Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2018

Open Access 01-12-2018 | Research

Longitudinal chemokine profile expression in a blood-brain barrier model from Alzheimer transgenic versus wild-type mice

Authors: J. Vérité, T. Janet, D. Chassaing, B. Fauconneau, H. Rabeony, G. Page

Published in: Journal of Neuroinflammation | Issue 1/2018

Login to get access

Abstract

Background

Alzheimer’s disease is widely described since the discovery of histopathological lesions in Mrs. Auguste Deter in 1906. However to date, there is no effective treatment to deal with the many cellular and molecular alterations. The complexity is even higher with the growing evidence of involvement of the peripheral blood mononuclear cells (PBMCs). Indeed, monocytes and T cells are shown in the cerebral parenchyma of AD patients, and these cells grafted to the periphery are able to go through the blood-brain barrier (BBB) in transgenic mouse models. It is known that BBB is disrupted at a late stage of AD. Chemokines represent major regulators of the transmigration of PBMCs, but many data were obtained on AD animal models. No data are available on the role of AD BBB in a healthy brain parenchyma. Therefore, the purpose of this study was to analyze the longitudinal chemokine profile expression in a BBB model from AD transgenic mice versus wild-type (WT) mice.

Methods

A primary mouse BBB model was used with a luminal compartment either AD or WT and an abluminal compartment WT consisting of astrocytes and microglia. PBMCs were extracted by a ficoll gradient and incubated in the transwell with a direct contact with the luminal side, including the endothelial cells and pericytes. Then, the complete BBB model was incubated during 48 h, before supernatants and cell lysates were collected. Chemokines were quantified by X-MAP® luminex technology.

Results

Abluminal CX3CL1 production increased in 12-month-old AD BBB while CX3CL1 levels decreased in luminal lysates. CCL3 in luminal compartment increased with aging and was significantly different compared to AD BBB at 12 months. In addition, abluminal CCL2 in 12-month-old AD BBB greatly decreased compared to levels in WT BBB. On the contrary, no modification was observed for CCL4, CCL5, and CXCL10.

