Wnt canonical pathway activator TWS119 drives microglial anti-inflammatory activation and facilitates neurological recovery following experimental stroke

Degang Song, Xiangjian Zhang, Junmin Chen, Xiaoxia Liu, Jing Xue, Lan Zhang, Xifa Lan, Degang Song, Xiangjian Zhang, Junmin Chen, Xiaoxia Liu, Jing Xue, Lan Zhang, Xifa Lan

Abstract

Background: Ischemic stroke is a leading cause of disability worldwide and characteristically accompanied by downregulation of the Wnt/β-catenin signaling. Activation of Wnt/β-catenin signaling emerges to attenuate neuroinflammation after ischemic stroke; however, its effect on modulating microglial polarization is largely unknown. Here, we explored whether Wnt/β-catenin pathway activator TWS119 facilitated long-term neurological recovery via modulating microglia polarization after experimental stroke.

Methods: Ischemic stroke mice model was induced by permanent distal middle cerebral artery occlusion plus 1 h hypoxia. TWS119 was administrated from day 1 to 14 after stroke. Neurological deficits were monitored up to 21 days after stroke. Angiogenesis, neural plasticity, microglial polarization, and microglia-associated inflammatory cytokines were detected in the peri-infarct cortex at days 14 and 21 after stroke. Primary microglia and mouse brain microvascular endothelial cell lines were employed to explore the underlying mechanism in vitro.

Results: TWS119 mitigated neurological deficits at days 14 and 21 after experimental stroke, paralleled by acceleration on angiogenesis and neural plasticity in the peri-infarct cortex. Mechanistically, cerebral ischemia induced production of microglia-associated proinflammatory cytokines and priming of activated microglia toward pro-inflammatory polarization, whereas TWS119 ameliorated microglia-mediated neuroinflammatory status following ischemic stroke and promoted angiogenesis by modulating microglia to anti-inflammatory phenotype. The beneficial efficacy of TWS119 in microglial polarization was largely reversed by selective Wnt/β-catenin pathway blockade in vitro, suggesting that TWS119-enabled pro-inflammatory to anti-inflammatory phenotype switch of microglia was possibly mediated by Wnt/β-catenin signaling.

Conclusions: Wnt/β-catenin pathway activator TWS119 ameliorated neuroinflammatory microenvironment following chronic cerebral ischemia via modulating microglia towards anti-inflammatory phenotype, and facilitates neurological recovery in an anti-inflammatory phenotype polarization-dependent manner. Activation of Wnt/β-catenin pathway following ischemic stroke might be a potential restorative strategy targeting microglia-mediated neuroinflammation.

