DOI QR코드

DOI QR Code

Ginsenoside Rb1 ameliorates cisplatin-induced learning and memory impairments

  • Chen, Chen (Department of Neurology, The First Affiliated Hospital of Zhengzhou University) ;
  • Zhang, Haifeng (Department of Neurology, The First Affiliated Hospital of Zhengzhou University) ;
  • Xu, Hongliang (Department of Neurology, The First Affiliated Hospital of Zhengzhou University) ;
  • Zheng, Yake (Department of Neurology, The First Affiliated Hospital of Zhengzhou University) ;
  • Wu, Tianwen (Department of Neurology, The First Affiliated Hospital of Zhengzhou University) ;
  • Lian, Yajun (Department of Neurology, The First Affiliated Hospital of Zhengzhou University)
  • Received : 2017.04.24
  • Accepted : 2017.07.20
  • Published : 2019.10.15

Abstract

Background: Ginsenoside Rb1 (Rb1), a dominant component from the extract of Panax ginseng root, exhibits neuroprotective functions in many neurological diseases. This study was intended to investigate whether Rb1 can attenuate cisplatin-induced memory impairments and explore the potential mechanisms. Methods: Cisplatin was injected intraperitoneally with a dose of 5 mg/kg/wk, and Rb1 was administered in drinking water at the dose of 2 mg/kg/d to rats for 5 consecutive wk. The novel objects recognition task and Morris water maze were used to detect the memory of rats. Nissl staining was used to examine the neuron numbers in the hippocampus. The activities of superoxide dismutase, glutathione peroxidase, cholineacetyltransferase, acetylcholinesterase, and the levels of malondialdehyde, reactive oxygen species, acetylcholine, tumor necrosis factor-${\alpha}$, interleukin-$1{\beta}$, and interleukin-10 were measured by ELISA to assay the oxidative stress, cholinergic function, and neuroinflammation in the hippocampus. Results: Rb1 administration effectively ameliorates the memory impairments caused by cisplatin in both novel objects recognition task and Morris water maze task. Rb1 also attenuates the neuronal loss induced by cisplatin in the different regions (CA1, CA3, and dentate gyrus) of the hippocampus. Meanwhile, Rb1 is able to rescue the cholinergic neuron function, inhibit the oxidative stress and neuroinflammation in cisplatin-induced rat brain. Conclusion: Rb1 rescues the cisplatin-induced memory impairment via restoring the neuronal loss by reducing oxidative stress and neuroinflammation and recovering the cholinergic neuron functions.

Keywords

1. Introduction

Chemotherapy is an effective treatment for many types of cancer; however, several side effects of chemotherapy have also been reported. Among them, postchemotherapy cognitive impairment (PCCI) is the most important. It encompasses a wide range of neurological disorders, including the impairments in cognition, clarity of thought, attention, executive functioning, and information processing speed [1]. Approximately 20-30% of patients who undergo chemotherapy suffer from different level of PCCI [2]. In the USA, up to 3.9 million individuals with cancer and cancer treatments are living with persistent cognitive problems. Cognitive deficits that occur from cancer or its treatment vary and may be subtle or dramatic, temporary or permanent, and stable or progressive [3]. However, the underlying mechanisms of PCCI are still elusive, and no effective treatments are currently available.

Cisplatin, cis-diamminedichloridoplatinum(II), is a drug used for treating various types of tumors by injecting into a vein [4]. Cisplatin could reportedly induce apoptosis in cancer cells by cross linking DNA with the purine bases, impairing the repair mechanisms of DNA and inducing DNA damage. Although it is very effective in cancer therapy, the highly toxic effects of cisplatin, especially neurotoxicity, have been widely reported including the PCCI. In mice and rats, administration of cisplatin can induce severe cognitive malformations via different mechanisms [5], such as elevation of reactive oxygen species (ROS) [6], induction of neuronal loss by promoting apoptosis and inhibiting neurogenesis [7], and stimulation of neuroinflammation [8]. Therefore, treatment with antioxidants and anti-inflammation drugs may restore the inflammation factors and endogenous antioxidants to physiological levels and thereby prevent the neuronal loss and memory function induced by cisplatin.

Ginseng, also known as Korean or Asian ginseng, is the root of Panax ginseng Meyer. The benefits of ginseng in traditional oriental medicine have been widely recognized for over a thousand years. The major bioactive components in ginseng are ginsenosides, and their neuroprotective effect on different neurologic diseases has been studied [9,10]. Numerous studies have proven that ginsenosides can ameliorate learning/memory deficits in different brain diseases and during senescence in humans and animals [11,12]. There are many important components in ginsenosides; Rg1 and Rb1 are two of them with slightly varying molecular structures. According to previous studies, the beneficial effects of ginseng root on learning and memory are often attributed to Rb1 [13]. Previous studies showed that Rb1 stimulated neurite outgrowth [14] and promoted synaptic vesicle release by enhancing the phosphorylation of synapsins through a cyclic adenosine monophosphatedependent protein kinase pathway [15]. Additionally, Rb1 was suggested to prevent from either ischemia [16] or neurodegeneration induced by glutamate in hippocampal neurons [17]. However, whether Rb1 is also protective to the memory impairments induced by cisplatin is not known.

In this study, we assessed whether ginsenoside Rb1 could protect against the cognitive deficit induced by cisplatin. We found that oral administration of Rb1 effectively attenuated the memory impairments caused by cisplatin. Rb1 also rescued the neuronal loss and cholinergic dysfunction. Finally, we reported that Rb1 reduces the oxidative stress and neuroinflammation in the hippocampus.

2. Materials and methods

2.1. Animals and drugs

Wistar rats (male, 250 ± 20 g) were purchased from the Henan Provincial Laboratory Animal Center (Zhengzhou, Henan). They were housed on a 12-h light/dark cycle (light on at 7:00 AM) for 1 wk before use with food and water ad libitum. The experiment protocols were approved by Care and Use of Laboratory Animals of HeNan Province. In total, 40 rats were divided randomly into four groups: control (n = 10); cisplatin (Cisp, n = 10); cisplatin plus Rb1 (Cisp + Rb1, n = 10); and Rb1 (Rb1, n = 10). The control group rats were subjected with 0.2-mL normal saline once per wk via intraperitoneal (i.p.) injection. The Cisp group rats received 5 mg/kg/wk i.p. cisplatin. The Cisp + Rb1 group rats were administered cisplatin (5 mg/kg/wk, i.p.) plus Rb1 (2 mg/kg/d in drinking water). The Rb1 group rats received Rb1 (2 mg/kg/d in drinking water). The effective doses of cisplatin and Rb1 were chosen based on literatures [18-20] (Fig. 1).

