Skip to main content

RETRACTED ARTICLE: Mechanisms of nanosized titanium dioxide-induced testicular oxidative stress and apoptosis in male mice

This article was retracted on 14 July 2015

Abstract

Background

Due to the increased application of titanium dioxide nanoparticles (TiO2 NPs) in the food industry and daily life, their potential toxic effects in humans and animals have been investigated. However, very few studies have focused on testicular oxidative stress and/or apoptosis.

Methods

In order to understand the possible molecular mechanisms of testicular lesions following exposure to TiO2 NPs, male mice were exposed to 2.5, 5, or 10 mg/kg body weight TiO2 NPs for 90 consecutive days. Testicular oxidative stress and apoptosis were then evaluated, and the testicular mRNA expression of several genes and their proteins involved in oxidative stress and/or apoptosis was investigated.

Results

TiO2 NPs entered Sertoli cells and caused severe testicular oxidative damage and/or apoptosis, accompanied by excessive production of reactive oxygen species and peroxidation of lipids, proteins and DNA as well as a significant reduction in antioxidant capacity. Furthermore, exposure to TiO2 NPs resulted in the up-regulation of caspase-3, Nrbp2, and cytochrome c expression, and caused down-regulation of SOD, CAT, GPx, GST, GR, Cyp1b1, Car3, Bcl-2, Acaa2, and Axud1 expression in mouse testis.

Conclusions

TiO2 NPs entered Sertoli cells via the blood-testis barrier and were deposited in mouse seminiferous cord and/or Sertoli cells, causing oxidative damage and apoptosis.

Background

The use of nanosized materials in consumer and industrial products has increased due to their distinctive physicochemical properties, including high reactivity, color changes, lower melting temperature, and greater solar radiation absorption. In particular, titanium dioxide nanoparticles (TiO2 NPs) have been used in crop production, dietary supplements, food additives, food packaging components, medicine, toothpastes, sunscreens, cosmetics, and waste water treatment [1-11]. This widespread use of TiO2 NPs has inevitably led to harmful biological responses in humans, and animals [12], particularly in the reproductive system [13].

Komatsu et al. suggested that TiO2 NPs could migrate to Leydig cells, decrease the viability and proliferation of these cells, and increase the gene expression levels of heme oxygenase-1 and steroidogenic acute regulatory protein of Leydig cells in mouse testis in vitro [14]. Hou et al. reported that exposure to TiO2 NPs led to obvious morphological changes in follicles, a reduction in follicle survival, reduced formation of antral follicles, and inhibition of rat follicle development and oocyte maturation in vitro [15]. Suzuki et al. found that TiO2 NPs entered the cytoplasm of cultured Chinese hamster ovary cells, but not the nucleus [16]. Di Virgilio et al. observed significant agglomeration of TiO2 NPs on both the plasma membrane and the cytoplasm inducing genotoxic and cytotoxic effects [17]. Exposure to TiO2 NPs can occur via inhalation, transdermal absorption, and ingestion, therefore, the ovaries and testes may also be target organs of toxicity in mammals exposed to TiO2 NPs. The importance of TiO2 NP exposure has been highlighted in in vivo studies. Ema et al. indicated that TiO2 NPs injected subcutaneously into pregnant mice resulted in male offspring with altered spermatogenesis and histopathological changes in the testes [18]. Takeda et al. also studied pregnant mice injected subcutaneously with TiO2 NPs, and TiO2 NPs were observed to migrate to spermatids, Sertoli cells and Leydig cells in the male offspring, resulting in disorganization in seminiferous tubules and reductions in daily sperm production, sperm motility and Sertoli cell number [19]. Male mice exposed to TiO2 NPs by intraperitoneal injection showed marked reductions in sperm numbers and motility, and elevated abnormal sperm and germ cell apoptosis in the testis [20]. Our recent studies demonstrated that TiO2 NPs can migrate to the ovary and testis, resulting in injury, a reduction in fertility, lower sperm concentration and sperm motility, higher malformation rates of sperm, and alterations in the gene expression profile in the damaged ovaries and testes in mice [21,22]. Many conditions or events associated with male infertility can induce oxidative stress and germ cell apoptosis. Given that spermatozoa are rich in polyunsaturated fatty acids, they are very susceptible to attack by reactive oxygen species (ROS) [23,24], while protection from lipid peroxidation is mainly due to the activity of glutathione peroxidase (GPx), an enzyme important for maintaining sperm motility and viability [25,26]. The reproductive system of male mammals mainly consists of the testis, epididymis and ductus deferens. The testis produces spermatozoa and the epididymis stores the spermatozoa and completes sperm maturation. We hypothesize that decreased sperm motility and viability caused by exposure to TiO2 NPs may be associated with testicular oxidative stress and/or apoptosis. To date, no studies have examined whether TiO2 NPs induce testicular oxidative stress and/or apoptosis by assessing the antioxidant capacity, and alterations in oxidative stress and/or apoptosis-related gene expression in male mice. Thus, the aims of the present study were to test the hypothesis that TiO2 NPs induce oxidative stress and/or apoptosis in the testis of mice by causing a reduction in antioxidant capacity and alterations in oxidative stress and/or apoptosis-related gene expression. In addition, the mechanisms of TiO2 NP-induced oxidative stress and/or apoptosis in mouse testis were evaluated.

Results

TiO2 NPs characteristics

XRD measurements showed that TiO2 NPs exhibited the anatase structure (Figure 1), and the average particle size calculated from the broadening of the (101) XRD peak of anatase was approximately 5.5 nm using Scherrer’s equation. TEM demonstrated that the average size of the powder particles (Figure 2a) and NPs suspended in HPMC solvent after incubation ranged from 5—6 nm, respectively (Figure 2b), which was consistent with the XRD results. The sample surface area was generally smaller than that estimated from the particle size, and it would seem that aggregation of the particles may have caused this decrease. To confirm the dispersion and stability of the TiO2 NP suspension, the aggregated size and the ζ potential of TiO2 NPs in HPMC were measured. Following 12, 18, and 24 h incubation, the mean hydrodynamic diameter of TiO2 NPs in HPMC solvent ranged from 210 to 450 nm (mainly 310 nm), as measured by DLS, which indicated that the majority of TiO2 NPs were clustered and aggregated in solution. In addition, the ζ potential was 7.57, 8.53, and 9.28 mV, respectively.

Figure 1
figure 1

The (101) X-ray diffraction peak of anatase TiO 2 NPs. The average particle size was about 5 nm calculated using Scherrer’s equation.

Figure 2
figure 2

Transmission electron microscope images of anatase TiO 2 NPs. (a) TiO2 NP powder; (b) TiO2 NPs suspended in HPMC solvent after incubation for 12 h. TEM images showed that the sizes of the TiO2 NP powder or TiO2 NPs suspended in HPMC solvent for 12 h ranged from 5 to 6 nm, respectively.

