Block of A1 astrocyte conversion by microglia is neuroprotective in models of Parkinson's disease

Seung Pil Yun, Tae-In Kam, Nikhil Panicker, SangMin Kim, Yumin Oh, Jong-Sung Park, Seung-Hwan Kwon, Yong Joo Park, Senthilkumar S Karuppagounder, Hyejin Park, Sangjune Kim, Nayeon Oh, Nayoung Alice Kim, Saebom Lee, Saurav Brahmachari, Xiaobo Mao, Jun Hee Lee, Manoj Kumar, Daniel An, Sung-Ung Kang, Yunjong Lee, Kang Choon Lee, Dong Hee Na, Donghoon Kim, Sang Hun Lee, Viktor V Roschke, Shane A Liddelow, Zoltan Mari, Ben A Barres, Valina L Dawson, Seulki Lee, Ted M Dawson, Han Seok Ko, Seung Pil Yun, Tae-In Kam, Nikhil Panicker, SangMin Kim, Yumin Oh, Jong-Sung Park, Seung-Hwan Kwon, Yong Joo Park, Senthilkumar S Karuppagounder, Hyejin Park, Sangjune Kim, Nayeon Oh, Nayoung Alice Kim, Saebom Lee, Saurav Brahmachari, Xiaobo Mao, Jun Hee Lee, Manoj Kumar, Daniel An, Sung-Ung Kang, Yunjong Lee, Kang Choon Lee, Dong Hee Na, Donghoon Kim, Sang Hun Lee, Viktor V Roschke, Shane A Liddelow, Zoltan Mari, Ben A Barres, Valina L Dawson, Seulki Lee, Ted M Dawson, Han Seok Ko

Abstract

Activation of microglia by classical inflammatory mediators can convert astrocytes into a neurotoxic A1 phenotype in a variety of neurological diseases1,2. Development of agents that could inhibit the formation of A1 reactive astrocytes could be used to treat these diseases for which there are no disease-modifying therapies. Glucagon-like peptide-1 receptor (GLP1R) agonists have been indicated as potential neuroprotective agents for neurologic disorders such as Alzheimer's disease and Parkinson's disease3-13. The mechanisms by which GLP1R agonists are neuroprotective are not known. Here we show that a potent, brain-penetrant long-acting GLP1R agonist, NLY01, protects against the loss of dopaminergic neurons and behavioral deficits in the α-synuclein preformed fibril (α-syn PFF) mouse model of sporadic Parkinson's disease14,15. NLY01 also prolongs the life and reduces the behavioral deficits and neuropathological abnormalities in the human A53T α-synuclein (hA53T) transgenic mouse model of α-synucleinopathy-induced neurodegeneration16. We found that NLY01 is a potent GLP1R agonist with favorable properties that is neuroprotective through the direct prevention of microglial-mediated conversion of astrocytes to an A1 neurotoxic phenotype. In light of its favorable properties, NLY01 should be evaluated in the treatment of Parkinson's disease and related neurologic disorders characterized by microglial activation.

Conflict of interest statement

Competing financial Interests Statement

Z.M., V.L.D., S.L., T.M.D., H.S.K are co-founders of Neuraly Inc. and hold ownership equity in the company. This arrangement has been reviewed and approved by the Johns Hopkins University in accordance with its conflict of interest policies. V.V.R. is the CSO of Neuraly Inc.