Fig. 1. Time-line of experimental procedures of the whole study. After 7 d adaption, Wistar rats were administered ginsenoside Rb1 (2 mg/kg/d) in drinking water for 5 wk. Cisplatin (0.2 mL, 5 mg/kg) or saline (0.9%, 0.2 mL) was injected intraperitoneally on Day 7, Day 14, Day 21, Day 28, and Day 35. Next, the learning and memory abilities of rats were assessed by the novel object recognition task and Morris water maze. After the behavioral tasks, the rats were killed on Day 45. Five rats were fixed for Nissl staining, and the other rats were prepared for making hippocampal samples to detect the cholinergic function and biochemical biomarkers. i.p., intraperitoneal; MWM, Morris water maze; NORT, novel object recognition task; p.o., per os.

Ginsenoside Rb1 and cisplatin were obtained from SigmaAldrich (St. Louis, MO, USA). Some commercial kits were obtained from Nanjing Jiancheng Bioengineering Institute (Nanjing, Jiang Su, China), including superoxide dismutase (SOD) assay kit, malondialdehyde (MDA) assay kit, glutathione peroxidase (GSH-Px) assay kit, acetylcholine (ACh) assay kit, cholineacetyltransferase (ChAT) assay kit, acetylcholinesterase (AChE) assay kit, tumor necrosis factor-α (TNF-α) assay kit, interleukin-1β assay kit (IL-1β), and interleukin-10 assay kit (IL-10).

2.2. Novel object recognition task

The novel object recognition task (NORT) was performed, as described previously [21]. The equipment comprises a Plexiglas box (45 cm × 45 cm × 50 cm) with identical objects located in the opposite corners. In the habituation period, rats were put into the center of the empty box for 5 min without objects. The training session was performed 24 h after the habituation. Two identical objects (familiar objects, A and A') were placed in opposite corners, and the rats were allowed to explore for 5 min. The latency to approach each object was recorded. The testing phase was carried out after a 24-h retention interval, and rats were returned to the testing environment for 5 min where the familiar object (A') was replaced by a novel object (B), which differed from the familiar object in shape and texture. Measures of interaction were recorded as the amount of time that the animal spent with its head and nose oriented toward and within 2 cm of the object. For both training and testing phases, the exploration was operationally defined as an active event in which the rats were facing, sniffing, or pawing at the object. During the whole task, all objects were cleaned with 70% ethanol between each session. The object preference value was calculated by the formula: Tnovel/(Tnovel + Tfamiliar) × 100, Tfamiliar/ (Tnovel + Tfamiliar) × 100. The discrimination ratio was calculated as the exploring novel object time compared with the familiar object relative to the total time, as per the following formula: (Tnovel - Tfamiliar)/Ttotal [22,23].

2.3. Morris water maze

Morris water maze was used to assess the spatial learning and memory of rats. The maze was a circular (180 cm in diameter) water tank filled with water (20-21°C) up to a height of 40 cm. Briefly, rats were first trained to find a hidden platform in the target quadrant that was submerged in 1-2 cm of water by using a stationary array of cues outside the pool. Acquisition training in the hidden platform was conducted for 5 d consecutively including 20 trials. In each trial, the rat was placed into the pool at one of the four possible locations (randomly ordered). All rats were given 60 s free time to find the hidden platform, and the time spent to reach the escape platform was defined as escape latency. If the rat did not find the platform within 60 s, it was gently guided by the observer to the platform for 30 s. After rest for 1 d, a 60-s probe test was conducted in which the platform was removed. Both during the training and test period, the swimming pathways, percentage of time spent in the target quadrant, platform area crossings, and swimming speed were recorded and calculated using a computer software (Smart V3.0, Panlab Harvard Apparatus) [24-26].

2.4. Nissl staining

We used Nissl staining to detect the neuronal population of hippocampus. Five rats in each group were anesthetized and fixed by perfusion of 4% paraformaldehyde phosphate-buffered solution. After the perfusion, the brains were harvested and post-fixed for 24 h. The hippocampal tissues were cut into 25 mm slices using a vibrate microtome (VT1000s, Leica, Germany). The slices were placed in Nissl staining fluid for 5-10 min. The cell numbers of hippocampal CA1, CA3, and dentate gyrus (DG) regions were counted in the six consecutive sections using a grid-installed light microscopy (BX51; Olympus, Tokyo, Japan). Three visual fields of the hippocampus (CA1, CA3, and DG areas) were found in each slice at low magnification and were used as statistical areas for counting neurons with nuclei. The average of six sections for every region was taken as the final value for that region [27].

2.5. Preparation of hippocampal samples

The hippocampi were homogenized according to the method described in previous studies, with minor modifications [28,29]. The rats were killed following measurement of spatial memory. Next, the right hippocampi were separated and homogenized with 10 volumes of ice-cold 50mM Tris-HCl, pH 7.4, 2mM EDTA. The homogenates were centrifuged at 4,000 ×g for 10 min at 4°C, and the resulting supernatants were stored at -80°C for assaying the cholinergic function and biochemical assays, including AChE, ChAT, ACh, TNF-α, IL-1β, IL-10, SOD, MDA, GSH-Px, and ROS levels.

2.6. Analysis of cholinergic function

The activity of ChAT and AChE and the level of ACh in the hippocampal supernatant were measured using ChAT assay kit, AChE assay kit, and ACh assay kit, respectively, according to the manufacturer’s instructions.

2.7. Assays of oxidative damage

The activities of SOD and GSH-Px as well as the level of MDA were assayed according to the manufacturer’s instructions. The ROS content was measured by the products of 2',7'-dichlorofluorescein (DCFH)-DA, as described previously. After the transfer into cells, DCFH-DA is cleaved to form DCFH, which in turn is transformed into highly fluorescent DCF upon reaction with ROS. DCF was quantified in each sample using a fluorescence monitor [30].

