LMTK2 regulates inflammation in lipopolysaccharide‑stimulated BV2 cells

  • Authors:
    • Qianyun Rui
    • Shugang Cao
    • Xiaozhu Wang
    • Xiaoyu Duan
    • Xinyi  Iao
    • Wanli Dong
    • Qi Fang
    • Xueguang Zhang
    • Qun Xue
  • View Affiliations

  • Published online on: January 18, 2021     https://doi.org/10.3892/etm.2021.9621
  • Article Number: 219
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Microglia activation plays vital roles in neuroinflammatory pathologys. Lemurs tyrosine kinase 2 (LMTK2) was reported to regulate NF‑κB signals. In the present study, the roles of LMTK2 were investigated in lipopolysaccharide (LPS)‑treated BV‑2 cells. Reverse transcription‑quantitative (RT‑q)PCR and western blotting (WB) were utilized to analyze LMTK2 levels in LPS‑treated BV2 cells. MTT assay determined cell viabilities. Nitric oxide (NO) and prostaglandin E2 (PGE2) levels were assessed through Griess and enzyme‑linked immunosorbent assay (ELISA), respectively. The expression level of inducible NO synthase (iNOS) and cyclooxygenase‑2 (COX‑2) were detected through RT‑qPCR and WB. The release of inflammatory mediators under LPS stimulation, tumor necrosis factor‑α (TNF‑α), interleukin‑1β (IL‑1β), IL‑6 and IL‑10, were analyzed through ELISA. WB was used to analyze the nuclear factor erythroid 2‑related factor 2 (Nrf2)/heme oxygenase 1 (HO‑1)/NAD(P)H dehydrogenase quinone 1 (NQO1) signal pathway. The results showed that the levels of the inflammatory mediators, iNOS, NO, COX‑2 and PGE2, along with pro‑inflammatory factors, TNF‑α, IL‑1β and IL‑6, were significantly decreased following the induction of exogenous LMTK2 expression by LMTK2 overexpression plasmids in LPS‑induced BV2 microglia. In contrast, anti‑inflammatory factor IL‑10 showed obvious decrease. Additionally, LMTK2 overexpression induced the elevation of Nrf2 in the cytoplasm and nucleus, along with the upregulation of HO‑1 and NQO1 expression. In conclusion, LMTK2 is postulated to regulate neuroinflammation possibly through Nrf2 pathway. The present study is essential to reveal the underlying function of LMTK2 and to identify novel therapeutic targets for drug development in treating neuroinflammation.

Introduction

Lemurs tyrosine kinase 2 (LMTK2) belongs to the transmembrane serine/threonine protein kinase family anchoring membrane with unique structure (1,2). LMTK2, widely expressed in brain, is involved in regulating key cellular events, apoptosis and cell differentiation (3-7). According to a review, LMTK2 affects the sensitivities of cells to cytotoxicity depending on apoptotic and survival pathways (3). The latest research shows that LMTK2 can activate the NRF/ARE signaling pathway to reduce neurons injury induced by ischemia reperfusion (8). LMTK2 could be phosphorylated through CDK5/p35 in neurons, however, there is only a small amount of the phosphorylated forms of LMTK2 in non-neuron cells with inactive CDK5 (9-12). Furthermore, LMTK2 can exert vital roles through phosphorylating downstream targets in non-neuron cells (3). It has been proved that LMTK2 regulates NF-κB signals through the PP1/GSK3β/p65 pathway or PP1/IKK pathway in colon cancer cells (13).

Neuroinflammation is associated with the progression of multiple neurological diseases. Microglia, as the main immune effector cells of the central nervous system (CNS), play a substantial role in CNS diseases (14). Although microglia have essential neuroprotection functions, including sensing changes of the environment, maintaining normal neuronal function and defensing these changes, they can damage neurons in response to a particular stimulus or with neuroinflammation (15). Activated microglia are involved in the pathologic processes of CNS diseases, such as neurodegenerative disease, pain, infection and brain trauma (16-19). The activation of microglia could damage neurons in the brain by releasing inflammatory cytokines and generating oxidative stress, which further triggers neurological diseases (20,21). Therefore, the present study aims to explore the role of LMTK2 in lipopolysaccharide (LPS)-induced microglia inflammation and to explore whether it can activate Nrf2 signaling. LPS was used to activate mouse microglia (BV2) cells to construct a cell model of neuroinflammation.

