1. Introduction
Chronic pain can be classified as neuropathic or inflammatory. While inflammatory pain has many therapeutic options, current available treatments for neuropathic pain (NP) are limited and usually accompanied by severe side effects, making their use controversial [
1]. The characteristics of NP are hyperalgesia and allodynia and several comorbidities such as anxiety and depression [
2,
3]. The complexity of the symptomatology makes NP the most difficult to treat chronic pain condition and novel and effective strategies to prevent this condition are an urgent medical need.
Even though the mechanisms responsible for the induction and maintenance of NP are not fully elucidated, evidence indicates excessive oxidative stress and oxidative species formation can contribute to the onset of chronic NP. In this condition there is an increase in the endogenous reactive oxygen species (ROS) production that overwhelms a cell’s antioxidant capability leading to cellular damage [
4]. An important ROS target is polyunsaturated fatty acids, which are extremely sensitive to oxidation. Thus, the high sensibility of neurons to oxidative damage originates from the high concentrations of axonal mitochondria, high cell membrane concentrations of phospholipids, and weak antioxidant defenses [
5], that makes oxidative stress a relevant target to NP management [
6].
Antioxidants obtained from natural sources include low-molecular-weight compounds, such as vitamins, plant-derived polyphenols, glutathione, and carotenoids. In recent years, the use of plant-derived preparations containing high levels of polyphenols for the management of pain has raised, thanks to their lower side effects compared to synthetic compounds [
7]. Several natural compounds have showed promising efficacy in different models of NP [
8] and numerous studies are currently focusing on the characterization and application of natural agents in various diseases for the reduction in and/or elimination of free radicals [
9]. Furthermore, natural antioxidants have contributed to developing new therapeutic interventions for the treatment of chronic NP [
10].
The bark of
Magnolia officinalis Rehder & E. Wilson, also known as "Houpo" in Chinese, has been used for thousands of years in traditional Eastern medicine, boasting more than 2,500 years of history [
11]. In Eastern culture, the uses of
Magnolia officinalis extracts are as varied as treating depression, anxiety, nervous disorders, gastrointestinal disorders, asthma and stroke, relieving migraine, muscle pain, and fever [
11]. The active component of
Magnolia officinalis extracts (MOE) are mainly polyphenolic neolignans, such as magnolol and honokiol [
12,
13]. These compounds are known for their wide range of pharmacological effects that are largely attributed to their antioxidant activity [
14]. In particular, the magnolol and honokiol inhibited oxygen consumption and malondialdehyde production by lipid peroxidation with a 1000 times higher efficacy than α-tocopherol [
15].
The imbalance between the oxidative and antioxidant mechanisms of the body, leads to oxidative stress and promotes inflammation. Evidence indicates the involvement of proinflammatory mediators and microglia-mediated neuroinflammation as processes underlying nociceptive responses [
16,
17], including conditions of NP [
2]. Thus, treatments endowed with anti-inflammatory and antioxidant activities could represent promising therapeutic option. In addition to the antioxidant activity, honokiol (HNK) displayed anti-inflammatory action in primary cultures of microglia and astrocytes stimulated by LPS, as demonstrated by the inhibition of expression of pro-inflammatory mediators, such as IL-6, IL-1β, iNOS and TNF-α [
18]. In the effort to identify new potential therapies for NP, the purpose of our research was to evaluate the analgesic activity of an HNK-rich standardized extract of
Magnolia officinalis Rehder & E. Wilson bark (MOE) in an animal model of neuropathic pain. The underlying molecular mechanisms of MOE and HNK were investigated in vitro. It has been observed that MOE and its main constituents magnolol and HNK can activate cannabinoid receptors. Specifically, magnolol behaved as a partial agonist for CB2 receptors while HNK showed full agonistic activity at CB1 receptors [
19]. Since the activation of the cannabinoid system promotes potent antioxidant and anti-inflammatory activity [
20], we also assess whether the analgesic effect might depend on modulation of this receptor class.
