Anti-inflammatory Effect of Glucagon Like Peptide-1 Receptor Agonist, Exendin-4, through Modulation of IB1/JIP1 Expression and JNK Signaling in Stroke

Soojin Kim, Jaewon Jeong, Hye-Seon Jung, Bokyung Kim, Ye-Eun Kim, Da-Sol Lim, So-Dam Kim, Yun Seon Song, Soojin Kim, Jaewon Jeong, Hye-Seon Jung, Bokyung Kim, Ye-Eun Kim, Da-Sol Lim, So-Dam Kim, Yun Seon Song

Abstract

Glucagon like peptide-1 (GLP-1) stimulates glucose-dependent insulin secretion. Dipeptidyl peptidase-4 (DPP-4) inhibitors, which block inactivation of GLP-1, are currently in clinical use for type 2 diabetes mellitus. Recently, GLP-1 has also been reported to have neuroprotective effects in cases of cerebral ischemia. We therefore investigated the neuroprotective effects of GLP-1 receptor (GLP-1R) agonist, exendin-4 (ex-4), after cerebral ischemia-reperfusion injury. Transient middle cerebral artery occlusion (tMCAO) was induced in rats by intracerebroventricular (i.c.v.) administration of ex-4 or ex9-39. Oxygen-glucose deprivation was also induced in primary neurons, bEnd.3 cells, and BV-2. Ischemia-reperfusion injury reduced expression of GLP-1R. Additionally, higher oxidative stress in SOD2 KO mice decreased expression of GLP-1R. Downregulation of GLP-1R by ischemic injury was 70% restored by GLP-1R agonist, ex-4, which resulted in significant reduction of infarct volume. Levels of intracellular cyclic AMP, a second messenger of GLP-1R, were also increased by 2.7-fold as a result of high GLP-1R expression. Moreover, our results showed that ex-4 attenuated pro-inflammatory cyclooxygenase-2 (COX-2) and prostaglandin E2 after MCAO. C-Jun NH2 terminal kinase (JNK) signaling, which stimulates activation of COX-2, was 36% inhibited by i.c.v. injection of ex-4 at 24 h. Islet-brain 1 (IB1), a scaffold regulator of JNK, was 1.7-fold increased by ex-4. GLP-1R activation by ex-4 resulted in reduction of COX-2 through increasing IB1 expression, resulting in anti-inflammatory neuroprotection during stroke. Our study suggests that the anti-inflammatory action of GLP-1 could be used as a new strategy for the treatment of neuroinflammation after stroke accompanied by hyperglycemia.

Keywords: Cerebral ischemia; Exendine-4; Glucagon like peptide-1 receptor; Islet-brain 1; Neuroinflammation; c-Jun NH2 terminal kinase.

