocol 5: 650-663.
  82. Li QZ, Sun J, Shen HT, Jia SF, Bai DS, et al. (2018) Cds nanoparticles of different lengths induce different responses in some of liver functions of mice. Bratist Lek Listy 119: 75-78.
  83. Wojcik EB, Szwajgier D, Oleszczuk P, Winiarska-Mieczan A (2019) Effects of titanium dioxide nanoparticles exposure on human health. Biol Trace Elem Res 193: 118-129.
  84. Sha B, Gao W, Wang S, Gou X, Li W, et al. (2014) Oxidative stress increased hepatotoxicity induced by nano-titanium dioxide in BRL-3A cells and Sprague Dawley rats. J Appl Toxicol 34: 345-356.
  85. Volkovova K, Handy RD, Staruchova M, Tulinska J, Kebis A, et al. (2015) Health effects of selected nanoparticles in vivo: Liver function and hepatotoxicity following intravenous injection of titanium dioxide and Na-oleate coated iron oxide nanoparticles in rodents. Nanotoxicology 1: 95-105.
  86. Yang J, Luo M, Tan Z, Dai M, Xie M, et al. (2017) Oral administration of titanium dioxide particle disrupts hepatic metabolic functions in a mouse model. Environ Toxicol Pharmacol 49: 112-118.
  87. Morgan A, Ibrahim MA, Galal MK, Ogaly HA, Abd-Elsalam RM (2018) Innovative perception on using tiron to modulate the hepatotoxicity induced by titanium dioxide nanoparticles in male rats. Biomed Parmacothernn 103: 553-561.
  88. Fadda M, Hagar H, Mohamed AM, Ali HM (2018) Quercetin and idebenone ameliorate oxidative stress, inflammation,DNA damage, and apoptosis induced by titanium dioxide nanoparticles in rat liver. Dose Response 16.
  89. Moradi A, Ziamazidi N, Ghafourikhosroshahi A, Abbasalipourkabir R (2019) Effects of vitamin A and vitamin E on attenuation of titanium dioxide nanoparticles-induced toxicity in the liver of male Wistar, rats. Mol Biol Rep 46: 2919-2932.
  90. Formenti P, Caquineau S, Desboeufs, Klaver A, Chevaillier S, et al. (2014) Mapping the physicochemical properties of mineral dust in western Africa: mineralogical composition. Atmos Chem Phys 14: 10663-10686.
  91. Kumari M, Rajak S, Singh SP, Murty US, Mahboob M, et al. (2013) Biochemical alterations induced by acute oral doses of iron oxide nanoparticles in Wistar rats. Drug Chem Toxicol 36: 296-305.
  92. Easo S L, Mohanan PV (2016) Hepatotoxicity evaluation of dextran stabilized iron oxide nanoparticles in Wistar rats. Int J Pharm 509: 28-34.
  93. He C, Jiang S, Yao H, Zhang L, Yang C, et al. (2018) Endoplasmic reticulum stress mediates inflammatory response triggered by ultra small superparamagnetic iron oxide nanoparticles in hepatocytes. Nanotoxicology 12: 1198-1214.
  94. Rana SVS (2020) Endoplasmic reticulum stress by toxic elements, a review, Biol Trace Elem Res.
  95. Che L, Yao H, Yang CL, Guo NJ, Huang J, et al. (2019) Cyclooxygenase -2 modulates ER-mitochondria crosstalk to mediate superparamagnetic iron oxide nanoparticles induced hepatotoxicity: An in vitro and in vivo study, Nanotoxicology 8: 1-19.
  96. Lei R, Wu C, Yang B, Ma H, Shi C, et al. (2008) Integrated metabolomic analysis of the nanosized copper particle-induced hepatotoxicity and nephrotoxicity in rats: A rapid screening method for nanotoxicity. Toxicol Appl Pharmacol 232: 292-301.
  97. Yang B, Wang Q, Lei R, Wu C, Shi C, et al. (2010) Systems toxicology used in nanotoxicity: mechanistic insights into the hepatotoxicity of nano copper particles from toxicogenomics. J Nanosci Nanotechnol 210: 8527-8537.
  98. Noureen A, Jabeen F, Tabish TA, Yaqub S, Ali M, et al. (2018) Assessment of copper nanoparticles (Cu-NPs) and copper (II) oxide (CuO) induced hemato-and hepatotoxicity in Cyprinus carpio. Nanotechnology 29: 144003.
  99. Ibrahim MA, Khalaf AA, Galal MK, Ogaly HA, Hassan A (2015) Ameliorative influence of green tea extract on copper nanoparticle induced hepatotoxicity in rats. Nanoscale Res Lett 363.
