Curcumin alleviates colistin-induced nephrotoxicity and neurotoxicity in rats via attenuation of oxidative stress, inflammation and apoptosis

Curcumin alleviates colistin-induced nephrotoxicity and neurotoxicity in rats via attenuation of oxidative stress, inflammation and apoptosis

Chemico-Biological Interactions 294 (2018) 56–64 Contents lists available at ScienceDirect Chemico-Biological Interactions journal homepage: www.els...

4MB Sizes 0 Downloads 607 Views

Chemico-Biological Interactions 294 (2018) 56–64

Contents lists available at ScienceDirect

Chemico-Biological Interactions journal homepage: www.elsevier.com/locate/chembioint

Curcumin alleviates colistin-induced nephrotoxicity and neurotoxicity in rats via attenuation of oxidative stress, inflammation and apoptosis

T

Nagah E. Edreesa, Azza A.A. Galala,∗, Aliaa R. Abdel Monaema, Rasha R. Beheiryb, Mohamed M.M. Metwallyc a

Department of Pharmacology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44511, Egypt Department of Histology and Cytology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44511, Egypt c Department of Pathology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44511, Egypt b

A R T I C LE I N FO

A B S T R A C T

Keywords: Colistin Curcumin Oxidative stress Pro-inflammatory cytokines Immunohistochemistry

Colistin is an effective antibiotic against multidrug-resistant (MDR) gram-negative bacterial infections; however, nephrotoxic and neurotoxic effects are fundamental dose-limiting factors for this treatment. This study was conducted to assess the potential protective effects of curcumin, a phenolic constituent of turmeric, against colistin-induced nephrotoxicity and neurotoxicity, and the possible mechanisms underlying any effect. Twentyfour adult male albino rats were randomly classified into 4 equal groups; the control group (orally received saline solution), the curcumin-treated group (orally administered 200 mg curcumin/kg/day), the colistin-treated group (IP administered 300,000 IU colistin/kg/day) and the concurrent group (orally received 200 mg curcumin/kg/day concurrently with colistin injection); all rats were treated for 6 successive days. Colistin administration significantly increased serum creatinine, urea and uric acid levels as well as brain gamma butyric acid (GABA) concentrations. In renal and brain tissues, colistin significantly increased malondialdehyde (MDA), nitric oxide (NO), tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and caspase-3 expression levels. In addition, colistin significantly decreased catalase (CAT), glutathione (GSH), and B-cell lymphoma 2 (Bcl-2) expressions. Curcumin administration in colistin-treated rats partially restored each of these altered biochemical, antioxidant, inflammatory and apoptotic markers. Histopathological changes in renal and brain tissues were also alleviated by curcumin co-treatment. Our study reveals a critical role of oxidative damage, inflammation and apoptosis in colistin-induced nephrotoxicity and neurotoxicity and showed that they were markedly ameliorated by curcumin co-administration. Therefore, curcumin could represent a promising agent for prevention of colistin-induced nephrotoxicity and neurotoxicity.

1. Introduction Colistin, also known as polymyxin E, is a glycopeptide antibiotic produced by Bacillus polymixa var colistinus [1]. It was discovered by Koyama in 1947, and since 1959, it has been utilized in the treatment of infections caused by multidrug-resistant (MDR) gram-negative bacteria [2,3]. Colistin induces bactericidal effects via the interaction of its cationic polypeptides with the anionic lipopolysaccharide (LPS) molecule of the gram-negative bacterial membrane, leading to displacement of Ca2+ and Mg+ of LPS; these changes in turn disturb membrane stability and increase membrane permeability, causing leakage of cell contents and ultimately cell death. It also binds to the endotoxin of gram-negative bacteria, the lipid A portion of the LPS molecule, and neutralizes it [1,2,4,5].



Colistin was efficacious and had good results in the treatment of infections caused by Escherichia coli, Pseudomonas aeruginosa, Klebsiella pneumonia, Acinetobacter baumannii, Salmonella sp., Enterobacter sp., Haemophilus influenza, and Shigella sp. The resistance of these bacteria against the drug was extremely low. In 1970, it was reported that colistin had nephrotoxic and neurotoxic side effects; thus, its use was temporarily stopped [6,7]. Renal toxicity is the most common side effect associated with colistin administration, because colistin is excreted primarily via the kidneys, and elevated blood levels may deteriorate renal function (Lewis and Lewis, 2004). Furthermore, neurological symptoms, such as confusion, dizziness, vertigo, seizures, and facial/ peripheral paresthesia, and less common fatal effects, including respiratory muscle weakness, apnea, and ataxia, were recorded in colistin-treated patients [8,9]. Clearly, identification of nephroprotective

Corresponding author. E-mail addresses: [email protected], [email protected] (A.A.A. Galal).

https://doi.org/10.1016/j.cbi.2018.08.012 Received 30 May 2018; Received in revised form 13 July 2018; Accepted 15 August 2018 Available online 20 August 2018 0009-2797/ © 2018 Elsevier B.V. All rights reserved.

Chemico-Biological Interactions 294 (2018) 56–64

N.E. Edrees et al.

described protocols [33–35]. Gamma amino butyric acid (GABA) concentration was determined in brain tissue homogenate using a specific rat ELISA kit in accordance with the manufacturer's protocol.

and neuroprotective agents that can be co-administered with colistin has the potential to allow the clinical application of this essential drug. A potential nephroprotective and neuroprotective candidate is curcumin, a compound found within the bright yellow spice turmeric, obtained from the rhizome of Curcuma longa Linn [10]. It has an outstanding safety profile and a number of pleiotropic actions, including anti-inflammatory [11–13], antioxidant and radical scavenging [14], cytotoxic and anti-apoptotic activities [15–17]. Additionally, it has hepatoprotective [18–20] and nephroprotective effects [21–23]. Notably, curcumin can cross the blood-brain barrier, suggesting a possible usage as a neuroprotective substance [24–27]. The spread of infections caused by MDR gram-negative bacteria and the lack of new antibiotics to fight them have led to a revival in colistin use [1]. Therefore, there is an urgent need to alleviate colistin-induced nephrotoxicity and neurotoxicity, as this would increase the therapeutic index of colistin, and thereby permit the administration of higher doses. Natural ingredients have been used to ameliorate the side effects of colistin [28–30]. Thus, the main aim of this work was to assess the mechanism by which colistin induces nephrotoxic and neurotoxic effects, and to determine whether curcumin can protect against colistin side effects via its antioxidant, anti-inflammatory and anti-apoptotic properties.

2.4. Evaluation of oxidative stress markers The brain and both kidneys were removed immediately after sacrifice and washed in physiological saline. One kidney and half of the brain were preserved at −80 °C until preparation of tissue homogenates, which were used for colorimetric assessment of glutathione (GSH), malondialdehyde (MDA) and catalase (CAT) levels using a CE1020 spectrophotometer [36–38]. 2.5. Evaluation of inflammatory markers Levels of tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) were measured from brain and kidney homogenates using a specific rat ELISA kit following the manufacturer's manual. Additionally, nitric oxide (NO) level was colorimetrically evaluated [39]. 2.6. Histopathological and immunohistochemical investigations The other kidney and the second half of the brain were fixed in 10% neutral buffered formalin solution immediately after sacrifice. Paraffin sections of 5–7 μm thickness were cut and stained with hematoxylin and eosin (H&E) and then examined microscopically [40]. Another group of embedded-paraffin sections was also prepared for immunodetection of caspase-3 using a rabbit polyclonal antibody (cat no: RB-1197-R7 Thermo Fisher Scientific, Waltham, MA, USA) and Bcl-2-positive cells using a mouse monoclonal antibody (cat no: MS-123-R7, ready-to-use Neomarkers, Thermo Fisher Scientific, Waltham, MA, USA) using an avidin-biotin-peroxidase (ABC) method [41,42]. Negative control sections were prepared by incubating with phosphate buffer saline (PBS) as an alternative to the primary antibodies. All stained sections were examined with a standard light microscope, and photographs were taken using AmScope Digital Imaging System. The reported histopathological lesions in the kidneys, and cerebral and cerebellar cortices in all groups were scored according to the following scoring system: (−) absence of the lesion in all animals of the group, (+) the lesion was rare within the group, (++) the lesion not so often observed in all animals of the group, (+++), the lesion observed in almost all animals of the group, (++++) the lesion often found in all animals of the group. Quantitative assessment of the Bcl-2 and caspase-3 expression in the kidneys, cerebral and cerebellar cortices was calculated based on the percentage of positive cells per five non-overlapping randomly selected high-power microscopic fields (400X)/section as following: 0 (negative to weak) = less than 10%, + (mild) = 10–25%, ++ (moderate) = 26–50%, +++ (strong) = 51–75%, and ++++ (severe) = more than 75% (severe).

