Anti‑inflammatory actions of gabapentin and pregabalin on the substance P‑induced mitogen‑activated protein kinase activation in U373 MG human glioblastoma astrocytoma cells

  • Authors:
    • Keisuke Yamaguchi
    • Seiichiro Kumakura
    • Akimasa Someya
    • Masako Iseki
    • Eiichi Inada
    • Isao Nagaoka
  • View Affiliations

  • Published online on: August 28, 2017     https://doi.org/10.3892/mmr.2017.7368
  • Pages: 6109-6115
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Gabapentin (GBP) and pregabalin (PGB) exert antinociceptive effects on chronic nociceptive responses with neuropathic or inflammatory conditions. Furthermore, it is considered that GBP and PGB exhibit anti‑inflammatory effects by modulating the substance P (SP)‑mediated neurokinin‑1 receptor (NK1R; a SP receptor) response. Thus, in the present study, the effects of GBP and PGB on SP‑induced activation were investigated in the human glioblastoma astrocytoma U373 MG cell line, which expresses high levels of functional high‑affinity NK1R, and produces interleukin (IL)‑6 and IL‑8 in response to SP. The results indicated that GBP and PGB suppressed the SP‑induced production of IL‑6, and IL‑8 in U373 MG cells. Furthermore, GBP and PGB inhibited the SP‑induced phosphorylation of p38 mitogen‑activated protein kinase (MAPK) and nuclear factor (NF)‑κB, and the nuclear translocation of NF‑κB in U373 MG cells. Together, these observations suggest that GBP and PGB likely prevent SP‑induced IL‑6 and IL‑8 production in U373 MG cells via the inhibition of signaling molecules, including p38 MAPK and NF‑κB, thereby exhibiting antineuroinflammatory effects.

Introduction

A neuropeptide substance P (SP) is an important mediator of neurogenic inflammation in the central and peripheral nervous systems. SP is implicated in pain, and also plays an important role in tumor cell proliferation, anti-apoptotic effects on tumor cells, angiogenesis, tumor cell invasion and metastasis (13). SP is released from primary afferent nociceptors and sympathetic postganglionic neurons, and activates neighboring receptors, thereby triggering spreading activation (4,5). Moreover, SP has been shown to induce the expression of proinflammatory cytokines and chemokines, such as interleukin (IL)-6 and IL-8 (6,7), which are involved in the pathogenesis of several human brain disorders (such as multiple sclerosis, dementia complex, and Alzheimer's disease) (8), although it is currently a matter of debate whether SP really plays a pathogenic role in these disorder. Previous report has shown that the activation of a SP receptor [neurokinin-1 receptor (NK1R)] elicits signals affecting inflammatory cytokine gene expression (7). In addition, it has been reported that SP potently triggers the activation of nuclear factor (NF)-κB, an important transcriptional activator, which regulates the production of various cytokines and other proinflammatory mediators (7).

Mitogen-activated protein kinases (MAPK), a family of protein Ser/Thr kinases, consist of at least three major subfamilies: i) the p42/44 MAPKs, which are also called extracellular signal regulated kinases (ERK-1 and ERK-2); ii) the c-Jun NH2-terminal kinase/stress-activated protein kinases (JNK/SAPK) including p46 JNK1 and p54 JNK2; and iii) the p38 MAPK subfamily. MAPKs are activated under stress conditions in response to a variety of extracellular stimuli, including oxidative stress. Among these MAPKs, phosphorylation of p38 MAPK is induced in dorsal horn of the spinal cord and dorsal root ganglia following peripheral nerve injury or inflammation (911).

Gabapentin (GBP) and pregabalin (PGB) are structural analogues of γ-amino butyric acid (GABA) with lipophilic characteristics, and were developed as potential anticonvulsants (12). However, many studies have shown that these substances do not act as GABA agonists and indeed have little demonstrable effect on any aspect of GABA transmission (13,14). Furthermore, a recent study has clarified the current understanding of gabapentinoid pharmacology that GBP and PGB do not inhibit any conventional subtype of voltage-gated calcium channel (VGCC), but rather selectively block calcium channels that contain the α2δ-1 subunit, with pharmacodynamics and cellular-specificities, depending on the structural and biochemical states of the α2δ-1 protein (15). It is believed that blockade of VGCC containing the α2δ-1 subunit is the predominant pharmacological mechanism of both GBP and PGB (16). PGB binds potently to the α2δ-1 subunit and modulates calcium influx at nerve terminals, thereby, reducing the release of several neurotransmitters, including glutamate, noradrenaline, serotonin and SP (1719). Interestingly, GBP could be a therapeutic agent, when given systemically, for treatment of neuropathic or postsurgical pain (2024), and also reduces the experimental pain in humans after sensitization of the skin with capsaicin and heat (25). Furthermore, GBP and its derivative PGB reduce nociceptive behaviors of animal models with neuropathic pain or inflammation such as nerve ligation, injection of immune antigens, herpes infection, arthritis, diabetes, postoperative pain and thermal injury (2630). In contrast, neither GBP nor PGB alters acute nociceptive responses (31,32). Thus, it is believed that the antinociceptive action of these substances depends on the chronic nociceptive responses with neuropathic or inflammatory conditions.

