Oxyntomodulin attenuates TNF‑α induced neuropathic pain by inhibiting the activation of the NF‑κB pathway

  • Authors:
    • Yu Zhang
    • Liyong Yuan
    • Yuebo Chen
    • Chunyan Lin
    • Guangyao Ye
  • View Affiliations

  • Published online on: October 25, 2019     https://doi.org/10.3892/mmr.2019.10770
  • Pages: 5223-5228
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Neuropathic pain is rarely diagnosed. Oxyntomodulin is peripherally and centrally distributed; however, the potential mechanisms underlying the effects of oxyntomodulin in attenuating nociception remain unclear; thus, we aimed to explore them in the present study. A neuropathic pain model in male C57BL/6 mice was induced by intrathecal injection of tumor necrosis factor‑α (TNF‑α), and the duration of nociceptive behavioral responses was measured with a stop‑watch timer within 30 min. Western blotting was used to explore the protein levels of ionized calcium binding adaptor molecule‑1 (IBA1), nuclear factor‑κB (NF‑κB) phosphorylated‑p65, interleukin (IL)‑6 and IL‑1β. We performed reverse transcription‑quantitative polymerase chain reaction and ELISA were performed to determine the mRNA and protein expression levels of IL‑6 and IL‑1β, respectively. An MTT assay was conducted to detect BV2 cell viability. Oxyntomodulin was observed to attenuate TNF‑α‑induced pain hypersensitivity in mice, as well as the expression of IBA1, NF‑κB p‑p65, IL‑6 and IL‑1β in the spinal cord. Oxyntomodulin exhibited no cytotoxicity on BV2 cells, and attenuated TNF‑α‑induced IL‑6 and IL‑1β production and release in BV2 cells and culture medium, respectively. Collectively, we proposed oxyntomodulin to attenuate TNF‑α induced neuropathic pain associated with the release of glial cytokines IL‑6 and IL‑1β via inhibiting the activation of the NF‑κB pathway.

Introduction

Neuropathic pain is defined as ‘pain caused by a lesion or disease of the somatosensory system’ by the International Association for the Study of Pain (1). Nociceptive pain involves peripheral noxious stimulation, for instance, by inflammatory factors (2). In 174 clinical trials, the efficacies of multiple drugs with analgesic effects, such as, local anesthetic drugs, opioids, antidepressants, NMDA receptor antagonists, botulinum toxin and topical capsaicin have been compared with placebo agents in patients with neuropathic pain (3). Despite the high prevalence of neuropathic pain, it is rarely diagnosed, and adequately and effectively treated. Thus, is important to develop novel potential therapies for treating patients with neuropathic pain.

Tumor necrosis factor (TNF)-α, which is a major proinflammatory cytokine and is produced during the transmission of pain (4), serves a role in the pathogenesis of neuropathic pain (5). Spinal nerve injury in rats induces mechanical allodynia and thermal hyperalgesia, which are accompanied with concomitant increases of interleukin (IL)-1β and TNF-α in the brain (6).

Increasing evidence has suggested that oxyntomodulin is peripherally and centrally distributed (7,8). Oxyntomodulin exhibits numerous functions. For instance, in mice administered a high-fat diet, intraperitoneal and intravenous treatment of oxyntomodulin ameliorates glucose intolerance (9), and increases insulin/glucose density in the plasma (10); additionally, oxyntomodulin regulates the intrinsic heart rate of mice (11). Furthermore, oxyntomodulin exhibits antinociceptive effects in mice (12). However, the potential molecules responsible for the attenuation of nociception by oxyntomodulin require further investigation of which we aimed to determine in the present study.

Materials and methods

Ethics statement

Each experimental procedure in the present study was approved by the Ethical Committee for the Use of Laboratory Animals of Ningbo No. 6 Hospital, and was conducted strictly according to the ‘Guide for Care and Use of Laboratory Animals published by the National Institutes of Health (13) and the ethical guidelines of the International Association for the Study of Pain (14)’.

