Tyrosine Kinase Inhibition Regulates Early Systemic Immune Changes and Modulates the Neuroimmune Response in α-Synucleinopathy

Michaeline L Hebron, Irina Lonskaya, Paul Olopade, Sandra T Selby, Fernando Pagan, Charbel E-H Moussa, Michaeline L Hebron, Irina Lonskaya, Paul Olopade, Sandra T Selby, Fernando Pagan, Charbel E-H Moussa

Abstract

Objectives: Neuro-inflammation is common in α-Synucleinopathies and Tauopathies; and evidence suggests a link between the tyrosine kinase Abl and neurodegeneration. Abl upregulates α-Synuclein and promotes Tau hyper-phosphorylation (p-Tau), while Abl inhibitors facilitate autophagic clearance.

Methods: A model of α-Synucleinopathy harboring human mutant A53T α-Synuclein and exhibits concomitant increase in murine p-Tau was used to determine the immunological response to Abl inhibition.

Results: Age-dependent alterations of brain immunity, including loss of IL-10 and decreased levels of IL-2 and IL-3 were observed in old A53T mice. Brain CCL2 and CCL5 were decreased, but CX3CL1 remained constantly elevated. Young A53T mice exhibited differential systemic and central immune profiles in parallel with increased blood markers of adaptive immunity, suggesting an early systemic immune response. Tyrosine kinase inhibitors (TKIs), including nilotinib and bosutinib reduced brain and peripheral α-Synuclein and p-Tau and modulated blood immunological responses. TKIs did not affect brain IL-10, but they changed the levels of all measured blood immune markers, except CX3CL1. TKIs altered microglia morphology and reduced the number of astrocyte and dendritic cells, suggesting beneficial regulation of microglia.

Conclusions: These data indicate that tyrosine kinase inhibition affects neuro-inflammation via early changes of the peripheral immune profile, leading to modulation of the neuro-immune response to α-Synuclein and p-Tau.

Keywords: Abl; Bosutinib; Inflammation; Microglia; Nilotinib; Tau; α-Synuclein.

Conflict of interest statement

Conflict of Interest

Dr. Charbel Moussa has a provisional patent application to use nilotinib and bosutinib to treat neurodegenerative diseases. Other authors declare no conflict of interest in association with this manuscript.