Keywords: Cerebral ischemia; Microglial polarization; Neuroinflammation; Neurological recovery; TWS119; WNT/β-catenin.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Fig. 1
Fig. 1
Experimental outline and schematic diagram of brain section. a Experimental outline: TWS119 or BrdU were administrated intraperitoneally once daily from day 1 to 14 or from day 7 to 14 after stroke. Neurobehavioral tests were performed at days 1, 7, 14, and 21 after stroke. Angiogenesis, neural plasticity, microglial polarization and inflammatory cytokines were detected at the indicated time point. Histological assessment was assessed at day 21. The number of mice in each group for each test were shown in parentheses. b Schematic diagram of brain section. Green squares indicated the region of interest in the ipsilateral peri-infarct cortex, in which immunofluorescence images were collected. Yellow strip (0.5 mm wide) indicated peri-infarct region, in which brain samples for qRT-PCR and ELISA were harvested. stroke, focal cerebral ischemia; IF, immunofluorescence; qRT-PCR, quantitative real time polymerase chain reaction; ELISA, enzyme linked immunosorbent assay
Fig. 2
Fig. 2
TWS119 improved post-stroke neurological function. CatWalk test (a–c), Adhesive Removal test (d, e), Accelerated Rotarod test (f) and mNSS (g) were performed between vehicle group and TWS119 group at day 1, 7, 14 and 21 after stroke. a-e TWS119 treatment, as opposed to saline injection, accelerated LF functional recovery. f TWS119 mice displayed better performance in Accelerated Rotarod test compared with vehicle mice. g TWS119 mice had lower mNSS compared with vehicle mice. LF, left forelimb, mNSS, modified neurological severity scores. (n = 8 per group, * P < 0.05, ** P < 0.01, *** P < 0.001, N.S = no statistical difference)
Fig. 3
Fig. 3
TWS119 facilitated post-stroke angiogenesis. a Representative images of coronal sections labeled with CD31 (cerebral microvascular endothelial cells marker). b Area of CD31-positive microvessels were increased in ischemic mice at day 14 and 21 after stroke, and further upregulated by TWS119 treatment at day 21. c Cerebral ischemia enhanced the density of CD31-positive microvessels in peri-infarct cortex at day 14 and 21 after stroke, this change was more significant in TWS119 mice. d Representative images of coronal sections labeled with CD31 or/and BrdU (cell proliferation marker). e TWS119 increased the percentage of CD31+BrdU+ cells in total CD31+ cells at day 14 after stroke. (n = 6 per group, * P < 0.05, ** P < 0.01)
Fig. 4
Fig. 4
TWS119 stimulated post-stroke neural plasticity. a Representative image of coronal sections labeled with GAP43 (axon marker), and PSD95 (postsynaptic structure marker). b TWS119 mice showed a higher density of GAP43+ puncta and PSD95+ puncta at day 21 after stroke compared with vehicle mice. c Representative image of coronal sections labeled with SMI312 (neurofilament marker), and Synaptophysin (presynaptic structure marker). d TWS119 also upregulated the density of SMI312+ puncta or Synaptophysin+ puncta at day 21 after stroke. (n = 6 per group, * P < 0.05, ** P < 0.01)
Fig. 5
Fig. 5
