Acessibilidade / Reportar erro

Involvement of redox status and the nuclear-related factor 2 in protecting against cadmium-induced renal injury with Sana Makki (Cassia senna L.) pre-treatment in male rats

Abstract

This study was designed to investigate the potential defensive strategy of Sana Makki extract (SME) against Cd-induced in vivo nephrotoxicity and its underlying mechanisms. Male albino rats were used in a thirty days study comparing control, SME-treated, CdCl2-treated, and combined SME and Cd treatment. Pre-treatment with SME significantly reduced serum kidney biomarkers (urea and creatinine), the concentration of renal KIM-1, and kidney index values. Additionally, SME also attenuated CdCl2-induced oxidative and nitrosative stress in renal tissue; significantly reducing malondialdehyde (MDA) and nitric oxide (NO) concentrations and significantly increasing antioxidant enzymes in kidney tissue. Molecularly, SME significantly upregulated antioxidant gene expression (SOD2, GR, GPx1, and CAT) caused by Cd. Notably, the augmented mRNA expression of nuclear-related factor 2 (Nrf2) by Cd was enhanced by SME administration. SME markedly suppressed the Cd-induced rise in pro-inflammatory cytokines. The combination of Cd and SME relieved the Cd-induced apoptotic damage by enhancing Bcl2 and suppressing Bax and Cas-3 levels in renal tissue. The renal tissue histoarchitecture confirmed the biochemical and molecular findings. Collectively, our data indicate that SME can counteract Cd-induced renal intoxication through anti-oxidative, anti-inflammatory, and anti-apoptotic mechanisms.

Key words
Sana Makki; cadmium; apoptosis; oxidative stress; nephrotoxicity

INTRODUCTION

Nephrotoxicity is a major reason for kidney injury and subsequently leads to renal disease, and finally renal failure. Exposure to environmental pollutants, particularly industrial ones, represents a significant public health concern (Almeer et al. 2019ALMEER RS, ALBASHER GI, ALARIFI S, ALKAHTANI S, ALI D & ABDEL MONEIM AE. 2019. Royal jelly attenuates cadmium-induced nephrotoxicity in male mice. Sci Rep 9: 5825.a). Cadmium (Cd) is an extremely toxic industrial metal and is considered the seventh most toxic agent among all known hazardous pollutants (Al Olayan et al. 2020). The main sources for Cd exposure are contaminated food, water, tobacco products, as well as the occupational risks from mining activities, electroplating, and working in the battery industry (Dkhil et al. 2014DKHIL MA, AL-QURAISHY S, DIAB MM, OTHMAN MS, AREF AM & ABDEL MONEIM AE. 2014. The potential protective role of Physalis peruviana L. fruit in cadmium-induced hepatotoxicity and nephrotoxicity. Food Chem Toxicol 74: 98-106.). Chronic Cd exposure causes the accumulation of this metal in proximal renal tubules of the kidney, its primary target organ (Elkhadragy et al. 2018ELKHADRAGY MF, AL-OLAYAN EM, AL-AMIERY AA & ABDEL MONEIM AE. 2018. Protective Effects of Fragaria ananassa Extract Against Cadmium Chloride-Induced Acute Renal Toxicity in Rats. Biol Trace Elem Res, 181, 378-387.). The Cd bound to metallothionein, a cysteine-rich metal-binding protein, is filtered through the glomeruli then presented for tubular reabsorption (Prozialeck et al. 2016PROZIALECK WC, LAMAR PC & EDWARDS JR. 2016. Effects of sub-chronic Cd exposure on levels of copper, selenium, zinc, iron and other essential metals in rat renal cortex. Toxicol Rep 3: 740-746.). The majority of Cd is accumulated in the epithelial cells within the proximal tubules at a level of 450–600 µg/g dry weight and has a slow excretion rate of approximately 1-2 μg per day (Roels et al. 1983ROELS H, LAUWERYS R & DARDENNE A. 1983. The critical level of cadmium in human renal cortex: a reevaluation. Toxicol Lett 15: 357-360., Zhu et al. 2019ZHU L, DUAN P, HU X, WANG Y, CHEN C, WAN J, DAI M, LIANG X, LI J & TAN Y. 2019. Exposure to cadmium and mono‐(2‐ethylhexyl) phthalate induce biochemical changes in rat liver, spleen, lung and kidney as determined by attenuated total reflection-Fourier transform infrared spectroscopy. J Applied Toxicol 39: 783-797.).

The generation of free radicals, reactive oxygen species (ROS), and reactive nitrogen species (RNS), leads to oxidative stress when their presence exceeds the antioxidant capacity of the cells to counterbalance their effects (Azab et al. 2013AZAB SS, ABDEL-DAIM M & ELDAHSHAN OA. 2013. Phytochemical, cytotoxic, hepatoprotective and antioxidant properties of Delonix regia leaves extract. Med Chem Res 22: 4269-4277., Mumtaz et al. 2020MUMTAZ F, ALBELTAGY RS, DIAB MSM, ABDEL MONEIM AE & EL-HABIT OH. 2020. Exposure to arsenite and cadmium induces organotoxicity and miRNAs deregulation in male rats. Environ Sci Pollut Res Int: 10.1007/s11356-020-08306-1.). Almeer et al. (2019a) stated that Cd enhances the production of free radicals as well as suppresses the scavenging system in cells, resulting in oxidative stress, lipid peroxidation, and apoptosis by disturbing mitochondrial function and redox metabolism. Additionally, Cd can induce the inflammatory cascade, with a subsequent rise in interleukins and TNF-α levels. High TNF-α contributes to further renal damage through activation of the pro-inflammatory signaling pathways, vasoconstriction, and cytotoxicity (Mehaffey & Majid 2017MEHAFFEY E & MAJID DS. 2017. Tumor necrosis factor-α, kidney function, and hypertension. Am J Physiol Renal Physiol 313: F1005-F1008.).

A recent focus involves understanding the role of plant-based medicines with antioxidant properties to ameliorate the undesirable effects of drugs and xenobiotics. Sana Makki (Cassia senna L.), a widely used herbal drug with many pharmacological activities, is found in Africa, India, and Asia. All parts of Cassia, including leaves, pods, and fruits are being used pharmacologically (Wang et al. 2020WANG X, WANG T, PAN T, HUANG M, REN W, XU G, AMIN HK, KASSAB RB & ABDEL MONEIM AE. 2020. Senna alexandrina extract supplementation reverses hepatic oxidative, inflammatory, and apoptotic effects of cadmium chloride administration in rats. Environ Sci Pollut Res Int 27: 5981-5992.). The leaves and pods are the main constituents of commercial herbal teas which are used for the treatment of constipation. Moreover, it can be used as a purgative, an antipyretic, or a diuretic (Farag et al. 2015FARAG MA, PORZEL A, MAHROUS EA, MO’MEN M & WESSJOHANN LA. 2015. Integrated comparative metabolite profiling via MS and NMR techniques for Senna drug quality control analysis. Anal Bioanal Chem 407: 1937-1949., Balasankar et al. 2013BALASANKAR D, VANILARASU K, PREETHA P, RAJESWARI S, UMADEVI M & BHOWMIK D. 2013. Senna–A medical miracle plant. J Med Plants Studies 1: 41-47.). Farag et al. (2015)FARAG MA, PORZEL A, MAHROUS EA, MO’MEN M & WESSJOHANN LA. 2015. Integrated comparative metabolite profiling via MS and NMR techniques for Senna drug quality control analysis. Anal Bioanal Chem 407: 1937-1949. investigated the metabolic profiling of Sana Makki extract (SME) and many phytochemical molecules present in the extract have been shown to combat oxidative damage to DNA in vitro by stabilizing the –OH moiety (Lin et al. 2014LIN J, LI X, HAN L, LI F, LU W, BAI Y & CHEN D. 2014. Folium Sennae protects against hydroxyl radical-induced DNA damage via antioxidant mechanism: an in vitro study. Botanical Studies 55: 16.). Thus, the present study aimed to evaluate the toxic effects of cadmium chloride on male renal function and the possible nephroprotective role of SME in albino rats.