Figures

Figure 1. Age-dependent alterations of brain immunity…
Figure 1. Age-dependent alterations of brain immunity in A53T mice
Histograms represent ELISA levels of A) α-Synuclein and p-Tau in the brain of A53T mice, and B) shows caspase-3 activity in young and older mice. Graphs represent the levels of mouse A53T brain immune markers, including C) pro-inflammatory IL-1α, IL-1β, IL-6 and TNF-α, D) anti-inflammatory IL-4 and IL-10, E) modulators of immune memory IL-2 and IL3, F) VEGF and IFN-γ and G) chemokines CCL2, CCL5 and CX3CL1. n= 5 for each strain at each time point. ANOVA, Neuman Keuls, Mean±SD, * indicates significantly different than WT with p<0.05, **p<0.01, ***p<0.001, **** p<0.0001.
Figure 2. Differential levels of immune markers…
Figure 2. Differential levels of immune markers in A53T brain and blood
Histograms represent levels of A) pro-inflammatory cytokines IL-1α, IL-1β, IL-6 and TNF-α in the brain and total blood of A53T mice, B) anti-inflammatory cytokines IL-4 and IL-10, C) modulators of immune memory IL-2 and IL3, D) VEGF and IFN-γ and E) chemokines CCL2, CCL5 and CX3CL1. n=4 for each strain at each time point. ANOVA, Neuman Keuls, Mean±SD, * indicates significantly different than WT with p<0.05, **p<0.01, ***p<0.001. Histograms represent ELISA levels of F) α-Synuclein and p-Tau and G) caspase-3 activity in the brain of A53T mice treated I.P with 10 mg/kg nilotinib or 5 mg/kg bosutinib or 3 μL DMSO every other day for 6 weeks. n=4 for each strain at each time point. ANOVA, Neuman Keuls, Mean±SD, * indicates significantly different than WT with p<0.05, **p<0.01, ***p<0.001.
Figure 3. Nilotinib and bosutinib decrease CNS…
Figure 3. Nilotinib and bosutinib decrease CNS and peripheral levels of α-Synuclein and p-Tau and modulate cytokine levels
Histograms represent ELISA levels of A) α-Synuclein and B) p-Tau in the spleen, heart, muscle, intestine and blood of A53T mice treated I.P with 10 mg/kg nilotinib or 5 mg/kg bosutinib or 3 μL DMSO every other day for 6 weeks. C) Western blot analysis on 10% S NuPAGE gel showing analysis of muscle and intestine homogenized in 1xSTEN buffer to compare nilotinib and bosutinib effects with DMSO in A53T mice. D) brain and E) blood levels of pro-inflammatory IL-1α, IL-1β, IL-6 and TNF-α. F) brain and G) blood levels of anti-inflammatory IL-4 and IL-10. n= 5 for each strain at each time point. ANOVA, Neuman Keuls, Mean±SD, * indicates significantly different than W, p<0.05.
Figure 4. Nilotinib and bosutinib modulate changes…
Figure 4. Nilotinib and bosutinib modulate changes in the blood immunological profiles of A53T mice
Histograms represent ELISA levels in A53T mice treated I.P with 10 mg/kg nilotinib or 5 mg/kg bosutinib or 3 μL DMSO every other day for 6 weeks in A) brain and B) blood levels of modulators of immune memory IL-2 and IL3, C) brain and D) blood VEGF and IFN-γ and E) and F) blood levels chemokines CCL2, CCL5 and CX3CL1. n=5 for each strain at each time point. ANOVA, Neuman Keuls, Mean±SD, * indicates significantly different than WT with p<0.05, **p<0.01.
Figure 5. Nilotinib and bosutinib alter microglia…
Figure 5. Nilotinib and bosutinib alter microglia morphology and reduce the number of astrocyte and dendritic cells
Coronal 20 μm thick brain sections show IBA-1 and nuclear DAPI staining of microglia in the striatum of A) WT mice, B) A53T mice treated with DMSO, insert is higher magnification, C) A53T mice treated with bosutinib, insert is higher magnification, and D) A53T mice treated with nilotinib, insert is higher magnification. E) histograms represent stereological quantification. Coronal 20 μm thick brain sections show GFAP and nuclear DAPI staining of astrocytes in the striatum of F) WT mice, G) A53T mice treated with DMSO, H) A53T mice treated with bosutinib, and I) A53T mice treated with nilotinib. J) histograms represent stereological quantification. Dendritic cells stained with CD11b and nuclear DAPI labeling in the striatum of K) WT mice, L) A53T mice treated with DMSO, M) A53T mice treated with bosutinib, and N) A53T mice treated with nilotinib. O) histograms represent stereological quantification. n=5 for each strain at each time point. n=4, ANOVA, Neuman Keuls, Mean±SD, * indicates significantly different than WT with p<0.05, **p<0.01.