Materials and methods

Cells

Mouse microglial cells, BV2, were purchased (The Institute of Cell Biology, Chinese Academy of Sciences, China) and cultured with DMEM, a high-glucose medium containing 10% fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc.), at a constant temperature of 37˚C in 5% CO2. The microglia were used in the experiment when the cells reached the logarithmic growth stage.

Western blotting (WB)

BV2 cell suspension was placed in a 6-well plate (2x105 cells/ml). The cells were stimulated with LPS at different concentrations (100, 200 and 500 ng/ml; Sigma-Aldrich; Merck KGaA) for 24 h. For LMTK2 overexpression, the cells were transfected with plasmids overexpressing LMTK2. After 24 h, the cells were stimulated with LPS (500 ng/ml) for 24 h. Subsequently, cells were collected to extract total protein using RIPA lysis solution (cat. no. R0278; Sigma-Aldrich; Merck KGaA). The protein concentrations were detected by BCA method. Then, 10% SDS-PAGE electrophoresis was performed to separate the proteins (40 µg protein in each well). Skim milk powder (5%) was utilized to block the PVDF membrane for 40 min at room temperature. Primary antibodies (LMTK2 (1:500; cat. no. DF3344; Affinity Biosciences), inducible nitric oxide synthase (1:1,000; iNOS; cat. no. ab178945; Abcam), cyclooxygenase 2 (1:1,000; COX2; cat. no. ab179800), nuclear factor erythroid 2-related factor 2 (1:1,000; NRF2; ab137550; Abcam), heme oxygenase-1 (1:2,000; HO-1; cat. no. ab189491; Abcam), NAD(P)H dehydrogenase quinone 1 (1:20,000; NQO1; cat. no. ab28947; Abcam), Histone H3 (1:2,000; cat. no. ab1791; Abcam), GAPDH (1:5,000; cat. no. ab8245; Abcam) were incubated with the membrane at 4˚C overnight. This was followed by incubation with the secondary antibodies goat anti-rabbit IgG (1:10,000; cat. no. ab6721; Abcam) and rabbit anti-mouse IgG (1:10,000; cat. no. ab6728; Abcam) at room temperature for 1 h. ECL was used to visualize the protein bands which then was quantified with ImageJ 1.52v software (National Institutes of Health).

Plasmids transfection

The plasmids overexpressing LMTK2 were constructed by Shanghai GenePharma Co., Ltd. BV2 cells were seeded into 6-well plates (2x105 cells/ml). LMTK2 overexpression plasmids (Oe-LM; 4 µg) and empty plasmids (Oe-NC; 4 µg) were respectively transfected into BV2 cells using Lipofectamine™3000 (Invitrogen; Thermo Fisher Scientific, Inc.) according to manufacturer's protocol. After transfection of 24 h at 37˚C, cells were activated by LPS (500 ng/ml).

Reverse transcription-quantitative (RT-q)PCR

BV2 cell suspension was placed in a 6-well plate (2x105 cells/ml). After experimental treatment, total RNA was extracted using TRIzol method and reverse transcribed into cDNA 42˚C for 1 h (RevertaidTM First Strand cDNA Synthesis kit, Fermentas; Thermo Fisher Scientific, Inc.). Relative determination of LMTK2 and GADPH mRNA was performed by SYBR-Green dye method (Clontech Laboratories, Inc.) and calculated using the 2-ΔΔCq method (22). The primers of LMTK2 mRNA were as follows: Forward, 5'-TTGCCCGCCACAGTCTAAAC-3' and reverse, 5'-GATGACTCTTGCTACGCTAGT-3'; The primers of GADPH mRNA were as follows: Forward 5'-GCCTTCCGTGTTCCTACCC -3' and reverse 5'-TGCCTG CTTCACCACCTTC-3'. The target mRNA was amplified in the following thermocycling conditions: Predenaturation at 95˚C for 3 min; 30 cycles of 95˚C 30 sec, 58˚C for 30 sec and 72˚C for 1 min. The total extension at 72˚C for 10 min.