MATERIALS AND METHODS

Chemicals and reagents

Cadmium chloride was purchased from Sigma-Aldrich Chemical Co. (St. Louis, MO, USA).

All other reagents used were of high analytical grade.

Plant material

Leaves and pods of Sana Makki were purchased from a local herbal seller in Riyadh, SA. The leaves and pods were identified and authenticated by a taxonomy specialist in the Botany Department at Saud University, Riyadh. After carefully washing and drying the plant material, they were pulverized into fine pieces using an electrical blender. Subsequently, the fine powder was extracted three consecutive times using 70% ethanol at 4°C at a 1:10 (w/v) ratio of plant powder to ethanol. The resulting alcoholic extract was filtered, concentrated in a rotary evaporator, and lyophilized to get rid of any remaining methanol. The final Sana Makki extract was a semi-solid that was stored at 4°C until use.

Animals and experimental protocol

Adult male albino rats (n=40), weighing 200 to 220 g (9-11 weeks old), were obtained from the Biological Department of Faculty of Science, at Princes Nourah bint Abdulrahman University. They were raised in plastic cages under optimal environmental conditions as follows: the temperature was 22-25°C, the humidity was 50-60%, and they were maintained in a 12-hour alternating light/dark cycle. Rats were fed a standard balanced diet with ad libitum access to food and water. All experimental procedures were performed at Princes Nourah bint Abdulrahman University (Riyadh, KSA; approval no, H-01-R-059/19-0168). The experimental rats were acclimatized for one week prior to the experiment and then randomly allocated into four groups, with ten rats per group (n=10) as follows:

Group 1 (control; CTR): the rats were first administered orally 3 ml isotonic NaCl solution kg-1 day-1 and after 2 h they intraperitoneally (i.p.) injected with 1 ml isotonic NaCl solution kg-1 day-1 over 30 successive days.

Group 2 (Sana Makki extract; SME): the rats received 100 mg SME, kg−1 bw day−1 orally by gastric tube for 30 successive days. The selected dose was based on a pilot study conducted to mitigate CdCl2 toxicity. The rats of this group were also received i.p. injection of 1 ml isotonic NaCl solution kg-1 day-1 after 2 h of SME administration.

Group 3 (Cadmium chloride; CdCl2): the rats received CdCl2 (dissolved in an isotonic NaCl solution) at a dose of 3.5 mg kg−1 bw day−1 via i.p. injections for 30 successive days (Kara et al. 2005KARA H, KARATAS F, CANATAN H & SERVI K. 2005. Effects of exogenous metallothionein on acute cadmium toxicity in rats. Biol Trace Elem Res 104: 223.), at the same time 2 h before the CdCl2 injection, the rats were received orally 3 ml isotonic NaCl solution kg-1 day-1.

Group 4 (a mixture of SME and CdCl2): the rats were orally administered 100 mg SME kg−1 bw day−1 two hour before the i.p. injection of CdCl2 at 3.5 mg kg−1 bw day−1 for 30 successive days.

One-day after receiving the last treatment, rats were euthanized with pentobarbital (400 mg/kg bodyweight). Blood samples were collected from the inferior vena cava of each rat, clots were allowed to form, and then samples were centrifuged. Serum was harvested and used in renal functional tests. The kidney was immediately dissected and weighted. Renal tissue was placed in ice-cold 50 mM Tris-HCl buffer (pH 7.4) and homogenates were generated using a tissue homogenizer, producing a 10% (w/v) tissue homogenate. The homogenate was centrifuged at 3,000 × g for 10 min at 4°C and the supernatant was stored at -70°C for biochemical tests.

Kidney functional and injury assays

Kidney functional biomarkers were measured in the serum samples using commercial kits from RANDOX Reagents (USA) for urea (Cat. No. UR3825) and creatinine (Cat. No. CR510) and were performed following the manufacturer’s protocols.

Kidney injury molecule-1 (Kim-1) assay

The concentration of kidney injury molecule-1 (Kim-1) in renal tissue homogenates was assessed using an enzyme-linked immunosorbent assay (ELISA) kit purchased from Abcam (Catalog No. ab119597; Cambridge, UK). The manufacturer’s instructions were followed when performing the test.

Kidney index

The kidney index was estimated using the following formula:

Kidney index=Left kidney weightBodyweight× 100

Renal oxidative stress and antioxidant biomarkers

The activities of enzymatic antioxidant markers, including superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), and glutathione reductase (GR) were assessed using a spectrophotometer according to the methods described by Sun et al. (1988)SUN Y, OBERLEY LW & LI Y. 1988. A simple method for clinical assay of superoxide dismutase. Clin Chem 34: 497-500., Luck (1965)LUCK H. 1965. Catalase. In: Bergmeyer HU (Ed), Methods of enzymatic analysis. New York: Academic Press., Paglia & Valentine (1967)PAGLIA DE & VALENTINE WN. 1967. Studies on the quantitative and qualitative characterization of erythrocyte glutathione peroxidase. J Lab Clin Med 70: 158-169., and Factor et al. (1998)FACTOR VM, KISS A, WOITACH JT, WIRTH PJ & THORGEIRSSON SS. 1998. Disruption of redox homeostasis in the transforming growth factor-alpha/c-myc transgenic mouse model of accelerated hepatocarcinogenesis. J Biol Chem 273: 15846-15853., respectively. Additionally, the levels of non-enzymatic markers such as glutathione (GSH), in addition to the oxidative markers namely, malondialdehyde (MDA), and nitric oxide (NO) were measured based on the methods described by Ellman (1959)ELLMAN GL. 1959. Tissue sulfhydryl groups. Arch Biochem Biophys 82: 70-77., Ohkawa et. al. (1979), and Green et al. (1982)GREEN LC, WAGNER DA, GLOGOWSKI J, SKIPPER PL, WISHNOK JS & TANNENBAUM SR. 1982. Analysis of nitrate, nitrite, and [15N]nitrate in biological fluids. Anal Biochem 126: 131-138., respectively.

Biomarkers for renal inflammation and apoptosis

The levels of the pro-inflammatory cytokines TNF-α (tumor necrosis factor-α) and IL-1β (interleukin-1β) were estimated using ELISA kits (Peprotech, Rocky Hill, NJ, USA) and following the manufacturer’s instructions.

Apoptotic protein markers

The concentrations of Cas-3, Bax, and Bcl-2 were measured. Commercial ELISA kits (Elabscience, Houston, TX, USA) were used for measuring Bax and Bcl-2 according to the manufacturer’s instructions. The caspase-3 concentration was estimated using a colorimetric assay (Sigma-Aldrich, St. Louis, MO, USA).

Gene expression studies

Total RNA was isolated from the renal tissue using TRIzol reagent (Sigma-Aldrich, St. Louis, MO, USA). Reverse transcription of the total RNA was performed immediately using the EasyScript Plus cDNA Synthesis Kit (ABM, Richmond, BC, Canada) to obtain cDNA. Real-time PCR was performed using Power SYBR Green (Life Technologies, CA, USA) on an Applied Biosystems 7500 instrument. The PCR program was as follows: 95°C for 4 minutes, 40 cycles of 95°C for 10 seconds, 60°C for 30 seconds, and 72°C for 10 seconds. The forward and reverse primers used in the PCR reactions were purchased from Sigma-Aldrich (Louis, MO, USA) and the NCBI Primer-BLAST program was used to design the primers. The relative gene expression of SOD2, CAT, GPx1, GR, Bcl2, Bax, Cas3, iNOS, and Nrf2 was determined in comparison with the control. Primer sequences and accession numbers used in the PCR reaction were previously published (Almeer & Abdel Moneim 2018). The housekeeping gene, glyceraldehyde-3-phosphate dehydrogenase (GAPDH), was used as it had fixed concentrations in all treatments.

