Effects of Caffeic Acid on Multi-walled Carbon Nanotubes -Induced Mitochondrial Toxicity in Rat Kidney

authors:

avatar Maryam Salehcheh ORCID 1 , avatar Leila Zeidooni ORCID 2 , avatar Mohammad Amin Dehghani 2 , avatar Soheila Alboghobeish 1 , avatar Maryam Shirani ORCID 2 , *

Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
Toxicology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran

how to cite: Salehcheh M, Zeidooni L, Dehghani M A, Alboghobeish S , Shirani M. Effects of Caffeic Acid on Multi-walled Carbon Nanotubes -Induced Mitochondrial Toxicity in Rat Kidney. Jundishapur J Nat Pharm Prod. 2023;18(2):e133171. https://doi.org/10.5812/jjnpp-133171.

Abstract

Multi-walled carbon nanotubes (MWCNTs) are being utilized in various fields. With regard to their numerous applications, MWCNT-induced toxicity has not been extensively investigated. Thus, the present study sheds light on the protective effect of caffeic acid (CA) on mitochondrial toxicity in the kidney caused by MWCNTs in Wistar rats employing the MTT assay as well as reactive oxygen species (ROS) indices, based on measuring glutathione (GSH), mitochondrial membrane potential (MMP), malondialdehyde (MDA), and catalase (CAT) activity. According to the MTT assay, using MWCNTs could significantly diminish mitochondrial viability based on doses. Furthermore, the study findings suggested that MWCNTs could reduce GSH content and CAT activity and subsequently improve mitochondrial ROS formation and damage the mitochondrial membrane of the kidney. The findings also implied that CA could protect renal mitochondria against toxicity induced by MWCNTs by lowering oxidative stress.

1. Background

With one main dimension of less than 100 mm as a minimum, nanomaterials are endowed with novel properties. Their high potential for further applications also characterizes them compared with their original forms (1). Despite numerous uses of such materials, few research studies have focused on their effects on humans and the environment. Accordingly, investigating toxic effects and their mechanisms is of utmost importance since they can generate harmless products (2). Among nanoparticles, carbon nanotubes (CNTs) are approving of interest for their electrical, mechanical, and magnetic properties. In this respect, single-walled carbon nanotubes (SWCNTs) and multi-walled carbon nanotubes (MWCNTs) have been recognized as two forms of nanotubes (3). However, there are few studies concerning their toxicity, whose results are often inconsistent. It has also been reported that such materials can enter the working environment as suspended solids in absorbing quantities and possibly, cause industrial hazards (4). In addition, it has been stated that they can produce CNTs in sizes < 2.5 µm due to the burning of methane and propane of natural gas in ordinary stoves inside and outside houses (4). Studies on the toxic effects of CNTs merely center on the toxicity of these substances on inhalation and epidermal contacts because the greatest possible contacts with nanomaterials are through the skin or the respiratory tract (5, 6). It has been found that CNTs can be absorbed into the circulatory system after entering the lungs and then move into other organs, including the spleen and the kidney, or even the brain (7). It has also been reported that these substances can induce asbestos-like pathogenicity with the risk of carcinogenesis (8). In this regard, Wang et al. confirmed that mitochondrial toxicity could play an important role in apoptosis created by CNTs (9). Mitochondria also have a significant role in oxidation, apoptosis, cell cycle regulation, cell growth, and metabolism (10). Changing the structure of mitochondria can thus induce a significant decrease in cellular metabolism and lead to cell death (11). It is accepted that one of the main causes of damage to the kidney cells is elevated oxidative stress.

2. Objectives

Moreover, studies have shown that CNTs can result in inflammation, apoptosis, and cytotoxicity by inducing oxidative stress, thereby lowering cell viability (12). Nevertheless, no study was found on MWCNT-induced toxicity by oxidative stress in renal mitochondria. Accordingly, this study investigated the effects of MWCNTs on oxidative stress and survival in mitotic mitochondria in rats.

Caffeic acid is a natural phenolic compound in coffee, some fruits, and traditional Chinese medicine (13). This substance and its analogs have a variety of pharmaceutical activities, i.e., anti-oxidant, anti-viral, anti-inflammatory, and anti-cancer properties (14). Therefore, CA was selected as an antioxidant to reduce oxidative stress produced by MWCNTs.

3. Methods

3.1. Materials

MWCNTs (> %95, OD: 5 - 15 nm), CA, and other chemicals, solvents, and reagents (Sigma Chemical Co.) were purchased from Pars Biochemistry Store (Ahvaz, Iran).

