Next Article in Journal
Protein Structures among Bio-Ethanol Co-Products and Its Relationships with Ruminal and Intestinal Availability of Protein in Dairy Cattle
Next Article in Special Issue
Preparation and Characterization of Nanoliposomes Entrapping Medium-Chain Fatty Acids and Vitamin C by Lyophilization
Previous Article in Journal
Endogenous Protease Nexin-1 Protects against Cerebral Ischemia
Previous Article in Special Issue
Galactosylated Chitosan Oligosaccharide Nanoparticles for Hepatocellular Carcinoma Cell-Targeted Delivery of Adenosine Triphosphate
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Effects of Nanomaterials as Endocrine Disruptors

Institute of Public Health, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, Roma 00168, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2013, 14(8), 16732-16801; https://doi.org/10.3390/ijms140816732
Submission received: 30 May 2013 / Revised: 8 July 2013 / Accepted: 25 July 2013 / Published: 14 August 2013
(This article belongs to the Special Issue Bioactive Nanoparticles 2013)

Abstract

:
In recent years, nanoparticles have been increasingly used in several industrial, consumer and medical applications because of their unique physico-chemical properties. However, in vitro and in vivo studies have demonstrated that these properties are also closely associated with detrimental health effects. There is a serious lack of information on the potential nanoparticle hazard to human health, particularly on their possible toxic effects on the endocrine system. This topic is of primary importance since the disruption of endocrine functions is associated with severe adverse effects on human health. Consequently, in order to gather information on the hazardous effects of nanoparticles on endocrine organs, we reviewed the data available in the literature regarding the endocrine effects of in vitro and in vivo exposure to different types of nanoparticles. Our aim was to understand the potential endocrine disrupting risks posed by nanoparticles, to assess their underlying mechanisms of action and identify areas in which further investigation is needed in order to obtain a deeper understanding of the role of nanoparticles as endocrine disruptors. Current data support the notion that different types of nanoparticles are capable of altering the normal and physiological activity of the endocrine system. However, a critical evaluation of these findings suggests the need to interpret these results with caution since information on potential endocrine interactions and the toxicity of nanoparticles is quite limited.

1. Introduction

Over the past 50 years, epidemiological data have revealed a significant increase in the incidence and prevalence of a number of adverse effects on human health such as alterations in the development and growth process, disorders of the immune and neurological systems, reduced fertility and the onset of some important diseases such as diabetes, obesity, breast, ovary, testicle and prostate cancer [13]. A possible explanation for the increase in these diseases lies in a growing exposure of workers and the general population to contaminants that may exert adverse effects on account of their action as endocrine disrupting chemicals (EDCs). In fact, most studies performed on EDCs have revealed that these compounds may play an important role in the onset of the aforementioned diseases by altering hormonal and homeostatic systems [410].
In 2002, the World Health Organization defined EDCs as “an exogenous substance or mixture that alters functions of the endocrine system and consequently causes adverse health effects in an intact organism, or its progeny, or (sub)populations” [11]. The group of chemical substances that belongs to this category is highly heterogeneous: it includes industrial solvents and lubricants and their by-products, dioxins, bisphenol A, polychlorinated biphenyls, persistent organic pollutants (POP), plastic compounds, plasticizers, pesticides such as chlorinated insecticides, imidazoles and triazoles, pharmaceutical agents, chemical compounds that are widely used in cosmetics such as phthalates, and heavy metals such as cadmium, mercury, arsenic, lead, manganese and zinc [1,2,12]. EDCs are found in virtually all regions of the world and typical human exposure in the general population occurs via environmental contamination of the food chain, especially fresh water fish and meat, or through contact with contaminated environmental matrices, while occupational exposure can occur during the production, use and disposal of the aforementioned chemical substances [13,14]. Since exposure of the general population and workers to EDCs is ubiquitous and unavoidable, and the impact on human health due to the known or unknown effects of these chemicals on hormonal systems is great, there is an urgent need to increase efforts to identify the compounds that can behave as endocrine disruptors and to study their molecular mechanisms of action. The Endocrine Disruptor Priority List (EDPL), developed as part of the European Union strategy for EDCs, represents a practical attempt to achieve a wider and more comprehensive knowledge of these substances. It provides a list and categorization (see Table 1) of chemicals that are likely or suspected to be EDCs [1518].
However, despite the studies carried out in recent years, current knowledge of EDCs is still limited and most research into this issue has concentrated on a few groups of chemical substances such as pesticides or POP, whereas data on a number of other xenobiotics that may act as EDCs is still scant and incomplete [1]. Therefore, to adequately address the issue of EDCs, it is our belief that the first step should be to identify all the possible compounds that can interfere and disrupt the homeostasis and regulatory mechanisms of the endocrine system. This provision is particularly urgent for those chemicals that have recently been used in workplaces and consumer products and whose toxicological profile has not yet been clearly and unequivocally defined. This is the case of nanoparticles (NPs).
The International Organization for Standardization, in ISO/TS 27687, defines a nanoparticle as a nano-object with all three external dimensions in the size range of approximately 1–100 nm [19]. NPs can be subdivided into two main categories, engineered NPs that are intentionally produced with very specific properties or compositions, and incidental NPs, also known as ultrafine particles, that are typically byproducts of processes such as combustion and vaporization and working activities such as welding or grinding [20]. Reducing particle size increases surface area and modifies unique physicochemical properties such as high conductivity, strength, durability, and chemical reactivity [21]. Furthermore, because of their very small size, NPs can enter cells directly by penetrating the cell membrane and may cause interference of important cell functions. The internalization of NPs can occur in a variety of ways and particle size largely influences their endocytic processes and cellular uptake ability. Additionally, several experimental reports have shown that nanoparticle size also affects their internalization in terms of efficiency [22,23]. The industrial applications of NPs are very wide-ranging and include those that may lead to more efficient water purification, stronger and lighter building materials, increased computing power and speed, enhanced generation and conservation of energy, and new tools for the diagnosis and treatment of diseases [24]. In recent years, several in vitro studies have assessed the potential adverse health effects of NPs, pointing out their ability to produce reactive oxygen species (ROS), releasetoxic ions, disrupt electron/ion cell membrane transport activity and cause oxidative damage and lipid peroxidation, while the results of in vivo studies have shown that these materials can induce adverse effects on the respiratory, cardiovascular and nervous systems [2527]. However, while nanotechnology and exposure to NPs are growing exponentially, research into their toxicological impact and possible hazard for human health and the environment is still fragmentary and largely incomplete. Consequently, even if nanotechnology and the commercialization of nanoenabled products and devices could help to address serious global problems (energy, transportation, pollution, health, medicine, and food), these benefits may not materialize unless a concerted effort is made to evaluate safety and health concerns regarding NPs.
By summarizing the findings of the in vitro and in vivo studies that have investigated the effects of NPs on the endocrine system, this review aims to evaluate the potential role of different types of NPs as endocrine disruptors, focusing in particular on their molecular mechanisms of action and on the relationship between their physical and chemical properties and the induction of alterations in endocrine function.

2. Impact of Endocrine Disrupting NPs on Reproductive Health

Hormones play a key role in influencing the development of the reproductive system and subsequently in controlling its activities once developed. For this reason, most of the research carried out on EDCs in the last two decades has focused its attention on reproductive health. With regard to the male reproductive system, numerous in vitro and in vivo study findings have demonstrated that EDCs can exert a number of detrimental effects such as malformed reproductive tissue, poor semen quality (low sperm counts, low ejaculate volume, high number of abnormal sperm, low number of motile sperm), prostate diseases, testicular cancer, and other recognized abnormalities of male reproductive tissues [3,28]. There is also evidence that EDCs may interfere with female reproductive development and function causing adverse effects such as fibrocystic disease of the breast, polycystic ovarian syndrome, endometriosis, uterine fibroids and pelvic inflammatory diseases, breast and reproductive organ tissue cancers and declining sex ratio [3,29].
Recently, the results of studies conducted to assess the potential toxic effects of NPs, have suggested that some of these may pose risks to male and female reproductive health by altering normal testis and ovarian structure, spermatogenesis and sperm quality, oogenesis, follicle maturation and sex hormone levels.

2.1. Effects of NPs on the Male Reproductive System

Most of the adverse effects of NPs on male reproductive function are mainly due to modification of the testicular structure (see Figure 1), impairment of spermatogenesis and alteration in the biosynthetic and catabolic pathways of testosterone (see Table 2).
To investigate damage caused by NPs to testicular constituent cells, three in vitro studies evaluated the cytotoxic effects of different types of NPs on mouse Leydig TM3 cells and on the RTG-2 cell line. The exposure of TM3 cells to increasing concentrations (0–1000 μg/mL) of titanium dioxide NPs (TiO2-NPs) induced a remarkable and dose-dependent reduction of cell viability [30]. Similarly, in the RTG-2 cell line, exposure to 50 μg/mL (the highest concentration used in this study) of TiO2-NPs produced a significant cytotoxicity [31]. In contrast, carbon black NPs (CB-NPs) and diesel exhaust NPs (DE-NPs) caused only slight cytotoxicity in TM3 cells [28], while cerium oxide NPs (CeO2-NPs) proved non-cytotoxic on the RTG-2 cell line [32]. With regard to these in vitro studies it should be noted that the different degree of observed cytotoxicity may depend on several factors. In fact the effect on the cytotoxicity of cells varies with different NPs and different cells, presumably because of the differing sensibility of cells and the varying sizes, doses, components and physico-chemical properties of the NPs. Furthermore, even when the experimental conditions (cell lines, particle size and concentrations) are quite similar for example in the studies of Vevers et al. [31] and Rosenkranz et al. [32], the conflicting results could be explained by taking into account the mode of toxicity that may be very different for the various metal oxide NPs, as has been shown for inflammogenic responses [62].
Several in vivo studies (see Table 2) have demonstrated that carbon-based NPs, nanoparticle-rich diesel exhaust (NRDE-NPs) and metal-based NPs are able to induce changes in the testicular histology of laboratory animals. These alterations appeared of great importance since they could affect the “micro-environment” required for a correct reproductive function. Intratracheal administration of a 0.1 mg/kg body weight dose of different CB-NPs caused partial vacuolization of the seminiferous tubules in ICR mice [36]. The same research group reported clear seminiferous tubule damage and low cellular adhesion of seminiferous epithelia in the offspring of this animal model after prenatal exposure to 0.2 mg/kg body weight of CB-NPs, demonstrating in utero developmental sensitivity to the CB-NP endocrine disrupting insult [38]. Another type of carbon-based NP induced similar damage (reduced thickness of the germinative layer, lower spermatogonia number, partial disappearance or vacuolization of Sertoli cells, vasodilatation and hyperemia) in the testes of BALB/c mice treated with intravenous injections (5 mg/kg/dose) of amine- or carboxylate-functionalized Multi Walled Carbon Nanotubes (MWCNTs) [37]. Severe adverse effects on seminiferous tubules, such as degenerative and necrotic changes, interstitial edema, desquamation of the seminiferous epithelium, and loss of spermatozoa were also observed in Fischer rats exposed via inhalation to different doses (15.37, 36.35 and 168.84 μg/m3) of NRDE-NPs [33]. The findings of a similar study, carried out in pregnant rats treated with 148.86 μg/m3 of the same NPs, detected the loss of germ cells in the seminiferous tubules of offspring [34]. Since carbon-based NPs of different types and particle sizes, administered to animals by different routes and in a variety of exposure doses caused similar damages, these NPs may share a common mechanism of action that could be the induction of oxidative stress as is suggested by the findings of Bai et al. [37] who observed an increase in malondialdehyde levels 15 days after treatment.
With regard to the testicular morphological alterations in offspring, the pre-natal exposure to TiO2-NPs of Slc:ICR mice, subcutaneously injected with 100 μg/kg body weight/day, also led to disruption of seminiferous tubules and lower numbers of Sertoli cells in 6 week-old pups [35]. It is worth noting that in this study TiO2-NPs were detected in Leydig cells, Sertoli cells and spermatids of the testis of offspring at both 4 days and 6 weeks of age suggesting a direct NP induced insult on the reproductive organ. In contrast, studies carried out by Yoshida et al. [38] and Li et al. [34] failed to detect NPs in the testis of pups prenatally exposed to CB-NPs and NRDE-NPs, respectively. Consequently, no definitive conclusion can be reached concerning the role NPs may play in causing direct damage to this organ. The damage observed may be related to accumulation of NPs in the placenta, thereby resulting in malnutrition of the foetus. However, the Authors observed no significant differences in body and organ weights between the exposed and control groups in either study.
In contrast with the previous studies, the intravenous administration of 45 and 225 mg/kg ω-methoxy and 45 mg/kg ω-aminoethyl poly(ethylene glycol) capped Au-NPs (mPEG@Au-NP and PEG-NH2@Au-NP, respectively) in ICR mice did not cause testicular morphological changes or germ cell apoptosis, even if these NPs had accumulated in mouse testes, passed through the blood-testis barrier, and entered germ cells [39]. Similarly, Morishita et al. [40] demonstrated that amorphous silica NPs were able to penetrate the blood-testis barrier and be internalized in cytoplasm and the nuclei of spermatocytes but did not induce any apparent testicular injury in BALB/c mice intravenously injected with 0.8 mg of the aforementioned NPs. Surprisingly, although the total doses of nanomaterials that were administered in these studies were significantly higher than the total dose used in the MWCNT report [37], no testicular morphological alterations were observed. These findings may be due to the different physico-chemical characteristics of the NPs studied, particulalrly their chemical composition, shape and surface functionalizations, since these properties significantly affect the toxicological profile of NPs. Morishita et al. [40] have suggested that NP testicular distribution and penetration may depend on the type of material but also on surface modifications as demonstrated by a higher accumulation in the testes of PEG-NH2@Au-NP compared to mPEG@Au-NP. Concerning alterations in spermatogenesis (see Table 2), several in vitro studies have investigated the potential toxic effects of different types of NPs (mainly metal- and carbon-based NPs) on spermatozoa and their maturation process, principally by evaluating cytotoxicity, DNA damage and modifications in sperm quality parameters. Significant dose-dependent cytotoxicity was observed in the C18-4 spermatogonial stem cell line exposed to concentrations ranging from 0 to 100 μg/mL of silver NPs (Ag-NPs), molybdenum trioxide NPs (MoO3-NPs) and aluminum NPs (Al-NPs) [42]. The Ag-NPs were found to be the most toxic. Interestingly, when evaluating the cytotoxicity of several Ag-NPs with different diameters and coatings, the same Authors demonstrated that smaller and hydrocarbon- coated Ag-NPs induced a greater reduction in cell viability [47]. Furthermore, Ag-NPs have been shown to interact with proteins and potentially block signaling by binding to glial cell line-derived neurotrophic factor or its receptor Ret, thereby eliminating ligand receptor interaction, or by interfering with intracellular signaling molecules. The same metal-based NPs and TiO2-NPs showed a dose and time dependent cytotoxic effect also in the Ntera2 (NT2, human testicular embryonic carcinoma) cell line and in primary testicular cells obtained from wild type (WT) C57BL6 mice and 8-oxoguanine DNA glycosylase knock-out (Ogg1−/−) (KO) genotype treated with concentrations of NPs ranging from 0 to 100 μg/mL [51]. Similar results were obtained by Terzuoli et al. [50] in human spermatozoa treated with different doses (0–500 μM) of Ag-NPs. However, the toxic effects of NPs on germ cells were not confirmed by the studies of Rafeeqi and Kaul and Murugan et al. [41,63] who demonstrated a clear biocompatibility between carbon nanotubes (CNTs) and spermatogonial cells and a fullerenol protective effect on the potential oxidative stress damage on spermatozoa, respectively. Once again the findings of these studies demonstrated the importance of different NP physico-chemical properties in determining the toxic effects. In fact, they show the importance of NP size, functionalization and chemical composition in influencing the biologic response of cell systems [41,42,47,63].
With regard to DNA damage, the treatment of mouse epididymal sperm with 0.5 × 1015 or 1.0 × 1015 particles/mL of gold nanoparticles (Au-NPs) resulted in inhibition of the chromatin decondensation process in gametes [49]. Unfortunately, this adverse effect was not observed by the same research group in a subsequent study conducted on mature gametes [64]. However, a concentration-dependent induction of sperm DNA damage was also observed in human spermatozoa treated with different doses of TiO2-NPs (3.73–59.7 μg/mL) and Zinc Oxide nanoparticles (ZnO-NPs) (11.5–93.2 μg/mL) [45]. Conflicting results were obtained for alterations in sperm motility induced by metal-based NPs, a parameter used to evaluate sperm quality. A complete loss of bovine spermatozoa motility was reported after exposure to 2.5 mg/mL of europium oxide NPs (Eu2O3-NPs) [44], while a 22% loss of motility was detected after treatment with 0.5–50 μM of Au-NPs [48,65]. The latter metal-based NPs also caused a reduction of motility in human fresh semen mixed with 500 μL of Au-NPs and the results of this study showed the ability of NPs to penetrate the heads and tails of spermatozoa inducing fragmentation of the sperm [46]. Finally, in the aforementioned study carried out by Terzuoli et al. [50], increasing concentrations of Ag-NPs significantly reduced human sperm motility. Conversely, motility and acrosome reactions of bovine sperm cells were not affected by exposure to 7.35 mM of magnetic iron oxide NPs (Fe3O4-NPs) coated with poly(vinyl alcohol) [43], or to 2.5 mg/mL of europium hydroxide NPs (EuOH3-NPs) conjugated with polyvinyl alcohol or polyvinyl piyrolidone [44]. Interestingly, these results suggest that the toxic potential of NPs may be altered by modifying NP surface functionalization.
The detrimental effects of NPs on the maturation process of male germ cells have also been confirmed by numerous in vivo studies. An increase in the testicular apoptotic index and the frequency of round spermatidis with two or more nuclei were observed in CBAB6F1 mice treated via oral gavage with increasing (0–1000 mg/kg) concentrations of TiO2-NPs [55]. Similar findings (clear reduction in sperm density and motility) were obtained by Guo et al. [52] in ICR mice intraperitoneally injected with 500 mg/kg of TiO2-NPs. The intravenous administration to Wistar rats of 5 or 10 mg/kg of Ag-NPs, with different diameters, determined a decrease in epididymal sperm count and an increase in DNA damage in germ cells [56]. In this study, the most severe adverse effects were caused by the smaller particles (20 nm vs. 200 nm), suggesting a relevant role of diameter in inducing greater biological reactivity. This finding should be given careful consideration as it may be linked to the different interaction between the NP surface and biological substances present in biological fluids. In fact, NPs may change their physico-chemical characteristics when transferred to body fluids. This may be due to protein corona formed on the NP surface which facilitates the uptake of NPs into cells and cell inner structures, consequently inducing greater toxicity of particles in the nanometer size. Interestingly, the in utero exposure of male offspring in pregnant BALB/c mice treated with 50–300 mg/kg of dimercaptosuccinic acid-coated Fe3O4-NPs also induced a significant reduction in spermatogonia, spermatocytes, spermatids and mature sperm [54]. However, in this study, histological examinations were conducted only on placental tissue and fetus liver, and even if the Fe3O4-NPs were passed via blood-placenta barrier, it is not clear whether the aforementioned findings were due the direct action of NPs on testes or to an accumulation in the placenta tissue. On the contrary, the administration to ICR mice of 45 and 225 mg/kg of mPEG@Au-NPs and of 45 mg/kg of PEG-NH2@Au-NPs failed to result in abnormalities in the sperm collected from the cauda epididymis of the animals [39].
Regarding daily sperm production (DSP) and other sperm functional features such as motility and density, adverse effects were reported after both fetal and adulthood exposure to TiO2-NPs and CB-NPs. Takeda et al. [35] observed a decrease in DSP and epidydimal sperm motility in Slc:ICR mice prenatally exposed to TiO2-NPs and the exposure, via inhalation or intratracheal administration, of pregnant C57BL/6J mice to 42 mg/m3 of nanosized TiO2 or to 67 μg/animal of CB-NPs caused a reduction of DSP in male F1 offspring and lowered sperm production in the F2 offspring, whose fathers were prenatally exposed to CB-NPs [58]. DSP was significantly reduced also in 5 week-old and 10–15 week-old male ICR mice after fetal exposure to 0.2 mg of CB-NPs [38], whereas adult mice subchronically exposed to 0.1 mg of CB-NPs showed a significant reduction in DSP [36]. However, other in vivo studies that evaluated alterations in spermatogenesis and sperm quality, using MWCNTs and other carbon-based NPs, failed to confirm the abovementioned findings. In fact, no alterations in the total sperm concentration, motility, percentage of abnormal semen and loss of acrosome integrity were detected in the study conducted by Bai et al. [37]. Likewise, Tang et al. [57], when evaluating the long term effects of short MWCNTs and polyethylene glycol functionalized s-MWCNTs (PEG-s-MWCNTs) in Kunming mice intravenously treated with a single dose of 100 μg/kg body weight, failed to observe sperm cell toxicity or changes in sperm morphologyFurthermore, in healthy and streptozotocin-induced diabetic male Wistar rats that were administered C60 fullerene (C60HyFn) at the dose of 4 μg/kg via the oral route, these NPs not only failed to cause significant toxic effects in testicular tissues, but actually increased both sperm motility and epididymal sperm concentration and decreased the abnormal sperm rate, indicating an important anti-oxidant activity [53]. It is worth noting that the lack of effects on the sperm functional features was observed mainly using functionalized and highly pure MWCNTs. Therefore, it can be assumed that the chemical composition and the functionalization of this type of NP play a key role in defining their toxicological profile. For instance, covalent modification of MWCNTs by linking PEG chains to the outside surface of MWCNTs has many advantages because it increases solubility, prolongs circulation half-life in vivo and reduces immunogenicity [57]. Another issue involving the capacity of NPs to interfere with the male reproductive system concerns the disruption of normal levels of sex hormones, particularly testosterone (see Table 2 and Figure 2). In fact, the results of two in vitro and ex vivo studies demonstrated that DE-NPs and CB-NPs are able to affect testosterone production in Leydig cells. In the study carried out by Komatsu et al. [30], after 48 h incubation, exposure of the TM3 Leydig cell line to 0–1000 μg/mL of DE-NPs and CB-NPs induced gene expression of the steroidogenic acute regulatory protein (StAR) which is an important molecule in the process of testosterone synthesis, involved as a carrier in cholesterol transfer from the outer to the inner mithocondrial membrane [66]. Moreover, the evaluation of testosterone production in interstitial testicular cells, dissected from male C57BL/Jcl mice exposed to 152.01 μg/m3 of NRDE-NPs, showed that this parameter significantly increased both with and without human chorionic gonadotropin stimulus [59].
Alterations in testosterone levels leading to impaired male reproductive function have also been investigated in numerous in vivo studies. Li et al. [33] observed that, in Fischer 344 rats, inhalation exposure to low (15.4 ± 1.0 μg/m3) and medium (36.4 ± 1.2 μg/m3) concentrations of NRDE-NPs significantly increased plasma and testicular testosterone, whereas this did not occur with exposure to a high concentration (168.8 ± 2.7 μg/m3), thereby clearly indicating an inverted-U dose-response. However, this phenomenon should be carefully evaluated since it may be due to agglomeration of NPs at the highest dose that reduces the activity of the nanoform. Plasma luteinizing hormone (LH) and follicle stimulating hormone (FSH) concentrations did not change significantly, while increased concentrations of the plasma and testicular immunoreactive (ir-) inhibin, a protein complex which inhibits FSH synthesis and secretion were observed in exposed groups. These findings suggest that the increased levels of testosterone were not related to LH stimulation but were due to the disrupted balance between androgen-metabolizing- and testosterone biosynthetic-enzymes. When attempting to clarify the mechanism responsible for the rise in testosterone levels, the same research group [60] confirmed results obtained by Komatsu et al. [30], thus proving that increased testosterone biosynthesis could be attributed to an increase in the mRNA expression of StAR and cytochrome P450 side-chain cleavage (P450scc), an enzyme responsible for the conversion of transported cholesterol to pregnenolone in Leydig cells [67]. Similar findings and action mechanism were obtained in the same animal model exposed to high (149 ± 8 μg/m3) and low (38 ± 3 μg/m3) NRDE-NPs concentrations [61]. Furthermore, higher levels of testosterone were also observed in male C57BL/Jcl mice treated with 152.01 μg/m3 of NRDE-NPs and the increase in this hormone was related to an enhanced expression of genes involved in testicular cholesterol synthesis, such as HMG-CoA, LDL-R, SR-B1, PBR, P450scc, P450 17α, and 17β-HSD [59]. Interestingly, in pregnant Fischer F344 rats, exposure to 148.86 μg/m3 of NRDE-NPs caused changes in offspring hormone levels involving decreased FSH and testosterone and increased ir-inhibin concentrations that were quite different from those observed in adult animals [34]. The possibility of NPs affecting reproductive function in different ways depending on the period of exposure i.e., during adulthood or the fetal period, is also corroborated by the findings of Yoshida [36,38] who reported elevated testosterone levels in ICR adult mice treated with CB-NPs but no significant increase in the offspring after in utero exposure. Elevated testosterone levels were also detected following the administration of PEG-NH2@Au-NP in ICR mice, while no alterations in LH and FSH plasma values were observed [39]. Taken together, these results suggested that modified-Au-NPs may act as endocrine disruptors that have a direct influence on testicular hormonal production, since pituitary LH and FSH levels remained unmodified. Moreover, NP surface chemistry would seem to play an important role in inducing hormonal alterations, in fact, unlike PEG-NH2@Au-NP, the mPEG@Au-NPs did not cause an increase in testosterone concentration. As confirmation of the importance of the functionalization of NPs in affecting sex hormone levels, no alterations in testosterone, LH and FSH plasma levels were observed in BALB/c mice, subacutely exposed to functionalized MWCNTs [37].
The aformentioned studies that investigated the adverse effects of NPs on male reproductive function used different types of NPs, cell lines, exposure routes and doses. Consequently, caution is needed when interpreting their findings, and reaching a definite conclusion poses a considerable challenge. Any evaluation of these results may take into account the fact that the exposure concentrations used in most of the studies were very high (up to 2.5 mg/mL and 1000 mg/kg for in vitro and in vivo studies respectively) and would probably be difficult to find in an environmental exposure context. Finally, it should be noted that the different toxic effects induced by NPs were strongly related to their physico-chemical properties which, in most of the studies we reviewed, were not adequately analyzed and defined. In fact, NP characterization often focuses only on the chemical composition and particle size (see Table 2) while, in our opinion it is extremely important to assess other relevant parameters such as surface area, reactivity and functionalization, charge, solubility, degree of agglomeration and number concentration.