Histopathological and immunohistochemical examinations

Samples from the kidney tissue were immersed in 10% neutral-buffered formalin for 24 h at 25°C for fixation. The samples were embedded in paraffin and sliced into sections. The tissue sections (5 μm thickness) were deparaffinized and stained with hematoxylin and eosin (H&E) and caspase-3 antibody as described previously (Yuan et al. 2014YUAN G ET AL. 2014. Sub-chronic lead and cadmium co-induce apoptosis protein expression in liver and kidney of rats. Int J Clin Exp Pathol 7: 2905-2914.). Kidney sections were examined under a light microscope (Kontron Electronic Imaging System KS300, Zeiss, Germany) for studying the pathological histoarchitecture at a magnification of 400×.

Statistical analysis

All the obtained data were analyzed using a one-way ANOVA followed by Duncan’s post hoc test. P values less than 0.05 were considered to be statistically significant. The analyzed values were expressed as the mean ± standard deviation (SD).

RESULTS

Bodyweight and clinical signs

Neither mortalities nor abnormal signs were observed in all treated rats. However, Cd-treated rats consumed less food when compared to the controls or SME-treated rats. A non-significant drop in the bodyweight of Cd-treated rats was noticed compared to control rats.

Kidney function, kidney index, and Kim-1 assays

Intraperitoneal injections of rats with Cd for 30 days resulted in a significant elevation (p<0.05) in serum urea and creatinine levels compared to the controls. In addition, the Kim-1 concentration and kidney index were significantly higher (p<0.05) in Cd-treated rats compared to the controls. Pre-treatment with SME before Cd treatment resulted in a significant decline (p<0.05) in all of the above mentioned renal parameters (Figure 1).

Figure 1
The kidney index, and serum level of kidney function markers (urea and creatinine) and kidney injury molecule-1 in adult male rats treated with CdCl2 (3.5 mg/kg) and/or SME (100 mg/kg). Values are expressed as the mean ± SD (n = 10). a p<0.05 vs. the control group; b p<0.05 vs. the CdCl2-treated rats.

Renal oxidative stress parameters

When compared to the control rats, the Cd-injected rats exhibited a significant reduction (p<0.05) in the renal activities of SOD, CAT, GPx and GR, and content of GSH, in addition to a significant elevation (p<0.05) in the MDA and NO content. In contrast, administration of SME shortly before CdCl2 markedly improved the renal oxidative state. It significantly increased the antioxidant parameters and lowered the NO and MDA in renal tissue. Further, oral SME administration alone significantly increased the activity of CAT and the content of GSH only when compared to the control group (Figures 2, 3).

Figure 2
The oxidative indices (malondialdehyde (MDA), nitric oxide, and glutathione) in the renal tissue of adult male rats treated with CdCl2 (3.5 mg/kg) and/or SME (100 mg/kg). Values are presented as mean ± SD (n = 10). a p < 0.05 compared to the control group; b p < 0.05 compared to the CdCl2-alone treatment group.
Figure 3
The activity of superoxide dismutase, catalase, glutathione reductase, and glutathione peroxidase in the kidney of adult male rats treated with CdCl2 (3.5 mg/kg) and/or SME (100 mg/kg). Values are presented as mean ± SD (n = 10). a p < 0.05 compared to the control group; b p < 0.05 compared to CdCl2-alone treatment group.

As shown in Figure 4, quantitative real-time polymerase chain reaction (qRT-PCR) analysis revealed that the fold change in gene expression of renal antioxidant genes showed marked downregulation (p<0.05) in Nrf2, SOD2, CAT, GPx1, and GR mRNA following Cd treatment compared to control rats. Meanwhile, the rats exposed to both Cd and SME exhibited upregulated gene expression of Nrf2, SOD2, CAT, GPx1, and GR compared to the rats injected with Cd only. The treatment with SME alone enhanced gene expression of Nrf2 and SOD2 in renal tissue compared to the control group (Figure 4).

Figure 4
The Nrf2/ARE antioxidant signaling pathway gene expression in the renal tissue of adult male rats treated with CdCl2 (3.5 mg/kg) and/or SME (100 mg/kg). Values are presented as mean ± SD of two assays and normalized to GAPDH and expressed as fold change (log2 scale) relative to mRNA levels in controls; a p < 0.05 compared to the control group; b p < 0.05 compared to the CdCl2-alone treatment group.

Kidney inflammatory assays

Cd-intoxicated rats demonstrated markedly high levels of renal IL-1β and TNF-α compared with control rats. Similarly, iNOS mRNA expression in renal tissue was significantly upregulated after Cd treatment. Contrastingly, pre-treatment with SME significantly reduced levels of both renal inflammatory cytokines and iNOS mRNA expression (Figure 5).

Figure 5
Pro-inflammatory cytokine production (TNF-α and IL-1β) and iNOS mRNA expression in the renal homogenate of adult male rats treated with CdCl2 (3.5 mg/kg) and/or SME (100 mg/kg). Values are presented as mean ± SD (n = 10). a p < 0.05 compared to the control group; b p < 0.05 compared to the CdCl2-alone treatment group. iNOS mRNA expression is presented as mean ± SD of two assays relative to GAPDH in the control group.

Apoptotic protein markers

Cd intoxication was associated with a significant disruption to apoptotic signaling pathways. A significant increase (p<0.05) in renal tissue Bax concentrations was noticed, while Bcl-2 levels declined (p<0.05) in Cd-treated rats compared to the control group. Further, significant increases (p<0.05) in mRNA expression of Bax and Cas3 were detected with a significant downregulation of Bcl2 after Cd injection in respect to the control group. These alterations were notably reversed following SME administration shortly before CdCl2 (Figure 6). Constant with the biochemical and molecular findings, caspase-3 detected with immunohistochical method was upregulated with Cd injection (Figure 7c) and downregulated with SME pretreatment (Figure 7d).

Figure 6
The apoptotic markers (Bcl-2, Bax, and caspase-3) and Bcl2, Bax, and Cas-3 mRNA expression in the renal homogenates. Values are presented as mean ± SD (n = 10). a p < 0.05 compared to the control group; b p < 0.05 compared to the CdCl2-alone treatment group. Cas-3, Bax and Bcl2 mRNA levels are presented as mean ± SD of two assays relative to GAPDH in the control group.
Figure 7
The expression of caspase-3 in the renal tissue of adult male rats treated with CdCl2 (3.5 mg/kg) and/or SME (100 mg/kg). (a) Control group; (b) SME administered group; (c) CdCl2 treatment group; (d) SME+CdCl2 treatment group; scale bar = 100.

Histopathological findings

The control and SME-treated alone groups showed normal morphology with no evidence of histopathological changes in the renal tissue (Figure 8a and 8b). However, the rats intoxicated with CdCl2 showed severe alterations in the renal tubules and glomeruli, including hydropic degeneration (blue star) and hypertrophy of the proximal tubular epithelium (red star), tubular swelling, glomerular shrinkage (black star), and cellular vacuolization. In addition, desquamation and subsequent degeneration of cells and focal necrosis were detected in some of the tubules (Figure 8c). In contrast, SME pre-treatment minimized the changes of CdCl2 in renal tubules and glomeruli (Figure 8d).