2.8. Detection of inflammatory cytokines

The inflammatory cytokine levels in the hippocampus were assessed using TNF-α, IL-1β, and IL-10 assay kits according to the manufacturer’s instructions. The concentrations of these inflammatory cytokines were calculated according to the optical density determined spectrometrically using a micro ELISA reader.

2.9. Statistical analysis

Statistical analysis was performed using SPSS version 17.0 (SPSS Inc., Chicago, IL, USA). The differences between the groups were tested by ANOVA, followed by Fisher’s least significant difference post hoc test. For a single comparison, the significance of differences was determined by the t test. A value of p < 0.05 is considered to be statistically significant.

3. Results

3.1. Effect of ginsenoside Rb1 on cisplatin-induced object recognition memory impairment

After the completion of different treatment, rates were subjected to a series of behavioral tasks to assess their cognitive function. In the NORT test, as shown in Fig. 2, we found that control rats spent more time exploring the novel object than the familiar object (t18 = 6.12, p < 0.01; Fig. 2A). In contrast, cisplatin-treated rats could not distinguish between the novel and familiar objects (t18 = 1.08, p > 0.05). Rb1 treatment blocked cisplatin-induced memory impairment without affecting normal recognition function (Cisp, t18 = 3.86, p < 0.05; Rb1, t18 = 5.72, p < 0.01). The recognition index was significantly decreased in cisplatin-treated group, and this was restored by ginsenoside Rb1 [F(3,36) = 6.792, p < 0.01; Fig. 2B]. All rats spent a similar amount of time on object exploration during training and test sessions, indicating that the significant differences of each group were not due to the alternation of exploratory behavior on objects (Figs. 2C and 2D). These results suggest that ginsenoside Rb1 improves cisplatin-induced memory impairment.

Fig. 2. Effect of ginsenoside Rb1 on cisplatin (cisp)-induced object recognition memory impairment. (A) The amount of time that the animals spent exploring each object in test, which was performed 24 h after the training session. (B) Quantitative comparison of the recognition index in memory test session. (C) Total exploration time in training session. (D) Total exploration time in test session. Data were represented as mean  standard error of the mean. n ¼ 10, each group. * p < 0.05; ** p < 0.01.

3.2. Effect of Rb1 on cisplatin-induced spatial memory deficit

We also detected the spatial cognitive abilities of rats using Morris water maze. We found that the cisplatin group rats spent more time to find the hidden platform from the 4th d compared with the control in the training period; however, the Cisp + Rb1 group rats showed learning latency similar to that of control rats (Fig. 3A). During the probe test, the cisplatin group rats showed poor memory retention, spent less time in the target quadrant [F(1,18) = 10.66, p < 0.01; Fig. 3B], and less crossing times of the platform’s previous location [F(1,18) = 6.14, p < 0.05; Fig. 3C] as compared with the controls. Similarly, the learning and memory impairment induced by cisplatin was significantly arrested by ginsenoside Rb1 [F(1,18) = 6.14, p < 0.05]. The rats could remember the previous platform location and exhibit more exploring time in target quadrant [F(1,18) = 10.24, p < 0.05]. Throughout the tasks, the swimming speed of rats among each group showed no significant difference, indicating that the effect of drugs is on learning and memory rather than swimming ability (Fig. 3D).

Fig. 3. Effect of ginsenoside Rb1 on cisplatin (cisp)-induced spatial memory impairment. (A) Escape latency to find the platform during the training stages. (B) The time spent in target quadrant of probe trial. (C) The crossing numbers over the platform area. (D) The swimming speed of rats. All values are expressed as the mean  standard error of the mean (n ¼ 10). * p < 0.05; ** p < 0.01 versus control group; *** p < 0.05; **** p < 0.01 versus Cisp group.

3.3. Ginsenoside Rb1 promotes neuronal survival in the hippocampus

As we know, hippocampus is an important structure for learning and memory [31], and selective hippocampal lesions can induce place and cue memory impairment [32]. In this study, the behavioral tests showed most obvious cognition impairment in mice, and we thought these changes were related with hippocampus. The effect of cisplatin or/and ginsenoside Rb1 on the neuronal population of hippocampus was detected via Nissl staining. A large number of neuron cell bodies were seen at the CA1, CA3, and DG areas in hippocampi, distributed with big and round nuclei. The results of statistics revealed a significant reduction of hippocampal neuronal population in the cisplatin-treated group [F(3,12) = 4.46, p < 0.05]. The least significant difference post hoc test revealed a considerable reduction in the hippocampal neuronal population of cisplatin animals (p < 0.05). Ginsenoside Rb1 treatment increased the survival of neurons in the hippocampus of rats (p < 0.05), whereas ginsenoside Rb1 single treatment showed no significant effect compared with the controls (Fig. 4).

Fig. 4. Comparisons of the number of hippocampal neurons in hippocampus. (A) Nissl staining of hippocampal neurons of rats in each group (400). (B) Changes of hippocampal neuron numbers of rats in each group. Bar ¼ 50 mm. * p < 0.05 versus control group; ** p < 0.05 versus Cisp group. Cisp, cisplatin; DG, dentate gyrus.

3.4. Ginsenoside Rb1 rescues cholinergic dysfunction induced by cisplatin

Cholinergic dysfunction is a primary cause related to cognitive degeneration; therefore, we measured the ACh level and AChE and ChAT activities in the hippocampi of rats. From Fig. 5, we found that cisplatin caused a significant decrease in ACh levels (control, 18.68 ± 1.12 μg/mg; cisplatin, 10.69 ± 1.54 μg/mg, p < 0.01), increased AChE activity (control, 0.84 ± 0.05 U/mg; cisplatin, 1.192 ± 0.09 U/mg, p < 0.01), and a dramatic reduction in ChAT activity (control, 276.70 ± 14.71 U/mg; cisplatin, 209.26 ± 20.68 U/ mg, p < 0.01) in the hippocampus compared with the controls. These results verified that cisplatin impairs cholinergic function. The treatment of ginsenoside Rb1 could rescue these cholinergic abnormalities, as indicated by elevated ACh levels (16.57 ± 1.55 μg/ mg, p < 0.01), increased ChAT activity (269.42 ± 18.46 U/mg, p < 0.05), and reduced AChE activity (1.04 ± 0.04 U/mg, p < 0.01). Meanwhile, administration of ginsenoside Rb1 alone did not alter cholinergic dysfunction significantly. These data support the behavioral test results and suggest that ginsenoside Rb1 may exert its neuroprotection by rescuing cholinergic function.