3.2. Animals

For the given tests, Wistar male rats (about 200 - 250 g) were received from Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran, and all experimental methods were fulfilled in accordance with ethical protocols introduced by the Animal Ethics Committee at Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran (IR.AJUMS.REC.1396.45).

3.3. Preparation of Nanoparticles

Stoke suspension (600 mg/mL) of MWCNTs was made using distilled water. Three drops of Triton X-100 were added to the sample to describe the particles better. It was then shaken for 20 seconds using a vortex and dispersed using a Sonicator Probe (Ultra Sonic VCX-750 W model) at 4°C and 60 cycles for 30 minutes. The suspension was diluted with distilled water to make the desired concentration and then dispersed again by a Sonicator Probe for 10 minutes (2).

3.4. Size and Morphology of Nanoparticles

3.4.1. Isolation of Mitochondria

The mitochondria of the tissues were isolated by differential centrifugation. First, the kidneys were cleaved into small pieces and then homogenized in mitochondria separation buffer (cold mannitol solution, 225 mM (and subsequently followed for separation of nuclei, unopened cells, and other noncellular tissues by centrifugation at 1500 x g and a temperature of 4°C for 10 minutes. The upper layers were centrifuged at 4°C at 12000 x g for 10 minutes. After that, the upper layer was cast off. The mitochondria were re-suspended in a separation medium (75 mM sucrose, pH 7.4, 0.2 mM EDTA, and 0.225 M D-mannitol) and once again centrifuged for 10 minutes at 12000 x g. Ultimately, the mitochondria were suspended at 4°C in a Tris buffer with 0.05 M Tris-HCl, 0.25 M sucrose, 20 M KCl, 2.0 mM MgCl2, pH 4, and 1.0 mM Na2HPO4. In this study, mitochondria were newly provided for each test. They were then used after 5 hours, except for mitochondria employed in ROS tests and MMP suspended in respiration buffer (10 mM Tris, 0.5 mM MgCl2, 5 mM sodium succinate, 0.32 mM sucrose, 50 µM EGTA, 0.1 mM KH2PO4, 20 mM Mops), and buffer (68 mM D-mannitol, 220 mM sucrose, 10 mM KCl, 2 µM rotenone, 50 µM EGTA, 5 mM KH2PO4, 10 mM HEPES, 5 mM sodium succinate, and 2 mM MgCl2) (15, 16). To ensure high-quality mitochondrial separation, all steps were performed on ice.

3.4.2. Protein Measurements

The mitochondrial protein concentration was established by binding protein molecules to Coomassie brilliant blue and using a BSA as a standard (17). To meet normalization in all the mitochondrial tests, mitochondrial samples were also utilized.

3.5. Dose Assessment of MWCNTs

To select the best dose, the given doses were exposed to mitochondria for one hour in an incubator based on previous reports (2, 18). To this end, the mitochondria were divided into six groups: (1) control (negative control); (2, 3, 4, 5, and 6) MWCNTs (20, 40, 80, 160, and 320 µg/mL). Then, the mitochondrial viability rate was measured using the MTT assay according to the reduction of yellow tetrazole to purple formazone due to mitochondrial respiration in live cells. Moreover, the viability rate was shown as a percentage control, which is 100% assumed. After selecting the effective dose of nanoparticles, the mitochondria were divided into six groups: control group; mitochondria group exposed initially in the environment for 1 hour and then exposed to the CA (80 µM) and subsequently exposed to the environment for 1 hour; groups 3, 4, and 5, exposed to CA (160, 80, 40 and 20 µM) for 1 hour and then exposed to MWCNTs (320 µg/mL) for 1 hour; and group 6, exposed to MWCNTs for 1 hour (320 µg/mL) (19).

3.6. Determination of Complex II Activity Using MTT Assay

The MTT assay is known as a sensitive and reliable indicator of the activity of mitochondrial complex II. In brief; for 1 minute, 1 mL of mitochondrial suspensions of the kidney was centrifuged in 10000 x g. Mitochondria were then suspended in 970 µL of isolation buffer and 500 µL of 0.5 µM MTT solution and incubated at 37°C for 45 minutes. Then, they were dissolved in 800 µL DMSO to dissolve the created purple formazan. The absorbance was also determined at 570 nm through a spectrophotometer (UV-1650 PC, Shimadzu, Japan) (3, 18, 20).