Histopathological evaluation

The histological changes in the testis specimens are shown in Figure 3. Unexposed testicular samples showed normal histology. The seminiferous tubules in these samples showed numerous spermatogonia, primary spermatocytes, secondary spermatocytes, spermatids, spermatozoids, Sertoli cells and Leydig cells (Figure 3). However, the samples from the mice exposed to TiO2 NPs exhibited the following severe pathological changes: a significant reduction in the thickness of the germinal layer, a reduction in the number of spermatogonia, rupture of the seminiferous tubules, vacuolation, spermatocyte and Sertoli cell apoptosis or necrosis, and irregular arrangement of spermatocytes and Sertoli cells in the seminiferous tubules (Figure 3). In addition, we also observed a significant number of black agglomerates in the seminiferous tubules exposed to 10 mg/kg of TiO2 NPs (Figure 3). Confocal Raman microscopy further showed a characteristic TiO2 peak in the black agglomerates (148 cm−1), which further confirmed the deposition of TiO2 in the testis (see Raman intensity insets in Figure 3).

Figure 3
figure 3

Histopathological observations of mouse testis following intragastric administration of TiO 2 NPs for 90 consecutive days. The blue circle indicates few sperm, breakages or vacuolation, the yellow circle indicates apoptosis and/or necrosis of Sertoli cells, and the blue arrow indicates damaged seminiferous tubule. The red arrows indicate vacuolation, the yellow arrows indicate irregular arrangement of Sertoli cells, and the red circle indicates black agglomerates in the testis. The green arrow spot denotes a representative cell loaded with TiO2 NPs. The right panel shows the corresponding Raman spectra identifying specific peaks at about 148 cm−1.

Sperm morphology

As shown in Figure 4, exposure to TiO2 NPs induced abnormalies in the sperm head and tail. Head abnormalities included amorphous, hookless, banana-shaped and double-headed sperm. TiO2 NPs also caused the formation of folded, coiled, bent and double-tailed sperm. In addition to these morphological alterations, many sperm tails appeared in the smears of treated mice with detached heads. Sperm also showed an acute bend at the cephalocaudal junction from which fibrils projected out.

Figure 4
figure 4

Morphological changes in mouse sperm following intragastric administration of TiO 2 NPs for 90 consecutive days.

Sertoli cell ultrastructure

Changes in the cell ultrastructure in mouse testicular samples are shown in Figure 5. Sertoli cells in the control group contained elliptical nuclei with homogeneous chromatin (Figure 5); however, cell ultrastructure in mouse testicular tissue treated with TiO2 NPs indicated typical apoptosis, including significant mitochondrial swelling and cristae disappearance, nuclear membrane collapse and chromatin marginalization (Figure 5). In addition, we also observed a significant number of black agglomerates in the cytoplasm and/or nucleus in the TiO2 NP-exposed testicular tissues (Figure 5). Confocal Raman microscopy showed a characteristic TiO2 NP peak in the black agglomerate (148 cm−1), which further demonstrated the aggregation of TiO2 NPs in cells (see the Raman intensity insets in Figure 5). These results suggested that chronic exposure to TiO2 NPs resulted in Sertoli apoptosis in the testis, which may be due to excessive ROS production and increased peroxidation levels.

Figure 5
figure 5

Ultrastructure of Sertoli cells in mouse testis caused by intragastric administration of TiO 2 NPs for 90 consecutive days. The yellow arrow indicates mitochondrial swelling, blushing, and/or cristae disappearance, the green arrow indicates TiO2 NP aggregation in mitochondria, and the red arrow indicates nuclear membrane collapse and chromatin marginalization in Sertoli cells. The green arrow spot denotes a representative cell loaded with TiO2 NPs. The right panel shows the corresponding Raman spectra identifying specific peaks at about 148 cm−1.

Oxidative damage of testis

To confirm the molecular mechanism of TiO2 NP-induced oxidative stress and/or apoptosis of testicular cells, oxidative indices including O2 ·−, H2O2, MDA, PC as well as 8-OHdG levels were examined. As shown in Table 1, O2 ·−, H2O2, MDA, and PC levels were significantly elevated and increased with increasing TiO2 NPs dose (P < 0.05). A marked increase in testicular 8-OHdG level was observed in the TiO2 NP-treated groups (P < 0.05 or 0.01), indicating that excessive ROS production led to DNA damage in the testis due to TiO2 NP–induced toxicity.

Table 1 Oxidative stress in testis of male mice caused by intragastric administration with TiO 2 NPs for 90 consecutive days

Antioxidant defense

The activities of antioxidative enzymes in the testis, including SOD, CAT, APx, GR and GST were examined (Table 2). These enzymes in testes exposed to TiO2 NPs were significantly decreased with increasing TiO2 NP dose (Table 2, P < 0.05). To further explore the effects of TiO2 NP stress on antioxidant capacity, the contents of GSH and ascorbic acid in the testis were examined and are shown in Table 2. Marked decreases in GSH and ascorbic acid in the three TiO2 NP-treated groups were observed (P < 0.05). These results suggested that exposure to TiO2 NPs resulted in decreased ROS removal in mouse testis.

Table 2 Alterations of antioxidative capacity in mouse testis after intragastric administration with TiO 2 NPs for 90 consecutive days

Assay of gene and protein expression

In the present study, GDPH was chosen as the endogenous control gene. The expression level of the GDPH gene was constant, with an expression ratio of almost one in all the samples (data not listed). Therefore, using this gene as a reference, changes in the expression levels of the 13 oxidative stress and/or apoptosis-related genes were evaluated and compared following exposure to TiO2 NPs for 90 consecutive days (Figure 6). The expression levels of SOD, CAT, GPx, GST, GR, Cyp1b1, Car3, Bcl-2, Acaa2, and Axud1 decreased gradually, whereas the expression levels of caspase-3, Nrbp2, and cytochrome c increased significantly in mouse testis with increasing TiO2 NP dose (P < 0.05 or 0.01), respectively. TiO2 NP-induced oxidative stress and/or apoptosis-related molecule expression was also examined at the protein level following exposure to various doses of TiO2 NPs for 90 consecutive days (Figure 7). ELISA showed that TiO2 NPs caused significant reductions in SOD, CAT, GPx, GST, GR, Cyp1b1, Car3, Bcl-2, Acaa2, and Axud1 protein expression, and elevated the levels of caspase-3, Nrbp2, and cytochrome c protein expression in mouse testis (P < 0.05 or 0.01), respectively.

Figure 6
figure 6

Alterations in the mRNA expression of oxidative stress and/or apoptosis-related genes in mouse testis caused by intragastric administration of TiO 2 NPs for 90 consecutive days. *p < 0.05, **p < 0.01, and ***p < 0.001. Values represent mean ± SD (n = 5).

Figure 7
figure 7

Alterations in the expression of oxidative stress and/or apoptosis-related proteins in mouse testis caused by intragastric administration of TiO 2 NPs for 90 consecutive days. *p < 0.05, **p < 0.01, and ***p < 0.001. Values represent mean ± SD (n = 5).

These results are consistent with the histological findings and Sertoli cell ultrastructure in the testicular sections observed in the treated mice.

Discussion

Studies on the oxidative impairments in the male reproductive system in animals due to nanosized materials are limited. In addition, it is not known whether TiO2 NPs enter Sertoli cells inducing oxidative disturbances and/or apoptosis, and the mechanisms involved have not been defined. This has relevance as elevated oxidative stress in the testicular milieu was demonstrated to have profound implications for testicular physiology and sperm function following exposure to nanosized materials [13,24,27].