2. Materials and methods Colistin (Colomycin®) vials were produced by Forest Laboratories UK, Ltd. Each vial contains 1 million IU colistimethate sodium powder for injection. Crystalline 99% extra pure curcumin was purchased from Loba Chemie Pvt Ltd-India. Colistin and curcumin were dissolved in sterile normal saline solution. 2.1. Experimental animals Experiments were conducted on 24 adult male albino rats weighing 150–180 g obtained from the animal house at the Faculty of Veterinary Medicine, Zagazig University. The rats were housed in metal cages at 23 ± 2 °C and 40–60% relative humidity with a 12 h light cycle. Food and water was provided ad libitum throughout the experimental period. The rats were adapted to the experimental location for two weeks prior to testing. Animal housing and care and the experimental protocols were conducted as stipulated in the Guide for the Care and Use of Laboratory Animals by the National Institutes of Health (NIH) and as approved by the local authorities of Zagazig University, Zagazig, Egypt. All efforts were made to minimize animal suffering. 2.2. Experimental design Rats were randomly classified into four equal groups (n = 6 each) as follows: Group 1, control group: each rat received 1/2 ml sterile saline solution orally as well as intraperitoneally once daily for 6 successive days. Group 2, curcumin-treated group (Curcumin): each rat was gavaged with curcumin (200 mg/kg/day) [31]. Group 3, colistin-treated group (Colistin): each rat received colistin (300,000 IU/kg/day) intraperitoneally for 6 successive days [32]. Group 4, (Concurrent): each rat received curcumin (200 mg/kg/day) 1 h before colistin (300,000 IU/kg/day) administration at for 6 days.

2.7. Statistical analysis The data are presented as the mean ± SE for each group. The variation between groups was statistically analyzed using one-way analysis of variance (ANOVA) followed by Duncan's multiple range post hoc test for pairwise comparisons. Differences were considered significant at p < 0.05.

2.3. Evaluation of biochemical parameters

3. Results

At the end of the 6-day experimental period, rats were fasted overnight and then sacrificed. Blood samples were collected from each rat in a glass tube without EDTA, left for 20 min to coagulate at room temperature and then centrifuged at 3000 rpm for 20 min to obtain serum. Serum samples were preserved at −20 °C until use for colorimetric evaluation of serum urea, creatinine and uric acid concentrations using a CE1020 spectrophotometer, according to previously

3.1. Effects of curcumin on several biochemical parameters of colistintreated rats Table 1 shows that rats treated only with colistin had significantly higher levels of serum creatinine (0.86 mg/dl), uric acid (4.32 mg/dl) 57

Chemico-Biological Interactions 294 (2018) 56–64

N.E. Edrees et al.

Table 1 Effects of oral administration of curcumin and/or colistin on serum levels of creatinine, uric acid and urea and on brain GABA concentrations. Groups

Parameters Creatinine (mg/dl) c

Control Curcumin Colistin Concurrent

Uric acid (mg/dl)

Urea (mg/dl)

bc

3.45 ± 0.17 3.34 ± 0.05c 4.32 ± 0.13a 3.81 ± 0.1b p = 0.002

0.58 ± 0.03 0.64 ± 0.04bc 0.86 ± 0.03a 0.72 ± 0.02b p = 0.001

GABA (ng/ml) b

18.83 ± 1.08 18.02 ± 1.53b 27.97 ± 1.03a 22.79 ± 2.29b p = 0.007

0.105 ± 0.014bc 0.089 ± 0.024c 0.204 ± 0.004a 0.147 ± 0.01b p = 0.003

Values are represented as the mean ± SE (n = 5). Control: received oral and intraperitonealsterile saline solution once daily, curcumin: gavaged with curcumin (200 mg/kg/day), colistin: injected with colistin (IP, 300,000 IU/kg/day), concurrent: received curcumin orally 1 h before IP colistin administration. All treatments were administered for 6 successive days. Means within the same column (in each parameter) carrying different superscripts (a, b, c) are significantly different at p < 0.05. GABA: Gamma amino butyric acid.

curcumin-treated rats reflected a normal histological structure in both renal corpuscles and tubules (Fig. 1A and B). By contrast, the kidneys of the colistin-treated rats revealed various inflammatory, degenerative and necrotic alterations manifested by vascular congestion, interstitial edema with mononuclear cell infiltration, cellular and hyaline cast formation, and acute tubular necrosis (Fig. 1C and D). Other alterations including tubular dilatation and vacuolar degeneration of the tubular epithelium particularly of the proximal and distal convoluted tubules were commonly encountered. No apparent histological changes were seen in the glomeruli or the basement membranes, but mild glomerular congestion was noticed in a few cases. In the concurrent group, the inflammatory and necrotic changes were less noticeable compared with the colistin-treated animals and the most encountered lesions were vascular congestion, epithelial vacuolation and mild interstitial mononuclear cell infiltration (Fig. 1 E). Immunohistochemically, the renal expression of Bcl-2 and caspase-3 in all groups were shown in Fig. 1(F–J) and (K-O) respectively.

and urea (27.97 mg/dl) as well as brain GABA (0.204 ng/ml) in comparison to control rats (0.58 mg/dl, 3.45 mg/dl, 18.83 mg/dl and 0.105 ng/ml, respectively). However, rats that were co-administered with both curcumin and colistin were intermediate in terms of all markers between colistin-only and control rats, for both renal function (creatinine 0.72 mg/dl, uric acid 3.81 mg/dl; urea (22.79 mg/dl) and brain function (GABA 0.147 ng/ml). 3.2. Effects of curcumin on renal and brain oxidant/antioxidant status following colistin-treatment Intraperitoneal administration of colistin resulted in a significant decrease in the CAT and GSH levels as well as a significant increase in MDA concentrations in kidney and brain tissues compared to the control group. However, administration of curcumin concurrently with colistin induced a significant elevation in the CAT and GSH levels as well as a significant (p < 0.05) reduction in MDA concentrations in brain and renal tissues compared to those of the colistin-treated group (Table 2).

3.4.2. Cerebral cortex The light microscopic panorama showed the normal histological structure in the of the control and curcumin-treated animals (Fig. 2A and B). The cerebral cortices of the colistin-treated rats revealed numerous neuropathic alterations as some neurons appeared shrunken with pyknotic nuclei and scanty cell bodies while others were swollen with marked eosinophilic cytoplasm. This was accompanied with vacuolations of the neuropil and vascular congestion of the cortices and choroid plexuses (Fig. 2C and D). Meningeal congestion, focal gliosis with perivascular lymphocytic cuffing were also observed. The neuropathic changes in the cerebral cortices of the concurrent group were minimal as they demonstrated almost the same histological picture as the control. However, they displayed neuropil vacuolations and mild perivascular cuffing (Fig. 2E). Immunohistochemically, the expression of Bcl-2 and caspase-3 in the cerebral cortices of all groups were shown

3.3. Effects of curcumin on inflammatory markers in renal and brain tissues of colistin-treated rats Treatment of adult male rats intraperitoneally with colistin significantly (p < 0.05) increased TNF-α, IL-6 and NO concentrations in kidney and brain homogenates compared to those of the control group. Interestingly, co-administration of curcumin with colistin significantly (p < 0.05) ameliorated each of these parameters (Table 3). 3.4. Histopathological and immunohistochemical findings 3.4.1. Kidneys The light microscopic investigation of the kidneys of the control and