2.2. Effects of NPs on the Female Reproductive System

In recent years, numerous in vitro and in vivo studies have evaluated the potential adverse effects of NP exposure on the female reproductive system. Study findings have demonstrated that several NPs can interfere with normal female reproductive function by inducing cytotoxic effects on ovarian structural cells, impairing oogenesis and follicle maturation, and altering normal sex hormone levels (see Table 3).
Different types of NPs, in particular TiO2-NPs, carbon-based NPs and silica NPs, were studied in order to define their cytotoxic potential on Chinese hamster ovary cells (CHO), epithelial-like ovarian constituent cells. With regard to metal-based NPs, data currently available have shown the ability of TiO2-NPs to be internalised by CHO-K1 and to induce a dose-dependent decrease in cell viability following both acute [68,71,72] and subacute exposure [76,77]. Anatase TiO2-NPs reduced cell viability in relation to the increasing dose applied (0–100 μg/mL) and the increasing ROS concentrations induced [68]. Similarly, anatase and rutile TiO2 NPs caused a cytotoxic effect in CHO cells treated with concentrations ranging from 25 to 325 μg/mL [71]. The results of this study showed that particle size and crystal phase composition can significantly affect the cytotoxic behaviour of NPs since 10–20 nm anatase yielded the highest level of cytotoxicity followed by 50–60 nm anatase and 50–60 nm rutile. Comparable results were obtained by Di Virgilio et al. [72] on treating CHO-K1 cells with 0–100 μg/mL of non-characterized (in terms of crystal phase composition) TiO2-NPs. Moreover, cell viability was negatively affected by acute and subacute exposure to concentrations of >50 μg/mL of anatase TiO2-NPs [76,77]. The most significant toxic effects were observed following treatment with particularly high doses of TiO2-NPs (100 and 200 μg/mL). However, these results should be interpreted with caution since the applied doses may not be representative of exposure levels in the general population or in occupationally exposed subjects. Aluminium oxide NPs (Al2O3-NPs), another type of metal-based NP, were able to penetrate the cytoplasm of CHO-K1 cells treated with 0–100 μg/mL [72], inducing dose-dependent cytotoxic damage. However, this was not so severe as the damage caused by TiO2-NPs and began at higher concentrations suggesting that the chemical composition of NPs is also important in causing toxic effects.
In a study on carbon-based NPs that examined the effects on CHO cell viability, of silicon carbide nanowires (SiCNWs) obtained from the reaction of MWCNTs with silicon monoxide, Jiang et al. [73] reported an important decrease in the reproduction rate. The main mechanism leading to SiCNW cellular toxicity was identified in the induction of the mitogen-activated protein kinase cellular signaling pathway and in the over expression of cyclossigenase-2. In the same cell line, transfected with the macrophage receptor with collagenous structure (MARCO), treatment with increasing concentrations (1–100 μg/mL) of MWCNTs caused significant cytotoxicity that was probably related to the incomplete inclusion of NPs by the cell membrane [79]. Furthermore, treatment of female granulosa cells, collected from infertile women, with calcium phosphate-NPs (10 and 100 μM), caused an accumulation in the S phase of the cell cycle with an increased apoptotic rate [74]. With regard to cytotoxicity in ovarian cells it is interesting to note that metal-based and carbon-based NPs may produce an effect through different molecular mechanisms of action such as the induction of oxidative stress and the activation of MAPKs cellular signaling/over-expression of COX-2, respectively.
In contrast, when CHO cells were seeded on an aligned MWCNT substrate, no toxicity was observed: the cells grew well and were aligned, and only a few cells elongated along the axis of the MWCNT bundles [75]. Studies have also demonstrated good biocompatibility of mesoporous silica NPs with human ovary cells. In fact, when naked mesoporous silica NPs were applied to ovarian NIH-OVCAR3 epithelial cancer cells (30, 75 μg/mL), no altered cell morphology, metabolism or cell loss was reported, although there was evidence of internalization of the NPs [78]. Comparable results were obtained in NIH-OVCAR3 and SKOV3 cancer cells treated with 20 μg/mL of greater or differently functionalized (carboxyl or amine modified) mesoporous silica NPs. In the same study, carboxyl-modified polystyrene-NPs were not toxic for NIH-OVCAR3 and SKOV3 cells at a concentration of 75 μg/mL, while polystyrene NPs functionalized with amine groups showed significant cytotoxic effects [78]. The absence of toxic effects of silica NPs in the latter study, compared to those previously analyzed, may be related to a number of factors, including the different type of NPs used (mesoporous silica NPs vs metal and carbon-based NPs), cell models adopted (NIH-OVCAR3 and SKOV3 vs CHO) and lower exposure concentrations, whereas the different functionalization of polystyrene-NPs seems to play a decisive role in the induction of cytotoxic effects.
Conflicting results were obtained for the genotoxic effects of metal-based NPs on the CHO cell line. In fact, at doses < 5000 μg/mL, anatase and rutile TiO2-NPs failed to produce any increase in chromosomal aberration frequencies in CHO-WBL cells, either in the absence or presence of UV [69]. Similarly, no structural or numerical chromosome aberrations were observed in CHO cells exposed to several doses (25–2500 μg/mL) of rutile TiO2-NPs [70] and no genotoxic effects were detected in the CHO-K1 cell line treated with increasing concentrations (0, 10, 20, or 40 μg/mL) of anatase TiO2-NPs [76]. Conversely, studies carried out by Zhu et al. [71] and Di Virgilio et al. [72] reported an increased frequency of sister chromatid exchange, micronuclei and DNA strand breaks. Moreover, Al2O3-NPs induced a dose-dependent response of micronuclei frequency in the whole dose-range studied [72]. Comparable results were obtained with SiCNW exposure [73]. The different experimental design as well as the chemical composition of the NPs and the crystal form of the TiO2-NPs were probably responsible for most of the inconsistencies observed. In particular, it is worth noting that 3 h [69] and 4 h [70] exposure periods may not be long enough to detect genotoxic effects, indicating that the response is also dependent on the exposure time.
Oocyte viability and maturation are extremely sensitive to microenvironment changes and extracellular chemical compounds [81]. Consequently, several in vitro studies have been carried out to assess the potential adverse effects of NPs on female germ cells (see Table 3). The findings of these studies, which indicated an alteration in oocyte maturation and fertilization and their subsequent embryonic development, point to the role of TiO2-NPs and Quantum Dots (QDs) as endocrine disruptors. In fact, Hou et al. [80] demonstrated that TiO2-NPs (12.5–50 μg/mL) inhibited rat follicle development and oocyte maturation, with a dose-dependent reduction in the survival rate of follicles, formation rate of antral follicles and release rate of cumulus-oocyte cell complexes. Similar results, (dose-dependent decrease in oocyte maturation rate, reduced fertilization, impairment of cell proliferation and dose-dependent enhanced blastocyst apoptotic rate) were observed on oocytes isolated from female ICR mice exposed to 0–500 nM of cadmium selenium core QDs (CdSe-core-QDs). However, the coating of QDs with a zinc sulfide shell prevented the cytotoxic effects on oocytes, thereby suggesting that QD surface chemistry plays an important role [81]. The results of this study are particularly intriguing since they point out the importance of surface functionalization of QDs in altering their biological effects. In fact, the Authors suggested that the ZnS coating of CdSe QDs prevented cytotoxicity by blocking surface oxidation and the subsequent release of Cd2+ ions, mechanisms that are correlated with the induction of cell death by CdSe-core QDs. Therefore, it is plausible to assume that NPs may exert their toxic effects by dissolving to become a source/reservoir of toxic cations. The possible release of ions from metal-based NPs and QDs is an important topic that needs careful consideration when evaluating the potential toxic effects of these NPs. The same research group investigated the effects of CdTe/ZnTe QD-Transferrin (QD-Tf) bioconjugates on follicle development and oocyte maturation in female Kunmimg mice. Their findings revealed a dose-dependent up-take of QDs-Tf by theca and granulosa cells and a significant decrease in the rate of antrum cavity formation and of the ratio of oocytes to first polar body [84]. These results suggested that the high uptake of QDs-Tf could potentially disturb oocytes to form antrum cavity and advance in maturation, as previously reported by Xu et al. [82] and Wang et al. [83] who, after treating Kunming mice immatured oocytes with CdSe/CdS/ZnS QDs, observed a significant reduction in the oocyte maturation process. The high uptake of QDs-Tf was probably due to the small hydrodynamic diameter and the bioconjugation of QDs that can facilitate penetration and increase impact on oocytes.
Impairment of the female reproductive function, due to some metal-based NPs and QDs that modify oogenesis and follicle maturation, has also been observed in several in vivo studies (see Table 3). Wang et al. [85] reported that administration of TiO2-NPs to zebrafish Danio rerio at concentrations of 0.1 and 1.0 mg/L, skewed the distribution of follicular developmental stages toward immature statuses and reduced the expression of gene coding for growth factors implicated as paracrine stimuli in oocyte maturation. Interestingly, when zebrafish Danio rerio were exposed to the same NP concentrations for a shorter period of time, female gonads showed a normal spread of oocyte development stages in all exposed groups [88]. However, the group treated with the highest TiO2-NPs dose (1.0 mg/L) yielded a much lower production of eggs and viable embryos. The differences in oocyte development stages observed in these studies could be due to the different exposure periods (13 weeks vs. 14 days). Accumulation of anatase TiO2-NPs in the ovary tissues of CD-1 (ICR) female mice, intragastrically treated with 10 mg/kg of these NPs, caused atresia of primary and secondary follicle development and led to increased Ca, Na, K, and Zn contents, and decreased Mg, Cu, and Fe contents [86]. Modification of the equilibrium of these elements in the ovaries might adversely affect their normal physiology and consequently lead to alterations in the female germ cell maturation process. Gene expression analysis in ovary tissues of adult sheepshead minnows (Cyprinodon Variegatus) exposed to 10 μg/L of Ag-NPs, revealed a dramatic transcriptional response indicating that exposure to these NPs can cause reproductive dysfunction in estuarine fish, even though the absence of ovary morphological and developmental alterations did not support this hypothesis [87].
It has been demonstrated that a number of NPs are able to alter physiological sex hormone levels in the female as well as the male reproductive system (see Table 3 and Figure 2). In in vitro studies, Au-NPs and QDs were found to alter estrogenic hormonal levels. The exposure of rat ovarian granulosa cells to 2.85 × 1010 NPs/mL of metal-based NPs induced a greater output of estradiol after 1–5 h of treatment, while a significant decrease was observed after 24 h. Initial estradiol accumulation could be ascribed to alterations induced by the internalized Au-NPs in the cleavage activity of cholesterol into pregnenolone, and in mitochondrial permeability, while the subsequent inhibition of estradiol production was probably due to impairment of the mitochondrial steroidogenic machinery [89]. The same mechanism of action has been proposed to explain the increase, at the 4th–8th day post-treatment, in 17 β-estradiol secretion into granulosa cells treated with the highest QD-Tf concentration. 17 β-estradiol secretion was less than in controls at lower QD-Tf concentrations [84]. Unlike previous studies, the production of progesterone and estradiol in the granulosa cells was not affected by treatment with calcium phosphate-NPs and no alterations were observed in the levels and expression of mRNA encoding P450scc, P450arom, and StAR [74]. These results did not agree with the findings of Stelzer and Hutz [89] and Xu et al. [84] and it is possible that these disparities may be due to differences in dosage, chemicals, particle size, and cell model in the aforementioned studies.
The issue concerning alterations in sex hormone levels caused by exposure to metal-based NPs and NRDE-NPs, has also been addressed by some in vivo studies which have, however, provided conflicting results. In fact, serum levels of estradiol significantly increased, while progesterone, FSH, LH and testosterone levels diminished in female CD-1 (ICR) mice sub-chronically treated with TiO2-NPs [86]. Nevertheless, the administration of zinc oxide NPs (ZnO-NPs) to female Wistar rats at a concentration of 333.33 mg/kg did not alter serum sex hormone levels such as FSH, LH and estradiol [91]. Once again the conflicting results observed in these two studies may be related to several factors such as the different type and size of NPs, and the varying doses of exposure and length of treatment. With regard to the latter parameter, it is worth noting that a subchronic intragastric exposure (90 days) [86] was able to induce a significant increase in estradiol levels, whereas acute administration via the same route [91] failed to determine hormonal alterations. This highlights the need, when extrapolating results from in vivo studies, to carefully evaluate also the periods of time used for an investigation. Finally, with regard to NRDE-NPs, exposure of pregnant Fischer 344 rats to 148.86 μg/m3 of these NPs led to an increase in LH and a reduction in progesterone levels, whereas no significant differences were observed in FSH, prolactin, ir-inhibin and testosterone levels [90].
As observed for the male reproductive system the effects reported in these studies are largely dependent on many factors related to the experimental design and the physico-chemical characteristics of the NPs investigated. Evaluation of these results should therefore take these variables into account. Moreover, when analysing the endocrine effects on the female reproductive system, it should be noted that the exposure concentrations used in both in vitro and in vivo studies were very high, up to 5 mg/L and 333.33 mg/kg, respectively.

2.3. Estrogenic Effects of NPs

Several in vivo studies have investigated the potential estrogenic effects of different types of NPs in male and female organisms (see Table 4). However, their findings are limited, fragmentary and often conflicting, especially with regard to the adverse effects on male organisms.
The estrogenic effects of Ag-NPs and QDs were investigated in aquatic species through the analysis of two proteins: vitellogenin (VTG), the yolk-precursor lipoprotein, and choriogenin (Chg), the precursor protein of the inner layer subunits egg envelope (chorion) of the teleosts which are under estrogenic control and consequently have a role as a biomarker of estrogens and endocrine disruption in oviparous organisms. In this context, Pham et al. [92] observed a conspicuous mRNA induction of VTG-1 and Chg-L in the livers of male Medaka (Oryzias latipes) fish exposed to Ag-NPs at concentrations of 1 and 25 μg/L. On the contrary, exposure of sexually immature rainbow trout (Onchorynchus mykiss) to concentrations of Ag-NPs ranging from 0 to 6 μg/L induced a significant reduction in liver expression of VTG-like proteins [93]. Similarly, studies to investigate the estrogenic effects of QDs also provided conflicting results since the administration of (CdS)/CdTe capped-QDs at doses of 1, 2 and 6 μg/L induced a clear up-regulation of VTG and a down-regulation of its receptor in juvenile rainbow trout (Oncorhynchus mykiss) [94], while the exposure of male sticklebacks (Gasterosteus aculeatus) to 0–500 μg/L of CdS-QDs failed to show any alteration in VTG expression [95]. With regard to the effects caused by Ag-NPs the different induction of VTG could be due to the variation in exposure doses used in the studies of Pham et al. [92] and Gagnè et al. [93] (1, 25 μg/L vs. 0.06, 0.6 and 6 μg/L, respectively). Neverthless, in the QD studies the up-regulation of VTG was evident at the lowest exposure concentrations (1, 2 and 6 μg/L vs. 0–500 μg/L) thus suggesting that the effects may be correlated to other aspects such as the different functionalization of QDs. However, an assessment of these estrogenic effects should take into consideration differencies in species and sexual maturation.
Likewise, the evaluation of progesterone production in male laboratory animals exposed to NRDE-NPs did not provide consistent results. In fact, Li et al. [96] demonstrated a lower basal release of progesterone and corticosterone from adrenal cells obtained from male C57BL/Jcl mice exposed to 152.01 μg/m3 of NRDE-NPs, while greater concentrations were determined in the low-dose (41.73 μg/m3) NRDE-NP treated group compared to controls. However, the same research group observed increased plasma progesterone levels in male F344 rats exposed to middle (36.35 μg/m3) or high (168.84 μg/m3) NRDE-NP concentrations and decreased hormone levels in rats administered with low (15.37 μg/m3) or high (168.84 μg/m3) NRDE-NP doses after 4 and 8 weeks treatment, respectively [33]. Furthermore, immature male F344 rats prenatally exposed to the same NPs (148.86 μg/m3 to pregnant mice) showed significantly reduced serum progesterone concentrations [34]. Finally, a study on the estrogenic effects of carbon-based NPs reported that treatment of male zebrafish (Danio rerio) with C60 fullerene reduced the biodisponibility of the synthetic estrogen, 17α-ethinylestradiol [97,98].
In female organisms, the estrogenic effects of QDs and CB-NPs appear to be more evident. In fact Jain et al. [99] demonstrated the metalloestrogenic effects of CdTe-QDs on human breast cancer MCF-7 cells. These NPs were found to induce estrogen-related genomic and non-genomic signaling pathways and increase estrogen receptor-α activation and biphasic phosphorylation of AKT and ERK1/2, similar to 17β-estradiol action [99]. In the same study, the treatment of BALB/C prepubescent and virgin ovariectomized female mice with CdTe-QDs caused a two to three fold increase in uterine weight, comparable or greater than that induced by 17β-estradiol. Interestingly, these results suggest that Cd2+ released by CdTe-QDs is able to bind to the estrogenic receptor (ER). Exposure of C57BL/6 mice to CB-NPs at a total dose of 11, 54 and 268 μg/kg body weight during gestation, induced a significantly earlier onset of puberty in the offspring, assessed as time of vaginal opening [100].

3. Impact of Endocrine Disrupting NPs on Thyroid Function

Thyroid hormones are involved in numerous physiological processes as regulators of metabolism, bone remodeling, cardiac function and mental status. Moreover, thyroid hormones are of special importance in fetal development, particularly in brain development, since the absence or alteration of their physiological levels reduces neuronal growth and differentiation in the cerebral cortex, hippocampus, and cerebellum [101,102]. In recent years, epidemiological studies have indicated that even minor changes in thyroid homeostasis may affect fetal neurological development [103]. Since exposure to thyroid EDCs may significantly affect public health, and studies on most chemicals with thyroid-disrupting potential have been sporadic and have failed to provide consistent results [104], there is clearly a need to evaluate the possible action of endocrine disrupting NPs on thyroid function.
To date, in both in vitro and in vivo models, conflicting results have been reported regarding the role that different metal-based NPs and QDs play in influencing thyroid hormonal pathways (see Table 5). Hinther et al. [105] investigated the effects of exposure to Ag-NPs, ZnO-NPs and QDs on thyroid hormone (TH) signalling in frog tissue using a cultured tail fin biopsy assay derived from Rana catesbeiana tadpoles. Results showed that exposure to 5–10 nM of Ag-NPs and to 0.1 nM of QDs alone induced a reduction in levels of transcripts encoding the TH-induced receptor β (TRβ) and TH-repressed Rana larval keratin type I (RLKI), whereas treatment with ZnO-NPs had no effect on TRβ and RLKI transcript levels. Furthermore, Ag-NPs were able to affect TH-action on TRβ and RLKI transcripts according to a nonmonotonic “inverted-U” hormesis response that is characteristic of endocrine disruption. In contrast, the administration of chromium NPs (Cr-NPs) to heat-stressed Sprague-Dawley rats did not bring about significant modifications in thyrotropic-stimulating hormone (TSH), free triiodothyronine (FT3) and free thyroxine (FT4) serum levels, indicating that these NPs do not affect the metabolism of thyroid hormones [106].
Clearly, current data concerning the impact of endocrine disrupting NPs on thyroid function are conflicting and very limited. Moreover, since and adequate comparison of findings cannot be made on account of differences in experimental conditions, it is not possible to evaluate the overall effects of NPs on thyroid.

4. Impact of Endocrine Disrupting NPs on Insulin Action and Metabolism

The findings of several toxicology and epidemiology studies have suggested that various EDCs are involved in an increasing number of metabolic disorders, including insulin resistance (IR) and IR-related co morbidities, such as obesity, type 2 diabetes mellitus (T2DM) and polycystic ovary syndrome [12,107,108]. These particular EDCs are also known as metabolic disruptors [109]. Recently, a number of in vitro and in vivo studies have evaluated possible metabolic disruption of insulin-signalling pathways and insulin production caused by exposure to different metal-based NPs (see Table 5).
In this context, Gurevitch et al. [110] investigated the potential role of TiO2-NPs in the etiology of metabolic/endocrine disorders such as obesity and closely- related IR. In this study, the exposure of Fao rat hepatoma cells to 50 and 200 μg/mL of TiO2-NPs impaired insulin response and induced insulin resistance through direct interference with insulin-signalling pathways and indirect inflammatory activation of the macrophages. In another in vitro study, an increase in cell viability, ATP/ADP ratio and secretion of insulin in response to glucose stimuli was observed in isolated pancreatic islets treated with CeO2-NPs at a concentration of 100 nmol/L, either alone or in combination with 30 nmol/L sodium selenite [111].These findings could possibly be ascribed to the proinflammatory action of TiO2-NPs [110] and to the anti-oxidant potential of CeO2-NPs [111] which may exert a different effect on the insulin cascade and release, respectively.
In contrast, dietary addition of 300 and 450 μg/kg of Cr-NPs caused a significant decrease in serum insulin levels in Sprague-Dawley rats [113]. Similarly, lower concentrations of insulin and cortisol and higher levels of insulin growth-factor (IGF)-1 were detected in the same animal model treated with a basal diet supplemented with 150–450 μg/kg of Cr-NPs [106]. Comparable results were obtained by Wang et al. [112] when administering 200 μg/kg of Cr-NPs to crossbred pigs, while an increase in serum levels of IGF-1 was reported in Swiss mice intranasally instilled with 1.5 mg/kg of double walled-CNTs [114]. The reduction in peripheral insulin levels may have been due to an enhanced binding of insulin to its receptors on targeted tissues such as muscle and fat since Cr is thought to facilitate the interactions between insulin and insulin receptors and to promote hormone internalization into cells [125]. The same Authors provided confirmation of the protective effects of CeO2-NPs on pancreatic islets in streptozotocin induced diabetic Wistar rats intraperitoneally injected with these NPs (60 mg/kg) alone, or in combination with sodium selenite (5 μmol/kg/day). The findings of this study showed a significant improvement in diabetes biomarkers including oxidative stress, energy compensation (ADP/ATP), and the glycemic and lipid profile [115].