Conclusion

These first findings highlighted the impact of AD luminal compartment on chemokine signature in a healthy brain parenchyma, suggesting new therapeutic or diagnostic approaches.
Appendix
Available only for authorised users
Literature
3.
5.
go back to reference Goossens J, Bjerke M, Struyfs H, Niemantsverdriet E, Somers C, Van den Bossche T, Van Mossevelde S, De Vil B, Sieben A, Martin JJ, et al. No added diagnostic value of non-phosphorylated tau fraction (p-taurel) in CSF as a biomarker for differential dementia diagnosis. Alzheimers Res Ther. 2017;9:49.CrossRefPubMedPubMedCentral Goossens J, Bjerke M, Struyfs H, Niemantsverdriet E, Somers C, Van den Bossche T, Van Mossevelde S, De Vil B, Sieben A, Martin JJ, et al. No added diagnostic value of non-phosphorylated tau fraction (p-taurel) in CSF as a biomarker for differential dementia diagnosis. Alzheimers Res Ther. 2017;9:49.CrossRefPubMedPubMedCentral
9.
go back to reference Reinhold AK, Rittner HL. Barrier function in the peripheral and central nervous system—a review. Pflugers Arch. 2017;469:123–34.CrossRefPubMed Reinhold AK, Rittner HL. Barrier function in the peripheral and central nervous system—a review. Pflugers Arch. 2017;469:123–34.CrossRefPubMed
10.
go back to reference Mietelska-Porowska A, Wojda U. T lymphocytes and inflammatory mediators in the interplay between brain and blood in Alzheimer’s disease: potential pools of new biomarkers. J Immunol Res. 2017;2017:4626540.CrossRefPubMedPubMedCentral Mietelska-Porowska A, Wojda U. T lymphocytes and inflammatory mediators in the interplay between brain and blood in Alzheimer’s disease: potential pools of new biomarkers. J Immunol Res. 2017;2017:4626540.CrossRefPubMedPubMedCentral
11.
go back to reference Wang J, Gu BJ, Masters CL, Wang YJ. A systemic view of Alzheimer disease—insights from amyloid-beta metabolism beyond the brain. Nat Rev Neurol. 2017;13:612–23.CrossRefPubMed Wang J, Gu BJ, Masters CL, Wang YJ. A systemic view of Alzheimer disease—insights from amyloid-beta metabolism beyond the brain. Nat Rev Neurol. 2017;13:612–23.CrossRefPubMed
12.
go back to reference Gosselet F, Saint-Pol J, Candela P, Fenart L. Amyloid-beta peptides, Alzheimer’s disease and the blood-brain barrier. Curr Alzheimer Res. 2013;10:1015–33.CrossRefPubMed Gosselet F, Saint-Pol J, Candela P, Fenart L. Amyloid-beta peptides, Alzheimer’s disease and the blood-brain barrier. Curr Alzheimer Res. 2013;10:1015–33.CrossRefPubMed
13.
go back to reference Da Mesquita S, Ferreira AC, Sousa JC, Correia-Neves M, Sousa N, Marques F. Insights on the pathophysiology of Alzheimer’s disease: the crosstalk between amyloid pathology, neuroinflammation and the peripheral immune system. Neurosci Biobehav Rev. 2016;68:547–62.CrossRefPubMed Da Mesquita S, Ferreira AC, Sousa JC, Correia-Neves M, Sousa N, Marques F. Insights on the pathophysiology of Alzheimer’s disease: the crosstalk between amyloid pathology, neuroinflammation and the peripheral immune system. Neurosci Biobehav Rev. 2016;68:547–62.CrossRefPubMed
14.
go back to reference Kisler K, Nelson AR, Montagne A, Zlokovic BV. Cerebral blood flow regulation and neurovascular dysfunction in Alzheimer disease. Nat Rev Neurosci. 2017;18:419–34.CrossRefPubMedPubMedCentral Kisler K, Nelson AR, Montagne A, Zlokovic BV. Cerebral blood flow regulation and neurovascular dysfunction in Alzheimer disease. Nat Rev Neurosci. 2017;18:419–34.CrossRefPubMedPubMedCentral
15.
go back to reference Di Marco LY, Venneri A, Farkas E, Evans PC, Marzo A, Frangi AF. Vascular dysfunction in the pathogenesis of Alzheimer’s disease—a review of endothelium-mediated mechanisms and ensuing vicious circles. Neurobiol Dis. 2015;82:593–606.CrossRefPubMed Di Marco LY, Venneri A, Farkas E, Evans PC, Marzo A, Frangi AF. Vascular dysfunction in the pathogenesis of Alzheimer’s disease—a review of endothelium-mediated mechanisms and ensuing vicious circles. Neurobiol Dis. 2015;82:593–606.CrossRefPubMed
16.
go back to reference Giri R, Selvaraj S, Miller CA, Hofman F, Yan SD, Stern D, Zlokovic BV, Kalra VK. Effect of endothelial cell polarity on beta-amyloid-induced migration of monocytes across normal and AD endothelium. Am J Physiol Cell Physiol. 2002;283:C895–904.CrossRefPubMed Giri R, Selvaraj S, Miller CA, Hofman F, Yan SD, Stern D, Zlokovic BV, Kalra VK. Effect of endothelial cell polarity on beta-amyloid-induced migration of monocytes across normal and AD endothelium. Am J Physiol Cell Physiol. 2002;283:C895–904.CrossRefPubMed
17.
go back to reference Gonzalez-Velasquez F, Reed JW, Fuseler JW, Matherly EE, Kotarek JA, Soto-Ortega DD, Moss MA. Activation of brain endothelium by soluble aggregates of the amyloid-beta protein involves nuclear factor-kappaB. Curr Alzheimer Res. 2011;8:81–94.CrossRefPubMed Gonzalez-Velasquez F, Reed JW, Fuseler JW, Matherly EE, Kotarek JA, Soto-Ortega DD, Moss MA. Activation of brain endothelium by soluble aggregates of the amyloid-beta protein involves nuclear factor-kappaB. Curr Alzheimer Res. 2011;8:81–94.CrossRefPubMed
18.
go back to reference Naert G, Rivest S. A deficiency in CCR2+ monocytes: the hidden side of Alzheimer’s disease. J Mol Cell Biol. 2013;5:284–93.CrossRefPubMed Naert G, Rivest S. A deficiency in CCR2+ monocytes: the hidden side of Alzheimer’s disease. J Mol Cell Biol. 2013;5:284–93.CrossRefPubMed
19.
go back to reference El Khoury J, Toft M, Hickman SE, Means TK, Terada K, Geula C, Luster AD. Ccr2 deficiency impairs microglial accumulation and accelerates progression of Alzheimer-like disease. Nat Med. 2007;13:432–8.CrossRefPubMed El Khoury J, Toft M, Hickman SE, Means TK, Terada K, Geula C, Luster AD. Ccr2 deficiency impairs microglial accumulation and accelerates progression of Alzheimer-like disease. Nat Med. 2007;13:432–8.CrossRefPubMed
21.