Table 2 Effects of oral administration of curcumin and/or colistin on renal and brain CAT, GSH and MDA concentrations. Groups

Parameters CAT (U/g tissue)

Control Curcumin Colistin Concurrent

GSH (mmol/g tissue)

MDA (nmol/g tissue)

Kidney

Brain

Kidney

Brain

Kidney

Brain

0.205 ± .0.004a 0.219 ± 0.01a 0.12 ± 0.01c 0.16 ± 0.01b p = 0.001

0.207 ± .0.003a 0.214 ± 0.02a 0.105 ± 0.01c 0.163 ± 0.013b p = 0.000

0.219 ± 0.003a 0.209 ± 0.003ab 0.105 ± 0.014c 0.186 ± 0.01b p = 0.000

0.283 ± 0.01a 0.282 ± 0.02a 0.118 ± 0.01c 0.186 ± 0.014b p = 0.000

0.132 ± 0.01c 0.131 ± 0.002c 0.379 ± 0.04a 0.250 ± 0.01b p = 0.000

0.125 ± 0.01c 0.102 ± 0.01c 0.304 ± 0.004a 0.252 ± 0.02b p = 0.000

Values are represented as the mean ± SE (n = 5). Control: received oral and intraperitoneal sterile saline solution once daily, curcumin: gavaged with curcumin (200 mg/kg/day), colistin: injected with colistin (IP, 300,000 IU/kg/day), concurrent: received curcumin orally 1 h before IP colistin administration. All treatments applied for 6 successive days. Means within the same column (in each parameter) carrying different superscripts (a, b, c) are significantly different at p < 0.05. CAT: Catalase. GSH: Reduced glutathione. MDA, Malondialdehyde. 58

Chemico-Biological Interactions 294 (2018) 56–64

N.E. Edrees et al.

Table 3 Effects of oral administration of curcumin and/or colistin on renal and brain NO, TNF-α and IL-6 concentrations. Groups

Parameters NO (μmol/g tissue)

Control Curcumin Colistin Concurrent

TNF-α (pg/ml)

IL-6 (pg/ml)

Kidney

Brain

Kidney

Brain

Kidney

Brain

0.099 ± 0.01c 0.104 ± 0.012c 0.289 ± 0.01a 0.208 ± 0.003b p = 0.000

0.114 ± 0.011c 0.102 ± 0.014c 0.243 ± 0.017a 0.183 ± 0.021b p = 0.001

0.179 ± 0.014b 0.169 ± 0.016b 0.311 ± 0.011a 0.198 ± 0.005b p = 0.000

0.117 ± 0.004c 0.114 ± 0.01c 0.287 ± 0.024a 0.186 ± 0.01b p = 0.000

0.13 ± 0.015c 0.119 ± 0.01c 0.302 ± 0.024a 0.189 ± 0.014b p = 0.000

0.096 ± 0.022c 0.104 ± 0.03c 0.269 ± 0.021a 0.184 ± 0.012b p = 0.002

Values are represented as the mean ± SE (n = 5). Control: received oral and intraperitoneal sterile saline solution once daily, curcumin: gavaged with curcumin (200 mg/kg/day), colistin: injected with colistin (IP, 300,000 IU/kg/day), concurrent: received curcumin orally 1 h before IP colistin administration. All treatments applied for 6 successive days. Means within the same column (in each parameter) carrying different superscripts (a, b, c) are significantly different at p < 0.05. NO: Nitric oxide. TNF-α: Tumor necrosis factor-α. IL-6, Interleukin-6.

Fig. 1. (A–E) Photomicrograph of H&E stained renal sections showing normal histological structure of the glomeruli (arrows) and renal tubules (arrowheads) in the control (A), and curcumin-treated rats (B). In colistin-treated rats there is vascular congestion (red arrow), extravasated RBCs (red arrowhead), interstitial mononuclear cell infiltration (blue arrow), perivascular edema (blue arrowhead), tubular casts (black arrow) (C), acute tubular necrosis (black arrowhead), pyknosis of the tubular epithelium (blue arrowhead), and glomerular (red arrow) and capillary (red arrow head) congestions (D). Vascular congestion (arrow) and interstitial mononuclear cell infiltration (arrowhead) are seen in the concurrent group (E). (F–J) Photomicrograph of the renal tissue showing the immunoexpression of Bcl-2 (red arrows) as follow: negative in the negative control (F), moderate in the control (G), strong in the curcumin-treated (H), mild in the colistin-treated (I), and moderate in the colistin-curcumin-treated rats (J). (K–O) Photomicrograph of the renal tissue showing the immunoexpression of caspase-3 (red arrows) as follow: negative in the negative control (K), control (K), and curcumin-treated (M), moderate in the colistin-treated (N), and mild in the colistin-curcumin-treated rats (O). (For interpretation of the references to color in this figure legend, the reader is referred to the Web version of this article.)

3.4.4. Lesion scoring All the encountered lesions in the kidneys, cerebral and cerebellar cortices in all groups were scored in Table 4. The frequency of the lesion in different groups declared that curcumin treatment ameliorated the colistin-induced nephro and neuropathy.

in Fig. 2(F–J) and (K-O) respectively.

3.4.3. Cerebellar cortex The light microscopy of the cerebellar cortices of the control and curcumin-treated groups showed a normal histological picture (Fig. 3A and B). In the colistin-treated group, the effect was concentrated in the Purkinje cell layer, where some Purkinje cells were disorganized, and others were pyknotic, lost their normal pyriform shape and surrounded with perineural vacuolations and in few cases, it could not be detected in some areas. Congestion of the granular layers and meninges was commonly detected (Fig. 3C and D). The cerebella of the concurrent group almost maintained their normal histology except for perineural vacuolation and few disorganized Purkinje cells (Fig. 3E). Immunohistochemically, the expression of Bcl-2 and caspase-3 in the cerebellar cortices of all groups were shown in Fig. 3(F–J) and (K-O) respectively.

3.4.5. Immunohistochemical quantitative assessment Additionally, the expression of Bcl-2 and caspase-3 in these organs was scored in Table 5. Briefly, the control group demonstrated moderate Bcl-2 expression (26–50%) in the renal and cerebral cortices, and mild expression (10–25%) in the cerebellar cortices tissue sections. The curcumin-treated group revealed strong Bcl-2 expression (51–75%) in the renal and cerebral cortices, and moderate expression (26–50%) in cerebellar cortices tissue sections. Colistin treatment downregulated Bcl-2 expression to mild (10–25%) in renal and cerebral cortices and into weak (less than 10%) in the cerebellar cortices tissue sections. Treatment with curcumin succeeded to upregulate Bcl-2 expression to moderate (26–50%) in the renal and cerebral cortices compared to the 59

Chemico-Biological Interactions 294 (2018) 56–64

N.E. Edrees et al.

