Next Article in Journal
Botulinum Toxin as a Pain Killer: Players and Actions in Antinociception
Next Article in Special Issue
Repetitive Treatment with Diluted Bee Venom Attenuates the Induction of Below-Level Neuropathic Pain Behaviors in a Rat Spinal Cord Injury Model
Previous Article in Journal / Special Issue
Therapeutic Effects of Bee Venom on Immunological and Neurological Diseases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Analgesic Effects of Bee Venom Derived Phospholipase A2 in a Mouse Model of Oxaliplatin-Induced Neuropathic Pain

1
Department of Physiology, College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdamoon-gu, Seoul 130-701, Korea
2
Department of East-West Medicine, Graduate School, Kyung Hee University, 26 Kyungheedae-ro, Dongdamoon-gu, Seoul 130-701, Korea
3
Department of Herbology, College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdamoon-gu, Seoul 130-701, Korea
*
Author to whom correspondence should be addressed.
Toxins 2015, 7(7), 2422-2434; https://doi.org/10.3390/toxins7072422
Submission received: 15 May 2015 / Revised: 11 June 2015 / Accepted: 23 June 2015 / Published: 29 June 2015

Abstract

:
A single infusion of oxaliplatin, which is widely used to treat metastatic colorectal cancer, induces specific sensory neurotoxicity signs that are triggered or aggravated when exposed to cold or mechanical stimuli. Bee Venom (BV) has been traditionally used in Korea to treat various pain symptoms. Our recent study demonstrated that BV alleviates oxaliplatin-induced cold allodynia in rats, via noradrenergic and serotonergic analgesic pathways. In this study, we have further investigated whether BV derived phospholipase A2 (bvPLA2) attenuates oxaliplatin-induced cold and mechanical allodynia in mice and its mechanism. The behavioral signs of cold and mechanical allodynia were evaluated by acetone and a von Frey hair test on the hind paw, respectively. The significant allodynia signs were observed from one day after an oxaliplatin injection (6 mg/kg, i.p.). Daily administration of bvPLA2 (0.2 mg/kg, i.p.) for five consecutive days markedly attenuated cold and mechanical allodynia, which was more potent than the effect of BV (1 mg/kg, i.p.). The depletion of noradrenaline by an injection of N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine hydrochloride (DSP4, 50 mg/kg, i.p.) blocked the analgesic effect of bvPLA2, whereas the depletion of serotonin by injecting DL-p-chlorophenylalanine (PCPA, 150 mg/kg, i.p.) for three successive days did not. Furthermore, idazoxan (α2-adrenegic receptor antagonist, 1 mg/kg, i.p.) completely blocked bvPLA2-induced anti-allodynic action, whereas prazosin (α1-adrenegic antagonist, 10 mg/kg, i.p.) did not. These results suggest that bvPLA2 treatment strongly alleviates oxaliplatin-induced acute cold and mechanical allodynia in mice through the activation of the noradrenergic system, via α2-adrenegic receptors, but not via the serotonergic system.

1. Introduction

Oxaliplatin is an effective platinum derivative, which is widely used in the treatment of colorectal carcinoma [1,2], but causes neurotoxicity predominantly within the peripheral nervous system [3,4]. Two different types of oxaliplatin-induced peripheral neuropathy have been described hitherto, i.e., cold and mechanical hypersensitivity [5,6]. However, effective treatment for oxaliplatin-induced cold and mechanical hypersensitivity still remains to be elucidated. Hence, it is required to discover therapeutic options for the management of oxaliplatin-induced neuropathic pain.
Bee Venom (BV) has been traditionally used in Korea to relieve pain and to treat chronic inflammatory diseases [7,8,9,10,11,12]. Previous studies have demonstrated that the analgesic effects of BV in various pain models are mediated mainly by activation of α2-adrenergic and/or serotonergic receptors [12,13,14,15,16]. Rho and his colleagues have reported that subcutaneous injections of BV attenuated heat hyperalgesia and cold and mechanical allodynia in the rats with nerve injury-induced neuropathic pain through the activation of the endogenous noradrenergic system [17,18]. In a rat model of oxaliplatin-induced neuropathic pain, we found that the anti-allodynic effect of BV is at least partially mediated by the noradrenergic and serotonergic system, but not by the opioid system [9,16].
Phospholipase A2 from BV (bvPLA2), a prototypic group III enzyme that hydrolyzes fatty acids in membrane phospholipids, is one of the major active components of BV [19,20]. Several studies have shown that this bvPLA2 prevents neuronal cell death and spinal cord injury [21,22]. Our previous study demonstrated that BV mitigates cisplatin-induced nephrotoxicity [23] and found that bvPLA2 can reduce such nephrotoxicity more potently than BV [24]. However, the effect of PLA2 on oxaliplatin-induced neuropathic pain and its mechanism have not been studied yet.
The aim of this study was to evaluate and compare the analgesic effect of BV and bvPLA2 on oxaliplatin-induced cold and mechanical allodynia in mice. In addition, we examined whether the anti-allodynic effect of bvPLA2 is mediated by the serotonergic or noradrenergic pain inhibitory system.