TWS119 modulated microglia/Macrophages to anti-inflammatory phenotype after ischemic stroke. a Pro-inflammatory-type marker (CD-16, TNF-α, iNOS, IL-1β and IL-6) and anti-inflammatory-type marker (CD206, IL-10, TGF-β, Arg-1 and YM1/2) of microglia were analyzed by qRT-PCR 14 days after stroke. The mRNA levels of CD-16, TNF-α, iNOS, IL-1β, IL-6 and IL-10 were increased in vehicle mice compared with sham mice, TWS119 treatment decreased the level of CD-16, TNF-α and iNOS, and upregulated the level of CD206, IL-10, TGF-β, Arg-1 and YM1/2 compared with saline treatment. b Representative images of coronal sections labeled with Iba1 (microglia marker), and CD16/32. c TWS119 reduced the ratio of CD16/32+Iba1+ cells at 14 days after stroke. d Representative images of coronal sections labeled with Iba1, and CD206. e TWS119 increased the ratio of CD206+Iba1+ cells at 14 and 21 days after stroke. (n = 6 per group, * P < 0.05, ** P < 0.01)
Fig. 6
Fig. 6
TWS119 ameliorated local inflammatory microenvironment in peri-infarct cortex. a TNF-α (pro-inflammatory cytokine) and IL-10 (anti-inflammatory cytokine) in per-infarct region ware measured by ELISA. The production of TNF-α was obviously increased at day 14 and 21 after stroke. TWS119 significantly reduced the production of TNF-α at day 14, and increased the production of IL-10 at day 14 and 21. b The production of IL-10 had a positive correlation with the residence time of Accelerated Rotarod test, while the production of IL-10 had a negative correlation with the LF-removal time in Adhesive Removal test and the mNSS at day 21 after stroke. LF, left forelimb; mNSS, modified neurological severity scores. (n = 8 per group, * P < 0.05, ** P < 0.01)
Fig. 7
Fig. 7
TWS119 promoted pro-inflammatory to anti-inflammatory phenotype switch of microglia probably via Wnt/β-catenin pathway. Pro-inflammatory polarization was stimulated by LPS plus IFN-γ for 24 h. a CCK-8 assay was performed to determine the cytotoxicity of TWS119, TWS119 with the doses (≥ 100 μM) exerted cytotoxic effect on microglia cells. b Using ELISA, 10 μM was determined the effective dose. TWS119 with doses (10 μM, 20 μM) increased the secretion of IL-10 in microglia cells stimulated with LPS + IFN-γ, while TWS119 with doses (5 μM, 40 μM) had no similar effect. c mRNA expression of CD16 was enhanced in LPS + IFN-γ-stimulated microglia cells, this enhancement was corrected by TWS119 treatment, IWR-1 reversed the effect of TWS119 on CD16. TWS119 increased the level of CD206, which was reversed by IWR-1. d TWS119 reduced the percentage of CD16/32 positive cells and raised CD206 positive cells in Flow Cytometry, which was reversed by IWR-1. e TWS119 increased the level of β-catenin, which was reversed by IWR-1. IFN-γ, interferon-γ; IWR-1, a reversible inhibitor of Wnt/β-catenin signaling pathway; LPS, lipopolysaccharide. (n = 5 per group, 5 independent experiments from 5 different microglia preps. * P < 0.05, ** P < 0.01, *** P < 0.001, N.S means no statistical significance)
Fig. 8
Fig. 8
TWS119 promoted angiogenesis in vitro via an amelioration of inflammatory microenvironment by altering microglia polarization. a Tube formation of bEend3 cells in conventional medium (5 × magnification). OGD injury reduced vascular tubular structures, which was not corrected by TWS119 treatment. b Supernatants of each microglia culture were used as CM. Proinflammatory cytokines (TNF-α, Nitrite, IL-1β) and anti-inflammatory cytokines (IL-10, TGF-β) were detected in each CM. c Representative images of tube formation in OGDbEend.3 cells incubated with CM (5 × magnification). Interestingly, only CM co-treated with LPS plus IFN-γ and TWS119 significantly increased the formation of pro-angiogenic structures in OGDbEnd.3 cells. Other CM had no similar function. bEend3, mouse-derived brain microvascular cell line; IFN-γ, interferon-γ; CM, conditioned media; LPS, lipopolysaccharide; OGD, oxygen-glucose deprivation. (n = 5, *P < 0.05, ***P < 0.05, N.S means no statistical significance)