Oxidative damage can affect many classes of molecules including lipids, proteins, nucleic acids, and sugars. Thus, the cell nucleus, mitochondrial membranes, structural and cytoplasmic proteins, complex carbohydrates, RNA, and DNA may all potentially be affected by oxidative stress [28]. In testicular tissue with high rates of metabolism and cell replication, oxidative stress can be especially damaging, making the antioxidant capacity of the tissue very important. Our data revealed that TiO2 NPs crossed the blood–testis barrier and were deposited in the seminiferous cord, were translocated to Sertoli cells and/or the nucleus, and caused severe pathological changes such as few sperm, sperm breakage, rupture of the seminiferous tubules, vacuolation, irregular arrangement of Sertoli cells, apoptosis, and significant oxidative impairment in the mouse testis. The pathological changes in the testis were similar to those in our previous report [22]. However, Guo et al. suggested that male mice exposed to rutile TiO2 NPs (30 nm) or ZnO NPs (30 nm) by intraperitoneal injection for one week showed significantly increased germ cell apoptosis in the testis without exhibiting significant testicular and/or epididymal lesions [20,29]. Bai et al. suggested that repeated intravenous injections of water-soluble multi-walled carbon nanotubes (20–30 nm) in male mice for 15 days caused oxidative stress in the testis without affecting fertility, and the oxidative stress and testicular damage were repaired by day 60 and 90, respectively [27]. Exposure to black carbon NPs was demonstrated to increase partial vacuolation of the seminiferous tubules [30]. Furthermore, mice intravenously injected with amorphous nanosilica particles (70 nm) for one week, confirmed that the NPs penetrated the blood–testis barrier and the nuclear membranes of spermatocytes, but did not produce any apparent testicular injuries in male mice [31]. We consider that the inconsistent findings from these reports may be due to the different NPs type, size, exposure time, and particularly clearance rates from the circulation and tissues.

Evidence in favor of our central hypothesis was the enhanced levels of MDA, PC and 8-OHdG, as well as ROS levels, and decreased antioxidative enzyme activities including SOD, CAT, GPx, GR and GST and antioxidant contents such as GSH and ascorbic acid in the testes due to TiO2 NP exposure. The occurrence of oxidative impairments in the testis raises the question as to whether this effect could be interpreted as a primary effect of TiO2 NPs in the testis as TiO2 NPs are known prooxidants and their potential to generate free radicals, damage DNA, and induce apoptosis/necrosis is well documented in vitro [32] and in vivo [33]. While, it is difficult to precisely answer this question based on the data obtained in the current study, it can be reasonably postulated that TiO2 NPs do cause significant oxidative stress in mouse testis.

TiO2 NP-induced oxidative damage in the testis responded to modulation of antioxidant capacity. Decreased antioxidant enzymes, ascorbic acid and GSH markedly aggravated TiO2 NPs induced oxidative damage in the testis as shown by elevated MDA and ROS levels in the cytosol and mitochondria. The major antioxidant enzymes in mammals are SOD, CAT, and GPX, the latter necessitating a number of other enzymes, e.g. GR and GST, required for the recycling or elimination of GSH. All of these antioxidant enzymes are expressed in the testis [34-36]. SOD converts O2 ·− to H2O2 and O2, and CAT and GPX reduce H2O2 to H2O and O2. GR reduces glutathione disulfide (GSSG) to the sulfhydryl form, GSH, which is an important cellular antioxidant. Elevated GR activity can increase the GSH/GSSG ratio, which is required for ascorbic acid regeneration [37]. Furthermore, GST, previously known as ligandin, is from a family of eukaryotic and prokaryotic phase II metabolic isozymes best known for their ability to catalyze the conjugation of GSH to xenobiotic substrates for the purpose of detoxification [38]. In the present study, our data suggest that exposure to TiO2 NPs significantly decreased the activities of SOD, CAT, GPX, GR, and GST, and their gene and protein levels, resulting in a higher level of ROS production, increased ROS toxicity, testicular tissue and cell damage in male mice following exposure to TiO2 NPs. A number of non-enzyme factors also function as antioxidants in the testis. Of these, ascorbic acid and GSH have proven efficacy in reducing testicular oxidative stress by removing ROS either directly or indirectly by being involved in the ascorbic acid–GSH cycle under different conditions [39-43]. Ascorbic acid is considered to be a major antioxidant in the testis, and neutralizes ROS and prevents sperm agglutination. It is an electron donor in redox systems, prevents lipid peroxidation, recycles vitamin E and protects against DNA damage induced by the hydrogen peroxide radical [44]. Oral administration of 200 mg of ascorbic acid together with vitamin E and GSH for a period of 2 months significantly reduced hydroxyguanine (8-OH-dG) levels in spermatozoa and increased the sperm count [45]. In the present study, however, the depletion of ascorbic acid and GSH in the testis caused by TiO2 NPs was associated with increases in ROS, MDA, PC, and 8-OH-dG, suggesting that the testis was using antioxidant defenses to prevent oxidative stress, which caused severe oxidative damage in the mouse testis. The oxidative damage and reductions in antioxidants caused by TiO2 NPs were consistent with those in mouse liver [46], kidney [47,48], lung [49,50], brain [51], and ovary [21].

