M. A. Anderson, Y. Ao, and M. V. Sofroniew, Heterogeneity of reactive astrocytes, Neurosci Lett, vol.565, pp.23-29, 2014.

A. Araque, G. Carmignoto, P. G. Haydon, S. H. Oliet, R. Robitaille et al., Gliotransmitters travel in time and space, Neuron, vol.81, pp.728-739, 2014.

A. Badin, R. Spinnewyn, B. Gaillard, M. C. , J. C. Malgorn et al., IRC-082451, a novel multitargeting molecule, reduces L-DOPA-induced dyskinesias in MPTP Parkinsonian primates, PLoS One, vol.8, p.52680, 2013.
URL : https://hal.archives-ouvertes.fr/inserm-00857944

B. Haim, L. Carrillo-de-sauvage, M. A. Ceyzeriat, K. Escartin, and C. , Elusive roles for reactive astrocytes in neurodegenerative diseases, Frontiers in cellular neuroscience, vol.9, p.278, 2015.
URL : https://hal.archives-ouvertes.fr/cea-02142599

B. Haim, L. Ceyzeriat, K. Carrillo-de-sauvage, M. A. , A. F. Auregan et al., The JAK/STAT3 Pathway Is a Common Inducer of Astrocyte Reactivity in Alzheimer's and Huntington's Diseases, J Neurosci, vol.35, pp.2817-2829, 2015.

J. F. Bromberg, M. H. Wrzeszczynska, G. Devgan, Y. Zhao, R. G. Pestell et al., Stat3 as an oncogene, Cell, vol.98, pp.295-303, 1999.

J. E. Burda and M. V. Sofroniew, Reactive gliosis and the multicellular response to CNS damage and disease, Neuron, vol.81, pp.229-248, 2014.

I. Carrero, M. R. Gonzalo, B. Martin, J. M. Sanz-anquela, J. Arevalo-serrano et al., Oligomers of beta-amyloid protein (Abeta1-42) induce the activation of cyclooxygenase-2 in astrocytes via an interaction with interleukin-1beta, tumour necrosis factor-alpha, and a nuclear factor kappa-B mechanism in the rat brain, Exp Neurol, vol.236, pp.215-227, 2012.

K. Ceyzeriat, L. Abjean, M. A. Carrillo-de-sauvage, B. Haim, L. Escartin et al., The complex STATes of astrocyte reactivity: How are they controlled by the JAK-STAT3 pathway, Neuroscience, vol.330, pp.205-218, 2016.
URL : https://hal.archives-ouvertes.fr/cea-02142559

Y. H. Chung, K. M. Joo, H. C. Lim, M. H. Cho, D. Kim et al., Immunohistochemical study on the distribution of phosphorylated extracellular signal-regulated kinase (ERK) in the central nervous system of SOD1G93A transgenic mice, Brain Res, vol.1050, pp.203-209, 2005.

A. Colin, M. Faideau, N. Dufour, G. Auregan, R. Hassig et al., Engineered lentiviral vector targeting astrocytes in vivo, Glia, vol.57, pp.667-679, 2009.
URL : https://hal.archives-ouvertes.fr/tel-00348988

C. Crosio, C. Valle, A. Casciati, C. Iaccarino, C. Mt-;-d'orange et al., Astroglial inhibition of NF-kappaB does not ameliorate disease onset and progression in a mouse model for amyotrophic lateral sclerosis (ALS), 2011.

A. Dobin, C. A. Davis, F. Schlesinger, J. Drenkow, C. Zaleski et al., STAR: ultrafast universal RNA-seq aligner, Bioinformatics, vol.29, pp.15-21, 2013.

G. B. Ehret, P. Reichenbach, U. Schindler, C. M. Horvath, S. Fritz et al., DNA binding specificity of different STAT proteins. Comparison of in vitro specificity with natural target sites, J Biol Chem, vol.276, pp.6675-6688, 2001.

C. Escartin, E. Brouillet, P. Gubellini, Y. Trioulier, C. Jacquard et al., Ciliary neurotrophic factor activates astrocytes, redistributes their glutamate transporters GLAST and GLT-1 to raft microdomains, and improves glutamate handling in vivo, J Neurosci, vol.26, pp.5978-5989, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00088896

R. Eulenfeld, A. Dittrich, C. Khouri, P. J. Muller, B. Mutze et al., Interleukin-6 signalling: more than Jaks and STATs, European journal of cell biology, vol.91, pp.486-495, 2012.

Y. Fan, R. Mao, and J. Yang, NF-kappaB and STAT3 signaling pathways collaboratively link inflammation to cancer, Protein & cell, vol.4, pp.176-185, 2013.

A. M. Fernandez, S. Jimenez, M. Mecha, D. Davila, C. Guaza et al., Regulation of the phosphatase calcineurin by insulin-like growth factor I unveils a key role of astrocytes in Alzheimer's pathology, Molecular psychiatry, vol.17, pp.705-718, 2012.

T. A. Ferreira, A. V. Blackman, J. Oyrer, S. Jayabal, A. J. Chung et al., Neuronal morphometry directly from bitmap images, Nature methods, vol.11, pp.982-984, 2014.

A. E. Frakes, L. Ferraiuolo, A. M. Haidet-phillips, L. Schmelzer, L. Braun et al., Microglia induce motor neuron death via the classical NF-kappaB pathway in amyotrophic lateral sclerosis, Neuron, vol.81, pp.1009-1023, 2014.

J. L. Furman, D. M. Sama, J. C. Gant, T. L. Beckett, M. P. Murphy et al., Targeting astrocytes ameliorates neurologic changes in a mouse model of Alzheimer's disease, J Neurosci, vol.32, pp.16129-16140, 2012.

S. Haan, S. Wuller, J. Kaczor, C. Rolvering, T. Nocker et al., SOCS-mediated downregulation of mutant Jak2 (V617F, T875N and K539L) counteracts cytokine-independent signaling, Oncogene, vol.28, pp.3069-3080, 2009.

M. Hara, K. Kobayakawa, Y. Ohkawa, H. Kumamaru, K. Yokota et al., Interaction of reactive astrocytes with type I collagen induces astrocytic scar formation through the integrin-N-cadherin pathway after spinal cord injury, Nat Med, vol.23, pp.818-828, 2017.

M. T. Heneka, M. J. Carson, E. Khoury, J. Landreth, G. E. Brosseron et al., Neuroinflammation in Alzheimer's disease, vol.14, pp.388-405, 2015.

M. T. Heneka, M. P. Kummer, and E. Latz, Innate immune activation in neurodegenerative disease, Nature reviews Immunology, vol.14, pp.463-477, 2014.

E. M. Hol and M. Pekny, Glial fibrillary acidic protein (GFAP) and the astrocyte intermediate filament system in diseases of the central nervous system, Current opinion in cell biology, vol.32, pp.121-130, 2015.

S. Hong, V. F. Beja-glasser, B. M. Nfonoyim, A. Frouin, S. Li et al., Complement and microglia mediate early synapse loss in Alzheimer mouse models, Science, vol.352, pp.712-716, 2016.

H. Y. Hsiao, Y. C. Chen, H. M. Chen, P. H. Tu, and Y. Chern, A critical role of astrocyte-mediated nuclear factor-kappaB-dependent inflammation in Huntington's disease, Hum Mol Genet, 2013.

W. Huang-da, B. T. Sherman, and R. A. Lempicki, Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources, Nature protocols, vol.4, pp.44-57, 2009.

J. L. Jankowsky, D. J. Fadale, J. Anderson, G. M. Xu, V. Gonzales et al., Mutant presenilins specifically elevate the levels of the 42 residue beta-amyloid peptide in vivo: evidence for augmentation of a 42-specific gamma secretase, Hum Mol Genet, vol.13, pp.159-170, 2004.

S. Jo, O. Yarishkin, Y. J. Hwang, Y. E. Chun, M. Park et al., GABA from reactive astrocytes impairs memory in mouse models of Alzheimer's disease, 2014.

J. Lin, C. C. Yu, K. Hatcher, A. Huang, T. W. Lee et al., Identification of diverse astrocyte populations and their malignant analogs, Nat Neurosci, vol.20, pp.396-405, 2017.

W. Kang and J. M. Hebert, Signaling pathways in reactive astrocytes, a genetic perspective, Mol Neurobiol, vol.43, pp.147-154, 2011.

S. K. Kim, J. Nabekura, and S. Koizumi, Astrocyte-mediated synapse remodeling in the pathological brain, Glia, vol.65, pp.1719-1727, 2017.

H. F. Kuipers, J. Yoon, J. Van-horssen, M. H. Han, P. L. Bollyky et al., Phosphorylation of alphaB-crystallin supports reactive astrogliosis in demyelination, Proc Natl Acad Sci U S A, vol.114, pp.1745-1754, 2017.

P. Langfelder and S. Horvath, WGCNA: an R package for weighted correlation network analysis, BMC bioinformatics, vol.9, p.559, 2008.

M. D. Lecomte, I. S. Shimada, C. Sherwin, and J. L. Spees, Notch1-STAT3-ETBR signaling axis controls reactive astrocyte proliferation after brain injury, Proc Natl Acad Sci U S A, vol.112, pp.8726-8731, 2015.

Y. Lee, A. Messing, M. Su, and M. Brenner, GFAP promoter elements required for region-specific and astrocyte-specific expression, Glia, vol.56, pp.481-493, 2008.

H. Lian, L. Yang, A. Cole, L. Sun, A. C. Chiang et al., NFkappaB-Activated Astroglial Release of Complement C3 Compromises Neuronal Morphology and Function Associated with Alzheimer's Disease, Neuron, vol.85, pp.101-115, 2015.

S. A. Liddelow and B. A. Barres, Reactive Astrocytes: Production, Function, and Therapeutic Potential, Immunity, vol.46, pp.957-967, 2017.

S. A. Liddelow, K. A. Guttenplan, L. E. Clarke, F. C. Bennett, C. J. Bohlen et al., Neurotoxic reactive astrocytes are induced by activated microglia, Nature, 2017.

E. M. Linossi, J. J. Babon, D. J. Hilton, and S. E. Nicholson, Suppression of cytokine signaling: the SOCS perspective, Cytokine & growth factor reviews, vol.24, pp.241-248, 2013.

M. I. Love, W. Huber, and S. Anders, Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2, Genome Biol, vol.15, p.550, 2014.

X. Ma, S. L. Reynolds, B. J. Baker, X. Li, E. N. Benveniste et al., IL-17 enhancement of the IL-6 signaling cascade in astrocytes, J Immunol, vol.184, pp.4898-4906, 2010.

V. Matyash and H. Kettenmann, Heterogeneity in astrocyte morphology and physiology, Brain Res Rev, vol.63, pp.2-10, 2010.

R. Medeiros and F. M. Laferla, Astrocytes: conductors of the Alzheimer disease neuroinflammatory symphony, Exp Neurol, vol.239, pp.133-138, 2013.

C. Mertens, J. E. Darnell, and J. , SnapShot: JAK-STAT signaling, Cell, vol.131, p.612, 2007.

P. J. Murray, The JAK-STAT signaling pathway: input and output integration, J Immunol, vol.178, pp.2623-2629, 2007.

C. S. Nicolas, S. Peineau, M. Amici, Z. Csaba, A. Fafouri et al., The Jak/STAT pathway is involved in synaptic plasticity, Neuron, vol.73, pp.374-390, 2012.

L. Noli, A. Capalbo, C. Ogilvie, Y. Khalaf, and D. Ilic, Discordant Growth of Monozygotic Twins Starts at the Blastocyst Stage: A Case Study, Stem cell reports, vol.5, pp.946-953, 2015.

J. P. O'callaghan and K. Sriram, Focused microwave irradiation of the brain preserves in vivo protein phosphorylation: comparison with other methods of sacrifice and analysis of multiple phosphoproteins, J Neurosci Methods, vol.135, pp.159-168, 2004.

S. Oddo, A. Caccamo, J. D. Shepherd, M. P. Murphy, T. E. Golde et al., Triple-transgenic model of Alzheimer's disease with plaques and tangles: intracellular Abeta and synaptic dysfunction, Neuron, vol.39, pp.409-421, 2003.

P. Oeckl, M. Lattke, T. Wirth, B. Baumann, and B. Ferger, Astrocyte-specific IKK2 activation in mice is sufficient to induce neuroinflammation but does not increase susceptibility to MPTP, Neurobiol Dis, vol.48, pp.481-487, 2012.

M. Orre, W. Kamphuis, L. M. Osborn, A. H. Jansen, L. Kooijman et al., Isolation of glia from Alzheimer's mice reveals inflammation and dysfunction, Neurobiol Aging, vol.35, pp.2746-2760, 2014.

P. I. Ortinski, J. Dong, A. Mungenast, C. Yue, H. Takano et al., Selective induction of astrocytic gliosis generates deficits in neuronal inhibition, Nat Neurosci, vol.13, pp.584-591, 2010.

A. Panatier, J. Vallee, M. Haber, K. K. Murai, J. C. Lacaille et al., Astrocytes are endogenous regulators of basal transmission at central synapses, Cell, vol.146, pp.785-798, 2011.

S. A. Robertson, R. I. Koleva, L. S. Argetsinger, C. Carter-su, J. A. Marto et al., Regulation of Jak2 function by phosphorylation of Tyr317 and Tyr637 during cytokine signaling, Mol Cell Biol, vol.29, pp.3367-3378, 2009.

V. Rothhammer, I. D. Mascanfroni, L. Bunse, M. C. Takenaka, J. E. Kenison et al., ) Type I interferons and microbial metabolites of tryptophan modulate astrocyte activity and central nervous system inflammation via the aryl hydrocarbon receptor, Nat Med, vol.22, pp.586-597, 2016.

K. Schindowski, A. Bretteville, K. Leroy, S. Begard, J. P. Brion et al., Alzheimer's disease-like tau neuropathology leads to memory deficits and loss of functional synapses in a novel mutated tau transgenic mouse without any motor deficits, Am J Pathol, vol.169, pp.599-616, 2006.

K. Sharma, S. Schmitt, C. G. Bergner, S. Tyanova, N. Kannaiyan et al., Cell type-and brain regionresolved mouse brain proteome, Nat Neurosci, vol.18, pp.1819-1831, 2015.

K. Shuai and B. Liu, Regulation of JAK-STAT signalling in the immune system, Nature reviews Immunology, vol.3, pp.900-911, 2003.

A. Singh and W. C. Abraham, Astrocytes and synaptic plasticity in health and disease, Experimental brain research, vol.235, pp.1645-1655, 2017.

S. Sirko, G. Behrendt, P. A. Johansson, P. Tripathi, M. Costa et al., Reactive glia in the injured brain acquire stem cell properties in response to sonic hedgehog glia, Cell stem cell, vol.12, pp.426-439, 2013.

K. Srinivasan, B. A. Friedman, J. L. Larson, B. E. Lauffer, L. D. Goldstein et al., Untangling the brain's neuroinflammatory and neurodegenerative transcriptional responses, Nature communications, vol.7, p.11295, 2016.

A. H. Stephan, B. A. Barres, and B. Stevens, The complement system: an unexpected role in synaptic pruning during development and disease, Annual review of neuroscience, vol.35, pp.369-389, 2012.

G. E. Tyzack, S. Sitnikov, D. Barson, K. L. Adams-carr, N. K. Lau et al., Astrocyte response to motor neuron injury promotes structural synaptic plasticity via STAT3-regulated TSP-1 expression, Nature communications, vol.5, p.4294, 2014.

S. Viana-da-silva, M. G. Haberl, P. Zhang, P. Bethge, C. Lemos et al., Early synaptic deficits in the APP/PS1 mouse model of Alzheimer's disease involve neuronal adenosine A2A receptors, Nature communications, vol.7, p.11915, 2016.

A. Volianskis, R. Kostner, M. Molgaard, S. Hass, and M. S. Jensen, Episodic memory deficits are not related to altered glutamatergic synaptic transmission and plasticity in the CA1 hippocampus of the APPswe/PS1deltaE9-deleted transgenic mice model of ss-amyloidosis, Neurobiol Aging, vol.31, pp.1173-1187, 2010.

A. M. Wojtowicz, A. Dvorzhak, M. Semtner, and R. Grantyn, Reduced tonic inhibition in striatal output neurons from Huntington mice due to loss of astrocytic GABA release through GAT-3, Frontiers in neural circuits, vol.7, p.188, 2013.

Z. Wu, Z. Guo, M. Gearing, and G. Chen, Tonic inhibition in dentate gyrus impairs long-term potentiation and memory in an Alzhiemer's disease model, Nature communications, vol.5, p.4159, 2014.

J. Yang, X. Liao, M. K. Agarwal, L. Barnes, P. E. Auron et al., Unphosphorylated STAT3 accumulates in response to IL-6 and activates transcription by binding to NFkappaB, Genes & development, vol.21, pp.1396-1408, 2007.

J. L. Zamanian, L. Xu, L. C. Foo, N. Nouri, L. Zhou et al., Genomic analysis of reactive astrogliosis, J Neurosci, vol.32, pp.6391-6410, 2012.

Y. Zhang and B. A. Barres, Astrocyte heterogeneity: an underappreciated topic in neurobiology, Curr Opin Neurobiol, vol.20, pp.588-594, 2010.

R. Abeti, A. Y. Abramov, and M. R. Duchen, Beta-amyloid activates PARP causing astrocytic metabolic failure and neuronal death, Brain, vol.134, pp.1658-1672, 2011.

I. Allaman, M. Belanger, and P. J. Magistretti, Astrocyte-neuron metabolic relationships: for better and for worse, Trends Neurosci, vol.34, pp.76-87, 2011.

I. Allaman, M. Gavillet, M. Belanger, T. Laroche, D. Viertl et al., Amyloid-beta aggregates cause alterations of astrocytic metabolic phenotype: impact on neuronal viability, J Neurosci, vol.30, pp.3326-3338, 2010.

M. A. Anderson, Y. Ao, and M. V. Sofroniew, Heterogeneity of reactive astrocytes, Neurosci Lett, vol.565, pp.23-29, 2014.

M. A. Anderson, J. E. Burda, Y. Ren, Y. Ao, O. Shea et al., Astrocyte scar formation aids central nervous system axon regeneration, Nature, vol.532, pp.195-200, 2016.

A. Araque, G. Carmignoto, P. G. Haydon, S. H. Oliet, R. Robitaille et al., Gliotransmitters travel in time and space, Neuron, vol.81, pp.728-739, 2014.

B. A. Barres, The mystery and magic of glia: a perspective on their roles in health and disease, Neuron, vol.60, pp.430-440, 2008.

M. Belanger, I. Allaman, and P. J. Magistretti, Brain energy metabolism: focus on astrocyte-neuron metabolic cooperation, Cell metabolism, vol.14, pp.724-738, 2011.

L. Ben-haim-ceyzériat, C. De-sauvage, M. A. , A. D. Abjean, L. Gipstein et al.,

B. Haim, L. Carrillo-de-sauvage, M. A. Ceyzeriat, K. Escartin, and C. , Elusive roles for reactive astrocytes in neurodegenerative diseases, Frontiers in cellular neuroscience, vol.9, p.278, 2015.
URL : https://hal.archives-ouvertes.fr/cea-02142599

B. Haim, L. Ceyzeriat, K. Carrillo-de-sauvage, M. A. , A. F. Auregan et al., The JAK/STAT3 Pathway Is a Common Inducer of Astrocyte Reactivity in Alzheimer's and Huntington's Diseases, J Neurosci, vol.35, pp.2817-2829, 2015.

S. L. Cole and R. Vassar, The Alzheimer's disease beta-secretase enzyme, BACE1. Molecular neurodegeneration, vol.2, p.22, 2007.

D. V. Dabir, M. B. Robinson, E. Swanson, B. Zhang, J. Q. Trojanowski et al., Impaired glutamate transport in a mouse model of tau pathology in astrocytes, J Neurosci, vol.26, pp.644-654, 2006.

C. Dansokho, A. Ahmed, D. Aid, S. Toly-ndour, C. Chaigneau et al., Regulatory T cells delay disease progression in Alzheimer-like pathology, Brain, vol.139, pp.1237-1251, 2016.

D. Strooper, B. Karran, and E. , The Cellular Phase of Alzheimer's Disease, Cell, vol.164, pp.603-615, 2016.

A. Delekate, M. Fuchtemeier, T. Schumacher, C. Ulbrich, M. Foddis et al., Metabotropic P2Y1 receptor signalling mediates astrocytic hyperactivity in vivo in an Alzheimer's disease mouse model, Nature communications, vol.5, p.5422, 2014.

C. Escartin, E. Brouillet, P. Gubellini, Y. Trioulier, C. Jacquard et al., Ciliary neurotrophic factor activates astrocytes, redistributes their glutamate transporters GLAST and GLT-1 to raft microdomains, and improves glutamate handling in vivo, J Neurosci, vol.26, pp.5978-5989, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00088896

C. Escartin, K. Pierre, C. A. Brouillet, E. Delzescaux, T. Guillermier et al., Activation of astrocytes by CNTF induces metabolic plasticity and increases resistance to metabolic insults, J Neurosci, vol.27, pp.7094-7104, 2007.
URL : https://hal.archives-ouvertes.fr/cea-02290623

J. L. Furman, D. M. Sama, J. C. Gant, T. L. Beckett, M. P. Murphy et al., Targeting astrocytes ameliorates neurologic changes in a mouse model of Alzheimer's disease, J Neurosci, vol.32, pp.16129-16140, 2012.

M. V. Guillot-sestier, K. R. Doty, D. Gate, J. Rodriguez, J. Leung et al., Il10 deficiency rebalances innate immunity to mitigate Alzheimer-like pathology, Neuron, vol.85, pp.534-548, 2015.

S. Haan, S. Wuller, J. Kaczor, C. Rolvering, T. Nocker et al., SOCS-mediated downregulation of mutant Jak2 (V617F, T875N and K539L) counteracts cytokine-independent signaling, Oncogene, vol.28, pp.3069-3080, 2009.

L. Hamelin, J. Lagarde, D. G. Leroy, C. Labit, M. Comley et al., Clinical It (2016) Early and protective microglial activation in Alzheimer's disease: a prospective study using 18F-DPA-714 PET imaging, Brain, vol.139, pp.1252-1264

M. T. Heneka, M. J. Carson, E. Khoury, J. Landreth, G. E. Brosseron et al., Neuroinflammation in Alzheimer's disease, vol.14, pp.388-405, 2015.

M. T. Heneka, M. Sastre, L. Dumitrescu-ozimek, I. Dewachter, J. Walter et al., Focal glial activation coincides with increased BACE1 activation and precedes amyloid plaque deposition in APP[V717I] transgenic mice, J Neuroinflammation, vol.2, p.22, 2005.

K. Herrup, The case for rejecting the amyloid cascade hypothesis, Nat Neurosci, vol.18, pp.794-799, 2015.

E. M. Hol and M. Pekny, Glial fibrillary acidic protein (GFAP) and the astrocyte intermediate filament system in diseases of the central nervous system, Current opinion in cell biology, vol.32, pp.121-130, 2015.

S. Hong, V. F. Beja-glasser, B. M. Nfonoyim, A. Frouin, S. Li et al., Complement and microglia mediate early synapse loss in Alzheimer mouse models, Science, vol.352, pp.712-716, 2016.

J. L. Jankowsky, D. J. Fadale, J. Anderson, G. M. Xu, V. Gonzales et al., Mutant presenilins specifically elevate the levels of the 42 residue beta-amyloid peptide in vivo: evidence for augmentation of a 42-specific gamma secretase, Hum Mol Genet, vol.13, pp.159-170, 2004.

S. Jo, O. Yarishkin, Y. J. Hwang, Y. E. Chun, M. Park et al., GABA from reactive astrocytes impairs memory in mouse models of Alzheimer's disease, 2014.

W. Kamphuis, L. Kooijman, M. Orre, O. Stassen, M. Pekny et al., GFAP and vimentin deficiency alters gene expression in astrocytes and microglia in wild-type mice and changes the transcriptional response of reactive glia in mouse model for Alzheimer's disease, Glia, 2015.

H. Keren-shaul, A. Spinrad, A. Weiner, O. Matcovitch-natan, R. Dvir-szternfeld et al., A Unique Microglia Type Associated with Restricting Development of Alzheimer's Disease, Cell, vol.169, pp.1276-1290, 2017.

M. Kim, J. Gillen, B. A. Landman, J. Zhou, and P. C. Van-zijl, Water saturation shift referencing (WASSR) for chemical exchange saturation transfer (CEST) experiments, Magn Reson Med, vol.61, pp.1441-1450, 2009.

M. Koistinaho, S. Lin, X. Wu, M. Esterman, D. Koger et al., Apolipoprotein E promotes astrocyte colocalization and degradation of deposited amyloid-beta peptides, Nat Med, vol.10, pp.719-726, 2004.

A. W. Kraft, X. Hu, H. Yoon, P. Yan, X. Q. Wang et al., Attenuating astrocyte activation accelerates plaque pathogenesis in APP/PS1 mice, FASEB J, vol.27, pp.187-198, 2013.

M. Krauthausen, M. P. Kummer, J. Zimmermann, E. Reyes-irisarri, D. Terwel et al., CXCR3 promotes plaque formation and behavioral deficits in an Alzheimer's disease model, J Clin Invest, vol.125, pp.365-378, 2015.

K. V. Kuchibhotla, C. R. Lattarulo, B. T. Hyman, and B. J. Bacskai, Synchronous hyperactivity and intercellular calcium waves in astrocytes in Alzheimer mice, Science, vol.323, pp.1211-1215, 2009.

C. Y. Lee and G. E. Landreth, The role of microglia in amyloid clearance from the AD brain, Journal of neural transmission, vol.117, pp.949-960, 2010.

Y. Lee, A. Messing, M. Su, and M. Brenner, GFAP promoter elements required for region-specific and astrocyte-specific expression, Glia, vol.56, pp.481-493, 2008.

C. Leyns and D. M. Holtzman, Glial contributions to neurodegeneration in tauopathies, Molecular neurodegeneration, vol.12, p.50, 2017.

H. Lian, L. Yang, A. Cole, L. Sun, A. C. Chiang et al., NFkappaB-Activated Astroglial Release of Complement C3 Compromises Neuronal Morphology and Function Associated with Alzheimer's Disease, Neuron, vol.85, pp.101-115, 2015.

M. C. Liao, C. R. Muratore, T. M. Gierahn, S. E. Sullivan, P. Srikanth et al., Single-Cell Detection of Secreted Abeta and sAPPalpha from Human IPSC-Derived Neurons and Astrocytes, J Neurosci, vol.36, pp.1730-1746, 2016.

S. A. Liddelow and B. A. Barres, Reactive Astrocytes: Production, Function, and Therapeutic Potential, Immunity, vol.46, pp.957-967, 2017.

S. A. Liddelow, K. A. Guttenplan, L. E. Clarke, F. C. Bennett, C. J. Bohlen et al., Neurotoxic reactive astrocytes are induced by activated microglia, Nature, 2017.

E. Masliah, M. Alford, R. Deteresa, M. Mallory, and L. Hansen, Deficient glutamate transport is associated with neurodegeneration in Alzheimer's disease, Ann Neurol, vol.40, pp.759-766, 1996.

E. S. Musiek and D. M. Holtzman, Three dimensions of the amyloid hypothesis: time, space and 'wingmen', Nat Neurosci, vol.18, pp.800-806, 2015.

S. Oddo, A. Caccamo, J. D. Shepherd, M. P. Murphy, T. E. Golde et al., Triple-transgenic model of Alzheimer's disease with plaques and tangles: intracellular Abeta and synaptic dysfunction, Neuron, vol.39, pp.409-421, 2003.

S. H. Oliet, R. Piet, and D. A. Poulain, Control of glutamate clearance and synaptic efficacy by glial coverage of neurons, Science, vol.292, pp.923-926, 2001.
URL : https://hal.archives-ouvertes.fr/inserm-00000059

M. Orre, W. Kamphuis, L. M. Osborn, A. H. Jansen, L. Kooijman et al., Isolation of glia from Alzheimer's mice reveals inflammation and dysfunction, Neurobiol Aging, vol.35, pp.2746-2760, 2014.

A. Panatier, D. T. Theodosis, J. P. Mothet, B. Touquet, L. Pollegioni et al., Glia-derived D-serine controls NMDA receptor activity and synaptic memory, Cell, vol.125, pp.775-784, 2006.
URL : https://hal.archives-ouvertes.fr/inserm-00078312

A. Panatier, J. Vallee, M. Haber, K. K. Murai, J. C. Lacaille et al., Astrocytes are endogenous regulators of basal transmission at central synapses, Cell, vol.146, pp.785-798, 2011.

U. Pannasch, D. Freche, G. Dallerac, G. Ghezali, C. Escartin et al., Connexin 30 sets synaptic strength by controlling astroglial synapse invasion, Nat Neurosci, vol.17, pp.549-558, 2014.
URL : https://hal.archives-ouvertes.fr/cea-02290543

J. Pepin, L. Francelle, M. A. Carrillo-de-sauvage, L. De-longprez, P. Gipchtein et al., In vivo imaging of brain glutamate defects in a knock-in mouse model of Huntington's disease, Neuroimage, vol.139, pp.53-64, 2016.

S. W. Provencher, Estimation of metabolite concentrations from localized in vivo proton NMR spectra, Magn Reson Med, vol.30, pp.672-679, 1993.

H. W. Querfurth and F. M. Laferla, Alzheimer's disease, N Engl J Med, vol.362, pp.329-344, 2010.

H. Sancheti, I. Patil, K. Kanamori, R. Diaz-brinton, W. Zhang et al., Hypermetabolic state in the 7-month-old triple transgenic mouse model of Alzheimer's disease and the effect of lipoic acid: a 13C-NMR study, J Cereb Blood Flow Metab, vol.34, pp.1749-1760, 2014.

Q. Shi, S. Chowdhury, R. Ma, K. X. Le, S. Hong et al., Complement C3 deficiency protects against neurodegeneration in aged plaque-rich APP/PS1 mice, Science translational, vol.9, 2017.

P. Sompol, J. L. Furman, M. M. Pleiss, S. D. Kraner, I. A. Artiushin et al., , 2017.

H. M. Abdul, M. A. Sama, J. L. Furman, D. M. Mathis, T. L. Beckett et al., Cognitive decline in Alzheimer's disease is associated with selective changes in calcineurin/NFAT signaling, J. Neurosci, vol.29, pp.12957-12969, 2009.

R. Abeti, A. Y. Abramov, and M. R. Duchen, Beta-amyloid activates PARP causing astrocytic metabolic failure and neuronal death, Brain, vol.134, pp.1658-1672, 2011.

A. Y. Abramov, L. Canevari, and M. R. Duchen, Beta-amyloid peptides induce mitochondrial dysfunction and oxidative stress in astrocytes and death of neurons through activation of NADPH oxidase, J. Neurosci, vol.24, pp.565-575, 2004.

Y. Agid, Parkinson's disease: pathophysiology, Lancet, vol.337, pp.1321-1324, 1991.

C. Agulhon, M. Y. Sun, T. Murphy, T. Myers, K. Lauderdale et al., Calcium signaling and gliotransmission in normal vs. reactive astrocytes, Front. Pharmacol, vol.3, p.139, 2012.

I. Allaman, M. Belanger, and P. J. Magistretti, Astrocyte-neuron metabolic relationships: for better and for worse, Trends Neurosci, vol.34, pp.76-87, 2011.

I. Allaman, M. Gavillet, M. Belanger, T. Laroche, D. Viertl et al., Amyloid-beta aggregates cause alterations of astrocytic metabolic phenotype: impact on neuronal viability, J. Neurosci, vol.30, pp.3326-3338, 2010.

M. A. Anderson, Y. Ao, and M. V. Sofroniew, Heterogeneity of reactive astrocytes, Neurosci. Lett, vol.565, pp.23-29, 2014.

J. Apelt, K. Ach, and R. Schliebs, Aging-related down-regulation of neprilysin, a putative beta-amyloid-degrading enzyme, in transgenic Tg2576 Alzheimer-like mouse brain is accompanied by an astroglial upregulation in the vicinity of beta-amyloid plaques, Neurosci. Lett, vol.339, pp.183-186, 2003.

B. Appel, Nonmammalian vertebrate glia, Neuroglia, pp.24-31, 2013.

A. Araque, G. Carmignoto, P. G. Haydon, S. H. Oliet, R. Robitaille et al., Gliotransmitters travel in time and space, Neuron, vol.81, pp.728-739, 2014.

T. Arzberger, K. Krampfl, S. Leimgruber, and A. Weindl, Changes of NMDA receptor subunit (NR1, NR2B) and glutamate transporter (GLT1) mRNA expression in Huntington's disease-an in situ hybridization study, 1997.