References

    1. Go AS, Mozaffarian D, Roger VL, Benjamin EJ, Berry JD, Borden WB, Bravata DM, Dai S, Ford ES, Fox CS, et al. Executive summary: heart disease and stroke statistics--2013 update: a report from the American Heart Association. Circulation. 2013;127:143–152. doi: 10.1161/CIR.0b013e318282ab8f.
    1. Powers WJ, Derdeyn CP, Biller J, Coffey CS, Hoh BL, Jauch EC, Johnston KC, Johnston SC, Khalessi AA, Kidwell CS, et al. American Heart Association/American Stroke Association Focused Update of the 2013 Guidelines for the Early Management of Patients With Acute Ischemic Stroke Regarding Endovascular Treatment: A Guideline for Healthcare Professionals From the American Heart Association/American Stroke Association. Stroke. 2015;2015(46):3020–3035. doi: 10.1161/STR.0000000000000074.
    1. Shuaib A, Butcher K, Mohammad AA, Saqqur M, Liebeskind DS. Collateral blood vessels in acute ischaemic stroke: a potential therapeutic target. Lancet Neurol. 2011;10:909–921. doi: 10.1016/S1474-4422(11)70195-8.
    1. Stokowska A, Atkins AL, Moran J, Pekny T, Bulmer L, Pascoe MC, Barnum SR, Wetsel RA, Nilsson JA, Dragunow M, Pekna M. Complement peptide C3a stimulates neural plasticity after experimental brain ischaemia. Brain. 2017;140:353–369. doi: 10.1093/brain/aww314.
    1. Cramer SC. Repairing the human brain after stroke. II. Restorative therapies. Ann Neurol. 2008;63:549–560.
    1. Wang X, Xuan W, Zhu ZY, Li Y, Zhu H, Zhu L, Fu DY, Yang LQ, Li PY, Yu WF. The evolving role of neuro-immune interaction in brain repair after cerebral ischemic stroke. CNS Neurosci Ther. 2018;24:1100–1114. doi: 10.1111/cns.13077.
    1. Jin Q, Cheng J, Liu Y, Wu J, Wang X, Wei S, Zhou X, Qin Z, Jia J, Zhen X. Improvement of functional recovery by chronic metformin treatment is associated with enhanced alternative activation of microglia/macrophages and increased angiogenesis and neurogenesis following experimental stroke. Brain Behav Immun. 2014;40:131–142. doi: 10.1016/j.bbi.2014.03.003.
    1. Qin C, Fan WH, Liu Q, Shang K, Murugan M, Wu LJ, Wang W, Tian DS. Fingolimod Protects Against Ischemic White Matter Damage by Modulating Microglia Toward M2 Polarization via STAT3 Pathway. Stroke. 2017;48:3336–3346. doi: 10.1161/STROKEAHA.117.018505.
    1. Hu X, Leak RK, Shi Y, Suenaga J, Gao Y, Zheng P, Chen J. Microglial and macrophage polarization-new prospects for brain repair. Nat Rev Neurol. 2015;11:56–64. doi: 10.1038/nrneurol.2014.207.
    1. Ma Y, Wang J, Wang Y, Yang GY. The biphasic function of microglia in ischemic stroke. Prog Neurobiol. 2017;157:247–272. doi: 10.1016/j.pneurobio.2016.01.005.
    1. Charriaut-Marlangue C, Leconte C, Csaba Z, Chafa L, Pansiot J, Talatizi M, Simon K, Moretti R, Marchand-Leroux C, Baud O, Besson VC. Sex differences in the effects of PARP inhibition on microglial phenotypes following neonatal stroke. Brain Behav Immun. 2018;73:375–389. doi: 10.1016/j.bbi.2018.05.022.
    1. Ille F, Sommer L. Wnt signaling: multiple functions in neural development. Cell Mol Life Sci. 2005;62:1100–1108. doi: 10.1007/s00018-005-4552-2.
    1. Libro R, Bramanti P, Mazzon E. The role of the Wnt canonical signaling in neurodegenerative diseases. Life Sci. 2016;158:78–88. doi: 10.1016/j.lfs.2016.06.024.
    1. Matias D, Dubois LG, Pontes B, Rosario L, Ferrer VP, Balca-Silva J, Fonseca ACC, Macharia LW, Romao L, TCLS ES, et al. GBM-Derived Wnt3a Induces M2-Like Phenotype in Microglial Cells Through Wnt/beta-Catenin Signaling. Mol Neurobiol. 2019;56:1517–1530. doi: 10.1007/s12035-018-1150-5.
    1. Ajmone-Cat MA, D'Urso MC, di Blasio G, Brignone MS, De Simone R, Minghetti L. Glycogen synthase kinase 3 is part of the molecular machinery regulating the adaptive response to LPS stimulation in microglial cells. Brain Behav Immun. 2016;55:225–235. doi: 10.1016/j.bbi.2015.11.012.