It has been demonstrated that the release of peptidergic neurotransmitters (including SP) from sensory neurons is increased during inflammation or in neuropathic pain models (33). In addition, SP is axonally transported to peripheral nerve endings, where it is released in response to traumatic stimuli and induces various biological effects (33), Importantly, GBP and PGB modulate the release of SP under the conditions corresponding to significant inflammation-induced sensitization of the spinal cord, which involves the action of SP on NK1 (SP) receptor (NK1R) (34). Based on the findings, we hypothesized that GBP and PGB may exert antiinflammatory action by modulating the SP-mediated NK1R response. In this study, therefore, we investigated the antiinflammatory effects of GBP and PGB on SP-induced activation of a human glioblastoma astrocytoma cell line U373 MG cell, which expresses the high levels of NK1R.

Materials and methods

Materials

U373 MG cell line (Uppsala; ECACC08061901) was purchased from European Collection of Cell Cultures (ECACC; Salisbury, UK). SP was obtained from Sigma-Aldrich Co., LLC., (St. Louis, MO, USA); GBP from Tokyo Chemical Industry Co., Ltd. (Tokyo, Japan); PGB from Toronto Research Chemicals (Toronto, ON, Canada); Minimum essential medium (MEM), non-essential amino acids (NEAA), sodium pyruvate and fetal bovine serum (FBS) from Gibco BRL Life Technologies (Grand Island, NY, USA). Phosphate-buffered saline (PBS), RIPA buffer containing protease inhibitor cocktail, sample buffer solution containing reducing reagent (6X) for SDS-PAGE, running buffer solution (10X) for SDS-PAGE, Blocking One, WB Stripping Solution Strong, and Protein Ladder One Multi-color (Broad Range) for SDS-PAGE from Nacalai Tesque, Inc., (Kyoto, Japan). BCA protein assay reagent kit and enhanced chemiluminescence reagent, SuperSignal West Dura and NE-PER nuclear and cytoplasmic extraction reagents from Thermo Fisher Scientific, Inc. (Waltham, MA, USA). Mini-PROTEAN® TGX™ Precast Gel and Trans-Blot® Turbo™ Mini PVDF Transfer Packs from Bio-Rad Laboratories, Inc., (Hercules, CA, USA).

Antibodies

Anti-phospho-ERK1/2 MAPK (Thr202/Tyr204) rabbit antibody (no. 9101), anti-ERK1/2 MAPK rabbit antibody (no. 9102), anti-phospho-NF-κB p65 (Ser536) rabbit monoclonal antibody (mAb; no. 3033), anti-NF-κB p65 rabbit mAb (no. 8242), and histone H3 (D1H2) XP rabbit mAb (no. 4499) from Cell Signaling Technology, Inc. (Danvers, MA, USA), anti-phospho p38 MAPK rabbit antibody (Thr180/Tyr182; V121A) from Promega Corporation (Madison, WI, USA), anti-p38 MAPK (p38/SAPK2α) mouse mAb (no. 612168) from BD Biosciences (San Jose, CA, USA). Horseradish peroxidase (HRP)-conjugated goat anti-rabbit IgG (AP132P), HRP-conjugated goat anti-mouse IgG/IgM (AP308P) from Chemicon International (Temecula, CA, USA).

Cell culture

U373 MG cells were cultured in MEM supplemented with 1% (v/v) penicillin/streptomycin, and NEAA, 1 mM sodium pyruvate and 10% heat-inactivated FBS. The cells were maintained at 37°C in a 5% CO2 humidified atmosphere.

Preparation of whole cell lysate and western blot analysis

U373 MG cells were plated into 12-well tissue culture plates at a density of 1×105 cells/well and incubated in MEM with 10% FBS for 12 h, followed by incubation in MEM with 0.5% FBS for 12 h at 37°C. Subsequently, the cells were incubated with GBP or PGB (1 mM) for 60 min, and then stimulated with SP (100 nM) for 10 or 15 min. Thereafter, the cells were washed three times with ice-cold PBS and lysed in 0.1 ml of RIPA buffer (50 mmol/l Tris-HCl pH 7.6, 150 mmol/l NaCl, 1% Nonidet P40, 0.5% sodium deoxycholate, 0.1% SDS and Protease Inhibitor Cocktail). Protein concentrations of cell lysates were measured with BCA protein assay reagent (Thermo Scientific). The lysates were mixed with SDS-polyacrylamide gel electrophoresis (PAGE) sample buffer (62.5 mM Tris-HCl, pH 6.8, 2% SDS, 10% glycerol, 0.05% bromphenol blue, and 5% 2-mercaptoethanol), and applied to SDS-PAGE in 10% gels (Mini-PROTEAN® TGX™ Precast Gel; 10–20 µg protein/lane). Thereafter, separated proteins were electroblotted onto polyvinylidine fluoride membranes (Trans-Blot® Turbo™ Mini PVDF Transfer Packs). After incubation with Blocking One (Nacalai Tesque, Inc.), blots were proved with a 1,000-fold dilution of rabbit anti-phospho p38 MAPK antibody, anti-phospho ERK1/2 antibody or anti-phospho NF-κB antibody, and further proved with a 10,000-fold dilution of HRP-conjugated goat anti-rabbit IgG. Signals were detected with SuperSignal West Dura Chemiluminescent Substrate (Thermo Fisher Scientific, Inc.), and quantified using LAS-3000 luminescent image analyzer (Fujifilm, Tokyo, Japan) and MultiGauge software (Fujifilm). Thereafter, the antibody was stripped using WB Stripping Solution Strong (Nacalai Tesque, Inc.) at room temperature for 15 min. Blots were proved with a 1,000-fold dilution of mouse anti-p38 MAPK antibody, rabbit anti-ERK1/2 antibody or anti-NF-κB antibody, and further proved with a 10,000-fold dilution of HRP-conjugated goat anti-rabbit IgG or HRP-conjugated goat anti-mouse IgG/IgM. Signals were detected and analyzed, as described above.