go back to reference Togo T, Akiyama H, Iseki E, Kondo H, Ikeda K, Kato M, Oda T, Tsuchiya K, Kosaka K. Occurrence of T cells in the brain of Alzheimer’s disease and other neurological diseases. J Neuroimmunol. 2002;124:83–92.CrossRefPubMed Togo T, Akiyama H, Iseki E, Kondo H, Ikeda K, Kato M, Oda T, Tsuchiya K, Kosaka K. Occurrence of T cells in the brain of Alzheimer’s disease and other neurological diseases. J Neuroimmunol. 2002;124:83–92.CrossRefPubMed
22.
go back to reference Town T, Tan J, Flavell RA, Mullan M. T-cells in Alzheimer’s disease. NeuroMolecular Med. 2005;7:255–64.CrossRefPubMed Town T, Tan J, Flavell RA, Mullan M. T-cells in Alzheimer’s disease. NeuroMolecular Med. 2005;7:255–64.CrossRefPubMed
23.
go back to reference Rogers J, Luber-Narod J, Styren SD, Civin WH. Expression of immune system-associated antigens by cells of the human central nervous system: relationship to the pathology of Alzheimer’s disease. Neurobiol Aging. 1988;9:339–49.CrossRefPubMed Rogers J, Luber-Narod J, Styren SD, Civin WH. Expression of immune system-associated antigens by cells of the human central nervous system: relationship to the pathology of Alzheimer’s disease. Neurobiol Aging. 1988;9:339–49.CrossRefPubMed
24.
go back to reference Jevtic S, Sengar AS, Salter MW, McLaurin J. The role of the immune system in Alzheimer disease: etiology and treatment. Ageing Res Rev. 2017;40:84–94.CrossRefPubMed Jevtic S, Sengar AS, Salter MW, McLaurin J. The role of the immune system in Alzheimer disease: etiology and treatment. Ageing Res Rev. 2017;40:84–94.CrossRefPubMed
26.
go back to reference Ulrich JD, Ulland TK, Colonna M, Holtzman DM. Elucidating the role of TREM2 in Alzheimer’s disease. Neuron. 2017;94:237–48.CrossRefPubMed Ulrich JD, Ulland TK, Colonna M, Holtzman DM. Elucidating the role of TREM2 in Alzheimer’s disease. Neuron. 2017;94:237–48.CrossRefPubMed
27.
go back to reference Jay TR, Hirsch AM, Broihier ML, Miller CM, Neilson LE, Ransohoff RM, Lamb BT, Landreth GE. Disease progression-dependent effects of TREM2 deficiency in a mouse model of Alzheimer’s disease. J Neurosci. 2017;37:637–47.CrossRefPubMedPubMedCentral Jay TR, Hirsch AM, Broihier ML, Miller CM, Neilson LE, Ransohoff RM, Lamb BT, Landreth GE. Disease progression-dependent effects of TREM2 deficiency in a mouse model of Alzheimer’s disease. J Neurosci. 2017;37:637–47.CrossRefPubMedPubMedCentral
28.
go back to reference Dansokho C, Ait Ahmed D, Aid S, Toly-Ndour C, Chaigneau T, Calle V, Cagnard N, Holzenberger M, Piaggio E, Aucouturier P, Dorothee G. Regulatory T cells delay disease progression in Alzheimer-like pathology. Brain. 2016;139:1237–51.CrossRefPubMed Dansokho C, Ait Ahmed D, Aid S, Toly-Ndour C, Chaigneau T, Calle V, Cagnard N, Holzenberger M, Piaggio E, Aucouturier P, Dorothee G. Regulatory T cells delay disease progression in Alzheimer-like pathology. Brain. 2016;139:1237–51.CrossRefPubMed
29.
go back to reference Montagne A, Nation DA, Pa J, Sweeney MD, Toga AW, Zlokovic BV. Brain imaging of neurovascular dysfunction in Alzheimer’s disease. Acta Neuropathol. 2016;131:687–707.CrossRefPubMedPubMedCentral Montagne A, Nation DA, Pa J, Sweeney MD, Toga AW, Zlokovic BV. Brain imaging of neurovascular dysfunction in Alzheimer’s disease. Acta Neuropathol. 2016;131:687–707.CrossRefPubMedPubMedCentral
30.
go back to reference Le Thuc O, Blondeau N, Nahon JL, Rovere C. The complex contribution of chemokines to neuroinflammation: switching from beneficial to detrimental effects. Ann N Y Acad Sci. 2015;1351:127–40.CrossRefPubMed Le Thuc O, Blondeau N, Nahon JL, Rovere C. The complex contribution of chemokines to neuroinflammation: switching from beneficial to detrimental effects. Ann N Y Acad Sci. 2015;1351:127–40.CrossRefPubMed
31.
go back to reference Gualtierotti R, Guarnaccia L, Beretta M, Navone SE, Campanella R, Riboni L, Rampini P, Marfia G. Modulation of neuroinflammation in the central nervous system: role of chemokines and sphingolipids. Adv Ther. 2017;34:396–420.CrossRefPubMed Gualtierotti R, Guarnaccia L, Beretta M, Navone SE, Campanella R, Riboni L, Rampini P, Marfia G. Modulation of neuroinflammation in the central nervous system: role of chemokines and sphingolipids. Adv Ther. 2017;34:396–420.CrossRefPubMed
32.
go back to reference Rostene W, Dansereau MA, Godefroy D, Van Steenwinckel J, Reaux-Le Goazigo A, Melik-Parsadaniantz S, Apartis E, Hunot S, Beaudet N, Sarret P. Neurochemokines: a menage a trois providing new insights on the functions of chemokines in the central nervous system. J Neurochem. 2011;118:680–94.CrossRefPubMed Rostene W, Dansereau MA, Godefroy D, Van Steenwinckel J, Reaux-Le Goazigo A, Melik-Parsadaniantz S, Apartis E, Hunot S, Beaudet N, Sarret P. Neurochemokines: a menage a trois providing new insights on the functions of chemokines in the central nervous system. J Neurochem. 2011;118:680–94.CrossRefPubMed
33.
go back to reference Ruan L, Kong Y, Wang JM, Le Y. Chemoattractants and receptors in Alzheimer’s disease. Front Biosci (Schol Ed). 2010;2:504-14. Ruan L, Kong Y, Wang JM, Le Y. Chemoattractants and receptors in Alzheimer’s disease. Front Biosci (Schol Ed). 2010;2:504-14.
35.
go back to reference Liu C, Cui G, Zhu M, Kang X, Guo H. Neuroinflammation in Alzheimer’s disease: chemokines produced by astrocytes and chemokine receptors. Int J Clin Exp Pathol. 2014;7:8342–55.PubMedPubMedCentral Liu C, Cui G, Zhu M, Kang X, Guo H. Neuroinflammation in Alzheimer’s disease: chemokines produced by astrocytes and chemokine receptors. Int J Clin Exp Pathol. 2014;7:8342–55.PubMedPubMedCentral
36.
go back to reference El Khoury JB, Moore KJ, Means TK, Leung J, Terada K, Toft M, Freeman MW, Luster AD. CD36 mediates the innate host response to beta-amyloid. J Exp Med. 2003;197:1657–66.CrossRefPubMedPubMedCentral El Khoury JB, Moore KJ, Means TK, Leung J, Terada K, Toft M, Freeman MW, Luster AD. CD36 mediates the innate host response to beta-amyloid. J Exp Med. 2003;197:1657–66.CrossRefPubMedPubMedCentral
37.