MTT assay

Cells viability was detected as per the manufacturer's protocol. BV2 cells were seeded into 96-well plates (1x105 cells/ml). After cells were treated, MTT solution of 5 g/l (APExBIO Technology LLC) was added and placed in an incubator at 37˚C and 5% CO2 for 4 h. The dimethyl sulfoxide (DMSO) solution of 100 µl was added to each well. After shaking the mixture, the absorbance (A) value at the wavelength of 490 nm was detected by a microplate reader and the cell survival rate was calculated.

Griess assays

NO2 was formed from NO in aqueous and reacted with Griess reagent (Beyotime Institute of Biotechnology). Therefore, NO levels in the supernatant were indirectly detected. Cells were seeded into 24-well plates (2x105 cells/ml). After LPS treatment (500 ng/ml) for 24 h, the supernatant in the medium was collected and then 50 µl of Griess reagent was supplemented into the supernatant. After 10 min, the absorbance at 540 nm was detected.

Enzyme-linked immunosorbent assay (ELISA)

BV2 cells were cultured in 6-well plates (2x105 cells/ml). After treatment, the supernatant in medium was collected. Interleukin (IL)-1β, IL-6 and IL-10 levels were analyzed through ELISA kits (Mouse IL-1β ELISA kit; cat. no. PI301; IL-6, Mouse IL-6 ELISA kit; cat. no. PI326; and IL-10; Mouse IL-10 ELISA kit, cat. no. PI522; all from Beyotime Institute of Biotechnology), along with the detection of tumor necrosis factor (Mouse TNF-α ELISA Standard Recombinant Protein; cat. no. 29-8321-65; Invitrogen; Thermo Fisher Scientific, Inc.) and prostaglandin E2 (PGE2) levels (Prostaglandin E2 ELISA; ab133021; Abcam).

Statistical analysis

GraphPad Prism 8.0 software (GraphPad Software, Inc.) was used for statistical analysis of data, and one-way ANOVA was used to perform the comparison among groups, followed by Turkey's test. P<0.05 was considered to indicate a statistically significant difference.

Results

LMTK2 was significantly decreased in LPS-induced BV2 cells

LPS of different concentrations (100, 200 and 500 ng/ml) was used to stimulate BV2 cells. The LMTK2 protein and mRNA levels presented a gradual decrease with the increasing dose of LPS (Fig. 1A and B). Therefore, 500 ng/ml LPS was utilized to perform the follow-up experiments. Subsequently, the plasmids Oe-LM and Oe-NC were utilized to pre-treat BV2 cells. A significant upregulation of LMTK2 mRNA levels was observed in cells transfected with Oe-LM. (Fig. S1). Following plasmid transfection for 24 h, LPS was used to activate BV2 cells. A marked upregulation of LMTK2 protein and mRNA levels was observed in BV2 cells transfected with Oe-LM compared with Oe-NC (Fig. 1C and D).

LMTK2 overexpression notably decreased the levels of pro-inflammatory mediators in LPS-stimulated BV2 cells

In subsequent experiments, cell viability was evaluated through MTT assay in BV2 cells stimulated with LPS. Compared with the control group, LPS stimulation increased the cell viability of BV2 cells (Fig. 2A). As shown in a previous study (23), the levels of cytoskeletal protein α-tubulin and Iba1 were significantly increased in BV2 cells following LPS induction. Moreover, succinic acid dehydrogenase decreased exogenous MTT into water-insoluble blue-purple crystal formazan, contributing to the increase in OD value. In the present study, LMTK2 overexpression significantly decreased cell viability compared with cells treated with LPS alone (Fig. 2A). Subsequently, it was observed that the levels of proinflammatory mediators, consisting of NO generated by iNOS, PGE2 generated by COX-2), iNOS and COX-2, showed significant decreases in the presence of LMTK2 overexpression in LPS-activated BV2 cells (Fig. 2B-E).