Preparation of nuclear extract and western blot analysis

U373 MG cells were plated into 12-well tissue culture plates at a density of 2×105 cells/well and incubated in MEM with 10% FBS for 12 h, followed by incubation in MEM with 0.5% FBS for 12 h at 37°C. Subsequently, the cells were incubated with GBP or PGB (1.0 mM) for 60 min, and then stimulated with SP (100 nM) for 15 min. Thereafter, the cells were washed three times with ice-cold PBS. Cells were detached by trypsin treatment, and washed in ice-cold PBS containing phosphatase inhibitors, and then centrifuged at 300 g for 5 min. Nuclear fractions were prepared using NE-PER nuclear and cytoplasmic extraction reagents by suspending the cell pellet in a hypotonic buffer, and centrifugation at 14,000 × g for 30 min. After collection of the supernatant (cytoplasmic fraction), the pellet (nuclear fraction) was lysed and solubilized in lysis buffer containing proteasome inhibitors. Protein concentrations were determined using with BCA protein assay reagent. The nuclear extract was mixed with SDS-PAGE sample buffer), and applied to SDS-PAGE in 10% gels, followed by western blot analysis using a 1,000-fold dilution of rabbit anti-NF-κB antibody and a 10,000-fold dilution of HRP-conjugated goat anti-rabbit IgG, and a 1,000-fold dilution of rabbit histone H3 and a 10,000-fold dilution of HRP-conjugated goat anti-rabbit IgG, as described in the above section.

Quantification of IL-6 and IL-8

U373 MG cells were plated into 12-well tissue culture plates at a density of 1×105 cells/well and incubated in MEM with 10% FBS for 12 h, followed by incubation in MEM with 0.5% FBS for 12 h at 37°C. Then, the cells were incubated with GBP or PGB (1 mM) for 60 min, and stimulated with SP (100 nM) for 24 h. Culture media were recovered, centrifuged for 10 min at 12,000 × g, and the levels of IL-6 in the supernatants were measured by a sandwich enzyme-linked immunosorbent assay (ELISA) kit, according to the manufacturer's instructions (eBioscience, San Diego, CA, USA). Moreover, the levels of IL-8 in the media were measured by ELISA kit, according to the manufacturer's instructions (R&D systems, Inc., Minneapolis, MN, USA).

Statistical analysis

Data are expressed as mean ± standard deviation, and analyzed for significant difference by a one-way ANOVA with multiple comparison test using GraphPad Prism version 6.0 for Windows (GraphPad Software, San Diego, CA, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

Suppression of SP-induced activation of p38 MAPK and ERK1/2 by GBP and PGB

First, we evaluated the effect of GBP and PGB on the phosphorylation of p38 MAPK. As shown in Fig. 1A, SP stimulation (100 nM) clearly induced the phosphorylation of p38 MAPK in U373 MG cells. Interestingly, GBP (1 mM) and PGB (1 mM) substantially suppressed the SP-induced phosphorylation of p38 MAPK, although only the suppression by PGB was statistically significant (P<0.05).

Next, we evaluated the effect of GBP and PGB on the phosphorylation of ERK1/2. As shown in Fig. 1B, SP stimulation (100 nM) markedly induced the phosphorylation of ERK1/2 in U373 MG cells; however, GBP (1 mM) and PGB (1 mM) did not essentially suppress the SP-induced phosphorylation of ERK1/2.

Suppression of SP-induced activation of NF-κB and nuclear translocation of p65 by GBP and PGB

Furthermore, we evaluated the effect of GBP and PGB on the phosphorylation of NF-κB. SP stimulation (100 nM) substantially induced the phosphorylation of NF-κB in U373 MG cells. Importantly, GBP and PGB significantly abolished the SP-induced phosphorylation of NF-κB (Fig. 2A; P<0.05).

Figure 2.

Evaluation of the effects of gabapentin (GBP) and pregabalin (PGB) on substance P (SP)-induced phosporylation of nuclear factor (NF)-κB and SP-induced nuclear translocation of NF-κB. U373 MG cells were plated into 12-well tissue culture plates at a density of 2×105 cells/well and incubated in minimum essential medium (MEM) with 10% FBS for 12 h, followed by incubation in MEM with 0.5% fetal bovine serum (FBS) for 12 h at 37°C. Subsequently, the cells were incubated with or without GBP (1 mM) or PGB (1 mM) for 60 min, and then incubated with or without SP (100 nM) for 15 min. (A) The expression of phosphorylation of NF-κB were detected by probing with anti-phospho NF-κB p65 (Ser536) rabbit mAb and HRP-conjugated goat anti-rabbit immunoglobulin G (IgG). In order to confirm that equal amount of proteins were analyzed in each samples, the blots were stripped, and total NF-κB were detected by reprobing with anti-NF-κB p65 rabbit mAb and horseradish peroxidase (HRP)-conjugated goat anti-rabbit IgG. (B) After incubation of U373 MG cells with SP in the absence or presence of GBP or PGB, nuclear fractions were prepared, and the nuclear levels of NF-κB p65 were analyzed by probing with anti-NF-κB p65 rabbit mAb and HRP-conjugated goat anti-rabbit IgG. In order to confirm that equal amount of proteins were analyzed in each samples, the blots were stripped, and histone H3 were detected by reprobing with anti-histone H3 and HRP-conjugated goat anti-rabbit IgG. A representative image is shown. Data are the means ± standard deviation of 3 separate experiments, and expressed as relative to the cells incubated without SP, GBP, and PGB. Data are compared between the SP-stimulated cells incubated without and with GBP or PGB. *P<0.05.