Figures

Fig. 1. Reduction of GLP-1 receptor levels…
Fig. 1. Reduction of GLP-1 receptor levels after tMCAO, and its relevance to oxidative stress in mice. (A) One-hour tMCAO reduced GLP-1R mRNA expression at 6 h and 24 h reperfusion (Rep) in mice (n =4, *p<0.05, compared to sham-operated group). (B) Four-hour OGD was performed in primary cultured neurons, and showed decreased GLP-1R mRNA levels compared to Nor. (C) SOD 2 KO mice, which have a high superoxide level, had significantly lower expression of GLP-1R than WT mice under normal or tMCAO (n =3, **p<0.01, compared to sham-operated WT group, ††p<0.01, comparison between sham-operated WT and KO group, #p<0.05, comparison between WT and KO mice at 24 h). (D) In SOD1 Tg mice, GLP-1R mRNA levels were higher than WT mice under sham or tMCAO.
Fig. 2. Neuroprotective effects of ex-4 through…
Fig. 2. Neuroprotective effects of ex-4 through the G protein-coupled GLP-1 receptor. (A) Infarct volume was assessed by TTC staining after tMCAO in rats. Treatment with ex-4, GLP-1R agonist, resulted in a significant reduction of brain tissue damage after ischemic injury. Ex9-39, a GLP-1R antagonist, reduced brain infarction volume; however, it was less than that of ex-4 (n =3, **p<0.01, ***p<0.001, compared to sham-operated group, ##p<0.01, ###p<0.001, compared to each chemical-treated group). (B) Reduction of GLP-1R protein and mRNA expression were noted at 48 h after 1 h MCAO. Ex-4 efficiently counteracted decreases in GLP-1R in the ischemic rat brain. Treatment of ex9-39 reduced the level of GLP-1R as much as the vehicle group. Ischemic reperfusion injury significantly increased GLP-1 levels (n =5, *p<0.05, ***p<0.001, compared to sham-operated group, #p<0.05, ##p<0.01, compared to each chemical-treated group). (C) In primary neurons, mouse brain microglia cells, and endothelial cells, ex-4 increased the reduced GLP-1R protein levels after OGD compared to the vehicle group (n =5, **p<0.01, compared to Nor, ##p<0.01, compared to each chemical-treated group). (D) Cyclic AMP levels were evaluated to investigate GLP-1R signaling. Cyclic AMP levels were decreased after 1 h tMCAO, but showed a significant increase in the ex-4 treated animal group (n =4, ##p<0.01, compared to vehicle group).
Fig. 3. Anti-inflammatory effects of ex-4 on…
Fig. 3. Anti-inflammatory effects of ex-4 on COX-2 and PGE2. (A) The level of COX-2 was significantly increased at 48 h after tMCAO. Treatment with ex-4 restored COX-2 to the basal level after tMCAO in the rat brain. Ex9-39 treatment increased COX-2 levels as much as vehicle group. (B) Ex-4 also reduced the COX-2 mRNA levels in the rat brain after tMCAO (n =5, **p<0.01, compared to sham-operated group, ##p<0.01, compared to chemical-treated group). (C) Results were consistent with the COX-2 promoter assay. Ex-4 attenuated COX-2 luciferase activity after OGD in bEnd.3 cells. Ex9-39 increased COX-2 activity as much as vehicle group. (D) The level of PGE2, which is product of COX-2 activity, was increased by 1 h tMCAO, but this level was attenuated by ex-4 (n =5, **p<0.01, compared to the sham-operated group).
Fig. 4. Attenuation of the SAPK/JNK pathway…
Fig. 4. Attenuation of the SAPK/JNK pathway by ex-4 through the GLP-1R. (A) Phosphorylation of SAPK/JNK was elevated following ischemic reperfusion injury in rats. Treatment with ex-4 prior to 1 h transient middle cerebral artery occlusion inhibited up-regulation of phospho-JNK and COX-2, while treatment with ex-9-39 increased phospho-JNK levels. Ex-4 did not significantly change other MAPK signaling. (B) Quantitative analyses showed significant augmentation in JNK-mediated COX-2 expression in rat (n =4, **p<0.01, ***p<0.001, compared to sham-operated group, ##p<0.01, compared to chemical-treated group at the same time point).
Fig. 5. Analysis of the interaction between…
Fig. 5. Analysis of the interaction between IB1/JIP1 and GLP-1R after tMCAO in rat. (A) IB1/JIP1 protein levels were down-regulated by 1 h tMCAO, and following reperfusion. Ex-4 treatment dramatically increased IB1/JIP1 levels in the rat brain, while ex9-39 did not. Up-regulation of IB1/JIP1 by ex-4 treatment led to reduced expression of phospho-JNK (n =4, #p<0.05, compared to chemical treated group at the same time point). (B) Endogenous IB1/JIP1 was captured by immunoprecipitation with an anti-IB1/JIP1 antibody in the total fraction of infarcted brain. Results were detected by immunoblotting using antibodies to GLP-1R, COX-2, and phospho-SAPK/JNK. Decreased GLP-1R was detected in the vehicle group, while the ex-4 treated group showed significant interactions between GLP-1R and IB1/JIP1 compared to the sham group. Input lysates showed no differences (n =4, *p<0.05, compared to sham-operated group, #p<0.01, compared to chemical-treated group).

