156
Views
0
CrossRef citations to date
0
Altmetric
Research Article

Silver nanoparticle-induced cell damage via impaired mtROS-JNK/MnSOD signaling pathway

, , , &
Received 22 Feb 2024, Accepted 28 Apr 2024, Published online: 13 May 2024

Abstract

This study investigated the mechanism of silver nanoparticle (AgNP) cytotoxicity from a mitochondrial perspective. The effect of AgNP on manganese superoxide dismutase (MnSOD), a mitochondrial antioxidant enzyme, against oxidative stress has not been studied in detail. We demonstrated that AgNP decreased MnSOD mRNA level, protein expression, and activity in human Chang liver cells in a time-dependent manner. AgNP induced the production of mitochondrial reactive oxygen species (mtROS), particularly superoxide anion. AgNP was found to increase mitochondrial calcium level and disrupt mitochondrial function, leading to reduced ATP level, succinate dehydrogenase activity, and mitochondrial permeability. AgNP induced cytochrome c release from the mitochondria into the cytoplasm, attenuated the expression of the anti-apoptotic proteins phospho Bcl-2 and Mcl-1, and induced the expression of the pro-apoptotic proteins Bim and Bax. In addition, c-Jun N-terminal kinase (JNK) phosphorylation was significantly increased by AgNP. Treatment with elamipretide (a mitochondria-targeted antioxidant) and SP600125 (a JNK inhibitor) showed the involvement of MnSOD and JNK in these processes. These results indicated that AgNP damaged human Chang liver cells by destroying mitochondrial function through the accumulation of mtROS.

Introduction

Since the discovery that silver nanoparticle (AgNP) products were harmful to human health, strong evidence supported the association between AgNP-mediated reactive oxygen species (ROS), subsequent oxidative stress, and cytotoxicity (Flores-López et al. Citation2019; Tortella et al. Citation2020; Salama et al. Citation2023). Many studies have demonstrated that the mechanism of AgNP-induced cytotoxicity was caused by oxidative stress, DNA damage, and apoptosis (Noga et al. Citation2023; Suthar et al. Citation2023).

Mitochondria are the main oxygen-consuming sites for oxidative phosphorylation to produce ATP, and ROS in mitochondria are produced as byproducts of oxygen (O2) metabolism. Manganese superoxide dismutase (MnSOD) is a major mitochondrial antioxidant enzyme that scavenges ROS to protect mitochondria from oxidative stress and mitochondrial dysfunction (Latham et al. Citation2023). It also maintains the mitochondrial integrity of cells exposed to oxidative stress (Liu et al. Citation2020). MnSOD overexpression prevents oxidative stress-induced cell death and tissue damage (Tavleeva et al. Citation2022).

Mitochondrial ROS (mtROS) production is not only determined by ROS production and disposal rates but is also regulated by several factors, such as mitochondrial membrane potential (ΔΨm), mitochondrial metabolic state, and O2 levels (Mailloux Citation2020). AgNP-induced ROS production is closely related to mitochondria (Zhang et al. Citation2022). Previously, we demonstrated that AgNP caused cytotoxicity by oxidative stress-induced apoptosis and damage to cellular components (Piao et al. Citation2011). In apoptosis, c-Jun N-terminal kinase (JNK) plays an important role in regulating mitochondrial pro- and anti-apoptotic proteins (Xu and Hu Citation2020). When JNK is phosphorylated, signaling is activated, and the activated JNK modulates cell survival or death via altering stress-related protein transcription (Liu et al. Citation2020). Previous studies have reported that MnSOD transcription is regulated via the mtROS-driven activation of JNK signaling (Liu et al. Citation2020; Trombetti et al. Citation2021). Although much research on cytotoxicity of AgNP have already been conducted, research on the mitochondria-related mechanism underlying AgNP-induced cell damage is still insufficient; hence, this study aimed to analyze this further.

Material and methods

Materials

AgNP was provided by Professor Jinhee Choi (Seoul University, Seoul, Republic of Korea) and characterized as previously described (Eom and Choi Citation2010). AgNP with a size smaller than 100 nm was evenly dispersed in the tetrahydrofuran (THF) solution by sonication for 3 h, stirred for 3 h to evaporate the THF, refilled with deionized water, and filtered through a cellulose membrane with a pore size of 100 nm to remove AgNP aggregates. Then, the remaining THF was detected using a nuclear magnetic resonance spectrometer until no free THF was detected. According to our previous study, an AgNP concentration of 4 μg/mL was set as the optimal concentration for this study (Piao et al. Citation2011). The other materials were listed in .

Table 1. Reagent list.