To further characterize the effect of GBP and PGB on the SP-induced activation of NF-κB, we evaluated the nuclear translocation of NF-κB. Thus, U373 cells were pretreated with GBP (1 mM) and PGB (1 mM) followed by stimulation with SP (100 nM), and the nuclear levels of p65 were analyzed. Nuclear p65 level was increased after SP stimulation compared with unstimulated cells; however, SP-induced p65 level was significantly reduced by GBP and PGB (Fig. 2B; P<0.05).

Suppression of SP-induced IL-6 and IL-8 production by GBP and PGB

Finally, we evaluated the effect of GBP and PGB on the IL-6 and IL-8 production by U373 MG cells. As shown in Fig. 3, both IL-6 and IL-8 were produced by resting U373 MG cells without SP. Importantly, the production of IL-6 and IL-8 was substantially increased by SP stimulation (100 nM). Noticeably, GBP and PGB (1 mM) significantly suppressed both the IL-6 and IL-8 production (P<0.05).

Discussion

To our knowledge, this is the first study to demonstrate the effects of GBP and PGB on the SP-induced inflammatory responses in glioblastoma astrocytoma U373 MG cells. U373 MG glioblastoma astrocytoma cells express a functional high-affinity NK1R (a SP receptor) (35) and are able to produce IL-6 and IL-8 in response to SP (6). In this study, we revealed that GBP and PGB suppressed the SP-induced production of IL-6 and IL-8 in U373 MG cells. Furthermore, GBP and PGB inhibited the SP-induced phosphorylation of p38 MAPK and NF-κB, and nuclear translocation of NF-κB in U373 MG cells. Thus, GBP and PGB likely prevent the SP-induced IL-6 and IL-8 production in U373 MG cells by inhibiting signaling molecules including p38 MAPK and NF-κB, thereby exhibiting anti-inflammatory action.

According to pharmacokinetic study (36,37), the maximum plasma concentration of GBP after administration of a single 400 mg GBP tablet to 12 volunteers was 3.33±1.19 µg/ml (approximately 20 µM). Moreover, it has been shown that 25 µM GBP and PGB reduced the SP-induced NF-κB activation in glioblastoma cell line in vitro (38). In our preliminary experiments, we observed that 100 µM GBP weakly but 1 mM GBP clearly suppressed the SP-induced phosphorylation of p38 MAPK and NF-κB, and production of IL-6 and IL-8. Thus, we examined the effect of GBP and PGB at a concentration of 1 mM on the SP-induced phosphorylation of signaling molecules, and production of cytokines.

The concentration of GBP used in this study (1 mM) was 50-fold higher than that of the maximum plasma concentration; however, we observed that GBP at 100 µM weakly suppressed the SP-induced phosphorylation of p38 MAPK and NF-κB, and production of IL-6 and IL-8 (data not shown). Thus, we speculate that GBP could exert the anti-inflammatory action in vivo, based on our findings.

GBP and PGB are anticonvulsants originally developed as spasmolytic agents for the management of generalized or partial epileptic seizures resistant to conventional therapies (12). However, subsequent single center and multicenter, randomized double-blind trials demonstrated that GBP is also effective for the management of pain of inflammatory and neuropathic origin, such as post herpetic neuralgia and painful diabetic neuropathy (39,40). Although PGB has not been extensively investigated as GBP, recent double-blind trials showed that PGB is also effective in the management of postoperative pain and diabetic neuropathy (39). In animal models of nociception, GBP reduces the mechanical or thermal hypersensitivity associated with nerve injury (31,41), incisional injury (26), inflammatory injury (27,41,42), and formalin-induced injury (4244). PGB similarly reduces the mechanical or thermal hypersensitivity associated with injuries, described above (27,42,45). In contrast, both compounds do not exhibit substantive effect on acute pain in uninjured animal models.

An interesting action of SP is the induction and modulation of proinflammatory cytokine secretion by glial cells (7). The mechanism of SP-induced signal transduction is incompletely understood. However, it has been suggested that the activation of NF-κB is involved in SP-induced cytokine expression (7). Importantly, it is demonstrated that GBP and PGB decrease SP-induced NF-κB activation in human neuroblastoma and rat glioma cells, and that these drugs also inhibit NF-κB activation in rat spinal dorsal root ganglia cells pre-treated with SP (38). Interestingly, it has been reported that GBP and PGB attenuate the release of neuropeptides (such as SP) from inflamed spinal cord (34). Importantly, the present study demonstrated that GBP and PGB prevent the SP-induced IL-6 and IL-8 production in U373 MG cells by inhibiting p38 MAPK and NF-κB activation. Based on these findings, it could be speculated that gabapentinoids may exhibit anti-inflammatory action by suppressing not only the SP-induced cytokine production via the inhibition of p38 MAPK and NF-κB activation, but also the secretion of inflammatory neuropeptides (such as SP) at the inflamed neural tissue. In fact, it has been demonstrated that PGB binds potently to the α2δ-1 subunit of calcium channels and modulates calcium influx at nerve terminals, thereby reducing the release of several neurotransmitters, including SP (46). However, it has been also reported that PGB and GBP suppress the degradation of IκB, thereby inhibiting nuclear localization of NF-κB (p65) in SH-SY5Y glioblastoma cells (45). Thus, gabapentinoid-mediated modulation of nuclear localization of NF-κB is also considered as a molecular mechanism for the actions of GBP and PGB.