Fig. 2. (A–E) Photomicrograph of H&E stained cerebral cortices tissue sections showing normal histological picture in the control (A) and curcumin-treated rats (B). The colistin-treated rats showed shrunken neurons with pyknotic nuclei and scanty cell bodies (black arrowhead), swollen neurons with marked eosinophilic cytoplasm (black arrow), neuropil vacuolations (blue arrowheads), vascular congestion (red arrow) and hemorrhage (red arrowhead) (C) and congested choroid plexus (red arrows) (D). Neuropil vacuolations (arrowheads) and perivascular lymphocytic cuffing (arrow) are seen in the curcumin-colistin-treated rats (E). (F–J) Photomicrograph of the cerebral cortices tissue sections showing the immunoexpression of Bcl-2 (red arrows) as follow: negative in the negative control (F), moderate in the control (G), strong in the curcumin-treated (H), mild in the colistin-treated (I), and moderate in the colistin-curcumin-treated rats (J). (K–O) Photomicrograph of the cerebral cortices tissue sections showing the immunoexpression caspase-3 (red arrows) as follow: negative in the negative control (K), control (K), and curcumin-treated (M), moderate in the colistin-treated (N), and mild in the colistin-curcumin-treated rats (O). (For interpretation of the references to color in this figure legend, the reader is referred to the Web version of this article.)

upregulated the expression to moderate (26–50%) in the renal and cerebral cortices and mild (10–25%) in cerebellar cortices compared to the control group. Curcumin treatment regained the normal values for caspase-3 tissue expression in the cerebellar cortices and

colistin-treated group, with no effect on the cerebellar cortices. The caspase-3 expression was negative in the kidneys, and cerebral and cerebellar cortices in the control, curcumin-treated groups. The effect of colistin treatment on the caspase-3 tissue expression was obvious as it

Fig. 3. (A–E) Photomicrograph of H&E stained cerebellar cortices tissue sections showing normal histological picture in the control (A) and curcumin-treated rats (B). The colistin-treated rats showing congested granular layer (red arrow), pyknotic Purkinje cells (arrowhead), absent Purkinje cells in some areas (black line), and perineural vacuolations (black arrows) (C) and meningeal congestion (red arrowheads) (D). Neuropil vacuolations (arrow) and disorganized Purkinje cells (arrowhead) is shown in the curcumin-colistin-treated rats (E). (F–J) Photomicrograph of the cerebellar cortices tissue sections showing the immunoexpression of Bcl-2 (red arrows) as follow: negative in the negative control (F), mild in the control (G), moderate in the curcumin-treated (H), weak in the colistin-treated (I), and mild in the colistin-curcumin-treated rats (J). (K–O) Photomicrograph of the cerebellar cortices tissue sections showing the immunoexpression caspase-3 (red arrows) as follow: negative in the negative control (K), control (K), and curcumin-treated (M), mild in the colistin-treated (N), and negative in the colistin-curcumin-treated rats (O). (For interpretation of the references to color in this figure legend, the reader is referred to the Web version of this article.) 60

Chemico-Biological Interactions 294 (2018) 56–64

N.E. Edrees et al.

and GSH levels and elevation of MDA concentrations [29,30,51]. Our results are also in line with those of Dai, Tang, Deng, Zhang, Zhou, Velkov, Li and Xiao [51] and Dai, Tang, Wang, Velkov and Xiao [52], who found that intravenous administration of colistin induced a significant reduction in CAT and GSH levels and a significant elevation in lipid peroxidation of mouse renal tissues. Similarly, the in vitro results of Liu, Dai, Gao and Li [53] showed a significant elevation of ROS and a significant reduction of GSH in PC12 cells. Furthermore, colistin provoked ROS generation in a dosedependent manner in cultured human proximal tubular cells [54]. Additionally, Dai, Ciccotosto, Cappai, Tang, Li, Xie, Xiao and Velkov [55] and Dai, Ciccotosto, Cappai, Wang, Tang, Xiao and Velkov [56] found that colistin significantly increased the intracellular ROS and decreased CAT and GSH levels in neuroblastoma-2a (N2a) cells. These results suggest the role of oxidative mechanisms in colistininduced tissue damage and may be attributed to the exhaustion of the antioxidants in combating ROS that were generated by colistin administration, leading to oxidative damage to the cell membrane, as indicated by the increased MDA concentration [57]. These results are supported by the histopathological findings from the brain and kidney tissues of colistin-treated rats. Interestingly, our results indicated that administration of curcumin with colistin protected renal and brain tissues against colistin-induced oxidative stress via improvements in antioxidant status (CAT and GSH levels) and consequently reduced lipid peroxidation (MDA concentration). This amelioration of the oxidant/antioxidant status of renal and brain tissues by curcumin could be attributed to a direct reduction of ROS generation and release [58], scavenging of free radicals and subsequent inhibition of oxygenation reactions, as curcumin has been reported to be a good antioxidant and free radical scavenger which inhibits lipid peroxidation [14]. Indeed, curcumin may reduce lipid peroxidation by enhancing the activities of antioxidant enzymes and GSH levels [59–61]. Together, these mechanisms might explain, at least in part, the cytoprotective effects of curcumin which confirmed by the improvement of renal and brain structure of concurrent group. Similarly, Gonzalez-Reyes, Guzman-Beltran, Medina-Campos and PedrazaChaverri [62] suggested that curcumin protects cerebellar granule neurons against hemin-induced neuronal death via reduction of ROS production as well as induction of nuclear factor (erythroid-2)-related factor 2 (Nrf2), a master regulator of the cell antioxidant response, GSH and antioxidant enzymes that may play an important role in the protective effect of this antioxidant. Inflammation occurs as a consequence of tissue and organ exposure to harmful stimuli, such as microbial pathogens, toxic cellular components or irritants [63]. Oxidative stress and inflammation are closely linked pathophysiological processes, as one can be easily stimulated by the other [64]. Indeed, oxidative stress can induce inflammatory cytokine production and vice versa [65]. Moreover, oxidative stress can induce expression of transcription factors which themselves cause expression of genes involved in inflammatory pathways [66]. NO is considered a pro-inflammatory mediator [67], and its overproduction can cause direct DNA damage, mitochondrial membrane damage, or apoptosis [28]. Moreover, TNF-α is a potent pro-inflammatory cytokine and an important mediator of inflammatory tissue damage [68,69]. In pathological conditions, microglia release high levels of TNF-α; this de novo production of TNF-α is an important component of the neuroinflammatory response that is linked to many neurological disturbances [70]. Pro-inflammatory cytokines also enhance neutrophil aggregation and the production of IL-6 [71], which is immediately and transiently released in response to infections and tissue injuries [72]. Our results indicated the involvement of inflammation in colistin-induced toxicities via elevation of NO, TNF-α and IL-6 in renal and brain tissues of colistin-treated rats. Oxidative stress induced by colistin might play an essential role in the activation of the inflammatory response via the release of pro-inflammatory cytokine (TNF- α and IL-6) and the migration of inflammatory cells to affected tissues. Additionally, it has

Table 4 Lesion scoring in the kidneys and cerebral and cerebellar cortices in all groups. Organ

lesion

Control

Curcumin

Colistin

Kidney

Vascular congestion Hemorrhages Interstitial edema Interstitial mononuclear cell infiltration Tubular dilatation Tubular epithelial vacuolation Tubular necrosis Cast formation Glomerulitis Congestion Cerebral cortex Meninges Choroid plexus Pyknotic neurons Swollen eosinophilic neurons Neuropil vacuolation Gliosis Perivascular cuffing Congestion Granular layer Meninges Purkinje cell Disorganization Pyknosis Loss of pyriform shape Perineural vacuolation Necrosis

– – – –

– – – –

+++ + ++ +++

– – – – – – – – – – – – – – – – – –

– – – – – – – – – – – – – – – – – –

++ +++ ++ +++ – +++ +++ + ++++ +++ ++++ ++ ++ + ++ +++ ++ ++





+++





+

Cerebral cortex

Cerebellar cortex

The score system was designed as the following: (−) absence of the lesion in all animals of the group, (+) the lesion was rare within the group, (++) the lesion not so often observed in all animals of the group, (+++), the lesion observed in almost all animals of the group, (++++) the lesion often found in all animals of the group. Table 5 The scores of renal, cerebral and cerebellar expressions of Bcl-2 and Caspase-3 in all groups. Organ

Marker

Negative control

Control

Curcumin

Colistin

Concurrent

Kidneys

Bcl-2 Caspase-3 Bcl-2 Caspase-3 Bcl-2 Caspase-3

0 0 0 0 0 0

++ 0 ++ 0 + 0

+++ 0 +++ 0 ++ 0

+ ++ + ++ 0 +

++ + ++ + + 0

Cerebral cortex Cerebellar cortex

Sample scores were calculated based on the percentage of positive cells per five non-repeated randomly selected microscopic fields (40X) as following: 0 (negative to weak) = less than 10%, + (mild) = 10–25%, ++ (moderate) = 26–50%, +++ (strong) = 51–75%, and ++++ (severe) = more than 75% (severe).