References

    1. Béjot Y, Giroud M. Stroke in diabetic patients. Diabetes Metab. 2010;36(Suppl 3):S84–S87.
    1. Kruyt ND, Biessels GJ, Devries JH, Roos YB. Hyperglycemia in acute ischemic stroke: pathophysiology and clinical management. Nat Rev Neurol. 2010;6:145–155.
    1. Cull CA, Neil HA, Holman RR. Changing aspirin use in patients with Type 2 diabetes in the UKPDS. Diabet Med. 2004;21:1368–1371.
    1. Riddle MC. Effects of intensive glucose lowering in the management of patients with type 2 diabetes mellitus in the Action to Control Cardiovascular Risk in Diabetes (ACCORD) trial. Circulation. 2010;122:844–846.
    1. Wilcox R, Bousser MG, Betteridge DJ, Schernthaner G, Pirags V, Kupfer S, Dormandy J PROactive Investigators. Effects of pioglitazone in patients with type 2 diabetes with or without previous stroke: results from PROactive (PROspective pioglitAzone Clinical Trial In macroVascular Events 04) Stroke. 2007;38:865–873.
    1. Yusta B, Baggio LL, Estall JL, Koehler JA, Holland DP, Li H, Pipeleers D, Ling Z, Drucker DJ. GLP-1 receptor activation improves beta cell function and survival following induction of endoplasmic reticulum stress. Cell Metab. 2006;4:391–406.
    1. Holst JJ, Burcelin R, Nathanson E. Neuroprotective properties of GLP-1: theoretical and practical applications. Curr Med Res Opin. 2011;27:547–558.
    1. During MJ, Cao L, Zuzga DS, Francis JS, Fitzsimons HL, Jiao X, Bland RJ, Klugmann M, Banks WA, Drucker DJ, Haile CN. Glucagon-like peptide-1 receptor is involved in learning and neuroprotection. Nat Med. 2003;9:1173–1179.
    1. Li Y, Perry T, Kindy MS, Harvey BK, Tweedie D, Holloway HW, Powers K, Shen H, Egan JM, Sambamurti K, Brossi A, Lahiri DK, Mattson MP, Hoffer BJ, Wang Y, Greig NH. GLP-1 receptor stimulation preserves primary cortical and dopaminergic neurons in cellular and rodent models of stroke and Parkinsonism. Proc Natl Acad Sci U S A. 2009;106:1285–1290.
    1. Teramoto S, Miyamoto N, Yatomi K, Tanaka Y, Oishi H, Arai H, Hattori N, Urabe T. Exendin-4, a glucagon-like peptide-1 receptor agonist, provides neuroprotection in mice transient focal cerebral ischemia. J Cereb Blood Flow Metab. 2011;31:1696–1705.
    1. Nogawa S, Zhang F, Ross ME, Iadecola C. Cyclo-oxygenase-2 gene expression in neurons contributes to ischemic brain damage. J Neurosci. 1997;17:2746–2755.
    1. Schroer K, Zhu Y, Saunders MA, Deng WG, Xu XM, Meyer-Kirchrath J, Wu KK. Obligatory role of cyclic adenosine monophosphate response element in cyclooxygenase-2 promoter induction and feedback regulation by inflammatory mediators. Circulation. 2002;105:2760–2765.
    1. Chen TH, Wo HT, Wu CC, Wang JL, Wang CC, Hsieh IC, Kuo CY, Liu CT. Exendin-4 attenuates lipopolysaccharides induced inflammatory response but does not protects H9c2 cells from apoptosis. Immunopharmacol Immunotoxicol. 2012;34:484–490.
    1. Whitmarsh AJ, Cavanagh J, Tournier C, Yasuda J, Davis RJ. A mammalian scaffold complex that selectively mediates MAP kinase activation. Science. 1998;281:1671–1674.
    1. Dickens M, Rogers JS, Cavanagh J, Raitano A, Xia Z, Halpern JR, Greenberg ME, Sawyers CL, Davis RJ. A cytoplasmic inhibitor of the JNK signal transduction pathway. Science. 1997;277:693–696.
    1. Nihalani D, Wong HN, Holzman LB. Recruitment of JNK to JIP1 and JNK-dependent JIP1 phosphorylation regulates JNK module dynamics and activation. J Biol Chem. 2003;278:28694–28702.
    1. Bonny C, Oberson A, Steinmann M, Schorderet DF, Nicod P, Waeber G. IB1 reduces cytokine-induced apoptosis of insulin-secreting cells. J Biol Chem. 2000;275:16466–16472.