The α2δ-1 subunit of VGCC is involved in propagation of excitatory signals mediated by glutamate, calcitonin gene-related protein (CGRP), and SP (34). Its upregulation under pathological conditions is associated with hyperexcitatory states such as seizures and neuropathic pain. The causal link between α2δ-1-subunit expression and hyperalgesia has been demonstrated in several experimental animal models of spinal nerve injury (47,48). Notably, PGB binds potently to the α2δ-1 subunit and suppresses calcium influx at nerve terminals, thereby modulating hyperexcitatory states of neuronal synapse.

SP stimulates a number of intracellular signaling molecules including the members of MAPK family (ERK1/2 and p38 MAPK) via the action of NK1R. Moreover, SP induce the IL-6 and IL-8 production via the activation of p38 MAPK, ERK1/2 and NF-κB by U373 MG (49). In this study, GBP and PGB did not reduce the SP-induced ERK1/2 phosphorylation (Fig. 1B) but reduced p38 MAPK phosphorylation in U373 MG cells (Fig. 1A). Thus, our results suggest that GBP and PGB reduce the SP-induced production of IL-6 and IL-8 by the NK1R (a principal receptor for SP)-expressing U373 MG cells via the inhibition of signaling molecules p38 MAPK but not ERK1/2. It is possible that GBP and PGB may reduce the expression of NK1R, thereby suppressing the SP-induced production of IL-6 and IL-8. However, GBP and PGB did not reduce the SP-induced ERK1/2 phosphorylation, as mentioned above. Thus, GBP and PGB unlikely modulate the expression of NK1R.

In a neuropathic pain model, it is well documented that microglial activation, accompanying with p38 MAPK phosphorylation in the spinal cord, plays an important role in the development of neuropathic pain (5052). Both peripheral inflammation and nerve injury induce p38 MAPK activation in spinal microglia, and p38 MAPK inhibitor SB203580 suppresses the inflammation-induced thermal hyperalgesia and spinal nerve ligation-induced mechanical allodynia (53). Moreover, intrathecal injection of minocycline, which inhibits spinal microglia activation by blocking p38 MAPK, exerts antinociceptive effect in both inflammation and neuropathic pain models (52). These observations indicate that p38 MAPK activation plays important role in development of neuroinflammation. In addition, IL-6 is speculated to be involved in the development of neuropathic pain, because IL-6 mRNA was significantly elevated in both the dorsal and ventral horns in a neuropathic pain model of spinal nerve cryoneurolysis and spinal nerve tight ligation (54,55). IL-8 also contributes to the pathophysiology of inflammatory pain, because the expression of IL-8 was critically upregulated in the ipsilateral spinal cord dorsal horn after chronic constriction injury (56,57). In this study, GBP and PGB reduced IL-6 and IL-8 production possibly by suppressing p38 MAPK in U373 MG cell, and the results explain the pharmacological action of GBP and PGB on neuroinflammation (neuropathic pain).

In conclusion, we demonstrated that GBP and PGB suppressed the production of IL-6 and IL-8 in U373 MG cells. Furthermore, GBP and PGB inhibited the SP-induced activation of p38 MAPK, and NF-κB in U373 MG cells. Thus, GBP and PGB likely prevent the SP-induced IL-6 and IL-8 production by U373 MG cells via the inhibition of signaling molecules including p38 MAPK and NF-κB, thereby exhibiting anti-neuroinflammatory action.

Acknowledgements

This study was supported by Grant-in-Aid for Scientific Research (C) (15K10564).

References

1 

Samsam M, Coveñas R, Ahangari R, Yajeya J, Narváez JA and Tramu G: Simultaneous depletion of neurokinin A substance P and calcitonin gene-related peptide from the caudal trigeminal nucleus of the rat during electrical stimulation of the trigeminal ganglion. Pain. 84:389–395. 2000. View Article : Google Scholar : PubMed/NCBI

2 

Johnson MB, Young AD and Marriott I: The therapeutic potential of targeting substance P/NK-1R interactions in inflammatory CNS disorders. Front Cell Neurosci. 10:2962017. View Article : Google Scholar : PubMed/NCBI

3 

Muñoz M and Coveñas R: Involvement of substance P and the NK-1 receptor in cancer progression. Peptides. 48:1–9. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Ebner K and Singewald N: The role of substance P in stress and anxiety responses. Amino Acids. 31:251–272. 2006. View Article : Google Scholar : PubMed/NCBI

5 

Garcia-Recio S and Gascón P: Biological and pharmacological aspects of the NK1-receptor. Biomed Res Int. 2015:4957042015. View Article : Google Scholar : PubMed/NCBI

6 

Lieb K, Schaller H, Bauer J, Berger M, Schulze-Osthoff K and Fiebich BL: Substance P and histamine induce interleukin-6 expression in human astrocytoma cells by a mechanism involving protein kinase C and nuclear factor-IL-6. J Neurochem. 70:1577–1583. 1998. View Article : Google Scholar : PubMed/NCBI