Fig. 5. Ginsenoside Rb1 restores the cholinergic system dysfunctions. After the behavioral test, the cholinergic system function in hippocampus was assayed using (A) ACh, (B) AChE, and (C) ChAT kits. All values are expressed as the mean  standard deviation (n ¼ 5). * p < 0.05; ** p < 0.01 versus control group; *** p < 0.05; **** p < 0.01 versus Cisp group. ACh, acetylcholine; AChE, acetylcholinesterase; ChAT, cholineacetyltransferase; Cisp, cisplatin.

3.5. Ginsenoside Rb1 reduces oxidative stress in hippocampus

It is reported that cisplatin can interrupt the cholinergic neurotransmitter through trigger peroxidation and can cause memory impairment [33,34]; therefore, we measured the activity of SOD and GSH-Px as well as the levels of MDA and ROS in the brains of rats (Fig. 6). We found cisplatin treatment decreased the activities of SOD (control, 12.23 ± 0.41 U/mg; Cisp, 8.12 ± 1.12 U/mg, p < 0.01) and GSH-Px (control, 6.58 ± 0.31 U/mg; Cisp, 3.88 ± 0.25 U/mg, p < 0.01) compared with the treatment for control group, whereas ginsenoside Rb1 significantly increased the activities of SOD (10.22 ± 0.42 U/mg) and GSH-Px (4.91 ± 0.29 U/ mg) compared with the Cisp group (p < 0.05). MDA and ROS levels significantly increased in the brain tissue of the cisplatin group rats compared with the control group rats (p < 0.01); ginsenoside Rb1 administration ameliorated this increase (p < 0.05 and p < 0.01).

Fig. 6. Ginsenoside Rb1 suppresses the oxidative stress in hippocampus. (A) SOD activity, (B) GSH-Px activity, (C) MDA levels, and (D) ROS activity in the hippocampus were measured using commercial kits. Data are expressed as mean  standard deviation (n ¼ 5). * p < 0.05; ** p < 0.01 versus control group; *** p < 0.05; **** p < 0.01 versus Cisp group. Cisp, cisplatin; MDA, malondialdehyde; GSH-Px, glutathione peroxidase; ROS, reactive oxygen species; SOD, superoxide dismutase.

3.6. Ginsenoside Rb1 attenuates the cisplatin-induced inflammatory response

At last, we detected the changes in the hippocampus interleukin levels using ELISA kits. The results showed that cisplatin significantly enhanced the levels of TNF-α and IL-1β (p < 0.01), implying stronger inflammatory response, which was prevented by Rb1 treatment (p < 0.01 and p < 0.05; Figs. 7A and 7B). In contrast, the anti-inflammatory IL-10 level in the cisplatin group showed a decline compared with that in the control group (p < 0.01; Fig. 7C), and Rb1 partly arrested the effect of cisplatin. We did not find significant difference in all inflammatory factor levels between the rats treated with Ginsenoside Rb1 alone and the controls, neither.

Fig. 7. Ginsenoside Rb1 attenuates cisplatin-induced inflammation. Ginsenoside Rb1 attenuates the levels of (A) TNF-a, (B) IL-1b, and (C) IL-10. Data are expressed as mean  standard deviation (n ¼ 5). * p < 0.01 versus control group; ** p < 0.05; *** p < 0.01 versus Cisp group. Cisp, cisplatin; IL-1b, interleukin-1b; IL-10, interleukin-10; TNF-a, tumor necrosis factor-a.

4. Discussion

As a platinum-based compound, cisplatin was widely used as an anticancer chemotherapeutic agent and was reported to elicit a lot of side-effects, including hepatotoxicity [35], nephrotoxicity [36], ototoxicity [37], neurotoxicity [38], and nausea/vomiting [39]. Among them, neurotoxicity is particularly important. In this study, we first evaluated the learning and memory abilities by NORT and Morris water maze task. We reported that cisplatin-treated rats displayed lower discrimination ratio in the novel object recognition and longer swimming latency, and shorter duration and less crossing times in the Morris water maze task, suggesting impairment in the learning and memory induced by cisplatin. These findings are consistent with those of previous reports [18,40,41]. Meanwhile, we also observed that upon the treatment of cisplatin, the neuron numbers were reduced in the different subregions of hippocampus. In accordance to our finding, cisplatin was found to cause a dramatic decrement in the number of DCX-positive neurons in the DG region of the adult rat hippocampus when compared with vehicle-treated ones [42]. Furthermore, the hypernitration and LMO4 down-regulation play important roles in cisplatininduced neuronal apoptosis [43]. Thus, the loss of neurons upon cisplatin treatment may be due to the reduction of neurogenesis and promotion of neuron apoptosis. In our study, we also demonstrated that application of Rb1 rescued the memory impairments both in the novel object recognition and Morris water maze. Simultaneously, the neuron numbers were restored after the Rb1 treatment, suggesting neuroprotection of Rb1. As reported before, Rb1 was shown to inhibit cell death caused by kainic acid in both CA1 and CA3 regions of rat hippocampus and protected hippocampal neurons from ischemia [44]. Rb1 injection also increases brain-derived neurotrophic factor level, reduces casapase-3 activity, and promotes neurogenesis in the hippocampus [45]. Then, the application of Rb1 could both enhance the neurogenesis and protect against neuron apoptosis, which is effective in neuroprotection upon cisplatin insult.