go back to reference Goldeck D, Witkowski JM, Fulop T, Pawelec G. Peripheral immune signatures in Alzheimer disease. Curr Alzheimer Res. 2016;13:739–49.CrossRefPubMed Goldeck D, Witkowski JM, Fulop T, Pawelec G. Peripheral immune signatures in Alzheimer disease. Curr Alzheimer Res. 2016;13:739–49.CrossRefPubMed
38.
go back to reference Liu YJ, Guo DW, Tian L, Shang DS, Zhao WD, Li B, Fang WG, Zhu L, Chen YH. Peripheral T cells derived from Alzheimer’s disease patients overexpress CXCR2 contributing to its transendothelial migration, which is microglial TNF-alpha-dependent. Neurobiol Aging. 2010;31:175–88.CrossRefPubMed Liu YJ, Guo DW, Tian L, Shang DS, Zhao WD, Li B, Fang WG, Zhu L, Chen YH. Peripheral T cells derived from Alzheimer’s disease patients overexpress CXCR2 contributing to its transendothelial migration, which is microglial TNF-alpha-dependent. Neurobiol Aging. 2010;31:175–88.CrossRefPubMed
39.
go back to reference Lee YK, Kwak DH, Oh KW, Nam SY, Lee BJ, Yun YW, Kim YB, Han SB, Hong JT. CCR5 deficiency induces astrocyte activation, Abeta deposit and impaired memory function. Neurobiol Learn Mem. 2009;92:356–63.CrossRefPubMed Lee YK, Kwak DH, Oh KW, Nam SY, Lee BJ, Yun YW, Kim YB, Han SB, Hong JT. CCR5 deficiency induces astrocyte activation, Abeta deposit and impaired memory function. Neurobiol Learn Mem. 2009;92:356–63.CrossRefPubMed
40.
go back to reference Kiyota T, Yamamoto M, Schroder B, Jacobsen MT, Swan RJ, Lambert MP, Klein WL, Gendelman HE, Ransohoff RM, Ikezu T. AAV1/2-mediated CNS gene delivery of dominant-negative CCL2 mutant suppresses gliosis, beta-amyloidosis, and learning impairment of APP/PS1 mice. Mol Ther. 2009;17:803–9.CrossRefPubMedPubMedCentral Kiyota T, Yamamoto M, Schroder B, Jacobsen MT, Swan RJ, Lambert MP, Klein WL, Gendelman HE, Ransohoff RM, Ikezu T. AAV1/2-mediated CNS gene delivery of dominant-negative CCL2 mutant suppresses gliosis, beta-amyloidosis, and learning impairment of APP/PS1 mice. Mol Ther. 2009;17:803–9.CrossRefPubMedPubMedCentral
41.
go back to reference Fuhrmann M, Bittner T, Jung CK, Burgold S, Page RM, Mitteregger G, Haass C, LaFerla FM, Kretzschmar H, Herms J. Microglial Cx3cr1 knockout prevents neuron loss in a mouse model of Alzheimer’s disease. Nat Neurosci. 2010;13:411–3.CrossRefPubMedPubMedCentral Fuhrmann M, Bittner T, Jung CK, Burgold S, Page RM, Mitteregger G, Haass C, LaFerla FM, Kretzschmar H, Herms J. Microglial Cx3cr1 knockout prevents neuron loss in a mouse model of Alzheimer’s disease. Nat Neurosci. 2010;13:411–3.CrossRefPubMedPubMedCentral
42.
go back to reference Martin E, Boucher C, Fontaine B, Delarasse C. Distinct inflammatory phenotypes of microglia and monocyte-derived macrophages in Alzheimer’s disease models: effects of aging and amyloid pathology. Aging Cell. 2017;16:27–38.CrossRefPubMed Martin E, Boucher C, Fontaine B, Delarasse C. Distinct inflammatory phenotypes of microglia and monocyte-derived macrophages in Alzheimer’s disease models: effects of aging and amyloid pathology. Aging Cell. 2017;16:27–38.CrossRefPubMed
43.
go back to reference Couturier J, Paccalin M, Morel M, Terro F, Milin S, Pontcharraud R, Fauconneau B, Page G. Prevention of the beta-amyloid peptide-induced inflammatory process by inhibition of double-stranded RNA-dependent protein kinase in primary murine mixed co-cultures. J Neuroinflammation. 2011;8:72.CrossRefPubMedPubMedCentral Couturier J, Paccalin M, Morel M, Terro F, Milin S, Pontcharraud R, Fauconneau B, Page G. Prevention of the beta-amyloid peptide-induced inflammatory process by inhibition of double-stranded RNA-dependent protein kinase in primary murine mixed co-cultures. J Neuroinflammation. 2011;8:72.CrossRefPubMedPubMedCentral
44.
go back to reference Francois A, Terro F, Janet T, Rioux Bilan A, Paccalin M, Page G. Involvement of interleukin-1beta in the autophagic process of microglia: relevance to Alzheimer’s disease. J Neuroinflammation. 2013;10:151.CrossRefPubMedPubMedCentral Francois A, Terro F, Janet T, Rioux Bilan A, Paccalin M, Page G. Involvement of interleukin-1beta in the autophagic process of microglia: relevance to Alzheimer’s disease. J Neuroinflammation. 2013;10:151.CrossRefPubMedPubMedCentral
45.
go back to reference Ruan L, Kang Z, Pei G, Le Y. Amyloid deposition and inflammation in APPswe/PS1dE9 mouse model of Alzheimer’s disease. Curr Alzheimer Res. 2009;6:531–40.CrossRefPubMed Ruan L, Kang Z, Pei G, Le Y. Amyloid deposition and inflammation in APPswe/PS1dE9 mouse model of Alzheimer’s disease. Curr Alzheimer Res. 2009;6:531–40.CrossRefPubMed
46.
go back to reference Couturier J, Paccalin M, Lafay-Chebassier C, Chalon S, Ingrand I, Pinguet J, Pontcharraud R, Guillard O, Fauconneau B, Page G. Pharmacological inhibition of PKR in APPswePS1dE9 mice transiently prevents inflammation at 12 months of age but increases Abeta42 levels in the late stages of the Alzheimer’s disease. Curr Alzheimer Res. 2012;9:344–60.CrossRefPubMed Couturier J, Paccalin M, Lafay-Chebassier C, Chalon S, Ingrand I, Pinguet J, Pontcharraud R, Guillard O, Fauconneau B, Page G. Pharmacological inhibition of PKR in APPswePS1dE9 mice transiently prevents inflammation at 12 months of age but increases Abeta42 levels in the late stages of the Alzheimer’s disease. Curr Alzheimer Res. 2012;9:344–60.CrossRefPubMed
48.
go back to reference Couturier J, Page G, Morel M, Gontier C, Claude J, Pontcharraud R, Fauconneau B, Paccalin M. Inhibition of double-stranded RNA-dependent protein kinase strongly decreases cytokine production and release in peripheral blood mononuclear cells from patients with Alzheimer’s disease. J Alzheimers Dis. 2010;21:1217–31.CrossRefPubMed Couturier J, Page G, Morel M, Gontier C, Claude J, Pontcharraud R, Fauconneau B, Paccalin M. Inhibition of double-stranded RNA-dependent protein kinase strongly decreases cytokine production and release in peripheral blood mononuclear cells from patients with Alzheimer’s disease. J Alzheimers Dis. 2010;21:1217–31.CrossRefPubMed
49.