2. Materials and Methods
2.1. Drug administration
Magnolia officinalis Rehder & E.H. Wilson bark extract (MOE, extraction solvent: ethanol 96% v/v, standardized to contain 40% honokiol, Naturex Inc., South Hackensack, NJ, USA) was used. For behavioral tests, MOE was dissolved in saline (0.9% NaCl) and administered in a volume of 10 ml/kg by gavage (p.o.) 45 min before the experimental procedure at the dose of 30 mg/kg, except for the dose–response curve where doses of MOE ranged from 1 to 60 mg/kg. For in vitro studies MOE was dissolved in Dulbecco’s modified Eagle’s medium (DMEM) (Sigma Aldrich, Milan, Italy) to reach the final concentrations of 0.1, 1 and 10 μg/ml, according to cell viability experiments for cell culture studies. AM251 (Tocris, Bristol, UK), a known CB1 antagonist, was dissolved in dimethyl sulfoxide/Tween 80/0.9% saline (1:1:18) and administered (0.5 and 3 mg/kg i.p) 30 min before behavioral tests. HNK (0.1 -10 μg/ml; Sigma Aldrich, Milan, Italy) and DAPT (3 µM; Sigma Aldrich, Milan, Italy) was dissolved in 0.5% DMSO. Pregabalin (30 mg/kg i.p.; Sigma Aldrich, Milan, Italy), used as antiallodynic reference drug, was dissolved in saline solution, and administered 3 h before testing.
2.2. Animals
CD1 male mice (20-22 g) from the Envigo (Varese, Italy) were used. Mice were randomly assigned to standard cages, with four to five animals per cage. The cages were placed in the experimental room 24 h before behavioural testing for acclimatization. The animals were fed a standard laboratory diet and tap water ad libitum and kept at 23 °C with a 12 h light/dark cycle, light on at 07:00 h. All animal care and experimental protocols were in compliance with international laws and policies (Directive 2010/63/EU of the European parliament and of the council of 22 September 2010 on the protection of animals used for scientific purposes; Guide for the Care and Use of Laboratory Animals, US National Research Council, 2011). Animal studies are reported in compliance with the animal research: reporting of in vivo experiments (ARRIVE) guidelines [
21,
22]. All efforts were taken to minimize the number of animals used and their suffering. Mice were sacrificed by cervical dislocation for removal of spinal cord for in vitro analysis. The number of animals per experiment was based on a power analysis and calculated by G power software [
23]. To determine the anti-nociceptive effect, each tested group comprised 8 animals.
2.3. SNI model
As previously reported [
24], mice were anesthetized with a mixture of 4% isoflurane in O
2/N
2O (30:70 V/V) and placed in a prone position. The right hind limb was slightly elevated, and a skin incision was made on the lateral surface of the thigh. The sciatic nerve was exposed and both tibial and common peroneal nerves were ligated with a microsurgical forceps (5.0 silk, Ethicon; Johnson & Johnson Intl, Brussels, Belgium) and transacted together. Uninjured sural extensions are used to measure the mechanical allodynia and thermal hyperalgesia associated with the model. The sham procedure consisted of the same surgery without ligation and transection of the nerves.
2.4. Nociceptive tests
Behavioral nociceptive tests were performed before surgery, to establish a baseline for comparison with postsurgical values, and 7 days after surgery by a blinded operator.
2.4.1. Hot Plate Test
The hot plate test involves the evaluation of thermal hyperalgesia using a circular metal surface (24 cm diameter) electrically heated to a temperature of about 52.5 °C. The mice are placed on the hot plate surrounded by a transparent acrylic cage and the response time of the animals to the hyperalgesic stimulus is measured. Response latency (measured in s) consists of a leap, licking, or shaking of the paw. The mouse is immediately removed from the plate when it exhibits any of these symptoms. The animals were tested one at a time and did not undergo a period of adaptation to the experimental system prior to testing [
25].
2.4.2. Von Frey filaments
The Von Frey test was used to evaluate mechanical allodynia [
26]. The tests were carried out both before the operations, using the data obtained as a reference, and afterwards for comparison. Mechanical nociception was measured by Von Frey monofilaments. Mice were placed in single plexiglass chambers [8.5 x 3.4 x 3.4 (h) cm]. After a settling period of 1 h inside the chambers, the mechanical threshold was measured through a stimulus using Von Frey monofilaments with increasing degree of strength (0.04, 0.07, 0.16, 0.4, 0.6, 1.0, 1.4, 2.0 g) on both legs, ipsilateral and contralateral. The response was defined by the withdrawal of the paw three times out of five stimuli performed. In the event of a negative response, the next higher-grade strand was applied, and the averages of the responses were finally calculated.