The neurotoxic effects of cisplatin have been widely studied, and it is suggested that the overproduction of ROS [6], high levels of Pt-DNA binding and the following neuronal apoptosis are the main reasons [46]. It is known that ROS is mainly generated from the mitochondria, and exposure to cisplatin results in the increment of intracellular ROS in normal cells. For example, an in vivo study has shown that cisplatin exposure induces an increase in the activity of SOD and MDA, and also levels of H2O2, and a reduction in glutathione (GSH) and activity of GSH reductase in the cochlear [47]. Another in vitro study found that cisplatin could initiate ROS response via a mitochondria-dependent pathway by directly affecting the mitochondrial DNA. This impairs the synthesis of electron transport chain proteins and accelerates the nuclear DNA impairments, which leads to the enhancement of cytotoxic effect [48]. Thus, application of molecules with antioxidant property will be benefit to protect the neurotoxicity induced by cisplatin. Here, we also detected the oxidative stress level on cisplatin administration. A reduction in the activities of SOD and GSH-Px and elevation in the levels of MDA and ROS were found after the cisplatin treatment. This indicated that the overproduction of oxidative free radicals and inhibition of anti-oxidative enzymes in the hippocampus were induced by cisplatin. However, treatment of ginsenoside Rb1 significantly reduced the oxidative stress level as demonstrated by restoring the activities of SOD and GSH-Px, suppressing the MDA and ROS. In many previous studies, Rb1 had been shown to protect against oxidative insult in numerous neurological disorders [49]. The possible mechanism by which Rb1 protects against the oxidative insult is via Nrf2 pathway activation and Gb1/ PI3K/Akt pathway in ER-dependent manner and heme oxygenase-1 pathway [50,51].

In the current study, we also reported that Rb1 alleviates the cisplatin-induced neuroinflammation by reducing the levels of TNF-α and IL-1β and restoring the level of IL-10. In some previous studies, cisplatin was found to stimulate the generation of proinflammatory cytokines, such as TNF-α, IL-1β, IL-6, and transforming growth factor-β1 [52,53]. TNF-α is a prototypical inflammatory cytokine; it activates other cytokines [54] to mediate the cisplatininduced renal injury [55]. In a mice obesity model induced by high-fat diet, Rb1 supplement decreased the levels of TNF-α, IL-6, and/or IL-1β [56]. Additionally, the reduction of TNF-α expression by Rb1 could inhibit the severity of inflammatory response in collagen-induced arthritis mice [57]. It has also been shown that IL-10 acts as the anti-inflammatory cytokine to protect against acute renal injury induced by cisplatin [58,59]. And Rb1 treatment attenuated carbon tetrachloride-induced liver fibrosis by elevating IL-10 level in rats [60]. Thus, the Rb1 application could rebalance the inflammation and anti-inflammation factors in the hippocampus and then exert its neuroprotective effects. Furthermore, Rb1 administration here decreased the dysfunction of cholinergic neurons by restoring the ACh level, ChAT activity, and reducing the AChE activity. The physiological cholinergic signaling could modulate peripheral cytokine production by activation of the cholinergic anti-inflammatory pathway. Thus, the rescuing holinergic function also participates in the neuroprotection of Rb1 on cisplatin-induced neuroinflammation.

Conclusively, we investigated the neuroprotective effects of ginsenoside Rb1 on cisplatin-treated rats. We demonstrated that Rb1 markedly improved the cognitive decline and neuronal loss by inhibition of oxidative stress, rescuing the cholinergic function, and suppression of neuroinflammation in the hippocampus. Taken together, we concluded that Rb1 might be used as a potent neuroprotector against cisplatin-induced learning and memory impairments.

Conflicts of interest

The authors have declared that there is no conflict of interest.