Cell condition

Human Chang liver cells were purchased from the American Type Culture Collection (Rockville, MD, USA) and cultured in RPMI-1640 medium supplemented with 10% fetal bovine serum (Gibco Life Technologies, New York, NY, USA) and 1% antibiotic-antimycotic (Gibco). The experimental conditions were conducted according to .

Table 2. Cell experimental conditions.

MnSOD mRNA

MnSOD mRNA was detected using reverse transcription–polymerase chain reaction (RT-PCR). A total RNA extraction solution was added to the cells, the total RNA was isolated, and cDNA was synthesized. After PCR according to the primers and PCR conditions in and , the amplified products were separated on a 1% agarose gel and imaged using Amersham ImageQuant (GE HealthCare, Chicago, IL, USA).

Table 3. Primers.

Table 4. Polymerase chain reaction (PCR) reaction condition.

Protein expression

For western blot analysis, protein samples (30 µg) were subjected to electrophoresis, nitrocellulose membrane transfer, blocking with 3% bovine serum albumin, and then incubation with the primary antibodies MnSOD, COX4, cytochrome c, Bax, Bim, Mcl-1, phospho Bcl-2, JNK, phospho JNK, and actin. Secondary antibodies were used immunoglobulin-G–horseradish peroxidase secondary antibody conjugates (Pierce, Rockford, IL, USA). Protein bands were visualized with an enhanced chemiluminescence detection reagent (Amersham, Little Chalfont, Buckinghamshire, UK), which was then exposed to X-ray film in the dark. For immunocytochemistry, after fixing the cells with 100% ethanol, permeabilizing with 0.5% Triton X-100, and blocking with 5% fetal bovine serum, the cells were incubated with MnSOD or phospho JNK antibody, followed by incubation with a fluorescein isothiocyanate-coupled secondary antibody. MitoTracker™ Red FM staining was performed and slides were made via mounting on a medium containing 4′,6-diamidino-2-phenylindole (DAPI). Images were acquired using a confocal microscope (FluoView FV1200; Olympus, Tokyo, Japan).

MnSOD activity

The cells were suspended in 10 mM phosphate buffer (pH 7.5) and sonicated twice for 15 sec on ice for lysis. Triton X-100 (1%) was added, and cells were incubated on ice for 10 min, followed by centrifugation at 5000 × g to determine the protein content in the supernatant. After adding potassium cyanide solution, the inhibition rate of epinephrine auto-oxidation was measured at 480 nm, and the quantity of enzyme required for 50% inhibition was defined as one unit of enzyme activity. MnSOD activity was expressed as units/mg protein.

Mitochondrial ROS and superoxide anion levels

Cells were treated as described in . First, mtROS were detected using a spectrofluorometer (Perkin Elmer LS-5B; Perkin Elmer, Waltham, MA, USA) according to the instructions of the mtROS detection kit (Cayman Chemical, Ann Arbor, MI, USA). Next, 20 μM dihydrorhodamine 123 (DHR123), for detection of mtROS, or dihydroethidium (DHE), for detection of superoxide anion, was added to 6-well plates and incubated for 30 min, the supernatant was aspirated, rinsed with phosphate-buffered saline (PBS), trypsinized, and harvested. The fluorescence intensity of mtROS was detected using a flow cytometer (Becton Dickinson, San Jose, CA, USA). Finally, 20 μM DHR123 or DHE was added to each slide well and incubated at 37 °C for another 30 min. After rinsing with PBS, the cells were mounted with a mounting medium (DAKO, Carpinteria, CA, USA). Images were acquired on a confocal microscope (FV1200; Olympus).

Mitochondrial Ca2+ level

Cells were treated as described in . Cells were incubated with the mitochondrial Ca2+ indicator Rhod-2 AM. Cell fluorescence was measured using a flow cytometer (Becton Dickinson) or confocal microscope (FV1200; Olympus) after mounting with a DAPI-containing mounting medium.

Intracellular ATP level

Cells were lysed for 30 min by adding 200 µL lysis buffer (25 mM Tris [pH 7.8], 270 mM sucrose, 1 mM EDTA) on ice and sonicated 3 times for 15 sec. The supernatant was collected by centrifugation at 16 000 × g for 10 min at 4 °C and measured with luciferase/luciferin ATP determination kit (Molecular Probes, Eugene, OR, USA).

Succinate dehydrogenase activity

Cells were treated as described in . 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) solution was added to each well and incubated for 4 h and centrifuged at 800 × g for 5 min; then, the supernatant was aspirated. Formazan crystals were dissolved by adding dimethyl sulfoxide to each well, and the absorbance was read at 540 nm using a microplate reader (Molecular Devices, San Jose, CA, USA).