3.7. Mitochondrial ROS Level Assay

Employing a fluorescent DCF-DA probe, the ROS rate in mitochondria was also measured. In sum, isolated mitochondria (protein 0.5 mg/mL) were washed with buffer phosphate (PBS) and incubated for 10 minutes with 1.6 mM DCM-DA at 37°C. Fluorescence was then measured via a fluorescence spectrometry of Perkin Elmer LS-50B (California, USA) at 500 and 520 nm emission and excitation wavelength, respectively (21, 22).

3.8. Measurement of Malondialdehyde Levels

Determining the thiobarbituric acid reactive substances (TBARS), MDA levels of renal mitochondria were assessed. To this end, 500 µL of mitochondrial suspension was added to 1.5 mL of TBARS (10%). Then, 2 mL of TBARS (0.67%) was mixed with the supernatant (1.5 mL) and placed for 15 minutes in a boiling water bath. Next, butanol (2 mL) was added and then centrifuged (4000 x g in 15 minutes) after cooling the samples, and the absorbance was assessed at 535 nm. Using various dilutions of 1, 1, 3, and 3-tetramethoxpropane, the standard MDA curve was prepared (23, 24).

3.9. Measurement of GSH Level

To determine the amount of GSH mitochondria, firstly, 5-sulfosalicylic acid (5%) was added to the samples. To remove the precipitated protein, the samples were centrifuged for 15 minutes at 13000 x g and 4°C, and subsequently, the upper layer was collected. Afterward, 5-Dithiobis (2-nitrobenzoic acid; DTNB) was added to the upper layer, and the total TNB was measured using a spectrophotometer at a wavelength of 412 nm (25).

3.10. Determination of CAT Activity

The CAT activity was investigated using the L.Goth method. For this purpose, 500 µL Tris-HCl 0.05 mM, 1 mL H2O2, and 50 µL of mitochondrial suppression were mixed and incubated for 10 minutes. Then the reaction was blocked by adding 500 mL of 4% ammonium molybdate solution. It should be noted that the absorbance was read at 410 nm, and the result was expressed as protein U/mg (22).

3.11. MMP Measurement

Rhodamine 123 (Rh123), as the mitochondrial absorption of cationic fluorescent dye, was utilized to evaluate the MMP. Rh123 is recognized as a lipophilic cation that can bind to parts of the mitochondrial membrane selectively and with a negative charge. Following the membrane hyperpolarization, the Rh123 enters the mitochondria and reduces fluorescence. The mitochondrial suspension (0.5 mg protein/mL) containing 10 µM Rh123 was reacted in MMP buffer. Then, using a Shimadzu RF-5000U Florescence Spectrophotometer, fluorescence was also examined in λEx = 490 nm and λEm = 535 nm (26, 27).

3.12. Statistical Analyses

Descriptive statistics, i.e., mean ± SD, were used to report the results. The statistical analyses were performed through one-way analysis of variance (ANOVA) using Prism Software (version 8). Using Tukey’s post-hoc test, group differences were also calculated.

4. Results

4.1. Dose Optimization of MWCNTs

To select the best dose of MWCNTs, mitochondria were exposed to various concentrations of MWCNTs (20, 40, 80, 160, and 320 µg/mL) for 1 hour; after that, the viability rate was tested using the MTT assay. The viability of the groups compared with H2O2 50 µM as control was also assumed to be equal to 100%. When mitochondria were exposed to 160 µg/mL of MWCNTs, the reduction in viability was approximately 50% compared with mitochondria treated with 50 µM H2O2 (P < 0.01). Therefore, 160 µg/mL of MWCNTs were used in the next experiments (Figure 1).

Effect of MWCNTs on the mitochondrial complex II after 30 and 60 min. Values are represented as mean ± SD and (n = 6). Difference between the control and other groups is significant at P < 0.01 (*), P < 0.001 (**), and P < 0.0001 (***).
Effect of MWCNTs on the mitochondrial complex II after 30 and 60 min. Values are represented as mean ± SD and (n = 6). Difference between the control and other groups is significant at P < 0.01 (*), P < 0.001 (**), and P < 0.0001 (***).

4.2. Effect of CA on Viability Rate of Mitochondria Exposed to MWCNTs

Cell viability was decreased to about 45% when the mitochondria were exposed to 160 µg/mL of MWCNTs for 1 hour. Then, the effect of different doses of CA on the viability rate was investigated. MTT assay showed that pre-treatment of mitochondria with 80 µM of CA could significantly increase the viability rate (P < 0.0001) (See Figure 2). However, there was no significant change in cell viability compared to the control group following 1-hour exposure to 80 µM CA.