7 

Lieb K, Fiebich BL, Berger M, Bauer J and Schulze-Osthoff K: The neuropeptide substance P activates transcription factor NF-kappa B and kappa B-dependent gene expression in human astrocytoma cells. J Immunol. 159:4952–4958. 1997.PubMed/NCBI

8 

Horuk R, Martin AW, Wang Z, Schweitzer L, Gerassimides A, Guo H, Lu Z, Hesselgesser J, Perez HD, Kim J, et al: Expression of chemokine receptors by subsets of neurons in the central nervous system. J Immunol. 158:2882–2890. 1997.PubMed/NCBI

9 

Kim SY, Bae JC, Kim JY, Lee HL, Lee KM, Kim DS and Cho HJ: Activation of p38 MAP kinase in the rat dorsal root ganglia and spinal cord following peripheral inflammation and nerve injury. Neuroreport. 13:2483–2486. 2002. View Article : Google Scholar : PubMed/NCBI

10 

Ji RR, Befort K, Brenner GJ and Woolf CJ: ERK MAP kinase activation in superficial spinal cord neurons induces prodynorphin and NK-1 upregulation and contributes to persistent inflammatory pain hypersensitivity. J Neurosci. 22:478–585. 2002.PubMed/NCBI

11 

Jin SX, Zhuang ZY, Woolf CJ and Ji RR: p38 mitogen-activated protein kinase is activated after a spinal nerve ligation in spinal cord microglia and dorsal root ganglion neurons and contributes to the generation of neuropathic pain. J Neurosci. 23:4017–4022. 2003.PubMed/NCBI

12 

Bryans JS and Wustrow DJ: 3-substituted GABA analogs with central nervous system activity: a review. Med Res Rev. 19:149–177. 1999. View Article : Google Scholar : PubMed/NCBI

13 

Taylor CP, Gee NS, Su TZ, Kocsis JD, Welty DF, Brown JP, Dooley DJ, Boden P and Singh L: A summary of mechanistic hypotheses of gabapentin pharmacology. Epilepsy Res. 29:233–249. 1998. View Article : Google Scholar : PubMed/NCBI

14 

Maneuf YP, Gonzalez MI, Sutton KS, Chung FZ, Pinnock RD and Lee K: Cellular and molecular action of the putative GABA-mimetic, gabapentin. Cell Mol Life Sci. 60:742–750. 2003. View Article : Google Scholar : PubMed/NCBI

15 

Brown JT and Randall A: Gabapentin fails to alter P/Q-type Ca2+ channel-mediated synaptic transmission in the hippocampus in vitro. Synapse. 55:262–269. 2005. View Article : Google Scholar : PubMed/NCBI

16 

SILLS G: The mechanisms of action of gabapentin and pregabalin. Curr Opin Pharmacol. 6:108–113. 2006. View Article : Google Scholar : PubMed/NCBI

17 

Maneuf YP, Hughes J and McKnight AT: Gabapentin inhibits the substance P-facilitated K (+)-evoked release of [(3)H]glutamate from rat caudial trigeminal nucleus slices. Pain. 93:191–196. 2001. View Article : Google Scholar : PubMed/NCBI

18 

Cunningham MO, Woodhall GL, Thompson SE, Dooley DJ and Jones RSG: Dual effects of gabapentin and pregabalin on glutamate release at rat entorhinal synapses in vitro. Eur J Neurosci. 20:1566–1576. 2004. View Article : Google Scholar : PubMed/NCBI

19 

Dooley DJ, Mieske CA and Borosky SA: Inhibition of K(+)-evoked glutamate release from rat neocortical and hippocampal slices by gabapentin. Neurosci Lett. 280:107–110. 2000. View Article : Google Scholar : PubMed/NCBI

20 

Rosenberg JM, Harrell C, Ristic H, Werner RA and de Rosayro AM: The effect of gabapentin on neuropathic pain. Clin J Pain. 13:251–255. 1997. View Article : Google Scholar : PubMed/NCBI

21 

Backonja M, Beydoun A, Edwards KR, Schwartz SL, Fonseca V, Hes M, LaMoreaux L and Garofalo E: Gabapentin for the symptomatic treatment of painful neuropathy in patients with diabetes mellitus: A randomized controlled trial. JAMA. 280:1831–1836. 1998. View Article : Google Scholar : PubMed/NCBI

22 

Rowbotham M, Harden N, Stacey B, Bernstein P and Magnus-Miller L: Gabapentin for the treatment of postherpetic neuralgia: A randomized controlled trial. JAMA. 280:1837–1842. 1998. View Article : Google Scholar : PubMed/NCBI

23 

Dirks J, Fredensborg BB, Christensen D, Fomsgaard JS, Flyger H and Dahl JB: A randomized study of the effects of single-dose gabapentin versus placebo on postoperative pain and morphine consumption after mastectomy. Anesthesiology. 97:560–564. 2002. View Article : Google Scholar : PubMed/NCBI

24 

Serpell MG; Neuropathic pain study group, : Gabapentin in neuropathic pain syndromes: A randomised, double-blind, placebo-controlled trial. Pain. 99:557–566. 2002. View Article : Google Scholar : PubMed/NCBI

25 

Werner MU, Perkins FM, Holte K, Pedersen JL and Kehlet H: Effects of gabapentin in acute inflammatory pain in humans. Reg Anesth Pain Med. 26:322–328. View Article : Google Scholar : PubMed/NCBI