, J. Neuropathol. Exp. Neurol, vol.56, pp.440-454

R. M. Bachoo, R. S. Kim, K. L. Ligon, E. A. Maher, C. Brennan et al., Molecular diversity of astrocytes with implications for neurological disorders, Proc. Natl. Acad. Sci. U.S.A, vol.101, pp.8384-8389, 2004.

L. H. Barbeito, M. Pehar, P. Cassina, M. R. Vargas, H. Peluffo et al., A role for astrocytes in motor neuron loss in amyotrophic lateral sclerosis, Brain Res. Brain Res. Rev, vol.47, pp.263-274, 2004.

C. Barcia, A. Sanchez-bahillo, E. Fernandez-villalba, V. Bautista, Y. P. Poza et al., Evidence of active microglia in substantia nigra pars compacta of parkinsonian monkeys 1 year after MPTP exposure, Glia, vol.46, pp.402-409, 2004.

S. Bardehle, M. Kruger, F. Buggenthin, J. Schwausch, J. Ninkovic et al., Live imaging of astrocyte responses to acute injury reveals selective juxtavascular proliferation, Nat. Neurosci, vol.16, pp.580-586, 2013.

M. F. Beal, Parkinson's disease: a model dilemma, Nature, vol.466, pp.8-10, 2010.

M. Belanger, I. Allaman, and P. J. Magistretti, Brain energy metabolism: focus on astrocyte-neuron metabolic cooperation, Cell Metab, vol.14, pp.724-738, 2011.

L. Ben-haim, K. Ceyzeriat, M. A. Carrillo-de-sauvage, F. Aubry, G. Auregan et al., The JAK/STAT3 Pathway is a common inducer of astrocyte reactivity in Alzheimer's and Huntington's diseases, J. Neurosci, vol.35, pp.2817-2829, 2015.

C. Bendotti, C. Atzori, R. Piva, M. Tortarolo, M. J. Strong et al., Activated p38MAPK is a novel component of the intracellular inclusions found in human amyotrophic lateral sclerosis and mutant SOD1 transgenic mice, J. Neuropathol. Exp. Neurol, vol.63, pp.113-119, 2004.

C. Bendotti, M. Tortarolo, S. K. Suchak, N. Calvaresi, L. Carvelli et al., Transgenic SOD1 G93A mice develop reduced GLT-1 in spinal cord without alterations in cerebrospinal fluid glutamate levels, J. Neurochem, vol.79, pp.737-746, 2001.

Y. Bernardinelli, J. Randall, E. Janett, I. Nikonenko, S. Konig et al., Activity-dependent structural plasticity of perisynaptic astrocytic domains promotes excitatory synapse stability, Curr. Biol, vol.24, pp.1679-1688, 2014.

C. Beurrier, M. Faideau, K. E. Bennouar, C. Escartin, . Kerkerian-le et al., Ciliary neurotrophic factor protects striatal neurons against excitotoxicity by enhancing glial glutamate uptake, PLoS ONE, vol.5, p.8550, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00848866

O. G. Bhalala, L. Pan, V. Sahni, T. L. Mcguire, K. Gruner et al., microRNA-21 regulates astrocytic response following spinal cord injury, J. Neurosci, vol.32, pp.17935-17947, 2012.

O. G. Bhalala, M. Srikanth, and J. A. Kessler, The emerging roles of microRNAs in CNS injuries, Nat. Rev. Neurol, vol.9, pp.328-339, 2013.

A. Bignami and D. Dahl, The astroglial response to stabbing. Immunofluorescence studies with antibodies to astrocyte-specific protein (GFA) in mammalian and submammalian vertebrates, Neuropathol. Appl. Neurobiol, vol.2, pp.130-140, 1976.

, Frontiers in Cellular Neuroscience | www.frontiersin.org, vol.20, p.278, 2015.

M. Bosch and T. Kielian, Hemichannels in neurodegenerative diseases: is there a link to pathology? Front, Cell. Neurosci, vol.8, p.242, 2014.

L. Boussicault, A. S. Herard, N. Calingasan, F. Petit, C. Malgorn et al., Impaired brain energy metabolism in the BACHD mouse model of Huntington's disease: critical role of astrocyte-neuron interactions, J. Cereb. Blood Flow Metab, vol.34, pp.1500-1510, 2014.

J. Bradford, J. Y. Shin, M. Roberts, C. E. Wang, X. J. Li et al., Expression of mutant huntingtin in mouse brain astrocytes causes age-dependent neurological symptoms, Proc. Natl. Acad. Sci. U.S.A, vol.106, pp.22480-22485, 2009.

G. Bu, Apolipoprotein E and its receptors in Alzheimer's disease: pathways, pathogenesis and therapy, Nat. Rev. Neurosci, vol.10, pp.333-344, 2009.

A. Buffo, C. Rolando, and S. Ceruti, Astrocytes in the damaged brain: molecular and cellular insights into their reactive response and healing potential, Biochem. Pharmacol, vol.79, pp.77-89, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00535828

J. E. Burda and M. V. Sofroniew, Reactive gliosis and the multicellular response to CNS damage and disease, Neuron, vol.81, pp.229-248, 2014.

R. S. Burns, C. C. Chiueh, S. P. Markey, M. H. Ebert, D. M. Jacobowitz et al., A primate model of parkinsonism: selective destruction of dopaminergic neurons in the pars compacta of the substantia nigra by Nmethyl-4-phenyl-1,2,3,6-tetrahydropyridine, Proc. Natl. Acad. Sci. U.S.A, vol.80, pp.4546-4550, 1983.

T. G. Bush, N. Puvanachandra, C. H. Horner, A. Polito, T. Ostenfeld et al., Leukocyte infiltration, neuronal degeneration, and neurite outgrowth after ablation of scar-forming, reactive astrocytes in adult transgenic mice, Neuron, vol.23, pp.297-308, 1999.

E. A. Bushong, M. E. Martone, Y. Z. Jones, and M. H. Ellisman, Protoplasmic astrocytes in CA1 stratum radiatum occupy separate anatomical domains, J. Neurosci, vol.22, pp.183-192, 2002.

M. J. Calkins, M. R. Vargas, D. A. Johnson, J. , and J. A. , Astrocytespecific overexpression of Nrf2 protects striatal neurons from mitochondrial complex II inhibition, Toxicol. Sci, vol.115, pp.557-568, 2010.

I. Carrero, M. R. Gonzalo, B. Martin, J. M. Sanz-anquela, J. Arevalo-serrano et al., Oligomers of beta-amyloid protein (Abeta1-42) induce the activation of cyclooxygenase-2 in astrocytes via an interaction with interleukin-1beta, tumour necrosis factor-alpha, and a nuclear factor kappa-B mechanism in the rat brain, Exp. Neurol, vol.236, pp.215-227, 2012.

M. Carrillo-de-sauvage, J. Flament, Y. Bramoulle, L. Ben-haim, M. Guillermier et al., The neuroprotective agent CNTF decreases neuronal metabolites in the rat striatum: an in vivo multimodal magnetic resonance imaging study, J. Cereb. Blood Flow Metab, vol.35, pp.917-921, 2015.
URL : https://hal.archives-ouvertes.fr/cea-02168367

S. F. Carter, M. Scholl, O. Almkvist, A. Wall, H. Engler et al., Evidence for astrocytosis in prodromal Alzheimer disease provided by 11C-deuterium-L-deprenyl: a multitracer PET paradigm combining 11C-Pittsburgh compound B and 18F-FDG, J. Nucl. Med, vol.53, pp.37-46, 2012.

P. Cassina, A. Cassina, M. Pehar, R. Castellanos, M. Gandelman et al., Mitochondrial dysfunction in SOD1G93A-bearing astrocytes promotes motor neuron degeneration: prevention by mitochondrial-targeted antioxidants, J. Neurosci, vol.28, pp.4115-4122, 2008.

G. Charron, E. Doudnikoff, M. H. Canron, Q. Li, C. Vega et al., Astrocytosis in parkinsonism: considering tripartite striatal synapses in physiopathology? Front, Aging Neurosci, vol.6, p.258, 2014.

F. Chauveau, H. Boutin, N. Van-camp, F. Dolle, and B. Tavitian, Nuclear imaging of neuroinflammation: a comprehensive review of [11C]PK11195 challengers, Eur. J. Nucl. Med. Mol. Imaging, vol.35, pp.2304-2319, 2008.

P. C. Chen, M. R. Vargas, A. K. Pani, R. J. Smeyne, D. A. Johnson et al., Nrf2-mediated neuroprotection in the MPTP mouse model of Parkinson's disease: critical role for the astrocyte, Proc. Natl. Acad. Sci. U.S.A, vol.106, pp.2933-2938, 2009.

J. K. Choi, A. Dedeoglu, and B. G. Jenkins, Application of MRS to mouse models of neurodegenerative illness, NMR Biomed, vol.20, pp.216-237, 2007.

S. Y. Chou, J. Y. Weng, H. L. Lai, F. Liao, S. H. Sun et al., Expanded-polyglutamine huntingtin protein suppresses the secretion and production of a chemokine (CCL5/RANTES) by astrocytes, J. Neurosci, vol.28, pp.3277-3290, 2008.

Y. H. Chung, K. M. Joo, H. C. Lim, M. H. Cho, D. Kim et al., Immunohistochemical study on the distribution of phosphorylated extracellular signal-regulated kinase (ERK) in the central nervous system of SOD1G93A transgenic mice, Brain Res, vol.1050, pp.203-209, 2005.

S. L. Cole and R. Vassar, The Alzheimer's disease beta-secretase enzyme, BACE1, Mol. Neurodegener, vol.2, p.22, 2007.

E. H. Corder, A. M. Saunders, W. J. Strittmatter, D. E. Schmechel, P. C. Gaskell et al., Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer's disease in late onset families, Science, vol.261, pp.921-923, 1993.

C. Crosio, C. Valle, A. Casciati, C. Iaccarino, C. et al., Astroglial inhibition of NF-kappaB does not ameliorate disease onset and progression in a mouse model for amyotrophic lateral sclerosis (ALS), PLoS ONE, vol.6, p.17187, 2011.

M. E. Cudkowicz, S. Titus, M. Kearney, H. Yu, A. Sherman et al., Safety and efficacy of ceftriaxone for amyotrophic lateral sclerosis: a multi-stage, randomised, double-blind, placebo-controlled trial, Lancet Neurol, vol.13, pp.1083-1091, 2014.

J. G. Cui, Y. Y. Li, Y. Zhao, S. Bhattacharjee, and W. J. Lukiw, Differential regulation of interleukin-1 receptor-associated kinase-1 (IRAK, 2010.

, IRAK-2 by microRNA-146a and NF-kappaB in stressed human astroglial cells and in Alzheimer disease, J. Biol. Chem, vol.285, pp.38951-38960

Y. Cui, K. Masaki, R. Yamasaki, S. Imamura, S. O. Suzuki et al., Extensive dysregulations of oligodendrocytic and astrocytic connexins are associated with disease progression in an amyotrophic lateral sclerosis mouse model, J. Neuroinflammation, vol.11, p.42, 2014.

D. V. Dabir, M. B. Robinson, E. Swanson, B. Zhang, J. Q. Trojanowski et al., Impaired glutamate transport in a mouse model of tau pathology in astrocytes, J. Neurosci, vol.26, pp.644-654, 2006.

M. D'amelio, V. Cavallucci, S. Middei, C. Marchetti, S. Pacioni et al., Caspase-3 triggers early synaptic dysfunction in a mouse model of Alzheimer's disease, Nat. Neurosci, vol.14, pp.69-76, 2011.

P. Damier, E. C. Hirsch, P. Zhang, Y. Agid, and F. Javoy-agid, , 1993.

, Glutathione peroxidase, glial cells and Parkinson's disease, Neuroscience, vol.52, pp.1-6

N. C. Danbolt, Glutamate uptake, Prog. Neurobiol, vol.65, pp.1-105, 2001.

N. P. Dantuma and L. C. Bott, The ubiquitin-proteasome system in neurodegenerative diseases: precipitating factor, yet part of the solution, Front. Mol. Neurosci, vol.7, p.70, 2014.

D. Davila, K. Thibault, T. A. Fiacco, A. , and C. , Recent molecular approaches to understanding astrocyte function, Front. Cell. Neurosci, vol.7, p.272, 2013.

L. P. De-almeida, C. A. Ross, D. Zala, P. Aebischer, and N. Deglon, , 2002.

, Lentiviral-mediated delivery of mutant huntingtin in the striatum of rats induces a selective neuropathology modulated by polyglutamine repeat size, huntingtin expression levels, and protein length, J. Neurosci, vol.22, pp.3473-3483

R. A. De-graaf, D. L. Rothman, and K. L. Behar, State of the art direct 13C and indirect 1H-[13C] NMR spectroscopy in vivo. A practical guide, NMR Biomed, vol.24, pp.958-972, 2011.

A. Delekate, M. Fuchtemeier, T. Schumacher, C. Ulbrich, M. Foddis et al., Metabotropic P2Y1 receptor signalling mediates astrocytic hyperactivity in vivo in an Alzheimer's disease mouse model, Nat. Commun, vol.5, p.5422, 2014.

L. Dimou and M. Gotz, Glial cells as progenitors and stem cells: new roles in the healthy and diseased brain, Physiol. Rev, vol.94, pp.709-737, 2014.

J. Doherty, A. E. Sheehan, R. Bradshaw, A. N. Fox, T. Y. Lu et al., PI3K signaling and Stat92E converge to modulate glial responsiveness to axonal injury, PLoS Biol, vol.12, p.1001985, 2014.

V. B. Dorfman, L. Pasquini, M. Riudavets, J. J. Lopez-costa, A. Villegas et al., Differential cerebral deposition of IDE and NEP in sporadic and familial Alzheimer's disease, Neurobiol. Aging, vol.31, pp.1743-1757, 2010.

C. Duyckaerts, M. C. Potier, and B. Delatour, Alzheimer disease models and human neuropathology: similarities and differences, Acta Neuropathol, vol.115, pp.5-38, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00320572

J. G. Emsley and J. D. Macklis, Astroglial heterogeneity closely reflects the neuronal-defined anatomy of the adult murine CNS, Neuron Glia Biol, vol.2, pp.175-186, 2006.

L. F. Eng, R. S. Ghirnikar, and Y. L. Lee, Glial fibrillary acidic protein: GFAP-thirty-one years, Neurochem. Res, vol.25, pp.1439-1451, 1969.

L. F. Eng, J. J. Vanderhaeghen, A. Bignami, and B. Gerstl, An acidic protein isolated from fibrous astrocytes, Brain Res, vol.28, pp.351-354, 1971.

C. Escartin, G. ;. Bonvento, E. Brouillet, P. Gubellini, Y. Trioulier et al., Ciliary neurotrophic factor activates astrocytes, redistributes their glutamate transporters GLAST and GLT-1 to raft microdomains, and improves glutamate handling in vivo, Mol. Neurobiol, vol.38, pp.5978-5989, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00088896

C. Escartin, K. Pierre, A. Colin, E. Brouillet, T. Delzescaux et al., Activation of astrocytes by CNTF induces metabolic plasticity and increases resistance to metabolic insults, J. Neurosci, vol.27, pp.7094-7104, 2007.
URL : https://hal.archives-ouvertes.fr/cea-02290623

C. Escartin and N. Rouach, Astroglial networking contributes to neurometabolic coupling, Front. Neuroenergetics, vol.5, p.4, 2013.
URL : https://hal.archives-ouvertes.fr/cea-02142608

M. Faideau, J. Kim, K. Cormier, R. Gilmore, M. Welch et al., In vivo expression of polyglutamine-expanded huntingtin by mouse striatal astrocytes impairs glutamate transport: a correlation with Huntington's disease subjects, Hum. Mol. Genet, vol.19, pp.3053-3067, 2010.

C. Farina, F. Aloisi, and E. Meinl, Astrocytes are active players in cerebral innate immunity, Trends Immunol, vol.28, pp.138-145, 2007.

A. M. Fernandez, S. Fernandez, P. Carrero, M. Garcia-garcia, and I. Torres-aleman, Calcineurin in reactive astrocytes plays a key role in the interplay between proinflammatory and anti-inflammatory signals, J. Neurosci, vol.27, pp.8745-8756, 2007.

A. M. Fernandez, S. Jimenez, M. Mecha, D. Davila, C. Guaza et al., Regulation of the phosphatase calcineurin by insulin-like growth factor I unveils a key role of astrocytes in Alzheimer's pathology, Mol. Psychiatry, vol.17, pp.705-718, 2012.

L. Ferraiuolo, A. Higginbottom, P. R. Heath, S. Barber, D. Greenald et al., Dysregulation of astrocyte-motoneuron cross-talk in mutant superoxide dismutase 1-related amyotrophic lateral sclerosis, Brain, vol.134, pp.2627-2641, 2011.

I. Ferrer, R. Blanco, M. Carmona, and B. Puig, Phosphorylated mitogenactivated protein kinase (MAPK/ERK-P), protein kinase of 38 kDa (p38-P), stress-activated protein kinase (SAPK/JNK-P), and calcium/calmodulindependent kinase II (CaM kinase II) are differentially expressed in tau deposits in neurons and glial cells in tauopathies, J. Neural Transm, vol.108, pp.1397-1415, 2001.

I. Ferrer, E. Marti, E. Lopez, and A. Tortosa, NF-kB immunoreactivity is observed in association with beta A4 diffuse plaques in patients with Alzheimer's disease, Neuropathol. Appl. Neurobiol, vol.24, pp.271-277, 1998.

L. C. Foo, N. J. Allen, E. A. Bushong, P. B. Ventura, W. S. Chung et al., Development of a method for the purification and culture of rodent astrocytes, Neuron, vol.71, pp.799-811, 2011.

L. S. Forno, L. E. Delanney, I. Irwin, D. Di-monte, and J. W. Langston, Astrocytes and Parkinson's disease. Prog, Brain Res, vol.94, pp.429-436, 1992.

A. E. Frakes, L. Ferraiuolo, A. M. Haidet-phillips, L. Schmelzer, L. Braun et al., Microglia induce motor neuron death via the classical NF-kappaB pathway in amyotrophic lateral sclerosis, Neuron, vol.81, pp.1009-1023, 2014.

M. R. Freeman, Drosophila central nervous system Glia, Cold Spring Harb. Perspect Biol, 2015.

H. Fukuyama, M. Ogawa, H. Yamauchi, S. Yamaguchi, J. Kimura et al., Altered cerebral energy metabolism in Alzheimer's disease: a PET study, J. Nucl. Med, vol.35, pp.1-6, 1994.

H. Funato, M. Yoshimura, T. Yamazaki, T. C. Saido, Y. Ito et al., Astrocytes containing amyloid beta-protein (Abeta)-positive granules are associated with Abeta40-positive diffuse plaques in the aged human brain, Am. J. Pathol, vol.152, pp.983-992, 1998.

J. L. Furman and C. M. Norris, Calcineurin and glial signaling: neuroinflammation and beyond, J. Neuroinflammation, vol.11, p.158, 2014.

J. L. Furman, D. M. Sama, J. C. Gant, T. L. Beckett, M. P. Murphy et al., Targeting astrocytes ameliorates neurologic changes in a mouse model of Alzheimer's disease, J. Neurosci, vol.32, pp.16129-16140, 2012.

L. Gan, M. R. Vargas, D. A. Johnson, J. , and J. A. , Astrocyte-specific overexpression of Nrf2 delays motor pathology and synuclein aggregation throughout the CNS in the alpha-synuclein mutant (A53T) mouse model, 2012.

, J. Neurosci, vol.32, pp.17775-17787

C. Genoud, C. Quairiaux, P. Steiner, H. Hirling, E. Welker et al., Plasticity of astrocytic coverage and glutamate transporter expression in adult mouse cortex, PLoS Biol, vol.4, p.343, 2006.

S. Ghavami, S. Shojaei, B. Yeganeh, S. R. Ande, J. R. Jangamreddy et al., Autophagy and apoptosis dysfunction in neurodegenerative disorders, Prog. Neurobiol, vol.112, pp.24-49, 2014.

C. Giaume, A. Koulakoff, L. Roux, D. Holcman, and N. Rouach, Astroglial networks: a step further in neuroglial and gliovascular interactions, Nat. Rev. Neurosci, vol.11, pp.87-99, 2010.

Y. H. Gong, A. S. Parsadanian, A. Andreeva, W. D. Snider, and J. L. Elliott, Restricted expression of G86R Cu/Zn superoxide dismutase in astrocytes results in astrocytosis but does not cause motoneuron degeneration, 2000.

, J. Neurosci, vol.20, pp.660-665

J. Gotz and L. M. Ittner, Animal models of Alzheimer's disease and frontotemporal dementia, Nat. Rev. Neurosci, vol.9, pp.532-544, 2008.

S. T. Grafton, J. C. Mazziotta, J. J. Pahl, P. St-george-hyslop, J. L. Haines et al., Serial changes of cerebral glucose metabolism and caudate size in persons at risk for Huntington's disease, Arch. Neurol, vol.49, pp.1161-1167, 1992.

M. Gray, D. I. Shirasaki, C. Cepeda, V. M. Andre, B. Wilburn et al., Full-length human mutant huntingtin with a stable polyglutamine repeat can elicit progressive and selective neuropathogenesis in BACHD mice, J. Neurosci, vol.28, pp.6182-6195, 2008.

S. I. Grivennikov, K. , and M. , Dangerous liaisons: STAT3 and NF-kappaB collaboration and crosstalk in cancer, Cytokine Growth Factor Rev, vol.21, pp.11-19, 2010.

X. L. Gu, C. X. Long, L. Sun, C. Xie, X. Lin et al., Astrocytic expression of Parkinson's disease-related A53T alpha-synuclein causes neurodegeneration in mice, Mol. Brain, vol.3, p.12, 2010.

M. E. Gurney, H. Pu, A. Y. Chiu, M. C. Dal-canto, C. Y. Polchow et al., Motor neuron degeneration in mice that express a human Cu,Zn superoxide dismutase mutation, Science, vol.264, pp.1772-1775, 1994.

A. M. Haidet-phillips, M. E. Hester, C. J. Miranda, K. Meyer, L. Braun et al., Astrocytes from familial and sporadic ALS patients are toxic to motor neurons, Nat. Biotechnol, vol.29, pp.824-828, 2011.

E. D. Hall, J. A. Oostveen, and M. E. Gurney, Relationship of microglial and astrocytic activation to disease onset and progression in a transgenic model of familial ALS, Glia, vol.23, pp.249-256, 1998.

A. Halle, V. Hornung, G. C. Petzold, C. R. Stewart, B. G. Monks et al., The NALP3 inflammasome is involved in the innate immune response to amyloid-beta, Nat. Immunol, vol.9, pp.857-865, 2008.

U. K. Hanisch and H. Kettenmann, Microglia: active sensor and versatile effector cells in the normal and pathologic brain, Nat. Neurosci, vol.10, pp.1387-1394, 2007.

B. Hassel, S. Tessler, R. L. Faull, and P. C. Emson, Glutamate uptake is reduced in prefrontal cortex in Huntington's disease, Neurochem. Res, vol.33, pp.232-237, 2008.

F. He, W. Ge, K. Martinowich, S. Becker-catania, V. Coskun et al., A positive autoregulatory loop of Jak-STAT signaling controls the onset of astrogliogenesis, Nat. Neurosci, vol.8, pp.616-625, 2005.

R. E. Heikkila, A. Hess, and R. C. Duvoisin, Dopaminergic neurotoxicity of 1-methyl-4-phenyl-1,2,5,6-tetrahydropyridine in mice, Science, vol.224, pp.1451-1453, 1984.

M. T. Heneka, M. P. Kummer, and E. Latz, Innate immune activation in neurodegenerative disease, Nat. Rev. Immunol, vol.14, pp.463-477, 2014.

M. T. Heneka, M. Sastre, L. Dumitrescu-ozimek, I. Dewachter, J. Walter et al., Focal glial activation coincides with increased BACE1 activation and precedes amyloid plaque deposition in APP[V717I] transgenic mice, J. Neuroinflammation, vol.2, p.22, 2005.

K. Hensley, R. A. Floyd, N. Y. Zheng, R. Nael, K. A. Robinson et al., p38 kinase is activated in the Alzheimer's disease brain, J. Neurochem, vol.72, pp.2053-2058, 1999.

J. E. Herrmann, T. Imura, B. Song, J. Qi, Y. Ao et al., STAT3 is a critical regulator of astrogliosis and scar formation after spinal cord injury, J. Neurosci, vol.28, pp.7231-7243, 2008.

E. C. Hirsch and S. Hunot, Neuroinflammation in Parkinson's disease: a target for neuroprotection?, Lancet Neurol, vol.8, pp.70062-70068, 2009.

B. Hoesel and J. A. Schmid, The complexity of NF-kappaB signaling in inflammation and cancer, Mol. Cancer, vol.12, p.86, 2013.

P. G. Hogan, L. Chen, J. Nardone, and A. Rao, Transcriptional regulation by calcium, calcineurin, and NFAT, Genes Dev, vol.17, pp.2205-2232, 2003.

D. S. Howland, J. Liu, Y. She, B. Goad, N. J. Maragakis et al., Focal loss of the glutamate transporter EAAT2 in a transgenic rat model of SOD1 mutant-mediated amyotrophic lateral sclerosis (ALS), Proc. Natl. Acad. Sci. U.S.A, vol.99, pp.1604-1609, 2002.

H. Y. Hsiao, Y. C. Chen, H. M. Chen, P. H. Tu, and Y. Chern, A critical role of astrocyte-mediated nuclear factor-kappaB-dependent inflammation in Huntington's disease, Hum. Mol. Genet, vol.22, pp.1826-1842, 2013.

K. Hsiao, P. Chapman, S. Nilsen, C. Eckman, Y. Harigaya et al., Correlative memory deficits, Abeta elevation, and amyloid plaques in transgenic mice, Science, vol.274, pp.99-102, 1996.

Y. Huang and L. Mucke, Alzheimer mechanisms and therapeutic strategies, Cell, vol.148, pp.1204-1222, 2012.

S. Hunot, B. Brugg, D. Ricard, P. P. Michel, M. P. Muriel et al., Nuclear translocation of NF-kappaB is increased in dopaminergic neurons of patients with parkinson disease, Proc. Natl. Acad. Sci. U.S.A, vol.94, pp.7531-7536, 1997.

A. P. Hutchins, D. Diez, Y. Takahashi, S. Ahmad, R. Jauch et al., Distinct transcriptional regulatory modules underlie STAT3 s cell typeindependent and cell type-specific functions, Nucleic Acids Res, vol.41, pp.2155-2170, 2013.

E. R. Hutchison, E. M. Kawamoto, D. D. Taub, A. Lal, K. Abdelmohsen et al., Evidence for miR-181 involvement in neuroinflammatory responses of astrocytes, Glia, vol.61, pp.1018-1028, 2013.

N. Jain, T. Zhang, S. L. Fong, C. P. Lim, and X. Cao, Repression of Stat3 activity by activation of mitogen-activated protein kinase (MAPK), Oncogene, vol.17, pp.3157-3167, 1998.

J. L. Jankowsky, D. J. Fadale, J. Anderson, G. M. Xu, V. Gonzales et al., Mutant presenilins specifically elevate the levels of the 42 residue beta-amyloid peptide in vivo: evidence for augmentation of a 42-specific gamma secretase, Hum. Mol. Genet, vol.13, pp.159-170, 2004.

A. H. Jansen, E. A. Reits, and E. M. Hol, The ubiquitin proteasome system in glia and its role in neurodegenerative diseases, Front. Mol. Neurosci, vol.7, p.73, 2014.

K. L. Jeffrey, M. Camps, C. Rommel, and C. R. Mackay, Targeting dualspecificity phosphatases: manipulating MAP kinase signalling and immune responses, Nat. Rev. Drug Discov, vol.6, pp.391-403, 2007.

S. Jo, O. Yarishkin, Y. J. Hwang, Y. E. Chun, M. Park et al., GABA from reactive astrocytes impairs memory in mouse models of Alzheimer's disease, Nat. Med, vol.20, pp.886-896, 2014.

A. Johansson, H. Engler, G. Blomquist, B. Scott, A. Wall et al., Evidence for astrocytosis in ALS demonstrated by [11C](L)-deprenyl-D2 PET, J. Neurol. Sci, vol.255, pp.17-22, 2007.

E. S. Jung, K. An, H. S. Hong, J. H. Kim, M. et al., Astrocyteoriginated ATP protects Abeta(1-42)-induced impairment of synaptic plasticity, J. Neurosci, vol.32, pp.3081-3087, 2012.

M. Kaiser, I. Maletzki, S. Hulsmann, B. Holtmann, W. Schulz-schaeffer et al., Progressive loss of a glial potassium channel (KCNJ10) in the spinal cord of the SOD1 (G93A) transgenic mouse model of amyotrophic lateral sclerosis, J. Neurochem, vol.99, pp.900-912, 2006.

B. Kaltschmidt, M. Uherek, B. Volk, P. A. Baeuerle, and C. Kaltschmidt, Transcription factor NF-kappaB is activated in primary neurons by amyloid beta peptides and in neurons surrounding early plaques from patients with Alzheimer disease, Proc. Natl. Acad. Sci. U.S.A, vol.94, pp.2642-2647, 1997.

B. Kaltschmidt, D. Widera, and C. Kaltschmidt, Signaling via NF-kappaB in the nervous system, Biochim. Biophys. Acta, vol.1745, pp.287-299, 2005.

B. Kaminska, A. Gozdz, M. Zawadzka, A. Ellert-miklaszewska, and M. Lipko, MAPK signal transduction underlying brain inflammation and gliosis as therapeutic target, Anat. Rec, vol.292, pp.1902-1913, 2009.

W. Kamphuis, L. Kooijman, M. Orre, O. Stassen, M. Pekny et al., GFAP and vimentin deficiency alters gene expression in astrocytes and microglia in wild-type mice and changes the transcriptional response of reactive glia in mouse model for Alzheimer's disease, Glia, vol.63, pp.1036-1056, 2015.

W. Kamphuis, M. Orre, L. Kooijman, M. Dahmen, and E. M. Hol, Differential cell proliferation in the cortex of the APPswePS1dE9 Alzheimer's disease mouse model, Glia, vol.60, pp.615-629, 2012.

W. Kang, F. Balordi, N. Su, L. Chen, G. Fishell et al., Astrocyte activation is suppressed in both normal and injured brain by FGF signaling, Proc. Natl. Acad. Sci. U.S.A, vol.111, 2014.

R. Kanski, M. E. Van-strien, P. Van-tijn, and E. M. Hol, A star is born: new insights into the mechanism of astrogenesis, Cell. Mol. Life Sci, vol.71, pp.433-447, 2014.

H. Kawamata, S. K. Ng, N. Diaz, S. Burstein, L. Morel et al., Abnormal intracellular calcium signaling and SNARE-dependent exocytosis contributes to SOD1G93A astrocyte-mediated toxicity in amyotrophic lateral sclerosis, J. Neurosci, vol.34, pp.2331-2348, 2014.

N. J. Kershaw, A. Laktyushin, N. A. Nicola, and J. J. Babon, Reconstruction of an active SOCS3-based E3 ubiquitin ligase complex in vitro: identification of the active components and JAK2 and gp130 as substrates, Growth Factors, vol.32, pp.1-10, 2014.

H. Kettenmann and B. R. Ransom, The concept of neuroglia: a historical perspective, pp.1-16, 2004.

A. Khoshnan, J. Ko, E. E. Watkin, L. A. Paige, P. H. Reinhart et al., Activation of the IkappaB kinase complex and nuclear factor-kappaB contributes to mutant huntingtin neurotoxicity, J. Neurosci, vol.24, pp.7999-8008, 2004.

K. A. Kigerl, . De-rivero, J. P. Vaccari, W. D. Dietrich, P. G. Popovich et al., Pattern recognition receptors and central nervous system repair, Exp. Neurol, vol.258, pp.5-16, 2014.

M. Kohutnicka, E. Lewandowska, I. Kurkowska-jastrzebska, A. Czlonkowski, and A. Czlonkowska, Microglial and astrocytic involvement in a murine model of Parkinson's disease induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), Immunopharmacology, vol.39, pp.167-180, 1998.

M. Koistinaho, M. I. Kettunen, G. Goldsteins, R. Keinanen, A. Salminen et al., Beta-amyloid precursor protein transgenic mice that harbor diffuse A beta deposits but do not form plaques show increased ischemic vulnerability: role of inflammation, Proc. Natl. Acad. Sci. U.S.A, vol.99, pp.1610-1615, 2002.