References

    1. Akiyama H, Barger S, Barnum S, Bradt B, Bauer J, et al. Inflammation and Alzheimer’s disease. Neurobiol Aging. 2000;21:383–421.
    1. Roodveldt C, Christodoulou J, Dobson CM. Immunological features of alpha-synuclein in Parkinson’s disease. J Cell Mol Med. 2008;12:1820–1829.
    1. Reynolds AD, Kadiu I, Garg SK, Glanzer JG, Nordgren T, et al. Nitrated alpha-synuclein and microglial neuroregulatory activities. J Neuroimmune Pharmacol. 2008;3:59–74.
    1. Zhang W, Wang T, Pei Z, Miller DS, Wu X, et al. Aggregated alpha-synuclein activates microglia: a process leading to disease progression in Parkinson’s disease. FASEB J. 2005;19:533–542.
    1. Reynolds AD, Banerjee R, Liu J, Gendelman HE, Mosley RL. Neuroprotective activities of CD4+CD25+ regulatory T cells in an animal model of Parkinson’s disease. J Leukoc Biol. 2007;82:1083–1094.
    1. Su X, Maguire-Zeiss KA, Giuliano R, Prifti L, Venkatesh K, et al. Synuclein activates microglia in a model of Parkinson’s disease. Neurobiol Aging. 2008;29:1690–1701.
    1. Gebicke-Haerter PJ. Microglia in neurodegeneration: molecular aspects. Microsc Res Tech. 2001;54:47–58.
    1. Gerhard A, Pavese N, Hotton G, Turkheimer F, Es M, et al. In vivo imaging of microglial activation with [11C](R)-PK11195 PET in idiopathic Parkinson’s disease. Neurobiol Dis. 2006;21:404–412.
    1. Ishizawa K, Dickson DW. Microglial activation parallels system degeneration in progressive supranuclear palsy and corticobasal degeneration. J Neuropathol Exp Neurol. 2001;60:647–657.
    1. Khandelwal PJ, Dumanis SB, Feng LR, Maguire-Zeiss K, Rebeck G, et al. Parkinson-related parkin reduces alpha-Synuclein phosphorylation in a gene transfer model. Mol Neurodegener. 2010;5:47.
    1. Khandelwal PJ, Dumanis SB, Herman AM, Rebeck GW, Moussa CE. Wild type and P301L mutant Tau promote neuro-inflammation and alpha-Synuclein accumulation in lentiviral gene delivery models. Mol Cell Neurosci. 2012;49:44–53.
    1. Li Y, Liu L, Barger SW, Griffin WS. Interleukin-1 mediates pathological effects of microglia on tau phosphorylation and on synaptophysin synthesis in cortical neurons through a p38-MAPK pathway. J Neurosci. 2003;23:1605–1611.
    1. Quintanilla RA, Orellana DI, Gonzalez-Billault C, Maccioni RB. Interleukin-6 induces Alzheimer-type phosphorylation of tau protein by deregulating the cdk5/p35 pathway. Exp Cell Res. 2004;295:245–257.
    1. Saez TE, Pehar M, Vargas M, Barbeito L, Maccioni RB. Astrocytic nitric oxide triggers tau hyperphosphorylation in hippocampal neurons. In Vivo. 2004;18:275–280.
    1. Kitazawa M, Yamasaki TR, LaFerla FM. Microglia as a potential bridge between the amyloid beta-peptide and tau. Ann N Y Acad Sci. 2004;1035:85–103.
    1. Yoshiyama Y, Higuchi M, Zhang B, Huang SM, Iwata N, et al. Synapse loss and microglial activation precede tangles in a P301S tauopathy mouse model. Neuron. 2007;53:337–351.
    1. Schlatterer SD, Suh HS, Conejero-Goldberg C, Chen S, Acker CM, et al. Neuronal c-Abl activation leads to induction of cell cycle and interferon signaling pathways. J Neuroinflammation. 2012;9:208.
    1. Schlatterer SD, Acker CM, Davies P. c-Abl in neurodegenerative disease. J Mol Neurosci. 2011;45:445–452.
    1. Schlatterer SD, Tremblay MA, Acker CM, Davies P. Neuronal cAbl overexpression leads to neuronal loss and neuroinflammation in the mouse forebrain. J Alzheimers Dis. 2011;25:119–133.
    1. Hebron ML, Lonskaya I, Moussa CE. Tyrosine kinase inhibition facilitates autophagic SNCA/alpha-synuclein clearance. Autophagy. 2013;9:1249–1250.
    1. Hebron ML, Lonskaya I, Moussa CE. Nilotinib reverses loss of dopamine neurons and improves motor behavior via autophagic degradation of alpha-synuclein in Parkinson’s disease models. Hum Mol Genet. 2013;22:3315–3328.