    1. Wang Y, Bao DJ, Xu B, Cheng CD, Dong YF, Wei XP, Niu CS. Neuroprotection mediated by the Wnt/Frizzled signaling pathway in early brain injury induced by subarachnoid hemorrhage. Neural Regen Res. 2019;14:1013–1024. doi: 10.4103/1673-5374.250620.
    1. Wang W, Li M, Wang Y, Li Q, Deng G, Wan J, Yang Q, Chen Q, Wang J. GSK-3beta inhibitor TWS119 attenuates rtPA-induced hemorrhagic transformation and activates the Wnt/beta-catenin signaling pathway after acute ischemic stroke in rats. Mol Neurobiol. 2016;53:7028–7036. doi: 10.1007/s12035-015-9607-2.
    1. Sabatino M, Hu J, Sommariva M, Gautam S, Fellowes V, Hocker JD, Dougherty S, Qin H, Klebanoff CA, Fry TJ, et al. Generation of clinical-grade CD19-specific CAR-modified CD8+ memory stem cells for the treatment of human B-cell malignancies. Blood. 2016;128:519–528. doi: 10.1182/blood-2015-11-683847.
    1. Chen YQ, Zheng L, Aldarouish M, Zhou ZH, Pan N, Liu JQ, Chen FX, Wang LX. Wnt pathway activator TWS119 enhances the proliferation and cytolytic activity of human gammadeltaT cells against colon cancer. Exp Cell Res. 2018;362:63–71. doi: 10.1016/j.yexcr.2017.11.003.
    1. Doyle KP, Fathali N, Siddiqui MR, Buckwalter MS. Distal hypoxic stroke: a new mouse model of stroke with high throughput, low variability and a quantifiable functional deficit. J Neurosci Methods. 2012;207:31–40. doi: 10.1016/j.jneumeth.2012.03.003.
    1. Cheng J, Alkayed NJ, Hurn PD. Deleterious effects of dihydrotestosterone on cerebral ischemic injury. J Cereb Blood Flow Metab. 2007;27:1553–1562. doi: 10.1038/sj.jcbfm.9600457.
    1. Hu X, Li P, Guo Y, Wang H, Leak RK, Chen S, Gao Y, Chen J. Microglia/macrophage polarization dynamics reveal novel mechanism of injury expansion after focal cerebral ischemia. Stroke. 2012;43:3063–3070. doi: 10.1161/STROKEAHA.112.659656.
    1. Liu Y, Tang G, Li Y, Wang Y, Chen X, Gu X, Zhang Z, Wang Y, Yang GY. Metformin attenuates blood-brain barrier disruption in mice following middle cerebral artery occlusion. J Neuroinflammation. 2014;11:177. doi: 10.1186/s12974-014-0177-4.
    1. Caballero-Garrido E, Pena-Philippides JC, Galochkina Z, Erhardt E, Roitbak T. Characterization of long-term gait deficits in mouse dMCAO, using the CatWalk system. Behav Brain Res. 2017;331:282–296. doi: 10.1016/j.bbr.2017.05.042.
    1. Freret T, Bouet V, Leconte C, Roussel S, Chazalviel L, Divoux D, Schumann-Bard P, Boulouard M. Behavioral deficits after distal focal cerebral ischemia in mice: Usefulness of adhesive removal test. Behav Neurosci. 2009;123:224–230. doi: 10.1037/a0014157.
    1. Caballero-Garrido E, Pena-Philippides JC, Lordkipanidze T, Bragin D, Yang Y, Erhardt EB, Roitbak T. In Vivo Inhibition of miR-155 Promotes Recovery after Experimental Mouse Stroke. J Neurosci. 2015;35:12446–12464. doi: 10.1523/JNEUROSCI.1641-15.2015.
    1. Hayashi-Takagi A, Yagishita S, Nakamura M, Shirai F, Wu YI, Loshbaugh AL, Kuhlman B, Hahn KM, Kasai H. Labelling and optical erasure of synaptic memory traces in the motor cortex. Nature. 2015;525:333–338. doi: 10.1038/nature15257.
    1. Gao C, Qian Y, Huang J, Wang D, Su W, Wang P, Guo L, Quan W, An S, Zhang J, Jiang R. A Three-Day Consecutive Fingolimod Administration Improves Neurological Functions and Modulates Multiple Immune Responses of CCI Mice. Mol Neurobiol. 2017;54:8348–8360. doi: 10.1007/s12035-016-0318-0.
    1. Jin Y, Barnett A, Zhang Y, Yu X, Luo Y. Poststroke Sonic Hedgehog Agonist Treatment Improves Functional Recovery by Enhancing Neurogenesis and Angiogenesis. Stroke. 2017;48:1636–1645. doi: 10.1161/STROKEAHA.117.016650.
    1. Zhu L, Kalimuthu S, Gangadaran P, Oh JM, Lee HW, Baek SH, Jeong SY, Lee SW, Lee J, Ahn BC. Exosomes Derived From Natural Killer Cells Exert Therapeutic Effect in Melanoma. Theranostics. 2017;7:2732–2745. doi: 10.7150/thno.18752.