Figure 8
The histopathological alterations in the renal tissue of adult male rats treated with CdCl2 (3.5 mg/kg) and/or SME (100 mg/kg). (a) Control group; (b) SME administered group; (c) CdCl2 treatment group; (d) SME+CdCl2 treatment group; scale bar = 100.

DISCUSSION

The kidney is a vital organ responsible for the eradication of metabolic waste products from the body, management of body fluids, and maintenance of electrolyte balance (Sindhu et al. 2015SINDHU G, NISHANTHI E & SHARMILA R. 2015. Nephroprotective effect of vanillic acid against cisplatin induced nephrotoxicity in wistar rats: a biochemical and molecular study. Environ Toxicol Pharmacol 39: 392-404.). Cd is a heavy metal and represents one of the most hazardous pollutants, particularly in industrialized countries. Chronic Cd exposure results in injuries to multiple organs, especially the kidney. Cd can persist in the body for up to 30 years due to its prolonged half-life (Wongmekiat et al. 2018WONGMEKIAT O, PEERAPANYASUT W & KOBROOB A. 2018. Catechin supplementation prevents kidney damage in rats repeatedly exposed to cadmium through mitochondrial protection. Naunyn-Schmiedeberg’s Arch Pharmacol 391: 385-394.YANG SH, LI P, YU LH, LI L, LONG M, LIU MD & HE JB. 2019. Sulforaphane Protect Against Cadmium-Induced Oxidative Damage in mouse Leydigs Cells by Activating Nrf2/ARE Signaling Pathway. Int J Mol Sci 20: 630.). Previous studies demonstrated the negative impact of Cd exposure, including oxidative status, inflammation, mitochondrial damage, and apoptosis in renal tissue (Almeer et al. 2019ALMEER RS, ALBASHER GI, ALARIFI S, ALKAHTANI S, ALI D & ABDEL MONEIM AE. 2019. Royal jelly attenuates cadmium-induced nephrotoxicity in male mice. Sci Rep 9: 5825.b, Rani et al. 2014RANI A, KUMAR A, LAL A & PANT M. 2014. Cellular mechanisms of cadmium-induced toxicity a review. Int J Environ Health Res 24: 378-399., Ansari et al. 2017ANSARI MA, RAISH M, AHMAD A, ALKHARFY KM, AHMAD SF, ATTIA SM, ALSAAD AM & BAKHEET SA. 2017. Sinapic acid ameliorate cadmium-induced nephrotoxicity: in vivo possible involvement of oxidative stress, apoptosis, and inflammation via NF-κB downregulation. Environ Toxicol Pharmacol 51: 100-107., Joardar et al. 2019JOARDAR S, DEWANJEE S, BHOWMICK S, DUA TK, DAS S, SAHA A & DE FEO V. 2019. Rosmarinic Acid Attenuates Cadmium-Induced Nephrotoxicity via Inhibition of Oxidative Stress, Apoptosis, Inflammation and Fibrosis. Int J Mol Sci 20: 2027.).

In the current study, the pre-treatment with SME was tested to determine whether it can protect the renal system from the toxicological insult of Cd. The results showed that Cd injections for 30 days resulted in marked alterations in renal structure and function, with an increase in oxidative stress, inflammation, and apoptosis. Further, the nephroprotective efficacy of SME reversed the renal damage created by Cd injections.

After absorption, Cd exists in the blood in two forms, free Cd and Cd-MT complexes. This complex undergoes glomerular filtration and reabsorption by the proximal tubule via endocytosis. In the epithelial cells of the proximal tubules, the acid protease dissociates the Cd-MT complexes liberating cadmium, which is transported back to the cytosol of the proximal tubule by the aid of Zn2+ and/or Cu2+ transporters (Thevenod 2003THEVENOD F. 2003. Nephrotoxicity and the proximal tubule. Insights from cadmium. Nephron Physiol 93: 87-93.). Previous studies concluded that the majority of the transported Cd was detected in the epithelial lining of the proximal tubules. According to the findings of Almeer et al. (2019b) and Jafarpour et al. (2017)JAFARPOUR D, SHEKARFOROUSH SS, GHAISARI HR, NAZIFI S, SAJEDIANFARD J & ESKANDARI MH. 2017. Protective effects of synbiotic diets of Bacillus coagulans, Lactobacillus plantarum and inulin against acute cadmium toxicity in rats. BMC Complement Altern Med 17: 291., Cd concentrations increased significantly in Cd-injected rats compared to the control. Farag et al. (2015)FARAG MA, PORZEL A, MAHROUS EA, MO’MEN M & WESSJOHANN LA. 2015. Integrated comparative metabolite profiling via MS and NMR techniques for Senna drug quality control analysis. Anal Bioanal Chem 407: 1937-1949. detected the existence of flavonoids, such as kaempferol, quercetin, and isorhamentin, in the 1H-NMR spectrum of Cassia senna. Furthermore, Cherrak et al. (2016)CHERRAK SA, MOKHTARI-SOULIMANE N, BERROUKECHE F, BENSENANE B, CHERBONNEL A, MERZOUK H & ELHABIRI M. 2016. In vitro antioxidant versus metal ion chelating properties of flavonoids: A structure-activity investigation. PLoS One 11: e0165575. stated that such flavonoids can bind with metals in human red blood cells in vitro.

The present study supports the results of previous studies (Poontawee et al. 2016POONTAWEE W, NATAKANKITKUL S & WONGMEKIAT O. 2016. Protective effect of Cleistocalyx nervosum var. paniala fruit extract against oxidative renal damage caused by cadmium. Molecules 21: 133., Wongmekiat et al. 2018WONGMEKIAT O, PEERAPANYASUT W & KOBROOB A. 2018. Catechin supplementation prevents kidney damage in rats repeatedly exposed to cadmium through mitochondrial protection. Naunyn-Schmiedeberg’s Arch Pharmacol 391: 385-394.YANG SH, LI P, YU LH, LI L, LONG M, LIU MD & HE JB. 2019. Sulforaphane Protect Against Cadmium-Induced Oxidative Damage in mouse Leydigs Cells by Activating Nrf2/ARE Signaling Pathway. Int J Mol Sci 20: 630., Sadek et al. 2017SADEK KM, LEBDA MA, ABOUZED TK, NASR SM & SHOUKRY M. 2017. Neuro-and nephrotoxicity of subchronic cadmium chloride exposure and the potential chemoprotective effects of selenium nanoparticles. Metab Brain Dis 32: 1659-1673.), where the Cd injection elevated serum biomarkers of renal malfunction (urea and creatinine) as well as the renal Kim-1 concentrations and kidney index in respect to control. Kim-1 is a transmembrane protein that is not detectable in the healthy kidney. However, with renal injury, it is upregulated and is detected in the urine (Yin et al. 2020YIN M, JIANG N, GUO L, NI Z, AL-BRAKATI AY, OTHMAN MS, ABDEL MONEIM AE & KASSAB RB. 2020. Oleuropein suppresses oxidative, inflammatory, and apoptotic responses following glycerol-induced acute kidney injury in rats. Life Sci 232: 116634.). It was reported that Cd exposure enhances the expression of Kim-1, even at a mild stage of apoptosis in the proximal renal tubule and before the onset of cell death (Prozialeck et al. 2009PROZIALECK WC, EDWARDS JR, LAMAR PC, LIU J, VAIDYA VS & BONVENTRE JV. 2009. Expression of kidney injury molecule-1 (Kim-1) in relation to necrosis and apoptosis during the early stages of Cd-induced proximal tubule injury. Toxicol Applied Pharmacol 238: 306-314.). Moreover, the present renal histopathological findings showed severe damage in renal tissue treated with Cd owing to its disturbing effect on glomerular filtration and tubular reabsorption. Meanwhile, pre-administration with SME markedly augmented the adverse impacts of Cd on renal function and morphology.