M. Koistinaho, S. Lin, X. Wu, M. Esterman, D. Koger et al.,

, Apolipoprotein E promotes astrocyte colocalization and degradation of deposited amyloid-beta peptides, Nat. Med, vol.10, pp.719-726

A. W. Kraft, X. Hu, H. Yoon, P. Yan, Q. Xiao et al., Attenuating astrocyte activation accelerates plaque pathogenesis in APP/PS1 mice, FASEB J, vol.27, pp.187-198, 2013.

R. Krencik and E. M. Ullian, A cellular star atlas: using astrocytes from human pluripotent stem cells for disease studies, Front. Cell. Neurosci, vol.7, p.25, 2013.

K. V. Kuchibhotla, C. R. Lattarulo, B. T. Hyman, and B. J. Bacskai, Synchronous hyperactivity and intercellular calcium waves in astrocytes in Alzheimer mice, Science, vol.323, pp.1211-1215, 2009.

Y. Kuge, H. Kawashima, K. Minematsu, Y. Hasegawa, T. Yamaguchi et al., Octanoate as a PET tracer for studying ischemic stroke: evaluation in a canine model of thromboembolic stroke with positron emission tomography, Biol. Pharm. Bull, vol.23, pp.984-988, 2000.

C. M. Lauderback, J. M. Hackett, F. F. Huang, J. N. Keller, L. I. Szweda et al., The glial glutamate transporter, GLT-1, is oxidatively modified by 4-hydroxy-2-nonenal in the Alzheimer's disease brain: the role of Abeta1-42, J. Neurochem, vol.78, pp.413-416, 2001.

S. Lavisse, M. Guillermier, A. S. Herard, F. Petit, M. Delahaye et al., Reactive Astrocytes Overexpress TSPO and are detected by TSPO positron emission tomography imaging, J. Neurosci, vol.32, pp.10809-10818, 2012.
URL : https://hal.archives-ouvertes.fr/hal-02110993

L. Prince, G. Delaere, P. Fages, C. Lefrancois, T. Touret et al., Glutamine synthetase (GS) expression is reduced in senile dementia of the Alzheimer type, Neurochem. Res, vol.20, pp.859-862, 1995.

V. Lebon, K. F. Petersen, G. W. Cline, J. Shen, G. F. Mason et al., Astroglial contribution to brain energy metabolism in humans revealed by 13C nuclear magnetic resonance spectroscopy: elucidation of the dominant pathway for neurotransmitter glutamate repletion and measurement of astrocytic oxidative metabolism, J. Neurosci, vol.22, pp.1523-1531, 2002.

W. Lee, R. C. Reyes, M. K. Gottipati, K. Lewis, M. Lesort et al., Enhanced Ca(2+)-dependent glutamate release from astrocytes of the BACHD Huntington's disease mouse model, Neurobiol. Dis, vol.58, pp.192-199, 2013.

V. Leoni and C. Caccia, Study of cholesterol metabolism in Huntington's disease, Biochem. Biophys. Res. Commun, vol.446, pp.697-701, 2014.

A. C. Lepore, C. Dejea, J. Carmen, B. Rauck, D. A. Kerr et al., Selective ablation of proliferating astrocytes does not affect disease outcome in either acute or chronic models of motor neuron degeneration, Exp. Neurol, vol.211, pp.423-432, 2008.

A. C. Lepore, B. Rauck, C. Dejea, A. C. Pardo, M. S. Rao et al., Focal transplantation-based astrocyte replacement is neuroprotective in a model of motor neuron disease, Nat. Neurosci, vol.11, pp.1294-1301, 2008.

D. E. Levy and J. E. Darnell, Stats: transcriptional control and biological impact, Nat. Rev. Mol. Cell Biol, vol.3, pp.651-662, 2002.

Y. Y. Li, J. G. Cui, J. M. Hill, S. Bhattacharjee, Y. Zhao et al., Increased expression of miRNA-146a in Alzheimer's disease transgenic mouse models, Neurosci. Lett, vol.487, pp.94-98, 2011.

H. Lian, L. Yang, A. Cole, L. Sun, A. C. Chiang et al., NFkappaB-Activated astroglial release of complement C3 compromises neuronal morphology and function associated with Alzheimer's Disease, Neuron, vol.85, pp.101-115, 2015.

C. M. Liberto, P. J. Albrecht, L. M. Herx, V. W. Yong, and S. W. Levison, Pro-regenerative properties of cytokine-activated astrocytes, J. Neurochem, vol.89, pp.1092-1100, 2004.

J. C. Lievens, T. Rival, M. Iche, H. Chneiweiss, and S. Birman, Expanded polyglutamine peptides disrupt EGF receptor signaling and glutamate transporter expression in Drosophila, Hum. Mol. Genet, vol.14, pp.713-724, 2005.
URL : https://hal.archives-ouvertes.fr/hal-00118469

J. C. Lievens, B. Woodman, A. Mahal, O. Spasic-boscovic, D. Samuel et al., Impaired glutamate uptake in the R6 Huntington's disease transgenic mice, Neurobiol. Dis, vol.8, pp.807-821, 2001.

D. Lim, A. Iyer, V. Ronco, A. A. Grolla, P. L. Canonico et al., , 2013.

, Amyloid beta deregulates astroglial mGluR5-mediated calcium signaling via calcineurin and Nf-kB, Glia, vol.61, pp.1134-1145

M. T. Lin and M. F. Beal, Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases, Nature, vol.443, pp.787-795, 2006.

K. M. Lucin and T. Wyss-coray, Immune activation in brain aging and neurodegeneration: too much or too little?, Neuron, vol.64, pp.110-122, 2009.

L. Mangiarini, K. Sathasivam, A. Mahal, R. Mott, M. Seller et al., Instability of highly expanded CAG repeats in mice transgenic for the Huntington's disease mutation, Nat. Genet, vol.15, pp.297-197, 1997.

N. J. Maragakis and J. D. Rothstein, Glutamate transporters: animal models to neurologic disease, Neurobiol. Dis, vol.15, pp.461-473, 2004.

N. J. Maragakis and J. D. Rothstein, Mechanisms of Disease: astrocytes in neurodegenerative disease, Nat. Clin. Pract. Neurol, vol.2, pp.679-689, 2006.

E. Marcora and M. B. Kennedy, The Huntington's disease mutation impairs Huntingtin's role in the transport of NF-kappaB from the synapse to the nucleus, Hum. Mol. Genet, vol.19, pp.4373-4384, 2010.

J. Margulis and S. Finkbeiner, Proteostasis in striatal cells and selective neurodegeneration in Huntington's disease. Front, Cell. Neurosci, vol.8, p.218, 2014.

J. Marik, A. Ogasawara, B. Martin-mcnulty, J. Ross, J. E. Flores et al., PET of glial metabolism using 2-18F-fluoroacetate, J. Nucl. Med, vol.50, pp.982-990, 2009.

F. Martorana, L. Brambilla, C. F. Valori, C. Bergamaschi, C. Roncoroni et al., The BH4 domain of Bcl-X(L) rescues astrocyte degeneration in amyotrophic lateral sclerosis by modulating intracellular calcium signals, Hum. Mol. Genet, vol.21, pp.826-840, 2012.

E. Masliah, M. Alford, R. Deteresa, M. Mallory, and L. Hansen, Deficient glutamate transport is associated with neurodegeneration in Alzheimer's disease, Ann. Neurol, vol.40, pp.759-766, 1996.

E. Masliah, M. Alford, M. Mallory, E. Rockenstein, D. Moechars et al., Abnormal glutamate transport function in mutant amyloid precursor protein transgenic mice, Exp. Neurol, vol.163, pp.381-387, 2000.

M. P. Mattson and M. K. Meffert, Roles for NF-kappaB in nerve cell survival, plasticity, and disease, Cell Death Differ, vol.13, pp.852-860, 2006.

V. Matyash and H. Kettenmann, Heterogeneity in astrocyte morphology and physiology, Brain Res. Rev, vol.63, 2010.

K. D. Mccarthy, D. Vellis, and J. , Preparation of separate astroglial and oligodendroglial cell cultures from rat cerebral tissue, J. Cell Biol, vol.85, pp.890-902, 1980.

X. Mei, P. Ezan, C. Giaume, and A. Koulakoff, Astroglial connexin immunoreactivity is specifically altered at beta-amyloid plaques in betaamyloid precursor protein/presenilin1 mice, Neuroscience, vol.171, pp.92-105, 2010.

L. B. Menalled, A. E. Kudwa, S. Miller, J. Fitzpatrick, J. Watson-johnson et al., Comprehensive behavioral and molecular characterization of a new knock-in mouse model of Huntington's disease: zQ175, PLoS ONE, vol.7, p.49838, 2012.

L. B. Menalled, J. D. Sison, I. Dragatsis, S. Zeitlin, and M. F. Chesselet, Time course of early motor and neuropathological anomalies in a knock-in mouse model of Huntington's disease with 140 CAG repeats, J. Comp. Neurol, vol.465, pp.11-26, 2003.

A. Migheli, R. Piva, C. Atzori, D. Troost, and D. Schiffer, c-Jun, JNK/SAPK kinases and transcription factor NF-kappa B are selectively activated in astrocytes, but not motor neurons, 1997.

, J. Neuropathol. Exp. Neurol, vol.56, pp.1314-1322

B. R. Miller, J. L. Dorner, M. Shou, Y. Sari, S. J. Barton et al., Up-regulation of GLT1 expression increases glutamate uptake and attenuates the Huntington's disease phenotype in the R6/2 mouse, Neuroscience, vol.153, pp.329-337, 2008.

J. Minkiewicz, . De-rivero, J. P. Vaccari, and R. W. Keane, Human astrocytes express a novel NLRP2 inflammasome, Glia, vol.61, pp.1113-1121, 2013.

A. V. Molofsky, R. Krencik, E. M. Ullian, H. H. Tsai, B. Deneen et al., Astrocytes and disease: a neurodevelopmental perspective, Genes Dev, vol.26, pp.891-907, 2012.

E. Motori, J. Puyal, N. Toni, A. Ghanem, C. Angeloni et al., Inflammation-induced alteration of astrocyte mitochondrial dynamics requires autophagy for mitochondrial network maintenance, Cell Metab, vol.18, pp.844-859, 2013.

H. W. Muller, U. Junghans, and J. Kappler, Astroglial neurotrophic and neurite-promoting factors, Pharmacol. Ther, vol.65, pp.47-54, 1995.

R. G. Nagele, M. R. D'andrea, H. Lee, V. Venkataraman, and H. Y. Wang, Astrocytes accumulate A beta 42 and give rise to astrocytic amyloid plaques in Alzheimer disease brains, Brain Res, vol.971, issue.03, pp.2361-2369, 2003.

J. I. Nagy, W. Li, E. L. Hertzberg, and C. A. Marotta, Elevated connexin43 immunoreactivity at sites of amyloid plaques in Alzheimer's disease, Brain Res, vol.717, pp.173-178, 1996.

L. H. Nilsen, C. Rae, L. M. Ittner, J. Gotz, and U. Sonnewald, Glutamate metabolism is impaired in transgenic mice with tau hyperphosphorylation, 2013.

J. Cereb, Blood Flow Metab, vol.33, pp.684-691

L. H. Nilsen, M. P. Witter, and U. Sonnewald, Neuronal and astrocytic metabolism in a transgenic rat model of Alzheimer's disease, J. Cereb. Blood Flow Metab, vol.34, pp.906-914, 2014.

C. M. Norris, I. Kadish, E. M. Blalock, K. C. Chen, V. Thibault et al., Calcineurin triggers reactive/inflammatory processes in astrocytes and is upregulated in aging and Alzheimer's models, J. Neurosci, vol.25, pp.4649-4658, 2005.

N. A. Oberheim, G. F. Tian, X. Han, W. Peng, T. Takano et al., Loss of astrocytic domain organization in the epileptic brain, J. Neurosci, vol.28, pp.3264-3276, 2008.

E. R. O'brien, C. Howarth, and N. R. Sibson, The role of astrocytes in CNS tumors: pre-clinical models and novel imaging approaches, Front. Cell Neurosci, vol.7, p.40, 2013.

S. Oddo, A. Caccamo, J. D. Shepherd, M. P. Murphy, T. E. Golde et al., Triple-transgenic model of Alzheimer's disease with plaques and tangles: intracellular Abeta and synaptic dysfunction, Neuron, vol.39, pp.409-421, 2003.

A. Oeckinghaus, M. S. Hayden, and S. Ghosh, Crosstalk in NF-kappaB signaling pathways, Nat. Immunol, vol.12, pp.695-708, 2011.

S. Okada, M. Nakamura, H. Katoh, T. Miyao, T. Shimazaki et al., Conditional ablation of Stat3 or Socs3 discloses a dual role for reactive astrocytes after spinal cord injury, Nat. Med, vol.12, pp.829-834, 2006.

M. Olabarria, H. N. Noristani, A. Verkhratsky, and J. J. Rodriguez, , 2010.

, Concomitant astroglial atrophy and astrogliosis in a triple transgenic animal model of Alzheimer's disease, Glia, vol.58, pp.831-838

M. Olabarria, H. N. Noristani, A. Verkhratsky, and J. J. Rodriguez, Agedependent decrease in glutamine synthetase expression in the hippocampal astroglia of the triple transgenic Alzheimer's disease mouse model: mechanism for deficient glutamatergic transmission?, Mol. Neurodegener, vol.6, p.55, 2011.

S. H. Oliet, R. Piet, and D. A. Poulain, Control of glutamate clearance and synaptic efficacy by glial coverage of neurons, Science, vol.292, pp.923-926, 2001.
URL : https://hal.archives-ouvertes.fr/inserm-00000059

J. A. Orellana, K. F. Shoji, V. Abudara, P. Ezan, E. Amigou et al., Amyloid beta-induced death in neurons involves glial and neuronal hemichannels, J. Neurosci, vol.31, pp.4962-4977, 2011.

M. Orre, W. Kamphuis, S. Dooves, L. Kooijman, E. T. Chan et al., Reactive glia show increased immunoproteasome activity in Alzheimer's disease, Brain, vol.136, pp.1415-1431, 2013.

M. Orre, W. Kamphuis, L. M. Osborn, A. H. Jansen, L. Kooijman et al., Isolation of glia from Alzheimer's mice reveals inflammation and dysfunction, Neurobiol. Aging, vol.35, pp.2746-2760, 2014.

P. I. Ortinski, J. Dong, A. Mungenast, C. Yue, H. Takano et al., Selective induction of astrocytic gliosis generates deficits in neuronal inhibition, Nat. Neurosci, vol.13, pp.584-591, 2010.

J. B. Owen, F. Di-domenico, R. Sultana, M. Perluigi, C. Cini et al., Proteomics-determined differences in the concanavalin-Afractionated proteome of hippocampus and inferior parietal lobule in subjects with Alzheimer's disease and mild cognitive impairment: implications for progression of AD, J. Proteome Res, vol.8, pp.471-482, 2009.

A. Panatier, M. Arizono, and U. V. Nagerl, Dissecting tripartite synapses with STED microscopy, Philos. Trans. R. Soc. Lond. B Biol. Sci, vol.369, 2014.

M. M. Pearce, E. J. Spartz, W. Hong, L. Luo, and R. R. Kopito, Prionlike transmission of neuronal huntingtin aggregates to phagocytic glia in the Drosophila brain, Nat. Commun, vol.6, p.6768, 2015.

M. Pehar, P. Cassina, M. R. Vargas, R. Castellanos, L. Viera et al., Astrocytic production of nerve growth factor in motor neuron apoptosis: implications for amyotrophic lateral sclerosis, J. Neurochem, vol.89, pp.464-473, 2004.

L. Pellerin and P. J. Magistretti, Glutamate uptake into astrocytes stimulates aerobic glycolysis: a mechanism coupling neuronal activity to glucose utilization, Proc. Natl. Acad. Sci. U.S.A, vol.91, pp.10625-10629, 1994.

F. W. Pfrieger and N. Ungerer, Cholesterol metabolism in neurons and astrocytes, Prog. Lipid Res, vol.50, pp.357-371, 2011.

T. Philips and W. Robberecht, Neuroinflammation in amyotrophic lateral sclerosis: role of glial activation in motor neuron disease, Lancet Neurol, vol.10, pp.253-263, 2011.

T. M. Pirttimaki, N. K. Codadu, A. Awni, P. Pratik, D. A. Nagel et al., alpha7 Nicotinic receptor-mediated astrocytic gliotransmitter release: Abeta effects in a preclinical Alzheimer's mouse model, PLoS ONE, vol.8, p.81828, 2013.

S. Pons and I. Torres-aleman, Insulin-like growth factor-I stimulates dephosphorylation of ikappa B through the serine phosphatase calcineurin (protein phosphatase 2B), J. Biol. Chem, vol.275, pp.38620-38625, 2000.

D. Popovic, D. Vucic, and I. Dikic, Ubiquitination in disease pathogenesis and treatment, Nat. Med, vol.20, pp.1242-1253, 2014.

T. B. Puschmann, C. Zanden, Y. De-pablo, F. Kirchhoff, M. Pekna et al., Bioactive 3D cell culture system minimizes cellular stress and maintains the in vivo-like morphological complexity of astroglial cells, Glia, vol.61, pp.432-440, 2013.

K. Puttaparthi and J. L. Elliott, Non-neuronal induction of immunoproteasome subunits in an ALS model: possible mediation by cytokines, Exp. Neurol, vol.196, pp.441-451, 2005.

K. Puttaparthi, L. Van-kaer, and J. L. Elliott, Assessing the role of immunoproteasomes in a mouse model of familial ALS, Exp. Neurol, vol.206, pp.53-58, 2007.

H. W. Querfurth and F. M. Laferla, Alzheimer's disease, N. Engl. J. Med, vol.362, pp.329-344, 2010.

R. Radde, T. Bolmont, S. A. Kaeser, J. Coomaraswamy, D. Lindau et al., Abeta42-driven cerebral amyloidosis in transgenic mice reveals early and robust pathology, EMBO Rep, vol.7, pp.940-946, 2006.

G. V. Rebec, Dysregulation of corticostriatal ascorbate release and glutamate uptake in transgenic models of Huntington's disease, Antioxid. Redox Signal, vol.19, pp.2115-2128, 2013.

G. V. Rebec, S. J. Barton, and M. D. Ennis, Dysregulation of ascorbate release in the striatum of behaving mice expressing the Huntington's disease gene, J Neurosci, vol.22, p.202, 2002.

W. Robberecht and T. Philips, The changing scene of amyotrophic lateral sclerosis, Nat. Rev. Neurosci, vol.14, pp.248-264, 2013.

S. Robel, S. C. Buckingham, J. L. Boni, S. L. Campbell, N. C. Danbolt et al., Reactive astrogliosis causes the development of spontaneous seizures, J. Neurosci, vol.35, pp.3330-3345, 2015.

S. Robel, T. Mori, S. Zoubaa, J. Schlegel, S. Sirko et al., Conditional deletion of beta1-integrin in astroglia causes partial reactive gliosis, Glia, vol.57, pp.1630-1647, 2009.

F. Rojas, N. Cortes, S. Abarzua, A. Dyrda, and B. Van-zundert, Astrocytes expressing mutant SOD1 and TDP43 trigger motoneuron death that is mediated via sodium channels and nitroxidative stress, Front. Cell. Neurosci, vol.8, p.24, 2014.

B. Ross, A. Lin, K. Harris, P. Bhattacharya, and B. Schweinsburg, Clinical experience with 13C MRS in vivo, NMR Biomed, vol.16, pp.358-369, 2003.

C. A. Ross and M. A. Poirier, Protein aggregation and neurodegenerative disease, Nat. Med, vol.10, pp.10-17, 2004.

D. Rossi, L. Brambilla, C. F. Valori, A. Crugnola, G. Giaccone et al., Defective tumor necrosis factor-alpha-dependent control of astrocyte glutamate release in a transgenic mouse model of Alzheimer disease, J. Biol. Chem, vol.280, pp.42088-42096, 2005.

J. D. Rothstein, L. J. Martin, and R. W. Kuncl, Decreased glutamate transport by the brain and spinal cord in amyotrophic lateral sclerosis, N. Engl. J. Med, vol.326, pp.1464-1468, 1992.

J. D. Rothstein, S. Patel, M. R. Regan, C. Haenggeli, Y. H. Huang et al., Beta-lactam antibiotics offer neuroprotection by increasing glutamate transporter expression, Nature, vol.433, pp.73-77, 2005.

J. D. Rothstein, M. Van-kammen, A. I. Levey, L. J. Martin, and R. W. Kuncl, Selective loss of glial glutamate transporter GLT-1 in amyotrophic lateral sclerosis, Ann. Neurol, vol.38, pp.73-84, 1995.

N. Rouach, A. Koulakoff, V. Abudara, K. Willecke, and C. Giaume, , 2008.

, Astroglial metabolic networks sustain hippocampal synaptic transmission, Science, vol.322, pp.1551-1555

M. Rufer, S. B. Wirth, A. Hofer, R. Dermietzel, A. Pastor et al., Regulation of connexin-43, GFAP, and FGF-2 is not accompanied by changes in astroglial coupling in MPTP-lesioned, FGF-2-treated parkinsonian mice, J. Neurosci. Res, vol.46, pp.606-617, 1996.

H. Sancheti, I. Patil, K. Kanamori, R. Diaz-brinton, W. Zhang et al., Hypermetabolic state in the 7-month-old triple transgenic mouse model of Alzheimer's disease and the effect of lipoic acid, p.13, 2014.

J. Cereb, Blood Flow Metab, vol.34, pp.1749-1760

M. Santello and A. Volterra, TNFalpha in synaptic function: switching gears, Trends Neurosci, vol.35, pp.638-647, 2012.

A. Scimemi, J. S. Meabon, R. L. Woltjer, J. M. Sullivan, J. S. Diamond et al., Amyloid-beta1-42 slows clearance of synaptically released glutamate by mislocalizing astrocytic GLT-1, J. Neurosci, vol.33, pp.5312-5318, 2013.

H. A. Scott, F. M. Gebhardt, A. D. Mitrovic, R. J. Vandenberg, and P. R. Dodd, Glutamate transporter variants reduce glutamate uptake in Alzheimer's disease, Neurobiol. Aging, vol.32, pp.1-11, 2011.

J. L. Seidel, M. Faideau, I. Aiba, U. Pannasch, C. Escartin et al., Ciliary neurotrophic factor (CNTF) activation of astrocytes decreases spreading depolarization susceptibility and increases potassium clearance, Glia, vol.63, pp.91-103, 2014.
URL : https://hal.archives-ouvertes.fr/cea-02168392

S. Sekar, J. Mcdonald, L. Cuyugan, J. Aldrich, A. Kurdoglu et al., Alzheimer's disease is associated with altered expression of genes involved in immune response and mitochondrial processes in astrocytes, Neurobiol. Aging, vol.36, pp.583-591, 2015.

A. Serrano-pozo, T. Gomez-isla, J. H. Growdon, M. P. Frosch, and B. T. Hyman, A phenotypic change but not proliferation underlies glial responses in Alzheimer disease, Am. J. Pathol, vol.182, pp.2332-2344, 2013.

A. Sharif and V. Prevot, Isolation and culture of human astrocytes, Methods Mol. Biol, vol.814, pp.137-151, 2012.

J. G. Sheng, S. Shirabe, N. Nishiyama, and J. P. Schwartz, Alterations in striatal glial fibrillary acidic protein expression in response to 6-hydroxydopamine-induced denervation, Exp. Brain Res, vol.95, pp.450-456, 1993.

N. Shibata, A. Kakita, H. Takahashi, Y. Ihara, K. Nobukuni et al., Activation of signal transducer and activator of transcription-3 in the spinal cord of sporadic amyotrophic lateral sclerosis patients, Neurodegener. Dis, vol.6, pp.118-126, 2009.

N. Shibata, T. Yamamoto, A. Hiroi, Y. Omi, Y. Kato et al., Activation of STAT3 and inhibitory effects of pioglitazone on STAT3 activity in a mouse model of SOD1-mutated amyotrophic lateral sclerosis, Neuropathology, vol.30, pp.353-360, 2010.

K. Shibuki, H. Gomi, L. Chen, S. Bao, J. J. Kim et al., Deficient cerebellar long-term depression, impaired eyeblink conditioning, and normal motor coordination in GFAP mutant mice, Neuron, vol.16, pp.587-599, 1996.

N. R. Sibson, J. P. Lowe, A. M. Blamire, M. J. Martin, T. P. Obrenovitch et al., Acute astrocyte activation in brain detected by MRI: new insights into T(1) hypointensity, J. Cereb. Blood Flow Metab, vol.28, pp.621-632, 2008.

J. E. Simpson, P. G. Ince, G. Lace, G. Forster, P. J. Shaw et al., Astrocyte phenotype in relation to Alzheimer-type pathology in the ageing brain, Neurobiol. Aging, vol.31, pp.578-590, 2010.

J. E. Simpson, P. G. Ince, P. J. Shaw, P. R. Heath, R. Raman et al., Microarray analysis of the astrocyte transcriptome in the aging brain: relationship to Alzheimer's pathology and APOE genotype, Neurobiol. Aging, vol.32, pp.1795-1807, 2011.

S. K. Singhrao, J. W. Neal, B. P. Morgan, and P. Gasque, Increased complement biosynthesis by microglia and complement activation on neurons in Huntington's disease, Exp. Neurol, vol.159, pp.362-376, 1999.

S. Sirko, G. Behrendt, P. A. Johansson, P. Tripathi, M. Costa et al., Reactive glia in the injured brain acquire stem cell properties in response to sonic hedgehog glia, Cell Stem Cell, vol.12, pp.426-439, 2013.

S. A. Sloan and B. A. Barres, Looks can be deceiving: reconsidering the evidence for gliotransmission, Neuron, vol.84, pp.1112-1115, 2014.

M. V. Sofroniew, Molecular dissection of reactive astrogliosis and glial scar formation, Trends Neurosci, vol.32, pp.638-647, 2009.

M. V. Sofroniew, Multiple roles for astrocytes as effectors of cytokines and inflammatory mediators, Neuroscientist, vol.20, pp.160-172, 2014.

M. V. Sofroniew, Astrocyte barriers to neurotoxic inflammation, Nat. Rev. Neurosci, vol.16, pp.249-263, 2015.

M. V. Sofroniew and H. V. Vinters, Astrocytes: biology and pathology, Acta Neuropathol, vol.119, pp.7-35, 2010.

G. G. Somjen, Nervenkitt: notes on the history of the concept of neuroglia, Glia, vol.1, pp.2-9, 1988.

N. Soni, B. V. Reddy, and P. Kumar, GLT-1 transporter: an effective pharmacological target for various neurological disorders, Pharmacol. Biochem. Behav, vol.127, pp.70-81, 2014.

K. Sriram, S. A. Benkovic, M. A. Hebert, D. B. Miller, O. 'callaghan et al., Induction of gp130-related cytokines and activation of JAK2/STAT3 pathway in astrocytes precedes up-regulation of glial fibrillary acidic protein in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine model of neurodegeneration: key signaling pathway for astrogliosis in vivo?, J. Biol. Chem, vol.279, 2004.

T. Takano, X. Han, R. Deane, B. Zlokovic, and M. Nedergaard, Twophoton imaging of astrocytic Ca2+ signaling and the microvasculature in experimental mice models of Alzheimer's disease, Ann. N.Y. Acad. Sci, vol.1097, pp.40-50, 2007.

M. Talantova, S. Sanz-blasco, X. Zhang, P. Xia, M. W. Akhtar et al., Abeta induces astrocytic glutamate release, extrasynaptic NMDA receptor activation, and synaptic loss, Proc. Natl. Acad. Sci. U.S.A, vol.110, 2013.

K. Terai, A. Matsuo, and P. L. Mcgeer, Enhancement of immunoreactivity for NF-kappa B in the hippocampal formation and cerebral cortex of Alzheimer's disease, Brain Res, vol.735, pp.159-168, 1996.

D. R. Thal, The role of astrocytes in amyloid beta-protein toxicity and clearance, Exp. Neurol, vol.236, pp.1-5, 2012.

X. Tong, Y. Ao, G. C. Faas, S. E. Nwaobi, J. Xu et al., Astrocyte Kir4.1 ion channel deficits contribute to neuronal dysfunction in Huntington's disease model mice, Nat. Neurosci, vol.17, pp.694-703, 2014.

M. Tortarolo, P. Veglianese, N. Calvaresi, A. Botturi, C. Rossi et al., Persistent activation of p38 mitogen-activated protein kinase in a mouse model of familial amyotrophic lateral sclerosis correlates with disease progression, Mol. Cell. Neurosci, vol.23, issue.03, pp.22-30, 2003.

U. Trager, R. Andre, N. Lahiri, A. Magnusson-lind, A. Weiss et al., HTT-lowering reverses Huntington's disease immune dysfunction caused by NFkappaB pathway dysregulation, Brain, vol.137, pp.819-833, 2014.

H. H. Tsai, H. Li, L. C. Fuentealba, A. V. Molofsky, R. Taveira-marques et al., Regional astrocyte allocation regulates CNS synaptogenesis and repair, Science, vol.337, pp.358-362, 2012.

B. J. Turner, T. , and K. , Transgenics, toxicity and therapeutics in rodent models of mutant SOD1-mediated familial ALS, Prog. Neurobiol, vol.85, pp.94-134, 2008.

S. Tydlacka, C. E. Wang, X. Wang, S. Li, and X. J. Li, Differential activities of the ubiquitin-proteasome system in neurons versus glia may account for the preferential accumulation of misfolded proteins in neurons, J. Neurosci, vol.28, pp.13285-13295, 2008.

M. Valenza, V. Leoni, J. M. Karasinska, L. Petricca, J. Fan et al., Cholesterol defect is marked across multiple rodent models of Huntington's disease and is manifest in astrocytes, J. Neurosci, vol.30, pp.10844-10850, 2010.

M. Valenza, M. Marullo, E. Di-paolo, E. Cesana, C. Zuccato et al., Disruption of astrocyte-neuron cholesterol cross talk affects neuronal function in Huntington's disease, Cell Death Differ, vol.22, pp.690-702, 2015.

M. R. Vargas, D. A. Johnson, D. W. Sirkis, A. Messing, J. et al., Nrf2 activation in astrocytes protects against neurodegeneration in mouse models of familial amyotrophic lateral sclerosis, J. Neurosci, vol.28, pp.13574-13581, 2008.

M. R. Vargas, J. , and J. A. , The Nrf2-ARE cytoprotective pathway in astrocytes, Expert Rev. Mol. Med, vol.11, 2009.

M. R. Vargas, M. Pehar, P. Cassina, L. Martinez-palma, J. A. Thompson et al., Fibroblast growth factor-1 induces heme oxygenase-1 via nuclear factor erythroid 2-related factor 2 (Nrf2) in spinal cord astrocytes: consequences for motor neuron survival, J. Biol. Chem, vol.280, pp.25571-25579, 2005.

S. Venneti, B. J. Lopresti, and C. A. Wiley, The peripheral benzodiazepine receptor (Translocator protein 18kDa) in microglia: from pathology to imaging, Prog. Neurobiol, vol.80, pp.308-322, 2006.

A. J. Vincent, R. Gasperini, L. Foa, and D. H. Small, Astrocytes in Alzheimer's disease: emerging roles in calcium dysregulation and synaptic plasticity, J. Alzheimers. Dis, vol.22, pp.699-714, 2010.

R. Virchow, , 1856.

, Gesammelte Abhandlungen zur wissenschaftlichen Medizin. Frankfurt: Verlag von Meidinger Sohn & Comp

J. C. Vis, L. F. Nicholson, R. L. Faull, W. H. Evans, N. J. Severs et al., Connexin expression in Huntington's diseased human brain, Cell Biol. Int, vol.22, pp.837-847, 1998.

J. P. Vonsattel, R. H. Myers, T. J. Stevens, R. J. Ferrante, E. D. Bird et al., Neuropathological classification of Huntington's disease, J. Neuropathol. Exp. Neurol, vol.44, pp.559-577, 1985.

C. Y. Wang, S. H. Yang, and S. F. Tzeng, MicroRNA-145 as one negative regulator of astrogliosis, Glia, vol.63, pp.194-205, 2015.

L. Wang, F. Lin, J. Wang, J. Wu, R. Han et al., Expression of mutant N-terminal huntingtin fragment (htt552-100Q) in astrocytes suppresses the secretion of BDNF, Brain Res, vol.1449, pp.69-82, 2012.

K. B. Washburn and J. T. Neary, P2 purinergic receptors signal to STAT3 in astrocytes: difference in STAT3 responses to P2Y and P2X receptor activation, Neuroscience, vol.142, pp.411-423, 2006.

C. Weigert, Beiträge zur Kenntnis der normalen menschlichen Neuroglia, Zeitschrift für Psychologie und Physiologie der Sinnesorgane, ed Liepmann, 1895.