Mitochondrial permeability pore

Cells were treated as described in . The mitochondrial permeability pore (mtPTP) was measured via flow cytometry or confocal microscopy using an mtPTP assay kit according to the manufacturer’s instructions.

Hoechst 33342 staining

Cells were treated as described in . Hoechst 33342 reagent (20 µM) was added for staining for 10 min, and images were acquired using a fluorescence microscope (Olympus).

Caspase-3 activity

Cells were treated as described in . Cells were tested using the Caspase-Glo®-3/7 assay kit according to the manufacturer’s instructions.

Mitochondria morphology

Cells were treated as described in . First, the cell shape was imaged at 100× magnification through a phase-contrast inverted microscope. Next, the cells were treated with 100 nM MitoTracker™ Red FM, mounted with DAPI-containing mounting medium, and imaged under a confocal microscope.

Statistical analysis

Test results were presented as mean ± standard error. SigmaStat version 3.5 (Systat Software Inc., San Jose, CA, USA) was used to perform analysis of variance and Tukey’s test. A p < 0.05 was considered significant.

Results

AgNP decreased MnSOD mRNA, protein expression, and activity

In human Chang liver cells, AgNP decreased MnSOD mRNA and protein expression in a time-dependent manner 3 h after treatment (). Confocal microscopy images showed that AgNP treatment reduced MnSOD expression after 24 h, but the mitochondrial targeting antioxidant elamipretide (MTP-131) restored it (). Similarly, MnSOD activity showed a significant downward trend beginning at 1 h (). This suggested that AgNP could disrupt the mitochondrial antioxidant system.

Figure 1. AgNP reduced MnSOD mRNA, protein expression, and activity. (A) MnSOD mRNA expression was analyzed using RT-PCR. GAPDH was used as a loading control. (B) MnSOD protein expression was analyzed using western blotting. COX4 was used as a mitochondrial loading control. (C) Confocal microscopic images using MitoTracker™ Red FM and DAPI staining showed the MnSOD expression (green), mitochondrial (red), nucleus locations (blue), and the merged image indicated the mitochondria localization of MnSOD. (D) MnSOD enzyme activity was expressed as units/mg protein. *,**Significantly different from the control group (p < 0.05, p < 0.001).

Figure 1. AgNP reduced MnSOD mRNA, protein expression, and activity. (A) MnSOD mRNA expression was analyzed using RT-PCR. GAPDH was used as a loading control. (B) MnSOD protein expression was analyzed using western blotting. COX4 was used as a mitochondrial loading control. (C) Confocal microscopic images using MitoTracker™ Red FM and DAPI staining showed the MnSOD expression (green), mitochondrial (red), nucleus locations (blue), and the merged image indicated the mitochondria localization of MnSOD. (D) MnSOD enzyme activity was expressed as units/mg protein. *,**Significantly different from the control group (p < 0.05, p < 0.001).

AgNP induced mtROS production including superoxide anion

A decrease in the expression or activity of antioxidant enzymes, which are intracellular antioxidant defense systems, is also associated with the production of excessive ROS (Latham et al. Citation2023). AgNP induced significant mtROS production within 1–12 h, with the maximum amount generated at 3 h (). Antimycin A was used as a positive control for mtROS production. Additionally, when mtROS were detected using flow cytometer and confocal microscope after staining of the cell permeant reagent DHR123, AgNP induced mtROS production, which was significantly inhibited by the mitochondrial targeting antioxidant MTP-131 (). In addition, as a result of measuring using flow cytometer and confocal microscope using DHE, a superoxide anion indicator, AgNP induced high level of superoxide anion, but it was suppressed by SOD (). These results suggested that AgNP induced excessive ROS in mitochondria.

Figure 2. AgNP-induced mitochondrial ROS production, including superoxide anion. (A–C) mtROS generated were detected using (A) mtROS detection kit, (B) flow cytometry, or (C) confocal microscopy after treatment with DHR123. Antimycin a was used as a positive control for mtROS production. MTP-131 is a mitochondrial-targeting antioxidant. (D, E) Superoxide anion was detected using (D) flow cytometry and (E) confocal microscopy after staining with DHE. (B, D) FI indicated the fluorescence intensity. (A) *,**,***Significantly different from the control group (p < 0.05, p < 0.01, p < 0.001). (B) *,**Significantly different from the control group (p < 0.001, p < 0.01), #,##significantly different from the AgNP group (p < 0.001, p < 0.01). (D) *,**Significantly different from the control group (p < 0.01, p < 0.001), #,##significantly different from the AgNP group (p < 0.05, p < 0.01).