Cadmium induced its nephrotoxicity via indirect enhancement of ROS and RNS formation and exhaustion of anti-oxidant defenses (Sanjeev et al. 2019SANJEEV S, BIDANCHI RM, MURTHY MK, GURUSUBRAMANIAN G & ROY VK. 2019. Influence of ferulic acid consumption in ameliorating the cadmium-induced liver and renal oxidative damage in rats. Environ Sci Pollut Res 26(20): 20631-20653.). The marked rise in tissue MDA along with NO concentration were indications of oxidative and nitrosative stress created by Cd. Due to the competition on the binding site, Cd can displace iron resulting in a high content of free redox-active ions. These free redox metals directly boost the generation of hydroxyl radicals via the Fenton reaction (Valko et al. 2016VALKO M, JOMOVA K, RHODES CJ, KUČA K & MUSILEK K. 2016. Redox-and non-redox-metal-induced formation of free radicals and their role in human disease. Arch Toxicol 90: 1-37.). Further, Cd enhances the NADPH oxidase activity resulting in the production of superoxide anion and upregulation of inducible nitric oxide synthase, which synthesizes excess NO and active nitrogen for increased lipid peroxidation (Li et al. 2017LI X, JIANG X, SUN J, ZHU C, LI X, TIAN L, LIU L & BAI W. 2017. Cytoprotective effects of dietary flavonoids against cadmium‐induced toxicity. Ann N Y Acad Sci 1398: 5-19.). In parallel with the high MDA and NO, significant suppression of SOD, CAT, GPx, and GR activities and GSH levels were detected after Cd exposure. Glutathione is the major thiol antioxidant and is considered to be the primary line of defense against Cd toxicity. Cd has a high affinity for thiol-containing compounds. Therefore, GSH is primarily attacked by free Cd-ions (Cuypers et al., 2010). Cd can also substitute the Zn2+ bound to SOD, increasing the free radical output and reducing its antioxidant activity. Catalase is an iron-containing enzyme responsible for hydrogen peroxide (H2O2) detoxification by converting H2O2 into water and divalent oxygen (Cuypers et al. 2010CUYPERS A, PLUSQUIN M, REMANS T, JOZEFCZAK M, KEUNEN E, GIELEN H, OPDENAKKER K, NAIR AR, MUNTERS E & ARTOIS TJ. 2010. Cadmium stress: an oxidative challenge. Biometals 23: 927-940.). The present findings revealed obvious exhaustion of the antioxidant capacity, leading to an overwhelming overproduction of free radicals induced by Cd. These results are in concordance with previous studies (Wongmekiat et al. 2018WONGMEKIAT O, PEERAPANYASUT W & KOBROOB A. 2018. Catechin supplementation prevents kidney damage in rats repeatedly exposed to cadmium through mitochondrial protection. Naunyn-Schmiedeberg’s Arch Pharmacol 391: 385-394.YANG SH, LI P, YU LH, LI L, LONG M, LIU MD & HE JB. 2019. Sulforaphane Protect Against Cadmium-Induced Oxidative Damage in mouse Leydigs Cells by Activating Nrf2/ARE Signaling Pathway. Int J Mol Sci 20: 630., Sadek et al. 2017SADEK KM, LEBDA MA, ABOUZED TK, NASR SM & SHOUKRY M. 2017. Neuro-and nephrotoxicity of subchronic cadmium chloride exposure and the potential chemoprotective effects of selenium nanoparticles. Metab Brain Dis 32: 1659-1673., Li et al. 2017LI X, JIANG X, SUN J, ZHU C, LI X, TIAN L, LIU L & BAI W. 2017. Cytoprotective effects of dietary flavonoids against cadmium‐induced toxicity. Ann N Y Acad Sci 1398: 5-19.).

Additionally, the fold-change in mRNA expression of antioxidant genes were assessed by qRT- PCR in kidney tissue to provide a better understanding regarding the molecular mechanism that regulates various enzymatic activities. Our results illustrated that there was a significant downregulation in mRNA for SOD2, CAT, GPx1, and GR following Cd injections. Nuclear factor erythroid 2-related factor 2 (Nrf2) regulates the gene expression of antioxidant response element (ARE)-dependent genes, which guard cells against oxidative damage (Almeer et al. 2019ALMEER RS, ALBASHER GI, ALARIFI S, ALKAHTANI S, ALI D & ABDEL MONEIM AE. 2019. Royal jelly attenuates cadmium-induced nephrotoxicity in male mice. Sci Rep 9: 5825.b). Nrf2 is a key factor in commencing Nrf2/ARE signaling pathway, one of the major antioxidant pathways. Normally, Nrf2 exists in an inactive form in the cytosol, as it is bound to Kelch-like ECH-associated protein 1 (keap1). During oxidative stress, the Nrf2 transcription factor is activated by its liberation from the Nrf2/keap1 complex. The free Nrf2 then moves to the nucleus and binds to the ARE and activates targets encoding antioxidant enzymes (Rao et al. 2010RAO VA, KLEIN SR, BONAR SJ, ZIELONKA J, MIZUNO N, DICKEY JS, KELLER PW, JOSEPH J, KALYANARAMAN B & SHACTER E. 2010. The antioxidant transcription factor Nrf2 negatively regulates autophagy and growth arrest induced by the anticancer redox agent mitoquinone. J Biol Chem 285: 34447-34459.). In accordance with other authors (Rao et al. 2010RAO VA, KLEIN SR, BONAR SJ, ZIELONKA J, MIZUNO N, DICKEY JS, KELLER PW, JOSEPH J, KALYANARAMAN B & SHACTER E. 2010. The antioxidant transcription factor Nrf2 negatively regulates autophagy and growth arrest induced by the anticancer redox agent mitoquinone. J Biol Chem 285: 34447-34459., Yang et al. 2019), Cd exposure induced activation of the Nrf2/ARE signaling pathway to protect the cellular components against oxidative insults.

Remarkably, the pre-treatment with SME before the Cd injection significantly reduced oxidative and nitrosative stress by enhancing the enzymatic and non-enzymatic antioxidant parameters as well as antioxidant gene expression levels. Flavonoids, the active compounds in SME, act as essential scavengers of free radicals through their ability to stabilize (Cherrak et al. 2016CHERRAK SA, MOKHTARI-SOULIMANE N, BERROUKECHE F, BENSENANE B, CHERBONNEL A, MERZOUK H & ELHABIRI M. 2016. In vitro antioxidant versus metal ion chelating properties of flavonoids: A structure-activity investigation. PLoS One 11: e0165575.). The redox properties of flavonoid phenolic hydroxyl groups can be easily oxidized and lead to activation of detoxifying enzymes such as NAD(P)H-quinone oxidoreductase, GST, or UDP-glucuronosyl transferase, which all fight against cellular oxidative stress (Burda & Oleszek 2001BURDA S & OLESZEK W. 2001. Antioxidant and antiradical activities of flavonoids. J Agric Food Chem 49: 2774-2779., Nijveldt et al. 2001NIJVELDT RJ, VAN NOOD E, VAN HOORN DE, BOELENS PG, VAN NORREN K & VAN LEEUWEN PA. 2001. Flavonoids: a review of probable mechanisms of action and potential applications. Am J Clin Nutr 74: 418-425., Heim et al. 2002HEIM KE, TAGLIAFERRO AR & BOBILYA DJ. 2002. Flavonoid antioxidants: chemistry, metabolism and structure-activity relationships. J Nutr Biochem 13: 572-584.).