Reagents

For in vivo experiments, 5 µl oxyntomodulin (0.2 µg/µl; Phoenix Pharmaceuticals) was dissolved in saline and administered to mice via an intrathecal (i.t.) injection according to a previous study (12). A total of 5 µl TNF-α (20 pg/µl; Sigma-Aldrich; Merck KGaA) was i.t. injected into mice (12).

For in vitro experiments, BV2 cells were purchased from American Type Culture Collection and were treated with 0.01, 0.1 and 1 µM of oxyntomodulin (15). Then, 1 pg/µl TNF-α was added into the culture medium (16).

Animals

A total of 24 adult (8-week old, weight 20 g) male C57BL/6 mice were obtained from and housed in Ningbo No.6 Hospital. Mice were housed under controlled conditions with a temperature of 23±1°C, humidity of 70±10% and a light-dark cycle of 12 h, with ad libitum access to water and food. All of the experiments in the current study were performed in the light phase of the light/dark cycle.

Mice were injected intrathecally with oxyntomodulin to determine the potential antinociceptive effects of oxyntomodulin. Thereafter, 24 mice were randomly divided into three different groups as described in a previously reported study (12): Control group, i.t. injection of 5 µl saline and 5 µl saline (n=8); TNF-α group, i.t. injection with 5 µl saline and 5 µl TNF-α (20 pg/µl) (n=8); oxyntomodulin + TNF-α group, i.t. injection with 5 µl oxyntomodulin (0.2 µg/µl) prior to 5 µl TNF-α (20 pg/µl) (n=8). Each mouse was euthanized by thoracic dislocation directly after the behavior tests (described below).

Nociceptive behavioral testing

Mice were acclimatized to the new condition for nociceptive behavioral testing for ~1 h before the experiments. After i.t. injection with TNF-α, mice were placed in a chamber (20 cm high, 20 cm diameter). Within half an hour, the cumulative response time of nociceptive behavioral responses of each mouse, including licking, scratching and biting episodes to the lumbar and caudal spinal cord were recorded with a stop-watch timer (17).

Western blotting

Lumbar 4–6 spinal cords were obtained mixed with radioimmunoprecipitation assay lysis (Roche Diagnostics) buffer containing phosphatase and proteinase inhibitors (Roche Diagnostics). The concentration of each protein sample was determined with a BCA kit (Beyotime Institute of Biotechnology). Protein samples (15 µg) were separated by 8% SDS-PAGE followed by transferring to a polyvinylidene difluoride (PVDF) membrane (EMD Millipore). The membranes were incubated with corresponding primary antibodies against IL-6 (cat. no. 12912; dilution 1:1,000), IL-1β (cat. no. 31202; dilution 1:1,000), phosphorylated-NF-κB (Ser536, cat. no. 3033; dilution 1:1,000), p65 (cat. no. 59674; dilution 1:1,000) and GAPDH (cat. no. 5174; dilution 1:1,000), which acted as a loading control. All the above primary antibodies were obtained from Cell Signaling Technology, Inc. and applied at 4°C overnight. The antibody for IBA1 (cat. no. ab178846; dilution 1:1,000) was obtained from Abcam and applied at 4°C overnight. Prior, the PVDF membrane was blocked with 5% skim milk at room temperature for ~1 h. The next day, the PVDF membrane was incubated with an anti-rabbit horseradish peroxidase-conjugated secondary antibody (cat. no. 7074; dilution 1:200, Cell Signaling Technology, Inc.) at room temperature for 1 h. Then, the protein bands were visualized via enhanced chemiluminescence (Thermo Fisher Scientific, Inc.). The quantity of each target protein band was analyzed by Image Quant LAS 500 (GE Healthcare Life Sciences).