    1. Mahul-Mellier AL, Fauvet B, Gysbers A, Dikiy I, Oueslati A, et al. c-Abl phosphorylates alpha-synuclein and regulates its degradation: implication for alpha-synuclein clearance and contribution to the pathogenesis of Parkinson’s disease. Hum Mol Genet. 2014;23:2858–2879.
    1. Schlatterer SD, Acker CM, Davies P. c-Abl in neurodegenerative disease. J Mol Neurosci. 2011;45:445–452.
    1. Tremblay MA, Acker CM, Davies P. Tau phosphorylated at tyrosine 394 is found in Alzheimer’s disease tangles and can be a product of the Abl-related kinase, Arg. J Alzheimers Dis. 2010;19:721–733.
    1. Derkinderen P, Scales TM, Hanger DP, Leung KY, Byers HL, et al. Tyrosine 394 is phosphorylated in Alzheimer’s paired helical filament tau and in fetal tau with c-Abl as the candidate tyrosine kinase. J Neurosci. 2005;25:6584–6593.
    1. Jing Z, Caltagarone J, Bowser R. Altered subcellular distribution of c-Abl in Alzheimer’s disease. J Alzheimers Dis. 2009;17:409–422.
    1. Ittner LM, Ke YD, Delerue F, Bi M, Gladbach A, et al. Dendritic function of tau mediates amyloid-beta toxicity in Alzheimer’s disease mouse models. Cell. 2010;142:387–397.
    1. Reynolds CH, Garwood CJ, Wray S, Price C, Kellie S, et al. Phosphorylation regulates tau interactions with Src homology 3 domains of phosphatidylinositol 3-kinase, phospholipase Cgamma, Grb2, and Src family kinases. J Biol Chem. 2008;283:18177–18186.
    1. Lee G. Tau and src family tyrosine kinases. Biochim Biophys Acta. 2005;1739:323–330.
    1. Lonskaya I, Desforges NM, Hebron ML, Moussa CE. Ubiquitination increases parkin activity to promote autophagic alpha-synuclein clearance. PLoS One. 2013;8:e83914.
    1. Lonskaya I, Hebron ML, Algarzae NK, Desforges N, Moussa CE. Decreased parkin solubility is associated with impairment of autophagy in the nigrostriatum of sporadic Parkinson’s disease. Neuroscience. 2012;232C:90.
    1. Lonskaya I, Hebron ML, Desforges NM, Franjie A, Moussa CE. Tyrosine kinase inhibition increases functional parkin-Beclin-1 interaction and enhances amyloid clearance and cognitive performance. EMBO Mol Med. 2013;5:1247–1262.
    1. Lonskaya I, Hebron ML, Desforges NM, Schachter JB, Moussa CE. Nilotinib-induced autophagic changes increase endogenous parkin level and ubiquitination, leading to amyloid clearance. J Mol Med (Berl) 2014;92:373–386.
    1. Kantarjian HM, Giles F, Gattermann N, Bhalla K, Alimena G, et al. Nilotinib (formerly AMN107), a highly selective BCR-ABL tyrosine kinase inhibitor, is effective in patients with Philadelphia chromosome-positive chronic myelogenous leukemia in chronic phase following imatinib resistance and intolerance. Blood. 2007;110:3540–3546.
    1. Musumeci F, Schenone S, Brullo C, Botta M. An update on dual Src/Abl inhibitors. Future Med Chem. 2012;4:799–822.
    1. Giasson BI, Duda JE, Quinn SM, Zhang B, Trojanowski JQ, et al. Neuronal alpha-synucleinopathy with severe movement disorder in mice expressing A53T human alpha-synuclein. Neuron. 2002;34:521–533.
    1. Ihle JN, Pepersack L, Rebar L. Regulation of T cell differentiation: in vitro induction of 20 alpha-hydroxysteroid dehydrogenase in splenic lymphocytes from athymic mice by a unique lymphokine. J Immunol. 1981;126:2184–2189.
    1. Naylor SL, Sakaguchi AY, Shows TB, Law ML, Goeddel DV, et al. Human immune interferon gene is located on chromosome 12. J Exp Med. 1983;157:1020–1027.
    1. Schoenborn JR, Wilson CB. Regulation of interferon-gamma during innate and adaptive immune responses. Adv Immunol. 2007;96:41–101.