  100. Khalaf AA, Zaki AR, Galal MK, Ogaly HA, Ibrahim MA, et al. (2017) The potential protective effect of alpha lipoic acid against nanocopper particle induced hepatotoxicity in male rats. Hum Exp Toxicol 36: 881-891.
  101. Hashem RM, Rashd LA, Hashem KS, Soliman HM (2015) Cerium oxide nanoparticles alleviate oxidative stress and decreases Nrf-2/HO-1 in D-GALN/LPS induced hepatotoxicity. Biomed Pharmacother 73: 80-86.
  102. Amin KA, Hassan MS, Awad el-ST, Hashem KS (2011) The protective effects of cerium oxide nanoparticles against hepatic oxidative damage induced by monocrotaline. Int J Nanomedicine 6: 143-149.
  103. Adebayo OA, Akinloye O, Adaramoye OA (2020) Cerium oxide nanoparticles attenuate oxidative stress and inflammation in the liver of diethylnitrosamine treated mice. Biol Trace Elem Res 193: 214-225.
  104. Nishimori H, Kondoh M, Isoda K, Tsunoda S, Tsutsumi Y, et al. (2009) Silica nanoparticles as hepatotoxicants. Eur J Pharm Biopharm 72: 496-501.
  105. Hasezaki T, Isoda K, Kondoh M, Tsutsumi Y, Yagi K (2011) Hepatotoxicity of silica nanoparticles with a diameter of 100nm, Pharmazie 66: 698-703.
  106. Ahmad J, Ahamed M, Akhtar MJ, Alokayan SA, Siddiqui MA, et al. (2012) Apoptosis induction by silica nanoparticles mediated through reactive oxygen species in human liver cell line HepG2. Toxicol Appl Pharmacol 259: 160-168.
  107. Parveen A, Rizvi SH, Gupta A, Singh R, Ahmad I, et al. (2012) NMR base metabonomics study of sub-acute hepatotoxicity induced by silica nanoparticles in rats after intranasal exposure. Cell Mol Biol 58: 196-203.
  108. Chatterjee N, Jeong J, Yoon D, Kim S, Choi J, et al. (2018) Global metabolomics approach in in vitro and in vivo models reveals hepatic ,glutathione depletion induced by amorphous silica nanoparticles, Chem Biol Interact 293: 100-106.
  109. Cornu R, Rougier N, Pellequer Y, Lamprecht A, Hamon P, et al. (2018) Interspecies differences in the cytochrome P450 activity of hepatocytes exposed to PLGA and silica nanoparticles: An in vitro and in vivo investigation, Nanoscale 10: 5171-5181.
  110. Zhang X, Luan J, Chen W, Fan J, Nan Y, et al. (2018) Mesoporous silica nanoparticles induced hepatotoxicity via NLRP3 inflammasome activation and caspase -1-dependent pyroptosis. Nanoscale 10: 9141-9152.
  111. Almansour M, Alarifi S, Jarrar B (2018) In vivo investigation on the chronic hepatotoxicity induced by intraperitoneal administration of 10-nm silicon dioxide nanoparticles. Int J Nanomedicine 13: 2685-2696.
  112. Neerman MF, Chen HT, Parrish AR, Simanek EE (2004) Reduction of drug toxicity using dendrimers based on melamine, Mol Pharm 1: 390-393.
  113. Neerman MF, Zhang W, Parrish AR, Simanek EE (2004) In vitro and in vivo evaluation of a melamine dendrimer as a vehicle for drug delivery. J Int Pharm 281: 129-132.
  114. Gupta U, Agashe HB, Asthana A, Jain NK (2006) A review of in vitro and in vivo investigations on dendrimers: The novel nanoscopic drug carriers. Nanomedicine 2: 66-73.
  115. Li Y, Zeng S, Wang S, Sun Y, Wang Z, et al. (2015) Inhibition of autophagy protects against PAMAM dendrimers-induced hepatotoxicity. Nanotoxicology 9: 344-355.
  116. Gao X, Li R, Sprando RL, Yourick JJ (2020) Concentration dependent- toxicogenomic changes of silver nanoparticles in hepatocyte-like cells derived from human induced pluripotent stem cells. Cell Biol Toxicol.

Citation

Rana SVS (2020) A Comprehensive Assessment of Hepatotoxicity Induced by Engineered Nanoparticles- A Review. J Toxicol Risk Assess 6:035. doi.org/10.23937/2572-4061.1510035