downregulated its expression to mild (10–25%) in the renal and cerebral cortices but failed to normalize it. 4. Discussion The emergence of MDR gram-negative bacteria has renewed interest in colistin use, which had fallen out of favor because of reports of nephrotoxicity and neurotoxicity [43]. Amelioration of these side effects would increase the therapeutic appeal of colistin and permit administration of higher doses of colistin. Oxidative stress plays a pivotal role in many diseases and in a variety of drug-induced hepatic, cardiac, renal and neurotoxicities [44–47]. Increased production of ROS is an important mechanism in colistin-induced nephrotoxicity and neurotoxicity [29,48–50]. Consistent with previous studies, our findings reveal that intraperitoneal administration of colistin induced oxidative stress in renal and brain tissues, as shown by the significant reduction in CAT 61

Chemico-Biological Interactions 294 (2018) 56–64

N.E. Edrees et al.

oxidative stress, inflammation and apoptosis in renal tissues of these animals. Histologically, degenerative changes in renal tubules with tubular dilatation, the presence of casts in the lumen of some renal tubules and vacuolations in the epithelial lining of some renal tubules with interstitial leukocytic infiltration were recorded in colistin-treated rats. Similarly, Ozkan, Ulusoy, Orem, Alkanat, Mungan, Yulug and Yucesan [28] stated that IP injection of colistin significantly increased blood urea nitrogen and creatinine levels. Furthermore, IP injection of colistin (480,000 IU/kg/day) resulted in a significant elevation of serum creatinine and an increased number of apoptotic cells [90]. In this context, curcumin co-administration in colistin-treated rats significantly decreased the levels of serum creatinine, urea and uric acid, and improved histological parameters, again perhaps via the antioxidant, anti-inflammatory and anti-apoptotic effects of curcumin. Histologically, the renal tubules showed regenerative attempts in the 4th group., curcumin administration to gentamicin-treated rats decreased the concentrations of urea and creatinine, elevated GSH and superoxide dismutase (SOD) levels and ameliorated the histopathological findings [91]. Curcumin significantly reduced the lipid peroxidation and renal dysfunction in cyclosporine-treated rats, while elevating their levels of antioxidant enzymes and normalizing their altered renal structures [92]. Finally, treatment of methotrexate-intoxicated rats with curcumin resulted in nephroprotective effects as evidenced by the significant decrease in levels of serum creatinine and urea as well as renal MDA, NO, and TNF-α with a concurrent increase in renal GSH-Px and SOD activities compared to nephrotoxic untreated rats (Morsy et al., 2013). GABA is the most important inhibitory neurotransmitter in the mammalian CNS [93]. Our results showed that intraperitoneal injection of colistin induced a significant increase in GABA concentration in brain tissue. Since glutamate content can be elevated via excess TNF-α release from microglial cells under pathological conditions [70], our results may be attributed to an elevation of glutamate content in brain tissue [94] that was converted into GABA via glutamic acid decarboxylase. Therefore, the anti-inflammatory effect of curcumin could contribute to the decreased GABA concentration in brain tissues of colistin-treated rats. Similarly, Wang, Yi, Chen, Muhammad, Liu, Li, Li and Li [94] stated that colistin accumulates in the mouse brain and markedly elevated glutamate and GABA contents and increased the mRNA expression levels of GABA type A and B receptors. Our findings were supported by those of Dai, Ciccotosto, Cappai, Tang, Li, Xie, Xiao and Velkov [55] who stated that a potential role for curcumin for treating colistin-induced neurotoxicity through the modulation of NF-κB signaling and its potent anti-oxidative and anti-apoptotic effects.

been shown to promote the expression of the pro-inflammatory enzyme cyclooxygenase 2 (COX-2) and inflammatory transcription factors, such as nuclear factor-kappa B (NF-κB), in N2a cells [55]. The anti-inflammatory effects of curcumin observed in the rats simultaneously treated with colistin and curcumin may be mediated through the ability of curcumin to suppress the pro-inflammatory cytokines TNF-α and IL-6 in renal and brain tissues. Ghosh, Banerjee and Sil [27] suggested that the anti-inflammatory effects of curcumin are due to its ability to reduce TNF-α, IL-1, IL-6, COX-2 and NF-κB. Curcumin also binds to TNF-α directly, and can inhibit both the production and activity of this cytokine [73]. Furthermore, it down-regulated the expression of COX-2 and NF-κB in colistin-exposed-N2a cells and markedly decreased serum TNF-α and IL-6 levels in the late phase of acute pancreatitis [74,75]. The elevated NO concentration in renal and brain tissues we observed may be due to the activation of NO synthase enzymes by colistin administration. Indeed, our results were supported by those of Ozyilmaz, Ebinc, Derici, Gulbahar, Goktas, Elmas, Oguzulgen and Sindel [32] and Ozkan, Ulusoy, Orem, Alkanat, Mungan, Yulug and Yucesan [28], who stated that intraperitoneal injection of 300,000 IU colistin/kg/day for 6 consecutive days resulted in overexpression of inducible nitric oxide synthase (iNOS) and endothelial nitric oxide synthase (eNOS) in renal tissues of rat. Our findings indicated that curcumin treatment may be a strategy to reduce the NO concentration of renal and brain tissues, perhaps via inhibition of NO synthase. Similarly, Jung, Lee, Cho, Shin, Rhee, Kim, Kang, Kim, Hong and Kang [76] and Song, Yim, Yim, Kang, Rho, Kim, Yhim, Lee, Song, Kwak, Sohn and Yim [77] reported that curcumin decreased NO production and iNOS gene expression in LPS-stimulated microglial cells and in a mouse ascites tumor model. Furthermore, Parada, Buendia, Navarro, Avendaño, Egea and López [78] stated that curcumin suppressed iNOS and TNF-α in LPS-activated microglial cells. Apoptosis is a normal process for eliminating unwanted cells during development and for maintaining tissue homeostasis [79]. Deregulation of this process causes several disorders, including renal and neurodegenerative disorders [80,81]. Caspase-3 is a frequently stimulated death protease, as it enhances the specific cleavage of many key cellular proteins [82] and leads to DNA breakdown, one of the characteristic cellular changes of apoptosis [83]. Bcl-2 inhibits apoptosis either by sequestering caspases or by inhibiting the release of mitochondrial apoptogenic factors, cytochrome c, and apoptosis-inducing factor into the cytoplasm [84]. We observed the up-regulation of caspase-3 expression as well as down-regulation of Bcl-2 expression in the brain and kidney tissues of the colistin-treated rats, suggesting that apoptosis may be involved in the pathogenesis of colistin toxicity. Jiang, Li, Zhou, Wang, Zhang and Wang [85] also found that colistin sulfate increased ROS levels, causing cytochrome c release and DNA damage. DNA damage, in turn, can promote p53, which leads to an imbalance of Bax/Bcl-2, promoting further cytochrome c release and resulting in caspase-9 activation and the subsequent caspase-3 activation, to ultimately induce apoptosis. Similarly, intravenous administration of colistin in mice caused down-regulation of Bcl-2 and stimulation of caspase-3 in renal tissues [86]. Moreover, this treatment could induce oxidative stress and apoptotic cell death in RSC96 Schwann cells via up-regulation of caspase-3 and down-regulation of Bcl-2 expression [87]. In the present study, curcumin ameliorated the effect of colistin on the expression levels of capase-3 and Bcl-2 in the 4th group. These results may be caused by the ability of curcumin to prevent GSH decrease, thus protecting cells against caspase-3 activation and DNA fragmentation [88]. Likewise, Park and Chun [89] found down-regulation of caspase-3 expression and ROS levels as well as elevation of Bcl-2 expression and intracellular GSH levels in manganese-exposed BV-2 microglial cells. In the colistin-treated rats, renal injury was evident by the notable elevation in the serum levels of urea, creatinine and uric acid caused by

5. Conclusion To our knowledge, this is the first in vivo study that assessed the protective effects of curcumin on colistin-induced nephrotoxicity and neurotoxicity. We revealed the involvement of oxidative stress, inflammation and apoptosis in colistin-induced toxicities. Our findings are consistent with a beneficial effect of curcumin administration in ameliorating these adverse effects via its antioxidant, anti-inflammatory and anti-apoptotic activities. Therefore, curcumin could represent a promising agent for prevention of colistin-induced nephrotoxicity and neurotoxicity. Supplementary studies are required to identify the possible additional effects, appropriate doses, and duration of the curcumin therapy on colistin-induced toxicities.