    1. Plastira I, Bernhart E, Goeritzer M, DeVaney T, Reicher H, Hammer A, Lohberger B, Wintersperger A, Zucol B, Graier WF, et al. Lysophosphatidic acid via LPA-receptor 5/protein kinase D-dependent pathways induces a motile and pro-inflammatory microglial phenotype. J Neuroinflammation. 2017;14:253. doi: 10.1186/s12974-017-1024-1.
    1. Li Y, Zhang X, Cui L, Chen R, Zhang Y, Zhang C, Zhu X, He T, Shen Z, Dong L, et al. Salvianolic acids enhance cerebral angiogenesis and neurological recovery by activating JAK2/STAT3 signaling pathway after ischemic stroke in mice. J Neurochem. 2017;143:87–99. doi: 10.1111/jnc.14140.
    1. Zhou YX, Shi Z, Singh P, Yin H, Yu YN, Li L, Walsh MP, Gui Y, Zheng XL. Potential Role of Glycogen Synthase Kinase-3beta in Regulation of Myocardin Activity in Human Vascular Smooth Muscle Cells. J Cell Physiol. 2016;231:393–402. doi: 10.1002/jcp.25084.
    1. Ding S, Wu TY, Brinker A, Peters EC, Hur W, Gray NS, Schultz PG. Synthetic small molecules that control stem cell fate. Proc Natl Acad Sci U S A. 2003;100:7632–7637. doi: 10.1073/pnas.0732087100.
    1. Wang Y, Liao J, Tang SJ, Shu J, Zhang W. HIV-1 gp120 Upregulates Brain-Derived Neurotrophic Factor (BDNF) Expression in BV2 Cells via the Wnt/beta-Catenin Signaling Pathway. J Mol Neurosci. 2017;62:199–208. doi: 10.1007/s12031-017-0931-z.
    1. Golpich M, Amini E, Hemmati F, Ibrahim NM, Rahmani B, Mohamed Z, Raymond AA, Dargahi L, Ghasemi R, Ahmadiani A. Glycogen synthase kinase-3 beta (GSK-3beta) signaling: Implications for Parkinson's disease. Pharmacol Res. 2015;97:16–26. doi: 10.1016/j.phrs.2015.03.010.
    1. Ransohoff RM. A polarizing question: do M1 and M2 microglia exist? Nat Neurosci. 2016;19:987–991. doi: 10.1038/nn.4338.
    1. Ren C, Yao Y, Han R, Huang Q, Li H, Wang B, Li S, Li M, Mao Y, Mao X, et al. Cerebral ischemia induces angiogenesis in the peri-infarct regions via Notch1 signaling activation. Exp Neurol. 2018;304:30–40. doi: 10.1016/j.expneurol.2018.02.013.
    1. Tian Y, Zhu P, Liu S, Jin Z, Li D, Zhao H, Zhu X, Shu C, Yan D, Dong Z. IL-4-polarized BV2 microglia cells promote angiogenesis by secreting exosomes. Adv Clin Exp Med. 2019;28:421–430. doi: 10.17219/acem/91826.
    1. Hoffmann CJ, Harms U, Rex A, Szulzewsky F, Wolf SA, Grittner U, Lattig-Tunnemann G, Sendtner M, Kettenmann H, Dirnagl U, et al. Vascular signal transducer and activator of transcription-3 promotes angiogenesis and neuroplasticity long-term after stroke. Circulation. 2015;131:1772–1782. doi: 10.1161/CIRCULATIONAHA.114.013003.
    1. Xu Y, Zhang G, Kang Z, Xu Y, Jiang W, Zhang S. Cornin increases angiogenesis and improves functional recovery after stroke via the Ang1/Tie2 axis and the Wnt/beta-catenin pathway. Arch Pharm Res. 2016;39:133–142. doi: 10.1007/s12272-015-0652-1.
    1. Esposito E, Hayakawa K, Ahn BJ, Chan SJ, Xing C, Liang AC, Kim KW, Arai K, Lo EH. Effects of ischemic post-conditioning on neuronal VEGF regulation and microglial polarization in a rat model of focal cerebral ischemia. J Neurochem. 2018;146:160–172. doi: 10.1111/jnc.14337.
    1. Sellgren CM, Sheridan SD, Gracias J, Xuan D, Fu T, Perlis RH. Patient-specific models of microglia-mediated engulfment of synapses and neural progenitors. Mol Psychiatry. 2017;22:170–177. doi: 10.1038/mp.2016.220.
    1. Kwon MJ, Kim J, Shin H, Jeong SR, Kang YM, Choi JY, Hwang DH, Kim BG. Contribution of macrophages to enhanced regenerative capacity of dorsal root ganglia sensory neurons by conditioning injury. J Neurosci. 2013;33:15095–15008. doi: 10.1523/JNEUROSCI.0278-13.2013.
    1. Hilla AM, Diekmann H, Fischer D. Microglia Are Irrelevant for Neuronal Degeneration and Axon Regeneration after Acute Injury. J Neurosci. 2017;37:6113–6124. doi: 10.1523/JNEUROSCI.0584-17.2017.

Source: PubMed

3
Abonner