    1. Centeno C, Repici M, Chatton JY, Riederer BM, Bonny C, Nicod P, Price M, Clarke PG, Papa S, Franzoso G, Borsello T. Role of the JNK pathway in NMDA-mediated excitotoxicity of cortical neurons. Cell Death Differ. 2007;14:240–253.
    1. Ferdaoussi M, Abdelli S, Yang JY, Cornu M, Niederhauser G, Favre D, Widmann C, Regazzi R, Thorens B, Waeber G, Abderrahmani A. Exendin-4 protects beta-cells from interleukin-1 beta-induced apoptosis by interfering with the c-Jun NH2-terminal kinase pathway. Diabetes. 2008;57:1205–1215.
    1. Magara F, Haefliger JA, Thompson N, Riederer B, Welker E, Nicod P, Waeber G. Increased vulnerability to kainic acid-induced epileptic seizures in mice underexpressing the scaffold protein Islet-Brain 1/JIP-1. Eur J Neurosci. 2003;17:2602–2610.
    1. Dong Z, Zhou L, Del Villar K, Ghanevati M, Tashjian V, Miller CA. JIP1 regulates neuronal apoptosis in response to stress. Brain Res Mol Brain Res. 2005;134:282–293.
    1. Epstein CJ, Avraham KB, Lovett M, Smith S, Elroy-Stein O, Rotman G, Bry C, Groner Y. Transgenic mice with increased Cu/Zn-superoxide dismutase activity: animal model of dosage effects in Down syndrome. Proc Natl Acad Sci U S A. 1987;84:8044–8048.
    1. Li Y, Copin JC, Reola LF, Calagui B, Gobbel GT, Chen SF, Sato S, Epstein CJ, Chan PH. Reduced mitochondrial manganese-superoxide dismutase activity exacerbates glutamate toxicity in cultured mouse cortical neurons. Brain Res. 1998;814:164–170.
    1. Kinouchi H, Epstein CJ, Mizui T, Carlson E, Chen SF, Chan PH. Attenuation of focal cerebral ischemic injury in transgenic mice overexpressing CuZn superoxide dismutase. Proc Natl Acad Sci U S A. 1991;88:11158–11162.
    1. Kim GW, Kondo T, Noshita N, Chan PH. Manganese superoxide dismutase deficiency exacerbates cerebral infarction after focal cerebral ischemia/reperfusion in mice: implications for the production and role of superoxide radicals. Stroke. 2002;33:809–815.
    1. Allen CL, Bayraktutan U. Oxidative stress and its role in the pathogenesis of ischaemic stroke. Int J Stroke. 2009;4:461–470.
    1. Perry T, Greig NH. The glucagon-like peptides: a double-edged therapeutic sword? Trends Pharmacol Sci. 2003;24:377–383.
    1. Briyal S, Gulati K, Gulati A. Repeated administration of exendin-4 reduces focal cerebral ischemia-induced infarction in rats. Brain Res. 2012;1427:23–34.
    1. Drucker DJ. The biology of incretin hormones. Cell Metab. 2006;3:153–165.
    1. Candelario-Jalil E, González-Falcón A, García-Cabrera M, Alvarez D, Al-Dalain S, Martínez G, León OS, Springer JE. Assessment of the relative contribution of COX-1 and COX-2 isoforms to ischemia-induced oxidative damage and neurodegeneration following transient global cerebral ischemia. J Neurochem. 2003;86:545–555.
    1. Williams TJ, Peck MJ. Role of prostaglandin-mediated vasodilatation in inflammation. Nature. 1977;270:530–532.
    1. Bonny C, Oberson A, Negri S, Sauser C, Schorderet DF. Cell-permeable peptide inhibitors of JNK: novel blockers of beta-cell death. Diabetes. 2001;50:77–82.
    1. Perry T, Lahiri DK, Chen D, Zhou J, Shaw KT, Egan JM, Greig NH. A novel neurotrophic property of glucagon-like peptide 1: a promoter of nerve growth factor-mediated differentiation in PC12 cells. J Pharmacol Exp Ther. 2002;300:958–966.
    1. Luciani P, Deledda C, Benvenuti S, Cellai I, Squecco R, Monici M, Cialdai F, Luciani G, Danza G, Di Stefano C, Francini F, Peri A. Differentiating effects of the glucagon-like peptide-1 analogue exendin-4 in a human neuronal cell model. Cell Mol Life Sci. 2010;67:3711–3723.
    1. Tang-Christensen M, Larsen PJ, Göke R, Fink-Jensen A, Jessop DS, Møller M, Sheikh SP. Central administration of GLP-1-(7-36) amide inhibits food and water intake in rats. Am J Physiol. 1996;271:R848–R856.