Figure 2. AgNP-induced mitochondrial ROS production, including superoxide anion. (A–C) mtROS generated were detected using (A) mtROS detection kit, (B) flow cytometry, or (C) confocal microscopy after treatment with DHR123. Antimycin a was used as a positive control for mtROS production. MTP-131 is a mitochondrial-targeting antioxidant. (D, E) Superoxide anion was detected using (D) flow cytometry and (E) confocal microscopy after staining with DHE. (B, D) FI indicated the fluorescence intensity. (A) *,**,***Significantly different from the control group (p < 0.05, p < 0.01, p < 0.001). (B) *,**Significantly different from the control group (p < 0.001, p < 0.01), #,##significantly different from the AgNP group (p < 0.001, p < 0.01). (D) *,**Significantly different from the control group (p < 0.01, p < 0.001), #,##significantly different from the AgNP group (p < 0.05, p < 0.01).

AgNP increased mitochondrial Ca2+ level

ROS can induce Ca2+ release from the mitochondria, increasing the cytosolic Ca2+ level, which may lead to cell apoptosis (Ding et al. Citation2021). Flow cytometry results showed that the fluorescence intensity of mitochondrial Ca2+ measured using the Rhod-2 AM, a mitochondrial Ca2+ indicator, at 12 h after AgNP treatment was significantly higher than that of the control group (). In addition, confocal microscopy revealed that AgNP treatment significantly increased the red fluorescence intensity of Rhod-2 AM (). These data suggested that mitochondrial Ca2+ level increased due to mtROS generated by AgNP.

Figure 3. AgNP increased the mitochondrial Ca2+ level. (A, B) After Rhod-2 AM staining, the mitochondrial Ca2+ level was determined using (A) flow cytometry and (B) confocal microscopy. (A) FI indicated the fluorescence intensity of Rhod-2 AM. *Significantly different from the control group (p < 0.01). (B) Confocal microscopic images showed the mitochondrial Ca2+ level (red).

Figure 3. AgNP increased the mitochondrial Ca2+ level. (A, B) After Rhod-2 AM staining, the mitochondrial Ca2+ level was determined using (A) flow cytometry and (B) confocal microscopy. (A) FI indicated the fluorescence intensity of Rhod-2 AM. *Significantly different from the control group (p < 0.01). (B) Confocal microscopic images showed the mitochondrial Ca2+ level (red).

AgNP damaged the mitochondrial function

Mitochondrial dysfunction is characterized by ATP depletion, depolarization of mitochondrial action potential, and mitochondrial membrane permeability, and it can be caused by excessive mtROS and Ca2+ level (Matuz-Mares et al. Citation2022). AgNP reduced ATP level in a time-dependent manner (). These data suggested that AgNP impaired mitochondrial function by causing a loss of mitochondrial membrane integrity. Succinate dehydrogenase activity in the mitochondrial respiratory chain was assessed using an MTT assay. Succinate dehydrogenase activity was reduced by AgNP treatment in a time-dependent manner (from 3 to 24 h) compared with that in the control group, and succinate dehydrogenase activity was reduced to 44% at 24 h (). Flow cytometric analysis of mtPTP opening experiment showed that AgNP impaired mitochondrial permeability, whereas MTP-131 ameliorated AgNP-induced mitochondrial dysfunction (). Similar results were observed in images captured using confocal microscopy (). Overall, these results indicated that AgNP damaged mitochondrial function through loss of ATP level, succinate dehydrogenase activity, and permeabilization of mitochondrial membrane via excessive mtROS and elevation of Ca2+ level caused by AgNP.

Figure 4. AgNP damaged mitochondrial function. (A) The ATP content was determined using an ATP determination kit. *Significantly different from the control group (p < 0.001). (B) Succinate dehydrogenase activity was detected using an MTT reagent. *,**Significantly different from the control group (p < 0.01, p < 0.001). (C, D) The mtPTP opening was measured using (C) flow cytometry or (D) confocal microscopy using the mitochondrial PT pore assay kit.

Figure 4. AgNP damaged mitochondrial function. (A) The ATP content was determined using an ATP determination kit. *Significantly different from the control group (p < 0.001). (B) Succinate dehydrogenase activity was detected using an MTT reagent. *,**Significantly different from the control group (p < 0.01, p < 0.001). (C, D) The mtPTP opening was measured using (C) flow cytometry or (D) confocal microscopy using the mitochondrial PT pore assay kit.