2. Results

2.1. Effects of BV and bvPLA2 on Oxaliplatin-Induced Cold and Mechanical Hypersensitivity

First, we investigated the effects of a single administration of oxaliplatin (6 mg/kg, i.p.) on behavioral sensitivity to cold and mechanical stimuli in mice (Figure 1). The administration significantly increased the frequency of licking and shaking of the hind paw in response to cold acetone stimuli. A significant cold allodynia was observed at day 1, peaked at day 3 and lasted for at least seven days after the oxaliplatin administration, compared to the vehicle group (Figure 1a). Similarly, an administration of oxaliplatin significantly increased the withdrawal responses of the hind paw to von Frey filament applications (as expressed % of withdrawal response) at day 1, peaked at day 3–4 and maintained up to day 5 (Figure 1b).
Figure 1. Effects of BV and bvPLA2 on oxaliplatin-induced cold and mechanical allodynia in mice. The behavioral tests for cold (a) and mechanical (b) allodynia were performed before (PRE) and after the administration of oxaliplatin (6 mg/kg, i.p.). Vehicle (5% glucose) + PBS, Oxaliplatin + PBS, Oxaliplatin + BV, and Oxaliplatin + bvPLA2 groups received daily injection of PBS, BV (1 mg/kg, i.p.) or bvPLA2 (0.2 mg/kg, i.p.) for five days after an oxaliplatin or vehicle administration. Results are expressed as mean ± SEM; n = 6 mice/group; The data was analyzed with one-way ANOVA followed by the Tukey’s multiple comparison test. ** p < 0.01, *** p < 0.001, vs. Vehicle + PBS; # p < 0.05, ## p < 0.01, ### p < 0.001 vs. Oxaliplatin + PBS; $ p < 0.05, $$ p < 0.01, $$$ p < 0.001, vs. Oxaliplatin + BV.
Figure 1. Effects of BV and bvPLA2 on oxaliplatin-induced cold and mechanical allodynia in mice. The behavioral tests for cold (a) and mechanical (b) allodynia were performed before (PRE) and after the administration of oxaliplatin (6 mg/kg, i.p.). Vehicle (5% glucose) + PBS, Oxaliplatin + PBS, Oxaliplatin + BV, and Oxaliplatin + bvPLA2 groups received daily injection of PBS, BV (1 mg/kg, i.p.) or bvPLA2 (0.2 mg/kg, i.p.) for five days after an oxaliplatin or vehicle administration. Results are expressed as mean ± SEM; n = 6 mice/group; The data was analyzed with one-way ANOVA followed by the Tukey’s multiple comparison test. ** p < 0.01, *** p < 0.001, vs. Vehicle + PBS; # p < 0.05, ## p < 0.01, ### p < 0.001 vs. Oxaliplatin + PBS; $ p < 0.05, $$ p < 0.01, $$$ p < 0.001, vs. Oxaliplatin + BV.
Toxins 07 02422 g001
Daily BV treatment (1 mg/kg, i.p.) for five consecutive days significantly reduced the cold allodynia from three days after the oxaliplatin administration and such analgesic effect last up to day 5. In addition, bvPLA2 treatment (0.2 mg/kg, i.p.) significantly attenuated the cold allodynia from day 1 after the oxaliplatin injection and such effect endured at least for the following six days (Figure 1a). BV treatment also significantly attenuated mechanical allodynia from day 2 after the oxaliplatin injection and this BV analgesia was continued up to day 4. Moreover, bvPLA2 treatment showed a significant reduction in mechanical allodynia from day 2 after the oxaliplatin injection and such effect lasted up to day 5 (Figure 1b). The relieving effects of bvPLA2 on oxaliplatin-induced cold and mechanical allodynia were significantly more potent than those of BV (Figure 1).

2.2. Effects of BvPLA2 on Oxaliplatin-Induced Cold and Mechanical Allodynia in Serotonin Depleted Mice

We investigated the effects of bvPLA2 on oxaliplatin-induced cold and mechanical allodynia in serotonin depleted mice by injecting DL-p-chlorophenylalanine (PCPA, 150 mg/kg, i.p.) for three successive days [25,26]. PCPA pretreatment itself did not affect the behavioral signs of cold and mechanical allodynia induced by oxaliplatin (p > 0.05, Oxaliplatin + PBS + PCPA [n = 6] vs. Oxaliplatin + PBS + NS [n = 4], Cold (frequency): 1.83 ± 0.04 vs. 1.92 ± 0.05 at 0d, 4.61 ± 0.07 vs. 4.67 ± 0.07 at 3d, 3.83 ± 0.06 vs. 3.79 ± 0.08 at 5d, 3.25 ± 0.07 vs. 3.04 ± 0.10 at 7d; Mechanical (%): 30.00 ± 1.83 vs. 30.00 ± 0.00 at 0d, 94.17 ± 0.83 vs. 93.75 ± 1.25 at 3d, 72.03 ± 1.54 vs. 68.75 ± 1.25 at 5d, 50 ± 2.24 vs. 51.25 ± 1.25 at 7d). Thus, we pooled the data from the two groups as a control group (Oxaliplatin + PBS + PCPA/NS, Figure 2). Compared to this control group, bvPLA2 treatment in mice without serotonin depletion (Oxaliplatin + PLA2 + NS group) significantly attenuated the cold and mechanical hypersensitivity (p < 0.01 at days 3 and 5). Such anti-allodynic effects of bvPLA2 were not blocked by PCPA pretreatment (Figure 2), indicating that the serotonergic mechanism is not involved in the analgesic effect of bvPLA2 on oxaliplatin-induced neuropathic pain.
Figure 2. Effects of bvPLA2 on oxaliplatin-induced cold and mechanical allodynia in serotonin depleted mice. The behavioral tests for cold (a) and mechanical (b) allodynia were performed before and after an administration of oxaliplatin (Oxa, 6 mg/kg, i.p.). Serotonin was depleted by daily injections of PCPA (150 mg/kg, i.p.) for three consecutive days prior to an oxaliplatin administration. Oxa + PBS + PCPA/NS, n = 10; Other groups, n = 6 mice/group; Results are expressed as mean ± SEM; N.S, no significance (p > 0.05), The data were analyzed with one-way ANOVA followed by the Tukey’s multiple comparison test.
Figure 2. Effects of bvPLA2 on oxaliplatin-induced cold and mechanical allodynia in serotonin depleted mice. The behavioral tests for cold (a) and mechanical (b) allodynia were performed before and after an administration of oxaliplatin (Oxa, 6 mg/kg, i.p.). Serotonin was depleted by daily injections of PCPA (150 mg/kg, i.p.) for three consecutive days prior to an oxaliplatin administration. Oxa + PBS + PCPA/NS, n = 10; Other groups, n = 6 mice/group; Results are expressed as mean ± SEM; N.S, no significance (p > 0.05), The data were analyzed with one-way ANOVA followed by the Tukey’s multiple comparison test.
Toxins 07 02422 g002