Transparency document Transparency document related to this article can be found online at https://doi.org/10.1016/j.cbi.2018.08.012.

62

Chemico-Biological Interactions 294 (2018) 56–64

N.E. Edrees et al.

References [31]

[1] M.E. Falagas, S.K. Kasiakou, Colistin: the revival of polymyxins for the management of multidrug-resistant gram-negative bacterial infections, Clin. Infect. Dis. : Offic. Publi. Infect. Dis. Soci. Am. 40 (2005) 1333–1341. [2] J. Li, R.L. Nation, R.W. Milne, J.D. Turnidge, K. Coulthard, Evaluation of colistin as an agent against multi-resistant Gram-negative bacteria, Int. J. Antimicrob. Agents 25 (2005) 11–25. [3] T. Loho, A. Dharmayanti, Colistin: an antibiotic and its role in multiresistant Gramnegative infections, Acta Med. Indones. 47 (2015) 157–168. [4] S.D. Davis, A. Iannetta, R.J. Wedgwood, Activity of colistin against Pseudomonas aeruginosa: inhibition by calcium, J. Infect. Dis. 124 (1971) 610–612. [5] M. Schindler, M.J. Osborn, Interaction of divalent cations and polymyxin B with lipopolysaccharide, Biochemistry 18 (1979) 4425–4430. [6] J. Li, R.L. Nation, J.D. Turnidge, R.W. Milne, K. Coulthard, C.R. Rayner, D.L. Paterson, Colistin: the re-emerging antibiotic for multidrug-resistant Gramnegative bacterial infections, the Lancet, Infect. Dis. 6 (2006) 589–601. [7] H. Spapen, R. Jacobs, V. Van Gorp, J. Troubleyn, P.M. Honoré, Renal and neurological side effects of colistin in critically ill patients, Ann. Intensive Care 1 (2011) 14-14. [8] K. Wahby, T. Chopra, P. Chandrasekar, Intravenous and inhalational colistin-induced respiratory failure, Clin. Infect. Dis. :Offic. Publi. Infect. Dis. Soci. Am. 50 (2010) e38–40. [9] P.M. Honore, R. Jacobs, S. Lochy, E. De Waele, V. Van Gorp, J. De Regt, G. Martens, O. Joannes-Boyau, W. Boer, H.D. Spapen, Acute respiratory muscle weakness and apnea in a critically ill patient induced by colistin neurotoxicity: key potential role of hemoadsorption elimination during continuous venovenous hemofiltration, Int. J. Nephrol. Renovascular Dis. 6 (2013) 107–111. [10] A. Noorafshan, S. Ashkani-Esfahani, A review of therapeutic effects of curcumin, Curr. Pharmaceut. Des. 19 (2013) 2032–2046. [11] X. Gao, J. Kuo, H. Jiang, D. Deeb, Y. Liu, G. Divine, R.A. Chapman, S.A. Dulchavsky, S.C. Gautam, Immunomodulatory activity of curcumin: suppression of lymphocyte proliferation, development of cell-mediated cytotoxicity, and cytokine production in vitro, Biochem. Pharmacol. 68 (2004) 51–61. [12] H. Hanai, K. Sugimoto, Curcumin has bright prospects for the treatment of inflammatory bowel disease, Curr. Pharmaceut. Des. 15 (2009) 2087–2094. [13] M.M. Chan, H.I. Huang, M.R. Fenton, D. Fong, In vivo inhibition of nitric oxide synthase gene expression by curcumin, a cancer preventive natural product with anti-inflammatory properties, Biochem. Pharmacol. 55 (1998) 1955–1962. [14] T. Ak, İ. Gülçin, Antioxidant and radical scavenging properties of curcumin, Chem. Biol. Interact. 174 (2008) 27–37. [15] W.H. Chan, H.J. Wu, Anti-apoptotic effects of curcumin on photosensitized human epidermal carcinoma A431 cells, J. Cell. Biochem. 92 (2004) 200–212. [16] C. Aktas, M. Kanter, M. Erboga, S. Ozturk, Anti-apoptotic effects of curcumin on cadmium-induced apoptosis in rat testes, Toxicol. Ind. Health 28 (2012) 122–130. [17] M. Dikmen, Z. Canturk, Y. Ozturk, Y. Tunali, Investigation of the apoptotic effect of curcumin in human leukemia HL-60 cells by using flow cytometry, Cancer Biother. Radiopharm. 25 (2010) 749–755. [18] S. Palipoch, C. Punsawad, P. Koomhin, P. Suwannalert, Hepatoprotective effect of curcumin and alpha-tocopherol against cisplatin-induced oxidative stress, BMC Compl. Alternative Med. 14 (2014) 111. [19] R. Singh, P. Sharma, Hepatoprotective effect of curcumin on lindane-induced oxidative stress in male wistar rats, Toxicol. Int. 18 (2011) 124–129. [20] D. Cerny, N. Lekic, K. Vanova, L. Muchova, A. Horinek, E. Kmonickova, Z. Zidek, L. Kamenikova, H. Farghali, Hepatoprotective effect of curcumin in lipopolysaccharide/-galactosamine model of liver injury in rats: relationship to HO-1/CO antioxidant system, Fitoterapia 82 (2011) 786–791. [21] J. Trujillo, Y.I. Chirino, E. Molina-Jijón, A.C. Andérica-Romero, E. Tapia, J. Pedraza-Chaverrí, Renoprotective effect of the antioxidant curcumin: recent findings, Redox Biol. 1 (2013) 448–456. [22] S. Ugur, R. Ulu, A. Dogukan, A. Gurel, I.P. Yigit, N. Gozel, B. Aygen, N. Ilhan, The renoprotective effect of curcumin in cisplatin-induced nephrotoxicity, Ren. Fail. 37 (2015) 332–336. [23] E.A. Hashish, S.A. Elgaml, Hepatoprotective and nephroprotective effect of curcumin against copper toxicity in rats, Indian J. Clin. Biochem. 31 (2016) 270–277. [24] G.M. Cole, B. Teter, S.A. Frautschy, Neuroprotective effects of curcumin, Adv. Exp. Med. Biol. 595 (2007) 197–212. [25] C.P. Kuo, C.H. Lu, L.L. Wen, C.H. Cherng, C.S. Wong, C.O. Borel, D.T. Ju, C.M. Chen, C.T. Wu, Neuroprotective effect of curcumin in an experimental rat model of subarachnoid hemorrhage, Anesthesiology 115 (2011) 1229–1238. [26] D.K. Khatri, A.R. Juvekar, Neuroprotective effect of curcumin as evinced by abrogation of rotenone-induced motor deficits, oxidative and mitochondrial dysfunctions in mouse model of Parkinson's disease, Pharmacol. Biochem. Behav. 150–151 (2016) 39–47. [27] S. Ghosh, S. Banerjee, P.C. Sil, The beneficial role of curcumin on inflammation, diabetes and neurodegenerative disease: a recent update, Food Chem. Toxicol. : Int. J. Publ. Br. Ind. Biol. Res. Assoc. 83 (2015) 111–124. [28] G. Ozkan, S. Ulusoy, A. Orem, M. Alkanat, S. Mungan, E. Yulug, F.B. Yucesan, How does colistin-induced nephropathy develop and can it be treated? AAC (Antimicrob. Agents Chemother.) 57 (2013) 3463–3469. [29] J.M. Yousef, G. Chen, P.A. Hill, R.L. Nation, J. Li, Melatonin attenuates colistininduced nephrotoxicity in rats, AAC (Antimicrob. Agents Chemother.) 55 (2011) 4044–4049. [30] Z. Ghlissi, A. Hakim, A. Sila, H. Mnif, K. Zeghal, T. Rebai, A. Bougatef, Z. Sahnoun, Evaluation of efficacy of natural astaxanthin and vitamin E in prevention of colistin-