The overexpression of LMTK2 regulated the release of inflammatory factors in LPS-induced BV2 cells

The levels of proinflammatory and anti-inflammatory factors were analyzed in LPS-treated BV2 cells with or without transfection of Oe-LM plasmids. As the result displayed, the proinflammatory mediators, TNF-α, IL-1β and IL-6, were markedly decreased in response to LMTK2 overexpression compared with LPS treatment alone (Fig. 3A). Subsequently, Nrf2 signals were analyzed by detecting the expression of Nrf2 and its downstream genes (HO-1 and NQO1). Nrf2 was upregulated in the cytoplasm and nucleus, as well as HO-1 and NQO1, following overexpression of LMTK2 in BV cells (Fig. 3B), implying that the activation of Nrf2 is dependent on LMTK2.

Discussion

The present study showed that exogenous LMTK2 significantly upregulated the expression of Nrf2 in the nucleus and the levels of HO-1 and NQO1 proteins in BV2 cells stimulated with LPS; which implied that LMTK2 promoted the transcription of Nrf2-mediated downstream genes. A recent study demonstrated that LMTK2 regulates GSK-3β/Nrf2/ARE signaling to ameliorate neuronal injury induced by oxygen-glucose deprivation/reoxygenation (8).

There is a crosstalk between the Nrf and NF-κB pathways (24). Lack of Nrf2 is associated with enhanced production of cytokines (25), which could lead to the neurodegenerative changes in Nrf2 knockdown animals (26,27). Previously, Nrf2 was reported to show anti-inflammatory abilities through the downregulation of COX-2, TNFα and iNOS in LPS-induced peritoneal macrophages (28). HO-1, as a Nrf2-mediated downstream protein, has been demonstrated to inhibit Nrf2-modulated NF-κB (29). Collectively, the upregulation of HO-1 expression could suppress NF-κB activation and cause the decrease in pro-inflammatory factors in the present study. LPS treatment frequently induces the activation of NF-κB, along with the enhancement of iNOS, COX-2, PGE2 and pro-inflammatory factors in microglia cells (30,31). PGE2 produced by microglial cells are the main source for neuroinflammation, showing marked increase upon LPS stimulation in microglia (32,33).

The aforementioned studies imply that the Nrf2 pathway could negatively regulate the NF-κB pathway. Taken together, LMTK2 overexpression reduced the levels of iNOS, COX-2 and pro-inflammatory factors, TNF-α, IL-1β and IL-6, but increased IL-10 level; possibly due to the dependence of LPS-induced microglia on Nrf2 pathway. However, no significant changes were observed in IL-10 levels following LPS stimulation, which was consistent with a previous report (34). IL-10 is an important inflammatory suppressor in vivo and can inhibit the release of pro-inflammatory cytokines in microglia cells in the central nervous system (35). Besides, LPS could induce the increase of NO, the production of iNOS, in microglia (36). In addition, NF-κB is considered upstream of NO and could initiate the synthesis of NO (37). A study has also shown that in lipoteichoic acid-induced microglia, matrix metalloprotease (MMP)-8 inhibitor regulates NF-κB and Nrf2 signals (38). Thus, LPS-mediated increase in pro-inflammatory factors were markedly reduced by inducement of exogenous LMTK2 expression, which implied the involvement of LMTK2 in regulating MMP-8 levels; however, this requires further study. In conclusion, the present study implies that LMTK2 regulates inflammation potentially by activating Nrf2 pathway.

Supplementary Material

Detection of LMTK2 mRNA through reverse transcription quantitative PCR in BV2 cells following transfection with LMTK2 overexpressing plasmids or empty plasmids. Data were shown as mean ± SD. ***P<0.001. LMTK2, Lemurs tyrosine kinase 2; NC, negative control.

Acknowledgements

Not applicable.

Funding

No funding was received.

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

QYR, and QX made substantial contributions to the conception and design of the study, acquired, analyzed and interpreted the data, and drafted and revised the manuscript for important intellectual content; QY, SGC, XZW, XYD, XI, WLD, QF and XGZ performed the experiments and interpreted the data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Tomomura M, Morita N, Yoshikawa F, Konishi A, Akiyama H, Furuichi T and Kamiguchi H: Structural and functional analysis of the apoptosis-associated tyrosine kinase (AATYK) family. Neuroscience. 148:510–521. 2007.PubMed/NCBI View Article : Google Scholar

2 

Wendler F: The LMTK-family of kinases: Emerging important players in cell physiology and disease pathogenesis. Biochim Biophys Acta Mol Basis Dis S0925-4439(18)30515-5, 2018.