E. J. Wild and S. J. Tabrizi, Targets for future clinical trials in Huntington's disease: what's in the pipeline?, Mov. Disord, vol.29, pp.1434-1445, 2014.

U. Wilhelmsson, E. A. Bushong, D. L. Price, B. L. Smarr, V. Phung et al., Redefining the concept of reactive astrocytes as cells that remain within their unique domains upon reaction to injury, Proc. Natl. Acad. Sci. U.S.A, vol.103, pp.17513-17518, 2006.

T. Witjas, E. Kaphan, J. P. Azulay, O. Blin, M. Ceccaldi et al., Nonmotor fluctuations in Parkinson's disease: frequent and disabling, Neurology, vol.59, pp.408-413, 2002.

S. Witte and S. A. Muljo, Integrating non-coding RNAs in JAK-STAT regulatory networks, JAKSTAT, vol.3, p.28055, 2014.

A. M. Wojtowicz, A. Dvorzhak, M. Semtner, and R. Grantyn, Reduced tonic inhibition in striatal output neurons from Huntington mice due to loss of astrocytic GABA release through GAT-3, Front. Neural Circuits, vol.7, p.188, 2013.

R. L. Woltjer, K. Duerson, J. M. Fullmer, P. Mookherjee, A. M. Ryan et al., Aberrant detergent-insoluble excitatory amino acid transporter 2 accumulates in Alzheimer disease, J. Neuropathol. Exp. Neurol, vol.69, pp.667-676, 2010.

Z. Wu, Z. Guo, M. Gearing, C. , and G. , Tonic inhibition in dentate gyrus impairs long-term potentiation and memory in an Alzhiemer's disease model, Nat. Commun, vol.5, p.4159, 2014.

T. Wyss-coray, J. D. Loike, T. C. Brionne, E. Lu, R. Anankov et al., Adult mouse astrocytes degrade amyloid-beta in vitro and in situ, Nat. Med, vol.9, pp.453-457, 2003.

Q. Xiao, P. Yan, X. Ma, H. Liu, R. Perez et al., Enhancing astrocytic lysosome biogenesis facilitates abeta clearance and attenuates amyloid plaque pathogenesis, J. Neurosci, vol.34, pp.9607-9620, 2014.

P. Yan, X. Hu, H. Song, K. Yin, R. J. Bateman et al., Matrix metalloproteinase-9 degrades amyloid-beta fibrils in vitro and compact plaques in situ, J. Biol. Chem, vol.281, pp.24566-24574, 2006.

Y. Yoshii, A. Otomo, L. Pan, M. Ohtsuka, and S. Hadano, Loss of glial fibrillary acidic protein marginally accelerates disease progression in a SOD1(H46R) transgenic mouse model of ALS, Neurosci. Res, vol.70, pp.321-329, 2011.

J. L. Zamanian, L. Xu, L. C. Foo, N. Nouri, L. Zhou et al., Genomic analysis of reactive astrogliosis, J. Neurosci, vol.32, pp.6391-6410, 2012.

T. Zeis, I. Allaman, M. Gentner, K. Schroder, J. Tschopp et al., Metabolic gene expression changes in astrocytes in Multiple Sclerosis cerebral cortex are indicative of immune-mediated signaling, Brain Behav. Immun, 2015.

B. V. Zlokovic, The blood-brain barrier in health and chronic neurodegenerative disorders, Neuron, vol.57, pp.178-201, 2008.

. =>-?@a@bcdce>-fgf>-?hgi->-h?jkl@,

S. Wx-hgs->-ywf>-?hgi->-h,

. <=>?@?a-b=?@?c-b=de-f?bg-c-@?>c-?hiji?hi?g@e-g-k-=?>bk-c-jg-c-@?,

Y. <iz,

. J=g-<]=,

, IjG I?>C mIG @@D n@jG @>G FH^G <IZ

. J=g-&lt;]=9,

I. &lt;ik-i=k-io=?^o@d, . =kg, @. &gt;=?hg-ib&lt;?c-pfi&gt;g, and . Fh^g-&lt;iz,

H. I. J=g-&lt;]=^c-?g-&lt;ink-=c-?-:q9@imc, &. Big-&lt;i=bg-c-m=g-c-@?@eg-&lt;ij=g-&lt;]=^]c-g-&lt;nc-@b&lt;ioc-b=dr&lt;c, B. @d-@sc, and . Ib&lt;?c-pfi&gt;l,

C. &lt;i=bg-c-mc and ?. &lt;ij=g-&lt;]=^]c-g-&lt;k-ij@k-g-ik&gt;^&gt;g-io&gt;-t9@o@?c-g-@kg-k-=?&gt;bk-c-jg-c-@?=dc-?hfbg-c-@?l#177t%-l-q-#,

C. Zb{c, G @K89:9, p.4

C. 3|630-$-t9@o@?c-g-@kg-&lt;ie-@k-o=g and . Ic-?b@ojd-iji&gt;lo1,

. =g and . &lt;iz,

. J=g-&lt;]=t,

@. &lt;=k-o=b@d and . B=dc-?&lt;c, , p.39

?. Hc-g-c-@?=d,

. I&gt;&gt;c-@?@ec-?&lt;c-nc-g-@k and . &gt;@kh@oc, =?G?Is=G C mIoFG =?G >l9 30, vol.6, p.376

I. $-tjjk and . &gt;g-c-g-fg-c-mid=bg-c-miofg-=?g, >lP, vol.76, p.9

. !-#-!,

C. Pjepjp--&gt;&gt;?-!-&quot;-!-#-!-)-)-!-$&amp;$&amp;-&apos;-!-#6,

. #%&apos;-#&apos;-!-#-#)q-#%&apos;-%%&apos;-!-#,

R. 9=-.-!-&quot;-)--&gt;&gt;?-!-&quot;)-!-#-!-&quot;-!-#&quot;-!-%-%-!-#2-%,

T. 16788u,

, 67899= 6 R 3 X! ' . 67887:-&% ' . 6 788<:>Y @ ' . 6789Z= 6! #0 ! #1! & $! 3

. +&amp;!-&apos;, X% ' . 67899=, vol.6788

%. , , vol.67888

. &amp;#%-r!-#9-&amp;&apos;-z$&amp;#, , vol.96

!. 6. #3-c`&gt;`6-!-&apos; and . V-;-&amp;-!-$&apos;-&quot;!-)#-&amp;, #0 & ]4d->>? $ &1 ! ! ) 1! & ! "! .51" 6 )# & ->>?! ' '

, @# #) @#, vol.6789

. !-%$-;-!-#-&amp;$!-&apos;-$-!-&amp;!-$$-$%3 and . X%, ! ' 0 ! # 0 -+S3 5 ## ' . 6788z:/ ## ' . 6789?= . ->>?! ' $#! ' 0 " ' . 6789U= 6

, 7899= 6 0 c`>`! #2 ! #3 -! ' . 6788Z= #)! #

. $-!-$&amp;-&apos;#-&quot;!-#2-%, 3 >%) ' . 67899= . 4& ' .' %)"! '(# #0 0 )! # # #( ! "3 {,=0 0->>? ! #&! ! &! )

. $!-|-&apos;-&apos;-!-#$!-#,

#. , ! 6 &},' "' 0~x iy6 m lnh m n #) guvgj 1 )%) ->>?{,3 4& ' . 6789<= .a# ! %)! )

, # wx iyo pq lrgh ih j st s #(#! ! 0 % & ' ! &) &! $ #0->>?0 ! "! . ;# # ' 6 #(#! ! )! $' )%)

. &amp;$-$&amp;-#)aw`%$-(%&apos;-!-#0,

. S-#-!-#-;--&gt;&gt;?!-#-;-#!-#-!-&quot;!-!-#, ! # q uguym i.-f76 & q uguym i &' (0->>?6# ' (' ! ' ! "! 0 R# ' ! #2 % 3 {& ' . 6789Z= . >& %' &1 & ->>?! # ' (%' 0

C. Ilemc-g-s-#$, , p.61

, 6#% ! # ! # #) #% # ' ) & ( )% ' ' ' ! &, p.6

. $$-$&apos;-#, >>? ! " ! # &c-3 T% ' . 6789?= $&! ' '' !, pp.3-6

!. #%, 0T-3 -&! ' . 67898= #)' Y&! _ )! 3 {6

$. , , vol.3, pp.5-6

. #5-!-&apos;-.-6789&lt;=-.-/&amp;!-&apos;--&gt;&gt;?-!-&quot;-!-#!-#!-#-&apos;-)-)!-#-!-&quot;-!-#-,{-)-;-!-&apos;-!-#-&amp;-&apos;-!-&amp;#0-!, ! 3 ;#5 ! ' . 6789< 0 "! 1= .`& ' (! '! #&! ! ! #0]47 #) $! |4V 0gh ih j! # &c`>`)'0`{ #%, p.1

!. #&amp;!-!-!-#!-#-$&amp;, , pp.0-5

. #5-!-&apos;-.-6789&lt;=-.--v+-?&quot;-r$-!-#$-&quot;#-)-&gt;&gt;?-!-&quot;-!-#-#,

. &gt;&amp;]4d-&gt;&gt;?$-&amp;1-!-#,

%. +y-!-&apos;-.q and . !-#?, , vol.83, pp.789-78

!. %&amp;-&apos;#(#)&amp;-*-+&quot;$, $)#4 ":;<& 4

, & '#4 "<. #-)#4 "56 78?TU2-":V $.>. $& #4 "@ #A#@ )4 "-)& . $*

%. and ;. C#, #&-@ J 9EFFZ

, #))4 $"$%XY2DJ1'# ';?, 56 78>. $?$& #.4 ),#@ @<'-. -<& #. 4 _#

@. ,

$. B-a# and . #&lt;$, #. A#:4 "& '#. $:#"&56 78>. $?$& #.B U-+-)'4 ?-#&-@ J 9GbbbI Jc4 ?#"& 4 "9-"$& '#.4 "& #. ?#:4 -& #d@ -?#"& <'-. -<& #. 4 )& 4 <$%. #-<& 4 A#-),#@ @-)4 ??-& ;. #-)& . $<=& #), p.9

@. ,

, &)4 /"-@ 4 "/<-)<-:#$<<, <4 :#),4 & '& '##^>. #))4 $" $%/@ 4 -@?-. +#. )XY2D-":0GFFf-&`GZJ H4 "?4 <#B C# #&-@ J 9EFFHI J01213(4 ":)& $& '#56 78>. $?$& #

$. B-g,

&. =@--&amp;-#, iU2?#& '=@ -* l2W "-?$:#@$%1gW 9& '#>'$)

%. $. ?$%01213b-.-#:i-&lt;&lt;;?;@--&amp;-#)4, <=& #)$A#. #^>. #))4 "/XY2DB /. ##"I J0'-?* $>#. -& #:. -& ):4 )>@ -=@ $,XY2D-":"$>'$)>'$* 012134 ??;"$. #-<& 4 A4 & =JulW "& '#<$. & #^#^>$)#:& $1gW 9-<& 4 A-& 4 $"$%& '#L2MN01213>-& ',-=4 )-@ )$:#& #<& #

. &lt;@-#;)$%-)&amp;-.-$&lt;=&amp;-#)jlw,

. &gt;)&amp;-.-#-?-&lt;-)&lt;-:#).-#)&gt;$,

, #@ @-)L2MEB >L2MEI(;&"$&L2MGB >L2MGI JW "& #. #)& 4 "/@ =9012134 )>'$)>'$. =@ -& #:-&1=. hFH -),#@ @-)0#. hEhB 2vuwx'>$)&1gW 9y3'>$)&1gW 9?$:4 d#:% . $?9!@ 4 A-#&-@ J 9EFGEI JW, p.4

&. , #& 4 ?#):4 z<

@. ={, , vol.4

. &lt;@-#;)$%.-#-&lt;&amp;, A#-)& . $<=& #)4 )$()#. A#:4 "?-"=:4 )#-)#?$:#@ )B g#"C-4 ?#&-@ J 9EFGH-I J4lY$.#^-?>@ #9 01213-<& 4 A-& 4 $"B . #:I4 )$()#. A#:4 "XY2D~. #-<& 4 A#-)& . $<=& #)B /. ##"I 94 "& '#'4 >>$<-?>;)$%2DDV D0G:`b?4 <#9-& . -")/#"4 <?$:#@$%2iJ T#-<& 4 A#-)& . $<=& #),4 & ')& . $"/01213-<& 4 A-& 4 $"-. ##)>#<4 -@ @, vol.4

#. Jxy2d, w29g HF?9i EH?J EGF MJ}#=_#

-. *4+, , vol.80623, pp.436-60645

. /-)1a,

, 1A)@, -36$4+2 9, vol.6, p.4

, 82 %88 4:42 ? 3 +, <522, vol.9, pp.3-4

!. %&amp;, 1) 79 8 * '+7 ,&, vol.179, p.7

, > 9, vol.7, pp.8-7187

!. and &. , 6%8 11179 7*'A 3 71 >8 7 77)1-.18 12610 B/23BB9

7. 1d3 and &. , , vol.18, pp.1371-1372

&. , , p.6

, ##9%&7 * +9 79 6! ), vol.79, p.718779

$. %. , , vol.9

, + 9 +9 1''8 18+*9 1 )7 )! ?%'8

&. , , vol.79, p.718779

. =79-+, , vol.6

$. &amp;&amp;%, % ! 1% ! &% )*! 7 2+, vol.7, pp.212-213

$. and -. , , vol.3

, >$ 2955" ! 27 ?<@ 38 7 11!7! ! 1! " ! $7&2? & ! 7 " ! 1% 5 231% 1% ! &% % 1%7 2% ! 1%%

, ! ! 17 2#1$$1% ! & 12 57! % 7 (7 1 >>3 22% $127 $1" % ! 7 21! 7 2A ! &)4*! 733+ -B

, >>37 21% ! &% +67 D! 27 2>>3! 2% 212

!. 1%,

, ! =&2452% 1051?145 ! 7*959123E5C'>5

+. 3bg, 3G)

. *!-$,

%. &lt;0512565012352650123e56&gt;5912350123,

, >>37&7 % 2 $7 " &7 232=&$)2! *1% 17H B+ :GG.,:) + ::3.:,)

, /<7 % 2! 7 1% 1% 35% *, vol.67, pp.32-39

. 2%497-7+, GG) 650123956754547 23957 45752125120 ,>! ">>37 21% 13&)% 13&+ BG.B3G), vol.8

, $$27 % &+1 17 23! " !>>3)*1%0(112!G+ BG

. 3+-/3g,

, ! 7 2121% 7 (1% 7 2">>37 2% 1% ! &%

&. , 92395029561! 2 4!4 GG:1% 1% 312% 1% :+$$7! " % " 1$7 &"7 321% ! 12! 121% 7 (1% ! "% ! 12! 7 %, vol.7

!. 17hg+,

. !-?%, " 1% !127 2%

A. Alvarez-buylla, J. M. García-verdugo, A. D. Tramontin, M. A. Crossref-medline-anderson, Y. Ao et al., Lentiviralmediated delivery of mutant huntingtin in the striatum of rats induces a selective neuropathology modulated by polyglutamine repeat size, huntingtin expression levels, and protein length, CrossRef Medline Arrasate M, Finkbeiner S (2012) Protein aggregates in Huntington's disease, vol.2, pp.5587-5593, 1993.

H. Y. Hsiao, Y. C. Chen, H. M. Chen, P. H. Tu, Y. Chern et al., Induction of gp130-related cytokines and activation of JAK2/STAT3 pathway in astrocytes precedes up-regulation of glial fibrillary acidic protein in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine model of neurodegeneration: key signaling pathway for astrogliosis in vivo, Mutant presenilins specifically elevate the levels of the 42 residue beta-amyloid peptide in vivo: evidence for augmentation of a 42-specific gamma secretase, vol.22, pp.6391-6410, 1982.

B. Haim, A Universal Trigger of, Astrocyte Reactivity J. Neurosci, vol.35, issue.6, pp.2817-2829, 2015.

. Eyo, , 2014.

O. Abiega, S. Beccari, I. Diaz-aparicio, A. Nadjar, S. Layé et al., Neuronal Hyperactivity Disturbs ATP Microgradients, Impairs Microglial Motility, and Reduces Phagocytic Receptor Expression Triggering Apoptosis/Microglial Phagocytosis Uncoupling, PLoS Biol, vol.14, pp.588-593, 2000.
URL : https://hal.archives-ouvertes.fr/hal-01605002

B. Ajami, J. L. Bennett, C. Krieger, W. Tetzlaff, and F. M. Rossi, Local self-renewal can sustain CNS microglia maintenance and function throughout adult life, Nat. Neurosci, vol.10, pp.1538-1543, 2007.

J. J. Alam, Selective Brain-Targeted Antagonism of p38 MAPK? Reduces Hippocampal IL-1? Levels and Improves Morris Water Maze Performance in Aged Rats, J. Alzheimers Dis. JAD, vol.48, pp.219-227, 2015.

W. A. Alaynick, Nuclear receptors, mitochondria and lipid metabolism. Mitochondrion, vol.8, pp.329-337, 2008.

R. Alonso, D. Pisa, A. I. Marina, E. Morato, A. Rábano et al., Fungal infection in patients with Alzheimer's disease, J. Alzheimers Dis. JAD, vol.41, pp.301-311, 2014.

R. Alonso, D. Pisa, A. Rábano, I. Rodal, and L. Carrasco, Cerebrospinal Fluid from Alzheimer's Disease Patients Contains Fungal Proteins and DNA, J. Alzheimers Dis. JAD, vol.47, pp.873-876, 2015.

K. Andersen, L. J. Launer, A. Ott, A. W. Hoes, M. M. Breteler et al., Do nonsteroidal anti-inflammatory drugs decrease the risk for Alzheimer's disease? The Rotterdam Study, Neurology, vol.45, pp.1441-1445, 1995.

G. Arsenault-lapierre, H. Chertkow, and S. Lupien, Seasonal effects on cortisol secretion in normal aging, mild cognitive impairment and Alzheimer's disease, Neurobiol. Aging, vol.31, pp.1051-1054, 2010.

A. Asea, M. Rehli, E. Kabingu, J. A. Boch, O. Bare et al., Novel signal transduction pathway utilized by extracellular HSP70: role of toll-like receptor (TLR) 2 and TLR4, J. Biol. Chem, vol.277, pp.15028-15034, 2002.

A. Schlunk, S. H. Mitchell, K. W. Brewster, C. E. Hudson, M. J. Cole et al., Fractalkine and CX 3 CR1 regulate hippocampal neurogenesis in adult and aged rats, Neurobiol. Aging, vol.32, pp.2030-2044, 2011.

D. Baglietto-vargas, Y. Chen, D. Suh, R. R. Ager, C. J. Rodriguez-ortiz et al., Short-term modern life-like stress exacerbates A?-pathology and synapse loss in 3xTg-AD mice, J. Neurochem, vol.134, pp.915-926, 2015.

L. D. Baker, L. L. Frank, K. Foster-schubert, P. S. Green, C. W. Wilkinson et al., Effects of aerobic exercise on mild cognitive impairment: a controlled trial, Arch. Neurol, vol.67, pp.71-79, 2010.

M. Baker, I. R. Mackenzie, S. M. Pickering-brown, J. Gass, R. Rademakers et al., Oxidative damage and progression to Alzheimer's disease in patients with mild cognitive impairment, J. Alzheimers Dis. JAD, vol.442, pp.1165-1177, 2006.

M. E. Bamberger, M. E. Harris, D. R. Mcdonald, J. Husemann, and G. E. Landreth, A cell surface receptor complex for fibrillar betaamyloid mediates microglial activation, J. Neurosci. Off. J. Soc. Neurosci, vol.23, pp.2665-2674, 2003.

R. Barbalat, S. E. Ewald, M. L. Mouchess, and G. M. Barton, Nucleic acid recognition by the innate immune system, Annu. Rev. Immunol, vol.29, pp.185-214, 2011.

D. E. Barnes, Y. , and K. , The projected effect of risk factor reduction on Alzheimer's disease prevalence, Lancet Neurol, vol.10, pp.819-828, 2011.

A. Baroja-mazo, F. Martín-sánchez, A. I. Gomez, C. M. Martínez, J. Amores-iniesta et al., , 2014.

G. Bartzokis, Age-related myelin breakdown: a developmental model of cognitive decline and Alzheimer's disease, Neurobiol. Aging, vol.15, pp.5-18, 2004.

G. Bartzokis, Alzheimer's disease as homeostatic responses to age-related myelin breakdown, Neurobiol. Aging, vol.32, pp.1341-1371, 2011.

G. Bartzokis, P. H. Lu, J. Mintz, K. Baruch, A. Deczkowska et al., Quantifying age-related myelin breakdown with MRI: novel therapeutic targets for preventing cognitive decline and Alzheimer's disease, J. Alzheimers Dis. JAD, vol.6, pp.135-137, 2004.

S. Basu, R. J. Binder, R. Suto, K. M. Anderson, and P. K. Srivastava, Necrotic but not apoptotic cell death releases heat shock proteins, which deliver a partial maturation signal to dendritic cells and activate the NF-kappa B pathway, Int. Immunol, vol.12, pp.1539-1546, 2000.

K. Bhaskar, M. Konerth, O. N. Kokiko-cochran, A. Cardona, R. M. Ransohoff et al., Regulation of tau pathology by the microglial fractalkine receptor, Neuron, vol.68, pp.19-31, 2010.

M. E. Bianchi, DAMPs, PAMPs and alarmins: all we need to know about danger, J. Leukoc. Biol, vol.81, pp.1-5, 2007.

M. E. Bianchi, HMGB1 loves company, J. Leukoc. Biol, vol.86, pp.573-576, 2009.

K. Biber, H. Neumann, K. Inoue, and H. W. Boddeke, Neuronal "On" and "Off" signals control microglia, Trends Neurosci, vol.30, pp.596-602, 2007.

G. J. Biessels, M. W. Strachan, F. L. Visseren, L. J. Kappelle, and R. A. Whitmer, , 2014.

, Dementia and cognitive decline in type 2 diabetes and prediabetic stages: towards targeted interventions, Lancet Diabetes Endocrinol, vol.2, pp.246-255

K. Bisht, K. P. Sharma, C. Lecours, M. G. Sánchez, H. El-hajj et al., Reduced physical activity and risk of chronic disease: the biology behind the consequences, Eur. J. Appl. Physiol, vol.64, pp.381-390, 2008.

L. M. Boulanger, Immune proteins in brain development and synaptic plasticity, Neuron, vol.64, pp.93-109, 2009.

J. C. Breitner, B. A. Gau, K. A. Welsh, B. L. Plassman, W. M. Mcdonald et al., Inverse association of antiinflammatory treatments and Alzheimer's disease: initial results of a co-twin control study, Neurology, vol.44, pp.227-232, 1994.

J. C. Breitner, L. D. Baker, and T. J. Montine,

C. L. Meinert, C. G. Lyketsos, K. H. Ashe, J. Brandt, S. Craft et al., , 2011.

, Alzheimers Dement. J. Alzheimers Assoc, vol.7, pp.402-411

M. Brundel, L. J. Kappelle, and G. J. Biessels, Brain imaging in type 2 diabetes, Eur. Neuropsychopharmacol. J. Eur. Coll. Neuropsychopharmacol, vol.24, pp.1967-1981, 2014.

J. M. Busillo and J. A. Cidlowski, The five Rs of glucocorticoid action during inflammation: ready, reinforce, repress, resolve, and restore, Trends Endocrinol. Metab. TEM, vol.24, pp.109-119, 2013.

C. , W. , G. , X. Sa, T. Wj et al., resistance, metabolic defects, apoptosis, and molecular signatures of immune activation in response to infections, Biochem. Biophys. Res. Commun, vol.482, pp.282-288, 2017.

A. Cagnin, D. J. Brooks, A. M. Kennedy, R. N. Gunn, R. Myers et al., In-vivo measurement of activated microglia in dementia, Lancet Lond, 2001.

. Engl, , vol.358, pp.461-467

D. Cai, Neuroinflammation and neurodegeneration in overnutrition-induced diseases, Trends Endocrinol. Metab. TEM, vol.24, p.40, 2013.

A. E. Cardona, E. P. Pioro, M. E. Sasse, V. Kostenko, S. M. Cardona et al., Control of microglial neurotoxicity by the fractalkine receptor, Nat. Neurosci, vol.9, pp.917-924, 2006.

I. Carreras, A. C. Mckee, J. Choi, N. Aytan, N. W. Kowall et al., R-flurbiprofen improves tau, but not Aß pathology in a triple transgenic model of Alzheimer's disease, Brain Res, vol.1541, pp.115-127, 2013.

S. F. Carter, M. Schöll, O. Almkvist, A. Wall, H. Engler et al., Evidence for astrocytosis in prodromal Alzheimer disease provided by 11C-deuterium-Ldeprenyl: a multitracer PET paradigm combining 11C-Pittsburgh compound B and 18F-FDG, J. Nucl. Med. Off. Publ. Soc. Nucl. Med, vol.53, pp.37-46, 2012.

P. Castellani, E. Balza, and A. Rubartelli, , 2014.

, DAMPs, tumor development, and progression: a vicious circle orchestrated by redox signaling, Antioxid. Redox Signal, vol.20, pp.1086-1097

J. K. Cataldo, J. J. Prochaska, S. A. Glantz, P. Chakrabarty, A. Li et al., Cigarette smoking is a risk factor for Alzheimer's Disease: an analysis controlling for tobacco industry affiliation, J. Alzheimers Dis. JAD, vol.19, pp.519-533, 2010.

K. A. Chalmers and S. Love, Neurofibrillary tangles may interfere with Smad 2/3 signaling in neurons, J. Neuropathol. Exp. Neurol, vol.66, pp.158-167, 2007.

C. C. Chao, T. A. Ala, S. Hu, K. B. Crossley, R. E. Sherman et al., Serum cytokine levels in patients with Alzheimer's disease, Clin. Diagn. Lab. Immunol, vol.1, pp.433-436, 1994.

F. Chauveau, H. Boutin, N. Van-camp, F. Dollé, and B. Tavitian, Nuclear imaging of neuroinflammation: a comprehensive review of [11C]PK11195 challengers, Eur. J. Nucl. Med, 2008.

, Mol. Imaging, vol.35, pp.2304-2319

Z. Chen, W. Jalabi, K. B. Shpargel, K. T. Farabaugh, R. Dutta et al., , 2012.

, Lipopolysaccharide-induced microglial activation and neuroprotection against experimental brain injury is independent of hematogenous TLR4, J. Neurosci. Off. J. Soc. Neurosci, vol.32, pp.11706-11715

Z. Chen, W. Jalabi, W. Hu, H. Park, J. T. Gale et al., Microglial displacement of inhibitory synapses provides neuroprotection in the adult brain, Am. J. Geriatr. Psychiatry Off. J. Am. Assoc. Geriatr. Psychiatry, vol.5, pp.63-74, 2014.

S. H. Cho, B. Sun, Y. Zhou, T. M. Kauppinen, B. Halabisky 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, vol.286, pp.32713-32722, 2011.

I. L. Choo, S. F. Carter, M. L. Schöll, and A. Nordberg, , p.11, 2014.

, C-deprenyl PET correlates with decrease in gray matter density in the parahippocampus of prodromal Alzheimer's patients, Eur. J. Nucl. Med. Mol. Imaging, vol.41, pp.2120-2126

J. Ciesielski-treska, G. Ulrich, S. Chasserot-golaz, J. Zwiller, M. O. Revel et al., Mechanisms underlying neuronal death induced by chromogranin A-activated microglia, J. Biol. Chem, vol.276, pp.13113-13120, 2001.

J. D. Clements, R. A. Lester, G. Tong, C. E. Jahr, and G. L. Westbrook, The time course of glutamate in the synaptic cleft, Science, vol.258, pp.1498-1501, 1992.

R. C. Coll, A. A. Robertson, J. J. Chae, S. C. Higgins, R. Muñoz-planillo et al.,

, Nat. Med, vol.21, pp.248-255

H. C. Comijs, L. Gerritsen, B. W. Penninx, M. A. Bremmer, D. J. Deeg et al., The association between serum cortisol and cognitive decline in older persons, 2010.

, J. Geriatr. Psychiatry Off. J. Am. Assoc. Geriatr. Psychiatry, vol.18, pp.42-50

D. Commenges, V. Scotet, S. Renaud, H. Jacqmin-gadda, P. Barberger-gateau et al., Intake of flavonoids and risk of dementia, Eur. J. Epidemiol, vol.16, pp.357-363, 2000.

A. W. Corona, D. M. Norden, J. P. Skendelas, Y. Huang, J. C. O&apos;connor et al., , 2013.

, Brain. Behav. Immun, vol.31, pp.134-142

M. Cruts, I. Gijselinck, J. Van-der-zee, S. Engelborghs, H. Wils et al., Null mutations in progranulin cause ubiquitin-positive frontotemporal dementia linked to chromosome 17q21, Nature, vol.442, pp.920-924, 2006.

H. A. Crystal, E. Ortof, W. H. Frishman, A. Gruber, D. Hershman et al., Serum vitamin B12 levels and incidence of dementia in a healthy elderly population: a report from the Bronx Longitudinal Aging Study, J. Am. Geriatr. Soc, vol.42, pp.933-936, 1994.

J. G. Csernansky, H. Dong, A. M. Fagan, L. Wang, C. Xiong et al., Plasma cortisol and progression of dementia in subjects with Alzheimer-type dementia, Am. J. Psychiatry, vol.163, pp.2164-2169, 2006.

M. R. Damani, L. Zhao, A. M. Fontainhas, J. Amaral, R. N. Fariss et al., , 2011.

, Age-related alterations in the dynamic behavior of microglia, Aging Cell, vol.10, pp.263-276

N. C. Danbolt, Glutamate uptake, Prog. Neurobiol, vol.65, pp.1-105, 2001.

M. J. Daniels, J. Rivers-auty, T. Schilling, N. G. Spencer, W. Watremez et al., Fenamate NSAIDs inhibit the NLRP3 inflammasome and protect against Alzheimer's disease in rodent models, Brain J. Neurol, vol.7, pp.1237-1251, 2016.

F. G. De-felice, D. Wu, M. P. Lambert, S. J. Fernandez, P. T. Velasco et al., Alzheimer's disease-type neuronal tau hyperphosphorylation induced by A beta oligomers, Neurobiol. Aging, vol.29, pp.1334-1347, 2008.

B. Degryse, T. Bonaldi, P. Scaffidi, S. Müller, M. Resnati et al., The high mobility group (HMG) boxes of the nuclear protein HMG1 induce chemotaxis and cytoskeleton reorganization in rat smooth muscle cells, J. Cell Biol, vol.152, pp.1197-1206, 2001.

L. A. Denner, J. Rodriguez-rivera, S. J. Haidacher, J. B. Jahrling, J. R. Carmical et al., Cognitive enhancement with rosiglitazone links the hippocampal PPAR? and ERK MAPK signaling pathways, J. Neurosci. Off. J. Soc. Neurosci, vol.32, pp.16725-16735, 2012.

M. K. Desai, M. A. Mastrangelo, D. A. Ryan, K. L. Sudol, W. C. Narrow et al., Early oligodendrocyte/myelin pathology in Alzheimer's disease mice constitutes a novel therapeutic target, Am. J. Pathol, vol.177, pp.1422-1435, 2010.

B. E. Deverman and P. H. Patterson, , 2009.

C. Cytokines and . Development, Neuron, vol.64, pp.61-78

C. Dickey, C. Kraft, U. Jinwal, J. Koren, A. Johnson et al., Aging analysis reveals slowed tau turnover and enhanced stress response in a mouse model of tauopathy, Nat. Rev. Drug Discov, vol.174, pp.633-652, 2009.

L. Dissing-olesen, J. M. Ledue, R. L. Rungta, J. K. Hefendehl, H. B. Choi et al., Activation of neuronal NMDA receptors triggers transient ATP-mediated microglial process outgrowth, J. Neurosci. Off. J. Soc. Neurosci, vol.34, pp.10511-10527, 2014.

H. Dong, C. M. Yuede, H. Yoo, M. V. Martin, C. Deal et al., Corticosterone and related receptor expression are associated with increased betaamyloid plaques in isolated Tg2576 mice, Neuroscience, vol.155, pp.154-163, 2008.

X. Dong, Y. Wang, and Z. Qin, , 2009.

, Acta Pharmacol. Sin, vol.30, pp.379-387

R. S. Duan, X. Yang, Z. G. Chen, M. O. Lu, C. Morris et al., , 2008.

, Decreased fractalkine and increased IP-10 expression in aged brain of APPswe transgenic mice, Neurochem. Res, vol.33, pp.1085-1089

N. Dunn, M. Mullee, V. H. Perry, and C. Holmes, Association between dementia and infectious disease: evidence from a case-control study, Alzheimer Dis. Assoc. Disord, vol.19, pp.91-94, 2005.