References

  1. Argyriou AA, Assimakopoulos K, Iconomou G, Giannakopoulou F, Kalofonos HP. Either called “chemobrain” or “chemofog,” the long-term chemotherapy-induced cognitive decline in cancer survivors is real. J Pain Symptom Manage 2011;41:126-39. https://doi.org/10.1016/j.jpainsymman.2010.04.021
  2. Tannock IF, Ahles TA, Ganz PA, Van Dam FS. Cognitive impairment associated with chemotherapy for cancer: report of a workshop. J Clin Oncol 2004;22:2233-9. https://doi.org/10.1200/JCO.2004.08.094
  3. Schagen SB, Hamburger HL, Muller MJ, Boogerd W, van Dam FS. Neurophysiological evaluation of late effects of adjuvant high-dose chemotherapy on cognitive function. J Neurooncol 2001;51:159-65. https://doi.org/10.1023/A:1010635229762
  4. Stathopoulos GP. Liposomal cisplatin: a new cisplatin formulation. Anticancer Drugs 2010;21:732-6. https://doi.org/10.1097/CAD.0b013e32833d9adf
  5. Troy L, McFarland K, Littman-Power S, Kelly BJ, Walpole ET, Wyld D, Thomson D. Cisplatin-based therapy: a neurological and neuropsychological review. Psychooncology 2000;9:29-39. https://doi.org/10.1002/(SICI)1099-1611(200001/02)9:1<29::AID-PON428>3.0.CO;2-Z
  6. Ravi R, Somani SM, Rybak LP. Mechanism of cisplatin ototoxicity: antioxidant system. Pharmacol Toxicol 1995;76:386-94. https://doi.org/10.1111/j.1600-0773.1995.tb00167.x
  7. Winocur G, Wojtowicz JM, Tannock IF. Memory loss in chemotherapy-treated rats is exacerbated in high-interference conditions and related to suppression of hippocampal neurogenesis. Behav Brain Res 2015;281:239-44. https://doi.org/10.1016/j.bbr.2014.12.028
  8. Briones TL, Woods J. Dysregulation in myelination mediated by persistent neuroinflammation: possible mechanisms in chemotherapy-related cognitive impairment. Brain Behav Immun 2014;35:23-32. https://doi.org/10.1016/j.bbi.2013.07.175
  9. Kim J, Kim SH, Lee DS, Lee DJ, Kim SH, Chung S, Yang HO. Effects of fermented ginseng on memory impairment and beta-amyloid reduction in Alzheimer's disease experimental models. J Ginseng Res 2013;37:100-7. https://doi.org/10.5142/jgr.2013.37.100
  10. Zhao Z, Kim YW, Wu Y, Zhang J, Lee JH, Li X, Cho IJ, Park SM, Jung DH, Yang CH, et al. Korean Red Ginseng attenuates anxiety-like behavior during ethanol withdrawal in rats. J Ginseng Res 2014;38:256-63. https://doi.org/10.1016/j.jgr.2014.05.010
  11. Petkov VD, Kehayov R, Belcheva S, Konstantinova E, Petkov VV, Getova D, Markovska V. Memory effects of standardized extracts of Panax ginseng (G115), Ginkgo biloba (GK 501) and their combination Gincosan (PHL-00701). Planta Med 1993;59:106-14. https://doi.org/10.1055/s-2006-959623
  12. Attele AS, Wu JA, Yuan CS. Ginseng pharmacology: multiple constituents and multiple actions. Biochem Pharmacol 1999;58:1685-93. https://doi.org/10.1016/S0006-2952(99)00212-9
  13. Tawab MA, Bahr U, Karas M, Wurglics M, Schubert-Zsilavecz M. Degradation of ginsenosides in humans after oral administration. Drug Metab Dispos 2003;31:1065-71. https://doi.org/10.1124/dmd.31.8.1065
  14. Nishiyama N, Cho SI, Kitagawa I, Saito H. Malonylginsenoside Rb1 potentiates nerve growth factor (NGF)-induced neurite outgrowth of cultured chick embryonic dorsal root ganglia. Biol Pharm Bull 1994;17:509-13. https://doi.org/10.1248/bpb.17.509
  15. Xue JF, Liu ZJ, Hu JF, Chen H, Zhang JT, Chen NH. Ginsenoside Rb1 promotes neurotransmitter release by modulating phosphorylation of synapsins through a cAMP-dependent protein kinase pathway. Brain Res 2006;1106:91-8. https://doi.org/10.1016/j.brainres.2006.05.106
  16. Lim JH, Wen TC, Matsuda S, Tanaka J, Maeda N, Peng H, Aburaya J, Ishihara K, Sakanaka M. Protection of ischemic hippocampal neurons by ginsenoside Rb1, a main ingredient of ginseng root. Neurosci Res 1997;28:191-200. https://doi.org/10.1016/S0168-0102(97)00041-2
  17. Kim YC, Kim SR, Markelonis GJ, Oh TH. Ginsenosides Rb1 and Rg3 protect cultured rat cortical cells from glutamate-induced neurodegeneration. J Neurosci Res 1998;53:426-32. https://doi.org/10.1002/(SICI)1097-4547(19980815)53:4<426::AID-JNR4>3.0.CO;2-8
  18. Oz M, Nurullahoglu Atalik KE, Yerlikaya FH, Demir EA. Curcumin alleviates cisplatin-induced learning and memory impairments. Neurobiol Learn Mem 2015;123:43-9. https://doi.org/10.1016/j.nlm.2015.05.001
  19. Song XY, Hu JF, Chu SF, Zhang Z, Xu S, Yuan YH, Han N, Liu Y, Niu F, He X, et al. Ginsenoside Rg1 attenuates okadaic acid induced spatial memory impairment by the GSK3beta/tau signaling pathway and the Abeta formation prevention in rats. Eur J Pharmacol 2013;710:29-38. https://doi.org/10.1016/j.ejphar.2013.03.051
  20. Golchin L, Shabani M, Harandi S, Razavinasab M. Pistachio supplementation attenuates motor and cognition impairments induced by cisplatin or vincristine in rats. Adv Biomed Res 2015;4:92. https://doi.org/10.4103/2277-9175.156656
  21. Ennaceur A, Delacour J. A new one-trial test for neurobiological studies of memory in rats. 1: Behavioral data. Behav Brain Res 1988;31:47-59. https://doi.org/10.1016/0166-4328(88)90157-X
  22. Kim DH, Choi SM, Jho J, Park MS, Kang J, Park SJ, Ryu JH, Jo J, Kim HH, Kim BC. Infliximab ameliorates AD-associated object recognition memory impairment. Behav Brain Res 2016;311:384-91. https://doi.org/10.1016/j.bbr.2016.06.001
  23. Fernandez F, Morishita W, Zuniga E, Nguyen J, Blank M, Malenka RC, Garner CC. Pharmacotherapy for cognitive impairment in a mouse model of Down syndrome. Nat Neurosci 2007;10:411-3. https://doi.org/10.1038/nn1860
  24. Tabari SS, Babri S, Mirzaie F, Farajdokht F, Mohaddes G. Enduring amnesia induced by ICV scopolamine is reversed by sesame oil in male rats. Acta Cir Bras 2016;31:520-6. https://doi.org/10.1590/S0102-865020160080000004
  25. Zhang W, Bai M, Xi Y, Hao J, Liu L, Mao N, Su C, Miao J, Li Z. Early memory deficits precede plaque deposition in APPswe/PS1dE9 mice: involvement of oxidative stress and cholinergic dysfunction. Free Radic Biol Med 2012;52:1443-52. https://doi.org/10.1016/j.freeradbiomed.2012.01.023
  26. Liu D, Tang H, Li XY, Deng MF, Wei N, Wang X, Zhou YF, Wang DQ, Fu P, Wang JZ, et al. Targeting the HDAC2/HNF-4A/miR-101b/AMPK pathway rescues tauopathy and dendritic abnormalities in alzheimer's disease. Mol Ther 2017;25:752-64. https://doi.org/10.1016/j.ymthe.2017.01.018
  27. Hu J, Huang HZ, Wang X, Xie AJ, Wang X, Liu D, Wang JZ, Zhu LQ. Activation of glycogen synthase kinase-3 mediates the olfactory deficit-induced hippocampal impairments. Mol Neurobiol 2015;52:1601-17. https://doi.org/10.1007/s12035-014-8953-9
  28. Ray RS, Rai S, Katyal A. Cholinergic receptor blockade by scopolamine and mecamylamine exacerbates global cerebral ischemia induced memory dysfunction in C57BL/6J mice. Nitric Oxide 2014;43:62-73. https://doi.org/10.1016/j.niox.2014.08.009
  29. Abd-El-Fattah MA, Abdelakader NF, Zaki HF. Pyrrolidine dithiocarbamate protects against scopolamine-induced cognitive impairment in rats. Eur J Pharmacol 2014;723:330-8. https://doi.org/10.1016/j.ejphar.2013.11.008
  30. Tang J, Yuan Y, Wei C, Liao X, Yuan J, Nanberg E, Zhang Y, Bornehag C-G, Yang X. Neurobehavioral changes induced by di (2-ethylhexyl) phthalate and the protective effects of vitamin E in Kunming mice. Toxicol Res 2015;4:1006-15. https://doi.org/10.1039/C4TX00250D