Figures

Figure 1
Figure 1
α-syn PFF-induced PD like pathology is rescued by NLY01. (a) Schematic diagram of the α-syn PFF experimental design. (b) Representative double-immunostaining for p-α-synser129 (p-α-syn) (green) and TH (red) in the SNpc. White arrows point to DA neuron loss. Low power images were generated by the tile scan algorithm in the Zen software (n=6, biologically independent animals). Scale bar, 100 μm or 10 μm, respectively. (c) Percentage of TH neurons with p-α-synser129 positive inclusion in the SNpc region. Error bars represent the mean ± S.E.M. (n=6, biologically independent animals, p value = 0.0007). Unpaired two-tailed Student’s test were used for statistical analysis. (d) Representative photomicrographs from coronal mesencephalon sections containing TH-positive neurons in the SNpc region (n=10, biologically independent animals). Scale bar, 500 μm. (e) Unbiased stereologic counts of TH and (f) Nissl-positive neurons in the SNpc region. Error bars represent the mean ± S.E.M. (n=10, biologically independent animals). (g) HPLC assessment of DA concentrations in the striatum of PBS and α-syn PFF stereotaxic injected mice that were treated with vehicle or NLY01. Error bars represent the mean ± S.E.M. (n=5, biologically independent animals). (h-i) Behavioral tests after PBS or α-syn PFF stereotaxic intrastriatal injection at six-months in vehicle or NLY01 treated mice. (h) Amphetamine rotation test, and (i) pole test. Error bars represent the mean ± S.E.M. (n=12 PBS with vehicle, n=13 PBS with NLY01, n=14 α-syn PFF with vehicle, and n=11 α-syn PFF with NLY01, biologically independent animals). Two-way ANOVA was used for statistical analysis followed by Tukey’s multiple comparisons test. #P < 0.05, ##P < 0.01, ###P < 0.001 vs. PBS stereotaxic injected mice with vehicle; *P < 0.05, **P < 0.01, ***P < 0.001 vs. α-syn PFF stereotaxic injected mice with NLY01. Maximum time to climb down the pole was limited to 60 sec.
Figure 2
Figure 2
NLY01 reduces the pathology in the hA53T α-syn transgenic mice. (a) Kaplan-Meier survival curves of littermate wild type (WT) and hA53T α-syn Tg mice treated with vehicle or NLY01 (3 mg/kg, n=20, biologically independent animals, p value < 0.0001). Statistical analysis for the survival curves were performed by Log-rank (Mantel-Cox) test. (b, d) Representative p-α-synser129 (p-α-syn) immunohistochemistry and immunostaining (red) in the lateral vestibular nucleus (LVe) of the brainstem (n=5, biologically independent animals). Low power images were generated by the tile scan algorithm in the Zen software. White arrow head (tile image line). Scale bar, 200 μm or 25 μm, respectively. (c, e) Quantification of LVe neurons containing p-α-synser129 positive-immunoreactivity inclusions. Error bars represent the mean ± S.E.M. (n=5, biologically independent animals, p value < 0.0001 for panel c and p value = 0.0003 for panel e). (f) Representative ubiquitin immunostaining (green) in the LVe (n=5, biologically independent animals). Scale bar, 200 μm or 25 μm. (g, h) Quantification of LVe neurons that are ubiquitin-positive. Error bars represent the mean ± S.E.M (n=5, biologically independent animals). (i) Representative immunoblots of α-syn, p-α-synser129 and β-actin in detergent (Triton-X100) insoluble fractions and detergent soluble fractions of the brainstem from ten-month-old WT and hA53T α-syn Tg mice treated with vehicle or NLY01 for four months (cropped blot images are shown, see Supplementary Fig. 21 for full immunoblots). (j) Quantification of α-syn monomer, aggregation, and p-α-synser129 protein levels in detergent insoluble fractions normalized to β-actin. Error bars represent the mean ± S.E.M. (n=5, biologically independent animals, p value < 0.0001). (k) Quantification of α-syn monomer and p-α-synser129 protein levels in detergent soluble fractions normalized to β-actin. Error bars represent the mean ± S.E.M. (n=5, biologically independent animals, α-syn, p value < 0.0001 and p-α-syn, p value = 0.0002). Unpaired two-tailed Student’s test or two-way ANOVA was used for statistical analysis followed by Tukey’s multiple comparisons test. #P < 0.05, ###P < 0.001 vs. WT with vehicle; *P < 0.05, ***P < 0.001 vs. hA53T α-syn Tg with NLY01.
Figure 3
Figure 3