5. Impact of Endocrine Disrupting NPs on the Neuroendocrine System

Chromaffin cells are neuroendocrine cells found mainly in the medulla of the adrenal gland and in other ganglia of the sympathetic nervous system. In these cells the release of numerous catecholamines into the blood e.g., adrenaline, noradrenaline, enkephalin and a range of biologically active peptides occurs through multiple intracellular (chromaffin cell excitability, Ca2+ signaling, exocytosis, endocytosis) and intercellular pathways (splanchnic nerve-mediated synaptic transmission, paracrine and endocrine communication, gap junctional coupling) in response to a wide variety of stress-related signals [126,127]. Chronic hypoxia is also included among these signals and can significantly affect catecholamine secretion by increasing the number of secreted vesicles at low voltages, although it does not alter the mechanism of catecholamine release [128]. Catecholamine secretion usually takes place in situations of fear, anxiety or organic stress. However, the release of hormones from adrenomedullary chromaffin cells can also be triggered by a number of chemicals that reach the adrenal medulla via the bloodstream [129]. For this reason, in recent years several in vitro studies have been performed in order to verify the endocrine disrupting activity of NPs (especially metal-based NPs), on chromaffin cells (see Table 5). Study findings have shown that NPs have the potential to disrupt the physiological function of neuroendocrine cells by altering their catecholamine levels or compromising the exocytotic machinery.
Hussain et al. [117] observed a dose-dependent depletion of dopamine (DA) and dihydroxyphenylacetic acid (DOPAC) in PC-12 cells exposed to 1–100 μg/mL of manganese oxide NPs (MnO-NPs). In the same study, treatment with Ag-NPs induced similar results but only at a concentration of 50 μg/mL, while homovanillic acid (HVA) levels decreased at 50 μg/mL in both MnO-NP and Ag-NP exposure. Comparable results were obtained by Wang et al. [118] who, on exposing PC-12 cells to increasing concentrations of copper NPs (Cu-NPs), found a significant reduction in the levels of DA, DOPAC and HVA, probably caused by oxidative stress and enzymatic alterations. The toxicological behavior of citrate-reduced Au-NPs and Ag-NPs was studied by Love and Haynes [119] on primary cultures of adrenal medullary chromaffin cells. Treatment with 0.01–1 nM of metal-based NPs did not affect cell viability, but caused less and slower secretion of epinephrine molecules, indicating the ability of these NPs to interfere with chemical messenger delivery and the vesicular kinetic. These findings were confirmed by a subsequent study conducted on murine adrenal medullary chromaffin cells exposed to different sizes of Ag-NPs (1 nM) and to polyethilen-glycol (PEG) and heparin surface-modified Au-NPs (10 μg/mL). In fact, the results of this study showed that noble metal NPs disrupt exocytosis, typically by altering the number of molecules and release kinetics, and enhance direct disruption of the vesicle matrix [123]. The functionality of the exocytotic machinery of mouse (C57BL/6J) chromaffin cells was also severely damaged by exposure to 5–36 nM of carboxyl QDs with CdSe core and ZnS shell since these were able to impair Ca2+ influx and consequently compromise the overall catecholamine supply from chromaffin cells [120]. Interestingly, Ag-NP (coated with citrate or polyvinylpyrrolidone) exposure (30 μM) also impairs neurodevelopment in PC12 cells as demonstrated by a depleted emergence of the acetylcholine-phenotype and an enhanced differentiation into a DA-phenotype after treatment with citrate-coated Ag-NPs and polyvinylpyrrolidone-coated Ag-NPs, respectively [121].
In a study on fullerene C60, Corona-Morales et al. [116] observed that exposure of Wistar rat adrenal chromaffin cells to 100 μM of these NPs, remarkably increased cell survival and prevented cell death, including apoptosis. The beneficial effects observed in this study were probably due to the strong antioxidant capacity of fullerene C60. Conversely, the exposure of cultured mouse chromaffin cells to increasing doses (30–263 μg/mL) of MWCNTs caused a dose-dependent reduction both in cell membrane input resistance and the number of spontaneously active cells [130]. Finally, exposure of the PC12 cell line to 25–200 μg/mL of silica NPs resulted in a dose-dependent reduction in DA synthesis associated with redundant ROS generation in the cells [122].

6. Other Effects of Endocrine Disrupting NPs

A limited number of studies have investigated the adverse effects of Cr-NPs on the pituitary gland (see Table 5) by evaluating secretion and the levels of growth hormone (GH). The exposure of Sprague-Dawley rats to 150, 300, and 450 μg/kg of Cr-NPs did not cause significant alterations in the concentrations of pituitary-produced GH [106]. Similar results (no significant difference in GH level between the exposed and control groups) were obtained in crossbred pigs (Duroc x Landrace x Yorkshire) treated with 200 μg/kg of Cr-NPs [112], while, in a subsequent study, the same research group observed that these NPs were able to alter GH pulsatile secretion and pituitary GH-mRNA expression by inducing an increase in the mean level, lowest value, peak value and peak duration of GH secretion and by significantly improving pituitary GH-mRNA expression [124]. These conflicting results are probably due to the different experimental design of the studies. In fact, given the pulsatile nature of GH release in most mammals, a measurement technique based on a single blood sample [112] is not sufficiently sensitive to assess the effects of treatment on GH.
The secretory function of adrenal glands may also be affected by exposure to NPs as demonstrated by some ex vivo and in vivo studies carried out using NRDE-NPs (see Table 5). In fact, the treatment of male C57BL/Jcl mice with 41.73 μg/m3 and 152.01 μg/m3 of NRDE-NPs inhibited, at the highest dose, both basal and ACTH-stimulated corticosterone release by adrenal cells, while the lowest NRDE-NP concentration increased synthesis and release of the hormone following a biphasic dose response curve [96]. However, the same research group demonstrated that there were no significant differences in plasma corticosterone concentrations in adult male Fischer rats and in male C57BL/Jcl mice exposed to 15.37, 36.35 and 168.84 μg/m3 and to 41.73 μg/m3 and 152.01 μg/m3 of NRDE-NPs, respectively [33,96]. Conversely, an increase in serum corticosterone levels was evident in pregnant Fischer rats treated with NRDE-NPs (148.86 μg/m3) [90], while in immature male Fischer rats, prenatally exposed to these NPs, there was a significant reduction in serum hormone concentrations [34]. Finally, with regard to cortisol secretion, Zha et al. [106] showed that the administration of 150–450 μg/kg of Cr-NPs to Sprague-Dawley rats reduced plasma cortisol levels.

7. Impact of Endocrine Disrupting NPs on Invertebrate Species

Currently, most of the studies to assess the adverse effects of EDCs are performed on vertebrates, while significantly less attention is paid to the effects on invertebrates. However, invertebrates account for most animal biodiversity and are important in routine ecotoxicological testing and environmental monitoring [131]. Thus, understanding the adverse effects on invertebrate endocrinology caused by endocrine disrupting NPs may be useful since this species could act as sentinel organisms to evaluate the environmental impact of this category of endocrine disruptors.
Several studies that evaluated the potential endocrine disrupting activity of different types (metal- and carbon-based NPs and QDs) of NPs in Daphnia magna, have reported a significant impact on the reproductive health of this invertebrate. A reduced number of offspring per female was observed in Daphnia magna chronically treated with different sized TiO2-NPs at concentrations ranging from 0.01 to 100 mg/L [132]. Similar results (reduced number of offspring and complete inhibition of reproduction at the highest dose) were obtained on exposing Daphnia magna to 0.1–5 mg/L of Degussa P25 TiO2-NPs [133] or to 0.02–2 mg/L of different types of the same metal-based NPs for 21 days [134]. A significant inhibition of reproduction was also observed in Daphnia magna exposed to 5–500 μg/mL of Ag-NPs [135]. A study on carbon-based NPs, [136] showed that the administration of 2.5 ppm of fullerene C60 could delay molting and reduce offspring production in Daphnia magna, whereas at sub-lethal doses of exposure, Tao et al. [137] reported observing the accumulation of reproductive responses such as delayed maturation of the female offspring of gestating exposed daphnids and the inability to reproduce of mother daphnids after fullerene C60 exposure. A decrease in the total number of neonates per female was also detected for nanodiamonds at concentrations ≥ 1.3 mg/L [138]. In order to assess the estrogenic effects of QDs on this aquatic species, Kim et al. [139] exposed juvenile Daphnia magna to concentrations ranging from 0 to 30 μg/L of CdSe/ZnSe QDs coated with tri-n-octylphosphine oxide/gum arabic. The results of this study failed to reveal changes in VTG mRNA expression, however, the administration of 3-mercaptopropionic acid-coated CdSe/ZnSe QDs with relevant levels of UV-B, induced a dose-dependent reduction in VTG expression. In another invertebrate aquatic organism, the marine amphipod Corophium volutator, exposure to 0.2–1 mg/L of ZnO-NPs caused a delay in sexual maturation and reduced fecundity [140].
The endocrine disrupting effects of metal-based NPs were also observed in Chironomus riparius and Drosophila Melanogaster. Exposure of Chironomus riparius midge larvae to 0–1 mg/L of these NPs reduced pupation, adult emergence, the male/female ratio and egg number, probably through an up-regulation of the gonadotropin releasing hormone (GnRH) mRNA gene expression [141]. Moreover, Ag-NPs may interfere with the Chironomus riparius midge ecdysteroid system by altering ecdysteroid receptor mRNA expression [142]. In Drosophila Melanogaster, the addition to food (0.005%–0.5% w/v) of TiO2-NPs and Ag-NPs led to a significant decrease in developmental success and, in the case of exposure to TiO2-NPs, to a significant progeny loss, as shown by the decline in female fecundity. Similarly, a substantial reduction in the Drosophila melanogaster lifespan and fertility performance was observed after the ingestion of 0.114–467 mg/g of citrate-capped Au-NPs [143].
A number of studies have reported reproductive toxicity in worms in a terrestrial environment due to various metal oxide NPs. Five days of dietary exposure to TiO2-NPs (>47.9 mg/L), ZnO-NPs (1.2 and 4.6 mg/L) and Al2O3-NPs (>102 mg/L) reduced nematode Caenorhabditis Elegans growth, the number of eggs and offspring per worm, indicating NP-induced reproductive impairment in this species [144,145]. Comparable results that demonstrated the ability of metal-based NPs (Ag-NPs, CeO2-NPs, TiO2-NPs and ZnO-NPs) to affect the reproductive potential of Caenorhabditis Elegans were obtained by Roh et al. [146,147] and by Ma et al. [148]. A subsequent study carried out by Lim et al. [149] suggested that oxidative stress was the leading mechanism in inducing Ag-NP reproductive impairment in this invertebrate species. In addition to metal-based NPs, QDs, carbon-based and silica NPs also produced significant detrimental effects on the reproductive function of Caenorhabditis Elegans. In fact, egg alterations were observed after exposure to 200 nM of CdSe/ZnS or CdTe-QDs [150], while a reduction in the reproductive rate, assessed as the number of eggs per animal, was induced by treatment with 100 μg/mL of hydroxylated fullerene [151]. Furthermore, the administration of 2.5 mg/mL of silica-NPs caused a reduction in progeny and a manifestly premature cessation of progeny production due to age-related degeneration of the reproductive organs [152]. Disruption of the reproductive function related to several types of metal, metal oxide and carbon-based NPs, was also observed in Eisenia fetida and Eisenia veneta earthworms. In fact, exposure to 2.5–25 mg/kg of Au-NPs caused a dose-dependent decrease in the number of offspring [153], while acute exposure to PVP-coated Ag-NPs (801 ± 74.9 and 773.3 ± 11.2 mg/kg) [154] and treatment with 3000 and 10,000 mg/kg of Al2O3-NPs (11 nm) resulted in a lower number of cocoons [155]. Similar results were also observed in Eisenia fetida and Eisenia veneta exposed to ZnO-NPs and TiO2-NPs [156] and to ZnO-NPs [157], respectively. In contrast, the exposure of Eisenia fetida to Cu-NPs at concentrations of 5–50 mg/kg dry mass did not cause any reproductive effects [158]. With regards to carbon-based NPs, treatment of Eisenia veneta earthworms with double walled-CNTs led to a significant reduction in cocoon production [159] and similarly, the cocoon number diminished in Eisenia veneta, Eisenia fetida and Lumbricus rebellus exposed to C60 fullerene [159161]. However, a study conducted by Pakarinen et al. [162] failed to confirm the reproductive toxicity of C60 fullerene in Lumbriculus variegatus administered with 10 and 50 mg/kg of the aforementioned carbon-based NPs.

8. Discussion and Conclusions

Recent advances in engineering and technology have led to the development of many types of NPs currently used in numerous industrial sectors. The rapidly developing field of nanotechnology, which is creating materials with size-dependent properties, is likely to become an important source of exposure to NPs. In fact, as more and more consumer products containing NPs become available on the market, exposure of the general population will inevitably increase. However, as with any new technology, the earliest and most extensive exposure to hazards is most likely to occur in the workplaces and workers in nanotechnology-related industries may be particularly exposed to uniquely engineered materials with innovative sizes, shapes, and physical and chemical properties. The growing NP exposure of workers and the general population to NPs has heightened concern about the potential human toxicity and environmental impact of these particles. Consequently, in the past few years, nanotoxicology has emerged as a new discipline designed to elucidate the relationship between the physical and chemical properties of NPs and the induction of toxic biological responses [163]. However, since there is still a lack of knowledge about the possible risks to human health and environmental safety posed by the expanding development and use of NPs, there seems to be an urgent need to gather information on this subject.
In this context, we have summarized the data currently available in the literature that report the adverse effects on the endocrine system caused by in vitro and in vivo exposure to different types of NPs. Our aim was to understand the risk of NP endocrine disruption, assess the underlying mechanisms that can affect endocrine function and identify research areas where further study needs to be carried out to reach a deeper understanding of the role of NPs as endocrine disruptors. Current data support the notion that different types of NPs are capable of altering the normal and physiological activity of the endocrine system (see Tables 2, 3, 4 and 5). The male and female reproductive systems are the endocrine organs that have received most attention. In the male reproductive system (see Table 2), findings showed that NPs are able to affect cell viability in gonadal tissues, testicular morphology and the process of spermatogenesis, while in the female reproductive system (see Table 3), NPs exerted cytotoxic and/or genotoxic effects on ovarian structural cells and damaged oogenesis and follicle maturation. Furthermore, NPs caused significant alterations in normal sex hormone levels in both systems. However, some studies failed to confirm these adverse effects, since no abnormalities in male and female reproductive function were found after NP exposure (see Tables 2 and 3). These conflicting results are probably due to the different intrinsic properties of NPs used in the in vitro and in vivo studies. In fact, research has shown that the physico-chemical characteristics (particle size, shape, surface area, charge, chemical properties, solubility, oxidant generation potential, and degree of agglomeration) of NPs can influence their effects on biological systems [20].
This supposition is also confirmed by the findings of some of the studies reviewed, since the physico-chemical properties of NPs seem to influence the toxic effects induced on endocrine function. For instance, NP size impacted spermatogenesis, sperm quality and testosterone levels in different ways [47,56], while surface functionalization appeared to bring about variations in both male and female reproductive NP effects [39,44,47,78,81]. Moreover, Love et al. [123] suggested that different NP physico-chemical properties, including size and surface coating, modulate changes in the cell communication of murine chromaffin cells exposed to different types of Ag-NPs and Au-NPs. Therefore, since the biological and toxic effects of NPs are highly dependent on their physico-chemical properties as well as on dosage, route of administration and exposure time [41], it is evident that a clear and precise evaluation of the effects of NPs as endocrine disruptors should also include a homogeneous exposure classification to ascertain exactly how the physico-chemical properties of NPs correlate with their adverse effects on the endocrine system. Useful parameters for a systematic categorization should include chemical composition, morphology, number concentration, surface area, mass concentration, weighted size distribution, state of agglomeration and surface reactivity (e.g., the ability to produce radicals and zeta potential). This is fundamental for an assessment of the endocrine toxicity of NPs, but unfortunately much of this characterization is largely lacking in current studies. For example, most available studies provide an indication of the chemical composition and size of the NPs used, while only a few of the reviewed studies go beyond this basic information to include data on other parameters such as functionalization [37,39,4244,57,69,78,81,82,84,94,120,121,123], number concentration [33,34,49,58,60,89,96], surface area [35,36,70,79,86,88], shape [72] and degree of aggregation [85].
Furthermore, in addition to the “intrinsic” NP properties, it may be extremely useful to evaluate the properties “extrinsically” acquired by NPs when interacting with biological fluids [164]. In fact, when NPs enter a physiological environment, they can rapidly adsorb proteins forming what is known as the protein corona. The protein corona provides the NPs with a biological identity that is substantially different from their synthetic identity [165], and, by affecting interaction with biomolecules, membranes and physical barriers, leads to specific toxic effects. Consequently, it is possible that quite different nanomaterials that share similar corona properties may have similar early biological interactions [164]. Interestingly, each physiological compartment has its own distinct set of proteins that interact in a unique way with the NPs. This is an important aspect to consider when comparing in vitro and in vivo experiments, but also when evaluating in vivo studies in which different routes of exposure were used [166]. In fact, since the protein corona reflects the traiectory of NPs in the body, a nanomaterial that enters the blood through the lung may have a dramatically different protein corona composition and resulting physiological response than the same nanomaterial directly injected into the blood [167]. Another important issue regarding the potential role of NPs in endocrine disruption is related to the molecular mechanisms of action underlying the adverse effects observed. In several studies, oxidative stress was hypothesized as the principal damaging mechanism [37,56,71,86]. However, oxidative stress is not the only mechanism of action capable of altering the functionality of the endocrine system, since Ag-NPs have been shown to inhibit cell proliferation by disrupting the proliferation signaling cascade of spermatogonial stem cells, rather than by inducing an oxidative reaction [47]. A similar mechanism has also been reported for SiCNWs on ovarian cells [73]. Furthermore, direct DNA damage has been reported as a potential toxic mechanism in mouse and human spermatozoa in in vitro [45,49] and in vivo models [55,56]. Numerous studies have also shown that NP endocrine toxicity occurs due to its ability to modify the gene expression of proteins and enzymes involved in sex hormone biosynthesis, metabolism and release [30,33,34,5961,86,89,90,120]. Another important mechanism of action by means of which EDCs are able to alter functions of the endocrine system is related to their ability to bind to hormone receptors. This mechanism of action may also be used by NPs and may play a significant role in the induction of endocrine effects. Indeed, the estrogenic effects exerted in mammalian cells and animal models may be dependent on NP interaction with nuclear and plasma membrane receptors, leading to activation of the estrogenic signaling pathway. For instance the findings of a study conducted by Jain et al. [99] suggest that the gradual and sustained release of ionic cadmium may contribute to the metalloestrogenic effects of QDs with a cadmium core. Cd2+ may bind to ER to trigger estrogenic effects including nongenomic activation of MAPK and PI3K as well as genomic activation, which involves ER dimerization and binding to the estrogen response element to induce gene transcription. To confirm that the estrogenic signaling induced by QDs was mediated via the estrogen receptor, the Authors found that the addition of the selective estrogen receptor antagonist, ICI 182780, completely annulled all QD-induced estrogenic effects [99].
The results of this study and the findings of Terzuoli [50], who suggested that the release of Ag+ under aqueous conditions could damage sperm membrane, endorse the idea that the release of ions by metallic NPs or QDs may play a relevant role in the induction of endocrine impairments in in vitro and in vivo models. Dissolution processes, consisting in the release of metallic cations, are greater in NPs than in bulk materials and are influenced by numerous factors such as reduced size, high surface-to-mass ratio, high radii of curvature, and corresponding low coordinated atoms at the surface [168]. This phenomenon is able to modify NP morphology and determine detrimental health effects [169171]. In fact, different studies have demonstrated the correlation between NP toxicity and ions released from Ag-NPs [172174], CdSe-QDs [175,176] and ZnO-NPs [170]. Therefore, the adverse effects on the endocrine system, potentially caused by the release of metal ions, should be more extensively evaluated in future studies. Moreover, NPs may also act by impairing the ligand-elicited signaling cascade as demonstrated by the direct interruption of insulin signaling in liver cells due to exposure to TiO2-NPs [110]. In this study, NPs reduced the phosphorylation on tyrosine residues of insulin receptor substrate proteins causing a decrease in the lipid messenger phosphatidylinositol 3,4,5 triphosphate wich is needed for most metabolic effects of insulin. The systematic identification of the molecular mechanisms that NPs may use to cause adverse effects on the endocrine system is particularly difficult because changes in their different physico-chemical properties may trigger different mechanisms of toxic action. Consequently, once more, accurate physico-chemical characterization of NPs seems essential in order to obtain a better understanding of the mechanisms of action.
Toxicity testing of NPs using in vitro or in vivo assays is aimed at identifying a potential risk by establishing dose-response relationships that characterize this type of hazard. However, toxicological studies (even those that have investigated effects on the endocrine system), frequently use excessive and unrealistically high mass doses of NPs [177]. It is therefore debatable whether the findings observed in these studies can be extrapolated to refer to humans in realistic, lower exposure scenarios since the applied doses have little relevance to real-world conditions. Although it is not easy to determine a realistic exposure dose to be used in these toxicological studies, as available data on exposure levels in workers and the general population are still very limited [20], it seems clear that future studies need to focus on the potentially adverse effects on the endocrine system of low-level NP exposure especially through the use of exposure doses similar to those identified in environmental monitoring studies. Furthermore, considering that in recent years additional metrics such as number concentration and surface area are being taken into account in the assessment of exposure to NPs [178], it would be interesting to express exposure doses also in terms of number concentration and surface area as has already been done in some studies [3336,49,58,60,70,79,86,88,89,96].
The results of some studies involving the nature of the dose-response curve indicate that exposure to NPs may exert a hormetic response in endocrine organs. In fact, when evaluating changes in testosterone levels caused by NRDE-NPs, Li et al. [33] showed a clear inverted-U dose-response curve, while Ag-NPs were able to affect the TH-action on TRβ and RLKI transcripts according to an inverted-U hormesis response [105]. Hormesis has been observed in a number of studies that investigated the health effects of NPs and recently, Iavicoli et al. [24] have reviewed studies that reveal similar hormetic dose-responses in a variety of nanomaterials (e.g., single walled carbon nanotubes; silver molybdenum and aluminum nanoparticles, graphite nanofibers, titanium dioxide nanoparticles). However, at present, little is known of the reasons underlying possible NP induction of hormesis for some endpoints. Further research is therefore needed to better understand the physico-chemical properties of NPs that may induce hormesis, and the hazard endpoints that are most likely to be involved. It should be noted that factors such as aggregation and agglomeration can significantly influence NP characteristics in exposure and hazard studies [26]. In this context, a possible explanation of NP-induced hormesis may also lie in the aggregation of NPs at higher concentrations. Correct evaluation of this phenomenon therefore requires appropriate characterisation of the NPs in the test medium, since these can greatly influence the extent of particle agglomeration. Finally, to verify the actual presence of hormesis in NP dose-responses and interpret study findings, it might be helpful to apply the methodology suggested by Nascarella and Calabrese [24,179]. Summing up, future studies could provide invaluable information by ascertaining whether this particular phenomenon also occurs in other endocrine organs, or whether it is a result of exposure to other, different types of NPs.
A critical issue concerning exposure to EDCs is the possibility that these substances may cause not only somatic but also intergenerational adverse effects on the endocrine system. In fact, in recent years, numerous studies have provided evidence that fetal or perinatal exposure to EDCs results in disturbed endocrine functions, especially in the sexual development of offspring [9]. In this context, some study findings have revealed that in utero exposure to CB-NPs and NRDE-NPs can also lead to testicular morphological and functional alterations in the male offspring of rats and mice [34,38]. Whether the toxic effects on pup gonads derived from direct NP action or were the result of alterations induced in the maternal endocrine/reproductive system is an open question. Therefore, since exposure to EDCs during critical periods of development may cause adverse consequences later in life, and since NPs may play a role as EDCs, future studies on the potential endocrine toxicity of NPs should also take into account the age, period of exposure and mechanisms of action of possible epigenetically-mediated intergenerational effects.
Of all the studies on different types of NPs, it is those that have investigated possible adverse effects on the endocrine system due to DE-NPs that have provided the most intriguing results from an environmental point of view. DE-NPs are the most common incidental combustion-derived NPs in the urban environment and can also occur in occupational settings [33]. The results of the studies reviewed [33,39,5961,90,96] point to an endocrine disrupting activity on the part of DE-NPs, although lack of evidence regarding the relationship between the exposure doses used in the aforementioned studies and real environmental concentrations make it difficult to extrapolate these results to humans. Furthermore, since environmental contamination is generally due to a mixture of compounds, the disrupting effects of DE-NPs may be due to chemical substances around or attached to them which then exert an additive, synergic or antagonistic influence. It follows that further investigation on the toxicological profile of DE-NPs and the environmental pollutant mixture that generally envelops them is urgently needed in order to clarify their impact on endocrine function.
The results of studies conducted on invertebrates provide valuable information for assessing the biota impact of NPs as endocrine disruptors. In fact, several parameters such as invertebrate growth and molting, changes in vitellogenin levels, and reduced fecundity are useful indicators of endocrine disruption activity. However, further research is also needed in this area to identify the endocrine disrupting mechanisms and thereby determine the specific biomarkers that would be effective in measuring these alterations.
Finally, a complete and systematic assessment of the endocrine disrupting activity of NPs should be based on an analysis of the adverse effects they exert on the entire endocrine system. Currently, there is a lack of balance between studies that have evaluated their toxic effects on the male and female reproductive system and studies that have investigated the same effects on other endocrine organs such as the thyroid, neuroendocrine system, pituitary and adrenal glands (see Table 5).
In conclusion, the data currently available indicate that several types of NPs can adversely affect the endocrine system, and in particular the male and female reproductive system. In fact, the results of the studies presented in this review suggest that NPs are able to disrupt the endocrine system by exerting cytotoxic effects and damaging the costituent cells of endocrine organs, thus influencing endocrine homeostasis, altering hormone byosinthesis, metabolism and release and interfering with the ligand elicited signaling. However, a more critical assessment of these findings suggests the need to interpret these results with caution since the information on the potential interations and toxicity of NPs on the endocrine system is quite limited and in some cases conflicting. This means it is not possible to to reach a broad and shared consensus on the role of NPs as endocrine disruptors on the basis of data currently available and further investigation is clearly required. Moreover, the current difficulty in reaching definitive conclusions on this topic is due to the heterogeneity of experimental study designs and to the numerous variables that can influence the toxicological profile of NPs. Obtaining an overview of results is complicated and challenging when quite different cell lines or laboratory animals, routes, times and doses of exposure (most of which are excessively high or not representative of a real human exposure) have been used in the studies reviewed. Clearly there is a need to establish a systematic and rational approach in the design of future studies. Moreover, the adverse effects of NPs on human health often depend on the chemical structure, shape, size, agglomeration state, surface area and functionalization of NPs. These are therefore among the factors to be considered in studying nanotoxic effects on the endocrine system. Finally, a key issue such as the molecular mechanisms underlying the potential endocrine toxicity of NPs is still largely unknown or unexplored.
It is hoped that this systematic review will provide helpful information and a stimulus for discussion on aspects that should be the focus of future studies. In our opinion, it is necessary to (i) carry out in vitro and in vivo studies using NPs whose chemical and physical properties have been characterized in a clear and detailed manner, (ii) clarify all the different potential molecular mechanisms of action underlying the adverse effects of NPs on endocrine organs, including those responsible for intergenerational effects (iii) study the biological responses of the endocrine system in relation to low-level exposures to NPs (iv) identify the presence of hormesis and (v) evaluate the endocrine disrupting effects of NPs on the entire endocrine system.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