2.5. Evaluation of locomotor side effects
2.5.1. Rotarod test
Rotarod was used to assess any impairment to the mouse motor coordination induced by the treatments. The test was performed before and 15, 30, 45, 60, 90, and 120 min after oral administration in naïve mice. The motor function was evaluated by counting the number of falls in 30 s, as previously described [
27].
2.5.2. Hole board test
The spontaneous mobility and the exploratory activity were recorded as counts in 5 min by means of the hole board test, as described [
27]. The test was performed 45 min after oral administration in naïve mice.
2.6. DPPH Radical Scavenging Assay
The capacity of MOE and HNK to scavenge 1,1-diphenyl-2-picryl-hydrazyl (DPPH) free radicals was measured by a spectrophotometric method. Crude extract and HNK were solved in methanol. One hundred microliters of extract, or HNK at different concentrations, were mixed with 100 μl of a DPPH methanolic solution (0.04 mg/ml). Methanol served as a blank and DPPH in methanol without the extract or HNK served as positive control. Ascorbic acid was used as reference drug. After 30 min of reaction at room temperature, the absorbance was measured at 517 nm. Controls contained all the reaction reagents except the plant extract, honokiol or reference substance. Background interferences from solvents were deducted from the activities of the corresponding extracts prior to calculating radical scavenging capacity as follows:
The antioxidant activity of honokiol was expressed as IC50, which is defined as the concentration (μg/ml) required to scavenge 50% of DPPH radicals. IC50 values were estimated by a non-linear regression (GraphPad Prism version 10.0). A lower IC50 value indicates higher antioxidant activity. The results are given as a mean ± SEM of experiments done in triplicate.
2.7. Preparation of tissue and cell lysates
Proteins extraction from tissues and cells were performed as previously reported [
26]. Briefly, mice were sacrificed 7 days after SNI, and the lumbar spinal cord tissue was removed. Samples were homogenized in a lysis buffer containing 0.1% SDS (Sigma-Aldrich). The homogenate was centrifuged at 12000 × g for 30 min at 4 °C and the pellet was discarded. Proteins from BV2 cells were extracted by radioimmunoprecipitation assay buffer (RIPA) buffer (Sigma-Aldrich) and the insoluble pellet was separated by centrifugation (12000 × g for 30 min, 4 °C). The total protein concentration in the supernatant was measured using Bradford colorimetric method (Sigma-Aldrich).
2.8. Western blotting
Protein samples (20-40 µg of protein/lane) were separated by 10% SDS PAGE [
28]. Proteins were then blotted onto nitrocellulose membranes (120 min at 100 V) using standard procedures. Membranes were blocked in PBS with 1% tween 20 (PBST) containing 5% non-fat dry milk for 120 min and incubated overnight at 4 °C with primary antibodies p-ERK1/2 (Thr202/Tyr204) (1:1000; Cell Signaling), p-p38 (Thr180/Tyr182) (1:1000; Cell Signaling), p-JNK1 (Thr183/Tyr185) (1:750; Cell Signaling), p-p65 (Ser536) (1:1000; Santa Cruz Biotechnologies), iNOS (1:500; Santa Cruz Biotechnologies), anti-IL-1β (1:1000; Bioss Antibodies, MA, USA), IL-10 (1:500; Santa Cruz Biotechnologies); Jagged1 (1:500; Santa Cruz Biotechnologies). The blots were rinsed three times with PBST and incubated for 2 h at rt with HRP-conjugated mouse anti-rabbit (1:3000; Santa Cruz Biotechnology) and goat anti-mouse (1:5000, bs-0296G,) (Bioss Antibodies) and then detected by chemiluminescence detection system (Pierce, Milan, Italy). For each sample, the signal intensity was normalized to that of total protein stained by Ponceau S and the acquired images were quantified using Image Lab software.