2.3. Noradrenergic Mechanism of the Anti-Allodynic Effects of BvPLA2 in Oxaliplatin-Administered Mice

We evaluated the effects of bvPLA2 on oxaliplatin-induced allodynia in noradrenaline depleted mice by a pretreatment of DSP4 [27]. The anti-allodynic effects of bvPLA2 (Oxa + PLA2 + NS group, p < 0.01 vs. control Oxa + PBS + DSP4/NS group at days 3, 5 and 7) were significantly blocked by DSP4 pretreatment (Figure 3), unlike the aforementioned PCPA pretreatment. These results suggest that activation of the noradrenergic pain inhibitory pathway at least partially mediates the bvPLA2-induced anti-allodynic action in oxaliplatin-administered mice.
Figure 3. Effects of bvPLA2 on oxaliplatin-induced cold and mechanical allodynia in noradrenaline depleted mice. The behavioral tests for cold (a) and mechanical (b) allodynia were performed before and after an administration of oxaliplatin (6 mg/kg, i.p.). Noradrenaline was depleted by an injection of DSP4 (50 mg/kg, i.p.) at a day before an oxaliplatin administration. Since DSP4 pretreatment itself did not affect the cold and mechanical allodynia signs induced by oxaliplatin (p > 0.05, Oxa + PBS + DSP4 [n = 6] vs. Oxa + PBS + NS [n = 6]), we pooled the data from the two groups as a control (Oxa + PBS + DSP4/NS, n = 12). Other groups, n = 6 mice/group; Results are expressed as mean ± SEM; N.S, no significance (p > 0.05), * p < 0.05, ** p < 0.01, *** p < 0.001, The data was analyzed with one-way ANOVA followed by the Tukey’s multiple comparison test.
Figure 3. Effects of bvPLA2 on oxaliplatin-induced cold and mechanical allodynia in noradrenaline depleted mice. The behavioral tests for cold (a) and mechanical (b) allodynia were performed before and after an administration of oxaliplatin (6 mg/kg, i.p.). Noradrenaline was depleted by an injection of DSP4 (50 mg/kg, i.p.) at a day before an oxaliplatin administration. Since DSP4 pretreatment itself did not affect the cold and mechanical allodynia signs induced by oxaliplatin (p > 0.05, Oxa + PBS + DSP4 [n = 6] vs. Oxa + PBS + NS [n = 6]), we pooled the data from the two groups as a control (Oxa + PBS + DSP4/NS, n = 12). Other groups, n = 6 mice/group; Results are expressed as mean ± SEM; N.S, no significance (p > 0.05), * p < 0.05, ** p < 0.01, *** p < 0.001, The data was analyzed with one-way ANOVA followed by the Tukey’s multiple comparison test.
Toxins 07 02422 g003
Figure 4. Effect of α2-adrenergic receptor antagonist, idazoxan, on bvPLA2-induced anti-allodynic action in oxaliplatin-administered mice. The behavioral tests for cold (a) and mechanical (b) allodynia were performed before and after an administration of oxaliplatin (6 mg/kg, i.p.). Idazoxan (IDA, 1 mg/kg, i.p.) was administered 30 min prior to bvPLA2 injection. Since IDA pretreatment itself did not affect the cold and mechanical allodynia signs induced by oxaliplatin (p > 0.05, Oxa + PBS + IDA [n = 6] vs. Oxa + PBS + NS [n = 6]), we pooled the data from the two groups as a control (Oxa + PBS + IDA/NS, n = 12). Other groups, n = 6 mice/group; Results are expressed as mean ± SEM; * p < 0.05, ** p < 0.01, *** p < 0.001, The data was analyzed with one-way ANOVA followed by the Tukey’s multiple comparison test.
Figure 4. Effect of α2-adrenergic receptor antagonist, idazoxan, on bvPLA2-induced anti-allodynic action in oxaliplatin-administered mice. The behavioral tests for cold (a) and mechanical (b) allodynia were performed before and after an administration of oxaliplatin (6 mg/kg, i.p.). Idazoxan (IDA, 1 mg/kg, i.p.) was administered 30 min prior to bvPLA2 injection. Since IDA pretreatment itself did not affect the cold and mechanical allodynia signs induced by oxaliplatin (p > 0.05, Oxa + PBS + IDA [n = 6] vs. Oxa + PBS + NS [n = 6]), we pooled the data from the two groups as a control (Oxa + PBS + IDA/NS, n = 12). Other groups, n = 6 mice/group; Results are expressed as mean ± SEM; * p < 0.05, ** p < 0.01, *** p < 0.001, The data was analyzed with one-way ANOVA followed by the Tukey’s multiple comparison test.
Toxins 07 02422 g004
To identify which adrenergic receptor subtype mediates the analgesic effects of bvPLA2 on oxaliplatin-induced neuropathic pain in mice, we examined the effect of prazosin (PRA, α1-adrenergic receptor antagonist) or idazoxan (IDA, α2-adrenergic receptor antagonist) on the bvPLA2-induced anti-allodynic action. As shown in Figure 4, IDA (1 mg/kg, i.p.) significantly blocked the relieving effect of bvPLA2 on oxaliplatin-induced cold and mechanical allodynia. However, PRA (10 mg/kg, i.p.) did not affect the bvPLA2 effect, because there were no significant differences in cold and mechanical sensitivity between the Oxa + PLA2 + NS and Oxa + PLA2 + PRA groups (p > 0.05, Table 1). These results indicate that bvPLA2 treatment alleviates oxaliplatin-induced acute cold and mechanical allodynia in mice via activation of α2-adrenergic receptors, but not α1-adrenergic receptors.
Table 1. Effect of α1-adrenergic receptor antagonist, prazosin (PRA) on bvPLA2-induced anti-allodynic action in oxaliplatin-administered mice.
Table 1. Effect of α1-adrenergic receptor antagonist, prazosin (PRA) on bvPLA2-induced anti-allodynic action in oxaliplatin-administered mice.
Post-Oxaliplatin DayD0D3D5D7
Acetone test Frequency
Oxa + PBS + PRA/NS2.1 ± 0.044.4 ± 0.083.6 ± 0.153.3 ± 0.09
Oxa + PLA2 + NS2.0 ± 0.063.5 ± 0.15 ***3.0 ± 0.10 **2.6 ± 0.10 ***
Oxa + PLA2 + PRA2.1 ± 0.063.1 ± 0.12 ***3.1 ± 0.09 *2.8 ± 0.07 **
von Frey test <0.4 g> % withdrawalresponse
Oxa + PBS + PRA/NS40.8 ± 0.8376.7 ± 1.6755.8 ± 1.5444.2 ± 1.54
Oxa + PLA2 + NS40.0 ± 1.8359.2 ± 2.39 ***48.1 ± 1.29 **41.5 ± 1.12
Oxa + PLA2 + PRA40.8 ± 2.0157.5 ± 1.71 ***47.5 ± 1.71 **40.33 ± 1.67
Oxa + PBS + PRA/NS, n = 12; Other groups, n = 6 mice/group; Results are expressed as mean ± SEM; * p < 0.05, ** p < 0.01, *** p < 0.001, vs. Oxa + PBS + PRA/NS, one-way ANOVA followed by the Tukey’s multiple comparison test.