8-OHdG
8-hydroxy-2-deoxyguanosine
ACTH
AdrenoCorticoTropic Hormone
ADP
Adenosine Diphosphate
Ag-HC-NPs
Hydrocarbon-coated silver Nanoparticles
Ag-NPs
Silver Nanoparticles
Ag-PS-NPs
Polysaccharide-coated silver Nanoparticles
Al-NPs
Aluminum Nanoparticles
Al2O3-NPs
Aluminium Oxide Nanoparticles
ATP
Adenosine Triphosphate
Au-NPs
Gold Nanoparticles
C60HyFn
hydrated C60 fullerene
CB
Carbon Black
CB-NPs
Carbon Black Nanoparticles
CdS-QDs
Cadmium Sulfide Quantum Dots
CdS/CdTe capped-QDs
Cadmium Sulfide/Cadmium Tellurium capped-Quantum Dots
CdSe-core-QDs
Cadmium Selenium core Quantum Dots
CdTe-QDs
Cadmium Tellurium Quantum Dots
CdTe/ZnTe QDs
Cadmium Tellurium/Zinc Tellurium Quantum Dots
CeO2-NPs
Cerium Oxide Nanoparticles
Chg
Choriogenin
CHO
Chinese Hamster Ovary cells
CNTs
Carbon Nanotubes
COX-2
Cyclooxygenase-2
CrCl3-NPs
Chromium Chloride Nanoparticles
Cr-NPs
Chromium Nanoparticles
Cu-NPs
Copper nanoparticles
DA
Dopamine
DE-NPs
Diesel Exhaust Nanoparticles
DOPAC
dihydroxyphenylacetic acid
DSP
Daily Sperm Production
EDCs
Endocrine Disrupting Chemicals
EDPL
Endocrine Disruptor Priority List
ER
Estrogenic Receptor
ERK
extracellular signal-regulated kinase
EuOH3-NPs
Europium Hydroxied Nanoparticles
Eu2O3-NPs
Europium Oxide Nanoparticles
Fe3O4-NPs
Magnetic Iron Oxide Nanoparticles
FSH
Follicle Stimulating Hormone
FT3
free triiodothyronine
FT4
free thyroxine
GH
Growth Hormone
GnRH
Gonadotropin Releasing Hormone
HMG-CoA
3-hydroxy-3-methylglutaryl-coenzyme A
HVA
HomoVanillic Acid
IGF-I
Insulin-like Growth Factor I
IR
Insulin Resistance
LDL-R
Low Density Lipoprotein Receptor
LH
Luteinizing Hormone
MAPKs
Mitogen-Activated Protein Kinases
MARCO
Macrophage Receptor with Collagenous structure
MN
Micro Nuclei
MnO-NPs
Manganese Oxide Nanoparticles
MoO3-NPs
Molybdenum Trioxide Nanoparticles
mPEG@Au-NP
ω-methoxy poly(ethylene glycol) capped gold-NPs
MTT
3-(4,5-dimethyl-2-thiazol)-2,5-diphenyl- 2H-tetrazolium bromide
MWCNTs
Multi Walled Carbon Nanotubes
NPs
NanoParticles
NRDE-NPs
Nanoparticle-Rich Diesel Exhaust Nanoparticles
nSP
amorphous nanosilica particles
Ntera2, NT2
human testicular embryonic carcinoma cell line
P450 17α
Cytochrome P450 17α
P450 17β-HSD
Cytochrome P450 17β-HydroxySteroid Dehydrogenase
P450arom
Aromatase P450
P450scc
Cytochrome P450 side-chain cleavage
PBR
Peripheral-type Benzodiazepine Receptor
PEG
PolyEthilen-Glycol
PEG-NH2@Au-NP
ω-aminoethyl poly(ethylene glycol) capped gold-NPs
POP
Persistent Organic Pollutants
QDs
Quantum Dots
RLKI
Rana Larval Keratin Type I
ROS
Reactive Oxygen Species
SCE
Sister Chromatid Exchanges
SiCNWs
Silicon Carbide Nanowires
s-MWCNTs-PEG
synthesized functionalized MWCNTs with polyethylene glycol
SR-B1
Scavenger Receptor class B type 1
SSC
Spermatogonial Stem Cells
StAR
Steroidogenic Acute Regulatory protein
T2DM
Type 2 Diabetes Mellitus
TH
Thyroid Hormone
TiO2
Titanium Dioxide
TiO2-NPs
Titanium Dioxide nanoparticles
TRβ
Thyroid hormone Receptor β
TSH
Thyrotropic-Stimulating Hormone
UV
Ultra Violet
VTG
Vitellogenin
ZnO-NPs
Zinc Oxide Nanoparticles
ZnS
Zinc Sulfide
ZnS-QDs
Zinc Sulfide Quantum Dots