Supporting the results of former investigations (Sanjeev et al. 2019SANJEEV S, BIDANCHI RM, MURTHY MK, GURUSUBRAMANIAN G & ROY VK. 2019. Influence of ferulic acid consumption in ameliorating the cadmium-induced liver and renal oxidative damage in rats. Environ Sci Pollut Res 26(20): 20631-20653., Wongmekiat et al. 2018WONGMEKIAT O, PEERAPANYASUT W & KOBROOB A. 2018. Catechin supplementation prevents kidney damage in rats repeatedly exposed to cadmium through mitochondrial protection. Naunyn-Schmiedeberg’s Arch Pharmacol 391: 385-394.YANG SH, LI P, YU LH, LI L, LONG M, LIU MD & HE JB. 2019. Sulforaphane Protect Against Cadmium-Induced Oxidative Damage in mouse Leydigs Cells by Activating Nrf2/ARE Signaling Pathway. Int J Mol Sci 20: 630., Turley et al. 2019TURLEY AE, ZAGORSKI JW, KENNEDY RC, FREEBORN RA, BURSLEY JK, EDWARDS JR & ROCKWELL CE. 2019. Chronic low-level cadmium exposure in rats affects cytokine production by activated T cells. Toxicol Res 8: 227-237., Olszowski et al. 2012OLSZOWSKI T, BARANOWSKA-BOSIACKA I, GUTOWSKA I & CHLUBEK D. 2012. Pro-inflammatory properties of cadmium. Acta Biochim Pol 59: 475-482., Phuagkhaopong et al. 2017PHUAGKHAOPONG S, OSPONDPANT D, KASEMSUK T, SIBMOOH N, SOODVILAI S, POWER C & VIVITHANAPORN P. 2017. Cadmium-induced IL-6 and IL-8 expression and release from astrocytes are mediated by MAPK and NF-κB pathways. Neurotoxicology 60: 82-91.), this study revealed significant increases in the serum pro-inflammatory cytokines and renal iNOS gene expression following Cd treatment. TNF-α is produced by macrophages and considered to self-feed the inflammatory cycle because it can start and control the expression of other pro-inflammatory genes involved its expression (Vazquez Prieto et al. 2015). IL-1β is mainly produced by bone marrow mononuclear cells and is rapidly synthesized and released by inactive tissue as a proactive cytoplasmic precursor (pro-IL-1β) when a pathogen triggers a Toll-like receptor (TLR) or other pattern recognition receptor (Dinarello 2018DINARELLO CA. 2018. Overview of the IL‐1 family in innate inflammation and acquired immunity. Immunol Rev 281: 8-27.). Oxidative events can initiate an inflammatory cascade within the cell via the nuclear factor kappa-activated B cell (NF-κB) pathway, thus, activating pro-inflammatory cytokines. Previous reports showed that the high TNF-α levels elicit renal malfunction by direct cytotoxicity, limiting the blood supply, which triggers the inflammatory cycle and subsequently affects tubular function (Mehaffey & Majid 2017MEHAFFEY E & MAJID DS. 2017. Tumor necrosis factor-α, kidney function, and hypertension. Am J Physiol Renal Physiol 313: F1005-F1008.). Interestingly, SME could significantly reverse the elevation of IL-1β, TNF-α levels, and iNOS gene expression caused by Cd exposure. This may be attributed to the presence of anthraquinone substances that can be isolated from Cassia seeds and possess anti-inflammatory activities through modulation of the NF-κB pathway (Hou et al. 2018HOU J, GU Y, ZHAO S, HUO M, WANG S, ZHANG Y, QIAO Y & LI X. 2018. Anti-Inflammatory Effects of Aurantio-Obtusin from Seed of Cassia obtusifolia L. through Modulation of the NF-κB Pathway. Molecules 23: 3093.).

Apoptosis is a form of programmed cell death which can be induced by various stimuli, including physiologic and pathologic, causing a disruption in tissue homeostasis and affecting organ function. Caspases play a major role in mediating apoptotic death as they regulate the associated cellular alterations. Caspases are activated through two distinct pathways, which include the intrinsic (mitochondrial-dependent) pathway and the extrinsic (death receptor-dependent) pathway (Khafaga & El-Sayed 2018KHAFAGA AF & EL-SAYED YS. 2018. All-trans-retinoic acid ameliorates doxorubicin-induced cardiotoxicity: in vivo potential involvement of oxidative stress, inflammation, and apoptosis via caspase-3 and p53 down-expression. Naunyn-Schmiedeberg’s Arch Pharmacol 391: 59-70.). The equilibrium between B-cell lymphoma-2 (Bcl-2) as an anti-apoptotic protein and Bcl-2-associated X (Bax) as a pro-apoptotic protein is essential for tissue homeostasis. Bcl-2 suppresses the deleterious impacts of ROS by preventing cytochrome-c liberation in the cytoplasm. Additionally, Bcl2 gene expression is increased was concomitantly high glutathione levels (Sadek et al. 2017SADEK KM, LEBDA MA, ABOUZED TK, NASR SM & SHOUKRY M. 2017. Neuro-and nephrotoxicity of subchronic cadmium chloride exposure and the potential chemoprotective effects of selenium nanoparticles. Metab Brain Dis 32: 1659-1673.). Cd can induce apoptosis in tissues and in vitro cell culture (Wongmekiat et al. 2018WONGMEKIAT O, PEERAPANYASUT W & KOBROOB A. 2018. Catechin supplementation prevents kidney damage in rats repeatedly exposed to cadmium through mitochondrial protection. Naunyn-Schmiedeberg’s Arch Pharmacol 391: 385-394.YANG SH, LI P, YU LH, LI L, LONG M, LIU MD & HE JB. 2019. Sulforaphane Protect Against Cadmium-Induced Oxidative Damage in mouse Leydigs Cells by Activating Nrf2/ARE Signaling Pathway. Int J Mol Sci 20: 630., Sadek et al. 2017SADEK KM, LEBDA MA, ABOUZED TK, NASR SM & SHOUKRY M. 2017. Neuro-and nephrotoxicity of subchronic cadmium chloride exposure and the potential chemoprotective effects of selenium nanoparticles. Metab Brain Dis 32: 1659-1673., Teng et al. 2019TENG X, ZHANG W, SONG Y, WANG H, GE M & ZHANG R. 2019. Protective effects of Ganoderma lucidum triterpenoids on oxidative stress and apoptosis in the spleen of chickens induced by cadmium. Environ Sci Pollut Res Int 26(23): 23967-23980., Liu et al. 2016LIU G, ZOU H, LUO T, LONG M, BIAN J, LIU X, GU J, YUAN Y, SONG R & WANG Y. 2016. Caspase-dependent and caspase-independent pathways are involved in cadmium-induced apoptosis in primary rat proximal tubular cell culture. PloS One 11: e0166823.). In accordance with other Cd-intoxication studies, this study revealed that Cd markedly upregulated Bax and Cas-3 and downregulated Bcl2, but after pre-treatment with SME a significant improvement was observed. This is consistent with a study which reported that Cassia species have anti-apoptotic properties by limiting the ROS formation and causing anti-inflammatory effects (Sobeh et al. 2017SOBEH M, MAHMOUD M, HASAN R, CHENG H, EL-SHAZLY A & WINK M. 2017. Senna singueana: Antioxidant, hepatoprotective, antiapoptotic properties and phytochemical profiling of a methanol bark extract. Molecules 22: 1502.).

CONCLUSIONS

Collectively, the findings of the present study explained the nephroprotective efficacy of SME against renal injury elicited by 30-days Cd injections in male rats. This ameliorative effect was mediated by inhibition of lipid peroxidation, enhancement of antioxidant enzymes, and upregulation of genes related to antioxidant and apoptosis mechanisms, minimizing the damage to DNA and other cellular molecules. Therefore, SME can be used as a remedy to improve Cd-induced renal malfunction and toxicity in rats.