Reverse transcription-quantitative-polymerase chain reaction (RT-qPCR)

Total RNA was extracted from the lumbar 4–6 spinal cords and BV2 cells using TRIzol® (Thermo Fisher Scientific, Inc.). cDNA was synthesized from RNA with a PrimerScript RT reagent kit (Takara Bio, Inc.) according to the manufacturer's protocols. RT-qPCR was performed by SYBR Premix Ex Taq (Takara Bio, Inc.) and conducted by a CFX96™ real-time PCR system (Bio-Rad Laboratories, Inc.). The thermocycling conditions were: Denaturation (95°C, 5 min), followed by 36 cycles of amplification (95°C, 25 sec) and quantification (62°C, 40 sec), the melting curve was them obtained (95°C, 25 sec and 60°C, 1 min). The 2−ΔΔCq method was used to quantify the expression level of each gene (18). GAPDH acted as a control. The primer sequences were as listed: IL-6, forward, 5′-GGCTACTGCTTTCCCTACCC-3′ and reverse, 5′-TTTTCTGCCAGTGCCTCTT-3′; IL-1β, forward, 5′-TCCAGGATGAGGACATGAGCAC-3′ and reverse, 5′-GAACGTCACCCAGCAGGTTA-3′; GAPDH, forward, 5′-GTGATGCTGGTGCTGAGTATC-3′ and reverse, 5′-GTGATGGCATGGACKGTGG-3′.

Cell culture

BV2 microglial cells were seeded into 6-well plates, cultured with Dulbecco's Modified Eagle's medium (DMEM; Thermo Fisher Scientific, Inc.) supplemented with 10% fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc.), 1% penicillin/streptomycin (Invitrogen; Thermo Fisher Scientific, Inc.). Cells were maintained at 37°C in an incubator with 5% CO2 for 24 h. Cells (3×105) were divided into three groups and maintained at 37°C in an incubator with 5% CO2 with the treatments as followed: Control group, cultured with only culture medium; TNF-α group, treated with 1 pg/µl TNF-α at 37°C for 24 h (16); oxyntomodulin + TNF-α group, treated with 0.01, 0.1 and 1 µM of oxyntomodulin at 37°C at 24 h (15) prior to the treatment of 1 pg/µl TNF-α.

MTT assay

The effects of oxyntomodulin on BV2 cell viability were analyzed via an MTT assay (Sigma-Aldrich; Merck KGaA). Cells (4×106 cells/well) were first incubated for 8 h at 37°C in an incubator with 5% CO2. Thereafter, 10 µl MTT was added into each well and cells were incubated for further 4 h at 37°C. The optical density (OD) at 570 nm was measured with a microplate reader (Bio-Rad 550; Bio-Rad Laboratories, Inc.).

ELISA

Supernatants of the culture medium were cleared of cells by spinning the 96-well plates at 400 × g at room temperature for 5 min. The supernatants were transferred to the round bottom, non-treated 96-well plates followed by spinning at 400 × g at room temperature for 5 min. Thereafter, the supernatants were transferred to a new 96-well plate for the subsequent experiments. The protein expression levels of IL-1β (SMLB00C, mouse IL-1 β/IL-1F2 Quantikine ELISA Kit) and IL-6 (SM6000B, mouse IL-6 Quantikine ELISA Kit) in the supernatants of the culture medium were tested by ELISA (R&D Systems, Inc.) according to the manufacturer's protocols. The OD at 490 nm was measured with a microplate reader (Spectra MAX 340PC Molecular Devices LLC).

Statistical analysis

Each experiment was repeated three times. Statistical analyses were conducted using GraphPad Prism 6 software (GraphPad Software, Inc.). Data were presented as the mean ± standard error of the mean. One-way ANOVA followed by a Student-Newman-Keuls test were applied for the analyses of differences among 3 or more groups. P<0.05 indicated a statistically significant difference.