    1. Shin YJ, Choi JS, Choi JY, Hou Y, Cha JH, et al. Induction of vascular endothelial growth factor receptor-3 mRNA in glial cells following focal cerebral ischemia in rats. J Neuroimmunol. 2010;229:81–90.
    1. Rosenfeld PJ, Brown DM, Heier JS, Boyer DS, Kaiser PK, et al. Ranibizumab for neovascular age-related macular degeneration. N Engl J Med. 2006;355:1419–1431.
    1. Machein MR, Plate KH. Role of VEGF in developmental angiogenesis and in tumor angiogenesis in the brain. Cancer Treat Res. 2004;117:191–218.
    1. Storkebaum E, Lambrechts D, Carmeliet P. VEGF: once regarded as a specific angiogenic factor, now implicated in neuroprotection. Bioessays. 2004;26:943–954.
    1. Lambrechts D, Carmeliet P. VEGF at the neurovascular interface: therapeutic implications for motor neuron disease. Biochim Biophys Acta. 2006;1762:1109–1121.
    1. Carr MW, Roth SJ, Luther E, Rose SS, Springer TA. Monocyte chemoattractant protein 1 acts as a T-lymphocyte chemoattractant. Proc Natl Acad Sci U S A. 1994;91:3652–3656.
    1. Xu LL, Warren MK, Rose WL, Gong W, Wang JM. Human recombinant monocyte chemotactic protein and other C-C chemokines bind and induce directional migration of dendritic cells in vitro. J Leukoc Biol. 1996;60:365–371.
    1. Maghazachi AA, Al-Aoukaty A, Schall TJ. CC chemokines induce the generation of killer cells from CD56+ cells. Eur J Immunol. 1996;26:315–319.
    1. Vangelista L, Secchi M, Liu X, Bachi A, Jia L, et al. Engineering of Lactobacillus jensenii to secrete RANTES and a CCR5 antagonist analogue as live HIV-1 blockers. Antimicrob Agents Chemother. 2010;54:2994–3001.
    1. Pan Y, Lloyd C, Zhou H, Dolich S, Deeds J, et al. Neurotactin, a membrane-anchored chemokine upregulated in brain inflammation. Nature. 1997;387:611–617.
    1. Pabon MM, Bachstetter AD, Hudson CE, Gemma C, Bickford PC. CX3CL1 reduces neurotoxicity and microglial activation in a rat model of Parkinson’s disease. J Neuroinflammation. 2011;8:9.
    1. Shan S, Hong-Min T, Yi F, Jun-Peng G, Yue F, et al. New evidences for fractalkine/CX3CL1 involved in substantia nigral microglial activation and behavioral changes in a rat model of Parkinson’s disease. Neurobiol Aging. 2011;32:443–458.
    1. Lee S, Varvel NH, Konerth ME, Xu G, Cardona AE, et al. CX3CR1 deficiency alters microglial activation and reduces beta-amyloid deposition in two Alzheimer’s disease mouse models. Am J Pathol. 2010;177:2549–2562.
    1. Fuhrmann M, Bittner T, Jung CK, Burgold S, Page RM, et al. Microglial Cx3cr1 knockout prevents neuron loss in a mouse model of Alzheimer’s disease. Nat Neurosci. 2010;13:411–413.
    1. Apte SH, Baz A, Groves P, Kelso A, Kienzle N. Interferon-gamma and interleukin-4 reciprocally regulate CD8 expression in CD8+ T cells. Proc Natl Acad Sci U S A. 2008;105:17475–17480.
    1. Sokol CL, Barton GM, Farr AG, Medzhitov R. A mechanism for the initiation of allergen-induced T helper type 2 responses. Nat Immunol. 2008;9:310–318.
    1. Maes T, Joos GF, Brusselle GG. Targeting interleukin-4 in asthma: lost in translation? Am J Respir Cell Mol Biol. 2012;47:261–270.
    1. Chatila TA. Interleukin-4 receptor signaling pathways in asthma pathogenesis. Trends Mol Med. 2004;10:493–499.
    1. Howard M, Paul WE. Interleukins for B lymphocytes. Lymphokine Res. 1982;1:1–4.
    1. Yokota T, Otsuka T, Mosmann T, Banchereau J, DeFrance T, et al. Isolation and characterization of a human interleukin cDNA clone, homologous to mouse B-cell stimulatory factor, that expresses B-cell-and T-cell-stimulating activities. Proc Natl Acad Sci U S A. 1986;83:5894–5898.
    1. Marone G, Florio G, Petraroli A, de Paulis A. Dysregulation of the IgE/Fc epsilon RI network in HIV-1 infection. J Allergy Clin Immunol. 2001;107:22–30.