References

  1. Iavicoli, I.; Fontana, L.; Bergamaschi, A. The effects of metals as endocrine disruptors. J. Toxicol. Environ. Health B 2009, 12, 206–223. [Google Scholar]
  2. De Coster, S.; van Larebeke, N. Endocrine-disrupting chemicals: Associated disorders and mechanisms of action. J. Environ. Public Health 2012. [Google Scholar] [CrossRef]
  3. World Health Organization (WHO), State of the Science of Endocrine Disrupting Chemicals–2012; World Health Organization: Geneva, Switzerland, 2012; pp. 23–237.
  4. Wormley, D.D.; Ramesh, A.; Hood, D.B. Environmental contaminant-mixture effects on CNS development, plasticity and behavior. Toxicol. Appl. Pharmacol 2004, 197, 49–65. [Google Scholar]
  5. Diamanti-Kandarakis, E.; Bourguignon, J.P.; Giudice, L.C.; Hauser, R.; Prins, G.S.; Soto, A.M.; Zoeller, R.T.; Gore, A.C. Endocrine-disrupting chemicals: An Endocrine Society scientific statement. Endocr. Rev 2009, 30, 293–342. [Google Scholar]
  6. Soto, A.M.; Sonnenschein, C. Environmental causes of cancer: Endocrine disruptors as carcinogens. Nat. Rev. Endocrinol 2010, 6, 363–370. [Google Scholar]
  7. Frye, C.A.; Bo, E.; Calamandrei, G.; Calzà, L.; Dessì-Fulgheri, F.; Fernández, M.; Fusani, L.; Kah, O.; Kajta, M.; Le Page, Y.; et al. Endocrine disrupters: A review of some sources, effects, and mechanisms of actions on behaviour and neuroendocrine systems. J. Neuroendocrinol 2012, 24, 144–159. [Google Scholar]
  8. Karoutsou, E.; Polymeris, A. Environmental endocrine disruptors and obesity. Endocr. Regul 2012, 46, 37–46. [Google Scholar]
  9. Meeker, J.D. Exposure to environmental endocrine disruptors and child development. Arch. Pediatr. Adolesc. Med 2012, 166, 952–958. [Google Scholar]
  10. Rogers, J.A.; Metz, L.; Yong, V.W. Endocrine disrupting chemicals and immune responses: A focus on bisphenol-A and its potential mechanisms. Mol. Immunol 2013, 53, 421–430. [Google Scholar]
  11. World Health Organization (WHO), Global Assessment of the State-of-the-Science of Endocrine Disruptors; World Health Organization: Geneva, Switzerland, 2002; pp. 1–3.
  12. Casals-Casas, C.; Desvergne, B. Endocrine disruptors: From endocrine to metabolic disruption. Annu. Rev. Physiol 2011, 73, 135–162. [Google Scholar]
  13. Baccarelli, A.; Pesatori, A.C.; Bertazzi, P.A. Occupational and environmental agents as endocrine disruptors: Experimental and human evidence. J. Endocrinol. Invest 2000, 23, 771–781. [Google Scholar]
  14. Ying, G.G. Endocrine Disrupting Chemicals. What? Where? In Analysis of Endocrine Disrupting Compounds in Food; Nollet, L.M.L., Ed.; Wiley-Blackwell: Oxford, UK, 2011; pp. 3–17. [Google Scholar]
  15. Groshart, C.; Okkerman, P.C. Towards the Establishment of a Priority List of Substances for Further Evaluation of Their Role in Endocrine Disruption—Preparation of a Candidate List of Substances as a Basis for Priority Setting; BKH Consulting Engineers for European Commission Directorate-General for the Environment: Delft, The Netherlands, 2000; pp. 1–29. [Google Scholar]
  16. Johnson, I.; Harvey, P. Study on the Scientific Evaluation of 12 Substances in the Context of Endocrine Disrupter Priority List of Actions; WRc-NSF for European Commission Directorate-General for the Environment: Medmenham Marlow, UK; p. 2002.
  17. Okkerman, P.C.; van der Putte, I. Endocrine Disrupters: Study on Gathering Information on 435 Substances with Insufficient Data; RPS BKH Consulting Engineers for European Commission Directorate-General for the Environment: Delft, The Netherlands, 2002; pp. 1–131. [Google Scholar]
  18. Petersen, G.; Rasmussen, D.; Gustavson, K. Study on Enhancing the Endocrine Disrupter Priority List with a Focus on Low Production Volume Chemical; DHI water & environment for European Commission Directorate-General for the Environment: Hørsholm, Denmark, 2007; pp. 1–249. [Google Scholar]
  19. International Organization for Standardization (ISO), Nanotechnologies—Terminology and Definitions for Nano-objects—Nanoparticle, Nanofibre and Nanoplate; International Organization for Standardization: Geneva, Switzerland, 2008; pp. 1–7.
  20. National Institute for Occupational Safety and Health (NIOSH), Approaches to Safe Nanotechnology: Managing the Health and Safety Concerns Associated with Engineered Nanomaterials; National Institute for Occupational Safety and Health, Department of Health and Human Services, Centers for Disease Control and Prevention: Atlanta, GA, USA, 2009; pp. 1–86.
  21. Nel, A.; Xia, T.; Madler, L.; Li, N. Toxic potential of materials at the nanolevel. Science 2006, 311, 622–627. [Google Scholar]
  22. Canton, I.; Battaglia, G. Endocytosis at the nanoscale. Chem. Soc. Rev 2012, 41, 2718–2739. [Google Scholar]
  23. Zhao, F.; Zhao, Y.; Liu, Y.; Chang, X.; Chen, C.; Zhao, Y. Cellular uptake, intracellular trafficking, and cytotoxicity of nanomaterials. Small 2011, 7, 1322–1337. [Google Scholar]
  24. Iavicoli, I.; Calabrese, E.J.; Nascarella, M.A. Exposure to nanoparticles and hormesis. Dose Response 2010, 8, 501–517. [Google Scholar]
  25. Italian Workers’ Compensation Authority (INAIL), White Book, Exposure to Engineered Nanomaterials and Occupational Health and Safety Effects; Italian Workers’ Compensation Authority, Department of Occupational Medicine, formerly ISPESL: Rome, Italy, 2011; pp. 1–211.
  26. Landsiedel, R.; Fabian, E.; Ma-Hock, L.; van Ravenzwaay, B.; Wohlleben, W.; Wiench, K.; Oesch, F. Toxico-/biokinetics of nanomaterials. Arch. Toxicol 2012, 86, 1021–1060. [Google Scholar]
  27. National Institute for Occupational Safety and Health (NIOSH), Filling the Knowledge Gaps for Safe Nanotechnology in the Workplace. A Progress Report from the NIOSH Nanotechnology Research Center 2004–2011; National Institute for Occupational Safety and Health, Department of Health and Human Services, Centers for Disease Control and Prevention: Atlanta, GA, USA, 2012; pp. 1–395.
  28. Knez, J. Endocrine-disrupting chemicals and male reproductive health. Reprod. Biomed. Online 2013, 26, 440–448. [Google Scholar]
  29. Fowler, P.A.; Bellingham, M.; Sinclair, K.D.; Evans, N.P.; Pocar, P.; Fischer, B.; Schaedlich, K.; Schmidt, J.S.; Amezaga, M.R.; Bhattacharya, S.; et al. Impact of endocrine-disrupting compounds (EDCs) on female reproductive health. Mol. Cell. Endocrinol 2012, 355, 231–239. [Google Scholar]
  30. Komatsu, T.; Tabata, M.; Kubo-Irie, M.; Shimizu, T.; Suzuki, K.; Nihei, Y.; Takeda, K. The effects of nanoparticles on mouse testis Leydig cells in vitro. Toxicol. In Vitro 2008, 22, 1825–1831. [Google Scholar]
  31. Vevers, W.F.; Jha, A.N. Genotoxic and cytotoxic potential of titanium dioxide (TiO2) nanoparticles on fish cells in vitro. Ecotoxicology 2008, 17, 410–420. [Google Scholar]
  32. Rosenkranz, P.; Fernández-Cruz, M.L.; Conde, E.; Ramírez-Fernández, M.B.; Flores, J.C.; Fernández, M.; Navas, J.M. Effects of cerium oxide nanoparticles to fish and mammalian cell lines: An assessment of cytotoxicity and methodology. Toxicol. In Vitro 2012, 26, 888–896. [Google Scholar]
  33. Li, C.; Taneda, S.; Taya, K.; Watanabe, G.; Li, X.; Fujitani, Y.; Ito, Y.; Nakajima, T.; Suzuki, A.K. Effects of inhaled nanoparticle-rich diesel exhaust on regulation of testicular function in adult male rats. Inhal. Toxicol 2009, 21, 803–811. [Google Scholar]
  34. Li, C.; Taneda, S.; Taya, K.; Watanabe, G.; Li, X.; Fujitani, Y.; Nakajima, T.; Suzuki, A.K. Effects of in utero exposure to nanoparticle-rich diesel exhaust on testicular function in immature male rats. Toxicol. Lett 2009, 185, 1–8. [Google Scholar]
  35. Takeda, K.; Suzuki, K.I.; Ishihara, A.; Kubo-Irie, M.; Fujimoto, R.; Tabata, M.; Oshio, S.; Nihei, Y.; Ihara, T.; Sugamata, M. Nanoparticles transferred from pregnant mice to their offspring can damage the genital and cranial nerve systems. J. Health Sci 2009, 55, 95–102. [Google Scholar]
  36. Yoshida, S.; Hiyoshi, K.; Ichinose, T.; Takano, H.; Oshio, S.; Sugawara, I.; Takeda, K.; Shibamoto, T. Effect of nanoparticles on the male reproductive system of mice. Int. J. Androl 2009, 32, 337–342. [Google Scholar]
  37. Bai, Y.; Zhang, Y.; Zhang, J.; Mu, Q.; Zhang, W.; Butch, E.R.; Snyder, S.E.; Yan, B. Repeated administrations of carbon nanotubes in male mice cause reversible testis damage without affecting fertility. Nat. Nanotechnol 2010, 5, 683–689. [Google Scholar]
  38. Yoshida, S.; Hiyoshi, K.; Oshio, S.; Takano, H.; Takeda, K.; Ichinose, T. Effects of fetal exposure to carbon nanoparticles on reproductive function in male offspring. Fertil. Steril 2010, 93, 1695–1699. [Google Scholar]
  39. Li, W.Q.; Wang, F.; Liu, Z.M.; Wang, Y.C.; Wang, J.; Sun, F. Gold nanoparticles elevate plasma testosterone levels in male mice without affecting fertility. Small 2013, 27, 1708–1714. [Google Scholar]
  40. Morishita, Y.; Yoshioka, Y.; Satoh, H.; Nojiri, N.; Nagano, K.; Abe, Y.; Kamada, H.; Tsunoda, S.; Nabeshi, H.; Yoshikawa, T.; et al. Distribution and histologic effects of intravenously administered amorphous nanosilica particles in the testes of mice. Biochem. Biophys. Res. Commun 2012, 420, 297–301. [Google Scholar]
  41. Murugan, M.A.; Gangadharan, B.; Mathur, P.P. Antioxidative effect of fullerenol on goat epididymal spermatozoa. Asian J. Androl 2002, 4, 149–152. [Google Scholar]
  42. Braydich-Stolle, L.; Hussain, S.; Schlager, J.J.; Hofmann, M.C. In vitro cytotoxicity of nanoparticles in mammalian germline stem cells. Toxicol. Sci 2005, 88, 412–419. [Google Scholar]
  43. Makhluf, S.B.D.; Qasem, R.; Rubinstein, S.; Gedanken, A.; Breibart, H. Loading magnetic nanoparticles into sperm cells does not affect their functionality. Langmuir 2006, 22, 9480–9482. [Google Scholar]
  44. Makhluf, S.B.D.; Arnon Patra, C.R.; Mukhopadhyay, D.; Gedanken, A.; Mukherjee, P.; Breitbart, H. Labeling of sperm cells via the spontaneous penetration of Eu3+ ions as nanoparticles complexed with PVA or PVP. J. Phys. Chem. C 2008, 112, 12801–12807. [Google Scholar]
  45. Gopalan, R.; Osman, I.; Amani, A.; de Matas, M.; Anderson, D. The effect of zinc oxide and titanium dioxide nanoparticles in the comet assay with UVA photoactivation of human sperm and lymphocytes. Nanotoxicology 2009, 3, 33–39. [Google Scholar]
  46. Wiwanitkit, V.; Sereemaspun, A.; Rojanathanes, R. Effect of gold nanoparticles on spermatozoa: The first world report. Fertil. Steril 2009, 91, e7–e8. [Google Scholar]
  47. Braydich-Stolle, L.K.; Lucas, B.; Schrand, A.; Murdock, R.C.; Lee, T.; Schlager, J.; Hussain, S.; Hofmann, M.C. Silver nanoparticles disrupt GDNF/Fyn kinase signalling in spermatogonial stem cells. Toxicol. Sci 2010, 116, 577–589. [Google Scholar]
  48. Taylor, U.; Petersen, S.; Barchanski, A.; Mittag, A.; Barcikowski, S.; Rath, D. Influence of gold nanoparticles on vitality parameters of bovine spermatozoa. Reprod. Domest. Anim 2010, 45, 60. [Google Scholar]
  49. Zakhidov, S.T.; Marshak, T.L.; Malolina, E.A.; Kulibin, A.Y.; Zelenina, I.A.; Pavluchenkova, S.M.; Rudoy, V.M.; Dementeva, O.V.; Skuridin, S.G.; Evdokimov, Y.M. Gold nanoparticles disturb nuclear chromatin decondensation in mouse sperm in vitro. Biol. Membr 2010, 4, 349–353. [Google Scholar]
  50. Terzuoli, G.; Iacoponi, F.; Moretti, E.; Renieri, T.; Baldi, G.; Collodel, G. In vitro effect of silver engineered nanoparticles on human spermatozoa. J. Siena Acad. Sci 2011, 3, 27–29. [Google Scholar]
  51. Asare, N.; Instanes, C.; Sandberg, W.J.; Refsnes, M.; Schwarze, P.; Kruszewski, M.; Brunborg, G. Cytotoxic and genotoxic effects of silver nanoparticles in testicular cells. Toxicology 2012, 291, 65–72. [Google Scholar]
  52. Guo, L.L.; Liu, X.H.; Qin, D.X.; Gao, L.; Zhang, H.M.; Liu, J.Y.; Cui, Y.G. Effects of nanosized titanium dioxide on the reproductive system of male mice. Zhonghua Nan Ke Xue 2009, 15, 517–522. [Google Scholar]
  53. Bal, R.; Türk, G.; Tuzcu, M.; Yilmaz, O.; Ozercan, I.; Kuloglu, T.; Gür, S.; Nedzvetsky, V.S.; Tykhomyrov, A.A.; Andrievsky, G.V.; et al. Protective effects of nanostructures of hydrated C(60) fullerene on reproductive function in streptozotocin-diabetic male rats. Toxicology 2011, 282, 69–81. [Google Scholar]
  54. Noori, A.; Parivar, K.; Modaresi, M.; Messripour, M.; Yousefi, M.H.; Amiri, G.R. Effect of magnetic iron oxide nanoparticles on pregnancy and testicular development of mice. Afr. J. Biotechnol 2011, 10, 1221–1227. [Google Scholar]
  55. Sycheva, L.P.; Zhurkov, V.S.; Iurchenko, V.V.; Daugel-Dauge, N.O.; Kovalenko, M.A.; Krivtsova, E.K.; Durnev, A.D. Investigation of genotoxic and cytotoxic effects of micro- and nanosized titanium dioxide in six organs of mice in vivo. Mutat. Res 2011, 726, 8–14. [Google Scholar]
  56. Gromadzka-Ostrowska, J.; Dziendzikowska, K.; Lankoff, A.; Dobrzyńska, M.; Instanes, C.; Brunborg, G.; Gajowik, A.; Radzikowska, J.; Wojewódzka, M.; Kruszewski, M. Silver nanoparticles effects on epididymal sperm in rats. Toxicol. Lett 2012, 214, 251–258. [Google Scholar]
  57. Tang, S.; Tang, Y.; Zhong, L.; Murat, K.; Asan, G.; Yu, J.; Jian, R.; Wang, C.; Zhou, P. Short- and long-term toxicities of multi-walled carbon nanotubes in vivo and in vitro. J. Appl. Toxicol 2012, 32, 900–912. [Google Scholar]
  58. Kyjovska, Z.O.; Boisen, A.M.; Jackson, P.; Wallin, H.; Vogel, U.; Hougaard, K.S. Daily sperm production: Application in studies of prenatal exposure to nanoparticles in mice. Reprod. Toxicol 2013, 36, 88–97. [Google Scholar]
  59. Li, C.; Li, X.; Jigami, J.; Hasegawa, C.; Suzuki, A.K.; Zhang, Y.; Fujitani, Y.; Nagaoka, K.; Watanabe, G.; Taya, K. Effect of nanoparticle-rich diesel exhaust on testosterone biosynthesis in adult male mice. Inhal. Toxicol 2012, 24, 599–608. [Google Scholar]
  60. Ramdhan, D.H.; Ito, Y.; Yanagiba, Y.; Yamagishi, N.; Hayashi, Y.; Li, C.; Taneda, S.; Suzuki, A.K.; Watanabe, G.; Taya, K.; et al. Nanoparticle-rich diesel exhaust may disrupt testosterone biosynthesis and metabolism via growth hormone. Toxicol. Lett 2009, 191, 103–108. [Google Scholar]
  61. Yamagishi, N.; Ito, Y.; Ramdhan, D.H.; Yanagiba, Y.; Hayashi, Y.; Wang, D.; Li, C.M.; Taneda, S.; Suzuki, A.K.; Taya, K.; et al. Effect of nanoparticle-rich diesel exhaust on testicular and hippocampus steroidogenesis in male rats. Inhal. Toxicol 2012, 24, 459–467. [Google Scholar]
  62. Cho, W.S.; Duffin, R.; Poland, C.A.; Howie, S.E.; MacNee, W.; Bradley, M.; Megson, I.L.; Donaldson, K. Metal oxide nanoparticles induce unique inflammatory footprints in the lung: Important implications for nanoparticle testing. Environ. Health Perspect 2010, 118, 1699–1706. [Google Scholar]
  63. Rafeeqi, T.; Kaul, G. Carbon nanotubes as a scaffold for spermatogonial cell maintenance. J. Biomed. Nanotechnol 2010, 6, 710–717. [Google Scholar]
  64. Zakhidov, S.T.; Pavliuchenkova, S.M.; Marshak, T.L.; Rudoĭ, V.M.; Dementeva, O.V.; Zelenina, I.A.; Skuridin, S.G.; Makarov, A.A.; Khokhlov, A.N.; Evdokimov, I.M. Effect of gold nanoparticles on mouse spermatogenesis. Izv. Akad. Nauk. Ser. Biol 2012, 3, 279–287. [Google Scholar]
  65. Taylor, U.; Barchanski, A.; Garrels, W.; Klein, S.; Kues, W.; Barcikowski, S.; Rath, D. Toxicity of gold nanoparticles on somatic and reproductive cells. Adv. Exp. Med. Biol 2012, 733, 125–133. [Google Scholar]
  66. West, L.A.; Horvat, R.D.; Roess, D.A.; Barisas, B.G.; Juengel, J.L.; Niswender, G.D. Steroidogenic acute regulatory protein and peripheral-type benzodiazepine receptor associate at the mitochondrial membrane. Endocrinology 2001, 142, 502–555. [Google Scholar]
  67. Payne, A.H. Hormonal regulation of cytochrome P450 enzymes, cholesterol side-chain cleavage and 17 alpha-hydroxylase/C17–20 lyase in Leydig cells. Biol. Reprod 1990, 42, 399–404. [Google Scholar]
  68. Uchino, T.; Tokunaga, H.; Ando, M.; Utsumi, H. Quantitative determination of OH radical generation and its cytotoxicity induced by TiO(2)-UVA treatment. Toxicol. In Vitro 2002, 16, 629–635. [Google Scholar]
  69. Theogaraj, E.; Riley, S.; Hughes, L.; Maier, M.; Kirkland, D. An investigation of the photo-clastogenic potential of ultrafine titanium dioxide particles. Mutat. Res 2007, 634, 205–219. [Google Scholar]
  70. Warheit, D.B.; Hoke, R.A.; Finlay, C.; Donner, E.M.; Reed, K.L.; Sayes, C.M. Development of a base set of toxicity tests using ultrafine TiO2 particles as a component of nanoparticle risk management. Toxicol. Lett 2007, 171, 99–110. [Google Scholar]
  71. Zhu, R.R.; Wang, S.L.; Chao, J.; Shi, D.L.; Zhang, R.; Sun, X.Y.; Yao, S.D. Bio-effects of nano-TiO2 on DNA and cellular ultrastructure with different polymorph and size. Mater. Sci. Eng. C 2009, 29, 691–696. [Google Scholar]
  72. Di Virgilio, A.L.; Reigosa, M.; Arnal, P.M.; Fernández Lorenzo de Mele, M. Comparative study of the cytotoxic and genotoxic effects of titanium oxide and aluminium oxide nanoparticles in Chinese hamster ovary (CHO-K1) cells. J. Hazard. Mater 2010, 177, 711–718. [Google Scholar]
  73. Jiang, J.; Wang, J.; Zhang, X.; Huo, K.; Wong, H.M.; Yeung, K.W.; Zhang, W.; Hu, T.; Chu, P.K. Activation of mitogen-activated protein kinases cellular signal transduction pathway in mammalian cells induced by silicon carbide nanowires. Biomaterials 2010, 31, 7856–7862. [Google Scholar]
  74. Liu, X.; Qin, D.; Cui, Y.; Chen, L.; Li, H.; Chen, Z.; Gao, L.; Li, Y.; Liu, J. The effect of calcium phosphate nanoparticles on hormone production and apoptosis in human granulosa cells. Reprod. Biol. Endocrinol 2010, 8, 32–39. [Google Scholar]
  75. Abdullah, C.A.; Asanithi, P.; Brunner, E.W.; Jurewicz, I.; Bo, C.; Azad, C.L.; Ovalle-Robles, R.; Fang, S.; Lima, M.D.; Lepro, X.; et al. Aligned, isotropic and patterned carbon nanotube substrates that control the growth and alignment of Chinese hamster ovary cells. Nanotechnology 2011, 22, 205102, :1–205102:11.. [Google Scholar]
  76. Wang, S.; Hunter, L.A.; Arslan, Z.; Wilkerson, M.G.; Wickliffe, J.K. Chronic exposure to nanosized, anatase titanium dioxide is not cyto- or genotoxic to Chinese hamster ovary cells. Environ. Mol. Mutagen 2011, 52, 614–622. [Google Scholar]
  77. Wang, S.; Yu, H.; Wickliffe, J.K. Limitation of the MTT and XTT assays for measuring cell viability due to superoxide formation induced by nano-scale TiO2. Toxicol. In Vitro 2011, 25, 2147–2151. [Google Scholar]
  78. Ekkapongpisit, M.; Giovia, A.; Follo, C.; Caputo, G.; Isidoro, C. Biocompatibility, endocytosis, and intracellular trafficking of mesoporous silica and polystyrene nanoparticles in ovarian cancer cells: Effects of size and surface charge groups. Int. J. Nanomed 2012, 7, 4147–4158. [Google Scholar]
  79. Hirano, S.; Fujitani, Y.; Furuyama, A.; Kanno, S. Macrophage receptor with collagenous structure (MARCO) is a dynamic adhesive molecule that enhances uptake of carbon nanotubes by CHO-K1 cells. Toxicol. Appl. Pharmacol 2012, 259, 96–103. [Google Scholar]
  80. Hou, J.; Wan, X.Y.; Wang, F.; Xu, G.F.; Liu, Z. Effects of titanium dioxide nanoparticles on development and maturation of rat preantral follicle in vitro. Acad. J. Second Mil. Med. Univ 2009, 29, 869–873. [Google Scholar]
  81. Hsieh, M.S.; Shiao, N.H.; Chan, W.H. Cytotoxic effects of CdSe quantum dots on maturation of mouse oocytes, fertilization, and fetal development. Int. J. Mol. Sci 2009, 10, 2122–2135. [Google Scholar]
  82. Xu, G.; Lin, X.; Yong, K.T.; Roy, I.; Qu, J.; Wang, X. Visualization of Reproduction Toxicity of QDs for in vitro Oocytes Maturation. In Progress in biomedical optics and imaging; Proceedings of SPIE 7519 Eighth International Conference on Photonics and Imaging in Biology and Medicine, Wuhan, China, 8–10 August 2009, Luo, Q., Wang, L.V., Tuchin, V.V., Li, P., Fu, L., Eds.; SPIE: Bellingham,WA, USA, 2009; 7519 1W; pp. 7519–7583. [Google Scholar]
  83. Wang, X.M.; Yong, K.T.; Xu, G.X.; Lin, X.T.; Zhou, X.Q.; Qu, J.L.; Chen, S.; Hanben, C. Invasion of CdSe/CdS/ZnS Quantum Dots for oocytes in vitro maturation. Chin. Laser 2010, 37, 2730–2734. [Google Scholar]
  84. Xu, G.; Lin, S.; Law, W.C.; Roy, I.; Lin, X.; Mei, S.; Ma, H.; Chen, S.; Niu, H.; Wang, X. The invasion and reproductive toxicity of QDs-transferrin bioconjugates on preantral follicle in vitro. Theranostics 2012, 2, 734–745. [Google Scholar]
  85. Wang, J.; Zhu, X.; Zhang, X.; Zhao, Z.; Liu, H.; George, R.; Wilson-Rawls, J.; Chang, Y.; Chen, Y. Disruption of zebrafish (Danio rerio) reproduction upon chronic exposure to TiO2 nanoparticles. Chemosphere 2011, 83, 461–467. [Google Scholar]
  86. Gao, G.; Ze, Y.; Li, B.; Zhao, X.; Zhang, T.; Sheng, L.; Hu, R.; Gui, S.; Sang, X.; Sun, Q.; et al. Ovarian dysfunction and gene-expressed characteristics of female mice caused by long-term exposure to titanium dioxide nanoparticles. J. Hazard. Mater 2012, 243, 19–27. [Google Scholar]
  87. Griffitt, R.J.; Brown-Peterson, N.J.; Savin, D.A.; Manning, C.S.; Boube, I.; Ryan, R.A.; Brouwer, M. Effects of chronic nanoparticulate silver exposure to adult and juvenile sheepshead minnows (Cyprinodon variegatus). Environ. Toxicol. Chem 2012, 31, 160–167. [Google Scholar]
  88. Ramsden, C.S.; Henry, T.B.; Handy, R.D. Sub-lethal effects of titanium dioxide nanoparticles on the physiology and reproduction of zebrafish. Aquat. Toxicol 2013, 126, 404–413. [Google Scholar]
  89. Stelzer, R.; Hutz, R.J. Gold nanoparticles enter rat ovarian granulosa cells and subcellular organelles, and alter in-vitro estrogen accumulation. J. Reprod. Dev 2009, 55, 685–690. [Google Scholar]
  90. Li, C.; Li, X.; Suzuki, A.K.; Zhang, Y.; Fujitani, Y.; Nagaoka, K.; Watanabe, G.; Taya, K. Effects of exposure to nanoparticle-rich diesel exhaust on pregnancy in rats. J. Reprod. Dev 2013, 59, 145–150. [Google Scholar]
  91. Esmaeillou, M.; Moharamnejad, M.; Hsankhani, R.; Tehrani, A.A.; Maadi, H. Toxicity of ZnO nanoparticles in healthy adult mice. Environ. Toxicol. Pharmacol 2013, 35, 67–71. [Google Scholar]
  92. Pham, C.H.; Yi, J.; Gu, M.B. Biomarker gene response in male Medaka (Oryzias latipes) chronically exposed to silver nanoparticle. Ecotoxicol. Environ. Saf 2012, 78, 239–245. [Google Scholar]
  93. Gagné, F.; André, C.; Skirrow, R.; Gélinas, M.; Auclair, J.; van Aggelen, G.; Turcotte, P.; Gagnon, C. Toxicity of silver nanoparticles to rainbow trout: A toxicogenomic approach. Chemosphere 2012, 89, 615–622. [Google Scholar]
  94. Gagné, F.; Fortier, M.; Yu, L.; Osachoff, H.L.; Skirrow, R.C.; van Aggelen, G.; Gagnon, C.; Fournier, M. Immunocompetence and alterations in hepatic gene expression in rainbow trout exposed to CdS/CdTe quantum dots. J. Environ. Monit 2010, 12, 1556–1565. [Google Scholar]
  95. Sanders, M.B.; Sebire, M.; Sturve, J.; Christian, P.; Katsiadaki, I.; Lyons, B.P.; Sheahan, D.; Weeks, J.M.; Feist, S.W. Exposure of sticklebacks (Gasterosteus aculeatus) to cadmium sulfide nanoparticles: Biological effects and the importance of experimental design. Mar. Environ. Res 2008, 66, 161–163. [Google Scholar]
  96. Li, C.; Li, X.; Suzuki, A.K.; Fujitani, Y.; Jigami, J.; Nagaoka, K.; Watanabe, G.; Taya, K. Effects of exposure to nanoparticle-rich diesel exhaust on adrenocortical function in adult male mice. Toxicol. Lett 2012, 209, 277–281. [Google Scholar]
  97. Park, J.W.; Henry, T.B.; Menn, F.M.; Compton, R.N.; Sayler, G. No bioavailability of 17α-ethinylestradiol when associated with nC60 aggregates during dietary exposure in adult male zebrafish (Danio rerio). Chemosphere 2010, 81, 1227–1232. [Google Scholar]
  98. Park, J.W.; Henry, T.B.; Ard, S.; Menn, F.M.; Compton, R.N.; Sayler, G.S. The association between nC60 and 17α-ethinylestradiol (EE2) decreases EE2 bioavailability in zebrafish and alters nanoaggregate characteristics. Nanotoxicology 2011, 5, 406–416. [Google Scholar]
  99. Jain, M.P.; Vaisheva, F.; Maysinger, D. Metalloestrogenic effects of quantum dots. Nanomedicine 2012, 7, 23–37. [Google Scholar]
  100. Jackson, P.; Vogel, U.; Wallin, H.; Hougaard, K.S. Prenatal exposure to carbon black (printex 90): Effects on sexual development and neurofunction. Basic Clin. Pharmacol. Toxicol 2011, 109, 434–437. [Google Scholar]