3 

Bencze J, Mórotz GM, Seo W, Bencs V, Kálmán J, Miller CCJ and Hortobágyi T: Biological function of Lemur tyrosine kinase 2 (LMTK2): Implications in neurodegeneration. Mol Brain. 11(20)2018.PubMed/NCBI View Article : Google Scholar

4 

Wang H and Brautigan DL: A novel transmembrane Ser/Thr kinase complexes with protein phosphatase-1 and inhibitor-2. J Biol Chem. 277:49605–49612. 2002.PubMed/NCBI View Article : Google Scholar

5 

Luz S, Cihil KM, Brautigan DL, Amaral MD, Farinha CM and Swiatecka-Urban A: LMTK2-mediated phosphorylation regulates CFTR endocytosis in human airway epithelial cells. J Biol Chem. 289:15080–15093. 2014.PubMed/NCBI View Article : Google Scholar

6 

Kawa S, Ito C, Toyama Y, Maekawa M, Tezuka T, Nakamura T, Nakazawa T, Yokoyama K, Yoshida N, Toshimori K, et al: Azoospermia in mice with targeted disruption of the Brek/Lmtk2 (brain-enriched kinase/lemur tyrosine kinase 2) gene. Proc Natl Acad Sci USA. 103:19344–19349. 2006.PubMed/NCBI View Article : Google Scholar

7 

Cruz DF, Farinha CM and Swiatecka-Urban A: Unraveling the Function of Lemur Tyrosine Kinase 2 Network. Front Pharmacol. 10(24)2019.PubMed/NCBI View Article : Google Scholar

8 

Bao H and Gao M: Overexpression of lemur tyrosine kinase-2 protects neurons from oxygen-glucose deprivation/reoxygenation-induced injury through reinforcement of Nrf2 signaling by modulating GSK-3β phosphorylation. Biochem Biophys Res Commun. 521:964–970. 2020.PubMed/NCBI View Article : Google Scholar

9 

Manser C, Vagnoni A, Guillot F, Davies J and Miller CCJ: Cdk5/p35 phosphorylates lemur tyrosine kinase-2 to regulate protein phosphatase-1C phosphorylation and activity. J Neurochem. 121:343–348. 2012.PubMed/NCBI View Article : Google Scholar

10 

Tsai LH, Delalle I, Caviness VS Jr, Chae T and Harlow E: p35 is a neural-specific regulatory subunit of cyclin-dependent kinase 5. Nature. 371:419–423. 1994.PubMed/NCBI View Article : Google Scholar

11 

Guidato S, Tsai LH, Woodgett J and Miller CC: Differential cellular phosphorylation of neurofilament heavy side-arms by glycogen synthase kinase-3 and cyclin-dependent kinase-5. J Neurochem. 66:1698–1706. 1996.PubMed/NCBI View Article : Google Scholar

12 

Li BS, Zhang L, Gu J, Amin ND and Pant HC: Integrin alpha(1) beta(1)-mediated activation of cyclin-dependent kinase 5 activity is involved in neurite outgrowth and human neurofilament protein H Lys-Ser-Pro tail domain phosphorylation. J Neurosci. 20:6055–6062. 2000.PubMed/NCBI View Article : Google Scholar

13 

Zhang R, Li X, Wei L, Qin Y and Fang J: Lemur tyrosine kinase 2 acts as a positive regulator of NF-κB activation and colon cancer cell proliferation. Cancer Lett. 454:70–77. 2019.PubMed/NCBI View Article : Google Scholar

14 

Goldmann T and Prinz M: Role of microglia in CNS autoimmunity. Clin Dev Immunol. 2013(208093)2013.PubMed/NCBI View Article : Google Scholar

15 

Hickman S, Izzy S, Sen P, Morsett L and El Khoury J: Microglia in neurodegeneration. Nat Neurosci. 21:1359–1369. 2018.PubMed/NCBI View Article : Google Scholar

16 

Lehnardt S: Innate immunity and neuroinflammation in the CNS: The role of microglia in Toll like receptor mediated neuronal injury. Glia. 58:253–623. 2010.PubMed/NCBI View Article : Google Scholar