The blood-testis barrier is formed by the tight junctions of Sertoli cells [52]. Therefore, TiO2 NP deposition in the testis is closely related to seminiferous tubule injury or apoptosis of Sertoli cells following exposure to TiO2 NPs. To understand the mechanisms of oxidative stress and/or apoptosis, the expression of oxidative stress and/or apoptosis-related genes and their proteins in the testis was detected and is listed in Figures 6 and 7. Of the genes involved in apoptosis and/or oxidative stress, caspase-3, Nrbp2, and cytochrome c were markedly up-regulated, while Cyp1b1, Car3, Bcl-2, Acaa2, Axud1, SOD, CAT, GPX, GR, and GST were significantly down-regulated in the mouse testis. Caspase-3 is activated in the apoptotic cell both by extrinsic (death ligand) and intrinsic (mitochondrial) pathways [53-56]. It has been reported that TiO2 NPs induced apoptosis in mouse spleen by activating caspase-3 [57]. The present study showed that caspase-3 expression in mouse testis was increased, in concordance with the above-mentioned report on the effects of TiO2 NPs in mouse spleen. Overexpression of Bcl-2 prevents cells from undergoing apoptosis in response to a variety of stimuli. Cytosolic cytochrome c is necessary for initiation of the apoptotic program, suggesting a possible connection between Bcl-2 and cytochrome c, which is normally located in the mitochondrial intermembrane space. Cells undergoing apoptosis were found to have elevated cytochrome c in the cytosol and a corresponding decrease in the mitochondria. Overexpression of Bcl-2 prevented the efflux of cytochrome c from the mitochondria and the initiation of apoptosis [58,59]. In the present study, Bcl-2 expression was significantly decreased, whereas cytochrome c was markedly increased in the TiO2 NP-exposed mouse testis, suggesting that Bcl-2 may play an important role in TiO2 NP-induced Sertoli apoptosis in the mouse testis. These results were similar to those of TiO2 NPs-induced apoptosis in mouse spleen [60]. Nrbp2 belongs to the protein kinase superfamily, and was found to be expressed in a subset of tumor cells in human medulloblastoma and protected nestin-positive neural progenitors against apoptosis during differentiation [61]. Nrbp2 upregulation in the mouse testis may prevent Sertoli cell apoptosis and may be a response to TiO2 NP-induced toxicity. Cao et al. indicated that Acaa2 abolished Bc1-2/adenovirus E1B 19 kD-interacting protein 3 (BNIP3)-mediated apoptosis and mitochondrial damage [62]. Axud1 is a member of the gene family encoding nuclear proteins which contain cysteine- and serine-rich domains, and is often considered to be related to reduced apoptosis [63]. Therefore, Sertoli apoptosis in testicular tissue caused by TiO2 NPs may be due to a marked reduction in Acaa2 and Axud1 expression. Of the genes and their proteins involved in oxidative stress, SOD, CAT, APX, GR, and GST were highly suppressed. SOD catalyses the dismutation of superoxide anion by successive oxidation-reduction of the transition metal at the enzyme’s active site [64]. CAT converts hydrogen peroxide to hydrogen and water. GPX catalyzes the breakdown of H2O2 and organic hydroperoxides. GPX can directly reduce phospholipid hydroperoxides inside biological membranes to lipid and water, and is essential for survival and important in repairing damage due to lipid peroxidation [65]. GPX also protects against mitochondrial apoptosis and decreases in mitochondrial ATP generation in the presence of oxidative stress, and is strongly expressed in the mitochondria of testis and spermatozoa [66,67]. GR plays a key role in oxidative stress by converting GSSG to GSH. Increased GR activity can elevate the GSH/GSSG ratio, which is required for ascorbic acid regeneration [37]. GST is able to conjugate GSH to the toxic reactive compounds, 4-hydroxynonenal and cholesterol α-oxide, which are generated during the oxidation of membranes. The GPx activity of some GST proteins also suggests that they may be important in the detoxification of organic hydroperoxides [68]. Thus, decreased expression of SOD, CAT, GPX, GR, and GST in the mouse testis aggravated TiO2 NP–induced oxidative stress. Car3 has been demonstrated to rapidly glutathionylate in vivo and in vitro when cells are exposed to oxidative stress, and it is one of the most carbonylated proteins in rodent liver [69]. Overexpression of Car3 decreased steady-state levels of intracellular ROS, increased the proliferation rate, and protected cells against H2O2-induced apoptosis [69]. Cyp1b1 is a member of the cytochrome P450 family of proteins and can remove cellular oxygenation products which induce oxidative stress, while a lack of Cyp1b1 increases oxidative stress in vivo and in vitro [70]. Therefore, the generation of oxidative stress in the TiO2 NP–exposed mouse testis may also be closely associated with significant reductions in Car3 and Cyp1b1 expression and removal of ROS.

In summary, TiO2 NPs entered Sertoli cells via the blood-testis barrier and were deposited in mouse testicular tissue and/or Sertoli cells, causing testicular oxidative damage and cell apoptosis. Furthermore, testicular damage following exposure to TiO2 NPs may be related to significant overproduction of ROS, peroxidation of lipids, proteins and DNA, and a significant reduction in antioxidant capacity. Sertoli cell apoptosis caused by TiO2 NPs was associated with up-regulation of caspase-3, Nrbp2, and cytochrome c expression, and down-regulation of SOD, CAT, GPx, GST, GR, Cyp1b1, Car3, bcl-2, Acaa2, and Axud1 expression in mouse testis. Therefore, the application of TiO2 NPs should be performed with caution to reduce potential risks to humans.

Materials and methods

Chemicals

Anatase TiO2 nanoparticles were prepared via controlled hydrolysis of titanium tetrabutoxide. The details of TiO2 NP synthesis were previously described [71,72]. Hydroxypropylmethylcellulose (HPMC) 0.5% w/v was used as a suspending agent. TiO2 powder was dispersed onto the surface of a 0.5% w/v HPMC solution, and then the suspension containing TiO2 particles was treated ultrasonically for 15–20 min and mechanically vibrated for 2 or 3 min.

The particle sizes of both the powder and nanoparticles suspended in 0.5% w/v HPMC solution (5 mg/L) following incubation were determined using a TecnaiG220 transmission electron microscope (TEM) (FEI Co., USA) operating at 100 kV, respectively. In brief, particles were deposited in suspension onto carbon film TEM grids, and allowed to air-dry. The mean particle size was determined by measuring > 100 randomly sampled individual particles. X-ray-diffraction (XRD) patterns of TiO2 NPs were obtained at room temperature with a charge-coupled device (CCD) diffractometer (Mercury 3 Versatile CCD Detector; Rigaku Corporation, Tokyo, Japan) using Ni-filtered Cu Kα radiation. The average aggregate or agglomerate size of the TiO2 NPs after incubation in 0.5% w/v HPMC solution (5 mg/L) for 0, 12 and 24 h was measured by dynamic light scattering (DLS) using a Zeta PALS + BI-90 Plus (Brookhaven Instruments Corp., USA) at a wavelength of 659 nm. The scattering angle was fixed at 90°.

Ethics statement

All experiments were conducted during the light phase, and were approved by the Animal Experimental Committee of Soochow University (Grant 2111270) in accordance with the National Institutes of Health Guidelines for the Care and Use of Laboratory Animals (NIH Guidelines).

Animals and treatment

One hundred sixty CD-1 (ICR) male mice (24 ± 2 g) were purchased from the Animal Center of Soochow University (China). The mice were housed in stainless steel cages in a ventilated animal room. The room temperature in the housing facility was maintained at 24 ± 2°C, with a relative humidity of 60 ± 10% and a 12-h light/dark cycle. Distilled water and sterilized food were available ad libitum. Before treatment, the mice were acclimated to this environment for five days. All the animals were handled in accordance with the guidelines and protocols approved by the Care and Use of Animals Committee of Soochow University (China).

TiO2 NP powder was dispersed onto the surface of 0.5% w/v HPMC and the suspension containing TiO2 NPs was treated ultrasonically for 30 min and mechanically vibrated for 5 min. The mice were randomly divided into four groups (n = 40 in each group), including a control group treated with 0.5% w/v HPMC and three experimental groups treated with 2.5, 5, or 10 mg/kg TiO2 NPs. The mice were weighed, a volume of TiO2 NP suspension was calculated for each mouse, and the fresh TiO2 NP suspension was administered by gavage using a gavage needle each day for 90 days. Symptoms, growth status, eating, drinking, activity and mortality were observed and carefully recorded daily during the 90-day period. After 90 days, the mice were weighed, anesthetized using ether, and then sacrificed. The testes were quickly removed, placed on ice, dissected, and then frozen at −80°C (except those for histopathological examination).

Histopathological examination of testis

All histopathological tests were performed using standard laboratory procedures [73]. Five sets of testicular tissues from 5 mice were embedded in paraffin blocks, sliced into 5 μm thick sections and then placed onto glass slides (five slices from each testis). Following hematoxylin–eosin staining, the slides were observed and photographed using an optical microscope (Nikon U-III Multi-point Sensor System, USA). The pathologist was blind to the identity and analysis of the pathology slides.