[32]

[33] [34] [35] [36] [37] [38] [39]

[40] [41]

[42]

[43]

[44] [45]

[46]

[47]

[48] [49]

[50]

[51]

[52]

[53] [54]

[55]

[56]

[57]

[58]

[59] [60]

[61]

63

induced nephrotoxicity in the rat model, Environ. Toxicol. Pharmacol. 37 (2014) 960–966. J. Wu, X. Pan, H. Fu, Y. Zheng, Y. Dai, Y. Yin, Q. Chen, Q. Hao, D. Bao, D. Hou, Effect of curcumin on glycerol-induced acute kidney injury in rats, Sci. Rep. 7 (2017) 10114. E. Ozyilmaz, F.A. Ebinc, U. Derici, O. Gulbahar, G. Goktas, C. Elmas, I.K. Oguzulgen, S. Sindel, Could nephrotoxicity due to colistin be ameliorated with the use of N-acetylcysteine? Intensive Care Med. 37 (2011) 141–146. J.J. Coulombe, L. Favreau, A new simple semimicro method for colorimetric determination of urea, Clin. Chem. 9 (1963) 102–108. K. Larsen, Creatinine assay in the presence of protein with LKB 8600 Reaction Rate Analyser, Clin. Chim. Acta; Int. J. Clin. Chem. 38 (1972) 475–476. T.P. Whitehead, E.A. Bevan, L. Miano, A. Leonardi, Defects in diagnostic kits for determination of urate in serum, Clin. Chem. 37 (1991) 879–881. H. Aebi, Catalase in vitro, Meth. Enzymol. 105 (1984) 121–126. E. Beutler, O. Duron, B.M. Kelly, Improved method for the determination of blood glutathione, J. Lab. Clin. Med. 61 (1963) 882–888. M. Mihara, M. Uchiyama, Determination of malonaldehyde precursor in tissues by thiobarbituric acid test, Anal. Biochem. 86 (1978) 271–278. L.C. Green, D.A. Wagner, J. Glogowski, P.L. Skipper, J.S. Wishnok, S.R. Tannenbaum, Analysis of nitrate, nitrite, and [15N]nitrate in biological fluids, Anal. Biochem. 126 (1982) 131–138. S.K. Suvarna, C. Layton, J.D. Bancroft, Bancroft's Theory and Practice of Histological Techniques, eighth ed., Churchill Livingstone, Elsevier, England, 2018. A.A. Mohamed, A.A. Galal, Y.H. Elewa, Comparative protective effects of royal jelly and cod liver oil against neurotoxic impact of tartrazine on male rat pups brain, Acta Histochemica 117 (2015) 649–658. M.M. Soliman, M. Abdo Nassan, T.A. Ismail, Immunohistochemical and molecular study on the protective effect of curcumin against hepatic toxicity induced by paracetamol in Wistar rats, BMC Compl. Alternative Med. 14 (2014) 457. J.D. Hartzell, R. Neff, J. Ake, R. Howard, S. Olson, K. Paolino, M. Vishnepolsky, A. Weintrob, G. Wortmann, Nephrotoxicity associated with intravenous colistin (colistimethate sodium) treatment at a tertiary care medical center, Clin. Infect. Dis. 48 (2009) 1724–1728. D.G. Deavall, E.A. Martin, J.M. Horner, R. Roberts, Drug-induced oxidative stress and toxicity, J. Toxicol. (2012) 13 (2012). C.V. Pereira, S. Nadanaciva, P.J. Oliveira, Y. Will, The contribution of oxidative stress to drug-induced organ toxicity and its detection in vitro and in vivo, Expet Opin. Drug Metabol. Toxicol. 8 (2012) 219–237. W.M. El-Hady, A.A.A. Galal, Neurotoxic outcomes of subchronic manganese chloride exposure via contaminated water in adult male rats and the potential benefits of ebselen, Biol. Trace Elem. Res. (2018). A.A.A. Galal, A.A.-R. Mohamed, S.I. Khater, M.M.M. Metwally, Beneficial role of biochanin A on cutaneous and renal tissues of ovariectomized rats treated with anastrozole, Life Sci. 201 (2018) 9–16. Y. Liu, C. Dai, R. Gao, J. Li, Ascorbic acid protects against colistin sulfate-induced neurotoxicity in PC12 cells, Toxicol. Mech. Meth. 23 (2013) 584–590. C. Dai, J. Li, J. Li, New insight in colistin induced neurotoxicity with the mitochondrial dysfunction in mice central nervous tissues, Exp. Toxicol. Pathol.: Offic. J. Gesellschaft Fur Toxikologische Pathol. 65 (2013) 941–948. C. Dai, D. Zhang, R. Gao, X. Zhang, J. Li, J. Li, In vitro toxicity of colistin on primary chick cortex neurons and its potential mechanism, Environ. Toxicol. Pharmacol. 36 (2013) 659–666. C. Dai, S. Tang, S. Deng, S. Zhang, Y. Zhou, T. Velkov, J. Li, X. Xiao, Lycopene attenuates colistin-induced nephrotoxicity in mice via activation of the nrf2/HO-1 pathway, AAC (Antimicrob. Agents Chemother.) 59 (2015) 579–585. C. Dai, S. Tang, Y. Wang, T. Velkov, X. Xiao, Baicalein acts as a nephroprotectant that ameliorates colistin-induced nephrotoxicity by activating the antioxidant defence mechanism of the kidneys and down-regulating the inflammatory response, J. Antimicrob. Chemother. 72 (2017) 2562–2569. Y. Liu, C. Dai, R. Gao, J. Li, Ascorbic acid protects against colistin sulfate-induced neurotoxicity in PC12 cells, Toxicol. Mech. Meth. 23 (2013) 584–590. S.H. Lee, J.S. Kim, K. Ravichandran, H.W. Gil, H.Y. Song, S.Y. Hong, P-glycoprotein induction ameliorates colistin induced nephrotoxicity in cultured human proximal tubular cells, PLoS One 10 (2015) e0136075. C. Dai, G.D. Ciccotosto, R. Cappai, S. Tang, D. Li, S. Xie, X. Xiao, T. Velkov, Curcumin attenuates colistin-induced neurotoxicity in N2a cells via anti-inflammatory activity, suppression of oxidative stress, and apoptosis, Mol. Neurobiol. 55 (2018) 421–434. C. Dai, G.D. Ciccotosto, R. Cappai, Y. Wang, S. Tang, X. Xiao, T. Velkov, Minocycline attenuates colistin-induced neurotoxicity via suppression of apoptosis, mitochondrial dysfunction and oxidative stress, J. Antimicrob. Chemother. 72 (2017) 1635–1645. A. Ayala, M.F. Munoz, S. Argüelles, Lipid peroxidation: production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal, Oxidative Med. Cell. Longev. 2014 (2014) 31. B. Joe, B.R. Lokesh, Role of capsaicin, curcumin and dietary n-3 fatty acids in lowering the generation of reactive oxygen species in rat peritoneal macrophages, Biochim. Biophys. Acta 1224 (1994) 255–263. G. Jagetia, G. Rajanikant, Curcumin stimulates the antioxidant mechanisms in mouse skin exposed to fractionated γ-irradiation, Antioxidants 4 (2015) 25. A.C. Reddy, B.R. Lokesh, Effect of dietary turmeric (Curcuma longa) on iron-induced lipid peroxidation in the rat liver, Food Chem. Toxicol. : An Int. J. Publ. Br. Ind. Biol. Res. Assoc. 32 (1994) 279–283. M. Susan, M.N. Rao, Induction of glutathione S-transferase activity by curcumin in mice, Arzneimittelforschung 42 (1992) 962–964.

Chemico-Biological Interactions 294 (2018) 56–64

N.E. Edrees et al.