Results

Oxyntomodulin attenuates TNF-α-induced pain hypersensitivity

The effects of oxyntomodulin on TNF-α-induced nociceptive behaviors in mice were assessed by nociceptive behavioral testing with a stop-watch timer. Compared with mice in the control group, mice in the TNF-α group presented acute licking, scratching, and biting the lumbar or caudal region which lasted for about half an hour; this was significantly longer than the control. On the contrary, a significant decrease in the duration of nociceptive behavior was reported following oxyntomodulin administration compared with TNF-α treatment alone (Fig. 1).

Oxyntomodulin decreases TNF-α-induced expression of IBA-1 and NF-κB p-p65 in the spinal cord

Over the past 15 years, neuropathic conditions have been proved to be related with the activation of glia in the central nervous system (CNS), which are important contributors to central sensitization (19). Molecular changes caused by glial activation leads to pain hypersensitivity, including upregulation of chemokine receptors and the release of glial cytokines (20).

Nuclear factor-κB is activated by IκB kinases, which then migrates to the nucleus (21). Spinal nerve injury in rats increases mechanical allodynia and thermal hyperalgesia which are accompanied with increases in NF-κB expression in the brain (6).

Therefore, we explored the expression levels of ionized calcium binding adaptor molecule-1 (IBA1, microglia marker) and NF-κB in the spinal cord of mice by western blotting. The results showed that, compared with the control group, TNF-α significantly induced the protein expression of IBA1 and NF-κB p-p65, but was attenuated by pretreatment with oxyntomodulin (Fig. 2A and B); no significant changes in NF-κB p65 expression were observed among the three groups.

Oxyntomodulin attenuates TNF-α-induced IL-6 and IL-1β production in the spinal cord

NF-κB activates the transcription of proinflammatory genes (18), while, NF-κB responds to ~150 different stimuli (22). Spinal nerve injury in rats induces mechanical allodynia and thermal hyperalgesia accompanied with concomitant increases of IL-1β and TNF-α in the brain (6). Several studies indicate that various cytokines can directly stimulate nociceptors (23,24). Cytokines (such as IL-1β and TNF-α) which are released after stress induce the activation of NF-κB (25).

Correspondingly, we detected the expression levels of IL-1β and TNF-α in the spinal cord of mice by RT-qPCR and western blotting. RT-qPCR demonstrated that compared with the control group, TNF-α significantly induced IL-6 and IL-1β mRNA expression in spinal cord, but was attenuated by oxyntomodulin administration (Fig. 3A and B). Western blotting demonstrated similar results to the findings of RT-qPCR (Fig. 3C and D).

Oxyntomodulin protects BV2 cells from TNF-α-induced toxicity

BV2 cell viability in different groups was investigated by an MTT assay. At 12 h after treatment, oxyntomodulin (0.01, 0.1 and 1 µM) did not affect BV2 cell viability as presented in Fig. 4A, indicating that oxyntomodulin exerted no toxicity on BV2 cells.

Compared with the control group, TNF-α significantly inhibited the cell viability of BV2 cells, which was rescued by oxyntomodulin treatment (0.01, 0.1 and 1 µM) (Fig. 4B).

Oxyntomodulin attenuates TNF-α-induced IL-6 and IL-1β production and release in BV2 cells and culture medium

Additionally, we detected the effects of oxyntomodulin on the release of glia cytokine. RT-qPCR was performed to determine the mRNA levels of IL-6 and IL-1β in BV2 cells. Compared with the control group, TNF-α significantly increased the mRNA levels of IL-6 and IL-1β in BV2 cells, which were significantly attenuated by oxyntomodulin treatment (0.1 and 1 µM) (Fig. 5A and B).

Furthermore, ELISA was used for the analyze the protein levels of IL-6 and IL-1β in the supernatants of the culture medium. The results demonstrated that compared with control group, TNF-α significantly increased protein levels of IL-6 and IL-1β, which were attenuated by oxyntomodulin treatment (0.1 and 1 µM) in the supernatants of culture medium (Fig. 5C and D).