ACKNOWLEGMENTS

The authors extend their appreciation to the Deanship of Scientific Research at King Saud University for funding this work through research group No (RG-1439-60).

REFERENCES

  • AL OLAYAN EM, ALOUFI AS, ALAMRI OD, EL-HABIT OH & ABDEL MONEIM AE. 2020. Protocatechuic acid mitigates cadmium-induced neurotoxicity in rats: Role of oxidative stress, inflammation and apoptosis. Sci Total Environ: 10.1016/j.scitotenv.2020.137969.
  • ALMEER RS & ABDEL MONEIM AE. 2018. Evaluation of the Protective Effect of Olive Leaf Extract on Cisplatin-Induced Testicular Damage in Rats. Oxid Med Cell Longev 2018: 11.
  • ALMEER RS, ALBASHER GI, ALARIFI S, ALKAHTANI S, ALI D & ABDEL MONEIM AE. 2019. Royal jelly attenuates cadmium-induced nephrotoxicity in male mice. Sci Rep 9: 5825.
  • ANSARI MA, RAISH M, AHMAD A, ALKHARFY KM, AHMAD SF, ATTIA SM, ALSAAD AM & BAKHEET SA. 2017. Sinapic acid ameliorate cadmium-induced nephrotoxicity: in vivo possible involvement of oxidative stress, apoptosis, and inflammation via NF-κB downregulation. Environ Toxicol Pharmacol 51: 100-107.
  • AZAB SS, ABDEL-DAIM M & ELDAHSHAN OA. 2013. Phytochemical, cytotoxic, hepatoprotective and antioxidant properties of Delonix regia leaves extract. Med Chem Res 22: 4269-4277.
  • BALASANKAR D, VANILARASU K, PREETHA P, RAJESWARI S, UMADEVI M & BHOWMIK D. 2013. Senna–A medical miracle plant. J Med Plants Studies 1: 41-47.
  • BURDA S & OLESZEK W. 2001. Antioxidant and antiradical activities of flavonoids. J Agric Food Chem 49: 2774-2779.
  • CHERRAK SA, MOKHTARI-SOULIMANE N, BERROUKECHE F, BENSENANE B, CHERBONNEL A, MERZOUK H & ELHABIRI M. 2016. In vitro antioxidant versus metal ion chelating properties of flavonoids: A structure-activity investigation. PLoS One 11: e0165575.
  • CUYPERS A, PLUSQUIN M, REMANS T, JOZEFCZAK M, KEUNEN E, GIELEN H, OPDENAKKER K, NAIR AR, MUNTERS E & ARTOIS TJ. 2010. Cadmium stress: an oxidative challenge. Biometals 23: 927-940.
  • DINARELLO CA. 2018. Overview of the IL‐1 family in innate inflammation and acquired immunity. Immunol Rev 281: 8-27.
  • DKHIL MA, AL-QURAISHY S, DIAB MM, OTHMAN MS, AREF AM & ABDEL MONEIM AE. 2014. The potential protective role of Physalis peruviana L. fruit in cadmium-induced hepatotoxicity and nephrotoxicity. Food Chem Toxicol 74: 98-106.
  • ELKHADRAGY MF, AL-OLAYAN EM, AL-AMIERY AA & ABDEL MONEIM AE. 2018. Protective Effects of Fragaria ananassa Extract Against Cadmium Chloride-Induced Acute Renal Toxicity in Rats. Biol Trace Elem Res, 181, 378-387.
  • ELLMAN GL. 1959. Tissue sulfhydryl groups. Arch Biochem Biophys 82: 70-77.
  • FACTOR VM, KISS A, WOITACH JT, WIRTH PJ & THORGEIRSSON SS. 1998. Disruption of redox homeostasis in the transforming growth factor-alpha/c-myc transgenic mouse model of accelerated hepatocarcinogenesis. J Biol Chem 273: 15846-15853.
  • FARAG MA, PORZEL A, MAHROUS EA, MO’MEN M & WESSJOHANN LA. 2015. Integrated comparative metabolite profiling via MS and NMR techniques for Senna drug quality control analysis. Anal Bioanal Chem 407: 1937-1949.
  • GREEN LC, WAGNER DA, GLOGOWSKI J, SKIPPER PL, WISHNOK JS & TANNENBAUM SR. 1982. Analysis of nitrate, nitrite, and [15N]nitrate in biological fluids. Anal Biochem 126: 131-138.
  • HEIM KE, TAGLIAFERRO AR & BOBILYA DJ. 2002. Flavonoid antioxidants: chemistry, metabolism and structure-activity relationships. J Nutr Biochem 13: 572-584.
  • HOU J, GU Y, ZHAO S, HUO M, WANG S, ZHANG Y, QIAO Y & LI X. 2018. Anti-Inflammatory Effects of Aurantio-Obtusin from Seed of Cassia obtusifolia L. through Modulation of the NF-κB Pathway. Molecules 23: 3093.
  • JAFARPOUR D, SHEKARFOROUSH SS, GHAISARI HR, NAZIFI S, SAJEDIANFARD J & ESKANDARI MH. 2017. Protective effects of synbiotic diets of Bacillus coagulans, Lactobacillus plantarum and inulin against acute cadmium toxicity in rats. BMC Complement Altern Med 17: 291.
  • JOARDAR S, DEWANJEE S, BHOWMICK S, DUA TK, DAS S, SAHA A & DE FEO V. 2019. Rosmarinic Acid Attenuates Cadmium-Induced Nephrotoxicity via Inhibition of Oxidative Stress, Apoptosis, Inflammation and Fibrosis. Int J Mol Sci 20: 2027.
  • KARA H, KARATAS F, CANATAN H & SERVI K. 2005. Effects of exogenous metallothionein on acute cadmium toxicity in rats. Biol Trace Elem Res 104: 223.
  • KHAFAGA AF & EL-SAYED YS. 2018. All-trans-retinoic acid ameliorates doxorubicin-induced cardiotoxicity: in vivo potential involvement of oxidative stress, inflammation, and apoptosis via caspase-3 and p53 down-expression. Naunyn-Schmiedeberg’s Arch Pharmacol 391: 59-70.
  • LI X, JIANG X, SUN J, ZHU C, LI X, TIAN L, LIU L & BAI W. 2017. Cytoprotective effects of dietary flavonoids against cadmium‐induced toxicity. Ann N Y Acad Sci 1398: 5-19.
  • LIN J, LI X, HAN L, LI F, LU W, BAI Y & CHEN D. 2014. Folium Sennae protects against hydroxyl radical-induced DNA damage via antioxidant mechanism: an in vitro study. Botanical Studies 55: 16.
  • LIU G, ZOU H, LUO T, LONG M, BIAN J, LIU X, GU J, YUAN Y, SONG R & WANG Y. 2016. Caspase-dependent and caspase-independent pathways are involved in cadmium-induced apoptosis in primary rat proximal tubular cell culture. PloS One 11: e0166823.
  • LUCK H. 1965. Catalase. In: Bergmeyer HU (Ed), Methods of enzymatic analysis. New York: Academic Press.
  • MEHAFFEY E & MAJID DS. 2017. Tumor necrosis factor-α, kidney function, and hypertension. Am J Physiol Renal Physiol 313: F1005-F1008.
  • MUMTAZ F, ALBELTAGY RS, DIAB MSM, ABDEL MONEIM AE & EL-HABIT OH. 2020. Exposure to arsenite and cadmium induces organotoxicity and miRNAs deregulation in male rats. Environ Sci Pollut Res Int: 10.1007/s11356-020-08306-1.
  • NIJVELDT RJ, VAN NOOD E, VAN HOORN DE, BOELENS PG, VAN NORREN K & VAN LEEUWEN PA. 2001. Flavonoids: a review of probable mechanisms of action and potential applications. Am J Clin Nutr 74: 418-425.