Inhibition of α-syn PFF-induced microglial activation by NLY01. (a) Glp1r mRNA expression was analyzed in various mouse brain regions. OB, olfactory bulb; FCtx, frontal cortex; Ctx, cerebral cortex; Str, striatum; Hippo, hippocampus; VMB, ventral midbrain; CB, cerebellum; BS, brainstem. Error bars represent the mean ± S.E.M. (n=4, biologically independent animals). (b) Predominant expression of GLP-1R in astrocytes and microglia. Mouse primary astrocytes (GFAP), microglia (Iba-1) and cortical neurons (Tuj1) were subjected to western blotting using GLP-1R antibody (cropped blot images are shown, see Supplementary Fig. 21 for full immunoblots). β-actin is provided as a loading control (n=3, biologically independent primary cells). (c) The Glp1r mRNA expression in α-syn PFF-activated primary cultured astrocyte, microglia, and neurons was analyzed by real-time RT-PCR. Bars represent the mean ± S.E.M. (n=3, biologically independent primary cells, primary microglia, p value = 0.0475 and primary cortical neuron, p value = 0.0045). (d) Schematic diagram showing treatment of α-syn PFF-astrocyte conditioned medium (ACM) or direct treatment of α-syn PFF into neurons. (e-g) Quantitative PCR analysis of NLY01 (1 μM) pretreatment on α-syn PFF-induced (1 μg/ml) microglial activation markers, (e) Il1a, (f), Tnfα, and (g) C1qa. Bars represent the mean ± S.E.M. (n=3, biologically independent primary microglia). (h-j) Cytokine analysis of α-syn PFF-activated microglia-conditioned medium (MCM) 18 hrs after α-syn PFF treatment by ELISA. NLY01 (1 μM) pretreatment prevented the increase of (h) IL-1α, (i) TNFα, and (j) C1q. Bars represent the mean ± S.E.M. (n=4, biologically independent primary microglia-conditioned medium). (k-m) Inhibition of α-syn PFF-induced microglial activation by NLY01 in vivo. (k) Representative immunohistochemical images of Iba-1. Scale bar, 50 μm. (l) Intensity of Iba-1 positive signals in the SNpc, (m) Quantification of Iba-1 positive cell number in the SNpc. Bars represent the mean ± S.E.M. (n=6, biologically independent animals). (n) Representative immunoblots of Iba-1 and β-actin in the ventral midbrain of PBS and α-syn PFF injected mice treated with vehicle or NLY01 (cropped blot images are shown, see Supplementary Fig. 21 for full immunoblots). (o) Quantification of Iba-1 levels in ventral midbrain normalized to β-actin. Bars represent the mean ± S.E.M. (n=3, biologically independent animals). (p-r) Quantitative PCR for (p) Il1a (q) Tnfα, and (r) C1qa in the ventral midbrain of α-syn PFF injected mice. Bars represent the mean ± S.E.M. (n=4, biologically independent animals). Unpaired two-tailed Student’s test or two-way ANOVA was used for statistical analysis followed by Tukey’s multiple comparisons test. #P < 0.05, ##P < 0.01, ###P < 0.001 vs. PBS with vehicle; *P < 0.05, **P < 0.01, ***P < 0.001 vs. or α-syn PFF with NLY01.
Figure 4
Figure 4
Inhibition of α-syn PFF-induced A1 reactive astrocytes by NLY01. (a) Schematic diagram showing the collection of astrocyte samples after treatment with MCM of α-syn PFF with or without NLY01 treated microglia. (b) Representative immunoblots of GFAP in primary astrocytes treated with α-syn PFF-MCM with vehicle or NLY01 (1 μM) (cropped blot images are shown, see Supplementary Fig. 21 for full immunoblots). (c) Quantification of GFAP levels. Bars represent the mean ± S.E.M. (n=3, biologically independent primary astrocytes). (d) Inhibition of α-syn PFF-induced A1 reactive astrocytes by NLY01 in purified primary astrocytes. Bars represent the mean ± S.E.M. (n=3, biologically independent primary astrocytes). (e, f) Inhibition of α-syn PFF-induced A1 reactive astrocytes by NLY01 in vivo. (e) Representative images of immunohistochemistry and (f) quantification of GFAP. Bars represent the mean ± S.E.M. (n=6, biologically independent animals). Scale bar, 50 μm. (g) Representative immunoblots of GFAP in the ventral midbrain region (cropped blot images are shown, see Supplementary Fig. 21 for full immunoblots). (h) Quantification of GFAP levels. Bars represent the mean ± S.E.M (n=4, biologically independent animals). (i) Co-localization of C3d and GFAP as assessed by confocal microscopy. Scale bar, 10 μm. (j) Percentage of GFAP positive cells that are C3d positive in the SNpc region. Bars represent the mean ± S.E.M. (n=5, biologically independent animals). (k) Increase in expression of C3 transcripts, which is prevented by NLY01 treatment in α-syn PFF injected mice. Bars represent the mean ± S.E.M. (n=4, biologically independent animals). (l) Quantitative PCR analysis of purified astrocytes from the ventral midbrain of α-syn PFF injected mice with vehicle or NLY01. Bars represent the mean ± S.E.M. (n=4, biologically independent animals). Two-way ANOVA was used for statistical analysis followed by Tukey’s multiple comparisons test. #P < 0.05, ##P < 0.01, ###P < 0.001 vs. PBS with vehicle; *P < 0.05, **P < 0.01, ***P < 0.001 vs. α-syn PFF with NLY01 or with Veh. n.s, not significant.