go back to reference Francois A, Julian A, Ragot S, Dugast E, Blanchard L, Brishoual S, Chassaing D, Page G, Paccalin M. Inflammatory stress on autophagy in peripheral blood mononuclear cells from patients with Alzheimer’s disease during 24 months of follow-up. PLoS One. 2015;10:e0138326.CrossRefPubMedPubMedCentral Francois A, Julian A, Ragot S, Dugast E, Blanchard L, Brishoual S, Chassaing D, Page G, Paccalin M. Inflammatory stress on autophagy in peripheral blood mononuclear cells from patients with Alzheimer’s disease during 24 months of follow-up. PLoS One. 2015;10:e0138326.CrossRefPubMedPubMedCentral
50.
go back to reference Julian A, Dugast E, Ragot S, Krolak-Salmon P, Berrut G, Dantoine T, Hommet C, Hanon O, Page G, Paccalin M. There is no correlation between peripheral inflammation and cognitive status at diagnosis in Alzheimer’s disease. Aging Clin Exp Res. 2015;27:589–94.CrossRefPubMed Julian A, Dugast E, Ragot S, Krolak-Salmon P, Berrut G, Dantoine T, Hommet C, Hanon O, Page G, Paccalin M. There is no correlation between peripheral inflammation and cognitive status at diagnosis in Alzheimer’s disease. Aging Clin Exp Res. 2015;27:589–94.CrossRefPubMed
51.
go back to reference Iarlori C, Gambi D, Gambi F, Lucci I, Feliciani C, Salvatore M, Reale M. Expression and production of two selected beta-chemokines in peripheral blood mononuclear cells from patients with Alzheimer’s disease. Exp Gerontol. 2005;40:605–11.CrossRefPubMed Iarlori C, Gambi D, Gambi F, Lucci I, Feliciani C, Salvatore M, Reale M. Expression and production of two selected beta-chemokines in peripheral blood mononuclear cells from patients with Alzheimer’s disease. Exp Gerontol. 2005;40:605–11.CrossRefPubMed
52.
go back to reference Magaki S, Mueller C, Dickson C, Kirsch W. Increased production of inflammatory cytokines in mild cognitive impairment. Exp Gerontol. 2007;42:233–40.CrossRefPubMed Magaki S, Mueller C, Dickson C, Kirsch W. Increased production of inflammatory cytokines in mild cognitive impairment. Exp Gerontol. 2007;42:233–40.CrossRefPubMed
53.
go back to reference Vedin I, Cederholm T, Freund-Levi Y, Basun H, Hjorth E, Irving GF, Eriksdotter-Jonhagen M, Schultzberg M, Wahlund LO, Palmblad J. Reduced prostaglandin F2 alpha release from blood mononuclear leukocytes after oral supplementation of omega3 fatty acids: the OmegAD study. J Lipid Res. 2010;51:1179–85.CrossRefPubMedPubMedCentral Vedin I, Cederholm T, Freund-Levi Y, Basun H, Hjorth E, Irving GF, Eriksdotter-Jonhagen M, Schultzberg M, Wahlund LO, Palmblad J. Reduced prostaglandin F2 alpha release from blood mononuclear leukocytes after oral supplementation of omega3 fatty acids: the OmegAD study. J Lipid Res. 2010;51:1179–85.CrossRefPubMedPubMedCentral
54.
go back to reference Poujol F, Monneret G, Pachot A, Textoris J, Venet F. Altered T lymphocyte proliferation upon lipopolysaccharide challenge ex vivo. PLoS One. 2015;10:e0144375.CrossRefPubMedPubMedCentral Poujol F, Monneret G, Pachot A, Textoris J, Venet F. Altered T lymphocyte proliferation upon lipopolysaccharide challenge ex vivo. PLoS One. 2015;10:e0144375.CrossRefPubMedPubMedCentral
55.
go back to reference Rocha NP, Teixeira AL, Coelho FM, Caramelli P, Guimaraes HC, Barbosa IG, da Silva TA, Mukhamedyarov MA, Zefirov AL, Rizvanov AA, et al. Peripheral blood mono-nuclear cells derived from Alzheimer’s disease patients show elevated baseline levels of secreted cytokines but resist stimulation with beta-amyloid peptide. Mol Cell Neurosci. 2012;49:77–84.CrossRefPubMed Rocha NP, Teixeira AL, Coelho FM, Caramelli P, Guimaraes HC, Barbosa IG, da Silva TA, Mukhamedyarov MA, Zefirov AL, Rizvanov AA, et al. Peripheral blood mono-nuclear cells derived from Alzheimer’s disease patients show elevated baseline levels of secreted cytokines but resist stimulation with beta-amyloid peptide. Mol Cell Neurosci. 2012;49:77–84.CrossRefPubMed
56.
go back to reference Wilhelm I, Fazakas C, Krizbai IA. In vitro models of the blood-brain barrier. Acta Neurobiol Exp (Wars). 2011;71:113–28. Wilhelm I, Fazakas C, Krizbai IA. In vitro models of the blood-brain barrier. Acta Neurobiol Exp (Wars). 2011;71:113–28.
57.
go back to reference Cordon-Cardo C, O'Brien JP, Casals D, Rittman-Grauer L, Biedler JL, Melamed MR, Bertino JR. Multidrug-resistance gene (P-glycoprotein) is expressed by endothelial cells at blood-brain barrier sites. Proc Natl Acad Sci U S A. 1989;86:695–8.CrossRefPubMedPubMedCentral Cordon-Cardo C, O'Brien JP, Casals D, Rittman-Grauer L, Biedler JL, Melamed MR, Bertino JR. Multidrug-resistance gene (P-glycoprotein) is expressed by endothelial cells at blood-brain barrier sites. Proc Natl Acad Sci U S A. 1989;86:695–8.CrossRefPubMedPubMedCentral
59.
go back to reference Heppner FL, Ransohoff RM, Becher B. Immune attack: the role of inflammation in Alzheimer disease. Nat Rev Neurosci. 2015;16:358–72.CrossRefPubMed Heppner FL, Ransohoff RM, Becher B. Immune attack: the role of inflammation in Alzheimer disease. Nat Rev Neurosci. 2015;16:358–72.CrossRefPubMed
60.
go back to reference Simard AR, Soulet D, Gowing G, Julien JP, Rivest S. Bone marrow-derived microglia play a critical role in restricting senile plaque formation in Alzheimer’s disease. Neuron. 2006;49:489–502.CrossRefPubMed Simard AR, Soulet D, Gowing G, Julien JP, Rivest S. Bone marrow-derived microglia play a critical role in restricting senile plaque formation in Alzheimer’s disease. Neuron. 2006;49:489–502.CrossRefPubMed
61.
go back to reference Fisher Y, Nemirovsky A, Baron R, Monsonego A. T cells specifically targeted to amyloid plaques enhance plaque clearance in a mouse model of Alzheimer’s disease. PLoS One. 2010;5:e10830.CrossRefPubMedPubMedCentral Fisher Y, Nemirovsky A, Baron R, Monsonego A. T cells specifically targeted to amyloid plaques enhance plaque clearance in a mouse model of Alzheimer’s disease. PLoS One. 2010;5:e10830.CrossRefPubMedPubMedCentral
62.