Discussion

The effects of oxyntomodulin in the CNS have not been well elucidated (26). One previous study indicated that D-Ser2-oxytomodulin (a protease-resistant oxyntomodulin analogue) functions in the CNS. For instance, it improves locomotor activities and protects dopaminergic neurons in a mouse model of Parkinson's Disease (PD) (27). Although oxyntomodulin is expressed in the brain and the spinal cord (7,8), the function and potential molecules responsible for the function of oxyntomodulin in nociception remain to be further explored.

TNF-α which is a major proinflammatory cytokine that is produced during the transmission of pain (4), serves a role in the pathogenesis of neuropathic pain (5). In the present study, we reported that oxyntomodulin attenuated TNF-α-induced neuropathic pain in mice in nociceptive behavior tests; however, the molecules that are responsible for the aforementioned changes remain to be further investigated.

As an important upstream molecule of NF-κB, TNF-α activates NF-κB, which further promotes the expression of IL-6 (28); TNF-α also leads to an increase of IL-1β expression (29). NF-κB plays a crucial role in the transcriptional regulation of various pro-inflammatory genes, including IL-1β and IL-6 (30). Our results showed that, compared with the control group, TNF-α induced the expression of NF-κB, as well as IL-6 and IL-1β, which were in agreement with the aforementioned findings, indicating that TNF-α induced the expression of IL-6 and IL-1β via activation of NF-κB (2830).

Furthermore, the effects of TNF-α on nociceptive behaviors, and NF-κB, IL-6 and IL-1β levels were found to be attenuated by pretreatment of oxyntomodulin in the present study. These findings were in agreement with a recent study (12) and a report that investigated the potential treatment of oxyntomodulin on PD (27).

In conclusion, we revealed for the first time, to the best of our knowledge, that oxyntomodulin attenuates TNF-α-induced neuropathic pain (release of glial cytokines IL-6 and IL-1β) via inhibiting the activation of NF-κB pathway, suggesting a potential role of oxyntomodulin the clinical management of pain in the future.

Unfortunately, there are several limitations in the present study: i) The time-dependent effects of oxyntomodulin on BV2 were not investigated, which could provide insight into the effects of intracellular cytokines; and ii) little evidence regarding the effects of oxyntomodulin on TNF-α-induced neuropathic pain has been reported; thus; further analysis should be conducted in the future.

Acknowledgements

Not applicable.

Funding

The present study was funded by Natural Science Foundation of Ningbo (grant no. 2009A610140).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

YZ, LY, YC, CL and GY performed the experiments and analyzed the data. LY prepared the manuscript.

Ethics approval and consent to participate

The present study was approved by the Ethics Committee for the Use of Laboratory Animals of Ningbo No. 6 Hospital, and was conducted strictly according to the ‘Guide for Care and Use of Laboratory Animals’ published by the National Institutes of Health (13) and the ethical guidelines of the International Association for the Study of Pain (14).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Jensen TS, Baron R, Haanpää M, Kalso E, Loeser JD, Rice AS and Treede RD: A new definition of neuropathic pain. Pain. 152:2204–2205. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Woolf CJ: American College of Physicians; American Physiological Society Pain: Moving from symptom control toward mechanism-specific pharmacologic management. Ann Intern Med. 140:441–451. 2004. View Article : Google Scholar : PubMed/NCBI

3 

Finnerup NB, Sindrup SH and Jensen TS: The evidence for pharmacological treatment of neuropathic pain. Pain. 150:573–581. 2010. View Article : Google Scholar : PubMed/NCBI

4 

Vicario N, Parenti R, Arico G, Turnaturi R, Scoto GM, Chiechio S and Parenti C: Repeated activation of delta opiod receptors counteracts nerve injury-induced TNF-α up-regulation in the sciatic nerve of rats with neuropathic pain: A possible correlation with delta opiod receptors-mediated antiallodinic effect. Mol Pain. 12:17448069166679492016. View Article : Google Scholar : PubMed/NCBI