AgNP changed the expression of mitochondria-related proteins

Opening of the pore leads to ΔΨm loss, which induces the release of cytochrome c from the mitochondria (Zhao et al. Citation2020). AgNP-induced changes in mitochondrial apoptosis-related proteins were also measured. AgNP induced the release of cytochrome c from the mitochondria into the cytoplasm (). As shown in , AgNP caused a time-dependent decrease in Mcl-1 expression and an increase in Bim and Bax expression. However, phospho Bcl-2 was present until 12 h and then decreased sharply at 24 h. It could be seen that these changes in the expression of apoptotic proteins were closely related to mitochondrial damage.

Figure 5. AgNP changed the expression of mitochondria-related proteins. (A, B) Western blot analysis was performed for the detection of (A) cytochrome c and (B) phospho Bcl-2, Mcl-1, Bim, and Bax proteins. COX4 was used as mitochondrial loading control and actin was used as cytosol or total loading control.

Figure 5. AgNP changed the expression of mitochondria-related proteins. (A, B) Western blot analysis was performed for the detection of (A) cytochrome c and (B) phospho Bcl-2, Mcl-1, Bim, and Bax proteins. COX4 was used as mitochondrial loading control and actin was used as cytosol or total loading control.

AgNP activated JNK signaling

We have previously verified that AgNP induced apoptosis via a caspase-dependent pathway involving the mitochondria (Piao et al. Citation2011). AgNP induced JNK phosphorylation in a time-dependent manner (from 3 to 24 h) (). Treatment with the mitochondrial targeting antioxidant MTP-131 and JNK inhibitor SP600125 confirmed the involvement of AgNP in mitochondrial damage and the JNK signaling in cell damage (). As shown in , cell viability decreased after AgNP treatment and was significantly recovered by treatment with MTP-131 or SP600125. Measurement of apoptotic body formation through Hoechst 33342 was also found to be protected by MTP-131 or SP600125 (). Caspase-3 activity was inhibited by MTP-131 and SP600125 (). These data confirmed that AgNP necessarily underwent JNK signaling.

Figure 6. AgNP activated JNK signaling. (A) Western blot analysis was performed to detect phospho JNK protein. JNK was used as loading control. (B) Phospho JNK was assessed using immunocytochemistry using a phospho JNK antibody and a fluorescein isothiocyanate-conjugated secondary antibody. Confocal microscopic image showed the phospho JNK expression (green), nucleus location (blue), and the merged image indicated the localization of phospho JNK. (C) Cell viability was detected using the MTT reagent. (D) Apoptotic bodies were detected by Hoechst 33342. The arrows indicated apoptotic bodies. (E) Caspase-3 activity was detected using the Caspase-Glo®-3/7 assay kit. (C, E) *Significantly different from the control group (p < 0.001), #significantly different from the AgNP group (p < 0.001).

Figure 6. AgNP activated JNK signaling. (A) Western blot analysis was performed to detect phospho JNK protein. JNK was used as loading control. (B) Phospho JNK was assessed using immunocytochemistry using a phospho JNK antibody and a fluorescein isothiocyanate-conjugated secondary antibody. Confocal microscopic image showed the phospho JNK expression (green), nucleus location (blue), and the merged image indicated the localization of phospho JNK. (C) Cell viability was detected using the MTT reagent. (D) Apoptotic bodies were detected by Hoechst 33342. The arrows indicated apoptotic bodies. (E) Caspase-3 activity was detected using the Caspase-Glo®-3/7 assay kit. (C, E) *Significantly different from the control group (p < 0.001), #significantly different from the AgNP group (p < 0.001).

AgNP changed mitochondria morphology

We observed time-dependent changes in cells after AgNP treatment using phase-contrast microscope. Morphological changes in damaged cells that did not completely match those of the control cells began to appear at 3 h, with clear changes observed after 6 h (). To demonstrate the effect of AgNP on mitochondrial damage, we further assessed the AgNP-induced changes in the mitochondrial morphological distribution via confocal microscopy using MitoTracker™ Red FM and DAPI dyes. In control cells, the mitochondria primarily contained spherical structures concentrated around the nucleus rather than being distributed throughout the cytoplasm; however, in AgNP-treated cells, this shape appeared distorted ().

Figure 7. AgNP changed the mitochondrial morphology. (A) AgNP-induced changes in cells were observed over time using a phase-contrast microscope (magnification 100×). (B) Changes in mitochondria morphology were observed using MitoTracker™ Red FM and DAPI staining. Confocal microscopic image showed the mitochondrial (red), nucleus locations (blue), and the merged image indicated the mitochondria localization.