2.9. BV2 Cell culture
A murine microglial line BV-2 (mouse, C57BL/6, brain, microglial cells, Tema Ricerca, Genova, Italy) was used for this study. The cells were thawed and placed in a 75 cm
2 flask (Sarstedt, Milan) in a medium containing RPMI with the addition of 10% of heat-inactivated (56 °C, 30 min) fetal bovine serum (FBS, Gibco®, Milan) and 1% glutamine. Cells were grown at 37 °C and 5% CO
2 with daily medium change [
29]. Microglial cells were seeded in 6-well plates (3 × 10
5 cells/well) until 70-80% confluence was achieved. The cells were then pretreated with MOE (0.1-10 µg mL
-1), HNK (3 µg mL
-1), or DAPT (SAHA, 3 µM) for 4 h and then stimulated with LPS, (250 ng mL
-1) for 24 h.
2.10. CCK-8 test
Cell viability was performed using a Cell Counting Kit (CCK-8, Merck) according to the manufacturer’s instructions. A total of 5.0 x 10
5 cells/well were seeded into 96 multi-well plates and grown to confluence. The absorbance was measured at 450 nm using a microplate photometer HiPo MPP-96 (Biosan, Riga, Latvia). The treatments were performed in six replicates in three independent experiments, and the cell viability was calculated by normalizing the values to the control’s mean [
30].
2.11. Statistical analysis
Data and statistical analysis in this study comply with the recommendation on experimental design and analysis in pharmacology [
31]. For in vitro analysis, data are expressed as the mean ± SEM of five experiments and assessed by one-way and two-way ANOVA followed by Tukey post hoc test. The behavioral data are presented as means ± SEM. Two-way analysis of variance (ANOVA) followed by Bonferroni post hoc was used for statistical analysis. For the locomotor activity, the unpaired sample t-test was performed. The software GraphPad Prism version 9.0 (GraphPad Software, San Diego, CA) was used in all statistical analysis. For each test a value of p < 0.05 was considered significant.
4. Discussion
Nowadays, available treatments for neuropathic pain are effective in less than 50 percent of patients because they have numerous side effects that limit their prolonged use [
1,
37]. The World Health Organization (WHO) reported that more and more people support the use of medicinal plant therapy for neuropathic pain [
38]. Based on the high compliance of patients towards herbal medicines and on the promising efficacy showed by natural antioxidants in the management of neuropathic pain, in the present work we investigated the capability of an HNK-rich ethanolic extract of
Magnolia officinalis (MOE), at antioxidant doses, to modulate pain perception after oral administration in mice. The role of the main constituent HNK in the mechanism of action was also assessed.
Magnolia officinalis is largely used in the traditional Chinese and Eastern medicine and some of its pharmacological properties, such as antioxidant, anti-inflammatory, antibiotic, and antispasmodic, are also exploited in the clinical practice in the treatment of gastrointestinal disorders, anxiety, depression, and allergies [
39]. Neolignans, particularly magnolol and HNK, represent the major constituents responsible for the biological and pharmacological properties of
Magnolia officinalis extracts [
13,
39], mainly related to their antioxidant effects. In the present study we investigated the analgesic activity of MOE in conditions of acute pain as well as its antiallodynic effect in the SNI model, a condition of trauma-induced NP. Acute oral administration of MOE induced thermal antinociception in naïve mice. We also present evidence on the attenuation of mechanical allodynia on trauma-induced NP by the extract. MOE pain-relieving activity was devoid of any alteration in the animals’ gross behavior and the extract did not produce any locomotor impairment.
Increased extracellular glutamate levels following painful stimuli lead to the activation of numerous intracellular pathways, including free radicals’ formation (i.e., ROS), which increase, resulting in oxidative stress. This condition activates cyclooxygenase (COX) enzymes and enhances the production of prostaglandins through the activation of transcription factors (i.e., nuclear factor kappa (NF-kB)), and MAPK [
40,
41]. MAPKs represent one of the most important signal transduction pathways associated with the onset and development of NP [
42]. Indeed, spinal activation of MAPKs has been detected in neurons and glial cells following nerve injury [
43]. Consistently, an increased phosphorylation of ERK1/2, p38 and JNK1 was observed in the spinal cord dorsal horn of SNI mice. MOE oral administration drastically reduced p-p38 and p-JNK1 levels with no effect on pERK1/2. In NP conditions p38 is largely involved in the modulation of microglia activity [
44], while JNK is mainly expressed in astrocytes cells [
45]. Thus, a prominent modulation of glia activation by the extract can be hypothesized.