[78] E. Parada, I. Buendia, E. Navarro, C. Avendaño, J. Egea, M.G. López, Microglial HO1 induction by curcumin provides antioxidant, antineuroinflammatory, and glioprotective effects, Mol. Nutr. Food Res. 59 (2015) 1690–1700. [79] M. Mendez-Armenta, C. Nava-Ruiz, D. Juarez-Rebollar, E. Rodriguez-Martinez, P.Y. Gomez, Oxidative stress associated with neuronal apoptosis in experimental models of epilepsy, Oxid Med Cell Longev 2014 (2014) 293689. [80] M. Agostini, P. Tucci, G. Melino, Cell death pathology: perspective for human diseases, Biochem. Biophys. Res. Commun. 414 (2011) 451–455. [81] R. Bonegio, W. Lieberthal, Role of apoptosis in the pathogenesis of acute renal failure, Curr. Opin. Nephrol. Hypertens. 11 (2002) 301–308. [82] A.G. Porter, R.U. Janicke, Emerging roles of caspase-3 in apoptosis, Cell Death Differ. 6 (1999) 99–104. [83] S. Faubel, D. Ljubanovic, L. Reznikov, H. Somerset, C.A. Dinarello, C.L. Edelstein, Caspase-1-deficient mice are protected against cisplatin-induced apoptosis and acute tubular necrosis, Kidney Int. 66 (2004) 2202–2213. [84] Y. Tsujimoto, Role of Bcl-2 family proteins in apoptosis: apoptosomes or mitochondria? Gene Cell. : Devoted Mol. Cell. Mech. 3 (1998) 697–707. [85] H. Jiang, J. Li, T. Zhou, C. Wang, H. Zhang, H. Wang, Colistin-induced apoptosis in PC12 cells: involvement of the mitochondrial apoptotic and death receptor pathways, Int. J. Mol. Med. 33 (2014) 1298–1304. [86] C. Dai, J. Li, S. Tang, J. Li, X. Xiao, Colistin-induced nephrotoxicity in mice involves the mitochondrial, death receptor, and endoplasmic reticulum pathways, AAC (Antimicrob. Agents Chemother.) 58 (2014) 4075–4085. [87] Z. Lu, G. Jiang, Y. Chen, J. Wang, I. Muhammad, L. Zhang, R. Wang, F. Liu, R. Li, F. Qian, J. Li, Salidroside attenuates colistin-induced neurotoxicity in RSC96 Schwann cells through PI3K/Akt pathway, Chem. Biol. Interact. 271 (2017) 67–78. [88] K. Piwocka, E. Jaruga, J. Skierski, I. Gradzka, E. Sikora, Effect of glutathione depletion on caspase-3 independent apoptosis pathway induced by curcumin in Jurkat cells, Free Radic. Biol. Med. 31 (2001) 670–678. [89] E. Park, H.S. Chun, Protective effects of curcumin on manganese-induced BV-2 microglial cell death, Biol. Pharm. Bull. 40 (2017) 1275–1281. [90] B.Y. Arslan, F. Arslan, K. Erkalp, A. Alagöl, M.S. Sevdi, G. Yıldız, S.H. Küçük, S. Altınay, Luteolin ameliorates colistin-induced nephrotoxicity in the rat models, Ren. Fail. 38 (2016) 1735–1740. [91] B.H. Ali, N. Al-Wabel, O. Mahmoud, H.M. Mousa, M. Hashad, Curcumin has a palliative action on gentamicin-induced nephrotoxicity in rats, Fund. Clin. Pharmacol. 19 (2005) 473–477. [92] N. Tirkey, G. Kaur, G. Vij, K. Chopra, Curcumin, a diferuloylmethane, attenuates cyclosporine-induced renal dysfunction and oxidative stress in rat kidneys, BMC Pharmacol. 5 (2005) 15. [93] N.G. Bowery, T.G. Smart, GABA and glycine as neurotransmitters: a brief history, Br. J. Pharmacol. 147 (2006) S109–S119. [94] J. Wang, M. Yi, X. Chen, I. Muhammad, F. Liu, R. Li, J. Li, J. Li, Effects of colistin on amino acid neurotransmitters and blood-brain barrier in the mouse brain, Neurotoxicol. Teratol. 55 (2016) 32–37.

[62] S. Gonzalez-Reyes, S. Guzman-Beltran, O.N. Medina-Campos, J. Pedraza-Chaverri, Curcumin pretreatment induces Nrf2 and an antioxidant response and prevents hemin-induced toxicity in primary cultures of cerebellar granule neurons of rats, Oxid. Med. Cell. Longev. 2013 (2013) 801418. [63] M.D. Turner, B. Nedjai, T. Hurst, D.J. Pennington, Cytokines and chemokines: at the crossroads of cell signalling and inflammatory disease, Biochim. Biophys. Acta Mol. Cell Res. 1843 (2014) 2563–2582. [64] S.K. Biswas, Does the interdependence between oxidative stress and inflammation explain the antioxidant paradox? Oxidative Med. Cell. Longev. 9 (2016) 2016. [65] A.A. Elmarakby, J.C. Sullivan, Relationship between oxidative stress and inflammatory cytokines in diabetic nephropathy, Cardiovascular Therapeut. 30 (2012) 49–59. [66] T. Hussain, B. Tan, Oxidative Stress and Inflammation: what Polyphenols Can Do for Us? 2016 (2016) 7432797. [67] J.N. Sharma, A. Al-Omran, S.S. Parvathy, Role of nitric oxide in inflammatory diseases, Inflammopharmacology 15 (2007) 252–259. [68] V. Vielhauer, T.N. Mayadas, Functions of TNF and its receptors in renal disease: distinct roles in inflammatory tissue injury and immune regulation, Semin. Nephrol. 27 (2007) 286–308. [69] N. Parameswaran, S. Patial, Tumor necrosis factor-alpha signaling in macrophages, Crit. Rev. Eukaryot. Gene Expr. 20 (2010) 87–103. [70] G. Olmos, J. Llado, Tumor Necrosis Factor Alpha: a Link between Neuroinflammation and Excitotoxicity, 2014 Mediators of inflammation, 2014861231. [71] T.C. Barnes, M.E. Anderson, R.J. Moots, The many faces of Interleukin-6: the role of IL-6 in inflammation, vasculopathy, and fibrosis in systemic sclerosis, Int. J. Rheumatol. 2011 (2011) 721608. [72] T. Tanaka, M. Narazaki, T. Kishimoto, IL-6 in inflammation, immunity, and disease, Cold Spring Harbor Perspect. Biol. 6 (2014) a016295. [73] B.B. Aggarwal, S.C. Gupta, B. Sung, Curcumin: an orally bioavailable blocker of TNF and other pro-inflammatory biomarkers, Br. J. Pharmacol. 169 (2013) 1672–1692. [74] C. Dai, G.D. Ciccotosto, R. Cappai, S. Tang, D. Li, S. Xie, X. Xiao, T. Velkov, Curcumin attenuates colistin-induced neurotoxicity in N2a cells via anti-inflammatory activity, suppression of oxidative stress, and apoptosis, Mol. Neurobiol. 55 (2018) 421–434. [75] A. Gulcubuk, K. Altunatmaz, K. Sonmez, D. Haktanir-Yatkin, H. Uzun, A. Gurel, S. Aydin, Effects of curcumin on tumour necrosis factor-alpha and interleukin-6 in the late phase of experimental acute pancreatitis, J. Vet. Med. A, Physiol., Pathol., Clin. Med. 53 (2006) 49–54. [76] K.K. Jung, H.S. Lee, J.Y. Cho, W.C. Shin, M.H. Rhee, T.G. Kim, J.H. Kang, S.H. Kim, S. Hong, S.Y. Kang, Inhibitory effect of curcumin on nitric oxide production from lipopolysaccharide-activated primary microglia, Life Sci. 79 (2006) 2022–2031. [77] M.Y. Song, J.Y. Yim, J.M. Yim, I.J. Kang, H.W. Rho, H.S. Kim, H.Y. Yhim, N.R. Lee, E.K. Song, J.Y. Kwak, M.H. Sohn, C.Y. Yim, Use of curcumin to decrease nitric oxide production during the induction of antitumor responses by IL-2, J. Immunother. 34 (2011) 149–164.

64