Figure 7. AgNP changed the mitochondrial morphology. (A) AgNP-induced changes in cells were observed over time using a phase-contrast microscope (magnification 100×). (B) Changes in mitochondria morphology were observed using MitoTracker™ Red FM and DAPI staining. Confocal microscopic image showed the mitochondrial (red), nucleus locations (blue), and the merged image indicated the mitochondria localization.

Discussion

In this study, we investigated that AgNP-induced human Chang liver cytotoxicity was related to reduced MnSOD expression and mitochondrial dysfunction caused by mtROS accumulation. Results showed that mtROS were significantly produced after 1 h of AgNP treatment, which continued until 12 h.

SODs are enzymes that cause the dismutation (or partitioning) of superoxide anion (O2) into ordinary O2 molecules or hydrogen peroxide; three subtypes of SOD have been identified, including copper-zinc SOD, MnSOD, and extracellular SOD. Among these, MnSOD initially scavenges superoxide anion from the mitochondrial matrix, thereby protecting mitochondria and other cellular components from oxidative stress (Palma et al. Citation2020; Liu et al. Citation2022). The physiological importance of MnSOD is highlighted in the finding that, in contrast to other SOD isoforms, MnSOD deficiency causes early neonatal death in gene-knockout mice (Liu et al. Citation2022). AgNP reduced MnSOD level in a time-dependent manner, which proved the downregulation of MnSOD via the accumulation of mtROS.

Mitochondria are ATP synthesis sites and the core of cellular energy metabolism. Ca2+ acts at several levels within organelles, is a key regulator of mitochondrial function, and stimulates ATP synthesis. Increased ROS levels owing to environmental stress increase mitochondrial Ca2+ level, causing mitochondrial dysfunction (Matuz-Mares et al. Citation2022). Our results showed that AgNP caused a significant increase in Ca2+ concentration at 12 h. However, studies have reported that Ca2+ accumulation in the mitochondrial matrix can increase ROS generation, triggering the permeability transition pore and cytochrome c release, thereby inducing apoptosis (Matuz-Mares et al. Citation2022). Therefore, mtROS production and Ca2+ accumulation have a mutually promoting relationship that maintains homeostasis via regulating intracellular antioxidants, such as MnSOD. The loss of ΔΨm causes the release of cytochrome c and activates the apoptotic pathway (Popov Citation2023). In previous study, we also demonstrated that AgNP destroyed ΔΨm (Piao et al. Citation2011).

In this study, we examined the expression of proteins involved in the mitochondria-related apoptotic pathways. The Bcl-2 family proteins, such as Bcl-2, Mcl-1, Bim, and Bax, perform a crucial role in regulating mitochondrial apoptosis by either inhibiting or promoting cell death. The interaction and balancing between Bcl-2 family members modulate the permeability of the outer layer of the mitochondrial membrane, triggering the release of activators of caspase cascade throughout the cytosol and subsequently triggering cell death (Czabotar and Garcia-Saez Citation2023). In general, Bcl-2 phosphorylation is known to effectively inhibit its activity and reduce its ability to prevent apoptosis, but phosphorylation at the serine 70 site increases the activity of Bcl-2 and decreases the apoptosis effect (Green Citation2022). JNK activation has been shown to lead to Bcl-2 phosphorylation and degradation, which activates caspase 9 and follows apoptosis (Xu and Hu Citation2020). After AgNP treatment, JNK began to be phosphorylated as early as 3 h, expression of anti-apoptotic proteins phospho Bcl-2 (serine 70) and Mcl-1 decreased at 24 h, and the expression of pro-apoptotic proteins Bim and Bax increased. AgNP induced the release of cytochrome c from the mitochondria into the cytosol. Mitochondrial injury is followed by depletion of intracellular ATP level. Succinate dehydrogenase, which is present in the mitochondrial respiratory chain of the mitochondrial membrane, is an indicator of mitochondrial membrane integrity (Cao et al. Citation2023). AgNP decreased ATP activity and succinate dehydrogenase levels in a time-dependent manner. Treatment with MTP-131, a mitochondria-targeting antioxidant, or SP600125, a JNK inhibitor, restored mitochondrial permeability, apoptotic bodies, and caspase-3 activity increased by AgNP, showing that the cell damage process of AgNP occurs through the mitochondria antioxidant system and JNK signaling. According to research, low levels of ROS intermediates lead to transient activation of JNK, but high ROS levels can activate the JNK pathway and prolong the activation time of JNK (Zhang et al. Citation2016). Our results showed that since the activation of JNK and the expression of MnSOD were significantly regulated from 3 h of AgNP treatment, it could be inferred that the accumulation of mtROS induced by AgNP activated JNK and reduced MnSOD, leading to cell damage. Regarding the study of the signaling pathway between JNK and MnSOD, mtROS can stimulate the transcription of MnSOD by activating JNK signaling (Liu et al. Citation2020), or MnSOD can effectively inhibit the mtROS-JNK-FOXO1 pathway, ultimately reducing cell death (Zhang et al. Citation2023). However, in our system, the phosphorylation of JNK and the inactivation of MnSOD by excess mtROS occurred almost simultaneously. We believe that either scenario is possible and depends on the degree of antioxidant system imbalance between mtROS and MnSOD. According to our observations, in vitro condition may not reflect the actual behavior or function in the liver and AgNP may have interactions with other tissues in the human body. In vitro assessment may not strongly represent the hazardous condition of AgNP or reflect the toxic effects due to the long-term exposure. Therefore, it is necessary to conduct in vivo assessment and clinical evaluation representing long-term exposure and dose-dependent exposure in further studies.