  31. Eichenbaum H. Hippocampus: cognitive processes and neural representations that underlie declarative memory. Neuron 2004;44:109-20. https://doi.org/10.1016/j.neuron.2004.08.028
  32. Jarrard LE. Selective hippocampal lesions and behavior: effects of kainic acid lesions on performance of place and cue tasks. Behav Neurosci 1983;97:873-89. https://doi.org/10.1037/0735-7044.97.6.873
  33. Lee JS, Kim HG, Han JM, Kim DW, Yi MH, Son SW, Kim YA, Lee JS, Choi MK, Son CG. Ethanol extract of Astragali Radix and Salviae Miltiorrhizae Radix, Myelophil, exerts anti-amnesic effect in a mouse model of scopolamineinduced memory deficits. J Ethnopharmacol 2014;153:782-92. https://doi.org/10.1016/j.jep.2014.03.048
  34. Shi J, Liu Q, Wang Y, Luo G. Coadministration of huperzine A and ligustrazine phosphate effectively reverses scopolamine-induced amnesia in rats. Pharmacol Biochem Behav 2010;96:449-53. https://doi.org/10.1016/j.pbb.2010.07.004
  35. Waseem M, Bhardwaj M, Tabassum H, Raisuddin S, Parvez S. Cisplatin hepatotoxicity mediated by mitochondrial stress. Drug Chem Toxicol 2015;38:452-9. https://doi.org/10.3109/01480545.2014.992437
  36. Loehrer PJ, Einhorn LH. Drugs five years later. Cisplatin. Ann Intern Med 1984;100:704-13. https://doi.org/10.7326/0003-4819-100-5-704
  37. Rybak LP, Mukherjea D, Jajoo S, Ramkumar V. Cisplatin ototoxicity and protection: clinical and experimental studies. Tohoku J Exp Med 2009;219:177-86. https://doi.org/10.1620/tjem.219.177
  38. Milosavljevic N, Duranton C, Djerbi N, Puech PH, Gounon P, Lagadic-Gossmann D, Dimanche-Boitrel MT, Rauch C, Tauc M, Counillon L, et al. Nongenomic effects of cisplatin: acute inhibition of mechanosensitive transporters and channels without actin remodeling. Cancer Res 2010;70:7514-22. https://doi.org/10.1158/0008-5472.CAN-10-1253
  39. Liaw CC, Wang CH, Chang HK, Kao CY, Huang JS. Prevention of acute and delayed cisplatin-induced nausea and vomiting with intravenous ondansetron plus intravenous dexamethasone. Chang Gung Med J 2000;23:413-9.
  40. Shabani M, Larizadeh MH, Parsania S, Hajali V, Shojaei A. Evaluation of destructive effects of exposure to cisplatin during developmental stage: no profound evidence for sex differences in impaired motor and memory performance. Int J Neurosci 2012;122:439-48. https://doi.org/10.3109/00207454.2012.673515
  41. Chiu GS, Maj MA, Rizvi S, Dantzer R, Vichaya EG, Laumet G, Kavelaars A, Heijnen CJ. Pifithrin-mu prevents cisplatin-induced chemobrain by preserving neuronal mitochondrial function. Cancer Res 2017;77:742-52. https://doi.org/10.1158/0008-5472.CAN-16-1817
  42. Dietrich J, Han R, Yang Y, Mayer-Proschel M, Noble M. CNS progenitor cells and oligodendrocytes are targets of chemotherapeutic agents in vitro and in vivo. J Biol 2006;5:22. https://doi.org/10.1186/jbiol50
  43. Rathinam R, Ghosh S, Neumann WL, Jamesdaniel S. Cisplatin-induced apoptosis in auditory, renal, and neuronal cells is associated with nitration and downregulation of LMO4. Cell Death Discov 2015;1:15052. https://doi.org/10.1038/cddiscovery.2015.52
  44. Lee JH, Kim SR, Bae CS, Kim D, Hong H, Nah S. Protective effect of ginsenosides, active ingredients of Panax ginseng, on kainic acid-induced neurotoxicity in rat hippocampus. Neurosci Lett 2002;325:129-33. https://doi.org/10.1016/S0304-3940(02)00256-2
  45. Gao XQ, Yang CX, Chen GJ, Wang GY, Chen B, Tan SK, Liu J, Yuan QL. Ginsenoside Rb1 regulates the expressions of brain-derived neurotrophic factor and caspase-3 and induces neurogenesis in rats with experimental cerebral ischemia. J Ethnopharmacol 2010;132:393-9. https://doi.org/10.1016/j.jep.2010.07.033
  46. Ta LE, Espeset L, Podratz J, Windebank AJ. Neurotoxicity of oxaliplatin and cisplatin for dorsal root ganglion neurons correlates with platinum-DNA binding. Neurotoxicology 2006;27:992-1002. https://doi.org/10.1016/j.neuro.2006.04.010
  47. Dehne N, Lautermann J, Petrat F, Rauen U, de Groot H. Cisplatin ototoxicity: involvement of iron and enhanced formation of superoxide anion radicals. Toxicol Appl Pharmacol 2001;174:27-34. https://doi.org/10.1006/taap.2001.9171
  48. Marullo R, Werner E, Degtyareva N, Moore B, Altavilla G, Ramalingam SS, Doetsch PW. Cisplatin induces a mitochondrial-ROS response that contributes to cytotoxicity depending on mitochondrial redox status and bioenergetic functions. PLoS One 2013;8:e81162. https://doi.org/10.1371/journal.pone.0081162
  49. Ahmed T, Raza SH, Maryam A, Setzer WN, Braidy N, Nabavi SF, de Oliveira MR, Nabavi SM. Ginsenoside Rb1 as a neuroprotective agent: a review. Brain Res Bull 2016;125:30-43. https://doi.org/10.1016/j.brainresbull.2016.04.002
  50. Ni N, Liu Q, Ren H, Wu D, Luo C, Li P, Wan JB, Su H. Ginsenoside Rb1 protects rat neural progenitor cells against oxidative injury. Molecules 2014;19:3012-24. https://doi.org/10.3390/molecules19033012
  51. Hwang YP, Jeong HG. Ginsenoside Rb1 protects against 6-hydroxydopamineinduced oxidative stress by increasing heme oxygenase-1 expression through an estrogen receptor-related PI3K/Akt/Nrf2-dependent pathway in human dopaminergic cells. Toxicol Appl Pharmacol 2010;242:18-28. https://doi.org/10.1016/j.taap.2009.09.009
  52. Miller RP, Tadagavadi RK, Ramesh G, Reeves WB. Mechanisms of cisplatin nephrotoxicity. Toxins (Basel) 2010;2:2490-518. https://doi.org/10.3390/toxins2112490
  53. Faubel S, Lewis EC, Reznikov L, Ljubanovic D, Hoke TS, Somerset H, Oh DJ, Lu L, Klein CL, Dinarello CA, et al. Cisplatin-induced acute renal failure is associated with an increase in the cytokines interleukin (IL)-1beta, IL-18, IL-6, and neutrophil infiltration in the kidney. J Pharmacol Exp Ther 2007;322:8-15. https://doi.org/10.1124/jpet.107.119792
  54. Amirshahrokhi K, Ghazi-khansari M, Mohammadi-Farani A, Karimian G. Effect of captopril on TNF-alpha and IL-10 in the livers of bile duct ligated rats. Iran J Immunol 2010;7:247-51.
  55. Ramesh G, Reeves WB. TNF-alpha mediates chemokine and cytokine expression and renal injury in cisplatin nephrotoxicity. J Clin Invest 2002;110:835-42. https://doi.org/10.1172/JCI200215606
  56. Wu Y, Yu Y, Szabo A, Han M, Huang XF. Central inflammation and leptin resistance are attenuated by ginsenoside Rb1 treatment in obese mice fed a high-fat diet. PLoS One 2014;9:e92618. https://doi.org/10.1371/journal.pone.0092618
  57. Kim HA, Kim S, Chang SH, Hwang HJ, Choi YN. Anti-arthritic effect of ginsenoside Rb1 on collagen induced arthritis in mice. Int Immunopharmacol 2007;7:1286-91. https://doi.org/10.1016/j.intimp.2007.05.006
  58. Deng J, Kohda Y, Chiao H, Wang Y, Hu X, Hewitt SM, Miyaji T, McLeroy P, Nibhanupudy B, Li S, et al. Interleukin-10 inhibits ischemic and cisplatininduced acute renal injury. Kidney Int 2001;60:2118-28. https://doi.org/10.1046/j.1523-1755.2001.00043.x
  59. Tadagavadi RK, Reeves WB. Endogenous IL-10 attenuates cisplatin nephrotoxicity: role of dendritic cells. J Immunol 2010;185:4904-11. https://doi.org/10.4049/jimmunol.1000383
  60. Hou YL, Tsai YH, Lin YH, Chao JC. Ginseng extract and ginsenoside Rb1 attenuate carbon tetrachloride-induced liver fibrosis in rats. BMC Complement Altern Med 2014;14:415. https://doi.org/10.1186/1472-6882-14-415