To further elucidate the mechanism of MOE, the effects on the main proinflammatory mediators was investigated. Increased iNOS expression has been observed in mice with neuropathy [
46] and elevated levels of microglial iNOS have been reported in proinflammatory conditions [
47,
48]. Another important mediator is NF-kB, commonly considered the driver of microglia activation. Inhibitors of this pathway represent promising candidate for neuropathic pain attenuation [
49]. A reversal of NF-kB, iNOS and IL-1ß overexpression in spinal cord samples from SNI following MOE administration was observed, along with an overexpression of the anti-inflammatory cytokine IL-10 [
50], further confirming the capability of MOE to attenuate spinal neuroinflammation. To confirm this hypothesis and to assess the role of the main constituent HNK in the MOE activity, the effect of the neolignan on LPS-stimulated microglial BV2 cells was evaluated. LPS-stimulated BV2 cells were used as a validated in vitro model for reproducing the effect of microglia activation in animals and they have been used to investigate the role of neuroinflammation in models of neuropathic pain [
51]. HNK restored the cell viability and homeostatic morphology in LPS-stimulated cells, in addition of reducing the number of cells in the proinflammatory state. These results are supported by previous studies on primary cultures of microglia and astrocytes stimulated by LPS in which HNK displayed anti-inflammatory activity by reducing pro-inflammatory mediators (iNOS, IL-6, IL-1β and TNF-α) [
18]. In another studies, HNK abrogated production of LPS-stimulated TNF-α, inhibited activity of NF-κB, COX-2, and peroxidation of lipid in mouse monocytes [
52].
Several studies have shown that neolignans present in the bark of
Magnolia officinalis interact with cannabinoid (CB) receptors and HNK has shown full agonistic activity at CB1 receptors [
19]. The presence of CB1 in the central nervous system (CNS) plays a crucial role in controlling normal synaptic activity, which, if altered, can underlie the onset of numerous pathologies, including neurodegenerative diseases and behavioral disorders [
53] An important role in neuropathic pain perception is attributed to CB1, as in vivo studies showed that reducing the activity of these receptors results in increased pain sensation [
54], and today, the endocannabinoid system represents one of the main targets for the modulation of pathways involved in neuropathic pain [
55].
CB1 involvement in the pain-relieving effect of MOE was evaluated by means of treatment with AM251, a known antagonist of this receptor class. A possible modulation of the endocannabinoid system in NP conditions by the extract was indicated by the prevention of the anti-allodynic effect of MOE in the presence of the CB1 antagonist. Concerning the molecular mechanisms involved, AM251 treatment selectively prevented p-p38, iNOS, NF-kB and IL-1ß increase, indicating a prevalent involvement of the CB1-mediated endocannabinoid system in the MOE-induced attenuation of pro-inflammatory events.
HNK was found to produce antitumor activity through the modulation of the Notch signaling pathway. An inhibition of cell proliferation, colony formation, and colon cancer markers by inhibiting the γ-secretase complex and the Notch signaling pathway was observed [
56]. Furthermore, treatment with HNK potently inhibited melanoma cells by reduction the expression of cleaved Notch and downstream proteins [
57]. Several studies showed that the Notch signaling pathway is involved in the promotion of NP in the SNI model [
58,
59]. Treatment with Notch inhibitors attenuates pain hypersensitivity in trauma-induced [
60] and diabetic [
61] neuropathies. Notch signaling occurs between two adjacent cells, a signal-sending cell that present the ligand (Delta-like 1, 3, and 4 and Jagged 1 and 2) and a signal-receiving cell that expresses the Notch receptor [
62]. By investigating the role of Notch signaling pathway in the mechanism of MOE, in spinal cord samples from SNI mice we observed and increased expression of Jagged1, the Notch ligand, and of the membrane-anchored Notch extracellular truncation (NEXT) fragment which is generated by Notch cleavage following Notch receptor activation. In MOE-treated mice we observed a reduction of the overexpression of the above-mentioned proteins. The role of HNK in this signaling cascade was investigated in LPS-stimulated BV2 cells that have been reported to express Jagged1 [
63,
64]. Both MOE and HNK treatments drastically reduced the Jagged1 overexpression, further confirming the involvement of the Notch signaling pathway in the MOE effect. Furthermore, present findings let hypothesize that the modulation of the Notch pathway is related to the content in HNK.