5 

Leung L and Cahill CM: TNF-alpha and neuropathic pain-a review. J Neuroinflammation. 7:272010. View Article : Google Scholar : PubMed/NCBI

6 

Xie W, Luo S, Xuan H, Chou C, Song G, Lv R, Jin Y, Li W and Xu J: Betamethasone affects cerebral expressions of NF-kappaB and cytokines that correlate with pain behavior in a rat model of neuropathy. Ann Clin Lab Sci. 36:39–46. 2006.PubMed/NCBI

7 

Blache P, Kervran A and Bataille D: Oxyntomodulin and glicentin: Brain-gut peptides in the rat. Endocrinology. 123:2782–2787. 1988. View Article : Google Scholar : PubMed/NCBI

8 

Vrang N and Larsen PJ: Preproglucagon derived peptides GLP-1, GLP-2 and oxyntomodulin in the CNS: Role of peripherally secreted and centrally produced peptides. Prog Neurobiol. 92:442–462. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Parlevliet ET, Heijboer AC, Schroder-van der Elst JP, Havekes LM, Romijn JA, Pijl H and Corssmit EP: Oxyntomodulin ameliorates glucose intolerance in mice fed a high-fat diet. Am J Physiol Endocrinol Metab. 294:E142–E147. 2008. View Article : Google Scholar : PubMed/NCBI

10 

ThanThan S, Zhao H, Yannaing S, Ishikawa T and Kuwayama H: Oxyntomodulin increases the concentrations of insulin and glucose in plasma but does not affect ghrelin secretion in Holstein cattle under normal physiological conditions. Domest Anim Endocrinol. 39:163–170. 2010. View Article : Google Scholar : PubMed/NCBI

11 

Sowden GL, Drucker DJ, Weinshenker D and Swoap SJ: Oxyntomodulin increases intrinsic heart rate in mice independent of the glucagon-like peptide-1 receptor. Am J Physiol Regul Integr Comp Physiol. 292:R962–R970. 2007. View Article : Google Scholar : PubMed/NCBI

12 

Park SH, Lee JR, Jang SP, Park SH, Lee HJ, Hong JW and Suh HW: Antinociceptive profiles and mechanisms of centrally administered oxyntomodulin in various mouse pain models. Neuropeptides. 68:7–14. 2018. View Article : Google Scholar : PubMed/NCBI

13 

Guide for the Care and Use of Laboratory Animals, . 8th. The National Academies Collection: Reports funded by National Institutes of HealthNational Academies Press; Washington, DC: 2011

14 

Ethical standards for investigations of experimental pain in animals. The Committee for Research and Ethical issues of the international association for the study of pain. Pain. 9:141–143. 1980. View Article : Google Scholar : PubMed/NCBI

15 

Li Y, Wu KJ, Yu SJ, Tamargo IA, Wang Y and Greig NH: Neurotrophic and neuroprotective effects of oxyntomodulin in neuronal cells and a rat model of stroke. Exp Neurol. 288:104–113. 2017. View Article : Google Scholar : PubMed/NCBI

16 

El Karim I, McCrudden MT, Linden GJ, Abdullah H, Curtis TM, McGahon M, About I, Irwin C and Lundy FT: TNF-α-induced p38MAPK activation regulates TRPA1 and TRPV4 activity in odontoblast-like cells. Am J Pathol. 185:2994–3002. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Hylden JL and Wilcox GL: Intrathecal substance P elicits a caudally-directed biting and scratching behavior in mice. Brain Res. 217:212–215. 1981. View Article : Google Scholar : PubMed/NCBI

18 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

19 

Tsuda M, Inoue K and Salter MW: Neuropathic pain and spinal microglia: A big problem from molecules in ‘small’ glia. Trends Neurosci. 28:101–107. 2005. View Article : Google Scholar : PubMed/NCBI