E. Dursun, D. Gezen-ak, H. Hana?as?, B. Bilgiç, E. Lohmann et al., , 2015.

, Alzheimer's disease, mild cognitive impairment or Parkinson's disease, J. Neuroimmunol, vol.283, pp.50-57

P. Edison, H. A. Archer, A. Gerhard, R. Hinz, N. Pavese et al., , 2008.

. Microglia, Alzheimer's disease: An [11C](R), p.11195

P. Study, Neurobiol. Dis, vol.32, pp.412-419

B. J. Eggen, D. Raj, U. Hanisch, and H. W. Boddeke, Microglial phenotype and adaptation, J. Neuroimmune Pharmacol. Off. J. Soc. NeuroImmune Pharmacol, vol.8, pp.807-823, 2013.

L. H. Eggermont, D. F. Swaab, E. M. Hol, and E. J. Scherder, Walking the line: a, 2009.

, Neurol. Neurosurg. Psychiatry, vol.80, pp.802-804

J. El-khoury, M. Toft, S. E. Hickman, T. K. Means, K. Terada et al., Ccr2 deficiency impairs microglial accumulation and accelerates progression of Alzheimer-like disease, Nat. Med, vol.13, pp.432-438, 2007.

L. J. Van-eldik and W. S. Griffin, S100 beta expression in Alzheimer's disease: relation to neuropathology in brain regions, Biochim. Biophys. Acta, vol.1223, pp.398-403, 1994.

M. F. Elias, P. K. Elias, L. M. Sullivan, P. A. Wolf, and R. B. Agostino, Obesity, diabetes and cognitive deficit: The Framingham Heart Study, Neurobiol. Aging, vol.26, pp.11-16, 2005.

M. R. Elmore, A. R. Najafi, M. A. Koike, N. N. Dagher, E. E. Spangenberg et al., Colony-stimulating factor 1 receptor signaling is necessary for microglia viability, unmasking a microglia progenitor cell in the adult brain, Neuron, vol.82, pp.380-397, 2014.

M. J. Engelhart, M. I. Geerlings, A. Ruitenberg, J. C. Van-swieten, A. Hofman et al., Diet and risk of dementia: Does fat matter, Neurology, vol.59, pp.1915-1921, 2002.

M. J. Engelhart, M. I. Geerlings, J. Meijer, A. Kiliaan, A. Ruitenberg et al., Inflammatory proteins in plasma and the risk of dementia: the rotterdam study, Arch. Neurol, vol.61, pp.668-672, 2004.

J. L. Etnier, R. J. Caselli, E. M. Reiman, G. E. Alexander, B. A. Sibley et al., Cognitive performance in older women relative to ApoE-epsilon4 genotype and aerobic fitness, Med. Sci. Sports Exerc, vol.39, pp.199-207, 2007.

B. Ettle, J. C. Schlachetzki, and J. Winkler, Oligodendroglia and Myelin in Neurodegenerative Diseases: More Than Just Bystanders?, Mol. Neurobiol, vol.53, pp.3046-3062, 2016.

U. B. Eyo, J. Peng, P. Swiatkowski, A. Mukherjee, A. Bispo et al., Neuronal hyperactivity recruits microglial processes via neuronal NMDA receptors and microglial P2Y12 receptors after status epilepticus, J. Neurosci. Off. J. Soc. Neurosci, vol.34, pp.10528-10540, 2014.

A. I. Faden and D. J. Loane, Chronic neurodegeneration after traumatic brain injury: Alzheimer disease, chronic traumatic encephalopathy, or persistent neuroinflammation?, Neurother. J. Am. Soc. Exp. Neurother, vol.12, pp.143-150, 2015.

A. I. Faden, J. Wu, B. A. Stoica, and D. J. Loane, Progressive inflammation-mediated neurodegeneration after traumatic brain or spinal cord injury, Br. J. Pharmacol, vol.173, pp.681-691, 2016.

Z. Fan, Y. Aman, I. Ahmed, G. Chetelat, B. Landeau et al., Influence of microglial activation on neuronal function in Alzheimer's and Parkinson's disease dementia, Alzheimers Dement. J. Alzheimers Assoc, vol.11, pp.3685-3698, 2015.

M. C. Farso, R. D. Shea, and P. M. Beart, Evidence group I mGluR drugs modulate the activation profile of lipopolysaccharideexposed microglia in culture, Neurochem. Res, vol.34, pp.1721-1728, 2009.

K. Fassbender, S. Walter, S. Kühl, R. Landmann, K. Ishii et al.,

, The LPS receptor (CD14) links innate immunity with Alzheimer's disease, FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol, vol.18, pp.203-205

H. H. Feldman, R. S. Doody, M. Kivipelto, D. L. Sparks, D. D. Waters et al., Randomized controlled trial of atorvastatin in mild to moderate Alzheimer disease: LEADe, Neurology, vol.74, pp.956-964, 2010.

M. A. Fishel, G. S. Watson, T. J. Montine, Q. Wang, P. S. Green et al.,

, Hyperinsulinemia provokes synchronous increases in central inflammation and betaamyloid in normal adults, Arch. Neurol, vol.62, pp.1539-1544

A. L. Fitzpatrick, L. H. Kuller, O. L. Lopez, C. H. Kawas, and W. Jagust, Survival following dementia onset: Alzheimer's disease and vascular dementia, J. Neurol. Sci. 229, vol.230, pp.43-49, 2005.

S. Fleminger, D. L. Oliver, S. Lovestone, S. Rabe-hesketh, and A. Giora, Head injury as a risk factor for Alzheimer's disease: the evidence 10 years on; a partial replication, J. Neurol. Neurosurg. Psychiatry, vol.74, pp.857-862, 2003.

N. C. Foley, R. H. Affoo, R. E. Martin, A. M. Fontainhas, M. Wang et al., A systematic review and meta-analysis examining pneumonia-associated mortality in dementia, Microglial morphology and dynamic behavior is regulated by ionotropic glutamatergic and GABAergic neurotransmission, vol.39, pp.52-67, 2011.

O. V. Forlenza, B. S. Diniz, L. L. Talib, V. A. Mendonça, E. B. Ojopi et al., Increased serum IL-1beta level in Alzheimer's disease and mild cognitive impairment, Dement. Geriatr. Cogn. Disord, vol.6, pp.507-512, 2009.

J. Frackowiak, H. M. Wisniewski, J. Wegiel, G. S. Merz, K. Iqbal et al., , 1992.

, Acta Neuropathol. (Berl.), vol.84, pp.225-233

R. P. Friedland, T. Fritsch, K. A. Smyth, E. Koss, A. J. Lerner et al., Patients with Alzheimer's disease have reduced activities in midlife compared with healthy control-group members, Proc. Natl. Acad. Sci. U. S. A, vol.98, pp.3440-3445, 2001.

D. Galimberti, C. Fenoglio, C. Lovati, E. Venturelli, I. Guidi et al., , 2006.

, Serum MCP-1 levels are increased in mild cognitive impairment and mild Alzheimer's disease, Neurobiol. Aging, vol.27, pp.1763-1768

L. Gasparini, E. Ongini, and G. Wenk, , 2004.

, Alzheimer's disease: old and new mechanisms of action, Non-steroidal anti-inflammatory drugs (NSAIDs) in, vol.91, pp.521-536

D. S. Geldmacher, T. Fritsch, M. J. Mcclendon, and G. Landreth, A randomized pilot clinical trial of the safety of pioglitazone in treatment of patients with Alzheimer disease, Arch. Neurol, vol.68, pp.45-50, 2011.

C. Gemma, A. D. Bachstetter, C. Gemma, A. D. Bachstetter, M. J. Cole et al., Blockade of caspase-1 increases neurogenesis in the aged hippocampus, Front. Cell. Neurosci, vol.7, pp.2795-2803, 2007.

H. C. Gérard, K. L. Wildt, J. A. Whittum-hudson, Z. Lai, J. Ager et al., The load of Chlamydia pneumoniae in the Alzheimer's brain varies with APOE genotype, Microb. Pathog, vol.39, pp.19-26, 2005.

H. C. Gérard, U. Dreses-werringloer, K. S. Wildt, S. Deka, C. Oszust et al., Chlamydophila (Chlamydia) pneumoniae in the Alzheimer's brain, FEMS Immunol. Med. Microbiol, vol.48, pp.355-366, 2006.

J. Gerson, D. L. Castillo-carranza, U. Sengupta, R. Bodani, D. S. Prough et al., Tau Oligomers Derived from Traumatic Brain Injury Cause Cognitive Impairment and Accelerate Onset of Pathology in Htau Mice, J. Neurotrauma, vol.33, pp.2034-2043, 2016.

F. Ginhoux, M. Greter, M. Leboeuf, S. Nandi, P. See et al., Fate mapping analysis reveals that adult microglia derive from primitive macrophages, Science, vol.330, pp.841-845, 2010.

B. Giunta, J. Deng, J. Jin, E. Sadic, S. Rum et al., EVALUATION OF HOW CIGARETTE SMOKE IS A DIRECT RISK FACTOR FOR ALZHEIMER'S DISEASE, Technol. Innov, vol.14, pp.39-48, 2012.

M. P. Zwiers, D. G. Norris, and F. De-leeuw,

, J. Neurol, vol.134, pp.2116-2124

H. F. Green, Y. M. Nolan, K. N. Green, L. M. Billings, B. Roozendaal et al., Unlocking mechanisms in interleukin-1?-induced changes in hippocampal neurogenesis--a role for GSK-3? and TLX. Transl. Psychiatry 2, e194, 2006.

, Glucocorticoids increase amyloid-beta and tau pathology in a mouse model of Alzheimer's disease, J. Neurosci. Off. J. Soc. Neurosci, vol.26, pp.9047-9056

A. Griciuc, A. Serrano-pozo, A. R. Parrado, A. N. Lesinski, C. N. Asselin et al.,

J. Guadagno, P. Swan, R. Shaikh, S. P. Cregan, M. Guillot-sestier et al., Microglia-derived IL-1? triggers p53-mediated cell cycle arrest and apoptosis in neural precursor cells, Cell Death Dis, vol.6, pp.534-548, 1779.

A. Halle, V. Hornung, G. C. Petzold, C. R. Stewart, B. G. Monks et al.,

L. Hamelin, J. Lagarde, G. Dorothée, C. Leroy, M. Labit et al., Early and protective microglial activation in Alzheimer's disease: a prospective study using 18F-DPA-714 PET imaging, Brain J. Neurol, vol.139, pp.1252-1264, 2016.

R. Haque and A. Nazir, Insulindegrading enzyme: a link between Alzheimer's and type 2 diabetes mellitus, CNS Neurol. Disord. Drug Targets, vol.13, pp.259-264, 2014.

J. K. Harrison, Y. Jiang, S. Chen, Y. Xia, D. Maciejewski et al., Role for neuronally derived fractalkine in mediating interactions between neurons and CX3CR1-expressing microglia, Proc. Natl. Acad. Sci. U. S. A, vol.95, pp.10896-10901, 1998.

G. J. Harry, Microglia during development and aging, Pharmacol. Ther, vol.139, pp.313-326, 2013.

C. J. Hauser, T. Sursal, E. K. Rodriguez, P. T. Appleton, Q. Zhang et al., , 2010.

M. He, H. Dong, Y. Huang, S. Lu, S. Zhang et al., Mitochondrial damage associated molecular patterns from femoral reamings activate neutrophils through formyl peptide receptors and P44/42 MAP kinase, Cell. Physiol. Biochem, vol.24, pp.859-870, 2016.

K. Heese, C. Hock, and U. Otten, Inflammatory signals induce neurotrophin expression in human microglial cells, J. Neurochem, vol.70, pp.699-707, 1998.

J. K. Hefendehl, J. J. Neher, R. B. Sühs, S. Kohsaka, A. Skodras et al., , 2014.

, Homeostatic and injury-induced microglia behavior in the aging brain, Aging Cell, vol.13, pp.60-69

S. Hellwig, A. Heinrich, K. Biber, M. T. Heneka, H. Wiesinger et al., Neuronal and glial coexpression of argininosuccinate synthetase and inducible nitric oxide synthase in Alzheimer disease, J. Neuropathol. Exp. Neurol, vol.7, pp.906-916, 2001.

M. T. Heneka, G. E. Landreth, and D. L. Feinstein, Role for peroxisome proliferatoractivated receptor-gamma in Alzheimer's disease, 2001.

M. T. Heneka, E. Galea, V. Gavriluyk, L. Dumitrescu-ozimek, J. Daeschner et al., Noradrenergic depletion potentiates beta -amyloid-induced cortical inflammation: implications for Alzheimer's disease, J. Neurosci. Off. J. Soc. Neurosci, vol.49, pp.2434-2442, 2002.

M. T. Heneka, V. Gavrilyuk, G. E. Landreth, M. K. O&apos;banion, G. Weinberg et al., Noradrenergic depletion increases inflammatory responses in brain: effects on IkappaB and HSP70 expression, J. Neurochem, vol.85, pp.387-398, 2003.

M. T. Heneka, M. Sastre, L. Dumitrescu-ozimek, A. Hanke, I. Dewachter et al., Acute treatment with the PPARgamma agonist pioglitazone and ibuprofen reduces glial inflammation and Abeta1-42 levels in APPV717I transgenic mice, Brain J. Neurol, vol.128, pp.1442-1453, 2005.

M. T. Heneka, M. Ramanathan, A. H. Jacobs, L. Dumitrescu-ozimek, A. Bilkei-gorzo et al., Locus ceruleus degeneration promotes Alzheimer pathogenesis in amyloid precursor protein 23 transgenic mice, J. Neurosci. Off. J. Soc. Neurosci, vol.26, pp.1343-1354, 2006.

M. T. Heneka, F. Nadrigny, T. Regen, A. Martinez-hernandez, L. Dumitrescu-ozimek et al., Locus ceruleus controls Alzheimer's disease pathology by modulating microglial functions through norepinephrine, Proc. Natl. Acad. Sci. U. S. A, vol.107, pp.6058-6063, 2010.

M. T. Heneka, M. P. Kummer, A. Stutz, A. Delekate, S. Schwartz et al.,

M. T. Heneka, D. T. Golenbock, and E. Latz, Innate immunity in Alzheimer's disease, 2015.

, Nat. Immunol, vol.16, pp.229-236

M. T. Heneka, A. Fink, and G. Doblhammer, Effect of pioglitazone medication on the incidence of dementia, Ann. Neurol, vol.78, pp.284-294, 2015.

S. E. Hickman, E. K. Allison, and J. Khoury, Microglial dysfunction and defective betaamyloid clearance pathways in aging Alzheimer's disease mice, J. Neurosci. Off. J. Soc. Neurosci, vol.28, pp.8354-8360, 2008.

Y. Ho, X. Yang, S. Yeung, K. Chiu, C. Lau et al., Cigarette Smoking Accelerated Brain Aging and Induced Pre-Alzheimer-Like Neuropathology in Rats, J. Neurol. Neurosurg. Psychiatry, vol.7, pp.788-789, 2003.

C. Holmes, C. Cunningham, E. Zotova, J. Woolford, C. Dean et al., Systemic inflammation and disease progression in Alzheimer disease, Neurology, vol.73, pp.768-774, 2009.

S. Holmseth, H. A. Scott, K. Real, K. P. Lehre, T. B. Leergaard et al., The concentrations and distributions of three C-terminal variants of the GLT1, 2009.

, Neuroscience, vol.162, pp.1055-1071

S. Hong, V. F. Beja-glasser, B. M. Nfonoyim, A. Frouin, S. Li et al., , 2016.

K. Honjo, R. Van-reekum, and N. P. Verhoeff, Alzheimer's disease and infection: do infectious agents contribute to progression of Alzheimer's disease?, Alzheimers Dement. J. Alzheimers Assoc, vol.5, pp.348-360, 2009.

W. T. Hu, D. M. Holtzman, A. M. Fagan, L. M. Shaw, R. Perrin et al., Plasma multianalyte profiling in mild cognitive impairment and Alzheimer disease, Neurology, vol.79, pp.897-905, 2012.

H. Huang, L. Wang, M. Cao, C. Marshall, J. Gao et al., , 2015.

, Isolation Housing Exacerbates Alzheimer's Disease-Like Pathophysiology in Aged APP/PS1

. Mice, . J. Int, N. C. Inestrosa, E. M. Toledo, R. F. Itzhaki et al., The role of Wnt signaling in neuronal dysfunction in Alzheimer's Disease, Mol. Neurodegener, vol.18, 1997.

T. J. Iwashyna, E. W. Ely, D. M. Smith, and K. M. Langa, Long-term cognitive impairment and functional disability among survivors of severe sepsis, Herpes simplex virus type 1 in brain and risk of Alzheimer's disease, vol.349, pp.1787-1794, 2010.

A. H. Jacobs and B. Tavitian, Noninvasive molecular imaging of neuroinflammation, J. Cereb. Blood Flow Metab. Off. J. Int. Soc. Cereb. Blood Flow Metab, vol.32, pp.1393-1415, 2012.

J. B. Jahrling, C. M. Hernandez, L. Denner, and K. T. Dineley, PPAR? recruitment to active ERK during memory consolidation is required for Alzheimer's disease-related cognitive enhancement, J. Neurosci. Off. J. Soc. Neurosci, vol.34, pp.4054-4063, 2014.

H. E. Jakobsson, A. M. Rodríguez-piñeiro, A. Schütte, A. Ermund, P. Boysen et al., The composition of the gut microbiota shapes the colon mucus barrier, EMBO Rep, vol.16, pp.164-177, 2015.

G. A. Jamieson, N. J. Maitland, G. K. Wilcock, J. Craske, and R. F. Itzhaki, Latent herpes simplex virus type 1 in normal and Alzheimer's disease brains, J. Med. Virol, vol.33, pp.224-227, 1991.

G. A. Jamieson, N. J. Maitland, G. K. Wilcock, C. M. Yates, and R. F. Itzhaki, Herpes simplex virus type 1 DNA is present in specific regions of brain from aged people with and without senile dementia of the Alzheimer type, J. Pathol, vol.167, pp.365-368, 1992.

C. S. Jensen, S. G. Hasselbalch, G. Waldemar, A. H. Simonsen, Y. H. Jeong et al., Chronic stress accelerates learning and memory impairments and increases amyloid deposition in APPV717I-CT100 transgenic mice, Front. Neurol, vol.6, 2006.

, FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol, vol.20, pp.729-731

K. Ji, G. Akgul, L. P. Wollmuth, S. E. Tsirka, L. Johansson et al., Midlife personality and risk of Alzheimer disease and distress: a 38-year followup, Neurology, vol.8, pp.1538-1544, 2013.

V. E. Johnson, W. Stewart, and D. H. Smith,

, Pathol. Zurich Switz, vol.22, pp.142-149

R. Kandimalla, V. Thirumala, and P. H. Reddy, Is Alzheimer's disease a Type 3 Diabetes? A critical appraisal, Biochim. Biophys. Acta, vol.1863, pp.1078-1089, 2017.

M. J. Kane, M. Angoa-pérez, D. I. Briggs, D. C. Viano, C. W. Kreipke et al., A mouse model of human repetitive mild traumatic brain injury, J. Neurosci. Methods, vol.203, pp.41-49, 2012.

V. Kaushal and L. C. Schlichter, Mechanisms of microglia-mediated neurotoxicity in a new model of the stroke penumbra, J. Neurosci. Off. J. Soc. Neurosci, vol.28, pp.2221-2230, 2008.

H. Kenche, C. J. Baty, K. Vedagiri, S. D. Shapiro, and A. Perry, , 2013.

, FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol, vol.27, pp.965-977

K. Kierdorf, D. Erny, T. Goldmann, V. Sander, C. Schulz et al.,

, Microglia emerge from erythromyeloid precursors via Pu.1-and Irf8-dependent pathways

, Nat. Neurosci, vol.16, pp.273-280

B. Kim, M. Shin, C. Kim, S. Baek, Y. Ko et al., Exerc. Rehabil, vol.10, pp.2-8, 2014.

T. Kim, H. Lim, J. Y. Lee, D. Kim, S. Park et al., , 2008.

, Alzheimer's disease, Neurosci. Lett, vol.436, pp.196-200

P. J. Kingham, M. L. Cuzner, and J. M. Pocock, Apoptotic pathways mobilized in microglia and neurones as a consequence of chromogranin A-induced microglial activation, J. Neurochem, vol.73, pp.538-547, 1999.

T. Kiyota, M. Yamamoto, H. Xiong, M. P. Lambert, W. L. Klein et al., Microglial phagocytosis induced by fibrillar beta-amyloid and IgGs are differentially regulated by proinflammatory cytokines, J. Neurosci. Off. J. Soc. Neurosci, vol.4, pp.8240-8249, 2005.

J. W. Koo and R. S. Duman, IL-1beta is an essential mediator of the antineurogenic and anhedonic effects of stress, Proc. Natl. Acad. Sci. U. S. A, vol.105, pp.751-756, 2008.

Y. Koronyo, B. C. Salumbides, J. Sheyn, L. Pelissier, S. Li et al., Therapeutic effects of glatiramer acetate and grafted CD115 + monocytes in a mouse model of Alzheimer's disease, Brain J. Neurol, vol.138, pp.2399-2422, 2015.

G. Krabbe, A. Halle, V. Matyash, J. L. Rinnenthal, G. D. Eom et al., Functional Impairment of Microglia Coincides with Beta-Amyloid Deposition in Mice with Alzheimer-Like Pathology, Brain J. Neurol, vol.8, pp.2228-2238, 2013.

Z. Kroner, The relationship between Alzheimer's disease and diabetes: Type 3 diabetes?, Altern. Med. Rev. J. Clin. Ther, vol.14, pp.373-379, 2009.

T. Kukar, S. Prescott, J. L. Eriksen, V. Holloway, M. P. Murphy et al., Chronic administration of R-flurbiprofen attenuates learning impairments in transgenic amyloid precursor protein mice, BMC Neurosci, vol.8, pp.833-844, 2007.

M. P. Kummer, T. Hammerschmidt, A. Martinez, D. Terwel, G. Eichele et al., , 2014.

, Ear2 deletion causes early memory and learning deficits in APP/PS1 mice, J. Neurosci. Off. J. Soc. Neurosci, vol.34, pp.8845-8854

G. E. Landreth and M. T. Heneka, Antiinflammatory actions of peroxisome proliferatoractivated receptor gamma agonists in Alzheimer's disease, Neurobiol. Aging, vol.22, pp.937-944, 2001.

S. Lavisse, M. Guillermier, A. Hérard, F. Petit, M. Delahaye et al., , 2012.

, Reactive astrocytes overexpress TSPO and are detected by TSPO positron emission tomography imaging, J. Neurosci. Off. J. Soc. Neurosci, vol.32, pp.10809-10818

D. C. Lee, J. Rizer, M. B. Selenica, P. Reid, C. Kraft et al., LPSinduced inflammation exacerbates phospho-tau pathology in rTg4510 mice, J. Neuroinflammation, vol.7, 2009.

, J. Neurosci. Res, vol.87, pp.2561-2570

Y. Li, X. Du, C. Liu, Z. Wen, and J. Du, Reciprocal regulation between resting microglial dynamics and neuronal activity in vivo, Dev. Cell, vol.23, pp.1189-1202, 2012.

G. P. Lim, F. Yang, T. Chu, E. Gahtan, O. Ubeda et al., , 2001.

G. Liu, R. J. Middleton, C. R. Hatty, W. W. Kam, .. Chan et al., The 18 kDa translocator protein, microglia and neuroinflammation, Brain Pathol. Zurich Switz, vol.24, pp.631-653, 2014.

S. Liu, Y. Liu, W. Hao, L. Wolf, A. J. Kiliaan et al., , 2012.

. Baltim and . Md, , vol.188, pp.1098-1107, 1950.

J. M. Lizcano, A. , and D. R. , The insulin signalling pathway, Curr. Biol. CB, vol.12, pp.236-238, 2002.

J. A. Luchsinger, M. Tang, S. Shea, R. Mayeux, S. Dj et al., Antioxidant Vitamin Intake and Risk of Alzheimer Disease, Arch. Neurol, vol.60, p.203, 2003.

, Progranulin Deficiency Promotes Circuit-Specific Synaptic Pruning by Microglia via Complement Activation, Cell, vol.165, pp.921-935

M. Luong, Y. Zhang, T. Chamberlain, T. Zhou, J. F. Wright et al., Amyloid-beta protein oligomer at low nanomolar concentrations activates microglia and induces microglial neurotoxicity, J. Inflamm. Lond. Engl, vol.9, pp.3693-3706, 2011.

S. Magaki, W. H. Yong, N. Khanlou, S. Tung, and H. V. Vinters, Comorbidity in dementia: update of an ongoing autopsy study, J. Am. Geriatr. Soc, vol.62, pp.1722-1728, 2014.

L. Maggi, M. Scianni, I. Branchi, I. D&apos;andrea, C. Lauro et al., CX(3)CR1 deficiency alters hippocampal-dependent plasticity phenomena blunting the effects of enriched environment, Expert Opin. Ther. Targets, vol.5, pp.1085-1097, 2011.

P. Mao, M. Manczak, M. J. Calkins, Q. Truong, T. P. Reddy et al., , 2012.

, Mitochondria-targeted catalase reduces abnormal APP processing, amyloid ? production and BACE1 in a mouse model of Alzheimer's disease: implications for neuroprotection and lifespan extension, Hum. Mol. Genet, vol.21, pp.2973-2990

K. Marosi, Z. Bori, N. Hart, L. Sárga, E. Koltai et al., Long-term exercise treatment reduces oxidative stress in the hippocampus of aging rats, Neuroscience, vol.226, pp.21-28, 2012.

E. Masliah, M. Alford, R. Deteresa, M. Mallory, and L. Hansen, Deficient glutamate transport is associated with neurodegeneration in Alzheimer's disease, Ann. Neurol, vol.40, pp.759-766, 1996.

M. Mathieu, N. Sawyer, G. M. Greig, M. Hamel, S. Kargman et al., The C3a receptor antagonist SB 290157 has agonist activity, Immunol. Lett, vol.100, pp.139-145, 2005.

K. M. Mcateer, F. Corrigan, E. Thornton, R. J. Turner, R. Vink et al., Short and Long Term Behavioral and Pathological Changes in a Novel Rodent Model of Repetitive Mild Traumatic Brain Injury, PLOS ONE, vol.11, 2008.

, Ibuprofen reduces Abeta, hyperphosphorylated tau and memory deficits in Alzheimer mice, Brain Res, vol.1207, pp.225-236

R. M. Mcmanus and M. T. Heneka, Role of neuroinflammation in neurodegeneration: new insights, Chronic disease long-term drug prevention trials: lessons from the Alzheimer's Disease Antiinflammatory Prevention Trial (ADAPT), vol.9, 2008.

, Alzheimers Dement. J. Alzheimers Assoc, vol.4, pp.7-14

J. J. Miguel-hidalgo, X. A. Alvarez, R. Cacabelos, and G. Quack, Neuroprotection by memantine against neurodegeneration induced by beta-amyloid(1-40), Brain Res, vol.958, pp.210-221, 2002.

S. Mitew, M. T. Kirkcaldie, G. M. Halliday, C. E. Shepherd, J. C. Vickers et al., Focal demyelination in Alzheimer's disease and transgenic mouse models, Acta Neuropathol. (Berl.), vol.119, pp.567-577, 2010.

C. Moran, T. G. Phan, J. Chen, L. Blizzard, R. Beare et al., Brain atrophy in type 2 diabetes: regional distribution and influence on cognition, Diabetes Care, vol.36, pp.4036-4042, 2013.

O. Moran, F. Conti, and P. Tammaro, Sodium channel heterologous expression in mammalian cells and the role of the endogenous beta1-subunits, Neurosci. Lett, vol.336, pp.175-179, 2003.

P. I. Moreira, C. Carvalho, X. Zhu, M. A. Smith, P. et al., Mitochondrial dysfunction is a trigger of Alzheimer's disease pathophysiology, Biochim. Biophys. Acta, vol.1802, pp.2-10, 2010.

I. Moreno-gonzalez, L. D. Estrada, E. Sanchez-mejias, C. Soto, T. Mori et al., Smoking exacerbates amyloid pathology in a mouse model of Alzheimer's disease, Nat. Commun, vol.4, 1495.

M. C. Morris, Nutritional determinants of cognitive aging and dementia, Proc. Nutr. Soc, vol.71, pp.1-13, 2012.

M. C. Morris, C. C. Tangney, Y. Wang, F. M. Sacks, L. L. Barnes et al., MIND diet slows cognitive decline with aging, Alzheimers Dement. J. Alzheimers Assoc, vol.11, pp.1015-1022, 2015.

M. C. Morris, C. C. Tangney, Y. Wang, F. M. Sacks, D. A. Bennett et al., MIND diet associated with reduced incidence of Alzheimer's disease, Alzheimers Dement. J. Alzheimers Assoc, vol.11, pp.1007-1014, 2015.

M. C. Morris, C. C. Tangney, Y. Wang, F. M. Sacks, L. L. Barnes et al., MIND diet slows cognitive decline with aging, Alzheimers Dement. J. Alzheimers Assoc, vol.11, pp.1015-1022, 2015.

J. A. Mortimer, C. M. Van-duijn, V. Chandra, L. Fratiglioni, A. B. Graves et al., Head trauma as a risk factor for Alzheimer's disease: a collaborative re-analysis of case-control studies. EURODEM Risk Factors Research Group, Int. J. Epidemiol, vol.20, issue.2, pp.28-35, 1991.

R. G. Nagele, M. R. D&apos;andrea, H. Lee, V. Venkataraman, and H. Wang, , 2003.

C. M. Nascimento, J. R. Pereira, L. P. De-andrade, M. Garuffi, L. L. Talib et al., Physical exercise in MCI elderly promotes reduction of pro-inflammatory cytokines and improvements on cognition and BDNF peripheral levels, Astrocytes accumulate A beta 42 and give rise to astrocytic amyloid plaques in Alzheimer disease brains, vol.971, pp.799-805, 2014.

K. R. Nash, D. C. Lee, J. B. Hunt, J. M. Morganti, M. L. Selenica et al., Fractalkine overexpression suppresses tau pathology in a mouse model of tauopathy, Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol, vol.34, pp.318-333, 2002.

A. Okello, P. Edison, H. A. Archer, F. E. Turkheimer, J. Kennedy et al., , 2009.

, Microglial activation and amyloid deposition in mild cognitive impairment: a PET study, Neurology, vol.72, pp.56-62

M. Orre, W. Kamphuis, L. M. Osborn, A. H. Jansen, L. Kooijman et al., Isolation of glia from Alzheimer's mice reveals inflammation and dysfunction, 2014.

, Aging, vol.35, pp.2746-2760

D. R. Owen, A. J. Yeo, R. N. Gunn, K. Song, G. Wadsworth et al., An 18-kDa translocator protein (TSPO) polymorphism explains differences in binding affinity of the PET radioligand PBR28, J. Cereb. Blood Flow Metab. Off. J. Int. Soc. Cereb. Blood Flow Metab, vol.32, pp.1-5, 2012.

S. L. Payão, G. M. Gonçalves, R. W. De-labio, L. Horiguchi, I. Mizumoto et al., Association of interleukin 1? polymorphisms and haplotypes with Alzheimer's disease, J. Neuroimmunol, vol.247, pp.59-62, 2012.

B. G. Perez-nievas, T. Hammerschmidt, M. P. Kummer, D. Terwel, J. C. Leza et al., , 2011.

V. H. Perry, Contribution of systemic inflammation to chronic neurodegeneration, Acta Neuropathol. (Berl.), vol.120, pp.277-286, 2010.

A. L. Petraglia, B. A. Plog, S. Dayawansa, M. L. Dashnaw, K. Czerniecka et al.,

D. Pisa, R. Alonso, A. Rábano, I. Rodal, L. Carrasco et al., Direct visualization of fungal infection in brains from patients with Alzheimer's disease, J. Alzheimers Dis. JAD, vol.5, pp.613-624, 2015.

J. M. Pocock and H. Kettenmann, , 2007.

, Neurotransmitter receptors on microglia, Trends Neurosci, vol.30, pp.527-535

L. J. Podewils, E. Guallar, L. H. Kuller, L. P. Fried, O. L. Lopez et al., Physical activity, APOE genotype, and dementia risk: findings from the Cardiovascular Health Cognition Study, Am. J. Epidemiol, vol.161, pp.639-651, 2005.

M. Popoli, Z. Yan, B. S. Mcewen, and G. Sanacora, The stressed synapse: the impact of stress and glucocorticoids on glutamate transmission, Nat. Rev. Neurosci, vol.13, pp.22-37, 2011.