17 

Tsuda M, Tozaki Saitoh H and Inoue K: Purinergic system, microglia and neuropathic pain. Curr Opin Pharmacol. 74–79. 2012.PubMed/NCBI View Article : Google Scholar

18 

Mao SS, Hua R, Zhao XP, Qin X, Sun ZQ, Zhang Y, Wu YQ, Jia MX, Cao JL and Zhang YM: Exogenous administration of PACAP alleviates traumatic brain injury in rats through a mechanism involving the TLR4/MyD88/NF-κB pathway. J Neurotrauma. 29:1941–1959. 2012.PubMed/NCBI View Article : Google Scholar

19 

Aravalli RN, Peterson PK and Lokensgard JR: Toll-like receptors in defense and damage of the central nervous system. J Neuroimmune Pharmacol. 2:297–312. 2007.PubMed/NCBI View Article : Google Scholar

20 

Hirsch EC and Hunot S: Neuroinflammation in Parkinson's disease: A target for neuroprotection? Lancet Neurol. 8:382–397. 2009.PubMed/NCBI View Article : Google Scholar

21 

Frank-Cannon TC, Alto LT, McAlpine FE and Tansey MG: Does neuroinflammation fan the flame in neurodegenerative diseases? Mol Neurodegener. 4(47)2009.PubMed/NCBI View Article : Google Scholar

22 

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.PubMed/NCBI View Article : Google Scholar

23 

Gupta M and Kaur G: Aqueous extract from the Withania somnifera leaves as a potential anti-neuroinflammatory agent: A mechanistic study. J Neuroinflammation. 13(193)2016.PubMed/NCBI View Article : Google Scholar

24 

Wardyn JD, Ponsford AH and Sanderson CM: Dissecting molecular cross-talk between Nrf2 and NF-κB response pathways. Biochem Soc Trans. 43:621–626. 2015.PubMed/NCBI View Article : Google Scholar

25 

Pan H, Wang H, Wang X, Zhu L and Mao L: The absence of Nrf2 enhances NF-κB-dependent inflammation following scratch injury in mouse primary cultured astrocytes. Mediators Inflamm. 2012(217580)2012.PubMed/NCBI View Article : Google Scholar

26 

Neymotin A, Calingasan NY, Wille E, Naseri N, Petri S, Damiano M, Liby KT, Risingsong R, Sporn M, Beal MF, et al: Neuroprotective effect of Nrf2/ARE activators, CDDO ethylamide and CDDO trifluoroethylamide, in a mouse model of amyotrophic lateral sclerosis. Free Radic Biol Med. 51:88–96. 2011.PubMed/NCBI View Article : Google Scholar

27 

Frakes AE, Ferraiuolo L, Haidet-Phillips AM, Schmelzer L, Braun L, Miranda CJ, Ladner KJ, Bevan AK, Foust KD, Godbout JP, et al: Microglia induce motor neuron death via the classical NF-κB pathway in amyotrophic lateral sclerosis. Neuron. 81:1009–1023. 2014.PubMed/NCBI View Article : Google Scholar

28 

Lin W, Wu RT, Wu T, Khor TO, Wang H and Kong AN: Sulforaphane suppressed LPS-induced inflammation in mouse peritoneal macrophages through Nrf2 dependent pathway. Biochem Pharmacol. 76:967–973. 2008.PubMed/NCBI View Article : Google Scholar

29 

Soares MP, Seldon MP, Gregoire IP, Vassilevskaia T, Berberat PO, Yu J, Tsui TY and Bach FH: Heme oxygenase-1 modulates the expression of adhesion molecules associated with endothelial cell activation. J Immunol. 172:3553–3563. 2004.PubMed/NCBI View Article : Google Scholar

30 

Xie Q, Wu GZ, Yang N, Shen YH, Tang J and Zhang WD: Delavatine A, an unusual isoquinoline alkaloid exerts anti-inflammation on LPS-induced proinflammatory cytokines production by suppressing NF-κB activation in BV-2 microglia. Biochem Biophys Res Commun. 502:202–208. 2018.PubMed/NCBI View Article : Google Scholar