3. Discussion

Oxaliplatin-induced neuropathic pain represents a major obstacle to successful cancer treatment as it restricts both individual and cumulative dosages. However, despite these limitations, it is widely used and many patients suffer from the development of long-lasting consequences (i.e., peripheral neuropathy) [28,29]. Patients may also experience cold-induced paresthesias, throat and jaw tightness, and occasionally focal weakness [30]. Oxaliplatin is structurally similar to other platinum based chemotherapy drugs, such as cisplatin and carboplatin. They all have neurotoxicity, however oxaliplatin has little nephrotoxicity and hematotoxicity [31]. It has been shown that oxaliplatin-induced acute neuropathy is characterized by a specific somatosensory profile, i.e., cold and mechanical hypersensitivity [32]. Therefore, cold and mechanical hypersensitivity is a hallmark of oxaliplatin-induced neuropathy. Indeed, in this study, a single intraperitoneal administration of oxaliplatin (6 mg/kg) significantly increased the cold and mechanical sensitivity in mice, corroborating the previous reports using rats [33,34]. This mouse model might help in exploring the molecular and genetic mechanism of oxaliplatin-induced neuropathic pain in the future studies, since transgenic and gene knock-out/knock-in animals have been developed primarily in the mouse [35,36,37].
There have been few reports about the effective treatment and prevention of oxaliplatin-induced neuropathic pain. However, our previous study has shown that BV has a beneficial role in reliving oxaliplatin-induced neuropathic pain symptoms in rats, suggesting that BV could be an alternative therapeutic option for the management of oxaliplatin-induced peripheral neuropathy [9,16]. Interestingly, we recently found that bvPLA2 mitigates cisplatin-induced nephrotoxicity and acetaminophen-induced hepatotoxicity, which was more potent than BV [24,38]. In the present study, we investigated for the first time whether bvPLA2 has an analgesic effect on oxaliplain-induced cold and mechanical allodynia. Our data showed that the treatment of bvPLA2 significantly alleviated the allodynia in oxaliplatin-administered mice and such bvPLA2 effect was superior to the BV effect. This powerful analgesic effect of bvPLA2 led us to investigate the underlying mechanisms.
The extensive data support a role for the monoamine neurotransmitters (i.e., serotonin and noradrenaline) and opioids, in the modulation of pain [39,40]. Serotonin and noradrenaline have been implicated as principal mediators of endogenous analgesic mechanisms in the descending pain pathways [39]. Our previous study suggested that the anti-allodynic effect of BV on oxaliplatin-induced cold allodynia in rats involves the noradrenergic, but not opioid, system [9]. In addition, spinal 5-HT3 receptors play an important role in the BV-induced anti-allodynic action in oxaliplatin-injected rats [16]. In contrast, the present study clearly showed that serotonin depletion by pretreatment of PCPA did not significantly affect the anti-allodynic effects of bvPLA2 in oxaliplatin-administered mice. This result suggests that the serotonergic inhibitory pathway is not involved in the analgesic effect of bvPLA2 on oxaliplatin-induced cold and mechanical hypersensitivity, unlike the case of BV. Other active components of BV, such as apamin [41], might be responsible for the serotonergic mechanism of BV-induced analgesia. However, in noradrenaline-depleted mice, the suppressive effect of bvPLA2 on oxaliplatin-induced cold and mechanical allodynia was significantly prevented. These results indicate that the noradrenergic analgesic system is at least partially involved in the analgesic effects of bvPLA2 in oxaliplatin-administered mice.
α-adrenergic receptors have been demonstrated to play an important role in the modulation of cold allodynia via the noradrenergic pain inhibitory system [42]. Previous studies showed that either α1- [43,44] or α2-adrenergic receptors [45,46] are responsible for the adrenergic sensitivity of nerve injury-induced neuropathic rats. A dual contribution of α1- and α2-adrenergic receptors to neuropathic pain was also suggested [47,48]. Also other articles have elucidated that the analgesic effects of BV are mediated mainly by activation of α2-adrenergic receptors in various pain models, such as nerve injury-induced neuropathic pain, acetic acid-induced visceral pain and inflammatory pain [12,13,14,15]. Our recent study also showed that α2-adrenergic receptors mediate the anti-allodynic effect of BV in oxaliplatin-induced neuropathic pain in rats [9]. In the present study, we further examined which adrenergic receptor subtype mediates the effects of bvPLA2 on cold and mechanical allodynia in oxaliplatin-administered mice. The current results showed that IDA (α2-adrenergic antagonist) was able to completely block the anti-allodynic effect of bvPLA2 on oxaliplatin-induced neuropathic pain in mice, whereas PRA (α1-adrenergic antagonist) did not. These results indicate that the noradrenergic mechanism of the analgesic effect of bvPLA2 on oxaliplatin-induced neuropathic pain is mediated by activation of α2-adrenergic, but not α1-adrenergic, receptors.
In this study, we have clearly shown that bvPLA2 treatment strongly alleviates oxaliplatin-induced acute cold and mechanical allodynia in mice through the activation of the noradrenergic system, via α2-adrenegic receptors. Besides such action through the neurochemical mechanism, bvPLA2 is known to have a potent immune modulatory effect. The major pathway of the bvPLA2-induced immune modulation is to increase peripheral regulatory T cells, which play a key role in the maintenance of tolerance in the immune system. Our recent studies showed that such strategies using bvPLA2 could be successful in the prevention of cisplatin-induced acute kidney and acetaminophen-induced acute liver injury, by suppressing immune response via the modulation of regulatory T cells [24,38]. Another previous study also demonstrated that regulatory T cells attenuate peripheral nerve injury-induced neuropathic pain in rats [49]. Thus, it would be of high interest to see if bvPLA2 treatment before an oxaliplatin administration prevents the development of neuropathic cold and mechanical allodynia by regulating peripheral immune response. To our best knowledge, there are no clinical trials for bvPLA2 treatment. Further research is required in this field. In addition, our current work is limited to behavioral and pharmacological approaches. Molecular and genetic studies using the advantage of the mouse model are now in progress to elucidate more detailed mechanism of bvPLA2-induced analgesia.