Effect of caffeic acid on cell viability in rat Kidney mitochondria exposed to MWCNTs. Results of treatment groups were compared with control group (** P < 0.01, *** P < 0.001) and MWCNTs 160 (## P < 0.01, ####P < 0.0001). Values are expressed as mean ± SD.
Effect of caffeic acid on cell viability in rat Kidney mitochondria exposed to MWCNTs. Results of treatment groups were compared with control group (** P < 0.01, *** P < 0.001) and MWCNTs 160 (## P < 0.01, ####P < 0.0001). Values are expressed as mean ± SD.

4.3. Effect of CA on Oxidative Stress in MWCNT-Treated Mitochondria

The study findings showed significant ROS production as mitochondria were exposed to 160 mM of MWCNTs (P < 0.05) (See Figure 3). However, pre-treatment with 80 µM of CA could significantly inhibit the production of ROS by MWCNTs (P < 0.0001). Pre-treatment with 80 µM of CA also showed significant changes compared with the control group (P < 0.001).

Effect of caffeic acid on ROS in rat Kidney mitochondria exposed to MWCNTs (dichlorodihydrofluorescein=DCF). Results of treatment groups were compared with the control group (*** P < 0.001, **** P < 0.0001) and MWCNTs 160 (####P < 0.0001).
Effect of caffeic acid on ROS in rat Kidney mitochondria exposed to MWCNTs (dichlorodihydrofluorescein=DCF). Results of treatment groups were compared with the control group (*** P < 0.001, **** P < 0.0001) and MWCNTs 160 (####P < 0.0001).

4.4. Effect of CA on MDA Levels in Mitochondria Exposed to MWCNTs

After incubating mitochondria with 160 µg/mL MWCNTs, MDA level (nmol/mg protein) augmented significantly compared with that in the control group (P < 0.001). Additionally, the 1-hour treatment of mitochondria with 40 µM of CA mitigated MDA level compared to that in the MWCNTs group (See Figure 4).

4.5. Effect of CA on GSH in Mitochondria Exposed to MWCNTs

The results of the GSH evaluation revealed a significant decline in the MWCNTs group compared with that in the control group (P < 0.0001). Additionally, 1-hour mitochondrial treatment with 80 µM of CA could prevent this reduction in GSH levels in mitochondria treated with MWCNTs (P < 0.001) (See Figure 4).

4.6. Effect of CA on CAT Activity in Mitochondria Exposed to MWCNTs

According to the study findings, the activity of CAT in mitochondria (unit/mg protein) declined significantly after adding MWCNTs to the mitochondria compared with that in the control group (P < 0.0001). Moreover, a significant increase was also reported in CAT activity after pre-treatment with 80 µM of CA (P < 0.01) compared with the MWCNTs group (See Figure 4).

4.7. Effect of CA on MMP in Mitochondria Exposed to MWCNTs

The cationic fluorochrome R123 pigment was utilized to assess MMP. In this respect, MWCNTs could significantly lower MMP in mitochondria (P < 0.01) (See Figure 4), but the 1-hour treatment of mitochondria with 80 µM of CA increased MMP compared with MWCNTs (P < 0.05).

Effect of caffeic acid on the mitochondrial MDA, GSH, and catalase activity in rat Kidney mitochondria exposed to MWCNTs. Results of treatment groups were compared with the control group ** P < 0.01, *** P < 0.001, **** P < 0.0001) and MWCNTs 160 (#P < 0.05, ## P < 0.01, ### P < 0.001). Values are expressed as mean ± SD.
Effect of caffeic acid on the mitochondrial MDA, GSH, and catalase activity in rat Kidney mitochondria exposed to MWCNTs. Results of treatment groups were compared with the control group ** P < 0.01, *** P < 0.001, **** P < 0.0001) and MWCNTs 160 (#P < 0.05, ## P < 0.01, ### P < 0.001). Values are expressed as mean ± SD.