Cited by

  1. Neuroprotective Properties of Asiatic Acid against 5-Fluorouracil Chemotherapy in the Hippocampus in an Adult Rat Model vol.10, pp.8, 2019, https://doi.org/10.3390/nu10081053
  2. Active ginseng components in cognitive impairment: Therapeutic potential and prospects for delivery and clinical study vol.9, pp.71, 2018, https://doi.org/10.18632/oncotarget.26035
  3. Effect of curcumin nanoparticles on the cisplatin-induced neurotoxicity in rat vol.42, pp.2, 2019, https://doi.org/10.1080/01480545.2018.1504058
  4. Traditional Chinese Medicine Jiuwei Zhenxin Granules in Treating Depression: An Overview vol.16, 2019, https://doi.org/10.2147/ndt.s273324
  5. Pro-Resolving Effect of Ginsenosides as an Anti-Inflammatory Mechanism of Panax ginseng vol.10, pp.3, 2020, https://doi.org/10.3390/biom10030444
  6. Preventive Effect of Muscone against Cisplatin Nephrotoxicity in LLC-PK1 Cells vol.10, pp.10, 2019, https://doi.org/10.3390/biom10101444
  7. Antioxidant Supplementation in the Treatment of Neurotoxicity Induced by Platinum-Based Chemotherapeutics—A Review vol.21, pp.20, 2020, https://doi.org/10.3390/ijms21207753
  8. A Proliferation-Inducing Ligand Regulation in Polymorphonuclear Neutrophils by Panax ginseng vol.68, pp.6, 2019, https://doi.org/10.1007/s00005-020-00597-z
  9. Peripherally administered cisplatin activates a parvocellular neuronal subtype expressing arginine vasopressin and enhanced green fluorescent protein in the paraventricular nucleus of a transgenic rat vol.70, pp.1, 2020, https://doi.org/10.1186/s12576-020-00764-z
  10. Protective effect of ginsenoside Rb1 against chronic restraint stress (CRS)-induced memory impairments in rats vol.405, 2021, https://doi.org/10.1016/j.bbr.2021.113146
  11. Ginsenoside from ginseng: a promising treatment for inflammatory bowel disease vol.73, pp.3, 2019, https://doi.org/10.1007/s43440-020-00213-z
  12. The promising therapeutic potentials of ginsenosides mediated through p38 MAPK signaling inhibition vol.7, pp.11, 2019, https://doi.org/10.1016/j.heliyon.2021.e08354
  13. Molecular docking, pharmacokinetic studies, and in vivo pharmacological study of indole derivative 2-(5-methoxy-2-methyl-1H-indole-3-yl)-N′-[(E)-(3-nitrophenyl) methylidene] acetohydrazide as a vol.11, pp.1, 2019, https://doi.org/10.1038/s41598-021-84748-y
  14. Nucleosides rich extract from Cordyceps cicadae alleviated cisplatin-induced neurotoxicity in rats: A behavioral, biochemical and histopathological study vol.15, pp.1, 2022, https://doi.org/10.1016/j.arabjc.2021.103476
  15. Total saponins from stems and leaves of Panax quinquefolius L. ameliorate podophyllotoxin‐induced myelosuppression and gastrointestinal toxicity vol.36, pp.2, 2019, https://doi.org/10.1002/bmc.5266