4. Experimental Section

4.1. Animals

Male C57BL/6 mice (6–8 weeks old) were purchased from the Daehan Biolink (Chungbuk, Korea). They were kept under specific pathogen-free conditions with air conditioning and a 12 h light/dark cycle. The mice had free access to food and water during the experiments. The study was approved by the Kyung Hee University Animal Care and Use Committee (KHUASP(SE)-15-024).

4.2. Behavioral Tests

Behavioral tests representing different sensory components of neuropathic pain were conducted before and after an oxaliplatin administration. The mice were habituated to handling by investigators and to all testing procedures for a week before the start of the experiments. The experimenters were blind to oxaliplatin and any treatments.
Cold sensitivity was measured by an acetone test [50]. Mice were placed in a clear plastic box (12 × 8 × 6 cm) with a wire mesh floor and allowed to habituate for 30 min prior to the testing. Acetone (10 μL, Reagents Chemical Ltd., Gyonggi-do, Korea) was sprayed onto the plantar skin of each hind paw 3 times and the frequency of licking and shaking of the affected paw was counted after the acetone spray for 30 s. The advantage of acetone test is that it is quite simple, economical, and that the assessment for multiple mice can be made in a short period of time. The disadvantage is that acetone can also induce some behavioral response in naïve mice. However, mice having allodynia (e.g., oxaliplatin-administered mice) show a significant increase in the level of response to acetone when compared to the control mice.
Mechanical sensitivity was measured by the von Frey hair test [51]. Mice were placed in a clear plastic box (12 × 8 × 6 cm) with a wire mesh floor and allowed to habituate for 30 min before the testing. A von Frey filament (Linton Instrumentation, Norfolk, UK) with a bending force of 0.4 g were applied to the midplantar skin (avoiding the base of the tori) of each hind paw 10 times, with each application held for 3 s [52]. The number of withdrawal responses to the von Frey filament applications from both hind paws were counted and then expressed as an overall percentage response.

4.3. Oxaliplatin Administration and BV/bvPLA2 Treatment

Oxaliplatin (Sigma, St. Louis, MO, USA) was dissolved in a 5% glucose solution at a concentration of 2 mg/mL and was intraperitoneally (i.p.) injected at 6 mg/kg [9,33]. The vehicle control group received the same volume of a 5% glucose solution through the same injection route.
BV (1 mg/kg, i.p., Sigma) or bvPLA2 (0.2 mg/kg, i.p., Sigma) [9,24,38] dissolved in phosphate buffered saline (PBS) was injected in oxaliplatin-administered mice for a continuous five days. Cold and mechanical sensitivity were measured by acetone test and von Frey hair test, respectively, before each bvPLA2 or BV treatment. Control group was treated intraperitoneally with PBS.

4.4. Depletion of Serotonin or Noradrenaline

DL-p-chlorophenylalanine (PCPA, Sigma, an inhibitor of serotonin synthesis, 150 mg/kg/day) or vehicle (normal saline, NS) was injected intraperitoneally to mice prior to oxaliplatin administration for three days. The dosage and treatment course of PCPA have been widely used to deplete 5-HT stores [25,26]. After 5-HT depletion with PCPA, oxaliplatin and bvPLA2 were administered as aforementioned.
N-(2-Chloroethyl)-N-ethyl-2-bromobenzylamine hydrochloride (DSP4, TOCRIS, 50 mg/kg) or vehicle (NS) was injected intraperitoneally to mice a day before an oxaliplatin administration. DSP4 at a concentration of 50 mg/kg has been shown to be an effective dose for maximal NE depletion [53], with the advantage that mice did not require special care following injection as no adverse effects could be observed.

4.5. α1- or α2-Adrenergic Receptor Antagonist

To test which adrenergic receptor subtype mediates the anti-allodynic effects of bvPLA2 in oxaliplatin-administered mice, specific antagonists were administered intraperitoneally 30 min prior to bvPLA2 treatement for five days: α1-adrenergic receptor antagonist (prazosin, 10 mg/kg, Sigma), α2-adrenergic receptor antagonist (idazoxan, 1 mg/kg, Sigma). The dose of each antagonist was determined based on the previous studies showing the selective and effective antagonistic action against adrenergic receptor-mediated responses [54,55,56].

4.6. Statistical Analyses

The data are presented as mean ± SEM and were analyzed by the unpaired t-test or one-way ANOVA followed by the Tukey’s multiple comparison test to determine the statistical differences among the groups. p < 0.05 was considered as statistically significant.

5. Conclusions

In conclusion, our findings reveal that cold and mechanical sensitivity were significantly increased after a single injection of oxaliplatin in mice. BV and bvPLA2 can exert significant relieving effects on oxaliplatin-induced cold and mechanical hypersensitivity, in which bvPLA2 is more potent than BV. The serotonergic mechanism is not involved in the analgesic effect of bvPLA2 on oxaliplatin-induced neuropathic pain, whereas the noradrenergic pain inhibitory system at least partially mediates the bvPLA2 effect. Finally, we demonstrated that bvPLA2 treatment alleviates oxaliplatin-induced acute cold and mechanical allodynia in mice via activation of α2-adrenergic receptors. These findings may provide clinically useful evidence for the application of bvPLA2 in the management of peripheral neuropathic pain that occurs after the oxaliplatin administration.

Acknowledgments

This research was supported by a grant of the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (grant number: HI14 C0738).