5. Discussion

Mitochondria are dynamic organelles with numerous functions of metabolism and homeostasis of the living cells. Moreover, they play a central role in generating cellular energy through oxidative phosphorylation, calcium homeostasis, caspase-dependent apoptosis initiation, and a cellular stress response (28). In this regard, it is required to find mitochondrial accuracy for the special functioning of the oxidative phosphorylation system. Mitochondria have five complexes along with two electron carriers that are combined into the oxidative phosphorylation system to reduce ROS levels (29). To regulate the production of cellular energy, mitochondria are thus able to send signals (mitochondrial ROS (mtROS) and transmembrane potential) to other cell organelles. Accordingly, excessive mtROS production is responsible for cellular pathology that can directly affect bio-molecules (30). CNTs can also cause oxidative stress in cells. As reported in the related literature, CNTs can directly interfere with cell membranes, activate inflammasomes, and also lead to ROS production (31). A number of research have further reported that MWCNTs cause tissue damage via ROS generation based on dose and time and consequently induce caspase three activity (30, 32). It has even been reported that MWCNTs have induced oxidative stress and reduced cell viability (12). In the present study, the protective effects of CA in renal mitochondria in rats against MWCNTs-induced oxidative stress damage were investigated. The present study also reported that exposure of mitochondria to MWCNTs had diminished cell viability based on doses. Then cell viability decreased up to 3 folds at the highest concentration (160 µg/mL). Correlated with ROS formation, there was also an increase in fluorescence in the MWCNTs group. The contemporary treatment of CA and MWCNT similarly boosted viability in a dose-dependent manner and showed its protective effect above 80 µM of CA. In this study, the effects of MWCNTs on the MMP of samples were similarly examined. In this respect, MWCNTs decreased MMP, as cited in the study by Guo et al. demonstrating that MWCNT could lead to a drop in MMP of cells consistent with cellular apoptosis (33). In this study, MMP treated with MWCNTs was lower than that of mitochondria treated with CA.

Additionally, MDA, GSH, and CAT activity were investigated in all groups. The results of this study imply that MWCNTs can significantly change GSH activity and diminish GSH in renal mitochondria. Decreased CAT levels were also observed in the MWCNTs groups, likely because of the high concentration of MWCNTs (160 mg/kg) employed in this study.

Formed within oxidative stress conditions, MDA is a major peroxidation product that can be used as a lipid peroxidation indicator (34). It should be noted that the instability of the plasma membrane originates from active oxygen atoms produced by peroxide (35). ROS detoxification is also of importance for cell viability. If ROS are not detoxificated by antioxidant materials, peroxides are produced by lipid peroxidation, and toxic aldehydes such as MDA can disrupt cell function (36). Nevertheless, significant rising trends were observed in mitochondrial MDA levels in the MWCNTs groups in this study, and MDA content in the MWCNTs was higher than in other groups. Together with the given changes in antioxidant enzyme activity, the MDA results indicated that the highest level of oxidative stress had occurred in the renal mitochondria of the MWCNTs-treated group.

It should be noted that CA is endowed with numerous biological activities, including ROS scavenging, immune stimulation, and lipid peroxidation quenching (37). The study results in this respect showed that antioxidant enzyme activities had significantly amplified in CA-treated groups compared with the MWCNTs group, suggesting the anti-oxidative effect of CA on mitochondria. The major finding of this research is that CA showed a protective effect against MWCNTs-induced mitochondrial toxicity. Using reactive oxygen metabolites, CA can also protect biopolymers and reduce oxidative DNA damage (38). The probable mechanism of this effect is that CA can act by increasing both the nuclear translocation of Nrf2 and the expression of antioxidant enzymes, decreasing oxidative stress (39). Previous research in this domain revealed that CA administration to rats affected with breast cancer had improved macro-molecular structure, including total protein content in the liver tissues, breast, and serums, indicating maintained cell structure and integrity along with modulated nucleic acids. Furthermore, CA mitigates membrane-bound marker enzyme and peroxidation reactions and increases the enzymic responses of the tricarboxylic acid cycle, adenosine triphosphatases, and antioxidants. These results indicate that mitochondria are a good candidate for the study of toxicity associated with high-dose MWCNTs.

5.1. Conclusions

To investigate toxicological effects induced by MWCNTs in rat mitochondria, a number of anti-oxidative defense system parameters were examined. According to the study results, antioxidant enzyme machinery could play an important role in coping with excessive ROS and protecting mitochondria against oxidative damage, even though they were inhibited after exposure to the highest concentrations of MWCNTs. Besides, CA could reduce oxidative stress and improve antioxidant status in mitochondria. There is a need to conduct further research studies to evaluate CA treatment efficiency against the effects of MWCNTs in animals.

References

  • 1.

    Jeevanandam J, Barhoum A, Chan YS, Dufresne A, Danquah MK. Review on nanoparticles and nanostructured materials: history, sources, toxicity and regulations. Beilstein J Nanotechnol. 2018;9:1050-74. [PubMed ID: 29719757]. [PubMed Central ID: PMC5905289]. https://doi.org/10.3762/bjnano.9.98.

  • 2.

    Ahangarpour A, Alboghobeish S, Oroojan AA, Dehghani MA. Mice pancreatic islets protection from oxidative stress induced by single-walled carbon nanotubes through naringin. Hum Exp Toxicol. 2018;37(12):1268-81. [PubMed ID: 29658312]. https://doi.org/10.1177/0960327118769704.