V. Porrini, A. Lanzillotta, C. Branca, M. Benarese, E. Parrella et al., , 2015.

, CSP-1103) induces microglia alternative activation in plaque-free Tg2576 mice and primary glial cultures exposed to betaamyloid, Neuroscience, vol.302, pp.112-120

M. Prinz and J. Priller, Microglia and brain macrophages in the molecular age: from origin to neuropsychiatric disease, Nat. Rev. Neurosci, vol.15, pp.300-312, 2014.

S. Prokop, K. R. Miller, N. Drost, S. Handrick, V. Mathur et al., Impact of peripheral myeloid cells on amyloid-? pathology in Alzheimer's disease-like mice, J. Exp. Med, vol.212, pp.1811-1818, 2015.

W. Q. Qiu and M. F. Folstein, Insulin, insulin-degrading enzyme and amyloid-beta peptide in Alzheimer's disease: review and hypothesis, Neurobiol. Aging, vol.27, pp.190-198, 2006.

Z. Radak, N. Hart, L. Sarga, E. Koltai, M. Atalay et al., Exercise plays a preventive role against Alzheimer's disease, J. Alzheimers Dis. JAD, vol.20, pp.777-783, 2010.

D. D. Raj, D. Jaarsma, I. R. Holtman, M. Olah, F. M. Ferreira et al., CD14 and toll-like receptors 2 and 4 are required for fibrillar A{beta}-stimulated microglial activation, J. Neurosci. Off. J. Soc. Neurosci, vol.35, pp.11982-11992, 2009.

R. Reshef, T. Kreisel, D. Beroukhim-kay, Y. , and R. , Microglia and their CX3CR1 signaling are involved in hippocampal-but not olfactory bulb-related memory and neurogenesis, Brain. Behav. Immun, vol.41, pp.239-250, 2014.

K. Riazi, M. A. Galic, J. B. Kuzmiski, W. Ho, K. A. Sharkey et al., , 2008.

, Microglial activation and TNFalpha production mediate altered CNS excitability following peripheral inflammation, Proc. Natl. Acad. Sci. U. S. A, vol.105, pp.17151-17156

M. Ries, M. ;. Sastre, M. E. Risner, A. M. Saunders, J. F. Altman et al., Mechanisms of A? Clearance and Degradation by Glial Cells. Front. Aging Neurosci. 8, 2016.

E. Rodriguez-vieitez, L. Saint-aubert, S. F. Carter, O. Almkvist, K. Farid et al., Diverging longitudinal changes in astrocytosis and amyloid PET in autosomal dominant Alzheimer's disease, Brain J. Neurol, vol.139, pp.922-936, 2016.

J. Rogers, L. C. Kirby, S. R. Hempelman, D. L. Berry, P. L. Mcgeer et al.,

, Clinical trial of indomethacin in Alzheimer's disease, Neurology, vol.43, pp.1609-1611

J. T. Rogers, J. M. Morganti, A. D. Bachstetter, C. E. Hudson, M. M. Peters et al.,

, Neurosci. Off. J. Soc. Neurosci, vol.31, pp.16241-16250

S. M. Rothman, N. Herdener, S. Camandola, S. J. Texel, M. R. Mughal et al., 3xTgAD mice exhibit altered behavior and elevated A? after chronic mild social stress, Neurobiol. Aging, vol.33, pp.1-12, 2012.

S. Rovio, I. Kåreholt, E. Helkala, M. Viitanen, B. Winblad et al., Leisuretime physical activity at midlife and the risk of dementia and Alzheimer's disease, Lancet Neurol, vol.4, pp.705-711, 2005.

A. Salminen, J. Ojala, A. Kauppinen, K. Kaarniranta, and T. Suuronen, Inflammation in Alzheimer's disease: amyloidbeta oligomers trigger innate immunity defence via pattern recognition receptors, Prog. Neurobiol, vol.87, pp.181-194, 2009.

A. F. Santillo, J. P. Gambini, L. Lannfelt, B. Långström, L. Ulla-marja et al., In vivo imaging of astrocytosis in Alzheimer's disease: an 11 C-L-deuteriodeprenyl and PIB PET study, Eur. J. Nucl. Med. Mol. Imaging, vol.38, pp.2202-2208, 2011.

T. Sato, H. Hanyu, K. Hirao, H. Kanetaka, H. Sakurai et al., Efficacy of PPAR-? agonist pioglitazone in mild Alzheimer disease, Neurobiol. Aging, vol.32, pp.1626-1633, 2011.

N. Scarmeas, Y. Stern, R. Mayeux, and J. A. Luchsinger, Mediterranean diet, Alzheimer disease, and vascular mediation, Arch. Neurol, vol.63, pp.1709-1717, 2006.

N. Scarmeas, Y. Stern, M. Tang, R. Mayeux, J. A. Luchsinger et al., Physical activity, diet, and risk of Alzheimer disease, Ann. Neurol, vol.59, pp.627-637, 2006.

F. M. Schmidt, J. Weschenfelder, C. Sander, J. Minkwitz, J. Thormann et al., Inflammatory cytokines in general and central obesity and modulating effects of physical activity, Med. Sci. Sports Exerc, vol.10, pp.772-777, 2001.

F. L. Sciacca, C. Ferri, F. Licastro, F. Veglia, I. Biunno et al., , 2003.

, Interleukin-1B polymorphism is associated with age at onset of Alzheimer's disease, Neurobiol. Aging, vol.24, pp.927-931

A. Scimemi, J. S. Meabon, R. L. Woltjer, J. M. Sullivan, J. S. Diamond et al., Amyloid-?1-42 slows clearance of synaptically released glutamate by mislocalizing astrocytic GLT-1, J. Neurosci. Off. J. Soc. Neurosci, vol.33, pp.5312-5318, 2013.

H. A. Scott, F. M. Gebhardt, A. D. Mitrovic, R. J. Vandenberg, and P. R. Dodd, , 2011.

, Glutamate transporter variants reduce glutamate uptake in Alzheimer's disease, Neurobiol. Aging, vol.32, pp.1-11

R. C. Seet, C. J. Lee, W. M. Loke, S. H. Huang, H. Huang et al., Biomarkers of oxidative damage in cigarette smokers: which biomarkers might reflect acute versus chronic oxidative stress? Free Radic, Biol. Med, vol.50, pp.1787-1793, 2011.

S. Sellner, R. Paricio-montesinos, A. Spieß, A. Masuch, D. Erny et al., Involvement of inflammation in Alzheimer's disease pathogenesis and therapeutic potential of anti-inflammatory agents, Acta Neuropathol. Commun, vol.4, pp.2106-2119, 2015.

C. Shen, Y. Chen, H. Liu, K. Zhang, T. Zhang et al., Hydrogen Peroxide Promotes A? Production through JNK-dependent Activation of ?-Secretase, J. Biol. Chem, vol.283, pp.17721-17730, 2008.

J. Shi, W. Shen, J. Chen, B. Wang, L. Zhong et al., Anti-TNF-? reduces amyloid plaques and tau phosphorylation and induces CD11c-positive dendritic-like cell in the APP/PS1 transgenic mouse brains, Brain Res, vol.1368, pp.239-247, 2011.

J. Shi, B. Wang, W. Jiang, J. Chen, Y. Zhu et al., Cognitive improvement with intrathecal administration of infliximab in a woman with Alzheimer's disease, J. Am. Geriatr, 2011.

. Soc, , vol.59, pp.1142-1144

H. Shishido, Y. Kishimoto, N. Kawai, Y. Toyota, M. Ueno et al., Traumatic brain injury accelerates amyloid-? deposition and impairs spatial learning in the triple-transgenic mouse model of Alzheimer's disease, Neurosci. Lett, vol.629, pp.62-67, 2016.

S. E. Shoelson, J. Lee, and A. B. Goldfine, Inflammation and insulin resistance, J. Clin. Invest, vol.116, pp.1793-1801, 2006.

A. Sierra, A. Gottfried-blackmore, T. A. Milner, B. S. Mcewen, and K. Bulloch, Steroid hormone receptor expression and function in microglia, Glia, vol.56, pp.659-674, 2008.

A. Sierra, J. M. Encinas, J. J. Deudero, J. H. Chancey, G. Enikolopov et al., Microglia shape adult hippocampal neurogenesis through apoptosis-coupled phagocytosis, Cell Stem Cell, vol.7, pp.483-495, 2010.

A. R. Simard, D. Soulet, G. Gowing, J. Julien, and S. Rivest, Bone marrow-derived, 2006.

M. Simons, F. Schwärzler, D. Lütjohann, K. Von-bergmann, K. Beyreuther et al., Treatment with simvastatin in normocholesterolemic patients with Alzheimer's disease: A 26-week randomized, placebocontrolled, double-blind trial, J. Neuropathol. Exp. Neurol, vol.52, pp.982-992, 1999.

D. H. Smith, V. E. Johnson, and W. Stewart, Chronic neuropathologies of single and repetitive TBI: substrates of dementia?, Nat. Rev. Neurol, vol.9, pp.211-221, 2013.

J. C. Smith, K. A. Nielson, J. L. Woodard, M. Seidenberg, and S. M. Rao, Physical Activity and Brain Function in Older Adults at Increased Risk for Alzheimer's Disease, Brain Sci, vol.3, pp.54-83, 2013.

A. Sokolova, M. D. Hill, F. Rahimi, L. A. Warden, G. M. Halliday et al., , 2009.

, Monocyte chemoattractant protein-1 plays a dominant role in the chronic inflammation observed in Alzheimer's disease, Brain Pathol, vol.19, pp.392-398

C. M. Sondag, G. Dhawan, C. K. Combs, D. L. Sparks, M. N. Sabbagh et al., Beta amyloid oligomers and fibrils stimulate differential activation of primary microglia, J. Neuroinflammation, vol.6, pp.753-757, 2005.

J. Stefaniak and J. Brien, Imaging of neuroinflammation in dementia: a review, J. Neurol. Neurosurg. Psychiatry, vol.87, pp.21-28, 2016.

D. Stellwagen and R. C. Malenka, , 2006.

, Synaptic scaling mediated by glial TNF-alpha, Nature, vol.440, pp.1054-1059

W. F. Stewart, C. Kawas, M. Corrada, and E. J. Metter, Risk of Alzheimer's disease and duration of NSAID use, Neurology, vol.48, pp.626-632, 1997.

F. Su, F. Bai, and Z. Zhang, , 2016.

, Inflammatory Cytokines and Alzheimer's Disease: A Review from the Perspective of Genetic Polymorphisms, Neurosci. Bull, vol.32, pp.469-480

I. Suridjan, B. G. Pollock, N. P. Verhoeff, A. N. Voineskos, T. Chow et al., In-vivo imaging of grey and white matter neuroinflammation in Alzheimer's disease: a positron emission tomography study with a novel radioligand, Mol. Psychiatry, vol.20, pp.1579-1587, 2015.

K. Tahara, H. Kim, J. Jin, J. A. Maxwell, L. Li et al., Role of toll-like receptor signalling in Abeta uptake and clearance, Brain J. Neurol, vol.129, pp.3006-3019, 2006.

K. Takata, Y. Kitamura, J. Kakimura, K. Shibagaki, D. Tsuchiya et al., Role of high mobility group protein-1 (HMG1) in amyloid-beta homeostasis, Biochem. Biophys. Res. Commun, vol.301, pp.699-703, 2003.

, Inflammatory markers and the risk of Alzheimer disease The Framingham Study, Neurology, vol.68, pp.1902-1908

K. F. Tanaka, H. Kashima, H. Suzuki, K. Ono, and M. Sawada, Existence of functional beta1-and beta2-adrenergic receptors on microglia, J. Neurosci. Res, vol.70, pp.232-237, 2002.

C. C. Tangney, M. J. Kwasny, H. Li, R. S. Wilson, D. A. Evans et al., , 2011.

, Am. J. Clin. Nutr, vol.93, pp.601-607

C. C. Tangney, H. Li, Y. Wang, L. Barnes, J. A. Schneider et al., Relation of DASH-and Mediterraneanlike dietary patterns to cognitive decline in older persons, Neurology, vol.83, pp.1410-1416, 2014.

L. Taupenot, J. E. Remacle, K. B. Helle, D. Aunis, and M. F. Bader, Recombinant human chromogranin A: expression, purification and characterization of the N-terminal derived peptides, Regul. Pept, vol.56, pp.71-88, 1995.

T. L. Tay, N. Hagemeyer, and M. Prinz, The force awakens: insights into the origin and formation of microglia, Curr. Opin. Neurobiol, vol.39, pp.30-37, 2016.

M. W. Teng, E. P. Bowman, J. J. Mcelwee, M. J. Smyth, J. Casanova et al., IL-12 and IL-23 cytokines: from discovery to targeted therapies for immunemediated inflammatory diseases, Nat. Med, vol.21, pp.719-729, 2015.

K. Terada, J. Yamada, Y. Hayashi, Z. Wu, Y. Uchiyama et al.,

P. Thériault, A. Elali, S. Rivest, O. Le-thuc, N. Blondeau et al., The complex contribution of chemokines to neuroinflammation: Switching from beneficial to detrimental effects, Alzheimers Res. Ther, vol.7, 2015.

. Acad and . Sci, , vol.1351, pp.127-140

J. Thundyil and K. Lim, DAMPs and neurodegeneration, Ageing Res. Rev, vol.24, pp.17-28, 2015.

E. L. Tobinick, H. Gross, E. Tobinick, H. Gross, A. Weinberger et al., Rapid cognitive improvement in Alzheimer's disease following perispinal etanercept administration, MedGenMed Medscape Gen. Med, vol.5, pp.681-687, 2006.

G. Trinchieri, S. Pflanz, and R. A. Kastelein, The IL-12 family of heterodimeric cytokines: new players in the regulation of T cell responses, Immunity, vol.19, pp.641-644, 2003.

G. Tsivgoulis, S. Judd, A. J. Letter, A. V. Alexandrov, G. Howard et al., Adherence to a Mediterranean diet and risk of incident cognitive impairment, Biochem. Soc. Trans, vol.80, pp.586-592, 2013.

G. G. Turrigiano, The self-tuning neuron: synaptic scaling of excitatory synapses, Cell, vol.135, pp.422-435, 2008.

S. L. Tyas, J. Manfreda, L. A. Strain, and P. R. Montgomery, Risk factors for Alzheimer's disease: a population-based, longitudinal study in Manitoba, Canada. Int. J. Epidemiol, vol.30, pp.590-597, 2001.

H. S. Um, E. B. Kang, Y. H. Leem, I. H. Cho, C. H. Yang et al., Exercise training acts as a therapeutic strategy for reduction of the pathogenic phenotypes for Alzheimer's disease in an NSE/APPsw-transgenic model, Lancet Lond. Engl, vol.22, pp.455-460, 2001.

R. E. Vance, R. R. Isberg, and D. A. Portnoy, Patterns of pathogenesis: discrimination of pathogenic and nonpathogenic microbes by the innate immune system, Cell Host Microbe, vol.6, pp.10-21, 2009.

J. Varley, D. J. Brooks, E. , and P. , Imaging neuroinflammation in Alzheimer's disease and other dementias: Recent advances and future directions, Alzheimers Dement. J. Alzheimers Assoc, vol.11, pp.1110-1120, 2015.

A. Varrone, V. Oikonen, A. Forsberg, J. Joutsa, A. Takano et al., Positron emission tomography imaging of the 18-kDa translocator protein (TSPO) with, 2015.

, Eur. J. Nucl. Med. Mol. Imaging, vol.42, pp.438-446

N. H. Varvel, S. A. Grathwohl, K. Degenhardt, C. Resch, . Bosch et al.,

, Med. 212, jem, 20150478.

M. J. Vasek, C. Garber, D. Dorsey, D. M. Durrant, B. Bollman et al., A complement-microglial axis drives synapse loss during virus-induced memory impairment, Nature, vol.534, pp.538-543, 2016.

S. Venneti, B. J. Lopresti, and C. A. Wiley, The peripheral benzodiazepine receptor (Translocator protein 18kDa) in microglia: from pathology to imaging, Prog. Neurobiol, vol.80, pp.308-322, 2006.

G. Verdile, S. J. Fuller, and R. N. Martins, The role of type 2 diabetes in neurodegeneration, Neurobiol. Dis, vol.84, pp.22-38, 2015.

R. Verreault, D. Laurin, J. Lindsay, and G. D. Serres, Past exposure to vaccines and subsequent risk of Alzheimer's disease, CMAJ Can. Med. Assoc. J, vol.165, pp.1495-1498, 2001.

A. J. Vincent, R. Gasperini, L. Foa, and D. H. Small, Astrocytes in Alzheimer's disease: emerging roles in calcium dysregulation and synaptic plasticity, J. Alzheimers Dis. JAD, vol.22, pp.699-714, 2010.

Y. Vodovotz, M. S. Lucia, K. C. Flanders, L. Chesler, Q. W. Xie et al.,

, J. Exp. Med, vol.184, pp.1425-1433

V. Berg, J. Prokop, S. Miller, K. R. Obst, J. Kälin et al., Inhibition of IL-12/IL-23 signaling reduces Alzheimer's disease-like pathology and cognitive decline, Neuroreport, vol.18, pp.69-72, 1993.

S. R. Waldstein and L. I. Katzel, , 2006.

. Obes, , vol.30, pp.201-207, 2005.

F. R. Walker, M. Nilsson, and K. Jones, , 2013.

, Acute and chronic stress-induced disturbances of microglial plasticity, phenotype and function, Curr. Drug Targets, vol.14, pp.1262-1276

H. X. Wang, A. Wahlin, H. Basun, J. Fastbom, B. Winblad et al., Vitamin B(12) and folate in relation to the development of Alzheimer's disease, Neurology, vol.56, pp.1188-1194, 2001.

Y. Wang, T. K. Ulland, J. D. Ulrich, W. Song, J. A. Tzaferis et al., Polypathology and dementia after brain trauma: Does brain injury trigger distinct neurodegenerative diseases, or should they be classified together as traumatic encephalopathy?, Exp. Neurol. 275 Pt, vol.213, pp.381-388, 2016.

G. S. Watson, B. A. Cholerton, M. A. Reger, L. D. Baker, S. R. Plymate et al., Preserved cognition in patients with early Alzheimer disease and amnestic mild cognitive impairment during treatment with rosiglitazone: a preliminary study, Am. J. Geriatr. Psychiatry Off. J. Am. Assoc. Geriatr. Psychiatry, vol.13, pp.950-958, 2005.

S. Weggen, J. L. Eriksen, P. Das, S. A. Sagi, R. Wang et al., A subset of NSAIDs lower amyloidogenic Abeta42 independently of cyclooxygenase activity, Nature, vol.414, pp.212-216, 2001.

R. Weiler, H. Lassmann, P. Fischer, K. Jellinger, and H. Winkler, A high ratio of chromogranin A to synaptin/synaptophysin is a common feature of brains in Alzheimer and Pick disease, FEBS Lett, vol.263, pp.337-339, 1990.

H. L. Weiner and D. Frenkel, , 2006.

, Immunology and immunotherapy of Alzheimer's disease, Nat. Rev. Immunol, vol.6, pp.404-416

G. L. Wenk, C. G. Parsons, and W. Danysz, Potential role of N-methyl-D-aspartate receptors as executors of neurodegeneration resulting from diverse insults: focus on memantine, Behav. Pharmacol, vol.17, pp.411-424, 2006.

K. Westin, P. Buchhave, H. Nielsen, L. Minthon, S. Janciauskiene et al., CCL2 is associated with a faster rate of cognitive decline during early stages of Alzheimer's disease, PLoS ONE, vol.7, pp.1-6, 2012.

R. A. Whitmer, D. R. Gustafson, E. Barrett-connor, M. N. Haan, E. P. Gunderson et al., Central obesity and increased risk of dementia more than three decades later, Neurology, vol.71, pp.1057-1064, 2008.

W. C. Willett, F. Sacks, A. Trichopoulou, G. Drescher, A. Ferro-luzzi et al., Microglial scavenger receptors and their roles in the pathogenesis of Alzheimer's disease, Int. J. Alzheimers Dis, vol.489456, 1995.

, J. Clin. Nutr, vol.61, pp.1402-1406

R. S. Wilson, D. A. Evans, J. L. Bienias, C. F. Mendes-de-leon, J. A. Schneider et al., Proneness to psychological distress is associated with risk of Alzheimer's disease, Neurology, vol.61, pp.1479-1485, 2003.

R. S. Wilson, J. A. Schneider, P. A. Boyle, S. E. Arnold, Y. Tang et al., Chronic distress and incidence of mild cognitive impairment, Neurology, vol.68, pp.2085-2092, 2007.

W. T. Wong, Microglial aging in the healthy CNS: phenotypes, drivers, and rejuvenation. Front. Cell. Neurosci. 7, 22. World Health Organization, 2004.

M. A. Wozniak, S. J. Shipley, M. Combrinck, G. K. Wilcock, and R. F. Itzhaki, , 2005.

, J. Med. Virol, vol.75, pp.300-306

M. D. Wu, S. L. Montgomery, F. Rivera-escalera, J. A. Olschowka, and M. K. Banion, , 2013.

T. Wyss-coray, J. Rogers, T. Wyss-coray, C. Lin, F. Yan et al., TGF-beta1 promotes microglial amyloid-beta clearance and reduces plaque burden in transgenic mice, Nat. Med, vol.7, pp.612-618, 2001.

T. Wyss-coray, J. D. Loike, T. C. Brionne, E. Lu, R. Anankov et al., Adult mouse astrocytes degrade amyloid-beta in vitro and in situ, 2003.

. Med, , vol.9, pp.453-457

Y. Yang, R. S. Turner, and J. R. Gaut, The chaperone BiP/GRP78 binds to amyloid precursor protein and decreases Abeta40 and Abeta42 secretion, J. Biol. Chem, vol.273, pp.25552-25555, 1998.

F. Yasuno, J. Kosaka, M. Ota, M. Higuchi, H. Ito et al., , 2012.

C. , Psychiatry Res, vol.203, pp.67-74

F. Yin, R. Banerjee, B. Thomas, P. Zhou, L. Qian et al., Exaggerated inflammation, impaired host defense, and neuropathology in progranulin-deficient mice, J. Exp. Med, vol.207, pp.117-128, 2010.

Y. Yin, Y. Liu, X. Pan, R. Chen, P. Li et al., Interleukin-1? Promoter Polymorphism Enhances the Risk of Sleep Disturbance in Alzheimer's Disease, Brain. Behav. Immun, vol.11, pp.181-213, 2011.

M. Yokokura, N. Mori, S. Yagi, E. Yoshikawa, M. Kikuchi et al., In vivo changes in microglial activation and amyloid deposits in brain regions with hypometabolism in Alzheimer's disease, Eur. J. Nucl. Med. Mol. Imaging, vol.38, pp.343-351, 2011.

H. Yuan, Q. Xia, P. Ge, S. Wu, H. Zetterberg et al., Genetic polymorphism of interleukin 1? -511C/T and susceptibility to sporadic Alzheimer's disease: a meta-analysis, Mol. Biol. Rep, vol.40, pp.194-196, 2004.

J. Zhang, A. Malik, H. B. Choi, R. W. Ko, L. Dissing-olesen et al., , 2014.

, Microglial CR3 activation triggers long-term synaptic depression in the hippocampus via NADPH oxidase, Neuron, vol.82, pp.195-207

K. Zhang, H. Xu, L. Cao, K. Li, and Q. Huang, Interleukin-1? inhibits the differentiation of hippocampal neural precursor cells into serotonergic neurons, Brain Res, vol.1490, pp.193-201, 2013.

X. Zhu, L. Tan, T. Jiang, M. Tan, W. Zhang et al., Association of IL-12A and IL-12B polymorphisms with Alzheimer's disease susceptibility in a Han Chinese population, J. Neuroimmunol, vol.274, pp.180-184, 2014.

P. A. Zunszain, C. Anacker, A. Cattaneo, S. Choudhury, K. Musaelyan et al., Interleukin-1?: a new regulator of the kynurenine pathway affecting human hippocampal neurogenesis, Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol, vol.37, pp.939-949, 2012.

A. Y. Références-abramov, L. Canevari, and M. R. Duchen, Beta-amyloid peptides induce mitochondrial dysfunction and oxidative stress in astrocytes and death of neurons through activation of NADPH oxidase, J. Neurosci. Off. J. Soc. Neurosci, vol.24, pp.565-575, 2004.

P. S. Aisen, The potential of anti-inflammatory drugs for the treatment of Alzheimer's disease, Lancet Neurol, vol.1, pp.279-284, 2002.

I. Allaman, M. Gavillet, M. Belanger, T. Laroche, D. Viertl et al.,

, Amyloid-beta aggregates cause alterations of astrocytic metabolic phenotype: impact on neuronal viability, J Neurosci, vol.30, pp.3326-3338

I. Allaman, M. Belanger, and P. J. Magistretti, Astrocyte-neuron metabolic relationships: for better and for worse, Trends Neurosci, vol.34, pp.76-87, 2011.

A. C. Alonso, T. Zaidi, I. Grundke-iqbal, and K. Iqbal, Role of abnormally phosphorylated tau in the breakdown of microtubules in Alzheimer disease, Proc. Natl. Acad. Sci. U. S. A, vol.91, pp.5562-5566, 1994.

A. C. Alonso, I. Grundke-iqbal, and K. Iqbal, Alzheimer's disease hyperphosphorylated tau sequesters normal tau into tangles of filaments and disassembles microtubules, Nat. Med, vol.2, pp.783-787, 1996.

A. D. Alonso, I. Grundke-iqbal, H. S. Barra, and K. Iqbal, Abnormal phosphorylation of tau and the mechanism of Alzheimer neurofibrillary degeneration: sequestration of microtubule-associated proteins 1 and 2 and the disassembly of microtubules by the abnormal tau, Proc. Natl. Acad. Sci. U. S. A, vol.94, pp.298-303, 1997.

J. I. Alvarez, T. Katayama, and A. Prat, Glial influence on the blood brain barrier, Glia, vol.61, pp.1939-1958, 2013.

A. Alzheimer, R. A. Stelzmann, H. N. Schnitzlein, and F. R. Murtagh, An English translation of Alzheimer's 1907 paper, Clin. Anat. N. Y. N, vol.8, pp.429-431, 1995.

M. A. Anderson, J. E. Burda, Y. Ren, Y. Ao, T. M. O&apos;shea et al., Astrocyte scar formation aids central nervous system axon regeneration, Nature, vol.532, pp.195-200, 2016.

A. Andreadis, Tau splicing and the intricacies of dementia, J. Cell. Physiol, vol.227, pp.1220-1225, 2012.

W. L. Andriezen, The Neuroglia Elements in the Human Brain, Br. Med. J, vol.2, pp.227-230, 1893.

M. C. Angulo, A. S. Kozlov, S. Charpak, A. , and E. , Glutamate released from glial cells synchronizes neuronal activity in the hippocampus, J. Neurosci. Off. J. Soc. Neurosci, vol.24, pp.6920-6927, 2004.

J. Aramburu, M. B. Yaffe, C. López-rodríguez, L. C. Cantley, P. G. Hogan et al., Affinitydriven peptide selection of an NFAT inhibitor more selective than cyclosporin A, Science, vol.285, pp.2129-2133, 1999.

A. Araque, V. Parpura, R. P. Sanzgiri, and P. G. Haydon, Tripartite synapses: glia, the unacknowledged partner, Trends Neurosci, vol.22, pp.208-215, 1999.

A. Araque, G. Carmignoto, P. G. Haydon, S. H. Oliet, R. Robitaille et al., , 2014.

, Gliotransmitters travel in time and space, Neuron, vol.81, pp.728-739

J. J. Babon, L. N. Varghese, N. , and N. A. , Inhibition of IL-6 family cytokines by SOCS3, Semin Immunol, vol.26, pp.13-19, 2014.

E. Bagyinszky, V. V. Giau, K. Shim, K. Suk, S. S. An et al., Role of inflammatory molecules in the Alzheimer's disease progression and diagnosis, J. Neurol. Sci, vol.376, pp.242-254, 2017.

F. Bao, L. Wicklund, P. N. Lacor, W. L. Klein, A. Nordberg et al., Different ?amyloid oligomer assemblies in Alzheimer brains correlate with age of disease onset and impaired cholinergic activity, Neurobiol. Aging, vol.33, pp.1-13, 2012.

F. Bard, C. Cannon, R. Barbour, R. L. Burke, D. Games et al., , 2000.

S. Bardehle, M. Kruger, F. Buggenthin, J. Schwausch, J. Ninkovic et al., Live imaging of astrocyte responses to acute injury reveals selective juxtavascular proliferation, Nat Neurosci, vol.16, pp.580-586, 2013.

S. R. Barnum, Complement biosynthesis in the central nervous system, Crit. Rev. Oral Biol, 1995.

, Med. Off. Publ. Am. Assoc. Oral Biol, vol.6, pp.132-146

C. S. Von-bartheld, J. Bahney, and S. Herculano-houzel, The search for true numbers of neurons and glial cells in the human brain: A review of 150 years of cell counting, J. Comp. Neurol, vol.524, pp.3865-3895, 2016.

K. Baumgärtel and I. M. Mansuy, Neural functions of calcineurin in synaptic plasticity and memory, Learn. Mem, vol.19, pp.375-384, 2012.

M. Belanger, I. Allaman, and P. J. Magistretti, Brain energy metabolism: focus on astrocyteneuron metabolic cooperation, Cell Metab, vol.14, pp.724-738, 2011.

L. Ben-haim, K. Ceyzeriat, M. A. Carrillo-de-sauvage, F. Aubry, G. Auregan et al., The JAK/STAT3 Pathway Is a Common Inducer of Astrocyte Reactivity in Alzheimer's and Huntington's Diseases, J Neurosci, vol.35, pp.2817-2829, 2015.

Y. Bernardinelli, J. Randall, E. Janett, I. Nikonenko, S. Konig et al., Activity-dependent structural plasticity of perisynaptic astrocytic domains promotes excitatory synapse stability, Curr Biol, vol.24, pp.1679-1688, 2014.

A. R. Bialas and B. Stevens, TGF-? signaling regulates neuronal C1q expression and developmental synaptic refinement, Nat. Neurosci, vol.16, pp.1773-1782, 2013.

A. Bignami and D. Dahl, The astroglial response to stabbing. Immunofluorescence studies with antibodies to astrocyte-specific protein (GFA) in mammalian and submammalian vertebrates, Neuropathol Appl Neuro, vol.2, pp.99-110, 1976.

L. M. Billings, S. Oddo, K. N. Green, J. L. Mcgaugh, and F. M. Laferla, Intraneuronal Abeta causes the onset of early Alzheimer's disease-related cognitive deficits in transgenic mice, Neuron, vol.45, pp.675-688, 2005.

E. Bindocci, I. Savtchouk, N. Liaudet, D. Becker, G. Carriero et al., Threedimensional Ca(2+) imaging advances understanding of astrocyte biology, Science, vol.356, 2017.

I. Blasko, R. Veerhuis, M. Stampfer-kountchev, M. Saurwein-teissl, P. Eikelenboom et al., Costimulatory effects of interferon-gamma and interleukin-1beta or tumor necrosis factor alpha on the synthesis of Abeta1-40 and Abeta1-42 by human astrocytes, Neurobiol. Dis, vol.7, pp.682-689, 2000.

L. Blazquez-llorca, S. Valero-freitag, E. F. Rodrigues, Á. Merchán-pérez, J. R. Rodríguez et al., High plasticity of axonal pathology in Alzheimer's disease mouse models, Acta Neuropathol. Commun, vol.5, p.14, 2017.

K. Blennow, M. J. De-leon, and H. Zetterberg, Alzheimer's disease, Lancet Lond. Engl, vol.368, pp.387-403, 2006.
URL : https://hal.archives-ouvertes.fr/hal-01098836

O. Bloom, Non-mammalian model systems for studying neuro-immune interactions after spinal cord injury, Exp Neurol, vol.258, pp.130-140, 2014.

D. S. Bouvier, E. V. Jones, G. Quesseveur, M. A. Davoli, T. et al., High Resolution Dissection of Reactive Glial Nets in Alzheimer's, Disease. Sci Rep, vol.6, p.24544, 2016.

G. L. Bowman, J. A. Kaye, M. Moore, D. Waichunas, N. E. Carlson et al., Blood-brain barrier impairment in Alzheimer disease: stability and functional significance, Neurology, vol.68, pp.1809-1814, 2007.

H. Braak and E. Braak, Neuropathological stageing of Alzheimer-related changes, Acta Neuropathol. (Berl.), vol.82, pp.239-259, 1991.