go back to reference Rezai-Zadeh K, Gate D, Gowing G, Town T. How to get from here to there: macrophage recruitment in Alzheimer’s disease. Curr Alzheimer Res. 2011;8:156–63.CrossRefPubMedPubMedCentral Rezai-Zadeh K, Gate D, Gowing G, Town T. How to get from here to there: macrophage recruitment in Alzheimer’s disease. Curr Alzheimer Res. 2011;8:156–63.CrossRefPubMedPubMedCentral
63.
go back to reference Streit WJ, Braak H, Xue QS, Bechmann I. Dystrophic (senescent) rather than activated microglial cells are associated with tau pathology and likely precede neurodegeneration in Alzheimer’s disease. Acta Neuropathol. 2009;118:475–85.CrossRefPubMedPubMedCentral Streit WJ, Braak H, Xue QS, Bechmann I. Dystrophic (senescent) rather than activated microglial cells are associated with tau pathology and likely precede neurodegeneration in Alzheimer’s disease. Acta Neuropathol. 2009;118:475–85.CrossRefPubMedPubMedCentral
64.
go back to reference Jawa RS, Quist E, Boyer CW, Shostrom VK, Mercer DW. Mesenteric ischemia-reperfusion injury up-regulates certain CC, CXC, and XC chemokines and results in multi-organ injury in a time-dependent manner. Eur Cytokine Netw. 2013;24:148–56.PubMed Jawa RS, Quist E, Boyer CW, Shostrom VK, Mercer DW. Mesenteric ischemia-reperfusion injury up-regulates certain CC, CXC, and XC chemokines and results in multi-organ injury in a time-dependent manner. Eur Cytokine Netw. 2013;24:148–56.PubMed
65.
go back to reference Fahey S, Dempsey E, Long A. The role of chemokines in acute and chronic hepatitis C infection. Cell Mol Immunol. 2014;11:25–40.CrossRefPubMed Fahey S, Dempsey E, Long A. The role of chemokines in acute and chronic hepatitis C infection. Cell Mol Immunol. 2014;11:25–40.CrossRefPubMed
67.
go back to reference Goodall EF, Wang C, Simpson JE, Baker DJ, Drew DR, Heath PR, Saffrey MJ, Romero IA, Wharton SB. Age-associated changes in the blood-brain barrier: comparative studies in human and mouse. Neuropathol Appl Neurobiol. 2018;44(3):328-40. https://doi.org/10.1111/nan.12408. Epub 2017 May 29. Goodall EF, Wang C, Simpson JE, Baker DJ, Drew DR, Heath PR, Saffrey MJ, Romero IA, Wharton SB. Age-associated changes in the blood-brain barrier: comparative studies in human and mouse. Neuropathol Appl Neurobiol. 2018;44(3):328-40. https://​doi.​org/​10.​1111/​nan.​12408. Epub 2017 May 29.
68.
go back to reference Vellonen KS, Ihalainen J, Boucau MC, Gosselet F, Picardat T, Gynther M, Kanninen KM, White AR, Malm T, Koistinaho J, et al. Disease-induced alterations in brain drug transporters in animal models of Alzheimer’s disease : theme: drug discovery, development and delivery in Alzheimer’s disease guest editor: Davide Brambilla. Pharm Res. 2017;34:2652–62.CrossRefPubMed Vellonen KS, Ihalainen J, Boucau MC, Gosselet F, Picardat T, Gynther M, Kanninen KM, White AR, Malm T, Koistinaho J, et al. Disease-induced alterations in brain drug transporters in animal models of Alzheimer’s disease : theme: drug discovery, development and delivery in Alzheimer’s disease guest editor: Davide Brambilla. Pharm Res. 2017;34:2652–62.CrossRefPubMed
69.
go back to reference Bien-Ly N, Boswell CA, Jeet S, Beach TG, Hoyte K, Luk W, Shihadeh V, Ulufatu S, Foreman O, Lu Y, et al. Lack of widespread BBB disruption in Alzheimer’s disease models: focus on therapeutic antibodies. Neuron. 2015;88:289–97.CrossRefPubMed Bien-Ly N, Boswell CA, Jeet S, Beach TG, Hoyte K, Luk W, Shihadeh V, Ulufatu S, Foreman O, Lu Y, et al. Lack of widespread BBB disruption in Alzheimer’s disease models: focus on therapeutic antibodies. Neuron. 2015;88:289–97.CrossRefPubMed
70.
go back to reference Chen P, Zhao W, Guo Y, Xu J, Yin M. CX3CL1/CX3CR1 in Alzheimer’s disease: a target for neuroprotection. Biomed Res Int. 2016;2016:8090918.PubMedPubMedCentral Chen P, Zhao W, Guo Y, Xu J, Yin M. CX3CL1/CX3CR1 in Alzheimer’s disease: a target for neuroprotection. Biomed Res Int. 2016;2016:8090918.PubMedPubMedCentral
71.
go back to reference Harrison JK, Jiang Y, Chen S, Xia Y, Maciejewski D, McNamara RK, Streit WJ, Salafranca MN, Adhikari S, Thompson DA, et al. Role for neuronally derived fractalkine in mediating interactions between neurons and CX3CR1-expressing microglia. Proc Natl Acad Sci U S A. 1998;95:10896–901.CrossRefPubMedPubMedCentral Harrison JK, Jiang Y, Chen S, Xia Y, Maciejewski D, McNamara RK, Streit WJ, Salafranca MN, Adhikari S, Thompson DA, et al. Role for neuronally derived fractalkine in mediating interactions between neurons and CX3CR1-expressing microglia. Proc Natl Acad Sci U S A. 1998;95:10896–901.CrossRefPubMedPubMedCentral
72.
73.
go back to reference Rustenhoven J, Jansson D, Smyth LC, Dragunow M. Brain pericytes as mediators of neuroinflammation. Trends Pharmacol Sci. 2017;38:291–304.CrossRefPubMed Rustenhoven J, Jansson D, Smyth LC, Dragunow M. Brain pericytes as mediators of neuroinflammation. Trends Pharmacol Sci. 2017;38:291–304.CrossRefPubMed
75.
go back to reference Kim KW, Vallon-Eberhard A, Zigmond E, Farache J, Shezen E, Shakhar G, Ludwig A, Lira SA, Jung S. In vivo structure/function and expression analysis of the CX3C chemokine fractalkine. Blood. 2011;118:e156–67.CrossRefPubMedPubMedCentral Kim KW, Vallon-Eberhard A, Zigmond E, Farache J, Shezen E, Shakhar G, Ludwig A, Lira SA, Jung S. In vivo structure/function and expression analysis of the CX3C chemokine fractalkine. Blood. 2011;118:e156–67.CrossRefPubMedPubMedCentral
76.
go back to reference Strobel S, Grunblatt E, Riederer P, Heinsen H, Arzberger T, Al-Sarraj S, Troakes C, Ferrer I, Monoranu CM. Changes in the expression of genes related to neuroinflammation over the course of sporadic Alzheimer’s disease progression: CX3CL1, TREM2, and PPARgamma. J Neural Transm (Vienna). 2015;122:1069–76.CrossRef Strobel S, Grunblatt E, Riederer P, Heinsen H, Arzberger T, Al-Sarraj S, Troakes C, Ferrer I, Monoranu CM. Changes in the expression of genes related to neuroinflammation over the course of sporadic Alzheimer’s disease progression: CX3CL1, TREM2, and PPARgamma. J Neural Transm (Vienna). 2015;122:1069–76.CrossRef