Figure 1.
Analgesic activity of MOE in condition of acute thermal pain. (A) Dose-response curve for the amalgesic effect of MOE (1-60 mg/kg p.o.) in the hot plate test. (B) Time-course curve for the analgesic effect of MOE (30 mg/kg p.o.). (C) Comparison of MOE activity vs morphine (MORPH; 7 mg/kg s.c.). CTRL: control untreated group. *P<0.05, **P<0.01 vs CTRL.
Figure 1.
Analgesic activity of MOE in condition of acute thermal pain. (A) Dose-response curve for the amalgesic effect of MOE (1-60 mg/kg p.o.) in the hot plate test. (B) Time-course curve for the analgesic effect of MOE (30 mg/kg p.o.). (C) Comparison of MOE activity vs morphine (MORPH; 7 mg/kg s.c.). CTRL: control untreated group. *P<0.05, **P<0.01 vs CTRL.
Figure 2.
Antiallodynic activity of MOE in SNI mice. (A)Dose-response curve for the attenuation of mechanical hypersensitivity in SNI mice by MOE (1-30 mg/kg p.o.). (B) Time-course evaluation of the increase in the mechanical threshold by MOE 30 mg/kg. (C) Comparison of the MOE activity with pregabalin (PREG) used as reference drug. (D) Determination of the non-hyperalgesic dose of AM251. (E) Prevention of MOE antiallodynic effect by AM251 (0.5 mg/kg). CONTRA: contralateral side; IPSI: ipsilateral side; CTR: control group. *P0.05, ***P<0.001 vs CTRL; °P<0.05, °°°P<0.001 vs IPSI.
Figure 2.
Antiallodynic activity of MOE in SNI mice. (A)Dose-response curve for the attenuation of mechanical hypersensitivity in SNI mice by MOE (1-30 mg/kg p.o.). (B) Time-course evaluation of the increase in the mechanical threshold by MOE 30 mg/kg. (C) Comparison of the MOE activity with pregabalin (PREG) used as reference drug. (D) Determination of the non-hyperalgesic dose of AM251. (E) Prevention of MOE antiallodynic effect by AM251 (0.5 mg/kg). CONTRA: contralateral side; IPSI: ipsilateral side; CTR: control group. *P0.05, ***P<0.001 vs CTRL; °P<0.05, °°°P<0.001 vs IPSI.
Figure 3.
Effect of MOE on locomotor behavior. (A) Lack of effect of MOE (30 mg/kg) on motor coordination evaluated by the rotarod test. The test was performed before and 15, 30, 45, 60, 90, and 120 min after oral administration in naïve mice. (B) Lack of impairment of spontaneous mobility and exploratory activity by MOE (30 mg/kg) in the hole board test. The test was performed 45 min after administration. ***P0.001 vs CTRL.
Figure 3.
Effect of MOE on locomotor behavior. (A) Lack of effect of MOE (30 mg/kg) on motor coordination evaluated by the rotarod test. The test was performed before and 15, 30, 45, 60, 90, and 120 min after oral administration in naïve mice. (B) Lack of impairment of spontaneous mobility and exploratory activity by MOE (30 mg/kg) in the hole board test. The test was performed 45 min after administration. ***P0.001 vs CTRL.
Figure 4.
Effect of MOE on MAPK phosphorylation in spinal cord tissue from SNI mice. Protein levels were evaluated by western blot experiments. Representative blots are reported. CONTRA: contralateral side; IPSI: ipsilateral side. *P0.05, **P<0.01, ***P<0.001 vs CONTRA; °P<0.05, °°P<0.01 vs IPSI; §P<0.05 vs IPSI+MOE.
Figure 4.
Effect of MOE on MAPK phosphorylation in spinal cord tissue from SNI mice. Protein levels were evaluated by western blot experiments. Representative blots are reported. CONTRA: contralateral side; IPSI: ipsilateral side. *P0.05, **P<0.01, ***P<0.001 vs CONTRA; °P<0.05, °°P<0.01 vs IPSI; §P<0.05 vs IPSI+MOE.
Figure 5.