Observation of testis ultrastructure

Testes (n = 5 in each group) were fixed in a fresh solution of 0.1 M sodium cacodylate buffer containing 2.5% glutaraldehyde and 2% formaldehyde followed by a 2 h fixation period at 4°C with 1% osmium tetroxide in 50 mM sodium cacodylate (pH 7.2-7.4). Staining was performed overnight with 0.5% aqueous uranyl acetate. The specimens were dehydrated in a graded series of ethanol (75, 85, 95, and 100%), and embedded in Epon 812. Ultrathin sections were obtained, contrasted with uranyl acetate and lead citrate, and observed with a HITACHI H600 TEM (HITACHI Co., Japan). Testicular apoptosis was determined based on the changes in nuclear morphology (e.g., chromatin condensation and fragmentation).

Confocal raman microscopy of testicular sections

Raman analysis was performed using backscattering geometry in a confocal configuration at room temperature using a HR-800 Raman microscope system equipped with a 632.817 nm HeNe laser (JY Co., Fort-de-France, Martinique). It was reported that when the size of the TiO2 NPs reached 6 nm, the Raman spectral peak was 148.7 cm−1 [74]. Laser power and resolution were approximately 20 mW and 0.3 cm−1, respectively, while the integration time was adjusted to 1 s. The testicular specimens (n = 5 in each group) were embedded in paraffin blocks, sliced into 5-μm thick sections, and placed onto glass slides. The slides were dewaxed, hydrated, and then scanned using the confocal Raman microscope.

Oxidative stress assay

Reactive oxygen species (ROS) (O2 ·− and H2O2) production and levels of malondialdehyde (MDA), protein carbonyl (PC), and 8-hydroxy deoxyguanosine (8-OHdG) in the testicular tissues (n = 5 in each group) were assayed using commercial enzyme-linked immunosorbent assay kits (Nanjing Jiancheng Bioengineering Institute, Jiangsu, China) according to the manufacturer’s instructions.

Antioxidant capacity

The activities of superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), glutathione S epoxide transferase (GST), and glutathione reductase (GR), and the contents of reduced glutathione (GSH) and ascorbic acid in the testicular tissues (n = 5 in each group) were determined using commercial assay kits (Nanjing Jiancheng Bioengineering Institute, Jiangsu, China) according to the manufacturer’s instructions.

Assay of gene and protein expression

Total RNA was extracted from individual testes using from the homogenates was isolated using Tripure Isolation Reagent (Roche, USA) according to the manufacturer’s instructions. Probes and cycling condition were optimized in accordance with MIQE guidelines for PCR [75]. Synthesized cDNA was used for the real-time PCR by employing primers designed using Primer Express Software according to the software guidelines. PCR primers used in the gene expression analysis are listed in Table 3. Gene expression levels were calculated as a ratio to the expression of the reference gene, GAPDH and data were analyzed using the ΔΔCt method. The probes for SOD, CAT, GPx, GST, GR, Cyp1b1, carbonic anhydrase III (Car3), caspase-3, Bcl-2, nuclear receptor binding protein 2 (Nrbp2), acetyl-coenzyme A acyltransferase 2 (Acaa2), Axin upregulated 1 (Axud1), and cytochrome c were designed by the manufacturer and purchased from Shinegene Company (Shanghai, China). The RT-qPCR data were processed with the sequence detection software version 1.3.1 following the method of Schefe et al. [76].

Table 3 Real time PCR primer pairs

To determine protein levels of SOD, CAT, GPx, GST, GR, Cyp1b1, Car3, caspase-3, Bcl-2, Nrbp2, Acaa2, Axud1, and cytochrome c in the testes, total protein from the frozen testicular tissues (N = 5 in each group) from experimental and control mice was extracted using Cell Lysis Kits (GENMED SCIENTIFICS INC.USA) and quantified using BCA protein assay kits (GENMED SCIENTIFICS INC.USA). ELISA was performed using commercial kits that were selective for each respective protein (R&D Systems, USA), following the manufacturer’s instructions. The absorbance was measured on a microplate reader at 450 nm (Varioskan Flash, Thermo Electron, Finland), and the concentrations of SOD, CAT, GPx, GST, GR, Cyp1b1, Car3, caspase-3, Bcl-2, Nrbp2, Acaa2, Axud1, and cytochrome c were calculated from a standard curve for each sample.

Statistical analysis

All results are expressed as means ± SD. One-way analysis of variance (ANOVA) was carried out to compare the differences of means among the multi-group data using SPSS 19 software (SPSS, Inc., Chicago, IL, USA). Dunnett’s test was performed when each dataset was compared with the solvent control data. Statistical significance for all tests was judged probability level of 0.05 (P < 0.05).

References

  1. Powell JJ, Faria N, Thomas McKay E, Pele LC: Origin and fate of dietary nanoparticles and microparticles in the gastrointestinal tract. J Autoimmun 2010, 34:226–233.

    Article  Google Scholar 

  2. Weir A, Westerhoff P, Fabricius L, Hristovski K, von Goetz N: Titanium dioxide nanoparticles in food and personal care products. Environ Sci Technol 2012, 46:2242–2250.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Fayaz AM, Balaji K, Girilal M, Kalaichelvan PT, Venkatesan R: Mycobased synthesis of silver nanoparticles and their incorporation into sodium alginate films for vegetable and fruit preservation. J Agric Food Chem 2009, 57:6246–6252.

    Article  Google Scholar 

  4. Lopez-Moreno ML, De La Rosa G, Hernandez-Viezcas JA, Peralta-Videa JR, Gardea-Torresdey JL: X-ray absorption spectroscopy (XAS) corroboration of the uptake and storage of CeO 2 nanoparticles and assessment of their differential toxicity in four hdible plant species. J Agric Food Chem 2010, 58:3689–3693.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Goutailler G, Guillard C, Faure R, Païssé O: Degradation pathway of dicyclanil in water in the presence of titanium dioxide. Comparison with photolysis. J Agric Food Chem 2002, 50:5115–5120.

    Article  CAS  PubMed  Google Scholar 

  6. Gimeno O, Rivas FJ, Beltrán FJ, Carbajo M: Photocatalytic ozonation of winery wastewaters. J Agric Food Chem 2007, 55:9944–9950.

    Article  CAS  PubMed  Google Scholar 

  7. Oberdörster G, Oberdörster E, Oberdörster J: Nanotoxicology: an emerging discipline evolving from studies of ultrafine particles. Environ Health Perspect 2005, 113(7):823–839.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Wang JJ, Sanderson BJS, Wang H: Cyto- and genotoxicity of ultrafine TiO 2 particles in cultured human lymphoblastoid cells. Mutat Res 2007, 628:99–106.

    Article  CAS  PubMed  Google Scholar 

  9. Wang HH, Wick RL, Xing BS: Toxicity of nanoparticulate and bulk ZnO, Al 2 O 3 and TiO 2 to the nematode caenorhabditis elegans. Environ Pollut 2009, 157:1171–1177.

    Article  CAS  PubMed  Google Scholar 

  10. Xiong D, Fang T, Yu L, Sima X, Zhu W: Effects of nano-scale TiO 2 , ZnO and their bulk counterparts on zebrafish: acute toxicity, oxidative stress and oxidative damage. Sci Total Environ 2011, 409(8):1444–1452.