31 

Zhao J, Bi W, Xiao S, Lan X, Cheng X, Zhang J, Lu D, Wei W, Wang Y, Li H, et al: Neuroinflammation induced by lipopolysaccharide causes cognitive impairment in mice. Sci Rep. 9(5790)2019.PubMed/NCBI View Article : Google Scholar

32 

Jung YS, Park JH, Kim H, Kim SY, Hwang JY, Hong KW, Bae SS, Choi BT, Lee SW and Shin HK: Probucol inhibits LPS-induced microglia activation and ameliorates brain ischemic injury in normal and hyperlipidemic mice. Acta Pharmacol Sin. 37:1031–1044. 2016.PubMed/NCBI View Article : Google Scholar

33 

Saliba SW, Jauch H, Gargouri B, Keil A, Hurrle T, Volz N, Mohr F, van der Stelt M, Bräse S and Fiebich BL: Anti-neuroinflammatory effects of GPR55 antagonists in LPS-activated primary microglial cells. J Neuroinflammation. 15(322)2018.PubMed/NCBI View Article : Google Scholar

34 

Bao Y, Zhu Y, He G, Ni H, Liu C, Ma L, Zhang L and Shi D: Dexmedetomidine attenuates neuroinflammation in LPS-stimulated BV2 microglia cells through upregulation of miR-340. Drug Des Devel Ther. 13:3465–3475. 2019.PubMed/NCBI View Article : Google Scholar

35 

Cianciulli A, Dragone T, Calvello R, Porro C, Trotta T, Lofrumento DD and Panaro MA: IL-10 plays a pivotal role in anti-inflammatory effects of resveratrol in activated microglia cells. Int Immunopharmacol. 24:369–376. 2015.PubMed/NCBI View Article : Google Scholar

36 

Posadas I, Terencio MC, Guillén I, Ferrándiz ML, Coloma J, Payá M and Alcaraz MJ: Co-regulation between cyclo-oxygenase-2 and inducible nitric oxide synthase expression in the time-course of murine inflammation. Naunyn Schmiedebergs Arch Pharmacol. 361:98–106. 2000.PubMed/NCBI View Article : Google Scholar

37 

El Moussawi L, Chakkour M and Kreydiyyeh S: The epinephrine-induced PGE2 reduces Na+/K+ ATPase activity in Caco-2 cells via PKC, NF-κB and NO. PLoS One. 14(e0220987)2019.PubMed/NCBI View Article : Google Scholar

38 

Lee EJ, Park JS, Lee YY, Kim DY, Kang JL and Kim HS: Anti-inflammatory and anti-oxidant mechanisms of an MMP-8 inhibitor in lipoteichoic acid-stimulated rat primary astrocytes: Involvement of NF-κB, Nrf2, and PPAR-γ signaling pathways. J Neuroinflammation. 15(326)2018.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

March-2021
Volume 21 Issue 3

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Rui Q, Cao S, Wang X, Duan X, Iao X, Dong W, Fang Q, Zhang X and Xue Q: LMTK2 regulates inflammation in lipopolysaccharide‑stimulated BV2 cells. Exp Ther Med 21: 219, 2021
APA
Rui, Q., Cao, S., Wang, X., Duan, X., Iao, X., Dong, W. ... Xue, Q. (2021). LMTK2 regulates inflammation in lipopolysaccharide‑stimulated BV2 cells. Experimental and Therapeutic Medicine, 21, 219. https://doi.org/10.3892/etm.2021.9621
MLA
Rui, Q., Cao, S., Wang, X., Duan, X., Iao, X., Dong, W., Fang, Q., Zhang, X., Xue, Q."LMTK2 regulates inflammation in lipopolysaccharide‑stimulated BV2 cells". Experimental and Therapeutic Medicine 21.3 (2021): 219.
Chicago
Rui, Q., Cao, S., Wang, X., Duan, X., Iao, X., Dong, W., Fang, Q., Zhang, X., Xue, Q."LMTK2 regulates inflammation in lipopolysaccharide‑stimulated BV2 cells". Experimental and Therapeutic Medicine 21, no. 3 (2021): 219. https://doi.org/10.3892/etm.2021.9621