    1. Filipov NM, Seegal RF, Lawrence DA. Manganese potentiates in vitro production of proinflammatory cytokines and nitric oxide by microglia through a nuclear factor kappa B-dependent mechanism. Toxicol Sci. 2005;84:139–148.
    1. Lumeng CN, Bodzin JL, Saltiel AR. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J Clin Invest. 2007;117:175–184.
    1. Yang YC, Ciarletta AB, Temple PA, Chung MP, Kovacic S, et al. Human IL-3 (multi-CSF): identification by expression cloning of a novel hematopoietic growth factor related to murine IL-3. Cell. 1986;47:3–10.
    1. Obituaries: Mrs Annie Isabella Anslow, Eileen Georgina Sherrard. N Z Nurs J. 1979;72:32–33. No authors listed.
    1. Cantrell DA, Smith KA. The interleukin-2 T-cell system: a new cell growth model. Science. 1984;224:1312–1316.
    1. Smith KA. Interleukin-2: inception, impact, and implications. Science. 1988;240:1169–1176.
    1. Stern JB, Smith KA. Interleukin-2 induction of T-cell G1 progression and c-myb expression. Science. 1986;233:203–206.
    1. Beadling C, Johnson KW, Smith KA. Isolation of interleukin 2-induced immediate-early genes. Proc Natl Acad Sci U S A. 1993;90:2719–2723.
    1. Beadling C, Smith KA. DNA array analysis of interleukin-2-regulated immediate/early genes. Med Immunol. 2002;1:2.
    1. Gerard C, Rollins BJ. Chemokines and disease. Nat Immunol. 2001;2:108–115.
    1. Banisadr G, Gosselin RD, Mechighel P, Kitabgi P, Rostene W, et al. Highly regionalized neuronal expression of monocyte chemoattractant protein-1 (MCP-1/CCL2) in rat brain: evidence for its colocalization with neurotransmitters and neuropeptides. J Comp Neurol. 2005;489:275–292.
    1. Foresti ML, Arisi GM, Katki K, Montanez A, Sanchez RM, et al. Chemokine CCL2 and its receptor CCR2 are increased in the hippocampus following pilocarpine-induced status epilepticus. J Neuroinflammation. 2009;6:40.
    1. Fabene PF, Bramanti P, Constantin G. The emerging role for chemokines in epilepsy. J Neuroimmunol. 2010;224:22–27.
    1. Kim JS, Gautam SC, Chopp M, Zaloga C, Jones ML, et al. Expression of monocyte chemoattractant protein-1 and macrophage inflammatory protein-1 after focal cerebral ischemia in the rat. J Neuroimmunol. 1995;56:127–134.
    1. Hickman SE, El Khoury J. Mechanisms of mononuclear phagocyte recruitment in Alzheimer’s disease. CNS Neurol Disord Drug Targets. 2010;9:168–173.
    1. Ransohoff RM, Hamilton TA, Tani M, Stoler MH, Shick HE, et al. Astrocyte expression of mRNA encoding cytokines IP-10 and JE/MCP-1 in experimental autoimmune encephalomyelitis. FASEB J. 1993;7:592–600.
    1. Semple BD, Bye N, Rancan M, Ziebell JM, Morganti-Kossmann MC. Role of CCL2 (MCP-1) in traumatic brain injury (TBI): evidence from severe TBI patients and CCL2−/−mice. J Cereb Blood Flow Metab. 2010;30:769–782.
    1. Cocchi F, DeVico AL, Garzino-Demo A, Arya SK, Gallo RC, et al. Identification of RANTES, MIP-1 alpha, and MIP-1 beta as the major HIV-suppressive factors produced by CD8+ T cells. Science. 1995;270:1811–1815.
    1. Struyf S, Menten P, Lenaerts JP, Put W, D’Haese A, et al. Diverging binding capacities of natural LD78beta isoforms of macrophage inflammatory protein-1alpha to the CC chemokine receptors, 3 and 5 affect their anti-HIV-1 activity and chemotactic potencies for neutrophils and eosinophils. Eur J Immunol. 2001;31:2170–2178.
    1. Schall TJ, Jongstra J, Dyer BJ, Jorgensen J, Clayberger C, et al. A human T cell-specific molecule is a member of a new gene family. J Immunol. 1988;141:1018–1025.
    1. Song A, Chen YF, Thamatrakoln K, Storm TA, Krensky AM. RFLAT-1: a new zinc finger transcription factor that activates RANTES gene expression in T lymphocytes. Immunity. 1999;10:93–103.