References

    1. Liddelow SA, Barres BA. Reactive Astrocytes: Production, Function, and Therapeutic Potential. Immunity. 2017;46:957–967.
    1. Liddelow SA, et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature. 2017;541:481–487.
    1. Athauda D, Foltynie T. The glucagon-like peptide 1 (GLP) receptor as a therapeutic target in Parkinson’s disease: mechanisms of action. Drug Discov Today. 2016;21:802–818.
    1. Aviles-Olmos I, et al. Exenatide and the treatment of patients with Parkinson’s disease. J Clin Invest. 2013;123:2730–2736.
    1. Chen S, et al. Glucagon-like peptide-1 protects hippocampal neurons against advanced glycation end product-induced tau hyperphosphorylation. Neuroscience. 2014;256:137–146.
    1. Harkavyi A, Whitton PS. Glucagon-like peptide 1 receptor stimulation as a means of neuroprotection. Br J Pharmacol. 2010;159:495–501.
    1. Li Y, et al. Liraglutide is neurotrophic and neuroprotective in neuronal cultures and mitigates mild traumatic brain injury in mice. J Neurochem. 2015;135:1203–1217.
    1. Li Y, et al. GLP-1 receptor stimulation preserves primary cortical and dopaminergic neurons in cellular and rodent models of stroke and Parkinsonism. Proceedings of the National Academy of Sciences of the United States of America. 2009;106:1285–1290.
    1. Meier JJ. GLP-1 receptor agonists for individualized treatment of type 2 diabetes mellitus. Nat Rev Endocrinol. 2012;8:728–742.
    1. Rachmany L, et al. Exendin-4 induced glucagon-like peptide-1 receptor activation reverses behavioral impairments of mild traumatic brain injury in mice. Age (Dordr) 2013;35:1621–1636.
    1. Teramoto S, et al. 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. Tweedie D, et al. Blast traumatic brain injury-induced cognitive deficits are attenuated by preinjury or postinjury treatment with the glucagon-like peptide-1 receptor agonist, exendin-4. Alzheimers Dement. 2016;12:34–48.
    1. Athauda D, et al. Exenatide once weekly versus placebo in Parkinson’s disease: a randomised, double-blind, placebo-controlled trial. Lancet. 2017;390:1664–1675.
    1. Luk KC, et al. Pathological alpha-synuclein transmission initiates Parkinson-like neurodegeneration in nontransgenic mice. Science. 2012;338:949–953.
    1. Mao X, et al. Pathological alpha-synuclein transmission initiated by binding lymphocyte-activation gene 3. Science. 2016;353
    1. Lee MK, et al. Human alpha-synuclein-harboring familial Parkinson’s disease-linked Ala-53 –> Thr mutation causes neurodegenerative disease with alpha-synuclein aggregation in transgenic mice. Proceedings of the National Academy of Sciences of the United States of America. 2002;99:8968–8973.
    1. Cabezas R, et al. Astrocytic modulation of blood brain barrier: perspectives on Parkinson’s disease. Front Cell Neurosci. 2014;8:211.
    1. Gray MT, Woulfe JM. Striatal blood-brain barrier permeability in Parkinson’s disease. J Cereb Blood Flow Metab. 2015;35:747–750.
    1. Polinski NK, et al. Best Practices for Generating and Using Alpha-Synuclein Pre-Formed Fibrils to Model Parkinson’s Disease in Rodents. J Parkinsons Dis. 2018
    1. Brahmachari S, et al. Activation of tyrosine kinase c-Abl contributes to alpha-synuclein-induced neurodegeneration. J Clin Invest. 2016;126:2970–2988.
    1. Zhang Y, et al. An RNA-sequencing transcriptome and splicing database of glia, neurons, and vascular cells of the cerebral cortex. J Neurosci. 2014;34:11929–11947.
    1. Kim C, Lee HJ, Masliah E, Lee SJ. Non-cell-autonomous Neurotoxicity of alpha-synuclein Through Microglial Toll-like Receptor 2. Exp Neurobiol. 2016;25:113–119.
    1. Daher JP, et al. Neurodegenerative phenotypes in an A53T alpha-synuclein transgenic mouse model are independent of LRRK2. Hum Mol Genet. 2012;21:2420–2431.