Conclusion

This study showed that AgNPs induced mitochondrial dysfunction through mtROS-JNK/MnSOD signaling, resulting in human Chang liver cell damage. Therefore, strengthening the antioxidant ability of MnSOD may prevent cell damage caused by AgNPs.

Abbreviations
AgNP=

silver nanoparticle

ΔΨm=

mitochondrial membrane potential

DAPI=

4′,6-diamidino-2-phenylindole

DHE=

dihydroethidium

DHR123=

dihydrorhodamine 123

JNK=

c-Jun N-terminal kinase

MnSOD=

manganese superoxide dismutase

mtPTP=

mitochondrial permeability pore

MTP-131=

elamipretide

mtROS=

mitochondrial reactive oxygen species

RT-PCR=

reverse transcription–polymerase chain reaction

THF=

tetrahydrofuran.

Disclosure statement

No potential conflict of interest was reported by the author(s).

Additional information

Funding

This study was supported by the Basic Science Research Program of the National Research Foundation of Korea (NRF), funded by the Ministry of Education [NRF-2021R1I1A1A01047212, RS-2023-00270936].

References

  • Cao K, Xu J, Cao W, Wang X, Lv W, Zeng M, Zou X, Liu J, Feng Z. 2023. Assembly of mitochondrial succinate dehydrogenase in human health and disease. Free Radic Biol Med. 207:247–259. doi:10.1016/j.freeradbiomed.2023.07.023.
  • Czabotar PE, Garcia-Saez AJ. 2023. Mechanisms of BCL-2 family proteins in mitochondrial apoptosis. Nat Rev Mol Cell Biol. 24(10):732–748. doi:10.1038/s41580-023-00629-4.
  • Ding R, Yin YL, Jiang LH. 2021. Reactive oxygen species-induced TRPM2-mediated Ca2+ signalling in endothelial cells. Antioxidants. 10(5):718. doi:10.3390/antiox10050718.
  • Eom HJ, Choi J. 2010. p38 MAPK activation, DNA damage, cell cycle arrest and apoptosis as mechanisms of toxicity of silver nanoparticles in Jurkat T cells. Environ Sci Technol. 44(21):8337–8342. doi:10.1021/es1020668.
  • Flores-López LZ, Espinoza-Gómez H, Somanathan R. 2019. Silver nanoparticles: electron transfer, reactive oxygen species, oxidative stress, beneficial and toxicological effects. Mini review. J Appl Toxicol. 39(1):16–26. doi:10.1002/jat.3654.
  • Green DR. 2022. The mitochondrial pathway of apoptosis Part II: the BCL-2 protein family. Cold Spring Harb Perspect Biol. 14(6):a041046. doi:10.1101/cshperspect.a041046.
  • Latham CM, Balawender PJ, Thomas NT, Keeble AR, Brightwell CR, Ismaeel A, Wen Y, Fry JL, Sullivan PG, Johnson DL, et al. 2023. Overexpression of manganese superoxide dismutase mitigates ACL injury-induced muscle atrophy, weakness and oxidative damage. Free Radic Biol Med. 212:191–198. doi:10.1016/j.freeradbiomed.
  • Liu M, Sun X, Chen B, Dai R, Xi Z, Xu H. 2022. Insights into manganese superoxide dismutase and human diseases. Int J Mol Sci. 23(24):15893. doi:10.3390/ijms232415893.
  • Liu Z, Xu S, Ji Z, Xu H, Zhao W, Xia Z, Xu R. 2020. Mechanistic study of mtROS-JNK-SOD2 signaling in bupivacaine-induced neuron oxidative stress. Aging. 12(13):13463–13476. doi:10.18632/aging.103447.
  • Mailloux RJ. 2020. An update on mitochondrial reactive oxygen species production. Antioxidants. 9(6):472. doi:10.3390/antiox9060472.
  • Matuz-Mares D, González-Andrade M, Araiza-Villanueva MG, Vilchis-Landeros MM, Vázquez-Meza H. 2022. Mitochondrial calcium: effects of its imbalance in disease. Antioxidants. 11(5):801. doi:10.3390/antiox11050801.