  • OHKAWA H, OHISHI N & YAGI K. 1979. Assay for lipid peroxides in animal tissues by thiobarbituric acid reaction. Anal Biochem 95: 351-358.
  • OLSZOWSKI T, BARANOWSKA-BOSIACKA I, GUTOWSKA I & CHLUBEK D. 2012. Pro-inflammatory properties of cadmium. Acta Biochim Pol 59: 475-482.
  • PAGLIA DE & VALENTINE WN. 1967. Studies on the quantitative and qualitative characterization of erythrocyte glutathione peroxidase. J Lab Clin Med 70: 158-169.
  • PHUAGKHAOPONG S, OSPONDPANT D, KASEMSUK T, SIBMOOH N, SOODVILAI S, POWER C & VIVITHANAPORN P. 2017. Cadmium-induced IL-6 and IL-8 expression and release from astrocytes are mediated by MAPK and NF-κB pathways. Neurotoxicology 60: 82-91.
  • POONTAWEE W, NATAKANKITKUL S & WONGMEKIAT O. 2016. Protective effect of Cleistocalyx nervosum var. paniala fruit extract against oxidative renal damage caused by cadmium. Molecules 21: 133.
  • PROZIALECK WC, EDWARDS JR, LAMAR PC, LIU J, VAIDYA VS & BONVENTRE JV. 2009. Expression of kidney injury molecule-1 (Kim-1) in relation to necrosis and apoptosis during the early stages of Cd-induced proximal tubule injury. Toxicol Applied Pharmacol 238: 306-314.
  • PROZIALECK WC, LAMAR PC & EDWARDS JR. 2016. Effects of sub-chronic Cd exposure on levels of copper, selenium, zinc, iron and other essential metals in rat renal cortex. Toxicol Rep 3: 740-746.
  • RANI A, KUMAR A, LAL A & PANT M. 2014. Cellular mechanisms of cadmium-induced toxicity a review. Int J Environ Health Res 24: 378-399.
  • RAO VA, KLEIN SR, BONAR SJ, ZIELONKA J, MIZUNO N, DICKEY JS, KELLER PW, JOSEPH J, KALYANARAMAN B & SHACTER E. 2010. The antioxidant transcription factor Nrf2 negatively regulates autophagy and growth arrest induced by the anticancer redox agent mitoquinone. J Biol Chem 285: 34447-34459.
  • ROELS H, LAUWERYS R & DARDENNE A. 1983. The critical level of cadmium in human renal cortex: a reevaluation. Toxicol Lett 15: 357-360.
  • SADEK KM, LEBDA MA, ABOUZED TK, NASR SM & SHOUKRY M. 2017. Neuro-and nephrotoxicity of subchronic cadmium chloride exposure and the potential chemoprotective effects of selenium nanoparticles. Metab Brain Dis 32: 1659-1673.
  • SANJEEV S, BIDANCHI RM, MURTHY MK, GURUSUBRAMANIAN G & ROY VK. 2019. Influence of ferulic acid consumption in ameliorating the cadmium-induced liver and renal oxidative damage in rats. Environ Sci Pollut Res 26(20): 20631-20653.
  • SINDHU G, NISHANTHI E & SHARMILA R. 2015. Nephroprotective effect of vanillic acid against cisplatin induced nephrotoxicity in wistar rats: a biochemical and molecular study. Environ Toxicol Pharmacol 39: 392-404.
  • SOBEH M, MAHMOUD M, HASAN R, CHENG H, EL-SHAZLY A & WINK M. 2017. Senna singueana: Antioxidant, hepatoprotective, antiapoptotic properties and phytochemical profiling of a methanol bark extract. Molecules 22: 1502.
  • SUN Y, OBERLEY LW & LI Y. 1988. A simple method for clinical assay of superoxide dismutase. Clin Chem 34: 497-500.
  • TENG X, ZHANG W, SONG Y, WANG H, GE M & ZHANG R. 2019. Protective effects of Ganoderma lucidum triterpenoids on oxidative stress and apoptosis in the spleen of chickens induced by cadmium. Environ Sci Pollut Res Int 26(23): 23967-23980.
  • THEVENOD F. 2003. Nephrotoxicity and the proximal tubule. Insights from cadmium. Nephron Physiol 93: 87-93.
  • TURLEY AE, ZAGORSKI JW, KENNEDY RC, FREEBORN RA, BURSLEY JK, EDWARDS JR & ROCKWELL CE. 2019. Chronic low-level cadmium exposure in rats affects cytokine production by activated T cells. Toxicol Res 8: 227-237.
  • VALKO M, JOMOVA K, RHODES CJ, KUČA K & MUSILEK K. 2016. Redox-and non-redox-metal-induced formation of free radicals and their role in human disease. Arch Toxicol 90: 1-37.
  • VAZQUEZ PRIETO MA, BETTAIEB A, RODRIGUEZ LANZI C, SOTO VC, PERDICARO DJ, GALMARINI CR, HAJ FG, MIATELLO RM & OTEIZA PI. 2015. Catechin and quercetin attenuate adipose inflammation in fructose-fed rats and 3T3-L1 adipocytes. Mol Nutr Food Res 59: 622-633.
  • WANG X, WANG T, PAN T, HUANG M, REN W, XU G, AMIN HK, KASSAB RB & ABDEL MONEIM AE. 2020. Senna alexandrina extract supplementation reverses hepatic oxidative, inflammatory, and apoptotic effects of cadmium chloride administration in rats. Environ Sci Pollut Res Int 27: 5981-5992.
  • WONGMEKIAT O, PEERAPANYASUT W & KOBROOB A. 2018. Catechin supplementation prevents kidney damage in rats repeatedly exposed to cadmium through mitochondrial protection. Naunyn-Schmiedeberg’s Arch Pharmacol 391: 385-394.YANG SH, LI P, YU LH, LI L, LONG M, LIU MD & HE JB. 2019. Sulforaphane Protect Against Cadmium-Induced Oxidative Damage in mouse Leydigs Cells by Activating Nrf2/ARE Signaling Pathway. Int J Mol Sci 20: 630.
  • YIN M, JIANG N, GUO L, NI Z, AL-BRAKATI AY, OTHMAN MS, ABDEL MONEIM AE & KASSAB RB. 2020. Oleuropein suppresses oxidative, inflammatory, and apoptotic responses following glycerol-induced acute kidney injury in rats. Life Sci 232: 116634.
  • YUAN G ET AL. 2014. Sub-chronic lead and cadmium co-induce apoptosis protein expression in liver and kidney of rats. Int J Clin Exp Pathol 7: 2905-2914.
  • ZHU L, DUAN P, HU X, WANG Y, CHEN C, WAN J, DAI M, LIANG X, LI J & TAN Y. 2019. Exposure to cadmium and mono‐(2‐ethylhexyl) phthalate induce biochemical changes in rat liver, spleen, lung and kidney as determined by attenuated total reflection-Fourier transform infrared spectroscopy. J Applied Toxicol 39: 783-797.

Publication Dates

  • Publication in this collection
    06 July 2020
  • Date of issue
    2020

History

  • Received
    9 Oct 2019
  • Accepted
    1 Dec 2019
Academia Brasileira de Ciências Rua Anfilófio de Carvalho, 29, 3º andar, 20030-060 Rio de Janeiro RJ Brasil, Tel: +55 21 3907-8100 - Rio de Janeiro - RJ - Brazil
E-mail: aabc@abc.org.br