Effect of MOE on the expression of proinflammatory and anti-inflammatory mediators in the spinal cord of SNI mice. Protein levels were evaluated by western blot experiments. Representative blots are reported. CONTRA: contralateral side; IPSI: ipsilateral side. *P0.05, **P<0.01 vs CONTRA; °P<0.05, °°P<0.01 vs IPSI; §P<0.05 vs IPSI+MOE.
Figure 5.
Effect of MOE on the expression of proinflammatory and anti-inflammatory mediators in the spinal cord of SNI mice. Protein levels were evaluated by western blot experiments. Representative blots are reported. CONTRA: contralateral side; IPSI: ipsilateral side. *P0.05, **P<0.01 vs CONTRA; °P<0.05, °°P<0.01 vs IPSI; §P<0.05 vs IPSI+MOE.
Figure 6.
Anti-inflammatory effect of HNK on LPS-stimulated BV2 cells. (A) Dose-response curve for the prevention by HNK (0.1-10 µg/ml) of cell viability reduction induced by LPS stimulation (250 ng for 24 h). (B) HNK (3 µg/ml) reduced the increase in the cell surface induced by LPS stimulation. (C) HNK (3 µg/ml) reduced the number of BV2 cells in the proinflammatory state after LPS stimulation. (D) Representative images of LPS-stimulated BV2 cells. *P<0.05, **P<0.01, ***P<0.001, ****P<0.0001.
Figure 6.
Anti-inflammatory effect of HNK on LPS-stimulated BV2 cells. (A) Dose-response curve for the prevention by HNK (0.1-10 µg/ml) of cell viability reduction induced by LPS stimulation (250 ng for 24 h). (B) HNK (3 µg/ml) reduced the increase in the cell surface induced by LPS stimulation. (C) HNK (3 µg/ml) reduced the number of BV2 cells in the proinflammatory state after LPS stimulation. (D) Representative images of LPS-stimulated BV2 cells. *P<0.05, **P<0.01, ***P<0.001, ****P<0.0001.
Figure 7.
Attenuation of MOE and HNK of the Notch signaling pathway. (A) Lack of impairment of cell viability by MOE (0.1-10 µg/ml) evaluated by the CCK-8 test in BV2 cells. (B) Reduction by MOE (10 µg/ml), HNK (3 µg/ml) and DAPT (3 µM) of the overexpression of Jagged1 induced by LPS stimulation in BV2 cells. Prevention of Jagged1 (C) and NEXT (D)overexpression by MOE treatment in spinal cord samples of SNI mice. Lack of effect by AM251 administration. Representative blots are reported. **P<0.01, ***P<0.001, ****P<0.0001.
Figure 7.
Attenuation of MOE and HNK of the Notch signaling pathway. (A) Lack of impairment of cell viability by MOE (0.1-10 µg/ml) evaluated by the CCK-8 test in BV2 cells. (B) Reduction by MOE (10 µg/ml), HNK (3 µg/ml) and DAPT (3 µM) of the overexpression of Jagged1 induced by LPS stimulation in BV2 cells. Prevention of Jagged1 (C) and NEXT (D)overexpression by MOE treatment in spinal cord samples of SNI mice. Lack of effect by AM251 administration. Representative blots are reported. **P<0.01, ***P<0.001, ****P<0.0001.
Figure 8.
Antioxidant activity of HNK and MOE. (A) Anti-radical scavenger activity of MOE and HNK (1-10 µg/µl) in the DPPH test. Ascorbic acid was used as refence drug. (B) Reduction of the overexpression of Nfr2 by MOE in spinal cord samples from SNI mice. *P<0.05.
Figure 8.
Antioxidant activity of HNK and MOE. (A) Anti-radical scavenger activity of MOE and HNK (1-10 µg/µl) in the DPPH test. Ascorbic acid was used as refence drug. (B) Reduction of the overexpression of Nfr2 by MOE in spinal cord samples from SNI mice. *P<0.05.
Figure 9.
MOE-induced increase of MBP content in the spinal cord of SNI mice. Lack of effect by AM251 treatment. MBP values were obtained by western blotting analysis. Representative blots are reported. *P0.05, **P<0.01.
Figure 9.
MOE-induced increase of MBP content in the spinal cord of SNI mice. Lack of effect by AM251 treatment. MBP values were obtained by western blotting analysis. Representative blots are reported. *P0.05, **P<0.01.