  • Noga M, Milan J, Frydrych A, Jurowski K. 2023. Toxicological aspects, safety assessment, and green toxicology of silver nanoparticles (AgNPs) – critical review: state of the art. Int J Mol Sci. 24(6):5133. doi:10.3390/ijms24065133.
  • Palma FR, He C, Danes JM, Paviani V, Coelho DR, Gantner BN, Bonini MG. 2020. Mitochondrial superoxide dismutase: what the established, the intriguing, and the novel reveal about a key cellular redox switch. Antioxid Redox Signal. 32(10):701–714. doi:10.1089/ars.2019.7962.
  • Piao MJ, Kang KA, Lee IK, Kim HS, Kim S, Choi JY, Choi J, Hyun JW. 2011. Silver nanoparticles induce oxidative cell damage in human liver cells through inhibition of reduced glutathione and induction of mitochondria-involved apoptosis. Toxicol Lett. 201(1):92–100. doi:10.1016/j.toxlet.2010.12.010.
  • Popov LD. 2023. Mitochondria as intracellular signalling organelles. An update. Cell Signal. 109:110794. doi:10.1016/j.cellsig.2023.110794.
  • Salama B, Alzahrani KJ, Alghamdi KS, Al-Amer O, Hassan KE, Elhefny MA, Albarakati AJA, Alharthi F, Althagafi HA, Al Sberi H, et al. 2023. Silver nanoparticles enhance oxidative stress, inflammation, and apoptosis in liver and kidney tissues: potential protective role of thymoquinone. Biol Trace Elem Res. 201(6):2942–2954. doi:10.1007/s12011-022-03399-w.
  • Suthar JK, Vaidya A, Ravindran S. 2023. Toxic implications of silver nanoparticles on the central nervous system: a systematic literature review. J Appl Toxicol. 43(1):4–21. doi:10.1002/jat.4317.
  • Tavleeva MM, Belykh ES, Rybak AV, Rasova EE, Chernykh AA, Ismailov ZB, Velegzhaninov IO. 2022. Effects of antioxidant gene overexpression on stress resistance and malignization in vitro and in vivo: a review. Antioxidants. 11(12):2316. doi:10.3390/antiox11122316.
  • Tortella GR, Rubilar O, Durán N, Diez MC, Martínez M, Parada J, Seabra AB. 2020. Silver nanoparticles: toxicity in model organisms as an overview of its hazard for human health and the environment. J Hazard Mater. 390:121974. doi:10.1016/j.jhazmat.2019.121974.
  • Trombetti S, Cesaro E, Catapano R, Sessa R, Lo Bianco A, Izzo P, Grosso M. 2021. Oxidative stress and ROS-mediated signaling in leukemia: novel promising perspectives to eradicate chemoresistant cells in myeloid leukemia. Int J Mol Sci. 22(5):2470. doi:10.3390/ijms22052470.
  • Xu R, Hu J. 2020. The role of JNK in prostate cancer progression and therapeutic strategies. Biomed Pharmacother. 121:109679. doi:10.1016/j.biopha.2019.109679.
  • Zhang J, Wang F, Yalamarty SSK, Filipczak N, Jin Y, Li X. 2022. Nano silver-induced toxicity and associated mechanisms. Int J Nanomedicine. 17:1851–1864. doi:10.2147/IJN.S355131.
  • Zhang J, Wang X, Vikash V, Ye Q, Wu D, Liu Y, Dong W. 2016. ROS and ROS-mediated cellular signaling. Oxid Med Cell Longev. 2016:4350965–4350918. doi:10.1155/2016/4350965.
  • Zhang X, Zhang D, Li H, Liu Z, Yang Y, Li J, Tang L, Tao J, Liu H, Shen M. 2023. Melatonin-mediated suppression of mtROS-JNK-FOXO1 pathway alleviates hypoxia-induced apoptosis in porcine granulosa cells. Antioxidants. 12(10):1881. doi:10.3390/antiox12101881.
  • Zhao Y, Guo Y, Chen Y, Liu S, Wu N, Jia D. 2020. Curculigoside attenuates myocardial ischemia‑reperfusion injury by inhibiting the opening of the mitochondrial permeability transition pore. Int J Mol Med. 45(5):1514–1524. doi:10.3892/ijmm.2020.4513.