    Article  CAS  PubMed  Google Scholar 

  11. Shi HB, Magaye R, Castranova V, Zhao JS: Titanium dioxide nanoparticles: a review of current toxicological data. Part Fibre Toxicol 2013, 10:15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Oberdörster G, Stone V, Donaldson K: Toxicology of nanoparticles: a historical perspective. Nanotoxicol 2007, 1:2–25.

    Article  Google Scholar 

  13. La Z, Yang WX: Nanoparticles and spermatogenesis: how do nanoparticles affect spermatogenesis and penetrate the blood–testis barrier. Nanomed 2012, 7(4):579–596.

    Article  Google Scholar 

  14. Komatsu T, Tabata M, Kubo-Irie M, Shimizu T, Suzuki KI, Nihei Y, Takeda K: The effects of nanoparticles on mouse testis Leydig cells in vitro . Toxicol in Vitro 2008, 22:1825–1831.

    Article  CAS  PubMed  Google Scholar 

  15. Hou J, Wan XY, Wang F, Xu GF, Liu Z, Zhang TB: Effects of titanium dioxide nanoparticles on development and maturation of rat preantral follicle in vitro . Acad J Second Military Med Univ 2009, 30(8):869–873 (In Chinese).

    Article  CAS  Google Scholar 

  16. Suzuki H, Toyooka T, Ibuki Y: Simple and easy method to evaluate uptake potential of nanoparticles in mammalian cells using a flow cytometric light scatter analysis. Environ Sci Technol 2007, 41(8):3018–3024.

    Article  CAS  PubMed  Google Scholar 

  17. Di Virgilio AL, Reigosa M, Arnal PM, 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.

    Article  PubMed  Google Scholar 

  18. Ema M, Kobayashi N, Naya M, Hanai S, Nakanishi J: Reproductive and developmental toxicity studies of manufactured nanomaterials. Reprod Toxicol 2010, 30(3):343–352.

    Article  CAS  PubMed  Google Scholar 

  19. Takeda K, Suzuki K, 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 2009, 55:95–102.

    CAS  Google Scholar 

  20. Guo LL, Liu XH, Qin DX, Gao L, Zhang HM, Liu JY, Cui YG: Effects of nanosized titanium dioxide on the reproductive system of male mice. Zhonghua Nan Ke Xue 2009, 15:517–522 (in Chinese).

    CAS  PubMed  Google Scholar 

  21. Gao GD, Ze YG, Li B, Zhao XY, Liu XR, Sheng L, Hu RP, Gui SX, Sang XZ, Sun QQ, Cheng J, Cheng Z, Wang L, Tang M, Hong FS: The ovarian dysfunction and its gene-expressed characteristics of female mice caused by long-term exposure to titanium dioxide nanoparticles. J Hazard Mater 2012, 243:19–27.

    Article  CAS  PubMed  Google Scholar 

  22. Gao GD, Ze YG, Zhao XY, Sang XZ, Zheng L, Ze X, Gui SX, Sheng L, Sun QQ, Hong J, Yu XH, Wang L, Hong FS, Zhang XG: Titanium dioxide nanoparticle-induced testicular damage, spermatogenesis suppression, and gene expression alterations in male mice. J Hazard Mater 2013, 258:133–143.

    Article  PubMed  Google Scholar 

  23. Sikka SC: Relative impact of oxidative stress on male reproductive function. Curr Med Chem 2001, 8:851–862.

    Article  CAS  PubMed  Google Scholar 

  24. Murugesan P, Muthusamy T, Balasbramanian K, Arunakarn J: Studies on the protective role of vit-amin C and E against polychlorinated biphenyl (Aroclor 1254)-induced oxidative damage in Leydig cells. Free Radic Res 2005, 39:1259–1272.

    Article  CAS  PubMed  Google Scholar 

  25. Griveau JF, Dumont E, Renard P, Callegari JP, Lannou DL: Reactive oxygen species, lipid-peroxdation and enzymatic defense systems in human spermatozoa. J Reprod Fert 1995, 103:17–26.

    Article  CAS  Google Scholar 

  26. Hu JH, Zan LS, Zhao XL, Li QW, Jiang ZL, Li YK, Li X: Effects of tehalose supplementation on semen quality and oxidative varioables in frozen-thawed bovine semen. J Anim Sci 2010, 88:1657–1662.

    Article  CAS  PubMed  Google Scholar 

  27. Bai YH, Zhang Y, Zhang JP, Mu QX, Zhang WD, Butch ER, Snyder SE, Yan B: Repeated administrations of carbon nanotubes in male mice cause reversible testis damage without affecting fertility. Nat Nanotechnol 2010, 5:683–689.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Pryor WA, Houk KN, Foote CS, Fukuto JM, Ignarro LJ, Squadrito GL, Davies KJA: Free radical biology and medicine: it’s a gas, man! Am J Physiol Regul Integr Comp Physiol 2006, 291:R491–R511.

    Article  CAS  PubMed  Google Scholar 

  29. Guo LL, Liu XH, Qin DX, Gao L, Zhang HM, Liu JY, Cui YG: Study on the effect of nanosized zinc oxide for reproductive system of male mice. J Med Postgra 2010, 23:357–364 (in Chinese).

    CAS  Google Scholar 

  30. 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 2008, 32:337–342.

    Article  PubMed  Google Scholar 

  31. Morishita Y, Yoshioka Y, Satoh H, Nojiri N, Nagano K, Abe Y, Kamada H, Tsunoda S, Nabeshi H, Yoshikawa T, Tsutsumi Y: Distribution and histologic effects of intravenously administered amorphous nanosilica particles in the testes of mice. Biochem Biophys Res Commun 2012, 420:297–301.

    Article  CAS  PubMed  Google Scholar 

  32. Iavicoli I, Leso V, Fontana L, Bergamaschi A: Toxicological effects of titanium dioxide nanoparticles: a review of in vitro studies. Eur Rev Med Pharmacol Sci 2011, 15(5):481–508.

    CAS  PubMed  Google Scholar 

  33. Iavicoli I, Leso V, Bergamaschi A: Toxicological effects of titanium dioxide nanoparticles: a review of in vivo studies. J Nanomater 2012, ᅟ:1–36. doi:10.1155/2012/964381.

    Google Scholar 

  34. Zini A, Schlegel PN: Cu/Zn superoxide dismutase, catalase and glutathione peroxidase mRNA expression in the rat testis after surgical cryptorchidism and efferent duct ligation. J Urol 1997, 158:659–663.

    Article  CAS  PubMed  Google Scholar 

  35. Ischi T, Matsuki S, Iuchi Y, Okada F, Toyosaki S, Tomita Y, Ikeda Y, Fujii J: Accelerated impairment of spermatogenic cells in SOD1-knockout mice under heat stress. Free Radic Res 2005, 39:695–705.

    Google Scholar 

  36. Maiorino M, Bosello V, Ursini F, Foresta C, Garolla A, Scapin M, Sztajer H, Flohe L: Genetic varia-tions of GSH-Px-4 and male infertility in hum ans. Biol Reprod 2003, 68:1134–1141.

    Article  CAS  PubMed  Google Scholar 

  37. Crawford NM, Kahn ML, Leustek T, Long SR: Nitrogen and sulfur. In Biochemistry and molecular biology of plants. Edited by Buchanan BB, Gruissem W, Jones RL. Rockville: ASPP; 2000:786–849.