    1. Göke R, Larsen PJ, Mikkelsen JD, Sheikh SP. Distribution of GLP-1 binding sites in the rat brain: evidence that exendin-4 is a ligand of brain GLP-1 binding sites. Eur J Neurosci. 1995;7:2294–2300.
    1. Dunphy JL, Taylor RG, Fuller PJ. Tissue distribution of rat glucagon receptor and GLP-1 receptor gene expression. Mol Cell Endocrinol. 1998;141:179–186.
    1. Timmers L, Henriques JP, de Kleijn DP, Devries JH, Kemperman H, Steendijk P, Verlaan CW, Kerver M, Piek JJ, Doevendans PA, Pasterkamp G, Hoefer IE. Exenatide reduces infarct size and improves cardiac function in a porcine model of ischemia and reperfusion injury. J Am Coll Cardiol. 2009;53:501–510.
    1. Hui H, Nourparvar A, Zhao X, Perfetti R. Glucagon-like peptide-1 inhibits apoptosis of insulin-secreting cells via a cyclic 5’-adenosine monophosphate-dependent protein kinase A- and a phosphatidylinositol 3-kinase-dependent pathway. Endocrinology. 2003;144:1444–1455.
    1. Walton M, Sirimanne E, Williams C, Gluckman P, Dragunow M. The role of the cyclic AMP-responsive element binding protein (CREB) in hypoxic-ischemic brain damage and repair. Brain Res Mol Brain Res. 1996;43:21–29.
    1. Masferrer JL, Zweifel BS, Manning PT, Hauser SD, Leahy KM, Smith WG, Isakson PC, Seibert K. Selective inhibition of inducible cyclooxygenase 2 in vivo is antiinflammatory and nonulcerogenic. Proc Natl Acad Sci U S A. 1994;91:3228–3232.
    1. Chun KS, Surh YJ. Signal transduction pathways regulating cyclooxygenase-2 expression: potential molecular targets for chemoprevention. Biochem Pharmacol. 2004;68:1089–1100.
    1. Abderrahmani A, Niederhauser G, Favre D, Abdelli S, Ferdaoussi M, Yang JY, Regazzi R, Widmann C, Waeber G. Human high-density lipoprotein particles prevent activation of the JNK pathway induced by human oxidised low-density lipoprotein particles in pancreatic beta cells. Diabetologia. 2007;50:1304–1314.
    1. Pellet JB, Haefliger JA, Staple JK, Widmann C, Welker E, Hirling H, Bonny C, Nicod P, Catsicas S, Waeber G, Riederer BM. Spatial, temporal and subcellular localization of islet-brain 1 (IB1), a homologue of JIP-1, in mouse brain. Eur J Neurosci. 2000;12:621–632.
    1. Helbecque N, Abderrahamani A, Meylan L, Riederer B, Mooser V, Miklossy J, Delplanque J, Boutin P, Nicod P, Haefliger JA, Cottel D, Amouyel P, Froguel P, Waeber G. Islet-brain1/C-Jun N-terminal kinase interacting protein-1 (IB1/JIP-1) promoter variant is associated with Alzheimer's disease. Mol Psychiatry. 2003;8:413–422. 363.
    1. Taru H, Iijima K, Hase M, Kirino Y, Yagi Y, Suzuki T. Interaction of Alzheimer’s beta -amyloid precursor family proteins with scaffold proteins of the JNK signaling cascade. J Biol Chem. 2002;277:20070–20078.
    1. Matsuda S, Yasukawa T, Homma Y, Ito Y, Niikura T, Hiraki T, Hirai S, Ohno S, Kita Y, Kawasumi M, Kouyama K, Yamamoto T, Kyriakis JM, Nishimoto I. c-Jun N-terminal kinase (JNK)-interacting protein-1b/islet-brain-1 scaffolds Alzheimer’s amyloid precursor protein with JNK. J Neurosci. 2001;21:6597–6607.
    1. Scheinfeld MH, Roncarati R, Vito P, Lopez PA, Abdallah M, D'Adamio L. Jun NH2-terminal kinase (JNK) interacting protein 1 (JIP1) binds the cytoplasmic domain of the Alzheimer’s beta-amyloid precursor protein (APP) J Biol Chem. 2002;277:3767–3775.
    1. Hayashi T, Sakai K, Sasaki C, Zhang WR, Warita H, Abe K. c-Jun N-terminal kinase (JNK) and JNK interacting protein response in rat brain after transient middle cerebral artery occlusion. Neurosci Lett. 2000;284:195–199.

Source: PubMed

3
Prenumerera