Author Contributions

Hyunsu Bae and Sun Kwang Kim conceived and designed the study. Dongxing Li and Younju Lee performed the experiments. Dongxing Li, Woojin Kim, Kyungjin Lee and Sun Kwang Kim analyzed the data. Dongxing Li, Younju Lee, Woojin Kim, Kyungjin Lee and Sun Kwang Kim wrote the manuscript. All authors have read and approved the final manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Andre, T.; Boni, C.; Mounedji-Boudiaf, L.; Navarro, M.; Tabernero, J.; Hickish, T.; Topham, C.; Zaninelli, M.; Clingan, P.; Bridgewater, J.; et al. Oxaliplatin, fluorouracil, and leucovorin as adjuvant treatment for colon cancer. N. Engl. J. Med. 2004, 350, 2343–2351. [Google Scholar] [CrossRef] [PubMed]
  2. De Gramont, A.; Figer, A.; Seymour, M.; Homerin, M.; Hmissi, A.; Cassidy, J.; Boni, C.; Cortes-Funes, H.; Cervantes, A.; Freyer, G.; et al. Leucovorin and fluorouracil with or without oxaliplatin as first-line treatment in advanced colorectal cancer. J. Clin. Oncol. 2000, 18, 2938–2947. [Google Scholar] [PubMed]
  3. Cassidy, J.; Misset, J.L. Oxaliplatin-related side effects: Characteristics and management. Semin. Oncol. 2002, 29, 11–20. [Google Scholar] [CrossRef] [PubMed]
  4. Extra, J.M.; Marty, M.; Brienza, S.; Misset, J.L. Pharmacokinetics and safety profile of oxaliplatin. Semin. Oncol. 1998, 25, 13–22. [Google Scholar] [PubMed]
  5. Pasetto, L.M.; D’Andrea, M.R.; Rossi, E.; Monfardini, S. Oxaliplatin-related neurotoxicity: How and why? Crit. Rev. Oncol. Hematol. 2006, 59, 159–168. [Google Scholar] [CrossRef] [PubMed]
  6. Grothey, A. Oxaliplatin-safety profile: Neurotoxicity. Semin. Oncol. 2003, 30, 5–13. [Google Scholar] [CrossRef]
  7. Billingham, M.E.; Morley, J.; Hanson, J.M.; Shipolini, R.A.; Vernon, C.A. Letter: An anti-inflammatory peptide from bee venom. Nature 1973, 245, 163–164. [Google Scholar] [CrossRef] [PubMed]
  8. Choi, M.S.; Park, S.; Choi, T.; Lee, G.; Haam, K.K.; Hong, M.C.; Min, B.I.; Bae, H. Bee venom ameliorates ovalbumin induced allergic asthma via modulating CD4+CD25+ regulatory T cells in mice. Cytokine 2013, 61, 256–265. [Google Scholar] [CrossRef] [PubMed]
  9. Lim, B.S.; Moon, H.J.; Li, D.X.; Gil, M.; Min, J.K.; Lee, G.; Bae, H.; Kim, S.K.; Min, B.I. Effect of bee venom acupuncture on oxaliplatin-induced cold allodynia in rats. Evid. Based Complement. Alternat. Med. 2013, 2013, 369324. [Google Scholar] [CrossRef] [PubMed]
  10. Yoon, S.Y.; Yeo, J.H.; Han, S.D.; Bong, D.J.; Oh, B.; Roh, D.H. Diluted bee venom injection reduces ipsilateral mechanical allodynia in oxaliplatin-induced neuropathic mice. Biol. Pharm. Bull. 2013, 36, 1787–1793. [Google Scholar] [CrossRef] [PubMed]
  11. Somerfield, S.D.; Brandwein, S. Bee venom and adjuvant arthritis. J. Rheumatol. 1988, 15, 1878. [Google Scholar] [PubMed]
  12. Roh, D.H.; Kwon, Y.B.; Kim, H.W.; Ham, T.W.; Yoon, S.Y.; Kang, S.Y.; Han, H.J.; Lee, H.J.; Beitz, A.J.; Lee, J.H. Acupoint stimulation with diluted bee venom (apipuncture) alleviates thermal hyperalgesia in a rodent neuropathic pain model: Involvement of spinal alpha 2-adrenoceptors. J. Pain 2004, 5, 297–303. [Google Scholar] [CrossRef] [PubMed]
  13. Baek, Y.H.; Huh, J.E.; Lee, J.D.; Choi do, Y.; Park, D.S. Antinociceptive effect and the mechanism of bee venom acupuncture (apipuncture) on inflammatory pain in the rat model of collagen-induced arthritis: Mediation by alpha2-adrenoceptors. Brain Res. 2006, 1073–1074, 305–310. [Google Scholar] [CrossRef] [PubMed]
  14. Kwon, Y.B.; Kang, M.S.; Han, H.J.; Beitz, A.J.; Lee, J.H. Visceral antinociception produced by bee venom stimulation of the Zhongwan acupuncture point in mice: Role of alpha(2) adrenoceptors. Neurosci. Lett. 2001, 308, 133–137. [Google Scholar] [CrossRef]
  15. Kim, H.W.; Kwon, Y.B.; Han, H.J.; Yang, I.S.; Beitz, A.J.; Lee, J.H. Antinociceptive mechanisms associated with diluted bee venom acupuncture (apipuncture) in the rat formalin test: Involvement of descending adrenergic and serotonergic pathways. Pharmacol. Res. 2005, 51, 183–188. [Google Scholar] [CrossRef] [PubMed]
  16. Lee, J.H.; Li, D.X.; Yoon, H.; Go, D.; Quan, F.S.; Min, B.I.; Kim, S.K. Serotonergic mechanism of the relieving effect of bee venom acupuncture on oxaliplatin-induced neuropathic cold allodynia in rats. BMC Complement. Altern. Med. 2014, 14, 471. [Google Scholar] [CrossRef] [PubMed]
  17. Kang, S.Y.; Roh, D.H.; Park, J.H.; Lee, H.J.; Lee, J.H. Activation of spinal alpha2-adrenoceptors using diluted bee venom stimulation reduces cold allodynia in neuropathic pain rats. Evid. Based Complement. Altern. Med. 2012, 2012, 784713. [Google Scholar] [CrossRef] [PubMed]
  18. Kang, S.Y.; Roh, D.H.; Yoon, S.Y.; Moon, J.Y.; Kim, H.W.; Lee, H.J.; Beitz, A.J.; Lee, J.H. Repetitive treatment with diluted bee venom reduces neuropathic pain via potentiation of locus coeruleus noradrenergic neuronal activity and modulation of spinal NR1 phosphorylation in rats. J. Pain 2012, 13, 155–166. [Google Scholar] [CrossRef] [PubMed]