    Google Scholar 

  38. Sivori JL, Cascorbic acidbe N, Zerba EN, Wood EJ: Induction of glutathione S-transferase activity in Tria-tomainfestans. Mem Inst Oswaldo Cruz 1997, 92:797–802.

    Article  CAS  PubMed  Google Scholar 

  39. Narra VR, Howell RW, Sastry KSR, Rao DV: Vitamin C as a radioprotector against iodine-131 in vivo. J Nucl Med 1993, 34:637–640.

    CAS  PubMed  Google Scholar 

  40. Kutlubay R, Oguz EO, Can B, Guven MC, Sinik Z, Tuncay OL, Vieamen E: Protection from testicular damage caused by intraperitoneal aluminium. Int J Toxicol 2007, 26:297–306.

    Article  CAS  PubMed  Google Scholar 

  41. Armstrong JS, Steinauer KK, Hornung B, Irish JM, Lecane P, Birrell GW, Peehl DM, Knox SJ: Role of glutathione depletion and reactive oxygen species generation in apoptotic signaling in a human B lymphoma cell line. Cell Death Differ 2002, 9:252–263.

    Article  CAS  PubMed  Google Scholar 

  42. Smirnoff N: Antioxidants and reactive oxygen species in plants. Oxford: Blackwell Publishing; 2005.

    Book  Google Scholar 

  43. Jovanović-Galović A, Blagojević DP, Grubor-Lajsić G, Worland R, Spasić MB: Role of antioxidant defense during different stages of preadult life cycle in European corn borer (Ostrinia nubilalis, Hubn.): Diapause and metamorphosis. Arch Insect Biochem Physiol 2004, 55(2):79–89.

    Article  PubMed  Google Scholar 

  44. Angulo C, Maldonado R, Pulgar E, Mancilla H, Córdova A, Villarroel F, Castro MA, Concha II: Vitamin C and oxidative stress in the seminiferous epithelium. Biol Res 2011, 44:169–180.

    Article  CAS  PubMed  Google Scholar 

  45. Kodama H, Yamaguchi R, Fukuda J, Kascorbic acidi H, Tanaka T: Increased oxidative deoxyribonucleic acid damage in the spermatozoa of infertile male patients. Fertl Steril 1997, 68:519–524.

    Article  CAS  Google Scholar 

  46. Cui YL, Gong XL, Duan YM, Li N, Hu RP, Liu HT, Hong MM, Zhou M, Wang L, Wang H, Hong FS: Hepatocyte apoptosis and its molecular mechanisms in mice caused by titanium dioxide nanoparticles. J Hazard Mater 2010, 183:874–880.

    Article  CAS  PubMed  Google Scholar 

  47. Gui SX, Sang XZ, Zheng L, Ze YG, Zhao XY, Sheng L, Sun QQ, Cheng Z, Cheng J, Hu RP, Wang L, Hong FS, Tang M: Intragastric exposure to titanium dioxide nanoparticles induced nephrotoxicity in mice, assessed by physiological and gene expression modifications. Part Fibre Toxicol 2013, 10:4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Gui SX, Li BY, Zhao XY, Sheng L, Hong J, Yu XH, Sang XZ, Sun QQ, Ze YG, Wang L, Hong FS: Renal injury and Nrf2 modulation in mouse kidney following chronic exposure to TiO 2 nanoparticles. Agriic Food Chem 2013, 61:8959–8968.

    Article  CAS  Google Scholar 

  49. Sun QQ, Tan DL, Zhou QP, Liu XR, Cheng Z, Liu G, Zhu M, Sang XZ, Gui SX, Cheng J, Hu RP, Tang M, Hong FS: Oxidative damage of lung and its protective mechanism in mice caused by long-term exposure to titanium dioxide nanoparticles. J Biomed Mater Res Part A 2012, 100A:2554–2562.

    Article  CAS  Google Scholar 

  50. Li B, Ze YG, Sun QQ, Zhang T, Sang XZ, Cui YL, Wang XC, Gui SX, Tan DL, Zhu M, Zhao XY, Sheng L, Wang L, Hong FS, Tang M: Molecular mechanisms of nanosized titanium dioxide–Induced pulmonary injury in mice. Plos One 2013, 8:e55563.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Hu RP, Zheng L, Zhang T, Cui YL, Gao GD, Cheng Z, Chen J, Tang M, Hong FS: Molecular mechanism of hippocampal apoptosis of mice following exposure to titanium dioxide nanoparticles. J Hazard Mater 2011, 191:32–40.

    Article  CAS  PubMed  Google Scholar 

  52. Cheng CY, Mruk DD: Cell junction dynamics in the testis: Sertoli-germ cell interactions and male contraceptive development. Physiol Rev 2002, 82:825–874.

    Article  CAS  PubMed  Google Scholar 

  53. Alnemri ES, Livingston DJ, Nicholson DW, Salvesen G, Thornberry NA, Wong WW, Yuan J: Human ICE/CED-3 protease nomenclature. Cell 1996, 87(2):171.

    Article  CAS  PubMed  Google Scholar 

  54. Salvesen GS: Caspases: opening the boxes and interpreting the arrows. Cell Death Differ 2002, 9(1):3–5.

    Article  PubMed  Google Scholar 

  55. Ghavami S, Hashemi M, Ande SR, Yeganeh B, Xiao W, Eshraghi M, Bus CJ, Kadkhoda K, Wiechec E, Halayko AJ, Los M: Apoptosis and cancer: mutations within caspase genes. J Med Genet 2009, 46(8):497–510.

    Article  CAS  PubMed  Google Scholar 

  56. Walters J, Pop C, Scott FL, Drag M, Swartz P, Mattos C, Salvesen GS, Clark AC: A constitutively active and uninhibitable caspase-3 zymogen efficiently induces apoptosis. Biochem J 2009, 424(3):335–345.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Li N, Duan YM, Hong MM, Zheng L, Fei M, Zhao XY, Wang Y, Cui YL, Liu HT, Cai JW, Gong SJ, Wang H, Hong FS: Spleen injury and apoptotic pathway in mice caused bytitanium dioxide nanoparticules. Toxicol Lett 2010, 195:161–168.

    Article  CAS  PubMed  Google Scholar 

  58. Yang J, Liu XS, Bhalla K, Kim CN, Ibrado AM, Cai J, Peng TI, Jones DP, Wang X: Prevention of apoptosis by Bcl-2: release of cytochrome c from mitochondria blocked. Science 1997, 275:1129–1132.

    Article  CAS  PubMed  Google Scholar 

  59. Howard S, Bottino C, Brooke S, Cheng E, Giffard RG, Sapolsky R: Neuroprotective effects of Bcl-2 overexpression in hippocampal cultures: inter-actions with pathways of oxidative damage. J Neurochem 2002, 83:914–923.

    Article  CAS  PubMed  Google Scholar 

  60. Starkov A, Polster B, Fiskum G: Regulation of hydrogen peroxide production by brain mitochondria by calcium and Bax. J Neurochem 2002, 83:220–228.