A. J. Brookes and D. St-clair, Synuclein proteins and Alzheimer's disease, Trends Neurosci, vol.17, pp.404-405, 1994.

A. J. Brooks, W. Dai, M. L. O&apos;mara, D. Abankwa, Y. Chhabra et al., Mechanism of activation of protein kinase JAK2 by the growth hormone receptor, Science, vol.344, p.1249783, 2014.

G. Bu, Apolipoprotein E and its receptors in Alzheimer's disease: pathways, pathogenesis and therapy, Nat. Rev. Neurosci, vol.10, pp.333-344, 2009.

J. E. Burda and M. V. Sofroniew, Reactive gliosis and the multicellular response to CNS damage and disease, Neuron, vol.81, pp.229-248, 2014.

J. E. Burda, A. M. Bernstein, and M. V. Sofroniew, Astrocyte roles in traumatic brain injury, Exp Neurol, vol.275, pp.305-315, 2016.

E. A. Bushong, M. E. Martone, Y. Z. Jones, and M. H. Ellisman, Protoplasmic astrocytes in CA1 stratum radiatum occupy separate anatomical domains, J Neurosci, vol.22, pp.183-192, 2002.

S. F. Carter, M. Scholl, O. Almkvist, A. Wall, H. Engler et al., Evidence for astrocytosis in prodromal Alzheimer disease provided by 11C-deuterium-L-deprenyl: a multitracer PET paradigm combining 11C-Pittsburgh compound B and 18F-FDG, J Nucl Med, vol.53, pp.37-46, 2012.

J. M. Castellano, R. Deane, A. J. Gottesdiener, P. B. Verghese, F. R. Stewart et al., Low-density lipoprotein receptor overexpression enhances the rate of brain-to, 2012.

, Proc. Natl. Acad. Sci. U. S. A, vol.109, pp.15502-15507

K. Ceyzeriat, L. Abjean, M. A. Carrillo-de-sauvage, L. Ben-haim, and C. Escartin, The complex STATes of astrocyte reactivity: How are they controlled by the JAK-STAT3 pathway?, Neuroscience, vol.330, pp.205-218, 2016.
URL : https://hal.archives-ouvertes.fr/cea-02142559

P. Chakrabarty, K. Jansen-west, A. Beccard, C. Ceballos-diaz, Y. Levites et al., Massive gliosis induced by interleukin-6 suppresses Abeta deposition in vivo: evidence against inflammation as a driving force for amyloid deposition, FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol, vol.24, pp.548-559, 2010.

P. Chakrabarty, C. Ceballos-diaz, A. Beccard, C. Janus, D. Dickson et al., , 2010.

, IFN-gamma promotes complement expression and attenuates amyloid plaque deposition in amyloid beta precursor protein transgenic mice, J. Immunol. Baltim. Md, vol.184, pp.5333-5343, 1950.

P. Chakrabarty, A. Herring, C. Ceballos-diaz, P. Das, and T. E. Golde, Hippocampal expression of murine TNF? results in attenuation of amyloid deposition in vivo, Mol. Neurodegener, vol.6, p.16, 2011.

P. Chakrabarty, A. Li, C. Ceballos-diaz, J. A. Eddy, C. C. Funk et al., IL-10 alters immunoproteostasis in APP mice, increasing plaque burden and worsening cognitive behavior, Neuron, vol.85, pp.519-533, 2015.

M. Cho, K. Cho, H. Kang, E. Jeon, H. Kim et al., Autophagy in microglia degrades extracellular ?-amyloid fibrils and regulates the NLRP3 inflammasome, Autophagy, vol.10, pp.1761-1775, 2014.

K. S. Christopherson, E. M. Ullian, C. C. Stokes, C. E. Mullowney, J. W. Hell et al., Thrombospondins are astrocyte-secreted proteins that promote CNS synaptogenesis, Cell, vol.120, pp.421-433, 2005.

W. S. Chung, L. E. Clarke, G. X. Wang, B. K. Stafford, A. Sher et al., Astrocytes mediate synapse elimination through MEGF10 and MERTK pathways, Nature, vol.504, pp.394-400, 2013.

W. Chung, N. J. Allen, and C. Eroglu, Astrocytes Control Synapse Formation, Function, and Elimination, Cold Spring Harb. Perspect. Biol, vol.7, 2015.

J. K. Clark, M. Furgerson, J. D. Crystal, M. Fechheimer, R. Furukawa et al., , 2015.

, Alterations in synaptic plasticity coincide with deficits in spatial working memory in presymptomatic 3xTg-AD mice, Neurobiol. Learn. Mem, vol.125, pp.152-162

L. K. Clinton, L. M. Billings, K. N. Green, A. Caccamo, J. Ngo et al., Age-dependent sexual dimorphism in cognition and stress response in the 3xTg-AD mice, Neurobiol. Dis, vol.28, pp.76-82, 2007.

C. K. Combs, J. C. Karlo, S. C. Kao, and G. E. Landreth, beta-Amyloid stimulation of microglia and monocytes results in TNFalpha-dependent expression of inducible nitric oxide synthase and neuronal apoptosis, J. Neurosci. Off. J. Soc. Neurosci, vol.21, pp.1179-1188, 2001.

E. H. Corder, A. M. Saunders, W. J. Strittmatter, D. E. Schmechel, P. C. Gaskell et al., , 1993.

A. Covelo, A. , and A. , Lateral regulation of synaptic transmission by astrocytes, Neuroscience, vol.323, pp.62-66, 2016.

A. Crowe, M. J. James, V. M. Lee, .. Smith, A. B. Trojanowski et al.,

, Aminothienopyridazines and methylene blue affect Tau fibrillization via cysteine oxidation, J. Biol. Chem, vol.288, pp.11024-11037

D. V. Dabir, M. B. Robinson, E. Swanson, B. Zhang, J. Q. Trojanowski et al., Impaired glutamate transport in a mouse model of tau pathology in astrocytes, J Neurosci, vol.26, pp.644-654, 2006.

R. Daneman and A. Prat, The Blood-Brain Barrier, Cold Spring Harb. Perspect. Biol, vol.7, p.20412, 2015.

J. W. Dani, A. Chernjavsky, and S. J. Smith, Neuronal activity triggers calcium waves in hippocampal astrocyte networks, Neuron, vol.8, pp.429-440, 1992.

C. Dansokho, D. Ahmed, S. Aid, C. Toly-ndour, T. Chaigneau et al., Regulatory T cells delay disease progression in Alzheimer-like pathology, Brain J. Neurol, vol.139, pp.1237-1251, 2016.

J. E. Darnell and . Jr, STATs and gene regulation, Science, vol.277, pp.1630-1635, 1997.

B. De-strooper and E. Karran, The Cellular Phase of Alzheimer's Disease, Cell, vol.164, pp.603-615, 2016.

W. Deng, J. B. Aimone, and F. H. Gage, New neurons and new memories: how does adult hippocampal neurogenesis affect learning and memory?, Nat. Rev. Neurosci, vol.11, pp.339-350, 2010.

C. A. Derby, L. C. Burns, C. Wang, M. J. Katz, M. E. Zimmerman et al., Screening for predementia AD: time-dependent operating characteristics of episodic memory tests, Neurology, vol.80, pp.1307-1314, 2013.

T. R. Desai, N. J. Leeper, K. L. Hynes, and B. L. Gewertz, , 2002.

A. Dobin, C. A. Davis, F. Schlesinger, J. Drenkow, C. Zaleski et al., STAR: ultrafast universal RNA-seq aligner, Bioinforma. Oxf. Engl, vol.29, pp.15-21, 2013.

J. Doherty, A. E. Sheehan, R. Bradshaw, A. N. Fox, T. Y. Lu et al., PI3K signaling and Stat92E converge to modulate glial responsiveness to axonal injury, PLoS Biol, vol.12, 2014.

B. Dubois, H. H. Feldman, C. Jacova, H. Hampel, J. L. Molinuevo et al., Advancing research diagnostic criteria for Alzheimer's disease: the IWG-2 criteria, Lancet Neurol, vol.13, pp.614-629, 2014.

C. Duyckaerts, M. C. Potier, and B. Delatour, Alzheimer disease models and human neuropathology: similarities and differences, Acta Neuropathol, vol.115, pp.5-38, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00320572

E. A. Eckman, D. K. Reed, and C. B. Eckman, Degradation of the Alzheimer's amyloid beta peptide by endothelin-converting enzyme, J. Biol. Chem, vol.276, pp.24540-24548, 2001.

Y. S. Eisele and C. Duyckaerts, Propagation of Aß pathology: hypotheses, discoveries, and yet unresolved questions from experimental and human brain studies, Acta Neuropathol. (Berl.), vol.131, pp.5-25, 2016.

M. R. Emmerling, M. D. Watson, C. A. Raby, and K. Spiegel, The role of complement in Alzheimer's disease pathology, Biochim. Biophys. Acta, vol.1502, pp.158-171, 2000.

L. F. Eng, J. J. Vanderhaeghen, A. Bignami, and B. Gerstl, An acidic protein isolated from fibrous astrocytes, Brain Res, vol.28, pp.351-354, 1971.

J. L. Eriksen, S. A. Sagi, T. E. Smith, S. Weggen, P. Das et al., NSAIDs and enantiomers of flurbiprofen target gamma-secretase and lower Abeta 42 in vivo, J. Clin. Invest, vol.112, pp.440-449, 2003.

C. Escartin, K. Pierre, A. Colin, E. Brouillet, T. Delzescaux et al., Activation of astrocytes by CNTF induces metabolic plasticity and increases resistance to metabolic insults, J Neurosci, vol.27, pp.7094-7104, 2007.
URL : https://hal.archives-ouvertes.fr/cea-02290623

R. Etcheberrigaray, M. Tan, I. Dewachter, C. Kuipéri, I. Van-der-auwera et al., Therapeutic effects of PKC activators in Alzheimer's disease transgenic mice, Proc. Natl. Acad. Sci. U. S. A, vol.101, pp.11141-11146, 2004.

S. Fedorff, Astrocytes Pt 1: Development, Morphology, and Regional Specialization of, Astrocytes, 2012.

T. Fellin, O. Pascual, S. Gobbo, T. Pozzan, P. G. Haydon et al., Neuronal synchrony mediated by astrocytic glutamate through activation of extrasynaptic NMDA receptors, Neuron, vol.43, pp.729-743, 2004.

A. M. Fernandez, S. Jimenez, M. Mecha, D. Davila, C. Guaza et al.,

T. P. Flaten, Aluminium as a risk factor in Alzheimer's disease, with emphasis on drinking water, Brain Res. Bull, vol.55, pp.187-196, 2001.

M. I. Fonseca, J. Zhou, M. Botto, and A. J. Tenner, Absence of C1q leads to less neuropathology in transgenic mouse models of Alzheimer's disease, J. Neurosci. Off. J. Soc. Neurosci, vol.24, pp.6457-6465, 2004.

M. R. Freeman, Drosophila Central Nervous System Glia, Cold Spring Harb. Perspect. Biol, vol.7, 2015.

H. Fukuyama, M. Ogawa, H. Yamauchi, S. Yamaguchi, J. Kimura et al.,

, Altered cerebral energy metabolism in Alzheimer's disease: a PET study, J Nucl Med, vol.35, pp.1-6

J. L. Furman, D. M. Sama, J. C. Gant, T. L. Beckett, M. P. Murphy et al., Targeting astrocytes ameliorates neurologic changes in a mouse model of Alzheimer's disease, J Neurosci, vol.32, pp.16129-16140, 2012.

D. Galimberti, C. Fenoglio, C. Lovati, E. Venturelli, I. Guidi et al., Serum MCP-1 levels are increased in mild cognitive impairment and mild Alzheimer's disease, Neurobiol. Aging, vol.27, pp.1763-1768, 2006.

S. Gao, H. C. Hendrie, K. S. Hall, and S. Hui, The relationships between age, sex, and the incidence of dementia and Alzheimer disease: a meta-analysis, Arch. Gen. Psychiatry, vol.55, pp.809-815, 1998.

M. Garcia-alloza, E. M. Robbins, S. X. Zhang-nunes, S. M. Purcell, R. A. Betensky et al., Characterization of amyloid deposition in the APPswe/PS1dE9 mouse model of Alzheimer disease, Neurobiol Dis, vol.24, pp.516-524, 2006.

V. Garcia-marin, P. Garcia-lopez, and M. Freire, Cajal's contributions to glia research, Trends Neurosci, vol.30, pp.479-487, 2007.

S. Gauthier, H. Loft, and J. Cummings, Improvement in behavioural symptoms in patients with moderate to severe Alzheimer's disease by memantine: a pooled data analysis, Int. J. Geriatr. Psychiatry, vol.23, pp.537-545, 2008.

S. Ghosh, M. D. Wu, S. S. Shaftel, S. Kyrkanides, F. M. Laferla et al., Sustained interleukin-1? overexpression exacerbates tau pathology despite reduced amyloid burden in an Alzheimer's mouse model, J. Neurosci. Off. J. Soc. Neurosci, vol.33, pp.5053-5064, 2013.

S. Gilman, M. Koller, R. S. Black, L. Jenkins, S. G. Griffith et al., Clinical effects of Abeta immunization (AN1792) in patients with AD in an interrupted trial, Neurology, vol.64, pp.1553-1562, 2005.

F. Ginhoux, M. Greter, M. Leboeuf, S. Nandi, P. See et al., Fate mapping analysis reveals that adult microglia derive from primitive macrophages, Science, vol.330, pp.841-845, 2010.

A. Goate, M. C. Chartier-harlin, M. Mullan, J. Brown, F. Crawford et al., Segregation of a missense mutation in the amyloid precursor protein gene with familial Alzheimer's disease, Nature, vol.349, pp.704-706, 1991.

M. Goedert, Alzheimer's and Parkinson's diseases: The prion concept in relation to assembled A?, tau, and ?-synuclein, Science, vol.349, p.1255555, 2015.

J. L. Griffin, M. Bollard, J. K. Nicholson, and K. Bhakoo, Spectral profiles of cultured neuronal and glial cells derived from HRMAS (1)H NMR spectroscopy, NMR Biomed, vol.15, pp.375-384, 2002.

T. Van-groen, A. J. Kiliaan, and I. Kadish, Deposition of mouse amyloid beta in human APP/PS1 double and single AD model transgenic mice, Neurobiol. Dis, vol.23, pp.653-662, 2006.

R. Guerreiro, A. Wojtas, J. Bras, M. Carrasquillo, E. Rogaeva et al., TREM2 variants in Alzheimer's disease, N Engl J Med, vol.368, pp.117-127, 2013.

M. V. Guillot-sestier, K. R. Doty, D. Gate, J. Rodriguez, . Jr et al.,

, Il10 deficiency rebalances innate immunity to mitigate Alzheimer-like pathology, Neuron, vol.85, pp.534-548

S. Haan, S. Wüller, J. Kaczor, C. Rolvering, T. Nöcker et al., SOCSmediated downregulation of mutant Jak2 (V617F, T875N and K539L) counteracts cytokineindependent signaling, Oncogene, vol.28, pp.3069-3080, 2009.

M. M. Halassa, T. Fellin, H. Takano, J. H. Dong, and P. G. Haydon, Synaptic islands defined by the territory of a single astrocyte, J Neurosci, vol.27, pp.6473-6477, 2007.

A. Halle, V. Hornung, G. C. Petzold, C. R. Stewart, B. G. Monks et al., The NALP3 inflammasome is involved in the innate immune response to amyloid-beta, Nat Immunol, vol.9, pp.857-865, 2008.

J. A. Hardy and G. A. Higgins, Alzheimer's disease: the amyloid cascade hypothesis, Science, vol.256, pp.184-185, 1992.

P. C. Heinrich, I. Behrmann, S. Haan, H. M. Hermanns, G. Muller-newen et al., Principles of interleukin (IL)-6-type cytokine signalling and its regulation, Biochem J, vol.374, pp.1-20, 2003.

M. T. Heneka, M. Sastre, L. Dumitrescu-ozimek, I. Dewachter, J. Walter et al., Focal glial activation coincides with increased BACE1 activation and precedes amyloid plaque deposition in APP[V717I] transgenic mice, J. Neuroinflammation, vol.2, 2005.

M. T. Heneka, M. P. Kummer, and E. Latz, Innate immune activation in neurodegenerative disease, Nat Rev Immunol, vol.14, pp.463-477, 2014.

M. T. Heneka, M. J. Carson, J. Khoury, G. E. Landreth, F. Brosseron et al., Neuroinflammation in Alzheimer's disease, vol.14, pp.388-405, 2015.

S. E. Hickman and J. Khoury, TREM2 and the neuroimmunology of Alzheimer's disease, 2014.

, Biochem Pharmacol, vol.88, pp.495-498

S. E. Hickman, E. K. Allison, and J. Khoury, Microglial dysfunction and defective betaamyloid clearance pathways in aging Alzheimer's disease mice, J Neurosci, vol.28, pp.8354-8360, 2008.

K. Hoglund, K. M. Thelen, S. Syversen, M. Sjogren, K. Von-bergmann et al., The effect of simvastatin treatment on the amyloid precursor protein and brain cholesterol metabolism, 2005.

, Dement. Geriatr. Cogn. Disord, vol.19, pp.256-265

E. M. Hol and M. Pekny, Glial fibrillary acidic protein (GFAP) and the astrocyte intermediate filament system in diseases of the central nervous system, Curr Opin Cell Biol, vol.32, pp.121-130, 2015.

L. Hong, H. Huang, and Z. Jiang, Relationship between amyloid-beta and the ubiquitinproteasome system in Alzheimer's disease, Neurol. Res, vol.36, pp.276-282, 2014.

S. Hong, V. F. Beja-glasser, B. M. Nfonoyim, A. Frouin, S. Li et al., Complement and microglia mediate early synapse loss in Alzheimer mouse models, Science, vol.352, pp.712-716, 2016.

D. W. Huang, B. T. Sherman, and R. A. Lempicki, Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources, Nat. Protoc, vol.4, pp.44-57, 2009.

Y. A. Huang, B. Zhou, M. Wernig, and T. C. Südhof, ApoE2, ApoE3, and ApoE4 Differentially Stimulate APP Transcription and A? Secretion, vol.168, pp.427-441, 2017.

A. P. Hutchins, D. Diez, Y. Takahashi, S. Ahmad, R. Jauch et al., Distinct transcriptional regulatory modules underlie STAT3's cell type-independent and cell type-specific functions, Nucleic Acids Res, vol.41, pp.2155-2170, 2013.

M. D. Ikonomovic, W. E. Klunk, E. E. Abrahamson, C. A. Mathis, J. C. Price et al., Post-mortem correlates of in vivo PiB-PET amyloid imaging in a typical case of Alzheimer's disease, Brain J. Neurol, vol.131, pp.1630-1645, 2008.

T. Iram, D. Trudler, D. Kain, S. Kanner, R. Galron et al., Astrocytes from old Alzheimer's disease mice are impaired in A? uptake and in neuroprotection, Neurobiol. Dis, vol.96, pp.84-94, 2016.

C. R. Jack, F. Barkhof, M. A. Bernstein, M. Cantillon, P. E. Cole et al., Steps to standardization and validation of hippocampal volumetry as a biomarker in clinical trials and diagnostic criterion for Alzheimer's disease, Alzheimers Dement. J. Alzheimers Assoc, vol.7, pp.474-485, 2011.

J. L. Jankowsky, D. J. Fadale, J. Anderson, G. M. Xu, V. Gonzales et al., Mutant presenilins specifically elevate the levels of the 42 residue beta-amyloid peptide in vivo: evidence for augmentation of a 42-specific gamma secretase, Hum Mol Genet, vol.13, pp.159-170, 2004.

A. H. Jansen, E. A. Reits, and E. M. Hol, The ubiquitin proteasome system in glia and its role in neurodegenerative diseases, Front Mol Neurosci, vol.7, p.73, 2014.

C. Janus, A. Y. Flores, G. Xu, and D. R. Borchelt, Behavioral abnormalities in APPSwe/PS1dE9 mouse model of AD-like pathology: comparative analysis across multiple behavioral domains, Neurobiol. Aging, vol.36, pp.2519-2532, 2015.

Z. Jaunmuktane, S. Mead, M. Ellis, J. D. Wadsworth, A. J. Nicoll et al., Evidence for human transmission of amyloid-? pathology and cerebral amyloid angiopathy, Nature, vol.525, pp.247-250, 2015.

T. R. Jay, C. M. Miller, P. J. Cheng, L. C. Graham, S. Bemiller et al., TREM2 deficiency eliminates TREM2+ inflammatory macrophages and ameliorates pathology in Alzheimer's disease mouse models, J. Exp. Med, vol.212, pp.287-295, 2015.

S. Jo, O. Yarishkin, Y. J. Hwang, Y. E. Chun, M. Park et al., GABA from reactive astrocytes impairs memory in mouse models of Alzheimer's disease, 2014.

, Nat Med, vol.20, pp.886-896

J. Lin, C. C. Yu, K. Hatcher, A. Huang, T. W. Lee et al., Identification of diverse astrocyte populations and their malignant analogs, Nat Neurosci, 2017.

T. Jonsson, H. Stefansson, S. Steinberg, I. Jonsdottir, P. V. Jonsson et al., Variant of TREM2 associated with the risk of Alzheimer's disease, N Engl J Med, vol.368, pp.107-116, 2013.

A. F. Jorm, History of depression as a risk factor for dementia: an updated review, Aust. N. Z. J. Psychiatry, vol.35, pp.776-781, 2001.

W. Kamphuis, L. Kooijman, M. Orre, O. Stassen, M. Pekny et al., GFAP and vimentin deficiency alters gene expression in astrocytes and microglia in wild-type mice and changes the transcriptional response of reactive glia in mouse model for Alzheimer's disease, Glia, vol.63, pp.1036-1056, 2015.

B. Kantor, R. M. Bailey, K. Wimberly, S. N. Kalburgi, and S. J. Gray, Methods for Gene Transfer to the Central Nervous System, Adv. Genet, vol.87, pp.125-197, 2014.

H. Keren-shaul, A. Spinrad, A. Weiner, O. Matcovitch-natan, R. Dvir-szternfeld et al., A Unique Microglia Type Associated with Restricting Development of Alzheimer's Disease, Cell, vol.169, pp.1276-1290, 2017.

N. J. Kershaw, J. M. Murphy, N. P. Liau, L. N. Varghese, A. Laktyushin et al., SOCS3 binds specific receptor-JAK complexes to control cytokine signaling by direct kinase inhibition, Nat Struct Mol Biol, vol.20, pp.469-476, 2013.

M. Kilgore, C. A. Miller, D. M. Fass, K. M. Hennig, S. J. Haggarty et al.,

, Alzheimer's disease, Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol, vol.35, pp.870-880

T. Kiyota, S. Okuyama, R. J. Swan, M. T. Jacobsen, H. E. Gendelman et al., CNS expression of anti-inflammatory cytokine interleukin-4 attenuates Alzheimer's disease-like pathogenesis in APP+PS1 bigenic mice, FASEB J, vol.24, pp.3093-3102, 2010.

T. Kiyota, K. L. Ingraham, R. J. Swan, M. T. Jacobsen, S. J. Andrews et al., AAV serotype 2/1-mediated gene delivery of anti-inflammatory interleukin-10 enhances neurogenesis and cognitive function in APP+PS1 mice, Gene Ther, vol.19, pp.724-733, 2012.

G. Kleinberger, Y. Yamanishi, M. Suarez-calvet, E. Czirr, E. Lohmann et al., TREM2 mutations implicated in neurodegeneration impair cell surface transport and phagocytosis, Sci Transl Med, vol.6, pp.243-86, 2014.

W. E. Klunk, H. Engler, A. Nordberg, Y. Wang, G. Blomqvist et al., Imaging brain amyloid in Alzheimer's disease with Pittsburgh Compound-B, Ann. Neurol, vol.55, pp.306-319, 2004.

M. Koistinaho, S. Lin, X. Wu, M. Esterman, D. Koger et al., Apolipoprotein E promotes astrocyte colocalization and degradation of deposited amyloid-beta peptides, Nat Med, vol.10, pp.719-726, 2004.

E. Kontsekova, N. Zilka, B. Kovacech, P. Novak, and M. Novak, First-in-man tau vaccine targeting structural determinants essential for pathological tau-tau interaction reduces tau oligomerisation and neurofibrillary degeneration in an Alzheimer's disease model, Alzheimers Res. Ther, vol.6, p.44, 2014.

N. A. Kootstra and I. M. Verma, Gene therapy with viral vectors, Annu. Rev. Pharmacol. Toxicol, vol.43, pp.413-439, 2003.

A. W. Kraft, X. Hu, H. Yoon, P. Yan, Q. Xiao et al., , 2013.

, FASEB J, vol.27, pp.187-198

M. Krauthausen, M. P. Kummer, J. Zimmermann, E. Reyes-irisarri, D. Terwel et al., CXCR3 promotes plaque formation and behavioral deficits in an Alzheimer's disease model, J. Clin. Invest, vol.125, pp.365-378, 2015.

C. Laurent, G. Dorothée, S. Hunot, E. Martin, Y. Monnet et al., Hippocampal T cell infiltration promotes neuroinflammation and cognitive decline in a mouse model of tauopathy, Brain J. Neurol, vol.140, pp.184-200, 2017.
URL : https://hal.archives-ouvertes.fr/inserm-01833350

C. Y. Lee and G. E. Landreth, The role of microglia in amyloid clearance from the AD brain, J. Neural Transm, vol.117, pp.949-960, 1996.

Y. Lee, A. Messing, M. Su, and M. Brenner, GFAP promoter elements required for regionspecific and astrocyte-specific expression, Glia, vol.56, pp.481-493, 2008.

D. E. Levy, J. E. Darnell, and . Jr, Stats: transcriptional control and biological impact, Nat Rev Mol Cell Biol, vol.3, pp.651-662, 2002.

E. Levy-lahad, W. Wasco, P. Poorkaj, D. M. Romano, J. Oshima et al., Candidate gene for the chromosome 1 familial Alzheimer's disease locus, Science, vol.269, pp.973-977, 1995.

B. Li, M. O. Chohan, I. Grundke-iqbal, and K. Iqbal, Disruption of microtubule network by Alzheimer abnormally hyperphosphorylated tau, Acta Neuropathol. (Berl.), vol.113, pp.501-511, 2007.

H. Li, A. Rao, and P. G. Hogan, Interaction of calcineurin with substrates and targeting proteins, Trends Cell Biol, vol.21, pp.91-103, 2011.

Y. Li, X. F. Du, C. S. Liu, Z. L. Wen, and J. L. Du, Reciprocal regulation between resting microglial dynamics and neuronal activity in vivo, Dev Cell, vol.23, pp.1189-1202, 2012.

H. Lian, L. Yang, A. Cole, L. Sun, A. C. Chiang et al., NF?B-activated astroglial release of complement C3 compromises neuronal morphology and function associated with Alzheimer's disease, Neuron, vol.85, pp.101-115, 2015.

H. Lian, A. Litvinchuk, A. C. Chiang, .. Aithmitti, N. Jankowsky et al., Astrocyte-Microglia Cross Talk through Complement Activation Modulates Amyloid Pathology in Mouse Models of Alzheimer's Disease, J. Neurosci. Off. J. Soc. Neurosci, vol.36, pp.577-589, 2016.

S. A. Liddelow, K. A. Guttenplan, L. E. Clarke, F. C. Bennett, C. J. Bohlen et al., Neurotoxic reactive astrocytes are induced by activated microglia, Nature, vol.541, pp.481-487, 2017.

C. P. Lim and X. Cao, Structure, function, and regulation of STAT proteins, Mol Biosyst, vol.2, pp.536-550, 2006.

M. T. Lin and M. F. Beal, Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases, Nature, vol.443, pp.787-795, 2006.

K. Lindsten, F. M. De-vrij, L. G. Verhoef, D. F. Fischer, F. W. Van-leeuwen et al., Mutant ubiquitin found in neurodegenerative disorders is a ubiquitin fusion degradation substrate that blocks proteasomal degradation, J. Cell Biol, vol.157, pp.417-427, 2002.

L. Liu, V. Drouet, J. W. Wu, M. P. Witter, S. A. Small et al., Trans-synaptic spread of tau pathology in vivo, PloS One, vol.7, p.31302, 2012.

Q. Y. Liu, A. E. Schaffner, Y. H. Chang, D. Maric, and J. L. Barker, Persistent activation of GABA(A) receptor/Cl(-) channels by astrocyte-derived GABA in cultured embryonic rat hippocampal neurons, J. Neurophysiol, vol.84, pp.1392-1403, 2000.

Y. Liu, S. Walter, M. Stagi, D. Cherny, M. Letiembre et al., LPS receptor (CD14): a receptor for phagocytosis of Alzheimer's amyloid peptide, Brain, vol.128, pp.1778-1789, 2005.

S. Lively and L. C. Schlichter, The microglial activation state regulates migration and roles of matrix-dissolving enzymes for invasion, J. Neuroinflammation, vol.10, p.75, 2013.

I. López-gonzález, P. Garcia-esparcia, F. Llorens, and I. Ferrer, Genetic and Transcriptomic Profiles of Inflammation in Neurodegenerative Diseases: Alzheimer, Parkinson, Creutzfeldt-Jakob and Tauopathies, Int. J. Mol. Sci, vol.17, p.206, 2016.

M. I. Love, W. Huber, A. , and S. , Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2, Genome Biol, vol.15, p.550, 2014.

S. Lovestone, M. Boada, B. Dubois, M. Hüll, J. O. Rinne et al., A phase II trial of tideglusib in Alzheimer's disease, J. Alzheimers Dis. JAD, vol.45, pp.75-88, 2015.

K. M. Lucin, C. E. O&apos;brien, G. Bieri, E. Czirr, K. I. Mosher et al., Microglial beclin 1 regulates retromer trafficking and phagocytosis and is impaired in Alzheimer's disease, Neuron, vol.79, pp.873-886, 2013.

J. Luna-muñoz, C. R. Harrington, C. M. Wischik, P. Flores-rodríguez, J. Avila et al., Phosphorylation of Tau Protein Associated as a Protective Mechanism in the Presence of Toxic, C-Terminally, 2013.

I. Lundgaard, B. Li, L. Xie, H. Kang, S. Sanggaard et al., Direct neuronal glucose uptake heralds activity-dependent increases in cerebral metabolism, Nat. Commun, vol.6, p.6807, 2015.

Y. Luo, B. Bolon, S. Kahn, B. D. Bennett, S. Babu-khan et al., Mice deficient in BACE1, the Alzheimer's beta-secretase, have normal phenotype and abolished beta-amyloid generation, Nat. Neurosci, vol.4, pp.231-232, 2001.

J. Ma, Y. Huang, S. Chen, and G. Halliday, Immunohistochemical evidence for macroautophagy in neurones and endothelial cells in Alzheimer's disease, Neuropathol. Appl. Neurobiol, vol.36, pp.312-319, 2010.

T. Ma, X. Du, J. E. Pick, G. Sui, M. Brownlee et al., Glucagon-like peptide-1 cleavage product GLP-1(9-36) amide rescues synaptic plasticity and memory deficits in Alzheimer's disease model mice, J. Neurosci. Off. J. Soc. Neurosci, vol.32, pp.13701-13708, 2012.

P. J. Magistretti, Neuron-glia metabolic coupling and plasticity, J Exp Biol, vol.209, pp.2304-2311, 2006.

T. K. Makar, M. Nedergaard, A. Preuss, A. S. Gelbard, A. S. Perumal et al., Vitamin E, ascorbate, glutathione, glutathione disulfide, and enzymes of glutathione metabolism in cultures of chick astrocytes and neurons: evidence that astrocytes play an important role in antioxidative processes in the brain, J Neurochem, vol.62, pp.45-53, 1994.

R. C. Malenka and M. F. Bear, LTP and LTD: an embarrassment of riches, Neuron, vol.44, pp.5-21, 2004.

T. Malm, J. Koistinaho, and K. Kanninen, Utilization of APPswe/PS1dE9 Transgenic Mice in Research of Alzheimer's Disease: Focus on Gene Therapy and Cell-Based Therapy Applications, Int. J. Alzheimers Dis, p.517160, 2011.

N. J. Maragakis and J. D. Rothstein, Glutamate transporters: animal models to neurologic disease, Neurobiol. Dis, vol.15, pp.461-473, 2004.

E. Martin, C. Boucher, B. Fontaine, and C. Delarasse, Distinct inflammatory phenotypes of microglia and monocyte-derived macrophages in Alzheimer's disease models: effects of aging and amyloid pathology, Aging Cell, vol.16, pp.27-38, 2017.

E. Masliah, M. Alford, R. Deteresa, M. Mallory, and L. Hansen, Deficient glutamate transport is associated with neurodegeneration in Alzheimer's disease, Ann Neurol, vol.40, pp.759-766, 1996.