77.
go back to reference Nash KR, Moran P, Finneran DJ, Hudson C, Robinson J, Morgan D, Bickford PC. Fractalkine over expression suppresses alpha-synuclein-mediated neurodegeneration. Mol Ther. 2015;23:17–23.CrossRefPubMed Nash KR, Moran P, Finneran DJ, Hudson C, Robinson J, Morgan D, Bickford PC. Fractalkine over expression suppresses alpha-synuclein-mediated neurodegeneration. Mol Ther. 2015;23:17–23.CrossRefPubMed
78.
go back to reference 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.CrossRefPubMed 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.CrossRefPubMed
79.
go back to reference Lee S, Varvel NH, Konerth ME, Xu G, Cardona AE, Ransohoff RM, Lamb BT. CX3CR1 deficiency alters microglial activation and reduces beta-amyloid deposition in two Alzheimer’s disease mouse models. Am J Pathol. 2010;177:2549–62.CrossRefPubMedPubMedCentral Lee S, Varvel NH, Konerth ME, Xu G, Cardona AE, Ransohoff RM, Lamb BT. CX3CR1 deficiency alters microglial activation and reduces beta-amyloid deposition in two Alzheimer’s disease mouse models. Am J Pathol. 2010;177:2549–62.CrossRefPubMedPubMedCentral
80.
go back to reference Febinger HY, Thomasy HE, Pavlova MN, Ringgold KM, Barf PR, George AM, Grillo JN, Bachstetter AD, Garcia JA, Cardona AE, et al. Time-dependent effects of CX3CR1 in a mouse model of mild traumatic brain injury. J Neuroinflammation. 2015;12:154.CrossRefPubMedPubMedCentral Febinger HY, Thomasy HE, Pavlova MN, Ringgold KM, Barf PR, George AM, Grillo JN, Bachstetter AD, Garcia JA, Cardona AE, et al. Time-dependent effects of CX3CR1 in a mouse model of mild traumatic brain injury. J Neuroinflammation. 2015;12:154.CrossRefPubMedPubMedCentral
81.
go back to reference McComb JG, Ranganathan M, Liu XH, Pilewski JM, Ray P, Watkins SC, Choi AM, Lee JS. CX3CL1 up-regulation is associated with recruitment of CX3CR1+ mononuclear phagocytes and T lymphocytes in the lungs during cigarette smoke-induced emphysema. Am J Pathol. 2008;173:949–61.CrossRefPubMedPubMedCentral McComb JG, Ranganathan M, Liu XH, Pilewski JM, Ray P, Watkins SC, Choi AM, Lee JS. CX3CL1 up-regulation is associated with recruitment of CX3CR1+ mononuclear phagocytes and T lymphocytes in the lungs during cigarette smoke-induced emphysema. Am J Pathol. 2008;173:949–61.CrossRefPubMedPubMedCentral
82.
go back to reference White GE, McNeill E, Channon KM, Greaves DR. Fractalkine promotes human monocyte survival via a reduction in oxidative stress. Arterioscler Thromb Vasc Biol. 2014;34:2554–62.CrossRefPubMedPubMedCentral White GE, McNeill E, Channon KM, Greaves DR. Fractalkine promotes human monocyte survival via a reduction in oxidative stress. Arterioscler Thromb Vasc Biol. 2014;34:2554–62.CrossRefPubMedPubMedCentral
83.
go back to reference Deshmane SL, Kremlev S, Amini S, Sawaya BE. Monocyte chemoattractant protein-1 (MCP-1): an overview. J Interf Cytokine Res. 2009;29:313–26.CrossRef Deshmane SL, Kremlev S, Amini S, Sawaya BE. Monocyte chemoattractant protein-1 (MCP-1): an overview. J Interf Cytokine Res. 2009;29:313–26.CrossRef
84.
go back to reference Galimberti D, Fenoglio C, Lovati C, Venturelli E, Guidi I, Corra B, Scalabrini D, Clerici F, Mariani C, Bresolin N, Scarpini E. Serum MCP-1 levels are increased in mild cognitive impairment and mild Alzheimer’s disease. Neurobiol Aging. 2006;27:1763–8.CrossRefPubMed Galimberti D, Fenoglio C, Lovati C, Venturelli E, Guidi I, Corra B, Scalabrini D, Clerici F, Mariani C, Bresolin N, Scarpini E. Serum MCP-1 levels are increased in mild cognitive impairment and mild Alzheimer’s disease. Neurobiol Aging. 2006;27:1763–8.CrossRefPubMed
85.
go back to reference Correa JD, Starling D, Teixeira AL, Caramelli P, Silva TA. Chemokines in CSF of Alzheimer’s disease patients. Arq Neuropsiquiatr. 2011;69:455–9.CrossRefPubMed Correa JD, Starling D, Teixeira AL, Caramelli P, Silva TA. Chemokines in CSF of Alzheimer’s disease patients. Arq Neuropsiquiatr. 2011;69:455–9.CrossRefPubMed
86.
go back to reference Galimberti D, Schoonenboom N, Scheltens P, Fenoglio C, Bouwman F, Venturelli E, Guidi I, Blankenstein MA, Bresolin N, Scarpini E. Intrathecal chemokine synthesis in mild cognitive impairment and Alzheimer disease. Arch Neurol. 2006;63:538–43.CrossRefPubMed Galimberti D, Schoonenboom N, Scheltens P, Fenoglio C, Bouwman F, Venturelli E, Guidi I, Blankenstein MA, Bresolin N, Scarpini E. Intrathecal chemokine synthesis in mild cognitive impairment and Alzheimer disease. Arch Neurol. 2006;63:538–43.CrossRefPubMed
87.
go back to reference Valiathan R, Ashman M, Asthana D. Effects of ageing on the immune system: infants to elderly. Scand J Immunol. 2016;83:255–66.CrossRefPubMed Valiathan R, Ashman M, Asthana D. Effects of ageing on the immune system: infants to elderly. Scand J Immunol. 2016;83:255–66.CrossRefPubMed
88.
go back to reference Song Y, Shen H, Schenten D, Shan P, Lee PJ, Goldstein DR. Aging enhances the basal production of IL-6 and CCL2 in vascular smooth muscle cells. Arterioscler Thromb Vasc Biol. 2012;32:103–9.CrossRefPubMed Song Y, Shen H, Schenten D, Shan P, Lee PJ, Goldstein DR. Aging enhances the basal production of IL-6 and CCL2 in vascular smooth muscle cells. Arterioscler Thromb Vasc Biol. 2012;32:103–9.CrossRefPubMed
89.
go back to reference Wang M, Jiang L, Monticone RE, Lakatta EG. Proinflammation: the key to arterial aging. Trends Endocrinol Metab. 2014;25:72–9.CrossRefPubMed Wang M, Jiang L, Monticone RE, Lakatta EG. Proinflammation: the key to arterial aging. Trends Endocrinol Metab. 2014;25:72–9.CrossRefPubMed
90.
go back to reference Kiyota T, Gendelman HE, Weir RA, Higgins EE, Zhang G, Jain M. CCL2 affects beta-amyloidosis and progressive neurocognitive dysfunction in a mouse model of Alzheimer’s disease. Neurobiol Aging. 2013;34:1060–8.CrossRefPubMed Kiyota T, Gendelman HE, Weir RA, Higgins EE, Zhang G, Jain M. CCL2 affects beta-amyloidosis and progressive neurocognitive dysfunction in a mouse model of Alzheimer’s disease. Neurobiol Aging. 2013;34:1060–8.CrossRefPubMed