26 

Field MJ, Holloman EF, McCleary S, Hughes J and Singh L: Evaluation of gabapentin and S-(+)-3-isobutylgaba in a rat model of postoperative pain. J Pharmacol Exp Ther. 282:1242–1246. 1997.PubMed/NCBI

27 

Houghton AK, Lu Y and Westlund KN: S-(+)-3-isobutylgaba and its stereoisomer reduces the amount of inflammation and hyperalgesia in an acute arthritis model in the rat. J Pharmacol Exp Ther. 285:533–538. 1998.PubMed/NCBI

28 

Partridge BJ, Chaplan SR, Sakamoto E and Yaksh TL: Characterization of the effects of gabapentin and 3-isobutyl-gamma-aminobutyric acid on substance P-induced thermal hyperalgesia. Anesthesiology. 88:196–205. 1998. View Article : Google Scholar : PubMed/NCBI

29 

Chen SR, Xu Z and Pan HL: Stereospecific effect of pregabalin on ectopic afferent discharges and neuropathic pain induced by sciatic nerve ligation in rats. Anesthesiology. 95:1473–1479. 2001. View Article : Google Scholar : PubMed/NCBI

30 

Takasaki I, Andoh T, Nojima H, Shiraki K and Kuraishi Y: Gabapentin antinociception in mice with acute herpetic pain induced by herpes simplex virus infection. J Pharmacol Exp Ther. 296:270–275. 2001.PubMed/NCBI

31 

Hunter JC, Gogas KR, Hedley LR, Jacobson LO, Kassotakis L, Thompson J and Fontana DJ: The effect of novel anti-epileptic drugs in rat experimental models of acute and chronic pain. Eur J Pharmacol. 324:153–160. 1997. View Article : Google Scholar : PubMed/NCBI

32 

Stanfa LC, Singh L, Williams RG and Dickenson AH: Gabapentin, ineffective in normal rats, markedly reduces C-fibre evoked responses after inflammation. Neuroreport. 8:587–590. 1997. View Article : Google Scholar : PubMed/NCBI

33 

Oku R, Satoh M and Takagi H: Release of substance P from the spinal dorsal horn is enhanced in polyarthritic rats. Neurosci Lett. 74:315–319. 1987. View Article : Google Scholar : PubMed/NCBI

34 

Fehrenbacher JC, Taylor CP and Vasko MR: Pregabalin and gabapentin reduce release of substance P and CGRP from rat spinal tissues only after inflammation or activation of protein kinase C. Pain. 105:133–141. 2003. View Article : Google Scholar : PubMed/NCBI

35 

Heuillet E, Ménager J, Fardin V, Flamand O, Bock M, Garret C, Crespo A, Fallourd AM and Doble A: Characterization of a human NK1 tachykinin receptor in the astrocytoma cell line U 373 MG. J Neurochem. 60:868–876. 1993. View Article : Google Scholar : PubMed/NCBI

36 

Bahrami G and Mohammadi B: Sensitive microanalysis of gabapentin by high-performance liquid chromatography in human serum using pre-column derivatization with 4-chloro-7-nitrobenzofurazan: Application to a bioequivalence study. J Chromatogr B Analyt Technol Biomed Life Sci. 837:24–28. 2006. View Article : Google Scholar : PubMed/NCBI

37 

Jalalizadeh H, Souri E, Tehrani MB and Jahangiri A: Validated HPLC method for the determination of gabapentin in human plasma using pre-column derivatization with 1-fluoro-2,4-dinitrobenzene and its application to a pharmacokinetic study. J Chromatogr B Analyt Technol Biomed Life Sci. 854:43–47. 2007. View Article : Google Scholar : PubMed/NCBI

38 

Park S, Ahn ES, Han DW, Lee JH, Min KT, Kim H and Hong YW: Pregabalin and gabapentin inhibit substance P-induced NF-kappaB activation in neuroblastoma and glioma cells. J Cell Biochem. 105:414–423. 2008. View Article : Google Scholar : PubMed/NCBI

39 

Tremont-Lukats IW, Megeff C and Backonja MM: Anticonvulsants for neuropathic pain syndromes: Mechanisms of action and place in therapy. Drugs. 60:1029–1052. 2000. View Article : Google Scholar : PubMed/NCBI

40 

Mao J and Chen LL: Gabapentin in pain management. Anesth Analg. 91:680–687. 2000. View Article : Google Scholar : PubMed/NCBI

41 

Patel S, Naeem S, Kesingland A, Froestl W, Capogna M, Urban L and Fox A: The effects of GABA(B) agonists and gabapentin on mechanical hyperalgesia in models of neuropathic and inflammatory pain in the rat. Pain. 90:217–226. 2001. View Article : Google Scholar : PubMed/NCBI

42 

Field MJ, Oles RJ, Lewis AS, McCleary S, Hughes J and Singh L: Gabapentin (neurontin) and S-(+)-3-isobutylgaba represent a novel class of selective antihyperalgesic agents. Br J Pharmacol. 121:1513–1522. 1997. View Article : Google Scholar : PubMed/NCBI

43 

Shimoyama N, Shimoyama M, Davis AM, Inturrisi CE and Elliott KJ: Spinal gabapentin is antinociceptive in the rat formalin test. Neurosci Lett. 222:65–67. 1997. View Article : Google Scholar : PubMed/NCBI