  101. Lavado-Autric, R.; Auso, E.; Garcia-Velasco, J.V.; Arufe, M.C.; Escobar, D.R.; Berbel, P.; Morreale, D.E. Early maternal hypothyroxinemia alters histogenesis and cerebral cortex cytoarchitecture of the progeny. J. Clin. Invest 2003, 111, 1073–1082. [Google Scholar]
  102. Auso, E.; Lavado-Autric, R.; Cuevas, E.; del Rey, F.E.; Morreale, D.E.; Berbel, P. A moderate and transient deficiency of maternal thyroid function at the beginning of fetal neocorticogenesis alters neuronal migration. Endocrinology 2004, 145, 4037–4047. [Google Scholar]
  103. Berbel, P.; Mestre, J.L.; Santamaria, A.; Palazon, I.; Franco, A.; Graells, M.; Gonzalez-Torga, A.; de Escobar, G.M. Delayed neurobehavioral development in children born to pregnant women with mild hypothyroxinemia during the first month of gestation: The importance of early iodine supplementation. Thyroid 2009, 19, 511–519. [Google Scholar]
  104. Boas, M.; Feldt-Rasmussen, U.; Main, K.M. Thyroid effects of endocrine disrupting chemicals. Mol. Cell. Endocrinol 2012, 355, 240–248. [Google Scholar]
  105. Hinther, A.; Vawda, S.; Skirrow, R.C.; Veldhoen, N.; Collins, P.; Cullen, J.T.; van Aggelen, G.; Helbing, C.C. Nanometals induce stress and alter thyroid hormone action in amphibia at or below North American water quality guidelines. Environ. Sci. Technol 2010, 44, 8314–8321. [Google Scholar]
  106. Zha, L.; Zeng, J.; Sun, S.; Deng, H.; Luo, H.; Li, W. Chromium(III) nanoparticles affect hormone and immune responses in heat-stressed rats. Biol. Trace Elem. Res 2009, 129, 157–169. [Google Scholar]
  107. Hatch, E.E.; Nelson, J.W.; Stahlhut, R.W.; Webster, T.F. Association of endocrine disruptors and obesity: Perspectives from epidemiological studies. Int. J. Androl 2010, 33, 324–332. [Google Scholar]
  108. Newbold, R.R. Impact of environmental endocrine disrupting chemicals on the development of obesity. Hormones 2010, 9, 206–217. [Google Scholar]
  109. Ben-Jonathan, N.; Hugo, E.R.; Brandebourg, T.D. Effects of bisphenol A on adipokine release from human adipose tissue: Implications for the metabolic syndrome. Mol. Cell. Endocrinol 2009, 304, 49–54. [Google Scholar]
  110. Gurevitch, D.; Shuster-Meiseles, T.; Nov, O.; Zick, Y.; Rudich, A.; Rudich, Y. TiO2 nanoparticles induce insulin resistance in liver-derived cells both directly and via macrophage activation. Nanotoxicology 2012, 6, 804–812. [Google Scholar]
  111. Pourkhalili, N.; Hosseini, A.; Nili-Ahmadabadi, A.; Rahimifard, M.; Navaei-Nigjeh, M.; Hassani, S.; Baeeri, M.; Abdollahi, M. Improvement of isolated rat pancreatic islets function by combination of cerium oxide nanoparticles/sodium selenite through reduction of oxidative stress. Toxicol. Mech. Methods 2012, 22, 476–482. [Google Scholar]
  112. Wang, M.Q.; Xu, Z.R.; Zha, L.Y.; Lindemann, M.D. Effects of chromium nanocomposite supplementation on blood metabolites, endocrine parameters and immune traits in finishing pigs. Anim. Feed. Sci. Technol 2007, 139, 69–80. [Google Scholar]
  113. Zha, L.Y.; Xu, Z.R.; Wang, M.Q.; Gu, L.Y. Effects of chromium nanoparticle dosage on growth, body composition, serum hormones and tissue chromium in Sprague-Dawley rats. J. Zhejiang Univ. Sci. B 2007, 8, 323–330. [Google Scholar]
  114. Crouzier, D.; Follot, S.; Gentilhomme, E.; Flahaut, E.; Arnaud, R.; Dabouis, V.; Castellarin, C.; Debouzy, J.C. Carbon nanotubes induce inflammation but decrease the production of reactive oxygen species in lung. Toxicology 2010, 272, 39–45. [Google Scholar] [Green Version]
  115. Pourkhalili, N.; Hosseini, A.; Nili-Ahmadabadi, A.; Hassani, S.; Pakzad, M.; Baeeri, M.; Mohammadirad, A.; Abdollahi, M. Biochemical and cellular evidence of the benefit of a combination of cerium oxide nanoparticles and selenium to diabetic rats. World J. Diabetes 2011, 2, 204–210. [Google Scholar]
  116. Corona-Morales, A.A.; Castell, A.; Escobar, A.; Drucker-Colín, R.; Zhang, L. Fullerene C60 and ascorbic acid protect cultured chromaffin cells against levodopa toxicity. J. Neurosci. Res 2003, 71, 121–126. [Google Scholar]
  117. Hussain, S.M.; Javorina, A.K.; Schrand, A.M.; Duhart, H.M.; Ali, S.F.; Schlager, J.J. The interaction of manganese nanoparticles with PC-12 cells induces dopamine depletion. Toxicol. Sci 2006, 92, 456–463. [Google Scholar]
  118. Wang, J.; Rahman, M.F.; Duhart, H.M.; Newport, G.D.; Patterson, T.A.; Murdock, R.C.; Hussain, S.M.; Schlager, J.J.; Ali, S.F. Expression changes of dopaminergic system-related genes in PC12 cells induced by manganese, silver, or copper nanoparticles. Neurotoxicology 2009, 30, 926–933. [Google Scholar]
  119. Love, S.A.; Haynes, C.L. Assessment of functional changes in nanoparticle-exposed neuroendocrine cells with amperometry: Exploring the generalizability of nanoparticle-vesicle matrix interactions. Anal. Bioanal. Chem 2010, 398, 677–688. [Google Scholar]
  120. Gosso, S.; Gavello, D.; Giachello, C.N.; Franchino, C.; Carbone, E.; Carabelli, V. The effect of CdSe-ZnS quantum dots on calcium currents and catecholamine secretion in mouse chromaffin cells. Biomaterials 2011, 32, 9040–9050. [Google Scholar]
  121. Powers, C.M.; Badireddy, A.R.; Ryde, I.T.; Seidler, F.J.; Slotkin, T.A. Silver nanoparticles compromise neurodevelopment in PC12 cells: Critical contributions of silver ion, particle size, coating, and composition. Environ. Health Perspect 2011, 119, 37–44. [Google Scholar]
  122. Wu, J.; Wang, C.; Sun, J.; Xue, Y. Neurotoxicity of silica nanoparticles: Brain localization and dopaminergic neurons damage pathways. ACS Nano 2011, 5, 4476–4489. [Google Scholar]
  123. Love, S.A.; Liu, Z.; Haynes, C.L. Examining changes in cellular communication in neuroendocrine cells after noble metal nanoparticle exposure. Analyst 2012, 137, 3004–3010. [Google Scholar]
  124. Wang, M.Q.; Xu, Z.R.; Li, W.F.; Jiang, Z.G. Effect of chromium nanocomposite supplementation on growth hormone pulsatile secretion and mRNA expression in finishing pigs. J. Anim. Physiol. Anim. Nutr 2009, 93, 520–525. [Google Scholar]
  125. Vincent, J.B. The biochemistry of chromium. J. Nutr 2001, 130, 715–718. [Google Scholar]
  126. Douglas, S.A.; Sreenivasan, D.; Carman, F.H.; Bunn, S.J. Cytokine interactions with adrenal medullary chromaffin cells. Cell. Mol. Neurobiol 2010, 30, 1467–1475. [Google Scholar]
  127. Guérineau, N.C.; Desarménien, M.G.; Carabelli, V.; Carbone, E. Functional chromaffin cell plasticity in response to stress: Focus on nicotinic, gap junction, and voltage-gated Ca(2+) channels. J. Mol. Neurosci 2012, 48, 368–386. [Google Scholar]
  128. Carabelli, V.; Marcantoni, A.; Comunanza, V.; de Luca, A.; Díaz, J.; Borges, R.; Carbone, E. Chronic hypoxia up-regulates alpha1H T-type channels and low-threshold catecholamine secretion in rat chromaffin cells. J. Physiol 2007, 584, 149–165. [Google Scholar]
  129. Pérez-Alvarez, A.; Hernández-Vivanco, A.; Albillos, A. Past, present and future of human chromaffin cells: Role in physiology and therapeutics. Cell. Mol. Neurobiol 2010, 30, 1407–1415. [Google Scholar]
  130. Gavello, D.; Vandael, D.H.; Cesa, R.; Premoselli, F.; Marcantoni, A.; Cesano, F.; Scarano, D.; Fubini, B.; Carbone, E.; Fenoglio, I.; Carabelli, V. Altered excitability of cultured chromaffin cells following exposure to multi-walled carbon nanotubes. Nanotoxicology 2012, 6, 47–60. [Google Scholar]
  131. Soin, T.; Verslycke, T.; Janssen, C.; Smagghe, G. Ecdysteroids and Their Importance in Endocrine Disruption Research. In Ecdysone: Structures and Functions; Smagghe, G., Ed.; Springer Science + Business Media B.V.: Dordrecht, The Netherlands, 2009; Volume Chapter 22, pp. 539–549. [Google Scholar]
  132. Wiench, K.; Wohlleben, W.; Hisgen, V.; Radke, K.; Salinas, E.; Zok, S.; Landsiedel, R. Acute and chronic effects of nano- and non-nano-scale TiO(2) and ZnO particles on mobility and reproduction of the freshwater invertebrate Daphnia magna. Chemosphere 2009, 76, 1356–1365. [Google Scholar]
  133. Zhu, X.; Chang, Y.; Chen, Y. Toxicity and bioaccumulation of TiO2 nanoparticle aggregates in Daphnia magna. Chemosphere 2010, 78, 209–215. [Google Scholar]
  134. Seitz, F.; Bundschuh, M.; Rosenfeldt, R.R.; Schulz, R. Nanoparticle toxicity in Daphnia magna reproduction studies: The importance of test design. Aquat. Toxicol 2013, 126, 163–168. [Google Scholar]
  135. Zhao, C.M.; Wang, W.X. Comparison of acute and chronic toxicity of silver nanoparticles and silver nitrate to Daphnia magna. Environ. Toxicol. Chem 2011, 30, 885–892. [Google Scholar]
  136. Oberdörster, E.; Zhu, S.; Blickley, T.M.; McClellan-Green, P.; Haasch, M.L. Ecotoxicology of carbon-based engineered nanoparticles: Effects of fullerene (C60) on aquatic organisms. Carbon 2006, 44, 1112–1120. [Google Scholar]
  137. Tao, X.; Fortner, J.D.; Zhang, B.; He, Y.; Chen, Y.; Hughes, J.B. Effects of aqueous stable fullerene nanocrystals (nC60) on Daphnia magna: Evaluation of sub-lethal reproductive responses and accumulation. Chemosphere 2009, 77, 1482–1487. [Google Scholar]
  138. Mendonça, E.; Diniz, M.; Silva, L.; Peres, I.; Castro, L.; Correia, J.B.; Picado, A. Effects of diamond nanoparticle exposure on the internal structure and reproduction of Daphnia magna. J. Hazard. Mater 2011, 186, 265–271. [Google Scholar]
  139. Kim, J.; Park, Y.; Yoon, T.H.; Yoon, C.S.; Choi, K. Phototoxicity of CdSe/ZnSe quantum dots with surface coatings of 3-mercaptopropionic acid or tri-n-octylphosphine oxide/gum arabic in Daphnia magna under environmentally relevant UV-B light. Aquat. Toxicol 2010, 97, 116–124. [Google Scholar]
  140. Fabrega, J.; Tantra, R.; Amer, A.; Stolpe, B.; Tomkins, J.; Fry, T.; Lead, J.R.; Tyler, C.R.; Galloway, T.S. Sequestration of zinc from zinc oxide nanoparticles and life cycle effects in the sediment dweller amphipod Corophium volutator. Environ. Sci. Technol 2012, 46, 1128–1135. [Google Scholar]
  141. Nair, P.M.; Park, S.Y.; Lee, S.W.; Choi, J. Differential expression of ribosomal protein gene, gonadotrophin releasing hormone gene and Balbiani ring protein gene in silver nanoparticles exposed Chironomus riparius. Aquat. Toxicol 2011, 101, 31–37. [Google Scholar]
  142. Nair, P.M.; Choi, J. Modulation in the mRNA expression of ecdysone receptor gene in aquatic midge, Chironomus riparius upon exposure to nonylphenol and silver nanoparticles. Environ. Toxicol. Pharmacol 2012, 33, 98–106. [Google Scholar]
  143. Vecchio, G.; Galeone, A.; Brunetti, V.; Maiorano, G.; Sabella, S.; Cingolani, R.; Pompa, P.P. Concentration-dependent, size-independent toxicity of citrate capped AuNPs in Drosophila melanogaster. PLoS One 2012, 7, e29980. [Google Scholar]
  144. Wang, H.; Wick, R.L.; Xing, B. Toxicity of nanoparticulate and bulk ZnO, Al2O3 and TiO2 to the nematode Caernorhabditis elegans. Environ. Poll 2009, 157, 1171–1177. [Google Scholar]
  145. Menard, A.; Drobne, D.; Jemec, A. Ecotoxicity of nanosized TiO2. Review of in vivo data. Environ. Pollut 2011, 159, 677–684. [Google Scholar]
  146. Roh, J.Y.; Park, Y.K.; Park, K.; Choi, J. Ecotoxicological investigation of CeO(2) and TiO(2) nanoparticles on the soil nematode Caenorhabditis elegans using gene expression, growth, fertility, and survival as endpoints. Environ. Toxicol. Pharmacol 2010, 29, 167–172. [Google Scholar]
  147. Roh, J.Y.; Sim, S.J.; Yi, J.; Park, K.; Chung, K.H.; Ryu, D.Y.; Choi, J. Ecotoxicity of silver nanoparticles on the soil nematode Caenorhabditis elegans using functional ecotoxicogenomics. Environ. Sci. Technol 2009, 43, 3933–3940. [Google Scholar]
  148. Ma, H.; Bertsch, P.M.; Glenn, T.C.; Kabengi, N.J.; Williams, P.L. Toxicity of manufactured zinc oxide nanoparticles in the nematode Caenorhabditis elegans. Environ. Toxicol. Chem 2009, 28, 1324–1330. [Google Scholar]
  149. Lim, D.; Roh, J.Y.; Eom, H.J.; Choi, J.Y.; Hyun, J.; Choi, J. Oxidative stress-related PMK-1 P38 MAPK activation as a mechanism for toxicity of silver nanoparticles to reproduction in the nematode Caenorhabditis elegans. Environ. Toxicol. Chem 2012, 31, 585–592. [Google Scholar]
  150. Qu, Y.; Li, W.; Zhou, Y.; Liu, X.; Zhang, L.; Wang, L.; Li, Y.F.; Iida, A.; Tang, Z.; Zhao, Y.; Chai, Z.; Chen, C. Full assessment of fate and physiological behavior of quantum dots utilizing Caenorhabditis elegans as a model organism. Nano Lett 2011, 11, 3174–3183. [Google Scholar]
  151. Cha, Y.J.; Lee, J.; Choi, S.S. Apoptosis-mediated in vivo toxicity of hydroxylated fullerene nanoparticles in soil nematode Caenorhabditis elegans. Chemosphere 2012, 87, 49–54. [Google Scholar]
  152. Pluskota, A.; Horzowski, E.; Bossinger, O.; von Mikecz, A. In Caenorhabditis elegans nanoparticle-bio-interactions become transparent: Silica-nanoparticles induce reproductive senescence. PLoS One 2009, 4, e6622. [Google Scholar]
  153. Unrine, J.M.; Hunyadi, S.E.; Tsyusko, O.V.; Rao, W.; Shoults-Wilson, W.A.; Bertsch, P.M. Evidence for bioavailability of Au nanoparticles from soil and biodistribution within earthworms (Eisenia fetida). Environ. Sci. Technol 2010, 44, 8308–8313. [Google Scholar]
  154. Schoults-Wilson, W.A.; Reinsch, B.C.; Tsyusko, O.V.; Bertsch, P.M.; Lowry, G.V.; Unrine, J.M. Role of particle size and soil type in toxicity of silver nanoparticles to earthworms. Soil Sci. Soc. Am. J 2011, 75, 365–377. [Google Scholar]
  155. Coleman, J.G.; Johnson, D.R.; Stanley, J.K.; Bednar, A.J.; Weiss, C.A., Jr; Boyd, R.E.; Steevens, J.A. Assessing the fate and effects of nano aluminum oxide in the terrestrial earthworm Eisenia fetida. Environ. Toxicol. Chem. 2010, 29, 1575–1580. [Google Scholar]
  156. Cañas, J.E.; Qi, B.; Li, S.; Maul, J.D.; Cox, S.B.; Das, S.; Green, M.J. Acute and reproductive toxicity of nano-sized metal oxides (ZnO and TiO2) to earthworms (Eisenia fetida). J. Environ. Monit 2011, 13, 3351–3357. [Google Scholar]
  157. Hooper, H.L.; Jurkschat, K.; Morgan, A.J.; Bailey, J.; Lawlor, A.J.; Spurgeon, D.J.; Svendsen, C. Comparative chronic toxicity of nanoparticulate and ionic zinc to the earthworm Eisenia veneta in a soil matrix. Environ. Int 2011, 37, 1111–1117. [Google Scholar]
  158. Unrine, J.M.; Tsyusko, O.V.; Hunyadi, S.E.; Judy, J.D.; Bertsch, P.M. Effects of particle size on chemical speciation and bioavailability of copper to earthworms (Eisenia fetida) exposed to copper nanoparticles. J. Environ. Qual 2010, 39, 1942–1953. [Google Scholar]
  159. Scott-Fordsmand, J.J.; Krogh, P.H.; Schaefer, M.; Johansen, A. The toxicity testing of double-walled nanotubes-contaminated food to Eisenia veneta earthworms. Ecotoxicol. Environ. Saf 2008, 71, 616–619. [Google Scholar]
  160. Li, D.; Alvarez, P.J. Avoidance, weight loss, and cocoon production assessment for Eisenia fetida exposed to C60 in soil. Environ. Toxicol. Chem 2011, 30, 2542–2545. [Google Scholar]
  161. Van der Ploeg, M.J.; Baveco, J.M.; van der Hout, A.; Bakker, R.; Rietjens, I.M.; van den Brink, N.W. Effects of C60 nanoparticle exposure on earthworms (Lumbricus rubellus) and implications for population dynamics. Environ. Pollut 2011, 159, 198–203. [Google Scholar]
  162. Pakarinen, K.; Petersen, E.J.; Leppänen, M.T.; Akkanen, J.; Kukkonen, J.V. Adverse effects of fullerenes (nC60) spiked to sediments on Lumbriculus variegatus (Oligochaeta). Environ. Pollut 2011, 159, 3750–3756. [Google Scholar]
  163. Lynch, I.; Dawson, K.A.; Linse, S. Detecting cryptic epitopes created by nanoparticles. Sci STKE 2006, 327. [Google Scholar] [CrossRef]
  164. Monopoli, M.P.; Aberg, C.; Salvati, A.; Dawson, K.A. Biomolecular coronas provide the biological identity of nanosized materials. Nat. Nanotechnol 2012, 7, 779–786. [Google Scholar]
  165. Walkey, C.D.; Chan, W.C. Understanding and controlling the interaction of nanomaterials with proteins in a physiological environment. Chem. Soc. Rev 2012, 41, 2780–2799. [Google Scholar]
  166. Monopoli, M.P.; Walczyk, D.; Campbell, A.; Elia, G.; Lynch, I.; Bombelli, F.B.; Dawson, K.A. Physical-chemical aspects of protein corona: Relevance to in vitro and in vivo biological impacts of nanoparticles. J. Am. Chem. Soc 2011, 133, 2525–2534. [Google Scholar]
  167. De Roe, C.; Courtoy, P.J.; Baudhuin, P. A model of protein-colloidal gold interactions. J. Histochem. Cytochem 1987, 35, 1191–1198. [Google Scholar]
  168. Casals, E.; Gonzalez, E.; Puntes, V.F. Reactivity of inorganic nanoparticles in biological environments: Insights into nanotoxicity mechanisms. J. Phys. D 2012, 45, 443001, :1–443001:15.. [Google Scholar]
  169. Auffan, M.; Rose, J.; Wiesner, M.R.; Bottero, J.Y. Chemical stability of metallic nanoparticles: A parameter controlling their potential cellular toxicity in vitro. Environ. Pollut 2009, 157, 1127–1133. [Google Scholar]
  170. Franklin, N.M.; Rogers, N.J.; Apte, S.C.; Batley, G.E.; Gadd, G.E.; Casey, P.S. Comparative toxicity of nanoparticulate ZnO, bulk ZnO, and ZnCl2 to a freshwater microalga (Pseudokirchneriella subcapitata): The importance of particle solubility. Environ. Sci. Technol 2007, 41, 8484–8490. [Google Scholar]
  171. Zheng, X.; Wu, R.; Chen, Y. Effects of ZnO nanoparticles on wastewater biological nitrogen and phosphorus removal. Environ. Sci. Technol 2011, 45, 2826–2832. [Google Scholar]
  172. Dibrov, P.; Dzioba, J.; Gosink, K.K.; Häse, C.C. Chemiosmotic mechanism of antimicrobial activity of Ag(+) in vibrio cholerae. Antimicrob. Agents Chemother 2002, 46, 2668–2670. [Google Scholar]
  173. Morones, J.R.; Elechiguerra, J.L.; Camacho, A.; Holt, K.; Kouri, J.B.; Ramírez, J.T.; Yacaman, M.J. The bactericidal effect of silver nanoparticles. Nanotechnology 2005, 16, 2346–2353. [Google Scholar]
  174. Nowack, B.; Krug, H.F.; Height, M. 120 years of nanosilver history: Implications for policy makers. Environ. Sci. Technol 2011, 45, 1177–1183. [Google Scholar]
  175. Derfus, A.M.; Chan, W.C.W.; Bhatia, S.N. Probing the cytotoxicity of semiconductor quantum dots. Nano Lett 2004, 4, 11–18. [Google Scholar]
  176. Chen, N.; He, Y.; Su, Y.; Li, X.; Huang, Q.; Wang, H.; Zhang, X.; Tai, R.; Fan, C. The cytotoxicity of cadmium-based quantum dots. Biomaterials 2012, 33, 1238–1244. [Google Scholar]
  177. Oberdörster, G. Safety assessment for nanotechnology and nanomedicine: Concepts of nanotoxicology. J. Intern. Med 2010, 267, 89–105. [Google Scholar]
  178. Iavicoli, I.; Leso, V.; Fontana, L.; Cottica, D.; Bergamaschi, A. Characterization of inhalable, thoracic, and respirable fractions and ultrafine particle exposure during grinding, brazing, and welding activities in a mechanical engineering factory. J. Occup. Environ. Med 2013, 55, 430–445. [Google Scholar]
  179. Nascarella, M.A.; Calabrese, E.J. A method to evaluate hormesis in nanoparticle dose-responses. Dose Response 2012, 10, 344–354. [Google Scholar]
Figure 1. Alterations and impairment of testicular structure induced by exposure to different types of NPs. Metal-based NPs, carbon-based NPs and nanoparticle-rich diesel exhaust (NRDE-NPs) were reported to induce changes in testicular histology of laboratory animals. Metal-based NPs (TiO2-NPs) induced the disruption of seminiferous tubules and decreased the number of Sertoli cells; Carbon-based NPs (carbon black-NPs, multi-walled carbon nanotubes) caused vacuolization of seminiferous tubules, seminiferous tubule damage and low cellular adhesion of seminiferous epithelia, partial disappearance or vacuolization of Sertoli cells, reduced thickness of the germinative layer, lower spermatogonia number, vasodilatation and hyperemia; NRDE-NPs induced degenerative and necrotic changes, interstitial edema, desquamation of the seminiferous epithelium, loss of germ cells and spermatozoa.
Figure 1. Alterations and impairment of testicular structure induced by exposure to different types of NPs. Metal-based NPs, carbon-based NPs and nanoparticle-rich diesel exhaust (NRDE-NPs) were reported to induce changes in testicular histology of laboratory animals. Metal-based NPs (TiO2-NPs) induced the disruption of seminiferous tubules and decreased the number of Sertoli cells; Carbon-based NPs (carbon black-NPs, multi-walled carbon nanotubes) caused vacuolization of seminiferous tubules, seminiferous tubule damage and low cellular adhesion of seminiferous epithelia, partial disappearance or vacuolization of Sertoli cells, reduced thickness of the germinative layer, lower spermatogonia number, vasodilatation and hyperemia; NRDE-NPs induced degenerative and necrotic changes, interstitial edema, desquamation of the seminiferous epithelium, loss of germ cells and spermatozoa.
Ijms 14 16732f1
Figure 2. Effects on pituitary-ovary and pituitary-testis axes caused by exposure to different types of NPs.Effects on pituitary-ovary axis: Au-NPs induced a greater output of estradiol from ovarian granulosa cells after 1–5 h of treatment, while they decreased the estradiol level after 24 h. TiO2-NP induced changes in hormone levels and ovarian injury in mice may be related to alterations in ovarian gene expression, e.g., Akr1c18, StAr, Cyp17a1, and Lgmn genes were up-regulated. Moreover, TiO2-NPs increased serum levels of estradiol while they decreased those of progesterone, FSH, LH and testosterone in female mice; NRDE-NP exposure led to an increase in LH and estradiol and a reduction in progesterone levels in rats. mRNA expression of the cytochrome P450scc, the 3β-hydroxysteroid dehydrogenase, and the LH receptor were significantly reduced, while that of estradiol-17β was increased by NR-DE exposure, suggesting an impairment in luteal function. NRDE-NP alteration in progesterone secretion was due to the suppression of the steroid enzyme activity and the suppression of the production of LH receptors in corpora lutea; Effects on pituitary-testis axis: Au-NPs induced elevated testosterone levels that directly affected the testicular hormone production, since no alterations in LH and FSH plasma values were observed; NRDE-NPs increased the biosynthesis of testicular testosterone. This could be attributed to an increase in the mRNA expression of StAR and cytochrome P450 side-chain cleavage in Leydig cells. Increased levels of serum and testicular testosterone were related to an enhanced expression of genes involved in testicular cholesterol synthesis, such as HMG-CoA, LDL-R, SR-B1, PBR, P450scc, P450 17α, and 17β-HSD induced by NRDE-NPs. Increased testosterone production was due to the impaired balance between androgen-metabolizing- and testosterone biosynthetic-enzymes and not to an LH stimulation, as demonstrated by the absence of alterations in the LH and FSH levels; CB-NPs can affect testosterone production through the induction of StAR gene expression.
Figure 2. Effects on pituitary-ovary and pituitary-testis axes caused by exposure to different types of NPs.Effects on pituitary-ovary axis: Au-NPs induced a greater output of estradiol from ovarian granulosa cells after 1–5 h of treatment, while they decreased the estradiol level after 24 h. TiO2-NP induced changes in hormone levels and ovarian injury in mice may be related to alterations in ovarian gene expression, e.g., Akr1c18, StAr, Cyp17a1, and Lgmn genes were up-regulated. Moreover, TiO2-NPs increased serum levels of estradiol while they decreased those of progesterone, FSH, LH and testosterone in female mice; NRDE-NP exposure led to an increase in LH and estradiol and a reduction in progesterone levels in rats. mRNA expression of the cytochrome P450scc, the 3β-hydroxysteroid dehydrogenase, and the LH receptor were significantly reduced, while that of estradiol-17β was increased by NR-DE exposure, suggesting an impairment in luteal function. NRDE-NP alteration in progesterone secretion was due to the suppression of the steroid enzyme activity and the suppression of the production of LH receptors in corpora lutea; Effects on pituitary-testis axis: Au-NPs induced elevated testosterone levels that directly affected the testicular hormone production, since no alterations in LH and FSH plasma values were observed; NRDE-NPs increased the biosynthesis of testicular testosterone. This could be attributed to an increase in the mRNA expression of StAR and cytochrome P450 side-chain cleavage in Leydig cells. Increased levels of serum and testicular testosterone were related to an enhanced expression of genes involved in testicular cholesterol synthesis, such as HMG-CoA, LDL-R, SR-B1, PBR, P450scc, P450 17α, and 17β-HSD induced by NRDE-NPs. Increased testosterone production was due to the impaired balance between androgen-metabolizing- and testosterone biosynthetic-enzymes and not to an LH stimulation, as demonstrated by the absence of alterations in the LH and FSH levels; CB-NPs can affect testosterone production through the induction of StAR gene expression.