20 

Ji RR, Berta T and Nedergaard M: Glia and pain: Is chronic pain a gliopathy? Pain. 154 (Suppl 1):S10–S28. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Ghosh S and Karin M: Missing pieces in the NF-kappaB puzzle. Cell. 109 (Suppl 1):S81–S96. 2002. View Article : Google Scholar : PubMed/NCBI

22 

Pahl HL: Activators and target genes of Rel/NF-kappaB transcription factors. Oncogene. 18:6853–6866. 1999. View Article : Google Scholar : PubMed/NCBI

23 

Obreja O, Rathee PK, Lips KS, Distler C and Kress M: IL-1 beta potentiates heat-activated currents in rat sensory neurons: Involvement of IL-1RI, tyrosine kinase, and protein kinase C. FASEB J. 16:1497–1503. 2002. View Article : Google Scholar : PubMed/NCBI

24 

Parada CA, Yeh JJ, Joseph EK and Levine JD: Tumor necrosis factor receptor type-1 in sensory neurons contributes to induction of chronic enhancement of inflammatory hyperalgesia in rat. Eur J Neurosci. 17:1847–1852. 2003. View Article : Google Scholar : PubMed/NCBI

25 

Allan SM and Rothwell NJ: Inflammation in central nervous system injury. Philos Trans R Soc Lond B Biol Sci. 358:1669–1677. 2003. View Article : Google Scholar : PubMed/NCBI

26 

Bataille D and Dalle S: The forgotten members of the glucagon family. Diabetes Res Clin Pract. 106:1–10. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Liu W, Li Y, Jalewa J, Saunders-Wood T, Li L and Hölscher C: Neuroprotective effects of an oxyntomodulin analogue in the MPTP mouse model of Parkinson's disease. Eur J Pharmacol. 765:284–290. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Lee KM, Jeon SM and Cho HJ: Tumor necrosis factor receptor 1 induces interleukin-6 upregulation through NF-kappaB in a rat neuropathic pain model. Eur J Pain. 13:794–806. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Segond von Banchet G, König C, Patzer J, Eitner A, Leuchtweis J, Ebbinghaus M, Boettger MK and Schaible HG: Long-lasting activation of the transcription factor CREB in sensory neurons by interleukin-1β during Antigen-induced arthritis in rats: A mechanism of persistent arthritis pain? Arthritis Rheumatol. 68:532–541. 2016. View Article : Google Scholar : PubMed/NCBI

30 

Hsing CH, Lin MC, Choi PC, Huang WC, Kai JI, Tsai CC, Cheng YL, Hsieh CY, Wang CY, Chang YP, et al: Anesthetic propofol reduces endotoxic inflammation by inhibiting reactive oxygen species-regulated Akt/IKKβ/NF-κB signaling. PLoS One. 6:e175982011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2019
Volume 20 Issue 6

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
Zhang Y, Yuan L, Chen Y, Lin C and Ye G: Oxyntomodulin attenuates TNF‑α induced neuropathic pain by inhibiting the activation of the NF‑κB pathway. Mol Med Rep 20: 5223-5228, 2019
APA
Zhang, Y., Yuan, L., Chen, Y., Lin, C., & Ye, G. (2019). Oxyntomodulin attenuates TNF‑α induced neuropathic pain by inhibiting the activation of the NF‑κB pathway. Molecular Medicine Reports, 20, 5223-5228. https://doi.org/10.3892/mmr.2019.10770
MLA
Zhang, Y., Yuan, L., Chen, Y., Lin, C., Ye, G."Oxyntomodulin attenuates TNF‑α induced neuropathic pain by inhibiting the activation of the NF‑κB pathway". Molecular Medicine Reports 20.6 (2019): 5223-5228.
Chicago
Zhang, Y., Yuan, L., Chen, Y., Lin, C., Ye, G."Oxyntomodulin attenuates TNF‑α induced neuropathic pain by inhibiting the activation of the NF‑κB pathway". Molecular Medicine Reports 20, no. 6 (2019): 5223-5228. https://doi.org/10.3892/mmr.2019.10770