  • 3.

    He H, Pham-Huy LA, Dramou P, Xiao D, Zuo P, Pham-Huy C. Carbon nanotubes: applications in pharmacy and medicine. Biomed Res Int. 2013;2013:578290. [PubMed ID: 24195076]. [PubMed Central ID: PMC3806157]. https://doi.org/10.1155/2013/578290.

  • 4.

    Lam CW, James JT, McCluskey R, Arepalli S, Hunter RL. A review of carbon nanotube toxicity and assessment of potential occupational and environmental health risks. Crit Rev Toxicol. 2006;36(3):189-217. [PubMed ID: 16686422]. https://doi.org/10.1080/10408440600570233.

  • 5.

    Yah CS, Iyuke SE, Simate GS. A review of nanoparticles toxicity and their routes of exposures. Iran J Pharm Sci. 2012;8(1):299-314. [PubMed ID: 22459480].

  • 6.

    Varkouhi AK, Scholte M, Storm G, Haisma HJ. Endosomal escape pathways for delivery of biologicals. J Control Release. 2011;151(3):220-8. [PubMed ID: 21078351]. https://doi.org/10.1016/j.jconrel.2010.11.004.

  • 7.

    Jain S. Toxicity Issues Related to Biomedical Applications of Carbon Nanotubes. Journal of Nanomedicine & Nanotechnology. 2012;3(5). https://doi.org/10.4172/2157-7439.1000140.

  • 8.

    Salih SJ, Ghobadi MZ. Evaluating the cytotoxicity and pathogenicity of multi-walled carbon nanotube through weighted gene co-expression network analysis: a nanotoxicogenomics study. BMC Genom Data. 2022;23(1):12. [PubMed ID: 35176998]. [PubMed Central ID: PMC8851761]. https://doi.org/10.1186/s12863-022-01031-3.

  • 9.

    Wang J, Sun P, Bao Y, Dou B, Song D, Li Y. Vitamin E renders protection to PC12 cells against oxidative damage and apoptosis induced by single-walled carbon nanotubes. Toxicol In Vitro. 2012;26(1):32-41. [PubMed ID: 22020378]. https://doi.org/10.1016/j.tiv.2011.10.004.

  • 10.

    Antico Arciuch VG, Elguero ME, Poderoso JJ, Carreras MC. Mitochondrial regulation of cell cycle and proliferation. Antioxid Redox Signal. 2012;16(10):1150-80. [PubMed ID: 21967640]. [PubMed Central ID: PMC3315176]. https://doi.org/10.1089/ars.2011.4085.

  • 11.

    Murray M, Dyari HR, Allison SE, Rawling T. Lipid analogues as potential drugs for the regulation of mitochondrial cell death. Br J Pharmacol. 2014;171(8):2051-66. [PubMed ID: 24111728]. [PubMed Central ID: PMC3976621]. https://doi.org/10.1111/bph.12417.

  • 12.

    Shvedova AA, Pietroiusti A, Fadeel B, Kagan VE. Mechanisms of carbon nanotube-induced toxicity: focus on oxidative stress. Toxicol Appl Pharmacol. 2012;261(2):121-33. [PubMed ID: 22513272]. [PubMed Central ID: PMC4686133]. https://doi.org/10.1016/j.taap.2012.03.023.

  • 13.

    Staniforth V, Chiu LT, Yang NS. Caffeic acid suppresses UVB radiation-induced expression of interleukin-10 and activation of mitogen-activated protein kinases in mouse. Carcinogenesis. 2006;27(9):1803-11. [PubMed ID: 16524889]. https://doi.org/10.1093/carcin/bgl006.

  • 14.

    Chao PC, Hsu CC, Yin MC. Anti-inflammatory and anti-coagulatory activities of caffeic acid and ellagic acid in cardiac tissue of diabetic mice. Nutr Metab (Lond). 2009;6:33. [PubMed ID: 19678956]. [PubMed Central ID: PMC2736962]. https://doi.org/10.1186/1743-7075-6-33.

  • 15.

    Ahangarpour A, Alboghobeish S, Rezaei M, Khodayar MJ, Oroojan AA, Zainvand M. Evaluation of Diabetogenic Mechanism of High Fat Diet in Combination with Arsenic Exposure in Male Mice. Iran J Pharm Res. 2018;17(1):164-83. [PubMed ID: 29755549]. [PubMed Central ID: PMC5937088].