E. Masliah, M. Alford, M. Mallory, E. Rockenstein, D. Moechars et al., , 2000.

C. L. Masters, R. Bateman, K. Blennow, C. C. Rowe, R. A. Sperling et al., , 2015.

, Alzheimer's disease, Nat. Rev. Dis. Primer, vol.1, p.15056

H. Mathys, C. Adaikkan, F. Gao, J. Z. Young, E. Manet et al., Temporal Tracking of Microglia Activation in Neurodegeneration at Single-Cell Resolution, Cell Rep, vol.21, pp.366-380, 2017.

P. L. Mcgeer, S. Itagaki, H. Tago, and E. G. Mcgeer, Occurrence of HLA-DR reactive microglia in Alzheimer's disease, Ann N Acad Sci, vol.540, pp.319-323, 1988.

D. X. Medina, A. Caccamo, and S. Oddo, Methylene blue reduces a? levels and rescues early cognitive deficit by increasing proteasome activity, Brain Pathol. Zurich Switz, vol.21, pp.140-149, 2011.

M. Merlini, E. P. Meyer, A. Ulmann-schuler, and R. M. Nitsch, Vascular ?-amyloid and early astrocyte alterations impair cerebrovascular function and cerebral metabolism in transgenic arcA? mice, Acta Neuropathol. (Berl.), vol.122, pp.293-311, 2011.

C. Métais, K. Brennan, A. J. Mably, M. Scott, D. M. Walsh et al., Simvastatin treatment preserves synaptic plasticity in A?PPswe/PS1dE9 mice, J. Alzheimers Dis. JAD, vol.39, pp.315-329, 2014.

J. Michaud, M. Bellavance, P. Préfontaine, and S. Rivest, Real-time in vivo imaging reveals the ability of monocytes to clear vascular amyloid beta, Cell Rep, vol.5, pp.646-653, 2013.

A. Mildner, B. Schlevogt, K. Kierdorf, C. Böttcher, D. Erny et al., Distinct and non-redundant roles of microglia and myeloid subsets in mouse models of Alzheimer's disease, J. Neurosci. Off. J. Soc. Neurosci, vol.31, pp.11159-11171, 2011.

R. H. Miller and M. C. Raff, Fibrous and protoplasmic astrocytes are biochemically and developmentally distinct, J. Neurosci. Off. J. Soc. Neurosci, vol.4, pp.585-592, 1984.

J. S. Miners, S. Baig, H. Tayler, P. G. Kehoe, and S. Love, Neprilysin and insulin-degrading enzyme levels are increased in Alzheimer disease in relation to disease severity, J. Neuropathol. Exp. Neurol, vol.68, pp.902-914, 2009.

J. Minkiewicz, . De-rivero, J. P. Vaccari, and R. W. Keane, Human astrocytes express a novel NLRP2 inflammasome, Glia, vol.61, pp.1113-1121, 2013.

M. A. Mintun, G. N. Larossa, Y. I. Sheline, C. S. Dence, S. Y. Lee et al., ). [11C]PIB in a nondemented population: potential antecedent marker of Alzheimer disease, Neurology, vol.67, pp.446-452, 2006.

S. S. Mirra, A. Heyman, D. Mckeel, S. M. Sumi, B. J. Crain et al., The Consortium to Establish a Registry for Alzheimer's Disease (CERAD). Part II. Standardization of the neuropathologic assessment of Alzheimer's disease, Neurology, vol.41, pp.479-486, 1991.

T. J. Montine, C. H. Phelps, T. G. Beach, E. H. Bigio, N. J. Cairns et al., National Institute on Aging-Alzheimer's Association guidelines for the neuropathologic assessment of Alzheimer's disease: a practical approach, Acta Neuropathol. (Berl.), vol.123, pp.1-11, 2012.

S. D. Mulder, R. Veerhuis, M. A. Blankenstein, and H. M. Nielsen, , 2012.

, Exp. Neurol, vol.233, pp.373-379

H. Nakanishi, Microglial functions and proteases, Mol. Neurobiol, vol.27, pp.163-176, 2003.

Y. Namba, M. Tomonaga, H. Kawasaki, E. Otomo, and K. Ikeda, Apolipoprotein E immunoreactivity in cerebral amyloid deposits and neurofibrillary tangles in Alzheimer's disease and kuru plaque amyloid in Creutzfeldt-Jakob disease, Brain Res, vol.541, pp.163-166, 1991.

L. H. Nelson and K. M. Lenz, The immune system as a novel regulator of sex differences in brain and behavioral development, J. Neurosci. Res, vol.95, pp.447-461, 2017.

T. J. Nelson, M. Sun, C. Lim, A. Sen, T. Khan et al., Bryostatin Effects on Cognitive Function and PKC? in Alzheimer's Disease Phase IIa and Expanded Access Trials, J. Alzheimers Dis. JAD, vol.58, pp.521-535, 2017.

P. J. Nestor, T. D. Fryer, and J. R. Hodges, Declarative memory impairments in Alzheimer's disease and semantic dementia, NeuroImage, vol.30, pp.1010-1020, 2006.

R. L. Neve, P. Harris, K. S. Kosik, D. M. Kurnit, and T. A. Donlon, Identification of cDNA clones for the human microtubule-associated protein tau and chromosomal localization of the genes for tau and microtubule-associated protein 2, Brain Res, vol.387, pp.271-280, 1986.

G. Neves, S. F. Cooke, and T. V. Bliss, Synaptic plasticity, memory and the hippocampus: a neural network approach to causality, Nat. Rev. Neurosci, vol.9, pp.65-75, 2008.

H. M. Nielsen, R. Veerhuis, B. Holmqvist, J. , and S. , Binding and uptake of A beta1-42 by primary human astrocytes in vitro, Glia, vol.57, pp.978-988, 2009.

R. A. Nixon, J. Wegiel, A. Kumar, W. H. Yu, C. Peterhoff et al., Extensive involvement of autophagy in Alzheimer disease: an immuno-electron microscopy study, J. Neuropathol. Exp. Neurol, vol.64, pp.113-122, 2005.

L. Noli, A. Capalbo, C. Ogilvie, Y. Khalaf, and D. Ilic, Discordant Growth of Monozygotic Twins Starts at the Blastocyst Stage: A Case Study, Stem Cell Rep, vol.5, pp.946-953, 2015.

C. Nyakas, I. Granic, L. G. Halmy, P. Banerjee, and P. G. Luiten, The basal forebrain cholinergic system in aging and dementia. Rescuing cholinergic neurons from neurotoxic amyloid-?42 with memantine, Behav. Brain Res, vol.221, pp.594-603, 2011.

S. Oddo, A. Caccamo, J. D. Shepherd, M. P. Murphy, T. E. Golde et al., Triple-transgenic model of Alzheimer's disease with plaques and tangles: intracellular Abeta and synaptic dysfunction, Neuron, vol.39, pp.409-421, 2003.

S. Oddo, A. Caccamo, M. Kitazawa, B. P. Tseng, and F. M. Laferla, Amyloid deposition precedes tangle formation in a triple transgenic model of Alzheimer's disease, Neurobiol Aging, vol.24, pp.1063-1070, 2003.

K. Oh, S. E. Perez, S. Lagalwar, L. Vana, L. Binder et al., Staging of Alzheimer's Pathology in Triple Transgenic Mice: A Light and Electron Microscopic Analysis, Int. J. Alzheimers Dis, 2010.

M. Olabarria, H. N. Noristani, A. Verkhratsky, and J. J. Rodriguez, Concomitant astroglial atrophy and astrogliosis in a triple transgenic animal model of Alzheimer's disease, Glia, vol.58, pp.831-838, 2010.

S. H. Oliet, R. Piet, and D. A. Poulain, Control of glutamate clearance and synaptic efficacy by glial coverage of neurons, Science, vol.292, pp.923-926, 2001.
URL : https://hal.archives-ouvertes.fr/inserm-00000059

G. Olmos and J. Lladó, Tumor necrosis factor alpha: a link between neuroinflammation and excitotoxicity, Mediators Inflamm, p.861231, 2014.

M. Orre, W. Kamphuis, S. Dooves, L. Kooijman, E. T. Chan et al., Reactive glia show increased immunoproteasome activity in Alzheimer's disease, Brain, vol.136, pp.1415-1431, 2013.

M. Orre, W. Kamphuis, L. M. Osborn, A. H. Jansen, L. Kooijman et al., Isolation of glia from Alzheimer's mice reveals inflammation and dysfunction, Neurobiol. Aging, vol.35, pp.2746-2760, 2014.

P. I. Ortinski, J. Dong, A. Mungenast, C. Yue, H. Takano et al., Selective induction of astrocytic gliosis generates deficits in neuronal inhibition, Nat Neurosci, vol.13, pp.584-591, 2010.

A. Panatier, D. T. Theodosis, J. P. Mothet, B. Touquet, L. Pollegioni et al.,

, Glia-derived D-serine controls NMDA receptor activity and synaptic memory, Cell, vol.125, pp.775-784

A. Panatier, J. Vallee, M. Haber, K. K. Murai, J. C. Lacaille et al., Astrocytes are endogenous regulators of basal transmission at central synapses, Cell, vol.146, pp.785-798, 2011.

F. Panza, V. Solfrizzi, D. Seripa, B. P. Imbimbo, M. Lozupone et al., Tau-Centric Targets and Drugs in Clinical Development for the Treatment of Alzheimer's Disease, BioMed Res. Int, p.3245935, 2016.

G. Perea, M. Navarrete, A. , and A. , Tripartite synapses: astrocytes process and control synaptic information, Trends Neurosci, vol.32, pp.421-431, 2009.

G. Perry, R. Friedman, G. Shaw, C. , and V. , Ubiquitin is detected in neurofibrillary tangles and senile plaque neurites of Alzheimer disease brains, Proc. Natl. Acad. Sci. U. S. A, vol.84, pp.3033-3036, 1987.

Y. Persidsky, S. H. Ramirez, J. Haorah, and G. D. Kanmogne, Blood-brain barrier: structural components and function under physiologic and pathologic conditions, J. Neuroimmune Pharmacol, 2006.

, J. Soc. NeuroImmune Pharmacol, vol.1, pp.223-236

D. Pfaff, U. Fiedler, A. , and H. G. , Emerging roles of the Angiopoietin-Tie and the ephrin-Eph systems as regulators of cell trafficking, J. Leukoc. Biol, vol.80, pp.719-726, 2006.

F. Pickford, E. Masliah, M. Britschgi, K. Lucin, R. Narasimhan et al., The autophagy-related protein beclin 1 shows reduced expression in early Alzheimer disease and regulates amyloid beta accumulation in mice, J. Clin. Invest, vol.118, pp.2190-2199, 2008.

L. Pini, M. Pievani, M. Bocchetta, D. Altomare, P. Bosco et al., Brain atrophy in Alzheimer's Disease and aging, Ageing Res. Rev, vol.30, pp.25-48, 2016.

C. Pomilio, P. Pavia, R. M. Gorojod, A. Vinuesa, A. Alaimo et al., Glial alterations from early to late stages in a model of Alzheimer's disease: Evidence of autophagy involvement in A? internalization, Hippocampus, vol.26, pp.194-210, 2016.

J. T. Porter and K. D. Mccarthy, Astrocytic neurotransmitter receptors in situ and in vivo, Prog Neurobiol, vol.51, pp.439-455, 1997.

C. Pottier, T. A. Ravenscroft, P. H. Brown, N. A. Finch, M. Baker et al., TYROBP genetic variants in early-onset Alzheimer's disease, vol.48, pp.9-222, 2016.

M. Prinz and J. Priller, The role of peripheral immune cells in the CNS in steady state and disease, Nat. Neurosci, vol.20, pp.136-144, 2017.

P. Raina, P. Santaguida, A. Ismaila, C. Patterson, D. Cowan et al., Effectiveness of cholinesterase inhibitors and memantine for treating dementia: evidence review for a clinical practice guideline, Ann. Intern. Med, vol.148, pp.379-397, 2008.

R. M. Ransohoff, A polarizing question: do M1 and M2 microglia exist?, Nat Neurosci, vol.19, pp.987-991, 2016.

L. M. Refolo, M. A. Pappolla, J. Lafrancois, B. Malester, S. D. Schmidt et al., A cholesterol-lowering drug reduces betaamyloid pathology in a transgenic mouse model of Alzheimer's disease, Neurobiol. Dis, vol.8, pp.890-899, 2001.

B. Reisberg, R. Doody, A. Stöffler, F. Schmitt, S. Ferris et al., Memantine in moderate-to-severe Alzheimer's disease, N. Engl. J. Med, vol.348, pp.1333-1341, 2003.

M. Ries and M. Sastre, Mechanisms of Abeta Clearance and Degradation by Glial Cells, Front Aging Neurosci, vol.8, p.160, 2016.

J. J. Rodríguez, V. C. Jones, M. Tabuchi, S. M. Allan, E. M. Knight et al., Impaired adult neurogenesis in the dentate gyrus of a triple transgenic mouse model of Alzheimer's disease, PloS One, vol.3, 2008.

J. J. Rodriguez, M. Olabarria, A. Chvatal, and A. Verkhratsky, Astroglia in dementia and Alzheimer's disease, Cell Death Differ, vol.16, pp.378-385, 2009.

E. Rodriguez-vieitez, L. Saint-aubert, S. F. Carter, O. Almkvist, K. Farid et al., Diverging longitudinal changes in astrocytosis and amyloid PET in autosomal dominant Alzheimer's disease, Brain, vol.139, pp.922-936, 2016.

A. E. Roher, J. D. Lowenson, S. Clarke, A. S. Woods, R. J. Cotter et al., beta-Amyloid-(1-42) is a major component of cerebrovascular amyloid deposits: implications for the pathology of Alzheimer disease, Proc. Natl. Acad. Sci. U. S. A, vol.90, pp.10836-10840, 1993.

S. Rossner, C. Lange-dohna, U. Zeitschel, and J. R. Perez-polo, Alzheimer's disease betasecretase BACE1 is not a neuron-specific enzyme, J. Neurochem, vol.92, pp.226-234, 2005.

C. C. Rudy, H. C. Hunsberger, D. S. Weitzner, and M. N. Reed, The role of the tripartite glutamatergic synapse in the pathophysiology of Alzheimer's disease, Aging Dis, vol.6, pp.131-148, 2015.

L. Saint-aubert, L. Lemoine, K. Chiotis, A. Leuzy, E. Rodriguez-vieitez et al., Tau PET imaging: present and future directions, Mol. Neurodegener, vol.12, p.19, 2017.

M. Sarazin, V. Chauviré, E. Gerardin, O. Colliot, S. Kinkingnéhun et al., The amnestic syndrome of hippocampal type in Alzheimer's disease: an MRI study, J. Alzheimers Dis. JAD, vol.22, pp.285-294, 2010.

C. Savarin-vuaillat and R. M. Ransohoff, Chemokines and chemokine receptors in neurological disease: raise, retain, or reduce?, Neurother. J. Am. Soc. Exp. Neurother, vol.4, pp.590-601, 2007.

G. D. Schellenberg, T. D. Bird, E. M. Wijsman, H. T. Orr, L. Anderson et al., , 1992.

R. Schliebs, A. , and T. , The significance of the cholinergic system in the brain during aging and in Alzheimer's disease, J. Neural Transm, vol.113, pp.1625-1644, 1996.

D. E. Schmechel, A. M. Saunders, W. J. Strittmatter, B. J. Crain, C. M. Hulette et al., Increased amyloid beta-peptide deposition in cerebral cortex as a consequence of apolipoprotein E genotype in late-onset Alzheimer disease, Proc. Natl. Acad. Sci. U. S. A, vol.90, pp.9649-9653, 1993.

M. Schöll, S. N. Lockhart, D. R. Schonhaut, J. P. O&apos;neil, M. Janabi et al., PET Imaging of Tau Deposition in the Aging Human Brain, vol.89, pp.971-982, 2016.

J. M. Schwarz and S. D. Bilbo, Sex, glia, and development: interactions in health and disease, 2012.

, Horm. Behav, vol.62, pp.243-253

J. D. Sedgwick, S. Schwender, H. Imrich, R. Dörries, G. W. Butcher et al., , 1991.

, Isolation and direct characterization of resident microglial cells from the normal and inflamed central nervous system, Proc. Natl. Acad. Sci. U. S. A, vol.88, pp.7438-7442

T. Del-ser, K. C. Steinwachs, H. J. Gertz, M. V. Andrés, B. Gómez-carrillo et al., Treatment of Alzheimer's disease with the GSK-3 inhibitor tideglusib: a pilot study, J. Alzheimers Dis. JAD, vol.33, pp.205-215, 2013.

C. Serguera and A. Bemelmans, Gene therapy of the central nervous system: general considerations on viral vectors for gene transfer into the brain, Rev. Neurol, vol.170, pp.727-738, 2014.

L. M. Shaffer, M. D. Dority, R. Gupta-bansal, R. C. Frederickson, S. G. Younkin et al.,

, Amyloid beta protein (A beta) removal by neuroglial cells in culture, Neurobiol. Aging, vol.16, pp.737-745

S. S. Shaftel, W. S. Griffin, and M. K. Banion, The role of interleukin-1 in neuroinflammation and Alzheimer disease: an evolving perspective, J. Neuroinflammation, vol.5, p.7, 2008.

Y. Shen, L. Lue, L. Yang, A. Roher, Y. Kuo et al., Complement activation by neurofibrillary tangles in Alzheimer's disease, Neurosci. Lett, vol.305, pp.165-168, 2001.

Y. Shen, L. Yang, L. , and R. , What does complement do in Alzheimer's disease? Old molecules with new insights, Transl. Neurodegener, vol.2, p.21, 2013.

Q. Shi, S. Chowdhury, R. Ma, K. X. Le, S. Hong et al., , 2017.

, Complement C3 deficiency protects against neurodegeneration in aged plaque-rich APP/PS1 mice, Sci. Transl. Med, vol.9, issue.392

K. Shibuki, H. Gomi, L. Chen, S. Bao, J. J. Kim et al., Deficient cerebellar long-term depression, impaired eyeblink conditioning, and normal motor coordination in GFAP mutant mice, Neuron, vol.16, pp.587-599, 1996.

K. Shuai, C. M. Horvath, L. H. Huang, S. A. Qureshi, D. Cowburn et al., , 1994.

, Interferon activation of the transcription factor Stat91 involves dimerization through SH2-phosphotyrosyl peptide interactions, Cell, vol.76, pp.821-828

T. Sigurdsson, V. Doyère, C. K. Cain, and J. E. Ledoux, Long-term potentiation in the amygdala: a cellular mechanism of fear learning and memory, Neuropharmacology, vol.52, pp.215-227, 2007.
URL : https://hal.archives-ouvertes.fr/hal-01468048

M. L. Simmons, C. G. Frondoza, and J. T. Coyle, Immunocytochemical localization of N-acetylaspartate with monoclonal antibodies, Neuroscience, vol.45, pp.37-45, 1991.

J. E. Simpson, P. G. Ince, G. Lace, G. Forster, P. J. Shaw et al., Astrocyte phenotype in relation to Alzheimer-type pathology in the ageing brain, Neurobiol Aging, vol.31, pp.578-590, 2010.

M. V. Sofroniew, Molecular dissection of reactive astrogliosis and glial scar formation, Trends Neurosci, vol.32, pp.638-647, 2009.

P. Sompol, J. L. Furman, M. M. Pleiss, S. D. Kraner, I. A. Artiushin et al., Calcineurin/NFAT Signaling in Activated Astrocytes Drives Network Hyperexcitability in A?-Bearing Mice, J. Neurosci. Off. J. Soc. Neurosci, vol.37, pp.6132-6148, 2017.

S. Song, S. Kim, Y. Hong, D. Jo, J. Lee et al., Essential role of E2-25K/Hip-2 in mediating amyloid-beta neurotoxicity, Mol. Cell, vol.12, pp.553-563, 2003.

A. Suzuki, S. A. Stern, O. Bozdagi, G. W. Huntley, R. H. Walker et al.,

, Astrocyte-neuron lactate transport is required for long-term memory formation, Cell, vol.144, pp.810-823

Y. Takeshita and R. M. Ransohoff, Inflammatory cell trafficking across the blood-brain barrier: chemokine regulation and in vitro models, Immunol. Rev, vol.248, pp.228-239, 2012.

J. Tanaka, K. Toku, B. Zhang, K. Ishihara, M. Sakanaka et al., Astrocytes prevent neuronal death induced by reactive oxygen and nitrogen species, Glia, vol.28, pp.85-96, 1999.

F. Tateno, R. Sakakibara, T. Kawai, M. Kishi, and T. Murano, Alpha-synuclein in the cerebrospinal fluid differentiates synucleinopathies (Parkinson Disease, dementia with Lewy bodies, multiple system atrophy) from Alzheimer disease, Alzheimer Dis. Assoc. Disord, vol.26, pp.213-216, 2012.

D. R. Thal, U. Rüb, M. Orantes, and H. Braak, Phases of A beta-deposition in the human brain and its relevance for the development of AD, Neurology, vol.58, pp.1791-1800, 2002.

D. R. Thal, W. S. Griffin, R. A. De-vos, and E. Ghebremedhin, Cerebral amyloid angiopathy and its relationship to Alzheimer's disease, Acta Neuropathol. (Berl.), vol.115, pp.599-609, 2008.

M. Theis and C. Giaume, Connexin-based intercellular communication and astrocyte heterogeneity, Brain Res, vol.1487, pp.88-98, 2012.

C. Theunis, N. Crespo-biel, V. Gafner, M. Pihlgren, M. P. López-deber et al., Efficacy and safety of a liposome-based vaccine against protein Tau, assessed in tau.P301L mice that model tauopathy, PloS One, vol.8, 2013.

D. Tweedie, R. A. Ferguson, K. Fishman, K. A. Frankola, H. Van-praag et al., Tumor necrosis factor-? synthesis inhibitor 3,6'-dithiothalidomide attenuates markers of inflammation, Alzheimer pathology and behavioral deficits in animal models of neuroinflammation and Alzheimer's disease, J. Neuroinflammation, vol.9, p.106, 2012.

M. F. Uchoa, V. A. Moser, and C. J. Pike, Interactions between inflammation, sex steroids, and Alzheimer's disease risk factors, Front. Neuroendocrinol, vol.43, pp.60-82, 2016.

J. D. Ulrich, M. B. Finn, Y. Wang, A. Shen, T. E. Mahan et al., Altered microglial response to Abeta plaques in APPPS1-21 mice heterozygous for TREM2, Mol Neurodegener, vol.9, p.20, 2014.

J. D. Ulrich, T. K. Ulland, M. Colonna, and D. M. Holtzman, Elucidating the Role of TREM2 in Alzheimer's Disease, Neuron, vol.94, pp.237-248, 2017.

J. Vandesompele, K. De-preter, F. Pattyn, B. Poppe, N. Van-roy et al.,

, Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple internal control genes, Genome Biol, vol.3, p.34

G. Vargas-alarcon, E. Juarez-cedillo, N. Martinez-rodriguez, J. M. Fragoso, N. Garcia-hernandez et al., Association of interleukin-10 polymorphisms with risk factors of Alzheimer's disease and other dementias (SADEM study), Immunol Lett, vol.177, pp.47-52, 2016.

M. M. Varnum and T. Ikezu, The classification of microglial activation phenotypes on neurodegeneration and regeneration in Alzheimer's disease brain, Arch. Immunol. Ther. Exp. (Warsz.), vol.60, pp.251-266, 2012.

M. J. Végh, C. M. Heldring, W. Kamphuis, S. Hijazi, A. J. Timmerman et al., Reducing hippocampal extracellular matrix reverses early memory deficits in a mouse model of Alzheimer's disease, Acta Neuropathol. Commun, vol.2, p.76, 2014.

C. Venegas and M. T. Heneka, Danger-associated molecular patterns in Alzheimer's disease, 2017.

, J. Leukoc. Biol, vol.101, pp.87-98

L. Verret, J. L. Jankowsky, G. M. Xu, D. R. Borchelt, R. et al., Alzheimer's-type amyloidosis in transgenic mice impairs survival of newborn neurons derived from adult hippocampal neurogenesis, J. Neurosci. Off. J. Soc. Neurosci, vol.27, pp.6771-6780, 2007.

S. Viana-da-silva, M. G. Haberl, P. Zhang, P. Bethge, C. Lemos et al., Early synaptic deficits in the APP/PS1 mouse model of Alzheimer's disease involve neuronal adenosine A2A receptors, Nat. Commun, vol.7, p.11915, 2016.

R. Virchow, , 1856.

, Gesammelte Abhandlungen zur wissenschaftlichen Medizin

A. Volianskis, R. Køstner, M. Mølgaard, S. Hass, and M. S. Jensen, Episodic memory deficits are not related to altered glutamatergic synaptic transmission and plasticity in the CA1 hippocampus of the APPswe/PS1?E9-deleted transgenic mice model of ß-amyloidosis, Neurobiol. Aging, vol.31, pp.1173-1187, 2010.

V. Berg, J. Prokop, S. Miller, K. R. Obst, J. Kälin et al., Inhibition of IL-12/IL-23 signaling reduces Alzheimer's diseaselike pathology and cognitive decline, Nat. Med, vol.18, pp.1812-1819, 2012.

D. M. Walsh and D. J. Selkoe, A beta oligomers -a decade of discovery, J. Neurochem, vol.101, pp.1172-1184, 2007.

Y. Wang and E. Mandelkow, Tau in physiology and pathology, Nat. Rev. Neurosci, vol.17, pp.5-21, 2016.

W. Wang, M. Tan, J. Yu, and L. Tan, Role of pro-inflammatory cytokines released from microglia in Alzheimer's disease, Ann. Transl. Med, vol.3, p.136, 2015.

Y. Wang, M. Cella, K. Mallinson, J. D. Ulrich, K. L. Young et al., TREM2 lipid sensing sustains the microglial response in an Alzheimer's disease model, Cell, vol.160, pp.1061-1071, 2015.

Y. Wang, T. K. Ulland, J. D. Ulrich, W. Song, J. A. Tzaferis et al., TREM2-mediated early microglial response limits diffusion and toxicity of amyloid plaques, J. Exp. Med, vol.213, pp.667-675, 2016.

Y. Wang, V. Balaji, S. Kaniyappan, L. Krüger, S. Irsen et al., The release and trans-synaptic transmission of Tau via exosomes, Mol. Neurodegener, vol.12, p.5, 2017.

S. Webster, R. , and J. , Relative efficacies of amyloid beta peptide (A beta) binding proteins in A beta aggregation, J. Neurosci. Res, vol.46, pp.58-66, 1996.

S. J. Webster, A. D. Bachstetter, and L. J. Van-eldik, Comprehensive behavioral characterization of an APP/PS-1 double knock-in mouse model of Alzheimer's disease, Alzheimers Res. Ther, vol.5, p.28, 2013.

C. Weigert, Beiträge zur Kenntnis der normalen menschlichen Neuroglia, Zeitschrift Für Psychologie Und Physiologie Der Sinnesorgane, Liepmann, 1895.

U. Wilhelmsson, E. A. Bushong, D. L. Price, B. L. Smarr, V. Phung et al., Redefining the concept of reactive astrocytes as cells that remain within their unique domains upon reaction to injury, Proc Natl Acad Sci U A, vol.103, pp.17513-17518, 2006.

O. Wirths and T. A. Bayer, Alpha-synuclein, Abeta and Alzheimer's disease, Prog. Neuropsychopharmacol. Biol. Psychiatry, vol.27, pp.103-108, 2003.

B. Wolozin, W. Kellman, P. Ruosseau, G. G. Celesia, and G. Siegel, Decreased prevalence of Alzheimer disease associated with 3-hydroxy-3-methyglutaryl coenzyme A reductase inhibitors, Arch. Neurol, vol.57, pp.1439-1443, 2000.

Z. Wu, Z. Guo, M. Gearing, C. , and G. , Tonic inhibition in dentate gyrus impairs long-term potentiation and memory in an Alzhiemer's disease model, Nat Commun, vol.5, p.4159, 2014.

T. Wyss-coray, C. Lin, F. Yan, G. Q. Yu, M. Rohde et al.,

, TGF-beta1 promotes microglial amyloid-beta clearance and reduces plaque burden in transgenic mice, Nat Med, vol.7, pp.612-618

T. Wyss-coray, J. D. Loike, T. C. Brionne, E. Lu, R. Anankov et al., Adult mouse astrocytes degrade amyloid-beta in vitro and in situ, Nat Med, vol.9, pp.453-457, 2003.

M. Q. Xia, S. X. Qin, L. J. Wu, C. R. Mackay, and B. T. Hyman, Immunohistochemical study of the beta-chemokine receptors CCR3 and CCR5 and their ligands in normal and Alzheimer's disease brains, Am. J. Pathol, vol.153, pp.31-37, 1998.

M. Q. Xia, B. J. Bacskai, R. B. Knowles, S. X. Qin, and B. T. Hyman, 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, vol.108, pp.227-235, 2000.

W. Xia, T. Yang, G. Shankar, I. M. Smith, Y. Shen et al., A specific enzyme-linked immunosorbent assay for measuring beta-amyloid protein oligomers in human plasma and brain tissue of patients with Alzheimer disease, Arch. Neurol, vol.66, pp.190-199, 2009.

H. Xiong, D. Callaghan, J. Wodzinska, J. Xu, M. Premyslova et al., Biochemical and behavioral characterization of the double transgenic mouse model (APPswe/PS1dE9) of Alzheimer's disease, Neurosci. Bull, vol.27, pp.221-232, 2011.

W. Xu, A. Marseglia, C. Ferrari, and H. Wang, Alzheimer's Disease: A Clinical Perspective, Neurodegenerative Diseases, 2013.

N. Yamamoto, M. Tanida, Y. Ono, R. Kasahara, Y. Fujii et al., , 2014.

, Leptin inhibits amyloid ?-protein degradation through decrease of neprilysin expression in primary cultured astrocytes, Biochem. Biophys. Res. Commun, vol.445, pp.214-217

R. Yan, Stepping closer to treating Alzheimer's disease patients with BACE1 inhibitor drugs, Transl. Neurodegener, vol.5, p.13, 2016.

P. Yan, X. Hu, H. Song, K. Yin, R. J. Bateman et al.,

, Matrix metalloproteinase-9 degrades amyloid-beta fibrils in vitro and compact plaques in situ, J Biol Chem, vol.281, pp.24566-24574

K. Yanamandra, N. Kfoury, H. Jiang, T. E. Mahan, S. Ma et al., Anti-tau antibodies that block tau aggregate seeding in vitro markedly decrease pathology and improve cognition in vivo, Neuron, vol.80, pp.402-414, 2013.

K. Yanamandra, H. Jiang, T. E. Mahan, S. E. Maloney, D. F. Wozniak et al., Anti-tau antibody reduces insoluble tau and decreases brain atrophy, Ann. Clin. Transl. Neurol, vol.2, pp.278-288, 2015.

Y. Yang, D. Shang, W. Zhao, W. Fang, C. et al., Microglial TNF-?dependent elevation of MHC class I expression on brain endothelium induced by amyloid-beta promotes T cell transendothelial migration, Neurochem. Res, vol.38, pp.2295-2304, 2013.

K. Yasojima, C. Schwab, E. G. Mcgeer, and P. L. Mcgeer, Up-regulated production and activation of the complement system in Alzheimer's disease brain, Am. J. Pathol, vol.154, pp.927-936, 1999.

W. H. Yu, A. Kumar, C. Peterhoff, L. Shapiro-kulnane, Y. Uchiyama et al., Autophagic vacuoles are enriched in amyloid precursor protein-secretase activities: implications for beta-amyloid peptide over-production and localization in Alzheimer's disease, Int. J. Biochem. Cell Biol, vol.36, pp.2531-2540, 2004.

W. H. Yu, A. M. Cuervo, A. Kumar, C. M. Peterhoff, S. D. Schmidt et al., Macroautophagy--a novel Beta-amyloid peptidegenerating pathway activated in Alzheimer's disease, J. Cell Biol, vol.171, pp.87-98, 2005.

X. Yuan, S. Sun, C. Tan, J. Yu, and L. Tan, The Role of ADAM10 in Alzheimer's Disease, J. Alzheimers Dis. JAD, vol.58, pp.303-322, 2017.

J. L. Zamanian, L. Xu, L. C. Foo, N. Nouri, L. Zhou et al., Genomic analysis of reactive astrogliosis, J Neurosci, vol.32, pp.6391-6410, 2012.

T. Zeis, I. Allaman, M. Gentner, K. Schroder, J. Tschopp et al.,

B. Zhang, C. Gaiteri, L. G. Bodea, Z. Wang, J. Mcelwee et al., Integrated systems approach identifies genetic nodes and networks in late-onset Alzheimer's disease, Cell, vol.153, pp.707-720, 2013.