  19. Monti, M.C.; Casapullo, A.; Santomauro, C.; D’Auria, M.V.; Riccio, R.; Gomez-Paloma, L. The molecular mechanism of bee venom phospholipase A2 inactivation by bolinaquinone. Chembiochem 2006, 7, 971–980. [Google Scholar] [CrossRef] [PubMed]
  20. Zhao, H.; Kinnunen, P.K. Modulation of the activity of secretory phospholipase A2 by antimicrobial peptides. Antimicrob. Agents Chemother. 2003, 47, 965–971. [Google Scholar] [CrossRef] [PubMed]
  21. Jeong, J.K.; Moon, M.H.; Bae, B.C.; Lee, Y.J.; Seol, J.W.; Park, S.Y. Bee venom phospholipase A2 prevents prion peptide induced-cell death in neuronal cells. Int. J. Mol. Med. 2011, 28, 867–873. [Google Scholar] [PubMed]
  22. Lopez-Vales, R.; Ghasemlou, N.; Redensek, A.; Kerr, B.J.; Barbayianni, E.; Antonopoulou, G.; Baskakis, C.; Rathore, K.I.; Constantinou-Kokotou, V.; Stephens, D.; et al. Phospholipase A2 superfamily members play divergent roles after spinal cord injury. FASEB J. 2011, 25, 4240–4252. [Google Scholar] [CrossRef] [PubMed]
  23. Burrage, P.S.; Mix, K.S.; Brinckerhoff, C.E. Matrix metalloproteinases: Role in arthritis. Front. Biosci. 2006, 11, 529–543. [Google Scholar] [CrossRef] [PubMed]
  24. Kim, H.; Lee, H.; Lee, G.; Jang, H.; Kim, S.-S.; Yoon, H.; Kang, G.-H.; Hwang, D.-S.; Kim, S.K.; Chung, H.-S.; et al. Phospholipase A2 inhibits cisplatin-induced acute kidney injury by modulating regulatory T cells via CD206 mannose receptor. Kidney Int. 2015, 10. [Google Scholar] [CrossRef]
  25. Zhu, J.X.; Zhu, X.Y.; Owyang, C.; Li, Y. Intestinal serotonin acts as a paracrine substance to mediate vagal signal transmission evoked by luminal factors in the rat. J. Physiol. 2001, 530, 431–442. [Google Scholar] [CrossRef] [PubMed]
  26. Maleki, N.; Nayebi, A.M.; Garjani, A. Effects of central and peripheral depletion of serotonergic system on carrageenan-induced paw oedema. Int. Immunopharmacol. 2005, 5, 1723–1730. [Google Scholar] [CrossRef] [PubMed]
  27. Kudo, T.; Kushikata, T.; Kudo, M.; Hirota, K. Antinociceptive effects of neurotropin in a rat model of central neuropathic pain: DSP-4 induced noradrenergic lesion. Neurosci. Lett. 2011, 503, 20–22. [Google Scholar] [CrossRef] [PubMed]
  28. Farquhar-Smith, P. Chemotherapy-induced neuropathic pain. Curr. Opin. Support. Palliat. Care 2011, 5, 1–7. [Google Scholar] [CrossRef] [PubMed]
  29. Kannarkat, G.; Lasher, E.E.; Schiff, D. Neurologic complications of chemotherapy agents. Curr. Opin. Neurol. 2007, 20, 719–725. [Google Scholar] [CrossRef] [PubMed]
  30. Lehky, T.J.; Leonard, G.D.; Wilson, R.H.; Grem, J.L.; Floeter, M.K. Oxaliplatin-induced neurotoxicity: Acute hyperexcitability and chronic neuropathy. Muscle Nerve 2004, 29, 387–392. [Google Scholar] [CrossRef] [PubMed]
  31. Desoize, B.; Madoulet, C. Particular aspects of platinum compounds used at present in cancer treatment. Crit. Rev. Oncol. Hematol. 2002, 42, 317–325. [Google Scholar] [CrossRef]
  32. Binder, A.; Stengel, M.; Maag, R.; Wasner, G.; Schoch, R.; Moosig, F.; Schommer, B.; Baron, R. Pain in oxaliplatin-induced neuropathy––Sensitisation in the peripheral and central nociceptive system. Eur. J. Cancer 2007, 43, 2658–2663. [Google Scholar] [CrossRef] [PubMed]
  33. Ling, B.; Coudore-Civiale, M.A.; Balayssac, D.; Eschalier, A.; Coudore, F.; Authier, N. Behavioral and immunohistological assessment of painful neuropathy induced by a single oxaliplatin injection in the rat. Toxicology 2007, 234, 176–184. [Google Scholar] [CrossRef] [PubMed]
  34. Ling, B.; Coudore, F.; Decalonne, L.; Eschalier, A.; Authier, N. Comparative antiallodynic activity of morphine, pregabalin and lidocaine in a rat model of neuropathic pain produced by one oxaliplatin injection. Neuropharmacology 2008, 55, 724–728. [Google Scholar] [CrossRef] [PubMed]
  35. De Felipe, C.; Herrero, J.F.; O’Brien, J.A.; Palmer, J.A.; Doyle, C.A.; Smith, A.J.; Laird, J.M.; Belmonte, C.; Cervero, F.; Hunt, S.P. Altered nociception, analgesia and aggression in mice lacking the receptor for substance P. Nature 1998, 392, 394–397. [Google Scholar] [PubMed]
  36. Honore, P.; Rogers, S.D.; Schwei, M.J.; Salak-Johnson, J.L.; Luger, N.M.; Sabino, M.C.; Clohisy, D.R.; Mantyh, P.W. Murine models of inflammatory, neuropathic and cancer pain each generates a unique set of neurochemical changes in the spinal cord and sensory neurons. Neuroscience 2000, 98, 585–598. [Google Scholar] [CrossRef]
  37. Back, S.K.; Sung, B.; Hong, S.K.; Na, H.S. A mouse model for peripheral neuropathy produced by a partial injury of the nerve supplying the tail. Neurosci. Lett. 2002, 322, 153–156. [Google Scholar] [CrossRef]
  38. Kim, H.; Keum, D.J.; Kwak, J.; Chung, H.S.; Bae, H. Bee venom phospholipase A2 protects against acetaminophen-induced acute liver injury by modulating regulatory T cells and IL-10 in mice. PLoS ONE 2014, 9, e114726. [Google Scholar] [CrossRef] [PubMed]