  • 16.

    Faizi M, Seydi E, Abarghuyi S, Salimi A, Nasoohi S, Pourahmad J. A Search for Mitochondrial Damage in Alzheimer's Disease Using Isolated Rat Brain Mitochondria. Iran J Pharm Res. 2016;15(Suppl):185-95. [PubMed ID: 28228816]. [PubMed Central ID: PMC5242364].

  • 17.

    Bradford MM. A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem. 1976;72:248-54. [PubMed ID: 942051]. https://doi.org/10.1006/abio.1976.9999.

  • 18.

    Ahangarpour A, Oroojan AA, Rezae M, Khodayar MJ, Alboghobeish S, Zeinvand M. Effects of butyric acid and arsenic on isolated pancreatic islets and liver mitochondria of male mouse. Gastroenterol Hepatol Bed Bench. 2017;10(1):44-53. [PubMed ID: 28331564]. [PubMed Central ID: PMC5346824].

  • 19.

    Pourkhalili N, Pournourmohammadi S, Rahimi F, Vosough-Ghanbari S, Baeeri M, Ostad SN, et al. Comparative effects of calcium channel blockers, autonomic nervous system blockers, and free radical scavengers on diazinon-induced hyposecretion of insulin from isolated islets of Langerhans in rats. Arh Hig Rada Toksikol. 2009;60(2):157-64. [PubMed ID: 19581208]. https://doi.org/10.2478/10004-1254-60-2009-1917.

  • 20.

    Zhao Y, Ye L, Liu H, Xia Q, Zhang Y, Yang X, et al. Vanadium compounds induced mitochondria permeability transition pore (PTP) opening related to oxidative stress. J Inorg Biochem. 2010;104(4):371-8. [PubMed ID: 20015552]. https://doi.org/10.1016/j.jinorgbio.2009.11.007.

  • 21.

    Keshtzar E, Khodayar MJ, Javadipour M, Ghaffari MA, Bolduc DL, Rezaei M. Ellagic acid protects against arsenic toxicity in isolated rat mitochondria possibly through the maintaining of complex II. Hum Exp Toxicol. 2016;35(10):1060-72. [PubMed ID: 26628001]. https://doi.org/10.1177/0960327115618247.

  • 22.

    Mahdavinia M, Alizadeh S, Raesi Vanani A, Dehghani MA, Shirani M, Alipour M, et al. Effects of quercetin on bisphenol A-induced mitochondrial toxicity in rat liver. Iran J Basic Med Sci. 2019;22(5):499-505. [PubMed ID: 31217929]. [PubMed Central ID: PMC6556511]. https://doi.org/10.22038/ijbms.2019.32486.7952.

  • 23.

    Colon J, Herrera L, Smith J, Patil S, Komanski C, Kupelian P, et al. Protection from radiation-induced pneumonitis using cerium oxide nanoparticles. Nanomedicine. 2009;5(2):225-31. [PubMed ID: 19285453]. https://doi.org/10.1016/j.nano.2008.10.003.

  • 24.

    Shirani M, Alizadeh S, Mahdavinia M, Dehghani MA. The ameliorative effect of quercetin on bisphenol A-induced toxicity in mitochondria isolated from rats. Environ Sci Pollut Res Int. 2019;26(8):7688-96. [PubMed ID: 30666577]. https://doi.org/10.1007/s11356-018-04119-5.

  • 25.

    Shang S, Yang S, Liu Z, Yang X. Oxidative damage in the kidney and brain of mice induced by different nano-materials. Front Biol. 2015;10(1):91-6. https://doi.org/10.1007/s11515-015-1345-3.

  • 26.

    Ahangarpour A, Zeidooni L, Rezaei M, Alboghobeish S, Samimi A, Oroojan AA. Protective effect of metformin on toxicity of butyric acid and arsenic in isolated liver mitochondria and langerhans islets in male mice: an in vitro study. Iran J Basic Med Sci. 2017;20(12):1297-305. [PubMed ID: 29238463]. [PubMed Central ID: PMC5722988]. https://doi.org/10.22038/IJBMS.2017.9567.

  • 27.

    Baracca A, Sgarbi G, Solaini G, Lenaz G. Rhodamine 123 as a probe of mitochondrial membrane potential: evaluation of proton flux through F(0) during ATP synthesis. Biochim Biophys Acta. 2003;1606(1-3):137-46. [PubMed ID: 14507434]. https://doi.org/10.1016/s0005-2728(03)00110-5.

  • 28.