    1. Song A, Nikolcheva T, Krensky AM. Transcriptional regulation of RANTES expression in T lymphocytes. Immunol Rev. 2000;177:236–245.
    1. Hebron ML, Algarzae NK, Lonskaya I2, Moussa C3. Fractalkine signaling and Tau hyper-phosphorylation are associated with autophagic alterations in lentiviral Tau and AI21–42 gene transfer models. Exp Neurol. 2014;251:127–138.
    1. Harrison JK, Jiang Y, Chen S, Xia Y, Maciejewski D, et al. Role for neuronally derived fractalkine in mediating interactions between neurons and CX3CR1-expressing microglia. Proc Natl Acad Sci U S A. 1998;95:10896–10901.
    1. Hatori K, Nagai A, Heisel R, Ryu JK, Kim SU. Fractalkine and fractalkine receptors in human neurons and glial cells. J Neurosci Res. 2002;69:418–426.
    1. Imai T, Hieshima K, Haskell C, Baba M, Nagira M, et al. Identification and molecular characterization of fractalkine receptor CX3CR, which mediates both leukocyte migration and adhesion. Cell. 1997;91:521–530.
    1. Bazan JF, Bacon KB, Hardiman G, Wang W, Soo K, et al. A new class of membrane-bound chemokine with a CX3C motif. Nature. 1997;385:640–644.
    1. Yoneda O, Imai T, Goda S, Inoue H, Yamauchi A, et al. Fractalkine-mediated endothelial cell injury by NK cells. J Immunol. 2000;164:4055–4062.
    1. Tong N, Perry SW, Zhang Q, James HJ, Guo H, et al. Neuronal fractalkine expression in HIV-1 encephalitis: roles for macrophage recruitment and neuroprotection in the central nervous system. J Immunol. 2000;164:1333–1339.
    1. Bhaskar K, Konerth M, Kokiko-Cochran ON, Cardona A, Ransohoff RM, et al. Regulation of tau pathology by the microglial fractalkine receptor. Neuron. 2010;68:19–31.
    1. Cardona AE, Sasse ME, Liu L, Cardona SM, Mizutani M, et al. Scavenging roles of chemokine receptors: chemokine receptor deficiency is associated with increased levels of ligand in circulation and tissues. Blood. 2008;112:256–263.
    1. Liu Z, Condello C, Schain A, Harb R, Grutzendler J. CX3CR1 in microglia regulates brain amyloid deposition through selective protofibrillar amyloid-I2 phagocytosis. J Neurosci. 2010;30:17091–17101.
    1. Cho SH, Sun B, Zhou Y, Kauppinen TM, Halabisky B, et al. CX3CR1 protein signaling modulates microglial activation and protects against plaque-independent cognitive deficits in a mouse model of Alzheimer disease. J Biol Chem. 2011;286:32713–32722.
    1. Meucci O, Fatatis A, Simen AA, Bushell TJ, Gray PW, et al. Chemokines regulate hippocampal neuronal signaling and gp120 neurotoxicity. Proc Natl Acad Sci U S A. 1998;95:14500–14505.
    1. Mizuno T, Kawanokuchi J, Numata K, Suzumura A. Production and neuroprotective functions of fractalkine in the central nervous system. Brain Res. 2003;979:65–70.
    1. Cardona AE, Pioro EP, Sasse ME, Kostenko V, Cardona SM, et al. Control of microglial neurotoxicity by the fractalkine receptor. Nat Neurosci. 2006;9:917–924.
    1. Denes A, Ferenczi S, Halasz J, Kornyei Z, Kovacs KJ. Role of CX3CR1 (fractalkine receptor) in brain damage and inflammation induced by focal cerebral ischemia in mouse. J Cereb Blood Flow Metab. 2008;28:1707–1721.
    1. Kastenbauer S, Koedel U, Wick M, Kieseier BC, Hartung HP, et al. CSF and serum levels of soluble fractalkine (CX3CL1) in inflammatory diseases of the nervous system. J Neuroimmunol. 2003;137:210–217.
    1. Rancan M, Bye N, Otto VI, Trentz O, Kossmann T, et al. The chemokine fractalkine in patients with severe traumatic brain injury and a mouse model of closed head injury. J Cereb Blood Flow Metab. 2004;24:1110–1118.
    1. Sporer B, Kastenbauer S, Koedel U, Arendt G, Pfister HW. Increased intrathecal release of soluble fractalkine in HIV-infected patients. AIDS Res Hum Retroviruses. 2003;19:111–116.

Source: PubMed

3
Abonneren