Ijms 14 16732f2
Table 1. Categories, criteria of classifications and numbers of chemical substances included in the Endocrine Disrupter Priority list (EDPL). The constitution and the integration of the EDPL is carried out by a process of analysis which consists of three steps. First of all, a working list of suspected endocrine disruptors chemicals is compiled, based on the information provided by various organizations and on the analysis of the literature (step 1). Subsequently the available information is reviewed to identify those chemicals that are more relevant, in term of exposure, for both humans and animals (step 2). Finally, the identified chemicals are studied to determine the strength of evidence for endocrine disruption and to enroll them in a specific category (step 3). Addition and removal of chemicals may be required in response to either developments in scientific knowledge or changes in chemical usage patterns.
Table 1. Categories, criteria of classifications and numbers of chemical substances included in the Endocrine Disrupter Priority list (EDPL). The constitution and the integration of the EDPL is carried out by a process of analysis which consists of three steps. First of all, a working list of suspected endocrine disruptors chemicals is compiled, based on the information provided by various organizations and on the analysis of the literature (step 1). Subsequently the available information is reviewed to identify those chemicals that are more relevant, in term of exposure, for both humans and animals (step 2). Finally, the identified chemicals are studied to determine the strength of evidence for endocrine disruption and to enroll them in a specific category (step 3). Addition and removal of chemicals may be required in response to either developments in scientific knowledge or changes in chemical usage patterns.
Type of categoryCategory 1Category 2Category 3aCategory 3bReferences
Criteria of classificationAt least one in vivo study providing clear evidence of endocrine disruption in an intact organismPotential for endocrine disruption. In vitro data indicating potential for endocrine disruption in intact organisms. Also includes effects in vivo that may, or may not be endocrine disruption-mediated.No scientific basis for inclusion in list. Endocrine disruption studies available but no indications of endocrine disruption effects.Substances with no or insufficient data gathered
Number of chemical substances in each category1941252386[1518]
Table 2. Effects of NPs on the male reproductive system.
Table 2. Effects of NPs on the male reproductive system.
Type of studyType and physico-chemical properties of NPsExperimental protocolCell line/laboratory animalsResultsReferences
Alterations and impairment of testicular structure
In vitroTiO2-NPs (25–70 nm); CB-NPs (14 nm); DE-NPsTreatment with 0–1000 μg/mL for 24 and 48 hMouse testis Leydig cell line TM3
  • Internalization of NPs;
  • Dose-dependent reduction of cell viability after 48-hour TiO2-NP exposure.
[30]
In vitro75% rutile and 25% anatase TiO2-NPs (24.5 nm)Treatment with 0.5–50 μg/mL for 4, 24 and 48 hRTG-2 cells
  • Significant cytotoxicity only at the highest concentration.
[31]
In vitroCeO2-NPs (10 nm and 20–25 nm)Treatment with 0–100 μg/mL for 24 and 72 hRTG-2 cells, derived from rainbow trout (Oncorhynchus mykiss) gonadal tissue
  • No cytotoxic effects.
[32]
In vivoNRDE-NPs—Average of mode diameter and number concentrations for low, middle and high doses were 22.48, 26.13 and 27.06 nm; 2.27 × 105, 5.11 × 105 and 1.36 × 106, respectivelyInhalation exposure to low (15.37), middle (36.35) and high (168.84) μg/m3 doses for 4, 8, or 12 weeks (5 h/day, 5 days/week)Fischer F344 rats
  • Seminiferous tubules showed degenerative and necrotic changes, interstitial edema, desquamation of the seminiferous epithelium, and loss of spermatozoa.
[33]
In vivoNRDE-NPs—Average of mode diameter and number concentrations were 26.81 nm and 1.83 × 106, respectivelyInhalation exposure to 148.86 μg/m3 for 5 h daily for 19 gestational daysPregnant Fischer F344 rats
  • Loss of germ cells in the seminiferous tubules of male offspring.
[34]
In vivoTiO2-NPs (anatase form, particle size of 25–70 nm, surface area 20–25 m2/g)Subcutaneous injections of 100 μL of 1mg/mL solution on 4 gestational daysPregnant Slc:ICR mice
  • Disruption of seminiferous tubules;
  • Decreased numbers of Sertoli cells.
[35]
In vivoCB-NPs (14, 56 and 95 nm with a surface area of 300, 45 and 20 m2/g, respectively)Intratracheal administration of 0.1 mg/kg body weight for 10 times every weekICR mice
  • Partial vacuolization of the seminiferous tubules;
  • The adverse effects depend on particle mass rather than particle number.
[36]
In vivoCarboxylate-functionalized (diameter of 20–30 nm, length 0.5–2.0 μm) and Amine-functionalized (diameter of 20–30 nm, length 0.5–2.0 μm) MWCNTsMice were randomly divided into 12 groups (15-day single dose, 15-day multi-dose, 60-day multi-dose, and 90-day multi-dose groups). Mice were given injections (5 mg/kg) via the tail vein once (single dose) or every 3 days for 5 timesBALB/c mice
  • Reduction in the thickness of the germinative layer; Reduced spermatogonia number; Partial disappearance or vacuolization of Sertoli cells; Induction of oxidative stress.
[37]
In vivoCB-NPs (14 nm)Intratracheal administration of 0.2 mg/kg body weight on days 7 and 14 of gestationPregnant ICR mice
  • Partial vacuolization of seminiferous tubules; Reduction of cellular adhesion of seminiferous epithelia.
[38]
In vivomPEG@Au-NP and PEG-NH2@Au-NP (14 nm)Intravenous injection of 45 and 225 mg/kg of mPEG@Au-NP and 45 mg/kg of PEG-NH2@ Au-NP at 48 h intervals for 5 daysICR mice
  • Penetration and accumulation of NPs in the testes; No testicular morphological changes; No germ cell apoptosis.
[39]
In vivoAmorphous silica particles (nSP70, 70-nm diameter; nSP300, 300-nm diameter)Intravenous injection of 0.4 and 0.8 mg of nSP70BALB/c mice
  • nSP70 can penetrate the bloodtestis barrier and the nuclear membranes of spermatocytes without producing apparent testicular injury.
[40]
Alterations and impairment of spermatogenesis
In vitroFullerenolTreatment with fullerenol (1, 10 and 100 μmol) for 3 h.Epididymal sperm samples collected from the fresh epididymis of adult goats
  • Protective effect, by increasing the activities of antioxidant enzymes and decreasing lipid peroxidation, on the potential oxidative stress damage on spermatozoa.
[41]
In vitroSilver (15 nm), molybdenum (30 nm), and aluminum (30 nm)Treatment with 5, 10, 25, 50, and 100 μg/mL culture medium for 48 hC18-4 spermatogonial stem cell line
  • concentration-dependent toxicity for all types of particles tested; Silver nanoparticles were the most toxic while molybdenum trioxide (MoO3) nanoparticles were the least toxic.
[42]
In vitroMagnetic iron oxide NPs (Fe3O4-NPs) coated with poly(vinyl alcohol)Treatment with 7.35 mM for 80 min and 4 hBovine sperm cells
  • No effects on the motility and the ability to undergo the acrosome reaction.
[43]
In vitroEu2O3-NPs (30 ± 10 nm); EuOH3-NPs conjugated with polyvinyl alcohol or polyvinyl piyrolidone (15.4 ± 3 nm and 9.3 ± 3 nm, respectively)1 mL of washed sperm cells was incubated for 24 h at 39 °C with 2.5 mg/mL of Eu2O3-NPs or of EuOH3-NPsBovine sperm cells
  • Loss of bovine spermatozoa motility after exposure to Eu2O3-NPs; No effects on motility and the ability to undergo acrosome reaction after exposure to EuOH3-NPs.
[44]
In vitroTiO2-NPs and ZnO-NPs (40–70 nm)Treatment with 3.73–59.7 μg/mL of TiO2-NPs and 11.5–93.2 μg/mL of ZnO-NPsHuman spermatozoa
  • Concentration - dependent induction of sperm DNA damage.
[45]
In vitroAu-NPs (9 nm)Analyses were conducted on a mixture of 500 μL of Au-NPs (44 ppm) solution and semenHuman spermatozoa
  • 25% of sperm were not motile; Penetration of Au-NPs into the sperm head and tails; Fragmentation of sperm.
[46]
In vitroHydrocarbon-coated silver (Ag-HC) nanoparticles of 15, 25, and 80 nm diameters and Polysaccharide-coated silver (Ag-PS) nanoparticles of 10, 25–30, and 80 nm diametersTreatment with 5, 10, 25, 50, and 100 μg/mL culture medium for 24 hC18-4 spermatogonial stem cell line
  • At concentrations ≥ 10 mg/mL, Ag-NPs induced a significant decline in SSC proliferation, which was also dependent on their size and coating; At a concentration of 10 mg/mL, Ag-NPs specifically interact with Fyn kinase downstream of Ret and impair SSC proliferation.
[47]
In vitroAu-NPs (5–65 nm)Treatment with 0.5–50 μM for 2 hBovine spermatozoa
  • 22% loss in sperm motility after exposure to the highest dose.
[48]
In vitroAu-NPs (~2.5 nm in diameter)Treatment with concentrations of 0.5 × 1015 or 1 × 1015 particles/mL for 20 and 40 minEpididymal sperm samples collected from the epididymis of hybrid mice CBA × C57B1/6
  • Au-NPs possess spermatotoxicity, causing an inhibition of chromatin decondensation process in gametes.
[49]
In vitroAg-NPs (65 nm)Aliquots of total semen were incubated at 37 °C for 60 min and 120 min at the concentration of 125, 250 and 500 μMSemen samples obtained from 10 healthy donors
  • Decline in sperm motility and viability even at the lowest concentration used; The cytotoxic effect occurs in a dose dependent manner; Sperm viability was always higher than the percentage of sperm motility meaning that spermatozoa were viable but immotile.
[50]
In vitroAg-NPs (20 nm) and TiO2-NPs (21 nm)Cells were exposed for 24, 48 and 72 h to 10, 50 and 100 μg/mL, equivalent to 7.8, 15.6 and 31 μg/cm2, respectivelyNtera2 (NT2, human testicular embryonic carcinoma cell line) and primary testicular cells from C57BL6 mice of wild type (WT) and 8- oxoguanine DNA glycosylase knock-out (KO, mOgg1−/−) genotype
  • Ag-NPs are more cytotoxic and cytostatic compared to TiO2-NPs, causing apoptosis, necrosis and decreased proliferation in a concentration- and time-dependent manner.
[51]
In vivoTiO2-NPsIntraperitoneal injection of 200 and 500 μg/kg every other day for five timesICR mice
  • No significant changes in the low dose group; Reduced sperm density and motility, increased sperm abnormality and germ cell apoptosis in the high dose group.
[52]
In vivoTiO2-NPs (anatase form, particle size of 25–70 nm, surface area 20–25 m2/g)Subcutaneous injections of 100 μL of 1 mg/mL solutionPregnant Slc: ICR mice
  • Decrease of DSP and epidydimal sperm motility in male offspring.
[35]
In vivoCB-NPs (14, 56 and 95 nm with a surface area of 300, 45 and 20 m2/g, respectively)Intratracheal administration of 0.1 mg/kg body weight every week for 10 timesICR mice
  • The DSP decreased by 33% in the 14 nm CB group, by 33% in the 56 nm CB group and by 23% in the 95 nm CB group.
[36]
In vivoCarboxylate-functionalized (diameter of 20–30 nm, length 0.5–2.0 μm) and Amine-functionalized (diameter of 20–30 nm, length 0.5–2.0 μm) MWCNTsMice were randomly divided into 12 groups (15-day single dose, 15-day multi-dose, 60-day multi-dose, and 90-day multi-dose groups). Mice were given injections (5 mg/kg) via the tail vein once (single dose) or every 3 days for 5 timesBALB/c mice
  • No alterations in the total sperm concentration, motility, percentage of abnormal semen and loss of acrosome integrity were detected in the treated mice.
[37]
In vivoCB-NPs (14 nm)Intratracheal administration of 0.2 mg/kg body weight of CB-NPs on days 7 and 14 of gestationPregnant female ICR mice
  • The DSP in the fetal carbon nanoparticle-exposed mice decreased by 47% at the age of 5 weeks, by 34% at the age of 10 weeks, and by 32% at the age of 15 weeks.
[38]
In vivoC60 fullerene (3–36 nm)Rats received distilled water containing aqueous solutions of C60HyFn at concentration of 4~μg/kg daily for 5 weeksHealthy and streptozotocin-induced diabetic male Wistar albino rats
  • No significant toxic effects in testicular tissues were observed; In the diabetic rats, C60 fullerene increased sperm motility, epididymal sperm concentration and decreased the abnormal sperm rate, showing an important anti-oxidant activity.
[53]
In vivoDimercaptosuccinic acid coated Fe3O4-NPs (3–9 nm)Intraperitoneal injection in a single dose of 50, 100, 200 and 300 mg/kgFemale pregnant Balb/C mice
  • In male infants of mothers treated with doses higher than 50 mg/kg a significant decrease in spermatogonia, spermatocytes, spermatids and mature sperm was observed.
[54]
In vivoTiO2-NPs (33.2 ± 16.7 nm)Administration by oral gavage of 40, 200 and 1000 mg/kg with volume of suspension 10 mL/kg of mouse weightMale CBAxB6 mice
  • Increase of the testis apoptotic index and of the frequency of round spermatidis with two or more nuclei.
[55]
In vivoAg-NPs with a nominal diameter of 20 ± 5 nmIntravenous injection with a single dose (5 mg/kg or 10 mg/kg) of Ag-NPsWistar rats
  • Decreased epididymal sperm count and increased DNA damage in germ cells at 24 h post-exposure; The most severe adverse effects were caused by the smaller particles.
[56]
In vivomPEG@Au-NP and PEG-NH2@Au-NP (14 nm)Intravenous injection of 45 and 225 mg/kg of mPEG@Au-NP and 45 mg/kg of PEG-NH2@Au-NP at 48 h intervals for 5 daysICR mice
  • No abnormalities in terms of irregular shapes or forms were observed in the sperm collected from the cauda epididymis of animals.
[39]
In vivoNude short MWCNTs (50–200 nm) and synthesized functionalized MWCNTs with polyethylene glycol (PEG) (s-MWCNTs-PEG).Intravenous administration with a single dose of 100 μg/kg body weightKunming mice
  • No toxicity of these nanotubes on male mouse sperm production or mutagenesis in the long term exposure was observed.
[57]
In vivoPowder of nanoparticulate TiO2 (primary particle size was 20.6 nm, in the exposure atmosphere the particle number concentration was 1.7 × 106 particles/cm3 and the major particle size mode was 97 nm); nanosized CB (particle size was 14 nm)Mice were exposed by whole body inhalation, 1 h/day from gestation day 8 to 18, to 42 mg/m3 of aerosolized powder of nanoparticulate TiO2; mice were intratracheally instilled four times during gestation, (days 7, 10, 15 and 18) with 67 μg/animal of nanosized CBPregnant female C57BL/6J mice
  • TiO2-NPs tended to reduce sperm count in the F1 generation; There was no effect on sperm production in the F2 generation originating after TiO2-NPs exposure; F2 offspring, whose fathers were prenatally exposed to CB-NPs, showed lowered sperm production.
[58]
Disruption of normal levels of sex hormones
In vitroTiO2-NPs (25–70 nm); CB-NPs (14 nm); DE-NPsTreatment with 0–1000 μg/mL for 24 and 48 hMouse testis Leydig cell line TM3
  • Induction of StAR gene expression.
[30]
In vitroNRDE-NPsMice were exposed to 152.01 μg/m3 of NRDE-NPs for 8 weeksInterstitial testicular cells, dissected from male C57BL/Jcl mice
  • Testosterone production in interstitial testicular cells was significantly increased both with and without human chorionic gonadotropin stimulus.
[59]
In vivoNRDE-NPs—Averages of mode diameter and number concentrations for low, middle and high doses were 22.48, 26.13 and 27.06 nm; 2.27 × 105, 5.11 × 105 and 1.36 × 106, respectivelyInhalation exposure to low (15.37), middle (36.35) and high (168.84) μg/m3 doses for 4, 8, or 12 weeks (5 h/day, 5 days/week)Fischer F344 rats
  • Exposure to low and medium concentrations of NRDE-NPs significantly increased plasma and testicular testosterone, whereas exposure to a high concentration did not (inverted-U dose-response); No alterations in LH and FSH production were observed.
[33]
In vivoNRDE-NPs—Averages of mode diameter and number concentrations were 26.81 nm and 1.83 × 106, respectivelyInhalation exposure to 148.86 μg/m3 for 5 h daily for 19 gestational daysPregnant Fischer F344 rats and male offspring
  • Decreased FSH and testosterone and increased ir-inhibin levels were observed in the offspring.
[34]
In vivoNRDE-NPs—Averages of mode diameter and number concentrations for low, middle and high doses were 22.48, 26.13 and 27.06 nm; 2.27 × 105, 5.11 × 105 and 1.36 × 106, respectivelyInhalation exposure to low (15.37), middle (36.35) and high (168.84) μg/m3 doses for 4, 8, or 12 weeks (5 h/day, 5 days/week)Fischer F344 rats
  • Low and medium exposure to NRDE-NPs significantly increased StAR- and P450scc-mRNA and their protein expressions in the testis of rats, in which the elevation pattern was very similar to that of plasma testosterone levels.
[60]
In vivoCB-NPs (14, 56 and 95 nm with a surface area of 300, 45 and 20 m2/g, respectively)Intratracheal administration of 0.1 mg/kg body weight for 10 times every weekICR mice
  • Increased testosterone levels.
[36]
In vivoCarboxylate-functionalized (diameter of 20–30 nm, length 0.5–2.0 μm) and Amine-functionalized (diameter of 20–30 nm, length 0.5–2.0 μm) MWCNTsMice were randomly divided into 12 groups (15-day single dose, 15-day multi-dose, 60-day multi-dose, and 90-day multi-dose groups). Mice were given injections (5 mg/kg) via the tail vein once (single dose) or every 3 days for 5 timesBALB/c mice
  • MWCNT exposure did not alter plasma levels of testosterone, LH, and FSH.
[37]
In vivoCB-NPs (14 nm)Intratracheal administration of 200 μg of CB-NPs on days 7 and 14 of gestationPregnant female ICR mice
  • No significant increase in testosterone levels was observed in fetal carbon nanoparticle- exposed mice.
[38]
In vivoNRDE-NPsMice were exposed to 152.01 μg/m3 of NRDE-NPs for 8 weeksC57BL/Jcl mice
  • Increased levels of testosterone; This result was related to the enhanced expression of genes involved in testicular cholesterol synthesis, such as HMG-CoA, LDL-R, SR-B1, PBR, P450scc, P450 17α, and 17β-HSD.
[59]
In vivomPEG@Au-NP and PEG-NH2@Au-NP (14 nm)Intravenous injection of 45 and 225 mg/kg of mPEG@Au-NP and 45 mg/kg of PEG-NH2@ Au-NP at 48 h intervals for 5 daysICR mice
  • The administration of PEG-NH2@Au-NP caused a significant increase in testosterone levels; No alterations in LH and FSH plasma values were detected.
[39]
In vivoNRDE-NPsInhalation exposure to low (38 ± 3 μg/m3) and high (149 ± 8 μg/m3) NRDE-NP concentrationsFischer F344 rats
  • High and low NRDE-NP concentrations increased plasma and testicular testosterone levels.
[61]
Ag-HC-NPs, Hydrocarbon-coated silver Nanoparticles; Ag-NPs, Silver Nanoparticles; Ag-PS-NPs Polysaccharide-coated silver Nanoparticles; Au-NPs, Gold Nanoparticles; C60HyFn, hydrated C60 fullerene; CB, Carbon Black; CB-NPs, Carbon Black Nanoparticles; CeO2-NPs, Cerium Oxide Nanoparticles; DE-NPs, Diesel Exhaust Nanoparticles; DSP, Daily Sperm Production; Eu2O3-NPs, Europium Oxide Nanoparticles; EuOH3-NPs, Europium Hydroxied Nanoparticles; Fe3O4-NPs, Iron Oxide Nanoparticles; FSH, Follicle Stimulating Hormone; HMG-CoA, 3-hydroxy-3-methylglutaryl-coenzyme A; LDL-R, Low Density Lipoprotein Receptor; LH, Luteinizing Hormone; mPEG@Au-NP, ω-methoxy poly(ethylene glycol) capped gold-NPs; MoO3, molybdenum trioxide; MWCNTs, Multi Walled Carbon Nanotubes; NRDE-NPs, Nanoparticle-Rich Diesel Exhaust Nanoparticles; nSP, amorphous nanosilica particles; P450 17α, Cytochrome P450 17α; P450 17β-HSD, Cytochrome P450 17β-HydroxySteroid Dehydrogenase; P450scc, Cytochrome P450 side-chain cleavage; PBR, Peripheral-type Benzodiazepine Receptor; PEG-NH2@Au-NP, ω-aminoethyl poly(ethylene glycol) capped gold-NPs; s-MWCNTs-PEG, synthesized functionalized MWCNTs with polyethylene glycol; SR-B1, Scavenger Receptor class B type 1; SSC, Spermatogonial Stem Cells; StAR, Steroidogenic Acute Regulatory protein; TiO2, Titanium Dioxide; TiO2-NPs, Titanium Dioxide nanoparticles; ZnO-NPs, Zinc Oxide Nanoparticles.
Table 3. Effects of NPs on the female reproductive system.
Table 3. Effects of NPs on the female reproductive system.
Type of studyType and physico-chemical properties of NPsExperimental protocolCell line/laboratory animalsResultsReferences
Effects on ovarian cells
In vitroAnatase TiO2-NPs (30 nm)Treatment with increasing concentrations of TiO2-NPs (0–100 μg/mL) for 24 hCHO-K1
  • Reduction of cell viability according to the increasing dose applied and the increasing ROS concentration induced.
[68]
In vitroTiO2-NPs (anatase-80% rutile-20% with organic coating, ~21 nm; anatase-80% rutile-20% doped with di-iron trioxide, ~21 nm; anatase-80% rutile-20%, ~21 nm; rutile 100% with inorganic and organic coating, 14 nm; anatase 100% with inorganic coating, 60 nm; rutile 100% with inorganic and organic coating, 20 nm; rutile 100% with inorganic and organic coating, 15 nm; rutile 100% with inorganic coating, 20–22 nm)Exposure for 3 h to different concentrations of TiO2-NPs (800, 1950, 3000, 5000 μg/mL)CHO-WBL
  • None of the titanium dioxide particles tested induced any increase in chromosomal aberration frequencies either in the absence or presence of UV.
[69]
In vitroTiO2-NPs (rutile-79% anatase-21%, particle size: 140 ± 44 nm, surface area: 38.5 m2/g)Cytogenetic evaluations were conducted exposing cells to 750, 1250, and 2500 μg/mL for the 4 h non-activated test condition, to 62.5, 125, and 250 μg/mL, for the 4 h activated test condition, and to 25, 50, and 100 μg/mL for the 20 h non-activated test condition.CHO
  • Genotoxicity tests demonstrated that TiO2-NPs were negative in in vitro mammalian chromosome aberration test with Chinese hamster ovary cells; TiO2-NPs did not induce structural or numerical chromosome aberrations in this study.
[70]
In vitroAnatase TiO2-NPs (10–20, 50–60 nm) and rutile TiO2-NPs (50–60 nm)Treatment with increasing concentrations of TiO2-NPs (25–325 μg/mL) for 24 hCHO
  • Results showed toxic effects on CHO cells with a toxic order as 10–20 nm anatase > 50–60 nm anatase > 50–60 nm rutile, which was consistent with that of 8-OHdG generation and DNA strand breaking activities.
[71]
In vitroTiO2-NPs (shape: complex; average particle sizes: 20 ± 7 nm; specific surface area: 142 m2/g) and Al2O3-NPs (shape: spherical; average particle sizes: 28 ± 19 nm; specific surface area: 39 m2/g)Treatment with increasing concentrations of NPs (0.5, 1, 5, 10, 25, 50 and 100 μg/mL) for 24 hCHO-K1
  • Results showed a dose-related cytotoxic effect manifested after 24 h by changes in lysosomal and mitochondrial dehydrogenase activity; The cytotoxic effects induced by Al2O3-NPs were weaker than those exerted by TiO2-NPs, and began at higher concentrations; Genotoxic effects were shown by MN frequencies which significantly increased at 0.5 and 1 μg/mL TiO2 and 0.5–10 μg/mL Al2O3; SCE frequencies were higher for cells treated with 1–5 μg/mL TiO2-NPs.
[72]
In vitroSiCNW (diameter of 80 nm; chemical composition: Si to C ratio close to 1:1)Exposure to different concentrations (0.5, 1.0, 5.0 and 10.0 μg/mL) of SiCNWs for 1, 3, and 5 daysCHO
  • CHO cells in contact with SiCNWs had significantly lower reproduction rates and genomic instability; Activation of the MAPKs cellular signaling pathway and over-expression of COX-2 were the main toxicity mechanisms; MN ratio rose sharply with increasing SiCNWs concentrations and very significant differences were found after exposure to 5 and 10 μg/mL SiCNWs.
[73]