44 

Kaneko M, Mestre C, Sánchez EH and Hammond DL: Intrathecally administered gabapentin inhibits formalin-evoked nociception and the expression of Fos-like immunoreactivity in the spinal cord of the rat. J Pharmacol Exp Ther. 292:743–751. 2000.PubMed/NCBI

45 

Field MJ, McCleary S, Hughes J and Singh L: Gabapentin and pregabalin, but not morphine and amitriptyline, block both static and dynamic components of mechanical allodynia induced by streptozocin in the rat. Pain. 80:391–398. 1999. View Article : Google Scholar : PubMed/NCBI

46 

Gajraj NM: Pregabalin: Its pharmacology and use in pain management. Anesth Analg. 105:1805–1815. 2007. View Article : Google Scholar : PubMed/NCBI

47 

Davies A, Hendrich J, Van Minh AT, Wratten J, Douglas L and Dolphin AC: Functional biology of the alpha(2)delta subunits of voltage-gated calcium channels. Trends Pharmacol Sci. 28:220–228. 2007. View Article : Google Scholar : PubMed/NCBI

48 

Luo ZD, Chaplan SR, Higuera ES, Sorkin LS, Stauderman KA, Williams ME and Yaksh TL: Upregulation of dorsal root ganglion (alpha)2(delta) calcium channel subunit and its correlation with allodynia in spinal nerve-injured rats. J Neurosci. 21:1868–1875. 2001.PubMed/NCBI

49 

Fiebich BL, Schleicher S, Butcher RD, Craig A and Lieb K: The neuropeptide substance P activates p38 mitogen-activated protein kinase resulting in IL-6 expression independently from NF-kappa B. J Immunol. 165:5606–5611. 2000. View Article : Google Scholar : PubMed/NCBI

50 

Hu P, Bembrick AL, Keay KA and McLachlan EM: Immune cell involvement in dorsal root ganglia and spinal cord after chronic constriction or transection of the rat sciatic nerve. Brain Behav Immun. 21:599–616. 2007. View Article : Google Scholar : PubMed/NCBI

51 

Scholz J, Abele A, Marian C, Häussler A, Herbert TA, Woolf CJ and Tegeder I: Low-dose methotrexate reduces peripheral nerve injury-evoked spinal microglial activation and neuropathic pain behavior in rats. Pain. 138:130–142. 2008. View Article : Google Scholar : PubMed/NCBI

52 

Cao H and Zhang YQ: Spinal glial activation contributes to pathological pain states. Neurosci Biobehav Rev. 32:972–983. 2008. View Article : Google Scholar : PubMed/NCBI

53 

Obata K and Noguchi K: MAPK activation in nociceptive neurons and pain hypersensitivity. Life Sci. 74:2643–2653. 2004. View Article : Google Scholar : PubMed/NCBI

54 

Arruda JL, Colburn RW, Rickman AJ, Rutkowski MD and DeLeo JA: Increase of interleukin-6 mRNA in the spinal cord following peripheral nerve injury in the rat: Potential role of IL-6 in neuropathic pain. Brain Res Mol Brain Res. 62:228–235. 1998. View Article : Google Scholar : PubMed/NCBI

55 

Lee HL, Lee KM, Son SJ, Hwang SH and Cho HJ: Temporal expression of cytokines and their receptors mRNAs in a neuropathic pain model. Neuroreport. 15:2807–2811. 2004.PubMed/NCBI

56 

Wang XM, Hamza M, Wu TX and Dionne RA: Upregulation of IL-6, IL-8 and CCL2 gene expression after acute inflammation: Correlation to clinical pain. Pain. 142:275–283. 2009. View Article : Google Scholar : PubMed/NCBI

57 

Wang F, Xu S, Shen X, Guo X, Peng Y and Yang J: Spinal macrophage migration inhibitory factor is a major contributor to rodent neuropathic pain-like hypersensitivity. Anesthesiology. 114:643–659. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2017
Volume 16 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yamaguchi K, Kumakura S, Someya A, Iseki M, Inada E and Nagaoka I: Anti‑inflammatory actions of gabapentin and pregabalin on the substance P‑induced mitogen‑activated protein kinase activation in U373 MG human glioblastoma astrocytoma cells. Mol Med Rep 16: 6109-6115, 2017
APA
Yamaguchi, K., Kumakura, S., Someya, A., Iseki, M., Inada, E., & Nagaoka, I. (2017). Anti‑inflammatory actions of gabapentin and pregabalin on the substance P‑induced mitogen‑activated protein kinase activation in U373 MG human glioblastoma astrocytoma cells. Molecular Medicine Reports, 16, 6109-6115. https://doi.org/10.3892/mmr.2017.7368
MLA
Yamaguchi, K., Kumakura, S., Someya, A., Iseki, M., Inada, E., Nagaoka, I."Anti‑inflammatory actions of gabapentin and pregabalin on the substance P‑induced mitogen‑activated protein kinase activation in U373 MG human glioblastoma astrocytoma cells". Molecular Medicine Reports 16.5 (2017): 6109-6115.
Chicago
Yamaguchi, K., Kumakura, S., Someya, A., Iseki, M., Inada, E., Nagaoka, I."Anti‑inflammatory actions of gabapentin and pregabalin on the substance P‑induced mitogen‑activated protein kinase activation in U373 MG human glioblastoma astrocytoma cells". Molecular Medicine Reports 16, no. 5 (2017): 6109-6115. https://doi.org/10.3892/mmr.2017.7368