    Bonora M, Morganti C, Morciano G, Giorgi C, Wieckowski MR, Pinton P. Comprehensive analysis of mitochondrial permeability transition pore activity in living cells using fluorescence-imaging-based techniques. Nat Protoc. 2016;11(6):1067-80. [PubMed ID: 27172167]. https://doi.org/10.1038/nprot.2016.064.

  • 29.

    Nelson G, Wordsworth J, Wang C, Jurk D, Lawless C, Martin-Ruiz C, et al. A senescent cell bystander effect: senescence-induced senescence. Aging Cell. 2012;11(2):345-9. [PubMed ID: 22321662]. [PubMed Central ID: PMC3488292]. https://doi.org/10.1111/j.1474-9726.2012.00795.x.

  • 30.

    Visalli G, Facciola A, Curro M, Lagana P, La Fauci V, Iannazzo D, et al. Mitochondrial Impairment Induced by Sub-Chronic Exposure to Multi-Walled Carbon Nanotubes. Int J Environ Res Public Health. 2019;16(5). [PubMed ID: 30841488]. [PubMed Central ID: PMC6427246]. https://doi.org/10.3390/ijerph16050792.

  • 31.

    Saria R, Mouchet F, Perrault A, Flahaut E, Laplanche C, Boutonnet JC, et al. Short term exposure to multi-walled carbon nanotubes induce oxidative stress and DNA damage in Xenopus laevis tadpoles. Ecotoxicol Environ Saf. 2014;107:22-9. [PubMed ID: 24905693]. https://doi.org/10.1016/j.ecoenv.2014.05.010.

  • 32.

    Alarifi S, Ali D. Mechanisms of Multi-walled Carbon Nanotubes-Induced Oxidative Stress and Genotoxicity in Mouse Fibroblast Cells. Int J Toxicol. 2015;34(3):258-65. [PubMed ID: 25998517]. https://doi.org/10.1177/1091581815584799.

  • 33.

    Guo B, Liao C, Liu X, Yi J. Preliminary study on conjugation of formononetin with multiwalled carbon nanotubes for inducing apoptosis via ROS production in HeLa cells. Drug Des Devel Ther. 2018;12:2815-26. [PubMed ID: 30233144]. [PubMed Central ID: PMC6135071]. https://doi.org/10.2147/DDDT.S169767.

  • 34.

    Wang J, Chen C, Li B, Yu H, Zhao Y, Sun J, et al. Antioxidative function and biodistribution of [Gd@C82(OH)22]n nanoparticles in tumor-bearing mice. Biochem Pharmacol. 2006;71(6):872-81. [PubMed ID: 16436273]. https://doi.org/10.1016/j.bcp.2005.12.001.

  • 35.

    Deng X, Jia G, Wang H, Sun H, Wang X, Yang S, et al. Translocation and fate of multi-walled carbon nanotubes in vivo. Carbon. 2007;45(7):1419-24. https://doi.org/10.1016/j.carbon.2007.03.035.

  • 36.

    Khedr NF. Protective effect of mirtazapine and hesperidin on cyclophosphamide-induced oxidative damage and infertility in rat ovaries. Exp Biol Med (Maywood). 2015;240(12):1682-9. [PubMed ID: 25787947]. [PubMed Central ID: PMC4935352]. https://doi.org/10.1177/1535370215576304.

  • 37.

    Jayanthi R, Subash P. Antioxidant effect of caffeic Acid on oxytetracycline induced lipid peroxidation in albino rats. Indian J Clin Biochem. 2010;25(4):371-5. [PubMed ID: 21966107]. [PubMed Central ID: PMC2994573]. https://doi.org/10.1007/s12291-010-0052-8.

  • 38.

    Li Y, Chen LJ, Jiang F, Yang Y, Wang XX, Zhang Z, et al. Caffeic acid improves cell viability and protects against DNA damage: involvement of reactive oxygen species and extracellular signal-regulated kinase. Braz J Med Biol Res. 2015;48(6):502-8. [PubMed ID: 25831202]. [PubMed Central ID: PMC4470308]. https://doi.org/10.1590/1414-431X20143729.

  • 39.

    Jamali N, Mostafavi-Pour Z, Zal F, Kasraeian M, Poordast T, Ramezani F, et al. Combination Effect of Caffeine and Caffeic Acid Treatment on the Oxidant Status of Ectopic Endometrial Cells Separated from Patients with Endometriosis. Iran J Med Sci. 2019;44(4):315-24. [PubMed ID: 31439975]. [PubMed Central ID: PMC6661517]. https://doi.org/10.30476/IJMS.2019.44970.