In vitroCalcium phosphate-NPs (20–30 nm, less than 10% particles were greater than 100 nm)Cells were divided into different groups of exposure: control (treated with 4-androstene-3,17-dione), group II (treated with 10 μM of Calcium phosphate-NPs and 4-androstene-3, 17-dione) and group III (treated with 100 μM of Calcium phosphate-NPs and 4-androstene-3, 17-dione)Granulosa cells collected from infertilite women
  • Increased percentage of cells in S phase; Increased apoptotic rate after the cells were treated with 100 μM of calcium phosphate-NPs for 48 h.
[74]
In vitroMWCNTs (diameter ~10 nm)In order to observe if the MWNT sheets have any toxic effect on cells, CHO cells were allowed to grow on the substrates until about 90% confluenceCHO
  • No toxicity was observed; The cells grew well and aligned and only a few cells elongated along the axis of the MWCNT bundles.
[75]
In vitroAnatase TiO2-NPs (<25 nm)Cells were maintained under exponential growth conditions and were continuously exposed for 1, 2, or 60 days to either 0, 10, 20, 40, 100 and 200 μg/mL of NPsCHO-K1
  • Cell viability or proliferative ability was negatively affected by acute exposure at concentrations of 100 μg/mL and 200 μg/mL as measured by cell counts and colony formation; No genotoxic effects.
[76]
In vitroAnatase TiO2-NPs (<25 nm)Treatment with different concentrations of NPs (0, 25, 50, 100 and 200 μg/mL). Cells were incubated for 24 h and 48 h.CHO-K1
  • Based on the trypan blue assay, viability decreased at the two higher concentrations; The decrease in cell viability using the MTT assay was only observed at the highest concentration and at 48 h incubation.
[77]
In vitroNaked mesoporous silica NPs (10 nm and 50 nm); carboxyl or amine modified mesoporous silica NPs; carboxyl-modified polystyrene-NPs (30 nm); polystyrene-NPs functionalized with amine groups (50 nm)Exposure of NIH-OVCAR3 for 48 h to 30 and 75 μg/mL of naked mesoporous silica NPs (10 nm); exposure of NIH-OVCAR3 and SKOV3 cells for 1 and 24 h to 20 μg/mL of naked mesoporous silica NPs (50 nm) and of carboxyl or amine modified mesoporous silica NPs; exposure of NIH-OVCAR3 and SKOV3 cells for 24 and 48 h to 75 μg/mL of carboxyl-modified polystyrene-NPs and polystyrene-NPs functionalized with amine groupsOvarian NIHOVCAR3 epithelial cancer cells; SKOV3 cancer cells
  • No altered cell morphology, metabolism or cell loss was detected in NIH-OVCAR3 and SKOV3 cells treated with naked mesoporous silica NPs or carboxyl or amine modified mesoporous silica NPs; Evident internalization of the NPs could be observed. Carboxyl-modified polystyrene-NPs were not toxic for NIH-OVCAR3 and SKOV3 cells; Polystyrene NPs functionalized with amine groups showed significant cytotoxic effects.
[78]
In vitroMWCNTs (average diameter: 67 nm; surface area: 26 m2/g; carbon purity: 99.79 wt%)Exposure for 24 h to 1, 10 and 100 μg/mL of NPsMARCO-transfected CHO-K1 cells
  • MWCNTs are highly cytotoxic; The toxicity of MWCNTs may be due to the incomplete inclusion of these NPs by the membrane structure.
[79]
Effects on oogenesis and follicle maturation
In vitroTiO2-NPs (25 nm)Exposure to increasing concentrations (12.5–50 μg/mL) of TiO2-NPsRat preantral follicles
  • TiO2-NPs inhibited rat follicle development and oocyte maturation; Survival rate of follicles, formation rate of antral follicles and release rate of cumulus-ocyte cell complexes were dose-dependently decreased.
[80]
In vitroCdSe-core-QDs and ZnS-coated CdSe QDs (3.5 nm)Exposure for 24 h to 0, 125, 250 and 500 nM of CdSe-core-QDs and to 500 nM of ZnS-coated CdSe QDsCumulus-oocyte complexes collected from female ICR mice
  • CdSe-core-QDs, but not ZnS-coated CdSe QDs, induced dose-dependent decrease in oocyte maturation rate, reduced fertilization, impairment of cell proliferation and dose-dependent enhanced blastocyst apoptotic rate.
[81]
In vitroLysine coated CdSe/CdS/ZnS QDs (~20 nm)Exposure for 4, 8, 16 and 24 h to 5.78 nmol/L and 29.8 nmol/L of Lysine coated CdSe/CdS/ZnS QDsImmature oocytes of 28 days Kunming mice
  • Oocyte maturation process has high susceptibility for disturbances induced by CdSe/CdS/ZnS QDs; CdSe/CdS/ZnS QDs interfere with the process of oocyte maturation disfunctioning the cumulus cells or disturbing the signal interaction between germ cell and somatic cell.
[82]
In vitroCdSe/CdS/ZnS QDsExposure for 4, 8 and 20 h to 28.9 nmol/L of QDsImmature oocytes were collected from Kunming mice
  • QDs enter cumulus cells and accumulate with co-culture time; After being treated for 20 h and being rejected by oocytes, QDs decrease the ratio of oocyte in vitro maturation dramatically.
[83]
In vitroCdTe/ZnTe QD-Transferrin bioconjugatesPreantral follicles were treated with escalating concentrations of (0.0289, 0.289, 2.89 and 28.9 nmol/L) CdTe/ZnTe QD-Transferrin bioconjugates for 8 daysPre antral follicles were collected from female Kunming mice
  • Dose-dependent up-take of QDs-Tf by theca and granulosa cells; A significant decrease in the rate of antrum cavity formation and the ratio of oocytes with first polar body was observed.
[84]
In vivoTiO2-NPs (anatase; NPs were found to aggregate in culture medium and consequently the mean sizes were 240–280 nm (0.1 mg/L) and 259–360 nm (1.0 mg/L)Reproductively active fish were exposed for 13 weeks to 0.1 and 1.0 mg/LZebrafish Danio rerio
  • Chronic exposure to 0.1 mg/L of TiO2-NPs can significantly impair zebrafish reproduction; Reduced expression of genes coding for growth factors implicated as paracrine stimuli in the oocyte maturation.
[85]
In vivoTiO2-NPs (anatase, average particle size: 6 nm, surface area: 174.8 m2/g)TiO2-NPs suspensions at different concentrations (2.5, 5, and 10 mg/kg of body weight) were administered to mice by intragastric administration for 90 consecutive days.CD-1 (ICR) female mice
  • TiO2-NPs can accumulate in the ovary and result in ovarian damage, causing an imbalance of mineral element distribution, decreased fertility or pregnancy rate and oxidative stress in mice.
[86]
In vivoAg-NPs (3 and 35 nm)Exposure to 10 μg/L of Ag-NPs for 35 daysSheepshead minnows (Cyprinodon Variegatus)
  • Gene expression analysis revealed a dramatic transcriptional response indicating that exposure to these NPs has the potential to cause reproductive dysfunction; Absence of ovary morphological and developmental alterations.
[87]
In vivoTiO2-NPs (anatase-75% rutile-25%, average primary particle size: 21 nm, specific surface area: 50 ± 15 m2/g)Reproductively active fish were exposed for 14 days to 0.1 and 1.0 mg/LZebrafish Danio rerio
  • Female gonad showed a normal spread of oocyte development stages; The highest TiO2-NPs dose caused a markedly lower production of eggs and viable embryos.
[88]
Disruption of normal levels of sex hormones
In vitroAu-NPs (10 nm)Exposure for 1,3,5 and 24 h to 2.85 × 1010 NPs/mLRat granulosa cells
  • Au-NPs induced a greater output of estradiol at 1–5 h of treatment; A significant decrease in estradiol was observed at 24 h.
[89]
In vitroCalcium phosphate-NPs (20–30 nm, less than 10% particles were greater than 100 nm)Cells were divided into different groups of exposure: control (treated with 4-androstene-3,17-dione), group II (treated with 10 μM of Calcium phosphate-NPs and 4-androstene-3, 17-dione) and group III (treated with 100 μM of Calcium phosphate-NPs and 4-androstene-3, 17-dione) for 48 hGranulosa cells collected from infertile women
  • Treatment with calcium phosphate-NPs did not significantly change either the progesterone or estradiol levels in culture fluid; No changes in the expression levels of mRNAs encoding P450scc, P450arom and StAR.
[74]
In vitroCdTe/ZnTe QD-Transferrin bioconjugatesPreantral follicles were treated with escalating concentrations of (0.0289, 0.289, 2.89 and 28.9 nmol/L) CdTe/ZnTe QD-Transferrin bioconjugates for 8 daysPre antral follicles were collected from female Kunming mice
  • Increase in 17 β-estradiol secretion, at the 4th-8th day post-treatment, at the highest QD-Tf concentration;
  • 17 β-estradiol secretion decreased compared to controls at lower QD-Tf concentrations.
[84]
In vivoTiO2-NPs (anatase, average particle size: 6 nm, surface area: 174.8 m2/g)TiO2-NP suspensions at different concentrations (2.5, 5, and 10 mg/kg of body weight) were administered to mice by intragastric administration for 90 consecutive days.CD-1 (ICR) female mice
  • Higher serum levels of estradiol; Lower serum levels of progesterone, FSH, LH and testosterone.
[86]
In vivoNRDE-NPs (22–27 nm; particle composition showed a higher percentage of organic carbon than elemental carbon)Inhalation exposure, for 5 h daily from day 1 to 19 of gestation, to 148.86 μg/m3Pregnant Fischer 344 rats
  • Increase in LH levels; Decrease in progesterone levels; No significant differences were observed in FSH, prolactin, ir-inhibin and testosterone levels.
[90]
In vivoZnO-NPs (20–30 nm)Oral administration of 333.33 mg/kg of ZnO-NPsWistar rats
  • The sexual hormone level (FSH, LH and estradiol) showed no significant difference between control and treatment group.
[91]
8-OHdG, 8-hydroxy-2-deoxyguanosine; Ag-NPs, Silver Nanoparticles; Al2O3-NPs, Aluminium oxide Nanoparticles; Au-NPs, Gold Nanoparticles; CdS-QDs, Cadmium Sulfide Quantum Dots; CdSe-QDs, Cadmium Selenium core Quantum Dots; CdTe/ZnTe QD, Cadmium Tellurium/Zinc Tellurium Quantum Dots; COX-2, Cyclooxygenase-2; FSH, Follicle Stimulating Hormone; LH, Luteinizing Hormone; MAPKs, Mitogen-Activated Protein Kinases; MN, Micro Nuclei; MARCO, Macrophage Receptor with Collagenous structure; MWCNTs, Multi Walled Carbon Nanotubes; MTT, 3-(4,5-dimethyl-2-thiazol)-2,5-diphenyl-2H-tetrazolium bromide; NRDE-NPs, Nanoparticle-Rich Diesel Exhaust Nanoparticles; P450arom, Aromatase P450; P450scc, Cytochrome P450 side-chain cleavage; ROS, Reactive Oxygen Species; SCE, Sister Chromatid Exchanges; SiCNWs, Silicon Carbide Nanowires; StAR, Steroidogenic Acute Regulatory protein; TiO2, Titanium Dioxide; TiO2-NPs, Titanium Dioxide nanoparticles; UV, Ultra Violet; ZnO-NPs, Zinc Oxide Nanoparticles; ZnS, Zinc Sulfide; ZnS-QDs, Zinc Sulfide Quantum Dots.
Table 4. Estrogenic effects of NPs.
Table 4. Estrogenic effects of NPs.
Type of studyType and physico-chemical properties of NPsExperimental protocolCell line/laboratory animalsResultsReferences
In vitroCdTe-QDs (~3 nm)Treatment with 0.5 and 10 μg/mL for time periods ranging from 5 min to 96 hHuman breast cancer MCF-7 cells
  • CdTe-QDs induced estrogen-related genomic and non-genomic signaling pathway; Increased estrogen receptor- α activation and biphasic phosphorylation of AKT and ERK1/2.
[99]
In vivoCdS-QDs (4.2 ± 1 nm)Exposure for 21 days to 5, 50 and 500 μg/L of CdS-QDsMale sticklebacks (Gasterosteus aculeatus)
  • Induction of VTG synthesis was not detected in any of the treatment groups.
[95]
In vivoNRDE-NPs—Averages of mode diameter and number concentrations for low, middle and high doses were 22.48, 26.13 and 27.06 nm; 2.27 × 105, 5.11 × 105 and 1.36 × 106, respectivelyInhalation exposure to low (15.37), middle (36.35) and high (168.84) μg/m3 doses for 4, 8, or 12 weeks (5 hours/day, 5 days/week)Fischer F344 rats
  • Higher plasma progesterone levels after exposure for 4 weeks to middle or high NRDE-NPs concentrations; Lower plasma progesterone levels in rats treated with low or high NRDE-NPs after 8 weeks.
[33]
In vivoNRDE-NPs—Averages of mode diameter and number concentrations were 26.81 nm and 1.83 × 106, respectivelyInhalation exposure to 148.86 μg/m3 for 5 h daily for 19 gestational daysPregnant Fischer F344 rats
  • Significantly decreased serum progesterone concentrations in male offspring.
[34]
In vivo(CdS)/CdTe capped-QDsExposure for 48 h to increasing concentrations of (CdS)/CdTe capped-QDs (1, 2 and 6 μg/L)Juvenile rainbow trout (Oncorhynchus mykiss)
  • Up-regulation of VTG; Down-regulation of VTG receptor.
[94]
In vivoC60 fullereneMale zebrafish (Danio rerio) were fed for 5 days with brine shrimp preparations that had accumulated a mixture of C60 fullerene (10% v/v of the 600 mg C60/900 mL water) and 1 μg/L of 17α-ethinylestradiol or C60 fullerene or 17α-ethinylestradiol alone.Male zebrafish (Danio rerio)
  • The treatment with C60 fullerene reduced the biodisponibility of synthetic estrogen, 17α-ethinylestradiol.
[97]
In vivoCB-NPs (50–60 nm)Intratracheal instillation at a total dose of 11, 54 and 268 μg/animal during gestation (7, 10, 15 and 18 gestation days)C57BL/6 mice
  • Exposure to 11 μg/animal induced a significantly earlier onset of puberty, assessed as time of vaginal opening.
[100]
In vivoC60 fullereneExposure of male zebrafish (Danio rerio) to increasing concentrations of C60 fullerene. See Park et al. (2010)Male zebrafish (Danio rerio)
  • Bioavailability of 17α-ethinylestradiol was reduced with increasing concentration of C60 fullerene and was related to computed surface area of C60 fullerene.
[98]
In vivoAg-NPs (20 nm)Exposure to increasing concentrations of Ag-NPs (0.06, 0.6 and 6 μg/L) for 96 hJuvenile rainbow trout (Oncorhynchus mykiss)
  • Ag-NPs induced a significant decrease in liver expression of VTG-like proteins.
[93]
In vivoNRDE-NPs Low dose group (average of mode diameter: 22.78 ± 0.39 nm; mass concentration: 41.73 ± 0.58 μg/m3; number concentration: 8.21 × 105 ± 3.1 × 105), High dose group (average of mode diameter: 26.31 ± 0.38 nm; mass concentration: 152.01 ± 1.18 μg/m3; number concentration: 1.8 × 106 ± 5.18 × 105)Exposure for 8 weeks (5 h/day, 5 days/week) to 41.73 μg/m3 and 152.01 μg/m3 of NRDE-NPsMale C57BL/Jcl mice
  • Corticosterone and progesterone concentrations increased significantly in mice exposed to low concentration; Corticosterone and progesterone concentrations decreased significantly in mice exposed to high-concentration.
[96]
In vivoAg-NPs (23.5 ± 4.4 nm)Exposure to 1 and 25 μg/L of Ag-NPs for 28 daysMale Medaka (Oryzias latipes) fish
  • Conspicuous mRNA induction of VTG-1 and Chg-L in livers of exposed fish.
[92]
Ag-NPs, Silver Nanoparticles; AKT, serine/threonine-protein kinases; CB-NPs, Carbon Black Nanoparticles; CdS-QDs, Cadmium Sulfide Quantum Dots; CdS/CdTe capped-QDs, Cadmium Sulfide/Cadmium Tellurium capped-Quantum Dots; CdTe-QDs, Cadmium Tellurium Quantum Dots; Chg-L, choriogenin L; ERK, extracellular signal-regulated kinase; NRDE-NPs, Nanoparticle-Rich Diesel Exhaust Nanoparticles; VTG, Vitellogenin.
Table 5. Other effects of NPs on endocrine system.
Table 5. Other effects of NPs on endocrine system.
Type of studyType and physico-chemical properties of NPsExperimental protocolCell line/laboratory animalsResultsReferences
Effects on thyroid function
In vitroAg-NPs (2–6 and 10 nm), ZnO-NPs (2–10 and 9 nm) and QDs composed of cadmium telluride (2–10 and 10–15 nm)Exposure of cultured tail fin biopsy to 0.06 μg/L–5.5 mg/L of Ag-NPs, 0.19–10 mg/L of ZnO-NPs and 0.25 μg/L–22 mg/L of QDs for 48 hCultured tail fin biopsy assay derived from Rana catesbeiana tadpoles
  • Reduced levels of transcripts encoding TRβ and RLKI were induced by exposure to 5–10 nM of Ag-NPs and to 0.1 nM of QDs; Treatment with ZnO-NPs had no effect on TRβ and RLKI transcript levels; Ag-NPs were able to affect the TH-action on TRβ and RLKI transcripts according to an “inverted-U” hormesis response.
[105]
In vivoCr-NPs (40–70 nm)Oral administration of 150, 300 and 450 μg/Kg of Cr-NPs for 8 weeksMale Sprague Dawley rats
  • Cr-NPs did not result in significant modifications in TSH, FT3 and FT4 serum levels.
[106]
Effects on insulin action and metabolism
In vitroTiO2-NPs (21 nm)Exposure for 2 h to 50 and 200 μg/mL of TiO2-NPsFao rat hepatoma cells
  • TiO2-NPs impaired insulin response and induced insulin resistance.
[110]
In vitroCeO2-NPs (100 nm)Exposure to 100 nmol/L of CeO2-NPs, alone or in combination with 30 nmol/L of sodium selenite for 1–6 daysPancreatic islets
  • Increased cell viability, ATP/ADP ratio and secretion of insulin in response to glucose stimuli.
[111]
In vivoCrCl3-NPs (40–70 nm)Dietary supplementation for 35 days with 200 μg/kg of CrCl3-NPsCrossbred pigs (Duroc X Landrace X Yorkshire)
  • The treatment with CrCl3-NPs reduced serum insulin and cortisol levels and increased serum insulin-like growth factor.
[112]
In vivoCr-NPs (40–50 nm)Oral administration for 6 weeks of 75, 150, 300, 450, 600 and 1200 ppb of Cr-NPsMale Sprague Dawley rats
  • Addition of 300 and 450 ppb of Cr-NPs reduced the serum insulin level.
[113]
In vivoCr-NPs (40–70 nm)Oral administration of 150, 300 and 450 μg/Kg of Cr-NPs for 8 weeksMale Sprague Dawley rats
  • Dietary supplementation of 150, 300, and 450 μg/kg significantly decreased serum concentrations of insulin and cortisol and increased sera levels of IGF-I.
[106]
In vivoDouble walled-CNTs (0.5–2.5 nm for inner tubes and 1.2–3.2 nm for outer tubes)Intranasal instillation of 1.5 mg/kgMale Swiss mice
  • Increased serum levels of IGF-1.
[114]
In vivoCeO2-NPsIntraperitoneal injection of 60 mg/Kg of CeO2-NPs for 2 weeks, alone or in combination with sodium selenite (5 μmol/kg/day)Male Wistar rats
  • Improvement in biomarkers of diabetes (oxidative stress, ADP/ATP, glycemic and lipid profile).
[115]
Effects on neuroendocrine system
In vitroC60 fullereneExposure for 4 h to 100 μM of C60 fullereneAdrenal chromaffin cells obtained from Wistar rats
  • Treatment with C60 fullerene notably increased cell survival and prevented cell death, including apoptosis.
[116]
In vitroMnO-NPs (40 nm) and Ag-NPs (15 nm)Exposure for 24 h to increasing concentrations (1–100 μg/mL ) of NPsPC-12 cells derived from Rattus norvegicus pheochromocytoma (CRL-1721)
  • MnO-NP dose dependently depleted DA and DOPAC;
  • Ag-NPs significantly reduced DA and DOPAC at concentrations of 50 μg/mL;
  • HVA levels decreased at 50 μg/mL in both MnO-NP and Ag-NP exposure.
[117]
In vitroCu-NPs (90.9 ± 19.3 nm), Mn-NPs (52.1 ± 23.8 nm) and Ag-NPs (18.3 ± 7.3 nm)Treatment for 24 h with 10 μg/mL of NPs and with increasing concentrations of Cu-NPs (2.5, 5, 7.5, 10 and 25 μg/mL)PC-12 cells
  • Cu-NPs induced a significant reduction in the content of DA, DOPAC, and HVA in PC12 cells.
[118]
In vitroAu-NPs (28 nm) and Ag-NPs (61 nm)Treatment for 24 and 48 h with 0.01–1 nM of NPsPrimary culture murine adrenal medullary chormaffin cells harvested from wild-type brown male mice (C57BL/6J)
  • Exposure to Au-NPs and Ag-NPs caused lower and slower secretion of epinephrine molecules.
[119]
In vitroCarboxyl QDs with CdSe core and ZnS shell (7–8 nm)Exposure for 24 h to increasing concentrations of QDs (5, 8, 16 and 36 nM)Mouse chromaffin cells obtained from young C57BL/6J male mice
  • Exposure to carboxyl QDs with CdSe core and ZnS shell impairs Ca2+ influx and severely interferes with the functionality of the exocytotic machinery, compromising the overall catecholamine supply from chromaffin cells.
[120]
In vitroAg-NPs coated with citrate (6 nm) or polyvinylpyrrolidone (21 nm)Exposure for 24 h to different concentrations (1–30 μM) of Ag-NPsPC-12 cells
  • Treatment with citrate- coated Ag-NPs reduced the emergence of the acetylcholine-phenotype; Treatment with polyvinylpyrrolidone -coated Ag-NPs enhanced differentiation into a DA-phenotype.
[121]
In vitroSilica NPs (15 nm)Exposure for 24 h to 25–200 μg/mL of silica NPsPC-12 cells
  • Silica NPs exerted a dose-dependent reduction in DA synthesis related to redundant ROS generation in the cells.
[122]
In vitroCitrated-capped Ag-NPs (from 15 to 60 nm); PEG (25.6 ± 7.8 nm) and heparin (20.6 ± 15.3 nm) surface modified Au NPs.Exposure for 24 h to citrated-capped Ag-NPs at 1 nM concentration and to PEG and heparin Au NPs at 10 μg/mLPrimary culture murine adrenal medullary chormaffin cells harvested from wild-type brown male mice (C57BL/6J)
  • Noble metal NPs disrupt exocytosis, typically altering the number of molecules and kinetics of release.
[123]
Effects on pituitary gland
In vivoCrCl3-NPs (40–70 nm)Dietary supplementation for 35 days with 200 μg/kg of CrCl3-NPsCrossbred pigs (Duroc X Landrace X Yorkshire)
  • No significant difference in GH level between exposed and control group.
[112]
In vivoCrCl3-NPs (40–70 nm)Dietary supplementation for 35 days with 200 μg/kg of CrCl3-NPsCrossbred pigs (Duroc X Landrace X Yorkshire)
  • CrCl3-NPs significantly increased the mean level, lowest value, peak value and peak duration of GH; Pituitary mRNA expression of GH improved significantly.
[124]
In vivoCr-NPs (40–70 nm)Oral administration of 150, 300 and 450 μg/Kg of Cr-NPs for 8 weeksMale Sprague Dawley rats
  • No significant alterations in the concentrations of pituitary produced GH.
[106]
Effects on adrenal gland
In vivoNRDE-NPs—Averages of mode diameter and number concentrations for low, middle and high doses were 22.48, 26.13 and 27.06 nm; 2.27 × 105, 5.11 × 105 and 1.36 × 106, respectivelyInhalation exposure to low (15.37), middle (36.35) and high (168.84) μg/m3 doses for 4, 8, or 12 weeks (5 h/day, 5 days/week)Fischer F344 rats
  • No significant differences in plasma concentrations of corticosterone were observed in the different exposure groups.
[33]
In vivoNRDE-NPs—Averages of mode diameter and number concentrations were 26.81 nm and 1.83 × 106, respectivelyInhalation exposure to 148.86 μg/m3for 5 h daily for 19 gestational daysPregnant Fischer F344 rats
  • Significantly lower corticosterone concentrations were observed in male offspring.
[34]
In vivoCr-NPs (40–70 nm)Oral administration of 150, 300 and 450 μg/Kg of Cr-NPs for 8 weeksMale Sprague Dawley rats
  • Exposure to Cr-NPs decreased plasma cortisol levels.
[106]
In vivoNRDE-NPs Low dose group (average of mode diameter: 22.78 ± 0.39 nm; mass concentration: 41.73 ± 0.58 μg/m3; number concentration: 8.21 × 105 ± 3.1 × 105), High dose group (average of mode diameter: 26.31 ± 0.38 nm; mass concentration: 152.01 ± 1.18 μg/m3; number concentration: 1.8 × 106 ± 5.18 × 105)Exposure for 8 weeks (5 h/day, 5 days/week) to 41.73 μg/m3 and 152.01 μg/m3 of NRDE-NPsMale C57BL/Jcl mice
  • At the highest exposure dose, NRDE-NPs inhibited basal and ACTH-stimulated corticosterone release; The lowest NRDE-NP concentration increased synthesis and release of corticosterone.
[96]
In vivoNRDE-NPs (22–27 nm; particle composition showed a higher percentage of organic carbon than elemental carbon)Inhalation exposure to 148.86 μg/m3 for 5 h daily from day 1 to 19 of gestationPregnant Fischer 344 rats
  • Increased serum levels of corticosterone.
[90]
ACTH, AdrenoCorticoTropic Hormone; ADP, Adenosine Diphosphate; ATP, Adenosine Triphosphate; Ag-NPs, Silver Nanoparticles; Au-NPs, Gold Nanoparticles; CdSe, Cadmium Selenium; CeO2-NPs, Cerium Oxide Nanoparticles; CNTs, Carbon Nanotubes; Cr-NPs, Chromium Nanoparticles; CrCl3-NPs, Chromium Chloride Nanoparticles; Cu-NPs, Copper nanoparticles; DA, Dopamine; DOPAC, dihydroxyphenylacetic acid; FT3, free triiodothyronine; FT4, free thyroxine; GH, Growth Hormone; HVA, HomoVanillic Acid; IGF-I, Insulin-like Growth Factor I; Mn-NPs, Manganese Nanoparticles; MnO-NPs, Manganese Oxide Nanoparticles; NRDE-NPs, Nanoparticle-Rich Diesel Exhaust Nanoparticles; PEG, PolyEthylene Glycol; QDs, Quantum Dots; RLKI, Rana Larval Keratin Type I; ROS, Reactive Oxygen Species; TiO2-NPs, Titanium Dioxide nanoparticles; TRβ, Thyroid hormone Receptor β; TSH, Thyrotropic-Stimulating Hormone; ZnO-NPs, Zinc Oxide Nanoparticles; ZnS, Zinc Sulfide.

Share and Cite

MDPI and ACS Style

Iavicoli, I.; Fontana, L.; Leso, V.; Bergamaschi, A. The Effects of Nanomaterials as Endocrine Disruptors. Int. J. Mol. Sci. 2013, 14, 16732-16801. https://doi.org/10.3390/ijms140816732

AMA Style

Iavicoli I, Fontana L, Leso V, Bergamaschi A. The Effects of Nanomaterials as Endocrine Disruptors. International Journal of Molecular Sciences. 2013; 14(8):16732-16801. https://doi.org/10.3390/ijms140816732

Chicago/Turabian Style

Iavicoli, Ivo, Luca Fontana, Veruscka Leso, and Antonio Bergamaschi. 2013. "The Effects of Nanomaterials as Endocrine Disruptors" International Journal of Molecular Sciences 14, no. 8: 16732-16801. https://doi.org/10.3390/ijms140816732

Article Metrics

Back to TopTop