91.
go back to reference El Khoury J, Luster AD. Mechanisms of microglia accumulation in Alzheimer’s disease: therapeutic implications. Trends Pharmacol Sci. 2008;29:626–32.CrossRefPubMed El Khoury J, Luster AD. Mechanisms of microglia accumulation in Alzheimer’s disease: therapeutic implications. Trends Pharmacol Sci. 2008;29:626–32.CrossRefPubMed
92.
go back to reference Yamamoto M, Horiba M, Buescher JL, Huang D, Gendelman HE, Ransohoff RM, Ikezu T. Overexpression of monocyte chemotactic protein-1/CCL2 in beta-amyloid precursor protein transgenic mice show accelerated diffuse beta-amyloid deposition. Am J Pathol. 2005;166:1475–85.CrossRefPubMedPubMedCentral Yamamoto M, Horiba M, Buescher JL, Huang D, Gendelman HE, Ransohoff RM, Ikezu T. Overexpression of monocyte chemotactic protein-1/CCL2 in beta-amyloid precursor protein transgenic mice show accelerated diffuse beta-amyloid deposition. Am J Pathol. 2005;166:1475–85.CrossRefPubMedPubMedCentral
93.
go back to reference Geppert AM, Losy J, Przedpelska-Ober E, Kozubski W. CCL3 correlates with the number of mood disturbances and personality changes in patients with Alzheimer’s disease. Psychiatry Res. 2010;176:261–4.CrossRefPubMed Geppert AM, Losy J, Przedpelska-Ober E, Kozubski W. CCL3 correlates with the number of mood disturbances and personality changes in patients with Alzheimer’s disease. Psychiatry Res. 2010;176:261–4.CrossRefPubMed
94.
go back to reference Blasko I, Lederer W, Oberbauer H, Walch T, Kemmler G, Hinterhuber H, Marksteiner J, Humpel C. Measurement of thirteen biological markers in CSF of patients with Alzheimer’s disease and other dementias. Dement Geriatr Cogn Disord. 2006;21:9–15.CrossRefPubMed Blasko I, Lederer W, Oberbauer H, Walch T, Kemmler G, Hinterhuber H, Marksteiner J, Humpel C. Measurement of thirteen biological markers in CSF of patients with Alzheimer’s disease and other dementias. Dement Geriatr Cogn Disord. 2006;21:9–15.CrossRefPubMed
95.
go back to reference Marciniak E, Faivre E, Dutar P, Alves Pires C, Demeyer D, Caillierez R, Laloux C, Buee L, Blum D, Humez S. The chemokine MIP-1alpha/CCL3 impairs mouse hippocampal synaptic transmission, plasticity and memory. Sci Rep. 2015;5:15862.CrossRefPubMedPubMedCentral Marciniak E, Faivre E, Dutar P, Alves Pires C, Demeyer D, Caillierez R, Laloux C, Buee L, Blum D, Humez S. The chemokine MIP-1alpha/CCL3 impairs mouse hippocampal synaptic transmission, plasticity and memory. Sci Rep. 2015;5:15862.CrossRefPubMedPubMedCentral
96.
go back to reference Man SM, Ma YR, Shang DS, Zhao WD, Li B, Guo DW, Fang WG, Zhu L, Chen YH. Peripheral T cells overexpress MIP-1alpha to enhance its transendothelial migration in Alzheimer’s disease. Neurobiol Aging. 2007;28:485–96.CrossRefPubMed Man SM, Ma YR, Shang DS, Zhao WD, Li B, Guo DW, Fang WG, Zhu L, Chen YH. Peripheral T cells overexpress MIP-1alpha to enhance its transendothelial migration in Alzheimer’s disease. Neurobiol Aging. 2007;28:485–96.CrossRefPubMed
97.
go back to reference Tripathy D, Thirumangalakudi L, Grammas P. RANTES upregulation in the Alzheimer’s disease brain: a possible neuroprotective role. Neurobiol Aging. 2010;31:8–16.CrossRefPubMed Tripathy D, Thirumangalakudi L, Grammas P. RANTES upregulation in the Alzheimer’s disease brain: a possible neuroprotective role. Neurobiol Aging. 2010;31:8–16.CrossRefPubMed
98.
go back to reference Haskins M, Jones TE, Lu Q, Bareiss SK. Early alterations in blood and brain RANTES and MCP-1 expression and the effect of exercise frequency in the 3xTg-AD mouse model of Alzheimer’s disease. Neurosci Lett. 2016;610:165–70.CrossRefPubMed Haskins M, Jones TE, Lu Q, Bareiss SK. Early alterations in blood and brain RANTES and MCP-1 expression and the effect of exercise frequency in the 3xTg-AD mouse model of Alzheimer’s disease. Neurosci Lett. 2016;610:165–70.CrossRefPubMed
99.
go back to reference Xia MQ, Bacskai BJ, Knowles RB, Qin SX, Hyman BT. Expression of the chemokine receptor CXCR3 on neurons and the elevated expression of its ligand IP-10 in reactive astrocytes: in vitro ERK1/2 activation and role in Alzheimer’s disease. J Neuroimmunol. 2000;108:227–35.CrossRefPubMed Xia MQ, Bacskai BJ, Knowles RB, Qin SX, Hyman BT. Expression of the chemokine receptor CXCR3 on neurons and the elevated expression of its ligand IP-10 in reactive astrocytes: in vitro ERK1/2 activation and role in Alzheimer’s disease. J Neuroimmunol. 2000;108:227–35.CrossRefPubMed
100.
go back to reference Duan RS, Yang X, Chen ZG, Lu MO, Morris C, Winblad B, Zhu J. Decreased fractalkine and increased IP-10 expression in aged brain of APP (swe) transgenic mice. Neurochem Res. 2008;33:1085–9.CrossRefPubMed Duan RS, Yang X, Chen ZG, Lu MO, Morris C, Winblad B, Zhu J. Decreased fractalkine and increased IP-10 expression in aged brain of APP (swe) transgenic mice. Neurochem Res. 2008;33:1085–9.CrossRefPubMed
101.
go back to reference Krauthausen M, Kummer MP, Zimmermann J, Reyes-Irisarri E, Terwel D, Bulic B, Heneka MT, Muller M. CXCR3 promotes plaque formation and behavioral deficits in an Alzheimer’s disease model. J Clin Invest. 2015;125:365–78.CrossRefPubMed Krauthausen M, Kummer MP, Zimmermann J, Reyes-Irisarri E, Terwel D, Bulic B, Heneka MT, Muller M. CXCR3 promotes plaque formation and behavioral deficits in an Alzheimer’s disease model. J Clin Invest. 2015;125:365–78.CrossRefPubMed
Metadata
Title
Longitudinal chemokine profile expression in a blood-brain barrier model from Alzheimer transgenic versus wild-type mice
Authors
J. Vérité
T. Janet
D. Chassaing
B. Fauconneau
H. Rabeony
G. Page
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2018
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-018-1220-7

Other articles of this Issue 1/2018

Journal of Neuroinflammation 1/2018 Go to the issue