    Article  CAS  PubMed  Google Scholar 

  61. Larsson J, Forsberg M, Brännvall K, Zhang XQ, Enarsson M, Hedborg F, Forsberg-Nilsson K: Nuclear receptor binding protein 2 is induced during neural progenitor differentiation and affects cell survival. Mole Cell Neurosci 2008, 39(1):32–39.

    Article  CAS  Google Scholar 

  62. Cao W, Liu NS, Tang S, Bao L, Shen L, Yuan HY, Zhao X, Lu H: Acetyl- Coenzyme A acyltransferase 2 attenuates the apoptotic effects of BNIP3 in two human cell lines. Biochim Biophys Acta 2008, 1780:873–888.

    Article  CAS  PubMed  Google Scholar 

  63. Gingras S, Pelletier S, Boyd K, Ihle JN: Characterization of a family of novel cysteine-serine-rich nuclear proteins (CSRNP). Plos One 2007, 2(8):e808.

    Article  PubMed  PubMed Central  Google Scholar 

  64. Hsieh Y, Guan Y, Tu C, Bratt PJ, Angerhofer JR, Lepock M, Hickey MJ, Tainer JA, Nick HS, Silverman DN: Probing the active site of human manganese superoxide disumtase: the role of glutamine 143. Biochem 1998, 37:4731–4739.

    Article  CAS  Google Scholar 

  65. Pons E, Sipila P, Britan A, Vernet P, Poutaneri M, Huhtaniemi I, Drevet JR: Epididymal expression of mouse GSH-PX proteins: analysis of the mechanisms of GSH-PX5 tissue and region-specific expression through in vitro and in vivo approaches. In Third International Conference on the Epididymis. Edited by Hinton BT, Turner TT. Charlottesville, VA: The Van Doren Company; 2003:74–93.

    Google Scholar 

  66. Liang H, Van Remmen H, Frohlich V, Lechleiter J, Richardson A, Ran Q: GSH-Px4 protects mitochondrial ATP generation against oxidative damage. Biochem Biophys Res Commun 2007, 356(4):893–898.

    Article  CAS  PubMed  Google Scholar 

  67. Imai H, Nakagawa Y: Biological significance of phospholipid hydroperoxide glutathione peroxidase (PHGSH-Px, GSH-Px4) in mammalian cells. Free Radic Biol Med 2003, 34(2):145–169.

    Article  CAS  PubMed  Google Scholar 

  68. Kim G, Lee TH, Wetzel P, Geers C, Robinson MA, Myers TG, Owens JW, Wehr NB, Eckhaus MW, Gros G, Wynshaw-Boris A, Levine RL: Carbonic anhydrase III is not required in the mouse for normal growth, development, and life span. Mol Cell Biol 2004, 24:9942–9947.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Raisanen S, Lehenkari P, Tascorbic Acidnen M, Rahikila P, Harkonen PL, Vaananen HK: Carbonic anhydrase III protects cells from hydrogen peroxide-induced apoptosis. FASEB J 1999, 13:513–522.

    CAS  PubMed  Google Scholar 

  70. Tang YX, Scheef EA, Wang SJ, Sorenson CM, Marcus CB, Jefcoate CR, Sheibani N: CYP1B1 expression promotes the proangiogenic phenotype of endothelium through decreased intracellular oxidative stress and thrombospondin-2 expression. Blood 2009, 113(3):751–754.

    Article  Google Scholar 

  71. Yang P, Lu C, Hua N, Du Y: Titanium dioxide nanoparticles co-doped with Fe 3+ and Eu 3+ ions for photocatalysis. Mater Lett 2002, 57:794–801.

    Article  CAS  Google Scholar 

  72. Sang XZ, Li B, Ze YG, Hong J, Ze X, Gui SX, Sun QQ, Liu HT, Zhao XY, Sheng L, Liu D, Yu XH, Wang L, Hong FS: Toxicological mechanisms of nanosized titanium dioxide-induced spleen injury in mice after repeated peroral application. Agric Food Chem 2013, 61:5590–5599.

    Article  CAS  Google Scholar 

  73. Tarabishy AB, Aldabagh B, Sun Y, Imamura Y, Mukherjee PK, Lass JH, Ghannoum MA, Pearlman E: MyD88 regulation of Fusarium keratitis is dependent on TLR4 and IL-1R1 but not TLR2. J Immunol 2008, 181:593–600.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Zhang WF, He YL, Zhang MS, Yin Z, Chen Q: Raman scattering study on anatase TiO 2 nanocrystals. Phys D: Appl Phys 2000, 33:912–991.

    Article  CAS  Google Scholar 

  75. Bustin SA, Benes V, Garson JA, Hellemans J, Huggett J, Kubista M, Mueller R, Nolan T, Pfaffl MW, Shipley GL, Vandesompele J, Wittwer CT: The MIQE guidelines: minimum information for publication of quantitative real-time PCR experiments. Clin Chem 2009, 55(4):611–622.

    Article  CAS  PubMed  Google Scholar 

  76. Schefe JH, Lehmann KE, Buschmann IR, Unger T, Funke-Kaiser H: Quantitative real-time RT-PCR data analysis: current concepts and the novel “gene expression’s CT difference” formula. J Mol Med 2006, 84:901–910.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by the National Natural Science Foundation of China (grant No. 81273036, 30901218), A Project Funded by the Priority Academic Program Development of Jiangsu Higher Education Institutions, and the Foundation of Cultivation Base of State Key Laboratory of Stem Cell and Biomaterials built together by Ministry of Science and Technology and Jiangsu Province.” To “This work was supported by the National Natural Science Foundation of China (grant No. 81473007, 81273036, 30901218), A Project Funded by the Priority Academic Program Development of Jiangsu Higher Education Institutions, and the Foundation of Cultivation Base of State Key Laboratory of Stem Cell and Biomaterials built together by Ministry of Science and Technology and Jiangsu Province.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Fashui Hong.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

Conceived and designed the experiments: FH, XZ, LS, and LW. Performed the experiments: FH, XZ, LS, LW, JH, and XY. Analyzed the data: FH, XZ, LS, LW, JH, XY, YZ, XS, and QS. Contributed reagents/materials/analysis tools: YZ, XS, and QS. Wrote the paper: FH, XZ, LS, and LW. All authors read and approved the final manuscript.

Xiaoyang Zhao, Lei Sheng and Ling Wang contributed equally.

This article has been retracted by the Editor. A committee at Soochow University has investigated this case and supports the decision to retract the article. Incorrect statistical methods were used to calculate mean and S.D. values and additional errors were made in determining 8-OHdG concentrations. It has come to light that Figure 1 and Figure 2 were published in a previous article. The committee also found that some of the original data were missing. We apologize to the readership of Particle and Fibre Toxicology

A retraction note to this article can be found online at http://dx.doi.org/10.1186/s12989-015-0098-0.

Rights and permissions

Open Access  This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made.

The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.

To view a copy of this licence, visit https://creativecommons.org/licenses/by/4.0/.

The Creative Commons Public Domain Dedication waiver (https://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhao, X., Sheng, L., Wang, L. et al. RETRACTED ARTICLE: Mechanisms of nanosized titanium dioxide-induced testicular oxidative stress and apoptosis in male mice. Part Fibre Toxicol 11, 47 (2014). https://doi.org/10.1186/s12989-014-0047-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12989-014-0047-3

Keywords