    1. Harms AS, et al. Peripheral monocyte entry is required for alpha-Synuclein induced inflammation and Neurodegeneration in a model of Parkinson disease. Experimental neurology. 2018;300:179–187.
    1. Martinez FO, Gordon S. The M1 and M2 paradigm of macrophage activation: time for reassessment. F1000Prime Rep. 2014;6:13.
    1. Allen Reish HE, Standaert DG. Role of alpha-synuclein in inducing innate and adaptive immunity in Parkinson disease. J Parkinsons Dis. 2015;5:1–19.
    1. Goncalves A, et al. Protective Effect of a GLP-1 Analog on Ischemia-Reperfusion Induced Blood-Retinal Barrier Breakdown and Inflammation. Invest Ophthalmol Vis Sci. 2016;57:2584–2592.
    1. Gullo F, et al. Plant Polyphenols and Exendin-4 Prevent Hyperactivity and TNF-alpha Release in LPS-Treated In vitro Neuron/Astrocyte/Microglial Networks. Front Neurosci. 2017;11:500.
    1. Kim S, Moon M, Park S. Exendin-4 protects dopaminergic neurons by inhibition of microglial activation and matrix metalloproteinase-3 expression in an animal model of Parkinson’s disease. J Endocrinol. 2009;202:431–439.
    1. Lee CH, et al. Activation of Glucagon-Like Peptide-1 Receptor Promotes Neuroprotection in Experimental Autoimmune Encephalomyelitis by Reducing Neuroinflammatory Responses. Mol Neurobiol. 2017
    1. Li Y, et al. Exendin-4 ameliorates motor neuron degeneration in cellular and animal models of amyotrophic lateral sclerosis. PLoS One. 2012;7:e32008.
    1. Wu HY, Tang XQ, Liu H, Mao XF, Wang YX. Both classic Gs-cAMP/PKA/CREB and alternative Gs-cAMP/PKA/p38beta/CREB signal pathways mediate exenatide-stimulated expression of M2 microglial markers. J Neuroimmunol. 2017
    1. Ventorp F, et al. Exendin-4 Treatment Improves LPS-Induced Depressive-Like Behavior Without Affecting Pro-Inflammatory Cytokines. J Parkinsons Dis. 2017;7:263–273.
    1. Ji C, et al. A novel dual GLP-1 and GIP receptor agonist is neuroprotective in the MPTP mouse model of Parkinson’s disease by increasing expression of BNDF. Brain Res. 2016;1634:1–11.
    1. Salcedo I, Tweedie D, Li Y, Greig NH. Neuroprotective and neurotrophic actions of glucagon-like peptide-1: an emerging opportunity to treat neurodegenerative and cerebrovascular disorders. Br J Pharmacol. 2012;166:1586–1599.
    1. Volpicelli-Daley LA, Luk KC, Lee VM. Addition of exogenous alpha-synuclein preformed fibrils to primary neuronal cultures to seed recruitment of endogenous alpha-synuclein to Lewy body and Lewy neurite-like aggregates. Nat Protoc. 2014;9:2135–2146.
    1. Kim TH, et al. Mono-PEGylated dimeric exendin-4 as high receptor binding and long-acting conjugates for type 2 anti-diabetes therapeutics. Bioconjug Chem. 2011;22:625–632.
    1. Kim TH, et al. Site-specific PEGylated Exendin-4 modified with a high molecular weight trimeric PEG reduces steric hindrance and increases type 2 antidiabetic therapeutic effects. Bioconjug Chem. 2012;23:2214–2220.
    1. Karuppagounder SS, et al. The c-Abl inhibitor, nilotinib, protects dopaminergic neurons in a preclinical animal model of Parkinson’s disease. Sci Rep. 2014;4:4874.
    1. Lee Y, et al. Parthanatos mediates AIMP2-activated age-dependent dopaminergic neuronal loss. Nat Neurosci. 2013;16:1392–1400.
    1. Kim S, et al. GBA1 deficiency negatively affects physiological alpha-synuclein tetramers and related multimers. Proceedings of the National Academy of Sciences of the United States of America. 2018;115:798–803.
    1. Panicker N, et al. Fyn Kinase Regulates Microglial Neuroinflammatory Responses in Cell Culture and Animal Models of Parkinson’s Disease. J Neurosci. 2015;35:10058–10077.
    1. Andrabi SA, et al. Iduna protects the brain from glutamate excitotoxicity and stroke by interfering with poly(ADP-ribose) polymer-induced cell death. Nat Med. 2011;17:692–699.
    1. Wang Y, et al. A nuclease that mediates cell death induced by DNA damage and poly(ADP-ribose) polymerase-1. Science. 2016;354

Source: PubMed

3
Sottoscrivi