  39. Lamont, L.A.; Tranquilli, W.J.; Grimm, K.A. Physiology of pain. Vet. Clin. N. Am. Small Anim. Pract. 2000, 30, 703–728. [Google Scholar] [CrossRef]
  40. Ossipov, M.H.; Morimura, K.; Porreca, F. Descending pain modulation and chronification of pain. Curr. Opin. Support. Palliat. Care 2014, 8, 143–151. [Google Scholar] [PubMed]
  41. Crespi, F. Apamin increases 5-HT cell firing in raphe dorsalis and extracellular 5-HT levels in amygdala: A concomitant in vivo study in anesthetized rats. Brain Res. 2009, 1281, 35–46. [Google Scholar] [CrossRef] [PubMed]
  42. Millan, M.J. Descending control of pain. Prog. Neurobiol. 2002, 66, 355–474. [Google Scholar] [CrossRef]
  43. Korenman, E.M.; Devor, M. Ectopic adrenergic sensitivity in damaged peripheral nerve axons in the rat. Exp. Neurol. 1981, 72, 63–81. [Google Scholar] [CrossRef]
  44. Lee, D.H.; Liu, X.; Kim, H.T.; Chung, K.; Chung, J.M. Receptor subtype mediating the adrenergic sensitivity of pain behavior and ectopic discharges in neuropathic Lewis rats. J. Neurophysiol. 1999, 81, 2226–2233. [Google Scholar] [PubMed]
  45. Leem, J.W.; Gwak, Y.S.; Nam, T.S.; Paik, K.S. Involvement of alpha2-adrenoceptors in mediating sympathetic excitation of injured dorsal root ganglion neurons in rats with spinal nerve ligation. Neurosci. Lett. 1997, 234, 39–42. [Google Scholar] [CrossRef]
  46. Zhang, J.M.; Song, X.J.; LaMotte, R.H. An in vitro study of ectopic discharge generation and adrenergic sensitivity in the intact, nerve-injured rat dorsal root ganglion. Pain 1997, 72, 51–57. [Google Scholar] [CrossRef]
  47. Hord, A.H.; Denson, D.D.; Stowe, B.; Haygood, R.M. Alpha-1 and alpha-2 adrenergic antagonists relieve thermal hyperalgesia in experimental mononeuropathy from chronic constriction injury. Anesth. Analg. 2001, 92, 1558–1562. [Google Scholar] [CrossRef] [PubMed]
  48. Tracey, D.J.; Cunningham, J.E.; Romm, M.A. Peripheral hyperalgesia in experimental neuropathy: Mediation by alpha 2-adrenoreceptors on post-ganglionic sympathetic terminals. Pain 1995, 60, 317–327. [Google Scholar] [CrossRef]
  49. Austin, P.J.; Kim, C.F.; Perera, C.J.; Moalem-Taylor, G. Regulatory T cells attenuate neuropathic pain following peripheral nerve injury and experimental autoimmune neuritis. Pain 2012, 153, 1916–1931. [Google Scholar] [CrossRef] [PubMed]
  50. Flatters, S.J.; Bennett, G.J. Ethosuximide reverses paclitaxel- and vincristine-induced painful peripheral neuropathy. Pain 2004, 109, 150–161. [Google Scholar] [CrossRef] [PubMed]
  51. Joseph, E.K.; Levine, J.D. Comparison of oxaliplatin- and cisplatin-induced painful peripheral neuropathy in the rat. J. Pain 2009, 10, 534–541. [Google Scholar] [CrossRef] [PubMed]
  52. Shibata, K.; Sugawara, T.; Fujishita, K.; Shinozaki, Y.; Matsukawa, T.; Suzuki, T.; Koizumi, S. The astrocyte-targeted therapy by bushi for the neuropathic pain in mice. PLoS ONE 2011, 6, e23510. [Google Scholar] [CrossRef] [PubMed]
  53. Scullion, G.A.; Kendall, D.A.; Sunter, D.; Marsden, C.A.; Pardon, M.C. Central noradrenergic depletion by DSP-4 prevents stress-induced memory impairments in the object recognition task. Neuroscience 2009, 164, 415–423. [Google Scholar] [CrossRef] [PubMed]
  54. Johnson, J.D.; Campisi, J.; Sharkey, C.M.; Kennedy, S.L.; Nickerson, M.; Greenwood, B.N.; Fleshner, M. Catecholamines mediate stress-induced increases in peripheral and central inflammatory cytokines. Neuroscience 2005, 135, 1295–1307. [Google Scholar] [CrossRef] [PubMed]
  55. Nelson, L.E.; Lu, J.; Guo, T.; Saper, C.B.; Franks, N.P.; Maze, M. The alpha2-adrenoceptor agonist dexmedetomidine converges on an endogenous sleep-promoting pathway to exert its sedative effects. Anesthesiology 2003, 98, 428–436. [Google Scholar] [CrossRef] [PubMed]
  56. Zarrindast, M.R.; Homayoun, H.; Khavandgar, S.; Fayaz-Dastgerdi, M. The effects of simultaneous administration of alpha(2)-adrenergic agents with L-NAME or L-arginine on the development and expression of morphine dependence in mice. Behav. Pharmacol. 2002, 13, 117–125. [Google Scholar] [CrossRef] [PubMed]

Share and Cite

MDPI and ACS Style

Li, D.; Lee, Y.; Kim, W.; Lee, K.; Bae, H.; Kim, S.K. Analgesic Effects of Bee Venom Derived Phospholipase A2 in a Mouse Model of Oxaliplatin-Induced Neuropathic Pain. Toxins 2015, 7, 2422-2434. https://doi.org/10.3390/toxins7072422

AMA Style

Li D, Lee Y, Kim W, Lee K, Bae H, Kim SK. Analgesic Effects of Bee Venom Derived Phospholipase A2 in a Mouse Model of Oxaliplatin-Induced Neuropathic Pain. Toxins. 2015; 7(7):2422-2434. https://doi.org/10.3390/toxins7072422

Chicago/Turabian Style

Li, Dongxing, Younju Lee, Woojin Kim, Kyungjin Lee, Hyunsu Bae, and Sun Kwang Kim. 2015. "Analgesic Effects of Bee Venom Derived Phospholipase A2 in a Mouse Model of Oxaliplatin-Induced Neuropathic Pain" Toxins 7, no. 7: 2422-2434. https://doi.org/10.3390/toxins7072422

Article Metrics

Back to TopTop